ArticlePDF AvailableLiterature Review

Abstract and Figures

On December 31, 2019, a novel coronavirus, being the third highly infective CoV and named as coronavirus disease 2019 (COVID-19) in the city of Wuhan, was announced by the World Health Organization. COVID-19 has a 2% mortality rate, is known as the third extremely infective CoV infection, and has a mortality rate less than MERS-CoV and SARS-CoV. The CoV family comprises a chief number of positive single-stranded ss (+) RNA viruses that are recognized in mammals. The 2019-nCoV patients showed that the angiotensin-converting enzyme II (ACE2) was the same for SARS-CoV. Structural proteins have an essential role in virus released and budding to various host cells. Notably, evidence indicated human-to-human transmission, along with several exported patients of virus infection worldwide. Nowadays, no licensed antivirals drugs or vaccines for being utilized against these coronavirus infections are recognized. There is an urgent requirement for an extensive research of CoV infections to disclose the route of extension, pathogenesis, and diagnosis and then to recognize the therapeutic targets to facilitate disease control and surveillance. In this article, we present an overview of the common biological criteria of CoVs and explain pathogenesis with a focus on the therapeutic approach to suggest potential goals for treating and monitoring this emerging zoonotic disease.
Content may be subject to copyright.
The Novel Insight of SARS-CoV-2 Molecular Biology
and Pathogenesis and Therapeutic Options
Arghavan Asghari,
1
Mohsen Naseri,
2
Hamidreza Safari,
3
Ehsan Saboory,
4
and Negin Parsamanesh
4
On December 31, 2019, a novel coronavirus, being the third highly infective CoV and named as coronavirus
disease 2019 (COVID-19) in the city of Wuhan, was announced by the World Health Organization. COVID-19
has a 2% mortality rate, is known as the third extremely infective CoV infection, and has a mortality rate less
than MERS-CoV and SARS-CoV. The CoV family comprises a chief number of positive single-stranded ss (+)
RNA viruses that are recognized in mammals. The 2019-nCoV patients showed that the angiotensin-converting
enzyme II (ACE2) was the same for SARS-CoV. Structural proteins have an essential role in virus released and
budding to various host cells. Notably, evidence indicated human-to-human transmission, along with several
exported patients of virus infection worldwide. Nowadays, no licensed antivirals drugs or vaccines for being
utilized against these coronavirus infections are recognized. There is an urgent requirement for an extensive
research of CoV infections to disclose the route of extension, pathogenesis, and diagnosis and then to recognize
the therapeutic targets to facilitate disease control and surveillance. In this article, we present an overview of the
common biological criteria of CoVs and explain pathogenesis with a focus on the therapeutic approach to
suggest potential goals for treating and monitoring this emerging zoonotic disease.
Keywords: coronaviruses, genome structure, pathogenesis, diagnosis, treatment
Introduction
The coronaviridae family contains a large number of
linear single-stranded positive-sense RNA viruses
(Ceraolo and Giorgi, 2020) that are found in fish, birds, and
mammals (Lea
˜oet al., 2020). Coronaviruses are genetically
categorized into four major genera: alpha coronavirus
(aCoV), beta coronavirus (bCoV), gamma coronavirus
(gCoV), and delta coronavirus (dCoV) (Li, 2016). aCoVs
and bCoVs tend to infect mammals, whereas dCoVs and
gCoVs typically infect birds. However, some dCoVs and
gCoVs can infect mammals under specific conditions. Se-
ven CoVs have been discovered in humans. Two aCoVs
(HCoV-229E and HCoV-NL63) and two bCoVs (HCoV-
OC43 and HCoV HKU1) may cause only moderate upper
respiratory disease such as common cold in immune-
competent hosts, especially a few cases of acute infection in
infants, children, and seniors (Su et al., 2016; Forni et al.,
2017) (Cui et al., 2019).
SARS-CoV-2 is a novel coronavirus with a 2% mortality
rate. It is considered the third highly infective CoV, which
has a lower mortality rate than SARS-CoV and MERS-CoV
(National Health and Health Commission and the provincial
health and health commission, including Hong Kong,
Macao, and Taiwan, 2020).
A very critical threshold associated with viral transmissi-
bility is the primary number of replication, which is usually
denoted by R0 (pronounced ‘‘R naught’’) (Organization,
2020). Transmissibility is significantly higher than SARS-
CoV-2 estimated mean R0 for SARS-CoV-2 (3.35.5) in the
early outbreak process (Zhao et al., 2020). It seems consid-
erably higher than SARS-CoV (R0: 2–5).
A recent study indicated that MERS-CoV R0 is less than
one, which means that it is impossible to trigger a pandemic
(Bauch and Oraby, 2013). The high transmission capacity of
the virus has led to 3,753,782 confirmed cases in 212 coun-
tries, areas, or territories according to the WHO statistics to
date (Thursday May 07, 2020). Proper early diagnosis and
treatment will reduce the mortality rate of this disease. Since
SARS-CoV-2 is a newly emerging disease and its diagnosis
and treatment is very controversial, a considerable number of
incorrect diagnoses caused 263,785 deaths up to now.
High transmissibility and pathogenicity of SARS-CoV-2
may be due to different genetic and protein structures such
as S protein compared with SARS-CoV and MERS-CoV
(Wang et al., 2020a). This review was conducted to study
1
Student Research Committee and
2
Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand, Iran.
3
Department of Immunology, Torbat Jam Faculty of Medical Sciences, Torbat Jam, Iran.
4
Zanjan Metabolic Diseases Research Center, Zanjan University of Medical Sciences, Zanjan, Iran.
DNA AND CELL BIOLOGY
Volume 39, Number 10, 2020
ªMary Ann Liebert, Inc.
Pp. 1–13
DOI: 10.1089/dna.2020.5703
1
Downloaded by 217.219.70.5 from www.liebertpub.com at 08/02/20. For personal use only.
the biological structure to determine appropriate diagnosis
and therapies for SARS-CoV-2.
Genomic Organization
Coronaviruse genome is a 30kb single-stranded positive-
sense RNA (+ssRNA). It has a 5¢cap head and a 3¢poly(A) tail.
The 5¢end of the gene contains a leader sequence and an
untranslated region (UTR) with several stem-loop structures
that are necessary to replicate and transcribe RNA.The 3¢UTR
also includes RNA structures that are essential for viral RNA
replication and synthesis. Coronavirus genome is organized by
5¢-leader-UTR-replication-ORF1a/b-Spike-3a-3b-envelope
protein-membrane-6p-7a-7b-Nucleocapsid-3¢UTR-poly(A)
tail with additional genes interspersed in structural genes at the
3¢end of the genome (Fig. 1) (Ceraolo and Giorgi, 2020).
The SARS-CoV-2 pathogen belongs to the coronavirus
family, bCoV genus, and Sarbecovirus subgenus. Although the
origins of the virus and its intermediate host are still contro-
versial, molecular studies have demonstrated that RaTG13, the
bat coronavirus, has 96.2% nucleotide homology with the
human coronavirus (Fehr and Perlman, 2015). Mutations in
various SARS-CoV-2 primarily occur in five genes, including
S, N, ORF8, ORF3a, and ORF1ab, with around 42% of the
variants being nonsynonymous (Kahn and McIntosh, 2005).
An increased degree of viral diversity has been observed
among patients diagnosed with SARS-CoV-2, indicating that
the virus has begun adapting to the human environment and its
genomes have started developing in populations (Li, 2016).
Virion Structure
Coronaviruses are spherical with diameters of *125 nm
(Ba
´rcena et al., 2009). This virus involves four main
structural proteins, including spike (S), membrane (M),
nucleocapsid (N), and envelope (E) proteins, which are
encoded inside the 3¢end of the viral genome.
S protein
This protein is located on the virus surface and contributes
to forming a corona, and this is the reason behind the cor-
onavirus getting its name. S protein is 150 kDa with 1300
amino acids that act as homotrimers protein and N-terminal
(NT) signal sequences for 20 asparagine-linked glycan in the
endoplasmic reticulum (ER) (Fehr and Perlman, 2015; Song
et al., 2018). S protein is anchored on virus envelope functions
in attaching coronavirus receptors and internalization. This
protein in coronaviruses consists of three major domains,
including the extracellular domain, which consists of the re-
ceptor binding (S1) and membrane-fusion subunit (S2). S1 is
divided into the N-terminal domain (S1-NTD) and receptor
binding domain (RDM) (Heald-Sargent and Gallagher,
2012). Certain structures are coiled coils consisting of three a-
helices labeled heptad repeat (HR1), besides three chains
known as HR2. The complexity between the HR1 and HR2
leads to providing a structure that is resistant to cellular pro-
teases during post-fusion (Yan et al., 2020).
S proteins in all coronaviruses are cleaved by host
proteases such as TMPRSS2 and lysosomal proteases ca-
thepsins during host–virus fusion (Shang et al., 2020).
SARS-CoV-2 S protein contains 33 specific amino acids
(2.59%), with larger variations being 439–449 and 482–505,
respectively (Li et al., 2020). Moreover, various investiga-
tions found that this virus has a unique peptide (PRRA)
insertion (Wang et al., 2020a) that affects cleaving. As a
consequence, S protein indicated a specific furin cleavage
(-PRAR-) inside the S1/S2 domain that overlaps with the
mentioned insertion (Wang et al., 2020a). Due to the key
FIG. 1. Coronavirus schematic diagram. Structural proteins are encoded by the four structural genes, including spike,
envelope, membrane, and nucleocapsid genes, and also genome organization and the encoded proteins of pp1ab and pp1a
and accessory proteins (3a, 3b, 6, 7a, 7b,8a, 8b, 9b, and ORFs). ORFs, open reading frames.
2 ASGHARI ET AL.
Downloaded by 217.219.70.5 from www.liebertpub.com at 08/02/20. For personal use only.
role of this protease in the entry of viruses into the host cell,
the inhibitory agents such as MI-1851 and camostat mesy-
late of this protease can be used to treat the SARS-CoV-2.
Another way to treat patients infected with SARS-CoV-2
is to inhibit protein S activity, which can block the virus
(Bestle et al., 2020; Hoffmann et al., 2020b).
Griffithsin is one of the drugs used to treat infected pa-
tients. It is a lectin drug that works by controlling S protein
by binding to glycoproteins (Lee, 2019). Antibody-based
therapies usually target S protein. Among them, we can
mention CR3022, which has been used to treat SARS dis-
ease. This protein acts by binding to the Receptor binding
domain site. Due to the high hematology of this site of
protein in SARS and SARS-CoV-2, it can be mentioned as a
treatment option ( Jiang et al., 2020; Tian et al., 2020).
M protein
Recent studies have shown that M protein in SARS-CoV-2
can be compared with MERS and SARS: It has a structural
identity of 39.2% and 90.1%, respectively (Ahmed et al.,
2020) M protein, a transmembrane glycoprotein type III, is an
important structural protein in coronaviruses (Arndt et al.,
2010). It is the most abundant protein on the surface of the
virus, which gives it shape. According to studies conducted
on the two viruses SARS-CoV and MERS-CoV, M protein
contains *230 amino acids in length, a 25–35 kDa, which is
the least in all structural proteins.
In silico analysis showed that M protein in the SARS-CoV-2
has a structure similar to prokaryotic sugar transport protein
and has a triple helix bundle, a single 3-transmembrane domain
(TMD).Basedonthis,itcanbeassumedthatthisproteincan
play a role in the entry of virus into the host cell and the
maturation of RNA viruses, but many studies must be done
to confirm this hypothesis (Thomas, 2020) .NT and
C-terminal domains (CTDs) are ecto domain and endo
domain, respectively. CTD has the ability to bind to RNA.
This protein is translated by ribosomes, which are attached
to the rough ER (co-translational) (Arndt et al., 2010). In
almost all coronaviridae representatives, an amphipathic
region at the end of the third TMD is highly preserved
(Arndt et al., 2010).
N-linked glycosylation in a,dand O-linked glycosylation
in b(Oostra et al., 2006) are found in this glycoprotein, which
leads to an antigenic role (Braakman and Van Anken, 2000).
N protein
This protein is 43–50 kD and is presented in coronaviruses.
This helical nucleocapsid structural protein can bind to RNA
with several lysine and arginine amino acids (Chang et al.,
2006). This protein contains NTD (RNA-binding site), CTD
(dimerization domain) and is intrinsically central disordered
(serine- and arginine-rich for phosphorylation) (Kumar et al.,
2020). N protein functions in attaching and assembling the
viral RNA genome to a long helical nucleocapsid structure or
matrix of the ribonucleoprotein (Kumar et al., 2020); how-
ever, it is remarkably sensitive to proteases (Macnaughton
et al., 1978). This protein is phosphorylated in multiple spe-
cific positions in various coronaviruses, which may alter
functions such as distinguishing nonviral and viral RNA
impaired binding of the monoclonal antibody to the virus
surface as well as virus maturation and assembling (Kuo et al.,
2016; Grunewald et al., 2018; Chen et al., 2019). However,
the reason behind phosphorylation has remained uncertain.
N protein can also be involved in transcription regulation,
viral transcription and increase the performance of replicative
or genomic RNA replication in reverse genetic systems
(Hu et al., 2017; Cong et al., 2020).
Accumulating evidence has indicated that N protein that
often exists in the cytoplasm of contaminated cells leads to
an arrest in the cell cycle in the G2/M phase (Wurm et al.,
2001). Further, protein crystallography has reported that N
protein structure is extremely varied in RNA binding sites of
severe infectious coronaviruses (SARS-CoV-2, SARS-CoV,
and MERS-CoV) and mild infectious viruses (HCoV-229E,
HCoV-NL63, HCoV-HKU1, and HCoV-OC43). SARS-
CoV-2 utilized a particular pattern to bind RNA with data
on atomic resolution (Kang et al., 2020). Since N protein is
a surface protein and has less variation than S protein, it can
be suggested as a vaccine candidate for SARS-CoV-2
(Ahmed et al., 2020; Fu et al., 2020). In silico analysis
suggests that the effect of the two drugs, Glycyrrhizic acid
and the phytochemical Theaflavi, on N proteins can be ex-
amined as one of the treatment options (Ray et al., 2020).
E protein
E protein is known as a small hydrophobic protein that is
*74–109 amino acids and 8.4–109 kDa. This protein consists
of three parts: NT (negatively charged), TMD (not recharged),
and CT (negatively charged) (Nieto-Torres et al., 2011). There
are three specific sites for cysteine that undergo palmitolate
changes (Liao et al., 2006; Tseng et al., 2014). Although the
accurate function of palmitolytic protein has been still con-
troversial, various roles for this protein are considered for
coronavirus. Changes in all three amino acids in MHV-CoV
can significantly weaken this virus. Several studies have pub-
lished that this modification may contribute to SARS-CoV Ag
shedding (Lopez et al., 2008; Tseng et al.,2014).Another
conserved residue in its structure is a proline that is protected in
the CT region of the b-coil-bmotif. It plays an important role in
the maturation of the protein in Golgi (Tseng et al.,2014).
E protein contributes to ion channel, viroporin activity,
and virus assembling to release the curvature of the cell
membrane in budding virus (Schoeman and Fielding, 2019).
PDZ-binding motif (PBM) is an important protein motif in
the CT region, and it plays a critical role in pathogenicity by
interfering in cell signaling. PBM is considered as a source of
the pathology of SARS-CoV (Hung and Sheng, 2002; Javier
and Rice, 2011). Post-translational changes with critical roles,
including glycosylation, palmitoylation, myristoylation, and
ubiquitination, occur in proteins (Schoeman and Fielding,
2019). Several studies show that E protein has a role in
pathway signaling, for example, one study of E protein
showed that the E protein in SARS-CoV-2 in the CT region
had changed in several amino acids, which could affect the
PALS1, which plays a key role in tight junction, causing the
virus to become more pathogenic than other coronaviruses
(De Maio et al.,2020).
Several pangenomic studies investigating beta cor-
onavirus sequences have revealed that two functional
characteristics, including an ion channel and a PBM, are
strictly conserved in all core gene clusters in SARS and
SARS-CoV-2 variants. These characteristics stimulate a
THE NOVEL INSIGHT OF SARS-COV-2 MOLECULAR BIOLOGY 3
Downloaded by 217.219.70.5 from www.liebertpub.com at 08/02/20. For personal use only.
cytokine storm, trigger the inflammasome, and, subse-
quently, increase edema in lungs. This process causes acute
respiratory distress syndrome and ultimately death in SARS-
CoV1 and SARS-CoV-2 infections. Most medications im-
pair this mechanism, such as Amantadine and Hexamethy-
leneamiloride (acting on ion channels) and SB203580
(effect on PBM).
E protein subcluster in SARS clade is quasi-identical for
the main functional regions of SARS-CoV1 and SARS-
CoV-2. Therefore, SARS E protein inhibitors are suggested
as an appropriate candidate for SARS-CoV-2 treatment, as
they are reported in animal models (Alam et al., 2020).
Nonstructural proteins
Sixteen nonstructural proteins (nsps) contribute to tran-
scription replication, which are originally provided by the
auto proteolytic of two primary proteins: pp1a and pp1ab.
Breaking down of pp1a protein leads to the production of
nsps 1–16, and the breaking down of pp1ab provides nsps
1–10 and nsps 12–16. This process is similar in all proteins,
except for those that are not produced by ribosomal fra-
meshifting. Pp1a autoproteolytic products play specific
roles, such as cell infection and assembling the RNA syn-
thesis system. Moreover, pp1ab products function in RNA
replication and transcription (Masters, 2006).
nsp1, which is located in the NT region of the pp1a
protein, contains 180 amino acids. The similarity of nsp1 in
SARS-CoV-2 is 82–84%, similar to that of SARS-CoV (Ren
et al., 2020). The difference is in the single amino acid,
which can naturally cause structural changes in the protein
(Wu et al., 2020). These changes are usually caused by
merging with the host sequence. Due to the epidemic of this
disease in human populations, point mutations increase,
which can lead to structural changes in vital proteins in the
virus.
nsp1 plays various roles, including suppression of host
gene expression through RNA degradation and 40s ribo-
somal disruption (Kamitani et al., 2006, 2009). Interest-
ingly, this protein can recognize emRNA modifications but
not a specific sequence of virus RNAs (Huang et al., 2011).
nsp1 regulates gene expression in the interferon type 1
signaling cascade by decreasing the amount of STAT1
protein phosphorylation and also through slight effects on
proteins such as STAT2, JAK1, and TYK2 (Wathelet et al.,
2007). nsp1 can be considered a virulence factor and con-
tributes to pathogenicity through favored replication of
SARS-CoV. Higher production of nsp1 influences the signal
pathway of calcineurin/NFAT, is eventually correlated with
the development of interleukin 2 (IL2), and may influence
its viral pathogenicity (Pfefferle et al., 2011). Cyclophilin
interfaces with nsp1 to alter the calcineurin signaling. The
effect of this drug on many coronaviruses has been inves-
tigated (de Wilde et al., 2013; Tanaka et al., 2013).
The degrade precursor nsp2–3 results in two nsp2 and
nsp3, which are 65 kDa and 22–240 kDa, respectively. Stu-
dies on protein nsp1 have failed to find the specific role of
this protein (Graham et al., 2005; Gadlage et al., 2008).
There are many mutations in nsp2 that have caused changes
in this protein (Menachery et al., 2017). Studies have shown
that this protein contains 61 amino acids varying from
SARS-CoV-2 and SARS-CoV (Wu et al., 2020). nsp2 was
observed as interacting with nsp8, which is involved in
replication machinery (von Brunn et al., 2007). Cor-
onaviruses create double-membrane vesicles (DMVs) in
infected cells that are filled with replication-transcription
complexes (RTCs). nsp2 is essential for the structure of
DMV-anchored RTCs (Hagemeijer et al., 2010). By inter-
acting with prohibitin 1 and prohibitin 2 cellular proteins,
nsp2 can interfere with host cell signaling, including cell
cycle death cell pathways and cell differentiation (Cornillez-
Ty et al., 2009).
One of the diagnostic methods is to suggest the identifi-
cation of coronavirus disease 2019 (COVID-19), which is
real-time reverse transcriptase PCR (RT-PCR) assay,
COVID-19-nsp2 real time, which is a more sensitive and
faster method than other conventional methods (Yip et al.,
2020).
nsp3 is a significant protein in coronaviruses and has eight
domains, including ubiquitin-like domain 1 (Ubl1), Glu-rich
acidic domain (also called ‘‘hypervariable region’’), macro-
domain (also named ‘‘X domain’’), ubiquitin-like domain 2
(Ubl2), papain-likeprotease2 (PL2
pro
), nsp3 ecto domain
(3Ecto, also called ‘‘zinc-finger domain’’), Y1 domain (with
unknown function as well as CoV-Y), and two conservation
regions, which are transmembrane (TM1 and TM2) (Lei
et al., 2018). This protein breaks down pp1a and pp1ab
through its protease activity in nsp3 (PL2
pro
) (Lei et al.,
2018).
nsp3 can disrupt the host cell cycles by affecting p53 and
calcium/calmodulin-dependent protein kinase II (Ma-Lauer
et al., 2016). This protein also plays a critical role in RTC
formation as well as in nsp2 (Angelini et al., 2013). Gen-
erally, the nsp3 multi-domain functions in infection with
coronavirus. nsp1, nsp2, and nsp3 are produced from the
polyproteins via nsp3 and cause replication/transcription
complex formation with other viral nsps as well as RNA. nsp3
performs post-translational structural changes on host protein
to suppress the host innate immunoresponse. nsp3 is affected
by 3Ecto N-glycosylation in infected cells (Lei et al., 2018).
This protein is also known as the principal target of lineage C
bCoVs development, based on a high frequency of positively
selected mutation sites (Forni et al., 2016).
Mounting evidence has shown that several structural
changes occurred in nsp2 and nsp3 during positive selection.
There are multiple varieties in the nsp2 endosome-associated
protein-like domain, which may justify the extreme conta-
gion characteristics in SARS-CoV-2. Another mutation near
the nsp3 phosphatase domain may cause the difference be-
tween SARS-CoV-2 and SARS-CoV (Angeletti et al., 2020).
SARS-CoV-2 sequencing has proposed that the papain-like
protease/deubiquitin domain, a unique protein in nsp3, can
be a promising viral inhibitor in medication (Shanker et al.,
2020). Two of the drugs that are prescribed for treatment are
lopinavir and ritonavir. These drugs inhibit the activity of the
virus by inhibiting protease activity. There is no agreement
in the articles on the use of these drugs.
Several strands of research find that these medications can
be used to cure patients (Lim et al., 2020; Xu et al., 2020).
Conversely, a new study consisting of a total of 199 patients
with laboratory-confirmed SARS-CoV-2 infection found
that lopinavir–ritonavir therapy outside routine care had no
positive effect in terms of clinical outcomes and deaths (Cao
et al., 2020). Disulfiram (inhibitor nsp3) and darunavir
4 ASGHARI ET AL.
Downloaded by 217.219.70.5 from www.liebertpub.com at 08/02/20. For personal use only.
(conformation changes PL2pro) are one of the drugs that
cause changes in the function of this protease and that can
be considered as treatment options (Zhou et al., 2015;
Hoffmann et al., 2020a; Lin et al., 2020).
nsp4 plays a role in the development, arrangement, and
function of these complexes for viral replication (Perlman
and Netland, 2009). Amino acid sequencing in MHV nsp4
projected that this protein has four TMDs (TM1–4). MHV
nsp4 can be a critical target to overcome contagious viruses
(Sparks et al., 2007). nsp4 is necessary for the structure of
RTCs anchored with DMV as well as with nsp2 (Gadlage
et al., 2010).
nsp5, which is often known as 3C-like protease, or the
main protease, plays a significant role in the synthesis of
viral proteins and generates several nonstructural viral
proteins through its protease activity (Lai and Cavanagh,
1997; Ziebuhr et al., 2000; Masters, 2006; Perlman and
Netland, 2009). Moreover, nsp5 can inhibit interferon I
signaling processes by intervening in the NF-kB process and
breaking down STAT 1 transcription factor (Zhu et al.,
2017a, 2017b). 3C-like protease plays a crucial role in the
coronaviruses life cycle and makes it a desirable target to
produce antiviral drugs (Macchiagodena et al., 2020). Pyr-
anones 3f, 3g, and 3m can impair neuraminidases prote-
ase and 3C-like protease and can be used as proper drugs to
treat SARS-CoV-2 (Kumar et al., 2016). Other drugs that
inhibit this protease include Paritaprevir and Raltegravir
Lasinavir, Brecanavir, Telinavir, Rotigaptide, 1,3-Bis-(2-
ethoxycarbonylchromon-5-yloxy)-2-(lysyloxy) propane, and
Pimelautide, which have been suggested for treatment
(Durdagi et al., 2020).
nsp6 has a membrane proliferation potential to cause
perinuclear vesicles situated around the microtubule orga-
nizing center. This protein, besides nsp3 and nsp4, can close
the DMVs as well as infected cells (Angelini et al., 2013).
nsp6 can induce cell autophagosis by affecting two vital
proteins of the autophagy signaling pathway, including
ATG5 and PIK3C3. nsp6 restricts autophagosomal expan-
sion, either directly by nsp6 or indirectly by deprivation or
MTOR signaling chemical suppression. Inhibition happens
at the omegasome formation level and, subsequently, pro-
hibits broad autolysosome development (Benvenuto et al.,
2020; Yang and Shen, 2020). One way to treat COVID-19 is
by its effect on the cell autophagosis signaling pathway
(Yang and Shen, 2020). For example, chloroquine is con-
sidered a treatment option by affecting the pH of lysosomes
(Degtyarev et al., 2008).
nsp8 and nsp7 are 22 and 10 kDa, respectively, and they
are considered co-factor components. These proteins create
a heterodimer and function in stabilizing the RNA binding
site in nsp12. The NT residue from nsp8 is crucial in the
capacity of the protein to interact with nsp7 (Zhai et al.,
2005; Kirchdoerfer and Ward, 2019). nsp8 has RNA-
dependent RNA polymerase (RdRp) replicase subunits that
are special for CoVs and can conduct de novo RNA
synthesis only with low fidelity on ssRNA templates
(Konkolova et al., 2020). These findings characterize the
complexofSARS-CoVnsp7andnsp8asaninteresting
multimeric RNA polymerase that can extend the primers
(Snijder et al., 2016; Konkolova et al., 2020).
nsp9 is an essential protein for linking coronavirus rep-
lication to RNA. Various ways of nsp9 dimerization im-
prove their binding affinity to nucleic acid (Zeng et al.,
2018). However, nsp9 requires the presence of nsp8 for
binding to RNA (Sutton et al., 2004).
nsp10 is also recognized as a significant replication reg-
ulator. nsp10 has148 amino acids containing two zinc finger
domains for enzymatic interaction. It can interact with
nsp14 and nsp16 (Ma et al., 2015; Rosas-Lemus et al.,
2020). nsp16 requires an interaction with nsp10 to perform
powerful methyltransferase, which can transform cap-0
(7MeGpppN) specifically into the cap-1 structure (Rosas-
Lemus et al., 2020). The interaction of nsp10 with nsp14
induces exonuclease function whereas it does not affect the
function of nsp14 methyl transferase (Ma et al., 2015).
nsp11 contains two main domains, including CTD and
NTD. It exhibits endo-ribonuclease activity (Li et al., 2014)
and plays a substantial role in the viral life cycle. This
protein can break down dsRNA and ssRNA by identifying
specific sites (3¢uridylate) (Nedialkova et al., 2009).
nsp11 has an inhibitory impact on TNF-adevelopment
and IL1 signaling (He et al., 2015). RNA microarray results
have revealed that nsp11 correlates with many host cell
pathways such as cell cycle and DNA replication, histon
modification, protein kinase signaling, and proteasome
processes (Sun et al., 2014a).
nsp12 is known for its RdRp activity. This protein has an
extended NT region, which binds to two cofactors (nsp7 and
nsp8) that are necessary for nsp12polymerase function. The
NT of this protein has nucleotidyltransferase activity (Leh-
mann et al., 2015), and an RdRp domain is located at the 12
CT (te Velthuis et al., 2009). Although binding nsps can
improve the efficiency of replication and transcription, at
least the complex that can make the protein nucleation
portion is nsp12, nsp8, and nsp7 (Sevajol et al., 2014;
Lehmann et al., 2015). nsp12 has two polymerase activities:
primer-dependent and primer-independent RNA synthesis
activities using homopolymeric RNA templates (Ahn et al.,
2012).
Emerging evidence has reported that vitamin B12 (me-
thylcobalamin) may bind to the active site of nsp12 and
inhibit it. Therefore, this vitamin can be used as an antiviral
drug in SARS-CoV-2 (Narayanan and Nair, 2020). Another
study has suggested that the Pan-Janus Kinase inhibitor
might be a treatment for this disease by disabling RdRp
protein activity (Mirza and Froeyen, 2020).
A significant group of drugs used for viral diseases are
nucleotide analogues (Remdesivir), which function by af-
fecting nsp12 (Wang et al., 2020b). Hence, this protein can
be regarded as one of the antiviral medications ( Ju et al.,
2020). Nucleoside analogues are usually derivatives of
adenosine and guanosine, which inhibit RdRp. This group of
drugs is commonly used in viral infections (De Clercq,
2019). Favipiravir, from the family of Nucleoside ana-
logues, inhibits the virus by targeting the catalytic domain of
nsp12 (Furuta et al., 2017). A study of patients with SARS-
CoV-2 showed that reducing the time to clear the virus and a
CT of the lungs of patients taking the drug were better than
those of those who did not take the drug (Cai et al., 2020).
Remedisir is another member of the nucleoside analog
family that is currently being recommended for the treat-
ment of infected individuals. It has previously been used to
treat MERS and SARS (Al-Tawfiq et al., 2020; Singh and
Sharma, 2020). It works by its effects on NSP12 and
THE NOVEL INSIGHT OF SARS-COV-2 MOLECULAR BIOLOGY 5
Downloaded by 217.219.70.5 from www.liebertpub.com at 08/02/20. For personal use only.
Replicase polyprotein 1ab (Agostini et al., 2018; Elfiky,
2020). This drug reduces the symptoms of the disease in
people infected with the SARS-CoV-2, such as: temperature
normalization, respiratory rate, oxygen saturation, and
cough alleviation (Al-Tawfiq et al., 2020).
nsp13 is characterized by helicase activity and belongs
tosuperfamily1(SF1)(Begumet al., 2020). This protein
can unwind both double-stranded DNA and RNA by hy-
drolyzing deoxyribonucleotide triphosphates (dNTPs) and
ribonucleotide triphosphates. nsp12 can enhance the un-
winding activity of nsp13 (Jia et al., 2019). nsp13 has
several domains, including NT Cys/His-rich domain (CH)
with three zinc atoms, b-barrel domain and CT SF1 heli-
case center with two RecA-like subdomains. This protein is
one of the most conserved ancestral proteins in nido-
viruses, rendering it an essential drug discovery (Hao et al.,
2017).
Mounting evidence has revealed that nsp13 can hydrolyze
all forms of NTPs and unwind NTP-dependent RNA helices
by its NTPase and RNA helicase activity. Some divalent
actions, such as Ca2
+
, Zn2
+
, and Mg2
+
, impair nsp13 ac-
tivity. A research report has shown that bismuth salts such
as potassium citrate (BPC), ranitidine bismuth citrate
(RBC), and bismuth citrate would inhibit NTPase and RNA
helix-unwinding behaviors of SARS-CoV-2 nsp13. Among
them, BPC or RBC can significantly impair SARS-CoV-2
nsp13-dependent NTPase and RNA helicase activities (Shu
et al., 2020).
Mounting evidence has revealed that nsp13 can hydrolyze
all forms of NTPs and unwind NTP-dependent RNA helices
by its NTPase and RNA helicase activity. Certain bismuth
salts can effectively inhibit nsp13 NTPase and RNA heli-
case activity (Shu et al., 2020).
nsp14 is known as an exonuclease; it functions in a 3¢-5¢
direction on ssRNAs and dsRNAs that depend on conserved
residues in the DEDD exonuclease super family ( Minskaia
et al., 2006). Further, it plays a role in RNA viral modifi-
cation (N7-methylguanosine [m7G]) (Chen et al., 2009).
The N7-MTase activity of nsp14 is related to protein
carboxy-terminal (Chen et al., 2009). Three natural and
microbial extracts of PF35468, PA48202, and PA48523
products may inhibit SARS-CoVnsp14 (Sun et al., 2014b).
One of the opposite approaches to COVID-19 can be to
inhibit this protein, which is one of the treatment options
based on the adenine dinucleoside SAM analogues (Ahmed-
Belkacem et al., 2020).
nsp15 is made of 345 amino acids and has three domains:
NT, central, and CT (catalytic NendoU) domain. This is a
hexameric endoribonucleases protein, and it is called En-
doU. Endoribonuclease breaks down the double-stranded
and single-stranded RNA by identifying uridine sites and
cleaves uridines 3¢(Bhardwaj et al., 2008).
nsp15 can detect multiple sensors such as MDA5, PKR,
and OAS/RNaseL and it can delay interferon signaling
(Deng et al., 2017; Kindler et al., 2017). nsp15 in COVID-
19 is a conserved protein that has 88% and 95% sequence
homology with other coronaviruses and SARS-CoV, re-
spectively. It means that certain variations can be related to
SARS-CoV-2 virulence (Kim et al., 2020). Recent research
on inhaled corticosteroid ciclesonide revealed that nsp15
inhibition might be an appropriate candidate for treating
SARS-CoV-2 (Matsuyama et al., 2020).
nsp16 is an S-adenosylmethionine-dependent operation
(nucleoside-2¢-O)-methyltransferase and generates nsp16/
nsp10 complex (Lugari et al., 2010). This protein, besides
the interfering interferon band ISRE signaling pathway, can
downregulate RIG1 and MDA5 proteins. nsp10 can also
accelerate this process (Shi et al., 2019); nsp10-derived
peptides K12 and K29 block nsp16 dose-dependent action in
SARS-CoV and suppress full MTase action of nsp10/nsp16
(Lugari et al., 2010). Ribavirin, known as an antiviral drug,
inhibits the nsp16. Ribavirincan can be blocked by 2¢-O-
methyltransferase activity of nsp16, which effectively in-
hibits the 5¢messenger RNA (mRNA) cappig (Te et al.,
2007; Elfiky, 2020) and contributes to a decrease in viral
gene expression by targeting the nucleotide binding site
(Tam et al., 2001). It also affects the immune system and
activates the antiviral response in the body (Tam, 2002)
Dolutegravir and Bictegravir are two drugs that are re-
commended to inhibit this enzyme in patients with SARS-
CoV-2 (Beck et al., 2020).
Replication and Pathogenesis
Attachment and entry
At this stage, S protein, which contains S1 and S2, plays a
critical role. S1 includes CTD (receptor binding and pro-
moting membrane fusion and has RBD) and NTD (sugar
receptors recognized).
S protein has two forms: the pre-fusion, which is observed
in mature virus and has a hemotrymeric arrangement; the
post-fusion, which is completed after membrane fusion
(Li et al., 2006). Coiled-coil structure is observed only in S2
and not S1 (Li et al., 2006; Walls et al., 2017). SARS-CoV-2
recognizes angiotensin-converting enzyme II (ACE2) recep-
tor protein by using the RBD motif of the S protein and binds
to the host cell (Minskaia et al., 2006; Chen et al., 2009; Sun
et al., 2014a). The binding of ACE2 to SARS-CoV-2 has
15 nM affinity, which is *10–20-fold higher than SARS-
CoV unexpectedly. This can be an important reason that
SARS-CoV-2 is exceedingly contagious (Wrapp et al., 2020).
ACE2 plays a significant role in controlling blood pressure
and it is found in tissues such as heart, liver, kidneys, etc.
The specialty of the host receptors triggers host tropism
and host selection (Bhardwaj et al., 2008). This is cleaved in
two places after attaching Sprotein to ACE2 via host pro-
teases, including the S1/S2 boundary by furin and the S2¢
site by extracellular proteases such as trypsin or TMPRSS2.
Furin is expressed in brain, lung, gastrointestinal tract, liver,
pancreas, and reproductive tissues. It can cut a particular
pattern (-PRRA-) at the border of S1 and S2 proteins that
activate the S protein for membrane fusion. It can be another
reason for high virulence and organ infections of COVID-19
(Wang et al., 2020a).
The unstable pre-fusion form is altered to the post-fusion
form via proteases and binds to viral receptors. Then, S2
helps the virus entering the host cell by creating a six-helix
bundle (Lee et al., 2016).
After binding the S protein to the ACE2 receptor, the
signaling pathway occurs through the phosphorylation of the
receptor by CK2. It can activate AP1 and ERK1/2 and
eventually leads to CCL2 expression, which refers to pul-
monary fibrosis (Fig. 2) (Heurich et al., 2014).
6 ASGHARI ET AL.
Downloaded by 217.219.70.5 from www.liebertpub.com at 08/02/20. For personal use only.
Uncoating
Virus membrane fused with host membrane resulted
in SARS-CoV-2 entering the host cell as enodocytosis
through clathrin-dependent or clathrin-independent ways
(We˛ drowska et al., 2020; Algarroba et al., 2020). Then, the
viral genome is released into the cytosolic space via lyso-
somal enzymes that are referred to as catapsin L and trypsin
(Hoffmann et al., 2020a).
Translation and the unfolded protein response
in coronavirus-infected cells
After releasing the viral genome into the host cell cyto-
solic space, the virus alters the transcription process in its
favor by disrupting the transcription process of the host cell.
Synthesized viral proteins are utilized instead of host cell
proteins, which contribute toward cellular pathogenesis
(Fung et al., 2016). Transmission of the virus genome oc-
curs after entering the 5¢region of the oRf1a and oRF1b and
products pp1a and pp1ab (Perlman and Netland, 2009).
Polyproteins generate nsps by affecting nsp3 (papain pro-
tease), nsp5 (3C protease), and cellular protease (Lai and
Cavanagh, 1997; Ziebuhr et al., 2000; Masters, 2006;
Perlman and Netland, 2009; Lei et al., 2018). The replica-
tion and transcription complex, which is produced in DMVs,
involves various proteins such as RdRp (nsp12), helicase
(nsp13), RNA cap-modifying methyltransferases (nsp14 and
nsp16), and an exoribonuclease (nsp14) (Perlman and Net-
land, 2009; Hao et al., 2017).
The RTC synthesizes a cluster, including subgenomic
RNAs in a discontinuous transcription manner. These sub-
genomic mRNAs have specific sequences of 5¢-leaders and
3¢-terminals. Several RNA processing enzymes, such as the
nsp14 exoribonuclease 3¢-5¢, are special among all RNA
viruses to CoVs and are likely to provide the RTC proof-
reading feature. The RTC instead uses the genome to syn-
thesize progeny genomes and a series of subgenomic
mRNAs, using negative-stranded intermediates. Structural
proteins such as M, S and accessory protein are translated by
ribosomes attached to the ER membrane and then, they
transit to the ER-Golgi intermediate compartment (ERGIC).
The N protein covers the progeny genomes by encapsidation
of these components along with the membrane-bound
components, creating virions by budding in the ERGIC.
Eventually, the vesicles, which include the viral particles,
combine with the plasma membrane to release the virus.
Diagnostic approach. Current screening and character-
istic ways help in epidemiologic monitoring, along with
successful control (Ozma et al., 2020). We have evaluated
the various procedures that are currently being prepared for
coronavirus detection.
CT imaging examination. The chest computerized to-
mography (CT) scan is one of the useful radiology tech-
niques or ways to observe pulmonary imaging variations in
suspected cases (Pan et al., 2020; Shi et al., 2020). The CT
scan is a complementary method to molecular-based meth-
ods and is a less costly and more successful treatment or
follow-up for a patient (Shi et al., 2020). Radiologists of the
patients with confirmed SARS-CoV-2 can obtain general
information regarding the infectious steps (Shi et al., 2020).
The CT finding taken from the SARS-CoV-2 patients con-
firmed various abnormalities in the lungs, such as consoli-
dation, centrilobular nodules, bronchial wall thickening,
vascular enlargement, crazy paving pattern, architectural
distortion, traction bronchiectasis, subpleural bands, and
reticulation, that cause pulmonary disease and need fast
FIG. 2. Schematic representation of SARS-CoV-2 and ACE2 receptor interaction. Interaction of spike protein on the
surface of the coronavirus and the cellular ACE2 receptors is needed for entrance into the target cell. ACE2, angiotensin-
converting enzyme II.
THE NOVEL INSIGHT OF SARS-COV-2 MOLECULAR BIOLOGY 7
Downloaded by 217.219.70.5 from www.liebertpub.com at 08/02/20. For personal use only.
diagnosis and viral therapy (Chung et al., 2020). However,
chest CT is a fast and easy way of diagnosis, beginning with
SARS-CoV-2 infection with high sensitivity for suitable
detection. Specific attention should be paid to the role of
radiologists in detecting this recent infectious disease (Shi
et al., 2020).
Molecular technique
PCR-based technique. PCR is the most common and
accurate technique that is used to detect pathogenic viruses
in blood and respiratory secretions (Uhlenhaut et al., 2012;
Setianingsih et al., 2019). The PCR-based technique has a
wide range of applications with high specificity and sensi-
tivity (Wan et al., 2016). This approach has become a
standard and effective method for the detection of cor-
onavirus. Coronavirus RNA is usually extracted, and then
PCR is done by a particular detection technique or instru-
ment (Lu et al., 2014).
RT-PCR is usually recommended for coronavirus detec-
tion because of its advantages as a precise and easy quan-
titative test (Wan et al., 2016). Also, real-time RT-PCR is
more specific and sensitive than traditional RT-PCR assays,
which aid in primary infection diagnosis (Corman et al.,
2012). For the SARS-CoV-2 molecular analysis, various
specimens such as oropharyngeal swabs, throat swabs, and
rectal swabs are examined by using RT-PCR as golden
clinical diagnosis method for virus detection according to
classical Koch‘s postulates (Corman et al., 2020).
Loop-mediated isothermal amplification-based tech-
nique. The Loop-mediated isothermal amplification
(LAMP) is a new isothermal nucleic acid amplification
technique with high efficiency. This is used to amplify
RNAs and DNAs with high specificity and sensitivity due to
its exponential amplification feature and six particular target
sequences diagnosed by four separate primers (Notomi
et al., 2000). The LAMP assay is rapid and does not need
high-priced reagents or equipment. In addition, the LAMP
test will help to decrease the cost of coronavirus detection.
Several strategies for the detection of coronavirus based on
LAMP will be defined here, which have been developed and
performed in clinical diagnosis. The gel electrophoresis
method is widely utilized for its study of the amplified items
to detect endpoints (Enosawa et al., 2003). Poon and co-
workers indicated a simple LAMP test for the SARS study
and showed the feasibility of the usage of these methods for
the SARS-CoV finding. The aim of the study was for the
amplified items to detect endpoints (Enosawa et al., 2003).
Microarray-based technique. The microarray-based
method is a quick and high-throughput detection device. For
this method, the SARS-CoV-2 RNA will first produce
cDNA by reverse transcription with different probes and
these are then hybridized with solid-phase oligonucleotides
into each well (Chen et al., 2010). Afterward, free DNAs are
separated by washing the solution. Because of this advan-
tage, the microarray test was usually used for coronavirus
detection (Li et al., 2014). According to the sequence of
TOR2, Shi and coworkers used a 60mer oligonucleotide
microarray and effectively utilized it for the SARS detection
of coronavirus in patient samples (Chen et al., 2010).
CRISPR technique. The evidence showed that RNA-
targeting detection with CRISPR-Cas13 for fast detection
and portable sensing of nucleic acids can be effective in
virus epidemiology, diagnosis, and prevention (Wright
et al., 2016; Gootenberg et al., 2017). Zhang et al. reported
a protocol for SARS-CoV-2 detection by CRISPR, that
Cas13/C2c2 can be programmed to target and destroy the
genomes of live cells and different diseases (Freije et al.,
2019). The test is carried out by isothermal amplification of
the nucleic acid extraction of patient clinical samples and
then viral RNA sequence amplified through Cas13/C2c2
and, consequently, readout in less than an hour by paper
dipstick (Freije et al., 2019). This method provides quick,
manageable, multiplexable, and quantitative detection plat-
forms of viral nucleic acids (Gootenberg et al., 2018).
In conclusion, this novel virus outbreak emerged and
spread immediately and has posed a challenge to medicine,
economy, and public health worldwide. Several strands of
evidence of this virus are proposed from the unknown in-
termediate host to cross-species, and human-to-human
transmission is established and is an alarm. Even so, assessing
the viral nature of COVID-19 and considering a significant
impairment of the human immune response in serious forms,
it is essential to balance the risk and benefit ratio before
beginning anti-inflammatory treatment. We also speculate on
a number of diagnostic approaches that contribute to the new
COVID-19 pneumonia, such as radiographic or laboratory
result, PCR, Microarray, LAMP, and CRISPR strategies.
To date, the number of infected cases is rapidly elevated.
This necessitates uncovering the viral mystery of the mo-
lecular pathways to provide a future investigation for
emerging targeted vaccines and antiviral treatment. We
expect that these strategies, in combination with the follow-
up laboratory studies aimed at assessing the computationally
anticipated antiviral agents, would allow us to have a
broader range of potential repurposed drugs in the event of
any possible pandemic virus.
Disclosure Statement
No competing financial interests exist.
Funding Information
No funding was received for this article.
References
Agostini, M.L., Andres, E.L., Sims, A.C., Graham, R.L.,
Sheahan, T.P., Lu, X., et al. (2018). Coronavirus suscepti-
bility to the antiviral remdesivir (GS-5734) is mediated by the
viral polymerase and the proofreading exoribonuclease. mBio
9, e00221–18.
Ahmed, S.F., Quadeer, A.A., and McKay, M.R.J.V. (2020).
Preliminary identification of potential vaccine targets for the
COVID-19 coronavirus (SARS-CoV-2) based on SARS-CoV
immunological studies. Viruses 12, 254.
Ahmed-Belkacem, R., Sutto-Ortiz, P., Guiraud, M., Canard, B.,
Vasseur, J.-J., Decroly, E., et al. (2020). Synthesis of adenine
dinucleosides SAM analogs as specific inhibitors of SARS-
CoV nsp14 RNA cap guanine-N7-methyltransferase. Eur J
Med Chem 201, 112557.
Ahn, D.-G., Choi, J.-K., Taylor, D.R., and Oh, J.-W. (2012).
Biochemical characterization of a recombinant SARS cor-
8 ASGHARI ET AL.
Downloaded by 217.219.70.5 from www.liebertpub.com at 08/02/20. For personal use only.
onavirus nsp12 RNA-dependent RNA polymerase capable of
copying viral RNA templates. Arch Virol 157, 2095–2104.
Alam, I., Kamau, A., Kulmanov, M., Arold, S.T., Pain, A.,
Gojobori, T., et al. (2020). Functional pangenome analysis
suggests inhibition of the protein E as a readily available
therapy for COVID-2019. BioRxiv [Preprint]. https://doi.org/
10.1101/2020.02.17.952895.
Algarroba, G.N., Rekawek, P., Vahanian, S.A., Khullar, P.,
Palaia, T., Peltier, M.R., et al. (2020). Visualization of SARS-
CoV-2 virus invading the human placenta using electron
microscopy. Am J Obstet Gynecol [Epub ahead of print];
DOI: 10.1016/j.ajog.2020.05.023.
Al-Tawfiq, J.A., Al-Homoud, A.H., Memish, Z.A.J.T.M., and
Disease, I. (2020). Remdesivir as a possible therapeutic op-
tion for the COVID-19. Travel Med Infect Dis 34, 101615
Angeletti, S., Benvenuto, D., Bianchi, M., Giovanetti, M.,
Pascarella, S., and Ciccozzi, M. (2020). COVID-2019: the
role of the nsp2 and nsp3 in its pathogenesis. J Med Virol 92,
584–588.
Angelini, M.M., Akhlaghpour, M., Neuman, B.W., and Buch-
meier, M.J.J.M. (2013). Severe acute respiratory syndrome
coronavirus nonstructural proteins 3, 4, and 6 induce double-
membrane vesicles. mBio 4, e00524-13.
Arndt, A.L., Larson, B.J., and Hogue, BGJJov. (2010). A con-
served domain in the coronavirus membrane protein tail is
important for virus assembly. J Virol 84, 11418–11428.
Ba
´rcena, M., Oostergetel, G.T., Bartelink, W., Faas, F.G.,
Verkleij, A., Rottier, P.J., et al. (2009). Cryo-electron to-
mography of mouse hepatitis virus: insights into the structure
of the coronavirion. Proc Natl Acad Sci U S A 106, 582–587.
Bauch, C.T., and Oraby, T.J.T.L. (2013). Assessing the pan-
demic potential of MERS-CoV. Lancet 382, 662–664.
Beck, B.R., Shin, B., Choi, Y., Park, S., Kang, K.J.C., and
journal sb. (2020). Predicting commercially available antivi-
ral drugs that may act on the novel coronavirus (SARS-CoV-
2) through a drug-target interaction deep learning model.
Comput Struct Biotechnol J 18, 784–790.
Begum, F., Banerjee, A.K., Tripathi, P.P., and Ray, U.
(2020). Two mutations P/L and Y/C in SARS-CoV-2 he-
licase domain exist together and influence helicase RNA
binding. BioRxiv [Preprint]. https://doi.org/10.1101/2020
.05.14.095224.
Benvenuto, D., Angeletti, S., Giovanetti, M., Bianchi, M.,
Pascarella, S., Cauda, R., et al. (2020). Evolutionary analysis
of SARS-CoV-2: how mutation of non-structural protein 6
(NSP6) could affect viral autophagy. J Infect 81, e24–e7.
Bestle, D., Heindl, M.R., Limburg, H., van, T.V.L., Pilgram, O.,
Moulton, H., et al. (2020) TMPRSS2 and furin are both es-
sential for proteolytic activation and spread of SARS-CoV-2
in human airway epithelial cells and provide promising drug
targets. bioRxiv 2020.04.15.042085.
Bhardwaj, K., Palaninathan, S., Alcantara JMO, Yi, L.L., Guar-
ino, L., Sacchettini, J.C., et al. (2008). Structural and functional
analyses of the severe acute respiratory syndrome coronavirus
endoribonuclease Nsp15. J Biol Chem 283, 3655–3664.
Braakman, I., and Van Anken, E.J.T. (2000). Folding of viral
envelope glycoproteins in the endoplasmic reticulum. Traffic
1, 533–539.
Cai, Q., Yang, M., Liu, D., Chen, J., Shu, D., Xia, J., et al.
(2020). Experimental treatment with favipiravir for COVID-
19: an open-label control study. Engineering (in press). http://
doi.org/10.1016/j.eng.2020.03.007.
Cao, B., Wang, Y., Wen, D., Liu, W., Wang, J., Fan, G. (2020).
A trial of lopinavir-ritonavir in adults hospitalized with severe
Covid-19 [published online ahead of print March 18, 2020].
J Med 382, 1787–1799.
Ceraolo, C., and Giorgi, F.M. (2020). Genomic variance of the
2019-nCoV coronavirus. J Med Virol 92, 522–528.
Chang, C.-K., Sue, S.-C., Yu, T.-H., Hsieh, C.-M., Tsai, C.-K.,
Chiang, Y.-C., et al. (2006). Modular organization of SARS
coronavirus nucleocapsid protein. J Biomed Sci 13, 59–72.
Chen, Q., Li, J., Deng, Z., Xiong, W., Wang, Q., and Hu, Y.-Q.
(2010). Comprehensive detection and identification of seven
animal coronaviruses and human respiratory coronavirus
229E with a microarray hybridization assay. Intervirology 53,
95–104.
Chen, Y., Cai, H., Pan Ja, Xiang, N., Tien, P., Ahola, T., et al.
(2009). Functional screen reveals SARS coronavirus non-
structural protein nsp14 as a novel cap N7 methyltransferase.
Proc Natl Acad Sci U S A 106, 3484–3489.
Chen, Y., Yu, Z., Yi, H., Wei, Y., Han, X., Li, Q., et al. (2019).
The phosphorylation of the N protein could affect PRRSV
virulence in vivo. Vet Microbiol 231, 226–231.
Chung, M., Bernheim, A., Mei, X., Zhang, N., Huang, M.,
Zeng, X., et al. (2020). CT imaging features of 2019 novel
coronavirus (2019-nCoV). Radiology 295, 202–207.
Cong, Y., Ulasli, M., Schepers, H., Mauthe, M., V’kovski P,
Kriegenburg, F., et al. (2020). Nucleocapsid protein recruit-
ment to replication-transcription complexes plays a crucial
role in coronaviral life cycle. J Virol 94, e01925–19.
Corman, V., Eckerle, I., Bleicker, T., Zaki, A., Landt, O.,
Eschbach-Bludau, M., et al. (2012). Detection of a novel
human coronavirus by real-time reverse-transcription poly-
merase chain reaction. Eurosurveillance 17, 20285.
Corman, V.M., Landt, O., Kaiser, M., Molenkamp, R., Meijer,
A., Chu, D.K., et al. (2020). Detection of 2019 novel cor-
onavirus (2019-nCoV) by real-time RT-PCR. Euro-
surveillance 25, 2000045.
Cornillez-Ty, C.T., Liao, L., Yates, J.R., Kuhn, P., and Buch-
meier, M.J. (2009). Severe acute respiratory syndrome cor-
onavirus nonstructural protein 2 interacts with a host protein
complex involved in mitochondrial biogenesis and intracel-
lular signaling. J Virol 83, 10314–10318.
Cui, J., Li, F., and Shi, Z.-LJNrM. (2019). Origin and evolution
of pathogenic coronaviruses. Nat Rev Microbiol 17, 181–192.
De Clercq, E.J.C.A.A.J. (2019). New nucleoside analogues for
the treatment of hemorrhagic fever virus infections. Chem
Asian J 14, 3962–3968.
Degtyarev, M., De Mazie
`re, A., Orr, C., Lin, J., Lee, B.B., Tien,
J.Y., et al. (2008). Akt inhibition promotes autophagy and
sensitizes PTEN-null tumors to lysosomotropic agents. J Cell
Biol 183, 101–116.
De Maio, F., Cascio, E.L., Babini, G., Sali, M., Della Longa, S.,
Tilocca, B., et al. (2020). Enhanced binding of SARS-CoV-2
Envelope protein to tight junction-associated PALS1 could
play a key role in COVID-19 pathogenesis. Research Square
[Preprint]. DOI: 10.21203/rs.3rs-30903/v1.
Deng, X., Hackbart, M., Mettelman, R.C., O’Brien, A., Mie-
lech, A.M., Yi, G., et al. (2017). Coronavirus nonstructural
protein 15 mediates evasion of dsRNA sensors and limits
apoptosis in macrophages. PNAS 114, E4251–E60.
de Wilde, A.H., Raj, V.S., Oudshoorn, D., Bestebroer, T.M.,
van Nieuwkoop, S., Limpens, R.W., et al. (2013). MERS-
coronavirus replication induces severe in vitro cytopathology
and is strongly inhibited by cyclosporin A or interferon-a
treatment. J Gen Virol 94, 1749.
Durdagi, S., Aksoydan, B., Dogan, B., Sahin, K., and Shahraki,
A. (2020). Screening of clinically approved and investigation
THE NOVEL INSIGHT OF SARS-COV-2 MOLECULAR BIOLOGY 9
Downloaded by 217.219.70.5 from www.liebertpub.com at 08/02/20. For personal use only.
drugs as potential inhibitors of COVID-19 main protease: A
virtual drug repurposing study. ResearchGate [Preprint]. DOI:
10.26434/chemrxiv.12032712.v1.
Elfiky, A.A.J.L.S. (2020). Ribavirin, remdesivir, sofosbuvir,
galidesivir, and tenofovir against SARS-CoV-2 RNA depen-
dent RNA polymerase (RdRp): a molecular docking study.
Life Sci 253, 117592.
Enosawa, M., Kageyama, S., Sawai, K., Watanabe, K., Notomi,
T., Onoe, S., et al. (2003). Use of loop-mediated isothermal
amplification of the IS900 sequence for rapid detection of
cultured Mycobacterium avium subsp. paratuberculosis.
J Clin Microbiol 41, 4359–4365.
Fehr, A.R., and Perlman, S. (2015). Coronaviruses: an overview
of their replication and pathogenesis. In Coronaviruses.
Springer, pp. 1–23.
Forni, D., Cagliani, R., Clerici, M., and Sironi, MJTim. (2017).
Molecular evolution of human coronavirus genomes. J Gen
Virol 25, 35–48.
Forni, D., Cagliani, R., Mozzi, A., Pozzoli, U., Al-Daghri, N.,
Clerici, M., et al. (2016). Extensive positive selection drives
the evolution of nonstructural proteins in lineage C betacor-
onaviruses. J Virol 90, 3627–3639.
Freije, C.A., Myhrvold, C., Boehm, C.K., Lin, A.E., Welch, N.L.,
Carter, A., et al. (2019). Programmable inhibition and detec-
tion of RNA viruses using Cas13. Mol Cell 76, 826–837. e11.
Fu, J., Chen, R., Hu, J., Qu, H., Zhao, Y., Cao, S., et al. (2020).
Identification of a novel linear B-cell epitope on the nucleo-
capsid protein of porcine deltacoronavirus. Int J Mol Sci 21,
648.
Fung, T.S., Liao, Y., and Liu, D.X.J.V. (2016). Regulation of
stress responses and translational control by coronavirus.
Viruses 8, 184.
Furuta, Y., Komeno, T., Nakamura, TJPotJA, and Series, B.
(2017). Favipiravir (T-705), a broad spectrum inhibitor of
viral RNA polymerase. Proc Jpn Acad Ser B Phys Biol Sci
93, 449–463.
Gadlage, M.J., Graham, R.L., and Denison, M.R. (2008).
Murine coronaviruses encoding nsp2 at different genomic loci
have altered replication, protein expression, and localization.
J Virol 82, 11964–11969.
Gadlage, M.J., Sparks, J.S., Beachboard, D.C., Cox, R.G.,
Doyle, J.D., Stobart, C.C., et al. (2010). Murine hepatitis
virus nonstructural protein 4 regulates virus-induced mem-
brane modifications and replication complex function. J Virol
84, 280–290.
Gootenberg, J.S., Abudayyeh, O.O., Kellner, M.J., Joung, J.,
Collins, J.J., and Zhang, F. (2018). Multiplexed and portable
nucleic acid detection platform with Cas13, Cas12a, and
Csm6. Science 360, 439–444.
Gootenberg, J.S., Abudayyeh, O.O., Lee, J.W., Essletzbichler,
P., Dy, A.J., Joung, J., et al. (2017). Nucleic acid detection
with CRISPR-Cas13a/C2c2. Science 356, 438–442.
Graham, R.L., Sims, A.C., Brockway, S.M., Baric, R.S., and
Denison, MRJJov. (2005). The nsp2 replicase proteins of
murine hepatitis virus and severe acute respiratory syndrome
coronavirus are dispensable for viral replication. J Virol 79,
13399–13411.
Grunewald, M.E., Fehr, A.R., Athmer, J., and Perlman, S.J.V.
(2018). The coronavirus nucleocapsid protein is ADP-
ribosylated. Virology 517, 62–68.
Hagemeijer, M.C., Verheije, M.H., Ulasli, M., Shaltie
¨l, I.A., de
Vries, L.A., Reggiori, F., et al. (2010). Dynamics of cor-
onavirus replication-transcription complexes. J Virol 84,
2134–2149.
Hao, W., Wojdyla, J.A., Zhao, R., Han, R., Das, R., Zlatev, I.,
et al. (2017). Crystal structure of Middle East respiratory
syndrome coronavirus helicase. PLoS Pathog 13, e1006474.
He, Q., Li, Y., Zhou, L., Ge, X., Guo, X., and Yang, HJVr.
(2015). Both Nsp1band Nsp11 are responsible for differential
TNF-aproduction induced by porcine reproductive and re-
spiratory syndrome virus strains with different pathogenicity
in vitro. Virus Res 201, 32–40.
Heald-Sargent, T., and Gallagher, T.J.V. (2012). Ready, set,
fuse! The coronavirus spike protein and acquisition of fusion
competence. Viruses 4, 557–580.
Heurich, A., Hofmann-Winkler, H., Gierer, S., Liepold, T., Jahn,
O., and Po
¨hlmann, SJJov. (2014). TMPRSS2 and ADAM17
cleave ACE2 differentially and only proteolysis by TMPRSS2
augments entry driven by the severe acute respiratory syn-
drome coronavirus spike protein. J Virol 88, 1293–1307.
Hoffmann, M., Kleine-Weber, H., Kru
¨ger, N., Mueller, M.A.,
Drosten, C., and Po
¨hlmann, S.J.B. (2020a). The novel cor-
onavirus 2019 (2019-nCoV) uses the SARS-coronavirus re-
ceptor ACE2 and the cellular protease TMPRSS2 for entry
into target cells. BioRxiv [Preprint]. https://doi.org/10.1101/
2020.01.31.929042.
Hoffmann, M., Kleine-Weber, H., Schroeder, S., Kru
¨ger, N.,
Herrler, T., Erichsen, S., et al. (2020b). SARS-CoV-2 cell
entry depends on ACE2 and TMPRSS2 and is blocked by a
clinically proven protease inhibitor. Cell 181, 271–280.e8.
Hu, Y., Li, W., Gao, T., Cui, Y., Jin, Y., Li, P., et al. (2017). The
severe acute respiratory syndrome coronavirus nucleocapsid
inhibits type I interferon production by interfering with
TRIM25-mediated RIG-I ubiquitination. J Virol 91, e02143–16.
Huang, C., Lokugamage, K.G., Rozovics, J.M., Narayanan, K.,
Semler, B.L., and Makino, S. (2011). SARS coronavirus nsp1
protein induces template-dependent endonucleolytic cleavage
of mRNAs: viral mRNAs are resistant to nsp1-induced RNA
cleavage. PLoS Pathog 7, e1002433-e.
Hung, A.Y., and Sheng, MJJoBC. (2002). PDZ domains:
structural modules for protein complex assembly. J Biol
Chem 277, 5699–5702.
Javier, R.T., and Rice, A.P. (2011). Emerging theme: cellular
PDZ proteins as common targets of pathogenic viruses.
J Virol 85, 11544–11556.
Jia, Z., Yan, L., Ren, Z., Wu, L., Wang, J., Guo, J., et al. (2019).
Delicate structural coordination of the Severe Acute Re-
spiratory Syndrome coronavirus Nsp13 upon ATP hydrolysis.
Nucleic Acids Res 47, 6538–6550.
Jiang, S., Hillyer, C., and Du, LJTiI. (2020). Neutralizing an-
tibodies against SARS-CoV-2 and other human cor-
onaviruses. Trends Immunol 41, 355–359.
Ju, J., Li, X., Kumar, S., Jockusch, S., Chien, M., Tao, C., et al.
(2020). Nucleotide analogues as inhibitors of SARS-CoV
polymerase. BioRxiv [Preprint]. https://doi.org/10.1101/2020
.03.12.989186.
Kahn, J.S., and Mcintoch, K. (2005). History and recent ad-
vances in coronavirus discovery. Pediatr Infect Dis J 24,
5223–5257.
Kamitani, W., Huang, C., Narayanan, K., Lokugamage, K.G.,
Makino, S.J.Ns., and Biology, M. (2009). A two-pronged
strategy to suppress host protein synthesis by SARS cor-
onavirus Nsp1 Protein 16, 1134.
Kamitani, W., Narayanan, K., Huang, C., Lokugamage, K.,
Ikegami, T., Ito, N., et al. (2006). Severe acute respiratory
syndrome coronavirus nsp1 protein suppresses host gene ex-
pression by promoting host mRNA degradation. Proc Natl
Acad Sci U S A 103, 12885–12890.
10 ASGHARI ET AL.
Downloaded by 217.219.70.5 from www.liebertpub.com at 08/02/20. For personal use only.
Kang, S., Yang, M., Hong, Z., Zhang, L., Huang, Z., Chen, X.,
et al. (2020). Crystal structure of SARS-CoV-2 nucleocapsid
protein RNA binding domain reveals potential unique drug
targeting sites. Acta Pharm Sin B (in press). http://doi.org/10
.1016/j.apsb.2020.04.009.
Kim, Y., Jedrzejczak, R., Maltseva, N.I., Endres, M., Godzik,
A., Michalska, K., et al. (2020). Crystal structure of Nsp15
endoribonuclease NendoU from SARS-CoV-2.
Kindler, E., Gil-Cruz, C., Spanier, J., Li, Y., Wilhelm, J., Ra-
bouw, H.H., et al. (2017). Early endonuclease-mediated
evasion of RNA sensing ensures efficient coronavirus repli-
cation. PLoS Pathog 13, e1006195.
Kirchdoerfer, R.N., and Ward, ABJNc. (2019). Structure of the
SARS-CoV nsp12 polymerase bound to nsp7 and nsp8 co-
factors. Nat Commun 10, 1–9.
Konkolova, E., Klima, M., Nencka, R., and Boura, E. (2020).
Structural analysis of the putative SARS-CoV-2 primase
complex. J Struct Biol 211, 107548.
Kumar, A., Parveen, A., Kumar, N., Bairy, S., Kaushik, V.,
Chandola, C., et al. (2020). Characterization of nucleocapsid
(N) protein from novel coronavirus SARS-CoV-2. Preprints
2020, 2020050413.
Kumar, V., Tan, K.-P., Wang, Y.-M., Lin, S.-W., Liang,
P.-H.J.B., and Chemistry, M. (2016). Identification, synthesis
and evaluation of SARS-CoV and MERS-CoV 3C-like pro-
tease inhibitors. Bioorg Med Chem 24, 3035–3042.
Kuo, L., Koetzner, C.A., and Masters, P.S.J.V. (2016). A key
role for the carboxy-terminal tail of the murine coronavirus
nucleocapsid protein in coordination of genome packaging.
Virology 494, 100–107.
Lai, M.M., and Cavanagh, D. (1997). The molecular biology
of coronaviruses. In Advances in Virus Research. Abany,
New York, Elsevier, pp. 1–100.
Lea
˜o, J.C., Gusmao, T.P.L., ZarZar, A.M., Filho, J.C.L., Faria,
A.B.S., Gueiros, L. et al. (2020). Coronaviridae-old friends,
new enemy! Oral Dis 001:1–9.
Lee, C. (2019). Griffithsin, a highly potent broad-spectrum
antiviral lectin from red algae: from discovery to clinical
application. Mar Drugs 17, 567.
Lee, Y.G., Park, J.H., Jeon, E.S., Kim, J.H., and Lim, B.K.
(2016). Fructus amomi cardamomi extract inhibits
coxsackievirus-B3 induced myocarditis in a murine myocar-
ditis model. J Microbiol Biotechnol 26, 2012–2018.
Lehmann, K.C., Gulyaeva, A., Zevenhoven-Dobbe, J.C., Jans-
sen, G.M., Ruben, M., Overkleeft, H.S., et al. (2015). Dis-
covery of an essential nucleotidylating activity associated
with a newly delineated conserved domain in the RNA
polymerase-containing protein of all nidoviruses. Nucleic
Acids Res 43, 8416–8434.
Lei, J., Kusov, Y., and Hilgenfeld, RJAr. (2018) Nsp3 of cor-
onaviruses: structures and functions of a large multi-domain
protein. Antiviral Res 149, 58–74.
Li, C., Yang, Y., Ren, L.J.I., and Genetics, Evolution. (2020).
Genetic evolution analysis of 2019 novel coronavirus and
coronavirus from other species. Infect Genet Evol 82,
104285.
Li, F. (2016). Structure, function, and evolution of coronavirus
spike proteins. Annu Rev 3, 237–261.
Li, F., Berardi, M., Li, W., Farzan, M., Dormitzer, P.R., and
Harrison, SCJJov. (2006). Conformational states of the severe
acute respiratory syndrome coronavirus spike protein ecto-
domain. J Virol 80, 6794–6800.
Li, Y., Zhou, L., Zhang, J., Ge, X., Zhou, R., Zheng, H., et al.
(2014). Nsp9 and Nsp10 contribute to the fatal virulence of
highly pathogenic porcine reproductive and respiratory syn-
drome virus emerging in China. PLoS Pathog 10, e1004216.
Liao, Y., Yuan, Q., Torres, J., Tam, J., and Liu DJV. (2006).
Biochemical and functional characterization of the membrane
association and membrane permeabilizing activity of the se-
vere acute respiratory syndrome coronavirus envelope pro-
tein. Virology 349, 264–275.
Lim, J., Jeon, S., Shin, H.-Y., Kim, M.J., Seong, Y.M., Lee,
W.J., et al. (2020). Case of the index patient who caused
tertiary transmission of COVID-19 infection in Korea: the
application of lopinavir/ritonavir for the treatment of COVID-
19 infected pneumonia monitored by quantitative RT-PCR.
J Korean Med Sci 35, e79.
Lin, S., Shen, R., He, J., Li, X., and Guo, X. (2020). Molecular
modeling evaluation of the binding effect of ritonavir, lopi-
navir and darunavir to severe acute respiratory syndrome
coronavirus 2 proteases. BioRxiv [Preprint]. http://doi.org/10
.1101/2020.01.31/929695.
Lopez, L.A., Riffle, A.J., Pike, S.L., Gardner, D., and Hogue,
BGJJov. (2008). Importance of conserved cysteine resi-
dues in the coronavirus envelope protein. J Virol 82, 3000–
3010.
Lu, X., Whitaker, B., Sakthivel, S.K.K., Kamili, S., Rose, L.E.,
Lowe, L., et al. (2014). Real-time reverse transcription-PCR
assay panel for Middle East respiratory syndrome cor-
onavirus. J Clin Microbiol 52, 67–75.
Lugari, A., Betzi, S., Decroly, E., Bonnaud, E., Hermant, A.,
Guillemot, J.-C., et al. (2010). Molecular mapping of the
RNA Cap 2¢-O-methyltransferase activation interface be-
tween severe acute respiratory syndrome coronavirus nsp10
and nsp16. J Biol Chem 285, 33230–33241.
Ma, Y., Wu, L., Shaw, N., Gao, Y., Wang, J., Sun, Y., et al.
(2015). Structural basis and functional analysis of the SARS
coronavirus nsp14–nsp10 complex. Proc Natl Acad Sci U S A
112, 9436–9441.
Macchiagodena, M., Pagliai, M., and Procacci, P. (2020).
Identification of potential binders of the main protease
3CL(pro) of the COVID-19 via structure-based ligand design
and molecular modeling. Chem Phys Lett 750, 137489.
Macnaughton, M., Davies, H.A., and Nermut, MJJoGV. (1978).
Ribonucleo protein-like structures from coronavirus particles.
J Gen Virol 39, 545–549.
Ma-Lauer, Y., Carbajo-Lozoya, J., Hein, M.Y., Mu
¨ller, M.A.,
Deng, W., Lei, J., et al. (2016). p53 down-regulates SARS
coronavirus replication and is targeted by the SARS-unique
domain and PLpro via E3 ubiquitin ligase RCHY1. PNAS
113, E5192–E201.
Masters, PSJAivr. (2006). The molecular biology of cor-
onaviruses. Adv Virus Res 66, 193–292.
Matsuyama, S., Kawase, M., Nao, N., Shirato, K., Ujike, M.,
Kamitani, W., et al. (2020). The inhaled corticosteroid ci-
clesonide blocks coronavirus RNA replication by targeting
viral NSP15.2020.03.11.987016. BioRxiv [Preprint]. http://
doi.org/10.1101/2020.03.11.987016.
Menachery, V.D., Graham, R.L., and Baric, RSJCoiv. (2017).
Jumping species—a mechanism for coronavirus persistence
and survival. Curr Opin Virol 23, 1–7.
Minskaia, E., Hertzig, T., Gorbalenya, A.E., Campanacci, V.,
Cambillau, C., Canard, B., et al. (2006). Discovery of an
RNA virus 3¢/5¢exoribonuclease that is critically involved
in coronavirus RNA synthesis. PNAS 103, 5108–5113.
Mirza, M.U., and Froeyen, M. (2020). Structural elucidation of
SARS-CoV-2 vital proteins: computational methods reveal
potential drug candidates against Main protease, Nsp12 RNA-
THE NOVEL INSIGHT OF SARS-COV-2 MOLECULAR BIOLOGY 11
Downloaded by 217.219.70.5 from www.liebertpub.com at 08/02/20. For personal use only.
dependent RNA polymerase and Nsp13 helicase. J Pharm
Anal [Epub ahead of print]; DOI: 10.1016/j.jpha.2020.04.008.
Narayanan, N., and Nair, D.T. (2020). Vitamin B12 may inhibit
RNA-dependent-RNA polymerase activity of nsp12 from the
COVID-19 virus.
National Health and Health Commission and the provincial
health and health commission, including Hong Kong, Macao,
and Taiwan. (2020). Distribution of New Coronavirus Pneu-
monia. [translated from Chinese]. http://2019ncov.chinacdc
.cn/2019-ncov/
Nedialkova, D.D., Ulferts, R., van den Born, E., Lauber, C.,
Gorbalenya, A.E., Ziebuhr, J., et al. (2009). Biochemical
characterization of arterivirus nonstructural protein 11 reveals
the nidovirus-wide conservation of a replicative en-
doribonuclease. J Virol 83, 5671–5682.
Nieto-Torres, J.L., DeDiego, M.L., A
´lvarez, E., Jime
´nez-
Guarden
˜o, J.M., Regla-Nava, J.A., Llorente, M., et al. (2011).
Subcellular location and topology of severe acute respiratory
syndrome coronavirus envelope protein. Virology 415,
69–82.
Notomi, T., Okayama, H., Masubuchi, H., Yonekawa, T., Wa-
tanabe, K., Amino, N., et al. (2000). Loop-mediated iso-
thermal amplification of DNA. Nucleic Acids Res 28, e63–e.
Oostra, M., De Haan, C.A.M., De Groot, R.J., and Rottier,
P.J.M. (2006). Glycosylation of the severe acute respiratory
syndrome coronavirus triple-spanning membrane proteins 3a
and M. J Virol 80, 2326–2336.
Ozma, M.A., Maroufi, P., Khodadadi, E., Ko
¨se, Sx., Esposito, I.,
Ganbarov, K., et al. (2020). Clinical manifestation, diagnosis,
prevention and control of SARS-CoV-2 (COVID-19) during
the outbreak period. Le Inf Med 28, 153–165.
Pan, Y., Guan, H., Zhou, S., Wang, Y., Li, Q., Zhu, T., et al.
(2020). Initial CT findings and temporal changes in patients
with the novel coronavirus pneumonia (2019-nCoV): a study
of 63 patients in Wuhan, China. Eur Radiol 30, 3306–3309.
Perlman, S., and Netland, J. (2009). Coronaviruses post-SARS:
update on replication and pathogenesis. Nat Rev Microbiol 7,
439–450.
Pfefferle, S., Scho
¨pf, J., Ko
¨gl, M., Friedel, C.C., Mu
¨ller, M.A.,
Carbajo-Lozoya, J., et al. (2011). The SARS-coronavirus-host
interactome: identification of cyclophilins as target for pan-
coronavirus inhibitors. Plos Pathog 7, e1002331.
Ray, M., Sarkar, S., Rath, S.N., and Rath, M.S.N. (2020).
Druggability for COVID19–In silico discovery of Potential
Drug Compounds against Nucleocapsid (N) Protein of SARS-
CoV-2. ChemRxiv [Preprint]. https://doi.org/10.26434/
chemrxiv.12387290.v1.
Ren, L.-L., Wang, Y.-M., Wu, Z.-Q., Xiang, Z.-C., Guo, L., Xu,
T., et al. (2020). Identification of a novel coronavirus causing
severe pneumonia in human: a descriptive study. Chin Med J
133, 1015–1024.
Rosas-Lemus, M., Minasov, G., Shuvalova, L., Inniss, N.,
Kiryukhina, O., Wiersum, G., et al. (2020). The crystal
structure of nsp10-nsp16 heterodimer from SARS-CoV-2 in
complex with S-adenosylmethionine. BioRxiv [Preprint].
https://doi.org/10.1101/2020.04.17.047498.
Schoeman, D., and Fielding, BCJVj. (2019). Coronavirus en-
velope protein: current knowledge. Virol J 16, 69.
Setianingsih, T.Y., Wiyatno, A., Hartono, T.S., Hindawati, E.,
Dewantari, A.K., Myint, K.S., et al. (2019). Detection of
multiple viral sequences in the respiratory tract samples of
suspected Middle East respiratory syndrome coronavirus pa-
tients in Jakarta, Indonesia 2015–2016. Int J Infect Dis 86,
102–107.
Sevajol, M., Subissi, L., Decroly, E., Canard, B., and Imbert,
IJVr. (2014). Insights into RNA synthesis, capping, and
proofreading mechanisms of SARS-coronavirus. Virus Res
194, 90–99.
Shang, J., Wan, Y., Luo, C., Ye, G., Geng, Q., Auerbach, A.,
et al. (2020). Cell entry mechanisms of SARS-CoV-2. Proc
Natl Acad Sci U S A 117, 11727.
Shanker, A., Bhanu, D., and Alluri, A. (2020). Analysis of
whole genome sequences and homology modelling of a 3C
like peptidase and a non-structural protein of the novel cor-
onavirus COVID-19 shows protein ligand interaction with an
Aza-peptide and a noncovalent lead inhibitor with possible
antiviral properties. ChemRxiv [Preprint]. https://doi.org/10
.26434/chemrxiv.11846943.v9.
Shi, H., Han, X., and Zheng, C. (2020). Evolution of CT
manifestations in a patient recovered from 2019 novel cor-
onavirus (2019-nCoV) pneumonia in Wuhan, China. Radi-
ology 295, 20.
Shi, P., Su, Y., Li, R., Liang, Z., Dong, S., and Huang, JJVr.
(2019). PEDV nsp16 negatively regulates innate immunity to
promote viral proliferation. Virus Res 265, 57–66.
Shu, T., Huang, M., Wu, D., Ren, Y., Zhang, X., Han, Y., et al.
(2020). SARS-coronavirus-2 nsp13 possesses NTPase and
RNA helicase activities. Viral Sin 30, 1–9.
Singh, S., and Sharma, B.B. (2020). Severe acute respiratory
syndrome-coronavirus 2 and novel coronavirus disease 2019:
an extraordinary pandemic. Lung India 37, 268–271.
Snijder, E., Decroly, E., and Ziebuhr, J. (2016). The nonstruc-
tural proteins directing coronavirus RNA synthesis and pro-
cessing. Adv Virus Res. 96, 59–126.
Song, W., Gui, M., Wang, X., and Xiang, Y. (2018). Cryo-EM
structure of the SARS coronavirus spike glycoprotein in
complex with its host cell receptor ACE2. PLoS Pathog 14,
e1007236.
Sparks, J.S., Lu, X., and Denison, MRJJov. (2007). Genetic
analysis of murine hepatitis virus nsp4 in virus replication.
J Virol 81, 12554–12563.
Su, S., Wong, G., Shi, W., Liu, J., Lai, A.C., Zhou, J., et al.
(2016). Epidemiology, genetic recombination, and patho-
genesis of coronaviruses. Trends Microbiol 24, 490–502.
Sun, Y., Li, D., Giri, S., Prasanth, S.G., and Yoo, DJBri.
(2014a). Differential host cell gene expression and regulation
of cell cycle progression by nonstructural protein 11 of por-
cine reproductive and respiratory syndrome virus. Biomed
Res Int 2014, 430508.
Sun, Y., Wang, Z., Tao, J., Wang, Y., Wu, A., Yang, Z., et al.
(2014b). Yeast-based assays for the high-throughput screen-
ing of inhibitors of coronavirus RNA cap guanine-N7-
methyltransferase. Antiviral Res 104, 156–164.
Sutton, G., Fry, E., Carter, L., Sainsbury, S., Walter, T., Nettleship,
J., et al. (2004). The nsp9 replicase protein of SARS-coronavirus,
structure and functional insights. Structure 12, 341–353.
Tam, R. (2002). Modulation of immune response by ribavirin.
In Google Patents. United States. Patent application, pub-
lication number: WO/2000/044388.
Tam, R.C., Lau, J.Y.N., and Hong, Z. (2001). Mechanisms of
action of ribavirin in antiviral therapies. Antiviral Chem
Chemother 12, 261–272.
Tanaka, Y., Sato, Y., and Sasaki, T.J.V. (2013). Suppression of
coronavirus replication by cyclophilin inhibitors. Viruses 5,
1250–1260.
Te, H.S., Randall, G., Jensen, D.M. (2007). Mechanism of ac-
tion of ribavirin in the treatment of chronic hepatitis C.
Gastroenterol Hepatol 3, 218.
12 ASGHARI ET AL.
Downloaded by 217.219.70.5 from www.liebertpub.com at 08/02/20. For personal use only.
te Velthuis, A.J.W., Arnold, J.J., Cameron, C.E., van den
Worm, S.H.E., and Snijder, E.J. (2009). The RNA polymerase
activity of SARS-coronavirus nsp12 is primer dependent.
Nucleic Acids Res 38, 203–214.
Thomas, S. (2020). The structure of the membrane protein
of SARS-CoV-2 resembles the sugar transporter semi-
SWEET. Preprints 2020040512 DOI: 10.20944/preprints
202004.0512.v1.
Tian, X., Li, C., Huang, A., Xia, S., Lu, S., Shi, Z., et al. (2020).
Potent binding of 2019 novel coronavirus spike protein by a
SARS coronavirus-specific human monoclonal antibody.
Emerg Microbes Infect 9, 382–385.
Tseng, Y.-T., Wang, S.-M., Huang, K.-J., and Wang, C.-T.
(2014). SARS-CoV envelope protein palmitoylation or nu-
cleocapid association is not required for promoting virus-like
particle production. J Biomed Sci 21, 34.
Uhlenhaut, C., Cohen, J.I., Pavletic, S., Illei, G., Gea-Bana-
cloche, J.C., Abu-Asab, M., et al. (2012). Use of a novel virus
detection assay to identify coronavirus HKU1 in the lungs of
a hematopoietic stem cell transplant recipient with fatal
pneumonia. Transplant Infect Dis 14, 79–85.
von Brunn, A., Teepe, C., Simpson, J.C., Pepperkok, R., Frie-
del, C.C., Zimmer, R., et al. (2007). Analysis of intraviral
protein-protein interactions of the SARS coronavirus OR-
Feome. PLoS One 2, e459–e.
Walls, A.C., Tortorici, M.A., Snijder, J., Xiong, X., Bosch, B.-
J., Rey, F.A., et al. (2017). Tectonic conformational changes
of a coronavirus spike glycoprotein promote membrane fu-
sion. Proc Natl Acad Sci U S A 114, 11157–11162.
Wan, Z., Zhang Yn, He, Z., Liu, J., Lan, K., Hu, Y., et al.
(2016). A melting curve-based multiplex RT-qPCR assay for
simultaneous detection of four human coronaviruses. Int J
Mol Sci 17, 1880.
Wang, Q., Qiu, Y., Li, J.-Y., Zhou, Z.-J., Liao, C.-H., and Ge,
X.-Y. (2020a). A unique protease cleavage site predicted in
the spike protein of the novel pneumonia coronavirus (2019-
nCoV) potentially related to viral transmissibility. Virol Sin
20, 1–3.
Wang, S., Zhang, Y., Liu, S., Peng, H., and Mackey,
V.J.J.I.D.T. (2020b). Coronaviruses and the associated po-
tential therapeutics for the viral infections. J Infect Dis 8, 2.
Wathelet, M.G., Orr, M., Frieman, M.B., and Baric, R.S.
(2007). Severe acute respiratory syndrome coronavirus
evades antiviral signaling: role of nsp1 and rational design of
an attenuated strain. J Virol 81, 11620–11633.
We˛drowska, E., Wandtke, T., Senderek, T., Piskorska, E., and
Kopin
´ski, P. (2020). Coronaviruses fusion with the membrane
and entry to the host cell. Ann Agri Environ Med 27, 175–
183.
World Health Organization. (2020). Statement on the meeting of
the International Health Regulations (2005) Emergency
Committee regarding the outbreak of novel coronavirus
(2019-nCoV).
Wrapp, D., Wang, N., Corbett, K.S., Goldsmith, J.A., Hsieh, C.-
L., Abiona, O., et al. (2020). Cryo-EM structure of the 2019-
nCoV spike in the prefusion conformation. Science 367,
1260–1263.
Wright, A.V., Nun
˜ez, J.K., and Doudna, J.A. (2016). Biology
and applications of CRISPR systems: harnessing nature’s
toolbox for genome engineering. Cell 164, 29–44.
Wu, A., Peng, Y., Huang, B., Ding, X., Wang, X., Niu, P., et al.
(2020). Genome composition and divergence of the novel
coronavirus (2019-nCoV) priginating in China. Cell Host
Microbe 27, 325–328.
Wurm, T., Chen, H., Hodgson, T., Britton, P., Brooks, G., and
Hiscox, J.A. (2001). Localization to the nucleolus is a com-
mon feature of coronavirus nucleoproteins, and the protein
may disrupt host cell division. J Virol 75, 9345–9356.
Xu, K., Cai, H., Shen, Y., Ni, Q., Chen, Y., Hu, S., et al. (2020).
Management of corona virus disease-19 (COVID-19): the
Zhejiang experience. Zhejiang Da Xue Xue Bao Yi Xue Ban
49, 0.
Yan, R., Zhang, Y., Li, Y., Xia, L., Guo, Y., and Zhou, Q.
(2020). Structural basis for the recognition of SARS-CoV-2
by full-length human ACE2. Science 367, 1444–1448.
Yang, N., and Shen, H.-MJIjobs. (2020). Targeting the en-
docytic pathway and autophagy process as a novel therapeutic
strategy in COVID-19. Int J Biol Sci 16, 1724.
Yip, C.C.-Y., Ho, C.-C., Chan, J.F.-W., To, K.K.-W., Chan,
H.S.-Y., Wong, S.C.-Y., et al. (2020). Development of a
novel, genome subtraction-derived, SARS-CoV-2-specific
COVID-19-nsp2 real-time RT-PCR assay and its evaluation
using clinical specimens. Int J Mol Sci 21, 2574.
Zeng, Z., Deng, F., Shi, K., Ye, G., Wang, G., Fang, L., et al.
(2018). Dimerization of coronavirus nsp9 with diverse modes
enhances its nucleic acid binding affinity. J Virol 92,
e00692–18.
Zhai, Y., Sun, F., Li, X., Pang, H., Xu, X., Bartlam, M., et al.
(2005). Insights into SARS-CoV transcription and replication
from the structure of the nsp7–nsp8 hexadecamer. Nat Struct
Mol Biol 12, 980–986.
Zhao, S., Lin, Q., Ran, J., Musa, S.S., Yang, G., Wang, W.,
et al. (2020). Preliminary estimation of the basic reproduction
number of novel coronavirus (2019-nCoV) in China, from
2019 to 2020: a data-driven analysis in the early phase of the
outbreak. Int J Infect Dis 92, 214–217.
Zhou, Y., Vedantham, P., Lu, K., Agudelo, J., Carrion, Jr., R.,
Nunneley, J.W., et al. (2015). Protease inhibitors targeting
coronavirus and filovirus entry. Antiviral Res 116, 76–84.
Zhu, X., Fang, L., Wang, D., Yang, Y., Chen, J., Ye, X., et al.
(2017a). Porcine deltacoronavirus nsp5 inhibits interferon-b
production through the cleavage of NEMO. Virology 502,
33–38.
Zhu, X., Wang, D., Zhou, J., Pan, T., Chen, J., Yang, Y., et al.
(2017b). Porcine deltacoronavirus nsp5 antagonizes type I
interferon signaling by cleaving STAT2. J Virol 91,
e00003–17.
Ziebuhr, J., Snijder, E.J., and Gorbalenya, A.E. (2000). Virus-
encoded proteinases and proteolytic processing in the Nido-
virales. J Gen Virol 81, 853–879.
Address correspondence to:
Negin Parsamanesh, PhD
Zanjan Metabolic Diseases Research Center
Zanjan University of Medical Science
Zanjan 4213956184
Iran
E-mail: neginparsa.684@gmail.com;
parsamanesh@zums.ac.ir
Received for publication May 13, 2020; received in revised
form June 24, 2020; accepted July 7, 2020.
THE NOVEL INSIGHT OF SARS-COV-2 MOLECULAR BIOLOGY 13
Downloaded by 217.219.70.5 from www.liebertpub.com at 08/02/20. For personal use only.
... Coronaviridae are single-stranded RNA viruses with a wide envelope. Their genomes range from 25 to 32 kb and have virion diameters of 118-136 nm (Asghari et al., 2020). The spike (S) glycoprotein that stretches 16-21 nm from the virus envelope distinguishes virions, which are approximately spherical. ...
... The receptor-binding domain (RBD), which may attach to angiotensin-converting enzyme 2, is found in S1 (ACE2). S2 aids the entrance of SARS-CoV-2 into the target site (Asghari et al., 2020;Mohamadian et al., 2021). Moreover, with COVID-19 sickness, ACE2 is a biologically relevant receptor (Fani et al., 2020). ...
... In humans, seven CoVs have been found, two CoVs (HCoV-229E and HCoV-NL63) and two CoVs (HCoV-OC43 and HCoV HKU1), which can cause only mild upper respiratory disease, such as the prevalent cold, with a few juvenile patients having acute infection (Sun et al., 2014;Forni et al., 2017).SARS-CoV-2 is the current CoV with a 2% death rate. It is also the third most contagious CoV, which is weaker than SARS-CoV and MERS-CoV (Asghari et al., 2020). The current COVID-19 pandemic, which began in Wuhan and has spread to 188 countries around the world, has caused havoc on civilization, prompting WHO to declare it an international health emergency on January 30, 2020 (Rasmussen et al., 2020). ...
Article
MicroRNAs (miRNAs) are small noncoding RNAs that regulate gene expression through recognition of cognate sequences and interference of transcriptional, translational, or epigenetic processes. Hundreds of miRNA genes have been found in diverse viruses, and many of these are phylogenetically conserved. Respiratory viruses are the most frequent causative agents of disease in humans, with a significant impact on morbidity and mortality worldwide. Recently, the role of miRNAs in respiratory viral gene regulation, as well as host gene regulation during disease progression, has become a field of interest. This review highlighted the importance of various miRNAs and their potential role in fighting with respiratory viruses as therapeutic molecules with a focus on COVID-19.
... The M protein is a type III glycoprotein, the smallest among other structural proteins. According to the studies performed on SARS-CoV and MERS-CoV viruses, the M protein consists of 230 amino acids, and its molecular mass is between 25-35 kDa (Asghari et al. 2020). It has a short ectodomain at the amino end, three transmembrane domains, and a long domain at the carboxyl end of the protein. ...
... This protein plays an important role in the pathogenesis of coronaviruses. SARS-CoV with abolished E protein channel activity was significantly less infectious (Nieto-Torres et al. 2014;Asghari et al. 2020). The analyses confirmed the cation-conducting ability of the SARS-CoV-2 E-protein, which is considered a potential ion channel and an important target for the anti-COVID-19 drug development strategy (Singh Tomar and Arkin 2020). ...
Article
Full-text available
The rapidly spreading Coronavirus Disease 2019 (COVID-19) pandemic has led to a global health crisis and has left a deep mark on society, culture, and the global economy. Despite considerable efforts made to contain the disease, SARS-CoV-2 still poses a threat on a global scale. The current epidemiological situation caused an urgent need to understand the basic mechanisms of the virus transmission and COVID-19 severe course. This review summarizes current knowledge on clinical courses, diagnostics, treatment, and prevention of COVID-19. Moreover, we have included the latest research results on the genetic characterization of SARS-CoV-2 and genetic determinants of susceptibility and severity to infection.
... On the other hand, two amino acids Cys141 and His41 act as catalysts in the active site of the protein. Recent studies have shown that methods can be used to produce drugs resistant to M pro-SARS-CoV-2 [275]. The major protease of SARS-CoV-2 NSP5 inhibits both RIG-I and mitochondrial antiviral signaling (MAVS) protein. ...
Article
Full-text available
Coronavirus disease 2019 (COVID-19) is caused by a new member of the Coronaviridae family known as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). There are structural and non-structural proteins (NSPs) in the genome of this virus. S, M, H, and E proteins are structural proteins, and NSPs include accessory and replicase proteins. The structural and NSP components of SARS-CoV-2 play an important role in its infectivity, and some of them may be important in the pathogenesis of chronic diseases, including cancer, coagulation disorders, neurodegenerative disorders, and cardiovascular diseases. The SARS-CoV-2 proteins interact with targets such as angiotensin-converting enzyme 2 (ACE2) receptor. In addition, SARS-CoV-2 can stimulate pathological intracellular signaling pathways by triggering transcription factor hypoxia-inducible factor-1 (HIF-1), neuropilin-1 (NRP-1), CD147, and Eph receptors, which play important roles in the progression of neurodegenerative diseases like Alzheimer's disease, epilepsy, and multiple sclerosis, and multiple cancers such as glioblastoma, lung malignancies, and leukemias. Several compounds such as polyphenols, doxazosin, baricitinib, and ruxolitinib could inhibit these interactions. It has been demonstrated that the SARS-CoV-2 spike protein has a stronger affinity for human ACE2 than the spike protein of SARS-CoV, leading the current study to hypothesize that the newly produced variant Omicron receptor-binding domain (RBD) binds to human ACE2 more strongly than the primary strain. SARS and Middle East respiratory syndrome (MERS) viruses against structural and NSPs have become resistant to previous vaccines. Therefore, the review of recent studies and the performance of current vaccines and their effects on COVID-19 and related diseases has become a vital need to deal with the current conditions. This review examines the potential role of these SARS-CoV-2 proteins in the initiation of chronic diseases, and it is anticipated that these proteins could serve as components of an effective vaccine or treatment for COVID-19 and related diseases.
... The cause of a serious respiratory disease epidemic that occurred in Wuhan City of China in December 2019 is the new type of coronavirus named as "novel coronavirus-2019" by the World Health Organization (WHO) (1,2). The coronaviruses that can cause disease in humans are as follows: SARS-CoV-2 (severe acute respiratory syndrome coronavirus 2), MERS-CoV, SARS-CoV HCoV-NL63, HCoV-OC43, HCoV-HKU1, and HCoV 229E (3,4). Intense symptoms associated with SARS-CoV-2 infection are shortness of breath, cough, fatigue, fever and muscle aches. ...
Article
Full-text available
Objective: The distinction between COVID-19 and other respiratory infections can be difficult during the flu and winter seasons. The aim of this study is to detect bacterial/viral microorganisms in nasopharyngeal swab samples and to evaluate routine laboratory results of patients with PCR (-) but suspected covid 19. Methods: Between 1 July 2021 and 31 December 2021, 78 patients who were hospitalized and followed up in the suspected Covid service were included in the study. The patients were divided into two groups as those with and without growth on the respiratory panel. Laboratory, demographic and radiological data were compared between groups. Results: C-reactive protein (CRP) and ferritin levels were found to be statistically significantly higher in the group with growth on the respiratory panel compared to the group without growth (p= .05, p= .041, respectively). Reproduction was detected in nasopharyngeal swab samples taken in 56.4% of the patients. More than half of the patients were radiologically defined as CO-RADS 3. Conclusion: It should not be forgotten that other respiratory viral and bacterial infections that mimic the COVID-19 clinic are also commonly observed during this period.
... SARS-CoV-2 belongs to the betacoronavirus and is closely related to the SARS-CoV branch [6]. It is mainly composed of spike protein, envelope protein, membrane protein, nucleocapsid protein, and nonstructural proteins [7,8]. All proteins could be used as antigens for vaccine development or targets for antiviral drugs therapy. ...
Article
Alginate derivatives have been demonstrated remarkable antiviral activities. Here we firstly identified polymannuronate phosphate (PMP) as a highly potential anti-SARS-CoV-2 agent. The structure-activity relationship showed polymannuronate monophosphate (PMPD, Mw: 5.8 kDa, P%: 8.7 %) was the most effective component to block the interaction of spike to ACE2 with an IC50 of 85.5 nM. Surface plasmon resonance study indicated that PMPD could bind to spike receptor binding domain (RBD) with the KD value of 78.59 nM. Molecular docking further suggested that the probable binding site of PMPD to spike RBD protein is the interaction interface between spike and ACE2. PMPD has the potential to inhibit the SARS-CoV-2 infection in an independent manner of heparan sulfate proteoglycans. In addition, polyguluronate sulfate (PGS) and propylene glycol alginate sodium sulfate (PSS) unexpectedly showed 3CLpro inhibition with an IC50 of 1.20 μM and 1.42 μM respectively. The polyguluronate backbone and sulfate group played pivotal roles in the 3CLpro inhibition. Overall, this study revealed the potential of PMPD as a novel agent against SARS-CoV-2. It also provided a theoretical basis for further study on the role of PGS and PSS as 3CLpro inhibitors.
... Other studies confirmed that, supplementing with a combination of L-cysteine and VD or glutathione precursor, rather than just VD, is helpful and aids in the effective implementation of VD administration [54]. Moreover, combining vitamin D and LC supplementation may be more beneficial in reducing the risk of oxidative stress and adverse effect related with type 2 diabetes or COVID-19 infection [55,56,57]. This attitude be able to reduce cellular damage in systemic inflammatory disease, such as hepatitis, obesity, diabetes, and hypertension caused by cytokine storm. ...
Article
Full-text available
According to the World Health Organization (WHO) report, viral hepatitis has been a problem in human society. Vitamins play a significant role in preventing the hepatocarcinoma and liver cirrhosis. In this report, we will first focus on the vitamin D function in the immune system reactions, and then investigate its role in the viral infections and the signaling pathway of hepatitis B virus. The existence of the cytochrome P450 (CYP) 27B1 enzyme, which is involved in vitamin D synthesis in immune system cells, has drawn researchers ' attention to the field of immune system. Toll like receptor (TLR) play a significant role in the immune system, and are one of the primary receptors of the innate immune system. In addition, the synthesis of inflammatory cytokines, such as Interferon γ (IFNγ) and Interleukin-2 (IL-2) is one of the key roles of T helper type 1 (Th1) cells; these cells can suppress two cited cytokines via vitamin D. In the chronic phase of hepatitis B, Cytotoxic T lymphocytes (CTLs) cells have weaker performance than the acute phase of the disease. The association between vitamin D physiologies with viral infections is also confirmed by genetic studies, carried out on genetic variations of vitamin D receptor (VDR) R-encoding disease susceptibility gene. Vitamin D affects different phases of the disease. Therefore, further experiments in this area are proposed.
... These vaccines are either whole inactivated viruses or have overlapping targets and origins which have been quite effective in mitigating the pandemic. SARS-CoV-2 has a genome encoding 28 proteins which play important roles in different stages of viral pathogenesis [8][9][10][11]. The mRNA (Pfizer, Moderna), adenoviral vector-based (AstraZeneca, Janssen) and inactivated (Sinovac, Sinopharm) vaccines use epitopes from the spike protein to generate an immunogenic response in the body, thus creating an immunogenic memory. However, since spike protein is very much prone to acquiring new mutations [12][13][14], these vaccines could lose their efficacies with the evolving viral genome (Center for Disease Control and Prevention). ...
Article
Full-text available
COVID-19 is caused by SARS-CoV-2 which has so far affected more than 500 million people worldwide and killed over 6 million as of 1 May 2022. The approved emergency-use vaccines were lifesaving in such a devastating pandemic. Inflammation-related pathways have been well documented to be upregulated in the case of SARS-CoV-2 in rodents, non-human primates and human samples. We reanalysed a previously published dataset to understand if certain molecular components of inflammation could be higher in infected samples. Mechanistically, viroporins are important players in the life cycle of SARS-CoV-2 and are primary to its pathogenesis. We studied the two prominent viroporins of SARS-CoV-2 (i) Orf3a and (ii) envelope (E) protein from a sequence and structural point of view. Orf3a is a cation-selective viral ion channel which has been shown to disrupt the endosomal pathways. E protein is one of the most conserved proteins among the SARS-CoV proteome which affects the ERGIC-related pathways. The aqueous medium through the viroporins mediates the non-selective translocation of cations, affecting ionic homeostasis in the host cellular compartments. We hypothesize a possible mechanistic approach whereby the ionic imbalance caused by viroporin action could potentially be one of the major pathogenic drivers leading to the increased inflammatory response in the host cell. Our results shed light into the transcriptomic, genomic and structural proteomics aspects of widely studied SARS-CoV-2 viroporins, which can be potentially leveraged for the development of antiviral therapeutics.
Chapter
Full-text available
Article
Severe acute respiratory syndrome coronavirus (SARS-CoV) is a single positive-strand RNA virus that is responsible for the current pandemic that the world has been facing since 2019. The primary route of transmission of SARS-CoV-2 is through respiratory tract transmission. However, other transmission routes such as fecal-oral, vertical transmission, and aerosol-eye also exist. In addition, it has been found that the pathogenesis of this virus involves the binding of the virus's S protein to its host cell surface receptor angiotensin-converting enzyme 2, which results in the subsequent membrane fusion that is required for SARS-CoV-2 to replicate and complete its entire life. The clinical symptoms of patients infected with SARS-CoV-2 can range from asymptomatic to severe. The most common symptoms seen include fever, dry cough, and fatigue. Once these symptoms are observed, a nucleic acid test is done using reverse transcription-polymerase chain reaction. This currently serves as the main confirmatory tool for COVID-19. Despite the fact that no cure has been found for SARS-CoV-2, prevention methods such as vaccines, specific facial mask, and social distancing have proven to be quite effective. It is imperative to have a complete understanding of the transmission and pathogenesis of this virus. To effectively develop new drugs as well as diagnostic tools, more knowledge about this virus would be needed.
Article
Full-text available
The human Betacoronavirus SARS-CoV-2 is a novel pathogen claiming millions of lives and causing a global pandemic that has disrupted international healthcare systems, economies, and communities. The virus is fast mutating and presenting more infectious but less lethal versions. Currently, some small-molecule therapeutics have received FDA emergency use authorization for the treatment of COVID-19, including Lagevrio (molnupiravir) and Paxlovid (nirmaltrevir/ritonavir), which target the RNA-dependent RNA polymerase and the 3CLpro main protease, respectively. Proteins downstream in the viral replication process, specifically the nonstructural proteins (Nsps1-16), are potential drug targets due to their crucial functions. Of these Nsps, Nsp4 is a particularly promising drug target due to its involvement in the SARS-CoV viral replication and double-membrane vesicle formation (mediated via interaction with Nsp3). Given the degree of sequence conservation of these two Nsps across the Betacoronavirus clade, their protein-protein interactions and functions are likely to be conserved as well in SARS-CoV-2. Through AlphaFold2 and its recent advancements, protein structures were generated of Nsp3 and 4 lumenal loops of interest. Then, using a combination of molecular docking suites and an existing library of lead-like compounds, we virtually screened 7 million ligands to identify five putative ligand inhibitors of Nsp4, which could present an alternative pharmaceutical approach against SARS-CoV-2. These ligands exhibit promising lead-like properties (ideal molecular weight and log P profiles), maintain fixed-Nsp4-ligand complexes in molecular dynamics (MD) simulations, and tightly associate with Nsp4 via hydrophobic interactions. Additionally, alternative peptide inhibitors based on Nsp3 were designed and shown in MD simulations to provide a highly stable binding to the Nsp4 protein. Finally, these therapeutics were attached to dendrimer structures to promote their multivalent binding with Nsp4, especially its large flexible luminal loop (Nsp4LLL). The therapeutics tested in this study represent many different approaches for targeting large flexible protein structures, especially those localized to the ER. This study is the first work targeting the membrane rearrangement system of viruses and will serve as a potential avenue for treating viruses with similar replicative function.
Article
Full-text available
The spreading of new viruses is known to provoke global human health threat. The current COVID-19 pandemic caused by the recently emerged coronavirus SARS-CoV-2 is one significant and unfortunate example of what the world will have to face in the future with emerging viruses in absence of appropriate treatment. The discovery of potent and specific antiviral inhibitors and/or vaccines to fight these massive outbreaks is an urgent research priority. Enzymes involved in the capping pathway of viruses and more specifically RNA N7- or 2’O-methyltransferases (MTases) are now admitted as potential targets for antiviral chemotherapy. We designed bisubstrate inhibitors by mimicking the transition state of the 2’-O-methylation of the cap RNA in order to block viral 2’-O MTases. This work resulted in the synthesis of 16 adenine dinucleosides with both adenosines connected by various nitrogen-containing linkers. Unexpectedly, all the bisubstrate compounds were barely active against 2’-O MTases of several flaviviruses or SARS-CoV but surprisingly, seven of them showed efficient and specific inhibition against SARS-CoV N7-MTase (nsp14) in the micromolar to submicromolar range. The most active nsp14 inhibitor identified is as potent as but particularly more specific than the broad-spectrum MTase inhibitor, sinefungin. Molecular docking suggests that the inhibitor binds to a pocket formed by the S-adenosyl methionine (SAM) and cap RNA binding sites, conserved among SARS-CoV nsp14. These dinucleoside SAM analogs will serve as starting points for the development of next inhibitors for SARS-CoV-2 nsp14 N7-MTase.
Article
Full-text available
The ongoing outbreak of Coronavirus Disease 2019 (COVID-19) has become a global public health emergency. SARS-coronavirus-2 (SARS-CoV-2), the causative pathogen of COVID-19, is a positive-sense single-stranded RNA virus belonging to the family Coronaviridae. For RNA viruses, virus-encoded RNA helicases have long been recognized to play pivotal roles during viral life cycles by facilitating the correct folding and replication of viral RNAs. Here, our studies show that SARS-CoV-2-encoded nonstructural protein 13 (nsp13) possesses the nucleoside triphosphate hydrolase (NTPase) and RNA helicase activities that can hydrolyze all types of NTPs and unwind RNA helices dependently of the presence of NTP, and further characterize the biochemical characteristics of these two enzymatic activities associated with SARS-CoV-2 nsp13. Moreover, we found that some bismuth salts could effectively inhibit both the NTPase and RNA helicase activities of SARS-CoV-2 nsp13 in a dose-dependent manner. Thus, our findings demonstrate the NTPase and helicase activities of SARS-CoV-2 nsp13, which may play an important role in SARS-CoV-2 replication and serve as a target for antivirals.
Article
Full-text available
Coronaviridae is a family of single‐stranded positive enveloped RNA viruses. This article aimed to review the history of these viruses in the last 60 years since their discovery to understand what lessons can be learned from the past. A review of the PubMed database was carried out, describing taxonomy, classification, virology, genetic recombination, host adaptation, and main symptoms related to each type of virus. SARS‐CoV‐2 is responsible for the ongoing global pandemic, SARS‐CoV and MERS‐CoV were responsible for causing severe respiratory illness and regional epidemics in the past while the four other strains of CoVs (229‐E OC43, NL63, and HKU1) circulate worldwide and normally only cause mild upper respiratory tract infections. Given the enormous diversity of coronavirus viruses in wildlife and their continuous evolution and adaptation to humans, future outbreaks would undoubtedly occur. Restricting or banning all trade in wild animals in wet markets would be a necessary measure to reduce future zoonotic infections.
Preprint
Full-text available
RNA helicases play pivotal role in RNA replication by catalysing the unwinding of complex RNA duplex structures into single strands in ATP/NTP dependent manner. SARS coronavirus 2 (SARS-CoV-2) is a single stranded positive sense RNA virus belonging to the family Coronaviridae. The viral RNA encodes non structural protein Nsp13 or the viral helicase protein that helps the viral RNA dependent RNA polymerase (RdRp) to execute RNA replication by unwinding the RNA duplexes. In this study we identified a novel mutation at position 541of the helicase where the tyrosine (Y) got substituted with cytosine (C). We found that Y541C is a destabilizing mutation increasing the molecular flexibility and leading to decreased affinity of helicase binding with RNA. Earlier we had reported a mutation P504L in the helicase protein for which had not performed RNA binding study. Here we report that P504L mutation leads to increased affinity of helicase RNA interaction. So, both these mutations have opposite effects on RNA binding. Moreover, we found a significant fraction of isolate population where both P504L and Y541C mutations were co-existing.
Article
Full-text available
Coronaviruses (CoVs) are positive-strand RNA viruses with the largest genome among all RNA viruses. They are able to infect many host, such as mammals or birds. Whereas CoVs were identified 1930s, they became known again in 2003 as the agents of the Severe Acute Respiratory Syndrome (SARS). The spike protein is thought to be essential in the process of CoVs entry, because it is associated with the binding to the receptor on the host cell. It is also involved in cell tropism and pathogenesis. Receptor recognition is the crucial step in the infection. CoVs are able to bind a variety of receptors, although the selection of receptor remains unclear. Coronaviruses were initially believed to enter cells by fusion with the plasma membrane. Further studies demonstrated that many of them involve endocytosis through clathrin-dependent, caveolae-dependent, clathrin-independent, as well as caveolae-independent mechanisms. The aim of this review is to summarise current knowledge about coronaviruses, focussing especially on CoVs entry into the host cell. Advances in understanding coronaviruses replication strategy and the functioning of the replicative structures are also highlighted. The development of host-directed antiviral therapy seems to be a promising way to treat infections with SARS-CoV or other pathogenic coronaviruses. There is still much to be discovered in the inventory of pro- and anti-viral host factors relevant for CoVs replication. The latest pandemic danger, originating from China, has given our previously prepared work even more of topicality.
Article
Full-text available
Significance A key to curbing SARS-CoV-2 is to understand how it enters cells. SARS-CoV-2 and SARS-CoV both use human ACE2 as entry receptor and human proteases as entry activators. Using biochemical and pseudovirus entry assays and SARS-CoV as a comparison, we have identified key cell entry mechanisms of SARS-CoV-2 that potentially contribute to the immune evasion, cell infectivity, and wide spread of the virus. This study also clarifies conflicting reports from recent studies on cell entry of SARS-CoV-2. Finally, by highlighting the potency and the evasiveness of SARS-CoV-2, the study provides insight into intervention strategies that target its cell entry mechanisms.
Article
Full-text available
The recently emerged SARS-CoV-2 caused a major outbreak of coronavirus disease 2019 (COVID-19) and instigated a widespread fear, threatening global health security. To date, no licensed antiviral drugs or vaccines are available against COVID-19 although several clinical trials are underway to test possible therapies. During this urgency situation, computational drug discovery methods provide an alternative to tiresome high-throughput screening, particularly in the hit-to-lead-optimization stage. Identification of small molecules that specifically target viral replication apparatus has indicated highest potential towards antiviral drug discovery. In this work, we present potential compounds that specifically target SARS-CoV-2 vital proteins, including the main protease, Nsp12 RNA polymerase and Nsp13 helicase. An integrative virtual screening and molecular dynamics simulations approach led to the identification of potential binding modes and favourable molecular interaction profile of corresponding compounds. Moreover, the identification of structurally important binding site residues in conserved motifs located inside the active site highlights relative importance of ligand binding based on residual energy decomposition analysis. Although the current study lacks experimental validation, the structural information obtained from this computational study has paved way for the design of targeted inhibitors to combat COVID-19 outbreak.
Article
We report the crystal structure of the SARS-CoV-2 putative primase composed of the nsp7 and nsp8 proteins. We observed a dimer of dimers (2:2 nsp7-nsp8) in the crystallographic asymmetric unit. The structure revealed a fold with a helical core of the heterotetramer formed by both nsp7 and nsp8 that is flanked with two symmetry-related nsp8 β-sheet subdomains. It was also revealed that two hydrophobic interfaces one of approx. 1340 Ų connects the nsp7 to nsp8 and a second one of approx. 950 Ų connects the dimers and form the observed heterotetramer. Interestingly, analysis of the surface electrostatic potential revealed a putative RNA binding site that is formed only within the heterotetramer.