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:

Fructose-1,6-bisphosphatase 1 dephosphorylates and inhibits TERT for tumor suppression

An Author Correction to this article was published on 22 April 2024

This article has been updated

Abstract

Telomere dysfunction is intricately linked to the aging process and stands out as a prominent cancer hallmark. Here we demonstrate that telomerase activity is differentially regulated in cancer and normal cells depending on the expression status of fructose-1,6-bisphosphatase 1 (FBP1). In FBP1-expressing cells, FBP1 directly interacts with and dephosphorylates telomerase reverse transcriptase (TERT) at Ser227. Dephosphorylated TERT fails to translocate into the nucleus, leading to the inhibition of telomerase activity, reduction in telomere lengths, enhanced senescence and suppressed tumor cell proliferation and growth in mice. Lipid nanoparticle-mediated delivery of FBP1 mRNA inhibits liver tumor growth. Additionally, FBP1 expression levels inversely correlate with TERT pSer227 levels in renal and hepatocellular carcinoma specimens and with poor prognosis of the patients. These findings demonstrate that FBP1 governs cell immortality through its protein phosphatase activity and uncover a unique telomerase regulation in tumor cells attributed to the downregulation or deficiency of FBP1 expression.

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: FBP1 acts as a protein phosphatase to dephosphorylate TERT.
Fig. 2: C129 of FBP1 is in a reduced state and forms a covalent phospho-C129 intermediate for TERT pS227 dephosphorylation.
Fig. 3: FBP1 reduces the nuclear accumulation of TERT, telomerase activity and telomere lengths and tumor cell senescence and proliferation inhibition.
Fig. 4: FBP1 expression inhibits tumor growth by inhibiting TERT activity.
Fig. 5: FBP1 expression is inversely correlated with TERT S227 phosphorylation level and is negatively associated with the clinical aggressiveness of ccRCC and HCC.
Fig. 6: LNP-mediated delivery of FBP1 mRNA effectively inhibits tumor growth.

Similar content being viewed by others

Data availability

The data that support the findings of this study are available in the main text, extended data figures and supplementary information file. Data are also available from the corresponding author upon reasonable request. The crystal structure of human liver FBP1 is available in the Protein Data Bank under accession code 5ZWK. The structure of TERT was predicted by AlphaFold (https://alphafold.ebi.ac.uk/entry/O14746). Mass spectrometry data have been deposited in the ProteomeXchange with accession code PXD050213. Source data are provided with this paper.

Change history

References

  1. Chakravarti, D., LaBella, K. A. & DePinho, R. A. Telomeres: history, health, and hallmarks of aging. Cell 184, 306–322 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Blackburn, E. H. & Gall, J. G. A tandemly repeated sequence at the termini of the extrachromosomal ribosomal RNA genes in tetrahymena. J. Mol. Biol. 120, 33–53 (1978).

    Article  CAS  PubMed  Google Scholar 

  3. Creighton, H. B. & McClintock, B. A correlation of cytological and genetical crossing-over in Zea mays. Proc. Natl Acad. Sci. USA 17, 492–497 (1931).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Sahin, E. & DePinho, R. A. Axis of ageing: telomeres, p53 and mitochondria. Nat. Rev. Mol. Cell Biol. 13, 397–404 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Nakamura, T. M. & Cech, T. R. Reversing time: origin of telomerase. Cell 92, 587–590 (1998).

    Article  CAS  PubMed  Google Scholar 

  6. Wang, J., Xie, L. Y., Allan, S., Beach, D. & Hannon, G. J. Myc activates telomerase. Genes Dev. 12, 1769–1774 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Li, X. et al. Programmable base editing of mutated TERT promoter inhibits brain tumour growth. Nat. Cell Biol. 22, 282–288 (2020).

    Article  CAS  PubMed  Google Scholar 

  8. Jeong, S. A. et al. Akt-mediated phosphorylation increases the binding affinity of hTERT for importin α to promote nuclear translocation. J. Cell Sci. 128, 2287–2301 (2015).

    Article  CAS  PubMed  Google Scholar 

  9. Tejwani, G. A. Regulation of fructose-bisphosphatase activity. Adv. Enzymol. Relat. Areas Mol. Biol. 54, 121–194 (1983).

    CAS  PubMed  Google Scholar 

  10. Huangyang, P. et al. Fructose-1,6-bisphosphatase 2 inhibits sarcoma progression by restraining mitochondrial biogenesis. Cell Metab. 31, 174–188 (2020).

    Article  CAS  PubMed  Google Scholar 

  11. Li, B. et al. Fructose-1,6-bisphosphatase opposes renal carcinoma progression. Nature 513, 251–255 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Li, F. et al. FBP1 loss disrupts liver metabolism and promotes tumorigenesis through a hepatic stellate cell senescence secretome. Nat. Cell Biol. 22, 728–739 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  13. Liao, K. et al. A feedback circuitry between polycomb signaling and fructose-1, 6-bisphosphatase enables hepatic and renal tumorigenesis. Cancer Res. 80, 675–688 (2020).

    Article  CAS  PubMed  Google Scholar 

  14. Wang, Z. et al. Fructose-1,6-bisphosphatase 1 functions as a protein phosphatase to dephosphorylate histone H3 and suppresses PPARα-regulated gene transcription and tumour growth. Nat. Cell Biol. 24, 1655–1665 (2022).

    Article  CAS  PubMed  Google Scholar 

  15. Li, H., Zhao, L. L., Funder, J. W. & Liu, J. P. Protein phosphatase 2A inhibits nuclear telomerase activity in human breast cancer cells. J. Biol. Chem. 272, 16729–16732 (1997).

    Article  CAS  PubMed  Google Scholar 

  16. Denu, J. M., Stuckey, J. A., Saper, M. A. & Dixon, J. E. Form and function in protein dephosphorylation. Cell 87, 361–364 (1996).

    Article  CAS  PubMed  Google Scholar 

  17. Walton, K. M. & Dixon, J. E. Protein tyrosine phosphatases. Annu. Rev. Biochem. 62, 101–120 (1993).

    Article  CAS  PubMed  Google Scholar 

  18. Qian, X. et al. PTEN suppresses glycolysis by dephosphorylating and inhibiting autophosphorylated PGK1. Mol. Cell 76, 516–527 (2019).

    Article  CAS  PubMed  Google Scholar 

  19. Cesare, A. J. & Reddel, R. R. Alternative lengthening of telomeres: models, mechanisms and implications. Nat. Rev. Genet. 11, 319–330 (2010).

    Article  CAS  PubMed  Google Scholar 

  20. Heaphy, C. M. et al. Altered telomeres in tumors with ATRX and DAXX mutations. Science 333, 425 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Brosnan-Cashman, J. A. et al. ATRX loss induces multiple hallmarks of the alternative lengthening of telomeres (ALT) phenotype in human glioma cell lines in a cell line-specific manner. PLoS ONE 13, e0204159 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  22. Tusell, L., Pampalona, J., Soler, D., Frias, C. & Genesca, A. Different outcomes of telomere-dependent anaphase bridges. Biochem. Soc. Trans. 38, 1698–1703 (2010).

    Article  CAS  PubMed  Google Scholar 

  23. Hou, X., Zaks, T., Langer, R. & Dong, Y. Lipid nanoparticles for mRNA delivery. Nat. Rev. Mater. 6, 1078–1094 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Kon, E., Ad-El, N., Hazan-Halevy, I., Stotsky-Oterin, L. & Peer, D. Targeting cancer with mRNA–lipid nanoparticles: key considerations and future prospects. Nat. Rev. Clin. Oncol. 20, 739–754 (2023).

    Article  CAS  PubMed  Google Scholar 

  25. Kubiatowicz, L. J., Mohapatra, A., Krishnan, N., Fang, R. H. & Zhang, L. mRNA nanomedicine: design and recent applications. Exploration (Beijing) 2, 20210217 (2022).

    Article  PubMed  Google Scholar 

  26. Gu, L. et al. Fructose-1,6-bisphosphatase is a nonenzymatic safety valve that curtails AKT activation to prevent insulin hyperresponsiveness. Cell Metab. 35, 1009–1021 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Zhu, W. et al. Fructose-1,6-bisphosphatase 1 dephosphorylates IκBα and suppresses colorectal tumorigenesis. Cell Res. 33, 245–257 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Chen, M. J., Dixon, J. E. & Manning, G. Genomics and evolution of protein phosphatases. Sci. Signal. 10, eaag1796 (2017).

  29. Romero, P. et al. Computational prediction of human metabolic pathways from the complete human genome. Genome Biol. 6, R2 (2005).

    Article  PubMed  Google Scholar 

  30. 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 

  31. Li, X., Egervari, G., Wang, Y., Berger, S. L. & Lu, Z. Regulation of chromatin and gene expression by metabolic enzymes and metabolites. Nat. Rev. Mol. Cell Biol. 19, 563–578 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Bian, X. et al. Regulation of gene expression by glycolytic and gluconeogenic enzymes. Trends Cell Biol. 32, 786–799 (2022).

    Article  CAS  PubMed  Google Scholar 

  33. Lu, Z. & Hunter, T. Metabolic kinases moonlighting as protein kinases. Trends Biochem. Sci. 43, 301–310 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Dasgupta, S. et al. Metabolic enzyme PFKFB4 activates transcriptional coactivator SRC-3 to drive breast cancer. Nature 556, 249–254 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Xu, D. et al. The gluconeogenic enzyme PCK1 phosphorylates INSIG1/2 for lipogenesis. Nature 580, 530–535 (2020).

    Article  CAS  PubMed  Google Scholar 

  36. Liu, R. et al. Choline kinase alpha 2 acts as a protein kinase to promote lipolysis of lipid droplets. Mol. Cell 81, 2722–2735 (2021).

    Article  CAS  PubMed  Google Scholar 

  37. Guo, D. et al. Aerobic glycolysis promotes tumor immune evasion by hexokinase2-mediated phosphorylation of IκBα. Cell Metab. 34, 1312–1324 (2022).

    Article  CAS  PubMed  Google Scholar 

  38. Liu, G. M. & Zhang, Y. M. Targeting FBPase is an emerging novel approach for cancer therapy. Cancer Cell Int. 18, 36 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  39. Chen, H., Xia, Y., Fang, D., Hawke, D. & Lu, Z. Caspase-10-mediated heat shock protein 90β cleavage promotes UVB irradiation-induced cell apoptosis. Mol. Cell. Biol. 29, 3657–3664 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Tong, Y. et al. SUCLA2-coupled regulation of GLS succinylation and activity counteracts oxidative stress in tumor cells. Mol. Cell 81, 2303–2316 (2021).

    Article  CAS  PubMed  Google Scholar 

  41. Qian, X. et al. Phosphoglycerate kinase 1 phosphorylates Beclin1 to induce autophagy. Mol. Cell 65, 917–931 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Li, X. et al. Nuclear PGK1 alleviates ADP-dependent inhibition of CDC7 to promote DNA replication. Mol. Cell 72, 650–660(2018).

    Article  CAS  PubMed  Google Scholar 

  43. Xu, D. et al. The protein kinase activity of fructokinase A specifies the antioxidant responses of tumor cells by phosphorylating p62. Sci. Adv. 5, eaav4570 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Kimura, M. et al. Measurement of telomere length by the Southern blot analysis of terminal restriction fragment lengths. Nat. Protoc. 5, 1596–1607 (2010).

    Article  CAS  PubMed  Google Scholar 

  45. Zhang, J. M., Yadav, T., Ouyang, J., Lan, L. & Zou, L. Alternative lengthening of telomeres through two distinct break-induced replication pathways. Cell Rep. 26, 955–968 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  46. Idilli, A. I., Segura-Bayona, S., Lippert, T. P. & Boulton, S. J. A C-circle assay for detection of alternative lengthening of telomere activity in FFPE tissue. STAR Protoc. 2, 100569 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Guan, K. L. & Dixon, J. E. Evidence for protein-tyrosine-phosphatase catalysis proceeding via a cysteine-phosphate intermediate. J. Biol. Chem. 266, 17026–17030 (1991).

    Article  CAS  PubMed  Google Scholar 

  48. He, X. et al. Loss of hepatic aldolase B activates Akt and promotes hepatocellular carcinogenesis by destabilizing the Aldob/Akt/PP2A protein complex. PLoS Biol. 18, e3000803 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Mowry, E. M. & Corboy, J. R. Another sphingosine 1-phosphate receptor modulator for the treatment of patients with multiple sclerosis. Lancet Neurol. 18, 983–985 (2019).

    Article  PubMed  Google Scholar 

  50. Du, L. et al. β-Catenin induces transcriptional expression of PD-L1 to promote glioblastoma immune evasion. J. Exp. Med. 217, e20191115 (2020).

  51. Yang, W. et al. Nuclear PKM2 regulates β-catenin transactivation upon EGFR activation. Nature 480, 118–122 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This study was supported by grants from the National Natural Science Foundation of China (82188102 and 82030074, Z.L.; 82072630, D.X.; 82173114, Z.W.; 82103351, M.L.; 82103354, X.H.; 82372816, D.G.), the Ministry of Science and Technology of the People’s Republic of China (2020YFA0803300, Z.L.; 2021YFA0805600, D.X.), the China Postdoctoral Science Foundation (2021M692827, X.H.), the Zhejiang Natural Science Foundation Key Project (LD22H160002, D.X.) and the Starry Night Science Fund of Zhejiang University Shanghai Institute for Advanced Study (SN-ZJU-SIAS-006). Z.L. is the Kuancheng Wang Distinguished Chair.

Author information

Authors and Affiliations

Authors

Contributions

Z.L. conceptualized the study. Z.L., D.X. and M.L. designed the study. M.L., Z.W., J.T., H.H., X.H., S.L., D.G., X.J., L.Y., H.Y., L.X. and Z.M. performed the molecular and animal experiments and analyzed the data. H.J., H.Z., R.Y. and J.F. performed molecular docking. T.L. provided tissue microarray. Z.L. wrote the manuscript, with comments from all other authors.

Corresponding authors

Correspondence to Daqian Xu or Zhimin Lu.

Ethics declarations

Competing interests

Z.L. owns shares in Signalway Life Sciences, which supplied the rabbit antibodies that recognize TERT pS227. Z.L.’s interest in this company had no bearing on its being chosen to supply these reagents. The remaining authors declare no competing interests.

Peer review

Peer review information

Nature Chemical Biology thanks Yuanyu Huang 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 FBP1 binds to TERT and acts as a protein phosphatase to dephosphorylate TERT.

b, c, g, i, j, l, m, Immunoprecipitation (IP) and immunoblotting analyses were performed with the indicated antibodies. The experiments were conducted at least three times with similar results. (a) FBP1 immunoprecipitants from Huh7 cells were analyzed by mass spectrometry. Selected peptide hits are shown. (b) 786-O cells were transfected with or without Flag-FBP1 (b, left) or FBP2 (b, right). Immunoprecipitation using the indicated antibody was performed. (c) 786-O cells were transfected with or without Flag-FBP1 as indicated. Cytosol and nuclear fractions were prepared. Immunoprecipitation was performed. (d, e) The metabolic activities (d), Km and Vmax (e) of WT His-FBP1 or N273A proteins purified from bacteria were determined. Data represent the mean ± s.d. Statistical significance was determined by two-way ANOVA. P values were shown. (f) Glycolytic flux in 786-O cells stably expressing WT Flag-FBP1 or N273A was determined. Data represent the mean ± s.d. (n = 3 biological independent replicates). Statistical significance was determined by two-tailed unpaired Student’s t test. P values were shown. (g) Immunoprecipitation analyses in 786-O cells expressing the indicated Flag-FBP1 proteins. (h) Specificity validation of the anti-TERT pS227 antibody by preincubation of the anti-TERT pS227 antibody with or without the TERT pS227 peptide before IHC analyses. Scale bars, 50 μm. (i) Immunoprecipitation was performed in 786-O cells with indicated antibodies. (j) Bacterially-purified WT His-FBP1 or S170D proteins were incubated with TERT pS227 peptide (j, top). FBP1 protein expressions were shown (j, bottom). The released phosphate was measured. Data represent the mean ± s.d. (n = 3 biological independent replicates). Statistical significance was determined by two-tailed unpaired Student’s t test. P values were shown. (k) Purified 293T-expressed PP2A proteins were incubated with the TERT pS227 peptide. The released phosphate was measured. Data represent the mean ± s.d. (n = 3 biological independent replicates). Statistical significance was determined by two-tailed unpaired Student’s t test. P values were shown. (l, m) Immunoblotting was performed in FBP1-deficient 786-O cells expressing PP2A WT or Y307F proteins (l), and cells expressing a scramble or PPP2CA shRNA (m).

Source data

Extended Data Fig. 2 FBP1 C129 is in a reduced state and forms a covalent phospho-C129 intermediate for TERT pS227 dephosphorylation.

b, f, g, Immunoprecipitation (IP) and immunoblotting analyses were performed with the indicated antibodies. The experiments were conducted at least three times with similar results. (a) Molecular docking analysis showing the structures of a TERT pS227 protein and FBP1 protein. (b) Immunoprecipitation was performed in 786-O cells expressing WT Flag-FBP1 or C129S. (c, d) The metabolic activities (c), Km and Vmax (d) of WT His-FBP1 or C129S proteins purified from bacteria were determined. Data represent the mean ± s.d. Statistical significance was determined by two-way ANOVA. P values were shown. (e) Glycolytic flux in 786-O cells stably expressing WT Flag-FBP1 or C129S was determined. Data represent the mean ± s.d. (n = 3 biological independent replicates). Statistical significance was determined by two-tailed unpaired Student’s t test. P values were shown. (f) Immunoblotting was performed in FBP1 shRNA expressing Huh7 cells with or without Flag-rFBP1 WT or C129S expression. (g) An in vitro kinase reaction was performed by incubating a TERT S227 peptide with purified active GST-AKT1 in the presence or absence of [γ-32P]-ATP. Autoradiography and immunoblot analyses were performed.

Source data

Extended Data Fig. 3 Depletion of FBP1 increases the nuclear accumulation of TERT, telomerase activity, and telomere lengths.

(a-d) Normal renal cells HK2 were stably transfected with a control (shNT) or FBP1 shRNA. Immunofluorescence analyses (a), telomerase activity (b) (n = 3 biological independent replicates), QFISH analyses (c) (n = 8 biological independent replicates) and southern blot analyses for telomere restriction fragment (TRF) (d) were determined. QFISH representative images were shown (c, left), and the immunofluorescence intensity was quantitated for eight cells using the ZEISS Zen Microscopy Software (c, right). Data are representative of three independent experiments. Scale bar, 10 μm. Tel, telomere. (e) Telomerase activity in 786-O cells with or without Flag-FBP2 expression was measured. Data represent the mean ± s.d. (n = 3 biological independent replicates). Statistical significance was determined by two-tailed unpaired Student’s t test.

Source data

Extended Data Fig. 4 FBP1 reduces the nuclear accumulation of TERT, telomerase activity, and telomere lengths and induces tumor cell senescence and inhibited proliferation.

(a) Immunofluorescence staining was performed with the 786-O cells expressing the indicated FBP1 proteins, scale bar, 10 μm. Data are representative of three independent experiments. (b) Cytosolic and nuclear fractions of the 786-O cells with or without expression of the indicated FBP1 protein were prepared. Immunoblotting analyses were performed with the indicated antibodies. The experiments were conducted at least three times with similar results. (c) Telomerase activity (n = 3 biological independent replicates) in FBP1 shRNA expressing Huh7 cells with or without the indicated Flag-rFBP1 protein overexpression was determined. Data represent the mean ± s.d. (n = 3 biological independent replicates). Statistical significance was determined by two-tailed unpaired Student’s t test. P values were shown. (d) Progressed QFISH analyses at the indicated time were performed (top) in 786-O cells, the immunofluorescence intensity was quantitated (bottom). (e) QFISH analyses in FBP1 shRNA expressing Huh7 cells with or without the indicated Flag-rFBP1 protein overexpression was determined. Representative images were shown (top), and the immunofluorescence intensity was quantitated (bottom). Immunofluorescence intensity (d, e) was quantitated for five cells using the ZEISS Zen Microscopy Software. Data represent the mean ± s.d. (n = 8 biological independent replicates in d, n = 5 biological independent replicates in e). Statistical significance was determined by two-tailed unpaired Student’s t test. P values were shown. Scale bar, 10 μm. Tel, telomere.

Source data

Extended Data Fig. 5 FBP1 does not alter ALT activity.

a, c, Immunoblotting analyses were performed with the indicated antibodies. The experiments were conducted at least three times with similar results. (a) Immunoblotting analyses of the indicated 786-O, Huh7 (left), and LN18 cells (right) were performed with the indicated antibodies. (b) C-circle in the 786-O cells expressing WT or the mutated FBP1 (80 days) was measured (n = 3 biological independent replicates). Data represent the mean ± s.d. Statistical significance was determined by two-tailed unpaired Student’s t test. (c) TERT shRNA expressing 786-O cells with or without Flag-FBP1 protein expression were stably transfected with TERT shRNA resistant V5-rTERT WT or S227D. Immunoblotting analyses were performed with the indicated antibodies.

Source data

Extended Data Fig. 6 FBP1 expression inhibits tumor growth by inhibiting TERT activity.

(a, b) Cell proliferation (a) (n = 3 biological independent replicates) and colony formation (b) were analyzed in FBP1 shRNA expressing Huh7 cells with or without the indicated Flag-rFBP1 protein overexpression. (c-e) Huh7 cells with or without depletion of endogenous FBP1 and with or without reconstituted overexpression of the indicated Flag-FBP1 proteins were subcutaneously injected into nude mice (n = 5 per group). Representative images are shown (c). Tumor weights (d) and tumor volumes (e) were measured. Data represent the mean ± s.d. Statistical significance was determined by two-tailed unpaired Student’s t test (a, d, e). P values were shown.

Source data

Extended Data Fig. 7 FBP1-mediated dephosphorylation of TERT disrupts telomere function, leading to senescence of tumor cells and tumor growth inhibition.

IHC analyses were performed with the indicated antibodies. (a) 786-O cells with or without stably expressing the indicated Flag-FBP1 protein were subcutaneously injected into nude mice. (b) FBP1 shRNA expressing Huh7 cells with or without the indicated Flag-rFBP1 protein overexpression were subcutaneously injected into nude mice. (c) TERT shRNA expressing 786-O cells with or without Flag-FBP1 protein expression were stably transfected with TERT shRNA resistant V5-rTERT WT, S227A or S227D. Cells were subcutaneously injected into nude mice. Representative images are shown (upper). The expression levels of the indicated proteins were quantified for ten microscopic fields of the tumor samples (lower). IHC Score=intensity×area. The intensity was graded as 0, 1, 2, 3. The area of positive staining was assessed by 0–25%, 26–50%, 51–75%, 76–100%, corresponding to 0, 1, 2, 3, 4 grades, respectively. Scale bars, 50 μm. Data represent the mean ± s.d. Statistical significance was determined by two-tailed unpaired Student’s t test. P values were shown.

Source data

Extended Data Fig. 8 FBP1 expression is inversely correlated with TERT S227 phosphorylation level and is negatively associated with the clinical aggressiveness of ccRCC and HCC.

(a) IHC analyses of human HCC specimens were performed with the indicated antibodies. Representative images are shown. Scale bars, 100 μm. (b) IHC staining of human HCC samples with the indicated antibodies was scored, and correlation analyses were performed. A Pearson correlation test was used (two-tailed) (n = 90). Some of the dots on the graphs represent more than one specimen (that is, some scores overlapped). P values were shown. (c) Kaplan-Meier plots of the overall survival rates of HCC patients (n = 90) in the groups with high (staining score, 4–8) and low (staining score, 0–3) expression of FBP1 and TERT pS227. The P values were calculated by the log-rank test. P values were shown.

Source data

Extended Data Fig. 9 LNP-mediated delivery of FBP mRNA was safe for mice.

(a) Schematic illustration of LNP preparation. A microfluidic-based mixing of lipids to construct LNPs encapsulating the indicated mRNA. (b-h) The MOCK LNPs or PBS (control group) were intravenously injected into 6-week-old nude mice every other day for a total of 2.5 weeks at a dose of 0.5 mg/kg body weight (n = 5 mice per group). Mice heart, liver, spleen, lung and kidney were harvested for H&E staining (b). Data are representative of six independent micrographs. Scale bars, 100 μm. Mice blood were collected for AST (c), ALT (d), CR (e), BUN (f), CK (g) and LDH (h) analysis (n = 5 mice per group). AST, aspartate transaminase; ALT, alanine aminotransferase; CR, creatinine; BUN, blood urea nitrogen; CK, creatine kinase; LDH, lactate dehydrogenase. (i-k) The 786-O cells were subcutaneously injected into 6-week-old nude mice. The LNPs with or without the indicated FBP1 mRNAs were intravenously injected every other day for a total of 2.5 weeks at a dose of 0.5 mg/kg body weight. [U-13C6]-glucose (1.5 mg/kg body weight) was injected into mouse tail veins before sacrificing the mice. Metabolic flux analyses of injected [U-13C6]-glucose in xenograft tumor (i), liver (j), and kidney (k) were measured (n = 3 mice per group). Data represent the mean ± s.d. Statistical significance was determined by two-tailed unpaired Student’s t test (c-k). P values were shown.

Source data

Extended Data Fig. 10 LNP-mediated delivery of FBP mRNA and effectively inhibits tumor growth.

(a-c) The 786-O cells were subcutaneously injected into 6-week-old nude mice (n = 5 per group). The LNPs with or without the indicated FBP1 mRNAs were intravenously injected every other day at a dose of 0.5 mg/kg body weight for a total of 2.5 weeks. Tumor volumes (a) and weights (b) were calculated. IHC analyses of mouse tumor tissues were performed with the indicated antibodies (c). Representative images are shown (c, upper). The expression levels of the indicated proteins were quantified for ten microscopic fields of the tumor samples (c, lower). Scale bars, 50 μm. Data represent the mean ± s.d. Statistical significance was determined by two-tailed unpaired Student’s t test. P values were shown.

Source data

Supplementary information

Source data

Source Data Fig. 1

Unprocessed western blots

Source Data Fig. 1

Statistical Source Data

Source Data Fig. 2

Unprocessed 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 Fig. 6

Statistical Source Data

Source Data Extended Data Fig. 1

Unprocessed western blots

Source Data Extended Data Fig. 1

Statistical Source Data

Source Data Extended Data Fig. 2

Unprocessed western blots

Source Data Extended Data Fig. 2

Statistical Source Data

Source Data Extended Data Fig. 3

Statistical Source Data

Source Data Extended Data Fig. 4

Unprocessed western blots

Source Data Extended Data Fig. 4

Statistical Source Data

Source Data Extended Data Fig. 5

Unprocessed western blots

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

Source Data Extended Data Fig. 10

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

Li, M., Wang, Z., Tao, J. et al. Fructose-1,6-bisphosphatase 1 dephosphorylates and inhibits TERT for tumor suppression. Nat Chem Biol (2024). https://doi.org/10.1038/s41589-024-01597-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1038/s41589-024-01597-2

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