1
|
Meng S, Miao A, Wu S, Du X, Gao F. Genetically modified chickens as bioreactors for protein-based drugs. Front Genome Ed 2025; 6:1522837. [PMID: 39845893 PMCID: PMC11753250 DOI: 10.3389/fgeed.2024.1522837] [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: 11/05/2024] [Accepted: 12/18/2024] [Indexed: 01/24/2025] Open
Abstract
Protein drug production encompasses various methods, among which animal bioreactors are emerging as a transgenic system. Animal bioreactors have the potential to reduce production costs and increase efficiency, thereby producing recombinant proteins that are crucial for therapeutic applications. Various species, including goats, cattle, rabbits, and poultry, have been genetically engineered to serve as bioreactors. This review delves into the analysis and comparison of different expression systems for protein drug production, highlighting the advantages and limitations of microbial, yeast, plant cell, and mammalian cell expression systems. Additionally, the emerging significance of genetically modified chickens as a potential bioreactor system for producing protein-based drugs is highlighted. The avian bioreactor enables the expression of target genes in ovarian cells, resulting in the production of corresponding gene expression products in egg whites. This production method boasts advantages such as a short cycle, high production efficiency, low research costs, and the expression products being closer to their natural state and easier to purify. It demonstrates immense potential in production applications, scientific research, and sustainable development. The utilization of advanced gene editing technologies, such as CRISPR/Cas9, has revolutionized the precision and efficiency of generating genetically modified chickens. This has paved the way for enhanced production of recombinant therapeutic proteins with desired glycosylation patterns and reduced immunogenic responses.
Collapse
Affiliation(s)
- Shujuan Meng
- Frontiers Science Center for Molecular Design Breeding (MOE), State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Aijun Miao
- Frontiers Science Center for Molecular Design Breeding (MOE), State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Sen Wu
- Frontiers Science Center for Molecular Design Breeding (MOE), State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, China
- Sanya Institute of China Agricultural University, Sanya, China
| | - Xuguang Du
- Frontiers Science Center for Molecular Design Breeding (MOE), State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, China
- Sanya Institute of China Agricultural University, Sanya, China
| | - Fei Gao
- Frontiers Science Center for Molecular Design Breeding (MOE), State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, China
| |
Collapse
|
2
|
Ge G, Li D, Ling Q, Xu L, Ata EB, Wang X, Li K, Hao W, Gong Q, Li J, Shi K, Leng X, Du R. IRF7-deficient MDBK cell based on CRISPR/Cas9 technology for enhancing IBRV replication. Front Microbiol 2024; 15:1483527. [PMID: 39691910 PMCID: PMC11649632 DOI: 10.3389/fmicb.2024.1483527] [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: 08/20/2024] [Accepted: 11/05/2024] [Indexed: 12/19/2024] Open
Abstract
Infectious bovine rhinotracheitis (IBR), characterized by acute respiratory lesions in cattle, is a major infectious disease caused by bovine alphaherpesvirus-1 (BoAHV-1). Control of this disease is primarily depending on vaccination. Madin-Darby bovine kidney cells (MDBK) being the main host cells and the important production platform for IBR vaccines. However, innate immune genes inhibit viral replication. Accordingly, the aim of this study was developing of IRF7 gene deleted MDBK cells to facilitate the production of high-titer vaccines. The CRISPR/Cas9 technology was used to knock out the IRF7 gene in MDBK cells and the impact on virus replication was examined using virus growth curves, CCK-8 assays, cell scratch assays, and qPCR. The knockout of the IRF7 gene in MDBK cells led to an increased replication capacity of IBRV and a significant reduction in type I interferons expression, specifically IFN-α and IFN-β. This indicates that IRF7 -/-MDBK cell lines can effectively result in production of IBRV with high-titer, which will enhance the development of inactivated or attenuated vaccines.
Collapse
Affiliation(s)
- Guiyang Ge
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Dongli Li
- Wengniute Banner Agriculture and Animal Husbandry Bureau, Chifeng, China
| | - Qian Ling
- College of Chinese Medicine Materials, Jilin Agricultural University, Changchun, China
| | - Lihui Xu
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Emad Beshir Ata
- Department of Parasitology and Animal Diseases, Veterinary Research Institute, National Research Centre, Giza, Egypt
| | - Xiaolin Wang
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Keyan Li
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Wen Hao
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Qinglong Gong
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Jianming Li
- College of Chinese Medicine Materials, Jilin Agricultural University, Changchun, China
| | - Kun Shi
- College of Chinese Medicine Materials, Jilin Agricultural University, Changchun, China
| | - Xue Leng
- College of Chinese Medicine Materials, Jilin Agricultural University, Changchun, China
| | - Rui Du
- College of Chinese Medicine Materials, Jilin Agricultural University, Changchun, China
| |
Collapse
|
3
|
Aboelhassan DM, Abozaid H. Opportunities for CRISPR-Cas9 application in farm animal genetic improvement. Mol Biol Rep 2024; 51:1108. [PMID: 39476174 DOI: 10.1007/s11033-024-10052-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 10/22/2024] [Indexed: 02/06/2025]
Abstract
CRISPR-Cas9 has emerged as a powerful tool in livestock breeding, enabling precise genetic modifications to address genetic diseases, enhance productivity, and develop disease-resistant animal breeds. A thorough analysis of previous research highlights the potential of CRISPR-Cas9 in overcoming genetic disorders by targeting specific mutations in genes. Furthermore, its integration with reproductive biotechnologies and genomic selection facilitates the production of gene-edited animals with high genomic value, contributing to genetic enhancement and improved productivity. Additionally, CRISPR-Cas9 opens new avenues for developing disease-resistant livestock and creating innovative breeding models for high-quality production. A key trend in the field is the development of multi-sgRNA vectors to correct mutations in various genes linked to productivity traits or certain diseases within individual genomes, thereby increasing resistance in animals. However, despite the potential advantages of CRISPR-Cas9, public acceptance of genetically modified agricultural products remains uncertain. Would consumers be willing to purchase such products? It is essential to advocate for bold and innovative research into genetically edited animals, with a focus on safety, careful promotion, and strict regulatory oversight to align with long-term goals and public acceptance. Continued advancements in this technology and its underlying mechanisms promise to improve poultry products and genetically modified livestock. Overall, CRISPR-Cas9 technology offers a promising pathway for advancing livestock breeding practices, with opportunities for genetic improvement, enhanced disease resistance, and greater productivity.
Collapse
Affiliation(s)
- Dalia M Aboelhassan
- Department of Cell Biology, Biotechnology Research Institute, National Research Centre, 33 El-Bohouth Street, P.O:12622, Dokki, Giza, Egypt.
| | - Hesham Abozaid
- Department of Animal Production, Agricultural and Biology Research Institute, National Research Centre, 33 El- Bohouth Street, P.O:12622, Dokki, Giza, Egypt
| |
Collapse
|
4
|
Dang W, Gao D, Lyu G, Irwin DM, Shang S, Chen J, Zhang J, Zhang S, Wang Z. A Nonsynonymous Substitution of Lhx3 Leads to Changes in Body Size in Dogs and Mice. Genes (Basel) 2024; 15:739. [PMID: 38927675 PMCID: PMC11202965 DOI: 10.3390/genes15060739] [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: 05/08/2024] [Revised: 05/30/2024] [Accepted: 06/01/2024] [Indexed: 06/28/2024] Open
Abstract
Lhx3 is a LIM-homeodomain transcription factor that affects body size in mammals by regulating the secretion of pituitary hormones. Akita, Shiba Inu, and Mame Shiba Inu dogs are Japanese native dog breeds that have different body sizes. To determine whether Lhx3 plays a role in the differing body sizes of these three dog breeds, we sequenced the Lhx3 gene in the three breeds, which led to the identification of an SNP in codon 280 (S280N) associated with body size. The allele frequency at this SNP differed significantly between the large Akita and the two kinds of smaller Shiba dogs. To validate the function of this SNP on body size, we introduced this change into the Lhx3 gene of mice. Homozygous mutant mice (S279N+/+) were found to have significantly increased body lengths and weights compared to heterozygous mutant (S279N+/-) and wild-type (S279N-/-) mice several weeks after weaning. These results demonstrate that a nonsynonymous substitution in Lhx3 plays an important role in regulating body size in mammals.
Collapse
Affiliation(s)
- Wanyi Dang
- College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang 110866, China
| | - Dali Gao
- College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang 110866, China
| | - Guangqi Lyu
- College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang 110866, China
| | - David M. Irwin
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Songyang Shang
- College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang 110866, China
| | - Junnan Chen
- College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang 110866, China
| | - Junpeng Zhang
- College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang 110866, China
| | - Shuyi Zhang
- College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang 110866, China
| | - Zhe Wang
- College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang 110866, China
| |
Collapse
|
5
|
Xiang C, Yang Z, Xiong T, Wang T, Yang J, Huang M, Liu D, Chen R. Construction and Transcriptomic Study of Chicken IFNAR1-Knockout Cell Line Reveals the Essential Roles of Cell Growth- and Apoptosis-Related Pathways in Duck Tembusu Virus Infection. Viruses 2022; 14:v14102225. [PMID: 36298780 PMCID: PMC9611459 DOI: 10.3390/v14102225] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 09/24/2022] [Accepted: 10/06/2022] [Indexed: 11/06/2022] Open
Abstract
For industrial vaccine production, overwhelming the existing antiviral innate immune response dominated by type I interferons (IFN-I) in cells would be a key factor improving the effectiveness and production cost of vaccines. In this study, we report the construction of an IFN-I receptor 1 (IFNAR1)-knockout DF-1 cell line (KO-IFNAR1), which supports much more efficient replication of the duck Tembusu virus (DTMUV), Newcastle disease virus (NDV) and gammacoronavirus infectious bronchitis virus (IBV). Transcriptomic analysis of DTMUV-infected KO-IFNAR1 cells demonstrated that DTMUV mainly activated genes and signaling pathways related to cell growth and apoptosis. Among them, JUN, MYC and NFKBIA were significantly up-regulated. Furthermore, knockdown of zinc-fingered helicase 2 (HELZ2) and interferon-α-inducible protein 6 (IFI6), the two genes up-regulated in both wild type and KO-IFNAR1 cells, significantly increased the replication of DTMUV RNA. This study paves the way for further studying the mechanism underlying the DTMUV-mediated IFN-I-independent regulation of virus replication, and meanwhile provides a potential cell resource for efficient production of cell-based avian virus vaccines.
Collapse
Affiliation(s)
- Chengwei Xiang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
- Zhaoqing Branch Center of Guangdong Laboratory for Lingnan Modern Agricultural Science and Technology, Zhaoqing 526000, China
| | - Zekun Yang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Ting Xiong
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Ting Wang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Jie Yang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Mei Huang
- Zhaoqing Institute of Biotechnology Co., Ltd., Zhaoqing 526238, China
| | - Dingxiang Liu
- Zhaoqing Branch Center of Guangdong Laboratory for Lingnan Modern Agricultural Science and Technology, Zhaoqing 526000, China
- Integrative Microbiology Research Centre, South China Agricultural University, Guangzhou 510642, China
- Correspondence: (D.L.); (R.C.)
| | - RuiAi Chen
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
- Zhaoqing Branch Center of Guangdong Laboratory for Lingnan Modern Agricultural Science and Technology, Zhaoqing 526000, China
- Correspondence: (D.L.); (R.C.)
| |
Collapse
|
6
|
Barkova OY, Larkina TA, Krutikova AA, Polteva EA, Shcherbakov YS, Peglivanyan GK, Pozovnikova MV. Innovative Approaches to Genome Editing in Chickens. CYTOL GENET+ 2022. [DOI: 10.3103/s0095452722020037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
7
|
Overexpression of bmp4, dazl, nanos3 and sycp2 in Hu Sheep Leydig Cells Using CRISPR/dcas9 System Promoted Male Germ Cell Related Gene Expression. BIOLOGY 2022; 11:biology11020289. [PMID: 35205154 PMCID: PMC8869737 DOI: 10.3390/biology11020289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Revised: 02/02/2022] [Accepted: 02/05/2022] [Indexed: 11/23/2022]
Abstract
Simple Summary Male germ cell development plays a crucial role in male reproduction, and gene expression also presents an essential regulatory role in its development. Many studies have been devoted to the induction and differentiation of pluripotent stem cells into germ cells in vitro. However, the culture system for pluripotent stem cells from domestic animals is not stable, especially in sheep. Our study attempted to transdifferentiate sheep somatic cells into germ cells in vitro by the overexpression of key germ cell related genes, with the aim of perfecting the construction of germ cell research models in vitro. Therefore, we explored the expression pattern of four crucial genes, bmp4, dazl, nanos3 and sycp2, in Hu sheep testicular development, and investigated the potential efficiency of overexpression of the four candidate genes using the CRISPR/dcas9 system in Leydig cells. We revealed that the overexpression of bmp4, dazl, nanos3 and sycp2 can promote the expression of male germ cell related genes. To the best of our knowledge, this is the first study to construct an overexpression induction system using CRISPR/dcas9 technology, and to induce sheep somatic cells into germ cells in vitro. Abstract Male germ cells directly affect the reproduction of males; however, their accurate isolation and culture in vitro is extremely challenging, hindering the study of germ cell development and function. CRISPR/dcas9, as an efficient gene reprogramming system, has been verified to promote the transdifferentiation of pluripotent stem cells into male germ cells by editing target genes. In our research, we explored the expression pattern of the germ cell related genes bmp4, dazl,nanos3 and sycp2 in Hu sheep testicular development and constructed the overexpression model using the CRISPR/dcas9 system. The results indicated that four genes showed more expression in testis tissue than in other tissues, and that bmp4, dazl and sycp2 present higher expression levels in nine-month-old sheep testes than in three-month-olds, while nanos3 expressed the opposite trend (p < 0.05). In addition, the expression of four potential genes in spermatogenic cells was slightly different, but they were all expressed in sheep Leydig cells. To verify the potential roles of the four genes in the process of inducing differentiation of male germ cells, we performed cell transfection in vitro. We found that the expression of the germ cell related genes Prdm1, Prdm14, Mvh and Sox17 were significantly increased after the overexpression of the four genes in Leydig cells, and the co-transfection effect was the most significant (p < 0.05). Our results illustrate the crucial functions of bmp4, dazl, nanos3 and sycp2 in Hu sheep testis development and verified the effectiveness of the overexpression model that was constructed using the CRISPR/dcas9 system, which provided a basis for further male germ cell differentiation in vitro.
Collapse
|
8
|
Sun C, Jin K, Zuo Q, Sun H, Song J, Zhang Y, Chen G, Li B. Characterization of Alternative Splicing (AS) Events during Chicken ( Gallus gallus) Male Germ-Line Stem Cell Differentiation with Single-Cell RNA-seq. Animals (Basel) 2021; 11:ani11051469. [PMID: 34065391 PMCID: PMC8160964 DOI: 10.3390/ani11051469] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 05/17/2021] [Accepted: 05/18/2021] [Indexed: 12/27/2022] Open
Abstract
Simple Summary Studies have shown that alternative splicing (AS) has been utilized in a wide variety of life processes. However, there are very few studies on AS during germ cell development. In this study, we preliminarily investigated the variation of variable shear events during the formation of chicken germ cells through the RNA-seq data analysis of embryonic stem cells (ESCs), gonad PGCs (gPGCs), and spermatogonia stem cells (SSCs), and the critical AS mode for several crucial stage-specific genes, which were identified during germ cell development. The results of this study lay a theoretical foundation for further analysis of the regulation mechanism of key genes involved in germ cell formation. Abstract Alternative splicing (AS) is a ubiquitous, co-transcriptional, and post-transcriptional regulation mechanism during certain developmental processes, such as germ cell differentiation. A thorough understanding of germ cell differentiation will help us to open new avenues for avian reproduction, stem cell biology, and advances in medicines for human consumption. Here, based on single-cell RNA-seq, we characterized genome-wide AS events in manifold chicken male germ cells: embryonic stem cells (ESCs), gonad primordial germ cells (gPGCs), and spermatogonia stem cells (SSCs). A total of 38,494 AS events from 15,338 genes were detected in ESCs, with a total of 48,955 events from 14,783 genes and 49,900 events from 15,089 genes observed in gPGCs and SSCs, respectively. Moreover, this distribution of AS events suggests the diverse splicing feature of ESCs, gPGCs, and SSCs. Finally, several crucial stage-specific genes, such as NANOG, POU5F3, LIN28B, BMP4, STRA8, and LHX9, were identified in AS events that were transmitted in ESCs, gPGCs, and SSCs. The gene expression results of the RNA-seq data were validated by qRT-PCR. In summary, we provided a comprehensive atlas of the genome-wide scale of the AS event landscape in male chicken germ-line cells and presented its distribution for the first time. This research may someday improve treatment options for men suffering from male infertility.
Collapse
Affiliation(s)
- Changhua Sun
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (C.S.); (K.J.); (Q.Z.); (H.S.); (Y.Z.); (G.C.)
- Department of Food Technology, College of Biochemical Engineering, Yangzhou Polytechnic College, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Kai Jin
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (C.S.); (K.J.); (Q.Z.); (H.S.); (Y.Z.); (G.C.)
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Qisheng Zuo
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (C.S.); (K.J.); (Q.Z.); (H.S.); (Y.Z.); (G.C.)
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Hongyan Sun
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (C.S.); (K.J.); (Q.Z.); (H.S.); (Y.Z.); (G.C.)
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Jiuzhou Song
- Animal & Avian Sciences, University of Maryland, College Park, MD 20741, USA;
| | - Yani Zhang
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (C.S.); (K.J.); (Q.Z.); (H.S.); (Y.Z.); (G.C.)
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Guohong Chen
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (C.S.); (K.J.); (Q.Z.); (H.S.); (Y.Z.); (G.C.)
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Bichun Li
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (C.S.); (K.J.); (Q.Z.); (H.S.); (Y.Z.); (G.C.)
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
- Correspondence:
| |
Collapse
|
9
|
Khwatenge CN, Nahashon SN. Recent Advances in the Application of CRISPR/Cas9 Gene Editing System in Poultry Species. Front Genet 2021; 12:627714. [PMID: 33679892 PMCID: PMC7933658 DOI: 10.3389/fgene.2021.627714] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 01/19/2021] [Indexed: 12/28/2022] Open
Abstract
CRISPR/Cas9 system genome editing is revolutionizing genetics research in a wide spectrum of animal models in the genetic era. Among these animals, is the poultry species. CRISPR technology is the newest and most advanced gene-editing tool that allows researchers to modify and alter gene functions for transcriptional regulation, gene targeting, epigenetic modification, gene therapy, and drug delivery in the animal genome. The applicability of the CRISPR/Cas9 system in gene editing and modification of genomes in the avian species is still emerging. Up to date, substantial progress in using CRISPR/Cas9 technology has been made in only two poultry species (chicken and quail), with chicken taking the lead. There have been major recent advances in the modification of the avian genome through their germ cell lineages. In the poultry industry, breeders and producers can utilize CRISPR-mediated approaches to enhance the many required genetic variations towards the poultry population that are absent in a given poultry flock. Thus, CRISPR allows the benefit of accessing genetic characteristics that cannot otherwise be used for poultry production. Therefore CRISPR/Cas9 becomes a very powerful and robust tool for editing genes that allow for the introduction or regulation of genetic information in poultry genomes. However, the CRISPR/Cas9 technology has several limitations that need to be addressed to enhance its use in the poultry industry. This review evaluates and provides a summary of recent advances in applying CRISPR/Cas9 gene editing technology in poultry research and explores its potential use in advancing poultry breeding and production with a major focus on chicken and quail. This could aid future advancements in the use of CRISPR technology to improve poultry production.
Collapse
Affiliation(s)
- Collins N. Khwatenge
- Department of Biological Sciences, Tennessee State University, Nashville, IN, United States
- Department of Agriculture and Environmental Sciences, Tennessee State University, Nashville, TN, United States
| | - Samuel N. Nahashon
- Department of Agriculture and Environmental Sciences, Tennessee State University, Nashville, TN, United States
| |
Collapse
|
10
|
Chojnacka-Puchta L, Sawicka D. CRISPR/Cas9 gene editing in a chicken model: current approaches and applications. J Appl Genet 2020; 61:221-229. [PMID: 31925767 PMCID: PMC7148258 DOI: 10.1007/s13353-020-00537-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Improvements in genome editing technology in birds using primordial germ cells (PGCs) have made the development of innovative era genome-edited avian models possible, including specific chicken bioreactors, production of knock-in/out chickens, low-allergenicity eggs, and disease-resistance models. New strategies, including CRISPR/Cas9, have made gene editing easy and highly efficient in comparison to the well-known process of homologous recombination. The clustered regularly interspaced short palindromic repeats (CRISPR) technique enables us to understand the function of genes and/or to modify the animal phenotype to fit a specific scientific or production target. To facilitate chicken genome engineering applications, we present a concise description of the method and current application of the CRISPR/Cas9 system in chickens. Different strategies for delivering sgRNAs and the Cas9 protein, we also present extensively. Furthermore, we describe a new gesicle technology as a way to deliver Cas9/sgRNA complexes into target cells, and we discuss the advantages and describe basal applications of the CRISPR/Cas9 system in a chicken model.
Collapse
Affiliation(s)
- Luiza Chojnacka-Puchta
- Department of Bioengineering, Lukasiewicz Research Network, Institute of Biotechnology and Antibiotics, Staroscinska 5, 02-516, Warsaw, Poland. .,Department of Chemical, Biological and Aerosol Hazards, Central Institute for Labour Protection-National Research Institute, Czerniakowska 16, 00-701, Warsaw, Poland.
| | - Dorota Sawicka
- Department of Bioengineering, Lukasiewicz Research Network, Institute of Biotechnology and Antibiotics, Staroscinska 5, 02-516, Warsaw, Poland.,Department of Chemical, Biological and Aerosol Hazards, Central Institute for Labour Protection-National Research Institute, Czerniakowska 16, 00-701, Warsaw, Poland
| |
Collapse
|
11
|
Xu K, Han CX, Zhou H, Ding JM, Xu Z, Yang LY, He C, Akinyemi F, Zheng YM, Qin C, Luo HX, Meng H. Effective MSTN Gene Knockout by AdV-Delivered CRISPR/Cas9 in Postnatal Chick Leg Muscle. Int J Mol Sci 2020; 21:ijms21072584. [PMID: 32276422 PMCID: PMC7177447 DOI: 10.3390/ijms21072584] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 04/02/2020] [Accepted: 04/06/2020] [Indexed: 02/07/2023] Open
Abstract
Muscle growth and development are important aspects of chicken meat production, but the underlying regulatory mechanisms remain unclear and need further exploration. CRISPR has been used for gene editing to study gene function in mice, but less has been done in chick muscles. To verify whether postnatal gene editing could be achieved in chick muscles and determine the transcriptomic changes, we knocked out Myostatin (MSTN), a potential inhibitor of muscle growth and development, in chicks and performed transcriptome analysis on knock-out (KO) muscles and wild-type (WT) muscles at two post-natal days: 3d (3-day-old) and 14d (14-day-old). Large fragment deletions of MSTN (>5 kb) were achieved in all KO muscles, and the MSTN gene expression was significantly downregulated at 14d. The transcriptomic results indicated the presence of 1339 differentially expressed genes (DEGs) between the 3d KO and 3d WT muscles, as well as 597 DEGs between 14d KO and 14d WT muscles. Many DEGs were found to be related to cell differentiation and proliferation, muscle growth and energy metabolism. This method provides a potential means of postnatal gene editing in chicks, and the results presented here could provide a basis for further investigation of the mechanisms involved in muscle growth and development.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - He Meng
- Correspondence: ; Tel.: +86-021-34206146
| |
Collapse
|
12
|
Bahrami S, Amiri-Yekta A, Daneshipour A, Jazayeri SH, Mozdziak PE, Sanati MH, Gourabi H. Designing A Transgenic Chicken: Applying New Approaches toward A Promising Bioreactor. CELL JOURNAL 2019; 22:133-139. [PMID: 31721526 PMCID: PMC6874784 DOI: 10.22074/cellj.2020.6738] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/19/2019] [Accepted: 04/15/2019] [Indexed: 12/26/2022]
Abstract
Specific developmental characteristics of the chicken make it an attractive model for the generation of transgenic organisms. Chicken possess a strong potential for recombinant protein production and can be used as a powerful bioreactor to produce pharmaceutical and nutritional proteins. Several transgenic chickens have been generated during the last two decades via viral and non-viral transfection. Culturing chicken primordial germ cells (PGCs) and their ability for germline transmission ushered in a new stage in this regard. With the advent of CRISPR/Cas9 system, a new phase of studies for manipulating genomes has begun. It is feasible to integrate a desired gene in a predetermined position of the genome using CRISPR/Cas9 system. In this review, we discuss the new approaches and technologies that can be applied to generate a transgenic chicken with regards to recombinant protein productions.
Collapse
Affiliation(s)
- Salahadin Bahrami
- Department of Genetics, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Amir Amiri-Yekta
- Department of Genetics, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Abbas Daneshipour
- Department of Genetics, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Seyedeh Hoda Jazayeri
- Department of Genetics, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | | | - Mohammad Hossein Sanati
- Department of Genetics, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran.Electronic Address: .,Department of Medical Genetics, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Hamid Gourabi
- Department of Genetics, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran. Electronic Address:
| |
Collapse
|
13
|
Xie T, Feng M, Dai M, Mo G, Ruan Z, Wang G, Shi M, Zhang X. Cholesterol-25-hydroxylase Is a Chicken ISG That Restricts ALV-J Infection by Producing 25-hydroxycholesterol. Viruses 2019; 11:E498. [PMID: 31151272 PMCID: PMC6631237 DOI: 10.3390/v11060498] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 05/20/2019] [Accepted: 05/21/2019] [Indexed: 12/29/2022] Open
Abstract
The avian leukosis virus subgroup J (ALV-J) belongs to the chicken retrovirus that causes enormous economic losses in the poultry industry. Interferon-stimulated genes (ISGs) are critical for controlling virus infections. Here, we identified 897 type I ISGs induced by interferon-α (IFN-α) in chicken peripheral blood mononuclear cell (PBMC) by RNA-Seq. In addition, we further identified 152 potential anti-ALV-J chicken type I ISGs. Among these potential anti-ALV-J ISGs, chicken cholesterol 25-hydroxylase (chCH25H) was selected for further antiviral mechanism studies in chicken embryo fibroblast cell lines (DF1). The gene chCH25H is located on chromosome 6 and clustered in a distinct group with mammals CH25H in the phylogenetic tree. The core promoter region of chCH25H was located within -75/-1 sequence. We found that chCH25H was induced by chicken IFN-α and ALV-J in DF1 cells. The overexpression of chCH25H significantly inhibited ALV-J replication in DF1 cells at 48 h post infection (hpi). In addition, ALV-J replication was significantly enhanced in the chCH25H- knockout DF1 cells. Furthermore, we demonstrated that chCH25H restricted ALV-J infection through the production of 25-hydroxycholesterol (25HC), rather than type I and II interferon. Our results identified 152 potential anti-ALV-J chicken type I ISGs and revealed that 25HC, the product of chCH25H, could be used as a natural antiviral agent to control ALV-J infection.
Collapse
Affiliation(s)
- Tingting Xie
- Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China.
- Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou 510642, China.
| | - Min Feng
- Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China.
| | - Manman Dai
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China.
| | - Guodong Mo
- Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China.
- Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou 510642, China.
| | - Zhuohao Ruan
- Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China.
- Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou 510642, China.
| | - Guiyan Wang
- Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China.
- Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou 510642, China.
| | - Meiqing Shi
- Division of Immunology, Virginia-Maryland Regional College of Veterinary Medicine, University of Maryland, College Park, MA 20742, USA.
| | - Xiquan Zhang
- Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China.
- Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou 510642, China.
| |
Collapse
|
14
|
Cheng Y, Lun M, Liu Y, Wang H, Yan Y, Sun J. CRISPR/Cas9-Mediated Chicken TBK1 Gene Knockout and Its Essential Role in STING-Mediated IFN-β Induction in Chicken Cells. Front Immunol 2019; 9:3010. [PMID: 30662438 PMCID: PMC6328437 DOI: 10.3389/fimmu.2018.03010] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 12/05/2018] [Indexed: 12/19/2022] Open
Abstract
TANK-binding kinase 1 (TBK1) is involved in innate immunity, prompting transcriptional induction of type I interferons in response to pathogenic infection. Many studies have focused on mammals but the function of TBK1 in chickens remains poorly defined. CRISPR/Cas9 system has made gene-knockout easy to accomplish. Although CRISPR/Cas9 has been used in chicken cells, low mutation efficiency limits its wide application in chickens. In this study, an effective gene-knockout system was developed based on the CRISPR/Cas9 system in chicken embryonic fibroblast DF-1. Two CRISPR/Cas9 plasmids were constructed, TBK1-g1 and TBK1-g2, which express gRNAs targeting different sequences of the chicken TBK1 gene. After transfection and enrichment with puromycin screening, the mutation rates as assessed via T7E1 assay were 88.05 and 89.55%, respectively, and subsequent sequence analysis showed mutation efficiencies of 86.67 and 93.33%. With the limiting-dilution method, a chTBK1 gene-deficiency monoclonal cell line was obtained and was named DF-1-TBK1-C3. The DF-1-TBK1-C3 cells exhibited normal morphology and maintained stable proliferation ability compared to wild-type cells. The gene-overexpression system and luciferase reporter assay showed that IFN-β induction induced by chSTING was almost completely blocked in DF-1-TBK1-C3 cells. With quantitative real-time PCR, we further confirmed the essential role of chTBK1 in the chSTING-mediated IFN-β induction. At last, the study demonstrated that the chTBK1 knockout system is also applicable in primary chick embryo fibroblasts (CEFs). In this study, an effective gene-knockout system was applied in chickens, a TBK1 gene-deleted DF-1 cell line was successfully created using this system, and with the chTBK1 knockout cells, chTBK1 was revealed to be indispensable in STING-mediated IFN-β activation in chicken cells.
Collapse
Affiliation(s)
| | | | | | | | | | - Jianhe Sun
- School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai Key Laboratory of Veterinary Biotechnology, Key Laboratory of Urban Agriculture (South), Ministry of Agriculture, Shanghai, China
| |
Collapse
|
15
|
Shen X, Niu C, Guo J, Xia M, Xia J, Hu Y, Zheng Y. Stra8 may inhibit apoptosis during mouse spermatogenesis via the AKT signaling pathway. Int J Mol Med 2018; 42:2819-2830. [PMID: 30106128 DOI: 10.3892/ijmm.2018.3825] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 08/02/2018] [Indexed: 11/06/2022] Open
Abstract
Stimulated by retinoic acid 8 (Stra8), one of genes induced by retinoic acid (RA), is required for the meiotic initiation of male spermatogenesis. The present study found that Stra8 inhibited apoptosis in male Stra8‑knockout mice, and in mice with vitamin A deficiency and vitamin A recovery in vivo. This phenotype was also verified in GC1 spermatogonia (spg) cells overexpressing Stra8. In addition, microarray analysis identified that there were nine differentially expressed genes (DEGs) in the Stra8‑overexpressed GC1 spg cells compared with the control groups; the expression of these nine genes was verified via mRNA expression levels. The DEGs were as follows: Phosphatidylinositol‑dependent kinase 1 (PDK1), a key gene upstream of protein kinase B (AKT); angiopoietin 2, a B‑cell lymphoma 2 (Bcl‑2)‑inhibited gene; transcription factor 4, glutathione S‑transferase P91 and ubiquitin‑specific protease 33, mitogen‑activated protein kinase (MAPK)‑related genes; oxidative stress induced growth inhibitor 1, related to the P53 pathway; Bcl‑2, P53, ERK (MAPK1/3), c‑Jun N‑terminal kinase (MAPK8/9), and P38 (MAPK14), all of which are key genes involved in the AKT signaling pathway. Therefore, the present study further verified these genes and found that the mRNA and protein expression levels of PDK1, AKT, Bcl‑2 and ERK were increased. Although the mRNA expression level of P53 was decreased, there was no significant difference in the protein expression level in Stra8‑overexpressing GC1 spg cells compared with controls. In addition, Caspase 3, one of the executioner caspases, was decreased in Stra8‑overexpressing GC1 spg cells compared with the control groups. Therefore, it was suggested that Stra8 may directly or indirectly inhibit caspases through the AKT signaling pathway and ultimately exert an anti‑apoptotic effect in the male reproductive system.
Collapse
Affiliation(s)
- Xueyi Shen
- Department of Histology and Embryology, School of Medicine, Yangzhou University, Yangzhou, Jiangsu 225001, P.R. China
| | - Changmin Niu
- Department of Histology and Embryology, School of Medicine, Yangzhou University, Yangzhou, Jiangsu 225001, P.R. China
| | - Jiaqian Guo
- Department of Histology and Embryology, School of Medicine, Yangzhou University, Yangzhou, Jiangsu 225001, P.R. China
| | - Mengmeng Xia
- Department of Histology and Embryology, School of Medicine, Yangzhou University, Yangzhou, Jiangsu 225001, P.R. China
| | - Jing Xia
- Department of Histology and Embryology, School of Medicine, Yangzhou University, Yangzhou, Jiangsu 225001, P.R. China
| | - Yanqiu Hu
- Center of Reproductive Medicine, Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu 225001, P.R. China
| | - Ying Zheng
- Department of Histology and Embryology, School of Medicine, Yangzhou University, Yangzhou, Jiangsu 225001, P.R. China
| |
Collapse
|
16
|
Roy B, Zhao J, Yang C, Luo W, Xiong T, Li Y, Fang X, Gao G, Singh CO, Madsen L, Zhou Y, Kristiansen K. CRISPR/Cascade 9-Mediated Genome Editing-Challenges and Opportunities. Front Genet 2018; 9:240. [PMID: 30026755 PMCID: PMC6042012 DOI: 10.3389/fgene.2018.00240] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 06/18/2018] [Indexed: 12/26/2022] Open
Abstract
Clustered Regularly Interspaced Palindromic Repeats (CRISPR) and Cascade 9 (also known as Cas9, CRISPR associated protein 9) confer protection against invading viruses or plasmids. The CRISPR/Cascade 9 system constitutes one of the most powerful genome technologies available to researchers today. So far, this technology has enabled efficient genome editing and modification in several model organisms and has successfully been used in biomedicine and biomedical engineering. However, challenges for efficient and safe genetic manipulation in several organisms persist. Here, we review functional approaches and future challenges associated with the use of the CRISPR/Cascade 9 genome editing system and discuss opportunities, ethical issues and future directions within this field.
Collapse
Affiliation(s)
| | - Jing Zhao
- BGI Genomics, BGI-Shenzhen, Shenzhen, China.,Laboratory of Genomics and Molecular Biomedicine, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Chao Yang
- BGI Genomics, BGI-Shenzhen, Shenzhen, China
| | - Wen Luo
- BGI Genomics, BGI-Shenzhen, Shenzhen, China
| | - Teng Xiong
- BGI Genomics, BGI-Shenzhen, Shenzhen, China
| | - Yong Li
- BGI Genomics, BGI-Shenzhen, Shenzhen, China
| | | | - Guanjun Gao
- School of Life Sciences, Tsinghua University, Beijing, China
| | - Chabungbam O Singh
- Institute of Sericulture and Apiculture, College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Lise Madsen
- BGI Genomics, BGI-Shenzhen, Shenzhen, China.,Laboratory of Genomics and Molecular Biomedicine, Department of Biology, University of Copenhagen, Copenhagen, Denmark.,Institute of Marine Research, Bergen, Norway
| | - Yong Zhou
- BGI Genomics, BGI-Shenzhen, Shenzhen, China
| | - Karsten Kristiansen
- BGI Genomics, BGI-Shenzhen, Shenzhen, China.,Laboratory of Genomics and Molecular Biomedicine, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
17
|
Zhang Y, Tang N, Sadigh Y, Baigent S, Shen Z, Nair V, Yao Y. Application of CRISPR/Cas9 Gene Editing System on MDV-1 Genome for the Study of Gene Function. Viruses 2018; 10:v10060279. [PMID: 29794970 PMCID: PMC6024840 DOI: 10.3390/v10060279] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 05/17/2018] [Accepted: 05/22/2018] [Indexed: 12/14/2022] Open
Abstract
Marek’s disease virus (MDV) is a member of alphaherpesviruses associated with Marek’s disease, a highly contagious neoplastic disease in chickens. Complete sequencing of the viral genome and recombineering techniques using infectious bacterial artificial chromosome (BAC) clones of Marek’s disease virus genome have identified major genes that are associated with pathogenicity. Recent advances in CRISPR/Cas9-based gene editing have given opportunities for precise editing of the viral genome for identifying pathogenic determinants. Here we describe the application of CRISPR/Cas9 gene editing approaches to delete the Meq and pp38 genes from the CVI988 vaccine strain of MDV. This powerful technology will speed up the MDV gene function studies significantly, leading to a better understanding of the molecular mechanisms of MDV pathogenesis.
Collapse
Affiliation(s)
- Yaoyao Zhang
- The Pirbright Institute & UK-China Centre of Excellence for Research on Avian Diseases, Pirbright, Ash Road, Guildford, Surrey GU24 0NF, UK.
| | - Na Tang
- The Pirbright Institute & UK-China Centre of Excellence for Research on Avian Diseases, Pirbright, Ash Road, Guildford, Surrey GU24 0NF, UK.
- Binzhou Animal Science and Veterinary Medicine Academy & UK-China Centre of Excellence for Research on Avian Diseases, Binzhou 256600, Shandong, China.
| | - Yashar Sadigh
- The Pirbright Institute & UK-China Centre of Excellence for Research on Avian Diseases, Pirbright, Ash Road, Guildford, Surrey GU24 0NF, UK.
| | - Susan Baigent
- The Pirbright Institute & UK-China Centre of Excellence for Research on Avian Diseases, Pirbright, Ash Road, Guildford, Surrey GU24 0NF, UK.
| | - Zhiqiang Shen
- Binzhou Animal Science and Veterinary Medicine Academy & UK-China Centre of Excellence for Research on Avian Diseases, Binzhou 256600, Shandong, China.
| | - Venugopal Nair
- The Pirbright Institute & UK-China Centre of Excellence for Research on Avian Diseases, Pirbright, Ash Road, Guildford, Surrey GU24 0NF, UK.
| | - Yongxiu Yao
- The Pirbright Institute & UK-China Centre of Excellence for Research on Avian Diseases, Pirbright, Ash Road, Guildford, Surrey GU24 0NF, UK.
| |
Collapse
|
18
|
Antonova E, Glazova O, Gaponova A, Eremyan A, Zvereva S, Grebenkina N, Volkova N, Volchkov P. Successful CRISPR/Cas9 mediated homologous recombination in a chicken cell line. F1000Res 2018; 7:238. [PMID: 29946437 PMCID: PMC6008848 DOI: 10.12688/f1000research.13457.2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/23/2018] [Indexed: 02/02/2023] Open
Abstract
Background: CRISPR/Cas9 system is becoming the dominant genome editing tool in a variety of organisms. CRISPR/Cas9 mediated knock out has been demonstrated both in chicken cell lines and in chicken germ cells that served to generate genetically modified birds. However, there is limited data about CRISPR/Cas9 dependent homology directed repair (HDR) for avian, even in cell culture. Few attempts have been made with integrations in safe harbor loci of chicken genome that induces constitutive expression of the inserted gene. Gene expression under an endogenous promoter would be more valuable than under a constitutive exogenous promoter, as it allows the gene expression to be tissue-specific. Methods: Three gRNAs were chosen to target chicken 3'-untranslated region of GAPDH gene. Cas9-mediated activity in the targeted locus for the gRNAs in DF-1 cells was estimated by T7E1 assay. To edit the locus, the HDR cassette was added along with CRISPR/Cas9. The inserted sequence contained eGFP in frame with a GAPDH coding sequence via P2A and Neomycin resistance gene ( neoR) under cytomegalovirus promoter. Correct integration of the cassette was confirmed with fluorescent microscopy, PCR analysis and sequencing. Enrichment of modified cells was done by G418 selection. Efficiency of integration was assessed with fluorescence activated cell sorting (FACS). Results: We have established a CRISPR/Cas9 system to target an endogenous locus and precisely insert a gene under endogenous control. In our system, we used positive and negative selection to enrich modified cells and remove cells with undesirable insertions. The efficiency of CRISPR/Cas9-mediated HDR was increased up to 90% via G418 enrichment. We have successfully inserted eGFP under control of the chicken GAPDH promoter. Conclusions: The approach can be used further to insert genes of interest under control of tissue-specific promoters in primordial germ cells in order to produce genetically modified birds with useful for biotechnological purposes features.
Collapse
Affiliation(s)
- Ekaterina Antonova
- Moscow Institute of Physics and Technology, Dolgoprudny, Moscow Region, 141701, Russian Federation
| | - Olga Glazova
- Moscow Institute of Physics and Technology, Dolgoprudny, Moscow Region, 141701, Russian Federation
| | - Anna Gaponova
- Moscow Institute of Physics and Technology, Dolgoprudny, Moscow Region, 141701, Russian Federation
| | - Aykaz Eremyan
- Moscow Institute of Physics and Technology, Dolgoprudny, Moscow Region, 141701, Russian Federation
| | - Svetlana Zvereva
- Moscow Institute of Physics and Technology, Dolgoprudny, Moscow Region, 141701, Russian Federation
| | - Natalya Grebenkina
- Moscow Institute of Physics and Technology, Dolgoprudny, Moscow Region, 141701, Russian Federation
| | - Natalya Volkova
- Ernst Institute of Animal Husbandry, Podolsk Municipal District, Moscow Region, 142132 , Russian Federation
| | - Pavel Volchkov
- Moscow Institute of Physics and Technology, Dolgoprudny, Moscow Region, 141701, Russian Federation
| |
Collapse
|
19
|
Tang N, Zhang Y, Pedrera M, Chang P, Baigent S, Moffat K, Shen Z, Nair V, Yao Y. A simple and rapid approach to develop recombinant avian herpesvirus vectored vaccines using CRISPR/Cas9 system. Vaccine 2018; 36:716-722. [PMID: 29269155 PMCID: PMC5783714 DOI: 10.1016/j.vaccine.2017.12.025] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 11/27/2017] [Accepted: 12/11/2017] [Indexed: 12/13/2022]
Abstract
Herpesvirus of turkeys (HVT) has been successfully used as live vaccine against Marek's disease (MD) worldwide for more than 40 years either alone or in combination with other serotypes. HVT is also widely used as a vector platform for generation of recombinant vaccines against a number of avian diseases such as infectious bursal disease (IBD), Newcastle disease (ND) and avian influenza (AI) using conventional recombination methods or recombineering tools on cloned viral genomes. In the present study, we describe the application of CRISPR/Cas9-based genome editing as a rapid and efficient method of generating HVT recombinants expressing VP2 protein of IBDV. This approach offers an efficient method to introduce other viral antigens into the HVT genome for rapid development of recombinant vaccines.
Collapse
Affiliation(s)
- Na Tang
- The Pirbright Institute & UK-China Centre of Excellence for Research on Avian Diseases, Pirbright, Ash Road, Guildford, Surrey GU24 0NF, United Kingdom; Binzhou Animal Science and Veterinary Medicine Academy & UK-China Centre of Excellence for Research on Avian Diseases, Binzhou 256600, Shandong, PR China
| | - Yaoyao Zhang
- The Pirbright Institute & UK-China Centre of Excellence for Research on Avian Diseases, Pirbright, Ash Road, Guildford, Surrey GU24 0NF, United Kingdom
| | - Miriam Pedrera
- The Pirbright Institute & UK-China Centre of Excellence for Research on Avian Diseases, Pirbright, Ash Road, Guildford, Surrey GU24 0NF, United Kingdom
| | - Pengxiang Chang
- The Pirbright Institute & UK-China Centre of Excellence for Research on Avian Diseases, Pirbright, Ash Road, Guildford, Surrey GU24 0NF, United Kingdom
| | - Susan Baigent
- The Pirbright Institute & UK-China Centre of Excellence for Research on Avian Diseases, Pirbright, Ash Road, Guildford, Surrey GU24 0NF, United Kingdom
| | - Katy Moffat
- The Pirbright Institute & UK-China Centre of Excellence for Research on Avian Diseases, Pirbright, Ash Road, Guildford, Surrey GU24 0NF, United Kingdom
| | - Zhiqiang Shen
- Binzhou Animal Science and Veterinary Medicine Academy & UK-China Centre of Excellence for Research on Avian Diseases, Binzhou 256600, Shandong, PR China
| | - Venugopal Nair
- The Pirbright Institute & UK-China Centre of Excellence for Research on Avian Diseases, Pirbright, Ash Road, Guildford, Surrey GU24 0NF, United Kingdom.
| | - Yongxiu Yao
- The Pirbright Institute & UK-China Centre of Excellence for Research on Avian Diseases, Pirbright, Ash Road, Guildford, Surrey GU24 0NF, United Kingdom.
| |
Collapse
|
20
|
Lee HJ, Kim YM, Ono T, Han JY. Genome Modification Technologies and Their Applications in Avian Species. Int J Mol Sci 2017; 18:ijms18112245. [PMID: 29072628 PMCID: PMC5713215 DOI: 10.3390/ijms18112245] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Revised: 10/22/2017] [Accepted: 10/23/2017] [Indexed: 12/01/2022] Open
Abstract
The rapid development of genome modification technology has provided many great benefits in diverse areas of research and industry. Genome modification technologies have also been actively used in a variety of research areas and fields of industry in avian species. Transgenic technologies such as lentiviral systems and piggyBac transposition have been used to produce transgenic birds for diverse purposes. In recent years, newly developed programmable genome editing tools such as transcription activator-like effector nuclease (TALEN) and clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 (CRISPR/Cas9) have also been successfully adopted in avian systems with primordial germ cell (PGC)-mediated genome modification. These genome modification technologies are expected to be applied to practical uses beyond system development itself. The technologies could be used to enhance economic traits in poultry such as acquiring a disease resistance or producing functional proteins in eggs. Furthermore, novel avian models of human diseases or embryonic development could also be established for research purposes. In this review, we discuss diverse genome modification technologies used in avian species, and future applications of avian biotechnology.
Collapse
Affiliation(s)
- Hong Jo Lee
- Department of Agricultural Biotechnology, College of Agriculture and Life Sciences, and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul 08826, Korea.
| | - Young Min Kim
- Department of Agricultural Biotechnology, College of Agriculture and Life Sciences, and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul 08826, Korea.
| | - Tamao Ono
- Faculty of Agriculture, Shinshu University, 8304 Minamiminowa, Kamiina, Nagano 399-4598, Japan.
| | - Jae Yong Han
- Department of Agricultural Biotechnology, College of Agriculture and Life Sciences, and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul 08826, Korea.
- Institute for Biomedical Sciences, Shinshu University, 8304 Minamiminowa, Kamiina, Nagano 399-4598, Japan.
| |
Collapse
|