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:

ENO1 promotes liver carcinogenesis through YAP1-dependent arachidonic acid metabolism

Abstract

Enolase 1 (ENO1) is a glycolytic enzyme that plays essential roles in various pathological activities including cancer development. However, the mechanisms underlying ENO1-contributed tumorigenesis are not well explained. Here, we uncover that ENO1, as an RNA-binding protein, binds to the cytosine-uracil-guanine-rich elements of YAP1 messenger RNA to promote its translation. ENO1 and YAP1 positively regulate alternative arachidonic acid (AA) metabolism by inverse regulation of PLCB1 and HPGD (15-hydroxyprostaglandin dehydrogenase). The YAP1/PLCB1/HPGD axis-mediated activation of AA metabolism and subsequent accumulation of prostaglandin E2 (PGE2) are responsible for ENO1-mediated cancer progression, which can be retarded by aspirin. Finally, aberrant activation of ENO1/YAP1/PLCB1 and decreased HPGD expression in clinical hepatocellular carcinoma samples indicate a potential correlation between ENO1-regulated AA metabolism and cancer development. These findings underline a new function of ENO1 in regulating AA metabolism and tumorigenesis, suggesting a therapeutic potential for aspirin in patients with liver cancer with aberrant expression of ENO1 or YAP1.

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: ENO1 promotes liver cancer progression by activating YAP1 translation as an RBP.
Fig. 2: ENO1 inversely regulates PLCB1 and HPGD expression in a YAP1-dependent manner.
Fig. 3: ENO1 activates the alterative AA biosynthetic pathway via YAP1-regulated PLCB1 and HPGD expression.
Fig. 4: YAP1-regulated AA metabolism via PLCB1 and HPGD expression is important for ENO1-mediated cancer progression.
Fig. 5: Aspirin inhibits ENO1- and YAP1-induced liver cancer progression in vivo.
Fig. 6: Activation of the ENO1-YAP1-AA axis predicts poor prognosis of patients with liver cancer.

Similar content being viewed by others

Data availability

RNA-seq data and RIP-seq data have been deposited in the Gene Expression Omnibus under accession code GSE183703. The KEGG used for gene pathway analysis of RNA-seq data can be accessed at https://www.genome.jp/kegg/. All other data are available in the Source data and Supplementary Information that are provided with this paper.

References

  1. Pavlova, N. N. & Thompson, C. B. The emerging hallmarks of cancer metabolism. Cell Metab. 23, 27–47 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Zhu, J. & Thompson, C. B. Metabolic regulation of cell growth and proliferation. Nat. Rev. Mol. Cell Biol. 20, 436–450 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Kim, J. W. & Dang, C. V. Multifaceted roles of glycolytic enzymes. Trends Biochem. Sci. 30, 142–150 (2005).

    Article  CAS  PubMed  Google Scholar 

  4. Yang, W. et al. PKM2 phosphorylates histone H3 and promotes gene transcription and tumorigenesis. Cell 150, 685–696 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Chang, C. H. et al. Posttranscriptional control of T cell effector function by aerobic glycolysis. Cell 153, 1239–1251 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Zhang, C. S. et al. Fructose-1,6-bisphosphate and aldolase mediate glucose sensing by AMPK. Nature 548, 112–116 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Yang, W. W. et al. Nuclear PKM2 regulates beta-catenin transactivation upon EGFR activation. Nature 480, 118–U289 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Xu, D. et al. The evolving landscape of noncanonical functions of metabolic enzymes in cancer and other pathologies. Cell Metab. 33, 33–50 (2021).

    Article  CAS  PubMed  Google Scholar 

  9. Schwanhausser, B. et al. Global quantification of mammalian gene expression control. Nature 473, 337–342 (2011).

    Article  PubMed  Google Scholar 

  10. Hein, M. Y. et al. A human interactome in three quantitative dimensions organized by stoichiometries and abundances. Cell 163, 712–723 (2015).

    Article  CAS  PubMed  Google Scholar 

  11. Itzhak, D. N., Tyanova, S., Cox, J. & Borner, G. H. H. Global, quantitative and dynamic mapping of protein subcellular localization. eLife 5, e16950 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  12. Diaz-Ramos, A., Roig-Borrellas, A., Garcia-Melero, A. & Lopez-Alemany, R. alpha-Enolase, a multifunctional protein: its role on pathophysiological situations. J. Biomed. Biotechnol. 2012, 156795 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  13. Pancholi, V. Multifunctional alpha-enolase: its role in diseases. Cell. Mol. Life Sci. 58, 902–920 (2001).

    Article  CAS  PubMed  Google Scholar 

  14. Huppertz, I. et al. Riboregulation of Enolase 1 activity controls glycolysis and embryonic stem cell differentiation. Mol. Cell. 82, 2666–2680.e11 (2022).

    Article  CAS  PubMed  Google Scholar 

  15. Kuhnel, K. & Luisi, B. F. Crystal structure of the Escherichia coli RNA degradosome component enolase. J. Mol. Biol. 313, 583–592 (2001).

    Article  CAS  PubMed  Google Scholar 

  16. Carpousis, A. J. The Escherichia coli RNA degradosome: structure, function and relationship in other ribonucleolytic multienzyme complexes. Biochem. Soc. Trans. 30, 150–155 (2002).

    Article  CAS  PubMed  Google Scholar 

  17. Entelis, N. et al. A glycolytic enzyme, enolase, is recruited as a cofactor of tRNA targeting toward mitochondria in Saccharomyces cerevisiae. Genes Dev. 20, 1609–1620 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Hernandez-Perez, L. et al. alpha-Enolase binds to RNA. Biochimie 93, 1520–1528 (2011).

    Article  CAS  PubMed  Google Scholar 

  19. Castello, A. et al. Insights into RNA biology from an atlas of mammalian mRNA-binding proteins. Cell 149, 1393–1406 (2012).

    Article  CAS  PubMed  Google Scholar 

  20. Zhang, T. et al. ENO1 suppresses cancer cell ferroptosis by degrading the mRNA of iron regulatory protein 1. Nat. Cancer 3, 75–89 (2022).

    Article  CAS  PubMed  Google Scholar 

  21. Tang, X., Edwards, E. M., Holmes, B. B., Falck, J. R. & Campbell, W. B. Role of phospholipase C and diacylglyceride lipase pathway in arachidonic acid release and acetylcholine-induced vascular relaxation in rabbit aorta. Am. J. Physiol.-Heart C. 290, H37–H45 (2006).

    Article  CAS  Google Scholar 

  22. Park, J. B. et al. Phospholipase signalling networks in cancer. Nat. Rev. Cancer 12, 782–792 (2012).

    Article  CAS  PubMed  Google Scholar 

  23. Wang, D. Z. & Dubois, R. N. Eicosanoids and cancer. Nat. Rev. Cancer 10, 181–193 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Chan, A. T., Ogino, S. & Fuchs, C. S. Aspirin use and survival after diagnosis of colorectal cancer. J. Am. Med Assoc. 302, 649–659 (2009).

    Article  CAS  Google Scholar 

  25. Holmes, M. D. et al. Aspirin intake and survival after breast cancer. J. Clin. Oncol. 28, 1467–1472 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Thun, M. J., Jacobs, E. J. & Patrono, C. The role of aspirin in cancer prevention. Nat. Rev. Clin. Oncol. 9, 259–267 (2012).

    Article  CAS  PubMed  Google Scholar 

  27. Drew, D. A., Cao, Y. & Chan, A. T. Aspirin and colorectal cancer: the promise of precision chemoprevention. Nat. Rev. Cancer 16, 173–186 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Hogan, D. J., Riordan, D. P., Gerber, A. P., Herschlag, D. & Brown, P. O. Diverse RNA-binding proteins interact with functionally related sets of RNAs, suggesting an extensive regulatory system. PLoS Biol. 6, e255 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  29. Moroishi, T., Hansen, C. G. & Guan, K. L. The emerging roles of YAP and TAZ in cancer. Nat. Rev. Cancer 15, 73–79 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Sanchez-Vega, F. et al. Oncogenic signaling pathways in the cancer genome atlas. Cell 173, 321–337 e310 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Zhao, B. et al. TEAD mediates YAP-dependent gene induction and growth control. Genes Dev. 22, 1962–1971 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Yu, F. X. et al. Regulation of the Hippo-YAP pathway by G-protein-coupled receptor signaling. Cell 150, 780–791 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Nguyen, L. T. et al. ERG activates the YAP1 transcriptional program and induces the development of age-related prostate tumors. Cancer Cell 27, 797–808 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Stein, C. et al. YAP1 exerts its transcriptional control via TEAD-mediated activation of enhancers. PLoS Genet. 11, e1005465 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  35. Sanchez, M., Galy, B., Muckenthaler, M. U. & Hentze, M. W. Iron-regulatory proteins limit hypoxia-inducible factor-2alpha expression in iron deficiency. Nat. Struct. Mol. Biol. 14, 420–426 (2007).

    Article  CAS  PubMed  Google Scholar 

  36. Wu, N. et al. Translation of yes-associated protein (YAP) was antagonized by its circular RNA via suppressing the assembly of the translation initiation machinery. Cell Death Differ. 26, 2758–2773 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Brown, S. Z. et al. The RNA-binding protein HuR posttranscriptionally regulates the protumorigenic activator YAP1 in pancreatic ductal adenocarcinoma. Mol. Cell. Biol. 42, e0001822 (2022).

    Article  PubMed  Google Scholar 

  38. Strnadel, J. et al. eIF5A-PEAK1 signaling regulates YAP1/TAZ protein expression and pancreatic cancer cell growth. Cancer Res. 77, 1997–2007 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Jin, D. et al. m6A mRNA methylation initiated by METTL3 directly promotes YAP translation and increases YAP activity by regulating the MALAT1-miR-1914-3p-YAP axis to induce NSCLC drug resistance and metastasis. J. Hematol. Oncol. 12, 135 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Hudson, W. H. & Ortlund, E. A. The structure, function and evolution of proteins that bind DNA and RNA. Nat. Rev. Mol. Cell Bio. 15, 749–760 (2014).

    Article  CAS  Google Scholar 

  41. Kang, H. J., Jung, S. K., Kim, S. J. & Chung, S. J. Structure of human alpha-enolase (hENO1), a multifunctional glycolytic enzyme. Acta Crystallogr. D. 64, 651–657 (2008).

    Article  CAS  PubMed  Google Scholar 

  42. Guo, X. C. et al. Single tumor-initiating cells evade immune clearance by recruiting type II macrophages. Genes Dev. 31, 247–259 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Li, W. et al. YAP transcriptionally regulates COX-2 expression and GCCSysm-4 (G-4), a dual YAP/COX-2 inhibitor, overcomes drug resistance in colorectal cancer. J. Exp. Clin. Cancer Res. 36, 144 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  44. Kim, H. B. et al. Prostaglandin E2 activates YAP and a positive-signaling loop to promote colon regeneration after colitis but also carcinogenesis in mice. Gastroenterology 152, 616–630 (2017).

    Article  CAS  PubMed  Google Scholar 

  45. Kim, M. K., Jang, J. W. & Bae, S. C. DNA binding partners of YAP/TAZ. BMB Rep. 51, 126–133 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Kim, M., Kim, T., Johnson, R. L. & Lim, D. S. Transcriptional co-repressor function of the Hippo pathway transducers YAP and TAZ. Cell Rep. 11, 270–282 (2015).

    Article  CAS  PubMed  Google Scholar 

  47. Lacoste, B., Raymond, V. A., Cassim, S., Lapierre, P. & Bilodeau, M. Highly tumorigenic hepatocellular carcinoma cell line with cancer stem cell-like properties. PLoS ONE 12, e0171215 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  48. Simsek, D. et al. The mammalian ribo-interactome reveals ribosome functional diversity and heterogeneity. Cell 169, 1051–1065 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Wu, J. et al. Intercellular interaction dictates cancer cell ferroptosis via NF2-YAP signalling. Nature 572, 402–406 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Yang, W. H. et al. The Hippo pathway effector YAP promotes ferroptosis via the E3 ligase SKP2. Mol. Cancer Res. 19, 1005–1014 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Zhang, X. et al. Endogenous glutamate determines ferroptosis sensitivity via ADCY10-dependent YAP suppression in lung adenocarcinoma. Theranostics 11, 5650–5674 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Gao, R. Z. et al. YAP/TAZ and ATF4 drive resistance to Sorafenib in hepatocellular carcinoma by preventing ferroptosis. EMBO Mol. Med. 13, e14351 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Demetz, E. et al. The arachidonic acid metabolome serves as a conserved regulator of cholesterol metabolism. Cell Metab. 20, 787–798 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Wang, T. et al. Selective interaction of the C2 domains of phospholipase C-beta1 and -beta2 with activated Galphaq subunits: an alternative function for C2-signaling modules. Proc. Natl Acad. Sci. USA 96, 7843–7846 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Rhee, S. G. Regulation of phosphoinositide-specific phospholipase C. Annu. Rev. Biochem. 70, 281–312 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Ha, S. D., Lewin, N., Li, S. S. C. & Kim, S. O. HDAC8 activates AKT through upregulating PLCB1 and suppressing DESC1 expression in MEK1/2 inhibition-resistant cells. Cells 10, 1101 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Balsinde, J., Winstead, M. V. & Dennis, E. A. Phospholipase A(2) regulation of arachidonic acid mobilization. FEBS Lett. 531, 2–6 (2002).

    Article  CAS  PubMed  Google Scholar 

  58. Norris, P. C., Gosselin, D., Reichart, D., Glass, C. K. & Dennis, E. A. Phospholipase A2 regulates eicosanoid class switching during inflammasome activation. Proc. Natl Acad. Sci. USA 111, 12746–12751 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Balboa, M. A., Perez, R. & Balsinde, J. Amplification mechanisms of inflammation: paracrine stimulation of arachidonic acid mobilization by secreted phospholipase A(2) is regulated by cytosolic phospholipase A(2)-derived hydroperoxyeicosatetraenoic acid. J. Immunol. 171, 989–994 (2003).

    Article  CAS  PubMed  Google Scholar 

  60. Leslie, C. C. Cytosolic phospholipase A(2): physiological function and role in disease. J. Lipid Res. 56, 1386–1402 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Kalyvas, A. et al. Differing roles for members of the phospholipase A2 superfamily in experimental autoimmune encephalomyelitis. Brain 132, 1221–1235 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  62. Simon, T. G. et al. Association between aspirin use and risk of hepatocellular carcinoma. J. Am. Med. Assoc. Oncol. 4, 1683–1690 (2018).

    Google Scholar 

  63. Lee, T. Y. et al. Association of daily aspirin therapy with risk of hepatocellular carcinoma in patients with chronic hepatitis B. J. Am. Med. Assoc. Intern Med 179, 633–640 (2019).

    Google Scholar 

  64. Simon, T. G. et al. Association of aspirin with hepatocellular carcinoma and liver-related mortality. N. Engl. J. Med. 382, 1018–1028 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Wang, S. J. et al. Association of aspirin therapy with risk of hepatocellular carcinoma: a systematic review and dose-response analysis of cohort studies with 2.5 million participants. Pharmacol. Res. 151, 104585 (2020).

    Article  CAS  PubMed  Google Scholar 

  66. Fox, R. K., Taddei, T. H. & Kaplan, D. E. Aspirin use and risk of hepatocellular carcinoma in hepatitis B. J. Am. Med. Assoc. Intern. Med. 179, 640–641 (2019).

    Google Scholar 

  67. Nair, A. B. & Jacob, S. A simple practice guide for dose conversion between animals and human. J. Basic Clin. Pharm. 7, 27–31 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  68. Sun, L. et al. cMyc-mediated activation of serine biosynthesis pathway is critical for cancer progression under nutrient deprivation conditions. Cell Res. 25, 429–444 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Wu, G. et al. MYC promotes cancer progression by modulating m(6) A modifications to suppress target gene translation. EMBO Rep. 22, e51519 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Liu, S. et al. METTL13 Methylation of eEF1A increases translational output to promote tumorigenesis. Cell 176, 491–504 e421 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Xing, S. et al. DIS3L2 promotes progression of hepatocellular carcinoma via hnRNP U-mediated alternative splicing. Cancer Res. 79, 4923–4936 (2019).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This work is supported in part by the National Natural Science Foundation of China (grant nos. 91957203 to P.G., 82192893, 81930083 and 81821001 to H.Z., 82130087 to P.G., 82273221 to L.S. and 82203556 to C.S.), the Chinese Academy of Sciences (grant no. XDB39020100 to H.Z.) and the National Key R&D Program of China (grant nos. 2022YFA1304504 to H.Z. and 2018YFA0800300 to P.G.).

Author information

Authors and Affiliations

Authors

Contributions

L.S., H.Z. and P.G. conceived the study and supervised the experiments. L.S., C.S., T.Z., S.S., H.Z. and P.G. designed the experiments. L.S. and T.Z. performed RIP and ChIP assays to identify the targets of ENO1 and YAP1, respectively. L.S. and C.S. performed animal studies and analysis of clinical HCC specimens. L.S., C.S. and T.Z. performed immunoblotting, cloning, cell biology and biochemistry experiments. S.S. analyzed RNA-seq and RIP-seq data. X.G., S.Q., P.Z., H.W., W.M., R.Y. and R.C. analyzed the data. W.J. provided clinical specimens. J.C. provided constructive guidance. L.S., C.S., T.Z., H.Z. and P.G. wrote the paper. All the authors read and approved the manuscript.

Corresponding authors

Correspondence to Linchong Sun, Huafeng Zhang or Ping Gao.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Chemical Biology thanks the anonymous reviewers 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 ENO1 regulates the expression of YAP1 target genes.

a, Protein copy numbers of glucose metabolism enzymes in NIH3T3 mouse fibroblast cells (Schwanhausser et al., 2011), and HeLa cells (Hein et al., 2015; Itzhak et al., 2016). b, Venn diagram showing the 21 overlapped target genes enriched by ENO1 IP in our RIP-seq data from HEK293T-Flag-ENO1 cells and the previously published HITS-GBP-CLIP data from HeLa-ENO1-YFP cells (Castello et al., 2012). c, qPCR-based expression analysis of known YAP1 target genes including NPPB, CYR61, CTGF, S1PR1, NEXN, NTF3, KDR, CPA3, PSG1, CCDC80, EGR3, EGR4, DDIT4, and TRAIL in PLC cells expression shRNAs targeting ENO1 or YAP1 (n = 3 independent assays). Error bars denote the mean ± s.e.m. (c).

Source data

Extended Data Fig. 2 ENO1 upregulates YAP1 expression by recruiting YTHDF3.

a, Co-IP assay in HEK293T cells transfected with Flag-ENO1 or Hep3B-Flag-ENO1 cells. b, Protein levels of eIF5A, YTHDF3, and YAP1 were measured by immunoblotting analysis in Hep3B cells with eIF5A or YTHDF3 knockdown. Actin served as a loading control. Par: parental; NC: negative control. c, Binding of endogenous YAP1 mRNA transcripts by Flag-ENO1 in Hep3B-Flag-ENO1 cells with or without YTHDF3 knockdown (n = 3 independent assays). Protein levels of ENO1, YTHDF3, and YAP1 were measured. Actin served as a loading control. d, HEK293T cells expressing shRNA targeting endogenous ENO1 were further transfected with plasmids to overexpress Flag-tagged wild-type ENO1, ENO1S40A, or ENO1D245R mutants (n = 1 sample detected at different time points). Then the ENO1 enzyme activity of these cells was measured by the consumption of NADH in an assay buffer that includes pyruvate kinase (PK), lactate dehydrogenase (LDHA), and the required substrates including 2-phospho-D-glycerate (2PG), phosphoenolpyruvate (PEP), and pyruvate. The consumption of NADH is monitored as a decrease of absorbance at 340 nm. e, Flag and YAP1 protein levels were measured in HEK293T cells expressing Flag-tagged wild-type ENO1 and indicated ENO1 mutant variants. Actin served as a negative control. f, g, Photographs showed xenografts described in Fig. 1l (f). ENO1 and YAP1 protein levels were measured (g). Actin served as a negative control. Immunoblots are representative of two independent experiments (a-c,e). Error bars denote the mean ± s.e.m. (c,d). Statistical analyses were performed by two-tailed Student’s t-test (c,d).

Source data

Extended Data Fig. 3 ENO1 and YAP1 transcriptionally activate arachidonic acid synthetic enzymes.

a, Venn diagram of the RNA-seq data showing the overlapped genes regulated by ENO1 and YAP1 in PLC cells with ENO1 or YAP1 knockdown. The statistical significance between ENO1-regulated genes and YAP1-regulated genes was determined using Fisher’s exact probability test, P < 0.001. b, KEGG mapper analysis of the pathways in which the co-regulated genes of ENO1 and YAP1 are enriched. 66 genes of metabolic pathways are shown in Supplementary Table 1. c, Gene Ontology (GO) enrichment analysis. The top ten GO terms are listed. Supplementary Table 2 provides a complete list of terms and corresponding gene lists. P-value was computed using the one-sided Fisher’s exact test and corrected for multiple hypothesis testing using false discovery rate (FDR). d, qPCR analysis of ENO1, YAP1, PLCB1, PTGS2 and HPGD expression in PLC cells expressing shRNAs targeting ENO1 or YAP1 (n = 3 independent assays). e, Immunoblotting analysis of ENO1, YAP1, and HPGD expression in PLC cells expressing shRNAs targeting ENO1 or YAP1. Actin served as a negative control. f, Protein levels of ENO1, YAP1, PLCB1, and HPGD were measured in Hepa 1–6 cells expressing shRNAs targeting ENO1 or YAP1. Actin served as a negative control. g, h, Protein levels of ENO1, YAP1, PLCB1, COX2, and HPGD were measured in PLC cells (g) or Hep3B cells (h) expressing ENO1 or YAP1. Actin served as a negative control. i, Protein levels of YAP1, PLCB1, COX2, and HPGD were measured in HEK293T cells expressing wild-type YAP1 or YAP-5SA mutant. Actin served as a negative control. Immunoblots are representative of three independent experiments. Error bars denote the mean ± s.e.m. (d). Statistical analyses were performed by two-tailed Student’s t-test (d).

Source data

Extended Data Fig. 4 The NuRD complex is required for ENO1/YAP1-mediated HPGD gene repression.

a, b, qPCR (a) and immunoblotting (b) analysis of HPGD expression levels in PLC cells treated with 1 μM TSA, or expressing shRNAs targeting MTA2, HDAC1, or HDAC2 (mRNA data: n = 3 independent assays). Actin served as a negative control. c, PLC cells expressing Flag-tagged YAP1 or ENO1 were further infected with shRNA viruses targeting HDAC1. Protein levels of YAP1, ENO1, Flag, HDAC1, and HPGD were measured by immunoblotting analysis. Actin served as a negative control. d, A schematic representation of the ENO1-mediated activation of YAP1 transcriptional regulation of the PLCB1 and HPGD loci. Illustration created with Biorender.com. Immunoblots are representative of three independent experiments. Error bars denote the mean ± s.e.m. (a). Statistical analyses were performed by two-tailed Student’s t-test (a).

Source data

Extended Data Fig. 5 ENO1 and YAP1 activate AA metabolism in a PLCB1- and HPGD-dependent manner.

a, AA and PGE2 contents were measured in PLC cells expressing ENO1 or YAP1 by ELISA (n = 3 independent assays). b, c, Immunoblotting and qPCR analysis of HPGD expression in PLC cells expressing shRNAs targeting HPGD (b), followed by the detection of PGE2 content and cell growth status (c) (n = 3 independent assays). Actin served as a negative control. d, Protein levels of ENO1 or YAP1, and HPGD (lower panel), and PGE2 content (upper panel) were measured in ENO1 or YAP1-overexpressing PLC cells expressing HPGD (n = 3 independent assays). Actin served as a negative control. e, AA content was measured in ENO1- or YAP1-overexpressing Hep3B cells infected with shRNA viruses targeting PLCB1 (n = 3 independent assays). f, Protein levels of HPGD were measured by immunoblotting in endogenous ENO1-knockdown PLC cells expressing wild-type ENO1, ENO1S40A, and ENO1D245R. AA and PGE2 contents in these cell lines were measured by ELISA (n = 3 independent assays). Actin served as a negative control. Immunoblots are representative of three independent experiments. Error bars denote the mean ± s.e.m. (a-f) or mean ± s.d. (c, lower panel). Statistical analyses were performed by two-tailed Student’s t-test (a-f), or two-way ANOVA (c, lower panel).

Source data

Extended Data Fig. 6 The accelerated tumor progression promoted by ENO1/YAP1 depends on PLCB1 expression.

a, Cell growth was measured in ENO1 or YAP1-overexpressing Hep3B cells infected with shRNA viruses targeting PLCB1 (n = 4 independent assays). b, Photographs show xenografts described in Fig. 4a at the end of the experiment. c, Protein levels of Flag, ENO1, YAP1, and PLCB1 in tumor lysates of Extended Data Fig. 6b were measured by immunoblotting analysis. Actin served as a loading control. d, Protein levels of YAP1 and PLCB1 were measured in YAP1-knockdown Hep3B cells further infected with shRNA viruses targeting PLCB1 (upper panel). Actin served as a negative control. Growth curves were measured by trypan blue counting (lower panel, n = 3 independent assays). e, f, Equal numbers of Hep3B cells expressing shYAP1 or/and shPLCB1 were injected subcutaneously into the flanks of BALB/c nude mice (n = 5 mice in each group). Tumor sizes were measured every 3 days using calipers (e). Tumor weights (f) were determined at the end of the experiment (day 30). g, Protein levels of YAP1 and PLCB1 in tumor lysates of Extended Data Fig. 6f were measured by immunoblotting analysis. Actin served as a loading control. Immunoblots are representative of two independent experiments (d). Error bars denote the mean ± s.e.m. (a,d-f). Statistical analyses were performed by two-tailed Student’s t-test (f), or two-way ANOVA with tukey’s multiple comparisons test (a,d,e).

Source data

Extended Data Fig. 7 The accelerated tumor progression promoted by ENO1/YAP1 depends on HPGD expression.

a, Cell growth was measured in ENO1 or YAP1-knockdown PLC cells infected with shRNA viruses targeting HPGD (n = 3 independent assays). b, Cell growth was measured in ENO1 or YAP1-overxpressing PLC cells expressing HPGD (n = 3 independent assays). c, Photographs show xenografts described in Fig. 4e at the end of the experiment. d, Protein levels of ENO1, YAP1, and HPGD in tumor lysates of Extended Data Fig. 7c were measured by immunoblotting analysis. Actin served as a loading control. e, PLC cells expressing shRNA targeting endogenous ENO1 were further transfected with plasmids to overexpress Flag-tagged wild-type ENO1, ENO1S40A, ENO1D245R, or ENO1M4mutants. Protein levels of ENO1 and Flag were measured by immunoblotting analysis. Actin served as a loading control. f, ENO1 enzyme activity of cells in Extended Data Fig. 7e was measured (n = 3 independent assays). g, Growth curves were measured by trypan blue counting in PLC cells expressing the EV, wild-type ENO1, or indicated ENO1 mutants with endogenous ENO1 knockdown (n = 3 independent assays). h, Photographs show xenografts described in Fig. 4k at the end of the experiment. i, Protein levels of Flag, ENO1, YAP1, and HPGD were detected in the tumor lysates described in Extended Data Fig. 7h. Actin served as a loading control. Immunoblots are representative of two independent experiments (e). Error bars denote the mean ± s.d. (a,b,g) or mean ± s.e.m. (f). Statistical analyses were performed by two-tailed Student’s t-test (f), two-way ANOVA (g), or two-way ANOVA with tukey’s multiple comparisons test (a,b).

Source data

Extended Data Fig. 8 COX2 is responsible for ENO1/YAP1-mediated tumor growth.

a, b, Cell growth was measured in ENO1- or YAP1-overexpressing PLC cells (a) or Hep3B cells (b) infected with shRNA viruses targeting COX2 (n = 3 independent assays). Protein levels of ENO1, YAP1, and COX2 were measured by immunoblotting analysis. Actin served as a loading control. c-f, Equal numbers of Hep3B cells expressing ENO or YAP1 that further knockdown COX2 were injected subcutaneously into the flanks of BALB/c nude mice (n = 5 mice in each group). Tumor sizes were measured every 3 days using calipers (c, d). Tumor weights (e, f) were determined at the end of the experiment (day 29). g, h, Protein levels of ENO1, YAP1, and COX2 in tumor lysates of Extended Data Fig. 8e, f were measured by immunoblotting analysis. Actin served as a loading control. Immunoblots are representative of three independent experiments (a,b). Error bars denote the mean ± s.d. (a,b) or mean ± s.e.m. (c-f). Statistical analyses were performed by two-tailed Student’s t-test (e,f), or two-way ANOVA with tukey’s multiple comparisons test (a-d).

Source data

Extended Data Fig. 9 Aspirin inhibits ENO1- and YAP1-induced liver cancer in vivo.

a, Protein levels of COX2 were measured in PLC cells treated with different doses of aspirin. Actin served as a negative control. b, COX2 enzyme activity was measured in the tumor lysates of Fig. 5b using ELISA (n = 10). c, Protein levels of ENO1 and YAP1 were measured in the tumor lysates of Fig. 5b. Actin served as a negative control. d, Protein levels of mus-ENO1 or mus-YAP1 were measured in Hepa 1–6 cells stably infected with viruses expressing mus-ENO1 or mus-YAP1. e, COX2 enzyme activity was measured in the tumor lysates of Fig. 5e, g using ELISA (n = 5). f, g, Protein levels of ENO1 (Flag), YAP1, PLCB1, and HPGD were measured in the tumor lysates of Fig. 5e, g. Calnexin served as a loading control. h, COX2 enzyme activity was measured in the tumor lysates of Fig. 5i using ELISA (n = 5). i, j, protein (i) and mRNA (j) levels of YAP1, PTGS2 (COX2), PLCB1, and HPGD were detected in the tumor lysates of Fig. 5i. Calnexin served as a loading control. Immunoblots are representative of three independent experiments (a,d). Error bars denote the mean ± s.e.m. (b,e,h,j). Statistical analyses were performed by two-tailed Student’s t-test (b,e,h,j).

Source data

Extended Data Fig. 10 ENO1 promotes liver cancer progression by regulating PLCB1 and HPGD-mediated arachidonic acid metabolism through YAP1.

a, Summary: ENO1, as an RNA-binding protein, activates YAP1 translation by binding to the (CUG)n triplet rich elements within the 3’UTR of YAP1 mRNA. YAP1 content increases and translocates into the nucleus to promote PLCB1 expression, but directly represses HPGD expression at transcriptional levels, which is responsible for the ENO1-mediated activation of the alterative arachidonic acid pathway, PGE2 accumulation, cell proliferation, and cancer progression. Aspirin, a blocker of the AA pathway by inhibiting COX2 activity and PGE2 production, could inhibit liver cancer progression, especially in liver cancers with elevated expression of ENO1 and YAP1. Illustration created with Biorender.com.

Supplementary information

Source data

Source Data Fig. 1

Unprocessed western blots.

Source Data Fig. 2

Unprocessed western blots.

Source Data Fig. 3

Unprocessed western blots.

Source Data Fig. 6

Unprocessed western blots.

Source Data Extended Data Fig. 2

Unprocessed western blots.

Source Data Extended Data Fig. 3

Unprocessed western blots.

Source Data Extended Data Fig. 4

Unprocessed western blots.

Source Data Extended Data Fig. 5

Unprocessed western blots.

Source Data Extended Data Fig. 6

Unprocessed western blots.

Source Data Extended Data Fig. 7

Unprocessed western blots.

Source Data Extended Data Fig. 8

Unprocessed western blots.

Source Data Extended Data Fig. 9

Unprocessed western blots.

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 Fig. 5

Statistical source data.

Source Data Fig. 6

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.

Source Data Extended Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 7

Statistical source data.

Source Data Extended Data Fig. 8

Statistical source data.

Source Data Extended Data Fig. 9

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

Sun, L., Suo, C., Zhang, T. et al. ENO1 promotes liver carcinogenesis through YAP1-dependent arachidonic acid metabolism. Nat Chem Biol 19, 1492–1503 (2023). https://doi.org/10.1038/s41589-023-01391-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41589-023-01391-6

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