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:

Induction of the nuclear receptor PPAR-γ by the cytokine GM-CSF is critical for the differentiation of fetal monocytes into alveolar macrophages

Abstract

Tissue-resident macrophages constitute heterogeneous populations with unique functions and distinct gene-expression signatures. While it has been established that they originate mostly from embryonic progenitor cells, the signals that induce a characteristic tissue-specific differentiation program remain unknown. We found that the nuclear receptor PPAR-γ determined the perinatal differentiation and identity of alveolar macrophages (AMs). In contrast, PPAR-γ was dispensable for the development of macrophages located in the peritoneum, liver, brain, heart, kidneys, intestine and fat. Transcriptome analysis of the precursors of AMs from newborn mice showed that PPAR-γ conferred a unique signature, including several transcription factors and genes associated with the differentiation and function of AMs. Expression of PPAR-γ in fetal lung monocytes was dependent on the cytokine GM-CSF. Therefore, GM-CSF has a lung-specific role in the perinatal development of AMs through the induction of PPAR-γ in fetal monocytes.

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

Figure 1: Impaired development and function of AMs in Cd11c-CrePpargfl/fl mice.
Figure 2: AMs develop perinatally from fetal monocytes through a pre-AM intermediate that requires PPAR-γ for terminal differentiation.
Figure 3: Cell-autonomous requirement for PPAR-γ during the terminal differentiation of AMs.
Figure 4: PPAR-γ confers the AM signature and identity.
Figure 5: Diminished lipid catabolism–associated gene expression and enhanced cholesterol esterification in PPAR-γ-deficient AMs.
Figure 6: GM-CSF drives the development of AMs from fetal monocytes by the induction of PPAR-γ.
Figure 7: Prenatal PPAR-γ is essential for the development of AMs.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

Referenced accessions

Gene Expression Omnibus

References

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

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  3. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Ginhoux, F. & Jung, S. Monocytes and macrophages: developmental pathways and tissue homeostasis. Nat. Rev. Immunol. 14, 392–404 (2014).

    CAS  PubMed  Google Scholar 

  6. Ginhoux, F. et al. Fate mapping analysis reveals that adult microglia derive from primitive macrophages. Science 330, 841–845 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Hoeffel, G. et al. Adult Langerhans cells derive predominantly from embryonic fetal liver monocytes with a minor contribution of yolk sac-derived macrophages. J. Exp. Med. 209, 1167–1181 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Guilliams, M. et al. Alveolar macrophages develop from fetal monocytes that differentiate into long-lived cells in the first week of life via GM-CSF. J. Exp. Med. 10, 1977–1992 (2013).

    Google Scholar 

  9. 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 

  10. 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 

  11. Tontonoz, P. & Spiegelman, B.M. Fat and beyond: the diverse biology of PPARγ. Annu. Rev. Biochem. 77, 289–312 (2008).

    CAS  PubMed  Google Scholar 

  12. Tontonoz, P., Nagy, L., Alvarez, J.G., Thomazy, V.A. & Evans, R.M. PPARγ promotes monocyte/macrophage differentiation and uptake of oxidized LDL. Cell 93, 241–252 (1998).

    CAS  PubMed  Google Scholar 

  13. Ricote, M. et al. Expression of the peroxisome proliferator-activated receptor γ (PPARγ) in human atherosclerosis and regulation in macrophages by colony stimulating factors and oxidized low density lipoprotein. Proc. Natl. Acad. Sci. USA 95, 7614–7619 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Ricote, M., Li, A.C., Willson, T.M., Kelly, C.J. & Glass, C.K. The peroxisome proliferator-activated receptor-γ is a negative regulator of macrophage activation. Nature 391, 79–82 (1998).

    CAS  PubMed  Google Scholar 

  15. Jiang, C., Ting, A.T. & Seed, B. PPAR-γ agonists inhibit production of monocyte inflammatory cytokines. Nature 391, 82–86 (1998).

    CAS  PubMed  Google Scholar 

  16. Szanto, A. et al. STAT6 transcription factor is a facilitator of the nuclear receptor PPARγ-regulated gene expression in macrophages and dendritic cells. Immunity 33, 699–712 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Bonfield, T.L. et al. Peroxisome proliferator-activated receptor-γ is deficient in alveolar macrophages from patients with alveolar proteinosis. Am. J. Respir. Cell Mol. Biol. 29, 677–682 (2003).

    CAS  PubMed  Google Scholar 

  18. Gautier, E.L. et al. Systemic analysis of PPARγ in mouse macrophage populations reveals marked diversity in expression with critical roles in resolution of inflammation and airway immunity. J. Immunol. 189, 2614–2624 (2012).

    CAS  PubMed  Google Scholar 

  19. Bonfield, T.L. et al. Peroxisome proliferator-activated receptor-γ regulates the expression of alveolar macrophage macrophage colony-stimulating factor. J. Immunol. 181, 235–242 (2008).

    CAS  PubMed  Google Scholar 

  20. Imai, T. et al. Peroxisome proliferator-activated receptor γ is required in mature white and brown adipocytes for their survival in the mouse. Proc. Natl. Acad. Sci. USA 101, 4543–4547 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Vermaelen, K. & Pauwels, R. Accurate and simple discrimination of mouse pulmonary dendritic cell and macrophage populations by flow cytometry: methodology and new insights. Cytometry 61, 170–177 (2004).

    PubMed  Google Scholar 

  22. Moore, K.J. & Tabas, I. Macrophages in the pathogenesis of atherosclerosis. Cell 145, 341–355 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Hochreiter-Hufford, A. & Ravichandran, K.S. Clearing the dead: apoptotic cell sensing, recognition, engulfment, and digestion. Cold Spring Harb. Perspect. Biol. 5, a008748 (2013).

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

    CAS  PubMed  Google Scholar 

  25. O'Neill, L.A.J. & Hardie, D.G. Metabolism of inflammation limited by AMPK and pseudo-starvation. Nature 493, 346–355 (2013).

    CAS  PubMed  Google Scholar 

  26. Schneider, C. et al. Alveolar macrophages are essential for protection from respiratory failure and associated morbidity following influenza virus infection. PLoS Pathog. 10, e1004053 (2014).

    PubMed  PubMed Central  Google Scholar 

  27. de Boer, J. et al. Transgenic mice with hematopoietic and lymphoid specific expression of Cre. Eur. J. Immunol. 33, 314–325 (2003).

    CAS  PubMed  Google Scholar 

  28. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Tall, A.R., Yvan-Charvet, L., Terasaka, N., Pagler, T. & Wang, N. HDL, ABC transporters, and cholesterol efflux: implications for the treatment of atherosclerosis. Cell Metab. 7, 365–375 (2008).

    CAS  PubMed  Google Scholar 

  31. Johnson, J.L. & Newby, A.C. Macrophage heterogeneity in atherosclerotic plaques. Curr. Opin. Lipidol. 20, 370–378 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Mallat, Z., Lambeau, G. & Tedgui, A. Lipoprotein-associated and secreted phospholipases A in cardiovascular disease: roles as biological effectors and biomarkers. Circulation 122, 2183–2200 (2010).

    PubMed  Google Scholar 

  33. Han, C.Z. & Ravichandran, K.S. Metabolic connections during apoptotic cell engulfment. Cell 147, 1442–1445 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Jakubzick, C. et al. Minimal differentiation of classical monocytes as they survey steady-state tissues and transport antigen to lymph nodes. Immunity 39, 599–610 (2013).

    CAS  PubMed  Google Scholar 

  35. Malur, A. et al. Deletion of PPAR γ in alveolar macrophages is associated with a Th-1 pulmonary inflammatory response. J. Immunol. 182, 5816–5822 (2009).

    CAS  PubMed  Google Scholar 

  36. Baker, A.D. et al. PPARγ regulates the expression of cholesterol metabolism genes in alveolar macrophages. Biochem. Biophys. Res. Commun. 393, 682–687 (2010).

    CAS  PubMed  Google Scholar 

  37. Kohyama, M. et al. Role for Spi-C in the development of red pulp macrophages and splenic iron homeostasis. Nature 457, 318–321 (2009).

    CAS  PubMed  Google Scholar 

  38. A-Gonzalez, N. et al. The nuclear receptor LXRα controls the functional specialization of splenic macrophages. Nat. Immunol. 14, 831–839 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Nakamura, A. et al. Transcription repressor Bach2 is required for pulmonary surfactant homeostasis and alveolar macrophage function. J. Exp. Med. 210, 2191–2204 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Okabe, Y. & Medzhitov, R. Tissue-specific signals control reversible program of localization and functional polarization of macrophages. Cell 157, 832–844 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Clausen, B.E., Burkhardt, C., Reith, W., Renkawitz, R. & Förster, I. Conditional gene targeting in macrophages and granulocytes using LysMcre mice. Transgenic Res. 8, 265–277 (1999).

    CAS  PubMed  Google Scholar 

  42. Caton, M.L., Smith-Raska, M.R. & Reizis, B. Notch-RBP-J signaling controls the homeostasis of CD8- dendritic cells in the spleen. J. Exp. Med. 204, 1653–1664 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Luche, H., Weber, O., Nageswara Rao, T., Blum, C. & Fehling, H.J. Faithful activation of an extra-bright red fluorescent protein in “knock-in” Cre-reporter mice ideally suited for lineage tracing studies. Eur. J. Immunol. 37, 43–53 (2007).

    CAS  PubMed  Google Scholar 

  44. Saeed, A.I. et al. TM4: a free, open-source system for microarray data management and analysis. Biotechniques 34, 374–378 (2003).

    CAS  PubMed  Google Scholar 

  45. Saeed, A.I. et al. TM4 microarray software suite. Methods Enzymol. 411, 134–193 (2006).

    CAS  PubMed  Google Scholar 

  46. Irizarry, R.A. Summaries of Affymetrix GeneChip probe level data. Nucleic Acids Res. 31, e15 (2003).

    PubMed  PubMed Central  Google Scholar 

  47. Thiele, C. et al. Tracing fatty acid metabolism by click chemistry. ACS Chem. Biol. 7, 2004–2011 (2012).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank P. Chambon (Université Louis Pasteur) for Ppargfl/fl mice20; B. Becher (University of Zurich) for Csf2−/− and Csf2rb−/− mice; and C. Halin (Swiss Federal Institute of Technology Zurich) for Rosa26-stopflox-tdRFP mice43. Supported by the Swiss National Science Foundation (310030-124922/1) and Swiss Federal Institute of Technology Zurich (ETH-34 13-1).

Author information

Authors and Affiliations

Authors

Contributions

C.S. and M.Ko. designed the experiments; C.S. performed and analyzed most of the experiments; S.P.N., M.Ku., H.R. and C.T. performed and analyzed specific experiments; and C.S. and M.Ko. wrote the manuscript.

Corresponding author

Correspondence to Manfred Kopf.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 PPAR-γ is dispensable for the development of tissue macrophages in the heart, kidneys, lamina propria and white adipose tissue.

Plots show the expression of F4/80 and CD11b (a) or F4/80 and CD11c (b) among CD45+ cells in the indicated organs (LP, lamina propria). Bar graphs display the percentage of cells gated as shown in flow cytometry plots. Data are from one experiment representative of two independent experiments (mean and s.d. of three mice per group).

Supplementary Figure 2 Cd11c-CrePpargfl/fl mice with functionally impaired AMs accumulate apoptotic cells in the bronchoalveolar space.

(a) CD11chiCD11bloautofluorescencehiSiglec-F+ Ppargfl/fl AM and CD11chiCD11bhiautofluorescencehiSiglec-F+ arrested immature AM from Cd11cCrePpargfl/fl mice were sorted by flow cytometry followed by cytospin and Oil Red O staining. Micrographs were taken at 20× magnification. Scale bar = 50 μm. (b) BAL of Ppargfl/fl, LysmCrePpargfl/fl and Cd11cCrePpargfl/fl mice was analyzed by flow cytometry for the presence of dead eFluor780+ cells. Representative pictures and plots of three to four mice per group are shown.

Supplementary Figure 3 Cell-autonomous requirement for PPAR-γ during AM development.

Mixed BM chimeras (1:4 mixture of CD45.1+WT:CD45.2+Cd11cCrePpargfl/fl or CD45.1+WT:CD45.2+Ppargfl/fl) were analyzed as in Fig. 3. (a) Reconstitution ratio in peripheral blood leukocyte populations shown as CD45.2+ fold over CD45.1+ cells. (b-d) Histograms show the levels of CD11b and Siglec-F in CD45.1+WT and CD45.2+Ppargfl/fl or CD45.2+Cd11cCrePpargfl/fl AM in the BAL of the same mouse (b) and the degree of CD11c expression and autofluorescence in the BAL (c) and lung (d). (e) Bar graphs display the frequencies of eF780+ apoptotic AM among CD45.2+Ppargfl/fl and CD45.2+Cd11cCrePpargfl/fl cells and their CD45.1+ WT counterparts. Data are from one experiment representative of two independent experiments (mean and s.d. of four chimeras per group, dot plots from one mouse representative of the group).

Supplementary Figure 4 PPAR-γ is required for the maintenance of AM identity.

Transcriptomes of Ppargfl/fl AM and arrested immature Cd11cCrePpargfl/fl AM sorted from adult mouse lungs by flow cytometry were analysed by microarray. (a,b) Heat maps representing mRNA levels in Ppargfl/fl and Cd11cCrePpargfl/fl AM of peritoneal macrophage signature-up-genes (a) and transcription factors (b). (c,d) Heat maps representing mRNA levels in Ppargfl/fl and Cd11cCrePpargfl/fl AM of microglia signature-up-genes (c) and transcription factors (d). The list of signature transcripts was obtained from Reference.

Supplementary Figure 5 PPAR-γ is required for the induction of an AM-specific gene-expression profile and maintenance of AM identity.

Transcriptomes of Ppargfl/fl AM and immature arrested Cd11cCrePpargfl/fl AM sorted from 11 days old and adult mice and of pre-AM from DAB2 were analysed by microarray. Bar graphs show relative expression levels plotted as log2-fold change in Cd11cCrePpargfl/fl cells compared to Ppargfl/fl. Effects of Pparg-deficiency on genes involved in phagocytosis of apoptotic cells (a), cytokines and modulators of inflammation (b), chemokines and chemokine receptors (c) and tissue remodeling factors (d).

Supplementary Figure 6 PPAR-γ is dispensable for the development and maintenance of most tissue macrophages.

(a) Fetal monocytes were sorted from lungs of the indicated strains on E17.5 and recombination of the Ppargfl/fl alleles was assessed by quantitative real-time PCR on genomic DNA. (b,c) E17.5 WT and Vav1CrePpargfl/fl fetuses were analyzed for the presence of fetal monocytes in the blood (b) and the liver (c). (d) Adult WT and Vav1CrePpargfl/fl mice were analyzed for the presence of macrophages in the indicated organs. Numbers represent the frequencies among total cells (blood), CD11b+CD19 (peritoneum), eF780CD45+ (brain, liver, perigonadal white adipose tissue (WAT)), eF780CD45+CD64+ (kidney), eF780CD45+CD64+autofluorescence+ (heart) or eF780CD45+CD11b+ cells (small intestine lamina propria (LP)). (e) Macrophages were sorted as gated in (d), from the indicated organs of adult WT and Vav1CrePpargfl/fl mice and recombination of the Ppargfl/fl alleles was assessed by quantitative real-time PCR on genomic DNA. Subsets of blood monocytes and peritoneal Mø subsets were pooled, respectively. NS, not significant (Student's t-test). Data are from one experiment (a; mean and s.d. of three to five mice per group), from one experiment representative of two independent experiments (b,c; dot plots of one mouse per group representative of three mice per group), from one experiment representative of two independent experiments (d; dot plots of one mouse per group representative of five mice per group) or from one experiment (e; mean and s.d. of four mice per group).

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–6 and Supplementary Table 1 (PDF 1854 kb)

Enrichment analysis in pathway maps for differentially expressed genes.

Enrichment analysis was performed using MetaCore. List of 100 pathways enriched in differentially expressed genes are displayed for upregulated and downregulated genes (log2 ratio ≥1, P<0.05). (XLS 125 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Schneider, C., Nobs, S., Kurrer, M. et al. Induction of the nuclear receptor PPAR-γ by the cytokine GM-CSF is critical for the differentiation of fetal monocytes into alveolar macrophages. Nat Immunol 15, 1026–1037 (2014). https://doi.org/10.1038/ni.3005

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ni.3005

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