1
|
Halurkar MS, Inoue O, Singh A, Mukherjee R, Ginugu M, Ahn C, Bonatto Paese CL, Duszynski M, Brugmann SA, Lim HW, Sanchez-Gurmaches J. The widely used Ucp1-Cre transgene elicits complex developmental and metabolic phenotypes. Nat Commun 2025; 16:770. [PMID: 39824816 PMCID: PMC11742029 DOI: 10.1038/s41467-024-54763-4] [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: 12/12/2023] [Accepted: 11/20/2024] [Indexed: 01/20/2025] Open
Abstract
Bacterial artificial chromosome transgenic models, including most Cre-recombinases, enable potent interrogation of gene function in vivo but require rigorous validation as limitations emerge. Due to its high relevance to metabolic studies, we perform comprehensive analysis of the Ucp1-CreEvdr line which is widely used for brown fat research. Hemizygotes exhibit major brown and white fat transcriptomic dysregulation, indicating potential altered tissue function. Ucp1-CreEvdr homozygotes also show high mortality, tissue specific growth defects, and craniofacial abnormalities. Mapping the transgene insertion site reveals insertion in chromosome 1 accompanied by large genomic alterations disrupting several genes expressed in a range of tissues. Notably, Ucp1-CreEvdr transgene retains an extra Ucp1 gene copy that may be highly expressed under high thermogenic burden. Our multi-faceted analysis highlights a complex phenotype arising from the presence of the Ucp1-CreEvdr transgene independently of intended genetic manipulations. Overall, comprehensive validation of transgenic mice is imperative to maximize discovery while mitigating unexpected, off-target effects.
Collapse
Affiliation(s)
- Manasi Suchit Halurkar
- Division of Endocrinology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Oto Inoue
- Division of Endocrinology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Archana Singh
- Division of Endocrinology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Rajib Mukherjee
- Division of Endocrinology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Pioneering Medicines, 140 First St., Suite 302, Cambridge, MA, USA
| | - Meghana Ginugu
- Division of Endocrinology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Christopher Ahn
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Christian Louis Bonatto Paese
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Surgery, Division of Plastic Surgery, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Molly Duszynski
- Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Samantha A Brugmann
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Surgery, Division of Plastic Surgery, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Hee-Woong Lim
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Joan Sanchez-Gurmaches
- Division of Endocrinology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
- Center for Stem Cell and Organoid Medicine (CuSTOM), Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
| |
Collapse
|
2
|
Zhu Z, Lu S, Wang H, Wang F, Xu W, Zhu Y, Xue J, Yang L. Innovations in Transgene Integration Analysis: A Comprehensive Review of Enrichment and Sequencing Strategies in Biotechnology. ACS APPLIED MATERIALS & INTERFACES 2025; 17:2716-2735. [PMID: 39760503 DOI: 10.1021/acsami.4c14208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2025]
Abstract
Understanding the integration of transgene DNA (T-DNA) in transgenic crops, animals, and clinical applications is paramount for ensuring the stability and expression of inserted genes, which directly influence desired traits and therapeutic outcomes. Analyzing T-DNA integration patterns is essential for identifying potential unintended effects and evaluating the safety and environmental implications of genetically modified organisms (GMOs). This knowledge is crucial for regulatory compliance and fostering public trust in biotechnology by demonstrating transparency in genetic modifications. This review highlights recent advancements in T-DNA integration analysis, specifically focusing on targeted DNA enrichment and sequencing strategies. We examine key technologies, such as polymerase chain reaction (PCR)-based methods, hybridization capture, RNA/DNA-guided endonuclease-mediated enrichment, and high-throughput resequencing, emphasizing their contributions to enhancing precision and efficiency in transgene integration analysis. We discuss the principles, applications, and recent developments in these techniques, underscoring their critical role in advancing biotechnological products. Additionally, we address the existing challenges and future directions in the field, offering a comprehensive overview of how innovative DNA-targeted enrichment and sequencing strategies are reshaping biotechnology and genomics.
Collapse
Affiliation(s)
- Zaobing Zhu
- Joint International Research Laboratory of Metabolic and Developmental Sciences, Yazhou Bay Institute of Deepsea Sci-Tech, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, People's Republic of China
- Institute of Plant Protection, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, People's Republic of China
- Zhejiang Yuzhi Biotechnology Company, Limited, Ningbo 315032, People's Republic of China
| | - Shengtao Lu
- Joint International Research Laboratory of Metabolic and Developmental Sciences, Yazhou Bay Institute of Deepsea Sci-Tech, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, People's Republic of China
- Zhejiang Yuzhi Biotechnology Company, Limited, Ningbo 315032, People's Republic of China
| | - Hongchun Wang
- Institute of Plant Protection, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, People's Republic of China
| | - Fan Wang
- Joint International Research Laboratory of Metabolic and Developmental Sciences, Yazhou Bay Institute of Deepsea Sci-Tech, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, People's Republic of China
| | - Wenting Xu
- Joint International Research Laboratory of Metabolic and Developmental Sciences, Yazhou Bay Institute of Deepsea Sci-Tech, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, People's Republic of China
| | - Yulei Zhu
- Institute of Plant Protection, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, People's Republic of China
| | - Jing Xue
- Institute of Plant Protection, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, People's Republic of China
| | - Litao Yang
- Joint International Research Laboratory of Metabolic and Developmental Sciences, Yazhou Bay Institute of Deepsea Sci-Tech, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, People's Republic of China
- Zhejiang Yuzhi Biotechnology Company, Limited, Ningbo 315032, People's Republic of China
| |
Collapse
|
3
|
Smirnov A, Nurislamov A, Koncevaya G, Serova I, Kabirova E, Chuyko E, Maltceva E, Savoskin M, Zadorozhny D, Svyatchenko VA, Protopopova EV, Taranov OS, Legostaev SS, Loktev VB, Serov O, Battulin N. Characterizing a lethal CAG-ACE2 transgenic mouse model for SARS-CoV-2 infection using Cas9-enhanced nanopore sequencing. Transgenic Res 2024; 33:453-466. [PMID: 39320390 DOI: 10.1007/s11248-024-00413-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 09/10/2024] [Indexed: 09/26/2024]
Abstract
The SARS-CoV-2 pandemic has underscored the necessity for functional transgenic animal models for testing. Mouse lines with overexpression of the human receptor ACE2 serve as the common animal model to study COVID-19 infection. Overexpression of ACE2 under a strong ubiquitous promoter facilitates convenient and sensitive testing of COVID-19 pathology. We performed pronuclear microinjections using a 5 kb CAG-ACE2 linear transgene construct and identified three founder lines with 140, 72, and 73 copies, respectively. Two of these lines were further analyzed for ACE2 expression profiles and sensitivity to SARS-CoV-2 infection. Both lines expressed ACE2 in all organs analyzed. Embryonic fibroblast cell lines derived from transgenic embryos demonstrated severe cytopathic effects following infection, even at low doses of SARS-CoV-2 (0,1-1.0 TCID50). Infected mice from the two lines began to show COVID-19 clinical signs three days post-infection and succumbed between days 4 and 7. Histological examination of lung tissues from terminally ill mice revealed severe pathological alterations. To further characterize the integration site in one of the lines, we applied nanopore sequencing combined with Cas9 enrichment to examine the internal transgene concatemer structure. Oxford Nanopore sequencing (ONT) is becoming the gold standard for transgene insert characterization, but it is relatively inefficient without targeted region enrichment. We digested genomic DNA with Cas9 and gRNA against the ACE2 transgene to create ends suitable for ONT adapter ligation. ONT data analysis revealed that most of the transgene copies were arranged in a head-to-tail configuration, with palindromic junctions being rare. We also detected occasional plasmid backbone fragments within the concatemer, likely co-purified during transgene gel extraction, which is a common occurrence in pronuclear microinjections.
Collapse
Affiliation(s)
- Alexander Smirnov
- Institute of Cytology and Genetics SB RAS, Novosibirsk, Russia, 630090
| | - Artem Nurislamov
- Institute of Cytology and Genetics SB RAS, Novosibirsk, Russia, 630090
- Novosibirsk State University, Novosibirsk, Russia, 630090
| | - Galina Koncevaya
- Institute of Cytology and Genetics SB RAS, Novosibirsk, Russia, 630090
| | - Irina Serova
- Institute of Cytology and Genetics SB RAS, Novosibirsk, Russia, 630090
| | - Evelyn Kabirova
- Institute of Cytology and Genetics SB RAS, Novosibirsk, Russia, 630090
| | - Eduard Chuyko
- Institute of Cytology and Genetics SB RAS, Novosibirsk, Russia, 630090
| | | | - Maxim Savoskin
- Novosibirsk State University, Novosibirsk, Russia, 630090
| | - Daniil Zadorozhny
- Center for Genetics and Life Science, Sirius University of Science and Technology, Sochi, Russia, 354340
| | - Victor A Svyatchenko
- State Research Center for Virology and Biotechnology "Vector", Novosibirsk, Koltsovo, Russia, 630559
| | - Elena V Protopopova
- State Research Center for Virology and Biotechnology "Vector", Novosibirsk, Koltsovo, Russia, 630559
| | - Oleg S Taranov
- State Research Center for Virology and Biotechnology "Vector", Novosibirsk, Koltsovo, Russia, 630559
| | - Stanislav S Legostaev
- State Research Center for Virology and Biotechnology "Vector", Novosibirsk, Koltsovo, Russia, 630559
| | - Valery B Loktev
- Institute of Cytology and Genetics SB RAS, Novosibirsk, Russia, 630090
- Novosibirsk State University, Novosibirsk, Russia, 630090
- State Research Center for Virology and Biotechnology "Vector", Novosibirsk, Koltsovo, Russia, 630559
| | - Oleg Serov
- Institute of Cytology and Genetics SB RAS, Novosibirsk, Russia, 630090
- Novosibirsk State University, Novosibirsk, Russia, 630090
| | - Nariman Battulin
- Institute of Cytology and Genetics SB RAS, Novosibirsk, Russia, 630090.
- Novosibirsk State University, Novosibirsk, Russia, 630090.
| |
Collapse
|
4
|
Adams PE, Thies JL, Sutton JM, Millwood JD, Caldwell GA, Caldwell KA, Fierst JL. Identifying transgene insertions in Caenorhabditis elegans genomes with Oxford Nanopore sequencing. PeerJ 2024; 12:e18100. [PMID: 39285918 PMCID: PMC11404476 DOI: 10.7717/peerj.18100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 08/26/2024] [Indexed: 09/19/2024] Open
Abstract
Genetically modified organisms are commonly used in disease research and agriculture but the precise genomic alterations underlying transgenic mutations are often unknown. The position and characteristics of transgenes, including the number of independent insertions, influences the expression of both transgenic and wild-type sequences. We used long-read, Oxford Nanopore Technologies (ONT) to sequence and assemble two transgenic strains of Caenorhabditis elegans commonly used in the research of neurodegenerative diseases: BY250 (pPdat-1::GFP) and UA44 (GFP and human α-synuclein), a model for Parkinson's research. After scaffolding to the reference, the final assembled sequences were ∼102 Mb with N50s of 17.9 Mb and 18.0 Mb, respectively, and L90s of six contiguous sequences, representing chromosome-level assemblies. Each of the assembled sequences contained more than 99.2% of the Nematoda BUSCO genes found in the C. elegans reference and 99.5% of the annotated C. elegans reference protein-coding genes. We identified the locations of the transgene insertions and confirmed that all transgene sequences were inserted in intergenic regions, leaving the organismal gene content intact. The transgenic C. elegans genomes presented here will be a valuable resource for Parkinson's research as well as other neurodegenerative diseases. Our work demonstrates that long-read sequencing is a fast, cost-effective way to assemble genome sequences and characterize mutant lines and strains.
Collapse
Affiliation(s)
- Paula E Adams
- Department of Biological Sciences, Auburn University, Auburn, AL, United States of America
- Department of Biological Sciences, University of Alabama - Tuscaloosa, Tuscaloosa, AL, United States of America
| | - Jennifer L Thies
- Department of Biological Sciences, University of Alabama - Tuscaloosa, Tuscaloosa, AL, United States of America
- Curriculum in Toxicology and Environmental Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - John M Sutton
- Department of Biological Sciences, University of Alabama - Tuscaloosa, Tuscaloosa, AL, United States of America
- Absci, Vancouver, WA, United States of America
| | - Joshua D Millwood
- Department of Biological Sciences, University of Alabama - Tuscaloosa, Tuscaloosa, AL, United States of America
- Department of Biological and Environmental Sciences, University of West Alabama, Livingston, AL, United States of America
| | - Guy A Caldwell
- Department of Biological Sciences, University of Alabama - Tuscaloosa, Tuscaloosa, AL, United States of America
| | - Kim A Caldwell
- Department of Biological Sciences, University of Alabama - Tuscaloosa, Tuscaloosa, AL, United States of America
| | - Janna L Fierst
- Department of Biological Sciences, Florida International University, Miami, FL, United States of America
- Biomolecular Sciences Institute, Florida International University, Miami, FL, United States of America
| |
Collapse
|
5
|
Perl AJ, Liu H, Hass M, Adhikari N, Chaturvedi P, Hu YC, Jiang R, Liu Y, Kopan R. Reduced Nephron Endowment in Six2-TGCtg Mice Is Due to Six3 Misexpression by Aberrant Enhancer-Promoter Interactions in the Transgene. J Am Soc Nephrol 2024; 35:566-577. [PMID: 38447671 PMCID: PMC11149036 DOI: 10.1681/asn.0000000000000324] [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: 02/03/2023] [Accepted: 02/27/2024] [Indexed: 03/08/2024] Open
Abstract
Key Points Aberrant enhancer–promoter interactions detected by Hi-C drive ectopic expression of Six3 in the Six2TGCtg line. Disruption of Six3 in the Six2TGCtg line restores nephron number, implicating SIX3 interference with SIX2 function in nephron progenitor cell renewal. Background Lifelong kidney function relies on the complement of nephrons generated during mammalian development from a mesenchymal nephron progenitor cell population. Low nephron endowment confers increased susceptibility to CKD. Reduced nephron numbers in the popular Six2TGC transgenic mouse line may be due to disruption of a regulatory gene at the integration site and/or ectopic expression of a gene(s) contained within the transgene. Methods Targeted locus amplification was performed to identify the integration site of the Six2TGC transgene. Genome-wide chromatin conformation capture (Hi-C) datasets were generated from nephron progenitor cells isolated from the Six2TGC +/tg mice, the Cited1 CreERT2/+ control mice, and the Six2TGC +/tg ; Tsc1 +/Flox mice that exhibited restored nephron number compared with Six2TGC +/tg mice. Modified transgenic mice lacking the C-terminal domain of Six3 were used to evaluate the mechanism of nephron number reduction in the Six2TGC +/tg mouse line. Results Targeted locus amplification revealed integration of the Six2TGC transgene within an intron of Cntnap5a on chr1, and Hi-C analysis mapped the precise integration of Six2TGC and Cited1 CreERT2 transgenes to chr1 and chr14, respectively. No changes in topology, accessibility, or expression were observed within the 50-megabase region centered on Cntnap5a in Six2TGC +/tg mice compared with control mice. By contrast, we identified an aberrant regulatory interaction between a Six2 distal enhancer and the Six3 promoter contained within the transgene. Increasing the Six2TGC tg to Six2 locus ratio or removing one Six2 allele in Six2TGC +/tg mice caused severe renal hypoplasia. Furthermore, clustered regularly interspaced short palindromic repeats disruption of Six3 within the transgene (Six2TGC ∆Six3CT ) restored nephron endowment to wild-type levels and abolished the stoichiometric effect. Conclusions These findings broadly demonstrate the utility of Hi-C data in mapping transgene integration sites and architecture. Data from genetic and biochemical studies together suggest that in Six2TGC kidneys, SIX3 interferes with SIX2 function in nephron progenitor cell renewal through its C-terminal domain.
Collapse
Affiliation(s)
- Alison J. Perl
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Han Liu
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Matthew Hass
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
- Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Nirpesh Adhikari
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Praneet Chaturvedi
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Yueh-Chiang Hu
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Rulang Jiang
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Yaping Liu
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Raphael Kopan
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| |
Collapse
|
6
|
Halurkar MS, Inoue O, Mukherjee R, Paese CLB, Duszynski M, Brugmann SA, Lim HW, Sanchez-Gurmaches J. The widely used Ucp1-CreEvdr transgene elicits complex developmental and metabolic phenotypes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.20.563165. [PMID: 37904917 PMCID: PMC10614962 DOI: 10.1101/2023.10.20.563165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/01/2023]
Abstract
Bacterial artificial chromosome transgenic models, including most Cre-recombinases, enable potent interrogation of gene function in vivo but require rigorous validation as limitations emerge. Due to its high relevance to metabolic studies, we performed comprehensive analysis of the Ucp1-CreEvdr line which is widely used for brown fat research. Hemizygotes exhibited major brown and white fat transcriptomic dysregulation, indicating potential altered tissue function. Ucp1-CreEvdr homozygotes also show high mortality, growth defects, and craniofacial abnormalities. Mapping the transgene insertion site revealed insertion in chromosome 1 accompanied by large genomic alterations disrupting several genes expressed in a range of tissues. Notably, Ucp1-CreEvdr transgene retains an extra Ucp1 gene copy that may be highly expressed under high thermogenic burden. Our multi-faceted analysis highlights a complex phenotype arising from the presence of the Ucp1-CreEvdr transgene independently of the intended genetic manipulations. Overall, comprehensive validation of transgenic mice is imperative to maximize discovery while mitigating unexpected, off-target effects.
Collapse
Affiliation(s)
- Manasi Suchit Halurkar
- Division of Endocrinology, Cincinnati Children’s Hospital Medical Center Cincinnati, OH 45229, USA
| | - Oto Inoue
- Division of Endocrinology, Cincinnati Children’s Hospital Medical Center Cincinnati, OH 45229, USA
| | - Rajib Mukherjee
- Division of Endocrinology, Cincinnati Children’s Hospital Medical Center Cincinnati, OH 45229, USA
| | | | - Molly Duszynski
- Division of Molecular Cardiovascular Biology, Cincinnati Children’s Hospital Medical Center Cincinnati, OH 45229, USA
| | - Samantha A. Brugmann
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center Cincinnati, OH 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45220, USA
- Department of Surgery, Division of Plastic Surgery, University of Cincinnati College of Medicine, Cincinnati, OH 45220, USA
| | - Hee-Woong Lim
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center Cincinnati, OH 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45220, USA
| | - Joan Sanchez-Gurmaches
- Division of Endocrinology, Cincinnati Children’s Hospital Medical Center Cincinnati, OH 45229, USA
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center Cincinnati, OH 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45220, USA
| |
Collapse
|
7
|
Perl AJ, Liu H, Hass M, Adhikari N, Chaturvedi P, Hu YC, Jiang R, Liu Y, Kopan R. Reduced nephron endowment in the common Six2-TGC tg mouse line is due to Six3 misexpression by aberrant enhancer-promoter interactions in the transgene. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.06.561202. [PMID: 37873415 PMCID: PMC10592608 DOI: 10.1101/2023.10.06.561202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Lifelong kidney function relies on the complement of nephrons generated during mammalian development from a mesenchymal nephron progenitor cell (NPC) population. Low nephron endowment confers increased susceptibility to chronic kidney disease. We asked whether reduced nephron numbers in the popular Six2TGC transgenic mouse line 1 was due to disruption of a regulatory gene at the integration site or to ectopic expression of a gene(s) contained within the transgene. Targeted locus amplification identified integration of the Six2TGC transgene within an intron of Cntnap5a on chr1. We generated Hi-C datasets from NPCs isolated from the Six2TGC tg/+ mice, the Cited1 CreERT2/+ control mice, and the Six2TGC tg/+ ; Tsc1 +/Flox,2 mice that exhibited restored nephron number compared with Six2TGC tg/+ mice, and mapped the precise integration of Six2TGC and Cited1 CreERT2 transgenes to chr1 and chr14, respectively. No changes in topology, accessibility, or expression were observed within the 50-megabase region centered on Cntnap5a in Six2TGC tg/+ mice compared with control mice. By contrast, we identified an aberrant regulatory interaction between a Six2 distal enhancer and the Six3 promoter contained within the transgene. Increasing the Six2TGC tg to Six2 locus ratio or removing one Six2 allele in Six2TGC tg/+ mice, caused severe renal hypoplasia. Furthermore, CRISPR disruption of Six3 within the transgene ( Six2TGC Δ Six3CT ) restored nephron endowment to wildtype levels and abolished the stoichiometric effect. Data from genetic and biochemical studies together suggest that in Six2TGC, SIX3 interferes with SIX2 function in NPC renewal through its C-terminal domain. Significance Using high-resolution chromatin conformation and accessibility datasets we mapped the integration site of two popular transgenes used in studies of nephron progenitor cells and kidney development. Aberrant enhancer-promoter interactions drive ectopic expression of Six3 in the Six2TGC tg line which was correlated with disruption of nephrogenesis. Disruption of Six3 within the transgene restored nephron numbers to control levels; further genetic and biochemical studies suggest that Six3 interferes with Six2 -mediated regulation of NPC renewal.
Collapse
|
8
|
Bryant WB, Yang A, Griffin SH, Zhang W, Rafiq AM, Han W, Deak F, Mills MK, Long X, Miano JM. CRISPR-Cas9 Long-Read Sequencing for Mapping Transgenes in the Mouse Genome. CRISPR J 2023; 6:163-175. [PMID: 37071672 PMCID: PMC10123806 DOI: 10.1089/crispr.2022.0099] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 02/12/2023] [Indexed: 04/19/2023] Open
Abstract
Microinjected transgenes, both large and small, are known to insert randomly into the mouse genome. Traditional methods of mapping a transgene are challenging, thus complicating breeding strategies and accurate interpretation of phenotypes, particularly when a transgene disrupts critical coding or noncoding sequences. As the vast majority of transgenic mouse lines remain unmapped, we developed CRISPR-Cas9 Long-Read Sequencing (CRISPR-LRS) to ascertain transgene integration loci. This novel approach mapped a wide size range of transgenes and uncovered more complex transgene-induced host genome re-arrangements than previously appreciated. CRISPR-LRS offers a facile, informative approach to establish robust breeding practices and will enable researchers to study a gene without confounding genetic issues. Finally, CRISPR-LRS will find utility in rapidly and accurately interrogating gene/genome editing fidelity in experimental and clinical settings.
Collapse
Affiliation(s)
- W. Bart Bryant
- Department of Medicine and Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| | - Allison Yang
- Department of Medicine and Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| | - Susan H. Griffin
- Department of Medicine and Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| | - Wei Zhang
- Department of Medicine and Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| | - Ashiq M. Rafiq
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| | - Weiping Han
- Department of Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, Republic of Singapore
| | - Ferenc Deak
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| | - Mary Katherine Mills
- Department of Department of Biology and Geology, University of South Carolina Aiken, Aiken, South Carolina, USA
| | - Xiaochun Long
- Department of Medicine and Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| | - Joseph M. Miano
- Department of Medicine and Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| |
Collapse
|
9
|
Burnet G, Bowles J, Spiller CM. Isolation and Culture Techniques for Fetal Mouse Germ Cells. Methods Mol Biol 2023; 2677:221-231. [PMID: 37464245 DOI: 10.1007/978-1-0716-3259-8_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/20/2023]
Abstract
The fetal gonad contains a great variety of differentiating cell populations, of which germ cells make up a relatively small percentage. In order to study germ cell-specific gene and protein expression, as well as determine direct effects of signaling molecules, it is necessary to prepare enriched populations of germ cells and maintain them in culture for several hours to multiple days. The protocols in this chapter are designed to provide a guide for the isolation or enrichment of primordial germ cells (from 9.5 days post coitum (dpc) to 18.5 dpc) by flow cytometry (Subheading 3.1) or magnetic sorting (Subheading 3.2), followed by feeder-free primary germ cell culture (Subheading 3.3).
Collapse
Affiliation(s)
- Guillaume Burnet
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, Australia
| | - Josephine Bowles
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, Australia
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - Cassy M Spiller
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, Australia.
| |
Collapse
|
10
|
Yu C, Caothien R, Pham A, Tam L, Alcantar T, Bacarro N, Reyes J, Jackson M, Nakao B, Roose-Girma M. ASIS-Seq: Transgene Insertion Site Mapping by Nanopore Adaptive Sampling. Methods Mol Biol 2023; 2631:135-153. [PMID: 36995666 DOI: 10.1007/978-1-0716-2990-1_5] [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
Generation of transgenic mice by direct microinjection of foreign DNA into fertilized ova has become a routine technique in biomedical research. It remains an essential tool for studying gene expression, developmental biology, genetic disease models, and their therapies. However, the random integration of foreign DNA into the host genome that is inherent to this technology can lead to confounding effects associated with insertional mutagenesis and transgene silencing. Locations of most transgenic lines remain unknown because the methods are often burdensome (Nicholls et al., G3: Genes Genomes Genetics 9:1481-1486, 2019) or have limitations (Goodwin et al., Genome Research 29:494-505, 2019). Here, we present a method that we call Adaptive Sampling Insertion Site Sequencing (ASIS-Seq) to locate transgene integration sites using targeted sequencing on Oxford Nanopore Technologies' (ONT) sequencers. ASIS-Seq requires only about 3 ug of genomic DNA, 3 hours of hands-on sample preparation time, and 3 days of sequencing time to locate transgenes in a host genome.
Collapse
Affiliation(s)
- Charles Yu
- Genentech, Inc., Department of Molecular Biology, South San Francisco, CA, USA
| | - Roger Caothien
- Genentech, Inc., Department of Molecular Biology, South San Francisco, CA, USA
| | - Anna Pham
- Genentech, Inc., Department of Molecular Biology, South San Francisco, CA, USA
| | - Lucinda Tam
- Genentech, Inc., Department of Molecular Biology, South San Francisco, CA, USA
| | - Tuija Alcantar
- Genentech, Inc., Department of Molecular Biology, South San Francisco, CA, USA
| | - Natasha Bacarro
- Genentech, Inc., Department of Molecular Biology, South San Francisco, CA, USA
| | - Juan Reyes
- Genentech, Inc., Department of Molecular Biology, South San Francisco, CA, USA
| | - Marques Jackson
- Genentech, Inc., Department of Molecular Biology, South San Francisco, CA, USA
| | - Brian Nakao
- Genentech, Inc., Department of Molecular Biology, South San Francisco, CA, USA
| | - Merone Roose-Girma
- Genentech, Inc., Department of Molecular Biology, South San Francisco, CA, USA.
| |
Collapse
|
11
|
Warthi G, Faulkner JL, Doja J, Ghanam AR, Gao P, Yang AC, Slivano OJ, Barris CT, Kress TC, Zawieja SD, Griffin SH, Xie X, Ashworth A, Christie CK, Bryant WB, Kumar A, Davis MJ, Long X, Gan L, de Chantemèle EJB, Lyu Q, Miano JM. Generation and Comparative Analysis of an Itga8-CreER T2 Mouse with Preferential Activity in Vascular Smooth Muscle Cells. NATURE CARDIOVASCULAR RESEARCH 2022; 1:1084-1100. [PMID: 36424917 PMCID: PMC9681021 DOI: 10.1038/s44161-022-00162-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Accepted: 09/29/2022] [Indexed: 11/12/2022]
Abstract
All current smooth muscle cell (SMC) Cre mice similarly recombine floxed alleles in vascular and visceral SMCs. Here, we present an Itga8-CreER T2 knock-in mouse and compare its activity with a Myh11-CreER T2 mouse. Both Cre drivers demonstrate equivalent recombination in vascular SMCs. However, Myh11-CreER T2 mice, but not Itga8-CreER T2 mice, display high activity in visceral SMC-containing tissues such as intestine, show early tamoxifen-independent activity, and produce high levels of CreERT2 protein. Whereas Myh11-CreER T2 -mediated knockout of serum response factor (Srf) causes a lethal intestinal phenotype precluding analysis of the vasculature, loss of Srf with Itga8-CreER T2 (Srf Itga8 ) yields viable mice with no evidence of intestinal pathology. Male and female Srf Itga8 mice exhibit vascular contractile incompetence, and angiotensin II causes elevated blood pressure in wild type, but not Srf Itga8 , male mice. These findings establish the Itga8-CreER T2 mouse as an alternative to existing SMC Cre mice for unfettered phenotyping of vascular SMCs following selective gene loss.
Collapse
Affiliation(s)
- Ganesh Warthi
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia 30912
| | - Jessica L. Faulkner
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia 30912
| | - Jaser Doja
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia 30912
| | - Amr R. Ghanam
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia 30912
| | - Pan Gao
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia 30912
| | - Allison C. Yang
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia 30912
| | - Orazio J. Slivano
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia 30912
| | - Candee T. Barris
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia 30912
| | - Taylor C. Kress
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia 30912
| | - Scott D. Zawieja
- Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, MO 65212
| | - Susan H. Griffin
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia 30912
| | - Xiaoling Xie
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, Georgia 30912
| | - Alan Ashworth
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, 94158
| | - Christine K. Christie
- Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, New York 14642
| | - William B. Bryant
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia 30912
| | - Ajay Kumar
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia 30912
| | - Michael J. Davis
- Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, MO 65212
| | - Xiaochun Long
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia 30912
| | - Lin Gan
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, Georgia 30912
| | | | - Qing Lyu
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia 30912
- Biomedical and Health Institute, Chongqing Institute of Green and Intelligence Technology, Chongqing, China 400714
- Chongqing General Hospital, Chongqing, China 401147
| | - Joseph M. Miano
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia 30912
| |
Collapse
|
12
|
Ghanam AR, Bryant WB, Miano JM. Of mice and human-specific long noncoding RNAs. Mamm Genome 2022; 33:281-292. [PMID: 35106622 PMCID: PMC8806012 DOI: 10.1007/s00335-022-09943-2] [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: 10/12/2021] [Accepted: 01/10/2022] [Indexed: 01/05/2023]
Abstract
The number of human LncRNAs has now exceeded all known protein-coding genes. Most studies of human LncRNAs have been conducted in cell culture systems where various mechanisms of action have been worked out. On the other hand, efforts to elucidate the function of human LncRNAs in an in vivo setting have been limited. In this brief review, we highlight some strengths and weaknesses of studying human LncRNAs in the mouse. Special consideration is given to bacterial artificial chromosome transgenesis and genome editing. The integration of these technical innovations offers an unprecedented opportunity to complement and extend the expansive literature of cell culture models for the study of human LncRNAs. Two different examples of how BAC transgenesis and genome editing can be leveraged to gain insight into human LncRNA regulation and function in mice are presented: the random integration of a vascular cell-enriched LncRNA and a targeted approach for a new LncRNA immediately upstream of the ACE2 gene, which encodes the receptor for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the etiologic agent underlying the coronavirus disease-19 (COVID-19) pandemic.
Collapse
Affiliation(s)
- Amr R Ghanam
- Vascular Biology Center, Department of Medicine, Medical College of Georgia at Augusta University, 1460 Laney Walker Blvd, CL-3060, Augusta, GA, 30912, USA
| | - William B Bryant
- Vascular Biology Center, Department of Medicine, Medical College of Georgia at Augusta University, 1460 Laney Walker Blvd, CL-3060, Augusta, GA, 30912, USA
| | - Joseph M Miano
- Vascular Biology Center, Department of Medicine, Medical College of Georgia at Augusta University, 1460 Laney Walker Blvd, CL-3060, Augusta, GA, 30912, USA.
| |
Collapse
|
13
|
Wang J, Lu C, Wei S. Whole-genome sequencing identifies I-SceI-mediated transgene integration sites in Xenopus tropicalis snai2:eGFP line. G3 (BETHESDA, MD.) 2022; 12:jkac037. [PMID: 35171990 PMCID: PMC9073676 DOI: 10.1093/g3journal/jkac037] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 01/21/2022] [Indexed: 11/23/2022]
Abstract
Transgenesis with the meganuclease I-SceI is a safe and efficient method, but the underlying mechanisms remain unclear due to the lack of information on transgene localization. Using I-SceI, we previously developed a transgenic Xenopus tropicalis line expressing enhanced green fluorescent protein driven by the neural crest-specific snai2 promoter/enhancer, which is a powerful tool for studying neural crest development and craniofacial morphogenesis. Here, we carried out whole-genome shotgun sequencing for the snai2:eGFP embryos to identify the transgene integration sites. With a 19x sequencing coverage, we estimated that 6 copies of the transgene were inserted into the Xenopus tropicalis genome in the hemizygous transgenic embryos. Two transgene integration loci adjacent to each other were identified in a noncoding region on chromosome 1, possibly as a result of duplication after a single transgene insertion. Interestingly, genomic DNA at the boundaries of the transgene integration loci contains short sequences homologous to the I-SceI recognition site, suggesting that the integration was not random but probably mediated by sequence homology. To our knowledge, our work represents the first genome-wide sequencing study on a transgenic organism generated with I-SceI, which is useful for evaluating the potential genetic effects of I-SceI-mediated transgenesis and further understanding the mechanisms underlying this transgenic method.
Collapse
Affiliation(s)
- Jian Wang
- Department of Biological Sciences and Center for Bioinformatics and Computational Biology, University of Delaware, Newark, DE 19716, USA
- Department of Computational Biology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Congyu Lu
- Department of Biological Sciences and Center for Bioinformatics and Computational Biology, University of Delaware, Newark, DE 19716, USA
| | - Shuo Wei
- Department of Biological Sciences and Center for Bioinformatics and Computational Biology, University of Delaware, Newark, DE 19716, USA
| |
Collapse
|
14
|
Smirnov A, Battulin N. Concatenation of Transgenic DNA: Random or Orchestrated? Genes (Basel) 2021; 12:genes12121969. [PMID: 34946918 PMCID: PMC8701086 DOI: 10.3390/genes12121969] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 12/08/2021] [Accepted: 12/09/2021] [Indexed: 12/18/2022] Open
Abstract
Generation of transgenic organisms by pronuclear microinjection has become a routine procedure. However, while the process of DNA integration in the genome is well understood, we still do not know much about the recombination between transgene molecules that happens in the first moments after DNA injection. Most of the time, injected molecules are joined together in head-to-tail tandem repeats-the so-called concatemers. In this review, we focused on the possible concatenation mechanisms and how they could be studied with genetic reporters tracking individual copies in concatemers. We also discuss various features of concatemers, including palindromic junctions and repeat-induced gene silencing (RIGS). Finally, we speculate how cooperation of DNA repair pathways creates a multicopy concatenated insert.
Collapse
Affiliation(s)
- Alexander Smirnov
- Laboratory of Developmental Genetics, Institute of Cytology and Genetics SB RAS, 630090 Novosibirsk, Russia;
| | - Nariman Battulin
- Laboratory of Developmental Genetics, Institute of Cytology and Genetics SB RAS, 630090 Novosibirsk, Russia;
- Institute of Genetic Technologies, Novosibirsk State University, 630090 Novosibirsk, Russia
- Correspondence:
| |
Collapse
|
15
|
Salnikov PA, Khabarova AA, Koksharova GS, Mungalov RV, Belokopytova PS, Pristyazhnuk IE, Nurislamov AR, Somatich P, Gridina MM, Fishman VS. Here and there: the double-side transgene localization. Vavilovskii Zhurnal Genet Selektsii 2021; 25:607-612. [PMID: 34755021 PMCID: PMC8553977 DOI: 10.18699/vj21.068] [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] [Received: 06/02/2021] [Revised: 06/22/2021] [Accepted: 07/01/2021] [Indexed: 11/19/2022] Open
Abstract
Random transgene integration is a powerful tool for developing new genome-wide screening approaches. These techniques have already been used for functional gene annotation by transposon-insertion sequencing, for identif ication of transcription factor binding sites and regulatory sequences, and for dissecting chromatin position effects. Precise localization of transgenes and accurate artifact f iltration are essential for this type of method. To date, many mapping assays have been developed, including Inverse-PCR, TLA, LAM-PCR, and splinkerette PCR. However, none of them is able to ensure localization of both transgene’s f lanking regions simultaneously, which would be necessary for some applications. Here we proposed a cheap and simple NGS-based approach that overcomes this limitation. The developed assay requires using intentionally designed vectors that lack recognition sites of one or a set of restriction enzymes used for DNA fragmentation. By looping and sequencing these DNA fragments, we obtain special data that allows us to link the two f lanking regions of the transposon. This can be useful for precise insertion mapping and for screening approaches in the f ield of chromosome engineering, where chromosomal recombination events between transgenes occur in a cell population. To demonstrate the method’s feasibility, we applied it for mapping SB transposon integration in the human HAP1 cell line. Our technique allowed us to eff iciently localize genomic transposon integrations, which was conf irmed via PCR analysis. For practical application of this approach, we proposed a set of recommendations and a normalization strategy. The developed method can be used for multiplex transgene localization and detection of rearrangements between them.
Collapse
Affiliation(s)
- P A Salnikov
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - A A Khabarova
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - G S Koksharova
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - R V Mungalov
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - P S Belokopytova
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - I E Pristyazhnuk
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - A R Nurislamov
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - P Somatich
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - M M Gridina
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - V S Fishman
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| |
Collapse
|
16
|
Zhang P, Ganesamoorthy D, Nguyen SH, Au R, Coin LJ, Tey SK. Nanopore sequencing as a scalable, cost-effective platform for analyzing polyclonal vector integration sites following clinical T cell therapy. J Immunother Cancer 2021; 8:jitc-2019-000299. [PMID: 32527930 PMCID: PMC7292043 DOI: 10.1136/jitc-2019-000299] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/28/2020] [Indexed: 11/21/2022] Open
Abstract
Background Analysis of vector integration sites in gene-modified cells can provide critical information on clonality and potential biological impact on nearby genes. Current short-read next-generation sequencing methods require specialized instruments and large batch runs. Methods We used nanopore sequencing to analyze the vector integration sites of T cells transduced by the gammaretroviral vector, SFG.iCasp9.2A.ΔCD19. DNA from oligoclonal cell lines and polyclonal clinical samples were restriction enzyme digested with two 6-cutters, NcoI and BspHI; and the flanking genomic DNA amplified by inverse PCR or cassette ligation PCR. Following nested PCR and barcoding, the amplicons were sequenced on the Oxford Nanopore platform. Reads were filtered for quality, trimmed, and aligned. Custom tool was developed to cluster reads and merge overlapping clusters. Results Both inverse PCR and cassette ligation PCR could successfully amplify flanking genomic DNA, with cassette ligation PCR showing less bias. The 4.8 million raw reads were grouped into 12,186 clusters and 6410 clones. The 3′long terminal repeat (LTR)-genome junction could be resolved within a 5-nucleotide span for a majority of clusters and within one nucleotide span for clusters with ≥5 reads. The chromosomal distributions of the insertional sites and their predilection for regions proximate to transcription start sites were consistent with previous reports for gammaretroviral vector integrants as analyzed by short-read next-generation sequencing. Conclusion Our study shows that it is feasible to use nanopore sequencing to map polyclonal vector integration sites. The assay is scalable and requires minimum capital, which together enable cost-effective and timely analysis. Further refinement is required to reduce amplification bias and improve single nucleotide resolution.
Collapse
Affiliation(s)
- Ping Zhang
- Department of Immunology, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Devika Ganesamoorthy
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia
| | - Son Hoang Nguyen
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia
| | - Raymond Au
- Department of Immunology, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Lachlan J Coin
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia.,Department of Clinical Pathology, The University of Melbourne, Melbourne, Victoria, Australia.,Department of Infectious Disease, Imperial College London, London, London, UK
| | - Siok-Keen Tey
- Department of Immunology, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia .,Department of Haematology and Bone Marrow Transplantation, Royal Brisbane and Women's Hospital, Herston, Queensland, Australia.,Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
17
|
Patocchi A, Keilwagen J, Berner T, Wenzel S, Broggini GAL, Altschmied L, Hanke MV, Flachowsky H. No Evidence of Unexpected Transgenic Insertions in T1190 - A Transgenic Apple Used in Rapid Cycle Breeding - Following Whole Genome Sequencing. FRONTIERS IN PLANT SCIENCE 2021; 12:715737. [PMID: 34456955 PMCID: PMC8386123 DOI: 10.3389/fpls.2021.715737] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 07/15/2021] [Indexed: 05/24/2023]
Abstract
Rapid cycle breeding uses transgenic early flowering plants as crossbreed parents to facilitate the shortening of breeding programs for perennial crops with long-lasting juvenility. Rapid cycle breeding in apple was established using the transgenic genotype T1190 expressing the BpMADS4 gene of silver birch. In this study, the genomes of T1190 and its non-transgenic wild-type PinS (F1-offspring of 'Pinova' and 'Idared') were sequenced by Illumina short-read sequencing in two separate experiments resulting in a mean sequencing depth of 182× for T1190 and 167× for PinS. The sequencing revealed 8,450 reads, which contain sequences of ≥20 bp identical to the plant transformation vector. These reads were assembled into 125 contigs, which were examined to see whether they contained transgenic insertions or if they are not using a five-step procedure. The sequence of one contig represents the known T-DNA insertion on chromosome 4 of T1190. The sequences of the remaining contigs were either equally present in T1190 and PinS, their part with sequence identity to the vector was equally present in apple reference genomes, or they seem to result from endophytic contaminations rather than from additional transgenic insertions. Therefore, we conclude that the transgenic apple plant T1190 contains only one transgenic insertion, located on chromosome 4, and shows no further partial insertions of the transformation vector. Accession Numbers: JQ974028.1.
Collapse
Affiliation(s)
- Andrea Patocchi
- Research Division Plant Breeding, Agroscope, Wädenswil, Switzerland
| | - Jens Keilwagen
- Julius Kühn-Institut (JKI), Federal Research Centre for Cultivated Plants, Institute for Biosafety in Plant Biotechnology, Quedlinburg, Germany
| | - Thomas Berner
- Julius Kühn-Institut (JKI), Federal Research Centre for Cultivated Plants, Institute for Biosafety in Plant Biotechnology, Quedlinburg, Germany
| | - Stefanie Wenzel
- Julius Kühn-Institut (JKI), Federal Research Centre for Cultivated Plants, Institute for Breeding Research on Fruit Crops, Dresden, Germany
| | - Giovanni A. L. Broggini
- Swiss Federal Institute of Technology, Molecular Plant Breeding, Institute of Agricultural Sciences, ETH Zurich, Zurich, Switzerland
| | - Lothar Altschmied
- Leibniz-Institut für Pflanzengenetik und Kulturpflanzenforschung (IPK), Gatersleben, Germany
| | - Magda-Viola Hanke
- Julius Kühn-Institut (JKI), Federal Research Centre for Cultivated Plants, Institute for Breeding Research on Fruit Crops, Dresden, Germany
| | - Henryk Flachowsky
- Julius Kühn-Institut (JKI), Federal Research Centre for Cultivated Plants, Institute for Breeding Research on Fruit Crops, Dresden, Germany
| |
Collapse
|
18
|
Willemin A, Lopez-Delisle L, Bolt CC, Gadolini ML, Duboule D, Rodriguez-Carballo E. Induction of a chromatin boundary in vivo upon insertion of a TAD border. PLoS Genet 2021; 17:e1009691. [PMID: 34292939 PMCID: PMC8330945 DOI: 10.1371/journal.pgen.1009691] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 08/03/2021] [Accepted: 06/30/2021] [Indexed: 12/19/2022] Open
Abstract
Mammalian genomes are partitioned into sub-megabase to megabase-sized units of preferential interactions called topologically associating domains or TADs, which are likely important for the proper implementation of gene regulatory processes. These domains provide structural scaffolds for distant cis regulatory elements to interact with their target genes within the three-dimensional nuclear space and architectural proteins such as CTCF as well as the cohesin complex participate in the formation of the boundaries between them. However, the importance of the genomic context in providing a given DNA sequence the capacity to act as a boundary element remains to be fully investigated. To address this question, we randomly relocated a topological boundary functionally associated with the mouse HoxD gene cluster and show that it can indeed act similarly outside its initial genomic context. In particular, the relocated DNA segment recruited the required architectural proteins and induced a significant depletion of contacts between genomic regions located across the integration site. The host chromatin landscape was re-organized, with the splitting of the TAD wherein the boundary had integrated. These results provide evidence that topological boundaries can function independently of their site of origin, under physiological conditions during mouse development. During development, enhancer sequences tightly regulate the spatio-temporal expression of target genes often located hundreds of kilobases away. This complex process is made possible by the folding of chromatin into domains, which are separated from one another by specific genomic regions referred to as boundaries. In order to understand whether such boundary sequences require their particular genomic contexts to achieve their isolating effect, we analyzed the impact of introducing one such boundary, taken from the HoxD locus, into a distinct topological domain. We show that this ectopic boundary splits the host domain into two sub-domains and affects the expression levels of a neighboring gene. We conclude that this sequence can work independently from its genomic context and thus carries all the information necessary to act as a boundary element.
Collapse
Affiliation(s)
- Andréa Willemin
- Department of Genetics and Evolution, Faculty of Science, University of Geneva, Geneva, Switzerland
| | - Lucille Lopez-Delisle
- School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Christopher Chase Bolt
- School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Marie-Laure Gadolini
- Department of Genetics and Evolution, Faculty of Science, University of Geneva, Geneva, Switzerland
| | - Denis Duboule
- Department of Genetics and Evolution, Faculty of Science, University of Geneva, Geneva, Switzerland
- School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
- Collège de France, Paris, France
- * E-mail: (DD); (ER-C)
| | - Eddie Rodriguez-Carballo
- Department of Genetics and Evolution, Faculty of Science, University of Geneva, Geneva, Switzerland
- * E-mail: (DD); (ER-C)
| |
Collapse
|
19
|
Sailer S, Coassin S, Lackner K, Fischer C, McNeill E, Streiter G, Kremser C, Maglione M, Green CM, Moralli D, Moschen AR, Keller MA, Golderer G, Werner-Felmayer G, Tegeder I, Channon KM, Davies B, Werner ER, Watschinger K. When the genome bluffs: a tandem duplication event during generation of a novel Agmo knockout mouse model fools routine genotyping. Cell Biosci 2021; 11:54. [PMID: 33726865 PMCID: PMC7962373 DOI: 10.1186/s13578-021-00566-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 02/25/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Genome editing in mice using either classical approaches like homologous recombination or CRISPR/Cas9 has been reported to harbor off target effects (insertion/deletion, frame shifts or gene segment duplications) that lead to mutations not only in close proximity to the target site but also outside. Only the genomes of few engineered mouse strains have been sequenced. Since the role of the ether-lipid cleaving enzyme alkylglycerol monooxygenase (AGMO) in physiology and pathophysiology remains enigmatic, we created a knockout mouse model for AGMO using EUCOMM stem cells but unforeseen genotyping issues that did not agree with Mendelian distribution and enzyme activity data prompted an in-depth genomic validation of the mouse model. RESULTS We report a gene segment tandem duplication event that occurred during the generation of an Agmo knockout-first allele by homologous recombination. Only low homology was seen between the breakpoints. While a single copy of the recombinant 18 kb cassette was integrated correctly around exon 2 of the Agmo gene, whole genome nanopore sequencing revealed a 94 kb duplication in the Agmo locus that contains Agmo wild-type exons 1-3. The duplication fooled genotyping by routine PCR, but could be resolved using qPCR-based genotyping, targeted locus amplification sequencing and nanopore sequencing. Despite this event, this Agmo knockout mouse model lacks AGMO enzyme activity and can therefore be used to study its physiological role. CONCLUSIONS A duplication event occurred at the exact locus of the homologous recombination and was not detected by conventional quality control filters such as FISH or long-range PCR over the recombination sites. Nanopore sequencing provides a cost convenient method to detect such underrated off-target effects, suggesting its use for additional quality assessment of gene editing in mice and also other model organisms.
Collapse
Affiliation(s)
- Sabrina Sailer
- Institute of Biological Chemistry, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Stefan Coassin
- Institute of Genetic Epidemiology, Department of Genetics and Pharmacology, Medical University of Innsbruck, Innsbruck, Austria
| | - Katharina Lackner
- Institute of Biological Chemistry, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Caroline Fischer
- Institute of Clinical Pharmacology of the Medical Faculty, Goethe-University, Frankfurt (Main), Germany
| | - Eileen McNeill
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
- Division of Cardiovascular Medicine, British Heart Foundation Centre of Research Excellence, University of Oxford, Oxford, United Kingdom
| | - Gertraud Streiter
- Institute of Genetic Epidemiology, Department of Genetics and Pharmacology, Medical University of Innsbruck, Innsbruck, Austria
| | - Christian Kremser
- Department of Radiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Manuel Maglione
- Department of Visceral, Transplant and Thoracic Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Catherine M Green
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Daniela Moralli
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Alexander R Moschen
- Department of Internal Medicine I, Gastroenterology, Endocrinology and Metabolism, Medical University of Innsbruck, Innsbruck, Austria
| | - Markus A Keller
- Institute of Human Genetics, Medical University of Innsbruck, Innsbruck, Austria
| | - Georg Golderer
- Institute of Biological Chemistry, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Gabriele Werner-Felmayer
- Institute of Biological Chemistry, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Irmgard Tegeder
- Institute of Clinical Pharmacology of the Medical Faculty, Goethe-University, Frankfurt (Main), Germany
| | - Keith M Channon
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
- Division of Cardiovascular Medicine, British Heart Foundation Centre of Research Excellence, University of Oxford, Oxford, United Kingdom
| | - Benjamin Davies
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Ernst R Werner
- Institute of Biological Chemistry, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Katrin Watschinger
- Institute of Biological Chemistry, Biocenter, Medical University of Innsbruck, Innsbruck, Austria.
- Institute of Biological Chemistry, Biocenter, Medical University of Innsbruck, Innrain 80, 6020, Innsbruck, Austria.
| |
Collapse
|
20
|
Giraldo PA, Shinozuka H, Spangenberg GC, Smith KF, Cogan NOI. Rapid and Detailed Characterization of Transgene Insertion Sites in Genetically Modified Plants via Nanopore Sequencing. FRONTIERS IN PLANT SCIENCE 2021; 11:602313. [PMID: 33613582 PMCID: PMC7889508 DOI: 10.3389/fpls.2020.602313] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 12/24/2020] [Indexed: 05/15/2023]
Abstract
Molecular characterization of genetically modified plants can provide crucial information for the development of detection and identification methods, to comply with traceability, and labeling requirements prior to commercialization. Detailed description of the genetic modification was previously a challenging step in the safety assessment, since it required the use of laborious and time-consuming techniques. In this study an accurate, simple, and fast method was developed for molecular characterization of genetically modified (GM) plants, following a user-friendly workflow for researchers with limited bioinformatic capabilities. Three GM events from a diverse array of crop species-perennial ryegrass, white clover, and canola-were used to test the approach that exploits long-read sequencing by the MinION device, from Oxford Nanopore Technologies. The method delivered a higher degree of resolution of the transgenic events within the host genome than has previously been possible with the standard Illumina short-range sequencing strategies. The flanking sequences, copy number, and presence of backbone sequences, and overall transgene insertion structure were determined for each of the plant genomes, with the additional identification of moderate-sized secondary insertions that would have previously been missed. The proposed workflow takes only about 1 week from DNA extraction to analyzed result, and the method will complement the existing approaches for molecular characterization of GM plants, since it makes the process faster, simpler, and more cost-effective.
Collapse
Affiliation(s)
- Paula A. Giraldo
- Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Parkville, VIC, Australia
- Agriculture Victoria Research, AgriBio, The Centre for AgriBioscience, Bundoora, VIC, Australia
| | - Hiroshi Shinozuka
- Agriculture Victoria Research, AgriBio, The Centre for AgriBioscience, Bundoora, VIC, Australia
| | - German C. Spangenberg
- Agriculture Victoria Research, AgriBio, The Centre for AgriBioscience, Bundoora, VIC, Australia
- AgriBio, The Centre for AgriBioscience, School of Applied Systems Biology, La Trobe University, Bundoora, VIC, Australia
| | - Kevin F. Smith
- Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Parkville, VIC, Australia
- Agriculture Victoria Research, Hamilton, VIC, Australia
| | - Noel O. I. Cogan
- Agriculture Victoria Research, AgriBio, The Centre for AgriBioscience, Bundoora, VIC, Australia
- AgriBio, The Centre for AgriBioscience, School of Applied Systems Biology, La Trobe University, Bundoora, VIC, Australia
| |
Collapse
|
21
|
Mikedis MM, Fan Y, Nicholls PK, Endo T, Jackson EK, Cobb SA, de Rooij DG, Page DC. DAZL mediates a broad translational program regulating expansion and differentiation of spermatogonial progenitors. eLife 2020; 9:56523. [PMID: 32686646 PMCID: PMC7445011 DOI: 10.7554/elife.56523] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 07/20/2020] [Indexed: 01/28/2023] Open
Abstract
Fertility across metazoa requires the germline-specific DAZ family of RNA-binding proteins. Here we examine whether DAZL directly regulates progenitor spermatogonia using a conditional genetic mouse model and in vivo biochemical approaches combined with chemical synchronization of spermatogenesis. We find that the absence of Dazl impairs both expansion and differentiation of the spermatogonial progenitor population. In undifferentiated spermatogonia, DAZL binds the 3' UTRs of ~2,500 protein-coding genes. Some targets are known regulators of spermatogonial proliferation and differentiation while others are broadly expressed, dosage-sensitive factors that control transcription and RNA metabolism. DAZL binds 3' UTR sites conserved across vertebrates at a UGUU(U/A) motif. By assessing ribosome occupancy in undifferentiated spermatogonia, we find that DAZL increases translation of its targets. In total, DAZL orchestrates a broad translational program that amplifies protein levels of key spermatogonial and gene regulatory factors to promote the expansion and differentiation of progenitor spermatogonia.
Collapse
Affiliation(s)
| | - Yuting Fan
- Whitehead Institute, Cambridge, United States.,Reproductive Medicine Center, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | | | | | - Emily K Jackson
- Whitehead Institute, Cambridge, United States.,Department of Biology, Massachusetts Institute of Technology, Cambridge, United States
| | | | | | - David C Page
- Whitehead Institute, Cambridge, United States.,Department of Biology, Massachusetts Institute of Technology, Cambridge, United States.,Howard Hughes Medical Institute, Whitehead Institute, Cambridge, United States
| |
Collapse
|
22
|
George J, Kahlke T, Abbriano RM, Kuzhiumparambil U, Ralph PJ, Fabris M. Metabolic Engineering Strategies in Diatoms Reveal Unique Phenotypes and Genetic Configurations With Implications for Algal Genetics and Synthetic Biology. Front Bioeng Biotechnol 2020; 8:513. [PMID: 32582656 PMCID: PMC7290003 DOI: 10.3389/fbioe.2020.00513] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 04/30/2020] [Indexed: 12/23/2022] Open
Abstract
Diatoms are photosynthetic microeukaryotes that dominate phytoplankton populations and have increasing applicability in biotechnology. Uncovering their complex biology and elevating strains to commercial standards depends heavily on robust genetic engineering tools. However, engineering microalgal genomes predominantly relies on random integration of transgenes into nuclear DNA, often resulting in detrimental “position-effects” such as transgene silencing, integration into transcriptionally-inactive regions, and endogenous sequence disruption. With the recent development of extrachromosomal transgene expression via independent episomes, it is timely to investigate both strategies at the phenotypic and genomic level. Here, we engineered the model diatom Phaeodactylum tricornutum to produce the high-value heterologous monoterpenoid geraniol, which, besides applications as fragrance and insect repellent, is a key intermediate of high-value pharmaceuticals. Using high-throughput phenotyping we confirmed the suitability of episomes for synthetic biology applications and identified superior geraniol-yielding strains following random integration. We used third generation long-read sequencing technology to generate a complete analysis of all transgene integration events including their genomic locations and arrangements associated with high-performing strains at a genome-wide scale with subchromosomal detail, never before reported in any microalga. This revealed very large, highly concatenated insertion islands, offering profound implications on diatom functional genetics and next generation genome editing technologies, and is key for developing more precise genome engineering approaches in diatoms, including possible genomic safe harbour locations to support high transgene expression for targeted integration approaches. Furthermore, we have demonstrated that exogenous DNA is not integrated inadvertently into the nuclear genome of extrachromosomal-expression clones, an important characterisation of this novel engineering approach that paves the road to synthetic biology applications.
Collapse
Affiliation(s)
- Jestin George
- University of Technology Sydney, Climate Change Cluster, Faculty of Science, Ultimo, NSW, Australia
| | - Tim Kahlke
- University of Technology Sydney, Climate Change Cluster, Faculty of Science, Ultimo, NSW, Australia
| | - Raffaela M Abbriano
- University of Technology Sydney, Climate Change Cluster, Faculty of Science, Ultimo, NSW, Australia
| | | | - Peter J Ralph
- University of Technology Sydney, Climate Change Cluster, Faculty of Science, Ultimo, NSW, Australia
| | - Michele Fabris
- University of Technology Sydney, Climate Change Cluster, Faculty of Science, Ultimo, NSW, Australia.,CSIRO Synthetic Biology Future Science Platform, Brisbane, QLD, Australia
| |
Collapse
|
23
|
Smirnov A, Fishman V, Yunusova A, Korablev A, Serova I, Skryabin BV, Rozhdestvensky TS, Battulin N. DNA barcoding reveals that injected transgenes are predominantly processed by homologous recombination in mouse zygote. Nucleic Acids Res 2020; 48:719-735. [PMID: 31740957 PMCID: PMC7145541 DOI: 10.1093/nar/gkz1085] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 10/29/2019] [Accepted: 11/05/2019] [Indexed: 12/14/2022] Open
Abstract
Mechanisms that ensure repair of double-strand DNA breaks (DSBs) are instrumental in the integration of foreign DNA into the genome of transgenic organisms. After pronuclear microinjection, exogenous DNA is usually found as a concatemer comprising multiple co-integrated transgene copies. Here, we investigated the contribution of various DSB repair pathways to the concatemer formation. We injected mouse zygotes with a pool of linear DNA molecules carrying unique barcodes at both ends and obtained 10 transgenic embryos with 1–300 transgene copies. Sequencing the barcodes allowed us to assign relative positions to the copies in concatemers and detect recombination events that occurred during integration. Cumulative analysis of approximately 1,000 integrated copies reveals that over 80% of them underwent recombination when their linear ends were processed by synthesis-dependent strand annealing (SDSA) or double-strand break repair (DSBR). We also observed evidence of double Holliday junction (dHJ) formation and crossing over during the concatemer formations. Sequencing indels at the junctions between copies shows that at least 10% of DNA molecules introduced into the zygotes are ligated by non-homologous end joining (NHEJ). Our barcoding approach, verified with Pacific Biosciences Single Molecule Real-Time (SMRT) long-range sequencing, documents high activity of homologous recombination after DNA microinjection.
Collapse
Affiliation(s)
| | - Veniamin Fishman
- Institute of Cytology and Genetics SB RAS, Novosibirsk, Russia.,Novosibirsk State University, Novosibirsk, Russia
| | | | - Alexey Korablev
- Institute of Cytology and Genetics SB RAS, Novosibirsk, Russia
| | - Irina Serova
- Institute of Cytology and Genetics SB RAS, Novosibirsk, Russia
| | - Boris V Skryabin
- Medical Faculty, Core Facility Transgenic animal and genetic engineering Models (TRAM), University of Muenster, Muenster, Germany
| | - Timofey S Rozhdestvensky
- Medical Faculty, Core Facility Transgenic animal and genetic engineering Models (TRAM), University of Muenster, Muenster, Germany
| | - Nariman Battulin
- Institute of Cytology and Genetics SB RAS, Novosibirsk, Russia.,Novosibirsk State University, Novosibirsk, Russia
| |
Collapse
|
24
|
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: 37] [Impact Index Per Article: 7.4] [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
|
25
|
Suzuki O, Koura M, Uchio-Yamada K, Sasaki M. Analysis of the transgene insertion pattern in a transgenic mouse strain using long-read sequencing. Exp Anim 2020; 69:279-286. [PMID: 32051389 PMCID: PMC7445054 DOI: 10.1538/expanim.19-0118] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Transgene insertion patterns are critical for the analysis of transgenic animals because
the influence of transgenes may change depending on the insertion pattern (such as copy
numbers and orientations of concatenations) and the insertion position in the genome. We
previously reported a genomic walking strategy to locate transgenes in the genomes of
transgenic mice (Exp. Anim. 53: 103–111, 2004) and to analyze transgene insertion patterns
(Exp. Anim. 55: 65–69, 2006). With such strategies, however, we could not determine the
copy number of transgenes or global genome modification induced by transgene insertion due
to read-length limitation. In this study, we used a long-read sequencer (MinION, Oxford
Nanopore Technologies) to overcome this limitation. We obtained 922,210 reads using MinION
with genomic DNA from a transgenic mouse strain (4C30, Proc. Jpn. Acad. Ser. B. Phys.
Biol. Sci. 87: 550–562, 2011). Among the reads, we found one 21,457-bp read containing the
transgene using a local BLAST search. Nucleotide dot plot analysis revealed that the
transgene was inserted in the genome as a tandem concatemer with an almost entire
construct (15–3,508 of 3,508 bp) and a partial fragment (4–660, 657 bp). Ensembl’s BLAST
search against the C57BL/6N genome revealed a 9,388-bp deletion at the insertion position
in the intron of the Sgcd gene, confirming that mutations such as a large
genomic deletion could occur at the time of transgene insertion. Thus, long-read
sequencers are useful tools for the analysis of transgene insertion patterns.
Collapse
Affiliation(s)
- Osamu Suzuki
- Laboratory of Animal Models for Human Diseases, National Institutes of Biomedical Innovation, Health and Nutrition,7-6-8 Saito-Asagi, Ibaraki, Ibaraki, Osaka 568-0085, Japan
| | - Minako Koura
- Laboratory of Animal Models for Human Diseases, National Institutes of Biomedical Innovation, Health and Nutrition,7-6-8 Saito-Asagi, Ibaraki, Ibaraki, Osaka 568-0085, Japan
| | - Kozue Uchio-Yamada
- Laboratory of Animal Models for Human Diseases, National Institutes of Biomedical Innovation, Health and Nutrition,7-6-8 Saito-Asagi, Ibaraki, Ibaraki, Osaka 568-0085, Japan
| | - Mitsuho Sasaki
- Laboratory of Animal Models for Human Diseases, National Institutes of Biomedical Innovation, Health and Nutrition,7-6-8 Saito-Asagi, Ibaraki, Ibaraki, Osaka 568-0085, Japan
| |
Collapse
|