scispace - formally typeset
Search or ask a question
Posted ContentDOI

Type-I interferon signatures in SARS-CoV-2 infected Huh7 cells

TL;DR: In this paper, the authors used a proteomics-based approach to identify that SARS-CoV-2 infection induces delayed and dysregulated Type-I interferon (IFN-I) signaling in Huh7 cells.
Abstract: Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) that causes Coronavirus disease 2019 (COVID-19) has caused a global health emergency A key feature of COVID-19 is dysregulated interferon-response Type-I interferon (IFN-I) is one of the earliest antiviral innate immune responses following viral infection and plays a significant role in the pathogenesis of SARS-CoV-2 In this study, using a proteomics-based approach, we identified that SARS-CoV-2 infection induces delayed and dysregulated IFN-I signaling in Huh7 cells We demonstrate that SARS-CoV-2 is able to inhibit RIG-I mediated IFN-{beta} production Our results also confirm the recent findings that IFN-I pretreatment is able to reduce susceptibility of Huh7 cells to SARS-CoV-2, but not post-treatment Moreover, senescent Huh7 cells, in spite of showing accentuated IFN-I response were more susceptible to SARS-CoV-2 infection, and the virus effectively inhibited IFIT1 in these cells Finally, proteomic comparison between SARS-CoV-2, SARS-CoV and MERS-CoV revealed a distinct differential regulatory signature of interferon-related proteins emphasizing that therapeutic strategies based on observations in SARS-CoV and MERS-CoV should be used with caution Our findings provide a better understanding of SARS-CoV-2 regulation of cellular interferon response and a perspective on its use as a treatment Investigation of different interferon stimulated genes and their role in inhibition of SARS-CoV-2 pathogenesis may direct novel antiviral strategies

Content maybe subject to copyright    Report

Chen et al. Cell Death Discovery (2021) 7:114
https://doi.org/10.1038/s41420-021-00487-z
Cell Death Discovery
ARTICLE Open Access
Type-I interferon signatures in SARS-CoV-2 infected
Huh7 cells
Xi Chen
1
, Elisa Saccon
1
,K.Soa Appelberg
2
, Flora Mikaeloff
1
, Jimmy Esneider Rodriguez
3
, Beatriz Vinhas
1
,
Teresa Frisan
4
, Ákos Végvári
3
,AliMirazimi
1,2
, Ujjwal Neogi
1,5
and Soham Gupta
1
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) that causes Coronavirus disease 2019 (COVID-19) has
caused a global health emergency. A key feature of COVID-19 is dysregulated interferon-response. Type-I interferon
(IFN-I) is one of the earliest antiviral innate immune responses following viral infection and plays a signicant role in
the pathogenesis of SARS-CoV-2. In this study, using a proteomics-based approach, we identi ed that SARS-CoV-2
infection induces delayed and dysregulated IFN-I signaling in Huh7 cells. We demonstrate that SARS-CoV-2 is able to
inhibit RIG-I mediated IFN-β production. Our results also conrm the recent ndings that IFN-I pretreatment is able to
reduce the susceptibility of Huh7 cells to SARS-CoV-2, but not post-treatment. Moreover , senescent Huh7 cells, in spit e
of showing accentuated IFN-I response were more susceptible to SARS-CoV-2 infection, and the virus effectively
inhibited IFIT1 in thes e cells. Finally, proteomic comparis on between SARS-CoV-2, SARS-CoV, and MERS-CoV revealed a
distinct differential regulatory signature of interferon-related proteins emphasizing that therapeutic strategies based
on observations in SARS-CoV and MERS-CoV should be used with caution. Our ndings provide a better understanding
of SARS-CoV-2 regulation of cellular interferon response and a perspective on its use as a treatment. Investigation of
different interferon-stimulated genes and their role in the inhibition of SARS-CoV-2 pathogenesis may direct novel
antiviral strategies.
Introduction
The novel severe acute respiratory syndrome cor-
onavirus 2 (SARS-CoV-2) caused a major ongoing pan-
demic with more than a million deaths worldwide by the
end of 2020
1
. SARS-CoV-2 shares similar clinical features
to two other well-known coronavirus infections, namely
SARS-CoV and MERS-CoV, but it presents a lower case
fatality compared to them
2,3
. However, the most severe
forms of coronavirus diseases are often associated with a
dysregulated type-I interferon (IFN-I) response
4
.
IFN-I response, majorly IFN-α and IFN-β, is one of the
rst lines of defense against viruses
5
. The early activation
of IFN responses against coronaviruses is initiated by
recognition of viral products by the host pattern recog-
nition receptors like Toll-like receptors (TLRs) and RIG-
I-like receptors (RLRs). RLRs can recognize the viral RNA
that promotes their oligomerization and subsequent
activation of a signaling cascade leading to the production
of IFNα and IFNβ
6
. Through autocrine and paracrine
signaling, the secreted IFN binds to IFN-α/β membrane
receptors, activating the JAK-STAT signaling cascade that
triggers the transcription of several interferon-stimulated
genes (ISGs) with diverse antiviral properties
7
. Cor-
onaviruses have evolved mechanisms to evade the host s
antiviral immune response. Several structural and non-
structural proteins in SARS-CoV
8
, in MERS-CoV
8,9
, and
in SARS-CoV-2
10,11
have been shown to be strong IFN-
antagonists. The dynamics of the IFN response varies
between these three coronaviruses
1214
. Distinct virus-
specic patterns in host cell response were also noted in
transcriptomics analysis
15
. Thus, a deeper understanding
of the SARS-CoV-2 mediated regulation of IFN response
© The Author(s) 2021
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction
in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a linktotheCreativeCommons license, and indicate if
changes were made. The images or other third party material in this article are included in the articles Creative Commons license, unless indicated otherwise in a credit line to the material. If
material is not included in the articles Creative Commons license and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain
permission directly from the copyright holder. To view a copy of this license, visit http://creativecommons.org/licenses/by/4.0/.
Correspondence: Soham Gupta (soham.gupta@ki.se)
1
Division of Clinical Microbiology, Department of Laboratory Medicine,
Karolinska Institutet, ANA Futura, Campus Flemingsberg, Stockholm, Sweden
2
Public Health Agency of Sweden, Solna, Sweden
Full list of author information is available at the end of the article
These authors contributed equally: Xi Chen, Elisa Saccon
Edited by Chiara Agrati
Ofcial journal of the Cell Death Differentiation Association
1234567890():,;
1234567890():,;
1234567890():,;
1234567890():,;

is necessary to develop rationale and novel therapeutic
approaches for SARS-CoV-2.
In this study, we characterized the SARS-CoV-2 medi-
ated dysregulation of IFN-signaling in Huh7 infected cells
using quantitative proteomics. We show a delayed acti-
vation of IFN-signaling with the ability of the virus to
evade RIG-I mediated IFN-signaling during early infec-
tion. In line with recent studies, susceptibility of Huh7
cells to SARS-CoV-2 decreased upon IFN-pretreatment,
but not post-treatment. We also determined the IFN-
signaling response pattern of SARS-CoV and MERS-CoV
infection in Huh7 cells using proteomics and show a
distinction compared to SARS-CoV-2. Together, the
results provide a perspective of immune regulation by
coronaviruses.
Results
Quantitative proteomics and transcriptomics of SARS-CoV-
2 infected Huh7 cells identies dysregulation in IFN-I
signaling pathways
To understand the modulation of IFN responses fol-
lowing SARS-CoV-2 infection, we reused the proteomics
and transcriptomics datasets from our earlier study
16
.We
rst analyzed the quantitative proteomics data on Huh7
cells that were either mock-infected or infected with
SARS-CoV-2 at a multiplicity of infection (MOI) of 1,
over a period of 24 and 48 h post infection (hpi). Genes
associated with the interferon response, including the
IFN-α/β signaling (Pathway:R-HSA-909733), IFN-γ sig-
naling (Pathway:R-HSA-877300), and the antiviral
mechanism by ISGs (Pathway:R-HSA-1169410) were
extracted from the data. For mock-infected, we con-
sidered the data for two replicates as the third one was a
major outlier as shown in the PCA plot (Fig. S1). No
major changes were observed in the ISGs at 24 hpi and
signicant modulation was only observed at 48 hpi after
infection as represented in the heatmap (Fig. 1A). Of the
94 proteins studied, a number of proteins showed a sig-
nicant reduction in abundance (n = 20), while a major
cluster of proteins showed an increase (n = 26) (LIMMA,
false-discovery rate (FDR) < 0.05). The log2fold change of
the signicantly regulated genes is represented as a vol-
cano plot (Fig. 1B). The protein-protein interaction net-
work of the signicantly changed genes showed two
denite clusters: cluster-1 involved proteins associated
with the RIG-I/DDX58 and IFN-I signaling, while cluster-
2 consisted of transporter proteins belonging to the
components of nucleoporin complex and karyopherin
family (Fig. 1C).
We also looked into the IFN-signaling genes in the
transcriptomics dataset and observed no major changes in
the differential expression of the transcripts related to this
pathway except for EIF4A2, STAT2, TRIM10 (upregu-
lated), and FLNA, JAK1, GBP2, MT2A, TRIM26
(downregulated) at 48 hpi (Fig. 2A). Of the genes corre-
sponding to the proteins that were altered in the pathway
(Fig. 2B) only EIF4A2, STAT2, JAK1, GBP2, and FLNA
showed transcript levels correlating with protein expres-
sion (Fig. 2C).
SARS-CoV-2 induces delayed and low-level activation of
RIG-I signaling and inhibits IFN-β in Huh7 cells
In our proteomics data, we observed a delayed activa-
tion of RIG-I and dysregulation of ISGs. RIG-I, a key
cytosolic receptor is responsible for the activation of IFN-
β (Fig. 3A). We next studied the effect of SARS-CoV-2 in
the induction of IFN-β. We did not observe any sig-
nicant changes in the levels of IFN-β specic messenger
RNAs (mRNAs) in SARS-CoV-2 infected Huh7 cells both
at 24 and 48 hpi, with a marginal increase with MOI 0.1 at
48 hpi (Fig. 3B). This effect was concomitant with a
marginal suppression of RIG-I and MDA-5 protein
expression at 24hpi and an observable increase at 48 hpi
detected in western blots probed with specic antibodies
(Fig. 3C, D). The Western blot data were in line with our
proteomics data.
Since we did not observ e any IFN-β induction or RIG-I
activation at 24 hpi, we next investigated whether SARS-
CoV-2 is abl e to inhibit IFN -β activation in Huh7 cells.
To determine this Huh7 cells were either mock-infected
or infected with SARS-CoV- 2 at MOI 0.1, followed by
IFN-β induction by treating with RIG-I agonists, aci-
tretin or polyI:C for 24 h. Treatment with acitretin or
polyI:C post infection did not inhibit the production of
the virus (Fig. 3E, G). SARS-CoV-2 was able to efciently
inhibit the IFN-β production i n the RIG-I activated cells
(Fig. 3F, H).
Effect of SARS-CoV-2 on ISGs
IFN-β can stimulate the expression of several ISGs with
antiviral properties using JAK-STAT signaling pathway
(Fig. 4A). Similar to our transcriptomics data, qPCR
analysis to detect IFIT1, RIG-I (DDX58), and MX2
expression in SARS-CoV-2 infected Huh7 cells did not
show any signicant changes compared to uninfected cells
(Fig. 4B). On contrary, our proteomics data showed an
increase in the protein level of several ISGs, including
ISG15 at 48 hpi. ISG15 can behave as an antiviral cytokine
in its free form and also can conjugate to diverse cellular
and viral proteins and regulate their functions
17,18
. The
mRNA levels of ISG15 in SARS-CoV-2 infected Huh7
cells at 24 and 48 hpi did not change signicantly (Fig.
4C). However, at the protein level, it was interesting to
note an observable decrease in the conjugated ISG15 at 24
hpi and a marked increase in host-protein ISGylation at
48 hpi (Fig. 4D, E) in a dose-dependent manner, sug-
gesting the virus can modulate protein ISGylation to alter
the cellular environment
11
.
Chen et al. Cell Death Discovery (2021) 7:114 Page 2 of 15
Ofcial journal of the Cell Death Differentiation Association

Fig. 1 SARS-CoV-2 induced a delayed and dysregulated IFN signaling response identied in proteomics data. A Heatmap of IFN-stimulated
proteins before infection and at 24 and 48 hpi. Data were quantile normalized and Z-score transformed. Lower values are represented in yellow and
higher values in purple. Signicant differentially expressed proteins between time points are indicated in blue if downregulated and in red if
upregulated. B Volcano plots of proteins with differential abundance between Mock and Huh7 cell 48 h after SARS-CoV-2 infection. Upregulated
proteins are represented in red while proteins downregulated are represented in green. FDR < 0.05. C Cytoscape network of differentially abundant
IFN-stimulated proteins. Proteins are represented as circles. Gradient color was applied on proteins depending on fold change (low = green to high
= red). The size of the circle is proportional to the fold change.
Chen et al. Cell Death Discovery (2021) 7:114 Page 3 of 15
Ofcial journal of the Cell Death Differentiation Association

Fig. 2 (See legend on next page.)
Chen et al. Cell Death Discovery (2021) 7:114 Page 4 of 15
Ofcial journal of the Cell Death Differentiation Association

SARS-CoV-2 is inhibited by IFN pretreatment
ISGs can also be stimulated in experimental models by
external treatment with IFNs. In order to evaluate the
susceptibility of SARS-CoV-2 to IFN-I, we either pre-
sensitized cells (16 h) with IFN-α2a (5000 IU) and IFN-β
(100 IU) or treated the cells with the same concentrations
of IFNs starting 1 hpi and continued for 24 h. Huh7 cells
were infected with SARS-CoV-2 at MOI 0.1 and at 24 hpi
the supernatant was collected to determine the virus
production in the presence or absence of different IFN-I
treatments. As shown in Fig. 4F, IFN pre-sensitization
lead to a signicant reduction in SARS-CoV-2 production
in the supernatant as compared to levels in supernatant
from untreated cells at 24 hpi. However, IFN-I treatment
after infection did not suppress virus production (Fig. 4H).
This observation suggests rstly that the presence of a
high level of IFN-response can suppress the incoming
virus and secondly that the virus has also developed
measures to counteract these responses when it has
already established infection. Then, we further looked into
the effect of IFN-I treatment and infection in transcrip-
tional activation of a few of the ISGs that were modulated
by SARS-CoV-2 infection. For this, we selected MX2,
IFIT1, and ISG15. While SARS-CoV-2 suppressed MX2
mRNA in untreated cells, MX2 did not show any activa-
tion following IFN-treatment (not shown). Both ISG15
and IFIT1 were signicantly induced following IFN-I
treatment, however, SARS-CoV-2 did not cause any sig-
nicant alterations to the mRNA levels (Fig. 4G, I).
Senescent Huh7 cells stimulate IFN-I response but
promote virus infectivity
Elderly people are more vulnerable to SARS-CoV-2
infection
19
and cellular senescence is postulated as a
factor for increased infection. Cellular senescence has
been observed to play a different role in either promoting
infection for some viruses or inhibiting infection for
others. To this end, we aimed to examine the suscept-
ibility of senescent Huh7 cells to SARS-CoV-2 and asso-
ciated IFN-I response. To induce cellular senescence
Huh7 cells were treated with 0.5 μM of etoposide for
6 days followed by 2 days without any treatment and then
infected with SARS-CoV-2 for 1 h and cells and
supernatants were harvested 24 hpi. Etoposide treatment
resulted in massive cell death and surviving cells were
large in size. Cellular senescence was determined by
detecting p21 mRNA levels (Fig. 5B). Senescent Huh7
cells showed a signicant increase in SARS-CoV-2 pro-
duction in cell supernatant of senescent Huh7 cells
compared to the untreated control cells (Fig. 5A). We
next investigated the IFN-response in senescence-induced
and non-induced cells by detecting mRNA transcripts of
IFN-β and ISGs such as ISG15, IFIT1, MX2, and RIG-I.
Cellular senescence induced an increase in the IFN-
response with a signicant increase in the levels of IFN-β
and other ISGs tested (Fig. 5B). SARS-CoV-2 failed to
signicantly alter the levels of any tested genes except for
IFIT1, where a signicant decrease in the mRNA levels
was noted upon infection (Fig. 5B). To determine if the
enhanced infectivity of senescent cells is specic to Huh7,
we tried to replicate the same experiment in Caco2 cells.
However, Caco2 cells were more resistant to 0.5 μM
etoposide treatment and did not show observable induc-
tion of senescence determined by qPCR of the p21 gene
(Fig. S2B). Most interestingly, in contrast to Huh7, even a
very low-level induction of p21 was sufcient to sig-
nicantly reduce SARS-CoV-2 susceptibility (Fig. S2A)
and among the ISGs IFIT1 showed an observable increase
upon infection (Fig. S2B). The results suggest that there is
a cell-type-specic regulation of SARS-CoV-2 and the
importance of IFIT1 as an anti-SARS-CoV-2 ISGs.
Global proteomic response to SARS-CoV-2 relative to
SARS-CoV and MERS-CoV in Huh7 cells
To explore the diff erences in pathogenicity of SARS-
CoV-2 in comparison with its predecessor human
pathogenic coronaviruses, we inf ected Huh7 cells with
SARS-CoV and MERS-CoV at MOI 1 and measured the
global proteomic changes by performing quantitative
proteomics. MERS-CoV was observed to be highly cyto-
pathic and by 48 hpi all the cells were dead restricting our
analysis t o 24 hpi, while SARS-CoV showed a slower
cytopathogenicity, and infected cells were collected both
at 24 and 48 hpi. Quanti tative proteomics was performed
utilizing a TMT-labeling strategy of mock-infected and
infected cells in triplicate as previously described by us
16
.
(see gure on previous page)
Fig. 2 SARS-CoV-2 induced transcriptional changes in the IFN-signaling genes in transcriptomics data. A Heatmap of IFN-stimulated
transcripts before infection and at 24 and 48 hpi. Data were log2 normalized and Z-score transformed. Lower values are represented in yellow and
higher values in purple. Signicant differentially expressed genes between time points are indicated in blue if downregulated and in red if
upregulated. B The scheme graph of the type I interferon signaling pathways created with BioRender, in which the regulated genes expression level
trend is noted. The signicantly changed proteins observed in the proteomics data are denoted by green arrows or letters (downregulated) or red
arrows or letters (upregulated). C Dot plot for each transcript that was detected as signicantly altered in proteomics. For each gene, the scaled
values in triplicates are represented in mock, 24 and 48 hpi and linked by the light red line, the average value is displayed in red. The name of the
genes is indicated in a colored box based on the proteomics data. The genes corresponding to increased protein levels are in red boxes and to
decreased protein levels in green boxes.
Chen et al. Cell Death Discovery (2021) 7:114 Page 5 of 15
Ofcial journal of the Cell Death Differentiation Association

Citations
More filters
Journal ArticleDOI
20 Apr 2021-iScience
TL;DR: In this article, the authors assessed the tropism and cytopathogenicity of the first Swedish isolate of SARS-CoV-2 in six different human cell lines, compared their growth characteristics, and performed quantitative proteomics for the susceptible cell lines.

45 citations

Posted ContentDOI
27 Mar 2021-bioRxiv
TL;DR: In this paper, the cytosolic RNA sensor MDA5 was identified as a cellular sensor for SARS-CoV-2 infection that induced type I and III interferons (IFNs) and proinflammatory factors such as IL6 and TNF.
Abstract: Human cells respond to infection by SARS-CoV-2, the virus that causes COVID-19, by producing cytokines including type I and III interferons (IFNs) and proinflammatory factors such as IL6 and TNF IFNs can limit SARS-CoV-2 replication but cytokine imbalance contributes to severe COVID-19 We studied how cells detect SARS-CoV-2 infection We report that the cytosolic RNA sensor MDA5 was required for type I and III IFN induction in the lung cancer cell line Calu-3 upon SARS-CoV-2 infection Type I and III IFN induction further required MAVS and IRF3 In contrast, induction of IL6 and TNF was independent of the MDA5-MAVS-IRF3 axis in this setting We further found that SARS-CoV-2 infection inhibited the ability of cells to respond to IFNs In sum, we identified MDA5 as a cellular sensor for SARS-CoV-2 infection that induced type I and III IFNs

3 citations

References
More filters
Journal ArticleDOI
17 Mar 2020-JAMA
TL;DR: The epidemiological and clinical characteristics of novel coronavirus (2019-nCoV)-infected pneumonia in Wuhan, China, and hospital-associated transmission as the presumed mechanism of infection for affected health professionals and hospitalized patients are described.
Abstract: Importance In December 2019, novel coronavirus (2019-nCoV)–infected pneumonia (NCIP) occurred in Wuhan, China. The number of cases has increased rapidly but information on the clinical characteristics of affected patients is limited. Objective To describe the epidemiological and clinical characteristics of NCIP. Design, Setting, and Participants Retrospective, single-center case series of the 138 consecutive hospitalized patients with confirmed NCIP at Zhongnan Hospital of Wuhan University in Wuhan, China, from January 1 to January 28, 2020; final date of follow-up was February 3, 2020. Exposures Documented NCIP. Main Outcomes and Measures Epidemiological, demographic, clinical, laboratory, radiological, and treatment data were collected and analyzed. Outcomes of critically ill patients and noncritically ill patients were compared. Presumed hospital-related transmission was suspected if a cluster of health professionals or hospitalized patients in the same wards became infected and a possible source of infection could be tracked. Results Of 138 hospitalized patients with NCIP, the median age was 56 years (interquartile range, 42-68; range, 22-92 years) and 75 (54.3%) were men. Hospital-associated transmission was suspected as the presumed mechanism of infection for affected health professionals (40 [29%]) and hospitalized patients (17 [12.3%]). Common symptoms included fever (136 [98.6%]), fatigue (96 [69.6%]), and dry cough (82 [59.4%]). Lymphopenia (lymphocyte count, 0.8 × 109/L [interquartile range {IQR}, 0.6-1.1]) occurred in 97 patients (70.3%), prolonged prothrombin time (13.0 seconds [IQR, 12.3-13.7]) in 80 patients (58%), and elevated lactate dehydrogenase (261 U/L [IQR, 182-403]) in 55 patients (39.9%). Chest computed tomographic scans showed bilateral patchy shadows or ground glass opacity in the lungs of all patients. Most patients received antiviral therapy (oseltamivir, 124 [89.9%]), and many received antibacterial therapy (moxifloxacin, 89 [64.4%]; ceftriaxone, 34 [24.6%]; azithromycin, 25 [18.1%]) and glucocorticoid therapy (62 [44.9%]). Thirty-six patients (26.1%) were transferred to the intensive care unit (ICU) because of complications, including acute respiratory distress syndrome (22 [61.1%]), arrhythmia (16 [44.4%]), and shock (11 [30.6%]). The median time from first symptom to dyspnea was 5.0 days, to hospital admission was 7.0 days, and to ARDS was 8.0 days. Patients treated in the ICU (n = 36), compared with patients not treated in the ICU (n = 102), were older (median age, 66 years vs 51 years), were more likely to have underlying comorbidities (26 [72.2%] vs 38 [37.3%]), and were more likely to have dyspnea (23 [63.9%] vs 20 [19.6%]), and anorexia (24 [66.7%] vs 31 [30.4%]). Of the 36 cases in the ICU, 4 (11.1%) received high-flow oxygen therapy, 15 (41.7%) received noninvasive ventilation, and 17 (47.2%) received invasive ventilation (4 were switched to extracorporeal membrane oxygenation). As of February 3, 47 patients (34.1%) were discharged and 6 died (overall mortality, 4.3%), but the remaining patients are still hospitalized. Among those discharged alive (n = 47), the median hospital stay was 10 days (IQR, 7.0-14.0). Conclusions and Relevance In this single-center case series of 138 hospitalized patients with confirmed NCIP in Wuhan, China, presumed hospital-related transmission of 2019-nCoV was suspected in 41% of patients, 26% of patients received ICU care, and mortality was 4.3%.

16,635 citations

Journal ArticleDOI
TL;DR: A validated diagnostic workflow for 2019-nCoV is presented, its design relying on close genetic relatedness of 2019- nCoV with SARS coronavirus, making use of synthetic nucleic acid technology.
Abstract: Background The ongoing outbreak of the recently emerged novel coronavirus (2019-nCoV) poses a challenge for public health laboratories as virus isolates are unavailable while there is growing evidence that the outbreak is more widespread than initially thought, and international spread through travellers does already occur. Aim We aimed to develop and deploy robust diagnostic methodology for use in public health laboratory settings without having virus material available. Methods Here we present a validated diagnostic workflow for 2019-nCoV, its design relying on close genetic relatedness of 2019-nCoV with SARS coronavirus, making use of synthetic nucleic acid technology. Results The workflow reliably detects 2019-nCoV, and further discriminates 2019-nCoV from SARS-CoV. Through coordination between academic and public laboratories, we confirmed assay exclusivity based on 297 original clinical specimens containing a full spectrum of human respiratory viruses. Control material is made available through European Virus Archive – Global (EVAg), a European Union infrastructure project. Conclusion The present study demonstrates the enormous response capacity achieved through coordination of academic and public laboratories in national and European research networks.

6,229 citations

Journal ArticleDOI
TL;DR: The data show that remdesivir was superior to placebo in shortening the time to recovery in adults who were hospitalized with Covid-19 and had evidence of lower respiratory tract infection.
Abstract: Background Although several therapeutic agents have been evaluated for the treatment of coronavirus disease 2019 (Covid-19), none have yet been shown to be efficacious. Methods We conducte...

5,532 citations

Journal ArticleDOI
David E. Gordon, Gwendolyn M. Jang, Mehdi Bouhaddou, Jiewei Xu, Kirsten Obernier, Kris M. White1, Matthew J. O’Meara2, Veronica V. Rezelj3, Jeffrey Z. Guo, Danielle L. Swaney, Tia A. Tummino4, Ruth Hüttenhain, Robyn M. Kaake, Alicia L. Richards, Beril Tutuncuoglu, Helene Foussard, Jyoti Batra, Kelsey M. Haas, Maya Modak, Minkyu Kim, Paige Haas, Benjamin J. Polacco, Hannes Braberg, Jacqueline M. Fabius, Manon Eckhardt, Margaret Soucheray, Melanie J. Bennett, Merve Cakir, Michael McGregor, Qiongyu Li, Bjoern Meyer3, Ferdinand Roesch3, Thomas Vallet3, Alice Mac Kain3, Lisa Miorin1, Elena Moreno1, Zun Zar Chi Naing, Yuan Zhou, Shiming Peng4, Ying Shi, Ziyang Zhang, Wenqi Shen, Ilsa T Kirby, James E. Melnyk, John S. Chorba, Kevin Lou, Shizhong Dai, Inigo Barrio-Hernandez5, Danish Memon5, Claudia Hernandez-Armenta5, Jiankun Lyu4, Christopher J.P. Mathy, Tina Perica4, Kala Bharath Pilla4, Sai J. Ganesan4, Daniel J. Saltzberg4, Rakesh Ramachandran4, Xi Liu4, Sara Brin Rosenthal6, Lorenzo Calviello4, Srivats Venkataramanan4, Jose Liboy-Lugo4, Yizhu Lin4, Xi Ping Huang7, Yongfeng Liu7, Stephanie A. Wankowicz, Markus Bohn4, Maliheh Safari4, Fatima S. Ugur, Cassandra Koh3, Nastaran Sadat Savar3, Quang Dinh Tran3, Djoshkun Shengjuler3, Sabrina J. Fletcher3, Michael C. O’Neal, Yiming Cai, Jason C.J. Chang, David J. Broadhurst, Saker Klippsten, Phillip P. Sharp4, Nicole A. Wenzell4, Duygu Kuzuoğlu-Öztürk4, Hao-Yuan Wang4, Raphael Trenker4, Janet M. Young8, Devin A. Cavero9, Devin A. Cavero4, Joseph Hiatt9, Joseph Hiatt4, Theodore L. Roth, Ujjwal Rathore9, Ujjwal Rathore4, Advait Subramanian4, Julia Noack4, Mathieu Hubert3, Robert M. Stroud4, Alan D. Frankel4, Oren S. Rosenberg, Kliment A. Verba4, David A. Agard4, Melanie Ott, Michael Emerman8, Natalia Jura, Mark von Zastrow, Eric Verdin10, Eric Verdin4, Alan Ashworth4, Olivier Schwartz3, Christophe d'Enfert3, Shaeri Mukherjee4, Matthew P. Jacobson4, Harmit S. Malik8, Danica Galonić Fujimori, Trey Ideker6, Charles S. Craik, Stephen N. Floor4, James S. Fraser4, John D. Gross4, Andrej Sali, Bryan L. Roth7, Davide Ruggero, Jack Taunton4, Tanja Kortemme, Pedro Beltrao5, Marco Vignuzzi3, Adolfo García-Sastre, Kevan M. Shokat, Brian K. Shoichet4, Nevan J. Krogan 
30 Apr 2020-Nature
TL;DR: A human–SARS-CoV-2 protein interaction map highlights cellular processes that are hijacked by the virus and that can be targeted by existing drugs, including inhibitors of mRNA translation and predicted regulators of the sigma receptors.
Abstract: A newly described coronavirus named severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which is the causative agent of coronavirus disease 2019 (COVID-19), has infected over 2.3 million people, led to the death of more than 160,000 individuals and caused worldwide social and economic disruption1,2. There are no antiviral drugs with proven clinical efficacy for the treatment of COVID-19, nor are there any vaccines that prevent infection with SARS-CoV-2, and efforts to develop drugs and vaccines are hampered by the limited knowledge of the molecular details of how SARS-CoV-2 infects cells. Here we cloned, tagged and expressed 26 of the 29 SARS-CoV-2 proteins in human cells and identified the human proteins that physically associated with each of the SARS-CoV-2 proteins using affinity-purification mass spectrometry, identifying 332 high-confidence protein–protein interactions between SARS-CoV-2 and human proteins. Among these, we identify 66 druggable human proteins or host factors targeted by 69 compounds (of which, 29 drugs are approved by the US Food and Drug Administration, 12 are in clinical trials and 28 are preclinical compounds). We screened a subset of these in multiple viral assays and found two sets of pharmacological agents that displayed antiviral activity: inhibitors of mRNA translation and predicted regulators of the sigma-1 and sigma-2 receptors. Further studies of these host-factor-targeting agents, including their combination with drugs that directly target viral enzymes, could lead to a therapeutic regimen to treat COVID-19. A human–SARS-CoV-2 protein interaction map highlights cellular processes that are hijacked by the virus and that can be targeted by existing drugs, including inhibitors of mRNA translation and predicted regulators of the sigma receptors.

3,319 citations

Journal ArticleDOI
28 May 2020-Cell
TL;DR: It is proposed that reduced innate antiviral defenses coupled with exuberant inflammatory cytokine production are the defining and driving features of COVID-19.

3,286 citations

Frequently Asked Questions (12)
Q1. What contributions have the authors mentioned in the paper "Type-i interferon signatures in sars-cov-2 infected huh7 cells" ?

Type-I interferon ( IFN-I ) is one of the earliest antiviral innate immune responses following viral infection and plays a significant role in the pathogenesis of SARS-CoV-2. In this study, using a proteomics-based approach, the authors identified that SARS-CoV-2 infection induces delayed and dysregulated IFN-I signaling in Huh7 cells. The authors demonstrate that SARS-CoV-2 is able to inhibit RIG-I mediated IFN-β production. 

ISG15 can behave as an antiviral cytokine in its free form and also can conjugate to diverse cellular and viral proteins and regulate their functions17,18. 

Senescent Huh7 cells stimulate IFN-I response but promote virus infectivity Elderly people are more vulnerable to SARS-CoV-2 infection19 and cellular senescence is postulated as a factor for increased infection. 

RNA viruses can stimulate the IFN-I response that is mediated by the RIG-I/RLR signaling cascade leading to the production and release of IFN-β20. 

while the nuclear transporter complex proteins KPNA1, KPNA2, and RAE1 were suppressed in SARS-CoV-2 infected cells, they were upregulated in MERS-CoV infected cells. 

Since the authors did not observe any IFN-β induction or RIG-I activation at 24 hpi, the authors next investigated whether SARSCoV-2 is able to inhibit IFN-β activation in Huh7 cells. 

The absence of visible detection of ORF1a or 3CL-pro peptides in MERSCoV infected cells further strengthens the role of these viral proteins in regulation of transport of cellular transcription factors to the nucleus. 

20. Gupta, S. et al. 14-3-3 scaffold proteins mediate the inactivation of trim25 and inhibition of the type The authorinterferon response by herpesvirus deconjugases. 

In concordance with earlier studies14,25,34, the authors observed that IFN pre-sensitized cells were more resistant to SARSCoV-2, but IFN-treatment following infection did not alter the susceptibility of the cells. 

Quantitative proteomics was performed utilizing a TMT-labeling strategy of mock-infected and infected cells in triplicate as previously described by us16.(see figure on previous page) Fig. 2 SARS-CoV-2 induced transcriptional changes in the IFN-signaling genes in transcriptomics data. 

it is reasonable to believe that people with naturally high level of IFN might better control the virus during early stages of infection and thus progressing towards better disease outcome and recovery. 

The protein-protein interaction network of the significantly changed genes showed two definite clusters: cluster-1 involved proteins associated with the RIG-I/DDX58 and IFN-I signaling, while cluster2 consisted of transporter proteins belonging to the components of nucleoporin complex and karyopherin family (Fig. 1C).