18
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      A high resolution atlas of gene expression in the domestic sheep ( Ovis aries)

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Sheep are a key source of meat, milk and fibre for the global livestock sector, and an important biomedical model. Global analysis of gene expression across multiple tissues has aided genome annotation and supported functional annotation of mammalian genes. We present a large-scale RNA-Seq dataset representing all the major organ systems from adult sheep and from several juvenile, neonatal and prenatal developmental time points. The Ovis aries reference genome (Oar v3.1) includes 27,504 genes (20,921 protein coding), of which 25,350 (19,921 protein coding) had detectable expression in at least one tissue in the sheep gene expression atlas dataset. Network-based cluster analysis of this dataset grouped genes according to their expression pattern. The principle of ‘guilt by association’ was used to infer the function of uncharacterised genes from their co-expression with genes of known function. We describe the overall transcriptional signatures present in the sheep gene expression atlas and assign those signatures, where possible, to specific cell populations or pathways. The findings are related to innate immunity by focusing on clusters with an immune signature, and to the advantages of cross-breeding by examining the patterns of genes exhibiting the greatest expression differences between purebred and crossbred animals. This high-resolution gene expression atlas for sheep is, to our knowledge, the largest transcriptomic dataset from any livestock species to date. It provides a resource to improve the annotation of the current reference genome for sheep, presenting a model transcriptome for ruminants and insight into gene, cell and tissue function at multiple developmental stages.

          Author summary

          Sheep are ruminant mammals kept as livestock for the production of meat, milk and wool in agricultural industries across the globe. Genetic and genomic information can be used to improve production traits such as growth rate and health and fitness traits including disease resilience. The sheep genome is, however, missing important information relating to gene function and many genes, which may be important for productivity, have no informative gene name. This can be remedied using RNA-Sequencing to generate a global expression profile of all protein-coding genes, across multiple organ systems and developmental stages. Clustering genes based on their expression profile across tissues and cells allows us to assign function to those genes. If, for example, a gene with no informative gene name is expressed in macrophages and is found within a cluster of known macrophage related genes it is likely to be involved in macrophage function and play a role in innate immunity. This information improves the quality of the reference genome and provides insight into biological processes underlying the complex traits that influence the productivity of sheep and other livestock species.

          Related collections

          Most cited references95

          • Record: found
          • Abstract: found
          • Article: not found

          The transcriptional landscape of the mammalian genome.

          This study describes comprehensive polling of transcription start and termination sites and analysis of previously unidentified full-length complementary DNAs derived from the mouse genome. We identify the 5' and 3' boundaries of 181,047 transcripts with extensive variation in transcripts arising from alternative promoter usage, splicing, and polyadenylation. There are 16,247 new mouse protein-coding transcripts, including 5154 encoding previously unidentified proteins. Genomic mapping of the transcriptome reveals transcriptional forests, with overlapping transcription on both strands, separated by deserts in which few transcripts are observed. The data provide a comprehensive platform for the comparative analysis of mammalian transcriptional regulation in differentiation and development.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            A mutation creating a potential illegitimate microRNA target site in the myostatin gene affects muscularity in sheep.

            Texel sheep are renowned for their exceptional meatiness. To identify the genes underlying this economically important feature, we performed a whole-genome scan in a Romanov x Texel F2 population. We mapped a quantitative trait locus with a major effect on muscle mass to chromosome 2 and subsequently fine-mapped it to a chromosome interval encompassing the myostatin (GDF8) gene. We herein demonstrate that the GDF8 allele of Texel sheep is characterized by a G to A transition in the 3' UTR that creates a target site for mir1 and mir206, microRNAs (miRNAs) that are highly expressed in skeletal muscle. This causes translational inhibition of the myostatin gene and hence contributes to the muscular hypertrophy of Texel sheep. Analysis of SNP databases for humans and mice demonstrates that mutations creating or destroying putative miRNA target sites are abundant and might be important effectors of phenotypic variation.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              IL-27, a heterodimeric cytokine composed of EBI3 and p28 protein, induces proliferation of naive CD4+ T cells.

              An efficient Th1-driven adaptive immune response requires activation of the T cell receptor and secretion of the T cell stimulatory cytokine IL-12 by activated antigen-presenting cells. IL-12 triggers Th1 polarization of naive CD4(+) T cells and secretion of IFN-gamma. We describe a new heterodimeric cytokine termed IL-27 that consists of EBI3, an IL-12p40-related protein, and p28, a newly discovered IL-12p35-related polypeptide. IL-27 is an early product of activated antigen-presenting cells and drives rapid clonal expansion of naive but not memory CD4(+) T cells. It also strongly synergizes with IL-12 to trigger IFN-gamma production of naive CD4(+) T cells. IL-27 mediates its biologic effects through the orphan cytokine receptor WSX-1/TCCR.
                Bookmark

                Author and article information

                Contributors
                Role: Data curationRole: Formal analysisRole: InvestigationRole: MethodologyRole: Project administrationRole: ValidationRole: VisualizationRole: Writing – original draft
                Role: Data curationRole: Formal analysisRole: InvestigationRole: MethodologyRole: SoftwareRole: ValidationRole: VisualizationRole: Writing – review & editing
                Role: Formal analysisRole: InvestigationRole: MethodologyRole: VisualizationRole: Writing – review & editing
                Role: InvestigationRole: MethodologyRole: Project administrationRole: Writing – review & editing
                Role: InvestigationRole: MethodologyRole: Writing – review & editing
                Role: InvestigationRole: MethodologyRole: Writing – review & editing
                Role: InvestigationRole: MethodologyRole: SupervisionRole: Writing – review & editing
                Role: Formal analysisRole: InvestigationRole: MethodologyRole: VisualizationRole: Writing – review & editing
                Role: Data curationRole: ResourcesRole: Software
                Role: Data curationRole: ResourcesRole: Software
                Role: ConceptualizationRole: Funding acquisitionRole: MethodologyRole: ValidationRole: Writing – review & editing
                Role: ConceptualizationRole: Funding acquisitionRole: Writing – review & editing
                Role: Formal analysisRole: MethodologyRole: VisualizationRole: Writing – review & editing
                Role: ConceptualizationRole: Formal analysisRole: Funding acquisitionRole: MethodologyRole: SupervisionRole: VisualizationRole: Writing – review & editing
                Role: ConceptualizationRole: Funding acquisitionRole: Project administrationRole: SupervisionRole: Writing – review & editing
                Role: ConceptualizationRole: Formal analysisRole: Funding acquisitionRole: MethodologyRole: Project administrationRole: SupervisionRole: Writing – review & editing
                Role: Editor
                Journal
                PLoS Genet
                PLoS Genet
                plos
                plosgen
                PLoS Genetics
                Public Library of Science (San Francisco, CA USA )
                1553-7390
                1553-7404
                15 September 2017
                September 2017
                : 13
                : 9
                : e1006997
                Affiliations
                [1 ] The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, Scotland, United Kingdom
                [2 ] Department of Integrative and Computational Biology, The Scripps Research Institute, La Jolla, CA, United States of America
                [3 ] Mater Research Institute and University of Queensland, Translational Research Institute, Woolloongabba, Queensland, Australia
                CSIRO, Australia, AUSTRALIA
                Author notes

                The authors have declared that no competing interests exist.

                [¤]

                Current address: Centre for Synthetic and Systems Biology, University of Edinburgh, Edinburgh, Scotland, United Kingdom.

                Author information
                http://orcid.org/0000-0002-9550-7407
                http://orcid.org/0000-0002-1049-0870
                http://orcid.org/0000-0002-2629-6124
                http://orcid.org/0000-0002-7084-4386
                Article
                PGENETICS-D-17-01139
                10.1371/journal.pgen.1006997
                5626511
                28915238
                a4048c0f-4534-4f32-a7b2-242876e0e626
                © 2017 Clark et al

                This is an open access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

                History
                : 8 June 2017
                : 24 August 2017
                Page count
                Figures: 6, Tables: 3, Pages: 38
                Funding
                Funded by: funder-id http://dx.doi.org/10.13039/501100000268, Biotechnology and Biological Sciences Research Council;
                Award ID: BB/L001209/1
                Award Recipient :
                Funded by: funder-id http://dx.doi.org/10.13039/501100000268, Biotechnology and Biological Sciences Research Council;
                Award ID: BBS/E/D/20211550
                Funded by: funder-id http://dx.doi.org/10.13039/501100000268, Biotechnology and Biological Sciences Research Council;
                Award ID: BBS/E/D/20211552
                Funded by: National Capability Grant
                Award ID: BBS/E/D/20310000
                Funded by: funder-id http://dx.doi.org/10.13039/100000057, National Institute of General Medical Sciences;
                Award ID: GM083924
                Funded by: National Environment Research Council
                Award ID: R8/H10/56
                Funded by: funder-id http://dx.doi.org/10.13039/501100000265, Medical Research Council;
                Award ID: MR/K001744/1
                Funded by: funder-id http://dx.doi.org/10.13039/501100000268, Biotechnology and Biological Sciences Research Council;
                Award ID: BB/J004243/1
                The work was supported by a Biotechnology and Biological Sciences Research Council (BBSRC) ( http://www.bbsrc.ac.uk) Grant BB/L001209/1 ('Functional Annotation of the Sheep Genome'). Also BBSRC Institute Strategic Program Grants: 'Farm Animal Genomics' (BBS/E/D/20211550), 'Transcriptomes, Networks and Systems' (BBS/E/D/20211552) and 'Blue Prints for Healthy Animals' (BB/P013732/1). Edinburgh Genomics is partly supported through core grants from BBSRC (BB/J004243/1), NERC ( http://www.nerc.ac.uk) (R8/H10/56) and the Medical Research Council (MRC) ( https://www.mrc.ac.uk) (MR/K001744/1). Upload of the data to BioGPS was supported by a National Institute of General Medical Sciences Grant ( https://www.nigms.nih.gov/) (GM083924). The funders had no role in the study design, data collection and analysis, decision to publish, or preparation of the manuscript.
                Categories
                Research Article
                Biology and Life Sciences
                Organisms
                Eukaryota
                Animals
                Vertebrates
                Amniotes
                Mammals
                Ruminants
                Sheep
                Biology and Life Sciences
                Genetics
                Gene Expression
                Biology and Life Sciences
                Agriculture
                Livestock
                Biology and Life Sciences
                Cell Biology
                Cellular Types
                Animal Cells
                Blood Cells
                White Blood Cells
                Macrophages
                Biology and Life Sciences
                Cell Biology
                Cellular Types
                Animal Cells
                Immune Cells
                White Blood Cells
                Macrophages
                Biology and Life Sciences
                Immunology
                Immune Cells
                White Blood Cells
                Macrophages
                Medicine and Health Sciences
                Immunology
                Immune Cells
                White Blood Cells
                Macrophages
                Biology and Life Sciences
                Organisms
                Eukaryota
                Animals
                Vertebrates
                Amniotes
                Mammals
                Swine
                Biology and life sciences
                Genetics
                DNA
                Forms of DNA
                Complementary DNA
                cDNA libraries
                Biology and life sciences
                Biochemistry
                Nucleic acids
                DNA
                Forms of DNA
                Complementary DNA
                cDNA libraries
                Biology and life sciences
                Genetics
                DNA
                DNA libraries
                cDNA libraries
                Biology and life sciences
                Biochemistry
                Nucleic acids
                DNA
                DNA libraries
                cDNA libraries
                Biology and Life Sciences
                Anatomy
                Reproductive System
                Ovaries
                Medicine and Health Sciences
                Anatomy
                Reproductive System
                Ovaries
                Biology and Life Sciences
                Genetics
                Genomics
                Animal Genomics
                Mammalian Genomics
                Custom metadata
                vor-update-to-uncorrected-proof
                2017-10-03
                The datasets supporting the conclusions of this article are available in the following locations. The raw read data for all new TxBF libraries generated for this project is deposited in the European Nucleotide Archive (ENA) under study accession number PRJEB19199 ( http://www.ebi.ac.uk/ena/data/view/PRJEB19199). We have made the files containing the expression estimates ( S1 Dataset, S2 Dataset and S3 Dataset) available for download through the University of Edinburgh DataShare portal ( http://dx.doi.org/10.7488/ds/2112), these can also be viewed and downloaded via BioGPS ( http://biogps.org/dataset/BDS_00015/sheep-atlas/). Sample metadata for all the tissue and cell samples collected has been deposited in the EBI BioSamples database under project identifier GSB-718 ( https://www.ebi.ac.uk/biosamples/groups/SAMEG317052). The raw read data from the 83 Texel samples incorporated into this dataset and previously published (Jiang et al. 2017; Science; 10.1126/science.1252806) is located in the European Nucleotide Archive (ENA) study accession PRJEB6169 ( http://www.ebi.ac.uk/ena/data/view/PRJEB6169). All experimental protocols are available on the FAANG consortium ftp site at http://ftp.faang.ebi.ac.uk/ftp/protocols/.

                Genetics
                Genetics

                Comments

                Comment on this article