Abstract
Tissue macrophages have an important role in the maintenance of liver homeostasis, and their functions are closely related to spatial localization. Here, through integration of whole liver lobe imaging and single-cell RNA sequencing analysis of CX3CR1+ cells in the mouse liver, we identified a dense network of CX3CR1+CD63+ liver portal area macrophages (LPAMs) that exhibited transcriptional and spatial differences compared with CX3CR1+CD207+ liver capsular macrophages. The survival of LPAMs was dependent on colony-stimulating factor 1 receptor (CSF1R). LPAMs colonized the hepatic portal area of mice after birth and were replenished from bone-marrow-derived cells during liver homeostasis. LPAMs efficiently captured antigens derived from hepatocytes and closely interacted with sympathetic nerves around the portal vein. Deletion of LPAMs led to increased neutrophil infiltration and worsened sympathetic nerve degeneration during hepatic nonalcoholic steatohepatitis. In summary, our results provide insights into a distinct subset of nerve-associated portal macrophages that function to maintain liver immune homeostasis.
This is a preview of subscription content, access via your institution
Access options
Access Nature and 54 other Nature Portfolio journals
Get Nature+, our best-value online-access subscription
$32.99 / 30 days
cancel any time
Subscribe to this journal
Receive 12 print issues and online access
$259.00 per year
only $21.58 per issue
Buy this article
- Purchase on SpringerLink
- Instant access to full article PDF
Prices may be subject to local taxes which are calculated during checkout








Similar content being viewed by others
Data availability
scRNA-seq and bulk RNA-seq data have been deposited in the SRA database (accession number: PRJNA974709). All other data are available in the main text or the supplementary materials. Source data are provided with this paper.
References
Mass, E., Nimmerjahn, F., Kierdorf, K. & Schlitzer, A. Tissue-specific macrophages: how they develop and choreograph tissue biology. Nat. Rev. Immunol. https://doi.org/10.1038/s41577-023-00848-y (2023).
Guilliams, M., Thierry, G. R., Bonnardel, J. & Bajenoff, M. Establishment and maintenance of the macrophage niche. Immunity 52, 434–451 (2020).
Kierdorf, K., Prinz, M., Geissmann, F. & Gomez Perdiguero, E. Development and function of tissue resident macrophages in mice. Semin. Immunol. 27, 369–378 (2015).
Okabe, Y. & Medzhitov, R. Tissue biology perspective on macrophages. Nat. Immunol. 17, 9–17 (2016).
Davies, L. C., Jenkins, S. J., Allen, J. E. & Taylor, P. R. Tissue-resident macrophages. Nat. Immunol. 14, 986–995 (2013).
Davies, L. C. et al. A quantifiable proliferative burst of tissue macrophages restores homeostatic macrophage populations after acute inflammation. Eur. J. Immunol. 41, 2155–2164 (2011).
Amit, I., Winter, D. R. & Jung, S. The role of the local environment and epigenetics in shaping macrophage identity and their effect on tissue homeostasis. Nat. Immunol. 17, 18–25 (2016).
Ural, B. B. et al. Identification of a nerve-associated, lung-resident interstitial macrophage subset with distinct localization and immunoregulatory properties. Sci. Immunol. 5, eaax8756 (2020).
Dawson, C. A. et al. Tissue-resident ductal macrophages survey the mammary epithelium and facilitate tissue remodelling. Nat. Cell Biol. 22, 546–558 (2020).
David, B. A. et al. Combination of mass cytometry and imaging analysis reveals origin, location, and functional repopulation of liver myeloid cells in mice. Gastroenterology 151, 1176–1191 (2016).
Aizarani, N. et al. A human liver cell atlas reveals heterogeneity and epithelial progenitors. Nature 572, 199–204 (2019).
Wen, Y., Lambrecht, J., Ju, C. & Tacke, F. Hepatic macrophages in liver homeostasis and diseases-diversity, plasticity and therapeutic opportunities. Cell. Mol. Immunol. 18, 45–56 (2021).
Racanelli, V. & Rehermann, B. The liver as an immunological organ. Hepatology 43, S54–S62 (2006).
Gola, A. et al. Commensal-driven immune zonation of the liver promotes host defence. Nature 589, 131–136 (2021).
Ginhoux, F. & Jung, S. Monocytes and macrophages: developmental pathways and tissue homeostasis. Nat. Rev. Immunol. 14, 392–404 (2014).
Sierro, F. et al. A liver capsular network of monocyte-derived macrophages restricts hepatic dissemination of intraperitoneal bacteria by neutrophil recruitment. Immunity 47, 374–388.e376 (2017).
Remmerie, A. et al. Osteopontin expression identifies a subset of recruited macrophages distinct from Kupffer cells in the fatty liver. Immunity 53, 641–657.e614 (2020).
Daemen, S. et al. Dynamic shifts in the composition of resident and recruited macrophages influence tissue remodeling in NASH. Cell Rep. 34, 108626 (2021).
Guilliams, M. et al. Spatial proteogenomics reveals distinct and evolutionarily conserved hepatic macrophage niches. Cell 185, 379–396.e338 (2022).
Hankeova, S. et al. DUCT reveals architectural mechanisms contributing to bile duct recovery in a mouse model for Alagille syndrome. eLife 10, e60916 (2021).
Huang, S. et al. Three-dimensional mapping of hepatic lymphatic vessels and transcriptome profiling of lymphatic endothelial cells in healthy and diseased livers. Theranostics 13, 639–658 (2023).
Murakami, T. C. et al. A three-dimensional single-cell-resolution whole-brain atlas using CUBIC-X expansion microscopy and tissue clearing. Nat. Neurosci. 21, 625–637 (2018).
Zhang, Q. et al. Multiscale reconstruction of various vessels in the intact murine liver lobe. Commun. Biol. 5, 260 (2022).
Liu, Z. et al. Mesoscale visualization of three-dimensional microvascular architecture and immunocyte distribution in intact mouse liver lobes. Theranostics 12, 5418–5433 (2022).
Muller, P. A. et al. Crosstalk between muscularis macrophages and enteric neurons regulates gastrointestinal motility. Cell 158, 300–313 (2014).
Jacobson, A., Yang, D., Vella, M. & Chiu, I. M. The intestinal neuro-immune axis: crosstalk between neurons, immune cells, and microbes. Mucosal Immunol. 14, 555–565 (2021).
Jin, X. & Yamashita, T. Microglia in central nervous system repair after injury. J. Biochem. 159, 491–496 (2016).
Miller, B. M., Oderberg, I. M. & Goessling, W. Hepatic nervous system in development, regeneration, and disease. Hepatology 74, 3513–3522 (2021).
Ydens, E. et al. Profiling peripheral nerve macrophages reveals two macrophage subsets with distinct localization, transcriptome and response to injury. Nat. Neurosci. 23, 676–689 (2020).
Wang, P. L. et al. Peripheral nerve resident macrophages share tissue-specific programming and features of activated microglia. Nat. Commun. 11, 2552 (2020).
Liu, F., Song, Y. & Liu, D. Hydrodynamics-based transfection in animals by systemic administration of plasmid DNA. Gene Ther. 6, 1258–1266 (1999).
Yu, S. & Vernia, S. A Transposon-based mouse model of hepatocellular carcinoma via hydrodynamic tail vein injection. Methods Mol. Biol. 2164, 129–143 (2020).
Bain, C. C. et al. Constant replenishment from circulating monocytes maintains the macrophage pool in the intestine of adult mice. Nat. Immunol. 15, 929–937 (2014).
Khosravi, A. et al. Gut microbiota promote hematopoiesis to control bacterial infection. Cell Host Microbe 15, 374–381 (2014).
Greter, M. et al. Stroma-derived interleukin-34 controls the development and maintenance of langerhans cells and the maintenance of microglia. Immunity 37, 1050–1060 (2012).
MacDonald, K. P. et al. An antibody against the colony-stimulating factor 1 receptor depletes the resident subset of monocytes and tissue- and tumor-associated macrophages but does not inhibit inflammation. Blood 116, 3955–3963 (2010).
Varol, C., Mildner, A. & Jung, S. Macrophages: development and tissue specialization. Annu. Rev. Immunol. 33, 643–675 (2015).
Guilliams, M. & Scott, C. L. Liver macrophages in health and disease. Immunity 55, 1515–1529 (2022).
Adori, C. et al. Disorganization and degeneration of liver sympathetic innervations in nonalcoholic fatty liver disease revealed by 3D imaging. Sci. Adv. 7, eabg5733 (2021).
Liu, K. et al. Metabolic stress drives sympathetic neuropathy within the liver. Cell Metab. 33, 666–675.e664 (2021).
Hu, M. et al. CD63 acts as a functional marker in maintaining hematopoietic stem cell quiescence through supporting TGFβ signaling in mice. Cell Death Differ. 29, 178–191 (2022).
Miyamoto, Y. et al. Periportal macrophages protect against commensal-driven liver inflammation. Nature 629, 901–909 (2024).
Dani, N. et al. A cellular and spatial map of the choroid plexus across brain ventricles and ages. Cell 184, 3056–3074.e3021 (2021).
Kim, Y. C. et al. Immaturity of immune cells around the dural venous sinuses contributes to viral meningoencephalitis in neonates. Sci. Immunol. 8, eadg6155 (2023).
Zou, J., Li, J., Wang, X., Tang, D. & Chen, R. Neuroimmune modulation in liver pathophysiology. J. Neuroinflammation 21, 188 (2024).
Hurr, C., Simonyan, H., Morgan, D. A., Rahmouni, K. & Young, C. N. Liver sympathetic denervation reverses obesity-induced hepatic steatosis. J. Physiol. 597, 4565–4580 (2019).
Renier, N. et al. iDISCO: a simple, rapid method to immunolabel large tissue samples for volume imaging. Cell 159, 896–910 (2014).
Lun, A. T. L. et al. EmptyDrops: distinguishing cells from empty droplets in droplet-based single-cell RNA sequencing data. Genome Biol. 20, 63 (2019).
McGinnis, C. S., Murrow, L. M. & Gartner, Z. J. DoubletFinder: doublet detection in single-cell RNA sequencing data using artificial nearest neighbors. Cell Syst. 8, 329–337.e324 (2019).
Hao, Y. et al. Integrated analysis of multimodal single-cell data. Cell 184, 3573–3587.e3529 (2021).
Wu, T. et al. clusterProfiler 4.0: a universal enrichment tool for interpreting omics data. Innovation 2, 100141 (2021).
Acknowledgements
We thank G. Shen from Tongji Medical College and L. G. Ng for discussions. We also thank the Optical Bioimaging Core Facility of WNLO-HUST for data acquisition assistance, the Analytical and Testing Center of HUST for spectral measurements, and the Optical Bioimaging Core Facility of WNLO-HUST and the Research Core Facilities for Life Science (HUST) for their support in data acquisition. This work was supported by the State Key Program of the National Natural Science Foundation of China (grant no. 32330048, Z.Z.), the National Key Research and Development Program of China (grant no. 2017YFA0700403, Z.Z.), the National Natural Science Foundation of China (grant no. 81827901, Q.L.) and the Science and Technology Talent Innovation Project in Hainan Province (grant no. KJRC2023B09, Z.H.Z.).
Author information
Authors and Affiliations
Contributions
Z.L., M.X., Z.Z. and Q.L. initiated and designed the project. M.X., Z.L., Z.C., X.L., S.H., X.W., J.Y., J.L., Z.F., Y.L., L.G. and Y.H. performed the experiments. M.X., Z.L., Q.P., B.L. and Y.Z. assisted with data processing. M.X., Z.L., Q.P. and Z.Z. analyzed the data and wrote the paper.
Corresponding authors
Ethics declarations
Competing interests
The authors declare no competing interests.
Peer review
Peer review information
Nature Immunology thanks Frederic Geissmann, Matteo Iannacone and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. 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 The distribution and phenotype of CX3CR1+ cells in liver lobe.
(a) Segmentation of CX3CR1+ cells in the intact liver lobe. HV in cyan, PV in magenta, the portal area CX3CR1+ cells in yellow, and the capsular CX3CR1+ cells in blue. Distance from CX3CR1+ cells to the liver capsule and the distance from CX3CR1+ cells to the portal region. The cut-offs range from 20–100 μm. The red line indicates CX3CR1+ cells located within 50 μm of the portal region, while the blue line represents CX3CR1+ cells located within 50 μm of the capsular region. (b) Whole liver lobe imaging of CX3CR1+ cells and portal structures with high resolution. PV in blue, BD in magenta, and CX3CR1+ cells in green. Scale bars, 1500 µm. Distances from CX3CR1+ cells to the PV and the BD were quantified based on the information obtained in Cx3cr1GFP/+ mice liver lobe. The red gate represents the distance from CX3CR1+ cells to PV was less than 20 μm, and the blue gate represents the distance from CX3CR1+ cells to BD was less than 20 μm. (c-d) Liver portal area (c) and capsular area (d) CX3CR1+ cells express macrophage markers. Immunofluorescence imaging of liver sections stained with antibodies specific for CD11b, CD64, CD68, Ly6C, and CD207 (white). CX3CR1+ cells in green. Scale bars, 50 µm. (e) Quantitative analysis of the proportion of CX3CR1+ cells expressing MHC-II, F4/80, CD11b, CD64, and CD68 in the portal area, n = 5 imaging regions for each group. Mean ± SD. (f) Confocal microscopic imaging of liver immunofluorescence sections stained with antibodies specific for MHC-II (blue), CD64 (magenta), and CD11c (yellow). CX3CR1+ cells in green. Segmentation of CX3CR1+ macrophage (CX3CR1+MHC-II+CD64+CD11c-), cDC2 (CX3CR1+MHC-II+CD64-CD11c+) and cDC1 (CX3CR1-MHC-II+CD64-CD11c+) in portal area. The macrophage is shown in green, cDC1 is shown in blue, and cDC2 is shown in yellow. (g) The pie chart was used to calculate the percentage of CD64+ macrophages and CD64- cells in CX3CR1+ MHC-II+ cells around the liver portal area. Data were collected from 6 imaging regions. The imaging data in b-d, f were from representative experiments.
Extended Data Fig. 2 Flow cytometry and GO enrichment analyses of Cx3cr1GFP/+ mouse liver cells.
(a) Flow cytometric analysis of liver LPAMs and LCMs gated on CD45+CX3CR1+ cells in whole Cx3cr1GFP/+ mice liver NPCs. (b) Flow cytometric analysis of liver LPAMs and LCMs gated on CD45+CX3CR1+ cells in vascular-enriched Cx3cr1GFP/+ mice liver NPCs. (c) The fluorescence minus one (FMO) control (without CD207 or CD63) for the flow cytometric gated strategy of LPAMs and LCMs. (d) Gene Ontology (GO) enrichment analysis in Pmepa1+Zmynd15+ LPAMs, Gpnmb+Spp1+ LPAMs, LCMs, Il1rn Macs and Ptprb Macs. (e) CD63 expression in human liver section: https://www.proteinatlas.org/ENSG00000135404-CD63/tissue/Liver#img. CD68 expression in human liver section: https://www.proteinatlas.org/ENSG00000129226-CD68/tissue/liver#img. C1QC expression in human liver section: https://www.proteinatlas.org/ENSG00000159189-C1QC/tissue/liver#img. HLA-DR expression in human liver section: https://www.proteinatlas.org/ENSG00000204287-HLA-DRA/tissue/liver#img. VSIG4 expression in human liver section:https://www.proteinatlas.org/ENSG00000155659-VSIG4/tissue/liver#. The green arrows indicated KCs in the hepatic sinusoid and the red arrows indicated portal macrophages.
Extended Data Fig. 3 Liver TH nerve distribution and nerve-associated macrophage comparison.
(a) Large volume imaging and different depths of the distribution of TH in the liver. Vascular in blue, TH in Green, liver boundary in Gray. Scale bars, 150 µm. This result was also supplied in the supplementary video 6. The imaging data were from representative experiments. (b) Comparison of transcriptional profiles among several nerve-associated macrophage populations. Hierarchical clustering based on transcriptomic data.
Extended Data Fig. 4 The response of LPAMs upon antigen or bacteria challenge.
(a) Imaging of LPAMs capturing hepatocyte-derived antigens. Scale bars, 20 µm. CX3CR1+ LPAMs in green, tfRFP-expressing HepG2 cells in magenta. The blue arrow indicates LPAMs that have phagocytosed tfRFP. (b) Apoptotic Raji cells (Raji-mcherry) were injected into Cx3cr1GFP/+ mice. Flow cytometry was used to detect the phagocytosis of cellular debris by LPAMs and KCs. The statistical results of the mean fluorescence intensity (MFI) of antigen and dead cells captured by LPAMs and KCs, as detected by flow cytometry, n = 4 per group. Mean ± SD, two-tailed unpaired t-test. (c) Imaging of LPAMs and KCs in the liver during infection with tfRFP-expressed E.coli-tfRFP. CX3CR1+ cells in green, F4/80 in white and E.coli-tfRFP in magenta. (d) GSEA analysis of the ‘Defense response to bacterium’ pathway between LPAMs versus LCMs, and LPAMs versus KCs. The top 20 genes with expression differences are listed on the right panel. The imaging data in a, c were from representative experiments.
Extended Data Fig. 5 The distribution of CX3CR1+ LPAMs and Treg cells at portal area.
(a) Immunofluorescence imaging of the liver portal area stained with DAPI (blue), MHC-II (magenta), CD3 (yellow), and the CX3CR1+ cells in green. Scale bar, 100 µm. The data in the white and yellow box were enlarged. Scale bars, 20 μm and 5 µm. (b) Immunofluorescence imaging of the liver portal area stained with an antibody specific for CD4 (magenta). CX3CR1+ cells in green. Scale bars, 100 µm. (c) Immunofluorescence imaging of liver sections stained with antibodies specific for F4/80 (red) and MHC-II (blue). Foxp3+ cells in green. Scale bars, 20 µm. (d) The 3D confocal imaging of a CUBIC-cleared liver section from a Foxp3GFP/+ mouse. Foxp3+ cells in green and Foxp3+ cells around PV in magenta. Scale bars, 200 µm. (e) The distribution of CX3CR1+ cells was detected by immunofluorescence. The CX3CR1+ cells in green, F4/80 in red, MHCII in white, CD63 in magenta. Scale bars, 30 µm. The imaging data in a-e were from representative experiments. (f-g) Percentage of the MHCII+ population in the CX3CR1+F4/80+CD63+ cells (f) as well as the percentage of mature LPAMs (CX3CR1+F4/80+MHC-II+CD63+) in live NPCs (g) from Cx3cr1GFP/+ mice at different weeks after birth. The data were pooled from three experiments, n = 5 (day 0), n = 3 (day 42), n = 4 in other days. Means ± SD in f, and means ± SEM in g, two-tailed unpaired t-test.
Extended Data Fig. 6 LPAMs maintenance depended on CSF1R.
(a) LPAMs and LCMs from control (Cx3cr1GFP/+ mice) and antibiotic-treated mice were stained and analyzed by flow cytometry to assess the influence of microbiota described in Fig. 6c, d. (b) Go enrichment analysis of the upregulated pathway in LCMs. (c) The upregulated genes in LCM from the ‘Cellular response to lipopolysaccharide’ pathway are listed on the right panel. (d) The imaging of liver sections for anti-IgG and anti-CSF1R injection Cx3cr1GFP/+ mice. Scale bars, 1000 μm. The data in the red and yellow boxes were enlarged from the left liver sections. Scale bars, 100 μm. (e) Imaging of liver sections from anti-IgG or anti-CSF1R treated liver and stained with antibody specific for F4/80 (red). The CX3CR1+ cells were shown in green. Scale bars, 100 µm. The imaging data in d-e were from representative experiments. (f) Flow cytometric analysis of CX3CR1+MHCII+ macrophages in anti-IgG or anti-CSF1R-treated liver. Mean ± SEM (two-tailed unpaired t-test), n = 3 mice per group.
Extended Data Fig. 7 The depletion efficiency of LPAM in Cx3cr1creER/+Csf1rfl/fl mice.
(a) Gating strategy for liver T cells (CD45+CD3+NK1.1−), NK cells (CD45+CD3-NK1.1+), NKT cells (CD45+CD3+NK1.1+), B cells (CD45+CD3-NK1.1-CD19+). (b) Gating strategy for liver cDC (CD45+CD64-CD11c+), cDC1 (CD45+CD64-CD11c+CD103+CD11b-), cDC2 (CD45+CD64-CD11c+CD103-CD11b+). (c) Gating strategy for circulating monocyte (CD45+CD11b+F4/80+). (d) Gating strategy for tissue macrophages (CD45+CD11b+F4/80+) from spleen, lung and intestine. (e) Gating strategy for LCM (CX3CR1+MHC-II+CD207+) in the liver. (f) The cell counts of liver T cells, Treg cells, NK cells, NKT cells, B cells, circulating monocytes, tissue macrophages, in the liver of tamoxifen (15 mg/kg) i.p. injected Cx3cr1creER/+Csf1rfl/fl mice (LPAMcDel) or Cx3cr1creER/+ mice (LPAMcCtr), mean ± SD, n = 3 mice per group; two-tailed unpaired t-test.
Extended Data Fig. 8 LPAM distribution in Cx3cr1GFP/+Flt3l−/− mice and MHCII expression in different LPAMs.
(a) The 3D confocal imaging of the mouse liver lobe from Cx3cr1GFP/+Flt3l−/− mice. Scale bar, 2000 μm. The imaging data were from representative experiments. (b) Calculate the percentage of MHC-II expression in CD45.1+ LPAMs (blood-borne) and CD45.2+ LPAMs (self-renewing) in CD45.2 mice. The data were pooled from three independent experiments, mean ± SD, n = 3 mice per group; two-tailed unpaired t-test.
Extended Data Fig. 9 Verification of LPAM depletion in Cx3cr1CreER-iDTR mice.
Cx3cr1CreER-iDTR mice were injected with different doses of TAMO (3 mg/kg, 15 mg/kg, and 75 mg/kg) and 50 µg/kg DT. Cx3cr1CreER-iDTR mice injected with TAMO and PBS were used as controls. (a-b) Gating strategy for LPAM (CX3CR1+MHC-II+CD63+), LCM (CX3CR1+MHC-II+CD207+), monocyte (CX3CR1+MHC-II-F4/80+), and DC (MHC-II+CD11c+CD64−) in the liver. (c) The depletion efficiency of LPAM, LCM, and monocyte in LPAMCtr and LPAMDel mice, n = 8 (Control, 15 mg/kg), n = 4 (3 mg/kg, 75 mg/kg). Mean ± SEM, two-tailed unpaired t-test. (d) The depletion efficiency of liver DCs in LPAMCtr and LPAMDel mice, n = 4 (Control), n = 3 (3 mg/kg, 15 mg/kg, 75 mg/kg). Mean ± SEM, two-tailed unpaired t-test. (e) The depletion efficiency of other tissue macrophages in LPAMCtr and LPAMDel mice, n = 4 in each group of lung and spleen Mac, n = 3 in each group of intestine Mac. Mean ± SEM, two-tailed unpaired t-test. (f-g) The biochemical analysis of liver ALT and AST, n = 7 (TAMO + PBS), n = 6 (TAMO + DT), n = 11 (MCD LPAMCtr), n = 8 (MCD LPAMDel). The data were pooled from at least two experiments (mean ± SEM, two-tailed unpaired t-test). (h-i) H&E and NAS score for NASH liver tissue, n = 5 (MCD LPAMCtr), n = 6 (MCD LPAMDel), n = 5 (MCD LPAMcCtr), n = 4 (MCD LPAMcDel). Mean ± SD, two-tailed unpaired t-test. The imaging data in g were from representative experiments.
Extended Data Fig. 10 The recovery of LPAMs and the numbers of different macrophages after MCD diet feeding.
(a) Percentage of LPAM in LPAMcDel mice at 7, 14, and 21 days post-tamoxifen, n = 13 (day 0), n = 6 (day 1), n = 4 (day 7), n = 7 (day 14), n = 4 (day 21). Mean ± SD, two-tailed unpaired t-test. (b) Percentage of BM monocytes in LPAMcDel mice at 7, 14, and 21 days post-tamoxifen, n = 7 (day 0), n = 3 (day 1), n = 4 (day 7), n = 3 (day 14), n = 4 (day 21). Mean ± SD, two-tailed unpaired t-test. (c-e) Flow cytometric analysis of the number of liver EmKCs, MoKCs, LAMs, LPAMs in LPAMcCtr mice or in LPAMcDel mice (mean ± SD, n = 3 mice per group; two-tailed unpaired t-test).
Supplementary information
Supplementary Information
Supplementary Methods and Tables 1–3.
Supplementary Video 1
The distribution of CX3CR1+ cells in Cx3cr1GFP/+ mouse liver. CX3CR1+ cells and vasculature from Fig. 1a were manually segmented with Imaris. The hepatic vein is shown in purple, the portal area GFP cells are shown in red, the capsular GFP cells are shown in cyan, and the lobular GFP cells are shown in yellow.
Supplementary Video 2
The distribution of CX3CR1+ cells in capsular and portal areas. The CX3CR1+ cells are shown in green, the hepatic vein is shown in cyan, the portal vein is shown in magenta, the portal area GFP cells are shown in yellow, and the capsular GFP cells are shown in blue.
Supplementary Video 3
Distribution of CX3CR1+ cells in the 745 × 745 × 746 μm3 portal area of the liver. CX3CR1+ cells are shown in green, the hepatic vein is shown in magenta, the portal vein is shown in gray, the lymphatic vessel (LV) is shown in yellow, the bile duct is shown in blue, and the hepatic artery is shown in red.
Supplementary Video 4
Portal vein CX3CR1+ cells and bile duct CX3CR1+ cells in intact liver lobe. CX3CR1+ cells are shown in green, the portal vein is shown in blue, the bile duct is shown in magenta, and the liver boundary is shown in gray.
Supplementary Video 5
LPAM interacts closely with nerves. Liver nerves were stained with TH (red); CX3CR1+ cells are shown in green.
Supplementary Video 6
Large-volume imaging of the distribution of TH in the liver. The liver vascular is shown in blue, TH is shown in green and the liver boundary is shown in gray
Supplementary Video 7
Portal CX3CR1+ cells interacted closely with T cells. Immunofluorescence imaging of the liver portal area stained with CD3 (yellow); CX3CR1+ cells are shown in green
Supplementary Video 8
Portal Foxp3+ cells contacted closely with F4/80+MHCII+ cells. Immunofluorescence imaging of the liver portal area stained with F4/80 (red) and MHCII (blue); Foxp3+ cells are shown in green.
Supplementary Video 9
The distribution of Foxp3+ cells in the liver. Foxp3+ cells are shown in green, and liver vessels are shown in blue.
Supplementary Video 10
Intact liver lobe at the age of P0–P21. CX3CR1+ cells are shown in green, and liver blood vessels are shown in red.
Supplementary Video 11
CX3CR1+ cells reconstituted by bone-marrow-derived cells. CX3CR1+ cells are shown in green, and liver vessels are shown in red.
Supplementary Video 12
Sympathetic innervations in the liver. Mice were fed with a standard chow diet (normal liver) or MCD (NASH liver). The cleared livers of the mice were processed for TH (green) immunolabeling.
Source data
Source Data Fig. 1
Statistical source data.
Source Data Fig. 5
Statistical source data.
Source Data Fig. 6
Statistical source data.
Source Data Fig. 7
Statistical source data.
Source Data Fig. 8
Statistical source data.
Source Data Extended Data Fig. 1
Statistical source data.
Source Data Extended Data Fig. 4
Statistical source data.
Source Data Extended Data Fig. 5
Statistical source data.
Source Data Extended Data Fig. 6
Statistical source data.
Source Data Extended Data Fig. 7
Statistical source data.
Source Data Extended Data Fig. 8
Statistical source data.
Source Data Extended Data Fig. 9
Statistical source data.
Source Data Extended Data Fig. 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.
About this article
Cite this article
Xu, M., Liu, Z., Pan, Q. et al. Neuroprotective liver portal area macrophages attenuate hepatic inflammation. Nat Immunol 26, 1048–1061 (2025). https://doi.org/10.1038/s41590-025-02190-y
Received:
Accepted:
Published:
Issue date:
DOI: https://doi.org/10.1038/s41590-025-02190-y
This article is cited by
-
Portal macrophages maintain liver homeostasis
Nature Immunology (2025)