Longitudinal follow-up of anti-SARS-CoV-2 antibody and neutralizing antibody productions after inoculation of inactivated SARS-CoV-2 vaccine A total of 11 healthy adult volunteers of both sexes, aged 24–47 years, with a BMI of 21.5–30.0 kg/m², were enrolled in this study (Fig. 1a and Supplementary Tables S1 and S2). SARS-CoV-2 vaccine (Vero Cell), inactivated (Beijing Institute of Biological Products Co. Ltd), was administered intramuscularly into the deltoid. Volunteers were divided into two cohorts; five participants (cohort A) were vaccinated with a full dose (4 µg) of inactivated SARS-CoV-2 Vaccine (Vero Cell) on days 1 and 14, and six participants (cohort B) received a full dose of the vaccine on days 1 and 28 (Fig. 1a). One of the volunteers in group B was tested positive for anti-SARS-CoV-2 IgM and IgG right before vaccination, suggestive of potential prior infections. However, there was no record of previous positivity by nucleic acid (NA) diagnosis for COVID-19 (marked green in Fig. 1a). For all follow-up examinations, data from this individual was marked green to track any possible influences from potential prior infections. Fig. 1: Schematic workflow and SARS-CoV-2 antibody/neutralizing antibody detection after vaccination. a Schematic description of vaccine inoculation strategies, blood sample collections, and measurements. b, c Antibody positivity changes (percent positive/total) over time in cohorts A and B. Volunteers in cohort A were inoculated on days 1 and 14, and in cohort B, on days 1 and 28. Red line represents IgM changes, and black, IgG. d Neutralizing antibody titer changes in plasma of volunteers in cohorts A and B after vaccination, as well as those from convalescent individuals tested. Full size image Adverse events were monitored daily during the first 7 days after each inoculation and then self-recorded by the participants on diary cards in the following weeks. Overall, adverse reactions were mild (grades 1 or 2) and transient (Supplementary Table S3). Blood samples were collected on days 0, 7, 14, 28, 42, 56, and 90, and urine samples were collected on days 0, 14, 28, 42, and 90. Plasma samples were subjected to anti-SARS-CoV-2 IgM/IgG testing using multiple diagnostic kits, results from the most sensitive kit were used for quantification (Fig. 1b, c). Testing results from cohort A demonstrated that prior to the 2nd inoculation 0% of the participants developed anti-SARS-CoV-2 IgG, but by day 28, which was 2 weeks post the 2nd inoculation, 100% of the participants were tested positive (Fig. 1b). Overall, IgM showed up earlier than IgG, which was expected. IgG and IgM positivity decreased by day 42 and remained at relatively low levels by day 90 in cohort A. For cohort B, no one developed IgG until after 2nd inoculation. Yet by day 42, IgG positivity reached 100% (Fig. 1c) and sustained until day 56, suggesting that the vaccination protocol for cohort B was more efficacious. By day 90, IgG positivity also reduced to 50%, indicating antibody production did not sustain for a long time. We further carried out tests for SARS-CoV-2 neutralizing antibodies18 (Fig. 1d), and results also indicated that two inoculations 28 days apart (cohort B) resulted in higher protective antibody titers as compared to two inoculations with 14 days apart (cohort A). On the other hand, it appeared that anti-SARS-CoV-2 neutralizing antibody titers were overall lower than those in COVID-19 convalescent individuals as reported before3 (Fig. 1d). By 90 days, neutralizing antibody titers dramatically decreased in all volunteers (Fig. 1d). Interestingly, the individual who was antibody positive prior to vaccination was not more prone to generating neutralizing antibodies as compared to the rest of the participants, suggesting that prior potential infection might not have occurred or may not generate long-lasting protection in the perspective of neutralizing antibody production. Alterations in clinical laboratory measurements after vaccination Clinical laboratory routine tests including infection-related indices, hematologic parameters, coagulation function, blood glucose, serum lipids, cardiac function-related enzymes, electrolytes, liver, and renal function-related biomarkers, were measured to reveal safety features of the vaccine (Fig. 2a and Supplementary Tables S4 and S5). White blood cell count was significantly, yet only slightly, increased after vaccination on day 7. No differences were detectable at the following time points (Fig. 2b). To our surprise, quite consistent increases in HbA1c levels were observed in healthy volunteers, regardless of whether they belonged to cohort A or B. By day 28 post the 1st inoculation, three out of 11 individuals reached the prediabetic range (Fig. 2c). By days 42 and 90, medium HbA1c levels appeared to revert back, yet were still significantly higher than those before vaccination. Previous work has demonstrated that diabetic patients with uncontrolled blood glucose levels are more prone to develop severe forms of COVID-1919. High blood glucose levels/glycolysis had been shown to promote SARS-CoV-2 replication in human monocytes via the production of mitochondrial reactive oxygen species and activation of HIF1A20, therefore presenting a disadvantageous feature. Fig. 2: Temporal changes of clinical laboratory measurements after vaccination. a Clinical laboratory routine tests include hematologic and coagulation parameters, blood glucose-related and infection-related indices, lipid profile, cardiac enzymes, electrolytes, liver- and renal function-related biomarkers. More information could be found in Supplementary Tables S4 and S5. Laboratory test values of white blood cell count (b), HbA1c (c), potassium (d, left panel), sodium (d, right panel), APTT (e, left panel), PT (e, right panel), total cholesterol (f), total bile acid (g), eGFR (h, left panel), creatinine (h, right panel). Data points represent the values of each individual. Box plots showed the 25th, 50th (median), and 75th percentiles. Horizontal dashed lines showed upper normal limits (red) in b, c, d (left panels), e (left panel), f, h and the lower normal limits (blue) in d (left panel) and h. The P values were calculated by the Wilcoxon sign-rank test by comparing the laboratory measurements at each time with the baseline measurements. *P ≤ 0.05, **P ≤ 0.01, ***P ≤ 0.001. Full size image Serum potassium levels decreased significantly by days 28, 42, and 90 post the 1st inoculation, with one sample below the lower normal limit at day 42 (Fig. 2d, left panel). Similarly, serum sodium levels also decreased following vaccination (Fig. 2d, right panel), indicative of vaccine influences on electrolyte balance. Again, electrolyte imbalance has also been linked to COVID-1921. Coagulopathy is another COVID-19-induced clinical condition22. We found that coagulation profiles changed significantly after vaccination, in the short-term (7 days) after the 1st inoculation, coagulation profiles were leaning toward shorter Prothrombin Time (PT), whereas the long-term (28 and 42 days) effect was toward activated partial thromboplastin time (APTT) and PT prolongation (Fig. 2e). By day 90, the profiles returned back to those before vaccination (Fig. 2e). Moreover, we found elevated blood cholesterol levels at days 7, 28 after the 1st inoculation, and elevated total bile acid levels were also detected at day 7 (Fig. 2f, g). Renal dysfunction is another clinical condition linked to COVID-19, and by 28, 42, and 90 days after the first inoculation, serum creatinine levels were significantly higher than those before vaccination, resulting in reduced eGFR (Fig. 2h). Most of these clinical features have been reported to be associated with the development of severe symptoms in COVID-19 patients (Supplementary Table S6). Overall, there were no statistically significant differences between cohorts A and B, except for only a few indices (Supplementary Table S7), therefore data from two cohorts were pooled for clinical data presentation and subsequent analyses. scRNA-seq revealed dramatic alterations in gene expression of almost all immune cells after vaccination To explore the immunological features of healthy volunteers following vaccination, we performed droplet-based scRNA-seq (10× Genomics) to study transcriptomic profiles of PBMCs from volunteers belonging to either cohort A or B, before and 28 days after vaccination (Fig. 3a and Supplementary Fig. S1a). After preprocessing and low-quality cell elimination (see “Materials and methods”), we obtained 188,886 cells from all PBMC samples, among which 86,685 cells were from cohort A and 102,201 cells from cohort B. All qualified cells were integrated into the unified dataset and subjected to downstream analyses. Fig. 3: Changes in peripheral immune cell type and subtype compositions as well as gene expression before and 28 days after the 1st inoculation. a Cell-type UMAP representation of all merged samples. In total, 22 cell types were identified by cell-type-specific gene expression signatures. In total, 188,886 cells were depicted. b Dot plot for cell-type-specific signature genes. Color scale indicated expression levels and point size represented the percentage of cells per cluster/subtype expressing the corresponding gene. c UMAP representation representing cells before (blue) and after (orange) vaccination. d Heatmap of correlation amongst pseudo-bulk samples. e Percentages of specific immune cell subtypes in total PBMCs from each individual before and after vaccination. Box plot depicted sample distribution. Blue boxes represented samples before, and orange, after vaccination. P values were based on the Wilcoxon test for comparisons between groups before and after vaccination. f Box plots showed changes before and after vaccination in monocyte content from scRNA-seq data (left panel) and clinical laboratory measures (right panel). g Box plots showed changes in CD4+, CD8+ T-cell contents as well as lymphocyte (T + B + NK) contents before and after vaccination from scRNA-seq data (left 3 panels) and laboratory tests (right panel). h DEGs identified by pseudo-bulk samples before and after vaccination. i Overrepresentation analysis of HALLMARK gene sets from MSigDB demonstrating different immunological features before and after vaccination. Full size image Using graph-based clustering of uniform manifold approximation and projection (UMAP)23, Single-cell Recognition of cell types (SingleR) algorithm24, and manual annotation based on canonical gene markers, we identified 22 cell types or subtypes and performed differential expression analysis amongst all cell types (Fig. 3b and Supplementary Table S8). Cells (cell transcriptomes) from samples before (blue) and after (orange) vaccination were distinctly separated in the UMAP representation for both cohorts, which meant immunological features had changed quite drastically in almost all immune cell types detected, and consistently in all volunteers (Fig. 3c). Among the 11 pairs (before and after) of PBMC samples, 10 pairs were sequenced together and one pair was sequenced separately in a different batch. UMAP distributions were drastically similar regardless of the different batches, suggesting minimal sequencing batch effects (Supplementary Fig. S1b). Two independent batches of sequencing revealed similar changes before and after vaccination, suggesting the changes are real, whereas using the batch effect correction method (Harmony25) (Supplementary Fig. S1c–e) would result in over filtration and elimination of the real changes caused by vaccination. Moreover, sample clustering based on the Pearson Correlation coefficient of the transcriptomes indicated that samples from the two cohorts (A and B) intermingled well with each other both before and after vaccination, whereas vaccination-induced changes could clearly be observed (Fig. 3d). Therefore, to increase the statistical power, we combined the two cohorts for subsequent analyses. To reveal differences in cell-type compositions before and after vaccination, we calculated relative percentages of all cell types in PBMCs of each individual on the basis of scRNA-seq data (Fig. 3e). We observed decreases in contents of CD4+ regulatory T cells (CD4.Treg), CD8+ T cells (CD8.T), and proliferating CD8+ cells (CD8.Tprolif) after vaccination (Fig. 3e). Decreases in γδ-T cell (gd.T.Vd2) contents were also significant (Fig. 3e). In contrast, vaccination increased CD14+ classical monocyte (Mono.C) contents (Fig. 3e), consistent with clinical laboratory measurements (Fig. 3f). The overall lymphocyte contents, which included all CD4+ T cells, all CD8+ T cells, B cells, and NK cells, did not change significantly before and after vaccination, which was also confirmed by clinical laboratory measurements (Fig. 3g). We collected a published dataset from 196 COVID-19-infected patients and controls7, and analyzed our data together with that dataset. The result indicated that vaccination-induced changes in cell contents of all five different immune cell subtypes also changed in the same directions in COVID-19 patients as compared to controls, except for proliferating CD8+ T cells (Supplementary Fig. S2). To study detailed gene expression changes induced by vaccination, we merged individual samples into pseudo-bulk samples and used paired sample test to identify differentially expressed genes (DEGs) (Fig. 3h and Supplementary Table S9). Significantly upregulated genes were involved in “TNFα signaling via NF-κB”, “inflammatory responses”, and “cytokine-cytokine receptor interaction”, “IL6-JAK STAT3 signaling”, “coagulation”, “hypoxia”, which had been reported for COVID-19, while cell cycle-related pathways were downregulated (Fig. 3i). These results supported the notion that vaccination mimicked an infection6,7,8,9,10,11,12. Featured immune cell subtype-specific gene expression changes mirrored clinical laboratory alterations Prior to the elucidation of the functional heterogeneity and cell-type-specific gene expression changes between samples before and after vaccination, we grouped cells into 11 major types: (1) naive-state CD4+ T cells, (2) naive-state CD8+ T cells, (3) CD4+ helper T cells (including CD4.T, CD4.Treg, and CD4.Tprolif), (4) CD8+ cytotoxic T cells (including CD8.T, CD8B.T, and CD8.Tprolif), (5) MAIT, (6) γδ-T cells, (7) NK cells (including NK, NK proliferative), (8) B/plasmablast cells (including B cells and plasmablasts), (9) monocytes/dendritic cells (including classical mono, intermediate mono, non-classical mono, myeloid DC1, myeloid DC2, and plasmacytoid DC), (10) CD4+ terminal effector T cells, and (11) CD8+ terminal effector T cells. Following eleven major cell-type categorizations, we performed sample-level comparisons by aggregating gene expression across major cell types within each donor and then performed differential expression analysis using muscat26. We identified diferentially expressed genes (DEGs) among all major cell types (Fig. 4a and Supplementary Table S10) and conducted gene functional analysis (Fig. 4b). Echoing the clinical measurement results, genes related to “cholesterol homeostasis”, “coagulation”, and “inflammatory response” (CXCL8, CD14, IL6, and TNFRSF1B), “TNFα signaling via NF-κB” (NFKB1, NFKB2, NFKBIE, TNFAIP3, and TNFSF9) and “hypoxia” (HIF1A) were upregulated. In addition, “TGFβ signaling”, “IL2-STAT5 signaling” (IFNGR1, MAPKAPK2, and CASP3), and “IL6-JAK-STAT3 signaling”-related genes were also upregulated (Fig. 4c). To visualize which cell types were enriched for those signatures, we performed gene module scoring and displayed the scores on UMAP coordinates as well as grouped box plots (Fig. 4c and Supplementary Table S11). Interestingly, “inflammatory response” genes were highly expressed in monocytes and after vaccination further increased (Fig. 4c), suggesting monocytes were one of the major cell types participating in inflammatory responses after vaccination. In contrast, genes related to “glycolysis”, “bile acid metabolism”, and “type I interferon (IFN-α/β) response” were downregulated, consistent with our clinical data and the pathophysiology of COVID-1913 (Fig. 4d). Fig. 4: Subtype-specific differential gene expression and gene set overrepresentation analyses depicting common gene expression changes amongst different types of immune cells after vaccination. a 11 major immune cell-type-specific DEGs identified by pseudo-bulk data produced by combinations of samples before and after vaccination. Genes with logFC > 0.5 and adjust P < 0.05 were included. b Overrepresentation analysis of HALLMARK gene sets from MSigDB amongst 11 major cell types demonstrated common changes in gene sets representing altered immunological states before and after vaccination. c, d UMAP visualization colored by average expression scores (levels) based on differential enrichment pathway. Box plot depicting the expression score distribution before and after vaccination. Full size image Most common changes in multiple immune cell subtypes revealed increases in NF-κB signaling and decreases in IFN-α/β responses Given that clusters of genes changed their expression dramatically among all major cell types, we hypothesized that there might be some transcription factors serving as master regulators leading to immunological alterations. To solve the computational challenges associated with such a big dataset, we used the MetaCell algorithm27 to aggregate homogeneous groups of cells into metacells, and finally produced 1857 metacells (893 before and 964 after vaccination) to represent the whole structure of the scRNA-seq data (Fig. 5a). Those metacells were then applied to “single-cell regulatory network inference and clustering (SCENIC)”28,29 to construct the gene regulatory networks. The workflow produced a list of 157 “regulons”, which included transcription factors and their direct targets. Regulon activities were scored using AUCell to access averaged enrichment of all genes belonging to each regulon in each metacell, as well as averaged regulon gene enrichment in all 893 metacells before vaccination, and 964 metacells after vaccination. Top-ranked (most active) eight regulons upregulated and eight regulons downregulated after vaccination were identified (Fig. 5b). We selected 3 + 3 typical regulons to construct a regulatory network as presented in Fig. 5c (Supplementary Table S12). The network showed two distinct groups, one is consisted of IRF2, STAT1 and STAT2, which were downregulated after vaccination, and the other, contained RELB, NFKB2, and HIF1A, which were upregulated after inoculation. The GO terms of the upregulated network are predominantly related to lymphocyte differentiation, activation, and “Germinal Center Formation”, which suggested that T cells and B cells were activated after vaccination. In addition, NF-κB signaling was also elevated after vaccination. The downregulated network was enriched for many interferons-related pathways and Cytokine Secretion (Fig. 5d and Supplementary Table S13). This suggested that vaccination might inhibit interferon responses in the peripheral immune system, by reducing the activities of regulons STAT1, STAT2, and IRF2, which were thought to be master transcription factors driving type I and III interferon signaling30,31. Fig. 5: Identification of master regulons and their regulatory networks before and after vaccination. a Visualization for the “similarity-structure-associating” metacells on the original scRNA-seq data. Metacells were color-coded according to their cell-type annotations. The original scRNA-seq data were color-coded “blue” and “orange” to represent samples “before” and “after” vaccination, respectively. b Top panels: rank of regulons in samples before (left) and after (right) vaccination, based on Regulon Specificity Score (RSS). Bottom panels: heatmap of top-ranked regulon activities before (blue) and after (orange) vaccination based on AUCell scores. Names of the regulons are color (blue/orange) and number coded (1–8). c Network of regulons and their target genes. The table below indicated the proportion of genes within the regulons which were up- or downregulated after vaccination. d Gene functional annotation and related genes before (blue) and after (orange) vaccination. e Schematic overview of the experiment. f After treatment with IFN-α/β, PBMCs from volunteers after vaccination had reduced expression of genes associated with type I interferon responses as compared to those before vaccination. Paired Wilcoxon test was used. *P ≤ 0.05, n = 6. Full size image To confirm vaccination-induced inhibition of interferon responses revealed by scRNA-seq, we stimulated PBMCs from vaccinated individuals before and 28 days after vaccination with IFN-α/β. After 16 h of culturing and 12 h of stimulation, we used RT-qPCR to measure the relative expression of master regulators IRF2, IRF7, and STAT2. STAT2 and IRF7 were significantly downregulated after vaccination, yet IRF2 showed a trend of downregulation (Fig. 5e, f). The regulon analyses indicated that the states of the peripheral immune system after vaccination had reduced type I interferon responses, indicative of attenuated general antiviral abilities at least 28 days after the first inoculation. Vaccination-induced inflammatory responses in monocytes Recent reports have described conserved host immune response signatures to respiratory viral infections, namely the Meta-Virus Signature (MVS), which is also conserved in SARS-CoV-2 infection32,33. Higher MVS scores are associated with infection32,33. In all, 380 (158 positively- and 222 negatively contributed to MVS scores) out of 396 (161 positively- and 235 negatively contributed) genes selected for MVS measurement were detected in our dataset. To investigate host immune responses after vaccination with inactivated SARS-CoV-2, we separated the positive and negative gene sets and calculated MVS scores (Fig. 6a). The MVS scores were substantially higher after vaccination (Fig. 6b, c), suggesting that vaccination mimicked an infection. Interestingly, the positive MVS gene set was predominantly expressed in monocytes, while the negative set in lymphocytes, indicating different cell-type-specific immune responses would take place after vaccination (Supplementary Fig. S3a, b). Fig. 6: Monocytes displayed high MVS scores and MVS score-correlated pathways. a UMAP visualization colored by MVS scores. b MVS scores in pseudo-bulk samples combining all cell types showed upregulation after vaccination. c Box plots depicting the score distribution amongst 11 major immune cell types before and after vaccination. d Heatmap of correlation between MVS scores and enriched pathways of differentially expressed genes before and after vaccination. Full size image To investigate which pathways were associated with MVS-positive gene set and MVS-negative gene set, we calculated Spearman correlation among MVS gene sets scores and previously identified differentially enriched pathways using our scRNA-seq data (Fig. 6d). The most highly correlated pathway with MVS score and MVS-positive set was “Inflammatory response signaling”, which was strikingly upregulated in monocyte after vaccination, together with CD14, FPR1, C5AR1, NAMPT, NLRP3, CDKN1A, and IFNGR2. Whereas, MVS-negative set correlated well with “Cytotoxicity signature”, represented by NKG7, CCL4, CST7, PRF1, GZMA, GZMB, IFNG, and CCL3 expression, significantly decreased in many T-cell subtypes but not NK cells after vaccination (Supplementary Fig. S3c).