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:

cGAS suppresses hepatocellular carcinoma independent of its cGAMP synthase activity

Abstract

Cyclic GMP–AMP synthase (cGAS) is a key innate immune sensor that recognizes cytosolic DNA to induce immune responses against invading pathogens. The role of cGAS is conventionally recognized as a nucleotidyltransferase to catalyze the synthesis of cGAMP upon recognition of cytosolic DNA, which leads to the activation of STING and production of type I/III interferon to fight against the pathogen. However, given that hepatocytes are lack of functional STING expression, it is intriguing to define the role of cGAS in hepatocellular carcinoma (HCC), the liver parenchymal cells derived malignancy. In this study, we revealed that cGAS was significantly downregulated in clinical HCC tissues, and its dysregulation contributed to the progression of HCC. We further identified cGAS as an immune tyrosine inhibitory motif (ITIM) containing protein, and demonstrated that cGAS inhibited the progression of HCC and increased the response of HCC to sorafenib treatment by suppressing PI3K/AKT/mTORC1 pathway in cellular and animal models. Mechanistically, cGAS recruits SH2-containing tyrosine phosphatase 1 (SHP1) via ITIM, and dephosphorylates p85 in phosphatidylinositol 3-kinase (PI3K), which leads to the suppression of AKT-mTORC1 pathway. Thus, cGAS is identified as a novel tumor suppressor in HCC via its function independent of its conventional role as cGAMP synthase, which indicates a novel therapeutic strategy for advanced HCC by modulating cGAS signaling.

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: Loss of cGAS expression was correlated with poor prognosis in HCC patients.
Fig. 2: cGAS suppressed the tumorigenesis of HCC cells.
Fig. 3: cGAS inhibited the AKT-mTORC1 pathway in HCC cells.
Fig. 4: cGAS recruited and interacted with SHP1 in HCC cells.
Fig. 5: cGAS-SHP1 dephosphorylated p85 and suppressed PI3K–AKT pathway.
Fig. 6: cGAS inhibited HCC via the SHP1–p85 axis independent of its nucleotidyltransferase activity.
Fig. 7: cGAS reversed the resistance to sorafenib treatment in HCC by inhibiting AKT pathway.
Fig. 8: Schematic working model.

Similar content being viewed by others

Data availability

The datasets used during the current study are available from the corresponding authors (hanlihui@sdu.edu.cn) on reasonable request. Raw western blots are available in the Supplementary file.

References

  1. Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global Cancer Statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA: Cancer J Clin. 2021;71:209–49.

    PubMed  Google Scholar 

  2. Pinter M, Scheiner B, Peck-Radosavljevic M. Immunotherapy for advanced hepatocellular carcinoma: a focus on special subgroups. Gut. 2020;70:204–14.

    Article  PubMed  Google Scholar 

  3. Lee HW, Cho KJ, Park JY. Current status and future direction of immunotherapy in hepatocellular carcinoma: what do the data suggest? Immune Netw. 2020;20:e11.

    Article  PubMed  PubMed Central  Google Scholar 

  4. Armstrong SA, He AR. Immuno-oncology for hepatocellular carcinoma: the present and the future. Clin Liver Dis. 2020;24:739–53.

    Article  PubMed  Google Scholar 

  5. Gan Y, Li X, Han S, Liang Q, Ma X, Rong P, et al. The cGAS/STING pathway: a novel target for cancer therapy. Front Immunol. 2021;12:795401.

    Article  CAS  PubMed  Google Scholar 

  6. Dhanisha SS, Guruvayoorappan C. Potential role of cGAS/STING pathway in regulating cancer progression. Crit Rev Oncol/Hematol. 2022;178:103780.

    Article  PubMed  Google Scholar 

  7. Schadt L, Sparano C, Schweiger NA, Silina K, Cecconi V, Lucchiari G, et al. Cancer-cell-intrinsic cGAS expression mediates tumor immunogenicity. Cell Rep. 2019;29:1236–48. e1237

    Article  CAS  PubMed  Google Scholar 

  8. Song Y, Liu Y, Teo HY, Hanafi ZB, Mei Y, Zhu Y, et al. Manganese enhances the antitumor function of CD8(+) T cells by inducing type I interferon production. Cell Mol Immunol. 2021;18:1571–4.

    Article  CAS  PubMed  Google Scholar 

  9. Ablasser A, Chen ZJ. cGAS in action: expanding roles in immunity and inflammation. Science. 2019;363:eaat8657.

    Article  CAS  PubMed  Google Scholar 

  10. Hao F. Entanglement of methylation changes and cGAS-STING signaling in non-small-cell lung cancer. Comb Chem High Throughput Screen. 2023;26:224–35.

  11. Thomsen MK, Nandakumar R, Stadler D, Malo A, Valls RM, Wang F, et al. Lack of immunological DNA sensing in hepatocytes facilitates hepatitis B virus infection. Hepatology. 2016;64:746–59.

    Article  CAS  PubMed  Google Scholar 

  12. Thomsen MK, Skouboe MK, Boularan C, Vernejoul F, Lioux T, Leknes SL, et al. The cGAS-STING pathway is a therapeutic target in a preclinical model of hepatocellular carcinoma. Oncogene. 2020;39:1652–64.

    Article  CAS  PubMed  Google Scholar 

  13. Yu Y, Liu Y, An W, Song J, Zhang Y, Zhao X. STING-mediated inflammation in Kupffer cells contributes to progression of nonalcoholic steatohepatitis. J Clin Investig. 2019;129:546–55.

    Article  PubMed  Google Scholar 

  14. Luo X, Li H, Ma L, Zhou J, Guo X, Woo SL, et al. Expression of STING is increased in liver tissues from patients with NAFLD and promotes macrophage-mediated hepatic inflammation and fibrosis in mice. Gastroenterology. 2018;155:1971–84. e1974

    Article  CAS  PubMed  Google Scholar 

  15. Hopfner KP, Hornung V. Molecular mechanisms and cellular functions of cGAS-STING signalling. Nat Rev Mol Cell Biol. 2020;21:501–21.

    Article  CAS  PubMed  Google Scholar 

  16. Sun EJ, Wankell M, Palamuthusingam P, McFarlane C, Hebbard L. Targeting the PI3K/Akt/mTOR pathway in hepatocellular carcinoma. Biomedicines. 2021;9:1639.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Lu X, Paliogiannis P, Calvisi DF, Chen X. Role of the mammalian target of rapamycin pathway in liver cancer: from molecular genetics to targeted therapies. Hepatology. 2021;73:49–61.

    Article  CAS  PubMed  Google Scholar 

  18. Guo M, Li N, Zheng J, Wang W, Wu Y, Han X, et al. Epigenetic regulation of hepatocellular carcinoma progression through the mTOR signaling pathway. Can J Gastroenterol Hepatol. 2021;2021:5596712.

    Article  PubMed  PubMed Central  Google Scholar 

  19. Ritchie C, Carozza JA, Li L. Biochemistry, cell biology, and pathophysiology of the innate immune cGAS-cGAMP-STING pathway. Annu Rev Biochem. 2022;91:599–628.

    Article  CAS  PubMed  Google Scholar 

  20. Skopelja-Gardner S, An J, Elkon KB. Role of the cGAS-STING pathway in systemic and organ-specific diseases. Nat Rev Nephrol. 2022;18:558–72.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Daeron M, Jaeger S, Du Pasquier L, Vivier E. Immunoreceptor tyrosine-based inhibition motifs: a quest in the past and future. Immunol Rev. 2008;224:11–43.

    Article  CAS  PubMed  Google Scholar 

  22. Barrow AD, Trowsdale J. You say ITAM and I say ITIM, let’s call the whole thing off: the ambiguity of immunoreceptor signalling. Eur J Immunol. 2006;36:1646–53.

    Article  CAS  PubMed  Google Scholar 

  23. Neel BG, Gu H, Pao L. The ‘Shp’ing news: SH2 domain-containing tyrosine phosphatases in cell signaling. Trends Biochem Sci. 2003;28:284–93.

    Article  CAS  PubMed  Google Scholar 

  24. Zhang J, Somani AK, Siminovitch KA. Roles of the SHP-1 tyrosine phosphatase in the negative regulation of cell signalling. Semin Immunol. 2000;12:361–78.

    Article  CAS  PubMed  Google Scholar 

  25. Burke JE, Triscott J, Emerling BM, Hammond GRV. Beyond PI3Ks: targeting phosphoinositide kinases in disease. Nat Rev Drug Discov. 2022;22:357–86.

  26. Vasan N, Cantley LC. At a crossroads: how to translate the roles of PI3K in oncogenic and metabolic signalling into improvements in cancer therapy. Nat Rev Clin Oncol. 2022;19:471–85.

    Article  CAS  PubMed  Google Scholar 

  27. Cuevas BD, Lu Y, Mao M, Zhang J, LaPushin R, Siminovitch K, et al. Tyrosine phosphorylation of p85 relieves its inhibitory activity on phosphatidylinositol 3-kinase. J Biol Chem. 2001;276:27455–61.

    Article  CAS  PubMed  Google Scholar 

  28. Wen LZ, Ding K, Wang ZR, Ding CH, Lei SJ, Liu JP, et al. SHP-1 acts as a tumor suppressor in hepatocarcinogenesis and HCC progression. Cancer Res. 2018;78:4680–91.

    Article  CAS  PubMed  Google Scholar 

  29. Liu X, Dong M, Yao Y, Wang Y, Mao J, Hu L, et al. A tyrosine phosphoproteome analysis approach enabled by selective dephosphorylation with protein tyrosine phosphatase. Anal Chem. 2022;94:4155–64.

    Article  CAS  PubMed  Google Scholar 

  30. Dutta R, Mahato RI. Recent advances in hepatocellular carcinoma therapy. Pharmacol Ther. 2017;173:106–17.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Llovet JM, Ricci S, Mazzaferro V, Hilgard P, Gane E, Blanc JF, et al. Sorafenib in advanced hepatocellular carcinoma. New Engl J Med. 2008;359:378–90.

    Article  CAS  PubMed  Google Scholar 

  32. Galmiche A, Chauffert B, Barbare JC. New biological perspectives for the improvement of the efficacy of sorafenib in hepatocellular carcinoma. Cancer Lett. 2014;346:159–62.

    Article  CAS  PubMed  Google Scholar 

  33. Chen KF, Chen HL, Tai WT, Feng WC, Hsu CH, Chen PJ, et al. Activation of phosphatidylinositol 3-kinase/Akt signaling pathway mediates acquired resistance to sorafenib in hepatocellular carcinoma cells. J Pharmacol Exp Ther. 2011;337:155–61.

    Article  CAS  PubMed  Google Scholar 

  34. Zhang H, Wang Q, Liu J, Cao H. Inhibition of the PI3K/Akt signaling pathway reverses sorafenib-derived chemo-resistance in hepatocellular carcinoma. Oncol Lett. 2018;15:9377–84.

    PubMed  PubMed Central  Google Scholar 

  35. Harding JJ, Nandakumar S, Armenia J, Khalil DN, Albano M, Ly M, et al. Prospective genotyping of hepatocellular carcinoma: clinical implications of next-generation sequencing for matching patients to targeted and immune therapies. Clin Cancer Res. 2019;25:2116–26.

    Article  CAS  PubMed  Google Scholar 

  36. Tang W, Chen Z, Zhang W, Cheng Y, Zhang B, Wu F, et al. The mechanisms of sorafenib resistance in hepatocellular carcinoma: theoretical basis and therapeutic aspects. Signal Transduct Target Ther. 2020;5:87.

    Article  PubMed  PubMed Central  Google Scholar 

  37. Cheng Z, Dai T, He X, Zhang Z, Xie F, Wang S, et al. The interactions between cGAS-STING pathway and pathogens. Signal Transduct Target Ther. 2020;5:91.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Wang X, Liu Y, Xue C, Hu Y, Zhao Y, Cai K, et al. A protein-based cGAS-STING nanoagonist enhances T cell-mediated anti-tumor immune responses. Nat Commun. 2022;13:5685.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Zhao X, Hu S, Zeng L, Liu X, Song Y, Zhang Y, et al. Irradiation combined with PD-L1(-/-) and autophagy inhibition enhances the antitumor effect of lung cancer via cGAS-STING-mediated T cell activation. iScience. 2022;25:104690.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Wang H, Hu S, Chen X, Shi H, Chen C, Sun L, et al. cGAS is essential for the antitumor effect of immune checkpoint blockade. Proc Natl Acad Sci USA. 2017;114:1637–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Li T, Chen ZJ. The cGAS-cGAMP-STING pathway connects DNA damage to inflammation, senescence, and cancer. J Exp Med. 2018;215:1287–99.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Nassour J, Radford R, Correia A, Fuste JM, Schoell B, Jauch A, et al. Autophagic cell death restricts chromosomal instability during replicative crisis. Nature. 2019;565:659–63.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Verrier ER, Yim SA, Heydmann L, El Saghire H, Bach C, Turon-Lagot V, et al. Hepatitis B virus evasion from cyclic guanosine monophosphate-adenosine monophosphate synthase sensing in human hepatocytes. Hepatology. 2018;68:1695–709.

    Article  CAS  PubMed  Google Scholar 

  44. Cuevas B, Lu Y, Watt S, Kumar R, Zhang J, Siminovitch KA, et al. SHP-1 regulates Lck-induced phosphatidylinositol 3-kinase phosphorylation and activity. J Biol Chem. 1999;274:27583–9.

    Article  CAS  PubMed  Google Scholar 

  45. Liu GY, Sabatini DM. mTOR at the nexus of nutrition, growth, ageing and disease. Nat Rev Mol Cell Biol. 2020;21:183–203.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Melnik BC. Dairy consumption and hepatocellular carcinoma risk. Ann Transl Med. 2021;9:736.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Guo P, Ma X, Zhao W, Huai W, Li T, Qiu Y, et al. TRIM31 is upregulated in hepatocellular carcinoma and promotes disease progression by inducing ubiquitination of TSC1-TSC2 complex. Oncogene. 2018;37:478–88.

    Article  CAS  PubMed  Google Scholar 

  48. Ou Q, Yu Y, Li A, Chen J, Yu T, Xu X, et al. Association of survival and genomic mutation signature with immunotherapy in patients with hepatocellular carcinoma. Ann Transl Med. 2020;8:230.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Chen J, Jin R, Zhao J, Liu J, Ying H, Yan H, et al. Potential molecular, cellular and microenvironmental mechanism of sorafenib resistance in hepatocellular carcinoma. Cancer Lett. 2015;367:1–11.

    Article  CAS  PubMed  Google Scholar 

  50. Sun Y, Zhang H, Meng J, Guo F, Ren D, Wu H, et al. S-palmitoylation of PCSK9 induces sorafenib resistance in liver cancer by activating the PI3K/AKT pathway. Cell Rep. 2022;40:111194.

    Article  CAS  PubMed  Google Scholar 

  51. Wei Q, Mu K, Li T, Zhang Y, Yang Z, Jia X, et al. Deregulation of the NLRP3 inflammasome in hepatic parenchymal cells during liver cancer progression. Lab Investig. 2014;94:52–62.

    Article  CAS  PubMed  Google Scholar 

  52. Ma D, Yang M, Wang Q, Sun C, Shi H, Jing W, et al. Arginine methyltransferase PRMT5 negatively regulates cGAS-mediated antiviral immune response. Sci Adv. 2021;7:eabc1834.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Zhang X, Coker OO, Chu ES, Fu K, Lau HCH, Wang YX, et al. Dietary cholesterol drives fatty liver-associated liver cancer by modulating gut microbiota and metabolites. Gut. 2021;70:761–74.

    Article  CAS  PubMed  Google Scholar 

  54. Liu S, Dai J, Lan X, Fan B, Dong T, Zhang Y, et al. Intestinal bacteria are potential biomarkers and therapeutic targets for gastric cancer. Microb Pathog. 2021;151:104747.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors thank Dr. Q Sun (Sun Yat-sen University, China) for SHP1 plasmid, Dr. X Yang (Shandong University, China) for assisting with structural and sequences analysis of cGAS, Dr. D Yuan (Shandong University, China) and Dr. H Wen (Ohio State University, USA) for constructive discussion about this work. The authors thank the Translational Medicine Core Facility of Shandong University for consultation and instrument availability that supported this work.

Funding

This study is funded by National Natural Science Foundation of China (Nos. 81972275, 82171748, and 32200709); Distinguished Professor of Taishan Scholars (No. tstp20221109), Shandong Provincial Natural Science Foundation Joint Fund (No. ZR202306180008), and the Major Innovation Project of Shandong Province (No. 2021GXGC011305).

Author information

Authors and Affiliations

Authors

Contributions

DM and LH designed this study, analyzed the data, and wrote the manuscript. DM performed most of the experiments. MY assisted with the cell experiment. CS assisted with the confocal microscopy capture. XC, HC, and SL assisted with the animal experiments and related assay. XG assisted with the animal experiments. QW, WJ, XL, TL, and YZ assisted with the experiments and provided technical help. All authors read the final version of the manuscript and approved the submission.

Corresponding author

Correspondence to Lihui Han.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary information

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

Ma, D., Yang, M., Sun, C. et al. cGAS suppresses hepatocellular carcinoma independent of its cGAMP synthase activity. Cell Death Differ (2024). https://doi.org/10.1038/s41418-024-01291-9

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1038/s41418-024-01291-9

Search

Quick links