1
|
Agca Y, Amos-Landgraf J, Araiza R, Brennan J, Carlson C, Ciavatta D, Clary D, Franklin C, Korf I, Lutz C, Magnuson T, de Villena FPM, Mirochnitchenko O, Patel S, Port D, Reinholdt L, Lloyd KCK. The mutant mouse resource and research center (MMRRC) consortium: the US-based public mouse repository system. Mamm Genome 2024; 35:524-536. [PMID: 39304538 PMCID: PMC11522152 DOI: 10.1007/s00335-024-10070-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 09/10/2024] [Indexed: 09/22/2024]
Abstract
Now in its 25th year, the Mutant Mouse Resource and Research Center (MMRRC) consortium continues to serve the United States and international biomedical scientific community as a public repository and distribution archive of laboratory mouse models of human disease for research. Supported by the National Institutes of Health (NIH), the MMRRC consists of 4 regionally distributed and dedicated vivaria, offices, and specialized laboratory facilities and an Informatics Coordination and Service Center (ICSC). The overarching purpose of the MMRRC is to facilitate groundbreaking biomedical research by offering an extensive repertoire of mutant mice that are essential for advancing the understanding of human physiology and disease. The function of the MMRRC is to identify, acquire, evaluate, characterize, cryopreserve, and distribute mutant mouse strains to qualified biomedical investigators around the nation and the globe. Mouse strains accepted from the research community are held to the highest scientific standards to optimize reproducibility and enhance scientific rigor and transparency. All submitted strains are thoroughly reviewed, documented, and validated using extensive scientific quality control measures. In addition, the MMRRC conducts resource-related research on cryopreservation, mouse genetics, environmental conditions, and other topics that enhance operations of the MMRRC. Today, the MMRRC maintains an archive of mice, cryopreserved embryos and sperm, embryonic stem (ES) cell lines, and murine hybridomas for nearly 65,000 alleles. Since its inception, the MMRRC has fulfilled more than 20,000 orders from 13,651 scientists at 8441 institutions worldwide. The MMRRC also provides numerous services to assist researchers, including scientific consultation, technical assistance, genetic assays, microbiome analysis, analytical phenotyping, pathology, cryorecovery, husbandry, breeding and colony management, infectious disease surveillance, and disease modeling. The ICSC coordinates MMRRC operations, interacts with researchers, and manages the website (mmrrc.org) and online catalogue. Researchers benefit from an expansive list of well-defined mouse models of disease that meet the highest scientific standards while submitting investigators benefit by having their mouse strains cryopreserved, protected, and distributed in compliance with NIH policies.
Collapse
Affiliation(s)
- Yuksel Agca
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, MO, USA
| | - James Amos-Landgraf
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, MO, USA
| | - Renee Araiza
- Department of Molecular and Cellular Biology, College of Biological Sciences and Bioinformatics Core, Genome Center, University of California, Davis, CA, USA
- Mouse Biology Program, University of California, Davis, CA, USA
| | - Jennifer Brennan
- Department of Genetics and Lineberger Comprehensive Cancer Center, University of North Carolina-Chapel Hill, Chapel Hill, NC, USA
| | - Charisse Carlson
- Department of Molecular and Cellular Biology, College of Biological Sciences and Bioinformatics Core, Genome Center, University of California, Davis, CA, USA
- Mouse Biology Program, University of California, Davis, CA, USA
| | - Dominic Ciavatta
- Department of Genetics and Lineberger Comprehensive Cancer Center, University of North Carolina-Chapel Hill, Chapel Hill, NC, USA
| | - Dave Clary
- Department of Molecular and Cellular Biology, College of Biological Sciences and Bioinformatics Core, Genome Center, University of California, Davis, CA, USA
- Mouse Biology Program, University of California, Davis, CA, USA
| | - Craig Franklin
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, MO, USA
| | - Ian Korf
- Department of Molecular and Cellular Biology, College of Biological Sciences and Bioinformatics Core, Genome Center, University of California, Davis, CA, USA
| | | | - Terry Magnuson
- Department of Genetics and Lineberger Comprehensive Cancer Center, University of North Carolina-Chapel Hill, Chapel Hill, NC, USA
| | - Fernando Pardo-Manuel de Villena
- Department of Genetics and Lineberger Comprehensive Cancer Center, University of North Carolina-Chapel Hill, Chapel Hill, NC, USA
| | - Oleg Mirochnitchenko
- Division of Comparative Medicine, Office of Research Infrastructure Programs, Division of Program Coordination, Planning, and Strategic Initiatives, Office of the Director, National Institutes of Health, Bethesda, USA
| | - Samit Patel
- Department of Molecular and Cellular Biology, College of Biological Sciences and Bioinformatics Core, Genome Center, University of California, Davis, CA, USA
| | - Dan Port
- Department of Molecular and Cellular Biology, College of Biological Sciences and Bioinformatics Core, Genome Center, University of California, Davis, CA, USA
- Mouse Biology Program, University of California, Davis, CA, USA
| | | | - K C Kent Lloyd
- Mouse Biology Program, University of California, Davis, CA, USA.
- Department of Surgery, School of Medicine, University of California, Davis, CA, USA.
| |
Collapse
|
2
|
Fernandez ME, Martinez-Romero J, Aon MA, Bernier M, Price NL, de Cabo R. How is Big Data reshaping preclinical aging research? Lab Anim (NY) 2023; 52:289-314. [PMID: 38017182 DOI: 10.1038/s41684-023-01286-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 10/10/2023] [Indexed: 11/30/2023]
Abstract
The exponential scientific and technological progress during the past 30 years has favored the comprehensive characterization of aging processes with their multivariate nature, leading to the advent of Big Data in preclinical aging research. Spanning from molecular omics to organism-level deep phenotyping, Big Data demands large computational resources for storage and analysis, as well as new analytical tools and conceptual frameworks to gain novel insights leading to discovery. Systems biology has emerged as a paradigm that utilizes Big Data to gain insightful information enabling a better understanding of living organisms, visualized as multilayered networks of interacting molecules, cells, tissues and organs at different spatiotemporal scales. In this framework, where aging, health and disease represent emergent states from an evolving dynamic complex system, context given by, for example, strain, sex and feeding times, becomes paramount for defining the biological trajectory of an organism. Using bioinformatics and artificial intelligence, the systems biology approach is leading to remarkable advances in our understanding of the underlying mechanism of aging biology and assisting in creative experimental study designs in animal models. Future in-depth knowledge acquisition will depend on the ability to fully integrate information from different spatiotemporal scales in organisms, which will probably require the adoption of theories and methods from the field of complex systems. Here we review state-of-the-art approaches in preclinical research, with a focus on rodent models, that are leading to conceptual and/or technical advances in leveraging Big Data to understand basic aging biology and its full translational potential.
Collapse
Affiliation(s)
- Maria Emilia Fernandez
- Experimental Gerontology Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Jorge Martinez-Romero
- Experimental Gerontology Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
- Laboratory of Epidemiology and Population Science, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Miguel A Aon
- Experimental Gerontology Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Michel Bernier
- Experimental Gerontology Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Nathan L Price
- Experimental Gerontology Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Rafael de Cabo
- Experimental Gerontology Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA.
| |
Collapse
|
3
|
Sundberg JP, Rice RH. Phenotyping mice with skin, hair, or nail abnormalities: A systematic approach and methodologies from simple to complex. Vet Pathol 2023; 60:829-842. [PMID: 37191004 DOI: 10.1177/03009858231170329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
The skin and adnexa can be difficult to interpret because they change dramatically with the hair cycle throughout life. However, a variety of methods are commonly available to collect skin and perform assays that can be useful for figuring out morphological and molecular changes. This overview provides information on basic approaches to evaluate skin and its molecular phenotype, with references for more detail, and interpretation of results on the skin and adnexa in the mouse. These approaches range from mouse genetic nomenclature, setting up a cutaneous phenotyping study, skin grafts, hair follicle reconstitution, wax stripping, electron microscopy, and Köbner reaction to very specific approaches such as lipid and protein analyses on a large scale.
Collapse
Affiliation(s)
- John P Sundberg
- The Jackson Laboratory, Bar Harbor, ME
- Vanderbilt University Medical Center, Nashville, TN
| | | |
Collapse
|
4
|
Bustin KA, Shishikura K, Chen I, Lin Z, McKnight N, Chang Y, Wang X, Li JJ, Arellano E, Pei L, Morton PD, Gregus AM, Buczynski MW, Matthews ML. Phenelzine-based probes reveal Secernin-3 is involved in thermal nociception. Mol Cell Neurosci 2023; 125:103842. [PMID: 36924917 PMCID: PMC10247460 DOI: 10.1016/j.mcn.2023.103842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 03/05/2023] [Accepted: 03/09/2023] [Indexed: 03/17/2023] Open
Abstract
Chemical platforms that facilitate both the identification and elucidation of new areas for therapeutic development are necessary but lacking. Activity-based protein profiling (ABPP) leverages active site-directed chemical probes as target discovery tools that resolve activity from expression and immediately marry the targets identified with lead compounds for drug design. However, this approach has traditionally focused on predictable and intrinsic enzyme functionality. Here, we applied our activity-based proteomics discovery platform to map non-encoded and post-translationally acquired enzyme functionalities (e.g. cofactors) in vivo using chemical probes that exploit the nucleophilic hydrazine pharmacophores found in a classic antidepressant drug (e.g. phenelzine, Nardil®). We show the probes are in vivo active and can map proteome-wide tissue-specific target engagement of the drug. In addition to engaging targets (flavoenzymes monoamine oxidase A/B) that are associated with the known therapeutic mechanism as well as several other members of the flavoenzyme family, the probes captured the previously discovered N-terminal glyoxylyl (Glox) group of Secernin-3 (SCRN3) in vivo through a divergent mechanism, indicating this functional feature has biochemical activity in the brain. SCRN3 protein is ubiquitously expressed in the brain, yet gene expression is regulated by inflammatory stimuli. In an inflammatory pain mouse model, behavioral assessment of nociception showed Scrn3 male knockout mice selectively exhibited impaired thermal nociceptive sensitivity. Our study provides a guided workflow to entangle molecular (off)targets and pharmacological mechanisms for therapeutic development.
Collapse
Affiliation(s)
- Katelyn A Bustin
- Department of Chemistry, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kyosuke Shishikura
- Department of Chemistry, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Irene Chen
- School of Neuroscience, Virginia Polytechnic and State University, Blacksburg, VA 24061, USA
| | - Zongtao Lin
- Department of Chemistry, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Nate McKnight
- Department of Chemistry, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yuxuan Chang
- Department of Chemistry, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Xie Wang
- Department of Chemistry, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jing Jing Li
- Center for Mitochondrial and Epigenomic Medicine, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Eric Arellano
- Department of Chemistry, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Liming Pei
- Center for Mitochondrial and Epigenomic Medicine, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Paul D Morton
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic and State University, Blacksburg, VA, 24060, USA
| | - Ann M Gregus
- School of Neuroscience, Virginia Polytechnic and State University, Blacksburg, VA 24061, USA.
| | - Matthew W Buczynski
- School of Neuroscience, Virginia Polytechnic and State University, Blacksburg, VA 24061, USA.
| | - Megan L Matthews
- Department of Chemistry, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
5
|
da Silva-Buttkus P, Spielmann N, Klein-Rodewald T, Schütt C, Aguilar-Pimentel A, Amarie OV, Becker L, Calzada-Wack J, Garrett L, Gerlini R, Kraiger M, Leuchtenberger S, Östereicher MA, Rathkolb B, Sanz-Moreno A, Stöger C, Hölter SM, Seisenberger C, Marschall S, Fuchs H, Gailus-Durner V, Hrabě de Angelis M. Knockout mouse models as a resource for the study of rare diseases. Mamm Genome 2023; 34:244-261. [PMID: 37160609 PMCID: PMC10290595 DOI: 10.1007/s00335-023-09986-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 03/07/2023] [Indexed: 05/11/2023]
Abstract
Rare diseases (RDs) are a challenge for medicine due to their heterogeneous clinical manifestations and low prevalence. There is a lack of specific treatments and only a few hundred of the approximately 7,000 RDs have an approved regime. Rapid technological development in genome sequencing enables the mass identification of potential candidates that in their mutated form could trigger diseases but are often not confirmed to be causal. Knockout (KO) mouse models are essential to understand the causality of genes by allowing highly standardized research into the pathogenesis of diseases. The German Mouse Clinic (GMC) is one of the pioneers in mouse research and successfully uses (preclinical) data obtained from single-gene KO mutants for research into monogenic RDs. As part of the International Mouse Phenotyping Consortium (IMPC) and INFRAFRONTIER, the pan-European consortium for modeling human diseases, the GMC expands these preclinical data toward global collaborative approaches with researchers, clinicians, and patient groups.Here, we highlight proprietary genes that when deleted mimic clinical phenotypes associated with known RD targets (Nacc1, Bach2, Klotho alpha). We focus on recognized RD genes with no pre-existing KO mouse models (Kansl1l, Acsf3, Pcdhgb2, Rabgap1, Cox7a2) which highlight novel phenotypes capable of optimizing clinical diagnosis. In addition, we present genes with intriguing phenotypic data (Zdhhc5, Wsb2) that are not presently associated with known human RDs.This report provides comprehensive evidence for genes that when deleted cause differences in the KO mouse across multiple organs, providing a huge translational potential for further understanding monogenic RDs and their clinical spectrum. Genetic KO studies in mice are valuable to further explore the underlying physiological mechanisms and their overall therapeutic potential.
Collapse
Affiliation(s)
- Patricia da Silva-Buttkus
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstaedter Landstrasse 1, Neuherberg, Germany
| | - Nadine Spielmann
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstaedter Landstrasse 1, Neuherberg, Germany
| | - Tanja Klein-Rodewald
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstaedter Landstrasse 1, Neuherberg, Germany
| | - Christine Schütt
- Institute of Experimental Genetics, Applied Computational Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstaedter Landstrasse 1, Neuherberg, Germany
| | - Antonio Aguilar-Pimentel
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstaedter Landstrasse 1, Neuherberg, Germany
| | - Oana V Amarie
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstaedter Landstrasse 1, Neuherberg, Germany
| | - Lore Becker
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstaedter Landstrasse 1, Neuherberg, Germany
| | - Julia Calzada-Wack
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstaedter Landstrasse 1, Neuherberg, Germany
| | - Lillian Garrett
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstaedter Landstrasse 1, Neuherberg, Germany
- Institute of Developmental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstaedter Landstrasse 1, Neuherberg, Germany
| | - Raffaele Gerlini
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstaedter Landstrasse 1, Neuherberg, Germany
| | - Markus Kraiger
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstaedter Landstrasse 1, Neuherberg, Germany
| | - Stefanie Leuchtenberger
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstaedter Landstrasse 1, Neuherberg, Germany
| | - Manuela A Östereicher
- Institute of Experimental Genetics, Applied Computational Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstaedter Landstrasse 1, Neuherberg, Germany
| | - Birgit Rathkolb
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstaedter Landstrasse 1, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Ingolstaedter Landstrasse 1, 85764, Neuherberg, Germany
- Institute of Molecular Animal Breeding and Biotechnology, Gene Center, Ludwig-Maximilians-Universität München, Feodor-Lynen Strasse 25, 81377, Munich, Germany
| | - Adrián Sanz-Moreno
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstaedter Landstrasse 1, Neuherberg, Germany
| | - Claudia Stöger
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstaedter Landstrasse 1, Neuherberg, Germany
| | - Sabine M Hölter
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstaedter Landstrasse 1, Neuherberg, Germany
- Institute of Developmental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstaedter Landstrasse 1, Neuherberg, Germany
| | - Claudia Seisenberger
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstaedter Landstrasse 1, Neuherberg, Germany
| | - Susan Marschall
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstaedter Landstrasse 1, Neuherberg, Germany
| | - Helmut Fuchs
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstaedter Landstrasse 1, Neuherberg, Germany
| | - Valerie Gailus-Durner
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstaedter Landstrasse 1, Neuherberg, Germany
| | - Martin Hrabě de Angelis
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstaedter Landstrasse 1, Neuherberg, Germany.
- German Center for Diabetes Research (DZD), Ingolstaedter Landstrasse 1, 85764, Neuherberg, Germany.
- Chair of Experimental Genetics, TUM School of Life Sciences, Technische Universität München, Alte Akademie 8, 85354, Freising, Germany.
| |
Collapse
|
6
|
Holroyd NA, Walsh C, Gourmet L, Walker-Samuel S. Quantitative Image Processing for Three-Dimensional Episcopic Images of Biological Structures: Current State and Future Directions. Biomedicines 2023; 11:909. [PMID: 36979887 PMCID: PMC10045950 DOI: 10.3390/biomedicines11030909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 03/03/2023] [Accepted: 03/10/2023] [Indexed: 03/17/2023] Open
Abstract
Episcopic imaging using techniques such as High Resolution Episcopic Microscopy (HREM) and its variants, allows biological samples to be visualized in three dimensions over a large field of view. Quantitative analysis of episcopic image data is undertaken using a range of methods. In this systematic review, we look at trends in quantitative analysis of episcopic images and discuss avenues for further research. Papers published between 2011 and 2022 were analyzed for details about quantitative analysis approaches, methods of image annotation and choice of image processing software. It is shown that quantitative processing is becoming more common in episcopic microscopy and that manual annotation is the predominant method of image analysis. Our meta-analysis highlights where tools and methods require further development in this field, and we discuss what this means for the future of quantitative episcopic imaging, as well as how annotation and quantification may be automated and standardized across the field.
Collapse
Affiliation(s)
| | - Claire Walsh
- Centre for Computational Medicine, University College London, London WC1E 6DD, UK
- Department of Mechanical Engineering, University College London, London WC1E 7JE, UK
| | - Lucie Gourmet
- Centre for Computational Medicine, University College London, London WC1E 6DD, UK
| | - Simon Walker-Samuel
- Centre for Computational Medicine, University College London, London WC1E 6DD, UK
| |
Collapse
|
7
|
Atf7ip Inhibits Osteoblast Differentiation via Negative Regulation of the Sp7 Transcription Factor. Int J Mol Sci 2023; 24:ijms24054305. [PMID: 36901736 PMCID: PMC10002255 DOI: 10.3390/ijms24054305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 02/03/2023] [Accepted: 02/15/2023] [Indexed: 02/24/2023] Open
Abstract
Epigenetic modifications are critical for cell differentiation and growth. As a regulator of H3K9 methylation, Setdb1 is implicated in osteoblast proliferation and differentiation. The activity and nucleus localization of Setdb1 are regulated by its binding partner, Atf7ip. However, whether Atf7ip is involved in the regulation of osteoblast differentiation remains largely unclear. In the present study, we found that Atf7ip expression was upregulated during the osteogenesis of primary bone marrow stromal cells and MC3T3-E1 cells, and was induced in PTH-treated cells. The overexpression of Atf7ip impaired osteoblast differentiation in MC3T3-E1 cells regardless of PTH treatment, as measured by the expression of osteoblast differentiation markers, Alp-positive cells, Alp activity, and calcium deposition. Conversely, the depletion of Atf7ip in MC3T3-E1 cells promoted osteoblast differentiation. Compared with the control mice, animals with Atf7ip deletion in the osteoblasts (Oc-Cre;Atf7ipf/f) showed more bone formation and a significant increase in the bone trabeculae microarchitecture, as reflected by μ-CT and bone histomorphometry. Mechanistically, Atf7ip contributed to the nucleus localization of Setdb1 in MC3T3-E1, but did not affect Setdb1 expression. Atf7ip negatively regulated Sp7 expression, and through specific siRNA, Sp7 knockdown attenuated the enhancing role of Atf7ip deletion in osteoblast differentiation. Through these data, we identified Atf7ip as a novel negative regulator of osteogenesis, possibly via its epigenetic regulation of Sp7 expression, and demonstrated that Atf7ip inhibition is a potential therapeutic measure for enhancing bone formation.
Collapse
|
8
|
Bustin KA, Shishikura K, Chen I, Lin Z, McKnight N, Chang Y, Wang X, Li JJ, Arellano E, Pei L, Morton PD, Gregus AM, Buczynski MW, Matthews ML. Phenelzine-based probes reveal Secernin-3 is involved in thermal nociception. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.02.526866. [PMID: 36778412 PMCID: PMC9915563 DOI: 10.1101/2023.02.02.526866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Chemical platforms that facilitate both the identification and elucidation of new areas for therapeutic development are necessary but lacking. Activity-based protein profiling (ABPP) leverages active site-directed chemical probes as target discovery tools that resolve activity from expression and immediately marry the targets identified with lead compounds for drug design. However, this approach has traditionally focused on predictable and intrinsic enzyme functionality. Here, we applied our activity-based proteomics discovery platform to map non-encoded and post-translationally acquired enzyme functionalities (e.g. cofactors) in vivo using chemical probes that exploit the nucleophilic hydrazine pharmacophores found in a classic antidepressant drug (e.g. phenelzine, Nardil ® ). We show the probes are in vivo active and can map proteome-wide tissue-specific target engagement of the drug. In addition to engaging targets (flavoenzymes monoamine oxidase A/B) that are associated with the known therapeutic mechanism as well as several other members of the flavoenzyme family, the probes captured the previously discovered N -terminal glyoxylyl (Glox) group of Secernin-3 (SCRN3) in vivo through a divergent mechanism, indicating this functional feature has biochemical activity in the brain. SCRN3 protein is ubiquitously expressed in the brain, yet gene expression is regulated by inflammatory stimuli. In an inflammatory pain mouse model, behavioral assessment of nociception showed Scrn3 male knockout mice selectively exhibited impaired thermal nociceptive sensitivity. Our study provides a guided workflow to entangle molecular (off)targets and pharmacological mechanisms for therapeutic development.
Collapse
|
9
|
Chee JM, Lanoue L, Clary D, Higgins K, Bower L, Flenniken A, Guo R, Adams DJ, Bosch F, Braun RE, Brown SDM, Chin HJG, Dickinson ME, Hsu CW, Dobbie M, Gao X, Galande S, Grobler A, Heaney JD, Herault Y, de Angelis MH, Mammano F, Nutter LMJ, Parkinson H, Qin C, Shiroishi T, Sedlacek R, Seong JK, Xu Y, Brooks B, McKerlie C, Lloyd KCK, Westerberg H, Moshiri A. Genome-wide screening reveals the genetic basis of mammalian embryonic eye development. BMC Biol 2023; 21:22. [PMID: 36737727 PMCID: PMC9898963 DOI: 10.1186/s12915-022-01475-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 11/23/2022] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Microphthalmia, anophthalmia, and coloboma (MAC) spectrum disease encompasses a group of eye malformations which play a role in childhood visual impairment. Although the predominant cause of eye malformations is known to be heritable in nature, with 80% of cases displaying loss-of-function mutations in the ocular developmental genes OTX2 or SOX2, the genetic abnormalities underlying the remaining cases of MAC are incompletely understood. This study intended to identify the novel genes and pathways required for early eye development. Additionally, pathways involved in eye formation during embryogenesis are also incompletely understood. This study aims to identify the novel genes and pathways required for early eye development through systematic forward screening of the mammalian genome. RESULTS Query of the International Mouse Phenotyping Consortium (IMPC) database (data release 17.0, August 01, 2022) identified 74 unique knockout lines (genes) with genetically associated eye defects in mouse embryos. The vast majority of eye abnormalities were small or absent eyes, findings most relevant to MAC spectrum disease in humans. A literature search showed that 27 of the 74 lines had previously published knockout mouse models, of which only 15 had ocular defects identified in the original publications. These 12 previously published gene knockouts with no reported ocular abnormalities and the 47 unpublished knockouts with ocular abnormalities identified by the IMPC represent 59 genes not previously associated with early eye development in mice. Of these 59, we identified 19 genes with a reported human eye phenotype. Overall, mining of the IMPC data yielded 40 previously unimplicated genes linked to mammalian eye development. Bioinformatic analysis showed that several of the IMPC genes colocalized to several protein anabolic and pluripotency pathways in early eye development. Of note, our analysis suggests that the serine-glycine pathway producing glycine, a mitochondrial one-carbon donator to folate one-carbon metabolism (FOCM), is essential for eye formation. CONCLUSIONS Using genome-wide phenotype screening of single-gene knockout mouse lines, STRING analysis, and bioinformatic methods, this study identified genes heretofore unassociated with MAC phenotypes providing models to research novel molecular and cellular mechanisms involved in eye development. These findings have the potential to hasten the diagnosis and treatment of this congenital blinding disease.
Collapse
Affiliation(s)
- Justine M Chee
- Oakland University William Beaumont School of Medicine, Rochester, MI, USA
| | - Louise Lanoue
- Mouse Biology Program, University of California Davis, Davis, CA, USA
| | - Dave Clary
- Mouse Biology Program, University of California Davis, Davis, CA, USA
| | - Kendall Higgins
- University of Miami: Miller School of Medicine, Miami, FL, USA
| | - Lynette Bower
- Mouse Biology Program, University of California Davis, Davis, CA, USA
| | - Ann Flenniken
- The Centre for Phenogenomics, Toronto, ON, Canada
- Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, ON, Canada
| | - Ruolin Guo
- The Centre for Phenogenomics, Toronto, ON, Canada
- The Hospital for Sick Children, Toronto, ON, Canada
| | - David J Adams
- The Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Fatima Bosch
- Centre of Animal Biotechnology and Gene Therapy (CBATEG), Universitat Autònoma de Barcelona, Barcelona, Spain
| | | | - Steve D M Brown
- Medical Research Council Harwell Institute, Mammalian Genetics Unit and Mary Lyon Centre, Harwell Campus, Oxfordshire, UK
| | - H-J Genie Chin
- National Laboratory Animal Center, National Applied Research Laboratories (NARLabs), Taipei City, Taiwan
| | - Mary E Dickinson
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Chih-Wei Hsu
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Michael Dobbie
- Phenomics Australia, The John Curtin School of Medical Research, Canberra, Australia
| | - Xiang Gao
- Nanjing Biomedical Research Institute, Nanjing University, Nanjing, China
| | - Sanjeev Galande
- Indian Institutes of Science Education and Research, Pune, India
| | - Anne Grobler
- Faculty of Health Sciences, PCDDP North-West University, Potchefstroom, South Africa
| | - Jason D Heaney
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Yann Herault
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Université de Strasbourg, Illkirch, France
| | - Martin Hrabe de Angelis
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, Neuherberg, Germany
| | - Fabio Mammano
- Monterotondo Mouse Clinic, Italian National Research Council (CNR), Monterotondo Scalo, Italy
| | - Lauryl M J Nutter
- The Centre for Phenogenomics, Toronto, ON, Canada
- The Hospital for Sick Children, Toronto, ON, Canada
| | - Helen Parkinson
- European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridgeshire, UK
| | - Chuan Qin
- National Laboratory Animal Center, National Applied Research Laboratories, Beijing, China
| | | | - Radislav Sedlacek
- Czech Center for Phenogenomics, Institute of Molecular Genetics of the Czech Academy of Sciences, Vestec, Czech Republic
| | - J-K Seong
- Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| | - Ying Xu
- CAM-SU Genomic Resource Center, Soochow University, Suzhou, China
| | - Brian Brooks
- Ophthalmic Genetics and Visual Function Branch, National Eye Institute, NIH, Bethesda, MD, 20892, USA
| | - Colin McKerlie
- The Hospital for Sick Children, Toronto, ON, Canada
- Department of Laboratory Medicine & Pathobiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - K C Kent Lloyd
- Mouse Biology Program, University of California Davis, Davis, CA, USA
- Department of Surgery, School of Medicine, University of California Davis, Sacramento, CA, USA
| | - Henrik Westerberg
- Medical Research Council Harwell Institute, Mammalian Genetics Unit and Mary Lyon Centre, Harwell Campus, Oxfordshire, UK
| | - Ala Moshiri
- Department of Ophthalmology & Vision Science, School of Medicine, University of California Davis, Sacramento, CA, USA.
- UC Davis Eye Center, 4860 Y St., Ste. 2400, Sacramento, CA, 95817, USA.
| |
Collapse
|
10
|
Morohoshi A, Miyata H, Tokuhiro K, Iida-Norita R, Noda T, Fujihara Y, Ikawa M. Testis-enriched ferlin, FER1L5, is required for Ca 2+-activated acrosome reaction and male fertility. SCIENCE ADVANCES 2023; 9:eade7607. [PMID: 36696506 PMCID: PMC9876558 DOI: 10.1126/sciadv.ade7607] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 12/23/2022] [Indexed: 05/28/2023]
Abstract
Spermatozoa need to undergo an exocytotic event called the acrosome reaction before fusing with eggs. Although calcium ion (Ca2+) is essential for the acrosome reaction, its molecular mechanism remains unknown. Ferlin is a single transmembrane protein with multiple Ca2+-binding C2 domains, and there are six ferlins, dysferlin (DYSF), otoferlin (OTOF), myoferlin (MYOF), fer-1-like 4 (FER1L4), FER1L5, and FER1L6, in mammals. Dysf, Otof, and Myof knockout mice have been generated, and each knockout mouse line exhibited membrane fusion disorders such as muscular dystrophy in Dysf, deafness in Otof, and abnormal myogenesis in Myof. Here, by generating mutant mice of Fer1l4, Fer1l5, and Fer1l6, we found that only Fer1l5 is required for male fertility. Fer1l5 mutant spermatozoa could migrate in the female reproductive tract and reach eggs, but no acrosome reaction took place. Even a Ca2+ ionophore cannot induce the acrosome reaction in Fer1l5 mutant spermatozoa. These results suggest that FER1L5 is the missing link between Ca2+ and the acrosome reaction.
Collapse
Affiliation(s)
- Akane Morohoshi
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 5650871 Japan
- Graduate School of Medicine, Osaka University, Suita, Osaka 5650871 Japan
| | - Haruhiko Miyata
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 5650871 Japan
| | - Keizo Tokuhiro
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 5650871 Japan
- Institute of Biomedical Science, Kansai Medical University, Hirakata, Osaka 5731191 Japan
| | - Rie Iida-Norita
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 5650871 Japan
| | - Taichi Noda
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 5650871 Japan
- Institute of Resource Development and Analysis, Kumamoto University, Kumamoto, Kumamoto 8600811 Japan
- Priority Organization for Innovation and Excellence, Kumamoto University, Kumamoto, Kumamoto 8608555 Japan
| | - Yoshitaka Fujihara
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 5650871 Japan
- Department of Bioscience and Genetics, National Cerebral and Cardiovascular Center, Suita, Osaka 5648565, Japan
| | - Masahito Ikawa
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 5650871 Japan
- Graduate School of Medicine, Osaka University, Suita, Osaka 5650871 Japan
- The Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo 1088639 Japan
- Center for Infectious Disease Education and Research (CiDER), Osaka University, Suita, Osaka 5650871 Japan
| |
Collapse
|
11
|
Higgins K, Moore BA, Berberovic Z, Adissu HA, Eskandarian M, Flenniken AM, Shao A, Imai DM, Clary D, Lanoue L, Newbigging S, Nutter LMJ, Adams DJ, Bosch F, Braun RE, Brown SDM, Dickinson ME, Dobbie M, Flicek P, Gao X, Galande S, Grobler A, Heaney JD, Herault Y, de Angelis MH, Chin HJG, Mammano F, Qin C, Shiroishi T, Sedlacek R, Seong JK, Xu Y, Lloyd KCK, McKerlie C, Moshiri A. Analysis of genome-wide knockout mouse database identifies candidate ciliopathy genes. Sci Rep 2022; 12:20791. [PMID: 36456625 PMCID: PMC9715561 DOI: 10.1038/s41598-022-19710-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 09/02/2022] [Indexed: 12/05/2022] Open
Abstract
We searched a database of single-gene knockout (KO) mice produced by the International Mouse Phenotyping Consortium (IMPC) to identify candidate ciliopathy genes. We first screened for phenotypes in mouse lines with both ocular and renal or reproductive trait abnormalities. The STRING protein interaction tool was used to identify interactions between known cilia gene products and those encoded by the genes in individual knockout mouse strains in order to generate a list of "candidate ciliopathy genes." From this list, 32 genes encoded proteins predicted to interact with known ciliopathy proteins. Of these, 25 had no previously described roles in ciliary pathobiology. Histological and morphological evidence of phenotypes found in ciliopathies in knockout mouse lines are presented as examples (genes Abi2, Wdr62, Ap4e1, Dync1li1, and Prkab1). Phenotyping data and descriptions generated on IMPC mouse line are useful for mechanistic studies, target discovery, rare disease diagnosis, and preclinical therapeutic development trials. Here we demonstrate the effective use of the IMPC phenotype data to uncover genes with no previous role in ciliary biology, which may be clinically relevant for identification of novel disease genes implicated in ciliopathies.
Collapse
Affiliation(s)
- Kendall Higgins
- The University of Miami Leonard M. Miller School of Medicine, Miami, FL, 33136, USA
| | - Bret A Moore
- Department of Small Animal Clinical Sciences, University of Florida, College of Veterinary Medicine, Gainesville, FL, 32608, USA
| | - Zorana Berberovic
- The Centre for Phenogenomics, Toronto, ON, Canada
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, M5G 1X5, Canada
| | | | - Mohammad Eskandarian
- The Centre for Phenogenomics, Toronto, ON, Canada
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, M5G 1X5, Canada
| | - Ann M Flenniken
- The Centre for Phenogenomics, Toronto, ON, Canada
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, M5G 1X5, Canada
| | - Andy Shao
- University of Reno, Nevada, School of Medicine, Reno, NV, 89557, USA
| | - Denise M Imai
- Comparative Pathology Laboratory, U.C. Davis, Davis, 95616, USA
| | - Dave Clary
- Mouse Biology Program, U.C. Davis, Davis, CA, 95618, USA
| | - Louise Lanoue
- Mouse Biology Program, U.C. Davis, Davis, CA, 95618, USA
| | - Susan Newbigging
- The Centre for Phenogenomics, Toronto, ON, Canada
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, M5G 1X5, Canada
| | - Lauryl M J Nutter
- The Centre for Phenogenomics, Toronto, ON, Canada
- The Hospital for Sick Children, 555 University Avenue, Toronto, ON, M5G 1X8, Canada
| | - David J Adams
- The Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK
| | - Fatima Bosch
- Centre of Animal Biotechnology and Gene Therapy (CBATEG), Universitat Autònoma de Barcelona, 08193, Barcelona, Spain
| | | | - Steve D M Brown
- Medical Research Council Harwell Institute (Mammalian Genetics Unit and Mary Lyon Centre), Harwell Campus, Oxfordshire, OX11 0RD, UK
| | - Mary E Dickinson
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Michael Dobbie
- Phenomics Australia, The Australian National University, 131 Garran Rd, Acton, Canberra, ACT, 2601, Australia
| | - Paul Flicek
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SD, UK
| | - Xiang Gao
- SKL of Pharmaceutical Biotechnology and Model Animal Research Center, Collaborative Innovation Center for Genetics and Development, Nanjing Biomedical Research Institute, Nanjing University, Nanjing, 210061, China
| | - Sanjeev Galande
- Indian Institutes of Science Education and Research, Dr. Homi Bhabha Rd, Ward No. 8, NCL Colony, Pashan, Pune, Maharashtra, 411008, India
| | - Anne Grobler
- Faculty of Health Sciences, PCDDP North-West University, North-West University Potchefstroom Campus 11 Hoffman Street, Potchefstroom, 2531, South Africa
| | - Jason D Heaney
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Yann Herault
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Université de Strasbourg, 67400, Illkirch, France
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Université de Strasbourg, 1 rue Laurent Fries, 67404, Illkirch, France
- Centre National de la Recherche Scientifique, UMR7104, Illkirch, France
- Institut National de la Santé et de la Recherche Médicale, U1258, Illkirch, France
- Université de Strasbourg, 1 rue Laurent Fries, 67404, Illkirch, France
- CELPHEDIA, PHENOMIN, Institut Clinique de la Souris (ICS), CNRS, INSERM, Université of Strasbourg, 1 rue Laurent Fries, 67404, Illkirch-Graffenstaden, France
| | - Martin Hrabe de Angelis
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstraße 1, 85764, Neuherberg, Germany
| | - Hsian-Jean Genie Chin
- National Laboratory Animal Center, National Applied Research Laboratories (NARLabs), 3F., No. 106, Sec. 2, Heping E. Rd., Da'an Dist., Taipei City, 106214, Taiwan (R.O.C.)
| | - Fabio Mammano
- Monterotondo Mouse Clinic, Italian National Research Council (CNR), Institute of Cell Biology and Neurobiology, Adriano Buzzati-Traverso Campus, Via Ramarini, 00015, Monterotondo Scalo, Italy
| | - Chuan Qin
- National Laboratory Animal Center, National Applied Research Laboratories (NARLabs), Beijing, China
- Institute of Laboratory Animal Sciences, Chinese Academy of Medical Science, 5 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | | | - Radislav Sedlacek
- Czech Center for Phenogenomics, Institute of Molecular Genetics of the Czech Academy of Sciences, IMG BIOCEV Building SO.02 Prumyslova 595, 252 50, Vestec, Czech Republic
| | - J-K Seong
- Korea Mouse Phenotyping Consortium (KMPC) and BK21 Program for Veterinary Science, Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, 599 Gwanangno, Gwanak-gu, Seoul, 08826, South Korea
| | - Ying Xu
- CAM-SU Genomic Resource Center, Soochow University, Organization Planning of No. 1 Shizi Street, Suzhou, 215123, China
| | - K C Kent Lloyd
- Mouse Biology Program, U.C. Davis, Davis, CA, 95618, USA
- Department of Surgery, School of Medicine, U.C. Davis, Sacramento, CA, 95817, USA
| | - Colin McKerlie
- The Hospital for Sick Children, 555 University Avenue, Toronto, ON, M5G 1X8, Canada.
- Department of Laboratory Medicine and Pathobiology, Hospital for Sick Children (SickKids), The Centre for Phenogenomics, Faculty of Medicine, University of Toronto, 25 Orde Street, Toronto, ON, M5T 3H7, USA.
| | - Ala Moshiri
- Department of Ophthalmology and Vision Science, School of Medicine, U.C. Davis Eye Center, 4860 Y. Street, Suite 2400, Sacramento, CA, 95817, USA.
| |
Collapse
|
12
|
Reissig LF, Geyer SH, Winkler V, Preineder E, Prin F, Wilson R, Galli A, Tudor C, White JK, Mohun TJ, Weninger WJ. Detailed characterizations of cranial nerve anatomy in E14.5 mouse embryos/fetuses and their use as reference for diagnosing subtle, but potentially lethal malformations in mutants. Front Cell Dev Biol 2022; 10:1006620. [PMID: 36438572 PMCID: PMC9682249 DOI: 10.3389/fcell.2022.1006620] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 10/28/2022] [Indexed: 01/03/2024] Open
Abstract
Careful phenotype analysis of genetically altered mouse embryos/fetuses is vital for deciphering the function of pre- and perinatally lethal genes. Usually this involves comparing the anatomy of mutants with that of wild types of identical developmental stages. Detailed three dimensional information on regular cranial nerve (CN) anatomy of prenatal mice is very scarce. We therefore set out to provide such information to be used as reference data and selected mutants to demonstrate its potential for diagnosing CN abnormalities. Digital volume data of 152 wild type mice, harvested on embryonic day (E)14.5 and of 18 mutants of the Col4a2, Arid1b, Rpgrip1l and Cc2d2a null lines were examined. The volume data had been created with High Resolution Episcopic Microscopy (HREM) as part of the deciphering the mechanisms of developmental disorders (DMDD) program. Employing volume and surface models, oblique slicing and digital measuring tools, we provide highly detailed anatomic descriptions of the CNs and measurements of the diameter of selected segments. Specifics of the developmental stages of E14.5 mice and anatomic norm variations were acknowledged. Using the provided data as reference enabled us to objectively diagnose CN abnormalities, such as abnormal formation of CN3 (Col4a2), neuroma of the motor portion of CN5 (Arid1b), thinning of CN7 (Rpgrip1l) and abnormal topology of CN12 (Cc2d2a). Although, in a first glimpse perceived as unspectacular, defects of the motor CN5 or CN7, like enlargement or thinning can cause death of newborns, by hindering feeding. Furthermore, abnormal topology of CN12 was recently identified as a highly reliable marker for low penetrating, but potentially lethal defects of the central nervous system.
Collapse
Affiliation(s)
- Lukas F. Reissig
- Division of Anatomy, Center for Anatomy and Cell Biology, Medical University of Vienna, Vienna, Austria
| | - Stefan H. Geyer
- Division of Anatomy, Center for Anatomy and Cell Biology, Medical University of Vienna, Vienna, Austria
| | - Viola Winkler
- Division of Anatomy, Center for Anatomy and Cell Biology, Medical University of Vienna, Vienna, Austria
| | - Ester Preineder
- Division of Anatomy, Center for Anatomy and Cell Biology, Medical University of Vienna, Vienna, Austria
| | - Fabrice Prin
- The Francis Crick Institute, London, United Kingdom
| | | | | | | | | | | | - Wolfgang J. Weninger
- Division of Anatomy, Center for Anatomy and Cell Biology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
13
|
Devine J, Vidal-García M, Liu W, Neves A, Lo Vercio LD, Green RM, Richbourg HA, Marchini M, Unger CM, Nickle AC, Radford B, Young NM, Gonzalez PN, Schuler RE, Bugacov A, Rolian C, Percival CJ, Williams T, Niswander L, Calof AL, Lander AD, Visel A, Jirik FR, Cheverud JM, Klein OD, Birnbaum RY, Merrill AE, Ackermann RR, Graf D, Hemberger M, Dean W, Forkert ND, Murray SA, Westerberg H, Marcucio RS, Hallgrímsson B. MusMorph, a database of standardized mouse morphology data for morphometric meta-analyses. Sci Data 2022; 9:230. [PMID: 35614082 PMCID: PMC9133120 DOI: 10.1038/s41597-022-01338-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 04/13/2022] [Indexed: 11/08/2022] Open
Abstract
Complex morphological traits are the product of many genes with transient or lasting developmental effects that interact in anatomical context. Mouse models are a key resource for disentangling such effects, because they offer myriad tools for manipulating the genome in a controlled environment. Unfortunately, phenotypic data are often obtained using laboratory-specific protocols, resulting in self-contained datasets that are difficult to relate to one another for larger scale analyses. To enable meta-analyses of morphological variation, particularly in the craniofacial complex and brain, we created MusMorph, a database of standardized mouse morphology data spanning numerous genotypes and developmental stages, including E10.5, E11.5, E14.5, E15.5, E18.5, and adulthood. To standardize data collection, we implemented an atlas-based phenotyping pipeline that combines techniques from image registration, deep learning, and morphometrics. Alongside stage-specific atlases, we provide aligned micro-computed tomography images, dense anatomical landmarks, and segmentations (if available) for each specimen (N = 10,056). Our workflow is open-source to encourage transparency and reproducible data collection. The MusMorph data and scripts are available on FaceBase ( www.facebase.org , https://doi.org/10.25550/3-HXMC ) and GitHub ( https://github.com/jaydevine/MusMorph ).
Collapse
Affiliation(s)
- Jay Devine
- Alberta Children's Hospital Research Institute, University of Calgary, 28 Oki Dr NW, Calgary, AB, T3B 6A8, Canada
- The McCaig Institute for Bone and Joint Health, University of Calgary, 3280 Hospital Dr NW, Calgary, AB, T2N 4Z6, Canada
- Department of Cell Biology and Anatomy, Cumming School of Medicine, University of Calgary, 3330 Hospital Dr NW, Calgary, AB, T2N 4N1, Canada
| | - Marta Vidal-García
- Alberta Children's Hospital Research Institute, University of Calgary, 28 Oki Dr NW, Calgary, AB, T3B 6A8, Canada
- The McCaig Institute for Bone and Joint Health, University of Calgary, 3280 Hospital Dr NW, Calgary, AB, T2N 4Z6, Canada
- Department of Cell Biology and Anatomy, Cumming School of Medicine, University of Calgary, 3330 Hospital Dr NW, Calgary, AB, T2N 4N1, Canada
| | - Wei Liu
- Alberta Children's Hospital Research Institute, University of Calgary, 28 Oki Dr NW, Calgary, AB, T3B 6A8, Canada
- The McCaig Institute for Bone and Joint Health, University of Calgary, 3280 Hospital Dr NW, Calgary, AB, T2N 4Z6, Canada
- Department of Cell Biology and Anatomy, Cumming School of Medicine, University of Calgary, 3330 Hospital Dr NW, Calgary, AB, T2N 4N1, Canada
| | - Amanda Neves
- Department of Biology, McMaster University, 1280 Main St W, Hamilton, ON, L8S 4L8, Canada
| | - Lucas D Lo Vercio
- Alberta Children's Hospital Research Institute, University of Calgary, 28 Oki Dr NW, Calgary, AB, T3B 6A8, Canada
- The McCaig Institute for Bone and Joint Health, University of Calgary, 3280 Hospital Dr NW, Calgary, AB, T2N 4Z6, Canada
- Department of Cell Biology and Anatomy, Cumming School of Medicine, University of Calgary, 3330 Hospital Dr NW, Calgary, AB, T2N 4N1, Canada
| | - Rebecca M Green
- School of Dental Medicine, University of Pittsburgh, 3501 Terrace St, Pittsburgh, PA, 15213, USA
| | - Heather A Richbourg
- Orthopaedic Trauma Institute, ZSFG, UCSF, 2550 23rd St, San Francisco, CA, 94110, USA
| | - Marta Marchini
- Alberta Children's Hospital Research Institute, University of Calgary, 28 Oki Dr NW, Calgary, AB, T3B 6A8, Canada
- The McCaig Institute for Bone and Joint Health, University of Calgary, 3280 Hospital Dr NW, Calgary, AB, T2N 4Z6, Canada
- Department of Cell Biology and Anatomy, Cumming School of Medicine, University of Calgary, 3330 Hospital Dr NW, Calgary, AB, T2N 4N1, Canada
| | - Colton M Unger
- The McCaig Institute for Bone and Joint Health, University of Calgary, 3280 Hospital Dr NW, Calgary, AB, T2N 4Z6, Canada
- Department of Biological Sciences, University of Calgary, 2500 University Dr NW, Calgary, AB, T2N 1N4, Canada
| | - Audrey C Nickle
- Center for Craniofacial Molecular Biology, Department of Biomedical Sciences, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, 2250 Alcazar St, Los Angeles, CA, 90033, USA
- Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, 1975 Zonal Ave, Los Angeles, CA, 90033, USA
| | - Bethany Radford
- Alberta Children's Hospital Research Institute, University of Calgary, 28 Oki Dr NW, Calgary, AB, T3B 6A8, Canada
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, 3330 Hospital Dr NW, Calgary, AB, T2N 4N1, Canada
| | - Nathan M Young
- Orthopaedic Trauma Institute, ZSFG, UCSF, 2550 23rd St, San Francisco, CA, 94110, USA
| | - Paula N Gonzalez
- Institute for Studies in Neuroscience and Complex Systems (ENyS) CONICET, Av. Calchaquí, 5402, Florencio Varela, Buenos Aires, Argentina
| | - Robert E Schuler
- Information Sciences Institute, Viterbi School of Engineering, University of Southern California, 4676 Admiralty Way, Marina del Rey, CA, 90292, USA
| | - Alejandro Bugacov
- Information Sciences Institute, Viterbi School of Engineering, University of Southern California, 4676 Admiralty Way, Marina del Rey, CA, 90292, USA
| | - Campbell Rolian
- The McCaig Institute for Bone and Joint Health, University of Calgary, 3280 Hospital Dr NW, Calgary, AB, T2N 4Z6, Canada
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, 3330 Hospital Dr NW, Calgary, AB, T2N 4N1, Canada
| | - Christopher J Percival
- Department of Anthropology, Stony Brook University, 100 Nicolls Rd, Stony Brook, NY, 11794, USA
| | - Trevor Williams
- Department of Craniofacial Biology, University of Colorado Anschutz Medical Campus, 12801 East 17th Ave, Aurora, CO, 80045, USA
| | - Lee Niswander
- Department of Molecular, Cellular and Developmental Biology, University of Colorado Boulder, Boulder, CO, 80309, USA
| | - Anne L Calof
- Department of Anatomy and Neurobiology, University of California, Irvine, Irvine, CA, 92697, USA
- Center for Complex Biological Systems, University of California, Irvine, Irvine, CA, 92697, USA
| | - Arthur D Lander
- Center for Complex Biological Systems, University of California, Irvine, Irvine, CA, 92697, USA
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, 92697, USA
| | - Axel Visel
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, 1 Cyclotron Rd, Berkeley, CA, 94720, USA
- U.S. Department of Energy Joint Genome Institute, Lawrence Berkeley National Laboratory, 1 Cyclotron Rd, Berkeley, CA, 94720, USA
- School of Natural Sciences, University of California, Merced, 5200 Lake Rd, Merced, CA, 95343, USA
| | - Frank R Jirik
- Alberta Children's Hospital Research Institute, University of Calgary, 28 Oki Dr NW, Calgary, AB, T3B 6A8, Canada
- The McCaig Institute for Bone and Joint Health, University of Calgary, 3280 Hospital Dr NW, Calgary, AB, T2N 4Z6, Canada
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, 3330 Hospital Dr NW, Calgary, AB, T2N 4N1, Canada
| | - James M Cheverud
- Department of Biology, Loyola University Chicago, 1032 W Sheridan Rd, Chicago, IL, 60660, USA
| | - Ophir D Klein
- Department of Orofacial Sciences and Program in Craniofacial Biology, University of California, San Francisco, 513 Parnassus Ave, San Francisco, CA, 94143, USA
- Department of Pediatrics and Institute for Human Genetics, University of California, San Francisco, 513 Parnassus Ave, San Francisco, CA, 94143, USA
- Department of Pediatrics, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA, 90048, USA
| | - Ramon Y Birnbaum
- Department of Life Sciences, Faculty of Natural Sciences, The Ben-Gurion University of the Negev, David Ben Gurion Blvd 1, Be'er Sheva, Israel
| | - Amy E Merrill
- Center for Craniofacial Molecular Biology, Department of Biomedical Sciences, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, 2250 Alcazar St, Los Angeles, CA, 90033, USA
- Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, 1975 Zonal Ave, Los Angeles, CA, 90033, USA
| | - Rebecca R Ackermann
- Department of Archaeology, University of Cape Town, Rondebosch, Cape Town, 7700, South Africa
- Human Evolution Research Institute, University of Cape Town, Rondebosch, Cape Town, 7700, South Africa
| | - Daniel Graf
- School of Dentistry, Faculty of Medicine and Dentistry, University of Alberta, 116 St. and 85 Ave, Edmonton, AB, T6G 2R3, Canada
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, 116 St. and 85 Ave, Edmonton, AB, T6G 2R3, Canada
| | - Myriam Hemberger
- Alberta Children's Hospital Research Institute, University of Calgary, 28 Oki Dr NW, Calgary, AB, T3B 6A8, Canada
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, 3330 Hospital Dr NW, Calgary, AB, T2N 4N1, Canada
| | - Wendy Dean
- Alberta Children's Hospital Research Institute, University of Calgary, 28 Oki Dr NW, Calgary, AB, T3B 6A8, Canada
- Department of Cell Biology and Anatomy, Cumming School of Medicine, University of Calgary, 3330 Hospital Dr NW, Calgary, AB, T2N 4N1, Canada
| | - Nils D Forkert
- Alberta Children's Hospital Research Institute, University of Calgary, 28 Oki Dr NW, Calgary, AB, T3B 6A8, Canada
- Department of Radiology, Cumming School of Medicine, University of Calgary, 3330 Hospital Dr NW, Calgary, AB, T2N 4N1, Canada
| | | | - Henrik Westerberg
- Department of Bioimaging Informatics, MRC Harwell Institute, Oxfordshire, OX11 0RD, UK
| | - Ralph S Marcucio
- Orthopaedic Trauma Institute, ZSFG, UCSF, 2550 23rd St, San Francisco, CA, 94110, USA
| | - Benedikt Hallgrímsson
- Alberta Children's Hospital Research Institute, University of Calgary, 28 Oki Dr NW, Calgary, AB, T3B 6A8, Canada.
- The McCaig Institute for Bone and Joint Health, University of Calgary, 3280 Hospital Dr NW, Calgary, AB, T2N 4Z6, Canada.
- Department of Cell Biology and Anatomy, Cumming School of Medicine, University of Calgary, 3330 Hospital Dr NW, Calgary, AB, T2N 4N1, Canada.
| |
Collapse
|
14
|
Salpeter EM, Leonard BC, Lopez AJ, Murphy CJ, Thomasy S, Imai DM, Grimsrud K, Lloyd KCK, Yan J, Sanchez Russo R, Shankar SP, Moshiri A. Retinal degeneration in mice and humans with neuronal ceroid lipofuscinosis type 8. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:1274. [PMID: 34532411 PMCID: PMC8421982 DOI: 10.21037/atm-20-4739] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 03/19/2021] [Indexed: 12/14/2022]
Abstract
Background Ceroid lipofuscinosis type 8 belongs to a heterogenous group of vision and life-threatening neurodegenerative diseases, neuronal ceroid lipofuscinosis (NCL). Effective therapy is limited to a single drug for treatment of ceroid lipofuscinosis type 2, necessitating animal disease models to facilitate further therapeutic development. Murine models are advantageous for therapeutic development due to easy genetic manipulation and rapid breeding, however appropriate genetic models need to be identified and characterized before being used for therapy testing. To date, murine models of ocular disease associated with ceroid lipofuscinosis type 8 have only been characterized in motor neuron degeneration mice. Methods Cln8−/− mice were produced by CRISPR/Cas9 genome editing through the International Mouse Phenotyping Consortium. Ophthalmic examination, optical coherence tomography, electroretinography, and ocular histology was performed on Cln8−/− mice and controls at 16 weeks of age. Quantification of all retinal layers, retinal pigmented epithelium, and the choriocapillaris was performed using images acquired with ocular coherence tomography and planimetry of histologic sections. Necropsy was performed to investigate concurrent systemic abnormalities. Clinical correlation with human patients with CLN8-associated retinopathy is provided. Results Retinal degeneration characterized by retinal pigment epithelium mottling, scattered drusen, and retinal vascular attenuation was noted in all Cln8−/− mice. Loss of inner and outer photoreceptor segment demarcation was noted on optical coherence tomography, with significant thinning of the whole retina (P=1e-9), outer nuclear layer (P=1e-9), and combined photoreceptor segments (P=1e-9). A global reduction in scotopic and photopic electroretinographic waveforms was noted in all Cln8−/− mice. Slight thickening of the inner plexiform layer (P=0.02) and inner nuclear layer (P=0.004), with significant thinning of the whole retina (P=0.03), outer nuclear layer (P=0.01), and outer photoreceptor segments (P=0.001) was appreciated on histologic sections. Scattered lipid vacuoles were noted in splenic red pulp of all Cln8−/− mice, though no gross systemic abnormalities were detected on necropsy. Retinal findings are consistent with those seen in patients with ceroid lipofuscinosis type 8. Conclusions This study provides detailed clinical characterization of retinopathy in adult Cln8−/− mice. Findings suggest that Cln8−/− mice may provide a useful murine model for development of novel therapeutics needed for treating ocular disease in patients with ceroid lipofuscinosis type 8.
Collapse
Affiliation(s)
- Elyse M Salpeter
- William R. Pritchard Veterinary Medical Teaching Hospital, School of Veterinary Medicine, University of California, Davis, Davis, California, USA
| | - Brian C Leonard
- William R. Pritchard Veterinary Medical Teaching Hospital, School of Veterinary Medicine, University of California, Davis, Davis, California, USA
| | - Antonio J Lopez
- Department of Ophthalmology & Vision Science, School of Medicine, University of California, Davis, Sacramento, California, USA
| | - Christopher J Murphy
- William R. Pritchard Veterinary Medical Teaching Hospital, School of Veterinary Medicine, University of California, Davis, Davis, California, USA.,Department of Ophthalmology & Vision Science, School of Medicine, University of California, Davis, Sacramento, California, USA
| | - Sara Thomasy
- William R. Pritchard Veterinary Medical Teaching Hospital, School of Veterinary Medicine, University of California, Davis, Davis, California, USA.,Department of Ophthalmology & Vision Science, School of Medicine, University of California, Davis, Sacramento, California, USA
| | - Denise M Imai
- Comparative Pathology Laboratory, School of Veterinary Medicine, UC Davis, Davis, California, USA
| | - Kristin Grimsrud
- Mouse Biology Program, University of California, Davis, Davis, California, USA.,Department of Pathology and Laboratory Medicine, School of Medicine, University of California, Davis, Sacramento, California, USA
| | - K C Kent Lloyd
- Mouse Biology Program, University of California, Davis, Davis, California, USA.,Department of Surgery, School of Medicine, University of California, Davis, Sacramento, California, USA
| | - Jiong Yan
- Department of Ophthalmology, Emory University, Atlanta, Georgia, USA
| | | | - Suma P Shankar
- Department of Pediatrics & Department of Ophthalmology & Vision Science, School of Medicine, University of California, Davis, Sacramento, California, USA
| | - Ala Moshiri
- Department of Ophthalmology & Vision Science, School of Medicine, University of California, Davis, Sacramento, California, USA
| |
Collapse
|
15
|
Singh R, Ha SE, Wei L, Jin B, Zogg H, Poudrier SM, Jorgensen BG, Park C, Ronkon CF, Bartlett A, Cho S, Morales A, Chung YH, Lee MY, Park JK, Gottfried-Blackmore A, Nguyen L, Sanders KM, Ro S. miR-10b-5p Rescues Diabetes and Gastrointestinal Dysmotility. Gastroenterology 2021; 160:1662-1678.e18. [PMID: 33421511 PMCID: PMC8532043 DOI: 10.1053/j.gastro.2020.12.062] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 12/18/2020] [Accepted: 12/26/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND & AIMS Interstitial cells of Cajal (ICCs) and pancreatic β cells require receptor tyrosine kinase (KIT) to develop and function properly. Degeneration of ICCs is linked to diabetic gastroparesis. The mechanisms linking diabetes and gastroparesis are unclear, but may involve microRNA (miRNA)-mediated post-transcriptional gene silencing in KIT+ cells. METHODS We performed miRNA-sequencing analysis from isolated ICCs in diabetic mice and plasma from patients with idiopathic and diabetic gastroparesis. miR-10b-5p target genes were identified and validated in mouse and human cell lines. For loss-of-function studies, we used KIT+ cell-restricted mir-10b knockout mice and KIT+ cell depletion mice. For gain-of-function studies, a synthetic miR-10b-5p mimic was injected in multiple diabetic mouse models. We compared the efficacy of miR-10b-5p mimic treatment vs antidiabetic and prokinetic medicines. RESULTS miR-10b-5p is highly expressed in ICCs from healthy mice, but drastically depleted in ICCs from diabetic mice. A conditional knockout of mir-10b in KIT+ cells or depletion of KIT+ cells in mice leads to degeneration of β cells and ICCs, resulting in diabetes and gastroparesis. miR-10b-5p targets the transcription factor Krüppel-like factor 11 (KLF11), which negatively regulates KIT expression. The miR-10b-5p mimic or Klf11 small interfering RNAs injected into mir-10b knockout mice, diet-induced diabetic mice, and TALLYHO polygenic diabetic mice rescue the diabetes and gastroparesis phenotype for an extended period of time. Furthermore, the miR-10b-5p mimic is more effective in improving glucose homoeostasis and gastrointestinal motility compared with common antidiabetic and prokinetic medications. CONCLUSIONS miR-10b-5p is a key regulator in diabetes and gastrointestinal dysmotility via the KLF11-KIT pathway. Restoration of miR-10b-5p may provide therapeutic benefits for these disorders.
Collapse
Affiliation(s)
- Rajan Singh
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada, Reno, NV, 89557, USA
| | - Se Eun Ha
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada, Reno, NV, 89557, USA
| | - Lai Wei
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada, Reno, NV, 89557, USA
| | - Byungchang Jin
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada, Reno, NV, 89557, USA
| | - Hannah Zogg
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada, Reno, NV, 89557, USA
| | - Sandra M. Poudrier
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada, Reno, NV, 89557, USA
| | - Brian G. Jorgensen
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada, Reno, NV, 89557, USA
| | - Chanjae Park
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada, Reno, NV, 89557, USA
| | - Charles F Ronkon
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada, Reno, NV, 89557, USA
| | - Allison Bartlett
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada, Reno, NV, 89557, USA
| | - Sung Cho
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada, Reno, NV, 89557, USA
| | - Addison Morales
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada, Reno, NV, 89557, USA
| | - Yu Heon Chung
- Division of Biological Sciences, Wonkwang University, Iksan, Chonbuk, Korea
| | - Moon Young Lee
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada, Reno, NV, 89557, USA,Department of Physiology, Wonkwang Digestive Disease Research Institute and Institute of Wonkwang Medical Science, School of Medicine, Wonkwang University, Iksan, Chonbuk, Korea
| | - Jong Kun Park
- Division of Biological Sciences, Wonkwang University, Iksan, Chonbuk, Korea
| | - Andrés Gottfried-Blackmore
- Division of Gastroenterology & Hepatology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Linda Nguyen
- Division of Gastroenterology & Hepatology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Kenton M. Sanders
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada, Reno, NV, 89557, USA
| | - Seungil Ro
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada, Reno, Nevada.
| |
Collapse
|
16
|
Geuther BQ, Peer A, He H, Sabnis G, Philip VM, Kumar V. Action detection using a neural network elucidates the genetics of mouse grooming behavior. eLife 2021; 10:e63207. [PMID: 33729153 PMCID: PMC8043749 DOI: 10.7554/elife.63207] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 03/05/2021] [Indexed: 12/25/2022] Open
Abstract
Automated detection of complex animal behaviors remains a challenging problem in neuroscience, particularly for behaviors that consist of disparate sequential motions. Grooming is a prototypical stereotyped behavior that is often used as an endophenotype in psychiatric genetics. Here, we used mouse grooming behavior as an example and developed a general purpose neural network architecture capable of dynamic action detection at human observer-level performance and operating across dozens of mouse strains with high visual diversity. We provide insights into the amount of human annotated training data that are needed to achieve such performance. We surveyed grooming behavior in the open field in 2457 mice across 62 strains, determined its heritable components, conducted GWAS to outline its genetic architecture, and performed PheWAS to link human psychiatric traits through shared underlying genetics. Our general machine learning solution that automatically classifies complex behaviors in large datasets will facilitate systematic studies of behavioral mechanisms.
Collapse
Affiliation(s)
| | - Asaf Peer
- The Jackson LaboratoryBar HarborUnited States
| | - Hao He
- The Jackson LaboratoryBar HarborUnited States
| | | | | | - Vivek Kumar
- The Jackson LaboratoryBar HarborUnited States
| |
Collapse
|
17
|
Sethi S, Vorontsov IE, Kulakovskiy IV, Greenaway S, Williams J, Makeev VJ, Brown SDM, Simon MM, Mallon AM. A holistic view of mouse enhancer architectures reveals analogous pleiotropic effects and correlation with human disease. BMC Genomics 2020; 21:754. [PMID: 33138777 PMCID: PMC7607678 DOI: 10.1186/s12864-020-07109-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 09/29/2020] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Efforts to elucidate the function of enhancers in vivo are underway but their vast numbers alongside differing enhancer architectures make it difficult to determine their impact on gene activity. By systematically annotating multiple mouse tissues with super- and typical-enhancers, we have explored their relationship with gene function and phenotype. RESULTS Though super-enhancers drive high total- and tissue-specific expression of their associated genes, we find that typical-enhancers also contribute heavily to the tissue-specific expression landscape on account of their large numbers in the genome. Unexpectedly, we demonstrate that both enhancer types are preferentially associated with relevant 'tissue-type' phenotypes and exhibit no difference in phenotype effect size or pleiotropy. Modelling regulatory data alongside molecular data, we built a predictive model to infer gene-phenotype associations and use this model to predict potentially novel disease-associated genes. CONCLUSION Overall our findings reveal that differing enhancer architectures have a similar impact on mammalian phenotypes whilst harbouring differing cellular and expression effects. Together, our results systematically characterise enhancers with predicted phenotypic traits endorsing the role for both types of enhancers in human disease and disorders.
Collapse
Affiliation(s)
- Siddharth Sethi
- Mammalian Genetics Unit, MRC Harwell Institute, Oxfordshire, OX11 0RD, UK
| | - Ilya E Vorontsov
- Vavilov Institute of General Genetics, Russian Academy of Sciences, Gubkina 3, Moscow, 119991, Russia
- Institute of Protein Research, Russian Academy of Sciences, Institutskaya 4, Pushchino, Moscow Region, 142290, Russia
| | - Ivan V Kulakovskiy
- Vavilov Institute of General Genetics, Russian Academy of Sciences, Gubkina 3, Moscow, 119991, Russia
- Institute of Protein Research, Russian Academy of Sciences, Institutskaya 4, Pushchino, Moscow Region, 142290, Russia
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilova 32, Moscow, 119991, Russia
| | - Simon Greenaway
- Mammalian Genetics Unit, MRC Harwell Institute, Oxfordshire, OX11 0RD, UK
| | - John Williams
- Mammalian Genetics Unit, MRC Harwell Institute, Oxfordshire, OX11 0RD, UK
- Institute of Translational Medicine, University Hospitals Birmingham NHS Foundation Trust, Birmingham, B15 2TH, UK
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - Vsevolod J Makeev
- Vavilov Institute of General Genetics, Russian Academy of Sciences, Gubkina 3, Moscow, 119991, Russia
- Institute of Protein Research, Russian Academy of Sciences, Institutskaya 4, Pushchino, Moscow Region, 142290, Russia
- Moscow Institute of Physics and Technology, 9 Institutskiy per., Dolgoprudny, Moscow Region, 141700, Russia
| | - Steve D M Brown
- Mammalian Genetics Unit, MRC Harwell Institute, Oxfordshire, OX11 0RD, UK
| | - Michelle M Simon
- Mammalian Genetics Unit, MRC Harwell Institute, Oxfordshire, OX11 0RD, UK.
| | - Ann-Marie Mallon
- Mammalian Genetics Unit, MRC Harwell Institute, Oxfordshire, OX11 0RD, UK.
| |
Collapse
|
18
|
Oh S, Boo K, Kim J, Baek SA, Jeon Y, You J, Lee H, Choi HJ, Park D, Lee JM, Baek SH. The chromatin-binding protein PHF6 functions as an E3 ubiquitin ligase of H2BK120 via H2BK12Ac recognition for activation of trophectodermal genes. Nucleic Acids Res 2020; 48:9037-9052. [PMID: 32735658 PMCID: PMC7498345 DOI: 10.1093/nar/gkaa626] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 07/07/2020] [Accepted: 07/14/2020] [Indexed: 12/19/2022] Open
Abstract
Epigenetic regulation is important for establishing lineage-specific gene expression during early development. Although signaling pathways have been well-studied for regulation of trophectoderm reprogramming, epigenetic regulation of trophectodermal genes with histone modification dynamics have been poorly understood. Here, we identify that plant homeodomain finger protein 6 (PHF6) is a key epigenetic regulator for activation of trophectodermal genes using RNA-sequencing and ChIP assays. PHF6 acts as an E3 ubiquitin ligase for ubiquitination of H2BK120 (H2BK120ub) via its extended plant homeodomain 1 (PHD1), while the extended PHD2 of PHF6 recognizes acetylation of H2BK12 (H2BK12Ac). Intriguingly, the recognition of H2BK12Ac by PHF6 is important for exerting its E3 ubiquitin ligase activity for H2BK120ub. Together, our data provide evidence that PHF6 is crucial for epigenetic regulation of trophectodermal gene expression by linking H2BK12Ac to H2BK120ub modification.
Collapse
Affiliation(s)
- Sungryong Oh
- Creative Research Initiatives Center for Epigenetic Code and Diseases, Department of Biological Sciences, Seoul National University, Seoul 08826, South Korea
| | - Kyungjin Boo
- Creative Research Initiatives Center for Epigenetic Code and Diseases, Department of Biological Sciences, Seoul National University, Seoul 08826, South Korea
| | - Jaebeom Kim
- Creative Research Initiatives Center for Epigenetic Code and Diseases, Department of Biological Sciences, Seoul National University, Seoul 08826, South Korea
| | - Seon Ah Baek
- Creative Research Initiatives Center for Epigenetic Code and Diseases, Department of Biological Sciences, Seoul National University, Seoul 08826, South Korea
| | - Yoon Jeon
- Graduate School of Cancer Science and Policy, Research Institute, National Cancer Center, Goyang 10408, South Korea
| | - Junghyun You
- Department of Biological Sciences, Seoul National University, Seoul 08826, South Korea
| | - Ho Lee
- Graduate School of Cancer Science and Policy, Research Institute, National Cancer Center, Goyang 10408, South Korea
| | - Hee-Jung Choi
- Department of Biological Sciences, Seoul National University, Seoul 08826, South Korea
| | - Daechan Park
- Department of Biological Sciences, College of Natural Sciences, Ajou University, Suwon 16499, South Korea
| | - Ji Min Lee
- Department of Molecular Bioscience, College of Biomedical Sciences, Kangwon National University, Chuncheon 24341, South Korea
| | - Sung Hee Baek
- Creative Research Initiatives Center for Epigenetic Code and Diseases, Department of Biological Sciences, Seoul National University, Seoul 08826, South Korea
| |
Collapse
|
19
|
Abstract
Sleep is a fundamental property conserved across species. The homeostatic induction of sleep indicates the presence of a mechanism that is progressively activated by the awake state and that induces sleep. Several lines of evidence support that such function, namely, sleep need, lies in the neuronal assemblies rather than specific brain regions and circuits. However, the molecular mechanism underlying the dynamics of sleep need is still unclear. This review aims to summarize recent studies mainly in rodents indicating that protein phosphorylation, especially at the synapses, could be the molecular entity associated with sleep need. Genetic studies in rodents have identified a set of kinases that promote sleep. The activity of sleep-promoting kinases appears to be elevated during the awake phase and in sleep deprivation. Furthermore, the proteomic analysis demonstrated that the phosphorylation status of synaptic protein is controlled by the sleep-wake cycle. Therefore, a plausible scenario may be that the awake-dependent activation of kinases modifies the phosphorylation status of synaptic proteins to promote sleep. We also discuss the possible importance of multisite phosphorylation on macromolecular protein complexes to achieve the slow dynamics and physiological functions of sleep in mammals.
Collapse
Affiliation(s)
- Koji L Ode
- Department of Systems Pharmacology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hiroki R Ueda
- Department of Systems Pharmacology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Laboratory for Synthetic Biology, RIKEN Center for Biosystems Dynamics, Osaka, Japan
| |
Collapse
|
20
|
Hosur V, Low BE, Li D, Stafford GA, Kohar V, Shultz LD, Wiles MV. Genes adapt to outsmart gene-targeting strategies in mutant mouse strains by skipping exons to reinitiate transcription and translation. Genome Biol 2020; 21:168. [PMID: 32646486 PMCID: PMC7350591 DOI: 10.1186/s13059-020-02086-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 06/29/2020] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Gene disruption in mouse embryonic stem cells or zygotes is a conventional genetics approach to identify gene function in vivo. However, because different gene disruption strategies use different mechanisms to disrupt genes, the strategies can result in diverse phenotypes in the resulting mouse model. To determine whether different gene disruption strategies affect the phenotype of resulting mutant mice, we characterized Rhbdf1 mouse mutant strains generated by three commonly used strategies-definitive-null, targeted knockout (KO)-first, and CRISPR/Cas9. RESULTS We find that Rhbdf1 responds differently to distinct KO strategies, for example, by skipping exons and reinitiating translation to potentially yield gain-of-function alleles rather than the expected null or severe hypomorphic alleles. Our analysis also revealed that at least 4% of mice generated using the KO-first strategy show conflicting phenotypes. CONCLUSIONS Exon skipping is a widespread phenomenon occurring across the genome. These findings have significant implications for the application of genome editing in both basic research and clinical practice.
Collapse
Affiliation(s)
- Vishnu Hosur
- The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME USA
| | - Benjamin E. Low
- The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME USA
| | - Daniel Li
- Arthritis and Tissue Degeneration Program, Hospital for Special Surgery at Weill Cornell Medicine, New York, NY 10021 USA
| | | | - Vivek Kohar
- The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME USA
| | | | - Michael V. Wiles
- The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME USA
| |
Collapse
|
21
|
Marshall KM, Kanczler JM, Oreffo ROC. Evolving applications of the egg: chorioallantoic membrane assay and ex vivo organotypic culture of materials for bone tissue engineering. J Tissue Eng 2020; 11:2041731420942734. [PMID: 33194169 PMCID: PMC7594486 DOI: 10.1177/2041731420942734] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 06/26/2020] [Indexed: 01/03/2023] Open
Abstract
The chick chorioallantoic membrane model has been around for over a century, applied in angiogenic, oncology, dental and xenograft research. Despite its often perceived archaic, redolent history, the chorioallantoic membrane assay offers new and exciting opportunities for material and growth factor evaluation in bone tissue engineering. Currently, superior/improved experimental methodology for the chorioallantoic membrane assay are difficult to identify, given an absence of scientific consensus in defining experimental approaches, including timing of inoculation with materials and the analysis of results. In addition, critically, regulatory and welfare issues impact upon experimental designs. Given such disparate points, this review details recent research using the ex vivo chorioallantoic membrane assay and the ex vivo organotypic culture to advance the field of bone tissue engineering, and highlights potential areas of improvement for their application based on recent developments within our group and the tissue engineering field.
Collapse
Affiliation(s)
- Karen M Marshall
- Bone and Joint Research Group, Centre for Human
Development, Stem Cells and Regeneration, Institute of Developmental Sciences,
University of Southampton, Southampton, UK
| | - Janos M Kanczler
- Bone and Joint Research Group, Centre for Human
Development, Stem Cells and Regeneration, Institute of Developmental Sciences,
University of Southampton, Southampton, UK
| | - Richard OC Oreffo
- Bone and Joint Research Group, Centre for Human
Development, Stem Cells and Regeneration, Institute of Developmental Sciences,
University of Southampton, Southampton, UK
| |
Collapse
|
22
|
Zi M, Stafford N, Prehar S, Baudoin F, Oceandy D, Wang X, Bui T, Shaheen M, Neyses L, Cartwright EJ. Cardiac hypertrophy or failure? - A systematic evaluation of the transverse aortic constriction model in C57BL/6NTac and C57BL/6J substrains. Curr Res Physiol 2019; 1:1-10. [PMID: 32699840 PMCID: PMC7357793 DOI: 10.1016/j.crphys.2019.10.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 10/20/2019] [Accepted: 10/21/2019] [Indexed: 12/13/2022] Open
Abstract
Background The mouse model of transverse aortic constriction (TAC) has been widely used as a cardiac stress in the investigation of the molecular mechanisms of cardiac hypertrophy. Recently, the International Knockout Mouse Consortium has selected the C57BL/6NTac (BL/6N) mouse strain to generate null alleles for all mouse genes; however, a range of genetic and cardiac phenotypic differences have been reported between this substrain and the commonly used C57BL/6J (BL/6J) substrain. It has been reported by Garcia-Menendez and colleagues that 12-week C57BL/6NTac mice are susceptible to heart failure but little is known about the cardiac remodeling in this substrain as cardiac function progresses from compensation to decompensation. Methods BL/6J and BL/6N mice were subjected to pressure overload via TAC. The impact of both age and duration of cardiac pressure overload induced by TAC on cardiac remodelling were systematically assessed. Results Our data showed that BL/6N mice developed eccentric hypertrophy with age- and time-dependent deterioration in cardiac function, accompanied by considerable interstitial fibrosis. In contrast, BL/6J mice were more resilient to TAC-induced cardiac stress and developed variable cardiac phenotypes independent of age and the duration of pressure overload. This was likely due to the greater variability in pre-TAC aortic arch dimension as measured by echocardiography. In addition to increased expression of brain natriuretic peptide and collagen gene type 1 and 3, BL/6N mice also had greater angiotensin II type 2 receptor (AT2R) gene expression than BL/6J counterparts at baseline and after 2-weeks TAC, which may contribute to the exacerbated interstitial fibrosis. Conclusions BL/6N and BL/6J mice have very different responses to TAC stimulation and these differences should be taken into consideration when using the substrains to investigate the mechanisms of hypertrophy and heart failure. The first study to show that adult BL/6NTac mice have an age-dependent cardiac response to pressure overload. The first study to show BL/6NTac mice have a time-dependent cardiac response to pressure overload. C57BL/6J mice have variable cardiac remodelling that positively correlates with the original size of the aorta.
Collapse
Affiliation(s)
- Min Zi
- Division of Cardiovascular Sciences, University of Manchester and Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Nicholas Stafford
- Division of Cardiovascular Sciences, University of Manchester and Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Sukhpal Prehar
- Division of Cardiovascular Sciences, University of Manchester and Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Florence Baudoin
- Division of Cardiovascular Sciences, University of Manchester and Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Delvac Oceandy
- Division of Cardiovascular Sciences, University of Manchester and Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Xin Wang
- Division of Cardiovascular Sciences, University of Manchester and Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Thuy Bui
- Division of Cardiovascular Sciences, University of Manchester and Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Mohamed Shaheen
- Division of Cardiovascular Sciences, University of Manchester and Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Ludwig Neyses
- Division of Cardiovascular Sciences, University of Manchester and Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Elizabeth J Cartwright
- Division of Cardiovascular Sciences, University of Manchester and Manchester Academic Health Science Centre, Manchester, United Kingdom
| |
Collapse
|
23
|
Elmore SA, Cardiff R, Cesta MF, Gkoutos GV, Hoehndorf R, Keenan CM, McKerlie C, Schofield PN, Sundberg JP, Ward JM. A Review of Current Standards and the Evolution of Histopathology Nomenclature for Laboratory Animals. ILAR J 2019; 59:29-39. [PMID: 30476141 DOI: 10.1093/ilar/ily005] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Revised: 05/04/2018] [Indexed: 12/14/2022] Open
Abstract
The need for international collaboration in rodent pathology has evolved since the 1970s and was initially driven by the new field of toxicologic pathology. First initiated by the World Health Organization's International Agency for Research on Cancer for rodents, it has evolved to include pathology of the major species (rats, mice, guinea pigs, nonhuman primates, pigs, dogs, fish, rabbits) used in medical research, safety assessment, and mouse pathology. The collaborative effort today is driven by the needs of the regulatory agencies in multiple countries, and by needs of research involving genetically engineered animals, for "basic" research and for more translational preclinical models of human disease. These efforts led to the establishment of an international rodent pathology nomenclature program. Since that time, multiple collaborations for standardization of laboratory animal pathology nomenclature and diagnostic criteria have been developed, and just a few are described herein. Recently, approaches to a nomenclature that is amenable to sophisticated computation have been made available and implemented for large-scale programs in functional genomics and aging. Most terminologies continue to evolve as the science of human and veterinary pathology continues to develop, but standardization and successful implementation remain critical for scientific communication now as ever in the history of veterinary nosology.
Collapse
Affiliation(s)
- Susan A Elmore
- Susan A. Elmore, MS, DVM, DCVP, DABT, FIATP, is NTP Pathologist and Staff Scientist at the National Toxicology Program, National Institute of Environmental Health Sciences in the Research Triangle Park, North Carolina. Robert D. Cardiff, MD, PhD, is Distinguished Professor of Pathology, Emeritus at the UCD Center for Comparative Medicine, University of California, and the Department of Pathology and Laboratory Medicine, School of Medicine, Davis, in Davis, California. Mark F. Cesta, DVM, PhD, DACVP, is NTP Pathologist and Staff Scientist, leading the effort for establishment of the online NTP Nonneoplastic Lesion Atlas at the National Toxicology Program, National Institute of Environmental Health Sciences in the Research Triangle Park, North Carolina. Georgios V. Gkoutos, PhD, DIC, is Professor of Clinical Bioinformatics at College of Medical and Dental Sciences, Institute of Cancer and Genomic Sciences Centre for Computational Biology, University of Birmingham in Birmingham, United Kingdom. Robert Hoehndorf, PhD, is Assistant Professor in Computer Science at the Computer, Electrical and Mathematical Sciences and Engineering Division, Computational Bioscience Research Center, King Abdullah University of Science and Technology in Thuwal, Kingdom of Saudi Arabia. Charlotte M. Keenan, VMD, DACVP, is a principle consultant at C.M. ToxPath Consulting in Doylestown, Pennsylvania, USA and leads the international STP effort for the publication of the harmonization of nomenclature and diagnostic criteria (INHAND) in toxicologic pathology. Colin McKerlie, DVM, DVSc, MRCVS, is a senior associate scientist in the Translational Medicine Research Program at The Hospital for Sick Children and a Professor in the Department of Pathobiology & Laboratory Medicine in the Faculty of Medicine at the University of Toronto, Toronto, Ontario, Canada. Paul N. Schofield, MA DPhil, is the University Reader in Biomedical Informatics at the Department of Physiology, Development & Neuroscience, University of Cambridge in Cambridge, United Kingdom and is also an adjunct professor at The Jackson Laboratory in Bar Harbor, Maine. John P. Sundberg, DVM, PhD, DACVP, is a professor at The Jackson Laboratory in Bar Harbor, Maine. Jerrold M. Ward, DVM, PhD, DACVP, FIATP, is a special volunteer at the National Cancer Institute, National Institutes of Health in Bethesda, MD and is also Adjunct Faculty at The Jackson Laboratory in Bar Harbor, Maine
| | - Robert Cardiff
- Susan A. Elmore, MS, DVM, DCVP, DABT, FIATP, is NTP Pathologist and Staff Scientist at the National Toxicology Program, National Institute of Environmental Health Sciences in the Research Triangle Park, North Carolina. Robert D. Cardiff, MD, PhD, is Distinguished Professor of Pathology, Emeritus at the UCD Center for Comparative Medicine, University of California, and the Department of Pathology and Laboratory Medicine, School of Medicine, Davis, in Davis, California. Mark F. Cesta, DVM, PhD, DACVP, is NTP Pathologist and Staff Scientist, leading the effort for establishment of the online NTP Nonneoplastic Lesion Atlas at the National Toxicology Program, National Institute of Environmental Health Sciences in the Research Triangle Park, North Carolina. Georgios V. Gkoutos, PhD, DIC, is Professor of Clinical Bioinformatics at College of Medical and Dental Sciences, Institute of Cancer and Genomic Sciences Centre for Computational Biology, University of Birmingham in Birmingham, United Kingdom. Robert Hoehndorf, PhD, is Assistant Professor in Computer Science at the Computer, Electrical and Mathematical Sciences and Engineering Division, Computational Bioscience Research Center, King Abdullah University of Science and Technology in Thuwal, Kingdom of Saudi Arabia. Charlotte M. Keenan, VMD, DACVP, is a principle consultant at C.M. ToxPath Consulting in Doylestown, Pennsylvania, USA and leads the international STP effort for the publication of the harmonization of nomenclature and diagnostic criteria (INHAND) in toxicologic pathology. Colin McKerlie, DVM, DVSc, MRCVS, is a senior associate scientist in the Translational Medicine Research Program at The Hospital for Sick Children and a Professor in the Department of Pathobiology & Laboratory Medicine in the Faculty of Medicine at the University of Toronto, Toronto, Ontario, Canada. Paul N. Schofield, MA DPhil, is the University Reader in Biomedical Informatics at the Department of Physiology, Development & Neuroscience, University of Cambridge in Cambridge, United Kingdom and is also an adjunct professor at The Jackson Laboratory in Bar Harbor, Maine. John P. Sundberg, DVM, PhD, DACVP, is a professor at The Jackson Laboratory in Bar Harbor, Maine. Jerrold M. Ward, DVM, PhD, DACVP, FIATP, is a special volunteer at the National Cancer Institute, National Institutes of Health in Bethesda, MD and is also Adjunct Faculty at The Jackson Laboratory in Bar Harbor, Maine
| | - Mark F Cesta
- Susan A. Elmore, MS, DVM, DCVP, DABT, FIATP, is NTP Pathologist and Staff Scientist at the National Toxicology Program, National Institute of Environmental Health Sciences in the Research Triangle Park, North Carolina. Robert D. Cardiff, MD, PhD, is Distinguished Professor of Pathology, Emeritus at the UCD Center for Comparative Medicine, University of California, and the Department of Pathology and Laboratory Medicine, School of Medicine, Davis, in Davis, California. Mark F. Cesta, DVM, PhD, DACVP, is NTP Pathologist and Staff Scientist, leading the effort for establishment of the online NTP Nonneoplastic Lesion Atlas at the National Toxicology Program, National Institute of Environmental Health Sciences in the Research Triangle Park, North Carolina. Georgios V. Gkoutos, PhD, DIC, is Professor of Clinical Bioinformatics at College of Medical and Dental Sciences, Institute of Cancer and Genomic Sciences Centre for Computational Biology, University of Birmingham in Birmingham, United Kingdom. Robert Hoehndorf, PhD, is Assistant Professor in Computer Science at the Computer, Electrical and Mathematical Sciences and Engineering Division, Computational Bioscience Research Center, King Abdullah University of Science and Technology in Thuwal, Kingdom of Saudi Arabia. Charlotte M. Keenan, VMD, DACVP, is a principle consultant at C.M. ToxPath Consulting in Doylestown, Pennsylvania, USA and leads the international STP effort for the publication of the harmonization of nomenclature and diagnostic criteria (INHAND) in toxicologic pathology. Colin McKerlie, DVM, DVSc, MRCVS, is a senior associate scientist in the Translational Medicine Research Program at The Hospital for Sick Children and a Professor in the Department of Pathobiology & Laboratory Medicine in the Faculty of Medicine at the University of Toronto, Toronto, Ontario, Canada. Paul N. Schofield, MA DPhil, is the University Reader in Biomedical Informatics at the Department of Physiology, Development & Neuroscience, University of Cambridge in Cambridge, United Kingdom and is also an adjunct professor at The Jackson Laboratory in Bar Harbor, Maine. John P. Sundberg, DVM, PhD, DACVP, is a professor at The Jackson Laboratory in Bar Harbor, Maine. Jerrold M. Ward, DVM, PhD, DACVP, FIATP, is a special volunteer at the National Cancer Institute, National Institutes of Health in Bethesda, MD and is also Adjunct Faculty at The Jackson Laboratory in Bar Harbor, Maine
| | - Georgios V Gkoutos
- Susan A. Elmore, MS, DVM, DCVP, DABT, FIATP, is NTP Pathologist and Staff Scientist at the National Toxicology Program, National Institute of Environmental Health Sciences in the Research Triangle Park, North Carolina. Robert D. Cardiff, MD, PhD, is Distinguished Professor of Pathology, Emeritus at the UCD Center for Comparative Medicine, University of California, and the Department of Pathology and Laboratory Medicine, School of Medicine, Davis, in Davis, California. Mark F. Cesta, DVM, PhD, DACVP, is NTP Pathologist and Staff Scientist, leading the effort for establishment of the online NTP Nonneoplastic Lesion Atlas at the National Toxicology Program, National Institute of Environmental Health Sciences in the Research Triangle Park, North Carolina. Georgios V. Gkoutos, PhD, DIC, is Professor of Clinical Bioinformatics at College of Medical and Dental Sciences, Institute of Cancer and Genomic Sciences Centre for Computational Biology, University of Birmingham in Birmingham, United Kingdom. Robert Hoehndorf, PhD, is Assistant Professor in Computer Science at the Computer, Electrical and Mathematical Sciences and Engineering Division, Computational Bioscience Research Center, King Abdullah University of Science and Technology in Thuwal, Kingdom of Saudi Arabia. Charlotte M. Keenan, VMD, DACVP, is a principle consultant at C.M. ToxPath Consulting in Doylestown, Pennsylvania, USA and leads the international STP effort for the publication of the harmonization of nomenclature and diagnostic criteria (INHAND) in toxicologic pathology. Colin McKerlie, DVM, DVSc, MRCVS, is a senior associate scientist in the Translational Medicine Research Program at The Hospital for Sick Children and a Professor in the Department of Pathobiology & Laboratory Medicine in the Faculty of Medicine at the University of Toronto, Toronto, Ontario, Canada. Paul N. Schofield, MA DPhil, is the University Reader in Biomedical Informatics at the Department of Physiology, Development & Neuroscience, University of Cambridge in Cambridge, United Kingdom and is also an adjunct professor at The Jackson Laboratory in Bar Harbor, Maine. John P. Sundberg, DVM, PhD, DACVP, is a professor at The Jackson Laboratory in Bar Harbor, Maine. Jerrold M. Ward, DVM, PhD, DACVP, FIATP, is a special volunteer at the National Cancer Institute, National Institutes of Health in Bethesda, MD and is also Adjunct Faculty at The Jackson Laboratory in Bar Harbor, Maine
| | - Robert Hoehndorf
- Susan A. Elmore, MS, DVM, DCVP, DABT, FIATP, is NTP Pathologist and Staff Scientist at the National Toxicology Program, National Institute of Environmental Health Sciences in the Research Triangle Park, North Carolina. Robert D. Cardiff, MD, PhD, is Distinguished Professor of Pathology, Emeritus at the UCD Center for Comparative Medicine, University of California, and the Department of Pathology and Laboratory Medicine, School of Medicine, Davis, in Davis, California. Mark F. Cesta, DVM, PhD, DACVP, is NTP Pathologist and Staff Scientist, leading the effort for establishment of the online NTP Nonneoplastic Lesion Atlas at the National Toxicology Program, National Institute of Environmental Health Sciences in the Research Triangle Park, North Carolina. Georgios V. Gkoutos, PhD, DIC, is Professor of Clinical Bioinformatics at College of Medical and Dental Sciences, Institute of Cancer and Genomic Sciences Centre for Computational Biology, University of Birmingham in Birmingham, United Kingdom. Robert Hoehndorf, PhD, is Assistant Professor in Computer Science at the Computer, Electrical and Mathematical Sciences and Engineering Division, Computational Bioscience Research Center, King Abdullah University of Science and Technology in Thuwal, Kingdom of Saudi Arabia. Charlotte M. Keenan, VMD, DACVP, is a principle consultant at C.M. ToxPath Consulting in Doylestown, Pennsylvania, USA and leads the international STP effort for the publication of the harmonization of nomenclature and diagnostic criteria (INHAND) in toxicologic pathology. Colin McKerlie, DVM, DVSc, MRCVS, is a senior associate scientist in the Translational Medicine Research Program at The Hospital for Sick Children and a Professor in the Department of Pathobiology & Laboratory Medicine in the Faculty of Medicine at the University of Toronto, Toronto, Ontario, Canada. Paul N. Schofield, MA DPhil, is the University Reader in Biomedical Informatics at the Department of Physiology, Development & Neuroscience, University of Cambridge in Cambridge, United Kingdom and is also an adjunct professor at The Jackson Laboratory in Bar Harbor, Maine. John P. Sundberg, DVM, PhD, DACVP, is a professor at The Jackson Laboratory in Bar Harbor, Maine. Jerrold M. Ward, DVM, PhD, DACVP, FIATP, is a special volunteer at the National Cancer Institute, National Institutes of Health in Bethesda, MD and is also Adjunct Faculty at The Jackson Laboratory in Bar Harbor, Maine
| | - Charlotte M Keenan
- Susan A. Elmore, MS, DVM, DCVP, DABT, FIATP, is NTP Pathologist and Staff Scientist at the National Toxicology Program, National Institute of Environmental Health Sciences in the Research Triangle Park, North Carolina. Robert D. Cardiff, MD, PhD, is Distinguished Professor of Pathology, Emeritus at the UCD Center for Comparative Medicine, University of California, and the Department of Pathology and Laboratory Medicine, School of Medicine, Davis, in Davis, California. Mark F. Cesta, DVM, PhD, DACVP, is NTP Pathologist and Staff Scientist, leading the effort for establishment of the online NTP Nonneoplastic Lesion Atlas at the National Toxicology Program, National Institute of Environmental Health Sciences in the Research Triangle Park, North Carolina. Georgios V. Gkoutos, PhD, DIC, is Professor of Clinical Bioinformatics at College of Medical and Dental Sciences, Institute of Cancer and Genomic Sciences Centre for Computational Biology, University of Birmingham in Birmingham, United Kingdom. Robert Hoehndorf, PhD, is Assistant Professor in Computer Science at the Computer, Electrical and Mathematical Sciences and Engineering Division, Computational Bioscience Research Center, King Abdullah University of Science and Technology in Thuwal, Kingdom of Saudi Arabia. Charlotte M. Keenan, VMD, DACVP, is a principle consultant at C.M. ToxPath Consulting in Doylestown, Pennsylvania, USA and leads the international STP effort for the publication of the harmonization of nomenclature and diagnostic criteria (INHAND) in toxicologic pathology. Colin McKerlie, DVM, DVSc, MRCVS, is a senior associate scientist in the Translational Medicine Research Program at The Hospital for Sick Children and a Professor in the Department of Pathobiology & Laboratory Medicine in the Faculty of Medicine at the University of Toronto, Toronto, Ontario, Canada. Paul N. Schofield, MA DPhil, is the University Reader in Biomedical Informatics at the Department of Physiology, Development & Neuroscience, University of Cambridge in Cambridge, United Kingdom and is also an adjunct professor at The Jackson Laboratory in Bar Harbor, Maine. John P. Sundberg, DVM, PhD, DACVP, is a professor at The Jackson Laboratory in Bar Harbor, Maine. Jerrold M. Ward, DVM, PhD, DACVP, FIATP, is a special volunteer at the National Cancer Institute, National Institutes of Health in Bethesda, MD and is also Adjunct Faculty at The Jackson Laboratory in Bar Harbor, Maine
| | - Colin McKerlie
- Susan A. Elmore, MS, DVM, DCVP, DABT, FIATP, is NTP Pathologist and Staff Scientist at the National Toxicology Program, National Institute of Environmental Health Sciences in the Research Triangle Park, North Carolina. Robert D. Cardiff, MD, PhD, is Distinguished Professor of Pathology, Emeritus at the UCD Center for Comparative Medicine, University of California, and the Department of Pathology and Laboratory Medicine, School of Medicine, Davis, in Davis, California. Mark F. Cesta, DVM, PhD, DACVP, is NTP Pathologist and Staff Scientist, leading the effort for establishment of the online NTP Nonneoplastic Lesion Atlas at the National Toxicology Program, National Institute of Environmental Health Sciences in the Research Triangle Park, North Carolina. Georgios V. Gkoutos, PhD, DIC, is Professor of Clinical Bioinformatics at College of Medical and Dental Sciences, Institute of Cancer and Genomic Sciences Centre for Computational Biology, University of Birmingham in Birmingham, United Kingdom. Robert Hoehndorf, PhD, is Assistant Professor in Computer Science at the Computer, Electrical and Mathematical Sciences and Engineering Division, Computational Bioscience Research Center, King Abdullah University of Science and Technology in Thuwal, Kingdom of Saudi Arabia. Charlotte M. Keenan, VMD, DACVP, is a principle consultant at C.M. ToxPath Consulting in Doylestown, Pennsylvania, USA and leads the international STP effort for the publication of the harmonization of nomenclature and diagnostic criteria (INHAND) in toxicologic pathology. Colin McKerlie, DVM, DVSc, MRCVS, is a senior associate scientist in the Translational Medicine Research Program at The Hospital for Sick Children and a Professor in the Department of Pathobiology & Laboratory Medicine in the Faculty of Medicine at the University of Toronto, Toronto, Ontario, Canada. Paul N. Schofield, MA DPhil, is the University Reader in Biomedical Informatics at the Department of Physiology, Development & Neuroscience, University of Cambridge in Cambridge, United Kingdom and is also an adjunct professor at The Jackson Laboratory in Bar Harbor, Maine. John P. Sundberg, DVM, PhD, DACVP, is a professor at The Jackson Laboratory in Bar Harbor, Maine. Jerrold M. Ward, DVM, PhD, DACVP, FIATP, is a special volunteer at the National Cancer Institute, National Institutes of Health in Bethesda, MD and is also Adjunct Faculty at The Jackson Laboratory in Bar Harbor, Maine
| | - Paul N Schofield
- Susan A. Elmore, MS, DVM, DCVP, DABT, FIATP, is NTP Pathologist and Staff Scientist at the National Toxicology Program, National Institute of Environmental Health Sciences in the Research Triangle Park, North Carolina. Robert D. Cardiff, MD, PhD, is Distinguished Professor of Pathology, Emeritus at the UCD Center for Comparative Medicine, University of California, and the Department of Pathology and Laboratory Medicine, School of Medicine, Davis, in Davis, California. Mark F. Cesta, DVM, PhD, DACVP, is NTP Pathologist and Staff Scientist, leading the effort for establishment of the online NTP Nonneoplastic Lesion Atlas at the National Toxicology Program, National Institute of Environmental Health Sciences in the Research Triangle Park, North Carolina. Georgios V. Gkoutos, PhD, DIC, is Professor of Clinical Bioinformatics at College of Medical and Dental Sciences, Institute of Cancer and Genomic Sciences Centre for Computational Biology, University of Birmingham in Birmingham, United Kingdom. Robert Hoehndorf, PhD, is Assistant Professor in Computer Science at the Computer, Electrical and Mathematical Sciences and Engineering Division, Computational Bioscience Research Center, King Abdullah University of Science and Technology in Thuwal, Kingdom of Saudi Arabia. Charlotte M. Keenan, VMD, DACVP, is a principle consultant at C.M. ToxPath Consulting in Doylestown, Pennsylvania, USA and leads the international STP effort for the publication of the harmonization of nomenclature and diagnostic criteria (INHAND) in toxicologic pathology. Colin McKerlie, DVM, DVSc, MRCVS, is a senior associate scientist in the Translational Medicine Research Program at The Hospital for Sick Children and a Professor in the Department of Pathobiology & Laboratory Medicine in the Faculty of Medicine at the University of Toronto, Toronto, Ontario, Canada. Paul N. Schofield, MA DPhil, is the University Reader in Biomedical Informatics at the Department of Physiology, Development & Neuroscience, University of Cambridge in Cambridge, United Kingdom and is also an adjunct professor at The Jackson Laboratory in Bar Harbor, Maine. John P. Sundberg, DVM, PhD, DACVP, is a professor at The Jackson Laboratory in Bar Harbor, Maine. Jerrold M. Ward, DVM, PhD, DACVP, FIATP, is a special volunteer at the National Cancer Institute, National Institutes of Health in Bethesda, MD and is also Adjunct Faculty at The Jackson Laboratory in Bar Harbor, Maine
| | - John P Sundberg
- Susan A. Elmore, MS, DVM, DCVP, DABT, FIATP, is NTP Pathologist and Staff Scientist at the National Toxicology Program, National Institute of Environmental Health Sciences in the Research Triangle Park, North Carolina. Robert D. Cardiff, MD, PhD, is Distinguished Professor of Pathology, Emeritus at the UCD Center for Comparative Medicine, University of California, and the Department of Pathology and Laboratory Medicine, School of Medicine, Davis, in Davis, California. Mark F. Cesta, DVM, PhD, DACVP, is NTP Pathologist and Staff Scientist, leading the effort for establishment of the online NTP Nonneoplastic Lesion Atlas at the National Toxicology Program, National Institute of Environmental Health Sciences in the Research Triangle Park, North Carolina. Georgios V. Gkoutos, PhD, DIC, is Professor of Clinical Bioinformatics at College of Medical and Dental Sciences, Institute of Cancer and Genomic Sciences Centre for Computational Biology, University of Birmingham in Birmingham, United Kingdom. Robert Hoehndorf, PhD, is Assistant Professor in Computer Science at the Computer, Electrical and Mathematical Sciences and Engineering Division, Computational Bioscience Research Center, King Abdullah University of Science and Technology in Thuwal, Kingdom of Saudi Arabia. Charlotte M. Keenan, VMD, DACVP, is a principle consultant at C.M. ToxPath Consulting in Doylestown, Pennsylvania, USA and leads the international STP effort for the publication of the harmonization of nomenclature and diagnostic criteria (INHAND) in toxicologic pathology. Colin McKerlie, DVM, DVSc, MRCVS, is a senior associate scientist in the Translational Medicine Research Program at The Hospital for Sick Children and a Professor in the Department of Pathobiology & Laboratory Medicine in the Faculty of Medicine at the University of Toronto, Toronto, Ontario, Canada. Paul N. Schofield, MA DPhil, is the University Reader in Biomedical Informatics at the Department of Physiology, Development & Neuroscience, University of Cambridge in Cambridge, United Kingdom and is also an adjunct professor at The Jackson Laboratory in Bar Harbor, Maine. John P. Sundberg, DVM, PhD, DACVP, is a professor at The Jackson Laboratory in Bar Harbor, Maine. Jerrold M. Ward, DVM, PhD, DACVP, FIATP, is a special volunteer at the National Cancer Institute, National Institutes of Health in Bethesda, MD and is also Adjunct Faculty at The Jackson Laboratory in Bar Harbor, Maine
| | - Jerrold M Ward
- Susan A. Elmore, MS, DVM, DCVP, DABT, FIATP, is NTP Pathologist and Staff Scientist at the National Toxicology Program, National Institute of Environmental Health Sciences in the Research Triangle Park, North Carolina. Robert D. Cardiff, MD, PhD, is Distinguished Professor of Pathology, Emeritus at the UCD Center for Comparative Medicine, University of California, and the Department of Pathology and Laboratory Medicine, School of Medicine, Davis, in Davis, California. Mark F. Cesta, DVM, PhD, DACVP, is NTP Pathologist and Staff Scientist, leading the effort for establishment of the online NTP Nonneoplastic Lesion Atlas at the National Toxicology Program, National Institute of Environmental Health Sciences in the Research Triangle Park, North Carolina. Georgios V. Gkoutos, PhD, DIC, is Professor of Clinical Bioinformatics at College of Medical and Dental Sciences, Institute of Cancer and Genomic Sciences Centre for Computational Biology, University of Birmingham in Birmingham, United Kingdom. Robert Hoehndorf, PhD, is Assistant Professor in Computer Science at the Computer, Electrical and Mathematical Sciences and Engineering Division, Computational Bioscience Research Center, King Abdullah University of Science and Technology in Thuwal, Kingdom of Saudi Arabia. Charlotte M. Keenan, VMD, DACVP, is a principle consultant at C.M. ToxPath Consulting in Doylestown, Pennsylvania, USA and leads the international STP effort for the publication of the harmonization of nomenclature and diagnostic criteria (INHAND) in toxicologic pathology. Colin McKerlie, DVM, DVSc, MRCVS, is a senior associate scientist in the Translational Medicine Research Program at The Hospital for Sick Children and a Professor in the Department of Pathobiology & Laboratory Medicine in the Faculty of Medicine at the University of Toronto, Toronto, Ontario, Canada. Paul N. Schofield, MA DPhil, is the University Reader in Biomedical Informatics at the Department of Physiology, Development & Neuroscience, University of Cambridge in Cambridge, United Kingdom and is also an adjunct professor at The Jackson Laboratory in Bar Harbor, Maine. John P. Sundberg, DVM, PhD, DACVP, is a professor at The Jackson Laboratory in Bar Harbor, Maine. Jerrold M. Ward, DVM, PhD, DACVP, FIATP, is a special volunteer at the National Cancer Institute, National Institutes of Health in Bethesda, MD and is also Adjunct Faculty at The Jackson Laboratory in Bar Harbor, Maine
| |
Collapse
|
24
|
Rixen S, Havemeyer A, Tyl-Bielicka A, Pysniak K, Gajewska M, Kulecka M, Ostrowski J, Mikula M, Clement B. Mitochondrial amidoxime-reducing component 2 (MARC2) has a significant role in N-reductive activity and energy metabolism. J Biol Chem 2019; 294:17593-17602. [PMID: 31554661 DOI: 10.1074/jbc.ra119.007606] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 09/19/2019] [Indexed: 01/29/2023] Open
Abstract
The mitochondrial amidoxime-reducing component (MARC) is a mammalian molybdenum-containing enzyme. All annotated mammalian genomes harbor two MARC genes, MARC1 and MARC2, which share a high degree of sequence similarity. Both molybdoenzymes reduce a variety of N-hydroxylated compounds. Besides their role in N-reductive drug metabolism, only little is known about their physiological functions. In this study, we characterized an existing KO mouse model lacking the functional MARC2 gene and fed a high-fat diet and also performed in vivo and in vitro experiments to characterize reductase activity toward known MARC substrates. MARC2 KO significantly decreased reductase activity toward several N-oxygenated substrates, and for typical MARC substrates, only small residual reductive activity was still detectable in MARC2 KO mice. The residual detected reductase activity in MARC2 KO mice could be explained by MARC1 expression that was hardly unaffected by KO, and we found no evidence of significant activity of other reductase enzymes. These results clearly indicate that MARC2 is mainly responsible for N-reductive biotransformation in mice. Striking phenotypical features of MARC2 KO mice were lower body weight, increased body temperature, decreased levels of total cholesterol, and increased glucose levels, supporting previous findings that MARC2 affects energy pathways. Of note, the MARC2 KO mice were resistant to high-fat diet-induced obesity. We propose that the MARC2 KO mouse model could be a powerful tool for predicting MARC-mediated drug metabolism and further investigating MARC's roles in energy homeostasis.
Collapse
Affiliation(s)
- Sophia Rixen
- Department of Pharmaceutical and Medicinal Chemistry, Pharmaceutical Institute, Christian Albrechts University, 24118 Kiel, Germany
| | - Antje Havemeyer
- Department of Pharmaceutical and Medicinal Chemistry, Pharmaceutical Institute, Christian Albrechts University, 24118 Kiel, Germany
| | - Anita Tyl-Bielicka
- Department of Genetics, Maria Sklodowska-Curie Institute, Cancer Center, 02-781 Warsaw, Poland
| | - Kazimiera Pysniak
- Department of Genetics, Maria Sklodowska-Curie Institute, Cancer Center, 02-781 Warsaw, Poland
| | - Marta Gajewska
- Department of Genetics, Maria Sklodowska-Curie Institute, Cancer Center, 02-781 Warsaw, Poland
| | - Maria Kulecka
- Department of Gastroenterology, Hepatology, and Clinical Oncology, Centre of Postgraduate Medical Education, 02-781 Warsaw, Poland
| | - Jerzy Ostrowski
- Department of Genetics, Maria Sklodowska-Curie Institute, Cancer Center, 02-781 Warsaw, Poland.,Department of Gastroenterology, Hepatology, and Clinical Oncology, Centre of Postgraduate Medical Education, 02-781 Warsaw, Poland
| | - Michal Mikula
- Department of Genetics, Maria Sklodowska-Curie Institute, Cancer Center, 02-781 Warsaw, Poland
| | - Bernd Clement
- Department of Pharmaceutical and Medicinal Chemistry, Pharmaceutical Institute, Christian Albrechts University, 24118 Kiel, Germany
| |
Collapse
|
25
|
Meagan Littrell O, Stoeger C, Maier H, Fuchs H, Hrabě de Angelis M, Cassis LA, Gerhardt GA, Grondin R, Gailus-Durner V. Costs of Implementing Quality in Research Practice. Handb Exp Pharmacol 2019; 257:399-423. [PMID: 31541322 DOI: 10.1007/164_2019_294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2023]
Abstract
Using standardized guidelines in preclinical research has received increased interest in light of recent concerns about transparency in data reporting and apparent variation in data quality, as evidenced by irreproducibility of results. Although the costs associated with supporting quality through a quality management system are often obvious line items in laboratory budgets, the treatment of the costs associated with quality failure is often overlooked and difficult to quantify. Thus, general estimations of quality costs can be misleading and inaccurate, effectively undervaluing costs recovered by reducing quality defects. Here, we provide examples of quality costs in preclinical research and describe how we have addressed misconceptions of quality management implementation as only marginally beneficial and/or unduly burdensome. We provide two examples of implementing a quality management system (QMS) in preclinical experimental (animal) research environments - one in Europe, the German Mouse Clinic, having established ISO 9001 and the other in the United States, the University of Kentucky (UK), having established Good Laboratory Practice-compliant infrastructure. We present a summary of benefits to having an effective QMS, as may be useful in guiding discussions with funders or administrators to promote interest and investment in a QMS, which ultimately supports shared, mutually beneficial outcomes.
Collapse
Affiliation(s)
- O Meagan Littrell
- University of Kentucky Good Research Practice Resource Center and Department of Neuroscience, Lexington, KY, USA
| | - Claudia Stoeger
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Holger Maier
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Helmut Fuchs
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Martin Hrabě de Angelis
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany.,Experimental Genetics, School of Life Science Weihenstephan, Technische Universität München, Freising, Germany.,German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Lisa A Cassis
- University of Kentucky Office of the Vice President for Research and Department of Pharmacology and Nutritional Sciences, Lexington, KY, USA
| | - Greg A Gerhardt
- University of Kentucky Good Research Practice Resource Center and Department of Neuroscience, Lexington, KY, USA
| | - Richard Grondin
- University of Kentucky Good Research Practice Resource Center and Department of Neuroscience, Lexington, KY, USA
| | - Valérie Gailus-Durner
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany.
| |
Collapse
|
26
|
Moore BA, Flenniken AM, Clary D, Moshiri AS, Nutter LMJ, Berberovic Z, Owen C, Newbigging S, Adissu H, Eskandarian M, McKerlie C, Thomasy SM, Lloyd KCK, Murphy CJ, Moshiri A. Genome-wide screening of mouse knockouts reveals novel genes required for normal integumentary and oculocutaneous structure and function. Sci Rep 2019; 9:11211. [PMID: 31371754 PMCID: PMC6672016 DOI: 10.1038/s41598-019-47286-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 06/17/2019] [Indexed: 11/18/2022] Open
Abstract
Oculocutaneous syndromes are often due to mutations in single genes. In some cases, mouse models for these diseases exist in spontaneously occurring mutations, or in mice resulting from forward mutatagenesis screens. Here we present novel genes that may be causative for oculocutaneous disease in humans, discovered as part of a genome-wide screen of knockout-mice in a targeted single-gene deletion project. The International Mouse Phenotyping Consortium (IMPC) database (data release 10.0) was interrogated for all mouse strains with integument abnormalities, which were then cross-referenced individually to identify knockouts with concomitant ocular abnormalities attributed to the same targeted gene deletion. The search yielded 307 knockout strains from unique genes with integument abnormalities, 226 of which have not been previously associated with oculocutaneous conditions. Of the 307 knockout strains with integument abnormalities, 52 were determined to have ocular changes attributed to the targeted deletion, 35 of which represent novel oculocutaneous genes. Some examples of various integument abnormalities are shown, as well as two examples of knockout strains with oculocutaneous phenotypes. Each of the novel genes provided here are potentially relevant to the pathophysiology of human integumentary, or oculocutaneous conditions, such as albinism, phakomatoses, or other multi-system syndromes. The novel genes reported here may implicate molecular pathways relevant to these human diseases and may contribute to the discovery of novel therapeutic targets.
Collapse
Affiliation(s)
- Bret A Moore
- William R. Pritchard Veterinary Medical Teaching Hospital, School of Veterinary Medicine, University of California Davis, Davis, CA, United States
| | - Ann M Flenniken
- The Centre for Phenogenomics, Toronto, ON, M5T 3H7, Canada.,Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, M5G 1X5, Canada
| | - Dave Clary
- Department of Surgery, School of Medicine, and Mouse Biology Program, University of California Davis, Davis, CA, United States
| | - Ata S Moshiri
- Division of Dermatology, Department of Medicine, University of Washington, Seattle, WA, United States
| | - Lauryl M J Nutter
- The Centre for Phenogenomics, Toronto, ON, M5T 3H7, Canada.,The Hospital for Sick Children, Toronto, ON, M5G 1X8, Canada
| | - Zorana Berberovic
- The Centre for Phenogenomics, Toronto, ON, M5T 3H7, Canada.,Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, M5G 1X5, Canada
| | - Celeste Owen
- The Centre for Phenogenomics, Toronto, ON, M5T 3H7, Canada.,Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, M5G 1X5, Canada
| | - Susan Newbigging
- The Centre for Phenogenomics, Toronto, ON, M5T 3H7, Canada.,The Hospital for Sick Children, Toronto, ON, M5G 1X8, Canada
| | - Hibret Adissu
- The Centre for Phenogenomics, Toronto, ON, M5T 3H7, Canada.,The Hospital for Sick Children, Toronto, ON, M5G 1X8, Canada
| | - Mohammad Eskandarian
- The Centre for Phenogenomics, Toronto, ON, M5T 3H7, Canada.,Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, M5G 1X5, Canada
| | - Colin McKerlie
- The Centre for Phenogenomics, Toronto, ON, M5T 3H7, Canada
| | | | - Sara M Thomasy
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California Davis, Davis, CA, United States.,Department of Ophthalmology & Vision Science, School of Medicine, University of California Davis, Sacramento, CA, United States
| | - K C Kent Lloyd
- Department of Surgery, School of Medicine, and Mouse Biology Program, University of California Davis, Davis, CA, United States
| | - Christopher J Murphy
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California Davis, Davis, CA, United States.,Department of Ophthalmology & Vision Science, School of Medicine, University of California Davis, Sacramento, CA, United States
| | - Ala Moshiri
- Department of Ophthalmology & Vision Science, School of Medicine, University of California Davis, Sacramento, CA, United States.
| |
Collapse
|
27
|
Balasubramanian K, Weis M, Eyre DR, Martin J, Ortiz-Sanchez J, Duran I, Vangala S, Wang J, Friedman RA, Krakow D, Cohn DH. The α2 chain of type IX collagen is essential for type IX collagen biosynthesis. Am J Med Genet A 2019; 179:1672-1677. [PMID: 31161720 DOI: 10.1002/ajmg.a.61208] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 04/22/2019] [Accepted: 04/24/2019] [Indexed: 11/08/2022]
Affiliation(s)
- Karthika Balasubramanian
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, California
| | - MaryAnn Weis
- Department of Orthopaedic Surgery, University of Washington, Seattle, Washington
| | - David R Eyre
- Department of Orthopaedic Surgery, University of Washington, Seattle, Washington
| | - Jorge Martin
- Department of Orthopaedic Surgery, University of California, Los Angeles, Los Angeles, California
| | - Jorge Ortiz-Sanchez
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, California
| | - Ivan Duran
- Department of Orthopaedic Surgery, University of California, Los Angeles, Los Angeles, California
| | - Sitaram Vangala
- Department of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Juemei Wang
- Caruso Department of Otolaryngology, Keck School of Medicine at USC, University of Southern California, Los Angeles, California
| | - Rick A Friedman
- Caruso Department of Otolaryngology, Keck School of Medicine at USC, University of Southern California, Los Angeles, California
| | - Deborah Krakow
- Department of Orthopaedic Surgery, University of California, Los Angeles, Los Angeles, California
| | - Daniel H Cohn
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, California.,Department of Orthopaedic Surgery, University of California, Los Angeles, Los Angeles, California
| |
Collapse
|
28
|
Barupal DK, Zhang Y, Shen T, Fan S, Roberts BS, Fitzgerald P, Wancewicz B, Valdiviez L, Wohlgemuth G, Byram G, Choy YY, Haffner B, Showalter MR, Vaniya A, Bloszies CS, Folz JS, Kind T, Flenniken AM, McKerlie C, Nutter LMJ, Lloyd KC, Fiehn O. A Comprehensive Plasma Metabolomics Dataset for a Cohort of Mouse Knockouts within the International Mouse Phenotyping Consortium. Metabolites 2019; 9:E101. [PMID: 31121816 PMCID: PMC6571919 DOI: 10.3390/metabo9050101] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 05/11/2019] [Accepted: 05/13/2019] [Indexed: 02/08/2023] Open
Abstract
Mouse knockouts facilitate the study ofgene functions. Often, multiple abnormal phenotypes are induced when a gene is inactivated. The International Mouse Phenotyping Consortium (IMPC) has generated thousands of mouse knockouts and catalogued their phenotype data. We have acquired metabolomics data from 220 plasma samples from 30 unique mouse gene knockouts and corresponding wildtype mice from the IMPC. To acquire comprehensive metabolomics data, we have used liquid chromatography (LC) combined with mass spectrometry (MS) for detecting polar and lipophilic compounds in an untargeted approach. We have also used targeted methods to measure bile acids, steroids and oxylipins. In addition, we have used gas chromatography GC-TOFMS for measuring primary metabolites. The metabolomics dataset reports 832 unique structurally identified metabolites from 124 chemical classes as determined by ChemRICH software. The GCMS and LCMS raw data files, intermediate and finalized data matrices, R-Scripts, annotation databases, and extracted ion chromatograms are provided in this data descriptor. The dataset can be used for subsequent studies to link genetic variants with molecular mechanisms and phenotypes.
Collapse
Affiliation(s)
- Dinesh K Barupal
- NIH-West Coast Metabolomics Center, University of California, Davis, 451 Health Sciences Drive, Davis, CA 95616, USA.
| | - Ying Zhang
- NIH-West Coast Metabolomics Center, University of California, Davis, 451 Health Sciences Drive, Davis, CA 95616, USA.
| | - Tong Shen
- NIH-West Coast Metabolomics Center, University of California, Davis, 451 Health Sciences Drive, Davis, CA 95616, USA.
| | - Sili Fan
- NIH-West Coast Metabolomics Center, University of California, Davis, 451 Health Sciences Drive, Davis, CA 95616, USA.
| | - Bryan S Roberts
- NIH-West Coast Metabolomics Center, University of California, Davis, 451 Health Sciences Drive, Davis, CA 95616, USA.
| | - Patrick Fitzgerald
- NIH-West Coast Metabolomics Center, University of California, Davis, 451 Health Sciences Drive, Davis, CA 95616, USA.
| | - Benjamin Wancewicz
- NIH-West Coast Metabolomics Center, University of California, Davis, 451 Health Sciences Drive, Davis, CA 95616, USA.
| | - Luis Valdiviez
- NIH-West Coast Metabolomics Center, University of California, Davis, 451 Health Sciences Drive, Davis, CA 95616, USA.
| | - Gert Wohlgemuth
- NIH-West Coast Metabolomics Center, University of California, Davis, 451 Health Sciences Drive, Davis, CA 95616, USA.
| | - Gregory Byram
- NIH-West Coast Metabolomics Center, University of California, Davis, 451 Health Sciences Drive, Davis, CA 95616, USA.
| | - Ying Yng Choy
- NIH-West Coast Metabolomics Center, University of California, Davis, 451 Health Sciences Drive, Davis, CA 95616, USA.
| | - Bennett Haffner
- NIH-West Coast Metabolomics Center, University of California, Davis, 451 Health Sciences Drive, Davis, CA 95616, USA.
| | - Megan R Showalter
- NIH-West Coast Metabolomics Center, University of California, Davis, 451 Health Sciences Drive, Davis, CA 95616, USA.
| | - Arpana Vaniya
- NIH-West Coast Metabolomics Center, University of California, Davis, 451 Health Sciences Drive, Davis, CA 95616, USA.
| | - Clayton S Bloszies
- NIH-West Coast Metabolomics Center, University of California, Davis, 451 Health Sciences Drive, Davis, CA 95616, USA.
| | - Jacob S Folz
- NIH-West Coast Metabolomics Center, University of California, Davis, 451 Health Sciences Drive, Davis, CA 95616, USA.
| | - Tobias Kind
- NIH-West Coast Metabolomics Center, University of California, Davis, 451 Health Sciences Drive, Davis, CA 95616, USA.
| | - Ann M Flenniken
- The Centre for Phenogenomics, Toronto, ON M5T 3H7, Canada.
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada.
| | - Colin McKerlie
- The Centre for Phenogenomics, Toronto, ON M5T 3H7, Canada.
- The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada.
| | - Lauryl M J Nutter
- The Centre for Phenogenomics, Toronto, ON M5T 3H7, Canada.
- The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada.
| | - Kent C Lloyd
- Mouse Biology Program, University of California, Davis, Davis, CA 95616, USA.
| | - Oliver Fiehn
- NIH-West Coast Metabolomics Center, University of California, Davis, 451 Health Sciences Drive, Davis, CA 95616, USA.
| |
Collapse
|
29
|
Fox CS. Using Human Genetics to Drive Drug Discovery: A Perspective. Am J Kidney Dis 2019; 74:111-119. [PMID: 30898364 DOI: 10.1053/j.ajkd.2018.12.045] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2018] [Accepted: 12/24/2018] [Indexed: 12/11/2022]
Abstract
The probability of success of developing medicines to treat human disease can be improved by leveraging human genetics. Different types of genetic data and techniques, including genome-wide association, whole-exome sequencing, and whole-genome sequencing, can be used to gain insight into human disease. Layering different types of genetic evidence from Mendelian disease, coding variants, and common variation can bolster support for a genetic target. Human knockouts offer the potential to perform reverse genetic screens in humans to identify physiologically relevant targets. Other components of a good genetic target include protective loss-of-function mutations, some degree of known biology, tractability, and a clean on-target safety profile. In addition to using human genetics to inspire new drug programs, phenome-wide association studies can be used to identify alternative indications or repurposing opportunities. This information can be combined into a 5-step approach for selecting a genetic target for validation, which is presented in detail in this review. Finally, current challenges in leveraging human genetics are highlighted, including the difficulties translating certain types of genetic data, relatively small number of bona fide disease-associated coding rare variants, and current sample sizes of large well-curated biobanks linked to comprehensive genetic information.
Collapse
|
30
|
Kang SK, Hawkins NA, Kearney JA. C57BL/6J and C57BL/6N substrains differentially influence phenotype severity in the Scn1a +/- mouse model of Dravet syndrome. Epilepsia Open 2019; 4:164-169. [PMID: 30868126 PMCID: PMC6398090 DOI: 10.1002/epi4.12287] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 10/05/2018] [Accepted: 11/13/2018] [Indexed: 01/01/2023] Open
Abstract
Many disease-relevant phenotypes modeled in inbred mice have been shown to be strain-dependent, indicating the important influence of genetic background on disease phenotypes. Although C57BL/6 mice are one of the most commonly used inbred strains in laboratory research, there are multiple substrains (eg, B6J vs B6N) that have been separated for more than 50 years. Thus, understanding the substrain differences is important for scientific rigor and reproducibility. In this study, seizure susceptibility, spontaneous seizures, and survival were compared between Scn1a +/- mice on (C57BL/6J × 129S6/SvEvTac)F1 (F1J) vs (C57BL/6N × 129S6/SvEvTac)F1 (F1N) strain backgrounds. F1N.Scn1a +/- mice were more susceptible to hyperthermia-induced seizures, yet had milder spontaneous seizures and improved survival relative to F1J.Scn1a +/- mice. Our results indicate that choice of C57BL/6 substrain may significantly alter disease phenotypes and should be considered carefully in experimental design using the Scn1a +/- Dravet mouse model, as well as other mouse models of epilepsy.
Collapse
Affiliation(s)
- Seok K. Kang
- Interdepartmental Neuroscience ProgramFeinberg School of MedicineNorthwestern UniversityChicagoIllinois
| | - Nicole A. Hawkins
- Department of PharmacologyFeinberg School of MedicineNorthwestern UniversityChicagoIllinois
| | - Jennifer A. Kearney
- Interdepartmental Neuroscience ProgramFeinberg School of MedicineNorthwestern UniversityChicagoIllinois
- Department of PharmacologyFeinberg School of MedicineNorthwestern UniversityChicagoIllinois
| |
Collapse
|
31
|
Hmeljak J, Justice MJ. From gene to treatment: supporting rare disease translational research through model systems. Dis Model Mech 2019; 12:12/2/dmm039271. [PMID: 30819728 PMCID: PMC6398488 DOI: 10.1242/dmm.039271] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Individual rare diseases may affect only a few people, making them difficult to recognize, diagnose or treat by studying humans alone. Instead, model organisms help to validate genetic associations, understand functional pathways and develop therapeutic interventions for rare diseases. In this Editorial, we point to the key parameters in face, construct, predictive and target validity for accurate disease modelling, with special emphasis on rare disease models. Raising the experimental standards for disease models will enhance successful clinical translation and benefit rare disease research. Summary: This Editorial discusses the importance of model systems with accurate face, construct, target and predictive validity for rare disease research.
Collapse
Affiliation(s)
- Julija Hmeljak
- Disease Models & Mechanisms, The Company of Biologists, Bidder Building, Station Road, Histon, Cambridge CB24 9LF, UK
| | - Monica J Justice
- Program in Genetics and Genome Biology, The Hospital for Sick Children, and Department of Molecular Genetics, The University of Toronto, Toronto, Ontario M5G 0A4 Canada
| |
Collapse
|
32
|
Moore BA, Roux MJ, Sebbag L, Cooper A, Edwards SG, Leonard BC, Imai DM, Griffey S, Bower L, Clary D, Lloyd KCK, Hérault Y, Thomasy SM, Murphy CJ, Moshiri A. A Population Study of Common Ocular Abnormalities in C57BL/6N rd8 Mice. Invest Ophthalmol Vis Sci 2019; 59:2252-2261. [PMID: 29847629 PMCID: PMC5935295 DOI: 10.1167/iovs.17-23513] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Purpose The purpose of this study was to quantify the frequency and severity of ocular abnormalities affecting wild-type C57BL/6N mice, the most common strain used worldwide for the creation of single-gene knockouts. Methods A total of 2773 animals (5546 eyes) were examined at one colony at UC Davis and in three more colonies at the Institut Clinique de la Souris in Strasbourg, France. Mice were examined at 15 to 16 weeks postnatal age by performing anterior segment biomicroscopy, posterior segment examination by indirect ophthalmoscopy, intraocular pressure measurement, and optical coherence tomography of anterior and posterior segment structures. Results Common ocular findings in the C57BL/6N strain included corneal deposits (3%), increased optical density of the anterior lens capsule (67%), punctate nuclear cataracts (98%), vitreous crystalline deposits (61%), hyaloid vascular remnant (6%), and retinal dysplasia attributed to the rd8 mutation (58%). Interestingly, retinal dysplasia was more common in male mice in all four breeding colonies evaluated in this study. The thickness of ocular tissues and compartments were measured by spectral-domain optical coherence tomography, including the central cornea, anterior chamber, vitreous, and retinal layers. Intraocular pressure was measured by rebound tonometry. Conclusions Ocular abnormalities are common in anterior and posterior segments of the C57BL/6N mouse, the most common background on which single-gene knockout mice have been made. It is important that vision scientists understand the extent and variability of ocular findings associated with this particular genetic background of mice.
Collapse
Affiliation(s)
- Bret A Moore
- William R. Pritchard Veterinary Medical Teaching Hospital, School of Veterinary Medicine, University of California-Davis, Davis, California, United States
| | - Michel J Roux
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Université de Strasbourg, Illkirch, France.,Centre National de la Recherche Scientifique, UMR7104, Illkirch, France.,Institut National de la Santé et de la Recherche Médicale, U964, Illkirch, France.,Université de Strasbourg, Illkirch, France
| | - Lionel Sebbag
- William R. Pritchard Veterinary Medical Teaching Hospital, School of Veterinary Medicine, University of California-Davis, Davis, California, United States
| | - Ann Cooper
- William R. Pritchard Veterinary Medical Teaching Hospital, School of Veterinary Medicine, University of California-Davis, Davis, California, United States
| | - Sydney G Edwards
- William R. Pritchard Veterinary Medical Teaching Hospital, School of Veterinary Medicine, University of California-Davis, Davis, California, United States
| | - Brian C Leonard
- William R. Pritchard Veterinary Medical Teaching Hospital, School of Veterinary Medicine, University of California-Davis, Davis, California, United States
| | - Denise M Imai
- Comparative Pathology Laboratory, School of Veterinary Medicine, UC Davis, Davis, California, United States
| | - Stephen Griffey
- Comparative Pathology Laboratory, School of Veterinary Medicine, UC Davis, Davis, California, United States
| | - Lynette Bower
- Mouse Biology Program, University of California-Davis, Davis, California, United States
| | - Dave Clary
- Mouse Biology Program, University of California-Davis, Davis, California, United States
| | - K C Kent Lloyd
- Mouse Biology Program, University of California-Davis, Davis, California, United States.,Department of Surgery, School of Medicine, University of California-Davis, Sacramento, California, United States
| | - Yann Hérault
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Université de Strasbourg, Illkirch, France.,Centre National de la Recherche Scientifique, UMR7104, Illkirch, France.,Institut National de la Santé et de la Recherche Médicale, U964, Illkirch, France.,Université de Strasbourg, Illkirch, France.,CELPHEDIA, PHENOMIN, Institut Clinique de la Souris (ICS), CNRS, INSERM, Université de Strasbourg, Illkirch, France
| | - Sara M Thomasy
- Department of Veterinary Surgical and Radiological Sciences, School of Veterinary Medicine, UC Davis, Davis, California, United States.,Department of Ophthalmology & Vision Science, School of Medicine, University of California-Davis, Sacramento, California, United States
| | - Christopher J Murphy
- Department of Veterinary Surgical and Radiological Sciences, School of Veterinary Medicine, UC Davis, Davis, California, United States.,Department of Ophthalmology & Vision Science, School of Medicine, University of California-Davis, Sacramento, California, United States
| | - Ala Moshiri
- Department of Ophthalmology & Vision Science, School of Medicine, University of California-Davis, Sacramento, California, United States
| |
Collapse
|
33
|
Moore BA, Leonard BC, Sebbag L, Edwards SG, Cooper A, Imai DM, Straiton E, Santos L, Reilly C, Griffey SM, Bower L, Clary D, Mason J, Roux MJ, Meziane H, Herault Y, McKerlie C, Flenniken AM, Nutter LMJ, Berberovic Z, Owen C, Newbigging S, Adissu H, Eskandarian M, Hsu CW, Kalaga S, Udensi U, Asomugha C, Bohat R, Gallegos JJ, Seavitt JR, Heaney JD, Beaudet AL, Dickinson ME, Justice MJ, Philip V, Kumar V, Svenson KL, Braun RE, Wells S, Cater H, Stewart M, Clementson-Mobbs S, Joynson R, Gao X, Suzuki T, Wakana S, Smedley D, Seong JK, Tocchini-Valentini G, Moore M, Fletcher C, Karp N, Ramirez-Solis R, White JK, de Angelis MH, Wurst W, Thomasy SM, Flicek P, Parkinson H, Brown SDM, Meehan TF, Nishina PM, Murray SA, Krebs MP, Mallon AM, Lloyd KCK, Murphy CJ, Moshiri A. Identification of genes required for eye development by high-throughput screening of mouse knockouts. Commun Biol 2018; 1:236. [PMID: 30588515 PMCID: PMC6303268 DOI: 10.1038/s42003-018-0226-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Accepted: 11/06/2018] [Indexed: 12/19/2022] Open
Abstract
Despite advances in next generation sequencing technologies, determining the genetic basis of ocular disease remains a major challenge due to the limited access and prohibitive cost of human forward genetics. Thus, less than 4,000 genes currently have available phenotype information for any organ system. Here we report the ophthalmic findings from the International Mouse Phenotyping Consortium, a large-scale functional genetic screen with the goal of generating and phenotyping a null mutant for every mouse gene. Of 4364 genes evaluated, 347 were identified to influence ocular phenotypes, 75% of which are entirely novel in ocular pathology. This discovery greatly increases the current number of genes known to contribute to ophthalmic disease, and it is likely that many of the genes will subsequently prove to be important in human ocular development and disease. Bret Moore et al. from the International Mouse Phenotyping Consortium report the identification of 347 mouse genes that influence ocular phenotypes when knocked out. 75% of the identified genes have not previously been associated with any ocular pathology.
Collapse
Affiliation(s)
- Bret A Moore
- William R. Pritchard Veterinary Medical Teaching Hospital, School of Veterinary Medicine, University of California-Davis, Davis, 95616, CA, USA
| | - Brian C Leonard
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California-Davis, Davis, CA, 95616, USA
| | - Lionel Sebbag
- William R. Pritchard Veterinary Medical Teaching Hospital, School of Veterinary Medicine, University of California-Davis, Davis, 95616, CA, USA
| | - Sydney G Edwards
- William R. Pritchard Veterinary Medical Teaching Hospital, School of Veterinary Medicine, University of California-Davis, Davis, 95616, CA, USA
| | - Ann Cooper
- William R. Pritchard Veterinary Medical Teaching Hospital, School of Veterinary Medicine, University of California-Davis, Davis, 95616, CA, USA
| | - Denise M Imai
- Comparative Pathology Laboratory, School of Veterinary Medicine, University of California-Davis, Davis, CA, 95616, USA
| | - Ewan Straiton
- Medical Research Council Harwell Institute (Mammalian Genetis Unit and Mary Lyon Center, Harwell, Oxfordshire, OX11 0RD, UK
| | - Luis Santos
- Medical Research Council Harwell Institute (Mammalian Genetis Unit and Mary Lyon Center, Harwell, Oxfordshire, OX11 0RD, UK
| | - Christopher Reilly
- Comparative Pathology Laboratory, School of Veterinary Medicine, University of California-Davis, Davis, CA, 95616, USA
| | - Stephen M Griffey
- Comparative Pathology Laboratory, School of Veterinary Medicine, University of California-Davis, Davis, CA, 95616, USA
| | - Lynette Bower
- Mouse Biology Program, and Department of Surgery, School of Medicine, University of California-Davis, Davis, CA, 95618, USA
| | - David Clary
- Mouse Biology Program, and Department of Surgery, School of Medicine, University of California-Davis, Davis, CA, 95618, USA
| | - Jeremy Mason
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1 SD, UK
| | - Michel J Roux
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Université de Strasbourg, 1 rue Laurent Fries, 67404, Illkirch, France.,Centre National de la Recherche Scientifique, UMR7104, Illkirch, France.,Institut National de la Santé et de la Recherche Médicale, U1258, Illkirch, France.,Université de Strasbourg, 1 rue Laurent Fries, 67404, Illkirch, France.,CELPHEDIA, PHENOMIN, Institut Clinique de la Souris (ICS), CNRS, INSERM, University of Strasbourg, 1 rue Laurent Fries, 67404, Illkirch-Graffenstaden, France
| | - Hamid Meziane
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Université de Strasbourg, 1 rue Laurent Fries, 67404, Illkirch, France.,Centre National de la Recherche Scientifique, UMR7104, Illkirch, France.,Institut National de la Santé et de la Recherche Médicale, U1258, Illkirch, France.,Université de Strasbourg, 1 rue Laurent Fries, 67404, Illkirch, France.,CELPHEDIA, PHENOMIN, Institut Clinique de la Souris (ICS), CNRS, INSERM, University of Strasbourg, 1 rue Laurent Fries, 67404, Illkirch-Graffenstaden, France
| | - Yann Herault
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Université de Strasbourg, 1 rue Laurent Fries, 67404, Illkirch, France.,Centre National de la Recherche Scientifique, UMR7104, Illkirch, France.,Institut National de la Santé et de la Recherche Médicale, U1258, Illkirch, France.,Université de Strasbourg, 1 rue Laurent Fries, 67404, Illkirch, France.,CELPHEDIA, PHENOMIN, Institut Clinique de la Souris (ICS), CNRS, INSERM, University of Strasbourg, 1 rue Laurent Fries, 67404, Illkirch-Graffenstaden, France
| | | | - Colin McKerlie
- The Centre for Phenogenomics, Toronto, ON, M5T 3H7, Canada.,The Hospital for Sick Children, Toronto, ON, M5G 1X8, Canada
| | - Ann M Flenniken
- The Centre for Phenogenomics, Toronto, ON, M5T 3H7, Canada.,Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, M5G 1X5, Canada
| | - Lauryl M J Nutter
- The Centre for Phenogenomics, Toronto, ON, M5T 3H7, Canada.,The Hospital for Sick Children, Toronto, ON, M5G 1X8, Canada
| | - Zorana Berberovic
- The Centre for Phenogenomics, Toronto, ON, M5T 3H7, Canada.,Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, M5G 1X5, Canada
| | - Celeste Owen
- The Centre for Phenogenomics, Toronto, ON, M5T 3H7, Canada.,Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, M5G 1X5, Canada
| | - Susan Newbigging
- The Centre for Phenogenomics, Toronto, ON, M5T 3H7, Canada.,Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, M5G 1X5, Canada
| | - Hibret Adissu
- The Centre for Phenogenomics, Toronto, ON, M5T 3H7, Canada.,Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, M5G 1X5, Canada
| | - Mohammed Eskandarian
- The Centre for Phenogenomics, Toronto, ON, M5T 3H7, Canada.,Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, M5G 1X5, Canada
| | - Chih-Wei Hsu
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Sowmya Kalaga
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Uchechukwu Udensi
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Chinwe Asomugha
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Ritu Bohat
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Juan J Gallegos
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - John R Seavitt
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Jason D Heaney
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Arthur L Beaudet
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Mary E Dickinson
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, 77030, USA.,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Monica J Justice
- The Centre for Phenogenomics, Toronto, ON, M5T 3H7, Canada.,The Hospital for Sick Children, Toronto, ON, M5G 1X8, Canada.,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Vivek Philip
- The Jackson Laboratory, Bar Harbor, ME, 04609, USA
| | - Vivek Kumar
- The Jackson Laboratory, Bar Harbor, ME, 04609, USA
| | | | | | - Sara Wells
- Medical Research Council Harwell Institute (Mammalian Genetis Unit and Mary Lyon Center, Harwell, Oxfordshire, OX11 0RD, UK
| | - Heather Cater
- Medical Research Council Harwell Institute (Mammalian Genetis Unit and Mary Lyon Center, Harwell, Oxfordshire, OX11 0RD, UK
| | - Michelle Stewart
- Medical Research Council Harwell Institute (Mammalian Genetis Unit and Mary Lyon Center, Harwell, Oxfordshire, OX11 0RD, UK
| | - Sharon Clementson-Mobbs
- Medical Research Council Harwell Institute (Mammalian Genetis Unit and Mary Lyon Center, Harwell, Oxfordshire, OX11 0RD, UK
| | - Russell Joynson
- Medical Research Council Harwell Institute (Mammalian Genetis Unit and Mary Lyon Center, Harwell, Oxfordshire, OX11 0RD, UK
| | - Xiang Gao
- SKL of Pharmaceutical Biotechnology and Model Animal Research Center, Collaborative Innovation Center for Genetics and Development, Nanjing Biomedical Research Institute, Nanjing University, Nanjing, 210061, China
| | | | | | - Damian Smedley
- Clinical Pharmacology, Charterhouse Square, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, EC1M 6BQ, UK
| | - J K Seong
- Korea Mouse Phenotyping Consortium (KMPC) and BK21 Program for Veterinary Science, Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, 599 Gwanangno, Gwanak-gu, Seoul, 08826, South Korea
| | - Glauco Tocchini-Valentini
- Monterotondo Mouse Clinic, Italian National Research Council (CNR), Institute of Cell Biology and Neurobiology, Adriano Buzzati-Traverso Campus, Via Ramarini, I-00015, Monterotondo Scalo, Italy
| | - Mark Moore
- International Mouse Phenotyping Consortium, San Anselmo, CA, 94960, USA
| | | | - Natasha Karp
- The Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK
| | - Ramiro Ramirez-Solis
- The Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK
| | - Jacqueline K White
- The Jackson Laboratory, Bar Harbor, ME, 04609, USA.,The Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK
| | - Martin Hrabe de Angelis
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstraße 1, 85764, Neuherberg, Germany
| | - Wolfgang Wurst
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstraße 1, 85764, Neuherberg, Germany
| | - Sara M Thomasy
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California-Davis, Davis, CA, 95616, USA.,Department of Ophthalmology & Vision Science, School of Medicine, U.C. Davis, Sacramento, CA, 95817, USA
| | - Paul Flicek
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1 SD, UK
| | - Helen Parkinson
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1 SD, UK
| | - Steve D M Brown
- Medical Research Council Harwell Institute (Mammalian Genetis Unit and Mary Lyon Center, Harwell, Oxfordshire, OX11 0RD, UK
| | - Terrence F Meehan
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1 SD, UK
| | | | | | - Mark P Krebs
- The Jackson Laboratory, Bar Harbor, ME, 04609, USA
| | - Ann-Marie Mallon
- Medical Research Council Harwell Institute (Mammalian Genetis Unit and Mary Lyon Center, Harwell, Oxfordshire, OX11 0RD, UK
| | - K C Kent Lloyd
- Mouse Biology Program, and Department of Surgery, School of Medicine, University of California-Davis, Davis, CA, 95618, USA
| | - Christopher J Murphy
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California-Davis, Davis, CA, 95616, USA. .,Department of Ophthalmology & Vision Science, School of Medicine, U.C. Davis, Sacramento, CA, 95817, USA.
| | - Ala Moshiri
- Department of Ophthalmology & Vision Science, School of Medicine, U.C. Davis, Sacramento, CA, 95817, USA.
| |
Collapse
|
34
|
Tian D, Wenlock S, Kabir M, Tzotzos G, Doig AJ, Hentges KE. Identifying mouse developmental essential genes using machine learning. Dis Model Mech 2018; 11:11/12/dmm034546. [PMID: 30563825 PMCID: PMC6307915 DOI: 10.1242/dmm.034546] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 10/19/2018] [Indexed: 12/20/2022] Open
Abstract
The genes that are required for organismal survival are annotated as ‘essential genes’. Identifying all the essential genes of an animal species can reveal critical functions that are needed during the development of the organism. To inform studies on mouse development, we developed a supervised machine learning classifier based on phenotype data from mouse knockout experiments. We used this classifier to predict the essentiality of mouse genes lacking experimental data. Validation of our predictions against a blind test set of recent mouse knockout experimental data indicated a high level of accuracy (>80%). We also validated our predictions for other mouse mutagenesis methodologies, demonstrating that the predictions are accurate for lethal phenotypes isolated in random chemical mutagenesis screens and embryonic stem cell screens. The biological functions that are enriched in essential and non-essential genes have been identified, showing that essential genes tend to encode intracellular proteins that interact with nucleic acids. The genome distribution of predicted essential and non-essential genes was analysed, demonstrating that the density of essential genes varies throughout the genome. A comparison with human essential and non-essential genes was performed, revealing conservation between human and mouse gene essentiality status. Our genome-wide predictions of mouse essential genes will be of value for the planning of mouse knockout experiments and phenotyping assays, for understanding the functional processes required during mouse development, and for the prioritisation of disease candidate genes identified in human genome and exome sequence datasets. Summary: Here, we used computer-based machine learning methodology to predict which genes in the mouse genome are essential for development, and present a database of mouse essential and non-essential genes.
Collapse
Affiliation(s)
- David Tian
- Division of Evolution and Genomic Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Stephanie Wenlock
- Division of Evolution and Genomic Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Mitra Kabir
- Division of Evolution and Genomic Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - George Tzotzos
- Department of Agriculture, Food and Environmental Sciences, Marche Polytechnic University, Ancona 60121, Italy
| | - Andrew J Doig
- Manchester Institute of Biotechnology, The University of Manchester, 131 Princess Street, Manchester M1 7DN, UK .,Division of Neuroscience and Experimental Psychology, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, UK
| | - Kathryn E Hentges
- Division of Evolution and Genomic Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Oxford Road, Manchester M13 9PT, UK
| |
Collapse
|
35
|
Fuentes R, Letelier J, Tajer B, Valdivia LE, Mullins MC. Fishing forward and reverse: Advances in zebrafish phenomics. Mech Dev 2018; 154:296-308. [PMID: 30130581 PMCID: PMC6289646 DOI: 10.1016/j.mod.2018.08.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 08/06/2018] [Accepted: 08/17/2018] [Indexed: 12/15/2022]
Abstract
Understanding how the genome instructs the phenotypic characteristics of an organism is one of the major scientific endeavors of our time. Advances in genetics have progressively deciphered the inheritance, identity and biological relevance of genetically encoded information, contributing to the rise of several, complementary omic disciplines. One of them is phenomics, an emergent area of biology dedicated to the systematic multi-scale analysis of phenotypic traits. This discipline provides valuable gene function information to the rapidly evolving field of genetics. Current molecular tools enable genome-wide analyses that link gene sequence to function in multi-cellular organisms, illuminating the genome-phenome relationship. Among vertebrates, zebrafish has emerged as an outstanding model organism for high-throughput phenotyping and modeling of human disorders. Advances in both systematic mutagenesis and phenotypic analyses of embryonic and post-embryonic stages in zebrafish have revealed the function of a valuable collection of genes and the general structure of several complex traits. In this review, we summarize multiple large-scale genetic efforts addressing parental, embryonic, and adult phenotyping in the zebrafish. The genetic and quantitative tools available in the zebrafish model, coupled with the broad spectrum of phenotypes that can be assayed, make it a powerful model for phenomics, well suited for the dissection of genotype-phenotype associations in development, physiology, health and disease.
Collapse
Affiliation(s)
- Ricardo Fuentes
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Joaquín Letelier
- Centro Andaluz de Biología del Desarrollo (CSIC/UPO/JA), Seville, Spain; Center for Integrative Biology, Facultad de Ciencias, Universidad Mayor, Santiago, Chile
| | - Benjamin Tajer
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Leonardo E Valdivia
- Center for Integrative Biology, Facultad de Ciencias, Universidad Mayor, Santiago, Chile.
| | - Mary C Mullins
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
36
|
Baldock RA, Armit C. eHistology image and annotation data from the Kaufman Atlas of Mouse Development. Gigascience 2018; 7:4768197. [PMID: 29272399 PMCID: PMC5827353 DOI: 10.1093/gigascience/gix131] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Accepted: 12/13/2017] [Indexed: 01/15/2023] Open
Abstract
“The Atlas of Mouse Development” by Kaufman is a classic paper atlas that is the de facto standard for the definition of mouse embryo anatomy in the context of standard histological images. We have redigitized the original haematoxylin and eosin–stained tissue sections used for the book at high resolution and transferred the hand-drawn annotations to digital form. We have augmented the annotations with standard ontological assignments (EMAPA anatomy) and made the data freely available via an online viewer (eHistology) and from the University of Edinburgh DataShare archive. The dataset captures and preserves the definitive anatomical knowledge of the original atlas, provides a core image set for deeper community annotation and teaching, and delivers a unique high-quality set of high-resolution histological images through mammalian development for manual and automated analysis.
Collapse
Affiliation(s)
- Richard A Baldock
- MRC Human Genetics Unit, Institute of Genomic and Molecular Medicine, University of Edinburgh, Crewe Road, Edinburgh EH4 2XU, UK
| | - Chris Armit
- MRC Human Genetics Unit, Institute of Genomic and Molecular Medicine, University of Edinburgh, Crewe Road, Edinburgh EH4 2XU, UK
| |
Collapse
|
37
|
Brown JM, Horner NR, Lawson TN, Fiegel T, Greenaway S, Morgan H, Ring N, Santos L, Sneddon D, Teboul L, Vibert J, Yaikhom G, Westerberg H, Mallon AM. A bioimage informatics platform for high-throughput embryo phenotyping. Brief Bioinform 2018; 19:41-51. [PMID: 27742664 PMCID: PMC5862285 DOI: 10.1093/bib/bbw101] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Indexed: 11/13/2022] Open
Abstract
High-throughput phenotyping is a cornerstone of numerous functional genomics projects. In recent years, imaging screens have become increasingly important in understanding gene-phenotype relationships in studies of cells, tissues and whole organisms. Three-dimensional (3D) imaging has risen to prominence in the field of developmental biology for its ability to capture whole embryo morphology and gene expression, as exemplified by the International Mouse Phenotyping Consortium (IMPC). Large volumes of image data are being acquired by multiple institutions around the world that encompass a range of modalities, proprietary software and metadata. To facilitate robust downstream analysis, images and metadata must be standardized to account for these differences. As an open scientific enterprise, making the data readily accessible is essential so that members of biomedical and clinical research communities can study the images for themselves without the need for highly specialized software or technical expertise. In this article, we present a platform of software tools that facilitate the upload, analysis and dissemination of 3D images for the IMPC. Over 750 reconstructions from 80 embryonic lethal and subviable lines have been captured to date, all of which are openly accessible at mousephenotype.org. Although designed for the IMPC, all software is available under an open-source licence for others to use and develop further. Ongoing developments aim to increase throughput and improve the analysis and dissemination of image data. Furthermore, we aim to ensure that images are searchable so that users can locate relevant images associated with genes, phenotypes or human diseases of interest.
Collapse
Affiliation(s)
- James M Brown
- MRC Harwell Institute, Harwell Campus, Oxfordshire
- Corresponding author: James Brown, MRC Harwell Institute, Harwell Campus, Oxfordshire, OX11 0RD. Tel. +44-0-1235-841237; Fax: +44-0-1235-841172; E-mail:
| | | | | | - Tanja Fiegel
- MRC Harwell Institute, Harwell Campus, Oxfordshire
| | | | - Hugh Morgan
- MRC Harwell Institute, Harwell Campus, Oxfordshire
| | - Natalie Ring
- MRC Harwell Institute, Harwell Campus, Oxfordshire
| | - Luis Santos
- MRC Harwell Institute, Harwell Campus, Oxfordshire
| | | | - Lydia Teboul
- MRC Harwell Institute, Harwell Campus, Oxfordshire
| | | | | | | | | |
Collapse
|
38
|
Zhao Y, Song G, Ren J, Li Q, Zhong S, Cui Z. Sleeping beauty transposon-mediated poly(A)-trapping and insertion mutagenesis in mouse embryonic stem cells. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2018; 59:687-697. [PMID: 30280432 DOI: 10.1002/em.22234] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 07/03/2018] [Accepted: 07/12/2018] [Indexed: 06/08/2023]
Abstract
Saturation mutagenesis of all endogenous genes within the mouse genome remains a challenging task, although a plenty of gene-editing approaches are available for this purpose. Here, a poly(A)-trap vector was generated for insertion mutagenesis in mouse embryonic stem (mES) cells. This vector contains an expression cassette of neomycin (Neo)-resistant gene lacking a poly(A) signal and flanked by two inverted terminal repeats of the Sleeping Beauty (SB) transposon. The whole poly(A)-trap cassette can transpose into target TA dinucleotides, properly splice with endogenous genes and effectively interrupt the transcription of trapped genes in mES cells after transient induction of SB expression by doxycycline (DOX)-treatment at 1 μg/ml, leading to the formation of multiple geneticin (G418)-resistant cell clones. In the first round of mutation screening, we identified six transposition events from 23 cell clones, including four inserted into an endogenous gene and two landed between endogenous genes. The abilities of self-renewal, totipotency, genetic stability and differentiation of syngap1+/- cells were not affected by DOX-treatment and G418-selection. These findings suggest that this SB transposon-mediated poly(A)-trap vector can be used as an alternative tool for a large-scale screening of mES cells with a gene mutation and for further generation of mutant mouse strains. Environ. Mol. Mutagen. 59:687-697, 2018. © 2018 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Yi Zhao
- Department of Genetics, School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei, China
| | - Guili Song
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - Jing Ren
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - Qing Li
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - Shan Zhong
- Department of Genetics, School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei, China
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, Hubei, China
- Hubei Province Key Laboratory of Allergy and Immunology, Wuhan, Hubei, China
| | - Zongbin Cui
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| |
Collapse
|
39
|
Roy AL, Conroy RS. Toward mapping the human body at a cellular resolution. Mol Biol Cell 2018; 29:1779-1785. [PMID: 30058989 PMCID: PMC6085824 DOI: 10.1091/mbc.e18-04-0260] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 06/01/2018] [Accepted: 06/07/2018] [Indexed: 12/21/2022] Open
Abstract
The adult human body is composed of nearly 37 trillion cells, each with potentially unique molecular characteristics. This Perspective describes some of the challenges and opportunities faced in mapping the molecular characteristics of these cells in specific regions of the body and highlights areas for international collaboration toward the broader goal of comprehensively mapping the human body with cellular resolution.
Collapse
Affiliation(s)
- Ananda L. Roy
- Office of Strategic Coordination, Division of Program Coordination, Planning, and Strategic Initiatives, Office of the Director, National Institutes of Health, Bethesda, MD 20892
| | - Richard S. Conroy
- Office of Strategic Coordination, Division of Program Coordination, Planning, and Strategic Initiatives, Office of the Director, National Institutes of Health, Bethesda, MD 20892
| |
Collapse
|
40
|
Lanza DG, Gaspero A, Lorenzo I, Liao L, Zheng P, Wang Y, Deng Y, Cheng C, Zhang C, Seavitt JR, DeMayo FJ, Xu J, Dickinson ME, Beaudet AL, Heaney JD. Comparative analysis of single-stranded DNA donors to generate conditional null mouse alleles. BMC Biol 2018; 16:69. [PMID: 29925370 PMCID: PMC6011517 DOI: 10.1186/s12915-018-0529-0] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Accepted: 05/09/2018] [Indexed: 01/29/2023] Open
Abstract
BACKGROUND The International Mouse Phenotyping Consortium is generating null allele mice for every protein-coding gene in the genome and characterizing these mice to identify gene-phenotype associations. While CRISPR/Cas9-mediated null allele production in mice is highly efficient, generation of conditional alleles has proven to be more difficult. To test the feasibility of using CRISPR/Cas9 gene editing to generate conditional knockout mice for this large-scale resource, we employed Cas9-initiated homology-driven repair (HDR) with short and long single stranded oligodeoxynucleotides (ssODNs and lssDNAs). RESULTS Using pairs of single guide RNAs and short ssODNs to introduce loxP sites around a critical exon or exons, we obtained putative conditional allele founder mice, harboring both loxP sites, for 23 out of 30 targeted genes. LoxP sites integrated in cis in at least one mouse for 18 of 23 genes. However, loxP sites were mutagenized in 4 of the 18 in cis lines. HDR efficiency correlated with Cas9 cutting efficiency but was minimally influenced by ssODN homology arm symmetry. By contrast, using pairs of guides and single lssDNAs to introduce loxP-flanked exons, conditional allele founders were generated for all four genes targeted, although one founder was found to harbor undesired mutations within the lssDNA sequence interval. Importantly, when employing either ssODNs or lssDNAs, random integration events were detected. CONCLUSIONS Our studies demonstrate that Cas9-mediated HDR with pairs of ssODNs can generate conditional null alleles at many loci, but reveal inefficiencies when applied at scale. In contrast, lssDNAs are amenable to high-throughput production of conditional alleles when they can be employed. Regardless of the single-stranded donor utilized, it is essential to screen for sequence errors at sites of HDR and random insertion of donor sequences into the genome.
Collapse
Affiliation(s)
- Denise G Lanza
- Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, MS BCM225, Houston, TX, 77030, USA
- Mouse ES Cell Core, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, USA
| | - Angelina Gaspero
- Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, MS BCM225, Houston, TX, 77030, USA
| | - Isabel Lorenzo
- Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, MS BCM225, Houston, TX, 77030, USA
- Mouse ES Cell Core, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, USA
| | - Lan Liao
- Department of Molecular Biology, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, USA
- Genetically Engineered Mouse Core, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, USA
| | - Ping Zheng
- Mouse ES Cell Core, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, USA
| | - Ying Wang
- Mouse ES Cell Core, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, USA
| | - Yu Deng
- Lester and Sue Smith Breast Center, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, USA
| | - Chonghui Cheng
- Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, MS BCM225, Houston, TX, 77030, USA
- Department of Molecular Biology, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, USA
- Lester and Sue Smith Breast Center, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, USA
| | - Chuansheng Zhang
- Department of Neuroscience, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, USA
| | - John R Seavitt
- Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, MS BCM225, Houston, TX, 77030, USA
| | - Francesco J DeMayo
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, Durham, NC, 27709, USA
| | - Jianming Xu
- Department of Molecular Biology, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, USA
- Genetically Engineered Mouse Core, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, USA
| | - Mary E Dickinson
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, USA
| | - Arthur L Beaudet
- Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, MS BCM225, Houston, TX, 77030, USA
| | - Jason D Heaney
- Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, MS BCM225, Houston, TX, 77030, USA.
- Mouse ES Cell Core, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, USA.
| |
Collapse
|
41
|
Doe B, Brown E, Boroviak K. Generating CRISPR/Cas9-Derived Mutant Mice by Zygote Cytoplasmic Injection Using an Automatic Microinjector. Methods Protoc 2018; 1:mps1010005. [PMID: 31164552 PMCID: PMC6526459 DOI: 10.3390/mps1010005] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Revised: 01/04/2018] [Accepted: 01/04/2018] [Indexed: 01/04/2023] Open
Abstract
Clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated (Cas) assisted generation of mutant animals has become the method of choice for the elucidation of gene function in development and disease due to the shortened timelines for generation of a desired mutant, the ease of producing materials in comparison to other methodologies (such as embryonic stem cells, ESCs) and the ability to simultaneously target multiple genes in one injection session. Here we describe a step by step protocol, from preparation of materials through to injection and validation of a cytoplasmic injection, which can be used to generate CRISPR mutants. This can be accomplished from start of injection to completion within 2–4 h with high survival and developmental rates of injected zygotes and offers significant advantages over pronuclear and other previously described methodologies for microinjection.
Collapse
Affiliation(s)
- Brendan Doe
- Wellcome Trust Sanger Institute, Hinxton, CB10 1SA, UK.
| | - Ellen Brown
- Wellcome Trust Sanger Institute, Hinxton, CB10 1SA, UK.
| | | |
Collapse
|
42
|
Fuchs H, Aguilar-Pimentel JA, Amarie OV, Becker L, Calzada-Wack J, Cho YL, Garrett L, Hölter SM, Irmler M, Kistler M, Kraiger M, Mayer-Kuckuk P, Moreth K, Rathkolb B, Rozman J, da Silva Buttkus P, Treise I, Zimprich A, Gampe K, Hutterer C, Stöger C, Leuchtenberger S, Maier H, Miller M, Scheideler A, Wu M, Beckers J, Bekeredjian R, Brielmeier M, Busch DH, Klingenspor M, Klopstock T, Ollert M, Schmidt-Weber C, Stöger T, Wolf E, Wurst W, Yildirim AÖ, Zimmer A, Gailus-Durner V, Hrabě de Angelis M. Understanding gene functions and disease mechanisms: Phenotyping pipelines in the German Mouse Clinic. Behav Brain Res 2017; 352:187-196. [PMID: 28966146 DOI: 10.1016/j.bbr.2017.09.048] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 09/18/2017] [Accepted: 09/25/2017] [Indexed: 02/07/2023]
Abstract
Since decades, model organisms have provided an important approach for understanding the mechanistic basis of human diseases. The German Mouse Clinic (GMC) was the first phenotyping facility that established a collaboration-based platform for phenotype characterization of mouse lines. In order to address individual projects by a tailor-made phenotyping strategy, the GMC advanced in developing a series of pipelines with tests for the analysis of specific disease areas. For a general broad analysis, there is a screening pipeline that covers the key parameters for the most relevant disease areas. For hypothesis-driven phenotypic analyses, there are thirteen additional pipelines with focus on neurological and behavioral disorders, metabolic dysfunction, respiratory system malfunctions, immune-system disorders and imaging techniques. In this article, we give an overview of the pipelines and describe the scientific rationale behind the different test combinations.
Collapse
Affiliation(s)
- Helmut Fuchs
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter-Landstr. 1, 85764 Neuherberg, Germany
| | - Juan Antonio Aguilar-Pimentel
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter-Landstr. 1, 85764 Neuherberg, Germany
| | - Oana V Amarie
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter-Landstr. 1, 85764 Neuherberg, Germany; Institute of Developmental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter-Landstr. 1, 85764 Neuherberg, Germany
| | - Lore Becker
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter-Landstr. 1, 85764 Neuherberg, Germany
| | - Julia Calzada-Wack
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter-Landstr. 1, 85764 Neuherberg, Germany
| | - Yi-Li Cho
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter-Landstr. 1, 85764 Neuherberg, Germany
| | - Lillian Garrett
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter-Landstr. 1, 85764 Neuherberg, Germany; Institute of Developmental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter-Landstr. 1, 85764 Neuherberg, Germany
| | - Sabine M Hölter
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter-Landstr. 1, 85764 Neuherberg, Germany; Institute of Developmental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter-Landstr. 1, 85764 Neuherberg, Germany
| | - Martin Irmler
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter-Landstr. 1, 85764 Neuherberg, Germany
| | - Martin Kistler
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter-Landstr. 1, 85764 Neuherberg, Germany
| | - Markus Kraiger
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter-Landstr. 1, 85764 Neuherberg, Germany
| | - Philipp Mayer-Kuckuk
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter-Landstr. 1, 85764 Neuherberg, Germany
| | - Kristin Moreth
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter-Landstr. 1, 85764 Neuherberg, Germany
| | - Birgit Rathkolb
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter-Landstr. 1, 85764 Neuherberg, Germany; German Center for Diabetes Research (DZD), Ingolstädter-Landstr. 1, 85764 Neuherberg, Germany; Institute of Molecular Animal Breeding and Biotechnology, Gene Center, Ludwig-Maximilians-University München, Feodor-Lynen-Str. 25, 81377 Munich, Germany
| | - Jan Rozman
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter-Landstr. 1, 85764 Neuherberg, Germany; German Center for Diabetes Research (DZD), Ingolstädter-Landstr. 1, 85764 Neuherberg, Germany
| | - Patricia da Silva Buttkus
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter-Landstr. 1, 85764 Neuherberg, Germany
| | - Irina Treise
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter-Landstr. 1, 85764 Neuherberg, Germany
| | - Annemarie Zimprich
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter-Landstr. 1, 85764 Neuherberg, Germany; Institute of Developmental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter-Landstr. 1, 85764 Neuherberg, Germany
| | - Kristine Gampe
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter-Landstr. 1, 85764 Neuherberg, Germany
| | - Christine Hutterer
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter-Landstr. 1, 85764 Neuherberg, Germany
| | - Claudia Stöger
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter-Landstr. 1, 85764 Neuherberg, Germany
| | - Stefanie Leuchtenberger
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter-Landstr. 1, 85764 Neuherberg, Germany
| | - Holger Maier
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter-Landstr. 1, 85764 Neuherberg, Germany
| | - Manuel Miller
- Research Unit Comparative Medicine, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter-Landstr. 1, 85764 Neuherberg, Germany
| | - Angelika Scheideler
- Research Unit Comparative Medicine, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter-Landstr. 1, 85764 Neuherberg, Germany
| | - Moya Wu
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter-Landstr. 1, 85764 Neuherberg, Germany
| | - Johannes Beckers
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter-Landstr. 1, 85764 Neuherberg, Germany; German Center for Diabetes Research (DZD), Ingolstädter-Landstr. 1, 85764 Neuherberg, Germany; Chair of Experimental Genetics, School of Life Science Weihenstephan, Technische Universität München, Alte Akademie 8, 85354 Freising, Germany
| | - Raffi Bekeredjian
- Department of Cardiology, University of Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Markus Brielmeier
- Research Unit Comparative Medicine, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter-Landstr. 1, 85764 Neuherberg, Germany
| | - Dirk H Busch
- Institute for Medical Microbiology, Immunology and Hygiene, Technische Universität München, Trogerstr. 30, 81675 Munich, Germany
| | - Martin Klingenspor
- Chair of Molecular Nutritional Medicine, Technical University Munich, EKFZ - Else Kröner Fresenius Center for Nutritional Medicine, Gregor-Mendel-Str. 2, 85350 Freising-Weihenstephan, Germany; ZIEL - Institute for Food and Health, Technical University Munich, Gregor-Mendel-Str. 2, 85350 Freising-Weihenstephan, Germany
| | - Thomas Klopstock
- Department of Neurology, Friedrich-Baur-Institute, Klinikum der Ludwig-Maximilians-Universität München, Ziemssenstr. 1a, 80336 Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), 80336 Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Adolf-Butenandt-Institut, Ludwig-Maximilians-Universität München, Schillerstr. 44, 80336 Munich, Germany; German Center for Vertigo and Balance Disorders, 81377 Munich, Germany
| | - Markus Ollert
- Department of Infection and Immunity, Luxembourg Institute of Health, 29, Rue Henri Koch, 4354 Esch-sur-Alzette, Luxembourg; Department of Dermatology and Allergy Center, Odense Research Center for Anaphylaxis, University of Southern Denmark, 5000 Odense C, Denmark
| | - Carsten Schmidt-Weber
- Center of Allergy & Environment (ZAUM), Technische Universität München, and Helmholtz Zentrum München, Ingolstädter-Landstr., 85764 Neuherberg, Germany
| | - Tobias Stöger
- Institute of Lung Biology and Disease, Member of the German Center for Lung Research (DZL), Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter-Landstr. 1, D-85764 Neuherberg, Germany
| | - Eckhard Wolf
- Institute of Molecular Animal Breeding and Biotechnology, Gene Center, Ludwig-Maximilians-University München, Feodor-Lynen-Str. 25, 81377 Munich, Germany
| | - Wolfgang Wurst
- Institute of Developmental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter-Landstr. 1, 85764 Neuherberg, Germany; German Center for Neurodegenerative Diseases (DZNE), 80336 Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Adolf-Butenandt-Institut, Ludwig-Maximilians-Universität München, Schillerstr. 44, 80336 Munich, Germany; Chair of Developmental Genetics, Technische Universität München Freising-Weihenstephan, c/o Helmholtz Zentrum München Ingolstädter-Landstr. 1, 85764 Neuherberg, Germany
| | - Ali Önder Yildirim
- Institute of Lung Biology and Disease, Member of the German Center for Lung Research (DZL), Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter-Landstr. 1, D-85764 Neuherberg, Germany
| | - Andreas Zimmer
- Institute of Molecular Psychiatry, Medical Faculty, University of Bonn, Sigmund Freud Str. 25, 53127 Bonn, Germany
| | - Valérie Gailus-Durner
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter-Landstr. 1, 85764 Neuherberg, Germany
| | - Martin Hrabě de Angelis
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter-Landstr. 1, 85764 Neuherberg, Germany; German Center for Diabetes Research (DZD), Ingolstädter-Landstr. 1, 85764 Neuherberg, Germany; Chair of Experimental Genetics, School of Life Science Weihenstephan, Technische Universität München, Alte Akademie 8, 85354 Freising, Germany.
| |
Collapse
|
43
|
Abstract
Improvements to RFID technology are making it an increasingly flexible tool for tracking genetically modified rodents, characterizing their natural behaviors, and improving data integrity.
Collapse
|
44
|
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: 6.9] [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
|
45
|
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: 162] [Impact Index Per Article: 23.1] [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
|
46
|
Hai T, Cao C, Shang H, Guo W, Mu Y, Yang S, Zhang Y, Zheng Q, Zhang T, Wang X, Liu Y, Kong Q, Li K, Wang D, Qi M, Hong Q, Zhang R, Wang X, Jia Q, Wang X, Qin G, Li Y, Luo A, Jin W, Yao J, Huang J, Zhang H, Li M, Xie X, Zheng X, Guo K, Wang Q, Zhang S, Li L, Xie F, Zhang Y, Weng X, Yin Z, Hu K, Cong Y, Zheng P, Zou H, Xin L, Xia J, Ruan J, Li H, Zhao W, Yuan J, Liu Z, Gu W, Li M, Wang Y, Wang H, Yang S, Liu Z, Wei H, Zhao J, Zhou Q, Meng A. Pilot study of large-scale production of mutant pigs by ENU mutagenesis. eLife 2017. [PMID: 28639938 PMCID: PMC5505698 DOI: 10.7554/elife.26248] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
N-ethyl-N-nitrosourea (ENU) mutagenesis is a powerful tool to generate mutants on a large scale efficiently, and to discover genes with novel functions at the whole-genome level in Caenorhabditis elegans, flies, zebrafish and mice, but it has never been tried in large model animals. We describe a successful systematic three-generation ENU mutagenesis screening in pigs with the establishment of the Chinese Swine Mutagenesis Consortium. A total of 6,770 G1 and 6,800 G3 pigs were screened, 36 dominant and 91 recessive novel pig families with various phenotypes were established. The causative mutations in 10 mutant families were further mapped. As examples, the mutation of SOX10 (R109W) in pig causes inner ear malfunctions and mimics human Mondini dysplasia, and upregulated expression of FBXO32 is associated with congenital splay legs. This study demonstrates the feasibility of artificial random mutagenesis in pigs and opens an avenue for generating a reservoir of mutants for agricultural production and biomedical research. DOI:http://dx.doi.org/10.7554/eLife.26248.001
Collapse
Affiliation(s)
- Tang Hai
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,Chinese Swine Mutagenesis Consortium Working Group, Chinese Swine Mutagenesis Consortium, Beijing, China
| | - Chunwei Cao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,Chinese Swine Mutagenesis Consortium Working Group, Chinese Swine Mutagenesis Consortium, Beijing, China
| | - Haitao Shang
- Chinese Swine Mutagenesis Consortium Working Group, Chinese Swine Mutagenesis Consortium, Beijing, China.,Department of Laboratory Animal Science, College of Basic Medicine, Third Military Medical University, Chongqing, China
| | - Weiwei Guo
- Department of Otolaryngology-Head and Neck Surgery, Institute of Otolaryngology, Chinese PLA General Hospital, Beijing, China
| | - Yanshuang Mu
- Chinese Swine Mutagenesis Consortium Working Group, Chinese Swine Mutagenesis Consortium, Beijing, China.,College of Life Science, Northeast Agricultural University of China, Harbin, China
| | - Shulin Yang
- Chinese Swine Mutagenesis Consortium Working Group, Chinese Swine Mutagenesis Consortium, Beijing, China.,Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Ying Zhang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,Chinese Swine Mutagenesis Consortium Working Group, Chinese Swine Mutagenesis Consortium, Beijing, China
| | - Qiantao Zheng
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,Chinese Swine Mutagenesis Consortium Working Group, Chinese Swine Mutagenesis Consortium, Beijing, China
| | - Tao Zhang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,Chinese Swine Mutagenesis Consortium Working Group, Chinese Swine Mutagenesis Consortium, Beijing, China
| | - Xianlong Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,Chinese Swine Mutagenesis Consortium Working Group, Chinese Swine Mutagenesis Consortium, Beijing, China
| | - Yu Liu
- Chinese Swine Mutagenesis Consortium Working Group, Chinese Swine Mutagenesis Consortium, Beijing, China.,Department of Laboratory Animal Science, College of Basic Medicine, Third Military Medical University, Chongqing, China
| | - Qingran Kong
- Chinese Swine Mutagenesis Consortium Working Group, Chinese Swine Mutagenesis Consortium, Beijing, China.,College of Life Science, Northeast Agricultural University of China, Harbin, China
| | - Kui Li
- Chinese Swine Mutagenesis Consortium Working Group, Chinese Swine Mutagenesis Consortium, Beijing, China.,Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Dayu Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,Chinese Swine Mutagenesis Consortium Working Group, Chinese Swine Mutagenesis Consortium, Beijing, China
| | - Meng Qi
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,Chinese Swine Mutagenesis Consortium Working Group, Chinese Swine Mutagenesis Consortium, Beijing, China
| | - Qianlong Hong
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,Chinese Swine Mutagenesis Consortium Working Group, Chinese Swine Mutagenesis Consortium, Beijing, China
| | - Rui Zhang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,Chinese Swine Mutagenesis Consortium Working Group, Chinese Swine Mutagenesis Consortium, Beijing, China
| | - Xiupeng Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,Chinese Swine Mutagenesis Consortium Working Group, Chinese Swine Mutagenesis Consortium, Beijing, China
| | - Qitao Jia
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,Chinese Swine Mutagenesis Consortium Working Group, Chinese Swine Mutagenesis Consortium, Beijing, China
| | - Xiao Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,Chinese Swine Mutagenesis Consortium Working Group, Chinese Swine Mutagenesis Consortium, Beijing, China
| | - Guosong Qin
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,Chinese Swine Mutagenesis Consortium Working Group, Chinese Swine Mutagenesis Consortium, Beijing, China
| | - Yongshun Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,Chinese Swine Mutagenesis Consortium Working Group, Chinese Swine Mutagenesis Consortium, Beijing, China
| | - Ailing Luo
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,Chinese Swine Mutagenesis Consortium Working Group, Chinese Swine Mutagenesis Consortium, Beijing, China
| | - Weiwu Jin
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,Chinese Swine Mutagenesis Consortium Working Group, Chinese Swine Mutagenesis Consortium, Beijing, China
| | - Jing Yao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,Chinese Swine Mutagenesis Consortium Working Group, Chinese Swine Mutagenesis Consortium, Beijing, China
| | - Jiaojiao Huang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,Chinese Swine Mutagenesis Consortium Working Group, Chinese Swine Mutagenesis Consortium, Beijing, China
| | - Hongyong Zhang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,Chinese Swine Mutagenesis Consortium Working Group, Chinese Swine Mutagenesis Consortium, Beijing, China
| | - Menghua Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,Chinese Swine Mutagenesis Consortium Working Group, Chinese Swine Mutagenesis Consortium, Beijing, China
| | - Xiangmo Xie
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,Chinese Swine Mutagenesis Consortium Working Group, Chinese Swine Mutagenesis Consortium, Beijing, China
| | - Xuejuan Zheng
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,Chinese Swine Mutagenesis Consortium Working Group, Chinese Swine Mutagenesis Consortium, Beijing, China
| | - Kenan Guo
- Chinese Swine Mutagenesis Consortium Working Group, Chinese Swine Mutagenesis Consortium, Beijing, China.,Department of Laboratory Animal Science, College of Basic Medicine, Third Military Medical University, Chongqing, China
| | - Qinghua Wang
- Chinese Swine Mutagenesis Consortium Working Group, Chinese Swine Mutagenesis Consortium, Beijing, China.,Department of Laboratory Animal Science, College of Basic Medicine, Third Military Medical University, Chongqing, China
| | - Shibin Zhang
- Chinese Swine Mutagenesis Consortium Working Group, Chinese Swine Mutagenesis Consortium, Beijing, China.,Department of Laboratory Animal Science, College of Basic Medicine, Third Military Medical University, Chongqing, China
| | - Liang Li
- Chinese Swine Mutagenesis Consortium Working Group, Chinese Swine Mutagenesis Consortium, Beijing, China.,Department of Laboratory Animal Science, College of Basic Medicine, Third Military Medical University, Chongqing, China
| | - Fei Xie
- Chinese Swine Mutagenesis Consortium Working Group, Chinese Swine Mutagenesis Consortium, Beijing, China.,Department of Laboratory Animal Science, College of Basic Medicine, Third Military Medical University, Chongqing, China
| | - Yu Zhang
- Chinese Swine Mutagenesis Consortium Working Group, Chinese Swine Mutagenesis Consortium, Beijing, China.,College of Life Science, Northeast Agricultural University of China, Harbin, China
| | - Xiaogang Weng
- Chinese Swine Mutagenesis Consortium Working Group, Chinese Swine Mutagenesis Consortium, Beijing, China.,College of Life Science, Northeast Agricultural University of China, Harbin, China
| | - Zhi Yin
- Chinese Swine Mutagenesis Consortium Working Group, Chinese Swine Mutagenesis Consortium, Beijing, China.,College of Life Science, Northeast Agricultural University of China, Harbin, China
| | - Kui Hu
- Chinese Swine Mutagenesis Consortium Working Group, Chinese Swine Mutagenesis Consortium, Beijing, China.,College of Life Science, Northeast Agricultural University of China, Harbin, China
| | - Yimei Cong
- Chinese Swine Mutagenesis Consortium Working Group, Chinese Swine Mutagenesis Consortium, Beijing, China.,College of Life Science, Northeast Agricultural University of China, Harbin, China
| | - Peng Zheng
- Chinese Swine Mutagenesis Consortium Working Group, Chinese Swine Mutagenesis Consortium, Beijing, China.,College of Life Science, Northeast Agricultural University of China, Harbin, China
| | - Hailong Zou
- Chinese Swine Mutagenesis Consortium Working Group, Chinese Swine Mutagenesis Consortium, Beijing, China.,College of Life Science, Northeast Agricultural University of China, Harbin, China
| | - Leilei Xin
- Chinese Swine Mutagenesis Consortium Working Group, Chinese Swine Mutagenesis Consortium, Beijing, China.,Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Jihan Xia
- Chinese Swine Mutagenesis Consortium Working Group, Chinese Swine Mutagenesis Consortium, Beijing, China.,Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Jinxue Ruan
- Chinese Swine Mutagenesis Consortium Working Group, Chinese Swine Mutagenesis Consortium, Beijing, China.,Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Hegang Li
- Chinese Swine Mutagenesis Consortium Working Group, Chinese Swine Mutagenesis Consortium, Beijing, China.,Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Weiming Zhao
- Chinese Swine Mutagenesis Consortium Working Group, Chinese Swine Mutagenesis Consortium, Beijing, China.,Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Jing Yuan
- Chinese Swine Mutagenesis Consortium Working Group, Chinese Swine Mutagenesis Consortium, Beijing, China.,Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Zizhan Liu
- Chinese Swine Mutagenesis Consortium Working Group, Chinese Swine Mutagenesis Consortium, Beijing, China.,Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Weiwang Gu
- Chinese Swine Mutagenesis Consortium Working Group, Chinese Swine Mutagenesis Consortium, Beijing, China.,Pearl Laboratory Animal Sci. & Tech. Co. Ltd, Guangzhou, China
| | - Ming Li
- Chinese Swine Mutagenesis Consortium Working Group, Chinese Swine Mutagenesis Consortium, Beijing, China.,Pearl Laboratory Animal Sci. & Tech. Co. Ltd, Guangzhou, China
| | - Yong Wang
- Chinese Swine Mutagenesis Consortium Working Group, Chinese Swine Mutagenesis Consortium, Beijing, China.,Department of Laboratory Animal Science, College of Basic Medicine, Third Military Medical University, Chongqing, China
| | - Hongmei Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,Chinese Swine Mutagenesis Consortium Guide Group, Chinese Swine Mutagenesis Consortium, Beijing, China
| | - Shiming Yang
- Department of Otolaryngology-Head and Neck Surgery, Institute of Otolaryngology, Chinese PLA General Hospital, Beijing, China.,Chinese Swine Mutagenesis Consortium Guide Group, Chinese Swine Mutagenesis Consortium, Beijing, China
| | - Zhonghua Liu
- College of Life Science, Northeast Agricultural University of China, Harbin, China.,Chinese Swine Mutagenesis Consortium Guide Group, Chinese Swine Mutagenesis Consortium, Beijing, China
| | - Hong Wei
- Department of Laboratory Animal Science, College of Basic Medicine, Third Military Medical University, Chongqing, China.,Chinese Swine Mutagenesis Consortium Guide Group, Chinese Swine Mutagenesis Consortium, Beijing, China
| | - Jianguo Zhao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,Chinese Swine Mutagenesis Consortium Guide Group, Chinese Swine Mutagenesis Consortium, Beijing, China
| | - Qi Zhou
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,Chinese Swine Mutagenesis Consortium Guide Group, Chinese Swine Mutagenesis Consortium, Beijing, China
| | - Anming Meng
- Chinese Swine Mutagenesis Consortium Guide Group, Chinese Swine Mutagenesis Consortium, Beijing, China.,School of Life Sciences, Tsinghua University, Beijing, China
| |
Collapse
|
47
|
Abstract
Organism-level systems biology in mammals aims to identify, analyze, control, and design molecular and cellular networks executing various biological functions in mammals. In particular, system-level identification and analysis of molecular and cellular networks can be accelerated by next-generation mammalian genetics. Mammalian genetics without crossing, where all production and phenotyping studies of genome-edited animals are completed within a single generation drastically reduce the time, space, and effort of conducting the systems research. Next-generation mammalian genetics is based on recent technological advancements in genome editing and developmental engineering. The process begins with introduction of double-strand breaks into genomic DNA by using site-specific endonucleases, which results in highly efficient genome editing in mammalian zygotes or embryonic stem cells. By using nuclease-mediated genome editing in zygotes, or ~100% embryonic stem cell-derived mouse technology, whole-body knock-out and knock-in mice can be produced within a single generation. These emerging technologies allow us to produce multiple knock-out or knock-in strains in high-throughput manner. In this review, we discuss the basic concepts and related technologies as well as current challenges and future opportunities for next-generation mammalian genetics in organism-level systems biology.
Collapse
Affiliation(s)
- Etsuo A Susaki
- Department of Systems Pharmacology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, , Bunkyo-ku, Tokyo 113-0033 Japan.,Laboratory for Synthetic Biology, RIKEN Quantitative Biology Center, 1-3 Yamadaoka, , Suita, Osaka 565-0871 Japan.,PRESTO, Japan Science and Technology Agency (JST), 4-1-8 Honcho, , Kawaguchi, Saitama 332-0012 Japan
| | - Hideki Ukai
- Laboratory for Synthetic Biology, RIKEN Quantitative Biology Center, 1-3 Yamadaoka, , Suita, Osaka 565-0871 Japan
| | - Hiroki R Ueda
- Department of Systems Pharmacology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, , Bunkyo-ku, Tokyo 113-0033 Japan.,Laboratory for Synthetic Biology, RIKEN Quantitative Biology Center, 1-3 Yamadaoka, , Suita, Osaka 565-0871 Japan
| |
Collapse
|
48
|
Zimprich A, Östereicher MA, Becker L, Dirscherl P, Ernst L, Fuchs H, Gailus-Durner V, Garrett L, Giesert F, Glasl L, Hummel A, Rozman J, de Angelis MH, Vogt-Weisenhorn D, Wurst W, Hölter SM. Analysis of locomotor behavior in the German Mouse Clinic. J Neurosci Methods 2017; 300:77-91. [PMID: 28483715 DOI: 10.1016/j.jneumeth.2017.05.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 05/04/2017] [Accepted: 05/05/2017] [Indexed: 10/19/2022]
Abstract
BACKGROUND Generation and phenotyping of mutant mouse models continues to increase along with the search for the most efficient phenotyping tests. Here we asked if a combination of different locomotor tests is necessary for comprehensive locomotor phenotyping, or if a large data set from an automated gait analysis with the CatWalk system would suffice. NEW METHOD First we endeavored to meaningfully reduce the large CatWalk data set by Principal Component Analysis (PCA) to decide on the most relevant parameters. We analyzed the influence of sex, body weight, genetic background and age. Then a combination of different locomotor tests was analyzed to investigate the possibility of redundancy between tests. RESULT The extracted 10 components describe 80% of the total variance in the CatWalk, characterizing different aspects of gait. With these, effects of CatWalk version, sex, body weight, age and genetic background were detected. In addition, the PCA on a combination of locomotor tests suggests that these are independent without significant redundancy in their locomotor measures. COMPARISON WITH EXISTING METHODS The PCA has permitted the refinement of the highly dimensional CatWalk (and other tests) data set for the extraction of individual component scores and subsequent analysis. CONCLUSION The outcome of the PCA suggests the possibility to focus on measures of the front and hind paws, and one measure of coordination in future experiments to detect phenotypic differences. Furthermore, although the CatWalk is sensitive for detecting locomotor phenotypes pertaining to gait, it is necessary to include other tests for comprehensive locomotor phenotyping.
Collapse
Affiliation(s)
- Annemarie Zimprich
- Developmental Genetics, Technische Universität München-Weihenstephan, c/o Helmholtz Zentrum München, Ingolstädter Landstr. 1, 85764 Neuherberg, Germany; Institute of Developmental Genetics, Helmholtz Zentrum München, Ingolstaedter Landstr.1, 85764 Neuherberg, Germany.
| | - Manuela A Östereicher
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, Ingolstaedter Landstr. 1, 85764 Neuherberg, Germany
| | - Lore Becker
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, Ingolstaedter Landstr. 1, 85764 Neuherberg, Germany
| | - Petra Dirscherl
- Institute of Developmental Genetics, Helmholtz Zentrum München, Ingolstaedter Landstr.1, 85764 Neuherberg, Germany
| | - Luise Ernst
- Developmental Genetics, Technische Universität München-Weihenstephan, c/o Helmholtz Zentrum München, Ingolstädter Landstr. 1, 85764 Neuherberg, Germany; Institute of Developmental Genetics, Helmholtz Zentrum München, Ingolstaedter Landstr.1, 85764 Neuherberg, Germany
| | - Helmut Fuchs
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, Ingolstaedter Landstr. 1, 85764 Neuherberg, Germany
| | - Valerie Gailus-Durner
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, Ingolstaedter Landstr. 1, 85764 Neuherberg, Germany
| | - Lillian Garrett
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, Ingolstaedter Landstr. 1, 85764 Neuherberg, Germany; Institute of Developmental Genetics, Helmholtz Zentrum München, Ingolstaedter Landstr.1, 85764 Neuherberg, Germany
| | - Florian Giesert
- Developmental Genetics, Technische Universität München-Weihenstephan, c/o Helmholtz Zentrum München, Ingolstädter Landstr. 1, 85764 Neuherberg, Germany; Institute of Developmental Genetics, Helmholtz Zentrum München, Ingolstaedter Landstr.1, 85764 Neuherberg, Germany
| | - Lisa Glasl
- Institute of Developmental Genetics, Helmholtz Zentrum München, Ingolstaedter Landstr.1, 85764 Neuherberg, Germany
| | - Angelika Hummel
- Institute of Developmental Genetics, Helmholtz Zentrum München, Ingolstaedter Landstr.1, 85764 Neuherberg, Germany
| | - Jan Rozman
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, Ingolstaedter Landstr. 1, 85764 Neuherberg, Germany; German Center for Diabetes Research (DZD), Helmholtz Zentrum München, Ingolstaedter Landstr. 1, 85764 Neuherberg, Germany
| | - Martin Hrabě de Angelis
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, Ingolstaedter Landstr. 1, 85764 Neuherberg, Germany; German Center for Diabetes Research (DZD), Helmholtz Zentrum München, Ingolstaedter Landstr. 1, 85764 Neuherberg, Germany; Chair of Experimental Genetics, Technische Universität München-Weihenstephan, c/o Helmholtz Zentrum München, Ingolstädter Landstr. 1, 85764 Neuherberg, Germany
| | - Daniela Vogt-Weisenhorn
- Institute of Developmental Genetics, Helmholtz Zentrum München, Ingolstaedter Landstr.1, 85764 Neuherberg, Germany
| | - Wolfgang Wurst
- Institute of Developmental Genetics, Helmholtz Zentrum München, Ingolstaedter Landstr.1, 85764 Neuherberg, Germany; Chair of Developmental Genetics, Technische Universität München-Weihenstephan, c/o Helmholtz Zentrum München, Ingolstädter Landstr. 1, 85764 Neuherberg, Germany; German Center for Neurodegenerative Diseases (DZNE), Site Munich, Feodor-Lynen-Str. 17, 81377 Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Adolf-Butenandt-Institut, Ludwig-Maximilians-Universität München, Schillerstr. 44, 80336 Munich, Germany
| | - Sabine M Hölter
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, Ingolstaedter Landstr. 1, 85764 Neuherberg, Germany; Institute of Developmental Genetics, Helmholtz Zentrum München, Ingolstaedter Landstr.1, 85764 Neuherberg, Germany
| |
Collapse
|
49
|
Human knockouts and phenotypic analysis in a cohort with a high rate of consanguinity. Nature 2017; 544:235-239. [PMID: 28406212 PMCID: PMC5600291 DOI: 10.1038/nature22034] [Citation(s) in RCA: 230] [Impact Index Per Article: 32.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Accepted: 03/05/2017] [Indexed: 02/02/2023]
Abstract
A major goal of biomedicine is to understand the function of every gene in the human genome. Loss-of-function mutations can disrupt both copies of a given gene in humans and phenotypic analysis of such 'human knockouts' can provide insight into gene function. Consanguineous unions are more likely to result in offspring carrying homozygous loss-of-function mutations. In Pakistan, consanguinity rates are notably high. Here we sequence the protein-coding regions of 10,503 adult participants in the Pakistan Risk of Myocardial Infarction Study (PROMIS), designed to understand the determinants of cardiometabolic diseases in individuals from South Asia. We identified individuals carrying homozygous predicted loss-of-function (pLoF) mutations, and performed phenotypic analysis involving more than 200 biochemical and disease traits. We enumerated 49,138 rare (<1% minor allele frequency) pLoF mutations. These pLoF mutations are estimated to knock out 1,317 genes, each in at least one participant. Homozygosity for pLoF mutations at PLA2G7 was associated with absent enzymatic activity of soluble lipoprotein-associated phospholipase A2; at CYP2F1, with higher plasma interleukin-8 concentrations; at TREH, with lower concentrations of apoB-containing lipoprotein subfractions; at either A3GALT2 or NRG4, with markedly reduced plasma insulin C-peptide concentrations; and at SLC9A3R1, with mediators of calcium and phosphate signalling. Heterozygous deficiency of APOC3 has been shown to protect against coronary heart disease; we identified APOC3 homozygous pLoF carriers in our cohort. We recruited these human knockouts and challenged them with an oral fat load. Compared with family members lacking the mutation, individuals with APOC3 knocked out displayed marked blunting of the usual post-prandial rise in plasma triglycerides. Overall, these observations provide a roadmap for a 'human knockout project', a systematic effort to understand the phenotypic consequences of complete disruption of genes in humans.
Collapse
|
50
|
Ingvorsen C, Karp NA, Lelliott CJ. The role of sex and body weight on the metabolic effects of high-fat diet in C57BL/6N mice. Nutr Diabetes 2017; 7:e261. [PMID: 28394359 PMCID: PMC5436097 DOI: 10.1038/nutd.2017.6] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Accepted: 01/11/2017] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Metabolic disorders are commonly investigated using knockout and transgenic mouse models on the C57BL/6N genetic background due to its genetic susceptibility to the deleterious metabolic effects of high-fat diet (HFD). There is growing awareness of the need to consider sex in disease progression, but limited attention has been paid to sexual dimorphism in mouse models and its impact in metabolic phenotypes. We assessed the effect of HFD and the impact of sex on metabolic variables in this strain. METHODS We generated a reference data set encompassing glucose tolerance, body composition and plasma chemistry data from 586 C57BL/6N mice fed a standard chow and 733 fed a HFD collected as part of a high-throughput phenotyping pipeline. Linear mixed model regression analysis was used in a dual analysis to assess the effect of HFD as an absolute change in phenotype, but also as a relative change accounting for the potential confounding effect of body weight. RESULTS HFD had a significant impact on all variables tested with an average absolute effect size of 29%. For the majority of variables (78%), the treatment effect was modified by sex and this was dominated by male-specific or a male stronger effect. On average, there was a 13.2% difference in the effect size between the male and female mice for sexually dimorphic variables. HFD led to a significant body weight phenotype (24% increase), which acts as a confounding effect on the other analysed variables. For 79% of the variables, body weight was found to be a significant source of variation, but even after accounting for this confounding effect, similar HFD-induced phenotypic changes were found to when not accounting for weight. CONCLUSION HFD and sex are powerful modifiers of metabolic parameters in C57BL/6N mice. We also demonstrate the value of considering body size as a covariate to obtain a richer understanding of metabolic phenotypes.
Collapse
Affiliation(s)
- C Ingvorsen
- Mouse Pipelines, Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Cambridge, UK
- University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Robinson Way, Cambridge, UK
| | - N A Karp
- Mouse Informatics Group, Wellcome Trust Sanger Institute, Cambridge, UK
| | - C J Lelliott
- Mouse Pipelines, Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Cambridge, UK
| |
Collapse
|