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:

Cryo-EM structure of the Hippo signaling integrator human STRIPAK

Abstract

The striatin-interacting phosphatase and kinase (STRIPAK) complex is a large, multisubunit protein phosphatase 2A (PP2A) assembly that integrates diverse cellular signals in the Hippo pathway to regulate cell proliferation and survival. The architecture and assembly mechanism of this critical complex are poorly understood. Using cryo-EM, we determine the structure of the human STRIPAK core comprising PP2AA, PP2AC, STRN3, STRIP1, and MOB4 at 3.2-Å resolution. Unlike the canonical trimeric PP2A holoenzyme, STRIPAK contains four copies of STRN3 and one copy of each the PP2AA–C heterodimer, STRIP1, and MOB4. The STRN3 coiled-coil domains form an elongated homotetrameric scaffold that links the complex together. An inositol hexakisphosphate (IP6) is identified as a structural cofactor of STRIP1. Mutations of key residues at subunit interfaces disrupt the integrity of STRIPAK, causing aberrant Hippo pathway activation. Thus, STRIPAK is established as a noncanonical PP2A complex with four copies of regulatory STRN3 for enhanced signal integration.

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

Access options

Buy this article

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

Fig. 1: Cryo-EM structure of human STRIPAK core.
Fig. 2: Interactions between PP2AA and the STRN3 coiled-coil tetramer.
Fig. 3: IP6 is a structural cofactor of STRIP1.
Fig. 4: STRIP1 scaffolds the STRIPAK core.
Fig. 5: MOB4 tethers the WD40 domain of STRN3 to STRIP1.

Similar content being viewed by others

Data Availability

Cryo-EM density maps and atomic coordinates for human STRIPAK have been deposited in the Electron Microscopy Data Bank and wwPDB, respectively, under accession codes EMD-22650 and PDB 7K36. Source data are provided with this paper.

References

  1. Shi, Y. Serine/threonine phosphatases: mechanism through structure. Cell 139, 468–484 (2009).

    Article  CAS  PubMed  Google Scholar 

  2. Glatter, T., Wepf, A., Aebersold, R. & Gstaiger, M. An integrated workflow for charting the human interaction proteome: insights into the PP2A system. Mol. Syst. Biol. 5, 237 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  3. Janssens, V., Goris, J. & Van Hoof, C. PP2A: the expected tumor suppressor. Curr. Opin. Genet. Dev. 15, 34–41 (2005).

    Article  CAS  PubMed  Google Scholar 

  4. Janssens, V. & Goris, J. Protein phosphatase 2A: a highly regulated family of serine/threonine phosphatases implicated in cell growth and signalling. Biochem. J. 353, 417–439 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Virshup, D. M. & Shenolikar, S. From promiscuity to precision: protein phosphatases get a makeover. Mol. Cell 33, 537–545 (2009).

    Article  CAS  PubMed  Google Scholar 

  6. Kamibayashi, C. et al. Comparison of heterotrimeric protein phosphatase 2A containing different B subunits. J. Biol. Chem. 269, 20139–20148 (1994).

    Article  CAS  PubMed  Google Scholar 

  7. Cho, U. S. & Xu, W. Crystal structure of a protein phosphatase 2A heterotrimeric holoenzyme. Nature 445, 53–57 (2007).

    Article  CAS  PubMed  Google Scholar 

  8. Lambrecht, C., Haesen, D., Sents, W., Ivanova, E. & Janssens, V. Structure, regulation, and pharmacological modulation of PP2A phosphatases. Methods Mol. Biol. 1053, 283–305 (2013).

    Article  CAS  PubMed  Google Scholar 

  9. Wlodarchak, N. et al. Structure of the Ca2+-dependent PP2A heterotrimer and insights into Cdc6 dephosphorylation. Cell Res. 23, 931–946 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Xu, Y., Chen, Y., Zhang, P., Jeffrey, P. D. & Shi, Y. Structure of a protein phosphatase 2A holoenzyme: insights into B55-mediated Tau dephosphorylation. Mol. Cell 31, 873–885 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Shi, Z., Jiao, S. & Zhou, Z. STRIPAK complexes in cell signaling and cancer. Oncogene https://doi.org/10.1038/onc.2016.9 (2016).

  12. Hwang, J. & Pallas, D. C. STRIPAK complexes: structure, biological function, and involvement in human diseases. Int. J. Biochem. Cell Biol. 47, 118–148 (2014).

    Article  CAS  PubMed  Google Scholar 

  13. Goudreault, M. et al. A PP2A phosphatase high density interaction network identifies a novel striatin-interacting phosphatase and kinase complex linked to the cerebral cavernous malformation 3 (CCM3) protein. Mol. Cell. Proteomics 8, 157–171 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Kuck, U., Radchenko, D. & Teichert, I. STRIPAK, a highly conserved signaling complex, controls multiple eukaryotic cellular and developmental processes and is linked with human diseases. Biol. Chem. https://doi.org/10.1515/hsz-2019-0173 (2019).

  15. Tang, Y. et al. Architecture, substructures, and dynamic assembly of STRIPAK complexes in Hippo signaling. Cell Discov. 5, 3 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Herzog, F. et al. Structural probing of a protein phosphatase 2A network by chemical cross-linking and mass spectrometry. Science 337, 1348–1352 (2012).

    Article  CAS  PubMed  Google Scholar 

  17. Moreno, C. S. et al. WD40 repeat proteins striatin and S/G2 nuclear autoantigen are members of a novel family of calmodulin-binding proteins that associate with protein phosphatase 2A. J. Biol. Chem. 275, 5257–5263 (2000).

    Article  CAS  PubMed  Google Scholar 

  18. Kean, M. J. et al. Structure-function analysis of core STRIPAK proteins: a signaling complex implicated in Golgi polarization. J. Biol. Chem. 286, 25065–25075 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Zheng, Y. & Pan, D. The Hippo signaling pathway in development and disease. Dev. Cell 50, 264–282 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Fu, V., Plouffe, S. W. & Guan, K. L. The Hippo pathway in organ development, homeostasis, and regeneration. Curr. Opin. Cell Biol. 49, 99–107 (2017).

    Article  CAS  PubMed  Google Scholar 

  21. Misra, J. R. & Irvine, K. D. The Hippo signaling network and its biological functions. Annu. Rev. Genet. https://doi.org/10.1146/annurev-genet-120417-031621 (2018).

  22. Yu, F. X., Zhao, B. & Guan, K. L. Hippo pathway in organ size control, tissue homeostasis, and cancer. Cell 163, 811–828 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Davis, J. R. & Tapon, N. Hippo signalling during development. Development https://doi.org/10.1242/dev.167106 (2019).

  24. Ribeiro, P. S. et al. Combined functional genomic and proteomic approaches identify a PP2A complex as a negative regulator of Hippo signaling. Mol. Cell 39, 521–534 (2010).

    Article  CAS  PubMed  Google Scholar 

  25. Couzens, A. L. et al. Protein interaction network of the mammalian Hippo pathway reveals mechanisms of kinase-phosphatase interactions. Sci. Signal 6, rs15 (2013).

    Article  PubMed  CAS  Google Scholar 

  26. Bae, S. J. et al. SAV1 promotes Hippo kinase activation through antagonizing the PP2A phosphatase STRIPAK. Elife https://doi.org/10.7554/eLife.30278 (2017).

  27. Zheng, Y. et al. Homeostatic control of Hpo/MST kinase activity through autophosphorylation-dependent recruitment of the STRIPAK PP2A phosphatase complex. Cell Rep. 21, 3612–3623 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Bae, S. J., Ni, L. & Luo, X. STK25 suppresses Hippo signaling by regulating SAV1-STRIPAK antagonism. Elife https://doi.org/10.7554/eLife.54863 (2020).

  29. Bae, S. J. & Luo, X. Activation mechanisms of the Hippo kinase signaling cascade. Biosci. Rep. https://doi.org/10.1042/BSR20171469 (2018).

  30. Meng, Z., Moroishi, T. & Guan, K. L. Mechanisms of Hippo pathway regulation. Genes Dev. 30, 1–17 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Ni, L. et al. Structural basis for autoactivation of human Mst2 kinase and its regulation by RASSF5. Structure 21, 1757–1768 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Ni, L., Zheng, Y., Hara, M., Pan, D. & Luo, X. Structural basis for Mob1-dependent activation of the core Mst–Lats kinase cascade in Hippo signaling. Genes Dev. 29, 1416–1431 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Liu, B. et al. Toll receptor-mediated Hippo signaling controls innate immunity in Drosophila. Cell 164, 406–419 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Chen, R., Xie, R., Meng, Z., Ma, S. & Guan, K. L. STRIPAK integrates upstream signals to initiate the Hippo kinase cascade. Nat. Cell Biol. 21, 1565–1577 (2019).

    Article  CAS  PubMed  Google Scholar 

  35. Gil-Ranedo, J. et al. STRIPAK members orchestrate Hippo and insulin receptor signaling to promote neural stem cell reactivation. Cell Rep. 27, 2921–2933.e5 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Tang, Y. et al. Selective inhibition of STRN3-containing PP2A phosphatase restores Hippo tumor-suppressor activity in gastric cancer. Cancer Cell 38, 115–128.e9 (2020).

    Article  CAS  PubMed  Google Scholar 

  37. Kim, J. W. et al. STRIPAK directs PP2A activity toward MAP4K4 to promote oncogenic transformation of human cells. Elife https://doi.org/10.7554/eLife.53003 (2020).

  38. Tanti, G. K., Pandey, S. & Goswami, S. K. SG2NA enhances cancer cell survival by stabilizing DJ-1 and thus activating Akt. Biochem. Biophys. Res. Commun. 463, 524–531 (2015).

    Article  CAS  PubMed  Google Scholar 

  39. Wong, M. et al. Silencing of STRN4 suppresses the malignant characteristics of cancer cells. Cancer Sci. 105, 1526–1532 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Weissmann, F. et al. biGBac enables rapid gene assembly for the expression of large multisubunit protein complexes. Proc. Natl Acad. Sci. USA 113, E2564–E2569 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Chen, C. et al. Striatins contain a noncanonical coiled coil that binds protein phosphatase 2A A subunit to form a 2:2 heterotetrameric core of striatin-interacting phosphatase and kinase (STRIPAK) complex. J. Biol. Chem. 289, 9651–9661 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Gordon, J. et al. Protein phosphatase 2a (PP2A) binds within the oligomerization domain of striatin and regulates the phosphorylation and activation of the mammalian Ste20-like kinase Mst3. BMC Biochem. 12, 54 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Moreno, C. S., Lane, W. S. & Pallas, D. C. A mammalian homolog of yeast MOB1 is both a member and a putative substrate of striatin family-protein phosphatase 2A complexes. J. Biol. Chem. 276, 24253–24260 (2001).

    Article  CAS  PubMed  Google Scholar 

  44. Castets, F. et al. Zinedin, SG2NA, and striatin are calmodulin-binding, WD repeat proteins principally expressed in the brain. J. Biol. Chem. 275, 19970–19977 (2000).

    Article  CAS  PubMed  Google Scholar 

  45. Groves, M. R. & Barford, D. Topological characteristics of helical repeat proteins. Curr. Opin. Struct. Biol. 9, 383–389 (1999).

    Article  CAS  PubMed  Google Scholar 

  46. Holm, L. DALI and the persistence of protein shape. Protein Sci. 29, 128–140 (2020).

    Article  CAS  PubMed  Google Scholar 

  47. Madsen, C. D. et al. STRIPAK components determine mode of cancer cell migration and metastasis. Nat. Cell Biol. 17, 68–80 (2015).

    Article  CAS  PubMed  Google Scholar 

  48. Duhart, J. C. & Raftery, L. A. Mob family proteins: regulatory partners in Hippo and Hippo-like intracellular signaling pathways. Front. Cell Dev. Biol. 8, 161 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  49. Kim, S. Y., Tachioka, Y., Mori, T. & Hakoshima, T. Structural basis for autoinhibition and its relief of MOB1 in the Hippo pathway. Sci. Rep. 6, 28488 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  50. Chen, M. et al. The MST4–MOB4 complex disrupts the MST1–MOB1 complex in the Hippo–YAP pathway and plays a pro-oncogenic role in pancreatic cancer. J. Biol. Chem. 293, 14455–14469 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Breitman, M., Zilberberg, A., Caspi, M. & Rosin-Arbesfeld, R. The armadillo repeat domain of the APC tumor suppressor protein interacts with striatin family members. Biochim. Biophys. Acta 1783, 1792–1802 (2008).

    Article  CAS  PubMed  Google Scholar 

  52. Buda, A. & Pignatelli, M. E-cadherin and the cytoskeletal network in colorectal cancer development and metastasis. Cell Commun. Adhes. 18, 133–143 (2011).

    Article  CAS  PubMed  Google Scholar 

  53. Mastronarde, D. N. Automated electron microscope tomography using robust prediction of specimen movements. J. Struct. Biol. 152, 36–51 (2005).

    Article  PubMed  Google Scholar 

  54. Zheng, S. Q. et al. MotionCor2: anisotropic correction of beam-induced motion for improved cryo-electron microscopy. Nat. Methods 14, 331–332 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Zhang, K. Gctf: real-time CTF determination and correction. J. Struct. Biol. 193, 1–12 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Zivanov, J. et al. New tools for automated high-resolution cryo-EM structure determination in RELION-3. Elife https://doi.org/10.7554/eLife.42166 (2018).

  57. Scheres, S. H. RELION: implementation of a Bayesian approach to cryo-EM structure determination. J. Struct. Biol. 180, 519–530 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Tang, G. et al. EMAN2: an extensible image processing suite for electron microscopy. J. Struct. Biol. 157, 38–46 (2007).

    Article  CAS  PubMed  Google Scholar 

  59. Scheres, S. H. & Chen, S. Prevention of overfitting in cryo-EM structure determination. Nat. Methods 9, 853–854 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Xu, Y. et al. Structure of the protein phosphatase 2A holoenzyme. Cell 127, 1239–1251 (2006).

    Article  CAS  PubMed  Google Scholar 

  61. Pettersen, E. F. et al. UCSF Chimera—a visualization system for exploratory research and analysis. J. Comput. Chem. 25, 1605–1612 (2004).

    Article  CAS  PubMed  Google Scholar 

  62. Emsley, P., Lohkamp, B., Scott, W. G. & Cowtan, K. Features and development of Coot. Acta Crystallogr. D Biol. Crystallogr. 66, 486–501 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Adams, P. D. et al. PHENIX: a comprehensive Python-based system for macromolecular structure solution. Acta Crystallogr. D Biol. Crystallogr. 66, 213–221 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Chen, V. B. et al. MolProbity: all-atom structure validation for macromolecular crystallography. Acta Crystallogr. D Biol. Crystallogr. 66, 12–21 (2010).

    Article  CAS  PubMed  Google Scholar 

  65. Gouet, P., Courcelle, E., Stuart, D. I. & Metoz, F. ESPript: analysis of multiple sequence alignments in PostScript. Bioinformatics 15, 305–308 (1999).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank H. Yu for helpful discussion. We thank S. Liu for assistance with mutant analysis. Single-particle cryo-EM data were collected at the University of Texas Southwestern Medical Center (UTSW) Cryo-Electron Microscopy Facility, which is funded by a Cancer Prevention and Research Institute of Texas (CPRIT) Core Facility Support Award (Grant no. RP170644). We thank D. Nicastro for facility access and data acquisition. This study is supported in part by grants from the National Institutes of Health (CA220283 to X.Z.; GM132275 to X.L.), and grants from the Welch Foundation (I-1702 to X.Z.; I-1944 to X.-c.B.; I-1932 to X.L.). X.Z. and X.-c.B. are Virginia Murchison Linthicum Scholars in Medical Research at UTSW.

Author information

Authors and Affiliations

Authors

Contributions

B.-C.J. performed protein purification, EM grid screening and preparation, structure docking and refinement, and functional studies in vitro. S.J.B. made the KO cell lines and performed functional studies in human cells. B.-C.J. and S.J.B. contributed to the initial draft of the manuscript. L.N. did the MS analysis. X.Z. assisted with structure determination and refinement. X.-c.B. prepared cryo-EM grids, collected cryo-EM data, and determined the cryo-EM structure. X.-c.B. and X.L. co-supervised the research and analyzed data. X.L. wrote the manuscript with help from all authors.

Corresponding authors

Correspondence to Xiao-chen Bai or Xuelian Luo.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Nature Structural & Molecular Biology thanks Lanfen Chen and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Peer reviewer reports are available. Florian Ullrich and Inês Chen were the primary editors on this article and managed its editorial process and peer review in collaboration with the rest of the editorial team.

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

Extended data

Extended Data Fig. 1 Purification of human STRIPAK complexes and single-particle cryo-EM analysis of the STRIPAK core.

a, Coomassie blue staining of fractions from size exclusion chromatography (SEC) purification of the STRIPAK core (left) and STRIPAK (right) complexes. Highlighted fraction (red) was used for cryo-EM analysis. b, Cryo-EM data processing for initial model generation. c, Cryo-EM density map and atomic model of the STRIPAK core at a resolution of ~3.9-Å. d, Final reconstruction with C1 symmetry colored based on local resolution.

Extended Data Fig. 2 Cryo-EM analyses of the STRIPAK complex.

a, Representative micrograph and 2D classes. b, Final reconstructions with C1 or C2 symmetry colored based on local resolution. c, Gold-standard Fourier shell correlation (FSC) curves of the final 3D reconstruction (C1 in orange and C2 in blue). d, Euler angle distribution of the particles used in the final C1 3D reconstruction.

Extended Data Fig. 3

Flow chart of cryo-EM image processing for the STRIPAK complex.

Extended Data Fig. 4 Cryo-EM map of the STRIPAK complex.

a, Overall cryo-EM map of human STRIPAK at a resolution of 3.5-Å in front and back views. b, Representative cryo-EM densities of the STRIPAK complex. The densities from the cryo-EM maps and the corresponding protein fragments are shown in surface and sticks, respectively. Residues numbers of each sample fragment are labeled. Representative residues from STRN3 WD40 or at the PP2AC–STRIP1 interface are labeled.

Extended Data Fig. 5 Mutational studies of STRN3 at the STRN3-PP2AA interface.

a, Effects of mutations in STRN3 on STRIPAK formation. Mock vector or FLAG-STRN3 mutants were transfected into control or STRN1/3/4-KO 293A cells as indicated. Lysates and FLAG IPs were subjected to immunoblotting. b, Effects of mutations in STRN3 on the ratios of pMOB1/MOB1, pLATS/LATS, and pYAP/YAP in control or STRN1/3/4-KO 293A cells transfected with mock vector or indicated plasmids. c, Immunofluorescence staining of YAP localization in 293A control or STRN1/3/4-KO cells transfected with mock vector or indicated plasmids. Scale bar, 10 µm. d, Relative expression of YAP target genes CYR61 in 293A control or STRN1/3/4-KO cells transfected with mock vector or indicated plasmids. Data in b,d are plotted as mean ± SEM of three independent experiments. Results were evaluated by Two-tailed unpaired t tests (*, P<0.05; **, P<0.01; ***, P<0.001; ****, P<0.0001; ns, non-significant). Source data for graphs are available online.

Source data

Extended Data Fig. 6 Functional validation of PP2AC-interacting residues of STRIP1.

a, Effects of mutations in STRIP1 on the ratios of pMOB1/MOB1, pLATS/LATS, and pYAP/YAP in control or STRIP1-KO 293A cells transfected with mock vector or indicated plasmids. b, Immunofluorescence staining of YAP localization in 293A control or STRIP1-KO cells transfected with mock vector or indicated plasmids. Scale bar, 10 µm. Data in a are plotted as mean ± SEM of three independent experiments. Results were evaluated by Two-tailed unpaired t tests (*, P<0.05; **, P<0.01; ***, P<0.001; ****, P<0.0001; ns, non-significant). Source data for graphs are available online.

Source data

Extended Data Fig. 7 Functional validation of the C-terminal tail (CTT) of STRIP1.

a, Effects of mutation in STRIP1 on STRIPAK formation. Control or STRIP1-KO 293A cells were transfected with mock vector, FLAG-STRIP1 WT or ∆CTT mutant plasmid as indicated. Lysates and FLAG IPs were subjected to immunoblotting. b, Immunoblot of lysates of control or STRIP1-KO 293A cells transfected with mock vector, STRIP1 WT or ∆CTT mutant plasmid as indicated. HM indicates hydrophobic motif in LATS1/2. c, Quantification of the ratios of phospho- and total proteins in b. d, Immunofluorescence staining of YAP localization in 293A control or STRIP1-KO cells transfected with mock vector, STRIP1 WT or ∆CTT mutant plasmids. Scale bar, 10 µm. Data in c are plotted as mean ± SEM of three independent experiments. Results were evaluated by Two-tailed unpaired t tests (*, P<0.05; **, P<0.01; ***, P<0.001; ****, P<0.0001; ns, non-significant). Source data for graphs are available online.

Source data

Extended Data Fig. 8 MOB4 interacts with STRIP1 and the WD40 domain of STRN3.

a, Cartoon representation of MOB4. The N-terminal extension (NTE) and core of MOB4 are colored in slate and yellow, respectively. b, Sequence alignment of human MOB NTEs. The N-terminal α helices (Nαs) and residue numbers of MOB1 and MOB4 are shown above and below the sequences, respectively. c, Superposition of MOB4 with the crystal structure of unphosphorylated MOB1 (colored in grey; PDB 5B5V). The NTE and core of MOB1 are colored in teal and grey, respectively. d, Superposition of MOB4–STRN3 WD40 (this study) and pMOB1–LATS1 (colored in grey and salmon, respectively; PDB 5BRK). The NTE of pMOB1 is colored in red. e, Interface between MOB4 and STRIP1 NTD (colored in yellow and wheat, respectively). Potential hydrogen bonds are indicated with red dashed lines. f, Interface between MOB4 core and STRN3 WD40 (colored in yellow and blue, respectively). g, Interface between MOB4 Nα1 and STRN3 WD40.

Extended Data Fig. 9 Interactions between MOB4 and STRN3 WD40.

a,b, Overall structure of the STRN3 WD40 domain with cartoon (a) and surface representation (b) in the same view. Residues at the MOB4–STRIP1 interface are colored salmon. The seven blades of the WD40 domain are numbered from 1 to 7, and the strands of each blade are numbered A-D from the innermost strand to the outermost strand. The N- and C-termini are indicated. c, Immunoblot of lysates of control or MOB4-KO 293A cells transfected with mock vector, MOB4 WT or ∆N40 mutant plasmids. d, Quantification of the ratios of phospho- and total proteins in c. Data in d are plotted as mean ± SEM of three independent experiments. Results were evaluated by Two-tailed unpaired t tests (*, P<0.05; **, P<0.01; ***, P<0.001; ****, P<0.0001; ns, non-significant). Source data for graphs are available online.

Source data

Extended Data Fig. 10 Interactions between STRN3 WD40 and MOB4.

a, Immunoblot of lysates of control or STRN1/3/4-KO 293A cells transfected with mock vector or indicated plasmids. b, Quantification of the ratios of phospho- and total proteins in a. c, Quantification of YAP localization in control or STRN1/3/4-KO 293A cells transfected with mock vector or indicated plasmids, derived from immunofluorescence staining analysis of YAP. Approximately 100 cells were counted for quantification. d, Relative expression of YAP target genes CTGF and CYR61 in control or STRN1/3/4-KO 293A cells transfected with mock vector or indicated plasmids. Data in b,d are plotted as mean ± SEM of three independent experiments. Results were evaluated by Two-tailed unpaired t tests (*, P<0.05; **, P<0.01; ***, P<0.001; ****, P<0.0001; ns, non-significant). Source data for graphs are available online.

Source data

Supplementary information

Supplementary Information

Supplementary Note 1 and Supplementary Figs. 1–4.

Reporting Summary

Peer Review Information

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 1

Unprocessed western blots.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 3

Unprocessed western blots.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 5

Unprocessed western blots.

Source Data Extended Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 7

Statistical source data.

Source Data Extended Data Fig. 7

Unprocessed western blots.

Source Data Extended Data Fig. 9

Statistical source data.

Source Data Extended Data Fig. 9

Unprocessed western blots.

Source Data Extended Data Fig. 10

Statistical source data.

Source Data Extended Data Fig. 10

Unprocessed western blots.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Jeong, BC., Bae, S.J., Ni, L. et al. Cryo-EM structure of the Hippo signaling integrator human STRIPAK. Nat Struct Mol Biol 28, 290–299 (2021). https://doi.org/10.1038/s41594-021-00564-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41594-021-00564-y

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