Exosomal tumor microRNA modulates premetastatic organ cells.
Journal: 2013/August - Neoplasia
ISSN: 1476-5586
PUBMED: 23479506
Abstract:
Tumor exosomes educate selected host tissues toward a prometastatic phenotype. We demonstrated this for exosomes of the metastatic rat adenocarcinoma BSp73ASML (ASML), which modulate draining lymph nodes and lung tissue to support settlement of poorly metastatic BSp73ASML-CD44v4-v7 knockdown (ASML-CD44v(kd)) cells. Now, we profiled mRNA and microRNA (miRNA) of ASML(wt) and ASML-CD44v(kd) exosomes to define the pathway(s), whereby exosomes prepare the premetastatic niche. ASML exosomes, recovered in draining lymph nodes after subcutaneous injection, preferentially are taken up by lymph node stroma cells (LnStr) and lung fibroblasts (LuFb) that were chosen as exosome targets. ASML(wt) and ASML-CD44v(kd) exosomes contain a restricted mRNA and miRNA repertoire that differs significantly between the two lines and exosomes thereof due to CD44v6 influencing gene and miRNA transcription/posttranscriptional regulation. Exosomal mRNA and miRNA are recovered in target cells, where transferred miRNA significantly affected mRNA translation. Besides others, this was exemplified for abundant ASML(wt)-exosomal miR-494 and miR-542-3p, which target cadherin-17 (cdh17). Concomitantly, matrix metalloproteinase transcription, accompanying cdh17 down-regulation, was upregulated in LnStr transfected with miR-494 or miR-542-3p or co-cultured with tumor exosomes. Thus, tumor exosomes target non-transformed cells in premetastatic organs and modulate premetastatic organ cells predominantly through transferred miRNA, where miRNA from a metastasizing tumor prepares premetastatic organ stroma cells for tumor cell hosting. Fitting the demands of metastasizing tumor cells, transferred exosomal miRNA mostly affected proteases, adhesion molecules, chemokine ligands, cell cycle- and angiogenesis-promoting genes, and genes engaged in oxidative stress response. The demonstration of function-competent exosomal miRNA in host target cells encourages exploiting exosomes as a therapeutic gene delivery system.
Relations:
Content
Citations
(108)
References
(64)
Diseases
(1)
Conditions
(1)
Chemicals
(1)
Organisms
(3)
Processes
(2)
Anatomy
(4)
Similar articles
Articles by the same authors
Discussion board
Neoplasia 15(3): 281-295

Exosomal Tumor MicroRNA Modulates Premetastatic Organ Cells<sup><a href="#FN1" rid="FN1" class=" fn">1</a>,</sup><sup><a href="#FN2" rid="FN2" class=" fn">2</a></sup>

Supplementary Figures and Tables:
Click here to view.(1.6M, pdf)
Department of Tumor Cell Biology, University Hospital of Surgery, Heidelberg, Germany
German Cancer Research Center, Heidelberg, Germany
Address all correspondence to: Margot Zöller, MD, Department of Tumor Cell Biology, University Hospital of Surgery, Im Neuenheimer Feld 365, D-69120 Heidelberg, Germany. E-mail: ed.grebledieh-inu@relleoz.togram
Address all correspondence to: Margot Zöller, MD, Department of Tumor Cell Biology, University Hospital of Surgery, Im Neuenheimer Feld 365, D-69120 Heidelberg, Germany. E-mail: ed.grebledieh-inu@relleoz.togram
Received 2012 Dec 2; Revised 2013 Jan 11; Accepted 2013 Jan 11.

Abstract

Tumor exosomes educate selected host tissues toward a prometastatic phenotype. We demonstrated this for exosomes of the metastatic rat adenocarcinoma BSp73ASML (ASML), which modulate draining lymph nodes and lung tissue to support settlement of poorly metastatic BSp73ASML-CD44v4-v7 knockdown (ASML-CD44v) cells. Now, we profiled mRNA and microRNA (miRNA) of ASML and ASML-CD44v exosomes to define the pathway(s), whereby exosomes prepare the premetastatic niche. ASML exosomes, recovered in draining lymph nodes after subcutaneous injection, preferentially are taken up by lymph node stroma cells (LnStr) and lung fibroblasts (LuFb) that were chosen as exosome targets. ASML and ASML-CD44v exosomes contain a restricted mRNA and miRNA repertoire that differs significantly between the two lines and exosomes thereof due to CD44v6 influencing gene and miRNA transcription/posttranscriptional regulation. Exosomal mRNA and miRNA are recovered in target cells, where transferred miRNA significantly affected mRNA translation. Besides others, this was exemplified for abundant ASML-exosomal miR-494 and miR-542-3p, which target cadherin-17 (cdh17). Concomitantly, matrix metalloproteinase transcription, accompanying cdh17 down-regulation, was upregulated in LnStr transfected with miR-494 or miR-542-3p or co-cultured with tumor exosomes. Thus, tumor exosomes target non-transformed cells in premetastatic organs and modulate premetastatic organ cells predominantly through transferred miRNA, where miRNA from a metastasizing tumor prepares premetastatic organ stroma cells for tumor cell hosting. Fitting the demands of metastasizing tumor cells, transferred exosomal miRNA mostly affected proteases, adhesion molecules, chemokine ligands, cell cycle- and angiogenesis-promoting genes, and genes engaged in oxidative stress response. The demonstration of function-competent exosomal miRNA in host target cells encourages exploiting exosomes as a therapeutic gene delivery system.

Abstract

Acknowledgments

We thank Shijing Yue and Florian Thuma for help with the luciferase reporter and in vitro mRNA translation assays.

Acknowledgments

Abbreviations

ASMLwtBSp73ASML
ASML-CD44vkdBSp73ASML-CD44v4-v7 knockdown
cdh17cadherin-17
CMconditioned medium
CM-exoexosome-depleted CM
ECsendothelial cells
ifpintrafootpad
KLF4Kruppel-like factor 4
LnStrlymph node stroma cells
LuFblung fibroblasts
MVBsmultivesicular bodies
CTthreshold cycle
Abbreviations

Footnotes

This work was supported by the Deutsche Krebshilfe (M.Z.), the Wilhelm Sander Foundation (M.Z.), and NCT Interdisciplinary Research Program (M.Z.). The authors declare no conflict of interest. Array data are deposited at http://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc={"type":"entrez-geo","attrs":{"text":"GSE34739","term_id":"34739"}}GSE34739.

This article refers to supplementary materials, which are designated by Tables W1 to W6 and Figures W1 to W7 and are available online at www.neoplasia.com.

Footnotes

References

  • 1. Kraljevic Pavelic S, Sedic M, Bosnjak H, Spaventi S, Pavelic KMetastasis: new perspectives on an old problem. Mol Cancer. 2011;10:22.[Google Scholar]
  • 2. Talmadge JE, Fidler IJAACR centennial series: the biology of cancer metastasis: historical perspective. Cancer Res. 2010;70:5649–5669.[Google Scholar]
  • 3. Geiger TR, Peeper DSMetastasis mechanisms. Biochim Biophys Acta. 2006;1796:293–308.[PubMed][Google Scholar]
  • 4. Kaplan RN, Rafii S, Lyden DPreparing the “soil”: the pre-metastatic niche. Cancer Res. 2006;66:11089–11093.[Google Scholar]
  • 5. McGovern M, Voutev R, Maciejowski J, Corsi AK, Hubbard EJA “latent niche” mechanism for tumor initiation. Proc Natl Acad Sci USA. 2009;106:11617–11622.[Google Scholar]
  • 6. Jung T, Castellana D, Klingbeil P, Cuesta Hernández I, Vitacolonna M, Orlicky DJ, Roffler SR, Brodt P, Zöller MCD44v6 dependence of premetastatic niche preparation by exosomes. Neoplasia. 2009;11:1093–1105.[Google Scholar]
  • 7. Chairoungdua A, Smith DL, Pochard P, Hull M, Caplan MJExosome release of β-catenin: a novel mechanism that antagonizes Wnt signaling. J Cell Biol. 2010;190:1079–1091.[Google Scholar]
  • 8. Hood JL, San Roman S, Wickline SAExosomes released by melanoma cells prepare sentinel lymph nodes for tumor metastasis. Cancer Res. 2011;71:3792–3801.[PubMed][Google Scholar]
  • 9. Lee TH, D'Asti E, Magnus N, Al-Nedawi K, Meehan B, Rak JMicrovesicles as mediators of intercellular communication in cancer—the emerging science of cellular ‘debris’ Semin Immunopathol. 2011;33:455–467.[PubMed][Google Scholar]
  • 10. Lima LG, Chammas R, Monteiro RQ, Moreira ME, Barcinski MA, Tumor-derived microvesicles modulate the establishment of metastatic melanoma in a phosphatidylserine-dependent manner. Cancer Lett. 2009;283:168–175.[PubMed]
  • 11. McCready J, Sims JD, Chan D, Jay DGSecretion of extracellular hsp90 via exosomes increases cancer cell motility: a role for plasminogen activation. BMC Cancer. 2010;10:294.[Google Scholar]
  • 12. Park JE, Tan HS, Datta A, Lai RC, Zhang H, Meng W, Lim SK, Sze SKHypoxic tumor cell modulates its microenvironment to enhance angiogenic and metastatic potential by secretion of proteins and exosomes. Mol Cell Proteomics. 2010;9:1085–1099.[Google Scholar]
  • 13. Peinado H, Lavotshkin S, Lyden DThe secreted factors responsible for pre-metastatic niche formation: old sayings and new thoughts. Semin Cancer Biol. 2011;21:139–146.[PubMed][Google Scholar]
  • 14. Rak JMicroparticles in cancer. Semin Thromb Hemost. 2010;36:888–906.[PubMed][Google Scholar]
  • 15. Ronquist KG, Ronquist G, Larsson A, Carlsson LProteomic analysis of prostate cancer metastasis-derived prostasomes. Anticancer Res. 2010;30:285–290.[PubMed][Google Scholar]
  • 16. Peinado H, Alečković M, Lavotshkin S, Matei I, Costa-Silva B, Moreno-Bueno G, Hergueta-Redondo M, Williams C, García-Santos G, Ghajar C, et al Melanoma exosomes educate bone marrow progenitor cells toward a pro-metastatic phenotype through MET. Nat Med. 2012;18:883–891.[Google Scholar]
  • 17. Mitchell PJ, Welton J, Staffurth J, Court J, Mason MD, Tabi Z, Clayton ACan urinary exosomes act as treatment response markers in prostate cancer? J Transl Med. 2009;7:4.[Google Scholar]
  • 18. Wittmann J, Jäck HMSerum microRNAs as powerful cancer biomarkers. Biochim Biophys Acta. 2010;1806:200–207.[PubMed][Google Scholar]
  • 19. Pap E, Pállinger E, Pásztói M, Falus AHighlights of a new type of intercellular communication:microvesicle-based information transfer. Inflamm Res. 2009;58:1–8.[PubMed][Google Scholar]
  • 20. Denzer K, Kleijmeer MJ, Heijnen HF, Stoorvogel W, Geuze HJExosome: from internal vesicle of the multivesicular body to intercellular signaling device. J Cell Sci. 2000;113(pt 19):3365–3374.[PubMed][Google Scholar]
  • 21. Lakkaraju A, Rodriguez-Boulan EItinerant exosomes: emerging roles in cell and tissue polarity. Trends Cell Biol. 2008;18:199–209.[Google Scholar]
  • 22. Mathivanan S, Ji H, Simpson RJExosomes: extracellular organelles important in intercellular communication. J Proteomics. 2010;73:1907–1920.[PubMed][Google Scholar]
  • 23. van Niel G, Porto-Carreiro I, Simoes S, Raposo GExosomes: a common pathway for a specialized function. J Biochem. 2006;140:13–21.[PubMed][Google Scholar]
  • 24. Raimondo F, Morosi L, Chinello C, Magni F, Pitto MAdvances in membranous vesicle and exosome proteomics improving biological understanding and biomarker discovery. Proteomics. 2010;11:709–720.[PubMed][Google Scholar]
  • 25. Zöller MTetraspanins: push and pull in suppressing and promoting metastasis. Nat Rev Cancer. 2009;9:40–55.[PubMed][Google Scholar]
  • 26. Valadi H, Ekström K, Bossios A, Sjöstrand M, Lee JJ, Lötvall JOExosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat Cell Biol. 2007;9:654–659.[PubMed][Google Scholar]
  • 27. Pant S, Hilton H, Burczynski METhe multifaceted exosome: biogenesis, role in normal and aberrant cellular function, and frontiers for pharmacological and biomarker opportunities. Biochem Pharmacol. 2012;83:1484–1494.[PubMed][Google Scholar]
  • 28. Gibbings DJ, Ciaudo C, Erhardt M, Voinnet OMultivesicular bodies associate with components of miRNA effector complexes and modulate miRNA activity. Nat Cell Biol. 2009;11:1143–1149.[PubMed][Google Scholar]
  • 29. Zomer A, Vendrig T, Hopmans ES, van Eijndhoven M, Middeldorp JM, Pegtel DMExosomes: fit to deliver small RNA. Commun Integr Biol. 2010;3:447–450.[Google Scholar]
  • 30. Atay S, Gercel-Taylor C, Taylor DDHuman trophoblast-derived exosomal fibronectin induces pro-inflammatory Il-1β production by macrophages. Am J Reprod Immunol. 2011;66:259–269.[PubMed][Google Scholar]
  • 31. Escrevente C, Keller S, Altevogt P, Costa JInteraction and uptake of exosomes by ovarian cancer cells. BMC Cancer. 2011;11:108.[Google Scholar]
  • 32. Rana S, Claas C, Kretz CC, Nazarenko I, Zöller MActivation-induced internalization differs for the tetraspanins CD9 and Tspan8: impact on tumor cell motility. Int J Biochem Cell Biol. 2011;43:106–119.[PubMed][Google Scholar]
  • 33. Rana S, Shijing Y, Stadel D, Zöller MToward tailored exosomes: the exosomal tetraspanin web contributes to target cell selection. Int J Biochem Cell Biol. 2012;44:1574–1584.[PubMed][Google Scholar]
  • 34. Kosaka N, Iguchi H, Yoshioka Y, Takeshita F, Matsuki Y, Ochiya TSecretory mechanisms and intercellular transfer of microRNAs in living cells. J Biol Chem. 2010;285:17442–17452.[Google Scholar]
  • 35. Lykke-Andersen S, Brodersen DE, Jensen THOrigins and activities of the eukaryotic exosome. J Cell Sci. 2009;122:1487–1494.[PubMed][Google Scholar]
  • 36. Hwang I, Ki DReceptor-mediated T cell absorption of antigen presenting cell-derived molecules. Front Biosci. 2011;16:411–421.[Google Scholar]
  • 37. Taylor DD, Gercel-Taylor CExosomes/microvesicles: mediators of cancer-associated immunosuppressive microenvironments. Semin Immunopathol. 2011;33:441–454.[PubMed][Google Scholar]
  • 38. Whiteside TL, Mandapathil M, Szczepanski M, Szajnik MMechanisms of tumor escape from the immune system: adenosine-producing Treg, exosomes and tumor-associated TLRs. Bull Cancer. 2011;98:25–31.[PubMed][Google Scholar]
  • 39. Matzku S, Komitowski D, Mildenberger M, Zöller MCharacterization of BSp73, a spontaneous rat tumor and its in vivo selected variants showing different metastasizing capacities. Invasion Metastasis. 1983;3:109–123.[PubMed][Google Scholar]
  • 40. Matzku S, Wenzel A, Liu S, Zöller MAntigenic differences between metastatic and nonmetastatic BSp73 rat tumor variants characterized by monoclonal antibodies. Cancer Res. 1989;49:1294–1299.[PubMed][Google Scholar]
  • 41. Günthert U, Hofmann M, Rudy W, Reber S, Zöller M, Haussmann I, Matzku S, Wenzel A, Ponta H, Herrlich PA new variant of glycoprotein CD44 confers metastatic potential to rat carcinoma cells. Cell. 1991;65:13–24.[PubMed][Google Scholar]
  • 42. Zöller MCD44: can a cancer-initiating cell profit from an abundantly expressed molecule? Nat Rev Cancer. 2011;11:254–267.[PubMed][Google Scholar]
  • 43. Klingbeil P, Marhaba R, Jung T, Kirmse R, Ludwig T, Zöller MCD44 variant isoforms promote metastasis formation by a tumor cell-matrix cross-talk that supports adhesion and apoptosis resistance. Mol Cancer Res. 2009;7:168–179.[PubMed][Google Scholar]
  • 44. LeBedis C, Chen K, Fallavollita L, Boutros T, Brodt PPeripheral lymph node stromal cells can promote growth and tumorigenicity of breast carcinoma cells through the release of IGF-I and EGF. Int J Cancer. 2002;100:2–8.[PubMed][Google Scholar]
  • 45. Zech D, Rana S, Büchler MW, Zöller MTumor-exosomes and leukocyte activation: an ambivalent crosstalk. Cell Commun Signal. 2012;10:37.[Google Scholar]
  • 46. Eldh M, Lötvall J, Malmhäll C, Ekström KImportance of RNA isolation methods for analysis of exosomal RNA: evaluation of different methods. Mol Immunol. 2012;50:278–286.[PubMed][Google Scholar]
  • 47. Liu QS, Zhang J, Liu M, Dong WGLentiviral-mediated miRNA against liver-intestine cadherin suppresses tumor growth and invasiveness of human gastric cancer. Cancer Sci. 2010;101:1807–1812.[PubMed][Google Scholar]
  • 48. Bussolati B, Grange C, Camussi GTumor exploits alternative strategies to achieve vascularization. FASEB J. 2011;25:2874–2882.[PubMed][Google Scholar]
  • 49. Bobrie A, Krumeich S, Reyal F, Recchi C, Moita LF, Seabra MC, Ostrowski M, Théry CRab27a supports exosome-dependent and -independent mechanisms that modify the tumor microenvironment and can promote tumor progression. Cancer Res. 2012;72:4920–4930.[PubMed][Google Scholar]
  • 50. Ohshima K, Inoue K, Fujiwara A, Hatakeyama K, Kanto K, Watanabe Y, Muramatsu K, Fukuda Y, Ogura S, Yamaguchi K, et al Let-7 microRNA family is selectively secreted into the extracellular environment via exosomes in a metastatic gastric cancer cell line. PLoS One. 2010;5:e13247.[Google Scholar]
  • 51. Liu C, Kelnar K, Liu B, Chen X, Calhoun-Davis T, Li H, Patrawala L, Yan H, Jeter C, Honorio S, et al The microRNA miR-34a inhibits prostate cancer stem cells and metastasis by directly repressing CD44. Nat Med. 2011;17:211–215.[Google Scholar]
  • 52. Ali S, Almhanna K, Chen W, Philip PA, Sarkar FHDifferentially expressed miRNAs in the plasma may provide a molecular signature for aggressive pancreatic cancer. Am J Transl Res. 2010;3:28–47.[Google Scholar]
  • 53. Liu L, Jiang Y, Zhang H, Greenlee AR, Han ZOverexpressed miR-494 down-regulates PTEN gene expression in cells transformed by antibenzo(a)pyrene-trans-7,8-dihydrodiol-9,10-epoxide. Life Sci. 2010;86:192–198.[PubMed][Google Scholar]
  • 54. Qin W, Shi Y, Zhao B, Yao C, Jin L, Ma J, Jin YmiR-24 regulates apoptosis by targeting the open reading frame (ORF) region of FAF1 in cancer cells. PLoS One. 2010;5:e9429.[Google Scholar]
  • 55. Garofalo M, Romano G, Di Leva G, Nuovo G, Jeon YJ, Ngankeu A, Sun J, Lovat F, Alder H, Condorelli G, et al EGFR and MET receptor tyrosine kinase-altered microRNA expression induces tumorigenesis and gefitinib resistance in lung cancers. Nat Med. 2011;18:74–82.[Google Scholar]
  • 56. Rana S, Zöller MExosome target cell selection and the importance of exosomal tetraspanins: a hypothesis. Biochem Soc Trans. 2011;39:559–562.[PubMed][Google Scholar]
  • 57. Simons K, Gerl MJRevitalizing membrane rafts: new tools and insights. Nat Rev Mol Cell Biol. 2010;11:688–699.[PubMed][Google Scholar]
  • 58. Nazarenko I, Rana S, Baumann A, McAlear J, Hellwig A, Trendelenburg M, Lochnit G, Preissner KT, Zöller MCell surface tetraspanin Tspan8 contributes to molecular pathways of exosome-induced endothelial cell activation. Cancer Res. 2010;70:1668–1678.[PubMed][Google Scholar]
  • 59. Brandt DT, Xu J, Steinbeisser H, Grosse RRegulation of myocardin-related transcriptional coactivators through cofactor interactions in differentiation and cancer. Cell Cycle. 2009;8:2523–2527.[PubMed][Google Scholar]
  • 60. Frank MMAL, a proteolipid in glycosphingolipid enriched domains: functional implications in myelin and beyond. Prog Neurobiol. 2000;60:531–544.[PubMed][Google Scholar]
  • 61. Lee NP, Poon RT, Shek FH, Ng IO, Luk JMRole of cadherin-17 in oncogenesis and potential therapeutic implications in hepatocellular carcinoma. Biochim Biophys Acta. 2010;1806:138–145.[PubMed][Google Scholar]
  • 62. Mathew SJ, Haubert D, Kronke M, Leptin MLooking beyond death: a morphogenetic role for the TNF signalling pathway. J Cell Sci. 2009;122:1939–1946.[PubMed][Google Scholar]
  • 63. Lin CC, Liu LZ, Addison JB, Wonderlin WF, Ivanov AV, Ruppert JMA KLF4-miRNA-206 autoregulatory feedback loop can promote or inhibit protein translation depending upon cell context. Mol Cell Biol. 2011;31:2513–2527.[Google Scholar]
  • 64. Papp B, Plath KReprogramming to pluripotency: stepwise resetting of the epigenetic landscape. Cell Res. 2011;21:486–501.[Google Scholar]
Collaboration tool especially designed for Life Science professionals.Drag-and-drop any entity to your messages.