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.

  • Article
  • Published:

XCR1+ type 1 conventional dendritic cells drive liver pathology in non-alcoholic steatohepatitis

A Publisher Correction to this article was published on 12 January 2022

This article has been updated

Abstract

Non-alcoholic fatty liver disease (NAFLD) and non-alcoholic steatohepatitis (NASH) are prevalent liver conditions that underlie the development of life-threatening cirrhosis, liver failure and liver cancer. Chronic necro-inflammation is a critical factor in development of NASH, yet the cellular and molecular mechanisms of immune dysregulation in this disease are poorly understood. Here, using single-cell transcriptomic analysis, we comprehensively profiled the immune composition of the mouse liver during NASH. We identified a significant pathology-associated increase in hepatic conventional dendritic cells (cDCs) and further defined their source as NASH-induced boost in cycling of cDC progenitors in the bone marrow. Analysis of blood and liver from patients on the NAFLD/NASH spectrum showed that type 1 cDCs (cDC1) were more abundant and activated in disease. Sequencing of physically interacting cDC-T cell pairs from liver-draining lymph nodes revealed that cDCs in NASH promote inflammatory T cell reprogramming, previously associated with NASH worsening. Finally, depletion of cDC1 in XCR1DTA mice or using anti-XCL1-blocking antibody attenuated liver pathology in NASH mouse models. Overall, our study provides a comprehensive characterization of cDC biology in NASH and identifies XCR1+ cDC1 as an important driver of liver pathology.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Fig. 1: scRNA-seq characterization of the liver immune niche during development of NASH.
Fig. 2: NASH-induced increase in conventional dendritic cell subtypes.
Fig. 3: cDCs in human NASH.
Fig. 4: NASH boosts DC-poiesis output in bone marrow.
Fig. 5: DCs in NASH induce more pro-inflammatory DC–T cell interaction signatures in liver lymph nodes.
Fig. 6: Depletion of Xcr1+ cDC1 protects the liver from NASH development and progression in diet-induced mouse models of NASH.

Similar content being viewed by others

Data availability

The scRNA-seq data generated in this study are available at the Gene Expression Omnibus under accession GSE169447.

Code availability

The Metacell package is available at https://github.com/tanaylab/metacell. The PIC-seq analysis package is available at https://github.com/aygoldberg/PIC-seq.

Change history

References

  1. Brunt, E. M. et al. Nonalcoholic fatty liver disease. Nat. Rev. Dis. Prim. 1, 15080 (2015).

    Article  PubMed  Google Scholar 

  2. Diehl, A. M. & Day, C. Cause, pathogenesis, and treatment of nonalcoholic steatohepatitis. N. Engl. J. Med. 377, 2063–2072 (2017).

    Article  CAS  PubMed  Google Scholar 

  3. Anstee, Q. M., Reeves, H. L., Kotsiliti, E., Govaere, O. & Heikenwälder, M. From NASH to HCC: current concepts and future challenges. Nat. Rev. Gastroenterol. Hepatol. 16, 411–428 (2019).

    Article  PubMed  Google Scholar 

  4. Friedman, S. L., Neuschwander-Tetri, B. A., Rinella, M. & Sanyal, A. J. Mechanisms of NAFLD development and therapeutic strategies. Nat. Med. 24, 908–922 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Chalasani, N. et al. The diagnosis and management of nonalcoholic fatty liver disease: practice guidance from the American Association for the Study of Liver Diseases. Hepatology 67, 328–357 (2018).

    Article  PubMed  Google Scholar 

  6. Macpherson, A. J., Heikenwälder, M. & Ganal-Vonarburg, S. C. The liver at the nexus of host-microbial interactions. Cell Host Microbe 20, 561–571 (2016).

    Article  CAS  PubMed  Google Scholar 

  7. Krenkel, O. & Tacke, F. Liver macrophages in tissue homeostasis and disease. Nat. Rev. Immunol. 17, 306–321 (2017).

    Article  CAS  PubMed  Google Scholar 

  8. Nishimura, S. et al. CD8+ effector T cells contribute to macrophage recruitment and adipose tissue inflammation in obesity. Nat. Med. 15, 914–920 (2009).

    Article  CAS  PubMed  Google Scholar 

  9. Bhattacharjee, J. et al. Hepatic natural killer T-cell and CD8+ T-cell signatures in mice with nonalcoholic steatohepatitis. Hepatol. Commun. 1, 299–310 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Wolf, M. J. et al. Metabolic activation of intrahepatic CD8+ T cells and NKT cells causes nonalcoholic steatohepatitis and liver cancer via cross-talk with hepatocytes. Cancer Cell 26, 549–564 (2014).

    Article  CAS  PubMed  Google Scholar 

  11. Breuer, D. A. et al. CD8+ T cells regulate liver injury in obesity-related nonalcoholic fatty liver disease. Am. J. Physiol. Gastrointest. liver Physiol. 318, g211–g224 (2020).

    Article  CAS  PubMed  Google Scholar 

  12. Pfister, D. et al. NASH limits anti-tumor surveillance in immunotherapy-treated HCC. Nature https://doi.org/10.1038/s41586-021-03362-0 (2021).

  13. Dudek, M. et al. Auto-aggressive CXCR6+ CD8 T cells cause liver immune pathology in NASH. Nature https://doi.org/10.1038/s41586-021-03233-8 (2021).

  14. Hart, K. M. et al. Type 2 immunity is protective in metabolic disease but exacerbates NAFLD collaboratively with TGF-β. Sci. Transl. Med. 9, eaal3694 (2017).

    Article  PubMed  Google Scholar 

  15. Jaitin, D. A. et al. Lipid-associated macrophages control metabolic homeostasis in a TREM2-dependent manner. Cell 178, 686–698 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Parhizkar, S. et al. Loss of TREM2 function increases amyloid seeding but reduces plaque-associated ApoE. Nat. Neurosci. 22, 191–204 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Xiong, X. et al. Landscape of intercellular crosstalk in healthy and NASH liver revealed by single-cell secretome gene analysis. Mol. Cell 75, 644–660 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Perugorria, M. J. et al. Non-parenchymal TREM-2 protects the liver from immune-mediated hepatocellular damage. Gut 68, 533 (2019).

    Article  CAS  PubMed  Google Scholar 

  19. Ramachandran, P. et al. Resolving the fibrotic niche of human liver cirrhosis at single-cell level. Nature https://doi.org/10.1038/s41586-019-1631-3 (2019).

  20. Montandon, S. A. et al. Multi-technique comparison of atherogenic and MCD NASH models highlights changes in sphingolipid metabolism. Sci. Rep. 9, 16810 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  21. Reid, D. T. & Eksteen, B. Murine models provide insight to the development of non-alcoholic fatty liver disease. Nutr. Res. Rev. 28, 133–142 (2015).

    Article  CAS  PubMed  Google Scholar 

  22. Baran, Y. et al. MetaCell: analysis of single-cell RNA-seq data using k-NN graph partitions. Genome Biol. 20, 206 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  23. Reid, D. T. et al. Kupffer cells undergo fundamental changes during the development of experimental NASH and are critical in initiating liver damage and inflammation. PLoS ONE 11, e0159524 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Tran, S. et al. Impaired Kupffer cell self-renewal alters the liver response to lipid overload during non-alcoholic steatohepatitis. Immunity 53, 627–640 (2020).

    Article  CAS  PubMed  Google Scholar 

  25. Devisscher, L. et al. Non-alcoholic steatohepatitis induces transient changes within the liver macrophage pool. Cell. Immunol. 322, 74–83 (2017).

    Article  CAS  PubMed  Google Scholar 

  26. Zang, S. et al. Neutrophils play a crucial role in the early stage of nonalcoholic steatohepatitis via neutrophil elastase in mice. Cell Biochem. Biophys. 73, 479–487 (2015).

    Article  CAS  PubMed  Google Scholar 

  27. Krenkel, O. et al. Myeloid cells in liver and bone marrow acquire a functionally distinct inflammatory phenotype during obesity-related steatohepatitis. Gut 69, 551–563 (2020).

    Article  CAS  PubMed  Google Scholar 

  28. Liaskou, E. et al. Monocyte subsets in human liver disease show distinct phenotypic and functional characteristics. Hepatology 57, 385–398 (2013).

    Article  CAS  PubMed  Google Scholar 

  29. Henning, J. R. et al. Dendritic cells limit fibroinflammatory injury in nonalcoholic steatohepatitis in mice. Hepatology 58, 589–602 (2013).

    Article  CAS  PubMed  Google Scholar 

  30. 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 

  31. Remmerie, A. et al. Osteopontin expression identifies a subset of recruited macrophages distinct from Kupffer cells in the fatty liver. Immunity 53, 641–657 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Daemen, S. et al. Dynamic shifts in the composition of resident and recruited macrophages influence tissue remodeling in NASH. Cell Rep. 34, 108626 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Guilliams, M. et al. Unsupervised high-dimensional analysis aligns dendritic cells across tissues and species. Immunity 45, 669–684 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Mederacke, I., Dapito, D. H., Affò, S., Uchinami, H. & Schwabe, R. F. High-yield and high-purity isolation of hepatic stellate cells from normal and fibrotic mouse livers. Nat. Protoc. 10, 305–315 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Trak-Smayra, V. et al. Pathology of the liver in obese and diabetic ob/ob and db/db mice fed a standard or high-calorie diet. Int J. Exp. Pathol. 92, 413–421 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Febbraio, M. A. et al. Preclinical models for studying NASH-driven HCC: how useful are they? Cell Metab. 29, 18–26 (2019).

    Article  CAS  PubMed  Google Scholar 

  37. Farrell, G. et al. Mouse models of nonalcoholic steatohepatitis: toward optimization of their relevance to human nonalcoholic steatohepatitis. Hepatology 69, 2241–2257 (2019).

    Article  PubMed  Google Scholar 

  38. Cabeza-Cabrerizo, M., Cardoso, A., Minutti, C. M., Pereira da Costa, M. & Reis e Sousa, C. Dendritic cells revisited. Ann. Rev. Immunol. https://doi.org/10.1146/annurev-immunol-061020-053707 (2021).

  39. Lavin, Y. et al. Tissue-resident macrophage enhancer landscapes are shaped by the local microenvironment. Cell 159, 1312–1326 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Malehmir, M. et al. Platelet GPIbα is a mediator and potential interventional target for NASH and subsequent liver cancer. Nat. Med. 25, 641–655 (2019).

    Article  CAS  PubMed  Google Scholar 

  41. Schlitzer, A., McGovern, N. & Ginhoux, F. in Seminars in Cell & Developmental Biology Vol. 41 9–22 (Elsevier, 2015).

  42. Schultze, J. L., Mass, E. & Schlitzer, A. Emerging principles in myelopoiesis at homeostasis and during infection and inflammation. Immunity 50, 288–301 (2019).

    Article  CAS  PubMed  Google Scholar 

  43. Schlitzer, A. et al. Identification of cDC1- and cDC2-committed DC progenitors reveals early lineage priming at the common DC progenitor stage in the bone marrow. Nat. Immunol. 16, 718–728 (2015).

    Article  CAS  PubMed  Google Scholar 

  44. Nutt, S. L. & Chopin, M. Transcriptional networks driving dendritic cell differentiation and function. Immunity 52, 942–956 (2020).

    Article  CAS  PubMed  Google Scholar 

  45. Yu, J. et al. The differential organogenesis and functionality of two liver-draining lymph nodes in mice. J. Autoimmun. 84, 109–121 (2017).

    Article  PubMed  Google Scholar 

  46. Giladi, A. et al. Dissecting cellular crosstalk by sequencing physically interacting cells. Nat. Biotechnol. 38, 629–637 (2020).

    Article  CAS  PubMed  Google Scholar 

  47. Böttcher, J. P. et al. NK cells stimulate recruitment of cDC1 into the tumor microenvironment promoting cancer immune control. Cell 172, 1022–1037 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  48. Wohn, C. et al. Absence of MHC class II on cDC1 dendritic cells triggers fatal autoimmunity to a cross-presented self-antigen. Sci. Immunol. 5, eaba1896 (2020).

    Article  CAS  PubMed  Google Scholar 

  49. Ghazarian, M. et al. Type I interferon responses drive intrahepatic T cells to promote metabolic syndrome. Sci. Immunol. 2, eaai7616 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  50. Jiao, J. et al. Dendritic cell regulation of carbon tetrachloride-induced murine liver fibrosis regression. Hepatology 55, 244–255 (2012).

    Article  PubMed  Google Scholar 

  51. Aarts, S. et al. Depletion of CD40 on CD11c+ cells worsens the metabolic syndrome and ameliorates hepatic inflammation during NASH. Sci. Rep. 9, 14702 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  52. Heier, E.-C. et al. Murine CD103+ dendritic cells protect against steatosis progression towards steatohepatitis. J. Hepatol. 66, 1241–1250 (2017).

    Article  CAS  PubMed  Google Scholar 

  53. Tussiwand, R. et al. Compensatory dendritic cell development mediated by BATF–IRF interactions. Nature 490, 502–507 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Seillet, C. et al. CD8α+ DCs can be induced in the absence of transcription factors Id2, Nfil3, and Batf3. Blood 121, 1574–1583 (2013).

    Article  CAS  PubMed  Google Scholar 

  55. Lee, W., Kim, H. S., Hwang, S. S. & Lee, G. R. The transcription factor Batf3 inhibits the differentiation of regulatory T cells in the periphery. Exp. Mol. Med. 49, e393–e393 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Li, J. et al. IL-9 and Th9 cells in health and diseases—from tolerance to immunopathology. Cytokine Growth Factor Rev. 37, 47–55 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Świderska, M. et al. The interplay between Th17 and T-regulatory responses as well as adipokines in the progression of non-alcoholic fatty liver disease. Clin. Exp. Hepatol. 3, 127–134 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  58. Zhang, Q. et al. Landscape and dynamics of single immune cells in hepatocellular carcinoma. Cell 179, 829–845 (2019).

    Article  CAS  PubMed  Google Scholar 

  59. Sun, Y. et al. Single-cell landscape of the ecosystem in early-relapse hepatocellular carcinoma. Cell 184, 404–421 (2021).

    Article  CAS  PubMed  Google Scholar 

  60. Zang, S. et al. Neutrophils play a crucial role in the early stage of nonalcoholic steatohepatitis via neutrophil elastase in mice. Cell Biochem. Biophys. 73, 479–487 (2015).

    Article  CAS  PubMed  Google Scholar 

  61. Tosello-Trampont, A. C. et al. NKp46(+) natural killer cells attenuate metabolism-induced hepatic fibrosis by regulating macrophage activation in mice. Hepatology 63, 799–812 (2016).

    Article  CAS  PubMed  Google Scholar 

  62. Finkin, S. et al. Ectopic lymphoid structures function as microniches for tumor progenitor cells in hepatocellular carcinoma. Nat. Immunol. 16, 1235–1244 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Halpern, K. B. et al. Single-cell spatial reconstruction reveals global division of labour in the mammalian liver. Nature 542, 352–356 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Keren-Shaul, H. et al. MARS-seq2.0: an experimental and analytical pipeline for indexed sorting combined with single-cell RNA sequencing. Nat. Protoc. 14, 1841–1862 (2019).

    Article  CAS  PubMed  Google Scholar 

  65. Soucie, E. L. et al. Lineage-specific enhancers activate self-renewal genes in macrophages and embryonic stem cells. Science 351, aad5510 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  66. Kleiner, D. E. et al. Design and validation of a histological scoring system for nonalcoholic fatty liver disease. Hepatology 41, 1313–1321 (2005).

    Article  PubMed  Google Scholar 

  67. Giladi, A. et al. Single-cell characterization of haematopoietic progenitors and their trajectories in homeostasis and perturbed haematopoiesis. Nat. Cell Biol. 20, 836–846 (2018).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank T. Wiesel and T. Bigdary from the Scientific Illustration unit of the Weizmann Institute for artwork, C. Raanan for histology, M. Guilliams, A. Schlitzer and members of the Amit laboratory for fruitful discussions. We thank A. Leshem, H. Keren-Shaul, F. Sheban, L. Geirsdottir, E. Tatirovsky, F. Müller, J. Hetzer and D. Heide for technical support. A.D. is a recipient of Short-Term EMBO Fellowship no. 7395 and is supported by Eden and Steven Romick. D.P. is supported by the Helmholtz Future; Inflammation and Immunology. M.H.W. is supported by an ERC Consolidator grant (HepatoMetaboPath), an EOS grant, SFBTR179, SFBTR 209, SFBTR1335, Horizon 2020, Research Foundation Flanders under grant 30826052 (EOS Convention MODEL-IDI), Deutsche Krebshilfe projects 70113166 and 70113167, German-Israeli Cooperation in Cancer Research (DKFZ-MOST) and the Helmholtz-Gemeinschaft, Zukunftsthema ‘Immunology and Inflammation’ (ZT-0027). E.E. is supported by the Leona M. and Harry B. Helmsley Charitable Trust; Adelis Foundation; Pearl Welinsky Merlo Scientific Progress Research Fund; Park Avenue Charitable Fund; The Hanna and Dr. Ludwik Wallach Cancer Research Fund; Daniel Morris Trust; The Wolfson Family Charitable Trust and The Wolfson Foundation; Ben B. and Joyce E. Eisenberg Foundation; White Rose International Foundation; Estate of Malka Moskowitz; Estate of Myron H. Ackerman; Estate of Bernard Bishin for the WIS-Clalit Program; Else Kroener Fresenius Foundation; Jeanne and Joseph Nissim Center for Life Sciences Research; Aliza Moussaieff; Miel de Botton; Vainboim Family; Alex Davidoff; the V. R. Schwartz Research Fellow Chair; the Swiss Society Institute for Cancer Prevention Research at the Weizmann Institute of Science, Rehovot, Israel and by grants funded by the European Research Council; Israel Science Foundation; Israel Ministry of Science and Technology; Israel Ministry of Health; the Helmholtz Foundation; Garvan Institute; European Crohn’s and Colitis Organization; Deutsch-Israelische Projektkooperation; IDSA Foundation; and Welcome Trust. E.E. is the incumbent of the Sir Marc and Lady Tania Feldmann Professorial Chair; a senior fellow, Canadian Institute of Advanced Research; and an international scholar, The Bill and Melinda Gates Foundation and Howard Hughes Medical Institute. I.A. is an Eden and Steven Romick Professorial Chair, supported by Merck KGaA, Darmstadt, Germany, the Chan Zuckerberg Initiative, the Howard Hughes Medical Institute International Scholar award, the European Research Council Consolidator Grant 724471-HemTree2.0, a Single Cell Analysis (SCA) award of the Wolfson Foundation and Family Charitable Trust, the Thompson Family Foundation, a Melanoma Research Alliance Established Investigator Award (509044), the Israel Science Foundation (703/15), the Ernest and Bonnie Beutler Research Program for Excellence in Genomic Medicine, the Helen and Martin Kimmel award for innovative investigation, the NeuroMac DFG/Transregional Collaborative Research Center Grant, an International Progressive MS Alliance/NMSS PA-1604 08459, the ISF Israel Precision Medicine Program (IPMP) 607/20 grant and an Adelis Foundation grant.

Author information

Authors and Affiliations

Authors

Contributions

A.D. and I.A. designed, analyzed and interpreted all experiments. A.D. wrote the manuscript. A.D., B.L., D.A.J., I.Y., M.C., O.B., S.S.-L. and C.G. performed experiments and analyzed the data. D.P., P.R. and K.S. under supervision of M.H., and Y.S. and M.Q. under the supervision of S.R. performed experiments and analyzed the data. A. Weiner and E.D. analyzed scRNA-seq data. A.G. analyzed PIC-seq data. C.G., D.G., H.H., E.L., G.B.-Y., O.C.-E., Y.D. and M.L. recruited the patients for the study. M.S. and Z.B.-A. prepared human blood for analysis and diagnosed liver pathology. A. Weber diagnosed liver pathology. S.H. and B.M. contributed resources. A.D., E.D. and A. Weiner prepared the figures. M.H., E.E. and I.A. supervised the study.

Corresponding authors

Correspondence to Aleksandra Deczkowska, Mathias Heikenwälder, Eran Elinav or Ido Amit.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Nature Medicine thanks Frank Tacke, Matteo Iannacone, Scott Friedman and the other, anonymous reviewer(s) for their contribution to the peer review of this work. Joao Monteiro was the primary editor on this article and managed its editorial process and peer review in collaboration with the rest of the editorial team.

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

Extended data

Extended Data Fig. 1 scRNA-seq characterization of the liver immune niche during development of NASH.

a, Representative pictures of H&E staining of livers in wild type C57/bl mice fed with ND or MCDD for 2, 4 and 8 weeks. b, Flow cytometry plots illustrating gating strategy for CD45 + immune cell selection for scRNA-seq analysis. c, Shown are number of reads, number of UMIs and percentage of cells analyzed per batch of 380 cells (that were pooled for library construction) for all cells collected for scRNA-seq in experiments presented in Figs. 1, 2 and 4. Black dots indicate cells used for downstream analysis and grey dots indicate cells which were filtered out.

Extended Data Fig. 2 Immune cell composition of the liver in NASH development.

a, Heat map of differential gene expression for all cells colored by the assigned metacells as in Fig. 1. Top bar graph shows relative contribution of ND and MCDD liver immune cells to each metacell. b, Density plots depicting distribution of cells in ND and MCDD condition (downsampled to 6699 cells) and enrichment analysis of MCDD as compared to ND (all time-points analyzed together) projected on the kNN graph. Red areas represent enrichment, blue areas represent depletion. c, kNN graph of liver immune cells of wild-type mice kept on ND and MCDD, down-sampled to 1000 cells at each time-point analyzed. d, relative abundance of immune cell subtypes in livers of ND or MCDD-fed wild type mice at each analyzed time-point.

Extended Data Fig. 3 Hepatic cDC increase in NASH.

a, Flow cytometry plots illustrating gating strategy for cDCs, cDC1, cDC2 and mDC quantification and sorting. b, Heat map of differential gene expression of cDC sorted from Xcr1Cre-mTFP1 reporter mice. Top plots illustrate relative fluorescent intensity of indicated FACS markers in each cell, ordered as in the heat map. c, Representative flow cytometry contour-plots of CD11c + MHC-II + cells out of CD45 + Lin- gate in livers of mice fed ND or MCDD for two weeks. Population frequencies represent mean ± s.e.m. d, Representative immunofluorescence images of CD11c (red), MHC-II (green) and CD31 (vasculature marker; magenta) in liver section of mice fed ND or MCDD for two weeks. Cell nuclei are stained with DAPI (blue). Scale bar, 30 μm. Images representative for two independent experiments. e, Representative pictures of H&E staining of livers in wild type C57/bl mice fed with ND or WD for 3 and 6 months. f, Percentage of CD11c + MHC-II + DCs among CD45 + cells in the livers of mice fed WD for 3 months, n = 6 per group, data are presented as mean ± s.e.m.; p-value was determined by two-tailed Student’s t-test. g, Representative pictures of H&E staining of livers in wild type C57/bl mice fed with ND or CDHFD for 6 months.

Extended Data Fig. 4 Proportions of hepatic cDC subtypes change in murine NASH and NAFLD models.

a, Representative pictures of H&E staining of livers in wild type C57/bl mice fed with ND or HFD for 6 months, 9 week-old Ob/Ob mouse and Ob/het control, 12 week-old Db/Db mouse and Db/het control. b, Percentage of DCs among CD45 + cells in the livers of wild type mice fed ND or HFD for 6 months, 9 week-old Ob/Ob mouse and Ob/het control, 12 week-old Db/Db mouse and Db/het control. c, Representative flow cytometry contour-plots of cDC1 and cDC2 out of cDC gate in livers of mice fed ND or MCDD for two weeks. Population frequencies represent mean ± s.e.m. d, Representative immunofluorescence image of XCR1 (red), CD11c (magenta), MHC-II (green) and DAPI (nuclear stain, blue) in liver section of mouse fed MCDD for two weeks. Scale bar, 30 μm. Images representative for two independent experiments. e, Representative immunofluorescence image of CD103 (white), CD11c (red), MHC-II (green) and DAPI (nuclear stain, blue) in liver section of mouse fed CDHFD for six months. Scale bar, 50 μm. Images representative for two independent experiments. f, Representative flow cytometry contour-plots of CCR7 + mDCs out of cDC gate in livers of mice fed ND or MCDD for two weeks. Population frequencies represent mean ± s.e.m. g-h, Percentage of cDC1 (g) and cDC2 (h) among CD45 + cells in the livers of wild type mice fed ND or HFD for 6 months, 9 week-old Ob/Ob mouse and Ob/het control, 12 week-old Db/Db mouse and Db/het control. In all graphs points indicate individual mice, n = 3-4 per group. Data are presented as mean ± s.e.m.; p-values were determined by two-tailed Student’s t-test; p < 0.001.

Extended Data Fig. 5 cDC in human NASH.

a, Full gating strategy used to select cDC from human livers. b-c, Number of UMIs (b) and percentage of QC-positive single cells (c) used for analysis out of all cells detected in scRNA-seq pipeline. d, Heat map of differential gene expression among CD45 + Lin-CD11C + cells sorted from healthy and NASH human livers. e, Relative abundance of cell subtypes among CD45 + Lin-CD11C + cells sorted from healthy and NASH human livers. f, Full gating strategy used to select cDC, cDC1 and cDC2 from human blood.

Extended Data Fig. 6 NASH boosts DC-poiesis.

a, Flow cytometry plots illustrating gating strategy for assessing BrdU incorporation in MDP, CDPs and preDCs in the bone marrow, blood and liver. b, Percentage of BrdU+ cells among MDP in the bone marrow of mice fed ND or MCDD for two weeks and pulsed with BrdU 10 h prior to DC progenitor analysis. n = 3 per group, data are presented as mean ± s.e.m. c, kNN graph of bone marrow-isolated CDP, pre-DC isolated form the bone marrow, blood and liver and liver DC isolated from wild-type mice kept on ND or fed MCDD for two weeks. d, Plot representing expression of marker genes in annotated clusters in cells showed in c. Color intensity indicates log2 of the mean UMI count; circle size represents percentage of cells within cluster expressing indicated genes. e, kNN graphs showing distribution of single cells from different organs and diet-regiments matched with bar plots showing relative contribution of each annotated cell type among CDP sorted from the bone marrow and pre-DCs sorted from the bone marrow, blood and liver of wild-type mice kept on ND or on MCDD for two weeks. Plots were down-sampled to 680 cells for CDP, 1404 for bone marrow preDC, 601 for blood preDC and 622 for liver preDC.

Extended Data Fig. 7 Characterization of genetic signatures of DC-T pairs in liver lymph nodes in NASH.

a, Full gating strategy for T and DC, as well as PIC-sorting from murine liver lymph nodes. b, Number of reads, number of UMIs and percentage of cells analyzed per batch of 380 cells (that were pooled for library construction) in PIC-seq experiments. c, Heat map of differential gene expression for all single cells sorted as DC and T cells, annotated based on their gene expression, and used to create ‘expected PICs’ for PIC-seq analysis. Highlighted are marker genes for the annotated DC and T subtypes. d,e, Gene-expression levels in observed PICs assigned to the cDC1-T identity (d) or cDC2-T identity (e), plotted against their expected levels as determined by PICseq in the liver lymph nodes of mice kept on ND or MCDD for two weeks. Highlighted genes are expressed differentially between observed and expected PICs in one or both conditions.

Extended Data Fig. 8 Partial depletion of hepatic cDC1 infiltration using anti-XCL1 antibody in CDHFD-fed mice attenuates NASH progression.

a, Levels of liver enzymes ALT and AST in the serum of wild type CDHFD-fed mice before anti-XCL1 or IgG isotype control treatment. b, Quantification of CD103 + MHC-II + CD11c + cells from 5–10 randomly selected areas per mouse c, Levels of AST, a measure of tissue injury, including liver, in the serum of wild type mice before feeding with CDHFD (grey), after 5 months of CDHFD but before treatment (black), and following 4 weeks of anti-XCL1 or control IgG treatment (blue). In a-c n = 6–7 per group. Data are presented as mean ± s.e.m. P-values were determined by two-way ANOVA with Tukey’s multiple comparisons test; p < 0.001, p < 0.0001 d,e, Representative pictures of H&E staining (d) and pathological evaluation of NAFLD activity score (e) in anti-XCL1 or IgG isotype control-treated, CDHFD-fed mice. Images representative for two experiments. f,g, Representative pictures (f) and quantification (g) of Sirius Red staining in anti-XCL1 or IgG isotype control-treated, CDHFD-fed mice. Images representative for two independent experiments. In e, g, data are presented as mean ± s.e.m.; n = 7 per group, two-tailed Student’s t-test h. Correlation analysis between MHC-II+CD11c+CD103+ cDC1 count and NASH pathology parameters in anti-XCL1-treated and IgG-treated mice. n = 14 mice, two-tailed Spearman correlation (crossed out points p > 0.05; color and circle size indicate R2).

Extended Data Fig. 9 Depletion of cDC1 in Xcr1DTA mice prevents NASH-induced immune infiltration.

a, Flow cytometry plots illustrating specific depletion of cDC1 in Xcr1DTA mice, compared to control mice kept both on ND and after two week of MCDD feeding. b, Representative pictures of H&E staining control mice and Xcr1DTA mice (lacking Xcr1 + cDC1) fed with ND or MCDD for two weeks. Images representative for two independent experiments. c,d, FACS gating strategy and quantification of CD45 + immune cell percentage among all cells isolated from livers of control and Xcr1DTA mice kept on ND or MCDD for two weeks. n = 4 per group. Data are presented as mean ± s.e.m.; p < 0.0001 by One-way ANOVA with Tukey’s multiple comparisons test.

Extended Data Fig. 10 cDC1 modulate hepatic immune cell populations in the MCDD model of NASH.

a,b, Representative flow cytometry plots (a) and flow cytometry-based quantification (b) of CD8 + cells in the livers of control and Xcr1DTA mice kept on ND or MCDD for two weeks. n = 4 per group. c,d, Representative microscopic images of IHC staining for CD8 (c), and image-based quantification (d) of the stained CD8 + cells in the livers of control and Xcr1DTA mice kept on ND or MCDD for two weeks. bar=100 μm. For quantification 5–10 randomly selected fields per mouse were averaged, n = 5 mice in control and Xcr1DTA-ND groups, n = 4 in the Xcr1DTA-MCDD group. e, FACS-based quantification of CD44 + CD62L + T central memory CD8 + T cells and naïve CD44-CD8 + T cells, separated as shown in (a) in the livers of control and Xcr1DTA mice kept on ND or MCDD for two weeks. n = 4 individual mice per group f, Flow cytometry plots representing gating strategy used to dissect CD4 + T cell subpopulations. g, Flow cytometry-based quantification of CD4 + T cell percentage among CD45 + immune cells in the livers of control and Xcr1DTA mice kept on ND or MCDD for two weeks. n = 4 mice per group h,i, Representative microscopic images of IHC staining for CD4 (h) and image based quantification (i) of CD4 + cells in the livers of control and Xcr1DTA mice kept on ND or MCDD for two weeks. bar = 100 μm. For quantification 5–10 randomly selected fields per mouse were averaged, n = 5 mice per group. j, Flow cytometry-based quantification of CD4 + T cell subsets in the livers of control and Xcr1DTA mice kept on ND or MCDD for two weeks. n = 4 mice per group k,l, Flow cytometry contour plots and flow cytometry-based quantification of NKT cells (k) and monocytes (l) in the livers of control and Xcr1DTA mice kept on ND or MCDD for two weeks. n = 4 mice per group m, Representative microscopic images of IHC staining for CD42b, a marker of platelets, and image based quantification of the stained cells in the livers of control and Xcr1DTA mice kept on ND or MCDD for two weeks. bar = 100 μm. For quantification 5–10 randomly selected fields per mouse were averaged, n = 5 mice per group. In all graphs data are presented as mean ± s.e.m. P-values were determined by one-way ANOVA with Tukey’s multiple comparisons test; p < 0.001, p < 0.0001.

Supplementary information

Reporting Summary

Supplementary Table 1

Gene lists used to perform PIC-seq.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Deczkowska, A., David, E., Ramadori, P. et al. XCR1+ type 1 conventional dendritic cells drive liver pathology in non-alcoholic steatohepatitis. Nat Med 27, 1043–1054 (2021). https://doi.org/10.1038/s41591-021-01344-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41591-021-01344-3

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