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:

STAT1-cooperative DNA binding distinguishes type 1 from type 2 interferon signaling

A Corrigendum to this article was published on 20 October 2014

This article has been updated

Abstract

STAT1 is an indispensable component of a heterotrimer (ISGF3) and a STAT1 homodimer (GAF) that function as transcription regulators in type 1 and type 2 interferon signaling, respectively. To investigate the importance of STAT1-cooperative DNA binding, we generated gene-targeted mice expressing cooperativity-deficient STAT1 with alanine substituted for Phe77. Neither ISGF3 nor GAF bound DNA cooperatively in the STAT1F77A mouse strain, but type 1 and type 2 interferon responses were affected differently. Type 2 interferon–mediated transcription and antibacterial immunity essentially disappeared owing to defective promoter recruitment of GAF. In contrast, STAT1 recruitment to ISGF3 binding sites and type 1 interferon–dependent responses, including antiviral protection, remained intact. We conclude that STAT1 cooperativity is essential for its biological activity and underlies the cellular responses to type 2, but not type 1 interferon.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Both GAF and ISGF3 cannot bind DNA cooperatively in STAT1F77A mice.
Figure 2: Antibacterial, not antiviral, immunity requires STAT1 cooperative DNA binding.
Figure 3: Type 2 IFN responses are defective in STATF77A MEFs.
Figure 4: Promoter recruitment of GAF, not ISGF3, requires STAT1 cooperativity.
Figure 5: Interferon priming cannot rescue defective IFN-γ signaling.
Figure 6: Effects of mutation F77A on STAT1 chromatin association and serine phosphorylation.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

Change history

  • 24 February 2014

    In the version of this article initially published, Klaus-Peter Knobeloch was incorrectly not included in the author list or Author Contributions section. This name should appear after "Filipa Antunes" in the author list and should be linked to the following affiliation: Institut für Neuropathologie, Universitätsklinikum Freiburg, Freiburg, Germany. The Author Contributions section should include the following revision to the sixth item: "K.-P.K. and R.N. contributed reagents and expertise for mouse genetics experiments" (and the thanks to "K.-P. Knobeloch, Universität Freiburg, for advice and cloning reagents" should be removed from the Acknowledgments section). The error has been corrected in the HTML and PDF versions of the article.

References

  1. Stark, G.R., Kerr, I.M., Williams, B.R., Silverman, R.H. & Schreiber, R.D. How cells respond to interferons. Annu. Rev. Biochem. 67, 227–264 (1998).

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

    CAS  PubMed  Google Scholar 

  4. Vinkemeier, U. et al. DNA binding of in vitro activated Stat1 alpha, Stat1 beta and truncated Stat1: interaction between NH2-terminal domains stabilizes binding of two dimers to tandem DNA sites. EMBO J. 15, 5616–5626 (1996).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Li, X., Leung, S., Burns, C. & Stark, G.R. Cooperative binding of Stat1-2 heterodimers and ISGF3 to tandem DNA elements. Biochimie 80, 703–710 (1998).

    CAS  PubMed  Google Scholar 

  6. Vinkemeier, U., Moarefi, I., Darnell, J.E. Jr. & Kuriyan, J. Structure of the amino-terminal protein interaction domain of STAT-4. Science 279, 1048–1052 (1998).

    CAS  PubMed  Google Scholar 

  7. Xu, X., Sun, Y.L. & Hoey, T. Cooperative DNA binding and sequence-selective recognition conferred by the STAT amino-terminal domain. Science 273, 794–797 (1996).

    CAS  PubMed  Google Scholar 

  8. Bonham, A.J. et al. STAT1:DNA sequence-dependent binding modulation by phosphorylation, protein:protein interactions and small-molecule inhibition. Nucleic Acids Res. 41, 754–763 (2013).

    CAS  PubMed  Google Scholar 

  9. Meyer, T., Hendry, L., Begitt, A., John, S. & Vinkemeier, U. A single residue modulates tyrosine dephosphorylation, oligomerization, and nuclear accumulation of Stat transcription factors. J. Biol. Chem. 279, 18998–19007 (2004).

    CAS  PubMed  Google Scholar 

  10. Mao, X. et al. Structural bases of unphosphorylated STAT1 association and receptor binding. Mol. Cell 17, 761–771 (2005).

    CAS  PubMed  Google Scholar 

  11. Wenta, N., Strauss, H., Meyer, S. & Vinkemeier, U. Tyrosine phosphorylation regulates the partitioning of STAT1 between different dimer conformations. Proc. Natl. Acad. Sci. USA 105, 9238–9243 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Ptashne, M. A Genetic Switch: Phage Lambda Revisited (Cold Spring Harbor Laboratory Press, Plainview, New York, USA, 2004).

  13. Lebrecht, D. et al. Bicoid cooperative DNA binding is critical for embryonic patterning in Drosophila. Proc. Natl. Acad. Sci. USA 102, 13176–13181 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Spitz, F. & Furlong, E.E. Transcription factors: from enhancer binding to developmental control. Nat. Rev. Genet. 13, 613–626 (2012).

    CAS  PubMed  Google Scholar 

  15. Lin, J.-X. et al. Critical Role of STAT5 transcription factor tetramerization for cytokine responses and normal immune function. Immunity 36, 586–599 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Giorgetti, L. et al. Noncooperative interactions between transcription factors and clustered DNA binding sites enable graded transcriptional responses to environmental inputs. Mol. Cell 37, 418–428 (2010).

    CAS  PubMed  Google Scholar 

  17. Durbin, J.E., Hackenmiller, R., Simon, M.C. & Levy, D.E. Targeted disruption of the mouse Stat1 gene results in compromised innate immunity to viral disease. Cell 84, 443–450 (1996).

    CAS  PubMed  Google Scholar 

  18. Zhong, M. et al. Implications of an antiparallel dimeric structure of nonphosphorylated STAT1 for the activation-inactivation cycle. Proc. Natl. Acad. Sci. USA 102, 3966–3971 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Qing, Y. & Stark, G.R. Alternative activation of STAT1 and STAT3 in response to interferon-gamma. J. Biol. Chem. 279, 41679–41685 (2004).

    CAS  PubMed  Google Scholar 

  20. Boisson-Dupuis, S. et al. Inborn errors of human STAT1: allelic heterogeneity governs the diversity of immunological and infectious phenotypes. Curr. Opin. Immunol. 24, 364–378 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    PubMed  Google Scholar 

  22. Buchmeier, N.A. & Schreiber, R.D. Requirement of endogenous interferon-gamma production for resolution of Listeria monocytogenes infection. Proc. Natl. Acad. Sci. USA 82, 7404–7408 (1985).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Lorsbach, R.B., Murphy, W.J., Lowenstein, C.J., Snyder, S.H. & Russell, S.W. Expression of the nitric oxide synthase gene in mouse macrophages activated for tumor cell killing. Molecular basis for the synergy between interferon-gamma and lipopolysaccharide. J. Biol. Chem. 268, 1908–1913 (1993).

    CAS  PubMed  Google Scholar 

  24. Stockinger, S. et al. Production of type I IFN sensitizes macrophages to cell death induced by Listeria monocytogenes. J. Immunol. 169, 6522–6529 (2002).

    CAS  PubMed  Google Scholar 

  25. Karaghiosoff, M. et al. Central role for type I interferons and Tyk2 in lipopolysaccharide-induced endotoxin shock. Nat. Immunol. 4, 471–477 (2003).

    CAS  PubMed  Google Scholar 

  26. Hartman, S.E. et al. Global changes in STAT target selection and transcription regulation upon interferon treatments. Genes Dev. 19, 2953–2968 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Ramana, C.V., Gil, M.P., Schreiber, R.D. & Stark, G.R. Stat1-dependent and -independent pathways in IFN-gamma-dependent signaling. Trends Immunol. 23, 96–101 (2002).

    CAS  PubMed  Google Scholar 

  28. Schmid, C.D. & Bucher, P. MER41 repeat sequences contain inducible STAT1 binding sites. PLoS ONE 5, e11425 (2010).

    PubMed  PubMed Central  Google Scholar 

  29. Hu, X. et al. Sensitization of IFN-gamma Jak-STAT signaling during macrophage activation. Nat. Immunol. 3, 859–866 (2002).

    CAS  PubMed  Google Scholar 

  30. Vinkemeier, U. Getting the message across, STAT! Design principles of a molecular signaling circuit. J. Cell Biol. 167, 197–201 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Lödige, I. et al. Nuclear export determines the cytokine sensitivity of STAT transcription factors. J. Biol. Chem. 280, 43087–43099 (2005).

    PubMed  Google Scholar 

  32. Meyer, T., Marg, A., Lemke, P., Wiesner, B. & Vinkemeier, U. DNA binding controls inactivation and nuclear accumulation of the transcription factor Stat1. Genes Dev. 17, 1992–2005 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Bluyssen, H.A. & Levy, D.E. Stat2 is a transcriptional activator that requires sequence-specific contacts provided by Stat1 and p48 for stable interaction with DNA. J. Biol. Chem. 272, 4600–4605 (1997).

    CAS  PubMed  Google Scholar 

  34. Sadzak, I. et al. Recruitment of Stat1 to chromatin is required for interferon-induced serine phosphorylation of Stat1 transactivation domain. Proc. Natl. Acad. Sci. USA 105, 8944–8949 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Wormald, S., Hilton, D.J., Smyth, G.K. & Speed, T.P. Proximal genomic localization of STAT1 binding and regulated transcriptional activity. BMC Genomics 7, 254 (2006).

    PubMed  PubMed Central  Google Scholar 

  36. Segal, E. & Widom, J. From DNA sequence to transcriptional behaviour: a quantitative approach. Nat. Rev. Genet. 10, 443–456 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Kessler, D.S., Veals, S.A., Fu, X.Y. & Levy, D.E. Interferon-alpha regulates nuclear translocation and DNA-binding affinity of ISGF3, a multimeric transcriptional activator. Genes Dev. 4, 1753–1765 (1990).

    CAS  PubMed  Google Scholar 

  38. Qureshi, S.A., Salditt-Georgieff, M. & Darnell, J.E. Jr. Tyrosine-phosphorylated Stat1 and Stat2 plus a 48-kDa protein all contact DNA in forming interferon-stimulated-gene factor 3. Proc. Natl. Acad. Sci. USA 92, 3829–3833 (1995).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Speil, J. et al. Activated STAT1 transcription factors conduct distinct saltatory movements in the cell nucleus. Biophys. J. 101, 2592–2600 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Cheon, H. et al. IFNβ-dependent increases in STAT1, STAT2, and IRF9 mediate resistance to viruses and DNA damage. EMBO J. 32, 2751–2763 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Dölken, L. et al. High-resolution gene expression profiling for simultaneous kinetic parameter analysis of RNA synthesis and decay. RNA 14, 1959–1972 (2008).

    PubMed  PubMed Central  Google Scholar 

  42. Mandal, M. et al. Epigenetic repression of the Igk locus by STAT5-mediated recruitment of the histone methyltransferase Ezh2. Nat. Immunol. 12, 1212–1220 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Chen, X. et al. A reinterpretation of the dimerization interface of the N-terminal domains of STATs. Protein Sci. 12, 361–365 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Stockinger, S. et al. Characterization of the interferon-producing cell in mice infected with Listeria monocytogenes. PLoS Pathog. 5, e1000355 (2009).

    PubMed  PubMed Central  Google Scholar 

  45. Detje, C.N. et al. Local type I IFN receptor signaling protects against virus spread within the central nervous system. J. Immunol. 182, 2297–2304 (2009).

    CAS  PubMed  Google Scholar 

  46. Begitt, A., Droescher, M., Knobeloch, K.P. & Vinkemeier, U. SUMO conjugation of STAT1 protects cells from hyperresponsiveness to IFNγ. Blood 118, 1002–1007 (2011).

    CAS  PubMed  Google Scholar 

  47. Zwaferink, H., Stockinger, S., Reipert, S. & Decker, T. Stimulation of inducible nitric oxide synthase expression by beta interferon increases necrotic death of macrophages upon Listeria monocytogenes infection. Infect. Immun. 76, 1649–1656 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Pelzel, C., Begitt, A., Wenta, N. & Vinkemeier, U. Evidence against a role for β-arrestin1 in STAT1 dephosphorylation and the inhibition of interferon-γ signaling. Mol. Cell 50, 149–156 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Antunes, F., Marg, A. & Vinkemeier, U. STAT1 signaling is not regulated by a phosphorylation-acetylation switch. Mol. Cell. Biol. 31, 3029–3037 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Meyer, T., Begitt, A. & Vinkemeier, U. Green fluorescent protein-tagging reduces the nucleocytoplasmic shuttling specifically of unphosphorylated STAT1. FEBS J. 274, 815–826 (2007).

    CAS  PubMed  Google Scholar 

  51. Meyer, L.R. et al. The UCSC Genome Browser database: extensions and updates 2013. Nucleic Acids Res. 41, D64–D69 (2013).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank M. Mee, C. Pelzel and N. Wenta for immunoblot or EMSA results; N. Salhat for data on Listeria-infected macrophages; V. Ruppert and J. Staab for animal data; U. Kalinke (Universität Hannover) for VSV; L. Olohan (University of Liverpool) for microarray scanning; and K. Weber, E. Louis and I. Macdonald for manuscript review. Supported by University of Nottingham Pump Priming (U.V.), Deutsche Forschungsgemeinschaft (VI 218/3 to U.V. and ME 1648/4-1 to T.M.), Austrian Science Foundation (SFB28 to T.D.), Biotechnology and Biological Sciences Research Council (BB/G019290/1 to U.V. and BB/I532353/1 to M.B.), Pfizer (M.B.) and the Wellcome Trust (A.B.).

Author information

Authors and Affiliations

Authors

Contributions

A.B. designed and executed gene expression and promoter binding studies; M.D. and T.M. generated the STAT1F77A mouse strain, and designed and performed biochemical experiments and animal studies; C.D.S. did bioinformatics promoter analyses; M.B. and M.R.O. generated mathematical models; F.A. designed and performed cell-based assays and generated microarray data; K.-P.K. and R.N. contributed reagents and expertise for mouse genetics experiments; T.D. provided reagents and expertise for bacterial infection experiments; U.V. conceived the research, directed the study and wrote the manuscript. All authors contributed to data analyses and manuscript editing.

Corresponding author

Correspondence to Uwe Vinkemeier.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Generation and characterization of STAT1F77A mice.

(a) Schematic diagram of the targeting construct used for homologous recombination in ES cells (top), the murine Stat1 gene locus encompassing exons 1-6 (second), a depiction of the targeted allele with insertion of a loxP-flanked neomycin gene (third), and the resulting Stat1 locus after Cre-induced deletion of the neomycin gene (bottom). The horizontal rectangle represents the position of probe P used for Southern blotting. (b) Southern blot analysis of genomic DNA after digestion with BamHI and hybridization with probe P. Homologous recombination with the targeting vector introduces an additional BamHI site that produces a 5.7 kB band representing the modified allele. (c) RFLP analyses using genomic DNA from wild-type (WT/WT), hetero- (WT/KI) and homozygous (KI/KI) STAT1F77A mice. Due to the loss of a Tsp509I site caused by mutation F77A, STAT1F77A-derived DNA showed a 234 bp fragment, which was cleaved into 110 bp and 120 bp fragments with wild-type-derived DNA. The undigested PCR amplicon (282 bp) is shown in lane 6; std, sizing standard. (d) Electropherograms showing DNA sequencing results for the non-coding strand from wild-type (WT/WT), hetero- (WT/KI) and homozygous (KI/KI) STAT1F77A animals.

Supplementary Figure 2 Type-2, but not type-1 IFN-stimulated NO production is defective in STAT1F77A-derived macrophages.

(a-c). Nitric oxide production of bone marrow-derived macrophages (BMMs) from wild-type (left) or STAT1F77A mice (right). Macrophages were seeded in 96-well plates at a density of 1 × 104 cells per well and kept in L cell-conditioned medium (10% L cell-supplemented DMEM with 10% calf serum). For the indicated times the medium was supplemented with different concentrations of IFN-γ (a) or IFN-α (b,c) alone or in combination with LPS. The concentrations of IFN and LPS are given in the keys. Nitrite concentrations in the culture supernatants were subsequently determined by Griess assay. Data are representative of two independent experiments done in duplicates

Supplementary Figure 3 Type-2 IFN responses are defective in STAT1F77A mice.

(a) Gene induction in immortalized MEFs after 6 h IFN-α (1000 U/ml) or IFN-γ (50 U/ml) as determined by qRT-PCR. (b) Validation of IFN-mediated gene repression. Cells were treated with IFNs for 6 h unless indicated otherwise. Note that Jun expression was below the 4-fold expression limit at the 6 h time point and hence was not detected in the microarray studies. For Pvrl4, qRT-PCR and microarray results (Supplementary Table 1) were contradictory. Bars represent the mean ± s.d. of three independent experiments. The 45 min time point for Jun was done once.

Supplementary Figure 4 Promoter recruitment of GAF, not ISGF3, requires STAT1 cooperativity.

Gels show recruitment of complexes to promoters for Gapdh (a), Tgtp (b), Igtp (c), Socs1 (d), Cxcl10 (e), Irf9 (f), and AldoB (g), determined by ChIP analysis of nuclear preparations from immortalized wild-type (WT) and STAT1F77A MEFs (KI), before or after treatment with 1,000 U/ml IFN-α or 50 U/ml IFN-γ for 45 min. Anti-STAT1 (S1), anti-STAT2 (S2), anti-IRF9 (IRF9), anti-acetylated histone 3 (Ac-H3), or anti-unspecific IgG (IgG) was used to immunoprecipitate (IP) the complexes, followed by PCR with primers designed to detect the promoters. The position and configuration of GAS and ISRE in each promoter sequence are indicated below the gels. bp, base pairs. Data are representative of one (g) or at least two independent experiments.

Supplementary Figure 5 Promoter recruitment of GAF, not ISGF3, requires STAT1 cooperativity.

Gels show recruitment of complexes to promoters for Pvalb (a), Npas (b), Pvrl4 (c), and Jun (d), determined by ChIP analysis of nuclear preparations from immortalized wild-type (WT) and STAT1F77A MEFs (KI), before or after treatment with 1,000 U/ml IFN-α or 50 U/ml IFN-γ for 45 min. Anti-STAT1 (S1), or anti-STAT2 (S2) was used to immunoprecipitate (IP) the complexes, followed by PCR with primers designed to detect the promoters. The position and configuration of GAS and ISRE in each promoter sequence are indicated below the gels. bp, base pairs. Data are representative of one (a) or at least two independent experiments.

Supplementary Figure 6 GAS and ISRE consensus sequence variants.

(a) Sequence logo with a graphical representation of the binding preferences of STAT1 as derived from ChIP-seq data28 Letter sizes indicate the frequency of the respective bases in the binding sequences. (b) Representative examples of sequences obtaining the corresponding PWM scores.

Supplementary Figure 7 In silico analyses of STAT1 cooperative DNA binding.

(a) Results of mathematical modeling of STAT1-DNA binding using the ‘single GAS polymer model’. Promoter recruitment was tested over a range of different affinities of STAT1 for non-GAS sites (key). The solid lines depict results for high cooperativity (Kd = 0.0017). For comparison, the dotted line shows the result for low cooperativity (Kd = 0.1) and a 50-fold difference in GAS to non-GAS STAT-DNA binding affinity, as displayed in Figure 6a. As the affinity of STAT1 for non-GAS sites decreases, the curves shift to the right of the plot, indicating that a greater STAT1 concentration would be needed to achieve the same level of recruitment. However, the dotted line shows that cooperativity facilitated promoter recruitment at all binding affinities tested. (b) Results of mathematical modeling of STAT1 DNA binding in a tetramer model. This model operates with two binding sites, either two GAS sites (blue), or a GAS and a non-GAS site (red). These configurations can be compared to the black lines (key) that show STAT1 recruitment to a single GAS site in the more realistic ‘single GAS polymer model’ as displayed in Figure 6a. The curves are shifted to the right slightly in the ‘single GAS polymer model’, indicating that the tetrameric model would overestimate the STAT1 concentration needed to achieve any particular level of STAT1 recruitment.

Supplementary Figure 8 Validation of STAT1 nuclear retention assay.

(a) Wide-field microscopy images of HeLa cells expressing the wild-type STAT1 or STAT1-NES. Shown is the GFP fluorescence, nuclei were stained with Hoechst dye. In unstimulated cells, STAT1-NES localizes predominantly in the cytoplasm (i). Addition of 10 ng/ml NES inactivatorleptomycin B (LMB, Sigma)50 had little effect on wild-type, but caused STAT1-NES to adopt a pancellular distribution (ii). Treatment with IFN-γ (50 U/ml) for 60 min expectedly resulted in nuclear accumulation of wild-type, but did not visibly change the distribution of STAT1-NES (iii). However, IFN-γ-induced nuclear accumulation of STAT1-NES occurred upon NES inactivation by LMB (iv), indicating that its nuclear import was functioning. Next, phosphatase inhibitor vanadate (0.8 mM)32 was used together with IFN-γ (v), which resulted in the nuclear accumulation of wild-type STAT1, demonstrating that vanadate did not inhibit STAT1 nuclear import. Vanadate, on the other hand, did not trigger nuclear accumulation of STAT1-NES in the IFN-γ-treated cells (v). A possible explanation for this outcome could have been that vanadate inhibited nuclear import. However, this was ruled out since wild-type STAT1 was imported normally in the presence of vanadate. As STAT1-NES remained predominantly cytoplasmic in the vanadate-treated cells, it was concluded furthermore that vanadate did not prevent the nuclear export of STAT1-NES either. Thus, vanadate did not preclude nuclear import and export of activated STAT1-NES. Since nuclear accumulation was not observed in the presence of phosphatase inhibitor vanadate, we concluded that NES-fusion enabled STAT1 to exit from the nucleus in the tyrosine-phosphorylated state.To test this hypothesis, the NES was inactivated in cells treated with both IFN-γ and vanadate (vi), which resulted in the indistinguishable nuclear accumulation of wild-type STAT1 and the NES fusion variant. Together, these results indicated that tyrosine phosphorylation was not sufficient for nuclear retention of STAT1-NES, contrary to wild-type STAT1 (ref. 30). (b) Immunoblot analysis of STAT1 phosphorylated at Tyr701 (pS1Y) and total STAT1 (S1) in HeLa cells transfected with GFP-tagged wild-type STAT1 or STAT1-NES. The cells were treated with 50 U/ml IFN-γ for the times indicated. (c) Representative quantitative confocal microscopy images of optical slices through the median of HeLa cells before (US) and after 1 h stimulation with IFN-γ (50 U/ml) or IFN-α (1000 U/ml). The cells expressed GFP-tagged STAT1-NES (WT) or the indicated mutants; amino acid changes are listed. Shown is the GFP fluorescence intensity in a color-coded rendering based on the scale shown at the bottom. Data are representative of three or more independent experiments.

Supplementary Figure 9 A schematic summarizing the conclusions concerning the roles of GAF and ISGF3 in type-1 and type-2 IFN-mediated gene induction and repression.

See key for explanations.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–9, Supplementary Tables 1–6 and Supplementary Note (PDF 5952 kb)

Source data

Rights and permissions

Reprints and permissions

About this article

Cite this article

Begitt, A., Droescher, M., Meyer, T. et al. STAT1-cooperative DNA binding distinguishes type 1 from type 2 interferon signaling. Nat Immunol 15, 168–176 (2014). https://doi.org/10.1038/ni.2794

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ni.2794

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