Skip to main content
Erschienen in: European Journal of Clinical Microbiology & Infectious Diseases 6/2021

Open Access 03.03.2021 | COVID-19 | Original Article

Automated Western immunoblotting detection of anti-SARS-CoV-2 serum antibodies

verfasst von: Sophie Edouard, Rita Jaafar, Nicolas Orain, Philippe Parola, Philippe Colson, Bernard La Scola, Pierre-Edouard Fournier, Didier Raoult, Michel Drancourt

Erschienen in: European Journal of Clinical Microbiology & Infectious Diseases | Ausgabe 6/2021

Abstract

ELISA and chemiluminescence serological assays for COVID-19 are currently incorporating only one or two SARS-CoV-2 antigens. We developed an automated Western immunoblotting as a complementary serologic assay for COVID-19. The JessTM Simple Western system, an automated capillary-based assay, was used, incorporating an inactivated SARS-CoV-2 lineage 20a strain as the source of antigen, and total immunoglobulins (IgG, IgM, IgA) detection. In total, 602 sera were tested including 223 from RT-PCR-confirmed COVID-19 patients, 76 from patients diagnosed with seasonal HCoVs and 303 from coronavirus-negative control sera. We also compared this assay with the EUROIMMUN® SARS-CoV-2 IgG ELISA kit. Among 223 sera obtained from RT-PCR-confirmed COVID-19 patients, 180/223 (81%) exhibited reactivity against the nucleocapsid and 70/223 (31%) against the spike protein. Nucleocapsid reactivity was further detected in 9/76 (14%) samples collected from patients diagnosed with seasonal HCoVs and in 15/303 (5%) coronavirus-negative control samples. In the subset of sera collected more than 2 weeks after the onset of symptoms, the sensitivity was 94% and the specificity 93%, the latter value probably reflecting cross-reactivity of SARS-CoV-2 with other coronaviruses. The automated Western immunoblotting presented a substantial agreement (90%) with the compared ELISA (Cohen’s Kappa=0.64). Automated Western immunoblotting may be used as a second line test to monitor exposure of people to HCoVs including SARS-CoV-2.

Introduction

To date, seven coronaviruses have been reported as human pathogens, including four seasonal coronaviruses (Alphacoronavirus 229E and NL63 and Betacoronavirus HKU1 and OC43) here referred to as HCoVs, which are associated with causing mild-to-severe upper and lower respiratory tract infections [1]. Two other betacoronaviruses that caused severe acute respiratory syndrome in 2002 in China (SARS-CoV) and the Middle East Respiratory Syndrome in 2012 in Saudi Arabia (MERS-CoV) [2] and the Betacoronavirus SARS-CoV-2 that is the agent of the COVID-19 pandemic have been demonstrated to infect a variety of animals and humans [3]. The latter is phylogenetically closely related to HCoV-HKU1 and presents a high sequence homology with SARS-CoV [2].
Serological assays used to explore exposure to seasonal HCoVs have previously indicated cross-immunity between all coronaviruses [46]. SARS-CoV-2 exhibits several antigens eliciting a serological response in COVID-19 patients, including spike glycoprotein, its N-terminal (S1), and C-terminal (S2) subunits as well as nucleocapsid [7]. Most of routinely used serological COVID-19 assays incorporated only one recombinant protein [810]. Second generation assays are combining two antigens to increase sensitivity and mostly specificity [7, 11].
We developed an automated Western immunoblotting (AWB) assay in order to characterize serological responses to SARS-CoV-2 and the potential cross-reactivity with HCoVs.

Patients and methods

Serum sample collections
A first set of 27 serum samples from 27 different patients with RT-PCR-documented COVID-19 [12], collected between March and April 2020, at least 2 weeks after the onset of symptoms were incorporated as a positive control group. All of them presented with an IgG titer ≥ 1:100 using in-house indirect immunofluorescence assay (IFA) [13]. Of these, 16 serum samples were used for conventional immunoblotting including 3 samples exhibiting low (1:200), moderate (1:800), and high (1:3200) IgG titers using IFA that were used to fix optimal conditions to be used for AWB (antigen, serum, and secondary antibodies concentrations). One serum collected in 2018, before the onset of COVID-19 (having a negative RT-PCR for HCoVs on homologous respiratory specimen), was included as negative control.
As for AWB, 223 serum samples (including the 27 serum samples described above) collected from 223 different RT-PCR-confirmed COVID-19 patients between March and September 2020 were incorporated as a positive control group. Twenty-seven of these sera were tested for antibodies to the recombinant S1 protein by EUROIMMUN® SARS-CoV-2 IgG ELISA (Euroimmun, Bussy Saint-Martin, France) performed using the Elispeed DUO system (Euroimmun) according to the manufacturer’s recommendations. The ratio (AUC sample/AUC calibrator) was interpreted as follows: <0.8 negative; ≥0.8 to <1.0 undetermined; ≥1.1 positive. We considered undetermined results as negative for statistical analyses. A negative control group (37 serum samples) consisted of (i) 10 serum samples obtained less than 5 days after the onset of symptoms in patients presenting high viral loads of SARS-CoV-2 (Ct values < 20) collected in March and April 2020; (ii) 14 sera from asymptomatic healthcare workers largely exposed to the virus but exhibiting negative results for RT-PCR and serology by IFA for SARS-CoV-2 during follow-up collected in March and April 2020; and (iii) 13 sera from patients collected between April and December 2019 before the pandemic and harboring negative RT-PCR results for the 4 HCoVs in their nasopharyngeal specimens. These 37 serum samples were also all tested by ELISA. A third group of 76 serum samples was retrieved from patients diagnosed with seasonal coronavirus infections (HCoV-NL63 (n=19), HCoV-OC43 (n=21), HCoV-229E (n=8), and HCoV-HKU1 (n=28)) between April and December 2019 an and were collected at least 2 weeks after the diagnosis, of which 45 were also tested by ELISA. A fourth group of 266 sera was collected between January 2018 and February 2020 from children and adults admitted in surgery departments (n=145) and other medical units (=121) before the pandemic, of which 88 serum samples were also tested by ELISA; their HCoVs status was unknown. Altogether, 197 sera tested by ELISA included 27 sera from COVID-19-positive patients and 170 from COVID-19-negative patients. Serum samples were collected from patients visiting four university hospitals in Marseille, France. They were stored frozen at −30°C and were tested retrospectively. No sample was collected specifically for this study which was approved by our institution’s ethics committee under No.2020-024.
Virus growth, purification, and concentration
The SARS-CoV-2 IHUMI2 strain (lineage 20a) was used as antigen as previously described [13]. One liter of infected cells was collected and clarified by centrifugation at 700g for 10 min and by filtering the supernatant through a 0.45-μm pore-sized filter (VWR, Radnor, PA, USA) and further a 0.2-μm pore-sized filter (Thermo Fisher Scientific, Waltham, MA, USA ). Virions were then aggregated by overnight precipitation at 4°C with 10% polyethylene glycol 8000 white flake type (PEG-8000, BioUltra, SIGMA-ALDRICH, USA) and 2.2% crystalline NaCl, with gentle swirling. Precipitated virus particles were then centrifuged at 10,000g for 30 min using a SORVALL Evolution centrifuge with SLA-3000 Recent 1 fixed angle rotor pre-cooled at 4°C (Kendro Laboratory Products, Newtown, CT, USA). The pellet was resuspended with HEPES-saline (0.9% NaCl, 10 mL of 1 M HEPES, 990 mL purified water) previously vacuum-sterilized through a 0.2-μm pore size membrane, swirled in the cold HEPES-saline until dissolution to avoid using pipetting as it may hurt viral spikes at this step. The resuspended pellet was then treated with a 30% sucrose cushion in 25 × 89 mm centrifuge tubes (Ultra-Clear, BECKMAN COULTER, CA, USA). Final purification was achieved by ultracentrifugation at 100,000g for 90 min at 4°C followed by two 30-min washes with Hanks Balanced Salt Solution using SORVALL Discovery 90SE with Surespin 630 rotor (Kendro Laboratory Products). The final pellet was resuspended in 400 μL of HEPES-buffered saline and heat-inactivated at 65°C for 1h.
Conventional Western immunoblotting
SARS-CoV-2 antigens diluted to 0.5 mg/mL were mixed (v/v) with 2X Laemmli Sample Buffer (Bio-Rad, Hercules, CA, USA) before a heating step of 5 min at 95°C. This preparation and a ladder were dispensed in wells shaped in a 5 % polyacrylamide stacking gel. The protein separation was then performed in a 10% polyacrylamide separating gel with a Mini Trans-blot cell device (Bio-Rad) at 160 V for 90 min. After transferring proteins from the gel to a 0.45-μm-pore size nitrocellulose membrane (Bio-Rad) at 100 V and 15°C for 90 min, the membrane was left at 4°C overnight with 5% non-fat milk powder in Tris buffered saline (TBS) with 0.5% Tween 20 (Euromedex, Souffelweyersheim, France). Blocked strips were incubated with sera diluted at 1:50 for 60 min. Three washes of 10 min were performed before a 90-min incubation of the strips with goat peroxidase-conjugated anti-human IgG/IgM/IgA (Jackson ImmunoResearch, Ely, UK) diluted 1:1000. Three washes of 10 min with TBS buffer with 0.5% Tween 20 were made. Strips were put in contact with ECL Western Blotting Substrate (Promega, Madison, WI, USA) and the reaction with secondary antibody peroxydases was revealed with a Fusion Fx chemiluminescence imaging system and analyzed with the Fusion software (Vilber, Marne-la-Vallée, France).
Automated Western immunoblotting
The JessTM Simple Western system (ProteinSimple, San Jose CA, USA,) is an automated capillary-based size separation and nano-immunoassay system. To quantify the absolute serological response to viral antigens, we followed the manufacturer’s standard method for 12-230-kDa Jess separation module (SM-W004). The SARS-CoV-2 antigen (1 μg/μL) was mixed with 0.1X Sample buffer and Fluorescent 5X Master mix (ProteinSimple) to achieve a final concentration of 0.25 μg/μL in the presence of fluorescent molecular weight markers and 400 mM dithiothreitol (ProteinSimple). This preparation was denatured at 95°C for 5 min. Ladder (12-230-kDa PS-ST01EZ) and SARS-CoV-2 proteins were separated in capillaries as they migrated through a separation matrix at 375 volts. A ProteinSimple proprietary photoactivated capture chemistry was used to immobilize separated viral proteins on the capillaries. Patients’ sera diluted at a 1:2 were added and incubated for 60 min. After a wash step, goat HRP-conjugated anti-human IgG/IgM/IgA antibodies (Jackson ImmunoResearch) diluted 1:500 were added for 30 min. The chemiluminescent revelation was established with peroxyde/luminol-S (ProteinSimple). Digital image of chemiluminescence of the capillary was captured with Compass Simple Western software (version 4.1.0, Protein Simple) that calculated automatically heights (chemiluminescence intensity), area, and signal/noise ratio. Results could be visualized as electropherograms representing peak of chemiluminescence intensity and as lane view from signal of chemiluminescence detected in the capillary. An internal system control was included in each run.
Statistical analysis
ROC curves were performed using XL stat. The agreement rate and Cohen’s Kappa value were determined for agreement between ELISA and AWB. For data comparisons and statistical analyses, the Fisher’s exact test, Chi-squared test, Mann-Whitney test, and standard statistical software (GraphPad Prism v7) were used. A p value < 0.05 was considered statistically significant.

Results

Fixing automated Western immunoblotting parameters
Protein profiles of the purified SARS-CoV-2 antigen and uninfected Vero cells were verified on silver-stained 2-D gels. As expected, the viral-specific and major-dominant proteins were N, S, S1, and S2 proteins at 42, 170, 110, and 90 kDa, respectively. All 16 serum samples collected from 16 different COVID-19 patients exhibited reactivity against the nucleocapsid and spike proteins. Parameter optimization to translate these data on AWB included an antigen concentration of 0.25 μg/μL, a serum dilution at 1:2, and a secondary antibody dilution of 1:500 (data not shown). In these conditions, AWB of positive serum samples yielded a prominent 56-kDa band interpreted as the nucleocapsid and a 217-kDa band interpreted as the spike protein (Fig. S1). Higher molecular weight values observed with AWB than with SDS-PAGE were due to the different composition of gels in the capillaries. In total, the 16 sera from COVID-19 patients tested with conventional and AWB gave similar results except that AWB failed to detect the spike protein in one sample. Furthermore, AWB yielded significantly higher S/N ratios, peak height, and area under curve for the nucleocapsid (p < 0.0001) and spike proteins (p < 0.0001) in the 27 serum samples from COVID-19 patients than in 37 serum samples collected in negative control patients (Table 1). The S/N ratio presented higher Youden Index for nucleocapsid detection, being therefore interpreted as the most pertinent parameter to interpret AWB results. Optimal threshold for the S/N ratio of 110.4 conferred a 96.3% sensitivity and 94.6% specificity for the nucleocapsid detection. Determination of a cut-off to interpret results of spike protein was not useful and could be based only on presence/absence of signal with sensitivity to 66.7% and 100% specificity (Table 2). Therefore, we further used the presence of antibodies to the nucleocapsid with S/N ratio ≥ 110.4 and/or to the spike protein, as criteria to define a positive AWB.
Table 1
Automated Western immunoblotting results of 27 sera from COVID-19-positive patients and 37 sera from negative controls used to fix automated Western immunoblotting parameters (results expressed as median with 25 and 75 percentile)
 
Negative controls (n=37)
Sera from COVID-19-positive patients (n=27)
 
Early sera from COVID-19-positive patients (n=10)
Sera from negative HCoVs patients collected before the pandemic (n=13)
Sera from healthcare workers highly exposed to SARS-CoV-2 (n=14)
All (n=37)
Nucleocapsid (56 kDa)
  S/N ratio
44 (29.98–64.4)
62.1 (33–78.7)
49.85 (34.95–70.95)
49.85 (34.95–70.95)
421 (214.1–666.8)
  Height
6511 (5674–7729)
8854 (6428–12,995)
4934 (4397–5605)
6100 (4586–9813)
64,065 (32,338–121,517)
  Area
82,169 (65588–94,895)
124,541 (84495–147,319)
67,780 (61427–76,234)
79,634 (66110–128,111)
839,470 (524,393–1,597,656)
Spike (217 kDa)
  S/N ratio
0
0
0
0
27.9 (0–43.1)
  Height
0
0
0
0
3174 (0–8621)
  Area
0
0
0
0
40,699 (0–182,108)
S/N signal/noise
Table 2
Sensitivity and specificity of automated Western immunoblotting determined with 27 sera from COVID-19-positive patients and 37 sera from non COVID-19 patients
n=64
AUC
Youden index
Optimal cut-off value
Sensitivity (%)
Specificity (%)
True positive (number)
True negative (number)
False positive (number)
False negative (number)
Nucleocapsid
  S/N
0.975
0.907
110.4
96.3
94.6
26
35
2
1
  Height
0.982
0.885
249
88.9
100
24
37
0
3
  Area
0.970
0.885
287
88.9
100
24
37
0
3
Spike
  S/N
0.833
0.667
0
66.7
100
18
37
0
9
  Height
0.833
0.667
0
66.7
100
18
37
0
9
  Area
0.833
0.667
0
66.7
100
18
37
0
9
AUC area under the curve
Automated Western immunoblotting results
AWB yielded 395/602 (66%) negative and 207/602 (34%) positive serum samples (Table 3), giving an 81% sensitivity as 181/223 COVID-19 patients were positive (nucleocapsid detected in 180/223 (76%) and spike in 67/223 (30%), respectively), and a 93% specificity as 26/379 (7%) non-COVID-19 patients were positive; applying above-reported cut-off criteria (Figs. 1 and 2a). Accordingly, positive (PPV) and negative predictive values (NPV) were of 87% and 89%, respectively. Sera from COVID-19 patients were collected with a median of 13 days (range 0 to 165) after the onset of symptoms. Sensitivity was 54% among sera collected less than 10 days after the onset of symptoms and increased to 94% among sera collected more than 10 days after the onset of symptoms (Fig. 3). AWB had a 90% agreement with the herein compared ELISA assay (Cohen’s Kappa=0.64) as the latter was positive in 22/27 (81.5%) COVID-19 patients and 6/170 (3.5%) non-COVID-19 patients, yielding a sensitivity of 81.5% and specificity of 97% (Table 4).
Table 3
Results of automated Western immunoblotting including the 602 sera tested
 
Negative controls
Sera from patients diagnosed with others HCoVs (n=76)
Sera collected before the pandemic from patients with unknown status for HCoVs (n=266)
Sera from COVID-19-positive patients (n=223)
Total (n=602)
 
Early sera from COVID-19-positive patients (n=10)
Sera from negative HCoVs patients collected before the pandemic (n=13)
Sera from healthcare workers highly exposed to SARS-CoV-2 (n=14)
All (n=37)
Positive sera
  Nucleocapsid reactivity
1 (10%)
0
2 (14%)
3 (8%)
9 (12%)
12 (4.5%)
180 (81%)
204 (34%)
  Spike reactivity
0
0
0
0
2 (3%)
0
67 (30%)
69 (11%)
  Total
1 (10%)
0
2 (14%)
3 (8%)
11 (14.5%)
12 (4.5%)
181 (81%)
207 (34%)
Table 4
Comparison between automated Western immunoblotting and commercial SARS-CoV-2 IgG ELISA on 197 sera
 
AWB IgT number (%)
ELISA IgG number (%)
Sera from COVID-19 positive patients (n=27)
27 (100)
22 (81)
Sera from negative control group (n=37)
3 (8)
0
  Early sera from COVID-19 positive patients (n=10)
1 (10)
0
  Sera from healthcare workers highly exposed to SARS-CoV-2 (n=14)
2 (14)
0
  Sera from negative HCoVs patients collected before the pandemic (n=13)
0
0
Sera from patients diagnosed with other HCoVs (n=45)
6 (13)
0
Sera collected before the pandemic from patients with unknown status for HCoVs (n=88)
5 (6)
6 (7)
Detailing false-positive AWB, antibodies to the nucleocapsid were detected in 3/37 (8%) negative control serum samples. Also, 9/76 (14%) of serum sampled from patients diagnosed with seasonal HCoVs reacted with the nucleocapsid which was detected in 5/28 (18%) of patients with HCoV-HKU1, 2/19 (10.5%) with HCoV-NL63, 2/21 (9.5%) with HCoV-OC43, but in none of HCoV-229E patients (Fig. 2b). In addition, one HCoV-HKU1 serum and one HCoV-NL63 serum reacted against the spike protein, increasing the number of total cross-reactions to 11/76 (14.5%) for this group. Among 266 serums sampled before the COVID-19 epidemic in France, albeit of unknown status for HCoVs, 12/266 (4.5%) reacted against the nucleocapsid but none against the spike protein.
Most cross-reactivities were detected in 46–65-year-old patients (7/63) and more than 65-year-old (4/43) patients (Fig. 2c). Cross-reactivity was more prevalent in subjects > 21 years (15/173) than in children ≤ 15 years (6/126) but this difference was not significant (p = 0.25, Fisher’s exact test).

Discussion

An AWB, incorporating whole SARS-CoV-2 viral particles, was demonstrated to be efficient in detecting specific antibodies against SARS-CoV-2 dual nucleocapsid and spike proteins, achieving a 87% PPV and a 89% NPV for COVID-19, in the population tested in this study. Accordingly, dual nucleocapsid and spike protein detections exhibited 81% sensitivity and 93% specificity. Indeed, the spike protein was detected in only two non-COVID-19 patients whereas the nucleocapsid protein was detected in 24 non-COVID-19 patients, including 11 patients diagnosed with HCoVs. In our study, AWB results were consistent with results obtained using a commercially available ELISA incorporating recombinant spike-1 protein. The serological observations obtained in this study therefore indicated that it is worth developing next generation serological assays incorporating both the nucleocapsid and the spike proteins, in order to achieve almost 100% sensitivity and 100% specificity of SARS-CoV-2 infection, which is not the situation with first generation, commercially available serological assays [7, 11, 14].
It should be noted that cross-reactivity was more prevalent in patients infected with other betacoronaviruses (accounting for 31% of cross-reactivity) than in patients infected with alphacoronaviruses (accounting for 12% of cross-reactivity), being mainly supported by SARS-CoV-2 nucleocapsid (in 92% of cross-reacting serum samples), and mostly found in adult patients older than 46 years (accounting for 52% of sera with cross-reactivity). Our observations are consistent with previous reports that cross-reactions were observed with nucleocapsid while serological assays incorporating the spike protein have been reported to be more specific but less sensitive [46, 1518]. Cross-reactivity has been described between endemic coronaviruses and SARS-CoV-2 [19]. Several studies reported the presence of antibodies reacting with SARS-CoV-2 spike and nucleocapsid proteins in serum sampled before the pandemic and in HCoVs patients [20, 21].
In a few previous reports of the described AWB [2224], a recombinant protein was used as the antigen whereas we used purified virus antigen directly produced in the biosafety level 3 laboratory [13]. This fact could explain in part the important difference of sensitivity for the spike protein compared to the nucleocapsid, in our assay. Thereby, serum dilution was a critical parameter as the spike protein was detected only for a low, 1:2 dilution of serum. Nevertheless, the herein described AWB assay demonstrated a better standardization and reproducibility than conventional Western immunoblotting, proved to be user-friendly, and enabled analyzing 24 serum samples in less than 4 h. Result interpretation was not only based on presence/absence and intensity of bands but a chemiluminescent image was automatically analyzed with software allowing noise reduction. The “virtual image” of reactions present in the capillaries could be represented by peaks on electropherogram or lane views.
In conclusion, the herein described AWB may be incorporated as a first line serological test for the diagnosis of exposure to SARS-CoV-2 if limited series have to be investigated, or as a second-line assay to confirm or exclude the diagnosis of COVID-19 as also previously suggested by others [25], especially in patients with negative, doubtful, and discrepant RT-PCR results, and may be used to measure past exposition to the virus.

Acknowledgements

The authors acknowledge the contribution of the technical staff of the IHU Méditerranée Infection Laboratory.

Declarations

The study was performed in accordance with the ethical standards as laid down in the 1964 Declaration of Helsinki and its later amendments. The non-interventional retrospective and anonymized nature of this study was approved by the Ethical Committee of the IHU Méditerranée Infection under no. 2020-024.
Not applicable.
Not applicable.

Conflict of interest

The authors declare no competing interests.
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 link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence 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 licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​.

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

Literatur
1.
Zurück zum Zitat Zimmermann P, Curtis N (2020) Coronavirus infections in children including COVID-19: an overview of the epidemiology, clinical features, diagnosis, treatment and prevention options in children. Pediatr Infect Dis J 39:355–368CrossRef Zimmermann P, Curtis N (2020) Coronavirus infections in children including COVID-19: an overview of the epidemiology, clinical features, diagnosis, treatment and prevention options in children. Pediatr Infect Dis J 39:355–368CrossRef
2.
Zurück zum Zitat Coronaviridae Study Group of the International Committee on Taxonomy of Viruses (2020) The species severe acute respiratory syndrome-related coronavirus: classifying 2019-nCoV and naming it SARS-CoV-2. Nat Microbiol 5:536–544CrossRef Coronaviridae Study Group of the International Committee on Taxonomy of Viruses (2020) The species severe acute respiratory syndrome-related coronavirus: classifying 2019-nCoV and naming it SARS-CoV-2. Nat Microbiol 5:536–544CrossRef
3.
Zurück zum Zitat Zhu N, Zhang D, Wang W, Li X, Yang B, Song J, Zhao X, Huang B, Shi W, Lu R, Niu P, Zhan F, Ma X, Wang D, Xu W, Wu G, Gao GF, Tan W, China Novel Coronavirus Investigating and Research Team (2020) A novel coronavirus from patients with pneumonia in China, 2019. N Engl J Med 382:727–733CrossRef Zhu N, Zhang D, Wang W, Li X, Yang B, Song J, Zhao X, Huang B, Shi W, Lu R, Niu P, Zhan F, Ma X, Wang D, Xu W, Wu G, Gao GF, Tan W, China Novel Coronavirus Investigating and Research Team (2020) A novel coronavirus from patients with pneumonia in China, 2019. N Engl J Med 382:727–733CrossRef
4.
Zurück zum Zitat Maache M, Komurian-Pradel F, Rajoharison A, Perret M, Berland J-L, Pouzol S, Bagnaud A, Duverger B, Xu J, Osuna A, Paranhos-Baccalà G (2006) False-positive results in a recombinant severe acute respiratory syndrome-associated coronavirus (SARS-CoV) nucleocapsid-based western blot assay were rectified by the use of two subunits (S1 and S2) of spike for detection of antibody to SARS-CoV. Clin Vaccine Immunol 13:409–414CrossRef Maache M, Komurian-Pradel F, Rajoharison A, Perret M, Berland J-L, Pouzol S, Bagnaud A, Duverger B, Xu J, Osuna A, Paranhos-Baccalà G (2006) False-positive results in a recombinant severe acute respiratory syndrome-associated coronavirus (SARS-CoV) nucleocapsid-based western blot assay were rectified by the use of two subunits (S1 and S2) of spike for detection of antibody to SARS-CoV. Clin Vaccine Immunol 13:409–414CrossRef
5.
Zurück zum Zitat Woo PCY, Lau SKP, Wong BHL, Chan K-H, Hui W-T, Kwan GSW, Peiris JSM, Couch RB, Yuen K-Y (2004) False-positive results in a recombinant severe acute respiratory syndrome-associated coronavirus (SARS-CoV) nucleocapsid enzyme-linked immunosorbent assay due to HCoV-OC43 and HCoV-229E rectified by Western blotting with recombinant SARS-CoV spike polypeptide. J Clin Microbiol 42:5885–5888CrossRef Woo PCY, Lau SKP, Wong BHL, Chan K-H, Hui W-T, Kwan GSW, Peiris JSM, Couch RB, Yuen K-Y (2004) False-positive results in a recombinant severe acute respiratory syndrome-associated coronavirus (SARS-CoV) nucleocapsid enzyme-linked immunosorbent assay due to HCoV-OC43 and HCoV-229E rectified by Western blotting with recombinant SARS-CoV spike polypeptide. J Clin Microbiol 42:5885–5888CrossRef
6.
Zurück zum Zitat Che X-Y, Qiu L-W, Liao Z-Y, Wang Y, Wen K, Pan Y-X, Hao W, Mei Y-B, Cheng VCC, Yuen K-Y (2005) Antigenic cross-reactivity between severe acute respiratory syndrome-associated coronavirus and human coronaviruses 229E and OC43. J Infect Dis 191:2033–2037CrossRef Che X-Y, Qiu L-W, Liao Z-Y, Wang Y, Wen K, Pan Y-X, Hao W, Mei Y-B, Cheng VCC, Yuen K-Y (2005) Antigenic cross-reactivity between severe acute respiratory syndrome-associated coronavirus and human coronaviruses 229E and OC43. J Infect Dis 191:2033–2037CrossRef
7.
Zurück zum Zitat Burbelo PD, Riedo FX, Morishima C, Rawlings S, Smith D, Das S, Strich JR, Chertow DS, Davey RT, Cohen JI (2020) Sensitivity in detection of antibodies to nucleocapsid and spike proteins of severe acute respiratory syndrome coronavirus 2 in patients with coronavirus disease 2019. J Infect Dis 222:206–213CrossRef Burbelo PD, Riedo FX, Morishima C, Rawlings S, Smith D, Das S, Strich JR, Chertow DS, Davey RT, Cohen JI (2020) Sensitivity in detection of antibodies to nucleocapsid and spike proteins of severe acute respiratory syndrome coronavirus 2 in patients with coronavirus disease 2019. J Infect Dis 222:206–213CrossRef
8.
Zurück zum Zitat Deeks JJ, Dinnes J, Takwoingi Y, Davenport C, Spijker R, Taylor-Phillips S, Adriano A, Beese S, Dretzke J, Ferrante di Ruffano L, Harris IM, Price MJ, Dittrich S, Emperador D, Hooft L, Leeflang MM, Van den Bruel A, Cochrane COVID-19 Diagnostic Test Accuracy Group (2020) Antibody tests for identification of current and past infection with SARS-CoV-2. Cochrane Database Syst Rev 6:CD013652PubMed Deeks JJ, Dinnes J, Takwoingi Y, Davenport C, Spijker R, Taylor-Phillips S, Adriano A, Beese S, Dretzke J, Ferrante di Ruffano L, Harris IM, Price MJ, Dittrich S, Emperador D, Hooft L, Leeflang MM, Van den Bruel A, Cochrane COVID-19 Diagnostic Test Accuracy Group (2020) Antibody tests for identification of current and past infection with SARS-CoV-2. Cochrane Database Syst Rev 6:CD013652PubMed
9.
Zurück zum Zitat Guo L, Ren L, Yang S, Xiao M, Chang D, Yang F, Dela Cruz CS, Wang Y, Wu C, Xiao Y, Zhang L, Han L, Dang S, Xu Y, Yang Q-W, Xu S-Y, Zhu H-D, Xu Y-C, Jin Q, Sharma L, Wang L, Wang J (2020) Profiling early humoral response to diagnose novel coronavirus disease (COVID-19). Clin Infect Dis 71:778–785CrossRef Guo L, Ren L, Yang S, Xiao M, Chang D, Yang F, Dela Cruz CS, Wang Y, Wu C, Xiao Y, Zhang L, Han L, Dang S, Xu Y, Yang Q-W, Xu S-Y, Zhu H-D, Xu Y-C, Jin Q, Sharma L, Wang L, Wang J (2020) Profiling early humoral response to diagnose novel coronavirus disease (COVID-19). Clin Infect Dis 71:778–785CrossRef
10.
Zurück zum Zitat Liu Z-L, Liu Y, Wan L-G, Xiang T-X, Le A-P, Liu P, Peiris M, Poon LLM, Zhang W (2020) Antibody profiles in mild and severe cases of COVID-19. Clin Chem 66:1102–1104CrossRef Liu Z-L, Liu Y, Wan L-G, Xiang T-X, Le A-P, Liu P, Peiris M, Poon LLM, Zhang W (2020) Antibody profiles in mild and severe cases of COVID-19. Clin Chem 66:1102–1104CrossRef
11.
Zurück zum Zitat Lynch KL, Whitman JD, Lacanienta NP, Beckerdite EW, Kastner SA, Shy BR, Goldgof GM, Levine AG, Bapat SP, Stramer SL, Esensten JH, Hightower AW, Bern C, Wu AHB (2020) Magnitude and kinetics of anti-SARS-CoV-2 antibody responses and their relationship to disease severity. Clin Infect Dis. https://doi.org/10.1093/cid/ciaa979 Lynch KL, Whitman JD, Lacanienta NP, Beckerdite EW, Kastner SA, Shy BR, Goldgof GM, Levine AG, Bapat SP, Stramer SL, Esensten JH, Hightower AW, Bern C, Wu AHB (2020) Magnitude and kinetics of anti-SARS-CoV-2 antibody responses and their relationship to disease severity. Clin Infect Dis. https://​doi.​org/​10.​1093/​cid/​ciaa979
12.
Zurück zum Zitat Amrane S, Tissot-Dupont H, Doudier B, Eldin C, Hocquart M, Mailhe M, Dudouet P, Ormières E, Ailhaud L, Parola P, Lagier J-C, Brouqui P, Zandotti C, Ninove L, Luciani L, Boschi C, La Scola B, Raoult D, Million M, Colson P, Gautret P (2020) Rapid viral diagnosis and ambulatory management of suspected COVID-19 cases presenting at the infectious diseases referral hospital in Marseille, France, - January 31st to March 1st, 2020: A respiratory virus snapshot. Travel Med Infect Dis 36:101632. https://doi.org/10.1016/j.tmaid.2020.101632 Amrane S, Tissot-Dupont H, Doudier B, Eldin C, Hocquart M, Mailhe M, Dudouet P, Ormières E, Ailhaud L, Parola P, Lagier J-C, Brouqui P, Zandotti C, Ninove L, Luciani L, Boschi C, La Scola B, Raoult D, Million M, Colson P, Gautret P (2020) Rapid viral diagnosis and ambulatory management of suspected COVID-19 cases presenting at the infectious diseases referral hospital in Marseille, France, - January 31st to March 1st, 2020: A respiratory virus snapshot. Travel Med Infect Dis 36:101632. https://​doi.​org/​10.​1016/​j.​tmaid.​2020.​101632
13.
Zurück zum Zitat Edouard S, Colson P, Melenotte C, Di Pinto F, Thomas L, La Scola B, Million M, Tissot-Dupont H, Gautret P, Stein A, Brouqui P, Parola P, Lagier J-C, Raoult D, Drancourt M (2020) Evaluating the serological status of COVID-19 patients using an indirect immunofluorescent assay, France. Eur J Clin Microbiol Infect Dis:1–11. https://doi.org/10.1007/s10096-020-04104-2 Edouard S, Colson P, Melenotte C, Di Pinto F, Thomas L, La Scola B, Million M, Tissot-Dupont H, Gautret P, Stein A, Brouqui P, Parola P, Lagier J-C, Raoult D, Drancourt M (2020) Evaluating the serological status of COVID-19 patients using an indirect immunofluorescent assay, France. Eur J Clin Microbiol Infect Dis:1–11. https://​doi.​org/​10.​1007/​s10096-020-04104-2
14.
Zurück zum Zitat Kohmer N, Westhaus S, Rühl C, Ciesek S, Rabenau HF (2020) Brief clinical evaluation of six high-throughput SARS-CoV-2 IgG antibody assays. J Clin Virol 129:104480CrossRef Kohmer N, Westhaus S, Rühl C, Ciesek S, Rabenau HF (2020) Brief clinical evaluation of six high-throughput SARS-CoV-2 IgG antibody assays. J Clin Virol 129:104480CrossRef
15.
Zurück zum Zitat Meyer B, Drosten C, Müller MA (2014) Serological assays for emerging coronaviruses: challenges and pitfalls. Virus Res 194:175–183CrossRef Meyer B, Drosten C, Müller MA (2014) Serological assays for emerging coronaviruses: challenges and pitfalls. Virus Res 194:175–183CrossRef
16.
Zurück zum Zitat Ou X, Liu Y, Lei X, Li P, Mi D, Ren L, Guo L, Guo R, Chen T, Hu J, Xiang Z, Mu Z, Chen X, Chen J, Hu K, Jin Q, Wang J, Qian Z (2020) Characterization of spike glycoprotein of SARS-CoV-2 on virus entry and its immune cross-reactivity with SARS-CoV. Nat Commun 11:1620CrossRef Ou X, Liu Y, Lei X, Li P, Mi D, Ren L, Guo L, Guo R, Chen T, Hu J, Xiang Z, Mu Z, Chen X, Chen J, Hu K, Jin Q, Wang J, Qian Z (2020) Characterization of spike glycoprotein of SARS-CoV-2 on virus entry and its immune cross-reactivity with SARS-CoV. Nat Commun 11:1620CrossRef
17.
Zurück zum Zitat Marien J, Michiels J, Heyndrickx L, Kerkhof K, Foque N, Widdowson M-A, Desombere I, Jansens H, Van Esbroeck M, Arien KK (2020) Evaluating SARS-CoV-2 spike and nucleocapsid proteins as targets for IgG antibody detection in severe and mild COVID-19 cases using a Luminex bead-based assay. medRxiv preprint 2020. https://doi.org/10.1101/2020.07.25.20161943 Marien J, Michiels J, Heyndrickx L, Kerkhof K, Foque N, Widdowson M-A, Desombere I, Jansens H, Van Esbroeck M, Arien KK (2020) Evaluating SARS-CoV-2 spike and nucleocapsid proteins as targets for IgG antibody detection in severe and mild COVID-19 cases using a Luminex bead-based assay. medRxiv preprint 2020. https://​doi.​org/​10.​1101/​2020.​07.​25.​20161943
18.
Zurück zum Zitat Van Elslande J, Decru B, Jonckheere S, Van Wijngaerden E, Houben E, Vandecandelaere P, Indevuyst C, Depypere M, Desmet S, André E, Van Ranst M, Lagrou K, Vermeersch P (2020) Antibody response against SARS-CoV-2 spike protein and nucleoprotein evaluated by four automated immunoassays and three ELISAs. Clin Microbiol Infect. https://doi.org/10.1016/j.cmi.2020.07.038 Van Elslande J, Decru B, Jonckheere S, Van Wijngaerden E, Houben E, Vandecandelaere P, Indevuyst C, Depypere M, Desmet S, André E, Van Ranst M, Lagrou K, Vermeersch P (2020) Antibody response against SARS-CoV-2 spike protein and nucleoprotein evaluated by four automated immunoassays and three ELISAs. Clin Microbiol Infect. https://​doi.​org/​10.​1016/​j.​cmi.​2020.​07.​038
19.
Zurück zum Zitat Huang AT, Garcia-Carreras B, Hitchings MDT, Yang B, Katzelnick LC, Rattigan SM, Borgert BA, Moreno CA, Solomon BD, Trimmer-Smith L, Etienne V, Rodriguez-Barraquer I, Lessler J, Salje H, Burke DS, Wesolowski A, Cummings DAT (2020) A systematic review of antibody mediated immunity to coronaviruses: kinetics, correlates of protection, and association with severity. Nat Commun 11:4704CrossRef Huang AT, Garcia-Carreras B, Hitchings MDT, Yang B, Katzelnick LC, Rattigan SM, Borgert BA, Moreno CA, Solomon BD, Trimmer-Smith L, Etienne V, Rodriguez-Barraquer I, Lessler J, Salje H, Burke DS, Wesolowski A, Cummings DAT (2020) A systematic review of antibody mediated immunity to coronaviruses: kinetics, correlates of protection, and association with severity. Nat Commun 11:4704CrossRef
20.
Zurück zum Zitat Khan S, Nakajima R, Jain A, de Assis RR, Jasinskas A, Obiero JM, Adenaiye O, Tai S, Hong F, Milton DK, Davies H, Felgner PL, Prometheus Study Group (2020) Analysis of serologic cross-reactivity between common human coronaviruses and SARS-CoV-2 using coronavirus antigen microarray. BioRxiv Prepr Serv Biol. https://doi.org/10.1101/2020.03.24.006544 Khan S, Nakajima R, Jain A, de Assis RR, Jasinskas A, Obiero JM, Adenaiye O, Tai S, Hong F, Milton DK, Davies H, Felgner PL, Prometheus Study Group (2020) Analysis of serologic cross-reactivity between common human coronaviruses and SARS-CoV-2 using coronavirus antigen microarray. BioRxiv Prepr Serv Biol. https://​doi.​org/​10.​1101/​2020.​03.​24.​006544
21.
Zurück zum Zitat Ng KW, Faulkner N, Cornish GH, Rosa A, Harvey R, Hussain S, Ulferts R, Earl C, Wrobel AG, Benton DJ, Roustan C, Bolland W, Thompson R, Agua-Doce A, Hobson P, Heaney J, Rickman H, Paraskevopoulou S, Houlihan CF, Thomson K, Sanchez E, Shin GY, Spyer MJ, Joshi D, O’Reilly N, Walker PA, Kjaer S, Riddell A, Moore C, Jebson BR, Wilkinson M, Marshall LR, Rosser EC, Radziszewska A, Peckham H, Ciurtin C, Wedderburn LR, Beale R, Swanton C, Gandhi S, Stockinger B, McCauley J, Gamblin SJ, McCoy LE, Cherepanov P, Nastouli E, Kassiotis G (2020) Preexisting and de novo humoral immunity to SARS-CoV-2 in humans. Science. https://doi.org/10.1126/science.abe1107 Ng KW, Faulkner N, Cornish GH, Rosa A, Harvey R, Hussain S, Ulferts R, Earl C, Wrobel AG, Benton DJ, Roustan C, Bolland W, Thompson R, Agua-Doce A, Hobson P, Heaney J, Rickman H, Paraskevopoulou S, Houlihan CF, Thomson K, Sanchez E, Shin GY, Spyer MJ, Joshi D, O’Reilly N, Walker PA, Kjaer S, Riddell A, Moore C, Jebson BR, Wilkinson M, Marshall LR, Rosser EC, Radziszewska A, Peckham H, Ciurtin C, Wedderburn LR, Beale R, Swanton C, Gandhi S, Stockinger B, McCauley J, Gamblin SJ, McCoy LE, Cherepanov P, Nastouli E, Kassiotis G (2020) Preexisting and de novo humoral immunity to SARS-CoV-2 in humans. Science. https://​doi.​org/​10.​1126/​science.​abe1107
22.
Zurück zum Zitat Yeh H-Y, Serrano KV, Acosta AS, Buhr RJ (2016) Production of recombinant Salmonella flagellar protein, FlgK, and its uses in detection of anti-Salmonella antibodies in chickens by automated capillary immunoassay. J Microbiol Methods 122:27–32CrossRef Yeh H-Y, Serrano KV, Acosta AS, Buhr RJ (2016) Production of recombinant Salmonella flagellar protein, FlgK, and its uses in detection of anti-Salmonella antibodies in chickens by automated capillary immunoassay. J Microbiol Methods 122:27–32CrossRef
23.
Zurück zum Zitat Fourier A, Dorey A, Perret-Liaudet A, Quadrio I (2018) Detection of CSF 14-3-3 protein in sporadic Creutzfeldt-Jakob disease patients using a new automated capillary Western assay. Mol Neurobiol 55:3537–3545CrossRef Fourier A, Dorey A, Perret-Liaudet A, Quadrio I (2018) Detection of CSF 14-3-3 protein in sporadic Creutzfeldt-Jakob disease patients using a new automated capillary Western assay. Mol Neurobiol 55:3537–3545CrossRef
24.
Zurück zum Zitat Kodani M, Martin M, de Castro VL, Drobeniuc J, Kamili S (2019) An automated immunoblot method for detection of IgG antibodies to hepatitis C virus: a potential supplemental antibody confirmatory assay. J Clin Microbiol 57:e01567–18. https://doi.org/10.1128/JCM.01567-18 Kodani M, Martin M, de Castro VL, Drobeniuc J, Kamili S (2019) An automated immunoblot method for detection of IgG antibodies to hepatitis C virus: a potential supplemental antibody confirmatory assay. J Clin Microbiol 57:e01567–18. https://​doi.​org/​10.​1128/​JCM.​01567-18
25.
Zurück zum Zitat Pavia C, Wormser G (2020) COVID-19: is there a role for Western blots and skin testing for determining immunity and development of a vaccine? Diagn Microbiol Infect Dis 98:115148CrossRef Pavia C, Wormser G (2020) COVID-19: is there a role for Western blots and skin testing for determining immunity and development of a vaccine? Diagn Microbiol Infect Dis 98:115148CrossRef
Metadaten
Titel
Automated Western immunoblotting detection of anti-SARS-CoV-2 serum antibodies
verfasst von
Sophie Edouard
Rita Jaafar
Nicolas Orain
Philippe Parola
Philippe Colson
Bernard La Scola
Pierre-Edouard Fournier
Didier Raoult
Michel Drancourt
Publikationsdatum
03.03.2021
Verlag
Springer Berlin Heidelberg
Schlagwort
COVID-19
Erschienen in
European Journal of Clinical Microbiology & Infectious Diseases / Ausgabe 6/2021
Print ISSN: 0934-9723
Elektronische ISSN: 1435-4373
DOI
https://doi.org/10.1007/s10096-021-04203-8

Weitere Artikel der Ausgabe 6/2021

European Journal of Clinical Microbiology & Infectious Diseases 6/2021 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Echinokokkose medikamentös behandeln oder operieren?

06.05.2024 DCK 2024 Kongressbericht

Die Therapie von Echinokokkosen sollte immer in spezialisierten Zentren erfolgen. Eine symptomlose Echinokokkose kann – egal ob von Hunde- oder Fuchsbandwurm ausgelöst – konservativ erfolgen. Wenn eine Op. nötig ist, kann es sinnvoll sein, vorher Zysten zu leeren und zu desinfizieren. 

Umsetzung der POMGAT-Leitlinie läuft

03.05.2024 DCK 2024 Kongressbericht

Seit November 2023 gibt es evidenzbasierte Empfehlungen zum perioperativen Management bei gastrointestinalen Tumoren (POMGAT) auf S3-Niveau. Vieles wird schon entsprechend der Empfehlungen durchgeführt. Wo es im Alltag noch hapert, zeigt eine Umfrage in einem Klinikverbund.

Proximale Humerusfraktur: Auch 100-Jährige operieren?

01.05.2024 DCK 2024 Kongressbericht

Mit dem demographischen Wandel versorgt auch die Chirurgie immer mehr betagte Menschen. Von Entwicklungen wie Fast-Track können auch ältere Menschen profitieren und bei proximaler Humerusfraktur können selbst manche 100-Jährige noch sicher operiert werden.

Die „Zehn Gebote“ des Endokarditis-Managements

30.04.2024 Endokarditis Leitlinie kompakt

Worauf kommt es beim Management von Personen mit infektiöser Endokarditis an? Eine Kardiologin und ein Kardiologe fassen die zehn wichtigsten Punkte der neuen ESC-Leitlinie zusammen.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.