1
|
Bertorello S, Cei F, Fink D, Niccolai E, Amedei A. The Future Exploring of Gut Microbiome-Immunity Interactions: From In Vivo/Vitro Models to In Silico Innovations. Microorganisms 2024; 12:1828. [PMID: 39338502 PMCID: PMC11434319 DOI: 10.3390/microorganisms12091828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 08/29/2024] [Accepted: 09/02/2024] [Indexed: 09/30/2024] Open
Abstract
Investigating the complex interactions between microbiota and immunity is crucial for a fruitful understanding progress of human health and disease. This review assesses animal models, next-generation in vitro models, and in silico approaches that are used to decipher the microbiome-immunity axis, evaluating their strengths and limitations. While animal models provide a comprehensive biological context, they also raise ethical and practical concerns. Conversely, modern in vitro models reduce animal involvement but require specific costs and materials. When considering the environmental impact of these models, in silico approaches emerge as promising for resource reduction, but they require robust experimental validation and ongoing refinement. Their potential is significant, paving the way for a more sustainable and ethical future in microbiome-immunity research.
Collapse
Affiliation(s)
- Sara Bertorello
- Department of Experimental and Clinical Medicine, University of Florence, 50139 Florence, Italy; (S.B.); (F.C.); (D.F.); (A.A.)
| | - Francesco Cei
- Department of Experimental and Clinical Medicine, University of Florence, 50139 Florence, Italy; (S.B.); (F.C.); (D.F.); (A.A.)
| | - Dorian Fink
- Department of Experimental and Clinical Medicine, University of Florence, 50139 Florence, Italy; (S.B.); (F.C.); (D.F.); (A.A.)
| | - Elena Niccolai
- Department of Experimental and Clinical Medicine, University of Florence, 50139 Florence, Italy; (S.B.); (F.C.); (D.F.); (A.A.)
- Laboratorio Congiunto MIA-LAB (Microbiome-Immunity Axis Research for a Circular Health), University of Florence, 50134 Florence, Italy
| | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Florence, 50139 Florence, Italy; (S.B.); (F.C.); (D.F.); (A.A.)
- Laboratorio Congiunto MIA-LAB (Microbiome-Immunity Axis Research for a Circular Health), University of Florence, 50134 Florence, Italy
| |
Collapse
|
2
|
Redel BK, Yoon J, Reese E, An H, Uh K, Chen PR, Prather RS, Lee K. Novel off-Targeting Events Identified after Genome Wide Analysis of CRISPR-Cas Edited Pigs. CRISPR J 2024; 7:141-149. [PMID: 38770737 PMCID: PMC11302191 DOI: 10.1089/crispr.2024.0012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 04/22/2024] [Indexed: 05/22/2024] Open
Abstract
CRISPR-Cas technology has transformed our ability to introduce targeted modifications, allowing unconventional animal models such as pigs to model human diseases and improve its value for food production. The main concern with using the technology is the possibility of introducing unwanted modifications in the genome. In this study, we illustrate a pipeline to comprehensively identify off-targeting events on a global scale in the genome of three different gene-edited pig models. Whole genome sequencing paired with an off-targeting prediction software tool filtered off-targeting events amongst natural variations present in gene-edited pigs. This pipeline confirmed two known off-targeting events in IGH knockout pigs, AR and RBFOX1, and identified other presumably off-targeted loci. Independent validation of the off-targeting events using other gene-edited DNA confirmed two novel off-targeting events in RAG2/IL2RG knockout pig models. This unique strategy offers a novel tool to detect off-targeting events in genetically heterogeneous species after genome editing.
Collapse
Affiliation(s)
| | - Junchul Yoon
- Division of Animal Sciences, University of Missouri, Columbia, Missouri, USA
| | - Emily Reese
- Division of Animal Sciences, University of Missouri, Columbia, Missouri, USA
| | - Hong An
- Bioinformatics and Analytics Core, University of Missouri, Columbia, Missouri, USA
| | - Kyungjun Uh
- Futuristic Animal Resource & Research Center (FARRC), Korea Research Institute of Bioscience and Biotechnology (KRIBB), Chungcheongbuk-do, Republic of Korea
| | - Paula R. Chen
- USDA-ARS, Plant Genetics Research Unit, Columbia, Missouri, USA
| | - Randall S. Prather
- Division of Animal Sciences, University of Missouri, Columbia, Missouri, USA
- National Swine Resource and Research Center, Columbia, Missouri, USA
| | - Kiho Lee
- Division of Animal Sciences, University of Missouri, Columbia, Missouri, USA
- National Swine Resource and Research Center, Columbia, Missouri, USA
| |
Collapse
|
3
|
Mao H, Li J, Gao M, Liu X, Zhang H, Zhuang Y, He T, Zuo W, Bai L, Bao J. Targeted Integration of siRNA against Porcine Cytomegalovirus (PCMV) Enhances the Resistance of Porcine Cells to PCMV. Microorganisms 2024; 12:837. [PMID: 38674781 PMCID: PMC11051760 DOI: 10.3390/microorganisms12040837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Revised: 04/15/2024] [Accepted: 04/17/2024] [Indexed: 04/28/2024] Open
Abstract
In the world's first pig-to-human cardiac cytomegalovirus (PCMV), xenotransplant and elevated levels of porcine key factors contributing to patient mortality were considered. This has renewed attention on PCMV, a virus widely prevalent in pigs. Currently, there are no effective drugs or vaccines targeting PCMV, and its high detection difficulty poses challenges for prevention and control research. In this study, antiviral small hairpin RNA (shRNA) was selected and inserted into the Rosa26 and miR-17-92 loci of pigs via a CRISPR/Cas9-mediated knock-in strategy. Further in vitro viral challenge experiments demonstrated that these genetically edited pig cells could effectively limit PCMV replication. Through this process, we constructed a PCMV-infected cell model, validated partial viral interference sites, enhanced gene knock-in efficiency, performed gene editing at two different gene loci, and ultimately demonstrated that RNA interference (RNAi) technology combined with CRISPR/Cas9 has the potential to generate pig cells with enhanced antiviral infection capabilities. This opens up possibilities for the future production of pig populations with antiviral functionalities.
Collapse
Affiliation(s)
- Hongzhen Mao
- Department of Pathology, Institute of Clinical Pathology, Key Laboratory of Transplant Engineering and Immunology, National Health Commission of China, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jinyang Li
- Department of Pathology, Institute of Clinical Pathology, Key Laboratory of Transplant Engineering and Immunology, National Health Commission of China, West China Hospital, Sichuan University, Chengdu 610041, China
- Department of Pathology, Regeneration Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Mengyu Gao
- Department of Pathology, Institute of Clinical Pathology, Key Laboratory of Transplant Engineering and Immunology, National Health Commission of China, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xinmei Liu
- Department of Pathology, Institute of Clinical Pathology, Key Laboratory of Transplant Engineering and Immunology, National Health Commission of China, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Haohan Zhang
- Department of Pathology, Institute of Clinical Pathology, Key Laboratory of Transplant Engineering and Immunology, National Health Commission of China, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yijia Zhuang
- Department of Pathology, Institute of Clinical Pathology, Key Laboratory of Transplant Engineering and Immunology, National Health Commission of China, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Tianyi He
- Department of Pathology, Institute of Clinical Pathology, Key Laboratory of Transplant Engineering and Immunology, National Health Commission of China, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Wei Zuo
- Key Laboratory of Transplant Engineering and Immunology, National Health Commission of China, West China Hospital, Sichuan University, Chengdu 610041, China
- Department of Organ Regeneration, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Lang Bai
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ji Bao
- Department of Pathology, Institute of Clinical Pathology, Key Laboratory of Transplant Engineering and Immunology, National Health Commission of China, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
4
|
Navarro-Serna S, Piñeiro-Silva C, Fernández-Martín I, Dehesa-Etxebeste M, López de Munain A, Gadea J. Oocyte electroporation prior to in vitro fertilization is an efficient method to generate single, double, and multiple knockout porcine embryos of interest in biomedicine and animal production. Theriogenology 2024; 218:111-118. [PMID: 38320372 DOI: 10.1016/j.theriogenology.2024.01.040] [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: 11/14/2023] [Revised: 01/31/2024] [Accepted: 01/31/2024] [Indexed: 02/08/2024]
Abstract
Genetically modified pigs play a critical role in mimicking human diseases, xenotransplantation, and the development of pigs resistant to viral diseases. The use of programmable endonucleases, including the CRISPR/Cas9 system, has revolutionized the generation of genetically modified pigs. This study evaluates the efficiency of electroporation of oocytes prior to fertilization in generating edited gene embryos for different models. For single gene editing, phospholipase C zeta (PLC ζ) and fused in sarcoma (FUS) genes were used, and the concentration of sgRNA and Cas9 complexes was optimized. The results showed that increasing the concentration resulted in higher mutation rates without affecting the blastocyst rate. Electroporation produced double knockouts for the TPC1/TPC2 genes with high efficiency (79 %). In addition, resistance to viral diseases such as PRRS and swine influenza was achieved by electroporation, allowing the generation of double knockout embryo pigs (63 %). The study also demonstrated the potential for multiple gene editing in a single step using electroporation, which is relevant for xenotransplantation. The technique resulted in the simultaneous mutation of 5 genes (GGTA1, B4GALNT2, pseudo B4GALNT2, CMAH and GHR). Overall, electroporation proved to be an efficient and versatile method to generate genetically modified embryonic pigs, offering significant advances in biomedical and agricultural research, xenotransplantation, and disease resistance. Electroporation led to the processing of numerous oocytes in a single session using less expensive equipment. We confirmed the generation of gene-edited porcine embryos for single, double, or quintuple genes simultaneously without altering embryo development to the blastocyst stage. The results provide valuable insights into the optimization of gene editing protocols for different models, opening new avenues for research and applications in this field.
Collapse
Affiliation(s)
- Sergio Navarro-Serna
- Department Physiology, University of Murcia, International Excellence Campus for Higher Education and Research "Campus Mare Nostrum" and Institute for Biomedical Research of Murcia (IMIB-Arrixaca), 30100, Murcia, Spain
| | - Celia Piñeiro-Silva
- Department Physiology, University of Murcia, International Excellence Campus for Higher Education and Research "Campus Mare Nostrum" and Institute for Biomedical Research of Murcia (IMIB-Arrixaca), 30100, Murcia, Spain
| | - Irene Fernández-Martín
- Department Physiology, University of Murcia, International Excellence Campus for Higher Education and Research "Campus Mare Nostrum" and Institute for Biomedical Research of Murcia (IMIB-Arrixaca), 30100, Murcia, Spain
| | | | - Adolfo López de Munain
- IIS Biodonostia, Neuroscience, San Sebastián, Spain; Department of Neurology. Hospital Universitario Donostia-OSAKIDETZA, San Sebastián, Spain; Department of Neurosciences. University of the Basque Country (UPV-EHU), San Sebastián, Spain; CIBERNED (CIBER), Institute Carlos III, Madrid, Spain
| | - Joaquín Gadea
- Department Physiology, University of Murcia, International Excellence Campus for Higher Education and Research "Campus Mare Nostrum" and Institute for Biomedical Research of Murcia (IMIB-Arrixaca), 30100, Murcia, Spain.
| |
Collapse
|
5
|
McCall MA. Pig Models in Retinal Research and Retinal Disease. Cold Spring Harb Perspect Med 2024; 14:a041296. [PMID: 37553210 PMCID: PMC10982707 DOI: 10.1101/cshperspect.a041296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/10/2023]
Abstract
The pig has been used as a large animal model in biomedical research for many years and its use continues to increase because induced mutations phenocopy several inherited human diseases. In addition, they are continuous breeders, can be propagated by artificial insemination, have large litter sizes (on the order of mice), and can be genetically manipulated using all of the techniques that are currently available in mice. The pioneering work of Petters and colleagues set the stage for the use of the pig as a model of inherited retinal disease. In the last 10 years, the pig has become a model of choice where specific disease-causing mutations that are not phenocopied in rodents need to be studied and therapeutic approaches explored. The pig is not only used for retinal eye disease but also for the study of the cornea and lens. This review attempts to show how broad the use of the pig has become and how it has contributed to the assessment of treatments for eye disease. In the last 10 years, there have been several reviews that included the use of the pig in biomedical research (see body of the review) that included information about retinal disease. None directly discuss the use of the pig as an animal model for retinal diseases, including inherited diseases, where a single genetic mutation has been identified or for multifactorial diseases such as glaucoma and diabetic retinopathy. Although the pig is used to explore diseases of the cornea and lens, this review focuses on how and why the pig, as a large animal model, is useful for research in neural retinal disease and its treatment.
Collapse
Affiliation(s)
- Maureen A McCall
- Departments of Ophthalmology & Visual Sciences and Anatomical Sciences & Neurobiology, University of Louisville, Louisville, Kentucky 40202, USA
| |
Collapse
|
6
|
Zheng Y, Li Y, Zhou K, Li T, VanDusen NJ, Hua Y. Precise genome-editing in human diseases: mechanisms, strategies and applications. Signal Transduct Target Ther 2024; 9:47. [PMID: 38409199 PMCID: PMC10897424 DOI: 10.1038/s41392-024-01750-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 01/15/2024] [Accepted: 01/17/2024] [Indexed: 02/28/2024] Open
Abstract
Precise genome-editing platforms are versatile tools for generating specific, site-directed DNA insertions, deletions, and substitutions. The continuous enhancement of these tools has led to a revolution in the life sciences, which promises to deliver novel therapies for genetic disease. Precise genome-editing can be traced back to the 1950s with the discovery of DNA's double-helix and, after 70 years of development, has evolved from crude in vitro applications to a wide range of sophisticated capabilities, including in vivo applications. Nonetheless, precise genome-editing faces constraints such as modest efficiency, delivery challenges, and off-target effects. In this review, we explore precise genome-editing, with a focus on introduction of the landmark events in its history, various platforms, delivery systems, and applications. First, we discuss the landmark events in the history of precise genome-editing. Second, we describe the current state of precise genome-editing strategies and explain how these techniques offer unprecedented precision and versatility for modifying the human genome. Third, we introduce the current delivery systems used to deploy precise genome-editing components through DNA, RNA, and RNPs. Finally, we summarize the current applications of precise genome-editing in labeling endogenous genes, screening genetic variants, molecular recording, generating disease models, and gene therapy, including ex vivo therapy and in vivo therapy, and discuss potential future advances.
Collapse
Affiliation(s)
- Yanjiang Zheng
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yifei Li
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Kaiyu Zhou
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Tiange Li
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Nathan J VanDusen
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
| | - Yimin Hua
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, 610041, China.
| |
Collapse
|
7
|
Patel A, Miles A, Strackhouse T, Cook L, Leng S, Patel S, Klinger K, Rudrabhatla S, Potlakayala SD. Methods of crop improvement and applications towards fortifying food security. Front Genome Ed 2023; 5:1171969. [PMID: 37484652 PMCID: PMC10361821 DOI: 10.3389/fgeed.2023.1171969] [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: 02/22/2023] [Accepted: 06/12/2023] [Indexed: 07/25/2023] Open
Abstract
Agriculture has supported human life from the beginning of civilization, despite a plethora of biotic (pests, pathogens) and abiotic (drought, cold) stressors being exerted on the global food demand. In the past 50 years, the enhanced understanding of cellular and molecular mechanisms in plants has led to novel innovations in biotechnology, resulting in the introduction of desired genes/traits through plant genetic engineering. Targeted genome editing technologies such as Zinc-Finger Nucleases (ZFNs), Transcription Activator-Like Effector Nucleases (TALENs), and Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR) have emerged as powerful tools for crop improvement. This new CRISPR technology is proving to be an efficient and straightforward process with low cost. It possesses applicability across most plant species, targets multiple genes, and is being used to engineer plant metabolic pathways to create resistance to pathogens and abiotic stressors. These novel genome editing (GE) technologies are poised to meet the UN's sustainable development goals of "zero hunger" and "good human health and wellbeing." These technologies could be more efficient in developing transgenic crops and aid in speeding up the regulatory approvals and risk assessments conducted by the US Departments of Agriculture (USDA), Food and Drug Administration (FDA), and Environmental Protection Agency (EPA).
Collapse
Affiliation(s)
- Aayushi Patel
- Penn State Harrisburg, Middletown, PA, United States
| | - Andrew Miles
- Penn State University Park, State College, University Park, PA, United States
| | | | - Logan Cook
- Penn State Harrisburg, Middletown, PA, United States
| | - Sining Leng
- Shanghai United Cell Biotechnology Co Ltd, Shanghai, China
| | - Shrina Patel
- Penn State Harrisburg, Middletown, PA, United States
| | | | | | | |
Collapse
|
8
|
Yang SP, Zhu XX, Qu ZX, Chen CY, Wu YB, Wu Y, Luo ZD, Wang XY, He CY, Fang JW, Wang LQ, Hong GL, Zheng ST, Zeng JM, Yan AF, Feng J, Liu L, Zhang XL, Zhang LG, Miao K, Tang DS. Production of MSTN knockout porcine cells using adenine base-editing-mediated exon skipping. In Vitro Cell Dev Biol Anim 2023:10.1007/s11626-023-00763-5. [PMID: 37099179 DOI: 10.1007/s11626-023-00763-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 03/24/2023] [Indexed: 04/27/2023]
Abstract
Gene-knockout pigs have important applications in agriculture and medicine. Compared with CRISPR/Cas9 and cytosine base editing (CBE) technologies, adenine base editing (ABE) shows better safety and accuracy in gene modification. However, because of the characteristics of gene sequences, the ABE system cannot be widely used in gene knockout. Alternative splicing of mRNA is an important biological mechanism in eukaryotes for the formation of proteins with different functional activities. The splicing apparatus recognizes conserved sequences of the 5' end splice donor and 3' end splice acceptor motifs of introns in pre-mRNA that can trigger exon skipping, leading to the production of new functional proteins, or causing gene inactivation through frameshift mutations. This study aimed to construct a MSTN knockout pig by inducing exon skipping with the aid of the ABE system to expand the application of the ABE system for the preparation of knockout pigs. In this study, first, we constructed ABEmaxAW and ABE8eV106W plasmid vectors and found that their editing efficiencies at the targets were at least sixfold and even 260-fold higher than that of ABEmaxAW by contrasting the editing efficiencies at the gene targets of endogenous CD163, IGF2, and MSTN in pigs. Subsequently, we used the ABE8eV106W system to realize adenine base (the base of the antisense strand is thymine) editing of the conserved splice donor sequence (5'-GT) of intron 2 of the porcine MSTN gene. A porcine single-cell clone carrying a homozygous mutation (5'-GC) in the conserved sequence (5'-GT) of the intron 2 splice donor of the MSTN gene was successfully generated after drug selection. Unfortunately, the MSTN gene was not expressed and, therefore, could not be characterized at this level. No detectable genomic off-target edits were identified by Sanger sequencing. In this study, we verified that the ABE8eV106W vector had higher editing efficiency and could expand the editing scope of ABE. Additionally, we successfully achieved the precise modification of the alternative splice acceptor of intron 2 of the porcine MSTN gene, which may provide a new strategy for gene knockout in pigs.
Collapse
Affiliation(s)
- Shuai-Peng Yang
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, School of Life Sciences and Engineering, Foshan University, Foshan, 528225, China
| | - Xiang-Xing Zhu
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, School of Life Sciences and Engineering, Foshan University, Foshan, 528225, China.
- Gene Editing Technology Center of Guangdong Province, School of Medicine, Foshan University, Foshan, 528225, China.
| | - Zi-Xiao Qu
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, School of Life Sciences and Engineering, Foshan University, Foshan, 528225, China
| | - Cai-Yue Chen
- Gene Editing Technology Center of Guangdong Province, School of Medicine, Foshan University, Foshan, 528225, China
| | - Yao-Bing Wu
- Gene Editing Technology Center of Guangdong Province, School of Medicine, Foshan University, Foshan, 528225, China
| | - Yue Wu
- Gene Editing Technology Center of Guangdong Province, School of Medicine, Foshan University, Foshan, 528225, China
| | - Zi-Dan Luo
- Gene Editing Technology Center of Guangdong Province, School of Medicine, Foshan University, Foshan, 528225, China
| | - Xin-Yi Wang
- Gene Editing Technology Center of Guangdong Province, School of Medicine, Foshan University, Foshan, 528225, China
| | - Chu-Yu He
- Gene Editing Technology Center of Guangdong Province, School of Medicine, Foshan University, Foshan, 528225, China
| | - Jia-Wen Fang
- Gene Editing Technology Center of Guangdong Province, School of Medicine, Foshan University, Foshan, 528225, China
| | - Ling-Qi Wang
- Gene Editing Technology Center of Guangdong Province, School of Medicine, Foshan University, Foshan, 528225, China
| | - Guang-Long Hong
- Gene Editing Technology Center of Guangdong Province, School of Medicine, Foshan University, Foshan, 528225, China
| | - Shu-Tao Zheng
- Gene Editing Technology Center of Guangdong Province, School of Medicine, Foshan University, Foshan, 528225, China
| | - Jie-Mei Zeng
- Gene Editing Technology Center of Guangdong Province, School of Medicine, Foshan University, Foshan, 528225, China
| | - Ai-Fen Yan
- Gene Editing Technology Center of Guangdong Province, School of Medicine, Foshan University, Foshan, 528225, China
| | - Juan Feng
- Gene Editing Technology Center of Guangdong Province, School of Medicine, Foshan University, Foshan, 528225, China
| | - Lian Liu
- Gene Editing Technology Center of Guangdong Province, School of Medicine, Foshan University, Foshan, 528225, China
| | - Xiao-Li Zhang
- Gene Editing Technology Center of Guangdong Province, School of Medicine, Foshan University, Foshan, 528225, China
| | - Li-Gang Zhang
- Gene Editing Technology Center of Guangdong Province, School of Medicine, Foshan University, Foshan, 528225, China
| | - Kai Miao
- Centre for Precision Medicine Research and Training, Faculty of Health Sciences, University of Macau, Macau SAR, China.
| | - Dong-Sheng Tang
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, School of Life Sciences and Engineering, Foshan University, Foshan, 528225, China.
- Gene Editing Technology Center of Guangdong Province, School of Medicine, Foshan University, Foshan, 528225, China.
| |
Collapse
|
9
|
Zakrzewska M, Burmistrz M. Mechanisms regulating the CRISPR-Cas systems. Front Microbiol 2023; 14:1060337. [PMID: 36925473 PMCID: PMC10013973 DOI: 10.3389/fmicb.2023.1060337] [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: 10/07/2022] [Accepted: 02/10/2023] [Indexed: 03/08/2023] Open
Abstract
The CRISPR-Cas (Clustered Regularly Interspaced Short Palindromic Repeats- CRISPR associated proteins) is a prokaryotic system that enables sequence specific recognition and cleavage of nucleic acids. This is possible due to cooperation between CRISPR array which contains short fragments of DNA called spacers that are complimentary to the targeted nucleic acid and Cas proteins, which take part in processes of: acquisition of new spacers, processing them into their functional form as well as recognition and cleavage of targeted nucleic acids. The primary role of CRISPR-Cas systems is to provide their host with an adaptive and hereditary immunity against exogenous nucleic acids. This system is present in many variants in both Bacteria and Archea. Due to its modular structure, and programmability CRISPR-Cas system become attractive tool for modern molecular biology. Since their discovery and implementation, the CRISPR-Cas systems revolutionized areas of gene editing and regulation of gene expression. Although our knowledge on how CRISPR-Cas systems work has increased rapidly in recent years, there is still little information on how these systems are controlled and how they interact with other cellular mechanisms. Such regulation can be the result of both auto-regulatory mechanisms as well as exogenous proteins of phage origin. Better understanding of these interaction networks would be beneficial for optimization of current and development of new CRISPR-Cas-based tools. In this review we summarize current knowledge on the various molecular mechanisms that affect activity of CRISPR-Cas systems.
Collapse
Affiliation(s)
- Marta Zakrzewska
- Department of Environmental Microbiology and Biotechnology, Faculty of Biology, Institute of Microbiology, University of Warsaw, Warsaw, Poland.,Department of Molecular Microbiology, Biological and Chemical Research Centre, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Michal Burmistrz
- Department of Molecular Microbiology, Biological and Chemical Research Centre, Faculty of Biology, University of Warsaw, Warsaw, Poland.,Centre of New Technologies, University of Warsaw, Warsaw, Poland
| |
Collapse
|
10
|
Preisinger D, Winogrodzki T, Klinger B, Schnieke A, Rieblinger B. Genome Editing in Pigs. Methods Mol Biol 2023; 2631:393-417. [PMID: 36995680 DOI: 10.1007/978-1-0716-2990-1_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/31/2023]
Abstract
The generation of genetically engineered (GE) pigs for disease modeling and xenotransplantation has been massively facilitated by the discovery of the CRISPR/Cas9 system. For livestock, genome editing is a powerful tool when used in combination with either somatic cell nuclear transfer (SCNT) or microinjection (MI) into fertilized oocytes. To generate either knockout or knock-in animals using SCNT, genome editing is carried out in vitro. This has the advantage that fully characterized cells are being employed to generate cloned pigs, predetermining their genetic makeups. However, this technique is labor-intensive and, hence, SCNT is better suited for more challenging projects such as the generation of multi-knockout- and knock-in pigs. Alternatively, CRISPR/Cas9 is introduced directly into fertilized zygotes via microinjection to produce knockout pigs more rapidly. Finally, the embryos are each transferred into recipient sows to deliver GE piglets.Both techniques, SCNT and MI, are technically challenging and therefore require skilled expertise, especially when applied for porcine embryos. Here, we present a detailed laboratory protocol for the generation of knockout and knock-in porcine somatic donor cells for SCNT and knockout pigs via microinjection. We describe the state-of-the-art method for isolation, cultivation, and manipulation of porcine somatic cells, which can then be used for SCNT. Moreover, we describe the isolation and maturation of porcine oocytes, their manipulation by microinjection, and the embryo transfer into surrogate sows.
Collapse
Affiliation(s)
- David Preisinger
- Department of Animal Sciences, School of Life Sciences Weihenstephan, Technical University Munich, Freising, Germany
| | - Thomas Winogrodzki
- Department of Animal Sciences, School of Life Sciences Weihenstephan, Technical University Munich, Freising, Germany
| | - Bernhard Klinger
- Department of Animal Sciences, School of Life Sciences Weihenstephan, Technical University Munich, Freising, Germany
| | - Angelika Schnieke
- Department of Animal Sciences, School of Life Sciences Weihenstephan, Technical University Munich, Freising, Germany
| | - Beate Rieblinger
- Department of Animal Sciences, School of Life Sciences Weihenstephan, Technical University Munich, Freising, Germany.
| |
Collapse
|
11
|
Moya-Jódar M, Coppiello G, Rodríguez-Madoz JR, Abizanda G, Barlabé P, Vilas-Zornoza A, Ullate-Agote A, Luongo C, Rodríguez-Tobón E, Navarro-Serna S, París-Oller E, Oficialdegui M, Carvajal-Vergara X, Ordovás L, Prósper F, García-Vázquez FA, Aranguren XL. One-Step In Vitro Generation of ETV2-Null Pig Embryos. Animals (Basel) 2022; 12:ani12141829. [PMID: 35883376 PMCID: PMC9311767 DOI: 10.3390/ani12141829] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 07/05/2022] [Accepted: 07/14/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary One of the latest goals in regenerative medicine is to use pluripotent stem cells to generate whole organs in vivo through the blastocyst complementation technique. This method consists of the microinjection of pluripotent stem cells into preimplantation embryos that have been genetically modified to ablate the development of a target organ. By taking advantage of the spatiotemporal clues present in the developing embryo, pluripotent stem cells are able to colonize the empty developmental niche and create the missing organ. Combining human pluripotent stem cells with genetically engineered pig embryos, it would be possible to obtain humanized organs that could be used for transplantation, and, therefore, solve the worldwide issue of insufficient availability of transplantable organs. As endothelial cells play a critical role in xenotransplantation rejection in all organs, in this study, we optimized a protocol to generate a vascular-disabled preimplantation pig embryo using the CRISPR/Cas9 system. This protocol could be used to generate avascular embryos for blastocyst complementation experiments and work towards the generation of rejection-free humanized organs in pigs. Abstract Each year, tens of thousands of people worldwide die of end-stage organ failure due to the limited availability of organs for use in transplantation. To meet this clinical demand, one of the last frontiers of regenerative medicine is the generation of humanized organs in pigs from pluripotent stem cells (PSCs) via blastocyst complementation. For this, organ-disabled pig models are needed. As endothelial cells (ECs) play a critical role in xenotransplantation rejection in every organ, we aimed to produce hematoendothelial-disabled pig embryos targeting the master transcription factor ETV2 via CRISPR-Cas9-mediated genome modification. In this study, we designed five different guide RNAs (gRNAs) against the DNA-binding domain of the porcine ETV2 gene, which were tested on porcine fibroblasts in vitro. Four out of five guides showed cleavage capacity and, subsequently, these four guides were microinjected individually as ribonucleoprotein complexes (RNPs) into one-cell-stage porcine embryos. Next, we combined the two gRNAs that showed the highest targeting efficiency and microinjected them at higher concentrations. Under these conditions, we significantly improved the rate of biallelic mutation. Hence, here, we describe an efficient one-step method for the generation of hematoendothelial-disabled pig embryos via CRISPR-Cas9 microinjection in zygotes. This model could be used in experimentation related to the in vivo generation of humanized organs.
Collapse
Affiliation(s)
- Marta Moya-Jódar
- Program of Regenerative Medicine, Centre for Applied Medical Research (CIMA), Instituto de Investigación Sanitaria de Navarra (IdiSNA), University of Navarra, 31008 Pamplona, Spain; (M.M.-J.); (G.C.); (J.R.R.-M.); (G.A.); (P.B.); (A.U.-A.); (X.C.-V.); (F.P.)
| | - Giulia Coppiello
- Program of Regenerative Medicine, Centre for Applied Medical Research (CIMA), Instituto de Investigación Sanitaria de Navarra (IdiSNA), University of Navarra, 31008 Pamplona, Spain; (M.M.-J.); (G.C.); (J.R.R.-M.); (G.A.); (P.B.); (A.U.-A.); (X.C.-V.); (F.P.)
| | - Juan Roberto Rodríguez-Madoz
- Program of Regenerative Medicine, Centre for Applied Medical Research (CIMA), Instituto de Investigación Sanitaria de Navarra (IdiSNA), University of Navarra, 31008 Pamplona, Spain; (M.M.-J.); (G.C.); (J.R.R.-M.); (G.A.); (P.B.); (A.U.-A.); (X.C.-V.); (F.P.)
| | - Gloria Abizanda
- Program of Regenerative Medicine, Centre for Applied Medical Research (CIMA), Instituto de Investigación Sanitaria de Navarra (IdiSNA), University of Navarra, 31008 Pamplona, Spain; (M.M.-J.); (G.C.); (J.R.R.-M.); (G.A.); (P.B.); (A.U.-A.); (X.C.-V.); (F.P.)
| | - Paula Barlabé
- Program of Regenerative Medicine, Centre for Applied Medical Research (CIMA), Instituto de Investigación Sanitaria de Navarra (IdiSNA), University of Navarra, 31008 Pamplona, Spain; (M.M.-J.); (G.C.); (J.R.R.-M.); (G.A.); (P.B.); (A.U.-A.); (X.C.-V.); (F.P.)
| | - Amaia Vilas-Zornoza
- Advanced Genomics Laboratory, Program of Hemato-Oncology, Center for Applied Medical Research (CIMA), University of Navarra, 31008 Pamplona, Spain;
| | - Asier Ullate-Agote
- Program of Regenerative Medicine, Centre for Applied Medical Research (CIMA), Instituto de Investigación Sanitaria de Navarra (IdiSNA), University of Navarra, 31008 Pamplona, Spain; (M.M.-J.); (G.C.); (J.R.R.-M.); (G.A.); (P.B.); (A.U.-A.); (X.C.-V.); (F.P.)
- Advanced Genomics Laboratory, Program of Hemato-Oncology, Center for Applied Medical Research (CIMA), University of Navarra, 31008 Pamplona, Spain;
| | - Chiara Luongo
- Department of Physiology, Veterinary School, International Excellence Campus for Higher Education and Research (Campus Mare Nostrum), University of Murcia, 30100 Murcia, Spain; (C.L.); (E.R.-T.); (S.N.-S.); (E.P.-O.)
- Institute for Biomedical Research of Murcia, IMIB-Arrixaca, 30100 Murcia, Spain
| | - Ernesto Rodríguez-Tobón
- Department of Physiology, Veterinary School, International Excellence Campus for Higher Education and Research (Campus Mare Nostrum), University of Murcia, 30100 Murcia, Spain; (C.L.); (E.R.-T.); (S.N.-S.); (E.P.-O.)
- Institute for Biomedical Research of Murcia, IMIB-Arrixaca, 30100 Murcia, Spain
| | - Sergio Navarro-Serna
- Department of Physiology, Veterinary School, International Excellence Campus for Higher Education and Research (Campus Mare Nostrum), University of Murcia, 30100 Murcia, Spain; (C.L.); (E.R.-T.); (S.N.-S.); (E.P.-O.)
- Institute for Biomedical Research of Murcia, IMIB-Arrixaca, 30100 Murcia, Spain
| | - Evelyne París-Oller
- Department of Physiology, Veterinary School, International Excellence Campus for Higher Education and Research (Campus Mare Nostrum), University of Murcia, 30100 Murcia, Spain; (C.L.); (E.R.-T.); (S.N.-S.); (E.P.-O.)
- Institute for Biomedical Research of Murcia, IMIB-Arrixaca, 30100 Murcia, Spain
| | | | - Xonia Carvajal-Vergara
- Program of Regenerative Medicine, Centre for Applied Medical Research (CIMA), Instituto de Investigación Sanitaria de Navarra (IdiSNA), University of Navarra, 31008 Pamplona, Spain; (M.M.-J.); (G.C.); (J.R.R.-M.); (G.A.); (P.B.); (A.U.-A.); (X.C.-V.); (F.P.)
| | - Laura Ordovás
- Aragon Agency for Research and Development (ARAID), 50018 Zaragoza, Spain;
- Biomedical Signal Interpretation and Computational Simulation (BSICoS), Institute of Engineering Research (I3A), University of Zaragoza & Instituto de Investigación Sanitaria (IIS), 50018 Zaragoza, Spain
| | - Felipe Prósper
- Program of Regenerative Medicine, Centre for Applied Medical Research (CIMA), Instituto de Investigación Sanitaria de Navarra (IdiSNA), University of Navarra, 31008 Pamplona, Spain; (M.M.-J.); (G.C.); (J.R.R.-M.); (G.A.); (P.B.); (A.U.-A.); (X.C.-V.); (F.P.)
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain
- Department of Hematology and Cell Therapy, Clínica Universidad de Navarra, 31008 Pamplona, Spain
| | - Francisco Alberto García-Vázquez
- Department of Physiology, Veterinary School, International Excellence Campus for Higher Education and Research (Campus Mare Nostrum), University of Murcia, 30100 Murcia, Spain; (C.L.); (E.R.-T.); (S.N.-S.); (E.P.-O.)
- Institute for Biomedical Research of Murcia, IMIB-Arrixaca, 30100 Murcia, Spain
- Correspondence: (F.A.G.-V.); (X.L.A.)
| | - Xabier L. Aranguren
- Program of Regenerative Medicine, Centre for Applied Medical Research (CIMA), Instituto de Investigación Sanitaria de Navarra (IdiSNA), University of Navarra, 31008 Pamplona, Spain; (M.M.-J.); (G.C.); (J.R.R.-M.); (G.A.); (P.B.); (A.U.-A.); (X.C.-V.); (F.P.)
- Correspondence: (F.A.G.-V.); (X.L.A.)
| |
Collapse
|
12
|
Whitworth KM, Green JA, Redel BK, Geisert RD, Lee K, Telugu BP, Wells KD, Prather RS. Improvements in pig agriculture through gene editing. CABI AGRICULTURE AND BIOSCIENCE 2022; 3:41. [PMID: 35755158 PMCID: PMC9209828 DOI: 10.1186/s43170-022-00111-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 06/12/2022] [Indexed: 05/06/2023]
Abstract
Genetic modification of animals via selective breeding is the basis for modern agriculture. The current breeding paradigm however has limitations, chief among them is the requirement for the beneficial trait to exist within the population. Desirable alleles in geographically isolated breeds, or breeds selected for a different conformation and commercial application, and more importantly animals from different genera or species cannot be introgressed into the population via selective breeding. Additionally, linkage disequilibrium results in low heritability and necessitates breeding over successive generations to fix a beneficial trait within a population. Given the need to sustainably improve animal production to feed an anticipated 9 billion global population by 2030 against a backdrop of infectious diseases and a looming threat from climate change, there is a pressing need for responsive, precise, and agile breeding strategies. The availability of genome editing tools that allow for the introduction of precise genetic modification at a single nucleotide resolution, while also facilitating large transgene integration in the target population, offers a solution. Concordant with the developments in genomic sequencing approaches, progress among germline editing efforts is expected to reach feverish pace. The current manuscript reviews past and current developments in germline engineering in pigs, and the many advantages they confer for advancing animal agriculture.
Collapse
Affiliation(s)
- Kristin M. Whitworth
- Division of Animal Science, College of Agriculture Food and Natural Resources, University of Missouri, 920 East Campus Drive, Columbia, MO 65211 USA
| | - Jonathan A. Green
- Division of Animal Science, College of Agriculture Food and Natural Resources, University of Missouri, 920 East Campus Drive, Columbia, MO 65211 USA
| | - Bethany K. Redel
- United States Department of Agriculture – Agriculture Research Service, Plant Genetics Research Unit, Columbia, MO 65211 USA
| | - Rodney D. Geisert
- Division of Animal Science, College of Agriculture Food and Natural Resources, University of Missouri, 920 East Campus Drive, Columbia, MO 65211 USA
| | - Kiho Lee
- Division of Animal Science, College of Agriculture Food and Natural Resources, University of Missouri, 920 East Campus Drive, Columbia, MO 65211 USA
| | - Bhanu P. Telugu
- Division of Animal Science, College of Agriculture Food and Natural Resources, University of Missouri, 920 East Campus Drive, Columbia, MO 65211 USA
| | - Kevin D. Wells
- Division of Animal Science, College of Agriculture Food and Natural Resources, University of Missouri, 920 East Campus Drive, Columbia, MO 65211 USA
| | - Randall S. Prather
- Division of Animal Science, College of Agriculture Food and Natural Resources, University of Missouri, 920 East Campus Drive, Columbia, MO 65211 USA
| |
Collapse
|
13
|
New pathogenic insights from large animal models of neurodegenerative diseases. Protein Cell 2022; 13:707-720. [PMID: 35334073 PMCID: PMC9233730 DOI: 10.1007/s13238-022-00912-8] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 02/23/2022] [Indexed: 12/12/2022] Open
Abstract
Animal models are essential for investigating the pathogenesis and developing the treatment of human diseases. Identification of genetic mutations responsible for neurodegenerative diseases has enabled the creation of a large number of small animal models that mimic genetic defects found in the affected individuals. Of the current animal models, rodents with genetic modifications are the most commonly used animal models and provided important insights into pathogenesis. However, most of genetically modified rodent models lack overt neurodegeneration, imposing challenges and obstacles in utilizing them to rigorously test the therapeutic effects on neurodegeneration. Recent studies that used CRISPR/Cas9-targeted large animal (pigs and monkeys) have uncovered important pathological events that resemble neurodegeneration in the patient’s brain but could not be produced in small animal models. Here we highlight the unique nature of large animals to model neurodegenerative diseases as well as the limitations and challenges in establishing large animal models of neurodegenerative diseases, with focus on Huntington disease, Amyotrophic lateral sclerosis, and Parkinson diseases. We also discuss how to use the important pathogenic insights from large animal models to make rodent models more capable of recapitulating important pathological features of neurodegenerative diseases.
Collapse
|
14
|
Gutierrez K, Glanzner WG, de Macedo MP, Rissi VB, Dicks N, Bohrer RC, Baldassarre H, Agellon LB, Bordignon V. Cell Cycle Stage and DNA Repair Pathway Influence CRISPR/Cas9 Gene Editing Efficiency in Porcine Embryos. Life (Basel) 2022; 12:life12020171. [PMID: 35207459 PMCID: PMC8876063 DOI: 10.3390/life12020171] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 01/18/2022] [Accepted: 01/21/2022] [Indexed: 11/21/2022] Open
Abstract
CRISPR/Cas9 technology is a powerful tool used for genome manipulation in different cell types and species. However, as with all new technologies, it still requires improvements. Different factors can affect CRISPR/Cas efficiency in zygotes, which influence the total cost and complexity for creating large-animal models for research. This study evaluated the importance of zygote cell cycle stage between early-injection (within 6 h post activation/fertilization) versus late-injection (14–16 h post activation/fertilization) when the CRISPR/Cas9 components were injected and the inhibition of the homologous recombination (HR) pathway of DNA repair on gene editing, embryo survival and development on embryos produced by fertilization, sperm injection, somatic cell nuclear transfer, and parthenogenetic activation technologies. Injections at the late cell cycle stage decreased embryo survival (measured as the proportion of unlysed embryos) and blastocyst formation (68.2%; 19.3%) compared to early-stage injection (86.3%; 28.8%). However, gene editing was higher in blastocysts from late-(73.8%) vs. early-(63.8%) injected zygotes. Inhibition of the HR repair pathway increased gene editing efficiency by 15.6% in blastocysts from early-injected zygotes without compromising embryo development. Our finding shows that injection at the early cell cycle stage along with HR inhibition improves both zygote viability and gene editing rate in pig blastocysts.
Collapse
Affiliation(s)
- Karina Gutierrez
- Department of Animal Science, McGill University, Sainte-Anne-de-Bellevue, QC H9X 3V9, Canada; (K.G.); (W.G.G.); (M.P.d.M.); (N.D.); (R.C.B.); (H.B.)
| | - Werner G. Glanzner
- Department of Animal Science, McGill University, Sainte-Anne-de-Bellevue, QC H9X 3V9, Canada; (K.G.); (W.G.G.); (M.P.d.M.); (N.D.); (R.C.B.); (H.B.)
| | - Mariana P. de Macedo
- Department of Animal Science, McGill University, Sainte-Anne-de-Bellevue, QC H9X 3V9, Canada; (K.G.); (W.G.G.); (M.P.d.M.); (N.D.); (R.C.B.); (H.B.)
| | - Vitor B. Rissi
- Department of Agriculture, Biodiversity and Forests, Federal University of Santa Catarina, Curitibanos 89520-000, Brazil;
| | - Naomi Dicks
- Department of Animal Science, McGill University, Sainte-Anne-de-Bellevue, QC H9X 3V9, Canada; (K.G.); (W.G.G.); (M.P.d.M.); (N.D.); (R.C.B.); (H.B.)
| | - Rodrigo C. Bohrer
- Department of Animal Science, McGill University, Sainte-Anne-de-Bellevue, QC H9X 3V9, Canada; (K.G.); (W.G.G.); (M.P.d.M.); (N.D.); (R.C.B.); (H.B.)
| | - Hernan Baldassarre
- Department of Animal Science, McGill University, Sainte-Anne-de-Bellevue, QC H9X 3V9, Canada; (K.G.); (W.G.G.); (M.P.d.M.); (N.D.); (R.C.B.); (H.B.)
| | - Luis B. Agellon
- School of Human Nutrition, McGill University, Sainte-Anne-de-Bellevue, QC H9X 3V9, Canada
- Correspondence: (L.B.A.); (V.B.)
| | - Vilceu Bordignon
- Department of Animal Science, McGill University, Sainte-Anne-de-Bellevue, QC H9X 3V9, Canada; (K.G.); (W.G.G.); (M.P.d.M.); (N.D.); (R.C.B.); (H.B.)
- Correspondence: (L.B.A.); (V.B.)
| |
Collapse
|
15
|
Adenine base-editing-mediated exon skipping induces gene knockout in cultured pig cells. Biotechnol Lett 2022; 44:59-76. [PMID: 34997407 DOI: 10.1007/s10529-021-03214-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 11/28/2021] [Indexed: 12/26/2022]
Abstract
Gene-knockout pigs have important applications in agriculture and medicine. Compared with CRISPR/Cas9, Adenine base editor (ABE) convert single A·T pairs to G·C pairs in the genome without generating DNA double-strand breaks, and this method has higher accuracy and biosafety in pig genetic modification. However, the application of ABE in pig gene knockout is limited by protospacer-adjacent motif sequences and the base-editing window. Alternative mRNA splicing is an important mechanism underlying the formation of proteins with diverse functions in eukaryotes. Spliceosome recognizes the conservative sequences of splice donors and acceptors in a precursor mRNA. Mutations in these conservative sequences induce exon skipping, leading to proteins with novel functions or to gene inactivation due to frameshift mutations. In this study, adenine base-editing-mediated exon skipping was used to expand the application of ABE in the generation of gene knockout pigs. We first constructed a modified "all-in-one" ABE vector suitable for porcine somatic cell transfection that contained an ABE for single-base editing and an sgRNA expression cassette. The "all-in-one" ABE vector induced efficient sgRNA-dependent A-to-G conversions in porcine cells during single base-editing of multiple endogenous gene loci. Subsequently, an ABE system was designed for single adenine editing of the conservative splice acceptor site (AG sequence at the 3' end of the intron 5) and splice donor site (GT sequence at the 5' end of the intron 6) in the porcine gene GHR; this method achieved highly efficient A-to-G conversion at the cellular level. Then, porcine single-cell colonies carrying a biallelic A-to-G conversion in the splice acceptor site in the intron 5 of GHR were generated. RT-PCR indicated exon 6 skipped at the mRNA level. Western blotting revealed GHR protein loss, and gene sequencing showed no sgRNA-dependent off-target effects. These results demonstrate accurate adenine base-editing-mediated exon skipping and gene knockout in porcine cells. This is the first proof-of-concept study of adenine base-editing-mediated exon skipping for gene regulation in pigs, and this work provides a new strategy for accurate and safe genetic modification of pigs for agricultural and medical applications.
Collapse
|
16
|
Tu CF, Chuang CK, Yang TS. The application of new breeding technology based on gene editing in pig industry. Anim Biosci 2022; 35:791-803. [PMID: 34991204 PMCID: PMC9066036 DOI: 10.5713/ab.21.0390] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 12/07/2021] [Indexed: 12/02/2022] Open
Abstract
Genome/gene-editing (GE) techniques, characterized by a low technological barrier, high efficiency, and broad application among organisms, are now being employed not only in medical science but also in agriculture/veterinary science. Different engineered CRISPR/Cas9s have been identified to expand the application of this technology. In pig production, GE is a precise new breeding technology (NBT), and promising outcomes in improving economic traits, such as growth, lean or healthy meat production, animal welfare, and disease resistance, have already been documented and reviewed. These promising achievements in porcine gene editing, including the Myostatin gene knockout (KO) in indigenous breeds to improve lean meat production, the uncoupling protein 1 (UCP1) gene knock-in to enhance piglet thermogenesis and survival under cold stress, the generation of GGTA1 and CMP-N-glycolylneuraminic acid hydroxylase (CMAH) gene double KO (dKO) pigs to produce healthy red meat, and the KO or deletion of exon 7 of the CD163 gene to confer resistance to porcine reproductive and respiratory syndrome virus infection, are described in the present article. Other related approaches for such purposes are also discussed. The current trend of global regulations or legislation for GE organisms is that they are exempted from classification as genetically modified organisms (GMOs) if no exogenes are integrated into the genome, according to product-based and not process-based methods. Moreover, an updated case study in the EU showed that current GMO legislation is not fit for purpose in term of NBTs, which contribute to the objectives of the EU’s Green Deal and biodiversity strategies and even meet the United Nations’ sustainable development goals for a more resilient and sustainable agri-food system. The GE pigs generated via NBT will be exempted from classification as GMOs, and their global valorization and commercialization can be foreseen.
Collapse
Affiliation(s)
- Ching-Fu Tu
- Division of Animal Technology, Animal Technology Laboratories, Agricultural Technology Research Institute, Hsinchu City 30093, Taiwan
| | - Chin-Kai Chuang
- Division of Animal Technology, Animal Technology Laboratories, Agricultural Technology Research Institute, Hsinchu City 30093, Taiwan
| | - Tien-Shuh Yang
- Division of Animal Technology, Animal Technology Laboratories, Agricultural Technology Research Institute, Hsinchu City 30093, Taiwan.,Department of Biotechnology and Animal Science, National Ilan University, Yilan City, 26047 Taiwan
| |
Collapse
|
17
|
Yang W, Chen X, Li S, Li XJ. Genetically modified large animal models for investigating neurodegenerative diseases. Cell Biosci 2021; 11:218. [PMID: 34933675 PMCID: PMC8690884 DOI: 10.1186/s13578-021-00729-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 12/07/2021] [Indexed: 12/02/2022] Open
Abstract
Neurodegenerative diseases represent a large group of neurological disorders including Alzheimer’s disease, amyotrophic lateral sclerosis, Parkinson’s disease, and Huntington’s disease. Although this group of diseases show heterogeneous clinical and pathological phenotypes, they share important pathological features characterized by the age-dependent and progressive degeneration of nerve cells that is caused by the accumulation of misfolded proteins. The association of genetic mutations with neurodegeneration diseases has enabled the establishment of various types of animal models that mimic genetic defects and have provided important insights into the pathogenesis. However, most of genetically modified rodent models lack the overt and selective neurodegeneration seen in the patient brains, making it difficult to use the small animal models to validate the effective treatment on neurodegeneration. Recent studies of pig and monkey models suggest that large animals can more faithfully recapitulate pathological features of neurodegenerative diseases. In this review, we discuss the important differences in animal models for modeling pathological features of neurodegenerative diseases, aiming to assist the use of animal models to better understand the pathogenesis and to develop effective therapeutic strategies.
Collapse
Affiliation(s)
- Weili Yang
- Guangdong Key Laboratory of Non-Human Primate Research, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China.
| | - Xiusheng Chen
- Guangdong Key Laboratory of Non-Human Primate Research, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Shihua Li
- Guangdong Key Laboratory of Non-Human Primate Research, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Xiao-Jiang Li
- Guangdong Key Laboratory of Non-Human Primate Research, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China.
| |
Collapse
|
18
|
Fang X, Tanga BM, Bang S, Seong G, Saadeldin IM, Lee S, Cho J. Oviduct epithelial cells-derived extracellular vesicles improve preimplantation developmental competence of in vitro produced porcine parthenogenetic and cloned embryos. Mol Reprod Dev 2021; 89:54-65. [PMID: 34843136 DOI: 10.1002/mrd.23550] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 11/22/2021] [Accepted: 11/23/2021] [Indexed: 12/15/2022]
Abstract
Extracellular vesicles (EVs) carry bioactive cargoes involved in the early preimplantation development. This study investigated the effects of EVs obtained from an oviductal epithelial cell (OEC) conditioned medium on the developmental competence of in parthenogenetic activation (PA) and somatic cell nuclear transfer (SCNT) porcine embryos. The OEC-EV-treated group showed significant increases in blastocyst formation and hatching rates compared to the control group (40.8% ± 2.2% and 20.1% ± 2.1% vs. 24.9% ± 2.0% and 5.3% ± 1.1%; p < 0.05), respectively. The 7 day OEC-EVs treatment group significantly increased blastocyst formation rate than the 3 day and 0 day-groups (45.0 ± 0.8 vs. 33.0 ± 0.7 and 26.7 ± 0.5; p < 0.05), respectively. SCNT revealed that the OEC-EV increased blastocyst formation rate compared to that of oviductal fluid EVs (OF-EVs) (35.4% ± 1.4% vs. 29.3% ± 1.3%; p < 0.05). Reactive oxygen species levels, apoptosis, and blastocyst lipid content were significantly decreased in the OEC-EVs group compared with the control group. OEC-EV group showed a significantly decreased BAX and increased BCL2, SOD1, POU5F1, SOX2, NANOG, GATA6, PNPLA2, LIPE, and MGLL gene expression than the control group (p < 0.05). In conclusion, OEC-EVs supplementation in embryo culture media improved the quality of porcine embryos, potentially helping porcine-cloned embryonic development possibly through transfer of messenger RNA and proteins to the early embryos.
Collapse
Affiliation(s)
- Xun Fang
- Laboratory of Theriogenology, College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Bereket Molla Tanga
- Laboratory of Theriogenology, College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Seonggyu Bang
- Laboratory of Theriogenology, College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Gyeonghwan Seong
- Laboratory of Theriogenology, College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Islam M Saadeldin
- Laboratory of Theriogenology, College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea.,Research Institute of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea.,Department of Physiology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt
| | - Sanghoon Lee
- Laboratory of Theriogenology, College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Jongki Cho
- Laboratory of Theriogenology, College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
| |
Collapse
|
19
|
Lucas CG, Redel BK, Chen PR, Spate LD, Prather RS, Wells KD. Effects of RAD51-stimulatory compound 1 (RS-1) and its vehicle, DMSO, on pig embryo culture. Reprod Toxicol 2021; 105:44-52. [PMID: 34407461 DOI: 10.1016/j.reprotox.2021.08.002] [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: 05/28/2021] [Revised: 08/01/2021] [Accepted: 08/11/2021] [Indexed: 10/20/2022]
Abstract
Pigs have become an important model for agricultural and biomedical purposes. The advent of genomic engineering tools, such as the CRISPR/Cas9 system, has facilitated the production of livestock models with desired modifications. However, precise site-specific modifications in pigs through the homology-directed repair (HDR) pathway remains a challenge. In mammalian embryos, the use of small molecules to inhibit non-homologous end joining (NHEJ) or to improve HDR have been tested, but little is known about their toxicity. The compound RS-1 stimulates the activity of the RAD51 protein, which plays a key role in the HDR mechanism, demonstrating enhancement of HDR events in rabbit and bovine zygotes. Thus, in this study, we evaluated the dosage and temporal effects of RS-1 on porcine embryo development and viability. Additionally, we assessed the effects of its vehicle, DMSO, during embryo in vitro culture. Transient exposure to 7.5 μM of RS-1 did not adversely affect early embryo development and was compatible with subsequent development to term. Additionally, low concentrations of its vehicle, DMSO, did not show any toxicity to in vitro produced embryos. The transient use of RS-1 at 7.5 μM during in vitro culture seems to be the best protocol of choice to reduce the potentially toxic effects of RS-1 while attempting to improve HDR in the pig. Direct injection of the CRISPR/Cas9 system, combined with strategies to increase the frequency of targeted modifications via HDR, have become an important tool to simplify and accelerate the production of genetically modified livestock models.
Collapse
Affiliation(s)
- C G Lucas
- National Swine Resource and Research Center, University of Missouri, Columbia, MO, USA; Division of Animal Science, University of Missouri, Columbia, MO, USA.
| | - B K Redel
- National Swine Resource and Research Center, University of Missouri, Columbia, MO, USA; USDA-ARS, Plant Genetics Unit, Columbia, MO, USA
| | - P R Chen
- Division of Animal Science, University of Missouri, Columbia, MO, USA
| | - L D Spate
- Division of Animal Science, University of Missouri, Columbia, MO, USA
| | - R S Prather
- National Swine Resource and Research Center, University of Missouri, Columbia, MO, USA; Division of Animal Science, University of Missouri, Columbia, MO, USA
| | - K D Wells
- National Swine Resource and Research Center, University of Missouri, Columbia, MO, USA; Division of Animal Science, University of Missouri, Columbia, MO, USA
| |
Collapse
|
20
|
Li Y, Adur MK, Wang W, Schultz RB, Hale B, Wierson W, Charley SE, McGrail M, Essner J, Tuggle CK, Ross JW. Effect of ARTEMIS (DCLRE1C) deficiency and microinjection timing on editing efficiency during somatic cell nuclear transfer and in vitro fertilization using the CRISPR/Cas9 system. Theriogenology 2021; 170:107-116. [PMID: 34004455 PMCID: PMC8243557 DOI: 10.1016/j.theriogenology.2021.04.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 03/10/2021] [Accepted: 04/14/2021] [Indexed: 01/17/2023]
Abstract
The ability to efficiently introduce site-specific genetic modifications to the mammalian genome has been dramatically improved with the use of the CRISPR/Cas9 system. CRISPR/Cas9 is a powerful tool used to generate genetic modifications by causing double-strand breaks (DSBs) in DNA. Artemis (ART; also known as DCLRE1C), is a nuclear protein and is essential for DSB end joining in DNA repair via the canonical non-homologous end joining (c-NHEJ) pathway. In this work, we tested whether ART deficiency affects DNA repair following CRISPR/Cas9 induced DSBs in somatic cells. We also demonstrated the effect of microinjection timing on embryo developmental ability and gene targeting efficiency of CRISPR/Cas9 system to disrupt the interleukin 2 receptor subunit gamma (IL2RG) locus using porcine in vitro fertilization (IVF) and somatic cell nuclear transfer (SCNT) derived embryos. In comparison to non-injected controls, CRISPR/Cas9 injection of IVF derived zygotes at 4 h and 8 h after fertilization did not impact cleavage and blastocyst rate. Gene modification rate was observed to be higher, 53.3% (9/16) in blastocysts injected 4 h post-fertilization as compared to 11.1% (1/9) in blastocysts injected 8 h post-fertilization. Microinjection 8 h after chemical activation of SCNT derived embryos decreased blastocyst development rate compared to non-injected controls but showed a higher gene modification efficiency of 66.7% as compared to 25% in the 4 h post-activation injection group. Furthermore, we observed that male ART-/- and ART+/- porcine fetal fibroblast (pFF) cells showed lower modification rates (2.5% and 1.9%, respectively) as compared to the ART intact cell line (8.3%). Interestingly, the female ART-/- and ART+/- pFF cells had modification rates (4.2% and 10.1%, respectively) similar to those seen in the ART intact cells. This study demonstrates the complex effect of various parameters such as microinjection timing and ART deficiency on gene editing efficiency in in vitro derived porcine embryos.
Collapse
Affiliation(s)
- Yunsheng Li
- Department of Animal Science, Iowa State University, Ames, Iowa, United States
| | - Malavika K. Adur
- Department of Animal Science, Iowa State University, Ames, Iowa, United States
| | - Wei Wang
- Department of Animal Science, Iowa State University, Ames, Iowa, United States
| | - R. Blythe Schultz
- Department of Animal Science, Iowa State University, Ames, Iowa, United States
| | - Benjamin Hale
- Department of Animal Science, Iowa State University, Ames, Iowa, United States
| | - Wesley Wierson
- Genetics, Development and Cell Biology, Iowa State University, Ames, Iowa, United States
| | - Sara E. Charley
- Department of Animal Science, Iowa State University, Ames, Iowa, United States
| | - Maura McGrail
- Genetics, Development and Cell Biology, Iowa State University, Ames, Iowa, United States
| | - Jeffrey Essner
- Genetics, Development and Cell Biology, Iowa State University, Ames, Iowa, United States
| | | | - Jason W. Ross
- Department of Animal Science, Iowa State University, Ames, Iowa, United States
| |
Collapse
|
21
|
Xu M, Weng Q, Ji J. Applications and advances of CRISPR/Cas9 in animal cancer model. Brief Funct Genomics 2021; 19:235-241. [PMID: 32124927 DOI: 10.1093/bfgp/elaa002] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 01/07/2020] [Indexed: 01/18/2023] Open
Abstract
The recent developments of clustered regularly interspaced short palindromic repeats(CRISPR)/-associate protein 9 (CRISPR/Cas9) have got scientific interests due to the straightforward, efficient and versatile talents of it. Furthermore, the CRISPR/Cas9 system has democratized access to gene editing in many biological fields, including cancer. Cancer development is a multistep process caused by innate and acquired mutations and leads to the initiation and progression of tumorigenesis. It is obvious that establishing appropriate animal cancer models which can simulate human cancers is crucial for cancer research currently. Since the emergence of CRISPR/Cas9, considerable efforts have been taken by researchers to apply this technology in generating animal cancer models. Although there is still a long way to go we are happy to see the achievements we have made and the promising future we have.
Collapse
|
22
|
Shim J, Ko N, Kim HJ, Lee Y, Lee JW, Jin DI, Kim H, Choi K. Human immune reactivity of GGTA1/CMAH/A3GALT2 triple knockout Yucatan miniature pigs. Transgenic Res 2021; 30:619-634. [PMID: 34232440 PMCID: PMC8478729 DOI: 10.1007/s11248-021-00271-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Accepted: 06/11/2021] [Indexed: 11/25/2022]
Abstract
In this study, we investigated the effect of a triple knockout of the genes alpha-1,3-galactosyltransferase (GGTA1), cytidine monophosphate-N-acetylneuraminic acid hydroxylase (CMAH), and alpha 1,3-galactosyltransferase 2 (A3GALT2) in Yucatan miniature pigs on human immune reactivity. We used the CRISPR/Cas9 system to create pigs lacking GGTA1 (GTKO) and GGTA1/CMAH/A3GALT2 triple gene knockout (TKO). The expression of all three xenoantigens was absent in TKO pigs, but there was no additional reduction in the level of Galα1,3Gal (αGal) epitopes expression in the A3GALT2 gene KO. Peripheral blood mononuclear cells (PBMCs), aorta endothelial cells (AECs), and cornea endothelial cells (CECs) were isolated from these pigs, and their ability to bind human IgM/IgG and their cytotoxicity in human sera were evaluated. Compared to wild type (WT) pigs, the level of human antibody binding of the PBMCs, AECs, and CECs of the transgenic pigs (GTKO and TKO) was significantly reduced. However, there were significant differences in human antibody binding between GTKO and TKO depending on the cell type. Human antibody binding of TKO pigs was less than that of GTKO on PBMCs but was similar between GTKO and TKO pigs for AECs and CECs. Cytotoxicity of transgenic pig (GTKO and TKO) PBMCs and AECs was significantly reduced compared to that of WT pigs. However, TKO pigs showed a reduction in cytotoxicity compared to GTKO pigs on PBMCs, whereas in AECs from both TKO and GTKO pigs, there was no difference. The cytotoxicity of transgenic pig CECs was significantly decreased from that of WT at 300 min, but there was no significant reduction in TKO pigs from GTKO. Our results indicate that genetic modification of donor pigs for xenotransplantation should be tailored to the target organ and silencing of additional genes such as CMAH or A3GALT2 based on GTKO might not be essential in Yucatan miniature pigs.
Collapse
Affiliation(s)
- Joohyun Shim
- Department of Transgenic Animal Research, Optipharm, Inc., Chungcheongbuk-do, Cheongju-si, 28158, Republic of Korea.,Department of Animal Science and Biotechnology, Chungnam National University, Daejeon, Republic of Korea
| | - Nayoung Ko
- Department of Transgenic Animal Research, Optipharm, Inc., Chungcheongbuk-do, Cheongju-si, 28158, Republic of Korea.,Department of Animal Science and Biotechnology, Chungnam National University, Daejeon, Republic of Korea
| | - Hyoung-Joo Kim
- Department of Transgenic Animal Research, Optipharm, Inc., Chungcheongbuk-do, Cheongju-si, 28158, Republic of Korea
| | - Yongjin Lee
- Department of Transgenic Animal Research, Optipharm, Inc., Chungcheongbuk-do, Cheongju-si, 28158, Republic of Korea
| | - Jeong-Woong Lee
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| | - Dong-Il Jin
- Department of Animal Science and Biotechnology, Chungnam National University, Daejeon, Republic of Korea
| | - Hyunil Kim
- Department of Transgenic Animal Research, Optipharm, Inc., Chungcheongbuk-do, Cheongju-si, 28158, Republic of Korea
| | - Kimyung Choi
- Department of Transgenic Animal Research, Optipharm, Inc., Chungcheongbuk-do, Cheongju-si, 28158, Republic of Korea.
| |
Collapse
|
23
|
Zhang J, Khazalwa EM, Abkallo HM, Zhou Y, Nie X, Ruan J, Zhao C, Wang J, Xu J, Li X, Zhao S, Zuo E, Steinaa L, Xie S. The advancements, challenges, and future implications of the CRISPR/Cas9 system in swine research. J Genet Genomics 2021; 48:347-360. [PMID: 34144928 DOI: 10.1016/j.jgg.2021.03.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 03/10/2021] [Accepted: 03/13/2021] [Indexed: 12/11/2022]
Abstract
Clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 (CRISPR/Cas9) genome editing technology has dramatically influenced swine research by enabling the production of high-quality disease-resistant pig breeds, thus improving yields. In addition, CRISPR/Cas9 has been used extensively in pigs as one of the tools in biomedical research. In this review, we present the advancements of the CRISPR/Cas9 system in swine research, such as animal breeding, vaccine development, xenotransplantation, and disease modeling. We also highlight the current challenges and some potential applications of the CRISPR/Cas9 technologies.
Collapse
Affiliation(s)
- Jinfu Zhang
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, PR China
| | - Emmanuel M Khazalwa
- Animal and Human Health Program, Biosciences, International Livestock Research Institute (ILRI), P.O. Box 30709, Nairobi 00100, Kenya
| | - Hussein M Abkallo
- Animal and Human Health Program, Biosciences, International Livestock Research Institute (ILRI), P.O. Box 30709, Nairobi 00100, Kenya
| | - Yuan Zhou
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, PR China
| | - Xiongwei Nie
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, PR China
| | - Jinxue Ruan
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, PR China
| | - Changzhi Zhao
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, PR China
| | - Jieru Wang
- Key Laboratory of Pig Molecular Quantitative Genetics of Anhui Academy of Agricultural Sciences, Livestock and Poultry Epidemic Diseases Research Center of Anhui Province, Anhui Provincial Key Laboratory of Livestock and Poultry Product Safety Engineering, Institute of Animal Husbandry and Veterinary Medicine, Anhui Academy of Agricultural Sciences, Hefei 230031, PR China
| | - Jing Xu
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, PR China
| | - Xinyun Li
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, PR China; The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan 430070, PR China
| | - Shuhong Zhao
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, PR China; The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan 430070, PR China
| | - Erwei Zuo
- Lingnan Guangdong Laboratory of Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518124, PR China.
| | - Lucilla Steinaa
- Animal and Human Health Program, Biosciences, International Livestock Research Institute (ILRI), P.O. Box 30709, Nairobi 00100, Kenya.
| | - Shengsong Xie
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, PR China; Animal and Human Health Program, Biosciences, International Livestock Research Institute (ILRI), P.O. Box 30709, Nairobi 00100, Kenya; The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan 430070, PR China.
| |
Collapse
|
24
|
Majumdar U, Yasuhara J, Garg V. In Vivo and In Vitro Genetic Models of Congenital Heart Disease. Cold Spring Harb Perspect Biol 2021; 13:cshperspect.a036764. [PMID: 31818859 DOI: 10.1101/cshperspect.a036764] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Congenital cardiovascular malformations represent the most common type of birth defect and encompass a spectrum of anomalies that range from mild to severe. The etiology of congenital heart disease (CHD) is becoming increasingly defined based on prior epidemiologic studies that supported the importance of genetic contributors and technological advances in human genome analysis. These have led to the discovery of a growing number of disease-contributing genetic abnormalities in individuals affected by CHD. The ever-growing population of adult CHD survivors, which are the result of reductions in mortality from CHD during childhood, and this newfound genetic knowledge have led to important questions regarding recurrence risks, the mechanisms by which these defects occur, the potential for novel approaches for prevention, and the prediction of long-term cardiovascular morbidity in adult CHD survivors. Here, we will review the current status of genetic models that accurately model human CHD as they provide an important tool to answer these questions and test novel therapeutic strategies.
Collapse
Affiliation(s)
- Uddalak Majumdar
- Center for Cardiovascular Research, Nationwide Children's Hospital, Columbus, Ohio 43205, USA.,The Heart Center, Nationwide Children's Hospital, Columbus, Ohio 43205, USA
| | - Jun Yasuhara
- Center for Cardiovascular Research, Nationwide Children's Hospital, Columbus, Ohio 43205, USA.,The Heart Center, Nationwide Children's Hospital, Columbus, Ohio 43205, USA
| | - Vidu Garg
- Center for Cardiovascular Research, Nationwide Children's Hospital, Columbus, Ohio 43205, USA.,The Heart Center, Nationwide Children's Hospital, Columbus, Ohio 43205, USA.,Department of Pediatrics, The Ohio State University, Columbus, Ohio 43205, USA.,Department of Molecular Genetics, The Ohio State University, Columbus, Ohio 43205, USA
| |
Collapse
|
25
|
Chen J, An B, Yu B, Peng X, Yuan H, Yang Q, Chen X, Yu T, Wang L, Zhang X, Wang H, Zou X, Pang D, Ouyang H, Tang X. CRISPR/Cas9-mediated knockin of human factor IX into swine factor IX locus effectively alleviates bleeding in hemophilia B pigs. Haematologica 2021; 106:829-837. [PMID: 31974191 PMCID: PMC7927883 DOI: 10.3324/haematol.2019.224063] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Indexed: 12/30/2022] Open
Abstract
Hemophilia B is an X-linked recessive bleeding disorder caused by abnormalities in the coagulation factor IX gene. Without prophylactic treatment, patients experience frequent spontaneous bleeding episodes. Well-characterized animal models are valuable for determining the pathobiology of the disease and for testing novel therapeutic innovations. Here, we generated a porcine model of hemophilia B (HB) using a combination of CRISPR/Cas9 and somatic cell nuclear transfer. We also tested the possibility of HB therapy by gene insertion. Frequent spontaneous joint bleeding episodes that occurred in HB pigs allowed a thorough investigation of the pathological process of hemophilic arthropathy. In contrast to the HB pigs, which showed a severe bleeding tendency and joint damage, the transgenic pigs carrying human coagulation factor IX exhibited a partial improvement in bleeding. In summary, this study not only offers a translational HB model for exploring the pathological process of hemophilic arthropathy, but also provides a possibility for the permanent correction of hemophilia in the future by genome editing in situ.
Collapse
Affiliation(s)
- Jiahuan Chen
- College of Animal Sciences, Jilin University, Changchun, China
| | - Beiying An
- Department of Medical Laboratory, the First Hospital of Jilin University, Changchun, China
| | - Biao Yu
- College of Animal Sciences, Jilin University, Changchun, China
| | - Xiaohuan Peng
- College of Animal Sciences, Jilin University, Changchun, China
| | - Hongming Yuan
- College of Animal Sciences, Jilin University, Changchun, China
| | - Qiangbing Yang
- College of Animal Sciences, Jilin University, Changchun, China
| | - Xue Chen
- College of Animal Sciences, Jilin University, Changchun, China
| | - Tingting Yu
- College of Animal Sciences, Jilin University, Changchun, China
| | - Lingyu Wang
- College of Animal Sciences, Jilin University, Changchun, China
| | - Xinwei Zhang
- College of Animal Sciences, Jilin University, Changchun, China
| | - He Wang
- College of Animal Sciences, Jilin University, Changchun, China
| | - Xiaodong Zou
- College of Animal Sciences, Jilin University, Changchun, China
| | - Daxin Pang
- College of Animal Sciences, Jilin University, Changchun, China
| | | | - Xiaochun Tang
- College of Animal Sciences, Jilin University, Changchun, China
| |
Collapse
|
26
|
Abstract
BACKGROUND Development of efficient strategies has always been one of the great perspectives for biotechnologists. During the last decade, genome editing of different organisms has been a fast advancing field and therefore has received a lot of attention from various researchers comprehensively reviewing latest achievements and offering opinions on future directions. This review presents a brief history, basic principles, advantages and disadvantages, as well as various aspects of each genome editing technology including the modes, applications, and challenges that face delivery of gene editing components. MAIN BODY Genetic modification techniques cover a wide range of studies, including the generation of transgenic animals, functional analysis of genes, model development for diseases, or drug development. The delivery of certain proteins such as monoclonal antibodies, enzymes, and growth hormones has been suffering from several obstacles because of their large size. These difficulties encouraged scientists to explore alternative approaches, leading to the progress in gene editing. The distinguished efforts and enormous experimentation have now been able to introduce methodologies that can change the genetic constitution of the living cell. The genome editing strategies have evolved during the last three decades, and nowadays, four types of "programmable" nucleases are available in this field: meganucleases, zinc finger nucleases, transcription activator-like effector nucleases, and the clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR associated protein 9 (Cas9) (CRISPR/Cas-9) system. Each group has its own characteristics necessary for researchers to select the most suitable method for gene editing tool for a range of applications. Genome engineering/editing technology will revolutionize the creation of precisely manipulated genomes of cells or organisms in order to modify a specific characteristic. Of the potential applications are those in human health and agriculture. Introducing constructs into target cells or organisms is the key step in genome engineering. CONCLUSIONS Despite the success already achieved, the genome editing techniques are still suffering certain difficulties. Challenges must be overcome before the full potential of genome editing can be realized.
Collapse
Affiliation(s)
- Ahmad M Khalil
- Department of Biological Sciences, Yarmouk University, Irbid, Jordan.
| |
Collapse
|
27
|
Abstract
BACKGROUND Development of efficient strategies has always been one of the great perspectives for biotechnologists. During the last decade, genome editing of different organisms has been a fast advancing field and therefore has received a lot of attention from various researchers comprehensively reviewing latest achievements and offering opinions on future directions. This review presents a brief history, basic principles, advantages and disadvantages, as well as various aspects of each genome editing technology including the modes, applications, and challenges that face delivery of gene editing components. MAIN BODY Genetic modification techniques cover a wide range of studies, including the generation of transgenic animals, functional analysis of genes, model development for diseases, or drug development. The delivery of certain proteins such as monoclonal antibodies, enzymes, and growth hormones has been suffering from several obstacles because of their large size. These difficulties encouraged scientists to explore alternative approaches, leading to the progress in gene editing. The distinguished efforts and enormous experimentation have now been able to introduce methodologies that can change the genetic constitution of the living cell. The genome editing strategies have evolved during the last three decades, and nowadays, four types of "programmable" nucleases are available in this field: meganucleases, zinc finger nucleases, transcription activator-like effector nucleases, and the clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR associated protein 9 (Cas9) (CRISPR/Cas-9) system. Each group has its own characteristics necessary for researchers to select the most suitable method for gene editing tool for a range of applications. Genome engineering/editing technology will revolutionize the creation of precisely manipulated genomes of cells or organisms in order to modify a specific characteristic. Of the potential applications are those in human health and agriculture. Introducing constructs into target cells or organisms is the key step in genome engineering. CONCLUSIONS Despite the success already achieved, the genome editing techniques are still suffering certain difficulties. Challenges must be overcome before the full potential of genome editing can be realized.
Collapse
Affiliation(s)
- Ahmad M Khalil
- Department of Biological Sciences, Yarmouk University, Irbid, Jordan.
| |
Collapse
|
28
|
Polkoff KM, Chung J, Simpson SG, Gleason K, Piedrahita JA. In Vitro Validation of Transgene Expression in Gene-Edited Pigs Using CRISPR Transcriptional Activators. CRISPR J 2020; 3:409-418. [PMID: 33095051 PMCID: PMC7580606 DOI: 10.1089/crispr.2020.0037] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The use of CRISPR-Cas and RNA-guided endonucleases has drastically changed research strategies for understanding and exploiting gene function, particularly for the generation of gene-edited animal models. This has resulted in an explosion in the number of gene-edited species, including highly biomedically relevant pig models. However, even with error-free DNA insertion or deletion, edited genes are occasionally not expressed and/or translated as expected. Therefore, there is a need to validate the expression outcomes gene modifications in vitro before investing in the costly generation of a gene-edited animal. Unfortunately, many gene targets are tissue specific and/or not expressed in cultured primary cells, making validation difficult without generating an animal. In this study, using pigs as a proof of concept, we show that CRISPR-dCas9 transcriptional activators can be used to validate functional transgene insertion in nonexpressing easily cultured cells such as fibroblasts. This is a tool that can be used across disciplines and animal species to save time and resources by verifying expected outcomes of gene edits before generating live animals.
Collapse
Affiliation(s)
- Kathryn M. Polkoff
- Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina, USA
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, USA
| | - Jaewook Chung
- Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina, USA
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, USA
| | - Sean G. Simpson
- Deparment of Animal and Avian Sciences, University of Maryland, College Park, Maryland, USA
- RenOVAte Biosciences, Inc., Reisterstown, Maryland, USA
| | - Katherine Gleason
- Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina, USA
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, USA
| | - Jorge A. Piedrahita
- Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina, USA
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, USA
| |
Collapse
|
29
|
Lucas CG, Spate AM, Samuel MS, Spate LD, Warren WC, Prather RS, Wells KD. A novel swine sex-linked marker and its application across different mammalian species. Transgenic Res 2020; 29:395-407. [PMID: 32607872 DOI: 10.1007/s11248-020-00204-z] [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: 03/13/2020] [Accepted: 06/18/2020] [Indexed: 01/25/2023]
Abstract
Advances in genome editing tools have reduced barriers to the creation of animal models. Due to their anatomical and physiological similarities to humans, there has been a growing need for pig models to study human diseases, for xenotransplantation and translational research. The ability to determine the sex of genetically modified embryos, cells or fetuses is beneficial for every project involving the production of transgenic animals. This strategy can improve the time-efficiency and lower the production costs. Additionally, sex assessment is very useful for wildlife studies to understand population behavior and structure. Thus, we developed a simple and fast PCR-based protocol for sex determination in pigs by using a unique primer set to amplify either the DDX3X or DDX3Y gene. The sex was 100% correctly assigned when tail genomic DNA, Day-35 fetus and hair samples from pigs were used. For both blastocysts and oocytes (84.6% and 96.5% of efficacy, respectively) the unidentified samples were potentially due to a limitation in sample size. Our assay also worked for domestic sheep (Ovis aries), American bison (Bison bison) and European cattle (Bos taurus) samples and by in silico analysis we confirmed X-Y amplicon length polymorphisms for the DDX3 gene in 12 other mammalian species. This PCR protocol for determining sex in pig tissues and cells showed to be simple, specific, highly reproducible and less time consuming as well as an important tool for other livestock species and wildlife studies.
Collapse
Affiliation(s)
- C G Lucas
- National Swine Resource and Research Center, University of Missouri, 920 East Campus Drive, Columbia, MO, 65211, USA.,Division of Animal Science, University of Missouri, Columbia, MO, USA
| | - A M Spate
- National Swine Resource and Research Center, University of Missouri, 920 East Campus Drive, Columbia, MO, 65211, USA.,Division of Animal Science, University of Missouri, Columbia, MO, USA
| | - M S Samuel
- National Swine Resource and Research Center, University of Missouri, 920 East Campus Drive, Columbia, MO, 65211, USA.,Division of Animal Science, University of Missouri, Columbia, MO, USA
| | - L D Spate
- Division of Animal Science, University of Missouri, Columbia, MO, USA
| | - W C Warren
- Bond Life Sciences Center, University of Missouri, Columbia, MO, USA
| | - R S Prather
- National Swine Resource and Research Center, University of Missouri, 920 East Campus Drive, Columbia, MO, 65211, USA.,Division of Animal Science, University of Missouri, Columbia, MO, USA
| | - K D Wells
- National Swine Resource and Research Center, University of Missouri, 920 East Campus Drive, Columbia, MO, 65211, USA. .,Division of Animal Science, University of Missouri, Columbia, MO, USA.
| |
Collapse
|
30
|
Abstract
Many factors influence the final oocyte maturation, fertilisation, and early embryo development, and there are both similarities and differences between species. When comparing the advancement of assisted reproductive technologies (ARTs), the development in the bovine species is not far behind the medical front, with around one million in vitro-produced bovine embryos each year. This rate of progress is not seen in the other domestic species. This review aims to give an overview of the development and specific difficulties of in vitro embryo production in various domestic animal species, with the main focus on cows, pigs, and cats. In production animals, the aim of ARTs is commonly to increase the genetic progress, not to treat reproductive failure. The ARTs are also used for preservation of genetic diversity for the future. However, specifically for oocyte maturation, fertilisation, and early embryonic development, domestic mammals such as the cow and pig can be used as models for humans. This is particularly attractive from an animal welfare point of view since bovine and porcine oocytes are available in large numbers from discarded slaughterhouse material, thereby decreasing the need for research animals. Both for researchers on the animal and human medical fronts, we aim for the development of in vitro production systems that will produce embryos and offspring that are no different from those conceived and developed in vivo. Species-comparative research and development can provide us with crucial knowledge to achieve this aim and hopefully help us avoid unnecessary problems in the future.
Collapse
Affiliation(s)
- Ylva Sjunnesson
- Department of Clinical Sciences, Reproduction, The Centre for Reproductive Biology in Uppsala (CRU), Swedish University of Agricultural Sciences (SLU), Uppsala, Sweden
- CONTACT Ylva Sjunnesson Department of Clinical Sciences, Reproduction, The Centre for Reproductive Biology in Uppsala (CRU), Swedish University of Agricultural Sciences (SLU), PO Box 7054, SE-750 07Uppsala, Sweden
| |
Collapse
|
31
|
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: 4.2] [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
|
32
|
Iqbal MA, Hong K, Kim JH, Choi Y. Severe combined immunodeficiency pig as an emerging animal model for human diseases and regenerative medicines. BMB Rep 2020. [PMID: 31722780 PMCID: PMC6889892 DOI: 10.5483/bmbrep.2019.52.11.267] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Severe combined immunodeficiency (SCID) is a group of inherited disorders characterized by compromised T lymphocyte differentiation related to abnormal development of other lymphocytes [i.e., B and/or natural killer (NK) cells], leading to death early in life unless treated immediately with hematopoietic stem cell transplant. Functional NK cells may impact engraftment success of life-saving procedures such as bone marrow transplantation in human SCID patients. Therefore, in animal models, a T cell−/B cell−/NK cell+ environment provides a valuable tool for understanding the function of the innate immune system and for developing targeted NK therapies against human immune diseases. In this review, we focus on underlying mechanisms of human SCID, recent progress in the development of SCID animal models, and utilization of SCID pig model in biomedical sciences. Numerous physiologies in pig are comparable to those in human such as immune system, X-linked heritability, typical T−B+NK− cellular phenotype, and anatomy. Due to analogous features of pig to those of human, studies have found that immunodeficient pig is the most appropriate model for human SCID.
Collapse
Affiliation(s)
- Muhammad Arsalan Iqbal
- Department of Stem Cell and Regenerative Biotechnology, Humanized Pig Research Center (SRC), Konkuk University, Seoul 05029, Korea
| | - Kwonho Hong
- Department of Stem Cell and Regenerative Biotechnology, Humanized Pig Research Center (SRC), Konkuk University, Seoul 05029, Korea
| | - Jin Hoi Kim
- Department of Stem Cell and Regenerative Biotechnology, Humanized Pig Research Center (SRC), Konkuk University, Seoul 05029, Korea
| | - Youngsok Choi
- Department of Stem Cell and Regenerative Biotechnology, Humanized Pig Research Center (SRC), Konkuk University, Seoul 05029, Korea
| |
Collapse
|
33
|
Zhu X, Wei Y, Zhan Q, Yan A, Feng J, Liu L, Tang D. CRISPR/Cas9-Mediated Biallelic Knockout of IRX3 Reduces the Production and Survival of Somatic Cell-Cloned Bama Minipigs. Animals (Basel) 2020; 10:E501. [PMID: 32192102 PMCID: PMC7142520 DOI: 10.3390/ani10030501] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 03/11/2020] [Accepted: 03/16/2020] [Indexed: 02/06/2023] Open
Abstract
Bama minipigs are a local pig breed that is unique to China and has a high development and utilization value. However, its high fat content, low feed utilization rate, and slow growth rate have limited its popularity and utilization. Compared with the long breeding cycle and high cost of traditional genetic breeding of pigs, clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated (Cas) endonuclease 9 system (CRISPR/Cas9)-mediated gene editing can cost-effectively implement targeted mutations in animal genomes, thereby providing a powerful tool for rapid improvement of the economic traits of Bama minipigs. The iroquois homeobox 3 (IRX3) gene has been implicated in human obesity. Mouse experiments have shown that knocking out IRX3 significantly enhances basal metabolism, reduces fat content, and controls body mass and composition. This study aimed to knock out IRX3 using the CRISPR/Cas9 gene editing method to breed Bama minipigs with significantly reduced fat content. First, the CRISPR/Cas9 gene editing method was used to efficiently obtain IRX3-/- cells. Then, the gene-edited cells were used as donor cells to produce surviving IRX3-/- Bama minipigs using somatic cell cloning. The results show that the use of IRX3-/- cells as donor cells for the production of somatic cell-cloned pigs results in a significant decrease in the average live litter size and a significant increase in the average number of stillbirths. Moreover, the birth weight of surviving IRX3-/- somatic cell-cloned pigs is significantly lower, and viability is poor such that all piglets die shortly after birth. Therefore, the preliminary results of this study suggest that IRX3 may have important biological functions in pigs, and IRX3 should not be used as a gene editing target to reduce fat content in Bama minipigs. Moreover, this study shows that knocking out IRX3 does not favor the survival of pigs, and whether targeted regulation of IRX3 in the treatment of human obesity will also induce severe adverse consequences requires further investigation.
Collapse
Affiliation(s)
- Xiangxing Zhu
- Guangdong Provincial Engineering and Technology Research Center for Gene Editing, School of Medical Engineering, Foshan University, Foshan 528225, China; (A.Y.); (J.F.); (L.L.)
| | - Yanyan Wei
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, School of Life Science and Engineering, Foshan University, Foshan 528225, China; (Y.W.); (Q.Z.)
| | - Qunmei Zhan
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, School of Life Science and Engineering, Foshan University, Foshan 528225, China; (Y.W.); (Q.Z.)
| | - Aifen Yan
- Guangdong Provincial Engineering and Technology Research Center for Gene Editing, School of Medical Engineering, Foshan University, Foshan 528225, China; (A.Y.); (J.F.); (L.L.)
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, School of Life Science and Engineering, Foshan University, Foshan 528225, China; (Y.W.); (Q.Z.)
| | - Juan Feng
- Guangdong Provincial Engineering and Technology Research Center for Gene Editing, School of Medical Engineering, Foshan University, Foshan 528225, China; (A.Y.); (J.F.); (L.L.)
| | - Lian Liu
- Guangdong Provincial Engineering and Technology Research Center for Gene Editing, School of Medical Engineering, Foshan University, Foshan 528225, China; (A.Y.); (J.F.); (L.L.)
| | - Dongsheng Tang
- Guangdong Provincial Engineering and Technology Research Center for Gene Editing, School of Medical Engineering, Foshan University, Foshan 528225, China; (A.Y.); (J.F.); (L.L.)
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, School of Life Science and Engineering, Foshan University, Foshan 528225, China; (Y.W.); (Q.Z.)
| |
Collapse
|
34
|
Abstract
This chapter highlights the importance of reproductive technologies that are applied to porcine breeds. Nowadays the porcine industry, part of a high technological and specialized sector, offers high-quality protein food. The development of the swine industry is founded in the development of breeding/genetics, nutrition, animal husbandry, and animal health. The implementation of reproductive technologies in swine has conducted to levels of productivity never reached before. In addition, the pig is becoming an important species for biomedicine. The generation of pig models for human disease, xenotransplantation, or production of therapeutic proteins for human medicine has in fact generated a growing field of interest.
Collapse
|
35
|
Abstract
In recent years, tremendous advances have been made in our ability to characterize complex microbial communities such as the gut microbiota, and numerous surveys of the human gut microbiota have identified countless associations between different compositional attributes of the gut microbiota and adverse health conditions. However, most of these findings in humans are purely correlative and animal models are required for prospective evaluation of such changes as causative factors in disease initiation or progression. As in most fields of biomedical research, microbiota-focused studies are predominantly performed in mouse or rat models. Depending on the field of research and experimental question or objective, non-rodent models may be preferable due to better translatability or an inability to use rodents for various reasons. The following review describes the utility and limitations of several non-rodent model species for research on the microbiota and its influence on host physiology and disease. In an effort to balance the breadth of potential model species with the amount of detail provided, four model species are discussed: zebrafish, dogs, pigs, and rabbits.
Collapse
Affiliation(s)
- Aaron C Ericsson
- Department of Veterinary Pathobiology, University of Missouri, United States of America
| |
Collapse
|
36
|
Savvulidi F, Ptacek M, Savvulidi Vargova K, Stadnik L. Manipulation of spermatogonial stem cells in livestock species. J Anim Sci Biotechnol 2019; 10:46. [PMID: 31205688 PMCID: PMC6560896 DOI: 10.1186/s40104-019-0355-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Accepted: 04/17/2019] [Indexed: 12/12/2022] Open
Abstract
We are entering an exciting epoch in livestock biotechnology during which the fundamental approaches (such as transgenesis, spermatozoa cryopreservation and artificial insemination) will be enhanced based on the modern understanding of the biology of spermatogonial stem cells (SSCs) combined with the outstanding recent advances in genomic editing technologies and in vitro cell culture systems. The general aim of this review is to outline comprehensively the promising applications of SSC manipulation that could in the nearest future find practical application in livestock breeding. Here, we will focus on 1) the basics of mammalian SSC biology; 2) the approaches for SSC isolation and purification; 3) the available in vitro systems for the stable expansion of isolated SSCs; 4) a discussion of how the manipulation of SSCs can accelerate livestock transgenesis; 5) a thorough overview of the techniques of SSC transplantation in livestock species (including the preparation of recipients for SSC transplantation, the ultrasonographic-guided SSC transplantation technique in large farm animals, and the perspectives to improve further the SSC transplantation efficiency), and finally, 6) why SSC transplantation is valuable to extend the techniques of spermatozoa cryopreservation and/or artificial insemination. For situations where no reliable data have yet been obtained for a particular livestock species, we will rely on the data obtained from studies conducted in rodents because the knowledge gained from rodent research is translatable to livestock species to a great extent. On the other hand, we will draw special attention to situations where such translation is not possible.
Collapse
Affiliation(s)
- Filipp Savvulidi
- Department of Animal Science, Faculty of Agrobiology, Food and Natural Resources, Czech University of Life Sciences Prague, Kamýcká 129, 165 00 Prague, Suchdol Czech Republic
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University in Prague, U Nemocnice 5, 128 53 Prague, Czech Republic
| | - Martin Ptacek
- Department of Animal Science, Faculty of Agrobiology, Food and Natural Resources, Czech University of Life Sciences Prague, Kamýcká 129, 165 00 Prague, Suchdol Czech Republic
| | - Karina Savvulidi Vargova
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University in Prague, U Nemocnice 5, 128 53 Prague, Czech Republic
| | - Ludek Stadnik
- Department of Animal Science, Faculty of Agrobiology, Food and Natural Resources, Czech University of Life Sciences Prague, Kamýcká 129, 165 00 Prague, Suchdol Czech Republic
| |
Collapse
|
37
|
Wu G, Bazer FW. Application of new biotechnologies for improvements in swine nutrition and pork production. J Anim Sci Biotechnol 2019; 10:28. [PMID: 31019685 PMCID: PMC6474057 DOI: 10.1186/s40104-019-0337-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 02/17/2019] [Indexed: 12/18/2022] Open
Abstract
Meeting the increasing demands for high-quality pork protein requires not only improved diets but also biotechnology-based breeding to generate swine with desired production traits. Biotechnology can be classified as the cloning of animals with identical genetic composition or genetic engineering (via recombinant DNA technology and gene editing) to produce genetically modified animals or microorganisms. Cloning helps to conserve species and breeds, particularly those with excellent biological and economical traits. Recombinant DNA technology combines genetic materials from multiple sources into single cells to generate proteins. Gene (genome) editing involves the deletion, insertion or silencing of genes to produce: (a) genetically modified pigs with important production traits; or (b) microorganisms without an ability to resist antimicrobial substances. Current gene-editing tools include the use of zinc finger nuclease (ZFN), transcription activator-like effector nuclease (TALEN), or clustered regularly interspaced short palindromic repeats-associated nuclease-9 (CRISPR/Cas9) as editors. ZFN, TALEN, or CRISPR/Cas9 components are delivered into target cells through transfection (lipid-based agents, electroporation, nucleofection, or microinjection) or bacteriophages, depending on cell type and plasmid. Compared to the ZFN and TALEN, CRISPR/Cas9 offers greater ease of design and greater flexibility in genetic engineering, but has a higher frequency of off-target effects. To date, genetically modified pigs have been generated to express bovine growth hormone, bacterial phytase, fungal carbohydrases, plant and C. elagan fatty acid desaturases, and uncoupling protein-1; and to lack myostatin, α-1,3-galactosyltransferase, or CD163 (a cellular receptor for the "blue ear disease" virus). Biotechnology holds promise in improving the efficiency of swine production and developing alternatives to antibiotics in the future.
Collapse
Affiliation(s)
- Guoyao Wu
- Department of Animal Science and Center for Animal Biotechnology and Genomics, Texas A&M University, College Station, TX 77843-2471 USA
| | - Fuller W Bazer
- Department of Animal Science and Center for Animal Biotechnology and Genomics, Texas A&M University, College Station, TX 77843-2471 USA
| |
Collapse
|
38
|
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. [PMID: 30786106 DOI: 10.1002/jgm.3082] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [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
|
39
|
Song YW, Pan ZQ. Reducing porcine corneal graft rejection, with an emphasis on porcine endogenous retrovirus transmission safety: a review. Int J Ophthalmol 2019; 12:324-332. [PMID: 30809491 DOI: 10.18240/ijo.2019.02.21] [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: 05/15/2018] [Accepted: 11/28/2018] [Indexed: 01/08/2023] Open
Abstract
Donor cornea shortage is a primary hurdle in the development of corneal transplantation. Of all species, porcine corneas are the ideal transplantation material for humans. However, the xenoimmune rejection induced by porcine corneal xenotransplantation compromises surgical efficacy. Although the binding of IgM/IgG in human serum to a genetically modified porcine cornea is significantly weaker than that of the wild type (WT), genetically modified porcine corneas do not display a prolonged graft survival time in vivo. Conversely, costimulatory blockade drugs, such as anti-CD40 antibodies, can reduce the xenoimmune response and prolong graft survival time in animal experiments. Moreover, porcine endothelial grafts can survive for more than 6mo with only the subconjunctival injection of a steroid-based immunosuppressants regime; therefore, they show great value for treating corneal endothelial disease. In addition, zoonotic transmission is a primary concern of xenotransplantation. Porcine endogenous retrovirus (PERV) is the most significant virus assessed by ophthalmologists. PERV integrates into the porcine genome and infects human cells in vitro. Fortunately, no evidence from in vivo studies has yet shown that PERV can be transmitted to hosts.
Collapse
Affiliation(s)
- Yao-Wen Song
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmology and Visual Science Key Laboratory, Beijing 100730, China
| | - Zhi-Qiang Pan
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmology and Visual Science Key Laboratory, Beijing 100730, China
| |
Collapse
|
40
|
Su X, Wang S, Su G, Zheng Z, Zhang J, Ma Y, Liu Z, Zhou H, Zhang Y, Zhang L. Production of microhomologous-mediated site-specific integrated LacS gene cow using TALENs. Theriogenology 2018; 119:282-288. [PMID: 30075414 DOI: 10.1016/j.theriogenology.2018.07.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Revised: 07/13/2018] [Accepted: 07/14/2018] [Indexed: 12/31/2022]
Abstract
Gene editing tools (Zinc-Finger Nucleases, ZFN; Transcription Activator-Like Effector Nucleases, TALEN; and Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)/CRISPR-associated (Cas)9, CRISPR-Cas9) provide us with a powerful means of performing genetic engineering procedures. A combinational approach that utilizes both somatic cell nuclear transfer (SCNT) and somatic cell gene editing facilitates the generation of genetically engineered animals. However, the associated research has utilized markers and/or selected genes, which constitute a potential threat to biosafety. Microhomologous-mediated end-joining (MMEJ) has showed the utilization of micro-homologous arms (5-25 bp) can mediate exogenous gene insertion. Dairy milk is a major source of nutrition worldwide. However, most people are not capable of optimally utilizing the nutrition in milk because of lactose intolerance. Sulfolobus solfataricus β-glycosidase (LacS) is a lactase derived from the extreme thermophilic archaeon Sulfolobus solfataricus. Our finally aim was to site-specific integrated LacS gene into cow's genome through TALEN-mediated MMEJ and produce low-lactose cow. Firstly, we constructed TALENs vectors which target to the cow's β-casein locus and LacS gene expression vector which contain TALEN reorganization sequence and micro-homologous arms. Then we co-transfected these vectors into fetal derived skin fibroblasts and cultured as monoclone. Positive cell clones were screened using 3' junction PCR amplification and sequencing analysis. The positive cells were used as donors for SCNT and embryo transfer (ET). Lastly, we detected the genotype through PCR of blood genomic DNA. This resulted in a LacS knock-in rate of 0.8% in TALEN-treated cattle fetal fibroblasts. The blastocyst rate of SCNT embryo was 27%. The 3 months pregnancy rate was 20%. Finally, we obtained 1 newborn cow (5%) and verified its genotype. We obtained 1 site-specific marker-free LacS transgenic cow. It provides a basis to solve lactose intolerance by gene engineering breeding. This study also provides us with a new strategy to facilitate gene knock-ins in livestock using techniques that exhibit improved biosafety and intuitive methodologies.
Collapse
Affiliation(s)
- Xiaohu Su
- Key Laboratory of Gene Engineering of the Ministry of Education, Guangzhou Key Laboratory of Healthy Aging Research and State Key Laboratory of Biocontrol, SYSU-BCM JointResearch Center, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, PR China
| | - Shenyuan Wang
- Key Laboratory of Biological Manufacturing of Inner Mongolia Autonomous Region, College of Life Sciences, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia Autonomous Region, 010018, PR China
| | - Guanghua Su
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Inner Mongolia University, Hohhot, Inner Mongolia Autonomous Region, 010018, PR China
| | - Zhong Zheng
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Inner Mongolia University, Hohhot, Inner Mongolia Autonomous Region, 010018, PR China
| | - Jiaqi Zhang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Yunlong Ma
- Key Laboratory of Biological Manufacturing of Inner Mongolia Autonomous Region, College of Life Sciences, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia Autonomous Region, 010018, PR China
| | - Zongzheng Liu
- Qingdao Animal Husbandry and Veterinary Research Institution, Qingdao, ShanDong, 266100, PR China
| | - Huanmin Zhou
- Key Laboratory of Biological Manufacturing of Inner Mongolia Autonomous Region, College of Life Sciences, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia Autonomous Region, 010018, PR China
| | - Yanru Zhang
- Key Laboratory of Biological Manufacturing of Inner Mongolia Autonomous Region, College of Life Sciences, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia Autonomous Region, 010018, PR China.
| | - Li Zhang
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Inner Mongolia University, Hohhot, Inner Mongolia Autonomous Region, 010018, PR China.
| |
Collapse
|
41
|
Karuppannan AK, Opriessnig T. Possible risks posed by single-stranded DNA viruses of pigs associated with xenotransplantation. Xenotransplantation 2018; 25:e12453. [PMID: 30264878 PMCID: PMC6120555 DOI: 10.1111/xen.12453] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 07/12/2018] [Accepted: 07/20/2018] [Indexed: 12/11/2022]
Abstract
Routine large-scale xenotransplantation from pigs to humans is getting closer to clinical reality owing to several state-of-the-art technologies, especially the ability to rapidly engineer genetically defined pigs. However, using pig organs in humans poses risks including unwanted cross-species transfer of viruses and adaption of these pig viruses to the human organ recipient. Recent developments in the field of virology, including the advent of metagenomic techniques to characterize entire viromes, have led to the identification of a plethora of viruses in many niches. Single-stranded DNA (ssDNA) viruses are the largest group prevalent in virome studies in mammals. Specifically, the ssDNA viral genomes are characterized by a high rate of nucleotide substitution, which confers a proclivity to adapt to new hosts and cross-species barriers. Pig-associated ssDNA viruses include torque teno sus viruses (TTSuV) in the Anelloviridae family, porcine parvoviruses (PPV), and porcine bocaviruses (PBoV) both in the family of Parvoviridae, and porcine circoviruses (PCV) in the Circoviridae family, some of which have been confirmed to be pathogenic to pigs. The risks of these viruses for the human recipient during xenotransplantation procedures are relatively unknown. Based on the scant knowledge available on the prevalence, predilection, and pathogenicity of pig-associated ssDNA viruses, careful screening and monitoring are required. In the case of positive identification, risk assessments and strategies to eliminate these viruses in xenotransplantation pig stock may be needed.
Collapse
Affiliation(s)
- Anbu K. Karuppannan
- Department of Veterinary Diagnostic and Production Animal MedicineCollege of Veterinary MedicineIowa State UniversityAmesIowa
| | - Tanja Opriessnig
- Department of Veterinary Diagnostic and Production Animal MedicineCollege of Veterinary MedicineIowa State UniversityAmesIowa
- The Roslin Institute and The Royal (Dick) School of Veterinary StudiesUniversity of EdinburghRoslinMidlothianUK
| |
Collapse
|
42
|
Gene-knocked out chimeric antigen receptor (CAR) T cells: Tuning up for the next generation cancer immunotherapy. Cancer Lett 2018; 423:95-104. [DOI: 10.1016/j.canlet.2018.03.010] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 03/06/2018] [Accepted: 03/07/2018] [Indexed: 12/15/2022]
|