key: cord-0996559-ry4mgywy authors: Bieberich, Florian; Vazquez-Lombardi, Rodrigo; Yermanos, Alexander; Ehling, Roy A.; Mason, Derek M.; Wagner, Bastian; Kapetanovic, Edo; Di Roberto, Raphael Brisset; Weber, Cédric R.; Savic, Miodrag; Rudolf, Fabian; Reddy, Sai T. title: A single-cell atlas of lymphocyte adaptive immune repertoires and transcriptomes reveals age-related differences in convalescent COVID-19 patients date: 2021-02-15 journal: bioRxiv DOI: 10.1101/2021.02.12.430907 sha: b4eef48af7862d5b39e73ad4e3cc6c26eac5125c doc_id: 996559 cord_uid: ry4mgywy COVID-19 disease outcome is highly dependent on adaptive immunity from T and B lymphocytes, which play a critical role in the control, clearance and long-term protection against SARS-CoV-2. To date, there is limited knowledge on the composition of the T and B cell immune receptor repertoires [T cell receptors (TCRs) and B cell receptors (BCRs)] and transcriptomes in convalescent COVID-19 patients of different age groups. Here, we utilize single-cell sequencing (scSeq) of lymphocyte immune repertoires and transcriptomes to quantitatively profile the adaptive immune response in COVID-19 patients of varying age. We discovered highly expanded T and B cells in multiple patients, with the most expanded clonotypes coming from the effector CD8+ T cell population. Highly expanded CD8+ and CD4+ T cell clones show elevated markers of cytotoxicity (CD8: PRF1, GZMH, GNLY; CD4: GZMA), whereas clonally expanded B cells show markers of transition into the plasma cell state and activation across patients. By comparing young and old convalescent COVID-19 patients (mean ages = 31 and 66.8 years, respectively), we found that clonally expanded B cells in young patients were predominantly of the IgA isotype and their BCRs had incurred higher levels of somatic hypermutation than elderly patients. In conclusion, our scSeq analysis defines the adaptive immune repertoire and transcriptome in convalescent COVID-19 patients and shows important age-related differences implicated in immunity against SARS-CoV-2. T and B lymphocytes are crucial for protection from SARS-CoV-2 infection, viral clearance and the formation of persisting antiviral immunity 1, 2 . Yet, adaptive immune responses have also been implicated in contributing to immunopathology during COVID-19, with higher mortality rates in elderly individuals 3-6 . However, the exact determinants of a successful adaptive immune response against SARS-CoV-2 and its variability between different age groups remain to be fully elucidated. Lymphocytes express either T cell receptors (TCR) or B cell receptors (BCR), which possess a highly diverse pair of variable chains [variable alpha (Vα) and beta (Vβ) for TCR and variable light (V L ) and heavy (V H ) for BCR] that are able to directly engage with antigen (e.g., viral proteins or peptides). Diversity in these variable chains are generated by somatic recombination of V-, D-and J-gene germline segments and along with combinatorial receptor chain pairing and somatic hypermutation (BCR only) results in an estimated human TCR and BCR diversity of 10 18 and 10 13 , respectively 7, 8 . Upon encountering cognate antigens, lymphocytes are phenotypically activated and undergo massive proliferation, also referred to as clonal selection and expansion. Deep sequencing of TCRs and BCRs has become a powerful strategy to profile the diversity of immune repertoires and to reveal insights on clonal selection, expansion and evolution (somatic hypermutation in B cells) [9] [10] [11] [12] and has been instrumental in studying long term effects following vaccination, infection and ageing [13] [14] [15] [16] [17] . In the context of COVID-19, immune repertoire sequencing has shown diminished TCR repertoire diversity and BCR isotype switching and respective expansion during early disease onset 18 . In recent years, single-cell sequencing (scSeq) of transcriptomes has progressed substantially through the development and integration of technologies such as cell sorting, microwells and droplet microfluidics 19,20 ; most notably commercial systems like those of 10X Genomics have been established and are providing standardized protocols for wider implementation of scSeq. To find interactions across multiple genes and cells, analysis and visualisation of this high dimensional single-cell data is facilitated by clustering and nonlinear dimensionality reduction algorithms [e.g., t-distributed stochastic neighbor embedding (t-SNE) or Uniform Manifold Approximation and Projection (UMAP)] 21, 22 . scSeq of transcriptomes has been used extensively to profile the gene expression signatures of T and B cells to identify novel cellular subsets and phenotypes as well as their response to vaccination, infection and cancer [23] [24] [25] [26] . Furthermore, clustering with scSeq data enables the unbiased identification of cellular states and analyses of the broad continuum of T and B cell populations as well as their differentiation trajectories 27 . In the context of patients with severe symptoms of COVID-19, scSeq has revealed a dysfunctional T cell response of interferon expression combined with elevated levels of exhaustion 28 . In addition to transcriptome sequencing, a major advantage of scSeq is that it also enables information on the native pairing of TCR Vα and Vβ chains and BCR V L and V H chains [29] [30] [31] [32] , which was not previously possible with the standard bulk sequencing of lymphocytes as these receptor chains are expressed as unique transcripts from separate chromosomes 33 . Coupling TCR or BCR sequence to the transcriptome within an individual cell enables phenotypic analyses of a clonal population of lymphocytes and their dynamics [34] [35] [36] . scSeq of transcriptomes and immune repertoires in COVID-19 patients with severe symptoms has shown a high level of clonal expansion in specific T cell subsets (Th1, Th2, and Th17) and preferential germline gene usage in clonally expanded B cells 28, 34, 37 ; while a more recent study found a positive correlation between clonal expansion of effector-like CD8+ T cells and disease severity 38 . An important question that remains to be answered is whether there are age-related differences in mounting a successful adaptive immune response against SARS-CoV-2. Here, we perform scSeq on the immune repertoires and transcriptomes of T and B cells derived from eight convalescent COVID-19 patients of two different age groups (mean ages = 31 and 66.8 years) at one month of convalescence following mild to moderate disease. We observed preferential clonal expansion of effector CD8+ T cells across all patients, although a significantly higher CD8-to-CD4 T cell ratio was detected in young patients of our cohort. Further, clonally expanded B cells in young patients displayed significantly higher levels of somatic hypermutation and an increased immunoglobulin (Ig) class-switching compared to clonally expanded B cells from older patients. Our analyses serve as a valuable resource for future scSeq characterization of SARS-CoV-2 adaptive immunity and highlight important age-related differences in the adaptive immune status of convalescent COVID-19 patients. We performed scSeq of immune receptor repertoires and transcriptomes of lymphocytes from convalescent COVID-19 patients to characterize the adaptive immune response against SARS-CoV-2. For this purpose, we selected eight patients enrolled in the SERO-BL-COVID-19 clinical study 39 Table 1 ). Since COVID-19 often affects older patients more severely 40 , subjects were divided into two groups according to their age, namely Group 1 (mean = 66.75 ± 6.9 years) and Group 2 (mean = 31 ± 5.9 years), with the aim of investigating potential differences in their responses against SARS-CoV-2. In addition to older age, significant differences in Group 1 versus Group 2 included elevated IgA/IgG SARS-CoV-2-specific antibody titers and an increased duration of COVID-19 symptoms ( Fig. 1b and Supplementary Fig.1 ). Patients from Group 1 also experienced an increased severity of COVID-19 symptoms relative to Group 2 ( Supplementary Table 1 ). Despite increased symptom duration in the older cohort, correlation of this parameter with age was only modest (R 2 = 0.4647), likely reflecting the small sample size ( Supplementary Fig. 1c ). To profile patient lymphocytes, we isolated peripheral blood mononuclear lymphocytes (PBMC) from blood and purified T cells and B cells by negative immunomagnetic enrichment. Plasma cells (PCs) were depleted from PBMC samples prior to this step for scSeq in a companion study (Ehling et al., manuscript in preparation), and thus were excluded from our analyses. After purification, T cells and B cells underwent the 10X genomics protocol for scSeq 5' library preparation, which included gel encapsulation single-cell barcoding of mRNA, followed by cDNA generation through polydT reverse transcription. Finally, after full-length V(D)J segment enrichment, construction of TCR and BCR V(D)J and transcriptome sequencing libraries was done according to the V(D)J enrichment and 5' library construction kits, respectively. Deep sequencing of immune repertoires and transcriptomes was performed using the Illumina NovaSeq with paired-end 26 x 91 bp cycles per read. For TCR and BCR V(D)J and transcriptome libraries, we recovered on average 20.000 and 10.000 reads per cell, respectively ( Fig. 1c ). Bioinformatic filtering was performed to exclude the following: Cell doublets, cells with a very low or high number of genes, and T cells with no detectable expression of CD8 and CD4 (see Methods), which resulted in the identification of 30,096 cells in total from all eight patients. Cells were then split into CD8+ T cell ( Fig. 2a , 7 ,353 cells), CD4+ T cell ( Fig. 2b , 8 ,334 cells) and B cell ( Fig. 2c , 14 ,409 cells) datasets. In order to reduce the dimensionality of the data, while preserving the global structure, we used UMAP for better visualisation and interpretation purposes 41 2g and 2h ). Interestingly, pseudotime analysis of B cell data not only showed naïve-MZ-memory and naïve-MZ-activated trajectories, but also a third MZ-memory-activated trajectory that suggests the presence of reactivated memory B cells, possibly through antigen encounter ( Fig. 2i ). Having defined the major T cell and B cell subsets from pooled patient data, we next compared their proportions across patients ( Figs. 2j-l ) and between different age groups ( Supplementary Fig. 3 ). We found that young patients had a significantly higher CD8-to-CD4 T cell ratio relative to older ones ( Supplementary Fig. 3a ) , which may reflect a previously reported age-dependent difference 42 . Interestingly, despite this reduction, there was a trend that older patients had a higher proportion of effector CD8+ T cells relative to their younger counterparts ( Supplementary Fig. 3b ). While this difference was not significant, it is consistent with the increased symptom severity experienced by older patients ( Supplementary Table 1 ), a feature that has been associated with elevated proportions of effector/exhausted CD8+ T cells in the periphery 28 . Of note, we found that older patients had a small but significant increase in CD4+ Tregs compared to young patients ( Supplementary Fig. 3h ), and that increased proportions of MZ B cells occurred in two of the older patients ( Supplementary Fig. 3l ). Taken together, our data highlights the diversity of elevated responses in specific patients across age groups, as exemplified by individuals with a high abundance of effector CD8+ T cells (e.g., Pt-2 and Pt-3) and/or activated B cells (e.g., Pt-2, Pt-5 and Pt-8). Single-cell profiling of immune receptor repertoires identifies highly expanded TCR and BCR clonotypes We next determined the clonal expansion levels of T cells and B cells in convalescent COVID-19 patients by quantifying the number of cells expressing unique TCRs ( Fig. 3a ) or BCRs ( Fig. 3c ) (clonotype definition in the methods section). We found substantial heterogeneity in T cell clonal expansion levels across patients, with the highest number of expanded TCR clonotypes occurring in four patients, namely Pt-2 and Pt-3 (Group 1), Pt-7 and Pt-8 (Group 2). Within these patients, Pt-2 displayed the largest amount of expanded TCR clonotypes, which is consistent with the high abundance of effector CD8+ T cells in this subject ( Fig. 2g ). Analysis of TCRα and TCRβ germline V-gene usage in the ten most expanded clonotypes per patient revealed a frequent occurrence of TRBV20-1 (7 out of 8 patients) and TRAV-29/DV5 genes (5 out of 8 patients), though pairing of these germline genes was not observed ( Fig. 3b and Supplementary Fig. 4 ). In agreement with the overall higher expansion of CD8+ effector over CD4+ effector T cell subsets ( Figs. 2g and 2h ), we found that the vast majority (85%) of the ten most expanded TCR clonotypes per patient originated from CD8+ T cells ( Supplementary Fig. 5 ). Based on this observation, we genotyped patient HLA class I alleles by means of amplicon deep sequencing ( Supplementary Table 2 ). We found that the two patients with the highest levels of T cell clonal expansion (i.e., Pt-2 and Pt-8) shared the HLA-A*0201 allele, as well as a number of TRBV and TRAV genes in their most expanded clonotypes, which indicates a possible convergence towards germlines that may be related to SARS-CoV-2 specificity. Analysis of single-cell BCR repertoire sequencing data revealed that highly expanded BCR clonotypes occurred more frequently in younger patients, for example in Pt-5, Pt-6 and Pt-8 ( Fig. 3c ). This is an unexpected finding, particularly as older patients in our cohort displayed significantly higher SARS-CoV-2-specific IgA/IgG titers in serum ( Supplementary Figs. 1e and 1f ). Thus, this suggests that older patients may harbor a higher diversity of relatively unexpanded SARS-CoV-2-specific B cells. Supporting this observation, we found that older patients had a wider range of heavy chain complementarity determining region 3 (CDR3H) lengths relative to younger ones, indicating a possible larger degree of variability in their antibody paratopes ( Supplementary Fig. 6 ). Analysis of heavy chain and light chain germline V-gene pairing in the ten most expanded BCR clonotypes per patient revealed a frequent occurrence of the IGHV-3-23 / IGKV-3-20 pairing (7 out 8 patients) ( Fig. 3d and Supplementary Fig. 7 ). However, as this pairing is the most frequently found in healthy cohorts 43, 44 , such antibodies may not necessarily be enriched for SARS-CoV-2 specificity. To further characterize BCR repertoires across different levels of clonal expansion we divided clonotypes into additional subsets: unexpanded (1 cell per clonotype), expanded (2-4 cells per clonotype) and highly expanded (≥5 cells per clonotype) and assessed their levels of somatic hypermutation (SHM). We found that the degree of SHM largely correlated with clonal expansion, with expanded and highly expanded clonotypes having higher SHM (i.e., more divergent from their germline V-genes) than unexpanded ones ( Fig. 3e ). Strikingly, highly expanded BCR clonotypes from young patients had significantly higher SHM levels compared to older patients, potentially indicating more efficient affinity maturation had occurred in response to SARS-CoV-2 antigens ( Fig. 3f ). Finally, we examined the distribution of Ig isotypes across clonal expansion groups. As expected, IgM was the most frequent isotype in unexpanded BCR clonotypes, with the proportion of this isotype being reduced in expanded BCR clonotypes (2-4 cells) of all patients. Conversely, the proportions of IgG and IgA isotypes in expanded clonotypes increased for all patients, thus indicating class-switching in response to clonal expansion. Analysis of Ig isotype distribution in highly expanded BCR clonotypes (≥5 cells) revealed that a subset of patients harbored a vast majority of class-switched IgA (Pt-5, Pt-6 and Pt-7) or IgG (Pt-4). Notably, we found that Ig isotype class-switching in highly expanded clonotypes was correlated with SHM levels across patients (Fig. 3e) , highlighting the temporal connection between affinity maturation and class-switching processes in the germinal center 45 . We next analysed single-cell BCR sequencing data to assess SHM levels in different B cell subsets. BCRs from memory B cells showed the highest levels of SHM across patients, while BCRs extracted from naïve and activated B cells displayed similarly low median SHM values. Notably, however, activated B cells expressed a larger number of high-SHM outliers than naïve B cells, suggesting ongoing affinity maturation in this subset ( Fig. 5c ) . Mapping of Ig isotype information onto the B cell transcriptional UMAP space, revealed an even distribution of IgM expression across B cell subsets, rare occurrence of IgD-expressing B cells and, most notably, confinement of class-switched IgG-and IgA-expressing B cells to the memory and MZ B cell regions ( Fig. 5d ). Finally, assessment of Ig isotype distribution across patients and B cell subsets revealed that as expected the vast majority of naïve B cells expressed the IgM isotype ( Fig. 5e ) , with minimal levels of class-switching observed in activated B cells but prominent class-switching to IgG and IgA isotypes in the memory B cell compartment across all patients ( Fig. 5e ). Together our results indicate ongoing transition of clonally-expanded B cells into antibody-producing plasma cells, as well as high levels of SHM and Ig isotype class-switching in the memory B cells of convalescent COVID-19 patients. Motivated by the high levels of CD8+ T cell clonal expansion and activation observed in convalescent COVID-19 patients, we further analyzed single-cell TCR repertoires for potential SARS-CoV-2 specificity. To this end, we applied GLIPH2, an algorithm developed by M. Davis and colleagues that clusters TCRs with a high probability of recognizing the same epitope into specificity groups (based on conserved motifs and similarity levels in CDR3β) 52 . In addition, the provision of HLA typing data enables the prediction of HLA restriction in specific TCR clusters. Analysis of 23,010 paired TCRα and TCRβ sequences derived from the CD8+ T cells of eight patients led to the identification of a total of 552 specificity groups with attributed HLA restriction (seven alleles). We observed distinct proportions of shared specificity groups between pairs of patients, with Pt-1:Pt-8, Pt-3:Pt-7 and Pt-1:Pt-7 showing the highest overlap ( Fig. 6a ) . Furthermore, the vast majority of clusters were attributed with HLA-A*01:01, HLA-A*03:01, HLA-B*13:02 or HLA-C*03:04 restriction, and HLA-A*02:01, A*24:02 and C*04:01 were attributed to less than 15% of TCR clusters ( Fig. 6b ). While some of these clusters may be defined by SARS-CoV-2 specificity, it is difficult to exclude reactivity to common human viruses (e.g., CMV, EBV). To further investigate potential for SARS-CoV-2 specificity, we analyzed the sequences of known HLA-A*02:01-restricted SARS-CoV-2-specific, CMV-specific and EBV-specific TCRs alongside those derived from patients expressing the HLA-A*02:01 allele (i.e., Pt-2, Pt-4 and Pt-8) ( Supplementary Tables 3 and 4 ). GLIPH2 identified 35 unique patient TCR sequences that clustered together with known SARS-CoV-2-specific TCRs, with the great majority originating from Pt-8 ( Fig. 6c and Table 1 ). Thus, such TCRs represent candidates for mediating CD8+ T cell immunity against SARS-CoV-2 infection. Our sample size of eight patients (four per age group) is small and reduces the number of conclusions that we can confidently make from the observed data. Nevertheless, single-cell data offers a deeper characterization of each patient than normal bulk transcriptome and repertoire studies. Furthermore, while the time between symptom onset and sample collection is highly uniform across patients, the time between symptom resolution and sample collection is significantly shorter in the old patient group due to prolonged symptom duration. This is an important variable that should be considered when interpreting the findings presented here. Here we apply scSeq for in-depth immune repertoire and transcriptomic analysis of T cells and B cells patients 28, 53 . For example, effector tissue-resident CD8+ T cells from bronchoalveolar lavage fluid were found to be highly clonally expanded in convalescent COVID-19 patients that experienced moderate but not severe infection 53 . In addition, scSeq of PBMCs revealed increases in cytotoxic effector CD4+ and CD8+ T cell subsets in non-severe convalescent COVID-19 patients only 28 . In our study, we observed high levels of clonal expansion of the effector CD8+ T cell subset but less evident expansion of CD4+ T cell subsets. Importantly, however, differential gene expression analysis revealed that both highly clonally pandemic but not prior to it 55, 56 . In line with these findings, our analysis of clonal expansion in lymphocyte subsets suggest a key role of CD8+ effector T cells in the clearance and protection against SARS-CoV-2 in patients with moderate disease. Notably, the fact that younger patients in our cohort had significantly higher CD8-to-CD4 T cell ratios might have contributed to reduced symptom duration relative to older patients. In this context, we predict that methods for the identification of CD8-derived SARS-CoV-2-specific TCRs including functional assays 54, 55 and the application of motif clustering 56 or machine learning 57 to TCR repertoire data, as well as methods for the identification of their corresponding epitopes 58,59 will become increasingly important tools for monitoring SARS-CoV-2 immunity. In contrast to T cells, we found modest levels of B cell clonal expansion that was not exclusively restricted to a particular subset but spanned activated, memory and MZ B cells. These low levels of B cell clonal expansion are in contrast to the high IgG and IgA serum titers found in all patients of our cohort, particularly those in the older patient group. This suggests that the B cell compartment may experience clonal contraction at one month of convalescence, with the disconnect from serum titers of IgG likely explained by the long half-lives of secreted IgG (~3 weeks) 60 . It should also be noted that antibody-producing plasma cells, which were not included in our analysis, may display clonal expansion levels that are more congruent with the observed levels of SARS-CoV-2-specific antibodies in serum. Although rare, highly expanded B cell clones showed elevated markers of plasma cell transition and activation, thus indicating possible ongoing differentiation into plasma cells, albeit at low levels, at one month after symptom onset. Analysis of BCR repertoires and transcriptomes revealed that the highest levels of SHM and Ig class-switching occurred in the memory B cell subset. This finding is consistent with the generation of germinal center-derived memory B cells following antigen encounter 61, 62 . Similarly, SHM and Ig class-switching levels were directly correlated with B cell clonal expansion. Remarkably, we found that highly expanded B cell clones from the young patient group had significantly higher SHM levels (median = 6.7% divergence from germline) than those derived from the old patient group (median = 2.5% divergence from germline). This occurred despite significantly higher serum titers of SARS-CoV-2-specific antibodies in the old patient group, and suggests that more effective affinity maturation may occur in younger patients. Initial studies characterizing SARS-CoV-2-specific antibodies reported low levels of SHM, with median divergence from germline ranging from 0.7-2% in convalescent patients of varied symptom severity analyzed at 20-40 days following symptom onset [63] [64] [65] . Notably, a recent report has described ongoing affinity maturation of SARS-CoV-2-specific antibodies at six months following symptom onset in non-severe patients, with SHM levels rising to 3% divergence from germline 2 . Interestingly, the cited study provides evidence of viable SARS-CoV-2 antigen in the gut of such patients, which has been proposed as a source of antigen for ongoing affinity maturation linked to elevated IgA serum titers. It is thus unclear whether increased SHM levels found in the young patients of our cohort are the result of a better capacity for affinity maturation upon initial antigen encounter or of ongoing affinity maturation resulting from longer exposure to antigen following symptom resolution. The clear dominance of IgA class-switched BCRs expressed by highly expanded B cells in 3 out of 4 patients in the young group appears to support the latter. In conclusion, our in-depth characterization integrating single-cell immune repertoire and transcriptome profiling of T and B cells represents a valuable resource to better understand the adaptive immune response and age-related differences in convalescent COVID-19 patients with moderate disease. The quality and concentrations of transcriptome (i.e., cDNA), TCR and BCR libraries were determined using a fragment analyzer (Agilent Bioanalyzer) at specific steps of library preparation, as recommended in the 10x Genomics scSeq protocol (CG000086 manual, RevM). Following multiplexing (Chromium i7 Multiplex Kit, #PN-120262, 10x Genomics), transcriptome libraries were treated with free adapter blocking (FAB) reagent to prevent index switching (#20024144, Illlumina). Paired-end sequencing of multiplexed transcriptome libraries was performed using a NovaSeq 6000 sequencer (Illumina) and SP100-cycle kit (#20027464, Illumina). TCR and BCR libraries were multiplexed, FAB-treated and paired-end-sequenced using a second SP100-cycle kit in a separate run. HLA class I transcripts were amplified and deep-sequenced from two overlapping RT-PCR reactions flanking exons 2 (PCR 1) or 3 (PCR 2) using barcoded primers designed to target conserved regions ( Supplementary Table 5 ) Reads from transcriptome scSeq (FASTQ format) were aligned to the GRCh38 reference human genome and output as filtered gene expression matrices using the 10x Genomics Cell Ranger software (version 3.1.0). Subsequent data QC and analysis was performed using R (version 3.6.2) and the Seurat package (version 3.1.5). QC steps consisted of the exclusion of TCR and BCR genes (prevention of clonotype influence on subsequent clustering), the exclusion of cells with lower than 150 or greater than 3500 genes (low quality cells), and the exclusion of cells in which more than 20% of UMIs were associated with mitochondrial genes (reduction of freeze-thaw metabolic effects) 67 . Patient datasets were merged into a Seurat object list using the merge and SplitObject function. Each patient dataset was then separately normalised using SCTransform . Variable integration features (3, 000) were calculated using the SelectIntegrationFeatures function from the R package Seurat 68 and setting them as variable features after merging the normalised patient datasets. Principal component analysis for dimensionality reduction was performed using the RunPCA function with up to 50 principal components. Potential batch effects between patient samples were addressed with the Harmony R package (version 1.0) using the RunHarmony function 69 . Finally, unsupervised clustering was performed using the FindNeighbours and FindClusters functions. Non-linear dimensionality reduction using the RunUMAP function was performed using the first 50 principal components to generate the final UMAP visualization of cell clusters. Initial T and B cell separation was performed by mapping of TCR and BCR (VDJ) cell-specific barcodes onto the scSeq transcriptome dataset. Double attribution of TCR and BCR to the same cell (i.e., barcode) was used to identify and exclude doublets. Separation of CD8+ and CD4+ T cells was performed using the WhichCells function, from the R package Seurat, based on the singular expression of CD8A and CD4, respectively. Additional filtering of B cells was done by discarding all B cells that showed expression of CD3E or SDC1 as well as excluding B cells whose cellular barcodes occured in the Plasma cell BCR (VDJ) cell barcodes (data not shown). The expression of specific markers in identified clusters was determined using the FindAllMarkers function using the Wilcoxon Rank Sum test. Cluster-specific markers were thresholded by having a log2(fold-change) greater than 0.25 between cells in the respective cluster and remaining cells; with marker expression occurring in at least 25% of cells in the cluster. Clusters were then attributed with specific cell states based on the expression of canonical markers Differentially expressed genes between two groups of cells were identified using the FindMarkers function. Genes were thresholded by being expressed in more than 50% of the cells and by having a log2(fold-change) greater than 0.5 between cells of the different groups using the Wilcoxon Rank Sum test. TCR and BCR reads in FASTQ format were aligned with the VDJ-GRCh38-alts-ensembl reference using the 10x Genomics Cell Ranger VDJ software (version 3.1.0). This generated single-cell VDJ sequences and annotations such as gene usage, clonotype frequency and cell-specific barcode information. As a QC step, only cells with one productive alpha and one productive beta chain (T cells) or with one productive heavy and one productive light chain (B cells)were retained for downstream analysis. Clonotype definition was adjusted to count all sequences as clonal if they met the following criteria: (1) Same V and J gene usage in both chains, (2) Same CDR3 length in both chains and (3) 80% amino acid sequence similarity in the CDR3 region of the TCRβ (T cells) or BCR heavy chain (B cells). Shared cellular barcode information between TCR/BCR (VDJ) scSeq and transcriptome scSeq data was used to project TCR and BCR clonotypes onto the UMAP plots (colour-coded by clonal expansion level). SHM levels in individual BCR clonotypes were determined using the change-o toolkit from the Immcantation portal as a wrapper to run IgBlast on the Cell Ranger VDJ output. The IgBlast output enabled assessment of germline similarity of single-cell BCR (VDJ) sequences. Germline identity was used as a proxy for somatic hypermutation levels and was calculated from alignments of BCR clonotypes with their corresponding VH and VL germline sequences. GLIPH2 clusters TCRs into specificity groups predicted to share the same antigen specificity based on sequence similarity 52 . We used this algorithm to cluster TCRs from HLA-A*0201 patients (Pt-2, Pt-4 and Pt-8) together with known SARS-CoV-2 binders as well as CMV and EBV binders, which were also from HLA-A*0201 background (obtained from VDJdb database). Specificity groups that were reported by GLIPH2, were filtered for groups that were significant according to the Fisher's Exact test (significance level < 5%) and contained at least one patient TCR and one TCR of known specificity (i.e., SARS-CoV-2, EBV or CMV). Specificity groups were identified with either global (0-1 amino acid differences in CDR3β) or local similarities (CDR3β share a common motif that is rare in the reference dataset). CD4 expressing clonotypes were filtered out. Pseudotime and trajectory inference was applied to scSeq transcriptome data using the slingshot function with default parameters from the Slingshot package in R 70 . The naive cluster from each CD8+ T cell, CD4+ T cell and B cell subgroup was set as the starting point for the minimum spanning tree. The previously generated UMAP clustering was set as the cellular embedding on which Slingshot performed trajectory inference computation. The data analysis pipeline followed the standard procedures as outlined in Cell Ranger and Seurat documentations. Custom scripts and functions for easier downstream analysis and visualisation purposes are available upon request. Biobase ( Immunological memory to SARS-CoV-2 assessed for up to 8 months after infection Evolution of antibody immunity to SARS-CoV-2 Targets of T Cell Responses to SARS-CoV-2 Coronavirus in Humans with COVID-19 Disease and Unexposed Individuals Potent neutralizing antibodies against multiple epitopes on SARS-CoV-2 spike Humoral and circulating follicular helper T cell responses in recovered patients with COVID-19 Adaptive immunity to SARS-CoV-2 Understanding the human antibody repertoire Overview of methodologies for T-cell receptor repertoire analysis Commonality despite exceptional diversity in the baseline human antibody repertoire High frequency of shared clonotypes in human B cell receptor repertoires Human T cell receptor occurrence patterns encode immune history, genetic background, and receptor specificity Systems Analysis Reveals High Genetic and Antigen-Driven Predetermination of Antibody Repertoires throughout B Cell Development Immunosequencing identifies signatures of cytomegalovirus exposure history and HLA-mediated effects on the T cell repertoire Analysis of B Cell Repertoire Dynamics Following Hepatitis B Vaccination in Humans, and Enrichment of Vaccine-specific Antibody Sequences The Changing Landscape of Naive T Cell Receptor Repertoire With Human Aging Monoclonal antibodies isolated without screening by analyzing the variable-gene repertoire of plasma cells Genetic measurement of memory B-cell recall using antibody repertoire sequencing Longitudinal Analysis of T and B Cell Receptor Repertoire Transcripts Reveal Dynamic Immune Response in COVID-19 Patients Highly Parallel Genome-wide Expression Profiling of Individual Cells Using Nanoliter Droplets Mapping the Mouse Cell Atlas by Microwell-Seq Visualizing non-metric similarities in multiple maps UMAP: Uniform Manifold Approximation and Projection A Distinct Gene Module for Dysfunction Uncoupled from Activation in Tumor-Infiltrating T Cells High-dimensional single cell analysis identifies stem-like cytotoxic CD8 T cells infiltrating human tumors Clonal CD4 T cells in the HIV-1 latent reservoir display a distinct gene profile upon reactivation Single-cell transcriptome and antigen-immunoglobin analysis reveals the diversity of B cells in non-small cell lung cancer The dynamics and regulators of cell fate decisions are revealed by pseudotemporal ordering of single cells Single-cell landscape of immunological responses in patients with COVID-19 In-depth determination and analysis of the human paired heavy-and light-chain antibody repertoire Simultaneous epitope and transcriptome measurement in single cells High-throughput pairing of T cell receptor α and β sequences High-Throughput Mapping of B Cell Receptor Sequences to Advanced Methodologies in High-Throughput Sequencing of Immune Repertoires Linking T-cell receptor sequence to functional phenotype at the single-cell level T cell fate and clonality inference from single-cell transcriptomes Single-cell RNA-seq and computational analysis using temporal mixture modelling resolves Th1/Tfh fate bifurcation in malaria The differential immune responses to COVID-19 in peripheral and lung revealed by single-cell RNA sequencing Multi-Omics Resolves a Sharp Disease-State Shift between Mild and Moderate COVID-19 Initial characterisation of ELISA assays and the immune response of the clinically correlated SARS-CoV-2 biobank SERO-BL-COVID-19 collected during the pandemic onset in Switzerland Age-dependent effects in the transmission and control of COVID-19 epidemics Dimensionality reduction for visualizing single-cell data using UMAP The effect of ageing on human lymphocyte subsets: comparison of males and females Deep Sequencing of B Cell Receptor Repertoires From COVID-19 Patients Reveals Strong Convergent Immune Signatures High-throughput sequencing of the paired human immunoglobulin heavy and light chain repertoire Affinity maturation and class switching Regulation of HLA-DR gene by IFN-gamma. Transcriptional and post-transcriptional control Lymphotoxin signalling in immune homeostasis and the control of microorganisms Dynamic reorganisation of intermediate filaments coordinates early B-cell activation XBP1, downstream of Blimp-1, expands the secretory apparatus and other organelles, and increases protein synthesis in plasma cell differentiation Stressed-out B cells? Plasma-cell differentiation and the unfolded protein response Role of MHC Class II on Memory B Cells in Post-Germinal Center B Cell Homeostasis and Memory Response Analyzing the Mycobacterium tuberculosis immune response by T-cell receptor clustering with GLIPH2 and genome-wide antigen screening Single-cell landscape of bronchoalveolar immune cells in patients with COVID-19 Broad and strong memory CD4 and CD8 T cells induced by SARS-CoV-2 in UK convalescent individuals following COVID-19 Robust T Cell Immunity in Convalescent Individuals with Asymptomatic or Mild COVID-19 SARS-CoV-2 epitopes are recognized by a public and diverse repertoire of human T cell receptors Use of machine learning to identify a T cell response to SARS-CoV-2 Unbiased Screens Show CD8 T Cells of COVID-19 Patients Recognize Shared Epitopes in SARS-CoV-2 that Largely Reside outside the Spike Protein Comprehensive analysis of T cell immunodominance and immunoprevalence of SARS-CoV-2 epitopes in COVID-19 cases IgG subclasses and allotypes: from structure to effector functions Plasma cell and memory B cell differentiation from the germinal center Transcriptional regulation of memory B cell differentiation Convergent antibody responses to SARS-CoV-2 in convalescent individuals Isolation of potent SARS-CoV-2 neutralizing antibodies and protection from disease in a small animal model Potent neutralizing antibodies from COVID-19 patients define multiple targets of vulnerability Ultra-high resolution HLA genotyping and allele discovery by highly multiplexed cDNA amplicon pyrosequencing Current best practices in single-cell RNA-seq analysis: a tutorial Comprehensive Integration of Single-Cell Data Fast, sensitive and accurate integration of single-cell data with Harmony Slingshot: cell lineage and pseudotime inference for single-cell transcriptomics We thank the Genomics Facility Basel (D-BSSE, ETH Zurich), in particular Ms. Ina Nissen and Dr.Christian Beisel, for their excellent support with the scSeq protocol and for performing deep sequencing runs. This study is supported by funding from the Personalized Health and Related Technologies Postdoctoral The authors declare no competing interests The raw FASTQ files from deep sequencing that support the findings of this study will be deposited under (DOI link from zenodo). Additional data that support the findings of this study are available from the corresponding author upon reasonable request.