Journal of Life Science and Biomedicine  
J Life Sci Biomed, 10 (1): 01-09, 2020  
ISSN 2251-9939  
CRISPR/Cas9 gene editing technology and its  
application to the coronavirus disease (COVID-19),  
a review  
Saeid CHEKANI-AZAR1, Ehsan GHARIB MOMBENI2, Mastewal BIRHAN3 and Mahshad YOUSEFI4  
1PhD, Faculty of Veterinary Medicine, Animal Physiology, Atatürk University, Turkey  
2PhD, Department of Pathobiology, Shahid Chamran University of Ahvaz, Iran  
3PhD, College of Veterinary Medicine and Animal Science, Department of Veterinary Paraclinical Studies, University of Gondar, Ethiopia  
4MD, Hamadan University of Medical Sciences, Hamadan, Iran  
Corresponding author’s Email: maste675@gmail.com;  
ABSTRACT  
Review Article  
Introduction. Clustered-Regularly Interspaced Short Palindromic Repeats (CRISPR), and  
CRISPR associated (Cas) protein (CRISPR/Cas) structures were first identified in E. coli in  
1987 and guard prokaryotic cells from any invading pathogens, harmful events and plasmids  
by recognizing and cutting foreign nucleic acid sequences that contain short palindromic  
repeats spacer sequences. Several genome editing approaches have been developed based on  
these mechanisms; the most recent is known as CRISPR/Cas. Before the CRISPR technique  
was revealed in 2012, editing the genomes of plants and animals took many years and cost  
hundreds of thousands of dollars. Thus, CRISPR/Cas has attracted significant interest in the  
scientific community, especially for disease diagnosis and treatment, as it is quicker, less  
expensive and more precise than other genome editing approaches. The evidence from gene  
mutations in specific patients generated using CRISPR/Cas can assist in the prediction of  
the optimal treatment schedule for individual patients and for innovation purposes in other  
researches like replication in cell culture of coronaviruses like severe acute respiratory  
syndrome coronavirus-2 (SARS-CoV2 or COVID-19). However, in numerous situations, the  
effects of the furthermost significant driver mutations are not yet understood and  
interpretation of the optimal treatment is impossible. CRISPR/Cas classifications feature  
highly sensitive and selective tools for the detection of various target genes. When we see  
the next steps of genomic research, it is obvious that genome-wide association studies are  
relatively new way to identify the genes involved in human disease. Furthermore,  
CRISPR/Cas provides a tool to manipulate non-coding regions and will thus accelerate  
examination of these poorly characterized regions of the genome and play a vital role in the  
progress of whole genome libraries. Aim. We aimed to review the history of CRISPR/Cas, the  
mechanisms of CRISPR techniques, its current status as a tool for studying both natural  
mutations and genomic manipulations, and explore how CRISPR/Cas may improve the  
treatment of diseases.  
PII: S225199392000001-10  
Rec. 22 December 2019  
Rev. 15 January 2020  
Pub. 25 January 2020  
Keywords  
CRISPR/Cas9,  
DNA-targeting,  
Palindromic,  
Plasmids,  
Genome sequencing,  
SARS-CoV2,  
COVID-19  
INTRODUCTION  
Clustered regularly interspaced short palindromic repeats (CRISPR) were first discovered in Escherichia coli in  
1987 and later found in other bacteria species. The role of these repeat sequences remained unclear until, in  
2005, several researchers described the similarities of the sequences` DNA, leading to the hypothesis that the  
sequences are part of adaptive immune system in bacteria (CRISPR/Cas9 for cancer research and therapy).  
As you know, CRISPR is also refers to an adaptive immune response in bacteria and archaea that is cast-off  
to target and cut down viral DNA by using endonuclease in specific ways. By reengineering this immune  
response to target parts of genetic material, scientists could make extremely precise genetic alterations tailored  
to the type of cell. This is the basis of CRISPR therapeutic and diagnostic platforms [1].  
CRISPR and CRISPR-associated proteins characterize the immune system of archaea and bacteria, and  
deliver protection against invasive nucleic acids, DNA, or RNA from phages, plasmids, and other exogenous  
DNA elements. At this time, two different classes, six types, and 21 subtypes of CRISPRCas systems have been  
identified [2]. The length of the repeat sequences varies between 25 and 40 nt, whereas the length of the spacer  
sequences varies between 21 and 71 nt [3].  
Citation: Chekani-Azar S, Gharib Mombeni E, Birhan M, and Yousefi M. CRISPR/Cas9 gene editing technology and its application to the coronavirus disease  
For the success of these, innumerable methods have been fashioned for targeted gene editing in cell and  
animal models. These include Zinc-Finger Nucleases (ZFNs), Transcription Activator-Like Effector Nucleases  
(TALENs). But, Clustered-Regularly Interspaced Short Palindromic Repeats (CRISPR), and CRISPR Associated  
(Cas) protein (CRISPR/Cas) is a rapid, simple, and often extremely efficient gene editing method [4]. That is why  
it is a powerful method for making changes and simple to manipulate the genome of many organisms [5]. Form  
these scientific truth, we can understand the advantage of genome editing tools made possible to modify a  
genomic DNA in a targeted fashion by applying of CRISPR/Cas [6].  
Recent developments in CRISPR/Cas gene editing have made it possible to introduce precise changes into  
a wide variety of genomes, including those of dsDNA viruses; in addition to this, it is pursuing relies on the co-  
expression of a prokaryotic Cas nuclease and an associated guide RNA (gRNA) sequence. Because, once gRNA-  
targeted Cas, it creates a double-stranded break (DSB) in the genome, by making of two main repair processes  
compete to repair the damage, leading to with some modifications of from the original sequence [7].  
The study of gene function in the recent decades has trusted on the generation of systemic and conditional  
gene knockouts to disrupt gene expression especially in coronaviruses. For example, de Wilde et al. [8]  
investigated the cyclophilin A (CypA) protein-dependence in the replication of three related nidoviruses in the  
same cell line (Huh7), in which CypA expression was knocked-out using CRISPR/Cas9 gene editing technology  
(Huh7-CypAKO cells). Therefore, even if successful studies and some innovation ideas and solution were done,  
but, it has been challenging to knockout a gene in specific cell types in the brain. The final target and goals of  
CRISPR was to revolutionize the treatment of hereditary diseases based therapeutics. Finally, the aim of this  
systematic review paper is to summarize the application of CRISPR/Case and its overall immune signal  
transduction and also discussing gene editing technology against novel diseases like coronavirus (COVID-19).  
Current status and significance of CRISPR/CAS and immune cells  
Types of CRISPR/Cas systems  
CRISPR/Cas systems have been categorized in details such as class 1 which utilizes multi-protein effector  
complexes and class 2 which utilizes single-protein effectors. Class 1 is further divided into different types I, III,  
and IV, while class 2 includes types II, V, and VI. It can also be divided into 19 different subtypes and it is likely to  
continue to expand as new CRISPR/Cas systems are identified. The greatest corporate Cas protein used for  
functional broadcast is a type II single-protein effector derived from Streptococcus pyogenes (SpCas9). It is a type  
of guide RNA to succor in successfully cleaving the target gene [9].  
This choice stems from the fact that types I and III are composed of a number of Cas proteins, although the  
Cas9 protein is the only protein constituent of type II CRISPR/Cas systems [10]. One of the difficulties in  
classifying and applying for the success is designing and construction of novel CRISPR/Cas systems with the  
researcher effort to demarcate targeting sequences in a simplified manner [11]. Type II system is composed of  
trans-activating crRNA (tracrRNA). CRISPR loci are prepared of “repeat-spacer” array and adjacent to small  
clusters of Cas genes including Cas9 endonuclease [12]. From this perspective, Cas proteins are of great  
necessity for the functioning of CRISPR/Cas immune system and are considered as indicators of the system’s  
activity [13].  
CRISPR in human pluripotent stem cells  
Human civilization has always faced two basic challenges: That are disease and hunger. Efforts to prevent  
or treat disease and accomplish food security endure. Despite of the progress made food insecurity and many.  
Diseases and pests persist; these problems have proved difficult to solve using available technologies.  
Nevertheless, hopes are high for solutions to many of these challenges with the advent of CRISPR technology  
[14]. There are different human cells those are, immortalized MRC5-hTERT fibroblasts were cultured in  
Dulbecco's modified Eagle medium, supplemented with fetal bovine serum (10%), 1% penicillin-streptomycin and  
4.5 g/L glucose, [7].  
Overview of CRISP-Cas-mediated genome editing  
Engineered nucleases, such as ZFN, TALEN, and Cas9, can induce DNA double-strand breaks (DSBs) at  
specific genomic loci, which are subsequently repaired by one of at least two endogenous cellular DNA repair  
pathways: non-homologous end joining (NHEJ) and homology-directed repair (HDR) [15]. Over the past two  
decades, the discovery of programmable DNA binding proteins (ZFs, Zinc fingers, and transcription activator-  
Citation: Chekani-Azar S, Gharib Mombeni E, Birhan M, and Yousefi M. CRISPR/Cas9 gene editing technology and its application to the coronavirus disease  
like effectors [TALEs]) has paved the way for the development of genome editing tools. Compared with ZFs,  
which contain repeated Cys2-His2 (C2H2), DNA binding domain (DBD) adapted from a family of eukaryotic  
transcription factors (TFs) [16].  
The CRISPR/Cas9 genome-editing technology is derived from the adaptive immune response in  
prokaryotes. This method has several advantages over conventional genome-editing technologies, including: a)  
simplicity in target designing, b) regulation, and c) the ability to target multiple genes. CRISPR consists of two  
major components: sgRNA and Cas protein. The sgRNA consists of a sequence, also called as scaffold. sgRNA  
includes a Cas enzyme binding site and a spacer sequence, called the target sequence, which is specific to the  
gene of interest. The spacer sequence contains ~20 nucleotides and is followed by protospacer adjacent motif  
(PAM), a 26-base pair DNA sequence immediately following the DNA sequence targeted by the Cas9 nuclease  
in the CRISPR. PAM sequences vary based upon the type of Cas9 enzyme [17].  
Figure 1. Promoting safety in genome editing [18].  
Editing efficiency  
There are a number of changes and factors that contribute for the difficulty of genome editing. The second  
major challenge in the genome editing is efficiency. The therapeutic efficacy of genome editing will be enhanced  
by improving the editing rates. For CRISPR/Cas, the activity of double-stranded break (DSB) restoration  
pathways plays an important regulatory role in its editing rates.  
Due to the different endogenous cellular DNA repair mechanisms mediated by non-homologous end-  
joining (NHEJ) and homology-directed repair (HDR), there are definitely more editing efficiency differences in  
both different cell types and also cellular states. HDR is differently operates during the S/G2 phase while NHEJ  
is active during the whole cell cycle, thus, it is generally more efficient for generating indels to knockout  
carcinogenic genes than HDR. The efficiency of HDR, as precise gene adjustment, is relatively lethargic  
depending on both homology arm length and DNA template type. HDR templates are typically single-stranded  
oligonucleotides or plasmids containing alleles and delivery of HDR templates has been accomplished with viral  
or non-viral vectors [19].  
CRISPR/Cas9 toolsets  
There are a number of online CRISPR tools and resource available, provided by both academic and  
commercial institutions, with a list and its application of the most widely used resources provided in Table 1.  
The function of these CRISPR resources is primarily to identify and recognized gRNAs, and provide the  
essential information required to select gRNAs for experimental applications. While some of the tools may be  
able to “mention” gRNAs for specific purposes. However for most applications, these CRISPR tools are just one  
component of the overall design process that starts with the identification of target locations [20].  
Citation: Chekani-Azar S, Gharib Mombeni E, Birhan M, and Yousefi M. CRISPR/Cas9 gene editing technology and its application to the coronavirus disease  
Table 1. List of common CRISPR resources [20].  
Name  
URL  
Addgene  
Benchling  
BreakingCas  
Broad Institute GPP  
CHOPCHOP  
CRISPOR  
CrisFlash  
Deskgen  
E-CRISP  
https://sg.idtdna.com/site/order/designtool/index/CRISPR_CUSTOM  
Horizon Discovery  
IDT  
Microsoft Research CRISPR  
RGEN Tools  
Synthego  
WTSI Genome Editing (WGE)  
WTSI: Wellcome Trust Sanger Institute; RGEN: RNA-Guided Endonucleases; GPP: Green Public Procurement,  
CRISPR applications and design  
For the applications systems of all CRISPR/Cas is depend on the gRNA’s ability to recognize specific target  
sequence, even though not all applications are dependent on the cleavage activity of the Cas-endonuclease.  
Because of both knock-in (KI) and knock-out (KO) solicitations are related to the formation of a DSB to insert  
DNA or either delete [20, 21]. Designing needs about the basic principles of CRISPR that involved to identifying  
a suitable target region or regions, tracked by the choices of suitable gRNAs based on positioning the target  
region: off-target and on-target scoring. This is very important for the applies of both targeted design and  
large-scale design approaches, even though the methods used for selecting and targeting gRNAs can be exact  
different between these approaches [20].  
There are invaluable information for systematic analyses of the genes which can be achieved by  
parameters like molecular characterization, mechanics behind biological processes, and screening of therapies  
and responses to drugs [22]. When we see for example like cancer: it is a genetic disease stemming from  
cumulative genetic/epigenetic aberrations, it is rational for us to envisage that modifying the oncogenic  
genome abnormalities through CRISPR/Cas might characterize a promising therapeutic strategy against  
cancer [19]. Introducing only a single DSB to stimulate the DNA-repair mechanisms can be sufficient, but even  
introducing more than one DSB for large deletions or insertions may be necessary. In the lack of a repair  
template, NHEJ pathway may introduce small idles within a coding sequence, which can disrupts protein  
translation by a frame shift and thus knockout a gene. An ideas suitable for DNA repair template is on condition  
that, the same double-stranded break (DSB) may be repaired by homology-directed repair (HDR), leading to the  
insertion or knock-in of any exogenous DNA included within the template. In contrast, CRISPR repression and  
activation (CRISPRa/i) uses a catalytically dead or inactivated Cas-endonuclease to target transcriptional  
repressor complexes or activator to a specific site within the promoter region [20].  
Today, the application of the CRISPR/Cas9 method has expanded into a variety of areas, including  
agriculture, husbandry, disease patterns and targeted therapies. This section emphasizes the therapeutic  
aspects for diseases with a genetic basis, especially monogenic disorders. In the ex vivo approach, the targeted  
cell population is removed from the patient’s body, and the designed nucleases are used to induce the desired  
changes and then returned to the patient by grafting. Avoiding issues such as transplant rejection and immune  
responses are important benefits of this method. Using in vivo gene therapy, the genomic modifying factors,  
such as programmatic nucleases and patterns, are put directly into the patient [23].  
In the case of Cas9; it is used to change endogenous genes which previously were problematic to be  
genetically manipulated, resulting in a more efficient and faster production of transgenic models. Those  
transgenic models that can be produced simply by injecting fertilized zygotes with customized Cas9 and  
transcribed single guide RNA for the manipulation of alleles and accomplish desired alterations in mammalians.  
Cas9 is also used in cellular models to study for the treatment and diagnosis of polygenic diseases such as  
diabetes and autism due to multiplexing properties [24].  
Citation: Chekani-Azar S, Gharib Mombeni E, Birhan M, and Yousefi M. CRISPR/Cas9 gene editing technology and its application to the coronavirus disease  
CRISPR/Cas9 gene editing technology applications against diseases like COVID-19  
CRISPR/Cas biotechnology is now expansively and broadly applied across numerous disciplines, including  
basic sciences, food/crop development, fuel generation, drug development, human genome engineering and  
gene editing technology against novel diseases like coronavirus (COVID-19 or SARS-CoV2) [8, 14, 19, 22, 25, 26].  
The Nidovirales that are consisted of the ronivirus, mesonivirus, arterivirus, and coronavirus families have  
minor or major economic and societal effects. In 20022003, the highly pathogenic outbreaks (ongoing), human  
coronaviruses (HCoVs), severe acute respiratory syndrome coronavirus (SARS-CoV) and Middle East  
respiratory syndrome coronavirus (MERS-CoV) occurred in zoonotic and transmitted to the human population,  
with alarming mortality. SARS-CoV outbreaks led to more than 8000 confirmed cases within a few months,  
(mortality rate: ~ 10%) [27] and for the MERS-CoV resulted in over 2000 human cases (~ 35% mortality rate)  
(http://www.who.int/emergencies/mers-cov/en/) [28]. But in December 2019, another pathogenic HCoV, 2019  
named novel coronavirus (2019-nCoV, COVID-19 or SARS-CoV2), was recognized in Wuhan, China [29, 30], and  
has caused serious mortality rate of ~ 35% (https://www.worldometers.info/coronavirus/). In most patients,  
COVID-19 infection is associated with a cytokine storm [27, 28, 30-33]. In recovered cases, the excessive immune  
responses lead to long-term lung damage and fibrosis, causing functional disability and reduced quality of life  
[34, 35]. There is no specific anti-viral treatment, and designing effective antiviral drugs against COVID-19 will  
take several years to develop and evaluate and also a range of existing host-directed safe therapies [36-38] could  
potentially be repurposed to treat COVID-19 infection. Hence, an urgent funding and scientific investments into  
advancing novel therapeutic interventions for coronavirus infections, is required.  
Therefore, the lack of proper strategies to control nidovirus-induced disease, increase the importance of  
knowledge of these virusesreplications and also their interactions with the host cell and specially role of  
CRISPR-Cas gene editing technology against these viruses. Nidoviruses are +RNA viruses with different  
genomes (arteriviruses with 13 to 16 kb, and coronaviruses with 2634 kb) [39, 40], with a complex genome  
translation for producing the nsps (polyprotein precursors of the nonstructural proteins) as well as a nested set  
of subgenomic (sg) mRNAs to express the structural proteins [41, 42]. Nidoviral nsps, probably with various host  
factors, assemble into replication and transcription complexes (RTCs) that drive viral RNA synthesis [43-46].  
These RTCs are probably associated with a virus-induced network of endoplasmic reticulum-derived membrane  
(ERDM) structures, including many double-membrane vesicles [43, 47-49].  
Nidovirus replication depends on membrane trafficking, cellular proteins and membranes, and host  
signaling pathways or host cell factors and processes [50-52]. The cyclophilin (Cyp) protein family members  
have been implicated in nidovirus replication. The Cyps family of peptidyl-prolyl isomerases (PPIases), act as  
chaperones to facilitate protein folding, trafficking and immune cell activation [53, 54]. Cyps family that are  
ubiquitous and abundant cytosolic proteins, are especially expressed CypA, an important factor that implicated  
in the replication of various RNA viruses. CypA have also specific roles in various viruses like human hepatitis C  
virus (HCV) and immunodeficiency virus-1 (HIV-1) infections. It assists HCV polyprotein processing, interacts  
with HCV NS5A for remodelling of cellular membranes into HCV replication organelles, and stabilizes HIV-1  
capsids to promote nuclear import of the HIV-1 genome [55].  
Studies with general Cyp inhibitors such as cyclosporine A (CsA) showed that, cyclophilins were initially  
implicated as host factors in nidovirus replication. In cell culture, the replication of a variety of arteriviruses and  
coronaviruses was found to be strongly inhibited by lowmicromolar concentrations of CsA and the non-  
immunosuppressive CsA analogs Alisporivir (ALV) and NIM-811 [56-63]. And then nidovirus replication can  
depend specifically on CypA and/or CypB.  
The replication in cell culture of the alphacoronaviruses feline coronavirus [64], the arterivirus equine  
arteritis virus (EAV) [65]; and human coronavirus (HCoV)-NL63 [56], and HCoV-229E [63] was reported to be  
affected by CypA knockout or knockdown. Finally, it seems that the normally cytosolic CypA was found to co-  
sediment with membrane structures containing EAV RTCs, have a direct association with the arteriviral RNA-  
synthesizing machinery [65].  
All the studies are depends on the nidoviruses types and cell lines, CypA expression levels, and readouts to  
measure viral replication efficiency. de Wilde et al. [8] investigated the CypA-dependence of the replication of  
three related nidoviruses in the same cell line (Huh7), in which CypA expression was knocked-out using  
CRISPR/Cas9 gene editing technology (Huh7-CypAKO cells). Using different cell lines, the replication of the  
arterivirus EAV [65] and the alphacoronavirus HCoV-229E [63], was previously concluded to depend on CypA.  
And the CypA dependence of betacoronaviruses like MERS-CoV has been documented for a first time with de  
Wilde et al. [8]. They reported that infection of Huh7-CypAKO cells with MERS-CoV revealed that its replication  
Citation: Chekani-Azar S, Gharib Mombeni E, Birhan M, and Yousefi M. CRISPR/Cas9 gene editing technology and its application to the coronavirus disease  
was only modestly affected by the absence of CypA, as opposed to equine arteritis virus which was strongly  
inhibited. While they stated that, HCoV-229E replication was not affected at all in the Huh7-CypAKO cells. They  
thus revealed major differences in CypA dependence of the arterivirus EAV in compared to other coronaviruses.  
There is a need to more evaluation of the CypA role in the replication of members of the latter virus family.  
The capacity of CRISPR diagnostics  
Diagnostic uses of CRISPR technologies help immense advantages in clinical effectiveness. A vast numbers  
of researchers have used CRISPR-based editing and diagnosis to correct the genetic basis of many diseases in  
isolated from the cells of animal models. The first groundswell of clinical trials using CRISPR enzymes to treat  
inherited disorders in humans involves eradicating a patient’s cells, editing ex vivo, and rein fusing the  
corrected cells. Such ex vivo genome editing is currently the most technically feasible approach, and has the  
potential to treat devastating blood disorders like sickle cell disease and β-thalassemia. The ex vivo strategy also  
underlies cancer immunotherapies [66].  
The role and function of CRISPR/Cas in gene editing in applications as diverse as fetal medicine,  
biodefense, and synthetic food makes for a stream of high-profile news. In 2018, several articles were published  
on the use of CRISPR in diagnostic tests for early-stage cancer detection or for infectious diseases. Further, it  
was seen as the most important innovation used to develop a rapid CRISPR-based assay for TB detection [25].  
CRISPR ethics  
Ethical decisions, particularly in biomedicine, are empirically informed and involve evaluating potential  
risk-benefit ratios, with the attempt of maximizing benefits while minimizing risk. To navigate ethical decision  
making, it is critical to consider the range of possible consequences, the probabilities of each instantiating, and  
the possible rationalizations driving results. The ethical concerns about CRISPR genome engineering  
technology are largely due to at least three important explanations. These include the possibilities of limited on-  
target editing efficiency [67].  
CRISPR/Cas9 is inexpensive, efficient and exact method to edit genes at the level of individual nucleotides,  
and also help to explore or explain many scientific questions. Moreover, this gene editing technology provides  
new potential treatments for many human diseases like novel coronavirus (COVID-19). In addition, the use of  
CRISPR/Cas9 gene editing technologies along with stem cells (i.e. induced pluripotent stem cells), can help to  
generate gametes for reproductive purposes or correct errors in their genome, and can also minimize the need  
for oocyte donation [68].  
In 2017, the US National Academies of Sciences, Engineering, and Medicine Committee on Human Gene  
Editing published reviews of scientific, legal or ethical concerns about the amazing progress of gene  
engineering technology. The astonishing report, was that heritable genome editing help to modification of the  
germ line with the aim of generating a new human being who could therefore transfer the genomic change to  
future generations-hold be impermissible now but eventually could be justified for certain medical indications.  
Currently, create, destroy, or modification of human embryos to include heritable genetic changes for research  
purposes, is unlawful for U.S. federal funds. The NASEM is still implies that if safety risks guaranteed, clinical  
trials conceivable would commence [69].  
CONCLUSIONS AND RECOMMENDATIONS  
Form these systems review; we can appreciate the most important components of CRISPR RNAs (crRNAs) and  
Cas effector proteins. CRISPR will be a very important and crucial study area for disease diagnosis and  
treatment in the future, with the best potential for research in the scientific community. Beside this, it will be  
important for correcting the mutations and hereditary diseases concerning immune cells and their system  
disturbances; it will be important even for immunological tolerance to correct wrongly activated immune cells  
that fail to identify self and non-self, leading to cancerous disturbances of immune balance. It is very crucial in  
facilitating the etiology of diseases and checking their causative agents. From its adaption, it has revolutionized  
molecular biology and genetics in general. In combination with parallel developments of the necessary  
supportive techniques, modern biotechnology has led to useful genetic modifications of micro-organisms,  
plants and animals it has generated, inter alia, various new therapeutics and diagnostics. Owing to such  
advancements, we can study and work with this environment in an easier manner. CRISPR gene editing and  
Citation: Chekani-Azar S, Gharib Mombeni E, Birhan M, and Yousefi M. CRISPR/Cas9 gene editing technology and its application to the coronavirus disease  
correcting methods are used for this purpose. Based on these, we can forward some recommendations  
pertaining to future studies related to CRISPR-Cas:  
All researchers, academicians, international NGOs & funders, the scientific community as well as the  
government should focus on CRISPR/Cas-based diagnostics to enable this.  
In the future, we can also focus on pharmacogenomics. This will allow for the development of designer  
drugs to treat a wide range of health problems and disorders occurring as a result, including cardiovascular  
diseases, Alzheimer`s, HIV/AIDS, asthma, cancer and the worldwide pandemics like coronavirus (COVID-19).  
DECLARATIONS  
Authors' contributions  
S.Chekani-Azar, E.Gharib Mombeni and *M.Birhan conceived the review, and M.Yousefi coordinated the  
overall activity and article processing.  
Conflict of interest  
The authors declare that there is no conflict of interest.  
Acknowledgment  
The authors’ heartfelt thanks are given to University of Gondar, Research and Community Service  
V/President Office, Ethiopia; Collage of Veterinary Medicine and Animal Sciences, Atatürk University, Erzurum,  
Turkey; Department of Pathobiology, Shahid Chamran University of Ahvaz, Iran; and Hamadan University of  
Medical Sciences, Hamadan, Iran and finally Journal of Life Science and Biomedicine for the resource  
supporting and free of charge publication of the review article.  
REFERENCES  
1. Pan A and Kraschel KL. Kraschel. CRISPR diagnostics: Underappreciated uses in perinatology. in Seminars in  
perinatology. 2018. Elsevier. DOI: https://doi.org/10.1053/j.semperi.2018.09.016.  
2. Hidalgo-Cantabrana C, Goh YJ and Barrangou R. Barrangou, Characterization and repurposing of type I and type II  
CRISPR-Cas  
systems  
in  
bacteria.  
Journal  
of  
molecular  
biology,  
2018;  
431(1):  
21-33.  
DOI:  
3. Qu D, Lu S, Wang P, Jiang M, Yi S, et al. Analysis of CRISPR/Cas system of Proteus and the factors affected the  
functional mechanism. Life sciences, 2019; 231: 116531. DOI: https://doi.org/10.1016/j.lfs.2019.06.006  
4. Mehravar M, Shirazi A, Nazari M and Banan M. Mosaicism in CRISPR/Cas9-mediated genome editing. Developmental  
5. Zhan T, Rindtorff N, Betge J, Ebert MP and Boutros M. CRISPR/Cas9 for cancer research and therapy. in Seminars in  
cancer biology. 2019; 55(2019): 106-119. DOI: https://doi.org/10.1016/j.semcancer.2018.04.001.  
6. Herai RH. Avoiding the off-target effects of CRISPR/cas9 system is still a challenging accomplishment for genetic  
transformation. Gene, 2019; 700: 176-178. DOI: https://doi.org/10.1016/j.gene.2019.03.019  
7. King MW and Munger J. Editing the human cytomegalovirus genome with the CRISPR/Cas9 system. Virology, 2019;  
8. de Wilde AH, Zevenhoven-Dobbe JC, Beugeling C, Chatterji U, de Jong D, et al. Coronaviruses and arteriviruses display  
striking differences in their cyclophilin a-dependence during replication in cell culture. Virology, 2018; 517: 148-156.  
9. Ford K, McDonald D and Mali P. Functional genomics via CRISPRCas. Journal of Molecular Biology, 2019; 431(1): 48-65.  
10. Makarova KS, Haft DH, Barrangou R, Brouns SJ, Charpentier E, et al. Evolution and classification of the CRISPRCas  
systems. Nature Reviews Microbiology, 2011; 9(6): 467. DOI: https://doi.org/10.1038/nrmicro2577  
11. Khatodia S, Bhatotia K, Passricha N, Khurana S and Tuteja N. The CRISPR/Cas genome-editing tool: application in  
improvement of crops. Frontiers in plant science, 2016; 7: 506. DOI: https://doi.org/10.3389/fpls.2016.00506  
12. Yamaguchi Handde Lecea L. In vivo cell type-specific CRISPR gene editing for sleep research. Journal of neuroscience  
methods, 2018; 15(316): 99-102. DOI: https://doi.org/10.1016/j.jneumeth.2018.10.016. Epub 2018 Nov 12.  
13. Deveau H, Barrangou R, Garneau JE, Labonté J, Fremaux C, et al. Phage response to CRISPR-encoded resistance in  
Streptococcus thermophilus. Journal of bacteriology, 2008; 190(4): 1390-1400. DOI: https://doi.org/10.1128/JB.01412-07  
14. Ogaugwu CE, Agbo SO and Adekoya MA. CRISPR in sub-Saharan Africa: applications and education. Trends in  
biotechnology, 2019. 37(3): p. 234-237. DOI: https://doi.org/10.1016/j.tibtech.2018.07.012  
15. Nishiyama J. Genome editing in the mammalian brain using the CRISPRCas system. Neuroscience research, 2019; 141:  
16. Rahman S, Datta M, Kim J and Jan AT. CRISPR/Cas: An intriguing genomic editing tool with prospects in treating  
neurodegenerative diseases. in Seminars in cell & developmental biology. 2019. Elsevier. 96: 22-31. DOI:  
Citation: Chekani-Azar S, Gharib Mombeni E, Birhan M, and Yousefi M. CRISPR/Cas9 gene editing technology and its application to the coronavirus disease  
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
17. Kaushik I, Ramachandran S and Srivastava SK. CRISPR-Cas9: A multifaceted therapeutic strategy for cancer treatment.  
in Seminars in cell & developmental biology. Elsevier. 2019; 96:4-12. DOI: https://doi.org/10.1016/j.semcdb.2019.04.018.  
18. Pineda M, Lear A, Collins JP and Kiani S. Safe CRISPR: Challenges and Possible Solutions. Trends in biotechnology, 2018;  
19. Chen M, Mao A, Xu M, Weng Q, Mao J, et al. CRISPR-Cas9 for cancer therapy: Opportunities and challenges. Cancer  
20. Thomas M, Parry-Smith DandIyer V. Best practice for CRISPR design using current tools and resources. Methods, 2019;  
21. Fu BXH, Smith JD, Fuchs RT, Mabuchi M, Curcuru J, et al. Target-dependent nickase activities of the CRISPR-Cas  
nucleases Cpf1 and Cas9. Nature microbiology, 2019; 4(5): 888-897. DOI: https://doi.org/10.1038/s41564-019-0382-0  
22. Shalem O, Sanjana NE, Hartenian E, Shi X, Scott DA, et al. Genome-scale CRISPR-Cas9 knockout screening in human  
cells. Science, 2014; 343(6166): 84-87. DOI: https://doi.org/10.1126/science.1247005  
23. Rezaei H, Farahani N, Hosseingholi EZ, Sathyapalan T and Hossein Sahebkar A. Harnessing CRISPR/Cas9 technology in  
cardiovascular  
disease.  
Trends  
in  
cardiovascular  
medicine,  
2019;  
30(2):  
93-101.  
DOI:  
24. Wang W, Zhang L, Wang X and Zeng Y. The advances in CRISPR technology and 3D genome. in Seminars in cell &  
developmental biology. 2018 Elsevier. DOI: https://doi.org/10.1016/j.semcdb.2018.07.009.  
25. Ai J-W, Zhou X, Xu T, Yang M, Chen Y, et al. CRISPR-based rapid and ultra-sensitive diagnostic test for Mycobacterium  
tuberculosis. Emerging microbes & infections, 2019; 8(1): 1361-1369. DOI: https://doi.org/10.1080/22221751.2019.1664939  
26. Strich JR and Chertow DS. CRISPR-Cas biology and its application to infectious diseases. Journal of Clinical  
Microbiology, 2019; 57(4): e01307-18. DOI: https://doi.org/doi.org/10.1128/JCM.01307-18  
27. Hui DS and Zumla A. Severe Acute Respiratory Syndrome: Historical, Epidemiologic, and Clinical Features. Infectious  
Disease Clinics, 2019; 33(4): 869-889. DOI: https://doi.org/10.1016/j.idc.2019.07.001  
28. Azhar EI, Hui DS, Memish ZA, Drosten C and Zumla A. The Middle East Respiratory Syndrome (MERS). Infectious  
Disease Clinics, 2019; 33(4): 891-905. DOI: https://doi.org/10.18683/germs.2019.1155  
29. Organization  
WH.  
Novel  
Coronavirus  
(2019-nCoV):  
situation  
report,  
3.  
2020.  
30. Huang C, Wang Y, Li X, Ren L, Zhao J, et al. Clinical features of patients infected with 2019 novel coronavirus in Wuhan,  
China. The Lancet, 2020; 395(10223): 497-506. DOI: https://doi.org/10.1016/S0140-6736(20)30183-5  
31. Zumla A, Hui DS, Azhar EI, Memish ZA and Maeurer M. Reducing mortality from 2019-nCoV: host-directed therapies  
should be an option. The Lancet, 2020; 395(10224): e35-e36. DOI: https://doi.org/10.1016/S0140-6736(20)30305-6  
32. Li G, Fan Y, Lai Y, Han T, Li Z, et al. Coronavirus infections and immune responses. Journal of Medical Virology, 2020;  
33. Channappanavar Channappanavar R and Perlman S. Pathogenic human coronavirus infections: causes and  
consequences of cytokine storm and immunopathology. in Seminars in immunopathology. 2017 Springer; p. 529-539.  
34. Batawi S, Tarazan N, Al-Raddadi R, Al Qasim E, Sindi A, et al. Quality of life reported by survivors after hospitalization  
for Middle East respiratory syndrome (MERS). Health and quality of life outcomes, 2019; 17(1): 101. DOI:  
35. Ngai JC, Ko FW, Ng SS, TO KW, Tong M, et al. The longterm impact of severe acute respiratory syndrome on  
pulmonary function, exercise capacity and health status. Respirology, 2010; 15(3): 543-550. DOI:  
36. Zumla A, Chan JF, Azhar EI, Hui DS and Yuen K-Y. Coronavirusesdrug discovery and therapeutic options. Nature  
reviews Drug discovery, 2016; 15(5): 327. DOI: https://doi.org/10.1038/nrd.2015.37  
37. Beigel JH, Nam HH, Adams PL, Krafft A, Ince WL, et al. Advances in respiratory virus therapeuticsA meeting report  
from  
the  
6th  
isirv  
Antiviral  
Group  
conference.  
Antiviral  
research,  
2019.  
DOI:  
38. Zumla A, Azhar EI, Arabi Y, Alotaibi B, Rao M, et al. Host-directed therapies for improving poor treatment outcomes  
associated with the middle east respiratory syndrome coronavirus infections. 2015 Elsevier. DOI:  
39. Gorbalenya AE, Enjuanes L, Ziebuhr J and Snijder EJ. Nidovirales: Evolving the largest RNA virus genome. Virus  
40. Nga PT, del Carmen Parquet M, Lauber C, Parida M, Nabeshima T, et al. Discovery of the first insect nidovirus, a  
missing evolutionary link in the emergence of the largest RNA virus genomes. PLoS pathogens, 2011; 7(9). DOI:  
41. de Wit E, van Doremalen N, Falzarano D and Munster VJ. SARS and MERS: recent insights into emerging  
coronaviruses. Nature Reviews Microbiology, 2016; 14(8): 523-534. DOI: https://doi.org/10.1038/nrmicro.2016.81  
42. Snijder EJ, Kikkert MandFang Y. Arterivirus molecular biology and pathogenesis. Journal of General Virology, 2013;  
43. Gosert R, Kanjanahaluethai A, Egger D, Bienz K and Baker SC. RNA replication of mouse hepatitis virus takes place at  
double-membrane vesicles. Journal of virology, 2002; 76(8): 3697-3708. DOI: https://doi.org/10.1128/JVI.76.8.3697-  
3708.2002  
Citation: Chekani-Azar S, Gharib Mombeni E, Birhan M, and Yousefi M. CRISPR/Cas9 gene editing technology and its application to the coronavirus disease  
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
44. Hagemeijer MC, Vonk AM, Monastyrska I, Rottier PJ and de Haan CA. Visualizing coronavirus RNA synthesis in time by  
using click chemistry. Journal of virology, 2012; 86(10): 5808-5816. DOI: https://doi.org/10.1128/JVI.07207-11  
45. Pedersen KW, van der Meer Y, Roos N and Snijder EJ. Open Reading Frame 1a-Encoded Subunits of the Arterivirus  
Replicase Induce Endoplasmic Reticulum-Derived Double-Membrane Vesicles Which Carry the Viral Replication  
Complex. Journal of Virology, 1999; 73(3): 2016-2026. PMID: 9971782; PMCID: PMC104444  
46. van Hemert MJ, van den Worm SHE, Knoops K, Mommaas AM, Gorbalenya AE, et al. SARS-Coronavirus  
Replication/Transcription Complexes Are Membrane-Protected and Need a Host Factor for Activity In Vitro. PLOS  
47. Knoops K, Bárcena M, Limpens RW, Koster AJ, Mommaas AM, et al. Ultrastructural characterization of arterivirus  
replication structures: reshaping the endoplasmic reticulum to accommodate viral RNA synthesis. Journal of Virology,  
48. Maier HJ, Hawes PC, Cottam EM, Mantell J, Verkade P, et al. Infectious bronchitis virus generates spherules from  
zippered endoplasmic reticulum membranes. MBio, 2013; 4(5): e00801-13. DOI: https://doi.org/10.1128/mBio.00801-13  
49. Ulasli M, Verheije MH, de Haan CAM and Reggiori F. Qualitative and quantitative ultrastructural analysis of the  
membrane rearrangements induced by coronavirus. Cellular Microbiology, 2010; 12(6): 844-861. DOI: 10.1111/j.1462-  
5822.2010.01437.x  
50. de Wilde AH, Snijder EJ, Kikkert Mandvan Hemert MJ. Host factors in coronavirus replication, in Roles of Host Gene  
and Non-coding RNA Expression in Virus Infection. 2017 Springer; p. 1-42. DOI: https://doi.org/10.1007/82_2017_25  
51. van der Hoeven B, Oudshoorn D, Koster AJ, Snijder EJ, Kikkert M, et al. Biogenesis and architecture of arterivirus  
replication organelles. Virus Research, 2016; 220: 70-90. DOI: https://doi.org/10.1016/j.virusres.2016.04.001  
52. Zhong Y, Tan YW and Liu DX. Recent Progress in Studies of Arterivirus- and Coronavirus-Host Interactions. Viruses,  
53. Naoumov NV. Cyclophilin inhibition as potential therapy for liver diseases. Journal of Hepatology, 2014; 61(5): 1166-1174.  
54. Nigro P, Pompilio G and Capogrossi MC. Cyclophilin A: a key player for human disease. Cell Death & Disease, 2013; 4(10):  
55. Hopkins S and Gallay PA. The role of immunophilins in viral infection. Biochimica et Biophysica Acta (BBA) - General  
Subjects, 2015; 1850(10): 2103-2110. DOI: https://doi.org/10.1016/j.bbagen.2014.11.011  
56. Carbajo-Lozoya J, Ma-Lauer Y, Malešević M, Theuerkorn M, Kahlert V, et al. Human coronavirus NL63 replication is  
cyclophilin A-dependent and inhibited by non-immunosuppressive cyclosporine A-derivatives including Alisporivir.  
Virus Research, 2014; 184: 44-53. DOI: https://doi.org/10.1016/j.virusres.2014.02.010  
57. de Wilde AH, Falzarano D, Zevenhoven-Dobbe JC, Beugeling C, Fett C, et al. Alisporivir inhibits MERS- and SARS-  
coronavirus replication in cell culture, but not SARS-coronavirus infection in a mouse model. Virus Research, 2017; 228:  
58. de Wilde AH, Raj VS, Oudshoorn D, Bestebroer TM, van Nieuwkoop S, et al. MERS-coronavirus replication induces  
severe in vitro cytopathology and is strongly inhibited by cyclosporin A or interferon-α treatment. Journal of General  
Virology, 2013; 94(8): 1749-1760. DOI: https://doi.org/10.1099/vir.0.052910-0  
59. de Wilde AH, Zevenhoven-Dobbe JC, van der Meer Y, Thiel V, Narayanan K, et al. Cyclosporin A inhibits the replication  
of diverse coronaviruses. Journal of General Virology, 2011; 92(11): 2542-2548. DOI: https://doi.org/10.1099/vir.0.034983-  
60. Kim YandLee C. Porcine epidemic diarrhea virus induces caspase-independent apoptosis through activation of  
mitochondrial  
apoptosis-inducing  
factor.  
Virology,  
2014;  
460-461:  
180-193.  
DOI:  
61. Tanaka Y, Sato Y, Osawa S, Inoue M, Tanaka S, et al. Suppression of feline coronavirus replication in vitro by cyclosporin  
A. Veterinary Research, 2012; 43(1): 41. DOI: https://doi.org/10.1186/1297-9716-43-41  
62. von Brunn A. Editorial overview: Engineering for viral resistance. Current Opinion in Virology, 2015. 14. DOI:  
63. von Brunn A, Ciesek S, von Brunn B and Carbajo-Lozoya J. Genetic deficiency and polymorphisms of cyclophilin A  
reveal its essential role for Human Coronavirus 229E replication. Current Opinion in Virology, 2015; 14: 56-61. DOI:  
64. Tanaka Y, Sato Y and Sasaki T. Feline coronavirus replication is affected by both cyclophilin A and cyclophilin B. Journal  
of General Virology, 2017; 98(2): 190-200. DOI: https://doi.org/10.1099/jgv.0.000663  
65. de Wilde AH, Li Y, van der Meer Y, Vuagniaux G, Lysek R, et al. Cyclophilin inhibitors block arterivirus replication by  
interfering with viral RNA synthesis. Journal of virology, 2013; 87(3): 1454-1464. DOI: https://doi.org/10.1128/JVI.02078-  
12  
66. Foss DV, Hochstrasser ML and Wilson RC. Clinical applications of CRISPRbased genome editing and diagnostics.  
Transfusion, 2019; 59(4): 1389-1399. DOI: https://doi.org/10.1111/trf.15126  
67. Brokowski C and Adli M. CRISPR ethics: moral considerations for applications of a powerful tool. Journal of molecular  
68. Sugarman J. Ethics and germline gene editing. EMBO reports, 2015; 16(8): 879-880. DOI:  
69. Brokowski C. Do CRISPR germline ethics statements cut it? The CRISPR journal, 2018; 1(2): 115-125. DOI:  
Citation: Chekani-Azar S, Gharib Mombeni E, Birhan M, and Yousefi M. CRISPR/Cas9 gene editing technology and its application to the coronavirus disease