1
|
Wang X, Qiao X, Sui L, Zhao H, Li F, Tang YD, Shi W, Guo Y, Jiang Y, Wang L, Zhou H, Tang L, Xu Y, Li Y. Establishment of stable Vero cell lines expressing TMPRSS2 and MSPL: A useful tool for propagating porcine epidemic diarrhea virus in the absence of exogenous trypsin. Virulence 2021; 11:669-685. [PMID: 32471322 PMCID: PMC7550007 DOI: 10.1080/21505594.2020.1770491] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Porcine epidemic diarrhea virus (PEDV) is the causative agent of porcine epidemic diarrhea, causing substantial economic losses to the swine industry worldwide. However, the development of PEDV vaccine is hampered by its low propagation titer in vitro, due to difficulty in adapting to the cells and complex culture conditions, even in the presence of trypsin. Furthermore, the frequent variation, recombination, and evolution of PEDV resulted in reemergence and vaccination failure. In this study, we established the Vero/TMPRSS2 and Vero/MSPL cell lines, constitutively expressing type II transmembrane serine protease TMPRSS2 and MSPL, in order to increase the stability and titer of PEDV culture and isolation in vitro. Our study revealed that the Vero/TMPRSS2, especially Vero/MSPL cell lines, can effectively facilitate the titer and multicycle replication of cell-adapted PEDV in the absence of exogenous trypsin, by cleaving and activating PEDV S protein. Furthermore, our results also highlighted that Vero/TMPRSS2 and Vero/MSPL cells can significantly enhance the isolation of PEDV from the clinical tissue samples as well as promote viral infection and replication by cell-cell fusion. The successful construction of the Vero/TMPRSS2 and Vero/MSPL cell lines provides a useful approach for the isolation and propagation of PEDV, simplification of virus culture, and large-scale production of industrial vaccine, and the cell lines are also an important system to research PEDV S protein cleaved by host protease.
Collapse
Affiliation(s)
- Xiaona Wang
- College of Veterinary Medicine, Northeast Agricultural University , Harbin, P.R. China
| | - Xinyuan Qiao
- College of Veterinary Medicine, Northeast Agricultural University , Harbin, P.R. China.,Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development , Harbin, P.R. China
| | - Ling Sui
- College of Veterinary Medicine, Northeast Agricultural University , Harbin, P.R. China
| | - Haiyuan Zhao
- Department of Swine Breeding, Jiangsu Hanswine Food Co., Ltd , Ma'anshan, Anhui Province, China
| | - Fengsai Li
- College of Veterinary Medicine, Northeast Agricultural University , Harbin, P.R. China
| | - Yan-Dong Tang
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences , Harbin, China
| | - Wen Shi
- College of Animal Science and Technology, Northeast Agricultural University , Harbin, P.R. China
| | - Yuyao Guo
- College of Veterinary Medicine, Northeast Agricultural University , Harbin, P.R. China
| | - Yanping Jiang
- College of Veterinary Medicine, Northeast Agricultural University , Harbin, P.R. China
| | - Li Wang
- College of Veterinary Medicine, Northeast Agricultural University , Harbin, P.R. China
| | - Han Zhou
- College of Veterinary Medicine, Northeast Agricultural University , Harbin, P.R. China
| | - Lijie Tang
- College of Veterinary Medicine, Northeast Agricultural University , Harbin, P.R. China.,Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development , Harbin, P.R. China
| | - Yigang Xu
- College of Veterinary Medicine, Northeast Agricultural University , Harbin, P.R. China.,Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development , Harbin, P.R. China
| | - Yijing Li
- College of Veterinary Medicine, Northeast Agricultural University , Harbin, P.R. China.,Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development , Harbin, P.R. China
| |
Collapse
|
2
|
Springer C, Wolf E, Simmet K. A New Toolbox in Experimental Embryology-Alternative Model Organisms for Studying Preimplantation Development. J Dev Biol 2021; 9:15. [PMID: 33918361 PMCID: PMC8167745 DOI: 10.3390/jdb9020015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 03/28/2021] [Accepted: 03/30/2021] [Indexed: 02/06/2023] Open
Abstract
Preimplantation development is well conserved across mammalian species, but major differences in developmental kinetics, regulation of early lineage differentiation and implantation require studies in different model organisms, especially to better understand human development. Large domestic species, such as cattle and pig, resemble human development in many different aspects, i.e., the timing of zygotic genome activation, mechanisms of early lineage differentiations and the period until blastocyst formation. In this article, we give an overview of different assisted reproductive technologies, which are well established in cattle and pig and make them easily accessible to study early embryonic development. We outline the available technologies to create genetically modified models and to modulate lineage differentiation as well as recent methodological developments in genome sequencing and imaging, which form an immense toolbox for research. Finally, we compare the most recent findings in regulation of the first lineage differentiations across species and show how alternative models enhance our understanding of preimplantation development.
Collapse
Affiliation(s)
- Claudia Springer
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, Ludwig-Maximilians-Universität München, 85764 Oberschleissheim, Germany; (C.S.); (E.W.)
| | - Eckhard Wolf
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, Ludwig-Maximilians-Universität München, 85764 Oberschleissheim, Germany; (C.S.); (E.W.)
- Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, Ludwig-Maximilians-Universität München, 81377 Munich, Germany
- Center for Innovative Medical Models (CiMM), Ludwig-Maximilians-Universität München, 85764 Oberschleissheim, Germany
| | - Kilian Simmet
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, Ludwig-Maximilians-Universität München, 85764 Oberschleissheim, Germany; (C.S.); (E.W.)
| |
Collapse
|
3
|
Wen J, Wu J, Cao T, Zhi S, Chen Y, Aagaard L, Zhen P, Huang Y, Zhong J, Huang J. Methylation silencing and reactivation of exogenous genes in lentivirus-mediated transgenic mice. Transgenic Res 2021; 30:63-76. [PMID: 33394315 DOI: 10.1007/s11248-020-00224-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 11/24/2020] [Indexed: 12/27/2022]
Abstract
Taking advantage of their ability to integrate their genomes into the host genome, lentiviruses have been used to rapidly produce transgenic mice in biomedical research. In most cases, transgenes delivered by lentiviral vectors have resisted silencing mediated by epigenetic modifications in mice. However, some studies revealed that methylation caused decreased transgene expression in mice. Therefore, there is conflicting evidence regarding the methylation-induced silencing of transgenes delivered by lentiviral transduction in mice. In this study, we present evidence that the human TTR transgene was silenced by DNA methylation in the liver of a transgenic mouse model generated by lentiviral transduction. The density of methylation on the transgene was increased during reproduction, and the expression of the transgene was completely silenced in mice of the F2 generation. Interestingly, 5-azacytidine (5-AzaC), a methyltransferase inhibitor, potently reactivated the silenced genes in neonatal mice whose hepatocytes were actively proliferating and led to stable transgene expression during development. However, 5-AzaC did not rescue liver transgene expression when administered to adult mice. Moreover, 5-AzaC at the given dose had low developmental toxicity in the newborn mice. In summary, we demonstrate the methylation-induced silencing of an exogenous gene in the liver of a mouse model generated by lentiviral transduction and show that the silenced transgene can be safely and efficiently reactivated by 5-AzaC treatment, providing an alternative way to obtain progeny with stable transgene expression in the case of the methylation of exogenous genes in transgenic mice generated by lentiviral transduction.
Collapse
Affiliation(s)
- Jinkun Wen
- Department of Neurology, Jiangmen Central Hospital, Affiliated Jiangmen Hospital of Sun Yat-Sen University, Jiangmen, 529030, China.,MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, China
| | - Jinni Wu
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, China
| | - Tianqi Cao
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, China
| | - Shengyao Zhi
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, China
| | - Yuxi Chen
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, China
| | - Lars Aagaard
- Department of Biomedicine, Aarhus University, 8000, Aarhus C, Denmark
| | - Peilin Zhen
- Department of Infectious Diseases, Jiangmen Central Hospital, Affiliated Jiangmen Hospital of Sun Yat-Sen University, Jiangmen, 529030, China
| | - Yanming Huang
- Clinical Experimental Center, Jiangmen Central Hospital, Affiliated Jiangmen Hospital of Sun Yat-Sen University, Jiangmen, 529030, China
| | - Jianxin Zhong
- Department of Neurology, Jiangmen Central Hospital, Affiliated Jiangmen Hospital of Sun Yat-Sen University, Jiangmen, 529030, China
| | - Junjiu Huang
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, China. .,Clinical Experimental Center, Jiangmen Central Hospital, Affiliated Jiangmen Hospital of Sun Yat-Sen University, Jiangmen, 529030, China. .,Key Laboratory of Reproductive Medicine of Guangdong Province, First Affiliated Hospital and School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, China.
| |
Collapse
|
4
|
Zettler S, Renner S, Kemter E, Hinrichs A, Klymiuk N, Backman M, Riedel EO, Mueller C, Streckel E, Braun-Reichhart C, Martins AS, Kurome M, Keßler B, Zakhartchenko V, Flenkenthaler F, Arnold GJ, Fröhlich T, Blum H, Blutke A, Wanke R, Wolf E. A decade of experience with genetically tailored pig models for diabetes and metabolic research. Anim Reprod 2020; 17:e20200064. [PMID: 33029223 PMCID: PMC7534555 DOI: 10.1590/1984-3143-ar2020-0064] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2023] Open
Abstract
The global prevalence of diabetes mellitus and other metabolic diseases is rapidly increasing. Animal models play pivotal roles in unravelling disease mechanisms and developing and testing therapeutic strategies. Rodents are the most widely used animal models but may have limitations in their resemblance to human disease mechanisms and phenotypes. Findings in rodent models are consequently often difficult to extrapolate to human clinical trials. To overcome this ‘translational gap’, we and other groups are developing porcine disease models. Pigs share many anatomical and physiological traits with humans and thus hold great promise as translational animal models. Importantly, the toolbox for genetic engineering of pigs is rapidly expanding. Human disease mechanisms and targets can therefore be reproduced in pigs on a molecular level, resulting in precise and predictive porcine (PPP) models. In this short review, we summarize our work on the development of genetically (pre)diabetic pig models and how they have been used to study disease mechanisms and test therapeutic strategies. This includes the generation of reporter pigs for studying beta-cell maturation and physiology. Furthermore, genetically engineered pigs are promising donors of pancreatic islets for xenotransplantation. In summary, genetically tailored pig models have become an important link in the chain of translational diabetes and metabolic research.
Collapse
Affiliation(s)
- Silja Zettler
- Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich, Germany.,Center for Innovative Medical Models, Department of Veterinary Sciences, LMU Munich, Oberschleißheim, Germany.,German Center for Diabetes Research, Neuherberg, Germany
| | - Simone Renner
- Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich, Germany.,Center for Innovative Medical Models, Department of Veterinary Sciences, LMU Munich, Oberschleißheim, Germany.,German Center for Diabetes Research, Neuherberg, Germany
| | - Elisabeth Kemter
- Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich, Germany.,Center for Innovative Medical Models, Department of Veterinary Sciences, LMU Munich, Oberschleißheim, Germany.,German Center for Diabetes Research, Neuherberg, Germany
| | - Arne Hinrichs
- Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich, Germany.,Center for Innovative Medical Models, Department of Veterinary Sciences, LMU Munich, Oberschleißheim, Germany
| | - Nikolai Klymiuk
- Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich, Germany.,Center for Innovative Medical Models, Department of Veterinary Sciences, LMU Munich, Oberschleißheim, Germany
| | - Mattias Backman
- Laboratory for Functional Genome Analysis, Gene Center, LMU Munich, Munich
| | | | - Christiane Mueller
- Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich, Germany.,Center for Innovative Medical Models, Department of Veterinary Sciences, LMU Munich, Oberschleißheim, Germany
| | - Elisabeth Streckel
- Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich, Germany.,Center for Innovative Medical Models, Department of Veterinary Sciences, LMU Munich, Oberschleißheim, Germany
| | - Christina Braun-Reichhart
- Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich, Germany.,Center for Innovative Medical Models, Department of Veterinary Sciences, LMU Munich, Oberschleißheim, Germany
| | - Ana Sofia Martins
- Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich, Germany.,Center for Innovative Medical Models, Department of Veterinary Sciences, LMU Munich, Oberschleißheim, Germany
| | - Mayuko Kurome
- Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich, Germany.,Center for Innovative Medical Models, Department of Veterinary Sciences, LMU Munich, Oberschleißheim, Germany
| | - Barbara Keßler
- Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich, Germany.,Center for Innovative Medical Models, Department of Veterinary Sciences, LMU Munich, Oberschleißheim, Germany
| | - Valeri Zakhartchenko
- Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich, Germany.,Center for Innovative Medical Models, Department of Veterinary Sciences, LMU Munich, Oberschleißheim, Germany
| | | | - Georg Josef Arnold
- Laboratory for Functional Genome Analysis, Gene Center, LMU Munich, Munich
| | - Thomas Fröhlich
- Laboratory for Functional Genome Analysis, Gene Center, LMU Munich, Munich
| | - Helmut Blum
- Laboratory for Functional Genome Analysis, Gene Center, LMU Munich, Munich
| | - Andreas Blutke
- Research Unit Analytical Pathology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Rüdiger Wanke
- Institute of Veterinary Pathology, Center for Clinical Veterinary Medicine, LMU Munich, Munich, Germany
| | - Eckhard Wolf
- Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich, Germany.,Center for Innovative Medical Models, Department of Veterinary Sciences, LMU Munich, Oberschleißheim, Germany.,German Center for Diabetes Research, Neuherberg, Germany.,Laboratory for Functional Genome Analysis, Gene Center, LMU Munich, Munich
| |
Collapse
|
5
|
Lee J, Kim DH, Lee K. Current Approaches and Applications in Avian Genome Editing. Int J Mol Sci 2020; 21:ijms21113937. [PMID: 32486292 PMCID: PMC7312999 DOI: 10.3390/ijms21113937] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 05/28/2020] [Accepted: 05/29/2020] [Indexed: 01/02/2023] Open
Abstract
Advances in genome-editing technologies and sequencing of animal genomes enable researchers to generate genome-edited (GE) livestock as valuable animal models that benefit biological researches and biomedical and agricultural industries. As birds are an important species in biology and agriculture, their genome editing has gained significant interest and is mainly performed by using a primordial germ cell (PGC)-mediated method because pronuclear injection is not practical in the avian species. In this method, PGCs can be isolated, cultured, genetically edited in vitro, and injected into a recipient embryo to produce GE offspring. Recently, a couple of GE quail have been generated by using the newly developed adenovirus-mediated method. Without technically required in vitro procedures of the PGC-mediated method, direct injection of adenovirus into the avian blastoderm in the freshly laid eggs resulted in the production of germ-line chimera and GE offspring. As more approaches are available in avian genome editing, avian research in various fields will progress rapidly. In this review, we describe the development of avian genome editing and scientific and industrial applications of GE avian species.
Collapse
Affiliation(s)
- Joonbum Lee
- Department of Animal Sciences, The Ohio State University, Columbus, OH 43210, USA; (J.L.); (D.-H.K.)
- The Ohio State University Interdisciplinary Human Nutrition Program, The Ohio State University, Columbus, OH 43210, USA
| | - Dong-Hwan Kim
- Department of Animal Sciences, The Ohio State University, Columbus, OH 43210, USA; (J.L.); (D.-H.K.)
| | - Kichoon Lee
- Department of Animal Sciences, The Ohio State University, Columbus, OH 43210, USA; (J.L.); (D.-H.K.)
- The Ohio State University Interdisciplinary Human Nutrition Program, The Ohio State University, Columbus, OH 43210, USA
- Correspondence: ; Tel.: +1-614-688-7963
| |
Collapse
|
6
|
Lee JG, Sung YH, Baek IJ. Generation of genetically-engineered animals using engineered endonucleases. Arch Pharm Res 2018; 41:885-897. [PMID: 29777358 PMCID: PMC6153862 DOI: 10.1007/s12272-018-1037-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2018] [Accepted: 05/01/2018] [Indexed: 02/06/2023]
Abstract
The key to successful drug discovery and development is to find the most suitable animal model of human diseases for the preclinical studies. The recent emergence of engineered endonucleases is allowing for efficient and precise genome editing, which can be used to develop potentially useful animal models for human diseases. In particular, zinc finger nucleases, transcription activator-like effector nucleases, and the clustered regularly interspaced short palindromic repeat systems are revolutionizing the generation of diverse genetically-engineered experimental animals including mice, rats, rabbits, dogs, pigs, and even non-human primates that are commonly used for preclinical studies of the drug discovery. Here, we describe recent advances in engineered endonucleases and their application in various laboratory animals. We also discuss the importance of genome editing in animal models for more closely mimicking human diseases.
Collapse
Affiliation(s)
- Jong Geol Lee
- ConveRgence mEDIcine research cenTer (CREDIT), Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea
- College of Veterinary Medicine, Chungbuk National University, Cheongju, Republic of Korea
| | - Young Hoon Sung
- ConveRgence mEDIcine research cenTer (CREDIT), Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea.
- Department of Convergence Medicine, ConveRgence mEDIcine research cenTer (CREDIT), Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.
| | - In-Jeoung Baek
- ConveRgence mEDIcine research cenTer (CREDIT), Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea.
- Department of Convergence Medicine, ConveRgence mEDIcine research cenTer (CREDIT), Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
7
|
Lu F, Luo C, Li N, Liu Q, Wei Y, Deng H, Wang X, Li X, Jiang J, Deng Y, Shi D. Efficient Generation of Transgenic Buffalos (Bubalus bubalis) by Nuclear Transfer of Fetal Fibroblasts Expressing Enhanced Green Fluorescent Protein. Sci Rep 2018; 8:6967. [PMID: 29725050 PMCID: PMC5934360 DOI: 10.1038/s41598-018-25120-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 04/11/2018] [Indexed: 01/01/2023] Open
Abstract
The possibility of producing transgenic cloned buffalos by nuclear transfer of fetal fibroblasts expressing enhanced green fluorescent protein (EGFP) was explored in this study. When buffalo fetal fibroblasts (BFFs) isolated from a male buffalo fetus were transfected with pEGFP-N1 (EGFP is driven by CMV and Neo is driven by SV-40) by means of electroporation, Lipofectamine-LTX and X-tremeGENE, the transfection efficiency of electroporation (35.5%) was higher than Lipofectamine-LTX (11.7%) and X-tremeGENE (25.4%, P < 0.05). When BFFs were transfected by means of electroporation, more embryos from BFFs transfected with pEGFP-IRES-Neo (EGFP and Neo are driven by promoter of human elongation factor) cleaved and developed to blastocysts (21.6%) compared to BFFs transfected with pEGFP-N1 (16.4%, P < 0.05). A total of 72 blastocysts were transferred into 36 recipients and six recipients became pregnant. In the end of gestation, the pregnant recipients delivered six healthy calves and one stillborn calf. These calves were confirmed to be derived from the transgenic cells by Southern blot and microsatellite analysis. These results indicate that electroporation is more efficient than lipofection in transfecting exogenous DNA into BFFs and transgenic buffalos can be produced effectively by nuclear transfer of BFFs transfected with pEGFP-IRES-Neo.
Collapse
Affiliation(s)
- Fenghua Lu
- Guangxi High Education Key Laboratory for Animal Reproduction and Biotechnology, State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning, 530005, China
| | - Chan Luo
- Guangxi High Education Key Laboratory for Animal Reproduction and Biotechnology, State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning, 530005, China
| | - Nan Li
- Guangxi High Education Key Laboratory for Animal Reproduction and Biotechnology, State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning, 530005, China.,Reproductive Center of Liuzhou Municipal Maternity and Child Healthcare Hospital, Liuzhou, 545001, China
| | - Qingyou Liu
- Guangxi High Education Key Laboratory for Animal Reproduction and Biotechnology, State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning, 530005, China
| | - Yingming Wei
- Guangxi High Education Key Laboratory for Animal Reproduction and Biotechnology, State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning, 530005, China
| | - Haiying Deng
- Guangxi High Education Key Laboratory for Animal Reproduction and Biotechnology, State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning, 530005, China
| | - Xiaoli Wang
- Guangxi High Education Key Laboratory for Animal Reproduction and Biotechnology, State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning, 530005, China
| | - Xiangping Li
- Guangxi High Education Key Laboratory for Animal Reproduction and Biotechnology, State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning, 530005, China
| | - Jianrong Jiang
- Guangxi High Education Key Laboratory for Animal Reproduction and Biotechnology, State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning, 530005, China
| | - Yanfei Deng
- Guangxi High Education Key Laboratory for Animal Reproduction and Biotechnology, State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning, 530005, China
| | - Deshun Shi
- Guangxi High Education Key Laboratory for Animal Reproduction and Biotechnology, State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning, 530005, China.
| |
Collapse
|
8
|
Sekhavati MH, Hosseini SM, Tahmoorespur M, Ghaedi K, Jafarpour F, Hajian M, Dormiani K, Nasr-Esfahani MH. PhiC31-based Site-Specific Transgenesis System for Production of Transgenic Bovine Embryos by Somatic Cell Nuclear Transfer and Intracytoplasmic Sperm Injection. CELL JOURNAL 2018; 20:98-107. [PMID: 29308625 PMCID: PMC5759686 DOI: 10.22074/cellj.2018.4385] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/19/2016] [Accepted: 03/01/2017] [Indexed: 01/10/2023]
Abstract
OBJECTIVES The Streptomyces phage phiC31 integrase offers a sequence-specific method of transgenesis with a robust long-term gene expression. PhiC31 has been successfully developed in a variety of tissues and organs for purpose of in vivo gene therapy. The objective of the present experiment was to evaluate PhiC31-based site-specific transgenesis system for production of transgenic bovine embryos by somatic cell nuclear transfer and intracytoplasmic sperm injection. MATERIALS AND METHODS In this experimental study, the application of phiC31 integrase system was evaluated for generating transgenic bovine embryos by somatic cell nuclear transfer (SCNT) and sperm mediated gene transfer (SMGT) approaches. RESULTS PhiC31 integrase mRNA and protein was produced in vitro and their functionality was confirmed. Seven phiC31 recognizable bovine pseudo attachment sites of phage (attP) sites were considered for evaluation of site specific recombination. The accuracy of these sites was validated in phic31 targeted bovine fibroblasts using polymerase chain reaction (PCR) and sequencing. The efficiency and site-specificity of phiC31 integrase system was also confirmed in generated transgenic bovine embryo which successfully obtained using SCNT and SMGT technique. CONCLUSIONS The results showed that both SMGT and SCNT-derived embryos were enhanced green fluorescent protein (EGFP) positive and phiC31 integrase could recombine the reporter gene in a site specific manner. These results demonstrate that attP site can be used as a proper location to conduct site directed transgenesis in both mammalian cells and embryos in phiC31 integrase system when even combinaed to SCNT and intracytoplasmic sperm injection (ICSI) method.
Collapse
Affiliation(s)
| | - Sayed Morteza Hosseini
- Department of Reproductive Biotechnology, Reproductive Biomedicine Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | | | - Kamran Ghaedi
- Department of Biology, Facualty of Sciences, Uneversity of Isfahan, Isfahan, Iran
- Department of Cellular Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Farnoosh Jafarpour
- Department of Reproductive Biotechnology, Reproductive Biomedicine Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Mehdi Hajian
- Department of Reproductive Biotechnology, Reproductive Biomedicine Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Kyanoosh Dormiani
- Department of Cellular Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Mohammad Hossain Nasr-Esfahani
- Department of Reproductive Biotechnology, Reproductive Biomedicine Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran.
| |
Collapse
|
9
|
Intratesticular injection followed by electroporation allows gene transfer in caprine spermatogenic cells. Sci Rep 2018; 8:3169. [PMID: 29453369 PMCID: PMC5816633 DOI: 10.1038/s41598-018-21558-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 02/07/2018] [Indexed: 01/09/2023] Open
Abstract
The production of transgenic livestock is constrained due to the limited success of currently available methods for transgenesis. Testis mediated gene transfer (TMGT) is an emerging method that shows a high success rate in generating transgenic mice. In this study, we report a newly developed protocol for electroporation-aided TMGT to produce a transgenic goat. The injectable volume and concentration of the transgene were first standardized, and then electroporation conditions were optimized in vitro. In vivo experiments were performed by injecting a transgenic construct (pIRES2-EGFP; enhanced green fluorescent protein) into the testicular interstitium followed by electroporation. Immunohistochemistry, quantitative real-time PCR (qPCR) and western blotting analyses confirmed the successful transfer of the transgene into seminiferous tubules and testicular cells. Furthermore, chromosomal integration of the transgene and its expression in sperm were evaluated d60 and d120 post-electroporation. Our protocol neither altered the seminal characteristics nor the fertilization capacity of the sperm cells. In vitro fertilization using transgenic sperm generated fluorescent embryos. Finally, natural mating of a pre-founder buck produced a transgenic baby goat. The present study demonstrates the first successful report of an electroporation-aided TMGT method for gene transfer in goats.
Collapse
|
10
|
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
|
11
|
Perleberg C, Kind A, Schnieke A. Genetically engineered pigs as models for human disease. Dis Model Mech 2018; 11:11/1/dmm030783. [PMID: 29419487 PMCID: PMC5818075 DOI: 10.1242/dmm.030783] [Citation(s) in RCA: 147] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Genetically modified animals are vital for gaining a proper understanding of disease mechanisms. Mice have long been the mainstay of basic research into a wide variety of diseases but are not always the most suitable means of translating basic knowledge into clinical application. The shortcomings of rodent preclinical studies are widely recognised, and regulatory agencies around the world now require preclinical trial data from nonrodent species. Pigs are well suited to biomedical research, sharing many similarities with humans, including body size, anatomical features, physiology and pathophysiology, and they already play an important role in translational studies. This role is set to increase as advanced genetic techniques simplify the generation of pigs with precisely tailored modifications designed to replicate lesions responsible for human disease. This article provides an overview of the most promising and clinically relevant genetically modified porcine models of human disease for translational biomedical research, including cardiovascular diseases, cancers, diabetes mellitus, Alzheimer's disease, cystic fibrosis and Duchenne muscular dystrophy. We briefly summarise the technologies involved and consider the future impact of recent technical advances. Summary: An overview of porcine models of human disease, including cardiovascular diseases, cancers, diabetes mellitus, Alzheimer's disease, cystic fibrosis and Duchenne muscular dystrophy. We summarise the technologies involved and potential future impact of recent technical advances.
Collapse
Affiliation(s)
- Carolin Perleberg
- Chair of Livestock Biotechnology, School of Life Sciences, Technische Universität München, 85354 Freising, Germany
| | - Alexander Kind
- Chair of Livestock Biotechnology, School of Life Sciences, Technische Universität München, 85354 Freising, Germany
| | - Angelika Schnieke
- Chair of Livestock Biotechnology, School of Life Sciences, Technische Universität München, 85354 Freising, Germany
| |
Collapse
|
12
|
Generation of DKK1 transgenic Tibet minipigs by somatic cell nuclear transfer (SCNT). Oncotarget 2017; 8:74331-74339. [PMID: 29088789 PMCID: PMC5650344 DOI: 10.18632/oncotarget.20604] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Accepted: 08/17/2017] [Indexed: 02/06/2023] Open
Abstract
Hairless mice have been widely applied in skin-related researches, while hairless pigs will be a useful model for skin-related study and other biomedical researches. Dickkopf-related protein 1 (DKK1) is inhibitor of Wnt signaling pathway. Transgenic mice expressing DKK1 transgene under control of a human keratin 14 (K14) promoter display hairless phenotype, which encouraged us to generate transgenic minipigs expressing pig DKK1 transgene under control of K14 promoter and finally achieve hairless minipigs. To generate transgenic cloned pigs, we constructed the lentiviral expression vector pERKDZG which contains two independent expression cassettes, the transcription of Tibet minipig DKK1 and EGFP genes are driven by K14 promoter, while mRFP is regulated under the control of Ef-1α promoter. Prior to generating the transgenic pig, the functionality of pERKDZG construct was verified by fluorescence assay and via checking pDKK1 expression. Subsequently, lentiviruses harboring ERKDZG transgene infected porcine embryonic fibroblasts (PEFs), followed by sorting RFP-positive PEFs by flow cytometry to obtain the purified PEFs carrying ERKDZG, designated DKK1-PEFs as donor cells used for somatic cell nuclear transfer (SCNT). Finally, we obtained 3 DKK1 transgenic cloned pigs with skin-specific expression of pDKK1 and EGFP transgenes, but unfortunately, DKK1 transgenic cloned pigs don't display hairless phenotype as expected. Taken together, we achieve DKK1 transgenic cloned pigs with skin-specific expression of pDKK1 transgene which provide a pig model for exploring DKK1 gene functions in pigs.
Collapse
|
13
|
Telugu BP, Park KE, Park CH. Genome editing and genetic engineering in livestock for advancing agricultural and biomedical applications. Mamm Genome 2017; 28:338-347. [PMID: 28712062 DOI: 10.1007/s00335-017-9709-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2017] [Accepted: 07/08/2017] [Indexed: 01/23/2023]
Abstract
Genetic modification of livestock has a longstanding and successful history, starting with domestication several thousand years ago. Modern animal breeding strategies predominantly based on marker-assisted and genomic selection, artificial insemination, and embryo transfer have led to significant improvement in the performance of domestic animals, and are the basis for regular supply of high quality animal derived food. However, the current strategy of breeding animals over multiple generations to introduce novel traits is not realistic in responding to the unprecedented challenges such as changing climate, pandemic diseases, and feeding an anticipated 3 billion increase in global population in the next three decades. Consequently, sophisticated genetic modifications that allow for seamless introgression of novel alleles or traits and introduction of precise modifications without affecting the overall genetic merit of the animal are required for addressing these pressing challenges. The requirement for precise modifications is especially important in the context of modeling human diseases for the development of therapeutic interventions. The animal science community envisions the genome editors as essential tools in addressing these critical priorities in agriculture and biomedicine, and for advancing livestock genetic engineering for agriculture, biomedical as well as "dual purpose" applications.
Collapse
Affiliation(s)
- Bhanu P Telugu
- Animal and Avian Science, University of Maryland, Bhanu Telugu, 2121 ANSC Building, College Park, MD, 20742, USA. .,Animal Bioscience and Biotechnology Laboratory, ARS, USDA, Beltsville, MD, USA. .,RenOVAte Biosciences Inc, Reisterstown, MD, USA.
| | - Ki-Eun Park
- Animal and Avian Science, University of Maryland, Bhanu Telugu, 2121 ANSC Building, College Park, MD, 20742, USA.,Animal Bioscience and Biotechnology Laboratory, ARS, USDA, Beltsville, MD, USA.,RenOVAte Biosciences Inc, Reisterstown, MD, USA
| | - Chi-Hun Park
- Animal and Avian Science, University of Maryland, Bhanu Telugu, 2121 ANSC Building, College Park, MD, 20742, USA.,Animal Bioscience and Biotechnology Laboratory, ARS, USDA, Beltsville, MD, USA
| |
Collapse
|
14
|
Desaulniers AT, Cederberg RA, Mills GA, Lents CA, White BR. Production of a gonadotropin-releasing hormone 2 receptor knockdown (GNRHR2 KD) swine line. Transgenic Res 2017; 26:567-575. [PMID: 28534229 PMCID: PMC5504211 DOI: 10.1007/s11248-017-0023-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 05/11/2017] [Indexed: 11/25/2022]
Abstract
Swine are the only livestock species that produce both the second mammalian isoform of gonadotropin-releasing hormone (GNRH2) and its receptor (GNRHR2). Previously, we reported that GNRH2 and GNRHR2 mediate LH-independent testosterone secretion from porcine testes. To further explore this ligand-receptor complex, a pig model with reduced GNRHR2 expression was developed. Small hairpin RNA sequences targeting porcine GNRHR2 were subcloned into a lentiviral-based vector, lentiviral particles were generated and microinjected into the perivitelline space of zygotes, and embryos were transferred into a recipient. One GNRHR2 knockdown (KD) female was born that subsequently produced 80 piglets from 6 litters with 46 hemizygous progeny (57% transgenic). Hemizygous GNRHR2 KD (n = 10) and littermate control (n = 7) males were monitored at 40, 100, 150, 190, 225 and 300 days of age; body weight and testis size were measured and serum was isolated and assayed for testosterone and luteinizing hormone (LH) concentrations. Body weight of GNRHR2 KD boars was not different from littermate controls (P = 0.14), but testes were smaller (P < 0.05; 331.8 vs. 374.8 cm3, respectively). Testosterone concentrations tended (P = 0.06) to be reduced in GNRHR2 KD (1.6 ng/ml) compared to littermate control (4.2 ng/ml) males, but LH levels were similar (P = 0.47). The abundance of GNRHR2 mRNA was reduced (P < 0.001) by 69% in testicular tissue from mature GNRHR2 KD (n = 5) versus littermate control (n = 4) animals. These swine represent the first genetically-engineered model to elucidate the function of GNRH2 and its receptor in mammals.
Collapse
Affiliation(s)
- A T Desaulniers
- Department of Animal Science, University of Nebraska-Lincoln, A224j Animal Science Building, 3940 Fair Street, Lincoln, NE, 68583-0908, USA
| | - R A Cederberg
- Department of Animal Science, University of Nebraska-Lincoln, A224j Animal Science Building, 3940 Fair Street, Lincoln, NE, 68583-0908, USA
| | - G A Mills
- Department of Animal Science, University of Nebraska-Lincoln, A224j Animal Science Building, 3940 Fair Street, Lincoln, NE, 68583-0908, USA
| | - C A Lents
- USDA, Agricultural Research Service, U.S. Meat Animal Research Center, Clay Center, NE, 68933-0166, USA
| | - B R White
- Department of Animal Science, University of Nebraska-Lincoln, A224j Animal Science Building, 3940 Fair Street, Lincoln, NE, 68583-0908, USA.
| |
Collapse
|
15
|
Zeng F, Li Z, Cai G, Gao W, Jiang G, Liu D, Urschitz J, Moisyadi S, Wu Z. Characterization of Growth and Reproduction Performance, Transgene Integration, Expression, and Transmission Patterns in Transgenic Pigs Produced by piggyBac Transposition-Mediated Gene Transfer. Anim Biotechnol 2017; 27:245-55. [PMID: 27565868 DOI: 10.1080/10495398.2016.1178140] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Previously we successfully produced a group of EGFP-expressing founder transgenic pigs by a newly developed efficient and simple pig transgenesis method based on cytoplasmic injection of piggyBac plasmids. In this study, we investigated the growth and reproduction performance and characterized the transgene insertion, transmission, and expression patterns in transgenic pigs generated by piggyBac transposition. Results showed that transgene has no injurious effect on the growth and reproduction of transgenic pigs. Multiple copies of monogenic EGFP transgene were inserted at noncoding sequences of host genome, and passed from founder transgenic pigs to their transgenic offspring in segregation or linkage manner. The EGFP transgene was ubiquitously expressed in transgenic pigs, and its expression intensity was associated with transgene copy number but not related to its promoter DNA methylation level. To the best of our knowledge, this is first study that fully described the growth and reproduction performance, transgene insertion, expression, and transmission profiles in transgenic pigs produced by piggyBac system. It not only demonstrates that piggyBac transposition-mediated gene transfer is an effective and favorable approach for pig transgenesis, but also provides scientific information for understanding the transgene insertion, expression and transmission patterns in transgenic animals produced by piggyBac transposition.
Collapse
Affiliation(s)
- Fang Zeng
- a National Engineering Research Center for Breeding Swine Industry, College of Animal Science , South China Agricultural University , Guangzhou , China.,b Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, College of Animal Science , South China Agricultural University , Guangzhou , China
| | - Zicong Li
- a National Engineering Research Center for Breeding Swine Industry, College of Animal Science , South China Agricultural University , Guangzhou , China.,b Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, College of Animal Science , South China Agricultural University , Guangzhou , China
| | - Gengyuan Cai
- c Institute of Animal Science , Guangdong Academy of Agricultural Sciences , Guangzhou , China
| | - Wenchao Gao
- a National Engineering Research Center for Breeding Swine Industry, College of Animal Science , South China Agricultural University , Guangzhou , China.,b Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, College of Animal Science , South China Agricultural University , Guangzhou , China
| | - Gelong Jiang
- a National Engineering Research Center for Breeding Swine Industry, College of Animal Science , South China Agricultural University , Guangzhou , China.,b Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, College of Animal Science , South China Agricultural University , Guangzhou , China
| | - Dewu Liu
- a National Engineering Research Center for Breeding Swine Industry, College of Animal Science , South China Agricultural University , Guangzhou , China.,b Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, College of Animal Science , South China Agricultural University , Guangzhou , China
| | - Johann Urschitz
- d Institute for Biogenesis Research, Department of Anatomy, Biochemistry and Physiology, John A. Burns School of Medicine , University of Hawaii at Manoa , Honolulu , Hawaii , USA
| | - Stefan Moisyadi
- d Institute for Biogenesis Research, Department of Anatomy, Biochemistry and Physiology, John A. Burns School of Medicine , University of Hawaii at Manoa , Honolulu , Hawaii , USA.,e Manoa BioSciences , Honolulu , Hawaii , USA
| | - Zhenfang Wu
- a National Engineering Research Center for Breeding Swine Industry, College of Animal Science , South China Agricultural University , Guangzhou , China.,b Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, College of Animal Science , South China Agricultural University , Guangzhou , China
| |
Collapse
|
16
|
Doran TJ, Cooper CA, Jenkins KA, Tizard MLV. Advances in genetic engineering of the avian genome: "Realising the promise". Transgenic Res 2016; 25:307-19. [PMID: 26820412 DOI: 10.1007/s11248-016-9926-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Accepted: 01/06/2016] [Indexed: 10/22/2022]
Abstract
This review provides an historic perspective of the key steps from those reported at the 1st Transgenic Animal Research Conference in 1997 through to the very latest developments in avian transgenesis. Eighteen years later, on the occasion of the 10th conference in this series, we have seen breakthrough advances in the use of viral vectors and transposons to transform the germline via the direct manipulation of the chicken embryo, through to the establishment of PGC cultures allowing in vitro modification, expansion into populations to analyse the genetic modifications and then injection of these cells into embryos to create germline chimeras. We have now reached an unprecedented time in the history of chicken transgenic research where we have the technology to introduce precise, targeted modifications into the chicken genome, ranging from; new transgenes that provide improved phenotypes such as increased resilience to economically important diseases; the targeted disruption of immunoglobulin genes and replacement with human sequences to generate transgenic chickens that express "humanised" antibodies for biopharming; and the deletion of specific nucleotides to generate targeted gene knockout chickens for functional genomics. The impact of these advances is set to be realised through applications in chickens, and other bird species as models in scientific research, for novel biotechnology and to protect and improve agricultural productivity.
Collapse
Affiliation(s)
- Timothy J Doran
- Australian Animal Health Laboratory, CSIRO Health and Biosecurity, Private Bag 24, Geelong, VIC, 3220, Australia.
| | - Caitlin A Cooper
- Australian Animal Health Laboratory, CSIRO Health and Biosecurity, Private Bag 24, Geelong, VIC, 3220, Australia
| | - Kristie A Jenkins
- Australian Animal Health Laboratory, CSIRO Health and Biosecurity, Private Bag 24, Geelong, VIC, 3220, Australia
| | - Mark L V Tizard
- Australian Animal Health Laboratory, CSIRO Health and Biosecurity, Private Bag 24, Geelong, VIC, 3220, Australia
| |
Collapse
|
17
|
Meng F, Li H, Wang X, Qin G, Oback B, Shi D. Optimized production of transgenic buffalo embryos and offspring by cytoplasmic zygote injection. J Anim Sci Biotechnol 2015; 6:44. [PMID: 26500768 PMCID: PMC4617447 DOI: 10.1186/s40104-015-0044-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Accepted: 09/29/2015] [Indexed: 12/05/2022] Open
Abstract
Background Cytoplasmic injection of exogenous DNA into zygotes is a promising technique to generate transgenic livestock. However, it is still relatively inefficient and has not yet been demonstrated to work in buffalo. We sought to improve two key technical parameters of the procedure, namely i) how much linear DNA to inject and ii) when to inject it. For this, we introduced a constitutively expressed enhanced green fluorescent protein (EGFP) plasmid into buffalo zygotes. Results First, we found that the proportion of EGFP-expressing blastocysts derived from zygotes injected with 20 or 50 ng/μL DNA was significantly higher than from those injected with 5 μg/mL. However, 50 ng/μL exogenous DNA compromised blastocyst development compared to non-injected IVF controls. Therefore the highest net yield of EGFP-positive blastocysts was achieved at 20 ng/μL DNA. Second, zygotes injected early (7–8 h post-insemination [hpi]) developed better than those injected at mid (12–13 hpi) or late (18–19 hpi) time points. Blastocysts derived from early injections were also more frequently EGFP-positive. As a consequence, the net yield of EGFP-expressing blastocysts was more than doubled using early vs late injections (16.4 % vs 7.7 %). With respect to blastocyst quality, we found no significant difference in cell numbers of EGFP-positive blastocysts vs non-injected blastocysts. Following embryo transfer of six EGFP-positive blastocysts into four recipient animals, two viable buffalo calves were born. Biopsied ear tissues from both buffalo calves were analyzed for transgene presence and expression by Southern blot, PCR and confocal laser scanning microscopy, respectively. This confirmed that both calves were transgenic. Conclusions Our cytoplasmic injection protocol improved generation of transgenic embryos and resulted in the first transgenic buffalo calves produced by this method.
Collapse
Affiliation(s)
- Fanli Meng
- Animal Reproduction Institute, State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, Guangxi University, 75 Xiuling Road, Nanning, 530005 P.R China ; Present address: AgResearch Ltd., Ruakura Research Centre, Reproductive Technologies, 10 Bisley Road, Private Bag 3123, Hamilton, New Zealand
| | - Hui Li
- Animal Reproduction Institute, State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, Guangxi University, 75 Xiuling Road, Nanning, 530005 P.R China
| | - Xiaoli Wang
- Animal Reproduction Institute, State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, Guangxi University, 75 Xiuling Road, Nanning, 530005 P.R China
| | - Guangsheng Qin
- Animal Reproduction Institute, State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, Guangxi University, 75 Xiuling Road, Nanning, 530005 P.R China
| | - Björn Oback
- Present address: AgResearch Ltd., Ruakura Research Centre, Reproductive Technologies, 10 Bisley Road, Private Bag 3123, Hamilton, New Zealand
| | - Deshun Shi
- Animal Reproduction Institute, State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, Guangxi University, 75 Xiuling Road, Nanning, 530005 P.R China
| |
Collapse
|
18
|
Watanabe M, Kobayashi M, Nagaya M, Matsunari H, Nakano K, Maehara M, Hayashida G, Takayanagi S, Sakai R, Umeyama K, Watanabe N, Onodera M, Nagashima H. Production of transgenic cloned pigs expressing the far-red fluorescent protein monomeric Plum. J Reprod Dev 2015; 61:169-177. [PMID: 25739316 PMCID: PMC4498373 DOI: 10.1262/jrd.2014-153] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Accepted: 12/31/2014] [Indexed: 12/22/2022] Open
Abstract
Monomeric Plum (Plum), a far-red fluorescent protein with photostability and photopermeability, is potentially suitable for in vivo imaging and detection of fluorescence in body tissues. The aim of this study was to generate transgenic cloned pigs exhibiting systemic expression of Plum using somatic cell nuclear transfer (SCNT) technology. Nuclear donor cells for SCNT were obtained by introducing a Plum-expression vector driven by a combination of the cytomegalovirus early enhancer and chicken beta-actin promoter into porcine fetal fibroblasts (PFFs). The cleavage and blastocyst formation rates of reconstructed SCNT embryos were 81.0% (34/42) and 78.6% (33/42), respectively. At 36-37 days of gestation, three fetuses systemically expressing Plum were obtained from one recipient to which 103 SCNT embryos were transferred (3/103, 2.9%). For generation of offspring expressing Plum, rejuvenated PFFs were established from one cloned fetus and used as nuclear donor cells. Four cloned offspring and one stillborn cloned offspring were produced from one recipient to which 117 SCNT embryos were transferred (5/117, 4.3%). All offspring exhibited high levels of Plum fluorescence in blood cells, such as lymphocytes, monocytes and granulocytes. In addition, the skin, heart, kidney, pancreas, liver and spleen also exhibited Plum expression. These observations demonstrated that transfer of the Plum gene did not interfere with the development of porcine SCNT embryos and resulted in the successful generation of transgenic cloned pigs that systemically expressed Plum. This is the first report of the generation and characterization of transgenic cloned pigs expressing the far-red fluorescent protein Plum.
Collapse
Affiliation(s)
- Masahito Watanabe
- Laboratory of Developmental Engineering, Department of Life Sciences, School of Agriculture, Meiji University, Kanagawa 214-8571, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Abstract
Traditional methods for DNA transfection are often inefficient and toxic for terminally differentiated cells, such as cardiac myocytes. Vector-based gene transfer is an efficient approach for introducing exogenous cDNA into these types of primary cell cultures. In this chapter, separate protocols for adult rat cardiac myocyte isolation and gene transfer with recombinant adenovirus are provided and are routinely utilized for studying the effects of sarcomeric proteins on myofilament function.
Collapse
|
20
|
Wang Y, Zhou XY, Xiang PY, Wang LL, Tang H, Xie F, Li L, Wei H. The meganuclease I-SceI containing nuclear localization signal (NLS-I-SceI) efficiently mediated mammalian germline transgenesis via embryo cytoplasmic microinjection. PLoS One 2014; 9:e108347. [PMID: 25250567 PMCID: PMC4177210 DOI: 10.1371/journal.pone.0108347] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2014] [Accepted: 08/18/2014] [Indexed: 12/23/2022] Open
Abstract
The meganuclease I-SceI has been effectively used to facilitate transgenesis in fish eggs for nearly a decade. I-SceI-mediated transgenesis is simply via embryo cytoplasmic microinjection and only involves plasmid vectors containing I-SceI recognition sequences, therefore regarding the transgenesis process and application of resulted transgenic organisms, I-SceI-mediated transgenesis is of minimal bio-safety concerns. However, currently no transgenic mammals derived from I-SceI-mediated transgenesis have been reported. In this work, we found that the native I-SceI molecule was not capable of facilitating transgenesis in mammalian embryos via cytoplasmic microinjection as it did in fish eggs. In contrast, the I-SceI molecule containing mammalian nuclear localization signal (NLS-I-SceI) was shown to be capable of transferring DNA fragments from cytoplasm into nuclear in porcine embryos, and cytoplasmic microinjection with NLS-I-SceI mRNA and circular I-SceI recognition sequence-containing transgene plasmids resulted in transgene expression in both mouse and porcine embryos. Besides, transfer of the cytoplasmically microinjected mouse and porcine embryos into synchronized recipient females both efficiently resulted in transgenic founders with germline transmission competence. These results provided a novel method to facilitate mammalian transgenesis using I-SceI, and using the NLS-I-SceI molecule, a simple, efficient and species-neutral transgenesis technology based on embryo cytoplasmic microinjection with minimal bio-safety concerns can be established for mammalian species. As far as we know, this is the first report for transgenic mammals derived from I-SceI-mediated transgenesis via embryo cytoplasmic microinjection.
Collapse
Affiliation(s)
- Yong Wang
- Department of Laboratory Animal Science, College of Basic Medical Sciences, Third Military Medical University, Chongqing, China
- * E-mail: (YW); (HW)
| | - Xiao-Yang Zhou
- Department of Laboratory Animal Science, College of Basic Medical Sciences, Third Military Medical University, Chongqing, China
| | - Peng-Ying Xiang
- Department of Laboratory Animal Science, College of Basic Medical Sciences, Third Military Medical University, Chongqing, China
| | - Lu-Lu Wang
- Department of Laboratory Animal Science, College of Basic Medical Sciences, Third Military Medical University, Chongqing, China
| | - Huan Tang
- China Three Gorges Museum, Chongqing, China
| | - Fei Xie
- Department of Laboratory Animal Science, College of Basic Medical Sciences, Third Military Medical University, Chongqing, China
| | - Liang Li
- Department of Laboratory Animal Science, College of Basic Medical Sciences, Third Military Medical University, Chongqing, China
| | - Hong Wei
- Department of Laboratory Animal Science, College of Basic Medical Sciences, Third Military Medical University, Chongqing, China
- * E-mail: (YW); (HW)
| |
Collapse
|
21
|
Embryo development, fetal growth and postnatal phenotype of eGFP lambs generated by lentiviral transgenesis. Transgenic Res 2014; 24:31-41. [PMID: 25048992 DOI: 10.1007/s11248-014-9816-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2014] [Accepted: 07/11/2014] [Indexed: 10/25/2022]
Abstract
Lentiviral technology has been recently proposed to generate transgenic farm animals more efficiently and easier than traditional techniques. The objective was to evaluate several parameters of lambs obtained by lentiviral transgenesis in comparison with non-transgenic counterparts. In vitro produced embryos were microinjected (TG group) at two-cell stage with a lentiviral construct containing enhanced green fluorescent protein (eGFP) gene, while embryos produced by in vitro fertilization (IVF group) or intrauterine insemination (IUI group) were not microinjected. Microinjection technique efficiently generated eight-cell transgenic embryos (97.4%; 114/117). Development rate on day 5 after fertilization was similar for TG (39.3%, 46/117) and IVF embryos (39.6%, 44/111). Pregnancy rate was detected in 50.0% (6/12) of recipient ewes with TG embryos, in 46.7% (7/15) with IVF embryos, and in 65.0% (13/20) of IUI ewes (P = NS). Nine lambs were born in TG group, six lambs in IVF group, and 16 lambs in IUI group. All TG lambs (9/9) were GFP positive to real-time PCR and eight (88.9%) showed a strong and evident GFP expression in mucosae, eyes and keratin tissues. Fetal growth monitored every 15 day by ultrasonography did not show significant differences. Transgenic lambs neither differ in morphometric variables in comparison with non transgenic IVF lambs within 3 months after birth. Transmission of the transgene to the progeny was observed in green fluorescent embryos produced by IVF using semen from the TG founder lambs. In conclusion, this study demonstrates the high efficiency of lentiviral technology to produce transgenic sheep, with no clinic differences in comparison with non transgenic lambs.
Collapse
|
22
|
Li Z, Zeng F, Meng F, Xu Z, Zhang X, Huang X, Tang F, Gao W, Shi J, He X, Liu D, Wang C, Urschitz J, Moisyadi S, Wu Z. Generation of transgenic pigs by cytoplasmic injection of piggyBac transposase-based pmGENIE-3 plasmids. Biol Reprod 2014; 90:93. [PMID: 24671876 DOI: 10.1095/biolreprod.113.116905] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
The process of transgenesis involves the introduction of a foreign gene, the transgene, into the genome of an animal. Gene transfer by pronuclear microinjection (PNI) is the predominant method used to produce transgenic animals. However, this technique does not always result in germline transgenic offspring and has a low success rate for livestock. Alternate approaches, such as somatic cell nuclear transfer using transgenic fibroblasts, do not show an increase in efficiency compared to PNI, while viral-based transgenesis is hampered by issues regarding transgene size and biosafety considerations. We have recently described highly successful transgenesis experiments with mice using a piggyBac transposase-based vector, pmhyGENIE-3. This construct, a single and self-inactivating plasmid, contains all the transpositional elements necessary for successful gene transfer. In this series of experiments, our laboratories have implemented cytoplasmic injection (CTI) of pmGENIE-3 for transgene delivery into in vivo-fertilized pig zygotes. More than 8.00% of the injected embryos developed into transgenic animals containing monogenic and often single transgenes in their genome. However, the CTI technique was unsuccessful during the injection of in vitro-fertilized pig zygotes. In summary, here we have described a method that is not only easy to implement, but also demonstrated the highest efficiency rate for nonviral livestock transgenesis.
Collapse
Affiliation(s)
- Zicong Li
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Takahashi T, Hanazawa K, Inoue T, Sato K, Sedohara A, Okahara J, Suemizu H, Yagihashi C, Yamamoto M, Eto T, Konno Y, Okano H, Suematsu M, Sasaki E. Birth of healthy offspring following ICSI in in vitro-matured common marmoset (Callithrix jacchus) oocytes. PLoS One 2014; 9:e95560. [PMID: 24751978 PMCID: PMC3994092 DOI: 10.1371/journal.pone.0095560] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2013] [Accepted: 03/27/2014] [Indexed: 11/18/2022] Open
Abstract
Intracytoplasmic sperm injection (ICSI), an important method used to treat male subfertility, is applied in the transgenic technology of sperm-mediated gene transfer. However, no study has described successful generation of offspring using ICSI in the common marmoset, a small non-human primate used as a model for biomedical translational research. In this study, we investigated blastocyst development and the subsequent live offspring stages of marmoset oocytes matured in vitro and fertilized by ICSI. To investigate the optimal timing of performing ICSI, corrected immature oocytes were matured in vitro and ICSI was performed at various time points (1–2 h, 2–4 h, 4–6 h, 6–8 h, and 8–10 h after extrusion of the first polar body (PB)). Matured oocytes were then divided randomly into two groups: one was used for in vitro fertilization (IVF) and the other for ICSI. To investigate in vivo development of embryos followed by ICSI, 6-cell- to 8-cell-stage embryos and blastocysts were nonsurgically transferred into recipient marmosets. Although no significant differences were observed in the fertilization rate of blastocysts among ICSI timing after the first PB extrusion, the blastocyst rate at 1–2 h was lowest among groups at 2–4 h, 4–6 h, 6–8 h, and 8–10 h. Comparing ICSI to IVF, the fertilization rates obtained in ICSI were higher than in IVF (p>0.05). No significant difference was noted in the cleaved blastocyst rate between ICSI and IVF. Following the transfer of 37 ICSI blastocysts, 4 of 20 recipients became pregnant, while with the transfer of 21 6-cell- to 8-cell-stage ICSI embryos, 3 of 8 recipients became pregnant. Four healthy offspring were produced and grew normally. These are the first marmoset offspring produced by ICSI, making it an effective fertilization method for marmosets.
Collapse
Affiliation(s)
- Tsukasa Takahashi
- Department of Applied Developmental Biology, Central Institute for Experimental Animals, Kawasaki-ku, Kawasaki, Kanagawa, Japan
- Department of Biochemistry, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Kisaburo Hanazawa
- Department of Oncology, Juntendo University Nerima Hospital, Nerima-ku, Tokyo, Japan
| | - Takashi Inoue
- Marmoset Research Department, Central Institute for Experimental Animals, Kawasaki-ku, Kawasaki, Kanagawa, Japan
| | - Kenya Sato
- Department of Applied Developmental Biology, Central Institute for Experimental Animals, Kawasaki-ku, Kawasaki, Kanagawa, Japan
| | - Ayako Sedohara
- Department of Applied Developmental Biology, Central Institute for Experimental Animals, Kawasaki-ku, Kawasaki, Kanagawa, Japan
| | - Junko Okahara
- Department of Applied Developmental Biology, Central Institute for Experimental Animals, Kawasaki-ku, Kawasaki, Kanagawa, Japan
| | - Hiroshi Suemizu
- Department of Biomedical Research, Central Institute for Experimental Animals, Kawasaki-ku, Kawasaki, Kanagawa, Japan
| | - Chie Yagihashi
- Department of Biomedical Research, Central Institute for Experimental Animals, Kawasaki-ku, Kawasaki, Kanagawa, Japan
| | - Masafumi Yamamoto
- Department of Biomedical Research, Central Institute for Experimental Animals, Kawasaki-ku, Kawasaki, Kanagawa, Japan
| | - Tomoo Eto
- Department of Applied Developmental Biology, Central Institute for Experimental Animals, Kawasaki-ku, Kawasaki, Kanagawa, Japan
| | - Yusuke Konno
- Altair Corporation, Kohoku-ku, Yokohama-shi, Kanagawa, Japan
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Makoto Suematsu
- Department of Biochemistry, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Erika Sasaki
- Department of Applied Developmental Biology, Central Institute for Experimental Animals, Kawasaki-ku, Kawasaki, Kanagawa, Japan
- Keio Advanced Research Center, Keio University, Shinjuku-ku, Tokyo, Japan
- * E-mail:
| |
Collapse
|
24
|
Germline transgenesis in pigs by cytoplasmic microinjection of Sleeping Beauty transposons. Nat Protoc 2014; 9:810-27. [DOI: 10.1038/nprot.2014.010] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
25
|
Larsson HM, Gorostidi F, Hubbell JA, Barrandon Y, Frey P. Clonal, self-renewing and differentiating human and porcine urothelial cells, a novel stem cell population. PLoS One 2014; 9:e90006. [PMID: 24587183 PMCID: PMC3935977 DOI: 10.1371/journal.pone.0090006] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2014] [Accepted: 01/24/2014] [Indexed: 01/01/2023] Open
Abstract
Although urothelial progenitor-like cells have been described in the human urinary tract, the existence of stem cells remains to be proven. Using a culture system that favors clonogenic epithelial cell growth, we evaluated and characterized clonal human urothelial cells. We isolated human urothelial cells that were clonogenic, capable of self-renewal and could develop into fully differentiated urothelium once re-implanted into the subcapsular space of nude mice. In addition to final urothelial cell differentiation, spontaneous formation of bladder-like microstructures was observed. By examining an epithelial stem cell signature marker, we found p63 to correlate with the self-renewal capacity of the isolated human urothelial clonal populations. Since a clinically relevant, long-term model for functional reconstitution of human cells does not exist, we sought to establish a culture method for porcine urothelial cells in a clinically relevant porcine model. We isolated cells from porcine ureter, urethra and bladder that were clonogenic and capable of self-renewal and differentiation into fully mature urothelium. In conclusion, we could isolate human and porcine cell populations, behaving as urothelial stem cells and showing clonogenicity, self-renewal and, once re-implanted, morphological differentiation.
Collapse
Affiliation(s)
- Hans M. Larsson
- Laboratory for Regenerative Medicine and Pharmacobiology, Institute for Bioengineering, School of Life Sciences and School of Engineering, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Francois Gorostidi
- Laboratory of Experimental Surgery, Department of Experimental Surgery, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
- Laboratory of Stem Cell Dynamics, Institute for Bioengineering, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Jeffrey A. Hubbell
- Laboratory for Regenerative Medicine and Pharmacobiology, Institute for Bioengineering, School of Life Sciences and School of Engineering, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Yann Barrandon
- Laboratory of Experimental Surgery, Department of Experimental Surgery, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
- Laboratory of Stem Cell Dynamics, Institute for Bioengineering, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Peter Frey
- Laboratory for Regenerative Medicine and Pharmacobiology, Institute for Bioengineering, School of Life Sciences and School of Engineering, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
- Laboratory of Experimental Pediatric Urology, Department of Pediatric Urology, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
- * E-mail:
| |
Collapse
|
26
|
Genetically engineered pig models for diabetes research. Transgenic Res 2013; 23:27-38. [PMID: 24065178 DOI: 10.1007/s11248-013-9755-y] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2013] [Accepted: 09/13/2013] [Indexed: 12/15/2022]
Abstract
Diabetes mellitus (DM) has emerged into a steadily increasing health problem and the predicted future dimension of the global DM epidemic is alarming: an increase from currently 346 million to over 400 million affected people worldwide by the year 2030 was extrapolated. Thus concerted research efforts are imperative to gain insight into disease mechanisms and to expand the basis for development of preventive and therapeutic strategies. Diabetic rodent models have traditionally been used to follow these goals, but have limitations for translational research. The pig is another classical animal model for diabetes research. Genetic engineering now facilitates tailoring pig models which mimic human disease mechanisms at the molecular level. This article reviews the existing genetically engineered pig models for diabetes research and their current and future applications. Further, the potential role of the pig as donor of pancreatic islets for xenotransplantation or as host for growing human pancreas is outlined.
Collapse
|
27
|
Bi Y, Liu X, Zhang L, Shao C, Ma Z, Hua Z, Zhang L, Li L, Hua W, Xiao H, Wei Q, Zheng X. Pseudo attP sites in favor of transgene integration and expression in cultured porcine cells identified by Streptomyces phage phiC31 integrase. BMC Mol Biol 2013; 14:20. [PMID: 24010979 PMCID: PMC3844521 DOI: 10.1186/1471-2199-14-20] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2013] [Accepted: 08/27/2013] [Indexed: 12/30/2022] Open
Abstract
Phage PhiC31 integrase integrates attB-containing plasmid into pseudo attP site in eukaryotic genomes in a unidirectional site-specific manner and maintains robust transgene expression. Few studies, however, explore its potential in livestock. This study aims to discover the molecular basis of PhiC31 integrase-mediated site-specific recombination in pig cells. We show that PhiC31 integrase can mediate site-specific transgene integration into the genome of pig kidney PK15 cells. Intramolecular recombination in pig PK15 cell line occurred at maximum frequency of 82% with transiently transfected attB- and attP-containing plasmids. An optimal molar ratio of pCMV-Int to pEGFP-N1-attB at 5:1 was observed for maximum number of cell clones under drug selection. Four candidate pseudo attP sites were identified by TAIL-PCR from those cell clones with single-copy transgene integration. Two of them gave rise to higher integration frequency occurred at 33%. 5′ and 3′ junction PCR showed that transgene integration mediated by PhiC31 integrase was mono-allelic. Micro- deletion and insertion were observed by sequencing the integration border, indicating that double strand break was induced by the recombination. We then constructed rescue reporter plasmids by ABI-REC cloning of the four pseudo attP sites into pBCPB + plasmid. Transfection of these rescue plasmids and pCMV-Int resulted in expected intramolecular recombination between attB and pseudo attP sites. This proved that the endogenous pseudo attP sites were functional substrates for PhiC31 integrase-mediated site-specific recombination. Two pseudo attP sites maintained robust extracellular and intracellular EGFP expression. Alamar blue assay showed that transgene integration into these specific sites had little effect on cell proliferation. This is the first report to document the potential use of PhiC31 integrase to mediate site-specific recombination in pig cells. Our work established an ideal model to study the position effect of identical transgene located in diverse chromosomal contexts. These findings also form the basis for targeted pig genome engineering and may be used to produce genetically modified pigs for agricultural and biomedical uses.
Collapse
Affiliation(s)
- Yanzhen Bi
- Hubei Key Laboratory of Animal Embryo Engineering and Molecular Breeding, Institute of Animal Science and Veterinary Medicine, Hubei Academy of Agricultural Science, Wuhan 430064, China.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Li X, Fan P, Jin J, Su W, An D, Xu L, Sun S, Zhang Y, Meng X, Gao F, Kong W, Jiang C. Establishment of cell lines with increased susceptibility to EV71/CA16 by stable overexpression of SCARB2. Virol J 2013; 10:250. [PMID: 23919614 PMCID: PMC3765843 DOI: 10.1186/1743-422x-10-250] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2013] [Accepted: 07/31/2013] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Human enterovirus type 71 (EV71) and Coxsackievirus A group type 16 (CA16) belong to human Enterovirus species A of the family Picornaviridae. These viruses are recognized as the major pathogens responsible for epidemics of hand-foot-mouth disease (HFMD), which presents with fever and vesicular eruptions of palms, soles of the feet or mouth. Human scavenger receptor class B, member 2 (SCARB2) has been identified as the receptor for both EV71 and CA16, as overexpression of SCARB2 in cells can enhance virus replication significantly. METHODS In this study, we used a lentivirus packaging vector to transduce the SCARB2 gene into human embryonic kidney cells (293), human rhabdomyosarcoma cells (RD) and African green monkey kidney cells (Vero) to create stable expression lines. Expression of SCARB2 in the resulting three transgenic cell lines was confirmed by real-time RT-PCR, immunofluorescence and flow cytometry. RESULTS Levels of SCARB2 mRNA determined by real-time RT-PCR in 293-SCARB2 (293S) or RD-SCARB2 (RDS) transgenic cell lines were approximately 2 × 10(2) times higher than those in 293 and RD cells, respectively, and three times higher in Vero-SCARB2 (VeroS) than in Vero cells. Furthermore, EV71 and CA16 virus titers in 293S and RDS cells were 10(2)-10(3)-fold higher (detected in RD cell) than those in the parental cells, and a 10-fold higher titer of EV71 was achieved in VeroS cells compared with that in Vero cells. CONCLUSIONS We established for the first time three cell lines stably overexpressing SCARB2, which showed drastic increases in susceptibility to EV71/CA16 infection. These optimal cell lines may be utilized to develop inactivated vaccines for EV71/CA16 and facilitate rapid detection and isolation of HFMD pathogens or other Enterovirus serotypes. Furthermore, these stable cell lines also can serve as tools to facilitate drug screenings as well as molecular studies on virus-host interactions and pathogenesis of causative agents for HFMD.
Collapse
Affiliation(s)
- Xiaojun Li
- School of Life Sciences, Jilin University, Changchun, P.R. China
- National Engineering Laboratory for AIDS Vaccine, Jilin University, Changchun, P.R. China
| | - Peihun Fan
- School of Life Sciences, Jilin University, Changchun, P.R. China
- National Engineering Laboratory for AIDS Vaccine, Jilin University, Changchun, P.R. China
| | - Jun Jin
- School of Life Sciences, Jilin University, Changchun, P.R. China
- National Engineering Laboratory for AIDS Vaccine, Jilin University, Changchun, P.R. China
| | - Weiheng Su
- School of Life Sciences, Jilin University, Changchun, P.R. China
- National Engineering Laboratory for AIDS Vaccine, Jilin University, Changchun, P.R. China
| | - Dong An
- School of Life Sciences, Jilin University, Changchun, P.R. China
- National Engineering Laboratory for AIDS Vaccine, Jilin University, Changchun, P.R. China
| | - Lin Xu
- School of Life Sciences, Jilin University, Changchun, P.R. China
- National Engineering Laboratory for AIDS Vaccine, Jilin University, Changchun, P.R. China
| | - Shiyang Sun
- School of Life Sciences, Jilin University, Changchun, P.R. China
- National Engineering Laboratory for AIDS Vaccine, Jilin University, Changchun, P.R. China
| | - Yan Zhang
- School of Life Sciences, Jilin University, Changchun, P.R. China
- National Engineering Laboratory for AIDS Vaccine, Jilin University, Changchun, P.R. China
| | - Xiangyu Meng
- School of Life Sciences, Jilin University, Changchun, P.R. China
- National Engineering Laboratory for AIDS Vaccine, Jilin University, Changchun, P.R. China
| | - Feng Gao
- School of Life Sciences, Jilin University, Changchun, P.R. China
- National Engineering Laboratory for AIDS Vaccine, Jilin University, Changchun, P.R. China
- Human Vaccine Institute, Duke University Medical Centre, Durham, NC 27710, USA
| | - Wei Kong
- School of Life Sciences, Jilin University, Changchun, P.R. China
- National Engineering Laboratory for AIDS Vaccine, Jilin University, Changchun, P.R. China
- Key Laboratory for Molecular Enzymology & Engineering, the Ministry of Education, Jilin University, 2699 Qianjin Street, Changchun 130012, P.R. China
| | - Chunlai Jiang
- School of Life Sciences, Jilin University, Changchun, P.R. China
- National Engineering Laboratory for AIDS Vaccine, Jilin University, Changchun, P.R. China
- Key Laboratory for Molecular Enzymology & Engineering, the Ministry of Education, Jilin University, 2699 Qianjin Street, Changchun 130012, P.R. China
| |
Collapse
|
29
|
Miao X. Recent advances in the development of new transgenic animal technology. Cell Mol Life Sci 2013; 70:815-28. [PMID: 22833168 PMCID: PMC11113483 DOI: 10.1007/s00018-012-1081-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2012] [Revised: 06/30/2012] [Accepted: 07/03/2012] [Indexed: 12/14/2022]
Abstract
Transgenic animal technology is one of the fastest growing biotechnology areas. It is used to integrate exogenous genes into the animal genome by genetic engineering technology so that these genes can be inherited and expressed by offspring. The transgenic efficiency and precise control of gene expression are the key limiting factors in the production of transgenic animals. A variety of transgenic technologies are available. Each has its own advantages and disadvantages and needs further study because of unresolved technical and safety issues. Further studies will allow transgenic technology to explore gene function, animal genetic improvement, bioreactors, animal disease models, and organ transplantation. This article reviews the recently developed animal transgenic technologies, including the germ line stem cell-mediated method to improve efficiency, gene targeting to improve accuracy, RNA interference-mediated gene silencing technology, zinc-finger nuclease gene targeting technology and induced pluripotent stem cell technology. These new transgenic techniques can provide a better platform to develop transgenic animals for breeding new animal varieties and promote the development of medical sciences, livestock production, and other fields.
Collapse
Affiliation(s)
- Xiangyang Miao
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193, China.
| |
Collapse
|
30
|
Liu C, Wang L, Li W, Zhang X, Tian Y, Zhang N, He S, Chen T, Huang J, Liu M. Highly efficient generation of transgenic sheep by lentivirus accompanying the alteration of methylation status. PLoS One 2013; 8:e54614. [PMID: 23382924 PMCID: PMC3558511 DOI: 10.1371/journal.pone.0054614] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2012] [Accepted: 12/13/2012] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND Low efficiency of gene transfer and silence of transgene expression are the critical factors hampering the development of transgenic livestock. Recently, transfer of recombinant lentivirus has been demonstrated to be an efficient transgene delivery method in various animals. However, the lentiviral transgenesis and the methylation status of transgene in sheep have not been well addressed. METHODOLOGY/PRINCIPLE FINDINGS EGFP transgenic sheep were generated by injecting recombinant lentivirus into zygotes. Of the 13 lambs born, 8 carried the EGFP transgene, and its chromosomal integration was identified in all tested tissues. Western blotting showed that GFP was expressed in all transgenic founders and their various tissues. Analysis of CpG methylation status of CMV promoter by bisulfate sequencing unraveled remarkable variation of methylation levels in transgenic sheep. The average methylation levels ranged from 37.6% to 79.1% in the transgenic individuals and 34.7% to 83% in the tested tissues. Correlative analysis of methylation status with GFP expression revealed that the GFP expression level was inversely correlated with methylation density. The similar phenomenon was also observed in tested tissues. Transgene integration determined by Southern blotting presented multiple integrants ranging from 2 to 6 copies in the genome of transgenic sheep. CONCLUSIONS/SIGNIFICANCE Injection of lentiviral transgene into zygotes could be a promising efficient gene delivery system to generate transgenic sheep and achieved widespread transgene expression. The promoter of integrants transferred by lentiviral vector was subjected to dramatic alteration of methylation status and the transgene expression level was inversely correlative with promoter methylation density. Our work illustrated for the first time that generation of transgenic sheep by injecting recombinant lentivirus into zygote could be an efficient tool to improve sheep performance by genetic modification.
Collapse
Affiliation(s)
- Chenxi Liu
- Xinjiang Laboratory of Animal Biotechnology, Urumqi, Xinjiang, China
- Key Laboratory of Genetics, Breeding and Reproduction of Grass Feeding Livestock, Ministry of Agriculture, Urumqi, Xinjiang, China
- College of Life Science and Technology, Xinjiang University, Urumqi, Xinjiang, China
| | - Liqin Wang
- Xinjiang Laboratory of Animal Biotechnology, Urumqi, Xinjiang, China
- Key Laboratory of Genetics, Breeding and Reproduction of Grass Feeding Livestock, Ministry of Agriculture, Urumqi, Xinjiang, China
- Animal Biotechnology Research Center, Xinjiang Academy of Animal Science, Urumqi, Xinjiang, China
| | - Wenrong Li
- Xinjiang Laboratory of Animal Biotechnology, Urumqi, Xinjiang, China
- Key Laboratory of Genetics, Breeding and Reproduction of Grass Feeding Livestock, Ministry of Agriculture, Urumqi, Xinjiang, China
- Animal Biotechnology Research Center, Xinjiang Academy of Animal Science, Urumqi, Xinjiang, China
| | - Xuemei Zhang
- Xinjiang Laboratory of Animal Biotechnology, Urumqi, Xinjiang, China
- Key Laboratory of Genetics, Breeding and Reproduction of Grass Feeding Livestock, Ministry of Agriculture, Urumqi, Xinjiang, China
- Animal Biotechnology Research Center, Xinjiang Academy of Animal Science, Urumqi, Xinjiang, China
| | - Yongzhi Tian
- Xinjiang Laboratory of Animal Biotechnology, Urumqi, Xinjiang, China
- Key Laboratory of Genetics, Breeding and Reproduction of Grass Feeding Livestock, Ministry of Agriculture, Urumqi, Xinjiang, China
- Animal Biotechnology Research Center, Xinjiang Academy of Animal Science, Urumqi, Xinjiang, China
| | - Ning Zhang
- Xinjiang Laboratory of Animal Biotechnology, Urumqi, Xinjiang, China
- Key Laboratory of Genetics, Breeding and Reproduction of Grass Feeding Livestock, Ministry of Agriculture, Urumqi, Xinjiang, China
- Animal Biotechnology Research Center, Xinjiang Academy of Animal Science, Urumqi, Xinjiang, China
| | - Sangang He
- Xinjiang Laboratory of Animal Biotechnology, Urumqi, Xinjiang, China
- Key Laboratory of Genetics, Breeding and Reproduction of Grass Feeding Livestock, Ministry of Agriculture, Urumqi, Xinjiang, China
- Animal Biotechnology Research Center, Xinjiang Academy of Animal Science, Urumqi, Xinjiang, China
| | - Tong Chen
- Xinjiang Laboratory of Animal Biotechnology, Urumqi, Xinjiang, China
- Key Laboratory of Genetics, Breeding and Reproduction of Grass Feeding Livestock, Ministry of Agriculture, Urumqi, Xinjiang, China
- Animal Biotechnology Research Center, Xinjiang Academy of Animal Science, Urumqi, Xinjiang, China
| | - Juncheng Huang
- Xinjiang Laboratory of Animal Biotechnology, Urumqi, Xinjiang, China
- Key Laboratory of Genetics, Breeding and Reproduction of Grass Feeding Livestock, Ministry of Agriculture, Urumqi, Xinjiang, China
- Animal Biotechnology Research Center, Xinjiang Academy of Animal Science, Urumqi, Xinjiang, China
| | - Mingjun Liu
- Xinjiang Laboratory of Animal Biotechnology, Urumqi, Xinjiang, China
- Key Laboratory of Genetics, Breeding and Reproduction of Grass Feeding Livestock, Ministry of Agriculture, Urumqi, Xinjiang, China
- Animal Biotechnology Research Center, Xinjiang Academy of Animal Science, Urumqi, Xinjiang, China
- * E-mail:
| |
Collapse
|
31
|
Prather RS, Lorson M, Ross JW, Whyte JJ, Walters E. Genetically engineered pig models for human diseases. Annu Rev Anim Biosci 2013; 1:203-19. [PMID: 25387017 DOI: 10.1146/annurev-animal-031412-103715] [Citation(s) in RCA: 124] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Although pigs are used widely as models of human disease, their utility as models has been enhanced by genetic engineering. Initially, transgenes were added randomly to the genome, but with the application of homologous recombination, zinc finger nucleases, and transcription activator-like effector nuclease (TALEN) technologies, now most any genetic change that can be envisioned can be completed. To date these genetic modifications have resulted in animals that have the potential to provide new insights into human diseases for which a good animal model did not exist previously. These new animal models should provide the preclinical data for treatments that are developed for diseases such as Alzheimer's disease, cystic fibrosis, retinitis pigmentosa, spinal muscular atrophy, diabetes, and organ failure. These new models will help to uncover aspects and treatments of these diseases that were otherwise unattainable. The focus of this review is to describe genetically engineered pigs that have resulted in models of human diseases.
Collapse
Affiliation(s)
- Randall S Prather
- Division of Animal Science, National Swine Resource and Research Center, and
| | | | | | | | | |
Collapse
|
32
|
Walters EM, Wolf E, Whyte JJ, Mao J, Renner S, Nagashima H, Kobayashi E, Zhao J, Wells KD, Critser JK, Riley LK, Prather RS. Completion of the swine genome will simplify the production of swine as a large animal biomedical model. BMC Med Genomics 2012; 5:55. [PMID: 23151353 PMCID: PMC3499190 DOI: 10.1186/1755-8794-5-55] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2011] [Accepted: 10/28/2011] [Indexed: 12/25/2022] Open
Abstract
Background Anatomic and physiological similarities to the human make swine an excellent large animal model for human health and disease. Methods Cloning from a modified somatic cell, which can be determined in cells prior to making the animal, is the only method available for the production of targeted modifications in swine. Results Since some strains of swine are similar in size to humans, technologies that have been developed for swine can be readily adapted to humans and vice versa. Here the importance of swine as a biomedical model, current technologies to produce genetically enhanced swine, current biomedical models, and how the completion of the swine genome will promote swine as a biomedical model are discussed. Conclusions The completion of the swine genome will enhance the continued use and development of swine as models of human health, syndromes and conditions.
Collapse
Affiliation(s)
- Eric M Walters
- National Swine Resource and Research Center, University of Missouri, Columbia, MO 65211, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Hyperactive self-inactivating piggyBac for transposase-enhanced pronuclear microinjection transgenesis. Proc Natl Acad Sci U S A 2012; 109:19184-9. [PMID: 23093669 DOI: 10.1073/pnas.1216473109] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
We have developed a unique method for mouse transgenesis. The transposase-enhanced pronuclear microinjection (PNI) technique described herein uses the hyperactive piggyBac transposase to insert a large transgene into the mouse genome. This procedure increased transgene integration efficiency by fivefold compared with conventional PNI or intracytoplasmic sperm injection-mediated transgenesis. Our data indicate that the transposase-enhanced PNI technique additionally requires fewer embryos to be microinjected than traditional methods to obtain transgenic animals. This transposase-mediated approach is also very efficient for single-cell embryo cytoplasmic injections, offering an easy-to-implement transgenesis method to the scientific community.
Collapse
|
34
|
Garrels W, Holler S, Cleve N, Niemann H, Ivics Z, Kues WA. Assessment of fecundity and germ line transmission in two transgenic pig lines produced by sleeping beauty transposition. Genes (Basel) 2012; 3:615-33. [PMID: 24705079 PMCID: PMC3899982 DOI: 10.3390/genes3040615] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2012] [Revised: 09/10/2012] [Accepted: 09/14/2012] [Indexed: 01/12/2023] Open
Abstract
Recently, we described a simplified injection method for producing transgenic pigs using a non-autonomous Sleeping Beauty transposon system. The founder animals showed ubiquitous expression of the Venus fluorophore in almost all cell types. To assess, whether expression of the reporter fluorophore affects animal welfare or fecundity, we analyzed reproductive parameters of two founder boars, germ line transmission, and organ and cell specific transgene expression in animals of the F1 and F2 generation. Molecular analysis of ejaculated sperm cells suggested three monomeric integrations of the Venus transposon in both founders. To test germ line transmission of the three monomeric transposon integrations, wild-type sows were artificially inseminated. The offspring were nursed to sexual maturity and hemizygous lines were established. A clear segregation of the monomeric transposons following the Mendelian rules was observed in the F1 and F2 offspring. Apparently, almost all somatic cells, as well as oocytes and spermatozoa, expressed the Venus fluorophore at cell-type specific levels. No detrimental effects of Venus expression on animal health or fecundity were found. Importantly, all hemizygous lines expressed the fluorophore in comparable levels, and no case of transgene silencing or variegated expression was found after germ line transmission, suggesting that the insertions occurred at transcriptionally permissive loci. The results show that Sleeping Beauty transposase-catalyzed transposition is a promising approach for stable genetic modification of the pig genome.
Collapse
Affiliation(s)
- Wiebke Garrels
- Friedrich-Loeffler-Institut, Institute of Farm Animal Genetics, Höltystraße 10, 31535 Neustadt, Germany.
| | - Stephanie Holler
- Friedrich-Loeffler-Institut, Institute of Farm Animal Genetics, Höltystraße 10, 31535 Neustadt, Germany.
| | - Nicole Cleve
- Friedrich-Loeffler-Institut, Institute of Farm Animal Genetics, Höltystraße 10, 31535 Neustadt, Germany.
| | - Heiner Niemann
- Friedrich-Loeffler-Institut, Institute of Farm Animal Genetics, Höltystraße 10, 31535 Neustadt, Germany.
| | - Zoltan Ivics
- Paul-Ehrlich-Institute, Paul-Ehrlich-Straße 51-59, 63225 Langen, Germany.
| | - Wilfried A Kues
- Friedrich-Loeffler-Institut, Institute of Farm Animal Genetics, Höltystraße 10, 31535 Neustadt, Germany.
| |
Collapse
|
35
|
Zhang S, Xiong K, Xie Z, Nan W, Liu H, Chen J. Stable silencing of β-lactoglobulin (BLG) gene by lentivirus-mediated RNAi in goat fetal fibroblasts. Genet Mol Biol 2012; 35:680-5. [PMID: 23055809 PMCID: PMC3459420 DOI: 10.1590/s1415-47572012005000042] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2011] [Accepted: 02/04/2012] [Indexed: 12/11/2022] Open
Abstract
β-lactoglobulin (BLG), a dominant allergen in goat milk, is difficult to remove by traditional biochemical methods. Its elimination from goat milk by genetic modification therefore poses a major challenge for modern goat breeders. A shRNA targeting BLG mRNA with high interference efficiency was identified, with which lentiviral vectors were used for mediating stable shRNA interference in goat-fetal fibroblast cells. Apart from high efficiency in the knockdown of BLG expression in these cells, lentivector-mediated RNAi manifested stable integration into the goat genome itself. Consequently, an in vitro model for goat BLG-content control was compiled, and a goat-cell line for accompanying transgenetic goat production created.
Collapse
Affiliation(s)
- Shumin Zhang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, People's Republic of China
| | | | | | | | | | | |
Collapse
|
36
|
Zhang Y, Xi Q, Ding J, Cai W, Meng F, Zhou J, Li H, Jiang Q, Shu G, Wang S, Zhu X, Gao P, Wu Z. Production of transgenic pigs mediated by pseudotyped lentivirus and sperm. PLoS One 2012; 7:e35335. [PMID: 22536374 PMCID: PMC3335058 DOI: 10.1371/journal.pone.0035335] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2011] [Accepted: 03/14/2012] [Indexed: 11/18/2022] Open
Abstract
Sperm-mediated gene transfer can be a very efficient method to produce transgenic pigs, however, the results from different laboratories had not been widely repeated. Genomic integration of transgene by injection of pseudotyped lentivirus to the perivitelline space has been proved to be a reliable route to generate transgenic animals. To test whether transgene in the lentivirus can be delivered by sperm, we studied incubation of pseudotyped lentiviruses and sperm before insemination. After incubation with pig spermatozoa, 62±3 lentiviral particles were detected per 100 sperm cells using quantitative real-time RT-PCR. The association of lentivirus with sperm was further confirmed by electron microscopy. The sperm incubated with lentiviral particles were artificially inseminated into pigs. Of the 59 piglets born from inseminated 5 sows, 6 piglets (10.17%) carried the transgene based on the PCR identification. Foreign gene and EGFP was successfully detected in ear tissue biopsies from two PCR-positive pigs, revealed via in situ hybridization and immunohistochemistry. Offspring of one PCR-positive boar with normal sows showed PCR-positive. Two PCR-positive founders and offsprings of PCR-positive boar were further identified by Southern-blot analysis, out of which the two founders and two offsprings were positive in Southern blotting, strongly indicating integration of foreign gene into genome. The results indicate that incubation of sperm with pseudotyped lentiviruses can incorporated with sperm-mediated gene transfer to produce transgenic pigs with improved efficiency.
Collapse
Affiliation(s)
- Yongliang Zhang
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Qianyun Xi
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
- * E-mail: (QYX); (ZFW)
| | - Jinghua Ding
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Weiguang Cai
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Fanmin Meng
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Junyun Zhou
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Hongyi Li
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Qingyan Jiang
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Gang Shu
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Songbo Wang
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Xiaotong Zhu
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Ping Gao
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Zhenfang Wu
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
- * E-mail: (QYX); (ZFW)
| |
Collapse
|
37
|
Extensive methylation of promoter sequences silences lentiviral transgene expression during stem cell differentiation in vivo. Mol Ther 2012; 20:1014-21. [PMID: 22434137 DOI: 10.1038/mt.2012.46] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Lentiviral vectors (LV) are widely used to stably transfer genes into target cells investigating or treating gene functions. In addition, gene transfer into early murine embryos may be improved to efficiently generate transgenic mice. We applied lentiviral gene transfer to generate a mouse model transgenic for SET binding protein-1 (Setbp1) and enhanced green fluorescent protein (eGFP). Neither transgenic founders nor their vector-positive offspring transcribed or expressed the transgenes. Bisulfite sequencing of the internal spleen focus-forming virus (SFFV) promoter demonstrated extensive methylation of all analyzed CpGs in the transgenic mice. To analyze the impact of Setbp1 on epigenetic silencing, embryonic stem cells (ESC) were differentiated into cardiomyocytes (CM) in vitro. In contrast to human promoters in LV, virally derived promoter sequences were strongly methylated during differentiation, independent of the transgene. Moreover, the commonly used SFFV promoter (SFFVp) was highly methylated with remarkable strength and frequency during hematopoietic differentiation in vivo in LV but less in γ-retroviral (γ-RV) backbones. In summary, we conclude that LV using an internal SFFVp are not suitable to generate transgenic mice or perform constitutive expression studies in differentiating cells. Choosing the appropriate promoter is also crucial to allow stable transgene expression in clinical gene therapy.
Collapse
|
38
|
Welfare assessment in transgenic pigs expressing green fluorescent protein (GFP). Transgenic Res 2011; 21:773-84. [PMID: 22173943 DOI: 10.1007/s11248-011-9571-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2011] [Accepted: 10/12/2011] [Indexed: 10/14/2022]
Abstract
Since large animal transgenesis has been successfully attempted for the first time about 25 years ago, the technology has been applied in various lines of transgenic pigs. Nevertheless one of the concerns with the technology--animal welfare--has not been approached through systematic assessment and statements regarding the welfare of transgenic pigs have been based on anecdotal observations during early stages of transgenic programs. The main aim of the present study was therefore to perform an extensive welfare assessment comparing heterozygous transgenic animals expressing GFP with wildtype animals along various stages of post natal development. The protocol used covered reproductory performance and behaviour in GFP and wildtype sows and general health and development, social behaviour, exploratory behaviour and emotionality in GFP and wildtype littermates from birth until an age of roughly 4 months. The absence of significant differences between GFP and wildtype animals in the parameters observed suggests that the transgenic animals in question are unlikely to suffer from deleterious effects of transgene expression on their welfare and thus support existing anecdotal observations of pigs expressing GFP as healthy. Although the results are not surprising in the light of previous experience, they give a more solid fundament to the evaluation of GFP expression as being relatively non-invasive in pigs. The present study may furthermore serve as starting point for researchers aiming at a systematic characterization of welfare relevant effects in the line of transgenic pigs they are working with.
Collapse
|
39
|
Tessanne K, Golding MC, Long CR, Peoples MD, Hannon G, Westhusin ME. Production of transgenic calves expressing an shRNA targeting myostatin. Mol Reprod Dev 2011; 79:176-85. [PMID: 22139943 PMCID: PMC3288734 DOI: 10.1002/mrd.22007] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2011] [Accepted: 11/14/2011] [Indexed: 11/22/2022]
Abstract
Myostatin (MSTN) is a well-known negative regulator of muscle growth. Animals that possess mutations within this gene display an enhanced muscling phenotype, a desirable agricultural trait. Increased neonatal morbidity is common, however, resulting from complications arising from the birth of offspring with increased fetal muscle mass. The objective of the current research was to generate an attenuated MSTN-null phenotype in a large-animal model using RNA interference to enhance muscle development without the detrimental consequences of an inactivating mutation. To this end, we identified a series of short interfering RNAs that demonstrated effective suppression of MSTN mRNA and protein levels. To produce transgenic offspring capable of stable MSTN suppression in vivo, a recombinant lentiviral vector expressing a short hairpin RNA (shRNA) targeting MSTN for silencing was introduced into bovine fetal fibroblasts. These cells were used as nucleus donors for somatic cell nuclear transfer (SCNT). Twenty blastocysts were transferred into seven recipient cows resulting in five pregnancies. One transgenic calf developed to term, but died following delivery by Caesarean-section. As an alternative strategy, microinjection of recombinant lentiviral particles into the perivitelline space of in vitro-produced bovine zygotes was utilized to produce 40 transgenic blastocysts that were transferred into 14 recipient cows, resulting in 7 pregnancies. Five transgenic calves were produced, of which three expressed the transgene. This is the first report of transgenic livestock produced by direct injection of a recombinant lentivirus, and expressing transgenes encoding shRNAs targeting an endogenous gene (myostatin) for silencing.
Collapse
Affiliation(s)
- K Tessanne
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine, Texas A&M University, College Station, Texas, USA
| | | | | | | | | | | |
Collapse
|
40
|
Garrels W, Holler S, Taylor U, Herrmann D, Struckmann C, Klein S, Barg-Kues B, Nowak-Imialek M, Ehling C, Rath D, Ivics Z, Niemann H, Kues WA. Genotype-independent transmission of transgenic fluorophore protein by boar spermatozoa. PLoS One 2011; 6:e27563. [PMID: 22110672 PMCID: PMC3217978 DOI: 10.1371/journal.pone.0027563] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2011] [Accepted: 10/19/2011] [Indexed: 12/24/2022] Open
Abstract
Recently, we generated transposon-transgenic boars (Sus scrofa), which carry three monomeric copies of a fluorophore marker gene. Amazingly, a ubiquitous fluorophore expression in somatic, as well as in germ cells was found. Here, we characterized the prominent fluorophore load in mature spermatozoa of these animals. Sperm samples were analyzed for general fertility parameters, sorted according to X and Y chromosome-bearing sperm fractions, assessed for potential detrimental effects of the reporter, and used for inseminations into estrous sows. Independent of their genotype, all spermatozoa were uniformly fluorescent with a subcellular compartmentalization of the fluorophore protein in postacrosomal sheath, mid piece and tail. Transmission of the fluorophore protein to fertilized oocytes was shown by confocal microscopic analysis of zygotes. The monomeric copies of the transgene segregated during meiosis, rendering a certain fraction of the spermatozoa non-transgenic (about 10% based on analysis of 74 F1 offspring). The genotype-independent transmission of the fluorophore protein by spermatozoa to oocytes represents a non-genetic contribution to the mammalian embryo.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Detlef Rath
- Friedrich-Loeffler-Institut, Mariensee, Germany
| | - Zoltán Ivics
- Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | | | | |
Collapse
|
41
|
Skin-specifically transgenic expression of biologically active human cytoxic T-lymphocyte associated antigen4-immunoglobulin (hCTLA4Ig) in mice using lentiviral vector. Transgenic Res 2011; 21:579-91. [PMID: 21983813 DOI: 10.1007/s11248-011-9559-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2011] [Accepted: 09/14/2011] [Indexed: 02/07/2023]
Abstract
Xenogeneic skin, especially porcine skin, has already been used to cover large wounds in clinic practice of wound care. Our previous data showed that transgenic expression of human cytoxic T-lymphocyte associated antigen4-immunoglobulin (hCTLA4Ig) in murine skin graft remarkably prolonged its survival in xenogeneic burn wounds without extensive immunosuppression in recipients, suggesting that transgenic hCTLA4Ig expression in skin graft may be an effective and safe method to prolong its survival in xenogeneic wounds for coverage. Lentiviral transgenesis provides an extremely efficient and cost-effective method to produce transgenic animals. However, tissue-targeted transgenic expression of biologically functional protein by lentiviral transgenesis is rarely reported. In this work, a recombinant lentiviral vector (LV), named FKCW in this article, was constructed by inserting a skin-specific hCTLA4Ig expression cassette consisting of keratin 14 (K14) promoter, hCTLA4Ig coding sequence and an intronic fragment. Its efficacy for transgenesis and skin-specific expression of bio-active hCTLA4Ig protein was tested using mice as models. The LV FKCW was readily to be packaged and concentrated to high titres (1.287-6.254 × 10(9) TU/ml) by conventional lentivirus package system. Using eggs collected from only five mated females having been subjected to conventional super-ovulation treatment, 8 hCTLA4Ig transgenic founder mice were generated with the concentrated FKCW vector, and transgenic founder per injected and transferred egg was 6.3%, which was nearly 9-fold higher than that for DNA micro-injection with a similar transgene construct in our previous work. The lentiviral transgenic hCTLA4Ig exhibited strictly skin-specific expression at a level comparable to or even slightly higher than that of transgenic hCTLA4Ig delivered by micro-injection in a similar cassette. Lentiviral transgenic hCTLA4Ig protein remarkably suppressed human lymphocyte proliferation in vitro to a degree comparable to that of commercially purchased purified hCTLA4Ig protein with defined activity at similar concentrations. Besides, lentiviral hCTLA4Ig transgenic mouse skin grafted into rat burn wounds exhibited remarkably extended survival compared to wild-type skin of the same strain (13.8 ± 3.8 vs. 6.8 ± 3.0 days), indicating that lentiviral transgenic hCTLA4Ig did inhibit immune rejection against xenogeneic skin graft in vivo. These results laid down the foundation to further efficiently generate transgenic pigs skin-specifically expressing bio-active hCTLA4Ig by lentiviral transgenesis, and provided a demonstration that transgenic animals with tissue-targeted expression of biologically functional protein can be efficiently produced using LV.
Collapse
|
42
|
Whyte JJ, Prather RS. Genetic modifications of pigs for medicine and agriculture. Mol Reprod Dev 2011; 78:879-91. [PMID: 21671302 PMCID: PMC3522184 DOI: 10.1002/mrd.21333] [Citation(s) in RCA: 142] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2011] [Accepted: 05/09/2011] [Indexed: 12/18/2022]
Abstract
Genetically modified swine hold great promise in the fields of agriculture and medicine. Currently, these swine are being used to optimize production of quality meat, to improve our understanding of the biology of disease resistance, and to reduced waste. In the field of biomedicine, swine are anatomically and physiologically analogous to humans. Alterations of key swine genes in disease pathways provide model animals to improve our understanding of the causes and potential treatments of many human genetic disorders. The completed sequencing of the swine genome will significantly enhance the specificity of genetic modifications, and allow for more accurate representations of human disease based on syntenic genes between the two species. Improvements in both methods of gene alteration and efficiency of model animal production are key to enabling routine use of these swine models in medicine and agriculture.
Collapse
Affiliation(s)
- Jeffrey J. Whyte
- National Swine Resource and Research Center, University of Missouri, Columbia, MO, U.S.A
- Department of Biomedical Sciences, University of Missouri, Columbia, MO, U.S.A
- Division of Animal Science, University of Missouri, Columbia, MO, U.S.A
| | - Randall S. Prather
- National Swine Resource and Research Center, University of Missouri, Columbia, MO, U.S.A
- Division of Animal Science, University of Missouri, Columbia, MO, U.S.A
| |
Collapse
|
43
|
Garrels W, Mátés L, Holler S, Dalda A, Taylor U, Petersen B, Niemann H, Izsvák Z, Ivics Z, Kues WA. Germline transgenic pigs by Sleeping Beauty transposition in porcine zygotes and targeted integration in the pig genome. PLoS One 2011; 6:e23573. [PMID: 21897845 PMCID: PMC3163581 DOI: 10.1371/journal.pone.0023573] [Citation(s) in RCA: 99] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2011] [Accepted: 07/20/2011] [Indexed: 12/21/2022] Open
Abstract
Genetic engineering can expand the utility of pigs for modeling human diseases, and for developing advanced therapeutic approaches. However, the inefficient production of transgenic pigs represents a technological bottleneck. Here, we assessed the hyperactive Sleeping Beauty (SB100X) transposon system for enzyme-catalyzed transgene integration into the embryonic porcine genome. The components of the transposon vector system were microinjected as circular plasmids into the cytoplasm of porcine zygotes, resulting in high frequencies of transgenic fetuses and piglets. The transgenic animals showed normal development and persistent reporter gene expression for >12 months. Molecular hallmarks of transposition were confirmed by analysis of 25 genomic insertion sites. We demonstrate germ-line transmission, segregation of individual transposons, and continued, copy number-dependent transgene expression in F1-offspring. In addition, we demonstrate target-selected gene insertion into transposon-tagged genomic loci by Cre-loxP-based cassette exchange in somatic cells followed by nuclear transfer. Transposase-catalyzed transgenesis in a large mammalian species expands the arsenal of transgenic technologies for use in domestic animals and will facilitate the development of large animal models for human diseases.
Collapse
Affiliation(s)
- Wiebke Garrels
- Institut für Nutztiergenetik, Friedrich-Loeffler-Institut, Neustadt, Germany
| | - Lajos Mátés
- Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Stephanie Holler
- Institut für Nutztiergenetik, Friedrich-Loeffler-Institut, Neustadt, Germany
| | - Anna Dalda
- Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Ulrike Taylor
- Institut für Nutztiergenetik, Friedrich-Loeffler-Institut, Neustadt, Germany
| | - Björn Petersen
- Institut für Nutztiergenetik, Friedrich-Loeffler-Institut, Neustadt, Germany
| | - Heiner Niemann
- Institut für Nutztiergenetik, Friedrich-Loeffler-Institut, Neustadt, Germany
| | - Zsuzsanna Izsvák
- Max Delbrück Center for Molecular Medicine, Berlin, Germany
- University of Debrecen, Debrecen, Hungary
| | - Zoltán Ivics
- Max Delbrück Center for Molecular Medicine, Berlin, Germany
- University of Debrecen, Debrecen, Hungary
- * E-mail: (WAK); (ZI)
| | - Wilfried A. Kues
- Institut für Nutztiergenetik, Friedrich-Loeffler-Institut, Neustadt, Germany
- * E-mail: (WAK); (ZI)
| |
Collapse
|
44
|
Carlson DF, Garbe JR, Tan W, Martin MJ, Dobrinsky JR, Hackett PB, Clark KJ, Fahrenkrug SC. Strategies for selection marker-free swine transgenesis using the Sleeping Beauty transposon system. Transgenic Res 2011; 20:1125-37. [PMID: 21221779 DOI: 10.1007/s11248-010-9481-7] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2010] [Accepted: 12/22/2010] [Indexed: 12/11/2022]
Abstract
Swine transgenesis by pronuclear injection or cloning has traditionally relied on illegitimate recombination of DNA into the pig genome. This often results in animals containing concatemeric arrays of transgenes that complicate characterization and can impair long-term transgene stability and expression. This is inconsistent with regulatory guidance for transgenic livestock, which also discourages the use of selection markers, particularly antibiotic resistance genes. We demonstrate that the Sleeping Beauty (SB) transposon system effectively delivers monomeric, multi-copy transgenes to the pig embryo genome by pronuclear injection without markers, as well as to donor cells for founder generation by cloning. Here we show that our method of transposon-mediated transgenesis yielded 38 cloned founder pigs that altogether harbored 100 integrants for five distinct transposons encoding either human APOBEC3G or YFP-Cre. Two strategies were employed to facilitate elimination of antibiotic genes from transgenic pigs, one based on Cre-recombinase and the other by segregation of independently transposed transgenes upon breeding.
Collapse
Affiliation(s)
- Daniel F Carlson
- The Center for Genome Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Sakuma T, De Ravin SS, Tonne JM, Thatava T, Ohmine S, Takeuchi Y, Malech HL, Ikeda Y. Characterization of retroviral and lentiviral vectors pseudotyped with xenotropic murine leukemia virus-related virus envelope glycoprotein. Hum Gene Ther 2010; 21:1665-73. [PMID: 20507233 PMCID: PMC2999575 DOI: 10.1089/hum.2010.063] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2010] [Accepted: 05/26/2010] [Indexed: 12/18/2022] Open
Abstract
Retroviral and lentiviral vectors are effective gene delivery vehicles that are being evaluated in clinical trials. Variations in the viral envelope (Env) glycoproteins, which are used to pseudotype retroviral or lentiviral vectors, can alter vector performance, including stability, titers, host range, and tissue tropism. Xenotropic murine leukemia virus (MLV)-related virus (XMRV) is a novel human retrovirus identified in patients with prostate cancer. XMRV targets XPR1 cell surface receptor, which is expressed in a broad range of human tissues including hematopoietic stem cells. Pseudotyping with XMRV Env would allow targeting of XPR1-expressing tissues. Here, we characterized XMRV Env-pseudotyped retroviral and lentiviral vectors. Although HIV and MLV vectors were poorly pseudotyped with wild-type XMRV Env, replacement of the C-terminal 11 amino acid residues in the transmembrane domain of XMRV Env with the corresponding 6 amino acid residues of amphotropic MLV Env (XMRV/R(ampho)) significantly increased XMRV Env-pseudotyped HIV and MLV vector titers. The transduction efficiency in human CD34(+) cells when using the XMRV/R(ampho)-pseudotyped HIV vector (10-20%) was comparable to that achieved when using the same infectious units of vesicular stomatitis virus G glycoprotein-pseudotyped vector (25%); thus the modified XMRV Env offers an alternative pseudotyping strategy for XPR1-mediated gene delivery.
Collapse
Affiliation(s)
- Toshie Sakuma
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905
| | - Suk See De Ravin
- Genetic Immunotherapy Section, Laboratory of Host Defenses, National Institute of Allergy and Infectious Diseases, National Institute of Health, Bethesda, MD 20892
| | - Jason M. Tonne
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905
| | | | - Seiga Ohmine
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905
| | - Yasuhiro Takeuchi
- Division of Infection and Immunity, Wohl Virion Centre, Windeyer Institute, University College London, London W1T 4JF, UK
| | - Harry L. Malech
- Genetic Immunotherapy Section, Laboratory of Host Defenses, National Institute of Allergy and Infectious Diseases, National Institute of Health, Bethesda, MD 20892
| | - Yasuhiro Ikeda
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905
| |
Collapse
|
46
|
Jakobsen JE, Li J, Kragh PM, Moldt B, Lin L, Liu Y, Schmidt M, Winther KD, Schyth BD, Holm IE, Vajta G, Bolund L, Callesen H, Jørgensen AL, Nielsen AL, Mikkelsen JG. Pig transgenesis by Sleeping Beauty DNA transposition. Transgenic Res 2010; 20:533-45. [PMID: 20803249 DOI: 10.1007/s11248-010-9438-x] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2010] [Accepted: 08/11/2010] [Indexed: 01/24/2023]
Abstract
Modelling of human disease in genetically engineered pigs provides unique possibilities in biomedical research and in studies of disease intervention. Establishment of methodologies that allow efficient gene insertion by non-viral gene carriers is an important step towards development of new disease models. In this report, we present transgenic pigs created by Sleeping Beauty DNA transposition in primary porcine fibroblasts in combination with somatic cell nuclear transfer by handmade cloning. Göttingen minipigs expressing green fluorescent protein are produced by transgenesis with DNA transposon vectors carrying the transgene driven by the human ubiquitin C promoter. These animals carry multiple copies (from 8 to 13) of the transgene and show systemic transgene expression. Transgene-expressing pigs carry both transposase-catalyzed insertions and at least one copy of randomly inserted plasmid DNA. Our findings illustrate critical issues related to DNA transposon-directed transgenesis, including coincidental plasmid insertion and relatively low Sleeping Beauty transposition activity in porcine fibroblasts, but also provide a platform for future development of porcine disease models using the Sleeping Beauty gene insertion technology.
Collapse
Affiliation(s)
- Jannik E Jakobsen
- Department of Human Genetics, University of Aarhus, Wilh. Meyers Allé 1240, 8000, Aarhus C, Denmark
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Miao K, Guo M, An L, Xu XL, Wu H, Wang D, Wu ZH, Tian JH. A new method to efficiently produce transgenic embryos and mice from low-titer lentiviral vectors. Transgenic Res 2010; 20:357-63. [PMID: 20585977 DOI: 10.1007/s11248-010-9414-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2010] [Accepted: 05/25/2010] [Indexed: 11/30/2022]
Abstract
Vector injection into the perivitelline space has emerged as the standard delivery method to transduce lentivirus to mammalian oocytes or one-cell embryos, but its application is limited by the need for high titers of lentivirus. Herein we developed a new method by using a Piezo impact micro-manipulator for injecting low titer of lentivirus into the subzonal space of two-cell embryos or the perivitelline space of one-cell embryos that were shrunk with a highly concentrated sucrose solution. The survival rate of embryos was greater than 98% using this micromanipulation strategy, which was increased compared to the normal one-cell embryo injection method. More than 90% of injected embryos were GFP positive after subzonal injection of a lentivirus vector carrying the GFP gene with titers of 2 × 10⁸ I.U./ml. Even when a low titer of lentivirus (2 × 10⁶ I.U./ml) was used, 53.26% and 40.85% transgenic embryos were obtained after two-cell embryonic injection and one-cell sucrose treated embryonic injection, respectively. The GFP-positive rates were also greater than in the conventional method of injecting one-cell embryos (25.39%). In addition, blastocysts from the two-cell embryo injection group displayed stronger GFP fluorescence than the one-cell embryo injection groups treated with or without the sucrose solution. Increased expression of GFP suggests that the embryos obtained from this injection method have higher exogenous gene expression levels compared to previous methods. Therefore, in contrast with the traditional injection method, we have demonstrated a simplified and efficient lentivirus-mediated gene transfer method based on a low-titer virus preparation.
Collapse
Affiliation(s)
- Kai Miao
- Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Sciences and Technology, China Agricultural University, 100193, Beijing, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Renner S, Fehlings C, Herbach N, Hofmann A, von Waldthausen DC, Kessler B, Ulrichs K, Chodnevskaja I, Moskalenko V, Amselgruber W, Göke B, Pfeifer A, Wanke R, Wolf E. Glucose intolerance and reduced proliferation of pancreatic beta-cells in transgenic pigs with impaired glucose-dependent insulinotropic polypeptide function. Diabetes 2010; 59:1228-38. [PMID: 20185813 PMCID: PMC2857903 DOI: 10.2337/db09-0519] [Citation(s) in RCA: 138] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
OBJECTIVE The insulinotropic action of the incretin glucose-dependent insulinotropic polypeptide (GIP) is impaired in type 2 diabetes, while the effect of glucagon-like peptide-1 (GLP-1) is preserved. To evaluate the role of impaired GIP function in glucose homeostasis and development of the endocrine pancreas in a large animal model, we generated transgenic pigs expressing a dominant-negative GIP receptor (GIPR(dn)) in pancreatic islets. RESEARCH DESIGN AND METHODS GIPR(dn) transgenic pigs were generated using lentiviral transgenesis. Metabolic tests and quantitative stereological analyses of the different endocrine islet cell populations were performed, and beta-cell proliferation and apoptosis were quantified to characterize this novel animal model. RESULTS Eleven-week-old GIPR(dn) transgenic pigs exhibited significantly reduced oral glucose tolerance due to delayed insulin secretion, whereas intravenous glucose tolerance and pancreatic beta-cell mass were not different from controls. The insulinotropic effect of GIP was significantly reduced, whereas insulin secretion in response to the GLP-1 receptor agonist exendin-4 was enhanced in GIPR(dn) transgenic versus control pigs. With increasing age, glucose control deteriorated in GIPR(dn) transgenic pigs, as shown by reduced oral and intravenous glucose tolerance due to impaired insulin secretion. Importantly, beta-cell proliferation was reduced by 60% in 11-week-old GIPR(dn) transgenic pigs, leading to a reduction of beta-cell mass by 35% and 58% in 5-month-old and 1- to 1.4-year-old transgenic pigs compared with age-matched controls, respectively. CONCLUSIONS The first large animal model with impaired incretin function demonstrates an essential role of GIP for insulin secretion, proliferation of beta-cells, and physiological expansion of beta-cell mass.
Collapse
Affiliation(s)
- Simone Renner
- Chair for Molecular Animal Breeding and Biotechnology and Laboratory for Functional Genome Analysis, Gene Center, Ludwig Maximilians University (LMU) Munich, Munich, Germany
| | - Christiane Fehlings
- Chair for Molecular Animal Breeding and Biotechnology and Laboratory for Functional Genome Analysis, Gene Center, Ludwig Maximilians University (LMU) Munich, Munich, Germany
| | - Nadja Herbach
- Institute of Veterinary Pathology, Faculty of Veterinary Medicine, LMU Munich, Munich, Germany
| | - Andreas Hofmann
- Institute of Pharmacology and Toxicology, University of Bonn, Bonn, Germany
| | - Dagmar C. von Waldthausen
- Chair for Molecular Animal Breeding and Biotechnology and Laboratory for Functional Genome Analysis, Gene Center, Ludwig Maximilians University (LMU) Munich, Munich, Germany
| | - Barbara Kessler
- Chair for Molecular Animal Breeding and Biotechnology and Laboratory for Functional Genome Analysis, Gene Center, Ludwig Maximilians University (LMU) Munich, Munich, Germany
| | - Karin Ulrichs
- Department of Experimental Transplantation Immunology, Surgical Clinic I, University Hospital of Würzburg, Würzburg, Germany
| | - Irina Chodnevskaja
- Department of Experimental Transplantation Immunology, Surgical Clinic I, University Hospital of Würzburg, Würzburg, Germany
| | - Vasiliy Moskalenko
- Department of Experimental Transplantation Immunology, Surgical Clinic I, University Hospital of Würzburg, Würzburg, Germany
| | - Werner Amselgruber
- Institute of Anatomy and Physiology, University of Stuttgart-Hohenheim, Stuttgart, Germany
| | - Burkhard Göke
- Medical Clinic II, Klinikum Grosshadern, LMU Munich, Munich, Germany
| | - Alexander Pfeifer
- Institute of Pharmacology and Toxicology, University of Bonn, Bonn, Germany
- Pharma Center Bonn, University of Bonn, Bonn, Germany
| | - Rüdiger Wanke
- Institute of Veterinary Pathology, Faculty of Veterinary Medicine, LMU Munich, Munich, Germany
| | - Eckhard Wolf
- Chair for Molecular Animal Breeding and Biotechnology and Laboratory for Functional Genome Analysis, Gene Center, Ludwig Maximilians University (LMU) Munich, Munich, Germany
- Corresponding author: Eckhard Wolf,
| |
Collapse
|
49
|
Carlson DF, Geurts AM, Garbe JR, Park CW, Rangel-Filho A, O'Grady SM, Jacob HJ, Steer CJ, Largaespada DA, Fahrenkrug SC. Efficient mammalian germline transgenesis by cis-enhanced Sleeping Beauty transposition. Transgenic Res 2010; 20:29-45. [PMID: 20352328 DOI: 10.1007/s11248-010-9386-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2009] [Accepted: 03/11/2010] [Indexed: 01/18/2023]
Abstract
Heightened interest in relevant models for human disease increases the need for improved methods for germline transgenesis. We describe a significant improvement in the creation of transgenic laboratory mice and rats by chemical modification of Sleeping Beauty transposons. Germline transgenesis in mice and rats was significantly enhanced by in vitro cytosine-phosphodiester-guanine methylation of transposons prior to injection. Heritability of transgene alleles was also greater from founder mice generated with methylated versus non-methylated transposon. The artificial methylation was reprogrammed in the early embryo, leading to founders that express the transgenes. We also noted differences in transgene insertion number and structure (single-insert versus concatemer) based on the influence of methylation and plasmid conformation (linear versus supercoiled), with supercoiled substrate resulting in efficient transpositional transgenesis (TnT) with near elimination of concatemer insertion. Combined, these substrate modifications resulted in increases in both the frequency of transgenic founders and the number of transgenes per founder, significantly elevating the number of potential transgenic lines. Given its simplicity, versatility and high efficiency, TnT with enhanced Sleeping Beauty components represents a compelling non-viral approach to modifying the mammalian germline.
Collapse
|
50
|
Transgenic pigs as models for translational biomedical research. J Mol Med (Berl) 2010; 88:653-64. [PMID: 20339830 DOI: 10.1007/s00109-010-0610-9] [Citation(s) in RCA: 192] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2009] [Revised: 02/26/2010] [Accepted: 03/02/2010] [Indexed: 12/23/2022]
Abstract
The translation of novel discoveries from basic research to clinical application is a long, often inefficient, and thus costly process. Accordingly, the process of drug development requires optimization both for economic and for ethical reasons, in order to provide patients with appropriate treatments in a reasonable time frame. Consequently, "Translational Medicine" became a top priority in national and international roadmaps of human health research. Appropriate animal models for the evaluation of efficacy and safety of new drugs or therapeutic concepts are critical for the success of translational research. In this context rodent models are most widely used. At present, transgenic pigs are increasingly being established as large animal models for selected human diseases. The first pig whole genome sequence and many other genomic resources will be available in the near future. Importantly, efficient and precise techniques for the genetic modification of pigs have been established, facilitating the generation of tailored disease models. This article provides an overview of the current techniques for genetic modification of pigs and the transgenic pig models established for neurodegenerative diseases, cardiovascular diseases, cystic fibrosis, and diabetes mellitus.
Collapse
|