101
|
Su X, Wang S, Su G, Zheng Z, Zhang J, Ma Y, Liu Z, Zhou H, Zhang Y, Zhang L. Production of microhomologous-mediated site-specific integrated LacS gene cow using TALENs. Theriogenology 2018; 119:282-288. [PMID: 30075414 DOI: 10.1016/j.theriogenology.2018.07.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Revised: 07/13/2018] [Accepted: 07/14/2018] [Indexed: 12/31/2022]
Abstract
Gene editing tools (Zinc-Finger Nucleases, ZFN; Transcription Activator-Like Effector Nucleases, TALEN; and Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)/CRISPR-associated (Cas)9, CRISPR-Cas9) provide us with a powerful means of performing genetic engineering procedures. A combinational approach that utilizes both somatic cell nuclear transfer (SCNT) and somatic cell gene editing facilitates the generation of genetically engineered animals. However, the associated research has utilized markers and/or selected genes, which constitute a potential threat to biosafety. Microhomologous-mediated end-joining (MMEJ) has showed the utilization of micro-homologous arms (5-25 bp) can mediate exogenous gene insertion. Dairy milk is a major source of nutrition worldwide. However, most people are not capable of optimally utilizing the nutrition in milk because of lactose intolerance. Sulfolobus solfataricus β-glycosidase (LacS) is a lactase derived from the extreme thermophilic archaeon Sulfolobus solfataricus. Our finally aim was to site-specific integrated LacS gene into cow's genome through TALEN-mediated MMEJ and produce low-lactose cow. Firstly, we constructed TALENs vectors which target to the cow's β-casein locus and LacS gene expression vector which contain TALEN reorganization sequence and micro-homologous arms. Then we co-transfected these vectors into fetal derived skin fibroblasts and cultured as monoclone. Positive cell clones were screened using 3' junction PCR amplification and sequencing analysis. The positive cells were used as donors for SCNT and embryo transfer (ET). Lastly, we detected the genotype through PCR of blood genomic DNA. This resulted in a LacS knock-in rate of 0.8% in TALEN-treated cattle fetal fibroblasts. The blastocyst rate of SCNT embryo was 27%. The 3 months pregnancy rate was 20%. Finally, we obtained 1 newborn cow (5%) and verified its genotype. We obtained 1 site-specific marker-free LacS transgenic cow. It provides a basis to solve lactose intolerance by gene engineering breeding. This study also provides us with a new strategy to facilitate gene knock-ins in livestock using techniques that exhibit improved biosafety and intuitive methodologies.
Collapse
Affiliation(s)
- Xiaohu Su
- Key Laboratory of Gene Engineering of the Ministry of Education, Guangzhou Key Laboratory of Healthy Aging Research and State Key Laboratory of Biocontrol, SYSU-BCM JointResearch Center, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, PR China
| | - Shenyuan Wang
- Key Laboratory of Biological Manufacturing of Inner Mongolia Autonomous Region, College of Life Sciences, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia Autonomous Region, 010018, PR China
| | - Guanghua Su
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Inner Mongolia University, Hohhot, Inner Mongolia Autonomous Region, 010018, PR China
| | - Zhong Zheng
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Inner Mongolia University, Hohhot, Inner Mongolia Autonomous Region, 010018, PR China
| | - Jiaqi Zhang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Yunlong Ma
- Key Laboratory of Biological Manufacturing of Inner Mongolia Autonomous Region, College of Life Sciences, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia Autonomous Region, 010018, PR China
| | - Zongzheng Liu
- Qingdao Animal Husbandry and Veterinary Research Institution, Qingdao, ShanDong, 266100, PR China
| | - Huanmin Zhou
- Key Laboratory of Biological Manufacturing of Inner Mongolia Autonomous Region, College of Life Sciences, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia Autonomous Region, 010018, PR China
| | - Yanru Zhang
- Key Laboratory of Biological Manufacturing of Inner Mongolia Autonomous Region, College of Life Sciences, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia Autonomous Region, 010018, PR China.
| | - Li Zhang
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Inner Mongolia University, Hohhot, Inner Mongolia Autonomous Region, 010018, PR China.
| |
Collapse
|
102
|
Sato M, Ohtsuka M, Nakamura S, Sakurai T, Watanabe S, Gurumurthy CB. In vivo genome editing targeted towards the female reproductive system. Arch Pharm Res 2018; 41:898-910. [PMID: 29974342 DOI: 10.1007/s12272-018-1053-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 06/27/2018] [Indexed: 11/30/2022]
Abstract
The discovery of sequence specific nucleases such as ZFNs, TALENs, and CRISPR/Cas9 has revolutionized genome editing. The CRISPR/Cas9 system has particularly emerged as a highly simple and efficient approach towards generating genome-edited animal models of most of the experimental species. The limitation of these novel genome editing tools is that, till date, they depend on traditional pronuclear injection (PI)-based transgenic technologies developed over the last three decades. PI requires expensive micromanipulator systems and the equipment operators must possess a high level of skill. Therefore, since the establishment of PI-based transgenesis, various research groups worldwide have attempted to develop alternative and simple gene delivery methods. However, owing to the failure of chromosomal integration of the transgene, none of these methods gained the level of confidence as that by the PI method in order to be adapted as a routine approach. The recently developed genome editing systems do not require complicated techniques. Therefore, presently, attention is being focused on non-PI-based gene delivery into germ cells for simple and rapid production of genetically engineered animals. For example, a few reports during the previous 1-2 years demonstrated the use of electroporation (EP) in isolated zygotes that helped to overcome the absolute dependency on PI techniques. Recently, another breakthrough technology called genome editing via oviductal nucleic acids delivery (GONAD) that directly delivers nucleic acids into zygotes within the oviducts in situ was developed. This technology completely relieves the bottlenecks of animal transgenesis as it does not require PI and ex vivo handling of embryos. This review discusses in detail the in vivo gene delivery methods targeted towards female reproductive tissues as these methods that have been developed over the past 2-3 decades can now be re-evaluated for their suitability to deliver the CRISPR/Cas9 components to produce transgenic animals. This review also provides an overview of the latest advances in CRISPR-enabled delivery technologies that have caused paradigm shifts in animal transgenesis methodologies.
Collapse
Affiliation(s)
- Masahiro Sato
- Section of Gene Expression Regulation, Frontier Science Research Center, Kagoshima University, Kagoshima, 890-8544, Japan.
| | - Masato Ohtsuka
- Division of Basic Medical Science and Molecular Medicine, School of Medicine, Tokai University, Kanagawa, 259-1193, Japan.,Center for Matrix Biology and Medicine, Graduate School of Medicine, Tokai University, Kanagawa, 259-1193, Japan.,The Institute of Medical Sciences, Tokai University, Kanagawa, 259-1193, Japan
| | - Shingo Nakamura
- Division of Biomedical Engineering, National Defense Medical College Research Institute, Saitama, 359-8513, Japan
| | - Takayuki Sakurai
- Department of Cardiovascular Research, Graduate School of Medicine, Shinshu University, Nagano, 390-8621, Japan.,Basic Research Division for Next-Generation Disease Models and Fundamental Technology, Research Center for Next Generation Medicine, Shinshu University, Nagano, 390-8621, Japan
| | - Satoshi Watanabe
- Animal Genome Research Unit, Division of Animal Science, National Institute of Agrobiological Sciences, Ibaraki, 305-8602, Japan
| | - Channabasavaiah B Gurumurthy
- Mouse Genome Engineering Core Facility, University of Nebraska Medical Center, Omaha, NE, 68198, USA.,Developmental Neuroscience, Munro Meyer Institute for Genetics and Rehabilitation, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| |
Collapse
|
103
|
Bogdanove AJ, Bohm A, Miller JC, Morgan RD, Stoddard BL. Engineering altered protein-DNA recognition specificity. Nucleic Acids Res 2018; 46:4845-4871. [PMID: 29718463 PMCID: PMC6007267 DOI: 10.1093/nar/gky289] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 04/03/2018] [Accepted: 04/06/2018] [Indexed: 02/07/2023] Open
Abstract
Protein engineering is used to generate novel protein folds and assemblages, to impart new properties and functions onto existing proteins, and to enhance our understanding of principles that govern protein structure. While such approaches can be employed to reprogram protein-protein interactions, modifying protein-DNA interactions is more difficult. This may be related to the structural features of protein-DNA interfaces, which display more charged groups, directional hydrogen bonds, ordered solvent molecules and counterions than comparable protein interfaces. Nevertheless, progress has been made in the redesign of protein-DNA specificity, much of it driven by the development of engineered enzymes for genome modification. Here, we summarize the creation of novel DNA specificities for zinc finger proteins, meganucleases, TAL effectors, recombinases and restriction endonucleases. The ease of re-engineering each system is related both to the modularity of the protein and the extent to which the proteins have evolved to be capable of readily modifying their recognition specificities in response to natural selection. The development of engineered DNA binding proteins that display an ideal combination of activity, specificity, deliverability, and outcomes is not a fully solved problem, however each of the current platforms offers unique advantages, offset by behaviors and properties requiring further study and development.
Collapse
Affiliation(s)
- Adam J Bogdanove
- Plant Pathology and Plant-Microbe Biology Section, School of Integrative Plant Science, Cornell University, Ithaca, NY 14853, USA
| | - Andrew Bohm
- Sackler School of Graduate Biomedical Sciences, Tufts University, 136 Harrison Avenue, Boston, MA 02111, USA
| | - Jeffrey C Miller
- Sangamo Therapeutics Inc. 501 Canal Blvd., Richmond, CA 94804, USA
| | - Richard D Morgan
- New England Biolabs, Inc., 240 County Road, Ipswich, MA 01938, USA
| | - Barry L Stoddard
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave. N., Seattle, WA 98019, USA
| |
Collapse
|
104
|
Banerjee TD, Monteiro A. CRISPR-Cas9 Mediated Genome Editing in Bicyclus anynana Butterflies. Methods Protoc 2018; 1:E16. [PMID: 31164559 PMCID: PMC6526417 DOI: 10.3390/mps1020016] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 05/04/2018] [Accepted: 05/04/2018] [Indexed: 01/19/2023] Open
Abstract
CRISPR-Cas9 is revolutionizing the field of genome editing in non-model organisms. The robustness, ease of use, replicability and affordability of the technology has resulted in its widespread adoption among researchers. The African butterfly Bicyclus anynana is an emerging model lepidopteran species in the field of evo-devo, with a sequenced genome and amenable to germ line transformation. However, efficient genome editing tools to accelerate the pace of functional genetic research in this species have only recently become available with CRISPR-Cas9 technology. Here, we provide a detailed explanation of the CRISPR-Cas9 protocol we follow in the lab. The technique has been successfully implemented to knock-out genes associated with eyespot development and melanin pigmentation.
Collapse
Affiliation(s)
- Tirtha Das Banerjee
- Department of Biological Sciences, National University of Singapore, 14 Science Drive 4, Singapore 117543, Singapore.
| | - Antónia Monteiro
- Department of Biological Sciences, National University of Singapore, 14 Science Drive 4, Singapore 117543, Singapore.
- Yale-NUS College, 10 College Avenue West, Singapore 138609, Singapore.
| |
Collapse
|
105
|
Liang P, Zhang X, Chen Y, Huang J. Developmental history and application of CRISPR in human disease. J Gene Med 2018. [PMID: 28623876 DOI: 10.1002/jgm.2963] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Genome-editing tools are programmable artificial nucleases, mainly including zinc-finger nucleases, transcription activator-like effector nucleases and clustered regularly interspaced short palindromic repeat (CRISPR). By recognizing and cleaving specific DNA sequences, genome-editing tools make it possible to generate site-specific DNA double-strand breaks (DSBs) in the genome. DSBs will then be repaired by either error-prone nonhomologous end joining or high-fidelity homologous recombination mechanisms. Through these two different mechanisms, endogenous genes can be knocked out or precisely repaired/modified. Rapid developments in genome-editing tools, especially CRISPR, have revolutionized human disease models generation, for example, various zebrafish, mouse, rat, pig, monkey and human cell lines have been constructed. Here, we review the developmental history of CRISPR and its application in studies of human diseases. In addition, we also briefly discussed the therapeutic application of CRISPR in the near future.
Collapse
Affiliation(s)
- Puping Liang
- Key Laboratory of Gene Engineering of the Ministry of Education and State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China.,Key Laboratory of Reproductive Medicine of G uangdong Province, The Third Affiliated Hospital, Guangzhou Medical University and School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Xiya Zhang
- Key Laboratory of Gene Engineering of the Ministry of Education and State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Yuxi Chen
- Key Laboratory of Gene Engineering of the Ministry of Education and State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Junjiu Huang
- Key Laboratory of Gene Engineering of the Ministry of Education and State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China.,Key Laboratory of Reproductive Medicine of G uangdong Province, The Third Affiliated Hospital, Guangzhou Medical University and School of Life Sciences, Sun Yat-sen University, Guangzhou, China.,State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
106
|
Gentsch GE, Spruce T, Monteiro RS, Owens NDL, Martin SR, Smith JC. Innate Immune Response and Off-Target Mis-splicing Are Common Morpholino-Induced Side Effects in Xenopus. Dev Cell 2018; 44:597-610.e10. [PMID: 29478923 PMCID: PMC5861998 DOI: 10.1016/j.devcel.2018.01.022] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Revised: 11/21/2017] [Accepted: 01/26/2018] [Indexed: 12/20/2022]
Abstract
Antisense morpholino oligomers (MOs) have been indispensable tools for developmental biologists to transiently knock down (KD) genes rather than to knock them out (KO). Here we report on the implications of genetic KO versus MO-mediated KD of the mesoderm-specifying Brachyury paralogs in the frog Xenopus tropicalis. While both KO and KD embryos fail to activate the same core gene regulatory network, resulting in virtually identical morphological defects, embryos injected with control or target MOs also show a systemic GC content-dependent immune response and many off-target splicing defects. Optimization of MO dosage and increasing incubation temperatures can mitigate, but not eliminate, these MO side effects, which are consistent with the high affinity measured between MO and off-target sequence in vitro. We conclude that while MOs can be useful to profile loss-of-function phenotypes at a molecular level, careful attention must be paid to their immunogenic and off-target side effects.
Collapse
Affiliation(s)
- George E Gentsch
- The Francis Crick Institute, Developmental Biology Laboratory, 1 Midland Road, London NW1 1AT, UK.
| | - Thomas Spruce
- The Francis Crick Institute, Developmental Biology Laboratory, 1 Midland Road, London NW1 1AT, UK
| | - Rita S Monteiro
- The Francis Crick Institute, Developmental Biology Laboratory, 1 Midland Road, London NW1 1AT, UK
| | - Nick D L Owens
- The Francis Crick Institute, Developmental Biology Laboratory, 1 Midland Road, London NW1 1AT, UK
| | - Stephen R Martin
- The Francis Crick Institute, Structural Biology Science Technology Platform, 1 Midland Road, London NW1 1AT, UK
| | - James C Smith
- The Francis Crick Institute, Developmental Biology Laboratory, 1 Midland Road, London NW1 1AT, UK.
| |
Collapse
|
107
|
Petersen B. Basics of genome editing technology and its application in livestock species. Reprod Domest Anim 2018; 52 Suppl 3:4-13. [PMID: 28815851 DOI: 10.1111/rda.13012] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
In the last decade, the research community has witnessed a blooming of targeted genome editing tools and applications. Novel programmable DNA nucleases such as zinc finger nucleases (ZFNs), transcription activator-like endonucleases (TALENs) and the clustered regularly interspaced short palindromic repeats/Cas9 system (CRISPR/Cas9) possess long recognition sites and are capable of cutting DNA in a very specific manner. These DNA nucleases mediate targeted genetic alterations by enhancing the DNA mutation rate via induction of double-strand breaks at a predetermined genomic site. Compared to conventional homologous recombination-based gene targeting, DNA nucleases, also referred to as Genome Editors (GEs), can increase the targeting rate around 10,000- to 100,000-fold. The successful application of different GEs has been shown in a myriad of different organisms, including insects, amphibians, plants, nematodes and several mammalian species, including human cells and embryos. In contrast to all other DNA nucleases, that rely on protein-DNA binding, CRISPR/Cas9 uses RNA to establish a specific binding of its DNA nuclease. Besides its capability to facilitate multiplexed genomic modifications in one shot, the CRISPR/Cas is much easier to design compared to all other DNA nucleases. Current results indicate that any DNA nuclease can be successfully employed in a broad range of organisms which renders them useful for improving the understanding of complex physiological systems such as reproduction, producing transgenic animals, including creating large animal models for human diseases, creating specific cell lines, and plants, and even for treating human genetic diseases. This review provides an update on DNA nucleases, their underlying mechanism and focuses on their application to edit the genome of livestock species.
Collapse
Affiliation(s)
- Bjoern Petersen
- Friedrich-Loeffler-Institut, Institute of Farm Animal Genetics, Neustadt am Rbge, Germany
| |
Collapse
|
108
|
Fan HC, Chi CS, Lee YJ, Tsai JD, Lin SZ, Harn HJ. The Role of Gene Editing in Neurodegenerative Diseases. Cell Transplant 2018; 27:364-378. [PMID: 29766738 PMCID: PMC6038035 DOI: 10.1177/0963689717753378] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Revised: 01/22/2017] [Accepted: 02/19/2017] [Indexed: 12/26/2022] Open
Abstract
Neurodegenerative diseases (NDs), at least including Alzheimer's, Huntington's, and Parkinson's diseases, have become the most dreaded maladies because there are no precise diagnostic tools or definite treatments for these debilitating diseases. The increased prevalence and a substantial impact on the social-economic and medical care of NDs propel governments to develop policies to counteract the impact. Although the etiologies of NDs are still unknown, growing evidence suggests that genetic, cellular, and circuit alternations may cause the generation of abnormal misfolded proteins, which uncontrolledly accumulate to damage and eventually overwhelm the protein-disposal mechanisms of these neurons, leading to a common pathological feature of NDs. If the functions and the connectivity can be restored, alterations and accumulated damages may improve. The gene-editing tools including zinc-finger nucleases (ZFNs), transcription activator-like effector nucleases (TALENs), and clustered regularly interspaced short palindromic repeats-associated nucleases (CRISPR/CAS) have emerged as a novel tool not only for generating specific ND animal models for interrogating the mechanisms and screening potential drugs against NDs but also for the editing sequence-specific genes to help patients with NDs to regain function and connectivity. This review introduces the clinical manifestations of three distinct NDs and the applications of the gene-editing technology on these debilitating diseases.
Collapse
Affiliation(s)
- Hueng-Chuen Fan
- Department of Pediatrics, Tungs’ Taichung Metroharbor Hospital, Taichung, Taiwan
- Department of Medical Research, Tungs’ Taichung Metroharbor Hospital, Taichung, Taiwan
- Department of Rehabilitation, Jen-Teh Junior College of Medicine, Nursing and Management, Miaoli, Taiwan
| | - Ching-Shiang Chi
- Department of Pediatrics, Tungs’ Taichung Metroharbor Hospital, Taichung, Taiwan
| | - Yih-Jing Lee
- School of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Jeng-Dau Tsai
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Pediatrics, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Shinn-Zong Lin
- Bioinnovation Center, Tzu Chi Foundation, Department of Neurosurgery, Buddhist Tzu Chi General Hospital, Tzu Chi University, Hualien, Taiwan
| | - Horng-Jyh Harn
- Bioinnovation Center, Tzu Chi Foundation, Department of Pathology, Buddhist Tzu Chi General Hospital, Tzu Chi University, Hualien, Taiwan
| |
Collapse
|
109
|
Insight into the molecular mechanism of miR-192 regulating Escherichia coli resistance in piglets. Biosci Rep 2018; 38:BSR20171160. [PMID: 29363554 PMCID: PMC5821941 DOI: 10.1042/bsr20171160] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Revised: 12/31/2017] [Accepted: 01/23/2018] [Indexed: 11/21/2022] Open
Abstract
MicroRNAs (miRNAs) have important roles in many cellular processes, including cell proliferation, growth and development, and disease control. Previous study demonstrated that the expression of two highly homologous miRNAs (miR-192 and miR-215) was up-regulated in weaned piglets with Escherichia coli F18 infection. However, the potential molecular mechanism of miR-192 in regulating E. coli infection remains unclear in pigs. In the present study, we analyzed the relationship between level of miR-192 and degree of E. coli resistance using transcription activator-like effector nuclease (TALEN), in vitro bacterial adhesion assays, and target genes research. A TALEN expression vector that specifically recognizes the pig miR-192 was constructed and then monoclonal epithelial cells defective in miR-192 were established. We found that miR-192 knockout led to enhance the adhesion ability of the E. coli strains F18ab, F18ac and K88ac, meanwhile increase the expression of target genes (DLG5 and ALCAM) by qPCR and Western blotting analysis. The results suggested that miR-192 and its key target genes (DLG5 and ALCAM) could have a key role in E. coli infection. Based on our findings, we propose that further investigation of miR-192 function is likely to lead to insights into the molecular mechanisms of E. coli infection.
Collapse
|
110
|
Tang L, Bondareva A, González R, Rodriguez-Sosa JR, Carlson DF, Webster D, Fahrenkrug S, Dobrinski I. TALEN-mediated gene targeting in porcine spermatogonia. Mol Reprod Dev 2018; 85:250-261. [PMID: 29393557 DOI: 10.1002/mrd.22961] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 01/22/2018] [Accepted: 01/25/2018] [Indexed: 01/05/2023]
Abstract
Spermatogonia represent a diploid germ cell population that includes spermatogonial stem cells. In this report, we describe new methods for isolation of highly enriched porcine spermatogonia based on light scatter properties, and for targeted mutagenesis in porcine spermatogonia using nucleofection and TALENs. We optimized a nucleofection protocol to deliver TALENs specifically targeting the DMD locus in porcine spermatogonia. We also validated specific sorting of porcine spermatogonia based on light scatter properties. We were able to obtain a highly enriched germ cell population with over 90% of cells being UCH-L1 positive undifferentiated spermatogonia. After gene targeting in porcine spermatogonia, indel (insertion or deletion) mutations as a result of non-homologous end joining (NHEJ) were detected in up to 18% of transfected cells. Our report demonstrates for the first time an approach to obtain a live cell population highly enriched in undifferentiated spermatogonia from immature porcine testes, and that gene targeting can be achieved in porcine spermatogonia which will enable germ line modification.
Collapse
Affiliation(s)
- Lin Tang
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, Canada
| | - Alla Bondareva
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, Canada
| | - Raquel González
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, Canada
| | - Jose R Rodriguez-Sosa
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, Canada
| | | | | | | | - Ina Dobrinski
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, Canada
| |
Collapse
|
111
|
Use of gene-editing technology to introduce targeted modifications in pigs. J Anim Sci Biotechnol 2018; 9:5. [PMID: 29423214 PMCID: PMC5787920 DOI: 10.1186/s40104-017-0228-7] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Accepted: 12/22/2017] [Indexed: 01/06/2023] Open
Abstract
Pigs are an important resource in agriculture and serve as a model for human diseases. Due to their physiological and anatomical similarities with humans, pigs can recapitulate symptoms of human diseases, making them a useful model in biomedicine. However, in the past pig models have not been widely used partially because of the difficulty in genetic modification. The lack of true embryonic stem cells in pigs forced researchers to utilize genetic modification in somatic cells and somatic cell nuclear transfer (SCNT) to generate genetically engineered (GE) pigs carrying site-specific modifications. Although possible, this approach is extremely inefficient and GE pigs born through this method often presented developmental defects associated with the cloning process. Advancement in the gene-editing systems such as Zinc-Finger Nucleases (ZFNs), Transcription activator-like effector nucleases (TALENs), and the Clustered regularly interspaced short palindromic repeat (CRISPR)/CRISPR-associated 9 (Cas9) system have dramatically increased the efficiency of producing GE pigs. These gene-editing systems, specifically engineered endonucleases, are based on inducing double-stranded breaks (DSBs) at a specific location, and then site-specific modifications can be introduced through one of the two DNA repair pathways: non-homologous end joining (NHEJ) or homology direct repair (HDR). Random insertions or deletions (indels) can be introduced through NHEJ and specific nucleotide sequences can be introduced through HDR, if donor DNA is provided. Use of these engineered endonucleases provides a higher success in genetic modifications, multiallelic modification of the genome, and an opportunity to introduce site-specific modifications during embryogenesis, thus bypassing the need of SCNT in GE pig production. This review will provide a historical prospective of GE pig production and examples of how the gene-editing system, led by engineered endonucleases, have improved GE pig production. We will also present some of our current progress related to the optimal use of CRISPR/Cas9 system during embryogenesis.
Collapse
|
112
|
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: 117] [Impact Index Per Article: 19.5] [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
|
113
|
Veil M, Schaechtle MA, Gao M, Kirner V, Buryanova L, Grethen R, Onichtchouk D. Maternal Nanog is required for zebrafish embryo architecture and for cell viability during gastrulation. Development 2018; 145:dev.155366. [PMID: 29180568 DOI: 10.1242/dev.155366] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 11/07/2017] [Indexed: 12/22/2022]
Abstract
Nanog has been implicated in establishment of pluripotency in mammals and in zygotic genome activation in zebrafish. In this study, we characterize the development of MZnanog (maternal and zygotic null) mutant zebrafish embryos. Without functional Nanog, epiboly is severely affected, embryo axes do not form and massive cell death starts at the end of gastrulation. We show that three independent defects in MZnanog mutants contribute to epiboly failure: yolk microtubule organization required for epiboly is abnormal, maternal mRNA fails to degrade owing to the absence of miR-430, and actin structure of the yolk syncytial layer does not form properly. We further demonstrate that the cell death in MZnanog embryos is cell-autonomous. Nanog is necessary for correct spatial expression of the ventral-specifying genes bmp2b, vox and vent, and the neural transcription factor her3 It is also required for the correctly timed activation of endoderm genes and for the degradation of maternal eomesa mRNA via miR-430. Our findings suggest that maternal Nanog coordinates several gene regulatory networks that shape the embryo during gastrulation.
Collapse
Affiliation(s)
- Marina Veil
- Developmental Biology, Institute Biology I, Faculty of Biology, Albert Ludwigs University of Freiburg, 79104 Freiburg, Germany
| | - Melanie Anna Schaechtle
- Developmental Biology, Institute Biology I, Faculty of Biology, Albert Ludwigs University of Freiburg, 79104 Freiburg, Germany.,BIOSS Centre for Biological Signalling Studies, Albert Ludwigs University of Freiburg, 79104 Freiburg, Germany
| | - Meijiang Gao
- Developmental Biology, Institute Biology I, Faculty of Biology, Albert Ludwigs University of Freiburg, 79104 Freiburg, Germany.,BIOSS Centre for Biological Signalling Studies, Albert Ludwigs University of Freiburg, 79104 Freiburg, Germany
| | - Viola Kirner
- Developmental Biology, Institute Biology I, Faculty of Biology, Albert Ludwigs University of Freiburg, 79104 Freiburg, Germany
| | - Lenka Buryanova
- Developmental Biology, Institute Biology I, Faculty of Biology, Albert Ludwigs University of Freiburg, 79104 Freiburg, Germany
| | - Rachel Grethen
- Developmental Biology, Institute Biology I, Faculty of Biology, Albert Ludwigs University of Freiburg, 79104 Freiburg, Germany
| | - Daria Onichtchouk
- Developmental Biology, Institute Biology I, Faculty of Biology, Albert Ludwigs University of Freiburg, 79104 Freiburg, Germany .,BIOSS Centre for Biological Signalling Studies, Albert Ludwigs University of Freiburg, 79104 Freiburg, Germany.,Institute of Developmental Biology RAS, 119991 Moscow, Russia
| |
Collapse
|
114
|
Khan MHU, Khan SU, Muhammad A, Hu L, Yang Y, Fan C. Induced mutation and epigenetics modification in plants for crop improvement by targeting CRISPR/Cas9 technology. J Cell Physiol 2018; 233:4578-4594. [PMID: 29194606 DOI: 10.1002/jcp.26299] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 11/28/2017] [Indexed: 12/26/2022]
Abstract
Clustered regularly interspaced palindromic repeats associated protein Cas9 (CRISPR-Cas9), originally an adaptive immunity system of prokaryotes, is revolutionizing genome editing technologies with minimal off-targets in the present era. The CRISPR/Cas9 is now highly emergent, advanced, and highly specific tool for genome engineering. The technology is widely used to animal and plant genomes to achieve desirable results. The present review will encompass how CRISPR-Cas9 is revealing its beneficial role in characterizing plant genetic functions, genomic rearrangement, how it advances the site-specific mutagenesis, and epigenetics modification in plants to improve the yield of field crops with minimal side-effects. The possible pitfalls of using and designing CRISPR-Cas9 for plant genome editing are also discussed for its more appropriate applications in plant biology. Therefore, CRISPR/Cas9 system has multiple benefits that mostly scientists select for genome editing in several biological systems.
Collapse
Affiliation(s)
| | - Shahid U Khan
- National Key Laboratory of Crop Genetic Improvement, Huazhong Agricultural University, Wuhan, China
| | - Ali Muhammad
- College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Limin Hu
- National Key Laboratory of Crop Genetic Improvement, Huazhong Agricultural University, Wuhan, China
| | - Yang Yang
- National Key Laboratory of Crop Genetic Improvement, Huazhong Agricultural University, Wuhan, China
| | - Chuchuan Fan
- National Key Laboratory of Crop Genetic Improvement, Huazhong Agricultural University, Wuhan, China
| |
Collapse
|
115
|
Kang JD, Kim H, Jin L, Guo Q, Cui CD, Li WX, Kim S, Kim JS, Yin XJ. Apancreatic pigs cloned using Pdx1-disrupted fibroblasts created via TALEN-mediated mutagenesis. Oncotarget 2017; 8:115480-115489. [PMID: 29383175 PMCID: PMC5777787 DOI: 10.18632/oncotarget.23301] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Accepted: 12/05/2017] [Indexed: 12/18/2022] Open
Abstract
Pancreatic and duodenal homeobox 1 (PDX1) plays a crucial role in pancreas development, β-cell differentiation, and maintenance of mature β-cell function. In this study, we designed a strategy to produce PDX1-knockout (KO) pigs. A transcription activator-like effector nuclease (TALEN) pair targeting exon 1 of the swine PDX1 gene was constructed. Porcine fetal fibroblasts (PFFs) were transfected with the TALEN plasmids plus a surrogate reporter plasmid. PDX1-mutated PFFs were enriched by magnetic separation and used to produce homozygous PDX1-KO pigs via a two-step somatic cell nuclear transfer (SCNT) cloning process. In the first SCNT step, we obtained eight fetuses, established PFF cell lines, and analyzed PDX1 gene mutations by T7 endonuclease 1 assays and Sanger sequencing. Five fetuses showed mutations at the PDX1 loci with two biallelic mutations and three monoallelic mutations (mutation rate of 62.5%). In the second step, a PDX1 biallelic mutant PFF cell line with a 2 bp deletion in one allele and a 4 bp insertion in the other allele was used as a donor to generate cloned pigs via SCNT. From 462 cloned embryos transferred into two surrogates, nine live piglets were delivered. These piglets at birth were not clearly distinguishable phenotypically from wild-type piglets, but soon developed severe diarrhea and vomiting and all died within 2 days after birth. Dissection of PDX1-KO piglets revealed that the liver, gallbladder, spleen, stomach, common bile duct, and other viscera were present and normal, but the pancreas was absent in all cases.
Collapse
Affiliation(s)
- Jin-Dan Kang
- Jilin Provincial Key Laboratory of Transgenic Animal and Embryo Engineering and Department of Animal Science, Yanbian University, Yanji 133002, China
| | - Hyojin Kim
- Center for Genome Engineering, Institute for Basic Science, Gwanak-gu, Seoul 151-747, South Korea.,Present/Current address: Department of Biosystems Science and Engineering, ETH Zurich, Basel CH-4058, Switzerland
| | - Long Jin
- Jilin Provincial Key Laboratory of Transgenic Animal and Embryo Engineering and Department of Animal Science, Yanbian University, Yanji 133002, China
| | - Qing Guo
- Jilin Provincial Key Laboratory of Transgenic Animal and Embryo Engineering and Department of Animal Science, Yanbian University, Yanji 133002, China
| | - Cheng-Du Cui
- Jilin Provincial Key Laboratory of Transgenic Animal and Embryo Engineering and Department of Animal Science, Yanbian University, Yanji 133002, China
| | - Wen-Xue Li
- Jilin Provincial Key Laboratory of Transgenic Animal and Embryo Engineering and Department of Animal Science, Yanbian University, Yanji 133002, China
| | - Seokjoong Kim
- ToolGen, Inc., Byucksan Kyoungin Digital Valley 2-Cha, Geumcheon-Gu, Seoul 153-023, South Korea
| | - Jin-Soo Kim
- Center for Genome Engineering, Institute for Basic Science, Gwanak-gu, Seoul 151-747, South Korea
| | - Xi-Jun Yin
- Jilin Provincial Key Laboratory of Transgenic Animal and Embryo Engineering and Department of Animal Science, Yanbian University, Yanji 133002, China
| |
Collapse
|
116
|
Sanagala R, Moola AK, Bollipo Diana RK. A review on advanced methods in plant gene targeting. J Genet Eng Biotechnol 2017; 15:317-321. [PMID: 30647669 PMCID: PMC6296621 DOI: 10.1016/j.jgeb.2017.07.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2016] [Revised: 06/29/2017] [Accepted: 07/03/2017] [Indexed: 12/26/2022]
Abstract
Plant genetic engineering is one of the most significant tools implemented in the modern molecular crop breeding techniques. The conventional approaches of plant genetic transformation include Agrobacterium tumefaciens, particle bombardment, DNA uptake into protoplast. The transgenic events derived by these methods carry the transgenes that are integrated at random sites in the plant genome. Novel techniques that mediate integration of foreign genes at specific pre-determined locations circumvent many problems associated with the existing methods of gene transfer. The recent years have witnessed the emergence of gene targeting techniques by employing zinc finger nucleases (ZFNs), transcription activator-like effector nucleases (TALENs), and clustered regularly interspaced short palindrome repeats (CRISPR). The present review focuses on the various approaches and their performance of plant gene targeting and suggests future directions in the important areas of plant molecular biology.
Collapse
Affiliation(s)
- Raghavendrarao Sanagala
- National Research Centre on Plant Biotechnology, Lal Bahadur Shastri Building, Pusa Campus, New Delhi 110012, India
- Department of Botany, Bharathidasan University, Tiruchirappalli, Tamil Nadu 620024, India
| | - Anil Kumar Moola
- Department of Botany, Bharathidasan University, Tiruchirappalli, Tamil Nadu 620024, India
| | | |
Collapse
|
117
|
Cooper DKC, Cowan P, Fishman JA, Hering BJ, Mohiuddin MM, Pierson RN, Sachs DH, Schuurman HJ, Dennis JU, Tönjes RR. Joint FDA‐IXA Symposium, September 20, 2017. Xenotransplantation 2017; 24. [PMID: 29193342 DOI: 10.1111/xen.12365] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- David K C Cooper
- Xenotransplantation Program, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Peter Cowan
- Immunology Research Centre, St Vincent's Hospital, Melbourne, Victoria, Australia
| | - Jay A Fishman
- Infectious Disease Division and MGH Transplant Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Bernhard J Hering
- Department of Surgery, University of Minnesota, Minneapolis, MN, USA
| | - Muhammad M Mohiuddin
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Richard N Pierson
- Department of Surgery, University of Maryland School of Medicine, Baltimore VA Medical Center, Baltimore, MD, USA
| | - David H Sachs
- Columbia University Medical Center, New York City, NY, USA.,Harvard Medical School and Massachusetts General Hospital, Boston, MA, USA
| | | | - John U Dennis
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Ralf R Tönjes
- Paul-Ehrlich-Institut, Federal Institute for Vaccines and Biomedicines, Division of Medical Biotechnology, Langen, Germany
| |
Collapse
|
118
|
Jeon S, Lim JM, Lee HG, Shin SE, Kang NK, Park YI, Oh HM, Jeong WJ, Jeong BR, Chang YK. Current status and perspectives of genome editing technology for microalgae. BIOTECHNOLOGY FOR BIOFUELS 2017; 10:267. [PMID: 29163669 PMCID: PMC5686953 DOI: 10.1186/s13068-017-0957-z] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 11/04/2017] [Indexed: 05/25/2023]
Abstract
Genome editing techniques are critical for manipulating genes not only to investigate their functions in biology but also to improve traits for genetic engineering in biotechnology. Genome editing has been greatly facilitated by engineered nucleases, dubbed molecular scissors, including zinc-finger nuclease (ZFN), TAL effector endonuclease (TALEN) and clustered regularly interspaced palindromic sequences (CRISPR)/Cas9. In particular, CRISPR/Cas9 has revolutionized genome editing fields with its simplicity, efficiency and accuracy compared to previous nucleases. CRISPR/Cas9-induced genome editing is being used in numerous organisms including microalgae. Microalgae have been subjected to extensive genetic and biological engineering due to their great potential as sustainable biofuel and chemical feedstocks. However, progress in microalgal engineering is slow mainly due to a lack of a proper transformation toolbox, and the same problem also applies to genome editing techniques. Given these problems, there are a few reports on successful genome editing in microalgae. It is, thus, time to consider the problems and solutions of genome editing in microalgae as well as further applications of this exciting technology for other scientific and engineering purposes.
Collapse
Affiliation(s)
- Seungjib Jeon
- Advanced Biomass Research and Development Center (ABC), 291 Daehak-ro, Yuseong-gu, Daejeon, 34141 Republic of Korea
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon, 34141 Republic of Korea
| | - Jong-Min Lim
- Plant Systems Engineering Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141 Republic of Korea
| | - Hyung-Gwan Lee
- Cell Factory Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141 Republic of Korea
| | - Sung-Eun Shin
- LG Chem, 188 Munji-ro, Yuseong-gu, Daejeon, 34122 Republic of Korea
| | - Nam Kyu Kang
- Advanced Biomass Research and Development Center (ABC), 291 Daehak-ro, Yuseong-gu, Daejeon, 34141 Republic of Korea
| | - Youn-Il Park
- Department of Biological Sciences, Chungnam National University, Daejeon, 34134 Republic of Korea
| | - Hee-Mock Oh
- Cell Factory Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141 Republic of Korea
| | - Won-Joong Jeong
- Plant Systems Engineering Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141 Republic of Korea
| | - Byeong-ryool Jeong
- Advanced Biomass Research and Development Center (ABC), 291 Daehak-ro, Yuseong-gu, Daejeon, 34141 Republic of Korea
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon, 34141 Republic of Korea
| | - Yong Keun Chang
- Advanced Biomass Research and Development Center (ABC), 291 Daehak-ro, Yuseong-gu, Daejeon, 34141 Republic of Korea
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon, 34141 Republic of Korea
| |
Collapse
|
119
|
Shen Y, Xu K, Yuan Z, Guo J, Zhao H, Zhang X, Zhao L, Qing Y, Li H, Pan W, Jia B, Zhao HY, Wei HJ. Efficient generation of P53 biallelic knockout Diannan miniature pigs via TALENs and somatic cell nuclear transfer. J Transl Med 2017; 15:224. [PMID: 29100547 PMCID: PMC5670695 DOI: 10.1186/s12967-017-1327-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 10/27/2017] [Indexed: 11/13/2022] Open
Abstract
Background Pigs have many features that make them attractive as biomedical models for various diseases, including cancer. P53 is an important tumor suppressor gene that exerts a central role in protecting cells from oncogenic transformation and is mutated in a large number of human cancers. P53 mutations occur in almost every type of tumor and in over 50% of all tumors. In a recent publication, pigs with a mutated P53 gene were generated that resulted in lymphoma and renal and osteogenic tumors. However, approximately 80% of human tumors have dysfunctional P53. A P53-deficient pig model is still required to elucidate. Methods Transcription activator-like effector nucleases (TALENs) were designed to target porcine P53 exon 4. The targeting activity was evaluated using a luciferase SSA recombination assay. P53 biallelic knockout (KO) cell lines were established from single-cell colonies of fetal fibroblasts derived from Diannan miniature pigs followed by electroporation with TALENs plasmids. One cell line was selected as the donor cell line for somatic cell nuclear transfer (SCNT) for the generation of P53 KO pigs. P53 KO stillborn fetuses and living piglets were obtained. Gene typing of the collected cloned individuals was performed by T7EI assay and sequencing. Fibroblast cells from Diannan miniature piglets with a P53 biallelic knockout or wild type were analyzed for the P53 response to doxorubicin treatment by confocal microscopy and western blotting. Results The luciferase SSA recombination assay revealed that the targeting activities of the designed TALENs were 55.35-fold higher than those of the control. Eight cell lines (8/19) were mutated for P53, and five of them were biallelic knockouts. One of the biallelic knockout cell lines was selected as nuclear donor cells for SCNT. The cloned embryos were transferred into five recipient gilts, three of them becoming pregnant. Five live fetuses were obtained from one surrogate by caesarean section after 38 days of gestation for genotyping. Finally, six live piglets and one stillborn piglet were collected from two recipients by caesarean section. Sequencing analyses of the target site confirmed the P53 biallelic knockout in all fetuses and piglets, consistent with the genotype of the donor cells. The qPCR analysis showed that the expression of the P53 mRNA had significant reduction in various tissues of the knockout piglets. Furthermore, confocal microscopy and western blotting analyses demonstrated that the fibroblast cells of Diannan miniature piglets with a P53 biallelic knockout were defective in mediating DNA damage when incubated with doxorubicin. Conclusion TALENs combined with SCNT was successfully used to generate P53 KO Diannan miniature pigs. Although these genetically engineered Diannan miniature pigs had no tumorigenic signs, the P53 gene was dysfunctional. We believe that these pigs will provide powerful new resources for preclinical oncology and basic cancer research. Electronic supplementary material The online version of this article (10.1186/s12967-017-1327-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Youfeng Shen
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming, 650201, China.,College of Animal Science and Technology, Yunnan Agricultural University, Kunming, China
| | - Kaixiang Xu
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming, 650201, China
| | - Zaimei Yuan
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming, 650201, China.,College of Animal Science and Technology, Yunnan Agricultural University, Kunming, China
| | - Jianxiong Guo
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming, 650201, China
| | - Heng Zhao
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming, 650201, China.,College of Animal Science and Technology, Yunnan Agricultural University, Kunming, China
| | - Xuezeng Zhang
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming, 650201, China.,Key Laboratory Animal Nutrition and Feed of Yunnan Province, Yunnan Agricultural University, Kunming, 650201, China
| | - Lu Zhao
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming, 650201, China.,Key Laboratory Animal Nutrition and Feed of Yunnan Province, Yunnan Agricultural University, Kunming, 650201, China
| | - Yubo Qing
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming, 650201, China.,College of Animal Science and Technology, Yunnan Agricultural University, Kunming, China
| | - Honghui Li
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming, 650201, China.,College of Animal Science and Technology, Yunnan Agricultural University, Kunming, China
| | - Weirong Pan
- College of Animal Science and Technology, Yunnan Agricultural University, Kunming, China
| | - Baoyu Jia
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming, 650201, China.,College of Animal Science and Technology, Yunnan Agricultural University, Kunming, China
| | - Hong-Ye Zhao
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming, 650201, China.
| | - Hong-Jiang Wei
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming, 650201, China. .,College of Animal Science and Technology, Yunnan Agricultural University, Kunming, China. .,Key Laboratory Animal Nutrition and Feed of Yunnan Province, Yunnan Agricultural University, Kunming, 650201, China.
| |
Collapse
|
120
|
Genome editing in livestock: Are we ready for a revolution in animal breeding industry? Transgenic Res 2017; 26:715-726. [DOI: 10.1007/s11248-017-0049-7] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2017] [Accepted: 10/24/2017] [Indexed: 12/25/2022]
|
121
|
Bhat SA, Malik AA, Ahmad SM, Shah RA, Ganai NA, Shafi SS, Shabir N. Advances in genome editing for improved animal breeding: A review. Vet World 2017; 10:1361-1366. [PMID: 29263600 PMCID: PMC5732344 DOI: 10.14202/vetworld.2017.1361-1366] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 10/20/2017] [Indexed: 01/05/2023] Open
Abstract
Since centuries, the traits for production and disease resistance are being targeted while improving the genetic merit of domestic animals, using conventional breeding programs such as inbreeding, outbreeding, or introduction of marker-assisted selection. The arrival of new scientific concepts, such as cloning and genome engineering, has added a new and promising research dimension to the existing animal breeding programs. Development of genome editing technologies such as transcription activator-like effector nuclease, zinc finger nuclease, and clustered regularly interspaced short palindromic repeats systems begun a fresh era of genome editing, through which any change in the genome, including specific DNA sequence or indels, can be made with unprecedented precision and specificity. Furthermore, it offers an opportunity of intensification in the frequency of desirable alleles in an animal population through gene-edited individuals more rapidly than conventional breeding. The specific research is evolving swiftly with a focus on improvement of economically important animal species or their traits all of which form an important subject of this review. It also discusses the hurdles to commercialization of these techniques despite several patent applications owing to the ambiguous legal status of genome-editing methods on account of their disputed classification. Nonetheless, barring ethical concerns gene-editing entailing economically important genes offers a tremendous potential for breeding animals with desirable traits.
Collapse
Affiliation(s)
- Shakil Ahmad Bhat
- Division of Animal Biotechnology, Faculty of Veterinary Sciences and Animal Husbandry, Sher-e-Kashmir University of Agricultural Sciences and Technology of Kashmir, Srinagar - 190 006, Jammu and Kashmir, India
| | - Abrar Ahad Malik
- Division of Animal Biotechnology, Faculty of Veterinary Sciences and Animal Husbandry, Sher-e-Kashmir University of Agricultural Sciences and Technology of Kashmir, Srinagar - 190 006, Jammu and Kashmir, India
| | - Syed Mudasir Ahmad
- Division of Animal Biotechnology, Faculty of Veterinary Sciences and Animal Husbandry, Sher-e-Kashmir University of Agricultural Sciences and Technology of Kashmir, Srinagar - 190 006, Jammu and Kashmir, India
| | - Riaz Ahmad Shah
- Division of Animal Biotechnology, Faculty of Veterinary Sciences and Animal Husbandry, Sher-e-Kashmir University of Agricultural Sciences and Technology of Kashmir, Srinagar - 190 006, Jammu and Kashmir, India
| | - Nazir Ahmad Ganai
- Division of Animal Genetics and Breeding, Faculty of Veterinary Sciences and Animal Husbandry, Sher-e-Kashmir University of Agricultural Sciences and Technology of Kashmir, Srinagar - 190 006, Jammu and Kashmir, India
| | - Syed Shanaz Shafi
- Division of Animal Genetics and Breeding, Faculty of Veterinary Sciences and Animal Husbandry, Sher-e-Kashmir University of Agricultural Sciences and Technology of Kashmir, Srinagar - 190 006, Jammu and Kashmir, India
| | - Nadeem Shabir
- Division of Animal Biotechnology, Faculty of Veterinary Sciences and Animal Husbandry, Sher-e-Kashmir University of Agricultural Sciences and Technology of Kashmir, Srinagar - 190 006, Jammu and Kashmir, India
| |
Collapse
|
122
|
Wang M, Sun Z, Yu T, Ding F, Li L, Wang X, Fu M, Wang H, Huang J, Li N, Dai Y. Large-scale production of recombinant human lactoferrin from high-expression, marker-free transgenic cloned cows. Sci Rep 2017; 7:10733. [PMID: 28878310 PMCID: PMC5587717 DOI: 10.1038/s41598-017-11462-z] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Accepted: 08/25/2017] [Indexed: 11/23/2022] Open
Abstract
Human lactoferrin (hLF) is a valuable protein for pharmaceutical products and functional foods, and worldwide demand for this protein has steadily increased. However, large-scale recombinant human lactoferrin (rhLF) production using current animal bioreactor techniques is limited by the low expression of foreign proteins, the use of antibiotic resistance genes and the down-regulation of endogenous milk proteins. Here, we generated a herd of marker-free, hLF bacterial artificial chromosome (BAC) transgenic cloned cows, as confirmed by Polymerase chain reaction, Southern blot and Western blot analyses. These transgenic cloned cows produced rhLF in milk at concentrations of 4.5–13.6 g/L. Moreover, the total protein content of the milk was increased. Over two hundred transgenic cloned cows were propagated by multiple ovulation and embryo transfer (MOET). A total of 400–450 g of rhLF protein, which shows similar enzymatic activity to natural hLF in iron binding and release, can be purified on a large scale from >100 L of milk per day. Our results suggested that transgenic bovine mammary bioreactors have the potential for large-scale protein production.
Collapse
Affiliation(s)
- Ming Wang
- State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Zhaolin Sun
- State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Tian Yu
- Kejienuo Biotechnology Company, Wuxi, China
| | - Fangrong Ding
- State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Ling Li
- State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Xi Wang
- Kejienuo Biotechnology Company, Wuxi, China
| | - Mingbo Fu
- Kejienuo Biotechnology Company, Wuxi, China
| | - Haiping Wang
- State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Jinming Huang
- Dairy cattle Research Center, Academy of Agricultural Sciences, Shandong, China
| | - Ning Li
- State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China.
| | - Yunping Dai
- State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China.
| |
Collapse
|
123
|
Li C, Peng Q, Wan X, Sun H, Tang J. C-terminal motifs in promyelocytic leukemia protein isoforms critically regulate PML nuclear body formation. J Cell Sci 2017; 130:3496-3506. [PMID: 28851805 DOI: 10.1242/jcs.202879] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 08/10/2017] [Indexed: 12/11/2022] Open
Abstract
Promyelocytic leukemia protein (PML) nuclear bodies (NBs), which are sub-nuclear protein structures, are involved in a variety of important cellular functions. PML-NBs are assembled by PML isoforms, and contact between small ubiquitin-like modifiers (SUMOs) with the SUMO interaction motif (SIM) are critically involved in this process. PML isoforms contain a common N-terminal region and a variable C-terminus. However, the contribution of the C-terminal regions to PML-NB formation remains poorly defined. Here, using high-resolution microscopy, we show that mutation of the SIM distinctively influences the structure of NBs formed by each individual PML isoform, with that of PML-III and PML-V minimally changed, and PML-I and PML-IV dramatically impaired. We further identify several C-terminal elements that are important in regulating NB structure and provide strong evidence to suggest that the 8b element in PML-IV possesses a strong ability to interact with SUMO-1 and SUMO-2, and critically participates in NB formation. Our findings highlight the importance of PML C-termini in NB assembly and function, and provide molecular insight into the PML-NB assembly of each distinctive isoform.
Collapse
Affiliation(s)
- Chuang Li
- State Key Laboratory of Agrobiotechnology and College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Qiongfang Peng
- State Key Laboratory of Agrobiotechnology and College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Xiao Wan
- State Key Laboratory of Agrobiotechnology and College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Haili Sun
- State Key Laboratory of Agrobiotechnology and College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Jun Tang
- State Key Laboratory of Agrobiotechnology and College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| |
Collapse
|
124
|
Woodcock ME, Idoko-Akoh A, McGrew MJ. Gene editing in birds takes flight. Mamm Genome 2017; 28:315-323. [PMID: 28612238 PMCID: PMC5569130 DOI: 10.1007/s00335-017-9701-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 06/05/2017] [Indexed: 12/28/2022]
Abstract
The application of gene editing (GE) technology to create precise changes to the genome of bird species will provide new and exciting opportunities for the biomedical, agricultural and biotechnology industries, as well as providing new approaches for producing research models. Recent advances in modifying both the somatic and germ cell lineages in chicken indicate that this species, and conceivably soon other avian species, has joined a growing number of model organisms in the gene editing revolution.
Collapse
Affiliation(s)
- Mark E Woodcock
- The Roslin Institute and Royal Dick School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, Midlothian, EH25 9RG, UK.
| | - Alewo Idoko-Akoh
- The Roslin Institute and Royal Dick School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, Midlothian, EH25 9RG, UK
| | - Michael J McGrew
- The Roslin Institute and Royal Dick School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, Midlothian, EH25 9RG, UK
| |
Collapse
|
125
|
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: 2.0] [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
|
126
|
Swine models, genomic tools and services to enhance our understanding of human health and diseases. Lab Anim (NY) 2017; 46:167-172. [PMID: 28328880 PMCID: PMC7091812 DOI: 10.1038/laban.1215] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Accepted: 12/07/2016] [Indexed: 12/20/2022]
Abstract
The pig is becoming increasingly important as a biomedical model. Given the similarities between pigs and humans, a greater understanding of the underlying biology of human health and diseases may come from the pig rather than from classical rodent models. With an increasing need for swine models, it is essential that the genomic tools, models and services be readily available to the scientific community. Many of these are available through the National Swine Resource and Research Center (NSRRC), a facility funded by the US National Institutes of Health at the University of Missouri. The goal of the NSRRC is to provide high-quality biomedical swine models to the scientific community.
Collapse
|
127
|
Liu P, Wang W, Wei D. Use of transcription activator-like effector for efficient gene modification and transcription in the filamentous fungus Trichoderma reesei. J Ind Microbiol Biotechnol 2017; 44:1367-1373. [PMID: 28674932 DOI: 10.1007/s10295-017-1963-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Accepted: 06/17/2017] [Indexed: 12/01/2022]
Abstract
Filamentous fungi have wide applications in the field of biotechnology. The use of transcription activator-like effectors (TALEs) is a powerful genome-engineering tool, which can facilitate genome editing and transcriptional modulation, and has been used for genetic modification in a variety of organisms. However, a transcription activator-like effectors nuclease (TALEN) approach has not been used in filamentous fungi so far. Here, we aimed to establish the shortest TALEN and TALE-transcription factor (TALE-TF) proteins for use in gene modification and transcription in the filamentous fungus Trichoderma reesei. The alternative tandem repeat unit of the TALEs was first established by the ligation of two isocaudamers: XbaI and SpeI, instead of the natural unit. In addition, we adopted T. reesei expression assays to monitor the activities of the TALENs and TALE-TF in vivo in T. reesei. Our results showed that TALEs are a powerful genome-manipulating tool for use in T. reesei and other filamentous fungal species and that their use might facilitate studies on functional genomics and strain improvement in these filamentous fungi.
Collapse
Affiliation(s)
- Pei Liu
- State Key Lab of Bioreactor Engineering, New World Institute of Biotechnology, East China University of Science and Technology, P.O.B. 311, 130 Meilong Road, Shanghai, 200237, China
| | - Wei Wang
- State Key Lab of Bioreactor Engineering, New World Institute of Biotechnology, East China University of Science and Technology, P.O.B. 311, 130 Meilong Road, Shanghai, 200237, China.
| | - Dongzhi Wei
- State Key Lab of Bioreactor Engineering, New World Institute of Biotechnology, East China University of Science and Technology, P.O.B. 311, 130 Meilong Road, Shanghai, 200237, China
| |
Collapse
|
128
|
Jennings CG, Landman R, Zhou Y, Sharma J, Hyman J, Movshon JA, Qiu Z, Roberts AC, Roe AW, Wang X, Zhou H, Wang L, Zhang F, Desimone R, Feng G. Opportunities and challenges in modeling human brain disorders in transgenic primates. Nat Neurosci 2017; 19:1123-30. [PMID: 27571191 DOI: 10.1038/nn.4362] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 07/19/2016] [Indexed: 12/15/2022]
Abstract
Molecular genetic tools have had a profound impact on neuroscience, but until recently their application has largely been confined to a few model species, most notably mouse, zebrafish, Drosophila melanogaster and Caenorhabditis elegans. With the development of new genome engineering technologies such as CRISPR, it is becoming increasingly feasible to apply these molecular tools in a wider range of species, including nonhuman primates. This will lead to many opportunities for brain research, but it will also pose challenges. Here we identify some of these opportunities and challenges in light of recent and foreseeable technological advances and offer some suggestions. Our main focus is on the creation of new primate disease models for understanding the pathological mechanisms of brain disorders and for developing new approaches to effective treatment. However, we also emphasize that primate genetic models have great potential to address many fundamental questions about brain function, providing an essential foundation for future progress in disease research.
Collapse
Affiliation(s)
- Charles G Jennings
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA.,Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Rogier Landman
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA.,Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA.,Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Yang Zhou
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA.,Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Jitendra Sharma
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA.,Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Julia Hyman
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA.,Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - J Anthony Movshon
- Center for Neural Science, New York University, New York, New York, USA
| | - Zilong Qiu
- Institute of Neuroscience, CAS Key Laboratory of Primate Neurobiology, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Angela C Roberts
- Department of Physiology, Development and Neuroscience, Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, UK
| | - Anna Wang Roe
- Interdisciplinary Institute of Neuroscience and Technology, Zhejiang University, Hangzhou, China
| | - Xiaoqin Wang
- Laboratory of Auditory Neurophysiology, Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Huihui Zhou
- The Brain Cognition and Brain Disease Institute (BCBDI) for Collaboration Research of SIAT at CAS and McGovern Institute at MIT, Shenzhen Institutes of Advanced Technology (SIAT), Chinese Academy of Science, Shenzhen, China
| | - Liping Wang
- Shenzhen Key Lab of Neuropsychiatric Modulation and Collaborative Innovation Center for Brain Science, CAS Center for Excellence in Brain Science and Intelligence Technology, The Brain Cognition and Brain Disease Institute (BCBDI) for Collaboration Research of SIAT at CAS and McGovern Institute at MIT, Shenzhen Institutes of Advanced Technology (SIAT), Chinese Academy of Science, Shenzhen, China
| | - Feng Zhang
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA.,Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA.,Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Robert Desimone
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA.,Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Guoping Feng
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA.,Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA.,Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| |
Collapse
|
129
|
Bahbahani H, Tijjani A, Mukasa C, Wragg D, Almathen F, Nash O, Akpa GN, Mbole-Kariuki M, Malla S, Woolhouse M, Sonstegard T, Van Tassell C, Blythe M, Huson H, Hanotte O. Signatures of Selection for Environmental Adaptation and Zebu × Taurine Hybrid Fitness in East African Shorthorn Zebu. Front Genet 2017. [PMID: 28642786 PMCID: PMC5462927 DOI: 10.3389/fgene.2017.00068] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The East African Shorthorn Zebu (EASZ) cattle are ancient hybrid between Asian zebu × African taurine cattle preferred by local farmers due to their adaptability to the African environment. The genetic controls of these adaptabilities are not clearly understood yet. Here, we genotyped 92 EASZ samples from Kenya (KEASZ) with more than 770,000 SNPs and sequenced the genome of a pool of 10 KEASZ. We observe an even admixed autosomal zebu × taurine genomic structure in the population. A total of 101 and 165 candidate regions of positive selection, based on genome-wide SNP analyses (meta-SS, Rsb, iHS, and ΔAF) and pooled heterozygosity (Hp) full genome sequence analysis, are identified, in which 35 regions are shared between them. A total of 142 functional variants, one novel, have been detected within these regions, in which 30 and 26 were classified as of zebu and African taurine origins, respectively. High density genome-wide SNP analysis of zebu × taurine admixed cattle populations from Uganda and Nigeria show that 25 of these regions are shared between KEASZ and Uganda cattle, and seven regions are shared across the KEASZ, Uganda, and Nigeria cattle. The identification of common candidate regions allows us to fine map 18 regions. These regions intersect with genes and QTL associated with reproduction and environmental stress (e.g., immunity and heat stress) suggesting that the genome of the zebu × taurine admixed cattle has been uniquely selected to maximize hybrid fitness both in terms of reproduction and survivability.
Collapse
Affiliation(s)
- Hussain Bahbahani
- Department of Biological Sciences, Faculty of Science, Kuwait UniversityKuwait, Kuwait
| | - Abdulfatai Tijjani
- School of Life Sciences, University of NottinghamNottingham, United Kingdom.,Centre for Genomics Research and Innovation, National Biotechnology Development AgencyAbuja, Nigeria
| | | | - David Wragg
- Centre for Tropical Livestock Genetics and Health, Roslin InstituteEdinburgh, United Kingdom
| | - Faisal Almathen
- Department of Veterinary Public Health and Animal Husbandry, College of Veterinary Medicine, King Faisal UniversityAl-Hasa, Saudi Arabia
| | - Oyekanmi Nash
- Centre for Genomics Research and Innovation, National Biotechnology Development AgencyAbuja, Nigeria
| | - Gerald N Akpa
- Department of Animal Science, Ahmadu Bello UniversityZaria, Nigeria
| | - Mary Mbole-Kariuki
- School of Life Sciences, University of NottinghamNottingham, United Kingdom
| | - Sunir Malla
- Deep Seq Department, University of NottinghamNottingham, United Kingdom
| | - Mark Woolhouse
- Ashworth Laboratories, Centre for Immunity, Infection and Evolution, University of EdinburghEdinburgh, United Kingdom
| | | | - Curtis Van Tassell
- Animal Genomics and Improvement Laboratory, United States Department of Agriculture, Agricultural Research ServiceBeltsville, MD, United States
| | - Martin Blythe
- Deep Seq Department, University of NottinghamNottingham, United Kingdom
| | - Heather Huson
- Animal Genomics and Improvement Laboratory, United States Department of Agriculture, Agricultural Research ServiceBeltsville, MD, United States
| | - Olivier Hanotte
- School of Life Sciences, University of NottinghamNottingham, United Kingdom.,International Livestock Research Institute (ILRI)Addis Ababa, Ethiopia
| |
Collapse
|
130
|
Wells KD, Prather RS. Genome-editing technologies to improve research, reproduction, and production in pigs. Mol Reprod Dev 2017; 84:1012-1017. [PMID: 28394093 DOI: 10.1002/mrd.22812] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 04/04/2017] [Indexed: 12/22/2022]
Abstract
The ability to directly manipulate the pig genome through genetic engineering has been available to the research community for over three decades. This technology has progressed from the random insertion of foreign DNA, via a variety of techniques (pronuclear microinjection, sperm mediated gene transfer, and integration of mobile genetic elements), to manipulation of endogenous genes, via homologous recombination in somatic cells followed by somatic cell nuclear transfer. Over the last few years, designer nucleases facilitated the development of techniques that provide efficient ways to introduce foreign DNA or to modify endogenous genes in eggs, zygotes, or somatic cells. Together, these genome-editing technologies have essentially removed the obstacles to gene manipulation in swine. Although the regulatory environment is still unclear for agricultural applications, genetic engineering of pigs will continue to advance biomedicine and biology. In addition, genetic engineering is now sufficiently simple and efficient that agricultural research can now ask basic and applied questions that are not hampered by limited funding.
Collapse
Affiliation(s)
- Kevin D Wells
- Division of Animal Science, University of Missouri, Columbia, Missouri
| | - Randall S Prather
- Division of Animal Science, University of Missouri, Columbia, Missouri
| |
Collapse
|
131
|
Fernández A, Josa S, Montoliu L. A history of genome editing in mammals. Mamm Genome 2017; 28:237-246. [DOI: 10.1007/s00335-017-9699-2] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 05/31/2017] [Indexed: 12/28/2022]
|
132
|
Cooper CA, Challagulla A, Jenkins KA, Wise TG, O'Neil TE, Morris KR, Tizard ML, Doran TJ. Generation of gene edited birds in one generation using sperm transfection assisted gene editing (STAGE). Transgenic Res 2017; 26:331-347. [PMID: 27896535 DOI: 10.1007/s11248-016-0003-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 11/16/2016] [Indexed: 12/28/2022]
Abstract
Generating transgenic and gene edited mammals involves in vitro manipulation of oocytes or single cell embryos. Due to the comparative inaccessibility of avian oocytes and single cell embryos, novel protocols have been developed to produce transgenic and gene edited birds. While these protocols are relatively efficient, they involve two generation intervals before reaching complete somatic and germline expressing transgenic or gene edited birds. Most of this work has been done with chickens, and many protocols require in vitro culturing of primordial germ cells (PGCs). However, for many other bird species no methodology for long term culture of PGCs exists. Developing methodologies to produce germline transgenic or gene edited birds in the first generation would save significant amounts of time and resource. Furthermore, developing protocols that can be readily adapted to a wide variety of avian species would open up new research opportunities. Here we report a method using sperm as a delivery mechanism for gene editing vectors which we call sperm transfection assisted gene editing (STAGE). We have successfully used this method to generate GFP knockout embryos and chickens, as well as generate embryos with mutations in the doublesex and mab-3 related transcription factor 1 (DMRT1) gene using the CRISPR/Cas9 system. The efficiency of the method varies from as low as 0% to as high as 26% with multiple factors such as CRISPR guide efficiency and mRNA stability likely impacting the outcome. This straightforward methodology could simplify gene editing in many bird species including those for which no methodology currently exists.
Collapse
Affiliation(s)
- Caitlin A Cooper
- CSIRO Health and Biosecurity, Australian Animal Health Laboratory, Geelong, VIC, Australia
| | - Arjun Challagulla
- CSIRO Health and Biosecurity, Australian Animal Health Laboratory, Geelong, VIC, Australia
| | - Kristie A Jenkins
- CSIRO Health and Biosecurity, Australian Animal Health Laboratory, Geelong, VIC, Australia
| | - Terry G Wise
- CSIRO Health and Biosecurity, Australian Animal Health Laboratory, Geelong, VIC, Australia
| | - Terri E O'Neil
- CSIRO Health and Biosecurity, Australian Animal Health Laboratory, Geelong, VIC, Australia
| | - Kirsten R Morris
- CSIRO Health and Biosecurity, Australian Animal Health Laboratory, Geelong, VIC, Australia
| | - Mark L Tizard
- CSIRO Health and Biosecurity, Australian Animal Health Laboratory, Geelong, VIC, Australia
| | - Timothy J Doran
- CSIRO Health and Biosecurity, Australian Animal Health Laboratory, Geelong, VIC, Australia.
| |
Collapse
|
133
|
Liška F, Landa V, Zídek V, Mlejnek P, Šilhavý J, Šimáková M, Strnad H, Trnovská J, Škop V, Kazdová L, Starker CG, Voytas DF, Izsvák Z, Mancini M, Šeda O, Křen V, Pravenec M. Downregulation of
Plzf
Gene Ameliorates Metabolic and Cardiac Traits in the Spontaneously Hypertensive Rat. Hypertension 2017; 69:1084-1091. [DOI: 10.1161/hypertensionaha.116.08798] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Revised: 12/06/2016] [Accepted: 03/09/2017] [Indexed: 12/20/2022]
Abstract
The spontaneously hypertensive rat (SHR), one of the most widely used model of essential hypertension, is predisposed to left ventricular hypertrophy, myocardial fibrosis, and metabolic disturbances. Recently, quantitative trait loci influencing blood pressure, left ventricular mass, and heart interstitial fibrosis were genetically isolated within a minimal congenic subline that contains only 7 genes, including mutant
Plzf
(promyelocytic leukemia zinc finger) candidate gene. To identify
Plzf
as a quantitative trait gene, we targeted
Plzf
in the SHR using the transcription activator-like effector nuclease technique and obtained SHR line harboring targeted
Plzf
gene with a premature stop codon. Because the
Plzf
targeted allele is semilethal, morphologically normal heterozygous rats were used for metabolic and hemodynamic analyses. SHR-
Plzf
+/−
heterozygotes versus SHR wild-type controls exhibited reduced body weight and relative weight of epididymal fat, lower serum and liver triglycerides and cholesterol, and better glucose tolerance. In addition, SHR-
Plzf
+/−
rats exhibited significantly increased sensitivity of adipose and muscle tissue to insulin action when compared with wild-type controls. Blood pressure was comparable in SHR versus SHR-
Plzf
+/−
; however, there was significant amelioration of cardiomyocyte hypertrophy and cardiac fibrosis in SHR-
Plzf
+/−
rats. Gene expression profiles in the liver and expression of selected genes in the heart revealed differentially expressed genes that play a role in metabolic pathways, PPAR (peroxisome proliferator-activated receptor) signaling, and cell cycle regulation. These results provide evidence for an important role of
Plzf
in regulation of metabolic and cardiac traits in the rat and suggest a cross talk between cell cycle regulators, metabolism, cardiac hypertrophy, and fibrosis.
Collapse
Affiliation(s)
- František Liška
- From the Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University, Prague, Czech Republic (F.L., O.Š., V.K., M.P.); Institute of Physiology (V.L., V.Z., P.M., J.Š., M.Š., M.P.) and Institute of Molecular Genetics (H.S.), Czech Academy of Sciences, Prague, Czech Republic; Institute for Experimental Medicine, Prague, Czech Republic (J.T., V.Š., L.K.); Department of Genetics, Cell Biology, and Development and Center for Genome Engineering, University of Minnesota,
| | - Vladimír Landa
- From the Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University, Prague, Czech Republic (F.L., O.Š., V.K., M.P.); Institute of Physiology (V.L., V.Z., P.M., J.Š., M.Š., M.P.) and Institute of Molecular Genetics (H.S.), Czech Academy of Sciences, Prague, Czech Republic; Institute for Experimental Medicine, Prague, Czech Republic (J.T., V.Š., L.K.); Department of Genetics, Cell Biology, and Development and Center for Genome Engineering, University of Minnesota,
| | - Václav Zídek
- From the Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University, Prague, Czech Republic (F.L., O.Š., V.K., M.P.); Institute of Physiology (V.L., V.Z., P.M., J.Š., M.Š., M.P.) and Institute of Molecular Genetics (H.S.), Czech Academy of Sciences, Prague, Czech Republic; Institute for Experimental Medicine, Prague, Czech Republic (J.T., V.Š., L.K.); Department of Genetics, Cell Biology, and Development and Center for Genome Engineering, University of Minnesota,
| | - Petr Mlejnek
- From the Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University, Prague, Czech Republic (F.L., O.Š., V.K., M.P.); Institute of Physiology (V.L., V.Z., P.M., J.Š., M.Š., M.P.) and Institute of Molecular Genetics (H.S.), Czech Academy of Sciences, Prague, Czech Republic; Institute for Experimental Medicine, Prague, Czech Republic (J.T., V.Š., L.K.); Department of Genetics, Cell Biology, and Development and Center for Genome Engineering, University of Minnesota,
| | - Jan Šilhavý
- From the Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University, Prague, Czech Republic (F.L., O.Š., V.K., M.P.); Institute of Physiology (V.L., V.Z., P.M., J.Š., M.Š., M.P.) and Institute of Molecular Genetics (H.S.), Czech Academy of Sciences, Prague, Czech Republic; Institute for Experimental Medicine, Prague, Czech Republic (J.T., V.Š., L.K.); Department of Genetics, Cell Biology, and Development and Center for Genome Engineering, University of Minnesota,
| | - Miroslava Šimáková
- From the Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University, Prague, Czech Republic (F.L., O.Š., V.K., M.P.); Institute of Physiology (V.L., V.Z., P.M., J.Š., M.Š., M.P.) and Institute of Molecular Genetics (H.S.), Czech Academy of Sciences, Prague, Czech Republic; Institute for Experimental Medicine, Prague, Czech Republic (J.T., V.Š., L.K.); Department of Genetics, Cell Biology, and Development and Center for Genome Engineering, University of Minnesota,
| | - Hynek Strnad
- From the Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University, Prague, Czech Republic (F.L., O.Š., V.K., M.P.); Institute of Physiology (V.L., V.Z., P.M., J.Š., M.Š., M.P.) and Institute of Molecular Genetics (H.S.), Czech Academy of Sciences, Prague, Czech Republic; Institute for Experimental Medicine, Prague, Czech Republic (J.T., V.Š., L.K.); Department of Genetics, Cell Biology, and Development and Center for Genome Engineering, University of Minnesota,
| | - Jaroslava Trnovská
- From the Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University, Prague, Czech Republic (F.L., O.Š., V.K., M.P.); Institute of Physiology (V.L., V.Z., P.M., J.Š., M.Š., M.P.) and Institute of Molecular Genetics (H.S.), Czech Academy of Sciences, Prague, Czech Republic; Institute for Experimental Medicine, Prague, Czech Republic (J.T., V.Š., L.K.); Department of Genetics, Cell Biology, and Development and Center for Genome Engineering, University of Minnesota,
| | - Vojtěch Škop
- From the Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University, Prague, Czech Republic (F.L., O.Š., V.K., M.P.); Institute of Physiology (V.L., V.Z., P.M., J.Š., M.Š., M.P.) and Institute of Molecular Genetics (H.S.), Czech Academy of Sciences, Prague, Czech Republic; Institute for Experimental Medicine, Prague, Czech Republic (J.T., V.Š., L.K.); Department of Genetics, Cell Biology, and Development and Center for Genome Engineering, University of Minnesota,
| | - Ludmila Kazdová
- From the Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University, Prague, Czech Republic (F.L., O.Š., V.K., M.P.); Institute of Physiology (V.L., V.Z., P.M., J.Š., M.Š., M.P.) and Institute of Molecular Genetics (H.S.), Czech Academy of Sciences, Prague, Czech Republic; Institute for Experimental Medicine, Prague, Czech Republic (J.T., V.Š., L.K.); Department of Genetics, Cell Biology, and Development and Center for Genome Engineering, University of Minnesota,
| | - Colby G. Starker
- From the Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University, Prague, Czech Republic (F.L., O.Š., V.K., M.P.); Institute of Physiology (V.L., V.Z., P.M., J.Š., M.Š., M.P.) and Institute of Molecular Genetics (H.S.), Czech Academy of Sciences, Prague, Czech Republic; Institute for Experimental Medicine, Prague, Czech Republic (J.T., V.Š., L.K.); Department of Genetics, Cell Biology, and Development and Center for Genome Engineering, University of Minnesota,
| | - Daniel F. Voytas
- From the Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University, Prague, Czech Republic (F.L., O.Š., V.K., M.P.); Institute of Physiology (V.L., V.Z., P.M., J.Š., M.Š., M.P.) and Institute of Molecular Genetics (H.S.), Czech Academy of Sciences, Prague, Czech Republic; Institute for Experimental Medicine, Prague, Czech Republic (J.T., V.Š., L.K.); Department of Genetics, Cell Biology, and Development and Center for Genome Engineering, University of Minnesota,
| | - Zsuzsanna Izsvák
- From the Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University, Prague, Czech Republic (F.L., O.Š., V.K., M.P.); Institute of Physiology (V.L., V.Z., P.M., J.Š., M.Š., M.P.) and Institute of Molecular Genetics (H.S.), Czech Academy of Sciences, Prague, Czech Republic; Institute for Experimental Medicine, Prague, Czech Republic (J.T., V.Š., L.K.); Department of Genetics, Cell Biology, and Development and Center for Genome Engineering, University of Minnesota,
| | - Massimiliano Mancini
- From the Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University, Prague, Czech Republic (F.L., O.Š., V.K., M.P.); Institute of Physiology (V.L., V.Z., P.M., J.Š., M.Š., M.P.) and Institute of Molecular Genetics (H.S.), Czech Academy of Sciences, Prague, Czech Republic; Institute for Experimental Medicine, Prague, Czech Republic (J.T., V.Š., L.K.); Department of Genetics, Cell Biology, and Development and Center for Genome Engineering, University of Minnesota,
| | - Ondřej Šeda
- From the Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University, Prague, Czech Republic (F.L., O.Š., V.K., M.P.); Institute of Physiology (V.L., V.Z., P.M., J.Š., M.Š., M.P.) and Institute of Molecular Genetics (H.S.), Czech Academy of Sciences, Prague, Czech Republic; Institute for Experimental Medicine, Prague, Czech Republic (J.T., V.Š., L.K.); Department of Genetics, Cell Biology, and Development and Center for Genome Engineering, University of Minnesota,
| | - Vladimír Křen
- From the Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University, Prague, Czech Republic (F.L., O.Š., V.K., M.P.); Institute of Physiology (V.L., V.Z., P.M., J.Š., M.Š., M.P.) and Institute of Molecular Genetics (H.S.), Czech Academy of Sciences, Prague, Czech Republic; Institute for Experimental Medicine, Prague, Czech Republic (J.T., V.Š., L.K.); Department of Genetics, Cell Biology, and Development and Center for Genome Engineering, University of Minnesota,
| | - Michal Pravenec
- From the Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University, Prague, Czech Republic (F.L., O.Š., V.K., M.P.); Institute of Physiology (V.L., V.Z., P.M., J.Š., M.Š., M.P.) and Institute of Molecular Genetics (H.S.), Czech Academy of Sciences, Prague, Czech Republic; Institute for Experimental Medicine, Prague, Czech Republic (J.T., V.Š., L.K.); Department of Genetics, Cell Biology, and Development and Center for Genome Engineering, University of Minnesota,
| |
Collapse
|
134
|
Generation of CMAHKO/GTKO/shTNFRI-Fc/HO-1 quadruple gene modified pigs. Transgenic Res 2017; 26:435-445. [PMID: 28553699 DOI: 10.1007/s11248-017-0021-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2016] [Accepted: 04/25/2017] [Indexed: 12/16/2022]
Abstract
As an alternative source of organs for transplantation into humans, attention has been directed to pigs due to their similarities in biological features and organ size. However, severe immune rejection has prevented successful xenotransplantation using pig organs and tissues. To overcome immune rejection, recently developed genetic engineering systems such as TALEN coupled with somatic cell nuclear transfer (SCNT) to make embryos could be used to produce pigs compatible with xenotransplantation. We used the TALEN system to target the non-Gal antigen cytidine monophosphate-N-acetylneuraminic acid hydroxylase (CMAH) gene in pigs that is naturally deleted in humans. Gal-deleted cells expressing both soluble human tumor necrosis factor receptor I IgG1-Fc (shTNFRI-Fc) and human hemagglutinin -tagged-human heme oxygenase-1 (hHO-1) were transfected with a TALEN target for CMAH. Cells lacking CMAH were negatively selected using N-glyconeuraminic acid (Neu5Gc)/magnetic beads and the level of Neu5Gc expression of isolated cells were analyzed by FACS and DNA sequencing. Cloned embryos using 3 different genetically modified cell clones were respectively transferred into 3 recipients, with 55.6% (5/9) becoming pregnant and three cloned pigs were produced. Successful genetic disruption of the CMAH gene was confirmed by sequencing, showing lack of expression of CMAH in tail-derived fibroblasts of the cloned piglets. Besides decreased expression of Neu5Gc in piglets produced by SCNT, antibody-mediated complement-dependent cytotoxicity assays and natural antibody binding for examining immuno-reactivity of the quadruple gene modified pigs derived from endothelial cells and fibroblasts were reduced significantly compared to those of wild type animals. We conclude that by combining the TALEN system and transgenic cells, targeting of multiple genes could be useful for generating organs for xenotransplantation. We produced miniature pigs with quadruple modified genes CMAHKO/GTKO/shTNFRI-Fc/hHO-1 that will be suitable for xenotransplantation by overcoming hyperacute, acute and anti-inflammatory rejection.
Collapse
|
135
|
Song M. The CRISPR/Cas9 system: Their delivery, in vivo and ex vivo applications and clinical development by startups. Biotechnol Prog 2017; 33:1035-1045. [PMID: 28440027 DOI: 10.1002/btpr.2484] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 04/09/2017] [Indexed: 12/23/2022]
Abstract
The CRISPR/Cas9 gene editing system was originally derived from the prokaryotic adaptive immune system mediated by Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR) and CRISPR-associated proteins (Cas). The system has been successfully applied to genome editing in eukaryotes and has contributed to remarkable advances in the life sciences, in areas ranging from agriculture to genetic disease therapies. For efficient editing and extending the influence of this system, proper delivery of its components is crucial. Both viral and nonviral delivery methods are reviewed here, along with the advantages and disadvantages of each. In addition, we review ex vivo and in vivo CRISPR/Cas9 applications for disease therapies. Related remarkable studies are highlighted and relevant startup companies and their drug development pipelines are described. © 2017 American Institute of Chemical Engineers Biotechnol. Prog., 33:1035-1045, 2017.
Collapse
Affiliation(s)
- Minjung Song
- Dept. of Food Biotechnology, Silla University, Baekyangdaero 700, Sasang-ku, Busan, Korea
| |
Collapse
|
136
|
Kamoshita M, Kato T, Fujiwara K, Namiki T, Matsumura K, Hyon SH, Ito J, Kashiwazaki N. Successful vitrification of pronuclear-stage pig embryos with a novel cryoprotective agent, carboxylated ε-poly-L-lysine. PLoS One 2017; 12:e0176711. [PMID: 28448636 PMCID: PMC5407792 DOI: 10.1371/journal.pone.0176711] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Accepted: 04/15/2017] [Indexed: 11/18/2022] Open
Abstract
Vitrification is a powerful tool for the efficient production of offspring derived from cryopreserved oocytes or embryos in mammalian species including domestic animals. Genome editing technologies such as transcription activator-like effector nucleases (TALENs) and clustered regularly interspaced short palindromic repeats (CRISPR)/ CRISPR-associated (Cas)9 are now available even for domestic species, suggesting that the vitrification of embryos at the pronuclear stage (PN) will be more important because they could provide genomic host cells to be targeted by TALENs or CRISPR/Cas9. Although we reported the successful production of piglets derived from vitrified PN embryos by a solid-surface vitrification method with glutathione supplementation, further improvements are required. The cryoprotective agent (CPA) carboxylated ε-poly-L-lysine (COOH-PLL) was introduced in 2009. COOH-PLL reduces the physical and physiological damage caused by cryopreservation in mammalian stem cells and the vitrification of mouse oocytes and embryos. Those results suggested that vitrification of COOH-PLL may help improve the developmental ability of pig embryos vitrified at the PN stage. However, it remains unclear whether COOH-PLL is available as a CPA for the vitrification of embryos in domestic species. In this study, we evaluated COOH-PLL as a CPA with ethylene glycol (EG) and Cryotop as a device for the vitrification of PN pig embryos. Exposure to vitrification solution supplemented with COOH-PLL up to 30% did not decrease developmental ability to the 2-cell stage and the blastocyst stage. After warming, most of the vitrified embryos survived regardless of the concentration of COOH-PLL (76.0 ± 11.8% to 91.8 ± 4.6%). However, the vitrified embryos without COOH-PLL showed a lower development rate up to the blastocyst stage (1.3 ± 1.0%) compared to the fresh embryos (28.4 ± 5.0%) (p<0.05). In contrast, supplementation of 20% (w/v) COOH-PLL in the vitrification solution dramatically improved the developmental ability to blastocysts of the vitrified embryos (19.4 ± 4.6%) compared to those without COOH-PLL (p<0.05). After the transfer of embryos vitrified with 30% (v/v) EG and 20% (w/v) COOH-PLL, we successfully obtained 15 piglets from 8 recipients. Taken together, our present findings demonstrate for the first time that COOH-PLL is an effective CPA for embryo vitrification in the pig. COOH-PLL is a promising CPA for further improvements in the vitrification of oocytes and embryos in mammalian species.
Collapse
Affiliation(s)
- Maki Kamoshita
- Laboratory of Animal Reproduction, Graduate School of Veterinary Science, Azabu University, Sagamihara, Japan
| | - Tsubasa Kato
- Laboratory of Animal Reproduction, Graduate School of Veterinary Science, Azabu University, Sagamihara, Japan
| | - Katsuyoshi Fujiwara
- Laboratory of Animal Reproduction, Graduate School of Veterinary Science, Azabu University, Sagamihara, Japan
| | - Takafumi Namiki
- Laboratory of Animal Reproduction, Graduate School of Veterinary Science, Azabu University, Sagamihara, Japan
| | | | - Suong-Hyu Hyon
- Center for Fiber and Textile Science, Kyoto Institute of Technology, Matsugasaki, Kyoto, Japan
| | - Junya Ito
- Laboratory of Animal Reproduction, Graduate School of Veterinary Science, Azabu University, Sagamihara, Japan
- School of Veterinary Medicine, Azabu University, Sagamihara, Japan
| | - Naomi Kashiwazaki
- Laboratory of Animal Reproduction, Graduate School of Veterinary Science, Azabu University, Sagamihara, Japan
- School of Veterinary Medicine, Azabu University, Sagamihara, Japan
| |
Collapse
|
137
|
Affiliation(s)
- Tetsuya Ishii
- Office of Health and Safety, Hokkaido University, Sapporo 060-0808, Hokkaido, Japan
| |
Collapse
|
138
|
Temperature effect on CRISPR-Cas9 mediated genome editing. J Genet Genomics 2017; 44:199-205. [PMID: 28412228 DOI: 10.1016/j.jgg.2017.03.004] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Revised: 02/15/2017] [Accepted: 03/06/2017] [Indexed: 11/22/2022]
Abstract
Zinc-finger nuclease (ZFN), transcription activator-like effector nuclease (TALEN), and clustered regularly interspaced short palindromic repeats/CRISPR-associated protein 9 (CRISPR-Cas9) are the most commonly used genome editing tools. Previous studies demonstrated that hypothermia treatment increased the mutation rates induced by ZFNs and TALENs in mammalian cells. Here, we characterize the effect of different culture temperatures on CRISPR-Cas9 mediated genome editing and find that the genome editing efficiency of CRISPR-Cas9 is significantly hampered by hypothermia treatment, unlike ZFN and TALEN. In addition, hyperthermia culture condition enhances genome editing by CRISPR-Cas9 in some cell lines, due to the higher enzyme activity and sgRNA expression level at higher temperature. Our study has implications on CRISPR-Cas9 applications in a broad spectrum of species, many of which do not live at 37°C.
Collapse
|
139
|
Genome editing using FACS enrichment of nuclease-expressing cells and indel detection by amplicon analysis. Nat Protoc 2017; 12:581-603. [PMID: 28207001 DOI: 10.1038/nprot.2016.165] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
This protocol describes methods for increasing and evaluating the efficiency of genome editing based on the CRISPR-Cas9 (clustered regularly interspaced short palindromic repeats-CRISPR-associated 9) system, transcription activator-like effector nucleases (TALENs) or zinc-finger nucleases (ZFNs). First, Indel Detection by Amplicon Analysis (IDAA) determines the size and frequency of insertions and deletions elicited by nucleases in cells, tissues or embryos through analysis of fluorophore-labeled PCR amplicons covering the nuclease target site by capillary electrophoresis in a sequenator. Second, FACS enrichment of cells expressing nucleases linked to fluorescent proteins can be used to maximize knockout or knock-in editing efficiencies or to balance editing efficiency and toxic/off-target effects. The two methods can be combined to form a pipeline for cell-line editing that facilitates the testing of new nuclease reagents and the generation of edited cell pools or clonal cell lines, reducing the number of clones that need to be generated and increasing the ease with which they are screened. The pipeline shortens the time line, but it most prominently reduces the workload of cell-line editing, which may be completed within 4 weeks.
Collapse
|
140
|
Knipping F, Osborn MJ, Petri K, Tolar J, Glimm H, von Kalle C, Schmidt M, Gabriel R. Genome-wide Specificity of Highly Efficient TALENs and CRISPR/Cas9 for T Cell Receptor Modification. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2017; 4:213-224. [PMID: 28345006 PMCID: PMC5363317 DOI: 10.1016/j.omtm.2017.01.005] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Accepted: 01/25/2017] [Indexed: 12/11/2022]
Abstract
In T cells with transgenic high-avidity T cell receptors (TCRs), endogenous and transferred TCR chains compete for surface expression and may pair inappropriately, potentially causing autoimmunity. To knock out endogenous TCR expression, we assembled 12 transcription activator-like effector nucleases (TALENs) and five guide RNAs (gRNAs) from the clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated (Cas9) system. Using TALEN mRNA, TCR knockout was successful in up to 81% of T cells. Additionally, we were able to verify targeted gene addition of a GFP gene by homology-directed repair at the TALEN target site, using a donor suitable for replacement of the reporter transgene with therapeutic TCR chains. Remarkably, analysis of TALEN and CRISPR/Cas9 specificity using integrase-defective lentiviral vector capture revealed only one off-target site for one of the gRNAs and three off-target sites for both of the TALENs, indicating a high level of specificity. Collectively, our work shows highly efficient and specific nucleases for T cell engineering.
Collapse
Affiliation(s)
- Friederike Knipping
- Department of Translational Oncology, National Center for Tumor Diseases and German Cancer Research Center, 69120 Heidelberg, Germany
| | - Mark J Osborn
- Division of Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA; Center for Genome Engineering, University of Minnesota, Minneapolis, MN 55455, USA; Stem Cell Institute, University of Minnesota, Minneapolis, MN 55455, USA; Asan-Minnesota Institute for Innovating Transplantation, Seoul 05505, Republic of Korea
| | - Karl Petri
- Department of Translational Oncology, National Center for Tumor Diseases and German Cancer Research Center, 69120 Heidelberg, Germany
| | - Jakub Tolar
- Division of Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA; Center for Genome Engineering, University of Minnesota, Minneapolis, MN 55455, USA; Stem Cell Institute, University of Minnesota, Minneapolis, MN 55455, USA; Asan-Minnesota Institute for Innovating Transplantation, Seoul 05505, Republic of Korea
| | - Hanno Glimm
- Department of Translational Oncology, National Center for Tumor Diseases and German Cancer Research Center, 69120 Heidelberg, Germany
| | - Christof von Kalle
- Department of Translational Oncology, National Center for Tumor Diseases and German Cancer Research Center, 69120 Heidelberg, Germany
| | - Manfred Schmidt
- Department of Translational Oncology, National Center for Tumor Diseases and German Cancer Research Center, 69120 Heidelberg, Germany
| | - Richard Gabriel
- Department of Translational Oncology, National Center for Tumor Diseases and German Cancer Research Center, 69120 Heidelberg, Germany
| |
Collapse
|
141
|
Taylor L, Carlson DF, Nandi S, Sherman A, Fahrenkrug SC, McGrew MJ. Efficient TALEN-mediated gene targeting of chicken primordial germ cells. Development 2017; 144:928-934. [PMID: 28174243 PMCID: PMC5374353 DOI: 10.1242/dev.145367] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2016] [Accepted: 01/12/2017] [Indexed: 12/28/2022]
Abstract
In this work we use TALE nucleases (TALENs) to target a reporter construct to the DDX4 (vasa) locus in chicken primordial germ cells (PGCs). Vasa is a key germ cell determinant in many animal species and is posited to control avian germ cell formation. We show that TALENs mediate homology-directed repair of the DDX4 locus on the Z sex chromosome at high (8.1%) efficiencies. Large genetic deletions of 30 kb encompassing the entire DDX4 locus were also created using a single TALEN pair. The targeted PGCs were germline competent and were used to produce DDX4 null offspring. In DDX4 knockout chickens, PGCs are initially formed but are lost during meiosis in the developing ovary, leading to adult female sterility. TALEN-mediated gene targeting in avian PGCs is therefore an efficient process. Summary: TALE nucleases are used to target the DDX4 (vasa) locus in chicken primordial germ cells and generate DDX4 knockouts, which provide insights into DDX4 function in early chick development.
Collapse
Affiliation(s)
- Lorna Taylor
- The Roslin Institute and Royal Dick School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, Midlothian EH25 9RG, UK
| | - Daniel F Carlson
- Recombinetics Inc, 1246 University Avenue West, Suite 300, Saint Paul, MN 55104, USA
| | - Sunil Nandi
- The Roslin Institute and Royal Dick School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, Midlothian EH25 9RG, UK
| | - Adrian Sherman
- The Roslin Institute and Royal Dick School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, Midlothian EH25 9RG, UK
| | - Scott C Fahrenkrug
- Recombinetics Inc, 1246 University Avenue West, Suite 300, Saint Paul, MN 55104, USA
| | - Michael J McGrew
- The Roslin Institute and Royal Dick School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, Midlothian EH25 9RG, UK
| |
Collapse
|
142
|
Schomberg DT, Miranpuri GS, Chopra A, Patel K, Meudt JJ, Tellez A, Resnick DK, Shanmuganayagam D. Translational Relevance of Swine Models of Spinal Cord Injury. J Neurotrauma 2017; 34:541-551. [DOI: 10.1089/neu.2016.4567] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Affiliation(s)
- Dominic T. Schomberg
- Biomedical and Genomic Research Group, Department of Animal Sciences, University of Wisconsin–Madison, Wisconsin
| | - Gurwattan S. Miranpuri
- Department of Neurological Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Abhishek Chopra
- Department of Neurological Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Kush Patel
- Department of Neurological Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Jennifer J. Meudt
- Biomedical and Genomic Research Group, Department of Animal Sciences, University of Wisconsin–Madison, Wisconsin
| | | | - Daniel K. Resnick
- Department of Neurological Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Dhanansayan Shanmuganayagam
- Biomedical and Genomic Research Group, Department of Animal Sciences, University of Wisconsin–Madison, Wisconsin
| |
Collapse
|
143
|
Abstract
In the past few years, new technologies have arisen that enable higher efficiency of gene editing. With the increase ease of using gene editing technologies, it is important to consider the best method for transferring new genetic material to livestock animals. Microinjection is a technique that has proven to be effective in mice but is less efficient in large livestock animals. Over the years, a variety of methods have been used for cloning as well as gene transfer including; nuclear transfer, sperm mediated gene transfer (SMGT), and liposome-mediated DNA transfer. This review looks at the different success rate of these methods and how they have evolved to become more efficient. As well as gene editing technologies, including Zinc-finger nucleases (ZFNs), transcription activator-like effector nucleases (TALENs), and the most recent clustered regulatory interspaced short palindromic repeats (CRISPRs). Through the advancements in gene-editing technologies, generating transgenic animals is now more accessible and affordable. The goals of producing transgenic animals are to 1) increase our understanding of biology and biomedical science; 2) increase our ability to produce more efficient animals; and 3) produce disease resistant animals. ZFNs, TALENs, and CRISPRs combined with gene transfer methods increase the possibility of achieving these goals.
Collapse
Key Words
- BLG, β-lactoglobulin
- CRISPR
- CRISPRs, clustered regulatory interspaced short palindromic repeats
- EG, embryonic germ
- ES, Embryonic stem
- ESC, Embryonic stem cell
- HDR, homology directed repair
- ICM, inner cell mass
- ICSI, intracytoplasmic sperm injection
- NHEJ, non-homologous end joining
- NT, nuclear transfers
- OBCT, oocyte bisection technique
- PAM, protospacer adjacent motif
- PCR, polymerase chain reaction
- PGCS, primordial germ cells
- RVDs, repeat variable diresidues
- SMGT
- SMGT, sperm mediated gene transfer
- SV40, simian virus 40
- TALEN
- TALENs, transcription activator-like effector nucleases
- ZFN
- ZFN, Zinc-finger nucleases
- gene editing
- gene transfer
- iPSC, induced pluripotent stem cells
- nuclear transfer
- ssODN, single strand oligo nucleotide
Collapse
Affiliation(s)
- Samantha N Lotti
- a Department of Animal Sciences , University of Illinois at Urbana-Champaign , Urbana , Illinois , USA
| | - Kathryn M Polkoff
- a Department of Animal Sciences , University of Illinois at Urbana-Champaign , Urbana , Illinois , USA
| | - Marcello Rubessa
- b Carl R. Woese Institute for Genomic Biology, University of Illinois , Urbana , IL , USA
| | - Matthew B Wheeler
- a Department of Animal Sciences , University of Illinois at Urbana-Champaign , Urbana , Illinois , USA.,b Carl R. Woese Institute for Genomic Biology, University of Illinois , Urbana , IL , USA
| |
Collapse
|
144
|
Park KE, Kaucher AV, Powell A, Waqas MS, Sandmaier SES, Oatley MJ, Park CH, Tibary A, Donovan DM, Blomberg LA, Lillico SG, Whitelaw CBA, Mileham A, Telugu BP, Oatley JM. Generation of germline ablated male pigs by CRISPR/Cas9 editing of the NANOS2 gene. Sci Rep 2017; 7:40176. [PMID: 28071690 PMCID: PMC5223215 DOI: 10.1038/srep40176] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Accepted: 12/02/2016] [Indexed: 12/21/2022] Open
Abstract
Genome editing tools have revolutionized the generation of genetically modified animals including livestock. In particular, the domestic pig is a proven model of human physiology and an agriculturally important species. In this study, we utilized the CRISPR/Cas9 system to edit the NANOS2 gene in pig embryos to generate offspring with mono-allelic and bi-allelic mutations. We found that NANOS2 knockout pigs phenocopy knockout mice with male specific germline ablation but other aspects of testicular development are normal. Moreover, male pigs with one intact NANOS2 allele and female knockout pigs are fertile. From an agriculture perspective, NANOS2 knockout male pigs are expected to serve as an ideal surrogate for transplantation of donor spermatogonial stem cells to expand the availability of gametes from genetically desirable sires.
Collapse
Affiliation(s)
- Ki-Eun Park
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD 20742, USA.,Animal Bioscience and Biotechnology Laboratory, USDA, ARS, Beltsville, MD 20705, USA.,Renovate Biosciences Inc, Riesterstown, MD, USA
| | - Amy V Kaucher
- Center for Reproductive Biology, College of Veterinary Medicine, Washington State University, Pullman, WA 99164, USA
| | - Anne Powell
- Animal Bioscience and Biotechnology Laboratory, USDA, ARS, Beltsville, MD 20705, USA
| | - Muhammad Salman Waqas
- Center for Reproductive Biology, College of Veterinary Medicine, Washington State University, Pullman, WA 99164, USA
| | - Shelley E S Sandmaier
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD 20742, USA.,Animal Bioscience and Biotechnology Laboratory, USDA, ARS, Beltsville, MD 20705, USA
| | - Melissa J Oatley
- Center for Reproductive Biology, College of Veterinary Medicine, Washington State University, Pullman, WA 99164, USA
| | - Chi-Hun Park
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD 20742, USA.,Animal Bioscience and Biotechnology Laboratory, USDA, ARS, Beltsville, MD 20705, USA
| | - Ahmed Tibary
- Center for Reproductive Biology, College of Veterinary Medicine, Washington State University, Pullman, WA 99164, USA
| | - David M Donovan
- Animal Bioscience and Biotechnology Laboratory, USDA, ARS, Beltsville, MD 20705, USA
| | - Le Ann Blomberg
- Animal Bioscience and Biotechnology Laboratory, USDA, ARS, Beltsville, MD 20705, USA
| | - Simon G Lillico
- Roslin Institute, University of Edinburgh, Edinburgh, Scotland
| | | | | | - Bhanu P Telugu
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD 20742, USA.,Animal Bioscience and Biotechnology Laboratory, USDA, ARS, Beltsville, MD 20705, USA.,Renovate Biosciences Inc, Riesterstown, MD, USA
| | - Jon M Oatley
- Center for Reproductive Biology, College of Veterinary Medicine, Washington State University, Pullman, WA 99164, USA
| |
Collapse
|
145
|
Polejaeva IA, Rutigliano HM, Wells KD. Livestock in biomedical research: history, current status and future prospective. Reprod Fertil Dev 2017; 28:112-24. [PMID: 27062879 DOI: 10.1071/rd15343] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Livestock models have contributed significantly to biomedical and surgical advances. Their contribution is particularly prominent in the areas of physiology and assisted reproductive technologies, including understanding developmental processes and disorders, from ancient to modern times. Over the past 25 years, biomedical research that traditionally embraced a diverse species approach shifted to a small number of model species (e.g. mice and rats). The initial reasons for focusing the main efforts on the mouse were the availability of murine embryonic stem cells (ESCs) and genome sequence data. This powerful combination allowed for precise manipulation of the mouse genome (knockouts, knockins, transcriptional switches etc.) leading to ground-breaking discoveries on gene functions and regulation, and their role in health and disease. Despite the enormous contribution to biomedical research, mouse models have some major limitations. Their substantial differences compared with humans in body and organ size, lifespan and inbreeding result in pronounced metabolic, physiological and behavioural differences. Comparative studies of strategically chosen domestic species can complement mouse research and yield more rigorous findings. Because genome sequence and gene manipulation tools are now available for farm animals (cattle, pigs, sheep and goats), a larger number of livestock genetically engineered (GE) models will be accessible for biomedical research. This paper discusses the use of cattle, goats, sheep and pigs in biomedical research, provides an overview of transgenic technology in farm animals and highlights some of the beneficial characteristics of large animal models of human disease compared with the mouse. In addition, status and origin of current regulation of GE biomedical models is also reviewed.
Collapse
Affiliation(s)
- Irina A Polejaeva
- Department of Animal, Dairy and Veterinary Sciences, Utah State University, Logan, UT 84322, USA
| | - Heloisa M Rutigliano
- Department of Animal, Dairy and Veterinary Sciences, Utah State University, Logan, UT 84322, USA
| | - Kevin D Wells
- Division of Animal Sciences, Animal Sciences Research Center, University of Missouri, Columbia, MO 65211, USA
| |
Collapse
|
146
|
Josa S, Seruggia D, Fernández A, Montoliu L. Concepts and tools for gene editing. Reprod Fertil Dev 2017; 29:1-7. [DOI: 10.1071/rd16396] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Gene editing is a relatively recent concept in the molecular biology field. Traditional genetic modifications in animals relied on a classical toolbox that, aside from some technical improvements and additions, remained unchanged for many years. Classical methods involved direct delivery of DNA sequences into embryos or the use of embryonic stem cells for those few species (mice and rats) where it was possible to establish them. For livestock, the advent of somatic cell nuclear transfer platforms provided alternative, but technically challenging, approaches for the genetic alteration of loci at will. However, the entire landscape changed with the appearance of different classes of genome editors, from initial zinc finger nucleases, to transcription activator-like effector nucleases and, most recently, with the development of clustered regularly interspaced short palindromic repeats (CRISPR) and CRISPR-associated proteins (Cas). Gene editing is currently achieved by CRISPR–Cas-mediated methods, and this technological advancement has boosted our capacity to generate almost any genetically altered animal that can be envisaged.
Collapse
|
147
|
Kang JD, Kim S, Zhu HY, Jin L, Guo Q, Li XC, Zhang YC, Xing XX, Xuan MF, Zhang GL, Luo QR, Kim YS, Cui CD, Li WX, Cui ZY, Kim JS, Yin XJ. Generation of cloned adult muscular pigs with myostatin gene mutation by genetic engineering. RSC Adv 2017. [DOI: 10.1039/c6ra28579a] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Skeletal muscle is the most economically valuable tissue in meat-producing animals and enhancing muscle growth in these species may enhance the efficiency of meat production.
Collapse
|
148
|
Abstract
Pigs are important livestock for food and have been used in various biomedical studies, particularly translational research, as experimental animals because of their anatomical and physiological similarity to humans. The recent development of genome editing techniques, such as ZFN, TALEN, and CRISPR/Cas9, has rapidly expanded the use of genome editing tools in a variety of animals, resulting in the relatively easy and efficient generation of gene knock-out pigs. In the past few years, there has been a sustained increase in reports describing the development of genetically modified pigs. This chapter introduces our workflow for establishing the genetically modified cells (nuclear donor cells) necessary to create gene knock-out pigs using somatic cell nuclear transfer and focuses on the actual generation of gene knock-out pigs using a cytoplasmic injection method.
Collapse
Affiliation(s)
- Masahito Watanabe
- Meiji University International Institute for Bio-Resource Research, Kawasaki, 214-8571, Japan.,Laboratory of Developmental Engineering, Department of Life Sciences, School of Agriculture, Meiji University, Kawasaki, 214-8571, Japan
| | - Hiroshi Nagashima
- Meiji University International Institute for Bio-Resource Research, Kawasaki, 214-8571, Japan. .,Laboratory of Developmental Engineering, Department of Life Sciences, School of Agriculture, Meiji University, Kawasaki, 214-8571, Japan.
| |
Collapse
|
149
|
Yuan YG, Song SZ, Zhu MM, He ZY, Lu R, Zhang T, Mi F, Wang JY, Cheng Y. Human lactoferrin efficiently targeted into caprine beta-lactoglobulin locus with transcription activator-like effector nucleases. ASIAN-AUSTRALASIAN JOURNAL OF ANIMAL SCIENCES 2016; 30:1175-1182. [PMID: 28002927 PMCID: PMC5494492 DOI: 10.5713/ajas.16.0697] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Revised: 11/11/2016] [Accepted: 12/13/2016] [Indexed: 02/06/2023]
Abstract
Objective To create genetically modified goat as a biopharming source of recombinant human lacotoferrin (hLF) with transcription activator-like effector nucleases. Methods TALENs and targeting vector were transferred into cultured fibroblasts to insert hLF cDNA in the goat beta-lactoglobulin (BLG) locus with homology-directed repair. The gene targeted efficiency was checked using sequencing and TE7I assay. The bi-allelic gene targeted colonies were isolated and confirmed with polymerase chain reaction, and used as donor cells for somatic cell nuclear transfer (SCNT). Results The targeted efficiency for BLG gene was approximately 10%. Among 12 Bi-allelic gene targeted colonies, five were used in first round SCNT and 4 recipients (23%) were confirmed pregnant at 30 d. In second round SCNT, 7 (53%), 4 (31%), and 3 (23%) recipients were confirmed to be pregnant by ultrasound on 30 d, 60 d, and 90 d. Conclusion This finding signifies the combined use of TALENs and SCNT can generate bi-allelic knock-in fibroblasts that can be cloned in a fetus. Therefore, it might lay the foundation for transgenic hLF goat generation and possible use of their mammary gland as a bioreactor for large-scale production of recombinant hLF.
Collapse
Affiliation(s)
- Yu-Guo Yuan
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis/College of animal science and technology, Yangzhou University, Yangzhou, Jiangsu 225009, China.,Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou, 225001, China
| | - Shao-Zheng Song
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis/College of animal science and technology, Yangzhou University, Yangzhou, Jiangsu 225009, China
| | - Meng-Ming Zhu
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis/College of animal science and technology, Yangzhou University, Yangzhou, Jiangsu 225009, China
| | - Zheng-Yi He
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis/College of animal science and technology, Yangzhou University, Yangzhou, Jiangsu 225009, China
| | - Rui Lu
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis/College of animal science and technology, Yangzhou University, Yangzhou, Jiangsu 225009, China
| | - Ting Zhang
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis/College of animal science and technology, Yangzhou University, Yangzhou, Jiangsu 225009, China
| | - Fei Mi
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis/College of animal science and technology, Yangzhou University, Yangzhou, Jiangsu 225009, China
| | - Jin-Yu Wang
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis/College of animal science and technology, Yangzhou University, Yangzhou, Jiangsu 225009, China
| | - Yong Cheng
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis/College of animal science and technology, Yangzhou University, Yangzhou, Jiangsu 225009, China.,Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou, 225001, China
| |
Collapse
|
150
|
Wang Y, Du Y, Zhou X, Wang L, Li J, Wang F, Huang Z, Huang X, Wei H. Efficient generation of B2m-null pigs via injection of zygote with TALENs. Sci Rep 2016; 6:38854. [PMID: 27982048 PMCID: PMC5159787 DOI: 10.1038/srep38854] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2015] [Accepted: 11/14/2016] [Indexed: 02/07/2023] Open
Abstract
Donor major histocompatibility complex class I (MHC I) molecules are the main targets of the host immune response after organ allotransplantation. Whether and how MHC I-deficiency of pig donor tissues affects rejection after xenotransplantation has not been assessed. Beta2-microglobulin (B2M) is indispensable for the assembly of MHC I receptors and therefore provides an effective target to disrupt cell surface MHC I expression. Here, we report the one-step generation of mutant pigs with targeted disruptions in B2m by injection of porcine zygotes with B2m exon 2-specific TALENs. After germline transmission of mutant B2m alleles, we obtained F1 pigs with biallelic B2m frameshift mutations. F1 pigs lacked detectable B2M expression in tissues derived from the three germ layers, and their lymphocytes were devoid of MHC I surface receptors. Skin grafts from B2M deficient pigs exhibited remarkably prolonged survival on xenogeneic wounds compared to tissues of non-mutant littermates. Mutant founder pigs with bi-allelic disruption in B2m and B2M deficient F1 offspring did not display visible abnormalities, suggesting that pigs are tolerant to B2M deficiency. In summary, we show the efficient generation of pigs with germline mutations in B2m, and demonstrate a beneficial effect of donor MHC I-deficiency on xenotransplantation.
Collapse
Affiliation(s)
- Yong Wang
- Department of Laboratory Animal Science, College of Basic Medical Sciences, Third Military Medical University, Chongqing 400038, China
| | - Yinan Du
- MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center of Nanjing University, National Resource Center for Mutant Mice, Nanjing 210061, China.,School of Life Science and Technology, ShanghaiTech University, 100 Haike Rd., Pudong New Area, Shanghai 201210, China
| | - Xiaoyang Zhou
- Department of Laboratory Animal Science, College of Basic Medical Sciences, Third Military Medical University, Chongqing 400038, China
| | - Lulu Wang
- Department of Laboratory Animal Science, College of Basic Medical Sciences, Third Military Medical University, Chongqing 400038, China
| | - Jian Li
- Department of Immunology, College of Basic Medical Sciences, Third Military Medical University, Chongqing 400038, China
| | - Fengchao Wang
- Institute of Combined Injury, College of Military Preventive Medicine, Third Military Medical University, Chongqing 400038, China
| | - Zhengen Huang
- Research Institute of Burns, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Xingxu Huang
- MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center of Nanjing University, National Resource Center for Mutant Mice, Nanjing 210061, China.,School of Life Science and Technology, ShanghaiTech University, 100 Haike Rd., Pudong New Area, Shanghai 201210, China
| | - Hong Wei
- Department of Laboratory Animal Science, College of Basic Medical Sciences, Third Military Medical University, Chongqing 400038, China
| |
Collapse
|