1
|
Kwon DH, Gim GM, Yum SY, Eom KH, Lee SJ, Han SE, Kim HS, Kim HJ, Lee WS, Choi WJ, Lee JH, Kim DY, Jung DJ, Kim DH, Yi JK, Moon BH, Lee WY, Jang G. Cas9-expressing cattle using the PiggyBac transposon all-in-one system. BMC Genomics 2025; 26:217. [PMID: 40045211 PMCID: PMC11881473 DOI: 10.1186/s12864-025-11381-8] [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: 10/15/2024] [Accepted: 02/19/2025] [Indexed: 03/09/2025] Open
Abstract
BACKGROUND Livestock, particularly cattle, are crucial for biotechnology fields, such as genetic breeding, infectious diseases, bioreactors, and specific disease models. However, genetic engineering in cattle has lagged due to long gestation periods, single embryo pregnancies, and high rearing costs. Additionally, the slow validation of germline transmission and the absence of germline-competent embryonic stem cells hinder progress. With the development of genome editing technologies like ZFN, TALEN, and CRISPR-Cas9, recent advancements have shown that Cas9-expressing pigs and chickens have been successfully produced. We hypothesize that generating CRISPR/Cas9-expressing cattle and their resources will provide a powerful resource for bovine genome editing, advancing our understanding of bovine genetics and disease resistance. RESULTS In this study, two types of Cas9-expressing cattle were successfully produced: Cas9-RFP-fatty acid dehydrogenase I (FatI), Cas9-GFP-sgRNA for the prion protein (sgPRNP). Somatic cells from these cattle were induced to mutate multiple target genes when single-guide RNAs (sgRNAs) were transfected into the somatic cells. Additionally, semen from Cas9 expressing male cattle was frozen and used to fertilize wild-type oocytes, successfully transmitting the transgene (Cas9, reporter genes, FatI), and sgPRNP) to the next generation. Furthermore, the gene editing capabilities of Cas9, including knockout and high-efficiency knock-in, were confirmed in embryos derived from F1 semen through in vitro production. CONCLUSION These data demonstrate, for the first time, that Cas9-expressing cattle were successfully born, and this transgene was transmitted to the next-generation calves (F1) and F2 embryos. In addition, somatic and germ cells derived from F0 and F1generations were used to evaluate the potential for gene editing (knockout and knock-in) in multiple genes. PRNP-mutated F1 cattle are currently being raised as a resistance model for bovine spongiform encephalopathy. These transgenic bovine models and their derivatives will serve as a valuable resource for both in vitro and in vivo genome editing, advancing our genetic understanding of bovine genomics and diseases.
Collapse
Affiliation(s)
- Dong-Hyeok Kwon
- Department of Theriogenology, College of Veterinary Medicine and the Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, Republic of Korea
- Bk21 FOUR Future Veterinary Medicine Leading Education & Research Center, Seoul National University, Seoul, 08826, Republic of Korea
| | - Gyeong-Min Gim
- Department of Theriogenology, College of Veterinary Medicine and the Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, Republic of Korea
- LARTBio Inc, Seoul, 14322, Republic of Korea
| | - Soo-Young Yum
- Department of Theriogenology, College of Veterinary Medicine and the Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, Republic of Korea
- LARTBio Inc, Seoul, 14322, Republic of Korea
| | - Kyeong-Hyeon Eom
- Department of Theriogenology, College of Veterinary Medicine and the Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, Republic of Korea
- LARTBio Inc, Seoul, 14322, Republic of Korea
| | - Song-Jeon Lee
- Embryo Research Center, Seoul Milk Coop, Gyeonggi-Do, 12528, Republic of Korea
| | - Sang-Eun Han
- Department of Theriogenology, College of Veterinary Medicine and the Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, Republic of Korea
| | - Hee-Soo Kim
- Embryo Research Center, Seoul Milk Coop, Gyeonggi-Do, 12528, Republic of Korea
| | - Hyeong-Jong Kim
- Embryo Research Center, Seoul Milk Coop, Gyeonggi-Do, 12528, Republic of Korea
| | - Woo-Sung Lee
- Embryo Research Center, Seoul Milk Coop, Gyeonggi-Do, 12528, Republic of Korea
| | - Woo-Jae Choi
- Department of Theriogenology, College of Veterinary Medicine and the Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, Republic of Korea
| | - Ji-Hyun Lee
- LARTBio Inc, Seoul, 14322, Republic of Korea
| | - Do-Yoon Kim
- Gyeongsangbukdo Livestock Research Institute, GyeongSang Buk-Do, Yeongju, 36052, Republic of Korea
| | - Dae-Jin Jung
- Gyeongsangbukdo Livestock Research Institute, GyeongSang Buk-Do, Yeongju, 36052, Republic of Korea
| | - Dae-Hyun Kim
- Department of Animal Science, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Jun-Koo Yi
- School of Animal Life Convergence Science, Hankyong National University, Anseong, Gyeonggi-Do, 17579, Republic of Korea
| | - Byeong-Ho Moon
- Department of Theriogenology, College of Veterinary Medicine and the Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, Republic of Korea
| | - Won-You Lee
- LARTBio Inc, Seoul, 14322, Republic of Korea
| | - Goo Jang
- Department of Theriogenology, College of Veterinary Medicine and the Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, Republic of Korea.
- Bk21 FOUR Future Veterinary Medicine Leading Education & Research Center, Seoul National University, Seoul, 08826, Republic of Korea.
- LARTBio Inc, Seoul, 14322, Republic of Korea.
| |
Collapse
|
2
|
Gim GM, Jang G. Outlook on genome editing application to cattle. J Vet Sci 2024; 25:e10. [PMID: 38311323 PMCID: PMC10839183 DOI: 10.4142/jvs.23133] [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: 05/15/2023] [Revised: 08/04/2023] [Accepted: 08/20/2023] [Indexed: 02/07/2024] Open
Abstract
In livestock industry, there is growing interest in methods to increase the production efficiency of livestock to address food shortages, given the increasing global population. With the advancements in gene engineering technology, it is a valuable tool and has been intensively utilized in research specifically focused on human disease. In historically, this technology has been used with livestock to create human disease models or to produce recombinant proteins from their byproducts. However, in recent years, utilizing gene editing technology, cattle with identified genes related to productivity can be edited, thereby enhancing productivity in response to climate change or specific disease instead of producing recombinant proteins. Furthermore, with the advancement in the efficiency of gene editing, it has become possible to edit multiple genes simultaneously. This cattle breed improvement has been achieved by discovering the genes through the comprehensive analysis of the entire genome of cattle. The cattle industry has been able to address gene bottlenecks that were previously impossible through conventional breeding systems. This review concludes that gene editing is necessary to expand the cattle industry, improving productivity in the future. Additionally, the enhancement of cattle through gene editing is expected to contribute to addressing environmental challenges associated with the cattle industry. Further research and development in gene editing, coupled with genomic analysis technologies, will significantly contribute to solving issues that conventional breeding systems have not been able to address.
Collapse
Affiliation(s)
| | - Goo Jang
- LARTBio Inco, Seoul 06221, Korea
- Laboratory of Theriogenology and Biotechnology, Department of Veterinary Clinical Science, College of Veterinary Medicine and the Research Institute of Veterinary Science, Seoul National University, Seoul 08826, Korea
- Comparative medicine Disease Research Center, Seoul National University, Seoul 08826, Korea
- Faculty of Veterinary Medicine, Universitas Airlangga, Surabaya 60115, Indonesia.
| |
Collapse
|
3
|
Liu S, Hur YH, Cai X, Cong Q, Yang Y, Xu C, Bilate AM, Gonzales KAU, Parigi SM, Cowley CJ, Hurwitz B, Luo JD, Tseng T, Gur-Cohen S, Sribour M, Omelchenko T, Levorse J, Pasolli HA, Thompson CB, Mucida D, Fuchs E. A tissue injury sensing and repair pathway distinct from host pathogen defense. Cell 2023; 186:2127-2143.e22. [PMID: 37098344 PMCID: PMC10321318 DOI: 10.1016/j.cell.2023.03.031] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 02/03/2023] [Accepted: 03/27/2023] [Indexed: 04/27/2023]
Abstract
Pathogen infection and tissue injury are universal insults that disrupt homeostasis. Innate immunity senses microbial infections and induces cytokines/chemokines to activate resistance mechanisms. Here, we show that, in contrast to most pathogen-induced cytokines, interleukin-24 (IL-24) is predominately induced by barrier epithelial progenitors after tissue injury and is independent of microbiome or adaptive immunity. Moreover, Il24 ablation in mice impedes not only epidermal proliferation and re-epithelialization but also capillary and fibroblast regeneration within the dermal wound bed. Conversely, ectopic IL-24 induction in the homeostatic epidermis triggers global epithelial-mesenchymal tissue repair responses. Mechanistically, Il24 expression depends upon both epithelial IL24-receptor/STAT3 signaling and hypoxia-stabilized HIF1α, which converge following injury to trigger autocrine and paracrine signaling involving IL-24-mediated receptor signaling and metabolic regulation. Thus, parallel to innate immune sensing of pathogens to resolve infections, epithelial stem cells sense injury signals to orchestrate IL-24-mediated tissue repair.
Collapse
Affiliation(s)
- Siqi Liu
- Robin Chemers Neustein Laboratory of Mammalian Development and Cell Biology, Howard Hughes Medical Institute, The Rockefeller University, New York, NY 10065, USA
| | - Yun Ha Hur
- Robin Chemers Neustein Laboratory of Mammalian Development and Cell Biology, Howard Hughes Medical Institute, The Rockefeller University, New York, NY 10065, USA
| | - Xin Cai
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Qian Cong
- McDermott Center for Human Growth and Development, Department of Biophysics, and Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Yihao Yang
- Robin Chemers Neustein Laboratory of Mammalian Development and Cell Biology, Howard Hughes Medical Institute, The Rockefeller University, New York, NY 10065, USA
| | - Chiwei Xu
- Robin Chemers Neustein Laboratory of Mammalian Development and Cell Biology, Howard Hughes Medical Institute, The Rockefeller University, New York, NY 10065, USA
| | - Angelina M Bilate
- Laboratory of Mucosal Immunology, Howard Hughes Medical Institute, The Rockefeller University, New York, NY 10065, USA
| | - Kevin Andrew Uy Gonzales
- Robin Chemers Neustein Laboratory of Mammalian Development and Cell Biology, Howard Hughes Medical Institute, The Rockefeller University, New York, NY 10065, USA
| | - S Martina Parigi
- Robin Chemers Neustein Laboratory of Mammalian Development and Cell Biology, Howard Hughes Medical Institute, The Rockefeller University, New York, NY 10065, USA
| | - Christopher J Cowley
- Robin Chemers Neustein Laboratory of Mammalian Development and Cell Biology, Howard Hughes Medical Institute, The Rockefeller University, New York, NY 10065, USA
| | - Brian Hurwitz
- Robin Chemers Neustein Laboratory of Mammalian Development and Cell Biology, Howard Hughes Medical Institute, The Rockefeller University, New York, NY 10065, USA
| | - Ji-Dung Luo
- Bioinformatics Resource Center, The Rockefeller University, New York, NY 10065, USA
| | - Tiffany Tseng
- Robin Chemers Neustein Laboratory of Mammalian Development and Cell Biology, Howard Hughes Medical Institute, The Rockefeller University, New York, NY 10065, USA
| | - Shiri Gur-Cohen
- Robin Chemers Neustein Laboratory of Mammalian Development and Cell Biology, Howard Hughes Medical Institute, The Rockefeller University, New York, NY 10065, USA
| | - Megan Sribour
- Robin Chemers Neustein Laboratory of Mammalian Development and Cell Biology, Howard Hughes Medical Institute, The Rockefeller University, New York, NY 10065, USA
| | - Tatiana Omelchenko
- Robin Chemers Neustein Laboratory of Mammalian Development and Cell Biology, Howard Hughes Medical Institute, The Rockefeller University, New York, NY 10065, USA
| | - John Levorse
- Robin Chemers Neustein Laboratory of Mammalian Development and Cell Biology, Howard Hughes Medical Institute, The Rockefeller University, New York, NY 10065, USA
| | - Hilda Amalia Pasolli
- Electron Microscopy Resource Center, The Rockefeller University, New York, NY 10065, USA
| | - Craig B Thompson
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Daniel Mucida
- Laboratory of Mucosal Immunology, Howard Hughes Medical Institute, The Rockefeller University, New York, NY 10065, USA
| | - Elaine Fuchs
- Robin Chemers Neustein Laboratory of Mammalian Development and Cell Biology, Howard Hughes Medical Institute, The Rockefeller University, New York, NY 10065, USA.
| |
Collapse
|
4
|
Xie Y, Wang M, Gu L, Wang Y. CRISPR/Cas9-mediated knock-in strategy at the Rosa26 locus in cattle fetal fibroblasts. PLoS One 2022; 17:e0276811. [PMID: 36441701 PMCID: PMC9704577 DOI: 10.1371/journal.pone.0276811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Accepted: 10/14/2022] [Indexed: 11/29/2022] Open
Abstract
The genetic modification of cattle has many agricultural and biomedical applications. However, random integration often leads to the unstable or differentially expression of the exogenous genes, which limit the application and development of transgenic technologies. Finding a safe locus suitable for site-specific insertion and efficient expression of exogenous genes is a good way to overcome these hurdles. In this study, we efficiently integrated three targeted vector into the cattle Rosa26 (cRosa26) by CRISPR/Cas9 technology in which EGFP was driven by CAG, EF1a, PGK and cRosa26 endogenous promoter respectively. The CRISPR/Cas9 knock-in system allows highly efficient gene insertion of different expression units at the cRosa26 locus. We also find that in the four cell lines, EGFP was stable expressed at different times, and the CAG promoter has the highest activity to activate the expression of EGFP, when compared with the cRosa26, EF1a and PGK promoter. Our results proved that cRosa26 was a locus that could integrate different expression units efficiently, and supported the friendly expression of different expression units. Our findings described here will be useful for a variety of studies using cattle.
Collapse
Affiliation(s)
- Yuxuan Xie
- Key Laboratory of Songliao Aquatic Environment, Ministry of Education, Jilin jianzhu University, Changchun, Jilin Province, People’s Republic of China
| | - Ming Wang
- College of Animal Science and Technology, China Agricultural University, Beijing, People’s Republic of China
| | - Liang Gu
- Key Laboratory of Songliao Aquatic Environment, Ministry of Education, Jilin jianzhu University, Changchun, Jilin Province, People’s Republic of China
| | - Yang Wang
- Key Laboratory of Songliao Aquatic Environment, Ministry of Education, Jilin jianzhu University, Changchun, Jilin Province, People’s Republic of China
- * E-mail:
| |
Collapse
|
5
|
Kwon DH, Gim GM, Eom KH, Lee JH, Jang G. Application of transposon systems in the transgenesis of bovine somatic and germ cells. BMC Vet Res 2022; 18:156. [PMID: 35477562 PMCID: PMC9044889 DOI: 10.1186/s12917-022-03252-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Accepted: 04/14/2022] [Indexed: 12/12/2022] Open
Abstract
Background Several DNA transposons including PiggyBac (PB), Sleeping Beauty (SB), and Tol2 have been applied as effective means for of transgenesis in many species. Cattle are not typically experimental animals, and relatively little verification has been presented on this species. Thus, the goal here was to determine the applicability of three transposon systems in somatic and embryo cells in cattle, while also investigating which of the three systems is appropriate for each cell type. Green fluorescent protein (GFP)-expressing transposon systems were used for electroporation and microinjection in the somatic cells and embryo stage, respectively. After transfection, the GFP-positive cells or blastocysts were observed through fluorescence, while the transfection efficiency was calculated by FACS. Results In bovine somatic cells, the PB (63.97 ± 11.56) showed the highest efficiency of the three systems (SB: 50.74 ± 13.02 and Tol2: 16.55 ± 5.96). Conversely, Tol2 (75.00%) and SB (70.00%) presented a higher tendency in the embryonic cells compared to PB (42.86%). Conclusions These results demonstrate that these three transposon systems can be used in bovine somatic cells and embryos as gene engineering experimental methods. Moreover, they demonstrate which type of transposon system to apply depending on the cell type. Supplementary Information The online version contains supplementary material available at 10.1186/s12917-022-03252-1.
Collapse
Affiliation(s)
- Dong-Hyeok Kwon
- Laboratory of Theriogenology, College of Veterinary Medicine, Seoul National University, Gwanak-ro, Gwanak-gu, Seoul, Republic of Korea, 08826.,BK21 PLUS program, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
| | - Gyeong-Min Gim
- Laboratory of Theriogenology, College of Veterinary Medicine, Seoul National University, Gwanak-ro, Gwanak-gu, Seoul, Republic of Korea, 08826.,BK21 PLUS program, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
| | - Kyeong-Hyeon Eom
- Laboratory of Theriogenology, College of Veterinary Medicine, Seoul National University, Gwanak-ro, Gwanak-gu, Seoul, Republic of Korea, 08826.,BK21 PLUS program, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
| | | | - Goo Jang
- Laboratory of Theriogenology, College of Veterinary Medicine, Seoul National University, Gwanak-ro, Gwanak-gu, Seoul, Republic of Korea, 08826. .,BK21 PLUS program, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea. .,LARTBio Inc, Seoul, Republic of Korea.
| |
Collapse
|
6
|
Dehdilani N, Taemeh SY, Goshayeshi L, Dehghani H. Genetically engineered birds; pre-CRISPR and CRISPR era. Biol Reprod 2021; 106:24-46. [PMID: 34668968 DOI: 10.1093/biolre/ioab196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 10/08/2021] [Accepted: 10/14/2021] [Indexed: 11/14/2022] Open
Abstract
Generating biopharmaceuticals in genetically engineered bioreactors continues to reign supreme. Hence, genetically engineered birds have attracted considerable attention from the biopharmaceutical industry. Fairly recent genome engineering methods have made genome manipulation an easy and affordable task. In this review, we first provide a broad overview of the approaches and main impediments ahead of generating efficient and reliable genetically engineered birds, and various factors that affect the fate of a transgene. This section provides an essential background for the rest of the review, in which we discuss and compare different genome manipulation methods in the pre-CRISPR and CRISPR era in the field of avian genome engineering.
Collapse
Affiliation(s)
- Nima Dehdilani
- Stem Cell Biology and Regenerative Medicine Research Group, Research Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Sara Yousefi Taemeh
- Stem Cell Biology and Regenerative Medicine Research Group, Research Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Lena Goshayeshi
- Stem Cell Biology and Regenerative Medicine Research Group, Research Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Hesam Dehghani
- Stem Cell Biology and Regenerative Medicine Research Group, Research Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran.,Division of Biotechnology, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran.,Department of Basic Sciences, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran
| |
Collapse
|
7
|
Altgilbers S, Klein S, Dierks C, Weigend S, Kues WA. Cultivation and characterization of primordial germ cells from blue layer hybrids (Araucana crossbreeds) and generation of germline chimeric chickens. Sci Rep 2021; 11:12923. [PMID: 34155221 PMCID: PMC8217269 DOI: 10.1038/s41598-021-91490-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 05/21/2021] [Indexed: 11/29/2022] Open
Abstract
The chicken (Gallus gallus) is one of the most common and widespread domestic species, with an estimated total population of 25 billion birds worldwide. The vast majority of chickens in agriculture originate from hybrid breeding programs and is concentrated on few commercially used high performance lines, whereas numerous local and indigenous breeds are at risk to become extinct. To preserve the genomic resources of rare and endangered chicken breeds innovative methods are necessary. Here, we established a solid workflow for the derivation and biobanking of chicken primordial germ cells (PGCs) from blue layer hybrids. To achieve this, embryos of a cross of heterozygous blue egg layers were sampled to obtain blood derived and gonadal male as well as female PGCs of different genotypes (homozygous, heterozygous and nullizygous blue-allele bearing). The total efficiency of established PGC lines was 45% (47/104) within an average of 49 days until they reached sufficient numbers of cells for cryopreservation. The stem-cell character of the cultivated PGCs was confirmed by SSEA-1 immunostaining, and RT-PCR amplification of the pluripotency- and PGC-specific genes cPOUV, cNANOG, cDAZL and CVH. The Sleeping Beauty transposon system allowed to generate a stable integration of a Venus fluorophore reporter into the chicken genome. Finally, we demonstrated that, after re-transfer into chicken embryos, Venus-positive PGCs migrated and colonized the forming gonads. Semen samples of 13 raised cell chimeric roosters were analyzed by flow cytometry for the efficiency of germline colonization by the transferred PGCs carrying the Venus reporter and their proper differentiation into vital spermatids. Thus, we provide a proof-of-concept study for the potential use of PGCs for the cryobanking of rare breeds or rare alleles.
Collapse
Affiliation(s)
- Stefanie Altgilbers
- Department of Biotechnology, Institute of Farm Animal Genetics, Friedrich-Loeffler-Institut, 31535, Neustadt, Germany
| | - Sabine Klein
- Department of Biotechnology, Institute of Farm Animal Genetics, Friedrich-Loeffler-Institut, 31535, Neustadt, Germany
| | - Claudia Dierks
- Department of Genetic Ressources, Institute of Farm Animal Genetics, Friedrich-Loeffler-Institut, 31535, Neustadt, Germany
| | - Steffen Weigend
- Department of Genetic Ressources, Institute of Farm Animal Genetics, Friedrich-Loeffler-Institut, 31535, Neustadt, Germany
| | - Wilfried A Kues
- Department of Biotechnology, Institute of Farm Animal Genetics, Friedrich-Loeffler-Institut, 31535, Neustadt, Germany.
| |
Collapse
|
8
|
Patel YD, Brown AJ, Zhu J, Rosignoli G, Gibson SJ, Hatton D, James DC. Control of Multigene Expression Stoichiometry in Mammalian Cells Using Synthetic Promoters. ACS Synth Biol 2021; 10:1155-1165. [PMID: 33939428 PMCID: PMC8296667 DOI: 10.1021/acssynbio.0c00643] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Indexed: 01/22/2023]
Abstract
To successfully engineer mammalian cells for a desired purpose, multiple recombinant genes are required to be coexpressed at a specific and optimal ratio. In this study, we hypothesized that synthetic promoters varying in transcriptional activity could be used to create single multigene expression vectors coexpressing recombinant genes at a predictable relative stoichiometry. A library of 27 multigene constructs was created comprising three discrete fluorescent reporter gene transcriptional units in fixed series, each under the control of either a relatively low, medium, or high transcriptional strength synthetic promoter in every possible combination. Expression of each reporter gene was determined by absolute quantitation qRT-PCR in CHO cells. The synthetic promoters did generally function as designed within a multigene vector context; however, significant divergences from predicted promoter-mediated transcriptional activity were observed. First, expression of all three genes within a multigene vector was repressed at varying levels relative to coexpression of identical reporter genes on separate single gene vectors at equivalent gene copies. Second, gene positional effects were evident across all constructs where expression of the reporter genes in positions 2 and 3 was generally reduced relative to position 1. Finally, after accounting for general repression, synthetic promoter transcriptional activity within a local multigene vector format deviated from that expected. Taken together, our data reveal that mammalian synthetic promoters can be employed in vectors to mediate expression of multiple genes at predictable relative stoichiometries. However, empirical validation of functional performance is a necessary prerequisite, as vector and promoter design features can significantly impact performance.
Collapse
Affiliation(s)
- Yash D. Patel
- Department
of Chemical and Biological Engineering, The University of Sheffield, Mappin Street, Sheffield, S1 3JD, U.K.
| | - Adam J. Brown
- Department
of Chemical and Biological Engineering, The University of Sheffield, Mappin Street, Sheffield, S1 3JD, U.K.
| | - Jie Zhu
- Cell
Culture and Fermentation Sciences, BioPharmaceuticals Development,
R&D, AstraZeneca, Gaithersburg, Maryland 20878, United States
| | - Guglielmo Rosignoli
- Dynamic
Omics, Antibody Discovery & Protein Engineering, R&D, AstraZeneca, Cambridge, CB21 6GH, U.K.
| | - Suzanne J. Gibson
- Cell
Culture and Fermentation Sciences, BioPharmaceuticals Development,
R&D, AstraZeneca, Cambridge, CB21 6GH, U.K.
| | - Diane Hatton
- Cell
Culture and Fermentation Sciences, BioPharmaceuticals Development,
R&D, AstraZeneca, Cambridge, CB21 6GH, U.K.
| | - David C. James
- Department
of Chemical and Biological Engineering, The University of Sheffield, Mappin Street, Sheffield, S1 3JD, U.K.
| |
Collapse
|
9
|
Sandoval-Villegas N, Nurieva W, Amberger M, Ivics Z. Contemporary Transposon Tools: A Review and Guide through Mechanisms and Applications of Sleeping Beauty, piggyBac and Tol2 for Genome Engineering. Int J Mol Sci 2021; 22:ijms22105084. [PMID: 34064900 PMCID: PMC8151067 DOI: 10.3390/ijms22105084] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 04/30/2021] [Accepted: 05/05/2021] [Indexed: 01/19/2023] Open
Abstract
Transposons are mobile genetic elements evolved to execute highly efficient integration of their genes into the genomes of their host cells. These natural DNA transfer vehicles have been harnessed as experimental tools for stably introducing a wide variety of foreign DNA sequences, including selectable marker genes, reporters, shRNA expression cassettes, mutagenic gene trap cassettes, and therapeutic gene constructs into the genomes of target cells in a regulated and highly efficient manner. Given that transposon components are typically supplied as naked nucleic acids (DNA and RNA) or recombinant protein, their use is simple, safe, and economically competitive. Thus, transposons enable several avenues for genome manipulations in vertebrates, including transgenesis for the generation of transgenic cells in tissue culture comprising the generation of pluripotent stem cells, the production of germline-transgenic animals for basic and applied research, forward genetic screens for functional gene annotation in model species and therapy of genetic disorders in humans. This review describes the molecular mechanisms involved in transposition reactions of the three most widely used transposon systems currently available (Sleeping Beauty, piggyBac, and Tol2), and discusses the various parameters and considerations pertinent to their experimental use, highlighting the state-of-the-art in transposon technology in diverse genetic applications.
Collapse
Affiliation(s)
| | | | | | - Zoltán Ivics
- Correspondence: ; Tel.: +49-6103-77-6000; Fax: +49-6103-77-1280
| |
Collapse
|
10
|
Drummer C, Vogt EJ, Heistermann M, Roshani B, Becker T, Mätz-Rensing K, Kues WA, Kügler S, Behr R. Generation and Breeding of EGFP-Transgenic Marmoset Monkeys: Cell Chimerism and Implications for Disease Modeling. Cells 2021; 10:505. [PMID: 33673402 PMCID: PMC7996964 DOI: 10.3390/cells10030505] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 02/18/2021] [Accepted: 02/22/2021] [Indexed: 02/07/2023] Open
Abstract
Genetic modification of non-human primates (NHP) paves the way for realistic disease models. The common marmoset is a NHP species increasingly used in biomedical research. Despite the invention of RNA-guided nucleases, one strategy for protein overexpression in NHP is still lentiviral transduction. We generated three male and one female enhanced green fluorescent protein (EGFP)-transgenic founder marmosets via lentiviral transduction of natural preimplantation embryos. All founders accomplished germline transmission of the transgene by natural mating, yielding 20 transgenic offspring together (in total, 45 pups; 44% transgenic). This demonstrates that the transgenic gametes are capable of natural fertilization even when in competition with wildtype gametes. Importantly, 90% of the transgenic offspring showed transgene silencing, which is in sharp contrast to rodents, where the identical transgene facilitated robust EGFP expression. Furthermore, we consistently discovered somatic, but so far, no germ cell chimerism in mixed wildtype/transgenic litters. Somatic cell chimerism resulted in false-positive genotyping of the respective wildtype littermates. For the discrimination of transgenic from transgene-chimeric animals by polymerase chain reaction on skin samples, a chimeric cell depletion protocol was established. In summary, it is possible to establish a cohort of genetically modified marmosets by natural mating, but specific requirements including careful promoter selection are essential.
Collapse
Affiliation(s)
- Charis Drummer
- Platform Degenerative Diseases, German Primate Center–Leibniz Institute for Primate Research, Kellnerweg 4, 37077 Göttingen, Germany;
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, 37099 Göttingen, Germany
| | - Edgar-John Vogt
- Platform Degenerative Diseases, German Primate Center–Leibniz Institute for Primate Research, Kellnerweg 4, 37077 Göttingen, Germany;
| | - Michael Heistermann
- Endocrinology Laboratory, German Primate Center–Leibniz Institute for Primate Research, Kellnerweg 4, 37077 Göttingen, Germany;
| | - Berit Roshani
- Unit of Infection Models, German Primate Center–Leibniz Institute for Primate Research, Kellnerweg 4, 37077 Göttingen, Germany;
| | - Tamara Becker
- Primate Husbandry, German Primate Center–Leibniz Institute for Primate Research, Kellnerweg 4, 37077 Göttingen, Germany;
| | - Kerstin Mätz-Rensing
- Pathology Unit, German Primate Center–Leibniz-Institute for Primate Research, Kellnerweg 4, 37077 Göttingen, Germany;
| | - Wilfried A. Kues
- Friedrich-Loeffler-Institut, Institut für Nutztiergenetik, Mariensee, 31535 Neustadt, Germany;
| | - Sebastian Kügler
- Center for Nanoscale Microscopy and Physiology of the Brain (CNMPB) at Department of Neurology, University of Göttingen, Waldweg 33, 37073 Göttingen, Germany;
| | - Rüdiger Behr
- Platform Degenerative Diseases, German Primate Center–Leibniz Institute for Primate Research, Kellnerweg 4, 37077 Göttingen, Germany;
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, 37099 Göttingen, Germany
| |
Collapse
|
11
|
Singh B, Mal G, Kues WA, Yadav PS. The domesticated buffalo - An emerging model for experimental and therapeutic use of extraembryonic tissues. Theriogenology 2020; 151:95-102. [PMID: 32320839 DOI: 10.1016/j.theriogenology.2020.04.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 03/12/2020] [Accepted: 04/04/2020] [Indexed: 12/16/2022]
Abstract
Large animals play important roles as model animals for biomedical sciences and translational research. The water buffalo (Bubalus bubalis) is an economically important, multipurpose livestock species. Important assisted reproduction techniques, such as in vitro fertilization, cryo-conservation of sperm and embryos, embryo transfer, somatic cell nuclear transfer, genetic engineering, and genome editing have been successfully applied to buffaloes. Recently, detailed whole genome data and transcriptome maps have been generated. In addition, rapid progress has been made in stem cell biology of the buffalo. Apart from embryonic stem cells, bubaline extra-embryonic stem cells have gained particular interest. The multipotency of non-embryonic stem cells has been revealed, and their utility in basic and applied research is currently investigated. In particular, success achieved in bubaline extra-embryonic stem cells may have important roles in experimental biology and therapeutic regenerative medicine. Progress in other farm animals in assisted reproduction techniques, stem cell biology and genetic engineering, which could be of importance for buffalo, will also be briefly summarized.
Collapse
Affiliation(s)
- Birbal Singh
- ICAR-Indian Veterinary Research Institute, Regional Station Palampur, 176 061, India
| | - Gorakh Mal
- ICAR-Indian Veterinary Research Institute, Regional Station Palampur, 176 061, India
| | | | - Prem S Yadav
- ICAR-Central Institute for Research on Buffaloes, Hisar, 125001, India.
| |
Collapse
|
12
|
Kesselring L, Miskey C, Zuliani C, Querques I, Kapitonov V, Laukó A, Fehér A, Palazzo A, Diem T, Lustig J, Sebe A, Wang Y, Dinnyés A, Izsvák Z, Barabas O, Ivics Z. A single amino acid switch converts the Sleeping Beauty transposase into an efficient unidirectional excisionase with utility in stem cell reprogramming. Nucleic Acids Res 2020; 48:316-331. [PMID: 31777924 PMCID: PMC6943129 DOI: 10.1093/nar/gkz1119] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 11/07/2019] [Accepted: 11/22/2019] [Indexed: 12/26/2022] Open
Abstract
The Sleeping Beauty (SB) transposon is an advanced tool for genetic engineering and a useful model to investigate cut-and-paste DNA transposition in vertebrate cells. Here, we identify novel SB transposase mutants that display efficient and canonical excision but practically unmeasurable genomic re-integration. Based on phylogenetic analyses, we establish compensating amino acid replacements that fully rescue the integration defect of these mutants, suggesting epistasis between these amino acid residues. We further show that the transposons excised by the exc+/int− transposase mutants form extrachromosomal circles that cannot undergo a further round of transposition, thereby representing dead-end products of the excision reaction. Finally, we demonstrate the utility of the exc+/int− transposase in cassette removal for the generation of reprogramming factor-free induced pluripotent stem cells. Lack of genomic integration and formation of transposon circles following excision is reminiscent of signal sequence removal during V(D)J recombination, and implies that cut-and-paste DNA transposition can be converted to a unidirectional process by a single amino acid change.
Collapse
Affiliation(s)
- Lisa Kesselring
- Transposition and Genome Engineering, Division of Medical Biotechnology, Paul Ehrlich Institute, Langen, Germany
| | - Csaba Miskey
- Transposition and Genome Engineering, Division of Medical Biotechnology, Paul Ehrlich Institute, Langen, Germany
| | - Cecilia Zuliani
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg 69117, Germany
| | - Irma Querques
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg 69117, Germany
| | - Vladimir Kapitonov
- Transposition and Genome Engineering, Division of Medical Biotechnology, Paul Ehrlich Institute, Langen, Germany
| | | | - Anita Fehér
- BioTalentum Ltd, Gödöllő, 2100 Gödöllő, Hungary
| | - Antonio Palazzo
- Department of Biology, University of Bari 'Aldo Moro', Italy
| | - Tanja Diem
- Transposition and Genome Engineering, Division of Medical Biotechnology, Paul Ehrlich Institute, Langen, Germany
| | - Janna Lustig
- Transposition and Genome Engineering, Division of Medical Biotechnology, Paul Ehrlich Institute, Langen, Germany
| | - Attila Sebe
- Transposition and Genome Engineering, Division of Medical Biotechnology, Paul Ehrlich Institute, Langen, Germany
| | - Yongming Wang
- Mobile DNA, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | | | - Zsuzsanna Izsvák
- Mobile DNA, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Orsolya Barabas
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg 69117, Germany
| | - Zoltán Ivics
- Transposition and Genome Engineering, Division of Medical Biotechnology, Paul Ehrlich Institute, Langen, Germany
| |
Collapse
|
13
|
Hyder I, Eghbalsaied S, Kues WA. Systematic optimization of square-wave electroporation conditions for bovine primary fibroblasts. BMC Mol Cell Biol 2020; 21:9. [PMID: 32111153 PMCID: PMC7049184 DOI: 10.1186/s12860-020-00254-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 02/19/2020] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Gene transfer by electroporation is an established method for the non-viral mediated transfection of mammalian cells. Primary cells pose a particular challenge for electroporation-mediated gene transfer, since they are more vulnerable than immortalized cells, and have a limited proliferative capacity. Improving the gene transfer by using square wave electroporation in difficult to transfect cells, like bovine fetal fibroblasts, is a prerequisite for transgenic and further downstream experiments. RESULTS Here, bovine fetal fibroblasts were used for square-wave electroporation experiments in which the following parameters were systematically tested: electroporation buffer, electroporation temperature, pulse voltage, pulse duration, pulse number, cuvette type and plasmid DNA amount. For the experiments a commercially available square-wave generator was applied. Post electroporation, the bovine fetal fibroblasts were observed after 24 h for viability and reporter expression. The best results were obtained with a single 10 millisecond square-wave pulse of 400 V using 10 μg supercoiled plasmid DNA and 0.3 × 106 cells in 100 μl of Opti-MEM medium in 4 mm cuvettes. Importantly, the electroporation at room temperature was considerably better than with pre-cooled conditions. CONCLUSIONS The optimized electroporation conditions will be relevant for gene transfer experiments in bovine fetal fibroblasts to obtain genetically engineered donor cells for somatic cell nuclear transfer and for reprogramming experiments in this species.
Collapse
Affiliation(s)
- Iqbal Hyder
- Institute of Farm Animal Genetics, Friedrich-Loeffler-Institute, 31535, Neustadt, Germany.,Department of Veterinary Physiology, NTR College of Veterinary Science, Gannavaram, India
| | - Shahin Eghbalsaied
- Institute of Farm Animal Genetics, Friedrich-Loeffler-Institute, 31535, Neustadt, Germany.,Transgenesis Center of Excellence, Isfahan (Khorasgan) branch, Islamic Azad University, Isfahan, Iran
| | - Wilfried A Kues
- Institute of Farm Animal Genetics, Friedrich-Loeffler-Institute, 31535, Neustadt, Germany.
| |
Collapse
|
14
|
Mehta P, Kaushik R, Singh KP, Sharma A, Singh MK, Chauhan MS, Palta P, Singla SK, Manik RS. Comparative analysis of buffalo (Bubalus bubalis) non-transgenic and transgenic embryos containing human insulin gene, produced by SCNT. Theriogenology 2019; 135:25-32. [PMID: 31195358 DOI: 10.1016/j.theriogenology.2019.06.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 06/03/2019] [Accepted: 06/04/2019] [Indexed: 12/11/2022]
Abstract
Somatic cell nuclear transfer (SCNT), using transgenic donor cells, is a highly efficient method for producing transgenic embryos. We compared the developmental competence, quality and gene expression of transgenic embryos produced by Hand-made cloning from buffalo fetal fibroblasts (BFFs) containing human insulin gene, with non-transgenic embryos produced from BFFs (Controls). The expression vector (pAcISUBC), constructed by inserting human insulin gene between DNA fragments containing mammary gland-specific buffalo β-lactoglobulin (buBLG) promoter and terminator buBLG 3'UTR regions into pAcGFP-N1 vector, was used for obtaining the 11 kb insert for transfection of BFFs by nucleofection. Presence of the transgene in embryos was confirmed by examining GFP expression by RT-PCR and immunofluorescence. The blastocyst rate was lower (P < 0.05) for transgenic embryos than for controls (35.7 ± 1.8% vs 48.7 ± 2.4%). The apoptotic index was higher (P < 0.05) for transgenic than for control blastocysts which, in turn, was higher (P < 0.05) than for IVF counterparts (6.9 ± 0.9, 3.8 ± 0.5 and 1.8 ± 0.3, respectively). The total cell number was similar for transgenic and non-transgenic blastocysts (143.2 ± 17.0 and 137.2 ± 7.6, respectively). The expression level of pro-apoptotic genes BAX and BID but not that of CASP3 and CASP9, and cell cycle check point control-related gene P53 was higher (P < 0.05), and that of development- (IGF-1R and G6PD) and pluripotency-related gene NANOG was lower (P < 0.05) in transgenic than in control embryos. The expression level of epigenetic-related genes DNMT1, DNMT3a and HDAC1 and pluripotency-related gene OCT4 was similar in the two groups. The expression level of BAX, BID, CASP9, P53, DNMT1 and DNMT3a was higher (P < 0.05) and that of OCT4, NANOG IGF-1R and G6PD was lower (P < 0.05) in cloned transgenic than in IVF blastocysts whereas, that of CASP3 and HDAC1 was similar between the two groups. In conclusion, these results suggest that transgenic embryos produced by SCNT have lower developmental competence and quality, and altered gene expression compared to non-transgenic embryos.
Collapse
Affiliation(s)
- P Mehta
- Embryo Biotechnology Laboratory, Animal Biotechnology Centre, ICAR-National Dairy Research Institute, Karnal, 132001, India.
| | - R Kaushik
- Embryo Biotechnology Laboratory, Animal Biotechnology Centre, ICAR-National Dairy Research Institute, Karnal, 132001, India
| | - K P Singh
- Embryo Biotechnology Laboratory, Animal Biotechnology Centre, ICAR-National Dairy Research Institute, Karnal, 132001, India
| | - A Sharma
- Embryo Biotechnology Laboratory, Animal Biotechnology Centre, ICAR-National Dairy Research Institute, Karnal, 132001, India
| | - M K Singh
- Embryo Biotechnology Laboratory, Animal Biotechnology Centre, ICAR-National Dairy Research Institute, Karnal, 132001, India
| | - M S Chauhan
- Embryo Biotechnology Laboratory, Animal Biotechnology Centre, ICAR-National Dairy Research Institute, Karnal, 132001, India
| | - P Palta
- Embryo Biotechnology Laboratory, Animal Biotechnology Centre, ICAR-National Dairy Research Institute, Karnal, 132001, India
| | - S K Singla
- Embryo Biotechnology Laboratory, Animal Biotechnology Centre, ICAR-National Dairy Research Institute, Karnal, 132001, India
| | - R S Manik
- Embryo Biotechnology Laboratory, Animal Biotechnology Centre, ICAR-National Dairy Research Institute, Karnal, 132001, India
| |
Collapse
|
15
|
Hodge R, Narayanavari SA, Izsvák Z, Ivics Z. Wide Awake and Ready to Move: 20 Years of Non-Viral Therapeutic Genome Engineering with the Sleeping Beauty Transposon System. Hum Gene Ther 2018; 28:842-855. [PMID: 28870121 DOI: 10.1089/hum.2017.130] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Gene therapies will only become a widespread tool in the clinical treatment of human diseases with the advent of gene transfer vectors that integrate genetic information stably, safely, effectively, and economically. Two decades after the discovery of the Sleeping Beauty (SB) transposon, it has been transformed into a vector system that is fulfilling these requirements. SB may well overcome some of the limitations associated with viral gene transfer vectors and transient non-viral gene delivery approaches that are being used in the majority of ongoing clinical trials. The SB system has achieved a high level of stable gene transfer and sustained transgene expression in multiple primary human somatic cell types, representing crucial steps that may permit its clinical use in the near future. This article reviews the most important aspects of SB as a tool for gene therapy, including aspects of its vectorization and genomic integration. As an illustration, the clinical development of the SB system toward gene therapy of age-related macular degeneration and cancer immunotherapy is highlighted.
Collapse
Affiliation(s)
- Russ Hodge
- 1 Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC) , Berlin, Germany
| | - Suneel A Narayanavari
- 1 Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC) , Berlin, Germany
| | - Zsuzsanna Izsvák
- 1 Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC) , Berlin, Germany
| | - Zoltán Ivics
- 2 Division of Medical Biotechnology, Paul Ehrlich Institute , Langen, Germany
| |
Collapse
|
16
|
Yum SY, Lee SJ, Park SG, Shin IG, Hahn SE, Choi WJ, Kim HS, Kim HJ, Bae SH, Lee JH, Moon JY, Lee WS, Lee JH, Lee CI, Kim SJ, Jang G. Long-term health and germline transmission in transgenic cattle following transposon-mediated gene transfer. BMC Genomics 2018; 19:387. [PMID: 29792157 PMCID: PMC5966871 DOI: 10.1186/s12864-018-4760-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Accepted: 05/04/2018] [Indexed: 12/25/2022] Open
Abstract
Background Transposon-mediated, non-viral gene delivery is a powerful tool for generating stable cell lines and transgenic animals. However, as multi-copy insertion is the preferred integration pattern, there is the potential for uncontrolled changes in endogenous gene expression and detrimental effects in cells or animals. Our group has previously reported on the generation of several transgenic cattle by using microinjection of the Sleeping Beauty (SB) and PiggyBac (PB) transposons and seeks to explore the long-term effects of this technology on cattle. Results Transgenic cattle, one female (SNU-SB-1) and one male (SNU-PB-1), reached over 36 months of age with no significant health issues and normal blood parameters. The detection of transgene integration and fluorescent signal in oocytes and sperm suggested the capacity for germline transmission in both of the founder animals. After natural breeding, the founder transgenic cow delivered a male calf and secreted milk containing fluorescent transgenic proteins. The calf expressed green fluorescent protein in primary cells from ear skin, with no significant change in overall genomic stability and blood parameters. Three sites of transgene integration were identified by next-generation sequencing of the calf’s genome. Conclusions Overall, these data demonstrate that transposon-mediated transgenesis can be applied to cattle without being detrimental to their long-term genomic stability or general health. We further suggest that this technology may be usefully applied in other fields, such as the generation of transgenic animal models. Electronic supplementary material The online version of this article (10.1186/s12864-018-4760-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Soo-Young Yum
- Department of Theriogenology, College of Veterinary Medicine and the Research Institute of Veterinary Science, Seoul National University, #631 Building 85, Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea
| | - Song-Jeon Lee
- Embryo Research Center, Seoul Milk Coop, Gyeonggi-do, 12528, Republic of Korea
| | - Sin-Gi Park
- Bioinformatics Team, Theragen Etex Bio Institute, Advanced Institutes of Convergence Technology, Kwanggyo Technovalley, Suwon, 16229, Republic of Korea
| | - In-Gang Shin
- Bioinformatics Team, Theragen Etex Bio Institute, Advanced Institutes of Convergence Technology, Kwanggyo Technovalley, Suwon, 16229, Republic of Korea
| | - Sang-Eun Hahn
- Department of Theriogenology, College of Veterinary Medicine and the Research Institute of Veterinary Science, Seoul National University, #631 Building 85, Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea
| | - Woo-Jae Choi
- Department of Theriogenology, College of Veterinary Medicine and the Research Institute of Veterinary Science, Seoul National University, #631 Building 85, Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea
| | - Hee-Soo Kim
- Embryo Research Center, Seoul Milk Coop, Gyeonggi-do, 12528, Republic of Korea
| | - Hyeong-Jong Kim
- Embryo Research Center, Seoul Milk Coop, Gyeonggi-do, 12528, Republic of Korea
| | - Seong-Hun Bae
- Embryo Research Center, Seoul Milk Coop, Gyeonggi-do, 12528, Republic of Korea
| | - Je-Hyeong Lee
- Embryo Research Center, Seoul Milk Coop, Gyeonggi-do, 12528, Republic of Korea
| | - Joo-Yeong Moon
- Embryo Research Center, Seoul Milk Coop, Gyeonggi-do, 12528, Republic of Korea
| | - Woo-Sung Lee
- Embryo Research Center, Seoul Milk Coop, Gyeonggi-do, 12528, Republic of Korea
| | - Ji-Hyun Lee
- Department of Theriogenology, College of Veterinary Medicine and the Research Institute of Veterinary Science, Seoul National University, #631 Building 85, Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea
| | - Choong-Il Lee
- Department of Theriogenology, College of Veterinary Medicine and the Research Institute of Veterinary Science, Seoul National University, #631 Building 85, Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea
| | - Seong-Jin Kim
- Bioinformatics Team, Theragen Etex Bio Institute, Advanced Institutes of Convergence Technology, Kwanggyo Technovalley, Suwon, 16229, Republic of Korea
| | - Goo Jang
- Department of Theriogenology, College of Veterinary Medicine and the Research Institute of Veterinary Science, Seoul National University, #631 Building 85, Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea. .,Emergence Center for Food-Medicine Personalized Therapy System, Advanced Institutes of Convergence Technology, Seoul National University, Gyeonggi-do, 16229, Republic of Korea.
| |
Collapse
|
17
|
Generation of Venus fluorochrome expressing transgenic handmade cloned buffalo embryos using Sleeping Beauty transposon. Tissue Cell 2018; 51:49-55. [DOI: 10.1016/j.tice.2018.02.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 02/21/2018] [Accepted: 02/23/2018] [Indexed: 12/27/2022]
|
18
|
Amann RP, Saacke RG, Barbato GF, Waberski D. Measuring Male-to-Male Differences in Fertility or Effects of Semen Treatments. Annu Rev Anim Biosci 2018; 6:255-286. [DOI: 10.1146/annurev-animal-030117-014829] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Rupert P. Amann
- Animal Reproduction and Biotechnology, Colorado State University, Fort Collins, Colorado 80523, USA
| | - Richard G. Saacke
- Department of Dairy Science, Virginia Polytechnic Institute and State University, Blacksburg, Virginia 24061, USA
| | - Guy F. Barbato
- Biology Program, Stockton University, Galloway, New Jersey 08205, USA
| | - Dagmar Waberski
- Unit for Reproductive Medicine of Clinics/Clinic for Pigs and Small Ruminants, University of Veterinary Medicine Hannover, 30559 Hannover, Germany
| |
Collapse
|
19
|
Yum SY, Youn KY, Choi WJ, Jang G. Development of genome engineering technologies in cattle: from random to specific. J Anim Sci Biotechnol 2018; 9:16. [PMID: 29423215 PMCID: PMC5789629 DOI: 10.1186/s40104-018-0232-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 01/09/2018] [Indexed: 12/16/2022] Open
Abstract
The production of transgenic farm animals (e.g., cattle) via genome engineering for the gain or loss of gene functions is an important undertaking. In the initial stages of genome engineering, DNA micro-injection into one-cell stage embryos (zygotes) followed by embryo transfer into a recipient was performed because of the ease of the procedure. However, as this approach resulted in severe mosaicism and has a low efficiency, it is not typically employed in the cattle as priority, unlike in mice. To overcome the above issue with micro-injection in cattle, somatic cell nuclear transfer (SCNT) was introduced and successfully used to produce cloned livestock. The application of SCNT for the production of transgenic livestock represents a significant advancement, but its development speed is relatively slow because of abnormal reprogramming and low gene targeting efficiency. Recent genome editing technologies (e.g., ZFN, TALEN, and CRISPR-Cas9) have been rapidly adapted for applications in cattle and great results have been achieved in several fields such as disease models and bioreactors. In the future, genome engineering technologies will accelerate our understanding of genetic traits in bovine and will be readily adapted for bio-medical applications in cattle.
Collapse
Affiliation(s)
- Soo-Young Yum
- 1Department of Veterinary Clinical Science, College of Veterinary Medicine and the Research Institute of Veterinary Science, Seoul National University, Seoul, 08826 Republic of Korea
| | - Ki-Young Youn
- 1Department of Veterinary Clinical Science, College of Veterinary Medicine and the Research Institute of Veterinary Science, Seoul National University, Seoul, 08826 Republic of Korea
| | - Woo-Jae Choi
- 1Department of Veterinary Clinical Science, College of Veterinary Medicine and the Research Institute of Veterinary Science, Seoul National University, Seoul, 08826 Republic of Korea
| | - Goo Jang
- 1Department of Veterinary Clinical Science, College of Veterinary Medicine and the Research Institute of Veterinary Science, Seoul National University, Seoul, 08826 Republic of Korea.,2Farm Animal Clinical Training and Research Center, Institute of GreenBio Science Technology, Seoul National University, PyeongChang-Gun, Gangwon-do 25354 Republic of Korea.,3Emergence Center for Food-Medicine Personalized Therapy System, Advanced Institutes of Convergence Technology, Seoul National University, SuWon, Gyeonggi-do 16629 Republic of Korea.,4College of Veterinary Medicine, Seoul National University, #85, Room631, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826 Republic of Korea
| |
Collapse
|
20
|
The abundance of health-associated bacteria is altered in PAH polluted soils-Implications for health in urban areas? PLoS One 2017; 12:e0187852. [PMID: 29145477 PMCID: PMC5690629 DOI: 10.1371/journal.pone.0187852] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2017] [Accepted: 10/29/2017] [Indexed: 01/01/2023] Open
Abstract
Long-term exposure to polyaromatic hydrocarbons (PAHs) has been connected to chronic human health disorders. It is also well-known that i) PAH contamination alters soil bacterial communities, ii) human microbiome is associated with environmental microbiome, and iii) alteration in the abundance of members in several bacterial phyla is associated with adverse or beneficial human health effects. We hypothesized that soil pollution by PAHs altered soil bacterial communities that had known associations with human health. The rationale behind our study was to increase understanding and potentially facilitate reconsidering factors that lead to health disorders in areas characterized by PAH contamination. Large containers filled with either spruce forest soil, pine forest soil, peat, or glacial sand were left to incubate or contaminated with creosote. Biological degradation of PAHs was monitored using GC-MS, and the bacterial community composition was analyzed using 454 pyrosequencing. Proteobacteria had higher and Actinobacteria and Bacteroidetes had lower relative abundance in creosote contaminated soils than in non-contaminated soils. Earlier studies have demonstrated that an increase in the abundance of Proteobacteria and decreased abundance of the phyla Actinobacteria and Bacteroidetes are particularly associated with adverse health outcomes and immunological disorders. Therefore, we propose that pollution-induced shifts in natural soil bacterial community, like in PAH-polluted areas, can contribute to the prevalence of chronic diseases. We encourage studies that simultaneously address the classic “adverse toxin effect” paradigm and our novel “altered environmental microbiome” hypothesis.
Collapse
|
21
|
Kebriaei P, Izsvák Z, Narayanavari SA, Singh H, Ivics Z. Gene Therapy with the Sleeping Beauty Transposon System. Trends Genet 2017; 33:852-870. [PMID: 28964527 DOI: 10.1016/j.tig.2017.08.008] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2017] [Revised: 08/24/2017] [Accepted: 08/31/2017] [Indexed: 11/16/2022]
Abstract
The widespread clinical implementation of gene therapy requires the ability to stably integrate genetic information through gene transfer vectors in a safe, effective, and economical manner. The latest generation of Sleeping Beauty (SB) transposon vectors fulfills these requirements, and may overcome limitations associated with viral gene transfer vectors and transient nonviral gene delivery approaches that are prevalent in ongoing clinical trials. The SB system enables high-level stable gene transfer and sustained transgene expression in multiple primary human somatic cell types, thereby representing a highly attractive gene transfer strategy for clinical use. Here, we review the most important aspects of using SB for gene therapy, including vectorization as well as genomic integration features. We also illustrate the path to successful clinical implementation by highlighting the application of chimeric antigen receptor (CAR)-modified T cells in cancer immunotherapy.
Collapse
Affiliation(s)
- Partow Kebriaei
- Department of Stem Cell Transplant and Cellular Therapy, MD Anderson Cancer Center, Houston, TX, USA
| | - Zsuzsanna Izsvák
- Mobile DNA, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Suneel A Narayanavari
- Mobile DNA, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Harjeet Singh
- Department of Pediatrics, MD Anderson Cancer Center, Houston, TX, USA
| | - Zoltán Ivics
- Division of Medical Biotechnology, Paul Ehrlich Institute, Langen, Germany.
| |
Collapse
|
22
|
Wang M, Sun Z, Yu T, Ding F, Li L, Wang X, Fu M, Wang H, Huang J, Li N, Dai Y. Large-scale production of recombinant human lactoferrin from high-expression, marker-free transgenic cloned cows. Sci Rep 2017; 7:10733. [PMID: 28878310 PMCID: PMC5587717 DOI: 10.1038/s41598-017-11462-z] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Accepted: 08/25/2017] [Indexed: 11/23/2022] Open
Abstract
Human lactoferrin (hLF) is a valuable protein for pharmaceutical products and functional foods, and worldwide demand for this protein has steadily increased. However, large-scale recombinant human lactoferrin (rhLF) production using current animal bioreactor techniques is limited by the low expression of foreign proteins, the use of antibiotic resistance genes and the down-regulation of endogenous milk proteins. Here, we generated a herd of marker-free, hLF bacterial artificial chromosome (BAC) transgenic cloned cows, as confirmed by Polymerase chain reaction, Southern blot and Western blot analyses. These transgenic cloned cows produced rhLF in milk at concentrations of 4.5–13.6 g/L. Moreover, the total protein content of the milk was increased. Over two hundred transgenic cloned cows were propagated by multiple ovulation and embryo transfer (MOET). A total of 400–450 g of rhLF protein, which shows similar enzymatic activity to natural hLF in iron binding and release, can be purified on a large scale from >100 L of milk per day. Our results suggested that transgenic bovine mammary bioreactors have the potential for large-scale protein production.
Collapse
Affiliation(s)
- Ming Wang
- State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Zhaolin Sun
- State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Tian Yu
- Kejienuo Biotechnology Company, Wuxi, China
| | - Fangrong Ding
- State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Ling Li
- State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Xi Wang
- Kejienuo Biotechnology Company, Wuxi, China
| | - Mingbo Fu
- Kejienuo Biotechnology Company, Wuxi, China
| | - Haiping Wang
- State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Jinming Huang
- Dairy cattle Research Center, Academy of Agricultural Sciences, Shandong, China
| | - Ning Li
- State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China.
| | - Yunping Dai
- State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China.
| |
Collapse
|
23
|
RNAi combining Sleeping Beauty transposon system inhibits ex vivo expression of foot-and-mouth disease virus VP1 in transgenic sheep cells. Sci Rep 2017; 7:10065. [PMID: 28855524 PMCID: PMC5577316 DOI: 10.1038/s41598-017-09302-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Accepted: 07/25/2017] [Indexed: 11/08/2022] Open
Abstract
Foot and mouth disease, which is induced by the foot and mouth disease virus (FMDV), takes its toll on the cloven-hoofed domestic animals. The VP1 gene in FMDV genome encodes the viral capsid, a vital element for FMDV replication. Sleeping Beauty (SB) is an active DNA-transposon system responsible for genetic transformation and insertional mutagenesis in vertebrates. In this study, a conserved VP1-shRNA which specifically targets the ovine FMDV-VP1 gene was constructed and combined with SB transposase and transposon. Then, they were microinjected into pronuclear embryos to breed transgenic sheep. Ninety-two lambs were born and the VP1-shRNA was positively integrated into eight of them. The rate of transgenic sheep production in SB transposon system was significantly higher than that in controls (13.04% vs. 3.57% and 7.14%, P < 0.05). The ear fibroblasts of the transgenic lambs transfected with the PsiCheck2-VP1 vector had a significant inhibitory effect on the VP1 gene of the FMDV. In conclusion, the VP1-shRNA transgenic sheep were successfully generated by the current new method. The ear fibroblasts from these transgenic sheep possess a great resistance to FMDV. The result indicated that RNAi technology combining the "Sleeping Beauty" transposon system is an efficient method to produce transgenic animals.
Collapse
|
24
|
A Novel Ideal Radionuclide Imaging System for Non-invasively Cell Monitoring built on Baculovirus Backbone by Introducing Sleeping Beauty Transposon. Sci Rep 2017; 7:43879. [PMID: 28262785 PMCID: PMC5338331 DOI: 10.1038/srep43879] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Accepted: 01/30/2017] [Indexed: 12/17/2022] Open
Abstract
Sleeping Beauty (SB) transposon is an attractive tool in stable transgene integration both in vitro and in vivo; and we introduced SB transposon into recombinant sodium-iodide symporter baculovirus system (Bac-NIS system) to facilitate long-term expression of recombinant sodium-iodide symporter. In our study, two hybrid baculovirus systems (Bac-eGFP-SB-NeoR and Bac-NIS-SB-NeoR) were successfully constructed and used to infect U87 glioma cells. After G418 selection screening, the Bac-eGFP-SB-NeoR-U87 cells remained eGFP positive, at the 18th and 196th day post transfection (96.03 ± 0.21% and 97.43 ± 0.81%), while eGFP positive population declined significantly at 18 days in cells transfected with unmodified baculovirus construct. NIS gene expression by Bac-NIS-SB-NeoR-U87 cells was also maintained for 28 weeks as determined by radioiodine uptake assay, reverse transcription-polymerase chain reaction (RT-PCR) and Western Blot (WB) assay. When transplanted in mice, Bac-NIS-SB-NeoR-U87 cells also expressed NIS gene stably as monitored by SPECT imaging for 43 days until the tumor-bearing mice were sacrificed. Herein, we showed that incorporation of SB in Bac-NIS system (hybrid Bac-NIS-SB-NeoR) can achieve a long-term transgene expression and can improve radionuclide imaging in cell tracking and monitoring in vivo.
Collapse
|
25
|
Vargas JE, Chicaybam L, Stein RT, Tanuri A, Delgado-Cañedo A, Bonamino MH. Retroviral vectors and transposons for stable gene therapy: advances, current challenges and perspectives. J Transl Med 2016; 14:288. [PMID: 27729044 PMCID: PMC5059932 DOI: 10.1186/s12967-016-1047-x] [Citation(s) in RCA: 94] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Accepted: 10/03/2016] [Indexed: 12/15/2022] Open
Abstract
Gene therapy protocols require robust and long-term gene expression. For two decades, retrovirus family vectors have offered several attractive properties as stable gene-delivery vehicles. These vectors represent a technology with widespread use in basic biology and translational studies that require persistent gene expression for treatment of several monogenic diseases. Immunogenicity and insertional mutagenesis represent the main obstacles to a wider clinical use of these vectors. Efficient and safe non-viral vectors are emerging as a promising alternative and facilitate clinical gene therapy studies. Here, we present an updated review for beginners and expert readers on retro and lentiviruses and the latest generation of transposon vectors (sleeping beauty and piggyBac) used in stable gene transfer and gene therapy clinical trials. We discuss the potential advantages and disadvantages of these systems such as cellular responses (immunogenicity or genome modification of the target cell) following exogenous DNA integration. Additionally, we discuss potential implications of these genome modification tools in gene therapy and other basic and applied science contexts.
Collapse
Affiliation(s)
- José Eduardo Vargas
- Centro Infantil-Pontifícia Universidade Católica do Rio Grande do Sul-PUCRS, Porto Alegre, Brazil
| | - Leonardo Chicaybam
- Programa de Carcinogênese Molecular, Instituto Nacional de Câncer (INCA), Rua Andre Cavalcanti 37/6º andar, Centro, Rio de Janeiro, 20231-050, Brazil.,Vice-presidência de Pesquisa e Laboratórios de Referência, Fundação Instituto Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Renato Tetelbom Stein
- Centro Infantil-Pontifícia Universidade Católica do Rio Grande do Sul-PUCRS, Porto Alegre, Brazil
| | - Amilcar Tanuri
- Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Martin H Bonamino
- Programa de Carcinogênese Molecular, Instituto Nacional de Câncer (INCA), Rua Andre Cavalcanti 37/6º andar, Centro, Rio de Janeiro, 20231-050, Brazil. .,Vice-presidência de Pesquisa e Laboratórios de Referência, Fundação Instituto Oswaldo Cruz, Rio de Janeiro, Brazil.
| |
Collapse
|
26
|
Narayanavari SA, Chilkunda SS, Ivics Z, Izsvák Z. Sleeping Beauty transposition: from biology to applications. Crit Rev Biochem Mol Biol 2016; 52:18-44. [PMID: 27696897 DOI: 10.1080/10409238.2016.1237935] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Sleeping Beauty (SB) is the first synthetic DNA transposon that was shown to be active in a wide variety of species. Here, we review studies from the last two decades addressing both basic biology and applications of this transposon. We discuss how host-transposon interaction modulates transposition at different steps of the transposition reaction. We also discuss how the transposon was translated for gene delivery and gene discovery purposes. We critically review the system in clinical, pre-clinical and non-clinical settings as a non-viral gene delivery tool in comparison with viral technologies. We also discuss emerging SB-based hybrid vectors aimed at combining the attractive safety features of the transposon with effective viral delivery. The success of the SB-based technology can be fundamentally attributed to being able to insert fairly randomly into genomic regions that allow stable long-term expression of the delivered transgene cassette. SB has emerged as an efficient and economical toolkit for safe and efficient gene delivery for medical applications.
Collapse
Affiliation(s)
- Suneel A Narayanavari
- a Mobile DNA , Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC) , Berlin , Germany
| | - Shreevathsa S Chilkunda
- a Mobile DNA , Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC) , Berlin , Germany
| | - Zoltán Ivics
- b Division of Medical Biotechnology , Paul Ehrlich Institute , Langen , Germany
| | - Zsuzsanna Izsvák
- a Mobile DNA , Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC) , Berlin , Germany
| |
Collapse
|
27
|
Mukherjee A, Garrels W, Talluri TR, Tiedemann D, Bősze Z, Ivics Z, Kues WA. Expression of Active Fluorophore Proteins in the Milk of Transgenic Pigs Bypassing the Secretory Pathway. Sci Rep 2016; 6:24464. [PMID: 27086548 PMCID: PMC4834472 DOI: 10.1038/srep24464] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Accepted: 03/30/2016] [Indexed: 12/12/2022] Open
Abstract
We describe the expression of recombinant fluorescent proteins in the milk of two lines of transgenic pigs generated by Sleeping Beauty transposon-mediated genetic engineering. The Sleeping Beauty transposon consisted of an ubiquitously active CAGGS promoter driving a fluorophore cDNA, encoding either Venus or mCherry. Importantly, the fluorophore cDNAs did not encode for a signal peptide for the secretory pathway, and in previous studies of the transgenic animals a cytoplasmic localization of the fluorophore proteins was found. Unexpectedly, milk samples from lactating sows contained high levels of bioactive Venus or mCherry fluorophores. A detailed analysis suggested that exfoliated cells of the mammary epithelium carried the recombinant proteins passively into the milk. This is the first description of reporter fluorophore expression in the milk of livestock, and the findings may contribute to the development of an alternative concept for the production of bioactive recombinant proteins in the udder.
Collapse
Affiliation(s)
- Ayan Mukherjee
- Friedrich-Loeffler-Institut, Institut für Nutztiergenetik, Mariensee, Germany
| | - Wiebke Garrels
- Medical School Hannover, Institute of Laboratory Animal Sciences, Hannover, Germany
| | | | - Daniela Tiedemann
- Friedrich-Loeffler-Institut, Institut für Nutztiergenetik, Mariensee, Germany
| | - Zsuzsanna Bősze
- NARIC- Agricultural Biotechnology Institute, Gödöllö, Hungary
| | | | - Wilfried A. Kues
- Friedrich-Loeffler-Institut, Institut für Nutztiergenetik, Mariensee, Germany
| |
Collapse
|