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:

Limited proliferation capacity of aortic intima resident macrophages requires monocyte recruitment for atherosclerotic plaque progression

Abstract

Early atherosclerosis depends upon responses by immune cells resident in the intimal aortic wall. Specifically, the healthy intima is thought to be populated by vascular dendritic cells (DCs) that, during hypercholesterolemia, initiate atherosclerosis by being the first to accumulate cholesterol. Whether these cells remain key players in later stages of disease is unknown. Using murine lineage-tracing models and gene expression profiling, we reveal that myeloid cells present in the intima of the aortic arch are not DCs but instead specialized aortic intima resident macrophages (MacAIR) that depend upon colony-stimulating factor 1 and are sustained by local proliferation. Although MacAIR comprise the earliest foam cells in plaques, their proliferation during plaque progression is limited. After months of hypercholesterolemia, their presence in plaques is overtaken by recruited monocytes, which induce MacAIR-defining genes. These data redefine the lineage of intimal phagocytes and suggest that proliferation is insufficient to sustain generations of macrophages during plaque progression.

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: Profiling MacAIR in the steady state.
Fig. 2: MacAIR are Csf1-dependent.
Fig. 3: MacAIR develop from bone marrow progenitors and seed the aorta at birth.
Fig. 4: MacAIR are maintained independent of circulating progenitors and proliferate within the tissue.
Fig. 5: MacAIR promote monocyte recruitment in early atherosclerotic lesions.
Fig. 6: Fate mapping MacAIR in the progression of atherosclerosis.
Fig. 7: Aortic intima microenvironment promotes shared gene programs between resident macrophages in the steady state and disease progression.

Similar content being viewed by others

Data availability

Gene expression data (bulk RNA-seq or scRNA-seq) have been uploaded to the Gene Expression Omnibus (GEO) repository for public availability under accession codes GSE116271, GSE116239, GSE154817 and GSE154921. All other data that support the findings of this study are available from the corresponding author upon reasonable request.

References

  1. Geovanini, G. R. & Libby, P. Atherosclerosis and inflammation: overview and updates. Clin. Sci. 132, 1243–1252 (2018).

    Article  CAS  Google Scholar 

  2. Benjamin, E. J. et al. Heart disease and stroke statistics—2019 update: a report from the American Heart Association. Circulation 139, e56–e528 (2019).

    Article  PubMed  Google Scholar 

  3. Libby, P., Lichtman, A. H. & Hansson, G. K. Immune effector mechanisms implicated in atherosclerosis: from mice to humans. Immunity 38, 1092–1104 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Williams, J. W., Huang, L. H. & Randolph, G. J. Cytokine circuits in cardiovascular disease. Immunity 50, 941–954 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Ridker, P. M. et al. Anti-inflammatory therapy with canakinumab for atherosclerotic disease. N. Engl. J. Med. 377, 1119–1131 (2017).

    Article  CAS  PubMed  Google Scholar 

  6. Randolph, G. J. Mechanisms that regulate macrophage burden in atherosclerosis. Circ. Res. 114, 1757–1771 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Dick, S. A., Zaman, R. & Epelman, S. Using high-dimensional approaches to probe monocytes and macrophages in cardiovascular disease. Front. Immunol. 10, 2146 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Jongstra-Bilen, J. et al. Low-grade chronic inflammation in regions of the normal mouse arterial intima predisposed to atherosclerosis. J. Exp. Med. 203, 2073–2083 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Choi, J. H. et al. Identification of antigen-presenting dendritic cells in mouse aorta and cardiac valves. J. Exp. Med. 206, 497–505 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Meredith, M. M. et al. Expression of the zinc finger transcription factor zDC (Zbtb46, Btbd4) defines the classical dendritic cell lineage. J. Exp. Med. 209, 1153–1165 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Satpathy, A. T. et al. Zbtb46 expression distinguishes classical dendritic cells and their committed progenitors from other immune lineages. J. Exp. Med. 209, 1135–1152 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Zhu, S. N., Chen, M., Jongstra-Bilen, J. & Cybulsky, M. I. GM-CSF regulates intimal cell proliferation in nascent atherosclerotic lesions. J. Exp. Med. 206, 2141–2149 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Kim, K.-W. et al. MHC II+ resident peritoneal and pleural macrophages rely on IRF4 for development from circulating monocytes. J. Exp. Med. 213, 1951–1959 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Choi, J. H. et al. Flt3 signaling-dependent dendritic cells protect against atherosclerosis. Immunity 35, 819–831 (2011).

    Article  CAS  PubMed  Google Scholar 

  15. Roufaiel, M. et al. CCL19-CCR7–dependent reverse transendothelial migration of myeloid cells clears Chlamydia muridarum from the arterial intima. Nat. Immunol. 17, 1263–1272 (2016).

    Article  CAS  PubMed  Google Scholar 

  16. Paulson, K. E. et al. Resident intimal dendritic cells accumulate lipid and contribute to the initiation of atherosclerosis. Circ. Res. 106, 383–390 (2010).

    Article  CAS  PubMed  Google Scholar 

  17. Robbins, C. S. et al. Local proliferation dominates lesional macrophage accumulation in atherosclerosis. Nat. Med. 19, 1166–1172 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Gautier, E. L. et al. Gene expression profiles and transcriptional regulatory pathways that underlie the identity and diversity of mouse tissue macrophages. Nat. Immunol. 13, 1118–1128 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Kim, K. et al. Transcriptome analysis reveals nonfoamy rather than foamy plaque macrophages are proinflammatory in atherosclerotic murine models. Circ. Res. 123, 1127–1142 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Ensan, S. et al. Self-renewing resident arterial macrophages arise from embryonic CX3CR1+ precursors and circulating monocytes immediately after birth. Nat. Immunol. 17, 159–168 (2016).

    Article  CAS  PubMed  Google Scholar 

  21. Lim, H. Y. et al. Hyaluronan receptor LYVE-1-expressing macrophages maintain arterial tone through hyaluronan-mediated regulation of smooth muscle cell collagen. Immunity 49, 326–341 (2018).

    Article  CAS  PubMed  Google Scholar 

  22. Cochain, C. et al. Single-cell RNA-seq reveals the transcriptional landscape and heterogeneity of aortic macrophages in murine atherosclerosis. Circ. Res. 122, 1661–1674 (2018).

    Article  CAS  PubMed  Google Scholar 

  23. Lin, J. D. et al. Single-cell analysis of fate-mapped macrophages reveals heterogeneity, including stem-like properties, during atherosclerosis progression and regression. JCI Insight 4, e124574 (2019).

    Article  PubMed Central  Google Scholar 

  24. Wumesh, K. C. et al. L-Myc expression by dendritic cells is required for optimal T-cell priming. Nature 507, 243–247 (2014).

    Article  PubMed Central  CAS  Google Scholar 

  25. Perdiguero, E. G. & Geissmann, F. The development and maintenance of resident macrophages. Nat. Immunol. 17, 2–8 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Wang, Y. et al. IL-34 is a tissue-restricted ligand of CSF1R required for the development of Langerhans cells and microglia. Nat. Immunol. 13, 753–760 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Williams, J. W., Giannarelli, C., Rahman, A., Randolph, G. J. & Kovacic, J. C. Macrophage biology, classification and phenotype in cardiovascular disease: JACC macrophage in CVD series (part 1). J. Am. Coll. Cardiol. 72, 2166–2180 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Serbina, N. V. & Pamer, E. G. Monocyte emigration from bone marrow during bacterial infection requires signals mediated by chemokine receptor CCR2. Nat. Immunol. 7, 311–317 (2006).

    Article  CAS  PubMed  Google Scholar 

  29. 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 

  30. Zhu, Y. et al. Tissue-resident macrophages in pancreatic ductal adenocarcinoma originate from embryonic hematopoiesis and promote tumor progression. Immunity 47, 323–338 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Epelman, S. et al. Embryonic and adult-derived resident cardiac macrophages are maintained through distinct mechanisms at steady state and during inflammation. Immunity 40, 91–104 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Hashimoto, D. et al. Tissue-resident macrophages self-maintain locally throughout adult life with minimal contribution from circulating monocytes. Immunity 38, 792–804 (2013).

    Article  CAS  PubMed  Google Scholar 

  33. Davies, L. C. et al. Distinct bone marrow-derived and tissue-resident macrophage lineages proliferate at key stages during inflammation. Nat. Commun. 4, 1886 (2013).

  34. Scholzen, T. & Gerdes, J. The Ki-67 protein: from the known and the unknown. J. Cell. Physiol. 182, 311–322 (2000).

    Article  CAS  PubMed  Google Scholar 

  35. Williams, J. W. et al. Limited macrophage positional dynamics in progressing or regressing murine atherosclerotic plaques—brief report. Arterioscler. Thromb. Vasc. Biol. 38, 1702–1710 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Yona, S. et al. Fate mapping reveals origins and dynamics of monocytes and tissue macrophages under homeostasis. Immunity 38, 79–91 (2013).

    Article  CAS  PubMed  Google Scholar 

  37. Wang, M. et al. Interleukin-3/granulocyte macrophage colony-stimulating factor receptor promotes stem cell expansion, monocytosis and atheroma macrophage burden in mice with hematopoietic ApoE deficiency. Arterioscler. Thromb. Vasc. Biol. 34, 976–984 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Subramanian, M., Thorp, E. & Tabas, I. Identification of a non-growth factor role for GM-CSF in advanced atherosclerosis: promotion of macrophage apoptosis and plaque necrosis through IL-23 signaling. Circ. Res. 116, e13–e24 (2015).

    Article  CAS  PubMed  Google Scholar 

  39. Ingersoll, M. A. et al. Comparison of gene expression profiles between human and mouse monocyte subsets. Blood 115, e10–e19 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Fernandez, D. M. et al. Single-cell immune landscape of human atherosclerotic plaques. Nat. Med. 25, 1576–1588 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. 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 

  42. 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 

  43. Ridker, P. M. et al. Low-dose methotrexate for the prevention of atherosclerotic events. N. Engl. J. Med. 380, 752–762 (2019).

    Article  CAS  PubMed  Google Scholar 

  44. Zernecke, A. et al. Meta-analysis of leukocyte diversity in atherosclerotic mouse aortas. Circ. Res. 127, 402–426 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Wiesmann, F. et al. Developmental changes of cardiac function and mass assessed with MRI in neonatal, juvenile and adult mice. Am. J. Physiol. Heart Circ. Physiol. 278, H652–H657 (2000).

    Article  CAS  PubMed  Google Scholar 

  46. Wagenseil, J. E. & Mecham, R. P. Vascular extracellular matrix and arterial mechanics. Physiol. Rev. 89, 957–989 (2009).

    Article  CAS  PubMed  Google Scholar 

  47. Mackarehtschian, K. et al. Targeted disruption of the flk2/flt3 gene leads to deficiencies in primitive hematopoietic progenitors. Immunity 3, 147–161 (1995).

    Article  CAS  PubMed  Google Scholar 

  48. Benz, C., Martins, V. C., Radtke, F. & Bleul, C. C. The stream of precursors that colonizes the thymus proceeds selectively through the early T lineage precursor stage of T cell development. J. Exp. Med. 205, 1187–1199 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Ohta, T. et al. Crucial roles of XCR1-expressing dendritic cells and the XCR1-XCL1 chemokine axis in intestinal immune homeostasis. Sci. Rep. 6, 23505 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Stuart, T. et al. Comprehensive integration of single-cell data. Cell 177, 1888–1902 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Finak, G. et al. MAST: a flexible statistical framework for assessing transcriptional changes and characterizing heterogeneity in single-cell RNA sequencing data. Genome Biol. 16, 278 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  52. Hafemeister, C. & Satija, R. Normalization and variance stabilization of single-cell RNA-seq data using regularized negative binomial regression. Genome Biol. 20, 296 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Linderman, G. C., Rachh, M., Hoskins, J. G., Steinerberger, S. & Kluger, Y. Fast interpolation-based t-SNE for improved visualization of single-cell RNA-seq data. Nat. Methods 16, 243–245 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Williams, J. W. et al. Thermoneutrality but not UCP1 deficiency suppresses monocyte mobilization into blood. Circ. Res. 121, 662–676 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Flt3Cre mice were provided by D. Mann (Washington University School of Medicine; WUSM); Flt3−/−, Flt3l−/−, SNZ22GFP, L-MycGFP and Zbtb46GFP mice were provided by K. Murphy (WUSM); IL-34−/− and CCR2GFP mice were provided by M. Colonna (WUSM); op/op mice were provided by E. Unanue (WUSM); and Csf2rb−/− mice were provided B. Edelson (WUSM). We also thank M. Vail (University of Minnesota), the WUSM Flow Cytometry Core Facility, WUSM McDonnell Genome Institute and WUSM Genome Technology Access Center for technical assistance on this study. Research was supported by the National Institutes of Health (NIH) R00 HL138163 (to J.W.W.), AHA 16SDGG30480008 (to B.H.Z.), T32 AI007313 (to C.G.T.), P01 AI35296 (to B.T.F.) and NIH R37 AI049653 and DP1DK109668 (to G.J.R.). J.W.W. was supported by NIH 2T32DK007120-41 and AHA 17POST33410473. J-H.C. was supported by the Korean Health Technology R&D project HI15C0399 and Ministry of Health, Welfare & Family Affairs (South Korea). K.Z. was supported by the Government of Russian Federation (grant no. 08-08).

Author information

Authors and Affiliations

Authors

Contributions

This project was conceived and designed by J.W.W., M.N.A., B.H.Z., J-H.C. and G.J.R. Samples and materials were provided by J.W.W., B.T.F., B.H.Z., J-H.C. and G.J.R. Experiments were performed and analyzed by J.W.W., K.Z., K-W.K., S.I., B.T.S., P.R.S., K.K., A.E., S.H.K., C.G.T., M.W., S.E., K.J.L., B.H.Z. and J-H.C. All authors contributed to the preparation of this manuscript.

Corresponding author

Correspondence to Jesse W. Williams.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Jamie D. K. Wilson 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 Profiling aorta macrophage populations by bulk RNA-seq.

Bulk sorted MacAIR from C57BL/6 mice and adventitia, intima non-foamy, and intima foamy macrophages from 26-week HFD fed ApoE−/− mice were profiled for gene expression by RNA-seq19. Gene groupings for (a) macrophage and DC genes, (b) foamy and adventitia macrophage genes, (c) MacAIR enriched genes, and (d) genes associated with chemokine signaling in myeloid cells. Data are mean expression values derived from pooled macrophages from two biological replicates for MacAIR derived from 10-pooled aorta each, and three biological replicates each for ApoE−/− samples from 6-pooled aorta each.

Extended Data Fig. 2 MacAIR are detected across multiple scRNA-seq approaches.

(a) scRNA-seq data from C57BL/6 total CD45+ cells was integrated with two published studies where CD45+ aorta cells from chow-fed Ldlr−/− mice22 and atherosclerosis regression samples23, identifying 10 unique clusters. (b) Top 12 enriched MacAIR genes were used to define MacAIR cluster as cluster 4, presented as relative expression. (c) MacAIR were detectable in all three datasets with a relative abundance between 2.00–9.28%. (d) cDC1 genes (Zbtb46, Flt3, Itgae, Xcr1, Snx22, Mycl, and Rab43) were interrogated against the integrated dataset, defining a unique DC population. (e) Itgae and Xcr1 gene expression plotted on integrated t-SNE cluster map.

Extended Data Fig. 3 Differential gene expression between clusters of integrated dataset of C57BL/6, Ldlr−/−, and regression scRNA-seq experiments.

Top differentially expressed genes are listed with expression level across the 10 unique groups identified by unsupervised clustering of the integrated data of C57BL/6, chow fed Ldlr-/−22, and regression23 scRNA-seq datasets.

Extended Data Fig. 4 Profiling myeloid cells in the aorta.

Spleen and aorta samples from (a) SNX22gfp and (b) L-Mycgfp reporter mice were assessed for presence of cDC1 in respective tissues, GFP (green) and Dapi (blue). In panel B aorta, sample was co-stained with CD45 (red) and MHC II (white) antibody. Aorta from XCR1-venus reporter mice were isolated and stained for MHC II (red) or CD45 (white) expression by antibody labeling, then imaged in whole-mount for presence of cDC1 in the (c) aortic arch intima, (d) adventitia, and (e) aortic valve. C57Bl/6 (WT) aorta was isolated and stained for CD103 (green), MHC II (red), and CD45 (white) by antibody labeling and imaged by confocal microscopy for cDC1 presence in the (f) intima of the aortic arch or (g) the adventitia of the aorta. Embryonic labeling was performed using CD115creER Rosa26-mTmG mice. Aorta were collected from adult animals and assessed for GFP (green) and Tomato (red) expression in mice labeled at (h) E11.5, and co-stained with CD45 (white) in mice labeled at (i) E14.5. Data are representative of (a) n = 3, (b) n = 3, (ce) n = 6 in two experiments, (f, g) n = 6 in two experiments, (h) n = 3, and (i) n = 5 in two experiments.

Extended Data Fig. 5 Atherosclerotic plaque development in the aorta of hypercholesterolemic mice following acute depletion of MacAIR.

(a) Following the schematic, CX3CR1creER CD115-stop-DTR mice were used to conditionally express DTR on MacAIR by gavage with tamoxifen, waiting three weeks to allow circulating cells to repopulation from DTR-negative progenitors. MacAIR were depleted by i.p. injection of diphtheria toxin and hypercholesterolemia induced by i.v. injection of AAV-PCSK9. Mice were started on HFD the following day. Mice were sacrificed and assessed for plaque area in the aortic arch by oil red o (ORO) staining. (b) Representative images of ORO staining and en face imaging. (c) Quantification of plaque area on the aortic arch. Data are from n = 7, Cntl and n = 4 for DTR-group and are the result of a single experiment, error bars represent SEM.

Extended Data Fig. 6 Fate-mapping MacAIR during atherosclerosis progression in bone marrow transplant model.

Recipient Ldlr−/− mice were lethally irradiated and reconstituted with CX3CR1creER Rosa26-lsl-Tomato bone marrow. Mice were rested for 8-weeks for full hematopoietic reconstitution, then treated with tamoxifen by gavage to induce Tomato expression in CX3CR1-expressing cells, including MacAIR. (a) Three weeks after tamoxifen labeling, MacAIR remained Tomato+ (red) positive. Mice were then fed HFD and assessed for plaque development at (b) 10 days and (c) 28 days. MacAIR can be observed by Tomato-expression, whereas recruited cells were stained with CD68 (green) antibody. Samples are all en face whole mount confocal images of the aortic arch. Images are representative of n = 3 for each time point.

Extended Data Fig. 7 Csf2rb−/− Ldlr−/− mice fed HFD for 12 days.

Csf2rb+/- Ldlr−/− or Csf2rb−/− Ldlr−/− mice were fed HFD for 12 days to induce atherosclerotic lesions in the aortic arch. Aorta were stained for MHC II (red) and imaged by tile scanning whole mount confocal microscopy for macrophage burden and morphologic changes associated with foamy cell development. Three representative images (n = 7 for Csf2rb+/- Ldlr−/−) or (n = 4 Csf2rb−/− Ldlr−/−) were selected from a single experiment.

Extended Data Fig. 8 Differential gene expression analysis of scRNA-seq of CD45+ cells from Ldlr−/− aorta after 21-days HFD feeding.

The top 10 enriched genes for each cluster are reported for scRNA-seq analysis of total CD45+ cells from Ldlr−/− mice fed HFD for 21 days.

Extended Data Fig. 9 Integrated scRNA-seq analysis across atherosclerosis progression.

(a) scRNA-seq data from sorted CD45+ aorta cells isolated from C57Bl/6, 21-day HFD Ldlr−/−, and 12-week HFD Ldlr−/− mice was integrated and clustered into 16 populations. (b) All clusters were represented across unique time points. (c) MacAIR geneset (Mmp12, Il1b, Lgals3, Nes, Rgs1, Acp5, Asb2, Itgax, Cadm1, Gngt2, Cd9, Bcl2a1a) expression analysis of integrated cluster map and across time points. (d) Foamy macrophage geneset (Fabp4, Ctsl, Atp6v0d2, Gpnmb, Fabp5, Htra1, Epb41l3, Pld3) expression analysis of integrated cluster map and across time points. (e) Human foamy macrophage geneset (Gpx1, Pfdn5, Tpt1, Eef1a1, Ctb, B2m, Tmsb4x, Fth1, Ftl, Tmsb10, Cd63, Lgals1, Fcer1g, Npc2, Serf2, Ybx1, Psap, Apoc1, Apoe, Cstb, Ctsb, Vim, RnaseI, Fabp5, Plin2, Ccl2) expression analysis in the murine integrated scRNA-seq cluster map, suggesting shared gene expression programs between human and murine foamy macrophage subsets.

Extended Data Fig. 10 Differential gene expression from integrated scRNA-seq analysis across atherosclerosis time course.

scRNA-seq data from sorted CD45+ aorta cells isolated from C57Bl/6, 21-day HFD Ldlr−/−, and 12-week HFD Ldlr−/− mice were integrated and clustered into 16 populations. Top 10 differentially expressed genes are shown for each cluster.

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Williams, J.W., Zaitsev, K., Kim, KW. et al. Limited proliferation capacity of aortic intima resident macrophages requires monocyte recruitment for atherosclerotic plaque progression. Nat Immunol 21, 1194–1204 (2020). https://doi.org/10.1038/s41590-020-0768-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-020-0768-4

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