51
|
Eppig JT. Mouse Genome Informatics (MGI) Resource: Genetic, Genomic, and Biological Knowledgebase for the Laboratory Mouse. ILAR J 2017; 58:17-41. [PMID: 28838066 PMCID: PMC5886341 DOI: 10.1093/ilar/ilx013] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Revised: 03/14/2017] [Accepted: 03/28/2017] [Indexed: 12/13/2022] Open
Abstract
The Mouse Genome Informatics (MGI) Resource supports basic, translational, and computational research by providing high-quality, integrated data on the genetics, genomics, and biology of the laboratory mouse. MGI serves a strategic role for the scientific community in facilitating biomedical, experimental, and computational studies investigating the genetics and processes of diseases and enabling the development and testing of new disease models and therapeutic interventions. This review describes the nexus of the body of growing genetic and biological data and the advances in computer technology in the late 1980s, including the World Wide Web, that together launched the beginnings of MGI. MGI develops and maintains a gold-standard resource that reflects the current state of knowledge, provides semantic and contextual data integration that fosters hypothesis testing, continually develops new and improved tools for searching and analysis, and partners with the scientific community to assure research data needs are met. Here we describe one slice of MGI relating to the development of community-wide large-scale mutagenesis and phenotyping projects and introduce ways to access and use these MGI data. References and links to additional MGI aspects are provided.
Collapse
Affiliation(s)
- Janan T. Eppig
- Janan T. Eppig, PhD, is Professor Emeritus at The Jackson Laboratory in Bar Harbor, Maine
| |
Collapse
|
52
|
Karp NA, Mason J, Beaudet AL, Benjamini Y, Bower L, Braun RE, Brown SDM, Chesler EJ, Dickinson ME, Flenniken AM, Fuchs H, Angelis MHD, Gao X, Guo S, Greenaway S, Heller R, Herault Y, Justice MJ, Kurbatova N, Lelliott CJ, Lloyd KCK, Mallon AM, Mank JE, Masuya H, McKerlie C, Meehan TF, Mott RF, Murray SA, Parkinson H, Ramirez-Solis R, Santos L, Seavitt JR, Smedley D, Sorg T, Speak AO, Steel KP, Svenson KL, Wakana S, West D, Wells S, Westerberg H, Yaacoby S, White JK. Prevalence of sexual dimorphism in mammalian phenotypic traits. Nat Commun 2017. [PMID: 28650954 PMCID: PMC5490203 DOI: 10.1038/ncomms15475] [Citation(s) in RCA: 174] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The role of sex in biomedical studies has often been overlooked, despite evidence of sexually dimorphic effects in some biological studies. Here, we used high-throughput phenotype data from 14,250 wildtype and 40,192 mutant mice (representing 2,186 knockout lines), analysed for up to 234 traits, and found a large proportion of mammalian traits both in wildtype and mutants are influenced by sex. This result has implications for interpreting disease phenotypes in animal models and humans. Systemic dissection of sexually dimorphic phenotypes in mice is lacking. Here, Karp and the International Mouse Phenotype Consortium show that approximately 10% of qualitative traits and 56% of quantitative traits in mice as measured in laboratory setting are sexually dimorphic.
Collapse
Affiliation(s)
- Natasha A Karp
- Mouse Informatics Group, The Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1SA, UK.,Quantitative Biology, AstraZeneca, Unit 310, Cambridge Science Park, Cambridge CB4 0WG, UK
| | - Jeremy Mason
- European Bioinformatics Institute (EMBL-EBI), European Molecular Biology Laboratory, Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| | - Arthur L Beaudet
- Human and Molecular Genetics, Baylor College of Medicine, 1 Baylor Plaza, Houston, Texas 77030, USA
| | - Yoav Benjamini
- Department of Statistics and O.R. School of Mathematical Sciences, Tel Aviv University, Tel Aviv 69978, Israel
| | - Lynette Bower
- Mouse Biology Program, University of California, 2795 Second Street, Suite 400, Davis, California 95618, USA
| | - Robert E Braun
- The Jackson Laboratory, 600 Main Street, Bar Harbor, Maine 04609, USA
| | | | - Elissa J Chesler
- The Jackson Laboratory, 600 Main Street, Bar Harbor, Maine 04609, USA
| | - Mary E Dickinson
- Molecular Physiology and Biophysics, Baylor College of Medicine, 1 Baylor Plaza, Houston, Texas 77030, USA
| | - Ann M Flenniken
- The Centre for Phenogenomics, 25 Orde Street, Toronto, Ontario, Canada M5T 3H7
| | - Helmut Fuchs
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, Ingolstädter Landstraße 1, Neuherberg 85764, Germany
| | - Martin Hrabe de Angelis
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, Ingolstädter Landstraße 1, Neuherberg 85764, Germany.,School of Life Science Weihenstephan, Technische Universität München, Alte Akademie 8, Freising 85354, Germany.,German Center for Diabetes Research (DZD), Ingostädter Landstr. 1, Neuherberg 85764, Germany
| | - Xiang Gao
- Model Animal Research Center, Nanjing University, 12 Xuefu Road, Pukou, Nanjing, Jiangsu 210061, China
| | - Shiying Guo
- Model Animal Research Center, Nanjing University, 12 Xuefu Road, Pukou, Nanjing, Jiangsu 210061, China
| | | | - Ruth Heller
- Department of Statistics and O.R. School of Mathematical Sciences, Tel Aviv University, Tel Aviv 69978, Israel
| | - Yann Herault
- CELPHEDIA, PHENOMIN, Institut Clinique de la Souris, 1 Rue Laurent Fries, Illkirch 67404, France.,Institut de Génétique et de Biologie Moléculaire et Cellulaire, 1 Rue Laurent Fries, Illkirch 67404, France.,Centre National de la Recherche Scientifique, UMR7104, 1 rue Laurent Fries, Illkirch 67404, France.,Institut National de la Santé et de la Recherche Médicale, U964, 1 rue Laurent Fries, Illkirch 67404, France.,Université de Strasbourg, 1 rue Laurent Fries, Illkirch 67404, France
| | - Monica J Justice
- The Hospital for Sick Children, 555 University Avenue, Toronto, Ontario, Canada M5G 1X8
| | - Natalja Kurbatova
- Department of Statistics and O.R. School of Mathematical Sciences, Tel Aviv University, Tel Aviv 69978, Israel
| | - Christopher J Lelliott
- Mouse Genetics Project, The Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1SA, UK
| | - K C Kent Lloyd
- Mouse Biology Program, University of California, 2795 Second Street, Suite 400, Davis, California 95618, USA
| | | | - Judith E Mank
- Department of Genetics, Evolution &Environment, University College London, Gower Street, London WC1E 6BT, UK
| | - Hiroshi Masuya
- BioResource Center, RIKEN, 3-1-1 Koyadai, Tsukuba, Ibaraki 305-0074, Japan
| | - Colin McKerlie
- The Centre for Phenogenomics, 25 Orde Street, Toronto, Ontario, Canada M5T 3H7.,The Hospital for Sick Children, 686 Bay Street, Toronto, Ontario, Canada M5G 0A4
| | - Terrence F Meehan
- European Bioinformatics Institute (EMBL-EBI), European Molecular Biology Laboratory, Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| | - Richard F Mott
- Genetics Institute, University College London, Gower Street, London WC1E 6BT, UK
| | - Stephen A Murray
- The Jackson Laboratory, 600 Main Street, Bar Harbor, Maine 04609, USA
| | - Helen Parkinson
- European Bioinformatics Institute (EMBL-EBI), European Molecular Biology Laboratory, Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| | - Ramiro Ramirez-Solis
- Mouse Genetics Project, The Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1SA, UK
| | - Luis Santos
- MRC Harwell Institute, Harwell Campus, Harwell OX11 0RD, UK
| | - John R Seavitt
- Human and Molecular Genetics, Baylor College of Medicine, 1 Baylor Plaza, Houston, Texas 77030, USA
| | - Damian Smedley
- Clinical Pharmacology, Queen Mary University of London, Gower Street, London WC1E 6BT, UK
| | - Tania Sorg
- CELPHEDIA, PHENOMIN, Institut Clinique de la Souris, 1 Rue Laurent Fries, Illkirch 67404, France.,Institut de Génétique et de Biologie Moléculaire et Cellulaire, 1 Rue Laurent Fries, Illkirch 67404, France.,Centre National de la Recherche Scientifique, UMR7104, 1 rue Laurent Fries, Illkirch 67404, France.,Institut National de la Santé et de la Recherche Médicale, U964, 1 rue Laurent Fries, Illkirch 67404, France.,Université de Strasbourg, 1 rue Laurent Fries, Illkirch 67404, France
| | - Anneliese O Speak
- Mouse Genetics Project, The Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1SA, UK
| | - Karen P Steel
- Mouse Genetics Project, The Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1SA, UK.,Wolfson Centre for Age-Related Diseases, King's College London, Wolfson Wing, Hodgkin Building, Guys Campus, London SE1 1UL, UK
| | - Karen L Svenson
- The Jackson Laboratory, 600 Main Street, Bar Harbor, Maine 04609, USA
| | | | - Shigeharu Wakana
- BioResource Center, RIKEN, 3-1-1 Koyadai, Tsukuba, Ibaraki 305-0074, Japan
| | - David West
- Children's Hospital Oakland Research Institute, 5700 Martin Luther King Jr Way, Oakland, California 94609, USA
| | - Sara Wells
- MRC Harwell Institute, Harwell Campus, Harwell OX11 0RD, UK
| | | | - Shay Yaacoby
- Department of Statistics and O.R. School of Mathematical Sciences, Tel Aviv University, Tel Aviv 69978, Israel
| | - Jacqueline K White
- The Jackson Laboratory, 600 Main Street, Bar Harbor, Maine 04609, USA.,Mouse Genetics Project, The Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1SA, UK
| |
Collapse
|
53
|
Gerasimčik N, He M, Baptista MAP, Severinson E, Westerberg LS. Deletion of Dock10 in B Cells Results in Normal Development but a Mild Deficiency upon In Vivo and In Vitro Stimulations. Front Immunol 2017; 8:491. [PMID: 28507547 PMCID: PMC5410582 DOI: 10.3389/fimmu.2017.00491] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 04/10/2017] [Indexed: 11/25/2022] Open
Abstract
We sought to identify genes necessary to induce cytoskeletal change in B cells. Using gene expression microarray, we compared B cells stimulated with interleukin-4 (IL-4) and anti-CD40 antibodies that induce B cell spreading, cell motility, tight aggregates, and extensive microvilli with B cells stimulated with lipopolysaccharide that lack these cytoskeletal changes. We identified 84 genes with 10-fold or greater expression in anti-CD40 + IL-4 stimulated B cells, one of these encoded the guanine nucleotide exchange factor (GEF) dedicator of cytokinesis 10 (Dock10). IL-4 selectively induced Dock10 expression in B cells. Using lacZ expression to monitor Dock10 promoter activity, we found that Dock10 was expressed at all stages during B cell development. However, specific deletion of Dock10 in B cells was associated with a mild phenotype with normal B cell development and normal B cell spreading, polarization, motility, chemotaxis, aggregation, and Ig class switching. Dock10-deficient B cells showed lower proliferation in response to anti-CD40 and IL-4 stimulation. Moreover, the IgG response to soluble antigen in vivo was lower when Dock10 was specifically deleted in B cells. Together, we found that most B cell responses were intact in the absence of Dock10. However, specific deletion of Dock10 in B cells was associated with a mild reduction in B cell activation in vitro and in vivo.
Collapse
Affiliation(s)
- Natalija Gerasimčik
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden.,Department of Rheumatology and Inflammation Research, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Minghui He
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden.,Department of Microbiology Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Marisa A P Baptista
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden.,Department of Microbiology Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden.,Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| | - Eva Severinson
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Lisa S Westerberg
- Department of Microbiology Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
54
|
Estimating the selective effects of heterozygous protein-truncating variants from human exome data. Nat Genet 2017; 49:806-810. [PMID: 28369035 PMCID: PMC5618255 DOI: 10.1038/ng.3831] [Citation(s) in RCA: 105] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 03/07/2017] [Indexed: 12/14/2022]
Abstract
The dispensability of individual genes for viability has interested generations of geneticists. For some genes it is essential to maintain two functional chromosomal copies, while others may tolerate the loss of one or both copies. Exome sequence data from 60,706 individuals provide sufficient observations of rare protein truncating variants (PTVs) to make genome-wide estimates of selection against heterozygous loss of gene function. The cumulative frequency of rare deleterious PTVs is primarily determined by the balance between incoming mutations and purifying selection rather than genetic drift. This enables the estimation of the genome-wide distribution of selection coefficients for heterozygous PTVs and corresponding Bayesian estimates for individual genes. The strength of selection can discriminate the severity, age of onset, and mode of inheritance in Mendelian exome sequencing cases. We find that genes under the strongest selection are enriched in embryonic lethal mouse knockouts, putatively cell-essential genes, Mendelian disease genes, and regulators of transcription. Screening by essentiality, we find a large set of genes under strong selection that likely have critical function but have not yet been extensively annotated in published literature.
Collapse
|
55
|
Thomas DC, Clare S, Sowerby JM, Pardo M, Juss JK, Goulding DA, van der Weyden L, Storisteanu D, Prakash A, Espéli M, Flint S, Lee JC, Hoenderdos K, Kane L, Harcourt K, Mukhopadhyay S, Umrania Y, Antrobus R, Nathan JA, Adams DJ, Bateman A, Choudhary JS, Lyons PA, Condliffe AM, Chilvers ER, Dougan G, Smith KG. Eros is a novel transmembrane protein that controls the phagocyte respiratory burst and is essential for innate immunity. J Exp Med 2017; 214:1111-1128. [PMID: 28351984 PMCID: PMC5379978 DOI: 10.1084/jem.20161382] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Revised: 12/20/2016] [Accepted: 01/20/2017] [Indexed: 02/02/2023] Open
Abstract
The phagocyte respiratory burst is crucial for innate immunity. The transfer of electrons to oxygen is mediated by a membrane-bound heterodimer, comprising gp91phox and p22phox subunits. Deficiency of either subunit leads to severe immunodeficiency. We describe Eros (essential for reactive oxygen species), a protein encoded by the previously undefined mouse gene bc017643, and show that it is essential for host defense via the phagocyte NAPDH oxidase. Eros is required for expression of the NADPH oxidase components, gp91phox and p22phox Consequently, Eros-deficient mice quickly succumb to infection. Eros also contributes to the formation of neutrophil extracellular traps (NETS) and impacts on the immune response to melanoma metastases. Eros is an ortholog of the plant protein Ycf4, which is necessary for expression of proteins of the photosynthetic photosystem 1 complex, itself also an NADPH oxio-reductase. We thus describe the key role of the previously uncharacterized protein Eros in host defense.
Collapse
Affiliation(s)
- David C. Thomas
- Department of Medicine, University of Cambridge, University of Cambridge School of Clinical Medicine, Cambridge CB2 0QQ, England, UK
| | - Simon Clare
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridgeshire CB10 1SA, England, UK
| | - John M. Sowerby
- Department of Medicine, University of Cambridge, University of Cambridge School of Clinical Medicine, Cambridge CB2 0QQ, England, UK
| | - Mercedes Pardo
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridgeshire CB10 1SA, England, UK
| | - Jatinder K. Juss
- Department of Medicine, University of Cambridge, University of Cambridge School of Clinical Medicine, Cambridge CB2 0QQ, England, UK
| | - David A. Goulding
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridgeshire CB10 1SA, England, UK
| | - Louise van der Weyden
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridgeshire CB10 1SA, England, UK
| | - Daniel Storisteanu
- Department of Medicine, University of Cambridge, University of Cambridge School of Clinical Medicine, Cambridge CB2 0QQ, England, UK
| | - Ananth Prakash
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SD, England, UK
| | - Marion Espéli
- Department of Medicine, University of Cambridge, University of Cambridge School of Clinical Medicine, Cambridge CB2 0QQ, England, UK
| | - Shaun Flint
- Department of Medicine, University of Cambridge, University of Cambridge School of Clinical Medicine, Cambridge CB2 0QQ, England, UK
| | - James C. Lee
- Department of Medicine, University of Cambridge, University of Cambridge School of Clinical Medicine, Cambridge CB2 0QQ, England, UK
| | - Kim Hoenderdos
- Cambridge Institute for Medical Research, University of Cambridge School of Clinical Medicine, Wellcome Trust/MRC Building, Cambridge Biomedical Campus, Cambridge CB2 0XY, England, UK
| | - Leanne Kane
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridgeshire CB10 1SA, England, UK
| | - Katherine Harcourt
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridgeshire CB10 1SA, England, UK
| | - Subhankar Mukhopadhyay
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridgeshire CB10 1SA, England, UK
| | - Yagnesh Umrania
- Cambridge Institute for Medical Research, University of Cambridge School of Clinical Medicine, Wellcome Trust/MRC Building, Cambridge Biomedical Campus, Cambridge CB2 0XY, England, UK
| | - Robin Antrobus
- Cambridge Institute for Medical Research, University of Cambridge School of Clinical Medicine, Wellcome Trust/MRC Building, Cambridge Biomedical Campus, Cambridge CB2 0XY, England, UK
| | - James A. Nathan
- Cambridge Institute for Medical Research, University of Cambridge School of Clinical Medicine, Wellcome Trust/MRC Building, Cambridge Biomedical Campus, Cambridge CB2 0XY, England, UK
| | - David J. Adams
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridgeshire CB10 1SA, England, UK
| | - Alex Bateman
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SD, England, UK
| | - Jyoti S. Choudhary
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridgeshire CB10 1SA, England, UK
| | - Paul A. Lyons
- Department of Medicine, University of Cambridge, University of Cambridge School of Clinical Medicine, Cambridge CB2 0QQ, England, UK
| | - Alison M. Condliffe
- Department of Medicine, University of Cambridge, University of Cambridge School of Clinical Medicine, Cambridge CB2 0QQ, England, UK
| | - Edwin R. Chilvers
- Department of Medicine, University of Cambridge, University of Cambridge School of Clinical Medicine, Cambridge CB2 0QQ, England, UK
| | - Gordon Dougan
- Department of Medicine, University of Cambridge, University of Cambridge School of Clinical Medicine, Cambridge CB2 0QQ, England, UK
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridgeshire CB10 1SA, England, UK
| | - Kenneth G.C. Smith
- Department of Medicine, University of Cambridge, University of Cambridge School of Clinical Medicine, Cambridge CB2 0QQ, England, UK
| |
Collapse
|
56
|
Geyer SH, Reissig L, Rose J, Wilson R, Prin F, Szumska D, Ramirez-Solis R, Tudor C, White J, Mohun TJ, Weninger WJ. A staging system for correct phenotype interpretation of mouse embryos harvested on embryonic day 14 (E14.5). J Anat 2017; 230:710-719. [PMID: 28185240 PMCID: PMC5382591 DOI: 10.1111/joa.12590] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/02/2016] [Indexed: 01/09/2023] Open
Abstract
We present a simple and quick system for accurately scoring the developmental progress of mouse embryos harvested on embryonic day 14 (E14.5). Based solely on the external appearance of the maturing forelimb, we provide a convenient way to distinguish six developmental sub‐stages. Using a variety of objective morphometric data obtained from the commonly used C57BL/6N mouse strain, we show that these stages correlate precisely with the growth of the entire embryo and its organs. Applying the new staging system to phenotype analyses of E14.5 embryos of 58 embryonic lethal null mutant lines from the DMDD research programme (https://dmdd.org.uk) and its pilot, we show that homozygous mutant embryos are frequently delayed in development. To demonstrate the importance of our staging system for correct phenotype interpretation, we describe stage‐specific changes of the palate, heart and gut, and provide examples in which correct diagnosis of malformations relies on correct staging.
Collapse
Affiliation(s)
- Stefan H Geyer
- Centre for Anatomy and Cell Biology & MIC, Medical University of Vienna, Vienna, Austria
| | - Lukas Reissig
- Centre for Anatomy and Cell Biology & MIC, Medical University of Vienna, Vienna, Austria
| | - Julia Rose
- Centre for Anatomy and Cell Biology & MIC, Medical University of Vienna, Vienna, Austria
| | - Robert Wilson
- The Francis Crick Institute Mill Hill Laboratory, London, UK
| | - Fabrice Prin
- The Francis Crick Institute Mill Hill Laboratory, London, UK
| | | | | | | | - Jacqui White
- Wellcome Trust Sanger Institute, Hinxton, Cambridge, UK
| | - Timothy J Mohun
- The Francis Crick Institute Mill Hill Laboratory, London, UK
| | - Wolfgang J Weninger
- Centre for Anatomy and Cell Biology & MIC, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
57
|
With mouse age comes wisdom: A review and suggestions of relevant mouse models for age-related conditions. Mech Ageing Dev 2016; 160:54-68. [DOI: 10.1016/j.mad.2016.07.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Revised: 07/07/2016] [Accepted: 07/15/2016] [Indexed: 12/14/2022]
|
58
|
Kõks S, Dogan S, Tuna BG, González-Navarro H, Potter P, Vandenbroucke RE. Mouse models of ageing and their relevance to disease. Mech Ageing Dev 2016; 160:41-53. [PMID: 27717883 DOI: 10.1016/j.mad.2016.10.001] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Revised: 09/26/2016] [Accepted: 10/03/2016] [Indexed: 12/28/2022]
Abstract
Ageing is a process that gradually increases the organism's vulnerability to death. It affects different biological pathways, and the underlying cellular mechanisms are complex. In view of the growing disease burden of ageing populations, increasing efforts are being invested in understanding the pathways and mechanisms of ageing. We review some mouse models commonly used in studies on ageing, highlight the advantages and disadvantages of the different strategies, and discuss their relevance to disease susceptibility. In addition to addressing the genetics and phenotypic analysis of mice, we discuss examples of models of delayed or accelerated ageing and their modulation by caloric restriction.
Collapse
Affiliation(s)
- Sulev Kõks
- University of Tartu, Tartu, Estonia and Estonian University of Life Sciences, Tartu, Estonia.
| | - Soner Dogan
- Yeditepe University, School of Medicine, Department of Medical Biology, Istanbul, Turkey.
| | - Bilge Guvenc Tuna
- Yeditepe University, School of Medicine, Department of Biophysics, Istanbul, Turkey.
| | - Herminia González-Navarro
- Institute of Health Research-INCLIVA, 46010 Valencia, Spain and CIBER de Diabetes y Enfermedades Metabólicas (CIBERDEM), 28029 Madrid, Spain.
| | - Paul Potter
- Mammalian Genetics Unit, MRC Harwell, Oxfordshire, UK.
| | - Roosmarijn E Vandenbroucke
- Inflammation Research Center, VIB, Ghent, Belgium, Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium.
| |
Collapse
|
59
|
Dickinson ME, Flenniken AM, Ji X, Teboul L, Wong MD, White JK, Meehan TF, Weninger WJ, Westerberg H, Adissu H, Baker CN, Bower L, Brown JM, Caddle LB, Chiani F, Clary D, Cleak J, Daly MJ, Denegre JM, Doe B, Dolan ME, Edie SM, Fuchs H, Gailus-Durner V, Galli A, Gambadoro A, Gallegos J, Guo S, Horner NR, Hsu CW, Johnson SJ, Kalaga S, Keith LC, Lanoue L, Lawson TN, Lek M, Mark M, Marschall S, Mason J, McElwee ML, Newbigging S, Nutter LM, Peterson KA, Ramirez-Solis R, Rowland DJ, Ryder E, Samocha KE, Seavitt JR, Selloum M, Szoke-Kovacs Z, Tamura M, Trainor AG, Tudose I, Wakana S, Warren J, Wendling O, West DB, Wong L, Yoshiki A, MacArthur DG, Tocchini-Valentini GP, Gao X, Flicek P, Bradley A, Skarnes WC, Justice MJ, Parkinson HE, Moore M, Wells S, Braun RE, Svenson KL, de Angelis MH, Herault Y, Mohun T, Mallon AM, Henkelman RM, Brown SD, Adams DJ, Lloyd KK, McKerlie C, Beaudet AL, Bucan M, Murray SA. High-throughput discovery of novel developmental phenotypes. Nature 2016; 537:508-514. [PMID: 27626380 PMCID: PMC5295821 DOI: 10.1038/nature19356] [Citation(s) in RCA: 844] [Impact Index Per Article: 93.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Accepted: 08/10/2016] [Indexed: 12/29/2022]
Abstract
Approximately one-third of all mammalian genes are essential for life. Phenotypes resulting from knockouts of these genes in mice have provided tremendous insight into gene function and congenital disorders. As part of the International Mouse Phenotyping Consortium effort to generate and phenotypically characterize 5,000 knockout mouse lines, here we identify 410 lethal genes during the production of the first 1,751 unique gene knockouts. Using a standardized phenotyping platform that incorporates high-resolution 3D imaging, we identify phenotypes at multiple time points for previously uncharacterized genes and additional phenotypes for genes with previously reported mutant phenotypes. Unexpectedly, our analysis reveals that incomplete penetrance and variable expressivity are common even on a defined genetic background. In addition, we show that human disease genes are enriched for essential genes, thus providing a dataset that facilitates the prioritization and validation of mutations identified in clinical sequencing efforts.
Collapse
Affiliation(s)
- Mary E. Dickinson
- Department of Molecular Physiology and Biophysics, Houston, Texas, USA
| | - Ann M. Flenniken
- The Centre for Phenogenomics, Toronto, Ontario, Canada
- Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Xiao Ji
- Genomics and Computational Biology Program, Perelman School of Medicine, University of Pennsylvania, Philadelphia PA 19104
| | - Lydia Teboul
- Medical Research Council Harwell (Mammalian Genetics Unit and Mary Lyon Centre), Harwell, Oxfordshire, UK
| | - Michael D. Wong
- The Centre for Phenogenomics, Toronto, Ontario, Canada
- Mouse Imaging Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Jacqueline K. White
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, UK
| | - Terrence F. Meehan
- European Molecular Biology Laboratory- European Bioinformatics Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, UK
| | - Wolfgang J. Weninger
- Centre for Anatomy and Cell Biology, Medical University of Vienna, Vienna, Austria
| | - Henrik Westerberg
- Medical Research Council Harwell (Mammalian Genetics Unit and Mary Lyon Centre), Harwell, Oxfordshire, UK
| | - Hibret Adissu
- The Centre for Phenogenomics, Toronto, Ontario, Canada
- The Hospital for Sick Children, Toronto, Ontario, Canada
| | | | - Lynette Bower
- Mouse Biology Program, University of California, Davis
| | - James M. Brown
- Medical Research Council Harwell (Mammalian Genetics Unit and Mary Lyon Centre), Harwell, Oxfordshire, UK
| | | | - Francesco Chiani
- Monterotondo Mouse Clinic, Italian National Research Council (CNR), Institute of Cell Biology and Neurobiology, Monterotondo Scalo, Itally
| | - Dave Clary
- Mouse Biology Program, University of California, Davis
| | - James Cleak
- Medical Research Council Harwell (Mammalian Genetics Unit and Mary Lyon Centre), Harwell, Oxfordshire, UK
| | - Mark J. Daly
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston MA, USA
- Program in Medical and Population Genetics, Broad Institute MIT and Harvard, Cambridge, MA, USA
| | | | - Brendan Doe
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, UK
| | | | | | - Helmut Fuchs
- Helmholtz Zentrum München, German Research Center for Environmental Health, Institute of Experimental Genetics and German Mouse Clinic, Neuherberg, Germany
| | - Valerie Gailus-Durner
- Helmholtz Zentrum München, German Research Center for Environmental Health, Institute of Experimental Genetics and German Mouse Clinic, Neuherberg, Germany
| | - Antonella Galli
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, UK
| | - Alessia Gambadoro
- Monterotondo Mouse Clinic, Italian National Research Council (CNR), Institute of Cell Biology and Neurobiology, Monterotondo Scalo, Itally
| | - Juan Gallegos
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX USA
| | - Shiying Guo
- SKL of Pharmaceutical Biotechnology and Model Animal Research Center, Collaborative Innovation Center for Genetics and Development, Nanjing Biomedical Research Institute, Nanjing University, China
| | - Neil R. Horner
- Medical Research Council Harwell (Mammalian Genetics Unit and Mary Lyon Centre), Harwell, Oxfordshire, UK
| | - Chih-wei Hsu
- Department of Molecular Physiology and Biophysics, Houston, Texas, USA
| | - Sara J. Johnson
- Medical Research Council Harwell (Mammalian Genetics Unit and Mary Lyon Centre), Harwell, Oxfordshire, UK
| | - Sowmya Kalaga
- Department of Molecular Physiology and Biophysics, Houston, Texas, USA
| | - Lance C. Keith
- Department of Molecular Physiology and Biophysics, Houston, Texas, USA
| | - Louise Lanoue
- Mouse Biology Program, University of California, Davis
| | - Thomas N. Lawson
- Medical Research Council Harwell (Mammalian Genetics Unit and Mary Lyon Centre), Harwell, Oxfordshire, UK
| | - Monkol Lek
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston MA, USA
- Program in Medical and Population Genetics, Broad Institute MIT and Harvard, Cambridge, MA, USA
| | - Manuel Mark
- Infrastructure Nationale PHENOMIN, Institut Clinique de la Souris (ICS), et Institut de Génétique Biologie Moléculaire et Cellulaire (IGBMC) CNRS, INSERM, University of Strasbourg, Illkirch-Graffenstaden, France
| | - Susan Marschall
- Helmholtz Zentrum München, German Research Center for Environmental Health, Institute of Experimental Genetics and German Mouse Clinic, Neuherberg, Germany
| | - Jeremy Mason
- European Molecular Biology Laboratory- European Bioinformatics Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, UK
| | | | - Susan Newbigging
- The Centre for Phenogenomics, Toronto, Ontario, Canada
- The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Lauryl M.J. Nutter
- The Centre for Phenogenomics, Toronto, Ontario, Canada
- The Hospital for Sick Children, Toronto, Ontario, Canada
| | | | - Ramiro Ramirez-Solis
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, UK
| | | | - Edward Ryder
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, UK
| | - Kaitlin E. Samocha
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston MA, USA
- Program in Medical and Population Genetics, Broad Institute MIT and Harvard, Cambridge, MA, USA
| | - John R. Seavitt
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX USA
| | - Mohammed Selloum
- Infrastructure Nationale PHENOMIN, Institut Clinique de la Souris (ICS), et Institut de Génétique Biologie Moléculaire et Cellulaire (IGBMC) CNRS, INSERM, University of Strasbourg, Illkirch-Graffenstaden, France
| | - Zsombor Szoke-Kovacs
- Medical Research Council Harwell (Mammalian Genetics Unit and Mary Lyon Centre), Harwell, Oxfordshire, UK
| | | | | | - Ilinca Tudose
- European Molecular Biology Laboratory- European Bioinformatics Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, UK
| | | | - Jonathan Warren
- European Molecular Biology Laboratory- European Bioinformatics Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, UK
| | - Olivia Wendling
- Infrastructure Nationale PHENOMIN, Institut Clinique de la Souris (ICS), et Institut de Génétique Biologie Moléculaire et Cellulaire (IGBMC) CNRS, INSERM, University of Strasbourg, Illkirch-Graffenstaden, France
| | - David B. West
- Children’s Hospital Oakland Research Institute, Oakland, CA 94609
| | - Leeyean Wong
- Department of Molecular Physiology and Biophysics, Houston, Texas, USA
| | | | | | - Daniel G. MacArthur
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston MA, USA
- Program in Medical and Population Genetics, Broad Institute MIT and Harvard, Cambridge, MA, USA
| | - Glauco P. Tocchini-Valentini
- Monterotondo Mouse Clinic, Italian National Research Council (CNR), Institute of Cell Biology and Neurobiology, Monterotondo Scalo, Itally
| | - Xiang Gao
- SKL of Pharmaceutical Biotechnology and Model Animal Research Center, Collaborative Innovation Center for Genetics and Development, Nanjing Biomedical Research Institute, Nanjing University, China
| | - Paul Flicek
- European Molecular Biology Laboratory- European Bioinformatics Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, UK
| | - Allan Bradley
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, UK
| | - William C. Skarnes
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, UK
| | - Monica J. Justice
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX USA
- The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Helen E. Parkinson
- European Molecular Biology Laboratory- European Bioinformatics Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, UK
| | | | - Sara Wells
- Medical Research Council Harwell (Mammalian Genetics Unit and Mary Lyon Centre), Harwell, Oxfordshire, UK
| | | | | | - Martin Hrabe de Angelis
- Helmholtz Zentrum München, German Research Center for Environmental Health, Institute of Experimental Genetics and German Mouse Clinic, Neuherberg, Germany
- Chair of Experimental Genetics, School of Life Science Weihenstephan, Technische Universität München, Freising
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Yann Herault
- Infrastructure Nationale PHENOMIN, Institut Clinique de la Souris (ICS), et Institut de Génétique Biologie Moléculaire et Cellulaire (IGBMC) CNRS, INSERM, University of Strasbourg, Illkirch-Graffenstaden, France
| | - Tim Mohun
- The Francis Crick Institute Mill Hill Laboratory, The Ridgeway, Mill Hill, London, UK
| | - Ann-Marie Mallon
- Medical Research Council Harwell (Mammalian Genetics Unit and Mary Lyon Centre), Harwell, Oxfordshire, UK
| | - R. Mark Henkelman
- The Centre for Phenogenomics, Toronto, Ontario, Canada
- Mouse Imaging Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Steve D.M. Brown
- Medical Research Council Harwell (Mammalian Genetics Unit and Mary Lyon Centre), Harwell, Oxfordshire, UK
| | - David J. Adams
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, UK
| | | | - Colin McKerlie
- The Centre for Phenogenomics, Toronto, Ontario, Canada
- The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Arthur L. Beaudet
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX USA
| | - Maja Bucan
- Departments of Genetics and Psychiatry, Perlman School of Medicine, University of Pennsylvania, Philadelphia PA 19104
| | | |
Collapse
|
60
|
Mouse Models for the Study of Synthesis, Secretion, and Action of Pituitary Gonadotropins. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2016; 143:49-84. [PMID: 27697204 DOI: 10.1016/bs.pmbts.2016.08.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Gonadotropins play fundamental roles in reproduction. More than 30years ago, Cga transgenic mice were generated, and more than 20years ago, the phenotypes of Cga null mice were reported. Since then, numerous mouse strains have been generated and characterized to address several questions in reproductive biology involving gonadotropin synthesis, secretion, and action. More recently, extragonadal expression, and in some cases, functions of gonadotropins in nongonadal tissues have been identified. Several genomic and proteomic approaches including novel mouse genome editing tools are available now. It is anticipated that these and other emerging technologies will be useful to build an integrated network of gonadotropin signaling pathways in various tissues. Undoubtedly, research on gonadotropins will continue to provide new knowledge and allow us transcend from benchside to the bedside.
Collapse
|
61
|
Jagannathan S, Bradley RK. Translational plasticity facilitates the accumulation of nonsense genetic variants in the human population. Genome Res 2016; 26:1639-1650. [PMID: 27646533 PMCID: PMC5131816 DOI: 10.1101/gr.205070.116] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 09/16/2016] [Indexed: 01/12/2023]
Abstract
Genetic variants that disrupt protein-coding DNA are ubiquitous in the human population, with about 100 such loss-of-function variants per individual. While most loss-of-function variants are rare, a subset have risen to high frequency and occur in a homozygous state in healthy individuals. It is unknown why these common variants are well tolerated, even though some affect essential genes implicated in Mendelian disease. Here, we combine genomic, proteomic, and biochemical data to demonstrate that many common nonsense variants do not ablate protein production from their host genes. We provide computational and experimental evidence for diverse mechanisms of gene rescue, including alternative splicing, stop codon readthrough, alternative translation initiation, and C-terminal truncation. Our results suggest a molecular explanation for the mild fitness costs of many common nonsense variants and indicate that translational plasticity plays a prominent role in shaping human genetic diversity.
Collapse
Affiliation(s)
- Sujatha Jagannathan
- Computational Biology Program, Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington 98109, USA.,Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington 98109, USA.,Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, Washington 98109, USA
| | - Robert K Bradley
- Computational Biology Program, Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington 98109, USA.,Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington 98109, USA
| |
Collapse
|
62
|
Dyment NA, Jiang X, Chen L, Hong SH, Adams DJ, Ackert-Bicknell C, Shin DG, Rowe DW. High-Throughput, Multi-Image Cryohistology of Mineralized Tissues. J Vis Exp 2016. [PMID: 27684089 DOI: 10.3791/54468] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
There is an increasing need for efficient phenotyping and histopathology of a variety of tissues. This phenotyping need is evident with the ambitious projects to disrupt every gene in the mouse genome. The research community needs rapid and inexpensive means to phenotype tissues via histology. Histological analyses of skeletal tissues are often time consuming and semi-quantitative at best, regularly requiring subjective interpretation of slides from trained individuals. Here, we present a cryohistological paradigm for efficient and inexpensive phenotyping of mineralized tissues. First, we present a novel method of tape-stabilized cryosectioning that preserves the morphology of mineralized tissues. These sections are then adhered rigidly to glass slides and imaged repeatedly over several rounds of staining. The resultant images are then aligned either manually or via computer software to yield composite stacks of several layered images. The protocol allows for co-localization of numerous molecular signals to specific cells within a given section. In addition, these fluorescent signals can be quantified objectively via computer software. This protocol overcomes many of the shortcomings associated with histology of mineralized tissues and can serve as a platform for high-throughput, high-content phenotyping of musculoskeletal tissues moving forward.
Collapse
Affiliation(s)
- Nathaniel A Dyment
- Department of Reconstructive Sciences, University of Connecticut Health Center;
| | - Xi Jiang
- Department of Reconstructive Sciences, University of Connecticut Health Center
| | - Li Chen
- Department of Reconstructive Sciences, University of Connecticut Health Center
| | - Seung-Hyun Hong
- Department of Computer Science and Engineering, University of Connecticut
| | - Douglas J Adams
- Department of Orthopaedic Surgery, University of Connecticut Health Center
| | | | - Dong-Guk Shin
- Department of Computer Science and Engineering, University of Connecticut
| | - David W Rowe
- Department of Reconstructive Sciences, University of Connecticut Health Center;
| |
Collapse
|
63
|
Longitudinal imaging of the ageing mouse. Mech Ageing Dev 2016; 160:93-116. [PMID: 27530773 DOI: 10.1016/j.mad.2016.08.001] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 07/30/2016] [Accepted: 08/04/2016] [Indexed: 12/13/2022]
Abstract
Several non-invasive imaging techniques are used to investigate the effect of pathologies and treatments over time in mouse models. Each preclinical in vivo technique provides longitudinal and quantitative measurements of changes in tissues and organs, which are fundamental for the evaluation of alterations in phenotype due to pathologies, interventions and treatments. However, it is still unclear how these imaging modalities can be used to study ageing with mice models. Almost all age related pathologies in mice such as osteoporosis, arthritis, diabetes, cancer, thrombi, dementia, to name a few, can be imaged in vivo by at least one longitudinal imaging modality. These measurements are the basis for quantification of treatment effects in the development phase of a novel treatment prior to its clinical testing. Furthermore, the non-invasive nature of such investigations allows the assessment of different tissue and organ phenotypes in the same animal and over time, providing the opportunity to study the dysfunction of multiple tissues associated with the ageing process. This review paper aims to provide an overview of the applications of the most commonly used in vivo imaging modalities used in mouse studies: micro-computed-tomography, preclinical magnetic-resonance-imaging, preclinical positron-emission-tomography, preclinical single photon emission computed tomography, ultrasound, intravital microscopy, and whole body optical imaging.
Collapse
|
64
|
Blitz IL, Fish MB, Cho KWY. Leapfrogging: primordial germ cell transplantation permits recovery of CRISPR/Cas9-induced mutations in essential genes. Development 2016; 143:2868-75. [PMID: 27385011 PMCID: PMC5004912 DOI: 10.1242/dev.138057] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Accepted: 06/15/2016] [Indexed: 01/07/2023]
Abstract
CRISPR/Cas9 genome editing is revolutionizing genetic loss-of-function analysis but technical limitations remain that slow progress when creating mutant lines. First, in conventional genetic breeding schemes, mosaic founder animals carrying mutant alleles are outcrossed to produce F1 heterozygotes. Phenotypic analysis occurs in the F2 generation following F1 intercrosses. Thus, mutant analyses will require multi-generational studies. Second, when targeting essential genes, efficient mutagenesis of founders is often lethal, preventing the acquisition of mature animals. Reducing mutagenesis levels may improve founder survival, but results in lower, more variable rates of germline transmission. Therefore, an efficient approach to study lethal mutations would be useful. To overcome these shortfalls, we introduce 'leapfrogging', a method combining efficient CRISPR mutagenesis with transplantation of mutated primordial germ cells into a wild-type host. Tested using Xenopus tropicalis, we show that founders containing transplants transmit mutant alleles with high efficiency. F1 offspring from intercrosses between F0 animals that carry embryonic lethal alleles recapitulate loss-of-function phenotypes, circumventing an entire generation of breeding. We anticipate that leapfrogging will be transferable to other species.
Collapse
Affiliation(s)
- Ira L Blitz
- 4410 Natural Sciences Building 2, Department of Developmental and Cell Biology, University of California, Irvine, CA 92697, USA
| | - Margaret B Fish
- 4410 Natural Sciences Building 2, Department of Developmental and Cell Biology, University of California, Irvine, CA 92697, USA
| | - Ken W Y Cho
- 4410 Natural Sciences Building 2, Department of Developmental and Cell Biology, University of California, Irvine, CA 92697, USA
| |
Collapse
|
65
|
Omori H, Ogaki S, Sakano D, Sato M, Umeda K, Takeda N, Nakagata N, Kume S. Changes in expression of C2cd4c in pancreatic endocrine cells during pancreatic development. FEBS Lett 2016; 590:2584-93. [PMID: 27349930 PMCID: PMC5129588 DOI: 10.1002/1873-3468.12271] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Revised: 06/20/2016] [Accepted: 06/21/2016] [Indexed: 11/10/2022]
Abstract
C2cd4c, encoded by a gene belonging to the C2cd4 family, contains a C2 domain conserved across species and is localized to the cytoplasm. To examine the role of C2cd4c in the pancreas, we studied its localization and generated C2cd4c knockout (KO) mice. C2cd4c was expressed in pancreatic endocrine progenitors at early embryonic stages. When endocrine cells arise from their precursors, C2cd4c is gradually confined to the insulin‐ and pancreatic polypeptide‐expressing cells of the endocrine. In the adult pancreas, C2cd4c is restricted to the beta cells. C2cd4c KO mice showed normal embryonic pancreatic development and adult pancreatic function. Thus, our results suggest that C2cd4c is dispensable for pancreatic development.
Collapse
Affiliation(s)
- Hisayoshi Omori
- Institute of Molecular Embryology and Genetics, Kumamoto University, Japan
| | - Soichiro Ogaki
- Institute of Molecular Embryology and Genetics, Kumamoto University, Japan.,Department of Life Science and Technology, School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Japan.,Division of Pharmacology, National Institute of Health Science, Kamiyoga, Setagaya-ku, Tokyo, Japan
| | - Daisuke Sakano
- Institute of Molecular Embryology and Genetics, Kumamoto University, Japan.,Department of Life Science and Technology, School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Japan
| | - Mutsumi Sato
- Institute of Molecular Embryology and Genetics, Kumamoto University, Japan
| | - Kahoko Umeda
- Institute of Molecular Embryology and Genetics, Kumamoto University, Japan.,HIGO program, Kumamoto University, Japan
| | - Naoki Takeda
- Division of Developmental Genetics, Institute of Resource Development and Analysis, Kumamoto University, Japan
| | - Naomi Nakagata
- Division of Reproductive Engineering, Institute of Resource Development and Analysis, Kumamoto University, Japan
| | - Shoen Kume
- Institute of Molecular Embryology and Genetics, Kumamoto University, Japan.,Department of Life Science and Technology, School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Japan
| |
Collapse
|
66
|
Delezie J, Dumont S, Sandu C, Reibel S, Pevet P, Challet E. Rev-erbα in the brain is essential for circadian food entrainment. Sci Rep 2016; 6:29386. [PMID: 27380954 PMCID: PMC4933951 DOI: 10.1038/srep29386] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Accepted: 06/20/2016] [Indexed: 01/07/2023] Open
Abstract
Foraging is costly in terms of time and energy. An endogenous food-entrainable system allows anticipation of predictable changes of food resources in nature. Yet the molecular mechanism that controls food anticipation in mammals remains elusive. Here we report that deletion of the clock component Rev-erbα impairs food entrainment in mice. Rev-erbα global knockout (GKO) mice subjected to restricted feeding showed reduced elevations of locomotor activity and body temperature prior to mealtime, regardless of the lighting conditions. The failure to properly anticipate food arrival was accompanied by a lack of phase-adjustment to mealtime of the clock protein PERIOD2 in the cerebellum, and by diminished expression of phosphorylated ERK 1/2 (p-ERK) during mealtime in the mediobasal hypothalamus and cerebellum. Furthermore, brain-specific knockout (BKO) mice for Rev-erbα display a defective suprachiasmatic clock, as evidenced by blunted daily activity under a light-dark cycle, altered free-running rhythm in constant darkness and impaired clock gene expression. Notably, brain deletion of Rev-erbα totally prevented food-anticipatory behaviour and thermogenesis. In response to restricted feeding, brain deletion of Rev-erbα impaired changes in clock gene expression in the hippocampus and cerebellum, but not in the liver. Our findings indicate that Rev-erbα is required for neural network-based prediction of food availability.
Collapse
Affiliation(s)
- Julien Delezie
- Regulation of circadian clocks team, Institute of Cellular and Integrative Neurosciences, UPR3212, Centre National de la Recherche Scientifique, University of Strasbourg, France
| | - Stéphanie Dumont
- Regulation of circadian clocks team, Institute of Cellular and Integrative Neurosciences, UPR3212, Centre National de la Recherche Scientifique, University of Strasbourg, France
| | - Cristina Sandu
- Regulation of circadian clocks team, Institute of Cellular and Integrative Neurosciences, UPR3212, Centre National de la Recherche Scientifique, University of Strasbourg, France
| | - Sophie Reibel
- Chronobiotron, UMS3415, CNRS, University of Strasbourg, France
| | - Paul Pevet
- Regulation of circadian clocks team, Institute of Cellular and Integrative Neurosciences, UPR3212, Centre National de la Recherche Scientifique, University of Strasbourg, France
| | - Etienne Challet
- Regulation of circadian clocks team, Institute of Cellular and Integrative Neurosciences, UPR3212, Centre National de la Recherche Scientifique, University of Strasbourg, France
| |
Collapse
|
67
|
Pedersen MT, Kooistra SM, Radzisheuskaya A, Laugesen A, Johansen JV, Hayward DG, Nilsson J, Agger K, Helin K. Continual removal of H3K9 promoter methylation by Jmjd2 demethylases is vital for ESC self-renewal and early development. EMBO J 2016; 35:1550-64. [PMID: 27266524 DOI: 10.15252/embj.201593317] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 05/06/2016] [Indexed: 12/12/2022] Open
Abstract
Chromatin-associated proteins are essential for the specification and maintenance of cell identity. They exert these functions through modulating and maintaining transcriptional patterns. To elucidate the functions of the Jmjd2 family of H3K9/H3K36 histone demethylases, we generated conditional Jmjd2a/Kdm4a, Jmjd2b/Kdm4b and Jmjd2c/Kdm4c/Gasc1 single, double and triple knockout mouse embryonic stem cells (ESCs). We report that while individual Jmjd2 family members are dispensable for ESC maintenance and embryogenesis, combined deficiency for specifically Jmjd2a and Jmjd2c leads to early embryonic lethality and impaired ESC self-renewal, with spontaneous differentiation towards primitive endoderm under permissive culture conditions. We further show that Jmjd2a and Jmjd2c both localize to H3K4me3-positive promoters, where they have widespread and redundant roles in preventing accumulation of H3K9me3 and H3K36me3. Jmjd2 catalytic activity is required for ESC maintenance, and increased H3K9me3 levels in knockout ESCs compromise the expression of several Jmjd2a/c targets, including genes that are important for ESC self-renewal. Thus, continual removal of H3K9 promoter methylation by Jmjd2 demethylases represents a novel mechanism ensuring transcriptional competence and stability of the pluripotent cell identity.
Collapse
Affiliation(s)
- Marianne Terndrup Pedersen
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark Centre for Epigenetics, University of Copenhagen, Copenhagen, Denmark
| | - Susanne Marije Kooistra
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark Centre for Epigenetics, University of Copenhagen, Copenhagen, Denmark
| | - Aliaksandra Radzisheuskaya
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark Centre for Epigenetics, University of Copenhagen, Copenhagen, Denmark
| | - Anne Laugesen
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark Centre for Epigenetics, University of Copenhagen, Copenhagen, Denmark The Danish Stem Cell Center (Danstem), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jens Vilstrup Johansen
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark
| | - Daniel Geoffrey Hayward
- The Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jakob Nilsson
- The Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Karl Agger
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark Centre for Epigenetics, University of Copenhagen, Copenhagen, Denmark
| | - Kristian Helin
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark Centre for Epigenetics, University of Copenhagen, Copenhagen, Denmark The Danish Stem Cell Center (Danstem), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
68
|
Bone WP, Washington NL, Buske OJ, Adams DR, Davis J, Draper D, Flynn ED, Girdea M, Godfrey R, Golas G, Groden C, Jacobsen J, Köhler S, Lee EMJ, Links AE, Markello TC, Mungall CJ, Nehrebecky M, Robinson PN, Sincan M, Soldatos AG, Tifft CJ, Toro C, Trang H, Valkanas E, Vasilevsky N, Wahl C, Wolfe LA, Boerkoel CF, Brudno M, Haendel MA, Gahl WA, Smedley D. Computational evaluation of exome sequence data using human and model organism phenotypes improves diagnostic efficiency. Genet Med 2016; 18:608-17. [PMID: 26562225 PMCID: PMC4916229 DOI: 10.1038/gim.2015.137] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2015] [Accepted: 08/27/2015] [Indexed: 01/18/2023] Open
Abstract
PURPOSE Medical diagnosis and molecular or biochemical confirmation typically rely on the knowledge of the clinician. Although this is very difficult in extremely rare diseases, we hypothesized that the recording of patient phenotypes in Human Phenotype Ontology (HPO) terms and computationally ranking putative disease-associated sequence variants improves diagnosis, particularly for patients with atypical clinical profiles. METHODS Using simulated exomes and the National Institutes of Health Undiagnosed Diseases Program (UDP) patient cohort and associated exome sequence, we tested our hypothesis using Exomiser. Exomiser ranks candidate variants based on patient phenotype similarity to (i) known disease-gene phenotypes, (ii) model organism phenotypes of candidate orthologs, and (iii) phenotypes of protein-protein association neighbors. RESULTS Benchmarking showed Exomiser ranked the causal variant as the top hit in 97% of known disease-gene associations and ranked the correct seeded variant in up to 87% when detectable disease-gene associations were unavailable. Using UDP data, Exomiser ranked the causative variant(s) within the top 10 variants for 11 previously diagnosed variants and achieved a diagnosis for 4 of 23 cases undiagnosed by clinical evaluation. CONCLUSION Structured phenotyping of patients and computational analysis are effective adjuncts for diagnosing patients with genetic disorders.Genet Med 18 6, 608-617.
Collapse
Affiliation(s)
- William P. Bone
- Undiagnosed Diseases Program, Common Fund, Office of the Director, National Institutes of Health, Bethesda, Maryland, USA
| | - Nicole L. Washington
- Genomics Division, Lawrence Berkeley National Laboratory, Berkeley, California, USA
| | - Orion J. Buske
- Centre for Computational Medicine Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Computer Science, University of Toronto, Toronto, Ontario, Canada
| | - David R. Adams
- Undiagnosed Diseases Program, Common Fund, Office of the Director, National Institutes of Health, Bethesda, Maryland, USA
- Medical Genetics Branch, National Human Genome Research Institute, Bethesda, Maryland, USA
| | - Joie Davis
- Undiagnosed Diseases Program, Common Fund, Office of the Director, National Institutes of Health, Bethesda, Maryland, USA
| | - David Draper
- Undiagnosed Diseases Program, Common Fund, Office of the Director, National Institutes of Health, Bethesda, Maryland, USA
| | - Elise D. Flynn
- Undiagnosed Diseases Program, Common Fund, Office of the Director, National Institutes of Health, Bethesda, Maryland, USA
| | - Marta Girdea
- Centre for Computational Medicine Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Computer Science, University of Toronto, Toronto, Ontario, Canada
| | - Rena Godfrey
- Undiagnosed Diseases Program, Common Fund, Office of the Director, National Institutes of Health, Bethesda, Maryland, USA
| | - Gretchen Golas
- Undiagnosed Diseases Program, Common Fund, Office of the Director, National Institutes of Health, Bethesda, Maryland, USA
| | - Catherine Groden
- Undiagnosed Diseases Program, Common Fund, Office of the Director, National Institutes of Health, Bethesda, Maryland, USA
| | - Julius Jacobsen
- Skarnes Faculty group, Wellcome Trust Sanger Institute, Hinxton, UK
| | - Sebastian Köhler
- Institute for Medical Genetics and Human Genetics, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Elizabeth M. J. Lee
- Undiagnosed Diseases Program, Common Fund, Office of the Director, National Institutes of Health, Bethesda, Maryland, USA
| | - Amanda E. Links
- Undiagnosed Diseases Program, Common Fund, Office of the Director, National Institutes of Health, Bethesda, Maryland, USA
| | - Thomas C. Markello
- Undiagnosed Diseases Program, Common Fund, Office of the Director, National Institutes of Health, Bethesda, Maryland, USA
| | | | - Michele Nehrebecky
- Undiagnosed Diseases Program, Common Fund, Office of the Director, National Institutes of Health, Bethesda, Maryland, USA
| | - Peter N. Robinson
- Institute for Medical Genetics and Human Genetics, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Murat Sincan
- Undiagnosed Diseases Program, Common Fund, Office of the Director, National Institutes of Health, Bethesda, Maryland, USA
| | - Ariane G. Soldatos
- Undiagnosed Diseases Program, Common Fund, Office of the Director, National Institutes of Health, Bethesda, Maryland, USA
| | - Cynthia J. Tifft
- Undiagnosed Diseases Program, Common Fund, Office of the Director, National Institutes of Health, Bethesda, Maryland, USA
- Medical Genetics Branch, National Human Genome Research Institute, Bethesda, Maryland, USA
| | - Camilo Toro
- Undiagnosed Diseases Program, Common Fund, Office of the Director, National Institutes of Health, Bethesda, Maryland, USA
| | - Heather Trang
- Centre for Computational Medicine Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Computer Science, University of Toronto, Toronto, Ontario, Canada
| | - Elise Valkanas
- Undiagnosed Diseases Program, Common Fund, Office of the Director, National Institutes of Health, Bethesda, Maryland, USA
| | - Nicole Vasilevsky
- Library; and Department of Medical Informatics and Epidemiology, Oregon Health & Science University, Portland, Oregon, USA
| | - Colleen Wahl
- Undiagnosed Diseases Program, Common Fund, Office of the Director, National Institutes of Health, Bethesda, Maryland, USA
| | - Lynne A. Wolfe
- Undiagnosed Diseases Program, Common Fund, Office of the Director, National Institutes of Health, Bethesda, Maryland, USA
| | - Cornelius F. Boerkoel
- Undiagnosed Diseases Program, Common Fund, Office of the Director, National Institutes of Health, Bethesda, Maryland, USA
| | - Michael Brudno
- Centre for Computational Medicine Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Computer Science, University of Toronto, Toronto, Ontario, Canada
| | - Melissa A. Haendel
- Library; and Department of Medical Informatics and Epidemiology, Oregon Health & Science University, Portland, Oregon, USA
| | - William A. Gahl
- Undiagnosed Diseases Program, Common Fund, Office of the Director, National Institutes of Health, Bethesda, Maryland, USA
- Medical Genetics Branch, National Human Genome Research Institute, Bethesda, Maryland, USA
| | - Damian Smedley
- Skarnes Faculty group, Wellcome Trust Sanger Institute, Hinxton, UK
| |
Collapse
|
69
|
McDonald JU, Kaforou M, Clare S, Hale C, Ivanova M, Huntley D, Dorner M, Wright VJ, Levin M, Martinon-Torres F, Herberg JA, Tregoning JS. A Simple Screening Approach To Prioritize Genes for Functional Analysis Identifies a Role for Interferon Regulatory Factor 7 in the Control of Respiratory Syncytial Virus Disease. mSystems 2016; 1:e00051-16. [PMID: 27822537 PMCID: PMC5069771 DOI: 10.1128/msystems.00051-16] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2016] [Accepted: 05/26/2016] [Indexed: 12/21/2022] Open
Abstract
Greater understanding of the functions of host gene products in response to infection is required. While many of these genes enable pathogen clearance, some enhance pathogen growth or contribute to disease symptoms. Many studies have profiled transcriptomic and proteomic responses to infection, generating large data sets, but selecting targets for further study is challenging. Here we propose a novel data-mining approach combining multiple heterogeneous data sets to prioritize genes for further study by using respiratory syncytial virus (RSV) infection as a model pathogen with a significant health care impact. The assumption was that the more frequently a gene is detected across multiple studies, the more important its role is. A literature search was performed to find data sets of genes and proteins that change after RSV infection. The data sets were standardized, collated into a single database, and then panned to determine which genes occurred in multiple data sets, generating a candidate gene list. This candidate gene list was validated by using both a clinical cohort and in vitro screening. We identified several genes that were frequently expressed following RSV infection with no assigned function in RSV control, including IFI27, IFIT3, IFI44L, GBP1, OAS3, IFI44, and IRF7. Drilling down into the function of these genes, we demonstrate a role in disease for the gene for interferon regulatory factor 7, which was highly ranked on the list, but not for IRF1, which was not. Thus, we have developed and validated an approach for collating published data sets into a manageable list of candidates, identifying novel targets for future analysis. IMPORTANCE Making the most of "big data" is one of the core challenges of current biology. There is a large array of heterogeneous data sets of host gene responses to infection, but these data sets do not inform us about gene function and require specialized skill sets and training for their utilization. Here we describe an approach that combines and simplifies these data sets, distilling this information into a single list of genes commonly upregulated in response to infection with RSV as a model pathogen. Many of the genes on the list have unknown functions in RSV disease. We validated the gene list with new clinical, in vitro, and in vivo data. This approach allows the rapid selection of genes of interest for further, more-detailed studies, thus reducing time and costs. Furthermore, the approach is simple to use and widely applicable to a range of diseases.
Collapse
Affiliation(s)
- Jacqueline U. McDonald
- Mucosal Infection and Immunity Group, Section of Virology, Imperial College London, St. Mary’s Campus, London, United Kingdom
| | - Myrsini Kaforou
- Section of Paediatrics, Imperial College London, St. Mary’s Campus, London, United Kingdom
| | - Simon Clare
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, United Kingdom
| | - Christine Hale
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, United Kingdom
| | - Maria Ivanova
- Mucosal Infection and Immunity Group, Section of Virology, Imperial College London, St. Mary’s Campus, London, United Kingdom
| | - Derek Huntley
- Imperial College Centre for Integrative Systems Biology and Bioinformatics, Imperial College London, London, United Kingdom
| | - Marcus Dorner
- Molecular Virology, Section of Virology, Imperial College London, St. Mary’s Campus, London, United Kingdom
| | - Victoria J. Wright
- Section of Paediatrics, Imperial College London, St. Mary’s Campus, London, United Kingdom
| | - Michael Levin
- Section of Paediatrics, Imperial College London, St. Mary’s Campus, London, United Kingdom
| | - Federico Martinon-Torres
- Department of Paediatrics, Hospital Clínico Universitario de Santiago, Santiago de Compostela, Spain
| | - Jethro A. Herberg
- Section of Paediatrics, Imperial College London, St. Mary’s Campus, London, United Kingdom
| | - John S. Tregoning
- Mucosal Infection and Immunity Group, Section of Virology, Imperial College London, St. Mary’s Campus, London, United Kingdom
| |
Collapse
|
70
|
Bartha I, Rausell A, McLaren PJ, Mohammadi P, Tardaguila M, Chaturvedi N, Fellay J, Telenti A. The Characteristics of Heterozygous Protein Truncating Variants in the Human Genome. PLoS Comput Biol 2015; 11:e1004647. [PMID: 26642228 PMCID: PMC4671652 DOI: 10.1371/journal.pcbi.1004647] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Accepted: 11/06/2015] [Indexed: 11/18/2022] Open
Abstract
Sequencing projects have identified large numbers of rare stop-gain and frameshift variants in the human genome. As most of these are observed in the heterozygous state, they test a gene’s tolerance to haploinsufficiency and dominant loss of function. We analyzed the distribution of truncating variants across 16,260 autosomal protein coding genes in 11,546 individuals. We observed 39,893 truncating variants affecting 12,062 genes, which significantly differed from an expectation of 12,916 genes under a model of neutral de novo mutation (p<10−4). Extrapolating this to increasing numbers of sequenced individuals, we estimate that 10.8% of human genes do not tolerate heterozygous truncating variants. An additional 10 to 15% of truncated genes may be rescued by incomplete penetrance or compensatory mutations, or because the truncating variants are of limited functional impact. The study of protein truncating variants delineates the essential genome and, more generally, identifies rare heterozygous variants as an unexplored source of diversity of phenotypic traits and diseases. Genome sequencing provides evidence for large numbers of putative protein truncating variants in humans. Most truncating variants are only observed in few individuals but are collectively prevalent and widely distributed across the coding genome. Most of the truncating variants are so rare that they are only observed in heterozygosis. The current study identifies 10% of genes where heterozygous truncations are not observed and describes their biological characteristics. In addition, for genes where rare truncations are observed, we argue that these are an unexplored source of diversity of phenotypic traits and diseases.
Collapse
Affiliation(s)
- István Bartha
- SIB Swiss Institute of Bioinformatics, Lausanne and Basel, Switzerland
- School of Life Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Antonio Rausell
- SIB Swiss Institute of Bioinformatics, Lausanne and Basel, Switzerland
- Vital-IT group, SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Paul J. McLaren
- SIB Swiss Institute of Bioinformatics, Lausanne and Basel, Switzerland
- School of Life Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Pejman Mohammadi
- SIB Swiss Institute of Bioinformatics, Lausanne and Basel, Switzerland
- Computational Biology Group, ETH Zurich, Zurich, Switzerland
| | - Manuel Tardaguila
- SIB Swiss Institute of Bioinformatics, Lausanne and Basel, Switzerland
- Vital-IT group, SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Nimisha Chaturvedi
- SIB Swiss Institute of Bioinformatics, Lausanne and Basel, Switzerland
- School of Life Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Jacques Fellay
- SIB Swiss Institute of Bioinformatics, Lausanne and Basel, Switzerland
- School of Life Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Amalio Telenti
- J. Craig Venter Institute, La Jolla, California, United States of America
- * E-mail:
| |
Collapse
|
71
|
Alazami AM, Awad SM, Coskun S, Al-Hassan S, Hijazi H, Abdulwahab FM, Poizat C, Alkuraya FS. TLE6 mutation causes the earliest known human embryonic lethality. Genome Biol 2015; 16:240. [PMID: 26537248 PMCID: PMC4634911 DOI: 10.1186/s13059-015-0792-0] [Citation(s) in RCA: 143] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Accepted: 09/28/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Embryonic lethality is a recognized phenotypic expression of individual gene mutations in model organisms. However, identifying embryonic lethal genes in humans is challenging, especially when the phenotype is manifested at the preimplantation stage. RESULTS In an ongoing effort to exploit the highly consanguineous nature of the Saudi population to catalog recessively acting embryonic lethal genes in humans, we have identified two families with a female-limited infertility phenotype. Using autozygosity mapping and whole exome sequencing, we map this phenotype to a single mutation in TLE6, a maternal effect gene that encodes a member of the subcortical maternal complex in mammalian oocytes. Consistent with the published phenotype of mouse Tle6 mutants, embryos from female patients who are homozygous for the TLE6 mutation fail to undergo early cleavage, with resulting sterility. The human mutation abrogates TLE6 phosphorylation, a step that is reported to be critical for the PKA-mediated progression of oocyte meiosis II. Furthermore, the TLE6 mutation impairs its binding to components of the subcortical maternal complex. CONCLUSION In this first report of a human defect in a member of the subcortical maternal subcritical maternal complex, we show that the TLE6 mutation is gender-specific and leads to the earliest known human embryonic lethality phenotype.
Collapse
Affiliation(s)
- Anas M Alazami
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Salma M Awad
- Cardiovascular Research Program, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Serdar Coskun
- Department of Pathology and Laboratory Medicine, King Faisal Specialist Hospital and Research Center, Alfaisal University, Riyadh, Saudi Arabia
| | - Saad Al-Hassan
- Department of Obstetrics and Gynecology, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Hadia Hijazi
- Cardiovascular Research Program, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Firdous M Abdulwahab
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Coralie Poizat
- Cardiovascular Research Program, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia.
| | - Fowzan S Alkuraya
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia. .,Department of Anatomy and Cell Biology, College of Medicine, Alfaisal University, Riyadh, Saudi Arabia.
| |
Collapse
|
72
|
Wilson R, McGuire C, Mohun T. Deciphering the mechanisms of developmental disorders: phenotype analysis of embryos from mutant mouse lines. Nucleic Acids Res 2015; 44:D855-61. [PMID: 26519470 PMCID: PMC4702824 DOI: 10.1093/nar/gkv1138] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Accepted: 10/18/2015] [Indexed: 01/09/2023] Open
Abstract
The Deciphering the Mechanisms of Developmental Disorders (DMDD) consortium is a research programme set up to identify genes in the mouse, which if mutated (or knocked-out) result in embryonic lethality when homozygous, and initiate the study of why disruption of their function has such profound effects on embryo development and survival. The project uses a combination of comprehensive high resolution 3D imaging and tissue histology to identify abnormalities in embryo and placental structures of embryonic lethal lines. The image data we have collected and the phenotypes scored are freely available through the project website (http://dmdd.org.uk). In this article we describe the web interface to the images that allows the embryo data to be viewed at full resolution in different planes, discuss how to search the database for a phenotype, and our approach to organising the data for an embryo and a mutant line so it is easy to comprehend and intuitive to navigate.
Collapse
Affiliation(s)
- Robert Wilson
- The Francis Crick Institute Mill Hill Laboratory, The Ridgeway, Mill Hill, London NW7 1AA, UK
| | - Christina McGuire
- The Francis Crick Institute Mill Hill Laboratory, The Ridgeway, Mill Hill, London NW7 1AA, UK
| | - Timothy Mohun
- The Francis Crick Institute Mill Hill Laboratory, The Ridgeway, Mill Hill, London NW7 1AA, UK
| | | |
Collapse
|
73
|
Reporter Gene Silencing in Targeted Mouse Mutants Is Associated with Promoter CpG Island Methylation. PLoS One 2015; 10:e0134155. [PMID: 26275310 PMCID: PMC4537176 DOI: 10.1371/journal.pone.0134155] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Accepted: 07/06/2015] [Indexed: 11/19/2022] Open
Abstract
Targeted mutations in mouse disrupt local chromatin structure and may lead to unanticipated local effects. We evaluated targeted gene promoter silencing in a group of six mutants carrying the tm1a Knockout Mouse Project allele containing both a LacZ reporter gene driven by the native promoter and a neo selection cassette. Messenger RNA levels of the reporter gene and targeted gene were assessed by qRT-PCR, and methylation of the promoter CpG islands and LacZ coding sequence were evaluated by sequencing of bisulfite-treated DNA. Mutants were stratified by LacZ staining into presumed Silenced and Expressed reporter genes. Silenced mutants had reduced relative quantities LacZ mRNA and greater CpG Island methylation compared with the Expressed mutant group. Within the silenced group, LacZ coding sequence methylation was significantly and positively correlated with CpG Island methylation, while promoter CpG methylation was only weakly correlated with LacZ gene mRNA. The results support the conclusion that there is promoter silencing in a subset of mutants carrying the tm1a allele. The features of targeted genes which promote local silencing when targeted remain unknown.
Collapse
|
74
|
Chong J, Buckingham K, Jhangiani S, Boehm C, Sobreira N, Smith J, Harrell T, McMillin M, Wiszniewski W, Gambin T, Coban Akdemir Z, Doheny K, Scott A, Avramopoulos D, Chakravarti A, Hoover-Fong J, Mathews D, Witmer P, Ling H, Hetrick K, Watkins L, Patterson K, Reinier F, Blue E, Muzny D, Kircher M, Bilguvar K, López-Giráldez F, Sutton V, Tabor H, Leal S, Gunel M, Mane S, Gibbs R, Boerwinkle E, Hamosh A, Shendure J, Lupski J, Lifton R, Valle D, Nickerson D, Bamshad M, Bamshad MJ. The Genetic Basis of Mendelian Phenotypes: Discoveries, Challenges, and Opportunities. Am J Hum Genet 2015; 97:199-215. [PMID: 26166479 DOI: 10.1016/j.ajhg.2015.06.009] [Citation(s) in RCA: 459] [Impact Index Per Article: 45.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Indexed: 01/06/2023] Open
Abstract
Discovering the genetic basis of a Mendelian phenotype establishes a causal link between genotype and phenotype, making possible carrier and population screening and direct diagnosis. Such discoveries also contribute to our knowledge of gene function, gene regulation, development, and biological mechanisms that can be used for developing new therapeutics. As of February 2015, 2,937 genes underlying 4,163 Mendelian phenotypes have been discovered, but the genes underlying ∼50% (i.e., 3,152) of all known Mendelian phenotypes are still unknown, and many more Mendelian conditions have yet to be recognized. This is a formidable gap in biomedical knowledge. Accordingly, in December 2011, the NIH established the Centers for Mendelian Genomics (CMGs) to provide the collaborative framework and infrastructure necessary for undertaking large-scale whole-exome sequencing and discovery of the genetic variants responsible for Mendelian phenotypes. In partnership with 529 investigators from 261 institutions in 36 countries, the CMGs assessed 18,863 samples from 8,838 families representing 579 known and 470 novel Mendelian phenotypes as of January 2015. This collaborative effort has identified 956 genes, including 375 not previously associated with human health, that underlie a Mendelian phenotype. These results provide insight into study design and analytical strategies, identify novel mechanisms of disease, and reveal the extensive clinical variability of Mendelian phenotypes. Discovering the gene underlying every Mendelian phenotype will require tackling challenges such as worldwide ascertainment and phenotypic characterization of families affected by Mendelian conditions, improvement in sequencing and analytical techniques, and pervasive sharing of phenotypic and genomic data among researchers, clinicians, and families.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Michael J Bamshad
- Department of Pediatrics, University of Washington, Seattle, WA 98195, USA; Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA; Division of Genetic Medicine, Seattle Children's Hospital, Seattle, WA 98105, USA.
| |
Collapse
|
75
|
Palmer K, Fairfield H, Borgeia S, Curtain M, Hassan MG, Dionne L, Yong Karst S, Coombs H, Bronson RT, Reinholdt LG, Bergstrom DE, Donahue LR, Cox TC, Murray SA. Discovery and characterization of spontaneous mouse models of craniofacial dysmorphology. Dev Biol 2015; 415:216-227. [PMID: 26234751 DOI: 10.1016/j.ydbio.2015.07.023] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 07/29/2015] [Indexed: 11/30/2022]
Abstract
Craniofacial abnormalities are among the most common features of human genetic syndromes and disorders. The etiology of these conditions is often complex, influenced by both genetic context and the environment. Frequently, craniofacial abnormalities present as part of a syndrome with clear comorbid phenotypes, providing additional insight into mechanisms of the causative gene or pathway. The mouse has been a key tool in our understanding of the genetic mechanisms of craniofacial development and disease, and can provide excellent models for human craniofacial abnormalities. While powerful genetic engineering tools in the mouse have contributed significantly our understanding of craniofacial development and dysmorphology, forward genetic approaches provide an unbiased means to identify new genes and pathways. Moreover, spontaneous mutations can occur on any number of genetic backgrounds, potentially revealing critical genes that require a specific genetic context. Here we report discovery and phenotyping of 43 craniofacial mouse models, derived primarily from a screen for spontaneous mutations in production colonies at the Jackson Laboratory. We identify the causative gene for 33 lines, including novel genes in pathways not previously connected to craniofacial development, and novel alleles of known genes that present with unique phenotypes. Together with our detailed characterization, this work provides a valuable gene discovery resource for the craniofacial community, and a rich source of mouse models for further investigation.
Collapse
Affiliation(s)
- Kristina Palmer
- The Jackson Laboratory, 600 Main St., Bar Harbor, ME 04609, USA
| | | | - Suhaib Borgeia
- Seattle Children's Research Institute, Seattle, WA 98101, USA
| | | | - Mohamed G Hassan
- Seattle Children's Research Institute, Seattle, WA 98101, USA; Faculty of Oral and Dental Medicine, South Valley University, Qena, Egypt
| | - Louise Dionne
- The Jackson Laboratory, 600 Main St., Bar Harbor, ME 04609, USA
| | - Son Yong Karst
- The Jackson Laboratory, 600 Main St., Bar Harbor, ME 04609, USA
| | - Harold Coombs
- The Jackson Laboratory, 600 Main St., Bar Harbor, ME 04609, USA
| | | | | | | | | | - Timothy C Cox
- Seattle Children's Research Institute, Seattle, WA 98101, USA; University of Washington, Department of Pediatrics (Craniofacial Medicine), Seattle, WA 98195, USA
| | - Stephen A Murray
- The Jackson Laboratory, 600 Main St., Bar Harbor, ME 04609, USA.
| |
Collapse
|
76
|
Eppig JT, Richardson JE, Kadin JA, Smith CL, Blake JA, Bult CJ. Mouse Genome Database: From sequence to phenotypes and disease models. Genesis 2015; 53:458-73. [PMID: 26150326 PMCID: PMC4545690 DOI: 10.1002/dvg.22874] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Revised: 06/30/2015] [Accepted: 07/02/2015] [Indexed: 12/19/2022]
Abstract
The Mouse Genome Database (MGD, www.informatics.jax.org) is the international scientific database for genetic, genomic, and biological data on the laboratory mouse to support the research requirements of the biomedical community. To accomplish this goal, MGD provides broad data coverage, serves as the authoritative standard for mouse nomenclature for genes, mutants, and strains, and curates and integrates many types of data from literature and electronic sources. Among the key data sets MGD supports are: the complete catalog of mouse genes and genome features, comparative homology data for mouse and vertebrate genes, the authoritative set of Gene Ontology (GO) annotations for mouse gene functions, a comprehensive catalog of mouse mutations and their phenotypes, and a curated compendium of mouse models of human diseases. Here, we describe the data acquisition process, specifics about MGD's key data areas, methods to access and query MGD data, and outreach and user help facilities. genesis 53:458–473, 2015. © 2015 The Authors. Genesis Published by Wiley Periodicals, Inc.
Collapse
|
77
|
Scavizzi F, Ryder E, Newman S, Raspa M, Gleeson D, Wardle-Jones H, Montoliu L, Fernandez A, Dessain ML, Larrigaldie V, Khorshidi Z, Vuolteenaho R, Soininen R, André P, Jacquot S, Hong Y, de Angelis MH, Ramirez-Solis R, Doe B. Blastocyst genotyping for quality control of mouse mutant archives: an ethical and economical approach. Transgenic Res 2015; 24:921-7. [PMID: 26178246 PMCID: PMC4569667 DOI: 10.1007/s11248-015-9897-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Accepted: 07/09/2015] [Indexed: 01/22/2023]
Abstract
With the advent of modern developmental biology and molecular genetics, the scientific community has generated thousands of newly genetically altered strains of laboratory mice with the aim of elucidating gene function. To this end, a large group of Institutions which form the International Mouse Phenotyping Consortium is generating and phenotyping a knockout mouse strain for each of the ~20,000 protein-coding genes using the mutant ES cell resource produced by the International Knockout Mouse Consortium. These strains are made available to the research community via public repositories, mostly as cryopreserved sperm or embryos. To ensure the quality of this frozen resource there is a requirement that for each strain the frozen sperm/embryos are proven able to produce viable mutant progeny, before the live animal resource is removed from cages. Given the current requirement to generate live pups to demonstrate their mutant genotype, this quality control check necessitates the use and generation of many animals and requires considerable time, cage space, technical and economic resources. Here, we describe a simple and efficient method of genotyping pre-implantation stage blastocysts with significant ethical and economic advantages especially beneficial for current and future large-scale mouse mutagenesis projects.
Collapse
Affiliation(s)
- Ferdinando Scavizzi
- Consiglio Nazionale delle Ricerche (IBCN), CNR-Campus International Development (EMMA-INFRAFRONTIER- IMPC), A. Buzzati-Traverso Campus, Via E. Ramarini 32, 00015, Monterotondo Scalo, Roma, Italy
| | - Edward Ryder
- Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, CB10 1SA, UK
| | - Stuart Newman
- Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, CB10 1SA, UK
| | - Marcello Raspa
- Consiglio Nazionale delle Ricerche (IBCN), CNR-Campus International Development (EMMA-INFRAFRONTIER- IMPC), A. Buzzati-Traverso Campus, Via E. Ramarini 32, 00015, Monterotondo Scalo, Roma, Italy
| | - Diane Gleeson
- Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, CB10 1SA, UK
| | | | - Lluis Montoliu
- Department of Molecular and Cellular Biology, National Centre for Biotechnology (CNB-CSIC), Campus de Cantoblanco, Darwin 3, 28049, Madrid, Spain
- CIBERER, ISCIII, Madrid, Spain
| | - Almudena Fernandez
- Department of Molecular and Cellular Biology, National Centre for Biotechnology (CNB-CSIC), Campus de Cantoblanco, Darwin 3, 28049, Madrid, Spain
- CIBERER, ISCIII, Madrid, Spain
| | - Marie-Laure Dessain
- CNRS, TAAM-CDTA UPS44, 3B rue de la Férollerie, CS 20057 45071, Orléans Cedex 2, France
| | - Vanessa Larrigaldie
- CNRS, TAAM-CDTA UPS44, 3B rue de la Férollerie, CS 20057 45071, Orléans Cedex 2, France
| | - Zuzana Khorshidi
- Karolinska Center for Transgene Technologies, Comparative Medicine, Karolinska Institutet, von Eulers väg 4a, 171 77, Stockholm, Sweden
| | | | - Raija Soininen
- Biocenter Oulu, University of Oulu, Aapistie 5 A, 90220, Oulu, Finland
| | - Philippe André
- ICS France Institut Clinique de la Souris, PHENOMIN, ICS-MCI, CNRS, INSERM, Université de Strasbourg, 1 rue Laurent Fries, 67404, Illkirch, France
| | - Sylvie Jacquot
- ICS France Institut Clinique de la Souris, PHENOMIN, ICS-MCI, CNRS, INSERM, Université de Strasbourg, 1 rue Laurent Fries, 67404, Illkirch, France
| | - Yi Hong
- Institute of Experimental Genetics, Helmholtz Zentrum München-German Research Center for Environmental Health (GmbH), Neuherberg, Germany
| | - Martin Hrabe de Angelis
- Institute of Experimental Genetics, Helmholtz Zentrum München-German Research Center for Environmental Health (GmbH), Neuherberg, Germany
| | | | - Brendan Doe
- Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, CB10 1SA, UK.
| |
Collapse
|
78
|
PhenStat: A Tool Kit for Standardized Analysis of High Throughput Phenotypic Data. PLoS One 2015; 10:e0131274. [PMID: 26147094 PMCID: PMC4493137 DOI: 10.1371/journal.pone.0131274] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 05/06/2015] [Indexed: 12/31/2022] Open
Abstract
The lack of reproducibility with animal phenotyping experiments is a growing concern among the biomedical community. One contributing factor is the inadequate description of statistical analysis methods that prevents researchers from replicating results even when the original data are provided. Here we present PhenStat – a freely available R package that provides a variety of statistical methods for the identification of phenotypic associations. The methods have been developed for high throughput phenotyping pipelines implemented across various experimental designs with an emphasis on managing temporal variation. PhenStat is targeted to two user groups: small-scale users who wish to interact and test data from large resources and large-scale users who require an automated statistical analysis pipeline. The software provides guidance to the user for selecting appropriate analysis methods based on the dataset and is designed to allow for additions and modifications as needed. The package was tested on mouse and rat data and is used by the International Mouse Phenotyping Consortium (IMPC). By providing raw data and the version of PhenStat used, resources like the IMPC give users the ability to replicate and explore results within their own computing environment.
Collapse
|
79
|
Shamseldin HE, Tulbah M, Kurdi W, Nemer M, Alsahan N, Al Mardawi E, Khalifa O, Hashem A, Kurdi A, Babay Z, Bubshait DK, Ibrahim N, Abdulwahab F, Rahbeeni Z, Hashem M, Alkuraya FS. Identification of embryonic lethal genes in humans by autozygosity mapping and exome sequencing in consanguineous families. Genome Biol 2015; 16:116. [PMID: 26036949 PMCID: PMC4491988 DOI: 10.1186/s13059-015-0681-6] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Accepted: 05/21/2015] [Indexed: 01/23/2023] Open
Abstract
Background Identifying genetic variants that lead to discernible phenotypes is the core of Mendelian genetics. An approach that considers embryonic lethality as a bona fide Mendelian phenotype has the potential to reveal novel genetic causes, which will further our understanding of early human development at a molecular level. Consanguineous families in which embryonic lethality segregates as a recessive Mendelian phenotype offer a unique opportunity for high throughput novel gene discovery as has been established for other recessive postnatal phenotypes. Results We have studied 24 eligible families using autozygosity mapping and whole-exome sequencing. In addition to revealing mutations in genes previously linked to embryonic lethality in severe cases, our approach revealed seven novel candidate genes (THSD1, PIGC, UBN1, MYOM1, DNAH14, GALNT14, and FZD6). A founder mutation in one of these genes, THSD1, which has been linked to vascular permeability, accounted for embryonic lethality in three of the study families. Unlike the other six candidate genes, we were able to identify a second mutation in THSD1 in a family with a less severe phenotype consisting of hydrops fetalis and persistent postnatal edema, which provides further support for the proposed link between this gene and embryonic lethality. Conclusions Our study represents an important step towards the systematic analysis of “embryonic lethal genes” in humans. Electronic supplementary material The online version of this article (doi:10.1186/s13059-015-0681-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Hanan E Shamseldin
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia.
| | - Maha Tulbah
- Department of Obstetrics and Gynecology, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia.
| | - Wesam Kurdi
- Department of Obstetrics and Gynecology, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia.
| | - Maha Nemer
- Department of Obstetrics and Gynecology, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia.
| | - Nada Alsahan
- Department of Obstetrics and Gynecology, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia.
| | - Elham Al Mardawi
- Department of Obstetrics and Gynecology, Security Forces Hospital, Riyadh, Saudi Arabia.
| | - Ola Khalifa
- Department of Medical Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia. .,Department of Pediatrics, Faculty of Medicine, Ain Shams University, Cairo, Egypt.
| | - Amal Hashem
- Department of Pediatrics, Prince Sultan Military Medical City, Riyadh, Saudi Arabia.
| | - Ahmed Kurdi
- Department of Obstetrics and Gynecology, Prince Sultan Military Medical City, Riyadh, Saudi Arabia.
| | - Zainab Babay
- Department of Obstetrics and Gynecology, College of Medicine, King Saud University, Riyadh, Saudi Arabia.
| | - Dalal K Bubshait
- Department of Medical Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia. .,Department of Pediatrics, King Fahd Hospital of the University, University of Dammam, Dammam, Saudi Arabia.
| | - Niema Ibrahim
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia.
| | - Firdous Abdulwahab
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia.
| | - Zuhair Rahbeeni
- Department of Medical Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia.
| | - Mais Hashem
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia.
| | - Fowzan S Alkuraya
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia. .,Department of Anatomy and Cell Biology, College of Medicine, Alfaisal University, Riyadh, Saudi Arabia.
| |
Collapse
|
80
|
Proetzel G, Wiles MV, Roopenian DC. Genetically engineered humanized mouse models for preclinical antibody studies. BioDrugs 2015; 28:171-80. [PMID: 24150980 DOI: 10.1007/s40259-013-0071-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The use of genetic engineering has vastly improved our capabilities to create animal models relevant in preclinical research. With the recent advances in gene-editing technologies, it is now possible to very rapidly create highly tunable mouse models as needs arise. Here, we provide an overview of genetic engineering methods, as well as the development of humanized neonatal Fc receptor (FcRn) models and their use for monoclonal antibody in vivo studies.
Collapse
|
81
|
Probert F, Rice P, Scudamore CL, Wells S, Williams R, Hough TA, Cox IJ. 1H NMR Metabolic Profiling of Plasma Reveals Additional Phenotypes in Knockout Mouse Models. J Proteome Res 2015; 14:2036-45. [DOI: 10.1021/pr501039k] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Fay Probert
- Mary
Lyon Centre, MRC Harwell, Oxfordshire OX11 0RD, United Kingdom
- Institute of Hepatology, Foundation for Liver Research, 69-75 Chenies Mews, London WC1E 6HX, United Kingdom
| | - Paul Rice
- Mary
Lyon Centre, MRC Harwell, Oxfordshire OX11 0RD, United Kingdom
| | | | - Sara Wells
- Mary
Lyon Centre, MRC Harwell, Oxfordshire OX11 0RD, United Kingdom
| | - Roger Williams
- Institute of Hepatology, Foundation for Liver Research, 69-75 Chenies Mews, London WC1E 6HX, United Kingdom
| | - Tertius A. Hough
- Mary
Lyon Centre, MRC Harwell, Oxfordshire OX11 0RD, United Kingdom
| | - I. Jane Cox
- Institute of Hepatology, Foundation for Liver Research, 69-75 Chenies Mews, London WC1E 6HX, United Kingdom
| |
Collapse
|
82
|
Fox CS, Hall JL, Arnett DK, Ashley EA, Delles C, Engler MB, Freeman MW, Johnson JA, Lanfear DE, Liggett SB, Lusis AJ, Loscalzo J, MacRae CA, Musunuru K, Newby LK, O'Donnell CJ, Rich SS, Terzic A. Future translational applications from the contemporary genomics era: a scientific statement from the American Heart Association. Circulation 2015; 131:1715-36. [PMID: 25882488 DOI: 10.1161/cir.0000000000000211] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The field of genetics and genomics has advanced considerably with the achievement of recent milestones encompassing the identification of many loci for cardiovascular disease and variable drug responses. Despite this achievement, a gap exists in the understanding and advancement to meaningful translation that directly affects disease prevention and clinical care. The purpose of this scientific statement is to address the gap between genetic discoveries and their practical application to cardiovascular clinical care. In brief, this scientific statement assesses the current timeline for effective translation of basic discoveries to clinical advances, highlighting past successes. Current discoveries in the area of genetics and genomics are covered next, followed by future expectations, tools, and competencies for achieving the goal of improving clinical care.
Collapse
|
83
|
Smith CL, Eppig JT. Expanding the mammalian phenotype ontology to support automated exchange of high throughput mouse phenotyping data generated by large-scale mouse knockout screens. J Biomed Semantics 2015; 6:11. [PMID: 25825651 PMCID: PMC4378007 DOI: 10.1186/s13326-015-0009-1] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Accepted: 03/03/2015] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND A vast array of data is about to emerge from the large scale high-throughput mouse knockout phenotyping projects worldwide. It is critical that this information is captured in a standardized manner, made accessible, and is fully integrated with other phenotype data sets for comprehensive querying and analysis across all phenotype data types. The volume of data generated by the high-throughput phenotyping screens is expected to grow exponentially, thus, automated methods and standards to exchange phenotype data are required. RESULTS The IMPC (International Mouse Phenotyping Consortium) is using the Mammalian Phenotype (MP) ontology in the automated annotation of phenodeviant data from high throughput phenotyping screens. 287 new term additions with additional hierarchy revisions were made in multiple branches of the MP ontology to accurately describe the results generated by these high throughput screens. CONCLUSIONS Because these large scale phenotyping data sets will be reported using the MP as the common data standard for annotation and data exchange, automated importation of these data to MGI (Mouse Genome Informatics) and other resources is possible without curatorial effort. Maximum biomedical value of these mutant mice will come from integrating primary high-throughput phenotyping data with secondary, comprehensive phenotypic analyses combined with published phenotype details on these and related mutants at MGI and other resources.
Collapse
Affiliation(s)
- Cynthia L Smith
- Mouse Genome Informatics, The Jackson Laboratory, Bar Harbor, ME 04609 USA
| | - Janan T Eppig
- Mouse Genome Informatics, The Jackson Laboratory, Bar Harbor, ME 04609 USA
| |
Collapse
|
84
|
Identification of a large set of rare complete human knockouts. Nat Genet 2015; 47:448-52. [PMID: 25807282 DOI: 10.1038/ng.3243] [Citation(s) in RCA: 167] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Accepted: 02/13/2015] [Indexed: 12/17/2022]
Abstract
Loss-of-function mutations cause many mendelian diseases. Here we aimed to create a catalog of autosomal genes that are completely knocked out in humans by rare loss-of-function mutations. We sequenced the whole genomes of 2,636 Icelanders and imputed the sequence variants identified in this set into 101,584 additional chip-genotyped and phased Icelanders. We found a total of 6,795 autosomal loss-of-function SNPs and indels in 4,924 genes. Of the genotyped Icelanders, 7.7% are homozygotes or compound heterozygotes for loss-of-function mutations with a minor allele frequency (MAF) below 2% in 1,171 genes (complete knockouts). Genes that are highly expressed in the brain are less often completely knocked out than other genes. Homozygous loss-of-function offspring of two heterozygous parents occurred less frequently than expected (deficit of 136 per 10,000 transmissions for variants with MAF <2%, 95% confidence interval (CI) = 10-261).
Collapse
|
85
|
|
86
|
Affiliation(s)
- Karl F Hoffmann
- Institute of Biological, Environmental and Rural Sciences (IBERS), Aberystwyth University, Aberystwyth SY23 3DA, UK.
| | - Paul J Brindley
- Department of Microbiology, Immunology and Tropical Medicine and Research Center for Neglected Diseases of Poverty, School of Medicine and Health Sciences, George Washington University, Washington, DC 20037, USA
| | - Matthew Berriman
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SA, UK
| |
Collapse
|
87
|
Brommage R, Liu J, Hansen GM, Kirkpatrick LL, Potter DG, Sands AT, Zambrowicz B, Powell DR, Vogel P. High-throughput screening of mouse gene knockouts identifies established and novel skeletal phenotypes. Bone Res 2014; 2:14034. [PMID: 26273529 PMCID: PMC4472125 DOI: 10.1038/boneres.2014.34] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Revised: 07/29/2014] [Accepted: 07/31/2014] [Indexed: 12/13/2022] Open
Abstract
Screening gene function in vivo is a powerful approach to discover novel drug targets. We present high-throughput screening (HTS) data for 3 762 distinct global gene knockout (KO) mouse lines with viable adult homozygous mice generated using either gene-trap or homologous recombination technologies. Bone mass was determined from DEXA scans of male and female mice at 14 weeks of age and by microCT analyses of bones from male mice at 16 weeks of age. Wild-type (WT) cagemates/littermates were examined for each gene KO. Lethality was observed in an additional 850 KO lines. Since primary HTS are susceptible to false positive findings, additional cohorts of mice from KO lines with intriguing HTS bone data were examined. Aging, ovariectomy, histomorphometry and bone strength studies were performed and possible non-skeletal phenotypes were explored. Together, these screens identified multiple genes affecting bone mass: 23 previously reported genes (Calcr, Cebpb, Crtap, Dcstamp, Dkk1, Duoxa2, Enpp1, Fgf23, Kiss1/Kiss1r, Kl (Klotho), Lrp5, Mstn, Neo1, Npr2, Ostm1, Postn, Sfrp4, Slc30a5, Slc39a13, Sost, Sumf1, Src, Wnt10b), five novel genes extensively characterized (Cldn18, Fam20c, Lrrk1, Sgpl1, Wnt16), five novel genes with preliminary characterization (Agpat2, Rassf5, Slc10a7, Slc26a7, Slc30a10) and three novel undisclosed genes coding for potential osteoporosis drug targets.
Collapse
Affiliation(s)
| | - Jeff Liu
- Lexicon Pharmaceuticals , The Woodlands, TX, USA
| | | | | | | | | | | | | | - Peter Vogel
- Lexicon Pharmaceuticals , The Woodlands, TX, USA
| |
Collapse
|
88
|
Mouse ENU Mutagenesis to Understand Immunity to Infection: Methods, Selected Examples, and Perspectives. Genes (Basel) 2014; 5:887-925. [PMID: 25268389 PMCID: PMC4276919 DOI: 10.3390/genes5040887] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Revised: 08/19/2014] [Accepted: 08/21/2014] [Indexed: 12/30/2022] Open
Abstract
Infectious diseases are responsible for over 25% of deaths globally, but many more individuals are exposed to deadly pathogens. The outcome of infection results from a set of diverse factors including pathogen virulence factors, the environment, and the genetic make-up of the host. The completion of the human reference genome sequence in 2004 along with technological advances have tremendously accelerated and renovated the tools to study the genetic etiology of infectious diseases in humans and its best characterized mammalian model, the mouse. Advancements in mouse genomic resources have accelerated genome-wide functional approaches, such as gene-driven and phenotype-driven mutagenesis, bringing to the fore the use of mouse models that reproduce accurately many aspects of the pathogenesis of human infectious diseases. Treatment with the mutagen N-ethyl-N-nitrosourea (ENU) has become the most popular phenotype-driven approach. Our team and others have employed mouse ENU mutagenesis to identify host genes that directly impact susceptibility to pathogens of global significance. In this review, we first describe the strategies and tools used in mouse genetics to understand immunity to infection with special emphasis on chemical mutagenesis of the mouse germ-line together with current strategies to efficiently identify functional mutations using next generation sequencing. Then, we highlight illustrative examples of genes, proteins, and cellular signatures that have been revealed by ENU screens and have been shown to be involved in susceptibility or resistance to infectious diseases caused by parasites, bacteria, and viruses.
Collapse
|
89
|
Karunamuni GH, Gu S, Ford MR, Peterson LM, Ma P, Wang YT, Rollins AM, Jenkins MW, Watanabe M. Capturing structure and function in an embryonic heart with biophotonic tools. Front Physiol 2014; 5:351. [PMID: 25309451 PMCID: PMC4173643 DOI: 10.3389/fphys.2014.00351] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Accepted: 08/27/2014] [Indexed: 11/17/2022] Open
Abstract
Disturbed cardiac function at an early stage of development has been shown to correlate with cellular/molecular, structural as well as functional cardiac anomalies at later stages culminating in the congenital heart defects (CHDs) that present at birth. While our knowledge of cellular and molecular steps in cardiac development is growing rapidly, our understanding of the role of cardiovascular function in the embryo is still in an early phase. One reason for the scanty information in this area is that the tools to study early cardiac function are limited. Recently developed and adapted biophotonic tools may overcome some of the challenges of studying the tiny fragile beating heart. In this chapter, we describe and discuss our experience in developing and implementing biophotonic tools to study the role of function in heart development with emphasis on optical coherence tomography (OCT). OCT can be used for detailed structural and functional studies of the tubular and looping embryo heart under physiological conditions. The same heart can be rapidly and quantitatively phenotyped at early and again at later stages using OCT. When combined with other tools such as optical mapping (OM) and optical pacing (OP), OCT has the potential to reveal in spatial and temporal detail the biophysical changes that can impact mechanotransduction pathways. This information may provide better explanations for the etiology of the CHDs when interwoven with our understanding of morphogenesis and the molecular pathways that have been described to be involved. Future directions for advances in the creation and use of biophotonic tools are discussed.
Collapse
Affiliation(s)
- Ganga H Karunamuni
- Department of Pediatrics, Case Western Reserve University School of Medicine Cleveland, OH, USA
| | - Shi Gu
- Department of Biomedical Engineering, Case Western Reserve University School of Engineering Cleveland, OH, USA
| | - Matthew R Ford
- Department of Biomedical Engineering, Case Western Reserve University School of Engineering Cleveland, OH, USA
| | - Lindsy M Peterson
- Department of Biomedical Engineering, Case Western Reserve University School of Engineering Cleveland, OH, USA
| | - Pei Ma
- Department of Biomedical Engineering, Case Western Reserve University School of Engineering Cleveland, OH, USA
| | - Yves T Wang
- Department of Pediatrics, Case Western Reserve University School of Medicine Cleveland, OH, USA ; Department of Biomedical Engineering, Case Western Reserve University School of Engineering Cleveland, OH, USA
| | - Andrew M Rollins
- Department of Biomedical Engineering, Case Western Reserve University School of Engineering Cleveland, OH, USA
| | - Michael W Jenkins
- Department of Pediatrics, Case Western Reserve University School of Medicine Cleveland, OH, USA ; Department of Biomedical Engineering, Case Western Reserve University School of Engineering Cleveland, OH, USA
| | - Michiko Watanabe
- Department of Pediatrics, Case Western Reserve University School of Medicine Cleveland, OH, USA
| |
Collapse
|
90
|
Belizário JE. The humankind genome: from genetic diversity to the origin of human diseases. Genome 2014; 56:705-16. [PMID: 24433206 DOI: 10.1139/gen-2013-0125] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Genome-wide association studies have failed to establish common variant risk for the majority of common human diseases. The underlying reasons for this failure are explained by recent studies of resequencing and comparison of over 1200 human genomes and 10 000 exomes, together with the delineation of DNA methylation patterns (epigenome) and full characterization of coding and noncoding RNAs (transcriptome) being transcribed. These studies have provided the most comprehensive catalogues of functional elements and genetic variants that are now available for global integrative analysis and experimental validation in prospective cohort studies. With these datasets, researchers will have unparalleled opportunities for the alignment, mining, and testing of hypotheses for the roles of specific genetic variants, including copy number variations, single nucleotide polymorphisms, and indels as the cause of specific phenotypes and diseases. Through the use of next-generation sequencing technologies for genotyping and standardized ontological annotation to systematically analyze the effects of genomic variation on humans and model organism phenotypes, we will be able to find candidate genes and new clues for disease's etiology and treatment. This article describes essential concepts in genetics and genomic technologies as well as the emerging computational framework to comprehensively search websites and platforms available for the analysis and interpretation of genomic data.
Collapse
Affiliation(s)
- Jose E Belizário
- Departamento de Farmacologia, Instituto de Ciências Biomédicas da Universidade de São Paulo, Avenida Lineu Prestes, 1524 CEP 05508-900, São Paulo, SP, Brazil
| |
Collapse
|
91
|
Bartman CM, Egelston J, Ren X, Das R, Phiel CJ. A simple and efficient method for transfecting mouse embryonic stem cells using polyethylenimine. Exp Cell Res 2014; 330:178-85. [PMID: 25102378 DOI: 10.1016/j.yexcr.2014.07.020] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Revised: 07/12/2014] [Accepted: 07/15/2014] [Indexed: 12/24/2022]
Abstract
Mouse embryonic stem cells (ESCs) can be transfected by electroporation, liposomal reagents, and viral transduction methods. The cationic polymer polyethylenimine (PEI) has been shown to transfect a variety of differentiated mammalian cell types, including mouse ESCs, but existing methods require the use of additional equipment that is not readily accessible to most labs. Here we describe conditions that permit for the efficient transfection of mouse ESCs with low cytotoxicity and without the need for specialized equipment. Our goal was to devise a protocol for the PEI-mediated transfection of mouse ESCs that was comparable in ease to commercial transfection reagents. For these studies, we compared PEI transfection efficiency and cytotoxicity to a well-known liposomal transfection reagent, Lipofectamine2000(™) (LF2K), using fluorescence microscopy, flow cytometry, cell viability assays, and Western blotting. We provide evidence that PEI transfection of mouse ESCs compares favorably to LF2K. Our optimized protocol for efficient transfection of mouse ESCs with PEI is detailed in this report.
Collapse
Affiliation(s)
- Colleen M Bartman
- Department of Integrative Biology, University of Colorado Denver, S4111, 1201 Fifth Street, Denver, CO 80204, United States
| | - Jennifer Egelston
- Department of Integrative Biology, University of Colorado Denver, S4111, 1201 Fifth Street, Denver, CO 80204, United States
| | - Xiaojun Ren
- Department of Chemistry, University of Colorado Denver, 1201 Fifth Street, Denver, CO 80204, United States
| | - Raibatak Das
- Department of Integrative Biology, University of Colorado Denver, S4111, 1201 Fifth Street, Denver, CO 80204, United States
| | - Christopher J Phiel
- Department of Integrative Biology, University of Colorado Denver, S4111, 1201 Fifth Street, Denver, CO 80204, United States.
| |
Collapse
|
92
|
Gaytán BD, Vulpe CD. Functional toxicology: tools to advance the future of toxicity testing. Front Genet 2014; 5:110. [PMID: 24847352 PMCID: PMC4017141 DOI: 10.3389/fgene.2014.00110] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2014] [Accepted: 04/12/2014] [Indexed: 11/16/2022] Open
Abstract
The increased presence of chemical contaminants in the environment is an undeniable concern to human health and ecosystems. Historically, by relying heavily upon costly and laborious animal-based toxicity assays, the field of toxicology has often neglected examinations of the cellular and molecular mechanisms of toxicity for the majority of compounds—information that, if available, would strengthen risk assessment analyses. Functional toxicology, where cells or organisms with gene deletions or depleted proteins are used to assess genetic requirements for chemical tolerance, can advance the field of toxicity testing by contributing data regarding chemical mechanisms of toxicity. Functional toxicology can be accomplished using available genetic tools in yeasts, other fungi and bacteria, and eukaryotes of increased complexity, including zebrafish, fruit flies, rodents, and human cell lines. Underscored is the value of using less complex systems such as yeasts to direct further studies in more complex systems such as human cell lines. Functional techniques can yield (1) novel insights into chemical toxicity; (2) pathways and mechanisms deserving of further study; and (3) candidate human toxicant susceptibility or resistance genes.
Collapse
Affiliation(s)
- Brandon D Gaytán
- Department of Nutritional Science and Toxicology, University of California Berkeley Berkeley, CA, USA
| | - Chris D Vulpe
- Department of Nutritional Science and Toxicology, University of California Berkeley Berkeley, CA, USA
| |
Collapse
|
93
|
Abstract
The use of model organisms as tools for the investigation of human genetic variation has significantly and rapidly advanced our understanding of the aetiologies underlying hereditary traits. However, while equivalences in the DNA sequence of two species may be readily inferred through evolutionary models, the identification of equivalence in the phenotypic consequences resulting from comparable genetic variation is far from straightforward, limiting the value of the modelling paradigm. In this review, we provide an overview of the emerging statistical and computational approaches to objectively identify phenotypic equivalence between human and model organisms with examples from the vertebrate models, mouse and zebrafish. Firstly, we discuss enrichment approaches, which deem the most frequent phenotype among the orthologues of a set of genes associated with a common human phenotype as the orthologous phenotype, or phenolog, in the model species. Secondly, we introduce and discuss computational reasoning approaches to identify phenotypic equivalences made possible through the development of intra- and interspecies ontologies. Finally, we consider the particular challenges involved in modelling neuropsychiatric disorders, which illustrate many of the remaining difficulties in developing comprehensive and unequivocal interspecies phenotype mappings.
Collapse
Affiliation(s)
- Peter N. Robinson
- Institute for Medical Genetics and Human Genetics, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Berlin Brandenburg Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin Berlin, Berlin, Germany
- Max Planck Institute for Molecular Genetics, Berlin, Germany
- Institute for Bioinformatics, Department of Mathematics and Computer Science, Freie Universität Berlin, Berlin, Germany
- * E-mail: (PNR); (CW)
| | - Caleb Webber
- MRC Functional Genomics Unit, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
- * E-mail: (PNR); (CW)
| |
Collapse
|
94
|
Abstract
The epidermis functions as a physical barrier to the external environment and works to prevent loss of water from the skin. Numerous factors have been implicated in the formation of epidermal barriers, such as cornified envelopes, corneocytes, lipids, junctional proteins, proteases, protease inhibitors, antimicrobial peptides, and transcription factors. This review illustrates human diseases (ichthyoses) and animal models in which the epidermal barrier is disrupted or dysfunctional at steady state owing to ablation of one or more of the above factors. These diseases and animal models help us to understand the complicated mechanisms of epidermal barrier formation and give further insights on epidermal development.
Collapse
|
95
|
Cowling BS, Chevremont T, Prokic I, Kretz C, Ferry A, Coirault C, Koutsopoulos O, Laugel V, Romero NB, Laporte J. Reducing dynamin 2 expression rescues X-linked centronuclear myopathy. J Clin Invest 2014; 124:1350-63. [PMID: 24569376 DOI: 10.1172/jci71206] [Citation(s) in RCA: 100] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2013] [Accepted: 11/21/2013] [Indexed: 12/29/2022] Open
Abstract
Centronuclear myopathies (CNM) are congenital disorders associated with muscle weakness and abnormally located nuclei in skeletal muscle. An autosomal dominant form of CNM results from mutations in the gene encoding dynamin 2 (DNM2), and loss-of-function mutations in the gene encoding myotubularin (MTM1) result in X-linked CNM (XLCNM, also called myotubular myopathy), which promotes severe neonatal hypotonia and early death. Currently, no effective treatments exist for XLCNM. Here, we found increased DNM2 levels in XLCNM patients and a mouse model of XLCNM (Mtm1(-/y)). Generation of Mtm1(-/y) mice that were heterozygous for Dnm2 revealed that reduction of DNM2 in XLCNM mice restored life span, whole-body strength, and diaphragm function and increased muscle strength. Additionally, classic CNM-associated histological features, including fiber atrophy and nuclei mispositioning, were absent or reduced. Ultrastructural analysis revealed improvement of sarcomere organization and triad structures. Skeletal muscle-specific decrease of Dnm2 during embryogenesis or in young mice after disease onset revealed that the rescue associated with downregulation of Dnm2 is cell autonomous and is able to stop and potentially revert XLCNM progression. These data indicate that MTM1 and DNM2 regulate muscle organization and force through a common pathway. Furthermore, despite DNM2 being a key mechanoenzyme, its reduction is beneficial for XLCNM and represents a potential therapeutic approach for patients.
Collapse
MESH Headings
- Animals
- Diaphragm/physiopathology
- Down-Regulation
- Dynamin II/genetics
- Dynamin II/metabolism
- Female
- Gene Expression
- Humans
- Male
- Mice
- Mice, Knockout
- Muscle Contraction
- Muscle Strength
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/pathology
- Myopathies, Structural, Congenital/metabolism
- Myopathies, Structural, Congenital/pathology
- Myopathies, Structural, Congenital/therapy
- Phenotype
- Protein Tyrosine Phosphatases, Non-Receptor/metabolism
Collapse
|
96
|
Abstract
The endolysosomal system and autophagy are essential components of macromolecular turnover in eukaryotic cells. The low-abundance signaling lipid PI(3,5)P2 is a key regulator of this pathway. Analysis of mouse models with defects in PI(3,5)P2 biosynthesis has revealed the unique dependence of the mammalian nervous system on this signaling pathway. This insight led to the discovery of the molecular basis for several human neurological disorders, including Charcot-Marie-Tooth disease and Yunis-Varon syndrome. Spontaneous mutants, conditional knockouts, transgenic lines, and gene-trap alleles of Fig4, Vac14, and Pikfyve (Fab1) in the mouse have provided novel information regarding the role of PI(3,5)P2in vivo. This review summarizes what has been learned from mouse models and highlights the utility of manipulating complex signaling pathways in vivo.
Collapse
Affiliation(s)
- Guy M Lenk
- Department of Human Genetics, University of Michigan, Ann Arbor, Michigan, USA
| | - Miriam H Meisler
- Department of Human Genetics, University of Michigan, Ann Arbor, Michigan, USA.
| |
Collapse
|
97
|
Mashimo T. Gene targeting technologies in rats: zinc finger nucleases, transcription activator-like effector nucleases, and clustered regularly interspaced short palindromic repeats. Dev Growth Differ 2013; 56:46-52. [PMID: 24372523 DOI: 10.1111/dgd.12110] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2013] [Revised: 11/18/2013] [Accepted: 11/18/2013] [Indexed: 12/21/2022]
Abstract
The laboratory rat has been widely used as an animal model in biomedical science for more than 150 years. Applying zinc-finger nucleases or transcription activator-like effector nucleases to rat embryos via microinjection is an efficient genome editing tool for generating targeted knockout rats. Recently, clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated endonucleases have been used as an effective tool for precise and multiplex genome editing in mice and rats. In this review, the advantages and disadvantages of these site-specific nuclease technologies for genetic analysis and manipulation in rats are discussed.
Collapse
Affiliation(s)
- Tomoji Mashimo
- Institute of Laboratory Animals, Graduate School of Medicine, Kyoto University, Yoshidakonoe-cho Sakyo-ku, Kyoto, 606-8501, Japan
| |
Collapse
|
98
|
Everitt AR, Clare S, McDonald JU, Kane L, Harcourt K, Ahras M, Lall A, Hale C, Rodgers A, Young DB, Haque A, Billker O, Tregoning JS, Dougan G, Kellam P. Defining the range of pathogens susceptible to Ifitm3 restriction using a knockout mouse model. PLoS One 2013; 8:e80723. [PMID: 24278312 PMCID: PMC3836756 DOI: 10.1371/journal.pone.0080723] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Accepted: 10/16/2013] [Indexed: 12/22/2022] Open
Abstract
The interferon-inducible transmembrane (IFITM) family of proteins has been shown to restrict a broad range of viruses in vitro and in vivo by halting progress through the late endosomal pathway. Further, single nucleotide polymorphisms (SNPs) in its sequence have been linked with risk of developing severe influenza virus infections in humans. The number of viruses restricted by this host protein has continued to grow since it was first demonstrated as playing an antiviral role; all of which enter cells via the endosomal pathway. We therefore sought to test the limits of antimicrobial restriction by Ifitm3 using a knockout mouse model. We showed that Ifitm3 does not impact on the restriction or pathogenesis of bacterial (Salmonella typhimurium, Citrobacter rodentium, Mycobacterium tuberculosis) or protozoan (Plasmodium berghei) pathogens, despite in vitro evidence. However, Ifitm3 is capable of restricting respiratory syncytial virus (RSV) in vivo either through directly restricting RSV cell infection, or by exerting a previously uncharacterised function controlling disease pathogenesis. This represents the first demonstration of a virus that enters directly through the plasma membrane, without the need for the endosomal pathway, being restricted by the IFITM family; therefore further defining the role of these antiviral proteins.
Collapse
Affiliation(s)
- Aaron R. Everitt
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, United Kingdom
- * E-mail:
| | - Simon Clare
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, United Kingdom
| | - Jacqueline U. McDonald
- Mucosal Infection and Immunity Group, Section of Infectious Diseases, Department of Medicine, Imperial College London, London, United Kingdom
| | - Leanne Kane
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, United Kingdom
| | - Katherine Harcourt
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, United Kingdom
| | - Malika Ahras
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, United Kingdom
| | - Amar Lall
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Christine Hale
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, United Kingdom
| | - Angela Rodgers
- Medical Research Council National Institute for Medical Research, London, United Kingdom
| | - Douglas B. Young
- Medical Research Council National Institute for Medical Research, London, United Kingdom
| | - Ashraful Haque
- Malaria Immunology Laboratory, Queensland Institute of Medical Research and The Australian Centre for Vaccine Development, Herston, Brisbane, Queensland, Australia
| | - Oliver Billker
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, United Kingdom
| | - John S. Tregoning
- Mucosal Infection and Immunity Group, Section of Infectious Diseases, Department of Medicine, Imperial College London, London, United Kingdom
| | - Gordon Dougan
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, United Kingdom
| | - Paul Kellam
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, United Kingdom
- Department of Infection, University College London, London, United Kingdom
| |
Collapse
|
99
|
Robinson PN, Köhler S, Oellrich A, Wang K, Mungall CJ, Lewis SE, Washington N, Bauer S, Seelow D, Krawitz P, Gilissen C, Haendel M, Smedley D. Improved exome prioritization of disease genes through cross-species phenotype comparison. Genome Res 2013; 24:340-8. [PMID: 24162188 PMCID: PMC3912424 DOI: 10.1101/gr.160325.113] [Citation(s) in RCA: 248] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Numerous new disease-gene associations have been identified by whole-exome sequencing studies in the last few years. However, many cases remain unsolved due to the sheer number of candidate variants remaining after common filtering strategies such as removing low quality and common variants and those deemed unlikely to be pathogenic. The observation that each of our genomes contains about 100 genuine loss-of-function variants makes identification of the causative mutation problematic when using these strategies alone. We propose using the wealth of genotype to phenotype data that already exists from model organism studies to assess the potential impact of these exome variants. Here, we introduce PHenotypic Interpretation of Variants in Exomes (PHIVE), an algorithm that integrates the calculation of phenotype similarity between human diseases and genetically modified mouse models with evaluation of the variants according to allele frequency, pathogenicity, and mode of inheritance approaches in our Exomiser tool. Large-scale validation of PHIVE analysis using 100,000 exomes containing known mutations demonstrated a substantial improvement (up to 54.1-fold) over purely variant-based (frequency and pathogenicity) methods with the correct gene recalled as the top hit in up to 83% of samples, corresponding to an area under the ROC curve of >95%. We conclude that incorporation of phenotype data can play a vital role in translational bioinformatics and propose that exome sequencing projects should systematically capture clinical phenotypes to take advantage of the strategy presented here.
Collapse
Affiliation(s)
- Peter N Robinson
- Institute for Medical and Human Genetics, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
100
|
White JK, Gerdin AK, Karp NA, Ryder E, Buljan M, Bussell JN, Salisbury J, Clare S, Ingham NJ, Podrini C, Houghton R, Estabel J, Bottomley JR, Melvin DG, Sunter D, Adams NC, Tannahill D, Logan DW, Macarthur DG, Flint J, Mahajan VB, Tsang SH, Smyth I, Watt FM, Skarnes WC, Dougan G, Adams DJ, Ramirez-Solis R, Bradley A, Steel KP. Genome-wide generation and systematic phenotyping of knockout mice reveals new roles for many genes. Cell 2013; 154:452-64. [PMID: 23870131 PMCID: PMC3717207 DOI: 10.1016/j.cell.2013.06.022] [Citation(s) in RCA: 393] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2013] [Revised: 05/10/2013] [Accepted: 06/17/2013] [Indexed: 02/06/2023]
Abstract
Mutations in whole organisms are powerful ways of interrogating gene function in a realistic context. We describe a program, the Sanger Institute Mouse Genetics Project, that provides a step toward the aim of knocking out all genes and screening each line for a broad range of traits. We found that hitherto unpublished genes were as likely to reveal phenotypes as known genes, suggesting that novel genes represent a rich resource for investigating the molecular basis of disease. We found many unexpected phenotypes detected only because we screened for them, emphasizing the value of screening all mutants for a wide range of traits. Haploinsufficiency and pleiotropy were both surprisingly common. Forty-two percent of genes were essential for viability, and these were less likely to have a paralog and more likely to contribute to a protein complex than other genes. Phenotypic data and more than 900 mutants are openly available for further analysis. PaperClip
Large openly available resource of targeted mouse mutants and phenotypic data Screen for broad range of disease features and traits Many novel phenotypes suggest functions for both studied and unstudied genes Haploinsufficiency and pleiotropy are common
Collapse
|