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:

Single-cell atlas of human liver and blood immune cells across fatty liver disease stages reveals distinct signatures linked to liver dysfunction and fibrogenesis

Abstract

Immune cells play a central yet poorly understood role in metabolic dysfunction-associated steatotic liver disease and metabolic dysfunction-associated steatohepatitis (MASLD/MASH), a global cause of liver disease with limited treatment. Limited access to human livers and lack of studies across MASLD/MASH stages thwart identification of stage-specific immunological targets. Here we provide a unique single-cell RNA sequencing atlas of paired peripheral blood and liver fine-needle aspirates from a full-spectrum MASLD/MASH human cohort. Our findings included heightened immunoregulatory programs with MASH progression, such as enriched hepatic regulatory T cells, monocytic myeloid-derived suppressor cells, TREM2+S100A9+ macrophages and S100hiHLAlo type 2 conventional dendritic cells. Hepatic cytotoxic T cell functions increased with inflammation, but decreased with fibrosis, while acquiring an exhausted signature, whereas natural killer cell-driven toxicity intensified. Our dataset proposes immunological mechanisms for increased fibrogenesis and vulnerability to liver cancer and infections in MASH and provides a basis for a deeper understanding of human immunological dysfunction in chronic liver disease and a roadmap to new targeted therapies.

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

Access options

Buy this article

USD 39.95

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

Fig. 1: The landscape of liver and peripheral blood immune cells across the MASLD/MASH spectrum.
Fig. 2: Monocytes, monocyte-derived cells and their connectivity across the MASLD/MASH spectrum.
Fig. 3: Macrophage and pre-macrophage subsets and association with MASLD/MASH progression.
Fig. 4: DC subsets and association with MASLD/MASH progression.
Fig. 5: T cell subsets and association with MASLD/MASH progression.
Fig. 6: B cell subsets and association with MASLD/MASH progression.
Fig. 7: NK cell subsets and association with MASLD/MASH progression.
Fig. 8: Hepatic immune cell crosstalk and changes with MASLD/MASH progression.

Similar content being viewed by others

Data availability

The human RNA-seq data are deposited in the NIH dbGaP portal (accession no. phs004044) and can be used only for studying health, medical or biomedical conditions. Source data are provided with this paper.

References

  1. Younossi, Z. M. et al. The global epidemiology of nonalcoholic fatty liver disease (NAFLD) and nonalcoholic steatohepatitis (NASH): a systematic review. Hepatology 77, 1335–1347 (2023).

    Article  PubMed  Google Scholar 

  2. Taylor, R. S. et al. Association between fibrosis stage and outcomes of patients with nonalcoholic fatty liver disease: a systematic review and meta-analysis. Gastroenterology 158, 1611–1625.e12 (2020).

    Article  CAS  PubMed  Google Scholar 

  3. Lazarus, J. V. et al. Opportunities and challenges following approval of resmetirom for MASH liver disease. Nat. Med. 30, 3402–3405 (2024).

    Article  CAS  PubMed  Google Scholar 

  4. Huby, T. & Gautier, E. L. Immune cell-mediated features of non-alcoholic steatohepatitis. Nat. Rev. Immunol. 22, 429–443 (2022).

    Article  CAS  PubMed  Google Scholar 

  5. Guilliams, M. et al. Spatial proteogenomics reveals distinct and evolutionarily conserved hepatic macrophage niches. Cell 185, 379–396.e38 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Mueller, J. L. et al. Circulating soluble CD163 is associated with steatohepatitis and advanced fibrosis in nonalcoholic fatty liver disease. Clin. Transl. Gastroenterol. 6, e114 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Lidofsky, A. et al. Macrophage activation marker soluble CD163 is a dynamic marker of liver fibrogenesis in human immunodeficiency virus/hepatitis C virus coinfection. J. Infect. Dis. 218, 1394–1403 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  8. Cui, A. et al. Single-cell atlas of the liver myeloid compartment before and after cure of chronic viral hepatitis. J. Hepatol. 80, 251–267 (2024).

    Article  CAS  PubMed  Google Scholar 

  9. Racanelli, V. & Rehermann, B. The liver as an immunological organ. Hepatology 43, S54–S62 (2006).

    Article  CAS  PubMed  Google Scholar 

  10. Canton, M. et al. Reactive oxygen species in macrophages: sources and targets. Front. Immunol. 12, 734229 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Katze, M. G., He, Y. & Gale, M. Viruses and interferon: a fight for supremacy. Nat. Rev. Immunol. 2, 675–687 (2002).

    Article  CAS  PubMed  Google Scholar 

  12. Mohlenberg, M. et al. The role of IFN in the development of NAFLD and NASH. Cytokine 124, 154519 (2019).

    Article  PubMed  Google Scholar 

  13. Lou, Y. et al. Essential roles of S100A10 in Toll-like receptor signaling and immunity to infection. Cell. Mol. Immunol. 17, 1053–1062 (2020).

    Article  CAS  PubMed  Google Scholar 

  14. Ray, A., Chakraborty, K. & Ray, P. Immunosuppressive MDSCs induced by TLR signaling during infection and role in resolution of inflammation. Front. Cell. Infect. Microbiol. 3, 52 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  15. Mastio, J. et al. Identification of monocyte-like precursors of granulocytes in cancer as a mechanism for accumulation of PMN-MDSCs. J. Exp. Med. 216, 2150–2169 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Bekaert, M., Verhelst, X., Geerts, A., Lapauw, B. & Calders, P. Association of recently described adipokines with liver histology in biopsy-proven non-alcoholic fatty liver disease: a systematic review. Obes. Rev. 17, 68–80 (2016).

    Article  CAS  PubMed  Google Scholar 

  17. Krautbauer, S. et al. Chemerin is highly expressed in hepatocytes and is induced in non-alcoholic steatohepatitis liver. Exp. Mol. Pathol. 95, 199–205 (2013).

    Article  CAS  PubMed  Google Scholar 

  18. Street, K. et al. Slingshot: cell lineage and pseudotime inference for single-cell transcriptomics. BMC Genom. 19, 477 (2018).

    Article  Google Scholar 

  19. Kwak, T. et al. Distinct populations of immune-suppressive macrophages differentiate from monocytic myeloid-derived suppressor cells in cancer. Cell Rep. 33, 108571 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. La Manno, G. et al. RNA velocity of single cells. Nature 560, 494–498 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  21. Seidman, J. S. et al. Niche-specific reprogramming of epigenetic landscapes drives myeloid cell diversity in nonalcoholic steatohepatitis. Immunity 52, 1057–1074.e7 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Barbosa, C. M. V., Bincoletto, C., Barros, C. C., Ferreira, A. T. & Paredes-Gamero, E. J. PLCγ2 and PKC are important to myeloid lineage commitment triggered by M-SCF and G-CSF. J. Cell. Biochem. 115, 42–51 (2014).

    Article  CAS  PubMed  Google Scholar 

  23. Hollmén, M. et al. G-CSF regulates macrophage phenotype and associates with poor overall survival in human triple-negative breast cancer. Oncoimmunology 5, e1115177 (2016).

    Article  PubMed  Google Scholar 

  24. Ganguly, S. et al. Lipid-associated macrophages’ promotion of fibrosis resolution during MASH regression requires TREM2. Proc. Natl Acad. Sci. USA 121, e2405746121 (2024).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Xu, R. et al. Lipid-associated macrophages between aggravation and alleviation of metabolic diseases. Trends Endocrinol. Metab. 35, 981–995 (2024).

    Article  CAS  PubMed  Google Scholar 

  26. Wei, G. et al. Comparison of murine steatohepatitis models identifies a dietary intervention with robust fibrosis, ductular reaction, and rapid progression to cirrhosis and cancer. Am. J. Physiol. Gastrointest. Liver Physiol. 318, G174–G188 (2020).

    Article  PubMed  Google Scholar 

  27. Jahrsdörfer, B. et al. Granzyme B produced by human plasmacytoid dendritic cells suppresses T-cell expansion. Blood 115, 1156–1165 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  28. Heger, L. et al. XCR1 expression distinguishes human conventional dendritic cell type 1 with full effector functions from their immediate precursors. Proc. Natl Acad. Sci. USA 120, e2300343120 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Averill, M. M. et al. S100A9 differentially modifies phenotypic states of neutrophils, macrophages, and dendritic cells. Circulation 123, 1216–1226 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Plebanek, M. P. et al. A lactate-SREBP2 signaling axis drives tolerogenic dendritic cell maturation and promotes cancer progression. Sci. Immunol. 9, eadi4191 (2024).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Haas, J. T. et al. Transcriptional network analysis implicates altered hepatic immune function in NASH development and resolution. Nat. Metab. 1, 604–614 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Rau, M. et al. Progression from nonalcoholic fatty liver to nonalcoholic steatohepatitis is marked by a higher frequency of Th17 cells in the liver and an increased Th17/resting regulatory T cell ratio in peripheral blood and in the liver. J. Immunol. 196, 97–105 (2016).

    Article  CAS  PubMed  Google Scholar 

  33. Woestemeier, A. et al. Multicytokine-producing CD4+ T cells characterize the livers of patients with NASH. JCI Insight 8, e15381 (2023).

    Article  Google Scholar 

  34. Dudek, M. et al. Auto-aggressive CXCR6+ CD8 T cells cause liver immune pathology in NASH. Nature 592, 444–449 (2021).

    Article  CAS  PubMed  Google Scholar 

  35. Pfister, D. et al. NASH limits anti-tumour surveillance in immunotherapy-treated HCC. Nature 592, 450–456 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Yasumizu, Y. et al. Single-cell transcriptome landscape of circulating CD4+ T cell populations in autoimmune diseases. Cell Genom. 4, 100473 (2024).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Stewart, A. et al. Single-cell transcriptomic analyses define distinct peripheral B cell subsets and discrete development pathways. Front. Immunol. 12, 602539 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Hikida, M., Johmura, S., Hashimoto, A., Takezaki, M. & Kurosaki, T. Coupling between B cell receptor and phospholipase C-gamma2 is essential for mature B cell development. J. Exp. Med. 198, 581–589 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Svensson, A., Patzi Churqui, M., Schlüter, K., Lind, L. & Eriksson, K. Maturation-dependent expression of AIM2 in human B-cells. PLoS ONE 12, e0183268 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  40. Li, H., Borrego, F., Nagata, S. & Tolnay, M. Fc receptor-like 5 expression distinguishes two distinct subsets of human circulating tissue-like memory B cells. J. Immunol. 196, 4064–4074 (2016).

    Article  CAS  PubMed  Google Scholar 

  41. Andreani, V., Ramamoorthy, S., Fässler, R. & Grosschedl, R. Integrin β1 regulates marginal zone B cell differentiation and PI3K signaling. J. Exp. Med. 220, e20220342 (2023).

    Article  CAS  PubMed  Google Scholar 

  42. Maleki, I. et al. Serum immunoglobulin A concentration is a reliable biomarker for liver fibrosis in non-alcoholic fatty liver disease. World J. Gastroenterol. 20, 12566–12573 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. De Roza, M. A. et al. Immunoglobulin G in non-alcoholic steatohepatitis predicts clinical outcome: A prospective multi-centre cohort study. World J. Gastroenterol. 27, 7563–7571 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  44. Chen, Y. et al. The potential role of CMC1 as an immunometabolic checkpoint in T cell immunity. Oncoimmunology 13, 2344905 (2024).

    Article  PubMed  PubMed Central  Google Scholar 

  45. Castriconi, R. et al. Molecular mechanisms directing migration and retention of natural killer cells in human tissues. Front. Immunol. 9, 2324 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  46. Jin, S., Plikus, M. V. & Nie, Q. CellChat for systematic analysis of cell-cell communication from single-cell transcriptomics. Nat. Protoc. 20, 180–219 (2025).

    Article  CAS  PubMed  Google Scholar 

  47. Jiang, Y. & Lu, L. New insight into the agonism of protease-activated receptors as an immunotherapeutic strategy. J. Biol. Chem. 300, 105614 (2024).

    Article  CAS  PubMed  Google Scholar 

  48. Radaeva, S. et al. Natural killer cells ameliorate liver fibrosis by killing activated stellate cells in NKG2D-dependent and tumor necrosis factor-related apoptosis-inducing ligand-dependent manners. Gastroenterology 130, 435–452 (2006).

    Article  CAS  PubMed  Google Scholar 

  49. Seitz, T. et al. Fibroblast growth factor 9 is expressed by activated hepatic stellate cells and promotes progression of hepatocellular carcinoma. Sci. Rep. 10, 4546 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Zhou, L. et al. Integrated analysis highlights the immunosuppressive role of TREM2+ macrophages in hepatocellular carcinoma. Front. Immunol. 13, 848367 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Ahmad, J. N. et al. Bordetella adenylate cyclase toxin inhibits monocyte-to-macrophage transition and dedifferentiates human alveolar macrophages into monocyte-like cells. mBio 10, e01743-19 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  52. Wahlang, B., McClain, C., Barve, S. & Gobejishvili, L. Role of cAMP and phosphodiesterase signaling in liver health and disease. Cell. Signal. 49, 105–115 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Deczkowska, A. et al. XCR1+ type 1 conventional dendritic cells drive liver pathology in non-alcoholic steatohepatitis. Nat. Med. 27, 1043–1054 (2021).

    Article  CAS  PubMed  Google Scholar 

  54. Wimmers, F. et al. Single-cell analysis reveals that stochasticity and paracrine signaling control interferon-alpha production by plasmacytoid dendritic cells. Nat. Commun. 9, 3317 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank the Akoya scientist, A. Gad, for his valuable input with the Opal 3-plex tissue immunostaining used for validation. We thank Z. Li for helping organize the raw sequencing data for database deposition. This work was financially supported by Bristol Myers Squibb and grants from the National Institutes of Health (NIH) and the National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK) (grant nos. R01DK098079 to R.T.C. and R56DK134251 to N.A. and R.T.C.). The content of this work, however, is solely the responsibility of the authors and does not necessarily represent the official views of the sponsors.

Author information

Authors and Affiliations

Authors

Contributions

Conceptualization: R.T.C. and N.A. Methodology: C.O., M.S.W., H.B.P., O.P.M., L.P.W., P.H., U.K., Y.V.P., R.I.S. and N.A. Software and formal analysis: O.P.M., M.S.W. and C.O. Investigation: O.P.M., M.S.W., C.O., H.B.P., R.I.S., R.T.C. and N.A. Resources: M.D.B., J.E., C.C., S. Salloum, Z.R., A.O., S. Shroff, K.E.C. and E.D.C. Writing—original draft: M.S.W., O.P.M., C.O. and N.A. Writing—review and editing: O.P.M., M.S.W., E.D.C., R.T.C., R.I.S. and N.A. Funding acquisition: R.T.C. and N.A. Supervision: N.A.

Corresponding author

Correspondence to Nadia Alatrakchi.

Ethics declarations

Competing interests

E.D.C. was formerly employed by Bristol Myers Squibb. R.T.C. received research grants to the institution from Abbvie, Gilead Sciences, Merck, Boehringer, Janssen and BMS. N.A. received a research grant to the institution from Boehringer for unrelated work. The other authors declare no competing interests.

Peer review

Peer review information

Nature Immunology thanks Haydn Kissick and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Peer reviewer reports are available. Primary Handling Editor: Ioana Staicu, in collaboration with the Nature Immunology team.

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 Serum levels of monocyte and macrophage activation marker, soluble (s)CD163.

Levels of sCD163 significantly correlated with (a) non-invasive markers of fibrosis, liver stiffness (kPa) and APRI score; and (b) liver injury enzymes, ALT and AST, two-sided Spearman correlation. (c) Levels of sCD163 were significantly higher in patients with histologically defined MASH, compared to healthy controls (n = 10), and in advanced MASH (n = 9), compared to NS-SS (n = 6); and (d) in patients with high NAS score (>4, n = 11) compared to low NAS score (<5, n = 14) (the box plots represent the median and interquartile ranges, and the whiskers depict the minimum and maximum of the data set), two-sided Mann–Whitney unpaired U test. *P < .05, **P < .01, ***P < .001, ****P < .0001. Abbreviations: kPa, kilopascal; APRI, aspartate aminotransferase–to-platelet ratio index; ALT, Alanine aminotransferase; AST, Aspartate aminotransferase; NAS; MASLD (former NAFLD) activity score. NS, no steatosis/inflammation/fibrosis.

Extended Data Fig. 2 Additional information on the quality of scRNA-seq dataset.

(a) Ridge plot of the housekeeping gene B2M expression per sample, per compartment. (b) Stacked bar chart of sample proportion in each cell type, per compartment. (c) Separate clustering UMAPs for liver FNAs (n = 55,234) and PBMCs (n = 123,753), color-coded by cell type (clustering settings: 20 PCs, Louvain Resolution=0.05).

Extended Data Fig. 3 Broad characterization of cell type enrichment per compartment and per liver disease stage.

(a) Box plots showing per-sample (n = 25) frequencies of CD16 NK cells, MAIT cells, CD8+ T cells and B cells, and the ratio of CD4+/CD8+ T cells, per compartment: data are presented as median values (horizontal line), 25th/75th quartile values (bounds of boxes), and non-outlier minimum/maximum (whiskers); two-sided Wilcoxon. (b) Box plot of the proportion of each cell type as a percentage of each sample in FNA (left) and PBMC (right): data are presented as median values (horizontal line), 25th/75th quartile values (bounds of boxes), and non-outlier minimum/maximum (whiskers); NS-SS (n = 6), early MASH (n = 10), advanced MASH (n = 9); two-sided permutation test; significance (*) determined by false-discovery rate (FDR) < 0.05 and log2 false discovery (log2FD)>0.58. (c) Scatter plot of average pathway enrichment score per patient (n = 25) of (left) ROS/RNS production in macrophages by controlled attenuation parameter (CAP reflects steatosis), (middle) collagen formation in hepatic stellate cells (HSCs) by MASLD/NAFLD activity score (NAS), and (right) insulin-growth factor binding protein (IGF) and IGF-binding protein (IGFBP) in hepatocytes by liver enzyme ALT: error bands represent 95% confidence interval; two-sided Pearson.

Extended Data Fig. 4 Interferon-induced protein validation and liver sources of interferons.

Box plot by cell type per patient of average (a) ISG score per disease stage (NS-SS (n = 6), early MASH (n = 10), advanced MASH (n = 9)) in liver FNA (left) and PBMCs (right), (b) IFNG expression in liver FNAs (n = 25), and (c) upstream gene expression for interferon-alpha (left) and interferon-beta (right) in liver FNAs (n = 25): data are presented as median values (horizontal line), 25th/75th quartile values (bounds of boxes), and non-outlier minimum/maximum (whiskers); two-sided Wilcoxon; upstream genes are shown in Extended Data Table 2. (d) Magnification of Extended Data Fig. 3a: box plot of the percentage of pDCs out of each sample in FNA (left) and PBMC (right): data presented as in 5a; two-sided permutation test; significance (*) determined by false-discovery rate (FDR) < 0.05 and log2 false discovery (log2FD)>0.58. (e) Comparative ISG expression in liver monocytes (left) and macrophages (right) between patients without steatosis or with steatosis (NS-SS), those with MASH and those with untreated HCV: data are presented as median values (horizontal line); two-sided Wilcoxon; ***P < .001. (f) Immuno-fluorescent (IF) validation of MX1 and IFIT3 in macrophages in human liver tissue biopsies. Representative image merged and per channel (left) and expression fractions of MX1 + , IFIT3 + , and MX1 + IFIT3+ out of macrophages (CD68 + ) for three disease stages (right): simple steatosis (SS, n = 1), early MASH (F1, n = 1), and advanced MASH (F2, n = 1).

Extended Data Fig. 5 Additional data on monocyte association with MASLD/MASH progression.

(a) Monocyte chemotaxis receptors: box plots of average CCRL2 (left) and CMKLR1 (right) expression in FNA by patient: data are presented as median values (horizontal line), 25th/75th quartile values (bounds of boxes), and non-outlier minimum/maximum (whiskers); NS-SS (n = 6), early MASH (n = 10), advanced MASH (n = 9); two-sided Wilcoxon (b) Monocyte chemotaxis ligands: scatter plot of average RARRES2 expression in hepatocytes per patient (n = 25) by CAP score: error bands represent 95% confidence interval; two-sided Pearson.

Extended Data Fig. 6 Investigation of the monocyte and monocyte-derived cell paths without specifying a starting point of cell progression.

(a) Velocity analysis of monocyte and monocyte-derived cells (macrophages and cDCs) shown by UMAP in early MASH (left) and advanced MASH (right) and (b) unbiased pseudotime via slingshot analysis, color-coded per cell type.

Extended Data Fig. 7 Investigation of T-cell activation marker ICOS, its ligand, and B-cell light chain kappa versus lambda expression.

(a) Immunofluorescent staining of ICOS in human liver biopsies for three disease stages: simple steatosis (SS, steatosis grade 1, n = 1), early MASH (F1, steatosis grade 1, n = 1), and advanced MASH (F2, steatosis grade 2, n = 1). Box plots of (b) average ICOS expression by T-cell subpopulation per patient FNA (n = 25), (c) average Treg ICOS expression by histological steatosis grade per patient FNA (0-1: n = 10; 2: n = 11, 3 n = 3), two-sided Wilcoxon, and (d) average ICOSLG expression by cell type per patient FNA (n = 25): data are presented as median values (horizontal line), 25th/75th quartile values (bounds of boxes), and non-outlier minimum/maximum (whiskers). (e) Bar chart showing the fraction of B cells that express kappa (IGKC) or lambda (IGLC2-7) light chain genes for liver FNA (left) and PBMC (right).

Extended Data Fig. 8 Additional data on immune cell crosstalk.

Changes in predicted individual ligand-receptor interactions between (a) SS versus early MASH and (b) early versus advanced MASH, from HSCs to immune cells (left) and from immune cells to HSCs (right). (c) Radar plot of the total number of cell-cell interactions (both incoming and outgoing) between macrophages and other cell types (left) and between pDCs and other cell types (right).

Extended Data Table 1 Patient characteristics
Extended Data Table 2 Gene sets used for custom scoring

Supplementary information

Source data

Source Data Fig. 3

Composite IF images for SS, MASH F1 and MASH F2 in Fig. 3h.

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

Martin, O.P., Wallace, M.S., Oetheimer, C. et al. Single-cell atlas of human liver and blood immune cells across fatty liver disease stages reveals distinct signatures linked to liver dysfunction and fibrogenesis. Nat Immunol 26, 1596–1611 (2025). https://doi.org/10.1038/s41590-025-02255-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Version of record:

  • Issue date:

  • DOI: https://doi.org/10.1038/s41590-025-02255-y

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