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:

Targeted deubiquitination rescues distinct trafficking-deficient ion channelopathies

Abstract

Impaired protein stability or trafficking underlies diverse ion channelopathies and represents an unexploited unifying principle for developing common treatments for otherwise dissimilar diseases. Ubiquitination limits ion channel surface density, but targeting this pathway for the purposes of basic study or therapy is challenging because of its prevalent role in proteostasis. We developed engineered deubiquitinases (enDUBs) that enable selective ubiquitin chain removal from target proteins to rescue the functional expression of disparate mutant ion channels that underlie long QT syndrome (LQT) and cystic fibrosis (CF). In an LQT type 1 (LQT1) cardiomyocyte model, enDUB treatment restored delayed rectifier potassium currents and normalized action potential duration. CF-targeted enDUBs synergistically rescued common (ΔF508) and pharmacotherapy-resistant (N1303K) CF mutations when combined with the US Food and Drug Administation (FDA)-approved drugs Orkambi (lumacaftor/ivacaftor) and Trikafta (elexacaftor/tezacaftor/ivacaftor and ivacaftor). Altogether, targeted deubiquitination via enDUBs provides a powerful protein stabilization method that not only corrects diverse diseases caused by impaired ion channel trafficking, but also introduces a new tool for deconstructing the ubiquitin code in situ.

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: enDUBs reverse NEDD4L-mediated ubiquitination of KCNQ1.
Fig. 2: enDUBs rescue trafficking-deficient mutant LQT1 channels in HEK293 cells.
Fig. 3: enDUBs restore action potential duration in an LQT1 cardiomyocyte model.
Fig. 4: enDUBs facilitate rescue of mutant CFTR channels in combination with Orkambi.
Fig. 5: CF-targeted enDUB combination therapy functionally rescues rare trafficking-deficient CFTR mutations.
Fig. 6: Dual-acting enDUBs synergize to improve the functional rescue and apical localization of the most common ΔF508 mutation.

Similar content being viewed by others

Data availability

All data generated or analyzed during this study are included in this published article (and its Extended Data files). All supporting data are available from the corresponding author upon reasonable request. The CFTR structure was reproduced from PDB (PDB: 5UAK). The nanobody structure was reproduced from PDB (3K1K).

References

  1. Kullmann, D. M. Neurological channelopathies. Annu. Rev. Neurosci. 33, 151–172 (2010).

    CAS  PubMed  Google Scholar 

  2. Bohnen, M. S. et al. Molecular pathophysiology of congenital long QT syndrome. Physiol. Rev. 97, 89–134 (2016).

    PubMed Central  Google Scholar 

  3. Cutting, G. R. Cystic fibrosis genetics: from molecular understanding to clinical application. Nat. Rev. Genet. 16, 45–56 (2014).

    PubMed  PubMed Central  Google Scholar 

  4. Ashcroft, F. M. & Rorsman, P. KATP channels and islet hormone secretion: new insights and controversies. Nat. Rev. Endocrinol. 9, 660–669 (2013).

    PubMed Central  Google Scholar 

  5. Imbrici, P. et al. Therapeutic approaches to genetic ion channelopathies and perspectives in drug discovery. Front. Pharmacol. 7, 121 (2016).

    PubMed  PubMed Central  Google Scholar 

  6. Wulff, H., Christophersen, P., Colussi, P., Chandy, G. K. & Yarov-Yarovoy, V. Antibodies and venom peptides: new modalities for ion channels. Nat. Rev. Drug Discov. 18, 339–357 (2019).

  7. Tester, D. J., Will, M. L., Haglund, C. M. & Ackerman, M. J. Compendium of cardiac channel mutations in 541 consecutive unrelated patients referred for long QT syndrome genetic testing. Heart Rhythm 2, 507–517 (2005).

    PubMed  Google Scholar 

  8. Wilson, A. J., Quinn, K. V., Graves, F. M., Bitner-Glindzicz, M. & Tinker, A. Abnormal KCNQ1 trafficking influences disease pathogenesis in hereditary long QT syndromes (LQT1). Cardiovasc. Res. 67, 476–486 (2005).

    CAS  PubMed  Google Scholar 

  9. Haardt, M., Benharouga, M., Lechardeur, D., Kartner, N. & Lukacs, G. L. C-terminal truncations destabilize the cystic fibrosis transmembrane conductance regulator without impairing its biogenesis. A novel class of mutation. J. Biol. Chem. 274, 21873–21877 (1999).

    CAS  PubMed  Google Scholar 

  10. Cheng, S. H. et al. Defective intracellular transport and processing of CFTR is the molecular basis of most cystic fibrosis. Cell 63, 827–834 (1990).

    CAS  PubMed  Google Scholar 

  11. Curran, J. & Mohler, P. J. Alternative paradigms for ion channelopathies: disorders of ion channel membrane trafficking and posttranslational modification. Annu. Rev. Physiol. 77, 505–524 (2015).

    CAS  PubMed  Google Scholar 

  12. Huang, H. et al. Mechanisms of KCNQ1 channel dysfunction in long QT syndrome involving voltage sensor domain mutations. Sci. Adv. 4, eaar2631 (2018).

    PubMed  PubMed Central  Google Scholar 

  13. Foot, N., Henshall, T. & Kumar, S. Ubiquitination and the regulation of membrane proteins. Physiol. Rev. 97, 253–281 (2017).

    PubMed  Google Scholar 

  14. Nalepa, G., Rolfe, M. & Harper, W. J. Drug discovery in the ubiquitin–proteasome system. Nat. Rev. Drug Discov. 5, 596–613 (2006).

    CAS  PubMed  Google Scholar 

  15. Huang, X. & Dixit, V. M. Drugging the undruggables: exploring the ubiquitin system for drug development. Cell Res. 26, 484–498 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Jespersen, T. et al. The KCNQ1 potassium channel is down-regulated by ubiquitylating enzymes of the Nedd4/Nedd4-like family. Cardiovasc. Res. 74, 64–74 (2007).

    CAS  PubMed  Google Scholar 

  17. Mevissen, T. et al. OTU deubiquitinases reveal mechanisms of linkage specificity and enable ubiquitin chain restriction analysis. Cell 154, 169–184 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Rothbauer, U. et al. A versatile nanotrap for biochemical and functional studies with fluorescent fusion proteins. Mol. Cell. Proteomics 7, 282–289 (2008).

    CAS  PubMed  Google Scholar 

  19. Abbott, G. W. et al. KCNQ1, KCNE2, and Na+-coupled solute transporters form reciprocally regulating complexes that affect neuronal excitability. Sci. Signal. 7, ra22 (2014).

    PubMed  PubMed Central  Google Scholar 

  20. Aromolaran, A. S., Subramanyam, P., Chang, D. D., Kobertz, W. R. & Colecraft, H. M. LQT1 mutations in KCNQ1 C-terminus assembly domain suppress IKs using different mechanisms. Cardiovasc. Res. 104, 501–511 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Peroz, D., Dahimène, S., Baró, I., Loussouarn, G. & Mérot, J. LQT1-associated mutations increase KCNQ1 proteasomal degradation independently of Derlin-1. J. Biol. Chem. 284, 5250–5256 (2009).

    CAS  PubMed  Google Scholar 

  22. Mattmann, M. E. et al. Identification of (R)-N-(4-(4-methoxyphenyl)thiazol-2-yl)-1-tosylpiperidine-2-carboxamide, ML277, as a novel, potent and selective Kv7.1 (KCNQ1) potassium channel activator. Bioorg. Med. Chem. Lett. 22, 5936–5941 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Veit, G. et al. From CFTR biology toward combinatorial pharmacotherapy: expanded classification of cystic fibrosis mutations. Mol. Biol. Cell 27, 424–433 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Boeck, K. & Amaral, M. D. Progress in therapies for cystic fibrosis. Lancet Respir. Med. 4, 662–674 (2016).

    PubMed  Google Scholar 

  25. Wainwright, C. E. et al. Lumacaftor–ivacaftor in patients with cystic fibrosis homozygous for Phe508del CFTR. N. Engl. J. Med. 373, 220–231 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Goor, F. et al. Correction of the F508del-CFTR protein processing defect in vitro by the investigational drug VX-809. Proc. Natl Acad. Sci. USA 108, 18843–18848 (2011).

    PubMed  Google Scholar 

  27. Goor, F. et al. Rescue of CF airway epithelial cell function in vitro by a CFTR potentiator, VX-770. Proc. Natl Acad. Sci. USA 106, 18825–18830 (2009).

    PubMed  Google Scholar 

  28. Farinha, C. M. & Matos, P. Repairing the basic defect in cystic fibrosis—one approach is not enough. FEBS J. 283, 246–264 (2016).

    CAS  PubMed  Google Scholar 

  29. Faesen, A. C. et al. The differential modulation of USP activity by internal regulatory domains, interactors and eight ubiquitin chain types. Chem. Biol. 18, 1550–1561 (2011).

    CAS  PubMed  Google Scholar 

  30. Liu, F., Zhang, Z., Csanády, L., Gadsby, D. C. & Chen, J. Molecular structure of the human CFTR ion channel. Cell 169, 85–95 (2017).

    CAS  PubMed  Google Scholar 

  31. McMahon, C. et al. Yeast surface display platform for rapid discovery of conformationally selective nanobodies. Nat. Struct. Mol. Biol. 25, 289–296 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Galietta, L. V., Jayaraman, S. & Verkman, A. S. Cell-based assay for high-throughput quantitative screening of CFTR chloride transport agonists. Am. J. Physiol. Cell Physiol. 281, C1734–C1742 (2001).

    Google Scholar 

  33. Durmowicz, A. G., Lim, R., Rogers, H., Rosebraugh, C. J. & Chowdhury, B. A. The U.S. Food and Drug Administration’s experience with ivacaftor in cystic fibrosis. Establishing efficacy using in vitro data in lieu of a clinical trial. Ann. Am. Thorac. Soc. 15, 1–2 (2018).

    PubMed  Google Scholar 

  34. Han, S. T. et al. Residual function of cystic fibrosis mutants predicts response to small molecule CFTR modulators. JCI Insight 3, e121159 (2018).

  35. Goor, F., Yu, H., Burton, B. & Hoffman, B. J. Effect of ivacaftor on CFTR forms with missense mutations associated with defects in protein processing or function. J. Cyst. Fibros. 13, 29–36 (2014).

    PubMed  Google Scholar 

  36. Middleton, P. G. et al. Elexacaftor–tezacaftor–ivacaftor for cystic fibrosis with a single Phe508del allele. N. Engl. J. Med. 381, 1809–1819 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Lukacs, G. L. & Verkman, A. S. CFTR: folding, misfolding and correcting the ΔF508 conformational defect. Trends Mol. Med. 18, 81–91 (2012).

    CAS  PubMed  Google Scholar 

  38. Okiyoneda, T. et al. Peripheral protein quality control removes unfolded CFTR from the plasma membrane. Science 329, 805–810 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Sigoillot, M. et al. Domain-interface dynamics of CFTR revealed by stabilizing nanobodies. Nat. Commun. 10, 2636 (2019).

  40. Kreda, S. M. et al. Characterization of wild-type and ΔF508 cystic fibrosis transmembrane regulator in human respiratory epithelia. Mol. Biol. Cell 16, 2154–2167 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. van Meegen, M. A., Terheggen-Lagro, S. W., Koymans, K. J., van der Ent, C. K. & Beekman, J. M. Apical CFTR expression in human nasal epithelium correlates with lung disease in cystic fibrosis. PLoS ONE 8, e57617 (2013).

    PubMed  PubMed Central  Google Scholar 

  42. Fukuda, R. & Okiyoneda, T. Peripheral protein quality control as a novel drug target for CFTR stabilizer. Front. Pharmacol. 9, 1100 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Meacham, G. C., Patterson, C., Zhang, W., Younger, J. M. & Cyr, D. M. The Hsc70 co-chaperone CHIP targets immature CFTR for proteasomal degradation. Nat. Cell Biol. 3, 100–105 (2001).

    CAS  PubMed  Google Scholar 

  44. Delisle, B. P. et al. Biology of cardiac arrhythmias. Circ. Res. 94, 1418–1428 (2004).

    CAS  PubMed  Google Scholar 

  45. Schapira, M., Calabrese, M. F., Bullock, A. N. & Crews, C. M. Targeted protein degradation: expanding the toolbox. Nat. Rev. Drug Discov. 18, 949–963 (2019).

    CAS  PubMed  Google Scholar 

  46. Kanner, S. A., Morgenstern, T. & Colecraft, H. M. Sculpting ion channel functional expression with engineered ubiquitin ligases. eLife 6, e29744 (2017).

  47. Caussinus, E., Kanca, O. & Affolter, M. Fluorescent fusion protein knockout mediated by anti-GFP nanobody. Nat. Struct. Mol. Biol. 19, 117–121 (2011).

    PubMed  Google Scholar 

  48. Mullard, A. First targeted protein degrader hits the clinic. Nat. Rev. Drug Discov. https://doi.org/10.1038/d41573-019-00043-6 (2019).

    Article  PubMed  Google Scholar 

  49. Wu, T. et al. Targeted protein degradation as a powerful research tool in basic biology and drug target discovery. Nat. Struct. Mol. Biol. 27, 605–614 (2020).

    CAS  PubMed  Google Scholar 

  50. Shi, D. & Grossman, S. R. Ubiquitin becomes ubiquitous in cancer: emerging roles of ubiquitin ligases and deubiquitinases in tumorigenesis and as therapeutic targets. Cancer Biol. Ther. 10, 737–747 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Isaacson, M. K. & Ploegh, H. L. Ubiquitination, ubiquitin-like modifiers, and deubiquitination in viral infection. Cell Host Microbe 5, 559–570 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Fridy, P. C. et al. A robust pipeline for rapid production of versatile nanobody repertoires. Nat. Methods 11, 1253–1260 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Sekine-Aizawa, Y. & Huganir, R. L. Imaging of receptor trafficking by using α-bungarotoxin-binding-site-tagged receptors. Proc. Natl Acad. Sci. USA 101, 17114–17119 (2004).

    CAS  PubMed  Google Scholar 

  54. Gao, S. et al. Ubiquitin ligase Nedd4L targets activated Smad2/3 to limit TGF-β signaling. Mol. Cell 36, 457–468 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Peters, K. W. et al. CFTR folding consortium: methods available for studies of CFTR folding and correction. Methods Mol. Biol. 742, 335–353 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Galietta, L. J., Haggie, P. M. & Verkman, A. S. Green fluorescent protein-based halide indicators with improved chloride and iodide affinities. FEBS Lett. 499, 220–224 (2001).

    CAS  PubMed  Google Scholar 

  57. Gentzsch, M. et al. Pharmacological rescue of conditionally reprogrammed cystic fibrosis bronchial epithelial cells. Am. J. Respir. Cell Mol. Biol. 56, 568–574 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  58. Zhang, C. et al. Long-term in vitro expansion of epithelial stem cells enabled by pharmacological inhibition of PAK1–ROCK–Myosin II and TGF-β signaling. Cell Rep. 25, 598–610 (2018).

    PubMed  PubMed Central  Google Scholar 

  59. Rayner, R. E., Makena, P., Prasad, G. L. & Cormet-Boyaka, E. Optimization of normal human bronchial epithelial (NHBE) cell 3D cultures for in vitro lung model studies. Sci. Rep. 9, 500 (2019).

    PubMed  PubMed Central  Google Scholar 

  60. Kanner, S. A., Jain, A. & Colecraft, H. M. Development of a high-throughput flow cytometry assay to monitor defective trafficking and rescue of long QT2 mutant hERG channels. Front. Physiol. 9, 397 (2018).

    PubMed  PubMed Central  Google Scholar 

  61. Lee, S.-R., Sang, L. & Yue, D. T. Uncovering aberrant mutant PKA function with flow cytometric FRET. Cell Rep. 14, 3019–3029 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank M. Chen for technical support, M.B. Johny (Columbia University) for help with the flow cytometry–FRET assay and helpful discussions and L. Pon (Columbia University) for guidance in initiating yeast cultures. This work was supported by grants RO1-HL121253 and 1RO1-HL122421 from the NIH (to H.M.C.). S.A.K. was supported by a Medical Scientist Training Program grant (T32 GM007367) and an NHLBI National Research Service Award (1F30-HL140878). This work was also supported by the TRx Accelerator of the CUMC Irving Institute, as supported by the NCATS, NIH (UL1TR001873). Flow cytometry experiments were performed in the CCTI Flow Cytometry Core, supported in part by the NIH (S10RR027050). Confocal images were collected in the HICCC Confocal and Specialized Microscopy Shared Resource, supported by the NIH (P30 CA013696).

Author information

Authors and Affiliations

Authors

Contributions

S.A.K. and H.M.C. conceived of the study and designed experiments. Z.S. performed CFTR electrophysiological measurements and analyses. P.C. contributed to molecular biology and the isolation of cardiomyocytes. A.J. contributed to KCNQ1 flow cytometry experiments. S.A.K. and H.M.C. wrote the manuscript; H.M.C. supervised the project.

Corresponding author

Correspondence to Henry M. Colecraft.

Ethics declarations

Competing interests

S.A.K. and H.M.C. have filed a patent application through Columbia University based on this work.

Additional information

Peer review information Rita Strack was the primary editor 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 KCNQ1 pulldown and ubiquitin analyses after cell lysis with modified RIPA buffer containing 1% SDS.

a, Left, KCNQ1 pulldowns probed with anti-KCNQ1 antibody from HEK293 cells expressing KCNQ1-YFP ± NEDD4L with nano alone or enDUB-O1. The four bands represent KCNQ1 monomer, dimer, trimer, and tetrameric species, respectively. Right, Anti-ubiquitin labeling of KCNQ1 pulldowns after stripping previous blot. b, Relative KCNQ1 ubiquitination computed by ratio of anti-ubiquitin to anti-KCNQ1 signal intensity (n = 3 independent experiments; mean). **p < 0.003, one-way ANOVA with Tukey’s multiple comparison test.

Source data

Extended Data Fig. 2 Flow cytometry gating strategy for BTX-647 surface labeling experiments.

a, Flow cytometry pseudocolor dot plots displaying an initial selection gate (left) for cells (vs debris); and a second selection gate (right) for singlets (vs doublets). b, Single color fluorescent controls after applying the gating strategy in a. c, Analysis gate established with single color controls in b to quantify channel surface density (BTX-647) in YFP- and CFP-positive cells (left) with cumulative distribution histograms (right). Sample 1 is exemplified by BBS-Q1-YFP + CFP-P2a-nano. Sample 2 is exemplified by BBS-Q1-YFP + CFP-P2a-nano + NEDD4L.

Extended Data Fig. 3 enDUB-O1 requires catalytic activity and target specificity for ubiquitin-dependent rescue of KCNQ1 channels.

a, (Left) Schematic of experimental strategy; BBS-Q1-YFP was co-transfected with either nanobody alone (grey line), NEDD4L + nano (red line), or NEDD4L + enDUB-O1 (blue line). (Right) Cumulative distribution histograms of Alexa647 fluorescence from flow cytometry analyses (data adapted from Fig. 1). Plot generated from population of YFP- and CFP-positive cells (n ≥ 5000 cells per experiment; N = 4). b, Same experiment as in a, but using catalytically inactive enDUB-O1* with C320S (N = 2). c, Same experiment as in a, but with untagged BBS-Q1 co-expressed with enDUB-O1 as a control for target specificity (N = 2).

Extended Data Fig. 4 enDUB-U21 has greater efficacy than enDUB-O1 in surface rescue of N1303K CFTR mutant channels.

a, Cumulative distribution histograms of Alexa647 fluorescence from flow cytometry analyses for cells expressing WT BBS-CFTR-YFP + nano (dotted line) and N1303K mutation co-expressing nano alone (red line), enDUB-O1 (cyan line), enDUB-U21 (blue line). Plot generated from population of YFP- and CFP-positive cells (n ≥ 5000 cells per experiment; N = 2). b, Same experimental design as above but with 24 hour incubation of VX809 with nano (green line), enDUB-O1 (cyan line) and enDUB-U21 (blue line) (N = 2).

Extended Data Fig. 5 enDUB-U21 requires catalytic activity and target specificity for ubiquitin-dependent rescue of CFTR mutants.

a, (Left) Schematic of experimental strategy; WT BBS-CFTR-YFP + nano (dashed line) or N1303K mutants co-transfected with nano (red line) or enDUB-U21 (blue line). Cumulative distribution histograms (middle) and quantification (right) of Alexa647 fluorescence from flow cytometry analyses (data adapted from Fig. 4). Plots generated from population of YFP- and CFP-positive cells (n ≥ 5000 cells per experiment; N = 3; mean ± s.e.m). Data are normalized to values from the WT CFTR control group (dotted line). b, Same experiment as in a, but using catalytically inactive enDUB-U21* with C221S (N = 3). c, Same experiment as in a, but with an mCherry-targeted nanobody, m-enDUB-U21, as a control for target specificity (N = 4).

Source data

Extended Data Fig. 6 enDUB-U21 increases functional rescue of 4326delTC CFTR mutant channels in combination with lumacaftor ± ivacaftor.

a, Exemplar family of basal (top, black), forskolin-activated (middle, red), and VX770-potentiated (bottom, green) currents for 4326delTC mutant channels after 24 hr VX809 treatment (3 µM) and co-expression with nano (left) or enDUB-U21 (right). b, Population I-V curves for basal (black squares), forskolin-activated (red circles), and VX770-potentiated (green triangles) currents from 4326delTC mutants co-expressing nano alone (left; n = 15) or enDUB-U21 (right; n = 14). c, Same format as a, but with N1303K mutants. d, Same format as b, but with N1303K mutants co-expressing nano alone (left; n = 9) or enDUB-U21 (right; n = 11). Data for VX770-potentiated currents adapted from Fig. 4.

Source data

Extended Data Fig. 7 Development of NBD1 binders from a yeast surface display nanobody library.

a, On-yeast binding affinity measurements of 9 nanobody clones using serial dilutions of purified FLAG-NBD1. b, Flow cytometric surface labeling assay and cumulative distribution histograms of WT CFTR surface density alone (dotted line) or when co-expressed with nanobody clones.

Extended Data Fig. 8 enDUB-U21CF.E3h deubiquitinates WT CFTR and N1303K.

a, Anti-ubiquitin labeling of WT CFTR and N1303K pulldowns. Lane 1 – untransfected cells; lane 2 – mCherry-CFTR + nanobody; lane 3 – mCherry-CFTR + enDUB-U21CF.E3h; lane 4 – mCherry-N1303K + nanobody; lane 5 – mCherry-N1303K + enDUB-U21CF.E3h. Proteins were pulled down with anti-mCherry and probed with anti-ubiquitin antibody (n = 2 independent experiments). b, Anti-CFTR labeling of WT CFTR and N1303K pulldowns. The top blot was stripped and probed with anti-CFTR antibody.

Source data

Extended Data Fig. 9 enDUB-U21 restores functional currents of N1303K and F508del CFTR mutant channels in combination with Trikafta.

a, Population I-V curves for forskolin-activated currents from FRT cells stably expressing WT CFTR (black circles, n = 57) and N1303K + CFP (red squares, n = 8), and forskolin-activated, VX770-potentiated currents from N1303K + CFP treated with VX661 + VX445 (green triangles, n = 9); N1303K + enDUB-U21CF.E3h treated with VX661 + VX445 (blue triangles, n = 9); n cells examined over ≥3 independent experiments (mean ± s.e.m). Data for N1303K + CFP adapted from Fig. 5. b, Population I-V curves for forskolin-activated currents from FRT cells stably expressing WT CFTR (black circles, n = 57) and F508del + CFP (red squares, n = 12), and forskolin-activated, VX770-potentiated currents from F508del + CFP treated with VX661 + VX445 (green triangles, n = 10); F508del + enDUB-U21CF.T2a treated with VX661 + VX445 (blue triangles, n = 9); n cells examined over ≥3 independent experiments (mean ± s.e.m). Data for F508del + CFP adapted from Fig. 6. *p < 0.03, **p < 0.009,***p < 0.0001, two-way ANOVA with Tukey’s multiple comparison test.

Source data

Extended Data Fig. 10 Mucociliary differentiation of human bronchial epithelial cells (hBECs) cultured at air-liquid interface (ALI).

a, H&E staining of immature (left; 5 day old) and mature hBEC ALI cultures (middle; 6 weeks), featuring a pseudostratified epithelium with mucin-containing goblet cells (*) (right). Apical (ap) and basal (bs) compartments labeled. b, Immunofluorescence staining of mature ALI cultures, featuring ciliated cells (green; acetylated-tubulin), mucin-containing goblet cells (pink; MUC5AC), and a basal cell layer (red; CK5), and merged image with DAPI staining (blue; nuclei). c, Immunostaining of mature WT hBEC cultures, with anti-CFTR signal (heat map) and anti-ezrin (green) labeling of the apical membrane. White box inset highlights the apical membrane (far right).

Supplementary information

Source data

Source Data Fig. 1

Data for bar charts and unprocessed western blots

Source Data Fig. 2

Source data for bar charts, IV curves and unprocessed western blots

Source Data Fig. 3

Source data for Ipeak and APD90 bar charts

Source Data Fig. 4

Source data for surface density bar charts and IV curves

Source Data Fig. 5

Source data for K-slope bar charts and IV curves

Source Data Fig. 6

Source data for surface density bar charts and IV curves

Source Data Extended Data Fig. 1

Source data for ubiquitin bar charts and unprocessed western blots

Source Data Extended Data Fig. 5

Source data for surface density bar charts

Source Data Extended Data Fig. 6

Source data for IV curves

Source Data Extended Data Fig. 8

Unprocessed Western blots

Source Data Extended Data Fig. 9

Source data for IV curves

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Kanner, S.A., Shuja, Z., Choudhury, P. et al. Targeted deubiquitination rescues distinct trafficking-deficient ion channelopathies. Nat Methods 17, 1245–1253 (2020). https://doi.org/10.1038/s41592-020-00992-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41592-020-00992-6

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