key: cord-0882248-qfzb8o7w authors: Mitchel, Jonathan; Gordon, M. Grace; Perez, Richard K.; Biederstedt, Evan; Bueno, Raymund; Ye, Chun Jimmie; Kharchenko, Peter V. title: Tensor decomposition reveals coordinated multicellular patterns of transcriptional variation that distinguish and stratify disease individuals date: 2022-02-18 journal: bioRxiv DOI: 10.1101/2022.02.16.480703 sha: 6379ec959fd8fd46388369dabecb4a41d504cc6f doc_id: 882248 cord_uid: qfzb8o7w Tissue- and organism-level biological processes often involve coordinated action of multiple distinct cell types. Current computational methods for the analysis of single-cell RNA-sequencing (scRNA-seq) data, however, are not designed to capture co-variation of cell states across samples, in part due to the low number of biological samples in most scRNA-seq datasets. Recent advances in sample multiplexing have enabled population-scale scRNA-seq measurements of tens to hundreds of samples. To take advantage of such datasets, here we introduce a computational approach called single-cell Interpretable Tensor Decomposition (scITD). This method extracts “multicellular gene expression patterns” that vary across different biological samples. These patterns capture how changes in one cell type are connected to changes in other cell types. The multicellular patterns can be further associated with known covariates (e.g., disease, treatment, or technical batch effects) and used to stratify heterogeneous samples. We first validated the performance of scITD using in vitro experimental data and simulations. We then applied scITD to scRNA-seq data on peripheral blood mononuclear cells (PBMCs) from 115 patients with systemic lupus erythematosus and 56 healthy controls. We recapitulated a well-established pan-cell-type signature of interferon-signaling that was associated with the presence of anti-dsDNA autoantibodies and a disease activity index. We further identified a novel multicellular pattern that appears to potentiate renal involvement for patients with anti-dsDNA autoantibodies. This pattern was characterized by an expansion of activated memory B cells along with helper T cell activation and is predicted to be mediated by an increase in ICOSLG-ICOS interaction between monocytes and helper T cells. Finally, we applied scITD to two PBMC datasets from patients with COVID-19 and identified reproducible multicellular patterns that stratify patients by disease severity. Overall, scITD is a flexible method for exploring co-variation of cell states in multi-sample single-cell datasets, which can yield new insights into complex non-cell-autonomous dependencies that define and stratify disease. Gene expression is a defining feature that distinguishes different cell types. However, gene expression profiles derived from the same cell type can also vary across individuals, driven by a combination of genetics and environment. Most often, gene expression is compared between individuals in case-control studies or is used to infer sample subgroupings. Alternatively, studies such as the Genotype-Tissue Expression (GTEx) project have revealed the genetic basis of tissue-specific gene expression by mapping natural genetic variation associated with expression differences in human populations (Consortium, 2015; Melé et al., 2015) . Applications of singlecell RNA-sequencing (scRNA-seq) have so far focused on the characterization of transcriptional differences among different cell types and cell states, and analysis of inter-individual variation has been hampered by small sample sizes and the presence of technical batch effects that are often difficult to separate from biological variation. Experimentally, this stimulated the development of multiplexed designs where samples from multiple individuals could be profiled in one run, thereby reducing confounding by technical batches (Kang et al., 2018; Stoeckius et al., 2018) . Analytically, a variety of approaches have been developed to perform dataset alignment (Butler et al., 2018; Haghverdi et al., 2018; Barkas et al., 2019; Stuart et al., 2019) . These tools establish correspondence between samples, effectively treating the difference between individuals as a problem to be overcome. These differences (e.g., between cases and controls or genetically different individuals) are often central for the downstream biological interpretation, but relatively few approaches exist to explore them systematically. The existing methods to compare single-cell transcriptomes between individuals generally require pre-defined groups of samples such as in case/control studies. Comparisons between sample groups can be made using cell-level data or aggregated counts for all cells within a given type or cluster (often referred to as a "pseudobulk" operation) Crowell et al., 2020) . After computing the pseudobulk profiles, differential expression (DE) tools designed for bulk RNAseq analysis can be used to compare sample groups one cell type/cluster at a time. Pseudobulk analysis has been demonstrated to provide a superior balance of robustness, performance, and runtime (Crowell et al., 2020; Squair et al., 2021) and has been used in several single-cell casecontrol studies to date (Kang et al., 2018; Mathys et al., 2019; Corridoni et al., 2020; Liu et al., 2021; Ren et al., 2021; van der Wijst et al., 2021) . However, case-control DE approaches are unable to stratify patients into subgroups and do not account for expression dysregulation that may occur jointly in multiple cell types. Patient subgrouping is of interest when the covariate defining a group has not been well-captured (e.g., disease status has been only partially recorded or recorded with errors) or when additional heterogeneity across samples exists. There is a lack of unsupervised single-cell analysis methods designed for capturing inter-individual variation. Standard matrix decomposition methods such as principal components analysis (PCA) and non-negative matrix factorization can, in principle, be used to describe gene expression variation across pseudobulk samples one cell type/cluster at a time. However, as we will illustrate, it is more informative to consider inter-individual variation within multiple cell populations jointly. Such a joint decomposition would more naturally describe scenarios where different cell types respond specifically to the same external signals. It would also improve our ability to infer dependencies between transcriptional programs across cell types (e.g., due to cell-cell communication or interaction) (Browaeys et al., 2020; Cabello-Aguilar et al., 2020; Efremova et al., 2020; Jin et al., 2021) . For example, a sample undergoing an innate immune response may display increased chemokine expression in myeloid cells and increased expression of chemotaxis genes in neutrophils, as these processes tend to occur together. Overall, by extracting such patterns of gene expression variation, we hypothesize that we can better characterize the molecular bases of complex phenotypes. Here, we developed an unsupervised computational method, called single-cell Interpretable Tensor Decomposition (scITD), that can infer multicellular patterns of gene expression ( Figure 1A ). We define a "multicellular pattern" to be a collection of genes in various cell types that covary together across samples. The multicellular patterns inferred by scITD can be linked with various clinical annotations, genetics, technical batch effects, and other sample metadata, leading to a richer understanding of the system under study. We first assessed the performance of scITD using simulated and real data from an in vitro experiment. Then, we applied scITD to investigate inter-individual heterogeneity in peripheral blood mononuclear cell (PBMC) expression using a dataset with 115 systemic lupus erythematosus (SLE) patients and 56 healthy controls. SLE is a heterogeneous autoimmune disease that can manifest with a wide array of symptoms and has few available targeted therapies (Fava and Petri, 2019; Allen et al., 2021) . We identified six multicellular patterns of gene expression that stratify SLE patients, and we show that these are associated with clinical variables including disease activity and nephritis, one of the most severe complications of SLE. These patterns were examined in depth to identify channels of intercellular communication and changes in cell-type composition. We also applied scITD to a PBMC dataset consisting of 83 patients with COVID-19 and 20 healthy controls, revealing multicellular patterns associated with disease severity. These were validated in an independent study of 49 COVID-19 patients and 11 controls, pointing to conserved mechanisms in IL-16 signaling that could lead to new therapeutic opportunities. Finally, we compared multicellular patterns from the COVID-19 dataset to those from the SLE dataset, revealing similarities and differences in type-1 interferon-stimulated gene response that predispose individuals to autoimmunity but may be protective in acute viral infection. To extract multicellular patterns that vary across individuals, we first generate normalized pseudobulk expression profiles per sample per cell type (Methods). When cell populations from samples are collapsed into pseudobulk profiles, the dataset can be represented as a 3dimensional matrix -a tensor with dimensions × × , where is the number of genes ( Figure 1B left) . Key to scITD, to capture recurrent patterns of transcriptional variation across individuals, we applied Tucker tensor decomposition (Tucker, 1966) to extract the most informative factors. In this context, each factor consists of two elements ( Figure 1A middle). The first element is a gene-by-cell type matrix of loadings values, representing a multicellular pattern (Figure 1B right and 1E right) , and the second element is a vector of sample scores indicating the relative amount of the multicellular pattern present in each sample (Figure 1B middle and 1E left) . Therefore, the output structure for a decomposition to factors consists of a factor-by-gene-bycell type ( × × ) tensor (Figure 1B right) and a sample-by-factor ( × ) matrix (Figure 1B middle). We will refer to the former as the "loadings tensor" and the latter as the "sample scores matrix" or "donor scores matrix" (if there is only one sample per donor). These two data objects can be multiplied together to reconstruct an approximation of the original gene expression tensor, . This representation has a notable advantage, in that each horizontal slice of the loadings tensor represents a multicellular pattern of gene expression that varies across samples according to the corresponding sample scores. We first demonstrate scITD by applying it to a dataset with one primary driving source of variation across samples. This dataset consists of 16 samples of PBMCs from SLE patients, half of which were stimulated in vitro with interferon-beta (IFN-beta) (Kang et al., 2018) (Figure 1C ). For simplicity of the demonstration, we limited the analysis to just classical monocytes (cMonocytes) and CD4+ T cells (Th cells). Differential expression (DE) between stimulated and control samples revealed both shared and cell-type-specific gene expression changes ( Figure 1D ). For example, the gene MX1 becomes upregulated in both cell types after IFN-beta stimulation, whereas ANXA5 becomes upregulated specifically in the monocytes. After applying scITD to this dataset, we examined the sample scores and loadings matrix for the first factor ( Figure 1E ). The scores for this factor perfectly separate the IFN-beta stimulated samples from the control samples ( Figure 1E left). The corresponding factor slice of the loadings tensor reveals a similar pattern with both shared and cell-type-specific perturbed genes ( Figure 1E right). Next, we computed associations between sample scores of factor 1 and the expression of each gene in each cell type to determine the genes with statistically significant contributions in the loadings matrix. The Benjamini-Hochberg (BH) procedure was used for multiple hypothesis test correction here as well as for other p-value adjustments throughout the study (Benjamini and Hochberg, 1995) . The gene expression-factor associations showed a high concordance with the p-values obtained from the regular DE analysis ( Figure 1F ). Similarly, we observed a high correlation between the factor loadings for all genes and the log fold-change values from the DE analysis ( Figure 1G ). This basic example demonstrates that scITD can accurately extract multicellular expression patterns that involve both shared and cell-type-specific genes. (A) The overall goal of scITD. The tool takes clustered and annotated scRNA-seq data from multiple samples/donors as input (left). scITD then identifies multicellular patterns of gene expression that vary across the samples (middle). These patterns can be further analyzed to reveal biological processes that are jointly active in multiple cell types (right). (B) Structure of the output from scITD (middle and right) applied to a single-cell pseudobulk expression tensor (left). An approximation of the expression tensor is reconstructed when the sample scores matrix (middle) and loadings tensor (right) are multiplied together. Sample scores and loadings for one factor are highlighted in green. (C) scRNA-seq data from an IFN-beta stimulation experiment and t-SNE plot with cells colored by their corresponding sample stimulation condition. (D) Sample-level pseudobulk gene expression of DE genes between control and IFN-beta stimulated samples. Rows are genes and columns are pseudobulked samples. Genes that are significant in at least one of the two cell types below an adjusted p-value of 0.01 were included. Genes are grouped (left annotation) by the cell types where they were DE across conditions. A few DE genes are shown labeled on the right. (E) The sample scores (left) and loadings (right) for factor 1 after applying scITD to the IFNbeta stimulation data. Samples in the sample scores vector are labeled by their condition. Only loadings for significant genes in each of the two cell types are shown in the loadings heatmap (Methods). Rows of the loadings heatmap are hierarchically clustered. The same DE gene callouts from (D) are shown as labels on the right. (F) Comparison of DE adjusted p-values (from D) to gene expression-factor 1 sample score association p-values (Methods). Each point is a gene in one of the two cell types. The dashed red lines are located at an adjusted p-value of 0.05. (G) Comparison of DE log2 fold-change values (from D) to loadings values from factor 1. Each point is a gene in one of the two cell types. The Spearman correlation is shown in the upper-right corner. Red dots represent genes that are significantly associated with factor 1 at an adjusted p-value < 0.05. To further evaluate the performance of scITD, we simulated a scRNA-seq dataset with 40 donors (1 sample per donor) and two cell types (Figure S1A left). The dataset was designed to include two multicellular patterns that varied across donors (Figure S1A right), and each pattern involved mostly different genes in each cell type. scITD correctly prioritized the relevant donors and genes involved in each pattern ( Figure S1B , S1C, and S1D). We further downsampled the simulated and IFN-beta datasets to assess the impact of cell number on performance, and we observed good performance above an average of 60 cells per donor per cell type ( Figure S1E and S1G). We also developed an approach to determine the appropriate number of factors into which the initial tensor should be decomposed (Methods). When applying this approach to the simulated dataset, it recommended a dimensionality that yielded accurate recovery (high AUCs) of true DE genes ( Figure S1F and S1D). Figure S1 . Evaluating the performance of scITD with simulated data. (A) UMAP of the simulated scRNA-seq dataset with two cell types (left). Also shown are two multicellular patterns that separate groups of donors (right). Arrows point to the cells from donors with upregulation of a given multicellular pattern. (B) Donor scores matrix after applying scITD to extract two factors. Rows are hierarchically clustered. Labels indicating which donors were assigned to have upregulation of each multicellular pattern are shown on the left annotation. (C) Loadings matrices for the two factors limited to significant genes only. The left matrices show the loading values, the middle matrices show the association significance p-values of each gene in each cell type with the factor, and the right matrices show the true DE genes in each cell type (as set with simulation parameters). Rows are hierarchically clustered. (D) ROC curves and AUC values for predicting ground truth DE genes in each cell type from each gene's expression-factor association p-value. (E) Performance of the method on the simulated dataset downsampled to a varying average number of cells per donor per cell type. AUC is computed the same way as in (D) and is shown for each multicellular pattern as distinct colors. Each point represents a different downsampling iteration (n=5), and the line is the mean AUC at a given dataset size. (F) Rank determination by SVD applied to the full simulated dataset. The left plots show the relative error when performing SVD on the unfolded tensor to varying numbers of factors. Mode 1 error refers to the reconstruction error when SVD is run on the tensor unfolded along the donor dimension. Mode 2 error refers to the reconstruction error when the SVD is run on the tensor unfolded along the gene dimension. The right plots show the change in relative error when incrementing the number of factors. Green bars show the results for the full simulated dataset, whereas purple bars show the results for the dataset after randomly shuffling cell-to-donor assignments (n=50 shuffling iterations). Error bars for the shuffled samples represent standard deviation. (G) Spearman correlations between loadings and log2FC from the IFN-beta experiment data downsampled to a varying average number of cells per donor per cell type. Each point represents a different downsampling iteration (n=5), and the line is the mean Spearman correlation at a given dataset size. Next, we applied scITD to a large scRNA-seq dataset of PBMCs from 115 SLE patients and 56 healthy donors ( Figure 2A ). We focused our analysis on 7 cell types annotated at a coarse-grained level so that we would have a sufficient number of cells per donor per cell type ( Figure 2B left). After transforming expression counts to pseudobulked counts, we further applied batch correction, as groups of donors were pooled and processed together in different 10X Chromium lanes (Methods). We then applied scITD to extract 7 factors, several of which were significantly associated with metadata variables such as SLE status, sex, ethnicity, and age ( Figure 2C ). We first investigated factor 1, which had a strong association with SLE status and explained the most variation in the dataset ( Figure 2C ). The loadings matrix for this factor ( Figure 2D top) revealed a core expression program consisting of interferon-stimulated genes (ISGs) impacting multiple cell types. Since the ISGs had large positive loadings, the interpretation is that they are upregulated in donors with large positive sample scores ( Figure 2D top and 2E). Therefore, this factor distinguishes SLE patients from healthy donors and delineates those patients who have high ISG expression across all cell types. Interferon signaling has been reported by many other groups in the context of SLE, and it is often present in roughly half of all SLE patients (Baechler et al., 2003; Bennett et al., 2003; Crow et al., 2003; Han et al., 2003; Hooks et al., 2010; Nehar-Belaid et al., 2020) . Applying gene set enrichment analysis (GSEA) per cell type for this factor ( Figure 2D bottom) yielded enrichment of the "response to type I interferon" gene set in all cell types as well as other gene sets enriched in specific cell types, especially monocytes. Some of the monocyte-specific gene sets included interleukin (IL)-1 production, IL-6 production, IL-10 production, and TNF production among others ( Figure 2D bottom). These may simply represent the monocyte-specific responses to interferon. Supporting this, roughly 70% of the significant genes in cMonocytes were differentially expressed for this cell type in the IFN-beta stimulation experiment discussed above (adjusted p-values < 0.05). The ISG-high donors also showed higher expression of genes from various other biological processes including cell-cycle in CD8+ T cells (Tc), apoptosis in NK and Tc cells, and proteolysis in multiple cell types ( Figure 2D bottom). Certain proteasome subunits (PSMB8, PSMB9, and PSMB10) are reported to be upregulated by interferon during infections, functioning to enhance antigen presentation (Yang et al., 1992; Shin et al., 2006) . Interestingly, these SLE patients also had upregulation of a pathway for regulatory T cell differentiation ( Figure 2D bottom) marked by the elevated expression of the canonical Treg transcription factor, FOXP3, among helper T cells in these donors ( Figure 2D top). Consistent with this, we observed a significant increase in Treg cell proportions (Tregs are a subcluster within Th cells) for donors with high factor 1 scores (p-value=8.7x10 -19 ). Previous studies have also shown increased numbers of Tregs in SLE patients compared to healthy donors and often accompanying high ISG expression (Suen and Chiang, 2012; Ferreira et al., 2019) . In addition to this finding, we also found a significant reduction in the proportion of naïve Th cells for donors with high ISG expression (p-value = 0.001). Whereas the naïve Th association was also shown in the main analysis of this dataset (Perez et al.) , the Treg association was not previously described. This highlights the ability of scITD to identify coordinated cell-type-specific transcriptional modules within a cohort of patients with autoimmunity. When using scITD on the data without any batch correction, we were able to isolate batch effects into distinct factors ( Figure 2F ). Notably, the number of batch-associated factors remained stable when continually increasing the total number of factors. Therefore, a user can apply scITD to nonbatch-corrected data and then choose to analyze the factors that are not batch-associated. However, this also provides a unique opportunity to explicitly study the multicellular nature of batch effects. We demonstrated this using our SLE dataset and showed that 10X Chromium laneassociated factors often have consistent expression patterns across different cell types ( Figure S2A and S2B). We further analyzed these patterns for associations with gene-level attributes such as GC-content, ambient RNA expression, or gene ontologies. For example, 10X laneassociated genes shared between cell types were more likely to be present at higher fractions in droplets containing ambient RNA compared to the cell-type-specific genes ( Figure S2C ). This analysis provides a unique use case for scITD to examine how batch effects impact multiple cell types and may eventually pave the way for improved batch-correction techniques. One unique attribute of the Tucker tensor decomposition is that the factors can be rotated to improve their interpretability (Unkel et al., 2011; Zhou and Cichocki, 2012) . Therefore, we compared rotations applied to either the loadings or the donor scores of the SLE decomposition ( Figure S3A ) (Methods). We show that by rotating the loadings, the factor patterns become less similar to one another and more modular ( Figure S3B ). This independence enables us to interpret individual factors as functionally interconnected multicellular patterns. In contrast, a rotation of the donor scores can produce factors that more strongly stratify patients into groups, although the multicellular patterns become less modular ( Figure S3C and S3B). From a user perspective, it is important to keep these distinctions in mind and to select an appropriate rotation based on the goals of the analysis. For most analyses in this study, we opted to use a rotation on loadings. In addition to identifying multicellular patterns broadly associated with disease status, scITD can also be used to identify those that stratify samples, which may be particularly useful for studying a heterogeneous disease like SLE. Therefore, we next applied scITD to only the samples from SLE patients to help identify multicellular patterns that may be associated with various clinical annotations. The resulting decomposition yielded factors that were highly similar to the previous ones ( Figure S4C ). Factors 1, 2, 3, and 7 involve multiple cell types, factors 4 and 5 primarily involve cytotoxic T cells (Tc) and Th cells, and factor 6 shows consistent expression variation for all cell types, as this factor is associated with sex ( Figure S4A and S4B). We further demonstrated that the factors have high stability by randomly subsampling to 85% of the donors, recomputing the decomposition, and assessing factor correlations with those from the full SLE-only decomposition ( Figure S4D ). (C) Analysis of factor 1 loadings for association with ambient RNA content of batch dmx_YE_7-19. The left plot shows the association between all genes that are upregulated in this batch and their fractional representation in the ambient RNA "soup" for the batch. The right plot shows ambient RNA fractions for genes that were significantly upregulated across all cell types compared to those upregulated only in some cell types. Associations are calculated by two-sample t-tests for (G-H). "NS genes" for (G-H) includes all genes that were not significantly upregulated. (D) Associations between upregulated genes in each factor and GC content. This is shown separately for the batch-associated factors (top) or non-batch-associated factors (bottom). iteration, the dataset was subsampled to 85% of the donors. Values represent the maximum absolute value factor-factor correlation coefficients for each of the original factors mapped to the factors from each subsampled dataset decomposition. This is shown separately for max donor score correlations (left) and max loadings correlations (right). Next, we associated these seven factors (from the SLE-only decomposition) with 41 clinical features including SLE symptoms and medication use. One of these is the SLE disease activity index (SLEDAI), which is a composite score derived from 24 manifestations and immunologic features spanning 9 organ systems (Bombardier et al., 1992) . Factor 1, which was again described by pan-cellular ISG expression, was significantly associated with the presence of anti-dsDNA and anti-smith autoantibodies ( Figure 3A ) as well as higher SLEDAI scores ( Figure 3B ). The connection between higher ISG expression and these clinical features has been previously shown in several studies (Hooks et al., 1979; Bennett et al., 2003; Kirou et al., 2005; Nikpour et al., 2008; Weckerle et al., 2011) . The Pearson correlation between factor 1 sample scores and SLEDAI was 0.321, which is similar to that which was observed in other studies (Catalina et al., 2019; Enocsson et al., 2021; Juárez-Vicuña et al., 2021) . Lupus nephritis is one of the most severe complications of SLE, and anti-dsDNA autoantibodies are a critical though insufficient component to its development (Yung and Chan, 2015) . Therefore, we sought to identify multicellular patterns that are associated with nephritis when autoantibodies are present. Factor 2 exhibited a significant association with the frequency of lupus nephritis among SLE patients who were positive for anti-dsDNA autoantibodies ( Figure 3C ). The frequency of patients with nephritis was computed using a sliding window along the factor scores to calculate the number of patients positive for lupus nephritis among those positive for anti-dsDNA autoantibodies (Methods). Notably, there is no association in patients negative for anti-dsDNA autoantibodies, and co-occurrence of anti-smith autoantibodies with lupus nephritis for this factor was less significant (p-value ~ 0.05). This finding suggests that the factor 2 multicellular pattern might exacerbate the adverse effects of anti-dsDNA autoantibodies specifically, leading to lupus nephritis. GSEA analysis reveals that factor 2 is enriched for stress response as well as altered cell cycle, apoptosis, cell migration, and cell adhesion pathways in multiple cell types ( Figure 3D) . Directionally, the genes in these pathways were overexpressed in the patients with high factor 2 scores. The observation of p38 MAPK signaling in several cell types was also intriguing because there is evidence that this pathway is an important mediator in the development of lupus nephritis, acting in concert with anti-dsDNA autoantibodies (Iwata et al., 2003; Yung et al., 2010; Liu et al., 2016) . (C) Factor 2 association with frequency of lupus nephritis among patients positive for anti-dsDNA autoantibodies. A sliding window was used to compute the percent of patients within the window that had lupus nephritis (Methods). Each point represents the sliding window center. (D) Factor 2 select enriched gene sets computed for each cell type. Enrichment significance was calculated using the FGSEA R package with GOBP gene sets (also applies to G). Rows are hierarchically clustered. (E) Factor 3 association with prednisone use. The significance of each association was computed using logistic regression with a likelihood-ratio test. (F) Factor 3 association with prednisone dose. The outlier with the highest prednisone dose was not included in the calculation of the linear model p-value but is still shown in the plot. The p-value was calculated using a linear model F-test. The line is a linear model. (G) Factor 3 select enriched gene sets computed for each cell type. Enrichment significance was calculated using the FGSEA R package with GOBP gene sets. Rows are hierarchically clustered. Association analysis of treatment revealed that factor 3 was strongly associated with both the use and dosage of the corticosteroid prednisone ( Figure 3E and 3F). GSEA analysis for this factor confirmed the expected enrichment of corticosteroid and hormone response genes in multiple cell types ( Figure 3G ). There were also several enriched pathways specific to Th cells. We further noted that several of the SLE patients taking prednisone did not have high scores for this factor. In fact, the SLE patient taking the highest prednisone dose had practically none of this multicellular pattern ( Figure 3F ). These few individuals may exhibit resistance to prednisone, as previous reports have shown that up to a third of SLE patients may have some degree of resistance to the medication (Luijten et al., 2013) . Overall, this illustrates one of the main benefits of using scITD over simple DE analyses, as DE would be underpowered to detect the prednisone-associated genes with such outliers. We also sought to identify LR interactions that are candidate mediators of the multicellular patterns identified by scITD. Our approach to inferring LR interactions differs from standard approaches in several ways. Most single-cell LR methods identify interactions based on the upregulation of ligands and their cognate receptors in pairs of cell clusters without regard to the sample of origin. Here, we explicitly test for interactions that are differentially active between samples. This is done by associating ligand expression in a source cell type with the expression of WGCNA co-expression gene modules in a target cell type, which we use as a proxy to represent the downstream effects of an LR signaling event ( Figure 4A ) (Methods). Applying this strategy to the SLE samples, we identified a set of ligands and gene modules significantly associated with each other across samples ( Figure 4B left). These candidate interactions can be further associated with the inferred multicellular patterns ( Figure 4B right). We further demonstrated that our approach enriches for plausible LR interactions and outperforms a standard LR inference approach (Methods) ( Figure S5A and S5B). Next, we more closely examined several of the top candidate LR interactions. One such prediction is the interaction of the ligand TNFSF13B expressed from cMonocytes with its cognate receptor on B cells. Specifically, we identified a positive association between monocytic expression of the ligand and a co-expression gene module in the B cells, B_m1 ( Figure 4B blue arrows and 4C left). The protein that TNFSF13B codes for is more commonly referred to as the B-lymphocyte stimulator (BLyS) or B-cell activating factor (BAFF). Notably, this protein is the target of the recently developed therapeutic, belimumab, which acts to reduce B cell activation and autoantibody production in SLE (Allen et al., 2021) . Here, the genes in the associated B_m1 coexpression module were significantly enriched for genes in B-cell receptor activation gene sets as well as differentiation and cell-cycle gene sets (adjusted p-values < 0.05), as would be expected from BAFF stimulation. We observed that donors with high ISG expression (high factor 1 scoring donors) had significantly higher expression of TNFSF13B ( Figure 4B right and 4C right). This is consistent with previous reports that the TNFSF13B ligand is itself a known target upregulated by interferon (Sjöstrand et al., 2016) . As we have shown, connecting our LR inference results to our scITD multicellular patterns enables contextualization of candidate interactions, improving our confidence in them. Another particularly strong candidate that we identified is the ICOSLG-ICOS interaction from cMonocytes to Th cells ( Figure 4D ). Specifically, we found that donors with higher ICOSLG expression in cMonocytes had significantly higher expression of gene module Th_m5 in Th cells. The Th_m5 gene module was enriched for genes involved in T-cell receptor activation, cell cycle, and p38 MAPK pathways ( Figure 4E ) consistent with the known co-stimulatory role for ICOSLG-ICOS binding in T cell activation (Dodeller and Schulze-Koops, 2006; Wikenheiser and Stumhofer, 2016) . We also observed that genes in the Th_m5 module had significantly higher NicheNet regulatory potential scores compared to genes in other modules ( Figure 4F ). For a given ligand, genes with higher regulatory potential scores are more likely to be downstream targets of that ligand (Browaeys et al., 2020) . Therefore, this result is consistent with the hypothesis that donors who overexpress ICOSLG have increased functional ICOSLG-ICOS signaling between monocytes and Th cells. We also noted that the donors with increased ICOSLG expression had significantly higher scores for factor 2 ( Figure 4B right and 4D right), suggesting the interaction between ICOSLG-ICOS may promoting T cell proliferation and MAPK activation to increase the frequency of lupus nephritis ( Figure 4G ). Corroborating this result, a previous study showed that T cell ICOS stimulation by myeloid cells contributed to the development of lupus nephritis and was mediated by increased T cell survival (Teichmann et al., 2015) . We also identified a significant interaction channel that appeared active for donors with functional prednisone response. Specifically, we found that the ligand THBS1 in cDCs was positively correlated with the Th_m9 co-expression module ( Figure 4H left) . The THBS1 ligand itself was also positively correlated with factor 3 donor scores, indicating that it is overexpressed with functional prednisone response ( Figure 4H right). Similar to factor 3 ( Figure 3G ), module Th_m9 was significantly enriched (adjusted p-values < 0.01) for genes involved in response to hormone (e.g., KLF9, TXNIP) and regulation of T cell activation (e.g., SOCS1, NFKBIZ). Previous studies have also shown that dendritic cell-derived THBS1 can promote Treg development when interacting with integrin-associated protein (CD47) (Grimbert et al., 2006 ). Therefore, we tested for an association between the factor 3 scores and Treg proportions. This association was statistically significant (p-value = 0.002), with a relative expansion of this Th subpopulation in the prednisone patients. Unlike the factor 1-high donors, however, the factor 3-high patients did not have increased ISG expression. We also observed significantly higher THBS1 regulatory potential scores for genes in the Th_m9 gene module compared to genes in all other modules (p-value = 2.8x10 -8 ), supporting the inference of this interaction. These results highlight a high confidence shift in intercellular communication upon prednisone use that may mechanistically contribute to its anti-inflammatory effects. Association between THBS1 expression in cDCs with donor scores for factor 3 (right). Figure S5 . Validation of the scITD LR inference approach using NicheNet regulatory potential scores. (A) LR channels inferred using scITD with the CellChat LR pair database. The height of each bar indicates the adjusted p-value (calculated with the Wilcoxon rank-sum test) comparing NicheNet regulatory potential for the top 200 potential target genes to the rest (Methods). The red bar indicates an adjusted p-value of 0.05. (B) The number of inferred LR channels using scITD compared to a simple LR pair association approach (left). The plot on the right shows the fraction of inferred LR channels with significant NicheNet enrichment (as calculated in G). The "random" method is a random selection of 250 LR channels as a background comparison. Error bars are the standard error of the mean. Different scenarios may underlie the inter-individual transcriptional variation captured by the scITD factors. The variation may represent differences in gene expression within a given cell type (e.g., different activation states of CD4 T cells), or it may reflect altered proportions of cell subtypes (e.g., regulatory versus effector CD4 T cells) in different samples. We implemented a strategy to identify cell subtype "compositional shifts" in an automated fashion ( Figure 5A ). For a given major cell type, we first perform subclustering to various resolutions. As finer resolution clusters may capture disease-specific expression states, we generally check that non-canonical subtypes are present to a minimal extent in the samples from healthy donors. For a given subclustering, we can then test whether the estimated cell subtype proportions are significantly associated with a given factor. This is demonstrated with the major Th cell cluster using the SLEonly portion of the dataset ( Figure 5B ). We found that certain subclustering resolutions yielded significant associations with the factor scores, indicating that compositional shifts likely contribute to the observed inter-individual variation. We then selected a single subclustering resolution for each major cell population to investigate cell subtypes further ( Figure 5C , 5D, S6A, and S6B). Firstly, we observed that donors with high factor 1 scores (those who had high ISG expression) tended to have lower fractions of the Th_1 subtype ( Figure 5E and 5F). This subtype overexpressed genes such as CCR7 and SELL indicating that it likely represents naïve Th cells ( Figure 5D left) . This result is consistent with the aforementioned shift in naïve Th cells when using the previously ascribed subtype annotations. Factor 1 donor scores were also significantly associated with altered proportions of B cell subtypes ( Figure 5G ). Specifically, the ISG expressing patients appeared to have expanded populations of activated memory B cells and transitional B cells. Markers for the activated memory B cells, B_4 ( Figure 5D right), match those from a recent single-cell study of healthy individuals (King et al., 2021) . The transitional B cell subtype (B_3) was annotated by marker genes, CD72 and TCL1A, identified from previous studies (Shen et al., 2020; Stewart et al., 2020) . These findings are also consistent with our observation of TNFSF13B upregulation in these patients (identified in our LR analysis), as BAFF has been shown to enhance survival of these B cell subtypes (Hsu et al., 2002) . Transitional B cells have previously been shown to be at increased proportions in SLE patients and they are known to play a role in regulating Th differentiation and reducing their proliferation (Simon et al., 2016; Dieudonné et al., 2019; Liu et al., 2019; Zhou et al., 2020) . Along these lines, we expected to see reduced Th numbers overall. Therefore, we tested whether factor 1 donor scores were also associated with the composition of the major cell types. We observed a significant overall association, with the Th cells at reduced proportions for the SLE patients with high ISG expression ( Figure 5H ). Analysis of cell subtype compositional shifts with the other factors also yielded several significant associations with relevant connections to their corresponding multicellular patterns ( Figure S6C -G). genes were determined from DE tests between subclusters using Conos. (C) Association significance between B cell subcluster proportions and factor 2. The standard error is used for the error bounds, and the p-value is calculated as in ( Figure 5B ). This also applies to (D-G). (D) Association significance between cMonocyte subcluster proportions and factor 3. (E) Association significance between Th subcluster proportions and factor 3. (F) Association significance between Tc subcluster proportions and factor 4. (G) Association significance between Th subcluster proportions and factor 4. Next, we applied scITD to analyze a large scRNA-seq dataset consisting of 83 COVID-19 patients and 20 healthy controls (Stephenson et al., 2021) . The patients demonstrated varying degrees of disease severity at the time of sample collection, ranging from asymptomatic to critical. We again limited our analysis to the major cell populations ( Figure 6A ). Similar to our SLE analysis, the decomposition of this data yielded one factor characterized by pan-cell type ISG expression (factor 1) ( Figure 6C ). High ISG expression was seen in a subset of the COVID-19 patients but not in the healthy controls ( Figure 6D) . Interestingly, the patients with critical disease also had significantly lower ISG expression compared to other patients ( Figure 6D ). This may be partially due to the presence of anti-interferon autoantibodies as indicated by recent reports (Bastard et al., 2020; Wang et al., 2021; van der Wijst et al., 2021) . As ISG seems to play opposite roles in COVID-19 (protective role) compared to SLE (pathogenic role), we aimed to identify differences in ISG expression between acute viral infection and autoimmunity. In comparing the ISG multicellular patterns extracted from the COVID-19 dataset versus the SLE dataset, we found many overlapping significant genes per cell type ( Figure 6E , S7A, and S7B), and the most strongly associated canonical ISGs (e.g., IFI6, MX1, XAF1, etc.) were all significant in both datasets ( Figure 6E and S7A). However, the ISG multicellular pattern in the SLE dataset contained many genes that were not significant in the COVID dataset ( Figure S7B ). These SLE-specific factor 1 genes were enriched for biological processes such as ATP metabolic process (Tc cells), cell activation (NK cells), and cell-cell adhesion (NK cells) among others ( Figure 6F ). Enrichment of ATP metabolic processes in Tc cells is particularly intriguing as one recent SLE study demonstrated striking metabolic changes in Tc cells that take place only in response to chronic interferon exposure and were absent with short-term exposure (Buang et al., 2021) . As a negative control, we compared the ISG factor of this COVID-19 dataset to that of another COVID-19 scRNA-seq dataset (van der Wijst et al., 2021) and observed no significant gene sets among the dataset-specific genes. We also noticed that many of the SLE-unique factor 1 genes represented components of the proteasome, including the immunoproteasome subunit, PSMB10, upregulated in all cell types analyzed. While proteasome subunits are significantly upregulated with ISGs in both COVID-19 and SLE ( Figure 2D and 6C ), there appears to be a far stronger enrichment of these genes in the SLE ISG factor ( Figure 6G ). This was observed for Tc cells, Th cells, and NK cells ( Figure S7C ). Two genetic studies of ISG expression in SLE found a significant variant in PPM1H, a gene that can remove proteolysis signals (Kariuki et al., 2010; Ghodke-Puranik et al., 2020) . Therefore, genetics or other disease-specific factors may play a role in differentiating the ISG multicellular pattern in SLE from that seen in other illnesses. Unlike the ISG factor, factor 2 of the COVID-19 decomposition significantly stratified patients along a continuous spectrum disease severity ( Figure 7A ). The multicellular pattern for this factor consisted of multiple cell-type-specific biological processes including cell-cycle (NK and T cells) and various signaling cascades (in cMonocytes, NK, and T cells) among others ( Figure 7B ). We further examined this factor for associations with cell proportions. We found that the more severe patients had reduced proportions of activated Th cells (IL22+) as well as increased proportions of terminal effector Tc cells ( Figure S7D ). Similar subcluster proportion associations were also reported in the original study that generated this dataset (Stephenson et al., 2021) . We also found that the factor 2-high donors had significantly increased proportions of proliferating Th cells, Tc cells, and NK cells ( Figure S7D ). This is consistent with the observed enrichment of cell-cycle processes in these cell types for these patients ( Figure 7B ). By further applying our LR inference technique, we identified a strong candidate LR interaction connected to this multicellular pattern. The interaction included the ligand IL16 expressed from Th cells interacting with the CD4 receptor on cMonocytes. Specifically, we observed high correlations between IL16 expression in Th cells and various co-expression gene modules in cMonocytes, including cMono_m14 ( Figure 7D right) . Module cMono_m14 was significantly enriched for MHC Class II genes ( Figure 7E ), indicating a possible role of Th derived IL16 in regulating the expression of these genes in monocytes. Corroborating this, earlier studies have shown that IL16 can upregulate monocytic MHC Class II genes, including HLA-DR (Cruikshank et al., 1987) . Here, we further found Th IL16 to be downregulated in donors with high factor 2 scores ( Figure 7D left), and the factor 2 multicellular pattern shows that these donors also have downregulation of HLA-DR expression in monocytes ( Figure 7B top) . Interestingly, reduced expression of HLA-DR in monocytes has also been found in patients with sepsis, a process that shares many of the same pathophysiological features as severe COVID-19 (Winkler et al., 2017; Olwal et al., 2021) . Two recent studies have also identified reduced levels of HLA-DR protein on monocytes of critically ill COVID-19 patients (Giamarellos-Bourboulis et al., 2020; Spinetti et al., 2020) . Given our result and the prior literature, further studies should be conducted to determine whether reduced IL16 contributes to increased COVID-19 severity via reduced HLA-DR expression. Finally, we aimed to replicate this severity-associated multicellular pattern by analyzing another COVID-19 scRNA-seq dataset. The test dataset (van der Wijst et al., 2021) consisted of a smaller number of donors but included the same cell types and a similar range of disease severity (not including asymptomatic or mild cases) ( Figure S7E ). We projected the factor 2 pattern extracted from the Stephenson et al. dataset onto the van der Wijst et al. dataset and similarly found that this pattern significantly stratified patients by disease severity ( Figure 7C) . A meta-analysis combining the result from these two datasets yielded a Fisher p-value of 4.3x10 -17 . The severe and critical patients in the test dataset also had significantly reduced IL16 expression in their Th cells ( Figure 7F left) matching our observation from the Stephenson et al. data. Similarly, IL16 was again positively associated with a co-expression module in cMonocytes ( Figure 7F right) that was enriched for MHC Class II genes ( Figure 7G ). The consistency of these results across independently generated datasets bolsters our confidence in the connection between this multicellular pattern and COVID-19 disease severity and highlights the conserved nature of a potentially crucial cell-cell interaction. Transcriptional response of complex biological tissues to different conditions or diseases can involve coordinated changes across multiple cell types. These multicellular events may represent the joint reactions of multiple cell types to a common set of stimuli as well as the cell-cell interactions that contribute to the associated phenotypes. Here, we developed a single-cell computational tool, scITD, to identify coordinated multicellular patterns of expression variation across individuals. Our approach can be applied to any scRNA-seq dataset with multiple samples, though it is best geared for use with datasets consisting of many source donors. We validated that the tool can extract meaningful and accurate multicellular patterns using simulated data as well as data from an in vitro IFN-beta stimulation experiment. We also developed a component of our method to infer cell-cell interactions and showed that it outperformed a widely used LR inference strategy. The primary use cases for scITD involve the study of inter-individual variation within different types of sample collections, including those sampling normal individuals, case-control studies, or disease subtyping studies. In this study, we principally applied the tool to discover multicellular patterns that stratify SLE patients. From this dataset, we extracted a multicellular pattern involving ISG expression that separated SLE patients from the healthy controls. This pattern was significantly associated with the presence of autoantibodies and SLE disease activity, recapitulating similar associations from previous studies. Our method uniquely allowed us to identify additional cell-type-specific biological processes co-occurring with ISG expression (including B cell activation, Treg expansion, etc.) suggestive of interactions between circulating cell types. By further connecting the other factors to clinical metadata, we were able to identify a multicellular pattern that was linked to increased frequency of lupus nephritis among the patients positive for anti-dsDNA autoantibodies. While several of the cell-type-specific components of this multicellular pattern have been previously validated and linked to lupus nephritis (e.g., P38 MAPK signaling and ICOSLG signaling from myeloid cells to T cells), our analysis suggests that these pathways may be important in explaining why anti-dsDNA autoantibodies are necessary but insufficient to cause these severe symptoms in SLE. We further showed that our method can yield insightful patterns in other large patient cohorts by applying it to a COVID-19 scRNA-seq dataset. We extracted a factor consisting of pan-cell-type ISG expression that separated healthy donors from a subset of COVID-19 patients. This ISG multicellular pattern appeared to play a protective role in the disease as the critically ill patients had reduced ISG expression. We compared this ISG pattern to that from the SLE dataset and identified cell-type-specific differences that may be connected to the chronic nature of SLE. Another factor from the COVID-19 dataset was strongly associated with the continuous spectrum of disease severity and displayed a more complex multicellular pattern of expression. We demonstrated that this pattern was also predictive of COVID-19 severity in an independently generated scRNA-seq dataset, highlighting the robustness and generalizability of the extracted multicellular patterns. Lastly, we highlighted a high-confidence LR interaction involving the ligand IL16 expressed in T cells and interacting with the CD4 receptor on cMonocytes. This interaction appeared to be downregulated for the most severe COVID-19 patients, highlighting a need for follow-up studies elucidating its role in this disease and evaluating its therapeutic potential. Finally, this work can be extended in several directions. For one, scITD can be applied to more thoroughly investigate how different technologies, processing techniques, and disease models impact expression jointly within each cell type. We briefly demonstrated how scITD can be used in this way, showing that ambient RNA within 10X Chromium lanes tends to alter expression across all cell types. More work in this area could lead to better-informed designs of batchcorrection methods tailored for various scenarios. In another application area, it may also be valuable to connect the scITD multicellular patterns with genetic variants. By further applying techniques such as Mendelian randomization, it will be possible to test causal hypotheses for genes that may be upstream drivers of the observed patterns. Another use case for the scITD is in the study of multi-tissue patterns of gene expression (as opposed to multi-cell type patterns). Since scITD uses scRNA-seq data at the pseudobulk-level, the tool can be directly applied to bulk RNA-seq datasets generated from multiple donors with multiple tissue types (e.g., the GTEx studies). This could allow one to connect gene expression changes in the blood with the expression states of less accessible tissues. Overall, scITD offers a novel approach to single-cell data analysis, extending our ability to study the complex biological processes that stratify individuals in health and disease. The first step is to transform a gene-by-cells UMI counts matrix into a pseudobulked tensor for decomposition. For cells of a given sample and cell type, all counts for each gene are summed and divided by the total counts from all genes. Then, the trimmed-mean of M values (TMM) method in edgeR (Robinson et al., 2010 ) is used to adjust library sizes of the pseudobulked counts and the data are normalized and log-transformed. Before this previous step, we only retain donors with at least a minimum number of cells in all cell types, as the Tucker decomposition does not allow for NA values in the tensor. Next, we then compute a measure of normalized variance for each gene, as is calculated in the R package, pagoda2 (Barkas et al., 2021) . Overdispersed genes are selected from each cell type and each gene-by-sample matrix is reduced to the union of overdispersed genes from all cell types. After normalization, the data are centered and unit scaled across samples. We then rescale genes by their normalized variance calculated previously. This is done by multiplying the expression by the normalized variance value to some power. The power should be set to 0.5 for the resulting variance to equal the normalized variance. We note that increasing the value of the power slightly (often between 1-2) can sometimes improve the quality of the decomposition. Finally, the pseudobulked cell-type matrices are stacked together to form the tensor. Next, we apply the Tucker tensor decomposition to the resulting tensor. For this, we use the R package, rTensor (Li et al., 2018) , which implements Higher-Order Orthogonal Iteration (HOOI) to compute the Tucker decomposition. The formal algorithmic procedure for HOOI can be viewed in the following publication (Sheehan and Saad, 2007) . For our case with three dimensions, HOOI outputs three separate factor matrices and a core tensor that can be multiplied together to reconstruct the approximation of the starting tensor. The three separate factor matrices are of dimensions donors-by-donor factors, genes-by-gene factors, and cell-types-by-cell-type factors. The core tensor is of dimensions donor factors-by-gene factors-by-cell-type factors. The standard data reconstruction using these objects is as follows: Here, is the reconstructed tensor, is the donor factor matrix, is the gene factor matrix, is the cell type factor matrix, and is the core tensor. The operator × $ indicates multiplication of the matrix on the right side of the operator by the tensor on the left side of the operator along the n th mode of the tensor. We then rearrange the terms in the above equation to yield a "donor-centric" view of the decomposition. To do this, we compute a loadings tensor by multiplying the core tensor only by the gene factor-and cell type factor matrices. This is valid to do because the order of multiplication does not matter when reconstructing the data, as long as the multiplication mode also changes accordingly (Kolda and Bader, 2009 ). This reordering and simplification appear as follows: Here, is the loadings tensor of dimensions donor factors-by-genes-by-cell types, the rest of the terms are the same as in the previous equation. In Figure 1B , this reconstruction is shown backward as × ! simply to demonstrate how the tensor times matrix multiplication yields the reconstructed tensor of correct dimensions. As noted previously, the Tucker decomposition to a given number of factors does not have one unique solution. The factor matrices can be rotated by any non-singular square matrix, and as long as the core tensor is counter-rotated by the inverse of the rotation matrix, the reconstruction error will remain unchanged. The core tensor can also be rotated similarly as long as the factor matrices are counter-rotated accordingly. Taking advantage of this property, several groups have found that rotating the factor matrices with independent component analysis (ICA) can improve the interpretability of the factors (Bro; Unkel et al., 2011; Zhou and Cichocki, 2012) . Here, we explored the use of various rotations. One such approach we tested was applying ICA rotation to the donor scores matrix, and counter-rotating the core tensor before generating the final donor-centric view of the decomposition. As a note, we needed to normalize the rotated donor scores matrix because ICA does not preserve lengths. After rotating the donor scores matrix, the core tensor is counter-rotated as follows: Here, 0 is the new core tensor, 1 % is the transpose of the ICA rotated (normalized) donor factor matrix, % is the transpose of the cell type factor matrix, % is the transpose of the gene factor matrix. Then, we substitute 0 for when calculating the loadings tensor in equation 2. The subscript refers to the cell type being used for calculating explained variance. The subscripts , , refer to the donor, gene, and cell type index of the tensor, respectively. To generate the simulations we used the R package, Splatter (Zappia et al., 2017) . Specifically, we applied the Splatter to generate four subpopulations for each of the two cell types (Figure 2A ). To generate a set of four subpopulations, two separate group simulations were run and concatenated together. Each simulation generated two groups of cells separated by some DE genes. Half of the cells from the first population of the first simulation were matched to those of the first population in the second simulation. Likewise, half of the cells from the second population of the first simulation were matched to the other half of the cells from the first population of the second simulation and so on. We then assigned groups of donors (by those with upregulation of each multicellular pattern) randomly to cells from the specified cell subpopulations. We ran the data through the standard scITD pipeline. However, in this analysis, we did not reduce the tensor to only the most variable genes, so that it would be possible to use all genes in the AUC calculation. We computed the Tucker decomposition to two donor factors and four gene factors, as suggested by our rank determination method. We then calculated gene significance p-values for each cell type in each factor. This was done using linear model F-tests with expression as the explanatory variable and donor scores as the response variable. This technique is also used in the main dataset analyses below to identify significant genes per factor. Then, we used these p-values to calculate the ROC AUC for predicting ground truth DE genes that distinguish each of the two multicellular patterns. We further subsampled the simulated dataset to varying sizes to determine the sensitivity of our method to a reduced signal-to-noise ratio. The SLE scRNA-seq dataset was originally demultiplexed using an updated version of demuxlet (Kang et al., 2018) , and quality control measures were applied using Scanpy (Wolf et al., 2018) with the default parameters. We further filtered out cells with over 10% of their UMIs attributed to mitochondrial genes. This dataset originally contained over 200 donors with transcriptomes from over 1.2 million cells. To make it possible to use this dataset with scITD in its current framework, we reduced the dataset down to one sample per donor, used only the largest cell clusters, and restricted it to only those donors with at least 20 cells in each major cell cluster. Cell clusters and annotations from the original study were used. This left us with 171 donors and 632,733 cells. The median number of cells per donor for B, NK, Th, Tc, cDC, cMonocytes, and ncMonocytes were 421, 264, 1145, 688, 50, 939 , and 144 cells respectively. According to our simulation study discussed previously, these quantities were more than sufficient to extract multicellular patterns with high accuracy. We formed the expression tensor using the standard scITD pipeline, and we also applied ComBat batch correction (Johnson et al., 2007) at the level of 10X lanes to each celltype slice of the tensor. For our primary SLE analysis, we used the hybrid rotation method and decomposed the data into 7 donor factors and 20 gene factors. We used the same ranks parameters for the decomposition run on only the SLE donors. We used the SLE-only decomposition to compare factor 1 cMonocyte dysregulated genes with DE genes from the IFNbeta experiment. We then computed associations between the donor scores for each factor and the metadata variables displayed in Figure 2C using linear model F-tests with donor scores as the response variable. The statistical tests used for computing associations between factor donor scores and the other clinical variables depended on the type of the variable being tested. For the ordinal variables such as SLEDAI score and SLICC score, we employed the ordinal logistic regression with the "probit" method, and p-values were calculated using the resulting t-statistics. For binary variables such as the presence of symptoms or prednisone use, we employed a logistic regression with a chi-square test for significance. We only tested the binary variables that were present in at least 20 donors. Multiple hypothesis test correction was applied with the BH procedure. To test for the association of factor 2 with the co-occurrence of lupus nephritis and anti-dsDNA autoantibodies, we first removed any donor scores for donors that did not have anti-dsDNA autoantibodies present. Then we used a sliding window of size 19 to calculate the number of donors within the window that also had lupus nephritis. To calculate a p-value we randomly shuffled donor scores and computed the Spearman correlation between the donor scores and the sliding window count. We repeated this procedure 10000 times to generate a null distribution of correlation values. The null distribution was then used to calculate a p-value, by counting the number of null instances with a larger correlation than the one we observed with the unshuffled data. We only kept the cell barcodes labeled as singlets and used the cell-type annotations ascribed by the authors who generated the data (Kang et al., 2018) . DE analysis results were also used directly from the original paper where the data were generated. We ran the Tucker decomposition to extract two donor factors, four gene factors. We used the ICA rotation method on donor scores for this dataset, although the results were practically identical when obtained using the other rotations as well. The downsampling procedure was performed as described above with the simulated data, except that instead of AUC, we report the Spearman correlation between the loadings and log2FC values from the DE analysis. To help determine the appropriate ranks to decompose the tensor to, we developed a method similar to those commonly used with matrix decompositions. The method works by unfolding the starting tensor along a given mode and computing the SVD to an increasing number of factors. The reconstruction error is calculated for each decomposition. Then, we repeat this procedure multiple times but with a shuffled version of the tensor. The shuffling is done by randomly reassigning cells to donors before tensor formation. This procedure allows us to evaluate when the reduction in real explained variance no longer exceeds that of the randomized scenario. To test the stability of our decomposition, we designed a procedure whereby the data tensor is subsampled to some fraction of donors. For our analysis, we subsampled to 85% of the donors. We then recomputed the decomposition to the same number of factors. The new factors found on the subsampled data were then linked back to the original factors by identifying, for each original factor, which new factor had the highest absolute value correlation with it. We repeated this procedure 500 times and report the max correlations for each original factor with the new factors from the subsampled data. To compute enriched gene sets among genes prioritized within each cell type for a given factor, we use the R package FGSEA (Korotkevich et al., 2021) . In tests of applying GSEA directly to an individual column of a factor loadings matrix, we noticed some spurious results appear as a result of the low-magnitude non-significant loadings being biased toward either positive or negative loadings. To avoid getting these false-positive hits, we compute a new value for each gene. This is calculated as the sum of unit scaled expression values multiplied by donor scores as follows: 6,2,( = ∑ 6,2,7 * (,7 7 Here, 6,2,8 is the new score for gene in cell type for factor . 6,2,7 is the scaled expression of gene in cell type for donor . And (,7 is the donor score for factor and donor . We then apply GSEA to the new gene scores for each cell-type factor combination and apply BH multiple hypothesis test corrections. This has shown to be much more robust in preventing false-positive enriched gene sets, while still yielding gene sets that are expected based on the significant genes. To systematically identify shifts in subtype composition, we first aligned cells across the 10X lane batch variable using Conos (Barkas et al., 2019) . Then we subclustered each major cell population to varying resolutions using the findSubcommunities function from Conos. To get marker genes that distinguish the cell subclusters, we used the getDifferentialGenes function. Marker genes were plotted using the DotPlot function in Seurat (Satija et al., 2015) . To test for associations between a factor and cell subtype composition, we calculated the cell proportions for each donor as the number of their cells from a given subcluster divided by the total number of cells from the corresponding major cell cluster. Next, we convert these proportions to balances using the isometric log-ratio transformation (Egozcue et al., 2003) . This converts the dependent set of proportions to an independent set of p-1 variables, where p is the number of cell subtypes for a given major cell cluster. By making this conversion, it allows one to use these variables with standard statistical tests that require covariates to be independent. As a note, we also add a pseudocount of 1 to each cell proportion numerator to avoid infinities when calculating balances. Then, we use the balances as explanatory variables in a multiple linear regression against the donor scores of each factor. An F-test is used to determine whether a given cell type composition is significantly associated with a factor. The procedure is the same for computing the significance of factor associations with the overall major cell type composition. For this analysis, we used a database of protein ligands and receptors from CellChat (Jin et al., 2021) . We first identified clusters of co-expressed genes in the pseudobulk data for each cell type using the R package WGCNA (Langfelder and Horvath, 2008) . Specifically, we used the signed network and TOM similarity matrix with a module tree cut height of 0.25. Then, we calculated the association between each module eigengene and each ligand from the list of cognate LR pairs. This is only done if the receptor was present in the module cell type to some minimal level (expressed in at least 5 cells for each of the top 15% of donors by ligand expression). Filtering was applied such that we only tested ligands where at least 1% of donors have scaled-adjusted expression above 0.2 (the same is applied to both ligands and receptors in the LR association test below). This removed ligands where high expression is observed in only a few donors or if the ligand has low normalized variance across donors. We also only tested for interactions between different cell types, as we would likely find many false-positive associations between ligands and modules found in the same cell type (due to regulation of the ligand by the same upstream transcription factor regulating the module). The association p-values were calculated by a linear model F-tests with the module eigengene as the response variable and the scaled expression of a ligand as the explanatory variable. P-values were adjusted using the BH procedure. When the CellChat database listed multiple receptor components as required for a specific LR interaction, we required all components of the receptor complex to be expressed. The heatmap in Figure 4B only includes rows with at least one adjusted p-value below 5.0x10-11 and columns with at least one adjusted p-value below 0.001 to reduce the number of results for visual purposes. Finally, we computed the overrepresentation of gene sets within gene co-expression modules of interest. We employed the hypergeometric test to determine whether a module contained a significantly higher proportion of genes from specific gene sets compared to the rest of the genes not found in the module but still included in the analysis. For the B_m1 enrichment tests, we included gene sets from GOBP, KEGG, and Reactome databases. For benchmarking our approach, we compared it to a standard LR association method and a random selection of LR channels. The LR association test inferred an LR interaction if the expression of the ligand from one cell type was significantly associated (adjusted p-value < 0.05) with the expression of its cognate receptor in another cell type. For the scITD LR inference method, we used a more stringent adjusted p-value of 0.0001 to make the number of inferred interactions roughly the same for at least one of the LR pair databases (CellChat database in this case). To demonstrate that our method could enrich for a more confident set of LR interactions, we used NicheNet regulatory potential scores for each ligand. For a given LR channel consisting of a ligand, a source cell type, and a target cell type, we first identified the top 200 genes in the target cell type by the highest absolute value Pearson correlation with the ligand's expression in the source cell type. Then, we compared the regulatory potential scores for these top 200 genes with those from all other genes. We used a Wilcoxon rank-sum test to test for differences in regulatory potential scores between these two groups. As a note, we also applied the normalize_correlation function from the spqn package to correct for gene-gene correlation biases with mean expression (Wang et al., 2020) . To calculate the soup profile for batch dmx_YE_7-19, we calculated the fraction of each gene's UMI counts over the total using all empty droplets with 10 or fewer UMIs. GC content from each gene was retrieved using the EDASeq package in R (Risso et al., 2011) . To test whether the upregulated genes of a given batch factor had a significant association with GC content, we used a two-sample t-test comparing GC contents for upregulated genes versus all other genes included in the analysis. For the non-batch factors, we arbitrarily selected the positive loading direction to call upregulated genes. After removing batch effects and retesting for GC associations, we tested both directions for GC content associations. To evaluate the impact of different factor rotations on the output we compared our hybrid rotation loadings to ICA on donor scores. We used the full SLE dataset for this analysis. For the loadings rotations, we ran the Tucker decomposition to extract 7 donor factors and 20 gene factors as in our main SLE analysis. For the donor scores rotation, we ran the Tucker decomposition to extract 9 donor factors and 20 gene factors because the explained variance was distributed more evenly among the factors for this rotation compared to the loadings rotation. To calculate ISG associations with each factor, we calculated the linear model r-squared value for regressing each ISG against the factor donor scores. The core set of ISGs used included ISG15, IFI27, IRF7, HERC5, LY6E, MX1, OAS2, OAS3, RSAD2, USP18, and GBP5, which were also used to represent ISG expression in a previous study (Davenport et al., 2018) . For the ethnicity association strength comparison, we used the factor from each rotation that was most strongly associated with ethnicity. We evaluated the statistical significance of the associations using linear model F-tests. We used the quality-controlled data that was made publicly available (Stephenson et al., 2021) . A few donors had multiple samples taken at various time points. We kept only the samples labeled with collection day "D0", so that the dataset contained 1 sample per donor. We also excluded patients with other diseases besides COVID-19, and we also excluded the healthy donors given LPS. We used the previous annotations labeled as "full_clustering" and grouped cell subtypes to form the major cell type clusters. Specifically, for B cells we included "B_exhausted", "B_immature", "B_naive", "B_non-switched_memory", and "B_switched_memory". For cMonocytes, we included "CD14_mono" and "CD83_CD14_mono". For Th cells we included "CD4.CM", "CD4.EM", "CD4.IL22", "CD4.Naive", "CD4.prolif", "CD4.Tfh", and "CD4.Th1". For Tc cells, we included "CD8.EM", "CD8.Naive", "CD8.Prolif", and "CD8.TE". For NK cells we included "NK_16hi", "NK_56hi", and "NK_prolif". We also noticed that some of the cells previously labeled as "B cells" clustered with the plasmablasts and expressed the same gene and protein markers as the plasmablasts. Therefore, these were excluded from the B-cell pseudobulks. After removing donors with less than 2 cells in any of the included cell types, we were left with 103 donors and 452,740 cells from the major cell populations used in the analysis. The median number of cells per donor for B, Tc, Th, NK, and cMonocytes were 386, 558, 573, 593, and 717 cells, respectively. The samples were processed at one of three different sites including Cambridge, NCL, and Sanger. Therefore, we applied ComBat batch correction to account for these technical differences as was done with the SLE dataset. We ran the Tucker decomposition to 9 donor factors and 26 gene factors using our hybrid rotation method. To compare the interferon factor from this dataset to that of the SLE dataset, we computed the association T-statistics for all genes that were present in both datasets against the first factors For the genes which were significant only for SLE factor 1 but not COVID factor 1, we computed enriched GO gene sets using the union of significant factor 1 genes from both datasets as the background with a hypergeometric test. We further used the FGSEA package for computing enrichment of donors by status healthy, in either high or low factor one scores. For enrichment of critical patients in factor 1, we removed the healthy donors. We used the quality-controlled data that was made publicly available (van der Wijst et al., 2021). We removed cases that were negative for SARS-CoV-2. We only used samples measured at day 0, such that we retained 1 sample per donor. Broad cell clusters and annotations were used from the original study. After removing donors with less than 2 cells in any of the included cell types, we were left with 60 donors and 166,970 cells from the major cell populations used in the analysis. The median number of cells per donor for B, Tc, Th, NK, and cMonocytes were 208, 459.5, 637, 278.5, and 747 cells, respectively. We also applied ComBat batch correction at the level of 10X lanes. Finally, we ran the Tucker decomposition to 10 donor factors and 30 gene factors using our hybrid rotation method. To project the factor 2 pattern from the Stephenson et al. dataset, we used the intersection of genes from the tensors of both datasets and carried out the projection as described above. We used Fisher's method to compute the meta-analysis p-value combining the severity-association test with that of the larger dataset. To compare factor 2 loadings to the factor loadings from the van der Wijst et al. decomposition, we computed Spearman correlations for each cell type separately. The IFN-beta stimulation dataset can be found in the Gene Expression Omnibus (GEO) at accession number GSE96583. The count-matrices for the SLE scRNA-seq dataset are available in GEO at accession GSE137029. The Stephenson et al. COVID-19 dataset is publicly available at https://www.covid19cellatlas.org/index.patient.html, titled "COVID-19 PBMC Ncl-Cambridge-UCL". The van der Wijst et al. COVID-19 dataset can be found at https://cellxgene.cziscience.com/collections/7d7cabfd-1d1f-40af-96b7-26a0825a306d . Our computational method, scITD, and its associated tutorials can be found at https://github.com/kharchenkolab/scITD. scITD is also available on The Comprehensive R Archive Network (CRAN) at https://cloud.r-project.org/web/packages/scITD/index.html. The code used to produce all figures in this paper can be found at https://github.com/j-mitchel/scITD-Analysis/tree/main/figure_generation. Leveraging Heterogeneity in Systemic Lupus Erythematosus for New Therapies Interferon-inducible gene expression signature in peripheral blood cells of patients with severe lupus Joint analysis of heterogeneous single-cell RNA-seq dataset collections pagoda2: Single Cell Analysis and Differential Expression Autoantibodies against type I IFNs in patients with life-threatening COVID-19 Controlling the False Discovery Rate: A Practical and Powerful Approach to Multiple Testing Interferon and Granulopoiesis Signatures in Systemic Lupus Erythematosus Blood Derivation of the sledai. A disease activity index for lupus patients The N-way on-line course on PARAFAC and PLS NicheNet: modeling intercellular communication by linking ligands to target genes Type I interferons affect the metabolic fitness of CD8+ T cells from patients with systemic lupus erythematosus Integrating single-cell transcriptomic data across different conditions, technologies, and species SingleCellSignalR: inference of intercellular networks from single-cell transcriptomics Gene expression analysis delineates the potential roles of multiple interferons in systemic lupus erythematosus Dissecting heterogeneous cell populations across drug and disease conditions with PopAlign The Genotype-Tissue Expression (GTEx) pilot analysis: Multitissue gene regulation in humans Single-cell atlas of colonic CD8 + T cells in ulcerative colitis Microarray Analysis of Interferon-regulated Genes in SLE muscat detects subpopulation-specific state transitions from multisample multi-condition single-cell transcriptomics data Lymphokine activation of T4+ T lymphocytes and monocytes Discovering in vivo cytokine-eQTL interactions from a lupus clinical trial Transitional B cells in quiescent SLE: an early checkpoint imprinted by IFN The p38 mitogen-activated protein kinase signaling cascade in CD4 T cells CellPhoneDB: inferring cell-cell communication from combined expression of multi-subunit ligand-receptor complexes Isometric Logratio Transformations for Compositional Data Analysis Comparison of Surrogate Markers of the Type I Interferon Response and Their Ability to Mirror Disease Activity in Systemic Lupus Erythematosus Systemic Lupus Erythematosus: Diagnosis and Clinical Management Chronic Immune Activation in Systemic Lupus Erythematosus and the Autoimmune PTPN22 Trp620 Risk Allele Drive the Expansion of FOXP3+ Regulatory T Cells and PD-1 Expression Novel genetic associations with interferon in systemic lupus erythematosus identified by replication and fine-mapping of traitstratified genome-wide screen Complex Immune Dysregulation in COVID-19 Patients with Severe Respiratory Failure Thrombospondin/CD47 Interaction: A Pathway to Generate Regulatory T Cells from Human CD4+CD25− T Cells in Response to Inflammation Batch effects in single-cell RNA-sequencing data are corrected by matching mutual nearest neighbors Analysis of gene expression profiles in human systemic lupus erythematosus using oligonucleotide microarray Immune Interferon in the Circulation of Patients with Autoimmune Disease Immune Interferon in the Circulation of Patients with Autoimmune Disease Cutting edge: BLyS enables survival of transitional and mature B cells through distinct mediators p38 Mitogen-Activated Protein Kinase Contributes to Autoimmune Renal Injury in MRL-Faslpr Mice Inference and analysis of cell-cell communication using CellChat Adjusting batch effects in microarray expression data using empirical Bayes methods Polymorphisms Is Not Associated With Susceptibility to Systemic Lupus Erythematosus, Although It Regulates OASL Expression in Patients With SLE Multiplexed droplet single-cell RNAsequencing using natural genetic variation Trait-stratified genome-wide association study identifies novel and diverse genetic associations with serologic and cytokine phenotypes in systemic lupus erythematosus Single-cell analysis of human B cell maturation predicts how antibody class switching shapes selection dynamics Activation of the interferon-alpha pathway identifies a subgroup of systemic lupus erythematosus patients with distinct serologic features and active disease Tensor Decompositions and Applications Fast gene set enrichment analysis WGCNA: an R package for weighted correlation network analysis rTensor: An R Package for Multidimensional Array (Tensor) Unfolding, Multiplication, and Decomposition Time-resolved systems immunology reveals a late juncture linked to fatal COVID-19 Type I interferons promote the survival and proinflammatory properties of transitional B cells in systemic lupus erythematosus patients Tumor Necrosis Factor-Like Weak Inducer of Apoptosis Accelerates the Progression of Renal Fibrosis in Lupus Nephritis by Activating SMAD and p38 MAPK in TGF-β1 Signaling Pathway The use of glucocorticoids in Systemic Lupus Erythematosus. After 60years still more an art than science Single-cell transcriptomic analysis of Alzheimer's disease The human transcriptome across tissues and individuals Mapping systemic lupus erythematosus heterogeneity at the single-cell level Association of a gene expression profile from whole blood with disease activity in systemic lupus erythaematosus Parallels in Sepsis and COVID-19 Conditions: Implications for Managing Severe COVID-19 Single-cell RNA-seq reveals the cell-type-specific molecular and genetic associations to lupus COVID-19 immune features revealed by a large-scale single-cell transcriptome atlas GC-Content Normalization for RNA-Seq Data edgeR: a Bioconductor package for differential expression analysis of digital gene expression data Spatial reconstruction of single-cell gene expression data Higher Order Orthogonal Iteration of Tensors (HOOI) and its Relation to PCA and GLRAM A regulatory role for CD72 expression on B cells and increased soluble CD72 in primary Sjogren's syndrome Virus-induced type I IFN stimulates generation of immunoproteasomes at the site of infection In-depth characterization of CD24(high)CD38(high) transitional human B cells reveals different regulatory profiles The Expression of BAFF Is Controlled by IRF Transcription Factors Reduced Monocytic Human Leukocyte Antigen-DR Expression Indicates Immunosuppression in Critically Ill COVID-19 Patients Confronting false discoveries in single-cell differential expression Single-cell multi-omics analysis of the immune response in COVID-19 Singlecell transcriptomic analyses define distinct peripheral B cell subsets and discrete development pathways Cell Hashing with barcoded antibodies enables multiplexing and doublet detection for single cell genomics Comprehensive Integration of Single-Cell Data CD4+FoxP3+ regulatory T-cells in human systemic lupus erythematosus Local Triggering of the ICOS Coreceptor by CD11c+ Myeloid Cells Drives Organ Inflammation in Lupus Some mathematical notes on three-mode factor analysis Independent Component Analysis for Three-Way Data With an Application From Atmospheric Science Diverse functional autoantibodies in patients with COVID-19 Co-expression analysis is biased by a meancorrelation relationship Network Analysis of Associations between Serum Interferon Alpha Activity, Autoantibodies, and Clinical Features in Systemic Lupus Erythematosus Longitudinal single-cell epitope and RNA-sequencing reveals the immunological impact of type 1 interferon autoantibodies in critical COVID-19 ICOS Co-Stimulation: Friend or Foe? Front Human leucocyte antigen (HLA-DR) gene expression is reduced in sepsis and correlates with impaired TNFα response: A diagnostic tool for immunosuppression SCANPY: large-scale single-cell gene expression data analysis Proteasomes are regulated by interferon gamma: implications for antigen processing Mechanisms of Kidney Injury in Lupus Nephritis -the Role of Anti-dsDNA Antibodies Anti-dsDNA Antibodies Bind to Mesangial Annexin II in Lupus Nephritis Splatter: simulation of single-cell RNA sequencing data Fast and Unique Tucker Decompositions via Multiway Blind Source Separation Transitional B cells involved in autoimmunity and their impact on neuroimmunological diseases We thank Anna Igolkina (St. Petersburg Technical University, Russia) for advice on the cell proportion analysis. The primary approach used in most analyses of this study involves a two-step rotation procedure which is a hybrid of ICA and varimax applied to the distinct components that make up the loadings tensor. The intuition behind this approach is to create a core tensor, where each donor factor represents some combination of biologically distinct gene sets (gene factors) in the different cell types. Furthermore, donor factors of the core tensor should be rotated to a simple structure to make the multicellular patterns more modular. This helps to ensure that each gene factor only partakes primarily in one donor factor. This can be achieved by applying varimax to the core tensor. However, it is also necessary to ensure that all gene factors are independent of one another. Otherwise, the optimized core tensor may still yield donor factor loadings with similar sets of relevant genes. To accomplish this, we apply rotations in two separate steps. In the first step, we apply ICA applied to the gene factor matrix and counter-rotate the core tensor (equation 4A). In the second step, we optimize the core tensor by the varimax rotation and counter-rotate the donor matrix (equation 4B). This is calculated as follows:StepStepHere, all variables are as previously described, with the addition of 0 , which represents the ICA rotated gene factor matrix, and which now represents the orthonormal rotation matrix found by varimax when applied to 0 (!) % . The symbol ⊗ is the Kronecker product. For this approach, we use the identity matrix for . For all of our analyses, we also set the ranks of equal to the number of cell types, since we used a relatively small number of cell types. To project a multicellular pattern onto new data to obtain donor scores (as in Figure 7F ), we first compute loadings as (!) = 0 (!) * 4 ⊗ 0 6 % without rotating the core. Then, we calculate the new donor scores by ()*+ = (!) * (!) % . Then, the columns of the new scores are normalized to a magnitude of 1 and are rotated by applying the varimax rotation matrix from the core optimization as 1 ()*+ = ()*+ * % !" .For visualization purposes, we order the factors in the donor scores matrix from highest to lowest explained variance. To calculate the variance explained by a given factor, we first compute the reconstructed tensor 9 using only the selected factor. Then variance explained is simply the calculation for the coefficient of variation:The subscript indicates the factor used for reconstructing the data. The subscript on the double brackets indicates the Frobenius norm. In the loadings matrices, we also display the