1
|
Arzik Y, Kizilaslan M, Behrem S, Piel LMW, White SN, Çınar MU. Exploring Genetic Factors Associated with Moniezia spp. Tapeworm Resistance in Central Anatolian Merino Sheep via GWAS Approach. Animals (Basel) 2025; 15:812. [PMID: 40150341 PMCID: PMC11939720 DOI: 10.3390/ani15060812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 02/19/2025] [Accepted: 03/10/2025] [Indexed: 03/29/2025] Open
Abstract
Gastrointestinal parasite (GIP) infections pose significant challenges in pasture-based sheep farming, leading to economic losses and welfare concerns. This study aimed to uncover the genetic basis of resistance to Moniezia spp. infections in Central Anatolian Merino (CAM) sheep. Genome-Wide Association Analysis (GWAS) was conducted between Moniezia spp. egg burden and genomic data from 226 CAM lambs. Thirteen significant Single-Nucleotide Polymorphisms (SNPs) were identified, with five surpassing the genome-wide threshold and eight exceeding the chromosome-wide threshold. Functional annotation revealed associations with genes involved in immune function, notably CD79A and MAP3K7. CD79A, integral to B-cell activation and antibody production, plays a key role in the immune response against parasitic infections. Its interaction with helminth-derived proteins modulates B-cell function, highlighting its potential as a therapeutic target. MAP3K7, a central regulator of immune signaling pathways, modulates host responses to helminth infections by influencing NF-κB activity. Additionally, it regulates macrophage function in bacterial infections, showcasing its versatility in mediating immune responses against diverse pathogens. From a practical perspective, the findings of the current research underscore the potential of integrating genomic information into breeding programs to bolster disease resilience in livestock populations for sustainable production purposes. However, further research is needed to elucidate the functional significance of identified SNPs and associated genes. This study underscores the potential of genomic approaches in combating parasitic diseases and promoting sustainable agriculture in sheep production systems.
Collapse
Affiliation(s)
- Yunus Arzik
- Department of Animal Science, Faculty of Veterinary Medicine, Aksaray University, 68000 Aksaray, Türkiye; (Y.A.); (S.B.)
| | - Mehmet Kizilaslan
- Department of Animal Science, Faculty of Agriculture, Erciyes University, 38039 Kayseri, Türkiye;
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Sedat Behrem
- Department of Animal Science, Faculty of Veterinary Medicine, Aksaray University, 68000 Aksaray, Türkiye; (Y.A.); (S.B.)
| | - Lindsay M. W. Piel
- USDA-ARS Animal Disease Research 3003 ADBF, Washington State University, Pullman, WA 99164, USA;
| | - Stephen N. White
- USDA-ARS Poultry Microbiological Safety and Processing Research, US National Poultry Research Center, Athens, GA 30605, USA;
- Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, Pullman, WA 99164, USA
| | - Mehmet Ulaş Çınar
- Department of Animal Science, Faculty of Agriculture, Erciyes University, 38039 Kayseri, Türkiye;
- Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, Pullman, WA 99164, USA
| |
Collapse
|
2
|
Smith SP, Smith OS, Mostafavi H, Peng D, Berg JJ, Edge MD, Harpak A. A Litmus Test for Confounding in Polygenic Scores. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.01.635985. [PMID: 39975133 PMCID: PMC11838432 DOI: 10.1101/2025.02.01.635985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Polygenic scores (PGSs) are being rapidly adopted for trait prediction in the clinic and beyond. PGSs are often thought of as capturing the direct genetic effect of one's genotype on their phenotype. However, because PGSs are constructed from population-level associations, they are influenced by factors other than direct genetic effects, including stratification, assortative mating, and dynastic effects ("SAD effects"). Our interpretation and application of PGSs may hinge on the relative impact of SAD effects, since they may often be environmentally or culturally mediated. We developed a method that estimates the proportion of variance in a PGS (in a given sample) that is driven by direct effects, SAD effects, and their covariance. We leverage a comparison of a PGS of interest based on a standard GWAS with a PGS based on a sibling GWAS-which is largely immune to SAD effects-to quantify the relative contribution of each type of effect to variance in the PGS of interest. Our method, Partitioning Genetic Scores Using Siblings (PGSUS, pron. "Pegasus"), breaks down variance components further by axes of genetic ancestry, allowing for a nuanced interpretation of SAD effects. In particular, PGSUS can detect stratification along major axes of ancestry as well as SAD variance that is "isotropic" with respect to axes of ancestry. Applying PGSUS, we found evidence of stratification in PGSs constructed using large meta-analyses of height and educational attainment as well as in a range of PGSs constructed using the UK Biobank. In some instances, a given PGS appears to be stratified along a major axis of ancestry in one prediction sample but not in another (for example, in comparisons of prediction in samples from different countries, or in ancient DNA vs. contemporary samples). Finally, we show that different approaches for adjustment for population structure in GWASs have distinct advantages with respect to mitigation of ancestry-axis-specific and isotropic SAD variance in PGS. Our study illustrates how family-based designs can be combined with standard population-based designs to guide the interpretation and application of genomic predictors.
Collapse
Affiliation(s)
- Samuel Pattillo Smith
- Department of Population Health, University of Texas at Austin, Austin, TX
- Department of Integrative Biology, University of Texas at Austin, Austin, TX
| | - Olivia S. Smith
- Department of Population Health, University of Texas at Austin, Austin, TX
- Department of Integrative Biology, University of Texas at Austin, Austin, TX
| | | | - Dandan Peng
- Department of Computational Biology, University of Southern California, Los Angeles, CA
| | - Jeremy J. Berg
- Department of Human Genetics, University of Chicago, Chicago, IL
| | - Michael D. Edge
- Department of Computational Biology, University of Southern California, Los Angeles, CA
| | - Arbel Harpak
- Department of Population Health, University of Texas at Austin, Austin, TX
- Department of Integrative Biology, University of Texas at Austin, Austin, TX
| |
Collapse
|
3
|
Kizilaslan M, Arzik Y, Behrem S, Yavuz E, White SN, Cinar MU. Unravelling the Genetic Architecture of Serum Biochemical Indicators in Sheep. Genes (Basel) 2024; 15:990. [PMID: 39202351 PMCID: PMC11353979 DOI: 10.3390/genes15080990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 07/16/2024] [Accepted: 07/23/2024] [Indexed: 09/03/2024] Open
Abstract
Serum biochemical indicators serve as vital proxies that reflect the physiological state and functions of different organs. The genetic parameters and molecular mechanisms underlying serum biochemical indicators of sheep (Ovis aries) have not been well understood. Therefore, the aim of the present study was to identify the genetic architecture and genomic loci underlying ten serum biochemical indicators in sheep, including alanine transaminase, aspartate transferase, lactate dehydrogenase, cholesterol, glucose, phosphorus, calcium, creatinine, urea and total protein levels. We implemented genetic parameter estimations and GWASs for each trait in 422 Akkaraman lambs. Overall, low to moderate heritability estimates were found in the range of 0.14-0.55. Additionally, low to high genetic correlations were observed among traits. In total, 23 SNP loci were associated with serum biochemical indicators leading to 19 genes. These were SPTA1, MGST2, CACUL1, IGFBP7, PARD3, PHB1, SLC15A5, TRIM35, RGS6, NUP93, CNTNAP2, SLC7A11, B3GALT5, DPP10, HST2ST1, NRP1, LRP1B, MAP3K9 and ENSOARG00020040484.1, as well as LOC101103187, LOC101117162, LOC105611309 and LOC101118029. To our knowledge, these data provide the first associations between SPTA1 and serum cholesterol and between ENSOARG00020040484.1 and serum glucose. The current findings provide a comprehensive inventory of the relationships between serum biochemical parameters, genetic variants and disease-relevant characteristics. This information may facilitate the identification of therapeutic targets and fluid biomarkers and establish a strong framework for comprehending the pathobiology of complex diseases as well as providing targets for sheep genetic improvement programs.
Collapse
Affiliation(s)
- Mehmet Kizilaslan
- Faculty of Agriculture, Department of Animal Science, Erciyes University, Kayseri 38039, Türkiye; (M.K.); (Y.A.)
- International Center for Livestock Research and Training, Ministry of Agriculture and Forestry, Ankara 06852, Türkiye
| | - Yunus Arzik
- Faculty of Agriculture, Department of Animal Science, Erciyes University, Kayseri 38039, Türkiye; (M.K.); (Y.A.)
- International Center for Livestock Research and Training, Ministry of Agriculture and Forestry, Ankara 06852, Türkiye
| | - Sedat Behrem
- Department of Veterinary Sciences, Aksaray University, Aksaray 68100, Türkiye
| | - Esra Yavuz
- International Center for Livestock Research and Training, Ministry of Agriculture and Forestry, Ankara 06852, Türkiye
| | - Stephen N. White
- Department of Veterinary Microbiology & Pathology, College of Veterinary Medicine, Washington State University, Pullman, WA 99164, USA;
| | - Mehmet Ulas Cinar
- Faculty of Agriculture, Department of Animal Science, Erciyes University, Kayseri 38039, Türkiye; (M.K.); (Y.A.)
- Department of Veterinary Microbiology & Pathology, College of Veterinary Medicine, Washington State University, Pullman, WA 99164, USA;
| |
Collapse
|
4
|
Greenwood JR, Lacorte-Apostol V, Kroj T, Padilla J, Telebanco-Yanoria MJ, Glaus AN, Roulin A, Padilla A, Zhou B, Keller B, Krattinger SG. Genome-wide association analysis uncovers rice blast resistance alleles of Ptr and Pia. Commun Biol 2024; 7:607. [PMID: 38769168 PMCID: PMC11106262 DOI: 10.1038/s42003-024-06244-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 04/24/2024] [Indexed: 05/22/2024] Open
Abstract
A critical step to maximize the usefulness of genome-wide association studies (GWAS) in plant breeding is the identification and validation of candidate genes underlying genetic associations. This is of particular importance in disease resistance breeding where allelic variants of resistance genes often confer resistance to distinct populations, or races, of a pathogen. Here, we perform a genome-wide association analysis of rice blast resistance in 500 genetically diverse rice accessions. To facilitate candidate gene identification, we produce de-novo genome assemblies of ten rice accessions with various rice blast resistance associations. These genome assemblies facilitate the identification and functional validation of novel alleles of the rice blast resistance genes Ptr and Pia. We uncover an allelic series for the unusual Ptr rice blast resistance gene, and additional alleles of the Pia resistance genes RGA4 and RGA5. By linking these associations to three thousand rice genomes we provide a useful tool to inform future rice blast breeding efforts. Our work shows that GWAS in combination with whole-genome sequencing is a powerful tool for gene cloning and to facilitate selection of specific resistance alleles for plant breeding.
Collapse
Affiliation(s)
- Julian R Greenwood
- Department of Plant and Microbial Biology, University of Zürich, Zürich, Switzerland.
- Research School of Biology, Australian National University, Canberra, ACT, Australia.
| | | | - Thomas Kroj
- PHIM Plant Health Institute, University of Montpellier, INRAE, CIRAD, Institut Agro, IRD, Montpellier, France
| | - Jonas Padilla
- International Rice Research Institute, Los Baños, Philippines
| | | | - Anna N Glaus
- Department of Plant and Microbial Biology, University of Zürich, Zürich, Switzerland
| | - Anne Roulin
- Agroscope, Müller-Thurgau-Strasse 29, 8820, Wädenswil, Switzerland
| | - André Padilla
- Centre de Biologie Structurale, CBS, University of Montpellier, CNRS UMR 5048, INSERM U, 1054, Montpellier, France
| | - Bo Zhou
- International Rice Research Institute, Los Baños, Philippines.
| | - Beat Keller
- Department of Plant and Microbial Biology, University of Zürich, Zürich, Switzerland.
| | - Simon G Krattinger
- Plant Science Program, Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia.
- Center for Desert Agriculture, KAUST, Thuwal, 23955-6900, Saudi Arabia.
| |
Collapse
|
5
|
Mini A, Touzy G, Beauchêne K, Cohan JP, Heumez E, Oury FX, Rincent R, Lafarge S, Le Gouis J. Genetic regions determine tolerance to nitrogen deficiency in European elite bread wheats grown under contrasting nitrogen stress scenarios. TAG. THEORETICAL AND APPLIED GENETICS. THEORETISCHE UND ANGEWANDTE GENETIK 2023; 136:218. [PMID: 37815653 DOI: 10.1007/s00122-023-04468-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 09/20/2023] [Indexed: 10/11/2023]
Abstract
KEY MESSAGE Clustering 24 environments in four contrasting nitrogen stress scenarios enabled the detection of genetic regions determining tolerance to nitrogen deficiency in European elite bread wheats. Increasing the nitrogen use efficiency of wheat varieties is an important goal for breeding. However, most genetic studies of wheat grown at different nitrogen levels in the field report significant interactions with the genotype. The chromosomal regions possibly involved in these interactions are largely unknown. The objective of this study was to quantify the response of elite bread wheat cultivars to different nitrogen field stress scenarios and identify genomic regions involved in this response. For this purpose, 212 elite bread wheat varieties were grown in a multi-environment trial at different nitrogen levels. Genomic regions associated with grain yield, protein concentration and grain protein deviation responses to nitrogen deficiency were identified. Environments were clustered according to adjusted means for grain yield, yield components and grain protein concentration. Four nitrogen availability scenarios were identified: optimal condition, moderate early deficiency, severe late deficiency, and severe continuous deficiency. A large range of tolerance to nitrogen deficiency was observed among varieties, which were ranked differently in different nitrogen deficiency scenarios. The well-known negative correlation between grain yield and grain protein concentration also existed between their respective tolerance indices. Interestingly, the tolerance indices for grain yield and grain protein deviation were either null or weakly positive meaning that breeding for the two traits should be less difficult than expected. Twenty-two QTL regions were identified for the tolerance indices. By selecting associated markers, these regions may be selected separately or combined to improve the tolerance to N deficiency within a breeding programme.
Collapse
Affiliation(s)
- Agathe Mini
- UMR GDEC, INRAE, Université Clermont Auvergne, 63100, Clermont-Ferrand, France
- Biogemma, Centre de Recherche de Chappes, Route d'Ennezat CS90216, 63720, Chappes, France
| | - Gaëtan Touzy
- Biogemma, Centre de Recherche de Chappes, Route d'Ennezat CS90216, 63720, Chappes, France
- Arvalis-Institut du Végétal, 41240, Beauce la Romaine, France
| | - Katia Beauchêne
- Arvalis-Institut du Végétal, 41240, Beauce la Romaine, France
| | - Jean-Pierre Cohan
- Arvalis-Institut du Végétal, Station Expérimentale, 91190, Villiers le Bâcle, France
| | | | | | - Renaud Rincent
- UMR GDEC, INRAE, Université Clermont Auvergne, 63100, Clermont-Ferrand, France
| | - Stéphane Lafarge
- Biogemma, Centre de Recherche de Chappes, Route d'Ennezat CS90216, 63720, Chappes, France
| | - Jacques Le Gouis
- UMR GDEC, INRAE, Université Clermont Auvergne, 63100, Clermont-Ferrand, France.
| |
Collapse
|
6
|
Yousri NA, Albagha OME, Hunt SC. Integrated epigenome, whole genome sequence and metabolome analyses identify novel multi-omics pathways in type 2 diabetes: a Middle Eastern study. BMC Med 2023; 21:347. [PMID: 37679740 PMCID: PMC10485955 DOI: 10.1186/s12916-023-03027-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 08/09/2023] [Indexed: 09/09/2023] Open
Abstract
BACKGROUND T2D is of high prevalence in the middle east and thus studying its mechanisms is of a significant importance. Using 1026 Qatar BioBank samples, epigenetics, whole genome sequencing and metabolomics were combined to further elucidate the biological mechanisms of T2D in a population with a high prevalence of T2D. METHODS An epigenome-wide association study (EWAS) with T2D was performed using the Infinium 850K EPIC array, followed by whole genome-wide sequencing SNP-CpG association analysis (> 5.5 million SNPs) and a methylome-metabolome (CpG-metabolite) analysis of the identified T2D sites. RESULTS A total of 66 T2D-CpG associations were identified, including 63 novel sites in pathways of fructose and mannose metabolism, insulin signaling, galactose, starch and sucrose metabolism, and carbohydrate absorption and digestion. Whole genome SNP associations with the 66 CpGs resulted in 688 significant CpG-SNP associations comprising 22 unique CpGs (33% of the 66 CPGs) and included 181 novel pairs or pairs in novel loci. Fourteen of the loci overlapped published GWAS loci for diabetes related traits and were used to identify causal associations of HK1 and PFKFB2 with HbA1c. Methylome-metabolome analysis identified 66 significant CpG-metabolite pairs among which 61 pairs were novel. Using the identified methylome-metabolome associations, methylation QTLs, and metabolic networks, a multi-omics network was constructed which suggested a number of metabolic mechanisms underlying T2D methylated genes. 1-palmitoyl-2-oleoyl-GPE (16:0/18:1) - a triglyceride-associated metabolite, shared a common network with 13 methylated CpGs, including TXNIP, PFKFB2, OCIAD1, and BLCAP. Mannonate - a food component/plant shared a common network with 6 methylated genes, including TXNIP, BLCAP, THBS4 and PEF1, pointing to a common possible cause of methylation in those genes. A subnetwork with alanine, glutamine, urea cycle (citrulline, arginine), and 1-carboxyethylvaline linked to PFKFB2 and TXNIP revealed associations with kidney function, hypertension and triglyceride metabolism. The pathway containing STYXL1-POR was associated with a sphingosine-ceramides subnetwork associated with HDL-C and LDL-C and point to steroid perturbations in T2D. CONCLUSIONS This study revealed several novel methylated genes in T2D, with their genomic variants and associated metabolic pathways with several implications for future clinical use of multi-omics associations in disease and for studying therapeutic targets.
Collapse
Affiliation(s)
- Noha A Yousri
- Genetic Medicine, Weill Cornell Medicine-Qatar, Doha, Qatar.
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar.
- Computer and Systems Engineering, Alexandria University, Alexandria, Egypt.
| | - Omar M E Albagha
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
| | - Steven C Hunt
- Genetic Medicine, Weill Cornell Medicine-Qatar, Doha, Qatar
| |
Collapse
|
7
|
Valente J, Gerin F, Mini A, Richard R, Le Gouis J, Prigent-Combaret C, Moënne-Loccoz Y. Symbiotic Variations among Wheat Genotypes and Detection of Quantitative Trait Loci for Molecular Interaction with Auxin-Producing Azospirillum PGPR. Microorganisms 2023; 11:1615. [PMID: 37375117 DOI: 10.3390/microorganisms11061615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 06/13/2023] [Accepted: 06/17/2023] [Indexed: 06/29/2023] Open
Abstract
Crop varieties differ in their ability to interact with Plant Growth-Promoting Rhizobacteria (PGPR), but the genetic basis for these differences is unknown. This issue was addressed with the PGPR Azospirillum baldaniorum Sp245, using 187 wheat accessions. We screened the accessions based on the seedling colonization by the PGPR and the expression of the phenylpyruvate decarboxylase gene ppdC (for synthesis of the auxin indole-3-acetic acid), using gusA fusions. Then, the effects of the PGPR on the selected accessions stimulating Sp245 (or not) were compared in soil under stress. Finally, a genome-wide association approach was implemented to identify the quantitative trait loci (QTL) associated with PGPR interaction. Overall, the ancient genotypes were more effective than the modern genotypes for Azospirillum root colonization and ppdC expression. In non-sterile soil, A. baldaniorum Sp245 improved wheat performance for three of the four PGPR-stimulating genotypes and none of the four non-PGPR-stimulating genotypes. The genome-wide association did not identify any region for root colonization but revealed 22 regions spread on 11 wheat chromosomes for ppdC expression and/or ppdC induction rate. This is the first QTL study focusing on molecular interaction with PGPR bacteria. The molecular markers identified provide the possibility to improve the capacity of modern wheat genotypes to interact with Sp245, as well as, potentially, other Azospirillum strains.
Collapse
Affiliation(s)
- Jordan Valente
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS, INRAE, VetAgro Sup, UMR5557 Ecologie Microbienne, 43 Bd du 11 Novembre 1918, F-69622 Villeurbanne, France
| | - Florence Gerin
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS, INRAE, VetAgro Sup, UMR5557 Ecologie Microbienne, 43 Bd du 11 Novembre 1918, F-69622 Villeurbanne, France
| | - Agathe Mini
- GDEC, INRAE, UCA, F-63000 Clermont-Ferrand, France
| | | | | | - Claire Prigent-Combaret
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS, INRAE, VetAgro Sup, UMR5557 Ecologie Microbienne, 43 Bd du 11 Novembre 1918, F-69622 Villeurbanne, France
| | - Yvan Moënne-Loccoz
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS, INRAE, VetAgro Sup, UMR5557 Ecologie Microbienne, 43 Bd du 11 Novembre 1918, F-69622 Villeurbanne, France
| |
Collapse
|
8
|
Majumdar S, Basu S, McGue M, Chatterjee S. Simultaneous selection of multiple important single nucleotide polymorphisms in familial genome wide association studies data. Sci Rep 2023; 13:8476. [PMID: 37231056 PMCID: PMC10213008 DOI: 10.1038/s41598-023-35379-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 05/17/2023] [Indexed: 05/27/2023] Open
Abstract
We propose a resampling-based fast variable selection technique for detecting relevant single nucleotide polymorphisms (SNP) in a multi-marker mixed effect model. Due to computational complexity, current practice primarily involves testing the effect of one SNP at a time, commonly termed as 'single SNP association analysis'. Joint modeling of genetic variants within a gene or pathway may have better power to detect associated genetic variants, especially the ones with weak effects. In this paper, we propose a computationally efficient model selection approach-based on the e-values framework-for single SNP detection in families while utilizing information on multiple SNPs simultaneously. To overcome computational bottleneck of traditional model selection methods, our method trains one single model, and utilizes a fast and scalable bootstrap procedure. We illustrate through numerical studies that our proposed method is more effective in detecting SNPs associated with a trait than either single-marker analysis using family data or model selection methods that ignore the familial dependency structure. Further, we perform gene-level analysis in Minnesota Center for Twin and Family Research (MCTFR) dataset using our method to detect several SNPs using this that have been implicated to be associated with alcohol consumption.
Collapse
Affiliation(s)
- Subhabrata Majumdar
- University of Minnesota Twin Cities, Minneapolis, USA.
- AI Risk and Vulnerability Alliance, Seattle, USA.
| | - Saonli Basu
- University of Minnesota Twin Cities, Minneapolis, USA
| | - Matt McGue
- University of Minnesota Twin Cities, Minneapolis, USA
| | | |
Collapse
|
9
|
Arzik Y, Kizilaslan M, Behrem S, White SN, Piel LMW, Cinar MU. Genome-Wide Scan of Wool Production Traits in Akkaraman Sheep. Genes (Basel) 2023; 14:genes14030713. [PMID: 36980985 PMCID: PMC10048666 DOI: 10.3390/genes14030713] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 12/26/2022] [Accepted: 03/10/2023] [Indexed: 03/17/2023] Open
Abstract
The objective of this study was to uncover the genetic background of wool quality, a production trait, by estimating genomic heritability and implementing GWAS in Akkaraman sheep. The wool characteristics measured included fibre diameter (FD) and staple length (SL) at the age of 8 months and yearling fibre diameter (YFD), yearling staple length (YSL) and yearling greasy fleece weight (YGFW) at 18 months of age. Animals were genotyped using the Axiom 50 K Ovine Genotyping Array. Maximum likelihood estimations of a linear mixed model (LMM) were used to estimate genomic heritability, where GWAS was conducted following a score test of each trait. Genomic heritability estimates for the traits ranged between 0.22 and 0.63, indicating that phenotypes have a moderate range of heritability. One genome- and six chromosome-wide significant SNPs were associated with the wool traits in Akkaraman lambs. Accordingly, TRIM2, MND1, TLR2, RNF175, CEP290, TMTC3, RERE, SLC45A1, SOX2, MORN1, SKI, FAAP20, PRKCZ, GABRD, CFAP74, CALML6 and TMEM52 genes as well as nine uncharacterized regions (LOC101118971, LOC105609137, LOC105603067, LOC101122892, LOC106991694, LOC106991467, LOC106991455, LOC105616534 and LOC105609719) were defined as plausible candidates. The findings of this study shed light on the genetics of wool quality and yield for the Akkaraman breed and suggests targets for breeders during systematic breeding programmes.
Collapse
Affiliation(s)
- Yunus Arzik
- Department of Animal Science, Faculty of Agriculture, Erciyes University, 38039 Kayseri, Türkiye
- International Center for Livestock Research and Training Center, Ministry of Agriculture and Forestry, 06852 Ankara, Türkiye
| | - Mehmet Kizilaslan
- Department of Animal Science, Faculty of Agriculture, Erciyes University, 38039 Kayseri, Türkiye
- International Center for Livestock Research and Training Center, Ministry of Agriculture and Forestry, 06852 Ankara, Türkiye
- Correspondence: ; Tel.: +90-553-6725986
| | - Sedat Behrem
- Department of Animal Science, Faculty of Veterinary Medicine, Aksaray University, 68100 Aksaray, Türkiye
| | - Stephen N. White
- Department of Veterinary Microbiology & Pathology, College of Veterinary Medicine, Washington State University, Pullman, WA 99164, USA
| | - Lindsay M. W. Piel
- USDA-ARS Animal Disease Research 3003 ADBF, Washington State University, Pullman, WA 99164, USA
| | - Mehmet Ulas Cinar
- Department of Animal Science, Faculty of Agriculture, Erciyes University, 38039 Kayseri, Türkiye
- Department of Veterinary Microbiology & Pathology, College of Veterinary Medicine, Washington State University, Pullman, WA 99164, USA
| |
Collapse
|
10
|
Alamin M, Sultana MH, Lou X, Jin W, Xu H. Dissecting Complex Traits Using Omics Data: A Review on the Linear Mixed Models and Their Application in GWAS. PLANTS (BASEL, SWITZERLAND) 2022; 11:3277. [PMID: 36501317 PMCID: PMC9739826 DOI: 10.3390/plants11233277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 11/23/2022] [Accepted: 11/25/2022] [Indexed: 06/17/2023]
Abstract
Genome-wide association study (GWAS) is the most popular approach to dissecting complex traits in plants, humans, and animals. Numerous methods and tools have been proposed to discover the causal variants for GWAS data analysis. Among them, linear mixed models (LMMs) are widely used statistical methods for regulating confounding factors, including population structure, resulting in increased computational proficiency and statistical power in GWAS studies. Recently more attention has been paid to pleiotropy, multi-trait, gene-gene interaction, gene-environment interaction, and multi-locus methods with the growing availability of large-scale GWAS data and relevant phenotype samples. In this review, we have demonstrated all possible LMMs-based methods available in the literature for GWAS. We briefly discuss the different LMM methods, software packages, and available open-source applications in GWAS. Then, we include the advantages and weaknesses of the LMMs in GWAS. Finally, we discuss the future perspective and conclusion. The present review paper would be helpful to the researchers for selecting appropriate LMM models and methods quickly for GWAS data analysis and would benefit the scientific society.
Collapse
Affiliation(s)
- Md. Alamin
- Institute of Bioinformatics, Zhejiang University, Hangzhou 310058, China
- Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | | | - Xiangyang Lou
- Department of Biostatistics, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Wenfei Jin
- Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Haiming Xu
- Institute of Bioinformatics, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
11
|
Estimates of genomic heritability and genome-wide association studies for blood parameters in Akkaraman sheep. Sci Rep 2022; 12:18477. [PMID: 36323871 PMCID: PMC9630504 DOI: 10.1038/s41598-022-22966-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 10/21/2022] [Indexed: 11/06/2022] Open
Abstract
The aim of this study was to estimate genomic heritability and the impact that genetic backgrounds have on blood parameters in Akkaraman sheep by conducting genome-wide association studies and regional heritability mapping analysis. Genomic heritability estimates for blood parameters ranged from 0.00 to 0.55, indicating that measured phenotypes have a low to moderate heritability. A total of 7 genome- and 13 chromosome-wide significant SNPs were associated with phenotypic changes in 15 blood parameters tested. Accordingly, SCN7A, SCN9A, MYADM-like, CCDC67, ITGA9, MGAT5, SLC19A1, AMPH, NTRK2, MSRA, SLC35F3, SIRT6, CREB3L3, and NAV3 genes as well as three undefined regions (LOC101117887, LOC106991526 and LOC105608461) were suggested as candidates. Most of the identified genes were involved in basic biological processes that are essential to immune system function and cellular growth; specific functions include cellular transport, histone deacetylation, cell differentiation, erythropoiesis, and endocytosis. The top significant SNP for HCT, MCH, and MCHC was found within a genomic region mainly populated by the MYADM-like gene family. This region was previously suggested to be under historical selection pressure in many sheep breeds from various parts of the world. These results have implications on animal breeding program studies due to the effect that the genetic background has on blood parameters, which underlying many productive and wellness related traits.
Collapse
|
12
|
Kizilaslan M, Arzik Y, White SN, Piel LMW, Cinar MU. Genetic Parameters and Genomic Regions Underlying Growth and Linear Type Traits in Akkaraman Sheep. Genes (Basel) 2022; 13:genes13081414. [PMID: 36011330 PMCID: PMC9407525 DOI: 10.3390/genes13081414] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 08/06/2022] [Accepted: 08/08/2022] [Indexed: 02/07/2023] Open
Abstract
In the current study, the genetic architecture of growth and linear type traits were investigated in Akkaraman sheep. Estimations of genomic heritability, genetic correlations, and phenotypic correlations were implemented for 17 growth and linear type traits of 473 Akkaraman lambs by the univariate and multivariate analysis of animal mixed models. Correspondingly, moderate heritability estimates, as well as high and positive genetic/phenotypic correlations were found between growth and type traits. On the other hand, 2 genome-wide and 19 chromosome-wide significant single nucleotide polymorphisms were found to be associated with the traits as a result of animal mixed model-based genome-wide association analyses. Accordingly, we propose several genes located on different chromosomes (e.g., PRDM2, PTGDR, PTPRG, KCND2, ZNF260, CPE, GRID2, SCD5, SPIDR, ZNF407, HCN3, TMEM50A, FKBP1A, TLE4, SP1, SLC44A1, and MYOM3) as putative quantitative trait loci for the 22 growth and linear type traits studied. In our study, specific genes (e.g., TLE4, PTGDR, and SCD5) were found common between the traits studied, suggesting an interplay between the genetic backgrounds of these traits. The fact that four of the proposed genes (TLE4, MYOM3, SLC44A1, and TMEM50A) are located on sheep chromosome 2 confirms the importance of these genomic regions for growth and morphological structure in sheep. The results of our study are therefore of great importance for the development of efficient selection indices and marker-assisted selection programs, as well as for the understanding of the genetic architecture of growth and linear traits in sheep.
Collapse
Affiliation(s)
- Mehmet Kizilaslan
- Department of Animal Science, Faculty of Agriculture, Erciyes University, Kayseri 38039, Turkey
- International Center for Livestock Research and Training Center, Ministry of Agriculture and Forestry, Ankara 06852, Turkey
| | - Yunus Arzik
- Department of Animal Science, Faculty of Agriculture, Erciyes University, Kayseri 38039, Turkey
- International Center for Livestock Research and Training Center, Ministry of Agriculture and Forestry, Ankara 06852, Turkey
| | - Stephen N. White
- Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, Pullman, WA 99164, USA
| | - Lindsay M. W. Piel
- USDA-ARS Animal Disease Research, Washington State University Pullman, Pullman, WA 991646630, USA
| | - Mehmet Ulas Cinar
- Department of Animal Science, Faculty of Agriculture, Erciyes University, Kayseri 38039, Turkey
- Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, Pullman, WA 99164, USA
- Correspondence: ; Tel.: +90-352-2076666-38601
| |
Collapse
|
13
|
Ray D, Vergara C, Taub MA, Wojcik G, Ladd‐Acosta C, Beaty TH, Duggal P. Benchmarking statistical methods for analyzing parent-child dyads in genetic association studies. Genet Epidemiol 2022; 46:266-284. [PMID: 35451532 PMCID: PMC9356976 DOI: 10.1002/gepi.22453] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 02/06/2022] [Accepted: 03/15/2022] [Indexed: 11/24/2022]
Abstract
Genetic association studies of child health outcomes often employ family-based study designs. One of the most popular family-based designs is the case-parent trio design that considers the smallest possible nuclear family consisting of two parents and their affected child. This trio design is particularly advantageous for studying relatively rare disorders because it is less prone to type 1 error inflation due to population stratification compared to population-based study designs (e.g., case-control studies). However, obtaining genetic data from both parents is difficult, from a practical perspective, and many large studies predominantly measure genetic variants in mother-child dyads. While some statistical methods for analyzing parent-child dyad data (most commonly involving mother-child pairs) exist, it is not clear if they provide the same advantage as trio methods in protecting against population stratification, or if a specific dyad design (e.g., case-mother dyads vs. case-mother/control-mother dyads) is more advantageous. In this article, we review existing statistical methods for analyzing genome-wide marker data on dyads and perform extensive simulation experiments to benchmark their type I errors and statistical power under different scenarios. We extend our evaluation to existing methods for analyzing a combination of case-parent trios and dyads together. We apply these methods on genotyped and imputed data from multiethnic mother-child pairs only, case-parent trios only or combinations of both dyads and trios from the Gene, Environment Association Studies consortium (GENEVA), where each family was ascertained through a child affected by nonsyndromic cleft lip with or without cleft palate. Results from the GENEVA study corroborate the findings from our simulation experiments. Finally, we provide recommendations for using statistical genetic association methods for dyads.
Collapse
Affiliation(s)
- Debashree Ray
- Department of Epidemiology, Bloomberg School of Public HealthJohns Hopkins UniversityBaltimoreMarylandUSA
- Department of Biostatistics, Bloomberg School of Public HealthJohns Hopkins UniversityBaltimoreMarylandUSA
| | - Candelaria Vergara
- Department of Epidemiology, Bloomberg School of Public HealthJohns Hopkins UniversityBaltimoreMarylandUSA
| | - Margaret A. Taub
- Department of Biostatistics, Bloomberg School of Public HealthJohns Hopkins UniversityBaltimoreMarylandUSA
| | - Genevieve Wojcik
- Department of Epidemiology, Bloomberg School of Public HealthJohns Hopkins UniversityBaltimoreMarylandUSA
| | - Christine Ladd‐Acosta
- Department of Epidemiology, Bloomberg School of Public HealthJohns Hopkins UniversityBaltimoreMarylandUSA
| | - Terri H. Beaty
- Department of Epidemiology, Bloomberg School of Public HealthJohns Hopkins UniversityBaltimoreMarylandUSA
| | - Priya Duggal
- Department of Epidemiology, Bloomberg School of Public HealthJohns Hopkins UniversityBaltimoreMarylandUSA
| |
Collapse
|
14
|
Zhuang W, Camacho L, Silva CS, Thomson M, Snyder K. A robust biostatistical method leverages informative but uncertainly determined qPCR data for biomarker detection, early diagnosis, and treatment. PLoS One 2022; 17:e0263070. [PMID: 35100319 PMCID: PMC8803186 DOI: 10.1371/journal.pone.0263070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 01/11/2022] [Indexed: 11/19/2022] Open
Abstract
As a common medium-throughput technique, qPCR (quantitative real-time polymerase chain reaction) is widely used to measure levels of nucleic acids. In addition to accurate and complete data, experimenters have unavoidably observed some incomplete and uncertainly determined qPCR data because of intrinsically low overall amounts of biological materials, such as nucleic acids present in biofluids. When there are samples with uncertainly determined qPCR data, some investigators apply the statistical complete-case method by excluding the subset of samples with uncertainly determined data from analysis (CO), while others simply choose not to analyze (CNA) these datasets altogether. To include as many observations as possible in analysis for interesting differential changes between groups, some investigators set incomplete observations equal to the maximum quality qPCR cycle (MC), such as 32 and 40. Although straightforward, these methods may decrease the sample size, skew the data distribution, and compromise statistical power and research reproducibility across replicate qPCR studies. To overcome the shortcomings of the existing, commonly-used qPCR data analysis methods and to join the efforts in advancing statistical analysis in rigorous preclinical research, we propose a robust nonparametric statistical cycle-to-threshold method (CTOT) to analyze incomplete qPCR data for two-group comparisons. CTOT incorporates important characteristics of qPCR data and time-to-event statistical methodology, resulting in a novel analytical method for qPCR data that is built around good quality data from all subjects, certainly determined or not. Considering the benchmark full data (BFD), we compared the abilities of CTOT, CO, MC, and CNA statistical methods to detect interesting differential changes between groups with informative but uncertainly determined qPCR data. Our simulations and applications show that CTOT improves the power of detecting and confirming differential changes in many situations over the three commonly used methods without excess type I errors. The robust nonparametric statistical method of CTOT helps leverage qPCR technology and increase the power to detect differential changes that may assist decision making with respect to biomarker detection and early diagnosis, with the goal of improving the management of patient healthcare.
Collapse
Affiliation(s)
- Wei Zhuang
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, Arkansas, United States of America
| | - Luísa Camacho
- Division of Biochemical Toxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, Arkansas, United States of America
| | - Camila S. Silva
- Division of Biochemical Toxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, Arkansas, United States of America
| | - Michael Thomson
- Office of New Drugs, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, United States of America
| | - Kevin Snyder
- Office of New Drugs, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, United States of America
| |
Collapse
|
15
|
Controlling for background genetic effects using polygenic scores improves the power of genome-wide association studies. Sci Rep 2021; 11:19571. [PMID: 34599249 PMCID: PMC8486788 DOI: 10.1038/s41598-021-99031-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 09/14/2021] [Indexed: 11/18/2022] Open
Abstract
Ongoing increases in the size of human genotype and phenotype collections offer the promise of improved understanding of the genetics of complex diseases. In addition to the biological insights that can be gained from the nature of the variants that contribute to the genetic component of complex trait variability, these data bring forward the prospect of predicting complex traits and the risk of complex genetic diseases from genotype data. Here we show that advances in phenotype prediction can be applied to improve the power of genome-wide association studies. We demonstrate a simple and efficient method to model genetic background effects using polygenic scores derived from SNPs that are not on the same chromosome as the target SNP. Using simulated and real data we found that this can result in a substantial increase in the number of variants passing genome-wide significance thresholds. This increase in power to detect trait-associated variants also translates into an increase in the accuracy with which the resulting polygenic score predicts the phenotype from genotype data. Our results suggest that advances in methods for phenotype prediction can be exploited to improve the control of background genetic effects, leading to more accurate GWAS results and further improvements in phenotype prediction.
Collapse
|
16
|
Yilmaz O, Kizilaslan M, Arzik Y, Behrem S, Ata N, Karaca O, Elmaci C, Cemal I. Genome-wide association studies of preweaning growth and in vivo carcass composition traits in Esme sheep. J Anim Breed Genet 2021; 139:26-39. [PMID: 34331347 DOI: 10.1111/jbg.12640] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 07/06/2021] [Accepted: 07/22/2021] [Indexed: 01/15/2023]
Abstract
Sheep are considered as a major contributor of global food security. Moreover, sheep preweaning growth traits as well as in vivo carcass composition traits such as ultrasonic measurements of Longissimus dorsi muscle depth (UMD) and back-fat thickness (UFD) are crucially important indicators of meat yield and hot carcass composition. Despite their relative importance for productivity and profitability of a sheep production system, detected QTL for these traits are quite scarce. Therefore, we implemented GWAS for these traits using animal mixed model-based association approach provided by GenABEL in Esme sheep. Three genome-wide and 14 individual chromosome-wide associated SNPs were discovered. As a result, ESRP1, LOC105613082, ZNF641, DUSP5, TEAD1, SMOX, PTPRT, RALYL, POM121C, PHIP, LOC101106051, ZIM3, PEG3, TRPC7, FBXL4, LOC105610397, LOC105616489 and DNAAF2 were suggested as candidates. Some of the discovered genes and involved pathways were already annotated to contribute growth and development in various species including human, mice and cattle. All in all, the results of this study are expected to strongly contribute to shed a light on the underlying molecular mechanisms behind growth and carcass composition traits, with potential implications on studies aiming faster genetic improvement, targeted low-resolution SNP panel designs and genome-editing studies.
Collapse
Affiliation(s)
- Onur Yilmaz
- Department of Animal Science, Faculty of Agriculture, Aydin Adnan Menderes University, Aydin, Turkey
| | - Mehmet Kizilaslan
- Department of Animal Breeding and Genetics, International Center for Livestock Research and Training, Ankara, Turkey
| | - Yunus Arzik
- Department of Animal Breeding and Genetics, International Center for Livestock Research and Training, Ankara, Turkey
| | - Sedat Behrem
- Department of Animal Breeding and Genetics, International Center for Livestock Research and Training, Ankara, Turkey
| | - Nezih Ata
- Department of Animal Science, Faculty of Agriculture, Aydin Adnan Menderes University, Aydin, Turkey
| | - Orhan Karaca
- Department of Animal Science, Faculty of Agriculture, Aydin Adnan Menderes University, Aydin, Turkey
| | - Cengiz Elmaci
- Animal Science Department, Agriculture Faculty, Bursa Uludag University, Bursa, Turkey
| | - Ibrahim Cemal
- Department of Animal Science, Faculty of Agriculture, Aydin Adnan Menderes University, Aydin, Turkey
| |
Collapse
|
17
|
Li LY, Xiao SJ, Tu JM, Zhang ZK, Zheng H, Huang LB, Huang ZY, Yan M, Liu XD, Guo YM. A further survey of the quantitative trait loci affecting swine body size and carcass traits in five related pig populations. Anim Genet 2021; 52:621-632. [PMID: 34182604 DOI: 10.1111/age.13112] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/16/2021] [Indexed: 12/13/2022]
Abstract
Breeding for good meat quality performance while maintaining large body size and desirable carcass traits has been the major challenge for modern swine selective breeding. To address this goal, in the present work we studied five related populations produced by two commercial breeds (Berkshire and Duroc) and two Chinese breeds (Licha black pig and Lulai black pig). A single-trait GWAS performed on 20 body size and carcass traits using a self-developed China Chip-1 porcine SNP50K BeadChip identified 11 genome-wide significant QTL on nine chromosomes and 22 suggestive QTL on 15 chromosomes. For the 11 genome-wide significant QTL, eight were detected in at least two populations, and the rest were population-specific and only mapped in Shanxia black pig. Most of the genome-wide significant QTL were pleiotropic; for example, the QTL around 75.65 Mb on SSC4 was associated with four traits at genome-wide significance level. After screening the genes within 50 kb of the top SNP for each genome-wide significant QTL, NR6A1 and VRTN were chosen as candidate genes for vertebrae number; PLAG1 and BMP2 were identified as candidate genes for body size; and MC4R was the strong candidate gene for body weight. The four genes have been reported as candidates for thoracic vertebrae number, lumbar vertebrae number, carcass length and body weight respectively in previous studies. The effects of VRTN on thoracic vertebrae number, carcass length and body length have been verified in Shanxia black pig. Therefore, the VRTN genotype could be used in gene-assisted selection, and this could accelerate genetic improvement of body size and carcass traits in Shanxia black pig.
Collapse
Affiliation(s)
- L-Y Li
- State Key Laboratory of Pig Genetic Improvement and Production Technology, Jiangxi Agricultural University, Nanchang, Jiangxi, 330045, China
| | - S-J Xiao
- State Key Laboratory of Pig Genetic Improvement and Production Technology, Jiangxi Agricultural University, Nanchang, Jiangxi, 330045, China
| | - J-M Tu
- Jiangxi Shanxia Swine Genetic Investment Company Limited, Dingnan, Jiangxi, 341900, China
| | - Z-K Zhang
- State Key Laboratory of Pig Genetic Improvement and Production Technology, Jiangxi Agricultural University, Nanchang, Jiangxi, 330045, China
| | - H Zheng
- State Key Laboratory of Pig Genetic Improvement and Production Technology, Jiangxi Agricultural University, Nanchang, Jiangxi, 330045, China.,Jiangxi Shanxia Swine Genetic Investment Company Limited, Dingnan, Jiangxi, 341900, China
| | - L-B Huang
- Jiangxi Shanxia Swine Genetic Investment Company Limited, Dingnan, Jiangxi, 341900, China
| | - Z-Y Huang
- Jiangxi Shanxia Swine Genetic Investment Company Limited, Dingnan, Jiangxi, 341900, China
| | - M Yan
- State Key Laboratory of Pig Genetic Improvement and Production Technology, Jiangxi Agricultural University, Nanchang, Jiangxi, 330045, China
| | - X-D Liu
- State Key Laboratory of Pig Genetic Improvement and Production Technology, Jiangxi Agricultural University, Nanchang, Jiangxi, 330045, China
| | - Y-M Guo
- State Key Laboratory of Pig Genetic Improvement and Production Technology, Jiangxi Agricultural University, Nanchang, Jiangxi, 330045, China
| |
Collapse
|
18
|
Martínez P, Robledo D, Taboada X, Blanco A, Moser M, Maroso F, Hermida M, Gómez-Tato A, Álvarez-Blázquez B, Cabaleiro S, Piferrer F, Bouza C, Lien S, Viñas AM. A genome-wide association study, supported by a new chromosome-level genome assembly, suggests sox2 as a main driver of the undifferentiatiated ZZ/ZW sex determination of turbot (Scophthalmus maximus). Genomics 2021; 113:1705-1718. [PMID: 33838278 DOI: 10.1016/j.ygeno.2021.04.007] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 03/20/2021] [Accepted: 04/05/2021] [Indexed: 01/10/2023]
Abstract
BACKGROUND Understanding sex determination (SD) across taxa is a major challenge for evolutionary biology. The new genomic tools are paving the way to identify genomic features underlying SD in fish, a group frequently showing limited sex chromosome differentiation and high SD evolutionary turnover. Turbot (Scophthalmus maximus) is a commercially important flatfish with an undifferentiated ZW/ZZ SD system and remarkable sexual dimorphism. Here we describe a new long-read turbot genome assembly used to disentangle the genetic architecture of turbot SD by combining genomics and classical genetics approaches. RESULTS The new turbot genome assembly consists of 145 contigs (N50 = 22.9 Mb), 27 of them representing >95% of its estimated genome size. A genome wide association study (GWAS) identified a ~ 6.8 Mb region on chromosome 12 associated with sex in 69.4% of the 36 families analyzed. The highest associated markers flanked sox2, the only gene in the region showing differential expression between sexes before gonad differentiation. A single SNP showed consistent differences between Z and W chromosomes. The analysis of a broad sample of families suggested the presence of additional genetic and/or environmental factors on turbot SD. CONCLUSIONS The new chromosome-level turbot genome assembly, one of the most contiguous fish assemblies to date, facilitated the identification of sox2 as a consistent candidate gene putatively driving SD in this species. This chromosome SD system barely showed any signs of differentiation, and other factors beyond the main QTL seem to control SD in a certain proportion of families.
Collapse
Affiliation(s)
- Paulino Martínez
- Departament of Zoology, Genetics and Physical Anthropology, Faculty of Veterinary, Universidade de Santiago de Compostela, Campus de Lugo, 27002 Lugo, Spain.
| | - Diego Robledo
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian EH25 9RG, UK.
| | - Xoana Taboada
- Departament of Zoology, Genetics and Physical Anthropology, Faculty of Biology, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Andrés Blanco
- Departament of Zoology, Genetics and Physical Anthropology, Faculty of Veterinary, Universidade de Santiago de Compostela, Campus de Lugo, 27002 Lugo, Spain.
| | - Michel Moser
- Centre for Integrative Genetics, Department of Animal and Aquacultural Sciences, Faculty of Biosciences, Norwegian University of Life Sciences, Ås, Norway.
| | - Francesco Maroso
- Department of Life Science and Biotechnology, University of Ferrara, 44121 Ferrara, Italy
| | - Miguel Hermida
- Departament of Zoology, Genetics and Physical Anthropology, Faculty of Veterinary, Universidade de Santiago de Compostela, Campus de Lugo, 27002 Lugo, Spain.
| | - Antonio Gómez-Tato
- Departament of Mathematics, Faculty of Mathematics, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain.
| | - Blanca Álvarez-Blázquez
- Instituto Español de Oceanografía (IEO), Centro Oceanográfico de Vigo, Cabo Estay-Canido, 36280 Vigo, Spain.
| | - Santiago Cabaleiro
- Cluster de Acuicultura de Galicia (Punta do Couso), Aguiño-Ribeira, 15695 A Coruña, Spain.
| | - Francesc Piferrer
- Institut de Ciències del Mar, Consejo Superior de Investigaciones Científicas (CSIC), 08003 Barcelona, Spain.
| | - Carmen Bouza
- Departament of Zoology, Genetics and Physical Anthropology, Faculty of Veterinary, Universidade de Santiago de Compostela, Campus de Lugo, 27002 Lugo, Spain.
| | - Sigbjørn Lien
- Centre for Integrative Genetics, Department of Animal and Aquacultural Sciences, Faculty of Biosciences, Norwegian University of Life Sciences, Ås, Norway.
| | - Ana M Viñas
- Departament of Zoology, Genetics and Physical Anthropology, Faculty of Biology, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain.
| |
Collapse
|
19
|
Ziyatdinov A, Kim J, Prokopenko D, Privé F, Laporte F, Loh PR, Kraft P, Aschard H. Estimating the effective sample size in association studies of quantitative traits. G3-GENES GENOMES GENETICS 2021; 11:6178001. [PMID: 33734375 PMCID: PMC8495748 DOI: 10.1093/g3journal/jkab057] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 01/06/2021] [Indexed: 01/08/2023]
Abstract
The effective sample size (ESS) is a metric used to summarize in a single term the amount of correlation in a sample. It is of particular interest when predicting the statistical power of genome-wide association studies (GWAS) based on linear mixed models. Here, we introduce an analytical form of the ESS for mixed-model GWAS of quantitative traits and relate it to empirical estimators recently proposed. Using our framework, we derived approximations of the ESS for analyses of related and unrelated samples and for both marginal genetic and gene-environment interaction tests. We conducted simulations to validate our approximations and to provide a quantitative perspective on the statistical power of various scenarios, including power loss due to family relatedness and power gains due to conditioning on the polygenic signal. Our analyses also demonstrate that the power of gene-environment interaction GWAS in related individuals strongly depends on the family structure and exposure distribution. Finally, we performed a series of mixed-model GWAS on data from the UK Biobank and confirmed the simulation results. We notably found that the expected power drop due to family relatedness in the UK Biobank is negligible.
Collapse
Affiliation(s)
- Andrey Ziyatdinov
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Jihye Kim
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Dmitry Prokopenko
- Genetics and Aging Unit and McCance Center for Brain Health, Department of Neurology, Massachusetts General Hospital, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA
| | - Florian Privé
- National Centre for Register-Based Research, Aarhus University, Aarhus, 8210, Denmark
| | - Fabien Laporte
- Department of Computational Biology-USR 3756 CNRS, Institut Pasteur, 75015 Paris, France
| | - Po-Ru Loh
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.,Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Peter Kraft
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Hugues Aschard
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA.,Department of Computational Biology-USR 3756 CNRS, Institut Pasteur, 75015 Paris, France
| |
Collapse
|
20
|
Moraleda CP, Robledo D, Gutiérrez AP, Del-Pozo J, Yáñez JM, Houston RD. Investigating mechanisms underlying genetic resistance to Salmon Rickettsial Syndrome in Atlantic salmon using RNA sequencing. BMC Genomics 2021; 22:156. [PMID: 33676414 PMCID: PMC7936450 DOI: 10.1186/s12864-021-07443-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 02/11/2021] [Indexed: 12/29/2022] Open
Abstract
Background Salmon Rickettsial Syndrome (SRS), caused by Piscirickettsia salmonis, is one of the primary causes of morbidity and mortality in Atlantic salmon aquaculture, particularly in Chile. Host resistance is a heritable trait, and functional genomic studies have highlighted genes and pathways important in the response of salmon to the bacteria. However, the functional mechanisms underpinning genetic resistance are not yet well understood. In the current study, a large population of salmon pre-smolts were challenged with P. salmonis, with mortality levels recorded and samples taken for genotyping. In parallel, head kidney and liver samples were taken from animals of the same population with high and low genomic breeding values for resistance, and used for RNA-Sequencing to compare their transcriptome profile both pre and post infection. Results A significant and moderate heritability (h2 = 0.43) was shown for the trait of binary survival. Genome-wide association analyses using 38 K imputed SNP genotypes across 2265 animals highlighted that resistance is a polygenic trait. Several thousand genes were identified as differentially expressed between controls and infected samples, and enriched pathways related to the host immune response were highlighted. In addition, several networks with significant correlation with SRS resistance breeding values were identified, suggesting their involvement in mediating genetic resistance. These included apoptosis, cytoskeletal organisation, and the inflammasome. Conclusions While resistance to SRS is a polygenic trait, this study has highlighted several relevant networks and genes that are likely to play a role in mediating genetic resistance. These genes may be future targets for functional studies, including genome editing, to further elucidate their role underpinning genetic variation in host resistance. Supplementary Information The online version contains supplementary material available at 10.1186/s12864-021-07443-2.
Collapse
Affiliation(s)
- Carolina P Moraleda
- The Roslin Institute and Royal (Dick) School of Veterinary Sciences, The University of Edinburgh, Edinburgh, UK
| | - Diego Robledo
- The Roslin Institute and Royal (Dick) School of Veterinary Sciences, The University of Edinburgh, Edinburgh, UK
| | - Alejandro P Gutiérrez
- The Roslin Institute and Royal (Dick) School of Veterinary Sciences, The University of Edinburgh, Edinburgh, UK
| | - Jorge Del-Pozo
- The Roslin Institute and Royal (Dick) School of Veterinary Sciences, The University of Edinburgh, Edinburgh, UK
| | - José M Yáñez
- Faculty of Veterinary and Livestock Sciences, University of Chile, Santiago, Chile.
| | - Ross D Houston
- The Roslin Institute and Royal (Dick) School of Veterinary Sciences, The University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
21
|
Liu Y, Ran S, Lin Y, Zhang YX, Yang XL, Wei XT, Jiang ZX, He X, Zhang H, Feng GJ, Shen H, Tian Q, Deng HW, Zhang L, Pei YF. Four pleiotropic loci associated with fat mass and lean mass. Int J Obes (Lond) 2020; 44:2113-2123. [PMID: 32719433 PMCID: PMC7912634 DOI: 10.1038/s41366-020-0645-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 06/23/2020] [Accepted: 07/16/2020] [Indexed: 11/08/2022]
Abstract
BACKGROUND Fat mass and lean mass are two biggest components of body mass. Both fat mass and lean mass are under strong genetic determinants and are correlated. METHODS We performed a bivariate genome-wide association meta-analysis of (lean adjusted) leg fat mass and (fat adjusted) leg lean mass in 12,517 subjects from 6 samples, and followed by in silico replication in large-scale UK biobank cohort sample (N = 370 097). RESULTS We identified four loci that were significant at the genome-wide significance (GWS, α = 5.0 × 10-8) level at the discovery meta-analysis, and successfully replicated in the replication sample: 2q36.3 (rs1024137, pdiscovery = 3.32 × 10-8, preplication = 4.07 × 10-13), 5q13.1 (rs4976033, pdiscovery = 1.93 × 10-9, preplication = 6.35 × 10-7), 12q24.31 (rs4765528, pdiscovery = 7.19 × 10-12, preplication = 1.88 × 10-11) and 18q21.32 (rs371326986, pdiscovery = 9.04 × 10-9, preplication = 2.35 × 10-95). The above four pleiotropic loci may play a pleiotropic role for fat mass and lean mass development. CONCLUSIONS Our findings further enhance the understanding of the genetic association between fat mass and lean mass and provide a new theoretical basis for their understanding.
Collapse
Affiliation(s)
- Yu Liu
- Center for Genetic Epidemiology and Genomics, School of Public Health, Medical College of Soochow University, Jiangsu, PR China
- School of Medical Instruments and Food Engineering, University of Shanghai for Science and Technology, Shanghai, PR China
| | - Shu Ran
- School of Medical Instruments and Food Engineering, University of Shanghai for Science and Technology, Shanghai, PR China
| | - Yong Lin
- School of Medical Instruments and Food Engineering, University of Shanghai for Science and Technology, Shanghai, PR China
| | - Yu-Xue Zhang
- School of Medical Instruments and Food Engineering, University of Shanghai for Science and Technology, Shanghai, PR China
| | - Xiao-Lin Yang
- Center for Genetic Epidemiology and Genomics, School of Public Health, Medical College of Soochow University, Jiangsu, PR China
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Jiangsu, PR China
| | - Xin-Tong Wei
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Jiangsu, PR China
- Department of Epidemiology and Health Statistics, Medical College of Soochow University, Jiangsu, PR China
| | - Zi-Xuan Jiang
- School of Medical Instruments and Food Engineering, University of Shanghai for Science and Technology, Shanghai, PR China
| | - Xiao He
- School of Medical Instruments and Food Engineering, University of Shanghai for Science and Technology, Shanghai, PR China
| | - Hong Zhang
- Center for Genetic Epidemiology and Genomics, School of Public Health, Medical College of Soochow University, Jiangsu, PR China
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Jiangsu, PR China
| | - Gui-Juan Feng
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Jiangsu, PR China
- Department of Epidemiology and Health Statistics, Medical College of Soochow University, Jiangsu, PR China
| | - Hui Shen
- Department of Biostatistics and Bioinformatics, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, USA
| | - Qing Tian
- Department of Biostatistics and Bioinformatics, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, USA
| | - Hong-Wen Deng
- Department of Biostatistics and Bioinformatics, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, USA.
| | - Lei Zhang
- Center for Genetic Epidemiology and Genomics, School of Public Health, Medical College of Soochow University, Jiangsu, PR China.
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Jiangsu, PR China.
| | - Yu-Fang Pei
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Jiangsu, PR China.
- Department of Epidemiology and Health Statistics, Medical College of Soochow University, Jiangsu, PR China.
| |
Collapse
|
22
|
Diao G, Lin DY. Statistically efficient association analysis of quantitative traits with haplotypes and untyped SNPs in family studies. BMC Genet 2020; 21:99. [PMID: 32894040 PMCID: PMC7487716 DOI: 10.1186/s12863-020-00902-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 08/17/2020] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Associations between haplotypes and quantitative traits provide valuable information about the genetic basis of complex human diseases. Haplotypes also provide an effective way to deal with untyped SNPs. Two major challenges arise in haplotype-based association analysis of family data. First, haplotypes may not be inferred with certainty from genotype data. Second, the trait values within a family tend to be correlated because of common genetic and environmental factors. RESULTS To address these challenges, we present an efficient likelihood-based approach to analyzing associations of quantitative traits with haplotypes or untyped SNPs. This approach properly accounts for within-family trait correlations and can handle general pedigrees with arbitrary patterns of missing genotypes. We characterize the genetic effects on the quantitative trait by a linear regression model with random effects and develop efficient likelihood-based inference procedures. Extensive simulation studies are conducted to examine the performance of the proposed methods. An application to family data from the Childhood Asthma Management Program Ancillary Genetic Study is provided. A computer program is freely available. CONCLUSIONS Results from extensive simulation studies show that the proposed methods for testing the haplotype effects on quantitative traits have correct type I error rates and are more powerful than some existing methods.
Collapse
Affiliation(s)
- Guoqing Diao
- Department of Biostatistics and Bioinformatics, The George Washington University, Washington, District of Columbia, USA.
| | - Dan-Yu Lin
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
23
|
Carlson JC, Weeks DE, Hawley NL, Sun G, Cheng H, Naseri T, Reupena MS, Tuitele J, Deka R, McGarvey ST, Minster RL. Genome-wide association studies in Samoans give insight into the genetic architecture of fasting serum lipid levels. J Hum Genet 2020; 66:111-121. [PMID: 32759990 DOI: 10.1038/s10038-020-0816-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 07/16/2020] [Accepted: 07/20/2020] [Indexed: 11/09/2022]
Abstract
The current understanding of the genetic architecture of lipids has largely come from genome-wide association studies (GWAS). To date, few GWAS have examined the genetic architecture of lipids in Polynesians, and none have in Samoans, whose unique population history, including many population bottlenecks, may provide insight into the biological foundations of variation in lipid levels. Here we performed a GWAS of four fasting serum lipid levels: total cholesterol (TC), high-density lipoprotein (HDL), low-density lipoprotein (LDL), and triglycerides (TG) in a sample of 2849 Samoans, with validation genotyping for associations in a replication cohort comprising 1798 Samoans and American Samoans. We identified multiple genome-wide significant associations (P < 5 × 10-8) previously seen in other populations-APOA1 with TG, CETP with HDL, and APOE with TC and LDL-and several suggestive associations (P < 1 × 10-5), including an association of variants downstream of MGAT1 and RAB21 with HDL. However, we observed different association signals for variants near APOE than what has been previously reported in non-Polynesian populations. The association with several known lipid loci combined with the newly identified associations with variants near MGAT1 and RAB21 suggest that while some of the genetic architecture of lipids is shared between Samoans and other populations, part of the genetic architecture may be Polynesian-specific.
Collapse
Affiliation(s)
- Jenna C Carlson
- Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA.,Department of Biostatistics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Daniel E Weeks
- Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA.,Department of Biostatistics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Nicola L Hawley
- Department of Chronic Disease Epidemiology, Yale School of Public Health, New Haven, CT, USA
| | - Guangyun Sun
- Department of Environmental Health, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Hong Cheng
- Department of Environmental Health, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Take Naseri
- Ministry of Health, Government of Samoa, Apia, Samoa
| | | | - John Tuitele
- Department of Public Health, Government of American Samoa, Pago Pago, AS, USA
| | - Ranjan Deka
- Department of Environmental Health, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Stephen T McGarvey
- International Health Institute and Department of Epidemiology, School of Public Health, Brown University, Providence, RI, USA.,Department of Anthropology, Brown University, Providence, RI, USA
| | - Ryan L Minster
- Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
24
|
Neamatallah M, El-Bendary M, Elalfy H, Besheer T, El-Maksoud MA, Elhammady D, Abed S, Elegezy M, Kandeel L, Eldeib D, Mousa N, Abd El-Hafeez M, El-Gilany AH, Esmat G. Impact of Toll-like Receptors 2(TLR2) and TLR 4 Gene Variations on HCV Susceptibility, Response to Treatment and Development of Hepatocellular Carcinoma in Cirrhotic HCV Patients. Immunol Invest 2020; 49:462-476. [PMID: 31615295 DOI: 10.1080/08820139.2019.1673772] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Background and Aims: Genetic polymorphisms of Toll-like receptors (TLRs) have been proposed to affect susceptibility to HCV infection and progression to end-stage liver disease. This study was conducted to clarify the association of SNPS of TLR2 and TLR4 with clinical outcome of hepatitis C, response to treatment and development of HCC.Methods: The current study examined 3295 individuals from 725 families that were categorized into groups comprising chronic HCV (CH), spontaneous viral clearance (SC) and control subjects. Treated patients were classified into responders (RT) and non-responders (NRT). In addition, patients with liver cirrhotic (LC), and hepatocellular carcinoma (HCC) were also included. All subjects were genotyped for five single nucleotide polymorphisms (SNPs) of TLR2 and four SNPs of TLR4 and their haplotypes using allelic discrimination real-time PCR.Results: Results demonstrated strong association with allele A of rs13105517 of TLR2 and allele C of rs10116253 of TLR4 with CH in comparison to SC group. However, The peak of risk of HCC was observed with allele C of rs3804099 of TLR2 and C allele of rs10116253 TLR4 (p < 0.001).A strong association was found with allele T of rs1816702 of TLR2 and allele A of rs5030728 of TLR4 in non responder group in comparison to responders (p < 0.001). Haplotypes CAGT of TLR4 and ATAC of TLR2 showed significant association with CH and HCC groups in comparison to other groups.Conclusions: This study shows an association of minor alleles of TLR2 and TLR4 with outcome of HCV infection, response to therapy and development of HCC in cirrhotic patients.
Collapse
Affiliation(s)
- Mustafa Neamatallah
- Medical Biochemistry Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Mahmoud El-Bendary
- Tropical Medicine and Hepatology Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Hatem Elalfy
- Tropical Medicine and Hepatology Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Tarek Besheer
- Tropical Medicine and Hepatology Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Mohamed Abd El-Maksoud
- Tropical Medicine and Hepatology Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Dina Elhammady
- Tropical Medicine and Hepatology Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Sally Abed
- Tropical Medicine and Hepatology Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Mohamed Elegezy
- Tropical Medicine and Hepatology Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Lamiaa Kandeel
- Ministry of Health, Sherbeen General Hospital, Sherbeen city, Egypt
| | - Dalia Eldeib
- Biochemistry department, Faculty of Science, Mansoura University, Mansoura, Egypt
| | - Naser Mousa
- Tropical Medicine and Hepatology Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | | | - Abdel-Hady El-Gilany
- Public Health Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Gamal Esmat
- Tropical Medicine and Hepatology Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| |
Collapse
|
25
|
Sosa‐Madrid BS, Hernández P, Blasco A, Haley CS, Fontanesi L, Santacreu MA, Pena RN, Navarro P, Ibáñez‐Escriche N. Genomic regions influencing intramuscular fat in divergently selected rabbit lines. Anim Genet 2020; 51:58-69. [PMID: 31696970 PMCID: PMC7004202 DOI: 10.1111/age.12873] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/04/2019] [Indexed: 12/12/2022]
Abstract
Intramuscular fat (IMF) is one of the main meat quality traits for breeding programmes in livestock species. The main objective of this study was to identify genomic regions associated with IMF content comparing two rabbit populations divergently selected for this trait, and to generate a list of putative candidate genes. Animals were genotyped using the Affymetrix Axiom OrcunSNP Array (200k). After quality control, the data involved 477 animals and 93 540 SNPs. Two methods were used in this research: single marker regressions with the data adjusted by genomic relatedness, and a Bayesian multiple marker regression. Associated genomic regions were located on the rabbit chromosomes (OCU) OCU1, OCU8 and OCU13. The highest value for the percentage of the genomic variance explained by a genomic region was found in two consecutive genomic windows on OCU8 (7.34%). Genes in the associated regions of OCU1 and OCU8 presented biological functions related to the control of adipose cell function, lipid binding, transportation and localisation (APOLD1, PLBD1, PDE6H, GPRC5D and GPRC5A) and lipid metabolic processes (MTMR2). The EWSR1 gene, underlying the OCU13 region, is linked to the development of brown adipocytes. The findings suggest that there is a large component of polygenic effect behind the differences in IMF content in these two lines, as the variance explained by most of the windows was low. The genomic regions of OCU1, OCU8 and OCU13 revealed novel candidate genes. Further studies would be needed to validate the associations and explore their possible application in selection programmes.
Collapse
Affiliation(s)
- Bolívar S. Sosa‐Madrid
- Institute for Animal Science and TechnologyUniversitat Politècnica de València46022 ValenciaSpain
| | - Pilar Hernández
- Institute for Animal Science and TechnologyUniversitat Politècnica de València46022 ValenciaSpain
| | - Agustín Blasco
- Institute for Animal Science and TechnologyUniversitat Politècnica de València46022 ValenciaSpain
| | - Chris S. Haley
- MRC Human Genetics UnitMRC Institute of Genetics and Molecular MedicineUniversity of EdinburghCrewe Road, Edinburgh EH4 2XUUnited Kingdom
- Roslin Institute and Royal (Dick) School of Veterinary StudiesUniversity of EdinburghMidlothian EH25 9RGUnited Kingdom
| | - Luca Fontanesi
- Division of Animal SciencesDepartment of Agricultural and Food SciencesUniversity of Bologna40127 BolognaItaly
| | - María A. Santacreu
- Institute for Animal Science and TechnologyUniversitat Politècnica de València46022 ValenciaSpain
| | - Romi N. Pena
- Departament de Ciència AnimalUniversitat de Lleida–Agrotecnio CentreE-25198 LleidaCatalonia, Spain
| | - Pau Navarro
- MRC Human Genetics UnitMRC Institute of Genetics and Molecular MedicineUniversity of EdinburghCrewe Road, Edinburgh EH4 2XUUnited Kingdom
| | - Noelia Ibáñez‐Escriche
- Institute for Animal Science and TechnologyUniversitat Politècnica de València46022 ValenciaSpain
| |
Collapse
|
26
|
Mikkola L, Holopainen S, Pessa-Morikawa T, Lappalainen AK, Hytönen MK, Lohi H, Iivanainen A. Genetic dissection of canine hip dysplasia phenotypes and osteoarthritis reveals three novel loci. BMC Genomics 2019; 20:1027. [PMID: 31881848 PMCID: PMC6935090 DOI: 10.1186/s12864-019-6422-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 12/22/2019] [Indexed: 12/15/2022] Open
Abstract
Background Hip dysplasia and osteoarthritis continue to be prevalent problems in veterinary and human medicine. Canine hip dysplasia is particularly problematic as it massively affects several large-sized breeds and can cause a severe impairment of the quality of life. In Finland, the complex condition is categorized to five classes from normal to severe dysplasia, but the categorization includes several sub-traits: congruity of the joint, Norberg angle, subluxation degree of the joint, shape and depth of the acetabulum, and osteoarthritis. Hip dysplasia and osteoarthritis have been proposed to have separate genetic etiologies. Results Using Fédération Cynologique Internationale -standardized ventrodorsal radiographs, German shepherds were rigorously phenotyped for osteoarthritis, and for joint incongruity by Norberg angle and femoral head center position in relation to dorsal acetabular edge. The affected dogs were categorized into mild, moderate and severe dysplastic phenotypes using official hip scores. Three different genome-wide significant loci were uncovered. The strongest candidate genes for hip joint incongruity were noggin (NOG), a bone and joint developmental gene on chromosome 9, and nanos C2HC-type zinc finger 1 (NANOS1), a regulator of matrix metalloproteinase 14 (MMP14) on chromosome 28. Osteoarthritis mapped to a long intergenic region on chromosome 1, between genes encoding for NADPH oxidase 3 (NOX3), an intriguing candidate for articular cartilage degradation, and AT-rich interactive domain 1B (ARID1B) that has been previously linked to joint laxity. Conclusions Our findings highlight the complexity of canine hip dysplasia phenotypes. In particular, the results of this study point to the potential involvement of specific and partially distinct loci and genes or pathways in the development of incongruity, mild dysplasia, moderate-to-severe dysplasia and osteoarthritis of canine hip joints. Further studies should unravel the unique and common mechanisms for the various sub-traits.
Collapse
Affiliation(s)
- Lea Mikkola
- Department of Veterinary Biosciences, University of Helsinki, P.O. Box 66 (Mustialankatu 1), FI-00014, Helsinki, Finland.,Department of Medical and Clinical Genetics, University of Helsinki, Helsinki, Finland.,Folkhälsan Research Center, Helsinki, Finland
| | - Saila Holopainen
- Department of Veterinary Biosciences, University of Helsinki, P.O. Box 66 (Mustialankatu 1), FI-00014, Helsinki, Finland.,Department of Medical and Clinical Genetics, University of Helsinki, Helsinki, Finland.,Folkhälsan Research Center, Helsinki, Finland.,Department of Equine and Small Animal Medicine, University of Helsinki, Helsinki, Finland
| | - Tiina Pessa-Morikawa
- Department of Veterinary Biosciences, University of Helsinki, P.O. Box 66 (Mustialankatu 1), FI-00014, Helsinki, Finland
| | - Anu K Lappalainen
- Department of Equine and Small Animal Medicine, University of Helsinki, Helsinki, Finland
| | - Marjo K Hytönen
- Department of Veterinary Biosciences, University of Helsinki, P.O. Box 66 (Mustialankatu 1), FI-00014, Helsinki, Finland.,Department of Medical and Clinical Genetics, University of Helsinki, Helsinki, Finland.,Folkhälsan Research Center, Helsinki, Finland
| | - Hannes Lohi
- Department of Veterinary Biosciences, University of Helsinki, P.O. Box 66 (Mustialankatu 1), FI-00014, Helsinki, Finland.,Department of Medical and Clinical Genetics, University of Helsinki, Helsinki, Finland.,Folkhälsan Research Center, Helsinki, Finland
| | - Antti Iivanainen
- Department of Veterinary Biosciences, University of Helsinki, P.O. Box 66 (Mustialankatu 1), FI-00014, Helsinki, Finland.
| |
Collapse
|
27
|
Rowland K, Persia ME, Rothschild MF, Schmidt C, Lamont SJ. Venous blood gas and chemistry components are moderately heritable in commercial white egg-laying hens under acute or chronic heat exposure. Poult Sci 2019; 98:3426-3430. [PMID: 31002114 PMCID: PMC6698185 DOI: 10.3382/ps/pez204] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 03/21/2019] [Indexed: 11/20/2022] Open
Abstract
Heat stress has a large negative impact on poultry around the world in both intensive and small-scale production systems. Better understanding of genetic factors contributing to response to high ambient temperatures would provide a basis to develop strategies for alleviating negative impacts of heat on poultry production. The objective of this work was to characterize the genetic control (heritability estimate and quantitative trait loci (QTL)) of blood chemistry components before and after exposure to acute and chronic high ambient temperature in a commercial egg laying line Hy-Line W-36 female parent line mature hens were exposed to 4 wk of daily cyclic heat exposure. Blood was collected pre-heat, on the first day of heat, and 2 and 4 wk post heat initiation and analyzed immediately using an i-STAT® hand-held blood analyzer. Thirteen blood components were quantified at the 4 time points: pH, pCO2, pO2, HCO3, TCO2, sO2, iCa, Na, K, base excess, glucose, "hematocrit" (estimated from blood electrical conductivity, BEC), and "hemoglobin" (calculated from BEC). Heritabilities were estimated using genomic relationship information obtained from 600k SNP chip data. All 13 parameters exhibited a significant change after 5 h of heat exposure and most did not return to pre-heat levels throughout the duration of the study. Eight parameters (base excess, glucose, hemoglobin, HCO3, hematocrit, K, pCO2, TCO2) had heritability estimates differing from zero at one or more time points (0.21 to 0.45). The traits with significant heritability would be good candidates for use as biomarkers in a selection program if they are correlated with traits of economic importance that are more difficult to measure. QTL were identified for nine of the traits at one or more time point. These nine traits, however, did not have significant heritability estimates suggesting that while some QTL have been identified their effects are generally small.
Collapse
Affiliation(s)
- Kaylee Rowland
- Department of Animal Science, Iowa State University, Ames, IA 50011
| | - Michael E Persia
- Virginia Tech, Department of Animal and Poultry Sciences, Blacksburg, VA 24061
| | - Max F Rothschild
- Department of Animal Science, Iowa State University, Ames, IA 50011
| | - Carl Schmidt
- University of Delaware, Animal and Food Sciences, Newark, DE 19716
| | - Susan J Lamont
- Department of Animal Science, Iowa State University, Ames, IA 50011
| |
Collapse
|
28
|
Bojnord NR, Honarvar M, Aminafshara M, Kashan NEJ. Imputation of non-genotyped individuals using their genotyped progeny implementing machine learning algorithm. GENE REPORTS 2019. [DOI: 10.1016/j.genrep.2019.100435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
29
|
Kess T, Boulding EG. Genome-wide association analyses reveal polygenic genomic architecture underlying divergent shell morphology in Spanish Littorina saxatilis ecotypes. Ecol Evol 2019; 9:9427-9441. [PMID: 31534666 PMCID: PMC6745682 DOI: 10.1002/ece3.5378] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 05/24/2019] [Indexed: 12/18/2022] Open
Abstract
Gene flow between diverging populations experiencing dissimilar ecological conditions can theoretically constrain adaptive evolution. To minimize the effect of gene flow, alleles underlying traits essential for local adaptation are predicted to be located in linked genome regions with reduced recombination. Local reduction in gene flow caused by selection is expected to produce elevated divergence in these regions. The highly divergent crab-adapted and wave-adapted ecotypes of the marine snail Littorina saxatilis present a model system to test these predictions. We used genome-wide association (GWA) analysis of geometric morphometric shell traits associated with microgeographic divergence between the two L. saxatilis ecotypes within three separate sampling sites. A total of 477 snails that had individual geometric morphometric data and individual genotypes at 4,066 single nucleotide polymorphisms (SNPs) were analyzed using GWA methods that corrected for population structure among the three sites. This approach allowed dissection of the genomic architecture of shell shape divergence between ecotypes across a wide geographic range, spanning two glacial lineages. GWA revealed 216 quantitative trait loci (QTL) with shell size or shape differences between ecotypes, with most loci explaining a small proportion of phenotypic variation. We found that QTL were evenly distributed across 17 linkage groups, and exhibited elevated interchromosomal linkage, suggesting a genome-wide response to divergent selection on shell shape between the two ecotypes. Shell shape trait-associated loci showed partial overlap with previously identified outlier loci under divergent selection between the two ecotypes, supporting the hypothesis of diversifying selection on these genomic regions. These results suggest that divergence in shell shape between the crab-adapted and wave-adapted ecotypes is produced predominantly by a polygenic genomic architecture with positive linkage disequilibrium among loci of small effect.
Collapse
Affiliation(s)
- Tony Kess
- Department of Integrative BiologyUniversity of GuelphGuelphONCanada
- Present address:
Fisheries and Oceans CanadaSt. John'sNLCanada
| | | |
Collapse
|
30
|
Abstract
Causal inference is essential across the biomedical, behavioural and social sciences.By progressing from confounded statistical associations to evidence of causal relationships, causal inference can reveal complex pathways underlying traits and diseases and help to prioritize targets for intervention. Recent progress in genetic epidemiology - including statistical innovation, massive genotyped data sets and novel computational tools for deep data mining - has fostered the intense development of methods exploiting genetic data and relatedness to strengthen causal inference in observational research. In this Review, we describe how such genetically informed methods differ in their rationale, applicability and inherent limitations and outline how they should be integrated in the future to offer a rich causal inference toolbox.
Collapse
|
31
|
Mikkola LI, Holopainen S, Lappalainen AK, Pessa-Morikawa T, Augustine TJP, Arumilli M, Hytönen MK, Hakosalo O, Lohi H, Iivanainen A. Novel protective and risk loci in hip dysplasia in German Shepherds. PLoS Genet 2019; 15:e1008197. [PMID: 31323019 PMCID: PMC6668854 DOI: 10.1371/journal.pgen.1008197] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Revised: 07/31/2019] [Accepted: 05/14/2019] [Indexed: 12/15/2022] Open
Abstract
Canine hip dysplasia is a common, non-congenital, complex and hereditary disorder. It can inflict severe pain via secondary osteoarthritis and lead to euthanasia. An analogous disorder exists in humans. The genetic background of hip dysplasia in both species has remained ambiguous despite rigorous studies. We aimed to investigate the genetic causes of this disorder in one of the high-risk breeds, the German Shepherd. We performed genetic analyses with carefully phenotyped case-control cohorts comprising 525 German Shepherds. In our genome-wide association studies we identified four suggestive loci on chromosomes 1 and 9. Targeted resequencing of the two loci on chromosome 9 from 24 affected and 24 control German Shepherds revealed deletions of variable sizes in a putative enhancer element of the NOG gene. NOG encodes for noggin, a well-described bone morphogenetic protein inhibitor affecting multiple developmental processes, including joint development. The deletion was associated with the healthy controls and mildly dysplastic dogs suggesting a protective role against canine hip dysplasia. Two enhancer variants displayed a decreased activity in a dual luciferase reporter assay. Our study identifies novel loci and candidate genes for canine hip dysplasia, with potential regulatory variants in the NOG gene. Further research is warranted to elucidate how the identified variants affect the expression of noggin in canine hips, and what the potential effects of the other identified loci are.
Collapse
Affiliation(s)
- Lea I. Mikkola
- Department of Veterinary Biosciences, University of Helsinki, Helsinki, Finland
- Department of Molecular Genetics, Folkhälsan Institute of Genetics, Helsinki, Finland
- Research Programs Unit, Molecular Neurology, University of Helsinki, Helsinki, Finland
| | - Saila Holopainen
- Department of Veterinary Biosciences, University of Helsinki, Helsinki, Finland
- Department of Molecular Genetics, Folkhälsan Institute of Genetics, Helsinki, Finland
- Research Programs Unit, Molecular Neurology, University of Helsinki, Helsinki, Finland
- Department of Equine and Small Animal Medicine, University of Helsinki, Helsinki, Finland
| | - Anu K. Lappalainen
- Department of Equine and Small Animal Medicine, University of Helsinki, Helsinki, Finland
| | | | | | - Meharji Arumilli
- Department of Veterinary Biosciences, University of Helsinki, Helsinki, Finland
- Department of Molecular Genetics, Folkhälsan Institute of Genetics, Helsinki, Finland
- Research Programs Unit, Molecular Neurology, University of Helsinki, Helsinki, Finland
| | - Marjo K. Hytönen
- Department of Veterinary Biosciences, University of Helsinki, Helsinki, Finland
- Department of Molecular Genetics, Folkhälsan Institute of Genetics, Helsinki, Finland
- Research Programs Unit, Molecular Neurology, University of Helsinki, Helsinki, Finland
| | - Osmo Hakosalo
- Department of Veterinary Biosciences, University of Helsinki, Helsinki, Finland
- Department of Molecular Genetics, Folkhälsan Institute of Genetics, Helsinki, Finland
- Research Programs Unit, Molecular Neurology, University of Helsinki, Helsinki, Finland
| | - Hannes Lohi
- Department of Veterinary Biosciences, University of Helsinki, Helsinki, Finland
- Department of Molecular Genetics, Folkhälsan Institute of Genetics, Helsinki, Finland
- Research Programs Unit, Molecular Neurology, University of Helsinki, Helsinki, Finland
| | - Antti Iivanainen
- Department of Veterinary Biosciences, University of Helsinki, Helsinki, Finland
| |
Collapse
|
32
|
Rowland K, Ashwell CM, Persia ME, Rothschild MF, Schmidt C, Lamont SJ. Genetic analysis of production, physiological, and egg quality traits in heat-challenged commercial white egg-laying hens using 600k SNP array data. Genet Sel Evol 2019; 51:31. [PMID: 31238874 PMCID: PMC6593552 DOI: 10.1186/s12711-019-0474-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 06/13/2019] [Indexed: 12/02/2022] Open
Abstract
Background Heat stress negatively affects the welfare and production of chickens. High ambient temperature is considered one of the most ubiquitous abiotic environmental challenges to laying hens around the world. In this study, we recorded several production traits, feed intake, body weight, digestibility, and egg quality of 400 commercial white egg-laying hens before and during a 4-week heat treatment. For the phenotypes that had estimated heritabilities (using 600k SNP chip data) higher than 0, SNP associations were tested using the same 600k genotype data. Results Seventeen phenotypes had heritability estimates higher than 0, including measurements at various time points for feed intake, feed efficiency, body weight, albumen weight, egg quality expressed in Haugh units, egg mass, and also for change in egg mass from prior to heat exposure to various time points during the 4-week heat treatment. Quantitative trait loci (QTL) were identified for 10 of these 17 phenotypes. Some of the phenotypes shared QTL including Haugh units before heat exposure and after 4 weeks of heat treatment. Conclusions Estimated heritabilities differed from 0 for 17 traits, which indicates that they are under genetic control and that there is potential for improving these traits through selective breeding. The association of different QTL with the same phenotypes before heat exposure and during heat treatment indicates that genomic control of traits under heat stress is distinct from that under thermoneutral conditions. This study contributes to the knowledge on the genomic control of response to heat stress in laying hens. Electronic supplementary material The online version of this article (10.1186/s12711-019-0474-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Kaylee Rowland
- Department of Animal Science, Iowa State University, Ames, USA
| | - Chris M Ashwell
- Prestage Department of Poultry Science, North Carolina State University, Raleigh, USA
| | - Michael E Persia
- Department of Animal and Poultry Sciences, Virginia Tech, Blacksburg, USA
| | | | - Carl Schmidt
- University of Delaware, Animal and Food Sciences, Newark, USA
| | - Susan J Lamont
- Department of Animal Science, Iowa State University, Ames, USA.
| |
Collapse
|
33
|
Gmel AI, Druml T, von Niederhäusern R, Leeb T, Neuditschko M. Genome-Wide Association Studies Based on Equine Joint Angle Measurements Reveal New QTL Affecting the Conformation of Horses. Genes (Basel) 2019; 10:genes10050370. [PMID: 31091839 PMCID: PMC6562990 DOI: 10.3390/genes10050370] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 04/30/2019] [Accepted: 05/03/2019] [Indexed: 12/30/2022] Open
Abstract
The evaluation of conformation traits is an important part of selection for breeding stallions and mares. Some of these judged conformation traits involve joint angles that are associated with performance, health, and longevity. To improve our understanding of the genetic background of joint angles in horses, we have objectively measured the angles of the poll, elbow, carpal, fetlock (front and hind), hip, stifle, and hock joints based on one photograph of each of the 300 Franches-Montagnes (FM) and 224 Lipizzan (LIP) horses. After quality control, genome-wide association studies (GWASs) for these traits were performed on 495 horses, using 374,070 genome-wide single nucleotide polymorphisms (SNPs) in a mixed-effect model. We identified two significant quantitative trait loci (QTL) for the poll angle on ECA28 (p = 1.36 × 10−7), 50 kb downstream of the ALX1 gene, involved in cranial morphology, and for the elbow joint on ECA29 (p = 1.69 × 10−7), 49 kb downstream of the RSU1 gene, and 75 kb upstream of the PTER gene. Both genes are associated with bone mineral density in humans. Furthermore, we identified other suggestive QTL associated with the stifle joint on ECA8 (p = 3.10 × 10−7); the poll on ECA1 (p = 6.83 × 10−7); the fetlock joint of the hind limb on ECA27 (p = 5.42 × 10−7); and the carpal joint angle on ECA3 (p = 6.24 × 10−7), ECA4 (p = 6.07 × 10−7), and ECA7 (p = 8.83 × 10−7). The application of angular measurements in genetic studies may increase our understanding of the underlying genetic effects of important traits in equine breeding.
Collapse
Affiliation(s)
- Annik Imogen Gmel
- Agroscope-Swiss National Stud Farm, Les Longs-Prés, 1580 Avenches, Switzerland.
- Institute of Genetics, Vetsuisse Faculty, University of Bern, Bremgartenstrasse 109a, 3012 Bern, Switzerland.
| | - Thomas Druml
- Institute of Animal Breeding and Genetics, Veterinary University Vienna, Veterinärplatz 1, A-1210 Vienna, Austria.
| | | | - Tosso Leeb
- Institute of Genetics, Vetsuisse Faculty, University of Bern, Bremgartenstrasse 109a, 3012 Bern, Switzerland.
| | - Markus Neuditschko
- Agroscope-Swiss National Stud Farm, Les Longs-Prés, 1580 Avenches, Switzerland.
| |
Collapse
|
34
|
Allais S, Hennequet-Antier C, Berri C, Salles L, Demeure O, Le Bihan-Duval E. Mapping of QTL for chicken body weight, carcass composition, and meat quality traits in a slow-growing line. Poult Sci 2019; 98:1960-1967. [PMID: 30535096 DOI: 10.3382/ps/pey549] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 11/09/2018] [Indexed: 01/28/2023] Open
Abstract
Slow-growing chicken lines are valuable genetic resources for the development of well-perceived alternative free-range production. While there is no constraint on increasing growth rate, breeding programs have to evolve in order to include new traits improving the positioning of such lines in the growing market for parts and processed products. In this study, we used dense genotyping to fine map QTL for chicken growth, body composition, and meat quality traits in view of developing new tools for selection of a slow-growing line. The dataset included a total of 836 birds (10 sires, 87 dams, 739 descendants) and 40,203 SNP. QTL for the 15 traits analyzed were detected by 3 different methods, i.e., linkage and linkage disequilibrium haplotype-based analysis (LDLA), family-based single marker association (FASTA), and Bayesian multi-marker regression (Bayes Cπ). After filtering for QTL redundancy, we found 16, 16, and 9 QTL when using the FASTA, LDLA, and Bayes Cπ methods, respectively, with a threshold of 2.49 × 10-5 for FASTA and LDLA, and a Bayes factor of 150 for the Bayes Cπ analysis. They comprised 17 QTL for body weight, 9 QTL for body composition, and 15 QTL for breast meat quality or behavior at slaughter. The 3 methods agreed in the detection of highly significant QTL such as that detected on GGA24 for body weight at 3, 6, and 9 wk, and the 2 QTL detected on GGA17 and GGA18 for breast meat yield. Several significant QTL were also detected for the different components of breast meat quality. This study provided new locations for investigation in order to improve our understanding of the genetic architecture of growth, carcass composition, and meat quality in the chicken and to develop molecular tools for the selection of these traits in a slow-growing line.
Collapse
Affiliation(s)
- S Allais
- PEGASE, Agrocampus Ouest, INRA, 35590 Saint-Gilles, France
| | | | - C Berri
- BOA, INRA, Université de Tours, 37380 Nouzilly, France
| | | | - O Demeure
- PEGASE, Agrocampus Ouest, INRA, 35590 Saint-Gilles, France
| | | |
Collapse
|
35
|
Zhou H, Sinsheimer JS, Bates DM, Chu BB, German CA, Ji SS, Keys KL, Kim J, Ko S, Mosher GD, Papp JC, Sobel EM, Zhai J, Zhou JJ, Lange K. OPENMENDEL: a cooperative programming project for statistical genetics. Hum Genet 2019; 139:61-71. [PMID: 30915546 DOI: 10.1007/s00439-019-02001-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 03/15/2019] [Indexed: 01/06/2023]
Abstract
Statistical methods for genome-wide association studies (GWAS) continue to improve. However, the increasing volume and variety of genetic and genomic data make computational speed and ease of data manipulation mandatory in future software. In our view, a collaborative effort of statistical geneticists is required to develop open source software targeted to genetic epidemiology. Our attempt to meet this need is called the OPENMENDEL project (https://openmendel.github.io). It aims to (1) enable interactive and reproducible analyses with informative intermediate results, (2) scale to big data analytics, (3) embrace parallel and distributed computing, (4) adapt to rapid hardware evolution, (5) allow cloud computing, (6) allow integration of varied genetic data types, and (7) foster easy communication between clinicians, geneticists, statisticians, and computer scientists. This article reviews and makes recommendations to the genetic epidemiology community in the context of the OPENMENDEL project.
Collapse
Affiliation(s)
- Hua Zhou
- Department of Biostatistics, UCLA Fielding School of Public Health, Los Angeles, USA.
| | - Janet S Sinsheimer
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, USA.
| | - Douglas M Bates
- Department of Statistics, University of Wisconsin, Madison, USA
| | - Benjamin B Chu
- Department of Biomathematics, David Geffen School of Medicine at UCLA, Los Angeles, USA
| | - Christopher A German
- Department of Biostatistics, UCLA Fielding School of Public Health, Los Angeles, USA
| | - Sarah S Ji
- Department of Biostatistics, UCLA Fielding School of Public Health, Los Angeles, USA
| | - Kevin L Keys
- Department of Medicine, University of California, San Francisco, USA
| | - Juhyun Kim
- Department of Biostatistics, UCLA Fielding School of Public Health, Los Angeles, USA
| | - Seyoon Ko
- Department of Statistics, Seoul National University, Seoul, South Korea
| | - Gordon D Mosher
- Departments of Statistics and Computer Science, University of California, Riverside, USA
| | - Jeanette C Papp
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, USA
| | - Eric M Sobel
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, USA
| | - Jing Zhai
- Department of Epidemiology and Biostatistics, Mel and Enid Zuckerman College of Public Health, University of Arizona, Tucson, USA
| | - Jin J Zhou
- Department of Epidemiology and Biostatistics, Mel and Enid Zuckerman College of Public Health, University of Arizona, Tucson, USA
| | - Kenneth Lange
- Department of Biomathematics, David Geffen School of Medicine at UCLA, Los Angeles, USA.
| |
Collapse
|
36
|
Fan BJ, Chen X, Sondhi N, Sharmila PF, Soumittra N, Sripriya S, Sacikala S, Asokan R, Friedman DS, Pasquale LR, Gao XR, Vijaya L, Cooke Bailey J, Vitart V, MacGregor S, Hammond CJ, Khor CC, Haines JL, George R, Wiggs JL. Family-Based Genome-Wide Association Study of South Indian Pedigrees Supports WNT7B as a Central Corneal Thickness Locus. Invest Ophthalmol Vis Sci 2019; 59:2495-2502. [PMID: 29847655 PMCID: PMC5961220 DOI: 10.1167/iovs.17-23536] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Purpose To identify genetic risk factors contributing to central corneal thickness (CCT) in individuals from South India, a population with a high prevalence of ocular disorders. Methods One hundred ninety-five individuals from 15 large South Indian pedigrees were genotyped using the Omni2.5 bead array. Family-based association for CCT was conducted using the score test in MERLIN. Results Genome-wide association study (GWAS) identified strongest association for single nucleotide polymorphisms (SNPs) in the first intron of WNT7B and CCT (top SNP rs9330813; β = −0.57, 95% confidence interval [CI]: −0.78 to −0.36; P = 1.7 × 10−7). We further investigated rs9330813 in a Latino cohort and four independent European cohorts. A meta-analysis of these data sets demonstrated statistically significant association between rs9330813 and CCT (β = −3.94, 95% CI: −5.23 to −2.66; P = 1.7 × 10−9). WNT7B SNPs located in the same genomic region that includes rs9330813 have previously been associated with CCT in Latinos but with other ocular quantitative traits related to myopia (corneal curvature and axial length) in a Japanese population (rs10453441 and rs200329677). To evaluate the specificity of the observed WNT7B association with CCT in the South Indian families, we completed an ocular phenome-wide association study (PheWAS) for the top WNT7B SNPs using 45 ocular traits measured in these same families including corneal curvature and axial length. The ocular PheWAS results indicate that in the South Indian families WNT7B SNPs are primarily associated with CCT. Conclusions The results indicate robust evidence for association between WNT7B SNPs and CCT in South Indian pedigrees, and suggest that WNT7B SNPs can have population-specific effects on ocular quantitative traits.
Collapse
Affiliation(s)
- Bao Jian Fan
- Department of Ophthalmology, Harvard Medical School, Massachusetts Eye and Ear Infirmary, Boston, Massachusetts, United States
| | - Xueli Chen
- Department of Ophthalmology & Visual Science, Eye & Ear Nose Throat Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Nisha Sondhi
- Department of Ophthalmology, Harvard Medical School, Massachusetts Eye and Ear Infirmary, Boston, Massachusetts, United States
| | - P Ferdinamarie Sharmila
- SNONGC Department of Genetics and Molecular Biology, Vision Research Foundation, Sankara Nethralaya, Chennai, India
| | - Nagasamy Soumittra
- SNONGC Department of Genetics and Molecular Biology, Vision Research Foundation, Sankara Nethralaya, Chennai, India
| | - Sarangapani Sripriya
- SNONGC Department of Genetics and Molecular Biology, Vision Research Foundation, Sankara Nethralaya, Chennai, India
| | - Srinivasan Sacikala
- SNONGC Department of Genetics and Molecular Biology, Vision Research Foundation, Sankara Nethralaya, Chennai, India
| | - Rashima Asokan
- Medical Research Foundation, Sankara Nethralaya, Chennai, India
| | - David S Friedman
- The Dana Center for Preventive Ophthalmology, Johns Hopkins Medical School, Wilmer Eye Institute, Baltimore, Maryland, United States
| | - Louis R Pasquale
- Department of Ophthalmology, Harvard Medical School, Massachusetts Eye and Ear Infirmary, Boston, Massachusetts, United States.,Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States
| | - X Raymond Gao
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, Illinois, United States
| | - Lingam Vijaya
- Medical Research Foundation, Sankara Nethralaya, Chennai, India
| | - Jessica Cooke Bailey
- Department of Epidemiology and Biostatistics, Institute for Computational Biology, Case Western Reserve University School of Medicine, Cleveland, Ohio, United States
| | - Veronique Vitart
- MRC Human Genetics Unit, Institute for Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Stuart MacGregor
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Christopher J Hammond
- Department of Twin Research and Genetic Epidemiology, King's College London, London, United Kingdom
| | - Chiea Chuen Khor
- Division of Human Genetics, Genome Institute of Singapore, Singapore
| | - Jonathan L Haines
- Department of Epidemiology and Biostatistics, Institute for Computational Biology, Case Western Reserve University School of Medicine, Cleveland, Ohio, United States
| | - Ronnie George
- Medical Research Foundation, Sankara Nethralaya, Chennai, India
| | - Janey L Wiggs
- Department of Ophthalmology, Harvard Medical School, Massachusetts Eye and Ear Infirmary, Boston, Massachusetts, United States
| | | |
Collapse
|
37
|
Revisit Population-based and Family-based Genotype Imputation. Sci Rep 2019; 9:1800. [PMID: 30755687 PMCID: PMC6372660 DOI: 10.1038/s41598-018-38469-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 12/27/2018] [Indexed: 11/12/2022] Open
Abstract
Genome-Wide Association (GWA) with population-based imputation (PBI) has been successful in identifying common variants associated with complex diseases; however, much heritability remains to be explained and low frequency variants (LFV) may contribute. To identify LFV, a study of unrelated individuals may no longer be as efficient as a family study, where rare population variants can be frequent in families. Family-based imputation (FBI) provides an opportunity to evaluate LFV. To compare the performance of PBI and FBI, we conducted extensive simulations, generating genotypes using SeqSIMLA from various reference panels for families. We masked genotype information for variants unavailable in Framingham 550 K GWA genotype data in less informative subjects selected by GIGI-Pick. We implemented IMPUTE2 with duoHMM in SHAPEIT (Impute2_duoHMM) for PBI, MERLIN and GIGI for FBI and PedBLIMP for a hybrid approach. In general, FBI in both MERLIN and GIGI outperformed other approaches with imputation accuracy greater than 0.99 for the squared correlation and imputation quality scores (IQS) especially for LFV, although imputation accuracy from MERLIN depends on pedigree splitting for larger families. PBI performed worst with the exception of good imputation accuracy for common variants when a closely ancestry matched reference is used. In summary, linkage disequilibrium (LD) information from large available genotype resources provides good imputation for common variants with well-selected reference panels without requiring densely sequenced data in family members, while imputation of LFV with FBI benefits more from information on inheritance patterns within families yielding better imputation.
Collapse
|
38
|
Robledo D, Gutiérrez AP, Barría A, Lhorente JP, Houston RD, Yáñez JM. Discovery and Functional Annotation of Quantitative Trait Loci Affecting Resistance to Sea Lice in Atlantic Salmon. Front Genet 2019; 10:56. [PMID: 30800143 PMCID: PMC6375901 DOI: 10.3389/fgene.2019.00056] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 01/23/2019] [Indexed: 12/22/2022] Open
Abstract
Sea lice (Caligus rogercresseyi) are ectoparasitic copepods which have a large negative economic and welfare impact in Atlantic salmon (Salmo salar) aquaculture, particularly in Chile. A multi-faceted prevention and control strategy is required to tackle lice, and selective breeding contributes via cumulative improvement of host resistance to the parasite. While host resistance has been shown to be heritable, little is yet known about the individual loci that contribute to this resistance, the potential underlying genes, and their mechanisms of action. In this study we took a multifaceted approach to identify and characterize quantitative trait loci (QTL) affecting host resistance in a population of 2,688 Caligus-challenged Atlantic salmon post-smolts from a commercial breeding program. We used low and medium density genotyping with imputation to collect genome-wide SNP marker data for all animals. Moderate heritability estimates of 0.28 and 0.24 were obtained for lice density (as a measure of host resistance) and growth during infestation, respectively. Three QTL explaining between 7 and 13% of the genetic variation in resistance to sea lice (as represented by the traits of lice density) were detected on chromosomes 3, 18, and 21. Characterisation of these QTL regions was undertaken using RNA sequencing and pooled whole genome sequencing data. This resulted in the identification of a shortlist of potential underlying causative genes, and candidate functional mutations for further study. For example, candidates within the chromosome 3 QTL include a putative premature stop mutation in TOB1 (an anti-proliferative transcription factor involved in T cell regulation) and an uncharacterized protein which showed significant differential allelic expression (implying the existence of a cis-acting regulatory mutation). While host resistance to sea lice is polygenic in nature, the results of this study highlight significant QTL regions together explaining between 7 and 13 % of the heritability of the trait. Future investigation of these QTL may enable improved knowledge of the functional mechanisms of host resistance to sea lice, and incorporation of functional variants to improve genomic selection accuracy.
Collapse
Affiliation(s)
- Diego Robledo
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Edinburgh, United Kingdom
| | - Alejandro P. Gutiérrez
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Edinburgh, United Kingdom
| | - Agustín Barría
- Facultad de Ciencias Veterinarias y Pecuarias, Universidad de Chile, Santiago, Chile
| | | | - Ross D. Houston
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Edinburgh, United Kingdom
| | - José M. Yáñez
- Facultad de Ciencias Veterinarias y Pecuarias, Universidad de Chile, Santiago, Chile
- Núcleo Milenio INVASAL, Concepción, Chile
| |
Collapse
|
39
|
Saad M, Wijsman EM. Association score testing for rare variants and binary traits in family data with shared controls. Brief Bioinform 2019; 20:245-253. [PMID: 28968627 DOI: 10.1093/bib/bbx107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Indexed: 11/12/2022] Open
Abstract
Genome-wide association studies have been an important approach used to localize trait loci, with primary focus on common variants. The multiple rare variant-common disease hypothesis may explain the missing heritability remaining after accounting for identified common variants. Advances of sequencing technologies with their decreasing costs, coupled with methodological advances in the context of association studies in large samples, now make the study of rare variants at a genome-wide scale feasible. The resurgence of family-based association designs because of their advantage in studying rare variants has also stimulated more methods development, mainly based on linear mixed models (LMMs). Other tests such as score tests can have advantages over the LMMs, but to date have mainly been proposed for single-marker association tests. In this article, we extend several score tests (χcorrected2, WQLS, and SKAT) to the multiple variant association framework. We evaluate and compare their statistical performances relative with the LMM. Moreover, we show that three tests can be cast as the difference between marker allele frequencies (AFs) estimated in each of the group of affected and unaffected subjects. We show that these tests are flexible, as they can be based on related, unrelated or both related and unrelated subjects. They also make feasible an increasingly common design that only sequences a subset of affected subjects (related or unrelated) and uses for comparison publicly available AFs estimated in a group of healthy subjects. Finally, we show the great impact of linkage disequilibrium on the performance of all these tests.
Collapse
Affiliation(s)
- Mohamad Saad
- Department of Biostatistics, University of Washington, Seattle, USA.,Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, USA.,Qatar Computing Research Institute, Hamad Bin Khalifa University, Doha, Qatar
| | - Ellen M Wijsman
- Department of Biostatistics, University of Washington, Seattle, USA.,Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, USA
| |
Collapse
|
40
|
Cecchi F, Dadousis C, Bozzi R, Fratini F, Russo C, Bandecchi P, Cantile C, Mazzei M. Genome scan for the possibility of identifying candidate resistance genes for goat lentiviral infections in the Italian Garfagnina goat breed. Trop Anim Health Prod 2018; 51:729-733. [PMID: 30350159 DOI: 10.1007/s11250-018-1728-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 10/12/2018] [Indexed: 11/28/2022]
Abstract
Small ruminant lentiviruses (SRLVs) are a heterogeneous group of viruses of sheep, goat, and wild ruminants responsible of lifelong persistent infection leading to a multisystem chronic disease. Increased evidences indicate that host genetic factors could influence the individual SRLV resistance. The present study was conducted on the Garfagnina goat breed, an Italian goat population registered on the Tuscan regional repertory of genetic resources at risk of extinction. Forty-eight adult goats belonging to a single flock were studied. SRLV diagnosis was achieved by serological tests and 21 serologically positive animals were identified. All animals were genotyped with the Illumina GoatSNP60 BeadChip and a genome-wide scan was then performed on the individual marker genotypes, in an attempt to identify genomic regions associated with the infection. One SNP was found significant (P < 5 × 10-5) on CHR 18 at 62,360,918 bp. The SNP was an intron of the zinc finger protein 331 (ZNF331) protein. In the region 1 Mb upstream the significant SNP, the NLRP12 (NLR family pyrin domain containing 12), the PRKCG (protein kinase C gamma), and the CACNG7 (calcium voltage-gated channel auxiliary subunit gamma 7) were found.
Collapse
Affiliation(s)
- Francesca Cecchi
- Department of Veterinary Sciences, University of Pisa, Viale delle Piagge 2, Pisa, Italy.
| | - Christos Dadousis
- DISPAA-Sez. Scienze Animali, Università degli Studi di Firenze, Piazzale delle Cascine, 18, Firenze, Italy
| | - Riccardo Bozzi
- DISPAA-Sez. Scienze Animali, Università degli Studi di Firenze, Piazzale delle Cascine, 18, Firenze, Italy
| | - Filippo Fratini
- Department of Veterinary Sciences, University of Pisa, Viale delle Piagge 2, Pisa, Italy
| | - Claudia Russo
- Department of Veterinary Sciences, University of Pisa, Viale delle Piagge 2, Pisa, Italy
| | - Patrizia Bandecchi
- Department of Veterinary Sciences, University of Pisa, Viale delle Piagge 2, Pisa, Italy
| | - Carlo Cantile
- Department of Veterinary Sciences, University of Pisa, Viale delle Piagge 2, Pisa, Italy
| | - Maurizio Mazzei
- Department of Veterinary Sciences, University of Pisa, Viale delle Piagge 2, Pisa, Italy
| |
Collapse
|
41
|
Gutierrez AP, Matika O, Bean TP, Houston RD. Genomic Selection for Growth Traits in Pacific Oyster ( Crassostrea gigas): Potential of Low-Density Marker Panels for Breeding Value Prediction. Front Genet 2018; 9:391. [PMID: 30283494 PMCID: PMC6156352 DOI: 10.3389/fgene.2018.00391] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 08/29/2018] [Indexed: 12/20/2022] Open
Abstract
Pacific oysters are a key aquaculture species globally, and genetic improvement via selective breeding is a major target. Genomic selection has the potential to expedite genetic gain for key target traits of a breeding program, but has not yet been evaluated in oyster. The recent development of SNP arrays for Pacific oyster (Crassostrea gigas) raises the opportunity to test genomic selection strategies for polygenic traits. In this study, a population of 820 oysters (comprising 23 full-sibling families) were genotyped using a medium density SNP array (23 K informative SNPs), and the genetic architecture of growth-related traits [shell height (SH), shell length (SL), and wet weight (WW)] was evaluated. Heritability was estimated to be moderate for the three traits (0.26 ± 0.06 for SH, 0.23 ± 0.06 for SL and 0.35 ± 0.05 for WW), and results of a GWAS indicated that the underlying genetic architecture was polygenic. Genomic prediction approaches were used to estimate breeding values for growth, and compared to pedigree based approaches. The accuracy of the genomic prediction models (GBLUP) outperformed the traditional pedigree approach (PBLUP) by ∼25% for SL and WW, and ∼30% for SH. Further, reduction in SNP marker density had little impact on prediction accuracy, even when density was reduced to a few hundred SNPs. These results suggest that the use of genomic selection in oyster breeding could offer benefits for the selection of breeding candidates to improve complex economic traits at relatively modest cost.
Collapse
Affiliation(s)
- Alejandro P Gutierrez
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Edinburgh, United Kingdom
| | - Oswald Matika
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Edinburgh, United Kingdom
| | - Tim P Bean
- Weymouth Laboratory, Centre for Environment, Fisheries and Aquaculture Science (CEFAS), Weymouth, United Kingdom
| | - Ross D Houston
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
42
|
İnanç ME, Tekin K, Akkurt MY, Olgac KT, Yılmaz B, Çil B, Kızılaslan M, Taşdemir U, Tuncer PB, Büyükleblebici S, Uysal O, Çınar Kul B. Genomewide association of male reproductive traits in Aksaray Malakli dogs. Reprod Domest Anim 2018; 53:1555-1562. [DOI: 10.1111/rda.13302] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 07/30/2018] [Indexed: 11/29/2022]
Affiliation(s)
- Muhammed Enes İnanç
- Faculty of Veterinary Medicine Department of Reproduction and Artificial Insemination Mehmet Akif Ersoy University Burdur Turkey
| | - Koray Tekin
- Faculty of Veterinary Medicine Department of Reproduction and Artificial Insemination Ankara University Ankara Turkey
| | - Mustafa Yenal Akkurt
- Faculty of Veterinary Medicine Department of Genetics Ankara University Ankara Turkey
| | - Kemal Tuna Olgac
- Faculty of Veterinary Medicine Department of Reproduction and Artificial Insemination Ankara University Ankara Turkey
| | - Burak Yılmaz
- Faculty of Veterinary Medicine Department of Reproduction and Artificial Insemination Ankara University Ankara Turkey
| | - Beste Çil
- Faculty of Veterinary Medicine Department of Reproduction and Artificial Insemination Ankara University Ankara Turkey
| | - Mehmet Kızılaslan
- International Center for Livestock Research and Training Ankara Turkey
| | - Umut Taşdemir
- Technical Sciences Vocational School Aksaray University Aksaray Turkey
| | | | | | - Ongun Uysal
- Faculty of Veterinary Medicine Department of Reproduction and Artificial Insemination Ankara University Ankara Turkey
| | - Bengi Çınar Kul
- Faculty of Veterinary Medicine Department of Genetics Ankara University Ankara Turkey
| |
Collapse
|
43
|
Hsu Y, Auerbach J, Zheng T, Lo SH. Coping with family structure in genome-wide association studies: a comparative evaluation. BMC Proc 2018; 12:42. [PMID: 30263047 PMCID: PMC6156900 DOI: 10.1186/s12919-018-0151-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
In this paper, a fully statistical investigation of the control of family structure as random effects is analyzed and discussed, using both the genome-wide association studies (GWAS) data and simulated data. Three modeling strategies are proposed and the analysis results suggest the hybrid use of results from all possible models should be combined in practice.
Collapse
|
44
|
Wang X, Zhang Z, Morris N, Cai T, Lee S, Wang C, Yu TW, Walsh CA, Lin X. Rare variant association test in family-based sequencing studies. Brief Bioinform 2018; 18:954-961. [PMID: 27677958 DOI: 10.1093/bib/bbw083] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Indexed: 12/20/2022] Open
Abstract
The objective of this article is to introduce valid and robust methods for the analysis of rare variants for family-based exome chips, whole-exome sequencing or whole-genome sequencing data. Family-based designs provide unique opportunities to detect genetic variants that complement studies of unrelated individuals. Currently, limited methods and software tools have been developed to assist family-based association studies with rare variants, especially for analyzing binary traits. In this article, we address this gap by extending existing burden and kernel-based gene set association tests for population data to related samples, with a particular emphasis on binary phenotypes. The proposed approach blends the strengths of kernel machine methods and generalized estimating equations. Importantly, the efficient generalized kernel score test can be applied as a mega-analysis framework to combine studies with different designs. We illustrate the application of the proposed method using data from an exome sequencing study of autism. Methods discussed in this article are implemented in an R package 'gskat', which is available on CRAN and GitHub.
Collapse
|
45
|
Rowland K, Wolc A, Gallardo RA, Kelly T, Zhou H, Dekkers JCM, Lamont SJ. Genetic Analysis of a Commercial Egg Laying Line Challenged With Newcastle Disease Virus. Front Genet 2018; 9:326. [PMID: 30177951 PMCID: PMC6110172 DOI: 10.3389/fgene.2018.00326] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 07/30/2018] [Indexed: 01/17/2023] Open
Abstract
In low income countries, chickens play a vital role in daily life. They provide a critical source of protein through egg production and meat. Newcastle disease, caused by avian paramyxovirus type 1, has been ranked as the most devastating disease for scavenging chickens in Africa and Asia. High mortality among flocks infected with velogenic strains leads to a devastating loss of dietary protein and buying power for rural households. Improving the genetic resistance of chickens to Newcastle Disease virus (NDV), in addition to vaccination, is a practical target for improvement of poultry production in low income countries. Because response to NDV has a component of genetic control, it can be influenced through selective breeding. Adding genomic information to a breeding program can increase the amount of genetic progress per generation. In this study, we challenged a commercial egg-laying line with a lentogenic strain of NDV, measured phenotypic responses, collected genotypes, and associated genotypes with phenotypes. Collected phenotypes included viral load at 2 and 6 days post-infection (dpi), antibody levels pre-challenge and 10 dpi, and growth rates pre- and post-challenge. Six suggestive QTL associated with response to NDV and/or growth were identified, including novel and known QTL confirming previously reported associations with related traits. Additionally, previous RNA-seq analysis provided support for several of the genes located in or near the identified QTL. Considering the trend of negative genetic correlation between antibody and Newcastle Disease tolerance (growth under disease) and estimates of moderate to high heritability, we provide evidence that these NDV response traits can be influenced through selective breeding. Producing chickens that perform favorably in challenging environments will ultimately increase the supply of quality protein for human consumption.
Collapse
Affiliation(s)
- Kaylee Rowland
- Department of Animal Science, Iowa State University, Ames, IA, United States
| | - Anna Wolc
- Department of Animal Science, Iowa State University, Ames, IA, United States.,Hy-Line International, Dallas Center, IA, United States
| | - Rodrigo A Gallardo
- School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
| | - Terra Kelly
- School of Veterinary Medicine, University of California, Davis, Davis, CA, United States.,Department of Animal Science, University of California, Davis, Davis, CA, United States
| | - Huaijun Zhou
- Department of Animal Science, University of California, Davis, Davis, CA, United States
| | - Jack C M Dekkers
- Department of Animal Science, Iowa State University, Ames, IA, United States
| | - Susan J Lamont
- Department of Animal Science, Iowa State University, Ames, IA, United States
| |
Collapse
|
46
|
Krishnan M, Major TJ, Topless RK, Dewes O, Yu L, Thompson JMD, McCowan L, de Zoysa J, Stamp LK, Dalbeth N, Harré Hindmarsh J, Rapana N, Deka R, Eng WWH, Weeks DE, Minster RL, McGarvey ST, Viali S, Naseri T, Sefuiva Reupena M, Wilcox P, Grattan D, Shepherd PR, Shelling AN, Murphy R, Merriman TR. Discordant association of the CREBRF rs373863828 A allele with increased BMI and protection from type 2 diabetes in Māori and Pacific (Polynesian) people living in Aotearoa/New Zealand. Diabetologia 2018; 61:1603-1613. [PMID: 29721634 PMCID: PMC6434933 DOI: 10.1007/s00125-018-4623-1] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2017] [Accepted: 03/16/2018] [Indexed: 12/28/2022]
Abstract
AIMS/HYPOTHESIS The A (minor) allele of CREBRF rs373863828 has been associated with increased BMI and reduced risk of type 2 diabetes in the Samoan populations of Samoa and American Samoa. Our aim was to test rs373863828 for associations with BMI and the odds of type 2 diabetes, gout and chronic kidney disease (CKD) in Māori and Pacific (Polynesian) people living in Aotearoa/New Zealand. METHODS Linear and logistic regression models were used to analyse the association of the A allele of CREBRF rs373863828 with BMI, log-transformed BMI, waist circumference, type 2 diabetes, gout and CKD in 2286 adults. The primary analyses were adjusted for age, sex, the first four genome-wide principal components and (where appropriate) BMI, waist circumference and type 2 diabetes. The primary analysis was conducted in ancestrally defined groups and association effects were combined using meta-analysis. RESULTS For the A allele of rs373863828, the effect size was 0.038 (95% CI 0.022, 0.055, p = 4.8 × 10-6) for log-transformed BMI, with OR 0.59 (95% CI 0.47, 0.73, p = 1.9 × 10-6) for type 2 diabetes. There was no evidence for an association of genotype with variance in BMI (p = 0.13), and nor was there evidence for associations with serum urate (β = 0.012 mmol/l, pcorrected = 0.10), gout (OR 1.00, p = 0.98) or CKD (OR 0.91, p = 0.59). CONCLUSIONS/INTERPRETATION Our results in New Zealand Polynesian adults replicate, with very similar effect sizes, the association of the A allele of rs373863828 with higher BMI but lower odds of type 2 diabetes among Samoan adults living in Samoa and American Samoa.
Collapse
Affiliation(s)
- Mohanraj Krishnan
- Department of Obstetrics and Gynaecology, University of Auckland, Auckland, New Zealand
- Department of Medicine, Faculty of Medical and Health Sciences, University of Auckland University of Auckland, Auckland, New Zealand
| | - Tanya J Major
- Department of Biochemistry, University of Otago, 710 Cumberland Street, Dunedin, 9054, New Zealand
| | - Ruth K Topless
- Department of Biochemistry, University of Otago, 710 Cumberland Street, Dunedin, 9054, New Zealand
| | - Ofa Dewes
- Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, New Zealand
| | - Lennex Yu
- Department of Anatomy and Medical Imaging, University of Auckland, Auckland, New Zealand
| | - John M D Thompson
- Department of Obstetrics and Gynaecology, University of Auckland, Auckland, New Zealand
- Department of Paediatrics, Child and Youth Health, University of Auckland, Auckland, New Zealand
| | - Lesley McCowan
- Department of Obstetrics and Gynaecology, University of Auckland, Auckland, New Zealand
| | - Janak de Zoysa
- Renal Services, Waitemata District Health Board, Auckland, New Zealand
| | - Lisa K Stamp
- Department of Medicine, University of Otago Christchurch, Christchurch, New Zealand
| | - Nicola Dalbeth
- Department of Medicine, Faculty of Medical and Health Sciences, University of Auckland University of Auckland, Auckland, New Zealand
| | - Jennie Harré Hindmarsh
- Ngāti Porou Hauora Charitable Trust, Te Puia Springs, Tairāwhiti East Coast, New Zealand
| | - Nuku Rapana
- Pukapuka Community of New Zealand Inc., Mangere, Auckland, New Zealand
| | - Ranjan Deka
- Department of Environmental Health, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Winston W H Eng
- Department of Biostatistics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Daniel E Weeks
- Department of Biostatistics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ryan L Minster
- Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Stephen T McGarvey
- International Health Institute, Department of Epidemiology, Brown University School of Public Health, and Department of Anthropology, Brown University, Providence, RI, USA
| | | | - Take Naseri
- Ministry of Health, Government of Samoa, Apia, Samoa
| | | | - Phillip Wilcox
- Department of Mathematics and Statistics, University of Otago, Dunedin, New Zealand
| | - David Grattan
- Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, New Zealand
- Department of Anatomy, University of Otago, Dunedin, New Zealand
| | - Peter R Shepherd
- Department of Medicine, Faculty of Medical and Health Sciences, University of Auckland University of Auckland, Auckland, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, New Zealand
| | - Andrew N Shelling
- Department of Obstetrics and Gynaecology, University of Auckland, Auckland, New Zealand
| | - Rinki Murphy
- Department of Medicine, Faculty of Medical and Health Sciences, University of Auckland University of Auckland, Auckland, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, New Zealand
| | - Tony R Merriman
- Department of Biochemistry, University of Otago, 710 Cumberland Street, Dunedin, 9054, New Zealand.
- Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, New Zealand.
| |
Collapse
|
47
|
Raymond B, Johansson AM, McCormack HA, Fleming RH, Schmutz M, Dunn IC, De Koning DJ. Genome-wide association study for bone strength in laying hens. J Anim Sci 2018; 96:2525-2535. [PMID: 29701819 PMCID: PMC6159512 DOI: 10.1093/jas/sky157] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 04/24/2018] [Indexed: 01/07/2023] Open
Abstract
Bone fracture in egg laying hens is a growing welfare and economic concern in the industry. Although environmental conditions and management (especially nutrition) can exacerbate it, the primary cause of bone weakness and the resulting fractures is believed to have a genetic basis. To test this hypothesis, we performed a genome-wide association study to identify the loci associated with bone strength in laying hens. Genotype and phenotype data were obtained from 752 laying hens belonging to the same pure line population. These hens were genotyped for 580,961 SNPs, with 232,021 SNPs remaining after quality control. Each of the SNPs were tested for association with tibial breaking strength using the family-based score test for association. A total of 52 SNPs across chromosomes 1, 3, 8, and 16 were significantly associated with tibial breaking strength with the genome-wide significance threshold set as a corrected P value of 10e-5. Based on the local linkage disequilibrium around the significant SNPs, 5 distinct and novel QTLs were identified on chromosomes 1 (2 QTLs), 3 (1 QTL), 8 (1 QTL) and 16 (1 QTL). The strongest association was detected within the QTL region on chromosome 8, with the most significant SNP having a corrected P value of 4e-7. A number of candidate genes were identified within the QTL regions, including the BRD2 gene that is required for normal bone physiology. Bone-related pathways involving some of the genes were also identified including chloride channel activity, which regulates bone reabsorption, and intermediate filament organization, which plays a role in the regulation of bone mass. Our result supports previous studies that suggest that bone strength is highly regulated by genetics. It is therefore possible to reduce bone fractures in laying hens through genetic selection and ultimately improve hen welfare.
Collapse
Affiliation(s)
- Biaty Raymond
- Swedish University of Agricultural Sciences, Department of Animal Breeding and Genetics, Uppsala, Sweden
| | - Anna Maria Johansson
- Swedish University of Agricultural Sciences, Department of Animal Breeding and Genetics, Uppsala, Sweden
| | - Heather Anne McCormack
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Scotland, UK
| | - Robert Hall Fleming
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Scotland, UK
| | | | - Ian Chisholm Dunn
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Scotland, UK
| | - Dirk Jan De Koning
- Swedish University of Agricultural Sciences, Department of Animal Breeding and Genetics, Uppsala, Sweden
| |
Collapse
|
48
|
Tedja MS, Wojciechowski R, Hysi PG, Eriksson N, Furlotte NA, Verhoeven VJ, Iglesias AI, Meester-Smoor MA, Tompson SW, Fan Q, Khawaja AP, Cheng CY, Höhn R, Yamashiro K, Wenocur A, Grazal C, Haller T, Metspalu A, Wedenoja J, Jonas JB, Wang YX, Xie J, Mitchell P, Foster PJ, Klein BE, Klein R, Paterson AD, Hosseini SM, Shah RL, Williams C, Teo YY, Tham YC, Gupta P, Zhao W, Shi Y, Saw WY, Tai ES, Sim XL, Huffman JE, Polašek O, Hayward C, Bencic G, Rudan I, Wilson JF, Joshi PK, Tsujikawa A, Matsuda F, Whisenhunt KN, Zeller T, van der Spek PJ, Haak R, Meijers-Heijboer H, van Leeuwen EM, Iyengar SK, Lass JH, Hofman A, Rivadeneira F, Uitterlinden AG, Vingerling JR, Lehtimäki T, Raitakari OT, Biino G, Concas MP, Schwantes-An TH, Igo RP, Cuellar-Partida G, Martin NG, Craig JE, Gharahkhani P, Williams KM, Nag A, Rahi JS, Cumberland PM, Delcourt C, Bellenguez C, Ried JS, Bergen AA, Meitinger T, Gieger C, Wong TY, Hewitt AW, Mackey DA, Simpson CL, Pfeiffer N, Pärssinen O, Baird PN, Vitart V, Amin N, van Duijn CM, Bailey-Wilson JE, Young TL, Saw SM, Stambolian D, MacGregor S, Guggenheim JA, Tung JY, Hammond CJ, Klaver CC. Genome-wide association meta-analysis highlights light-induced signaling as a driver for refractive error. Nat Genet 2018; 50:834-848. [PMID: 29808027 PMCID: PMC5980758 DOI: 10.1038/s41588-018-0127-7] [Citation(s) in RCA: 193] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Accepted: 03/26/2018] [Indexed: 12/18/2022]
Abstract
Refractive errors, including myopia, are the most frequent eye disorders worldwide and an increasingly common cause of blindness. This genome-wide association meta-analysis in 160,420 participants and replication in 95,505 participants increased the number of established independent signals from 37 to 161 and showed high genetic correlation between Europeans and Asians (>0.78). Expression experiments and comprehensive in silico analyses identified retinal cell physiology and light processing as prominent mechanisms, and also identified functional contributions to refractive-error development in all cell types of the neurosensory retina, retinal pigment epithelium, vascular endothelium and extracellular matrix. Newly identified genes implicate novel mechanisms such as rod-and-cone bipolar synaptic neurotransmission, anterior-segment morphology and angiogenesis. Thirty-one loci resided in or near regions transcribing small RNAs, thus suggesting a role for post-transcriptional regulation. Our results support the notion that refractive errors are caused by a light-dependent retina-to-sclera signaling cascade and delineate potential pathobiological molecular drivers.
Collapse
Affiliation(s)
- Milly S. Tedja
- Department of Ophthalmology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Robert Wojciechowski
- Department of Epidemiology and Medicine, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
- Computational and Statistical Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, USA
- Wilmer Eye Institute, Johns Hopkins Medical Institutions, Baltimore, Maryland, USA
| | - Pirro G. Hysi
- Section of Academic Ophthalmology, School of Life Course Sciences, King’s College London, London, UK
| | | | | | - Virginie J.M. Verhoeven
- Department of Ophthalmology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Adriana I. Iglesias
- Department of Ophthalmology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Magda A. Meester-Smoor
- Department of Ophthalmology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Stuart W. Tompson
- Department of Ophthalmology and Visual Sciences, University of Wisconsin–Madison, Madison, Wisconsin, USA
| | - Qiao Fan
- Centre for Quantitative Medicine, DUKE-National University of Singapore, Singapore
| | - Anthony P. Khawaja
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- NIHR Biomedical Research Centre, Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London, UK
| | - Ching-Yu Cheng
- Centre for Quantitative Medicine, DUKE-National University of Singapore, Singapore
- Ocular Epidemiology Research Group, Singapore Eye Research Institute, Singapore National Eye Centre, Singapore
| | - René Höhn
- Department of Ophthalmology, University Hospital Bern, Inselspital, University of Bern, Bern, Switzerland
- Department of Ophthalmology, University Medical Center Mainz, Mainz, Germany
| | - Kenji Yamashiro
- Department of Ophthalmology and Visual Sciences, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Adam Wenocur
- Department of Ophthalmology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Clare Grazal
- Department of Ophthalmology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Toomas Haller
- Estonian Genome Center, University of Tartu, Tartu, Estonia
| | | | - Juho Wedenoja
- Department of Ophthalmology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Department of Public Health, University of Helsinki, Helsinki, Finland
| | - Jost B. Jonas
- Department of Ophthalmology, Medical Faculty Mannheim of the Ruprecht-Karls-University of Heidelberg, Mannheim, Germany
- Beijing Institute of Ophthalmology, Beijing Key Laboratory of Ophthalmology and Visual Sciences, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Ya Xing Wang
- Beijing Institute of Ophthalmology, Beijing Key Laboratory of Ophthalmology and Visual Sciences, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Jing Xie
- Centre for Eye Research Australia, Ophthalmology, Department of Surgery, University of Melbourne, Royal Victorian Eye and Ear Hospital, Melbourne, Australia
| | - Paul Mitchell
- Department of Ophthalmology, Centre for Vision Research, Westmead Institute for Medical Research, University of Sydney, Sydney, Australia
| | - Paul J. Foster
- NIHR Biomedical Research Centre, Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London, UK
| | - Barbara E.K. Klein
- Department of Ophthalmology and Visual Sciences, University of Wisconsin–Madison, Madison, Wisconsin, USA
| | - Ronald Klein
- Department of Ophthalmology and Visual Sciences, University of Wisconsin–Madison, Madison, Wisconsin, USA
| | - Andrew D. Paterson
- Program in Genetics and Genome Biology, Hospital for Sick Children and University of Toronto, Toronto, Ontario, Canada
| | - S. Mohsen Hosseini
- Program in Genetics and Genome Biology, Hospital for Sick Children and University of Toronto, Toronto, Ontario, Canada
| | - Rupal L. Shah
- School of Optometry & Vision Sciences, Cardiff University, Cardiff, UK
| | - Cathy Williams
- Department of Population Health Sciences, Bristol Medical School, Bristol, UK
| | - Yik Ying Teo
- Department of Statistics and Applied Probability, National University of Singapore, Singapore
- Saw Swee Hock School of Public Health, National University Health Systems, National University of Singapore, Singapore
| | - Yih Chung Tham
- Ocular Epidemiology Research Group, Singapore Eye Research Institute, Singapore National Eye Centre, Singapore
| | - Preeti Gupta
- Department of Health Service Research, Singapore Eye Research Institute, Singapore National Eye Centre, Singapore
| | - Wanting Zhao
- Centre for Quantitative Medicine, DUKE-National University of Singapore, Singapore
- Statistics Support Platform, Singapore Eye Research Institute, Singapore National Eye Centre, Singapore
| | - Yuan Shi
- Statistics Support Platform, Singapore Eye Research Institute, Singapore National Eye Centre, Singapore
| | - Woei-Yuh Saw
- Life Sciences Institute, National University of Singapore, Singapore
| | - E-Shyong Tai
- Saw Swee Hock School of Public Health, National University Health Systems, National University of Singapore, Singapore
| | - Xue Ling Sim
- Saw Swee Hock School of Public Health, National University Health Systems, National University of Singapore, Singapore
| | - Jennifer E. Huffman
- MRC Human Genetics Unit, MRC Institute of Genetics & Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Ozren Polašek
- Faculty of Medicine, University of Split, Split, Croatia
| | - Caroline Hayward
- MRC Human Genetics Unit, MRC Institute of Genetics & Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Goran Bencic
- Department of Ophthalmology, Sisters of Mercy University Hospital, Zagreb, Croatia
| | - Igor Rudan
- Centre for Global Health Research, Usher Institute for Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, UK
| | - James F. Wilson
- MRC Human Genetics Unit, MRC Institute of Genetics & Molecular Medicine, University of Edinburgh, Edinburgh, UK
- Centre for Global Health Research, Usher Institute for Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, UK
| | | | | | | | - Peter K. Joshi
- Centre for Global Health Research, Usher Institute for Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, UK
| | - Akitaka Tsujikawa
- Department of Ophthalmology and Visual Sciences, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Fumihiko Matsuda
- Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Kristina N. Whisenhunt
- Department of Ophthalmology and Visual Sciences, University of Wisconsin–Madison, Madison, Wisconsin, USA
| | - Tanja Zeller
- Clinic for General and Interventional Cardiology, University Heart Center Hamburg, Hamburg, Germany
| | | | - Roxanna Haak
- Department of Bioinformatics, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Hanne Meijers-Heijboer
- Department of Clinical Genetics, Academic Medical Center, Amsterdam, The Netherlands
- Department of Clinical Genetics, VU University Medical Center, Amsterdam, The Netherlands
| | - Elisabeth M. van Leeuwen
- Department of Ophthalmology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Sudha K. Iyengar
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, Ohio, USA
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University and University Hospitals Eye Institute, Cleveland, Ohio, USA
- Department of Genetics, Case Western Reserve University, Cleveland, Ohio, USA
| | - Jonathan H. Lass
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, Ohio, USA
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University and University Hospitals Eye Institute, Cleveland, Ohio, USA
| | - Albert Hofman
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Epidemiology, Harvard T.HChan School of Public Health, Boston, Massachusetts, USA
- Netherlands Consortium for Healthy Ageing, Netherlands Genomics Initiative, the Hague, the Netherlands
| | - Fernando Rivadeneira
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
- Netherlands Consortium for Healthy Ageing, Netherlands Genomics Initiative, the Hague, the Netherlands
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - André G. Uitterlinden
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
- Netherlands Consortium for Healthy Ageing, Netherlands Genomics Initiative, the Hague, the Netherlands
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | | | - Terho Lehtimäki
- Department of Clinical Chemistry, Finnish Cardiovascular Research Center-Tampere, Faculty of Medicine and Life Sciences, University of Tampere
- Department of Clinical Chemistry, Fimlab Laboratories, University of Tampere, Tampere, Finland
| | - Olli T. Raitakari
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland
- Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, Turku, Finland
| | - Ginevra Biino
- Institute of Molecular Genetics, National Research Council of Italy, Sassari, Italy
| | - Maria Pina Concas
- Institute for Maternal and Child Health - IRCCS “Burlo Garofolo”, Trieste, Italy
| | - Tae-Hwi Schwantes-An
- Computational and Statistical Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, USA
- Department of Medical and Molecular Genetics, Indiana University, School of Medicine, Indianapolis, Indiana, USA
| | - Robert P. Igo
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, Ohio, USA
| | | | - Nicholas G. Martin
- Genetic Epidemiology, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Jamie E. Craig
- Department of Ophthalmology, Flinders University, Adelaide, Australia
| | - Puya Gharahkhani
- Statistical Genetics, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Katie M. Williams
- Section of Academic Ophthalmology, School of Life Course Sciences, King’s College London, London, UK
| | - Abhishek Nag
- Department of Twin Research and Genetic Epidemiology, King’s College London, London, UK
| | - Jugnoo S. Rahi
- NIHR Biomedical Research Centre, Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London, UK
- Great Ormond Street Institute of Child Health, University College London, London, UK
- Ulverscroft Vision Research Group, University College London, London, UK
| | | | - Cécile Delcourt
- Université de Bordeaux, Inserm, Bordeaux Population Health Research Center, team LEHA, UMR 1219, F-33000 Bordeaux, France
| | - Céline Bellenguez
- Institut Pasteur de Lille, Lille, France
- Inserm, U1167, RID-AGE - Risk factors and molecular determinants of aging-related diseases, Lille, France
- Université de Lille, U1167 - Excellence Laboratory LabEx DISTALZ, Lille, France
| | - Janina S. Ried
- Institute of Genetic Epidemiology, Helmholtz Zentrum München—German Research Center for Environmental Health, Neuherberg, Germany
| | - Arthur A. Bergen
- Department of Clinical Genetics, Academic Medical Center, Amsterdam, The Netherlands
- Department of Ophthalmology, Academic Medical Center, Amsterdam, The Netherlands
- The Netherlands Institute for Neurosciences (NIN-KNAW), Amsterdam, The Netherlands
| | - Thomas Meitinger
- Institute of Human Genetics, Helmholtz Zentrum München, Neuherberg, Germany
- Institute of Human Genetics, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Christian Gieger
- Institute of Genetic Epidemiology, Helmholtz Zentrum München—German Research Center for Environmental Health, Neuherberg, Germany
| | - Tien Yin Wong
- Academic Medicine Research Institute, Singapore
- Retino Center, Singapore National Eye Centre, Singapore, Singapore
| | - Alex W. Hewitt
- Centre for Eye Research Australia, Ophthalmology, Department of Surgery, University of Melbourne, Royal Victorian Eye and Ear Hospital, Melbourne, Australia
- Department of Ophthalmology, Menzies Institute of Medical Research, University of Tasmania, Hobart, Australia
- Centre for Ophthalmology and Visual Science, Lions Eye Institute, University of Western Australia, Perth, Australia
| | - David A. Mackey
- Centre for Eye Research Australia, Ophthalmology, Department of Surgery, University of Melbourne, Royal Victorian Eye and Ear Hospital, Melbourne, Australia
- Department of Ophthalmology, Menzies Institute of Medical Research, University of Tasmania, Hobart, Australia
- Centre for Ophthalmology and Visual Science, Lions Eye Institute, University of Western Australia, Perth, Australia
| | - Claire L. Simpson
- Computational and Statistical Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, USA
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Sciences Center, Memphis, Tenessee
| | - Norbert Pfeiffer
- Department of Ophthalmology, University Medical Center Mainz, Mainz, Germany
| | - Olavi Pärssinen
- Department of Ophthalmology, Central Hospital of Central Finland, Jyväskylä, Finland
- Gerontology Research Center, Faculty of Sport and Health Sciences, University of Jyväskylä, Jyväskylä, Finland
| | - Paul N. Baird
- Centre for Eye Research Australia, Ophthalmology, Department of Surgery, University of Melbourne, Royal Victorian Eye and Ear Hospital, Melbourne, Australia
| | - Veronique Vitart
- MRC Human Genetics Unit, MRC Institute of Genetics & Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Najaf Amin
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | | | - Joan E. Bailey-Wilson
- Computational and Statistical Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Terri L. Young
- Department of Ophthalmology and Visual Sciences, University of Wisconsin–Madison, Madison, Wisconsin, USA
| | - Seang-Mei Saw
- Saw Swee Hock School of Public Health, National University Health Systems, National University of Singapore, Singapore
- Myopia Research Group, Singapore Eye Research Institute, Singapore National Eye Centre, Singapore
| | - Dwight Stambolian
- Department of Ophthalmology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Stuart MacGregor
- Statistical Genetics, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | | | | | - Christopher J. Hammond
- Section of Academic Ophthalmology, School of Life Course Sciences, King’s College London, London, UK
| | - Caroline C.W. Klaver
- Department of Ophthalmology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Ophthalmology, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
49
|
Agrawal A, Chou YL, Carey CE, Baranger DAA, Zhang B, Sherva R, Wetherill L, Kapoor M, Wang JC, Bertelsen S, Anokhin AP, Hesselbrock V, Kramer J, Lynskey MT, Meyers JL, Nurnberger JI, Rice JP, Tischfield J, Bierut LJ, Degenhardt L, Farrer LA, Gelernter J, Hariri AR, Heath AC, Kranzler HR, Madden PAF, Martin NG, Montgomery GW, Porjesz B, Wang T, Whitfield JB, Edenberg HJ, Foroud T, Goate AM, Bogdan R, Nelson EC. Genome-wide association study identifies a novel locus for cannabis dependence. Mol Psychiatry 2018; 23:1293-1302. [PMID: 29112194 PMCID: PMC5938138 DOI: 10.1038/mp.2017.200] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Revised: 06/26/2017] [Accepted: 07/13/2017] [Indexed: 01/01/2023]
Abstract
Despite moderate heritability, only one study has identified genome-wide significant loci for cannabis-related phenotypes. We conducted meta-analyses of genome-wide association study data on 2080 cannabis-dependent cases and 6435 cannabis-exposed controls of European descent. A cluster of correlated single-nucleotide polymorphisms (SNPs) in a novel region on chromosome 10 was genome-wide significant (lowest P=1.3E-8). Among the SNPs, rs1409568 showed enrichment for H3K4me1 and H3K427ac marks, suggesting its role as an enhancer in addiction-relevant brain regions, such as the dorsolateral prefrontal cortex and the angular and cingulate gyri. This SNP is also predicted to modify binding scores for several transcription factors. We found modest evidence for replication for rs1409568 in an independent cohort of African American (896 cases and 1591 controls; P=0.03) but not European American (EA; 781 cases and 1905 controls) participants. The combined meta-analysis (3757 cases and 9931 controls) indicated trend-level significance for rs1409568 (P=2.85E-7). No genome-wide significant loci emerged for cannabis dependence criterion count (n=8050). There was also evidence that the minor allele of rs1409568 was associated with a 2.1% increase in right hippocampal volume in an independent sample of 430 EA college students (fwe-P=0.008). The identification and characterization of genome-wide significant loci for cannabis dependence is among the first steps toward understanding the biological contributions to the etiology of this psychiatric disorder, which appears to be rising in some developed nations.
Collapse
Affiliation(s)
- Arpana Agrawal
- Washington University School of Medicine, Dept. of Psychiatry, 660 S. Euclid, CB 8134, Saint Louis, MO 63110, USA
| | - Yi-Ling Chou
- Washington University School of Medicine, Dept. of Psychiatry, 660 S. Euclid, CB 8134, Saint Louis, MO 63110, USA
| | - Caitlin E. Carey
- Washington University in St. Louis, Dept. of Psychological and Brain Sciences, St. Louis, MO, USA
| | - David A. A. Baranger
- Washington University in St. Louis, Dept. of Psychological and Brain Sciences, St. Louis, MO, USA
| | - Bo Zhang
- Washington University School of Medicine, Dept. of Developmental Biology, St. Louis, MO, USA
| | - Richard Sherva
- Boston University School of Medicine, Dept. of Medicine (Biomedical Genetics), Boston, MA, USA
| | - Leah Wetherill
- Indiana University School of Medicine, Dept. of Medical and Molecular Genetics, Indianapolis, IN, USA
| | - Manav Kapoor
- Icahn School of Medicine at Mount Sinai, Dept. of Neuroscience, New York, NY USA
| | - Jen-Chyong Wang
- Icahn School of Medicine at Mount Sinai, Dept. of Neuroscience, New York, NY USA
| | - Sarah Bertelsen
- Icahn School of Medicine at Mount Sinai, Dept. of Neuroscience, New York, NY USA
| | - Andrey P Anokhin
- Washington University School of Medicine, Dept. of Psychiatry, 660 S. Euclid, CB 8134, Saint Louis, MO 63110, USA
| | - Victor Hesselbrock
- University of Connecticut Health, Dept. of Psychiatry, Farmington, CT, USA
| | - John Kramer
- University of Iowa Carver College of Medicine, Dept. of Psychiatry, Iowa City, IA USA
| | - Michael T. Lynskey
- King’s College, Institute of Psychiatry, Psychology and Neuroscience, Addictions Department, London, UK
| | - Jacquelyn L. Meyers
- State University of New York, Downstate Medical Center, Dept. of Psychiatry, Brooklyn, NY USA
| | - John I Nurnberger
- Indiana University School of Medicine, Depts. of Psychiatry and Medical and Molecular Genetics, and Stark Neuroscience Center, Indianapolis, IN, USA
| | - John P. Rice
- Washington University School of Medicine, Dept. of Psychiatry, 660 S. Euclid, CB 8134, Saint Louis, MO 63110, USA
| | - Jay Tischfield
- Rutgers, The State University of New Jersey: New Brunswick, NJ, United States
| | - Laura J. Bierut
- Washington University School of Medicine, Dept. of Psychiatry, 660 S. Euclid, CB 8134, Saint Louis, MO 63110, USA
| | - Louisa Degenhardt
- National Drug and Alcohol Research Centre, University of New South Wales, Sydney, Australia
| | - Lindsay A Farrer
- Boston University School of Medicine, Dept. of Medicine (Biomedical Genetics), Boston, MA, USA
| | - Joel Gelernter
- Yale University School of Medicine: New Haven, CT, USA
- US Department of Veterans Affairs: West Haven, CT, USA
| | - Ahmad R. Hariri
- Duke University, Department of Psychology and Neuroscience, Durham, NC, USA
| | - Andrew C. Heath
- Washington University School of Medicine, Dept. of Psychiatry, 660 S. Euclid, CB 8134, Saint Louis, MO 63110, USA
| | - Henry R. Kranzler
- Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, VISN 4 MIRECC, Crescenz VAMC, Philadelphia, PA, USA
| | - Pamela A. F. Madden
- Washington University School of Medicine, Dept. of Psychiatry, 660 S. Euclid, CB 8134, Saint Louis, MO 63110, USA
| | | | - Grant W Montgomery
- University of Queensland, Institute for Molecular Bioscience, Queensland, Australia
| | - Bernice Porjesz
- State University of New York, Downstate Medical Center, Dept. of Psychiatry, Brooklyn, NY USA
| | - Ting Wang
- Washington University School of Medicine, Department of Genetics, St. Louis, MO, USA
| | | | - Howard J. Edenberg
- Indiana University School of Medicine, Dept. of Medical and Molecular Genetics, Indianapolis, IN, USA
- Indiana University, Dept. of Biochemistry and Molecular Biology, Indianapolis, IN, USA
| | - Tatiana Foroud
- Indiana University School of Medicine, Dept. of Medical and Molecular Genetics, Indianapolis, IN, USA
| | - Alison M. Goate
- Icahn School of Medicine at Mount Sinai, Dept. of Neuroscience, New York, NY USA
| | - Ryan Bogdan
- Washington University in St. Louis, Dept. of Psychological and Brain Sciences, St. Louis, MO, USA
| | - Elliot C. Nelson
- Washington University School of Medicine, Dept. of Psychiatry, 660 S. Euclid, CB 8134, Saint Louis, MO 63110, USA
| |
Collapse
|
50
|
A Genome-Wide Association Study for Host Resistance to Ostreid Herpesvirus in Pacific Oysters ( Crassostrea gigas). G3-GENES GENOMES GENETICS 2018; 8:1273-1280. [PMID: 29472307 PMCID: PMC5873916 DOI: 10.1534/g3.118.200113] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Ostreid herpesvirus (OsHV) can cause mass mortality events in Pacific oyster aquaculture. While various factors impact on the severity of outbreaks, it is clear that genetic resistance of the host is an important determinant of mortality levels. This raises the possibility of selective breeding strategies to improve the genetic resistance of farmed oyster stocks, thereby contributing to disease control. Traditional selective breeding can be augmented by use of genetic markers, either via marker-assisted or genomic selection. The aim of the current study was to investigate the genetic architecture of resistance to OsHV in Pacific oyster, to identify genomic regions containing putative resistance genes, and to inform the use of genomics to enhance efforts to breed for resistance. To achieve this, a population of ∼1,000 juvenile oysters were experimentally challenged with a virulent form of OsHV, with samples taken from mortalities and survivors for genotyping and qPCR measurement of viral load. The samples were genotyped using a recently-developed SNP array, and the genotype data were used to reconstruct the pedigree. Using these pedigree and genotype data, the first high density linkage map was constructed for Pacific oyster, containing 20,353 SNPs mapped to the ten pairs of chromosomes. Genetic parameters for resistance to OsHV were estimated, indicating a significant but low heritability for the binary trait of survival and also for viral load measures (h2 0.12 – 0.25). A genome-wide association study highlighted a region of linkage group 6 containing a significant QTL affecting host resistance. These results are an important step toward identification of genes underlying resistance to OsHV in oyster, and a step toward applying genomic data to enhance selective breeding for disease resistance in oyster aquaculture.
Collapse
|