CRISPR/Cas9-Mediated Genome Editing of Herpesviruses Limits Productive and Latent Infections

Herpesviruses infect the majority of the human population and can cause significant morbidity and mortality. Herpes simplex virus (HSV) type 1 causes cold sores and herpes simplex keratitis, whereas HSV-2 is responsible for genital herpes. Human cytomegalovirus (HCMV) is the most common viral cause of congenital defects and is responsible for serious disease in immuno-compromised individuals. Epstein-Barr virus (EBV) is associated with infectious mononucleosis and a broad range of malignancies, including Burkitt’s lymphoma, nasopharyngeal carcinoma, Hodgkin’s disease, and post-transplant lymphomas. Herpesviruses persist in their host for life by establishing a latent infection that is interrupted by periodic reactivation events during which replication occurs. Current antiviral drug treatments target the clinical manifestations of this productive stage, but they are ineffective at eliminating these viruses from the infected host. Here, we set out to combat both productive and latent herpesvirus infections by exploiting the CRISPR/Cas9 system to target viral genetic elements important for virus fitness. We show effective abrogation of HCMV and HSV-1 replication by targeting gRNAs to essential viral genes. Simultaneous targeting of HSV-1 with multiple gRNAs completely abolished the production of infectious particles from human cells. Using the same approach, EBV can be almost completely cleared from latently infected EBV-transformed human tumor cells. Our studies indicate that the CRISPR/Cas9 system can be effectively targeted to herpesvirus genomes as a potent prophylactic and therapeutic anti-viral strategy that may be used to impair viral replication and clear latent virus infection.

[1]  J. Yates,et al.  A vector that replicates as a plasmid and can be efficiently selected in B-lymphoblasts transformed by Epstein-Barr virus , 1985, Molecular and cellular biology.

[2]  G. Hayward,et al.  Sequence-specific DNA binding of the Epstein-Barr virus nuclear antigen (EBNA-1) to clustered sites in the plasmid maintenance region , 1985, Cell.

[3]  D. Reisman,et al.  A putative origin of replication of plasmids derived from Epstein-Barr virus is composed of two cis-acting components , 1985, Molecular and cellular biology.

[4]  J. Yates,et al.  Stable replication of plasmids derived from Epstein–Barr virus in various mammalian cells , 1985, Nature.

[5]  C. M. Preston,et al.  An in vitro latency system for herpes simplex virus type 2. , 1986, The Journal of general virology.

[6]  D. Reisman,et al.  trans activation of an Epstein-Barr viral transcriptional enhancer by the Epstein-Barr viral nuclear antigen 1 , 1986, Molecular and cellular biology.

[7]  C. M. Preston,et al.  Herpes simplex virus genes involved in latency in vitro. , 1987, The Journal of general virology.

[8]  I. Lehman,et al.  A DNA helicase induced by herpes simplex virus type 1. , 1988, Nucleic acids research.

[9]  L. Melton,et al.  Epidemiology of ocular herpes simplex. Incidence in Rochester, Minn, 1950 through 1982. , 1989, Archives of ophthalmology.

[10]  I. Lehman,et al.  Herpes simplex virus 1 helicase-primase: a complex of three herpes-encoded gene products. , 1989, Proceedings of the National Academy of Sciences of the United States of America.

[11]  U. Nater,et al.  Epstein-Barr virus. , 1991, The Journal of family practice.

[12]  S. Takada,et al.  Quantitative analysis of genomic polymorphism of herpes simplex virus type 1 strains from six countries: studies of molecular evolution and molecular epidemiology of the virus. , 1994, The Journal of general virology.

[13]  R. Samulski,et al.  Selective and rapid uptake of adeno-associated virus type 2 in brain. , 1998, Human gene therapy.

[14]  L. Hutt-Fletcher,et al.  Epstein-Barr Virus gH Is Essential for Penetration of B Cells but Also Plays a Role in Attachment of Virus to Epithelial Cells , 2000, Journal of Virology.

[15]  J. Rajčáni,et al.  Mechanisms of replication of alpha- and betaherpesviruses and their pathogenesis. , 2001, Bratislavske lekarske listy.

[16]  E. Robertson,et al.  OF KAPOSI ’ S SARCOMA-ASSOCIATED HERPESVIRUS , 2002 .

[17]  T. Shenk,et al.  Functional map of human cytomegalovirus AD169 defined by global mutational analysis , 2003, Proceedings of the National Academy of Sciences of the United States of America.

[18]  V. Štolc,et al.  Functional profiling of a human cytomegalovirus genome , 2003, Proceedings of the National Academy of Sciences of the United States of America.

[19]  Martin Kühne,et al.  A pathway of double-strand break rejoining dependent upon ATM, Artemis, and proteins locating to gamma-H2AX foci. , 2004, Molecular cell.

[20]  A. Izeta,et al.  Compartmentalization of VP16 in Cells Infected with Recombinant Herpes Simplex Virus Expressing VP16-Green Fluorescent Protein Fusion Proteins , 2004, Journal of Virology.

[21]  S. Kenney,et al.  Lytic Induction Therapy for Epstein-Barr Virus-Positive B-Cell Lymphomas , 2004, Journal of Virology.

[22]  P. Sharp,et al.  Cre-lox-regulated conditional RNA interference from transgenes. , 2004, Proceedings of the National Academy of Sciences of the United States of America.

[23]  Y. Stierhof,et al.  Human Cytomegalovirus Labeled with Green Fluorescent Protein for Live Analysis of Intracellular Particle Movements , 2005, Journal of Virology.

[24]  J. Tolar,et al.  A picornaviral 2A-like sequence-based tricistronic vector allowing for high-level therapeutic gene expression coupled to a dual-reporter system. , 2005, Molecular therapy : the journal of the American Society of Gene Therapy.

[25]  H. Mizuguchi,et al.  Eradication of Epstein-Barr virus episome and associated inhibition of infected tumor cell growth by adenovirus vector-mediated transduction of dominant-negative EBNA1. , 2005, Molecular therapy : the journal of the American Society of Gene Therapy.

[26]  E. Flemington,et al.  siRNAs against the Epstein Barr virus latency replication factor, EBNA1, inhibit its function and growth of EBV-dependent tumor cells. , 2006, Virology.

[27]  Andrew J Davison,et al.  Topics in herpesvirus genomics and evolution. , 2006, Virus research.

[28]  T. Shenk,et al.  Human Cytomegalovirus UL38 Protein Blocks Apoptosis , 2007, Journal of Virology.

[29]  D. Dittmer,et al.  Kaposi-Sarcoma-Associated Herpesvirus , 2008 .

[30]  V. Connor,et al.  Histone modifications associated with herpes simplex virus type 1 genomes during quiescence and following ICP0-mediated de-repression , 2008, The Journal of general virology.

[31]  E. Clercq,et al.  Viral DNA Polymerase Inhibitors , 2009 .

[32]  Andrew Rambaut,et al.  Evolutionary analysis of the dynamics of viral infectious disease , 2009, Nature Reviews Genetics.

[33]  C. Cameron,et al.  Viral genome replication , 2009 .

[34]  Gonçalo R. Abecasis,et al.  The Sequence Alignment/Map format and SAMtools , 2009, Bioinform..

[35]  R. Barrangou,et al.  CRISPR/Cas, the Immune System of Bacteria and Archaea , 2010, Science.

[36]  Kristen D Yetming,et al.  UL84-independent replication of human cytomegalovirus strain TB40/E. , 2010, Virology.

[37]  N. DeLuca,et al.  Activities of ICP0 Involved in the Reversal of Silencing of Quiescent Herpes Simplex Virus 1 , 2011, Journal of Virology.

[38]  E. Kieff,et al.  Roscovitine Inhibits EBNA1 Serine 393 Phosphorylation, Nuclear Localization, Transcription, and Episome Maintenance , 2011, Journal of Virology.

[39]  M. Nicoll,et al.  The molecular basis of herpes simplex virus latency , 2012, FEMS microbiology reviews.

[40]  Steven L Salzberg,et al.  Fast gapped-read alignment with Bowtie 2 , 2012, Nature Methods.

[41]  J. Doudna,et al.  A Programmable Dual-RNA–Guided DNA Endonuclease in Adaptive Bacterial Immunity , 2012, Science.

[42]  J. Doudna,et al.  RNA-guided genetic silencing systems in bacteria and archaea , 2012, Nature.

[43]  Rudolf Jaenisch,et al.  One-Step Generation of Mice Carrying Mutations in Multiple Genes by CRISPR/Cas-Mediated Genome Engineering , 2013, Cell.

[44]  Yongxiang Zhao,et al.  Heritable gene targeting in the mouse and rat using a CRISPR-Cas system , 2013, Nature Biotechnology.

[45]  R. Jaenisch,et al.  One-Step Generation of Mice Carrying Reporter and Conditional Alleles by CRISPR/Cas-Mediated Genome Engineering , 2013, Cell.

[46]  Paul D. Shaw,et al.  Using Tablet for visual exploration of second-generation sequencing data , 2013, Briefings Bioinform..

[47]  Le Cong,et al.  Multiplex Genome Engineering Using CRISPR/Cas Systems , 2013, Science.

[48]  Yoshio Koyanagi,et al.  Harnessing the CRISPR/Cas9 system to disrupt latent HIV-1 provirus , 2013, Scientific Reports.

[49]  S. Y. Kim,et al.  Characterization of naturally Epstein-Barr virus-infected gastric carcinoma cell line YCCEL1. , 2013, The Journal of general virology.

[50]  G. Church,et al.  Cas9 as a versatile tool for engineering biology , 2013, Nature Methods.

[51]  T. Flotte Birth of a new therapeutic platform: 47 years of adeno-associated virus biology from virus discovery to licensed gene therapy. , 2013, Molecular therapy : the journal of the American Society of Gene Therapy.

[52]  James E. DiCarlo,et al.  RNA-Guided Human Genome Engineering via Cas9 , 2013, Science.

[53]  David A. Scott,et al.  Double Nicking by RNA-Guided CRISPR Cas9 for Enhanced Genome Editing Specificity , 2013, Cell.

[54]  J. Keith Joung,et al.  High frequency off-target mutagenesis induced by CRISPR-Cas nucleases in human cells , 2013, Nature Biotechnology.

[55]  Geoffrey M. Hill,et al.  Herpes simplex keratitis. , 2014, Disease-a-month : DM.

[56]  R. Cohrs,et al.  Varicella Zoster Virus , 2014 .

[57]  Mazhar Adli,et al.  Genome-wide analysis reveals characteristics of off-target sites bound by the Cas9 endonuclease , 2014, Nature Biotechnology.

[58]  G. Gao,et al.  State-of-the-art human gene therapy: part II. Gene therapy strategies and clinical applications. , 2014, Discovery medicine.

[59]  V. Barbaro,et al.  Gene transfer of integration defective anti-HSV-1 meganuclease to human corneas ex vivo , 2014, Gene Therapy.

[60]  S. Schaffert,et al.  Precise gene deletion and replacement using the CRISPR/Cas9 system in human cells. , 2014, BioTechniques.

[61]  G. Gao,et al.  State-of-the-art human gene therapy: part I. Gene delivery technologies. , 2014, Discovery medicine.

[62]  E. Kieff,et al.  Small molecule inhibition of Epstein-Barr virus nuclear antigen-1 DNA binding activity interferes with replication and persistence of the viral genome. , 2014, Antiviral research.

[63]  J. Keith Joung,et al.  Improving CRISPR-Cas nuclease specificity using truncated guide RNAs , 2014, Nature Biotechnology.

[64]  S. Quake,et al.  RNA-guided endonuclease provides a therapeutic strategy to cure latent herpesviridae infection , 2014, Proceedings of the National Academy of Sciences.

[65]  Michael T. McManus,et al.  A high-coverage shRNA screen identifies TMEM129 as an E3 ligase involved in ER-associated protein degradation , 2014, Nature Communications.

[66]  David Bryder,et al.  Efficient ablation of genes in human hematopoietic stem and effector cells using CRISPR/Cas9. , 2014, Cell stem cell.

[67]  T. Suenaga,et al.  Engineering large viral DNA genomes using the CRISPR‐Cas9 system , 2014, Microbiology and immunology.

[68]  Qihan Li,et al.  High-Efficiency Targeted Editing of Large Viral Genomes by RNA-Guided Nucleases , 2014, PLoS pathogens.

[69]  David A. Scott,et al.  Genome-wide binding of the CRISPR endonuclease Cas9 in mammalian cells , 2014, Nature Biotechnology.

[70]  G. Boivin,et al.  Antiviral drug resistance in herpesviruses other than cytomegalovirus , 2014, Reviews in medical virology.

[71]  Meagan E. Sullender,et al.  Rational design of highly active sgRNAs for CRISPR-Cas9–mediated gene inactivation , 2014, Nature Biotechnology.

[72]  Yoshimitsu Takahashi,et al.  In vitro and in vivo growth suppression of human papillomavirus 16-positive cervical cancer cells by CRISPR/Cas9. , 2014, Biochemical and biophysical research communications.

[73]  A. Scharenberg,et al.  In vitro Inactivation of Latent HSV by Targeted Mutagenesis Using an HSV-specific Homing Endonuclease , 2014, Molecular therapy. Nucleic acids.

[74]  Bryan R. Cullen,et al.  Inactivation of the Human Papillomavirus E6 or E7 Gene in Cervical Carcinoma Cells by Using a Bacterial CRISPR/Cas RNA-Guided Endonuclease , 2014, Journal of Virology.

[75]  E. Lander,et al.  Development and Applications of CRISPR-Cas9 for Genome Engineering , 2014, Cell.

[76]  M. Luftig,et al.  Dynamic Epstein-Barr virus gene expression on the path to B-cell transformation. , 2014, Advances in virus research.

[77]  Fan Yang,et al.  RNA-directed gene editing specifically eradicates latent and prevents new HIV-1 infection , 2014, Proceedings of the National Academy of Sciences.

[78]  B. Cullen,et al.  Suppression of hepatitis B virus DNA accumulation in chronically infected cells using a bacterial CRISPR/Cas RNA-guided DNA endonuclease. , 2015, Virology.

[79]  Hans Clevers,et al.  Efficient Intracellular Delivery of Native Proteins , 2015, Cell.

[80]  K. Khalili,et al.  CRISPR/Cas9 System as an Agent for Eliminating Polyomavirus JC Infection , 2015, PloS one.

[81]  J. Englund,et al.  Cytomegalovirus in immunocompromised children , 2015, Current opinion in infectious diseases.

[82]  David S. Weiss,et al.  Cas9-mediated targeting of viral RNA in eukaryotic cells , 2015, Proceedings of the National Academy of Sciences.

[83]  D. Tscharke,et al.  Engineering herpes simplex viruses by infection-transfection methods including recombination site targeting by CRISPR/Cas9 nucleases. , 2015, Journal of virological methods.

[84]  Martin J. Aryee,et al.  Engineered CRISPR-Cas9 nucleases with altered PAM specificities , 2015, Nature.

[85]  J. Doudna,et al.  Expanding the Biologist's Toolkit with CRISPR-Cas9. , 2015, Molecular cell.

[86]  J. Keith Joung,et al.  Efficient Delivery of Genome-Editing Proteins In Vitro and In Vivo , 2014, Nature Biotechnology.

[87]  Keiichiro Suzuki,et al.  Use of the CRISPR/Cas9 system as an intracellular defense against HIV-1 infection in human cells , 2015, Nature Communications.

[88]  Won-Bin Young,et al.  CRISPR/gRNA-directed synergistic activation mediator (SAM) induces specific, persistent and robust reactivation of the HIV-1 latent reservoirs , 2015, Scientific Reports.

[89]  Honglin Chen,et al.  CRISPR/Cas9-mediated genome editing of Epstein-Barr virus in human cells. , 2015, The Journal of general virology.

[90]  David A. Scott,et al.  Rationally engineered Cas9 nucleases with improved specificity , 2015, Science.

[91]  J. Joung,et al.  High-fidelity CRISPR-Cas9 variants with undetectable genome-wide off-targets , 2015, Nature.

[92]  J. Karn,et al.  Elimination of HIV-1 Genomes from Human T-lymphoid Cells by CRISPR / Cas 9 Gene Editing SUPPLEMENTARY MATERIALS , 2016 .

[93]  J. Keith Joung,et al.  731. High-Fidelity CRISPR-Cas9 Nucleases with No Detectable Genome-Wide Off-Target Effects , 2016 .

[94]  Derrick J Chen,et al.  Epstein-Barr Virus , 2016, Succinct Pediatrics: Evaluation and Management of Infectious Diseases and Dermatology.