Skip to main content
Top
Published in: Journal of Translational Medicine 1/2019

Open Access 01-12-2019 | Melanoma | Research

Tumor-derived exosomes promote the in vitro osteotropism of melanoma cells by activating the SDF-1/CXCR4/CXCR7 axis

Authors: Francesco Mannavola, Marco Tucci, Claudia Felici, Anna Passarelli, Stella D’Oronzo, Francesco Silvestris

Published in: Journal of Translational Medicine | Issue 1/2019

Login to get access

Abstract

Background

Bone metastases occur rarely in patients suffering from malignant melanoma, although their onset severely worsens both prognosis and quality of life. Extracellular vesicles (EVs) including exosomes (Exos) are active players in melanoma progression involved in the formation of the pre-metastatic niche.

Methods

Trans-well assays explored the basal migratory and invasive potential of four melanoma cell lines and investigated their different propensity to be attracted toward the bone. Exosomes were purified from cell supernatants by ultracentrifugation and explored in their ability to influence the bone tropism of melanoma cells. The molecular machinery activated during this process was investigated by RT-PCR, droplet digital-PCR, flow-cytometry and Western blot, while loss of function studies with dedicated siRNAs defined the single contribute of CXCR4 and CXCR7 molecules.

Results

Melanoma cells revealed a variable propensity to be attracted toward bone fragments. Gene profiling of both osteotropic and not-osteotropic cells did not show a different expression of those genes notoriously correlated to chemotaxis and bone metastasis. However, bone conditioned medium significantly increased CXCR4, CXCR7 and PTHrP expression solely to osteotropic cells, while their Exos were able to revert the original poor bone tropism of not-osteotropic cells through CXCR7 up-regulation. Silencing experiments also demonstrated that membrane expression of CXCR7 is required by melanoma cells to promote their chemotaxis toward SDF-1 gradients.

Conclusions

Our data correlated the osteotropism of melanoma cells to the activation of the SDF-1/CXCR4/CXCR7 axis following the exposition of tumor cells to bone-derived soluble factors. Also, we demonstrated in vitro that tumor-derived Exos can reprogram the innate osteotropism of melanoma cells by up-regulating membrane CXCR7. These results may have a potential translation to future identification of druggable targets for the treatment of skeletal metastases from malignant melanoma.
Appendix
Available only for authorised users
Literature
1.
go back to reference Selby HM, Sherman RS, Pack GT. A roentgen study of bone metastases from melanoma. Radiology. 1956;67:224–8.CrossRef Selby HM, Sherman RS, Pack GT. A roentgen study of bone metastases from melanoma. Radiology. 1956;67:224–8.CrossRef
2.
go back to reference Zekri J, Marples M, Taylor D, Kandukurti K, McParland L, Brown JE. Complications of bone metastases from malignant melanoma. J Bone Oncol. 2017;8:13–7.CrossRef Zekri J, Marples M, Taylor D, Kandukurti K, McParland L, Brown JE. Complications of bone metastases from malignant melanoma. J Bone Oncol. 2017;8:13–7.CrossRef
3.
go back to reference Casimiro S, Ferreira AR, Mansinho A, Alho I, Costa L. Molecular mechanisms of bone metastasis: which targets came from the bench to the bedside? Int J Mol Sci. 2016;17:1415.CrossRef Casimiro S, Ferreira AR, Mansinho A, Alho I, Costa L. Molecular mechanisms of bone metastasis: which targets came from the bench to the bedside? Int J Mol Sci. 2016;17:1415.CrossRef
4.
go back to reference Sceneay J, Smyth MJ, Möller A. The pre-metastatic niche: finding common ground. Cancer Metastasis Rev. 2013;32:449–64.CrossRef Sceneay J, Smyth MJ, Möller A. The pre-metastatic niche: finding common ground. Cancer Metastasis Rev. 2013;32:449–64.CrossRef
5.
go back to reference Tucci M, Ciavarella S, Strippoli S, Brunetti O, Dammacco F, Silvestris F. Immature dendritic cells from patients with multiple myeloma are prone to osteoclast differentiation in vitro. Exp Hematol. 2011;39(773–83):e1. Tucci M, Ciavarella S, Strippoli S, Brunetti O, Dammacco F, Silvestris F. Immature dendritic cells from patients with multiple myeloma are prone to osteoclast differentiation in vitro. Exp Hematol. 2011;39(773–83):e1.
6.
go back to reference Javelaud D, Mohammad KS, McKenna CR, Fournier P, Luciani F, Niewolna M, et al. Stable overexpression of Smad7 in human melanoma cells impairs bone metastasis. Cancer Res. 2007;67:2317–24.CrossRef Javelaud D, Mohammad KS, McKenna CR, Fournier P, Luciani F, Niewolna M, et al. Stable overexpression of Smad7 in human melanoma cells impairs bone metastasis. Cancer Res. 2007;67:2317–24.CrossRef
7.
go back to reference Krzeszinski JY, Wan Y. New therapeutic targets for cancer bone metastasis. Trends Pharmacol Sci. 2015;36:360–73.CrossRef Krzeszinski JY, Wan Y. New therapeutic targets for cancer bone metastasis. Trends Pharmacol Sci. 2015;36:360–73.CrossRef
8.
go back to reference Cafforio P, Savonarola A, Stucci S, De Matteo M, Tucci M, Brunetti AE, et al. PTHrP produced by myeloma plasma cells regulates their survival and pro-osteoclast activity for bone disease progression. J Bone Miner Res. 2013;29:55–66.CrossRef Cafforio P, Savonarola A, Stucci S, De Matteo M, Tucci M, Brunetti AE, et al. PTHrP produced by myeloma plasma cells regulates their survival and pro-osteoclast activity for bone disease progression. J Bone Miner Res. 2013;29:55–66.CrossRef
9.
go back to reference Wang J, Loberg R, Taichman RS. The pivotal role of CXCL12 (SDF-1)/CXCR9 axis in bone metastasis. Cancer Metastasis Rev. 2006;25:573–87.CrossRef Wang J, Loberg R, Taichman RS. The pivotal role of CXCL12 (SDF-1)/CXCR9 axis in bone metastasis. Cancer Metastasis Rev. 2006;25:573–87.CrossRef
10.
go back to reference Raposo G, Stoorvogel W. Extracellular vesicles: exosomes, microvesicles, and friends. J Cell Biol. 2013;200:373–83.CrossRef Raposo G, Stoorvogel W. Extracellular vesicles: exosomes, microvesicles, and friends. J Cell Biol. 2013;200:373–83.CrossRef
11.
go back to reference Tucci M, Stucci S, Passarelli A, Giudice G, Dammacco F, Silvestris F. The immune escape in melanoma: role of the impaired dendritic cell function. Expert Rev Clin Immunol. 2014;10:1395–404.CrossRef Tucci M, Stucci S, Passarelli A, Giudice G, Dammacco F, Silvestris F. The immune escape in melanoma: role of the impaired dendritic cell function. Expert Rev Clin Immunol. 2014;10:1395–404.CrossRef
12.
go back to reference Tucci M, Mannavola F, Passarelli A, Stucci LS, Cives M, Silvestris F. Exosomes in melanoma: a role in tumor progression, metastasis and impaired immune system activity. Oncotarget. 2018;9:20826–37.CrossRef Tucci M, Mannavola F, Passarelli A, Stucci LS, Cives M, Silvestris F. Exosomes in melanoma: a role in tumor progression, metastasis and impaired immune system activity. Oncotarget. 2018;9:20826–37.CrossRef
13.
go back to reference Passarelli A, Mannavola F, Stucci LS, Tucci M, Silvestris F. Immune system and melanoma biology: a balance between immunosurveillance and immune escape. Oncotarget. 2017;8:106132–42.CrossRef Passarelli A, Mannavola F, Stucci LS, Tucci M, Silvestris F. Immune system and melanoma biology: a balance between immunosurveillance and immune escape. Oncotarget. 2017;8:106132–42.CrossRef
14.
go back to reference Valencia K, Luis-Ravelo D, Bovy N, Antón I, Martínez-Canarias S, Zandueta C, et al. miRNA cargo within exosome-like vesicle transfer influences metastatic bone colonization. Mol Oncol. 2014;8:689.CrossRef Valencia K, Luis-Ravelo D, Bovy N, Antón I, Martínez-Canarias S, Zandueta C, et al. miRNA cargo within exosome-like vesicle transfer influences metastatic bone colonization. Mol Oncol. 2014;8:689.CrossRef
15.
go back to reference Théry C, Amigorena S, Raposo G, Clayton A. Isolation and characterization of exosomes from cell culture supernatants and biological fluids. Curr Protoc Cell Biol. 2006;3:3–22. Théry C, Amigorena S, Raposo G, Clayton A. Isolation and characterization of exosomes from cell culture supernatants and biological fluids. Curr Protoc Cell Biol. 2006;3:3–22.
16.
go back to reference Palmirotta R, Lovero D, Cafforio P, Felici C, Mannavola F, Pellè E, et al. Liquid biopsy of cancer: a multimodal diagnostic tool in clinical oncology. Ther Adv Med Oncol. 2018;10:1758835918794630.CrossRef Palmirotta R, Lovero D, Cafforio P, Felici C, Mannavola F, Pellè E, et al. Liquid biopsy of cancer: a multimodal diagnostic tool in clinical oncology. Ther Adv Med Oncol. 2018;10:1758835918794630.CrossRef
17.
go back to reference Tucci M, Passarelli A, Mannavola F, Stucci LS, Ascierto PA, Capone M, et al. Serum exosomes as predictors of clinical response to ipilimumab in metastatic melanoma. Oncoimmunology. 2017;7:e1387706.CrossRef Tucci M, Passarelli A, Mannavola F, Stucci LS, Ascierto PA, Capone M, et al. Serum exosomes as predictors of clinical response to ipilimumab in metastatic melanoma. Oncoimmunology. 2017;7:e1387706.CrossRef
18.
go back to reference Théry C, Witwer KW, Aikawa E, Alcaraz MJ, Anderson JD, Andriantsitohaina R, et al. Minimal information for studies of extracellular vesicles 2018 (MISEV2018): a position statement of the International Society for Extracellular Vesicles and update of the MISEV2014 guidelines. J Extracell Vesicles. 2018;7:1535750.CrossRef Théry C, Witwer KW, Aikawa E, Alcaraz MJ, Anderson JD, Andriantsitohaina R, et al. Minimal information for studies of extracellular vesicles 2018 (MISEV2018): a position statement of the International Society for Extracellular Vesicles and update of the MISEV2014 guidelines. J Extracell Vesicles. 2018;7:1535750.CrossRef
19.
go back to reference Hughes L, Malone C, Chumsri S, Burger AM, McDonnell S. Characterisation of breast cancer cell lines and establishment of a novel isogenic subclone to study migration, invasion and tumourigenicity. Clin Exp Metastasis. 2008;25:549–57.CrossRef Hughes L, Malone C, Chumsri S, Burger AM, McDonnell S. Characterisation of breast cancer cell lines and establishment of a novel isogenic subclone to study migration, invasion and tumourigenicity. Clin Exp Metastasis. 2008;25:549–57.CrossRef
20.
go back to reference Rocuts F, Ma Y, Zhang X, Gao W, Yue Y, Vartanian T, et al. Carbon monoxide suppresses membrane expression of TLR4 via myeloid differentiation factor-2 in betaTC3 cells. J Immunol. 2010;185:2134–9.CrossRef Rocuts F, Ma Y, Zhang X, Gao W, Yue Y, Vartanian T, et al. Carbon monoxide suppresses membrane expression of TLR4 via myeloid differentiation factor-2 in betaTC3 cells. J Immunol. 2010;185:2134–9.CrossRef
21.
go back to reference Cafforio P, Viggiano L, Mannavola F, Pellè E, Caporusso C, Maiorano E, et al. pIL6-TRAIL-engineered umbilical cord mesenchymal/stromal stem cells are highly cytotoxic for myeloma cells both in vitro and in vivo. Stem Cell Res Ther. 2017;8:206.CrossRef Cafforio P, Viggiano L, Mannavola F, Pellè E, Caporusso C, Maiorano E, et al. pIL6-TRAIL-engineered umbilical cord mesenchymal/stromal stem cells are highly cytotoxic for myeloma cells both in vitro and in vivo. Stem Cell Res Ther. 2017;8:206.CrossRef
22.
go back to reference Kawada K, Sonoshita M, Sakashita H, Takabayashi A, Yamaoka Y, Manabe T, et al. Pivotal role of CXCR22 in melanoma cell metastasis to lymph nodes. Can Res. 2004;64:4010–7.CrossRef Kawada K, Sonoshita M, Sakashita H, Takabayashi A, Yamaoka Y, Manabe T, et al. Pivotal role of CXCR22 in melanoma cell metastasis to lymph nodes. Can Res. 2004;64:4010–7.CrossRef
23.
go back to reference Murakami T, Cardones AR, Hwang ST. Chemokine receptors and melanoma metastasis. J Dermatol Sci. 2004;36:71–8.CrossRef Murakami T, Cardones AR, Hwang ST. Chemokine receptors and melanoma metastasis. J Dermatol Sci. 2004;36:71–8.CrossRef
24.
go back to reference Peinado H, Zhang H, Matei IR, Costa-Silva B, Hoshino A, Rodrigues G, et al. Pre-metastatic niches: organ-specific homes for metastases. Nat Rev Cancer. 2017;17:302–17.CrossRef Peinado H, Zhang H, Matei IR, Costa-Silva B, Hoshino A, Rodrigues G, et al. Pre-metastatic niches: organ-specific homes for metastases. Nat Rev Cancer. 2017;17:302–17.CrossRef
25.
go back to reference Maia J, Caja S, Moraes MCS, Couto N, Costa-Silva B. Exosome-based cell-cell communication in the tumor microenvironment. Front Cell Dev Biol. 2018;6:18.CrossRef Maia J, Caja S, Moraes MCS, Couto N, Costa-Silva B. Exosome-based cell-cell communication in the tumor microenvironment. Front Cell Dev Biol. 2018;6:18.CrossRef
26.
go back to reference Stucci S, Tucci M, Passarelli A, Silvestris F. Avβ3 integrin: pathogenetic role in osteotropic tumors. Crit Rev Oncol Hematol. 2015;96:183–93.CrossRef Stucci S, Tucci M, Passarelli A, Silvestris F. Avβ3 integrin: pathogenetic role in osteotropic tumors. Crit Rev Oncol Hematol. 2015;96:183–93.CrossRef
27.
go back to reference Hayashi C, Rittling S, Hayata T, Amagasa T, Denhardt D, Ezura Y, et al. Serum osteopontin, an enhancer of tumor metastasis to bone, promotes B16 melanoma cell migration. J Cell Biochem. 2007;101:979–86.CrossRef Hayashi C, Rittling S, Hayata T, Amagasa T, Denhardt D, Ezura Y, et al. Serum osteopontin, an enhancer of tumor metastasis to bone, promotes B16 melanoma cell migration. J Cell Biochem. 2007;101:979–86.CrossRef
28.
go back to reference Maruta S, Takiguchi S, Ueyama M, Kataoka Y, Oda Y, Tsuneyoshi M, et al. A role for leukemia inhibitory factor in melanoma-induced bone metastasis. Clin Exp Metastasis. 2008;26:133–41.CrossRef Maruta S, Takiguchi S, Ueyama M, Kataoka Y, Oda Y, Tsuneyoshi M, et al. A role for leukemia inhibitory factor in melanoma-induced bone metastasis. Clin Exp Metastasis. 2008;26:133–41.CrossRef
29.
go back to reference Inder KL, Ruelcke JE, Petelin L, Moon H, Choi E, Rae J, et al. Cavin-1/PTRF alters prostate cancer cell-derived extracellular vesicle content and internalization to attenuate extracellular vesicle-mediated osteoclastogenesis and osteoblast proliferation. J Extracell Vesicles. 2014;3:23784.CrossRef Inder KL, Ruelcke JE, Petelin L, Moon H, Choi E, Rae J, et al. Cavin-1/PTRF alters prostate cancer cell-derived extracellular vesicle content and internalization to attenuate extracellular vesicle-mediated osteoclastogenesis and osteoblast proliferation. J Extracell Vesicles. 2014;3:23784.CrossRef
30.
go back to reference Peinado H, Alečković M, Lavotshkin S, Matei I, Costa-Silva B, Moreno-Bueno G, et al. Melanoma exosomes educate bone marrow progenitor cells toward a pro-metastatic phenotype through MET. Nat Med. 2012;18:883.CrossRef Peinado H, Alečković M, Lavotshkin S, Matei I, Costa-Silva B, Moreno-Bueno G, et al. Melanoma exosomes educate bone marrow progenitor cells toward a pro-metastatic phenotype through MET. Nat Med. 2012;18:883.CrossRef
31.
go back to reference Hashimoto K, Ochi H, Sunamura S, Kosaka N, Mabuchi Y, Fukuda T, et al. Cancer-secreted hsa-miR-940 induces an osteoblastic phenotype in the bone metastatic microenvironment via targeting ARHGAP1 and FAM134A. Proc Natl Acad Sci USA. 2018;115:2204–9.CrossRef Hashimoto K, Ochi H, Sunamura S, Kosaka N, Mabuchi Y, Fukuda T, et al. Cancer-secreted hsa-miR-940 induces an osteoblastic phenotype in the bone metastatic microenvironment via targeting ARHGAP1 and FAM134A. Proc Natl Acad Sci USA. 2018;115:2204–9.CrossRef
32.
go back to reference Luis-Ravelo D, Antón I, Zandueta C, Valencia K, Ormazábal C, Martínez-Canarias S, et al. A gene signature of bone metastatic colonization sensitizes for tumor-induced osteolysis and predicts survival in lung cancer. Oncogene. 2014;33(43):5090.CrossRef Luis-Ravelo D, Antón I, Zandueta C, Valencia K, Ormazábal C, Martínez-Canarias S, et al. A gene signature of bone metastatic colonization sensitizes for tumor-induced osteolysis and predicts survival in lung cancer. Oncogene. 2014;33(43):5090.CrossRef
33.
go back to reference Hoshino A, Costa-Silva B, Shen T-L, Rodrigues G, Hashimoto A, Mark MT, et al. Tumour exosome integrins determine organotropic metastasis. Nature. 2015;527:329.CrossRef Hoshino A, Costa-Silva B, Shen T-L, Rodrigues G, Hashimoto A, Mark MT, et al. Tumour exosome integrins determine organotropic metastasis. Nature. 2015;527:329.CrossRef
34.
go back to reference Xiao D, Barry S, Kmetz D, Egger M, Pan J, Rai SN, et al. Melanoma cell-derived exosomes promote epithelial-mesenchymal transition in primary melanocytes through paracrine/autocrine signaling in the tumor microenvironment. Cancer Lett. 2016;376:318–27.CrossRef Xiao D, Barry S, Kmetz D, Egger M, Pan J, Rai SN, et al. Melanoma cell-derived exosomes promote epithelial-mesenchymal transition in primary melanocytes through paracrine/autocrine signaling in the tumor microenvironment. Cancer Lett. 2016;376:318–27.CrossRef
35.
go back to reference Li M, Lu Y, Xu Y, Wang J, Zhang C, Du Y, et al. Horizontal transfer of exosomal CXCR35 promotes murine hepatocarcinoma cell migration, invasion and lymphangiogenesis. Gene. 2018;676:101–9.CrossRef Li M, Lu Y, Xu Y, Wang J, Zhang C, Du Y, et al. Horizontal transfer of exosomal CXCR35 promotes murine hepatocarcinoma cell migration, invasion and lymphangiogenesis. Gene. 2018;676:101–9.CrossRef
36.
go back to reference Alonso SR, Tracey L, Ortiz P, Pérez-Gómez B, Palacios J, Pollán M, et al. A high-throughput study in melanoma identifies epithelial-mesenchymal transition as a major determinant of metastasis. Can Res. 2007;67:3450–60.CrossRef Alonso SR, Tracey L, Ortiz P, Pérez-Gómez B, Palacios J, Pollán M, et al. A high-throughput study in melanoma identifies epithelial-mesenchymal transition as a major determinant of metastasis. Can Res. 2007;67:3450–60.CrossRef
37.
go back to reference Schüler Y, Lee-Thedieck C, Geiger K, Kaiser T, Ino Y, Aicher WK, et al. Osteoblast-secreted factors enhance the expression of dysadherin and CCL2-dependent migration of renal carcinoma cells. Int J Cancer. 2011;130:288–99.CrossRef Schüler Y, Lee-Thedieck C, Geiger K, Kaiser T, Ino Y, Aicher WK, et al. Osteoblast-secreted factors enhance the expression of dysadherin and CCL2-dependent migration of renal carcinoma cells. Int J Cancer. 2011;130:288–99.CrossRef
38.
go back to reference Guo F, Wang Y, Liu J, Mok SC, Xue F, Zhang W. CXCL12/CXCR1: a symbiotic bridge linking cancer cells and their stromal neighbors in oncogenic communication networks. Oncogene. 2016;35(7):816.CrossRef Guo F, Wang Y, Liu J, Mok SC, Xue F, Zhang W. CXCL12/CXCR1: a symbiotic bridge linking cancer cells and their stromal neighbors in oncogenic communication networks. Oncogene. 2016;35(7):816.CrossRef
39.
go back to reference Shi J, Wei Y, Xia J, Wang S, Wu J, Chen F, et al. CXCL12-CXCR39 contributes to the implication of bone marrow in cancer metastasis. Future Oncol. 2014;10:749–59.CrossRef Shi J, Wei Y, Xia J, Wang S, Wu J, Chen F, et al. CXCL12-CXCR39 contributes to the implication of bone marrow in cancer metastasis. Future Oncol. 2014;10:749–59.CrossRef
40.
go back to reference Zabel BA, Wang Y, Lewén S, Berahovich RD, Penfold MET, Zhang P, et al. Elucidation of CXCR40-mediated signaling events and inhibition of CXCR40-mediated tumor cell transendothelial migration by CXCR40 ligands. J Immunol. 2009;183:3204–11.CrossRef Zabel BA, Wang Y, Lewén S, Berahovich RD, Penfold MET, Zhang P, et al. Elucidation of CXCR40-mediated signaling events and inhibition of CXCR40-mediated tumor cell transendothelial migration by CXCR40 ligands. J Immunol. 2009;183:3204–11.CrossRef
41.
go back to reference Rajagopal S, Kim J, Ahn S, Craig S, Lam CM, Gerard NP, et al. Beta-arrestin- but not G protein-mediated signaling by the “decoy” receptor CXCR41. Proc Natl Acad Sci USA. 2009;107:628–32.CrossRef Rajagopal S, Kim J, Ahn S, Craig S, Lam CM, Gerard NP, et al. Beta-arrestin- but not G protein-mediated signaling by the “decoy” receptor CXCR41. Proc Natl Acad Sci USA. 2009;107:628–32.CrossRef
42.
go back to reference Liedtke D, Erhard I, Abe K, Furutani-Seiki M, Kondoh H, Schartl M. Xmrk-induced melanoma progression is affected by Sdf1 signals through Cxcr7. Pigment Cell Melanoma Res. 2013;27:221–33.CrossRef Liedtke D, Erhard I, Abe K, Furutani-Seiki M, Kondoh H, Schartl M. Xmrk-induced melanoma progression is affected by Sdf1 signals through Cxcr7. Pigment Cell Melanoma Res. 2013;27:221–33.CrossRef
43.
go back to reference Boldajipour B, Mahabaleshwar H, Kardash E, Reichman-Fried M, Blaser H, Minina S, et al. Control of chemokine-guided cell migration by ligand sequestration. Cell. 2008;132:463–73.CrossRef Boldajipour B, Mahabaleshwar H, Kardash E, Reichman-Fried M, Blaser H, Minina S, et al. Control of chemokine-guided cell migration by ligand sequestration. Cell. 2008;132:463–73.CrossRef
44.
go back to reference Luker KE, Lewin SA, Mihalko LA, Schmidt BT, Winkler JS, Coggins NL, et al. Scavenging of CXCL12 by CXCR44 promotes tumor growth and metastasis of CXCR44-positive breast cancer cells. Oncogene. 2012;31:4750–8.CrossRef Luker KE, Lewin SA, Mihalko LA, Schmidt BT, Winkler JS, Coggins NL, et al. Scavenging of CXCL12 by CXCR44 promotes tumor growth and metastasis of CXCR44-positive breast cancer cells. Oncogene. 2012;31:4750–8.CrossRef
45.
go back to reference Naumann U, Cameroni E, Pruenster M, Mahabaleshwar H, Raz E, Zerwes H-G, et al. CXCR45 functions as a scavenger for CXCL12 and CXCL11. PLoS ONE. 2010;5:e9175.CrossRef Naumann U, Cameroni E, Pruenster M, Mahabaleshwar H, Raz E, Zerwes H-G, et al. CXCR45 functions as a scavenger for CXCL12 and CXCL11. PLoS ONE. 2010;5:e9175.CrossRef
46.
go back to reference Lazar I, Clement E, Ducoux-Petit M, Denat L, Soldan V, Dauvillier S, et al. Proteome characterization of melanoma exosomes reveals a specific signature for metastatic cell lines. Pigment Cell Melanoma Res. 2015;28:464–75.CrossRef Lazar I, Clement E, Ducoux-Petit M, Denat L, Soldan V, Dauvillier S, et al. Proteome characterization of melanoma exosomes reveals a specific signature for metastatic cell lines. Pigment Cell Melanoma Res. 2015;28:464–75.CrossRef
47.
go back to reference Mannavola F, Tucci M, Felici C, Stucci S, Silvestris F. miRNAs in melanoma: a defined role in tumor progression and metastasis. Expert Rev Clin Immunol. 2015;12:79–89.CrossRef Mannavola F, Tucci M, Felici C, Stucci S, Silvestris F. miRNAs in melanoma: a defined role in tumor progression and metastasis. Expert Rev Clin Immunol. 2015;12:79–89.CrossRef
Metadata
Title
Tumor-derived exosomes promote the in vitro osteotropism of melanoma cells by activating the SDF-1/CXCR4/CXCR7 axis
Authors
Francesco Mannavola
Marco Tucci
Claudia Felici
Anna Passarelli
Stella D’Oronzo
Francesco Silvestris
Publication date
01-12-2019
Publisher
BioMed Central
Published in
Journal of Translational Medicine / Issue 1/2019
Electronic ISSN: 1479-5876
DOI
https://doi.org/10.1186/s12967-019-1982-4

Other articles of this Issue 1/2019

Journal of Translational Medicine 1/2019 Go to the issue