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:

A role for the ITAM signaling module in specifying cytokine-receptor functions

Subjects

Abstract

Diverse cellular responses to external cues are controlled by a small number of signal-transduction pathways, but how the specificity of functional outcomes is achieved remains unclear. Here we describe a mechanism for signal integration based on the functional coupling of two distinct signaling pathways widely used in leukocytes: the ITAM pathway and the Jak-STAT pathway. Through the use of the receptor for interferon-γ (IFN-γR) and the ITAM adaptor Fcγ as an example, we found that IFN-γ modified responses of the phagocytic antibody receptor FcγRI (CD64) to specify cell-autonomous antimicrobial functions. Unexpectedly, we also found that in peritoneal macrophages, IFN-γR itself required tonic signaling from Fcγ through the kinase PI(3)K for the induction of a subset of IFN-γ-specific antimicrobial functions. Our findings may be generalizable to other ITAM and Jak-STAT signaling pathways and may help explain signal integration by those pathways.

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: IFN-γR is coupled with the FcγRI signalosome in phagocytes.
Figure 2: FcγR and IFN-γR are dispensable for each other's expression and canonical signaling.
Figure 3: Induction of subset of IFN-γ-regulated genes in peritoneal macrophages requires expression of Fcγ.
Figure 4: Induction of a subset of IFN-γ-regulated genes in peritoneal macrophages requires tonic PI(3)K signaling through FcγR and Fcγ.
Figure 5: Early IFN-γ-mediated control of infection with L. monocytogenes in vivo requires Fcγ.
Figure 6: IFN-γR and FcγRI function by the 'coincidence detection principle' to induce a specific transcriptional program in phagocytes.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

References

  1. Humphrey, M.B., Lanier, L.L. & Nakamura, M.C. Role of ITAM-containing adapter proteins and their receptors in the immune system and bone. Immunol. Rev. 208, 50–65 (2005).

    CAS  PubMed  Google Scholar 

  2. Stark, G.R. & Darnell, J.E. Jr. The JAK-STAT pathway at twenty. Immunity 36, 503–514 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Nathan, C. & Sporn, M. Cytokines in context. J. Cell Biol. 113, 981–986 (1991).

    CAS  PubMed  Google Scholar 

  4. Murray, P.J. The JAK-STAT signaling pathway: input and output integration. J. Immunol. 178, 2623–2629 (2007).

    CAS  PubMed  Google Scholar 

  5. Call, M.E. & Wucherpfennig, K.W. Common themes in the assembly and architecture of activating immune receptors. Nat. Rev. Immunol. 7, 841–850 (2007).

    CAS  PubMed  Google Scholar 

  6. Bezbradica, J.S. & Medzhitov, R. Role of ITAM signaling module in signal integration. Curr. Opin. Immunol. 24, 58–66 (2012).

    CAS  PubMed  Google Scholar 

  7. Hamerman, J.A., Ni, M., Killebrew, J.R., Chu, C.L. & Lowell, C.A. The expanding roles of ITAM adapters FcRγ and DAP12 in myeloid cells. Immunol. Rev. 232, 42–58 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Koga, T. et al. Costimulatory signals mediated by the ITAM motif cooperate with RANKL for bone homeostasis. Nature 428, 758–763 (2004).

    CAS  PubMed  Google Scholar 

  9. Zou, W., Reeve, J.L., Liu, Y., Teitelbaum, S.L. & Ross, F.P. DAP12 couples c-Fms activation to the osteoclast cytoskeleton by recruitment of Syk. Mol. Cell 31, 422–431 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Horng, T., Bezbradica, J.S. & Medzhitov, R. NKG2D signaling is coupled to the interleukin 15 receptor signaling pathway. Nat. Immunol. 8, 1345–1352 (2007).

    CAS  PubMed  Google Scholar 

  11. Hida, S. et al. Fc receptor γ-chain, a constitutive component of the IL-3 receptor, is required for IL-3-induced IL-4 production in basophils. Nat. Immunol. 10, 214–222 (2009).

    CAS  PubMed  Google Scholar 

  12. Vivier, E. & Malissen, B. Innate and adaptive immunity: specificities and signaling hierarchies revisited. Nat. Immunol. 6, 17–21 (2005).

    CAS  PubMed  Google Scholar 

  13. Schroder, K., Hertzog, P.J., Ravasi, T. & Hume, D.A. Interferon-γ: an overview of signals, mechanisms and functions. J. Leukoc. Biol. 75, 163–189 (2004).

    CAS  PubMed  Google Scholar 

  14. Nimmerjahn, F. & Ravetch, J.V. Fcγ receptors: old friends and new family members. Immunity 24, 19–28 (2006).

    CAS  PubMed  Google Scholar 

  15. Pearse, R.N., Feinman, R. & Ravetch, J.V. Characterization of the promoter of the human gene encoding the high-affinity IgG receptor: transcriptional induction by γ-interferon is mediated through common DNA response elements. Proc. Natl. Acad. Sci. USA 88, 11305–11309 (1991).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Beekman, J.M., van der Linden, J.A., van de Winkel, J.G. & Leusen, J.H. FcγRI (CD64) resides constitutively in lipid rafts. Immunol. Lett. 116, 149–155 (2008).

    CAS  PubMed  Google Scholar 

  17. Wang, L. et al. 'Tuning' of type I interferon-induced Jak-STAT1 signaling by calcium-dependent kinases in macrophages. Nat. Immunol. 9, 186–193 (2008).

    CAS  PubMed  Google Scholar 

  18. Sato, K. et al. Dectin-2 is a pattern recognition receptor for fungi that couples with the Fc receptor γ chain to induce innate immune responses. J. Biol. Chem. 281, 38854–38866 (2006).

    CAS  PubMed  Google Scholar 

  19. Agarwal, A., Salem, P. & Robbins, K.C. Involvement of p72syk, a protein-tyrosine kinase, in Fcγ receptor signaling. J. Biol. Chem. 268, 15900–15905 (1993).

    CAS  PubMed  Google Scholar 

  20. Crowley, M.T. et al. A critical role for Syk in signal transduction and phagocytosis mediated by Fcγ receptors on macrophages. J. Exp. Med. 186, 1027–1039 (1997).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Gamero, A.M. & Larner, A.C. Signaling via the T cell antigen receptor induces phosphorylation of Stat1 on serine 727. J. Biol. Chem. 275, 16574–16578 (2000).

    CAS  PubMed  Google Scholar 

  22. Xu, W., Nair, J.S., Malhotra, A. & Zhang, J.J. B cell antigen receptor signaling enhances IFN-γ-induced Stat1 target gene expression through calcium mobilization and activation of multiple serine kinase pathways. J. Interferon Cytokine Res. 25, 113–124 (2005).

    CAS  PubMed  Google Scholar 

  23. Kovarik, P., Stoiber, D., Novy, M. & Decker, T. Stat1 combines signals derived from IFN-γ and LPS receptors during macrophage activation. EMBO J. 17, 3660–3668 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Farlik, M. et al. Nonconventional initiation complex assembly by STAT and NF-κB transcription factors regulates nitric oxide synthase expression. Immunity 33, 25–34 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Wen, Z., Zhong, Z. & Darnell, J.E. Jr. Maximal activation of transcription by Stat1 and Stat3 requires both tyrosine and serine phosphorylation. Cell 82, 241–250 (1995).

    CAS  PubMed  Google Scholar 

  26. Nair, J.S. et al. Requirement of Ca2+ and CaMKII for Stat1 Ser-727 phosphorylation in response to IFN-γ. Proc. Natl. Acad. Sci. USA 99, 5971–5976 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Kovarik, P. et al. Stress-induced phosphorylation of STAT1 at Ser727 requires p38 mitogen-activated protein kinase whereas IFN-γ uses a different signaling pathway. Proc. Natl. Acad. Sci. USA 96, 13956–13961 (1999).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. van Boxel-Dezaire, A.H. & Stark, G.R. Cell type-specific signaling in response to interferon-γ. Curr. Top. Microbiol. Immunol. 316, 119–154 (2007).

    CAS  PubMed  Google Scholar 

  29. MacMicking, J., Xie, Q.W. & Nathan, C. Nitric oxide and macrophage function. Annu. Rev. Immunol. 15, 323–350 (1997).

    CAS  PubMed  Google Scholar 

  30. Kleinert, H., Schwarz, P.M. & Forstermann, U. Regulation of the expression of inducible nitric oxide synthase. Biol. Chem. 384, 1343–1364 (2003).

    CAS  PubMed  Google Scholar 

  31. Kamijo, R. et al. Requirement for transcription factor IRF-1 in NO synthase induction in macrophages. Science 263, 1612–1615 (1994).

    CAS  PubMed  Google Scholar 

  32. Lowenstein, C.J. et al. Macrophage nitric oxide synthase gene: two upstream regions mediate induction by interferon γ and lipopolysaccharide. Proc. Natl. Acad. Sci. USA 90, 9730–9734 (1993).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. MacMicking, J.D. Immune control of phagosomal bacteria by p47 GTPases. Curr. Opin. Microbiol. 8, 74–82 (2005).

    CAS  PubMed  Google Scholar 

  34. Schattgen, S.A. & Fitzgerald, K.A. The PYHIN protein family as mediators of host defenses. Immunol. Rev. 243, 109–118 (2011).

    CAS  PubMed  Google Scholar 

  35. Roberts, T.L. et al. HIN-200 proteins regulate caspase activation in response to foreign cytoplasmic DNA. Science 323, 1057–1060 (2009).

    CAS  PubMed  Google Scholar 

  36. Unkeless, J.C. & Eisen, H.N. Binding of monomeric immunoglobulins to Fc receptors of mouse macrophages. J. Exp. Med. 142, 1520–1533 (1975).

    CAS  PubMed  Google Scholar 

  37. Ioan-Facsinay, A. et al. FcγRI (CD64) contributes substantially to severity of arthritis, hypersensitivity responses, and protection from bacterial infection. Immunity 16, 391–402 (2002).

    CAS  PubMed  Google Scholar 

  38. Ivashkiv, L.B. A signal-switch hypothesis for cross-regulation of cytokine and TLR signalling pathways. Nat. Rev. Immunol. 8, 816–822 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Pamer, E.G. Immune responses to Listeria monocytogenes. Nat. Rev. Immunol. 4, 812–823 (2004).

    CAS  PubMed  Google Scholar 

  40. Trost, M. et al. The phagosomal proteome in interferon-γ-activated macrophages. Immunity 30, 143–154 (2009).

    CAS  PubMed  Google Scholar 

  41. Srinivasan, L. et al. PI3 kinase signals BCR-dependent mature B cell survival. Cell 139, 573–586 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Schweighoffer, E. et al. The BAFF receptor transduces survival signals by co-opting the B cell receptor signaling pathway. Immunity 38, 475–488 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Wu, J. et al. An activating immunoreceptor complex formed by NKG2D and DAP10. Science 285, 730–732 (1999).

    CAS  PubMed  Google Scholar 

  44. Raulet, D.H. Roles of the NKG2D immunoreceptor and its ligands. Nat. Rev. Immunol. 3, 781–790 (2003).

    CAS  PubMed  Google Scholar 

  45. Lu, J. et al. Structural recognition and functional activation of FcγR by innate pentraxins. Nature 456, 989–992 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Kastenmüller, W., Torabi-Parizi, P., Subramanian, N., Lammermann, T. & Germain, R.N. A spatially-organized multicellular innate immune response in lymph nodes limits systemic pathogen spread. Cell 150, 1235–1248 (2012).

    PubMed  PubMed Central  Google Scholar 

  47. Boekhoudt, G.H., Frazier-Jessen, M.R. & Feldman, G.M. Immune complexes suppress IFN-γ signaling by activation of the FcγRI pathway. J. Leukoc. Biol. 81, 1086–1092 (2007).

    CAS  PubMed  Google Scholar 

  48. Virgin, H.W., Wittenberg, G.F. & Unanue, E.R. Immune complex effects on murine macrophages. I. Immune complexes suppress interferon-γ induction of Ia expression. J. Immunol. 135, 3735–3743 (1985).

    CAS  PubMed  Google Scholar 

  49. Esparza, I., Green, R. & Schreiber, R.D. Inhibition of macrophage tumoricidal activity by immune complexes and altered erythrocytes. J. Immunol. 131, 2117–2121 (1983).

    CAS  PubMed  Google Scholar 

  50. Gallo, P., Goncalves, R. & Mosser, D.M. The influence of IgG density and macrophage Fcγ receptor cross-linking on phagocytosis and IL-10 production. Immunol. Lett. 133, 70–77 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank D. Stetson (University of Washington, Seattle) for modified retroviral vector pMSCV.hCD2 and vectors MIGR2 and pMSCV.IRES-GFP; members of the Medzhitov laboratory and S. Joyce for discussions and reading of the manuscript; and C. Annicelli and S. Cronin for assistance with animal work. Supported by the Howard Hughes Medical Institute (R.M. and J.S.B.), the US National Institutes of Health (1R56AI087725-01 AI046688, AI055502, AI089771 and DK071754 to R.M.) and the Damon Runyon Cancer Research Foundation (DRG-1968-08 to J.S.B.).

Author information

Authors and Affiliations

Authors

Contributions

J.S.B. and R.M. designed research and wrote manuscript; J.S.B. did and analyzed experiments; R.K.R. and J.S.B. cloned and initially characterized Fcγ and its mutants; I.B. and J.S.B. did in vitro L. monocytogenes infection experiments; and R.A.D. analyzed and interpreted ImageStream data.

Corresponding author

Correspondence to Ruslan Medzhitov.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Immunofluorescence analysis of actin, IFN-γRI and FcγRI on macrophages.

(a, b) Immunofluorescence analysis of IFN-γRI localization in BM macrophages from wild-type and indicated KO mice, stimulated with IgG2a-opsonised SRBC for 15 min in the absence in a or presence in b of IFN-γ. Cells were fixed, permeabilized and probed with IFN-γRI antibody (clone C-20). Uptake of SRBC (N) by macrophages is extremely low, and so is uptake of SRBC (O) by Fcγ-deficient macrophages. Thus, arrows were added to those images to indicate location of few phagocytosed SRBC. (c) ImageStream analysis of actin recruitment to the FcγR phagocytic cup of BM DC stimulated with rabbit IgG-opsonized, Alexa fluor 488-labeled beads for 5 or 10 min. Cells were fixed, permeabilized and stained with Phalloidin Alexa fluor 647. (d) ImageStream analysis showing surface distribution of IFN-γRI and FcγRI in BM DC stained with antibodies against IFN-γRI (biotinilated clone 2E2) and FcγRI. Cells were not fixed nor permeabilized because, unlike clone C-20 (used in a and b), clone 2E2 against IFN-γRI and antibody against FcγRI do not work well under those conditions. BDS score is shown as histogram on top right and representative images with BDS>2 are shown at the bottom. (e and f) Flow cytometry analysis of IFN-γRI and FcγRI in wild-type and indicated KO macrophages, indicating the specificity of antibodies used in (d). For un-stained control, mix of wild-type and KO cells were used. (g) Schematic representation of IFN-γR and FcγRI with their key signaling components.

Supplementary Figure 2 Serine-phosphorylation of STAT1 by IFN-γR and FcγRI

(a-d) Immunoblot analysis of signaling in wild-type BM macrophages stimulated as indicated on the top with SRBC (O) (in synchronized phagocytosis assay) or with IFN-γ.Cells were lysed and analyzed by IB.

Supplementary Figure 3 Gene expression analysis of Fcγ-deficient peritoneal macrophages after IFN-γ and IL-4 stimulation.

(a) Flow cytometry of peritoneal cells from wild-type (B6 or BALB/c) and Fcγ-deficient mice stained with macrophage and B cells specific markers. (b and c) Quantitative PCR analysis of gene transcription in wild-type and Fcγ-deficient peritoneal macrophages stimulated with IFN-γ in b or IL-4 in c for 0-6 h. (d and e) Immunoblot and flow cytometry analysis of wild-type Fcγ and its mutant protein expression upon retroviral transduction into Fcγ-deficient BM. Expression was monitored in differentiated macrophages using Flag immunoblot in d, of Flag surface staining in e. (f) Outline of the microarray data analysis of global gene expression changes in peritoneal macrophages after IFN-γ or IL-4 stimulation for 6 h. (g) Cluster analysis of genes differently expressed between wild type and Fcγ-deficient peritoneal macrophages relative to wild-type unstimulated sample after stimulation with IL-4 for 6 h. Data were analyzed and presented as in Fig. 3f. (h) Summary of the results in g. (i) Quantitative PCR analysis confirmation of selected genes of interest from g, results are presented relative to KO unstimulated sample, set as 1, because expression in wild-type was low to undetectable for Ifi202b. (j) Comparison of two arrays: Genes differently expressed between wild-type and Fcγ-deficient peritoneal macrophages relative to wild-type unstimulated sample in response to IFN-γ or IL-4, from the two microarrays shown in Fig. 3f and Supplementary fig. 3g, respectively, were compared. Gene groups A, B, C, E, F, and G from each of those arrays (i.e., where cluster profiles were different between WT STIM and KO STIM) were aligned and compared. Shared genes in two arrays were identified and clustered in two main groups: Group A, where expression patterns are similar between two arrays and Group B, where expression patterns are distinct.

Supplementary Figure 4 Peritoneal macrophage signaling in the presence of pathway inhibitors.

(a) Two models describing how Fcγ adapter could be linked to IFN-γR functions. (b) Griess assay measuring NO production in the sup of STAT1-deficient and wild type BM macrophages stimulated with SRBC (O) with or without IFN-γ for ~16-20 h. (c) Immunoblot analysis of signaling in BM macrophages pretreated with DMSO, NFAT inhibitor (Cyclosporine, 1 and 10 uM), PI3K inhibitor (Ly294002, 10 and 20 uM) or ERK inhibitor (PD98059, 20 and 40 uM) for 60 min and then stimulated with SRBC (O) for 15 min. (d) Immunoblot analysis of signaling in peritoneal macrophages pretreated with DMSO, PI3K inhibitor (Wortmannin or AktVIII), pan Jak inhibitor, ERK inhibitor (PD98059) or NFkB inhibitor (Bay11) for 30 min and then stimulated with SRBC (O) and IFN-γ or LPS Concentrations tested for Wortmannin and panJak: 1 uM, 500 nM, 100 nM; for AKTVIII, PD98059 and Bay11: 10 uM, 1 uM, 100 nM. LPS was used for Bay11 testing because it is more robust stimulator of NFkB pathway than SRBC (O). (e and f) Quantitative PCR analysis of gene transcription in peritoneal macrophages pretreated with DMSO, Bay11, PD98059 (ERK inh) or CyclosporineA (NFAT inh) in e or 100 nM panJak inhibitor in f for 30 min prior to IFN-γ stimulation for 6 h.

Supplementary Figure 5 L. monocytogenes uptake by wild-type, Fcγ-deficient and IFN-γRI-deficient macrophages.

B6, Fcγ-deficient and IFN-γRI-deficient BM macrophages were replated in 24 well TC dishes in antibiotic free media, stimulated in triplicates with IFN-γ, where indicated, for 16 h to induce iNOS expression before being infected with L. monocytogenes at MOI of 10. To synchronize infection bacteria were spun over macrophages for 5 min, infection was carried for 30 min at 37oC, and extracellular bacteria were killed by adding gentamycin to cultures at c=50 ug/ml for 1 h. Cells were then lysed in H2O, lysates were plated on streptomycin containing LB plates and bacterial loads calculated after 24 h.

Supplementary Figure 6 IFN-γ functional 'collaboration' with FcγRs during phagocytosis.

(a) BM macrophages microarray data analysis outline. (b) Primers used for quantitative PCR in this study. (c) To validate hybridoma sups used for opsonization, SRBC were opsonized for 1 h at RT. Cells were lysed in 1X SDS buffer and analyzed by IB. Normal mouse IgG was run on the left as control. (d) To validate that SRBC (O) signaling is Fcγ-dependent BM macrophages were stimulated with SRBC (N) (SRBC incubated with either in 10% DMEM, left, or with control hybridoma conditional media, middle) or with SRBC (O) (right) for 15 min. Cells were analyzed by immunoblot. (e, f) To validate that SRBC (O) responses are Fcγ-dependent BM macrophages were stimulated as indicated for 16 h. NO was measured in the culture supernatant using Griess assay.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–6 (PDF 6790 kb)

Supplementary Table 1

Peritoneal macrophage IFN-γ array (XLSX 149 kb)

Supplementary Table 2

Peritoneal macrophage IL-4 array (XLSX 128 kb)

Supplementary Table 3

Shared genes between IFN-γ and IL-4 array (XLSX 88 kb)

Supplementary Table 4

BM macrophages SRBC (O) and IFN-γ array (XLSX 77 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Bezbradica, J., Rosenstein, R., DeMarco, R. et al. A role for the ITAM signaling module in specifying cytokine-receptor functions. Nat Immunol 15, 333–342 (2014). https://doi.org/10.1038/ni.2845

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ni.2845

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