L. Gebert and I. J. Macrae, Regulation of microRNA function in animals, Nat Rev Mol Cell Biol, vol.20, issue.1, pp.21-37, 2019.

R. C. Friedman, K. K. Farh, C. B. Burge, and D. P. Bartel, Most mammalian mRNAs are conserved targets of microRNAs, Genome Res, vol.19, issue.1, pp.92-105, 2009.

A. Esquela-kerscher and F. J. Slack, Oncomirs -microRNAs with a role in cancer, Nat Rev Cancer, vol.6, issue.4, pp.259-69, 2006.

B. P. Lewis, C. B. Burge, and D. P. Bartel, Conserved seed pairing, often flanked by adenosines, indicates that thousands of human genes are microRNA targets, Cell, vol.120, issue.1, pp.15-20, 2005.

X. Chen, H. Liang, J. Zhang, K. Zen, and C. Zhang, Secreted microRNAs: a new form of intercellular communication, Trends Cell Biol, vol.22, issue.3, pp.125-157, 2012.

C. Villarroya-beltri, F. Baixauli, C. Gutierrez-vazquez, F. Sanchez-madrid, and M. Mittelbrunn, Sorting it out: regulation of exosome loading, Semin Cancer Biol, vol.28, pp.3-13, 2014.

N. Kosaka, H. Iguchi, K. Hagiwara, Y. Yoshioka, F. Takeshita et al., Neutral sphingomyelinase 2 (nSMase2)-dependent exosomal transfer of angiogenic microRNAs regulate cancer cell metastasis, J Biol Chem, vol.288, issue.15, pp.10849-59, 2013.

C. Villarroya-beltri, C. Gutierrez-vazquez, F. Sanchez-cabo, D. Perez-hernandez, J. Vazquez et al., Sumoylated hnRNPA2B1 controls the sorting of miRNAs into exosomes through binding to specific motifs, Nat Commun, vol.4, p.2980, 2013.

F. Hobor, A. Dallmann, N. J. Ball, C. Cicchini, C. Battistelli et al., A cryptic RNAbinding domain mediates Syncrip recognition and exosomal partitioning of miRNA targets, Nat Commun, vol.9, issue.1, p.831, 201826.

D. Koppers-lalic, M. Hackenberg, I. V. Bijnsdorp, M. A. Van-eijndhoven, P. Sadek et al., Nontemplated nucleotide additions distinguish the small RNA composition in cells from exosomes, Cell Rep, vol.8, issue.6, pp.1649-58, 2014.

F. Frank, N. Sonenberg, and B. Nagar, Structural basis for 5'-nucleotide base-specific recognition of guide RNA by human AGO2, Nature, vol.465, issue.7299, pp.818-840, 2010.

D. J. Gibbings, C. Ciaudo, M. Erhardt, and O. Voinnet, Multivesicular bodies associate with components of miRNA effector complexes and modulate miRNA activity, Nat Cell Biol, vol.11, issue.9, pp.1143-1152, 2009.
URL : https://hal.archives-ouvertes.fr/hal-00423288

J. Guduric-fuchs, O. Connor, A. Camp, B. O'neill, C. L. Medina et al., Selective extracellular vesicle-mediated export of an overlapping set of microRNAs from multiple cell types, BMC Genomics, vol.13, p.357, 2012.

M. L. Squadrito, C. Baer, F. Burdet, C. Maderna, G. D. Gilfillan et al., Endogenous RNAs modulate microRNA sorting to exosomes and transfer to acceptor cells, Cell Rep, vol.8, issue.5, pp.1432-1478, 2014.

D. M. Pegtel, K. Cosmopoulos, D. A. Thorley-lawson, M. A. Van-eijndhoven, E. S. Hopmans et al., Functional delivery of viral miRNAs via exosomes, Proc Natl Acad Sci, vol.107, issue.14, pp.6328-6361, 2010.

A. Rodríguez-galán, L. Fernández-messina, and F. Sánchez-madrid, Control of Immunoregulatory Molecules by miRNAs in T Cell Activation, Front Immunol, vol.9, p.2148, 2018.

P. T. Jindra, J. Bagley, J. G. Godwin, and J. Iacomini, Costimulation-dependent expression of microRNA-214 increases the ability of T cells to proliferate by targeting Pten, J Immunol, vol.185, issue.2, pp.990-997, 2010.

N. Zhang and M. J. Bevan, Dicer controls CD8 + T-cell activation, migration, and survival, Proc Natl Acad Sci, vol.107, issue.50, pp.21629-21663, 2010.

C. Gutiérrez-vázquez, A. Rodríguez-galán, M. Fernández-alfara, M. Mittelbrunn, F. Sánchez-cabo et al., miRNA profiling during antigen-dependent T cell activation: A role for miR-132-3p. Sci Rep, vol.7, p.3508, 2017.

M. Aubuchon, L. Bolt, M. Janssen-heijnen, S. Verleisdonk-bolhaar, A. Van-marion et al., Epidemiology, management and survival outcomes of primary cutaneous melanoma: a ten-year overview, Acta Chir Belg, vol.117, issue.1, pp.29-35, 2017.

C. Théry, S. Amigorena, G. Raposo, and A. Clayton, Isolation and characterization of exosomes from cell culture supernatants and biological fluids, Curr Protoc Cell Biol, 2006.

T. L. Bailey, J. Johnson, C. E. Grant, and W. S. Noble, The MEME Suite, Nucleic Acids Res, vol.43, issue.W1, pp.39-49, 2015.

L. G. Lima, R. Chammas, R. Q. Monteiro, M. Moreira, and M. A. Barcinski, Tumor-derived microvesicles modulate the establishment of metastatic melanoma in a phosphatidylserine-dependent manner, Cancer Lett, vol.283, issue.2, pp.168-75, 2009.

H. Zhang and W. E. Grizzle, Exosomes: A Novel Pathway of Local and Distant Intercellular Communication that Facilitates the Growth and Metastasis of Neoplastic Lesions, The American Journal of Pathology, vol.184, issue.1, pp.28-41, 2014.

P. P. Corsetti, L. A. De-almeida, A. Gonçalves, M. Gomes, E. S. Guimarães et al., miR-181a-5p Regulates TNF-? and miR-21a-5p Influences Gualynate-Binding Protein 5 and IL-10 Expression in Macrophages Affecting Host Control of Brucella abortus Infection, Front Immunol, vol.9, p.1331, 2018.

P. Santini, L. Politi, P. D. Vedova, R. Scandurra, and A. Scotto-d'abusco, The inflammatory circuitry of miR-149 as a pathological mechanism in osteoarthritis, Rheumatol Int, vol.34, issue.5, pp.711-717, 2014.

D. Xiao, J. Ohlendorf, Y. Chen, D. D. Taylor, S. N. Rai et al., Identifying mRNA, microRNA and protein profiles of melanoma exosomes, PLoS One, vol.7, issue.10, p.46874, 2012.

M. L. Hermiston, Z. Xu, and A. Weiss, CD45: a critical regulator of signaling thresholds in immune cells, Annu Rev Immunol, vol.21, pp.107-144, 2003.

G. A. Koretzky, J. Picus, M. L. Thomas, and A. Weiss, Tyrosine phosphatase CD45 is essential for coupling T-cell antigen receptor to the phosphatidyl inositol pathway, Nature, vol.346, issue.6279, pp.66-74, 1990.

J. T. Pingel and M. L. Thomas, Evidence that the leukocyte-common antigen is required for antigeninduced T lymphocyte proliferation, Cell, vol.58, issue.6, pp.1055-65, 1989.

R. Bayraktar, K. Van-roosbroeck, and G. A. Calin, Cell-to-cell communication: microRNAs as hormones, Mol Oncol, vol.11, issue.12, pp.1673-86, 2017.

I. S. Trowbridge and M. L. Thomas, CD45: an emerging role as a protein tyrosine phosphatase required for lymphocyte activation and development, Annu Rev Immunol, vol.12, pp.85-116, 1994.

I. Seki, M. Suzuki, N. Miyasaka, and H. Kohsaka, Expression of CD45 isoforms correlates with differential proliferative responses of peripheral CD4+ and CD8 + T cells, Immunol Lett, vol.129, issue.1, pp.39-46, 2010.

T. Mustelin, K. M. Coggeshall, and A. Altman, Rapid activation of the T-cell tyrosine protein kinase pp56lck by the CD45 phosphotyrosine phosphatase, Proc Natl Acad Sci, vol.86, issue.16, pp.6302-6308, 1989.

S. B. Ye, Z. L. Li, D. H. Luo, B. J. Huang, Y. S. Chen et al., Tumor-derived exosomes promote tumor progression and T-cell dysfunction through the regulation of enriched exosomal microRNAs in human nasopharyngeal carcinoma, Oncotarget, vol.5, issue.14, pp.5439-52, 2014.

, Author Manuscript Published OnlineFirst, 2019.

H. W. Hwang, E. A. Wentzel, and J. T. Mendell, A hexanucleotide element directs microRNA nuclear import. Science, vol.315, pp.97-100, 2007.

G. Wang, H. Chen, Y. Oktay, J. Zhang, A. E. Smith et al., PNPASE regulates RNA import into mitochondria, Cell, vol.142, issue.3, pp.456-67, 2010.

Y. Wu, W. Deng, E. C. Mcginley, and D. J. Klinke, Melanoma exosomes deliver a complex biological payload that upregulates PTPN11 to suppress T lymphocyte function, Pigment Cell Melanoma Res, vol.30, issue.2, pp.203-221, 2017.

B. T. Maybruck, L. W. Pfannenstiel, M. Diaz-montero, and B. R. Gastman, Tumor-derived exosomes induce CD8 + T cell suppressors, J Immunother Cancer, vol.15, issue.1, p.65, 2017.

A. J. Abusamra, Z. Zhong, X. Zheng, M. Li, T. E. Ichim et al., Tumor exosomes expressing Fas ligand mediate CD8 + T-cell apoptosis, Blood Cells Mol Dis, vol.35, issue.2, pp.169-73, 2005.

L. Muller, M. Mitsuhashi, P. Simms, W. E. Gooding, and T. L. Whiteside, Wilcoxon tests are applied. B) TNF? RNA expression in CTL03.1 cells exposed to M113 or M117-derived exosomes determined by qPCR after 5 hours of coculture. The 2???Ct method was used to calculate relative changes in expression (n = 3). Results were normalized to level of RPLP0 and PPIA gene expressions. C) TNF? secretion by CTL03.1 cells transfected with 50 ?M of candidate miRNAs (percent reduction relative to the nontransfected CTL03.1 cells). Wilcoxon tests are applied. Results are expressed as mean ± SEM (n = 3). D) Percent of relative luminescence unit (RLU) of HEK-293 cells transfected with candidate miRNAs, vol.6, 2016.

, UTR. A) Schematic representation of constructs wildtype (WT) and mutated in the miR-498 binding site. B) Percent of relative luminescence unit (RLU) of HEK-293 cells transfected with miR-498 (10 ?M) and our different constructs

, Representative flow cytometry plots of CD45 expression in CD8 + T cells. In blue, CD8 + T cells alone, in red, CD8 + T cells exposed to exoM113 and in grey, the isotype control. MFI: Median Fluorescent Intensity (n = 6). B) CD45 expression measured by flow cytometry, on CTL03.1 cells exposed to M113 or M117 exosomes expressed as percent reduction, Figure 5. Melanoma-derived exosomes decrease CD45 expression on CD8 + T cells. A)

. +-t-cells, Wilcoxon tests are applied. C) PTPRC expression in CTL03.1 cells exposed to M113 or M117 exosomes determined by qPCR after 5 hours of coculture. The 2???Ct method was used to calculate relative changes in expression. Results were normalized to level of RPLP0 and PPIA gene expressions (n = 3). D) CD45 expression in CTL03.1 transfected with 50 ?M of miR-3187-3p or miR-498, expressed as percent reduction (percent reduction relative to non-transfected CD8 + T cells). Results are expressed as mean ± SEM (n = 9). E) Relative luminescence unit (RLU) of HEK-293 cells transfected with miR-3187-3p. All assays were conducted in triplicate and were normalized to a control containing only transfection reagents. F) Relative luminescence unit (RLU) of HEK-293 cells, Results are expressed as mean ± SEM (n = 12)

, A) One representative FACS plot of CD45 expression in CTL03.1 transfected with hsa-miR-3187-3p alone or with anti-miR-31873p. In blue, CD8 + T cells alone, in pink, CTL03.1 cells transfected with hsa-miR-3187-3p mimic, in yellow CTL03.1 cells transfected with hsa-antimiR-3187-3p, in green CTL03.1 cells transfected with hsa-miR-3187-3p mimic and its antimir, Figure 6. hsa-miR-3187-3p from melanoma-derived exosomes targets PTPRC transcripts

, MFI: median fluorescent intensity. P values were calculated from 3 independent experiments in triplicate. B) One representative FACS plot of CD45 expression in CTL03.1 exposed to melanoma-Research

, American Association for Cancer cancerimmunolres.aacrjournals.org Downloaded from Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited, 2019.

, Author Manuscript Published OnlineFirst, 2019.

, CD8 + T cells exposed to exoM117, in green CTL03.1 cells exposed to exoM117 transfected with hsa-antimiR-3187-3p and in grey, the isotype control. MFI: median fluorescent intensity. P values were calculated from 3 independent experiments in triplicate. C) Cytotoxicity assay against M113 cells by CTL03, blue, CD8 + T cells alone

, Cytotoxicity was measured at different Target (T) / Effector (E) ratios with no exosome (white-filled triangle) and with CTL03.1 pre-incubated overnight with M113, p.117

, American Association for Cancer cancerimmunolres, 2019.