1
|
Nickl P, Jenickova I, Elias J, Kasparek P, Barinka C, Kopkanova J, Sedlacek R. Multistep allelic conversion in mouse pre-implantation embryos by AAV vectors. Sci Rep 2024; 14:20160. [PMID: 39215103 PMCID: PMC11364770 DOI: 10.1038/s41598-024-70853-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 08/21/2024] [Indexed: 09/04/2024] Open
Abstract
Site-specific recombinases (SSRs) are critical for achieving precise spatiotemporal control of engineered alleles. These enzymes play a key role in facilitating the deletion or inversion of loci flanked by recombination sites, resulting in the activation or repression of endogenous genes, selection markers or reporter elements. However, multiple recombination in complex alleles can be laborious. To address this, a new and efficient method using AAV vectors has been developed to simplify the conversion of systems based on Cre, FLP, Dre and Vika recombinases. In this study, we present an effective method for ex vivo allele conversion using Cre, FLP (flippase), Dre, and Vika recombinases, employing adeno-associated viruses (AAV) as delivery vectors. AAVs enable efficient allele conversion with minimal toxicity in a reporter mouse line. Moreover, AAVs facilitate sequential allele conversion, essential for fully converting alleles with multiple recombination sites, typically found in conditional knockout mouse models. While simple allele conversions show a 100% efficiency rate, complex multiple conversions consistently achieve an 80% conversion rate. Overall, this strategy markedly reduces the need for animals and significantly speeds up the process of allele conversion, representing a significant improvement in genome engineering techniques.
Collapse
Affiliation(s)
- Petr Nickl
- Czech Centre for Phenogenomics, Institute of Molecular Genetics of the Czech Academy of Sciences, Prumyslova 595, 252 50, Vestec, Czech Republic.
- Laboratory of Transgenic Models of Diseases, Institute of Molecular Genetics of the Czech Academy of Sciences, Videnska 1083, 142 20, Prague, Czech Republic.
| | - Irena Jenickova
- Czech Centre for Phenogenomics, Institute of Molecular Genetics of the Czech Academy of Sciences, Prumyslova 595, 252 50, Vestec, Czech Republic
| | - Jan Elias
- Czech Centre for Phenogenomics, Institute of Molecular Genetics of the Czech Academy of Sciences, Prumyslova 595, 252 50, Vestec, Czech Republic
- Laboratory of Transgenic Models of Diseases, Institute of Molecular Genetics of the Czech Academy of Sciences, Videnska 1083, 142 20, Prague, Czech Republic
| | - Petr Kasparek
- Czech Centre for Phenogenomics, Institute of Molecular Genetics of the Czech Academy of Sciences, Prumyslova 595, 252 50, Vestec, Czech Republic
- Laboratory of Transgenic Models of Diseases, Institute of Molecular Genetics of the Czech Academy of Sciences, Videnska 1083, 142 20, Prague, Czech Republic
| | - Cyril Barinka
- Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Prumyslova 595, 252 50, Vestec, Czech Republic
| | - Jana Kopkanova
- Czech Centre for Phenogenomics, Institute of Molecular Genetics of the Czech Academy of Sciences, Prumyslova 595, 252 50, Vestec, Czech Republic
| | - Radislav Sedlacek
- Czech Centre for Phenogenomics, Institute of Molecular Genetics of the Czech Academy of Sciences, Prumyslova 595, 252 50, Vestec, Czech Republic.
- Laboratory of Transgenic Models of Diseases, Institute of Molecular Genetics of the Czech Academy of Sciences, Videnska 1083, 142 20, Prague, Czech Republic.
| |
Collapse
|
2
|
Zhang C, Chang F, Miao H, Fu Y, Tong X, Feng Y, Zheng W, Ma X. Construction and application of a multifunctional CHO cell platform utilizing Cre/ lox and Dre/ rox site-specific recombination systems. Front Bioeng Biotechnol 2023; 11:1320841. [PMID: 38173869 PMCID: PMC10761530 DOI: 10.3389/fbioe.2023.1320841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 12/04/2023] [Indexed: 01/05/2024] Open
Abstract
During the development of traditional Chinese hamster ovary (CHO) cell lines, target genes randomly integrate into the genome upon entering the nucleus, resulting in unpredictable productivity of cell clones. The characterization and screening of high-yielding cell lines is a time-consuming and expensive process. Site-specific integration is recognized as an effective approach for overcoming random integration and improving production stability. We have designed a multifunctional expression cassette, called CDbox, which can be manipulated by the site-specific recombination systems Cre/lox and Dre/rox. The CDbox expression cassette was inserted at the Hipp11(H11) locus hotspot in the CHO-K1 genome using CRISPR/Cas9 technology, and a compliant CHO-CDbox cell platform was screened and obtained. The CHO-CDbox cell platform was transformed into a pool of EGFP-expressing cells using Cre/lox recombinase-mediated cassette exchange (RMCE) in only 2 weeks, and this expression remained stable for at least 75 generations without the need for drug stress. Subsequently, we used the Dre/rox system to directly eliminate the EGFP gene. In addition, two practical applications of the CHO-CDbox cell platform were presented. The first was the quick construction of the Pembrolizumab antibody stable expression strain, while the second was a protocol for the integration of surface-displayed and secreted antibodies on CHO cells. The previous research on site-specific integration of CHO cells has always focused on the single functionality of insertion of target genes. This newly developed CHO cell platform is expected to offer expanded applicability for protein production and gene function studies.
Collapse
Affiliation(s)
- Chen Zhang
- State Key Laboratory of Bioreactor Engineering, School of Biotechnology, East China University of Science and Technology, Shanghai, China
| | - Feng Chang
- State Key Laboratory of Bioreactor Engineering, School of Biotechnology, East China University of Science and Technology, Shanghai, China
| | - Hui Miao
- State Key Laboratory of Bioreactor Engineering, School of Biotechnology, East China University of Science and Technology, Shanghai, China
| | - Yunhui Fu
- State Key Laboratory of Bioreactor Engineering, School of Biotechnology, East China University of Science and Technology, Shanghai, China
| | - Xikui Tong
- State Key Laboratory of Bioreactor Engineering, School of Biotechnology, East China University of Science and Technology, Shanghai, China
| | - Yu Feng
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Wenyun Zheng
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Xingyuan Ma
- State Key Laboratory of Bioreactor Engineering, School of Biotechnology, East China University of Science and Technology, Shanghai, China
| |
Collapse
|
3
|
Zhao C, Ries C, Du Y, Zhang J, Sakimura K, Itoi K, Deussing JM. Differential CRH expression level determines efficiency of Cre- and Flp-dependent recombination. Front Neurosci 2023; 17:1163462. [PMID: 37599997 PMCID: PMC10434532 DOI: 10.3389/fnins.2023.1163462] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 07/21/2023] [Indexed: 08/22/2023] Open
Abstract
Corticotropin-releasing hormone expressing (CRH+) neurons are distributed throughout the brain and play a crucial role in shaping the stress responses. Mouse models expressing site-specific recombinases (SSRs) or reporter genes are important tools providing genetic access to defined cell types and have been widely used to address CRH+ neurons and connected brain circuits. Here, we investigated a recently generated CRH-FlpO driver line expanding the CRH system-related tool box. We directly compared it to a previously established and widely used CRH-Cre line with respect to the FlpO expression pattern and recombination efficiency. In the brain, FlpO mRNA distribution fully recapitulates the expression pattern of endogenous Crh. Combining both Crh locus driven SSRs driver lines with appropriate reporters revealed an overall coherence of respective spatial patterns of reporter gene activation validating CRH-FlpO mice as a valuable tool complementing existing CRH-Cre and reporter lines. However, a substantially lower number of reporter-expressing neurons was discerned in CRH-FlpO mice. Using an additional CRH reporter mouse line (CRH-Venus) and a mouse line allowing for conversion of Cre into FlpO activity (CAG-LSL-FlpO) in combination with intersectional and subtractive mouse genetic approaches, we were able to demonstrate that the reduced number of tdTomato reporter expressing CRH+ neurons can be ascribed to the lower recombination efficiency of FlpO compared to Cre recombinase. This discrepancy particularly manifests under conditions of low CRH expression and can be overcome by utilizing homozygous CRH-FlpO mice. These findings have direct experimental implications which have to be carefully considered when targeting CRH+ neurons using CRH-FlpO mice. However, the lower FlpO-dependent recombination efficiency also entails advantages as it provides a broader dynamic range of expression allowing for the visualization of cells showing stress-induced CRH expression which is not detectable in highly sensitive CRH-Cre mice as Cre-mediated recombination has largely been completed in all cells generally possessing the capacity to express CRH. These findings underscore the importance of a comprehensive evaluation of novel SSR driver lines prior to their application.
Collapse
Affiliation(s)
- Chen Zhao
- Molecular Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - Clemens Ries
- Molecular Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - Ying Du
- Molecular Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - Jingwei Zhang
- Molecular Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - Kenji Sakimura
- Department of Animal Model Development, Brain Research Institute, Niigata University, Niigata, Japan
| | - Keiichi Itoi
- Super-Network Brain Physiology, Graduate School of Life Sciences, Tohoku University, Sendai, Japan
| | - Jan M. Deussing
- Molecular Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| |
Collapse
|
4
|
Loder S, Patel N, Morgani S, Sambon M, Leucht P, Levi B. Genetic models for lineage tracing in musculoskeletal development, injury, and healing. Bone 2023; 173:116777. [PMID: 37156345 PMCID: PMC10860167 DOI: 10.1016/j.bone.2023.116777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 04/07/2023] [Accepted: 04/17/2023] [Indexed: 05/10/2023]
Abstract
Musculoskeletal development and later post-natal homeostasis are highly dynamic processes, marked by rapid structural and functional changes across very short periods of time. Adult anatomy and physiology are derived from pre-existing cellular and biochemical states. Consequently, these early developmental states guide and predict the future of the system as a whole. Tools have been developed to mark, trace, and follow specific cells and their progeny either from one developmental state to the next or between circumstances of health and disease. There are now many such technologies alongside a library of molecular markers which may be utilized in conjunction to allow for precise development of unique cell 'lineages'. In this review, we first describe the development of the musculoskeletal system beginning as an embryonic germ layer and at each of the key developmental stages that follow. We then discuss these structures in the context of adult tissues during homeostasis, injury, and repair. Special focus is given in each of these sections to the key genes involved which may serve as markers of lineage or later in post-natal tissues. We then finish with a technical assessment of lineage tracing and the techniques and technologies currently used to mark cells, tissues, and structures within the musculoskeletal system.
Collapse
Affiliation(s)
- Shawn Loder
- Department of Plastic Surgery, University of Pittsburgh, Scaife Hall, Suite 6B, 3550 Terrace Street, Pittsburgh, PA 15261, USA
| | - Nicole Patel
- Center for Organogenesis and Trauma, Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | | | | | | | - Benjamin Levi
- Center for Organogenesis and Trauma, Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
5
|
Dobrowolski P, Buch T, Nagel-Riedasch S. Nomenclature for standardized designation of diploid genotypes in genetically modified laboratory animals. Lab Anim 2023; 57:371-380. [PMID: 37672033 PMCID: PMC10506360 DOI: 10.1177/00236772231175727] [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: 12/07/2022] [Accepted: 04/25/2023] [Indexed: 09/07/2023]
Abstract
Information about the diploid genotype of a gene-modified or mutant laboratory animal is essential for breeding and experimental planning. It is also required for the exchange of animals between different research groups or for communication with professional genotyping service providers. While there are detailed, standardized rules for creating an allele name of a genome modification or mutation, the notation of the diploid genotype after biopsy and genotyping has not been standardized yet. Therefore, a uniform, generally understandable nomenclature for the diploid genotype of gene-modified laboratory animals is needed. With the here-proposed nomenclature recommendations from the Committee on Genetics and Breeding of Laboratory Animals of the German Society for Laboratory Animal Science (GV-SOLAS), we provide a practical, standardized representation of the genotype of gene-modified animals. It is intended to serve as a compact guide for animal care and scientific personnel in animal research facilities and to simplify data exchange between groups and with external service providers.
Collapse
Affiliation(s)
| | - Thorsten Buch
- Institute of Laboratory Animal Science, University of Zurich, Switzerland
| | - Stefan Nagel-Riedasch
- Research Facilities for Experimental Medicine, Charité – Universitätsmedizin Berlin, Germany
| |
Collapse
|
6
|
Tomoi T, Tameshige T, Betsuyaku E, Hamada S, Sakamoto J, Uchida N, Torii K, Shimizu KK, Tamada Y, Urawa H, Okada K, Fukuda H, Tatematsu K, Kamei Y, Betsuyaku S. Targeted single-cell gene induction by optimizing the dually regulated CRE/ loxP system by a newly defined heat-shock promoter and the steroid hormone in Arabidopsis thaliana. FRONTIERS IN PLANT SCIENCE 2023; 14:1171531. [PMID: 37351202 PMCID: PMC10283073 DOI: 10.3389/fpls.2023.1171531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 04/28/2023] [Indexed: 06/24/2023]
Abstract
Multicellular organisms rely on intercellular communication systems to organize their cellular functions. In studies focusing on intercellular communication, the key experimental techniques include the generation of chimeric tissue using transgenic DNA recombination systems represented by the CRE/loxP system. If an experimental system enables the induction of chimeras at highly targeted cell(s), it will facilitate the reproducibility and precision of experiments. However, multiple technical limitations have made this challenging. The stochastic nature of DNA recombination events, especially, hampers reproducible generation of intended chimeric patterns. Infrared laser-evoked gene operator (IR-LEGO), a microscopic system that irradiates targeted cells using an IR laser, can induce heat shock-mediated expression of transgenes, for example, CRE recombinase gene, in the cells. In this study, we developed a method that induces CRE/loxP recombination in the target cell(s) of plant roots and leaves in a highly specific manner. We combined IR-LEGO, an improved heat-shock-specific promoter, and dexamethasone-dependent regulation of CRE. The optimal IR-laser power and irradiation duration were estimated via exhaustive irradiation trials and subsequent statistical modeling. Under optimized conditions, CRE/loxP recombination was efficiently induced without cellular damage. We also found that the induction efficiency varied among tissue types and cellular sizes. The developed method offers an experimental system to generate a precisely designed chimeric tissue, and thus, will be useful for analyzing intercellular communication at high resolution in roots and leaves.
Collapse
Affiliation(s)
- Takumi Tomoi
- Center for Innovation Support, Institute for Social Innovation and Cooperation, Utsunomiya University, Utsunomiya, Japan
- School of Engineering, Utsunomiya University, Utsunomiya, Japan
- Laboratory for Biothermology, National Institute for Basic Biology, Okazaki, Japan
| | - Toshiaki Tameshige
- Kihara Institute for Biological Research (KIBR), Yokohama City University, Yokohama, Japan
- Division of Biological Sciences, Graduate School of Science and Technology, Nara Institute of Science and Technology, Ikoma, Japan
| | - Eriko Betsuyaku
- Department of Life Science, Faculty of Agriculture, Ryukoku University, Otsu, Japan
| | - Saki Hamada
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Tokyo, Japan
| | - Joe Sakamoto
- Laboratory for Biothermology, National Institute for Basic Biology, Okazaki, Japan
- Biophotonics Research Group, Exploratory Research Center on Life and Living Systems (ExCELLS), Okazaki, Japan
| | - Naoyuki Uchida
- Center for Gene Research, Nagoya University, Nagoya, Japan
- Institute of Transformative Bio-Molecules (WPI-ITbM), Nagoya University, Nagoya, Japan
| | - Keiko U. Torii
- Institute of Transformative Bio-Molecules (WPI-ITbM), Nagoya University, Nagoya, Japan
- Department of Molecular Biosciences and Howard Hughes Medical Institute, The University of Texas at Austin, Austin, TX, United States
| | - Kentaro K. Shimizu
- Kihara Institute for Biological Research (KIBR), Yokohama City University, Yokohama, Japan
- Department of Evolutionary Biology and Environmental Studies, University of Zurich, Zürich, Switzerland
| | - Yosuke Tamada
- School of Engineering, Utsunomiya University, Utsunomiya, Japan
- Center for Optical Research and Education (CORE), Utsunomiya University, Utsunomiya, Japan
- Robotics, Engineering and Agriculture-Technology Laboratory (REAL), Utsunomiya University, Utsunomiya, Japan
| | - Hiroko Urawa
- Faculty of Education, Gifu Shotoku Gakuen University, Gifu, Japan
- Laboratory of Plant Organ Development, National Institute for Basic Biology, Okazaki, Japan
| | - Kiyotaka Okada
- Laboratory of Plant Organ Development, National Institute for Basic Biology, Okazaki, Japan
- Ryukoku Extention Center Shiga, Ryukoku University, Otsu, Japan
| | - Hiroo Fukuda
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Tokyo, Japan
- Department of Bioscience and Biotechnology, Faculty of Bioenvironmental Sciences, Kyoto University of Advanced Science, Kyoto, Japan
| | - Kiyoshi Tatematsu
- Laboratory of Plant Organ Development, National Institute for Basic Biology, Okazaki, Japan
- The Graduate University for Advanced Studies (SOKENDAI), Okazaki, Japan
| | - Yasuhiro Kamei
- Laboratory for Biothermology, National Institute for Basic Biology, Okazaki, Japan
- Robotics, Engineering and Agriculture-Technology Laboratory (REAL), Utsunomiya University, Utsunomiya, Japan
- The Graduate University for Advanced Studies (SOKENDAI), Okazaki, Japan
- Optics and Imaging Facility, Trans-Scale Biology Center, National Institute for Basic Biology, Okazaki, Japan
| | - Shigeyuki Betsuyaku
- Department of Life Science, Faculty of Agriculture, Ryukoku University, Otsu, Japan
| |
Collapse
|
7
|
Bülck C, Nyström EE, Koudelka T, Mannbar-Frahm M, Andresen G, Radhouani M, Tran F, Scharfenberg F, Schrell F, Armbrust F, Dahlke E, Zhao B, Vervaeke A, Theilig F, Rosenstiel P, Starkl P, Rosshart SP, Fickenscher H, Tholey A, Hansson GC, Becker-Pauly C. Proteolytic processing of galectin-3 by meprin metalloproteases is crucial for host-microbiome homeostasis. SCIENCE ADVANCES 2023; 9:eadf4055. [PMID: 37000885 PMCID: PMC10065446 DOI: 10.1126/sciadv.adf4055] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 03/02/2023] [Indexed: 06/19/2023]
Abstract
The metalloproteases meprin α and meprin β are highly expressed in the healthy gut but significantly decreased in inflammatory bowel disease, implicating a protective role in mucosal homeostasis. In the colon, meprin α and meprin β form covalently linked heterodimers tethering meprin α to the plasma membrane, therefore presenting dual proteolytic activity in a unique enzyme complex. To unravel its function, we applied N-terminomics and identified galectin-3 as the major intestinal substrate for meprin α/β heterodimers. Galectin-3-deficient and meprin α/β double knockout mice show similar alterations in their microbiome in comparison to wild-type mice. We further demonstrate that meprin α/β heterodimers differentially process galectin-3 upon bacterial infection, in germ-free, conventionally housed (specific pathogen-free), or wildling mice, which in turn regulates the bacterial agglutination properties of galectin-3. Thus, the constitutive cleavage of galectin-3 by meprin α/β heterodimers may play a key role in colon host-microbiome homeostasis.
Collapse
Affiliation(s)
- Cynthia Bülck
- Institute of Biochemistry, University of Kiel, 24118 Kiel, Germany
| | | | - Tomas Koudelka
- Institute of Experimental Medicine, University of Kiel, 24188 Kiel, Germany
| | - Michael Mannbar-Frahm
- Institute of Infection Medicine, University of Kiel and University Medical Center Schleswig-Holstein, 24015 Kiel, Germany
| | - Gerrit Andresen
- Institute of Infection Medicine, University of Kiel and University Medical Center Schleswig-Holstein, 24015 Kiel, Germany
| | - Mariem Radhouani
- Division of Infection Biology, Department of Medicine I, Medical University of Vienna, 1090 Vienna, Austria
| | - Florian Tran
- Institute of Clinical Molecular Biology, Kiel University and University Medical Center Schleswig-Holstein, 24105 Kiel, Germany
| | | | | | - Fred Armbrust
- Institute of Biochemistry, University of Kiel, 24118 Kiel, Germany
| | - Eileen Dahlke
- Institute of Anatomy, University of Kiel, 24118 Kiel, Germany
| | - Bei Zhao
- Department of Microbiome Research, Friedrich-Alexander-University Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Alex Vervaeke
- Division of Infection Biology, Department of Medicine I, Medical University of Vienna, 1090 Vienna, Austria
| | | | - Philip Rosenstiel
- Institute of Clinical Molecular Biology, Kiel University and University Medical Center Schleswig-Holstein, 24105 Kiel, Germany
| | - Philipp Starkl
- Division of Infection Biology, Department of Medicine I, Medical University of Vienna, 1090 Vienna, Austria
| | - Stephan P. Rosshart
- Department of Microbiome Research, Friedrich-Alexander-University Erlangen-Nürnberg, 91054 Erlangen, Germany
- Department of Medicine II (Gastroenterology, Hepatology, Endocrinology, and Infectious Diseases), Medical Center–University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Helmut Fickenscher
- Institute of Infection Medicine, University of Kiel and University Medical Center Schleswig-Holstein, 24015 Kiel, Germany
| | - Andreas Tholey
- Institute of Experimental Medicine, University of Kiel, 24188 Kiel, Germany
| | - Gunnar C. Hansson
- Department of Medical Biochemistry and Cell Biology, University of Gothenburg, 405 30 Gothenburg, Sweden
| | | |
Collapse
|
8
|
Huang DF, Lin CW, Yang TY, Lien CC, Yang CH, Huang HS. An intersectional genetic approach for simultaneous cell type-specific labelling and gene knockout in the mouse. Development 2023; 150:287021. [PMID: 36786332 DOI: 10.1242/dev.201198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 01/17/2023] [Indexed: 02/15/2023]
Abstract
Precise genome manipulation in specific cell types and subtypes in vivo is crucial for neurobiological research because of the cellular heterogeneity of the brain. Site-specific recombinase systems in the mouse, such as Cre-loxP, improve cell type-specific genome manipulation; however, undesirable expression of cell type-specific Cre can occur. This could be due to transient expression during early development, natural expression in more than one cell type, kinetics of recombinases, sensitivity of the Cre reporter, and disruption in cis-regulatory elements by transgene insertion. Moreover, cell subtypes cannot be distinguished in cell type-specific Cre mice. To address these issues, we applied an intersectional genetic approach in mouse using triple recombination systems (Cre-loxP, Flp-FRT and Dre-rox). As a proof of principle, we labelled heterogeneous cell subtypes and deleted target genes within given cell subtypes by labelling neuropeptide Y (NPY)-, calretinin (calbindin 2) (CR)- and cholecystokinin (CCK)-expressing GABAergic neurons in the brain followed by deletion of RNA-binding Fox-1 homolog 3 (Rbfox3) in our engineered mice. Together, our study applies an intersectional genetic approach in vivo to generate engineered mice serving dual purposes of simultaneous cell subtype-specific labelling and gene knockout.
Collapse
Affiliation(s)
- De-Fong Huang
- Graduate Institute of Brain and Mind Sciences, College of Medicine, National Taiwan University, Taipei 10051, Taiwan
| | - Chao-Wen Lin
- Graduate Institute of Brain and Mind Sciences, College of Medicine, National Taiwan University, Taipei 10051, Taiwan
- Department of Ophthalmology, National Taiwan University Hospital, Taipei 100229, Taiwan
- Department of Ophthalmology, College of Medicine, National Taiwan University, Taipei 10051, Taiwan
| | - Tzu-Yin Yang
- Graduate Institute of Brain and Mind Sciences, College of Medicine, National Taiwan University, Taipei 10051, Taiwan
| | - Cheng-Chang Lien
- Institute of Neuroscience, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan
- Brain Research Center, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan
| | - Chang-Hao Yang
- Department of Ophthalmology, National Taiwan University Hospital, Taipei 100229, Taiwan
- Department of Ophthalmology, College of Medicine, National Taiwan University, Taipei 10051, Taiwan
| | - Hsien-Sung Huang
- Graduate Institute of Brain and Mind Sciences, College of Medicine, National Taiwan University, Taipei 10051, Taiwan
| |
Collapse
|
9
|
Abstract
The mouse is one of the most widely used model organisms for genetic study. The tools available to alter the mouse genome have developed over the preceding decades from forward screens to gene targeting in stem cells to the recent influx of CRISPR approaches. In this review, we first consider the history of mice in genetic study, the development of classic approaches to genome modification, and how such approaches have been used and improved in recent years. We then turn to the recent surge of nuclease-mediated techniques and how they are changing the field of mouse genetics. Finally, we survey common classes of alleles used in mice and discuss how they might be engineered using different methods.
Collapse
Affiliation(s)
- James F Clark
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mt. Sinai, New York, New York 10029, USA
| | - Colin J Dinsmore
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mt. Sinai, New York, New York 10029, USA
| | - Philippe Soriano
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mt. Sinai, New York, New York 10029, USA
| |
Collapse
|
10
|
Fenno LE, Ramakrishnan C, Kim YS, Evans KE, Lo M, Vesuna S, Inoue M, Cheung KYM, Yuen E, Pichamoorthy N, Hong ASO, Deisseroth K. Comprehensive Dual- and Triple-Feature Intersectional Single-Vector Delivery of Diverse Functional Payloads to Cells of Behaving Mammals. Neuron 2020; 107:836-853.e11. [PMID: 32574559 PMCID: PMC7687746 DOI: 10.1016/j.neuron.2020.06.003] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 05/08/2020] [Accepted: 05/29/2020] [Indexed: 01/12/2023]
Abstract
The resolution and dimensionality with which biologists can characterize cell types have expanded dramatically in recent years, and intersectional consideration of such features (e.g., multiple gene expression and anatomical parameters) is increasingly understood to be essential. At the same time, genetically targeted technology for writing in and reading out activity patterns for cells in living organisms has enabled causal investigation in physiology and behavior; however, cell-type-specific delivery of these tools (including microbial opsins for optogenetics and genetically encoded Ca2+ indicators) has thus far fallen short of versatile targeting to cells jointly defined by many individually selected features. Here, we develop a comprehensive intersectional targeting toolbox including 39 novel vectors for joint-feature-targeted delivery of 13 molecular payloads (including opsins, indicators, and fluorophores), systematic approaches for development and optimization of new intersectional tools, hardware for in vivo monitoring of expression dynamics, and the first versatile single-virus tools (Triplesect) that enable targeting of triply defined cell types.
Collapse
Affiliation(s)
- Lief E Fenno
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA; Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Charu Ramakrishnan
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Yoon Seok Kim
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Kathryn E Evans
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Maisie Lo
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Sam Vesuna
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Masatoshi Inoue
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Kathy Y M Cheung
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Elle Yuen
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | | | - Alice S O Hong
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Karl Deisseroth
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA; Department of Bioengineering, Stanford University, Stanford, CA 94305, USA; Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
11
|
Abstract
The endocannabinoid system (ECS) is a highly versatile signaling system within the nervous system. Despite its widespread localization, its functions within the context of distinct neural processes are very well discernable and specific. This is remarkable, and the question remains as to how such specificity is achieved. One key player in the ECS is the cannabinoid type 1 receptor (CB1), a G protein-coupled receptor characterized by the complexity of its cell-specific expression, cellular and subcellular localization, and its adaptable regulation of intracellular signaling cascades. CB1 receptors are involved in different synaptic and cellular plasticity processes and in the brain's bioenergetics in a context-specific manner. CB2 receptors are also important in several processes in neurons, glial cells, and immune cells of the brain. As polymorphisms in ECS components, as well as external impacts such as stress and metabolic challenges, can both lead to dysregulated ECS activity and subsequently to possible neuropsychiatric disorders, pharmacological intervention targeting the ECS is a promising therapeutic approach. Understanding the neurobiology of cannabinoid receptor signaling in depth will aid optimal design of therapeutic interventions, minimizing unwanted side effects.
.
Collapse
Affiliation(s)
- Beat Lutz
- Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| |
Collapse
|
12
|
Soni A, Augsburg M, Buchholz F, Pisabarro MT. Nearest-neighbor amino acids of specificity-determining residues influence the activity of engineered Cre-type recombinases. Sci Rep 2020; 10:13985. [PMID: 32814809 PMCID: PMC7438526 DOI: 10.1038/s41598-020-70867-5] [Citation(s) in RCA: 5] [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: 01/22/2020] [Accepted: 08/03/2020] [Indexed: 11/29/2022] Open
Abstract
The tyrosine-type site-specific DNA recombinase Cre recombines its target site, loxP, with high activity and specificity without cross-recombining the target sites of highly related recombinases. Understanding how Cre achieves this precision is key to be able to rationally engineer site-specific recombinases (SSRs) for genome editing applications. Previous work has revealed key residues for target site selectivity in the Cre/loxP and the related Dre/rox recombinase systems. However, enzymes in which these residues were changed to the respective counterpart only showed weak activity on the foreign target site. Here, we use molecular modeling and dynamics simulation techniques to comprehensively explore the mechanisms by which these residues determine target recognition in the context of their flanking regions in the protein-DNA interface, and we establish a structure-based rationale for the design of improved recombination activities. Our theoretical models reveal that nearest-neighbors to the specificity-determining residues are important players for enhancing SSR activity on the foreign target site. Based on the established rationale, we design new Cre variants with improved rox recombination activities, which we validate experimentally. Our work provides new insights into the target recognition mechanisms of Cre-like recombinases and represents an important step towards the rational design of SSRs for applied genome engineering.
Collapse
Affiliation(s)
- Anjali Soni
- Structural Bioinformatics, BIOTEC, TU Dresden, Tatzberg 47-51, 01307, Dresden, Germany
| | - Martina Augsburg
- University Carl Gustav Carus and Medical Faculty, UCC, Medical Systems Biology, TU Dresden, Fetscherstrasse 74, Dresden, Germany
| | - Frank Buchholz
- University Carl Gustav Carus and Medical Faculty, UCC, Medical Systems Biology, TU Dresden, Fetscherstrasse 74, Dresden, Germany
| | - M Teresa Pisabarro
- Structural Bioinformatics, BIOTEC, TU Dresden, Tatzberg 47-51, 01307, Dresden, Germany.
| |
Collapse
|
13
|
Tamura S, Yasuoka Y, Miura H, Takahashi G, Sato M, Ohtsuka M. Thy1 promoter activity in the Rosa26 locus in mice: lessons from Dre-rox conditional expression system. Exp Anim 2020; 69:287-294. [PMID: 32051391 PMCID: PMC7445056 DOI: 10.1538/expanim.20-0002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
The pronuclear injection (PI)-based targeted transgenesis (PITT) method allows the
generation of targeted transgenic (Tg) mice wherein a single copy of a transgene is
integrated into the Rosa26 locus following PI. The
Rosa26 locus allows unbiased ubiquitous expression of integrated
transgenes; however, it remains little known whether tissue-specific promoters retain
their functional properties when placed at the Rosa26 locus. We evaluated
tissue-specific activity and reproducibility of exogenous tissue-specific promoters
targeted to the Rosa26 locus by generating Thy1-Dre/Dre reporter mice
using PITT and assessed spatial expression patterns of the transgenes. The
Thy1 promoter targeted to the Rosa26 locus appeared
active in virtually all Purkinje cells in the cerebellum and hippocampus. However, mosaic
expression of the transgene under the Thy1 promoter was observed in many
other organs. This phenomenon was consistent in all the Tg lines generated by PITT,
indicating a high degree of reproducibility for this experiment.
Collapse
Affiliation(s)
- Saaki Tamura
- Department of Molecular Life Science, Division of Basic Medical Science and Molecular Medicine, Tokai University School of Medicine, 143 Shimokasuya, Isehara, Kanagawa 259-1193, Japan
| | - Yukiko Yasuoka
- Department of Physiology, Kitasato University, School of Medicine, 1-15-1 Kitasato, Minami-ku, Sagamihara, Kanagawa 252-0374, Japan
| | - Hiromi Miura
- Department of Molecular Life Science, Division of Basic Medical Science and Molecular Medicine, Tokai University School of Medicine, 143 Shimokasuya, Isehara, Kanagawa 259-1193, Japan.,Center for Matrix Biology and Medicine, Graduate School of Medicine, Tokai University, 143 Shimokasuya, Isehara, Kanagawa 259-1193, Japan
| | - Gou Takahashi
- Department of Molecular Life Science, Division of Basic Medical Science and Molecular Medicine, Tokai University School of Medicine, 143 Shimokasuya, Isehara, Kanagawa 259-1193, Japan.,Department of Bioproduction, Tokyo University of Agriculture, 196 Yasaka, Abashiri, Hokkaido, 099-2493, Japan
| | - Masahiro Sato
- Section of Gene Expression Regulation, Frontier Science Research Center, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima, Kagoshima 890-8544, Japan
| | - Masato Ohtsuka
- Department of Molecular Life Science, Division of Basic Medical Science and Molecular Medicine, Tokai University School of Medicine, 143 Shimokasuya, Isehara, Kanagawa 259-1193, Japan.,Center for Matrix Biology and Medicine, Graduate School of Medicine, Tokai University, 143 Shimokasuya, Isehara, Kanagawa 259-1193, Japan.,The Institute of Medical Sciences, Tokai University, 143 Shimokasuya, Isehara, Kanagawa 259-1193, Japan
| |
Collapse
|
14
|
Wang YB, de Lartigue G, Page AJ. Dissecting the Role of Subtypes of Gastrointestinal Vagal Afferents. Front Physiol 2020; 11:643. [PMID: 32595525 PMCID: PMC7300233 DOI: 10.3389/fphys.2020.00643] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 05/20/2020] [Indexed: 12/22/2022] Open
Abstract
Gastrointestinal (GI) vagal afferents convey sensory signals from the GI tract to the brain. Numerous subtypes of GI vagal afferent have been identified but their individual roles in gut function and feeding regulation are unclear. In the past decade, technical approaches to selectively target vagal afferent subtypes and to assess their function has significantly progressed. This review examines the classification of GI vagal afferent subtypes and discusses the current available techniques to study vagal afferents. Investigating the distribution of GI vagal afferent subtypes and understanding how to access and modulate individual populations are essential to dissect their fundamental roles in the gut-brain axis.
Collapse
Affiliation(s)
- Yoko B Wang
- Vagal Afferent Research Group, Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
| | - Guillaume de Lartigue
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, United States.,Center for Integrative Cardiovascular and Metabolic Disease, University of Florida, Gainesville, FL, United States
| | - Amanda J Page
- Vagal Afferent Research Group, Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia.,Nutrition, Diabetes and Gut Health, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| |
Collapse
|
15
|
Lanigan TM, Kopera HC, Saunders TL. Principles of Genetic Engineering. Genes (Basel) 2020; 11:E291. [PMID: 32164255 PMCID: PMC7140808 DOI: 10.3390/genes11030291] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 02/28/2020] [Accepted: 03/06/2020] [Indexed: 12/12/2022] Open
Abstract
Genetic engineering is the use of molecular biology technology to modify DNA sequence(s) in genomes, using a variety of approaches. For example, homologous recombination can be used to target specific sequences in mouse embryonic stem (ES) cell genomes or other cultured cells, but it is cumbersome, poorly efficient, and relies on drug positive/negative selection in cell culture for success. Other routinely applied methods include random integration of DNA after direct transfection (microinjection), transposon-mediated DNA insertion, or DNA insertion mediated by viral vectors for the production of transgenic mice and rats. Random integration of DNA occurs more frequently than homologous recombination, but has numerous drawbacks, despite its efficiency. The most elegant and effective method is technology based on guided endonucleases, because these can target specific DNA sequences. Since the advent of clustered regularly interspaced short palindromic repeats or CRISPR/Cas9 technology, endonuclease-mediated gene targeting has become the most widely applied method to engineer genomes, supplanting the use of zinc finger nucleases, transcription activator-like effector nucleases, and meganucleases. Future improvements in CRISPR/Cas9 gene editing may be achieved by increasing the efficiency of homology-directed repair. Here, we describe principles of genetic engineering and detail: (1) how common elements of current technologies include the need for a chromosome break to occur, (2) the use of specific and sensitive genotyping assays to detect altered genomes, and (3) delivery modalities that impact characterization of gene modifications. In summary, while some principles of genetic engineering remain steadfast, others change as technologies are ever-evolving and continue to revolutionize research in many fields.
Collapse
Affiliation(s)
- Thomas M. Lanigan
- Biomedical Research Core Facilities, Vector Core, University of Michigan, Ann Arbor, MI 48109, USA; (T.M.L.); (H.C.K.)
- Department of Internal Medicine, Division of Rheumatology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Huira C. Kopera
- Biomedical Research Core Facilities, Vector Core, University of Michigan, Ann Arbor, MI 48109, USA; (T.M.L.); (H.C.K.)
- Department of Human Genetics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Thomas L. Saunders
- Biomedical Research Core Facilities, Transgenic Animal Model Core, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Internal Medicine, Division of Genetic Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
16
|
Light-mediated control of Gene expression in mammalian cells. Neurosci Res 2020; 152:66-77. [DOI: 10.1016/j.neures.2019.12.018] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Revised: 12/05/2019] [Accepted: 12/06/2019] [Indexed: 01/07/2023]
|
17
|
Kohl J. Parenting - a paradigm for investigating the neural circuit basis of behavior. Curr Opin Neurobiol 2020; 60:84-91. [PMID: 31830690 PMCID: PMC7005672 DOI: 10.1016/j.conb.2019.11.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 11/11/2019] [Accepted: 11/12/2019] [Indexed: 02/06/2023]
Abstract
Parenting is essential for survival and wellbeing in many species. Since it can be performed with little prior experience and entails considerable sacrifices without immediate benefits for the caregiver, this behavior is likely orchestrated by evolutionarily shaped, hard-wired neural circuits. At the same time, experience, environmental factors and internal state also make parenting highly malleable. These characteristics have made parenting an attractive paradigm for linking complex, naturalistic behavior to its underlying neural mechanisms. Recent work - based on the identification of critical neuronal populations and improved tools for dissecting neural circuits - has uncovered novel functional principles and challenged simplistic models of parenting control. A better understanding of the neural basis of parenting will provide crucial clues to how complex behaviors are organized at the level of cells, circuits and computations. Here I review recent progress, discuss emerging functional principles of parental circuits, and outline future opportunities and challenges.
Collapse
Affiliation(s)
- Johannes Kohl
- The Francis Crick Institute, 1 Midland Rd., London NW1 1AT, UK.
| |
Collapse
|
18
|
Calvo M, Davies AJ, Hébert HL, Weir GA, Chesler EJ, Finnerup NB, Levitt RC, Smith BH, Neely GG, Costigan M, Bennett DL. The Genetics of Neuropathic Pain from Model Organisms to Clinical Application. Neuron 2019; 104:637-653. [PMID: 31751545 PMCID: PMC6868508 DOI: 10.1016/j.neuron.2019.09.018] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 09/05/2019] [Accepted: 09/12/2019] [Indexed: 12/14/2022]
Abstract
Neuropathic pain (NeuP) arises due to injury of the somatosensory nervous system and is both common and disabling, rendering an urgent need for non-addictive, effective new therapies. Given the high evolutionary conservation of pain, investigative approaches from Drosophila mutagenesis to human Mendelian genetics have aided our understanding of the maladaptive plasticity underlying NeuP. Successes include the identification of ion channel variants causing hyper-excitability and the importance of neuro-immune signaling. Recent developments encompass improved sensory phenotyping in animal models and patients, brain imaging, and electrophysiology-based pain biomarkers, the collection of large well-phenotyped population cohorts, neurons derived from patient stem cells, and high-precision CRISPR generated genetic editing. We will discuss how to harness these resources to understand the pathophysiological drivers of NeuP, define its relationship with comorbidities such as anxiety, depression, and sleep disorders, and explore how to apply these findings to the prediction, diagnosis, and treatment of NeuP in the clinic.
Collapse
Affiliation(s)
- Margarita Calvo
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alexander J Davies
- Neural Injury Group, Nuffield Department of Clinical Neuroscience, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Harry L Hébert
- Chronic Pain Research Group, Division of Population Health and Genomics, Mackenzie Building, Ninewells Hospital & Medical School, University of Dundee, Dundee, UK
| | - Greg A Weir
- Neural Injury Group, Nuffield Department of Clinical Neuroscience, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | | | - Nanna B Finnerup
- Department of Clinical Medicine, Danish Pain Research Center, Aarhus University, Aarhus 8000, Denmark
| | - Roy C Levitt
- Department of Anesthesiology, Perioperative Medicine and Pain Management, and John T. MacDonald Foundation Department of Human Genetics, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Blair H Smith
- Chronic Pain Research Group, Division of Population Health and Genomics, Mackenzie Building, Ninewells Hospital & Medical School, University of Dundee, Dundee, UK
| | - G Gregory Neely
- Dr. John and Anne Chong Lab for Functional Genomics, Camperdown, University of Sydney, Sydney, NSW, Australia
| | - Michael Costigan
- Departments of Anesthesia and Neurobiology, Children's Hospital Boston and Harvard Medical School, Boston, MA, USA.
| | - David L Bennett
- Neural Injury Group, Nuffield Department of Clinical Neuroscience, John Radcliffe Hospital, University of Oxford, Oxford, UK.
| |
Collapse
|
19
|
Hara T, Verma IM. Modeling Gliomas Using Two Recombinases. Cancer Res 2019; 79:3983-3991. [PMID: 31315836 PMCID: PMC6677610 DOI: 10.1158/0008-5472.can-19-0717] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 05/15/2019] [Accepted: 05/30/2019] [Indexed: 01/21/2023]
Abstract
Development of animal models to investigate the complex ecosystem of malignant gliomas using the Cre/loxP recombination system has significantly contributed to our understanding of the molecular underpinnings of this deadly disease. In these model systems, once the tumor is induced by activation of Cre-recombinase in a tissue-specific manner, further genetic manipulations to explore the progression of tumorigenesis are limited. To expand the application of mouse models for gliomas, we developed glial fibrillary acidic protein (GFAP)-FLP recombinase (FLPo) mice that express FLPo recombinase specifically in GFAP-positive cells. Lentivirus-based in vivo delivery of cancer genes conditioned by FLP/FRT-mediated recombination initiated gliomas in GFAP-FLPo mice. Using the Cre-mediated multifluorescent protein-expressing system, we demonstrated that the GFAP-FLPo mouse model enables the analysis of various stages of gliomagenesis. Collectively, we present a new mouse model that will expand our ability to dissect developmental processes of gliomagenesis and to provide new avenues for therapeutic approaches. SIGNIFICANCE: This study presents a new glioma mouse model derived using lentiviral vectors and two recombination systems that will expand the ability to dissect developmental processes of gliomagenesis.
Collapse
Affiliation(s)
- Toshiro Hara
- Laboratory of Genetics, Salk Institute for Biological Studies, La Jolla, California.
| | - Inder M Verma
- Laboratory of Genetics, Salk Institute for Biological Studies, La Jolla, California
| |
Collapse
|