Abstract
Hepatocellular carcinoma (HCC) is a highly heterogeneous solid tumor, with its biological characteristics intricately linked to the activation of oncogenes. This research specifically explored CCDC137, a molecule within the CCDC family exhibiting the closest association with HCC. Our investigation aimed to unravel the role, underlying mechanisms, and potential therapeutic implications of CCDC137 in the context of HCC. We observed a close correlation between elevated CCDC137 expression and poor prognosis in HCC patients, along with a promotive effect on HCC progression in vitro and in vivo. Mechanistically, we identified LZTS2, a negative regulator of β-catenin, as the binding protein of CCDC137. CCDC137 facilitated K48-linked poly-ubiquitination of LZTS2 at lysine 467 via recruiting E3 ubiquitin ligase β-TrCP in the nucleus, triggering AKT phosphorylation and activation of β-catenin pathway. Moreover, the 1-75 ___domain of CCDC137 was responsible for the formation of the CCDC137-LZTS2-β-TrCP complex. Subsequently, designed peptides targeting the 1-75 ___domain of CCDC137 to disrupt CCDC137-LZTS2 interaction demonstrated efficacy in inhibiting HCC progression. This promising outcome was further supported by HCC organoids and patient-derived xenograft (PDX) models, underscoring the potential clinical utility of the peptides. This study elucidated the mechanism of the CCDC137-LZTS2-β-TrCP protein complex in HCC and offered clinically significant therapeutic strategies targeting this complex.
This is a preview of subscription content, access via your institution
Access options
Subscribe to this journal
Receive 12 print issues and online access
269,00 € per year
only 22,42 € per issue
Buy this article
- Purchase on SpringerLink
- Instant access to full article PDF
Prices may be subject to local taxes which are calculated during checkout








Similar content being viewed by others
Data availability
The data supporting the findings of this study are available upon reasonable request to the corresponding author.
References
Llovet JM, Kelley RK, Villanueva A, Singal AG, Pikarsky E, Roayaie S, et al. Hepatocellular carcinoma. Nat Rev Dis Primers. 2021;7:6.
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.
Katyal S, Oliver JH 3rd, Peterson MS, Ferris JV, Carr BS, Baron RL. Extrahepatic metastases of hepatocellular carcinoma. Radiology. 2000;216:698–703.
Furuta M, Ueno M, Fujimoto A, Hayami S, Yasukawa S, Kojima F, et al. Whole genome sequencing discriminates hepatocellular carcinoma with intrahepatic metastasis from multi-centric tumors. J Hepatol. 2017;66:363–73.
Finn RS, Qin S, Ikeda M, Galle PR, Ducreux M, Kim TY, et al. Atezolizumab plus Bevacizumab in unresectable hepatocellular carcinoma. N Engl J Med. 2020;382:1894–905.
Yang C, Zhang H, Zhang L, Zhu AX, Bernards R, Qin W, et al. Evolving therapeutic landscape of advanced hepatocellular carcinoma. Nat Rev Gastroenterol Hepatol. 2023;20:203–22.
Valastyan S, Weinberg RA. Tumor metastasis: molecular insights and evolving paradigms. Cell. 2011;147:275–92.
Gerstberger S, Jiang Q, Ganesh K. Metastasis. Cell. 2023;186:1564–79.
Fidler IJ. The pathogenesis of cancer metastasis: the ‘seed and soil’ hypothesis revisited. Nat Rev Cancer. 2003;3:453–8.
Jiang WG, Sanders AJ, Katoh M, Ungefroren H, Gieseler F, Prince M, et al. Tissue invasion and metastasis: Molecular, biological and clinical perspectives. Semin Cancer Biol. 2015;35:S244–S275.
Dimri M, Humphries A, Laknaur A, Elattar S, Lee TJ, Sharma A, et al. NAD(P)H Quinone dehydrogenase 1 ablation inhibits activation of the phosphoinositide 3-kinase/akt serine/threonine kinase and mitogen-activated protein kinase/extracellular signal-regulated kinase pathways and blocks metabolic adaptation in hepatocellular carcinoma. Hepatology. 2020;71:549–68.
Zuo Q, He J, Zhang S, Wang H, Jin G, Jin H, et al. PPARgamma coactivator-1alpha suppresses metastasis of hepatocellular carcinoma by inhibiting Warburg effect by PPARgamma-dependent WNT/beta-catenin/pyruvate dehydrogenase kinase isozyme 1 axis. Hepatology. 2021;73:644–60.
Loesch R, Caruso S, Paradis V, Godard C, Gougelet A, Renault G, et al. Deleting the beta-catenin degradation ___domain in mouse hepatocytes drives hepatocellular carcinoma or hepatoblastoma-like tumor growth. J Hepatol. 2022;77:424–35.
Baik SH, Lee J, Lee YS, Jang JY, Kim CW. ANT2 shRNA downregulates miR-19a and miR-96 through the PI3K/Akt pathway and suppresses tumor growth in hepatocellular carcinoma cells. Exp Mol Med. 2016;48:e222.
Vogelstein B, Papadopoulos N, Velculescu VE, Zhou S, Diaz LA Jr, Kinzler KW. Cancer genome landscapes. Science. 2013;339:1546–58.
Liu Z, Yan W, Liu S, Liu Z, Xu P, Fang W. Regulatory network and targeted interventions for CCDC family in tumor pathogenesis. Cancer Lett. 2023;565:216225.
Guo L, Li B, Lu Z, Liang H, Yang H, Chen Y, et al. CCDC137 is a prognostic biomarker and correlates with immunosuppressive tumor microenvironment based on pan-cancer analysis. Front Mol Biosci. 2021;8:674863.
Dai W, Wu J, Peng X, Hou W, Huang H, Cheng Q, et al. CDK12 orchestrates super-enhancer-associated CCDC137 transcription to direct hepatic metastasis in colorectal cancer. Clin Transl Med. 2022;12:e1087.
Tao S, Xie SJ, Diao LT, Lv G, Hou YR, Hu YX, et al. RNA-binding protein CCDC137 activates AKT signaling and promotes hepatocellular carcinoma through a novel non-canonical role of DGCR8 in mRNA localization. J Exp Clin Cancer Res. 2023;42:194.
Lu Y, Li X, Liu H, Xue J, Zeng Z, Dong X, et al. beta-Trcp and CK1delta-mediated degradation of LZTS2 activates PI3K/AKT signaling to drive tumorigenesis and metastasis in hepatocellular carcinoma. Oncogene. 2021;40:1269–83.
Hu X, Zhao Y, Wei L, Zhu B, Song D, Wang J, et al. CCDC178 promotes hepatocellular carcinoma metastasis through modulation of anoikis. Oncogene. 2017;36:4047–59.
Cheng X, Wang Y, Gong Y, Li F, Guo Y, Hu S, et al. Structural basis of FYCO1 and MAP1LC3A interaction reveals a novel binding mode for Atg8-family proteins. Autophagy. 2016;12:1330–9.
Wang H, Zhang CZ, Lu SX, Zhang MF, Liu LL, Luo RZ, et al. A coiled-coil ___domain containing 50 splice variant is modulated by serine/arginine-rich splicing factor 3 and promotes hepatocellular carcinoma in mice by the ras signaling pathway. Hepatology. 2019;69:179–95.
Cabeza-Arvelaiz Y, Thompson TC, Sepulveda JL, Chinault AC. LAPSER1: a novel candidate tumor suppressor gene from 10q24.3. Oncogene. 2001;20:6707–17.
Thyssen G, Li TH, Lehmann L, Zhuo M, Sharma M, Sun Z. LZTS2 is a novel beta-catenin-interacting protein and regulates the nuclear export of beta-catenin. Mol Cell Biol. 2006;26:8857–67.
Xu S, Li Y, Lu Y, Huang J, Ren J, Zhang S, et al. LZTS2 inhibits PI3K/AKT activation and radioresistance in nasopharyngeal carcinoma by interacting with p85. Cancer Lett. 2018;420:38–48.
Hua F, Shang S, Yang YW, Zhang HZ, Xu TL, Yu JJ, et al. TRIB3 interacts with beta-catenin and TCF4 to increase stem cell features of colorectal cancer stem cells and tumorigenesis. Gastroenterology. 2019;156:708–721 e715.
Xu C, Xu Z, Zhang Y, Evert M, Calvisi DF, Chen X. beta-Catenin signaling in hepatocellular carcinoma. J Clin Invest. 2022;132:e154515.
Wang Q, Chen X, Hay N. Akt as a target for cancer therapy: more is not always better (lessons from studies in mice). Br J Cancer. 2017;117:159–63.
Rogel A, Willoughby JE, Buchan SL, Leonard HJ, Thirdborough SM, Al-Shamkhani A. Akt signaling is critical for memory CD8(+) T-cell development and tumor immune surveillance. Proc Natl Acad Sci USA. 2017;114:E1178–E1187.
Ruiz de Galarreta M, Bresnahan E, Molina-Sanchez P, Lindblad KE, Maier B, Sia D, et al. beta-Catenin activation promotes immune escape and resistance to anti-PD-1 therapy in hepatocellular carcinoma. Cancer Discov. 2019;9:1124–41.
Acknowledgements
We express our gratitude to Wang Guangxin from the Institute of Hydrobiology, Chinese Academy of Sciences, for providing technical support in confocal microscopy imaging. Additionally, we extend our thanks to Professor Li Yan from Zhongnan Hospital of Wuhan University, Wuhan, China, for generously supplying HCCLM9 cells.
Funding
This work was supported by the National Natural Science Foundation of China (82372994, 82003003, 81874189), Wuhan Science and Technology Bureau 2022 annual key research and development project (2022023502015182), 2021 Wuhan Young and Middle-aged Medical Backbone Talent Training Project (2021ZQNYX008).
Author information
Authors and Affiliations
Contributions
Jin Chen and Bixiang Zhang conceived and designed the study. Lei Xu, Qiumeng Liu and Hailing Liu performed the experiments. Shiwei Yue, Feimu Fan, Pengcheng Li and Jie Mo collected the clinical specimens and data. Zhicheng Liu, Renshun Dong and Xuewu Zhang performed the statistical analysis. Lei Xu drafted the manuscript. Hanhua Dong, Huifang Liang, Lin Chen, Bixiang Zhang and Jin Chen contributed to the critical revision of the paper. All authors read and approved the final manuscript.
Corresponding authors
Ethics declarations
Competing interests
The authors declare no competing interests.
Ethics approval
The utilization of human tissue samples in this study has been approved by the Ethics Committee of Tongji Hospital, and informed consent exemption was approved by the institutional ethics committee (TJ-IRB20211163). All animal experiments involved in this study were approved by the Ethics Committee of Tongji Hospital (TJ-202304015).
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.
About this article
Cite this article
Xu, L., Liu, Q., Liu, H. et al. Disrupting CCDC137-mediated LZTS2 and β-TrCP interaction in the nucleus inhibits hepatocellular carcinoma development via β-catenin and AKT. Cell Death Differ 32, 134–148 (2025). https://doi.org/10.1038/s41418-024-01328-z
Received:
Revised:
Accepted:
Published:
Issue Date:
DOI: https://doi.org/10.1038/s41418-024-01328-z