Abstract
Pancreatic ductal adenocarcinoma (PDAC) has an atypical, highly stromal tumour microenvironment (TME) that profoundly contributes to its poor prognosis1. Here, to better understand the intercellular signalling between cancer and stromal cells directly in PDAC tumours, we developed a multidimensional proteomic strategy called TMEPro. We applied TMEPro to profile the glycosylated secreted and plasma membrane proteome of 100 human pancreatic tissue samples to a great depth, define cell type origins and identify potential paracrine cross-talk, especially that mediated through tyrosine phosphorylation. Temporal dynamics during pancreatic tumour progression were investigated in a genetically engineered PDAC mouse model. Functionally, we revealed reciprocal signalling between stromal cells and cancer cells mediated by the stromal PDGFR–PTPN11–FOS signalling axis. Furthermore, we examined the generic shedding mechanism of plasma membrane proteins in PDAC tumours and revealed that matrix-metalloprotease-mediated shedding of the AXL receptor tyrosine kinase ectodomain provides an additional dimension of intercellular signalling regulation in the PDAC TME. Importantly, the level of shed AXL has a potential correlation with lymph node metastasis, and inhibition of AXL shedding and its kinase activity showed a substantial synergistic effect in inhibiting cancer cell growth. In summary, we provide TMEPro, a generically applicable clinical functional proteomic strategy, and a comprehensive resource for better understanding the PDAC TME and facilitating the discovery of new diagnostic and therapeutic targets.
This is a preview of subscription content, access via your institution
Access options
Access Nature and 54 other Nature Portfolio journals
Get Nature+, our best-value online-access subscription
27,99 € / 30 days
cancel any time
Subscribe to this journal
Receive 51 print issues and online access
199,00 € per year
only 3,90 € 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 MS proteomics data have been deposited to the ProteomeXchange Consortium via the PRIDE69 partner repository under dataset identifier PXD048644. The UniProt human and mouse proteome databases are available online (https://www.uniprot.org/). All other data supporting the findings of this study are available within the Article and its Supplementary Information. Source data are provided with this paper.
Code availability
No new algorithms were developed for this Article. Scripts to reproduce the figures are available from the corresponding authors on reasonable request.
References
Ho, W. J., Jaffee, E. M. & Zheng, L. The tumour microenvironment in pancreatic cancer—clinical challenges and opportunities. Nat. Rev. Clin. Oncol. 17, 527–540 (2020).
Siegel, R. L., Miller, K. D., Fuchs, H. E. & Jemal, A. Cancer statistics, 2021. CA Cancer J. Clin. 71, 7–33 (2021).
Siegel, R. L., Giaquinto, A. N. & Jemal, A. Cancer statistics, 2024. CA Cancer J. Clin. 74, 12–49 (2024).
Kleeff, J. et al. Pancreatic cancer. Nat. Rev. Dis. Primers 2, 16022 (2016).
Cao, L. et al. Proteogenomic characterization of pancreatic ductal adenocarcinoma. Cell 184, 5031–5052 (2021).
Jiang, L. et al. Prospective observational study on biomarkers of response in pancreatic ductal adenocarcinoma. Nat. Med. 30, 749–761 (2024).
Hanahan, D. & Weinberg, R. A. Hallmarks of cancer: the next generation. Cell 144, 646–674 (2011).
Grünwald, B. T. et al. Spatially confined sub-tumor microenvironments in pancreatic cancer. Cell 184, 5577–5592 (2021).
Scott, J. D. & Pawson, T. Cell signaling in space and time: where proteins come together and when they’re apart. Science 326, 1220–1224 (2009).
Uhlén, M. et al. Tissue-based map of the human proteome. Science 347, 1260419 (2015).
Hunter, T. Tyrosine phosphorylation: thirty years and counting. Curr. Opin. Cell Biol. 21, 140–146 (2009).
Huang, P., Gao, W., Fu, C. & Tian, R. Functional and clinical proteomic exploration of pancreatic cancer. Mol. Cell. Proteom. 22, 100575 (2023).
Tape, C. J. et al. Oncogenic KRAS regulates tumor cell signaling via stromal reciprocation. Cell 165, 910–920 (2016).
Shi, Y. et al. Targeting LIF-mediated paracrine interaction for pancreatic cancer therapy and monitoring. Nature 569, 131–135 (2019).
Pan, S., Chen, R., Aebersold, R. & Brentnall, T. A. Mass spectrometry based glycoproteomics—from a proteomics perspective. Mol. Cell. Proteom. 10, R110.003251 (2011).
Türei, D. et al. Integrated intra- and intercellular signaling knowledge for multicellular omics analysis. Mol. Syst. Biol. 17, e9923 (2021).
Gukovsky, I., Li, N., Todoric, J., Gukovskaya, A. & Karin, M. Inflammation, autophagy, and obesity: common features in the pathogenesis of pancreatitis and pancreatic cancer. Gastroenterology 144, 1199–1209 (2013).
Peng, J. et al. Single-cell RNA-seq highlights intra-tumoral heterogeneity and malignant progression in pancreatic ductal adenocarcinoma. Cell Res. 29, 725–738 (2019).
DeBerardinis, R. J., Lum, J. J., Hatzivassiliou, G. & Thompson, C. B. The biology of cancer: metabolic reprogramming fuels cell growth and proliferation. Cell Metab. 7, 11–20 (2008).
Olive, K. P. et al. Inhibition of Hedgehog signaling enhances delivery of chemotherapy in a mouse model of pancreatic cancer. Science 324, 1457–1461 (2009).
Del Rosario, A. M. & White, F. M. Quantifying oncogenic phosphotyrosine signaling networks through systems biology. Curr. Opin. Genet. Dev. 20, 23–30 (2010).
Lim, W. A. & Pawson, T. Phosphotyrosine signaling: evolving a new cellular communication system. Cell 142, 661–667 (2010).
Merilahti, J. A. M. & Elenius, K. Gamma-secretase-dependent signaling of receptor tyrosine kinases. Oncogene 38, 151–163 (2019).
Klein, T., Eckhard, U., Dufour, A., Solis, N. & Overall, C. M. Proteolytic cleavage-mechanisms, function, and “omic” approaches for a near-ubiquitous posttranslational modification. Chem. Rev. 118, 1137–1168 (2018).
Graham, D. K., DeRyckere, D., Davies, K. D. & Earp, H. S. The TAM family: phosphatidylserine sensing receptor tyrosine kinases gone awry in cancer. Nat. Rev. Cancer 14, 769–785 (2014).
Kariolis, M. S. et al. An engineered Axl ‘decoy receptor’ effectively silences the Gas6-Axl signaling axis. Nat. Chem. Biol. 10, 977–983 (2014).
Nieto, M. A., Huang, R. Y., Jackson, R. A. & Thiery, J. P. EMT: 2016. Cell 166, 21–45 (2016).
Chen, W. et al. Simple and integrated spintip-based technology applied for deep proteome profiling. Anal. Chem. 88, 4864–4871 (2016).
Erkan, M. et al. StellaTUM: current consensus and discussion on pancreatic stellate cell research. Gut 61, 172–178 (2012).
Rappsilber, J., Mann, M. & Ishihama, Y. Protocol for micro-purification, enrichment, pre-fractionation and storage of peptides for proteomics using StageTips. Nat. Protoc. 2, 1896–1906 (2007).
Huang, P. et al. Spatial proteome profiling by immunohistochemistry-based laser capture microdissection and data-independent acquisition proteomics. Anal. Chim. Acta 1127, 140–148 (2020).
Xu, R. et al. Spatial-resolution cell type proteome profiling of cancer tissue by fully integrated proteomics technology. Anal. Chem. 90, 5879–5886 (2018).
Bian, Y. et al. Ultra-deep tyrosine phosphoproteomics enabled by a phosphotyrosine superbinder. Nat. Chem. Biol. 12, 959–966 (2016).
Chu, B. et al. Photoaffinity-engineered protein scaffold for systematically exploring native phosphotyrosine signaling complexes in tumor samples. Proc. Natl Acad. Sci. USA 115, E8863–E8872 (2018).
Zhou, H. et al. Robust phosphoproteome enrichment using monodisperse microsphere-based immobilized titanium (IV) ion affinity chromatography. Nat. Protoc. 8, 461–480 (2013).
Tian, R. et al. Combinatorial proteomic analysis of intercellular signaling applied to the CD28 T-cell costimulatory receptor. Proc. Natl Acad. Sci. USA 112, E1594–E1603 (2015).
Huang, P., Li, H., Gao, W., Cai, Z. & Tian, R. A fully integrated spintip-based approach for sensitive and quantitative profiling of region-resolved in vivo brain glycoproteome. Anal. Chem. 91, 9181–9189 (2019).
Jedrychowski, M. P. et al. Detection and quantitation of circulating human irisin by tandem mass spectrometry. Cell Metab. 22, 734–740 (2015).
Bausch-Fluck, D. et al. The in silico human surfaceome. Proc. Natl Acad. Sci. USA 115, E10988–E10997 (2018).
Möller, S., Croning, M. D. & Apweiler, R. Evaluation of methods for the prediction of membrane spanning regions. Bioinformatics 17, 646–653 (2001).
Nielsen, H. Predicting secretory proteins with SignalP. Methods Mol. Biol. 1611, 59–73 (2017).
Käll, L., Krogh, A. & Sonnhammer, E. L. A combined transmembrane topology and signal peptide prediction method. J. Mol. Biol. 338, 1027–1036 (2004).
Ramilowski, J. A. et al. A draft network of ligand-receptor-mediated multicellular signalling in human. Nat. Commun. 6, 7866 (2015).
Graeber, T. G. & Eisenberg, D. Bioinformatic identification of potential autocrine signaling loops in cancers from gene expression profiles. Nat. Genet. 29, 295–300 (2001).
Sharman, J. L. et al. IUPHAR-DB: updated database content and new features. Nucleic Acids Res. 41, D1083–D1088 (2013).
Ben-Shlomo, I., Yu Hsu, S., Rauch, R., Kowalski, H. W. & Hsueh, A. J. Signaling receptome: a genomic and evolutionary perspective of plasma membrane receptors involved in signal transduction. Sci. STKE 2003, RE9 (2003).
Keshava Prasad, T. S. et al. Human Protein Reference Database—2009 update. Nucleic Acids Res. 37, D767–D772 (2009).
Szklarczyk, D. et al. The STRING database in 2021: customizable protein-protein networks, and functional characterization of user-uploaded gene/measurement sets. Nucleic Acids Res. 49, D605–D612 (2021).
Manning, G., Whyte, D. B., Martinez, R., Hunter, T. & Sudarsanam, S. The protein kinase complement of the human genome. Science 298, 1912–1934 (2002).
Lemmon, M. A. & Schlessinger, J. Cell signaling by receptor tyrosine kinases. Cell 141, 1117–1134 (2010).
Liu, B. A. et al. The human and mouse complement of SH2 ___domain proteins-establishing the boundaries of phosphotyrosine signaling. Mol. Cell 22, 851–868 (2006).
Wagner, M. J., Stacey, M. M., Liu, B. A. & Pawson, T. Molecular mechanisms of SH2- and PTB-___domain-containing proteins in receptor tyrosine kinase signaling. Cold Spring Harb. Perspect. Biol. 5, a008987 (2013).
Uhlik, M. T. et al. Structural and evolutionary division of phosphotyrosine binding (PTB) domains. J. Mol. Biol. 345, 1–20 (2005).
Alonso, A. et al. Protein tyrosine phosphatases in the human genome. Cell 117, 699–711 (2004).
Pao, L. I., Badour, K., Siminovitch, K. A. & Neel, B. G. Nonreceptor protein-tyrosine phosphatases in immune cell signaling. Annu. Rev. Immunol. 25, 473–523 (2007).
Zielinska, D. F., Gnad, F., Wisniewski, J. R. & Mann, M. Precision mapping of an in vivo N-glycoproteome reveals rigid topological and sequence constraints. Cell 141, 897–907 (2010).
Tyanova, S., Temu, T. & Cox, J. The MaxQuant computational platform for mass spectrometry-based shotgun proteomics. Nat. Protoc. 11, 2301–2319 (2016).
Newman, A. M. et al. Determining cell type abundance and expression from bulk tissues with digital cytometry. Nat. Biotechnol. 37, 773–782 (2019).
Futschik, M. E. & Carlisle, B. Noise-robust soft clustering of gene expression time-course data. J. Bioinform. Comput. Biol. 3, 965–988 (2005).
Morello, A., Sadelain, M. & Adusumilli, P. S. Mesothelin-targeted CARs: driving T cells to solid tumors. Cancer Discov. 6, 133–146 (2016).
Liu, X. et al. A new panel of pancreatic cancer biomarkers discovered using a mass spectrometry-based pipeline. Br. J. Cancer 117, 1846–1854 (2017).
Fredriksson, L., Li, H. & Eriksson, U. The PDGF family: four gene products form five dimeric isoforms. Cytokine Growth Factor Rev. 15, 197–204 (2004).
Fu, C. et al. The transcription factor ZFHX3 is crucial for the angiogenic function of hypoxia-inducible factor 1α in liver cancer cells. J. Biol. Chem. 295, 7060–7074 (2020).
Zhu, S. et al. GDF15 promotes glioma stem cell-like phenotype via regulation of ERK1/2-c-Fos-LIF signaling. Cell Death Discov. 7, 3 (2021).
Bolte, S. & Cordelières, F. P. A guided tour into subcellular colocalization analysis in light microscopy. J. Microsc. 224, 213–232 (2006).
Shi, X. et al. Integrated profiling of human pancreatic cancer organoids reveals chromatin accessibility features associated with drug sensitivity. Nat. Commun. 13, 2169 (2022).
Jaaks, P. et al. Effective drug combinations in breast, colon and pancreatic cancer cells. Nature 603, 166–173 (2022).
Liu, C. et al. Kindlin-2 enhances c-Myc translation through association with DDX3X to promote pancreatic ductal adenocarcinoma progression. Theranostics 13, 4333–4355 (2023).
Perez-Riverol, Y. et al. The PRIDE database resources in 2022: a hub for mass spectrometry-based proteomics evidences. Nucleic Acids Res. 50, D543–d552 (2022).
Matthews, M. L. et al. Chemoproteomic profiling and discovery of protein electrophiles in human cells. Nat. Chem. 9, 234–243 (2017).
Maurer, C. et al. Experimental microdissection enables functional harmonisation of pancreatic cancer subtypes. Gut 68, 1034–1043 (2019).
Elyada, E. et al. Cross-species single-cell analysis of pancreatic ductal adenocarcinoma reveals antigen-presenting cancer-associated fibroblasts. Cancer Discov. 9, 1102–1123 (2019).
Kim, J. et al. Detection of early pancreatic ductal adenocarcinoma with thrombospondin-2 and CA19-9 blood markers. Sci. Transl. Med. 9, eaah5583 (2017).
Cohen, J. D. et al. Detection and localization of surgically resectable cancers with a multi-analyte blood test. Science 359, 926–930 (2018).
Melo, S. A. et al. Glypican-1 identifies cancer exosomes and detects early pancreatic cancer. Nature 523, 177–182 (2015).
Seguin, L. et al. Galectin-3, a druggable vulnerability for KRAS-addicted cancers. Cancer Discov. 7, 1464–1479 (2017).
Jones, S. et al. Core signaling pathways in human pancreatic cancers revealed by global genomic analyses. Science 321, 1801–1806 (2008).
Makohon-Moore, A. & Iacobuzio-Donahue, C. A. Pancreatic cancer biology and genetics from an evolutionary perspective. Nat. Rev. Cancer 16, 553–565 (2016).
Acknowledgements
This study was supported by grants from the China State Key Basic Research Program Grants (2020YFE0202200, 2021YFA1301601, 2021YFA1301602, 2021YFA1302603, 2023YFC2506401, 2022YFC3401104, 2022YFA1105300-03 and 2020YFA0509002); the National Natural Science Foundation of China (22125403, 92253304, 91953118, 92359302, 32201218, 82341074, 32171433, 22104047, 22004056, 82273438 and 32125013); the Shenzhen Innovation of Science and Technology Commission (ZDSYS20230626090803004, JSGGZD20220822095200001, KJZD20230923114220041, JCYJ20200109141212325, JCYJ20210324120210029 and JCYJ20200109140814408); Guangdong province (2019B151502050); the Basic Frontier Science Research Program of Chinese Academy of Sciences (ZDBS-LY-SM015); the Shanghai Municipal Science and Technology Major Project; and the Innovative Research Team of High-level Local Universities in Shanghai (SHSMUZDCX20211800). Work in the T.H. group was supported by grants from the NCI (CA080100 and CA082683), NIH NCI 1 P01 (CA265762-01A1), Lustgarten Foundation Awards (388246 and 552873) and core services funded through an NCI Cancer Center Support Grant (CA014195). T.H. is a Frank and Elsa Schilling American Cancer Society Professor, and the Renato Dulbecco Chain in Cancer Research.
Author information
Authors and Affiliations
Contributions
R.T. conceived and designed the study. P.H. performed most MS-based experiments, ELISA and initial data analysis. W.G. performed analysis of proteomic data, with assistance from Yuan Li. C.F. performed the majority of the in vitro and part of the in vivo validation experiments and analysed the data with assistance from X.D. Yunguang Li performed organoid-related experiments and analysed the data with assistance from Y.Z. B.C. performed profiling of pTyr protein complexes and in vitro validation of AXL shedding. M.W., J.Y., H.W. and R.Q. collected and analysed clinical samples. Y.S. provided the genetically engineered mouse model of pancreatic cancer and analysed the data. A.H. synthesized the biotin-hydrazide probe. Q.K. assisted with phosphoproteomic-related experiments. P.H., W.G., C.F., Y.S., T.H. and R.T. wrote the manuscript with input from all of the authors. R.T., T.H., R.Q., D.G. and Y.S. supervised the study.
Corresponding authors
Ethics declarations
Competing interests
R.T. is a founder of BayOmics.
Peer review
Peer review information
Nature thanks the anonymous reviewers for their contribution to the peer review of this work. Peer reviewer reports are available.
Additional information
Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Extended data figures and tables
Extended Data Fig. 1 Optimization of N-glycoproteomic method using the synthesized long chain biotin-hydrazide probe.
a, Chemical structure of in-house synthesized biotin-hydrazide for glycoprotein labelling and enrichment. b, Labelling and enrichment efficiency of the biotin-hydrazide probe. Mouse pancreas proteins were extracted and labelled with 2 mM probe for 1 h (lane 2). Lysate without biotin-hydrazide labelling was subjected for streptavidin pull down as control (lane 3). Labelled proteins were pulled down by streptavidin beads (lane 4). Supernatant was collected from the streptavidin pull down (lane 5). c, Comparison of biotin-hydrazide probe-based method with conventional hydrazide bead-based method. Either biotin-hydrazide probe or hydrazide beads were incubated with 500 μg lysate for different times. After on-bead trypsin digestion to remove non-glycopeptides, glycopeptides were released from the streptavidin beads or hydrazide beads by PNGase F for LC–MS/MS analysis. Data are mean of two technical replicates. d,e, Subcellular ___location of proteins identified by glycopeptides (d) and sequence coverage comparison of 1,313 S–PM proteins commonly identified by glycopeptides and non-glycopeptides (e) using 6 pairs of tumour and NT samples. f, The comparison of analysis by non-glycopeptides and glycopeptides in terms of identification and quantification performance of S–PM proteins. The left panel is the number of S–PM proteins identified by glycopeptides and non-glycopeptides. Data are mean ± s.d. of 6 pairs of tumour and NT samples. The middle panel is the sum peptide intensities of all identified S–PM proteins from these 12 tissue samples. The right panel is the CV of 1,313 overlapped S–PM proteins quantified with LFQ intensities by glycopeptides and non-glycopeptides. g, The percentage of secreted (S), plasma membrane (PM), and other transmembrane (TM) proteins, non-S/PM/TM proteins, and nonspecifically labelled proteins reported by Matthews et al.70 to total proteins in terms of protein number (left panel) and sum LFQ intensity (right panel) as identified by glycopeptides and non-glycopeptides. h, The percentage of proteins nonspecifically absorbed on streptavidin beads to total proteins identified by glycoprotein enrichment. The experimental procedure for identification of nonspecifically absorbed proteins was the same as glycoprotein enrichment, except that no biotin-hydrazide probe was added. Proteins identified only in the streptavidin pull down sample without biotin-hydrazide probe or having 2-fold higher LFQ intensity as compared with glycoprotein enrichment were defined as nonspecifically absorbed proteins. i,j, Comparison of quantification performance of S–PM proteins by global proteomic method (Unenriched) and N-glycoproteomic method (Enriched) using 6 pairs of tumour and NT samples. S–PM proteins quantified by Unenriched and Enriched were compared in terms of protein number and intensities (i), and CV of 1,546 S–PM proteins commonly quantified by Unenriched and Enrich (j). k,l, Correlation of the average tumour/NT ratios of commonly quantified S–PM proteins between the two methods. Bio-replicates denote correlation of the 6 pairs of tumour and NT samples; correlations of glycoproteins identified with different number of N-glycosites are independently displayed (k); technical replicates denote correlation of 1 pair of tumour and NT samples for three technical replicates (l).
Extended Data Fig. 2 Sample quality control and functional annotation of S–PM proteome in PDAC.
a, Pearson’s correlation coefficients of LFQ intensities between any two samples from the same group of normal, NT, CP, or tumour tissues, respectively. b, Classification of S–PM proteins according to their roles in intercellular communications. The bar graph shows the top 20 intercellular communication roles. c, Clustering of top 50 GOMF terms of significantly changed S–PM proteins between Tumour and NT, related to Fig. 1g. d, Comparison of top 20 GOMF terms of significantly altered S–PM proteins between Tumour versus Normal and Tumour versus CP. P values are from hypergeometric distribution (c, d).
Extended Data Fig. 3 Extended analysis of spatially resolved and cell type-specific proteomic data and validation of PCC specific proteins.
a, Cell type deconvolution of 29 tumour samples using our bulk S–PM proteome in Fig. 1b and published scRNA-seq dataset of PDAC (Methods). Box plots are as defined in Fig. 2c. b, Representative images of IHC-guided and hematoxylin staining-based LCM of PCC and stromal regions from tumour tissue section. The stroma region was stained by IHC targeting PDGFRB, a marker of stroma cells. The PCC region was stained by IHC targeting KRT19, a marker of epithelial cells. c, The number of S–PM proteins identified in each of the tissue and cell line samples. d, Pearson’s correlation coefficients of LFQ intensities between any two biological replicates from the same region of PCCs or stroma. e, Comparison of proteome depth of our LCM-based proteomic dataset with a recently published LCM-based proteomic dataset of PDAC8. The raw files of the published dataset were downloaded and processed using the same workflow and criteria as our dataset. f, Comparison of the S–PM proteins identified in spatial proteomic dataset with genes of S–PM proteins from the transcriptomic dataset summarized from spatial or scRNA-seq analysis of PDAC18,71,72. All the identified genes of S–PM proteins from the three references were combined as the transcriptome dataset. g, Cell-type annotation of the S–PM proteins identified from N-glycoproteomic profiling of 29 bulk tumour tissues. h, TSA staining of 5 PCC-specific and tumour-upregulated PM proteins, including the aspartate/glutamate transporter SLC1A3, the PKD1 calcium channels, the TMPRSS4 protease, the CDON Hedgehog signalling regulator, and the LRIG2 protease regulator. KRT19 and PDGFRB were marker of PCCs and stroma, respectively. Box plots are as defined in Fig. 2c. The pearson’s r values indicates the colocalization potential of target proteins with KRT19 or PDGFRB (Methods). i,j, The number of ligands, receptors, and ligand–receptor pairs annotated from secreted ligands and plasma membrane receptors of Extended Data Fig. 2b by an in-house ligand–receptor database (see Methods) (h), and display of the 1,724 ligand–receptor pairs (j). The pairs of three classes of most enriched GOMF terms are shown as examples, with representative proteins displayed. k, Dot plot showing GOMF terms of the 1,724 ligand–receptor pairs. Top 20 GOMF terms according to pair number were annotated and terms in red were presented in j. l, Protein expression trend of the 1,724 pairs according to LFQ intensity ratio distribution of tumour and normal tissues. One dot represents one pair. The X and Y axes are the log2 transformed LFQ ratio between Tumour and Normal of ligand and receptor, respectively. The pie chart shows the percentage of pairs in each section to total pairs. m, The number of spatially resolved and cell type-specific ligands and receptors (top panel) and their predicated signalling flow (bottom panel) in 29 tumours. Data are mean ± s.d. P values are from two-tailed unpaired Student’s t-test (e,m), or hypergeometric distribution (k).
Extended Data Fig. 4 Extended analysis for time-resolved N-glycoproteomic profiling of KPC mouse model.
a, Pearson’s correlation coefficients of LFQ intensities between any two samples from the same group of NT, 3 weeks, 5 weeks, or 7 weeks of KPC mice, respectively. b, Consensus-clustering analysis of S–PM proteins differentially expressed between NT and different tumour stages of the KPC mouse model. c, The number of differentially expressed S–PM proteins in each stage of tumour tissues as compared with NT, respectively. d, Overlap and expression trend consistency of differentially expressed proteins between human PDAC and KPC mouse model. e, Relative expression levels of representative PDAC biomarkers, including Lif14, Thbs273, Timp174, Gpc175, Msln60, and Lgals376, in different stage tumour tissues of KPC mice and in human pancreatic tissues. The number of human and mouse tissue samples in each group is indicated in Figs. 1a and 3b, respectively. f, Top 5 GOMF terms of S–PM proteins in the three clusters. P values are calculated by hypergeometric distribution. g, Validation of TNFRSF11B and NPTX1 in PDAC plasma samples. Human plasma samples from training and validation cohorts were tested by ELISA kits. h, Annotation of ligand–receptor pairs in 29 bulk human tumour tissues on the basis of the indicated tumour progression trends. Data are mean ± s.d. All pairs refer to the pairs shown in Extended Data Fig. 3j in which the expression of at least ligand or receptor was significantly changed in human Tumour versus Normal in N-glycoproteomic dataset. Box plots are as defined in Fig. 2c (e, tissue results of g). Box plots of plasma results in g: centre line, median; boxes, interquartile range; whiskers, minimum to maximum. P values are from two-tailed unpaired Student’s t-test.
Extended Data Fig. 5 Extended analysis of pTyr protein complex and pTyr site profiling and integrated analysis of ligand–receptor–downstream signalling axes.
a, Clinical tissue samples used for N-glycoproteomic analysis, photo-pTyr-scaffold, and pTyr peptide enrichment. b, Overlap of pTyr writers, readers, and erasers identified by photo-pTyr-scaffold approach and pTyr peptide enrichment approach. c, Summary of all the pTyr writers, readers, and erasers identified by two approaches in Fig. 4b. The rectangle indicates proteins identified by the photo-pTyr-scaffold approach and the circle indicates the pTyr sites identified by pTyr peptide enrichment approach. Proteins are classified according to their molecular functions. d, Expression levels and pTyr sites of top ranked pTyr writers, erasers and readers in Fig. 4c. Box plots are as defined in Fig. 2c. P values are from two-tailed unpaired Student’s t-test. e, The overlapped GOBP terms of proteins identified by our multidimensional proteomics (S–PM and pTyr machinery proteins identified in tumour tissues by N-glycoproteomic profiling and pTyr-mediated complexes profiling) with the core signalling pathways summarized from global genomic analysis of PDAC77,78. The core signalling pathways summarized in these two references were combined. P values are from hypergeometric distribution. f, The four categories of activated ligand–receptor–downstream signalling axes in which the expression of both receptor and downstream protein were significantly changed in tumours in the pTyr signalling dataset (circles labelled with P). g, Spatial and temporal annotation of the 148 pairs in Fig. 4d. h, The activated paracrine signalling pairs from stromal cells to PCCs in tumour and their downstream proteins screened out from g.
Extended Data Fig. 6 Functional characterization of reciprocal signalling between stromal cells and PCCs mediated by the PDGFR–PTPN11–FOS signalling axis.
a, Expression levels of PDGFRA, PDGFRB, and PDGFC in the multidimensional proteomic datasets shown in Figs. 1–4. The number of samples for protein expression, cell type, tumour progression, and pTyr activation is indicated in Fig. 1a, Fig. 2a, Fig. 3b, and Extended Data Fig. 5a, respectively. Box plots are as defined in Fig. 2c. P values are from two-tailed unpaired Student’s t-test. b, Validation of PDGFR activation by PANC1 CM or recombinant PDGF-BB and inhibition by the PDGFR inhibitor crenolanib in HPaSteCs. Cells were starved in FBS-free medium for 12 h with or without PDGFR inhibitor crenolanib, and then stimulated with PDGF-BB or PANC1 CM for 5 min before cell lysis for WB analysis. c, Validation of signalling proteins downstream of PDGFRA and PDGFRB. HPaSteCs were transfected with siRNAs targeting PDGFRA or PDGFRB in normal medium for 24 h, followed by in FBS-free medium for 24 h, and then stimulated with PANC1 CM for 5 min before cell lysis for WB analysis. d, Validation of PTPN11 as an upstream regulator of ERK signalling but not AKT signalling. Cells were starved in FBS-free medium for 12 h with or without PDGFR inhibitor crenolanib or PTPN11 inhibitor SHP099 with indicated concentrations, and then stimulated with PDGF-BB for 5 min before cell lysis for WB analysis. e, Secretome analysis of HPaSteCs after PDGF-BB stimulation and PDGFR or PTPN11 inhibition (n = 3 biological replicates). HPaSteCs were treated with 100 ng ml−1 PDGF-BB, 0.5 μM crenolanib or 10 μM SHP099 for 24 h, respectively. f, Investigation of the signalling axis regulating LIF expression upon PDGF stimulation. Cells were treated with PDGFR inhibitor crenolanib (0.5 μM), JAK inhibitor ruxolitinib (1 μM), PI3K inhibitor LY294002 (10 μM), PTPN11 inhibitor SHP099 (10 μM), MEK inhibitor mirdametinib (10 μM), or ERK inhibitor ravoxertinib (10 μM) for 24 h before cell lysis for total RNA extraction and measurement of LIF mRNA levels by real-time qPCR. g, Effect of PTPN11 knockdown on LIF protein secretion from HPaSteCs with or without PDGF-BB stimulation. h, Effect of inhibitors targeting the PDGFR–PTPN11–ERK signalling axis on LIF promoter activity. HPaSteCs were stimulated with PDGF-BB with or without an inhibitor targeting PDGFR, PTPN11 or ERK. LIF promoter activity was measured by a dual-luciferase reporter assay system. i, Validation of FOS as a LIF gene transcription factor after PDGF-BB stimulation (100 ng ml−1, 24 h) as determined by ChIP assay. P1 to P4 indicate four promoter regions of LIF, the result shows that P2 region is the binding site of FOS. Due to similar molecular weight of proteins, samples were run on separate gels, with β-actin as sample processing control (b–d). All the bar graphs are mean ± s.d. of n = 3 (b–d,f,g,i) or n = 4 (h) biological replicates; P values are two-tailed unpaired Student’s t-test (a,f–i) or one-tailed paired Student’s t-test (b–d).
Extended Data Fig. 7 Workflow for identification of shed PM proteins and extended analysis for validation of MMP-mediated shedding of PM proteins.
a, Schematic representation of detail criteria for identification of shed PM proteins in tumours. In step 1, the unique peptide ratio of ECD/ICD for PM proteins (RatioPM) identified in the PM proteome by the N-glycoproteomic analysis of cell pellets (data presented in Extended Data Fig. 3c), the same ratio for PM proteins (RatioS) identified by the secretome analysis (data presented in Extended Data Fig. 3c), and the same ratio for PM proteins (RatioS–PM) identified in the S–PM proteome by the N-glycoproteomic analysis of 29 PDAC tumour tissues (data presented in Fig. 1b) were calculated after data filtering according to the indicated criteria, respectively. The strict cutoff ensures the reliability and reproducibility of quantification performance in each dataset. In step 2, to avoid false positive calculation of shedding proteins due to the intrinsic difference of sequence length of ECD and ICD, the RatioS were first normalized to (divided by) their corresponding RatioPM to generate the Indextraining values so as to identify shed PM proteins in the secretome of 8 cell lines. Then, 45 shed PM proteins were identified in cell lines. By using the same normalization step and indicated filtering criteria, 22 shed proteins were finally identified and validated in tumour tissues. b, Molecular functions of the 22 shed proteins identified in tumour tissues. c, Expression level of MMP1, MMP9, MMP11, MMP15, ADAM10, and ADAM17 in tumour and normal tissues by N-glycoproteomic profiling. d, Knockdown efficiency of each MMP by siRNA in PANC1 cells in Fig. 5e. Cells were transfected with siRNAs in normal medium for 24 h, followed by in FBS-free medium for 24 h before that WCLs were collected for total RNA extraction. The mRNA levels of MMPs were measured by real-time qRT–PCR. e, Statistic analysis of WB result in Fig. 5f, showing sAXL levels in PANC1 CM after knockdown of different MMPs. Cells were transfected with siRNA targeting MMP1, MMP9, MMP11, or MMP15 in normal medium for 24 h, followed by in FBS-free medium for 24 h before that CM and WCL were collected for WB analysis. f, Knockdown efficiency of each MMP by siRNA in MIA PaCa2 cells. g, Global profiling of shedding substrates of MMPs in MIA PaCa2 cells. The heatmap shows shedding level of 17 PM proteins identified as shed proteins in cell lines and tumour tissues. AXL levels are shown as an example. h, Validation of AXL shedding upon MMP knockdown by siRNAs in MIA PaCa2 cells. Cells were treated with siRNAs by following the same procedure as PANC1. sAXL in CM and WCL was detected by WB, β-actin was run on the same gel as loading control, and the bar graph shows band intensities of sAXL in CM of the WB image. Box plots are as defined in Fig. 2c (c,g). All the bar graphs are mean ± s.d. of n = 4 (d,f) or n = 3 (e,h) biological replicates. P values are from two-tailed unpaired Student’s t-test (c,d,f,g) or one-tailed paired Student’s t-test (e,h).
Extended Data Fig. 8 PRM quantification of AXL shedding in PCC and stromal regions of tumours.
a, Expression level of TAM family members AXL, MERTK and TYRO3 in multi-dimensional proteomic datasets. The number of samples for protein expression, tumour progression, and pTyr activation is indicated in Fig. 1a, Fig. 3b, and Extended Data Fig. 5a, respectively. Box plots are as defined in Fig. 2c. P values are from two-tailed unpaired Student’s t-test. b, The linearity of PRM quantification for stable isotope labelled heavy peptides of ECD and ICD of AXL, and GAS6. Different amount of heavy peptides were spiked into 100 ng of PANC1 peptides before PRM-MS analysis. c-e, Representative PRM-MS transition peaks of ICD (c) and ECD (d) of AXL, and GAS6 (e) from tumour samples. The transition peaks of stable isotope labelled heavy peptide spiked into the sample are shown on the right. The dark arrow indicates the retention time and mass error for the highest transition peak. f, IHC image of lymph node metastasis. Tumour cells metastasized into the lymph node were marked with IHC staining targeting KRT19. Ten lymph nodes were stained and representative image is presented. g–i, Classification of tumours according to lymph node metastasis status and relative levels of sAXL and GAS6 in PCC and stroma regions of 50 primary tumour samples (g,h), and in 41 samples showing consistent trend between PCCs and stroma (namely, the absolute amount of sAXL is higher or lower than GAS6 for both PCC and stromal regions isolated from the same tumour). Displayed are averages of PCC and stromal regions quantified from the same tumour for both sAXL and GAS6 (i). j, Boxplot of sAXL, GAS6, and CA19-9 levels in human plasma samples from the training cohort and validation cohort of normal controls and PDAC patients. Centre line, median; boxes, interquartile range; whiskers, minimum to maximum. The results from the training cohort and the validation cohort were combined for ROC curve analysis in Fig. 6d.
Extended Data Fig. 9 Combined inhibition of AXL shedding and AXL activity synergistically reduced cancer cell proliferation, migration, and tumour growth.
a, Inhibition of AXL shedding by the broad-spectrum MMP inhibitor BB-94. β-actin was run on the same gel as loading control. The bar graph is statistical analysis of sAXL level in PANC1 CM. b, Inhibition of AXL shedding in primary KPCP tumour cells by the broad-spectrum MMP inhibitor BB-94. Cells were maintained in FBS-free medium containing 10 μm BB-94 for 24 h, and secretome in CM was profiled by LC-MS/MS analysis. The LFQ intensity of AXL was extracted for comparison between control and BB-94 treatment. c, Statistic analysis of the WB results in Fig. 6e, showing relative band intensities of p-AKT after normalization to total AKT. d, PRM analysis of two AXL pTyr peptides after enrichment of phosphopeptides. The treatment of PANC1 cells was the same as in Fig. 6e. The left panel of each pTyr peptide is representative transition peaks. The dark arrow indicates the retention time and mass error for the highest transition peak. e, Schematic workflow for testing synergistic effect between BB-94 and R428 on PDOs. f, Histological characterization of PDAC tumour tissues and corresponding PDOs and PDOs-derived xenografts. PDOs and PDOs-derived xenografts formed typical glandular tubular structures similar to the corresponding patient tumours, and had similar expression levels of the epithelial cell marker KRT19 and SOX9, and cell proliferation marker Ki67. g,h, Solo or combined drug treatments on xenograft tumours generated by one case of PDO (DAC-71) with significant synergistic response (g) between BB-94 and R428 (statistics are presented in Fig. 6g). i–k, Validation of synergistic effect on xenograft models generated by one case of PDO (DAC-18) with no synergistic response (i). Data are mean + s.d. of n = 8 xenograft tumours per group (j,k). l, Orthotopic tumours of PDAC under different drug treatment. Orthotopic model was constructed by injecting KPCP primary cancer cells into pancreata of NSG mice. After tumour formation, mice were daily administrated with indicated drugs for 14 days. The dark red tissues were spleens, which were removed before measuring tumour weights. Tumour weights are presented as mean ± s.d. of n = 9 mice per condition. m, Correlation of MMP1 and MMP11 mRNA expression levels in 66 pancreatic PDO lines with drug combination response of BB-94 and R428. Box plots are as defined in Fig. 2c. n,o, Colony formation assay of MIA PaCa2 cells with stable knockdown of MMP1 or MMP11 by shRNAs. Cells were treated with DMSO as control or 0.5 μM R428 for testing of inhibition efficiency (n). Knockdown efficiency was measured by real-time qRT–PCR (o). p, Transwell migration assays of MIA PaCa2 cells under shedding inhibition and/or AXL inhibition. Shown are representative crystal violet-stained images of transwell migration assays of MIA PaCa2 cells under different treatments. Quantification of cell migration determined by counting the number of migratory cells per field and relative numbers of migratory cells were calculated by normalizing to control cells. q, Expression of EMT markers in cancer cells upon inhibition of shedding by BB-94. Due to similar molecular weight of proteins, samples were run on separate gels, with β-actin as sample processing control. All the bar graphs are mean ± s.d. of n = 3 biological replicates (the data points in p are relative cell numbers from 2 random views of each biological replicates). P values are from two-tailed unpaired Student’s t-test (b,d,k,l,m,n,o,p) or one-tailed paired Student’s t-test (a,c,q).
Supplementary information
Supplementary Information
Supplementary Methods, Supplementary Results, Supplementary Discussion, Supplementary Figs. 1–11, legends for the Supplementary Tables and Supplementary References.
Supplementary Tables
Supplementary Tables 1–7.
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
Huang, P., Gao, W., Fu, C. et al. Clinical functional proteomics of intercellular signalling in pancreatic cancer. Nature 637, 726–735 (2025). https://doi.org/10.1038/s41586-024-08225-y
Received:
Accepted:
Published:
Issue Date:
DOI: https://doi.org/10.1038/s41586-024-08225-y
This article is cited by
-
Advances in humanoid organoid-based research on inter-organ communications during cardiac organogenesis and cardiovascular diseases
Journal of Translational Medicine (2025)
-
CircERC1 facilitates chemoresistance through inhibiting pyroptosis and remodeling extracellular matrix in pancreatic cancer
Molecular Cancer (2025)