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:

CD9 identifies pancreatic cancer stem cells and modulates glutamine metabolism to fuel tumour growth

Abstract

Pancreatic ductal adenocarcinoma (PDAC) shows great cellular heterogeneity, with pronounced epithelial and mesenchymal cancer cell populations. However, the cellular hierarchy underlying PDAC cell diversity is unknown. Here we identify the tetraspanin CD9 as a marker of PDAC tumour-initiating cells. CD9high cells had increased organoid formation capability, and generated tumour grafts in vivo at limiting dilutions. Tumours initiated from CD9high cells recapitulated the cellular heterogeneity of primary PDAC, whereas CD9low cells produced only duct-like epithelial progeny. CD9 knockdown decreased the growth of PDAC organoids, and heterozygous CD9 deletion in Pdx1-Cre; LSL-KRasG12D; p53F/F mice prolonged overall survival. Mechanistically, CD9 promoted the plasma membrane localization of the glutamine transporter ASCT2, enhancing glutamine uptake in PDAC cells. Thus, our study identifies a PDAC subpopulation capable of initiating PDAC and giving rise to PDAC heterogeneity, suggesting that the cellular diversity of PDAC is generated by PDAC stem cell differentiation.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: CD9 identification.
Fig. 2: CD9 marks TICs.
Fig. 3: CD9high cells recapitulate tumour heterogeneity.
Fig. 4: CD9 is required for efficient PDAC development.
Fig. 5: CD9 facilitates glutamine import into TICs.
Fig. 6: CD9 expression in human PDAC and model of CD9 function in PDAC.

Similar content being viewed by others

Data availability

Microarray data supporting the findings of this study have been deposited in the Gene Expression Omnibus under the accession code GSE121864. Mass spectrometry data have been deposited in ProteomeXchange with the primary accession code PXD015439. Clinical data and normalized RSEM read count tables for pancreatic adenocarcinoma were downloaded from the Broad firehose data repository (https://gdac.broadinstitute.org/). Source data for Figs. 1 and 5 and Extended Data Figs. 1 and 57 are available online. All other relevant data are available from the corresponding author. Ethical consent for use of human biological samples is currently restricted to the Adult Stem Cell Laboratory at the Francis Crick Institute.

References

  1. Cronin, K. A. et al. Annual report to the nation on the status of cancer, part I: national cancer statistics. Cancer 124, 2785–2800 (2018).

    Article  PubMed  Google Scholar 

  2. Neoptolemos, J. P. et al. Therapeutic developments in pancreatic cancer: current and future perspectives. Nat. Rev. Gastroenterol. Hepatol. 15, 333–348 (2018).

    PubMed  Google Scholar 

  3. Dagogo-Jack, I. & Shaw, A. T. Tumour heterogeneity and resistance to cancer therapies. Nat. Rev. Clin. Oncol. 15, 81–94 (2017).

    PubMed  Google Scholar 

  4. Maddipati, R. & Stanger, B. Z. Pancreatic cancer metastases harbor evidence of polyclonality. Cancer Discov. 5, 1086–1097 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Kreso, A. & Dick, J. E. Evolution of the cancer stem cell model. Cell Stem Cell 14, 275–291 (2014).

    CAS  PubMed  Google Scholar 

  6. Li, C. et al. Identification of pancreatic cancer stem cells. Cancer Res. 67, 1030–1037 (2007).

    CAS  PubMed  Google Scholar 

  7. Li, C. et al. c-Met is a marker of pancreatic cancer stem cells and therapeutic target. Gastroenterology 141, 2218–2227 (2011).

    CAS  PubMed  Google Scholar 

  8. Hermann, P. C. et al. Distinct populations of cancer stem cells determine tumor growth and metastatic activity in human pancreatic cancer. Cell Stem Cell 1, 313–323 (2007).

    CAS  PubMed  Google Scholar 

  9. Fox, R. G. et al. Image-based detection and targeting of therapy resistance in pancreatic adenocarcinoma. Nature 534, 407–411 (2016).

    PubMed  PubMed Central  Google Scholar 

  10. Bailey, J. M. et al. DCLK1 marks a morphologically distinct subpopulation of cells with stem cell properties in preinvasive pancreatic cancer. Gastroenterology 146, 245–256 (2014).

    CAS  PubMed  Google Scholar 

  11. Abel, E. V. et al. HNF1A is a novel oncogene that regulates human pancreatic cancer stem cell properties. eLife 7, e33947 (2018).

    PubMed  PubMed Central  Google Scholar 

  12. Barker, N. et al. Crypt stem cells as the cells-of-origin of intestinal cancer. Nature 457, 608–611 (2009).

    CAS  PubMed  Google Scholar 

  13. Ferreira, R. M. M. et al. Duct- and acinar-derived pancreatic ductal adenocarcinomas show distinct tumor progression and marker expression. Cell Rep. 21, 966–978 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Went, P. T. et al. Frequent EpCam protein expression in human carcinomas. Hum. Pathol. 35, 122–128 (2004).

    CAS  PubMed  Google Scholar 

  15. Cruz-Monserrate, Z. et al. Detection of pancreatic cancer tumours and precursor lesions by cathepsin E activity in mouse models. Gut 61, 1315–1322 (2012).

    CAS  PubMed  Google Scholar 

  16. Uhlen, M. et al. Tissue-based map of the human proteome. Science 347, 1260419 (2015).

    PubMed  Google Scholar 

  17. Bardeesy, N. et al. Both p16-Ink4a and the p19-Arf-p53-pathway constrain progression of pancreatic adenocarcinoma in the mouse. Proc. Natl Acad. Sci. USA 103, 5947–5952 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Hemler, M. E. Tetraspanin proteins promote multiple cancer stages. Nat. Rev. Cancer 14, 49–60 (2014).

    CAS  PubMed  Google Scholar 

  19. Huch, M. et al. Unlimited in vitro expansion of adult bi‐potent pancreas progenitors through the Lgr5/R‐spondin axis. EMBO J. 32, 2708–2721 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Malanchi, I. et al. Interactions between cancer stem cells and their niche govern metastatic colonization. Nature 481, 85–89 (2012).

    CAS  Google Scholar 

  21. Reya, T., Morrison, S. J., Clarke, M. F. & Weissman, I. L. Stem cells, cancer, and cancer stem cells. Nature 414, 105–111 (2001).

    CAS  PubMed  Google Scholar 

  22. Hotz, B. et al. Epithelial to mesenchymal transition: expression of the regulators snail, slug, and twist in pancreatic cancer. Clin. Cancer Res. 13, 4769–4776 (2007).

    CAS  PubMed  Google Scholar 

  23. Zheng, X. et al. Epithelial-to-mesenchymal transition is dispensable for metastasis but induces chemoresistance in pancreatic cancer. Nature 527, 525–530 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Rhim, A. D. et al. EMT and dissemination precede pancreatic tumor formation. Cell 148, 349–361 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Miyado, K. et al. The fusing ability of sperm is bestowed by CD9-containing vesicles released from eggs in mice. Proc. Nal Acad. Sci. USA 105, 12921–12926 (2008).

    CAS  Google Scholar 

  26. Skarnes, W. C. et al. A conditional knockout resource for the genome-wide study of mouse gene function. Nature 474, 337–342 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Charrin, S. et al. The major CD9 and CD81 molecular partner: identification and characterization of the complexes. J. Biol. Chem. 276, 14329–14337 (2001).

    CAS  PubMed  Google Scholar 

  28. Le Naour, F. et al. Profiling of the tetraspanin web of human colon cancer cells. Mol. Cell. Proteom. 5, 845–857 (2006).

    Google Scholar 

  29. Kovalenko, O. V., Yang, X., Kolesnikova, T. V. & Hemler, M. E. Evidence for specific tetraspanin homodimers: inhibition of palmitoylation makes cysteine residues available for cross-linking. Biochem. J. 377, 407–417 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Xu, D. & Hemler, M. E. Metabolic activation-related CD147–CD98 complex. Mol. Cell. Proteom. 4, 1061–1071 (2005).

    CAS  Google Scholar 

  31. Son, J. et al. Glutamine supports pancreatic cancer growth through a KRAS-regulated metabolic pathway. Nature 496, 101–105 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Schonhuber, N. et al. A next-generation dual-recombinase system for time- and host-specific targeting of pancreatic cancer. Nat. Med. 20, 1340–1347 (2014).

    PubMed  PubMed Central  Google Scholar 

  33. Kaira, K. et al. Clinicopathological significance of ASC amino acid transporter-2 expression in pancreatic ductal carcinoma. Histopathology 66, 234–243 (2015).

    PubMed  Google Scholar 

  34. Biancur, D. E. et al. Compensatory metabolic networks in pancreatic cancers upon perturbation of glutamine metabolism. Nat. Commun. 8, 15965 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Schulte, M. L. et al. Pharmacological blockade of ASCT2-dependent glutamine transport leads to antitumor efficacy in preclinical models. Nat. Med. 24, 194–202 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Cerami, E. et al. The cBio Cancer Genomics Portal: an open platform for exploring multidimensional cancer genomics data. Cancer Discov. 2, 401–404 (2012).

    PubMed  Google Scholar 

  37. Witkiewicz, A. K. et al. Whole-exome sequencing of pancreatic cancer defines genetic diversity and therapeutic targets. Nat. Commun. 6, 6744 (2015).

    CAS  PubMed  Google Scholar 

  38. Mueller, S. et al. Evolutionary routes and KRAS dosage define pancreatic cancer phenotypes. Nature 554, 62–68 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Kim, M. P. et al. ALDH activity selectively defines an enhanced tumor-initiating cell population relative to CD133 expression in human pancreatic adenocarcinoma. PLoS ONE 6, e20636 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Miranda-Lorenzo, I. et al. Intracellular autofluorescence: a biomarker for epithelial cancer stem cells. Nat. Methods 11, 1161–1169 (2014).

    CAS  PubMed  Google Scholar 

  41. Dosch, J. S., Ziemke, E. K., Shettigar, A., Rehemtulla, A. & Sebolt-Leopold, J. S. Cancer stem cell marker phenotypes are reversible and functionally homogeneous in a preclinical model of pancreatic cancer. Cancer Res. 75, 4582–4592 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Zeng, A. et al. Prospectively isolated tetraspanin-positive neoblasts are adult pluripotent stem cells underlying planaria regeneration. Cell 173, 1593–1608 (2018).

    CAS  PubMed  Google Scholar 

  43. Kwon, H. Y. et al. Tetraspanin 3 is required for the development and propagation of acute myelogenous leukemia. Cell Stem Cell 17, 152–164 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Wang, G. P. & Han, X. F. CD9 modulates proliferation of human glioblastoma cells via epidermal growth factor receptor signaling. Mol. Med. Rep. 12, 1381–1386 (2015).

    CAS  PubMed  Google Scholar 

  45. Copeland, B. T., Bowman, M. J., Boucheix, C. & Ashman, L. K. Knockout of the tetraspanin Cd9 in the TRAMP model of de novo prostate cancer increases spontaneous metastases in an organ-specific manner. Int. J. Cancer 133, 1803–1812 (2013).

    CAS  PubMed  Google Scholar 

  46. Tang, M. et al. Downregulation of CD9 promotes pancreatic cancer growth and metastasis through upregulation of epidermal growth factor on the cell surface. Oncology Rep. 34, 350–358 (2015).

    CAS  Google Scholar 

  47. Grønborg, M. et al. Biomarker discovery from pancreatic cancer secretome using a differential proteomic approach. Mol. Cell. Proteom. 5, 157–171 (2006).

    Google Scholar 

  48. Shi, Y. et al. Tetraspanin CD9 stabilizes gp130 by preventing its ubiquitin-dependent lysosomal degradation to promote STAT3 activation in glioma stem cells. Cell Death Differ. 24, 167–180 (2017).

    Google Scholar 

  49. Podergajs, N. et al. Transmembrane protein CD9 is glioblastoma biomarker, relevant for maintenance of glioblastoma stem cells. Oncotarget 7, 593–609 (2016).

    PubMed  Google Scholar 

  50. Yamazaki, H. et al. Regulation of cancer stem cell properties by CD9 in human B-acute lymphoblastic leukemia. Biochem. Biophys. Res. Commun. 409, 14–21 (2011).

    CAS  PubMed  Google Scholar 

  51. Kamphorst, J. J. et al. Human pancreatic cancer tumors are nutrient poor and tumor cells actively scavenge extracellular protein. Cancer Res. 75, 544–553 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Roux, C. et al. Endogenous glutamine decrease is associated with pancreatic cancer progression. Oncotarget 8, 95361–95376 (2017).

    PubMed  PubMed Central  Google Scholar 

  53. Seo, J.-W. et al. Autophagy is required for PDAC glutamine metabolism. Sci. Rep. 6, 37594 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Commisso, C. et al. Macropinocytosis of protein is an amino acid supply route in Ras-transformed cells. Nature 497, 633–637 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Boj, S. F. et al. Organoid models of human and mouse ductal pancreatic cancer. Cell 160, 324–338 (2015).

    CAS  PubMed  Google Scholar 

  56. Means, A. L., Xu, Y., Zhao, A., Ray, K. C. & Gu, G. A. CK19(CreERT) knockin mouse line allows for conditional DNA recombination in epithelial cells in multiple endodermal organs. Genesis 46, 318–323 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Jandke, A. et al. The F-box protein Fbw7 is required for cerebellar development. Devel. Biol. 358, 201–212 (2011).

    CAS  Google Scholar 

  58. Jackson, E. L. et al. Analysis of lung tumor initiation and progression using conditional expression of oncogenic K-ras. Genes Devel. 15, 3243–3248 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  59. Marino, S., Vooijs, M., van Der Gulden, H., Jonkers, J. & Berns, A. Induction of medulloblastomas in p53-null mutant mice by somatic inactivation of Rb in the external granular layer cells of the cerebellum. Genes Devel. 14, 994–1004 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  60. Hingorani, S. R. et al. Preinvasive and invasive ductal pancreatic cancer and its early detection in the mouse. Cancer Cell 4, 437–450 (2003).

    CAS  PubMed  Google Scholar 

  61. Srinivas, S. et al. Cre reporter strains produced by targeted insertion of EYFP and ECFP into the ROSA26 locus. BMC Devel. Biol. 1, 4 (2001).

    CAS  Google Scholar 

  62. Flanagan, S. P. ‘Nude’, a new hairless gene with pleiotropic effects in the mouse. Genet. Res. 8, 295–309 (1966).

    CAS  PubMed  Google Scholar 

  63. Hayer, A. et al. Engulfed cadherin fingers are polarized junctional structures between collectively migrating endothelial cells. Nat. Cell Biol. 18, 1311–1323 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  64. Tyanova, S., Temu, T. & Cox, J. The MaxQuant computational platform for mass spectrometry-based shotgun proteomics. Nat. Protoc. 11, 2301–2319 (2016).

    CAS  PubMed  Google Scholar 

  65. MacRae, J. I. et al. Mitochondrial metabolism of sexual and asexual blood stages of the malaria parasite Plasmodium falciparum. BMC Biol. 11, 67 (2013).

    PubMed  PubMed Central  Google Scholar 

  66. Behrends, V., Tredwell, G. D. & Bundy, J. G. A software complement to AMDIS for processing GC-MS metabolomic data. Anal. Biochem. 415, 206–208 (2011).

    CAS  PubMed  Google Scholar 

  67. Zhang, T., Creek, D. J., Barrett, M. P., Blackburn, G. & Watson, D. G. Evaluation of coupling reversed phase, aqueous normal phase, and hydrophilic interaction liquid chromatography with Orbitrap mass spectrometry for metabolomic studies of human urine. Anal. Chem. 84, 1994–2001 (2012).

    CAS  PubMed  Google Scholar 

  68. Rey, G. et al. Metabolic oscillations on the circadian time scale in Drosophila cells lacking clock genes. Mol. Syst. Biol. 14, e8376 (2018).

    PubMed  PubMed Central  Google Scholar 

  69. Chong, J., Yamamoto, M. & Xia, J. MetaboAnalystR 2.0: from raw spectra to biological insights. Metabolites 9, e57 (2019).

    PubMed  Google Scholar 

  70. Barry, D. J., Durkin, C. H., Abella, J. V. & Way, M. Open source software for quantification of cell migration, protrusions, and fluorescence intensities. J. Cell Biol. 209, 163–180 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank D. Saur for providing the Pdx1-Flp, Trp53 FRT and FSF-KRasG12D mice, I. Malanchi and M. Yuneva for comments on the manuscript, C. Cremona for help in preparing the manuscript, the Yuneva laboratory for advice on organoid metabolomics and the V-9302 inhibitor, R. Mitter and S. Boeing from the Bioinformatics and Statistics Service at the Francis Crick Institute, K. Miyado for the EGFP−mCD9 fusion construct, D. Sarker, A. Prachalias, C. Cotoi, Y. Zen and A. Zamalloa for coordinating and providing the human samples used in this study, technicians from the Biological Research Facility at the Francis Crick Institute, and all other Behrens laboratory members for their feedback. T.E. was supported by a doctoral clinical fellowship from a CRUK Accelerator Award (C422/A23614). This work was supported by the Francis Crick Institute, which receives its core funding from Cancer Research UK (FC001039), the UK Medical Research Council (FC001039) and the Wellcome Trust (FC001039).

Author information

Authors and Affiliations

Authors

Contributions

R.M.M.F., V.M.-Y.W. and A.B. conceptualised this study and designed experiments. R.M.M.F., V.M.-Y.W., J.A., T.E., M.Z.T., N.L., D.F. and J.C. performed experiments. R.M.M.F., V.M.-Y.W., D.J.B., N.L., A.P.S., E.H., E.L.N. and J.I.M. analysed data. R.M.M.F. and V.M.-Y.W. wrote the manuscript and prepared figures. R.M.M.F., V.M.-Y.W., J.I.M. and A.B. reviewed and edited the manuscript. A.B. acquired funding and supervised this study. All authors approved the content of the manuscript. J.A., T.E., M.Z.T. and N.L. contributed equally.

Corresponding author

Correspondence to Axel Behrens.

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.

Extended data

Extended Data Fig. 1 CD9 identification.

a, YFP stain of pancreatic ducts of R26-LSL-YFP; Ck19-CreER mice 2 weeks post-tamoxifen. Scale bar, 50 μm, inset 50 µm x 50 µm. b, Flow cytometry analysis and genotyping of unrecombined and recombined cells from KFCkY pancreas 2 weeks post-tamoxifen. Expected band sizes (base pairs) are indicated; see Source Data for uncropped gels. c, CD44 and CD133 stain in consecutive pancreatic sections of KFCkY mice 2 weeks post-tamoxifen. Non-responsive regions magnified. Black arrows show apical expression of CD133 in non-responsive cells. Scale bar, 50 µm. d, CD44 gating strategy using rat IgG2b isotype control on KFCkY pancreas 2 weeks post-tamoxifen. Related to Fig. 1d. e, Validation of the CD44 antibody using rat IgG2b isotype control on KFCkY tumours; quantification of CD44+ cells. f, Quantification of transformed/CD44+ YFP+ foci in pancreatic tissues of KFCkY mice two (n = 5) and four (n = 3 biologically independent animals) weeks post-tamoxifen. Dot plot shows mean, s.d. g, Human PDAC markers among upregulated genes in microarray. Fold changes relative to NT cells. h, Flow cytometry analysis of CD44 and CD9 expression on live, YFP+ cells from KFCkY pancreas 4 weeks post-tamoxifen. i, Quantification of live tumour cells (DAPIYFP+, n = 3 biologically independent animals) from (h). Bar chart shows mean, s.d., p = 0.0013, two-sided t test. j, CD9, E-cadherin and DAPI staining on pancreatic tissues of KFCkY mice 4 weeks post-tamoxifen. Scale bar, 50 µm, 10 μm (inset). k, CD9 gating strategy using rat IgG2a isotype control on live KPCY tumour cells. Related to Fig. 1i. l, Validation of the CD9 antibody using rat IgG2a isotype control in live KPCY PDAC organoids; quantification of CD9+ cells. All flow cytometry plots and immuno-staining data shown in Extended Data Fig. 1 are representative of at least three biologically independent animals/experiments.

Source data

Source data

Extended Data Fig. 2 CD9 marks TICs.

a, Organoid formation assay (organoid number) of CD9high and CD9low YFP+ tumor cells freshly isolated from KFCkY PDAC (n = 4 biologically independent animals). Dot plot shows mean, s.d., p value from two-sided Mann-Whitney test. Right, representative brightfield images of generated organoids 10 days after plating. Scale bar, 250 µm. b, CD9 expression dynamics of two representative KPCY organoid cultures. Organoid cultures were dissociated at confluency and replated. The percentage of CD9high cells was measured by flow cytometry over time as the cultures grew. c, Flow cytometry analysis of a representative KPCY organoid culture at 4 and 8 days of culture, stained for CD9 and the proliferation marker Ki67. Proliferation is higher at 4 than 8 days, and only CD9high cells actively proliferate in the culture at both timepoints. Flow cytometry plots shown in (b) and (c) are representative of at least three biologically independent samples. d, Tumourigenic capacity of CD9high versus CD9low tumour cells from KPCY PDAC-derived organoids. 200,000, 20,000, 2,000 or 200 sorted tumour cells were injected into the flanks of Nu/Nu mice and the number of weeks to nodule detection (4 mm3) was registered. e, Bar charts of percentage of mice injected with 2,000 (above) or 200 (below) sorted CD9high or CD9low tumour cells that developed tumours by the given week. Each bar shows cumulative data of all mice harbouring tumours at a given week (n = 6 for 2,000 cells, n = 4 for 200 cells). f, Fold difference in tumour volumes between the CD9low- and CD9high-derived tumours at each dilution; n = 3, 5, 6 and 4 biologically independent animals for 200,000-, 20,000-, 2,000- and 200-cell injections, respectively. Dot plot shows mean, s.d., p values from two-sided t tests.

Source data

Extended Data Fig. 3 CD9 is required for efficient PDAC formation.

a, Organoid formation assay (total organoid area) of KPC organoids upon CD9 knockdown (representative of n = 3 biologically independent experiments). Dot plot shows mean, s.d., p values from two-sided t test. Related to Fig. 4b. b, Flow cytometry plot of HEK293T cells transiently transfected with EGFP control and EGFP-mCD9 vectors. The KMC8 antibody was used to specifically recognise murine CD9. c, Live confocal microscopy images showing cytoplasmic EGFP and predominantly plasma membrane EGFP-mCD9 localisation in primary KPC cells. Scale bar, 20 µm. Flow cytometry plots and fluorescence imaging shown in (b) and (c) are representative of three biologically independent samples. d, Organoid formation assay (total organoid area) of KPC organoids upon CD9 overexpression (n = 3 biologically independent experiments). Related to Fig. 4e. Dot plot shows mean, s.d., p < 0.0001, two-sided t test. e, Schematic of the EGFP control and EGFP-mCD9 overexpression system using an EF1α promoter. f, Organoid formation assay (organoid number) of KPC organoids upon CD9 overexpression using the EF1α vector (n = 2 biologically independent experiments). p = 0.001, two-sided t test. g, Organoid formation assay (total organoid area) of KPC organoids upon CD9 overexpression using the EF1α vector (n = 2 biologically independent experiments). p = 0.002, two-sided t test. Dot plots in (f) and (g) show mean, s.d.

Source data

Extended Data Fig. 4 Characterisation of KPC CD9 knock-out model.

a, Flow cytometry histogram of cells isolated directly from mouse pancreas of the indicated genotypes after gating on YFP+ cells. CD9 staining confirms heterozygous and complete knockout in CD9Δ/WT and CD9Δ/Δ cells, respectively. Flow cytometry plot is representative of three biologically independent samples. b, Cd9 mRNA levels measured by RT-qPCR in early passage (<5) KPC organoids of the indicated genotypes (n = 2 biologically independent animals per genotype); cells were isolated from endpoint tumours. Values were normalised to Gapdh and Actb, and fold changes calculated relative to CD9WT/WT cells. c, Representative histology of KPC pancreases at 8 weeks with indicated CD9 status (in the p53Δ/WT model, median survival of CD9WT/WT mice 17.4 weeks; n = 6, 4 and 8 biologically independent animals for CD9WT/WT, CD9Δ/WT and CD9Δ/Δ, respectively). d, Stacked bar graph showing quantification of precursor PanIN and PDAC lesions in KPC pancreases at 8 weeks from (c). KPC CD9Δ/WT mice exhibit later onset and/or slower tumor progression. e, Weight of subcutaneous grafts at endpoint after initial injection of 10,000 KPC CD9Δ/WT or KPC CD9Δ/Δ cells (n = 3 biologically independent animals, from organoid cultures derived from the p53Δ/Δ model at 4 weeks). P = 0.0252, two-sided t test. Dot plot shows mean, s.d.

Source data

Extended Data Fig. 5 CD9 facilitates glutamine import into TICs.

a, Co-IP of transiently transfected Myc-hMCT1 (left) or Myc-mASCT2 (right) with EGFP-mCD9 after EGFP pull-down in HEK293T cells. Blots are representative of 3 independent experiments. See Source Data for uncropped blots. b, Flow cytometry plots co-staining CD9 with cell surface proteins involved in metabolism in KPC organoids. Representative of 3 independent experiments. c, Membrane co-localisation (arrowheads) of ASCT2 and EGFP-CD9 in KPC organoids by co-immunofluorescence. Scale bar, 20 µm; insert 20 by 20 µm. d, Immunofluorescence of ASCT on sorted CD9low and CD9high KPC cells. Scale bars, 20 µm. Representative of 3 independent experiments. e, Immunofluorescence of KPF-CD9 organoids after control vehicle or 4-OHT treatment. Representative of 2 independent experiments. Scale bars, 20 µm. Related to Fig. 5e. f, Immunofluorescence of ASCT2 in a KPCY mouse pancreas: non-transformed area (top) and transformed areas (bottom). Scale bars, 100 µm low mag, 20 µm high mag. Representative of at least 3 biologically independent animals. g, YFP, CD44, ASCT2 and DAPI staining on pancreatic tissue of KFCkY mice 1 week (top) and 4 weeks (bottom) post-tamoxifen. Scale bars, 20 µm. Representative of at least 3 biologically independent animals. h, Metabolic profiles of KPF-CD9 organoids (after control Adeno-EGFP (CD9F/WT) or Adeno-CRE-EGFP (CD9Δ/WT) infection) grown in organoid media containing 4 mM 13C-glutamine for 4 h. i, Some metabolites, including glycolytic intermediates, are equally abundant under the conditions in (h), but did not contain any detectable 13C label. Data in (h) and (i) represent one experiment carried out with six technical replicates. Due to technical limitations, statistics for metabolite abundance and label incorporation were performed on technical replicates separately using a two-way ANOVA corrected for multiple comparisons using the Holm-Sidak method, α = 0.05; bar charts show mean, s.d.

Source data

Source data

Extended Data Fig. 6 CD9 facilitates glutamine import into TICs and metabolic compensation in KPC mice.

a, Western blot showing overexpression of ASCT2 in organoids. Blot shown is representative of 2 independent experiments. See Source Data for uncropped blots. b, Organoid formation assay (total organoid area) of KPF-CD9 organoids after vehicle/Adeno-EGFP or 4-OHT/Adeno-CRE-EGFP treatment in the presence of basal or elevated (4 mM) levels of glutamine, and with control pLV or pLV ASCT2 overexpression (n = 3 biologically independent experiments). Related to Fig. 5h. Black P values (CD9F/WT versus CD9Δ/WT) calculated using two-way ANOVA corrected for multiple comparisons, Holm-Sidak method, α = 0.05. Blue P values (within CD9Δ/WT conditions) calculated using two-way ANOVA corrected for multiple comparisons, Dunnett’s test, α = 0.05. Dot plot shows mean, s.d. c, Organoid formation assay (total organoid area) of CD9low KPC cells with control pLV or pLV ASCT2 overexpression (n = 3 biologically independent experiments). Related to Fig. 5i. P = 0.0091, two-sided t test. Dot plot shows mean, s.d. d, Histology of KPC pancreases at 4 weeks with indicated CD9 status (in the p53Δ/Δ model, median survival of CD9WT/WT mice 6.2 weeks). At this timepoint, all mice present with PDAC, regardless of CD9 status. Images are representative of at least 3 biologically independent animals. e, Slc1a5 mRNA levels measured by RT-qPCR in early passage (<5) KPC organoids of the indicated genotypes (n = 2 biologically independent animals per genotype); cells were isolated from endpoint tumors. Values normalised to Gapdh and Actb; fold changes calculated relative to CD9WT/WT cells. f, Indicated tetraspanin mRNA levels measured by RT-qPCR in early passage (<5) KPC organoids of the indicated genotypes (n = 2 biologically independent animals per genotype); cells were isolated from endpoint tumours. Values normalised to Gapdh and Actb; fold changes calculated relative to CD9WT/WT cells.

Source data

Source data

Extended Data Fig. 7 CD9 expression in human PDAC.

a, Co-IP of endogenous CD9 with endogenous ASCT2 after ASCT2 pull-down in the human pancreatic cancer cell line PANC-1. Input, left, 10% polyacrylamide gel; Co-IP, right, 20% polyacrylamide gel to separate bands from IgG heavy and light chains (asterisks). Blot representative of 2 independent experiments. See Source Data for uncropped blots. b, Co-immunofluorescence for CD9 and ASCT2 on human PDAC, showing punctate CD9 at the plasma membrane, with high levels of ASCT2 in CD9high cells; scale bars, 20 µm. Data are representative of 3 biologically independent human samples. c, Flow cytometry plots co-staining CD9 with other PDAC CSC cell surface proteins in primary human PDAC organoid culture. CD9 gate separates CD9high cells from bulk; gates on the x-axis separate marker-positive from marker-negative cells (based on fluorescence minus one controls). Plots are representative of 3 biologically independent primary organoid lines.

Source data

Supplementary information

Source data

Source Data Fig. 1

Statistical source data

Source Data Fig. 1

Unprocessed gels

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. 5

Unprocessed Western blots

Source Data Fig. 6

Statistical source data

Source Data Extended Data Fig. 1

Statistical source data

Source Data Extended Data Fig. 1

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

Statistical source data

Source Data Extended Data Fig. 5

Statistical source data

Source Data Extended Data Fig. 5

Unprocessed western blots

Source Data Extended Data Fig. 6

Statistical source data

Source Data Extended Data Fig. 6

Unprocessed western blots

Source Data Extended Fig. 7

Unprocessed western blots

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wang, V.MY., Ferreira, R.M.M., Almagro, J. et al. CD9 identifies pancreatic cancer stem cells and modulates glutamine metabolism to fuel tumour growth. Nat Cell Biol 21, 1425–1435 (2019). https://doi.org/10.1038/s41556-019-0407-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41556-019-0407-1

This article is cited by

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