Elsevier

Antiviral Research

Volume 130, June 2016, Pages 50-57
Antiviral Research

Establishment of a highly efficient virus-inducible CRISPR/Cas9 system in insect cells

https://doi.org/10.1016/j.antiviral.2016.03.009Get rights and content

Highlights

  • CRISPR/Cas9 system is capable of specifically disrupting ie-1 gene in BmNPV genome.

  • The CRISPR/Cas9 system effectively edits BmNPV genome and inhibits virus replication.

  • Virus-inducible CRISPR/Cas9 system is expected to reduce the probability of off-target effects in the insect cells.

Abstract

Although current antiviral strategies can inhibit baculovirus infection and decrease viral DNA replication to a certain extent, novel tools are required for specific and accurate elimination of baculovirus genomes from infected insects. Using the newly developed clustered regularly interspaced short palindromic repeats/associated protein 9 nuclease (CRISPR/Cas9) technology, we disrupted a viral genome in infected insect cells in vitro as a defense against viral infection. We optimized the CRISPR/Cas9 system to edit foreign and viral genome in insect cells. Using Bombyx mori nucleopolyhedrovirus (BmNPV) as a model, we found that the CRISPR/Cas9 system was capable of cleaving the replication key factor ie-1 in BmNPV thus effectively inhibiting virus proliferation. Furthermore, we constructed a virus-inducible CRISPR/Cas9 editing system, which minimized the probability of off-target effects and was rapidly activated after viral infection. This is the first report describing the application of the CRISPR/Cas9 system in insect antiviral research. Establishment of a highly efficient virus-inducible CRISPR/Cas9 system in insect cells provides insights to produce virus-resistant transgenic strains for future.

Introduction

Baculovirus plays an important role as a natural agent controlling the size of insect populations (Jiang and Xia, 2014, Zhang et al., 2014a). However, it is a serious threat for beneficial insects such as silkworm, which is an important Lepidopteran model insect of economic value (Jiang and Xia, 2014). Since silkworm is the only host of Bombyx mori nucleopolyhedrovirus (BmNPV), viral resistance strains of this lepidopteran will benefit the silk industry. Thus far, a number of antiviral strategies have been used to create transgenic silkworms; these include RNA interference of the BmNPV core gene, overexpression of an endogenous or exogenous antiviral gene and regulation of host immunity. Current antiviral strategies only reduce viral replication and suppress the virus in infected cells and tissues but do not eliminate the viral genome from an infected silkworm.

Recent studies have described an efficient gene editing system targeting viral genomes with clustered regularly interspaced short palindromic repeats/CRISPR-associated protein 9 nuclease (CRISPR/Cas9) technology (Lijuan et al., 2015, Ma et al., 2014, Pellagatti et al., 2015). CRISPR/Cas9 is a component of the adaptive immune defense system in bacteria and archaea, and is used to defend against invading viruses and exogenous DNA (Ebina et al., 2013, Lijuan et al., 2015, Pellagatti et al., 2015). Recently, the CRISPR/Cas9 genome editing tool has been used widely for applied to the systematic investigate mammalian genomes in cell cultures (Ran et al., 2013, Shalem et al., 2014, Wang et al., 2014). This genome editing technology has great potential to edit animal genomes, and has been used for somatic genome editing (Chiou et al., 2015, Sanchez-Rivera et al., 2014, Wang et al., 2015), to correct genetic disorders (Meissner et al., 2014, Zou et al., 2015) and treat infectious diseases (Ebina et al., 2013, Ramanan et al., 2015, Seeger and Sohn, 2014, Tu et al., 2015, Yuan et al., 2015b). However, there are huge challenges in the large-scale application of the CRISPR/Cas9 genome editing system due to the high potential for off-target effects and homologous sequence repairs.

Recently, Ebina et al. reported that the CRISPR/Cas9 system can edit HIV-1 genome in human cells resulting in the removal of the viral DNA from host cells (Ebina et al., 2013, Sanyal et al., 2013). The technology has been widely applied in gene therapy. Several studies have successfully applied the CRISPR/Cas9 system to treat infectious diseases by disrupting viral genes, editing viral DNA and eliminating the virus genome (Ebina et al., 2013, Pellagatti et al., 2015). More importantly, the editing of human herpes virus, human papillomavirus and hepatitis B virus genomes with the CRISPR/Cas9 system has achieved remarkable success (Ebina et al., 2013, Liao et al., 2015, Seeger and Sohn, 2014, Yuan et al., 2015b). However, the CRISPR/Cas9 system has not been applied to counter viral infections in insects.

In the present study, we applied a highly efficient virus-inducible CRISPR/Cas9 technology to disrupt BmNPV in infected silkworm. First, we improved the CRISPR/Cas9 system gene editing efficiency by linking Cas9 and sgRNA expression cassettes in the same vector. Furthermore, to reduce the probability of off-target effects and the possible toxicity of CRISPR/Cas9 system induced by long-term expression of exogenous proteins in host cells, we optimized the gene editing system. Our results showed the successful construction of a CRISPR/Cas9 genome editing system with reduced potential off-target of effects and high editing efficiency that can be used for antiviral research in insects.

Section snippets

Cells and viruses

The B. mori cell line BmN-SWU1, was cultured at 27 °C in TC-100 medium (United States Biological, USA) supplemented with 10% (V/V) fetal bovine serum (FBS) (Gibco, USA) and 10% (V/V) penicillin/streptomycin (Pan et al., 2010). Recombinant BmNPV (vA4prm-EGFP) containing an EGFP marker gene driven by the B. mori actin A4 promoter was constructed previously (Dong et al., 2014, Zhang et al., 2014a). Viruses were cultured in BmN-SWU1 cells, and viral titers were determined by 50% tissue culture

Excision of the ie1 gene from BmNPV genome

Baculoviruses quickly establish a systemic infection in B. mori. Therefore, it is critical to rapidly disable viral replication in B. mori. For this purpose, we used the CRISPR/Cas9 system and the immediate early gene, ie-1, in BmNPV as the editing target. Besides, we linked all three expression cassettes (Cas9, U6-sgRNA and mCherry) into a single vector to improve the efficiency of the CRISPR/Cas9 system (Fig. 1A). After transfecting pIZ-Cas9-sgIE1-mCherry and pIZ-Cas9-sgMock-mCherry into

Discussion

Antiviral studies in silkworm are essential to improve not only sericulture production but also to control insect pests (Dong et al., 2014, Jiang and Xia, 2014, Zhang et al., 2014a). Thus far, disrupting the viral genome with the CRISPR/Cas9 gene editing system has not been previously reported in the baculovirus (Pellagatti et al., 2015, Wei et al., 2014). To apply the CRISPR/Cas9 system for insect pest control and to treat infectious diseases, we constructed a highly efficient gene editing

Acknowledgments

This work was supported by grants from the National High-tech R&D Program of China (No. 2013AA102507), the National Natural Science Foundation of China (Nos. 31272505, 31472152 and 31572466), China Agriculture Research System (CARS-22), China Postdoctoral Science Foundation (2015M582502) and Fundamental Research Funds for the Central Universities (XDJK2015D002).

References (37)

  • J.H. Zhang et al.

    Improving the specificity and efficacy of CRISPR/CAS9 and gRNA through target specific DNA reporter

    J. Biotechnol.

    (2014)
  • B. Zou et al.

    The application of genome editing in studying hearing loss

    Hear. Res.

    (2015)
  • C.S. Asensio

    Application of Cas9/CRISPR to the study of synaptic function

    Med. Sci. M S

    (2015)
  • S.H. Chiou et al.

    Pancreatic cancer modeling using retrograde viral vector delivery and in vivo CRISPR/Cas9-mediated somatic genome editing

    Genes & Dev.

    (2015)
  • H. Ebina et al.

    Harnessing the CRISPR/Cas9 system to disrupt latent HIV-1 provirus

    Sci. Rep.

    (2013)
  • W. Jiang et al.

    CRISPR-Cas: new tools for genetic manipulations from bacterial immunity systems

    Annu. Rev. Microbiol.

    (2015)
  • B.P. Kleinstiver et al.

    High-fidelity CRISPR-Cas9 nucleases with no detectable genome-wide off-target effects

    Nature

    (2016)
  • J.F. Li et al.

    Targeted plant genome editing via the CRISPR/Cas9 technology

    Methods Mol. Biol.

    (2015)
  • Cited by (48)

    • Baculovirus LEF-11 interacts with BmIMPI to induce cell cycle arrest in the G2/M phase for viral replication

      2022, Pesticide Biochemistry and Physiology
      Citation Excerpt :

      The cDNA sequences for all target genes were cloned in-frame with an HA or FLAG epitope using Kpn I and BamH I restriction sites. For knockout vectors, we designed the target sequence using online software (http://crispr.dbcls.jp/), and oligos sgRNAs were cloned to pSL1180-IE1-Cas9-Ser-PA-U6-sgRNA gene editing vectors as previously described (Dong et al., 2016). For the yeast two-hybrid plasmids, pGBKT7-LEF11 and pGADT7-target genes were constructed to express the bait and prey proteins, respectively.

    • CRISPR/Cpf1 multiplex genome editing system increases silkworm tolerance to BmNPV

      2022, International Journal of Biological Macromolecules
      Citation Excerpt :

      Therefore, the release of gene editing lines carrying CRISPR/Cpf1 requires strict pre-release risk assessment of non-target effects to prevent accidental ecological consequences [36]. Previously, we constructed a virus-inducible gene editing system, which can avoid the release of Cas9 protein [25]. Therefore, we can construct a virus inducible multiplex CRISPR/Cpf1 genome editing system, which can activate Cpf1 only in virus infected cells and avoid the release of CRISPR/Cpf1 gene editing system.

    • Transgenic genome editing-derived antiviral therapy to nucleopolyhedrovirus infection in the industrial strain of the silkworm

      2021, Insect Biochemistry and Molecular Biology
      Citation Excerpt :

      Relative to WT specimens, our TG Jingsong strain displayed a slight decrease in number of offspring and cocoon shell rate, and we postulate that this may have been caused by the poisonousness exogenous CRISPR/Cas9 system (Haapaniemi et al., 2018; Kosicki et al., 2018; Xu et al., 2020). Perhaps by using a viral induce promoter expressing Cas9 will minimize the negative effects (Dong et al., 2016, 2018). Nevertheless, the TG Jingsong strain exhibited extremely enhanced antivirus capacity, which suggests its use in the sericulture industry may still bring economic benefits.

    View all citing articles on Scopus
    View full text