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:

Chronic interleukin-1 exposure drives haematopoietic stem cells towards precocious myeloid differentiation at the expense of self-renewal

This article has been updated

Abstract

Haematopoietic stem cells (HSCs) maintain lifelong blood production and increase blood cell numbers in response to chronic and acute injury. However, the mechanism(s) by which inflammatory insults are communicated to HSCs and their consequences for HSC activity remain largely unknown. Here, we demonstrate that interleukin-1 (IL-1), which functions as a key pro-inflammatory ‘emergency’ signal, directly accelerates cell division and myeloid differentiation of HSCs through precocious activation of a PU.1-dependent gene program. Although this effect is essential for rapid myeloid recovery following acute injury to the bone marrow, chronic IL-1 exposure restricts HSC lineage output, severely erodes HSC self-renewal capacity, and primes IL-1-exposed HSCs to fail massive replicative challenges such as transplantation. Importantly, these damaging effects are transient and fully reversible on IL-1 withdrawal. Our results identify a critical regulatory circuit that tailors HSC responses to acute needs, and is likely to underlie deregulated blood homeostasis in chronic inflammation conditions.

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

Figure 1: IL-1 accelerates HSC differentiation along the myeloid lineage.
Figure 2: IL-1 induces precocious activation of a PU.1 gene program in HSCs.
Figure 3: PU.1 activation requires direct and sustained IL-1R signalling.
Figure 4: IL-1 enforces HSC myeloid differentiation in vivo.
Figure 5: IL-1 accelerates myeloid regeneration following acute BM injury.
Figure 6: Chronic IL-1 exposure forces myeloid cell production at the expense of HSC engraftment.
Figure 7: Chronic IL-1 exposure impairs HSC self-renewal.
Figure 8: IL-1 effects are reversible on withdrawal.

Similar content being viewed by others

Change history

  • 28 April 2016

    In the version of this Article originally published online, in the Acknowledgements section, the NIH grant number 'K01 DK09831' should have read 'K01 DK098315'. This has been corrected in all versions of the Article.

References

  1. Orkin, S. H. & Zon, L. I. Hematopoiesis: an evolving paradigm for stem cell biology. Cell 132, 631–644 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  2. Iwasaki, H. & Akashi, K. Myeloid lineage commitment from the hematopoietic stem cell. Immunity 26, 726–740 (2007).

    CAS  PubMed  Google Scholar 

  3. Schepers, K., Campbell, T. C. & Passegué, E. Normal and leukemic stem cell niches: insights and therapeutic opportunities. Cell Stem Cell 16, 254–267 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  4. Wilson, A. et al. Hematopoietic stem cells reversibly switch from dormancy to self-renewal during homeostasis and repair. Cell 135, 1118–1129 (2008).

    CAS  PubMed  Google Scholar 

  5. Pietras, E. M., Warr, M. R. & Passegué, E. Cell cycle regulation in hematopoietic stem cells. J. Cell Biol. 195, 709–720 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Takizawa, H., Boettcher, S. & Manz, M. G. Demand-adapted regulation of early hematopoiesis in infection and inflammation. Blood 119, 2991–3002 (2012).

    CAS  PubMed  Google Scholar 

  7. Medzhitov, R. Origin and physiological roles of inflammation. Nature 454, 428–435 (2008).

    CAS  PubMed  Google Scholar 

  8. Nagai, Y. et al. Toll-like receptors on hematopoietic progenitor cells stimulate innate immune system replenishment. Immunity 24, 801–812 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Scheuttpelz, L. G. & Link, D. C. Regulation of hematopoietic stem cell activity by inflammation. Front. Immunol. 4, 204 (2013).

    Google Scholar 

  10. Mirantes, C., Passegué, E. & Pietras, E. M. Pro-inflammatory cytokines: emerging players regulating HSC function in normal and diseased hematopoiesis. Exp. Cell Res. 329, 248–254 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Essers, M. et al. IFNα activates dormant haematopoietic stem cells in vivo. Nature 458, 904–908 (2009).

    CAS  PubMed  Google Scholar 

  12. Baldridge, M. T., King, K. Y., Boles, N. C., Weksberg, D. C. & Goodell, M. A. Quiescent haematopoietic stem cells are activated by IFN-γ in response to chronic infection. Nature 465, 793–797 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Pietras, E. M. et al. Re-entry into quiescence protects hematopoietic stem cells from the killing effect of chronic exposure to type I interferons. J. Exp. Med. 211, 245–262 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Pronk, C. J., Veiby, O. P., Bryder, D. & Jacobsen, S. E. Tumor necrosis factor restricts hematopoietic stem cell activity in mice: involvement of two distinct receptors. J. Exp. Med. 208, 1563–1570 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Espín-Palazón, R. et al. Proinflammatory signaling regulates hematopoietic stem cell emergence. Cell 159, 1070–1085 (2014).

    PubMed  PubMed Central  Google Scholar 

  16. Li, Y. et al. Inflammatory signaling regulates embryonic hematopoietic stem and progenitor cell production. Genes Dev. 28, 2597–2612 (2014).

    PubMed  PubMed Central  Google Scholar 

  17. King, K. Y. & Goodell, M. A. Inflammatory modulation of HSCs: viewing the HSC as a foundation for the immune response. Nat. Rev. Immunol. 11, 685–692 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Sims, J. E. & Smith, D. E. The IL-1 family: regulators of immunity. Nat. Rev. Immunol. 10, 89–102 (2010).

    CAS  PubMed  Google Scholar 

  19. Dinarello, C. A., Simon, A. & van der Meer, J. W. Treating inflammation by blocking interleukin-1 in a broad spectrum of diseases. Nat. Rev. Drug Discov. 11, 633–652 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Martin, M. U. & Wesche, H. Summary and comparison of the signaling mechanisms of the Toll/interleukin-1 receptor family. BBA Mol. Cell Res. 1592, 265–280 (2002).

    CAS  Google Scholar 

  21. Morrissey, P., Charrier, K., Bressler, L. & Alpert, A. The influence of IL-1 treatment on the reconstitution of the hemopoietic and immune systems after sublethal irradiation. J. Immunol. 140, 4204–4210 (1988).

    CAS  PubMed  Google Scholar 

  22. Damia, G. et al. Prevention of acute chemotherapy-induced death in mice by recombinant human interleukin 1: protection from hematological and nonhematological toxicities. Cancer Res. 52, 4082–4089 (1992).

    CAS  PubMed  Google Scholar 

  23. Hestdal, K. et al. Interleukin-1 (IL-1) directly and indirectly promotes hematopoietic cell growth through type I IL-1 receptor. Blood 84, 125–132 (1994).

    CAS  PubMed  Google Scholar 

  24. Ueda, Y., Cain, D. W., Kuraoka, M., Kondo, M. & Kelsoe, G. IL-1R type I-dependent hematopoietic stem cell proliferation is necessary for inflammatory granulopoieisis and reactive neutrophilia. J. Immunol. 182, 6477–6484 (2009).

    CAS  PubMed  Google Scholar 

  25. Smith, M. A., Knight, S. M., Maddison, P. J. & Smith, J. G. Mechanism of anaemia in rheumatoid arthritis: effect of the blunted response to erythropoietin and of interleukin 1 production by marrow macrophages. Ann. Rheum. Dis. 51, 753–757 (1992).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Dinarello, C. A. Blocking IL-1 in systemic inflammation. J. Exp. Med. 201, 1355–1359 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Cain, D., Kondo, M., Chen, H. & Kelsoe, G. Effects of acute and chronic inflammation on B-cell development and differentiation. J. Invest. Dermatol. 129, 266–277 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Rieger, M. A., Hoppe, P. S., Smejkal, B. M., Eitelhuber, A. C. & Schroeder, T. Hematopoietic cytokines can instruct lineage choice. Science 325, 217–218 (2009).

    CAS  PubMed  Google Scholar 

  29. Cabezas-Wallscheid, N. et al. Identification of regulatory networks in HSCs and their immediate progeny via integrated proteome, transcriptome, and DNA methylome analysis. Cell Stem Cell 15, 507–522 (2014).

    CAS  PubMed  Google Scholar 

  30. Pietras, E. M. et al. Functionally distinct subsets of lineage-biased multipotent progenitors control blood production in normal and regenerative conditions. Cell Stem Cell 17, 35–46 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Enver, T. & May, G. Lineage specification: reading the instructions may help! Curr. Biol. 23, R662–R665 (2013).

    PubMed  Google Scholar 

  32. Staber, P. B. et al. Sustained PU.1 levels balance cell-cycle regulators to prevent exhaustion of adult hematopoietic stem cells. Mol. Cell 49, 934–946 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Rosenbauer, F. et al. Acute myeloid leukemia induced by graded reduction of a lineage-specific transcription factor, PU.1. Nat. Genet. 36, 624–630 (2013).

    Google Scholar 

  34. Steidl, U. et al. A distal single nucleotide polymorphism alters long-range regulation of the PU.1 gene in acute myeloid leukemia. J. Clin. Invest. 117, 2611–2620 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Bonadies, N. et al. PU.1 is regulated by NF-κB through a novel binding site in a 17 kb upstream enhancer element. Oncogene 18, 1062–1072 (2010).

    Google Scholar 

  36. Mossadegh-Keller, N. et al. M-CSF instructs myeloid lineage fate in single haematopoietic stem cells. Nature 497, 239–243 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Takizawa, H., Regoes, R. R., Boddupalli, C. S., Bonhoeffer, S. & Manz, M. G. Dynamic variation in cycling of hematopoietic stem cells in steady state and inflammation. J. Exp. Med. 208, 273–284 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Dorshkind, K. IL-1 inhibits B cell differentiation in long term bone marrow cultures. J. Immunol. 6, 1191–1197 (1988).

    Google Scholar 

  39. Schepers, K. et al. Myeloproliferative neoplasia remodels the endosteal bone marrow niche into a self-reinforcing leukemic niche. Cell Stem Cell 13, 285–299 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Cullen, S. P., Kearney, C. J., Clancy, D. M. & Martin, S. J. Diverse activators of the NLRP3 inflammasome promote IL-1β secretion by triggering necrosis. Cell Rep. 11, 1535–1548 (2015).

    CAS  PubMed  Google Scholar 

  41. Glaccum, M. B. et al. Phenotypic and functional characterization of mice that lack the type I receptor for IL-1. J. Immunol. 159, 3364–3371 (1997).

    CAS  PubMed  Google Scholar 

  42. Mendelson, A. & Frenette, P. S. Hematopoietic stem cell niche maintenance during homeostasis and regeneration. Nat. Med. 20, 833–846 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Gekas, C. & Graf, T. CD41 expression marks myeloid-biased adult hematopoietic stem cells and increases with age. Blood 121, 4463–4472 (2013).

    CAS  PubMed  Google Scholar 

  44. Yamamoto, R. et al. Clonal analysis unveils self-renewing lineage-restricted progenitors generated directly from hematopoietic stem cells. Cell 154, 1112–1126 (2013).

    CAS  PubMed  Google Scholar 

  45. Miyawaki, K. et al. CD41 marks the initial myelo-erythroid specification in adult mouse hematopoiesis: redefinition of murine common myeloid progenitor. Stem Cells 33, 976–987 (2015).

    CAS  PubMed  Google Scholar 

  46. Manz, M. G. & Boettcher, S. Emergency granulopoiesis. Nat. Rev. Immunol. 14, 302–314 (2014).

    CAS  PubMed  Google Scholar 

  47. Dakic, A., Wu, L. & Nutt, S. A. Is PU.1 a dosage-sensitive regulator of haemopoietic lineage commitment and leukaemogenesis? Trends Immunol. 28, 108–114 (2007).

    CAS  PubMed  Google Scholar 

  48. Adamson, J. W. The anemia of inflammation/malignancy: mechanisms and management. Hematol. Am. Soc. Hematol. Educ. Program. 2008, 159–165 (2008).

    Google Scholar 

  49. Zhang, P. et al. PU.1 inhibits GATA-1 function and erythroid differentiation by blocking GATA-1 DNA binding. Blood 96, 2641–2648 (2000).

    CAS  PubMed  Google Scholar 

  50. Zhao, J. L. et al. Conversion of danger signals into cytokine signals by hematopoietic stem and progenitor cells for regulation of stress-induced hematopoieisis. Cell Stem Cell 14, 445–459 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Hirai, H. et al. C/EBPβ is required for ‘emergency’ granulopoiesis. Nat. Immunol. 7, 732–739 (2006).

    CAS  PubMed  Google Scholar 

  52. Matatall, K. A., Shen, C. C., Challen, G. A. & King, K. Y. Type II interferon promotes differentiation of myeloid-biased hematopoietic stem cells. Stem Cells 32, 3023–3030 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Sun, J. et al. Clonal dynamics of native haematopoiesis. Nature 514, 322–327 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Busch, K. et al. Fundamental properties of unperturbed haematopoiesis from stem cells in vivo. Nature 518, 542–546 (2015).

    CAS  PubMed  Google Scholar 

  55. Haas, S. et al. Inflammation-induced emergency megakaryopoiesis driven by hematopoietic stem cell-like megakaryocyte progenitors. Cell Stem Cell 17, 422–434 (2015).

    CAS  PubMed  Google Scholar 

  56. Hu, Y. & Smith, K. ELDA: Extreme limiting dilution analysis for comparing depleted and enriched populations in stem cell and other assays. J. Immunol. Methods 346, 70–78 (2009).

    Google Scholar 

  57. Chow, A. et al. Bone marrow CD169+ macrophages promote the retention of hematopoietic stem and progenitor cells in the mesenchymal stem cell niche. J. Exp. Med. 208, 261–271 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  58. Mohrin, M. et al. Hematopoietic stem cell quiescence promotes error-prone DNA repair and mutagenesis. Cell Stem Cell 7, 174–185 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  59. Flach, J. et al. Replication stress is a potent driver of functional decline in ageing haematopoietic stem cells. Nature 512, 198–202 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank M. Kissner and M. Lee for management of our Flow Cytometry Core Facility and members of the Passegué laboratory for insights and suggestions. E.M.P. was supported by NIH F32 HL106989 and K01 DK098315, C.M.-B. by an Institute of Health Carlos III (ISCIII) fellowship, R.L. by a CIRM Bridges internship and M.G.M. by the Swiss National Science Foundation (310030_146528/1). E.P. and research were supported by a LLS Scholar Award and NIH R01 HL092471.

Author information

Authors and Affiliations

Authors

Contributions

E.M.P. performed all of the experiments with assistance from C.M.-B. and S.F. D.L. helped with the single-cell continuous tracking experiments that were performed in T.S. laboratory. R.L. helped with the Il1r1−/− and 5-FU analyses, J.-M.T. with the in vitro cultures, C.P.C. with analysis of single-cell tracking experiments, L.V.K. and M.G.M. with the in vivo CFSE dilution experiments, B.W. and U.S. with the PU.1ΔURE experiments, and C.N. provided PU.1–EYFP reporter mice. E.M.P., C.M.-B., S.F. and E.P. designed the experiments and interpreted the results. E.M.P. and E.P. wrote the manuscript.

Corresponding authors

Correspondence to Eric M. Pietras or Emmanuelle Passegué.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Gating strategy and effect of IL-1 on progenitor cell proliferation and survival.

(a) Gating strategy and representative FACS plots of stained c-Kit-enriched wild-type (WT) BM cells. MPP2 and MPP3 are often isolated as a combined MPP2/3 population for experiments. (b) Il1r1 expression in the indicated populations. Results are dye intensity levels from microarray analyses30. (n = 5 MPP4, n = 4 HSC and GMP, remainder n = 3). (c) Representative expansion in liquid culture in one of two independent experiments performed in triplicate. (d) Cleaved caspase-3 (CC3) activity expressed as relative luminescence units, RLU) after 12 hours culture in one of two independent experiments performed in triplicate (e) Experimental design and quantification of BrdU incorporation after 24 hours culture in one of two independent experiments performed in triplicate. (f) Mean absolute numbers of the indicated phenotypic cell populations in HSC cultures over time in two independent experiments. Source data for b are shown in Supplementary Table 1. Data are means, and shown in b as means ± S.D. Exact P-values, number of replicates used to derive statistical data (n) and statistical tests used are shown in Supplementary Table 2.

Supplementary Figure 2 Effect of IL-1 on progenitor cell differentiation.

(a,b) Myeloid differentiation in cultured MPP2/3, MPP4 and GMPs: (a) representative FACS plots, and (b) quantification of surface marker expression (n = 4 biological replicates/group). (c) Single cell clonogenic colony forming unit (CFU) assays in methylcellulose (n = 60 cells/group). Colonies were scored after 7 days. MkE: megakaryocyte/erythrocyte; M: macrophage; G: granulocyte; GM: granulocyte/macrophage; GMMkE: mix colony. (d) Representative colony-type (scale bar, 100 μm) and colony-morphology (scale bar, 10 μm; arrows indicate macrophages). (e) Representative FACS plots of myeloid differentiation in cultured HSCs and GMPs (n = 3 biological replicates). Source data for b are shown in Supplementary Table 1. Data in b are means ± S.D.; p ≤ 0.05; p ≤ 0.001. P-values in b were determined by Mann-Whitney u test. Exact P-values, number of replicates used to derive statistical data (n) and statistical tests used are shown in Supplementary Table 2.

Supplementary Figure 3 PU.1 activation by IL-1.

(a,b) qRT-PCR analyses of myeloid lineage genes the indicated populations after 12 hours culture (n = 4 biological replicates/group for HSC, MPP4 and GMP, n = 3 for MPP2/3): (a) experimental design, and (b) expression of individual genes. Results are expressed as fold changes compared to levels in each-IL-1β population (set to 0). (c) PU.1 levels in individual PU.1-eYFP HSCs at 0 and 15 hours culture (n = 140 and 186 individual HSC/group). Results are expressed in arbitrary units (AU) with box plots showing median (lines) and 10–90th percentile (whiskers). (d) Division kinetics of PU.1-eYFP HSCs from Fig. 1e re-analysed based on pre-division PU.1lo (≤4 AU) and PU.1hi (>4 AU) expression levels. (e) Experimental design, representative histograms and quantification of BrdU incorporation in HSCs after 24 hours culture (n = 3 biological replicates/group). (f) PU.1 levels in PU.1-eYFP HSCs after 12 hours culture with the indicated concentrations of IL-1β. (g) Representative histograms showing PU.1 levels in PU.1-eYFP HSCs after 12 hours culture ± IL-1β (25 pg/ml) and the indicated inhibitors. Results with Il1r1−/−::PU.1-eYFP HSCs are shown as negative controls. (h) Validation of the blocking activity of the anti-M-CSFR antibody (αMR) in BM cells cultured for 5 days? M-CSF (M, 100 ng/ml). Source data for b and e are shown in Supplementary Table 1. Data are means ± S.D.; p ≤ 0.05; p ≤ 0.001. P-values in b and e were determined by paired Student’s t test, and in c,d by one-way ANOVA with Tukey’s test. Exact P-values, number of replicates used to derive statistical data (n) and statistical tests used are shown in Supplementary Table 2.

Supplementary Figure 4 Gating strategies and effect of acute in vivo IL-1 treatment.

(ac) Gating strategies with representative FACS plots for the different hematopoietic stem, progenitor and mature cell populations analysed in peripheral blood (PB) and bone marrow (BM): (a) PB of mice injected ± IL-1β for 20 days, (b) and (c) BM of mice injected ± IL-1β for 1 day. Number in each gate indicates population frequency. (d) BM cellularity and size of the indicated BM populations in mice injected ± IL-1β for 1 day (n = 15 −IL-1β and 13 +IL-1β mice/group). (e) Numbers of HSCLT and MPP1 in Il1r1+/+ and Il1r1−/− mice in mice injected ± IL-1β for 1 day (n = 15 −IL-1β and +IL-1β Il1r1+/+ mice, 5 −IL-1β and 6 +IL-1β Il1r1−/− mice/group) Data are means ± S.D.; p ≤ 0.05; p ≤ 0.01; p ≤ 0.001. P-values in d and e were determined by Mann-Whitney u test. Exact P-values, number of replicates used to derive statistical data (n) and statistical tests used are shown in Supplementary Table 2.

Supplementary Figure 5 In vivo HSC division tracking and specificity of PU.1 activation.

(a) Representative FACS plots showing CFSE dilution in donor cells following 7 days treatment of the recipient mice ± IL-1β. (b) Gating strategy for identification of HSCLT in CFSE-labelled donor cells. (c) PU.1 levels in the indicated populations from PU.1-eYFP mice injected ± IL-1β for 1 or 20 days (n = 4 −IL-1β and 5 +IL-1β_ mice/group for day 1; n = 5 −IL-1β and 3 +IL-1β_ mice/group for day 20). Source data for c are shown in Supplementary Table 1. Data are means ± S.D.; p ≤ 0.05. P-values in c were determined by Mann-Whitney u test. Exact P-values, number of replicates used to derive statistical data (n) and statistical tests used are shown in Supplementary Table 2.

Supplementary Figure 6 Cytokine production following 5-FU myeloablation, identification of IL-1 producing cells and haematopoiesis in Il1r1−/− mice.

(a) Cytokine levels in BM plasma (top) and blood serum (bottom) of 5-FU-treated mice (mice/group: n = 7 day 0–8; n = 8 day 10; n = 6 day 12; n = 3 day 14; for M-CSF n = 4 day 0–12; n = 3 day 14). (b) Experimental design and gating strategy used to isolate BM stromal ECs, MSCs and OBCs, and hematopoietic granulocytes (Gr), monocytes (Mon), macrophages (MΦ), B cells and CD4 + T cells. Representative FACS plots of untreated d0 mice are shown. (c) Representative FACS plots and quantification of Ki67/DAPI cell cycle distribution in HSCs from Il1r1+/+ and Il1r1−/− mice (n = 4 Il1r1+/+ and 6 Il1r1−/− mice/group). (d) BM cellularity and size of the indicated BM populations in Il1r1+/+ and Il1r1−/− mice (n = 20 mice/group; for HSCLT and MPP1, n = 16 Il1r1+/+ and 13 Il1r1−/− mice/group) Source data for a are shown in Supplementary Table 1. Data are means ± S.D.; p ≤ 0.05; p ≤ 0.01; p ≤ 0.001. P-values in a were determined by one-way ANOVA with Dunnet’s test, and in c and d by Mann-Whitney u test. Exact P-values, number of replicates used to derive statistical data (n) and statistical tests used are shown in Supplementary Table 2.

Supplementary Figure 7 Short-term in vitro and in vivo tracking of HSCs.

(a) Representative methylcellulose colony forming-unit (CFU) and cleaved caspase-3 (CC3; expressed as relative luminescence units: (RLU) assays. Results are representative of one of two independent experiments performed in triplicate. b-c, Terminal analyses of mice transplanted with HSCs exposed ± IL-1β for 20 days in donor mice and another 30 days in recipient mice shown in Fig. 6a-d): representative FACS plots showing donor chimerism, and myeloid (My) and lymphoid (Ly) donor lineage distribution in (b) PB and (c) BM HSCs 30 days post-transplant. (d) Representative FACS plots showing the frequency of GFP+ control and PU.1-transduced HSCs prior to transplantation (d0 input) and after 30 days post-transplant in the BM (d30). Data in a are shown as means.

Supplementary Figure 8 Functional analysis of IL-1-exposed HSCs.

(ac) Limiting dilution analysis (LDA) of unfractionated BM cells from mice injected ± IL-1β for 20 days (106 cell dose: n = 4 −IL-1β and 4 +IL-1β mice/group; 105 cell dose: n = 4 mice/group; 104 cell dose: n = 4 −IL-1β and 3 +IL-1β mice/group): (a) experimental design, (b) LDA graph (values: estimated HSC frequency; solid lines: dose response curve; dashed lines: upper and lower confidence intervals), and (c) engraftment results at 16 weeks post-transplant, Mice with ≥0.5% donor chimerism were considered engrafted. (df) Analyses of mice injected ± IL-1β for 70 days n = 5 mice/group): (d) representative FACS plots and HSCLT number fractionated by CD41 expression (n = 4 −IL-1β and 5 +IL-1β mice/group), (e) size (means) of the indicated BM populations from two mice, and (f) methylcellulose colony forming-unit (CFU) assay in one experiment performed in triplicate. (gi) Secondary transplantation (2° txpl) of HSCs re-isolated from primary transplanted mice (1° txpl, shown in Fig. 7d–f) reconstituted with HSCLT from mice injected ± IL-1β for 70 days (n = 5 mice/group): (g) experimental design, (h) donor chimerism and (i) lineage distribution in PB over time. Source data for bd are shown in Supplementary Table 1. Data are means ± S.D. except when indicated. P-values in b were determined using Poisson statistical analysis, in c,d,h and i by Mann-Whitney u test. Exact P-values, number of replicates used to derive statistical data (n) and statistical tests used are shown in Supplementary Table 2.

Supplementary information

Supplementary Information

Supplementary Information (PDF 2632 kb)

Supplementary Table 1

Statistics source data. (XLSX 111 kb)

Supplementary Table 2

Exact P-values and statistical tests used in experiments. (XLSX 68 kb)

Supplementary Table 3

Antibodies used. (XLSX 51 kb)

Supplementary Table 4

Primers used for qRT-PCR and Fluidigm analysis. (XLSX 11 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Pietras, E., Mirantes-Barbeito, C., Fong, S. et al. Chronic interleukin-1 exposure drives haematopoietic stem cells towards precocious myeloid differentiation at the expense of self-renewal. Nat Cell Biol 18, 607–618 (2016). https://doi.org/10.1038/ncb3346

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ncb3346

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