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:

Neutrophils prime a long-lived effector macrophage phenotype that mediates accelerated helminth expulsion

Abstract

We examined the role of innate cells in acquired resistance to the natural murine parasitic nematode, Nippostrongylus brasiliensis. Macrophages obtained from lungs as late as 45 d after N. brasiliensis inoculation were able to transfer accelerated parasite clearance to naive recipients. Primed macrophages adhered to larvae in vitro and triggered increased mortality of parasites. Neutrophil depletion in primed mice abrogated the protective effects of transferred macrophages and inhibited their in vitro binding to larvae. Neutrophils in parasite-infected mice showed a distinct transcriptional profile and promoted alternatively activated M2 macrophage polarization through secretory factors including IL-13. Differentially activated neutrophils in the context of a type 2 immune response therefore prime a long-lived effector macrophage phenotype that directly mediates rapid nematode damage and clearance.

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: Eosinophils and macrophages surround parasitic larvae in the lung, and type 2 related cytokines are upregulated shortly after secondary inoculation with N. brasiliensis.
Figure 2: Macrophages from mice primed with N. brasiliensis directly damage larvae.
Figure 3: IL-4R signaling is required for resistance to N. brasiliensis mediated by primed macrophages.
Figure 4: Neutrophils are required for differentiation of effector M2 lung macrophages after primary inoculation with N. brasiliensis.
Figure 5: Depletion of neutrophils impairs effector lung macrophage polarization after primary inoculation.
Figure 6: N.brasiliensis–primed neutrophils show a distinct profile of gene expression.
Figure 7: N.brasiliensis–primed neutrophils polarize to an alternatively activated neutrophil phenotype that promotes M2 macrophage differentiation in vitro.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

References

  1. King, C.H. Health metrics for helminthic infections. Adv. Parasitol. 73, 51–69 (2010).

    Article  PubMed  Google Scholar 

  2. Salgame, P., Yap, G.S. & Gause, W.C. Effect of helminth-induced immunity on infections with microbial pathogens. Nat. Immunol. 14, 1118–1126 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Gaze, S. et al. Characterising the mucosal and systemic immune responses to experimental human hookworm infection. PLoS Pathog. 8, e1002520 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Anthony, R.M., Rutitzky, L.I., Urban, J.F. Jr., Stadecker, M.J. & Gause, W.C. Protective immune mechanisms in helminth infection. Nat. Rev. Immunol. 7, 975–987 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Gause, W.C., Wynn, T.A. & Allen, J.E. Type 2 immunity and wound healing: evolutionary refinement of adaptive immunity by helminths. Nat. Rev. Immunol. 13, 607–614 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Harvie, M. et al. The lung is an important site for priming CD4 T-cell-mediated protective immunity against gastrointestinal helminth parasites. Infect. Immun. 78, 3753–3762 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Maizels, R.M. et al. Immune modulation and modulators in Heligmosomoides polygyrus infection. Exp. Parasitol. 132, 76–89 (2012).

    Article  CAS  PubMed  Google Scholar 

  8. Zhao, A. et al. Dependence of IL-4, IL-13, and nematode-induced alterations in murine small intestinal smooth muscle contractility on Stat6 and enteric nerves. J. Immunol. 171, 948–954 (2003).

    Article  CAS  PubMed  Google Scholar 

  9. Harris, N. & Gause, W.C. To B or not to B: B cells and the Th2-type immune response to helminths. Trends Immunol. 32, 80–88 (2011).

    Article  CAS  PubMed  Google Scholar 

  10. Finkelman, F.D. et al. Interleukin-4- and interleukin-13-mediated host protection against intestinal nematode parasites. Immunol. Rev. 201, 139–155 (2004).

    Article  CAS  PubMed  Google Scholar 

  11. Herbert, D.R. et al. Intestinal epithelial cell secretion of RELM-β protects against gastrointestinal worm infection. J. Exp. Med. 206, 2947–2957 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Knott, M.L. et al. Impaired resistance in early secondary Nippostrongylus brasiliensis infections in mice with defective eosinophilopoeisis. Int. J. Parasitol. 37, 1367–1378 (2007).

    Article  CAS  PubMed  Google Scholar 

  13. Anthony, R.M. et al. Memory TH2 cells induce alternatively activated macrophages to mediate protection against nematode parasites. Nat. Med. 12, 955–960 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Yang, Z. et al. Macrophages as IL-25/IL-33-responsive cells play an important role in the induction of type 2 immunity. PLoS ONE 8, e59441 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Morimoto, M. et al. Peripheral CD4 T cells rapidly accumulate at the host: parasite interface during an inflammatory Th2 memory response. J. Immunol. 172, 2424–2430 (2004).

    Article  CAS  PubMed  Google Scholar 

  16. Bonne-Année, S. et al. Human and mouse macrophages collaborate with neutrophils to kill larval Strongyloides stercoralis. Infect. Immun. 81, 3346–3355 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  17. Harvie, M., Camberis, M. & Le Gros, G. Development of CD4 T cell dependent immunity against N. brasiliensis infection. Frontiers Immunol. 4, 74 (2013).

    Article  CAS  Google Scholar 

  18. Katona, I.M., Urban, J.F. Jr. & Finkelman, F.D. The role of L3T4+ and Lyt-2+ T cells in the IgE response and immunity to Nippostrongylus brasiliensis. J. Immunol. 140, 3206–3211 (1988).

    CAS  PubMed  Google Scholar 

  19. Liu, Q. et al. B cells have distinct roles in host protection against different nematode parasites. J. Immunol. 184, 5213–5223 (2010).

    Article  CAS  PubMed  Google Scholar 

  20. Patel, N., Kreider, T., Urban, J.F. Jr. & Gause, W.C. Characterisation of effector mechanisms at the host:parasite interface during the immune response to tissue-dwelling intestinal nematode parasites. Int. J. Parasitol. 39, 13–21 (2009).

    Article  CAS  PubMed  Google Scholar 

  21. Chen, F. et al. An essential role for TH2-type responses in limiting acute tissue damage during experimental helminth infection. Nat. Med. 18, 260–266 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Smith, K.A. et al. Type 2 innate immunity in helminth infection is induced redundantly and acts autonomously following CD11c+ cell depletion. Infect. Immun. 80, 3481–3489 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Obata-Ninomiya, K. et al. The skin is an important bulwark of acquired immunity against intestinal helminths. J. Exp. Med. 210, 2583–2589 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Kreider, T., Anthony, R.M., Urban, J.F. Jr. & Gause, W.C. Alternatively activated macrophages in helminth infections. Curr. Opin. Immunol. 19, 448–453 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Dobrovolskaia, M.A. & Vogel, S.N. Toll receptors, CD14, and macrophage activation and deactivation by LPS. Microbes Infect. 4, 903–914 (2002).

    Article  CAS  PubMed  Google Scholar 

  26. Murray, P.J. & Wynn, T.A. Protective and pathogenic functions of macrophage subsets. Nat. Rev. Immunol. 11, 723–737 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Morris, S.M. Jr. Recent advances in arginine metabolism: roles and regulation of the arginases. Br. J. Pharmacol. 157, 922–930 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Ckless, K. et al. Inhibition of arginase activity enhances inflammation in mice with allergic airway disease, in association with increases in protein S-nitrosylation and tyrosine nitration. J. Immunol. 181, 4255–4264 (2008).

    Article  CAS  PubMed  Google Scholar 

  29. Herbert, D.R. et al. Arginase I suppresses IL-12/IL-23p40-driven intestinal inflammation during acute schistosomiasis. J. Immunol. 184, 6438–6446 (2010).

    Article  CAS  PubMed  Google Scholar 

  30. Bonne-Année, S., Hess, J.A. & Abraham, D. Innate and adaptive immunity to the nematode Strongyloides stercoralis in a mouse model. Immunol. Res. 51, 205–214 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  31. Gong, D. et al. TGFβ signaling plays a critical role in promoting alternative macrophage activation. BMC Immunol. 13, 31 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Oo-puthinan, S. et al. The amino acids involved in the distinct carbohydrate specificities between macrophage galactose-type C-type lectins 1 and 2 (CD301a and b) of mice. Biochim. Biophys. Acta 1780, 89–100 (2008).

    Article  CAS  PubMed  Google Scholar 

  33. Staples, K.J. et al. Phenotypic characterization of lung macrophages in asthmatic patients: overexpression of CCL17. J. Allergy Clin. Immunol. 130, 1404.e7–1412.e7 (2012).

    Article  CAS  Google Scholar 

  34. Becker, H.M. et al. α1β1 integrin-mediated adhesion inhibits macrophage exit from a peripheral inflammatory lesion. J. Immunol. 190, 4305–4314 (2013).

    Article  CAS  PubMed  Google Scholar 

  35. Huang, D.W., Sherman, B.T. & Lempicki, R.A. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat. Protoc. 4, 44–57 (2009).

    Article  CAS  Google Scholar 

  36. Durrant, W.E. & Dong, X. Systemic acquired resistance. Annu. Rev. Phytopathol. 42, 185–209 (2004).

    Article  CAS  PubMed  Google Scholar 

  37. Kurtz, J. Specific memory within innate immune systems. Trends Immunol. 26, 186–192 (2005).

    Article  CAS  PubMed  Google Scholar 

  38. Netea, M.G., Quintin, J. & van der Meer, J.W. Trained immunity: a memory for innate host defense. Cell Host Microbe 9, 355–361 (2011).

    Article  CAS  PubMed  Google Scholar 

  39. Didierlaurent, A. et al. Sustained desensitization to bacterial Toll-like receptor ligands after resolution of respiratory influenza infection. J. Exp. Med. 205, 323–329 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Reece, J.J., Siracusa, M.C. & Scott, A.L. Innate immune responses to lung-stage helminth infection induce alternatively activated alveolar macrophages. Infect. Immun. 74, 4970–4981 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Marsland, B.J., Kurrer, M., Reissmann, R., Harris, N.L. & Kopf, M. Nippostrongylus brasiliensis infection leads to the development of emphysema associated with the induction of alternatively activated macrophages. Eur. J. Immunol. 38, 479–488 (2008).

    Article  CAS  PubMed  Google Scholar 

  42. Satoh, T. et al. The Jmjd3-Irf4 axis regulates M2 macrophage polarization and host responses against helminth infection. Nat. Immunol. 11, 936–944 (2010).

    Article  CAS  PubMed  Google Scholar 

  43. Monticelli, S. & Natoli, G. Short-term memory of danger signals and environmental stimuli in immune cells. Nat. Immunol. 14, 777–784 (2013).

    Article  CAS  PubMed  Google Scholar 

  44. Mantovani, A., Cassatella, M.A., Costantini, C. & Jaillon, S. Neutrophils in the activation and regulation of innate and adaptive immunity. Nat. Rev. Immunol. 11, 519–531 (2011).

    Article  CAS  PubMed  Google Scholar 

  45. Galioto, A.M. et al. Role of eosinophils and neutrophils in innate and adaptive protective immunity to larval strongyloides stercoralis in mice. Infect. Immun. 74, 5730–5738 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Tsuda, Y. et al. Three different neutrophil subsets exhibited in mice with different susceptibilities to infection by methicillin-resistant Staphylococcus aureus. Immunity 21, 215–226 (2004).

    Article  CAS  PubMed  Google Scholar 

  47. Soehnlein, O., Weber, C. & Lindbom, L. Neutrophil granule proteins tune monocytic cell function. Trends Immunol. 30, 538–546 (2009).

    Article  CAS  PubMed  Google Scholar 

  48. Rosenthal, L.A., Sutterwala, F.S., Kehrli, M.E. & Mosser, D.M. Leishmania major-human macrophage interactions: cooperation between Mac-1 (CD11b/CD18) and complement receptor type 1 (CD35) in promastigote adhesion. Infect. Immun. 64, 2206–2215 (1996).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Dent, L.A. et al. Interleukin-5 transgenic mice show enhanced resistance to primary infections with Nippostrongylus brasiliensis but not primary infections with Toxocara canis. Infect. Immun. 67, 989–993 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Voehringer, D. Protective and pathological roles of mast cells and basophils. Nat. Rev. Immunol. 13, 362–375 (2013).

    Article  CAS  PubMed  Google Scholar 

  51. Pesce, J.T. et al. Neutrophils clear bacteria associated with parasitic nematodes augmenting the development of an effective Th2-type response. J. Immunol. 180, 464–474 (2008).

    Article  CAS  PubMed  Google Scholar 

  52. Tusher, V.G., Tibshirani, R. & Chu, G. Significance analysis of microarrays applied to the ionizing radiation response. Proc. Natl. Acad. Sci. USA 98, 5116–5121 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Liu, Z. et al. IL-2 and autocrine IL-4 drive the in vivo development of antigen-specific Th2 T cells elicited by nematode parasites. J. Immunol. 174, 2242–2249 (2005).

    Article  CAS  PubMed  Google Scholar 

  54. Gause, W.C. & Adamovicz, J. The use of the PCR to quantitate gene expression. PCR Methods Appl. 3, S123–S135 (1994).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

Mention of trade names or commercial products in this publication is solely for the purpose of providing specific information and does not imply recommendation or endorsement by the US Department of Agriculture (USDA). The USDA is an equal opportunity provider and employer. Supported by National Institutes of Health (1R01AI107588 to W.C.G. and C.C.K.; 5R01AI031678 to W.C.G.; R00AI085035 to C.C.K.) and the Agricultural Research Service of the USDA (1235-51000-058 to J.F.U. Jr.).

Author information

Authors and Affiliations

Authors

Contributions

W.C.G., F.C., C.C.K. and J.F.U. Jr. designed the experiments; F.C., W.W., A.M., J.F.C., E.C. and N.P. did the experiments; J.L.B. prepared and provided the BEC; and W.C.G., F.C., W.W. and C.C.K. wrote the manuscript.

Corresponding author

Correspondence to William C Gause.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Nippostrongylus brasiliensis (Nb) migratory larvae induce long-term changes in immune cell populations.

At specific timepoints after Nb inoculation, mice were examined for changes in lung immune cell populations. (a) Representative haematoxylin and eosin staining of formalin-fixed lung sections (left small panels, scale bars, 20 μm; right panel, scale bar, 2 μm). Arrows indicate macrophages (Mac) and eosinophils (Eo), BV=blood vessel. (b) The numbers of lung immune cell populations obtained from a single lung lobule were determined by flow cytometry assay using antibodies specific for macrophages (F4/80+, CD11cvar, MHCIIint-hi), eosinophils (Siglec F+, MHCIIlo, CD11clo), neutrophils (Ly6G+), CD4+ T cells (CD3+CD4+), and CD8+ T cells (CD3+CD8+). Individual timepoints represent pooled samples from five mice per treatment group. (c) At three months after primary inoculation, mice were challenged and then examined at days two, four, and seven after secondary inoculation. The numbers of lung innate immune cells obtained from a single lung lobule was determined by flow cytometry (1° Nb, primary infection; 2° Nb, secondary infection). Individual timepoints represent pooled samples from five mice per treatment group. Similar results were obtained in two independent experiments.

Supplementary Figure 2 N. brasiliensis (Nb)-primed macrophages promote type 2 immune responses in lung.

(a) Macrophages (F4/80+, MHCII+, CD11c var) were electronically sorted from lung of Nb-inoculated mice and purity, including depletion of eosinophils, confirmed by G-W staining. (b) Nb-inoculated mice were sacrificed at day 7 and lung macrophages were sorted, labeled with CFSE, and transferred to recipient mice, to confirm the presence of transferred macrophages in the lung at day 3; (c) Total numbers of macrophages (F4/80+, CD11cvar, MHCIIint-hi), eosinophils (Siglec F+, MHCIIlo, CD11clo), and neutrophils (Ly6G+) in single lung lobule at day 2 and 3 after inoculation of recipient mice with transferred primed macrophages or control mice not receiving macrophage transfer. Individual timepoints represent values for pooled samples from 5 mice/treatment group; (d) and (e) as in (c), but lung tissue mRNA levels of: (d) Il4, Il5, Il13, and Il10 and (e) M2/M1 markers (Arg1, Chi3l3, and Nos2), as determined by qPCR. Gene expression is presented as the fold increase over naive WT controls after normalization to 18sRNA. Data shown for mRNA levels are the mean and SEM from five individual mice per group and are representative of two independent experiments. (f) Lung macrophages(F4/80hiMHCIIint-hiCD11cvar) were electronically sorted from mice at day 7 after Nb inoculation or were isolated from naive mice, transferred to recipient mice and two later inoculated with Nb; 2 days after Nb inoculatoin parasites were enumerated in the skin, as previously described23. (g) Lung macrophages were electronically sorted at day 7 after Nb inoculation or were isolated from naive mice, transferred to recipient mice, which were inoculated with Nb 2 days later. Five days after Nb inoculation parasites were enumerated in the gut. (h) Wild type and Jh–/– donor mice were inoculated with Nb and, at day 45 after inoculation, electronically sorted macrophages were transferred to naive recipient mice which were inoculated with Nb two days later. parasites were enumerated at five days post inoculation in the gut. (f-h) The mean and SEM of 5 individual mice per treatment group is shown and each experiment was repeated two times with similar results. *p<0.05.

Supplementary Figure 3 Effects of neutrophil depletion on lung and blood immune cell populations and on lung cytokine gene expression.

(a and b) Cell subpopulations at day 2 from the lung and blood of mice administered neutrophil-depleting anti-Ly6G antibody or isotype control at days -1 after N. brasiliensis inoculation. Data shown are mean of pooled samples from 3 mice per group, and this experiment was repeated two times with similar results. (c and d) Lung tissue from a single lung lobule was analyzed for changes in cytokine and M2 marker expression, by qPCR, at day 7 (c) or at day 1(d) after Nb inoculation and treatment with neutrophil depleting anti-Ly6G antibody or isotype control day -1, +3 (c) or day -1 (d). Gene expression is presented as the fold increase over naive WT controls after normalization to 18sRNA. Data shown are the mean and SEM from five individual mice per group and are representative of two independent experiments. *p<0.01

Supplementary Figure 4 Analysis of sorted neutrophils and the impact of CD4+ T cells on neutrophil recruitment to the lung during N. brasiliensis infection.

(a-b) Mice were infected with N. brasiliensis for 2 days and lung cell suspension collected for further analysis. (a) Gated CD11b+, LY6G+ neutrophil population selected for FACS sorting. (b) Frequency of basophils (FcɛRI+DX5+) was examined in total lung cells, the gated CD11b+Ly6G+ cell population, and electronically sorted CD11b+Ly6G+ cells. (c) Antibodies against CD4+ T cells were administered to BALB/c mice (800 µg, i.p.) 2 days before N. brasiliensis infection. Mice were sacrificed at day 2 and lung CD4+ T cells (CD3+CD4+) and neutrophils (CD11b+Ly6G+) were examined by flow analysis. Data were obtained from pool samples from 3 mice per group, and this experiment was repeated 3 times with similar results.

Supplementary Figure 5 Neutrophils, but not macrophages and CD4+ T cells are the source of IL-13 and IL-33 in the lung during N. brasiliensis infection at an early time point.

Mice were infected with N. brasiliensis for 2 days and then sacrificed. Whole lung cells from N. brasiliensis-infected mice at day 2 were stimulated by PMA/ion then stained with antibodies against cell surface marker CD3, CD4, CD11b, Ly6G, F4/80, and cytokines IL-13, IL-33. The expression of IL-13 and IL-33 on neutrophils, macrophages, or CD4+ T cells was examined by flow cytometry analysis. The flow plot is a representative of three mice per group, and the cell number graph is shown as mean and SEM from 3 individual mice per group and are representative of two independent experiments. *p<0.01

Supplementary Figure 6 Model of macrophage-mediated acquired resistance to the nematode parasite N. brasiliensis (Nb).

Initial Nb infection results in neutrophil infiltration of the lung within 2 days after inoculation. By day 4, the type 2 immune response develops, which includes alternatively activated neutrophil (N2) that then help promote the development of an anti-helminth effector macrophage phenotype, which develops by 7 days after inoculation and is sustained as a long-lived persistent phenotype capable of mediating accelerated parasite killing after secondary inoculation. These effector macrophages adhere to parasites through ITGAM-dependent mechanisms and mediate larval parasite damage and killing through mechanisms dependent on Arg1.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–6 (PDF 4211 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Chen, F., Wu, W., Millman, A. et al. Neutrophils prime a long-lived effector macrophage phenotype that mediates accelerated helminth expulsion. Nat Immunol 15, 938–946 (2014). https://doi.org/10.1038/ni.2984

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ni.2984

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