Skip to main content

Advertisement

Log in

Single-cell multi-omic and spatial profiling of human kidneys implicates the fibrotic microenvironment in kidney disease progression

  • Article
  • Published:

From Nature Genetics

View current issue Submit your manuscript

Abstract

Kidneys are intricate three-dimensional structures in the body, yet the spatial and molecular principles of kidney health and disease remain inadequately understood. We generated high-quality datasets for 81 samples, including single-cell, single-nuclear, spot-level (Visium) and single-cell resolution (CosMx) spatial-RNA expression and single-nuclear open chromatin, capturing cells from healthy, diabetic and hypertensive diseased human kidneys. Combining these data, we identify cell types and map them to their locations within the tissue. Unbiased deconvolution of the spatial data identifies the following four distinct microenvironments: glomerular, immune, tubule and fibrotic. We describe the complex organization of microenvironments in health and disease and find that the fibrotic microenvironment is able to molecularly classify human kidneys and offers an improved prognosis compared to traditional histopathology. We provide a comprehensive spatially resolved molecular roadmap of the human kidney and the fibrotic process, demonstrating the clinical utility of spatial transcriptomics.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1: Comprehensive integrated multimodal human kidney single-cell atlas.
Fig. 2: Spatially resolved human kidneys.
Fig. 3: Human kidney stromal cell atlas.
Fig. 4: Human kidney FME.
Fig. 5: iPT cells in diseased human kidneys.
Fig. 6: FME-GS successfully predicts disease prognosis in a large cohort of human kidney samples.

Similar content being viewed by others

Data availability

Raw data, processed data and metadata from the snRNA-seq, scRNA-seq, snATAC–seq and spRNA-seq have been deposited in Gene Expression Omnibus with the accession code GSE211785. The human bulk kidney RNA-seq data are available under the following accession numbers: GSE115098 and GSE173343. The single-cell and nuclear expression and open chromatin and spatial data are also available at www.susztaklab.com (https://susztaklab.com/hk_genemap/).

Code availability

All the codes used for the analysis were deposited on GitHub (https://github.com/amin69upenn/Human_Kidney_Multiomics_and_Spatial_Atlas_ and https://github.com/jlevins2010/FME_atlas).

References

  1. Gueutin, V., Deray, G. & Isnard-Bagnis, C. Renal physiology. Bull. Cancer 99, 237–249 (2012).

    Article  CAS  PubMed  Google Scholar 

  2. Smith, H. W. The fate of sodium and water in the renal tubules. Bull. N. Y. Acad. Med. 35, 293 (1959).

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Vart, P. et al. National trends in the prevalence of chronic kidney disease among racial/ethnic and socioeconomic status groups, 1988–2016. JAMA Netw. Open 3, e207932 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  4. Yinusa, A., Faezipour, M. & Faezipour, M. A study on CKD progression and health disparities using system dynamics modeling. Healthcare 10, 1628 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  5. Kakitapalli, Y., Ampolu, J., Madasu, S. D. & Kumar, M. S. Detailed review of chronic kidney disease. Kidney Dis. 6, 85–91 (2020).

    Article  Google Scholar 

  6. Balzer, M. S., Rohacs, T. & Susztak, K. How many cell types are in the kidney and what do they do? Annu. Rev. Physiol. 84, 507 (2022).

    Article  CAS  PubMed  Google Scholar 

  7. Schreibing, F. & Kramann, R. Mapping the human kidney using single-cell genomics. Nat. Rev. Nephrol. 18, 347–360 (2022).

    Article  PubMed  Google Scholar 

  8. Balzer, M. S. et al. Single-cell analysis highlights differences in druggable pathways underlying adaptive or fibrotic kidney regeneration. Nat. Commun. 13, 4018 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Miao, Z. et al. Single cell regulatory landscape of the mouse kidney highlights cellular differentiation programs and disease targets. Nat. Commun. 12, 2277 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Muto, Y. et al. Single cell transcriptional and chromatin accessibility profiling redefine cellular heterogeneity in the adult human kidney. Nat. Commun. 12, 2190 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Wilson, P. C. et al. The single-cell transcriptomic landscape of early human diabetic nephropathy. Proc. Natl Acad. Sci. USA 116, 19619–19625 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Young, M. D. et al. Single-cell transcriptomes from human kidneys reveal the cellular identity of renal tumors. Science 361, 594–599 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Dixon, E. E., Wu, H., Muto, Y., Wilson, P. C. & Humphreys, B. D. Spatially resolved transcriptomic analysis of acute kidney injury in a female murine model. J. Am. Soc. Nephrol. 33, 279–289 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Chen, T. K., Knicely, D. H. & Grams, M. E. Chronic kidney disease diagnosis and management: a review. JAMA 322, 1294–1304 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Palmer, M. B. et al. The role of glomerular epithelial injury in kidney function decline in patients with diabetic kidney disease in the TRIDENT cohort. Kidney Int. Rep. 6, 1066–1080 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  16. Armutcu, F. Organ crosstalk: the potent roles of inflammation and fibrotic changes in the course of organ interactions. Inflamm. Res. 68, 825–839 (2019).

    Article  CAS  PubMed  Google Scholar 

  17. Majo, J., Klinkhammer, B. M., Boor, P. & Tiniakos, D. Pathology and natural history of organ fibrosis. Curr. Opin. Pharmacol. 49, 82–89 (2019).

    Article  CAS  PubMed  Google Scholar 

  18. Deng, Z. et al. The extracellular matrix and mechanotransduction in pulmonary fibrosis. Int. J. Biochem. Cell Biol. 126, 105802 (2020).

    Article  CAS  PubMed  Google Scholar 

  19. Humphreys, B. D. Mechanisms of renal fibrosis. Annu. Rev. Physiol. 80, 309–326 (2018).

    Article  CAS  PubMed  Google Scholar 

  20. Parimon, T., Hohmann, M. S. & Yao, C. Cellular senescence: pathogenic mechanisms in lung fibrosis. Int. J. Mol. Sci. 22, 6214 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Lausecker, F., Lennon, R. & Randles, M. J. The kidney matrisome in health, aging and disease. Kidney Int. 102, 1000–1012 (2022).

    Article  CAS  PubMed  Google Scholar 

  22. Zhou, D. & Liu, Y. Understanding the mechanisms of kidney fibrosis. Nat. Rev. Nephrol. 12, 68–70 (2016).

    Article  CAS  PubMed  Google Scholar 

  23. Grams, M. E. et al. Predicting timing of clinical outcomes in patients with chronic kidney disease and severely decreased glomerular filtration rate. Kidney Int. 93, 1442–1451 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  24. Hallan, S. I. et al. Combining GFR and albuminuria to classify CKD improves prediction of ESRD. J. Am. Soc. Nephrol. 20, 1069–1077 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Neuen, B. L. et al. Changes in GFR and albuminuria in routine clinical practice and the risk of kidney disease progression. Am. J. Kidney Dis. 78, 350–360 (2021).

    Article  CAS  PubMed  Google Scholar 

  26. An, Y. et al. Renal histologic changes and the outcome in patients with diabetic nephropathy. Nephrol. Dial. Transplant. 30, 257–266 (2015).

    Article  CAS  PubMed  Google Scholar 

  27. Rodríguez-Iturbe, B., Johnson, R. R. & Herrera-Acosta, J. Tubulointerstitial damage and progression of renal failure. Kidney Int. 68, S82–S86 (2005).

    Article  Google Scholar 

  28. Srivastava, A. et al. The prognostic value of histopathologic lesions in native kidney biopsy specimens: results from the Boston Kidney Biopsy Cohort study. J. Am. Soc. Nephrol. 29, 2213–2224 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  29. Luecken, M. D. et al. Benchmarking atlas-level data integration in single-cell genomics. Nat. Methods 19, 41–50 (2022).

    Article  CAS  PubMed  Google Scholar 

  30. Lake, B. B. et al. An atlas of healthy and injured cell states and niches in the human kidney. Nature 619, 585–594 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Kleshchevnikov, V. et al. Cell2location maps fine-grained cell types in spatial transcriptomics. Nat. Biotechnol. 40, 661–671 (2022).

    Article  CAS  PubMed  Google Scholar 

  32. Wei, R. et al. Spatial charting of single-cell transcriptomes in tissues. Nat. Biotechnol. 40, 1190–1199 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Thul, P. J. & Lindskog, C. The Human Protein Atlas: a spatial map of the human proteome. Protein Sci. 27, 233–244 (2018).

    Article  CAS  PubMed  Google Scholar 

  34. Kuppe, C. et al. Decoding myofibroblast origins in human kidney fibrosis. Nature 589, 281–286 (2021).

    Article  CAS  PubMed  Google Scholar 

  35. Naba, A. et al. The extracellular matrix: tools and insights for the ‘omics’ era. Matrix Biol. 49, 10–24 (2016).

    Article  CAS  PubMed  Google Scholar 

  36. Crow, M., Paul, A., Ballouz, S., Huang, Z. J. & Gillis, J. Characterizing the replicability of cell types defined by single cell RNA-sequencing data using MetaNeighbor. Nat. Commun. 9, 884 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  37. Langfelder, P. & Horvath, S. WGCNA: an R package for weighted correlation network analysis. BMC Bioinformatics 9, 559 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  38. Morabito, S. et al. Single-nucleus chromatin accessibility and transcriptomic characterization of Alzheimer’s disease. Nat. Genet. 53, 1143–1155 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Nirmal, A. J. et al. The spatial landscape of progression and immunoediting in primary melanoma at single cell resolution. Cancer Discov. 12, 1518 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Sato, Y., Tamura, M. & Yanagita, M. Tertiary lymphoid tissues: a regional hub for kidney inflammation. Nephrol. Dial. Transplant. 38, 26–33 (2021).

    Article  Google Scholar 

  41. Jin, S. et al. Inference and analysis of cell–cell communication using CellChat. Nat. Commun. 12, 1088 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Cang, Z. et al. Screening cell–cell communication in spatial transcriptomics via collective optimal transport. Nat. Methods 20, 218–228 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Wilson, P. C. et al. Multimodal single cell sequencing implicates chromatin accessibility and genetic background in diabetic kidney disease progression. Nat. Commun. 13, 5253 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Wu, H. et al. Mapping the single-cell transcriptomic response of murine diabetic kidney disease to therapies. Cell Metab. 34, 1064–1078 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Kluger, A. Y. et al. Class effects of SGLT2 inhibitors on cardiorenal outcomes. Cardiovasc. Diabetol. 18, 99 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  46. Park, J. et al. Single-cell transcriptomics of the mouse kidney reveals potential cellular targets of kidney disease. Science 360, 758–763 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Zhang, L. et al. Genome-wide analysis of histone H3 lysine 4 trimethylation in peripheral blood mononuclear cells of minimal change nephrotic syndrome patients. Am. J. Nephrol. 30, 505–513 (2009).

    Article  PubMed  Google Scholar 

  48. Wynn, T. A. & Ramalingam, T. R. Mechanisms of fibrosis: therapeutic translation for fibrotic disease. Nat. Med. 18, 1028–1040 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Djudjaj, S. & Boor, P. Cellular and molecular mechanisms of kidney fibrosis. Mol. Aspects Med. 65, 16–36 (2019).

    Article  CAS  PubMed  Google Scholar 

  50. Liu, M. & Gomez, D. Smooth muscle cell phenotypic diversity: at the crossroads of lineage tracing and single-cell transcriptomics. Arterioscler. Thromb. Vasc. Biol. 39, 1715–1723 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Pedroza, A. J. et al. Single-cell transcriptomic profiling of vascular smooth muscle cell phenotype modulation in Marfan syndrome aortic aneurysm. Arterioscler. Thromb. Vasc. Biol. 40, 2195–2211 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Zhang, F., Guo, X., Xia, Y. & Mao, L. An update on the phenotypic switching of vascular smooth muscle cells in the pathogenesis of atherosclerosis. Cell. Mol. Life Sci. 79, 6 (2022).

    Article  CAS  Google Scholar 

  53. Aghajanian, H. et al. Targeting cardiac fibrosis with engineered T cells. Nature 573, 430–433 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Aghajanian, H., Rurik, J. G. & Epstein, J. A. CAR-based therapies: opportunities for immuno-medicine beyond cancer. Nat. Metab. 4, 163–169 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  55. Rurik, J. G. et al. CAR T cells produced in vivo to treat cardiac injury. Science 375, 91–96 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Song, J. et al. Understanding kidney injury molecule 1: a novel immune factor in kidney pathophysiology. Am. J. Transl. Res. 11, 1219–1229 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Zhao, J., Gu, S. & McDermaid, A. Predicting outcomes of chronic kidney disease from EMR data based on Random Forest Regression. Math. Biosci. 310, 24–30 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Work in the Susztak Lab is supported by the National Institutes of Health (NIH; P50DK114786, DK076077, DK087635, DK132630 and DK105821). The study is supported by GSK, Regeneron, Boehringer Ingelheim and Novo Nordisk. The funders have no influence on the reported results. The authors would like to thank the Molecular Pathology and Imaging Core (P30-DK050306) and Diabetes Research Center (P30-DK19525) at the University of Pennsylvania for their services. J.L. is supported by the ASN Ben Lipps Fellowship. B.D. is supported by the Deutsche Forschungsgemeinschaft (DFG; German Research Foundation, grant DU 2449/1-1). K.A.K. is supported by the MOLMED Ph.D. program at the Medical University of Graz, a Marietta-Blau Grant and an Austrian Marshall Plan Foundation scholarship. M.S.B. is supported by grants from the German Research Foundation (DFG, BA 6205/2-1), Else Kröner-Fresenius Foundation, Jackstädt Foundation and the Berlin Institute of Health at Charité – Universitätsmedizin Berlin Clinician Scientist Program.

Author information

Authors and Affiliations

Authors

Contributions

A.A., J.L., Z.M., J.F., R.S., P.D., D.T., A.M.B. and T.B. performed experiments. A.A., J.L., K.A.K., M.S.B., H.L., S.V., M.S.B., H.Y. and K.C. performed computational analysis. K.D., B.D., L.M., E.H., S.P., C.B.K., L.S.B., C.A.H., P.G., A.K., P.G., C.M.B., K.D.N, K.H.K. and M.L. offered experimental and analytical suggestions. K.S. was responsible for the overall design and oversight of the experiments. M.P. performed pathological scorings. K.S. supervised the experiment. A.A. and K.S. wrote the original draft. All authors contributed to and approved the final version of the manuscript.

Corresponding author

Correspondence to Katalin Susztak.

Ethics declarations

Competing interests

K.D. and L.M. are employees of Regeneron Pharmaceuticals. G.P., T.B., E.H. and L.S.B. are employees of GSK. S.P., C.M.B. and P.G. are employees of Boehringer Ingelheim. A.K. is an employee of Novo Nordisk. The remaining authors declare no competing interests.

Peer review

Peer review information

Nature Genetics thanks Rafel Kramann and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 Multimodal single-cell atlas.

(a) UMAP of snRNA-seq, scRNA-seq and snATAC-seq datasets before integration. (b) UMAP of integrated snRNA-seq, scRNA-seq and snATAC-seq datasets of 338,565 cells and nuclei using the SCVI tool. (c) Annotations of cell types on integrated UMAP. (d) The dot plots of marker genes used to annotate 44 main cell types in the integrated dataset. The size of the dot indicates the percent of positive cells, and the darkness of the color indicates average expression.

Extended Data Fig. 2 Integrations of snRNA-seq, scRNA-seq and snATAC-seq datasets from multiple sources.

(a) UMAP of integrated snRNA-seq, scRNA-seq and snATAC-seq datasets (n = 588,425 cells and nuclei) from Susztak Lab and KPMP using the SCVI tool. Left, the new annotation after integration of the dataset. Right, the original annotation used by KPMP. (b) Bar charts showing cell abundance in each cell cluster (Method, Lab, present study annotation and KPMP annotation).

Extended Data Fig. 3 CosMx cell populations.

(a) Original UMAP of all cells that passed QC along with their annotations. (b) Cell populations with annotatable markers of the CosMx data on UMAP. (c) UMAP of all CosMx cells by sample. (d) CosMx cell annotations across the UMAP with a single population being shown for a given UMAP demonstrating that these clusters are indeed relatively localized within the UMAP. (e) Dot plot showing markers for each annotated CosMx population. (f) Annotation frequency for each cell type.

Extended Data Fig. 4 CosMx SCVI integration with snRNA-seq data.

After integrating with the snRNA-seq data, we compared annotations from our CosMx analysis and the original snRNA-seq annotation. (a) UMAP of integrated data demonstrating the technology type of each cell within the UMAP. (b) Comparison of CosMx annotations and snRNA-seq annotations, demonstrating concordance of location within the integrated UMAP.

Extended Data Fig. 5 Location of CosMx annotated cell types within the slide.

(a) Location of annotated cell types within the two tissue sections. (b) Location of glomerular cell subtypes. (c) Location of iPT, fibroblasts and immune cells.

Extended Data Fig. 6 Microanatomy of the CosMx slide.

(a) Location of glomerular cell types within a subsection of tissue and in a single field of view (right). (b) Location of injured thick ascending limb, healthy injured thick ascending limb, principal cells and immune cell types within a subsection of tissue and in a single field of view (right). (c) Location of distal nephron cell subtypes within a subsection of tissue and in a single field of view (right). (d) Single field of view showing many cell types.

Extended Data Fig. 7 Neighborhood characteristics of CosMx slide.

(a) Relative type cell frequency between each sample. Orange indicates HK3039 (healthy), and blue indicates HK2844 (diseased). Right, frequency of neighbor annotations for each cell type for a 20-micron neighborhood. (b) Neighborhood enrichment by permuting annotations for the 20-micron neighborhood size. Lighter color indicates higher enrichment and colocalization of a given population. (c) Dot plots for iPT, PEC, podocytes and PT cells expression of iPT and PEC markers across genomics modalities and protein staining of VCAM1 in PECs from the Human Protein Atlas: https://www.proteinatlas.org/.

Extended Data Fig. 8 Cell–cell interaction analysis in the spRNA-seq dataset in fibrotic microenvironment.

(a) Weighted total interaction strength of the CXCL, SPP1, TGFβ and PDGF pathways in control and diseased samples in the fibrotic microenvironment (left). The spatial location of the identified cell–cell communications pathways (CXCL, SPP1, TGFβ and PDGF) in control and diseased sample in the fibrotic microenvironment (right). The arrows indicate the source and targets of the identified pathways. (b) Expression of CD34 and CDH5 as the markers of high endothelial venules (HEVs) in the fibrotic microenvironments. Scale bar is 1 mm in length. (c) Volcano plot of differentially expressed genes from CosMx data. Cells with an immune neighbor within 20 microns were compared against cells without an immune neighbor for both the PT and iPT population. log(fold change) >0 indicates increased expression in cells with an immune neighbor, while log(fold change) <0 indicates increased expression in cells without an immune neighbor. log10(p value) is indicated by the y axis. Genes with an adjusted p-value < 0.01 are marked in orange. (d) The dot plot of expression of ligands and receptors in regions of FME in integrated snRNA-seq/scRNA-seq and snATAC-seq data. The size of the dot indicates the percent positive cells and the darkness of the color indicates average expression (right). The gray indicates control, and the red indicates diseased group.

Extended Data Fig. 9 Spatial characteristics of injured PT subclusters on CosMx.

(a) The CosMx iPT population for each sample was subclustered using a Leiden algorithm with a resolution of 0.3. (b) The top 10 differentially expressed genes for each subcluster. (c) iPT subcluster localization within the entire slide. Views of specific regions indicated by inset boxes are shown in Supplementary Fig. 9d. (d) iPT subclusters visualized on H&E. Subset images showing populations on H&E. Blue cells correspond with cluster 0 (iPT_APOE), orange with cluster 1 (iPT_SPP1) and green with cluster 2 (iPT_KRT7). (e) Frequency of immune and fibroblast neighbors for each iPT subtype within each sample within 20 microns is shown below. We performed testing using a Wilcoxon rank-sum test between each population within a sample. These subtypes had significantly different immune neighbors and fibroblast neighbors with each sample. HK3039 fibroblasts: iPT_APOE vs iPT_SPP1, p-value = 7E-39. HK3039 immune cells: iPT_APOE vs iPT_SPP1, p-value = 4E-67. HK2844 fibroblasts iPT_APOE vs iPT_SPP1, p-value = 9E-9, iPT_KRT7 vs iPT_SPP1, p-value = 9E-46. HK2844 immune cells iPT_APOE vs iPT_SPP1, p-value = 3E-16, iPT_KRT7 vs iPT_SPP1, p-value = 9E-11. (f) iPT subcluster neighborhood enrichment within a 20-micron neighborhood size. Lighter color indicates higher enrichment and colocalization of a given population.

Extended Data Fig. 10 FME gene expression predicts kidney outcomes.

(a) Hierarchical clustering of 245 human kidney tubule samples based on the expression of 1,100 randomly picked genes. (b) Kaplan–Meier analysis with log-rank test was used to compare the survival of the 3 different clusters. Renal survival was defined as cases reaching end-stage renal disease or greater than 40% eGFR decline. (c) Single-cell expression enrichment of genes associated with eGFR decline. The heatmap shows the cell-type enrichment of genes associated with eGFR decline (red indicates more genes with cell-type expression). Endo, endothelial cells; Stroma, stromal cells; PEC, parietal epithelial cells; Podo, podocyte; PT, proximal tubule cells; LOH, loop of Henle; DCT, distal convoluted tubule; CNT, connecting tubule; PC, principal cells of collecting duct; IC_A, type alpha intercalated cells. Spatial expression and microenvironment enrichment of genes associated with eGFR decline. GME, glomerular; TME, tubule; FME, fibrosis; IME, immune microenvironment. (d) Using the LASSO regression of all FME genes against eGFR, Kaplan–Meier analysis was re-performed using clustering of subsets of gene—those with a LASSO coefficient that is nonzero and the genes with the most negative coefficients (Supplementary Table 13).

Supplementary information

Supplementary Information

Supplementary Note and Supplementary Figs. 1–27.

Reporting Summary

Supplementary Tables 1–26

Supplementary Table 1: Demographic and clinical characteristics of the participants in the study. Supplementary Table 2: QC metrics of the snRNA-seq, scRNA-seq, snATAC–seq and spRNA-seq data. Supplementary Table 3: Cell-type marker genes in the snRNA-seq data, 144 clusters. Supplementary Table 4: Cell-type marker genes in the snRNA-seq data, 44 clusters. Supplementary Table 5: Cell-type marker genes in the scRNA-seq data, 144 clusters. Supplementary Table 6: Cell-type marker genes in the scRNA-seq data, 44 clusters. Supplementary Table 7: Cell-type marker ATAC–seq, chromatin accessibility. Supplementary Table 8: Cell-type marker ATAC–seq, gene activity. Supplementary Table 9: Cell-type markers ATAC–seq, 144 clusters. Supplementary Table 10: Cell-type markers ATAC–seq, 44 clusters. Supplementary Table 11: Cell-type specific markers genes in the integrated snRNA-seq, snATAC–seq and scRNA-seq, 144 clusters. Supplementary Table 12: Cell-type-specific markers genes in the integrated snRNA-seq, snATAC–seq and scRNA-seq, 44 clusters. Supplementary Table 13: Cell-type-specific TF motif enrichment. Supplementary Table 14: Module-specific genes and scores in fibroblast (WGCNA). Supplementary Table 15: Module-specific genes and scores in myofibroblast (WGCNA). Supplementary Table 16: Differentially expressed genes between diseased and control samples in each cell type in the integrated snRNA-seq, snATAC–seq and scRNA-seq—DKD versus control. Supplementary Table 17: Differentially expressed genes between diseased and control samples in each cell type in the integrated snRNA-seq, snATAC–seq and scRNA-seq: all disease versus control. Supplementary Table 18: Differentially expressed genes between diseased and control samples in each cell type in the integrated snRNA-seq, snATAC–seq and scRNA-seq—CKD versus control. Supplementary Table 19: CellChat interaction scores, integrated. Supplementary Table 20: CellChat interaction scores, HK2671_ST. Supplementary Table 21: CellChat interaction scores, HK2844_ST. Supplementary Table 22: Differentially expressed genes along injured PT trajectory in the integrated snRNA-seq, snATAC–seq and scRNA-seq. Supplementary Table 23: Fibrotic microenvironment specific genes. Supplementary Table 24: Clinical characteristics of the 245 tubule bulk RNA-seq cohorts, summary. Supplementary Table 25: Clinical characteristics of the 245 tubule bulk RNA-seq cohorts, cluster analysis. Supplementary Table 26: LASSO score for each FME gene against eGFR.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Abedini, A., Levinsohn, J., Klötzer, K.A. et al. Single-cell multi-omic and spatial profiling of human kidneys implicates the fibrotic microenvironment in kidney disease progression. Nat Genet 56, 1712–1724 (2024). https://doi.org/10.1038/s41588-024-01802-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41588-024-01802-x

  • Springer Nature America, Inc.

This article is cited by

Navigation