open access

Vol 58, No 3 (2020)
Original paper
Submitted: 2020-07-29
Accepted: 2020-09-08
Published online: 2020-09-16
Get Citation

A knockdown of the herpes simplex virus type-1 gene in all-in-one CRISPR vectors

Nastaran Khodadad12, Mona Fani13, Saleh Jamehdor4, Rahil Nahidsamiei12, Manoochehr Makvandi12, Saeed Kaboli5, Ali Teimoori126, Jose Thekkiniath78
·
Pubmed: 32937678
·
Folia Histochem Cytobiol 2020;58(3):174-181.
Affiliations
  1. Cancer Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
  2. Department of Virology, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
  3. Department of Pathobiology and Laboratory Sciences, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
  4. Department of Biology, Faculty of Sciences, University of Sistan and Baluchestan, Zahedan, Iran
  5. Department of Medical Biotechnology and Cancer Gene Therapy Research Center, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
  6. Department of Virology, Faculty of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
  7. Fuller Laboratories, Fullerton, CA, USA
  8. Department of Internal Medicine, Section of Infectious Diseases, Yale School of Medicine, New Haven, CT, USA

open access

Vol 58, No 3 (2020)
ORIGINAL PAPERS
Submitted: 2020-07-29
Accepted: 2020-09-08
Published online: 2020-09-16

Abstract

Introduction. Herpes simplex virus type 1 (HSV-1) is a virus that causes serious human disease and establishes a long-term latent infection. The latent form of this virus has shown to be resistant to antiviral drugs. Clustered Regularly Interspace Short Palindromic Repeats (CRISPR), is an important tool in genome engineering and composed of guide RNA (gRNA) and Cas9 nuclease that makes an RNA-protein complex to digest exclusive target sequences implementation of gRNA. Moreover, CRISPR-Cas9 system effectively suppresses HSV-1 infection by knockout of some viral genes.

Materials and methods. To survey the efficacy of Cas9 system on HSV-1 genome destruction, we designed several guide RNAs (gRNAs) that all packaged in one vector. Additionally, we performed a one-step restriction using BamHI and Esp3I enzymes.

Results. CRISPR/Cas9 system targeted against the gD gene of HSV-1 was transfected into HEK-AD cells that showed a significant reduction of HSV-1 infection by plaque assay and real-time PCR.

Conclusion. The pCas-Guide-EF1a-GFP CRISPR vector can create a fast and efficient method for gRNA cloning by restriction enzymes (Esp3I (BsmBI) and BamHI). Therefore, the CRISPR/Cas9 system may be utilized for the screening of genes critical for the HSV-1 infection and developing new strategies for targeted therapy of viral infections caused by HSV-1.

Abstract

Introduction. Herpes simplex virus type 1 (HSV-1) is a virus that causes serious human disease and establishes a long-term latent infection. The latent form of this virus has shown to be resistant to antiviral drugs. Clustered Regularly Interspace Short Palindromic Repeats (CRISPR), is an important tool in genome engineering and composed of guide RNA (gRNA) and Cas9 nuclease that makes an RNA-protein complex to digest exclusive target sequences implementation of gRNA. Moreover, CRISPR-Cas9 system effectively suppresses HSV-1 infection by knockout of some viral genes.

Materials and methods. To survey the efficacy of Cas9 system on HSV-1 genome destruction, we designed several guide RNAs (gRNAs) that all packaged in one vector. Additionally, we performed a one-step restriction using BamHI and Esp3I enzymes.

Results. CRISPR/Cas9 system targeted against the gD gene of HSV-1 was transfected into HEK-AD cells that showed a significant reduction of HSV-1 infection by plaque assay and real-time PCR.

Conclusion. The pCas-Guide-EF1a-GFP CRISPR vector can create a fast and efficient method for gRNA cloning by restriction enzymes (Esp3I (BsmBI) and BamHI). Therefore, the CRISPR/Cas9 system may be utilized for the screening of genes critical for the HSV-1 infection and developing new strategies for targeted therapy of viral infections caused by HSV-1.

Get Citation

Keywords

CRISPR; Cas9; gRNA; genome editing; HSV-1; HEK-AD cells

About this article
Title

A knockdown of the herpes simplex virus type-1 gene in all-in-one CRISPR vectors

Journal

Folia Histochemica et Cytobiologica

Issue

Vol 58, No 3 (2020)

Article type

Original paper

Pages

174-181

Published online

2020-09-16

Page views

1796

Article views/downloads

1354

DOI

10.5603/FHC.a2020.0020

Pubmed

32937678

Bibliographic record

Folia Histochem Cytobiol 2020;58(3):174-181.

Keywords

CRISPR
Cas9
gRNA
genome editing
HSV-1
HEK-AD cells

Authors

Nastaran Khodadad
Mona Fani
Saleh Jamehdor
Rahil Nahidsamiei
Manoochehr Makvandi
Saeed Kaboli
Ali Teimoori
Jose Thekkiniath

References (30)
  1. Chen YC, Sheng J, Trang P, et al. Potential Application of the CRISPR/Cas9 System against Herpesvirus Infections. Viruses. 2018; 10(6).
  2. Fani M, Khodadad N, Ebrahimi S, et al. Zinc Sulfate in Narrow Range as an In Vitro Anti-HSV-1 Assay. Biol Trace Elem Res. 2020; 193(2): 410–413.
  3. Liu G, Hai R, Liu F. Detection of congenital cytomegalovirus in newborns using nucleic acid amplification techniques and its public health implications. Virol Sin. 2017; 32(5): 376–386.
  4. Komor AC, Kim YB, Packer MS, et al. Programmable editing of a target base in genomic DNA without double-stranded DNA cleavage. Nature. 2016; 533(7603): 420–424.
  5. Long C, McAnally JR, Shelton JM, et al. Prevention of muscular dystrophy in mice by CRISPR/Cas9-mediated editing of germline DNA. Science. 2014; 345(6201): 1184–1188.
  6. Jinek M, Chylinski K, Fonfara I, et al. A programmable dual-RNA-guided DNA endonuclease in adaptive bacterial immunity. Science. 2012; 337(6096): 816–821.
  7. Barrangou R, Marraffini LA. CRISPR-Cas systems: Prokaryotes upgrade to adaptive immunity. Mol Cell. 2014; 54(2): 234–244.
  8. Jansen R, Embden JD, Gaastra W, et al. Identification of genes that are associated with DNA repeats in prokaryotes. Mol Microbiol. 2002; 43(6): 1565–1575.
  9. Makarova K, Koonin E. Annotation and Classification of CRISPR-Cas Systems. CRISPR. 2015: 47–75.
  10. Chylinski K, Makarova K, Charpentier E, et al. Classification and evolution of type II CRISPR-Cas systems. Nucleic Acids Research. 2014; 42(10): 6091–6105.
  11. Ran FA, Hsu PD, Wright J, et al. Genome engineering using the CRISPR-Cas9 system. Nat Protoc. 2013; 8(11): 2281–2308.
  12. Chang HHY, Pannunzio NR, Adachi N, et al. Non-homologous DNA end joining and alternative pathways to double-strand break repair. Nat Rev Mol Cell Biol. 2017; 18(8): 495–506.
  13. Chen C, Fenk LA, de Bono M. Efficient genome editing in Caenorhabditis elegans by CRISPR-targeted homologous recombination. Nucleic Acids Res. 2013; 41(20): e193.
  14. Graham FL, Smiley J, Russell WC, et al. Characteristics of a human cell line transformed by DNA from human adenovirus type 5. J Gen Virol. 1977; 36(1): 59–74.
  15. Wang T, Wei JJ, Sabatini DM, et al. Genetic screens in human cells using the CRISPR-Cas9 system. Science. 2014; 343(6166): 80–84.
  16. Labun K, Montague TG, Krause M, et al. CHOPCHOP v3: expanding the CRISPR web toolbox beyond genome editing. Nucleic Acids Res. 2019; 47(W1): W171–W174.
  17. Karpov DS, Karpov VL, Klimova RR, et al. A Plasmid-Expressed CRISPR/Cas9 System Suppresses Replication of HSV Type I in a Vero Cell Culture. Molecular Biology. 2019; 53(1): 70–78.
  18. Kabadi AM, Ousterout DG, Hilton IB, et al. Multiplex CRISPR/Cas9-based genome engineering from a single lentiviral vector. Nucleic Acids Res. 2014; 42(19): e147.
  19. Kunkel GR, Maser RL, Calvet JP, et al. U6 small nuclear RNA is transcribed by RNA polymerase III. Proc Natl Acad Sci U S A. 1986; 83(22): 8575–8579.
  20. Szybalski W, Kim SC, Hasan N, et al. Class-IIS restriction enzymes--a review. Gene. 1991; 100: 13–26.
  21. Zon LI, Dorfman DM, Orkin SH. The polymerase chain reaction colony miniprep. Biotechniques. 1989; 7(7): 696–698.
  22. Doench JG, Hartenian E, Graham DB, et al. Rational design of highly active sgRNAs for CRISPR-Cas9-mediated gene inactivation. Nat Biotechnol. 2014; 32(12): 1262–1267.
  23. Doench JG, Fusi N, Sullender M, et al. Optimized sgRNA design to maximize activity and minimize off-target effects of CRISPR-Cas9. Nat Biotechnol. 2016; 34(2): 184–191.
  24. Xu H, Xiao T, Chen CH, et al. Sequence determinants of improved CRISPR sgRNA design. Genome Res. 2015; 25(8): 1147–1157.
  25. Moreno-Mateos MA, Vejnar CE, Beaudoin JD, et al. CRISPRscan: designing highly efficient sgRNAs for CRISPR-Cas9 targeting in vivo. Nat Methods. 2015; 12(10): 982–988.
  26. Shen B, Zhang W, Zhang J, et al. Efficient genome modification by CRISPR-Cas9 nickase with minimal off-target effects. Nat Methods. 2014; 11(4): 399–402.
  27. Sanson KR, Hanna RE, Hegde M, et al. Optimized libraries for CRISPR-Cas9 genetic screens with multiple modalities. Nat Commun. 2018; 9(1): 5416.
  28. Kim HK, Min S, Song M, et al. Deep learning improves prediction of CRISPR-Cpf1 guide RNA activity. Nat Biotechnol. 2018; 36(3): 239–241.
  29. Hsu PD, Scott DA, Weinstein JA, et al. DNA targeting specificity of RNA-guided Cas9 nucleases. Nat Biotechnol. 2013; 31(9): 827–832.
  30. Bae S, Park J, Kim JS. Cas-OFFinder: a fast and versatile algorithm that searches for potential off-target sites of Cas9 RNA-guided endonucleases. Bioinformatics. 2014; 30(10): 1473–1475.

Regulations

Important: This website uses cookies. More >>

The cookies allow us to identify your computer and find out details about your last visit. They remembering whether you've visited the site before, so that you remain logged in - or to help us work out how many new website visitors we get each month. Most internet browsers accept cookies automatically, but you can change the settings of your browser to erase cookies or prevent automatic acceptance if you prefer.

By VM Media Group sp z o.o., ul. Świętokrzyska 73, 80–180 Gdańsk

tel.:+48 58 320 94 94, faks:+48 58 320 94 60, e-mail:  viamedica@viamedica.pl