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:

Enzymatic assembly of the salinosporamide γ-lactam-β-lactone anticancer warhead

Abstract

The marine microbial natural product salinosporamide A (marizomib) is a potent proteasome inhibitor currently in clinical trials for the treatment of brain cancer. Salinosporamide A is characterized by a complex and densely functionalized γ-lactam-β-lactone bicyclic warhead, the assembly of which has long remained a biosynthetic mystery. Here, we report an enzymatic route to the salinosporamide core catalyzed by a standalone ketosynthase (KS), SalC. Chemoenzymatic synthesis of carrier protein-tethered substrates, as well as intact proteomics, allowed us to probe the reactivity of SalC and understand its role as an intramolecular aldolase/β-lactone synthase with roles in both transacylation and bond-forming reactions. Additionally, we present the 2.85-Å SalC crystal structure that, combined with site-directed mutagenesis, allowed us to propose a bicyclization reaction mechanism. This work challenges our current understanding of the role of KS enzymes and establishes a basis for future efforts toward streamlined production of a clinically relevant chemotherapeutic.

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: Microbial natural products assembled by terminal cyclization reactions.
Fig. 2: SalC is involved in late-stage salinosporamide A biosynthesis.
Fig. 3: SalC is a γ-lactam-β-lactone bicyclase.
Fig. 4: Transacylation of SalC by acylated SalB.
Fig. 5: SalC structure and active site mutagenesis.
Fig. 6: Abbreviated key bicyclization steps of the proposed SalC mechanism with 15.

Similar content being viewed by others

Data availability

Strains and plasmids used in this study are described in Supplementary Table 1. All oligonucleotides (Integrated DNA Technology) are shown in Supplementary Table 2. The salinosporamide (sal) BGC from S. tropica CNB440 is available in the MIBiG database (accession BGC0001041). Other salinosporamide BGCs and the cinnabaramide BGC used for alignments are available in the IMG JGI database. Functional KS and non-elongating KS protein sequences used for alignments can be found in the Supplementary material and through MIBiG. Atomic coordinates and structure factors for the reported crystal structures in this work have been deposited to the PDB under accession number 7S2X(native SalC). Additionally, the following PBD datasets were used for SalC structural comparison: 2HG4, 4WKY, 2QO3 and 4NA2. Source data are provided with this paper. Other relevant data supporting the findings of this study are available in this published article or its Supplementary files.

References

  1. Roth, P. et al. A phase III trial of marizomib in combination with temozolomide-based radiochemotherapy versus temozolomide-based radiochemotherapy alone in patients with newly diagnosed glioblastoma. J. Clin. Oncol. 39, 2004 (2021).

    Article  Google Scholar 

  2. Feling, R. H. et al. Salinosporamide A: a highly cytotoxic proteasome inhibitor from a novel microbial source, a marine bacterium of the new genus Salinospora. Angew. Chem. Int. Ed. Engl. 42, 355–357 (2003).

    Article  CAS  PubMed  Google Scholar 

  3. Williams, P. G. et al. New cytotoxic salinosporamides from the marine Actinomycete Salinispora tropica. J. Org. Chem. 70, 6196–6203 (2005).

    Article  CAS  PubMed  Google Scholar 

  4. Macherla, V. R. et al. Structure–activity relationship studies of salinosporamide A (NPI-0052), a novel marine derived proteasome inhibitor. J. Med. Chem. 48, 3684–3687 (2005).

    Article  CAS  PubMed  Google Scholar 

  5. McGlinchey, R. P. et al. Engineered biosynthesis of antiprotealide and other unnatural salinosporamide proteasome inhibitors. J. Am. Chem. Soc. 130, 7822–7823 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Nett, M., Gulder, T. A. M., Kale, A. J., Hughes, C. C. & Moore, B. S. Function-oriented biosynthesis of β-lactone proteasome inhibitors in Salinispora tropica. J. Med. Chem. 52, 6163–6167 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Groll, M., Huber, R. & Potts, B. C. M. Crystal structures of salinosporamide A (NPI-0052) and B (NPI-0047) in complex with the 20S proteasome reveal important consequences of β-lactone ring opening and a mechanism for irreversible binding. J. Am. Chem. Soc. 128, 5136–5141 (2006).

    Article  CAS  PubMed  Google Scholar 

  8. Beer, L. L. & Moore, B. S. Biosynthetic convergence of salinosporamides A and B in the marine actinomycete Salinispora tropica. Org. Lett. 9, 845–848 (2007).

    Article  CAS  PubMed  Google Scholar 

  9. Eustáquio, A. S. et al. Biosynthesis of the salinosporamide A polyketide synthase substrate chloroethylmalonyl-coenzyme A from S-adenosyl-l-methionine. Proc. Natl Acad. Sci. USA 106, 12295–12300 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  10. Mahlstedt, S., Fielding, E. N., Moore, B. S. & Walsh, C. T. Prephenate decarboxylases: a new prephenate-utilizing enzyme family that performs non-aromatizing decarboxylation en route to diverse secondary metabolites. Biochemistry 49, 9021–9023 (2010).

    Article  CAS  PubMed  Google Scholar 

  11. Ma, G., Nguyen, H. & Romo, D. Concise total synthesis of (±)-salinosporamide A, (±)-cinnabaramide A, and derivatives via a bis-cyclization process: implications for a biosynthetic pathway? Org. Lett. 9, 2143–2146 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Nguyen, H., Ma, G., Gladysheva, T., Fremgen, T. & Romo, D. Bioinspired total synthesis and human proteasome inhibitory activity of (–)-salinosporamide A, (–)-homosalinosporamide A, and derivatives obtained via organonucleophile promoted bis-cyclizations. J. Org. Chem. 76, 2–12 (2011).

    Article  CAS  PubMed  Google Scholar 

  13. Fischbach, M. A. & Walsh, C. T. Assembly-line enzymology for polyketide and nonribosomal peptide antibiotics: logic, machinery, and mechanisms. Chem. Rev. 106, 3468–3496 (2006).

    Article  CAS  PubMed  Google Scholar 

  14. Schaffer, J. E., Reck, M. R., Prasad, N. K. & Wencewicz, T. A. β-Lactone formation during product release from a nonribosomal peptide synthetase. Nat. Chem. Biol. 13, 737–744 (2017).

    Article  CAS  PubMed  Google Scholar 

  15. Gulder, T. A. M. & Moore, B. S. Salinosporamide natural products: potent 20S proteasome inhibitors as promising cancer chemotherapeutics. Angew. Chem. Int. Ed. Engl. 49, 9346–9367 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Gao, X. et al. Cyclization of fungal nonribosomal peptides by a terminal condensation-like domain. Nat. Chem. Biol. 8, 823–830 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Feng, K.-N. et al. A hydrolase-catalyzed cyclization forms the fused bicyclic β-lactone in vibralactone. Angew. Chem. Int. Ed. Engl. 59, 7209–7213 (2020).

    Article  CAS  PubMed  Google Scholar 

  18. Gaudelli, N. M., Long, D. H. & Townsend, C. A. β-Lactam formation by a non-ribosomal peptide synthetase during antibiotic biosynthesis. Nature 520, 383–387 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Yun, C.-S. et al. Unique features of the ketosynthase domain in a non-ribosomal peptide synthetase–polyketide synthase hybrid enzyme, tenuazonic acid synthetase 1. J. Biol. Chem. 295, 11602–11612 (2020).

  20. Yun, C.-S., Motoyama, T. & Osada, H. Biosynthesis of the mycotoxin tenuazonic acid by a fungal NRPS–PKS hybrid enzyme. Nat. Commun. 6, 8758 (2015).

    Article  CAS  PubMed  Google Scholar 

  21. Mo, X. & Gulder, T. A. M. Biosynthetic strategies for tetramic acid formation. Nat. Prod. Rep. 38, 1555–1566 (2021).

  22. Rachid, S. et al. Mining the cinnabaramide biosynthetic pathway to generate novel proteasome inhibitors. ChemBioChem 12, 922–931 (2011).

    Article  CAS  PubMed  Google Scholar 

  23. Altschul, S. F., Gish, W., Miller, W., Myers, E. W. & Lipman, D. J. Basic local alignment search tool. J. Mol. Biol. 215, 403–410 (1990).

    Article  CAS  PubMed  Google Scholar 

  24. Ziemert, N. et al. The Natural Product Domain Seeker NaPDoS: a phylogeny based bioinformatic tool to classify secondary metabolite gene diversity. PLoS ONE 7, e34064 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Robbins, T., Kapilivsky, J., Cane, D. E. & Khosla, C. Roles of conserved active site residues in the ketosynthase domain of an assembly line polyketide synthase. Biochemistry 55, 4476–4484 (2016).

    Article  CAS  PubMed  Google Scholar 

  26. Helfrich, E. J. N. & Piel, J. Biosynthesis of polyketides by trans-AT polyketide synthases. Nat. Prod. Rep. 33, 231–316 (2016).

    Article  CAS  PubMed  Google Scholar 

  27. Masschelein, J. et al. A dual transacylation mechanism for polyketide synthase chain release in enacyloxin antibiotic biosynthesis. Nat. Chem. 11, 906–912 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Satou, R. et al. Structural basis for cyclization specificity of two Azotobacter type III polyketide synthases. J. Biol. Chem. 288, 34146–34157 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Boddy, C. N., Schneider, T. L., Hotta, K., Walsh, C. T. & Khosla, C. Epothilone C macrolactonization and hydrolysis are catalyzed by the isolated thioesterase domain of epothilone polyketide synthase. J. Am. Chem. Soc. 125, 3428–3429 (2003).

    Article  CAS  PubMed  Google Scholar 

  30. Reed, K. A. et al. Salinosporamides D–J from the marine actinomycete Salinispora tropica, bromosalinosporamide, and thioester derivatives are potent inhibitors of the 20S proteasome. J. Nat. Prod. 70, 269–276 (2007).

    Article  CAS  PubMed  Google Scholar 

  31. Manam, R. R. et al. Antiprotealide is a natural product. J. Nat. Prod. 72, 295–297 (2009).

    Article  CAS  PubMed  Google Scholar 

  32. Ling, T., Macherla, V. R., Manam, R. R., McArthur, K. A. & Potts, B. C. M. Enantioselective total synthesis of (–)-salinosporamide A (NPI-0052). Org. Lett. 9, 2289–2292 (2007).

    Article  CAS  PubMed  Google Scholar 

  33. Agarwal, V. et al. Chemoenzymatic synthesis of acyl coenzyme A substrates enables in situ labeling of small molecules and proteins. Org. Lett. 17, 4452–4455 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Dorrestein, P. C. et al. Facile detection of acyl and peptidyl intermediates on thiotemplate carrier domains via phosphopantetheinyl elimination reactions during tandem mass spectrometry. Biochemistry 45, 12756–12766 (2006).

    Article  CAS  PubMed  Google Scholar 

  35. Khosla, C., Tang, Y., Chen, A. Y., Schnarr, N. A. & Cane, D. E. Structure and mechanism of the 6-deoxyerythronolide B synthase. Annu. Rev. Biochem. 76, 195–221 (2007).

    Article  CAS  PubMed  Google Scholar 

  36. Denora, N., Potts, B. C. M. & Stella, V. J. A mechanistic and kinetic study of the β-lactone hydrolysis of salinosporamide A (NPI-0052), a novel proteasome inhibitor. J. Pharm. Sci. 96, 2037–2047 (2007).

    Article  CAS  PubMed  Google Scholar 

  37. Tsueng, G. & Lam, K. S. Stabilization effect of resin on the production of potent proteasome inhibitor NPI-0052 during submerged fermentation of Salinispora tropica. J. Antibiot. 60, 469–472 (2007).

    Article  CAS  Google Scholar 

  38. Dutta, S. et al. Structure of a modular polyketide synthase. Nature 510, 512–517 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Holm, L. Using Dali for protein structure comparison. Methods Mol. Biol. 2112, 29–42 (2020).

    Article  CAS  PubMed  Google Scholar 

  40. Tang, Y., Chen, A. Y., Kim, C.-Y., Cane, D. E. & Khosla, C. Structural and mechanistic analysis of protein interactions in module 3 of the 6-deoxyerythronolide B synthase. Chem. Biol. 14, 931–943 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Lohman, J. R. et al. Structural and evolutionary relationships of ‘AT-less’ type I polyketide synthase ketosynthases. Proc. Natl Acad. Sci. USA 112, 12693–12698 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Gay, D. C. et al. A close look at a ketosynthase from a trans-acyltransferase modular polyketide synthase. Structure 22, 444–451 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Davies, C., Heath, R. J., White, S. W. & Rock, C. O. The 1.8 Å crystal structure and active-site architecture of β-ketoacyl-acyl carrier protein synthase III (FabH) from Escherichia coli. Structure 8, 185–195 (2000).

    Article  CAS  PubMed  Google Scholar 

  44. Cromm, P. M. & Crews, C. M. The proteasome in modern drug discovery: second life of a highly valuable drug target. ACS Cent. Sci. 3, 830–838 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Niewerth, D. et al. Antileukemic activity and mechanism of drug resistance to the marine Salinispora tropica proteasome inhibitor salinosporamide A (marizomib). Mol. Pharmacol. 86, 12–19 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  46. Hansen, D. A., Koch, A. A. & Sherman, D. H. Identification of a thioesterase bottleneck in the pikromycin pathway through full-module processing of unnatural pentaketides. J. Am. Chem. Soc. 139, 13450–13455 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Christenson, J. K. et al. β-Lactone synthetase found in the olefin biosynthesis pathway. Biochemistry 56, 348–351 (2017).

    Article  CAS  PubMed  Google Scholar 

  48. Robinson, S. L. et al. Global analysis of adenylate-forming enzymes reveals β-lactone biosynthesis pathway in pathogenic Nocardia. J. Biol. Chem. 295, 14826–14839 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Gilchrist, C. L. M. et al. cblaster: a remote search tool for rapid identification and visualisation of homologous gene clusters. Bioinformatics Adv. 1, vbab016 (2021).

  50. Gilchrist, C. L. M. & Chooi, Y.-H. clinker & clustermap.js: automatic generation of gene cluster comparison figures. Bioinformatics 37, 2473–2475 (2021).

  51. Kieser, T. Practical Streptomyces Genetics (John Innes Foundation, 2000).

  52. MacNeil, D. J. et al. Analysis of Streptomyces avermitilis genes required for avermectin biosynthesis utilizing a novel integration vector. Gene 111, 61–68 (1992).

    Article  CAS  PubMed  Google Scholar 

  53. Flett, F., Mersinias, V. & Smith, C. P. High efficiency intergeneric conjugal transfer of plasmid DNA from Escherichia coli to methyl DNA-restricting Streptomycetes. FEMS Microbiol. Lett. 155, 223–229 (1997).

    Article  CAS  PubMed  Google Scholar 

  54. Wilkinson, C. J. et al. Increasing the efficiency of heterologous promoters in actinomycetes. J. Mol. Microbiol. Biotechnol. 4, 417–426 (2002).

    CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  Google Scholar 

  57. Tang, Y., Kim, C.-Y., Mathews, I. I., Cane, D. E. & Khosla, C. The 2.7-Å crystal structure of a 194-kDa homodimeric fragment of the 6-deoxyerythronolide B synthase. Proc. Natl Acad. Sci. USA 103, 11124–11129 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Roy, A., Kucukural, A. & Zhang, Y. I-TASSER: a unified platform for automated protein structure and function prediction. Nat. Protoc. 5, 725–738 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. 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 

  60. Emsley, P. & Cowtan, K. Coot: model-building tools for molecular graphics. Acta Crystallogr. D Biol. Crystallogr. 60, 2126–2132 (2004).

    Article  PubMed  CAS  Google Scholar 

Download references

Acknowledgements

We are grateful to P.R. Jensen (Scripps Institution of Oceanography) for the S. tropica CNB440 strain, B.M. Dungan (UC San Diego) for assistance with NMR, K.E. Creamer (UC San Diego) for the NaPDoS2.0 analysis and helpful bioinformatics discussions, Y. Su (UC San Diego Molecular Mass Spectrometry Facility) for intact proteomics experiments and J.P. Noel and G. Louie (Salk Institute for Biological Studies) for beamline coordination. Beamline 8.2.2 of the Advanced Light Source, a U.S. DOE Office of Science User Facility under contract number DE-AC02-05CH11231, is supported in part by the ALS-ENABLE program funded by the National Institutes of Health, National Institute of General Medical Sciences, grant P30-GM124169-01. This work was supported by NIH grants R01CA127622 (B.S.M.), R01AI047818 (B.S.M.) and F31HD101307 (K.D.B.), R35GM134910 (D.R.), the Robert A. Welch Foundation (grant number AA-1280 (D.R.)) and the São Paulo Research Foundation (FAPESP; grant number 2011/21358-5 (D.B.B.T.)).

Author information

Authors and Affiliations

Authors

Contributions

K.D.B., V.V.S., T.A.M.G. and B.S.M. designed the study. K.D.B. performed protein expression and purification, enzymatic assays, mutagenesis and data analysis. V.V.S. synthesized and characterized all substrates in this study. P.Y.-T.C., D.B.B.T. and K.D.B. performed all crystallography experiments and subsequent data analysis. T.A.M.G. performed all gene inactivation experiments, preliminary protein expressions and initial substrate synthesis. S.V. and D.R. provided helpful initial synthetic discourse. K.D.B. and B.S.M. wrote the manuscript with input from all coauthors.

Corresponding author

Correspondence to Bradley S. Moore.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Chemical Biology thanks Andrew Gulick, Yi Tang 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 Salinosporamide A hydrolysis and subsequent THF ring formation is accelerated by the presence of SalC.

a, LCMS chromatograms of salinosporamide A (1) hydrolysis assay with and without SalC. * indicates compound retains chloride and no THF ring formation has occurred as evidenced by characteristic isotope pattern. b, Structures of compounds putatively identified by LCMS in a.

Extended Data Fig. 2

Naturally produced analogs of salinosporamide A that served as inspiration for simplisporamide.

Extended Data Fig. 3 SalC activity assay with diffusible substrates.

a, Reaction scheme depicting chemoenzymatic synthesis and subsequent SalB-PCP acylation assay to generate (15). b, LCMS chromatograms (BPCs and EICs m/z 266.18) of SalC activity assay with diffusible substrates shown in part (a) including pantetheine-activated (13), CoaA product-activated (19), CoaD product-activated (20), CoA activated (14), and carrier protein-activated substrate (15). Substrates were activated via in vitro CoA enzyme biosynthesis. All SalC activity assays contained apo-SalB-PCP as well.

Extended Data Fig. 4 Proteasome inhibitory activity of SalC assay product using purified human 20S proteasome.

a, Proteasome activity determined by reading fluorescence (AFUs) of the cleaved substrate (Suc-LLVY-AMC) at 355 nm (excitation) and 460 nm (emission) every five minutes after substrate was added for 30 min in the presence of various inhibitors. Blank (no proteasome added) = black, control (no inhibitor) = gray, epoxomicin (0.5 μM) = green, SalC reaction product = red, no SalC control = dark blue, no substrate control = orange, salinosporamide A (0.5 μM) = light blue. Samples run in duplicate, all data points shown. b, Magnification of y-axis of plot from a to examine successful inhibition of the 20S proteasome by epoxomicin, SalC reaction product, and salinosporamide A. c, Percent proteasome inhibition at 30 min, relative to control (no inhibitor, 0% inhibition). See Supplementary Fig. 14 for corresponding LCMS traces of extracts used in these assays.

Source data

Extended Data Fig. 5 Acylation of SalC with diffusible substrates.

a, Reaction scheme depicting chemoenzymatic synthesis to generate (22) b, UV chromatograms (215 nm) of intact protein LCMS for transacylation assay with diffusible substrates. Transacylation assay utilized linear mechanistic probe (21) activated in different ways (CoA-precursor-activated (23, 24), CoA-activated (25), and SalB-PCP-tethered, all generated in situ) and SalC. Transacylation assay with column purified 22 shown for comparison.

Extended Data Fig. 6 SalC overlay with a trans-AT KS.

SalC structure aligned with closest Dali server homolog, the trans-AT KS OzmN KS2 (PDB ID: 4WKY) from the hybrid NRPS/PKS oxazolomycin pathway, RMSD 0.842 Å. Overall structure of SalC dimer (colors shown as previous, KS monomers in brown and green, flanking subdomains in yellow and teal) with OzmN KS2 (gray).

Extended Data Fig. 7 SalC overlay with functional type I KS.

SalC KS aligned with DEBS KS3 (PDB: 2QO3), RMSD 1.225 Å. a, Overall structure of SalC dimer (colors shown as previous, KS monomers in brown and green, flanking subdomains in yellow and teal) with DEBS KS3 (gray). b, Active site overlay of SalC (green) and DEBS KS3 (gray).

Extended Data Fig. 8 Tyr284 is conserved in SalC homologs.

Condensed alignment showing conservation of Tyr284 in all SalC homologs from Salinispora and Streptomyces cinnabarigriseus JS360 (CinC) but not in canonical elongating KSs. All SalC homologs from Salinispora strains found in JBI IMG database. For functional KS sequences refer to Supplementary Figure 5.

Extended Data Fig. 9 SalC structure overlaid with bacillaene PKS (bae) KS2 bound to its natural intermediate.

SalC is shown in green and BaeKS2 is shown in grey, bae intermediate in gold. PDB ID: 4NA2, RMSD 1.071 Å.

Extended Data Fig. 10 Proposed active site mechanism of SalC.

Catalysis is initiated by deprotonation of Cys180 followed by transacylation of the SalB-PCP tethered substrate through a tetrahedral intermediate (not shown). Lys348 deprotonates His353, and hydrogen bonding of the thioamide carbonyl to Tyr284 facilitates deprotonation of the thioester α-proton by His353. An intramolecular aldol reaction forms the γ-lactam; the oxyanion is presumably stabilized by dipole interactions with backbone amides, as is hypothesized for KSs25. Subsequent β-lactonization through a tetrahedral intermediate leads to release of simplisporamide from SalC. Finally, Cys180 is reprotonated by His353.

Supplementary information

Supplementary Information

Supplementary Figs. 1–30, Tables 1–4, Notes Figs. 1–29 and Notes Table 1.

Reporting Summary

Source data

Source Data Extended Data Fig. 4

Unprocessed Arbitrary Fluorescence Units (AFUs) for proteasome inhibition assay.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Bauman, K.D., Shende, V.V., Chen, P.YT. et al. Enzymatic assembly of the salinosporamide γ-lactam-β-lactone anticancer warhead. Nat Chem Biol 18, 538–546 (2022). https://doi.org/10.1038/s41589-022-00993-w

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41589-022-00993-w

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