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:

Acidic chitinase primes the protective immune response to gastrointestinal nematodes

Abstract

Acidic mammalian chitinase (AMCase) is known to be induced by allergens and helminths, yet its role in immunity is unclear. Using AMCase-deficient mice, we show that AMCase deficiency reduced the number of group 2 innate lymphoid cells during allergen challenge but was not required for establishment of type 2 inflammation in the lung in response to allergens or helminths. In contrast, AMCase-deficient mice showed a profound defect in type 2 immunity following infection with the chitin-containing gastrointestinal nematodes Nippostrongylus brasiliensis and Heligmosomoides polygyrus bakeri. The impaired immunity was associated with reduced mucus production and decreased intestinal expression of the signature type 2 response genes Il13, Chil3, Retnlb, and Clca1. CD103+ dendritic cells, which regulate T cell homing, were also reduced in mesenteric lymph nodes of infected AMCase-deficient mice. Thus, AMCase functions as a critical initiator of protective type 2 responses to intestinal nematodes but is largely dispensable for allergic responses in the lung.

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: AMCase-deficient mice develop acute HDM-induced lung allergy despite diminished ILC2s.
Figure 2: Chronic HDM induces similar allergy in the lungs of wild-type and AMCase-deficient mice.
Figure 3: AMCase-deficient mice develop S. mansoni egg–induced lung granulomas.
Figure 4: AMCase is critical for type 2–mediated protection against N. brasiliensis.
Figure 5: Type 2 immunity against H. p. bakeri is impaired in AMCase-deficient mice.

Similar content being viewed by others

Accession codes

Primary accessions

NCBI Reference Sequence

References

  1. Lenardon, M.D., Munro, C.A. & Gow, N.A. Chitin synthesis and fungal pathogenesis. Curr. Opin. Microbiol. 13, 416–423 (2010).

    Article  CAS  Google Scholar 

  2. Neville, A.C., Parry, D.A. & Woodhead-Galloway, J. The chitin crystallite in arthropod cuticle. J. Cell Sci. 21, 73–82 (1976).

    CAS  PubMed  Google Scholar 

  3. Veronico, P. et al. Nematode chitin synthases: gene structure, expression and function in Caenorhabditis elegans and the plant parasitic nematode Meloidogyne artiellia. Mol. Genet. Genomics 266, 28–34 (2001).

    Article  CAS  Google Scholar 

  4. Foster, J.M., Zhang, Y., Kumar, S. & Carlow, C.K. Parasitic nematodes have two distinct chitin synthases. Mol. Biochem. Parasitol. 142, 126–132 (2005).

    Article  CAS  Google Scholar 

  5. Boot, R.G. et al. Identification of a novel acidic mammalian chitinase distinct from chitotriosidase. J. Biol. Chem. 276, 6770–6778 (2001).

    Article  CAS  Google Scholar 

  6. Renkema, G.H., Boot, R.G., Muijsers, A.O., Donker-Koopman, W.E. & Aerts, J.M. Purification and characterization of human chitotriosidase, a novel member of the chitinase family of proteins. J. Biol. Chem. 270, 2198–2202 (1995).

    Article  CAS  Google Scholar 

  7. Zhu, Z. et al. Acidic mammalian chitinase in asthmatic Th2 inflammation and IL-13 pathway activation. Science 304, 1678–1682 (2004).

    Article  CAS  Google Scholar 

  8. Reese, T.A. et al. Chitin induces accumulation in tissue of innate immune cells associated with allergy. Nature 447, 92–96 (2007).

    Article  CAS  Google Scholar 

  9. Ramanathan, M. Jr., Lee, W.K. & Lane, A.P. Increased expression of acidic mammalian chitinase in chronic rhinosinusitis with nasal polyps. Am. J. Rhinol. 20, 330–335 (2006).

    Article  Google Scholar 

  10. Fitz, L.J. et al. Acidic mammalian chitinase is not a critical target for allergic airway disease. Am. J. Respir. Cell Mol. Biol. 46, 71–79 (2012).

    Article  CAS  Google Scholar 

  11. Kim, L.K. et al. AMCase is a crucial regulator of type 2 immune responses to inhaled house dust mites. Proc. Natl. Acad. Sci. USA 112, E2891–E2899 (2015).

    Article  CAS  Google Scholar 

  12. Wynn, T.A. Type 2 cytokines: mechanisms and therapeutic strategies. Nat. Rev. Immunol. 15, 271–282 (2015).

    Article  CAS  Google Scholar 

  13. Nair, M.G. et al. Chitinase and Fizz family members are a generalized feature of nematode infection with selective upregulation of Ym1 and Fizz1 by antigen-presenting cells. Infect. Immun. 73, 385–394 (2005).

    Article  CAS  Google Scholar 

  14. Yagi, R. et al. The transcription factor GATA3 is critical for the development of all IL-7Rα-expressing innate lymphoid cells. Immunity 40, 378–388 (2014).

    Article  CAS  Google Scholar 

  15. Bierbaum, S. et al. Polymorphisms and haplotypes of acid mammalian chitinase are associated with bronchial asthma. Am. J. Respir. Crit. Care Med. 172, 1505–1509 (2005).

    Article  Google Scholar 

  16. Wiesner, D.L. et al. Chitin recognition via chitotriosidase promotes pathologic type-2 helper T cell responses to cryptococcal infection. PLoS Pathog. 11, e1004701 (2015).

    Article  Google Scholar 

  17. Seibold, M.A. et al. Chitotriosidase is the primary active chitinase in the human lung and is modulated by genotype and smoking habit. J. Allergy Clin. Immunol. 122, 944–950 (2008).

    Article  CAS  Google Scholar 

  18. Sandler, N.G., Mentink-Kane, M.M., Cheever, A.W. & Wynn, T.A. Global gene expression profiles during acute pathogen-induced pulmonary inflammation reveal divergent roles for Th1 and Th2 responses in tissue repair. J. Immunol. 171, 3655–3667 (2003).

    Article  CAS  Google Scholar 

  19. Albonico, M. et al. Controlling soil-transmitted helminthiasis in pre-school-age children through preventive chemotherapy. PLoS Negl. Trop. Dis. 2, e126 (2008).

    Article  Google Scholar 

  20. Zaph, C., Cooper, P.J. & Harris, N.L. Mucosal immune responses following intestinal nematode infection. Parasite Immunol. 36, 439–452 (2014).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  22. Sutherland, T.E. et al. Chitinase-like proteins promote IL-17-mediated neutrophilia in a tradeoff between nematode killing and host damage. Nat. Immunol. 15, 1116–1125 (2014).

    Article  CAS  Google Scholar 

  23. Sabo-Attwood, T. et al. Gene expression profiles reveal increased mClca3 (Gob5) expression and mucin production in a murine model of asbestos-induced fibrogenesis. Am. J. Pathol. 167, 1243–1256 (2005).

    Article  CAS  Google Scholar 

  24. Hasnain, S.Z. et al. Muc5ac: a critical component mediating the rejection of enteric nematodes. J. Exp. Med. 208, 893–900 (2011).

    Article  CAS  Google Scholar 

  25. McKenzie, G.J., Fallon, P.G., Emson, C.L., Grencis, R.K. & McKenzie, A.N. Simultaneous disruption of interleukin (IL)-4 and IL-13 defines individual roles in T helper cell type 2-mediated responses. J. Exp. Med. 189, 1565–1572 (1999).

    Article  CAS  Google Scholar 

  26. Urban, J.F. Jr. et al. IL-13, IL-4Rα, and Stat6 are required for the expulsion of the gastrointestinal nematode parasite Nippostrongylus brasiliensis. Immunity 8, 255–264 (1998).

    Article  CAS  Google Scholar 

  27. Johansson-Lindbom, B. et al. Functional specialization of gut CD103+ dendritic cells in the regulation of tissue-selective T cell homing. J. Exp. Med. 202, 1063–1073 (2005).

    Article  CAS  Google Scholar 

  28. Strobel, S., Roswag, A., Becker, N.I., Trenczek, T.E. & Encarnação, J.A. Insectivorous bats digest chitin in the stomach using acidic mammalian chitinase. PLoS One 8, e72770 (2013).

    Article  CAS  Google Scholar 

  29. Iwasaki, A. & Medzhitov, R. Control of adaptive immunity by the innate immune system. Nat. Immunol. 16, 343–353 (2015).

    Article  CAS  Google Scholar 

  30. Jaensson, E. et al. Small intestinal CD103+ dendritic cells display unique functional properties that are conserved between mice and humans. J. Exp. Med. 205, 2139–2149 (2008).

    Article  CAS  Google Scholar 

  31. Katona, I.M., Urban, J.F. Jr., Scher, I., Kanellopoulos-Langevin, C. & Finkelman, F.D. Induction of an IgE response in mice by Nippostrongylus brasiliensis: characterization of lymphoid cells with intracytoplasmic or surface IgE. J. Immunol. 130, 350–356 (1983).

    CAS  PubMed  Google Scholar 

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

    Article  CAS  Google Scholar 

Download references

Acknowledgements

This research was supported by the Intramural Research Program of the National Institutes of Health, National Institute of Allergy and Infectious Disease. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript. We thank MedImmune for generating the anti-AMCase rabbit sera, C. Mainhart for genotyping, T. Gieseck and K. Kindrachuk for discussions, and the animal care staffs of Buildings 50 and 14BS at the US National Institutes of Health's Bethesda, Maryland campus for the conscientious care of mice.

Author information

Authors and Affiliations

Authors

Contributions

K.M.V., T.R.R. and T.A.W. conceived and designed the experiments; K.M.V., A.D.S., K.M.H., L.A.B., R.W.T., S.W., J.F.U., R.d.Q.P. and J.S. performed the experiments; I.M. and K.B. performed immunofluorescence techniques; K.M.V., T.R.R., A.D.S., A.W.C., L.B., L.A.B., M.M.-K., T.A.W., J.F.U. and R.d.Q.P. analyzed the data; A.D.S., A.W.C., I.M., J.F.U. and L.J.F. contributed reagents; K.M.V., T.R.R. and T.A.W. wrote the paper.

Corresponding author

Correspondence to Thomas A Wynn.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Acute model of papain-induced type 2 allergic inflammation in the lung.

(a) Quantitative PCR analysis of gene expression in lung tissue from wild type mice or AMCase-KO mice sensitized and challenged intranasally with PBS (n = 3 mice per genotype) or papain (n =5 mice per genotype) expressed relative to water-treated wild type. (b) Quantification of eosinophils in lung tissue of mice in a (PBS: n = 3, papain: n = 5). (c) Intracellular cytokine analysis of lung tissue lymphocytes of mice in a (PBS: n = 3, HDM: n = 5). (d) Quantification of eosinophils in BALF of mice in a (PBS: n = 3, HDM: n = 5). (e) Intracellular cytokine analysis of BALF lymphocytes of mice in a (PBS: n = 3, HDM: n = 5). Error bars represent standard error of the mean, and each data point represents a value for an individual mouse.

Supplementary Figure 2 Acute model of SEA-induced type 2 allergic inflammation in the lung.

(a) Quantitative PCR analysis of gene expression in lung tissue from wild type mice or AMCase-KO mice sensitized and challenged intratracheally with PBS (n = 3 mice per genotype) or schistosoma egg antigen (SEA) (n = 9 mice per genotype) expressed relative to PBS-treated wild type. (b) Quantification of leukocytes in BALF of mice in a (PBS: n = 3, SEA: n = 6). (c) Intracellular cytokine analysis of lung tissue lymphocytes of mice in a (PBS: n = 3, WT/SEA: n = 9, KO/SEA: n = 5). (d) Quantification of eosinophils in lung tissue of mice in a (PBS: n = 3, WT/SEA: n = 9, KO/SEA: n = 5). Data are representative of three experiments with similar results. Error bars represent standard error of the mean, and each data point represents a value for an individual mouse.

Supplementary Figure 3 Gene expression in lung tissue during N. brasiliensis infection.

Quantitative PCR analysis of gene expression in lung tissue from wild type mice (n = 7) or AMCase-KO mice (n = 7) 8 days after primary infection with N. brasiliensis. Data shown relative to expression in lung tissue from naïve mice. *P<0.05 (Student’s t-test). Data are representative of two experiments with similar results. Error bars represent standard error of the mean, and each data point represents a value for an individual mouse.

Supplementary Figure 4 Wild type and AMCase-deficient T cells are equally competent at clearing H.p. bakeri infection.

Quantification of H.p. bakeri worms recovered from intestines of TCRα KO mice that were recipients of 2x105 T cells from previously infected wild type mice (triangles, n = 8), 2x105 T cells from previously infected AMCase-KO mice (squares, n = 3), or no T cells (circles, n = 4).

Supplementary Figure 5 Il33 expression in small intestine tissue 3.5 days after primary H. p. bakeri infection.

Quantitative PCR analysis of Il33 expression in proximal small intestine tissue from wild type mice or AMCase-KO mice 3.5 days post-infection (n = 9 per genotype) or from uninfected wild type or AMCase-KO (n = 3 per genotype). Error bars represent standard error of the mean, and each data point represents a value for an individual mouse.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–5 (PDF 1073 kb)

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Vannella, K., Ramalingam, T., Hart, K. et al. Acidic chitinase primes the protective immune response to gastrointestinal nematodes. Nat Immunol 17, 538–544 (2016). https://doi.org/10.1038/ni.3417

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ni.3417

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