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:

Bhlhe40 mediates tissue-specific control of macrophage proliferation in homeostasis and type 2 immunity

Abstract

Most tissue-resident macrophage populations develop during embryogenesis, self-renew in the steady state and expand during type 2 immunity. Whether shared mechanisms regulate the proliferation of macrophages in homeostasis and disease is unclear. Here we found that the transcription factor Bhlhe40 was required in a cell-intrinsic manner for the self-renewal and maintenance of large peritoneal macrophages (LPMs), but not that of other tissue-resident macrophages. Bhlhe40 was necessary for the proliferation, but not the polarization, of LPMs in response to the cytokine IL-4. During infection with the helminth Heligmosomoides polygyrus bakeri, Bhlhe40 was required for cell cycling of LPMs. Bhlhe40 repressed the expression of genes encoding the transcription factors c-Maf and Mafb and directly promoted expression of transcripts encoding cell cycle-related proteins to enable the proliferation of LPMs. In LPMs, Bhlhe40 bound to genomic sites co-bound by the macrophage lineage-determining factor PU.1 and to unique sites, including Maf and loci encoding cell-cycle-related proteins. Our findings demonstrate a tissue-specific control mechanism that regulates the proliferation of resident macrophages in homeostasis and type 2 immunity.

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: Loss of Bhlhe40 dysregulates the cell cycle in LPMs.
Fig. 2: Bhlhe40 is cell-intrinsically required in LPMs to regulate the cell cycle.
Fig. 3: Bhlhe40 regulates a distinct set of genes related to alternative activation in LPMs.
Fig. 4: Bhlhe40 is required for normal accumulation of resident, but not recruited, macrophages in the peritoneum.
Fig. 5: Bhlhe40 is required for normal cycling, but not polarization, of peritoneal macrophages during type 2 immunity.
Fig. 6: Bhlhe40 is required for LPM proliferation in response to H. polygyrus.
Fig. 7: Bhlhe40 regulates gene expression to modulate proliferation, but not alternative activation, in LPMs during type 2 immunity.
Fig. 8: Bhlhe40 directly regulates gene expression in LPMs in an activation state-dependent manner.

Similar content being viewed by others

Data availability

The data that support the findings of this study are available from the corresponding author upon request. The microarray and ChIP-sequencing data have been deposited in the GEO repository under accession code GSE125730.

References

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

    Article  CAS  Google Scholar 

  2. 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 

  3. T’Jonck, W., Guilliams, M. & Bonnardel, J. Niche signals and transcription factors involved in tissue-resident macrophage development. Cell. Immunol. 330, 43–53 (2018).

    Article  Google Scholar 

  4. 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  Google Scholar 

  5. 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 

  6. Aziz, A., Soucie, E., Sarrazin, S. & Sieweke, M. H. MafB/c-Maf deficiency enables self-renewal of differentiated functional macrophages. Science 326, 867–871 (2009).

    Article  CAS  Google Scholar 

  7. Soucie, E. L. et al. Lineage-specific enhancers activate self-renewal genes in macrophages and embryonic stem cells. Science 351, 680–693 (2016).

    Article  CAS  Google Scholar 

  8. Imperatore, F. et al. SIRT1 regulates macrophage self-renewal. EMBO J. 36, 2353–2372 (2017).

    Article  CAS  Google Scholar 

  9. Rosas, M. et al. The transcription factor Gata6 links tissue macrophage phenotype and proliferative renewal. Science 344, 645–648 (2014).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  11. Gautier, E. L. et al. Gata6 regulates aspartoacylase expression in resident peritoneal macrophages and controls their survival. J. Exp. Med. 211, 1525–1531 (2014).

    Article  CAS  Google Scholar 

  12. Jenkins, S. J. et al. Local macrophage proliferation, rather than recruitment from the blood, is a signature of TH2 inflammation. Science 332, 1284–1288 (2011).

    Article  CAS  Google Scholar 

  13. Jenkins, S. J. et al. IL-4 directly signals tissue-resident macrophages to proliferate beyond homeostatic levels controlled by CSF-1. J. Exp. Med. 210, 2477–2491 (2013).

    Article  CAS  Google Scholar 

  14. Ruckerl, D. & Allen, J. E. Macrophage proliferation, provenance, and plasticity in macroparasite infection. Immunol. Rev. 262, 113–133 (2014).

    Article  Google Scholar 

  15. Minutti, C. M. et al. Local amplifiers of IL-4Rα-mediated macrophage activation promote repair in lung and liver. Science 356, 1076–1080 (2017).

    Article  CAS  Google Scholar 

  16. Bosurgi, L. et al. Macrophage function in tissue repair and remodeling requires IL-4 or IL-13 with apoptotic cells. Science 356, 1072–1076 (2017).

    Article  CAS  Google Scholar 

  17. Gundra, U. M. et al. Alternatively activated macrophages derived from monocytes and tissue macrophages are phenotypically and functionally distinct. Blood 123, e110–e122 (2014).

    Article  CAS  Google Scholar 

  18. Gundra, U. M. et al. Vitamin A mediates conversion of monocyte-derived macrophages into tissue-resident macrophages during alternative activation. Nat. Immunol. 18, 642–653 (2017).

    Article  CAS  Google Scholar 

  19. Kato, Y., Kawamoto, T., Fujimoto, K. & Noshiro, M. DEC1/STRA13/SHARP2 and DEC2/SHARP1 coordinate physiological processes, including circadian rhythms in response to environmental stimuli. Curr. Top. Dev. Biol. 110, 339–372 (2014).

    Article  CAS  Google Scholar 

  20. Ow, J. R., Tan, Y. H., Jin, Y., Bahirvani, A. G. & Taneja, R. Stra13 and sharp-1, the non-grouchy regulators of development and disease. Curr. Top. Dev. Biol. 110, 317–338 (2014).

    Article  CAS  Google Scholar 

  21. Lin, C. C. et al. IL-1-induced Bhlhe40 identifies pathogenic T helper cells in a model of autoimmune neuroinflammation. J. Exp. Med. 213, 251–271 (2016).

    Article  CAS  Google Scholar 

  22. Sun, H. & Taneja, R. Stra13 expression is associated with growth arrest and represses transcription through histone deacetylase (HDAC)-dependent and HDAC-independent mechanisms. Proc. Natl Acad. Sci. USA 97, 4058–4063 (2000).

    Article  CAS  Google Scholar 

  23. St-Pierre, B., Flock, G., Zacksenhaus, E. & Egan, S. E. Stra13 homodimers repress transcription through class B E-box elements. J. Biol. Chem. 277, 46544–46551 (2002).

    Article  CAS  Google Scholar 

  24. Li, Y. et al. The expression of antiapoptotic protein survivin is transcriptionally upregulated by DEC1 primarily through multiple sp1 binding sites in the proximal promoter. Oncogene 25, 3296–3306 (2006).

    Article  CAS  Google Scholar 

  25. Qian, Y., Zhang, J., Jung, Y. S. & Chen, X. DEC1 coordinates with HDAC8 to differentially regulate TAp73 and DeltaNp73 expression. PLoS One 9, e84015 (2014).

    Article  Google Scholar 

  26. Seimiya, M. et al. Impaired lymphocyte development and function in Clast5/Stra13/DEC1-transgenic mice. Eur. J. Immunol. 34, 1322–1332 (2004).

    Article  CAS  Google Scholar 

  27. Kanda, M. et al. Transcriptional regulator Bhlhe40 works as a cofactor of T-bet in the regulation of IFN-γ production in iNKT cells. Proc. Natl Acad. Sci. USA 113, E3394–E3402 (2016).

    Article  CAS  Google Scholar 

  28. Kreslavsky, T. et al. Essential role for the transcription factor Bhlhe41 in regulating the development, self-renewal and BCR repertoire of B-1a cells. Nat. Immunol. 18, 442–455 (2017).

    Article  CAS  Google Scholar 

  29. Camponeschi, A. et al. DEC1/STRA13 is a key negative regulator of activation-induced proliferation of human B cells highly expressed in anergic cells. Immunol. Lett. 198, 7–11 (2018).

    Article  CAS  Google Scholar 

  30. Martinez-Llordella, M. et al. CD28-inducible transcription factor DEC1 is required for efficient autoreactive CD4+ T cell response. J. Exp. Med. 210, 1603–1619 (2013).

    Article  CAS  Google Scholar 

  31. Lin, C. C. et al. Bhlhe40 controls cytokine production by T cells and is essential for pathogenicity in autoimmune neuroinflammation. Nat. Commun. 5, 3551 (2014).

    Article  Google Scholar 

  32. Yu, F. et al. The transcription factor Bhlhe40 is a switch of inflammatory versus antiinflammatory Th1 cell fate determination. J. Exp. Med. 215, 1813–1821 (2018).

    Article  CAS  Google Scholar 

  33. Huynh, J. P. et al. Bhlhe40 is an essential repressor of IL-10 during Mycobacterium tuberculosis infection. J. Exp. Med. 215, 1823–1838 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Gabryšová, L. & O’Garra, A. Regulating the regulator: Bhlhe40 directly keeps IL-10 in check. J. Exp. Med. 215, 1767–1769 (2018).

    PubMed  PubMed Central  Google Scholar 

  35. Gosselin, D. et al. Environment drives selection and function of enhancers controlling tissue-specific macrophage identities. Cell 159, 1327–1340 (2014).

    Article  CAS  Google Scholar 

  36. 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 

  37. Lavin, Y. et al. Tissue-resident macrophage enhancer landscapes are shaped by the local microenvironment. Cell 159, 1312–1326 (2014).

    Article  CAS  Google Scholar 

  38. Scott, C. L. et al. Bone marrow-derived monocytes give rise to self-renewing and fully differentiated Kupffer cells. Nat. Commun. 7, 10321 (2016).

    Article  CAS  Google Scholar 

  39. Bain, C. C. et al. Long-lived self-renewing bone marrow-derived macrophages displace embryo-derived cells to inhabit adult serous cavities. Nat. Commun. 7, 11852 (2016).

    Article  Google Scholar 

  40. Shaw, T. N. et al. Tissue-resident macrophages in the intestine are long lived and defined by Tim-4 and CD4 expression. J. Exp. Med. 215, 1507–1518 (2018).

    Article  CAS  Google Scholar 

  41. 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  Google Scholar 

  42. Subramanian, A. et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc. Natl Acad. Sci. USA 102, 15545–15550 (2005).

    Article  CAS  Google Scholar 

  43. Gautier, E. L., Ivanov, S., Lesnik, P. & Randolph, G. J. Local apoptosis mediates clearance of macrophages from resolving inflammation in mice. Blood 122, 2714–2722 (2013).

    Article  CAS  Google Scholar 

  44. Bertoli, C., Skotheim, J. M. & de Bruin, R. A. Control of cell cycle transcription during G1 and S phases. Nat. Rev. Mol. Cell. Biol. 14, 518–528 (2013).

    Article  CAS  Google Scholar 

  45. Liberzon, A. et al. The Molecular Signatures Database (MSigDB) hallmark gene set collection. Cell Syst. 1, 417–425 (2015).

    Article  CAS  Google Scholar 

  46. Cain, D. W. et al. Identification of a tissue-specific, C/EBPβ-dependent pathway of differentiation for murine peritoneal macrophages. J. Immunol. 191, 4665–4675 (2013).

    Article  CAS  Google Scholar 

  47. Franklin, R. A. & Li, M. O. Ontogeny of tumor-associated macrophages and its implication in cancer regulation. Trends Cancer 2, 20–34 (2016).

    Article  Google Scholar 

  48. 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  Google Scholar 

  49. Loyher, P. L. et al. Macrophages of distinct origins contribute to tumor development in the lung. J. Exp. Med. 215, 2536–2553 (2018).

    Article  CAS  Google Scholar 

  50. Mantovani, A., Marchesi, F., Malesci, A., Laghi, L. & Allavena, P. Tumour-associated macrophages as treatment targets in oncology. Nat. Rev. Clin. Oncol. 14, 399–416 (2017).

    Article  CAS  Google Scholar 

  51. Sun, H., Lu, B., Li, R. Q., Flavell, R. A. & Taneja, R. Defective T cell activation and autoimmune disorder in Stra13-deficient mice. Nat. Immunol. 2, 1040–1047 (2001).

    Article  CAS  Google Scholar 

  52. Finkelman, F. D. et al. Anti-cytokine antibodies as carrier proteins. Prolongation of in vivo effects of exogenous cytokines by injection of cytokine-anti-cytokine antibody complexes. J. Immunol. 151, 1235–1244 (1993).

    CAS  Google Scholar 

  53. Camberis, M., Le Gros, G. & Urban, J. Jr. Animal model of Nippostrongylus brasiliensis and Heligmosomoides polygyrus. Curr. Protoc. Immunol. Chapter 19, Unit 19.12 (2003).

    PubMed  Google Scholar 

  54. Bando, J. K. et al. The tumor necrosis factor superfamily member RANKL suppresses effector cytokine production in group 3 innate lymphoid cells. Immunity 48, 1208–1219 (2018).

    Article  CAS  Google Scholar 

  55. Jablonski, K. A. et al. Novel markers to delineate murine M1 and M2 macrophages. PLoS One 10, e0145342 (2015).

    Article  Google Scholar 

  56. Zhang, Y. et al. Model-based analysis of ChIP-Seq (MACS). Genome Biol. 9, R137 (2008).

    Article  Google Scholar 

  57. Bailey, T. L. DREME: motif discovery in transcription factor ChIP-seq data. Bioinformatics 27, 1653–1659 (2011).

    Article  CAS  Google Scholar 

  58. Micallef, L. & Rodgers, P. EulerAPE: drawing area-proportional 3-Venn diagrams using ellipses. PLoS One 9, e101717 (2014).

    Article  Google Scholar 

Download references

Acknowledgements

This work was supported by the National Institute of Allergy and Infectious Disease (NIAID) (AI113118 and AI132653) (B.T.E.) and a Burroughs Wellcome Fund Career Award for Medical Scientists (B.T.E.). N.N.J. was supported by grant 5T32AI007163 from the NIAID. C.-C.L. was supported by the McDonnell International Scholars Academy at Washington University. S.C.-C.H. was supported by the Case Comprehensive Cancer Center American Cancer Society IRG Award (IRG−16–186–21). M.E.C. was supported by the National Science Foundation Graduate Research Fellowship program (DGE-1745038). Research reported in this publication was supported by the Washington University Institute of Clinical and Translational Sciences grant UL1TR002345 from the National Center for Advancing Translational Sciences (NCATS) of the National Institutes of Health (NIH). The content is solely the responsibility of the authors and does not necessarily represent the official view of the NIH. We thank E. Lantelme, A. Cullen and D. Brinja for help with fluorescence-activated cell sorting. We thank W. Beatty and L. Mwaghore for electron microscopy. We thank S. Van Dyken and C. Farnsworth for critical reading of the manuscript. We thank the members of the G. Randolph laboratory for helpful discussions about this project. We thank J. Bando, M. Robinette and T. Ai for help with flow cytometry of the gut.

Author information

Authors and Affiliations

Authors

Contributions

N.N.J designed, performed and analyzed experiments and wrote the manuscript. E.A.S., T.R.B., C.-C.L., M.E.C. and C.-W.L. performed experiments. S.C.-C.H. provided reagents, protocols and technical expertise. I.S. and M.N.A. analyzed ChIP-seq data. R.T. provided mice. T.S.S., G.J.R. and J.F.U. provided reagents and technical expertise. B.T.E. designed and analyzed experiments, supervised the studies and wrote the manuscript.

Corresponding author

Correspondence to Brian T. Edelson.

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 Fig. 1 Bhlhe40 is specifically required in peritoneal and pleural macrophages.

a, Flow cytometry of Bhlhe40GFP transgene reporter expression in pleural macrophages from Bhlhe40GFP+ and Bhlhe40GFP mice (representative of 2 experiments, n = 3 Bhlhe40GFP+, 2 Bhlhe40GFP-). b-e, Flow cytometry for AMs (b), red pulp macrophages (c), kidney macrophages (d), and Kupffer cells (e) from Bhlhe40+/+ and Bhlhe40-/- mice as in Fig. 1c. f, Flow cytometry for Tim4 expression on LPMs from Bhlhe40+/+ and Bhlhe40-/- mice (representative of 11 experiments, n = 40/group). g, Numbers of Tim4+ LPMs as in f. h, Flow cytometry for CD226 expression on SPMs from Bhlhe40+/+ and Bhlhe40-/- mice (representative of 8 experiments, n = 27/group). i, Numbers of CD226+ SPMs as in h. j, Numbers of peritoneal B cells from Bhlhe40+/+ and Bhlhe40-/- mice (pooled from 12 experiments, n = 35/group). k, Numbers of large pleural macrophages from Bhlhe40+/+ and Bhlhe40-/- mice (pooled from 6 experiments, n = 16 Bhlhe40+/+, 17 Bhlhe40-/-). l, Frequency of Ki67+ large pleural macrophages from Bhlhe40+/+ and Bhlhe40-/- mice (pooled from 3 experiments, n = 7 Bhlhe40+/+, 8 Bhlhe40-/-). m, frequency of 7-AAD+ LPMs from Bhlhe40+/+ and Bhlhe40-/- mice (pooled from 8 experiments, n = 19 Bhlhe40+/+, 18 Bhlhe40-/-). Data in g,i-m are mean ± s.e.m; each symbol represents an individual mouse (g,i-m). ***P < 0.001; ****P < 0.0001, unpaired two-sided Student’s t-test.

Supplementary Fig. 2 Bhlhe40 is cell-intrinsically required in LPMs.

a, Ratio of CD45.1 to CD45.2 cells for small pleural macrophages and pleural B cells from mixed bone marrow chimeras as in Fig. 2d. b, Flow cytometry for the discrimination of donor and recipient LPMs and peritoneal B cells from CD45.1/CD45.2 recipients of transferred peritoneal cells as in Fig. 2k, l. c,d, Gene expression microarray data from Bhlhe40+/+ and Bhlhe40-/- LPMs (in this study) and LysM-Cre- Gata6fl/fl, and LysM-Cre+ Gata6fl/fl LPMs (reanalyzed from 36) were analyzed for shared and unique Bhlhe40 and/or Gata6-dependent genes ( ≥ 2-fold differentially expressed, depicted as a Venn diagram) (c) and differentially expressed genes dependent on both Bhlhe40 and Gata6 (d). e, Gene expression microarray data were analyzed for expression of an LPM gene signature in LPMs and AMs from Bhlhe40+/+ and Bhlhe40-/- mice. Microarray data from LPMs (n = 3/group) and AMs (n = 2/group) are from a single experiment. Data in a are mean ± s.e.m; each symbol represents an individual mouse (a). Significance calculated with an unpaired two-sided Student’s t-test.

Supplementary Fig. 3 Further analysis of responses to IL-4c in Bhlhe40-deficient mice.

a, Flow cytometry of peritoneal macrophage subsets from Bhlhe40+/+, Bhlhe40-/-, and Bhlhe40-/- Il10-/- mice treated with PBS or IL-4c (representative of 2 experiments, n = 3 PBS-treated Bhlhe40+/+, 2 PBS-treated Bhlhe40-/-, 3 PBS-treated Bhlhe40-/- Il10-/-, 5 IL-4c-treated Bhlhe40+/+, 7 IL-4c-treated Bhlhe40-/-, 4 IL-4c-treated Bhlhe40-/- Il10-/-). b, Numbers of LPMs as in a. c, Immunoblotting of cyclins D1-3, cyclin-dependent kinase (CDK) 2, CDK4, CDK6, E2F2, and beta actin in lysates of LPMs from Bhlhe40+/+ and Bhlhe40-/- mice unstimulated or treated with IL-4c (representative of 2 experiments, n = 2/group). d, Frequency of BrdU+ Bhlhe40+/+ (CD45.1), Bhlhe40+/+ (CD45.2), or Bhlhe40-/- (CD45.2) LPMs from mixed bone marrow chimera mice (generated as in Fig. 2a–f) treated with PBS or IL-4c, with LPMs from each donor recovered from the same recipient connected by a line (pooled from 2 experiments, n = 2 PBS-treated [Bhlhe40+/+ (CD45.1) +Bhlhe40+/+ (CD45.2)] and [Bhlhe40+/+ (CD45.1) +Bhlhe40-/- (CD45.2)]; 4 IL-4c-treated [Bhlhe40+/+ (CD45.1) +Bhlhe40+/+ (CD45.2)]; 5 [Bhlhe40+/+ (CD45.1) +Bhlhe40-/- (CD45.2)])). Data in b,d are mean ± s.e.m; (b) each symbol or (d) paired symbols represent an individual mouse. ***P < 0.001, (b) unpaired or (d) paired two-sided Student’s t-test.

Supplementary Fig. 4 Loss of Bhlhe40 causes morphological changes in in vivo IL-4c-stimulated peritoneal macrophages.

a,b, Transmission electron microscopy images of LPMs from naïve (a) and IL-4c-treated (b) Bhlhe40+/+ and Bhlhe40-/- mice (representative of 2 experiments, n = 2 mice (45–50 images)/group). Scale bar, 2 μm. c-h, Transmission electron microscopy for cellular cross-sectional area (c), endoplasmic reticulum (ER) cross-sectional extent (d), ER luminal width (e), vesicle cross-sectional area (f), nucleoli/cross-section (g), mitochondria/cross-section (h) of LPMs from Bhlhe40+/+ and Bhlhe40-/- mice unstimulated or treated with IL-4c (pooled from 2 experiments, n = 2 mice/group [27–60 cells analyzed, except for 17–20 cells analyzed in (d)]). Data in c-h are mean ± s.e.m; each symbol represents an individual cell (c-h). *P ≤ 0.05; **P < 0.01; ***P < 0.001; ****P < 0.0001, unpaired two-sided Student’s t -test.

Supplementary Fig. 5 Bhlhe40 expression is tightly regulated in resident macrophages.

a, Flow cytometry of Bhlhe40GFP transgene reporter expression in LPMs, SPMs, red pulp macrophages, Kupffer cells, kidney macrophages, and AMs from Bhlhe40GFP+ and Bhlhe40GFP- mice after PBS or IL-4c treatment (representative of 1–2 experiments, n = 3 PBS-treated Bhlhe40GFP+, 1 PBS-treated Bhlhe40GFP-, 5 IL-4c-treated Bhlhe40GFP+, 3 IL-4c-treated Bhlhe40GFP-). b, Numbers of LPMs, red pulp macrophages, Kupffer cells, and AMs from Bhlhe40+/+ and Bhlhe40-/- mice treated with PBS or IL-4c (pooled from 2 experiments, n = 4 PBS-treated Bhlhe40+/+, 3 PBS-treated Bhlhe40-/-, 8 IL-4c-treated Bhlhe40+/+, 7 IL-4c-treated Bhlhe40-/-). c-e, Flow cytometry of BrdU incorporation by red pulp macrophages (c), Kupffer cells (d), and AMs (e) from Bhlhe40+/+ and Bhlhe40-/- mice treated with PBS or IL-4c as in Fig. 5g. Data in b are mean ± s.e.m; each symbol represents an individual mouse (b). *P ≤ 0.05, unpaired two-sided Student’s t-test.

Supplementary Fig. 6 Bhlhe40 is required for normal proliferation of thioglycollate-elicited macrophages during type 2 immunity.

a, Flow cytometry of BrdU incorporation by LPMs and Thio-elicited macrophages from Bhlhe40+/+ and Bhlhe40-/- mice treated with PBS, IL-4c, thioglycollate (Thio), or Thio and IL-4c as in Fig. 5j. b,c, Frequency of pHH3+ LPMs and Thio-elicited macrophages (b) (pooled from 2 experiments, n = 3 PBS-treated Bhlhe40+/+; 4 PBS-treated Bhlhe40-/-; 4 IL-4c-treated and Thio-treated Bhlhe40+/+ and Bhlhe40-/-; 6 Thio and IL-4c-treated Bhlhe40+/+ and Bhlhe40-/-) and RELMα+ LPMs and Thio-elicited macrophages (c) (pooled as in b) from Bhlhe40+/+ and Bhlhe40-/- mice treated with PBS, IL-4c, thioglycollate (Thio), or Thio and IL-4c. Data in b,c are mean ± s.e.m; each symbol represents an individual mouse (b,c). *P ≤ 0.05; ***P < 0.001, unpaired two-sided Student’s t-test.

Supplementary Fig. 7 Bhlhe40 directly regulates gene transcription in LPMs.

a, Gene expression microarray data were analyzed for expression of genes encoding selective regulators of LPM proliferation (Myo18a and C1q) in LPMs from Bhlhe40+/+, Bhlhe40-/-, and LysM-Cre+ Bhlhe40fl/fl mice unstimulated or treated with IL-4c. b-d, Tracings of Bhlhe40 binding, PU.1 binding, and vertebrate conservation at the Il10 (b), Ccl2 (c), and Plac8 (d) loci. e, Bhlhe40-bound, Bhlhe40-dependent genes ( ≥ 2-fold differentially expressed in Bhlhe40+/+ and Bhlhe40-/- LPMs) in LPMs from naïve mice and Bhlhe40-bound, Bhlhe40-dependent genes ( ≥ 2-fold differentially expressed in Bhlhe40+/+ and Bhlhe40-/- LPMs) in LPMs from IL-4c-treated mice, as in Fig. 8i. Underlined genes are highlighted elsewhere in this study. f-h, Tracings of Bhlhe40 binding, PU.1 binding, and vertebrate conservation at the Maf (distal) (f), Mafb (g), and Mafb (distal) (h) loci. LPM Bhlhe40 ChIP-seq data (n = 1/group), microarray data from naïve LPMs (n = 3/group), and microarray data from IL-4c-stimulated LPMs (n = 2/group) are from single separate experiments. LPM PU.1 ChIP-seq data reanalyzed from 35.

Supplementary Fig. 8 Bhlhe40 directly binds to cell cycle-related loci and is required to sustain normal gene expression.

a-d, The proportion of Bhlhe40-bound members of gene sets enriched in Bhlhe40+/+ compared to Bhlhe40-/- LPMs from IL-4c-treated mice. Tracings of Bhlhe40 binding, PU.1 binding, and vertebrate conservation for a representative member of the core enrichment signature for each gene set is presented. Hallmark E2F Targets (a), Hallmark Myc Targets (v1) (b), C5 Chromosome Organization (c), and C5 Cell Cycle Process (d). e-h, GSEA of expression of Bhlhe40-bound genes from gene expression microarray data from LPMs from Bhlhe40+/+ and Bhlhe40-/- mice treated with IL-4c for Hallmark E2F Targets (e), Hallmark Myc Targets v1 (f), C5 Chromosome Organization (g), and C5 Cell Cycle Process (h). C5 Cell Cycle Process is also presented in Fig. 8i. NES, normalized enrichment score. FWER, family-wise error rate. LPM Bhlhe40 ChIP-seq data (n = 1/group) and microarray data (n = 2/group) are from single separate experiments. LPM PU.1 ChIP-seq data reanalyzed from 35. (e-h) NES and FWER.

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Jarjour, N.N., Schwarzkopf, E.A., Bradstreet, T.R. et al. Bhlhe40 mediates tissue-specific control of macrophage proliferation in homeostasis and type 2 immunity. Nat Immunol 20, 687–700 (2019). https://doi.org/10.1038/s41590-019-0382-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-019-0382-5

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