Melanoma exosomes educate bone marrow progenitor cells toward a pro-metastatic phenotype through MET

Tumor-derived exosomes are emerging mediators of tumorigenesis. We explored the function of melanoma-derived exosomes in the formation of primary tumors and metastases in mice and human subjects. Exosomes from highly metastatic melanomas increased the metastatic behavior of primary tumors by permanently 'educating' bone marrow progenitors through the receptor tyrosine kinase MET. Melanoma-derived exosomes also induced vascular leakiness at pre-metastatic sites and reprogrammed bone marrow progenitors toward a pro-vasculogenic phenotype that was positive for c-Kit, the receptor tyrosine kinase Tie2 and Met. Reducing Met expression in exosomes diminished the pro-metastatic behavior of bone marrow cells. Notably, MET expression was elevated in circulating CD45−C-KITlow/+TIE2+ bone marrow progenitors from individuals with metastatic melanoma. RAB1A, RAB5B, RAB7 and RAB27A, regulators of membrane trafficking and exosome formation, were highly expressed in melanoma cells. Rab27A RNA interference decreased exosome production, preventing bone marrow education and reducing, tumor growth and metastasis. In addition, we identified an exosome-specific melanoma signature with prognostic and therapeutic potential comprised of TYRP2, VLA-4, HSP70, an HSP90 isoform and the MET oncoprotein. Our data show that exosome production, transfer and education of bone marrow cells supports tumor growth and metastasis, has prognostic value and offers promise for new therapeutic directions in the metastatic process.

[1]  I. Fidler Critical determinants of melanoma metastasis. , 1996, The journal of investigative dermatology. Symposium proceedings.

[2]  D. Bottaro,et al.  Targeting the c-Met signaling pathway in cancer. , 2008, Clinical cancer research : an official journal of the American Association for Cancer Research.

[3]  Hamid Cheshmi Glioblastoma microvesicles transport RNA and proteins that promote tumour growth and provide diagnostic biomarkers , 2011 .

[4]  S. Pomeroy,et al.  Tumour microvesicles contain retrotransposon elements and amplified oncogene sequences. , 2011, Nature communications.

[5]  S. Wickline,et al.  Exosomes released by melanoma cells prepare sentinel lymph nodes for tumor metastasis. , 2011, Cancer research.

[6]  Z. Shao,et al.  Enhanced Expression of Rab27A Gene by Breast Cancer Cells Promoting Invasiveness and the Metastasis Potential by Secretion of Insulin-Like Growth Factor-II , 2008, Molecular Cancer Research.

[7]  M. Zöller,et al.  CD44v6 dependence of premetastatic niche preparation by exosomes. , 2009, Neoplasia.

[8]  P. Comoglio,et al.  Targeting the MET oncogene in cancer and metastases , 2010, Expert opinion on investigational drugs.

[9]  Miguel C. Seabra,et al.  Rab27a and Rab27b control different steps of the exosome secretion pathway , 2010, Nature Cell Biology.

[10]  W. Grizzle,et al.  Contribution of MyD88 to the tumor exosome-mediated induction of myeloid derived suppressor cells. , 2010, The American journal of pathology.

[11]  Hubing Shi,et al.  Pulmonary vascular destabilization in the premetastatic phase facilitates lung metastasis. , 2009, Cancer research.

[12]  J. Catravas,et al.  Regulators of endothelial and epithelial barrier integrity and function in acute lung injury. , 2009, Biochemical pharmacology.

[13]  R. Kaplan,et al.  Priming the 'soil' for breast cancer metastasis: the pre-metastatic niche. , 2006, Breast disease.

[14]  J Ratajczak,et al.  Membrane-derived microvesicles: important and underappreciated mediators of cell-to-cell communication , 2006, Leukemia.

[15]  Hiroyuki Aburatani,et al.  Tumour-mediated upregulation of chemoattractants and recruitment of myeloid cells predetermines lung metastasis , 2006, Nature Cell Biology.

[16]  James Ewing,et al.  Neoplastic Diseases: A Treatise on Tumours , 1928, The Indian Medical Gazette.

[17]  O. De Wever,et al.  Effect of the secretory small GTPase Rab27B on breast cancer growth, invasion, and metastasis. , 2010, Journal of the National Cancer Institute.

[18]  J Ratajczak,et al.  Embryonic stem cell-derived microvesicles reprogram hematopoietic progenitors: evidence for horizontal transfer of mRNA and protein delivery , 2006, Leukemia.

[19]  J. Lötvall,et al.  Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells , 2007, Nature Cell Biology.

[20]  N. Altorki,et al.  Bone marrow-derived endothelial progenitor cells contribute to the angiogenic switch in tumor growth and metastatic progression. , 2009, Biochimica et biophysica acta.

[21]  H. Stenmark Rab GTPases as coordinators of vesicle traffic , 2009, Nature Reviews Molecular Cell Biology.

[22]  D. Pe’er,et al.  An Integrated Approach to Uncover Drivers of Cancer , 2010, Cell.

[23]  L. Trusolino,et al.  MET signalling: principles and functions in development, organ regeneration and cancer , 2010, Nature Reviews Molecular Cell Biology.

[24]  S. Rafii,et al.  VEGFR1-positive haematopoietic bone marrow progenitors initiate the pre-metastatic niche , 2005, Nature.

[25]  Jacopo Meldolesi,et al.  Shedding microvesicles: artefacts no more. , 2009, Trends in cell biology.

[26]  S Paget,et al.  THE DISTRIBUTION OF SECONDARY GROWTHS IN CANCER OF THE BREAST. , 1889 .

[27]  Yibin Kang,et al.  Unravelling the complexity of metastasis — molecular understanding and targeted therapies , 2011, Nature Reviews Cancer.

[28]  J. Erler,et al.  The pre-metastatic niche: is metastasis random? , 2012, BoneKEy reports.

[29]  K. Preissner,et al.  Cell surface tetraspanin Tspan8 contributes to molecular pathways of exosome-induced endothelial cell activation. , 2010, Cancer research.

[30]  P. Quesenberry,et al.  Alteration of Marrow Cell Gene Expression, Protein Production, and Engraftment into Lung by Lung‐Derived Microvesicles: A Novel Mechanism for Phenotype Modulation , 2007, Stem cells.

[31]  M. Zöller CD44: can a cancer-initiating cell profit from an abundantly expressed molecule? , 2011, Nature Reviews Cancer.

[32]  G. Parmiani,et al.  Tumour-released exosomes and their implications in cancer immunity , 2008, Cell Death and Differentiation.

[33]  D. Bottaro,et al.  Targeting the HGF/Met signalling pathway in cancer. , 2010, European journal of cancer.

[34]  M. Bar‐eli,et al.  Transcriptional control of melanoma metastasis: the importance of the tumor microenvironment. , 2011, Seminars in cancer biology.

[35]  J. Christensen,et al.  c-Met as a target for human cancer and characterization of inhibitors for therapeutic intervention. , 2005, Cancer letters.

[36]  G. Raposo,et al.  Exosomes: a common pathway for a specialized function. , 2006, Journal of biochemistry.

[37]  Hiroyuki Aburatani,et al.  The S100A8–serum amyloid A3–TLR4 paracrine cascade establishes a pre-metastatic phase , 2008, Nature Cell Biology.

[38]  W. Birchmeier,et al.  Met, metastasis, motility and more , 2003, Nature Reviews Molecular Cell Biology.

[39]  A. Poliakov,et al.  Induction of myeloid‐derived suppressor cells by tumor exosomes , 2009, International journal of cancer.

[40]  Bethan Psaila,et al.  The metastatic niche: adapting the foreign soil , 2009, Nature Reviews Cancer.

[41]  Miguel C. Seabra,et al.  1 Rab 27 a and Rab 27 b control different steps of the exosome secretion pathway , 2009 .

[42]  Gerald C. Chu,et al.  Proinvasion metastasis drivers in early-stage melanoma are oncogenes. , 2011, Cancer cell.

[43]  A. Turner,et al.  The HGF/c-Met axis synergizes with G-CSF in the mobilization of hematopoietic stem/progenitor cells. , 2010, Stem cells and development.

[44]  Massimo Spada,et al.  High Levels of Exosomes Expressing CD63 and Caveolin-1 in Plasma of Melanoma Patients , 2009, PloS one.

[45]  J. Pollard,et al.  Microenvironmental regulation of metastasis , 2009, Nature Reviews Cancer.

[46]  Djuro Josic,et al.  Microvesicle entry into marrow cells mediates tissue-specific changes in mRNA by direct delivery of mRNA and induction of transcription. , 2010, Experimental hematology.

[47]  M. Barcinski,et al.  Tumor-derived microvesicles modulate the establishment of metastatic melanoma in a phosphatidylserine-dependent manner. , 2009, Cancer letters.

[48]  J. Erler,et al.  Hypoxia-induced lysyl oxidase is a critical mediator of bone marrow cell recruitment to form the premetastatic niche. , 2009, Cancer cell.

[49]  A. Guha,et al.  Intercellular transfer of the oncogenic receptor EGFRvIII by microvesicles derived from tumour cells , 2008, Nature Cell Biology.

[50]  G. Nicolson,et al.  Organ selectivity for implantation survival and growth of B16 melanoma variant tumor lines. , 1976, Journal of the National Cancer Institute.

[51]  P. Comoglio,et al.  Invasive growth: a MET-driven genetic programme for cancer and stem cells , 2006, Nature Reviews Cancer.

[52]  T. Guise Examining the metastatic niche: targeting the microenvironment. , 2010, Seminars in oncology.

[53]  A. Rosenwald,et al.  Targeting the microenvironment , 2010, Leukemia & lymphoma.

[54]  E. Laurenti,et al.  Enhanced c-Met activity promotes G-CSF-induced mobilization of hematopoietic progenitor cells via ROS signaling. , 2011, Blood.

[55]  Laurence Zitvogel,et al.  Exosomes: composition, biogenesis and function , 2002, Nature Reviews Immunology.

[56]  C. Ramana,et al.  The Role of Hsp90N, a New Member of the Hsp90 Family, in Signal Transduction and Neoplastic Transformation* , 2002, The Journal of Biological Chemistry.

[57]  D. Lyden,et al.  The secreted factors responsible for pre-metastatic niche formation: old sayings and new thoughts. , 2011, Seminars in cancer biology.

[58]  Edi Brogi,et al.  Breast cancer cells produce tenascin C as a metastatic niche component to colonize the lungs , 2011, Nature Medicine.

[59]  Aled Clayton,et al.  Cancer exosomes trigger fibroblast to myofibroblast differentiation. , 2010, Cancer research.

[60]  J. Xiang,et al.  Epigenetic transfer of metastatic activity by uptake of highly metastatic B16 melanoma cell-released exosomes. , 2006, Experimental oncology.

[61]  Douglas D. Taylor,et al.  Characterization of plasma membrane shedding from murine melanoma cells , 1988, International journal of cancer.