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:

Internal motions prime cIAP1 for rapid activation

Abstract

Cellular inhibitor of apoptosis 1 (cIAP1) is a ubiquitin ligase with critical roles in the control of programmed cell death and NF-κB signaling. Under normal conditions, the protein exists as an autoinhibited monomer, but proapoptotic signals lead to its dimerization, activation and proteasomal degradation. This view of cIAP1 as a binary switch has been informed by static structural studies that cannot access the protein's dynamics. Here, we use NMR spectroscopy to study micro- and millisecond motions of specific domain interfaces in human cIAP1 and use time-resolved small-angle X-ray scattering to observe the global conformational changes necessary for activation. Although motions within each interface of the 'closed' monomer are insufficient to activate cIAP1, they enable associations with catalytic partners and activation factors. We propose that these internal motions facilitate rapid peptide-induced opening and dimerization of cIAP1, which undergoes a dramatic spring-loaded structural transition.

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: E2 binding to monomeric and dimeric cIAP1.
Figure 2: AVPW binding kinetics.
Figure 3: NMR spectra.
Figure 4: R2 dispersion data.
Figure 5: Standard SAXS data of closed, open and dimeric forms of cIAP1.
Figure 6: Monitoring peptide-induced conformational changes of cIAP1 by TR-SAXS.

Similar content being viewed by others

Accession codes

Accessions

Protein Data Bank

References

  1. Hanahan, D. & Weinberg, R.A. Hallmarks of cancer: the next generation. Cell 144, 646–674 (2011).

    Article  CAS  Google Scholar 

  2. Fuchs, Y. & Steller, H. Programmed cell death in animal development and disease. Cell 147, 742–758 (2011).

    Article  CAS  Google Scholar 

  3. Flygare, J.A. & Fairbrother, W.J. Small-molecule pan-IAP antagonists: a patent review. Expert Opin. Ther. Pat. 20, 251–267 (2010).

    Article  CAS  Google Scholar 

  4. Fulda, S. & Vucic, D. Targeting IAP proteins for therapeutic intervention in cancer. Nat. Rev. Drug Discov. 11, 109–124 (2012).

    Article  CAS  Google Scholar 

  5. Du, C., Fang, M., Li, Y., Li, L. & Wang, X. Smac, a mitochondrial protein that promotes cytochrome c-dependent caspase activation by eliminating IAP inhibition. Cell 102, 33–42 (2000).

    Article  CAS  Google Scholar 

  6. Verhagen, A.M. et al. Identification of DIABLO, a mammalian protein that promotes apoptosis by binding to and antagonizing IAP proteins. Cell 102, 43–53 (2000).

    Article  CAS  Google Scholar 

  7. Liu, Z. et al. Structural basis for binding of Smac/DIABLO to the XIAP BIR3 domain. Nature 408, 1004–1008 (2000).

    Article  CAS  Google Scholar 

  8. Wu, G. et al. Structural basis of IAP recognition by Smac/DIABLO. Nature 408, 1008–1012 (2000).

    Article  CAS  Google Scholar 

  9. Vucic, D. et al. Engineering ML-IAP to produce an extraordinarily potent caspase 9 inhibitor: implications for Smac-dependent anti-apoptotic activity of ML-IAP. Biochem. J. 385, 11–20 (2005).

    Article  CAS  Google Scholar 

  10. Zobel, K. et al. Design, synthesis, and biological activity of a potent Smac mimetic that sensitizes cancer cells to apoptosis by antagonizing IAPs. ACS Chem. Biol. 1, 525–533 (2006).

    Article  CAS  Google Scholar 

  11. Varfolomeev, E. et al. IAP antagonists induce autoubiquitination of c-IAPs, NF-κB activation, and TNFα-dependent apoptosis. Cell 131, 669–681 (2007).

    Article  CAS  Google Scholar 

  12. Gaither, A. et al. A Smac mimetic rescue screen reveals roles for inhibitor of apoptosis proteins in tumor necrosis factor-α signaling. Cancer Res. 67, 11493–11498 (2007).

    Article  CAS  Google Scholar 

  13. Vince, J.E. et al. IAP antagonists target cIAP1 to induce TNFα-dependent apoptosis. Cell 131, 682–693 (2007).

    Article  CAS  Google Scholar 

  14. Bertrand, M.J.M. et al. cIAP1 and cIAP2 facilitate cancer cell survival by functioning as E3 ligases that promote RIP1 ubiquitination. Mol. Cell 30, 689–700 (2008).

    Article  CAS  Google Scholar 

  15. Varfolomeev, E. et al. c-IAP1 and c-IAP2 are critical mediators of tumor necrosis factor alpha (TNFα)-induced NF-κB activation. J. Biol. Chem. 283, 24295–24299 (2008).

    Article  CAS  Google Scholar 

  16. Plechanovová, A., Jaffray, E.G., Tatham, M.H., Naismith, J.H. & Hay, R.T. Structure of a RING E3 ligase and ubiquitin-loaded E2 primed for catalysis. Nature 489, 115–120 (2012).

    Article  Google Scholar 

  17. Dou, H., Buetow, L., Sibbet, G.J., Cameron, K. & Huang, D.T. BIRC7–E2 ubiquitin conjugate structure reveals the mechanism of ubiquitin transfer by a RING dimer. Nat. Struct. Mol. Biol. 19, 876–883 (2012).

    Article  CAS  Google Scholar 

  18. Mace, P.D. et al. Structures of the cIAP2 RING domain reveal conformational changes associated with ubiquitin-conjugating enzyme (E2) recruitment. J. Biol. Chem. 283, 31633–31640 (2008).

    Article  CAS  Google Scholar 

  19. Feltham, R. et al. Smac mimetics activate the E3 ligase activity of cIAP1 protein by promoting RING domain dimerization. J. Biol. Chem. 286, 17015–17028 (2011).

    Article  CAS  Google Scholar 

  20. Dueber, E.C. et al. Antagonists induce a conformational change in cIAP1 that promotes autoubiquitination. Science 334, 376–380 (2011).

    Article  CAS  Google Scholar 

  21. Lopez, J. et al. CARD-mediated autoinhibition of cIAP1′s E3 ligase activity suppresses cell proliferation and migration. Mol. Cell 42, 569–583 (2011).

    Article  CAS  Google Scholar 

  22. Kleckner, I.R. & Foster, M.P. An introduction to NMR-based approaches for measuring protein dynamics. Biochim. Biophys. Acta 1814, 942–968 (2011).

    Article  CAS  Google Scholar 

  23. Tugarinov, V. & Kay, L.E. Methyl groups as probes of structure and dynamics in NMR studies of high-molecular-weight proteins. ChemBioChem 6, 1567–1577 (2005).

    Article  CAS  Google Scholar 

  24. Gelis, I. et al. Structural basis for signal-sequence recognition by the translocase motor SecA as determined by NMR. Cell 131, 756–769 (2007).

    Article  CAS  Google Scholar 

  25. Martel, A., Liu, P., Weiss, T.M., Niebuhr, M. & Tsuruta, H. An integrated high-throughput data acquisition system for biological solution X-ray scattering studies. J. Synchrotron Radiat. 19, 431–434 (2012).

    Article  CAS  Google Scholar 

  26. Bernadó, P., Mylonas, E., Petoukhov, M.V., Blackledge, M. & Svergun, D.I. Structural characterization of flexible proteins using small-angle X-ray scattering. J. Am. Chem. Soc. 129, 5656–5664 (2007).

    Article  Google Scholar 

  27. Konarev, P.V., Volkov, V.V., Sokolova, A.V., Koch, M.H.J. & Svergun, D.I. PRIMUS: a Windows PC-based system for small-angle scattering data analysis. J. Appl. Crystallogr. 36, 1277–1282 (2003).

    Article  CAS  Google Scholar 

  28. Henzler-Wildman, K. & Kern, D. Dynamic personalities of proteins. Nature 450, 964–972 (2007).

    Article  CAS  Google Scholar 

  29. Lorenz, S., Cantor, A.J., Rape, M. & Kuriyan, J. Macromolecular juggling by ubiquitylation enzymes. BMC Biol. 11, 65 (2013).

    Article  Google Scholar 

  30. Krumbiegel, M., Herrmann, A. & Blumenthal, R. Kinetics of the low pH-induced conformational changes and fusogenic activity of influenza hemagglutinin. Biophys. J. 67, 2355–2360 (1994).

    Article  CAS  Google Scholar 

  31. Carr, C.M. & Kim, P.S. A spring-loaded mechanism for the conformational change of influenza hemagglutinin. Cell 73, 823–832 (1993).

    Article  CAS  Google Scholar 

  32. Ohno, A. et al. Structure of the UBA domain of Dsk2p in complex with ubiquitin molecular determinants for ubiquitin recognition. Structure 13, 521–532 (2005).

    Article  CAS  Google Scholar 

  33. Brzovic, P.S., Lissounov, A., Christensen, D.E., Hoyt, D.W. & Klevit, R.E.A. UbcH5/ubiquitin noncovalent complex is required for processive BRCA1-directed ubiquitination. Mol. Cell 21, 873–880 (2006).

    Article  CAS  Google Scholar 

  34. Pruneda, J.N. et al. Structure of an E3:E2~Ub complex reveals an allosteric mechanism shared among RING/U-box ligases. Mol. Cell 47, 933–942 (2012).

    Article  CAS  Google Scholar 

  35. Bentley, M.L. et al. Recognition of UbcH5c and the nucleosome by the Bmi1/Ring1b ubiquitin ligase complex. EMBO J. 30, 3285–3297 (2011).

    Article  CAS  Google Scholar 

  36. Dong, K.C. et al. Preparation of distinct ubiquitin chain reagents of high purity and yield. Structure 19, 1053–1063 (2011).

    Article  CAS  Google Scholar 

  37. Smith, P.A. et al. A plasmid expression system for quantitative in vivo biotinylation of thioredoxin fusion proteins in Escherichia coli. Nucleic Acids Res. 26, 1414–1420 (1998).

    Article  CAS  Google Scholar 

  38. Cai, M. et al. An efficient and cost-effective isotope labeling protocol for proteins expressed in Escherichia coli. J. Biomol. NMR 11, 97–102 (1998).

    Article  CAS  Google Scholar 

  39. Isotope labeling strategies for the study of high-molecular-weight proteins by solution NMR spectroscopy. Nat. Protoc. 1, 749–754 (2006).

  40. Tollinger, M., Skrynnikov, N.R., Mulder, F.A., Forman-Kay, J.D. & Kay, L.E. Slow dynamics in folded and unfolded states of an SH3 domain. J. Am. Chem. Soc. 123, 11341–11352 (2001).

    Article  CAS  Google Scholar 

  41. Carver, J.P. & Richards, R. E. A general two-site solution for the chemical exchange produced dependence of T2 upon the Carr-Purcell pulse separation. J. Magn. Reson. 6, 89–105 (1972).

    CAS  Google Scholar 

  42. Kleckner, I.R. & Foster, M.P. GUARDD: user-friendly MATLAB software for rigorous analysis of CPMG RD NMR data. J. Biomol. NMR 52, 11–22 (2012).

    Article  CAS  Google Scholar 

  43. Tsuruta, H. et al. A wide-bandpass multilayer monochromator for biological small-angle scattering and fiber diffraction studies. J. Appl. Crystallogr. 31, 672–682 (1998).

    Article  CAS  Google Scholar 

  44. Svergun, D.I. Determination of the regularization parameter in indirect-transform methods using perceptual criteria. J. Appl. Crystallogr. 25, 495–503 (1992).

    Article  CAS  Google Scholar 

  45. Svergun, D.I., Petoukhov, M.V. & Koch, M. Determination of domain structure of proteins from X-ray solution scattering. Biophys. J. 80, 2946–2953 (2001).

    Article  CAS  Google Scholar 

  46. Volkov, V.V. & Svergun, D.I. Uniqueness of ab initio shape determination in small-angle scattering. J. Appl. Crystallogr. 36, 860–864 (2003).

    Article  CAS  Google Scholar 

  47. Petoukhov, M.V. & Svergun, D.I. Global rigid body modeling of macromolecular complexes against small-angle scattering data. Biophys. J. 89, 1237–1250 (2005).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank R. Klevit for helpful discussions and A. Taherbhoy for assistance in preparing ubiquitin-charged E2. We acknowledge use of the Central California 900-MHz Facility (supported by US National Institutes of Health (NIH) grant GM68933) and thank D. Wemmer and J. Pelton for assistance with the facility. Use of the Stanford Synchrotron Radiation Lightsource (SSRL), Stanford Linear Accelerator Center (SLAC) US National Accelerator Laboratory, is supported by the US Department of Energy (DOE), Office of Science, Office of Basic Energy Sciences under contract no. DE-AC02-76SF00515. The SSRL Structural Molecular Biology Program is supported by the DOE Office of Biological and Environmental Research and by the NIH National Institute of General Medical Sciences (NIGMS) (including grant P41GM103393). The contents of this publication are solely the responsibility of the authors and do not necessarily represent the official views of NIGMS or NIH.

Author information

Authors and Affiliations

Authors

Contributions

A.H.P. designed and performed NMR experiments, analyzed data and wrote the paper. A.J.S. designed and performed SAXS and biochemical experiments (except surface plasmon resonance experiments), analyzed data and wrote the paper. T.M. and T.M.W. designed the specialized stopped-flow SAXS instrumentation and performed SAXS experiments. K.Z. prepared peptide reagents. A.M.G. performed surface plasmon resonance experiments. J.W.B. performed selected NMR experiments. E.C.D. and W.J.F. supervised the work, designed experiments and contributed to the writing of the paper.

Corresponding authors

Correspondence to Erin C Dueber or Wayne J Fairbrother.

Ethics declarations

Competing interests

A.J.S., A.M.G., K.Z., E.C.D. and W.J.F. are all employed by Genentech, a member of the Roche group. J.W.B. is employed by Emergent Biosolutions.

Integrated supplementary information

Supplementary Figure 1 Definition and assessment of cIAP1 constructs.

(a) cIAP1 constructs used in this study are shown as primary structure schematics with domains colored as in the main text. Mutations and deletions are indicated with magenta lines. The mutations in the cIAP1-B3R construct reduce proteolysis and non-native disulfide formation1. (b) The binding of cIAP1-B3R (blue) and cIAP1-B3RMF-AA (gray) to monobiotinylated ubiquitin was tested using biolayer interferometry. Equilibrium response values are plotted against cIAP1 concentration. No detectable binding is observed by the cIAP1-B3RMF-AA construct. cIAP1-B3R binds ubiquitin with a KD of approximately 20 μM as determined by a single-site binding isotherm fit, in good agreement with past studies2. Recent reports have suggested that this mutation destabilizes cIAP13, which might contribute to the very slight decrease in E2 binding affinity we observe in the MF-AA variants. The use of cIAP1MF-AA in E2 binding studies removes the complicating effects of direct cIAP1-ubiquitin binding in the case of the E2-Ub conjugate. (c) The binding of cIAP1-B3R (blue) and cIAP1-B3R-ΔCARD (green) to E2SRCK was assessed using biolayer interferometry. Equilibrium response values were plotted as a function of E2 concentration, and the resulting curves were fit to a single-site binding isotherm. The affinities determined are approximately 27 μM (cIAP1-B3R) and 19 μM (cIAP1-B3R-ΔCARD), recapitulating the relative increase in affinity upon CARD deletion observed in the case of cIAP1-B3RMF-AA.

Supplementary Figure 2 UbcH5c does not induce dimerization of cIAP1.

(a) cIAP1-B3R responds to SMAC mimetics by dimerizing, as detected by native gel electrophoresis. Lane 1 shows a native molecular weight marker (Native Mark, Invitrogen) with molecular weights indicated at left. Lane 2 shows monomeric cIAP1 (64 μM). Lane 3 shows dimerized cIAP1 in the presence of AVPW (1 mM). Binding of the bivalent SMAC mimetic BV6 (1 mM) induces stronger and more compact dimerization because it can engage two BIR3 domains simultaneously. Lanes 4-7 demonstrate that UbcH5c (64 μM) does not affect the dimerization state of cIAP1. Lanes 8-10 demonstrate that the gel mobility of UbcH5c is not affected by the SMAC mimetics. All samples contain 1% DMSO. (b) cIAP1-B3R (64 μM) was incubated with UbcH5c at 0 to 128 μM. No detectable dimer is formed. cIAP1 bound to BV6 is included as a dimerization control.

Supplementary Figure 3 Methionine methyl groups assigned by mutagenesis.

1H-13C HMQC spectra of each methionine to leucine point mutant (blue) overlaid with the cIAP1-B3R spectrum (red). The M466L mutant is also shown with the contours drawn four times lower. The M266L, M391L, M392L, and M402L spectra display secondary chemical shift changes that may be due to structural perturbations as a result of the mutation.

Supplementary Figure 4 Dispersion profiles of UBA α3.

The residues in UBA α3 were fit to a single exchange process with kex = 970 ± 100 s-1 and pB = 2.2 ± 0.1 %. Data and fits at 900 MHz are shown in red, 800 MHz data and fits are in blue. Error bars were determined by treating the spectral noise level as the uncertainty in peak heights. The two residues in UBA α3 that do not have detectable R2 dispersion, Q411 and L418, are severely overlapped in the 1H-15N spectra. As shown in Fig. 4a, R2 dispersion was also detected at additional residues in the UBA (A388, V389, M390, A399, F404, K425, D429 and I430) as well as at five additional residues outside the UBA domain (L359, T534, I548, V581 and I608); however, as the residues do not cluster into large, contiguous surfaces we did not fit the data to extract kinetic or equilibrium parameters describing the exchange processes.

Supplementary Figure 5 Characterization of the conformational states of cIAP1 variants.

(a) Size-exclusion multi-angle light scattering (SEC-MALS) traces of cIAP1 constructs with and without SMAC mimetics. Apo-proteins are shown in black, AVPW-bound protein in blue, and BV6 bound proteins (as a dimerization control) in red. The UV absorbance for each peak was normalized to 1. As expected, cIAP1-B3R (top) dimerizes in response to both AVPW and BV6, while cIAP1-B3R-ΔCARD (center) is constitutively open and dimerizes only in the presence of BV6. The L617E construct (bottom) blocks AVPW-induced dimerization without disrupting the closed conformation. (b) L617 is positioned in the RING dimerization interface. The structure of the cIAP2 RING dimer (PDB code: 3EB5, ref. 4) is shown in cartoon representation with the homologous residue to L617, L603, colored magenta and shown as spheres. Zinc ions are shown as light gray spheres.

Supplementary Figure 6 Standard SAXS models.

Averaged, filtered SAXS models from 10 independent ab initio calculations are shown for cIAP1-B3R, cIAP1-B3R-∆C7 and cIAP1-B3RL617E with and without AVPW. Fits (dark blue lines) of the raw scattering data (red circles) to the best ab initio model from each set are shown. Chi values for those fits are also shown. Models were generated as described (see Methods) using GASBOR.

Supplementary Figure 7 Molecular models based on standard cIAP1 SAXS data.

(a) The best of ten molecular models of cIAP1-B3R-ΔC7 is shown superposed with the averaged, filtered ab initio model. A normalized spatial discrepancy (NSD) for the aligment is indicated. The fit (dark blue line) of the molecular model to the experimental data (red circles) is shown below the model, and the Chi value describing the fit is indicated. I stands for scattering intensity and q is proportional to the scattering angle (q = 4 sin(θ)/λ, where 2θ = the angle between the incident X-ray beam and the detector, and λ = the X-ray wavelength in Ångstroms). (b) The same data as in (a) is shown for cIAP1-B3R-ΔC7 in the presence of 1 mM AVPW. (c, d) Constraints used in the generation of closed monomeric molecular models are shown. All constraints are based on mutational data1. (e) The best of ten molecular models of cIAP1-B3R is shown superposed with the averaged, filtered ab initio model, in two views. The NSD between the two models is indicated. (f) The fit of the best cIAP1-B3R molecular model to the experimental data is shown. All colors and variables are as in (a). (g) A subset of calculated molecular models for cIAP1-B3R is shown aligned by their BIR3 domains to demonstrate the heterogeneity of the position of the CARD. The protein likely exists as an ensemble of states not fully reflected by any single model. (h) The distributions of Rg values in the ensembles of molecular models generated by EOM for cIAP1-B3R (black), cIAP1-B3RL617E (orange), cIAP1-B3R-ΔC7 (violet) and cIAP1-B3RL617E + AVPW (green) are shown. The initial, unoptimized pools are displayed as dotted lines, and the optimized ensembles as solid lines. Note that the closed states (cIAP1-B3R and cIAP1-B3RL617E) adopt tighter and smaller distributions than the open states (cIAP1-B3R-ΔC7 and cIAP1-B3RL617E + AVPW), reflecting the more rigid conformation of the closed states.

Supplementary Figure 8 Controls for TR-SAXS measurements.

(a,b) Time-resolved SAXS (TR-SAXS) experiments of cIAP1-B3R and cIAP1-B3RL617E mixed with buffer instead of AVPW. (c, d) TR-SAXS experiment of cIAP1-B3R-∆C7 mixed (c) with buffer and (d) with AVPW. (e) TR-SAXS analysis of cIAP1-B3RL617E with a higher concentration of AVPW (1 mM). The relative populations of the monomeric, open, and dimeric conformations of cIAP1 were extracted from the TR-SAXS data by deconvolution using the static scattering curves of each state as reference (See Methods). Each point represents and average from three experiments, plus or minus standard deviation. Data are fit to first order integrated rate equations. The closed monomer fraction is shown as blue squares, open fraction as hot pink circles, dimer fraction as purple diamonds.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–8 and Supplementary Tables 1–3 (PDF 1897 kb)

Supplementary Data Set 1

Uncropped gels from Figure 1d,e and Supplementary Figure 2 (PDF 15563 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Phillips, A., Schoeffler, A., Matsui, T. et al. Internal motions prime cIAP1 for rapid activation. Nat Struct Mol Biol 21, 1068–1074 (2014). https://doi.org/10.1038/nsmb.2916

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nsmb.2916

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