Data were fit with GraphPad version 5. Acknowledgments This work is supported by grants from the program ‘Investissements d’Avenir’ with reference ANR-11-LABX-0021-01-LipSTIC Labex, the Conseil Regional de Bourgogne, the INCa (Institut National du Cancer, POLYNOM-174), the Cancrop?le Grand-Est, la Ligue Nationale Contre le Cancer and the ANR (Agence Nationale de la Recherche, 07-PCV-0031 and SphingoDR). has been reported to both inhibit and enhance TRAIL-mediated apoptosis.36, 37, 38 It will be interesting in the future to test whether this might be related to TRAIL receptor glycosylation status. Keeping in mind that neoplastic C75 transformation involves drastic changes in glycosylation,39 galectin-3 expression40 and N-terminal sugar modifications,41 all should be considered as potentially important regulators of the TRAIL-mediated tumor killing. Altogether, our results provide the first evidence that TRAIL-R1 analysis Sequence alignment across species was performed using CLC Sequence Nr4a1 Viewer 6.5.2 software (CLC bio, Aarhus, Denmarkoctet). O– and N-glycosylated sites were predicted using the GlycoEP server (prediction of glycosides in eukaryotic glycoproteins),16 NetNGlyc1.0 and NetOGlyc 3.1 servers available at http://www.imtech.res.in/raghava/glycoep/ and at the CBS (Center for biological sequence analysis (http://www.cbs.dtu.dk/services/NetNGlyc/ or NetOGlyc/), respectivley. Representation of TRAIL-R1 and mTRAIL-R 3D structure prediction were inferred from TRAIL-R2 crystallographic structure using PHYRE2 Protein Fold Recognition server,17 at http://www.sbg.bio.ic.ac.uk/phyre2. Evolutionary history of primate and rodent TRAIL agonist receptors was inferred using the Neighbor-Joining method using the software MEGA 6.06 (Molecular Evolutionary Genetics Analysis). Statistical analysis Statistical analysis was performed using the Student’s t-test. All statistical analyses were performed using Prism version 5.0a software (GraphPad Software, San Diego, CA, USA). *P<0.05 and **P<0.01 were considered significant. Production of soluble TRAIL receptors and BLI biolayer C75 interferometry analysis Murine mTRAIL-R variants N99A, N122A, N150A mutants and human TRAIL-R1 variant fused to human Fc IgG1 were created by routine site-directed mutagenesis from pCR3-TRAIL-R1-hFc or pCR3-mTRAIL-R-hFc vectors using the following sets of primers: TRAIL-R1 forward 5-GGG TGT GGG TTA CAC CGC CGC TTC CAA CAA TTT G-3, reverse 5-CAA ATT GTT GGA AGC GGC GGT GTA ACC CAC ACC C-3 and primer sets for mTRAIL-R described in Plasmid constructions. All constructs were confirmed by sequencing. To produce these soluble recombinants receptors, 6 106 293?T cells were seeded in 10?cm tissue culture dish and cultured in DMEM C75 medium (Lonza) with 10% fetal calf serum for 24?h. 293?T cells were then transfected with pCR3-mTRAIL-R-WT-hFc, pCR3-mTRAIL-R-N99/122A-hFc, pCR3-mTRAIL-R-N99/122/150A-Fc, pCR3-TRAIL-R1-WT-hFc, pCR3-TRAIL-R1-N156A-WT-hFc using calcium phosphate transfection method. After 16?h, cells were washed twice with HBSS, then 10?ml of Opti-MEM (Invitrogen) were added in each 10 cm tissue culture dish. Seventy-two hours latter, cell culture supernatant was collected, cleared by centrifugation and filtered. Production of soluble hFc-fused WT or mutant mTRAIL-R or TRAIL-R1 was assessed by western blot using the anti-mouse TRAIL-R2 antibody from Leinco Technologies and the anti-TRAIL-R1antibody (wB-K32) from Gen-Probe (Diaclone, Besan?on, France). Purification of hFc fusion proteins was achieved by an overnight pull-down with protein A/G-coated beads (Millipore) at 4?C with mixing. Beads were washed four occasions with PBS, and pulled-down proteins was eluted in 100?mM glycine-HCl, pH 2. pH neutralization was achieved by adding 1M Tris, pH 9.0. Quantitation of hFc fusion proteins were decided using an Octet Red System with anti-human IgG quantitation (AHQ) biosensors (FortBIO). All Octet experiments were designed and analyzed with data acquisition software (7.1) and data analysis software (7.1), respectively. Data were fit with GraphPad version 5. Acknowledgments This work is supported by grants from the program 'Investissements d'Avenir' with reference ANR-11-LABX-0021-01-LipSTIC Labex, the Conseil Regional de Bourgogne, the INCa (Institut National du Cancer, POLYNOM-174), the Cancrop?le Grand-Est, la Ligue Nationale Contre le Cancer and the ANR (Agence Nationale de la Recherche, 07-PCV-0031 and SphingoDR). SS, FD, AM and GM were supported by fellowships from the INCa, ANR, the Ministry of Research and Education and the foundation ARC. PS is supported by C75 grants of the Swiss National Science Foundation, DMZ and CAB by the National Institute of Health ("type":"entrez-nucleotide","attrs":"text":"AI117530","term_id":"3517854","term_text":"AI117530"AI117530 and "type":"entrez-nucleotide","attrs":"text":"AI101423","term_id":"3706326","term_text":"AI101423"AI101423, respectively). CG's group has the label 'Ligue contre le Cancer team'. We are indebted to Pr Ali Bettaieb (EPHE, Dijon, France) for EMT6H cells, Pr Serge Lebecque (INSERM U1052, Lyon, France) for U2OS cells, Dr Thierry Guillaudeux (INSERM U917, Rennes, France) and Dr Jean-Ehrland Ricci (INSERM U1065, Nice, France) for B lymphoma cell lines. We thank the FEDER for their support. Author contributions OM and CAB designed research; FD, TR, SS, GP, AAC, AM, ZAB, SC and EH performed experiments; GM, EZ, DMZ, RS, GG, FP, GH, TG, CG, PS, CAB and OM analyzed data; and PS, CAB and OM wrote the paper. Footnotes Supplementary Information accompanies this paper on Cell Death and Differentiation website (http://www.nature.com/cdd) Edited by E Baehrecke The authors declare no conflict.