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:

A spatiotemporal atlas of cholestatic injury and repair in mice

Abstract

Cholestatic liver injuries, characterized by regional damage around the bile ductular region, lack curative therapies and cause considerable mortality. Here we generated a high-definition spatiotemporal atlas of gene expression during cholestatic injury and repair in mice by integrating spatial enhanced resolution omics sequencing and single-cell transcriptomics. Spatiotemporal analyses revealed a key role of cholangiocyte-driven signaling correlating with the periportal damage-repair response. Cholangiocytes express genes related to recruitment and differentiation of lipid-associated macrophages, which generate feedback signals enhancing ductular reaction. Moreover, cholangiocytes express high TGFβ in association with the conversion of liver progenitor-like cells into cholangiocytes during injury and the dampened proliferation of periportal hepatocytes during recovery. Notably, Atoh8 restricts hepatocyte proliferation during 3,5-diethoxycarbonyl-1,4-dihydro-collidin damage and is quickly downregulated after injury withdrawal, allowing hepatocytes to respond to growth signals. Our findings lay a keystone for in-depth studies of cellular dynamics and molecular mechanisms of cholestatic injuries, which may further develop into therapies for cholangiopathies.

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: Spatial and single-cell transcriptomic atlas of cholestatic liver injury and repair.
Fig. 2: Stereo-seq dissects the regional dynamics of tissue damage.
Fig. 3: Integrated transcriptomics identifies cholangiocyte-driven signaling possesses a hub-like function to coordinate the periportal damage responses.
Fig. 4: Identification of the spatial heterogeneity of LPLCs.
Fig. 5: Spatiotemporal dynamic of damage regression and cholangiocyte diminishment during liver repair.
Fig. 6: Spatiotemporal dynamic of regeneration regulating factors and regulons during liver regeneration.
Fig. 7: Atlas of cholestatic liver injury and repair.

Similar content being viewed by others

Data availability

All raw data generated by Stereo-seq and scRNA-seq have been deposited to CNGB Nucleotide Sequence Archive with accession number: CNP0003447 (ref. 71). Additional data, including processed spatially visually genes in Stereo-seq and gene expression of annotated cell types in scRNA-seq, can be accessed from ref. 19. All sequencing reads from Stereo-seq and scRNA-seq were mapped to the mouse reference genome (mm10). Raw and processed bulk RNA-seq data from this study are also available through the Gene Expression Omnibus under accession number GSE219169. Source data are provided with this paper.

Code availability

The upstream analyses are carried out using published and freely available software and code, which is indexed at https://github.com/MGI-tech-bioinformatics/DNBelab_C_Series_HT_scRNA-analysis-software for scRNA-seq analysis workflow and ref. 68 for Stereo-seq analysis workflow. The code used for reproducing downstream analyses and visualization in the main figure is indexed at https://github.com/XinyiST/CIRSTA_analysis (https://doi.org/10.5281/zenodo.10577701).

References

  1. Cunningham, R. P. & Porat-Shliom, N. Liver zonation—revisiting old questions with new technologies. Front. Physiol. 12, 732929 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  2. Sato, K. et al. Ductular reaction in liver diseases: pathological mechanisms and translational significances. Hepatology 69, 420–430 (2019).

    Article  PubMed  Google Scholar 

  3. Banales, J. M. et al. Cholangiocyte pathobiology. Nat. Rev. Gastroenterol. Hepatol. 16, 269–281 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  4. Mariotti, V., Strazzabosco, M., Fabris, L. & Calvisi, D. F. Animal models of biliary injury and altered bile acid metabolism. Biochim. Biophys. Acta Mol. Basis Dis. 1864, 1254–1261 (2018).

    Article  CAS  PubMed  Google Scholar 

  5. Planas-Paz, L. et al. YAP, but not RSPO-LGR4/5, signaling in biliary epithelial cells promotes a ductular reaction in response to liver injury. Cell Stem Cell 25, 39–53 e10 (2019).

    Article  CAS  PubMed  Google Scholar 

  6. Jörs, S. et al. Lineage fate of ductular reactions in liver injury and carcinogenesis. J. Clin. Invest. 125, 2445–2457 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  7. Li, W. et al. A homeostatic Arid1a-dependent permissive chromatin state licenses hepatocyte responsiveness to liver-injury-associated YAP signaling. Cell Stem Cell 25, 54–68 e5 (2019).

    Article  CAS  PubMed  Google Scholar 

  8. Tarlow, B. D. et al. Bipotential adult liver progenitors are derived from chronically injured mature hepatocytes. Cell Stem Cell 15, 605–618 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Gao, C. & Peng, J. All routes lead to Rome: multifaceted origin of hepatocytes during liver regeneration. Cell Regen. 10, 2 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. He, J., Deng, C., Krall, L. & Shan, Z. scRNA-seq and ST-seq in liver research. Cell Regen. 12, 11 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Chen, A. et al. Spatiotemporal transcriptomic atlas of mouse organogenesis using DNA nanoball-patterned arrays. Cell 185, 1777–1792.e21 (2022).

    Article  CAS  PubMed  Google Scholar 

  12. Karras, P. et al. A cellular hierarchy in melanoma uncouples growth and metastasis. Nature 610, 190–198 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. 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 

  14. Li, L. et al. Kupffer-cell-derived IL-6 is repurposed for hepatocyte dedifferentiation via activating progenitor genes from injury-specific enhancers. Cell Stem Cell 30, 283–299 e9 (2023).

    Article  CAS  PubMed  Google Scholar 

  15. 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 

  16. Cable, D. M. et al. Robust decomposition of cell type mixtures in spatial transcriptomics. Nat. Biotechnol. 40, 517–526 (2022).

    Article  CAS  PubMed  Google Scholar 

  17. Dobie, R. et al. Single-cell transcriptomics uncovers zonation of function in the mesenchyme during liver fibrosis. Cell Rep. 29, 1832–1847.e8 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Tanaka, M. & Iwakiri, Y. The hepatic lymphatic vascular system: structure, function, markers, and lymphangiogenesis. Cell Mol. Gastroenterol. Hepatol. 2, 733–749 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  19. CIRSTA: Cholestatic Injury and Repair Spatio-Temporal Atlas. STOmicsDB https://db.cngb.org/stomics/cirsta (2024).

  20. Andrade, R. J. et al. Drug-induced liver injury. Nat. Rev. Dis. Primers 5, 58 (2019).

    Article  PubMed  Google Scholar 

  21. Aravinthan, A. D. & Alexander, G. J. M. Senescence in chronic liver disease: is the future in aging? J. Hepatol. 65, 825–834 (2016).

    Article  CAS  PubMed  Google Scholar 

  22. Ma, Q. Role of nrf2 in oxidative stress and toxicity. Annu. Rev. Pharmacol. Toxicol. 53, 401–426 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Gong, T., Liu, L., Jiang, W. & Zhou, R. DAMP-sensing receptors in sterile inflammation and inflammatory diseases. Nat. Rev. Immunol. 20, 95–112 (2020).

    Article  CAS  PubMed  Google Scholar 

  24. Schaefer, L. Complexity of danger: the diverse nature of damage-associated molecular patterns. J. Biol. Chem. 289, 35237–35245 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Mederacke, I. The purinergic P2Y14 receptor links hepatocyte death to hepatic stellate cell activation and fibrogenesis in the liver. Sci. Transl. Med. 14, eabe5795 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Shetty, S., Lalor, P. F. & Adams, D. H. Liver sinusoidal endothelial cells—gatekeepers of hepatic immunity. Nat. Rev. Gastroenterol. Hepatol. 15, 555–567 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Lukacs-Kornek, V. The role of lymphatic endothelial cells in liver injury and tumor development. Front. Immunol. 7, 5848 (2016).

    Article  Google Scholar 

  28. Tamburini, B. A. J. et al. Chronic liver disease in humans causes expansion and differentiation of liver lymphatic endothelial cells. Front. Immunol. 10, 1036 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  29. Ye, C. et al. Single‐cell and spatial transcriptomics reveal the fibrosis‐related immune landscape of biliary atresia. Clin. Transl. Med. 12, e1070 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Casazza, A. et al. Impeding macrophage entry into hypoxic tumor areas by Sema3A/Nrp1 signaling blockade inhibits angiogenesis and restores antitumor immunity. Cancer Cell 24, 695–709 (2013).

    Article  CAS  PubMed  Google Scholar 

  31. Takase, H. M. et al. FGF7 is a functional niche signal required for stimulation of adult liver progenitor cells that support liver regeneration. Genes Dev. 27, 169–181 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Bird, T. G. et al. Bone marrow injection stimulates hepatic ductular reactions in the absence of injury via macrophage-mediated TWEAK signaling. Proc. Natl Acad. Sci. USA 110, 6542–6547 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Alvaro, D. et al. The intrahepatic biliary epithelium is a target of the growth hormone/insulin-like growth factor 1 axis. J. Hepatol. 42, 33 (2005).

    Article  Google Scholar 

  34. Cao, S., Liu, M., Sehrawat, T. S. & Shah, V. H. Regulation and functional roles of chemokines in liver diseases. Nat. Rev. Gastroenterol. Hepatol. 18, 630–647 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  35. Akagawa, K. S. et al. Functional heterogeneity of colony-stimulating factor-induced human monocyte-derived macrophages. Respirology 11, S32–S36 (2006).

    Article  PubMed  Google Scholar 

  36. Stutchfield, B. M. et al. CSF1 restores innate immunity after liver injury in mice and serum levels indicate outcomes of patients with acute liver failure. Gastroenterology 149, 1896–1909.e14 (2015).

    Article  CAS  PubMed  Google Scholar 

  37. Liu, Z. et al. Fate mapping via Ms4a3-expression history traces monocyte-derived cells. Cell 178, 1509–1525 e19 (2019).

    Article  CAS  PubMed  Google Scholar 

  38. Andrews, T. S. et al. Single-cell, single-nucleus, and spatial RNA sequencing of the human liver identifies cholangiocyte and mesenchymal heterogeneity. Hepatol. Commun. 6, 821–840 (2022).

    Article  CAS  PubMed  Google Scholar 

  39. Aizarani, N. et al. A human liver cell atlas reveals heterogeneity and epithelial progenitors. Nature 572, 199–204 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Ben-Moshe, S. et al. The spatiotemporal program of zonal liver regeneration following acute injury. Cell Stem Cell 29, 973–989 e10 (2022).

    Article  CAS  PubMed  Google Scholar 

  41. Yimlamai, D. et al. Hippo pathway activity influences liver cell fate. Cell 157, 1324–1338 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Yanger, K. et al. Robust cellular reprogramming occurs spontaneously during liver regeneration. Genes Dev. 27, 719–724 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Schaub, J. R. et al. De novo formation of the biliary system by TGFβ-mediated hepatocyte transdifferentiation. Nature 557, 247–251 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Ateeq, B. et al. Therapeutic targeting of SPINK1-positive prostate cancer. Sci. Transl. Med. 3, 72ra17 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  45. Nie, X. et al. Periostin: a potential therapeutic target for pulmonary hypertension? Circ. Res. 127, 1138–1152 (2020).

    Article  CAS  PubMed  Google Scholar 

  46. Cruciat, C. M. & Niehrs, C. Secreted and transmembrane wnt inhibitors and activators. Cold Spring Harb. Perspect. Biol. 5, a015081 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  47. Bird, T. G. & Müller, M. TGFβ inhibition restores a regenerative response in acute liver injury by suppressing paracrine senescence. Sci. Transl. Med. 10, eaan1230 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  48. Zhang, Y., Alexander, P. B. & Wang, X. F. TGF-β family signaling in the control of cell proliferation and survival. Cold Spring Harb. Perspect. Biol. 9, a022145 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  49. Takayama, K. et al. CCAAT/enhancer binding protein-mediated regulation of TGFβ receptor 2 expression determines the hepatoblast fate decision. Development 141, 91–100 (2014).

    Article  CAS  PubMed  Google Scholar 

  50. Sahoo, S., Mishra, A., Diehl, A. M. & Jolly, M. K. Dynamics of hepatocyte-cholangiocyte cell-fate decisions during liver development and regeneration. iScience 25, 104955 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Belenguer, G. et al. RNF43/ZNRF3 loss predisposes to hepatocellular-carcinoma by impairing liver regeneration and altering the liver lipid metabolic ground-state. Nat. Commun. 13, 334 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Sun, T. et al. ZNRF3 and RNF43 cooperate to safeguard metabolic liver zonation and hepatocyte proliferation. Cell Stem Cell 28, 1822–1837.e10 (2021).

    Article  CAS  PubMed  Google Scholar 

  53. Benhamouche, S. et al. Apc tumor suppressor gene is the “zonation-keeper” of mouse liver. Dev. Cell 10, 759–770 (2006).

    Article  CAS  PubMed  Google Scholar 

  54. Tago, K. & Nakamura, T. Inhibition of Wnt signaling by ICAT, a novel b-catenin-interacting protein. Genes Dev. 14, 1741–1749 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Thyssen, G. et al. LZTS2 is a novel β-catenin-interacting protein and regulates the nuclear export of β-catenin. Mol. Cell. Biol. 26, 8857–8867 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Aibar, S. et al. SCENIC: single-cell regulatory network inference and clustering. Nat. Methods 14, 1083–1086 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Song, Y. et al. Loss of ATOH8 increases stem cell features of hepatocellular carcinoma cells. Gastroenterology 149, 1068–1081.e5 (2015).

    Article  CAS  PubMed  Google Scholar 

  58. Liping Chen, J. Y. ATOH8 overexpression inhibits the tumor progression and monocyte chemotaxis in hepatocellular carcinoma. Int. J. Clin. Exp. Pathol. 13, 2534–2543 (2020).

    PubMed  PubMed Central  Google Scholar 

  59. Lei, L., Bruneau, A. & El Mourabit, H. Portal fibroblasts with mesenchymal stem cell features form a reservoir of proliferative myofibroblasts in liver fibrosis. Hepatology 76, 1360–1375 (2022).

    Article  CAS  PubMed  Google Scholar 

  60. Patel, N. et al. The transcription factor ATOH8 is regulated by erythropoietic activity and regulates HAMP transcription and cellular pSMAD1,5,8 levels. Br. J. Haematol. 164, 586–596 (2014).

    Article  CAS  PubMed  Google Scholar 

  61. The ENCODE Project Consortium. An integrated encyclopedia of DNA elements in the human genome. Nature 489, 57–74 (2012).

  62. Marra, F. & Tacke, F. Roles for chemokines in liver disease. Gastroenterology 147, 577–594 e1 (2014).

    Article  CAS  PubMed  Google Scholar 

  63. Campana, L., Esser, H., Huch, M. & Forbes, S. Liver regeneration and inflammation: from fundamental science to clinical applications. Nat. Rev. Mol. Cell Biol. 22, 608–624 (2021).

    Article  CAS  PubMed  Google Scholar 

  64. Andersson, E. R. In the zone for liver proliferation. Science 371, 887–888 (2021).

    Article  CAS  PubMed  Google Scholar 

  65. Itoh, T. The truth lies somewhere in the middle: the cells responsible for liver tissue maintenance finally identified. Cell Regen. 10, 28 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Deng, X. & Zhang, X. Hepatocyte generation in liver homeostasis, repair, and regeneration. Cell Regen. 23, 114 (2018).

    CAS  Google Scholar 

  67. Deng, X. et al. Chronic liver injury induces conversion of biliary epithelial cells into hepatocytes. Cell Stem Cell 23, 114–122 e3 (2018).

    Article  CAS  PubMed  Google Scholar 

  68. SAW. GitHub https://github.com/STOmics/SAW (2023).

  69. STOmics. BGI Research https://www.stomics.tech/helpcenter (2023).

  70. Li, R., Li, D. & Nie, Y. IL-6/gp130 signaling: a key unlocking regeneration. Cell Regen. 12, 16 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Xu, Z. et al. STOmicsDB: a comprehensive database for spatial transcriptomics data sharing, analysis and visualization. Nucleic Acids Res. 52, D1053–D1061 (2023).

    Article  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank Qi Zhou at Chinese Academy of Sciences for the long-term support in liver regeneration and cell therapy, Zhaoyuan Liu and Florent Ginhoux from the Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, for sharing transgenic mice, Shan Zhen from the Children’s Hospital of Fudan University for sharing human BA data, Hong Li, Luonan Chen and Gangqi Wang for constructive suggestions. L.H. group was supported by the National Natural Science Foundation of China (NSFC) (92368301 and 92168202), Shanghai Municipal Science and Technology Major Project, the National Key Research and Development Project (2019YFA0801503), Shanghai Science and Technology Committee (22JC1403001), the National Key Research and Development Project (2019YFA0802001) and the National Natural Science Foundation of China (NSFC) (32221002 and 31930030). X.X. was supported by the National Key Research and Development Program of China (2022YFC3400400). M.A.E. was supported by the National Natural Science Foundation of China (U20A2015). Yiwei L. was supported by National Natural Science Foundation of China (32200688). L.L. was supported by the Shenzhen Basic Research Project for Excellent Young Scholars (RCYX20200714114644191) and National Key Research and Development Program of China (2022YFC3400405). The project also received funding from the Guangdong Genomics Data Center (2021B1212100001). We thank all members of our laboratories for their support, and the China National GeneBank for providing technical support.

Author information

Authors and Affiliations

Authors

Contributions

B.W., X.S., H.N., P.G., Yiwei L., L.L., M.A.E. and L.H. conceived the idea; Yiwei L., L.L., M.A.E. and L.H. supervised the study; H.N., J.X., P.G., Yiwei L., M.A.E. and L.H. designed the experiment; H.N., J.X. and P.G. performed the majority of the experiments with the help of B.W., S.S., L.C. and J.C.; B.W., X.S., S.H. and Yiwei L. analyzed the data; Q.D., Y.W., Chang L., Y.S., X.L., Z.W., Y.Y., W.M., R.L., Chuanyu L., A.C. and X.X. provided technical support; Yikang L., Q.Q. and X.M. provided mouse disease tissues; W.D., T.L. and T.Y. constructed the CIRSTA website; B.W., X.S., H.N. and L.H. wrote the paper.

Corresponding authors

Correspondence to Yiwei Lai, Longqi Liu, Miguel A. Esteban or Lijian Hui.

Ethics declarations

Competing interests

The chip, procedure and applications of Stereo-seq are described in a pending patent, with A.C., X.X. and L.L. listed as the applicants. All other authors declare no competing interests.

Peer review

Peer review information

Meritxell Huch, Shalev Itzkovitz, Noémi Van Hu 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 Characterization of DDC treatment experiment and liver zonation in Stereo-seq.

a. H&E-staining on liver sections at all time points after DDC injury and repair. The staining was repeated with at least five independent biological replicates with similar results. Scale bars, 100 μm. b. Immunostaining images to determine the abundance of cholangiocyte (Ck19+, red, top) at D0, D17 and R21, LPLC (Opn+Hnf4a+, green and red, middle, indicated by arrows) at D0, D17 and R21, and proliferating hep (Ki67+Hnf4a+, green and red, bottom, indicated by arrows) at D0, D17 and R2. The staining was repeated with at least five independent biological replicates with similar results. Scale bars, 100 μm. c. Quantification of Ck19+ area, Opn+Hnf4a+ and Ki67+ hepatocytes at all time points (D0, n = 5; D8, n = 5; D17, n = 5; R2, n = 6; R7, n = 5; R21, n = 5). Data were presented as mean ± SD. d. Principal-component analysis of global gene expression profiles in biological replicates of Stereo-seq. e. Pearson’s correlation coefficient pairwise matrix for each biological replicate of Stereo-seq. f. The workflow of zonation landmark genes identification. g. Spatial visualization of Cyp2e1 and Cyp2f2 in representative Stereo-seq sections across all timepoints. Scale bars, 500 μm. h. Line chart showing the z-score of average expression per zonation layer of landmark genes used for defining the zonation layers in all Stereo-seq sections during injury and repair.

Source data

Extended Data Fig. 2 Additional characterization of liver zonation and spatial domain.

a. Spatial visualization of the nine zonation layers from central vein to portal vein in representative Stereo-seq sections at D8, R2, R7, and R21. Scale bars, 500 μm. b. Spatial visualization of the pericentral (CV), midzonal (MZ), and periportal (PV) area in representative Stereo-seq sections at D17. Scale bars, 500 μm. Black line indicated the position of median zonation layer, which matched with the midzonal area. c. Line chart showing zonation score of 9 zonation layers among each replicate in Stereo-seq sections for each timepoint. d. UMAP of unsupervised 17 clusters in Stereo-seq. e. Hierarchical clustering of expression profiles of 17 spatial clusters (Clusters 5 and 16, chol-domain; Cluster 2, LPLC domain; Cluster 9, portal vein-area; Other clusters, hepa-domain). f. Distinct markers for each domain from Fig. 1e (log2foldchange ≥ 0.25, q < 0.05). g. UMAP of annotated domains in Stereo-seq. h. Spatial visualization of canonical signaling pathways on a representative D17 slide. Scale bar, 500 μm for whole view and 100 μm for magnified view. i. Average intensity of canonical signaling pathways in each domain on Stereo-seq sections during injury.

Source data

Extended Data Fig. 3 Characterization of scRNA-seq data.

a. Expression of top DEGs (log2foldchange ≥ 0.25, q < 0.05) for each annotated cell type in scRNA-seq. b. GSEA analysis of reported RRG (reprogramming-related genes) in LPLC and hepatocyte pseudo-bulk scRNA-seq data during DDC injury. c. Adgre1, Clec4f, and LAM marker (Gpnmb and Fabp5) on non-KC macrophage UMAP of scRNA-seq. d. GSEA analysis of reported LAM-enriched genes in non-KC macrophages and KC pseudo-bulk scRNA-seq data during DDC injury. e. UMAP of dynamics of indicated cell types in scRNA-seq data at D0 and D17. Circle stood for the LAM populations. f. Magnified view of the cholangiocyte in the area selected from Fig. 1h and immunofluorescence staining for Ck19 (yellow) in the matched area from an adjacent section. Scale bar, 100 μm. g. Spatial correspondence of spatial domain and RCTD deconvolution result on a representative D17 slide. Left panel indicated the comparison of chol-domain and cholangiocyte in RCTD. Right panel indicated the comparison of LPLC-domain and LPLC in RCTD. Green points stood for the overlap bins of two methods. Scale bar, 500 μm.

Source data

Extended Data Fig. 4 The spatiotemporal and cellular dynamics of tissue damage.

a. TUNEL (green) staining at D0 and D17 (n = 5 each). Scale bar, 100 μm. b. Spatial visualization of damage response signaling activities in D0, D8, and D17 sections. Scale bar, 100 μm. Black line indicated position of median zonation layer. CV indicated pericentral region. Zonal module distribution at D17 (Kruskal-Wallis test, *q < 0.05). c. DAMP gene expression for each zonation layer in Stereo-seq sections upon injury. d. Bgn (green) immunostaining at D0 and D17 (n = 2 each; a single representative field of view is shown). Scale bar, 100 μm. e. Slc4a2 expression in cholangiocytes from scRNA-seq during injury. f. Expression of genes related to activation and fibrogenesis in fibroblasts and HSCs from scRNA-seq between D0 and injury (D8 and D17). As shown in Fig. 5b, most fibroblasts were distributed in PV, indicated they were portal fibroblasts. g. GO and KEGG biological pathway analysis of DEGs of each innate immune cell cluster (log2foldchange ≥ 0.25, q < 0.05). h. Top: experiment design of DAPM-induced and BDL model. Bottom left: Pdpn (green) and Ck19 (red) staining on liver sections of WT (n = 5), DAPM (n = 4) and BDL (n = 5) mice. Arrows indicated Pdpn+Ck19 LyECs. Scale bar, 100 μm. Bottom right: quantification of Pdpn+Ck19- LyECs of WT, DAPM and BDL mice. Mean ± SD; *p = 0.0113, ***p = 0.0001, two-sided t-test. i. Top: experiment design of Mdr2-/- model. Bottom left: Pdpn (green) and Ck19 (red) staining on liver sections of control and Mdr2-/- (n = 5 each). Arrows indicated Pdpn+Ck19- LyECs. Scale bar, 100 μm. Bottom right: quantification Pdpn+Ck19- LyECs of WT and Mdr2-/- mouse. Mean ± SD; ***p = 0.0002, two-sided t-test. j. LyEC specific gene expression in cell types identified from BA scRNA-seq. k. Left: UMAP of cell types in BA scRNA-seq data. Box indicates LyEC and myeloid cell populations. Right: UMAP of LyEC dynamics in scRNA-seq data of normal and BA liver. Circle indicates the LyEC population. l. Periportal region (cluster 1) from human BA 10x Visium is spatially corresponding to LyEC enriched bins. m. Interaction strength of Vegfc/d ligand-receptor pairs for each zonation layer in Stereo-seq sections upon injury. n. Vegfc/d expression of in scRNA-seq upon injury. o. GO and KEGG terms for LSEC and LyEC DEG enriched pathway (log2foldchange ≥ 0.25, q < 0.05). p. Sema3a expression in LyEC scRNA-seq upon injury. q. Spatial visualization of Sema3a on D0, D8, and D17 sections. Zonal module distribution at D17 (Kruskal-Wallis test, *q < 0.05).

Source data

Extended Data Fig. 5 Decoding cellular components and cell-cell interactions in chol-domain.

a. Cell score distribution of the major cell types among hepa-domain, LPLC-domain, and chol-domain at D17. Foldchange indicated the fold change of average scores between chol-domain and other domains. Min-max normalization was applied before comparison. Cell scores lower than 0.01 were presented in grey. b. Spatial visualization of ligand and receptor modules in 226 periportal ligand-receptor interactions on D0, D8, and D17 sections for a representative area. Asterisks indicated module was zonal distribution, based on module score across nine layers at D17 (Kruskal-Wallis test, p < 0.05). Black line indicated the position of median zonation layer. CV indicated the pericentral region of lobule. c. The outgoing interactions from cholangiocyte. Collagen and laminin related receptors were not shown. d. The interaction mode of injury upregulated periportal ligand-receptor pairs in each cell type from scRNA-seq. e. The functional types of injury upregulated periportal ligand-receptor pairs in each cell type from scRNA-seq. f. Incoming and outgoing interaction weights within all cell types identified by CellChat based on D17 scRNA-seq data. The total weights of incoming or outgoing interactions within each cell type were summarized and displayed in the bar plot, respectively. Cholangiocyte is high on both incoming and outgoing interactions. g. Communication possibility of ligand-receptor pairs in incoming signalings of cholangiocyte enriched in D17 than in D0 after filtration (Statistical significance of each ligand-receptor pair is calculated by CellChat, p < 0.01). h. Expression of cholangiocyte specific receptors in scRNA-seq across D0, D8 and D17. i. Expression of cholangiocyte specific ligands in scRNA-seq across D0, D8 and D17.

Source data

Extended Data Fig. 6 LAM characterization.

a. Expression of cholangiocyte-driven signaling genes in the domain cluster of human BA 10x Visium. Cluster 9 indicated the chol-domain, which expressed high levels of KRT7 and KRT19. b. Chol-domain (unsupervised cluster 9) presented in human BA 10x Visium, which were spatially corresponding to cholangiocyte-driven signaling signature enriched bins. c. Spatial visualization of indicated interactions on D0 and D17 section for a representative area. Scale bar, 100 μm. Asterisk indicated zonal distribution of interactions across nine layers at D17 (Kruskal-Wallis test, q < 0.05). Black line indicated the position of median zonation layer. CV indicated the pericentral region of lobule. d. Left: co-staining of F4/80 (green) and Clec4f (red) on liver sections of WT (n = 5), DAPM (n = 4) and BDL (n = 5) mice. Scale bars, 100 μm. Right: the ratio of Clec4f F4/80+ macrophage area per field of view. Data were presented as mean ± SD. ****p < 0.0001, ****p < 0.0001, two-sided unpaired Student’s t-test. e. Left: co-staining of F4/80 (green) and Clec4f (red) on liver sections of control and Mdr2-/- mouse (n = 5 for each group). Scale bars, 100 μm. Right: the ratio of Clec4f- F4/80+ macrophage area per field of view. Data were presented as mean ± SD. ****p < 0.0001, two-sided unpaired Student’s t-test. f. Expression of indicated LAM specific genes in the cell types identified from the BA scRNA-seq. g. UMAP of dynamics of LAM in scRNA-seq data of human normal and BA liver. Circle indicates the LAM population. h. Chol-domain (unsupervised cluster 9) presented in human BA 10x Visium, which are spatially corresponding to LAM signature enriched bins. i. Top GO and KEGG terms for LAMs in scRNA-seq during DDC injury (log2foldchange ≥ 0.25, q < 0.05). Metascape calculated the statistical significance of each term enrichment (p-value) based on the accumulative hypergeometric test. j. Spatial visualization of fatty acid biosynthesis pathway on D0, D8, and D17 sections for a representative area. Scale bar, 100 μm. k. Left: Oil red O staining of liver sections at D0 and D17 (n = 5 for each time point). Scale bar, 100 μm. Right: the ratio of Oil Red O area per field of view. Data were presented as mean ± SD. *p = 0.0105, two-sided unpaired Student’s t-test. l. The ratio of tdTomato+ monocyte to total Ly6c+ monocyte based on the flow cytometer data at DDC 7 days (n = 3).

Source data

Extended Data Fig. 7 The transcriptomic heterogeneity of LPLCs.

a. Pearson’s correlation coefficient pairwise matrix between each epithelial cell cluster. b. Expression of indicated genes for each epithelial cell cluster in scRNA-seq. c. Left: co-staining of Hnf1b (green), Hnf4a (red) and Opn (white) on liver sections at all time points after DDC injury and recovery (D0, n = 5; D8, n = 5; D17, n = 5; R2, n = 6; R7, n = 5; R21, n = 5). Scale bars, 100 μm. Arrows indicated Hnf4a+Opn+Hnf1b+ LPLC2. Arrowheads indicated Hnf4a+Opn+Hnf1b LPLC1. The periportal field of view was shown. Right: quantification of two subtypes of LPLCs at all time points after DDC injury and recovery. Data were presented as mean ± SD. d. Left: co-staining of Hnf1b (green), Hnf4a (red) and Opn (white) on liver sections of WT (n = 5), DAPM (n = 4) and BDL (n = 5) mice. Scale bars, 100 μm. Right: the ratio of two subtypes of LPLCs per field of view. Data were presented as mean ± SD. e. Left: co-staining of Hnf1b (green), Hnf4a (red) and Opn (white) on liver sections of control and Mdr2-/- mouse (n = 5 for each group). Scale bars, 100 μm. Right: the ratio of two subtypes of LPLCs per field of view. Data were presented as mean ± SD. f. GO and KEGG biological pathway analysis of the top DEGs of each epithelial cell scRNA-seq cluster at D17 (log2foldchange ≥ 0.25, q < 0.05). Heatmap color stood for -log10(p-value). g. Average module score of YAP, NOTCH, and TGFβ signaling pathways in scRNA-seq of each epithelial cell cluster. h. The ratio of cell types in Fig. 4d along the hepatocyte proliferation branch in pseudotime. i. Heatmap displaying DEG expression for each cluster along inferred differentiation to cholangiocyte branch of pseudotime. Cells are colored by pseudotime.

Source data

Extended Data Fig. 8 The spatial heterogeneity of LPLC-domains.

a. Left: spatial visualization of subcluster of LPLC-domain across all timepoints based on Fig. 1e. Scale bars, 500 μm. Black line indicated the position of median zonation layer. Right: time point compositions of each domain labeled in the left panel. Data were presented as mean ± SEM (Section number: D0, n = 2; D8, n = 4; D17, n = 3; R2, n = 4; R7, n = 4; R21, n = 5. Rep1 and Rep2 represent the biological replicates). b. Pearson’s correlation coefficient pairwise matrix between each domain. c. Pseudotime trajectory analysis corresponding to the designated domains in Extended Data Fig. 8a from Stereo-seq data via Monocle2. d. Violin plots showing the differential enrichment of LPLC-domain1 (top) or LPLC-domain2 (bottom) genes in LPLC1 and LPLC2 from scRNA-seq. e. Cell score distribution of the major cell types along hepa-domain, LPLC-domain subtypes, and chol-domain at D17. Foldchange indicates the fold change of average scores between two LPLC-domain subtypes. Min-max normalization was applied before comparison. Cell scores lower than 0.01 were presented in grey. f. Expression of representative common ligands in domain of LPLC subtypes. g. Spatial feature plots showing the expression pattern of indicated genes (Top layer, black spots) in LPLC-domain1 and LPLC-domain2 (Bottom layer, green and yellow). h. Expression of specific ligands of LPLC-domain subtypes in the cell types identified from scRNA-seq at D17. i. Expression of specific receptors in both LPLC subtypes or LPLC2 from scRNA-seq at D17.

Source data

Extended Data Fig. 9 The spatiotemporal cellular dynamics and extrinsic cues during liver regeneration.

a. Average intensity of indicated cell scores for each zonation layer in all Stereo-seq sections (Two-sided wilcoxon rank sum test, Monocyte: p < 2.22e-16; LAM: p < 2.22e-16; Neutrophil: p < 2.22e-16; Cholangiocyte: p = 0.22; Fibroblast: p = 0.97). b. Expression of indicated genes in all Stereo-seq sections at each time point. c. Spatial visualization of the Mki67+ bins in representative Stereo-seq sections. Bins were colored by zonation layer. Scale bar, 500 μm. d. Temporal dynamics of normalized expression of Mki67 in all Stereo-seq sections. e. Expression of Mki67 in hepatocyte from scRNA-seq at each time point during injury and repair (Two-sided wilcoxon rank sum test; At R2, Layer7-9 versus Layer1-2: p = 5.07e-11, Layer7-9 versus Layer3-6: p < 2.22e-16; Layer3-6 versus Layer1-2: p < 2.22e-16; At R7, Layer7-9 versus Layer1-2: p < 2.22e-16, Layer7-9 versus Layer3-6: p = 1.30e-05; Layer3-6 versus Layer1-2: p = 1.08e-13). The range of data in each layer at each time point is presented by grey boxes. f. Average overall, periportal and pericentral ligand-receptor interaction strength for each zonation layer in all Stereo-seq sections at each time point. g. Left: pericentral enriched ligand-receptor interaction strength for each zonation layers in all Stereo-seq sections. Right: spatial visualization of interactions in the left panel for a representative area. Asterisks indicated that interaction distribution was zonal, based on ligand-receptor interactions across nine layers at R2 (Kruskal-Wallis test, q < 0.05). Black line indicated the position of median zonation layer. CV indicated the pericentral region of lobule. h. Average normalized expression of WNT promoting ligands for each zonation layer in all Stereo-seq sections at each time point. i. Expression of WNT promoting ligands in each cell type from scRNA-seq at R2. j. Ppib (red, positive control) and DapB (red, negative control) mRNA ISH co-staining with DAPI on liver sections. The staining was repeated independently two times with similar results. The periportal field of view was shown. Scale bar, 100 μm. k. Expression of indicated ligands in each cell type from scRNA-seq at R2. l. Average module score of TGFb signaling pathways in pericentral (CV Hep), midzonal (MZ Hep), and periportal (PV Hep) hepatocyte from scRNA-seq at D17 and R2. m. Average expression of cell cycle inhibitors in each zone of hepatocyte from scRNA-seq at D17.

Source data

Extended Data Fig. 10 Atoh8 suppressed the hepatocyte regeneration.

a. WNT suppressors’ zonation layer expression from Stereo-seq. b. Atoh8 expression in hepatocyte from scRNA-seq. c. Atoh8 expression in liver at D0, D17, and R2 via qPCR (n = 5 each). Mean ± SD; **p = 0.0025, **p = 0.0047, two-sided t-test. d. Gene expression in microarray data (GSE159676) of human healthy (LTH), primary sclerosing cholangitis (PSC), primary biliary cirrhosis (PBC) and autoimmune hepatitis (AIH) (Two-sided t-test, PSC-LTH, p = 0.024; PBC-LTH, p = 0.51; AIH-LTH, p = 0.064). e. Atoh8 and Hamp expression in WT, shNC and shAtoh8 hepatocytes at D17 via qPCR (n = 5 each). Mean ± SD; *p = 0.0128, ****p < 0.0001, two-sided t-test. f. Top: Atoh8 knockdown experiment design under resting condition. Bottom: H&E staining on liver sections from shNC (n = 5) and shAtoh8 (n = 4) mice. Scale bar, 100 μm. g. Serum ALT and AST levels in WT (n = 5), shNC (n = 5) and shAtoh8 (n = 4) mice under resting condition. Mean ± SD; ns, not significant, two-sided t-test. h. Left: Ck19 (red) staining on liver sections from shNC (n = 5) and shAtoh8 (n = 4) mice under resting condition. Right: ratio of Ck19 positive area. Scale bars, 100 μm. Mean ± SD; ns, not significant, two-sided t-test. i. Left: co-staining of Ki67 (green) and Hnf4a (red) on liver sections from shNC (n = 5) and shAtoh8 (n = 4) mice under resting condition. Right: ratio of Ki67+ hepatocytes. Scale bars, 100 μm. Mean ± SD; ns, not significant, two-sided t-test. j. Serum ALT, AST and ALP levels in shNC (n = 5) and shAtoh8 (n = 5) mice at D17. Mean ± SD; ns, not significant, ****p < 0.0001, two-sided t-test. k. Left: Ck19 (red) staining on liver sections from shNC (n = 5) and shAtoh8 (n = 5) mice at D17. Right: ratio of Ck19 positive area. Scale bars, 100 μm. Mean ± SD; ns, not significant, two-sided t-test. l. Ccnd1 and Mki67 expression in WT, shNC and shAtoh8 hepatocytes at D17 via qPCR (n = 5 each). Mean ± SD; ***p = 0.0002, two-sided t-test. m. Ki67 hepatocyte quantification in periportal, middle, and pericentral region from shNC and shAtoh8 mice (n = 5 each). Mean ± SD; ns, not significant, *p = 0.0247, **p = 0.0031, ***p = 0.0002, ***p = 0.0002, two-way ANOVA with Sidak’s test. n. ATOH8 ChIP-seq signal tracks near cell cycle gene loci in human (GSE170527). Shaded regions are ATOH8 bounding sites. o. Left: ratio of Ki67+ hepatocytes to total hepatocytes on liver under DAPM treatment (0d,n = 5; 2d,n = 5; 4d,n = 5; 7d,n = 5; 10d,n = 3). Right: Atoh8 expression in liver at 0d, 2d and 4d via qPCR (n = 5 each). Mean ± SD; ns, not significant; **p = 0.0023, two-sided t-test.

Source data

Supplementary information

Supplementary Information

Supplementary Fig. 1, methods and Tables 1–6.

Reporting Summary

Supplementary Tables 1–6

Supplementary Tables 1–6.

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 5

Statistical source data.

Source Data Fig. 6

Statistical source data.

Source Data Extended Data Fig./Table 1

Statistical source data.

Source Data Extended Data Fig./Table 2

Statistical source data.

Source Data Extended Data Fig./Table 3

Statistical source data.

Source Data Extended Data Fig./Table 4

Statistical source data.

Source Data Extended Data Fig./Table 5

Statistical source data.

Source Data Extended Data Fig./Table 6

Statistical source data.

Source Data Extended Data Fig./Table 7

Statistical source data.

Source Data Extended Data Fig./Table 8

Statistical source data.

Source Data Extended Data Fig./Table 9

Statistical source data.

Source Data Extended Data Fig./Table 10

Statistical source data.

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

Wu, B., Shentu, X., Nan, H. et al. A spatiotemporal atlas of cholestatic injury and repair in mice. Nat Genet (2024). https://doi.org/10.1038/s41588-024-01687-w

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1038/s41588-024-01687-w

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