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.

  • Letter
  • Published:

The chaperone DNAJB6 surveils FG-nucleoporins and is required for interphase nuclear pore complex biogenesis

Abstract

Biogenesis of nuclear pore complexes (NPCs) includes the formation of the permeability barrier composed of phenylalanine-glycine-rich nucleoporins (FG-Nups) that regulate the selective passage of biomolecules across the nuclear envelope. The FG-Nups are intrinsically disordered and prone to liquid–liquid phase separation and aggregation when isolated. How FG-Nups are protected from making inappropriate interactions during NPC biogenesis is not fully understood. Here we find that DNAJB6, a molecular chaperone of the heat shock protein network, forms foci in close proximity to NPCs. The number of these foci decreases upon removal of proteins involved in the early steps of interphase NPC biogenesis. Conversely, when this process is stalled in the last steps, the number of DNAJB6-containing foci increases and these foci are identified as herniations at the nuclear envelope. Immunoelectron tomography shows that DNAJB6 localizes inside the lumen of the herniations arising at NPC biogenesis intermediates. Loss of DNAJB6 results in the accumulation of cytosolic annulate lamellae, which are structures containing partly assembled NPCs, a feature associated with disturbances in NPC biogenesis. We find that DNAJB6 binds to FG-Nups and can prevent the aggregation of the FG region of several FG-Nups in cells and in vitro. Together, our data show that the molecular chaperone DNAJB6 provides quality control during NPC biogenesis and is involved in the surveillance of native intrinsically disordered FG-Nups.

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: DNAJB6 localizes in foci to the NE, and disruption of DNAJB6 expression leads to accumulation of annulate lamellae.
Fig. 2: DNAJB6 foci formation is cell cycle dependent and is related to interphase NPC assembly.
Fig. 3: DNAJB6 localizes to herniations at the NE.
Fig. 4: DNAJB6 interacts with FG-Nup components of the NPC.
Fig. 5: DNAJB6b modulates the LLPS and aggregation of FG-Nup fragments in vitro.
Fig. 6: The S/T domain of DNAJB6 mediates the interaction with FG-Nups.

Similar content being viewed by others

Data availability

Large-scale EM data are available via nanotomy.org (http://www.nanotomy.org/OA/Kuiper2022NCB/). All other data supporting the findings of this study are available from the corresponding author on reasonable request. Source data are provided with this paper.

References

  1. Fernandez-Martinez, J. & Rout, M. P. One ring to rule them all? Structural and functional diversity in the nuclear pore complex. Trends Biochem. Sci. 46, 595–607 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Hampoelz, B., Andres-Pons, A., Kastritis, P. & Beck, M. Structure and assembly of the nuclear pore complex. Annu. Rev. Biophys. 48, 515–536 (2019).

    Article  CAS  PubMed  Google Scholar 

  3. Otsuka, S. & Ellenberg, J. Mechanisms of nuclear pore complex assembly—two different ways of building one molecular machine. FEBS Lett. 592, 475–488 (2018).

    Article  CAS  PubMed  Google Scholar 

  4. Dultz, E., Wojtynek, M., Medalia, O. & Onischenko, E. The nuclear pore complex: birth, life, and death of a cellular behemoth. Cells 11, 1456 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Frey, S., Richter, R. P. & Görlich, D. FG-rich repeats of nuclear pore proteins form a three-dimensional meshwork with hydrogel-like properties. Science 314, 815–817 (2006).

    Article  CAS  PubMed  Google Scholar 

  6. Schmidt, H. B. & Görlich, D. Transport selectivity of nuclear pores, phase separation, and membraneless organelles. Trends Biochem. Sci. 41, 46–61 (2016).

    Article  CAS  PubMed  Google Scholar 

  7. Milles, S. et al. Facilitated aggregation of FG nucleoporins under molecular crowding conditions. EMBO Rep. 14, 178–183 (2013).

    Article  CAS  PubMed  Google Scholar 

  8. D’Angelo, M. A., Raices, M., Panowski, S. H. & Hetzer, M. W. Age-dependent deterioration of nuclear pore complexes causes a loss of nuclear integrity in postmitotic cells. Cell 136, 284–295 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  9. Gasset-Rosa, F. et al. Polyglutamine-expanded huntingtin exacerbates age-related disruption of nuclear integrity and nucleocytoplasmic transport. Neuron 94, 48–57.e4 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Rempel, I. L. et al. Age-dependent deterioration of nuclear pore assembly in mitotic cells decreases transport dynamics. eLife 8, 1–26 (2019).

    Article  Google Scholar 

  11. Larrieu, D. et al. Inhibition of the acetyltransferase NAT10 normalizes progeric and aging cells by rebalancing the Transportin-1 nuclear import pathway. Sci. Signal 11, 1–16 (2018).

    Article  Google Scholar 

  12. Lautier, O. et al. Co-translational assembly and localized translation of nucleoporins in nuclear pore complex biogenesis. Mol. Cell 81, 2417–2427.e5 (2021).

    Article  CAS  PubMed  Google Scholar 

  13. Seidel, M. et al. Co-translational assembly orchestrates competing biogenesis pathways. Nat. Commun. 13, 1224 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Kampinga, H. H. & Craig, E. A. The HSP70 chaperone machinery: J proteins as drivers of functional specificity. Nat. Rev. Mol. Cell Biol. 11, 579–592 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Hageman, J. et al. A DNAJB chaperone subfamily with HDAC-dependent activities suppresses toxic protein aggregation. Mol. Cell 37, 355–369 (2010).

    Article  CAS  PubMed  Google Scholar 

  16. Kakkar, V. et al. The S/T-rich motif in the DNAJB6 chaperone delays polyglutamine aggregation and the onset of disease in a mouse model. Mol. Cell 62, 272–283 (2016).

    Article  CAS  PubMed  Google Scholar 

  17. Thiruvalluvan, A. et al. DNAJB6, a key factor in neuronal sensitivity to amyloidogenesis. Mol. Cell 78, 346–358.e9 (2020).

    Article  CAS  PubMed  Google Scholar 

  18. Månsson, C. et al. DNAJB6 is a peptide-binding chaperone which can suppress amyloid fibrillation of polyglutamine peptides at substoichiometric molar ratios. Cell Stress Chaperones 19, 227–239 (2014).

    Article  PubMed  Google Scholar 

  19. Aprile, F. A. et al. The molecular chaperones DNAJB6 and Hsp70 cooperate to suppress α-synuclein aggregation. Sci. Rep. 7, 1–11 (2017).

    Article  CAS  Google Scholar 

  20. Reidy, M., Sharma, R., Roberts, B. L. & Masison, D. C. Human J-protein DnaJB6b cures a subset of Saccharomyces cerevisiae prions and selectively blocks assembly of structurally related amyloids. J. Biol. Chem. 291, 4035–4047 (2016).

    Article  CAS  PubMed  Google Scholar 

  21. Stein, K. C., Bengoechea, R., Harms, M. B., Weihl, C. C. & True, H. L. Myopathy-causing mutations in an HSP40 chaperone disrupt processing of specific client conformers. J. Biol. Chem. 289, 21120–21130 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Kessel, R. G. Annulate lamellae: a last frontier in cellular organelles. Int. Rev. Cytol. 133, 43–120 (1992).

    Article  CAS  PubMed  Google Scholar 

  23. Kessel, R. G. The structure and function of annulate lamellae: porous cytoplasmic and intranuclear membranes. Int. Rev. Cytol. 82, 181–303 (1983).

    Article  CAS  PubMed  Google Scholar 

  24. Walther, T. C. et al. The conserved Nup107-160 complex is critical for nuclear pore complex assembly. Cell 113, 195–206 (2003).

    Article  CAS  PubMed  Google Scholar 

  25. Hampoelz, B. et al. Pre-assembled nuclear pores insert into the nuclear envelope during early development. Cell 166, 664–678 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Cordes, V. C., Reidenbach, S. & Franke, W. W. Cytoplasmic annulate lamellae in cultured cells: composition, distribution, and mitotic behavior. Cell Tissue Res 284, 177–191 (1996).

    Article  CAS  PubMed  Google Scholar 

  27. Makio, T. et al. The nucleoporins Nup170p and Nup157p are essential for nuclear pore complex assembly. J. Cell Biol. 185, 459–473 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Wu, X. et al. Disruption of the FG nucleoporin Nup98 causes selective changes in nuclear pore complex stoichiometry and function. Proc. Natl Acad. Sci. USA 98, 3191–3196 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Walther, T. C. et al. RanGTP mediates nuclear pore complex assembly. Nature 424, 689–694 (2003).

    Article  CAS  PubMed  Google Scholar 

  30. Yannarell, A., Schumm, D. E. & Webb, T. E. Age-dependence of nuclear RNA processing. Mech. Ageing Dev. 6, 259–264 (1977).

    Article  CAS  PubMed  Google Scholar 

  31. Freibaum, B. D. et al. GGGGCC repeat expansion in C9orf72 compromises nucleocytoplasmic transport. Nature 525, 129–133 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Kampinga, H. H. et al. Function, evolution, and structure of J-domain proteins. Cell Stress Chaperones 24, 7–15 (2019).

    Article  PubMed  Google Scholar 

  33. Doucet, C. M., Talamas, J. A. & Hetzer, M. W. Cell cycle-dependent differences in nuclear pore complex assembly in metazoa. Cell 141, 1030–1041 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Vollmer, B. et al. Nup153 recruits the Nup107-160 complex to the inner nuclear membrane for interphasic nuclear pore complex assembly. Dev. Cell 33, 717–728 (2015).

    Article  CAS  PubMed  Google Scholar 

  35. Rampello, A. J. et al. Torsin ATPase deficiency leads to defects in nuclear pore biogenesis and sequestration of MLF2. J. Cell Biol. 219, e201910185 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Chase, A. R. et al. Dynamic functional assembly of the Torsin AAA+ ATPase and its modulation by LAP1. Mol. Biol. Cell 28, 2765–2772 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Zhao, C., Brown, R. S. H., Chase, A. R., Eisele, M. R. & Schlieker, C. Regulation of Torsin ATPases by LAP1 and LULL1. Proc. Natl Acad. Sci. USA 110, E1545–E1554 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Esra Demircioglu, F., Sosa, B. A., Ingram, J., Ploegh, H. L. & Schwartz, T. U. Structures of torsinA and its disease-mutant complexed with an activator reveal the molecular basis for primary dystonia. eLife 5, 1–14 (2016).

    Google Scholar 

  39. Ozelius, L. J. et al. The early-onset torsion dystonia gene (DYT1) encodes an ATP-binding protein. Nat. Genet. 17, 40–48 (1997).

    Article  CAS  PubMed  Google Scholar 

  40. Laudermilch, E. & Schlieker, C. Torsin ATPases: structural insights and functional perspectives. Curr. Opin. Cell Biol. 40, 1–7 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Laudermilch, E. et al. Dissecting Torsin/cofactor function at the nuclear envelope: a genetic study. Mol. Biol. Cell 27, 3964–3971 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Szöllösi, M. S. & Szöllösi, D. ‘Blebbing’ of the nuclear envelope of mouse zygotes, early embryos and hybrid cells. J. Cell Sci. 91, 257–267 (1988).

    Article  PubMed  Google Scholar 

  43. Speese, S. D. et al. Nuclear envelope budding enables large ribonucleoprotein particle export during synaptic Wnt signaling. Cell 149, 832–846 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Slot, J. W. & Geuze, H. J. Cryosectioning and immunolabeling. Nat. Protoc. 2, 2480–2491 (2007).

    Article  CAS  PubMed  Google Scholar 

  45. Allegretti, M. et al. In-cell architecture of the nuclear pore and snapshots of its turnover. Nature 586, 796–800 (2020).

    Article  CAS  PubMed  Google Scholar 

  46. Shulga, N. & Goldfarb, D. S. Binding dynamics of structural nucleoporins govern nuclear pore complex permeability and may mediate channel gating. Mol. Cell. Biol. 23, 534–542 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Shi, K. Y. et al. Toxic PRn poly-dipeptides encoded by the C9orf72 repeat expansion block nuclear import and export. Proc. Natl Acad. Sci. USA 114, E1111–E1117 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Otsuka, S. et al. Nuclear pore assembly proceeds by an inside-out extrusion of the nuclear envelope. eLife 5, 1–23 (2016).

    Article  Google Scholar 

  49. Terry, L. J. & Wente, S. R. Flexible gates: dynamic topologies and functions for FG nucleoporins in nucleocytoplasmic transport. Eukaryot. Cell 8, 1814–1827 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Yamada, J. et al. A bimodal distribution of two distinct categories of intrinsically disordered structures with separate functions in FG nucleoporins. Mol. Cell. Proteom. 9, 2205–2224 (2010).

    Article  Google Scholar 

  51. Huang, K., Tagliazucchi, M., Park, S. H., Rabin, Y. & Szleifer, I. Nanocompartmentalization of the nuclear pore lumen. Biophys. J. 118, 219–231 (2020).

    Article  CAS  PubMed  Google Scholar 

  52. Schmidt, H. B. & Görlich, D. Nup98 FG domains from diverse species spontaneously phase-separate into particles with nuclear pore-like permselectivity. eLife 4, 1–30 (2015).

    Article  Google Scholar 

  53. Celetti, G. et al. The liquid state of FG-nucleoporins mimics permeability barrier properties of nuclear pore complexes. J. Cell Biol. 219, e201907157 (2019).

    Article  PubMed Central  Google Scholar 

  54. Konishi, H. A. & Yoshimura, S. H. Interactions between non-structured domains of FG- and non-FG-nucleoporins coordinate the ordered assembly of the nuclear pore complex in mitosis. FASEB J. 34, 1532–1545 (2020).

    Article  CAS  PubMed  Google Scholar 

  55. Meinema, A. C. et al. Long unfolded linkers facilitate membrane protein import through the nuclear pore complex. Science 333, 90–93 (2011).

    Article  CAS  PubMed  Google Scholar 

  56. Rempel, I. L. et al. Flexible and extended linker domains support efficient targeting of heh2 to the inner nuclear membrane. Structure 28, 185–195.e5 (2020).

    Article  CAS  PubMed  Google Scholar 

  57. Månsson, C. et al. Conserved S/T residues of the human chaperone DNAJB6 are required for effective inhibition of Aβ42 amyloid fibril formation. Biochemistry 57, 4891–4892 (2018).

    Article  PubMed  Google Scholar 

  58. Månsson, C. et al. Interaction of the molecular chaperone DNAJB6 with growing amyloid-β 42 (Aβ42) aggregates leads to sub-stoichiometric inhibition of amyloid formation. J. Biol. Chem. 289, 31066–31076 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  59. Söderberg, C. A. G. et al. Structural modelling of the DNAJB6 oligomeric chaperone shows a peptide-binding cleft lined with conserved S/T-residues at the dimer interface. Sci. Rep. 8, 1–15 (2018).

    Article  Google Scholar 

  60. Karamanos, T. K., Tugarinov, V. & Clore, G. M. Unraveling the structure and dynamics of the human DNAJB6b chaperone by NMR reveals insights into Hsp40-mediated proteostasis. Proc. Natl Acad. Sci. USA 116, 21529–21538 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Ruggieri, A. et al. Complete loss of the DNAJB6 G/F domain and novel missense mutations cause distal-onset DNAJB6 myopathy. Acta Neuropathol. Commun. 3, 44 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  62. Cascarina, S. M., King, D. C., Osborne Nishimura, E. & Ross, E. D. LCD-Composer: an intuitive, composition-centric method enabling the identification and detailed functional mapping of low-complexity domains. NAR Genom. Bioinform 3, 1–19 (2021).

    CAS  Google Scholar 

  63. Prophet, S. M. et al. Atypical nuclear envelope condensates linked to neurological disorders reveal nucleoporin-directed chaperone activities. Nat. Cell Biol. https://doi.org/10.1038/s41556-022-01001-y (2022).

  64. Kim, C. E., Perez, A., Perkins, G., Ellisman, M. H. & Dauer, W. T. A molecular mechanism underlying the neural-specific defect in torsinA mutant mice. Proc. Natl Acad. Sci. USA 107, 9861–9866 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Tanabe, L. M., Liang, C. C. & Dauer, W. T. Neuronal nuclear membrane budding occurs during a developmental window modulated by torsin paralogs. Cell Rep. 16, 3322–3333 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Ruggieri, A. et al. DNAJB6 myopathies: focused review on an emerging and expanding group of myopathies. Front Mol. Biosci. 3, 63 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  67. Sheffield, L. G., Miskiewicz, H. B., Tannenbaum, L. B. & Mirra, S. S. Nuclear pore complex proteins in Alzheimer disease. J. Neuropathol. Exp. Neurol. 65, 45–54 (2006).

    Article  CAS  PubMed  Google Scholar 

  68. Eftekharzadeh, B. et al. Tau protein disrupts nucleocytoplasmic transport in Alzheimer’s disease. Neuron 99, 925–940.e7 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Grima, J. C. et al. Mutant huntingtin disrupts the nuclear pore complex. Neuron 94, 93–107.e6 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Li, N. & Lagier-Tourenne, C. Nuclear pores: the gate to neurodegeneration. Nat. Neurosci. 21, 156–158 (2018).

    Article  CAS  PubMed  Google Scholar 

  71. Zhang, K. et al. The C9orf72 repeat expansion disrupts nucleocytoplasmic transport. Nature 525, 56–61 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Chou, C. C. et al. TDP-43 pathology disrupts nuclear pore complexes and nucleocytoplasmic transport in ALS/FTD. Nat. Neurosci. 21, 228–239 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Larson, M. H. et al. CRISPR interference (CRISPRi) for sequence-specific control of gene expression. Nat. Protoc. 8, 2180–2196 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Alberts, N., Mathangasinghe, Y. & Nillegoda, N. B. In situ monitoring of transiently formed molecular chaperone assemblies in bacteria, yeast, and human cells. J. Vis. Exp. 151, https://doi.org/10.3791/60172 (2019).

  75. Kuipers, J., Kalicharan, R. D., Wolters, A. H. G., van Ham, T. J. & Giepmans, B. N. G. Large-scale scanning transmission electron microscopy (nanotomy) of healthy and injured zebrafish brain. J. Vis. Exp. 111, 12–14 (2016).

    Google Scholar 

Download references

Acknowledgements

E.F.E.K. is supported by a topmaster fellowship from the Groningen University Institute for Drug Exploration (GUIDE). H.H.K. and S.B. are supported by a grant from the CTH (grant no. 686728). L.M.V., H.H.K., S.B., M.K.M., P.G., T.B. and A.S. are supported by an NWO Groot (grant no. 685709). L.M.V., P.G., T.B. and A.S. are supported by a Vici grant (VI.C.192.031). M.M. is supported by an ALW Open Programme (ALWOP.355). We thank J. Brunsting and A. Blaauwbroek for practical assistance. We thank E. Lemke for providing us with the GFP-Nup153AG construct, D. Görlich for the yNup116FG and yNup145NFG constructs, and C. Schlieker for the HeLa 4TorKO cell line. We thank S. Cascarina for assistance with the LCD-Composer. We thank W. Browne for his help with the Circular dichroism measurements. Fluorescence and live cell imaging was performed in the UMCG Microscopy and Imaging Center (UMIC), sponsored by ZonMW grant 91111.006 and NWO 175-010-2009-023. EM 2D imaging was performed at the UMIC. Tomography imaging was performed at the EM facility of the Faculty of Science and Engineering of Groningen, the Netherlands.

Author information

Authors and Affiliations

Authors

Contributions

E.F.E.K., H.H.K., L.M.V. and S.B. conceived the project. The experiments in Figs. 1, 2 and 4 were designed, performed and analysed by E.F.E.K., the TEM in Fig. 1f by J.K., the EM and tomography in Fig. 3 by M.M., the in vitro data in Figs. 5 and 6 by P.G., T.B. and A.S., and the PLA and DNAJB6b mutant experiments in Figs. 4b and 6c by M.K.M. Supervision was done by H.H.K., L.M.V., A.S., B.N.G.G. and S.B. The manuscript was written by E.F.E.K., S.B. and L.M.V., with input from all authors.

Corresponding authors

Correspondence to E. F. Elsiena Kuiper, Harm H. Kampinga, Liesbeth M. Veenhoff or Steven Bergink.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Cell Biology thanks Janine Kirstein, and the other, anonymous reviewer(s) for their contribution to the peer review of this work. Peer reviewer reports are available.

Additional information

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

Extended data

Extended Data Fig. 1 Disruption of DNAJB6 expression leads to accumulation of annulate lamellae.

a, DNAJB6 in HEK293T cells localizes to the cytoplasm and nucleus, and in foci at the nuclear rim. A Z-projection is shown. Graph depicts a quantification of the distribution of the number of DNAJB6 foci per cell. n = 3 independent experiments with 1272 cells analyzed; Log2 graph box and whiskers minimum to maximum with all data points; median indicated with a line and mean with a+. b, Different cell lines (BeWo, U2OS and HeLA) stained for DNAJB6 (green) and FG-Nups (mAb414, magenta). c, HEK293T DNAJB6 KO cells stained for DNAJB6 (green) and Lamin B1 (magenta). d, Images of mAb414 or Nup153-stained nuclear pores (focused on the top of the nucleus) in HEK293T and HEK293T DNAJB6 KO cells. Quantification of average nearest neighbour distance (in pixels) shows larger distances between NPCs in DNAJB6 KO cells. Every point represents one cell; WT n = 32 individual cells, DNAJB6 KO n = 33 individual cells; mean ± SEM. e, siRNA-mediated knockdown of DNAJB6 (green) (72 h) induces cytoplasmic accumulations of NPCs stained by mAb414 (magenta). f, HEK293T cells transfected with siRNA against the 5’UTR of DNAJB6 show accumulations of NPCs and by simultaneous expression of GFP-DNAJB6bWT formation of these can be prevented. With a quantification of mAb414-positive accumulations after knockdown of DNAJB6. n = 3 independent experiments; unpaired t-test, 2-tailed p-value; *p = 0.0139, **p = 0.0097; mean ± SEM. g, In DNAJB6 KO cells, NPCs in the cytoplasm (annulate lamellae) visualized by transmission electron microscopy and pseudo-colored in bright pink, can be distinguished in cytoplasmic strings. Arrows point at individually identifiable NPCs. Nucleus indicated with N, mitochondrion indicated with M. Scale bar represents 500 nm. h, As in g, but now the white arrow head points at individually identifiable NPCs in the NE and black arrow heads at NPCs in the annulate lamellae. i, Individual channels of Fig. 1h. RanGap1, and different nucleoporins of the NPC Nup358, Nup133, Nup205, Nup98, TPR, Nup50, and Nup153 are stained (magenta) together with DNAJB6 (green) in WT and DNAJB6 KO cells. Source numerical data are available in source data.

Source data

Extended Data Fig. 2 Closely related JDPs of DNAJB6 also accumulate in foci and deletion can induce annulate lamellae.

a, Representative images of HEK293T and HEK293T DNAJB6 KO cells cultured in medium containing the uridine analog 5-ethynyluridine (5-EU). 5-EU incorporation into newly transcribed RNA was detected by Cy3-azide (green). The ratio of nuclear to cytoplasmic signal intensities of RNA relative to HEK293T WT cells. Each individual point represents a single cell; WT n = 3 independent experiments with 327 cells examined, DNAJB6 KO n = 3 independent experiments with 323 cells examined; unpaired t-test, 2-tailed p-value; ***p < 0.0001; mean ± SEM. b, Colocalization of GFP-tagged DNAJB6b, DNAJB8, and DNAJB2 with endogenous DNAJB6. DNAJB1 and DNAJA1 do not colocalize with DNAJB6 foci at the NE. c, annulate lamellae formation under siRNA-mediated knockdown of DNAJB6, DNAJB2, DNAJB1, and DNAJA1. Knockdown of DNAJB6 and DNAJB2 leads to significant increase in annulate lamellae formation. All experiments n = 3 independent experiments with cells examined: control 4392, DNAJB6b 5073, DNAJB2 4118, DNAJB1 4059, DNAJA1 4184; unpaired t-test, 2-tailed p-value; **p = 0.0048 mock vs DNAJB6b, **p = 0.003 mock vs DNAJB2; mean ± SEM. Scale bars on all fluorescent images represent 10 μm, and on magnifications 2 μm. Source numerical data are available in source data.

Source data

Extended Data Fig. 3 DNAJB6 foci formation is cell cycle dependent and is related to interphase NPC assembly.

a, Cell cycle progression profiles as analyzed by fluorescence-activated cell sorting (FACS). Cells were stained with propidium iodide to detect DNA content. b, DNAJB6 foci in HEK293T cells in asynchronized cells, and cells 1.5 h and 12 h after release from a double thymidine cell cycle synchronization. c, siRNA-mediated knockdown of ELYS and Nup153 in HEK293T cells visualized on SDS-PAGE with specific antibodies. d, e, Bar graphs of the data in Fig. 2b depicting a distribution of the amount of DNAJB6 foci per cell for the indicated siRNA treatment. NB: these conditions share the same control. d, *p = 0.0129, e, *p = 0.0304, **p = 0.0080, ***p = 0.0006; Control n = 5 independent experiments with 8298 cells examined, ELYS n = 4 independent experiments with 6839 cells examined, Nup153 n = 4 independent experiments with 7690 cells examined, automated with ICY; mean ± SEM. f, Quantification of percentage of cells with cytoplasmic NPC accumulations (AL) stained with mAb414 under siRNA knockdown of ELYS, Nup153, or Nup358. Control n = 5 independent experiments with 7317 cells examined, ELYS n = 4 independent experiments with 6323 cells examined, Nup153 n = 3 independent experiments with 4798 cells examined, Nup358 n = 2 independent experiments with 2264 cells examined; unpaired t-test, 2-tailed p-value; **p = 0.0037, ***p < 0.0001; mean ± SEM. g, qPCR on mRNA levels of Torsin A and B under control or knockdown conditions, normalized to GAPDH. Tor1A control n = 3 independent experiments, Tor1A, Tor1B control, and Tor1B n = 2 independent experiments. h, Bar graph of the data in Fig. 2c, depicting a distribution of the amount of DNAJB6 foci per cell. Control n = 3 independent experiments with 1272 cells examined, gRNA TOR1A + TOR1B n = 3 independent experiments with 952 cells examined, automated with ICY; mean is indicated with a cross; unpaired t-test, 2-tailed p-value; only significant differences are indicated; ***p = 0.0005; mean ± SEM. i, HeLa WT and HeLa 4TorKO cells stained for DNAJB6 (green) and FG-Nups with mAb414 (magenta). j, Overexpression of either TorsinA-GFP wildtype or ΔE302 (green) leads to an increase in foci at the nuclear envelope, containing DNAJB6 (magenta). k, Ubiquitin (magenta) colocalizes with DNAJB6 (green) at foci at the nuclear envelope and are increased upon Torsin knockdown (gRNA vector containing mTurquoise2 in the blue channel). Scale bars on all fluorescent images represent 10 μm, and on magnifications 2 μm. Source numerical data are available in source data.

Source data

Extended Data Fig. 4 DNAJB6 localizes to herniations at the NE and is dependent on intact NPCs.

a, For immunoelectron tomography, surface labelling was used to select areas of interest prior to recording tilt series. Tomographic slice (Z slice 4) revealing the specific immunogold (15 nm) labelling on the surface of the cryosection. Asterisk indicating herniations. Indicated tomographic slices extracted from the tomogram showing herniations at the NE. In the bottom images, nuclear membranes are traced (pink). More electron dense areas can be observed at the neck of the herniations where the NE bends into the herniation, indicated by black arrow heads. NPCs are indicated with white arrow heads. Nucleus is indicated with N and nuclear envelope with NE. Scale bars represent 232 nm. Lower images are from the three-dimensional model (see Supplementary Video 2) of DNAJB6-positive herniations, obtained by electron tomography of 400 to 450 nm thick cryosections. The NE is represented in pink. Left is a view from the nuclear side showing the openings of the NPCs and of the herniations. Right shows the herniations with the localization of the DNAJB6 gold labelling in red. b, HSP70 (magenta) colocalizes with DNAJB6 (green) in foci at the NE. c, MLF2-GFP colocalizes with DNAJB6 (magenta) in foci at the NE. d, WT HEK293T cells treated with 5% 1,6-hexanediol for 2 minutes lose almost all DNAJB6 foci (green), while cells treated with 2,5-hexanediol have only a slight reduction in foci. Scale bars on all fluorescent images represent 5 μm, and on magnifications 2 μm.

Extended Data Fig. 5 The interaction of DNAJB6 with hNup153FG depends on the FG-motifs in hNup153FG.

a, Schematic overview of the BioID2-DNAJB6b fusion protein and how samples were obtained. Fluorescent image shows HEK293T cells expressing BioID2-DNAJB6b, treated for 24 h with 50 μM biotin and stained with Alexa Fluor 488-conjugated streptavidin, biotinylated substrates are visualized in foci at the NE. b, Streptavidin blot belonging Fig. 4a where HEK293T cells expressing V5-DNAJB6b or BioID2-DNAJB6b are treated for 24 h with 50 μM biotin. c, Proximity ligation assay (PLA) performed with antibodies against FG-Nups (mAb414) or DNAJB6 alone, or only both secondary antibodies as negative technical controls for PLA. d, SDS-PAGE showing increase in HSPA6 levels after heat shock (43 °C for 30 minutes) but not in DNAJB6 KO nor after overexpression of hNup153FG. e, GFP-Nup153 full length localizes predominantly to the NE in HEK293T cells. Due to overexpression, there is also small clustering at the NE, but no aggregates can be observed. f, GFP-Nup153AG overexpression in HEK293T cells is homogeneously distributed in the cytoplasm. g, HEK293T cells overexpressing GFP-Nup153FG and mCherry-DNAJB6b show colocalization in spherical accumulations (arrowheads). h, Filter-trap assay of HEK293T cells with GFP-Nup153AG or GFP-Nup153FG overexpression. Three serial 5-fold dilutions were loaded onto cellulose-acetate membranes and probed with anti-GFP antibodies to detect aggregation of the GFP-Nup153FG or GFP-Nup153AG fragment without or with co-overexpression of V5-DNAJB6b. i, HEK293T cells overexpressing V5-DNAJB6b or BioID2-DNAJB6b, either with GFP-Nup153FG or GFP-Nup153AG, are treated for 24 h with 50 μM biotin. Biotinylated close proximity partners of DNAJB6b are pulled down by biotin affinity immunoprecipitation with streptavidin beads, run on SDS-PAGE. GFP and DNAJB6 are detected with specific antibodies. Scale bars on all fluorescent images represent 10 μm. Source numerical data and unprocessed blots are available in source data.

Source data

Extended Data Fig. 6 The effect of crowding agents and DNAJB6b on the aggregation of human and yeast FG-Nup fragments in vitro.

a, 6 μM of purified FG-Nup fragments were incubated in assay buffer (150 mM NaCl, 50 mM TrisHCl, pH 8) containing the indicated concentration (w/v) of the crowding agents Serine, PEG3350, or Ficoll, at 25 °C for 1 hour, and loaded on a filter trap. Graphs show quantification of Nup band intensity in the described conditions. n = 3 independent experiments; mean ± SEM. hNup153FG did not form any quantifiable bands on filter trap with the addition of Serine or Ficoll. b-e, Individual repetitions of the data in Fig. 5a. The effect of DNAJB6b on the aggregation of human and yeast FG-Nup fragments in vitro. Filter trap assays to detect aggregation of purified FG-Nup fragments, incubated with the indicated molar ratios of DNAJB6b, probed with anti-His antibodies to detect FG-Nup fragments. hNup153FG (b), yNup100FG (c), and yNup116FG (d) were incubated in the assay buffer (150 mM NaCl, 50 mM TrisHCl, pH 8) containing 10% PEG3350 (w/v), at 25 °C for either 1 hour (hNup153FG, yNup100FG) or 3 hours (yNup116FG), and yeast Nup145NFG (e) was incubated in a buffer comprising 50 mM NaCl, 100 mM sodium phosphate, pH6, 10% PEG3350 (w/v), at 25 °C during 1 hour. Graphs show band intensities on filter trap quantified relative to the average intensity in the absence of DNAJB6b. Source numerical data and unprocessed blots are available in source data.

Source data

Extended Data Fig. 7 Individual repetitions of the data in Fig. 5b.

a, b, The effect of control proteins relative to DNAJB6b on the aggregation of human and yeast FG-Nup fragments in vitro. 6 μM of purified hNup153FG (a) or yNup100FG (b) were incubated in the absence (control) or presence of BSA, MBP-mCherry, DNAJA1, or DNAJB6b at a 6 μM concentration (molar ratio 1:1). BSA and MBP-mCherry served as non-chaperone controls and DNAJA1 served as chaperone control. Graphs represent band intensities relative to the average intensity of the FG-Nup fragments alone (control) of each replicate. n = 3 independent experiments. c, d, 6 μM of purified hNup153FG (c) or yNup100FG (d) were incubated in the absence (control) or presence of the GFP-tagged intrinsically disordered proteins (IDPs) 1 and 2 (IDP1 is the disordered region of yHeh2; IDP2 is a synthetic IDP; for details see methods) at a 6 μM concentration (molar ratio 1:1). Band intensities are quantified relative to the average intensity of the FG-Nup fragments alone. Source numerical data and unprocessed blots are available in source data.

Extended Data Fig. 8 Individual repetitions of the data in Fig. 5c.

a, b, The effect of DNAJB6 on the time-dependent formation of hNup153FG and yNup100FG aggregates in vitro. Formation of hNup153FG (a) and yNup100FG (b) aggregates captured on filter trap over time in the absence and presence of DNAJB6b at a molar ratio of 1:1. Time is indicated in minutes. Graphs depict quantification of Nup band intensity (in arbitrary units, a.u.) over time in the absence (green) or presence (grey) of DNAJB6b. Source numerical data and unprocessed blots are available in source data.

Extended Data Fig. 9 Condensation of DNAJB6b and FG-Nup fragments, and individual repetitions of the data in Fig. 6b.

a, DNAJB6b condensates by itself and is SDS soluble. Purified DNAJB6b was incubated with or without 10% PEG3350 (w/v). After 1 hour incubation, either 5% 2.5-hexanediol, 5% 1.6-hexanediol, or 0.5% SDS was added for 10 minutes and bright field images were taken. b, Fluorescence and bright field microscopy images showing co-localisation of Fluorescein-5-Maleimide labelled hNup153FG and yNup100FG in the absence or the presence of unlabelled DNAJB6b at a 3 μM concentration (molar ratio 1:1). Deconvolved fluorescence images are depicted as maximum Z-Projection of 30 consecutive slices of 0.2 micron. Scale bar represents 2 μm. c, Coomassie-stained SDS-PAGE showing one replicate of the sedimentation assay (quantified in Fig. 5e-f) to assess the soluble fraction of yNup100FG (3 μM) in the presence or absence of DNAJB6b (1.5 μM) for the indicated times. To determine the fraction 1,6-hexanediol soluble condensates, 10% 1,6-hexanediol was added for 10 minutes prior to centrifugation. Total yNup100FG protein is slightly shifted on the blot due to effects of PEG on mobility. d, Individual repetitions of the data in Fig. 6b. Filter-trap assays showing the effect of wildtype or indicated DNAJB6 mutants (18x S/T > A, 12x F > A, Δ79-115) on the aggregation of yNup100FG (3 μM; molar ratio 1:1). Graphs represent band intensities relative to the average intensity of the yNup100FG fragment alone (control). Source numerical data and unprocessed blots are available in source data.

Source data

Extended Data Fig. 10 Characterization of purified FG-Nup fragments, control proteins and DNAJB6 mutants.

a, Coomassie Brilliant Blue stained SDS-PAGE of indicated purified protein samples. b, Circular dichroism spectroscopy of DNAJB6b and DNAJB6b mutants (18x S/T > A, 12x F > A, and Δ79-115). Purified proteins were diluted to a concentration of either 0.2 μM (DNAJB6b WT), 0.67 μM (18xS/T > A) or 15 μM (12xF > A and Δ79-115) in the assay buffer (50 mM TrisHCl, 150 mM NaCl, pH 8.0). Ellipticity was recorded between 500 and 200 nm in a quartz (QS) cuvette. Data was converted to Molar ellipticity ([q]) to correct for protein concentration, cuvette pathlength and number of backbone peptide units. Source numerical data are available in source data.

Source data

Supplementary information

Reporting Summary

Peer Review File

Supplementary Table 1

Amino acid composition comparison with the S+T, G and F content of the S/T domain of DNAJB6.

Supplementary Video 1

HEK293T cells stained for DNAJB6 (green) showing foci at the NE, Z-stacks. Hoechst in blue.

Supplementary Video 2

Tomography 3D model of NE herniation; DNAJB6 stained with anti-DNAJB6.

Supplementary Video 3

Tomography 3D model of NE herniation; GFP-DNAJB6b stained with anti-GFP.

Source data

Source Data Fig. 1

Numerical source.

Source Data Fig. 2

Numerical source.

Source Data Fig. 4

Unprocessed blot.

Source Data Fig. 4

Numerical source.

Source Data Fig. 5

Unprocessed blot.

Source Data Fig. 5

Numerical source.

Source Data Fig. 6

Unprocessed blots and gels.

Source Data Fig. 6

Numerical source.

Source Data Extended Data Fig. 1

Numerical source.

Source Data Extended Data Fig. 2

Numerical source.

Source Data Extended Data Fig. 3

Unprocessed blot.

Source Data Extended Data Fig. 3

Numerical source.

Source Data Extended Data Fig. 5

Numerical source.

Source Data Extended Data Fig. 6

Unprocessed blot.

Source Data Extended Data Fig. 6

Numerical source.

Source Data Extended Data Fig. 9

Unprocessed blot.

Source Data Extended Data Fig. 10

Numerical source.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Kuiper, E.F.E., Gallardo, P., Bergsma, T. et al. The chaperone DNAJB6 surveils FG-nucleoporins and is required for interphase nuclear pore complex biogenesis. Nat Cell Biol 24, 1584–1594 (2022). https://doi.org/10.1038/s41556-022-01010-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41556-022-01010-x

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