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:

Chemoproteomic mapping of the glycolytic targetome in cancer cells

Abstract

Hyperactivated glycolysis is a metabolic hallmark of most cancer cells. Although sporadic information has revealed that glycolytic metabolites possess nonmetabolic functions as signaling molecules, how these metabolites interact with and functionally regulate their binding targets remains largely elusive. Here, we introduce a target-responsive accessibility profiling (TRAP) approach that measures changes in ligand binding-induced accessibility for target identification by globally labeling reactive proteinaceous lysines. With TRAP, we mapped 913 responsive target candidates and 2,487 interactions for 10 major glycolytic metabolites in a model cancer cell line. The wide targetome depicted by TRAP unveils diverse regulatory modalities of glycolytic metabolites, and these modalities involve direct perturbation of enzymes in carbohydrate metabolism, intervention of an orphan transcriptional protein’s activity and modulation of targetome-level acetylation. These results further our knowledge of how glycolysis orchestrates signaling pathways in cancer cells to support their survival, and inspire exploitation of the glycolytic targetome for cancer therapy.

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: Benchmarking the TRAP approach for targetome mining in native cell milieu.
Fig. 2: TRAP can identify thermally stable protein targets and hence complements thermal stability-based target-discovery approaches.
Fig. 3: TRAP mapped a global glycolytic targetome in HCT116 cells.
Fig. 4: Functional validation of glycolytic metabolites on the carbohydrate metabolism enzymes assigned as glycolytic targets by TRAP.
Fig. 5: Lactate engagement influenced the transcriptional activity of TRIM28.
Fig. 6: Glycolytic metabolites modulated acetylation levels of their binding targets.

Similar content being viewed by others

Data availability

Reprocessed Meltome datasets were accessed with the identifier PXD011929, reprocessed LiP-Quant dataset was accessed with the identifier PXD015446 and our data can be accessed with the identifier IPX0002602000/PXD022568 through the ProteomeXchange Consortium (http://proteomecentral.proteomexchange.org). RNA-seq data are stored in the NCBI GEO repository under accession number GSE225738. Source data are provided with this paper.

References

  1. Wishart, D. Emerging applications of metabolomics in drug discovery and precision medicine. Nat. Rev. Drug Discov. 15, 473–484 (2016).

    Article  CAS  PubMed  Google Scholar 

  2. Zasłona, Z. & O’Neill, L. Cytokine-like roles for metabolites in immunity. Mol. Cell 78, 814–823 (2020).

    Article  PubMed  Google Scholar 

  3. Dang, L. et al. Cancer-associated IDH1 mutations produce 2-hydroxyglutarate. Nature 462, 739–744 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Zhang, W. et al. Lactate is a natural suppressor of RLR signaling by targeting MAVS. Cell 178, 176–189.e15 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Piazza, I. et al. A map of protein–metabolite interactions reveals principles of chemical communication. Cell 172, 358–372.e23 (2018).

    Article  CAS  PubMed  Google Scholar 

  7. Geiger, R. et al. l-Arginine modulates T cell metabolism and enhances survival and anti-tumor activity. Cell 167, 829–842.e13 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Kornberg, M. D. et al. Dimethyl fumarate targets GAPDH and aerobic glycolysis to modulate immunity. Science 360, 449–453 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Humphries, F. et al. Succination inactivates gasdermin D and blocks pyroptosis. Science 369, 1633–1637 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Pavlova, N. N., Zhu, J. & Thompson, C. B. The hallmarks of cancer metabolism: still emerging. Cell Metab. 34, 355–377 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Zhang, D. et al. Metabolic regulation of gene expression by histone lactylation. Nature 574, 575–580 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Wan, N. et al. Cyclic immonium ion of lactyllysine reveals widespread lactylation in the human proteome. Nat. Methods 19, 854–864 (2022).

    Article  CAS  PubMed  Google Scholar 

  13. Niphakis, M. J. et al. A global map of lipid-binding proteins and their ligandability in cells. Cell 161, 1668–1680 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Hulce, J. J. et al. Proteome-wide mapping of cholesterol-interacting proteins in mammalian cells. Nat. Methods 10, 259–264 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  15. Qin, W. et al. Chemoproteomic profiling of itaconation by bioorthogonal probes in inflammatory macrophages. J. Am. Chem. Soc. 142, 10894–10898 (2020).

    Article  CAS  PubMed  Google Scholar 

  16. Zhang, Y. et al. Chemoproteomic profiling of itaconations in Salmonella. Chem. Sci. 12, 6059–6063 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Lomenick, B. et al. Target identification using drug affinity responsive target stability (DARTS). Proc. Natl Acad. Sci. USA 106, 21984–21989 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Savitski, M. M. et al. Tracking cancer drugs in living cells by thermal profiling of the proteome. Science 346, 1255784 (2014).

    Article  PubMed  Google Scholar 

  19. Martinez Molina, D. et al. Monitoring drug target engagement in cells and tissues using the cellular thermal shift assay. Science 341, 84–87 (2013).

    Article  PubMed  Google Scholar 

  20. Zhang, X. et al. Solvent-induced protein precipitation for drug target discovery on the proteomic scale. Anal. Chem. 92, 1363–1371 (2020).

    Article  CAS  PubMed  Google Scholar 

  21. West, G. M., Tang, L. & Fitzgerald, M. C. Thermodynamic analysis of protein stability and ligand binding using a chemical modification- and mass spectrometry-based strategy. Anal. Chem. 80, 4175–4185 (2008).

    Article  CAS  PubMed  Google Scholar 

  22. Sridharan, S. et al. Proteome-wide solubility and thermal stability profiling reveals distinct regulatory roles for ATP. Nat. Commun. 10, 1155 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  23. Reinhard, F. B. et al. Thermal proteome profiling monitors ligand interactions with cellular membrane proteins. Nat. Methods 12, 1129–1131 (2015).

    Article  CAS  PubMed  Google Scholar 

  24. Mendoza, V. L. & Vachet, R. W. Probing protein structure by amino acid-specific covalent labeling and mass spectrometry. Mass Spectrom. Rev. 28, 785–815 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. McKenzie-Coe, A., Montes, N. S. & Jones, L. M. Hydroxyl radical protein footprinting: a mass spectrometry-based structural method for studying the higher order structure of proteins. Chem. Rev. 122, 7532–7561 (2022).

    Article  CAS  PubMed  Google Scholar 

  26. Zhou, Y. et al. Prediction of ligand modulation patterns on membrane receptors via lysine reactivity profiling. Chem. Commun. 55, 4311–4314 (2019).

    Article  CAS  Google Scholar 

  27. Bamberger, C. et al. Protein footprinting via covalent protein painting reveals structural changes of the proteome in Alzheimer’s disease. J. Proteome Res. 20, 2762–2771 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Raines, R. T. Ribonuclease A. Chem. Rev. 98, 1045–1066 (1998).

    Article  CAS  PubMed  Google Scholar 

  29. Dombrauckas, J. D., Santarsiero, B. D. & Mesecar, A. D. Structural basis for tumor pyruvate kinase M2 allosteric regulation and catalysis. Biochemistry 44, 9417–9429 (2005).

    Article  CAS  PubMed  Google Scholar 

  30. Anastasiou, D. et al. Pyruvate kinase M2 activators promote tetramer formation and suppress tumorigenesis. Nat. Chem. Biol. 8, 839–847 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Jarzab, A. et al. Meltome atlas–thermal proteome stability across the tree of life. Nat. Methods 17, 495–503 (2020).

    Article  CAS  PubMed  Google Scholar 

  32. Schrader, J. et al. The inhibition mechanism of human 20S proteasomes enables next-generation inhibitor design. Science 353, 594–598 (2016).

    Article  CAS  PubMed  Google Scholar 

  33. Piazza, I. et al. A machine learning-based chemoproteomic approach to identify drug targets and binding sites in complex proteomes. Nat. Commun. 11, 4200 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  34. Guo, Y. et al. Cryo-EM structures of recombinant human sodium–potassium pump determined in three different states. Nat. Commun. 13, 3957 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Pavlovic, D. Endogenous cardiotonic steroids and cardiovascular disease, where to next? Cell Calcium 86, 102156 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Chang, A. et al. BRENDA, the ELIXIR core data resource in 2021: new developments and updates. Nucleic Acids Res. 49, D498–D508 (2021).

    Article  CAS  PubMed  Google Scholar 

  37. Hasmann, M. & Schemainda, I. FK866, a highly specific noncompetitive inhibitor of nicotinamide phosphoribosyltransferase, represents a novel mechanism for induction of tumor cell apoptosis. Cancer Res. 63, 7436–7442 (2003).

    CAS  PubMed  Google Scholar 

  38. Khan, J. A., Tao, X. & Tong, L. Molecular basis for the inhibition of human NMPRTase, a novel target for anticancer agents. Nat. Struct. Mol. Biol. 13, 582–588 (2006).

    Article  CAS  PubMed  Google Scholar 

  39. Lambert, S. A. et al. The human transcription factors. Cell 172, 650–665 (2018).

    Article  CAS  PubMed  Google Scholar 

  40. Addison, J. B. et al. KAP1 promotes proliferation and metastatic progression of breast cancer cells. Cancer Res. 75, 344–355 (2015).

    Article  CAS  PubMed  Google Scholar 

  41. Tang, Z. et al. GEPIA: a web server for cancer and normal gene expression profiling and interactive analyses. Nucleic Acids Res. 45, W98–W102 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Stoll, G. A. et al. Structure of KAP1 tripartite motif identifies molecular interfaces required for retroelement silencing. Proc. Natl Acad. Sci. USA 116, 15042–15051 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Iyengar, S. & Farnham, P. J. KAP1 protein: an enigmatic master regulator of the genome. J. Biol. Chem. 286, 26267–26276 (2021).

    Article  Google Scholar 

  44. Acharyya, S. et al. A CXCL1 paracrine network links cancer chemoresistance and metastasis. Cell 150, 165–178 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Kawada, K. et al. Chemokine receptor CXCR3 promotes colon cancer metastasis to lymph nodes. Oncogene 26, 4679–4688 (2007).

    Article  CAS  PubMed  Google Scholar 

  46. Huang, H. et al. iPTMnet: an integrated resource for protein post-translational modification network discovery. Nucleic Acids Res. 46, D542–D550 (2018).

    Article  CAS  PubMed  Google Scholar 

  47. Huang, H. et al. p300-mediated lysine 2-hydroxyisobutyrylation regulates glycolysis. Mol. Cell 70, 663–678.e6 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Das, C. et al. CBP/p300-mediated acetylation of histone H3 on lysine 56. Nature 459, 113–117 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Williams, R. J. Trichostatin A, an inhibitor of histone deacetylase, inhibits hypoxia-induced angiogenesis. Expert Opin. Investig. Drugs 10, 1571–1573 (2001).

    Article  CAS  PubMed  Google Scholar 

  50. Huang, J. P. & Ling, K. EZH2 and histone deacetylase inhibitors induce apoptosis in triple negative breast cancer cells by differentially increasing H3 Lys27 acetylation in the BIM gene promoter and enhancers. Oncol. Lett. 14, 5735–5742 (2017).

    PubMed  PubMed Central  Google Scholar 

  51. Liu, Y. et al. Novel histone deacetylase inhibitors derived from Magnolia officinalis significantly enhance TRAIL-induced apoptosis in non-small cell lung cancer. Pharmacol. Res. 111, 113–125 (2016).

    Article  CAS  PubMed  Google Scholar 

  52. Zhou, Y. et al. Probing the lysine proximal microenvironments within membrane protein complexes by active dimethyl labeling and mass spectrometry. Anal. Chem. 88, 12060–12065 (2016).

    Article  CAS  PubMed  Google Scholar 

  53. Villar, H. O. & Kauvar, L. M. Amino acid preferences at protein binding sites. FEBS Lett. 349, 125–130 (1994).

    Article  CAS  PubMed  Google Scholar 

  54. Lu, G. et al. Subresidue-resolution footprinting of ligand–protein interactions by carbene chemistry and ion mobility-mass spectrometry. Anal. Chem. 92, 947–956 (2020).

    Article  CAS  PubMed  Google Scholar 

  55. Fang, M. et al. Elucidating protein–ligand interactions in cell lysates using high-throughput hydrogen-deuterium exchange mass spectrometry with integrated protein thermal depletion. Anal. Chem. 95, 1805–1810 (2023).

    Article  CAS  Google Scholar 

  56. Yan, W. et al. Living cell-target responsive accessibility profiling reveals silibinin targeting ACSL4 for combating ferroptosis. Anal. Chem. 94, 14820–14826 (2022).

    Article  CAS  PubMed  Google Scholar 

  57. Zheng, X., Cai, X. & Hao, H. Emerging targetome and signalome landscape of gut microbial metabolites. Cell Metab. 34, 35–58 (2022).

    Article  CAS  PubMed  Google Scholar 

  58. Henley, M. J. & Koehler, A. N. Advances in targeting ‘undruggable’ transcription factors with small molecules. Nat. Rev. Drug Discov. 20, 669–688 (2021).

    Article  CAS  PubMed  Google Scholar 

  59. Karki, R. et al. NLRC3 is an inhibitory sensor of PI3K-mTOR pathways in cancer. Nature 540, 583–587 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Hoxhaj, G. & Manning, B. D. The PI3K-AKT network at the interface of oncogenic signalling and cancer metabolism. Nat. Rev. Cancer 20, 74–88 (2020).

    Article  CAS  PubMed  Google Scholar 

  61. Chen, W. W. et al. Absolute quantification of matrix metabolites reveals the dynamics of mitochondrial metabolism. Cell 166, 1324–1337.e11 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Røst, L. M. et al. Absolute quantification of the central carbon metabolome in eight commonly applied prokaryotic and eukaryotic model systems. Metabolites 10, 74 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  63. Miyo, M. et al. Metabolic adaptation to nutritional stress in human colorectal cancer. Sci. Rep. 6, 38415 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Feng, Y. et al. Global analysis of protein structural changes in complex proteomes. Nat. Biotechnol. 32, 1036–1044 (2014).

    Article  CAS  PubMed  Google Scholar 

  65. Haeussler, M. et al. The UCSC Genome Browser database: 2019 update. Nucleic Acids Res. 47, D853–D858 (2019).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This research was supported by the National Natural Science Foundation of China (grant 81930109 to H.H., grant 82173783 to H.Y.), the Natural Science Foundation of Jiangsu Province (BK20220088), the National Key Research and Development Program of China (2021YFA1301300), the Fundamental Research Funds for the Central Universities (2632022YC03), the Overseas Expertise Introduction Project for Discipline Innovation (G20582017001) and the Sanming Project of Medicine in Shenzhen (SZSM201801060). We thank Y. Xiao from China Pharmaceutical University, Q. Yu from Harvard University, B. Shan and W. Li from PEAKS Studio for useful discussions. We also acknowledge N. Wang in the Cellular and Molecular Biology Center of China Pharmaceutical University and W. Jiang in the State Key Laboratory of Natural Medicines of China Pharmaceutical University for their technical support.

Author information

Authors and Affiliations

Authors

Contributions

H. Hao, H.Y. and G.W. conceived the project. Y.T., N.W., H.Z., C.S., H.Y. and H. Hao designed the experiments. Y.T., N.W., H.Z., H.Y. and M.D. performed the proteomics experiments. Y.T., H.Z., C.S. and C.L. performed the flow cytometry and western blotting experiments. Q.B. performed the SPR experiments. K.Z. and S.C. carried out protein site mutation and purification experiments. H. Hu conducted the conservation analysis. N.W., Y.T., H.S., H.Y. and H. Hao analyzed the experimental data. N.W., H.Y. and H. Hao wrote the paper with input from coauthors.

Corresponding authors

Correspondence to Hui Ye or Haiping Hao.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Chemical Biology thanks Mikhail Savitski, Chu Wang 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 Benchmarking the TRAP approach in probing ligand-target interactions using RNase and PKM2.

(a) Docking analysis of RNase with its ligand CDP (PDB: 1ROB). (b) Native MS showing CDP and CTP bound to RNase at different affinities. (c) Microscale thermophoresis (MST) showing CTP possessed stronger affinity to RNase than CDP. The experiment was conducted once. (d) Time-resolved intact MS analysis showing impeded mass shift of TRAP-labeled RNase in response to CDP/CTP binding (n indicates the number of TRAP-labeled lysines on RNase). (e) Summarized accessibility changes of lysines (Rtreated/control) in RNase in response to CDP/CTP binding based on quantitative analysis of labeled lysine-containing peptides via TRAP. (f) Crystal structure of FBP (red, sphere)-induced PKM2 tetramerization (grey, cartoon) by an allosteric mechanism (PDB: 4B2D). The measured minimal Euclidean distances suggested that the PKM2-K422 residue (blue, stick) is distant from both FBP and pyruvate (in dashed lines). (g) Summarized accessibility changes of lysines (Rtreated/control) in human recombinant PKM2 in response to FBP incubation based on quantitative analysis of labeled lysine-containing peptides via TRAP. For (e) and (g), data represent the mean ± SEM (n=3 biologically independent samples) and P values were determined using an unpaired two-tailed Student’s t-test.

Extended Data Fig. 2 TRAP identified PKM2 as the target of TEPP-46 and pinpointed the binding site.

(a) Crystal structure suggesting the binding site of TEPP-46 (red, sphere) as PKM2-K305 (PDB: 3U2Z). The TRAP-assigned TRP bearing K305 was colored in blue. (b) Correlation analysis between SASA and labeling occupancy of lysine residues of PKM2 without (left panel) and with TEPP-46 (right panel). The Pearson’s correlation coefficients (r) and statistical significance of correlation (P) determined by unpaired two-tailed Student’s t-test are shown. In agreement with the decreased SASA, labeling occupancy of PKM2-K305 (in red), the binding site of TEPP-46, was also markedly reduced following TEPP-46 incubation. Specifically, labeling occupancy of each K was estimated by calculating the ratio between abundance of the peptides bearing this labeled K residue and summed abundance of the labeled K-containing peptides as well as those carrying this unlabeled K residue. (c) TRAP identified the labeled K305 and K311-bearing TRPs in human recombinant PKM2, both of which signified markedly decreased accessibility at K305/K311 following TEPP-46 incubation. RTEPP-46/control were calculated based on n=3 biologically independent samples, while representative EICs of the TRPs and the non-TRPs bearing K115/K188 are shown. (d) Summarized accessibility changes of lysines (Rtreated/control) using data in (c). (e) Rtreated/control of the labeled K305-containing TRP in E. coli lysates following TEPP-46 treatment (n=5 biologically independent samples). (f) Dose-responsive accessibility change curves for TEPP-46’s target candidates that were assigned by single-dose TRAP experiment shown in Fig. 1h (the bona fide target PKM2 excepted). The experiment was conducted once. For (d) and (e), data represent the mean ± SEM and P values were determined using an unpaired two-tailed Student’s t-test.

Extended Data Fig. 3 TRAP complements thermal stability-based target discovery approaches.

(a) Analysis of protein melting behaviors using the human HEK293T cell meltome data (PXD011929) identified five clusters as shown here and in Fig. 2a. (b) Nonmelters displaying resistance to thermal denaturation were identified in the meltome data of HaCaT, HepG2 and colon cancer spheroids cells (PXD011929). (c) GO BP analysis of the nonmelters shown in (b) using a modified Fisher’s Exact test from DAVID bioinformatics website. (d) Dose-responsive TRAP detected dose-dependent increased abundance of the TRPtype C of PSMB1-K164, implying dose-dependent decreased accessibility at K164 following bortezomib incubation. The experiment was conducted once. (e) Dose-responsive TRAP curves for bortezomib’s target candidates that were assigned by single-dose TRAP experiment shown in Fig. 2f (the bona fide target PSMB1 excepted). The experiment was conducted once.

Extended Data Fig. 4 TRAP complements proteolytic stability-based target discovery approaches.

(a) FT% distribution plots of the re-analyzed publicly available LiP-Quant proteome datasets (PXD015446). The numbers of the identified proteins were listed as n and suggested good proteome coverage. (b) Crystal structures of human ATP1A1 in the E1 (PDB: 7E1Z) and E2 states (PDB: 7E20). The identified TRPtype A bearing K91 was colored in red. (c) TRAP analysis identified the labeled K91-bearing TRPtype A spanning D75-R94 of ATP1A1. Its Rtreated/control showed dose-dependent response to digoxin incubation. Data represent the mean ± SEM (n=4 biologically independent samples) and P values were determined using an unpaired two-tailed Student’s t-test. (d) Illustrated protein coverage of ATP1A1 (PDB: 7E1Z) detected by TRAP and LiP. Only labeled lysine-bearing TRPtype A and LiP-produced HT peptides were used for sequence mapping.

Extended Data Fig. 5 Characterizing the TRAP-assigned glycolytic targetome using the multiplexed-TRAP data.

(a) Wide TRAP-labeling coverage of lysines based on analysis of the second batch of the multiplexed-TRAP data. (b-c) Chemical accessibility of proteinaceous lysines assessed by the labeled fraction of lysine residues for each quantified protein using the first (b) and second (c) batches of the multiplexed-TRAP data. (d) Analysis of TRAP labeling preference for high-order structures using the second batch of the multiplexed-TRAP data. (e) Summary of the numbers of glycolytic metabolite-protein interactions detected by TRAP. (f-g) Box plots (center lines mark the median, box borders represent the first and third quartiles, and the whiskers indicate the minimum and maximum values) of sequence coverage (f) and the number of detected unique peptides (g) for known glycolytic targets (retrieved from BRENDA, species: human) that can and cannot be identified by TRAP (unpaired two-tailed Student’s t-test).

Extended Data Fig. 6 Functional assessment and validation of the identified glycolytic metabolite-target interactions belonging to the carbohydrate metabolism pathway.

(a) GO MF analysis of the TRAP-identified glycolytic targetome showing enrichment in proteins ascribed to catalytic activity. (b) KEGG pathway annotation summarized the enriched pathways for the targets ascribed to catalytic activity. (c) Illustration of how the boundary of active site is defined using the multiplexed-TRAP data. The minimal Euclidean distances between the TRPs of the TRAP-identified targets that use the assayed metabolites as substrates and the corresponding active site (retrievable from PDB) were measured, and the resultant median of the collected distances was used to represent the active site boundary that can be probed by TRAP. (d) Summarized Rtreated/control values of the TRPs in PKM2 via the multiplexed-TRAP analysis of HCT116 cell lysates following FBP, F6P and G6P incubation, respectively. Of note, the letter before K denotes the type of the classified TRPs. (e) Relative (Rel.) PKM2 activity at different FBP concentrations normalized to the activity without FBP. (f) TRAP analysis delivering the Rtreated/control of the TRPtype B carrying PKM2-K433 following 3PG incubation. (g) DrugBank and non-DrugBank fractions of the quantified proteome (n=4778) vs. the TRAP-assigned glycolytic targetome (n=913) using the multiplexed-TRAP data. For (d, e, f), data represent the mean ± SEM (n=3 biologically independent samples). For (d, f), P values were determined using an unpaired two-tailed Student’s t-test.

Extended Data Fig. 7 Functional and structural characterization of the lactate-TRIM28 interaction.

(a) Gene expression profile of TRIM28 across given cancer types and paired normal tissues retrieved from the TCGA and the GTEx projects were plotted using GEPIA. (b) GEPIA-based analysis implying that high TRIM28 gene expression level is unfavored for patients’ survival for the examined cancer types using Log-rank test (median cutoff). (c) Summarized Rtreated/control values of the TRPs assigned for lactate via the LFQ-TRAP analysis of human recombinant TRIM28 (upper panel, all peptides shown here are TRPType A candidates) and the multiplexed-TRAP analysis of HCT116 cell lysates (bottom panel, the letter before K denotes the type of the classified TRPs). Data represent the mean ± SEM (n=3 biologically independent samples) and P values were determined using an unpaired two-tailed Student’s t-test.

Extended Data Fig. 8 Glycolytic metabolites binding regulated targetome acetylation.

(a) Fraction of lysines in quantified peptides vs. lysines in TRPs of the glycolytic targetome based on PTM annotation with iPTMnet. (b) Percentage of lysine acetylation in non-TRPs vs. TRPs retrieved from the same glycolytic target (n=913). Data represent the mean ± SEM and P value was determined by a paired, two-tailed Student’s t-test. (c) GO analysis showing diverse subcellular locations for the fraction of TRAP-assigned glycolytic targets that have been assigned as acetylation carriers by iPTMnet. The subcellular distribution pattern resembles that of the whole glycolytic targetome. (d) Distribution pattern of MFs summarized for the whole glycolytic targetome vs. MFs of the glycolytic targetome that have been documented as acetylation carriers by iPTMnet. (e) Number of acetylated lysines in TRPs of each assayed glycolytic metabolite based on iPTMnet.

Extended Data Fig. 9 Validation of ENO1 as the binding target of G3P.

(a) Summarized Rtreated/control values of the TRPs in ENO1 assigned for G3P using the multiplexed-TRAP data. Of note, the letter before K denotes the Type A/B/C of TRPs. Data represent the mean ± SEM (n=3 biologically independent samples) and P values were determined using an unpaired two-tailed Student’s t-test. (b) Thermal shift assay validating the engagement of G3P (500 μM) with ENO1 using HCT116 cell lysates. Experiments were repeated (n=3 biologically independent samples) with one representative sample shown. (c) Thermal shift assay showing G3P-mediated stabilization of ENO1 using HCT116 cell lysates. Experiments were repeated (n=2 biologically independent samples) with one representative sample shown. (d) Volcano plot of the TRPs of G3P (500 μM) assigned by TRAP analysis of the human recombinant ENO1 (n=3 biologically independent samples). TRPs (RG3P/control >1.5 or <0.67, p <0.05 by unpaired two-sided Student’s t-test) were determined and highlighted in red. (e) SPR analysis showing weakened affinity of G3P to the ENO1 carrying K330E point mutation (Mutant-ENO1). The experiment was conducted once.

Extended Data Fig. 10 Metabolomic and immunoblotting analysis verified the ability of pyruvate in entering HCT116 cells and rescuing TSA-induced apoptosis.

(a) EICs of intracellular pyruvate in HCT116 cells without and with pyruvate administration for 24 hr (n=3 biologically independent samples). (b) Relative ion abundance of intracellular pyruvate using data in (a). Data represent the mean ± SEM (n=3 biological samples) and P value was determined using an unpaired two-tailed Student’s t-test. (c) Gating strategy to sort TSA-induced apoptotic HCT116 cells without and with pyruvate pretreatment as specified in Fig. 6g. (d) Representative immunoblots of the apoptotic protein markers, including cleaved PARP and cleaved caspase-9, in HCT116 cells treated as specified in Fig. 6g. β-Tubulin was used as the loading control. Statistical analyses of the band intensities of the protein markers are shown in the bottom panel, where data represent the mean ± SEM (n=3 independent experiments) and P values were determined using ordinary one-way ANOVA with Tukey’s multiple comparisons test.

Source data

Supplementary information

Reporting Summary

Supplementary Dataset 1

Analysis of LiP-resisting proteins by assessing FT% from four publicly available LiP-proteome datasets.

Supplementary Dataset 2

Summary of glycolytic targetome and TRPs assigned by TRAP in HCT116 cells.

Supplementary Dataset 3

Summarized interactions of the known and quantified glycolytic metabolites-enzymatic targets as retrieved from BRENDA.

Supplementary Dataset 4

KEGG pathway analysis of the TRAP-assigned glycolytic targetome with catalytic activity.

Supplementary Dataset 5

Summary of the minimal Euclidean distances measured between the detected TRPs of enzymatic targets that use the examined metabolites as substrates and their active sites.

Supplementary Dataset 6

RNA-seq analysis of TRIM28-dependent gene expression changes in response to lactate treatment in HCT116 cells.

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 6

Unprocessed western blots.

Source Data Extended Data Fig. 10

Unprocessed 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

Tian, Y., Wan, N., Zhang, H. et al. Chemoproteomic mapping of the glycolytic targetome in cancer cells. Nat Chem Biol 19, 1480–1491 (2023). https://doi.org/10.1038/s41589-023-01355-w

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41589-023-01355-w

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