1
|
Tara A, Singh P, Gautam D, Tripathi G, Uppal C, Malhotra S, De S, Singh MK, Telugu BP, Selokar NL. CRISPR-mediated editing of β-lactoglobulin (BLG) gene in buffalo. Sci Rep 2024; 14:14822. [PMID: 38937564 PMCID: PMC11211398 DOI: 10.1038/s41598-024-65359-9] [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: 02/20/2024] [Accepted: 06/19/2024] [Indexed: 06/29/2024] Open
Abstract
Milk is a good source of nutrition but is also a source of allergenic proteins such as α-lactalbumin, β-lactoglobulin (BLG), casein, and immunoglobulins. The Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)/Cas technology has the potential to edit any gene, including milk allergens. Previously, CRISPR/Cas has been successfully employed in dairy cows and goats, but buffaloes remain unexplored for any milk trait. In this study, we utilized the CRISPR/Cas9 system to edit the major milk allergen BLG gene in buffaloes. First, the editing efficiency of designed sgRNAs was tested in fibroblast cells using the T7E assay and Sanger sequencing. The most effective sgRNA was selected to generate clonal lines of BLG-edited cells. Analysis of 15 single-cell clones, through TA cloning and Sanger sequencing, revealed that 7 clones exhibited bi-allelic (-/-) heterozygous, bi-allelic (-/-) homozygous, and mono-allelic (-/+) disruptions in BLG. Bioinformatics prediction analysis confirmed that non-multiple-of-3 edited nucleotide cell clones have frame shifts and early truncation of BLG protein, while multiple-of-3 edited nucleotides resulted in slightly disoriented protein structures. Somatic cell nuclear transfer (SCNT) method was used to produce blastocyst-stage embryos that have similar developmental rates and quality with wild-type embryos. This study demonstrated the successful bi-allelic editing (-/-) of BLG in buffalo cells through CRISPR/Cas, followed by the production of BLG-edited blastocyst stage embryos using SCNT. With CRISPR and SCNT methods described herein, our long-term goal is to generate gene-edited buffaloes with BLG-free milk.
Collapse
Affiliation(s)
- Aseem Tara
- Animal Biotechnology Division (ABTD), ICAR-National Dairy Research Institute, Karnal, Haryana, 132001, India
| | - Priyanka Singh
- Animal Biotechnology Division (ABTD), ICAR-National Dairy Research Institute, Karnal, Haryana, 132001, India
| | - Devika Gautam
- Animal Biotechnology Division (ABTD), ICAR-National Dairy Research Institute, Karnal, Haryana, 132001, India
| | - Gaurav Tripathi
- Animal Biotechnology Division (ABTD), ICAR-National Dairy Research Institute, Karnal, Haryana, 132001, India
| | - Chirag Uppal
- Animal Biotechnology Division (ABTD), ICAR-National Dairy Research Institute, Karnal, Haryana, 132001, India
| | - Shreya Malhotra
- Animal Biotechnology Division (ABTD), ICAR-National Dairy Research Institute, Karnal, Haryana, 132001, India
| | - Sacchinandan De
- Animal Biotechnology Division (ABTD), ICAR-National Dairy Research Institute, Karnal, Haryana, 132001, India
| | - Manoj K Singh
- Animal Biotechnology Division (ABTD), ICAR-National Dairy Research Institute, Karnal, Haryana, 132001, India
| | - Bhanu P Telugu
- Division of Animal Science, University of Missouri, Columbia, MO, 65211, USA
| | - Naresh L Selokar
- Animal Biotechnology Division (ABTD), ICAR-National Dairy Research Institute, Karnal, Haryana, 132001, India.
| |
Collapse
|
2
|
Zhang Y, Shi Q, Wang P, Huang C, Tang S, Zhou M, Hu Q, Wu L, Liang D. iPSC-derived NK cells with site-specific integration of CAR19 and IL24 at the multi-copy rDNA locus enhanced antitumor activity and proliferation. MedComm (Beijing) 2024; 5:e553. [PMID: 38737469 PMCID: PMC11082533 DOI: 10.1002/mco2.553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Revised: 02/22/2024] [Accepted: 04/01/2024] [Indexed: 05/14/2024] Open
Abstract
The generation of chimeric antigen receptor-modified natural killer (CAR-NK) cells using induced pluripotent stem cells (iPSCs) has emerged as one of the paradigms for manufacturing off-the-shelf universal immunotherapy. However, there are still some challenges in enhancing the potency, safety, and multiple actions of CAR-NK cells. Here, iPSCs were site-specifically integrated at the ribosomal DNA (rDNA) locus with interleukin 24 (IL24) and CD19-specific chimeric antigen receptor (CAR19), and successfully differentiated into iPSC-derived NK (iNK) cells, followed by expansion using magnetic beads in vitro. Compared with the CAR19-iNK cells, IL24 armored CAR19-iNK (CAR19-IL24-iNK) cells showed higher cytotoxic capacity and amplification ability in vitro and inhibited tumor progression more effectively with better survival in a B-cell acute lymphoblastic leukaemia (B-ALL) (Nalm-6 (Luc1))-bearing mouse model. Interestingly, RNA-sequencing analysis showed that IL24 may enhance iNK cell function through nuclear factor kappa B (NFκB) pathway-related genes while exerting a direct effect on tumor cells. This study proved the feasibility and potential of combining IL24 with CAR-iNK cell therapy, suggesting a novel and promising off-the-shelf immunotherapy strategy.
Collapse
Affiliation(s)
- Yuxuan Zhang
- Center for Medical Genetics & Hunan Key Laboratory of Medical GeneticsSchool of Life SciencesCentral South UniversityChangshaChina
| | - Qingxin Shi
- Center for Medical Genetics & Hunan Key Laboratory of Medical GeneticsSchool of Life SciencesCentral South UniversityChangshaChina
| | - Peiyun Wang
- Center for Medical Genetics & Hunan Key Laboratory of Medical GeneticsSchool of Life SciencesCentral South UniversityChangshaChina
| | - Chujun Huang
- Center for Medical Genetics & Hunan Key Laboratory of Medical GeneticsSchool of Life SciencesCentral South UniversityChangshaChina
| | - Shuqing Tang
- Center for Medical Genetics & Hunan Key Laboratory of Medical GeneticsSchool of Life SciencesCentral South UniversityChangshaChina
| | - Miaojin Zhou
- Center for Medical Genetics & Hunan Key Laboratory of Medical GeneticsSchool of Life SciencesCentral South UniversityChangshaChina
| | - Qian Hu
- Center for Medical Genetics & Hunan Key Laboratory of Medical GeneticsSchool of Life SciencesCentral South UniversityChangshaChina
| | - Lingqian Wu
- Center for Medical Genetics & Hunan Key Laboratory of Medical GeneticsSchool of Life SciencesCentral South UniversityChangshaChina
| | - Desheng Liang
- Center for Medical Genetics & Hunan Key Laboratory of Medical GeneticsSchool of Life SciencesCentral South UniversityChangshaChina
- Hunan Key Laboratory of Animal Models for Human DiseasesSchool of Life SciencesCentral South UniversityChangshaChina
| |
Collapse
|
3
|
Fauser F, Kadam BN, Arangundy-Franklin S, Davis JE, Vaidya V, Schmidt NJ, Lew G, Xia DF, Mureli R, Ng C, Zhou Y, Scarlott NA, Eshleman J, Bendaña YR, Shivak DA, Reik A, Li P, Davis GD, Miller JC. Compact zinc finger architecture utilizing toxin-derived cytidine deaminases for highly efficient base editing in human cells. Nat Commun 2024; 15:1181. [PMID: 38360922 PMCID: PMC10869815 DOI: 10.1038/s41467-024-45100-w] [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: 06/14/2023] [Accepted: 01/15/2024] [Indexed: 02/17/2024] Open
Abstract
Nucleobase editors represent an emerging technology that enables precise single-base edits to the genomes of eukaryotic cells. Most nucleobase editors use deaminase domains that act upon single-stranded DNA and require RNA-guided proteins such as Cas9 to unwind the DNA prior to editing. However, the most recent class of base editors utilizes a deaminase domain, DddAtox, that can act upon double-stranded DNA. Here, we target DddAtox fragments and a FokI-based nickase to the human CIITA gene by fusing these domains to arrays of engineered zinc fingers (ZFs). We also identify a broad variety of Toxin-Derived Deaminases (TDDs) orthologous to DddAtox that allow us to fine-tune properties such as targeting density and specificity. TDD-derived ZF base editors enable up to 73% base editing in T cells with good cell viability and favorable specificity.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Garrett Lew
- Sangamo Therapeutics, Inc., Brisbane, CA, USA
| | - Danny F Xia
- Sangamo Therapeutics, Inc., Brisbane, CA, USA
| | | | - Colman Ng
- Sangamo Therapeutics, Inc., Brisbane, CA, USA
| | | | | | | | | | | | | | - Patrick Li
- Sangamo Therapeutics, Inc., Brisbane, CA, USA
| | | | | |
Collapse
|
4
|
Yamashita MS, Melo EO. Animal Transgenesis and Cloning: Combined Development and Future Perspectives. Methods Mol Biol 2023; 2647:121-149. [PMID: 37041332 DOI: 10.1007/978-1-0716-3064-8_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/13/2023]
Abstract
The revolution in animal transgenesis began in 1981 and continues to become more efficient, cheaper, and faster to perform. New genome editing technologies, especially CRISPR-Cas9, are leading to a new era of genetically modified or edited organisms. Some researchers advocate this new era as the time of synthetic biology or re-engineering. Nonetheless, we are witnessing advances in high-throughput sequencing, artificial DNA synthesis, and design of artificial genomes at a fast pace. These advances in symbiosis with animal cloning by somatic cell nuclear transfer (SCNT) allow the development of improved livestock, animal models of human disease, and heterologous production of bioproducts for medical applications. In the context of genetic engineering, SCNT remains a useful technology to generate animals from genetically modified cells. This chapter addresses these fast-developing technologies driving this biotechnological revolution and their association with animal cloning technology.
Collapse
Affiliation(s)
- Melissa S Yamashita
- Embrapa Genetic Resources and Biotechnology, Brasília, Distrito Federal, Brazil
- Graduation Program in Animal Biology, University of Brasília, Brasília, Distrito Federal, Brazil
| | - Eduardo O Melo
- Embrapa Genetic Resources and Biotechnology, Brasília, Distrito Federal, Brazil.
- Graduation Program in Biotechnology, University of Tocantins, Gurupi, Tocantins, Brazil.
| |
Collapse
|
5
|
Use of Genome Editing Techniques to Produce Transgenic Farm Animals. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1354:279-297. [PMID: 34807447 PMCID: PMC9810480 DOI: 10.1007/978-3-030-85686-1_14] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Recombinant proteins are essential for the treatment and diagnosis of clinical human ailments. The availability and biological activity of recombinant proteins is heavily influenced by production platforms. Conventional production platforms such as yeast, bacteria, and mammalian cells have biological and economical challenges. Transgenic livestock species have been explored as an alternative production platform for recombinant proteins, predominantly through milk secretion; the strategy has been demonstrated to produce large quantities of biologically active proteins. The major limitation of utilizing livestock species as bioreactors has been efforts required to alter the genome of livestock. Advancements in the genome editing field have drastically improved the ability to genetically engineer livestock species. Specifically, genome editing tools such as the CRISPR/Cas9 system have lowered efforts required to generate genetically engineered livestock, thus minimizing restrictions on the type of genetic modification in livestock. In this review, we discuss characteristics of transgenic animal bioreactors and how the use of genome editing systems enhances design and availability of the animal models.
Collapse
|
6
|
Gutierrez-Reinoso MA, Aponte PM, Garcia-Herreros M. Genomic Analysis, Progress and Future Perspectives in Dairy Cattle Selection: A Review. Animals (Basel) 2021; 11:599. [PMID: 33668747 PMCID: PMC7996307 DOI: 10.3390/ani11030599] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 11/23/2020] [Accepted: 11/24/2020] [Indexed: 12/16/2022] Open
Abstract
Genomics comprises a set of current and valuable technologies implemented as selection tools in dairy cattle commercial breeding programs. The intensive progeny testing for production and reproductive traits based on genomic breeding values (GEBVs) has been crucial to increasing dairy cattle productivity. The knowledge of key genes and haplotypes, including their regulation mechanisms, as markers for productivity traits, may improve the strategies on the present and future for dairy cattle selection. Genome-wide association studies (GWAS) such as quantitative trait loci (QTL), single nucleotide polymorphisms (SNPs), or single-step genomic best linear unbiased prediction (ssGBLUP) methods have already been included in global dairy programs for the estimation of marker-assisted selection-derived effects. The increase in genetic progress based on genomic predicting accuracy has also contributed to the understanding of genetic effects in dairy cattle offspring. However, the crossing within inbred-lines critically increased homozygosis with accumulated negative effects of inbreeding like a decline in reproductive performance. Thus, inaccurate-biased estimations based on empirical-conventional models of dairy production systems face an increased risk of providing suboptimal results derived from errors in the selection of candidates of high genetic merit-based just on low-heritability phenotypic traits. This extends the generation intervals and increases costs due to the significant reduction of genetic gains. The remarkable progress of genomic prediction increases the accurate selection of superior candidates. The scope of the present review is to summarize and discuss the advances and challenges of genomic tools for dairy cattle selection for optimizing breeding programs and controlling negative inbreeding depression effects on productivity and consequently, achieving economic-effective advances in food production efficiency. Particular attention is given to the potential genomic selection-derived results to facilitate precision management on modern dairy farms, including an overview of novel genome editing methodologies as perspectives toward the future.
Collapse
Affiliation(s)
- Miguel A. Gutierrez-Reinoso
- Facultad de Ciencias Agropecuarias y Recursos Naturales, Carrera de Medicina Veterinaria, Universidad Técnica de Cotopaxi (UTC), Latacunga 05-0150, Ecuador
- Laboratorio de Biotecnología Animal, Departamento de Ciencia Animal, Facultad de Ciencias Veterinarias, Universidad de Concepción (UdeC), Chillán 3780000, Chile
| | - Pedro M. Aponte
- Colegio de Ciencias Biológicas y Ambientales (COCIBA), Universidad San Francisco de Quito (USFQ), Quito 170157, Ecuador
- Campus Cumbayá, Instituto de Investigaciones en Biomedicina “One-health”, Universidad San Francisco de Quito (USFQ), Quito 170157, Ecuador
| | - Manuel Garcia-Herreros
- Instituto Nacional de Investigação Agrária e Veterinária (INIAV), 2005-048 Santarém, Portugal
| |
Collapse
|
7
|
Lee K, Uh K, Farrell K. Current progress of genome editing in livestock. Theriogenology 2020; 150:229-235. [PMID: 32000993 DOI: 10.1016/j.theriogenology.2020.01.036] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 01/18/2020] [Indexed: 12/12/2022]
Abstract
Historically, genetic engineering in livestock proved to be challenging. Without stable embryonic stem cell lines to utilize, somatic cell nuclear transfer (SCNT) had to be employed to produce many of the genetically engineered (GE) livestock models. Through the genetic engineering of somatic cells followed by SCNT, GE livestock models could be generated carrying site-specific modifications. Although successful, only a few GE livestock models were generated because of low efficiency and associated birth defects. Recently, there have been major strides in the development of genome editing tools: Zinc-Finger Nucleases (ZFNs), Transcription activator-like effector nucleases (TALENS), and Clustered regularly interspaced short palindromic repeats (CRISPR) and CRISPR-associated 9 (Cas9) system. These tools rely on the generation of a double strand DNA break, followed by one of two repair pathways: non-homologous end joining (NHEJ) or homology directed repair (HDR). Compared to the traditional approaches, these tools dramatically reduce time and effort needed to establish a GE animal. Another benefit of utilizing genome editing tools is the application of direct injection into developing embryos to induce targeted mutations, therefore, eliminating side effects associated with SCNT. Emerging technological advancements of genome editing systems have dramatically improved efficiency to generate GE livestock models for both biomedical and agricultural purposes. Although the efficiency of genome editing tools has revolutionized GE livestock production, improvements for safe and consistent application are desired. This review will provide an overview of genome editing techniques, as well as examples of GE livestock models for agricultural and biomedical purposes.
Collapse
Affiliation(s)
- Kiho Lee
- Department of Animal and Poultry Sciences, Virginia Tech, Blacksburg, VA, USA.
| | - Kyungjun Uh
- Department of Animal and Poultry Sciences, Virginia Tech, Blacksburg, VA, USA
| | - Kayla Farrell
- Department of Animal and Poultry Sciences, Virginia Tech, Blacksburg, VA, USA
| |
Collapse
|
8
|
Zhao J, Lai L, Ji W, Zhou Q. Genome editing in large animals: current status and future prospects. Natl Sci Rev 2019; 6:402-420. [PMID: 34691891 PMCID: PMC8291540 DOI: 10.1093/nsr/nwz013] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 01/09/2019] [Accepted: 01/30/2019] [Indexed: 12/14/2022] Open
Abstract
Large animals (non-human primates, livestock and dogs) are playing important roles in biomedical research, and large livestock animals serve as important sources of meat and milk. The recently developed programmable DNA nucleases have revolutionized the generation of gene-modified large animals that are used for biological and biomedical research. In this review, we briefly introduce the recent advances in nuclease-meditated gene editing tools, and we outline these editing tools' applications in human disease modeling, regenerative medicine and agriculture. Additionally, we provide perspectives regarding the challenges and prospects of the new genome editing technology.
Collapse
Affiliation(s)
- Jianguo Zhao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing 100049, China
- Institute of Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
| | - Liangxue Lai
- South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Weizhi Ji
- Yunnan Key Laboratory of Primate Biomedicine Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming 650500, China
- CAS Center for Excellence in Brain Science and Intelligence Technology (CEBSIT), Shanghai 200031, China
| | - Qi Zhou
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing 100049, China
- Institute of Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
| |
Collapse
|
9
|
An L, Yang L, Huang Y, Cheng Y, Du F. Generating Goat Mammary Gland Bioreactors for Producing Recombinant Proteins by Gene Targeting. Methods Mol Biol 2019; 1874:391-401. [PMID: 30353527 DOI: 10.1007/978-1-4939-8831-0_23] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Exogenous genes can be site-specifically integrated into the genomic DNA of animals by homologous recombination, generating transgenic animals. These animals have a clear hereditary background, although position effects of the exogenous genes and potential functional disruption of host genes can be caused by the genetic inserts. Therefore, the generation of mammary gland bioreactors via gene-targeting methods is a great asset for producing recombinant proteins in milk. Here, we describe a method of generating gene-targeted goats with the human alpha-lactalbumin gene (hα-LA) integrated into the beta-lactoglobulin gene (BLG) locus. The milk from these goats will be less allergenic and will be enriched with components of human milk protein.
Collapse
Affiliation(s)
- Liyou An
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Lan Yang
- Lannuo Biotechnologies Wuxi Inc., Wuxi, China
| | - Yuejin Huang
- Shanghai Jenomed Biotech Co., Ltd., Shanghai, China
| | - Yong Cheng
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, China
| | - Fuliang Du
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China. .,Renova Life, Inc., College Park, MD, USA.
| |
Collapse
|
10
|
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
|
11
|
Wei J, Wagner S, Maclean P, Brophy B, Cole S, Smolenski G, Carlson DF, Fahrenkrug SC, Wells DN, Laible G. Cattle with a precise, zygote-mediated deletion safely eliminate the major milk allergen beta-lactoglobulin. Sci Rep 2018; 8:7661. [PMID: 29769555 PMCID: PMC5955954 DOI: 10.1038/s41598-018-25654-8] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 04/19/2018] [Indexed: 12/26/2022] Open
Abstract
We applied precise zygote-mediated genome editing to eliminate beta-lactoglobulin (BLG), a major allergen in cows’ milk. To efficiently generate LGB knockout cows, biopsied embryos were screened to transfer only appropriately modified embryos. Transfer of 13 pre-selected embryos into surrogate cows resulted in the birth of three calves, one dying shortly after birth. Deep sequencing results confirmed conversion of the genotype from wild type to the edited nine bp deletion by more than 97% in the two male calves. The third calf, a healthy female, had in addition to the expected nine bp deletion (81%), alleles with an in frame 21 bp deletion (<17%) at the target site. While her milk was free of any mature BLG, we detected low levels of a BLG variant derived from the minor deletion allele. This confirmed that the nine bp deletion genotype completely knocks out production of BLG. In addition, we showed that the LGB knockout animals are free of any TALEN-mediated off-target mutations or vector integration events using an unbiased whole genome analysis. Our study demonstrates the feasibility of generating precisely biallelically edited cattle by zygote-mediated editing for the safe production of hypoallergenic milk.
Collapse
Affiliation(s)
- Jingwei Wei
- AgResearch, Ruakura Research Centre, Hamilton, 3240, New Zealand
| | - Stefan Wagner
- AgResearch, Ruakura Research Centre, Hamilton, 3240, New Zealand.,Rowett Institute, Aberdeen, AB25 2ZD, United Kingdom
| | - Paul Maclean
- AgResearch, Ruakura Research Centre, Hamilton, 3240, New Zealand
| | - Brigid Brophy
- AgResearch, Ruakura Research Centre, Hamilton, 3240, New Zealand
| | - Sally Cole
- AgResearch, Ruakura Research Centre, Hamilton, 3240, New Zealand
| | - Grant Smolenski
- AgResearch, Ruakura Research Centre, Hamilton, 3240, New Zealand.,MS3 Solutions Ltd., Ruakura Research Centre, Hamilton, 3240, New Zealand
| | | | | | - David N Wells
- AgResearch, Ruakura Research Centre, Hamilton, 3240, New Zealand
| | - Götz Laible
- AgResearch, Ruakura Research Centre, Hamilton, 3240, New Zealand.
| |
Collapse
|
12
|
Ikeda M, Matsuyama S, Akagi S, Ohkoshi K, Nakamura S, Minabe S, Kimura K, Hosoe M. Correction of a Disease Mutation using CRISPR/Cas9-assisted Genome Editing in Japanese Black Cattle. Sci Rep 2017; 7:17827. [PMID: 29259316 PMCID: PMC5736618 DOI: 10.1038/s41598-017-17968-w] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Accepted: 12/04/2017] [Indexed: 12/20/2022] Open
Abstract
Isoleucyl-tRNA synthetase (IARS) syndrome is a recessive disease of Japanese Black cattle caused by a single nucleotide substitution. To repair the mutated IARS gene, we designed clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 (Cas9) to create a double-strand break near the mutation site. CRISPR/Cas9 and donor DNA that contained a synonymous codon for the correct amino acid and an Aequorea coerulescens Green Fluorescent Protein (AcGFP) cassette with a piggyBac transposase recognition site at both ends were introduced into bovine fetal fibroblast (BFF) cells isolated from a homozygous mutant calf. Recombinant cells were enriched on the basis of expression of AcGFP, and two cell lines that contained the repaired allele were subcloned. We generated somatic cell nuclear transfer (SCNT) embryos from the repaired cells and transferred 22 blastocysts to recipient cows. In total, five viable fetuses were retrieved at Days 34 and 36. PiggyBac transposase mRNA was introduced into BFF cells isolated from cloned foetuses and AcGFP-negative cells were used for second round of cloning. We transferred nine SCNT embryos to recipient cows and retrieved two fetuses at Day 34. Fetal genomic DNA analysis showed correct repair of the IARS mutation without any additional DNA footprint.
Collapse
Affiliation(s)
- Mitsumi Ikeda
- Institute of Agrobiological Sciences, NARO, Ikenodai 2, Tsukuba, Ibaraki, 305-8602, Japan
| | - Shuichi Matsuyama
- Institute of Livestock and Grassland Science, NARO, Senbonmatsu 768, Nasushiobara, Tochigi, 329-2793, Japan
| | - Satoshi Akagi
- Institute of Livestock and Grassland Science, NARO, Ikenodai 2, Tsukuba, Ibaraki, 305-0901, Japan
| | - Katsuhiro Ohkoshi
- Institute of Agrobiological Sciences, NARO, Ikenodai 2, Tsukuba, Ibaraki, 305-8602, Japan
| | - Sho Nakamura
- Institute of Livestock and Grassland Science, NARO, Senbonmatsu 768, Nasushiobara, Tochigi, 329-2793, Japan
| | - Shiori Minabe
- Institute of Livestock and Grassland Science, NARO, Senbonmatsu 768, Nasushiobara, Tochigi, 329-2793, Japan
| | - Koji Kimura
- Okayama University Graduate School of Environmental and Life Science, Tsushima-Naka 1-1-1, Kita-ku, Okayama, 700-8530, Japan
| | - Misa Hosoe
- Institute of Agrobiological Sciences, NARO, Ikenodai 2, Tsukuba, Ibaraki, 305-8602, Japan.
| |
Collapse
|
13
|
Refining strategies to translate genome editing to the clinic. Nat Med 2017; 23:415-423. [PMID: 28388605 DOI: 10.1038/nm.4313] [Citation(s) in RCA: 182] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Accepted: 02/27/2017] [Indexed: 12/17/2022]
Abstract
Recent progress in developing programmable nucleases, such as zinc-finger nucleases, transcription activator-like effector nucleases (TALENs) and clustered regularly interspaced short palindromic repeat (CRISPR)-Cas nucleases, have paved the way for gene editing to enter clinical practice. This translation is a result of combining high nuclease activity with high specificity and successfully applying this technology in various preclinical disease models, including infectious disease, primary immunodeficiencies, hemoglobinopathies, hemophilia and muscular dystrophy. Several clinical gene-editing trials, both ex vivo and in vivo, have been initiated in the past 2 years, including studies that aim to knockout genes as well as to add therapeutic transgenes. Here we discuss the advances made in the gene-editing field in recent years, and specify priorities that need to be addressed to expand therapeutic genome editing to further disease entities.
Collapse
|
14
|
Abstract
In the past few years, new technologies have arisen that enable higher efficiency of gene editing. With the increase ease of using gene editing technologies, it is important to consider the best method for transferring new genetic material to livestock animals. Microinjection is a technique that has proven to be effective in mice but is less efficient in large livestock animals. Over the years, a variety of methods have been used for cloning as well as gene transfer including; nuclear transfer, sperm mediated gene transfer (SMGT), and liposome-mediated DNA transfer. This review looks at the different success rate of these methods and how they have evolved to become more efficient. As well as gene editing technologies, including Zinc-finger nucleases (ZFNs), transcription activator-like effector nucleases (TALENs), and the most recent clustered regulatory interspaced short palindromic repeats (CRISPRs). Through the advancements in gene-editing technologies, generating transgenic animals is now more accessible and affordable. The goals of producing transgenic animals are to 1) increase our understanding of biology and biomedical science; 2) increase our ability to produce more efficient animals; and 3) produce disease resistant animals. ZFNs, TALENs, and CRISPRs combined with gene transfer methods increase the possibility of achieving these goals.
Collapse
Key Words
- BLG, β-lactoglobulin
- CRISPR
- CRISPRs, clustered regulatory interspaced short palindromic repeats
- EG, embryonic germ
- ES, Embryonic stem
- ESC, Embryonic stem cell
- HDR, homology directed repair
- ICM, inner cell mass
- ICSI, intracytoplasmic sperm injection
- NHEJ, non-homologous end joining
- NT, nuclear transfers
- OBCT, oocyte bisection technique
- PAM, protospacer adjacent motif
- PCR, polymerase chain reaction
- PGCS, primordial germ cells
- RVDs, repeat variable diresidues
- SMGT
- SMGT, sperm mediated gene transfer
- SV40, simian virus 40
- TALEN
- TALENs, transcription activator-like effector nucleases
- ZFN
- ZFN, Zinc-finger nucleases
- gene editing
- gene transfer
- iPSC, induced pluripotent stem cells
- nuclear transfer
- ssODN, single strand oligo nucleotide
Collapse
Affiliation(s)
- Samantha N Lotti
- a Department of Animal Sciences , University of Illinois at Urbana-Champaign , Urbana , Illinois , USA
| | - Kathryn M Polkoff
- a Department of Animal Sciences , University of Illinois at Urbana-Champaign , Urbana , Illinois , USA
| | - Marcello Rubessa
- b Carl R. Woese Institute for Genomic Biology, University of Illinois , Urbana , IL , USA
| | - Matthew B Wheeler
- a Department of Animal Sciences , University of Illinois at Urbana-Champaign , Urbana , Illinois , USA.,b Carl R. Woese Institute for Genomic Biology, University of Illinois , Urbana , IL , USA
| |
Collapse
|
15
|
Vriend LEM, Krawczyk PM. Nick-initiated homologous recombination: Protecting the genome, one strand at a time. DNA Repair (Amst) 2016; 50:1-13. [PMID: 28087249 DOI: 10.1016/j.dnarep.2016.12.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2016] [Accepted: 12/17/2016] [Indexed: 01/13/2023]
Abstract
Homologous recombination (HR) is an essential, widely conserved mechanism that utilizes a template for accurate repair of DNA breaks. Some early HR models, developed over five decades ago, anticipated single-strand breaks (nicks) as initiating lesions. Subsequent studies favored a more double-strand break (DSB)-centered view of HR initiation and at present this pathway is primarily considered to be associated with DSB repair. However, mounting evidence suggests that nicks can indeed initiate HR directly, without first being converted to DSBs. Moreover, recent studies reported on novel branches of nick-initiated HR (nickHR) that rely on single-, rather than double-stranded repair templates and that are characterized by mechanistically and genetically unique properties. The physiological significance of nickHR is not well documented, but its high-fidelity nature and low mutagenic potential are relevant in recently developed, precise gene editing approaches. Here, we review the evidence for stimulation of HR by nicks, as well as the data on the interactions of nickHR with other DNA repair pathways and on its mechanistic properties. We conclude that nickHR is a bona-fide pathway for nick repair, sharing the molecular machinery with the canonical HR but nevertheless characterized by unique properties that secure its inclusion in DNA repair models and warrant future investigations.
Collapse
Affiliation(s)
- Lianne E M Vriend
- Department of Cell Biology and Histology, Academic Medical Center, University of Amsterdam, Meibergdreef 15, 1105 AZ Amsterdam, The Netherlands
| | - Przemek M Krawczyk
- Department of Cell Biology and Histology, Academic Medical Center, University of Amsterdam, Meibergdreef 15, 1105 AZ Amsterdam, The Netherlands.
| |
Collapse
|