按钮
按钮
按钮
按钮
按钮
首页    盛普前沿    COVID-19    Characterization of anti-viral immunity in recovered individuals infected by SARS-CoV-2
1
 
 
 
Characterization of anti-viral immunity in recovered individuals infected
by SARS-CoV-2
 
Ling Ni
1, *
, Fang Ye
2, *
, Meng-Li Chen
3, *
, Yu Feng
1
, Yong-Qiang Deng
3
, Hui Zhao
3
, Peng
Wei
1
, Jiwan Ge
5
, Xiaoli Li
1
, Lin Sun
1
, Pengzhi Wang
1
, Peng Liang
4
, Han Guo
6
, Xinquan
Wang
5
, Cheng-Feng Qin
3
, Fang Chen
4,#
, Chen Dong
1,7,#
 
1. Institute for Immunology and School of Medicine, Tsinghua University, Beijing 100084,
China
2. Department of Hematology, Chui Yang Liu Hospital affiliated to Tsinghua University,
Beijing 100022, China
3. Department of Virology, State Key Laboratory of Pathogen and Biosecurity, Institute of
Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing 100071, China
4. Department of Cardiology, Chui Yang Liu Hospital affiliated to Tsinghua University,
Beijing100022, China
5. School of Life Sciences, Tsinghua University, Beijing 100084, China
6. Department of Orthopedics, Chui Yang Liu Hospital affiliated to Tsinghua University,
Beijing 100022, China
7. Beijing Key Lab for Immunological Research on Chronic Diseases, Beijing 100084,
China
 
*These authors contributed equally to this work.
 
#To whom correspondence should be addressed: Chen Dong, chendong@tsinghua.edu.cn;
or Fang Chen, anzhenchenfang@163.com
 
All rights reserved. No reuse allowed without permission.
perpetuity.
preprint (which was not certified by peer review) is the author/funder, who has granted medRxiv a license to display the preprint in
The copyright holder for thisthis version posted March 20, 2020. .https://doi.org/10.1101/2020.03.17.20036640doi: medRxiv preprint
 
 
2
 
Abstract
 
The WHO has declared SARS-CoV-2 outbreak a public health emergency of international
concern. However, to date, there was hardly any study in characterizing the immune
responses, especially adaptive immune responses to SARS-CoV-2 infection. In this study, we
collected blood from COVID-19 patients who have recently become virus-free and therefore
were discharged, and analyzed their SARS-CoV-2-specific antibody and T cell responses. We
observed SARS-CoV-2-specific humoral and cellular immunity in the patients. Both were
detected in newly discharged patients, suggesting both participate in immune-mediated
protection to viral infection. However, follow-up patients (2 weeks post discharge) exhibited
high titers of IgG antibodies, but with low levels of virus-specific T cells, suggesting that they
may enter a quiescent state. Our work has thus provided a basis for further analysis of
protective immunity to SARS-CoV-2, and understanding the pathogenesis of COVID-19,
especially in the severe cases. It has also implications in designing an effective vaccine to
protect and treat SARS-CoV-2 infection.
 
Keywords: SARS-CoV-2, COVID-19 patients, humoral immunity, cellular immunity
 
 
 
 
 
 
 
 
 
 
 
 
All rights reserved. No reuse allowed without permission.
perpetuity.
preprint (which was not certified by peer review) is the author/funder, who has granted medRxiv a license to display the preprint in
The copyright holder for thisthis version posted March 20, 2020. .https://doi.org/10.1101/2020.03.17.20036640doi: medRxiv preprint
 
 
3
 
 
At the end of 2019, patients with Coronavirus Disease 2019 (COVID-19) were identified
in Wuhan, China
1
, infected by a novel coronavirus, now named as severe acute respiratory
syndrome coronavirus 2, SARS-CoV-2. The WHO then declared this outbreak a public health
emergency of international concern
2
. The genome sequence of SARS-CoV-2 bears 96%
3
and 79.5% identity to that of a bat coronavirus and SARS-CoV, respectively
4
. Like SARS-
CoV and MERS-CoV, SARS-CoV-2 belongs to the beta genus Coronavirus in the
Corornaviridae family
5
. Clinically, several papers showed that most COVID-19 patients
developed lymphopenia as well as pneumonia with higher plasma levels of pro-inflammatory
cytokines in severe cases
6-8
, suggesting that the host immune system is involved in the
pathogenesis
9,10
. Patients infected by SARS-CoV or MERS-CoV were previously reported to
have antibody responses
11-14
, but exhibited defective expression of type I and II interferon
(IFN), indicative of poor protective immune responses
15-17
. However, to date, there was hardly
any study in characterizing the immune responses, especially adaptive immune responses to
SARS-CoV-2 infection. Only one COVID-19 patient was shown with nucleocapsid protein
(NP)-specific antibody response, in which IgM peaked at day 9 after disease onset and then
switched to IgG by week 2
3
, suggesting involvement of humoral immunity. SARS-CoV-2-
specific T lymphocyte response was unclear. In this study, we collected blood from COVID-
19 patients who have recently become virus-free and therefore were discharged, and
analyzed their SARS-CoV-2-specific antibody and T cell responses.
Clinical and pathological characteristics of the 12 COVID-19 patients in this study were
shown in Table 1. All the patients initially showed mild symptoms via CT scan and were
positive during SARS-CoV-2 nucleic acid testing. Of them, 6 (patients #1-6) were newly
discharged, while the remaining 6 were 2 weeks post discharge (follow-up patients, patients
#7-12). In line with the previous reports, 2 patients (#5, 8) showed lymphopenia (normal range
is 1.1-3.2X10e9 cells per L). However, only two travelled to Wuhan city within the past 3
months. One serum was obtained before the SARS-CoV-2 outbreak (healthy donor #1). 3
All rights reserved. No reuse allowed without permission.
perpetuity.
preprint (which was not certified by peer review) is the author/funder, who has granted medRxiv a license to display the preprint in
The copyright holder for thisthis version posted March 20, 2020. .https://doi.org/10.1101/2020.03.17.20036640doi: medRxiv preprint
 
 
4
 
additional healthy donors (#2-4) who had been in close contacts with the patients were
recruited in this study. Human AB serum (GemCell, CA) was used as a negative control.
In order to detect anti-viral immune responses, we first constructed recombinant pET28-
N-6XHis by linking 6 copies of His tag to the C-terminus of NP in the pET28-N vector (Biomed,
Cat. number: BM2640). Escherichia coli transformed with pET28-N-6xHis was lysed and
tested by Coomassie blue staining to confirm NP expression at 45.51 kDa. NP was further
purified by Ni-NTA affinity chromatography and gel filtration. The purity of NP was
approximately 90% (Figure S1A). The presence of NP was subsequently confirmed by anti-
Flag antibody (Figure S1B). The RBD region of S protein (S-RBD) and main protease (doi:
https://doi.org/10.1101/2020.02.19.956235) were produced by a baculovirus insect
expression system and purified to reach the purity of 90% (Figure S1A).
Using sera from patients and healthy donors, IgG and IgM against SARS-CoV-2 NP, main
protease and S-RBD antigens were analyzed. There was no significant antibody response to
main protease in sera from several patients (data not shown), suggesting that it may not serve
as an antigen for humoral immunity. We thus focused on NP and S-RBD. The serum from a
patient and human AB serum were titrated in order to determine optimal dilutions (data not
shown). Dilution of 1:50 was used for IgM and 1:150 for IgG. NP- and S-RBD-specific IgM and
IgG antibodies were both detected in the sera of newly discharged patients, compared with
healthy donor groups (Figure 1A). Anti-SARS-CoV-2 IgG antibodies were also more obviously
observed than IgM in the follow-up patients (#7-12), when compared with healthy donors
(Figure 1A). Taken together, these findings indicate that COVID-19 patients mounted IgG and
IgM responses to SARS-CoV-2 proteins, especially NP and S-RBD, and also suggest that
infected patients could maintain their IgG levels, at least for two weeks.
Since RBD domain of the S protein has been shown to bind to human receptor ACE2
3
,
the existence of antibodies against it may suggest neutralization of SARS-CoV-2 infection. To
assess this, we performed pseudovirus particle-based neutralization assay. As shown in
Figure 1B, patients #1, 2, 4 and 5, all within the discharged group, had high levels of
neutralizing antibody titers. These results demonstrate that most recently discharged patients
All rights reserved. No reuse allowed without permission.
perpetuity.
preprint (which was not certified by peer review) is the author/funder, who has granted medRxiv a license to display the preprint in
The copyright holder for thisthis version posted March 20, 2020. .https://doi.org/10.1101/2020.03.17.20036640doi: medRxiv preprint
 
 
5
 
had protective humoral immunity to SARS-CoV-2. All except patient #11, the follow-up
patients had lower levels of neutralizing antibody titers than recently discharged patients,
though all positive with the exception of patient #7 being negative. Whether protective humoral
immunity can be maintained needs further investigation.
To explore cellular immune responses to SARS-CoV-2, we isolated PBMCs from the
whole blood and phenotypically analyzed them by flow cytometry (Figure 2A). We found that
compared to discharged patients, there was a trend towards an increased frequency of NK
cells in the follow-up patients (Figure S2). However, there was no significant difference in
terms of the percentages of T cells among those two groups and the healthy donors.
To assess virus-specific cellular immunity, we then treated PBMCs with recombinant NP,
main protease and S-RBD, followed by IFN-g ELISpot analysis. The results were considered
positive if there were at least 1-fold increase in the numbers of IFN-g-secreting T cells in the
subject than in the healthy donors. As shown in Figure 2B, compared with healthy donors, the
numbers of IFN-g-secreting NP-specific T cells in patients #1, 2, 4 and 5 were much higher
than other patients, suggesting that they had developed SARS-CoV-2-specific T cell
responses. Of note, patients #1, 2, 4 and 5 developed both strong humoral and cellular
immune responses. Main protease-specific T cells were detected in patient #1, 2 and 5, while
patients # 1, 2, 4, 5 and 6 showed S-RBD-specific T cells. Although the numbers of IFN-g-
secreting S-RBD specific T cells were much lower than those of NP-specific T cells, they could
be detected in more patients than those for other viral proteins. S-RBD thus not only elicited
humoral immunity that may result in blockade of receptor binding during viral entry in host
cells, but also induced T cell immune responses, suggesting S-RBD is a promising target for
SARS-CoV-2 vaccines. In the follow-up patients, only patient #8 who showed lymphopenia
before treatment still had a high number of IFN-g-secreting T cells in response to NP, main
protease and S-RBD (Figure 2B), which suggests that anti-viral T cells may not be maintained
at high numbers in the PBMCs in the recovered patients. More interestingly, when combining
all 12 patients in our analysis, there was a significant correlation between the neutralizing
All rights reserved. No reuse allowed without permission.
perpetuity.
preprint (which was not certified by peer review) is the author/funder, who has granted medRxiv a license to display the preprint in
The copyright holder for thisthis version posted March 20, 2020. .https://doi.org/10.1101/2020.03.17.20036640doi: medRxiv preprint
 
 
6
 
antibody titers and the numbers of NP-specific T cells (Figure 2C), indicating that the
development of neutralizing antibodies may be correlated with the activation of anti-viral T
cells. Thus, effective clearance of virus may need collaborative humoral and cellular immune
responses.
In summary, we provided the first analysis of SARS-CoV-2-specific humoral and cellular
immunity. Both were detected in newly discharged patients, suggesting both participate in
immune-mediated protection to viral infection. However, follow-up patients exhibited high titers
of IgG antibodies, but with low levels of virus-specific T cells, suggesting that they may enter
a quiescent state.
Our work has thus provided a basis for further analysis of protective immunity to SARS-
CoV-2, and understanding the pathogenesis of COVID-19, especially in the severe cases. It
has also implications in designing an effective vaccine to protect and treat SARS-CoV-2
infection.
 
 
All rights reserved. No reuse allowed without permission.
perpetuity.
preprint (which was not certified by peer review) is the author/funder, who has granted medRxiv a license to display the preprint in
The copyright holder for thisthis version posted March 20, 2020. .https://doi.org/10.1101/2020.03.17.20036640doi: medRxiv preprint
 
 
7
 
Materials and Methods
Ethics statement
All procedures followed were in accordance with the ethical standards of the responsible
committee on human experimentation (the institutional review board at Tsinghua University)
and with the Helsinki Declaration of 1975, as revised in 2000. Informed consent was obtained
from all subjects for being included in the study. All patient data were anonymized before study
inclusion. The blood samples of COVID-19 patients and healthy donors were obtained from
Chui Yang Liu Hospital affiliated to Tsinghua University in Beijing.
 
Expression and Purification of recombinant proteins
The recombinant His-tagged NP of SARS-CoV-2 was expressed in E. coli by a T7
expression system, with 1 mM IPTG induction at 37 ºC for 4 h. pET-28aHis-tagged S-RBD
(amino acids 319-541) was expressed by a Baculovirus system in insect cells (doi:
https://doi.org/10.1101/2020.02.19.956235). Purified proteins were identified by SDS-PAGE
gels and stained with Coomassie blue. Western blot was performed to confirm their
antigenicity by mouse anti-His monoclonal antibody (Proteintech, HRP-66005).
 
Isolation of PBMC
PBMCs were isolated from anti-coagulant blood using Ficoll-Hypaque gradients (GE
Healthcare Life Sciences, Philadelphia, PA) as previously described
18
under the BSL-3 facility
in AMMS.
 
Anti-SARS-CoV-2 IgG/IgM ELISA
For IgM/IgG testing, 96-well ELISA plates were coated overnight with recombinant NP
and S-RBD (100 ng/well). The sera from COVID-19 patients were incubated for 1 h at 37°C.
An anti-Human IgG-biotin conjugated monoclonal antibody (Sino Biological Inc., Wayne, PA)
and streptavidin-HRP were used at a dilution of 1:500 and 1:250, respectively, and anti-human
All rights reserved. No reuse allowed without permission.
perpetuity.
preprint (which was not certified by peer review) is the author/funder, who has granted medRxiv a license to display the preprint in
The copyright holder for thisthis version posted March 20, 2020. .https://doi.org/10.1101/2020.03.17.20036640doi: medRxiv preprint
 
 
8
 
IgM-HRP conjugated monoclonal antibody (Inova Diagnostics, Inc., San Diego, CA). The OD
value at 450 was calculated.
 
Neutralizing antibody assay
Pseudovirus expressing the SARS-CoV-2 S protein was produced as described
previously
19
. pNL43Luci and GP-pCAGGS were co-transfected into 293T cells. 48 hours later,
SARS-CoV-2 pseudovirus-containing supernatants were mixed with at least 6 serially diluted
serum samples from the COVID-19 patients at 37°C for 1 hour. Then the mixtures were
transferred to 96-well plates containing monolayers of Huh-7 cells. 3 hours later, the medium
was replaced. After incubation for 48 h, the cells were washed, harvested in lysis buffer and
analyzed for luciferase activity by the addition of luciferase substrate. Inhibition rate = [1-(the
sample group- the cell control group) / (the virus control group- the cell control group)] x 100%.
The neutralizing antibody titer were calculated by performing S-fit analysis via Graphpad Prism
7 software.
 
Interferon Gamma (IFN-γ) ELISpot
IFN-g-secreting T cells were detected by Human IFN-g ELISpot
pro
kits (MABTECH AB,
Sweden) according to the manufacture protocol. Fresh PBMCs were plated in duplicate at
150k per well and then incubated 48h with 1uM of recombinant proteins. Spots were then
counted using an ELIspot Reader System (AT-Spot2100, atyx). The number of spots was
converted into the number of spots per million cells and the mean of duplicate wells plotted.
 
FACS staining
PBMCs were washed with PBS plus 2% FBS (Gibco, Grand Island, NY), and then Fc
blocking reagent (Meltenyi Biotec, Inc., Auburn, CA) was added followed by a wash with PBS
plus 2% FBS. Cells were then incubated for 30 min on ice with anti-CD45 (H130) (BioLegend),
anti-CD3 (OKT3) (BioLegend), anti-CD8 (SK1) (BD), anti-CD56 (HCD56) (BioLegend), anti-
All rights reserved. No reuse allowed without permission.
perpetuity.
preprint (which was not certified by peer review) is the author/funder, who has granted medRxiv a license to display the preprint in
The copyright holder for thisthis version posted March 20, 2020. .https://doi.org/10.1101/2020.03.17.20036640doi: medRxiv preprint
 
 
9
 
CD38 (HIT2) (BioLegend) and live/dead fixable aqua dye (eF660, eBioscience), washed twice
with PBS plus 2% FBS and then stored at 4 °C until acquired by FACS Verse (BD Biosciences,
San Jose, CA). Data were analyzed using FlowJo software (Version 10.0.8, Tree Star Inc.,
Ashland, Or).
 
Statistical analysis
Prism 7 software is used for statistical analysis. Student’s t test was performed for two-
group analysis. Pearson’s correlation coefficients were calculated. P values less than 0.05
were considered to be statistically significant.
 
ACKNOWLEDGEMENT
This work was supported in part by grants from Natural Science Foundation of China
(NSFC, Grant No. 31991173 to CD), National key research (Grant No. 2016YFC130390 to
LN) and an award from Zhejiang University Foundation (to CD).
 
Conflict of interest
LN, YF, WP and CD have filed a provisional patent on the methodology of detecting
SARS-CoV-2-specific antibody responses.
 
 
 
 
 
 
 
 
All rights reserved. No reuse allowed without permission.
perpetuity.
preprint (which was not certified by peer review) is the author/funder, who has granted medRxiv a license to display the preprint in
The copyright holder for thisthis version posted March 20, 2020. .https://doi.org/10.1101/2020.03.17.20036640doi: medRxiv preprint
 
 
10
 
References
 
 
1. Wang C, Horby PW, Hayden FG, Gao GF. A novel coronavirus outbreak of global health concern.
Lancet 2020; 395(10223): 470-3.
2. Phelan AL, Katz R, Gostin LO. The Novel Coronavirus Originating in Wuhan, China: Challenges
for Global Health Governance. JAMA 2020.
3. Zhou P, Yang XL, Wang XG, et al. A pneumonia outbreak associated with a new coronavirus of
probable bat origin. Nature 2020.
4. Zhu N, Zhang D, Wang W, et al. A Novel Coronavirus from Patients with Pneumonia in China,
2019. N Engl J Med 2020; 382(8): 727-33.
5. Lu R, Zhao X, Li J, et al. Genomic characterisation and epidemiology of 2019 novel coronavirus:
implications for virus origins and receptor binding. Lancet 2020; 395(10224): 565-74.
6. Wu F, Zhao S, Yu B, et al. A new coronavirus associated with human respiratory disease in
China. Nature 2020.
7. Huang C, Wang Y, Li X, et al. Clinical features of patients infected with 2019 novel coronavirus
in Wuhan, China. Lancet 2020; 395(10223): 497-506.
8. Chan JF, Yuan S, Kok KH, et al. A familial cluster of pneumonia associated with the 2019 novel
coronavirus indicating person-to-person transmission: a study of a family cluster. Lancet 2020;
395(10223): 514-23.
9. Mahallawi WH, Khabour OF, Zhang Q, Makhdoum HM, Suliman BA. MERS-CoV infection in
humans is associated with a pro-inflammatory Th1 and Th17 cytokine profile. Cytokine 2018; 104: 8-
13.
10. Nicholls JM, Poon LL, Lee KC, et al. Lung pathology of fatal severe acute respiratory syndrome.
Lancet 2003; 361(9371): 1773-8.
11. Wang W, Wang H, Deng Y, et al. Characterization of anti-MERS-CoV antibodies against various
recombinant structural antigens of MERS-CoV in an imported case in China. Emerg Microbes Infect
2016; 5(11): e113.
12. Ko JH, Muller MA, Seok H, et al. Serologic responses of 42 MERS-coronavirus-infected patients
according to the disease severity. Diagn Microbiol Infect Dis 2017; 89(2): 106-11.
13. Shi Y, Wan Z, Li L, et al. Antibody responses against SARS-coronavirus and its nucleocaspid in
SARS patients. J Clin Virol 2004; 31(1): 66-8.
14. Woo PC, Lau SK, Wong BH, et al. Detection of specific antibodies to severe acute respiratory
syndrome (SARS) coronavirus nucleocapsid protein for serodiagnosis of SARS coronavirus pneumonia.
J Clin Microbiol 2004; 42(5): 2306-9.
15. Vijay R, Perlman S. Middle East respiratory syndrome and severe acute respiratory syndrome.
Curr Opin Virol 2016; 16: 70-6.
16. Cameron MJ, Bermejo-Martin JF, Danesh A, Muller MP, Kelvin DJ. Human
immunopathogenesis of severe acute respiratory syndrome (SARS). Virus Res 2008; 133(1): 13-9.
17. Thiel V, Weber F. Interferon and cytokine responses to SARS-coronavirus infection. Cytokine
Growth Factor Rev 2008; 19(2): 121-32.
18. Xie S, Huang J, Qiao Q, et al. Expression of the inhibitory B7 family molecule VISTA in human
colorectal carcinoma tumors. Cancer Immunol Immunother 2018; 67(11): 1685-94.
19. Deng HK, Unutmaz D, KewalRamani VN, Littman DR. Expression cloning of new receptors used
by simian and human immunodeficiency viruses. Nature 1997; 388(6639): 296-300.
 
 
 
 
 
 
 
All rights reserved. No reuse allowed without permission.
perpetuity.
preprint (which was not certified by peer review) is the author/funder, who has granted medRxiv a license to display the preprint in
The copyright holder for thisthis version posted March 20, 2020. .https://doi.org/10.1101/2020.03.17.20036640doi: medRxiv preprint
 
 
11
 
 
Figure legends
 
Figure 1. Detection of antibody responses to recombinant SARS-CoV-2 proteins in
COVID-19 patients.
(A) Serological responses of 12 COVID-19 patients to recombinant NP (top) and S-RBD
(bottom). The experiment was performed in duplicates. (B) Measurement of neutralizing
antibody titers by pseudovirus-based assay. The experiment was performed in triplicates. NP,
nucleocapsid protein. S-RBD, receptor binding domain of spike protein. HD, healthy donor. Pt,
patient. HD#1, the serum was collected in 2018. HD#2-4, the sera were from close contacts.
*P<0.05, 0.05<**P<0.001, ***P<0.001.
 
Figure 2. T cell responses to recombinant SARS-CoV-2 proteins in COVID-19 patients.
(A) Phenotypic analysis of PBMCs from representative COVID-19 patients. (B) IFN-g ELISpot
analysis of COVID-19 patients to recombinant proteins. The experiment was performed in
duplicates. (C) Correlation analysis of the neutralizing antibody titers with the numbers of NP-
specific T cells (n=12). M protease, main protease. NP, nucleocapsid protein. S-RBD, receptor
binding domain of spike protein.
 
 
All rights reserved. No reuse allowed without permission.
perpetuity.
preprint (which was not certified by peer review) is the author/funder, who has granted medRxiv a license to display the preprint in
The copyright holder for thisthis version posted March 20, 2020. .https://doi.org/10.1101/2020.03.17.20036640doi: medRxiv preprint
 
 
 
Table 1 Clinical and pathological characteristics of the COVID-19 patients
 
Pt#
Sex
Age
Travel in
Wuhan
Fever
Fatigue
Lymphocyte
Count
Days in
hospital
Discharge
CT scan
NA test
CT scan
NA test
#1
F
51
yes
yes
yes
1.1X10
9
/L
33
Patchy ground
glass shadows on
both lungs
 
 
P
 
 
improvement
 
 
N
#2
F
42
no
no
no
2.5X10
9
/L
27
Multiple patchy
ground glass and
high-density
shadows in both
lungs
 
 
 
 
 
P
 
 
 
 
 
improvement
 
 
 
 
 
N
#3
M
32
no
yes
no
1.7X10
9
/L
36
Exudative lesion
of the right lower
lung
 
 
P
 
 
improvement
 
 
N
#4
M
49
no
yes
no
1.5X10
9
/L
32
Patchy ground
glass shadows on
both lungs
 
 
P
significant
improvement
 
 
N
#5
F
62
no
yes
yes
0.8X10
9
/L
37
Patchy ground
glass shadows on
both lungs
 
 
P
 
significant
improvement
 
 
N
#6
M
32
no
yes
yes
2.1X10
9
/L
17
Multiple ground
glass shadows in
both lungs
 
 
P
 
significant
improvement
 
N
#7
F
26
no
yes
no
2.9X10
9
/L
12
Right lung
inflammation
 
P
 
normal
 
N
#8
M
68
no
yes
no
0.7X10
9
/L
14
Multiple patchy
ground glass
shadows are seen
in the left lung,
and the upper
lobe of the left
lung is obvious
 
 
 
 
 
 
P
 
 
 
 
 
significant
improvement
 
 
 
 
 
 
N
#9
F
37
no
no
yes
1.9X10
9
/L
12
Double lung veins
thickened
 
P
 
normal
 
N
#10
F
29
no
yes
yes
1.9X10
9
/L
13
Ground glass in
the pleura of the
lower lobe of both
lungs
 
 
 
P
 
 
 
normal
 
 
 
N
#11
F
31
yes
yes
no
1.1X10
9
/L
19
Patchy ground
glass shadows on
both lungs
 
 
P
 
significant
improvement
 
 
N
#12
M
35
no
yes
yes
2.3X10
9
/L
11
Multiple ground
glass shadows in
both lungs
 
 
P
 
 
normal
 
 
N
 
Notes: pt, patient; F, female; M, male; P, positive; N, negative; NA, nucleic acid
All rights reserved. No reuse allowed without permission.
perpetuity.
preprint (which was not certified by peer review) is the author/funder, who has granted medRxiv a license to display the preprint in
The copyright holder for thisthis version posted March 20, 2020. .https://doi.org/10.1101/2020.03.17.20036640doi: medRxiv preprint
 
 
Figure 1
B
NP-based ELISA
S-RBD-based ELISA
B
IgM
IgG
IgM
IgG
A
IgG NP 1:50
AB serum
HD from Donglab
HD from hospital
Pts #1-6
Pts #7-12
0.0
0.5
1.0
1.5
2.0
2.5
Serum
OD
450
AB serum
HD from Donglab
HD from hospital
Pts #1-6
Pts #7-12
***
***
AB serum
HD#1
HD#2-5
Pts#1-6
Pts#7-12
#1
#2
#3
#4
#5
#1
#2
#3
#4
#5
#6
#7
#8
#9
#10
#11
#12
HD
Discharged pt.
Follow-up pt.
Neutralizing antibody titer
All rights reserved. No reuse allowed without permission.
perpetuity.
preprint (which was not certified by peer review) is the author/funder, who has granted medRxiv a license to display the preprint in
The copyright holder for thisthis version posted March 20, 2020. .https://doi.org/10.1101/2020.03.17.20036640doi: medRxiv preprint
 
 
none
NP
main protease
S-RBD
0
1000
2000
3000
4000
Spot counts per million PBMCs
Pt#7
Pt#8
Pt#9
Pt#10
Pt#11
Pt#11
none
NP
S
-
RBD
none
NP
main protease
S-RBD
0
1000
2000
3000
4000
Spot counts per million PBMCs
Pt#7
Pt#8
Pt#9
Pt#10
Pt#11
Pt#12
M protease
A
Gated on live
lymphocytes
Gated on live
CD3
+
CD56
-
cells
Gated on live
CD3
+
CD56
-
cells
CD56
CD3
CD8
CD3
CD38
CD3
HD
Discharged
Follow-up
Figure 2
B
Spot counts per million PBMCs
none
NP
M protease
C
Neutralizing antibody titer
NP-specific T cell No.
none
S-RBD
0
500
1000
1500
2000
Spot counts per million PBMCs
HD#1
HD#2
HD#3
Pt#1
Pt#2
Pt#3
Pt#4
Pt#5
Pt#6
none
S
-
RBD
none
NP
main protease
0
500
1000
1500
2000
2000
2500
3000
3500
4000
Spot counts per million PBMCs
HD#1
HD#2
HD#3
Pt#1
Pt#2
Pt#3
Pt#4
Pt#5
Pt#6
none
NP
main protease
S-RBD
0
500
1000
1500
2000
2000
2500
3000
3500
4000
Spot counts per million PBMCs
HD#1
HD#2
HD#3
Pt#1
Pt#2
Pt#3
Pt#4
Pt#5
Pt#6
All rights reserved. No reuse allowed without permission.
perpetuity.
preprint (which was not certified by peer review) is the author/funder, who has granted medRxiv a license to display the preprint in
The copyright holder for thisthis version posted March 20, 2020. .https://doi.org/10.1101/2020.03.17.20036640doi: medRxiv preprint