1
|
Nunley H, Shao B, Denberg D, Grover P, Singh J, Avdeeva M, Joyce B, Kim-Yip R, Kohrman A, Biswas A, Watters A, Gal Z, Kickuth A, Chalifoux M, Shvartsman SY, Brown LM, Posfai E. Nuclear instance segmentation and tracking for preimplantation mouse embryos. Development 2024; 151:dev202817. [PMID: 39373366 PMCID: PMC11574361 DOI: 10.1242/dev.202817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 09/23/2024] [Indexed: 10/08/2024]
Abstract
For investigations into fate specification and morphogenesis in time-lapse images of preimplantation embryos, automated 3D instance segmentation and tracking of nuclei are invaluable. Low signal-to-noise ratio, high voxel anisotropy, high nuclear density, and variable nuclear shapes can limit the performance of segmentation methods, while tracking is complicated by cell divisions, low frame rates, and sample movements. Supervised machine learning approaches can radically improve segmentation accuracy and enable easier tracking, but they often require large amounts of annotated 3D data. Here, we first report a previously unreported mouse line expressing near-infrared nuclear reporter H2B-miRFP720. We then generate a dataset (termed BlastoSPIM) of 3D images of H2B-miRFP720-expressing embryos with ground truth for nuclear instances. Using BlastoSPIM, we benchmark seven convolutional neural networks and identify Stardist-3D as the most accurate instance segmentation method. With our BlastoSPIM-trained Stardist-3D models, we construct a complete pipeline for nuclear instance segmentation and lineage tracking from the eight-cell stage to the end of preimplantation development (>100 nuclei). Finally, we demonstrate the usefulness of BlastoSPIM as pre-train data for related problems, both for a different imaging modality and for different model systems.
Collapse
Affiliation(s)
- Hayden Nunley
- Center for Computational Biology, Flatiron Institute - Simons Foundation, New York, NY 10010, USA
| | - Binglun Shao
- Center for Computational Biology, Flatiron Institute - Simons Foundation, New York, NY 10010, USA
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ 08544, USA
| | - David Denberg
- The Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
| | - Prateek Grover
- Center for Computational Biology, Flatiron Institute - Simons Foundation, New York, NY 10010, USA
| | - Jaspreet Singh
- Center for Computational Biology, Flatiron Institute - Simons Foundation, New York, NY 10010, USA
| | - Maria Avdeeva
- Center for Computational Biology, Flatiron Institute - Simons Foundation, New York, NY 10010, USA
| | - Bradley Joyce
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Rebecca Kim-Yip
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Abraham Kohrman
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Abhishek Biswas
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
- Research Computing, Princeton University, Princeton, NJ 08544, USA
| | - Aaron Watters
- Center for Computational Biology, Flatiron Institute - Simons Foundation, New York, NY 10010, USA
| | - Zsombor Gal
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Alison Kickuth
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Madeleine Chalifoux
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ 08544, USA
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Stanislav Y Shvartsman
- Center for Computational Biology, Flatiron Institute - Simons Foundation, New York, NY 10010, USA
- The Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Lisa M Brown
- Center for Computational Biology, Flatiron Institute - Simons Foundation, New York, NY 10010, USA
| | - Eszter Posfai
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| |
Collapse
|
2
|
Xie H, Linning-Duffy K, Demireva EY, Toh H, Abolibdeh B, Shi J, Zhou B, Iwase S, Yan L. CRISPR-based genome editing of a diurnal rodent, Nile grass rat (Arvicanthis niloticus). BMC Biol 2024; 22:144. [PMID: 38956550 PMCID: PMC11218167 DOI: 10.1186/s12915-024-01943-9] [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: 11/28/2023] [Accepted: 06/21/2024] [Indexed: 07/04/2024] Open
Abstract
BACKGROUND Diurnal and nocturnal mammals have evolved distinct pathways to optimize survival for their chronotype-specific lifestyles. Conventional rodent models, being nocturnal, may not sufficiently recapitulate the biology of diurnal humans in health and disease. Although diurnal rodents are potentially advantageous for translational research, until recently, they have not been genetically tractable. The present study aims to address this major limitation by developing experimental procedures necessary for genome editing in a well-established diurnal rodent model, the Nile grass rat (Arvicanthis niloticus). RESULTS A superovulation protocol was established, which yielded nearly 30 eggs per female grass rat. Fertilized eggs were cultured in a modified rat 1-cell embryo culture medium (mR1ECM), in which grass rat embryos developed from the 1-cell stage into blastocysts. A CRISPR-based approach was then used for gene editing in vivo and in vitro, targeting Retinoic acid-induced 1 (Rai1), the causal gene for Smith-Magenis Syndrome, a neurodevelopmental disorder. The CRISPR reagents were delivered in vivo by electroporation using an improved Genome-editing via Oviductal Nucleic Acids Delivery (i-GONAD) method. The in vivo approach produced several edited founder grass rats with Rai1 null mutations, which showed stable transmission of the targeted allele to the next generation. CRISPR reagents were also microinjected into 2-cell embryos in vitro. Large deletion of the Rai1 gene was confirmed in 70% of the embryos injected, demonstrating high-efficiency genome editing in vitro. CONCLUSION We have established a set of methods that enabled the first successful CRISPR-based genome editing in Nile grass rats. The methods developed will guide future genome editing of this and other diurnal rodent species, which will promote greater utility of these models in basic and translational research.
Collapse
Affiliation(s)
- Huirong Xie
- Transgenic and Genome Editing Facility, Institute for Quantitative Health Science & Engineering, Research Technology Support Facility, Michigan State University, East Lansing, MI, 48824, USA.
| | | | - Elena Y Demireva
- Transgenic and Genome Editing Facility, Institute for Quantitative Health Science & Engineering, Research Technology Support Facility, Michigan State University, East Lansing, MI, 48824, USA
| | - Huishi Toh
- Neuroscience Research Institute, University of California Santa Barbara, Santa Barbara, USA
| | - Bana Abolibdeh
- Transgenic and Genome Editing Facility, Institute for Quantitative Health Science & Engineering, Research Technology Support Facility, Michigan State University, East Lansing, MI, 48824, USA
| | - Jiaming Shi
- Department of Psychology, Michigan State University, East Lansing, MI, 48824, USA
| | - Bo Zhou
- Department of Human Genetics, University of Michigan Medical School, Ann Arbor, USA
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, USA
| | - Shigeki Iwase
- Department of Human Genetics, University of Michigan Medical School, Ann Arbor, USA
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, USA
| | - Lily Yan
- Department of Psychology, Michigan State University, East Lansing, MI, 48824, USA.
- Neuroscience Program, Michigan State University, East Lansing, USA.
| |
Collapse
|
3
|
Jiang D, Jiao L, Li Q, Xie R, Jia H, Wang S, Chen Y, Liu S, Huang D, Zheng J, Song W, Li Y, Chen J, Li J, Ying B, Yu L. Neutrophil-derived migrasomes are an essential part of the coagulation system. Nat Cell Biol 2024; 26:1110-1123. [PMID: 38997457 PMCID: PMC11251984 DOI: 10.1038/s41556-024-01440-9] [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: 03/13/2024] [Accepted: 05/14/2024] [Indexed: 07/14/2024]
Abstract
Migrasomes are organelles that are generated by migrating cells. Here we report the key role of neutrophil-derived migrasomes in haemostasis. We found that a large number of neutrophil-derived migrasomes exist in the blood of mice and humans. Compared with neutrophil cell bodies and platelets, these migrasomes adsorb and enrich coagulation factors on the surface. Moreover, they are highly enriched with adhesion molecules, which enable them to preferentially accumulate at sites of injury, where they trigger platelet activation and clot formation. Depletion of neutrophils, or genetic reduction of the number of these migrasomes, significantly decreases platelet plug formation and impairs coagulation. These defects can be rescued by intravenous injection of purified neutrophil-derived migrasomes. Our study reveals neutrophil-derived migrasomes as a previously unrecognized essential component of the haemostasis system, which may shed light on the cause of various coagulation disorders and open therapeutic possibilities.
Collapse
Affiliation(s)
- Dong Jiang
- State Key Laboratory of Membrane Biology, Tsinghua University-Peking University Joint Center for Life Sciences, Beijing Frontier Research Center for Biological Structure, School of Life Sciences, Tsinghua University, Beijing, China
| | - Lin Jiao
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Qing Li
- Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Renxiang Xie
- State Key Laboratory of Membrane Biology, Tsinghua University-Peking University Joint Center for Life Sciences, Beijing Frontier Research Center for Biological Structure, School of Life Sciences, Tsinghua University, Beijing, China
| | - Haohao Jia
- State Key Laboratory of Membrane Biology, Tsinghua University-Peking University Joint Center for Life Sciences, Beijing Frontier Research Center for Biological Structure, School of Life Sciences, Tsinghua University, Beijing, China
| | - ShiHui Wang
- State Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Yining Chen
- State Key Laboratory of Membrane Biology, Tsinghua University-Peking University Joint Center for Life Sciences, Beijing Frontier Research Center for Biological Structure, School of Life Sciences, Tsinghua University, Beijing, China
| | - Siyuan Liu
- State Key Laboratory of Membrane Biology, Tsinghua University-Peking University Joint Center for Life Sciences, Beijing Frontier Research Center for Biological Structure, School of Life Sciences, Tsinghua University, Beijing, China
| | - Dandan Huang
- State Key Laboratory of Membrane Biology, Tsinghua University-Peking University Joint Center for Life Sciences, Beijing Frontier Research Center for Biological Structure, School of Life Sciences, Tsinghua University, Beijing, China
| | - Jiajia Zheng
- Department of Laboratory Medicine, Peking University Third Hospital, Beijing, People's Republic of China
| | - Wenhao Song
- State Key Laboratory of Membrane Biology, Tsinghua University-Peking University Joint Center for Life Sciences, Beijing Frontier Research Center for Biological Structure, School of Life Sciences, Tsinghua University, Beijing, China
| | - Ying Li
- State Key Laboratory of Membrane Biology, Tsinghua University-Peking University Joint Center for Life Sciences, Beijing Frontier Research Center for Biological Structure, School of Life Sciences, Tsinghua University, Beijing, China
| | - JianFeng Chen
- State Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Jinsong Li
- Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Binwu Ying
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Li Yu
- State Key Laboratory of Membrane Biology, Tsinghua University-Peking University Joint Center for Life Sciences, Beijing Frontier Research Center for Biological Structure, School of Life Sciences, Tsinghua University, Beijing, China.
| |
Collapse
|
4
|
Maino E, Scott O, Rizvi SZ, Chan WS, Visuvanathan S, Zablah YB, Li H, Sengar AS, Salter MW, Jia Z, Rossant J, Cohn RD, Gu B, Ivakine EA. An Irak1-Mecp2 tandem duplication mouse model for the study of MECP2 duplication syndrome. Dis Model Mech 2024; 17:dmm050528. [PMID: 38881329 PMCID: PMC11552499 DOI: 10.1242/dmm.050528] [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: 10/16/2023] [Accepted: 06/06/2024] [Indexed: 06/18/2024] Open
Abstract
MECP2 duplication syndrome (MDS) is a neurodevelopmental disorder caused by tandem duplication of the MECP2 locus and its surrounding genes, including IRAK1. Current MDS mouse models involve transgenic expression of MECP2 only, limiting their applicability to the study of the disease. Herein, we show that an efficient and precise CRISPR/Cas9 fusion proximity-based approach can be utilized to generate an Irak1-Mecp2 tandem duplication mouse model ('Mecp2 Dup'). The Mecp2 Dup mouse model recapitulates the genomic landscape of human MDS by harboring a 160 kb tandem duplication encompassing Mecp2 and Irak1, representing the minimal disease-causing duplication, and the neighboring genes Opn1mw and Tex28. The Mecp2 Dup model exhibits neuro-behavioral abnormalities, and an abnormal immune response to infection not previously observed in other mouse models, possibly owing to Irak1 overexpression. The Mecp2 Dup model thus provides a tool to investigate MDS disease mechanisms and develop potential therapies applicable to patients.
Collapse
Affiliation(s)
- Eleonora Maino
- Program in Genetics and Genome Biology, the Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Ori Scott
- Program in Genetics and Genome Biology, the Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
- Division of Clinical Immunology and Allergy, Department of Pediatrics, the Hospital for Sick Children and University of Toronto, Toronto, ON M5G 1E8, Canada
| | - Samar Z. Rizvi
- Program in Genetics and Genome Biology, the Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Wing Suen Chan
- Program in Genetics and Genome Biology, the Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Shagana Visuvanathan
- Program in Genetics and Genome Biology, the Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Youssif Ben Zablah
- Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada
- Program in Neuroscience and Mental Health, the Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Hongbin Li
- Program in Neuroscience and Mental Health, the Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Ameet S. Sengar
- Program in Neuroscience and Mental Health, the Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Michael W. Salter
- Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada
- Program in Neuroscience and Mental Health, the Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Zhengping Jia
- Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada
- Program in Neuroscience and Mental Health, the Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Janet Rossant
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
- Program in Developmental and Stem Cell Biology, the Hospital for Sick Children Research Institute, Toronto, ON M5G 0A4, Canada
| | - Ronald D. Cohn
- Program in Genetics and Genome Biology, the Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
- Division of Clinical Immunology and Allergy, Department of Pediatrics, the Hospital for Sick Children and University of Toronto, Toronto, ON M5G 1E8, Canada
- Division of Clinical and Metabolic Genetics, Department of Pediatrics, the Hospital for Sick Children and University of Toronto, Toronto, ON M5G 1X8, Canada
| | - Bin Gu
- Department of Obstetrics, Gynecology and Reproductive Biology, Michigan State University, East Lansing, MI 48824, USA
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI 48824, USA
| | - Evgueni A. Ivakine
- Program in Genetics and Genome Biology, the Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
- Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| |
Collapse
|
5
|
Zheng D, Elnegiry AA, Luo C, Bendahou MA, Xie L, Bell D, Takahashi Y, Hanna E, Mias GI, Tsoi MF, Gu B. Brd4::Nutm1 fusion gene initiates NUT carcinoma in vivo. Life Sci Alliance 2024; 7:e202402602. [PMID: 38724194 PMCID: PMC11082452 DOI: 10.26508/lsa.202402602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 04/26/2024] [Accepted: 04/29/2024] [Indexed: 05/12/2024] Open
Abstract
NUT carcinoma (NC) is an aggressive cancer with no effective treatment. About 70% of NUT carcinoma is associated with chromosome translocation events that lead to the formation of a BRD4::NUTM1 fusion gene. Because the BRD4::NUTM1 gene is unequivocally cytotoxic when ectopically expressed in cell lines, questions remain on whether the fusion gene can initiate NC. Here, we report the first genetically engineered mouse model for NUT carcinoma that recapitulates the human t(15;19) chromosome translocation in mice. We demonstrated that the mouse t(2;17) syntenic chromosome translocation, forming the Brd4::Nutm1 fusion gene, could induce aggressive carcinomas in mice. The tumors present histopathological and molecular features similar to human NC, with enrichment of undifferentiated cells. Similar to the reports of human NC incidence, Brd4::Nutm1 can induce NC from a broad range of tissues with a strong phenotypical variability. The consistent induction of poorly differentiated carcinoma demonstrated a strong reprogramming activity of BRD4::NUTM1. The new mouse model provided a critical preclinical model for NC that will lead to better understanding and therapy development for NC.
Collapse
Affiliation(s)
- Dejin Zheng
- Department of Obstetrics, Gynecology and Reproductive Biology, College of Human Medicine, Michigan State University, East Lansing, MI, USA
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, USA
| | - Ahmed A Elnegiry
- Department of Obstetrics, Gynecology and Reproductive Biology, College of Human Medicine, Michigan State University, East Lansing, MI, USA
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, USA
- Home Institution: Department of Cytology and Histology, Faculty of Veterinary Medicine, Aswan University, Aswan, Egypt
| | - Chenxiang Luo
- Department of Obstetrics, Gynecology and Reproductive Biology, College of Human Medicine, Michigan State University, East Lansing, MI, USA
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, USA
- Home Institution: Center for Reproductive Medicine and Department of Gynecology & Obstetrics, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, PR China
| | - Mohammed Amine Bendahou
- Infection Biology and Cancer Biology Program, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Liangqi Xie
- Infection Biology and Cancer Biology Program, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Diana Bell
- City of Hope Comprehensive Cancer Center, Pathology, Duarte, CA, USA
| | - Yoko Takahashi
- Department of Head and Neck Surgery, the University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ehab Hanna
- Department of Head and Neck Surgery, the University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - George I Mias
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, USA
- Department of Biochemistry and Molecular Biology, College of Nature Science, Michigan State University, East Lansing, MI, USA
| | - Mayra F Tsoi
- Department of Pathobiology and Diagnostic Investigation, College of Veterinary Medicine, Michigan State University, East Lansing, MI, USA
| | - Bin Gu
- Department of Obstetrics, Gynecology and Reproductive Biology, College of Human Medicine, Michigan State University, East Lansing, MI, USA
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, USA
| |
Collapse
|
6
|
Jones RA, Trejo B, Sil P, Little KA, Pasolli HA, Joyce B, Posfai E, Devenport D. An mTurq2-Col4a1 mouse model allows for live visualization of mammalian basement membrane development. J Cell Biol 2024; 223:e202309074. [PMID: 38051393 PMCID: PMC10697824 DOI: 10.1083/jcb.202309074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 11/03/2023] [Accepted: 11/15/2023] [Indexed: 12/07/2023] Open
Abstract
Basement membranes (BMs) are specialized sheets of extracellular matrix that underlie epithelial and endothelial tissues. BMs regulate the traffic of cells and molecules between compartments, and participate in signaling, cell migration, and organogenesis. The dynamics of mammalian BMs, however, are poorly understood, largely due to a lack of models in which core BM components are endogenously labeled. Here, we describe the mTurquoise2-Col4a1 mouse in which we fluorescently tag collagen IV, the main component of BMs. Using an innovative planar-sagittal live imaging technique to visualize the BM of developing skin, we directly observe BM deformation during hair follicle budding and basal progenitor cell divisions. The BM's inherent pliability enables dividing cells to remain attached to and deform the BM, rather than lose adhesion as generally thought. Using FRAP, we show BM collagen IV is extremely stable, even during periods of rapid epidermal growth. These findings demonstrate the utility of the mTurq2-Col4a1 mouse to shed new light on mammalian BM developmental dynamics.
Collapse
Affiliation(s)
- Rebecca A. Jones
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Brandon Trejo
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Parijat Sil
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | | | - H. Amalia Pasolli
- Electron Microscopy Resource Center, The Rockefeller University, New York, NY, USA
| | - Bradley Joyce
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Eszter Posfai
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Danelle Devenport
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| |
Collapse
|
7
|
Jones RA, Trejo B, Sil P, Little KA, Pasolli HA, Joyce B, Posfai E, Devenport D. A Window into Mammalian Basement Membrane Development: Insights from the mTurq2-Col4a1 Mouse Model. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.27.559396. [PMID: 37808687 PMCID: PMC10557719 DOI: 10.1101/2023.09.27.559396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
Basement membranes (BMs) are specialized sheets of extracellular matrix that underlie epithelial and endothelial tissues. BMs regulate traffic of cells and molecules between compartments, and participate in signaling, cell migration and organogenesis. The dynamics of mammalian BMs, however, are poorly understood, largely due to a lack of models in which core BM components are endogenously labelled. Here, we describe the mTurquoise2-Col4a1 mouse, in which we fluorescently tag collagen IV, the main component of BMs. Using an innovative Planar-Sagittal live imaging technique to visualize the BM of developing skin, we directly observe BM deformation during hair follicle budding and basal progenitor cell divisions. The BM's inherent pliability enables dividing cells to remain attached to and deform the BM, rather than lose adhesion as generally thought. Using FRAP, we show BM collagen IV is extremely stable, even during periods of rapid epidermal growth. These findings demonstrate the utility of the mTurq2-Col4a1 mouse to shed new light on mammalian BM developmental dynamics.
Collapse
Affiliation(s)
- Rebecca A Jones
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544
| | - Brandon Trejo
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544
| | - Parijat Sil
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544
| | - Katherine A Little
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544
| | - H Amalia Pasolli
- Electron Microscopy Resource Center, The Rockefeller University, 1230 York Ave., New York, NY 10065
| | - Bradley Joyce
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544
| | - Eszter Posfai
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544
| | - Danelle Devenport
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544
| |
Collapse
|
8
|
Sowbhagya R, Muktha H, Ramakrishnaiah TN, Surendra AS, Tanvi Y, Nivitha K, Rajashekara S. CRISPR/Cas-mediated genome editing in mice for the development of drug delivery mechanism. Mol Biol Rep 2023; 50:7729-7743. [PMID: 37438488 DOI: 10.1007/s11033-023-08659-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 06/30/2023] [Indexed: 07/14/2023]
Abstract
BACKGROUND To manipulate particular locations in the bacterial genome, researchers have recently resorted to a group of unique sequences in bacterial genomes that are responsible for safeguarding bacteria against bacteriophages. Clustered regularly interspaced short palindromic repeats (CRISPR) and CRISPR-associated protein 9 (Cas9) are two such systems, each of which consists of an RNA component and an enzyme component. METHODS AND RESULTS This review focuses primarily on how CRISPR/Cas9 technology can be used to make models to study human diseases in mice. Creating RNA molecules that direct endonucleases to a specific position in the genome are crucial for achieving a specific genetic modification. CRISPR/Cas9 technology has allowed scientists to edit the genome with greater precision than ever before. Researchers can use knock-in and knock-out methods to model human diseases such as Neurological, cardiovascular disease, and cancer. CONCLUSIONS In terms of developing innovative methods to discover ailments for diseases/disorders, improved CRISPR/Cas9 technology will provide easier access to valuable novel animal models.
Collapse
Affiliation(s)
- Ramachandregowda Sowbhagya
- Department of Biotechnology and Genetics, M.S. Ramaiah College of Arts, Science and Commerce, 7th Main Rd, MSRIT, M S R Nagar, Mathikere, Bengaluru, Karnataka, 560 054, India
| | - Harsha Muktha
- Department of Biotechnology and Genetics, M.S. Ramaiah College of Arts, Science and Commerce, 7th Main Rd, MSRIT, M S R Nagar, Mathikere, Bengaluru, Karnataka, 560 054, India
| | - Thippenahalli Narasimhaiah Ramakrishnaiah
- Department of Biotechnology and Genetics, M.S. Ramaiah College of Arts, Science and Commerce, 7th Main Rd, MSRIT, M S R Nagar, Mathikere, Bengaluru, Karnataka, 560 054, India
| | - Adagur Sudarshan Surendra
- Department of Biochemistry, M.S. Ramaiah College of Arts, Science and Commerce, 7th Main Rd, MSRIT, M S R Nagar, Mathikere, Bengaluru, Karnataka, 560 054, India
| | - Yesudas Tanvi
- Department of Biotechnology and Genetics, M.S. Ramaiah College of Arts, Science and Commerce, 7th Main Rd, MSRIT, M S R Nagar, Mathikere, Bengaluru, Karnataka, 560 054, India
| | - Karayi Nivitha
- Department of Biotechnology and Genetics, M.S. Ramaiah College of Arts, Science and Commerce, 7th Main Rd, MSRIT, M S R Nagar, Mathikere, Bengaluru, Karnataka, 560 054, India
| | - Somashekara Rajashekara
- Centre for Applied Genetics, Department of Studies in Zoology, Bangalore University, Jnana Bharathi Campus, Off Mysuru Road, Bengaluru, Karnataka, 560 056, India.
| |
Collapse
|
9
|
Davis DJ, McNew JF, Maresca-Fichter H, Chen K, Telugu BP, Bryda EC. Efficient DNA knock-in using AAV-mediated delivery with 2-cell embryo CRISPR-Cas9 electroporation. Front Genome Ed 2023; 5:1256451. [PMID: 37694158 PMCID: PMC10485772 DOI: 10.3389/fgeed.2023.1256451] [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: 07/10/2023] [Accepted: 08/18/2023] [Indexed: 09/12/2023] Open
Abstract
Recent advances in CRISPR-Cas genome editing technology have been instrumental in improving the efficiency to produce genetically modified animal models. In this study we have combined four very promising approaches to come up with a highly effective pipeline to produce knock-in mouse and rat models. The four combined methods include: AAV-mediated DNA delivery, single-stranded DNA donor templates, 2-cell embryo modification, and CRISPR-Cas ribonucleoprotein (RNP) electroporation. Using this new combined approach, we were able to produce successfully targeted knock-in rat models containing either Cre or Flp recombinase sequences with knock-in efficiencies over 90%. Furthermore, we were able to produce a knock-in mouse model containing a Cre recombinase targeted insertion with over 50% knock-in efficiency directly comparing efficiencies to other commonly used approaches. Our modified AAV-mediated DNA delivery with 2-cell embryo CRISPR-Cas9 RNP electroporation technique has proven to be highly effective for generating both knock-in mouse and knock-in rat models.
Collapse
Affiliation(s)
- Daniel J. Davis
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, United States
| | - James F. McNew
- Comparative Medicine Program, University of Missouri, Columbia, MO, United States
| | - Hailey Maresca-Fichter
- School of Veterinary Medicine, Michigan State University, East Lansing, MI, United States
| | - Kaiwen Chen
- School of Veterinary Medicine, Kansas State University, Manhattan, KS, United States
| | - Bhanu P. Telugu
- Department of Animal Sciences, University of Missouri, Columbia, MO, United States
| | - Elizabeth C. Bryda
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, United States
- Rat Resource and Research Center, Columbia, MO, United States
| |
Collapse
|
10
|
Xie H, Linning-Duffy K, Demireva EY, Toh H, Abolibdeh B, Shi J, Zhou B, Iwase S, Yan L. CRISPR-based Genome Editing of a Diurnal Rodent, Nile Grass Rat ( Arvicanthis niloticus). BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.23.553600. [PMID: 37662225 PMCID: PMC10473663 DOI: 10.1101/2023.08.23.553600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
Diurnal and nocturnal mammals have evolved distinct pathways to optimize survival for their chronotype-specific lifestyles. Conventional rodent models, being nocturnal, may not sufficiently recapitulate the biology of diurnal humans in health and disease. Although diurnal rodents are potentially advantageous for translational research, until recently, they have not been genetically tractable. Here, we address this major limitation by demonstrating the first successful CRISPR genome editing of the Nile grass rat ( Arvicanthis niloticus ), a valuable diurnal rodent. We establish methods for superovulation; embryo development, manipulation, and culture; and pregnancy maintenance to guide future genome editing of this and other diurnal rodent species.
Collapse
|
11
|
Nunley H, Shao B, Grover P, Singh J, Joyce B, Kim-Yip R, Kohrman A, Watters A, Gal Z, Kickuth A, Chalifoux M, Shvartsman S, Posfai E, Brown LM. A novel ground truth dataset enables robust 3D nuclear instance segmentation in early mouse embryos. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.14.532646. [PMID: 36993260 PMCID: PMC10055179 DOI: 10.1101/2023.03.14.532646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
For investigations into fate specification and cell rearrangements in live images of preimplantation embryos, automated and accurate 3D instance segmentation of nuclei is invaluable; however, the performance of segmentation methods is limited by the images' low signal-to-noise ratio and high voxel anisotropy and the nuclei's dense packing and variable shapes. Supervised machine learning approaches have the potential to radically improve segmentation accuracy but are hampered by a lack of fully annotated 3D data. In this work, we first establish a novel mouse line expressing near-infrared nuclear reporter H2B-miRFP720. H2B-miRFP720 is the longest wavelength nuclear reporter in mice and can be imaged simultaneously with other reporters with minimal overlap. We then generate a dataset, which we call BlastoSPIM, of 3D microscopy images of H2B-miRFP720-expressing embryos with ground truth for nuclear instance segmentation. Using BlastoSPIM, we benchmark the performance of five convolutional neural networks and identify Stardist-3D as the most accurate instance segmentation method across preimplantation development. Stardist-3D, trained on BlastoSPIM, performs robustly up to the end of preimplantation development (> 100 nuclei) and enables studies of fate patterning in the late blastocyst. We, then, demonstrate BlastoSPIM's usefulness as pre-train data for related problems. BlastoSPIM and its corresponding Stardist-3D models are available at: blastospim.flatironinstitute.org.
Collapse
Affiliation(s)
- Hayden Nunley
- Center for Computational Biology, Flatiron Institute - Simons Foundation, New York, United States of America
| | - Binglun Shao
- Center for Computational Biology, Flatiron Institute - Simons Foundation, New York, United States of America
- Department of Chemical and Biological Engineering, Princeton University, Princeton, New Jersey, United States of America
| | - Prateek Grover
- Center for Computational Biology, Flatiron Institute - Simons Foundation, New York, United States of America
| | - Jaspreet Singh
- Center for Computational Biology, Flatiron Institute - Simons Foundation, New York, United States of America
| | - Bradley Joyce
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, United States of America
| | - Rebecca Kim-Yip
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, United States of America
| | - Abraham Kohrman
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, United States of America
| | - Aaron Watters
- Center for Computational Biology, Flatiron Institute - Simons Foundation, New York, United States of America
| | - Zsombor Gal
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, United States of America
| | - Alison Kickuth
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, United States of America
| | - Madeleine Chalifoux
- Department of Chemical and Biological Engineering, Princeton University, Princeton, New Jersey, United States of America
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, United States of America
| | - Stanislav Shvartsman
- Center for Computational Biology, Flatiron Institute - Simons Foundation, New York, United States of America
- Department of Chemical and Biological Engineering, Princeton University, Princeton, New Jersey, United States of America
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, United States of America
- The Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, New Jersey, United States of America
| | - Eszter Posfai
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, United States of America
| | - Lisa M. Brown
- Center for Computational Biology, Flatiron Institute - Simons Foundation, New York, United States of America
| |
Collapse
|
12
|
Phan HTL, Kim K, Lee H, Seong JK. Progress in and Prospects of Genome Editing Tools for Human Disease Model Development and Therapeutic Applications. Genes (Basel) 2023; 14:483. [PMID: 36833410 PMCID: PMC9957140 DOI: 10.3390/genes14020483] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 02/09/2023] [Accepted: 02/10/2023] [Indexed: 02/17/2023] Open
Abstract
Programmable nucleases, such as zinc finger nucleases (ZFNs), transcription activator-like effector nucleases (TALENs), and clustered regularly interspaced short palindromic repeats (CRISPR)/Cas, are widely accepted because of their diversity and enormous potential for targeted genomic modifications in eukaryotes and other animals. Moreover, rapid advances in genome editing tools have accelerated the ability to produce various genetically modified animal models for studying human diseases. Given the advances in gene editing tools, these animal models are gradually evolving toward mimicking human diseases through the introduction of human pathogenic mutations in their genome rather than the conventional gene knockout. In the present review, we summarize the current progress in and discuss the prospects for developing mouse models of human diseases and their therapeutic applications based on advances in the study of programmable nucleases.
Collapse
Affiliation(s)
- Hong Thi Lam Phan
- Department of Physiology, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Kyoungmi Kim
- Department of Physiology, Korea University College of Medicine, Seoul 02841, Republic of Korea
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Ho Lee
- Graduate School of Cancer Science and Policy, National Cancer Center, Goyang 10408, Republic of Korea
| | - Je Kyung Seong
- Korea Mouse Phenotyping Center, Seoul National University, Seoul 08826, Republic of Korea
- Laboratory of Developmental Biology and Genomics, BK21 PLUS Program for Creative Veterinary Science Research, Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea
- Interdisciplinary Program for Bioinformatics, Program for Cancer Biology, BIO-MAX/N-Bio Institute, Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
13
|
Strategies for generation of mice via CRISPR/HDR-mediated knock-in. Mol Biol Rep 2023; 50:3189-3204. [PMID: 36701041 DOI: 10.1007/s11033-023-08278-8] [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: 05/24/2022] [Accepted: 01/12/2023] [Indexed: 01/27/2023]
Abstract
CRISPR/Cas9 framework is generally used to generate genetically modified mouse models. The clustered regularly interspaced short palindromic repeat gene editing technique, can efficiently generate knock-outs using the non-homologous end-joining repair pathway. Small knock-ins also work precisely using a repair template with help of homology-directed-repair (HDR) mechanism. However, when the fragment size is larger than 4-5 kb, the knock-in tends to be error prone and the efficiency decreases. Certain types of modifications, in particular insertions of very large DNA fragments (10-100 kb) or entire gene replacements, are still difficult. The HDR process needs further streamlining and improvement. Here in this review, we describe methods to enhance the efficiency of the knock-in through checking each step from the guide design to the microinjection and choice of the oocyte donors. This helps in understanding the parameters that can be modified to get improved knock-in efficiency via CRISPR targeting.
Collapse
|
14
|
Lintott LG, Nutter LMJ. Genetic and Molecular Quality Control of Genetically Engineered Mice. Methods Mol Biol 2023; 2631:53-101. [PMID: 36995664 DOI: 10.1007/978-1-0716-2990-1_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/31/2023]
Abstract
Genetically engineered mice are used as avatars to understand mammalian gene function and develop therapies for human disease. During genetic modification, unintended changes can occur, and these changes may result in misassigned gene-phenotype relationships leading to incorrect or incomplete experimental interpretations. The types of unintended changes that may occur depend on the allele type being made and the genetic engineering approach used. Here we broadly categorize allele types as deletions, insertions, base changes, and transgenes derived from engineered embryonic stem (ES) cells or edited mouse embryos. However, the methods we describe can be adapted to other allele types and engineering strategies. We describe the sources and consequ ences of common unintended changes and best practices for detecting both intended and unintended changes by screening and genetic and molecular quality control (QC) of chimeras, founders, and their progeny. Employing these practices, along with careful allele design and good colony management, will increase the chance that investigations using genetically engineered mice will produce high-quality reproducible results, to enable a robust understanding of gene function, human disease etiology, and therapeutic development.
Collapse
Affiliation(s)
- Lauri G Lintott
- The Centre for Phenogenomics, Toronto, ON, Canada
- The Hospital for Sick Children, Toronto, ON, Canada
| | - Lauryl M J Nutter
- The Centre for Phenogenomics, Toronto, ON, Canada.
- The Hospital for Sick Children, Toronto, ON, Canada.
| |
Collapse
|
15
|
Buch T, Jerchow B, Zevnik B. Practical Application of the 3Rs in Rodent Transgenesis. Methods Mol Biol 2023; 2631:33-51. [PMID: 36995663 DOI: 10.1007/978-1-0716-2990-1_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/31/2023]
Abstract
The principles of the 3Rs (replace, reduce, refine), as originally published by Russell and Burch, are internationally acclaimed guidelines for meeting ethical and welfare standards in animal experimentation. Genome manipulation is a standard technique in biomedical research and beyond. The goal of this chapter is to give practical advice on the implementation of the 3Rs in laboratories generating genetically modified rodents. We cover 3R aspects from the planning phase through operations of the transgenic unit to the final genome-manipulated animals. The focus of our chapter is on an easy-to-use, concise protocol that is close to a checklist. While we focus on mice, the proposed methodological concepts can be easily adapted for the manipulation of other sentient animals.
Collapse
Affiliation(s)
- Thorsten Buch
- Institute of Laboratory Animal Science, University of Zurich, Zurich, Switzerland
| | - Boris Jerchow
- Novartis Institute for Biomedical Research (NIBR), Novartis Pharma AG, Basel, Switzerland
| | - Branko Zevnik
- In vivo Research Facility, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany.
- Cologne Excellence Cluster for Cellular Stress Responses in Aging-Associated Diseases (CECAD), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany.
| |
Collapse
|
16
|
Tozaki T, Ohnuma A, Nakamura K, Hano K, Takasu M, Takahashi Y, Tamura N, Sato F, Shimizu K, Kikuchi M, Ishige T, Kakoi H, Hirota KI, Hamilton NA, Nagata SI. Detection of Indiscriminate Genetic Manipulation in Thoroughbred Racehorses by Targeted Resequencing for Gene-Doping Control. Genes (Basel) 2022; 13:genes13091589. [PMID: 36140757 PMCID: PMC9498419 DOI: 10.3390/genes13091589] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Revised: 08/29/2022] [Accepted: 08/31/2022] [Indexed: 11/16/2022] Open
Abstract
The creation of genetically modified horses is prohibited in horse racing as it falls under the banner of gene doping. In this study, we developed a test to detect gene editing based on amplicon sequencing using next-generation sequencing (NGS). We designed 1012 amplicons to target 52 genes (481 exons) and 147 single-nucleotide variants (SNVs). NGS analyses showed that 97.7% of the targeted exons were sequenced to sufficient coverage (depth > 50) for calling variants. The targets of artificial editing were defined as homozygous alternative (HomoALT) and compound heterozygous alternative (ALT1/ALT2) insertion/deletion (INDEL) mutations in this study. Four models of gene editing (three homoALT with 1-bp insertions, one REF/ALT with 77-bp deletion) were constructed by editing the myostatin gene in horse fibroblasts using CRISPR/Cas9. The edited cells and 101 samples from thoroughbred horses were screened using the developed test, which was capable of identifying the three homoALT cells containing 1-bp insertions. Furthermore, 147 SNVs were investigated for their utility in confirming biological parentage. Of these, 120 SNVs were amenable to consistent and accurate genotyping. Surrogate (nonbiological) dams were excluded by 9.8 SNVs on average, indicating that the 120 SNV could be used to detect foals that have been produced by somatic cloning or embryo transfer, two practices that are prohibited in thoroughbred racing and breeding. These results indicate that gene-editing tests that include variant calling and SNV genotyping are useful to identify genetically modified racehorses.
Collapse
Affiliation(s)
- Teruaki Tozaki
- Genetic Analysis Department, Laboratory of Racing Chemistry, 1731-2, Tsurutamachi, Utsunomiya 320-0851, Japan
- Correspondence:
| | - Aoi Ohnuma
- Genetic Analysis Department, Laboratory of Racing Chemistry, 1731-2, Tsurutamachi, Utsunomiya 320-0851, Japan
| | - Kotono Nakamura
- Department of Veterinary Medicine, Faculty of Applied Biological Sciences, Gifu University, 1-1, Yanagido, Gifu 501-1193, Japan
| | - Kazuki Hano
- Department of Veterinary Medicine, Faculty of Applied Biological Sciences, Gifu University, 1-1, Yanagido, Gifu 501-1193, Japan
| | - Masaki Takasu
- Department of Veterinary Medicine, Faculty of Applied Biological Sciences, Gifu University, 1-1, Yanagido, Gifu 501-1193, Japan
| | - Yuji Takahashi
- Equine Research Institute, Japan Racing Association, 1400-4, Shiba, Shimotsuke 329-0412, Japan
| | - Norihisa Tamura
- Equine Research Institute, Japan Racing Association, 1400-4, Shiba, Shimotsuke 329-0412, Japan
| | - Fumio Sato
- Equine Research Institute, Japan Racing Association, 1400-4, Shiba, Shimotsuke 329-0412, Japan
| | - Kyo Shimizu
- Registration Department, Japan Association for International Racing and Stud Book, 4-5-4, Shimbashi, Minato, Tokyo 105-0004, Japan
| | - Mio Kikuchi
- Genetic Analysis Department, Laboratory of Racing Chemistry, 1731-2, Tsurutamachi, Utsunomiya 320-0851, Japan
| | - Taichiro Ishige
- Genetic Analysis Department, Laboratory of Racing Chemistry, 1731-2, Tsurutamachi, Utsunomiya 320-0851, Japan
| | - Hironaga Kakoi
- Genetic Analysis Department, Laboratory of Racing Chemistry, 1731-2, Tsurutamachi, Utsunomiya 320-0851, Japan
| | - Kei-ichi Hirota
- Genetic Analysis Department, Laboratory of Racing Chemistry, 1731-2, Tsurutamachi, Utsunomiya 320-0851, Japan
| | - Natasha A. Hamilton
- Equine Genetics Research Centre, Racing Australia, 2 Randwick Way, Scone, NSW 2337, Australia
| | - Shun-ichi Nagata
- Genetic Analysis Department, Laboratory of Racing Chemistry, 1731-2, Tsurutamachi, Utsunomiya 320-0851, Japan
| |
Collapse
|
17
|
Chuang HC, Hsueh CH, Hsu PM, Huang RH, Tsai CY, Chung NH, Chow YH, Tan TH. SARS-CoV-2 spike protein enhances MAP4K3/GLK-induced ACE2 stability in COVID-19. EMBO Mol Med 2022; 14:e15904. [PMID: 35894122 PMCID: PMC9353388 DOI: 10.15252/emmm.202215904] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 07/11/2022] [Accepted: 07/12/2022] [Indexed: 11/09/2022] Open
Abstract
ACE2 on epithelial cells is the SARS-CoV-2 entry receptor. Single-cell RNA-sequencing data derived from two COVID-19 cohorts revealed that MAP4K3/GLK-positive epithelial cells were increased in patients. SARS-CoV-2-induced GLK overexpression in epithelial cells was correlated with COVID-19 severity and vesicle secretion. GLK overexpression induced the epithelial cell-derived exosomes containing ACE2; the GLK-induced exosomes transported ACE2 proteins to recipient cells, facilitating pseudovirus infection. Consistently, ACE2 proteins were increased in the serum exosomes from another COVID-19 cohort. Remarkably, SARS-CoV-2 spike protein-stimulated GLK, and GLK stabilized ACE2 in epithelial cells. Mechanistically, GLK phosphorylated ACE2 at two serine residues (Ser776, Ser783), leading to the dissociation of ACE2 from its E3 ligase UBR4. Reduction in UBR4-induced Lys48-linked ubiquitination at three lysine residues (Lys26, Lys112, Lys114) of ACE2 prevented its degradation. Furthermore, SARS-CoV-2 pseudovirus or live virus infection in humanized ACE2 mice induced GLK and ACE2 protein levels, and ACE2-containing exosomes. Collectively, ACE2 stabilization by SARS-CoV-2-induced MAP4K3/GLK may contribute to the pathogenesis of COVID-19.
Collapse
Affiliation(s)
- Huai-Chia Chuang
- Immunology Research Center, National Health Research Institutes, Zhunan, Taiwan
| | - Chia-Hsin Hsueh
- Immunology Research Center, National Health Research Institutes, Zhunan, Taiwan
| | - Pu-Ming Hsu
- Immunology Research Center, National Health Research Institutes, Zhunan, Taiwan
| | - Rou-Huei Huang
- Immunology Research Center, National Health Research Institutes, Zhunan, Taiwan
| | - Ching-Yi Tsai
- Immunology Research Center, National Health Research Institutes, Zhunan, Taiwan
| | - Nai-Hsiang Chung
- National Institute of Infectious Disease and Vaccinology, National Health Research Institutes, Zhunan, Taiwan.,Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu, Taiwan
| | - Yen-Hung Chow
- National Institute of Infectious Disease and Vaccinology, National Health Research Institutes, Zhunan, Taiwan.,Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
| | - Tse-Hua Tan
- Immunology Research Center, National Health Research Institutes, Zhunan, Taiwan.,Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
18
|
Macdonald L, Taylor GC, Brisbane JM, Christodoulou E, Scott L, von Kriegsheim A, Rossant J, Gu B, Wood AJ. Rapid and specific degradation of endogenous proteins in mouse models using auxin-inducible degrons. eLife 2022; 11:e77987. [PMID: 35736539 PMCID: PMC9273210 DOI: 10.7554/elife.77987] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 05/19/2022] [Indexed: 11/13/2022] Open
Abstract
Auxin-inducible degrons are a chemical genetic tool for targeted protein degradation and are widely used to study protein function in cultured mammalian cells. Here, we develop CRISPR-engineered mouse lines that enable rapid and highly specific degradation of tagged endogenous proteins in vivo. Most but not all cell types are competent for degradation. By combining ligand titrations with genetic crosses to generate animals with different allelic combinations, we show that degradation kinetics depend upon the dose of the tagged protein, ligand, and the E3 ligase substrate receptor TIR1. Rapid degradation of condensin I and II - two essential regulators of mitotic chromosome structure - revealed that both complexes are individually required for cell division in precursor lymphocytes, but not in their differentiated peripheral lymphocyte derivatives. This generalisable approach provides unprecedented temporal control over the dose of endogenous proteins in mouse models, with implications for studying essential biological pathways and modelling drug activity in mammalian tissues.
Collapse
Affiliation(s)
- Lewis Macdonald
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of EdinburghEdinburghUnited Kingdom
| | - Gillian C Taylor
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of EdinburghEdinburghUnited Kingdom
| | - Jennifer Margaret Brisbane
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of EdinburghEdinburghUnited Kingdom
| | - Ersi Christodoulou
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of EdinburghEdinburghUnited Kingdom
| | - Lucy Scott
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of EdinburghEdinburghUnited Kingdom
| | - Alex von Kriegsheim
- Cancer Research UK Edinburgh Centre, Institute of Genetics and Cancer, University of EdinburghEdinburghUnited Kingdom
| | - Janet Rossant
- Program in Developmental and Stem Cell Biology, Hospital for Sick ChildrenTorontoCanada
| | - Bin Gu
- Department of Obstetrics, Gynecology and Reproductive Biology, Michigan State UniversityEast LansingUnited States
- Department of Biomedical Engineering; Michigan State UniversityEast LansingUnited States
- Institute for Quantitative Health Science and Engineering, Michigan State UniversityEast LansingUnited States
| | - Andrew J Wood
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of EdinburghEdinburghUnited Kingdom
| |
Collapse
|
19
|
3R measures in facilities for the production of genetically modified rodents. Lab Anim (NY) 2022; 51:162-177. [PMID: 35641635 DOI: 10.1038/s41684-022-00978-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 04/22/2022] [Indexed: 12/30/2022]
Abstract
Sociocultural changes in the human-animal relationship have led to increasing demands for animal welfare in biomedical research. The 3R concept is the basis for bringing this demand into practice: Replace animal experiments with alternatives where possible, Reduce the number of animals used to a scientifically justified minimum and Refine the procedure to minimize animal harm. The generation of gene-modified sentient animals such as mice and rats involves many steps that include various forms of manipulation. So far, no coherent analysis of the application of the 3Rs to gene manipulation has been performed. Here we provide guidelines from the Committee on Genetics and Breeding of Laboratory Animals of the German Society for Laboratory Animal Science to implement the 3Rs in every step during the generation of genetically modified animals. We provide recommendations for applying the 3Rs as well as success/intervention parameters for each step of the process, from experiment planning to choice of technology, harm-benefit analysis, husbandry conditions, management of genetically modified lines and actual procedures. We also discuss future challenges for animal welfare in the context of developing technologies. Taken together, we expect that our comprehensive analysis and our recommendations for the appropriate implementation of the 3Rs to technologies for genetic modifications of rodents will benefit scientists from a wide range of disciplines and will help to improve the welfare of a large number of laboratory animals worldwide.
Collapse
|
20
|
Ang JXD, Nevard K, Ireland R, Purusothaman DK, Verkuijl SAN, Shackleford L, Gonzalez E, Anderson MAE, Alphey L. Considerations for homology-based DNA repair in mosquitoes: Impact of sequence heterology and donor template source. PLoS Genet 2022; 18:e1010060. [PMID: 35180218 PMCID: PMC8893643 DOI: 10.1371/journal.pgen.1010060] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 03/03/2022] [Accepted: 01/28/2022] [Indexed: 12/23/2022] Open
Abstract
The increasing prevalence of insecticide resistance and the ongoing global burden of vector-borne diseases have encouraged new efforts in mosquito control. For Aedes aegypti, the most important arboviral vector, integration rates achieved in Cas9-based knock-ins so far have been rather low, highlighting the need to understand gene conversion patterns and other factors that influence homology-directed repair (HDR) events in this species. In this study, we report the effects of sequence mismatches or donor template forms on integration rates. We found that modest sequence differences between construct homology arms [DNA sequence in the donor template which resembles the region flanking the target cut] and genomic target comprising 1.2% nucleotide dissimilarity (heterology) significantly reduced integration rates. While most integrations (59-88%) from plasmid templates were the result of canonical [on target, perfect repair] HDR events, no canonical events were identified from other donor types (i.e. ssDNA, biotinylated ds/ssDNA). Sequencing of the transgene flanking region in 69 individuals with canonical integrations revealed 60% of conversion tracts to be unidirectional and extend up to 220 bp proximal to the break, though in three individuals bidirectional conversion of up to 725 bp was observed.
Collapse
Affiliation(s)
| | | | | | | | - Sebald A. N. Verkuijl
- The Pirbright Institute, Pirbright, Woking, United Kingdom
- Mathematical Ecology Research Group, Department of Zoology, University of Oxford, Oxford, United Kingdom
| | | | | | | | - Luke Alphey
- The Pirbright Institute, Pirbright, Woking, United Kingdom
| |
Collapse
|
21
|
Gu B, Bradshaw B, Zhu M, Sun Y, Hopyan S, Rossant J. Live imaging YAP signalling in mouse embryo development. Open Biol 2022; 12:210335. [PMID: 35042406 PMCID: PMC8767199 DOI: 10.1098/rsob.210335] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022] Open
Abstract
YAP protein is a critical regulator of mammalian embryonic development. By generating a near-infrared fusion YAP reporter mouse line, we have achieved high-resolution live imaging of YAP localization during mouse embryonic development. We have validated the reporter by demonstrating its predicted responses to blocking LATS kinase activity or blocking cell polarity. By time lapse imaging preimplantation embryos, we revealed a mitotic reset behaviour of YAP nuclear localization. We also demonstrated deep tissue live imaging in post-implantation embryos and revealed an intriguing nuclear YAP pattern in migrating cells. The YAP fusion reporter mice and imaging methods will open new opportunities for understanding dynamic YAP signalling in vivo in many different situations.
Collapse
Affiliation(s)
- Bin Gu
- Program in Developmental and Stem Cell Biology, Hospital for Sick Children, Toronto, Ontario, Canada M5G 0A4
| | - Brian Bradshaw
- Program in Developmental and Stem Cell Biology, Hospital for Sick Children, Toronto, Ontario, Canada M5G 0A4
| | - Min Zhu
- Program in Developmental and Stem Cell Biology, Hospital for Sick Children, Toronto, Ontario, Canada M5G 0A4.,Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada M5S 1A8.,Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, Ontario, Canada M5S 3G8
| | - Yu Sun
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, Ontario, Canada M5S 3G8.,Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada M5S 3G9.,Department of Electrical and Computer Engineering, University of Toronto, Toronto, Ontario, Canada M5S 3G4
| | - Sevan Hopyan
- Program in Developmental and Stem Cell Biology, Hospital for Sick Children, Toronto, Ontario, Canada M5G 0A4.,Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada M5S 1A8.,Division of Orthopaedic Surgery, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada M5G 1X8
| | - Janet Rossant
- Program in Developmental and Stem Cell Biology, Hospital for Sick Children, Toronto, Ontario, Canada M5G 0A4.,Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada M5S 1A8
| |
Collapse
|
22
|
Basta LP, Hill-Oliva M, Paramore SV, Sharan R, Goh A, Biswas A, Cortez M, Little KA, Posfai E, Devenport D. New mouse models for high resolution and live imaging of planar cell polarity proteins in vivo. Development 2021; 148:271988. [PMID: 34463728 DOI: 10.1242/dev.199695] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 08/24/2021] [Indexed: 01/10/2023]
Abstract
The collective polarization of cellular structures and behaviors across a tissue plane is a near universal feature of epithelia known as planar cell polarity (PCP). This property is controlled by the core PCP pathway, which consists of highly conserved membrane-associated protein complexes that localize asymmetrically at cell junctions. Here, we introduce three new mouse models for investigating the localization and dynamics of transmembrane PCP proteins: Celsr1, Fz6 and Vangl2. Using the skin epidermis as a model, we characterize and verify the expression, localization and function of endogenously tagged Celsr1-3xGFP, Fz6-3xGFP and tdTomato-Vangl2 fusion proteins. Live imaging of Fz6-3xGFP in basal epidermal progenitors reveals that the polarity of the tissue is not fixed through time. Rather, asymmetry dynamically shifts during cell rearrangements and divisions, while global, average polarity of the tissue is preserved. We show using super-resolution STED imaging that Fz6-3xGFP and tdTomato-Vangl2 can be resolved, enabling us to observe their complex localization along junctions. We further explore PCP fusion protein localization in the trachea and neural tube, and discover new patterns of PCP expression and localization throughout the mouse embryo.
Collapse
Affiliation(s)
- Lena P Basta
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544USA
| | - Michael Hill-Oliva
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544USA.,Department of Medicine, Columbia University, New York, NY 10032USA
| | - Sarah V Paramore
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544USA
| | - Rishabh Sharan
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544USA
| | - Audrey Goh
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544USA
| | - Abhishek Biswas
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544USA.,Research Computing, Office of Information Technology, Princeton University, Princeton, NJ 08544, USA
| | - Marvin Cortez
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544USA
| | - Katherine A Little
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544USA
| | - Eszter Posfai
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544USA
| | - Danelle Devenport
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544USA
| |
Collapse
|
23
|
Bandyopadhyay S, Douglass J, Kapell S, Khan N, Feitosa-Suntheimer F, Klein JA, Temple J, Brown-Culbertson J, Tavares AH, Saeed M, Lau NC. DNA templates with blocked long 3' end single-stranded overhangs (BL3SSO) promote bona fide Cas9-stimulated homology-directed repair of long transgenes into endogenous gene loci. G3-GENES GENOMES GENETICS 2021; 11:6275753. [PMID: 33989385 PMCID: PMC8496256 DOI: 10.1093/g3journal/jkab169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 05/03/2021] [Indexed: 11/16/2022]
Abstract
Knock-in of large transgenes by Cas9-mediated homology-directed repair (HDR) is an extremely inefficient process. Although the use of single-stranded oligonucleotides (ssODN) as an HDR donor has improved the integration of smaller transgenes, they do not support efficient insertion of large DNA sequences. In an effort to gain insights into the mechanism(s) governing the HDR-mediated integration of larger transgenes and to improve the technology, we conducted knock-in experiments targeting the human EMX1 locus and applied rigorous genomic PCR analyses in the human HEK293 cell line. This exercise revealed an unexpected molecular complication arising from the transgene HDR being initiated at the single homology arm and the subsequent genomic integration of plasmid backbone sequences. To pivot around this problem, we devised a novel PCR-constructed template containing blocked long 3' single-stranded overhangs (BL3SSO) that greatly improved the efficiency of bona fide Cas9-stimulated HDR at the EMX1 locus. We further refined BL3SSO technology and successfully used it to insert GFP transgenes into two important interferon-stimulated genes (ISGs) loci, Viperin/RSAD2, and ISG15. This study demonstrates the utility of the BL3SSO platform for inserting long DNA sequences into both constitutive and inducible endogenous loci to generate novel human cell lines for the study of important biological processes.
Collapse
Affiliation(s)
- Saptaparni Bandyopadhyay
- Department of Biochemistry, Boston University School of Medicine, Boston University, Boston, MA 02118, USA
| | - Joseph Douglass
- Department of Biochemistry, Boston University School of Medicine, Boston University, Boston, MA 02118, USA
| | - Sebastian Kapell
- Department of Biochemistry, Boston University School of Medicine, Boston University, Boston, MA 02118, USA.,National Emerging Infectious Diseases Laboratories (NEIDL), Boston University, Boston, MA 02118, USA
| | - Nazimuddin Khan
- Department of Biochemistry, Boston University School of Medicine, Boston University, Boston, MA 02118, USA.,National Emerging Infectious Diseases Laboratories (NEIDL), Boston University, Boston, MA 02118, USA
| | | | - Jenny A Klein
- Department of Biology, Brandeis University, Waltham, MA 02453, USA.,Department of Anatomy and Neurobiology, Boston University School of Medicine, Boston University, Boston, MA 02118, USA
| | - Jasmine Temple
- Department of Biology, Brandeis University, Waltham, MA 02453, USA
| | - Jayce Brown-Culbertson
- Department of Biochemistry, Boston University School of Medicine, Boston University, Boston, MA 02118, USA
| | - Alexander H Tavares
- Department of Biochemistry, Boston University School of Medicine, Boston University, Boston, MA 02118, USA.,National Emerging Infectious Diseases Laboratories (NEIDL), Boston University, Boston, MA 02118, USA
| | - Mohsan Saeed
- Department of Biochemistry, Boston University School of Medicine, Boston University, Boston, MA 02118, USA.,National Emerging Infectious Diseases Laboratories (NEIDL), Boston University, Boston, MA 02118, USA
| | - Nelson C Lau
- Department of Biochemistry, Boston University School of Medicine, Boston University, Boston, MA 02118, USA.,National Emerging Infectious Diseases Laboratories (NEIDL), Boston University, Boston, MA 02118, USA.,Genome Science Institute, Boston University School of Medicine, Boston University, Boston, MA 02118, USA
| |
Collapse
|
24
|
Park KE, Frey JF, Waters J, Simpson SG, Coutu C, Plummer S, Campbell M, Donovan DM, Telugu BP. One-Step Homology Mediated CRISPR-Cas Editing in Zygotes for Generating Genome Edited Cattle. CRISPR J 2020; 3:523-534. [PMID: 33252243 PMCID: PMC7757693 DOI: 10.1089/crispr.2020.0047] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Selective breeding and genetic modification have been the cornerstone of animal agriculture. However, the current strategy of breeding animals over multiple generations to introgress novel alleles is not practical in addressing global challenges such as climate change, pandemics, and the predicted need to feed a population of 9 billion by 2050. Consequently, genome editing in zygotes to allow for seamless introgression of novel alleles is required, especially in cattle with long generation intervals. We report for the first time the use of CRISPR-Cas genome editors to introduce novel PRNP allelic variants that have been shown to provide resilience towards human prion pandemics. From one round of embryo injections, we have established six pregnancies and birth of seven edited offspring, with two founders showing >90% targeted homology-directed repair modifications. This study lays out the framework for in vitro optimization, unbiased deep-sequencing to identify editing outcomes, and generation of high frequency homology-directed repair–edited calves.
Collapse
Affiliation(s)
- Ki-Eun Park
- Animal and Avian Sciences, University of Maryland, College Park, Maryland, USA.,RenOVAte Biosciences, Inc., Reisterstown, Maryland, USA
| | - Juli Foster Frey
- Animal Biosciences and Biotechnology Laboratory, USDA, ARS, Beltsville, Maryland, USA
| | - Jerel Waters
- RenOVAte Biosciences, Inc., Reisterstown, Maryland, USA
| | - Sean G Simpson
- Animal and Avian Sciences, University of Maryland, College Park, Maryland, USA.,RenOVAte Biosciences, Inc., Reisterstown, Maryland, USA
| | - Chris Coutu
- Thomas D. Morris Inc., Reisterstown, Maryland, USA
| | | | | | - David M Donovan
- Animal Biosciences and Biotechnology Laboratory, USDA, ARS, Beltsville, Maryland, USA
| | - Bhanu P Telugu
- Animal and Avian Sciences, University of Maryland, College Park, Maryland, USA.,RenOVAte Biosciences, Inc., Reisterstown, Maryland, USA
| |
Collapse
|
25
|
McEwan AR, MacKenzie A. Perspective: Quality Versus Quantity; Is It Important to Assess the Role of Enhancers in Complex Disease from an In Vivo Perspective? Int J Mol Sci 2020; 21:E7856. [PMID: 33113946 PMCID: PMC7660172 DOI: 10.3390/ijms21217856] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 10/15/2020] [Accepted: 10/20/2020] [Indexed: 12/18/2022] Open
Abstract
Sequencing of the human genome has permitted the development of genome-wide association studies (GWAS) to analyze the genetics of a number of complex disorders such as depression, anxiety and substance abuse. Thanks to their ability to analyze huge cohort sizes, these studies have successfully identified thousands of loci associated with a broad spectrum of complex diseases. Disconcertingly, the majority of these GWAS hits occur in non-coding regions of the genome, much of which controls the cell-type-specific expression of genes essential to health. In contrast to gene coding sequences, it is a challenge to understand the function of this non-coding regulatory genome using conventional biochemical techniques in cell lines. The current commentary scrutinizes the field of complex genetics from the standpoint of the large-scale whole-genome functional analysis of the promoters and cis-regulatory elements using chromatin markers. We contrast these large scale quantitative techniques against comparative genomics and in vivo analyses including CRISPR/CAS9 genome editing to determine the functional characteristics of these elements and to understand how polymorphic variation and epigenetic changes within these elements might contribute to complex disease and drug response. Most importantly, we suggest that, although the role of chromatin markers will continue to be important in identifying and characterizing enhancers, more emphasis must be placed on their analysis in relevant in-vivo models that take account of the appropriate cell-type-specific roles of these elements. It is hoped that offering these insights might refocus progress in analyzing the data tsunami of non-coding GWAS and whole-genome sequencing "hits" that threatens to overwhelm progress in the field.
Collapse
Affiliation(s)
| | - Alasdair MacKenzie
- School of Medicine, Medical Sciences and Nutrition, Institute of Medical Sciences, Foresterhill, University of Aberdeen, Aberdeen AB25 2ZD, UK;
| |
Collapse
|
26
|
Fernández A, Morín M, Muñoz‐Santos D, Josa S, Montero A, Rubio‐Fernández M, Cantero M, Fernández J, del Hierro MJ, Castrillo M, Moreno‐Pelayo MÁ, Montoliu L. Simple Protocol for Generating and Genotyping Genome‐Edited Mice With CRISPR‐Cas9 Reagents. ACTA ACUST UNITED AC 2020; 10:e69. [DOI: 10.1002/cpmo.69] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Affiliation(s)
- Almudena Fernández
- National Centre for Biotechnology (CNB)CSIC Madrid Spain
- CIBER of Rare Diseases (CIBERER)ISCIII Madrid Spain
| | - Matías Morín
- CIBER of Rare Diseases (CIBERER)ISCIII Madrid Spain
- Servicio de Genética, Ramón y Cajal University HospitalIRYCIS Madrid Spain
| | - Diego Muñoz‐Santos
- National Centre for Biotechnology (CNB)CSIC Madrid Spain
- CIBER of Rare Diseases (CIBERER)ISCIII Madrid Spain
| | - Santiago Josa
- National Centre for Biotechnology (CNB)CSIC Madrid Spain
- CIBER of Rare Diseases (CIBERER)ISCIII Madrid Spain
| | - Andrea Montero
- National Centre for Biotechnology (CNB)CSIC Madrid Spain
- CIBER of Rare Diseases (CIBERER)ISCIII Madrid Spain
| | - Marcos Rubio‐Fernández
- National Centre for Biotechnology (CNB)CSIC Madrid Spain
- CIBER of Rare Diseases (CIBERER)ISCIII Madrid Spain
| | - Marta Cantero
- National Centre for Biotechnology (CNB)CSIC Madrid Spain
- CIBER of Rare Diseases (CIBERER)ISCIII Madrid Spain
| | - Julia Fernández
- National Centre for Biotechnology (CNB)CSIC Madrid Spain
- CIBER of Rare Diseases (CIBERER)ISCIII Madrid Spain
| | | | | | - Miguel Ángel Moreno‐Pelayo
- CIBER of Rare Diseases (CIBERER)ISCIII Madrid Spain
- Servicio de Genética, Ramón y Cajal University HospitalIRYCIS Madrid Spain
| | - Lluís Montoliu
- National Centre for Biotechnology (CNB)CSIC Madrid Spain
- CIBER of Rare Diseases (CIBERER)ISCIII Madrid Spain
| |
Collapse
|