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:

Small-molecule inhibition of the archetypal UbiB protein COQ8

Abstract

Small-molecule tools have enabled mechanistic investigations and therapeutic targeting of the protein kinase-like (PKL) superfamily. However, such tools are still lacking for many PKL members, including the highly conserved and disease-related UbiB family. Here, we sought to develop and characterize an inhibitor for the archetypal UbiB member COQ8, whose function is essential for coenzyme Q (CoQ) biosynthesis. Guided by crystallography, activity assays and cellular CoQ measurements, we repurposed the 4-anilinoquinoline scaffold to selectively inhibit human COQ8A in cells. Our chemical tool promises to lend mechanistic insights into the activities of these widespread and understudied proteins and to offer potential therapeutic strategies for human diseases connected to their dysfunction.

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: Characterization of in vitro COQ8A inhibitors.
Fig. 2: UNC-CA157 decreases de novo CoQ production in COQ8BKO HAP1 cells.
Fig. 3: Mitochondrial targeting of TPP-UNC-CA157 increases cellular efficacy in HAP1 cells.
Fig. 4: TPP-UNC-CA157 kinome specificity.

Similar content being viewed by others

Data availability

LC–MS lipid measurement data that support the findings of this study were deposited into the MassIVE data repository under accession number MSV000090082. Structural data generated in this study have been deposited in the PDB with accession codes 7UDP and 7UDQ. Structural data that were used to support the findings in this study are available in the PDB with accession codes 5I35 and 4PED. Source data are provided with this paper.

References

  1. Manning, G., Whyte, D. B., Martinez, R., Hunter, T. & Sudarsanam, S. The protein kinase complement of the human genome. Science 298, 1912–1934 (2002).

    Article  CAS  Google Scholar 

  2. Dar, A. C. & Shokat, K. M. The evolution of protein kinase inhibitors from antagonists to agonists of cellular signaling. Annu. Rev. Biochem. 80, 769–795 (2011).

    Article  CAS  Google Scholar 

  3. Cohen, P., Cross, D. & Jänne, P. A. Kinase drug discovery 20 years after imatinib: progress and future directions. Nat. Rev. Drug Discov. 20, 551–569 (2021).

    Article  CAS  Google Scholar 

  4. Attwood, M. M., Fabbro, D., Sokolov, A. V., Knapp, S. & Schiöth, H. B. Trends in kinase drug discovery: targets, indications and inhibitor design. Nat. Rev. Drug Discov. 20, 839–861 (2021).

    Article  CAS  Google Scholar 

  5. Kung, J. E. & Jura, N. Prospects for pharmacological targeting of pseudokinases. Nat. Rev. Drug Discov. 18, 501–526 (2019).

    CAS  Google Scholar 

  6. Oprea, T. I. et al. Unexplored therapeutic opportunities in the human genome. Nat. Rev. Drug Discov. 17, 317–332 (2018).

    Article  CAS  Google Scholar 

  7. Leonard, C. J., Aravind, L. & Koonin, E. V. Novel families of putative protein kinases in bacteria and archaea: evolution of the “eukaryotic” protein kinase superfamily. Genome Res. 8, 1038–1047 (1998).

    Article  CAS  Google Scholar 

  8. Pagliarini, D. J. et al. A mitochondrial protein compendium elucidates complex I disease biology. Cell 134, 112–123 (2008).

    Article  CAS  Google Scholar 

  9. Lundquist, P. K., Davis, J. I. & van Wijk, K. J. ABC1K atypical kinases in plants: filling the organellar kinase void. Trends Plant Sci. 17, 546–555 (2012).

    Article  CAS  Google Scholar 

  10. Traschütz, A. et al. Clinico‐genetic, imaging and molecular delineation of COQ8A‐ataxia: a multicenter study of 59 patients. Ann. Neurol. 88, 251–263 (2020).

    Article  Google Scholar 

  11. Stefely, J. A. et al. Cerebellar ataxia and coenzyme Q deficiency through loss of unorthodox kinase activity. Mol. Cell 63, 608–620 (2016).

    Article  CAS  Google Scholar 

  12. Ashraf, S. et al. ADCK4 mutations promote steroid-resistant nephrotic syndrome through CoQ10 biosynthesis disruption. J. Clin. Invest. 123, 5179–5189 (2013).

    Article  CAS  Google Scholar 

  13. Poon, W. W. et al. Identification of Escherichia coli ubiB, a gene required for the first monooxygenase step in ubiquinone biosynthesis. J. Bacteriol. 182, 5139–5146 (2000).

    Article  CAS  Google Scholar 

  14. Do, T. Q., Hsu, A. Y., Jonassen, T., Lee, P. T. & Clarke, C. F. A defect in coenzyme Q biosynthesis is responsible for the respiratory deficiency in Saccharomyces cerevisiae abc1 mutants. J. Biol. Chem. 276, 18161–18168 (2001).

    Article  CAS  Google Scholar 

  15. Stefely, J. A. & Pagliarini, D. J. Biochemistry of mitochondrial coenzyme Q biosynthesis. Trends Biochem. Sci. 42, 824–843 (2017).

    Article  CAS  Google Scholar 

  16. Stefely, J. A. et al. Mitochondrial ADCK3 employs an atypical protein kinase-like fold to enable coenzyme Q biosynthesis. Mol. Cell 57, 83–94 (2015).

    Article  CAS  Google Scholar 

  17. Reidenbach, A. G. et al. Conserved lipid and small-molecule modulation of COQ8 reveals regulation of the ancient kinase-like UbiB family. Cell Chem. Biol. 25, 154–165 (2018).

    Article  CAS  Google Scholar 

  18. Asquith, C. R. M., Murray, N. H. & Pagliarini, D. J. ADCK3/COQ8A: the choice target of the UbiB protein kinase-like family. Nat. Rev. Drug Discov. 18, 815 (2019).

    Article  CAS  Google Scholar 

  19. Murray, N. H. et al. 2-Propylphenol allosterically modulates COQ8A to enhance ATPase activity. ACS Chem. Biol. 17, 2031–2038 (2022).

    Article  CAS  Google Scholar 

  20. Asquith, C. R. M. et al. SGC-GAK-1: a chemical probe for cyclin G associated kinase (GAK). J. Med. Chem. 62, 2830–2836 (2019).

    Article  CAS  Google Scholar 

  21. Wells, C. I. et al. The kinase chemogenomic set (KCGS): an open science resource for kinase vulnerability identification. Int. J. Mol. Sci. 22, 566 (2021).

    Article  CAS  Google Scholar 

  22. Robers, M. B. et al. Target engagement and drug residence time can be observed in living cells with BRET. Nat. Commun. 6, 10091 (2015).

    Article  CAS  Google Scholar 

  23. Vasta, J. D. et al. Quantitative, wide-spectrum kinase profiling in live cells for assessing the effect of cellular ATP on target engagement. Cell Chem. Biol. 25, 206–214 (2018).

    Article  CAS  Google Scholar 

  24. Lagier-Tourenne, C. et al. ADCK3, an ancestral kinase, is mutated in a form of recessive ataxia associated with coenzyme Q10 deficiency. Am. J. Hum. Genet. 82, 661–672 (2008).

    Article  CAS  Google Scholar 

  25. Fonseca, L. V. et al. Mutations in COQ8B (ADCK4) found in patients with steroid-resistant nephrotic syndrome alter COQ8B function. Hum. Mutat. 39, 406–414 (2017).

    Article  Google Scholar 

  26. Rensvold, J. W. et al. Defining mitochondrial protein functions through deep multiomic profiling. Nature 606, 382–388 (2022).

    Article  CAS  Google Scholar 

  27. Zielonka, J. et al. Mitochondria-targeted triphenylphosphonium-based compounds: syntheses, mechanisms of action, and therapeutic and diagnostic applications. Chem. Rev. 117, 10043–10120 (2017).

    Article  CAS  Google Scholar 

  28. Smith, R. A. J., Porteous, C. M., Gane, A. M. & Murphy, M. P. Delivery of bioactive molecules to mitochondria in vivo. Proc. Natl Acad. Sci. USA 100, 5407–5412 (2003).

    Article  CAS  Google Scholar 

  29. Trionnaire, S. L. et al. The synthesis and functional evaluation of a mitochondria-targeted hydrogen sulfide donor, (10-oxo-10-(4-(3-thioxo-3 H -1,2-dithiol-5-yl)phenoxy)decyl)triphenylphosphonium bromide (AP39). MedChemComm 5, 728–736 (2014).

    Article  Google Scholar 

  30. Trnka, J., Elkalaf, M. & Anděl, M. Lipophilic triphenylphosphonium cations inhibit mitochondrial electron transport chain and induce mitochondrial proton leak. PLoS ONE 10, e0121837 (2015).

    Article  Google Scholar 

  31. Reily, C. et al. Mitochondrially targeted compounds and their impact on cellular bioenergetics. Redox Biol. 1, 86–93 (2013).

    Article  CAS  Google Scholar 

  32. Duncan, J. S. et al. Dynamic reprogramming of the kinome in response to targeted MEK inhibition in triple-negative breast cancer. Cell 149, 307–321 (2012).

    Article  CAS  Google Scholar 

  33. Asquith, C. R. M. et al. Identification and optimization of 4-anilinoquinolines as inhibitors of cyclin G associated kinase. ChemMedChem 13, 48–66 (2018).

    Article  CAS  Google Scholar 

  34. Asquith, C. R. M. et al. Design of a cyclin G associated kinase (GAK)/epidermal growth factor receptor (EGFR) inhibitor set to interrogate the relationship of EGFR and GAK in chordoma. J. Med. Chem. 62, 4772–4778 (2019).

    Article  CAS  Google Scholar 

  35. Asquith, C. R. M. et al. Identification of 4-anilinoquin(az)oline as a cell-active protein kinase novel 3 (PKN3) inhibitor chemotype. ChemMedChem 17, e202200161 (2022).

  36. Asquith, C. R. M. et al. Design and analysis of the 4‐anilinoquin(az)oline kinase inhibition profiles of GAK/SLK/STK10 using quantitative structure–activity relationships. ChemMedChem 15, 26–49 (2020).

    Article  CAS  Google Scholar 

  37. Serafim, R. A. M., Elkins, J. M., Zuercher, W. J., Laufer, S. A. & Gehringer, M. Chemical probes for understudied kinases: challenges and opportunities. J. Med. Chem. 65, 1132–1170 (2022).

    Article  CAS  Google Scholar 

  38. Larsen, P. L. & Clarke, C. F. Extension of life-span in Caenorhabditis elegans by a diet lacking coenzyme Q. Science 295, 120–123 (2002).

    Article  CAS  Google Scholar 

  39. Liu, X. et al. Evolutionary conservation of the Clk-1-dependent mechanism of longevity: loss of Mclk1 increases cellular fitness and lifespan in mice. Gene Dev. 19, 2424–2434 (2005).

    Article  CAS  Google Scholar 

  40. Doll, S. et al. FSP1 is a glutathione-independent ferroptosis suppressor. Nature 575, 693–698 (2019).

    Article  CAS  Google Scholar 

  41. Bersuker, K. et al. The CoQ oxidoreductase FSP1 acts parallel to GPX4 to inhibit ferroptosis. Nature 575, 688–692 (2019).

    Article  CAS  Google Scholar 

  42. Forsman, U., Sjöberg, M., Turunen, M. & Sindelar, P. J. 4-Nitrobenzoate inhibits coenzyme Q biosynthesis in mammalian cell cultures. Nat. Chem. Biol. 6, 515–517 (2010).

    Article  CAS  Google Scholar 

  43. Wang, Y. et al. The anti-neurodegeneration drug clioquinol inhibits the aging-associated protein CLK-1. J. Biol. Chem. 284, 314–323 (2009).

    Article  CAS  Google Scholar 

  44. Tsuganezawa, K. et al. Identification of small molecule inhibitors of human COQ7. Bioorg. Med Chem. 28, 115182 (2020).

    Article  CAS  Google Scholar 

  45. Nara, T. et al. The ubiquinone synthesis pathway is a promising drug target for Chagas disease. PLoS ONE 16, e0243855 (2021).

    Article  CAS  Google Scholar 

  46. Fernández-del-Río, L. et al. Regulation of hepatic coenzyme Q biosynthesis by dietary omega-3 polyunsaturated fatty acids. Redox Biol. 46, 102061 (2021).

  47. Ward, R. A. et al. Challenges and opportunities in cancer drug resistance. Chem. Rev. 121, 3297–3351 (2021).

    Article  CAS  Google Scholar 

  48. Kelso, G. F. et al. Selective targeting of a redox-active ubiquinone to mitochondria within cells. J. Biol. Chem. 276, 4588–4596 (2001).

    Article  CAS  Google Scholar 

  49. Kulkarni, C. A. et al. A novel triphenylphosphonium carrier to target mitochondria without uncoupling oxidative phosphorylation. J. Med. Chem. 64, 662–676 (2021).

    Article  CAS  Google Scholar 

  50. Kemmerer, Z. A. et al. UbiB proteins regulate cellular CoQ distribution in Saccharomyces cerevisiae. Nat. Commun. 12, 4769 (2021).

    Article  CAS  Google Scholar 

  51. Asquith, C. R. M. et al. Targeting an EGFR water network with 4‐anilinoquin(az)oline inhibitors for chordoma. ChemMedChem 14, 1693–1700 (2019).

    Article  CAS  Google Scholar 

  52. Asquith, C. R. M., Treiber, D. K. & Zuercher, W. J. Utilizing comprehensive and mini-kinome panels to optimize the selectivity of quinoline inhibitors for cyclin G associated kinase (GAK). Bioorg. Med. Chem. Lett. 29, 1727–1731 (2019).

    Article  CAS  Google Scholar 

  53. Stepanov, S. et al. JBluIce-EPICS control system for macromolecular crystallography. Acta Crystallogr. D Biol. Crystallogr. 67, 176–188 (2011).

    Article  CAS  Google Scholar 

  54. Vonrhein, C. et al. Data processing and analysis with the autoPROC toolbox. Acta Crystallogr. D Biol. Crystallogr. 67, 293–302 (2011).

    Article  CAS  Google Scholar 

  55. Kabsch, W. XDS. Acta Crystallogr. D Biol. Crystallogr. 66, 125–132 (2010).

    Article  CAS  Google Scholar 

  56. McCoy, A. J. et al. Phaser crystallographic software. J. Appl. Crystallogr. 40, 658–674 (2007).

    Article  CAS  Google Scholar 

  57. Moriarty, N. W., Grosse-Kunstleve, R. W. & Adams, P. D. electronic Ligand Builder and Optimization Workbench (eLBOW): a tool for ligand coordinate and restraint generation. Acta Crystallogr. D Biol. Crystallogr. 65, 1074–1080 (2009).

    Article  CAS  Google Scholar 

  58. 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  Google Scholar 

  59. Afonine, P. V. et al. Towards automated crystallographic structure refinement with phenix.refine. Acta Crystallogr. D Biol. Crystallogr. 68, 352–367 (2012).

    Article  CAS  Google Scholar 

  60. Laskowski, R. A. & Swindells, M. B. LigPlot+: multiple ligand–protein interaction diagrams for drug discovery. J. Chem. Inf. Model. 51, 2778–2786 (2011).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank members of the Pagliarini lab for helpful discussions throughout this project. This work was supported by NIH awards R35GM131795 (D.J.P.) and T32GM008505 (N.H.M), funds from the BJC Investigator Program (D.J.P.) and NSF DGE-1747503 (N.H.M.). We would like to thank K. Overmyer, B. Paulson, E. Trujillo and J. Coon for assistance with LC–MS method development. We thank G. Hicks for graphic design assistance. This study made use of the Washington University in St. Louis Genome Engineering and iPSC Center for instrument use and the Washington University in St. Louis Center for Drug Discovery for synthesis services. The SGC is a registered charity (number 1097737) that receives funds from AbbVie, Bayer Pharma AG, Boehringer Ingelheim, Canada Foundation for Innovation, Eshelman Institute for Innovation, Genome Canada, Innovative Medicines Initiative (EU/EFPIA; ULTRA-DD grant number 115766), Janssen, Merck KGaA Darmstadt Germany, MSD, Novartis Pharma AG, Ontario Ministry of Economic Development and Innovation, Pfizer, São Paulo Research Foundation-FAPESP, Takeda and Wellcome (106169/ZZ14/Z). The NIH is acknowledged for support (1U24DK11604-01). We thank Biocenter Finland/DDCB for financial support and the CSC-IT Center for Science, Ltd. (Finland), for allocation of computational resources. We also thank B. Ehrmann and D.E. Weatherspoon for LC–MS/HRMS support provided by the Mass Spectrometry Core Laboratory at the University of North Carolina at Chapel Hill. The core is supported by the National Science Foundation under grant number CHE-1726291. We thank T. Pantsar (University of Eastern Finland) for useful discussions. This research used resources of the Advanced Photon Source, a US Department of Energy (DOE) Office of Science User Facility operated for the DOE Office of Science by Argonne National Laboratory under contract number DE-AC02-06CH11357. GM/CA@APS has been funded by the National Cancer Institute (ACB-12002) and the National Institute of General Medical Sciences (AGM-12006, P30GM138396). The Eiger 16M detector at GM/CA-XSD was funded by NIH grant S10 OD012289. We thank C. Ogata for beamline support. Use of the LS-CAT Sector 21 was supported by the Michigan Economic Development Corporation and the Michigan Technology Tri-Corridor (grant 085P1000817). The Collaborative Crystallography Core in the Department of Biochemistry, Univeristy of Wisconsin–Madison, received support from the Department of Biochemistry endowment.

Author information

Authors and Affiliations

Authors

Contributions

N.H.M. and D.J.P. wrote the manuscript. N.H.M., C.R.M.A. and D.J.P. conceived the overall project and its design. C.R.M.A. performed compound synthesis and contributed to inhibitor development. Z.F. performed and analyzed MS experiments. N.P. contributed reagents (cloning) and developed DSF methods. R.W.S. and C.A.B. performed crystallization trials and solved the crystal structure. J.D.V., C.A.Z., C.R.C. and M.B.R. performed NanoBRET analyses. M.P.E. and G.L.J. developed and performed the MIBS–MS analyses. All authors reviewed and edited the manuscript.

Corresponding author

Correspondence to David J. Pagliarini.

Ethics declarations

Competing interests

J.D.V., C.R.C., C.A.Z. and M.B.R. are employed by Promega Corporation. The remaining authors declare no competing interests.

Peer review

Peer review information

Nature Chemical Biology thanks Markus Seeliger and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

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 COQ8A inhibition in vitro.

a, Inhibition of COQ8AN∆250 ATPase activity by the top candidate inhibitors (n = 3 independent samples from one experiment, mean ± SD) (IC50 data are available in the supporting information). b, Top candidate inhibitor binding curves, as determined by DSF (n = 3 independent samples from one experiment, mean ± SD) (Kd,app data are available in the supporting information).

Source data

Extended Data Fig. 2 De novo CoQ10 biosynthesis measurements.

a, Schematic outlining the experimental plan to measure de novo CoQ production. b, De novo production of 13C6-PPHB10 in WT, COQ8AKO, COQ8BKO, and COQ8A/BDKO HAP1 cells after treatment with 10 µM 13C6-4-HB and either DMSO or 20 µM UNC-CA157 (grey = DMSO, red=UNC-CA157, n = 3 independent samples from one experiment, mean ± SD) (two sided students t-test, no adjustment for multiple comparisons). c, Unlabeled PPHB10 levels in WT, COQ8AKO, COQ8BKO, and COQ8A/BDKO HAP1 cells after treatment with 10 µM 13C6-4-HB and either DMSO or 20 µM UNC-CA157 (grey = DMSO, red=UNC-CA157, n = 3 independent samples from one experiment, mean ± SD) (two sided students t-test, no adjustment for multiple comparisons). d, Unlabeled CoQ10 levels in WT, COQ8AKO, COQ8BKO, and COQ8A/BDKO HAP1 cells after treatment with 10 µM 13C6-4-HB and either DMSO or 20 µM UNC-CA157 (grey = DMSO, red=UNC-CA157, n = 3 independent samples from one experiment, mean ± SD) (two sided students t-test, no adjustment for multiple comparisons).

Source data

Extended Data Fig. 3 F495 is required for potent inhibition.

a-c, Protein-ligand interaction networks for three independent complexes of COQ8AN∆254 and UNC-CA157 generated in LigPlot + (PDB: 7UDQ and 7UDP) (hydrogen bond = dashed line; hydrophobic contact = arc with spokes). d, Protein-ligand interaction network for COQ8AN∆254 R611K and AMPPNP generated in LigPlot + (PDB: 5I35) (hydrogen bond = dashed line; hydrophobic contact = arc with spokes). e, SDS-PAGE analysis of COQ8AN∆250 purifications. f, COQ8AN∆250 WT and binding pocket mutant thermal stabilization by UNC-CA157, as determined by DSF (n = 3 independent samples from one experiment, mean ± SD) (two sided students t-test, no adjustment for multiple comparisons, ***p = 1.2 × 10−10 F336A, ***p = 4.5 × 10−5 A356G, ***p = 2.4 × 10−9 K358A, ***p = 4.7 × 10−11 K358E, ***p = 1.7 × 10−6 L443A, *p = 0.0264 T445A, ***p = 1.8 × 10−10 L447A, ***p = 1.4 × 10−9 V448P, ***p = 9.5 × 10−12 F495A; all six WT-1 and WT-2 data points were used for statistical analysis). g, Stick representation of UNC-CA157 and adjacent F495 residue. h, Conservation of F495 across the UbiB protein family. i, Thermal stabilization of COQ8AN∆250 WT and F495L as well as Coq8pN∆41 WT and L353F by UNC-CA157 (n = 3 independent samples from one experiment, mean ± SD) (two sided students t-test, no adjustment for multiple comparisons, *p = 0.0126 COQ8A WT vs. F495L, ***p = 5.6 × 10−7 Coq8p WT vs. L353F). j, Inhibition of COQ8AN∆250 WT and F495L ATPase activity by UNC-CA157 (n = 3 independent samples from one experiment, mean ± SD).

Source data

Extended Data Fig. 4 Results from de novo CoQ production with TPP-UNC-CA157 and inhibitor toxicity analysis.

a, Unlabeled CoQ10 and PPHB10 levels in WT, COQ8AKO, COQ8BKO, and COQ8A/BDKO HAP1 cells after treatment with 10 µM 13C6-4-HB and either DMSO or 17.6 µM TPP-UNC-CA157 (n = 3 independent samples from one experiment, mean ± SD) (two sided students t-test, no adjustment for multiple comparisons, *p = 0.0228 WT PPHB10, ***p = 0.0003 COQ8AKO PPHB10). b-d, 13C6-CoQ10 levels in WT HAP1 cells after treatment with 10 µM 13C6-4-HB and indicated concentrations of UNC-CA157, TPP-UNC-CA157 or Alkyl-TPP (n = 3 independent samples from one experiment, mean ± SD) (two sided students t-test, no adjustment for multiple comparisons, *p = 0.0482 DMSO vs. 43.9 µM UNC-CA157, *p = 0.0118 DMSO vs. 22.0 µM TPP-UNC-CA157, **p = 0.0030 DMSO vs. 43.9 µM TPP-UNC-CA157, **p = 0.0026 DMSO vs. 87.8 µM TPP-UNC-CA157). e, Growth analysis of WT HAP1 cells in either 10 mM glucose or 10 mM galactose with indicated concentrations of TPP-UNC-CA157 (n = 3 biologically independent wells of cells in one experiment, mean ± SEM).

Source data

Supplementary information

Supplementary Information

Supplementary Tables 1–6, Figs. 1–4 and Note TPP-UNC-CA157 synthesis.

Reporting Summary

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 2

Statistical source data.

Source Data Extended Data Fig. 3

Statistical source data and uncropped gel.

Source Data Extended Data Fig. 4

Statistical source data.

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

Murray, N.H., Asquith, C.R.M., Fang, Z. et al. Small-molecule inhibition of the archetypal UbiB protein COQ8. Nat Chem Biol 19, 230–238 (2023). https://doi.org/10.1038/s41589-022-01168-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41589-022-01168-3

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