8
views
0
recommends
+1 Recommend
1 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: not found

      Zygote injection of CRISPR/Cas9 RNA successfully modifies the target gene without delaying blastocyst development or altering the sex ratio in pigs

      research-article

      Read this article at

      ScienceOpenPublisherPMC
      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          The CRISPR/Cas9 genome editing tool has increased the efficiency of creating genetically modified pigs for use as biomedical or agricultural models. The objectives were to determine if DNA editing resulted in a delay in development to the blastocyst stage or in a skewing of the sex ratio. Six DNA templates (gBlocks) that were designed to express guide RNAs that target the transmembrane protease, serine S1, member 2 ( TMPRSS2) gene were in vitro transcribed. Pairs of CRISPR guide RNAs that flanked the start codon and polyadenylated Cas9 were co-injected into the cytoplasm of zygotes and cultured in vitro to the blastocyst stage. Blastocysts were collected as they formed on days 5, 6 or 7. PCR was performed to determine genotype and sex of each embryo. Separately, embryos were surgically transferred into recipient gilts on day 4 of estrus. The rate of blastocyst development was not significantly different between CRISPR injection embryos or the non-injected controls at day 5, 6 or 7 ( p = 0.36, 0.09, 0.63, respectively). Injection of three CRISPR sets of guides resulted in a detectable INDEL in 92–100 % of the embryos analyzed. There was not a difference in the number of edits or sex ratio of male to female embryos when compared between days 5, 6 and 7 to the controls ( p > 0.22, >0.85). There were 12 resulting piglets and all 12 had biallelic edits of TMRPSS2. Zygote injection with CRISPR/Cas9 continues to be a highly efficient tool to genetically modify pig embryos.

          Electronic supplementary material

          The online version of this article (doi:10.1007/s11248-016-9989-6) contains supplementary material, which is available to authorized users.

          Related collections

          Most cited references28

          • Record: found
          • Abstract: found
          • Article: found
          Is Open Access

          One-step generation of knockout pigs by zygote injection of CRISPR/Cas system

          Dear Editor, The pig is an important livestock for food supply and an ideal model for various human diseases. Efficient and precise genetic engineering in pigs holds great promise in agriculture and biomedicine 1 . Using currently available approach, generating specific gene modifications in pigs requires two steps. First, site-specific nucleases such as zinc finger nucleases (ZFNs) and transcription activator-like effector nucleases (TALENs) are used to generate targeted mutations in pig somatic cells. Then the engineered somatic nucleus is used to generate cloned animals using somatic cell nuclear transfer (SCNT) technology 2,3 . The complex design and generation of ZFNs and TALENs, as well as the technical challenges of SCNT, greatly limit the application of this method. Recently a new type of site-specific nuclease was identified in CRISPR (clustered regularly interspaced short palindromic repeat)/Cas (CRISPR-associated) RNA-based adaptive immune system in bacteria and archaea 4 . The Cas9 endonuclease from Streptococcus pyogenes type II CRISPR/Cas system can be engineered to produce targeted genome modification under the guidance of a synthetic single guide RNA (sgRNA) with simple base-pair complementarities with a target genomic DNA sequence 5,6,7 . Although highly efficient one-step generation of mice and rats carrying single or multiple mutations have been achieved through direct injection of CRISPR/Cas system into one-cell embryo 8,9 , it remains to be tested whether the same strategy could be applied to generation of genome-engineered large animals. Here we report the efficient generation of biallelic knockout pigs in one step by direct cytoplasmic injection of Cas9 mRNA and sgRNA into zygotes. To demonstrate the application of genome engineering in disease modeling and agriculture, we chose to target vWF gene, whose deficiency in human causes severe von Willebrand disease (vWD) 10 (Supplementary information, Figure S1). As mouse model does not fully recapitulate the disease phenotype and response to treatment in human patients, a large animal model with similar size of organ and circulatory system is highly desirable. In food industry, bleeding is an important procedure before meat production, as the leftover blood is an ideal medium for bacterial growth. vWF knockout could improve bleeding efficiency for slaughtering procedures and blood collection. We designed an sgRNA targeting exon 5 of the pig vWF gene, which lies in the first trysin-inhibitor-like domain and its mutation could lead to the loss of function of vWF protein. The in vitro developmental efficiencies of embryos injected with Cas9 mRNA/sgRNA (∼79%) and embryos injected with water (∼77%) were both very high and comparable with each other, suggesting that the microinjection manipulation and the Cas9 mRNA/sgRNA had little effect on pig early embryonic development (Supplementary information, Table S1). We next transferred the injected embryos into surrogate pigs to produce piglets. A total of 76 injected embryos were transplanted into 5 surrogate mothers in 5 independent experiments. Three surrogates were successfully pregnant and delivered a total of 16 piglets (Figure 1A). T7 endonuclease I (T7EI) assay showed that 10 piglets contained insertions or deletions (indels) in the targeting site, indicating that vWF mutation efficiency reached about 63% (10/16) (Figure 1A and 1B). We further analyzed the genotype by Sanger sequencing of the PCR products containing the targeting site of each individual piglet. The sequencing results confirmed the indels and revealed that one mutant pig yielding uncut PCR product in the T7EI assay (fourth lane in Figure 1B) contained only one type of mutant allele. Most pigs contained more than two different alleles (Figure 1C, pig #4 and #11), suggesting that the Cas9-mediated DNA cleavage may occur at embryonic stage later than the one-cell stage, as previously observed with ZFN, TALEN and CRISPR-mediated targeting 11 . In each pig, if no wild-type allele was detected by sequencing, we categorized the pig as biallelic mutant. If both wild-type and mutant alleles were observed in the sequence reads, we categorized the pig as monoallelic mutant, although the ratio of wild-type to mutant alleles might not always be 1. In total, we identified six vWF biallelic mutant piglets and five monoallelic mutant piglets (11/16, 68%) (Figure 1A and 1C). Of these piglets, two died shortly after birth and the other 14 survived until now (Figure 1D). The high birth rate (16/76, 21%) and survival rate (14/16, 88%) indicated that Cas9 mRNA/sgRNA had little toxicity to pig embryonic development (Figure 1A). These results demonstrate that the CRISPR/Cas system could efficiently generate both monoallelic and biallelic mutant pigs in one step through direct zygote injection. We further examined whether the vWF mutations cause the vWD phenotypes. Western blot analysis showed that the vWF expression in lung was largely reduced in monoallelic mutant pigs, and was undetectable in biallelic mutant pigs, confirming the vWF knockout at protein levels (Figure 1E). We next performed hematological analysis with pigs having different genotypes. The plasma vWF antigen level was significantly reduced in the monoallelic mutant pig group, and was nearly undetectable in biallelic mutant group (Figure 1F). In human vWD, the level of coagulation factor FVIII may be reduced due to rapid degradation in the absence of vWF 10 . Consistently, the activated partial thromboplastin time (APTT)-based assay for FVIII activity showed a significantly lower activity in the biallelic mutant group, indicating that FVIII level was decreased in the plasma of vWF-null pigs (Figure 1G). No significant differences were observed between wild-type and knockout pigs with regard to other parameters in the hematological analysis (Supplementary information, Table S2). The most prominent phenotype resulting from vWF mutation is a severe bleeding tendency 10 . We found that the bleeding time of biallelic mutant pigs (512 ± 121 min) was much longer than that of wild-type pigs (34 ± 11 min) (Figure 1H). Taken together, our results demonstrate that the one-step generated biallelic mutant pigs developed similar phenotypes of the human vWD. A certain level of off-target cleavage by CRISPR/Cas system was suggested by recent studies in human cell lines 12 , indicating that Cas9-mediated DNA cleavage tolerates small numbers of mismatches between sgRNA and target DNA especially in protospacer adjacent motif (PAM)-distal region 12 . We thus sought to test the possible off-target effects in genome-modified pigs derived from zygote injection. By screening the genome based on sequence homology (> 14 base pair identity to 3′ end of the sgRNA spacer), around 20 potential off-target sites were found (Supplementary information, Figure S2). In all 11 mutant animals tested, none of them contained indels at 2 randomly chosen sites based on T7EI assay (Supplementary information, Figure S2). We further analyzed the potential off-target effects by Sanger sequencing of 10 randomly selected potential off-target sites in 3 biallelic mutant pigs (#9, #10 and #11). None of the sequencing reads had mutations, suggesting no off-target effects at these sites (Supplementary information, Figure S2). However, we could not exclude the possible existence of certain off-target effects due to the high number of potential off-target sites. Thus a systematic off-target evaluation would be needed in the future before the wide application of such knockout pig models. Also, modified CRISPR/Cas-methods with less off-target tendency can be used for genome engineering in pigs and other large animals 13 . Germline transmission of the mutations to next generation is essential for expanding the knockout pig population. To examine the germline transmission potential of the mutations, we dissected an ovary from a monoallelic mutant female pig and a testis from a biallelic mutant male pig by biopsy and analyzed their genotypes by Sanger sequencing. Both the ovary and the testis were morphologically normal (Supplementary information, Figure S3). More than half sequence reads of the ovary and all the sequence reads of the testis were mutant, indicating that the mutations had been efficiently transmitted into the germline (Supplementary information, Figure S3). Considering the highly efficient germline transmission in rodents using the same procedure 14 , germline transmission of the mutations to next generation is highly expected. In summary, we have shown that zygote injection of the CRISPR/Cas system can efficiently generate genome-modified pigs in one step, resulting in rapid establishment of large animal model for important human diseases. The high efficiency of Cas9-mediated genome editing in pigs demonstrated here, together with the versatile functionality of the CRISPR/Cas system such as multiplexed genome editing and epigenetic modification 5 , opens up unlimited possibilities of genome engineering in livestock for applications in agriculture and biomedicine.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Use of the CRISPR/Cas9 system to produce genetically engineered pigs from in vitro-derived oocytes and embryos.

            Targeted modification of the pig genome can be challenging. Recent applications of the CRISPR/Cas9 system hold promise for improving the efficacy of genome editing. When a designed CRISPR/Cas9 system targeting CD163 or CD1D was introduced into somatic cells, it was highly efficient in inducing mutations. When these mutated cells were used with somatic cell nuclear transfer, offspring with these modifications were created. When the CRISPR/Cas9 system was delivered into in vitro produced presumptive porcine zygotes, the system was effective in creating mutations in eGFP, CD163, and CD1D (100% targeting efficiency in blastocyst stage embryos); however, it also presented some embryo toxicity. We could also induce deletions in CD163 or CD1D by introducing two types of CRISPRs with Cas9. The system could also disrupt two genes, CD163 and eGFP, simultaneously when two CRISPRs targeting two genes with Cas9 were delivered into zygotes. Direct injection of CRISPR/Cas9 targeting CD163 or CD1D into zygotes resulted in piglets that have mutations on both alleles with only one CD1D pig having a mosaic genotype. We show here that the CRISPR/Cas9 system can be used by two methods. The system can be used to modify somatic cells followed by somatic cell nuclear transfer. System components can also be used in in vitro produced zygotes to generate pigs with specific genetic modifications. © 2014 by the Society for the Study of Reproduction, Inc.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Production of CFTR-null and CFTR-DeltaF508 heterozygous pigs by adeno-associated virus-mediated gene targeting and somatic cell nuclear transfer.

              Progress toward understanding the pathogenesis of cystic fibrosis (CF) and developing effective therapies has been hampered by lack of a relevant animal model. CF mice fail to develop the lung and pancreatic disease that cause most of the morbidity and mortality in patients with CF. Pigs may be better animals than mice in which to model human genetic diseases because their anatomy, biochemistry, physiology, size, and genetics are more similar to those of humans. However, to date, gene-targeted mammalian models of human genetic disease have not been reported for any species other than mice. Here we describe the first steps toward the generation of a pig model of CF. We used recombinant adeno-associated virus (rAAV) vectors to deliver genetic constructs targeting the CF transmembrane conductance receptor (CFTR) gene to pig fetal fibroblasts. We generated cells with the CFTR gene either disrupted or containing the most common CF-associated mutation (DeltaF508). These cells were used as nuclear donors for somatic cell nuclear transfer to porcine oocytes. We thereby generated heterozygote male piglets with each mutation. These pigs should be of value in producing new models of CF. In addition, because gene-modified mice often fail to replicate human diseases, this approach could be used to generate models of other human genetic diseases in species other than mice.
                Bookmark

                Author and article information

                Contributors
                PratherR@Missouri.edu
                Journal
                Transgenic Res
                Transgenic Res
                Transgenic Research
                Springer International Publishing (Cham )
                0962-8819
                1573-9368
                15 October 2016
                2017
                : 26
                : 1
                : 97-107
                Affiliations
                [1 ]GRID grid.134936.a, ISNI 0000000121623504, Division of Animal Sciences, , University of Missouri, ; 920 East Campus Dr., E125D ASRC, Columbia, MO 65211 USA
                [2 ]GRID grid.134936.a, ISNI 0000000121623504, National Swine Research and Resource Center, , University of Missouri, ; 920 East Campus Dr., E125D ASRC, Columbia, MO 65211 USA
                [3 ]GRID grid.36567.31, ISNI 0000000107371259, College of Veterinary Medicine, Diagnostic Medicine/Pathobiology, , Kansas State University, ; Manhattan, KS USA
                Article
                9989
                10.1007/s11248-016-9989-6
                5247313
                27744533
                d9cc934f-5622-41f7-bac2-31a7c295a173
                © Springer International Publishing Switzerland 2016

                This article is made available via the PMC Open Access Subset for unrestricted research re-use and secondary analysis in any form or by any means with acknowledgement of the original source. These permissions are granted for the duration of the World Health Organization (WHO) declaration of COVID-19 as a global pandemic.

                History
                : 13 July 2016
                : 4 October 2016
                Funding
                Funded by: FundRef http://dx.doi.org/10.13039/100000002, National Institutes of Health;
                Award ID: U42OD011140
                Award ID: HHSN266200700005C
                Award Recipient :
                Categories
                Original Paper
                Custom metadata
                © Springer International Publishing Switzerland 2017

                Genetics
                crispr/cas9,zygote injection,dna editing,gblock
                Genetics
                crispr/cas9, zygote injection, dna editing, gblock

                Comments

                Comment on this article