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:

The DNA-binding domain mediates both nuclear and cytosolic functions of p53

Abstract

Under conditions of genotoxic stress, human p53 activates the apoptotic effectors BAX or BAK to result in mitochondrial outer-membrane permeabilization and apoptosis. Antiapoptotic BCL-2 family member BCL-xL opposes this activity by sequestering cytosolic p53 via association with its DNA-binding domain, an interaction enhanced by p53 tetramerization. Here we characterized the BCL-xL–p53 complex by NMR spectroscopy and modulated it through mutagenesis to determine the relative contributions of BCL-xL's interactions with p53 or other BCL-2 family proteins to the BCL-xL–dependent inhibition of UV irradiation–induced apoptosis. Under our experimental conditions, one-third of the antiapoptotic activity of BCL-xL was mediated by p53 sequestration and the remaining two-thirds through sequestration of proapoptotic BCL-2 family members. Our studies define the contributions of cytosolic p53 to UV irradiation–induced apoptosis and provide opportunities to explore its contributions to other p53-dependent apoptotic signaling 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: Schematic representation of p53 constructs used in this study.
Figure 2: Titrations of various unlabeled p53 domain constructs into 15N-labeled BCL-xLΔC monitored by 1H-15N TROSY.
Figure 3: Titrations of unlabeled BCL-xLΔC into different 15N-labeled p53 constructs monitored by 1H-15N TROSY.
Figure 4: Structure of the complex between p53 DBD and BCL-xL.
Figure 5: Mutagenesis analysis of individual p53 N-D-T residue contributions to DNA or BCL-xL binding and BCL-xLΔC residue contributions to p53 binding.
Figure 6: Functional effects of mutations within p53 that destabilize its interaction with BCL-xL.
Figure 7: Functional effects of mutations within BCL-xL that destabilize its interaction with p53.

Similar content being viewed by others

Accession codes

Primary accessions

Biological Magnetic Resonance Data Bank

Protein Data Bank

Referenced accessions

Protein Data Bank

References

  1. Green, D.R. & Kroemer, G. Cytoplasmic functions of the tumour suppressor p53. Nature 458, 1127–1130 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Chipuk, J.E. et al. Direct activation of Bax by p53 mediates mitochondrial membrane permeabilization and apoptosis. Science 303, 1010–1014 (2004).

    Article  CAS  PubMed  Google Scholar 

  3. Leu, J.I., Dumont, P., Hafey, M., Murphy, M.E. & George, D.L. Mitochondrial p53 activates Bak and causes disruption of a Bak–Mcl1 complex. Nat. Cell Biol. 6, 443–450 (2004).

    Article  CAS  PubMed  Google Scholar 

  4. Chipuk, J.E., Bouchier-Hayes, L., Kuwana, T., Newmeyer, D.D. & Green, D.R. PUMA couples the nuclear and cytoplasmic proapoptotic function of p53. Science 309, 1732–1735 (2005).

    Article  CAS  PubMed  Google Scholar 

  5. Sattler, M. et al. Structure of Bcl-xL-Bak peptide complex: recognition between regulators of apoptosis. Science 275, 983–986 (1997).

    Article  CAS  PubMed  Google Scholar 

  6. Petros, A.M. et al. Rationale for Bcl-xL/Bad peptide complex formation from structure, mutagenesis, and biophysical studies. Protein Sci. 9, 2528–2534 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Feng, W., Huang, S., Wu, H. & Zhang, M. Molecular basis of Bcl-xL's target recognition versatility revealed by the structure of Bcl-xL in complex with the BH3 domain of Beclin-1. J. Mol. Biol. 372, 223–235 (2007).

    Article  CAS  PubMed  Google Scholar 

  8. Liu, X., Dai, S., Zhu, Y., Marrack, P. & Kappler, J.W. The structure of a Bcl-xL/Bim fragment complex: implications for Bim function. Immunity 19, 341–352 (2003).

    Article  CAS  PubMed  Google Scholar 

  9. Cheng, E.H. et al. BCL-2, BCL-X(L) sequester BH3 domain-only molecules preventing BAX- and BAK-mediated mitochondrial apoptosis. Mol. Cell 8, 705–711 (2001).

    Article  CAS  PubMed  Google Scholar 

  10. Kussie, P.H. et al. Structure of the MDM2 oncoprotein bound to the p53 tumor suppressor transactivation domain. Science 274, 948–953 (1996).

    Article  CAS  PubMed  Google Scholar 

  11. Wang, P. et al. p53 domains: structure, oligomerization, and transformation. Mol. Cell. Biol. 14, 5182–5191 (1994).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Petros, A.M., Gunasekera, A., Xu, N., Olejniczak, E.T. & Fesik, S.W. Defining the p53 DNA-binding domain/Bcl-x(L)-binding interface using NMR. FEBS Lett. 559, 171–174 (2004).

    Article  CAS  PubMed  Google Scholar 

  13. Sot, B., Freund, S.M. & Fersht, A.R. Comparative biophysical characterization of p53 with the pro-apoptotic BAK and the anti-apoptotic BCL-xL. J. Biol. Chem. 282, 29193–29200 (2007).

    Article  CAS  PubMed  Google Scholar 

  14. Hagn, F. et al. BclxL changes conformation upon binding to wild-type but not mutant p53 DNA binding domain. J. Biol. Chem. 285, 3439–3450 (2010).

    Article  CAS  PubMed  Google Scholar 

  15. Yao, H. et al. Anti-apoptosis proteins Mcl-1 and Bcl-xL have different p53-binding profiles. Biochemistry 52, 6324–6334 (2013).

    Article  CAS  PubMed  Google Scholar 

  16. Di Lello, P. et al. Structure of the Tfb1/p53 complex: insights into the interaction between the p62/Tfb1 subunit of TFIIH and the activation domain of p53. Mol. Cell 22, 731–740 (2006).

    Article  CAS  PubMed  Google Scholar 

  17. Lee, C.W., Martinez-Yamout, M.A., Dyson, H.J. & Wright, P.E. Structure of the p53 transactivation domain in complex with the nuclear receptor coactivator binding domain of CREB binding protein. Biochemistry 49, 9964–9971 (2010).

    Article  CAS  PubMed  Google Scholar 

  18. Xu, H., Tai, J., Ye, H., Kang, C.B. & Yoon, H.S. The N-terminal domain of tumor suppressor p53 is involved in the molecular interaction with the anti-apoptotic protein Bcl-Xl. Biochem. Biophys. Res. Commun. 341, 938–944 (2006).

    Article  CAS  PubMed  Google Scholar 

  19. Xu, H. et al. The MDM2-binding region in the transactivation domain of p53 also acts as a Bcl-X(L)-binding motif. Biochemistry 48, 12159–12168 (2009).

    Article  CAS  PubMed  Google Scholar 

  20. Ha, J.H. et al. Dual-site interactions of p53 protein transactivation domain with anti-apoptotic Bcl-2 family proteins reveal a highly convergent mechanism of divergent p53 pathways. J. Biol. Chem. 288, 7387–7398 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Livingstone, L.R. et al. Altered cell cycle arrest and gene amplification potential accompany loss of wild-type p53. Cell 70, 923–935 (1992).

    Article  CAS  PubMed  Google Scholar 

  22. Serrano, M., Lin, A.W., McCurrach, M.E., Beach, D. & Lowe, S.W. Oncogenic ras provokes premature cell senescence associated with accumulation of p53 and p16INK4a. Cell 88, 593–602 (1997).

    Article  CAS  PubMed  Google Scholar 

  23. Yonish-Rouach, E. et al. Wild-type p53 induces apoptosis of myeloid leukaemic cells that is inhibited by interleukin-6. Nature 352, 345–347 (1991).

    Article  CAS  PubMed  Google Scholar 

  24. Mietz, J.A., Unger, T., Huibregtse, J.M. & Howley, P.M. The transcriptional transactivation function of wild-type p53 is inhibited by SV40 large T-antigen and by HPV-16 E6 oncoprotein. EMBO J. 11, 5013–5020 (1992).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Schultz, L.B., Chehab, N.H., Malikzay, A. & Halazonetis, T.D. p53 binding protein 1 (53BP1) is an early participant in the cellular response to DNA double-strand breaks. J. Cell Biol. 151, 1381–1390 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Pervushin, K., Riek, R., Wider, G. & Wuthrich, K. Attenuated T-2 relaxation by mutual cancellation of dipole-dipole coupling and chemical shift anisotropy indicates an avenue to NMR structures of very large biological macromolecules in solution. Proc. Natl. Acad. Sci. USA 94, 12366–12371 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Muchmore, S.W. et al. X-ray and NMR structure of human Bcl-xL, an inhibitor of programmed cell death. Nature 381, 335–341 (1996).

    Article  CAS  PubMed  Google Scholar 

  28. Joerger, A.C., Allen, M.D. & Fersht, A.R. Crystal structure of a superstable mutant of human p53 core domain: insights into the mechanism of rescuing oncogenic mutations. J. Biol. Chem. 279, 1291–1296 (2004).

    Article  CAS  PubMed  Google Scholar 

  29. Follis, A.V. et al. PUMA binding induces partial unfolding within BCL-xL to disrupt p53 binding and promote apoptosis. Nat. Chem. Biol. 9, 163–168 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Galea, C., Bowman, P. & Kriwacki, R.W. Disruption of an intermonomer salt bridge in the p53 tetramerization domain results in an increased propensity to form amyloid fibrils. Protein Sci. 14, 2993–3003 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Johnson, C.R., Morin, P.E., Arrowsmith, C.H. & Freire, E. Thermodynamic analysis of the structural stability of the tetrameric oligomerization domain of p53 tumor-suppressor. Biochemistry 34, 5309–5316 (1995).

    Article  CAS  PubMed  Google Scholar 

  32. Kitayner, M. et al. Structural basis of DNA recognition by p53 tetramers. Mol. Cell 22, 741–753 (2006).

    Article  CAS  PubMed  Google Scholar 

  33. Cho, Y., Gorina, S., Jeffrey, P.D. & Pavletich, N.P. Crystal structure of a p53 tumor suppressor-DNA complex: understanding tumorigenic mutations. Science 265, 346–355 (1994).

    Article  CAS  PubMed  Google Scholar 

  34. Chipuk, J.E., Moldoveanu, T., Llambi, F., Parsons, M.J. & Green, D.R. The BCL-2 family reunion. Mol. Cell 37, 299–310 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Hollstein, M. et al. Database of p53 gene somatic mutations in human tumors and cell lines. Nucleic Acids Res. 22, 3551–3555 (1994).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Hernandez-Boussard, T., Rodriguez-Tome, P., Montesano, R. & Hainaut, P. IARC p53 mutation database: a relational database to compile and analyze p53 mutations in human tumors and cell lines. Hum. Mutat. 14, 1–8 (1999).

    Article  CAS  PubMed  Google Scholar 

  37. Bullock, A.N. et al. Thermodynamic stability of wild-type and mutant p53 core domain. Proc. Natl. Acad. Sci. USA 94, 14338–14342 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Inga, A. & Resnick, M.A. Novel human p53 mutations that are toxic to yeast can enhance transactivation of specific promoters and reactivate tumor p53 mutants. Oncogene 20, 3409–3419 (2001).

    Article  CAS  PubMed  Google Scholar 

  39. Bullock, A.N., Henckel, J. & Fersht, A.R. Quantitative analysis of residual folding and DNA binding in mutant p53 core domain: definition of mutant states for rescue in cancer therapy. Oncogene 19, 1245–1256 (2000).

    Article  CAS  PubMed  Google Scholar 

  40. Kato, S. et al. Understanding the function-structure and function-mutation relationships of p53 tumor suppressor protein by high-resolution missense mutation analysis. Proc. Natl. Acad. Sci. USA 100, 8424–8429 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Weinberg, R.L., Veprintsev, D.B. & Fersht, A.R. Cooperative binding of tetrameric p53 to DNA. J. Mol. Biol. 341, 1145–1159 (2004).

    Article  CAS  PubMed  Google Scholar 

  42. Hollstein, M., Sidransky, D., Vogelstein, B. & Harris, C.C. p53 mutations in human cancers. Science 253, 49–53 (1991).

    Article  CAS  PubMed  Google Scholar 

  43. Joo, W.S. et al. Structure of the 53BP1 BRCT region bound to p53 and its comparison to the Brca1 BRCT structure. Genes Dev. 16, 583–593 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Gorina, S. & Pavletich, N.P. Structure of the p53 tumor suppressor bound to the ankyrin and SH3 domains of 53BP2. Science 274, 1001–1005 (1996).

    Article  CAS  PubMed  Google Scholar 

  45. Lilyestrom, W., Klein, M.G., Zhang, R., Joachimiak, A. & Chen, X.S. Crystal structure of SV40 large T-antigen bound to p53: interplay between a viral oncoprotein and a cellular tumor suppressor. Genes Dev. 20, 2373–2382 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Kuwana, T. et al. Bid, Bax, and lipids cooperate to form supramolecular openings in the outer mitochondrial membrane. Cell 111, 331–342 (2002).

    Article  CAS  PubMed  Google Scholar 

  47. Asciolla, J.J., Renault, T.T. & Chipuk, J.E. Examining BCL-2 family function with large unilamellar vesicles. J. Vis. Exp. 10.3791/4291(5 October 2012).

  48. Llambi, F. et al. A unified model of mammalian BCL-2 protein family interactions at the mitochondria. Mol. Cell 44, 517–531 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Oltersdorf, T. et al. An inhibitor of Bcl-2 family proteins induces regression of solid tumours. Nature 435, 677–681 (2005).

    Article  CAS  PubMed  Google Scholar 

  50. Liu, Y. & Bodmer, W.F. Analysis of P53 mutations and their expression in 56 colorectal cancer cell lines. Proc. Natl. Acad. Sci. USA 103, 976–981 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Jones, S. & Thornton, J.M. Principles of protein-protein interactions. Proc. Natl. Acad. Sci. USA 93, 13–20 (1996).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Miyashita, T. & Reed, J.C. Tumor suppressor p53 is a direct transcriptional activator of the human bax gene. Cell 80, 293–299 (1995).

    Article  CAS  PubMed  Google Scholar 

  53. Wang, Y., Filippov, I., Richter, C., Luo, R. & Kriwacki, R.W. Solution NMR studies of an intrinsically unstructured protein within a dilute, 75 kDa eukaryotic protein assembly; probing the practical limits for efficiently assigning polypeptide backbone resonances. ChemBioChem 6, 2242–2246 (2005).

    Article  CAS  PubMed  Google Scholar 

  54. Bell, S., Hansen, S. & Buchner, J. Refolding and structural characterization of the human p53 tumor suppressor protein. Biophys. Chem. 96, 243–257 (2002).

    Article  CAS  PubMed  Google Scholar 

  55. Keller, R.J.J. Optimizing the process of nuclear magnetic resonance spectrum analysis and computer aided resonance assignment. PhD thesis, Eidgenossische Technishe Hochschule (2005).

  56. Güntert, P. Automated NMR structure calculation with CYANA. Methods Mol. Biol. 278, 353–378 (2004).

    PubMed  Google Scholar 

  57. Cornilescu, G., Delaglio, F. & Bax, A. Protein backbone angle restraints from searching a database for chemical shift and sequence homology. J. Biomol. NMR 13, 289–302 (1999).

    Article  CAS  PubMed  Google Scholar 

  58. Laskowski, R.A., Rullmannn, J.A., MacArthur, M.W., Kaptein, R. & Thornton, J.M. AQUA and PROCHECK-NMR: programs for checking the quality of protein structures solved by NMR. J. Biomol. NMR 8, 477–486 (1996).

    Article  CAS  PubMed  Google Scholar 

  59. de Vries, S.J., van Dijk, M. & Bonvin, A.M. The HADDOCK web server for data-driven biomolecular docking. Nat. Protoc. 5, 883–897 (2010).

    Article  CAS  PubMed  Google Scholar 

  60. Simon, B., Madl, T., Mackereth, C.D., Nilges, M. & Sattler, M. An efficient protocol for NMR-spectroscopy-based structure determination of protein complexes in solution. Angew. Chem. Int. Ed. Engl. 49, 1967–1970 (2010).

    Article  CAS  PubMed  Google Scholar 

  61. Wang, Y., Rosengarth, A. & Luecke, H. Structure of the human p53 core domain in the absence of DNA. Acta Crystallogr. D Biol. Crystallogr. 63, 276–281 (2007).

    Article  CAS  PubMed  Google Scholar 

  62. Reynolds, C., Damerell, D. & Jones, S. ProtorP: a protein-protein interaction analysis server. Bioinformatics 25, 413–414 (2009).

    Article  CAS  PubMed  Google Scholar 

  63. Saha, R.P., Bahadur, R.P., Pal, A., Mandal, S. & Chakrabarti, P. ProFace: a server for the analysis of the physicochemical features of protein-protein interfaces. BMC Struct. Biol. 6, 11 (2006).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  64. Gabdoulline, R.R., Wade, R.C. & Walther, D. MolSurfer: a macromolecular interface navigator. Nucleic Acids Res. 31, 3349–3351 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors acknowledge C.R. Grace (St. Jude Children's Research Hospital) for assistance with NMR experiments, C.-G. Park (St. Jude Children's Research Hospital) for assistance with protein preparation and isotope labeling and J. Chipuk (Mount Sinai Medical Center) for helpful discussion and advice. This work was supported by US National Institutes of Health (NIH) grants R01CA082491 and 1R01GM083159 (to R.W.K.), NIH R01GM52735 and R01GM96208 (to D.R.G.), US National Cancer Institute Cancer Center Support grant P30CA21765 (at St. Jude Children's Research Hospital) and the American Lebanese Syrian Associated Charities. A.V.F. is supported as the recipient of the Neoma Boadway Fellowship from St. Jude Children's Research Hospital.

Author information

Authors and Affiliations

Authors

Contributions

A.V.F. and L.O. contributed to NMR experiments and data analysis; A.V.F. contributed to structural calculations and biophysical and biochemical assays; A.V.F., F.L. and K.B. contributed to cellular assays; A.V.F., D.R.G. and R.W.K. wrote the manuscript. All authors contributed to different aspects of the design and conception of this study.

Corresponding authors

Correspondence to Douglas R Green or Richard W Kriwacki.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 NMR titrations between [15N]BCL-xLΔC and unlabeled p53 domains.

Overlaid full views of 1H-15N-TROSY spectra of 15N-BCL-xLΔC upon titration of unlabeled p53 NTD (a); p53 DBD (b); p53 N-D (c); p53 N-D-T (d); p53 TET (e). For panels a trough d the displayed spectra follow a blue to red transition color scheme at increasing p53 molar ratios. 15N-BCL-xLΔC concentration was 100 μM in all titrations (80 μM in e); p53 NTD (a) and p53 DBD (b) concentrations were 0, 10, 20, 40, 75, 110, 150, 200, 250, 300 μM; p53 N-D (c) concentrations were 0, 10, 25, 50, 75, 120, 180, 250, 300 μM; p53 N-D-T (d) concentrations were 0, 10, 25, 50, 75, 110, 150, 200 μM. f. Sequence plot of BCL-xLΔC resonance intensities relative to its free state in the presence of 180 μM p53 N-D (green trace) or 200 μM p53 N-D-T (purple trace).

Supplementary Figure 2 1H-15N TROSY titrations of unlabeled BCL-xLΔC into various 15N-labeled p53 constructs.

Titration of p53 NTD (a); p53 DBD (b) p53 N-D (c) p53 N-D-T (d). The displayed spectra follow a blue to red transition color scheme at increasing BCL-xLΔC molar ratios. The concentration of p53 NTD, p53 DBD and p53 N-D was 100 μM; that of p53 N-D-T 180 μM. BCL-xLΔC was titrated into 15N-p53 NTD (a) at 0, 50, 100, 200 μM; into 15N-p53 DBD (b) at 0, 25, 50, 100, 200, 300 μM; into p53 N-D (c) at 0, 25, 50, 100, 150, 200 μM; into p53 N-D-T (d) at 0 and 360 μM. e. Sequence plot of chemical shift perturbations detected in p53 N-D in the presence of 200 mM BCL-xLΔC. f. Mapping of the data displayed in panel e onto the p53 DBD structure indicating residues that showed backbone amide chemical shift perturbation > 0.06 ppm.

Supplementary Figure 3 Measurements of paramagnetic relaxation enhancements (PREs) for BCL-xLΔC–p53 DBD structure determination.

a. Overlaid 1H-15N-TROSY spectra of 15N-p53 DBD (100 μM) in the presence of reduced (blue) or oxidized (red) Cys151-MTSL labeled BCL-xLΔC (150 μM) and (b) of 15N-BCL-xLΔC (100 μM) in the presence of Zn2+ (blue) or Co2+-coordinating (red) p53 DBD (150 μM). c. Mapping onto the structure of p53 DBD of the paramagnetic relaxation enhancements (PREs) observed in a. d. Equivalent mapping of PREs observed in b onto the structure of BCL-xL.

Supplementary Figure 4 Intermolecular NOEs observed between [2H, 15N]p53 DBD and BCL-xLΔC at long NOE mixing time (160 ms).

The top two panels display slices of the 1H-15N-TROSY-NOESY 3D spectrum for selected resonances of 2H/15N-p53 DBD (450 μM) in the absence (a) and presence (b) of BCL-xLΔC at a 2:1 molar ratio. The bottom panel (c) displays slices of the 1H-15N-TROSY-TOCSY spectrum of free 15N-BCL-xL for the residues to which the observed inter-molecular NOEs were assigned. The inter-molecular NOESY peaks in the 3D NOESY spectrum were assigned by inference, with reference to the 15N-TROSY-TOCSY spectrum of free 15N-BCL-xL and with knowledge of preliminary structures based upon extensive PRE data. In a few cases, there are p53 binding-induced chemical shift changes between the two spectra (marked with an asterisk). d. Structure mapping of the p53 DBD (top) and BCL-xL (bottom) residues that exhibited intermolecular NOEs shown as spheres and color coded based on their reciprocal assignment.

Supplementary Figure 5 Conformational rearrangements take place within BCL-xL upon complex formation with p53 DBD.

a. Bundle of 20 lowest energy structures of apo BCL-xLΔC calculated with restraints obtained from 2H/13C/15N-BCL-xLΔC with selective 1H-labeling of Isoleucine, Leucine, Valine methyl moieties. b.. Equivalent representation of the BCL-xLΔC structure obtained from an identically labeled sample in the presence of p53 DBD. c. Overlay between the lowest energy structures in panels a (cyan) and b (pink). Flexible regions are omitted for clarity. A marked rearrangement of the α2-3 region of BCL-xL is observed upon binding of p53 DBD (highlighted with a red ellipse). d,e. Slices corresponding to the C-terminus of α2 of from the 1H-15N-TROSY-NOESY 3D spectrum of 2H/13C/15N-BCL-xLΔC with selective 1H-labeling of Isoleucine, Leucine, Valine methyl moieties in its apo form (d) and in the presence of p53 DBD (e) highlighting the subtle changes in local stabilization of α-helical structure that takes place in this region of BCL-xL upon binding of p53 DBD. Patterns of NOE cross-peaks indicative of a-helical structure are highlighted by dashed lines (red: i - i±1; yellow: longer range) and the approximate limits of strong a-helical NOE patterns are marked at the top of each panel along with residue labels. All samples contained each protein component at 600 μM. f. Sequence mapping of chemical shift-derived backbone dihedral restraints (calculated with the program Talos)58 for apo BCL-xLΔC (blue) and BCL-xLΔC bound to p53 DBD (red). The position of individual a-helices is marked above the plot. An increase in a-helical dihedral angles is observed at the C-terminus of α2 when BCL-xL is bound to p53-DBD.

Supplementary Figure 6 Structure-based mutagenesis of interfacial residues within the p53 DBD–BCL-xL complex.

a. Binding titrations between fluorescently labeled, wild-type BCL-xLΔC (F-BCL-xLΔC) and wild-type or mutated p53 N-D-T. Error bars, s.e.m. (n = 3 titrations). b. Mapping of the studied mutations on the p53 DBD surface depicted in the same orientation as Figure 4c. c. Competition experiments between F-BCL-xLΔC and unlabeled wild-type or mutated BCL-xLΔC for p53 binding. Error bars, s.e.m. (n = 3 titrations). d. Mapping of the studied mutations on the BCL-xL surface depicted in the same orientation as Figure 4d. e. Fluorescence polarization titration between F-BCL-xLΔC, pre-incubated with 1mM ABT-737 and p53 N-D-T. Error bars, s.e.m. (n = 3 titrations). f. Fluorescent polarization titration between F-BCL-xLΔC and full-length, wild-type human p53 expressed from Sf9 insect cells using a Baculovirus expression system (Bac-p53). Error bars, s.e.m. (n = 3 titrations). The measured binding affinity between these two proteins is equivalent to that between F-BCL-xLΔC and the p53 N-D-T bearing core stabilizing mutations.

Supplementary Figure 7 Binding titrations between fluorescently labeled, double-stranded oligonucleotides and wild-type or mutant p53 N-D-T.

a. Binding mode between p53 DBD (light grey) and DNA. The minimal binding unit is composed of one double stranded consensus sequence and two p53 DBD. Promoter regions in p53 target genes contain two closely spaced consensus sequences and are bound by p53 tetramers (in a 'dimer of dimers' fashion). b. Sequences of the studied oligonucleotides. The partial consensus segments are highlighted in bold characters in each sequence and nucleotides that differ between the two promoters are highlighted in red. The structural depictions at the bottom illustrate how sequence differences between the promoter regions correlate with residues within p53 whose mutation differentially altered the binding affinity for the two promoters. c. Fluorescence polarization binding titrations between wild-type or mutant p53 N-D-T and fluorescent p21 promoter. d. Analogous representation of binding titrations to the puma promoter. e. Structure representation of p53 bound to DNA highlighting interface regions and mutated residues that overlap with the p53 – BCL-xL binding interface. f. Competition experiments between F-BCL-xLΔC and an unlabeled p21 promoter double stranded oligonucleotide for p53 binding g. Fluorescence polarization titrations between Bac-p53 and fluorescently labeled DNA. Bac-p53 or p53 N-D-T display comparable binding affinities in vitro to these two oligonucleotides. Data in panels c,d,f,g represent the average of three independent titrations, error bars indicate s.e.m.

Supplementary Figure 8 Functional relevance of p53 mutations

a. Correlation between p21 promoter binding free energies and p21 transcription levels in yeast, as assessed by Kato et al.39, by wild-type and mutant p53. b. Equivalent analysis for the average DNA binding free energies (p21, puma promoter) and the average transcription levels in yeast of all eight p53 target genes analyzed by Kato et al.39. c. Western blot analysis of cytosolic (C), nuclear (N) and heavy membrane (HM) H1299 cell fractions transfected with empty vector or the indicated p53 constructs. Membranes were probed for p53, Glyceraldehyde 3-phosphate dehydrogenase (GAPDH, cytosolic marker) and Lamin B (nuclear marker). d. Effect of p53 mutations on its ability to activate BAX in vitro in a large unilamellar vesicle (LUV) permeabilization assay, analogous to the one reported in Figure 7a d but in the absence of BCL-xL inhibiting BAX activation. All tested mutants induced lower BAX activation compared to wild-type p53, indicating a possible overlap of interaction interfaces between BCL-xL or BAX and p53. Reduction in BAX activation was particularly pronounced for the H178D E180K double mutant, and roughly equivalent to the additive detrimental effect of each individual mutation. e. LUV permeabilization assay testing the ability of wild-type or mutant BCL-xLΔC to prevent liposome permeabilization by active BAX. Unlike the one displayed in Figure 7a, in this assay no agent was introduced to block the BH3 binding groove of BCL-xL (BAD BH3, ABT-737) therefore only limited LUV permeabilization was observed in the presence of wild-type or mutant BCL-xLΔC, likely because all these BCl-xL constructs are capable of sequestering partially active BAX by binding its exposed BH3 domain. f. LUV permeabilization assay testing the ability of wild-type or mutant BCL-xLΔC to prevent BAX activation by BIM or BID BH3 constructs. In this experiment, BIM-BID BH3 peptides or BIM-BID BH3-BCL-xL complexes were added to LUV samples at a lower concentration than the total amount of BAX present, therefore minimizing any potential effect of BCL-xL sequestration of active BAX. Both tested BCL-xL mutants showed equal ability to the wild-type protein to inhibit BIM-BID-dependent BAX activation.

Supplementary Figure 9 Full images of western blots displayed in cropped format in Figures 6 and 7

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–9, Supplementary Tables 1–3 and Supplementary Note (PDF 3012 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Follis, A., Llambi, F., Ou, L. et al. The DNA-binding domain mediates both nuclear and cytosolic functions of p53. Nat Struct Mol Biol 21, 535–543 (2014). https://doi.org/10.1038/nsmb.2829

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nsmb.2829

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