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:

STAM and Hrs interact sequentially with IFN-α Receptor to control spatiotemporal JAK–STAT endosomal activation

Abstract

Activation of the JAK–STAT pathway by type I interferons (IFNs) requires clathrin-dependent endocytosis of the IFN-α and -β receptor (IFNAR), indicating a role for endosomal sorting in this process. The molecular machinery that brings the selective activation of IFN-α/β-induced JAK–STAT signalling on endosomes remains unknown. Here we show that the constitutive association of STAM with IFNAR1 and TYK2 kinase at the plasma membrane prevents TYK2 activation by type I IFNs. IFN-α-stimulated IFNAR endocytosis delivers the STAM–IFNAR complex to early endosomes where it interacts with Hrs, thereby relieving TYK2 inhibition by STAM and triggering signalling of IFNAR at the endosome. In contrast, when stimulated by IFN-β, IFNAR signalling occurs independently of Hrs as IFNAR is sorted to a distinct endosomal subdomain. Our results identify the molecular machinery that controls the spatiotemporal activation of IFNAR by IFN-α and establish the central role of endosomal sorting in the differential regulation of JAK–STAT signalling by IFN-α and IFN-β.

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

Fig. 1: IFNAR1 interaction with Hrs.
Fig. 2: Interaction of IFNAR1 with Hrs is required for IFN-α-induced JAK–STAT signalling.
Fig. 3: IFNAR1 and IFNAR2 degradations are independent of Hrs.
Fig. 4: The constitutive association of STAM with TYK2 prevents TYK2 activation.
Fig. 5: IFNAR1 constitutively interacts with STAM at the plasma membrane and at the early endosome.
Fig. 6: STAM2A interacts with IFNAR1 and IFNAR2 independently of Hrs.
Fig. 7: IFNAR endocytosis is dispensable for efficient JAK–STAT signalling in cells lacking STAM.
Fig. 8: IFNAR1 interaction with Hrs relieves STAM inhibition of TYK2.

Similar content being viewed by others

Data availability

All data supporting the findings of this study are available from the corresponding authors on reasonable request. Source data are provided with this paper.

References

  1. O’Shea, J. J. et al. The JAK–STAT pathway: impact on human disease and therapeutic intervention. Ann. Rev. Med 66, 311–328 (2015).

    Article  PubMed  Google Scholar 

  2. Villarino, A. V., Kanno, Y. & O’Shea, J. J. Mechanisms and consequences of Jak–STAT signaling in the immune system. Nat. Immunol. 18, 374–384 (2017).

    Article  CAS  PubMed  Google Scholar 

  3. Lochte, S., Waichman, S., Beutel, O., You, C. & Piehler, J. Live cell micropatterning reveals the dynamics of signaling complexes at the plasma membrane. J. Cell Biol. 207, 407–418 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  4. Platanias, L. C. Mechanisms of type-I- and type-II-interferon-mediated signalling. Nat. Rev. Immunol. 5, 375–386 (2005).

    Article  CAS  PubMed  Google Scholar 

  5. Thomas, C. et al. Structural linkage between ligand discrimination and receptor activation by type I interferons. Cell 146, 621–632 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Ivashkiv, L. B. & Donlin, L. T. Regulation of type I interferon responses. Nat. Rev. Immunol. 14, 36–49 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Fox, L. E., Locke, M. C. & Lenschow, D. J. Context is key: delineating the unique functions of IFNα and IFNβ in disease. Front. Immunol. 11, 606874 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Jaitin, D. A. et al. Inquiring into the differential action of interferons (IFNs): an IFN-α2 mutant with enhanced affinity to IFNAR1 is functionally similar to IFN-β. Mol. Cell. Biol. 26, 1888–1897 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. de Weerd, N. A. et al. Structural basis of a unique interferon-β signaling axis mediated via the receptor IFNAR1. Nat. Immunol. 14, 901–907 (2013).

    Article  PubMed  Google Scholar 

  10. Schreiber, G. & Piehler, J. The molecular basis for functional plasticity in type I interferon signaling. Trends Immunol. 36, 139–149 (2015).

    Article  CAS  PubMed  Google Scholar 

  11. Marchetti, M. et al. Stat-mediated signaling induced by type I and type II interferons (IFNs) is differentially controlled through lipid microdomain association and clathrin-dependent endocytosis of IFN receptors. Mol. Biol. Cell 17, 2896–2909 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Gonnord, P., Blouin, C. M. & Lamaze, C. Membrane trafficking and signaling: two sides of the same coin. Semin. Cell Dev. Biol. 23, 154–164 (2012).

    Article  CAS  PubMed  Google Scholar 

  13. Zanin, N., de Lesegno, C. V., Lamaze, C. & Blouin, C.M. Interferon receptor trafficking and signaling: journey to the cross roads. Front. Immunol. 11, 615603 (2021).

  14. Vieira, A. V., Lamaze, C. & Schmid, S. L. Control of EGF receptor signaling by clathrin-mediated endocytosis. Science 274, 2086–2089 (1996).

    Article  CAS  PubMed  Google Scholar 

  15. Di Fiore, P.P. & von Zastrow, M. Endocytosis, signaling, and beyond. Cold Spring Harb. Perspect. Biol. 6, a016865 (2014).

  16. Irannejad, R., Tsvetanova, N. G., Lobingier, B. T. & von Zastrow, M. Effects of endocytosis on receptor-mediated signaling. Curr. Opin. Cell Biol. 35, 137–143 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Villasenor, R., Kalaidzidis, Y. & Zerial, M. Signal processing by the endosomal system. Curr. Opin. Cell Biol. 39, 53–60 (2016).

    Article  CAS  PubMed  Google Scholar 

  18. Asao, H. et al. Hrs is associated with STAM, a signal-transducing adaptor molecule. Its suppressive effect on cytokine-induced cell growth. J. Biol. Chem. 272, 32785–32791 (1997).

    Article  CAS  PubMed  Google Scholar 

  19. Bache, K. G., Raiborg, C., Mehlum, A. & Stenmark, H. STAM and Hrs are subunits of a multivalent ubiquitin-binding complex on early endosomes. J. Biol. Chem. 278, 12513–12521 (2003).

    Article  CAS  PubMed  Google Scholar 

  20. Mayers, J. R. et al. ESCRT-0 assembles as a heterotetrameric complex on membranes and binds multiple ubiquitinylated cargoes simultaneously. J. Biol. Chem. 286, 9636–9645 (2011).

    Article  CAS  PubMed  Google Scholar 

  21. Migliano, S. M., Wenzel, E. M. & Stenmark, H. Biophysical and molecular mechanisms of ESCRT functions, and their implications for disease. Curr. Opin. Cell Biol. 75, 102062 (2022).

    Article  CAS  PubMed  Google Scholar 

  22. Lohi, O. & Lehto, V. P. STAM/EAST/Hbp adapter proteins—integrators of signalling pathways. FEBS Lett. 508, 287–290 (2001).

    Article  CAS  PubMed  Google Scholar 

  23. Raiborg, C. et al. FYVE and coiled-coil domains determine the specific localisation of Hrs to early endosomes. J. Cell Sci. 114, 2255–2263 (2001).

    Article  CAS  PubMed  Google Scholar 

  24. Row, P. E., Clague, M. J. & Urbe, S. Growth factors induce differential phosphorylation profiles of the Hrs–STAM complex: a common node in signalling networks with signal-specific properties. Biochem. J. 389, 629–636 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Urbe, S., Mills, I. G., Stenmark, H., Kitamura, N. & Clague, M. J. Endosomal localization and receptor dynamics determine tyrosine phosphorylation of hepatocyte growth factor-regulated tyrosine kinase substrate. Mol. Cell. Biol. 20, 7685–7692 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Urbe, S. et al. The UIM domain of Hrs couples receptor sorting to vesicle formation. J. Cell Sci. 116, 4169–4179 (2003).

    Article  CAS  PubMed  Google Scholar 

  27. Wegner, C. S. et al. Ultrastructural characterization of giant endosomes induced by GTPase-deficient Rab5. Histochem. Cell Biol. 133, 41–55 (2010).

    Article  CAS  PubMed  Google Scholar 

  28. Bache, K. G., Raiborg, C., Mehlum, A., Madshus, I. H. & Stenmark, H. Phosphorylation of Hrs downstream of the epidermal growth factor receptor. Eur. J. Biochem. 269, 3881–3887 (2002).

    Article  CAS  PubMed  Google Scholar 

  29. Komada, M. & Kitamura, N. Growth factor-induced tyrosine phosphorylation of Hrs, a novel 115-kilodalton protein with a structurally conserved putative zinc finger domain. Mol. Cell. Biol. 15, 6213–6221 (1995).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Bache, K. G., Brech, A., Mehlum, A. & Stenmark, H. Hrs regulates multivesicular body formation via ESCRT recruitment to endosomes. J. Cell Biol. 162, 435–442 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Prag, G. et al. The Vps27/Hse1 complex is a GAT domain-based scaffold for ubiquitin-dependent sorting. Dev. Cell 12, 973–986 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Kumar, K. G. et al. Site-specific ubiquitination exposes a linear motif to promote interferon-α receptor endocytosis. J. Cell Biol. 179, 935–950 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Marijanovic, Z., Ragimbeau, J., Kumar, K. G., Fuchs, S. Y. & Pellegrini, S. TYK2 activity promotes ligand-induced IFNAR1 proteolysis. Biochem. J. 397, 31–38 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Kumar, K. G. et al. SCF(HOS) ubiquitin ligase mediates the ligand-induced down-regulation of the interferon-α receptor. EMBO J. 22, 5480–5490 (2003).

    Article  CAS  PubMed  Google Scholar 

  35. Marijanovic, Z., Ragimbeau, J., van der Heyden, J., Uze, G. & Pellegrini, S. Comparable potency of IFNα2 and IFNβ on immediate JAK/STAT activation but differential down-regulation of IFNAR2. Biochem. J. 407, 141–151 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Umebayashi, K., Stenmark, H. & Yoshimori, T. Ubc4/5 and c-Cbl continue to ubiquitinate EGF receptor after internalization to facilitate polyubiquitination and degradation. Mol. Biol. Cell 19, 3454–3462 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Endo, K. et al. STAM2, a new member of the STAM family, binding to the Janus kinases. FEBS Lett. 477, 55–61 (2000).

    Article  CAS  PubMed  Google Scholar 

  38. Takeshita, T. et al. STAM, signal transducing adaptor molecule, is associated with Janus kinases and involved in signaling for cell growth and c-myc induction. Immunity 6, 449–457 (1997).

    Article  CAS  PubMed  Google Scholar 

  39. Mizuno, E., Kawahata, K., Okamoto, A., Kitamura, N. & Komada, M. Association with Hrs is required for the early endosomal localization, stability, and function of STAM. J. Biochem. 135, 385–396 (2004).

    Article  CAS  PubMed  Google Scholar 

  40. Christ, L., Raiborg, C., Wenzel, E. M., Campsteijn, C. & Stenmark, H. Cellular functions and molecular mechanisms of the ESCRT membrane-scission machinery. Trends Biochem. Sci. 42, 42–56 (2017).

    Article  CAS  PubMed  Google Scholar 

  41. Raiborg, C. & Stenmark, H. The ESCRT machinery in endosomal sorting of ubiquitylated membrane proteins. Nature 458, 445–452 (2009).

    Article  CAS  PubMed  Google Scholar 

  42. Soderberg, O. et al. Direct observation of individual endogenous protein complexes in situ by proximity ligation. Nat. Methods 3, 995–1000 (2006).

    Article  PubMed  Google Scholar 

  43. Wallweber, H. J., Tam, C., Franke, Y., Starovasnik, M. A. & Lupardus, P. J. Structural basis of recognition of interferon-α receptor by tyrosine kinase 2. Nat. Struct. Mol. Biol. 21, 443–448 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Sezgin, E. et al. Elucidating membrane structure and protein behavior using giant plasma membrane vesicles. Nat. Protoc. 7, 1042–1051 (2012).

    Article  CAS  PubMed  Google Scholar 

  45. Dewulf, M. et al. Dystrophy-associated caveolin-3 mutations reveal that caveolae couple IL6/STAT3 signaling with mechanosensing in human muscle cells. Nat. Commun. 10, 1974 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  46. Christoforidis, S. et al. Phosphatidylinositol-3-OH kinases are Rab5 effectors. Nat. Cell Biol. 1, 249–252 (1999).

    Article  CAS  PubMed  Google Scholar 

  47. Lodhi, I. J. et al. Insulin stimulates phosphatidylinositol 3-phosphate production via the activation of Rab5. Mol. Biol. Cell 19, 2718–2728 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Elkin, S. R. et al. Ikarugamycin: a natural product inhibitor of clathrin-mediated endocytosis. Traffic 17, 1139–1149 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Benmerah, A. et al. AP-2/Eps15 interaction is required for receptor-mediated endocytosis. J. Cell Biol. 140, 1055–1062 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Heldin, C. H., Lu, B., Evans, R. & Gutkind, J. S. Signals and receptors. Cold Spring Harb. Perspect. Biol. 8, a005900 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  51. Platta, H. W. & Stenmark, H. Endocytosis and signaling. Curr. Opin. Cell Biol. 23, 393–403 (2011).

    Article  CAS  PubMed  Google Scholar 

  52. von Zastrow, M. & Sorkin, A. Mechanisms for regulating and organizing receptor signaling by endocytosis. Annu. Rev. Biochem. 90, 709–737 (2021).

    Article  Google Scholar 

  53. Kojima, K. et al. ESCRT-0 protein hepatocyte growth factor-regulated tyrosine kinase substrate (Hrs) is targeted to endosomes independently of signal-transducing adaptor molecule (STAM) and the complex formation with STAM promotes its endosomal dissociation. J. Biol. Chem. 289, 33296–33310 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Gong, J., Korner, R., Gaitanos, L. & Klein, R. Exosomes mediate cell contact-independent ephrin–Eph signaling during axon guidance. J. Cell Biol. 214, 35–44 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Hammond, D. E. et al. Endosomal dynamics of Met determine signaling output. Mol. Biol. Cell 14, 1346–1354 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Rodahl, L. M., Stuffers, S., Lobert, V. H. & Stenmark, H. The role of ESCRT proteins in attenuation of cell signalling. Biochem. Soc. Trans. 37, 137–142 (2009).

    Article  CAS  PubMed  Google Scholar 

  57. Hurley, J. H. ESCRTs are everywhere. EMBO J. 34, 2398–2407 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Pavlin, M. R. & Hurley, J.H. The ESCRTs—converging on mechanism. J. Cell. Sci. 133, jcs240333 (2020).

  59. van Boxel-Dezaire, A. H., Rani, M. R. & Stark, G. R. Complex modulation of cell type-specific signaling in response to type I interferons. Immunity 25, 361–372 (2006).

    Article  PubMed  Google Scholar 

  60. Piehler, J., Thomas, C., Christopher Garcia, K. & Schreiber, G. Structural and dynamic determinants of type I interferon receptor assembly and their functional interpretation. Immunol. Rev. 250, 317–334 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  61. Martinez-Fabregas, J. et al. Kinetics of cytokine receptor trafficking determine signaling and functional selectivity. eLife 8, e49314 (2019).

  62. Gorby, C. et al. Engineered IL-10 variants elicit potent immunomodulatory effects at low ligand doses. Sci. Signal. 13, eabc0653 (2020).

  63. Chmiest, D. et al. Spatiotemporal control of interferon-induced JAK/STAT signalling and gene transcription by the retromer complex. Nat. Commun. 7, 13476 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Yoshimura, A., Ito, M., Chikuma, S., Akanuma, T. & Nakatsukasa, H. Negative regulation of cytokine signaling in immunity. Cold Spring Harb. Perspect. Biol., 10, a028571 (2018).

  65. Piganis, R. A. et al. Suppressor of cytokine signaling (SOCS) 1 inhibits type I interferon (IFN) signaling via the interferon α receptor (IFNAR1)-associated tyrosine kinase Tyk2. J. Biol. Chem. 286, 33811–33818 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Liau, N. P. D. et al. The molecular basis of JAK/STAT inhibition by SOCS1. Nat. Commun. 9, 1558 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  67. Simonetti, B. et al. Molecular identification of a BAR domain-containing coat complex for endosomal recycling of transmembrane proteins. Nat. Cell Biol. 21, 1219–1233 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Norris, A. et al. SNX-1 and RME-8 oppose the assembly of HGRS-1/ESCRT-0 degradative microdomains on endosomes. Proc. Natl Acad. Sci. USA 114, E307–E316 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Blouin, C. M. et al. Glycosylation-dependent IFN-γR partitioning in lipid and actin nanodomains is critical for JAK activation. Cell 166, 920–934 (2016).

    Article  CAS  PubMed  Google Scholar 

  70. Schmidt-Arras, D. & Rose-John, S. Endosomes as signaling platforms for IL-6 family cytokine receptors. Front. Cell Dev. Biol. 9, 688314 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  71. Muller, U. et al. Functional role of type I and type II interferons in antiviral defense. Science 264, 1918–1921 (1994).

    Article  CAS  PubMed  Google Scholar 

  72. MacDonald, E. et al. HRS–WASH axis governs actin-mediated endosomal recycling and cell invasion. J. Cell Biol. 217, 2549–2564 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Kalie, E., Jaitin, D. A., Abramovich, R. & Schreiber, G. An interferon α2 mutant optimized by phage display for IFNAR1 binding confers specifically enhanced antitumor activities. J. Biol. Chem. 282, 11602–11611 (2007).

    Article  CAS  PubMed  Google Scholar 

  74. Bago, R. et al. Characterization of VPS34-IN1, a selective inhibitor of Vps34, reveals that the phosphatidylinositol 3-phosphate-binding SGK3 protein kinase is a downstream target of class III phosphoinositide 3-kinase. Biochem. J. 463, 413–427 (2014).

    Article  CAS  PubMed  Google Scholar 

  75. Bolte, S. & Cordelières, F. P. A guided tour into subcellular colocalization analysis in light microscopy. J. Microsc. 224, 213–232 (2006).

    Article  CAS  PubMed  Google Scholar 

  76. Schindelin, J. et al. Fiji: an open-source platform for biological-image analysis. Nat. Methods 9, 676–682 (2012).

    Article  CAS  PubMed  Google Scholar 

  77. Wedeking, T. et al. Spatiotemporally controlled reorganization of signaling complexes in the plasma membrane of living cells. Small 11, 5912–5918 (2015).

    Article  CAS  PubMed  Google Scholar 

  78. Wedeking, T. et al. Single cell GFP–Trap reveals stoichiometry and dynamics of cytosolic protein complexes. Nano Lett. 15, 3610–3615 (2015).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank the core facilities and the CurieCoreTech recombinant antibodies platform of Institut Curie, and the staff of the Cell and Tissue Imaging (PICT-IBiSA) and the Nikon Imaging Centre at Institut Curie as well as the members of the French National Research Infrastructure France–BioImaging (ANR10-INBS-04) for their scientific and technical assistance. In particular, we thank F. Waharte and L. Sengmanivong for their help with the FLIM–FRET and fluorescence microscopy experiments. We thank the following people for providing materials or expertise: G. Raposo and P. Benaroch (Institut Curie, Paris), and P. Eid (INSERM, Paris). We thank the Morph-Im core facility (UNamur, Belgium) for Airyscan imaging. This work was supported by institutional grants from the Curie Institute, INSERM, CNRS, and by specific grants from Agence Nationale de la Recherche (ANR NANOSTAT-15-CE11-0025-01 to C.L.) and Marie Curie Actions—Networks for Initial Training (FP7-PEOPLE-2010-ITN to C.L. and D.C.). Funding was also provided by the Deutsche Forschungsgemeinschaft to J. Piehler (NANOSTAT, PI 405/10-1). Fast FLIM imaging was carried out at the NeurImag Imaging core facility, part of the IPNP, INSERM 1266 unit and Université de Paris. Support from Fondation Leducq is acknowledged. N.Z. was supported by a PhD fellowship from Ministère de l’Enseignement Supérieur et de la Recherche and Ligue Nationale contre le Cancer, D.C. by a PhD fellowship from Fondation pour la Recherche Médicale and C.M.B. by a postdoctoral fellowship from Ligue Nationale contre le Cancer. The Lamaze team are members of Labex CelTisPhyBio ANR-10-LBX-0038, part of the IDEX PSL ANR-10-IDEX-0001-02.

Author information

Authors and Affiliations

Authors

Contributions

N.Z., C.M.B., C.V.d.L., J. Podkalicka and T.M. designed the study, performed experiments and analysed results. N.Z., C.M.B., C.V.d.L., J. Piehler and C.L. wrote the manuscript. D.C., P.G.T., P.B., L.D. and S.U. assisted with experiments and provided several reagents. C.L. supervised and directed the research. All authors discussed the results and commented on the manuscript.

Corresponding authors

Correspondence to Cédric M. Blouin or Christophe Lamaze.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Cell Biology thanks Paul Hertzog, Pier Paolo Di Fiore, and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Peer reviewer reports are available.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 IFN-α-dependent proximity of IFNAR1 and Hrs on endosomes.

a, Immunofluorescent labelling of Hrs, EEA1 and endocytosed IFNAR1 subunits after 10 min IFN-α (upper panels) or IFN-β (lower panels) stimulation in eGFP–Rab5 Q79L-transfected RPE1 cells observed by confocal microscopy with representative plots of co-localization profiles on endosomes. Quantification of Hrs and IFNAR1 co-localization is expressed as the Manders’ coefficient indicating the portion of IFNAR1 pixels containing Hrs pixels. Scale bar, 10 µm. Data represent the mean ± s.e.m., n = 8 cells examined from two independent experiments. Statistical significance was determined using a two-tailed Student’s t-test. ***P < 0,001; NS, not significant. b, Fast FLIM analysis of the IFNAR1–eGFP fluorescence lifetime in segmented endosomes of transfected RPE1 cells (with or without Hrs–mCherry expression) at steady state or following stimulation with IFN-α or IFN-β for 10 or 20 min (one dot is the mean lifetime of all endosomes from one cell); n = 28, 29, 29, 29, 29 and 29 cells, respectively, examined from four independent experiments. Data are the median ± 95% confidence interval. P values were calculated using a one-way ANOVA with the Kruskal–Wallis multiple comparison test. ***P < 0.001; NS, not significant. Source numerical data are provided.

Source data

Extended Data Fig. 2 Effect of Hrs depletion on JAK–STAT signalling and receptor degradation.

a, Nuclear translocation of pSTAT1 from wide-field microscopy imaging (left) and pSTAT1 nucleus/cytosol fluorescence ratio following stimulation with IFN-α, IFN-β or IFN-α2-YNS for 10 min in control RPE1 cells (shCtrl: α = 80, β = 142, α2-YNS = 90) or depleted for Hrs (shHrs: α = 90, β = 90, α2-YNS = 156; right). Data were pooled from two independent experiments. Scale bar, 50 µm. b, Immunoblots of tyrosine phosphorylation levels of IFNAR1 (pIFNAR1) in control (shCtrl) and shHrs RPE1 cells with or without IFN-α stimulation for 10 min, as indicated. c, Immunoblots and quantification of tyrosine phosphorylation levels of JAK1 (pJAK1) in shCtrl and shHrs HeLa S3-GFP cells (top) or S3-mHrs-GFP cells (bottom) with or without IFN-α or IFN-β stimulation for 10 min, as indicated. Control shCtrl + IFN-β was set at 100%. d, Immunofluorescent labelling (left) and quantification (right) of surface and total of IFNAR1 subunits in control or Hrs-depleted RPE1 cells analysed by confocal microscopy (the mean of the control condition was set to 100); n = 16 cells (shCtrl) and n = 28 cells (shHrs) examined from two independent experiments for surface staining and n = 28 cells (shCtrl) and n = 25 cells (shHrs) examined from three independent experiments for total staining. Scale bar, 10 µm. e,f, Immunoblot and quantification of IFNAR1 (e; three independent experiments) and EGFR (f; two independent experiments) degradation in RPE1 cells depleted or not for Hrs following IFN-β or EGF (100 ng ml−1) stimulation, respectively. Mean of the control t = 0 time point was set at 100%. a,d,e, Data are the mean ± s.e.m. Statistical significance was determined using an ANOVA with Dunnett’s multiple comparison test (a,d) or a one-way ANOVA with the Kruskal–Wallis multiple comparison test (e); ****P < 0,0001; NS, not significant. b,c, Data are representative of two independent experiments. Source numerical data and unprocessed blots are provided.

Source data

Extended Data Fig. 3 STAM constitutively interacts with IFNAR1 at the plasma membrane.

a, Immunofluorescent labelling of STAM2A and IFNAR1 subunits at steady state in RPE1 cells observed by confocal microscopy with representative plots of co-localization profiles in the vicinity of the plasma membrane. Scale bars, 10 µm. b, Co-immunoprecipitation of endogenous STAM1 and STAM2 with endogenous IFNAR1 subunit in control or TYK2-depleted RPE1 cells with or without IFN-α or IFN-β treatment for 10 min with. Data are representative of three independent experiments. c, Immunofluorescence imaging (left) of IFNAR1 uptake in control (siRNA scrambled; n = 35) or STAM1 and STAM2-depleted (n = 34) RPE1 cells stimulated for 10 min with IFN-α. Following fixation, the cells were co-labelled for EEA1 and analysed by confocal microscopy. Scale bar, 10 μm. Quantifications (right) of co-localizations are expressed as the Manders’ coefficient, indicating the proportion of IFNAR1 pixels containing EEA1 pixels in cells. Data pooled from three independent experiments and represent the mean ± s.e.m. d, Levels of IFNAR1 surface staining monitored by FACS in control or STAM1 and STAM2-depleted RPE1 cells stimulated with IFN-α or IFN-β for 10 min. The same gating strategy (red polygon, left) was used for all samples. Data are representative of two independent experiments; NS, not significant. Source numerical data and unprocessed blots are provided.

Source data

Extended Data Fig. 4 STAM constitutively interacts with TYK2 and is not required for IFNAR1 endocytosis.

a, RFP-Trap immunoprecipitation of TYK2–mCherry in RPE1 cells expressing eGFP–STAM2A. Control immunoblots for RFP-Trap at steady state (left) without any cells or with cells transfected with either eGFP–STAM2A or eGFP and with either TYK2–mCherry or mCherry. Immunoblots for RFP-Trap (right) on RPE1 cells transfected with TYK2–mCherry and eGFP–STAM2A with or without IFN-α or IFN-β stimulation for 10 min. Data are representative of two independent experiments. b, Immunofluorescence imaging (left) of IFNAR1 uptake in control (siRNA scrambled) or STAM1 and STAM2-depleted RPE1 cells stimulated for 10 min with IFN-α. Following fixation, the cells were co-labelled for EEA1 and analysed by confocal microscopy. Scale bars, 50 µm. Quantifications (right) of co-localizations are expressed as the Manders’ coefficient, indicating the proportion of IFNAR1 pixels containing EEA1 pixels in three independent experiments. c, Levels of IFNAR1 surface staining monitored by FACS in control or STAM1 and STAM2-depleted RPE1 cells stimulated with IFN-α or IFN-β for 10 min. Data are representative of two independent experiments. Statistical significance was determined using a two-way RM ANOVA with Tukey’s multiple comparisons test. d, Immunoblots for TYK2 expression in RPE1 cells with or without siRNA to TYK2 transfection for 72 h (left). PLA experiments monitoring the interaction between total IFNAR1 and STAM2A in control (n = 53) and TYK2 (n = 45)-depleted RPE1 cells pooled from three independent experiments (middle). The corresponding quantifications show the mean ± s.e.m. (right). Statistical significance was determined using an unpaired two-tailed Student’s t-test. NS, not significant. Scale bar, 10 µm. Source numerical data and unprocessed blots are provided.

Source data

Extended Data Fig. 5 Block of clathrin-dependent endocytosis in cells ectopically expressing Hrs at the plasma membrane.

a, Confocal microscopy of endogenous Hrs immunofluorescent labelling in RPE1 cells co-transfected with peGFP-FYVE and with constitutively active RFP-Rab5-CAAX (Q79L) and HA-TC10 (Q75L) to ectopically produce PI3-P at the plasma membrane. Insets: magnified views of the plasma membrane (boxes 1 and 2) and endosomal (box 3) areas. Scale bars, 10 µm. b, RPE1 cells treated with 4 µM ikarugamycin or dimethylsulfoxide (Ctrl) were incubated for 20 min at 4 °C with Alexa Fluor 546 transferrin, anti-IFNAR1 and IFN-α. The cells were then transferred to 37 °C for 10 min to allow endocytosis, fixed, immunostained for EEA1 and observed by confocal microscopy. Inset: magnified views of the plasma membrane areas (top) and the endosomal structures (bottom). Scale bars, 10 µm. Data are representative of three independent experiments.

Extended Data Fig. 6 Blocking of clathrin-dependent endocytosis by dominant-negative Eps15 mutant in cells ectopically expressing Hrs at the plasma membrane.

a, Wide-field microscopy of Alexa Fluor 546 transferrin uptake (10 min) in control Eps15 D3Δ2–eGFP (arrow)-or dominant-negative Eps15 DN–eGFP (arrowhead)-transfected cells. Data are representative of three independent experiments. b, Wide-field microscopy (right) and quantification (left) of pSTAT1 nuclear translocation in RPE1 cells with or without (ø) IFN-α or IFN-γ stimulation for 10 min cells transfected with control Eps15 D3Δ2–eGFP (ø = 24, α = 33, γ = 56), or dominant-negative Eps15 DN–eGFP (ø = 36, α = 28, γ = 35). The nucleus/cytosol fluorescence ratio for pSTAT1 was detremined from the fluorescence imaging. Nuclear translocation of pSTAT1 is inhibited by Eps15 DN–eGFP for IFN-α (white arrow) but not for IFN-γ (white arrowhead). c, Wide-field microscopy (right) and quantification (left) of pSTAT1 nuclear translocation in RPE1 cells with or without IFN-α or IFN-β stimulation for 10 min and co-transfected with control Eps15 D3Δ2–eGFP alone (ø = 162, α = 162, β = 167), RFP-Rab5-CAAX (Q79L) (ø = 158, α = 160, β = 161), dominant-negative Eps15 DN–eGFP alone (ø = 162, α = 167, β = 161) or RFP-Rab5-CAAX (Q79L) (ø = 162, α = 162, β = 161). pSTAT1 nuclear translocation is rescued for IFN-α (white arrowheads) but not for IFN-β (white arrows). The nucleus/cytosol fluorescence ratio for pSTAT1 was determined from the fluorescence imaging. b,c, Data were pooled from three independent experiments and are shown as the median ± 95% confidence interval. Statistical significance was determined using a one-way ANOVA with the Kruskal–Wallis multiple comparison test. ****P < 0.0001; ***P < 0.001; *P < 0.05; NS, not significant. Scale bars, 20 µm. Source numerical data are provided.

Source data

Extended Data Fig. 7 Competition between STAM2A and SOCS-1 for binding to the TYK2–IFNAR1 signalling complex.

Representative live-cell micropatterning experiment with micropatterned receptor HaloTag–mTagBFP–IFNAR1 as the bait protein (cyan), and TYK2–meGFP FERM–SH2 (FS) and full-length (FL) forms (green), STAM2A–mCherry (yellow) and SOCS-1–iRFP (red) as prey proteins. Scale bars, 10 µm. Interactions of TYK2, STAM2A and SOCS-1 with micropatterned IFNAR1 were quantified according to the contrast within micropatterned cells. Cells expressing TYK2-FS/SOCS-1 (n = 10), TYK2-FL/SOCS-1 (n = 15) or TYK2-FL/SOCS-1/STAM2A (n = 20) were analysed for contrast values and visualized in a box-and-whisker plot (representing the median, minima, maxima and the four quartiles); data were pooled from two independent experiments. Statistical significance was determined using a two-sample Kolmogorov–Smirnov tests. **P < 0.01. Source numerical data are provided.

Source data

Supplementary information

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 1

Unprocessed western blots.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 2

Unprocessed western blots.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 3

Unprocessed western blots.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 4

Unprocessed western blots.

Source Data Fig. 5

Statistical source data.

Source Data Fig. 5

Unprocessed western blots.

Source Data Fig. 6

Statistical source data.

Source Data Fig. 6

Unprocessed western blots.

Source Data Fig. 7

Statistical source data.

Source Data Fig. 7

Unprocessed western blots.

Source Data Fig. 8

Statistical source data.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 2

Statistical source data.

Source Data Extended Data Fig. 2

Unprocessed western blots.

Source Data Extended Data Fig. 3

Statistical source data.

Source Data Extended Data Fig. 3

Unprocessed western blots.

Source Data Extended Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 4

Unprocessed western blots.

Source Data Extended Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 7

Statistical source data.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zanin, N., Viaris de Lesegno, C., Podkalicka, J. et al. STAM and Hrs interact sequentially with IFN-α Receptor to control spatiotemporal JAK–STAT endosomal activation. Nat Cell Biol 25, 425–438 (2023). https://doi.org/10.1038/s41556-022-01085-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41556-022-01085-6

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