Abstract
Background
Vagal innervation plays a pivotal role in gastric tumorigenesis and tumor progression. However, the upstream signaling regulating acetylcholine receptors (AChRs) and its contribution to carcinogenesis remains largely elusive.
Methods
We constructed Trp53−/−; Cdh1−/− mouse gastric organoids to recapitulate the morphological and functional characteristics of diffuse-type gastric cancer (DGC) for FDA-approved drug (1464 compounds) screening. We investigated the effects of AChR inhibitors in diffuse-type patient-derived organoids (PDOs) by examining IC50 and xenograft tumorigenesis. Mass spectrometry and Co-immunoprecipitation was used to identify the interaction between cholinergic receptor muscarinic 3 (CHRM3) and PJA2. Ubiquitination and degradation assays were used to explore the regulation of CHRM3 by PJA2. 120 human gastric cancer specimens and GEO database were used to explore the clinical relevance of PJA2-CHRM3 signaling.
Results
mAChR inhibitors were identified as the most effective at suppressing Trp53−/−; Cdh1−/− organoids. Consistently, the viability of PDOs with aberrant expression of CHRM3 can be significantly inhibited by oxybutynin hydrochloride (OXY) and nortriptyline hydrochloride (NOR). Mechanistically, the RING E3 ligase PJA2 ubiquitinates and degrades CHRM3, subsequently suppressing downstream TGFβ-pSMAD3 signaling and tumor cell progression. Clinically, low PJA2 expression was correlated with high CHRM3, p-SMAD3 and choline acetyltransferase (ChAT) expression and predicted poor outcomes. Biologically, PJA2 but not its catalytically dead ΔRING mutant could suppress PDOs with aberrant CHRM3 signaling. PJA2 depletion enhanced tumor metastasis of gastric cancer cells, subsequently reversed by OXY and NOR treatment.
Conclusions
This previously unknown PJA2-CHRM3 signaling axis provides further understanding of cholinergic innervation as well as identifies a new therapeutic vulnerability in DGC.
This is a preview of subscription content, access via your institution
Access options
Subscribe to this journal
Receive 24 print issues and online access
$259.00 per year
only $10.79 per issue
Buy this article
- Purchase on SpringerLink
- Instant access to the full article PDF.
USD 39.95
Prices may be subject to local taxes which are calculated during checkout







Similar content being viewed by others
Data availability
The results displayed here are partially based on data generated by The Cancer Genome Atlas (TCGA) Research Network (https://www.cancer.gov/tcga), Asian Cancer Research Group (ACRG) database (https://consortiapedia.fastercures.org/consortia/acrg/) and Chinese Glioma Genome Atlas (CGGA) database (http://www.cgga.org.cn/). The publicly available dataset reused in this study, including Kumar et al.’ dataset (11 NT and 29 PT samples), was retrieved from GSE183904 (PubMed ID: 34642171). Kaplan–Meier Plotter (https://kmplot.com/analysis/) were used for survival analysis of patients with gastric cancer by survival curves tests. The RNA-Seq data has been uploaded to the Sequence Read Archive (SRA), BioProject ID: PRJNA1073869. All the data supporting the findings of this study are available from the corresponding authors upon reasonable request.
References
Zahalka AH, Frenette PS. Nerves in cancer. Nat Rev Cancer. 2020;20:143–57.
Senga SS, Grose RP. Hallmarks of cancer-the new testament. Open Biol. 2021;11:200358.
Zhao CM, Hayakawa Y, Kodama Y, Muthupalani S, Westphalen CB, Andersen GT, et al. Denervation suppresses gastric tumorigenesis. Sci Transl Med. 2014;6:250ra115.
Magnon C, Hall SJ, Lin J, Xue X, Gerber L, Freedland SJ, et al. Autonomic nerve development contributes to prostate cancer progression. Science. 2013;341:1236361.
Ayala GE, Dai H, Powell M, Li R, Ding Y, Wheeler TM, et al. Cancer-related axonogenesis and neurogenesis in prostate cancer. Clin Cancer Res. 2008;14:7593–603.
Albo D, Akay CL, Marshall CL, Wilks JA, Verstovsek G, Liu H, et al. Neurogenesis in colorectal cancer is a marker of aggressive tumor behavior and poor outcomes. Cancer. 2011;117:4834–45.
Demir IE, Friess H, Ceyhan GO. Neural plasticity in pancreatitis and pancreatic cancer. Nat Rev Gastroenterol Hepatol. 2015;12:649–59.
Bressy C, Lac S, Nigri J, Leca J, Roques J, Lavaut MN, et al. LIF drives neural remodeling in pancreatic cancer and offers a new candidate biomarker. Cancer Res. 2018;78:909–21.
You H, Shang W, Min X, Weinreb J, Li Q, Leapman M, et al. Sight and switch off: nerve density visualization for interventions targeting nerves in prostate cancer. Sci Adv. 2020;6:eaax6040.
Hanahan D, Monje M. Cancer hallmarks intersect with neuroscience in the tumor microenvironment. Cancer Cell. 2023;41:573–80.
Cassaro M, Rugge M, Gutierrez O, Leandro G, Graham DY, Genta RM. Topographic patterns of intestinal metaplasia and gastric cancer. Am J Gastroenterol. 2000;95:1431–8.
Vaes N, Idris M, Boesmans W, Alves MM, Melotte V. Nerves in gastrointestinal cancer: from mechanism to modulations. Nat Rev Gastroenterol Hepatol. 2022;19:768–84.
Bedine MS. Textbook of gastroenterology. Gastroenterology. 2000;118:984–5.
Xie G, Drachenberg C, Yamada M, Wess J, Raufman JP. Cholinergic agonist-induced pepsinogen secretion from murine gastric chief cells is mediated by M1 and M3 muscarinic receptors. Am J Physiol Gastrointest Liver Physiol. 2005;289:G521–G9.
Wang K, Zhao XH, Liu J, Zhang R, Li JP. Nervous system and gastric cancer. Biochim Biophys Acta Rev Cancer. 2020;1873:188313.
Bahmanyar S, Ye W, Dickman PW, Nyrén O. Long-term risk of gastric cancer by subsite in operated and unoperated patients hospitalized for peptic ulcer. Am J Gastroenterol. 2007;102:1185–91.
Wang N, Yao M, Xu J, Quan Y, Zhang K, Yang R, et al. Autocrine activation of CHRM3 promotes prostate cancer growth and castration resistance via CaM/CaMKK-mediated phosphorylation of Akt. Clin Cancer Res. 2015;21:4676–85.
Raufman J-P, Samimi R, Shah N, Khurana S, Shant J, Drachenberg C, et al. Genetic ablation of M3 muscarinic receptors attenuates murine colon epithelial cell proliferation and neoplasia. Cancer Res. 2008;68:3573–8.
Yan HHN, Siu HC, Law S, Ho SL, Yue SSK, Tsui WY, et al. A comprehensive human gastric cancer organoid biobank captures tumor subtype heterogeneity and enables therapeutic screening. Cell Stem Cell. 2018;23:882–97.
Lo YH, Kolahi KS, Du Y, Chang CY, Krokhotin A, Nair A, et al. A CRISPR/Cas9-engineered ARID1A-deficient human gastric cancer organoid model reveals essential and non-essential modes of oncogenic transformation. Cancer Discov. 2021;11:1562–81.
Li H, Fu X, Zhao J, Li C, Li L, Xia P, et al. EXOC4 promotes diffuse-type gastric cancer metastasis via activating FAK signal. Mol Cancer Res. 2022;20:1021–34.
Huang L, Zhao J, Chen H, Wan L, Inuzuka H, Guo J, et al. SCFFBW7-mediated degradation of Brg1 suppresses gastric cancer metastasis. Nat Commun. 2018;9:3569.
Zhao J, Li H, Min L, Han X, Shu P, Yang Y, et al. High expression of tumor necrosis factor receptor-associated factor 2 promotes tumor metastasis and is associated with unfavorable prognosis in gastric cancer. J Gastroenterol Hepatol. 2018;33:431–42.
Bass AJ, Thorsson V, Shmulevich I, Reynolds SM, Miller M, Bernard B, et al. Comprehensive molecular characterization of gastric adenocarcinoma. Nature. 2014;513:202–9.
Gravalos C, Jimeno A. HER2 in gastric cancer: a new prognostic factor and a novel therapeutic target. Ann Oncol. 2008;19:1523–9.
Bang YJ, Van Cutsem E, Feyereislova A, Chung HC, Shen L, Sawaki A, et al. Trastuzumab in combination with chemotherapy versus chemotherapy alone for treatment of HER2-positive advanced gastric or gastro-oesophageal junction cancer (ToGA): a phase 3, open-label, randomised controlled trial. Lancet. 2010;376:687–97.
Antman EM. Evaluating the cardiovascular safety of nonsteroidal anti-inflammatory drugs. Circulation. 2017;135:2062–72.
Hayakawa Y, Sakitani K, Konishi M, Asfaha S, Niikura R, Tomita H, et al. Nerve growth factor promotes gastric tumorigenesis through aberrant cholinergic signaling. Cancer Cell. 2017;31:21–34.
Wang L, Xu J, Xia Y, Yin K, Li Z, Li B, et al. Muscarinic acetylcholine receptor 3 mediates vagus nerve-induced gastric cancer. Oncogenesis. 2018;7:88.
Oki T, Kageyama A, Takagi Y, Uchida S, Yamada S. Comparative evaluation of central muscarinic receptor binding activity by oxybutynin, tolterodine and darifenacin used to treat overactive bladder. J Urol. 2007;177:766–70.
Maruyama S, Oki T, Otsuka A, Shinbo H, Ozono S, Kageyama S, et al. Human muscarinic receptor binding characteristics of antimuscarinic agents to treat overactive bladder. J Urol. 2006;175:365–9.
Mannan Baig A, Khan NA, Effendi V, Rana Z, Ahmad HR, Abbas F. Differential receptor dependencies: expression and significance of muscarinic M1 receptors in the biology of prostate cancer. Anticancer Drugs. 2017;28:75–87.
Gong M, Ye S, Li WX, Zhang J, Liu Y, Zhu J, et al. Regulatory function of praja ring finger ubiquitin ligase 2 mediated by the P2rx3/P2rx7 axis in mouse hippocampal neuronal cells. Am J Physiol Cell Physiol. 2020;318:C1123–C35.
Lipkowitz S, Weissman AM. RINGs of good and evil: RING finger ubiquitin ligases at the crossroads of tumour suppression and oncogenesis. Nat Rev Cancer. 2011;11:629–43.
Zheng L, Xu H, Di Y, Chen L, Liu J, Kang L, et al. ELK4 promotes the development of gastric cancer by inducing M2 polarization of macrophages through regulation of the KDM5A-PJA2-KSR1 axis. J Transl Med. 2021;19:342.
Zhao Z, Zhu L, Xing Y, Zhang Z. Praja2 suppresses the growth of gastric cancer by ubiquitylation of KSR1 and inhibiting MEK-ERK signal pathways. Aging. 2021;13:3886–97.
Rinaldi L, Delle Donne R, Sepe M, Porpora M, Garbi C, Chiuso F, et al. praja2 regulates KSR1 stability and mitogenic signaling. Cell Death Dis. 2016;7:e2230.
Lignitto L, Arcella A, Sepe M, Rinaldi L, Delle Donne R, Gallo A, et al. Proteolysis of MOB1 by the ubiquitin ligase praja2 attenuates Hippo signalling and supports glioblastoma growth. Nat Commun. 2013;4:1822.
Lignitto L, Carlucci A, Sepe M, Stefan E, Cuomo O, Nisticò R, et al. Control of PKA stability and signalling by the RING ligase praja2. Nat Cell Biol. 2011;13:412–22.
Mosser VA, Jones KT, Hoffman KM, McCarty NA, Jackson DA. Differential role of beta-arrestin ubiquitination in agonist-promoted down-regulation of M1 vs M2 muscarinic acetylcholine receptors. J Mol Signal. 2008;3:20.
Wang L, Zhi X, Zhang Q, Wei S, Li Z, Zhou J, et al. Muscarinic receptor M3 mediates cell proliferation induced by acetylcholine and contributes to apoptosis in gastric cancer. Tumour Biol. 2016;37:2105–17.
Ding S, Hsu C, Wang Z, Natesh NR, Millen R, Negrete M, et al. Patient-derived micro-organospheres enable clinical precision oncology. Cell Stem Cell. 2022;29:905–17.
Boj SF, Hwang CI, Baker LA, Chio II, Engle DD, Corbo V, et al. Organoid models of human and mouse ductal pancreatic cancer. Cell. 2015;160:324–38.
Kim M, Mun H, Sung CO, Cho EJ, Jeon HJ, Chun SM, et al. Patient-derived lung cancer organoids as in vitro cancer models for therapeutic screening. Nat Commun. 2019;10:3991.
Tatler AL, John AE, Jolly L, Habgood A, Porte J, Brightling C, et al. Integrin αvβ5-mediated TGF-β activation by airway smooth muscle cells in asthma. J Immunol. 2011;187:6094–107.
Oenema TA, Smit M, Smedinga L, Racké K, Halayko AJ, Meurs H, et al. Muscarinic receptor stimulation augments TGF-β1-induced contractile protein expression by airway smooth muscle cells. Am J Physiol Lung Cell Mol Physiol. 2012;303:L589–L97.
Jenkins RG. Ligation of protease-activated receptor 1 enhances αvβ6 integrin-dependent TGFβ-activation and promotes acute lung injury. J Clin Invest. 2006;116:1606–14.
Lu JJ, Xu GN, Yu P, Song Y, Wang XL, Zhu L, et al. The activation of M3 mAChR in airway epithelial cells promotes IL-8 and TGF-β1 secretion and airway smooth muscle cell migration. Respir Res. 2016;17:25.
Mohamed R, Shajimoon A, Afroz R, Gabr M, Thomas WG, Little PJ, et al. Akt acts as a switch for GPCR transactivation of the TGF‐β receptor type 1. FEBS J. 2021;289:2642–56.
Acknowledgements
We thank Dr. Rongkui Luo (Department of Pathology, Gastric Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China) for his kind help in evaluation, IHC staining, and Lauren classification of tissue microarrays. This work was supported by the National Natural Science Foundation of China to LYH (82472666, 82272994 and 82073098), and the Interdisciplinary Program of Shanghai Jiao Tong University (YG2022QN086 and YG2025QNB47).
Author information
Authors and Affiliations
Contributions
LYH designed and supervised the study; LML and JYY performed most of the experiments; LZ, WSL, YNN, LHW and GXF helped some biochemical experiments and data curation; DLL helped some animal experiments; LYH, LML and JYY wrote the manuscript; JJZ, YG, ZMW, GQH and XFW were responsible for writing–review and editing. All authors read and approved the final manuscript.
Corresponding author
Ethics declarations
Competing interests
The authors declare no competing interests.
Ethics approval and consent to participate
The animal studies were approved by the Institutional Animal Care and Use Committee (IACUC) of Shanghai Jiao Tong University (Approval Number: 20221035). This study, involving human participants, was approved by the Ethics Committee of Fudan University (Approval Number: B2021-449R) and was carried out in accordance with the ethical standards stipulated in the 1964 Declaration of Helsinki. Each patient receives written informed consent.
Additional information
Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
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
Li, L., Yin, J., Zhang, L. et al. Aberrant PJA2-CHRM3 signaling creates a therapeutic vulnerability in gastric tumor. Br J Cancer 133, 1122–1136 (2025). https://doi.org/10.1038/s41416-025-03145-8
Received:
Revised:
Accepted:
Published:
Version of record:
Issue date:
DOI: https://doi.org/10.1038/s41416-025-03145-8


