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.

  • Letter
  • Published:

Neutrophil ageing is regulated by the microbiome

This article has been updated

Abstract

Blood polymorphonuclear neutrophils provide immune protection against pathogens, but may also promote tissue injury in inflammatory diseases1,2. Although neutrophils are generally considered to be a relatively homogeneous population, evidence for heterogeneity is emerging3,4. Under steady-state conditions, neutrophil heterogeneity may arise from ageing and replenishment by newly released neutrophils from the bone marrow5. Aged neutrophils upregulate CXCR4, a receptor allowing their clearance in the bone marrow6,7, with feedback inhibition of neutrophil production via the IL-17/G-CSF axis8, and rhythmic modulation of the haematopoietic stem-cell niche5. The aged subset also expresses low levels of L-selectin5,9. Previous studies have suggested that in vitro-aged neutrophils exhibit impaired migration and reduced pro-inflammatory properties6,10. Here, using in vivo ageing analyses in mice, we show that neutrophil pro-inflammatory activity correlates positively with their ageing whilst in circulation. Aged neutrophils represent an overly active subset exhibiting enhanced αMβ2 integrin activation and neutrophil extracellular trap formation under inflammatory conditions. Neutrophil ageing is driven by the microbiota via Toll-like receptor and myeloid differentiation factor 88-mediated signalling pathways. Depletion of the microbiota significantly reduces the number of circulating aged neutrophils and dramatically improves the pathogenesis and inflammation-related organ damage in models of sickle-cell disease or endotoxin-induced septic shock. These results identify a role for the microbiota in regulating a disease-promoting neutrophil subset.

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: Aged neutrophils represent an overly active subset of neutrophils.
Figure 2: Neutrophil ageing is driven by the microbiota.
Figure 3: Microbiota-driven neutrophil ageing is mediated by neutrophil TLRs and Myd88 signalling.
Figure 4: Microbiota depletion reduces vaso-occlusive events in sickle-cell disease.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

Data deposits

Microarray data have been deposited in the Gene Expression Omnibus under accession code GSE69886.

Change history

  • 23 September 2015

    A minor change was made to the ‘Antibiotic treatment’ section in the Methods.

References

  1. Nauseef, W. M. & Borregaard, N. Neutrophils at work. Nature Immunol. 15, 602–611 (2014)

    Article  CAS  Google Scholar 

  2. Mayadas, T. N., Cullere, X. & Lowell, C. A. The multifaceted functions of neutrophils. Annu. Rev. Pathol. 9, 181–218 (2014)

    Article  CAS  Google Scholar 

  3. Taylor, P. R. et al. Activation of neutrophils by autocrine IL-17A-IL-17RC interactions during fungal infection is regulated by IL-6, IL-23, RORgammat and dectin-2. Nature Immunol. 15, 143–151 (2014)

    Article  CAS  Google Scholar 

  4. Woodfin, A. et al. The junctional adhesion molecule JAM-C regulates polarized transendothelial migration of neutrophils in vivo. Nature Immunol. 12, 761–769 (2011)

    Article  CAS  Google Scholar 

  5. Casanova-Acebes, M. et al. Rhythmic modulation of the hematopoietic niche through neutrophil clearance. Cell 153, 1025–1035 (2013)

    Article  CAS  Google Scholar 

  6. Rankin, S. M. The bone marrow: a site of neutrophil clearance. J. Leukoc. Biol. 88, 241–251 (2010)

    Article  CAS  Google Scholar 

  7. Martin, C. et al. Chemokines acting via CXCR2 and CXCR4 control the release of neutrophils from the bone marrow and their return following senescence. Immunity 19, 583–593 (2003)

    Article  CAS  Google Scholar 

  8. Stark, M. A. et al. Phagocytosis of apoptotic neutrophils regulates granulopoiesis via IL-23 and IL-17. Immunity 22, 285–294 (2005)

    Article  CAS  Google Scholar 

  9. Tanji-Matsuba, K. et al. Functional changes in aging polymorphonuclear leukocytes. Circulation 97, 91–98 (1998)

    Article  CAS  Google Scholar 

  10. Whyte, M. K., Meagher, L. C., MacDermot, J. & Haslett, C. Impairment of function in aging neutrophils is associated with apoptosis. J. Immunol. 150, 5124–5134 (1993)

    CAS  PubMed  Google Scholar 

  11. Hidalgo, A. et al. Heterotypic interactions enabled by polarized neutrophil microdomains mediate thromboinflammatory injury. Nature Med. 15, 384–391 (2009)

    Article  CAS  Google Scholar 

  12. Manwani, D. & Frenette, P. S. Vaso-occlusion in sickle cell disease: pathophysiology and novel targeted therapies. Blood 122, 3892–3898 (2013)

    Article  CAS  Google Scholar 

  13. Chiang, E. Y., Hidalgo, A., Chang, J. & Frenette, P. S. Imaging receptor microdomains on leukocyte subsets in live mice. Nature Methods 4, 219–222 (2007)

    Article  CAS  Google Scholar 

  14. Turhan, A., Weiss, L. A., Mohandas, N., Coller, B. S. & Frenette, P. S. Primary role for adherent leukocytes in sickle cell vascular occlusion: a new paradigm. Proc. Natl Acad. Sci. USA 99, 3047–3051 (2002)

    Article  ADS  CAS  Google Scholar 

  15. Scheiermann, C. et al. Adrenergic nerves govern circadian leukocyte recruitment to tissues. Immunity 37, 290–301 (2012)

    Article  CAS  Google Scholar 

  16. Gordy, C., Pua, H., Sempowski, G. D. & He, Y. W. Regulation of steady-state neutrophil homeostasis by macrophages. Blood 117, 618–629 (2011)

    Article  CAS  Google Scholar 

  17. 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)

    Article  CAS  Google Scholar 

  18. 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  ADS  CAS  Google Scholar 

  19. Mortha, A. et al. Microbiota-dependent crosstalk between macrophages and ILC3 promotes intestinal homeostasis. Science 343, 1249288 (2014)

    Article  Google Scholar 

  20. Brenchley, J. M. & Douek, D. C. Microbial translocation across the GI tract. Annu. Rev. Immunol. 30, 149–173 (2012)

    Article  CAS  Google Scholar 

  21. Khosravi, A. et al. Gut microbiota promote hematopoiesis to control bacterial infection. Cell Host Microbe 15, 374–381 (2014)

    Article  CAS  Google Scholar 

  22. Deshmukh, H. S. et al. The microbiota regulates neutrophil homeostasis and host resistance to Escherichia coli K1 sepsis in neonatal mice. Nature Med. 20, 524–530 (2014)

    Article  CAS  Google Scholar 

  23. Balmer, M. L. et al. Microbiota-derived compounds drive steady-state granulopoiesis via MyD88/TICAM signaling. J. Immunol. 193, 5273–5283 (2014)

    Article  CAS  Google Scholar 

  24. Clarke, T. B. et al. Recognition of peptidoglycan from the microbiota by Nod1 enhances systemic innate immunity. Nature Med. 16, 228–231 (2010)

    Article  CAS  Google Scholar 

  25. Furze, R. C. &. Rankin. S. M. The role of the bone marrow in neutrophil clearance under homeostatic conditions in the mouse. FASEB J. 22, 3111–3119 (2008)

    Article  CAS  Google Scholar 

  26. Sabroe, I. et al. Selective roles for Toll-like receptor (TLR)2 and TLR4 in the regulation of neutrophil activation and life span. J. Immunol. 170, 5268–5275 (2003)

    Article  CAS  Google Scholar 

  27. Magri, G. et al. Innate lymphoid cells integrate stromal and immunological signals to enhance antibody production by splenic marginal zone B cells. Nature Immunol. 15, 354–364 (2014)

    Article  CAS  Google Scholar 

  28. Li, P. et al. PAD4 is essential for antibacterial innate immunity mediated by neutrophil extracellular traps. J. Exp. Med. 207, 1853–1862 (2010)

    Article  CAS  Google Scholar 

  29. Chen, G. et al. Heme-induced neutrophil extracellular traps contribute to the pathogenesis of sickle cell disease. Blood 123, 3818–3827 (2014)

    Article  CAS  Google Scholar 

  30. Gaston, M. H. et al. Prophylaxis with oral penicillin in children with sickle cell anemia. A randomized trial. N. Engl. J. Med. 314, 1593–1599 (1986)

    Article  CAS  Google Scholar 

  31. Hanoun, M. et al. Acute myelogenous leukemia-induced sympathetic neuropathy promotes malignancy in an altered hematopoietic stem cell niche. Cell Stem Cell 15, 365–375 (2014)

    Article  CAS  Google Scholar 

  32. Hang, J. et al. 16S rRNA gene pyrosequencing of reference and clinical samples and investigation of the temperature stability of microbiome profiles. Microbiome 2, 31 (2014)

    Article  Google Scholar 

  33. Rothe, G. & Valet, G. Flow cytometric assays of oxidative burst activity in phagocytes. Methods Enzymol. 233, 539–548 (1994)

    Article  CAS  Google Scholar 

  34. Schnitt, S. J. et al. Myocardial fibrin deposition in experimental viral myocarditis that progresses to dilated cardiomyopathy. Circ. Res. 72, 914–920 (1993)

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We are grateful to C. Prophete and P. Ciero for expert technical assistance. We also thank E. Pamer (Memorial Sloan Kettering Cancer Center) for the gift of Tlr2−/− and Tlr4−/− mice; K. Ireland for assistance with the SCD patient study; Z. Chen for the taxonomic microbiota analysis; R. Ng for assistance with the germ-free mice; O. Uche and G. Wang for assistance in cell sorting; D. Reynolds and W. Tran for the microarray assay; and R. Sellers for histopathological analyses. This work was supported by a predoctoral fellowship from the American Heart Association (15PRE23010014 to D.Z.) and R01 grants from the National Institutes of Health (HL069438, DK056638, HL116340 to P.S.F.). Flow cytometry and cell sorting was supported by a Shared Facilities Award from the New York State Stem Cell Science (NYSTEM) Program.

Author information

Authors and Affiliations

Authors

Contributions

D.Z. designed and performed experiments, analysed results and wrote the manuscript; G.C., C.X., Y.K., R.B. and J.J. performed experiments and provided valuable inputs on the manuscript; A.M. provided LysM-cre/Myd88fl/fl and Csf2−/− mice and performed experiments; J.J.F. provided germ-free mice and performed experiments; D.M. provided human samples; C.S. and M.M. discussed data and provided valuable input on the manuscript; P.S.F. designed and supervised the study, discussed data and wrote the manuscript.

Corresponding author

Correspondence to Paul S. Frenette.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Extended data figures and tables

Extended Data Figure 1 Phenotypic and functional characterization of aged neutrophils.

a, Flow cytometry analysis of donor neutrophil ageing after adoptive transfer into recipients. Donor neutrophils gated by CD45.1+ and aged neutrophils gated by CD62LloCXCR4hi. b, Ageing and clearance kinetics of donor neutrophils after adoptive transfer into recipients (n = 3 mice). Left y axis, donor neutrophil number relative to the initial number of neutrophils transferred (black dashed line); right y axis, percentage of the aged subset in donor neutrophils (red line). c, d, MFIM analysis of Mac-1 activation of neutrophils harvested from wild-type or Selp−/− mice, labelled by PKH26 (red) and transferred into wild-type recipients. Scale bar, 10 μm. e, Plasma cytokine levels in wild-type and CD169-DTR mice 5 days after diphtheria toxin treatment (n = 5 mice). f, Percentages of adherent neutrophils that capture more than eight beads in diphtheria-toxin-treated wild-type and CD169-DTR mice (n = 8 mice). g, h, Flow cytometry analysis of surface marker expression (g), cell size (FSC) and granularity (SSC; h; n = 7 mice) on CD62Lhi young and CD62Llo aged neutrophils. i, CXCR4 expression levels on CD62Lhi young and CD62Llo aged neutrophils in wild-type, Selp−/−, and CD169-DTR mice (wild type, n = 13 mice; Selp−/−, n = 4 mice; CD169-DTR, n = 5 mice). Error bars, mean ± s.e.m. *P < 0.05, **P < 0.01, ***P < 0.001, data representing two or more independent experiments analysed with one-way ANOVA (b) or unpaired Student’s t-test (ei).

Extended Data Figure 2 Antibiotic treatment efficiently depletes and alters the composition of the microbiota.

a, Copy numbers of 16S ribosomal DNA in feces from control and antibiotics (ABX)-treated mice (n = 5 mice). b, Principal component analysis of the microbiome composition in control and ABX-treated mice (n = 5 mice). c, d, Percentage of each bacteria genus in total microbiome (n = 5 mice). Error bars, mean ± s.e.m. *P < 0.05, ***P < 0.001, data representing two or more independent experiments analysed with unpaired Student’s t-test (a, d) or permutational multivariate ANOVA (b).

Extended Data Figure 3 Microbiota-derived molecules regulate neutrophil homeostasis and ageing.

a, Numbers of circulating leukocyte subsets in control and antibiotics (ABX)-treated mice (n = 9 mice). b, Bone marrow cellularity and numbers of leukocyte subsets in the bone marrow of control and ABX-treated mice (n = 14 mice). c, Numbers of bone marrow haematopoietic stem and progenitor cells in control and ABX-treated mice (n = 9 mice). d, Spleen cellularity and numbers of leukocyte subsets in the spleen of control and ABX-treated mice (n = 7 mice). e, Flow cytometry analysis of neutrophil–LPS interactions in blood, bone marrow (BM) and spleen 1 h after LPS–FITC gavage (Ctrl, n = 4 mice; LPS–FITC, n = 5 mice). Histogram showing fluorescence intensity on neutrophils gated by Gr-1hi CD115lo SSAhi. f, Numbers of circulating aged neutrophils in control, ABX-treated, and ABX-treated mice fed with peptidoglycan (PGN) or mTriDAP (left, n = 11 (Ctrl), 9 (ABX), 9 (ABX+PGN) mice; right, n = 10 (Ctrl), 10 (ABX), 5 (ABX+mTriDAP) mice). Error bars, mean ± s.e.m. *P < 0.05, **P < 0.01, ***P < 0.001, data representing two or more independent experiments analysed with unpaired Student’s t-test.

Extended Data Figure 4 Neutrophil homeostasis is altered in germ-free mice.

a, Total white blood cell (WBC) counts and numbers of leukocyte subsets in blood of specific-pathogen-free (SPF) and germ-free (GF) mice (n = 5 mice). b, Total bone marrow (BM) cellularity and numbers of leukocyte subsets in the bone marrow of SPF and germ-free mice (SPF, n = 5 mice; germ-free, n = 4 mice). c, Total spleen cellularity and numbers of leukocyte subsets in the spleen of SPF and germ-free mice (SPF, n = 5 mice; germ-free, n = 4 mice). d, Copy numbers of 16S ribosomal DNA in feces from SPF mice, germ-free mice, germ-free mice reconstituted by fecal transplantation (GF-FT), and antibiotic-treated germ-free mice (GF-ABX; n = 5, 5, 5 and 4 mice, respectively). e, Numbers of total circulating neutrophils in SPF, germ-free, GF-FT, and GF-ABX mice (n = 5, 5, 5 and 3 mice, respectively). Error bars, mean ± s.e.m. *P < 0.05, **P < 0.01, ***P < 0.001, data representing two or more independent experiments analysed with unpaired Student’s t-tests.

Extended Data Figure 5 Microbiota-driven neutrophil ageing is independent of clearance mechanisms, and mediated by TLRs and Myd88 signalling.

a, Adhesion molecule expression on endothelial cells (ECs) in control and antibiotics (ABX)-treated mice (n = 4 mice). MFI, mean fluorescence intensity. b, Numbers of spleen and liver macrophages in control and ABX-treated mice (left, n = 7 mice; right, n = 4 mice). c, d, Numbers of bone marrow (BM) macrophages (c; n = 19, 19, 10, 10 mice, left to right) and circulating aged neutrophils (d; n = 12, 11, 10, 9 mice, left to right) in diphtheria toxin (DT)-treated control, ABX-treated mice, CD169-DTR, and ABX-treated CD169-DTR mice. e, Flow cytometry analysis of aged neutrophils in wild-type and LysM-cre/Myd88fl/fl mice (n = 12, 10 mice, respectively). f, Percentages of aged neutrophils in wild-type, Tlr4−/− and Tlr2−/− mice (n = 10, 10, 12 mice, respectively). g, Flow cytometry analysis of aged neutrophils in wild-type and Tnf−/− or Csf2−/− mice. h, Percentages of wild-type and LysM-cre/Myd88fl/fl or Tlr4−/− or Tlr2−/− neutrophils in total leukocyte population in chimaeric mice (n = 5 mice). i, Percentages of wild-type and LysM-cre/Myd88fl/fl or Tlr4−/− or Tlr2−/− neutrophils that capture more than eight beads in chimaeric mice (n = 5 mice). Error bars, mean ± s.e.m. *P < 0.05, **P < 0.01, ***P < 0.001, data representing two or more independent experiments analysed with unpaired Student’s t-test (af) or paired Student’s t-test (h, i).

Extended Data Figure 6 Microbiota depletion inhibits NET formation.

a, Flow cytometry analysis of aged neutrophils in isotype and anti-P/E-selectin antibody-treated mice (n = 6, 5 mice, respectively). b, ROS production of neutrophils from isotype and anti-P/E-selectin antibody-treated mice, as analysed by flow cytometry using dihydrorhodamine 123 (DHR-123; Isotype, n = 10; Abs (P/E), n = 11 mice). Grey lines, background fluorescence of neutrophils from both groups without LPS stimulation. ns, not significant. c, LPS-induced NET formation of neutrophils from control and antibiotics (ABX)-treated mice, as analysed by immunofluorescence staining of DNA (sytox orange), neutrophil elastase (NE) and citrullinated histone 3 (CitH3). Inset, isotype control. Scale bars, 10 μm. d, Quantification of NET formation of neutrophils from isotype and anti-P/E-selectin antibody-treated mice, or from control and ABX-treated mice (left, n = 4 (Isotype), 5 (Abs) mice; right, n = 4 mice). Error bars, mean ± s.e.m. *P < 0.05, **P < 0.01, ***P < 0.001, data representing two or more independent experiments analysed with unpaired Student’s t-test.

Extended Data Figure 7 Microbiota depletion benefits endotoxin-induced septic shock.

a, Representative images and quantification of in vivo NET formation in liver vasculature of control and antibiotics (ABX)-treated mice challenged with 30 mg kg −1 LPS (n = 3, 4 mice, respectively). Scale bar, 10 μm. b, Quantification of NET biomarkers, plasma nucleosome and DNA, in septic control and ABX-treated animals (n = 4 mice). c, d, Representative images showing CitH3+ neutrophil aggregates (c) and fibrin deposition associated with neutrophil aggregates (d) in septic liver of control and ABX-treated mice. Arrows, diffusive CitH3 and neutrophil elastase (NE) proteins. Insets, isotype controls. Scale bars, 10 μm. e, f, Numbers of CitH3+ neutrophils and neutrophil aggregates (e; left: n = 4 mice; right: n = 40 vessels from 4 mice) and quantification of fibrin deposition (f; n = 4 (Ctrl), 3 (ABX) mice) in septic liver of control and ABX-treated mice. g, Survival time of control, ABX-treated mice, and ABX-treated mice infused with 2 × 106 aged or young neutrophils in septic shock induced by 30 mg kg −1 LPS (n = 16, 10, 13, 6 mice, respectively). Error bars, mean ± s.e.m. *P < 0.05, **P < 0.01, data representing two or more independent experiments analysed with unpaired Student’s t-test (a, e (left), f), Mann–Whitney U-test (b, e (right)) or log-rank test (g).

Extended Data Figure 8 Microbiota depletion affects disease progression in sickle-cell disease.

a, Numbers of circulating leukocyte subsets in hemizygous control (SA), control SCD (SS Ctrl) and antibiotics-treated SCD (SS ABX) mice (SA: n = 8 mice; SS Ctrl: n = 9 mice; SS ABX: n = 9 mice). b, Haemodynamic parameters of mice analysed for neutrophil adhesion and integrin activation. c, Percentages of adherent neutrophils that capture more than eight beads in SA, SS Ctrl and SS ABX mice (n = 4, 3, 3 mice, respectively). d, Correlation between the survival times of SS control and SS ABX mice in acute vaso-occlusive crisis and their spleen weights. R2 = 0.45. e, Scoring of liver damage, liver fibrosis, inflammation and necrosis in SS control and SS ABX mice (n = 8, 9 mice, respectively). f, Flow cytometry analysis of aged neutrophils in healthy controls, SCD patients (SS), and SCD patients on penicillin V prophylaxis (SS-PV). g, Demographics of human subjects analysed for aged neutrophil numbers. ACS, acute chest syndrome; VOC, vaso-occlusive crisis. h, Aged neutrophil numbers in SCD patients grouped by age, gender, hydroxyurea (HU) and penicillin V (Pen V) treatment (Ctrl, n = 9 subjects; SS, n = 23 subjects; SS-PV, n = 11 subjects). Error bars, mean ± s.e.m. *P < 0.05, **P < 0.01, ***P < 0.001, data representing two or more independent experiments analysed with unpaired Student’s t-test (a, c, h) or Mann–Whitney U-test (e).

Extended Data Table 1 Pathways selected for the analysis of neutrophil functions
Extended Data Table 2 Gene set enrichment analysis of selected pathways in aged and activated neutrophils

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zhang, D., Chen, G., Manwani, D. et al. Neutrophil ageing is regulated by the microbiome. Nature 525, 528–532 (2015). https://doi.org/10.1038/nature15367

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature15367

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

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