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:

Depletion of creatine phosphagen energetics with a covalent creatine kinase inhibitor

Abstract

Creatine kinases (CKs) provide local ATP production in periods of elevated energetic demand, such as during rapid anabolism and growth. Thus, creatine energetics has emerged as a major metabolic liability in many rapidly proliferating cancers. Whether CKs can be targeted therapeutically is unknown because no potent or selective CK inhibitors have been developed. Here we leverage an active site cysteine present in all CK isoforms to develop a selective covalent inhibitor of creatine phosphagen energetics, CKi. Using deep chemoproteomics, we discover that CKi selectively engages the active site cysteine of CKs in cells. A co-crystal structure of CKi with creatine kinase B indicates active site inhibition that prevents bidirectional phosphotransfer. In cells, CKi and its analogs rapidly and selectively deplete creatine phosphate, and drive toxicity selectively in CK-dependent acute myeloid leukemia. Finally, we use CKi to uncover an essential role for CKs in the regulation of proinflammatory cytokine production in macrophages.

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: Ultra-deep chemoproteomics identifies a potent and selective small molecule that engages the CK active site cysteine.
Fig. 2: Structural basis for CK inhibition by CKi.
Fig. 3: CKi and MitoCKi deplete creatine phosphagen energetics in cells.
Fig. 4: CKi and MitoCKi exploit creatine-energetics dependency in AML to drive cytotoxicity.
Fig. 5: Macrophages and monocytes require the creatine phosphagen system to support proinflammatory cytokine production.

Similar content being viewed by others

Data availability

The datasets generated during and/or analyzed during the current study are available as Excel spreadsheets in Supplementary Tables 110. Source data are provided with this paper. All additional data is available upon request from the corresponding authors.

References

  1. Kazak, L. & Cohen, P. Creatine metabolism: energy homeostasis, immunity and cancer biology. Nat. Rev. Endocrinol. 16, 421–436 (2020).

    PubMed  Google Scholar 

  2. Fenouille, N. et al. The creatine kinase pathway is a metabolic vulnerability in EVI1-positive acute myeloid leukemia. Nat. Med. 23, 301–313 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Loo, J. M. et al. Extracellular metabolic energetics can promote cancer progression. Cell 160, 393–406 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  4. Maguire, O. A. et al. Creatine-mediated crosstalk between adipocytes and cancer cells regulates obesity-driven breast cancer. Cell Metab. 33, 499–512.e496 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Zhang, L. et al. Creatine promotes cancer metastasis through activation of Smad2/3. Cell Metab. 33, 1111–1123.e1114 (2021).

    CAS  PubMed  Google Scholar 

  6. Papalazarou, V. et al. The creatine–phosphagen system is mechanoresponsive in pancreatic adenocarcinoma and fuels invasion and metastasis. Nat. Metab. 2, 62–80 (2020).

    CAS  PubMed  Google Scholar 

  7. Streijger, F. et al. Mice lacking brain-type creatine kinase activity show defective thermoregulation. Physiol. Behav. 97, 76–86 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Streijger, F. et al. Structural and behavioural consequences of double deficiency for creatine kinases BCK and UbCKmit. Behav. Brain Res. 157, 219–234 (2005).

    CAS  PubMed  Google Scholar 

  9. Van der Zee, C. Hypothalamic plasticity of neuropeptide Y is lacking in brain-type creatine kinase double knockout mice with defective thermoregulation. Eur. J. Pharmacol. 719, 137–144 (2013).

    PubMed  Google Scholar 

  10. Steeghs, K. et al. Cytoarchitectural and metabolic adaptations in muscles with mitochondrial and cytosolic creatine kinase deficiencies. Mol. Cell. Biochem. 184, 183–194 (1998).

    CAS  PubMed  Google Scholar 

  11. Kurth, I. et al. Therapeutic targeting of SLC6A8 creatine transporter suppresses colon cancer progression and modulates human creatine levels. Sci. Adv. 7, eabi7511 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Kazak, L. et al. A creatine-driven substrate cycle enhances energy expenditure and thermogenesis in beige fat. Cell 163, 643–655 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Bong, S. M. et al. Structural studies of human brain-type creatine kinase complexed with the ADP–Mg2+–NO3–creatine transition-state analogue complex. FEBS Lett. 582, 3959–3965 (2008).

    CAS  PubMed  Google Scholar 

  14. Shen, Y. Q., Tang, L., Zhou, H. M. & Lin, Z. J. Structure of human muscle creatine kinase. Acta Crystallogr. D Biol. Crystallogr. 57, 1196–1200 (2001).

    CAS  PubMed  Google Scholar 

  15. Fritz-Wolf, K., Schnyder, T., Wallimann, T. & Kabsch, W. Structure of mitochondrial creatine kinase. Nature 381, 341–345 (1996).

    CAS  PubMed  Google Scholar 

  16. Eder, M., Fritz-Wolf, K., Kabsch, W., Wallimann, T. & Schlattner, U. Crystal structure of human ubiquitous mitochondrial creatine kinase. Proteins 39, 216–225 (2000).

    CAS  PubMed  Google Scholar 

  17. Xiao, H. et al. A quantitative tissue-specific landscape of protein redox regulation during aging. Cell 180, 968–983 e924 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Lu, W. et al. Correction: fragment-based covalent ligand discovery. RSC Chem. Biol. 2, 670–671 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Backus, K. M. et al. Proteome-wide covalent ligand discovery in native biological systems. Nature 534, 570–574 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Kuljanin, M. et al. Reimagining high-throughput profiling of reactive cysteines for cell-based screening of large electrophile libraries. Nat. Biotechnol. 39, 630–641 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Weerapana, E. et al. Quantitative reactivity profiling predicts functional cysteines in proteomes. Nature 468, 790–795 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Vinogradova, E. V. et al. An activity-guided map of electrophile–cysteine interactions in primary human T cells. Cell 182, 1009–1026.e1029 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Coles, B. F. & Kadlubar, F. F. Detoxification of electrophilic compounds by glutathione S-transferase catalysis: determinants of individual response to chemical carcinogens and chemotherapeutic drugs? Biofactors 17, 115–130 (2003).

    CAS  PubMed  Google Scholar 

  24. Li, Q. et al. Insights into the phosphoryl transfer mechanism of human ubiquitous mitochondrial creatine kinase. Sci. Rep. 6, 38088 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Smith, R. A., Hartley, R. C. & Murphy, M. P. Mitochondria-targeted small molecule therapeutics and probes. Antioxid. Redox Signal. 15, 3021–3038 (2011).

    CAS  PubMed  Google Scholar 

  26. Lillie, J. W. et al. Cyclocreatine (1-carboxymethyl-2-iminoimidazolidine) inhibits growth of a broad spectrum of cancer cells derived from solid tumors. Cancer Res. 53, 3172–3178 (1993).

    CAS  PubMed  Google Scholar 

  27. Naito, Y., Takagi, T. & Higashimura, Y. Heme oxygenase-1 and anti-inflammatory M2 macrophages. Arch. Biochem. Biophys. 564, 83–88 (2014).

    CAS  PubMed  Google Scholar 

  28. Hudalla, H. et al. Carbonic anhydrase inhibition ameliorates inflammation and experimental pulmonary hypertension. Am. J. Respir. Cell Mol. Biol. 61, 512–524 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Strowitzki, M. J. et al. Carbon dioxide sensing by immune cells occurs through carbonic anhydrase 2-dependent changes in intracellular pH. J. Immunol. 208, 2363–2375 (2022).

    CAS  PubMed  Google Scholar 

  30. Zhang, X. et al. Calcium/calmodulin-dependent protein kinase (CaMK) Ialpha mediates the macrophage inflammatory response to sepsis. J. Leukoc. Biol. 90, 249–261 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Reddy, A. et al. pH-Gated succinate secretion regulates muscle remodeling in response to exercise. Cell 183, 62–75 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Johnson, J. L. et al. An atlas of substrate specificities for the human serine/threonine kinome. Nature 613, 759–766 (2023).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Hutti, J. E. et al. A rapid method for determining protein kinase phosphorylation specificity. Nat. Methods 1, 27–29 (2004).

    CAS  PubMed  Google Scholar 

  34. Turk, B. E., Huang, L. L., Piro, E. T. & Cantley, L. C. Determination of protease cleavage site motifs using mixture-based oriented peptide libraries. Nat. Biotechnol. 19, 661–667 (2001).

    CAS  PubMed  Google Scholar 

  35. Kabsch, W. Integration, scaling, space-group assignment and post-refinement. Acta Crystallogr. D Biol. Crystallogr. 66, 133–144 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

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

    PubMed  Google Scholar 

  39. Darabedian, N., Chen, T. C., Molina, H., Pratt, M. R. & Schönthal, A. H. Bioorthogonal profiling of a cancer cell proteome identifies a large set of 3-bromopyruvate targets beyond glycolysis. ACS Chem. Biol. 13, 3054–3058 (2018).

    CAS  PubMed  Google Scholar 

  40. Mills, E. L. et al. Itaconate is an anti-inflammatory metabolite that activates Nrf2 via alkylation of KEAP1. Nature 556, 113–117 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Pettinger, J., Carter, M., Jones, K. & Cheeseman, M. D. Kinetic optimization of lysine-targeting covalent inhibitors of HSP72. J. Med. Chem. 62, 11383–11398 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This work was supported by the Claudia Adams Barr Program (E.T.C.), the Lavine Family Fund (E.T.C.), the Pew Charitable Trust (E.T.C.), National Institutes of Health (NIH) CA259739-01 (E.T.C.), AG071966-01 (E.T.C.), NIH DK123095 (E.T.C.), The Smith Family Foundation (E.T.C.), T32CA236754 (N.D.), the National Cancer Center (NCI) (H.X. and N.D.) NCI R35 CA210030 (K. Stegmaier) and P50 CA206963 (K. Stegmaier), K99AG073461 (H.X.), NCI R35 CA231991 (B.F.C.), the Linde Family Foundation (S.D.-P.), the Doris Duke Charitable Foundation (S.D.-P.), Deerfield 3DC (S.D.-P.), Taiho Pharmaceuticals (S.D.-P.), NCI K99 CA263161 (S.L), and NIH (4R00DK123321-03), Mathers Foundation (MF-2204-02617) and American Cancer Society (DBG-23-983219-01-TBE) (E.L.M). S.L. is a Fellow of the Leukemia & Lymphoma Society. This work was based upon research conducted at the Northeastern Collaborative Access Team beamlines, which were funded by the National Institute of General Medical Sciences from the National Institutes of Health (P30 GM124165). The Eiger 16M detector on the 24-ID-E beamline was funded by a NIH-Office of Research Infrastructure Programs High-End Instrumentation grant (S10OD021527). 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 No. DE-AC02-06CH11357.

Author information

Authors and Affiliations

Authors

Contributions

E.T.C. and N.S.G. conceived of and designed the study. N.D. and S.L. performed cellular experiments and analyzed data. W.J., M.F., J.C. and T.Z. designed and conducted chemical syntheses. N.D. E.M.H, J.L.J. and T.M.Y carried out and analyzed data from mass spectrometry experiments. N.D. assisted with protein expression and purification. H.X. developed the CPT screening platform. E.V.V., K. Senkane and B.F.C. conceived, designed and conducted gel-based screening experiments. E.L.M. performed macrophage experiments and metabolomics experiments. S.D.-P. oversaw CK expression and purification and crystallization. J.C. performed molecular modeling. K. Stegmaier and S.L. contributed to conceptual design and provided cellular reagents. E.T.C. and N.S.G. directed research, oversaw the experiments and wrote the manuscript with assistance from the other authors. L.C.C oversaw kinome profiling experiments.

Corresponding authors

Correspondence to Nathanael S. Gray or Edward T. Chouchani.

Ethics declarations

Competing interests

N.S.G. is a founder, science advisory board member (SAB) and equity holder in Syros, C4, Allorion, Lighthorse, Voronoi, Inception, Matchpoint, CobroVentures, GSK, Shenandoah (board member), Larkspur (board member) and Soltego (board member). The Gray lab receives or has received research funding from Novartis, Takeda, Astellas, Taiho, Jansen, Kinogen, Arbella, Deerfield, Springworks, Interline and Sanofi. E.T.C. is a founder, board member and equity holder in Matchpoint Therapeutics and Aevum Therapeutics. T.Z. and J.C. are founders, equity holders and consultants in Matchpoint Therapeutics. J.C. is a scientific co-founder M3 Bioinformatics & Technology Inc., and consultant and equity holder for Soltego and Allorion. K. Stegmaier has funding from Novartis Institute of Biomedical Research and Kronos Bio, consults for and has stock options in Auron Therapeutics and previously consulted for Kronos Bio and AstraZeneca. T.M.Y. is a co-founder, stockholder and on the board of directors of DESTROKE, Inc. T.Z., W.J., N.D., N.S.G., J.C. and E.T.C. are inventors on a patent for CK inhibitors WO 2022/087433 A8. All other authors declare no potential conflict of interest. B.F.C. is a founder and scientific advisor to Vividion Therapeutics.

Peer review

Peer review information

Nature Chemical Biology thanks Apirat Chaikuad 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 Characterizing CKi interaction with CKB.

(a) Establishing gel-based screening of recombinant CKB in Ramos cell lysates for engagement of cysteine 283 by competition labeling with iodoacetamide-rhodamine. Single experiment shown. (b) Fragment competition labeling of recombinant CKB in Ramos cell lysates identifies small molecules that compete with iodoacetamide-rhodamine labeling. CKi is highlighted in green. Single experiment shown. (c) Structure of CKi-alkyne. (d) Left: Intact protein MS of 800 nM human recombinant CKB incubated for 2 h with DMSO at 37 °C. Right: Intact protein MS of 800 nM human recombinant CKB incubated for 2 h with 1.6 μM CKi-alkyne at 37 °C. (e) UCSD-AML1 cells incubated with alkyne-CKi followed by conjugation to flouorophore for gel based detection. Fluorophore-conjugated iodoacetamide included as a promiscuous labeling control. (f) Crystal structure active site of human CKB bound to CKi highlighting Cys283, ADP, creatine, location of ligands based on PDB: 3B6R.

Extended Data Fig. 2 Effects of CKi on recombinant CKB.

(a) Electron density of CKi in the active site of CKB. A 2FO-FC map of CKi and its attached residue (Cys283) at sigma 1.0. (b) % CKi engagement with human recombinant CKB assessed by monitoring intact mass of CKB over 2 h at 37 °C. (c) Recombinant CKB phosphotransfer activity ± incubation with CKi for indicated times. n = 3 independent experiments. (d) Kinact/Ki determined using % CKi engagement with human recombinant CKB assessed by monitoring intact mass of CKB over 2 h at 37 °C at different concentrations of CKi. n = 3. (e) ATP/ADP ratio determined in UCSD-AML1 after treating with 0, 1.25, 2.5 5, 10, and 20 μM CKi for 16 h using ADP/ATP ratio assay kit. n = 3 biological cell replicates. (f) Structure of select CKi analogs with increasing hydrocarbon lengths. (g) Recombinant CKB phosphotransfer activity ± 2 h preincubation with CKi analogs with increasing hydrocarbon length. (h) Creatine and phosphocreatine abundance in UCSD-AML1 and A549 cells treated with 10 μM CKi. n = 5. (i) HEK293 cells over-expressing (OE) Flag-CKB exhibit significant increase in total cellular CKB content. Representative blot from 2 independent experiments shown.

Source data

Extended Data Fig. 3 Phosphoproteome remodeling induced by CKi and MitoCKi.

(a) The effect of CKi on cell viability in a range of cancer cell lines. HEK; HEK293 cells, HEKOECKB are cells generated in Extended Data Fig. 2i. n = 8. (b) Cell type specific phosphoproteomic signature of UCSD-AML1 and A549 cells treated for 16 h with 10 μM CKi or 5 μM MitoCKi. n = 5. (c) Full phosphoproteomic analysis of UCSD-AML1 or A549 cells treated with 10 μM CKi or 5 μM MitoCKi for 16 h. (n = 5). Upregulated and downregulated sites were then subjected to kinase enrichment analysis as describe in Methods. (d) Volcano plots showing phosphoproteomic analysis of UCSD-AML1 or A549 cells treated with 10 μM CKi or 5 μM MitoCKi for 16 h. n = 5. (e) Volcano plots showing phosphoproteomic analysis of UCSD-AML1 or A549 cells treated with 10 μM CKi or 5 μM MitoCKi for 16 h. n = 5.

Extended Data Fig. 4 CKi flow cytometry analysis.

(a) Flow cytometry analysis using Annexin V and 7-AAD staining of UCSD-AML1 or A549 cells treated with DMSO or 10 μM CKi for 16 h. n = 3. (b) Flow cytometry analysis using Click-iT Plus EdU Alexa Fluor 647 and FxCycle Violet staining of UCSD-AML1 or A549 cells treated with DMSO or 10 μM CKi for 16 h. n = 3.

Extended Data Fig. 5 CKi and MitoCKi flow cytometry analyses.

(a) Flow cytometry analysis using Annexin V and 7-AAD staining of UCSD-AML1 or A549 cells treated with DMSO, 2.5 or 5 μM MitoCKi for 16 h. (b) Quantification of fraction of cells that are apoptotic after 16 h of treatment with DMSO, 2.5 or 5 μM MitoCKi. n = 3. (c) Flow cytometry analysis using Click-iT Plus EdU Alexa Fluor 647 and FxCycle Violet staining of UCSD-AML1 or A549 cells treated with DMSO, 2.5 or 5 μM MitoCKi for 16 h. (d) Quantification of fraction of cells that are in S, G0/G1 and G2/M phase of the cell cycle after 16 h of treatment with DMSO, 2.5 or 5 μM MitoCKi. n = 3.

Source data

Extended Data Fig. 6 Kinome analysis and TLR pathway analysis of CKi.

(a) Kinome-wide selectivity profile for compound CKi at 10 μM. CKi was tested at 10 μM on a panel of 468 kinases (Creatine Kinases are not in the panel). The results are displayed as red circles with their sizes correlating with the inhibitor’s binding affinity (percent DMSO control). Complete dataset is included in Supplementary Table 7. (b) Immunoblot of IκBα and phosphorylated IκBα in THP1 cells ± CKi and ± LPS. Representative blot from 3 independent experiments shown.

Source data

Supplementary information

Supplementary Information

Supplementary note containing all chemical synthesis information.

Reporting Summary

Supplementary Tables

Supplementary Tables 1–10.

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 1

Uncropped western blots.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 5

Statistical source data.

Source Data Extended Data Fig.2

Statistical source data.

Source Data Extended Data Fig. 2

Uncropped western blots.

Source Data Extended Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 6

Uncropped western blots.

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

Darabedian, N., Ji, W., Fan, M. et al. Depletion of creatine phosphagen energetics with a covalent creatine kinase inhibitor. Nat Chem Biol 19, 815–824 (2023). https://doi.org/10.1038/s41589-023-01273-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41589-023-01273-x

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer