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:

Self-renewing resident cardiac macrophages limit adverse remodeling following myocardial infarction

A Publisher Correction to this article was published on 12 March 2019

This article has been updated

Abstract

Macrophages promote both injury and repair after myocardial infarction, but discriminating functions within mixed populations remains challenging. Here we used fate mapping, parabiosis and single-cell transcriptomics to demonstrate that at steady state, TIMD4+LYVE1+MHC-IIloCCR2 resident cardiac macrophages self-renew with negligible blood monocyte input. Monocytes partially replaced resident TIMD4LYVE1MHC-IIhiCCR2 macrophages and fully replaced TIMD4LYVE1MHC-IIhiCCR2+ macrophages, revealing a hierarchy of monocyte contribution to functionally distinct macrophage subsets. Ischemic injury reduced TIMD4+ and TIMD4 resident macrophage abundance, whereas CCR2+ monocyte-derived macrophages adopted multiple cell fates within infarcted tissue, including those nearly indistinguishable from resident macrophages. Recruited macrophages did not express TIMD4, highlighting the ability of TIMD4 to track a subset of resident macrophages in the absence of fate mapping. Despite this similarity, inducible depletion of resident macrophages using a Cx3cr1-based system led to impaired cardiac function and promoted adverse remodeling primarily within the peri-infarct zone, revealing a nonredundant, cardioprotective role of resident cardiac macrophages.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Fig. 1: Single-cell RNA sequencing reveals distinct cardiac macrophage and dendritic cell subsets at steady state.
Fig. 2: High-dimensional single-cell mapping and trajectory analysis reveals four macrophage clusters that occupy distinct activation states.
Fig. 3: TIMD4 and CCR2 are durable markers of resident and recruited macrophages.
Fig. 4: Resident macrophages are lost in infarcted cardiac tissue and return via local proliferation.
Fig. 5: High-dimensional analyses reveal plasticity of recruited macrophages to adopt both unique and resident-like transcriptional states within infarcted tissue.
Fig. 6: Selective depletion of resident cardiac macrophages results in impaired infarct healing and adverse cardiac remodeling primarily within the peri-infarct region.

Similar content being viewed by others

Data availability

Single-cell sequence data that support the findings of this study are available through the National Center for Biotechnology Information Gene Expression Omnibus (NCBI GEO) (GSE119355). The Agilent gene array data that support the findings of this study are available through the NCBI GEO (GSE119515).

Change history

  • 12 March 2019

    In the version of this article initially published, the equal contribution of the third author was omitted. The footnote links for that author should be “Sara Nejat1,11” and the correct statement is as follows: “11These authors contributed equally: Sarah A. Dick, Jillian A. Macklin, Sara Nejat.” The error has been corrected in the HTML and PDF versions of the article.

References

  1. Lozano, R. et al. Global and regional mortality from 235 causes of death for 20 age groups in 1990 and 2010: a systematic analysis for the Global Burden of Disease Study 2010. Lancet 380, 2095–2128 (2012).

    Article  Google Scholar 

  2. Ismahil, M. A. et al. Remodeling of the mononuclear phagocyte network underlies chronic inflammation and disease progression in heart failure: critical importance of the cardiosplenic axis. Circ. Res. 114, 266–282 (2014).

    Article  CAS  Google Scholar 

  3. Epelman, S., Lavine, K. J. & Randolph, G. J. Origin and functions of tissue macrophages. Immunity 41, 21–35 (2014).

    Article  CAS  Google Scholar 

  4. Schulz, C. et al. A lineage of myeloid cells independent of Myb and hematopoietic stem cells. Science 336, 86–90 (2012).

    Article  CAS  Google Scholar 

  5. 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  Google Scholar 

  6. Ginhoux, F. & Guilliams, M. Tissue-resident macrophage ontogeny and homeostasis. Immunity 44, 439–449 (2016).

    Article  CAS  Google Scholar 

  7. Porrello, E. R. et al. Transient regenerative potential of the neonatal mouse heart. Science 331, 1078–1080 (2011).

    Article  CAS  Google Scholar 

  8. Lavine, K. et al. Distinct macrophage lineages contribute to disparate patterns of cardiac recovery and remodeling in the neonatal and adult heart. Proc. Natl Acad. Sci. USA 111, 16029–16034 (2014).

    Article  CAS  Google Scholar 

  9. Aurora, A. B. et al. Macrophages are required for neonatal heart regeneration. J. Clin. Invest. 124, 1382–1392 (2014).

    Article  CAS  Google Scholar 

  10. Leid, J. M. et al. Primitive embryonic macrophages are required for coronary development and maturation. Circ. Res. 118, 1498–1511 (2016).

    Article  CAS  Google Scholar 

  11. Panizzi, P. et al. Impaired infarct healing in atherosclerotic mice with Ly-6Chi monocytosis. J. Am. Coll. Cardiol. 55, 1629–1638 (2010).

    Article  Google Scholar 

  12. van Amerongen, M. J., Harmsen, M. C., van Rooijen, N., Petersen, A. H. & van Luyn, M. J. Macrophage depletion impairs wound healing and increases left ventricular remodeling after myocardial injury in mice. Am. J. Pathol. 170, 818–829 (2007).

    Article  Google Scholar 

  13. Molawi, K. et al. Progressive replacement of embryo-derived cardiac macrophages with age. J. Exp. Med. 211, 2151–2158 (2014).

    Article  CAS  Google Scholar 

  14. 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  Google Scholar 

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

    Article  CAS  Google Scholar 

  16. Hulsmans, M. et al. Macrophages facilitate electrical conduction in the heart. Cell 169, 510–522 e520 (2017).

    Article  CAS  Google Scholar 

  17. Miyanishi, M. et al. Identification of Tim4 as a phosphatidylserine receptor. Nature 450, 435–439 (2007).

    Article  CAS  Google Scholar 

  18. Clemente-Casares, X. et al. A CD103+ conventional dendritic cell surveillance system prevents development of overt heart failure during subclinical viral myocarditis. Immunity 47, 974–989 (2017).

    Article  CAS  Google Scholar 

  19. Van der Borght, K. et al. Myocardial infarction primes autoreactive T cells through activation of dendritic cells. Cell Rep. 18, 3005–3017 (2017).

    Article  Google Scholar 

  20. 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  Google Scholar 

  21. Tirosh, I. et al. Dissecting the multicellular ecosystem of metastatic melanoma by single-cell RNA-seq. Science 352, 189–196 (2016).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  23. 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  Google Scholar 

  24. Trapnell, C. et al. The dynamics and regulators of cell fate decisions are revealed by pseudotemporal ordering of single cells. Nat. Biotechnol. 32, 381–386 (2014).

    Article  CAS  Google Scholar 

  25. Chen, J., Schlitzer, A., Chakarov, S., Ginhoux, F. & Poidinger, M. Mpath maps multi-branching single-cell trajectories revealing progenitor cell progression during development. Nat. Commun. 7, 11988 (2016).

    Article  CAS  Google Scholar 

  26. Parkhurst, C. N. et al. Microglia promote learning-dependent synapse formation through brain-derived neurotrophic factor. Cell 155, 1596–1609 (2013).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  28. Leuschner, F. et al. Rapid monocyte kinetics in acute myocardial infarction are sustained by extramedullary monocytopoiesis. J. Exp. Med. 209, 123–137 (2012).

    Article  CAS  Google Scholar 

  29. Leuschner, F. et al. Angiotensin-converting enzyme inhibition prevents the release of monocytes from their splenic reservoir in mice with myocardial infarction. Circ. Res. 107, 1364–1373 (2010).

    Article  CAS  Google Scholar 

  30. Nahrendorf, M. et al. The healing myocardium sequentially mobilizes two monocyte subsets with divergent and complementary functions. J. Exp. Med. 204, 3037–3047 (2007).

    Article  CAS  Google Scholar 

  31. Wan, E. et al. Enhanced efferocytosis of apoptotic cardiomyocytes through myeloid-epithelial-reproductive tyrosine kinase links acute inflammation resolution to cardiac repair after infarction. Circ. Res. 113, 1004–1012 (2013).

    Article  CAS  Google Scholar 

  32. 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 e327 (2018).

    Article  CAS  Google Scholar 

  33. Majmudar, M. D. et al. Monocyte-directed RNAi targeting CCR2 improves infarct healing in atherosclerosis-prone mice. Circulation 127, 2038–2046 (2013).

    Article  CAS  Google Scholar 

  34. Bajpai, G. et al. The human heart contains distinct macrophage subsets with divergent origins and functions. Nat. Med. 24, 1234–1245 (2018).

    Article  CAS  Google Scholar 

  35. Satoh, T. et al. Identification of an atypical monocyte and committed progenitor involved in fibrosis. Nature 541, 96–101 (2017).

    Article  CAS  Google Scholar 

  36. Menezes, S. et al. The heterogeneity of Ly6Chi monocytes controls their differentiation into iNOS+ macrophages or monocyte-derived dendritic cells. Immunity 45, 1205–1218 (2016).

    Article  CAS  Google Scholar 

  37. Ikeda, N. et al. Emergence of immunoregulatory Ym1(+)Ly6C(hi) monocytes during recovery phase of tissue injury. Sci. Immunol. 3, eaat0207 (2018).

  38. Mrdjen, D. et al. High-dimensional single-cell mapping of central nervous system immune cells reveals distinct myeloid subsets in health, aging, and disease. Immunity 48, 599 (2018).

    Article  CAS  Google Scholar 

  39. Han, X. et al. Mapping the mouse cell atlas by microwell-seq. Cell 172, 1091–1107 e1017 (2018).

    Article  CAS  Google Scholar 

  40. Mass, E. et al. Specification of tissue-resident macrophages during organogenesis. Science 353, aaf4238 (2016).

  41. van de Laar, L. et al. Yolk sac macrophages, fetal liver, and adult monocytes can colonize an empty niche and develop into functional tissue-resident macrophages. Immunity 44, 755–768 (2016).

    Article  Google Scholar 

  42. Gosselin, D. et al. An environment-dependent transcriptional network specifies human microglia identity. Science 356, eaal3222 (2017).

  43. Aronoff, L., Epelman, S. & X, C.-C. Isolation and identification of extravascular immune cells of the heart. J. Vis. Exp. 138, 7 (2018).

    Google Scholar 

  44. Noyan-Ashraf, M. H. et al. GLP-1R agonist liraglutide activates cytoprotective pathways and improves outcomes after experimental myocardial infarction in mice. Diabetes 58, 975–983 (2009).

    Article  CAS  Google Scholar 

  45. Dobin, A. et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21 (2013).

    Google Scholar 

  46. Butler, A., Hoffman, P., Smibert, P., Papalexi, E. & Satija, R. Integrating single-cell transcriptomic data across different conditions, technologies, and species. Nat. Biotechnol. 36, 411–420 (2018).

    Article  CAS  Google Scholar 

  47. Lun, A., McCarthy, D. & Marioni, J. A step-by-step workflow for low-level analysis of single-cell RNA-seq data with Bioconductor. F1000 Res. 5, 2122 (2016).

    Google Scholar 

  48. McCarthy, D., Campbell, K., Lun, A. & Wills, Q. Scater: pre-processing, quality control, normalization and visualization of single-cell RNA-seq data in R. Bioinformatics 33, 1179–1186 (2017).

    CAS  Google Scholar 

  49. Blondel, V. D., Guillaume, J. L., Lambiotte, R. & Lefebvre, E. Fast unfolding of communities in large networks. J. Stat. Mech. 10, 10008 (2008).

  50. 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  Google Scholar 

  51. Reimand, J. et al. g:Profiler—a web server for functional interpretation of gene lists (2016update). Nucleic Acids Res. 44, W83–W89 (2016).

    Article  CAS  Google Scholar 

  52. Qiu, X. et al. Reversed graph embedding resolves complex single-cell trajectories. Nat. Meth. 14, 979–982 (2017).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

This work was supported by the Canadian Institutes of Health Research (S.E., PJT364831; J.A.M. and S.A.D.), Heart and Stroke Foundation (S.E.), March of Dimes (S.E.), Ted Rogers Centre for Heart Research (S.E., S.A.D. and J.A.M.), the Peter Munk Cardiac Centre (S.E.) and the National Institutes of Health (S.E. K08HL112826). We thank D. Mann for his insight and advice, and S. Wilson for editorial assistance. Thanks to N. Winegarden and N. Khuu for help with processing single-cell RNA sequencing samples.

Author information

Authors and Affiliations

Authors

Contributions

S.A.D. and J.A.M. designed and performed experiments with the help of X.C-C., S.H., C.K., M.G.A., A.W., L.A., R.Z., R.B. and I.B. A.M. performed all surgeries. M.H., K.J.L., B.R., F.G., M.I.C. and C.S.R. provided expertise and feedback. S.N. performed the bioinformatics analyses. J.C. performed the Mpath analysis. S.E. conceived the study, obtained funding and wrote the manuscript with S.A.D., S.N. and J.A.M.

Corresponding author

Correspondence to Slava Epelman.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

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

Integrated supplementary information

Supplementary Figure 1 Gene-expression analysis of cardiac macrophage clusters defined by single-cell RNA sequencing reveals five distinct populations.

a) Cardiac mononuclear phagocytes were sorted from adult c57bl/6j mice (~20 weeks old). Total macrophages (MFs) (R2; CD45+CD64lo–hiCD11b+) and total dendritic cells (DCs) (R3; CD45+CD64CD11c+MHC-IIint–hi). The two sorted populations were pooled in a 1:1 ratio before processing for scRNA-seq on the 10x Genomics Chromium platform. N=8 (4 male; 4 female); one experiment. b) The top ~65 differentially expressed genes in each cluster were input into the ImmGEN online portal for "My Geneset" http://rstats.immgen.org/MyGeneSet_New/index.html to generate a similarity score. We included all MF, monocyte and dendritic cell populations in the V1 array database for the assessment to generate a means-normalized expression scatter plot. Colored boxes highlight similarity in dendritic cell (light green), macrophage (dark green) and monocyte (red) populations to the sorted populations contributed to ImmGen. c) Violin plots of cluster defining genes in the 11 clusters defined in Fig. 1c. *p<0.01 (adjusted P value). Statistical analysis of scRNA-seq is defined in the material and methods, and was performed using the MAST method.

Supplementary Figure 2 Cx3cr1CreER:R26Td mice retain the ability to distinguish resident macrophages from recruited macrophages following irradiation and adoptive transplant.

a) Cx3cr1CreER–YFP:R26Td mice were used to determine Td reporter expression in cardiac macrophages (CD45+CD64+CD11b+), brain microglia (CD45lowCD64+CD11b+), liver Kupffer cells (CD45+CD64+CD11b+) and blood monocytes (CD115+CD11b+) in the absence of tamoxifen administration (No TAM) at the indicated times (from E17.5 to 77 days post-natal). Recombination in tissue macrophages without TAM was compared to pregnant mice that were given a TAM pulse at E18.5 in utero (via i.p. injection of 2mg TAM). N=3; center value, mean; error bars, SEM; one representative experiment, repeated twice. b) A second fate-mapping system was also used (Cx3cr1CreERT2)30 and bred to the same reporter R26Td line to assess for Td expression in cardiac macrophages (heart) and microglia (brain) at the indicated times either without TAM (No TAM) or following TAM administration at E18.5 (+TAM). N=3; center value, mean; one experiment. c) Bone marrow from adult Cx3cr1CreER–YFP:R26Td mice (Donor BM, CD45.2), not previously pulsed with tamoxifen, was isolated and the %Td+ macrophages was determined in blood CD115+CD11b+ monocytes and total CD45+ bone marrow. Donor Bone marrow was transplanted into non-lethal irradiated CD45.1 mice (Recipients). After 6 weeks, chimerism rates in the recipient were ~95%. Blood and cardiac tissue was isolated from the CD45.1 recipient. A subset of recipient mice was given a pulse of tamoxifen (TAM) (i.p. 2mg) 2 days prior to isolation to confirm the responsiveness of chimeric CD45.2 cells in the recipient compared to without TAM administration (No TAM). The %Td+ cells was determined in total CD45.2+ blood monocytes (CD115+CD11b+) and total CD45.2+ cardiac macrophages (MFs) (CD64+CD11b+). N=3,2; center value, mean; error bars, SEM; one experiment.

Supplementary Figure 3 Tracking resident cardiac macrophages post myocardial infarction.

a-f) 3-week-old Cx3cr1CreER–YFP:R26Td mice were fed tamoxifen (TAM)- containing chow for 10 days, and then mice were either left as non-infarcted controls for an additional 6 weeks before a myocardial infarction was performed (MI). a) Ischemic tissue was isolated at Day 2 and Day 28 post-MI and total resident cardiac macrophages (CD45+CD64+CD11b+Td+) were analyzed as TIMD4+ and TIMD4 subsets per mg/tissue compared to non-infarcted controls (No MI). N=3; repeated independently 2 times with similar results. b) Total CD45+CD64+CD11b+ cardiac macrophages (MFs) were partitioned into TIMD4+, CCR2+ and TIMD4CCR2 MF subsets. LYVE1 was detected by flow cytometry. N=3; repeated independently 2 times with similar results. c) Representative images of hearts isolated, fixed and sectioned longitudinally for immunofluorescence from Cx3cr1CreER–YFP:R26Td mice post-MI. Td expression was induced as in panel (a) above. Cardiac macrophages (CD68+, white) were counted based on their coexpression for the residency marker Td (red) and LYVE1 (green) in the infarct and peri-infarct regions, defined in Fig. 4a, from 3 individual images/region/mouse. N=3; one experiment. d) Hearts were isolated from the ischemic zone of Cx3cr1CreER–YFP:R26Td/DTR mice day 4 post-MI after a 2 hr-BrdU pulse. Total Td+ cardiac macrophages were subdivided into TIMD4+ and TIMD4 MF populations and %BrdU+ macrophages was quantified. N=3,3,4,4; one experiment. e) Heat map of the 20 most differentially expressed genes from clusters from Fig. 5b. Overlapping clusters (#1 to #6a), represent macrophages from control and post-MI samples that clustered together. For the overlapping clusters both the control and MI cells from each cluster are aligned next to each other in the heatmap. f) Means-normalized expression scatter plot produced by comparison of the differentially expressed genes from each unique cluster (7, 8, 9, 10 and 11) in Fig. 5b to the ImmGen data set. For all: *p<0.05, **p<0.01, ***p<0.001, ****p<0.0001; two-tailed Student’s T test; center value, mean; error bars, SEM.

Supplementary Figure 4 Chimeric Cx3cr1CreER–YFP:R26Td monocytes retain the ability to distinguish resident macrophages from recruited macrophages post myocardial infarction in the setting of parabiosis and shielded irradiation.

a) Cx3cr1CreER–YFP:R26Td mice (Donor CD45.2) were fed tamoxifen (TAM)-containing chow at 3 weeks of age for 1 week, TAM was then discontinued for additional 10 weeks. At this time, Cx3cr1CreER–YFP:R26Td mice were paired with recipient CD45.1 mice. Two weeks after pairing, recipient CD45.1 mice were infarcted by surgical ligation of the left anterior descending artery (MI). At day 5 post-MI, blood and cardiac tissue (ischemic zone) was quantified for %Td+ in chimeric CD45.2 macrophages (MFs) (CD45+CD64+CD11b+) and blood monocytes (CD115+CD11b+) in the CD45.1 recipient mouse. N=3. One experiment. b) Bone marrow was isolated from adult Cx3cr1CreER–YFP:R26Td mice (Donor BM, CD45.2) treated as in panel (a). The bone marrow cells were transplanted into non-lethally irradiated CD45.1 mice (Recipient) that had a lead shield covering the heart and thoracic cavity to minimize loss of resident cardiac macrophages. Following 6 weeks of reconstitution, surgical MI was performed on the recipient CD45.1 mouse and it recovered for an additional 4 weeks. Blood and cardiac tissue (separated into ischemic and remote zone, see Fig. 4a) was taken for flow cytometric analysis. A subset of mice was given a pulse of TAM (i.p.) 2 days prior to isolation to confirm the responsiveness of the CD45.2 cells to activate the TAM-inducible Td reporter compared to No TAM controls. Gated on total CD45.2+ blood monocytes (CD115+CD11b+) and total CD45.2+ cardiac macrophages (CD64+CD11b+) in the CD45.1 recipient and %Td+ was quantified. N=3; one experiment. For all: center value, mean; error bars, SEM.

Supplementary Figure 5 Similar cardiac macrophage subsets exist in human cardiomyopathy.

Left ventricular cardiac tissue was obtained from human patients with end-stage cardiomyopathy during implantation of left ventricular assist devices. a) Tissue was digested and analyzed by flow cytometry [gated similar to mouse (CD45+CD64+MerTK+CD14+)] or b) sorted for Hema-3 staining analysis and visualization. c) Cardiac macrophage (MF) populations were sorted from human tissue based on MHC-II and CCR2 expression. RNA was extracted, amplified, and gene expression was determined by gene array (Agilent). Genes that were most differentially expressed in the 5 Seurat MF clusters identified by scRNA-seq in the mouse (Fig. 1d) were compared to the gene expression across the three human MF subsets isolated to generate heatmaps. N=6 patient samples; one experiment.

Supplementary Figure 6 Resident cardiac macrophages proliferate post myocardial infarction in the remote myocardium, with resident macrophage depletion leading to increased cardiomyocyte hypertrophy without cardiac fibrosis.

3-week-old Cx3cr1CreER–YFP:R26Td mice were fed tamoxifen (TAM)- containing chow for 10 days, and then mice were either left as non-infarcted controls for an additional 6 weeks. a) Diphtheria toxin (DT) was then administered to Cx3cr1CreER–YFP:R26Td/DTR mice for 2 weeks at steady state. Blood monocytes (CD115+CD11b+Ly6C+), neutrophils (CD45+CD11b+CD64Ly6G+), lymphocytes (CD45+CD11b+CD64), TdCCR2+ cardiac macrophages, and Td+ cardiac macrophages were quantified by flow cytometry, normalized to DT-injected Cx3cr1+/+:R26Td/DTR control mice. N=5; repeated independently 3 times with similar results. b) Representative flow plots of resident macrophage depletion during steady state (Fig. 6a) from blood and cardiac tissue following a myocardial infarction demonstrating effective depletion of resident macrophages. c) Naive wild-type mice were given chronic PBS or 250ng DT injections (i.p.) for 35 days. Whole hearts were either cut longitudinally and sectioned to be stained with Fast Green for interstitial fibrosis, quantified as % Fibrosis using ImageJ software, or quantified by flow cytometry for the number of neutrophils or cardiac macrophages normalized per mg tissue. N=3, 7; one experiment. d) Hearts were isolated from adult TAM-fed Cx3cr1CreER–YFP:R26Td/DTR mice (as above) day 5 post-MI, comparing non-depleted or depleted mice (DT treated daily). Recruited macrophages (MF), monocytes, and neutrophils were quantified and normalized per mg tissue in the ischemic and remote cardiac zones. N=6,5,4; repeated independently 2 times with similar results. e-f) Hearts were isolated from adult Cx3cr1CreER–YFP:R26Td mice following TAM-induction (as above), and mice received BrdU injection 2 hours prior to harvest. Resident MFs (Td+), recruited MFs (Td) and monocytes were normalized per mg tissue in the remote zone at various time points (e), or %BrdU+ was determined (f), compared to an uninfarcted control (No MI). N(Td+)=13, 13, 6, 3, 10. N(Td, mono)= 12, 10, 7, 3, 6; Repeated 3 times for day 2, 2 times for day 4 and 28, 1 time for day 7, 6 times for control with similar results. g) Change in fractional shortening (contractile function) from day 7 to day 28 post-MI in the remote zone comparing mice with or without chronic resident macrophage depletion (Paired T-test) through the use of M-mode echocardiography. N=11; repeated 2 times independently with similar results. h-i) Longitudinally cut hearts from Cx3cr1CreER–YFP:R26Td/DTR mice (as above) isolated day 35 post-MI to compare post-MI control and depleted groups. Cardiomyocyte hypertrophy was detected using Wheat Germ Agglutinin staining in the remote zone. Hearts imaged at 20x magnification and cardiomyocytes measured in ImageJ, 8 images/zone/heart at 2 cutting levels. N=4, 6, 6; repeated once with similar results (h); Cardiac tissue fibrosis was detected using Fast Green staining. Percentage fibrotic area was quantified with ImageJ analysis, 4 images/zone/heart at 2 cutting levels. N = 9; repeated once with similar results. For all: *p<0.05, **p<0.01, ***p<0.001, ****p<0.0001; Two-tailed Student’s T test; center value, mean; error bars, SEM.

Supplementary information

Supplementary Figures 1-6

Supplementary Figures 1-6 Supplementary Table 1

Reporting Summary

Supplementary Table 2

DEgenes For Supplementary Figure 3e

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Dick, S.A., Macklin, J.A., Nejat, S. et al. Self-renewing resident cardiac macrophages limit adverse remodeling following myocardial infarction. Nat Immunol 20, 29–39 (2019). https://doi.org/10.1038/s41590-018-0272-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-018-0272-2

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