Nothing Special   »   [go: up one dir, main page]

CRISPR Cas Based Antiviral Strategy

Download as pdf or txt
Download as pdf or txt
You are on page 1of 15

HHS Public Access

Author manuscript
Semin Cell Dev Biol. Author manuscript; available in PMC 2020 December 01.
Author Manuscript

Published in final edited form as:


Semin Cell Dev Biol. 2019 December ; 96: 53–64. doi:10.1016/j.semcdb.2019.04.004.

CRISPR-Cas Based Targeting of Host and Viral Genes as an


Antiviral Strategy
Lulia Koujah1,2, Deepak Shukla1,2,*, Afsar R. Naqvi3,*
1Department of Microbiology and Immunology, University of Illinois at Chicago, Illinois, USA,
60612.
2Department of Ophthalmology and Visual Sciences, University of Illinois Medical Center,
Author Manuscript

Chicago, Illinois, USA, 60612.


3MucosalImmunology Lab, College of Dentistry, University of Illinois at Chicago, Chicago, Illinois,
USA, 60612.

Abstract
Viral infections in human are leading cause of mortality and morbidity across the globe. Several
viruses (including HIV and Herpesvirus), have evolved ingenious strategies to evade host-immune
system and persist life-long. Clustered Regularly Interspaced Short Palindromic Repeats
(CRISPR) and CRISPR-associated (Cas) is an ancient antiviral system recently discovered in
bacteria that has shown tremendous potential as a precise, invariant genome editing tool. Using
CRISPR-Cas based system to activate host defenses or genetic modification of viral genome can
Author Manuscript

provide novel, exciting and successful antiviral mechanisms and treatment modalities. In this
review, we will provide progress on the CRISPR-Cas based antiviral approaches that facilitate
clearance of virus-infected cells and/or prohibit virus infection or replication. We will discuss on
the possibilities of CRIPSR-Cas as prophylaxis and therapy in viral infections and review the
challenges of this potent gene editing technology.

Keywords
CRISPR-Cas; Viruses; Antiviral immunity; Gene editing; Restriction factors
Author Manuscript

*
Corresponding Author: Dr. Afsar R. Naqvi, University of Illinois at Chicago, College of Dentistry, Department of Periodontics, 561B
Dent MC 859, 801 South Paulina, Chicago, Illinois, 60612., afsarraz@uic.edu; Dr. Deepak Shukla, University of Illinois at Chicago,
Department of Ophthalmology, LIERI Room 233 MC 648, 1855 W. Taylor Street, Chicago, Illinois, 60612, dshukla@uic.edu.
Author Contributions
LK, DS and ARN conceptualized the review; LK and ARN prepared all the figures and compiled the tables; LK. DS and ARN wrote
the manuscript.
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our
customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of
the resulting proof before it is published in its final citable form. Please note that during the production process errors may be
discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
Conflict of Interest Statement
The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be
construed as a potential conflict of interest.
Koujah et al. Page 2

1. Introduction
Author Manuscript

1.1 Genome Engineering


Genetic engineering is a powerful technology that has resulted in the advancements of many
fields such as medicine, industrial biotechnology and agriculture [1,2]. Specifically, the
ability to alter genetic makeup of an organism has revolutionized medical research and
expanded our ability to understand disease development and prevention. The application of
various genome engineering tools such as zinc finger nucleases (ZFN), transcription
activator-like effector nucleases (TALENs) and, most recently, CRISPR-Cas systems have
advanced biological research and allowed for novel discoveries to be made.

ZFN and TALEN are both synthetic proteins with nucleus activity that achieve specific DNA
binding via protein-DNA interactions. However, retargeting the specific activity of these
nucleases is achieved through protein engineering or cloning which limits its versatility,
Author Manuscript

feasibility and application. Recently, harnessing the CRISPR-Cas system, initially observed
in prokaryotes, as a gene editing tool in eukaryotic cells has revolutionized the field as a
robust and efficient tool for precise genome editing [3-5]. CRISPR/Cas system is a
prokaryotic adaptive immune system that confers resistance to foreign genetic elements and
has been repurposed by researchers as an invariant genome editing tool that can be more
easily redirected to a new target [6].

The use of genome editing technology, primarily CRISPR, is reaching exciting stages and
surpassing milestones in basic science research with applications extending beyond genome
editing. More recently, CRISPR is being used as an antiviral by targeting genes in both the
human genome as well as the invading viral genome. Both in vitro and in vivo studies have
shown effective targeting of key viral genes important for the virus life cycle as well as host
Author Manuscript

genes involved in viral entry, replication and persistence [7].

1.2 CRISPR/Cas9: Precise genome editing


CRISPR is an abbreviation for clustered regularly interspaced short palindromic repeats,
which were initially observed in Escherichia coli, and found to be separated by non-repeated
DNA sequences called spacers. These spacers, known as CRISPR arrays, where later
identified as acquired copies of previously encountered foreign DNA and provides the
organism with memory of the plasmid and phage genomes [8-10]. Upon encountering the
foreign genetic element again, the bacteria produce RNA segments from the CRISPR arrays
to target the pathogen directly and employ an adaptive immune response. Adjacent to
CRISPR arrays are well conserved CRISPR-associated (Cas) genes that have been grouped
into families and subtypes based on sequence similarity of encoded proteins [11]. There are
Author Manuscript

six types of CRISPR/Cas system that have been divided into two classes: class 1 CRISPR-
Cas systems (types I, III, and IV) employ multi-Cas protein complexes for interference,
whereas in class 2 systems (types II, V, and VI), interference is accomplished by a single
effector protein [12].

Type II CRISPR/Cas9 system of Streptococcus pyogenes (Sp) is the most studied and used
system. Figure 1 describes generalized biogenesis and function of CRISPR/Cas9 system.
The CRISPR array, which contains unique protospacer sequences that have homology to

Semin Cell Dev Biol. Author manuscript; available in PMC 2020 December 01.
Koujah et al. Page 3

foreign DNA, is transcribed to make long precursor CRISPR RNA (pre-crRNA). Pre-crRNA
are then processed to individual crRNA which guide the interference machinery to cleave
Author Manuscript

complementary sequences or protospacers and ultimately eliminate the foreign DNA [13].
An invariant trans-activating CRISPR RNA (tracrRNA) is required to bind crRNA in a
sequence complementary manner and recruit RNAse III and CRISPR-associated 9 (Cas9)
enzymes [14]. Together, tracrRNA, RNAse III and Cas9 break up and form a complex with
each individual unique crRNAs [15]. Protospacer adjacent motifs (PAMs), located directly
after where the crRNA would bind, are critical to this system and contribute to the CRISPR
targeting specificity [16]. Cas protein motifs have helicase and nuclease activity that are
guided by crRNA resulting in DNA cleavage upon target binding, which stimulates non-
homologous end joining (NHEJ) or homology-directed repair (HDR)-mediated genome
editing, resulting in the elimination of invading foreign DNA [17-19].

Cas9 effector endonuclease have been repurposed by researchers as a tool for precise gene
Author Manuscript

editing to eliminate or integrate target genes in the human genome [6,20]. Specifically, the
tracrRNA:crRNA complex have been engineered into a single guide RNA (sgRNA), guiding
CRISPR-Cas9 to desired targets in the genome [21]. The use of this system and its gene
editing potential in eukaryotic cells proved to be successful and the application of CRIPSR/
Cas9 is now widely adapted in multiple research areas [5,6,20,22].

1.3 The need for a new antiviral: CRISPR/Cas9


Multiple viruses still persist and infect a large number of the human population while still
without a cure [23-26]. Current antiviral treatments are able to block active viral replication
or subside symptoms of infection; however, they lack the ability to completely abolish the
virus from the host, primarily due to escape mutants or establishment of latent infection
[27-29]. CRISPR/Cas system has been adapted from its natural function as an immune
Author Manuscript

response in prokaryotes against phage infection to studying its gene editing potential in
eukaryotes. This application has extended to the development of a novel antiviral; where
CRISPR/Cas systems are directed to target viral genome or host genes that aide in the
persistence of the virus [30,31]. In this review we will discuss these approaches that have
been studied as well as other potential applications against viruses including Herpesvirus,
Human immunodeficiency virus (HIV), Hepatitis B Virus (HBV) and Influenza A Virus
(IAV). Different CRISPR/Cas based antiviral strategies targeting host and viral genomes are
illustrated in Figure 2.

Of the viruses listed, herpesvirus has been recognized as the most adapted human pathogen
resulting in lifelong infections and the development of a wide range of innocuous diseases
[32-34]. The Herpesviridae family is divided into three subfamilies of double stranded DNA
Author Manuscript

viruses: Alpha-, Beta- and Gammaherpesvirinae [35]. Herpes simplex virus-1 and 2 (HSV-1
and HSV-2) are neurotropic viruses belonging to the Alphaherpesvirinae family and are
capable of establishing lifelong latency in the central nervous system. HSV-2 is responsible
for genital herpes resulting in painful ulcers while HSV-1 primarily infects the corneal
epithelium resulting in lesions and the development herpes simplex keratitis as well as other
less frequent diseases such as encephalitis [36-39]. Human Cytomegalovirus (HCMV),
belongs to the Betaherpesvirinae family, is capable of infecting multiple cell types including

Semin Cell Dev Biol. Author manuscript; available in PMC 2020 December 01.
Koujah et al. Page 4

epithelial, endothelial, myeloid, neuronal and fibroblast cells [40]. Epstein–Barr virus (EBV)
belongs to the Gammaherpesvirinae family, primarily infecting B cells and epithelial cells
Author Manuscript

causing infectious mononucleosis [41]. Acyclovir and Ganciclovir, nucleoside analogs that
inhibit viral replication upon incorporation into viral DNA, are the current drug choice for
treatment of HSV and CMV disease respectively [42,43]. While these drugs are highly
effective in most cases, drug resistant viral strains have evolved [44-46]. On the other hand,
EBV is reported to infect 95% of the population and no current treatment is available [47].

Another prominent virus, Hepatitis B Virus (HBV), remains a global health threat as
infection results in life-threatening complications including elevated risks for liver damage
and liver cancer [48]. HBV belongs to the family of Hepadnaviridae and replicates in
infected hepatocytes in the liver via reverse transcription. HBV virions contain a partial
dsDNA genome which is converted to double-stranded covalently closed circular (ccc)DNA
in the nucleus shortly after infection [49]. The cccDNA is maintained as a
Author Manuscript

‘minichromosome’ and serves as a template for transcription of viral pregenomic RNA and
mRNAs [50]. Current antivirals against HBV include IFN-α, which bolsters the host
antiviral immune response, and nucleoside/nucleotide analogues to inhibit reverse
transcriptase activity. However, due to persistence of cccDNA, long-term treatment is
required is not a feasible option for some patients and has resulted in drug resistance among
some strains [51].

Human Immunodeficiency virus (HIV) is also among the viruses that are highly prevalent
worldwide and still remain without a cure. HIV is a single-stranded enveloped RNA virus
that predominately infects human T cells and results in the development of acquired immune
deficiency syndrome (AIDS). An estimated number of 35 million individuals suffer from
HIV infection worldwide and require life-long treatment, which has resulted in many side
Author Manuscript

effects and the emergence of escape mutants [52,53]. Antiretroviral therapy is the current
mode of treatment and while it does efficiently limit virus replication to undetectable levels
in HIV patients, it suffers from serious drawbacks [54].

Influenza A virus (IAV) is considered the most serious influenza capable of causing
widespread outbreaks and disease [55]. IAV infection attacks the respiratory system and is
able to rapidly evolve, resulting in seasonal epidemics and the constant emergence of
variant- and drug-resistant strains [56]. The genome of IAV, which consists of multiple
segments of single-stranded negative-sense RNA, contributes to its susceptibility to
mutations which allow for evasion of the immune system and resistance to antiviral
strategies [57]. Current vaccinations are limited in their efficacy and therapies only alleviate
the symptoms and manage disease development to prevent hospitalization and death in
patients [58].
Author Manuscript

The conventional antiviral drugs pose many concerning disadvantages. First of all, resistance
among infected patients for all current FDA approved antiviral drugs, especially in the case
of IAV, have been repeatedly reported [27,58-60]. Also, while current antivirals are effective
in clearing productive viral replication in most cases, they are unable to eradicate the virus
from latently infected cells [61,62]. This causes the infection to persist throughout the
lifetime of the host with the potential of reactivation and recurrence. For these cases, life-

Semin Cell Dev Biol. Author manuscript; available in PMC 2020 December 01.
Koujah et al. Page 5

long treatments are prescribed to patients in order to suppress viral reactivation, which
results in side effects and escape mutants in many cases.
Author Manuscript

The viruses mentioned here remain a global health threat and continued efforts are aimed
towards developing an effective antiviral or vaccination that eliminates viral genomes from
the host. Most efforts to develop antivirals in the past have been directed towards targeting
the viral genome in order to dismantle the virus’ ability to infect. However, with the
emergence of escape mutants and drug resistance, efforts are now also being directed
towards targeting the host cell machinery [31,63]. To prevent viral reactivation, drug
resistance and protein expression, new antiviral strategies are needed that provide a durable
and feasible antiviral effect. Ideally, eradication of the integrated proviral DNA in all
infected cells, including the latent reservoir is required to effectively combat viral infection.

2. Targeting Host Factors Using CRISPR/Cas9


Author Manuscript

While targeting viral genomes with CRISPR/Cas9 may be subjected to the same
disadvantages as current therapeutics, targeting host factors provides a more promising
solution to abolish the virus from the host. Clinical advantages of host-based interventions
using CRISPR/Cas9 include lower chances of drug resistances due to genetic stability of
host factors, higher efficacy due to larger therapeutic timeframe and reduced side effects due
to minimal dosing requirements.

Upon infection, the virus exploits host factors to complete its life cycle [64]. While the
host’s innate immune system works to clear viral infection, the virus hijacks cell signaling
pathways to escape immune surveillance and persist. Exploiting the dynamic virus-host
interactions is a strategy used to block virus infection and make the virus less susceptible to
survival in the cellular environment [65]. CRISPR/Cas systems have been used in multiple
Author Manuscript

studies for genome editing of dispensable target host genes involved in viral replication and
persistence. Recent advancements in CRISPR/Cas9 technology has also allowed for
genome-wide CRISPR knockout (GeCKO) screening, providing a powerful tool to discover
host factors and to highlight biological pathways essential for the replication of intracellular
pathogens [66].

2.1 Host factors involved in viral entry


While current drug targets include viral proteins required for cell entry, most of the
developed drugs have been regarded as ineffective over long term control of infection. In an
effort to produce antivirals with long-lasting effects, reduced risks of developed resistance
and toxicity, new mechanisms of action need to be developed. With the recent advancements
made in gene editing, CRISPR/Cas9 and other tools have been used to target both the viral
Author Manuscript

and host genome for factors involved in viral entry (Table 1). Recent studies have focused on
inhibiting viral entry by targeting host factors such as cell surface receptors. Results from
these studies have shown been high antiviral efficacy against HIV and IAV.

2.1.1 Targeting HIV cell-surface entry receptors—CC chemokine receptor 5


(CCR5) and C-X-C chemokine receptor type 4 (CXCR4) are cell surface receptors involved
in viral entry during primary infection [67]. Individuals homozygous for a naturally

Semin Cell Dev Biol. Author manuscript; available in PMC 2020 December 01.
Koujah et al. Page 6

occurring deletion of 32 nt in the CCR5 gene prevents the expression of CCR5 on the cell
surface and deems them highly resistant to HIV infection [68]. This discovery proposed a
Author Manuscript

new antiviral approach to combat HIV: targeting cell surface entry receptors in order to
disrupt membrane fusion between the virus and cell [69]. A competitive inhibitor of CCR5,
maraviroc, was clinically approved and showed affective anti-HIV activity. However,
development of resistance by selection for escape mutants was later observed by viral
adaptations to the drug [70]. While CCR5 is the predominant receptor involved in HIV entry
and primary infection, viral tropism towards CXCR4 usage occurs at later stages of infection
and maraviroc was also dispensable in the presence of CXCR4-tropic HIV strains [71]. To
combat CCR5-tropic HIV strains, gene therapy approaches to block CCR5 expression were
evaluated and found to be promising both in vitro and in vivo [72]. Perez and colleagues
published a study using ZFN-guided genomic editing of CCR5 in primary human CD4+ T
lymphocytes showing high specificity, tolerance and robust protection against HIV-1 and
soon underwent clinical trials targeting mature CD4+ T cells [73,74].
Author Manuscript

More interest has been developed in the past decade following a report that an acute myeloid
leukemia patient who received a hematopoietic stem/progenitor cell (HSC) transplant from a
CCR5Δ32 donor was also cured from HIV-1 [75,76]. Subsequent studies in mice
transplanted with CCR5-disrupted HSCs showed significant reduction in viral load when
challenged with CCR5-tropic HIV-1 strain, as well as protection of human T cell
populations in key tissues that HIV-1 infects. Long-term control of HIV-1 replication was
observed for the first time using this technique [77]. This study suggested that modification
of autologous HSCs by CCR5-ZFNs could provide a permanent supply of HIV-resistant
progeny, leading to immune reconstitution and long-term control of viral replication [78].
While ZFN and TALEN are effective in enabling mutagenesis in a variety of cells, they both
exhibit challenges including variability in mutation frequencies, lack of specificity,
Author Manuscript

feasibility and off-target effects. The most important off-target sites associated with CCR5-
specific ZFNs and TALENs reside in CCR2, a close homolog of CCR5, located 15kpb
upstream of CCR5.

In order to increase specificity and reduce undesired off-target effects, CRISPR/Cas9 has
been employed to induce a site-specific genome modification in human cells and in vivo
using mouse model of infection [79-82]. Mulitiple groups successfully transduced primary
CD4+ T-lymphocytes and disrupted CCR5 by gene editing using adenovirus-delivered
CRISPR/Cas9 targeting the open reading frames (ORF) of CCR5. This resulted in inhibition
of HIV-1 infection with no significant off-target effects [81]. Targeting CCR5 in both HSC
and CD4+ T cells has shown promise as a gene-editing strategy for persistent resistance to
HIV. However, its ineffectiveness towards CXCR4-tropic HIV strains and therefore lacks
Author Manuscript

conclusive evidence for its use as a cure. Studies have also directed Cas9-mediated gene
editing of CXCR4 with high precision and efficacy, showing negligible off-target effects and
resistance to HIV infection [79,83]. Targeting both coreceptors CCR5 and CXCR4 may
provide more breadth in protection and use in clinical settings.

2.1.2 Targeting host factors involved in IAV entry—CRISPR/Cas9 has also been
used to limit IAV infection by targeting host factors involved in viral entry. IAV enters host
cells after attachment of its hemagglutinin (HA) to the cell surface receptor sialic acid. Entry

Semin Cell Dev Biol. Author manuscript; available in PMC 2020 December 01.
Koujah et al. Page 7

inhibitors have been developed in the case of IAV that either bind to the HA globular head to
prevent interaction with the receptor or by acting on the sialic acid receptor itself [84,85].
Author Manuscript

However, currently available drugs are ineffective against newly emerging viruses with
developed resistance. Recent studies using a genome-wide CRISPR knockout (GeCKO)
screen demonstrated that a CMP-sialic acid transporter (SLC35A1) is an essential host
factor for IAV entry [86]. Validation studies revealed that loss of SLC35A1 resulted in
resistance against IAV infection due to the absence of cell-surface sialic acid. Clonal
knockouts infected with H5N1, H1N1 and H3N2 observed a significant reduction in viral
titers, confirming the role of SLC35A1 in replication of multiple IAV strains. SLC35A1
knockout cell lines also showed inability to support recombinant HA binding and therefore
resulting in a robust reduction of IAV entry.

Genome wide overexpression screen using CRISPR/dCas9 was recently used as a tool to
uncover IAV restriction factors that, when overexpressed, are sufficient to prevent infection.
Author Manuscript

A glycosyltransferase (B4GALNT2) that modifies sialic acid-containing glycans, and thus


prevents IAV infection, was identified as a top hit, leading to greater than 100-fold reduction
in IAV infection. The inhibitory activity of B4GALNT2 was restricted to viruses with avian
a2,3-linked receptor which include avian influenza viruses H7N2, H9N2, H5N9, H10N4,
some of which infect humans as well [87].

2.2 Antiviral Host Effectors (Cell-intrinsic immunity)


Upon viral infection, the host triggers a robust innate immune response as a defense
mechanism to clear the pathogen [64]. The dynamic virus-host interaction that occurs at
early stages of infection determines whether the cell will overcome the viral infection or
succumb to it. While the host cells have evolved to express restriction factors that block
virus infection, the virus in turn exploits host factors and uses specific strategies to escape
Author Manuscript

immune surveillance by manipulating its environment. Studies of this dynamic interaction


has allowed for the identification of host effectors that aid or suppress the cell-intrinsic
immunity. These host effectors are now key targets for developing novel antiviral strategies
through the use of CRISPR/Cas9.

In regard to HIV, APOBEC3G (A3G) and APOBEC3B (A3B) are restriction factors in
human cells that have recently been targeted. A3G is expressed in human T cells and
macrophages, which are usually the target cells of HIV-1, and is capable of restricting the
virus. However, HIV-1 Vif protein is able to target and degrade this host effector. A3B on the
other hand is restricted to expression in pluripotent cell types and is not targeted for
degradation by Vif. In a recent study, a gain-of-function approach using a cleavage defective
Cas9 (dCas9) allowed for specific and effective induction of A3G and A3B genes in cell
Author Manuscript

culture [88]. Potent inhibition of HIV-1ΔVif infectivity was achieved by induced


endogenous A3B and A3G in 293 T cells, which typically lack both proteins, and a decrease
in HIV-1WT infectivity upon A3B expression. This study shows that CRISPR/Cas gene
editing technology can be used as a novel platform to combat infection by efficiently
targeting human genes that regulate virus replication.

Using a genome-wide CRISPR/Cas9 screen, key regulators of cell-intrinsic immunity upon


IAV infection have been identified and serve as targets for further investigation [86]. In this

Semin Cell Dev Biol. Author manuscript; available in PMC 2020 December 01.
Koujah et al. Page 8

study, capicua (CIC) was shown to play a role in suppressing antiviral gene expression by its
transcriptional repressor activity. Confirming this, knocking down CIC using CRISPR/Cas9
Author Manuscript

resulted in decreased replication of both IAV and VSV by the observed heightened antiviral
responses. Similarly, JAK2 was also identified as a host effector involved in negatively
regulating cell-intrinsic immunity by lowering antiviral gene expression. While more studies
are required to elucidate the exact role JAK2 plays at different stages of IAV infection, using
CRISPR/Cas9 this group was able to identify key factors that contribute to virus survival
against the innate immune system.

Similarly, a genome-wide CRISPR/Cas9 loss-of-function screen was conducted to identify


host effectors critical for EBV induced B cell-transformation leading to B cell malignancies
and how oncoproteins evade tumor suppressor responses [89]. The screen identified BATF,
IRF4, IRF2, CDK6 and cyclin D2 as lymphoblastoid cell lines (LCLs) dependency factors
that are highly upregulated in EBV infection of primary human B cells. Another gene,
Author Manuscript

cFLIP, was highlighted as a potential EBV therapeutic target for its role as a survival factor
in LCLs by blocking TNFα-mediated apoptosis and necroptosis.

Other host factors recently identified as having an integral role in viral infection are
interferon-induced transmembrane proteins (IFITM) which restrict the entry of diverse
enveloped viruses through unknown mechanism [90,91]. In the case of IAV, initial infection
upregulates p53, a protein involved in key cellular stress mechanisms, which in turn
downregulates IFITMs and therefore promotes virus survival in the host. Since IFITMs were
detected as downstream targets that are negatively regulated by p53, CRISPR/Cas9 was used
to generate p53null cells to investigate its role in influenza infectivity. While p53 was not
shown to affect initial IAV entry, overexpression of IFITMs reduced cell susceptibility to
IAV infection by blocking viral entry in A549 cells [92]. The role of IFITM on restricting
Author Manuscript

HIV-1 infection was also investigated using CRISPR/Cas9 by directly targeting IFITMs in
U87 CD4+ CCR5+ cells. While similar findings as those regarding IAV were concluded
from the study, the clear mechanism of IFITM upon viral infection is still poorly understood
and lacks in vivo relevance [93].

2.3 Host Factors involved in viral transcription and replication


Infectious particles take over various cellular pathways and processes in order to ensure its
survival and persistence in the host. Certain cellular genes are upregulated in the presence of
viral particles and aid in viral transcription and replication. For example, genes known to
support HBV replication include sodium-taurocholate cotransporting polypeptide (NTCP),
heat stress cognate 70 (Hsc70), heat shock protein Hsp90 and tyrosyl-DNA-
phosphodiesterase 2 (TDP2) [31]. While studies have not shown the antiviral effect of
Author Manuscript

knocking down these genes using CRISPR/Cas9, these are still potential targets to be
considered.

Various host factors identified as restricting HIV replication include APOBEC3C, TRIM5α,
SAMHD1, and tetherin, and also serve as potential CRISPR/Cas targets in developing
therapeutics [31,94,95]. Many of these are involved in eliciting robust immune responses
upon detection of HIV, and ultimately acting as restriction factors to terminate HIV
replication by blocking viral entry or release from the host cell. TRIM5α is associated with

Semin Cell Dev Biol. Author manuscript; available in PMC 2020 December 01.
Koujah et al. Page 9

antiviral activity by recognizing retroviral capsid proteins and, in turn, transducing diverse
cellular signaling pathways, including triggering innate immunity [95]. Studies showed
Author Manuscript

efficient antiviral activity using TRIM-cyclophilin fusion protein both in vitro and in vivo
with robust inhibition of HIV-1 replication [96]. Similarly, SAMHD1 is a restriction factor
that prevents HIV replication. SAMHD1 is a deoxynucleotide triphosphate (dNTP)
hydrolase that is activated by the binding of GTP to its allosteric site which consequently
reduces the intracellular dNTP pool below levels that support HIV-1 reverse transcription
[97]. This results in the blockage of HIV-1 replication and prevents the induction of IFN
responses. However, transducing the activity of SAMHD1 is counteracted by Viral Protein X
(VpX) which targets and degrades the protein, leading to the restoration of dNTP levels and
allowing for HIV-1 replication [98,99]. Tetherin is another restriction factor of HIV induced
by interferon and prevents viral replication by restricting viral particle release [100]. While
HIV-1 Viral Protein U (Vpu) counteracts tetherin activity, a single amino acid substitution in
tetherin results in Vpu-resistance [101]. These restriction factors serve as promising targets
Author Manuscript

for the use of CRIPSR/Cas9 in either activating these restriction factors or inducing the
mutations that make the proteins resistant to inhibition by viral proteins.

Similarly, various studies have investigated the host-pathogen interaction that occurs upon
IAV infection in order to identify key host genes involved. A genome-wide RNAi study
identified hundreds of host genes involved in restricting or promoting cellular infection by
RNA viruses, including IAV [102]. Of the genes identified, COPI-protein families, fibroblast
growth factor receptor (FGFR) proteins, glycogen synthase kinase 3 (GSK3)-beta and
calcium/calmodulin-dependent protein kinase (CaM kinase) II beta (CAMK2B) were
significantly enriched and found to be involved in early steps of influenza virus replication.
This study also showed that inhibition of these identified targets using small molecules
resulted in attenuated viral replication; further proving that these may be suitable targets
Author Manuscript

using CRISPR/Cas9.

A study conducting a genetic screening using CRISPR/Cas9 identified host factor


dependencies of flavivirus replication including proteins involved in endocytosis, heparin
sulfation and endoplasmic reticulum membrane complex (EMC) [103]. This study serves as
a resource and paves the way for drug targeting and development. More recently, CIRPSR/
Cas9 knock-out of viperin, an interferon stimulated gene (ISG) involved in innate immunity,
further elucidated its role in Zika virus (ZIKV) infection [104]. ZIKV was originally isolated
from a sentinel monkey in Uganda and belongs to the Flaviviridae family of positive-sense
RNA viruses that includes dengue (DENV), West Nile (WNV) and Japanese encephalitis
(JEV) [105]. Vanwalscappel and colleagues identified a TLR agonists with potent inhibition
of ZIKV infection and identified enrichment of viperin upon treatment with the TLR
Author Manuscript

agonist. Gene editing tools such as CRISPR/Cas9 may be used to target and induce antiviral
host machinery to combat the infection.

2.4 Boosting Host Immunity with CRISPR/Cas9


Dysregulated immune responses are central to pathogenesis of various diseases and
autoimmune disorders. Dysregulation could be a result of genetic mutation, pathogen-
mediated suppression of effector immune responses or altered phenotype of cells in diseased

Semin Cell Dev Biol. Author manuscript; available in PMC 2020 December 01.
Koujah et al. Page 10

microenvironment. Devising CRISPR/Cas system to correct these genetic errors or


enhancement of immune cell function by targeted modification can benefit humans against
Author Manuscript

numerous diseases. Multiple studies have shown positive outcomes with CRISPR/Cas-
mediated improvement of T cells that are central to adaptive immune response. While
specific role in boosting antiviral T cell immunity has not been studied, evidences support
CRISPR/Cas-based enhancement of T cell-mediated immunity in cancer immunotherapy.

To test CRISPR/Cas system-mediated enhancement of T cell function, Rupp et al. examined


disruption of programmed cell death protein 1 (PD-1) in chimeric antigen receptor (CAR) T
cells which are modified to detect specific tumor antigens [145]. Recognition of tumor cells
by CAR T cells alone is not sufficient to clear cancerous cells due to the immunosuppressive
tumor microenvironment and expression of inhibitory ligands, such as PD-L1, on both tumor
cells and surrounding tissues. PD-L1 can interact with its ligand PD-1 on T cells and trigger
immunosuppressive signals leading to T cell exhaustion and hypo-function. This scenario is
Author Manuscript

frequently observed in chronic viral infections and tumor infiltrating lymphocytes in patients
with advanced malignancy. Using a Cas9 RNP gene editing (targeting PDCD-1) and
lentiviral transduction (encoding the anti-CD19 CAR) of primary human T cells, PDCD-1
gene was ablated while CD19 CAR expression was simultaneously achieved in primary
human T cells. The genetically modified T cells (PD-1−: CD19 CAR+) when cultured with
K562 tumor cells (CD19+; PD-L1+) displayed efficient at killing PD-L1+ tumor cells. The
efficacy of PD-1−: CD19 CAR+ T cells was assessed in vivo in NOD scid gamma (NSG)
mice. Mice with adoptively transferred PD-1−: CD19 CAR+ T cells, following subcutaneous
injection of CD19+ PD-L1+ K562 cells and establishment of tumors, showed reduced tumor
growth, increased survival and accelerated tumor clearance compared with controls. Overall,
CRISPR-mediated targeted disruption of the Pdcd1 locus in human CAR T cells strongly
enhance in vivo anti-tumor efficacy.
Author Manuscript

Eyquem et al. employed a different approach of targeted delivery of CD19-specific CAR to


the T-cell receptor α constant (TRAC) locus using CRISPR/Cas system [146]. They
demonstrated that CAR expression is uniform in T cells and these genetically modified cells
are more potent as in their antitumor activity and outperform CAR T cell generated using
conventional approach in a mouse model of acute lymphoblastic leukaemia (AML). For
instance, markers of T cell exhaustion such as PD1, LAG3 and TIM3 were expressed at
higher levels in conventional (retrovirally delivered) CAR compared to TRAC-CAR T cells
and the former showed accelerated terminal differentiation further correlating with higher
antitumor activity of TRAC-CAR T cells. It can be noted that in vitro functional studies did
not reveal any notable differences between TRAC-encoded and randomly integrated 1928z,
in terms of both cytotoxicity and T-cell proliferation in response to weekly stimulation with
Author Manuscript

CD19+ antigen-presenting cells strongly suggesting that readouts of in vitro and in vivo
studies may marked vary thereby signifying the need to perform in vivo experiments. These
findings clearly show that targeted delivery of CAR using CRISPR/Cas can reduces tonic
signalling, averts accelerated T-cell differentiation and exhaustion, and increases the
therapeutic potency of engineered T cells in vivo.

In another study, a different strategy was employed to evaluate CRISPR/Cas-mediated


genomic editing to reprogram human T cells by correcting a gene locus in a genetic disease

Semin Cell Dev Biol. Author manuscript; available in PMC 2020 December 01.
Koujah et al. Page 11

as well as selective targeting of cancer cell [147]. An electroporation was used to deliver
crRNA and tracrRNA into T cells in order to generate genetically modified, highly viable,
Author Manuscript

therapeutic grade T cells with no significant evidence of off-target integrations. The efficacy
and targeted editing of the approach was tested on individuals (three siblings) with
monogenic primary immune deficiency, an autoimmune disease caused by recessive loss-of-
function mutations in the gene encoding the IL-2 alpha receptor (IL2RA), required for
healthy regulatory T cells [148-149]. Two different mutations were identified on the ILRA
of the subjects (1) c.530A>G, creates a premature stop codon and (2) c.800delA, causes a
frameshift in the reading frame. Following restoration of genetic mutation, T cells were
functionally assessed and active IL2 signaling was demonstrated by increased STAT5
phosphorylation upon IL-2 treatment, a hallmark of productive signaling. Furthermore, this
CRISPR/Cas strategy was employed to assess in vivo anti-tumor function of T cells. A pair
of T cell receptor (TCR) genes viz., TCR-β and TCR-α which recognizes the NY-ESO-1
tumor antigen was incorporated into the TRAC locus of polyclonal T cells isolated from
Author Manuscript

healthy human donors [150]. Interestingly, it was observed that NY-ESO-1 TCR knock-in T
cells preferentially localized to, persisted at, and proliferated in the tumor rather than the
spleen. Adoptive transfer of sorted NY-ESO-1 TCR T cells also reduced the tumor burden in
treated animals. These findings clearly support successful use of non-viral templates to
harness potency of CRISPR/Cas genome editing capability in human health and disease.
Most importantly, this methodology is cost-effective, absence of viral templates poses low
risk of adverse impact on human health and is easy to adapt good manufacturing practices
(GMP) for clinical use. However, a key limitation of using non-viral templates is low
efficiency of genome editing. All of these studies strongly support that CRISPR/Cas can be
utilized to boost T cell immunity and selectively targeting of cancer cells by T cells in vivo.
Further investigations on long-term impact on the functions of genetically-modified T cells
and how their phenotype is impacted in different individuals should be thoroughly
Author Manuscript

examined. It will be important to assess CRISPR/Cas technology in boosting antiviral


immunity with specific focus on CD8+ T cell and natural killer (NK) cell which are central
to antiviral immunity and actively participate in clearing virus infected cells.

3. Targeting Viral Genomes


CRISPR/Cas9 has also been used to directly target and edit viral genome to impair its
activity and eradicate it from the host (Table 2). While drugs targeting the virus, such as
inhibitors that neutralize viral particles and nucleoside analogues that terminate viral
replication have been developed, most have resulted in development of escape mutants from
repeated use or ineffective against some viral strains. Using CRISPR/Cas9 to target one or
multiple parts of the viral genome may be more effective in abolishing the virus from the
Author Manuscript

host.

The use of CRISPR/Cas9 to directly mutate viral genomes and impair viral replication was
demonstrated by multiple groups that targeted viral genetic elements important for both
productive and latent herpesvirus infection [106-108]. Diemen el al. designed gRNAs
targeting viral microRNAs BART5, BART6, and BART15 in latently infected gastric
carcinoma cell line NSU-719 and showed direct editing of the EBV genome [106]. This
study, along with others, showed clearance of latent viral infection in cell culture by

Semin Cell Dev Biol. Author manuscript; available in PMC 2020 December 01.
Koujah et al. Page 12

effectively targeting EBNA1, which is crucial for EBV functions and latent genome
replication, with CRISPR/Cas9. Direct editing of viral genome was proved to be possible,
Author Manuscript

allowing for continued efforts to be made in this direction.

Along the same lines, multiple gRNAs targeting essential genes of HSV-1 and HCMV also
showed high efficacy in abrogating virus replication in vitro. Specifically, the use of double
gRNAs showed complete blocking of HSV-1 replication and was suggested to be more
effective in preventing the outgrowth of resistant strains. Other studies have shown efficient
use of CRISPR/Cas9 to edit both essential and nonessential genes in the HSV genome and
this in turn has accelerated our understanding of HSV [106,109]. gRNAs targeting UL7
gene, a tegument protein of HSV, were designed by Xu et al. in 2016 using CRISPR/Cas9
and found that the replication capacity of this mutant strain was 10-fold lower than wild-type
HSV-1 [109]. Similarly, a different group showed high efficacy in directly targeting HSV-1
genome by modifying ICP0, an immediate early gene involved in HSV-1 gene expression
Author Manuscript

and replication, with no off-target modifications [107]. In 2018, a group designed a


multiplex antiviral CRISPR/Cas9 system targeting UL122/123 gene in the HCMV genome
which are important for regulating lytic replication and reactivation from latency. This study
showed effective targeting of the viral genome and abrogation of viral replication in primary
fibroblasts and U-251 MG cells [110]. The authors also suspect reduction in viral
reactivation from latency following the destruction of the IL122/123 HCMV gene.

Multiple studies have shown effective targeting of cccDNA genome of HBV by Cas9-
induced double-stranded DNA break [111,112]. Seeger et al. designed several sgRNAs
spanning the ENII/CP region and the PC of the HBV genome and showed effective
CRISPR/Cas9 targeting of HBV cccDNA in HEPG2 cells expressing HBV receptor sodium
taurocholate cotransporting polypeptide (NTCP). A different group showed the eradication
Author Manuscript

of persistent HBV infection both in vitro, using Huh-7 cells transfected with an HBV-
expression vector, and in vivo, using a hydrodynamics-HBV persistence mouse model, using
CRISPR/Cas9 [113]. In vitro, reduced production of HBV core and surface proteins was
observed using two out of the eight designed HBV-specific gRNAs. Reduced serum surface
antigen levels were observed in vivo, further suggesting a robust role of CRISPR/Cas9 -
mediated disruption of HBV infection. Soon after, multiple studies showed the complete
eradication of HBV infection and elimination of persistent HBV genome, including full-
length integrated HBV DNA and HBV cccDNA [113-121].

CRISPR/Cas9 was also used to directly target HIV-1 proviral genome and showed
elimination from latently infected human T-cells, and therefore inhibition of HIV-1
reactivation, without off-target effects [122]. Whole genome sequencing verified CRISPR/
Cas9 mediated-excision of provirus of two copies of HIV-1 by designing sgRNAs with
Author Manuscript

specific DNA sequences positioned within the HIV-1 promoter spanning the 5’ long terminal
sequence (LTR). In another approach to target latent HIV-1 provirus, viral reactivation was
achieved using Cas9 -synergistic activation mediators (dCas9-SAM) targeting the enhancer
of the HIV-1 LTR promoter. This was done in an effort to develop an additional “shock and
kill” therapeutic strategy where following viral reaction, the clearing of the viral genomes is
achieved by mechanisms of viral cytotoxicity or host immune defense. While this approach

Semin Cell Dev Biol. Author manuscript; available in PMC 2020 December 01.
Koujah et al. Page 13

suffers from limitations and requires more investigation, it proposes a unique therapeutic
strategy with clinical significance [123].
Author Manuscript

In a different study, 10 sgRNAs were designed targeting different regions of the HIV-1
genome: LTR, pol gene, and the second exon of tat/rev [124]. The site in the second exon of
Rev exhibited the highest level of mutation by Cas9 and as a result showed diminished levels
of HIV-1 gene expression and virus production. Combined use of different gRNAs, which is
predicted to diminish the change of HIV-1 escape mutants arising, targeting the viral
genome at once showed reduced viral yields by more than 24-fold. Similarly, Wang and
collogues further elucidate the impact of using combinatorial CRISPR/Cas9 as an antiviral
to extinguish HIV-1 DNA without consequences such as escape mutants and resistance
[125]. Most recently, Ophinni et al. demonstrated CRISPR/Cas9 transduction in 293T and
HeLa cells using tat and rev-targeting regions of HIV-1 genome [126]. This study confirms
no off-target effects as well as no effects on cell viability, further showing the therapeutic
Author Manuscript

potential of CRISPR/Cas9 as a single-time intervention to clear viral genomic persistence


and thus become a new cure for HIV-1.

Systemic removal of HIV from various cellular reservoirs is important for complete cure,
however, during latency, viruses may reside in more than one cell or tissue. CRISPR/Cas
technology has also been employed, both in vivo and ex vivo, to remove latent viruses (EBV,
HSV-1, HCMV, etc.) including viruses that incorporate into host genome like HIV-1 [108].
While various studies have demonstrated successful use of CRISPR/Cas system to inhibit
virus replication by targeting critical virus-encoded, efficient delivery of CRISPR/Cas
components may vary in vivo and ex vivo/in vitro and this may impact the excision of
desired pathogenic sequences hidden inside host cells. Kaminski et al. demonstrated
CRISPR/Cas-mediated excision of HIV sequence in mice and rats infected with HIV-1
Author Manuscript

[143]. They utilized a recombinant adeno-associated virus (AAV) to express modified


version of Staphylococcusaureus Cas9 (saCas9) and guide RNAs, targeting viral DNA
sequences within the 5’-LTR and the Gag gene, for systemic delivery using tail vein (mice)
or retroorbital inoculation (rats) in HIV infected animals. Deletion of target viral DNA
region was noticed as well as reduced expression of viral transcripts indicating efficient
systemic knockout of integrated viral genomes. The same research group further examined
the feasibility and efficiency of HIV-1 excision using previously tested recombinant AAV-
mediated delivery of saCas9 and a multiplex of sgRNA targeting various regions of HIV-
genome editing in humanized bone marrow/liver/thymus (BLT) mice inoculated with HIV-1
and in mice during an acute infection of EcoHIV [144]. While targeting of LTR was
sufficient to induce HIV genome excision, a combination of LTR and gal or pol targeting
induces potent excision of viral genome suggesting multiple targeting is more efficient.
Author Manuscript

However, further studies are required on possible off-target effects of CRISPR/Cas system in
vivo and how this system can be effectively used in patients with other systemic illnesses.
Additionally, the functional impact of CRISPR/Cas9 knockout on host cells should be
thoroughly investigated. Whether the effector functions of these cells are similar to normal
cells also demand a comprehensive analysis.

CRISPR/Cas9 has also successfully been used to edit the viral genomes of other viruses
such as Vaccinia virus (VACV). JC polyomavirus (JCPyV) and human papillomavirus

Semin Cell Dev Biol. Author manuscript; available in PMC 2020 December 01.
Koujah et al. Page 14

(HPV). In VACV, CRISPR/Cas9 has been used to generate efficient site-specific mutations
in the genome of the virus that can be employed as vaccine vectors against infectious
Author Manuscript

diseases. Specifically, sgRNAs targeting N1L and A46R genes were constructed due to their
role in virulence and regulating host immune response to VACV [127, 128]. Homologous
recombination was efficiently induced (62.5% and 85% respectively) by the gRNA-guided
Cas9, showing direct mutation of the viral genome. Another study utilized CRISPR/Cas9 to
inhibit virus replication and viral protein expression of JCPyV, a member of the
Polyomaviridae family, by targeting the noncoding control region and the late gene open
reading frame of the JCPyV genome [129]. Similarly, a study using CRISPR/Cas9 to
eliminate the oncogenic function of HPV constructed gRNAs targeting E6 and E7 genes
which are essential for malignant phenotype of cervical cancer by inactivating tumor
suppressor proteins p53 and pRb [130]. CRISPR/Cas9 has been applied to a variety of
viruses and shown to be both versatile and effective in most cases.
Author Manuscript

4. Current limitations to the use of CRISPR/Cas9 as an antiviral


While the use of CRISPR/Cas9 as an antiviral has shown to be both effective and precise in
various studies, limitations to its use still exist. Most of the published studies discussed in
this review have been performed in cell-culture-based systems and, while showing high
antiviral efficacy, lack convincing in vivo data from animal model studies. Although these
studies give an indication regarding the application of the system, they lack various
considerations and therefore fail to elucidate its therapeutic potential in humans. This
includes ensuring safe and efficient delivery of Cas9 and gRNA.

The time of addition, dosage and delivery vehicle of the CRISPR materials are all critical to
ensure proper use and exert the desired therapeutic benefits [131, 132]. Although various
Author Manuscript

studies suggest the activity of Cas9 is highly precise and specific, studies have indicated
unintended mutations resulting from the repair process of DNA breaks generated by
CRISPR/Cas [133, 134]. Additionally, the development of CRISPR/Cas9-induced virus
escape mutants and, as a result, the development of more pathogenic virus strains has been
reported in recent studies. This has been resolved by the use of multiple gRNAs directed
towards various sites of the genome but still presents itself as an issue in some cases [110,
136].

The consideration of viral vectors used for therapeutic delivery of Cas9:gRNA is also a key
component to ensure success and efficiency [30, 137]. The large size of Cas9 proteins and
delivery to specific cell types and tissues limits the vehicles that can be used. Lentiviruses
induce long-term expression by integration into the target cell genome, exhibit high infection
efficacy and have demonstrated safe delivery in vivo [138]. However; lentiviruses yield low
Author Manuscript

virus titers and makes them ineffective in cases where large numbers of virally infected cells
need to be targeted. Additionally, a high potential for insertional mutagenesis due to long-
term and systematic expression has been observed [139]. In contrast, adeno-associated virus
(AAV) is another common option that does not integrate into the genome, yields high viral
titers and is capable of infecting various cell types. AAV is suitable in many cases, however,
the relatively small viral genome size limits the DNA cargo that can be delivered to target
cells [140]. The development of a safe and efficient delivery system is crucial for the success

Semin Cell Dev Biol. Author manuscript; available in PMC 2020 December 01.
Koujah et al. Page 15

of CRIPSR/Cas9 in clinics and targeting tissues or cells in human body still remains to be a
challenge.
Author Manuscript

As we inch towards personalized gene editing and medicine, it is of utmost importance to


identify and evaluate adverse effect of antiviral sgRNAs on host physiology and absolutely
assuring that they do not impair genome integrity as well as transcriptome profiles. With
these challenges resolved, CRISPR/Cas will be an attractive tool to eradicate numerous viral
infections most of which remain without cure.

5. Conclusion
The recent advancements regarding gene editing technology has achieved broad application
and progressed our knowledge in multiple research areas, including microbiology and
immunology. Achieving precise genome editing in eukaryotic cells has long been studied
Author Manuscript

with programmable endonucleases, such as ZFNs and TALENs, to interfere with certain
genes. However, the feasibility and precision of these tools has been at question. CRISPR/
Cas9 presents a more powerful platform with wide application, easier retargeting ability and
reduced off-target events.

CRISPR/Cas9 has been widely used to target human genomic DNA and recently has been
directed towards viral DNA in hopes of finding a cure for viruses such as herpesvirus, HBV,
HIV and IAV. This review summarizes the application of CRISPR/Cas9 in abolishing these
viruses from the host by directly targeting viral genomes or particular host genes that aid in
viral replication and persistence. First and foremost, the goal is to identify host and virus
genes that are indispensable for virus infection. Once these targets are ascertained, a major
challenge will be to develop sgRNA that can specifically target gene-of-interest with no off-
target effect. Another challenge is to thoroughly characterize sgRNA impact on cellular
Author Manuscript

physiology both in vitro and in vivo. Using CRISPR/Cas9 in human as antiviral may be a
near future reality, however, researchers must proceed with extreme caution regarding
potential biological and ethical consequences.

Acknowledgments
Funding

Part of this work was funded by the NIH/NIDCR R21 DE026259-01A1 and R01 DE027980 to ARN and 2R01
EY024710-04 to DS.

References
[1]. Voytas DF, Gao C. Precision genome engineering and agriculture: opportunities and regulatory
Author Manuscript

challenges. PLoS Biology 2014;12[6].


[2]. Doudna JA, Charpentier E. The new frontier of genome engineering with CRISPR-Cas9. Science
2014;346[6213]:1258096. [PubMed: 25430774]
[3]. Wang H, La Russa M, Qi LS. CRISPR/Cas9 in Genome Editing and Beyond. Annu Rev Biochem
2016;85:227–264. [PubMed: 27145843]
[4]. Jiang F, Doudna JA. CRISPR-Cas9 Structures and Mechanisms. Annu Rev Biophys 2017;46:505–
529. [PubMed: 28375731]

Semin Cell Dev Biol. Author manuscript; available in PMC 2020 December 01.

You might also like