Autoantibodies against chemokines post-SARS-CoV-2 infection correlate with disease course

Material availability

Material used in the present study is available upon request from the lead contact and may require a Material Transfer Agreement (MTA). A key resources table is provided as Supplementary Table 8.

Study participants and ethical approvals

The Lugano COVID-19 cohort included 71 participants, diagnosed with COVID-19 at the Clinica Luganese Moncucco (CLM, Switzerland) between 8 March 2020 and 22 November 2020, who were enrolled in the study and divided into two groups, according to the severity of the acute disease. The hospitalized group included 50 participants; the outpatient group included 21 close contacts of the hospitalized group, who received only at-home care. Inclusion criteria for the hospitalized group were a SARS-CoV-2-positive nasopharyngeal swab test by quantitative PCR with reverse transcription (RT–qPCR) and age ≥ 18 yr. Inclusion criteria for the outpatient group were being a symptomatic close contact (living in the same household) of an individual enrolled in the hospitalized group and age ≥ 18 yr. Serologic tests confirmed COVID-19 positivity for all the participants (Fig. 1a and Extended Data Fig. 3a). At the 12-month visit, participants were asked to indicate the presence or absence of persisting symptoms related to COVID-19 according to a questionnaire (Supplementary Table 5). Patients who reported at least one symptom at month 12 were included in the long COVID group. The study was performed in compliance with all relevant ethical regulations and the study protocols were approved by the Ethical Committee of the Canton Ticino (ECCT): CE-3428 and CE-3960.

The Milan COVID-19 cohort included 44 participants, diagnosed with COVID-19 and hospitalized at the Humanitas Research Hospital (Milan, Italy) between 10 March 2020 and 29 March 2021, who were enrolled in the study. Inclusion criteria were a SARS-CoV-2-positive nasopharyngeal swab test by RT–qPCR and age ≥ 18 yr. Serologic tests confirmed COVID-19 positivity for the participants who were not tested by RT–qPCR. Individuals were stratified as mild or severe depending on duration of hospitalization (mild: ≤5 d; severe: ≥7 d). The study was performed in compliance with all relevant ethical regulations and the study protocols were approved by the Ethical Committee of Humanitas Research Hospital (authorization no. 738/20 and no. 956/20).

The Zurich COVID-19 cohort7 included 104 participants, diagnosed with COVID-19 at the University Hospital Zurich, the City Hospital Triemli Zurich, the Limmattal Hospital or the Uster Hospital between April 2020 and April 2021, who were included in the study and divided into two groups, according to the severity of the acute disease. The hospitalized group included 38 participants, whereas the outpatient group included 66 individuals, who received only at-home care. Inclusion criteria for the participants were a SARS-CoV-2-positive nasopharyngeal swab test by RT–qPCR and age ≥ 18 yr. At the 13-month visit, blood was collected and participants were asked by trained study physicians to indicate the presence or absence of persisting symptoms related to COVID-19. Patients who reported at least one symptom at month 13 were included in the long COVID group. The study was performed in compliance with all relevant ethical regulations and the study protocols were approved by the Cantonal Ethics Committee of Zurich (Business Administration System for Ethics Committees (BASEC) no. 2016-01440).

The healthy control cohort included 15 adult participants (≥18 yr) with self-reported absence of previous SARS-CoV-2 infection or vaccination, enrolled between November 2020 and June 2021. An additional eight pre-pandemic samples were obtained from blood bank donors (ECCT: CE-3428). Serologic tests confirmed COVID-19 negativity for all healthy controls (Fig. 1a and Extended Data Fig. 4a). These samples were used as controls for the COVID-19 convalescents (Lugano, Milano and Zurich) and HIV-1, AS, RA and SjS cohorts.

The vaccination cohort included 16 adult participants (≥18 yr) with self-reported absence of previous SARS-CoV-2 infection (confirmed by negative serologic test; Extended Data Fig. 4f) and who received two doses of mRNA-based COVID-19 vaccine37,38, enrolled on the day of first vaccine dose or earlier, between November 2020 and June 2021 (ECCT: CE-3428).

The HIV-1 and autoimmune diseases cohorts included pre-pandemic plasma samples obtained from the following participants: 24 HIV-1 positive (ECCT: CE-813)39, and 13 each with AS, RA (ECCT: CE-3065, and Ethical Committee of the Canton Zurich EK-515) or SjS (Istituto di ricovero e cura a carattere scientifico (IRCCS) Policlinico San Matteo Foundation Ethics Committee no. 20070001302).

The Lyme disease cohort included plasma samples of 27 individuals with erythema migrans (Lyme disease) and 30 healthy controls obtained at The Valley Hospital (Ridgewood, NJ, USA) and the Lyme and Tick-Borne Diseases Research Center at Columbia University Irving Medical Center (New York, NY, USA) between 2015 and 2019. All were 18–89 yr of age and all denied being immunocompromised. Lyme disease cohort: individuals had new or recent onset erythema migrans, exposure to a Lyme endemic area in the previous 30 d and received no more than 3 weeks of antibiotic treatment. Healthy control cohort: individuals reported being medically healthy, had an unremarkable physical exam and blood tests, had no signs or symptoms of infection or illness, denied having had a diagnosis and/or treatment for Lyme and/or another tick-borne disease within the past 5 yr and denied having a tick bite in the previous 6 months. The Lyme cohort samples were collected at the time of the erythema migrans and 6 months later on average. The study was performed in compliance with all relevant ethical regulations and the study protocol was approved by the New York State Psychiatric Institute Institutional Review Board (no. 6805).

Written, informed consent was obtained from all participants, and all samples were coded. No compensation was provided to the study participants. Demographic and clinical features of the cohorts are reported in Supplementary Table 2.

Blood collection, processing and storage

Blood was collected by venipuncture at approximately 6-month intervals and the PBMCs were isolated using Histopaque density centrifugation (Lugano and healthy control cohorts). Total PBMCs were aliquoted and frozen in liquid nitrogen in the presence of FCS and dimethylsulfoxide. Plasma was aliquoted and stored at −20 °C or less. Before use, plasma aliquots were heat-inactivated (56 °C for 1 h) and then stored at 4 °C. For chemotaxis assays, CD14+ monocytes and CD19+ B cells were enriched from fresh PBMCs derived from blood donors (Swiss Red Cross Laboratory; ECCT: CE-3428) through positive immunoselection (130-050-201 and 130-050-301, respectively, Miltenyi Biotec) according to the manufacturer’s instructions. After isolation, CD19+ B cells were rested overnight in RPMI-1640 medium supplemented with 10% (v/v) FBS, 1% (v/v) nonessential amino acids, 1 mM sodium pyruvate, 2 mM GlutaMAX, 50 μM β-mercaptoethanol and 50 U ml−1 penicillin/streptomycin (all from Gibco) before being used in chemotactic assays. For the other cohorts, see refs. 7,40.

ReagentsPeptides

Synthetic peptides containing the N-loop or the C-terminal sequence of human chemokines were designed and obtained (>75% purity) from GenScript. All peptides are biotinylated (biotin-Ahx) at the N terminus and amidated at the C terminus. In addition, the first 2–4 amino acids of each peptide (GS, GGS, GGGS or GGK depending on the length of the N-loop/C terminus of the chemokine) consist of a linker between the biotin and the chemokine sequence. Peptides are generally 25 amino acids long, or 22–25 amino acids when synthesis was problematic. The sequence of the IFN-α2 peptide (7–28) was based on a previously described immunoreactive epitope in patients with myasthenia gravis41, and that of the SARS-CoV-2 nucleocapsid protein (N) peptide (157–178) was described in ref. 42. An irrelevant peptide was used as negative control. The amino acid sequences of all peptides in this study are listed in Supplementary Table 3.

Proteins

CCL7, CCL20, CXCL8 and CXCL13 were synthesized using tBoc solid-phase chemistry43. CCL8 and CXCL16 were obtained from Peprotech (catalog no. 300-15 and catalog no. 300-55, respectively) or produced and purified in-house. Briefly, recombinant chemokines were expressed in E. coli, purified from inclusion bodies by immobilized-metal affinity chromatography and folded under N2 protection in an arginine-containing buffer (80 mM Tris-Cl (pH 8.5), 100 mM NaCl, 0.8 M arginine, 2 mM EDTA, 1 mM cysteine, 0.2 mM cystine) as previously described44. After recovery and concentration, the purification tag was cleaved with enterokinase, and the processed chemokine was purified by C18 reverse-phase chromatography. The SARS-CoV-2 RBD was produced and purified as described45.

Chemotaxis

The migration of primary human monocytes and B cells isolated from buffy coats or of murine preB 300.19 cells stably expressing the human chemokine receptors CCR2 (ref. 46), CCR6, CXCR1 (ref. 47) and CXCR6 (ref. 48) was assayed using 48-well Boyden chambers (Neuro Probe) with polyvinylpyrrolidone-free polycarbonate membranes with pore size of 3 µm for primary human B cells and 5 µm for the other cell types, as previously described49. Briefly, 105 primary human B cells or 5 × 104 primary human monocytes and murine preB 300.19 cells were diluted in RPMI-1640 supplemented with 20 mM HEPES, pH 7.4, and 1% pasteurized plasma protein solution (5% PPL SRK; Swiss Red Cross Laboratory). Cells were then added to the upper wells and the chemokine (with or without antibodies) to the bottom wells. After 120 min of incubation for primary human B cells and 90 min for the other cell types, the membrane was removed, washed on the upper side with PBS, fixed and stained with DiffQuik. All assays were done in triplicate, and for each well the migrated cells were counted at 100-fold magnification in five randomly selected high-power fields.

Inhibition of chemotaxis by monoclonal antibodies

Experiments were performed with monoclonal antibodies at a final concentration of 30 µg ml−1 (Extended Data Fig. 8g) or 50 µg ml−1 (Fig. 2e,g and Extended Data Fig. 8j). Baseline migration was determined in the absence of chemoattractant (buffer control).

Inhibition of chemotaxis by plasma purified IgGs

IgGs were purified from a subset of samples of the COVID-19 and healthy control cohorts using Protein G Sepharose 4 Fast Flow (Cytiva) according to the manufacturer’s instructions (plasma/resuspended beads at a 5:4 (v/v) ratio), buffer-exchanged and concentrated in PBS by Amicon Ultra-4 centrifugal filters (30-kDa cutoff, Millipore). Chemotaxis of preB 300.19 expressing CCR2 or CXCR1 was performed at a final IgG concentration of 200 µg ml−1 (IgG concentration in human serum: ~10,000 µg ml−1 (ref. 29)), in the presence of the chemokine concentration resulting in peak migration when no antibodies were added (CCL7 (100 nM), CCL8 (100 nM), CXCL8 (1 nM)) (Extended Data Fig. 8k).

ELISA

To evaluate the antibodies’ binding to chemokine peptides, 96-well plates (ThermoFisher, 442404) or 384-well plates (ThermoFisher, 464718) were coated with 50 μl (or 10 μl for 384-well plates) per well of a 2 μg ml−1 Neutravidin (Life Technologies, 31000) solution in PBS, overnight at room temperature. Plates were washed four times with washing buffer (PBS + 0.05% Tween-20 (Sigma-Aldrich)) and incubated with 50 μl (or 10 μl for 384-well plates) per well of a 50 nM biotinylated peptide solution in PBS for 1 h at room temperature. After washing four times with washing buffer, plates were incubated with 200 μl (or 50 μl for 384-well plates) per well of blocking buffer (PBS + 2% BSA + 0.05% Tween-20) for 2 h at room temperature. Plates were then washed four times with washing buffer, and serial dilutions of monoclonal antibodies or plasma were added in PBS + 0.05% Tween-20 and incubated for 1 h at room temperature. To screen for the presence of chemokine IgGs, plasma samples were assayed (unless otherwise stated) at 1:50 starting dilution followed by three fourfold serial dilutions (1:200, 1:800, 1:3,200). Monoclonal antibodies were tested at 5 μg ml−1 starting concentration followed by 11 threefold serial dilutions. Plates were subsequently washed four times with washing buffer and incubated with an antibody against human IgG secondary antibody conjugated to horseradish peroxidase (HRP) (GE Healthcare, NA933) at a 1:5,000 dilution in PBS + 0.05% Tween-20. Finally, after washing four times with washing buffer, plates were developed by the addition of 50 μl (or 10 μl for 384-well plates) per well of the HRP substrate tetramethylbenzidine (TMB) (ThermoFisher, 34021) for 10 min. The developing reaction was stopped with 50 μl (or 10 μl for 384-well plates) per well of a 1 M H2SO4 solution, and absorbance was measured at 450 nm with an ELISA microplate reader (BioTek) with Gen5 3.12 software. A positive control (broadly reactive plasma from donor CLM70) and negative control (uninfected participant) samples were included in each experiment. Since the basal average optical density likely also depends on intrinsic features of each peptide that is used to coat the ELISA plate, the presented values should be interpreted as relative rather than absolute. The area under the curve (AUC) was obtained from two independent experiments and plotted with GraphPad Prism v.9.0.2. The main findings were further confirmed by assaying subsets of samples belonging to the different groups, side-by-side on the same plates.

Lyme disease cohort

Plasma was assayed at a 1:100 starting dilution, followed by two additional fourfold dilutions (1:400 and 1:1,600) (Extended Data Fig. 10a).

Reactivity at month 6 versus 12

Experiments were performed with plasma samples from different time points side-by-side on the same plate. In Extended Data Fig. 4b, plasma was assayed at a 1:50 starting dilution, followed by four additional fivefold dilutions. RBD IgG antibody levels were measured in COVID-19 convalescents who had not received a COVID-19 mRNA vaccine between first and second visits (no vaccination) or in individuals with at least one dose of vaccine at least 10 d before blood sampling at the second visit (Fig. 1d, Extended Data Fig. 4b,c and Supplementary Table 2).

Kinetics of signature chemokine IgG antibodies

Experiments were performed with plasma samples from different time points assayed at 1:50 dilution side-by-side on the same plate, and the average optical density at 450 nm obtained from two independent experiments was plotted with GraphPad Prism v.9.0.2 (Extended Data Fig. 4e).

IgG antibodies binding to SARS-CoV-2 RBD

Experiments were performed with 96-well plates coated with 50 μl per well of a 5 μg ml−1 protein solution in PBS overnight at room temperature, and subsequently blocked and treated as described above. In this case, plasma samples were assayed at a 1:50 starting dilution either followed by seven additional threefold serial dilutions (Figs. 1a and 2a and Extended Data Figs. 3a,c,d and 6a) or followed by three additional fivefold serial dilutions (Extended Data Fig. 4b,f).

Chemokine quantification in plasma

Plasma levels of 14 chemokines were measured using the Luminex Discovery Assay—Human Premixed Multi-Analyte Kit (R&D Systems, LXSAHM-14) following the manufacturer’s instructions. Chemokines included in the panel were: CCL2, CCL3, CCL4, CCL19, CCL21, CCL22, CCL25, CXCL2, CXCL5, CXCL8, CXCL9, CXCL10, CXCL13 and CXCL16. Each sample was measured in duplicate using a Luminex FLEXMAP 3D system.

Single-cell sorting by flow cytometry

B cells were enriched from PBMCs of healthy controls or of COVID-19 convalescents 6 months after COVID-19 (participant CLM9 for CCL8 antibodies; CLM64 for CCL20 antibodies; CLM5, CLM7 and CLM33 for CXCL13 antibodies; and CLM8 and CLM30 for CXCL16 antibodies), using the pan-B-cell isolation kit according to manufacturer’s instructions (Miltenyi Biotec, 130-101-638). The enriched B cells were subsequently stained in FACS buffer (PBS + 2% FCS + 1 mM EDTA) with the following antibodies/reagents (all 1:200 diluted) for 30 min on ice: antibodies against CD20-PE-Cy7 (BD Biosciences, 335828), against CD14-APC-eFluor 780 (ThermoFisher, 47-0149-42), against CD16-APC-eFluor 780 (ThermoFisher, 47-0168-41), against CD3-APC-eFluor 780 (ThermoFisher, 47-0037-41), against CD8-APC-eFluor 780 (Invitrogen, 47-0086-42); Zombie NIR (BioLegend, 423105); as well as fluorophore-labeled ovalbumin (Ova) and N-loop peptides. Live single Zombie-NIR−CD14−CD16−CD3−CD8−CD20+Ova−N-loop-PE+N-loop-AF647+ B cells were single-cell sorted into 96-well plates containing 4 μl of lysis buffer (0.5 × PBS, 10 mM DTT, 3,000 U ml−1 RNasin Ribonuclease Inhibitors (Promega, N2615)) per well using a FACS Aria III, and the analysis was performed with FlowJo software. The CCL20 antibody sequences were obtained by sorting with a pool of 12 peptides; for all the others, a single peptide was used. The sorted cells were frozen on dry ice and stored at −80 °C.

Antibody sequencing, cloning, production and purification

Antibody genes were sequenced, cloned and expressed as previously reported50,51,52. Briefly, reverse transcription of RNA from FACS-sorted single cells was performed to obtain complementary DNA, which was then used for amplification of the immunoglobulin IGH, IGK and IGL genes by nested PCR. Amplicons from this first PCR reaction served as templates for sequence- and ligation-independent cloning into human IgG1 antibody expression vectors. Monoclonal antibodies were produced by transiently transfecting Expi293F cells cultured in Freestyle-293 Expression Medium (ThermoFisher) with equal amounts of immunoglobulin heavy and light chain expression vectors using polyethyleneimine Max (PEI-MAX, Polysciences) as a transfection reagent. After 6–7 d of culture, cell supernatants were filtered through 0.22-μm Millex-GP filters (Merck Millipore), and antibodies were purified using Protein G Sepharose 4 Fast Flow (Cytiva) according to the manufacturer’s instructions and buffer-exchanged and concentrated in PBS by Amicon Ultra-4 centrifugal filters (30-kDa cutoff, Millipore). Where indicated, the monoclonal antibody against Zika virus Z02150 was used as an isotype control.

Computational analysis of antibody sequences

Antibody sequences were analyzed using a collection of Perl and R scripts provided by IgPipeline and publicly available on GitHub (https://github.com/stratust/igpipeline)27. In brief, sequences were annotated using IgBlast53 v.1.14.0 with IMGT domain delineation system and the Change-O toolkit v.0.4.5 (ref. 54). CDR3 sequences were determined by aligning the IGHV and IGLV nucleotide sequences against their closest germlines using the blastn function of IgBlast.

SARS-CoV-2 pseudotyped reporter virus and neutralization assay

To generate (HIV-1/NanoLuc2AEGFP)-SARS-CoV-2 particles, HEK293T cells were co-transfected with the three plasmids pHIVNLGagPol, pCCNanoLuc2AEGFP and SARS-CoV-2 S as described elsewhere27,55. Supernatants containing virions were collected 48 h after transfection, and virion infectivity was determined by titration on 293TACE2 cells. The plasma neutralizing activity was measured as previously reported27,55. Briefly, threefold serially diluted plasma samples (from 1:50 to 1:328,050) were incubated with SARS-CoV-2 pseudotyped virus for 1 h at 37 °C, and the virus–plasma mixture was subsequently incubated with 293TACE2 cells for 48 h. Cells were then washed with PBS and lysed with Luciferase Cell Culture Lysis 5X reagent (Promega). Nanoluc Luciferase activity in cell lysates was measured using the Nano-Glo Luciferase Assay System (Promega) with Modulus II Microplate Reader user interface (TURNER BioSystems). The obtained relative luminescence units were normalized to those derived from cells infected with SARS-CoV-2 pseudotyped virus in the absence of plasma. The NT50 values were determined using four-parameter nonlinear regression with bottom and top constrains equal to 0 and 1, respectively (GraphPad Prism v.9.0.2). The dotted line (NT50 = 5) in the plots represents the lower limit of detection of the assay.

Model interaction between chemokine and chemokine receptor

The illustrative model in Extended Data Fig. 1a was generated from the structure of inactive CCR2 (PDB code: 5T1A)56, together with the electron microscopy structures of CCR5 and CCR6 (PDB codes: 6MEO and 6WWZ, respectively)57,58, by using the SWISS-MODEL59 server and the molecular graphics program PyMOL 2.5.0 for modeling the N and C termini of the receptor. The crystal structure of CCL8 (MCP-2) (PDB code: 1ESR)60 and the electron microscopy structure of CCR6 (ref. 58) were used to model the complex. The intracellular residues were removed for clarity.

Statistical analysisSample size definition

No statistical methods were used to pre-determine sample sizes but our sample sizes are similar to those reported in previous publications7.

Tests for statistical significance

Upon testing of parametric assumptions (Kolmogorov–Smirnov test for normality and Hartley’s Fmax test for homoskedasticity), statistical significance between two groups was determined using parametric paired two-tailed Student’s t-test, or nonparametric two-tailed Mann–Whitney U-tests (unpaired samples), or Wilcoxon signed-rank test (paired samples). Statistical significance between more than two groups was evaluated using Kruskal–Wallis test (followed by Dunn multiple comparisons), one-way analysis of variance (ANOVA) (followed by Tukey multiple comparisons) or two-way repeated measures ANOVA (followed by Šídák multiple comparisons), as described in the figure legends. Statistical significance of the signature chemokines (CCL19, CCL22, CXCL17, CXCL8, CCL25, CXCL5, CCL21, CXCL13 and CXCL16) was also confirmed when applying the Bonferroni criterion to guarantee a familywise level of significance equal to 0.05. Statistical significance from a 2 × 2 contingency table was determined with Fisher’s exact test. Correlations were assessed using Pearson correlation analysis. A P value of less than 0.05 was considered statistically significant. Data and statistical analyses were performed with GraphPad Prism v.9.0.2. Data collection and analysis were not performed blind to the conditions of the experiments.

t-SNE

t-SNE analysis was performed using the Rtsne R package v.0.15 (https://CRAN.R-project.org/package=Rtsne) using the AUC values for all chemokines. The theta parameter for the accuracy of the mapping was set to zero in all cases for exact t-SNE.

Clustering

Hierarchical clustering was created using the hclust R function v.4.1.1. Clustering analysis was performed using the correlation as distance and Ward’s method for agglomeration. Heatmaps were created with either GraphPad Prism v.9.0.2 (Fig. 1a and Extended Data Fig. 2d) or the Pretty Heatmaps (pheatmap) R package v.1.0.12 (Extended Data Fig. 10b). In Extended Data Fig. 10b, each column containing a distinct chemokine was scaled with the scaling function provided by R, which sets the mean and the standard deviation to 0 and 1, respectively.

Logistic regression and additional analyses

Logistic regression was performed using the GLM (Generalized Linear Models) function provided by the R package v.4.1.1. To identify which variables to include in the analysis, AUCs were ranked according to the P value obtained with a Mann–Whitney–Wilcoxon nonparametric test on the Lugano cohort. The first N variables minimizing the Akaike information criterion were then used in the fitting. Furthermore, the same set of variables was used to perform the fitting with the Milan and Zurich cohorts. In each plot, values from 0 to 0.5 and from 0.5 to 1 on the y axis represent the assignment of individuals to the A and B groups (of a Prediction A versus B; see gray backgrounds), respectively. On the x axis, samples are divided into the two groups and subsequently ordered according to sample identity as shown in Supplementary Table 2. Dots in the gray area represent individuals that are assigned to the correct group. We additionally performed chi-tests considering covariates that are known to influence COVID-19 severity (demographics (sex and age) and comorbidities (diabetes and cardiovascular diseases)) and found that none of them was significantly different between groups. Race/ethnicity was not analyzed because the cohort is nearly 100% White; similarly, immune deficiency was rare and was not considered. Logistic regression analysis using the combination of these covariates (age, sex, diabetes and cardiovascular diseases) allowed proper assignment with accuracies of 74.6% (COVID-19 severity; outpatient versus hospitalized) and 68.3% (long COVID; no long COVID versus long COVID). Notably, the accuracy using chemokine antibody values is even better (77.5% (COVID-19 severity) and 77.8% (long COVID)). These analyses are shown in Supplementary Table 9.

Reporting summary

Further information on research design is available in the Nature Portfolio Reporting Summary linked to this article.

留言 (0)

沒有登入
gif