1
|
Rodriguez-Villamil P, Beaton BP, Krisher RL. Gene editing in livestock: innovations and applications. Anim Reprod 2024; 21:e20240054. [PMID: 39372257 PMCID: PMC11452096 DOI: 10.1590/1984-3143-ar2024-0054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 08/05/2024] [Indexed: 10/08/2024] Open
Abstract
Gene editing technologies have revolutionized the field of livestock breeding, offering unprecedented opportunities to enhance animal welfare, productivity, and sustainability. This paper provides a comprehensive review of recent innovations and applications of gene editing in livestock, exploring the diverse applications of gene editing in livestock breeding, as well as the regulatory and ethical considerations, and the current challenges and prospects of the technology in the industry. Overall, this review underscores the transformative potential of gene editing in livestock breeding and its pivotal role in shaping the future of agriculture and biomedicine.
Collapse
|
2
|
Su H, Zhi D, Song Y, Yang Y, Wang D, Li X, Cao G. Exploring the formation mechanism of short-tailed phenotypes in animals using mutant mice with the TBXT gene c.G334T developed by CRISPR/Cas9. Gene 2024; 910:148310. [PMID: 38401832 DOI: 10.1016/j.gene.2024.148310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Revised: 02/01/2024] [Accepted: 02/21/2024] [Indexed: 02/26/2024]
Abstract
With the change in diet structure, individuals prefer to consume mutton with less fat. However, sheep tail has a lot of fat. We identified a breed of low-fat short-tailed sheep (i.e., Hulunbuir short-tailed sheep). It is necessary to develop an animal model that can promote research on the potential mechanisms of the short-tail phenotype in sheep, which results from the TBXT gene c.G334T mutation. To create animal models, we selected mice as experimental animals. Mouse embryos lacking the TBXT protein, which crucially regulates mouse embryonic development, cannot develop normally. We utilized CRISPR/Cas9 gene editing technology to generate site-specific mutation (c.G334T) in the TBXT gene of mice, and found that the mouse TBXT mutation (c.G334T) leads to a short-tail phenotype. Furthermore, we investigated the interaction between TBXT and Wnt signaling pathways. The expressions of TBXT, Axin2, Dkk1, Wnt3, Wnt3a, and Wnt5a were discovered to be significantly different between mutant embryos and wild embryos by obtaining mouse embryos at various developmental stages and examining the expression relationship between the TBXT and Wnt signaling pathway-related components in all of these embryos. Therefore, as a transcription factor, TBXT regulates the expression of the aforementioned Wnt signaling pathway components by forming a regulatory network for the normal development of mouse embryos. This study enriches the research on the functional role of the TBXT in the development of mouse embryos and the mechanism by which the short-tailed phenotype in sheep develops.
Collapse
Affiliation(s)
- Hong Su
- College of Veterinary Medicine, Inner Mongolia Agricultural University, China; Animal Embryo and Developmental Engineering Key Laboratory of Higher Education, Institutions of Inner Mongolia Autonomous Region, China; Inner Mongolia Autonomous Region Key Laboratory of Basic Veterinary Medicine, China.
| | - Dafu Zhi
- College of Veterinary Medicine, Inner Mongolia Agricultural University, China; Animal Embryo and Developmental Engineering Key Laboratory of Higher Education, Institutions of Inner Mongolia Autonomous Region, China; Inner Mongolia Autonomous Region Key Laboratory of Basic Veterinary Medicine, China.
| | - Yongli Song
- College of Life Sciences, Inner Mongolia University, China.
| | - Yanyan Yang
- Inner Mongolia Academy of Agriculture and Animal Husbandry, China.
| | - Daqing Wang
- College of Veterinary Medicine, Inner Mongolia Agricultural University, China; Animal Embryo and Developmental Engineering Key Laboratory of Higher Education, Institutions of Inner Mongolia Autonomous Region, China; Inner Mongolia Autonomous Region Key Laboratory of Basic Veterinary Medicine, China; Inner Mongolia Academy of Agriculture and Animal Husbandry, China.
| | - Xiunan Li
- College of Veterinary Medicine, Inner Mongolia Agricultural University, China; Animal Embryo and Developmental Engineering Key Laboratory of Higher Education, Institutions of Inner Mongolia Autonomous Region, China; Inner Mongolia Autonomous Region Key Laboratory of Basic Veterinary Medicine, China; Inner Mongolia Academy of Agriculture and Animal Husbandry, China.
| | - Guifang Cao
- College of Veterinary Medicine, Inner Mongolia Agricultural University, China; Animal Embryo and Developmental Engineering Key Laboratory of Higher Education, Institutions of Inner Mongolia Autonomous Region, China; Inner Mongolia Autonomous Region Key Laboratory of Basic Veterinary Medicine, China.
| |
Collapse
|
3
|
Briski O, La Motta GE, Ratner LD, Allegroni FA, Pillado S, Álvarez G, Gutierrez B, Tarragona L, Zaccagnini A, Acerbo M, Ciampi C, Fernández-Martin R, Salamone DF. Comparison of ICSI, IVF, and in vivo derived embryos to produce CRISPR-Cas9 gene-edited pigs for xenotransplantation. Theriogenology 2024; 220:43-55. [PMID: 38471390 DOI: 10.1016/j.theriogenology.2024.02.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 02/26/2024] [Accepted: 02/26/2024] [Indexed: 03/14/2024]
Abstract
Genome editing in pigs for xenotransplantation has seen significant advances in recent years. This study compared three methodologies to generate gene-edited embryos, including co-injection of sperm together with the CRISPR-Cas9 system into oocytes, named ICSI-MGE (mediated gene editing); microinjection of CRISPR-Cas9 components into oocytes followed by in vitro fertilization (IVF), and microinjection of in vivo fertilized zygotes with the CRISPR-Cas9 system. Our goal was to knock-out (KO) porcine genes involved in the biosynthesis of xenoantigens responsible for the hyperacute rejection of interspecific xenografts, namely GGTA1, CMAH, and β4GalNT2. Additionally, we attempted to KO the growth hormone receptor (GHR) gene with the aim of limiting the growth of porcine organs to a size that is physiologically suitable for human transplantation. Embryo development, pregnancy, and gene editing rates were evaluated. We found an efficient mutation of the GGTA1 gene following ICSI-MGE, comparable to the results obtained through the microinjection of oocytes followed by IVF. ICSI-MGE also showed higher rates of biallelic mutations compared to the other techniques. Five healthy piglets were born from in vivo-derived embryos, all of them exhibiting biallelic mutations in the GGTA1 gene, with three displaying mutations in the GHR gene. No mutations were observed in the CMAH and β4GalNT2 genes. In conclusion, in vitro methodologies showed high rates of gene-edited embryos. Specifically, ICSI-MGE proved to be an efficient technique for obtaining homozygous biallelic mutated embryos. Lastly, only live births were obtained from in vivo-derived embryos showing efficient multiple gene editing for GGTA1 and GHR.
Collapse
Affiliation(s)
- Olinda Briski
- CONICET-Universidad de Buenos Aires - Instituto de Investigaciones en Producción Animal (INPA), Ciudad Autónoma de Buenos Aires, C1425FQB, Argentina; Facultad de Agronomía, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, C1417DSE, Argentina
| | - Gastón Emilio La Motta
- CONICET-Universidad de Buenos Aires - Instituto de Investigaciones en Producción Animal (INPA), Ciudad Autónoma de Buenos Aires, C1425FQB, Argentina
| | - Laura Daniela Ratner
- CONICET-Universidad de Buenos Aires - Instituto de Investigaciones en Producción Animal (INPA), Ciudad Autónoma de Buenos Aires, C1425FQB, Argentina; Facultad de Agronomía, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, C1417DSE, Argentina
| | - Federico Andrés Allegroni
- Facultad de Agronomía, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, C1417DSE, Argentina
| | - Santiago Pillado
- Facultad de Ciencias Veterinarias, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, C1417DSE, Argentina
| | - Guadalupe Álvarez
- Facultad de Ciencias Veterinarias, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, C1417DSE, Argentina
| | - Betiana Gutierrez
- Facultad de Ciencias Veterinarias, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, C1417DSE, Argentina
| | - Lisa Tarragona
- Facultad de Ciencias Veterinarias, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, C1417DSE, Argentina
| | - Andrea Zaccagnini
- Facultad de Ciencias Veterinarias, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, C1417DSE, Argentina
| | - Marcelo Acerbo
- Facultad de Ciencias Veterinarias, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, C1417DSE, Argentina
| | - Carla Ciampi
- CONICET-Universidad de Buenos Aires - Instituto de Investigaciones en Producción Animal (INPA), Ciudad Autónoma de Buenos Aires, C1425FQB, Argentina; Facultad de Agronomía, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, C1417DSE, Argentina
| | - Rafael Fernández-Martin
- CONICET-Universidad de Buenos Aires - Instituto de Investigaciones en Producción Animal (INPA), Ciudad Autónoma de Buenos Aires, C1425FQB, Argentina; Facultad de Agronomía, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, C1417DSE, Argentina.
| | - Daniel Felipe Salamone
- CONICET-Universidad de Buenos Aires - Instituto de Investigaciones en Producción Animal (INPA), Ciudad Autónoma de Buenos Aires, C1425FQB, Argentina; Facultad de Agronomía, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, C1417DSE, Argentina.
| |
Collapse
|
4
|
Ge W, Zhao X, Gou S, Jin Q, Chen F, Ouyang Z, Lai C, Cui T, Mai B, Lu S, Zhong K, Liang Y, Chen T, Wu H, Li N, Ye Y, Lai L, Wang K. Evaluation of guide-free Cas9-induced genomic damage and transcriptome changes in pig embryos. MOLECULAR THERAPY. NUCLEIC ACIDS 2023; 34:102035. [PMID: 37808924 PMCID: PMC10551558 DOI: 10.1016/j.omtn.2023.102035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 09/15/2023] [Indexed: 10/10/2023]
Abstract
Cas9 protein without sgRNAs can induce genomic damage at the cellular level in vitro. However, whether the detrimental effects occur in embryos after Cas9 treatment remains unknown. Here, using pig embryos as subjects, we observed that Cas9 protein transcribed from injected Cas9 mRNA can persist until at least the blastocyst stage. Cas9 protein alone can induce genome damage in preimplantation embryos, represented by the increased number of phosphorylated histone H2AX foci on the chromatin fiber, which led to apoptosis and decreased cell number of blastocysts. In addition, single-blastocyst RNA sequencing confirmed that Cas9 protein without sgRNAs can cause changes in the blastocyst transcriptome, depressing embryo development signal pathways, such as cell cycle, metabolism, and cellular communication-related signal pathways, while activating apoptosis and necroptosis signal pathways, which together resulted in impaired preimplantation embryonic development. These results indicated that attention should be given to the detrimental effects caused by the Cas9 protein when using CRISPR-Cas9 for germline genome editing, especially for the targeted correction of human pathological mutations using germline gene therapy.
Collapse
Affiliation(s)
- Weikai Ge
- China-New Zealand Joint Laboratory on Biomedicine and Health, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Centre for Regenerative Medicine and Health, Hong Kong Institute of Science and Innovation, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
- Research Unit of Generation of Large Animal Disease Models, Chinese Academy of Medical Sciences (2019RU015), Guangzhou 510530, China
- Sanya Institute of Swine Resource, Hainan Provincial Research Centre of Laboratory Animals, Sanya 572000, China
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, Wuyi University, Jiangmen 529020, China
| | - Xiaozhu Zhao
- China-New Zealand Joint Laboratory on Biomedicine and Health, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Centre for Regenerative Medicine and Health, Hong Kong Institute of Science and Innovation, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shixue Gou
- China-New Zealand Joint Laboratory on Biomedicine and Health, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Centre for Regenerative Medicine and Health, Hong Kong Institute of Science and Innovation, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
- Research Unit of Generation of Large Animal Disease Models, Chinese Academy of Medical Sciences (2019RU015), Guangzhou 510530, China
- Sanya Institute of Swine Resource, Hainan Provincial Research Centre of Laboratory Animals, Sanya 572000, China
| | - Qin Jin
- China-New Zealand Joint Laboratory on Biomedicine and Health, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Centre for Regenerative Medicine and Health, Hong Kong Institute of Science and Innovation, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
- Research Unit of Generation of Large Animal Disease Models, Chinese Academy of Medical Sciences (2019RU015), Guangzhou 510530, China
- Sanya Institute of Swine Resource, Hainan Provincial Research Centre of Laboratory Animals, Sanya 572000, China
| | - Fangbing Chen
- China-New Zealand Joint Laboratory on Biomedicine and Health, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Centre for Regenerative Medicine and Health, Hong Kong Institute of Science and Innovation, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
- Research Unit of Generation of Large Animal Disease Models, Chinese Academy of Medical Sciences (2019RU015), Guangzhou 510530, China
- Sanya Institute of Swine Resource, Hainan Provincial Research Centre of Laboratory Animals, Sanya 572000, China
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, Wuyi University, Jiangmen 529020, China
| | - Zhen Ouyang
- China-New Zealand Joint Laboratory on Biomedicine and Health, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Centre for Regenerative Medicine and Health, Hong Kong Institute of Science and Innovation, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
- Research Unit of Generation of Large Animal Disease Models, Chinese Academy of Medical Sciences (2019RU015), Guangzhou 510530, China
- Sanya Institute of Swine Resource, Hainan Provincial Research Centre of Laboratory Animals, Sanya 572000, China
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, Wuyi University, Jiangmen 529020, China
| | - Chengdan Lai
- China-New Zealand Joint Laboratory on Biomedicine and Health, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Centre for Regenerative Medicine and Health, Hong Kong Institute of Science and Innovation, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
- Research Unit of Generation of Large Animal Disease Models, Chinese Academy of Medical Sciences (2019RU015), Guangzhou 510530, China
- Sanya Institute of Swine Resource, Hainan Provincial Research Centre of Laboratory Animals, Sanya 572000, China
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, Wuyi University, Jiangmen 529020, China
| | - Tao Cui
- College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Baoyi Mai
- Sanya Institute of Swine Resource, Hainan Provincial Research Centre of Laboratory Animals, Sanya 572000, China
| | - Sijia Lu
- Sanya Institute of Swine Resource, Hainan Provincial Research Centre of Laboratory Animals, Sanya 572000, China
| | - Kexin Zhong
- Sanya Institute of Swine Resource, Hainan Provincial Research Centre of Laboratory Animals, Sanya 572000, China
| | - Yanhui Liang
- China-New Zealand Joint Laboratory on Biomedicine and Health, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Centre for Regenerative Medicine and Health, Hong Kong Institute of Science and Innovation, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
- Research Unit of Generation of Large Animal Disease Models, Chinese Academy of Medical Sciences (2019RU015), Guangzhou 510530, China
- Sanya Institute of Swine Resource, Hainan Provincial Research Centre of Laboratory Animals, Sanya 572000, China
| | - Tao Chen
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, Wuyi University, Jiangmen 529020, China
| | - Han Wu
- China-New Zealand Joint Laboratory on Biomedicine and Health, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Centre for Regenerative Medicine and Health, Hong Kong Institute of Science and Innovation, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
- Research Unit of Generation of Large Animal Disease Models, Chinese Academy of Medical Sciences (2019RU015), Guangzhou 510530, China
- Sanya Institute of Swine Resource, Hainan Provincial Research Centre of Laboratory Animals, Sanya 572000, China
| | - Nan Li
- China-New Zealand Joint Laboratory on Biomedicine and Health, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Centre for Regenerative Medicine and Health, Hong Kong Institute of Science and Innovation, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
- Research Unit of Generation of Large Animal Disease Models, Chinese Academy of Medical Sciences (2019RU015), Guangzhou 510530, China
- Sanya Institute of Swine Resource, Hainan Provincial Research Centre of Laboratory Animals, Sanya 572000, China
| | - Yinghua Ye
- China-New Zealand Joint Laboratory on Biomedicine and Health, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Centre for Regenerative Medicine and Health, Hong Kong Institute of Science and Innovation, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
- Research Unit of Generation of Large Animal Disease Models, Chinese Academy of Medical Sciences (2019RU015), Guangzhou 510530, China
- Sanya Institute of Swine Resource, Hainan Provincial Research Centre of Laboratory Animals, Sanya 572000, China
| | - Liangxue Lai
- China-New Zealand Joint Laboratory on Biomedicine and Health, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Centre for Regenerative Medicine and Health, Hong Kong Institute of Science and Innovation, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
- Research Unit of Generation of Large Animal Disease Models, Chinese Academy of Medical Sciences (2019RU015), Guangzhou 510530, China
- Sanya Institute of Swine Resource, Hainan Provincial Research Centre of Laboratory Animals, Sanya 572000, China
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, Wuyi University, Jiangmen 529020, China
| | - Kepin Wang
- China-New Zealand Joint Laboratory on Biomedicine and Health, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Centre for Regenerative Medicine and Health, Hong Kong Institute of Science and Innovation, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
- Research Unit of Generation of Large Animal Disease Models, Chinese Academy of Medical Sciences (2019RU015), Guangzhou 510530, China
- Sanya Institute of Swine Resource, Hainan Provincial Research Centre of Laboratory Animals, Sanya 572000, China
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, Wuyi University, Jiangmen 529020, China
| |
Collapse
|
5
|
Popova J, Bets V, Kozhevnikova E. Perspectives in Genome-Editing Techniques for Livestock. Animals (Basel) 2023; 13:2580. [PMID: 37627370 PMCID: PMC10452040 DOI: 10.3390/ani13162580] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 08/08/2023] [Accepted: 08/08/2023] [Indexed: 08/27/2023] Open
Abstract
Genome editing of farm animals has undeniable practical applications. It helps to improve production traits, enhances the economic value of livestock, and increases disease resistance. Gene-modified animals are also used for biomedical research and drug production and demonstrate the potential to be used as xenograft donors for humans. The recent discovery of site-specific nucleases that allow precision genome editing of a single-cell embryo (or embryonic stem cells) and the development of new embryological delivery manipulations have revolutionized the transgenesis field. These relatively new approaches have already proven to be efficient and reliable for genome engineering and have wide potential for use in agriculture. A number of advanced methodologies have been tested in laboratory models and might be considered for application in livestock animals. At the same time, these methods must meet the requirements of safety, efficiency and availability of their application for a wide range of farm animals. This review aims at covering a brief history of livestock animal genome engineering and outlines possible future directions to design optimal and cost-effective tools for transgenesis in farm species.
Collapse
Affiliation(s)
- Julia Popova
- Laboratory of Bioengineering, Novosibirsk State Agrarian University, 630039 Novosibirsk, Russia; (J.P.); (V.B.)
| | - Victoria Bets
- Laboratory of Bioengineering, Novosibirsk State Agrarian University, 630039 Novosibirsk, Russia; (J.P.); (V.B.)
- Center of Technological Excellence, Novosibirsk State Technical University, 630073 Novosibirsk, Russia
| | - Elena Kozhevnikova
- Laboratory of Bioengineering, Novosibirsk State Agrarian University, 630039 Novosibirsk, Russia; (J.P.); (V.B.)
- Laboratory of Experimental Models of Cognitive and Emotional Disorders, Scientific-Research Institute of Neurosciences and Medicine, 630117 Novosibirsk, Russia
| |
Collapse
|
6
|
Niu M, Zou Q. SgRNA-RF: Identification of SgRNA On-Target Activity With Imbalanced Datasets. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2022; 19:2442-2453. [PMID: 33979289 DOI: 10.1109/tcbb.2021.3079116] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Single-guide RNA is a guide RNA (gRNA), which guides the insertion or deletion of uridine residues into kinetoplastid during RNA editing. It is a small non-coding RNA that can be combined with pre -mRNA pairing. SgRNA is a critical component of the CRISPR/Cas9 gene knockout system and play an important role in gene editing and gene regulation. It is important to accurately and quickly identify highly on-target activity sgRNAs. Due to its importance, several computational predictors have been proposed to predict sgRNAs on-target activity. All these methods have clearly contributed to the development of this very important field. However, they also have certain limitations. In the paper, we developed a new classifier SgRNA-RF, which extracts the features of nucleic acid composition and structure of on-target activity sgRNA sequence and identified by random forest algorithm. In addition to solving an imbalanced dataset, this paper proposed a new method called CS-Smote. We compared sgRNA-RF with state-of-the-art predictors on the five datasets, and found SgRNA-RF significantly improved the identification accuracy, with accuracies of 0.8636,0.9161,0.894,0.938,0.965,0.77,0.979,0.973, respectively. The user-friendly web server that implements sgRNA-RF is freely available at http://server.malab.cn/sgRNA-RF/.
Collapse
|
7
|
Abstract
Cardiovascular disease remains the leading cause of morbidity and mortality in the developed world. In recent decades, extraordinary effort has been devoted to defining the molecular and pathophysiological characteristics of the diseased heart and vasculature. Mouse models have been especially powerful in illuminating the complex signaling pathways, genetic and epigenetic regulatory circuits, and multicellular interactions that underlie cardiovascular disease. The advent of CRISPR genome editing has ushered in a new era of cardiovascular research and possibilities for genetic correction of disease. Next-generation sequencing technologies have greatly accelerated the identification of disease-causing mutations, and advances in gene editing have enabled the rapid modeling of these mutations in mice and patient-derived induced pluripotent stem cells. The ability to correct the genetic drivers of cardiovascular disease through delivery of gene editing components in vivo, while still facing challenges, represents an exciting therapeutic frontier. In this review, we provide an overview of cardiovascular disease mechanisms and the potential applications of CRISPR genome editing for disease modeling and correction. We also discuss the extent to which mice can faithfully model cardiovascular disease and the opportunities and challenges that lie ahead.
Collapse
Affiliation(s)
- Ning Liu
- Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas
| | - Eric N Olson
- Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas
| |
Collapse
|
8
|
Song R, Wang Y, Zheng Q, Yao J, Cao C, Wang Y, Zhao J. One-step base editing in multiple genes by direct embryo injection for pig trait improvement. SCIENCE CHINA. LIFE SCIENCES 2022; 65:739-752. [PMID: 35060075 DOI: 10.1007/s11427-021-2013-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 09/17/2021] [Indexed: 10/19/2022]
Abstract
The precise and simultaneous acquisition of multiple beneficial alleles in the genome is in great demand for the development of elite pig breeders. Cytidine base editors (CBEs) that convert C:G to T:A have emerged as powerful tools for single-nucleotide replacement. Whether CBEs can effectively mediate C-to-T substitution at multiple sites/loci for trait improvement by direct zygote injection has not been verified in large animals. Here, we determined the editing efficiency of four CBE variants in porcine embryonic fibroblast cells and embryos. The findings showed that hA3A-BE3-Y130F and hA3A-eBE-Y130F consistently resulted in increased base-editing efficiency and low toxic effects in embryonic development. Further, we verified that using a one-step approach, direct zygote microinjection of the CBE system can generate pigs harboring multiple point mutations. Our process resulted in a stop codon in CD163 and myostatin (MSTN) and introduced a beneficial allele in insulin-like growth factor-2 (IGF2). The pigs showed disrupted expression of CD163 and MSTN and increased expression of IGF2, which significantly improved growth performance and infectious disease resistance. Our approach allows immediate introduction of multiple mutations in transgene-free animals to comprehensively improve economic traits through direct embryo microinjection, providing a potential new route to produce elite pig breeders.
Collapse
Affiliation(s)
- Ruigao Song
- State Key Laboratory of Stem cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.,The Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Taiyuan, 030032, China
| | - Yu Wang
- State Key Laboratory of Stem cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.,University of Chinese Academy of Sciences, Beijing, 100049, China.,State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100081, China
| | - Qiantao Zheng
- State Key Laboratory of Stem cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Jing Yao
- State Key Laboratory of Stem cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.,University of Chinese Academy of Sciences, Beijing, 100049, China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.,Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China
| | - Chunwei Cao
- State Key Laboratory of Stem cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Yanfang Wang
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100081, China.
| | - Jianguo Zhao
- State Key Laboratory of Stem cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China. .,University of Chinese Academy of Sciences, Beijing, 100049, China. .,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China. .,Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China.
| |
Collapse
|
9
|
Hou N, Du X, Wu S. Advances in pig models of human diseases. Animal Model Exp Med 2022; 5:141-152. [PMID: 35343091 PMCID: PMC9043727 DOI: 10.1002/ame2.12223] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 02/14/2022] [Accepted: 03/02/2022] [Indexed: 01/07/2023] Open
Abstract
Animal models of human diseases play a critical role in medical research. Pigs are anatomically and physiologically more like humans than are small rodents such as mice, making pigs an attractive option for modeling human diseases. Advances in recent years in genetic engineering have facilitated the rapid rise of pig models for use in studies of human disease. In the present review, we summarize the current status of pig models for human cardiovascular, metabolic, neurodegenerative, and various genetic diseases. We also discuss areas that need to be improved. Animal models of human diseases play a critical role in medical research. Advances in recent years in genetic engineering have facilitated the rapid rise of pig models for use in studies of human disease. In the present review, we summarize the current status of pig models for human cardiovascular, metabolic, neurodegenerative, various genetic diseases and xenotransplantation.
Collapse
Affiliation(s)
- Naipeng Hou
- College of Animal Science and Technology, China Agricultural University, Beijing, China.,Sanya Institute of China Agricultural University, Sanya, China
| | - Xuguang Du
- Sanya Institute of China Agricultural University, Sanya, China.,State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Sen Wu
- College of Animal Science and Technology, China Agricultural University, Beijing, China.,Sanya Institute of China Agricultural University, Sanya, China.,State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| |
Collapse
|
10
|
Li G, Li X, Zhuang S, Wang L, Zhu Y, Chen Y, Sun W, Wu Z, Zhou Z, Chen J, Huang X, Wang J, Li D, Li W, Wang H, Wei W. Gene editing and its applications in biomedicine. SCIENCE CHINA. LIFE SCIENCES 2022; 65:660-700. [PMID: 35235150 PMCID: PMC8889061 DOI: 10.1007/s11427-021-2057-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 12/06/2021] [Indexed: 02/06/2023]
Abstract
The steady progress in genome editing, especially genome editing based on the use of clustered regularly interspaced short palindromic repeats (CRISPR) and programmable nucleases to make precise modifications to genetic material, has provided enormous opportunities to advance biomedical research and promote human health. The application of these technologies in basic biomedical research has yielded significant advances in identifying and studying key molecular targets relevant to human diseases and their treatment. The clinical translation of genome editing techniques offers unprecedented biomedical engineering capabilities in the diagnosis, prevention, and treatment of disease or disability. Here, we provide a general summary of emerging biomedical applications of genome editing, including open challenges. We also summarize the tools of genome editing and the insights derived from their applications, hoping to accelerate new discoveries and therapies in biomedicine.
Collapse
Affiliation(s)
- Guanglei Li
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Xiangyang Li
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Songkuan Zhuang
- Department of Clinical Laboratory, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, 518035, China
| | - Liren Wang
- Shanghai Frontiers Science Research Base of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Yifan Zhu
- Shanghai Frontiers Science Research Base of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Yangcan Chen
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- Institute for Stem Cell and Regenerative Medicine, Chinese Academy of Sciences, Beijing, 100101, China
| | - Wen Sun
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Institute for Stem Cell and Regenerative Medicine, Chinese Academy of Sciences, Beijing, 100101, China
| | - Zeguang Wu
- Biomedical Pioneering Innovation Center, Beijing Advanced Innovation Center for Genomics, Peking-Tsinghua Center for Life Sciences, Peking University Genome Editing Research Center, State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing, 100871, China
| | - Zhuo Zhou
- Biomedical Pioneering Innovation Center, Beijing Advanced Innovation Center for Genomics, Peking-Tsinghua Center for Life Sciences, Peking University Genome Editing Research Center, State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing, 100871, China
| | - Jia Chen
- Gene Editing Center, School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China.
| | - Xingxu Huang
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China.
| | - Jin Wang
- Department of Clinical Laboratory, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, 518035, China.
| | - Dali Li
- Shanghai Frontiers Science Research Base of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai, 200241, China.
| | - Wei Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- Institute for Stem Cell and Regenerative Medicine, Chinese Academy of Sciences, Beijing, 100101, China.
- Bejing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China.
- HIT Center for Life Sciences, Harbin Institute of Technology, Harbin, 150001, China.
| | - Haoyi Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- Institute for Stem Cell and Regenerative Medicine, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Wensheng Wei
- Biomedical Pioneering Innovation Center, Beijing Advanced Innovation Center for Genomics, Peking-Tsinghua Center for Life Sciences, Peking University Genome Editing Research Center, State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing, 100871, China.
| |
Collapse
|
11
|
Namula Z, Hirata M, Le QA, Lin Q, Takebayashi K, Yoshimura N, Tanihara F, Thongkittidilok C, Otoi T. Zona pellucida treatment before CRISPR/Cas9-mediated genome editing of porcine zygotes. Vet Med Sci 2021; 8:164-169. [PMID: 34674375 PMCID: PMC8788957 DOI: 10.1002/vms3.659] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Background Increasing the permeability of the zona pellucida (ZP) of oocytes before CRISPR/Cas9 electroporation may improve the efficiency of gene editing; however, the effects of this approach on subsequent developmental processes are unclear. In this study, the effects of ZP treatment before electroporation on embryonic development and gene editing in porcine embryos were evaluated. Methods The ZP of zygotes was weakened or removed by exposure to 0.5% actinase E, followed by electroporation of the Cas9 protein with guide RNA targeting GGTA1. Results The blastocyst formation rate of ZP‐free zygotes after electroporation was significantly lower (p < 0.05) than that of ZP‐intact zygotes. The mutation rate in blastocysts from ZP‐weakened zygotes was similar to that in ZP‐intact zygotes, whereas ZP removal increased the mutation rate. The mutation efficiency in blastocysts from electroporated zygotes did not differ among ZP treatment groups. Conclusions Our results indicate that weakening the ZP does not affect the developmental competence, mutation rate, or mutation efficiency of electroporated zygotes, whereas ZP removal has a detrimental effect on embryonic development but may increase the mutation rate.
Collapse
Affiliation(s)
- Zhao Namula
- Faculty of Veterinary Science, Guangdong Ocean University, Zhanjiang, China.,Faculty of Bioscience and Bioindustry, Tokushima University, Tokushima, Japan
| | - Maki Hirata
- Faculty of Bioscience and Bioindustry, Tokushima University, Tokushima, Japan
| | - Quynh Anh Le
- Faculty of Bioscience and Bioindustry, Tokushima University, Tokushima, Japan
| | - Qingyi Lin
- Faculty of Bioscience and Bioindustry, Tokushima University, Tokushima, Japan
| | - Koki Takebayashi
- Faculty of Bioscience and Bioindustry, Tokushima University, Tokushima, Japan
| | - Naoaki Yoshimura
- Faculty of Bioscience and Bioindustry, Tokushima University, Tokushima, Japan
| | - Fuminori Tanihara
- Faculty of Bioscience and Bioindustry, Tokushima University, Tokushima, Japan
| | | | - Takeshige Otoi
- Faculty of Bioscience and Bioindustry, Tokushima University, Tokushima, Japan
| |
Collapse
|
12
|
Abstract
Germline editing, the process by which the genome of an individual is edited in such a way that the change is heritable, has been applied to a wide variety of animals [D. A. Sorrell, A. F. Kolb, Biotechnol. Adv. 23, 431-469 (2005); D. Baltimore et al., Science 348, 36-38 (2015)]. Because of its relevancy in agricultural and biomedical research, the pig genome has been extensively modified using a multitude of technologies [K. Lee, K. Farrell, K. Uh, Reprod. Fertil. Dev. 32, 40-49 (2019); C. Proudfoot, S. Lillico, C. Tait-Burkard, Anim. Front. 9, 6-12 (2019)]. In this perspective, we will focus on using pigs as the model system to review the current methodologies, applications, and challenges of mammalian germline genome editing. We will also discuss the broad implications of animal germline editing and its clinical potential.
Collapse
|
13
|
Ratner LD, La Motta GE, Briski O, Salamone DF, Fernandez-Martin R. Practical Approaches for Knock-Out Gene Editing in Pigs. Front Genet 2021; 11:617850. [PMID: 33747029 PMCID: PMC7973260 DOI: 10.3389/fgene.2020.617850] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 12/30/2020] [Indexed: 12/18/2022] Open
Abstract
Pigs are an important resource for meat production and serve as a model for human diseases. Due to their physiological and anatomical similarities to humans, these animals can recapitulate symptoms of human diseases, becoming an effective model for biomedical research. Although, in the past pig have not been widely used partially because of the difficulty in genetic modification; nowadays, with the new revolutionary technology of programmable nucleases, and fundamentally of the CRISPR-Cas9 systems, it is possible for the first time to precisely modify the porcine genome as never before. To this purpose, it is necessary to introduce the system into early stage zygotes or to edit cells followed by somatic cell nuclear transfer. In this review, several strategies for pig knock-out gene editing, using the CRISPR-Cas9 system, will be summarized, as well as genotyping methods and different delivery techniques to introduce these tools into the embryos. Finally, the best approaches to produce homogeneous, biallelic edited animals will be discussed.
Collapse
Affiliation(s)
- Laura Daniela Ratner
- Laboratorio Biotecnología Animal (LabBA), Departamento de Producción Animal, Facultad de Agronomía, Universidad de Buenos Aires, Buenos Aires, Argentina.,Instituto de Investigaciones en Producción Animal (INPA), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Gaston Emilio La Motta
- Laboratorio Biotecnología Animal (LabBA), Departamento de Producción Animal, Facultad de Agronomía, Universidad de Buenos Aires, Buenos Aires, Argentina.,Instituto de Investigaciones en Producción Animal (INPA), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Olinda Briski
- Laboratorio Biotecnología Animal (LabBA), Departamento de Producción Animal, Facultad de Agronomía, Universidad de Buenos Aires, Buenos Aires, Argentina.,Instituto de Investigaciones en Producción Animal (INPA), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Daniel Felipe Salamone
- Laboratorio Biotecnología Animal (LabBA), Departamento de Producción Animal, Facultad de Agronomía, Universidad de Buenos Aires, Buenos Aires, Argentina.,Instituto de Investigaciones en Producción Animal (INPA), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Rafael Fernandez-Martin
- Laboratorio Biotecnología Animal (LabBA), Departamento de Producción Animal, Facultad de Agronomía, Universidad de Buenos Aires, Buenos Aires, Argentina.,Instituto de Investigaciones en Producción Animal (INPA), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
14
|
Niu M, Lin Y, Zou Q. sgRNACNN: identifying sgRNA on-target activity in four crops using ensembles of convolutional neural networks. PLANT MOLECULAR BIOLOGY 2021; 105:483-495. [PMID: 33385273 DOI: 10.1007/s11103-020-01102-y] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Accepted: 12/01/2020] [Indexed: 06/12/2023]
Abstract
KEY MESSAGE We proposed an ensemble convolutional neural network model to identify sgRNA high on-target activity in four crops and we used one-hot encoding and k-mers for sequence encoding. As an important component of the CRISPR/Cas9 system, single-guide RNA (sgRNA) plays an important role in gene redirection and editing. sgRNA has played an important role in the improvement of agronomic species, but there is a lack of effective bioinformatics tools to identify the activity of sgRNA in agronomic species. Therefore, it is necessary to develop a method based on machine learning to identify sgRNA high on-target activity. In this work, we proposed a simple convolutional neural network method to identify sgRNA high on-target activity. Our study used one-hot encoding and k-mers for sequence data conversion and a voting algorithm for constructing the convolutional neural network ensemble model sgRNACNN for the prediction of sgRNA activity. The ensemble model sgRNACNN was used for predictions in four crops: Glycine max, Zea mays, Sorghum bicolor and Triticum aestivum. The accuracy rates of the four crops in the sgRNACNN model were 82.43%, 80.33%, 78.25% and 87.49%, respectively. The experimental results showed that sgRNACNN realizes the identification of high on-target activity sgRNA of agronomic data and can meet the demands of sgRNA activity prediction in agronomy to a certain extent. These results have certain significance for guiding crop gene editing and academic research. The source code and relevant dataset can be found in the following link: https://github.com/nmt315320/sgRNACNN.git .
Collapse
Affiliation(s)
- Mengting Niu
- Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology of China, Chengdu, China
| | - Yuan Lin
- Department of System Integration, Sparebanken Vest, Bergen, Norway.
| | - Quan Zou
- Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology of China, Chengdu, China.
| |
Collapse
|
15
|
Lipofection-Mediated Introduction of CRISPR/Cas9 System into Porcine Oocytes and Embryos. Animals (Basel) 2021; 11:ani11020578. [PMID: 33672168 PMCID: PMC7926877 DOI: 10.3390/ani11020578] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 02/11/2021] [Accepted: 02/19/2021] [Indexed: 02/06/2023] Open
Abstract
Simple Summary Liposome-mediated gene transfer has become an alternative method for establishing a gene targeting framework, and the production of mutant animals may be feasible even in laboratories without specialized equipment. However, whether blastocyst genome editing can be performed by treatment with lipofection reagent, guide RNA, and Cas9, without performing electroporation or microinjection, remains unclear. In this study, we demonstrated that lipofection treatment successfully induced mutation into zygotes during in vitro fertilization and in embryos at the 2- and 4-cell stages. Although liposome-mediated gene editing is a feasible system for use with zona-pellucida-free oocytes/embryos, several challenges must be overcome. Abstract Liposome-mediated gene transfer has become an alternative method for establishing a gene targeting framework, and the production of mutant animals may be feasible even in laboratories without specialized equipment. However, how this system functions in mammalian oocytes and embryos remains unclear. The present study was conducted to clarify whether blastocyst genome editing can be performed by treatment with lipofection reagent, guide RNA, and Cas9 for 5 h without using electroporation or microinjection. A mosaic mutation was observed in blastocysts derived from zona pellucida (ZP)-free oocytes following lipofection treatment, regardless of the target genes. When lipofection treatment was performed after in vitro fertilization (IVF), no significant differences in the mutation rates or mutation efficiency were found between blastocysts derived from embryos treated at 24 and 29 h from the start of IVF. Only blastocysts from embryos exposed to lipofection treatment at 29 h after IVF contained biallelic mutant. Furthermore, there were no significant differences in the mutation rates or mutation efficiency between blastocysts derived from embryos at the 2- and 4-cell stages. This suggests that lipofection-mediated gene editing can be performed in ZP-free oocytes and ZP-free embryos; however, other factors affecting the system efficiency should be further investigated.
Collapse
|
16
|
Perisse IV, Fan Z, Singina GN, White KL, Polejaeva IA. Improvements in Gene Editing Technology Boost Its Applications in Livestock. Front Genet 2021; 11:614688. [PMID: 33603767 PMCID: PMC7885404 DOI: 10.3389/fgene.2020.614688] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 12/07/2020] [Indexed: 12/18/2022] Open
Abstract
Accelerated development of novel CRISPR/Cas9-based genome editing techniques provides a feasible approach to introduce a variety of precise modifications in the mammalian genome, including introduction of multiple edits simultaneously, efficient insertion of long DNA sequences into specific targeted loci as well as performing nucleotide transitions and transversions. Thus, the CRISPR/Cas9 tool has become the method of choice for introducing genome alterations in livestock species. The list of new CRISPR/Cas9-based genome editing tools is constantly expanding. Here, we discuss the methods developed to improve efficiency and specificity of gene editing tools as well as approaches that can be employed for gene regulation, base editing, and epigenetic modifications. Additionally, advantages and disadvantages of two primary methods used for the production of gene-edited farm animals: somatic cell nuclear transfer (SCNT or cloning) and zygote manipulations will be discussed. Furthermore, we will review agricultural and biomedical applications of gene editing technology.
Collapse
Affiliation(s)
- Iuri Viotti Perisse
- Department of Animal, Dairy and Veterinary Sciences, Utah State University, Logan, UT, United States
| | - Zhiqiang Fan
- Department of Animal, Dairy and Veterinary Sciences, Utah State University, Logan, UT, United States
| | - Galina N. Singina
- L.K. Ernst Federal Research Center for Animal Husbandry, Podolsk, Russia
| | - Kenneth L. White
- Department of Animal, Dairy and Veterinary Sciences, Utah State University, Logan, UT, United States
| | - Irina A. Polejaeva
- Department of Animal, Dairy and Veterinary Sciences, Utah State University, Logan, UT, United States
| |
Collapse
|
17
|
Le QA, Tanihara F, Wittayarat M, Namula Z, Sato Y, Lin Q, Takebayashi K, Hirata M, Otoi T. Comparison of the effects of introducing the CRISPR/Cas9 system by microinjection and electroporation into porcine embryos at different stages. BMC Res Notes 2021; 14:7. [PMID: 33407863 PMCID: PMC7788904 DOI: 10.1186/s13104-020-05412-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 12/03/2020] [Indexed: 11/10/2022] Open
Abstract
Objective Cytoplasmic microinjection and electroporation of the CRISPR/Cas9 system into zygotes are used for generating genetically modified pigs. However, these methods create mosaic mutations in embryos. In this study, we evaluated whether the gene editing method and embryonic stage for gene editing affect the gene editing efficiency of porcine embryos. Results First, we designed five guide RNAs (gRNAs) targeting the B4GALNT2 gene and evaluated mutation efficiency by introducing each gRNA with Cas9 protein into zygotes by electroporation. Next, the optimized gRNA with Cas9 protein was introduced into 1-cell and 2-cell stage embryos by either microinjection or electroporation. The sequence of gRNA affected the bi-allelic mutation rate and mutation efficiency of blastocysts derived from electroporated embryos. Microinjection significantly decreased the cleavage rates in each embryonic stage and blastocyst formation rates in 2-cell stage embryos compared with electroporation (p < 0.05). However, the bi-allelic mutation rate and mutation efficiency of blastocysts from the 1-cell stage embryos edited using microinjection were significantly higher (p < 0.05) than those of blastocysts from the 2-cell stage embryos edited by both methods. These results indicate that the gene editing method and embryonic stage for gene editing may affect the genotype and mutation efficiency of the resulting embryos.
Collapse
Affiliation(s)
- Quynh Anh Le
- Laboratory of Animal Reproduction, Faculty of Bioscience and Bioindustry, Tokushima University, 2272-1 Ishii, Myozai-gun, Tokushima, 779-3233, Japan
| | - Fuminori Tanihara
- Laboratory of Animal Reproduction, Faculty of Bioscience and Bioindustry, Tokushima University, 2272-1 Ishii, Myozai-gun, Tokushima, 779-3233, Japan.
| | - Manita Wittayarat
- Faculty of Veterinary Science, Prince of Songkla University, Songkhla, Thailand
| | - Zhao Namula
- Laboratory of Animal Reproduction, Faculty of Bioscience and Bioindustry, Tokushima University, 2272-1 Ishii, Myozai-gun, Tokushima, 779-3233, Japan.,College of Coastal Agricultural Sciences, Guangdong Ocean University, Guangdong, China
| | - Yoko Sato
- School of Biological Science, Tokai University, Sapporo, Japan
| | - Qingyi Lin
- Laboratory of Animal Reproduction, Faculty of Bioscience and Bioindustry, Tokushima University, 2272-1 Ishii, Myozai-gun, Tokushima, 779-3233, Japan
| | - Koki Takebayashi
- Laboratory of Animal Reproduction, Faculty of Bioscience and Bioindustry, Tokushima University, 2272-1 Ishii, Myozai-gun, Tokushima, 779-3233, Japan
| | - Maki Hirata
- Laboratory of Animal Reproduction, Faculty of Bioscience and Bioindustry, Tokushima University, 2272-1 Ishii, Myozai-gun, Tokushima, 779-3233, Japan
| | - Takeshige Otoi
- Laboratory of Animal Reproduction, Faculty of Bioscience and Bioindustry, Tokushima University, 2272-1 Ishii, Myozai-gun, Tokushima, 779-3233, Japan
| |
Collapse
|
18
|
Niu D, Ma X, Yuan T, Niu Y, Xu Y, Sun Z, Ping Y, Li W, Zhang J, Wang T, Church GM. Porcine genome engineering for xenotransplantation. Adv Drug Deliv Rev 2021; 168:229-245. [PMID: 32275950 DOI: 10.1016/j.addr.2020.04.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 03/28/2020] [Accepted: 04/06/2020] [Indexed: 02/06/2023]
Abstract
The extreme shortage of human donor organs for treatment of patients with end-stage organ failures is well known. Xenotransplantation, which might provide unlimited organ supply, is a most promising strategy to solve this problem. Domestic pigs are regarded as ideal organ-source animals owing to similarity in anatomy, physiology and organ size to humans as well as high reproductive capacity and low maintenance cost. However, several barriers, which include immune rejection, inflammation and coagulative dysfunctions, as well as the cross-species transmission risk of porcine endogenous retrovirus, blocked the pig-to-human xenotransplantation. With the rapid development of genome engineering technologies and the potent immunosuppressive medications in recent years, these barriers could be eliminated through genetic modification of pig genome together with the administration of effective immunosuppressants. A number of candidate genes involved in the regulation of immune response, inflammation and coagulation have been explored to optimize porcine xenograft survival in non-human primate recipients. PERV inactivation in pigs has also been accomplished to firmly address the safety issue in pig-to-human xenotransplantation. Many encouraging preclinical milestones have been achieved with some organs surviving for years. Therefore, the clinical trials of some promising organs, such as islet, kidney and heart, are aimed to be launched in the near future.
Collapse
Affiliation(s)
- Dong Niu
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, China-Australian Joint Laboratory for Animal Health Big Data Analytics, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection & Internet Technology, College of Animal Science and Technology & College of Veterinary Medicine of Zhejiang A&F University, Hangzhou, P.R. China
| | - Xiang Ma
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, China-Australian Joint Laboratory for Animal Health Big Data Analytics, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection & Internet Technology, College of Animal Science and Technology & College of Veterinary Medicine of Zhejiang A&F University, Hangzhou, P.R. China
| | - Taoyan Yuan
- Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Sciences, Hangzhou, Zhejiang 310021, China
| | - Yifan Niu
- Nanjing Kgene Genetic Engineering Co., Ltd, Nanjing, Jiangsu 211300, China
| | - Yibin Xu
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Zhongxin Sun
- Cosmetic & Plastic Surgery Department, Hangzhou First People's Hospital, Hangzhou, Zhejiang 310006, China
| | - Yuan Ping
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Weifen Li
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Jufang Zhang
- Cosmetic & Plastic Surgery Department, Hangzhou First People's Hospital, Hangzhou, Zhejiang 310006, China.
| | - Tao Wang
- Nanjing Kgene Genetic Engineering Co., Ltd, Nanjing, Jiangsu 211300, China.
| | - George M Church
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA 02138, USA.
| |
Collapse
|
19
|
Hennig SL, Owen JR, Lin JC, Young AE, Ross PJ, Van Eenennaam AL, Murray JD. Evaluation of mutation rates, mosaicism and off target mutations when injecting Cas9 mRNA or protein for genome editing of bovine embryos. Sci Rep 2020; 10:22309. [PMID: 33339870 PMCID: PMC7749171 DOI: 10.1038/s41598-020-78264-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 11/19/2020] [Indexed: 12/11/2022] Open
Abstract
The CRISPR/Cas9 genome editing tool has the potential to improve the livestock breeding industry by allowing for the introduction of desirable traits. Although an efficient and targeted tool, the CRISPR/Cas9 system can have some drawbacks, including off-target mutations and mosaicism, particularly when used in developing embryos. Here, we introduced genome editing reagents into single-cell bovine embryos to compare the effect of Cas9 mRNA and protein on the mutation efficiency, level of mosaicism, and evaluate potential off-target mutations utilizing next generation sequencing. We designed guide-RNAs targeting three loci (POLLED, H11, and ZFX) in the bovine genome and saw a significantly higher rate of mutation in embryos injected with Cas9 protein (84.2%) vs. Cas9 mRNA (68.5%). In addition, the level of mosaicism was higher in embryos injected with Cas9 mRNA (100%) compared to those injected with Cas9 protein (94.2%), with little to no unintended off-target mutations detected. This study demonstrated that the use of gRNA/Cas9 ribonucleoprotein complex resulted in a high editing efficiency at three different loci in bovine embryos and decreased levels of mosaicism relative to Cas9 mRNA. Additional optimization will be required to further reduce mosaicism to levels that make single-step embryo editing in cattle commercially feasible.
Collapse
Affiliation(s)
- Sadie L Hennig
- Department of Animal Science, University of California - Davis, Davis, CA, USA
| | - Joseph R Owen
- Department of Animal Science, University of California - Davis, Davis, CA, USA
| | - Jason C Lin
- Department of Animal Science, University of California - Davis, Davis, CA, USA
| | - Amy E Young
- Department of Animal Science, University of California - Davis, Davis, CA, USA
| | - Pablo J Ross
- Department of Animal Science, University of California - Davis, Davis, CA, USA
| | | | - James D Murray
- Department of Animal Science, University of California - Davis, Davis, CA, USA.,Department of Population Health and Reproduction, University of California - Davis, Davis, CA, USA
| |
Collapse
|
20
|
Park KE, Frey JF, Waters J, Simpson SG, Coutu C, Plummer S, Campbell M, Donovan DM, Telugu BP. One-Step Homology Mediated CRISPR-Cas Editing in Zygotes for Generating Genome Edited Cattle. CRISPR J 2020; 3:523-534. [PMID: 33252243 PMCID: PMC7757693 DOI: 10.1089/crispr.2020.0047] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Selective breeding and genetic modification have been the cornerstone of animal agriculture. However, the current strategy of breeding animals over multiple generations to introgress novel alleles is not practical in addressing global challenges such as climate change, pandemics, and the predicted need to feed a population of 9 billion by 2050. Consequently, genome editing in zygotes to allow for seamless introgression of novel alleles is required, especially in cattle with long generation intervals. We report for the first time the use of CRISPR-Cas genome editors to introduce novel PRNP allelic variants that have been shown to provide resilience towards human prion pandemics. From one round of embryo injections, we have established six pregnancies and birth of seven edited offspring, with two founders showing >90% targeted homology-directed repair modifications. This study lays out the framework for in vitro optimization, unbiased deep-sequencing to identify editing outcomes, and generation of high frequency homology-directed repair–edited calves.
Collapse
Affiliation(s)
- Ki-Eun Park
- Animal and Avian Sciences, University of Maryland, College Park, Maryland, USA.,RenOVAte Biosciences, Inc., Reisterstown, Maryland, USA
| | - Juli Foster Frey
- Animal Biosciences and Biotechnology Laboratory, USDA, ARS, Beltsville, Maryland, USA
| | - Jerel Waters
- RenOVAte Biosciences, Inc., Reisterstown, Maryland, USA
| | - Sean G Simpson
- Animal and Avian Sciences, University of Maryland, College Park, Maryland, USA.,RenOVAte Biosciences, Inc., Reisterstown, Maryland, USA
| | - Chris Coutu
- Thomas D. Morris Inc., Reisterstown, Maryland, USA
| | | | | | - David M Donovan
- Animal Biosciences and Biotechnology Laboratory, USDA, ARS, Beltsville, Maryland, USA
| | - Bhanu P Telugu
- Animal and Avian Sciences, University of Maryland, College Park, Maryland, USA.,RenOVAte Biosciences, Inc., Reisterstown, Maryland, USA
| |
Collapse
|
21
|
Xie F, Zhou X, Lin T, Wang L, Liu C, Luo X, Luo L, Chen H, Guo K, Wei H, Wang Y. Production of gene-edited pigs harboring orthologous human mutations via double cutting by CRISPR/Cas9 with long single-stranded DNAs as homology-directed repair templates by zygote injection. Transgenic Res 2020; 29:587-598. [PMID: 33170439 DOI: 10.1007/s11248-020-00218-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Accepted: 10/21/2020] [Indexed: 12/14/2022]
Abstract
Precise gene editing of model organisms is required for accurately modeling human diseases and deciphering gene functions. In this study, we used a pair of guide RNAs (sgRNAs), which in vitro transcribed along with other CRISPR RNA components, to generate two cleavage sites flanking pig GJB2 (pGJB2) CDS. By using long single-stranded DNAs (lssDNA) as homology-directed repair (HDR) templates, we efficiently obtained two gene-edited pigs, of which GJB2 CDS replaced with CDSs containing human GJB2 c.235delC mutation and orthologous human p.V37I mutation, respectively. These mutations were commonly observed in patients with hearing loss. Genetic analysis of the two gene-edited pigs showed that the HDR-derived gene-editing efficiency were as high as 80% (4/5) and 50% (2/4), respectively. While no mutation was observed in the group of single cutting with one sgRNA covering the 235th nucleotide C in pGJB2 CDS, using a short single-stranded oligo DNA containing c.235delC mutation as HDR template. Extra experiments proved that the intended mutations were successfully transmitted to offspring or extensively integrated into various tissues including gonad of founder pigs. Our work indicated that the new "double cutting with lssDNA template" gene editing method can expand sgRNA selection scope and avoids direct cutting of gene CDS. Additionally, can introduce precise mutations into mammalian genomic sites, especially those with unavailable proper protospacer sequence or being resistant to gene editing. Moreover, this method can be performed with CRISPR RNA reagents instead of CRISPR ribonucleoproteins applied in previous reports.
Collapse
Affiliation(s)
- Fei Xie
- Department of Laboratory Animal Science, College of Basic Medical Sciences, Army Medical University, Third Military Medical University, Chongqing, 400038, China
| | - Xiaoyang Zhou
- Department of Laboratory Animal Science, College of Basic Medical Sciences, Army Medical University, Third Military Medical University, Chongqing, 400038, China
| | - Tingting Lin
- Department of Laboratory Animal Science, College of Basic Medical Sciences, Army Medical University, Third Military Medical University, Chongqing, 400038, China
| | - Lulu Wang
- Department of Laboratory Animal Science, College of Basic Medical Sciences, Army Medical University, Third Military Medical University, Chongqing, 400038, China
| | - Chuanhong Liu
- Department of Laboratory Animal Science, College of Basic Medical Sciences, Army Medical University, Third Military Medical University, Chongqing, 400038, China
| | - Xi Luo
- Department of Foreign Languages Studies, College of Basic Medical Sciences, Army Medical University, Third Military Medical University, Chongqing, 400038, China
| | - Lihua Luo
- Department of Laboratory Animal Science, College of Basic Medical Sciences, Army Medical University, Third Military Medical University, Chongqing, 400038, China
| | - Huayu Chen
- Department of Laboratory Animal Science, College of Basic Medical Sciences, Army Medical University, Third Military Medical University, Chongqing, 400038, China
| | - Kenan Guo
- Department of Laboratory Animal Science, College of Basic Medical Sciences, Army Medical University, Third Military Medical University, Chongqing, 400038, China
| | - Hong Wei
- Department of Laboratory Animal Science, College of Basic Medical Sciences, Army Medical University, Third Military Medical University, Chongqing, 400038, China.
| | - Yong Wang
- Department of Laboratory Animal Science, College of Basic Medical Sciences, Army Medical University, Third Military Medical University, Chongqing, 400038, China.
| |
Collapse
|
22
|
Lee K, Farrell K, Uh K. Application of genome-editing systems to enhance available pig resources for agriculture and biomedicine. Reprod Fertil Dev 2020; 32:40-49. [PMID: 32188556 DOI: 10.1071/rd19273] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Traditionally, genetic engineering in the pig was a challenging task. Genetic engineering of somatic cells followed by somatic cell nuclear transfer (SCNT) could produce genetically engineered (GE) pigs carrying site-specific modifications. However, due to difficulties in engineering the genome of somatic cells and developmental defects associated with SCNT, a limited number of GE pig models were reported. Recent developments in genome-editing tools, such as zinc finger nucleases (ZFNs), transcription activator-like effector nucleases (TALENs) and the clustered regularly interspaced short palindromic repeat (CRISPR)/CRISPR-associated (Cas) 9 system, have markedly changed the effort and time required to produce GE pig models. The frequency of genetic engineering in somatic cells is now practical. In addition, SCNT is no longer essential in producing GE pigs carrying site-specific modifications, because direct injection of genome-editing systems into developing embryos introduces targeted modifications. To date, the CRISPR/Cas9 system is the most convenient, cost-effective, timely and commonly used genome-editing technology. Several applicable biomedical and agricultural pig models have been generated using the CRISPR/Cas9 system. Although the efficiency of genetic engineering has been markedly enhanced with the use of genome-editing systems, improvements are still needed to optimally use the emerging technology. Current and future advances in genome-editing strategies will have a monumental effect on pig models used in agriculture and biomedicine.
Collapse
Affiliation(s)
- Kiho Lee
- Department of Animal and Poultry Sciences, Litton-Reaves Hall, Virginia Tech, Blacksburg, Virgina 24061, USA; and Corresponding author.
| | - Kayla Farrell
- Department of Animal and Poultry Sciences, Litton-Reaves Hall, Virginia Tech, Blacksburg, Virgina 24061, USA
| | - Kyungjun Uh
- Department of Animal and Poultry Sciences, Litton-Reaves Hall, Virginia Tech, Blacksburg, Virgina 24061, USA
| |
Collapse
|
23
|
Su X, Chen W, Cai Q, Liang P, Chen Y, Cong P, Huang J. Effective generation of maternal genome point mutated porcine embryos by injection of cytosine base editor into germinal vesicle oocytes. SCIENCE CHINA. LIFE SCIENCES 2020; 63:996-1005. [PMID: 31974864 DOI: 10.1007/s11427-019-1611-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 12/23/2019] [Indexed: 01/19/2023]
Abstract
Cytosine and adenine base editors are promising new tools for introducing precise genetic modifications that are required to generate disease models and to improve traits in pigs. Base editors can catalyze the conversion of C→T (C>T) or A→G (A>G) in the target site through a single guide RNA. Injection of base editors into the zygote cytoplasm can result in the production of offspring with precise point mutations, but most F0 are mosaic, and breeding of F1 heterozygous pigs is time-intensive. Here, we developed a method called germinal vesicle oocyte base editing (GVBE) to produce point mutant F0 porcine embryos by editing the maternal alleles during the GV to MII transition. Injection of cytosine base editor 3 (BE3) mRNA and X-linked Dmd-specific guide RNAs into GVoocytes efficiently edited maternal Dmd during in vitro maturation and did not affect the maturation potential of the oocytes. The edited MII oocytes developed into blastocysts after parthenogenetic activation (PA) or in vitro fertilization (IVF). However, BE3 may reduce the developmental potential of IVF blastocysts from 31.5%±0.8% to 20.4% ±2.1%. There 40%-78.3% diploid PA blastocysts had no more than two different alleles, including up to 10% embryos that had only C>T mutation alleles. Genotyping of IVF blastocysts indicated that over 70% of the edited embryos had one allele or two different alleles of Dmd. Since the male embryos had only a copy of Dmd allele, all five (5/19) F0 male embryos are homozygous and three of them were Dmd precise C>T mutation. Nine (9/19) female IVF embryos had two different alleles including a WT and a C>T mutation. DNA sequencing showed that some of them might be heterozygous embryos. In conclusion, the GVBE method is a valuable method for generating F0 embryos with maternal point mutated alleles in a single step.
Collapse
Affiliation(s)
- Xiaohu Su
- Key Laboratory of Reproductive Medicine of Guangdong Province, the First Affiliated Hospital and School of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, China
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, China
| | - Wei Chen
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, China
| | - Qingqing Cai
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, China
| | - Puping Liang
- Key Laboratory of Reproductive Medicine of Guangdong Province, the First Affiliated Hospital and School of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, China
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, China
| | - Yaosheng Chen
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, China
| | - Peiqing Cong
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, China.
| | - Junjiu Huang
- Key Laboratory of Reproductive Medicine of Guangdong Province, the First Affiliated Hospital and School of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, China.
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, China.
- Key Laboratory of Reproductive Medicine of Guangdong Province, the Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China.
| |
Collapse
|
24
|
Menchaca A, Dos Santos-Neto PC, Mulet AP, Crispo M. CRISPR in livestock: From editing to printing. Theriogenology 2020; 150:247-254. [PMID: 32088034 PMCID: PMC7102594 DOI: 10.1016/j.theriogenology.2020.01.063] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 01/28/2020] [Indexed: 12/16/2022]
Abstract
Precise genome editing of large animals applied to livestock and biomedicine is nowadays possible since the CRISPR revolution. This review summarizes the latest advances and the main technical issues that determine the success of this technology. The pathway from editing to printing, from engineering the genome to achieving the desired animals, does not always imply an easy, fast and safe journey. When applied in large animals, CRISPR involves time- and cost-consuming projects, and it is mandatory not only to choose the best approach for genome editing, but also for embryo production, zygote microinjection or electroporation, cryopreservation and embryo transfer. The main technical refinements and most frequent questions to improve this disruptive biotechnology in large animals are presented. In addition, we discuss some CRISPR applications to enhance livestock production in the context of a growing global demand of food, in terms of increasing efficiency, reducing the impact of farming on the environment, enhancing pest control, animal welfare and health. The challenge is no longer technical. Controversies and consensus, opportunities and threats, benefits and risks, ethics and science should be reconsidered to enter into the CRISPR era.
Collapse
Affiliation(s)
- A Menchaca
- Instituto de Reproducción Animal Uruguay, Fundación IRAUy, Cruz del Sur 2350, Montevideo, Uruguay.
| | - P C Dos Santos-Neto
- Instituto de Reproducción Animal Uruguay, Fundación IRAUy, Cruz del Sur 2350, Montevideo, Uruguay
| | - A P Mulet
- Unidad de Animales Transgénicos y de Experimentación (UATE), Institut Pasteur de Montevideo, Mataojo, 2020, Montevideo, Uruguay
| | - M Crispo
- Unidad de Animales Transgénicos y de Experimentación (UATE), Institut Pasteur de Montevideo, Mataojo, 2020, Montevideo, Uruguay
| |
Collapse
|
25
|
Hryhorowicz M, Lipiński D, Hryhorowicz S, Nowak-Terpiłowska A, Ryczek N, Zeyland J. Application of Genetically Engineered Pigs in Biomedical Research. Genes (Basel) 2020; 11:genes11060670. [PMID: 32575461 PMCID: PMC7349405 DOI: 10.3390/genes11060670] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 06/16/2020] [Accepted: 06/17/2020] [Indexed: 02/07/2023] Open
Abstract
Progress in genetic engineering over the past few decades has made it possible to develop methods that have led to the production of transgenic animals. The development of transgenesis has created new directions in research and possibilities for its practical application. Generating transgenic animal species is not only aimed towards accelerating traditional breeding programs and improving animal health and the quality of animal products for consumption but can also be used in biomedicine. Animal studies are conducted to develop models used in gene function and regulation research and the genetic determinants of certain human diseases. Another direction of research, described in this review, focuses on the use of transgenic animals as a source of high-quality biopharmaceuticals, such as recombinant proteins. The further aspect discussed is the use of genetically modified animals as a source of cells, tissues, and organs for transplantation into human recipients, i.e., xenotransplantation. Numerous studies have shown that the pig (Sus scrofa domestica) is the most suitable species both as a research model for human diseases and as an optimal organ donor for xenotransplantation. Short pregnancy, short generation interval, and high litter size make the production of transgenic pigs less time-consuming in comparison with other livestock species This review describes genetically modified pigs used for biomedical research and the future challenges and perspectives for the use of the swine animal models.
Collapse
Affiliation(s)
- Magdalena Hryhorowicz
- Department of Biochemistry and Biotechnology, Poznan University of Life Sciences, Dojazd 11, 60-632 Poznań, Poland; (D.L.); (A.N.-T.); (N.R.); (J.Z.)
- Correspondence:
| | - Daniel Lipiński
- Department of Biochemistry and Biotechnology, Poznan University of Life Sciences, Dojazd 11, 60-632 Poznań, Poland; (D.L.); (A.N.-T.); (N.R.); (J.Z.)
| | - Szymon Hryhorowicz
- Institute of Human Genetics, Polish Academy of Sciences, Strzeszyńska 32, 60-479 Poznań, Poland;
| | - Agnieszka Nowak-Terpiłowska
- Department of Biochemistry and Biotechnology, Poznan University of Life Sciences, Dojazd 11, 60-632 Poznań, Poland; (D.L.); (A.N.-T.); (N.R.); (J.Z.)
| | - Natalia Ryczek
- Department of Biochemistry and Biotechnology, Poznan University of Life Sciences, Dojazd 11, 60-632 Poznań, Poland; (D.L.); (A.N.-T.); (N.R.); (J.Z.)
| | - Joanna Zeyland
- Department of Biochemistry and Biotechnology, Poznan University of Life Sciences, Dojazd 11, 60-632 Poznań, Poland; (D.L.); (A.N.-T.); (N.R.); (J.Z.)
| |
Collapse
|
26
|
Livestock Gene Editing by One-step Embryo Manipulation. J Equine Vet Sci 2020; 89:103025. [PMID: 32563448 DOI: 10.1016/j.jevs.2020.103025] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 04/06/2020] [Accepted: 04/07/2020] [Indexed: 12/11/2022]
Abstract
The breakthrough and rapid advance of clustered regularly interspaced short palindromic repeat (CRISPR)/CRISPR-associated protein 9 (Cas9) technology has enabled the efficient generation of gene-edited animals by one-step embryo manipulation. Clustered regularly interspaced short palindromic repeat/CRISPR-associated protein 9 delivery to the livestock embryos has been typically achieved by intracytoplasmic microinjection; however, recent studies show that electroporation may be a reliable, efficient, and practical method for CRISPR/Cas9 delivery. The source of embryos used to generate gene-edited animals varies from in vivo to in vitro produced, depending mostly on the species of interest. In addition, different Cas9 and gRNA reagents can be used for embryo editing, ranging from Cas9-coding plasmid or messenger RNA to Cas9 recombinant protein, which can be combined with in vitro transcribed or synthetic guide RNAs. Mosaicism is reported as one of the main problems with generation of animals by embryo editing. On the other hand, off-target mutations are rarely found in livestock derived from one-step editing. In this review, we discussed these and other aspects of generating gene-edited animals by single-step embryo manipulation.
Collapse
|
27
|
Yamashita S, Kogasaka Y, Hiradate Y, Tanemura K, Sendai Y. Suppression of mosaic mutation by co-delivery of CRISPR associated protein 9 and three-prime repair exonuclease 2 into porcine zygotes via electroporation. J Reprod Dev 2020; 66:41-48. [PMID: 31761839 PMCID: PMC7040215 DOI: 10.1262/jrd.2019-088] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 11/01/2019] [Indexed: 12/15/2022] Open
Abstract
Gene-modified animals, including pigs, can be generated efficiently by introducing CRISPR associated protein 9 (CRISPR/Cas9) into zygotes. However, in many cases, these zygotes tend to become mosaic mutants with various different mutant cell types, making it difficult to analyze the phenotype of gene-modified founder animals. To reduce the mosaic mutations, we introduced three-prime repair exonuclease 2 (Trex2), an exonuclease that improves gene editing efficiency, into porcine zygotes along with CRISPR/Cas9 via electroporation. Although the rate of porcine blastocyst formation decreased due to electroporation (25.9 ± 4.6% vs. 41.2 ± 2.0%), co-delivery of murine Trex2 (mTrex2) mRNA with CRISPR/Cas9 did not affect it any further (25.9 ± 4.6% vs. 31.0 ± 4.6%). In addition, there was no significant difference in the diameter of blastocysts carrying CRISPR/Cas9 (164.7 ± 10.2 μm), and those with CRISPR/Cas9 + mTrex2 (151.9 ± 5.1 μm) as compared to those from the control group (178.9 ± 9.0 μm). These results revealed that mTrex2 did not affect the development of pre-implantation embryo. We also found bi-allelic, as well as mono-allelic, non-mosaic homozygous mutations in the blastocysts. Most importantly, co-delivery of mTrex2 mRNA with CRISPR/Cas9 increased non-mosaic mutant blastocysts (29.3 ± 4.5%) and reduced mosaic mutant blastocysts (70.7 ± 4.5%) as compared to CRISPR/Cas9 alone (5.6 ± 6.4% and 92.6 ± 8.6%, respectively). These data suggest that the co-delivery of CRISPR/Cas9 and mTrex2 is a useful method to suppress mosaic mutation.
Collapse
Affiliation(s)
- Shiro Yamashita
- Biological Sciences Section, Central Research Institute for Feed and Livestock of Zen-noh, Ibaraki 300-4204, Japan
- Laboratory of Animal Reproduction and Development, Graduate School of Agricultural Science, Tohoku University, Miyagi 980-8572, Japan
| | - Yuhei Kogasaka
- Biological Sciences Section, Central Research Institute for Feed and Livestock of Zen-noh, Ibaraki 300-4204, Japan
| | - Yuuki Hiradate
- Laboratory of Animal Reproduction and Development, Graduate School of Agricultural Science, Tohoku University, Miyagi 980-8572, Japan
| | - Kentaro Tanemura
- Laboratory of Animal Reproduction and Development, Graduate School of Agricultural Science, Tohoku University, Miyagi 980-8572, Japan
| | - Yutaka Sendai
- Biological Sciences Section, Central Research Institute for Feed and Livestock of Zen-noh, Ibaraki 300-4204, Japan
| |
Collapse
|
28
|
Le QA, Hirata M, Nguyen NT, Takebayashi K, Wittayarat M, Sato Y, Namula Z, Nii M, Tanihara F, Otoi T. Effects of electroporation treatment using different concentrations of Cas9 protein with gRNA targeting Myostatin (MSTN) genes on the development and gene editing of porcine zygotes. Anim Sci J 2020; 91:e13386. [PMID: 32512638 DOI: 10.1111/asj.13386] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 04/15/2020] [Accepted: 04/30/2020] [Indexed: 12/29/2022]
Abstract
This study was conducted to investigate the effect of seven concentrations of Cas9 protein (0, 25, 50, 100, 200, 500, and 1,000 ng/µl) on the development and gene editing of porcine embryos. This included the target editing and off-target effect of embryos developed from zygotes that were edited via electroporation of the Cas9 protein with guide RNA targeting Myostatin genes. We found that the development to blastocysts of electroporated zygotes was not affected by the concentration of Cas9 protein. Although the editing rate, which was defined as the ratio of edited blastocysts to total examined blastocysts, did not differ with Cas9 protein concentration, the editing efficiency, which was defined as the frequency of indel mutations in each edited blastocyst, was significantly decreased in the edited blastocysts from zygotes electroporated with 25 ng/µl of Cas9 protein compared with that of blastocysts from zygotes electroporated with higher Cas9 protein concentrations. Moreover the frequency of indel events at the two possible off-target sites was not significantly different with different concentrations of Cas9 protein. These results indicate that the concentration of Cas9 protein affects gene editing efficiency in embryos but not the embryonic development, gene editing rate, and non-specific cleavage of off-target sites.
Collapse
Affiliation(s)
- Quynh A Le
- Faculty of Bioscience and Bioindustry, Tokushima University, Tokushima, Japan
| | - Maki Hirata
- Faculty of Bioscience and Bioindustry, Tokushima University, Tokushima, Japan
| | - Nhien T Nguyen
- Faculty of Bioscience and Bioindustry, Tokushima University, Tokushima, Japan
| | - Koki Takebayashi
- Faculty of Bioscience and Bioindustry, Tokushima University, Tokushima, Japan
| | - Manita Wittayarat
- Faculty of Veterinary Science, Prince of Songkla University, Songkhla, Thailand
| | - Yoko Sato
- School of Biological Science, Tokai University, Sapporo, Japan
| | - Zhao Namula
- Faculty of Veterinary Science, Guangdong Ocean University, Zhanjiang, China
| | - Masahiro Nii
- Tokushima Prefectural Livestock Research Institute, Tokushima, Japan
| | - Fuminori Tanihara
- Faculty of Bioscience and Bioindustry, Tokushima University, Tokushima, Japan
| | - Takeshige Otoi
- Faculty of Bioscience and Bioindustry, Tokushima University, Tokushima, Japan
| |
Collapse
|
29
|
Lamas-Toranzo I, Galiano-Cogolludo B, Cornudella-Ardiaca F, Cobos-Figueroa J, Ousinde O, Bermejo-Álvarez P. Strategies to reduce genetic mosaicism following CRISPR-mediated genome edition in bovine embryos. Sci Rep 2019; 9:14900. [PMID: 31624292 PMCID: PMC6797768 DOI: 10.1038/s41598-019-51366-8] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Accepted: 09/30/2019] [Indexed: 11/29/2022] Open
Abstract
Genetic mosaicism is the presence of more than two alleles on an individual and it is commonly observed following CRISPR microinjection of zygotes. This phenomenon appears when DNA replication precedes CRISPR-mediated genome edition and it is undesirable because it reduces greatly the odds for direct KO generation by randomly generated indels. In this study, we have developed alternative protocols to reduce mosaicism rates following CRISPR-mediated genome edition in bovine. In a preliminary study we observed by EdU incorporation that DNA replication has already occurred at the conventional microinjection time (20 hpi). Aiming to reduce mosaicism appearance, we have developed three alternative microinjection protocols: early zygote microinjection (10 hpi RNA) or oocyte microinjection before fertilization with either RNA or Ribonucleoprotein delivery (0 hpi RNA or 0 hpi RNP). All three alternative microinjection protocols resulted in similar blastocyst and genome edition rates compared to the conventional 20 hpi group, whereas mosaicism rates were significantly reduced in all early delivery groups (~10-30% of edited embryos being mosaic depending on the loci) compared to conventional 20 hpi microinjection (100% mosaicism rate). These strategies constitute an efficient way to reduce the number of indels, increasing the odds for direct KO generation.
Collapse
Affiliation(s)
| | | | | | | | - O Ousinde
- Animal Reproduction Department, INIA, Madrid, Spain
| | | |
Collapse
|
30
|
Perota A, Galli C. N-Glycolylneuraminic Acid (Neu5Gc) Null Large Animals by Targeting the CMP-Neu5Gc Hydroxylase (CMAH). Front Immunol 2019; 10:2396. [PMID: 31681287 PMCID: PMC6803385 DOI: 10.3389/fimmu.2019.02396] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 09/24/2019] [Indexed: 01/05/2023] Open
Abstract
The two major sialic acids described in mammalian cells are the N-glycolylneuraminic acid (Neu5Gc) and the N-acetylneuraminic acid (Neu5Ac). Neu5Gc synthesis starts from the N-acetylneuraminic acid (Neu5Ac) precursor modified by an hydroxylic group addition catalyzed by CMP-Neu5Ac hydroxylase enzyme (CMAH). In humans, CMAH was inactivated by a 92 bp deletion occurred 2-3 million years ago. Few other mammals do not synthetize Neu5Gc, however livestock species used for food production and as a source of biological materials for medical applications carry Neu5Gc. Trace amounts of Neu5Gc are up taken through the diet and incorporated into various tissues including epithelia and endothelia cells. Humans carry "natural," diet-induced Anti-Neu5Gc antibodies and when undertaking medical treatments or receiving transplants or devices that contain animal derived products they can cause immunological reaction affecting pharmacology, immune tolerance, and severe side effect like serum sickness disease (SSD). Neu5Gc null mice have been the main experimental model to study such phenotype. With the recent advances in genome editing, pigs and cattle KO for Neu5Gc have been generated always in association with the αGal KO. These large animals are normal and fertile and provide additional experimental models to study such mutation. Moreover, they will be the base for the development of new therapeutic applications like polyclonal IgG immunotherapy, Bioprosthetic Heart Valves, cells and tissues replacement.
Collapse
Affiliation(s)
- Andrea Perota
- Laboratory of Reproductive Technologies, Avantea, Cremona, Italy
| | - Cesare Galli
- Laboratory of Reproductive Technologies, Avantea, Cremona, Italy.,Fondazione Avantea, Cremona, Italy
| |
Collapse
|
31
|
Martinez CA, Cambra JM, Parrilla I, Lucas X, Rodriguez-Martinez H, Martinez EA, Izpisua JC, Cuello C, Gil MA. Three-to-5-day weaning-to-estrus intervals do not affect neither efficiency of collection nor in vitro developmental ability of in vivo-derived pig zygotes. Theriogenology 2019; 141:48-53. [PMID: 31518728 DOI: 10.1016/j.theriogenology.2019.09.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 09/03/2019] [Accepted: 09/04/2019] [Indexed: 12/11/2022]
Abstract
An efficient system to collect large numbers of vital zygotes is a pre-requisite for application of zygote genome-editing technology, including development of efficient models for xenotransplantation using pigs. Owing to the sub-optimal in vitro production of zygotes in pigs, efficient collection of in vivo developed zygotes is required. Timing of ovulation is a key factor to sustain efficiency since the interval between pronuclear formation and the first division is very short in pigs. The weaning-to-estrus interval can, due to its inverse relation with length of estrus and time of ovulation, interfere with ovulation and make it asynchronous, which reduces the probability of obtaining zygotes. This retrospective study compared the effects of three weaning-to-estrus intervals of 3, 4 or 5 days on zygote collection efficiency in a total of 17 trials over a 3-year period including 223 sows. Donor sows in groups of 10-15 animals were super-ovulated with eCG 24 h after weaning and those in estrus at 48-72 h post-eCG were immediately treated with hCG, followed by insemination 6 and 24 h thereafter. Collected structures during laparotomy on Day 2 (Day 0: onset of estrus) were morphologically evaluated and only those with a single cell and two visible polar bodies were considered as zygotes. Zygotes were injected with CRISPR-Cas9 editor mixture and cultured for 6 days to evaluate their developmental ability against non-injected control zygotes. Of all recovered structures (N = 5,468), 67.4%, 30.8% and 1.8% were zygotes, 2-cell embryos and oocytes-degenerated embryos, respectively. The different weaning-to-estrus intervals did not affect either the percentages of collected zygotes (range: 64.1%-70.0%) or the percentages of sows with zygotes at collection time (range: 69.0%-73.3%). The weaning-to-estrus intervals did not affect the in vitro developmental ability of zygotes. After 24 h of culture, 78.1 ± 2.0% and 95.1 ± 0.6 (P < 0.05) of injected (N = 2,345) and non-injected (N = 335) zygotes, respectively, developed to 2-to-4-cell embryo stage. The total efficiency of the system was 64.1 ± 2.2% and 85.8 ± 1.5% (P < 0.05) for injected and non-injected zygotes, respectively. In conclusion, the results indicate that neither the efficiency of collecting in vivo derived porcine zygotes from superovulated sows nor the zygote ability to develop to blastocyst after cytoplasmic genome-editing injection were affected by a weaning-to-estrus interval between 3-to-5 days.
Collapse
Affiliation(s)
- C A Martinez
- Department of Medicine and Animal Surgery, Faculty of Veterinary Medicine, International Excellence Campus for Higher Education and Research "Campus Mare Nostrum", University of Murcia, 30100, Murcia, Spain; Institute for Biomedical Research of Murcia (IMIB-Arrixaca), Campus de Ciencias de La Salud, Carretera Buenavista S/n, 30120 El, Palmar, Murcia, Spain
| | - J M Cambra
- Department of Medicine and Animal Surgery, Faculty of Veterinary Medicine, International Excellence Campus for Higher Education and Research "Campus Mare Nostrum", University of Murcia, 30100, Murcia, Spain; Institute for Biomedical Research of Murcia (IMIB-Arrixaca), Campus de Ciencias de La Salud, Carretera Buenavista S/n, 30120 El, Palmar, Murcia, Spain
| | - I Parrilla
- Department of Medicine and Animal Surgery, Faculty of Veterinary Medicine, International Excellence Campus for Higher Education and Research "Campus Mare Nostrum", University of Murcia, 30100, Murcia, Spain; Institute for Biomedical Research of Murcia (IMIB-Arrixaca), Campus de Ciencias de La Salud, Carretera Buenavista S/n, 30120 El, Palmar, Murcia, Spain
| | - X Lucas
- Department of Medicine and Animal Surgery, Faculty of Veterinary Medicine, International Excellence Campus for Higher Education and Research "Campus Mare Nostrum", University of Murcia, 30100, Murcia, Spain; Institute for Biomedical Research of Murcia (IMIB-Arrixaca), Campus de Ciencias de La Salud, Carretera Buenavista S/n, 30120 El, Palmar, Murcia, Spain
| | - H Rodriguez-Martinez
- Department of Clinical & Experimental Medicine (IKE), Linköping University, Campus US, 58183, Linköping, Sweden
| | - E A Martinez
- Department of Medicine and Animal Surgery, Faculty of Veterinary Medicine, International Excellence Campus for Higher Education and Research "Campus Mare Nostrum", University of Murcia, 30100, Murcia, Spain; Institute for Biomedical Research of Murcia (IMIB-Arrixaca), Campus de Ciencias de La Salud, Carretera Buenavista S/n, 30120 El, Palmar, Murcia, Spain
| | - J C Izpisua
- Salk Institute for Biological Studies, 10010 N. Torrey Pines Rd., La Jolla, CA, 92037, USA
| | - C Cuello
- Department of Medicine and Animal Surgery, Faculty of Veterinary Medicine, International Excellence Campus for Higher Education and Research "Campus Mare Nostrum", University of Murcia, 30100, Murcia, Spain; Institute for Biomedical Research of Murcia (IMIB-Arrixaca), Campus de Ciencias de La Salud, Carretera Buenavista S/n, 30120 El, Palmar, Murcia, Spain.
| | - M A Gil
- Department of Medicine and Animal Surgery, Faculty of Veterinary Medicine, International Excellence Campus for Higher Education and Research "Campus Mare Nostrum", University of Murcia, 30100, Murcia, Spain; Institute for Biomedical Research of Murcia (IMIB-Arrixaca), Campus de Ciencias de La Salud, Carretera Buenavista S/n, 30120 El, Palmar, Murcia, Spain
| |
Collapse
|
32
|
García-Martínez S, Sánchez Hurtado MA, Gutiérrez H, Sánchez Margallo FM, Romar R, Latorre R, Coy P, López Albors O. Mimicking physiological O2 tension in the female reproductive tract improves assisted reproduction outcomes in pig. Mol Hum Reprod 2019; 24:260-270. [PMID: 29490063 DOI: 10.1093/molehr/gay008] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 02/26/2018] [Indexed: 01/20/2023] Open
Abstract
STUDY QUESTION Is O2 tension in the pig oviduct and uterus affected by the estrous cycle stage and the animal's age, and can the outcome of in vitro embryo development be improved by mimicking these physiological values? SUMMARY ANSWER O2 tension within the pig reproductive organs is affected by the animal's age, and values close to those measured in vivo have a positive impact on embryo development and quality when used during IVF and embryo culture (EC). WHAT IS KNOWN ALREADY To obtain a healthy embryo in vitro, it is necessary to adopt a culture microenvironment that approximates physiological conditions. Despite advances in surgical procedures and sensitive probes that allow accurate assessment of in vivo O2 tension, few such studies have been conducted recently in mammals. In addition, no reference values of physiological O2 tension in the reproductive tract exist for large animal models such as pig, and the effect of O2 tension on ART outcomes is unknown. STUDY DESIGN, SIZE, DURATION This study was conducted in pigs. We measured oviductal and uterine O2 tension (n = 29 and 13, respectively) and then examined how the use of the physiological values in pig IVF and EC affected pig ART output (n = 1447 oocytes). PARTICIPANTS/MATERIALS, SETTING, METHODS The oviductal and uterine O2 tension at the different stages of the estrous cycle was monitored using a laparo-endoscopic single-site surgery (LESS) assisted approach along with a flexible and thin miniaturized luminescent probe. Two groups of pigs, Large-white × Landrace breed, were used: for the first group, 16 pre-pubertal gilts (5 months old and 95 kg) were induced to ovulate with equine chorionic gonadotropin (eCG) and human chorionic gonadotropin (hCG); in the second group 13 mature sows (24-48 months and 185 kg) were used. IVF and EC were performed at two different O2 tensions: Atmospheric O2 (20%) and the mean in vivo value measured (7%). At 18-20 h post-insemination (hpi), a small sample of presumptive zygotes were fixed, stained and examined under epifluorescence microscopy to assess the fertilization rates. At 48 hpi, cleavage was evaluated under the stereomicroscope. Finally, at 180 hpi, development to the blastocyst stage was quantified, blastocyst morphology was assessed, and embryos were fixed and stained to count the mean cell number per blastocyst. MAIN RESULTS AND THE ROLE OF CHANCE The mean O2 content within the pig oviduct and uterus was always lower than in ambient air. The average O2 percentage was higher in gilts (10.0%) than in sows (7.6%) (P < 0.0001). The cleavage rate of porcine in vitro fertilized embryos maintained under 7% O2 during IVF and EC was significantly higher (60.0 ± 2.3) compared with those cultured under 20% O2 (32.0 ± 2.2) (P < 0.05). An increase in the number of cells in embryos cultured under the low O2 concentration (88.9 ± 5.9) was observed compared to those cultured under 20% O2 (59.0 ± 5.0) (P < 0.05). LARGE SCALE DATA None. LIMITATIONS, REASONS FOR CAUTION Although minimally invasive surgery was used the effect of anesthesia and manipulations on O2 tension within the organs are unknown. WIDER IMPLICATIONS OF THE FINDINGS Using physiological oxygen concentrations in IVF/EC could improve ART outcomes. STUDY FUNDING AND COMPETING INTEREST(S) This study was funded by Spanish Ministry of Economy and Competitiveness (MINECO) and European Regional Development Fund (FEDER). Grants AGL2012-40180-C03-01 and AGL2015-66341-R. The authors declare no conflict of interest.
Collapse
Affiliation(s)
- S García-Martínez
- Dept. Physiology, Facultad de Veterinaria, Campus Internacional de Excelencia de Educación Superior (Campus Mare Nostrum), Universidad de Murcia, Murcia 30100, España.,Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca (IMIB-Arrixaca), Murcia, España
| | - M A Sánchez Hurtado
- Laparoscopy Unit, Minimally Invasive Surgery Center Jesús Usón, Cáceres 10071, España
| | - H Gutiérrez
- Dept. Animal Health, Faculty of Veterinary Medicine and Zootechnics, University Nacional of Colombia, Carrera 30 45-03, Bogotá, Colombia.,Dept. Anatomy & Comparative Physiology, Facultad de Veterinaria, Campus Internacional de Excelencia de Educación Superior (Campus Mare Nostrum), Universidad de Murcia, Murcia 30100, España
| | - F M Sánchez Margallo
- Laparoscopy Unit, Minimally Invasive Surgery Center Jesús Usón, Cáceres 10071, España
| | - R Romar
- Dept. Physiology, Facultad de Veterinaria, Campus Internacional de Excelencia de Educación Superior (Campus Mare Nostrum), Universidad de Murcia, Murcia 30100, España.,Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca (IMIB-Arrixaca), Murcia, España
| | - R Latorre
- Dept. Anatomy & Comparative Physiology, Facultad de Veterinaria, Campus Internacional de Excelencia de Educación Superior (Campus Mare Nostrum), Universidad de Murcia, Murcia 30100, España
| | - P Coy
- Dept. Physiology, Facultad de Veterinaria, Campus Internacional de Excelencia de Educación Superior (Campus Mare Nostrum), Universidad de Murcia, Murcia 30100, España.,Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca (IMIB-Arrixaca), Murcia, España
| | - O López Albors
- Dept. Anatomy & Comparative Physiology, Facultad de Veterinaria, Campus Internacional de Excelencia de Educación Superior (Campus Mare Nostrum), Universidad de Murcia, Murcia 30100, España
| |
Collapse
|
33
|
Zhao J, Lai L, Ji W, Zhou Q. Genome editing in large animals: current status and future prospects. Natl Sci Rev 2019; 6:402-420. [PMID: 34691891 PMCID: PMC8291540 DOI: 10.1093/nsr/nwz013] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 01/09/2019] [Accepted: 01/30/2019] [Indexed: 12/14/2022] Open
Abstract
Large animals (non-human primates, livestock and dogs) are playing important roles in biomedical research, and large livestock animals serve as important sources of meat and milk. The recently developed programmable DNA nucleases have revolutionized the generation of gene-modified large animals that are used for biological and biomedical research. In this review, we briefly introduce the recent advances in nuclease-meditated gene editing tools, and we outline these editing tools' applications in human disease modeling, regenerative medicine and agriculture. Additionally, we provide perspectives regarding the challenges and prospects of the new genome editing technology.
Collapse
Affiliation(s)
- Jianguo Zhao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing 100049, China
- Institute of Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
| | - Liangxue Lai
- South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Weizhi Ji
- Yunnan Key Laboratory of Primate Biomedicine Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming 650500, China
- CAS Center for Excellence in Brain Science and Intelligence Technology (CEBSIT), Shanghai 200031, China
| | - Qi Zhou
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing 100049, China
- Institute of Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
| |
Collapse
|
34
|
Hirata M, Tanihara F, Wittayarat M, Hirano T, Nguyen NT, Le QA, Namula Z, Nii M, Otoi T. Genome mutation after introduction of the gene editing by electroporation of Cas9 protein (GEEP) system in matured oocytes and putative zygotes. In Vitro Cell Dev Biol Anim 2019; 55:237-242. [PMID: 30820813 DOI: 10.1007/s11626-019-00338-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2018] [Accepted: 02/13/2019] [Indexed: 11/30/2022]
Abstract
The application of CRISPR/Cas9 strategy promises to rapidly increase the production of genetically engineered animals since it yields stably integrated transgenes. In the present study, we investigated the efficiency of target mutations after electroporation with the CRISPR/Cas9 system using sgRNAs to target the MSTN or FGF10 genes in porcine-matured oocytes and putative zygotes. Effects of pulse number (3-7 pulse repetitions) during electroporation on the embryonic development and mutation efficiency were also investigated. Our results showed that the cleavage rate of matured oocytes with electroporation treatment significantly decreased as compared with electroporated putative zygotes (p < 0.05). Moreover, the rates of blastocyst formation from oocytes/zygotes electroporated with more than 5 pulses decreased. Mutation efficiency was then assessed after sequencing the target sites in individual blastocysts derived from oocytes/zygotes electroporated by 3 and 5 pulses. No bi-allelic mutations in all examined blastocysts were observed in this study. There were no differences in the mutation rates (50-60%) between blastocysts derived from matured oocytes electroporated by 3 and 5 pulses, irrespective of targeting gene. In the targeting MSTN gene, however, the mutation rate (12.5%) of blastocysts derived from putative zygotes electroporated by 3 pulses tended to be lower than that (60%) from 5-pulsed electroporated putative zygotes. These data indicate that the type of eggs may influence not only their development after electroporation treatment but also the mutation rate in the resulting blastocysts.
Collapse
Affiliation(s)
- Maki Hirata
- Faculty of Bioscience and Bioindustry, Tokushima University, Tokushima, Japan
| | - Fuminori Tanihara
- Faculty of Bioscience and Bioindustry, Tokushima University, Tokushima, Japan.
| | - Manita Wittayarat
- Faculty of Veterinary Science, Prince of Songkla University, Songkhla, Thailand
| | - Takayuki Hirano
- Faculty of Bioscience and Bioindustry, Tokushima University, Tokushima, Japan
| | - Nhien Thi Nguyen
- Faculty of Bioscience and Bioindustry, Tokushima University, Tokushima, Japan
| | - Quynh Anh Le
- Faculty of Bioscience and Bioindustry, Tokushima University, Tokushima, Japan
| | - Zhao Namula
- Faculty of Veterinary Science, Guangdong Ocean University, Zhanjiang, China
| | - Masahiro Nii
- Tokushima Prefectural Livestock Research Institute, Tokushima, Japan
| | - Takeshige Otoi
- Faculty of Bioscience and Bioindustry, Tokushima University, Tokushima, Japan
| |
Collapse
|
35
|
Zarei A, Razban V, Hosseini SE, Tabei SMB. Creating cell and animal models of human disease by genome editing using CRISPR/Cas9. J Gene Med 2019; 21:e3082. [DOI: 10.1002/jgm.3082] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 02/02/2019] [Accepted: 02/02/2019] [Indexed: 12/26/2022] Open
Affiliation(s)
- Ali Zarei
- Department of Molecular Genetics, Marvdasht BranchIslamic Azad University Marvdasht Iran
- Department of Molecular Genetics, Science and Research BranchIslamic Azad University Fars Iran
| | - Vahid Razban
- Department of Molecular medicine, School of Advanced Medical Sciences and Technologies Shiraz Iran
- Stem Cell and Transgenic Technology Research CenterShiraz University of Medical Sciences Shiraz Iran
| | | | | |
Collapse
|
36
|
Thi Nguyen N, Hirata M, Tanihara F, Hirano T, Le QA, Nii M, Otoi T. Hypothermic storage of porcine zygotes in serum supplemented with chlorogenic acid. Reprod Domest Anim 2019; 54:750-755. [PMID: 30788874 DOI: 10.1111/rda.13417] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 02/08/2019] [Indexed: 12/14/2022]
Abstract
The current study was conducted to investigate the effects of 100% foetal bovine serum (FBS) and 100% porcine follicular fluid (pFF) as a storage medium on the developmental competence of porcine zygotes stored at 25°C for 24 hr. Moreover, we evaluated the additive effects of chlorogenic acid (CGA) in the storage medium. When in vitro-produced zygotes were stored at 25°C for 24 hr in tubes containing either tissue culture medium (TCM) 199 supplemented with 1 mg/ml bovine serum albumin (BSA), 100% of FBS or 100% of pFF, the rate of blastocyst formation was significantly higher in 100% of FBS than in BSA-containing TCM 199. When the effects of CGA supplementation in 100% of FBS on the development of zygotes stored at 25°C for 24 hr was evaluated, more zygotes stored with 50 µM CGA developed to blastocysts compared with the other concentrations of CGA. When the formation date and quality of blastocysts derived from zygotes stored in 100% of FBS supplemented with 50 µM CGA were investigated, the highest ratio of blastocysts formation in the storage group appeared 1 day later than in the non-stored control group. However, a higher proportion of blastocysts with apoptotic nuclei was observed in the stored group as compared to the non-stored group. In conclusion, 100% of FBS is available for a short storage medium of porcine zygotes. The supplementation of 50 µM CGA into the storage medium improves the rates of blastocyst formation of zygotes after storage, but the quality of embryos from the stored zygotes remains to be improved.
Collapse
Affiliation(s)
- Nhien Thi Nguyen
- Faculty of Bioscience and Bioindustry, Tokushima University, Tokushima, Japan
| | - Maki Hirata
- Faculty of Bioscience and Bioindustry, Tokushima University, Tokushima, Japan
| | - Fuminori Tanihara
- Faculty of Bioscience and Bioindustry, Tokushima University, Tokushima, Japan
| | - Takayuki Hirano
- Faculty of Bioscience and Bioindustry, Tokushima University, Tokushima, Japan
| | - Quynh Anh Le
- Faculty of Bioscience and Bioindustry, Tokushima University, Tokushima, Japan
| | - Masahiro Nii
- Tokushima Prefectural Livestock Research Institute, Tokushima, Japan
| | - Takeshige Otoi
- Faculty of Bioscience and Bioindustry, Tokushima University, Tokushima, Japan
| |
Collapse
|
37
|
Tanihara F, Hirata M, Nguyen NT, LE QA, Hirano T, Otoi T. Effects of concentration of CRISPR/Cas9 components on genetic mosaicism in cytoplasmic microinjected porcine embryos. J Reprod Dev 2019; 65:209-214. [PMID: 30726783 PMCID: PMC6584178 DOI: 10.1262/jrd.2018-116] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Cytoplasmic microinjection (CI) of the CRISPR/Cas9 system enabled the induction of site-specific mutations in porcine zygotes and resulting pigs. However, mosaicism is a serious problem for genetically modified pigs. In the present study, we investigated suitable timing and concentration of CRISPR/Cas9 components for introduction into oocytes/zygotes by CI, to reduce mosaicism in the resulting blastocysts. First, we introduced 20 ng/μl of Cas9 protein and guide RNA (gRNA), targeting the α-1,3-galactosyltransferase (GalT) gene in oocytes before in vitro fertilization (IVF), in zygotes after IVF, or in oocytes/zygotes before and after IVF, twice. CI treatment had no detrimental effects on blastocyst formation rates. The highest value of the rate of mutant blastocysts was observed in zygotes injected after IVF. Next, we injected Cas9 protein and gRNA into zygotes after IVF at a concentration of 20 ng/μl each (20 ng/μl group) or 100 ng/μl each (100 ng/μl group). The ratio of the number of blastocysts that carried mutations to the total number of blastocysts examined in the 100 ng/μl group was significantly higher (P < 0.05) than that in the 20 ng/μl group. Although no blastocysts from the 20 ng/μl group carried a biallelic mutation, 16.7% of blastocysts from the 100 ng/μl group carried a biallelic mutation. In conclusion, increasing the concentration of Cas9 protein and gRNA is effective in generating biallelic mutant blastocysts. To reduce mosaicism, however, further optimization of the timing of CI, and the concentration of CRISPR/Cas9 components, is needed.
Collapse
Affiliation(s)
- Fuminori Tanihara
- Faculty of Bioscience and Bioindustry, Tokushima University, Tokushima 770-8513, Japan
| | - Maki Hirata
- Faculty of Bioscience and Bioindustry, Tokushima University, Tokushima 770-8513, Japan
| | - Nhien Thi Nguyen
- Faculty of Bioscience and Bioindustry, Tokushima University, Tokushima 770-8513, Japan
| | - Quynh Anh LE
- Faculty of Bioscience and Bioindustry, Tokushima University, Tokushima 770-8513, Japan
| | - Takayuki Hirano
- Faculty of Bioscience and Bioindustry, Tokushima University, Tokushima 770-8513, Japan
| | - Takeshige Otoi
- Faculty of Bioscience and Bioindustry, Tokushima University, Tokushima 770-8513, Japan
| |
Collapse
|
38
|
Ding Y, Zhu S, Wu C, Qian L, Li D, Wang L, Wang Y, Zhang W, Yang M, Ding J, Wu X, Zhang X, Gao Y, Yin Z. Relationship between porcine miR-20a and its putative target low-density lipoprotein receptor based on dual luciferase reporter gene assays. ASIAN-AUSTRALASIAN JOURNAL OF ANIMAL SCIENCES 2019; 32:922-929. [PMID: 30744358 PMCID: PMC6601058 DOI: 10.5713/ajas.18.0510] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Accepted: 12/13/2018] [Indexed: 12/28/2022]
Abstract
Objective Mutations in low-density lipoprotein receptor (LDLR), which encodes a critical protein for cholesterol homeostasis and lipid metabolism in mammals, are involved in cardiometabolic diseases, such as familial hypercholesterolemia in pigs. Whereas microRNAs (miRNAs) can control LDLR regulation, their involvement in circulating cholesterol and lipid levels with respect to cardiometabolic diseases in pigs is unclear. We aimed to identify and analyze LDLR as a potential target gene of SSC-miR-20a. Methods Bioinformatic analysis predicted that porcine LDLR is a target of SSC-miR-20a. Wild-type and mutant LDLR 3′-untranslated region (UTR) fragments were generated by polymerase chain reaction (PCR) and cloned into the pGL3-Control vector to construct pGL3 Control LDLR wild-3′-UTR and pGL3 Control LDLR mutant-3′-UTR recombinant plasmids, respectively. An miR-20a expression plasmid was constructed by inserting the porcine pre-miR-20a-coding sequence between the HindIII and BamHI sites in pMR-mCherry, and constructs were confirmed by sequencing. HEK293T cells were co-transfected with the miR-20a expression or pMR-mCherry control plasmids and constructs harboring the corresponding 3′-UTR, and relative luciferase activity was determined. The relative expression levels of miR-20a and LDLR mRNA and their correlation in terms of expression levels in porcine liver tissue were analyzed using reverse-transcription quantitative PCR. Results Gel electrophoresis and sequencing showed that target gene fragments were successfully cloned, and the three recombinant vectors were successfully constructed. Compared to pMR-mCherry, the miR-20a expression vector significantly inhibited wild-type LDLR-3′-UTR-driven (p<0.01), but not mutant LDLR-3′-UTR-driven (p>0.05), luciferase reporter activity. Further, miR-20a and LDLR were expressed at relatively high levels in porcine liver tissues. Pearson correlation analysis revealed that porcine liver miR-20a and LDLR levels were significantly negatively correlated (r = −0.656, p<0.05). Conclusion LDLR is a potential target of miR-20a, which might directly bind the LDLR 3′-UTR to post-transcriptionally inhibit expression. These results have implications in understanding the pathogenesis and progression of porcine cardiovascular diseases.
Collapse
Affiliation(s)
- Yueyun Ding
- Anhui Provincial Laboratory of Local Animal Genetic Resource Conservation and Bio-Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Shujiao Zhu
- Anhui Provincial Laboratory of Local Animal Genetic Resource Conservation and Bio-Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Chaodong Wu
- Anhui Provincial Laboratory of Local Animal Genetic Resource Conservation and Bio-Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Li Qian
- Anhui Provincial Laboratory of Local Animal Genetic Resource Conservation and Bio-Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - DengTao Li
- Anhui Provincial Laboratory of Local Animal Genetic Resource Conservation and Bio-Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Li Wang
- Anhui Provincial Laboratory of Local Animal Genetic Resource Conservation and Bio-Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Yuanlang Wang
- Anhui Provincial Laboratory of Local Animal Genetic Resource Conservation and Bio-Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Wei Zhang
- Anhui Provincial Laboratory of Local Animal Genetic Resource Conservation and Bio-Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Min Yang
- Anhui Provincial Laboratory of Local Animal Genetic Resource Conservation and Bio-Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Jian Ding
- Anhui Provincial Laboratory of Local Animal Genetic Resource Conservation and Bio-Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Xudong Wu
- Anhui Provincial Laboratory of Local Animal Genetic Resource Conservation and Bio-Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Xiaodong Zhang
- Anhui Provincial Laboratory of Local Animal Genetic Resource Conservation and Bio-Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Yafei Gao
- Anhui Haoxiang Agriculture And Animal Husbandry Co. LTD, Bozhou, Anhui 236700, China
| | - Zongjun Yin
- Anhui Provincial Laboratory of Local Animal Genetic Resource Conservation and Bio-Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, China
| |
Collapse
|
39
|
Patel S, Athirasala A, Menezes PP, Ashwanikumar N, Zou T, Sahay G, Bertassoni LE. Messenger RNA Delivery for Tissue Engineering and Regenerative Medicine Applications. Tissue Eng Part A 2019; 25:91-112. [PMID: 29661055 PMCID: PMC6352544 DOI: 10.1089/ten.tea.2017.0444] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 04/09/2018] [Indexed: 12/25/2022] Open
Abstract
The ability to control cellular processes and precisely direct cellular reprogramming has revolutionized regenerative medicine. Recent advances in in vitro transcribed (IVT) mRNA technology with chemical modifications have led to development of methods that control spatiotemporal gene expression. Additionally, there is a current thrust toward the development of safe, integration-free approaches to gene therapy for translational purposes. In this review, we describe strategies of synthetic IVT mRNA modifications and nonviral technologies for intracellular delivery. We provide insights into the current tissue engineering approaches that use a hydrogel scaffold with genetic material. Furthermore, we discuss the transformative potential of novel mRNA formulations that when embedded in hydrogels can trigger controlled genetic manipulation to regenerate tissues and organs in vitro and in vivo. The role of mRNA delivery in vascularization, cytoprotection, and Cas9-mediated xenotransplantation is additionally highlighted. Harmonizing mRNA delivery vehicle interactions with polymeric scaffolds can be used to present genetic cues that lead to precise command over cellular reprogramming, differentiation, and secretome activity of stem cells-an ultimate goal for tissue engineering.
Collapse
Affiliation(s)
- Siddharth Patel
- Department of Pharmaceutical Sciences, College of Pharmacy, Collaborative Life Science Building, Oregon State University, Portland, Oregon
| | - Avathamsa Athirasala
- Division of Biomaterials and Biomechanics, Department of Restorative Dentistry, School of Dentistry, Oregon Health and Science University, Portland, Oregon
| | - Paula P. Menezes
- Division of Biomaterials and Biomechanics, Department of Restorative Dentistry, School of Dentistry, Oregon Health and Science University, Portland, Oregon
- Postgraduate Program in Health Sciences, Department of Pharmacy, Federal University of Sergipe, Aracaju, Sergipe, Brazil
| | - N. Ashwanikumar
- Department of Pharmaceutical Sciences, College of Pharmacy, Collaborative Life Science Building, Oregon State University, Portland, Oregon
| | - Ting Zou
- Division of Biomaterials and Biomechanics, Department of Restorative Dentistry, School of Dentistry, Oregon Health and Science University, Portland, Oregon
- Endodontology, Faculty of Dentistry, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Gaurav Sahay
- Department of Pharmaceutical Sciences, College of Pharmacy, Collaborative Life Science Building, Oregon State University, Portland, Oregon
- Department of Biomedical Engineering, Collaborative Life Science Building, Oregon Health and Science University, Portland, Oregon
| | - Luiz E. Bertassoni
- Division of Biomaterials and Biomechanics, Department of Restorative Dentistry, School of Dentistry, Oregon Health and Science University, Portland, Oregon
- Department of Biomedical Engineering, Collaborative Life Science Building, Oregon Health and Science University, Portland, Oregon
- Center for Regenerative Medicine, Oregon Health and Science University, Portland, Oregon
| |
Collapse
|
40
|
Abstract
Though making up nearly half of the known CRISPR-Cas9 family of enzymes, the Type II-C CRISPR-Cas9 has been underexplored for their molecular mechanisms and potential in safe gene editing applications. In comparison with the more popular Type II-A CRISPR-Cas9, the Type II-C enzymes are generally smaller in size and utilize longer base pairing in identification of their DNA substrates. These characteristics suggest easier portability and potentially less off-targets for Type II-C in gene editing applications. We describe identification and biochemical characterization of a thermophilic Type II-C CRISPR-Cas from Acidothermus cellulolyticus (AceCas9). We describe several library-based methods that enabled us to identify the PAM sequence and elements critical to protospacer mismatch surveillance of AceCas9.
Collapse
|
41
|
Hand TH, Das A, Roth MO, Smith CL, Jean-Baptiste UL, Li H. Phosphate Lock Residues of Acidothermus cellulolyticus Cas9 Are Critical to Its Substrate Specificity. ACS Synth Biol 2018; 7:2908-2917. [PMID: 30458109 PMCID: PMC6525624 DOI: 10.1021/acssynbio.8b00455] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Despite being utilized widely in genome sciences, CRISPR-Cas9 remains limited in achieving high fidelity in cleaving DNA. A better understanding of the molecular basis of Cas9 holds the key to improve Cas9-based tools. We employed direct evolution and in vitro characterizations to explore structural parameters that impact the specificity of the thermophilic Cas9 from Acidothermus cellulolyticus (AceCas9). By identifying variants that are able to cleave mismatched protospacers within the seed region, we found a critical role of the phosphate lock residues in substrate specificity in a manner that depends on their sizes and charges. Removal of the negative charge from the phosphate lock residues significantly decreases sensitivity to the guide-DNA mismatches. An increase in size of the substituted residues further reduces the sensitivity to mismatches at the first position of the protospacer. Our findings identify the phosphate lock residues as an important site for tuning the specificity and catalytic efficiency of Cas9.
Collapse
Affiliation(s)
- Travis H. Hand
- Institute of Molecular Biophysics, Florida State University, Tallahassee, FL 32306, USA
| | - Anuska Das
- Institute of Molecular Biophysics, Florida State University, Tallahassee, FL 32306, USA
| | - Mitchell O. Roth
- Institute of Molecular Biophysics, Florida State University, Tallahassee, FL 32306, USA
| | - Chardasia L. Smith
- Department of Chemistry and Biochemistry, Florida State University, Tallahassee, FL 32306, USA
| | - Uriel L. Jean-Baptiste
- Department of Chemistry and Biochemistry, Florida State University, Tallahassee, FL 32306, USA
| | - Hong Li
- Institute of Molecular Biophysics, Florida State University, Tallahassee, FL 32306, USA
- Department of Chemistry and Biochemistry, Florida State University, Tallahassee, FL 32306, USA
| |
Collapse
|
42
|
Characterization of Three Generations of Transgenic Pigs Expressing the HLA-E Gene. ANNALS OF ANIMAL SCIENCE 2018. [DOI: 10.2478/aoas-2018-0034] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Abstract
The use of pigs as a source of organs and tissues for xenotransplantation can overcome the growing shortage of human donors. Human NK cells play an important role in the cell-mediated rejection of pig-to-human xenografts. In this paper we report the generation and extensive characterization of three generations of transgenic pigs with HLA-E gene encoding the antigen which can inhibit the human NK cell-mediated response. The gene construct pHLAE-GFPBsd containing the human gene encoding the human leukocyte antigen under the promoter of the EF-1α elongation factor ensuring systemic expression was introduced by microinjection into a pronucleus of the fertilized porcine oocyte. PCR analysis revealed and FISH analysis confirmed that the pHLAE-GFPBsd gene construct was present in the genome of the founder female pig. As a result of inter-breeding, an additional 7 transgenic animals were obtained (one individual from F1 generation and six individuals from F2 generation). The transgene expression was shown by RT-PCR and flow cytometry. Real Time PCR analysis estimated the approximate number of transgene copies at 16–34. Karyotype analysis did not show any changes in the structure or the number of chromosomes. The expression level of the transgene was stable in the next generation of genetically modified pigs. An NK cell-mediated cytotoxicity assay showed the increased viability of the transgenic cells in comparison with the wild-type, which confirmed the protective influence of HLA-E expression.
Collapse
|
43
|
Tian H, Luo J, Zhang Z, Wu J, Zhang T, Busato S, Huang L, Song N, Bionaz M. CRISPR/Cas9-mediated Stearoyl-CoA Desaturase 1 (SCD1) Deficiency Affects Fatty Acid Metabolism in Goat Mammary Epithelial Cells. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2018; 66:10041-10052. [PMID: 30180552 DOI: 10.1021/acs.jafc.8b03545] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Stearoyl-CoA desaturase 1 (SCD1) is a fatty acid desaturase catalyzing cis-double-bond formation in the Δ9 position to produce monounsaturated fatty acids essential for the synthesis of milk fat. Previous studies using RNAi methods have provided support for a role of SCD1 in goat mammary epithelial cells (GMEC); however, RNAi presents several limitations that might preclude a truthful understanding of the biological function of SCD1. To explore the function of SCD1 on fatty acid metabolism in GMEC, we used CRISPR-Cas9-mediated SCD1 knockout through non-homologous end-joining (NHEJ) and homology-directed repair (HDR) pathways in GMEC. We successfully introduced nucleotide deletions and mutations in the SCD1 gene locus through the NHEJ pathway and disrupted its second exon via insertion of an EGFP-PuroR segment using the HDR pathway. In clones derived from the latter, gene- and protein-expression data indicated that we obtained a monoallelic SCD1 knockout. A T7EN1-mediated assay revealed no off-targets in the surveyed sites. The contents of triacylglycerol and cholesterol and the desaturase index were significantly decreased as a consequence of SCD1 knockout. The deletion of SCD1 decreased the expression of other genes involved in de novo fatty acid synthesis, including SREBF1 and FASN, as well the fatty acid transporters FABP3 and FABP4. The downregulation of these genes partly explains the decrease of intracellular triacylglycerols. Our results indicate a successful SCD1 knockout in goat mammary cells using CRISPR-Cas9. The demonstration of the successful use of CRISPR-Cas9 in GMEC is an important step to producing transgenic goats to study mammary biology in vivo.
Collapse
Affiliation(s)
- Huibin Tian
- Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology , Northwest A&F University , Yangling 712100 , China
| | - Jun Luo
- Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology , Northwest A&F University , Yangling 712100 , China
| | - Zhifei Zhang
- Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology , Northwest A&F University , Yangling 712100 , China
| | - Jiao Wu
- Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology , Northwest A&F University , Yangling 712100 , China
| | - Tianying Zhang
- Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology , Northwest A&F University , Yangling 712100 , China
| | - Sebastiano Busato
- Department of Animal and Rangeland Sciences , Oregon State University , Corvallis , Oregon 97331 , United States
| | - Lian Huang
- Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology , Northwest A&F University , Yangling 712100 , China
| | - Ning Song
- Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology , Northwest A&F University , Yangling 712100 , China
| | - Massimo Bionaz
- Department of Animal and Rangeland Sciences , Oregon State University , Corvallis , Oregon 97331 , United States
| |
Collapse
|
44
|
Lamas-Toranzo I, Ramos-Ibeas P, Pericuesta E, Bermejo-Álvarez P. Directions and applications of CRISPR technology in livestock research. Anim Reprod 2018; 15:292-300. [PMID: 34178152 PMCID: PMC8202460 DOI: 10.21451/1984-3143-ar2018-0075] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The ablation (KO) or targeted insertion (KI) of specific genes or sequences has been essential
to test their roles on a particular biological process. Unfortunately, such genome modifications
have been largely limited to the mouse model, as the only way to achieve targeted mutagenesis
in other mammals required from somatic cell nuclear transfer, a time- and resource-consuming
technique. This difficulty has left research in livestock species largely devoided of KO
and targeted KI models, crucial tools to uncover the molecular roots of any physiological
or pathological process. Luckily, the eruption of site-specific endonucleases, and particularly
CRISPR technology, has empowered farm animal scientists to consider projects that could
not develop before. In this sense, the availability of genome modification in livestock species
is meant to change the way research is performed on many fields, switching from descriptive
and correlational approaches to experimental research. In this review we will provide some
guidance about how the genome can be edited by CRISPR and the possible strategies to achieve
KO or KI, paying special attention to an initially overlooked phenomenon: mosaicism. Mosaicism
is produced when the zygote´s genome edition occurs after its DNA has replicated,
and is characterized by the presence of more than two alleles in the same individual, an undesirable
outcome when attempting direct KO generation. Finally, the possible applications on different
fields of livestock research, such as reproduction or infectious diseases are discussed.
Collapse
Affiliation(s)
| | | | - Eva Pericuesta
- Department Reproducción Animal, INIA, 28040 Madrid, Spain
| | | |
Collapse
|
45
|
Wei J, Wagner S, Maclean P, Brophy B, Cole S, Smolenski G, Carlson DF, Fahrenkrug SC, Wells DN, Laible G. Cattle with a precise, zygote-mediated deletion safely eliminate the major milk allergen beta-lactoglobulin. Sci Rep 2018; 8:7661. [PMID: 29769555 PMCID: PMC5955954 DOI: 10.1038/s41598-018-25654-8] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 04/19/2018] [Indexed: 12/26/2022] Open
Abstract
We applied precise zygote-mediated genome editing to eliminate beta-lactoglobulin (BLG), a major allergen in cows’ milk. To efficiently generate LGB knockout cows, biopsied embryos were screened to transfer only appropriately modified embryos. Transfer of 13 pre-selected embryos into surrogate cows resulted in the birth of three calves, one dying shortly after birth. Deep sequencing results confirmed conversion of the genotype from wild type to the edited nine bp deletion by more than 97% in the two male calves. The third calf, a healthy female, had in addition to the expected nine bp deletion (81%), alleles with an in frame 21 bp deletion (<17%) at the target site. While her milk was free of any mature BLG, we detected low levels of a BLG variant derived from the minor deletion allele. This confirmed that the nine bp deletion genotype completely knocks out production of BLG. In addition, we showed that the LGB knockout animals are free of any TALEN-mediated off-target mutations or vector integration events using an unbiased whole genome analysis. Our study demonstrates the feasibility of generating precisely biallelically edited cattle by zygote-mediated editing for the safe production of hypoallergenic milk.
Collapse
Affiliation(s)
- Jingwei Wei
- AgResearch, Ruakura Research Centre, Hamilton, 3240, New Zealand
| | - Stefan Wagner
- AgResearch, Ruakura Research Centre, Hamilton, 3240, New Zealand.,Rowett Institute, Aberdeen, AB25 2ZD, United Kingdom
| | - Paul Maclean
- AgResearch, Ruakura Research Centre, Hamilton, 3240, New Zealand
| | - Brigid Brophy
- AgResearch, Ruakura Research Centre, Hamilton, 3240, New Zealand
| | - Sally Cole
- AgResearch, Ruakura Research Centre, Hamilton, 3240, New Zealand
| | - Grant Smolenski
- AgResearch, Ruakura Research Centre, Hamilton, 3240, New Zealand.,MS3 Solutions Ltd., Ruakura Research Centre, Hamilton, 3240, New Zealand
| | | | | | - David N Wells
- AgResearch, Ruakura Research Centre, Hamilton, 3240, New Zealand
| | - Götz Laible
- AgResearch, Ruakura Research Centre, Hamilton, 3240, New Zealand.
| |
Collapse
|
46
|
Huang L, Hua Z, Xiao H, Cheng Y, Xu K, Gao Q, Xia Y, Liu Y, Zhang X, Zheng X, Mu Y, Li K. CRISPR/Cas9-mediated ApoE-/- and LDLR-/- double gene knockout in pigs elevates serum LDL-C and TC levels. Oncotarget 2018; 8:37751-37760. [PMID: 28465483 PMCID: PMC5514946 DOI: 10.18632/oncotarget.17154] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Accepted: 03/28/2017] [Indexed: 12/13/2022] Open
Abstract
The traditional method to establish a cardiovascular disease model induced by high fat and high cholesterol diets is time consuming and laborious and may not be appropriate in all circumstances. A suitable pig model to study metabolic disorders and subsequent atherosclerosis is not currently available. For this purpose, we applied the CRISPR/Cas9 system to Bama minipigs, targeting apolipoprotein E (ApoE) and low density lipoprotein receptor (LDLR) gene simultaneously. Six biallelic knockout pigs of these two genes were obtained successfully in a single step. No off-target incidents or mosaic mutations were detected by an unbiased analysis. Serum biochemical analyses of gene-modified piglets showed that the levels of low density lipoprotein choleserol (LDL-C), total cholesterol (TC) and apolipoprotein B (APOB) were elevated significantly. This model should prove valuable for the study of human cardiovascular disease and related translational research.
Collapse
Affiliation(s)
- Lei Huang
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China.,Animal Functional Genomics Group, Agricultural Genomes Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518120, China
| | - Zaidong Hua
- Hubei Key Laboratory of Animal Embryo Engineering and Molecular Breeding, Institute of Animal Science and Veterinary Medicine, Hubei Academy of Agricultural Science, Wuhan 430064, China
| | - Hongwei Xiao
- Hubei Key Laboratory of Animal Embryo Engineering and Molecular Breeding, Institute of Animal Science and Veterinary Medicine, Hubei Academy of Agricultural Science, Wuhan 430064, China
| | - Ying Cheng
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Kui Xu
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Qian Gao
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Ying Xia
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Yang Liu
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Xue Zhang
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Xinming Zheng
- Hubei Key Laboratory of Animal Embryo Engineering and Molecular Breeding, Institute of Animal Science and Veterinary Medicine, Hubei Academy of Agricultural Science, Wuhan 430064, China
| | - Yulian Mu
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Kui Li
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| |
Collapse
|
47
|
Abstract
Prokaryotic type II adaptive immune systems have been developed into the versatile CRISPR technology, which has been widely applied in site-specific genome editing and has revolutionized biomedical research due to its superior efficiency and flexibility. Recent studies have greatly diversified CRISPR technologies by coupling it with various DNA repair mechanisms and targeting strategies. These new advances have significantly expanded the generation of genetically modified animal models, either by including species in which targeted genetic modification could not be achieved previously, or through introducing complex genetic modifications that take multiple steps and cost years to achieve using traditional methods. Herein, we review the recent developments and applications of CRISPR-based technology in generating various animal models, and discuss the everlasting impact of this new progress on biomedical research.
Collapse
Affiliation(s)
- Xun Ma
- Key Laboratory for Regenerative Medicine in Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China.
| | - Avery Sum-Yu Wong
- Key Laboratory for Regenerative Medicine in Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China.
| | - Hei-Yin Tam
- Key Laboratory for Regenerative Medicine in Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China.
| | - Samuel Yung-Kin Tsui
- Key Laboratory for Regenerative Medicine in Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China.
| | - Dittman Lai-Shun Chung
- Key Laboratory for Regenerative Medicine in Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China.
| | - Bo Feng
- Key Laboratory for Regenerative Medicine in Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China. .,Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Guangdong 510530, China.,SBS Core Laboratory, CUHK Shenzhen Research Institute, Shenzhen Guangdong 518057, China
| |
Collapse
|
48
|
Nishio K, Tanihara F, Nguyen TV, Kunihara T, Nii M, Hirata M, Takemoto T, Otoi T. Effects of voltage strength during electroporation on the development and quality of in vitro-produced porcine embryos. Reprod Domest Anim 2018; 53:313-318. [PMID: 29135047 DOI: 10.1111/rda.13106] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Accepted: 10/10/2017] [Indexed: 12/20/2022]
Abstract
This study was conducted to determine suitable conditions for an experimental method in which the CRISPR/Cas9 system is introduced into in vitro-produced porcine zygotes by electroporation. In the first experiment, when putative zygotes derived from in vitro fertilization (IVF) were electroporated by either unipolar or bipolar pulses, keeping the voltage, pulse duration and pulse number fixed at 30 V/mm, 1 msec and five repeats, respectively, the rate of blastocyst formation from zygotes electroporated by bipolar pulses decreased compared to zygotes electroporated by unipolar pulses. In the second experiment, the putative zygotes were electroporated by electroporation voltages ranging from 20 V/mm-40 V/mm with five 1-msec unipolar pulses. The rate of cleavage and blastocyst formation of zygotes electroporated at 40 V/mm was significantly lower (p < .05) than that of zygotes electroporated at less than 30 V/mm. Moreover, the apoptotic nuclei indices of blastocysts derived from zygotes electroporated by voltages greater than 30 V/mm significantly increased compared with those from zygotes electroporated by voltages less than 25 V/mm (p < .05). When zygotes were electroporated with Cas9 mRNA and single-guide RNA (sgRNA) targeting site in the FGF10 exon 3, the proportions of blastocysts with targeted genomic sequences were 7.7% (2/26) and 3.6% (1/28) in the embryos derived from zygotes electroporated at 25 V/mm and 30 V/mm, respectively. Our results indicate that electroporation at 25 V/mm may be an acceptable condition for introducing Cas9 mRNA and sgRNA into pig IVF zygotes under which the viability of the embryos is not significantly affected.
Collapse
Affiliation(s)
- K Nishio
- Faculty of Bioscience and Bioindustry, Tokushima University, Tokushima, Japan
| | - F Tanihara
- Faculty of Bioscience and Bioindustry, Tokushima University, Tokushima, Japan
| | - T-V Nguyen
- Faculty of Bioscience and Bioindustry, Tokushima University, Tokushima, Japan
| | - T Kunihara
- Faculty of Bioscience and Bioindustry, Tokushima University, Tokushima, Japan
| | - M Nii
- Tokushima Prefectural Livestock Research Institute, Tokushima, Japan
| | - M Hirata
- Faculty of Bioscience and Bioindustry, Tokushima University, Tokushima, Japan
| | - T Takemoto
- Institute for Advanced Medical Sciences, Tokushima University, Tokushima, Japan
| | - T Otoi
- Faculty of Bioscience and Bioindustry, Tokushima University, Tokushima, Japan
| |
Collapse
|
49
|
Single step production of Cas9 mRNA for zygote injection. Biotechniques 2018; 64:118-124. [PMID: 29570443 DOI: 10.2144/btn-2017-0116] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 02/19/2018] [Indexed: 12/27/2022] Open
Abstract
Production of Cas9 mRNA in vitro typically requires the addition of a 5´ cap and 3´ polyadenylation. A plasmid was constructed that harbored the T7 promoter followed by the EMCV IRES and a Cas9 coding region. We hypothesized that the use of the metastasis associated lung adenocarcinoma transcript 1 (Malat1) triplex structure downstream of an IRES/Cas9 expression cassette would make polyadenylation of in vitro produced mRNA unnecessary. A sequence from the mMalat1 gene was cloned downstream of the IRES/Cas9 cassette described above. An mRNA concentration curve was constructed with either commercially available Cas9 mRNA or the IRES/ Cas9/triplex, by injection into porcine zygotes. Blastocysts were genotyped to determine if differences existed in the percent of embryos modified. The concentration curve identified differences due to concentration and RNA type injected. Single step production of Cas9 mRNA provides an alternative source of Cas9 for use in zygote injections.
Collapse
|
50
|
Abstract
The performance of the molecular tool using CRISPR-Cas9, which makes it possible to induce targeted modifications of the DNA, has found numerous applications in research and open promising prospects in human clinic. CRISPR-Cas9 has been widely used to generate transgenic animals after targeted modification of the genome at the zygotic stage. It was also tested on human embryos on an experimental basis. Although there are potential medical indications that may justify a targeted modification of the embryo or germ cell genome, the uncertainties regarding the efficacy and safety of the method do not allow us to consider implementing such germline gene therapy in the short-term. However, it is necessary to weigh the scientific and ethical issues involved in this approach.
Collapse
Affiliation(s)
- Pierre Jouannet
- Université Paris Descartes, 12 Rue de l'École de Médecine, 75006 Paris, France
| |
Collapse
|