1
|
Perisse IV, Fan Z, Singina GN, White KL, Polejaeva IA. Improvements in Gene Editing Technology Boost Its Applications in Livestock. Front Genet 2021; 11:614688. [PMID: 33603767 PMCID: PMC7885404 DOI: 10.3389/fgene.2020.614688] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 12/07/2020] [Indexed: 12/18/2022] Open
Abstract
Accelerated development of novel CRISPR/Cas9-based genome editing techniques provides a feasible approach to introduce a variety of precise modifications in the mammalian genome, including introduction of multiple edits simultaneously, efficient insertion of long DNA sequences into specific targeted loci as well as performing nucleotide transitions and transversions. Thus, the CRISPR/Cas9 tool has become the method of choice for introducing genome alterations in livestock species. The list of new CRISPR/Cas9-based genome editing tools is constantly expanding. Here, we discuss the methods developed to improve efficiency and specificity of gene editing tools as well as approaches that can be employed for gene regulation, base editing, and epigenetic modifications. Additionally, advantages and disadvantages of two primary methods used for the production of gene-edited farm animals: somatic cell nuclear transfer (SCNT or cloning) and zygote manipulations will be discussed. Furthermore, we will review agricultural and biomedical applications of gene editing technology.
Collapse
Affiliation(s)
- Iuri Viotti Perisse
- Department of Animal, Dairy and Veterinary Sciences, Utah State University, Logan, UT, United States
| | - Zhiqiang Fan
- Department of Animal, Dairy and Veterinary Sciences, Utah State University, Logan, UT, United States
| | - Galina N. Singina
- L.K. Ernst Federal Research Center for Animal Husbandry, Podolsk, Russia
| | - Kenneth L. White
- Department of Animal, Dairy and Veterinary Sciences, Utah State University, Logan, UT, United States
| | - Irina A. Polejaeva
- Department of Animal, Dairy and Veterinary Sciences, Utah State University, Logan, UT, United States
| |
Collapse
|
2
|
Mammalian-specific ectodermal enhancers control the expression of Hoxc genes in developing nails and hair follicles. Proc Natl Acad Sci U S A 2020; 117:30509-30519. [PMID: 33199643 PMCID: PMC7720164 DOI: 10.1073/pnas.2011078117] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Vertebrate Hox genes are critical for the establishment of structures during the development of the main body axis. Subsequently, they play important roles either in organizing secondary axial structures such as the appendages, or during homeostasis in postnatal stages and adulthood. Here, we set up to analyze their elusive function in the ectodermal compartment, using the mouse limb bud as a model. We report that the HoxC gene cluster was co-opted to be transcribed in the distal limb ectoderm, where it is activated following the rule of temporal colinearity. These ectodermal cells subsequently produce various keratinized organs such as nails or claws. Accordingly, deletion of the HoxC cluster led to mice lacking nails (anonychia), a condition stronger than the previously reported loss of function of Hoxc13, which is the causative gene of the ectodermal dysplasia 9 (ECTD9) in human patients. We further identified two mammalian-specific ectodermal enhancers located upstream of the HoxC gene cluster, which together regulate Hoxc gene expression in the hair and nail ectodermal organs. Deletion of these regulatory elements alone or in combination revealed a strong quantitative component in the regulation of Hoxc genes in the ectoderm, suggesting that these two enhancers may have evolved along with the mammalian taxon to provide the level of HOXC proteins necessary for the full development of hair and nail.
Collapse
|
3
|
Lee K, Farrell K, Uh K. Application of genome-editing systems to enhance available pig resources for agriculture and biomedicine. Reprod Fertil Dev 2020; 32:40-49. [PMID: 32188556 DOI: 10.1071/rd19273] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Traditionally, genetic engineering in the pig was a challenging task. Genetic engineering of somatic cells followed by somatic cell nuclear transfer (SCNT) could produce genetically engineered (GE) pigs carrying site-specific modifications. However, due to difficulties in engineering the genome of somatic cells and developmental defects associated with SCNT, a limited number of GE pig models were reported. Recent developments in genome-editing tools, such as zinc finger nucleases (ZFNs), transcription activator-like effector nucleases (TALENs) and the clustered regularly interspaced short palindromic repeat (CRISPR)/CRISPR-associated (Cas) 9 system, have markedly changed the effort and time required to produce GE pig models. The frequency of genetic engineering in somatic cells is now practical. In addition, SCNT is no longer essential in producing GE pigs carrying site-specific modifications, because direct injection of genome-editing systems into developing embryos introduces targeted modifications. To date, the CRISPR/Cas9 system is the most convenient, cost-effective, timely and commonly used genome-editing technology. Several applicable biomedical and agricultural pig models have been generated using the CRISPR/Cas9 system. Although the efficiency of genetic engineering has been markedly enhanced with the use of genome-editing systems, improvements are still needed to optimally use the emerging technology. Current and future advances in genome-editing strategies will have a monumental effect on pig models used in agriculture and biomedicine.
Collapse
Affiliation(s)
- Kiho Lee
- Department of Animal and Poultry Sciences, Litton-Reaves Hall, Virginia Tech, Blacksburg, Virgina 24061, USA; and Corresponding author.
| | - Kayla Farrell
- Department of Animal and Poultry Sciences, Litton-Reaves Hall, Virginia Tech, Blacksburg, Virgina 24061, USA
| | - Kyungjun Uh
- Department of Animal and Poultry Sciences, Litton-Reaves Hall, Virginia Tech, Blacksburg, Virgina 24061, USA
| |
Collapse
|
4
|
Dai B, Zhang M, Yuan JL, Ren LQ, Han XY, Liu DJ. Integrative Analysis of Methylation and Transcriptional Profiles to Reveal the Genetic Stability of Cashmere Traits in the Tβ4 Overexpression of Cashmere Goats. Animals (Basel) 2019; 9:ani9121002. [PMID: 31756916 PMCID: PMC6940810 DOI: 10.3390/ani9121002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Revised: 10/27/2019] [Accepted: 11/18/2019] [Indexed: 12/25/2022] Open
Abstract
Simple Summary Cashmere goats have double coats consisting of non-medullated fine inner hairs or cashmere fibers produced by secondary hair follicles (SHFs) and guard hairs produced by primary hair follicles (PHFs). Cashmere is an important economic product worldwide and the world market for cashmere is increasing while the current production of cashmere is limited. Thymosin β4 (Tβ4), a 4.9-kDa protein, contains 43 amino acids. Here, we produced Tβ4 overexpression (Tβ4-OE) offspring using two methods. The somatic cell nuclear transfer (SCNT) goats had increased hair follicle development and higher cashmere yields than wild type (WT) and natural mating (NM) goats. Taken together, our results showed that DNA methylation affected the expression of differentially expressed genes (DEGs) between generations and the genetic stability of cashmere traits. Abstract DNA methylation alteration is frequently observed in exogenous gene silencing and may play important roles in the genetic stability of traits. Cashmere is derived from the secondary hair follicles (SHFs) of cashmere goats, which are morphogenetically distinct from primary hair follicles (PHFs). Here, in light of having initially produced 15 Tβ4 overexpression (Tβ4-OE) cashmere goats which had more SHFs than the wild type (WT) goats, and produced more cashmere, we produced Tβ4-OE offsprings both via somatic cell nuclear transfer (SCNT) and via natural mating (NM). However, the desired trait exhibited lower fixation in the line-bred offspring compared to the SCNT offspring. Integrative analysis of methylation and transcriptional profiles showed that this might be due to the influence of methylation on the expression of differentially expressed genes (DEGs) between generations, which was mutually consistent with the results of the functional and pathway enrichment analysis of differentially methylated regions (DMRs) and DEGs. Overall, our study systematically describes the DNA methylation characteristics between generations of cashmere goats and provides a basis for improving genetic stability.
Collapse
|
5
|
Yin Y, Hao H, Xu X, Shen L, Wu W, Zhang J, Li Q. Generation of an MC3R knock-out pig by CRSPR/Cas9 combined with somatic cell nuclear transfer (SCNT) technology. Lipids Health Dis 2019; 18:122. [PMID: 31138220 PMCID: PMC6540458 DOI: 10.1186/s12944-019-1073-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2018] [Accepted: 05/17/2019] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Melanocortin 3 receptor (MC3R), a rhodopsin-like G protein-coupled receptor, is an important regulator of metabolism. Although MC3R knock-out (KO) mice and rats were generated in earlier studies, the function of MC3R remains elusive. Since pig models have many advantages over rodents in metabolism research, we generated an MC3R-KO pig using a CRSPR/Cas9-based system combined with somatic cell nuclear transfer (SCNT) technology. METHOD Four CRSPR/Cas9 target vectors were constructed and then their cleavage efficiency was tested in porcine fetal fibroblasts (PFFs). The pX330-sgRNA1 and pX330-sgRNA4 vectors were used to co-transfect PFFs to obtain positive colonies. PCR screening and sequencing were conducted to identify the genotype of the colonies. The biallelically modified colonies and wild-type control colonies were used simultaneously as donor cells for SCNT. A total of 1203 reconstructed embryos were transferred into 6 surrogates, of which one became pregnant. The genotypes of the resulting piglets were determined by PCR and sequencing, and off-target effects in the MC3R KO piglets were detected by sequencing. Then, offspring were obtained through breeding and six male KO pigs were used for the growth performance analysis. RESULTS Four vectors were constructed successfully, and their cleavage efficiencies were 27.96, 44.89, 32.72 and 38.86%, respectively. A total of 21 mutant colonies, including 11 MC3R-/- and 10 MC3R+/- clones, were obtained, corresponding to a gene targeting efficiency of 29.17%, with 15.28% biallelic mutations. A total of 6 piglets were born, and only two MC3R KO piglets were generated, one with malformations and a healthy one. No off-target effects were detected by sequencing in the healthy mutant. Six male MC3R KO pigs were obtained in the F2 generation and their body weight and body fat were both increased compared to wild-type full siblings. CONCLUSION A MC3R KO pig strain was generated using the CRSIPR/Cas9-based system, which makes it possible to study the biological function of MC3R in a non-rodent model.
Collapse
Affiliation(s)
- Yajun Yin
- College of Biological, Chemical Science and Engineering, Jiaxing University, Jiaxing, 314001, China
| | - Haiyang Hao
- State Key Laboratory of Agrobiotechnology & College of Biological Sciences, China Agricultural University, Beijing, 100193, China
| | - Xingbin Xu
- College of life science and biotechnology, Hebei Normal University of Science and Technology, Qinhuangdao, 066004, China
| | - Liangcai Shen
- State Key Laboratory of Agrobiotechnology & College of Biological Sciences, China Agricultural University, Beijing, 100193, China
| | - Wenjing Wu
- College of Biological, Chemical Science and Engineering, Jiaxing University, Jiaxing, 314001, China
| | - Jin Zhang
- College of Biological, Chemical Science and Engineering, Jiaxing University, Jiaxing, 314001, China.
- College of life science and biotechnology, Hebei Normal University of Science and Technology, Qinhuangdao, 066004, China.
| | - Qiuyan Li
- State Key Laboratory of Agrobiotechnology & College of Biological Sciences, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|
6
|
Carey K, Ryu J, Uh K, Lengi AJ, Clark-Deener S, Corl BA, Lee K. Frequency of off-targeting in genome edited pigs produced via direct injection of the CRISPR/Cas9 system into developing embryos. BMC Biotechnol 2019; 19:25. [PMID: 31060546 PMCID: PMC6501304 DOI: 10.1186/s12896-019-0517-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Accepted: 04/23/2019] [Indexed: 12/24/2022] Open
Abstract
Background The CRISPR/Cas9 system can effectively introduce site-specific modifications to the genome. The efficiency is high enough to induce targeted genome modifications during embryogenesis, thus increasing the efficiency of producing genetically modified animal models and having potential clinical applications as an assisted reproductive technology. Because most of the CRISPR/Cas9 systems introduce site-specific double-stranded breaks (DSBs) to induce site-specific modifications, a major concern is its potential off-targeting activity, which may hinder the application of the technology in clinics. In this study, we investigated off-targeting events in genome edited pigs/fetuses that were generated through direct injection of the CRISPR/Cas9 system into developing embryos; off-targeting activity of four different sgRNAs targeting RAG2, IL2RG, SCD5, and Ig Heavy chain were examined. Results First, bioinformatics analysis was applied to identify 27 potential off-targeting genes from the sgRNAs. Then, PCR amplification followed by sequencing analysis was used to verify the presence of off-targeting events. Off-targeting events were only identified from the sgRNA used to disrupt Ig Heavy chain in pigs; frequency of off-targeting was 80 and 70% on AR and RBFOX1 locus respectively. A potential PAM sequence was present in both of the off-targeting genes adjacent to probable sgRNA binding sites. Mismatches against sgRNA were present only on the 5′ side of AR, suggesting that off-targeting activities are systematic events. However, the mismatches on RBFOX1 were not limited to the 5′ side, indicating unpredictability of the events. Conclusions The prevalence of off-targeting is low via direct injection of CRISPR/Cas9 system into developing embryos, but the events cannot be accurately predicted. Off-targeting frequency of each CRISPR/Cas9 system should be deliberately assessed prior to its application in clinics. Electronic supplementary material The online version of this article (10.1186/s12896-019-0517-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Kayla Carey
- Department of Animal and Poultry Sciences, Virginia Tech, Blacksburg, VA, 24061, USA
| | - Junghyun Ryu
- Department of Animal and Poultry Sciences, Virginia Tech, Blacksburg, VA, 24061, USA
| | - Kyungjun Uh
- Department of Animal and Poultry Sciences, Virginia Tech, Blacksburg, VA, 24061, USA
| | - Andrea J Lengi
- Department of Dairy Science, Virginia Tech, Blacksburg, VA, 24061, USA
| | | | - Benjamin A Corl
- Department of Dairy Science, Virginia Tech, Blacksburg, VA, 24061, USA
| | - Kiho Lee
- Department of Animal and Poultry Sciences, Virginia Tech, Blacksburg, VA, 24061, USA.
| |
Collapse
|
7
|
Yan S, Li S, Li XJ. Use of large animal models to investigate Huntington's diseases. CELL REGENERATION 2019; 8:9-11. [PMID: 31205683 PMCID: PMC6557753 DOI: 10.1016/j.cr.2019.01.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 01/10/2019] [Accepted: 01/21/2019] [Indexed: 01/21/2023]
Abstract
Animal models that can mimic human diseases are the important tools for investigating the pathogenesis of the diseases and finding a way for treatment. There is no doubt that small animal models have provided a wealth of information regarding disease pathogenesis and also offered widely used tools to develop therapeutic strategies. Rodent models have been very valuable for investigators to understand the mechanisms underlying misfolded protein-mediated neuronal dysfunction and behavioral phenotypes in a variety of neurodegenerative diseases including Alzheimer's, Parkinson's, and Huntington's diseases (HD). However, most of genetically modified rodent models of these diseases lack the overt and selective neurodegeneration seen in the patient brains. Since large animals are more similar to humans than small animals and rodents, the large animal models are likely to mimic important neuropathological features in humans. Here we discuss the application of large animal models in neurodegenerative disease research with focus on the HD large animal models, aiming to provide insight into the application of animal models to study neurodegenerative diseases.
Collapse
Affiliation(s)
- Sen Yan
- Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Shihua Li
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Xiao-Jiang Li
- Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
- Corresponding author. Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou. 510632, China.
| |
Collapse
|
8
|
Zhu XX, Zhong YZ, Ge YW, Lu KH, Lu SS. CRISPR/Cas9-Mediated Generation of Guangxi Bama Minipigs Harboring Three Mutations in α-Synuclein Causing Parkinson's Disease. Sci Rep 2018; 8:12420. [PMID: 30127453 PMCID: PMC6102220 DOI: 10.1038/s41598-018-30436-3] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 07/25/2018] [Indexed: 12/31/2022] Open
Abstract
Parkinson’s disease (PD) is a common, progressive neurodegenerative disorder characterized by classical motor dysfunction and is associated with α-synuclein-immunopositive pathology and the loss of dopaminergic neurons in the substantia nigra (SN). Several missense mutations in the α-synuclein gene SCNA have been identified as cause of inherited PD, providing a practical strategy to generate genetically modified animal models for PD research. Since minipigs share many physiological and anatomical similarities to humans, we proposed that genetically modified minipigs carrying PD-causing mutations can serve as an ideal model for PD research. In the present study, we attempted to model PD by generating Guangxi Bama minipigs with three PD-causing missense mutations (E46K, H50Q and G51D) in SCNA using CRISPR/Cas9-mediated gene editing combining with somatic cell nuclear transfer (SCNT) technique. We successfully generated a total of eight SCNT-derived Guangxi Bama minipigs with the desired heterozygous SCNA mutations integrated into genome, and we also confirmed by DNA sequencing that these minipigs expressed mutant α-synuclein at the transcription level. However, immunohistochemical analysis was not able to detect PD-specific pathological changes such as α-synuclein-immunopositive pathology and loss of SN dopaminergic neurons in the gene-edited minipigs at 3 months of age. In summary, we successfully generated Guangxi Bama minipigs harboring three PD-casusing mutations (E46K, H50Q and G51D) in SCNA. As they continue to develop, these gene editing minipigs need to be regularly teseted for the presence of PD-like pathological features in order to validate the use of this large-animal model in PD research.
Collapse
Affiliation(s)
- Xiang-Xing Zhu
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources; Guangxi High Education Key Laboratory for Animal Reproduction and Biotechnology; College of Animal Science and Technology, Guangxi University, Nanning, 530004, China.
| | - Yi-Zhi Zhong
- Guangxi Nanning Yanleshang Biotechnology Co. LTD, Nanning, 530004, China
| | - Yao-Wen Ge
- Wuhan ViaGen Animal Breeding Resources Development Company, Wuhan, 430073, China
| | - Ke-Huan Lu
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources; Guangxi High Education Key Laboratory for Animal Reproduction and Biotechnology; College of Animal Science and Technology, Guangxi University, Nanning, 530004, China
| | - Sheng-Sheng Lu
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources; Guangxi High Education Key Laboratory for Animal Reproduction and Biotechnology; College of Animal Science and Technology, Guangxi University, Nanning, 530004, China.
| |
Collapse
|
9
|
Yan S, Tu Z, Liu Z, Fan N, Yang H, Yang S, Yang W, Zhao Y, Ouyang Z, Lai C, Yang H, Li L, Liu Q, Shi H, Xu G, Zhao H, Wei H, Pei Z, Li S, Lai L, Li XJ. A Huntingtin Knockin Pig Model Recapitulates Features of Selective Neurodegeneration in Huntington's Disease. Cell 2018; 173:989-1002.e13. [PMID: 29606351 DOI: 10.1016/j.cell.2018.03.005] [Citation(s) in RCA: 212] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2017] [Revised: 01/21/2018] [Accepted: 02/27/2018] [Indexed: 01/23/2023]
Abstract
Huntington's disease (HD) is characterized by preferential loss of the medium spiny neurons in the striatum. Using CRISPR/Cas9 and somatic nuclear transfer technology, we established a knockin (KI) pig model of HD that endogenously expresses full-length mutant huntingtin (HTT). By breeding this HD pig model, we have successfully obtained F1 and F2 generation KI pigs. Characterization of founder and F1 KI pigs shows consistent movement, behavioral abnormalities, and early death, which are germline transmittable. More importantly, brains of HD KI pig display striking and selective degeneration of striatal medium spiny neurons. Thus, using a large animal model of HD, we demonstrate for the first time that overt and selective neurodegeneration seen in HD patients can be recapitulated by endogenously expressed mutant proteins in large mammals, a finding that also underscores the importance of using large mammals to investigate the pathogenesis of neurodegenerative diseases and their therapeutics.
Collapse
Affiliation(s)
- Sen Yan
- Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, 510632 Guangzhou, China
| | - Zhuchi Tu
- Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, 510632 Guangzhou, China
| | - Zhaoming Liu
- Key Laboratory of Regenerative Biology, South China Institute for Stem Cell, Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 510530 Guangzhou, China
| | - Nana Fan
- Key Laboratory of Regenerative Biology, South China Institute for Stem Cell, Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 510530 Guangzhou, China
| | - Huiming Yang
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Su Yang
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Weili Yang
- Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, 510632 Guangzhou, China
| | - Yu Zhao
- Key Laboratory of Regenerative Biology, South China Institute for Stem Cell, Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 510530 Guangzhou, China
| | - Zhen Ouyang
- Key Laboratory of Regenerative Biology, South China Institute for Stem Cell, Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 510530 Guangzhou, China
| | - Chengdan Lai
- Key Laboratory of Regenerative Biology, South China Institute for Stem Cell, Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 510530 Guangzhou, China
| | - Huaqiang Yang
- Key Laboratory of Regenerative Biology, South China Institute for Stem Cell, Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 510530 Guangzhou, China
| | - Li Li
- Key Laboratory of Regenerative Biology, South China Institute for Stem Cell, Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 510530 Guangzhou, China
| | - Qishuai Liu
- Key Laboratory of Regenerative Biology, South China Institute for Stem Cell, Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 510530 Guangzhou, China
| | - Hui Shi
- Key Laboratory of Regenerative Biology, South China Institute for Stem Cell, Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 510530 Guangzhou, China
| | - Guangqing Xu
- Department of Neurology, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080 Guangdong, China
| | - Heng Zhao
- College of Animal Science and Technology, Yunnan Agricultural University, 650201 Kunming, China
| | - Hongjiang Wei
- College of Animal Science and Technology, Yunnan Agricultural University, 650201 Kunming, China
| | - Zhong Pei
- Department of Neurology, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080 Guangdong, China
| | - Shihua Li
- Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, 510632 Guangzhou, China; Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA.
| | - Liangxue Lai
- Key Laboratory of Regenerative Biology, South China Institute for Stem Cell, Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 510530 Guangzhou, China; Jilin Provincial Key Laboratory of Animal Embryo Engineering, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, 130062 Changchun, China.
| | - Xiao-Jiang Li
- Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, 510632 Guangzhou, China; Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA.
| |
Collapse
|
10
|
Li Z, Yang HY, Wang Y, Zhang ML, Liu XR, Xiong Q, Zhang LN, Jin Y, Mou LS, Liu Y, Li RF, Rao Y, Dai YF. Generation of tryptophan hydroxylase 2 gene knockout pigs by CRISPR/Cas9-mediated gene targeting. J Biomed Res 2017; 31:445-452. [PMID: 28866660 PMCID: PMC5706437 DOI: 10.7555/jbr.31.20170026] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Unbalanced brain serotonin (5-HT) levels have implications in various behavioral abnormalities and neuropsychiatric disorders. The biosynthesis of neuronal 5-HT is regulated by the rate-limiting enzyme, tryptophan hydroxylase-2 (TPH2). In the present study, the clustered regularly interspaced short palindromic repeat (CRISPR)/CRISPR-associated (Cas) system was used to target theTph2 gene in Bama mini pig fetal fibroblasts. It was found that CRISPR/Cas9 targeting efficiency could be as high as 61.5%, and the biallelic mutation efficiency reached at 38.5%. The biallelic modified colonies were used as donors for somatic cell nuclear transfer (SCNT) and 10Tph2 targeted piglets were successfully generated. These Tph2 KO piglets were viable and appeared normal at the birth. However, their central 5-HT levels were dramatically reduced, and their survival and growth rates were impaired before weaning. TheseTph2 KO pigs are valuable large-animal models for studies of 5-HT deficiency induced behavior abnomality.
Collapse
Affiliation(s)
- Ze Li
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Hai-Yuan Yang
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Ying Wang
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Man-Ling Zhang
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Xiao-Rui Liu
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Qiang Xiong
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Li-Ning Zhang
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Yong Jin
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Li-Sha Mou
- Shenzhen Xenotransplantation Medical Engineering Research and Development Center, Institute of Translational Medicine, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong 518035, China
| | - Yan Liu
- Peking-Tsinghua Center for Life Sciences, PKU-IDG/McGovern Institute for Brain Research, the School of Life Sciences, Peking University, Beijing 100871, China
| | - Rong-Feng Li
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Yi Rao
- Peking-Tsinghua Center for Life Sciences, PKU-IDG/McGovern Institute for Brain Research, the School of Life Sciences, Peking University, Beijing 100871, China
| | - Yi-Fan Dai
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| |
Collapse
|