Rekayasa Genetika
Rekayasa Genetika
https://doi.org/10.1007/s12033-021-00431-7
REVIEW
Received: 29 September 2021 / Accepted: 22 November 2021 / Published online: 29 January 2022
© The Author(s), under exclusive licence to Springer Science+Business Media, LLC, part of Springer Nature 2021
Abstract
Biotechnological approaches have always sought to utilize novel and efficient methods in the prevention, diagnosis, and
treatment of diseases. This science has consistently tried to revolutionize medical science by employing state-of-the-art
technologies in genomic and proteomic engineering. CRISPR–Cas system is one of the emerging techniques in the field of
biotechnology. To date, the CRISPR–Cas system has been extensively applied in gene editing, targeting genomic sequences
for diagnosis, treatment of diseases through genomic manipulation, and in creating animal models for preclinical researches.
With the emergence of the COVID-19 pandemic in 2019, there is need for the development and modification of novel tools
such as the CRISPR–Cas system for use in diagnostic emergencies. This system can compete with other existing biotech-
nological methods in accuracy, precision, and wide performance that could guarantee its future in these conditions. In this
article, we review the various platforms of the CRISPR–Cas system meant for SARS-CoV-2 diagnosis, anti-viral therapeutic
procedures, producing animal models for preclinical studies, and genome-wide screening studies toward drug and vaccine
development.
Keywords CRISPR–Cas systems · SARS-CoV-2 · Diagnosis · Anti-viral approach · Genome-wide association study ·
Animal model
Introduction for the third large-scale human outbreak of its family in the
last two decades. This zoonotic virus is a non-segmented
The SARS-CoV-2 virus, a member of the Coronaviri- enveloped positive-sense single-stranded RNA virus with
dae family, Betacoronavirus genus, and the causative agent a large genomic size that has high animal-to-human and
of Coronavirus Disease 2019 (COVID-19), is responsible human-to-human transmission compared to its family
members [1]. The genomic sequence of this new emerging
virus has a high rate of mutation and recombination, due to
* Gholam Ali Kardar its unique self-replication phenomenon [2]. At the begin-
gakardar@tums.ac.ir
ning of the pandemic, the complete sequence of the viral
Zeinab Yousefi Najafabadi genome was shared through metagenomic approaches and
z-yousefin@razi.tums.ac.ir
the three-dimensional structure of its key proteins was deter-
1
Department of Medical Biotechnology, School of Advanced mined [3]. The fourteen open reading frames (ORF) of the
Technologies in Medicine, Tehran University of Medical SARS-CoV-2 genome encode sixteen nonstructural proteins
Sciences, Tehran, Iran to form replicase complex (such as 3-chymotrypsin-like pro-
2
Immunology, Asthma Allergy Research Institute (IAARI), tease (3CLpro), papain-like protease (PLpro), helicase, and
Tehran University of Medical Sciences, Tehran, Iran RNA-dependent RNA polymerase (RdRp), nine accessory
3
Department of Immunology, School of Medicine, Iran proteins, and four structural proteins (spike (S), membrane
University of Medical Sciences, Tehran, Iran (M), envelope (E), and nucleocapsid (N)). Spike protein has
4
Department of Applied Biosciences and Biotechnology, two functional segments (S1 and S2) that are activated by
Faculty of Science and Technology, Midlands State host cell proteases (cathepsin L and transmembrane protease
University (MSU), Gweru, Zimbabwe
serine 2 (TMPRSS2) [4]. In SARS-CoV-2 the receptor-bind-
5
Department of Medical Biotechnology, School of Medicine, ing domain (RBD) of spike protein binds to the human cell
Zanjan University of Medical Sciences, Zanjan, Iran
13
Vol:.(1234567890)
Molecular Biotechnology (2023) 65:146–161 147
receptors, Angiotensin-Converting Enzyme 2 (ACE2), and [10]. Types II, V, and VI from class II of the CRISPR/
defines the tropism and the pathogenicity of the virus, While Cas system have enzymatic activity with a similar nucle-
the spike protein of other human coronaviruses can bind to ase domain in their effector protein through the detection
the other cellular entry receptors such as aminopeptidase N of target DNA or RNA sequences. In CRISPR/Cas com-
(APN) and dipeptidyl peptidase 4 (DPP4) too. Mutation in plex, the CRISPR RNA (crRNA) is responsible for target
RBD enhances the binding affinity of the virus to human sequence (RNA or DNA) identification and can depend
ACE2 and consequently higher transmissibility compared on protospacer-associated motif (PAM) or protospacer
to SARS-CoV and MERS-CoV [1, 5]. flanking site (PFS) based on CRISPR types. In addition
The creation of new and fast platforms at three compasses to crRNA and Cas complex, type II and type V-B have
of prevention, diagnosis, and treatment of virus infections additional trans-activating RNA (tracrRNA) which medi-
are remarkable biotechnological approaches in the COVID- ates the binding between them. The single-guide RNA
19 pandemic. Therefore, knowing more about the biology (sgRNA) sequence is chimerically designed with about
and pathogenesis of the virus will help in diagnosis, treat- 20 nucleotides complementary of the target sequence,
ment regimens, and vaccine design. Regions of RdRP gene instead of the crRNA-tracrRNA complex, in the labora-
sequences in the ORF1ab, envelope protein gene (E), and tory to use as a biotechnological tool. In these complexes,
nucleocapsid protein gene (N) are hotspot regions used for the nuclease domains of the Cas protein have cleavage
molecular detection of the SARS-CoV-2 virus due to their activity, resulting in blunt or overhang on-target sequences
conserved sequences [2]. The use of intervention strategies, [11, 12]. Table 1 summarizes the three most applied types
such as targeting the virus cell entry pathway, including of class II CRISPR–Cas systems [13–16].
RBD, as well as the virus replication complex, including
RdRp, are critical targets in the design of anti-viral molecu-
lar therapeutic platforms [6, 7].
Applications of CRISPR–Cas Systems
in COVID‑19 Disease
CRISPR–Cas System
Some aspects of the CRISPR system beyond genome
Clustered Regularly Interspaced Short Palindromic editing have been considered by researchers during this
Repeats (CRISPR) and its associated proteins (Cas) are course of the COVID-19 pandemic. In this review, we
encoded by bacteria and archaea as defensive mechanisms screened research articles from PubMed and LitCovid for
against invasive genetic agents such as viruses and plas- CRISPR–Cas system application in SARS-CoV-2 using
mids through a three-stage process of adaptation, matura- ‘‘COVID-19 and CRISPR’’ as keywords for the period
tion, and interference [8, 9]. This two-component system 2020 and 2021. We returned 290 articles and after delet-
(CRISPR array and Cas protein) is classified into class I ing articles that were unrelated, review, and preprint arti-
and class II, including different types and subtypes based cles, we were left with 86 articles focusing on diagnosis,
on computational sequence and protein analysis of their anti-viral therapeutics, preclinical models, and genome-
effector subunits. The discovery of the CRISPR system wide screening. The percent distribution of the diagnosis
has revolutionized biotechnology approaches because of researches and the other categorized research articles is
its genomic sequence recognition and enzymatic cleavage presented via a pie diagram in Fig. 1.
ability to genetic engineering of the genome and beyond
II (Cas9) II-A HNH and RuvC domains 3′ G-rich motif dsDNA blunt cleavage activity Diagnostic platforms
II-B Genome-wide screening
II-C Animal model designing
V (Cas12) V-A (Cas12a/Cpf1) RuvC and Nuc domains 5′ T-rich motif dsDNA overhang cleavage Diagnostic platforms
V-B (Cas12b/C2c1) RuvC domain activity and ssDNA collateral
cleavage activity
VI (Cas13) VI-A (Cas13a/C2c2) 2 HEPN domains 3′ none G PFS ssRNA overhang cleavage Diagnostic platforms
VI-D (Cas13d) None activity and ssRNA collateral Anti-viral therapy
cleavage activity
13
148 Molecular Biotechnology (2023) 65:146–161
13
Table 2 Notable CRISPR–Cas-based diagnostic platforms for SARS-CoV-2 designed in 2020 and 2021
CRISPR–Cas Technique name Attendant methods Target genes Limit of detection Duration time References
type (LOD)
Amplification Detection method
method
Type II (Cas9) Cas9/D10A with CRISPR array- PER Electrochemical Nucleocapsid (N) 5 nM for synthesized Three steps (CRISPR [33]
nickase activity mediated biosensing SARS-CoV-2 reactant, PER reac-
primer exchange genome in the tant, and detection)
reaction-based human cell lysate within 110 min
biochemical circuit
cascades
Molecular Biotechnology (2023) 65:146–161
Francisella novicidae CRISPR/Cas9- RT-RPA Lateral flow visual Simultaneous dual 100 RNA copies per Two steps at 37 °C [34]
Cas9 mediated triple-line readout genes; envelope (E) reaction (25 μl) within 40 min
lateral flow assay and Orf1ab
(TL-LFA)
Campylobacter Leveraging engi- RT-PCR or free- Polyacrylamide gel Spike (S) One copy, or 1.7 aM unspecified [31]
jejuni NCTC11168 neered tracrRNAs amplification electrophoresis or in the original dilu-
(CjeCas9) and on-target Bioanalyzer tion, of RNA com-
DNAs for parallel pared with 3 × 108
RNA detection copies, or 0.6 nM
(LEOPARD) in the original
dilution, without
preamplification
Type V (Cas12) Cas12a (Cpf1) DNA endonuclease- RT–LAMP Lateral flow visual Nucleocapsid (N) 10 copies per µl of Three steps within [35]
targeted CRISPR readout and envelope (E) nasopharyngeal 30–40 min
trans reporter swab samples
(DETECTR)
CRISPR-based fluo- RT-RPA Fluorescence-based Nucleocapsid (N) 2 copies per µl of Three steps within [36, 37]
rescent diagnosis detection in and envelope (E) nasal swab samples 50 min
system for COVID- 96-well microtiter
19 (COVID-19 plate and adaptable
CRISPR-FDS) to smartphone-read
chip format
In vitro Specific RT-LAMP Lateral flow and Nucleocapsid (N) 10 copies per µl of One and two steps [38]
CRISPR-based fluorescence-based and envelope (E) Nasopharyngeal at 62 °C within
Assay for Nucleic detection swab samples 60 min
acid detection
(iSCAN)
CRISPR–Cas12a RT-RAA Fluorescence-based orf1a, orf1b nucle- 10 copies per µl input Three steps within [39]
naked eye readout detection ocapsid (N) and 60 min
(CRISPR–Cas12a- envelope (E)
NER)
13
149
Table 2 (continued)
150
CRISPR–Cas Technique name Attendant methods Target genes Limit of detection Duration time References
type (LOD)
13
Amplification Detection method
method
Specific enhancer rRT-PCR Fluorescence-based Orf1ab (O) and 1.6 copies per µl of Three steps within [40]
for PCR-amplified detection nucleocapsid (N) pharyngeal and rRT-PCR time and
Nucleic Acid nasopharyngeal CRISPR detection
(SENA) swabs with 95% time
confidence
Manganese-enhanced RT-RAA Fluorescence-based Envelope (E) 5 copies with Mn2+ Three steps within [41]
Cas12a detection detection compared to 10 45 min
(MeCas12a) copies with Mg2+
ENHANCE system RT-LAMP Fluorescence-based Nucleocapsid (N) 3–300 copies per µl Three steps within [42]
assay and lateral 40–60 min
flow assay
One-pot visual RT-LAMP Fluorescence-based Spike (S) 5 copies per µl Three steps within [43]
SARS-CoV-2 assay 45 min
detection sys-
tem named
(opvCRISPR)
All-In-One Dual RT-RPA Fluorescence-based Nucleocapsid (N) 5 copies per µl One-step amplifica- [44]
CRISPR–Cas12a assay tion and detection
(AIOD-CRISPR) at room tempera-
ture (37 °C) within
20 min
Microfluidic Isota- RT-LAMP Fluorescence-based Nucleocapsid (N) 10 copies per µl of Within 30 min [45]
chophoresis (ITP)- assay and envelope (E) Nasopharyngeal
CRISPR-based swab samples
Digital warm-start RT-DAMP Fluorescence-based Nucleocapsid (N) 5 copies/μl RNA in One step in Quant- [46]
CRISPR (DWS- assay the chip tenfold Studio 3D digital
CRISPR) higher sensitivity chip initiating at
than tube-based above 50 °C
bulk assay format
Cas12b (C2c1) CRISPR-assisted RT-RAA Fluorescence-based RdRp 10 copies per µl one step at 42 °C [47]
detection (CAS- assay within 60 min
detec)
SHERLOCK Testing RT-LAMP Fluorescence-based Nucleocapsid (N) 100 copies/reaction One-step amplifica- [48]
in One Pot (STOP) assay and lateral tion and detection
flow assay at 60 °C within
45 min
Molecular Biotechnology (2023) 65:146–161
Table 2 (continued)
CRISPR–Cas Technique name Attendant methods Target genes Limit of detection Duration time References
type (LOD)
Amplification Detection method
method
Type VI (Cas13) Cas13a (C2c2) Specific High-sen- RT–RPA and T7 Fluorescence-based Spike (S), nucleopro- 42 RNA copies per Three steps within [26, 49]
sitivity Enzymatic transcription assay and lateral tein (N) replicase reaction of Naso- 60 min
Reporter unlocking flow assay polyprotein 1ab pharyngeal swab
(SHERLOCK) (Orf1ab)
Combinatorial PCR or RT-RPA Fluorescence- spike (S), nucleopro- 104 copies per µl for Three steps [50, 51]
arrayed reactions based assay using tein (N) replicase synthetic targets
Molecular Biotechnology (2023) 65:146–161
13
151
152 Molecular Biotechnology (2023) 65:146–161
Step I: Amplification
RPA
LAMP
Nasopharyngeal sample Inactivation Extraction of RAA
collection of SARS-CoV-2 SARS-CoV-2 RNA
Amplified DNA for target gene
T7 transcription
Cas9 detection-based Cas12 detection-based Cas13 detection-based
Binding to target
amplified biotinylated
DNA Cleaved reporters
Cleaved reporters
anti-FAM
antibody Cleaved
Biotin-FAM
reporter Cleaved
Quencher-
Fluorescent
Fluorophore
Positive band emission
Sample flow Control band Anti- Control band Positive band anti- reporter
streptavidin Sample flow
rabbit Antibody streptavidin FAM antibody coated
coated coated coated
Fig. 2 An overview of CRISPR-based diagnostic assays with three extraction of RNA of SARS-CoV-2, pass through three steps and can
types of Cas enzymes; Cas9, Cas12, and Cas13 detections with be visualized through the lateral flow assay or fluorescence-based
names of FELUDA, DETECTR, and SHERLOCK, respectively, after assay
respiratory swab, by lateral flow assay. The collaboration of combination with reverse transcriptase-recombinase poly-
this method with reverse transcriptase-loop-mediated iso- merase amplification (RT-RPA) and T7 transcription to
thermal amplification (RT-LAMP) to convert and amplify increase detection accuracy using a quenched fluorescent
RNA virus to DNA effectively increased the detection effi- ssRNA reporter suitable for visual or lateral flow read-
ciency [35]. The advantages of this method are laboratory out [55]. This platform was successful in the diagnosis of
instrument independence and rapidness, along with accept- the SARS-CoV-2 virus during its pandemic. It is the first
able accuracy and specificity [28, 53]. Other DETECTR-like CRISPR-based platform to be used on clinical samples with
platforms for COVID-19 are listed in Table 2. the US FDA-EUA approval and has acceptable results com-
The pioneer of the CRISPR-DX system is Specific High- pared with other diagnostic methods, such as next-genera-
sensitivity Enzymatic Reporter unlocking (SHERLOCK) tion sequencing (NGS) and RT-PCR (q-PCR) [22, 49, 56].
that uses type VI/Cas13 for detection [54]. This platform Discrimination of the wild and mutated SARS-CoV-2, such
was designed to detect pathogens, including viruses, in as D614G mutation with Cas13a-gRNA and Cas12a-gRNA
13
Molecular Biotechnology (2023) 65:146–161 153
Expressed Cas13-
SARS-CoV-2 infection
crRNA complex
Local delivery to
airway cells
SARS-CoV-2
Library of Cas13- RNA genome or mRNA
AAV
crRNA in AAV library
transfer vector for
SARS-CoV-2
conserved genomic Viral genome Blocking viral genome
regions degradation expression
mACE2 promoter
non-expressible
Exon-2 mouse ACE2 ACE2
Intranasal
inoculation of
CRISPR-Cas9 complex SARS-CoV-2
Severe infectious
No infectious
Sequencing &
data analysis
sgRNA
Library transduction
sgRNA
CRISPR-based platforms, is valuable for monitoring of platforms for SARS-CoV-2 virus detection are listed in
epidemiological analyses and can improve the ability of Table 2. Figure 2 shows the steps of the available three types
POC tests [57, 58]. Other notable CRISPR–Cas13-based of CRISPR-DX platforms.
13
154 Molecular Biotechnology (2023) 65:146–161
◂Fig. 3 a Cas13-crRNA complex through variable delivery systems off-targets in the DNA sequence of virus-infected host
(such as AAV delivery method) can target virus genes (such as cells [15, 64].The CRISPR–Cas13 system is not depend-
ORF1ab, RdRp, S, and N genes) to degrade viral genome and block
genome expression of virus. b Production of humanized ACE2 mouse
ent on the detection of PAM and the crRNA targets ssRNA
model through the microinjection of CRISPR–Cas9 complex and coding sequences of the virus without interfering with
human ACE2 template sequence in mouse zygote to substitute mouse the human transcriptome, causing sequence degradation,
ACE2 gene and express in the lung, intestine, and brain of a mouse interrupting gene expression, and eventually blocking viral
under its promoter. This humanized model is compared with a wild-
type mouse in SARS-CoV-2 infectious conditions. c The pooled or
function [65]. Therefore, using CRISPR–Cas13 is superior
arrayed CRISPR genome-wide screening is done for analysis of top- to CRISPR–Cas9 as an anti-viral programmable inhibition
ranked gene clusters in the pathway of virus infection in host cells. system.
In this direction, designing of sgRNA library for targeting candidate Prophylactic anti-viral CRISPR in human cells (PAC-
host cell gene to production of knocked out cells and challenging with
SARS-CoV-2 virus are done for determining of anti-viral and pro-
MAN), effectively can identify and degrade the virus
viral genes from sensitive and resistant cells sequence and its mutants in the human lung epithelial
cells. The PAC-MAN system uses the class 2, type VI-D,
CRISPR–Cas13d system derived from Ruminococcus fla-
CRISPR‑Based Therapeutics vefaciens XPD3002. PAC-MAN simultaneously identifies
more than 90% of all coronaviruses using a combination of
Conventional therapies for treating viral diseases are usu- 6 crRNAs by identifying protected regions (such as ORF1ab,
ally based on preventing the virus infections by targeting RdRp, and N genes) as well as detecting virus ssRNA in
biomolecules in the entrance pathway up to proliferation. their replication and transcription phases [66]. The com-
In the emergence of viruses, the first step in anti-viral ther- prehensive set of bioinformatics methods for receiving in
apy is drug repurposing to use existing drugs for the ben- silico optimal crRNA candidates can improve the efficiency
efit of treatment in the shortest possible time. The second of laboratory tests for combating SARS-CoV-2 in the least
approach could be target-based drug designing by focus- time [67]. Besides in silico designing and human cell line
ing on the genomics and proteomics of the desired virus tests, using animal models will improve preclinical studies
[59, 60]. Based on these two approaches, drug candidates with the simulation of respiratory infections in vivo and will
for the treatment of SARS-CoV-2 virus infectious usually help to screen the safety and effectiveness of CRISPR-based
focus on disruption of the enzymatic function of the virus platforms for anti-viral approaches [68]. The advantages of
(especially RNA-dependent RNA polymerase), prevention the CRISPR-based anti-viral systems are high flexibility in
of virus endocytosis, blocking viral proteins (envelope, identifying viral sequences, speed of detection, and direct-
membrane, nucleocapsid, and accessory proteins), helping ness against the virus [65, 66]. The Cas13d expression
innate immunity or suppressing the excessive inflamma- vector could be transmitted via the adeno-associated virus-
tory response, and prevention of SARS-CoV-2 replication packaging system (AAV delivery method) with high tropism
[61]. On the other hand, viruses that have new mutations for respiratory tissues and expressed under the induction of
and cause pandemics usually cannot respond to old drugs expression promoters in specific tissues, especially airway
and vaccines. These limitations in the treatment and pre- cells [65]. The basis of the anti-viral CRISPR-based system
vention of the COVID-19 could be applied to other viruses for SARS-CoV-2 is depicted in Fig. 3a.
of the same family, such as SARS and MERS. Therefore,
under these conditions, there is an urgent need for new CRISPR in preclinical Researches
therapies based on targeting the genomic sequences of the
virus. The disruption of gene function in anti-viral ther- Preclinical researches related to COVID-19 require suit-
apy incorporates biotechnological tools, such as siRNA or able animal model that can be infected with the SARS-
CRISPR–Cas system. One of the well-known applications CoV-2 virus and subsequently access mild-to-severe dis-
of the CRISPR–Cas system is the possibility of identifying ease. As in human cells, the target cells in the selected
the DNA or RNA sequence of the virus to cut them and animal should possess ACE2 as the SARS-CoV-2-binding
destroy their function [62]. receptor to let virus cell entrance. Animals such as ham-
Related studies have suggested CRISPR an acceptable sters, ferrets, African green monkeys, cynomolgus, and
candidate for targeting human pathogens such as HIV, hep- rhesus macaques have similar human ACE2 receptors. For
atitis B virus (HBV), herpesviruses, human papillomavirus extensive researches the best candidate is mice because
(HPV), and JC virus (JCV) [63]. The Cas9 and Cas13 as of reducing costs and increasing the number of animals
endonuclease enzyme subunits in the CRISPR system can but mouse ACE2 has much lower binding affinity for the
identify the DNA and RNA of viruses in infected mam- viral spike protein, compared to its human counterpart.
malian cells. Studies have shown that Cas9 has a lower This low binding affinity will develop a mild disease in
cleavage efficacy for ssRNA and also the ability to create mice models that is not suitable for multi-aspect studies
13
Table 3 Genome-wide CRISPR screens of pathways and processes involved in SARS-CoV-2 infection
Screened pathways & process Screened cell line Top-ranked gene clusters in Highlights Similar small molecule References
pathway
Resistant genes Sensitize genes
Cholesterol biosynthesis pathway A549 ATP6AP1 Loss of resistant genes such as RNA-sequencing of cells treated [84]
(part of the endosomal entry ATP6V1A RAB7A reduces viral entry by with amlodipine small molecule
pathway from the vacuolar NPC1 sequestering the ACE2 receptor shows a similar differential
ATPase proton pump, Retromer, RAB7A inside cell gene expression profile as seen
and Commander complexes) CCDC22 in CRISPR knock-out of genes
PIK3C3 in cholesterol biosynthesis
pathway
Molecular Biotechnology (2023) 65:146–161
Rab-GTPase requirements HAP1 SREBP/SCAP MRPS2 SCAP regulates lipid and choles- Fatostatin molecule as SCAP [83]
Glycosylphosphatidylinositol- Huh-7.5-Cas9 HS2ST1 MRPS5 terol homeostasis by sequester- inhibitors shows anti-viral
anchored biosynthesis EIF4E2 MRPS25 ing SREBPs in the ER in the properties
Cholesterol biosynthesis RAB2A MRPS27 presence of sterols 27-Hydroxycholesterol (27OHC)
RAB10 and 25OHC have SARS-CoV-2
RAB14 anti-viral activity in VeroE6
cells
Replication cycle A549 TMEM41B SARS-CoV-2 require TMEM41B None [85]
for replication cycles in cell
Chromatin remodeling Huh7.5 ARID1A TARDBP Viral RNA-RNA and RNA–pro- PFI-3, which targets the bromodo- [80, 81]
Histone modification VeroE6 DYRK1A tein interactions reveal specific mains of the SWI/SNF proteins
Cellular signaling KDM6A SARS-CoV-2-mediated mito- SMARCA4 and SMARCA2
RNA regulation CTSL chondrial dysfunction during SIS3, which targets the pro-viral
ACE2 infection pro-viral genes and gene SMAD3 identified in the
SMARCA4 pathways, including HMGB1, screen
DYRK1A and the SWI/SNF chroma-
KDM6A tin remodeling complex are
HMGB1 SARS-CoV-2 lineage HMGB1
HIRA is critical for SARS lineage viral
CABIN1 entry with critical role in ACE2
TRIP12 expression
BPTF
PIAS2
Glycosaminoglycan biosynthesis Huh-7.5 hepatoma cells (Huh-7.5- SCAP Absolute requirement for the None [86]
SREBP signaling Cas9) TMEM106B VTT-domain containing protein
Glycosylphosphatidylinositol TMEM41B TMEM41B for infection by
biosynthesis VAC14 SARS-CoV-2
Cholesterol biosynthesis ACE2
HMGCS1
MVK
PMVK
RAB6A
RAB10
13
155
Table 3 (continued)
156
Screened pathways & process Screened cell line Top-ranked gene clusters in Highlights Similar small molecule References
pathway
13
Resistant genes Sensitize genes
Phosphatidylinositol phosphate Huh7.5.1 hepatoma cells TMEM106B Deletions in TMEM106B caused None [87, 88]
biosynthesis B3GALT6 defects in lysosome traffick-
Cholesterol homeostasis B3GAT3 ing, impaired acidification, and
Heparan sulfate biosynthetic B4GALT7 EXT1 reduced levels of lysosomal
genes EXT2 enzymes but its precise molecu-
EXTL3 lar function remains enigmatic
FAM20B
NDST1
SLC35B2
UGDH
XYLT2
SREBP/SCAP
O-glycan biosynthesis HEK293T C1GalT1 Knocking-out N-glycan biosyn- Kifunensine small molecule [89]
N-glycan biosynthesis MGAT1 thesis on Spike-abrogated viral inhibits N-linked glycosylation
entry to reduce viral entry
Cholesterol biosynthesis pathway A549 ATP6AP1 Loss of resistant genes such as RNA-sequencing of cells treated [84]
(part of the endosomal entry ATP6V1A RAB7A reduces viral entry by with amlodipine small molecule
pathway from the vacuolar NPC1 sequestering the ACE2 receptor shows a similar differential
ATPase proton pump, Retromer, RAB7A inside cell gene expression profile as seen
and Commander complexes) CCDC22 in CRISPR knock-out of genes
PIK3C3 in cholesterol biosynthesis
pathway
Rab-GTPase requirements HAP1 SREBP/SCAP MRPS2 SCAP regulates lipid and choles- Fatostatin molecule as SCAP [83]
Glycosylphosphatidylinositol- Huh-7.5-Cas9 HS2ST1 MRPS5 terol homeostasis by sequester- inhibitors shows anti-viral
anchored biosynthesis EIF4E2 MRPS25 ing SREBPs in the ER in the properties
Cholesterol biosynthesis RAB2A MRPS27 presence of sterols 27-hydroxycholesterol (27OHC)
RAB10 and 25OHC have SARS-CoV-2
RAB14 anti-viral activity in VeroE6
cells
Replication cycle A549 TMEM41B SARS-CoV-2 require TMEM41B None [85]
for replication cycles in cell
Molecular Biotechnology (2023) 65:146–161
Table 3 (continued)
Screened pathways & process Screened cell line Top-ranked gene clusters in Highlights Similar small molecule References
pathway
Resistant genes Sensitize genes
Chromatin remodeling Huh7.5 ARID1A TARDBP Viral RNA-RNA and RNA–pro- PFI-3, which targets the bromodo- [80, 81]
Histone modification VeroE6 DYRK1A tein interactions reveal specific mains of the SWI/SNF proteins
Cellular signaling KDM6A SARS-CoV-2-mediated mito- SMARCA4 and SMARCA2
RNA regulation CTSL chondrial dysfunction during SIS3, which targets the pro-viral
ACE2 infection pro-viral genes and gene SMAD3 identified in the
SMARCA4 pathways, including HMGB1, screen
DYRK1A and the SWI/SNF chroma-
KDM6A tin remodeling complex are
Molecular Biotechnology (2023) 65:146–161
13
157
158 Molecular Biotechnology (2023) 65:146–161
[69, 70]. The application of transgenic mice can solve this host cell lines can be done by CRISPR–Cas9 genome-wide
problem by inserting the human ACE2 receptor gene into mutagenesis screening. Recently screening of the Norovi-
the mouse embryo genome in an engineered manner that ruses, Zika virus, West Nile virus, and HIV in host cell lines
will pass on to the next generation. In this case, human- has been performed by CRISPRing [77–79].
ized ACE2-incorporated mice will be more sensitive to The necessity to identify the pathogenesis of the SARS-
SARS-CoV-2 infection via intranasal inoculation. Notably, CoV-2 virus pandemic, new screening studies are extended
the pathological outcomes and subsequent lung damages for revealing of viral entry and pathogenesis of host genes
will be similar to human infection [71]. In addition to the through genome-wide pooled or array CRISPR libraries in
biotechnology-based techniques used to produce transgene the appropriate cell lines. In these studies, the candidate host
mice, CRISPR technology could also insert the target genes are determined by systems biology approaches and
gene into the mouse genome by the knock-in process. For host–viral protein interactome [61]. The targeted sgRNA
COVID-19 preclinical studies, the CRISPR–Cas9 knock-in libraries are designed for candidate host genes to produce
technology can help the expression of the human ACE2 knock-out cell lines. These mutant cells will be challenged
receptor in various mouse tissues. Humanized mice sen- by virus and finally, the sequencing of the target region
sitize to SARS-CoV-2 intranasal inoculation and intragas- of the sgRNAs on mutant cell genome will determine the
tric inoculation [72], which is depicted in Fig. 3b. The causative genes of resistance and sensitivity [80–82]. The
production of humanized mice has facilitated anti-viral process of the CRISPR genome-wide screening of SARS-
therapeutic research, the development of vaccines, study- CoV-2 host cell is depicted in Fig. 3c. After knocking-out
ing virus transmission and pathogenesis, and screening steps the genes that make the virus-resistant cells are pro-
severe symptoms in COVID-19. viral, and the genes that make the virus-sensitive cells are
anti-viral. By propagating resistant knock-out cells and
sequencing data analysis, anti-viral drugs would be des-
Genome‑Wide Screening by CRISPR ignable based on these gene pathways [83]. In Table 3, the
CRISPR-based genome-wide screening of important cellular
Functional studies of genes via conditional mutagenesis in pathways and processes to determine top-ranked resistant
mammalian cells have been considered for decades. Genome and sensitive genes involved in SARS-CoV-2 infection is
engineering tools such as Cre-recombinase, RNAi, and reported. In some cases, the candidate small molecules that
designer nucleases such as ZFN and TALENs have been mimic anti-viral gene pathways or blocking pro-viral gene
used for forwarding genetic screening until 2012. The dis- pathways have been specified. The studies show that due
covery of the CRISPR–Cas system as a novel biotechnologi- to the lack of adequate knowledge about specific genetic
cal method of designer nucleases generation is an alternative pathways involved in cytotoxicity and metabolism associated
technique to overcome the limitations. One of the advan- with drug repurposing in SARS-CoV-2, research in this field
tages of genome-wide CRISPR screening is the designing has begun by genome-wide CRISPR.
of sgRNA libraries to target thousands of genes simultane-
ously for pooled or arrayed screening and directly identify
the candidates of desired phenotype [73, 74]. In addition Conclusion
to genome-wide mutagenesis through CRISPR knock-out
approach, this system is a suitable tool for insertion and After less than two years of the COVID-19 pandemic,
deletion of the target genes. Also, site-directed base editing CRISPR technology has shown some potential in all appli-
and up/down gene regulation via gene-specific transcription cations, from diagnosis to treatment of this disease. All of
factors can be done with the new modifications of CRISPR these capabilities are as a result of CRISPR’s high sensi-
system. CRISPRing has provided an opportunity to under- tivity, flexibility, adaptability, and developable platform. In
stand the biology of mammalian cells related to diseases, addition, based on new SARS-CoV-2 mutations and the need
such as cancers and pathogen infectious, including viruses, for newly designed vaccines, we have to find other treatment
and designing the pharmacological studies through the crea- strategies like CRISPR technology for COVID-19 and other
tion of targeted mutant cell libraries [75, 76]. future viral infections. In addition to all advantages of using
Usually the host genes that coordinate the virus entrance the CRISPR system in the detection of the virus variants,
and pathogenesis processes are unknown. The identification producing a preclinical animal model for drug and vaccine
of principal host genes in the viral infection, regulation, or researches, its application in anti-viral therapeutics based
suppression pathway can provide a better understanding of on eliminating the virus in the infectious cells, and studies
the virus function in the host cell and can help to find novel on genes involved in the disease; this system certainly has
potential therapeutic marks such as antagonist drugs and shortcomings.
design an appropriate vaccine. All of these studies in the
13
Molecular Biotechnology (2023) 65:146–161 159
One of the disadvantages of this system in diagnosis is 8. Makarova, W. Y., & Koonim, E. (2018). Classification and nomen-
difficult to access results for clinicians in clinical studies, so clature of CRISPR-Cas systems: Where from here? The CRISPR
Journal, 1(5), 325–336.
diagnostic protocols based on CRISPR-DX platforms should 9. Makarova, K. S., et al. (2011). Evolution and classification of
be toward automation as well as better reading using the the CRISPR-Cas systems. Nature Reviews Microbiology, 9(6),
mobile application with provision for offline or online mode. 467–477.
In studies related to anti-viral applications of the CRISPR 10. Tang, Y., & Fu, Y. (2018). Class 2 CRISPR/Cas: An expanding
biotechnology toolbox for and beyond genome editing. Cell &
system, the biggest limitation is the development of effective Bioscience, 8, 59.
and safe in vivo delivery methods. Therefore, more stud- 11. Shmakov, S., et al. (2015). Discovery and functional characteri-
ies on the transfer of structures are needed. Also, sufficient zation of diverse class 2 CRISPR-Cas systems. Molecular Cell,
expression of CRISPR/Cas in the target cells has a direct 60(3), 385–397.
12. Shmakov, S., et al. (2017). Diversity and evolution of class 2
effect on achieving efficient viral inhibition through viral CRISPR-Cas systems. Nature reviews. Microbiology, 15(3),
genome cleavage. 169–182.
Off-target effects and limited on-target activity as well 13. Burmistrz, M., Krakowski, K., & Krawczyk-Balska, A. (2020).
as low knock-out efficiency for designed sgRNAs are RNA-targeting CRISPR-Cas systems and their applications.
International Journal of Molecular Sciences. https://doi.org/
other problems facing CRISPR genome-wide screening 10.3390/ijms21031122
researches, which require optimizing CRISPR systems 14. Jinek, M., Chylinski, K., Fonfara, I., Hauer, M., Doudna, J. A.,
through structural engineering and more precise design of & Charpentier, E. (2012). A Programmable dual-RNA—Guided
sgRNAs for the target genome. DNA endonuclease in adaptive bacterial immunity. Science,
337(6096), 816–821.
15. Freije, C. A., et al. (2019). Programmable inhibition and detec-
tion of RNA viruses using Cas13. Molecular Cell, 76(5), 826-
Author Contributions Conceptualization, ZY and SF; supervision, GK; 837.e11.
writing—original draft preparation, ZY; reviewing and editing of the 16. Zetsche, B., et al. (2015). Cpf1 is a single RNA-guided endonu-
manuscript, GK, RF, and SK; all authors read and agreed to the final clease of a class 2 CRISPR-Cas system. Cell, 163(3), 759–771.
version of the manuscript. 17. Esbin, M. N., Whitney, O. N., Chong, S., Maurer, A., Darzacq,
X., & Tjian, R. (2020). Overcoming the bottleneck to widespread
Funding This research was not funded. testing: A rapid review of nucleic acid testing approaches for
COVID-19 detection. RNA, 26(7), 771–783.
18. Shen, M., et al. (2020). Recent advances and perspectives of
Declarations nucleic acid detection for coronavirus. Journal of Pharmaceuti-
cal Analysis. https://doi.org/10.1016/j.jpha.2020.02.010
Conflict of interest The authors declare that they have no conflict of 19. Feng, W., et al. (2020). Molecular diagnosis of COVID-19:
interest. Challenges and research needs. Analytical Chemistry, 92(15),
10196–10209.
20. R. Jalandra et al., Strategies and perspectives to develop SARS-
CoV-2 detection methods and diagnostics. Biomed Pharmacother,
2020.
References 21. Murugan, K., Babu, K., Sundaresan, R., Rajan, R., & Sashital, D.
G. (2017). The Revolution continues: Newly discovered systems
1. Harrison, A. G., Lin, T., & Wang, P. (2020). Mechanisms of expand the CRISPR-Cas toolkit. Molecular Cell, 68(1), 15–25.
SARS-CoV-2 transmission and pathogenesis. Trends Immunol- 22. Guglielmi, G. (2020). First CRISPR test for the coronavirus
ogy. https://doi.org/10.1016/j.it.2020.10.004 approved in the United States. Nature. https://doi.org/10.1038/
2. Sood, S., et al. (2020). COVID-19 pandemic: From molecular d41586-020-01402-9
biology, pathogenesis, detection, and treatment to global societal 23. Sashital, D. G. (2018). Pathogen detection in the CRISPR-Cas era.
impact. Current Pharmacology Reports. https://doi.org/10.1007/ Genome Medicine, 10(1), 32.
s40495-020-00229-2 24. Jolany Vangah, S., Katalani, C., Booneh, H. A., Hajizade, A.,
3. Dömling, A., & Gao, L. (2020). Chemistry and biology of SARS- Sijercic, A., & Ahmadian, G. (2020). CRISPR-based diagnosis
CoV-2. Chem, 6(6), 1283–1295. of infectious and noninfectious diseases. Biological Procedures
4. V’kovski, P., Kratzel, A., Steiner, S., Stalder, H., & Thiel, V. Online. https://doi.org/10.1186/s12575-020-00135-3
(2020). Coronavirus biology and replication: Implications for 25. Azmi, I., et al. (2021). A saliva-based RNA extraction-free work-
SARS-CoV-2. Nature Reviews Microbiology. https://doi.org/10. flow integrated with Cas13a for SARS-CoV-2 detection. Frontiers
1038/s41579-020-00468-6 in Cellular and Infection Microbiology. https://doi.org/10.3389/
5. Shang, J., et al. (2020). Cell entry mechanisms of SARS-CoV-2. fcimb.2021.632646
PNAS, 117(21), 11727–11734. 26. Fozouni, P., et al. (2020). Amplification-free detection of SARS-
6. Morse, J. S., Lalonde, T., Xu, S., & Liu, W. R. (2020). Learning CoV-2 with CRISPR-Cas13a and mobile phone microscopy. Cell.
from the past: Possible urgent prevention and treatment options for https://doi.org/10.1016/j.cell.2020.12.001
severe acute respiratory infections caused by 2019-nCoV. Chem- 27. Arizti-Sanz, J., et al. (2020). Streamlined inactivation, amplifica-
BioChem, 21(5), 730–738. tion, and Cas13-based detection of SARS-CoV-2. Nature Com-
7. Elfiky, A. A. (2020). Ribavirin, remdesivir, sofosbuvir, galidesivir, munications, 11(1), 5921.
and tenofovir against SARS-CoV-2 RNA dependent RNA poly- 28. Chen, Y., et al. (2020). Contamination-free visual detection of
merase (RdRp): A molecular docking study. Life Science. https:// SARS-CoV-2 with CRISPR/Cas12a: A promising method in the
doi.org/10.1016/j.lfs.2020.117592
13
160 Molecular Biotechnology (2023) 65:146–161
point-of-care detection. Biosensors and Bioelectronics. https:// 49. Patchsung, M., et al. (2020). Clinical validation of a Cas13-based
doi.org/10.1016/j.bios.2020.112642 assay for the detection of SARS-CoV-2 RNA. Nature Biomedical
29. Jia, C., et al. (2018). New applications of CRISPR/Cas9 system Engineering, 4, 1140–1149.
on mutant DNA detection. Gene, 641, 55–62. 50. Ackerman, C. M., et al. (2020). Massively multiplexed nucleic
30. Wang, Q., Zhang, B., Xu, X., Long, F., & Wang, J. (2018). acid detection with Cas13. Nature, 582(7811), 277–282.
CRISPR-typing PCR (ctPCR), a new Cas9-based DNA detection 51. Storch, G. A. (2020). CRISPR tool scales up to interrogate a huge
method. Scientific Reports, 8(1), 14126. line-up of viral suspects. Nature, 582(7811), 188–189.
31. Jiao, C., et al. (2021). Noncanonical crRNAs derived from host 52. Chen, J. S., et al. (2018). CRISPR-Cas12a target binding
transcripts enable multiplexable RNA detection by Cas9. Science, unleashes indiscriminate single-stranded DNase activity. Sci-
372(6545), 941–948. ence, 360(6387), 436–439.
32. Hajian, R., et al. (2019). Detection of unamplified target genes via 53. Saha, A., Arantes, P. R., Hsu, R. V., Narkhede, Y. B., Jinek, M.,
CRISPR–Cas9 immobilized on a graphene field-effect transistor. & Palermo, G. (2020). Molecular dynamics reveals a DNA-
Nature Biomedical Engineering, 3(6), 427–437. induced dynamic switch triggering activation of CRISPR-
33. Dai, Y., Xu, W., Somoza, R. A., Welter, J. F., Caplan, A. I., & Liu, Cas12a. Journal of Chemical Information and Modeling.
C. C. (2020). Integrated multi-function heterogeneous biochemi- https://doi.org/10.1021/acs.jcim.0c00929
cal circuit for high-resolution electrochemistry based genetic 54. Gootenberg, J. S., et al. (2017). Nucleic acid detection with
analysis. Angewandte Chemie International Edition. https://doi. CRISPR-Cas13a/C2c2. Science, 356(6336), 438–442.
org/10.1002/anie.202010648 55. Gootenberg, J. S., Abudayyeh, O. O., Kellner, M. J., Joung, J.,
34. Xiong, E., et al. (2020). Simultaneous dual-gene diagnosis of Collins, J. J., & Zhang, F. (2018). Multiplexed and portable
SARS-CoV-2 based on CRISPR/Cas9-mediated lateral flow nucleic acid detection platform with Cas13, Cas12a, and Csm6.
assay. Angewandte Chemie International Edition. https://doi.org/ Science (New York, NY), 360(6387), 439–444.
10.1002/anie.202014506 56. Hou, T., et al. (2020). Development and evaluation of a rapid
35. Broughton, J. P., et al. (2020). CRISPR-Cas12-based detection CRISPR-based diagnostic for COVID-19. PLoS Pathogens.
of SARS-CoV-2. Nature Biotechnology. https://doi.org/10.1038/ https://doi.org/10.1371/journal.ppat.1008705
s41587-020-0513-4 57. Wang, Y., et al. (2021). Detection of SARS-CoV-2 and its
36. Huang, Z., et al. (2020). Ultra-sensitive and high-throughput mutated variants via CRISPR-Cas13-based transcription ampli-
CRISPR-p owered COVID-19 diagnosis. Biosensors and. https:// fication. Analytical Chemistry, 93(7), 3393–3402.
doi.org/10.1016/j.bios.2020.112316 58. Meng, Q., et al. (2021). Detection of the SARS-CoV-2 D614G
37. Ning, B., et al. (2020). A smartphone-read ultrasensitive and quan- mutation using engineered Cas12a guide RNA. Biotechnology
titative saliva test for COVID-19. Science Advances. https://doi. Journal. https://doi.org/10.1002/biot.202100040
org/10.1126/sciadv.abe3703 59. Saxena, A. (2020). Drug targets for COVID-19 therapeutics:
38. Ali, Z., et al. (2020). iSCAN: An RT-LAMP-coupled CRISPR- Ongoing global efforts. Journal of Biosciences, 45(1), 87.
Cas12 module for rapid, sensitive detection of SARS-CoV-2. 60. Chellapandi, P., & Saranya, S. (2020). Genomics insights of
Virus Research, 9, 221. SARS-CoV-2 (COVID-19) into target-based drug discov-
39. Guo, L., et al. (2020). SARS-CoV-2 detection with CRISPR diag- ery. Medicinal Chemistry Research. https://d oi.o rg/1 0.1 007/
nostics. Cell Discov, 6, 34. s00044-020-02610-8
40. Huang, W., et al. (2020). A CRISPR-Cas12a-based specific 61. Gordon, D. E., et al. (2020). A SARS-CoV-2 protein interaction
enhancer for more sensitive detection of SARS-CoV-2 infection. map reveals targets for drug repurposing. Nature, 583(7816),
EBioMedicine, 61, 103036. 459–468.
41. Ma, P., et al. (2020). MeCas12a, a highly sensitive and specific 62. Atzrodt, C. L., et al. (2020). A Guide to COVID-19: a global
system for COVID-19 detection. Advanced Science (Weinheim), pandemic caused by the novel coronavirus SARS-CoV-2. FEBS
7(20), 2001300. Journal. https://doi.org/10.1111/febs.15375
42. Nguyen, L. T., Smith, B. M., & Jain, P. K. (2020). Enhancement of 63. Soppe, J. A., & Lebbink, R. J. (2017). Antiviral goes viral:
trans-cleavage activity of Cas12a with engineered crRNA enables Harnessing CRISPR/Cas9 to combat viruses in humans. Trends
amplified nucleic acid detection. Nature Communications, 11(1), in Microbiology, 25(10), 833–850.
4906. 64. Price, A. A., Sampson, T. R., Ratner, H. K., Grakoui, A., &
43. Wang, R., et al. (2020). opvCRISPR: One-pot visual RT-LAMP- Weiss, D. S. (2015). Cas9-mediated targeting of viral RNA in
CRISPR platform for SARS-cov-2 detection. Biosens Bioelectron, eukaryotic cells. Proceedings of the National Academy of Sci-
172, 112766. ences USA, 112(19), 6164–6169.
44. Ding, X., et al. (2020). Ultrasensitive and visual detection of 65. Nguyen, T. M., Zhang, Y., & Pandolfi, P. P. (2020). Virus
SARS-CoV-2 using all-in-one dual CRISPR-Cas12a assay. Nature against virus: A potential treatment for 2019-nCov (SARS-
Communications, 11(1), 4711. CoV-2) and other RNA viruses. Cell Research, 30(3), 189–190.
45. Ramachandran, A., et al. (2020). Electric field-driven microflu- 66. T. R. Abbott et al., Development of CRISPR as an Antiviral
idics for rapid CRISPR-based diagnostics and its application to Strategy to Combat SARS-CoV-2 and Influenza. Cell, 2020.
detection of SARS-CoV-2. PNAS. https://doi.org/10.1073/pnas. 67. Wang, L., Zhou, J., Wang, Q., Wang, Y., & Kang, C. (2021).
2010254117 Rapid design and development of CRISPR-Cas13a targeting
46. Ding, X., Yin, K., Li, Z., Sfeir, M. M., & Liu, C. (2021). Sensi- SARS-CoV-2 spike protein. Theranostics, 11(2), 649–664.
tive quantitative detection of SARS-CoV-2 in clinical samples 68. Blanchard, E. L., et al. (2021). Treatment of influenza and
using digital warm-start CRISPR assay. Biosens Bioelectron, 184, SARS-CoV-2 infections via mRNA-encoded Cas13a in rodents.
113218. Nature Biotechnology, 64, 4893.
47. Teng, F., et al. (2019). CDetection: CRISPR-Cas12b-based DNA 69. Subbarao, K., & Roberts, A. (2006). Is there an ideal animal
detection with sub-attomolar sensitivity and single-base specific- model for SARS? Trends in Microbiology, 14(7), 299–303.
ity. Genome Biology, 20(1), 132. 70. Ni, W., et al. (2020). Role of angiotensin-converting enzyme 2
48. Joung, J., et al. (2020). Detection of SARS-CoV-2 with SHER- (ACE2) in COVID-19. Critical Care, 24(1), 422.
LOCK One-Pot Testing. New England Journal of Medicine,
383(15), 1492–1494.
13
Molecular Biotechnology (2023) 65:146–161 161
71. Lutz, C., Maher, L., Lee, C., & Kang, W. (2020). COVID-19 81. Flynn, R. A., et al. (2021). Discovery and functional interroga-
preclinical models: Human angiotensin-converting enzyme 2 tion of SARS-CoV-2 RNA-host protein interactions. Cell, 184(9),
transgenic mice. Human Genomics, 14(1), 20. 2394-2411.e16.
72. Sun, S. H., et al. (2020). A mouse model of SARS-CoV-2 infec- 82. Lu, F., & Tellier, M. (2020). CRISPRing for host genes regulat-
tion and pathogenesis. Cell Host & Microbe, 28(1), 124-133.e4. ing SARS-CoV-2. Nature Reviews Immunology. https://doi.org/
73. Agrotis, A., & Ketteler, R. (2015). A new age in functional 10.1038/s41577-020-0400-
genomics using CRISPR/Cas9 in arrayed library screening. 83. Hoffmann, H. H., et al. (2020). Functional interrogation of a
Frontiers in Genetics, 6, 300. SARS-CoV-2 host protein interactome identifies unique and
74. Francis Stewart, A., & von Melchner, H. (2019). New methods shared coronavirus host factors. Cell Host & Microbes, 5, 4921.
for extracting function from the mammalian genome. Methods, 84. Daniloski, Z., et al. (2020). Identification of required host factors
164–165, 1–2. for SARS-CoV-2 infection in human cells. Cell. https://doi.org/
75. Ludwig, M. L., et al. (2018). Generation and utilization of 10.1016/j.cell.2020.10.030
CRISPR/Cas9 screening libraries in mammalian cells. In K. 85. Hoffmann, H. H., et al. (2021). TMEM41B is a pan-flavivirus host
Appasani (Ed.), Genome editing and engineering: From TAL- factor. Cell, 184(1), 133-148.e20.
ENs, ZFNs and CRISPRs to molecular surgery (pp. 223–234). 86. Schneider, W. M., et al. (2021). Genome-scale identification of
Cambridge University Press. SARS-CoV-2 and pan-coronavirus host factor networks. Cell,
76. Yu, J. S. L., & Yusa, K. (2019). Genome-wide CRISPR-Cas9 184(1), 120-132.e14.
screening in mammalian cells. Methods, 164–165, 29–35. 87. Wang, R., et al. (2021). Genetic screens identify host factors for
77. Ma, H., et al. (2015). A CRISPR-based screen identifies genes SARS-CoV-2 and common cold coronaviruses. Cell, 184(1), 106-
essential for west-nile-virus-induced cell death. Cell Reports, 119.e14.
12(4), 673–683. 88. Baggen, J., et al. (2021). Genome-wide CRISPR screening iden-
78. Savidis, G., et al. (2016). Identification of Zika virus and dengue tifies TMEM106B as a proviral host factor for SARS-CoV-2.
virus dependency factors using functional genomics. Cell Reports, Nature Genetics, 53(4), 435–444.
16(1), 232–246. 89. Yang, Q., et al. (2020). Inhibition of SARS-CoV-2 viral entry
79. Park, R. J., et al. (2017). A genome-wide CRISPR screen identifies upon blocking N- and O-glycan elaboration,". Elife. https://doi.
a restricted set of HIV host dependency factors. Nature Genetics, org/10.7554/eLife.61552
49(2), 193–203.
80. Wei, J., et al. (2020). Genome-wide CRISPR screens reveal host Publisher's Note Springer Nature remains neutral with regard to
factors critical for SARS-CoV-2 infection. Cell. https://doi.org/ jurisdictional claims in published maps and institutional affiliations.
10.1016/j.cell.2020.10.028
13