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:

Axin cancer mutants form nanoaggregates to rewire the Wnt signaling network

Abstract

Signaling cascades depend on scaffold proteins that regulate the assembly of multiprotein complexes. Missense mutations in scaffold proteins are frequent in human cancer, but their relevance and mode of action are poorly understood. Here we show that cancer point mutations in the scaffold protein Axin derail Wnt signaling and promote tumor growth in vivo through a gain-of-function mechanism. The effect is conserved for both the human and Drosophila proteins. Mutated Axin forms nonamyloid nanometer-scale aggregates decorated with disordered tentacles, which 'rewire' the Axin interactome. Importantly, the tumor-suppressor activity of both the human and Drosophila Axin cancer mutants is rescued by preventing aggregation of a single nonconserved segment. Our findings establish a new paradigm for misregulation of signaling in cancer and show that targeting aggregation-prone stretches in mutated scaffolds holds attractive potential for cancer treatment.

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: Axin RGS missense mutations induce Wnt-pathway activation and structural destabilization.
Figure 2: Tumorigenic activity of Axin mutants depends on newly acquired properties of the destabilized RGS domain.
Figure 3: Axin L106R RGS forms oligomers with disordered extensions.
Figure 4: Interference with oligomerization rescues tumor-suppressor activity of Axin L106R.
Figure 5: TANGO prediction of aggregation-prone sequences in human and Drosophila Axin RGS.
Figure 6: In vivo temperature-dependent hyperplastic growth induced by Drosophila Axin cancer mutants is rescued by aggregon mutations.
Figure 7: Model for the mechanism of action of Axin RGS cancer variants.

Similar content being viewed by others

Accession codes

Accessions

NCBI Reference Sequence

Protein Data Bank

References

  1. Good, M.C., Zalatan, J.G. & Lim, W.A. Scaffold proteins: hubs for controlling the flow of cellular information. Science 332, 680–686 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Cortese, M.S., Uversky, V.N. & Dunker, A.K. Intrinsic disorder in scaffold proteins: getting more from less. Prog. Biophys. Mol. Biol. 98, 85–106 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Luo, W. & Lin, S.C. Axin: a master scaffold for multiple signaling pathways. Neurosignals 13, 99–113 (2004).

    Article  CAS  PubMed  Google Scholar 

  4. Noutsou, M. et al. Critical scaffolding regions of the tumor suppressor Axin1 are natively unfolded. J. Mol. Biol. 405, 773–786 (2011).

    Article  CAS  PubMed  Google Scholar 

  5. Mark, W.Y. et al. Characterization of segments from the central region of BRCA1: an intrinsically disordered scaffold for multiple protein-protein and protein-DNA interactions? J. Mol. Biol. 345, 275–287 (2005).

    Article  CAS  PubMed  Google Scholar 

  6. Minde, D.P., Anvarian, Z., Rüdiger, S.G.D. & Maurice, M.M. Messing up disorder: how do missense mutations in the tumor suppressor protein APC lead to cancer? Mol. Cancer 10, 101 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Szabo, C.I., Worley, T. & Monteiro, A.N. Understanding germ-line mutations in BRCA1. Cancer Biol. Ther. 3, 515–520 (2004).

    Article  CAS  PubMed  Google Scholar 

  8. Salahshor, S. & Woodgett, J.R. The links between axin and carcinogenesis. J. Clin. Pathol. 58, 225–236 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Polakis, P. The many ways of Wnt in cancer. Curr. Opin. Genet. Dev. 17, 45–51 (2007).

    Article  CAS  PubMed  Google Scholar 

  10. Hart, M.J., de los Santos, R., Albert, I.N., Rubinfeld, B. & Polakis, P. Downregulation of beta-catenin by human Axin and its association with the APC tumor suppressor, beta-catenin and GSK3 beta. Curr. Biol. 8, 573–581 (1998).

    Article  CAS  PubMed  Google Scholar 

  11. Ikeda, S. et al. Axin, a negative regulator of the Wnt signaling pathway, forms a complex with GSK-3beta and beta-catenin and promotes GSK-3beta-dependent phosphorylation of beta-catenin. EMBO J. 17, 1371–1384 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Itoh, K., Krupnik, V.E. & Sokol, S.Y. Axis determination in Xenopus involves biochemical interactions of axin, glycogen synthase kinase 3 and beta-catenin. Curr. Biol. 8, 591–594 (1998).

    Article  CAS  PubMed  Google Scholar 

  13. Sakanaka, C., Weiss, J.B. & Williams, L.T. Bridging of beta-catenin and glycogen synthase kinase-3beta by axin and inhibition of beta-catenin-mediated transcription. Proc. Natl. Acad. Sci. USA 95, 3020–3023 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Clevers, H. & Nusse, R. Wnt/β-catenin signaling and disease. Cell 149, 1192–1205 (2012).

    Article  CAS  PubMed  Google Scholar 

  15. MacDonald, B.T., Tamai, K. & He, X. Wnt/beta-catenin signaling: components, mechanisms, and diseases. Dev. Cell 17, 9–26 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Reya, T. & Clevers, H. Wnt signalling in stem cells and cancer. Nature 434, 843–850 (2005).

    Article  CAS  PubMed  Google Scholar 

  17. Dow, L.E. et al. Apc restoration promotes cellular differentiation and reestablishes crypt homeostasis in colorectal cancer. Cell 161, 1539–1552 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Spink, K.E., Polakis, P. & Weis, W.I. Structural basis of the Axin-adenomatous polyposis coli interaction. EMBO J. 19, 2270–2279 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Schwarz-Romond, T. et al. The DIX domain of Dishevelled confers Wnt signaling by dynamic polymerization. Nat. Struct. Mol. Biol. 14, 484–492 (2007).

    Article  CAS  PubMed  Google Scholar 

  20. Taniguchi, K. et al. Mutational spectrum of beta-catenin, AXIN1, and AXIN2 in hepatocellular carcinomas and hepatoblastomas. Oncogene 21, 4863–4871 (2002).

    Article  CAS  PubMed  Google Scholar 

  21. Webster, M.T. et al. Sequence variants of the axin gene in breast, colon, and other cancers: an analysis of mutations that interfere with GSK3 binding. Genes Chromosom. Cancer 28, 443–453 (2000).

    Article  CAS  PubMed  Google Scholar 

  22. Shimizu, Y. et al. Frequent alterations in the Wnt signaling pathway in colorectal cancer with microsatellite instability. Genes Chromosom. Cancer 33, 73–81 (2002).

    Article  CAS  PubMed  Google Scholar 

  23. Jin, L.H. et al. Detection of point mutations of the Axin1 gene in colorectal cancers. Int. J. Cancer 107, 696–699 (2003).

    Article  CAS  PubMed  Google Scholar 

  24. Major, M.B. et al. Wilms tumor suppressor WTX negatively regulates WNT/beta-catenin signaling. Science 316, 1043–1046 (2007).

    Article  CAS  PubMed  Google Scholar 

  25. Peterson-Nedry, W. et al. Unexpectedly robust assembly of the Axin destruction complex regulates Wnt/Wg signaling in Drosophila as revealed by analysis in vivo. Dev. Biol. 320, 226–241 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Oosterveen, T. et al. Two functionally distinct Axin-like proteins regulate canonical Wnt signaling in C. elegans. Dev. Biol. 308, 438–448 (2007).

    Article  CAS  PubMed  Google Scholar 

  27. Wüthrich, K. NMR studies of structure and function of biological macromolecules (Nobel Lecture). J. Biomol. NMR 27, 13–39 (2003).

    Article  PubMed  Google Scholar 

  28. Schwarz-Romond, T., Merrifield, C., Nichols, B.J. & Bienz, M. The Wnt signalling effector Dishevelled forms dynamic protein assemblies rather than stable associations with cytoplasmic vesicles. J. Cell Sci. 118, 5269–5277 (2005).

    Article  CAS  PubMed  Google Scholar 

  29. Johnston, J.A., Ward, C.L. & Kopito, R.R. Aggresomes: a cellular response to misfolded proteins. J. Cell Biol. 143, 1883–1898 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Kawaguchi, Y. et al. The deacetylase HDAC6 regulates aggresome formation and cell viability in response to misfolded protein stress. Cell 115, 727–738 (2003).

    Article  CAS  PubMed  Google Scholar 

  31. Fernandez-Escamilla, A.M., Rousseau, F., Schymkowitz, J. & Serrano, L. Prediction of sequence-dependent and mutational effects on the aggregation of peptides and proteins. Nat. Biotechnol. 22, 1302–1306 (2004).

    Article  CAS  PubMed  Google Scholar 

  32. Jensen, L.J. et al. STRING 8: a global view on proteins and their functional interactions in 630 organisms. Nucleic Acids Res. 37, D412–D416 (2009).

    Article  CAS  PubMed  Google Scholar 

  33. Menssen, R. et al. Exploring the topology of the Gid complex, the E3 ubiquitin ligase involved in catabolite-induced degradation of gluconeogenic enzymes. J. Biol. Chem. 287, 25602–25614 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Francis, O., Han, F. & Adams, J.C. Molecular phylogeny of a RING E3 ubiquitin ligase, conserved in eukaryotic cells and dominated by homologous components, the muskelin/RanBPM/CTLH complex. PLoS One 8, e75217 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Baena-Lopez, L.A., Alexandre, C., Mitchell, A., Pasakarnis, L. & Vincent, J.P. Accelerated homologous recombination and subsequent genome modification in Drosophila. Development 140, 4818–4825 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Vincent, J.P., Kolahgar, G., Gagliardi, M. & Piddini, E. Steep differences in wingless signaling trigger Myc-independent competitive cell interactions. Dev. Cell 21, 366–374 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Eisenberg, D. & Jucker, M. The amyloid state of proteins in human diseases. Cell 148, 1188–1203 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Chiti, F. & Dobson, C.M. Protein misfolding, functional amyloid, and human disease. Annu. Rev. Biochem. 75, 333–366 (2006).

    Article  CAS  PubMed  Google Scholar 

  39. Gillis, J. et al. The DNAJB6 and DNAJB8 protein chaperones prevent intracellular aggregation of polyglutamine peptides. J. Biol. Chem. 288, 17225–17237 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Bullock, A.N. & Fersht, A.R. Rescuing the function of mutant p53. Nat. Rev. Cancer 1, 68–76 (2001).

    Article  CAS  PubMed  Google Scholar 

  41. Freed-Pastor, W.A. & Prives, C. Mutant p53: one name, many proteins. Genes Dev. 26, 1268–1286 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Xu, J. et al. Gain of function of mutant p53 by coaggregation with multiple tumor suppressors. Nat. Chem. Biol. 7, 285–295 (2011).

    Article  CAS  PubMed  Google Scholar 

  43. Rowling, P.J., Cook, R. & Itzhaki, L.S. Toward classification of BRCA1 missense variants using a biophysical approach. J. Biol. Chem. 285, 20080–20087 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Stehr, H. et al. The structural impact of cancer-associated missense mutations in oncogenes and tumor suppressors. Mol. Cancer 10, 54 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  45. Willert, K. et al. Wnt proteins are lipid-modified and can act as stem cell growth factors. Nature 423, 448–452 (2003).

    Article  CAS  PubMed  Google Scholar 

  46. Tauriello, D.V. et al. Loss of the tumor suppressor CYLD enhances Wnt/beta-catenin signaling through K63-linked ubiquitination of Dvl. Mol. Cell 37, 607–619 (2010).

    Article  CAS  PubMed  Google Scholar 

  47. Cabrita, L.D. et al. Enhancing the stability and solubility of TEV protease using in silico design. Protein Sci. 16, 2360–2367 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Rudiger, S., Freund, S.M., Veprintsev, D.B. & Fersht, A.R. CRINEPT-TROSY NMR reveals p53 core domain bound in an unfolded form to the chaperone Hsp90. Proc. Natl. Acad. Sci. USA 99, 11085–11090 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Nolo, R., Abbott, L.A. & Bellen, H.J. Senseless, a Zn finger transcription factor, is necessary and sufficient for sensory organ development in Drosophila. Cell 102, 349–362 (2000).

    Article  CAS  PubMed  Google Scholar 

  50. Duncan, D.M., Burgess, E.A. & Duncan, I. Control of distal antennal identity and tarsal development in Drosophila by spineless-aristapedia, a homolog of the mammalian dioxin receptor. Genes Dev. 12, 1290–1303 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Bergmann, A., Fritz, G. & Glatter, O. Solving the generalized indirect Fourier transformation (GIFT) by Boltzmann simplex simulated annealing (BSSA). J. Appl. Crystallogr. 33, 1212–1216 (2000).

    Article  CAS  Google Scholar 

  52. 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 

  53. Guinier, A. La diffraction des rayons X aux très petits angles: application à l'étude de phénomènes ultramicroscopiques. Ann. Phys. 12, 161–237 (1939).

    Article  CAS  Google Scholar 

  54. Svergun, D., Barberato, C. & Koch, M.H.J. CRYSOL: a program to evaluate X-ray solution scattering of biological macromolecules from atomic coordinates. J. Appl. Crystallogr. 28, 768–773 (1995).

    Article  CAS  Google Scholar 

  55. Demeler, B., Brookes, E. & Nagel-Steger, L. Analysis of heterogeneity in molecular weight and shape by analytical ultracentrifugation using parallel distributed computing. Methods Enzymol. 454, 87–113 (2009).

    Article  CAS  PubMed  Google Scholar 

  56. Hayes, D.B. & Stafford, W.F. SEDVIEW: real-time sedimentation analysis. Macromol. Biosci. 10, 731–735 (2010).

    Article  CAS  PubMed  Google Scholar 

  57. Frese, C.K. et al. Improved peptide identification by targeted fragmentation using CID, HCD and ETD on an LTQ-Orbitrap Velos. J. Proteome Res. 10, 2377–2388 (2011).

    Article  CAS  PubMed  Google Scholar 

  58. Mellacheruvu, D. et al. The CRAPome: a contaminant repository for affinity purification-mass spectrometry data. Nat. Methods 10, 730–736 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Choi, H. et al. SAINT: probabilistic scoring of affinity purification–mass spectrometry data. Nat. Methods 8, 70–73 (2011).

    Article  CAS  PubMed  Google Scholar 

  60. Vizcaíno, J.A. et al. ProteomeXchange provides globally coordinated proteomics data submission and dissemination. Nat. Biotechnol. 32, 223–226 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

Download references

Acknowledgements

We thank members of the laboratories of M.M.M. and S.G.D.R. for experimental support, helpful discussions and suggestions. We thank B. Kleizen and I. Braakman (Cellular Protein Chemistry, Bijvoet Centre for Biomolecular Research, Utrecht University) for providing the CFTR ΔF508 construct and H. Bellen (Baylor College of Medicine), I. Duncan and D. Duncan (Washington University) for antibodies. This work was supported by the European Research Council ((ERC) starting grant 242958 to M.M.M. and ERC advanced grant 294523 to J.-P.V.); Utrecht University (High Potential Grants to M.M.M. and S.G.D.R.); Boehringer Ingelheim Fonds (PhD fellowship to E.C.v.K.); the European Union (Framework Programme (FP) 7 Marie Curie ITN 608180 'WntsApp' to M.M.M. and S.G.D.R., FP7 Marie Curie ITN-IDP 317371 'ManiFold' to S.G.D.R., FP6 Marie Curie Excellence Grant 25651 'chaperoning cascades' to S.G.D.R. and BioNMR project 261863 to R.B. and T.M.); the Netherlands Organization for Scientific Research ((NWO) VICI grant to M.M.M., Vidi career development grant to S.G.D.R. and instrumentation support for a TCI probe to R.B.); the Internationale Stichting Alzheimer Onderzoek ((ISAO) grant to S.G.D.R.); the Medical Research Council of Great Britain (grant U117584268 to J.P.V.); the European Molecular Biology Organization ((EMBO) ALTF 983-2009 to H.N.); the Uehara and Kanae Foundations (to H.N.); the Austrian Academy of Sciences (APART-fellowship to T.M.); the Bavarian Ministry of Sciences, Research and the Arts in the framework of the Bavarian Molecular Biosystems Research Network (to T.M.); and the German Research Foundation (Emmy Noether program MA 5703/1-1 to T.M.). T.Y.L. and A.J.R.H. were supported by the NWO embedded roadmap program Proteins@Work (project 184.032.201) and PRIME-XS, grant number 262067, funded by the European Union FP7. We thank the Deutsches Elektronen Synchrotron (DESY) synchrotron radiation facilities for support of the SAXS data collection and B. Demeler for providing a license for the UltraScan software package.

Author information

Authors and Affiliations

Authors

Contributions

Z.A., H.N., E.C.v.K., J.-P.V., S.G.D.R. and M.M.M. conceived and designed the experiments. Z.A., H.N., E.C.v.K., T.B., M.S., I.J., M.V., T.Y.L., R.C.v.S., I.K. and K.R. performed the experiments. Z.A., H.N., E.C.v.K., I.J., T.Y.L., M.V., T.M., R.B., A.J.R.H., J.-P.V., S.G.D.R. and M.M.M. analyzed the data. Z.A., J.-P.V., E.C.v.K., S.G.D.R. and M.M.M. wrote the manuscript. The other authors commented on the manuscript.

Corresponding authors

Correspondence to Stefan G D Rüdiger or Madelon M Maurice.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Alignment of Axin RGS domains and structural consequences of mutation-induced destabilization.

(a) Alignment of Axin RGS domain amino acid sequences of Homo sapiens and other indicated species. Mutated residues found in cancer patients are indicated in red and show full, partial or no conservation. (b) CD spectra of purified wild-type (WT) and mutant RGS domains are shown at the indicated temperatures. At low temperatures all CD spectra, with the exception of RGS-L106R, exhibit typical α-helical profiles showing two peak minima at 208 and 220 nm and a peak maximum at 190 nm. For each of these mutants, loss of α-helical structure is shown at or above the unfolding temperature that were calculated from fluorescence-based thermal denaturation (Fig. 1f). CD spectra of RGS-L106R do not display α-helical content and remain unaffected by temperature change. Results represent two independent experiments. (c) 2D NMR spectra showing signals of RGS-L106R. As compared to the 2D NMR spectrum of RGS-wt (Fig. 3b), Trp side chain signals (purple squares) as well as signals from folded regions (grey squares) are lost for RGS-L106R, indicating loss of regular 3D structure. By contrast, signals from disordered termini of the protein remain in place (blue squares).

Supplementary Figure 2 Characterization of Axin L106R aggresome-like structures and analysis of SDS solubility.

(a) Quantification of the percentage of Axin-L106R expressing cells carrying an aggresome-like structure. Per field >50 cells were counted. Error bars represent SDs of 7 fields (representing >350 cells) per condition. (b) γ-tubulin staining of cells expressing Axin-wt, Axin-L106R and Axin-L106R F119R reveals that a fraction of Axin-L106R and Axin-L106R F119R (Axin-L106R (F>R) proteins accumulate in a pericentriolar structure. Scale bars 10 μm. (c) Pericentriolar accumulations of Axin-L106R do not display enrichment of HDAC6 or vimentin, in contrast to previously described aggresomes formed by mutant CFTR-ΔF508 protein29. Scale bars 10 μm. (d) Axin-L106R is fully soluble in SDS-based lysis buffer (SDS), similar to the Axin-wt (WT) protein. SDS-resistant pellet fractions (dissolved in formic acid (FA)) were devoid of Axin protein. Actin control is shown for comparison.

Supplementary Figure 3 Excess Axin WT suppresses Axin L106R–induced β-catenin–mediated transcription.

Wnt luciferase reporter activity in HEK293T cells co-expressing Axin-L106R with increasing doses of Axin-wt protein. Axin-L106R induces basal β-catenin mediated transcription. Co-expression of increasing amounts of Axin-wt protein levels suppresses Axin-L106R induced β-catenin mediated transcription. Graph shows average (bars) and range (diamonds) of luc activity in duplicate cell cultures.

Supplementary Figure 4 Analysis of Axin L106R F119R’s structural stability and dose-response effects on tumor-suppressor activity in cells.

(a) Fluorescence-based thermal denaturation of RGS-L106R F119R at 340 nm emission. Tu, unfolding temperature. (b) Introduction of the F119R mutation into Axin-wt does not affect tumour suppressor activity. Graph shows average (bars) and range (diamonds) of luc activity in duplicate cell cultures. (c) Introduction of the F119R mutation into Axin-L106R rescues tumour suppressor activity. Wnt luciferase reporter activity was measured in HEK293T cells expressing increasing concentrations (6.25, 12.5, 25, 50, 100 and 200 ng of transfected DNA, respectively) of the indicated Axin variants. Graph shows average (bars) and range (diamonds) of luc activity in duplicate cell cultures. (d) Western blot indicating protein levels of Axin-wt (WT), Axin-L106R, rescue variants Axin-L106R F119R (F>R), Axin-L106R W118R F119R (WF>RR), and Axin-ΔRGS upon transfection of HEK293T cells with 100 ng of plasmid DNA. Ctr, control.

Supplementary Figure 5 STRING v10 analysis of lost and gained binders of Axin-variant interactomes.

(a) Interactome (grey dots) of Axin-wt (red dot) is shown. Shown interactors displayed a confidence score of >0.900 and interaction with at least one other partner in the network. Binding partners are clustered (grey circles) based on previously described protein-protein interactions and on shared activity in the regulation of cellular processes as revealed by GOTERM analysis. Loss of binding (blue dots) is indicated for (b) Axin-ΔRGS, (c) Axin-L106R and (d) Axin-L106R F119R. (e) Gained interactions (green dots) of Axin-L106R are shown, based on comparison with the Axin-wt interactome in (a). Partners that were trapped by Axin-L106R and set free by Axin-L106R F119R are indicated (yellow dots).

Supplementary Figure 6 Ectopic wingless signalling induced by Axin cancer-mutant expression in Drosophila wing discs is rescued by aggregon mutation.

Related to Figure 6. (a) Schematic presentation of gene targeting strategy to generate Axin knock-in flies. (b) Confirmation of genetic modification of Axin knock-out mutant flies. (c) Quantification of the average clone size of the P compartment occupied by GFP-positive tissue in different genotype (n = 5). Error bars represent SDs. *** p<0.0001. (d) Excess Wingless (Wnt) signalling in DAxin-V72R clones is rescued by aggregon mutation. Larvae were grown at 29°C. In GFP-negative cells, the indicated DAxin variants were knocked in the endogenous axn locus. Expression of the Wg target genes senseless (Sens) and distalless (Dll) was analysed using immunostaining. DAxin-V72R-expressing clones overgrow the posterior tissue (arrowheads) and show ectopic expression of both Sens and Dll. Wg target gene expression by the V72R cancer mutant is rescued by introduction of the secondary aggregon mutation I169R L170R. Boundary between anterior and posterior tissue in wing discs is marked by a dotted line. Scale bar 100 µm (e) Mutation of the region homologous to the human RGS aggregon does not rescue DAxin-V72R suppressor activity. In shown mosaic posterior compartments, the V72R Y86R F87R DAxin variant (homologue of human Axin suppressor mutant L106R F119R W118R) was knocked in the endogenous axn locus in GFP-negative cells. Mutant clones overgrow the posterior tissue at all temperatures tested.

Supplementary Figure 7 Aggregon mutation restores the ability of Drosophila Axin V72R to assemble in punctate cytosolic complexes.

(a) Confocal microscopy analysis of human Axin (hAxin)-wt, hAxin-L106R and hAxin-L106R F119R in HEK293T cells. Scale bars 10 μm. (b) Confocal microscopy analysis of Drosophila Axin (DAxin)-wt, DAxin-V72R, DAxin-V72R F87R (homologue of human aggregon suppressor), DAxin-V72R I169R L170R (Drosophila aggregon suppressor) in HEK293T cells. Note that the Drosophila aggregation suppressor mutation (I169R L170R) reverts the protein into puncta while the potential suppressor mutation at the position of the human aggregon (F87R) fails to do so. Scale bars 10 μm. Schematic RGS domain structures indicate the location of cancer (red) and aggregon (green) mutations.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–7 (PDF 1218 kb)

Supplementary Table 1

Analysis of interactomes of Axin variants (XLSX 318 kb)

Supplementary Data Set 1

Uncropped blots (PDF 6393 kb)

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Anvarian, Z., Nojima, H., van Kappel, E. et al. Axin cancer mutants form nanoaggregates to rewire the Wnt signaling network. Nat Struct Mol Biol 23, 324–332 (2016). https://doi.org/10.1038/nsmb.3191

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nsmb.3191

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer