Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Resource
  • Published:

The immune cell landscape in kidneys of patients with lupus nephritis

A Publisher Correction to this article was published on 13 August 2019

This article has been updated

Abstract

Lupus nephritis is a potentially fatal autoimmune disease for which the current treatment is ineffective and often toxic. To develop mechanistic hypotheses of disease, we analyzed kidney samples from patients with lupus nephritis and from healthy control subjects using single-cell RNA sequencing. Our analysis revealed 21 subsets of leukocytes active in disease, including multiple populations of myeloid cells, T cells, natural killer cells and B cells that demonstrated both pro-inflammatory responses and inflammation-resolving responses. We found evidence of local activation of B cells correlated with an age-associated B-cell signature and evidence of progressive stages of monocyte differentiation within the kidney. A clear interferon response was observed in most cells. Two chemokine receptors, CXCR4 and CX3CR1, were broadly expressed, implying a potentially central role in cell trafficking. Gene expression of immune cells in urine and kidney was highly correlated, which would suggest that urine might serve as a surrogate for kidney biopsies.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: An overview of the approach used for analyzing the cellular contents and molecular states of kidney and urine samples.
Fig. 2: A summary of the stepwise clustering of kidney cells.
Fig. 3: Focused analysis of kidney myeloid cells.
Fig. 4: Focused analysis of kidney T cells and NK cells.
Fig. 5: Focused analysis of kidney B cells.
Fig. 6: The expression of GWAS genes in LN kidneys.
Fig. 7: Chemokine- and cytokine-mediated cellular networks.
Fig. 8: Comparison of immune cells extracted from urine samples and from kidney samples.

Similar content being viewed by others

Data availability

The data reported in this publication, including the clinical and serological data of the study participants, are deposited in the ImmPort repository (accession code SDY997). The raw single-cell RNA-seq data are also deposited in dbGAP (accession code phs001457.v1.p1). The processed data can be viewed using an interactive browser at https://immunogenomics.io/ampsle, https://immunogenomics.io/cellbrowser/ and https://portals.broadinstitute.org/single_cell/study/amp-phase-1.

Code availability

All R scripts used to analyze the data reported in this publication are available from the corresponding authors on request.

Change history

  • 13 August 2019

    An amendment to this paper has been published and can be accessed via a link at the top of the paper.

References

  1. Costenbader, K. H. et al. Trends in the incidence, demographics, and outcomes of end-stage renal disease due to lupus nephritis in the US from 1995 to 2006. Arthritis Rheum. 63, 1681–1688 (2011).

    Article  Google Scholar 

  2. Narain, S. & Furie, R. Update on clinical trials in systemic lupus erythematosus. Curr. Opin. Rheumatol. 28, 477–487 (2016).

    Article  CAS  Google Scholar 

  3. Tektonidou, M. G., Dasgupta, A. & Ward, M. M. Risk of end-stage renal disease in patients with lupus nephritis, 1971–2015: a systematic review and Bayesian meta-analysis. Arthritis Rheumatol. 68, 1432–1441 (2016).

    Article  Google Scholar 

  4. Thanou, A. & Merrill, J. T. Treatment of systemic lupus erythematosus: new therapeutic avenues and blind alleys. Nat. Rev. Rheumatol. 10, 23–34 (2014).

    Article  CAS  Google Scholar 

  5. Banchereau, R. et al. Personalized immunomonitoring uncovers molecular networks that stratify lupus patients. Cell 165, 1548–1550 (2016).

    Article  CAS  Google Scholar 

  6. Liarski, V. M. et al. Cell distance mapping identifies functional T follicular helper cells in inflamed human renal tissue. Sci. Transl. Med. 6, 230ra246 (2014).

    Article  Google Scholar 

  7. Hutloff, A. et al. Involvement of inducible costimulator in the exaggerated memory B cell and plasma cell generation in systemic lupus erythematosus. Arthritis Rheum. 50, 3211–3220 (2004).

    Article  Google Scholar 

  8. Kassianos, A. J. et al. Increased tubulointerstitial recruitment of human CD141(hi) CLEC9A(+) and CD1c(+) myeloid dendritic cell subsets in renal fibrosis and chronic kidney disease. Am. J. Physiol. Renal Physiol. 305, F1391–F1401 (2013).

    Article  CAS  Google Scholar 

  9. Davidson, A. What is damaging the kidney in lupus nephritis? Nat. Rev. Rheumatol. 12, 143–153 (2016).

    Article  CAS  Google Scholar 

  10. Hashimshony, T. et al. CEL-Seq2: sensitive highly-multiplexed single-cell RNA-Seq. Genome Biol. 17, 77 (2016).

    Article  Google Scholar 

  11. Hooks, J. J. et al. Immune interferon in the circulation of patients with autoimmune disease. N. Engl. J. Med. 301, 5–8 (1979).

    Article  CAS  Google Scholar 

  12. Villani, A. C. et al. Single-cell RNA-seq reveals new types of human blood dendritic cells, monocytes, and progenitors. Science 356, eaah4573 (2017).

    Article  Google Scholar 

  13. Ghebrehiwet, B., Hosszu, K. H. & Peerschke, E. I. C1q as an autocrine and paracrine regulator of cellular functions. Mol. Immunol. 84, 26–33 (2017).

    Article  CAS  Google Scholar 

  14. Hulsebus, H. J., O’Conner, S. D., Smith, E. M., Jie, C. & Bohlson, S. S. Complement component C1q programs a pro-efferocytic phenotype while limiting TNFalpha production in primary mouse and human macrophages. Front. Immunol. 7, 230 (2016).

    Article  Google Scholar 

  15. Ikezumi, Y. et al. The sialoadhesin (CD169) expressing a macrophage subset in human proliferative glomerulonephritis. Nephrol. Dial. Transplant. 20, 2704–2713 (2005).

    Article  CAS  Google Scholar 

  16. Knutson, M. D. Iron transport proteins: gateways of cellular and systemic iron homeostasis. J. Biol. Chem. 292, 12735–12743 (2017).

    Article  CAS  Google Scholar 

  17. Adamson, S. E. et al. Disabled homolog 2 controls macrophage phenotypic polarization and adipose tissue inflammation. J. Clin. Invest. 126, 1311–1322 (2016).

    Article  Google Scholar 

  18. Spadaro, O. et al. IGF1 shapes macrophage activation in response to immunometabolic challenge. Cell Rep. 19, 225–234 (2017).

    Article  CAS  Google Scholar 

  19. Varghese, B., Paulos, C. & Low, P. S. Optimization of folate-targeted immunotherapy for the treatment of experimental arthritis. Inflammation 39, 1345–1353 (2016).

    Article  CAS  Google Scholar 

  20. Kreslavsky, T. et al. Essential role for the transcription factor Bhlhe41 in regulating the development, self-renewal and BCR repertoire of B-1a cells. Nat. Immunol. 18, 442–455 (2017).

    Article  CAS  Google Scholar 

  21. Angerer, P. et al. destiny: diffusion maps for large-scale single-cell data in R. Bioinformatics 32, 1241–1243 (2016).

    Article  CAS  Google Scholar 

  22. van den Maaten, L. J. P. H. G. Visualizing high-dimensional data using t-SNE. J. Mach. Learn. Res. 9, 2579–2605 (2008).

    Google Scholar 

  23. Dehn, S. & Thorp, E. B. Myeloid receptor CD36 is required for early phagocytosis of myocardial infarcts and induction of Nr4a1-dependent mechanisms of cardiac repair. FASEB J. 32, 254–264 (2017).

    Article  Google Scholar 

  24. Bengsch, B. et al. Deep immune profiling by mass cytometry links human T and NK cell differentiation and cytotoxic molecule expression patterns. J. Immunol. Methods 453, 3–10 (2017).

    Article  Google Scholar 

  25. Bratke, K., Kuepper, M., Bade, B., Virchow, J. C. Jr. & Luttmann, W. Differential expression of human granzymes A, B, and K in natural killer cells and during CD8+ T cell differentiation in peripheral blood. Eur. J. Immunol. 35, 2608–2616 (2005).

    Article  CAS  Google Scholar 

  26. Boddupalli, C. S. et al. ABC transporters and NR4A1 identify a quiescent subset of tissue-resident memory T cells. J. Clin. Invest. 126, 3905–3916 (2016).

    Article  Google Scholar 

  27. Mackay, L. K. et al. Hobit and Blimp1 instruct a universal transcriptional program of tissue residency in lymphocytes. Science 352, 459–463 (2016).

    Article  CAS  Google Scholar 

  28. Allan, D. S. J. et al. Transcriptome analysis reveals similarities between human blood CD3(−) CD56(bright) cells and mouse CD127(+) innate lymphoid cells. Sci. Rep. 7, 3501 (2017).

    Article  Google Scholar 

  29. McKinney, E. F., Lee, J. C., Jayne, D. R., Lyons, P. A. & Smith, K. G. T-cell exhaustion, co-stimulation and clinical outcome in autoimmunity and infection. Nature 523, 612–616 (2015).

    Article  CAS  Google Scholar 

  30. Tilstra, J. S. et al. Kidney-infiltrating T cells in murine lupus nephritis are metabolically and functionally exhausted. J. Clin. Invest. 128, 4884–4897 (2018).

    Article  Google Scholar 

  31. Wherry, E. J. et al. Molecular signature of CD8+ T cell exhaustion during chronic viral infection. Immunity 27, 670–684 (2007).

    Article  CAS  Google Scholar 

  32. Nish, S. A. et al. CD4+ T cell effector commitment coupled to self-renewal by asymmetric cell divisions. J. Exp. Med. 214, 39–47 (2017).

    Article  CAS  Google Scholar 

  33. Karnell, J. L. et al. Role of CD11c(+) T-bet(+) B cells in human health and disease. Cell. Immunol. 321, 40–45 (2017).

    Article  CAS  Google Scholar 

  34. Jenks, S. A. et al. Distinct effector B cells induced by unregulated Toll-like receptor 7 contribute to pathogenic responses in systemic lupus erythematosus. Immunity 49, 725–739.e6 (2018).

    Article  CAS  Google Scholar 

  35. The FANTOM Consortium and the RIKEN PMI and CLST (DGT). A promoter-level mammalian expression atlas. Nature 507, 462–470 (2014).

  36. Lizio, M. et al. Gateways to the FANTOM5 promoter level mammalian expression atlas. Genome Biol. 16, 22 (2015).

    Article  CAS  Google Scholar 

  37. Wang, S. et al. IL-21 drives expansion and plasma cell differentiation of autoreactive CD11chiT-bet+ B cells in SLE. Nat. Commun. 9, 1758 (2018).

    Article  Google Scholar 

  38. Chen, L., Morris, D. L. & Vyse, T. J. Genetic advances in systemic lupus erythematosus: an update. Curr. Opin. Rheumatol. 29, 423–433 (2017).

    Article  Google Scholar 

  39. Chung, S. A. et al. Lupus nephritis susceptibility loci in women with systemic lupus erythematosus. J. Am. Soc. Nephrol. 25, 2859–2870 (2014).

    Article  CAS  Google Scholar 

  40. Hua, Z. & Hou, B. TLR signaling in B-cell development and activation. Cell. Mol. Immunol. 10, 103–106 (2013).

    Article  CAS  Google Scholar 

  41. Kawasaki, T. & Kawai, T. Toll-like receptor signaling pathways. Front. Immunol. 5, 461 (2014).

    Article  Google Scholar 

  42. Madan-Lala, R. et al. Mycobacterium tuberculosis impairs dendritic cell functions through the serine hydrolase Hip1. J. Immunol. 192, 4263–4272 (2014).

    Article  CAS  Google Scholar 

  43. Matsuda, S. et al. Regulation of the cell cycle and inflammatory arthritis by the transcription cofactor LBH gene. J. Immunol. 199, 2316–2322 (2017).

    Article  CAS  Google Scholar 

  44. Botta, D. et al. Dynamic regulation of T follicular regulatory cell responses by interleukin 2 during influenza infection. Nat. Immunol. 18, 1249–1260 (2017).

    Article  CAS  Google Scholar 

  45. Wing, J. B. et al. A distinct subpopulation of CD25(−) T-follicular regulatory cells localizes in the germinal centers. Proc. Natl Acad. Sci. USA 114, E6400–e6409 (2017).

    Article  CAS  Google Scholar 

  46. Winchester, R. et al. Immunologic characteristics of intrarenal T cells: trafficking of expanded CD8+ T cell β-chain clonotypes in progressive lupus nephritis. Arthritis Rheum. 64, 1589–1600 (2012).

    Article  CAS  Google Scholar 

  47. Peterson, K. S. et al. Characterization of heterogeneity in the molecular pathogenesis of lupus nephritis from transcriptional profiles of laser-captured glomeruli. J. Clin. Invest. 113, 1722–1733 (2004).

    Article  CAS  Google Scholar 

  48. Davidson, A. Editorial: autoimmunity to vimentin and lupus nephritis. Arthritis Rheumatol. 66, 3251–3254 (2014).

    Article  CAS  Google Scholar 

  49. Myles, A., Gearhart, P. J. & Cancro, M. P. Signals that drive T-bet expression in B cells. Cell. Immunol. 321, 3–7 (2017).

    Article  CAS  Google Scholar 

  50. Huen, S. C. & Cantley, L. G. Macrophages in renal injury and repair. Ann. Rev. Physiol. 79, 449–469 (2017).

    Article  CAS  Google Scholar 

  51. Tabas, I. & Bornfeldt, K. E. Macrophage phenotype and function in different stages of atherosclerosis. Circ. Res. 118, 653–667 (2016).

    Article  CAS  Google Scholar 

  52. Auffray, C. et al. Monitoring of blood vessels and tissues by a population of monocytes with patrolling behavior. Science 317, 666–670 (2007).

    Article  CAS  Google Scholar 

  53. Picelli, S. et al. Full-length RNA-seq from single cells using Smart-seq2. Nat. Protoc. 9, 171–181 (2014).

    Article  CAS  Google Scholar 

  54. Nemesh, J. Drop-seq core computational protocol. McCarroll Laboratory http://mccarrolllab.com/wp-content/uploads/2016/03/Drop-seqAlignmentCookbookv1.2Jan2016.pdf (2016).

  55. Satija, R., Farrell, J. A., Gennert, D., Schier, A. F. & Regev, A. Spatial reconstruction of single-cell gene expression data. Nat. Biotechnol. 33, 495–502 (2015).

    Article  CAS  Google Scholar 

  56. McDavid, A. et al. Data exploration, quality control and testing in single-cell qPCR-based gene expression experiments. Bioinformatics 29, 461–467 (2013).

    Article  CAS  Google Scholar 

  57. Benjamini, Y. & Hochberg, Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J. R. Statist. Soc. B 57, 289–300 (1995).

    Google Scholar 

  58. Tirosh, I. et al. Dissecting the multicellular ecosystem of metastatic melanoma by single-cell RNA-seq. Science 352, 189–196 (2016).

    Article  CAS  Google Scholar 

  59. IUPHAR/BPS. Guide to Pharmacology. http://www.guidetopharmacology.org/download.jsp (2019).

Download references

Acknowledgements

This work was supported by the Accelerating Medicines Partnership (AMP) in Rheumatoid Arthritis and Lupus Network. AMP is a public–private partnership (AbbVie Inc., Arthritis Foundation, Bristol-Myers Squibb Company, Foundation for the National Institutes of Health, Janssen Pharmaceuticals, Lupus Foundation of America, Lupus Research Alliance, Merck Sharp & Dohme Corp., National Institute of Allergy and Infectious Diseases, National Institute of Arthritis and Musculoskeletal and Skin Diseases, Pfizer Inc., Rheumatology Research Foundation, Sanofi and Takeda Pharmaceuticals International, Inc.) created to develop new ways of identifying and validating promising biological targets for diagnostics and drug development. Funding was provided through grants from the National Institutes of Health (UH2-AR067676, UH2-AR067677, UH2-AR067679, UH2-AR067681, UH2-AR067685, UH2-AR067688, UH2-AR067689, UH2-AR067690, UH2-AR067691, UH2-AR067694 and UM2-AR067678). N.H. was supported by the David P. Ryan, MD Endowed Chair in Cancer Research. We thank participating Lupus Nephritis Trials Network clinical sites and participants.

Author information

Authors and Affiliations

Authors

Consortia

Contributions

A.A., D.A.R, A.D., P.J.H., A.H.J. and D.J.L. analyzed the data. D.A.R., C.C.B., T.M.E., E.P.B., J.A.L. and D.A.H. developed the sample collection and processing protocols. D.A.R., Y.L., P.J.H., A.C., A.N., D.S. and S.S. processed the samples. S.L., D.J.L., A.N., D.S. and S.S. developed the scRNA-seq library preparation protocol. F.Z. and K.S. developed the web-based browser of the data. D.E.S., P.T., E.M., M.D.E., M.P., D.L.K., R.A.F., F.P.S., W.F.P., E.A.M., J.P.B., M.A.P., C.P., K.C.K., E.S.W., D.A.H., D.W. and J.H.A. acquired samples. A.A., D.A.R., C.C.B., A.D., W.A., J.A.L., D.A.H., C.N., D.W., M.K., J.H.A., M.B.B., N.H. and B.D. designed the study. A.D., W.A., D.A.H., C.N., S.R., D.W., M.K., J.H.A., M.B.B., N.H. and B.D. supervised the work. A.A., D.A.R., C.C.B., A.D., P.J.H., N.H. and B.D. wrote the manuscript.

Corresponding authors

Correspondence to Nir Hacohen or Betty Diamond.

Ethics declarations

Competing interests

M.B.B. is a consultant to Roche in the area of stromal cells.

Integrated supplementary information

Supplementary Figure 1 Analysis of cells obtained from cryopreserved viable kidney tissue.

a, Leukocyte yields from kidney tissue samples processed fresh or after freeze/thaw. b, Leukocyte yields from kidney tissue samples cryopreserved either post-dissociation (frozen cells) or pre-dissociation (frozen tissue). n = 9 segments pooled from 4 different donors. * p < 0.01 by two-tailed Mann-Whitney U-test. c, Cell yields and leukocyte frequencies from kidney samples either cryopreserved or shipped overnight on wet ice. Wilcoxon matched-pairs signed rank test. d, Example of flow cytometry analysis of cells from a cryopreserved lupus nephritis kidney biopsy. e, RNA-seq quality metrics of bulk leukocytes from kidney cells analyzed before (fresh cells) and after freeze/thaw (frozen cells), or obtained from kidney tissue shipped overnight or shipped after tissue cryopreservation (frozen tissue). f-h, The distribution of the number of genes per cell on each processed 384-well plate, in kidney leukocytes (f), kidney epithelial cells (g) and urine leukocytes (h).

Supplementary Figure 2 An overview of the isolated cells.

a-d, Leukocyte yields and composition from LN kidney biopsies, as determined by flow cytometry. a, Number of leukocytes sorted as single cells from LN and control biopsy samples. b, Number of leukocytes sorted from LN biopsies stratified by histologic glomerulonephritis class. c, Flow cytometric assessment of the proportion of leukocytes that are B cells, T cells, monocyte/macrophages, or other in biopsy samples. d, Percentage of B cells among leukocytes in biopsy samples (p = 0.0031; two-tailed Mann-Whitney U-test). e, Projection of the gene expression data of kidney cells following principal component analysis (PCA), in the PC1-PC2 (left) and PC2-PC3 (right) planes. Cells are colored based on their identity as either leukocytes (cyan) or epithelial cells (pink), as determined by flow cytometry. The labeling of clusters in the PC2-PC3 plane is based on the identity of the leading genes in each principal component. f, Same as (e), except cells are colored based on the 384-well plate they were processed on. g, The expression of selected genes in the 10 low-resolution clusters; these were among the genes used to determine the putative lineage of each cell. h, The number of clusters encountered as a function of the number of patients included in the analysis. The presented curve was generated by randomly down-sampling the number of patients analyzed, while counting in each case the number of clusters spanned by the cells of these patients. Patients that had a small number of high quality cells (less than 70) were not included in this analysis, as such patients would lead to a false saturation. Furthermore, to account for the fact that for a cluster to be identified it should contain a sufficient number of cells, we only counted here clusters that had more cells than the smallest cluster identified when analyzing the full cohort (17 cells—cluster CB2b). i, A tSNE plot of the analyzed cells, highlighting the LD controls as opposed to the LN patients. j, A tSNE plot of the analyzed cells, colored by the donor identity.

Supplementary Figure 3 Focused analysis of kidney myeloid cells.

a, The expression of selected genes over the 5 myeloid cell clusters. b-c, The expression of the canonical markers CD16 (FCGR3A) and CD14 on the 5 myeloid clusters. d-f, The changes in the expression of 3 selected genes, along the trajectory from CM0 to CM4; ‘pseudotime’ represents the ordering of the cells along this trajectory. The violin plots (shades) show the distribution of expression levels in equally-spaced intervals along the pseudotime axis (and do not directly correspond to cell clusters). g, The scRNA-seq data of myeloid blood cells sorted from 2 LN patients, projected in the tSNE plane. Low-resolution clustering identified a cluster of CD16+ monocytes; other myeloid cells are grouped here into an ‘other’ set. h, The results of comparing the blood CD16+ monocytes to the 5 myeloid cells clusters found in kidney (CM0-CM4). For each blood cell, the most similar kidney cluster was determined; the bar plot denotes the percentage of myeloid cells most similar to each of the kidney myeloid clusters (only values larger than 0 are shown). i, The increase in the phagocytosis score, computed as the average expression of several genes associated with phagocytosis, in cluster CM1 compared to cluster CM0, in blood myeloid cells. j, Same as (i), but with regard to kidney myeloid cells. ***—p-value < 0.001 (two-tailed Mann-Whitney U-test).

Supplementary Figure 4 Focused analysis of kidney T cells and NK cells.

a, The expression of selected genes across the 7 high-level T/NK cell clusters. b, The expression of selected genes across the two subclusters of cluster CT5, and in comparison to cluster CT1. c, The expression of genes associated with exhaustion, across the different T/NK cell clusters. d, A heatmap showing the separation of LN patients (orange in line above heatmap) and healthy controls (blue), based on expression data of CD8+ T cells sorted from blood samples, and considering a published list of exhaustion markers. Both rows and columns are clustered based on Euclidian distance. Note that out of the 10 LN patients for which blood samples were available, only 8 had sufficient numbers of blood CD8+ T cells to allow sequencing. e, The exhaustion score as calculated in blood CD8+ T cells, comparing the 8 LN patients to the 10 healthy controls. Each point represents a single patient; lines represent the median per group. **—p-value < 0.01 (two-tailed Mann-Whitney U-test). f, The exhaustion score in kidney CD8+ T cells, comparing LN patients to LD controls (same 8 LN patients as in (e)). Each point refers to a cell. g, The expression of selected genes across the two subclusters of cluster CT3. h, The expression of selected genes across the two subclusters of cluster CT0a.

Supplementary Figure 5 Focused analysis of kidney B cells.

a, The expression of selected genes, across the 4 high-level B cell clusters. b, A tSNE projection of all B cells. Note that the cells of cluster CB2 (in cyan) occupy two main separate regions in the tSNE1-tSNE2 plane. c, The expression of selected genes, across the two subclusters of cluster CB2. d-f, The results of classifying the CB2 cells by correlating their gene expression with that of 3 sets of reference samples—FANTOM5 (d), 13 immune cell populations sorted from healthy individuals (Browne et al.; e), and the clusters identified in Villani et al. (f). For each of the 2 subclusters in CB2, the bars denote the percentage of cells most similar to each of the reference samples. For readability, only relevant reference samples are specified, and the rest are collapsed into an ‘other’ category. For all cells in all comparisons, the correlation used for classification was above the corresponding assignability threshold. Note that the reference samples in Villani et al. do not include B cells, and therefore are not relevant for the classification of the cells in cluster CB2a.

Supplementary Figure 6 Focused analysis of cluster C9.

a, The identification of 3 subclusters, the first of which pertains to cells undergoing mitosis. b, The percentage of reads mapped to mitochondrial genes in each subcluster. c, Classification results for each subcluster, comparing the cells of cluster C9 to reference samples found in FANTOM5.

Supplementary Figure 7 Analysis of chemokines, cytokines and their receptors.

a, The distribution of the maximum fraction of expressing cells (taken over all clusters for each receptor), for all receptors. Based on this distribution, a threshold of 0.3 was chosen to define frequently expressed receptors. b–f, The percentage of cells expressing selected chemokine receptors, specified for each cluster. The dashed line in all panels pertains to the above threshold. g–j, The distribution of expression levels of selected chemokines, for each cluster.

Supplementary Figure 8 Analysis of urine cells.

A comparison of gene expression in kidney (x axis) and urine (y axis), for 3 of the clusters represented in both compartments.

Supplementary information

Supplementary Information

Supplementary Figs. 1–8.

Reporting Summary

Supplementary Table 1: Clinical characteristics of kidney biopsy donors.

Two worksheets are provided—one containing a summary of the clinical characteristics of the studied cohort, the other containing detailed information per sample.

Supplementary Table 2: Sensitivity analysis of clustering parameters.

The results of a detailed sensitivity analysis with regard to the low-resolution clustering step, varying several parameters. Consistency of clustering across various parameter combinations, as defined using the Rand index, is reported.

Supplementary Table 3: Number of cells per cluster and patient.

Rows correspond to patients; columns correspond to clusters.

Supplementary Table 4: Number of cells per cluster and processed 384-well plate.

Rows correspond to plates; columns correspond to clusters.

Supplementary Table 5: Differential expression analysis, comparing LN and LD samples.

Results are reported (in separate worksheets) per cluster, looking only at clusters that had a sufficient number of cells in the LD samples

Supplementary Table 6: Gene lists.

The various gene lists that were used throughout the analysis, each in a separate worksheet.

Supplementary Table 7: Myeloid cells classification by correlation.

The Pearson correlation scores, comparing each kidney myeloid cell (rows) to each reference sample (columns) in Villani et al.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Arazi, A., Rao, D.A., Berthier, C.C. et al. The immune cell landscape in kidneys of patients with lupus nephritis. Nat Immunol 20, 902–914 (2019). https://doi.org/10.1038/s41590-019-0398-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-019-0398-x

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing