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:

Pancreatic cancer exosomes initiate pre-metastatic niche formation in the liver

Abstract

Pancreatic ductal adenocarcinomas (PDACs) are highly metastatic with poor prognosis, mainly due to delayed detection. We hypothesized that intercellular communication is critical for metastatic progression. Here, we show that PDAC-derived exosomes induce liver pre-metastatic niche formation in naive mice and consequently increase liver metastatic burden. Uptake of PDAC-derived exosomes by Kupffer cells caused transforming growth factor β secretion and upregulation of fibronectin production by hepatic stellate cells. This fibrotic microenvironment enhanced recruitment of bone marrow-derived macrophages. We found that macrophage migration inhibitory factor (MIF) was highly expressed in PDAC-derived exosomes, and its blockade prevented liver pre-metastatic niche formation and metastasis. Compared with patients whose pancreatic tumours did not progress, MIF was markedly higher in exosomes from stage I PDAC patients who later developed liver metastasis. These findings suggest that exosomal MIF primes the liver for metastasis and may be a prognostic marker for the development of PDAC liver metastasis.

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

Figure 1: PDAC-derived exosomes target and activate KCs, induce liver fibrosis pathways, and increase liver metastasis.
Figure 2: Role of PDAC-derived exosome education in ECM component expression and liver pre-metastatic niche formation.
Figure 3: PDAC-derived exosomes induce FN expression and migration of bone marrow-derived cells to the liver.
Figure 4: TGFβ signalling induces FN upregulation and macrophage recruitment to the liver pre-metastatic niche.
Figure 5: FN and macrophages play major roles in PAN02 exosome-mediated liver pre-metastatic niche formation.
Figure 6: MIF-expressing PAN02 exosomes induce liver pre-metastatic niche formation.
Figure 7: Liver pre-metastatic niche formation and increased exosomal MIF levels precede PDAC lesion development in PKCY mice.
Figure 8: Model for the sequential steps in liver pre-metastatic niche formation induced by PDAC-derived exosomes.

Similar content being viewed by others

References

  1. Saif, M. W. Pancreatic neoplasm in 2011: an update. JOP 12, 316–321 (2011).

    PubMed  Google Scholar 

  2. Chan, A., Diamandis, E. P. & Blasutig, I. M. Strategies for discovering novel pancreatic cancer biomarkers. J. Proteomics 81, 126–134 (2013).

    Article  CAS  PubMed  Google Scholar 

  3. Fesinmeyer, M. D., Austin, M. A., Li, C. I., De Roos, A. J. & Bowen, D. J. Differences in survival by histologic type of pancreatic cancer. Cancer Epidemiol. Biomarkers Prev. 14, 1766–1773 (2005).

    Article  PubMed  Google Scholar 

  4. Arscott, W. T. & Camphausen, K. A. EGFR isoforms in exosomes as a novel method for biomarker discovery in pancreatic cancer. Biomarkers Med. 5, 821 (2011).

    Article  Google Scholar 

  5. Record, M., Carayon, K., Poirot, M. & Silvente-Poirot, S. Exosomes as new vesicular lipid transporters involved in cell–cell communication and various pathophysiologies. Biochim. Biophys. Acta 1841, 108–120 (2014).

    Article  CAS  PubMed  Google Scholar 

  6. El Andaloussi, S., Mager, I., Breakefield, X. O. & Wood, M. J. Extracellular vesicles: biology and emerging therapeutic opportunities. Nat. Rev. Drug Discov. 12, 347–357 (2013).

    Article  CAS  PubMed  Google Scholar 

  7. Choi, D. S., Kim, D. K., Kim, Y. K. & Gho, Y. S. Proteomics, transcriptomics and lipidomics of exosomes and ectosomes. Proteomics 13, 1554–1571 (2013).

    Article  CAS  PubMed  Google Scholar 

  8. Martins, V. R., Dias, M. S. & Hainaut, P. Tumor-cell-derived microvesicles as carriers of molecular information in cancer. Curr. Opin. Oncol. 25, 66–75 (2013).

    Article  CAS  PubMed  Google Scholar 

  9. Peinado, H., Lavotshkin, S. & Lyden, D. The secreted factors responsible for pre-metastatic niche formation: old sayings and new thoughts. Semin. Cancer Biol. 21, 139–146 (2011).

    Article  CAS  PubMed  Google Scholar 

  10. Thakur, B. K. et al. Double-stranded DNA in exosomes: a novel biomarker in cancer detection. Cell Res. 24, 766–769 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Tetta, C., Ghigo, E., Silengo, L., Deregibus, M. C. & Camussi, G. Extracellular vesicles as an emerging mechanism of cell-to-cell communication. Endocrine 44, 11–19 (2013).

    Article  CAS  PubMed  Google Scholar 

  12. Valadi, H. et al. Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat. Cell Biol. 9, 654–659 (2007).

    Article  CAS  PubMed  Google Scholar 

  13. Zoller, M. Pancreatic cancer diagnosis by free and exosomal miRNA. World J. Gastrointest. Pathophysiol. 4, 74–90 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  14. Kaplan, R. N. et al. VEGFR1-positive haematopoietic bone marrow progenitors initiate the pre-metastatic niche. Nature 438, 820–827 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Sceneay, J., Smyth, M. J. & Moller, A. The pre-metastatic niche: finding common ground. Cancer Metastasis Rev. 32, 449–464 (2013).

    Article  CAS  PubMed  Google Scholar 

  16. Peinado, H. et al. Melanoma exosomes educate bone marrow progenitor cells toward a pro-metastatic phenotype through MET. Nat. Med. 18, 883–891 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Hood, J. L., San, R. S. & Wickline, S. A. Exosomes released by melanoma cells prepare sentinel lymph nodes for tumor metastasis. Cancer Res. 71, 3792–3801 (2011).

    Article  CAS  PubMed  Google Scholar 

  18. Corbett, T. H. et al. Induction and chemotherapeutic response of two transplantable ductal adenocarcinomas of the pancreas in C57BL/6 mice. Cancer Res. 44, 717–726 (1984).

    CAS  PubMed  Google Scholar 

  19. Little, E. C. et al. Novel immunocompetent murine models representing advanced local and metastatic pancreatic cancer. J. Surg. Res. 176, 359–366 (2012).

    Article  CAS  PubMed  Google Scholar 

  20. Hingorani, S. R. et al. Trp53R172H and KrasG12D cooperate to promote chromosomal instability and widely metastatic pancreatic ductal adenocarcinoma in mice. Cancer Cell 7, 469–483 (2005).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Achyut, B. R. & Yang, L. Transforming growth factor-β in the gastrointestinal and hepatic tumor microenvironment. Gastroenterology 141, 1167–1178 (2011).

    Article  CAS  PubMed  Google Scholar 

  23. Hayashi, H. & Sakai, T. Biological significance of local TGF-β activation in liver diseases. Front. Physiol. 3, 12 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Wight, T. N. & Potter-Perigo, S. The extracellular matrix: an active or passive player in fibrosis? Am. J. Physiol. Gastrointest. Liver Physiol. 301, G950–G955 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Gressner, A. M., Weiskirchen, R., Breitkopf, K. & Dooley, S. Roles of TGF-β in hepatic fibrosis. Front. Biosci. 7, d793–d807 (2002).

    Article  CAS  PubMed  Google Scholar 

  26. Cong, M., Iwaisako, K., Jiang, C. & Kisseleva, T. Cell signals influencing hepatic fibrosis. Int. J. Hepatol. 2012, 158547 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  27. Kawelke, N. et al. Fibronectin protects from excessive liver fibrosis by modulating the availability of and responsiveness of stellate cells to active TGF-β. PLoS ONE 6, e28181 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Xu, G. et al. Gene expression and synthesis of fibronectin isoforms in rat hepatic stellate cells. Comparison with liver parenchymal cells and skin fibroblasts. J. Pathol. 183, 90–98 (1997).

    Article  CAS  PubMed  Google Scholar 

  29. Tojo, M. et al. The ALK-5 inhibitor A-83-01 inhibits Smad signaling and epithelial-to-mesenchymal transition by transforming growth factor-β. Cancer Sci. 96, 791–800 (2005).

    Article  CAS  PubMed  Google Scholar 

  30. Duffield, J. S. et al. Selective depletion of macrophages reveals distinct, opposing roles during liver injury and repair. J. Clin. Invest. 115, 56–65 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Lau, C. et al. Role of pancreatic cancer-derived exosomes in salivary biomarker development. J. Biol. Chem. 288, 26888–26897 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Heinrichs, D. et al. Macrophage migration inhibitory factor (MIF) exerts antifibrotic effects in experimental liver fibrosis via CD74. Proc. Natl Acad. Sci. USA 108, 17444–17449 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Barnes, M. A. et al. Macrophage migration inhibitory factor contributes to ethanol-induced liver injury by mediating cell injury, steatohepatitis, and steatosis. Hepatology 57, 1980–1991 (2013).

    Article  CAS  PubMed  Google Scholar 

  34. Funamizu, N. et al. Macrophage migration inhibitory factor induces epithelial to mesenchymal transition, enhances tumor aggressiveness and predicts clinical outcome in resected pancreatic ductal adenocarcinoma. Int. J. Cancer 132, 785–794 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Nanji, A. A. et al. Macrophage migration inhibitory factor expression in male and female ethanol-fed rats. J. Interferon Cytokine Res. 21, 1055–1062 (2001).

    Article  CAS  PubMed  Google Scholar 

  36. Shin, H. N., Moon, H. H. & Ku, J. L. Stromal cell-derived factor-1α and macrophage migration-inhibitory factor induce metastatic behavior in CXCR4-expressing colon cancer cells. Int. J. Mol. Med. 30, 1537–1543 (2012).

    Article  CAS  PubMed  Google Scholar 

  37. Zhang, H. Y. et al. Macrophage migration inhibitory factor expression correlates with inflammatory changes in human chronic hepatitis B infection. Liver Int. 25, 571–579 (2005).

    Article  CAS  PubMed  Google Scholar 

  38. Adamali, H. et al. Macrophage migration inhibitory factor enzymatic activity, lung inflammation, and cystic fibrosis. Am. J. Respir. Crit. Care Med. 186, 162–169 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Kobayashi, S., Nishihira, J., Watanabe, S. & Todo, S. Prevention of lethal acute hepatic failure by antimacrophage migration inhibitory factor antibody in mice treated with bacille Calmette-Guerin and lipopolysaccharide. Hepatology 29, 1752–1759 (1999).

    Article  CAS  PubMed  Google Scholar 

  40. Yaddanapudi, K. et al. Control of tumor-associated macrophage alternative activation by macrophage migration inhibitory factor. J. Immunol. 190, 2984–2993 (2013).

    Article  CAS  PubMed  Google Scholar 

  41. Chen, P. F. et al. ISO-1, a macrophage migration inhibitory factor antagonist, inhibits airway remodeling in a murine model of chronic asthma. Mol. Med. 16, 400–408 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Javle, M. et al. Biomarkers of TGF-β signaling pathway and prognosis of pancreatic cancer. PLoS ONE 9, e85942 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  43. Ellermeier, J. et al. Therapeutic efficacy of bifunctional siRNA combining TGF-β1 silencing with RIG-I activation in pancreatic cancer. Cancer Res. 73, 1709–1720 (2013).

    Article  CAS  PubMed  Google Scholar 

  44. Gaspar, N. J. et al. Inhibition of transforming growth factor β signaling reduces pancreatic adenocarcinoma growth and invasiveness. Mol. Pharmacol. 72, 152–161 (2007).

    Article  CAS  PubMed  Google Scholar 

  45. Melisi, D. et al. LY2109761, a novel transforming growth factor β receptor type I and type II dual inhibitor, as a therapeutic approach to suppressing pancreatic cancer metastasis. Mol. Cancer Ther. 7, 829–840 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Pickup, M., Novitskiy, S. & Moses, H. L. The roles of TGFβ in the tumour microenvironment. Nat. Rev. Cancer 13, 788–799 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Ijichi, H. et al. Aggressive pancreatic ductal adenocarcinoma in mice caused by pancreas-specific blockade of transforming growth factor-β signaling in cooperation with active Kras expression. Genes Dev. 20, 3147–3160 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Hezel, A. F. et al. TGF-β and αvβ6 integrin act in a common pathway to suppress pancreatic cancer progression. Cancer Res. 72, 4840–4845 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Bayon, L. G. et al. Role of Kupffer cells in arresting circulating tumor cells and controlling metastatic growth in the liver. Hepatology 23, 1224–1231 (1996).

    Article  CAS  PubMed  Google Scholar 

  50. Kruse, J. et al. Macrophages promote tumour growth and liver metastasis in an orthotopic syngeneic mouse model of colon cancer. Int. J. Colorectal Dis. 28, 1337–1349 (2013).

    Article  CAS  PubMed  Google Scholar 

  51. Wen, S. W., Ager, E. I. & Christophi, C. Bimodal role of Kupffer cells during colorectal cancer liver metastasis. Cancer Biol. Ther. 14, 606–613 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Grzesiak, J. J. et al. Knockdown of the β(1) integrin subunit reduces primary tumor growth and inhibits pancreatic cancer metastasis. Int. J. Cancer 129, 2905–2915 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Saito, N. et al. Inhibition of hepatic metastasis in mice treated with cell-binding domain of human fibronectin and angiogenesis inhibitor TNP-470. Int. J. Clin. Oncol. 6, 215–220 (2001).

    Article  CAS  PubMed  Google Scholar 

  54. Zvibel, I., Halpern, Z. & Papa, M. Extracellular matrix modulates expression of growth factors and growth-factor receptors in liver-colonizing colon-cancer cell lines. Int. J. Cancer 77, 295–301 (1998).

    Article  CAS  PubMed  Google Scholar 

  55. Porembka, M. R. et al. Pancreatic adenocarcinoma induces bone marrow mobilization of myeloid-derived suppressor cells which promote primary tumor growth. Cancer Immunol. Immunother. 61, 1373–1385 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Yamamoto, M. et al. TSU68 prevents liver metastasis of colon cancer xenografts by modulating the premetastatic niche. Cancer Res. 68, 9754–9762 (2008).

    Article  CAS  PubMed  Google Scholar 

  57. Zhang, Y., Davis, C., Ryan, J., Janney, C. & Pena, M. M. Development and characterization of a reliable mouse model of colorectal cancer metastasis to the liver. Clin. Exp. Metastasis 30, 903–918 (2013).

    Article  PubMed  Google Scholar 

  58. Seubert, B. et al. Tissue inhibitor of metalloproteinases (TIMP)-1 creates a premetastatic niche in the liver through SDF-1/CXCR4-dependent neutrophil recruitment in mice. Hepatology 61, 238–248 (2015).

    Article  CAS  PubMed  Google Scholar 

  59. Kato, R. et al. A new type of antimetastatic peptide derived from fibronectin. Clin. Cancer Res. 8, 2455–2462 (2002).

    CAS  PubMed  Google Scholar 

  60. Bissell, D. M. Therapy for hepatic fibrosis: revisiting the preclinical models. Clin. Res. Hepatol. Gastroenterol. 35, 521–525 (2011).

    Article  PubMed  Google Scholar 

  61. Korpal, M. & Kang, Y. Targeting the transforming growth factor-β signalling pathway in metastatic cancer. Eur. J. Cancer 46, 1232–1240 (2010).

    Article  CAS  PubMed  Google Scholar 

  62. Noy, R. & Pollard, J. W. Tumor-associated macrophages: from mechanisms to therapy. Immunity 41, 49–61 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Gu, G., Brown, J. R. & Melton, D. A. Direct lineage tracing reveals the ontogeny of pancreatic cell fates during mouse embryogenesis. Mech. Dev. 120, 35–43 (2003).

    Article  CAS  PubMed  Google Scholar 

  64. Zhong, S. et al. High throughput illuma strand-specific RNA sequencing library preparation. Cold Spring Harb. Protoc. 2011, 940–949 (2011).

    Article  PubMed  Google Scholar 

  65. Sakai, T. et al. Plasma fibronectin supports neuronal survival and reduces brain injury following transient focal cerebral ischemia but is not essential for skin-wound healing and hemostasis. Nat. Med. 7, 324–330 (2001).

    Article  CAS  PubMed  Google Scholar 

  66. Suemizu, H. et al. A versatile technique for the in vivo imaging of human tumor xenografts using near-infrared fluorochrome-conjugated macromolecule probes. PLoS ONE 8, e82708 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  67. Morikawa, K., Walker, S. M., Jessup, J. M. & Fidler, I. J. In vivo selection of highly metastatic cells from surgical specimens of different primary human colon carcinomas implanted into nude mice. Cancer Res. 48, 1943–1948 (1988).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank D. L. Bajor (Vonderheide laboratory, University of Pennsylvania) for the gift of the R6560B cells. We thank L. Bojmar for carefully reviewing the paper. We thank S. Rudchenko and M. Barbu-Stevanovic at the Hospital for Special Surgery Fannie E. Rippel Foundation Flow Cytometry Core Facility for expert flow cytometry. We are supported by grants from the Children’s Cancer and Blood Foundation (H.P., D.L.), Manning Foundation (D.L.), Hartwell Foundation (D.L.), Champalimaud Foundation (D.L.), Fundacao para a Ciencia e a Tecnologia (D.L.), Nancy C and Daniel P Paduano Foundation (H.P., D.L.), Mary Kay Foundation (D.L.), Pediatric Oncology Experimental Therapeutic Investigator Consortium (D.L.), James Paduano Foundation (D.L., H.P.), Melanoma Research Alliance (H.P.), Sohn Conference Foundation (H.P.), Beth Tortolani Foundation (D.L., J.B.), Malcolm Hewitt Weiner Foundation (D.L.), Jose Carreras Leukemia Foundation (B.K.T.), Theodore Rapp Foundation (D.L.), American Hellenic Educational Progressive Association 5th District Cancer Research Foundation (D.L.), Charles and Marjorie Holloway Foundation (J.B.), Sussman Family Fund (J.B.), Lerner Foundation (J.B.), Breast Cancer Alliance (J.B.), and Manhasset Women’s Coalition Against Breast Cancer (J.B.).

Author information

Authors and Affiliations

Authors

Contributions

B.C-S. developed the hypothesis, designed the experimental approach, performed experiments, analysed the data, coordinated the project and wrote the manuscript. N.M.A. conducted experiments. S.S., G.R. and T-L.S. performed immunostaining. H.Z. and Y.H. extracted RNA. B.K.T. and A.B. performed western blots. A.H., T-M.T., C.W. and Y.A. maintained mouse colonies. M.T.M. and H.M. performed proteomic analysis. J.X. and T.Z. processed samples and analysed RNA sequencing data. G.G-S. and C.W. processed human samples. P.M.G., M.A.H., K.J.L., I.M.B.L., E.H.K., A.J.O., J.H., A.D., M.J., K.M., S.K.B. and W.R.J. collected patient samples and managed clinical records. S.H.E. contributed to writing the manuscript. R.E.S., I.M., H.P. and B.Z.S. contributed to hypothesis generation, experimental design and data interpretation. J.B. coordinated the project and interpreted data. D.L. conceived the hypothesis, led the project, interpreted data and wrote the manuscript.

Corresponding authors

Correspondence to Ben Z. Stanger, Jacqueline Bromberg or David Lyden.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Morphological and functional characterization of pancreatic ductal adenocarcinoma exosomes.

(a) A representative electron microscope image of exosomes isolated from PAN02 conditioned media. Scale bar, 100 nm. (b) Evaluation of liver metastasis by liver weight (grams) in mice pre-educated with exosomes isolated from a PKCY-mouse model tumor-derived primary cell line (PKCY) or a KPC-mouse model tumor-derived primary cell line (R6560B) before intra-splenic injection with PAN02 cells. Control tumor-bearing mice educated with PBS (TU); n = 4 (TU and PKCYexo + TU) and n = 3 (R6560Bexo + TU) mice from one experiment. P < 0.01,P < 0.05, N.S. stands for not significant by ANOVA. Scale bar, 1 cm. (c) Flow cytometric quantification of the frequency of liver and lung cells incorporating PKH67-labeled exosomes. Exosomes were isolated from normal pancreas (NP), PAN02, PKCY, and R6560B cells; n = 4 (NP) and n = 5 (R6560B) mice from one experiment, and n = 7 (PAN02) and n = 8 (PKCY) mice pooled from two experiments. Statistical source data can be found in Supplementary Table 4. P < 0.01 by ANOVA. (d) Percentage of PKH67-labeled exosome+ liver cells expressing CD11b and F480 markers (KCs). Exosomes were isolated from human (BxPC-3 and HPAF-II) and murine (R6560B) PDAC cell lines; n = 4 (BxPC-3 and HPAF-II) and n = 5 (R6560B) mice from one experiment. (e) Fluorescence microscopy analysis of PKH67-labeled exosomes (green) and αSMA+ cells, S100A4+ fibroblasts, CD31+ endothelial cells, or EpCAM+ epithelial cells (red). Arrows point to green fluorescent signal indicating exosome uptake. Scale bars, 50 μm. (f) Analysis of canonical pathways of genes upregulated by Kupffer cells following in vitro education with BxPC-3 exosomes or PBS treatment. The list is comprised of genes related to liver fibrosis. Data was obtained from one experiment performed in triplicate. Statistical source data can be found in Supplementary Table 2. All data are represented as mean ± s.e.m.

Supplementary Figure 2 Pancreatic ductal adenocarcinoma-derived exosomes induce αSMA and FN expression and increase F4/80+ cell frequency in the liver and migration of BM-derived macrophages to the liver.

(a) Analysis by fluorescence microscopy showing lack of co-localization of FN expression with CD31+ endothelial cells in the livers of mice educated with PAN02 exosomes. Scale bar, 50 μm. (b) Immunofluorescence quantification of αSMA and FN expression in arbitrary units (a.u.) in the livers of mice educated with PBS (CTL), normal pancreas (NP), PKCY, or R6560B exosomes; n = 4 (CTLαSMA; PKCY αSMA; R6560B αSMA and FN) and n = 5 (CTL FN; NP αSMA and FN; PKCY FN) mice from one experiment. Scale bars, 150 μm.P < 0.001; N.S. stands for not significant by ANOVA. (c) Immunofluorescence quantification of the frequency of F4/80+ cell recruited to the livers of mice educated with PBS (CTL), normal pancreas (NP), PKYC, or R6560B exosomes; n = 4 (NP and R6560B) and n = 5 (CTL and PKCY) mice from one experiment. Scale bars, 150 μm.P < 0.01; N.S. stands for not significant by ANOVA. All data are represented as mean ± s.e.m.

Supplementary Figure 3 Low magnification overview of αSMA, FN and F4/80 expression in the liver of mice educated with PBS (CTL), PAN02 exosomes (Exo), Exo in combination with the TGFβR inhibitor A83-01 (Exo + A83-01), Exo in combination with tamoxifen in the FN conditional knockout mouse model (Exo + TMX (CRE+), Exo in combination with Diphtheria toxin (DT) in the CD11b-DTR (Exo + DT(DTR+), and Exo derived from MIF knockdown pancreatic cancer cells (shMIF Exo).

Scale bars, 500 μm.

Supplementary Figure 4 Metastatic pancreatic cells localize adjacent to liver macrophages.

Fluorescence microscopy analysis of F4/80+ staining and mCherry+ PAN02 cell localization in early metastatic lesions of mice educated with PAN02 exosomes reveals close association between metastatic cells and liver macrophages. Scale bar, 150 μm.

Supplementary Figure 5 Evaluation of MIF knockdown exosome uptake by KCs in vivo and the functional consequences on hStC activation and early metastasis.

(a) Western blot analysis of MIF levels in cells and in exosomes (Exo) of non-infected parental PAN02 cells (WT), PAN02 cells infected with control shRNA (shCTL), MIF knockdown (shMIF), a second construct of MIF knockdown (shMIF(2)), and R6560B cells. Unprocessed scans of blots are presented in in Supplementary Fig. 6. (b) Left panel: flow cytometric quantification of PKH67-labeled exosome incorporation by CD11b+F4/80+ liver cells (KCs), represented as a percentage of all PKH67+ cells; n = 4 (WT and shMIF) and n = 5 (shCTL and shMIF(2)) mice from one experiment. Right panel: exosome protein quantification, represented as microgram (μg) per 106 exosomes; n = 3 (shCTL, shMIF, and shMIF(2)) and n = 4 (WT) independent exosome isolations from in vitro cell culture. N.S. stands for not significant by ANOVA. (c) Size distribution analysis of PAN02 exosomes by NanoSight. (d) Immunofluorescence quantification of mCherry+ PAN02 cells 24 hours after their intra-splenic injection into mice previously educated for three weeks with parental PAN02 exosomes (Exo) or PAN02shMIF exosomes (shMIF exo). Arrows In representative images indicate PAN02 cells. Control animals were educated with PBS (TU); n = 4 mice per cohort from one experiment. P < 0.01 by ANOVA. Scale bars, 100 μm. All data are represented as mean ± s.e.m.

Supplementary Figure 6 Unprocessed scans of immunoblots.

Supplementary information

Supplementary Information

Supplementary Information (PDF 2049 kb)

Supplementary Table 1

Supplementary Information (XLSX 242 kb)

Supplementary Table 2

Supplementary Information (XLSX 140 kb)

Supplementary Table 3

Supplementary Information (XLSX 86 kb)

Supplementary Table 4

Supplementary Information (XLSX 44 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Costa-Silva, B., Aiello, N., Ocean, A. et al. Pancreatic cancer exosomes initiate pre-metastatic niche formation in the liver. Nat Cell Biol 17, 816–826 (2015). https://doi.org/10.1038/ncb3169

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ncb3169

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