Phagocytic clearance of targeted cells with a synthetic ligand

Wait 5 sec.

AbstractDuring the process of engulfment, phosphatidylserine is exposed on the surface of dead cells as an ‘eat-me’ signal and is recognized by Protein S (ProS), a secreted factor that also binds to the Mer tyrosine kinase (MerTK) on phagocytes. Despite its robust activity, this engulfment mechanism has not been exploited for therapeutic purposes. Here we develop a synthetic protein modality called Crunch (connector for removal of unwanted cell habitat) by modifying ProS, inspired by the high engulfment capability of the ProS–MerTK pathway. In Crunch, the phosphatidylserine-binding motif of ProS is replaced with a nanobody or single-chain variable fragment that recognizes the surface proteins of targeted cells. Green fluorescent protein nanobody-conjugated Crunch eliminates green fluorescent protein-expressing melanoma cells in transplantation mouse models. In addition, CD19+B cells are eliminated by anti-CD19 single-chain variable fragment-conjugated Crunch, resulting in a therapeutic effect on systemic lupus erythematosus. Both mouse and human versions of Crunch are effective, establishing this synthetic ligand as a promising tool for the elimination of targeted cells.This is a preview of subscription content, access via your institutionAccess optionsAccess Nature and 54 other Nature Portfolio journalsGet Nature+, our best-value online-access subscription27,99 € / 30 dayscancel any timeLearn moreSubscribe to this journalReceive 12 digital issues and online access to articles118,99 € per yearonly 9,92 € per issueLearn moreBuy this articlePurchase on SpringerLinkInstant access to full article PDFBuy nowPrices may be subject to local taxes which are calculated during checkoutFig. 1: Design for Crunch.Fig. 2: MerTK dimerization and phosphorylation by Crunch.Fig. 3: Engulfment by Crunch in vitro and in vivo.Fig. 4: Suppression of tumour growth and metastasis by Crunch.Fig. 5: Removal of targeted through recognition of an endogenous marker by scFv Crunch.Fig. 6: Removal of targeted cells by scFv Crunch targets for disease treatment.Data availabilityHuman acral melanoma datasets for single-cell analysis were obtained from the GEO database under accession number GSE115978. Source data are provided with this paper.ReferencesSoussi, T. & Wiman, K. G. Shaping genetic alterations in human cancer: the p53 mutation paradigm. Cancer Cell 12, 303–312 (2007).CAS  PubMed  Google Scholar Rahman, A. & Isenberg, D. A. Systemic lupus erythematosus. N. Engl. J. Med. 358, 929–939 (2008).CAS  PubMed  Google Scholar Smolen, J. S. et al. Rheumatoid arthritis. Nat. Rev. Dis. Prim. 4, 1–23 (2018).Google Scholar Huston, J. S. et al. Protein engineering of antibody binding sites: recovery of specific activity in an anti-digoxin single-chain Fv analogue produced in Escherichia coli. Proc. Natl Acad. Sci. USA 85, 5879–5883 (1988).CAS  PubMed  PubMed Central  Google Scholar Brudno, J. N. & Kochenderfer, J. N. Current understanding and management of CAR T cell-associated toxicities. Nat. Rev. Clin. Oncol. 21, 501–521 (2024).PubMed  PubMed Central  Google Scholar Baker, D. J., Arany, Z., Baur, J. A., Epstein, J. A. & June, C. H. CAR T therapy beyond cancer: the evolution of a living drug. Nature 619, 707–715 (2023).CAS  PubMed  Google Scholar Leidi, M. et al. M2 macrophages phagocytose rituximab-opsonized leukemic targets more efficiently than M1 cells in vitro. J. Immunol. 182, 4415–4422 (2009).CAS  PubMed  Google Scholar Shi, Y. et al. Trastuzumab triggers phagocytic killing of high HER2 cancer cells in vitro and in vivo by interaction with Fcγ receptors on macrophages. J. Immunol. 194, 4379–4386 (2015).CAS  PubMed  Google Scholar Iwai, Y. et al. Involvement of PD-L1 on tumor cells in the escape from host immune system and tumor immunotherapy by PD-L1 blockade. Proc. Natl Acad. Sci. USA 99, 12293–12297 (2002).CAS  PubMed  PubMed Central  Google Scholar Bader, J. E. et al. Obesity induces PD-1 on macrophages to suppress anti-tumour immunity. Nature 630, 968–975 (2024).CAS  PubMed  PubMed Central  Google Scholar Ishida, Y., Agata, Y., Shibahara, K. & Honjo, T. Induced expression of PD-1, a novel member of the immunoglobulin gene superfamily, upon programmed cell death. EMBO J. 11, 3887–3895 (1992).CAS  PubMed  PubMed Central  Google Scholar Löffler, A. et al. A recombinant bispecific single-chain antibody, CD19 × CD3, induces rapid and high lymphoma-directed cytotoxicity by unstimulated T lymphocytes. Blood 95, 2098–2103 (2000).PubMed  Google Scholar Dreier, T. et al. Extremely potent, rapid and costimulation-independent cytotoxic T-cell response against lymphoma cells catalyzed by a single-chain bispecific antibody. Int. J. Cancer 100, 690–697 (2002).CAS  PubMed  Google Scholar Hosseini, S. S. et al. Bispecific monoclonal antibodies for targeted immunotherapy of solid tumors: recent advances and clinical trials. Int. J. Biol. Macromol. 167, 1030–1047 (2021).CAS  PubMed  Google Scholar Kris, M. G. et al. Using multiplexed assays of oncogenic drivers in lung cancers to select targeted drugs. JAMA 311, 1998–2006 (2014).PubMed  PubMed Central  Google Scholar Thress, K. S. et al. Acquired EGFR C797S mutation mediates resistance to AZD9291 in non–small cell lung cancer harboring EGFR T790M. Nat. Med. 21, 560–562 (2015).CAS  PubMed  PubMed Central  Google Scholar Arandjelovic, S. & Ravichandran, K. S. Phagocytosis of apoptotic cells in homeostasis. Nat. Immunol. 16, 907–917 (2015).CAS  PubMed  PubMed Central  Google Scholar Nagata, S. & Tanaka, M. Programmed cell death and the immune system. Nat. Rev. Immunol. 17, 333–340 (2017).CAS  PubMed  Google Scholar Suzuki, J., Denning, D. P., Imanishi, E., Horvitz, H. R. & Nagata, S. Xk-related protein 8 and CED-8 promote phosphatidylserine exposure in apoptotic cells. Science 341, 403–406 (2013).CAS  PubMed  Google Scholar Nyberg, P., He, X., Härdig, Y., Dahlbäck, B. & García De Frutos, P. Stimulation of Sky tyrosine phosphorylation by bovine Protein S. Eur. J. Biochem. 246, 147–154 (1997).CAS  PubMed  Google Scholar Lai, C. & Lemke, G. An extended family of protein-tyrosine kinase genes differentially expressed in the vertebrate nervous system. Neuron 6, 691–704 (1991).CAS  PubMed  Google Scholar Graham, D. K., Dawson, T. L., Mullaney, D. L., Snodgrass, H. R. & Earp, H. S. Cloning and mRNA expression analysis of a novel human protooncogene, c-mer. Cell Growth Differ. 5, 647–657 (1994).CAS  PubMed  Google Scholar Prasad, D. et al. TAM receptor function in the retinal pigment epithelium. Mol. Cell Neurosci. 33, 96–108 (2006).CAS  PubMed  Google Scholar Saerens, D. et al. Identification of a universal VHH framework to graft non-canonical antigen-binding loops of camel single-domain antibodies. J. Mol. Biol. 352, 597–607 (2005).CAS  PubMed  Google Scholar Kubala, M. H., Kovtun, O., Alexandrov, K. & Collins, B. M. Structural and thermodynamic analysis of the GFP:GFP-nanobody complex. Protein Sci. 19, 2389–2401 (2010).CAS  PubMed  PubMed Central  Google Scholar Suzuki, J., Umeda, M., Sims, P. J. & Nagata, S. Calcium-dependent phospholipid scrambling by TMEM16F. Nature 468, 834–838 (2010).CAS  PubMed  Google Scholar Maruoka, M. et al. Caspase cleavage releases a nuclear protein fragment that stimulates phospholipid scrambling at the plasma membrane. Mol. Cell 81, 1397–1410.e9 (2021).CAS  PubMed  Google Scholar Attallah, C., Etcheverrigaray, M., Kratje, R. & Oggero, M. A highly efficient modified human serum albumin signal peptide to secrete proteins in cells derived from different mammalian species. Protein Expr. Purif. 132, 27–33 (2017).CAS  PubMed  Google Scholar Massagué, J. Epidermal growth factor-like transforming growth factor. II. Interaction with epidermal growth factor receptors in human placenta membranes and A431 cells. J. Biol. Chem. 258, 13614–13620 (1983).PubMed  Google Scholar Revest, J. M., DeMoerlooze, L. & Dickson, C. Fibroblast growth factor 9 secretion is mediated by a non-cleaved amino-terminal signal sequence. J. Biol. Chem. 275, 8083–8090 (2000).CAS  PubMed  Google Scholar Sasada, R., Marumoto, R. & Igarashi, K. Secretion of human EGF and IgE in mammalian cells by recombinant DNA techniques; use of a IL-2 leader sequence. Cell Struct. Funct. 13, 129–141 (1988).CAS  PubMed  Google Scholar Kamiyama, D. et al. Versatile protein tagging in cells with split fluorescent protein. Nat. Commun. 7, 11046 (2016).CAS  PubMed  PubMed Central  Google Scholar Ling, L., Templeton, D. & Kung, H.-J. Identification of the major autophosphorylation sites of Nyk/Mer, an NCAM-related receptor tyrosine kinase*. J. Biol. Chem. 271, 18355–18362 (1996).CAS  PubMed  Google Scholar Nishi, C., Yanagihashi, Y., Segawa, K. & Nagata, S. MERTK tyrosine kinase receptor together with TIM4 phosphatidylserine receptor mediates distinct signal transduction pathways for efferocytosis and cell proliferation. J. Biol. Chem. 294, 7221–7230 (2019).CAS  PubMed  PubMed Central  Google Scholar Carbone, F. R., Sterry, S. J., Butler, J., Rodda, S. & Moore, M. W. T cell receptor alpha-chain pairing determines the specificity of residue 262 within the Kb-restricted, ovalbumin257-264 determinant. Int Immunol. 4, 861–867 (1992).CAS  PubMed  Google Scholar Price, P. A., Fraser, J. D. & Metz-Virca, G. Molecular cloning of matrix Gla protein: implications for substrate recognition by the vitamin K-dependent gamma-carboxylase. Proc. Natl Acad. Sci. USA 84, 8335–8339 (1987).CAS  PubMed  PubMed Central  Google Scholar van der Meer, J. H. M., van der Poll, T. & van ‘t Veer, C. TAM receptors, Gas6, and Protein S: roles in inflammation and hemostasis. Blood 123, 2460–2469 (2014).PubMed  Google Scholar Reynisson, B., Alvarez, B., Paul, S., Peters, B. & Nielsen, M. NetMHCpan-4.1 and NetMHCIIpan-4.0: improved predictions of MHC antigen presentation by concurrent motif deconvolution and integration of MS MHC eluted ligand data. Nucleic Acids Res. 48, W449–W454 (2020).CAS  PubMed  PubMed Central  Google Scholar Jespersen, M. C., Peters, B., Nielsen, M. & Marcatili, P. BepiPred-2.0: improving sequence-based B-cell epitope prediction using conformational epitopes. Nucleic Acids Res. 45, W24–W29 (2017).CAS  PubMed  PubMed Central  Google Scholar Naoi, Y. et al. CD106 in tumor-specific exhausted CD8+ T cells mediates immunosuppression by inhibiting TCR signaling. Cancer Res. 84, 2109–2122 (2024).CAS  PubMed  Google Scholar Beck, J. D. et al. Long-lasting mRNA-encoded interleukin-2 restores CD8+ T cell neoantigen immunity in MHC class I-deficient cancers. Cancer Cell 42, 568–582.e11 (2024).CAS  PubMed  Google Scholar Jerby-Arnon, L. et al. A cancer cell program promotes T cell exclusion and resistance to checkpoint blockade. Cell 175, 984–997.e24 (2018).CAS  PubMed  PubMed Central  Google Scholar Kochenderfer, J. N., Yu, Z., Frasheri, D., Restifo, N. P. & Rosenberg, S. A. Adoptive transfer of syngeneic T cells transduced with a chimeric antigen receptor that recognizes murine CD19 can eradicate lymphoma and normal B cells. Blood 116, 3875–3886 (2010).CAS  PubMed  PubMed Central  Google Scholar Mitchell, K. G. et al. High-volume hybridoma sequencing on the NeuroMabSeq platform enables efficient generation of recombinant monoclonal antibodies and scFvs for neuroscience research. Sci. Rep. 13, 16200 (2023).CAS  PubMed  PubMed Central  Google Scholar Schrand, B. et al. Hapten-mediated recruitment of polyclonal antibodies to tumors engenders antitumor immunity. Nat. Commun. 9, 3348 (2018).PubMed  PubMed Central  Google Scholar Carmi, Y. et al. Allogeneic IgG combined with dendritic cell stimuli induce antitumour T-cell immunity. Nature 521, 99–104 (2015).CAS  PubMed  PubMed Central  Google Scholar Patel, D. et al. Generation and characterization of a therapeutic human antibody to melanoma antigen TYRP1. Hum. Antibodies 16, 127–136 (2007).CAS  PubMed  Google Scholar Xiong, W. & Lahita, R. G. Pragmatic approaches to therapy for systemic lupus erythematosus. Nat. Rev. Rheumatol. 10, 97–107 (2014).CAS  PubMed  Google Scholar Mackensen, A. et al. Anti-CD19 CAR T cell therapy for refractory systemic lupus erythematosus. Nat. Med. 28, 2124–2132 (2022).CAS  PubMed  Google Scholar Doglio, M. et al. Regulatory T cells expressing CD19-targeted chimeric antigen receptor restore homeostasis in systemic lupus erythematosus. Nat. Commun. 15, 2542 (2024).CAS  PubMed  PubMed Central  Google Scholar Edman, N. I. et al. Modulation of FGF pathway signaling and vascular differentiation using designed oligomeric assemblies. Cell 187, 3726–3740.e43 (2024).CAS  PubMed  PubMed Central  Google Scholar Krishna, R. et al. Generalized biomolecular modeling and design with RoseTTAFold All-Atom. Science https://doi.org/10.1126/science.adl2528 (2024).PubMed  PubMed Central  Google Scholar Roy, A. et al. De novo design of highly selective miniprotein inhibitors of integrins αvβ6 and αvβ8. Nat. Commun. 14, 5660 (2023).CAS  PubMed  PubMed Central  Google Scholar Kunert, R. & Reinhart, D. Advances in recombinant antibody manufacturing. Appl. Microbiol. Biotechnol. 100, 3451–3461 (2016).CAS  PubMed  PubMed Central  Google Scholar Tihanyi, B. & Nyitray, L. Recent advances in CHO cell line development for recombinant protein production. Drug Discov. Today Technol. 38, 25–34 (2020).PubMed  Google Scholar Xu, W.-J., Lin, Y., Mi, C.-L., Pang, J.-Y. & Wang, T.-Y. Progress in fed-batch culture for recombinant protein production in CHO cells. Appl. Microbiol. Biotechnol. 107, 1063–1075 (2023).CAS  PubMed  PubMed Central  Google Scholar Thim, L. et al. Secretion and processing of insulin precursors in yeast. Proc. Natl Acad. Sci. USA 83, 6766–6770 (1986).CAS  PubMed  PubMed Central  Google Scholar Mukherjee, J. et al. Prolonged prophylactic protection from botulism with a single adenovirus treatment promoting serum expression of a VHH-based antitoxin protein. PLoS ONE 9, e106422 (2014).PubMed  PubMed Central  Google Scholar Mandrup, O. A. et al. Programmable half-life and anti-tumour effects of bispecific T-cell engager-albumin fusions with tuned FcRn affinity. Commun. Biol. 4, 1–11 (2021).Google Scholar Pyzik, M. et al. The neonatal Fc receptor (FcRn): a misnomer? Front Immunol. 10, 1540 (2019).CAS  PubMed  PubMed Central  Google Scholar Borgel, D. et al. Implication of protein S thrombin-sensitive region with membrane binding via conformational changes in the gamma-carboxyglutamic acid-rich domain. Biochem. J. 360, 499–506 (2001).CAS  PubMed  PubMed Central  Google Scholar Dahlbäck, B. & Villoutreix, B. O. Regulation of blood coagulation by the Protein C anticoagulant pathway. Arter. Thromb. Vasc. Biol. 25, 1311–1320 (2005).Google Scholar Griffin, J. H., Zlokovic, B. V. & Mosnier, L. O. Protein C anticoagulant and cytoprotective pathways. Int J. Hematol. 95, 333–345 (2012).CAS  PubMed  PubMed Central  Google Scholar de Córdoba, S. R. et al. The gene coding for the β-chain of C4b-binding protein (C4BPB) has become a pseudogene in the mouse. Genomics 21, 501–509 (1994).Google Scholar Hillarp, A., Thern, A. & Dahlbäck, B. Bovine C4b binding protein. Molecular cloning of the alpha- and beta-chains provides structural background for lack of complex formation with protein S. J. Immunol. 153, 4190–4199 (1994).CAS  PubMed  Google Scholar DeRyckere, D., Huelse, J. M., Earp, H. S. & Graham, D. K. TAM family kinases as therapeutic targets at the interface of cancer and immunity. Nat. Rev. Clin. Oncol. 20, 755–779 (2023).PubMed  Google Scholar Genetic dissection of TAM receptor–ligand interaction in retinal pigment epithelial cell phagocytosis. Neuron 76, 1123–1132 (2012).Sandahl, M., Hunter, D. B., Strunk, K. E., Earp, H. S. & Cook, R. S. Epithelial cell-directed efferocytosis in the post-partum mammary gland is necessary for tissue homeostasis and future lactation. BMC Dev. Biol. 10, 122 (2010).CAS  PubMed  PubMed Central  Google Scholar Lu, Q., Gore, M. & Zhang, Q. et al. Tyro-3 family receptors are essential regulators of mammalian spermatogenesis. Nature 398, 723–728 (1999).CAS  PubMed  Google Scholar Wu, J. et al. Disturbed flow impairs MerTK-mediated efferocytosis in aortic endothelial cells during atherosclerosis. Theranostics 14, 2427–2441 (2024).CAS  PubMed  PubMed Central  Google Scholar Albelda, S. M. CAR T cell therapy for patients with solid tumours: key lessons to learn and unlearn. Nat. Rev. Clin. Oncol. 21, 47–66 (2024).PubMed  Google Scholar Jung, H. et al. Anti-inflammatory clearance of amyloid-β by a chimeric Gas6 fusion protein. Nat. Med. 28, 1802–1812 (2022).CAS  PubMed  Google Scholar Morioka, S. et al. Chimeric efferocytic receptors improve apoptotic cell clearance and alleviate inflammation. Cell 185, 4887–4903.e17 (2022).CAS  PubMed  PubMed Central  Google Scholar Liu, X. et al. CD47 blockade triggers T cell–mediated destruction of immunogenic tumors. Nat. Med. 21, 1209–1215 (2015).CAS  PubMed  PubMed Central  Google Scholar Vonderheide, R. H. CD47 blockade as another immune checkpoint therapy for cancer. Nat. Med. 21, 1122–1123 (2015).CAS  PubMed  Google Scholar Niesen, F. H., Berglund, H. & Vedadi, M. The use of differential scanning fluorimetry to detect ligand interactions that promote protein stability. Nat. Protoc. 2, 2212–2221 (2007).CAS  PubMed  Google Scholar Foss, S. et al. Human IgG Fc-engineering for enhanced plasma half-life, mucosal distribution and killing of cancer cells and bacteria. Nat. Commun. 15, 2007 (2024).CAS  PubMed  PubMed Central  Google Scholar Tarhan, L. et al. Single Cell Portal: an interactive home for single-cell genomics data. Preprint at bioRxiv https://doi.org/10.1101/2023.07.13.548886 (2023).Muraoka, M. et al. IK cytokine ameliorates the progression of lupus nephritis in MRL/lpr mice. Arthritis Rheumatism 54, 3591–3600 (2006).CAS  PubMed  Google Scholar Download referencesAcknowledgementsWe thank A. Fujimoto for administrative assistance and A. Wee for proof-reading. This work was supported by JST-CREST (grant no. 1199566), Kusunoki125, Sumitomo Kyoto University Innovation Promotion System (SKIPS) to J. Suzuki. Computation time was provided by the Supercomputer System, Institute for Chemical Research, Kyoto University.Author informationAuthors and AffiliationsInstitute for Integrated Cell-Material Sciences, Kyoto University, Yoshida-Honmachi, Sakyoku, Kyoto, JapanYuki Yamato & Jun SuzukiGraduate School of Biostudies, Kyoto University, Konoe-cho, Yoshida, Sakyoku, Kyoto, JapanYuki Yamato & Jun SuzukiCenter for Integrated Biosystems, Institute of Biomedical Sciences, Academia Sinica, Taipei, TaiwanJun SuzukiCREST, Japan Science and Technology Agency, Kawaguchi, Saitama, JapanJun SuzukiAuthorsYuki YamatoView author publicationsSearch author on:PubMed Google ScholarJun SuzukiView author publicationsSearch author on:PubMed Google ScholarContributionsY.Y. and J.S. designed the overall research and interpreted experimental results. Y.Y. conducted all experiments. Y.Y. and J.S. wrote the paper.Corresponding authorCorrespondence to Jun Suzuki.Ethics declarationsCompeting interestsJ.S. and Y.Y. are inventors on a patent application (2024-191258) of a protein modality to eliminate unwanted cells. This work was supported in part by the Collaborative Research Grant from Sumitomo Pharma Co Ltd.Peer reviewPeer review informationNature Biomedical Engineering thanks Jeanette Leusen, Lun Tsou and Thomas Valerius and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Peer reviewer reports are available.Additional informationPublisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.Extended dataExtended Data Fig. 1 Production and characterization of Crunch.a–b, Generation of high Crunch-secreting CHO cells. CHO cells transduced with Crunch-IRES-RFP lentivirus were RFP-sorted twice (S2), reinfected, and sorted again to yield S3. Cells were seeded, switched to serum-free medium after 16 hr, and supernatant was collected at 48 hr. Crunch expression was analyzed by by immunoblotting with anti-FLAG antibody (a), and binding to GFPm⁺ BDKO cells was assessed by flow cytometry (b). c, Production kinetics. S3 cells were cultured as above, and daily supernatants were analyzed by ELISA (n = 3, independent biological samples). d–e, Crunch purification. Supernatant (540 ml, Day 6) was filtered, pH-adjusted (25 mM HEPES, pH 7.5), and incubated with Ni-NTA beads overnight. Input, flow-through, wash, and elution were assessed by immunoblotting with anti-FLAG (d). Eluates were buffer-exchanged to PBS and analyzed with BSA standards by CBB staining (e). Final concentration: 3.7 mg/ml; yield: ~9.61 mg/L. f, Glycosylation analysis. Crunch was treated with PNGaseF and analyzed by by immunoblotting with anti-FLAG. g, Binding affinity. ELISA using GFP-coated plates compared GFPNb-Crunch and GFP nanobody. Absorbance (450 nm) was used to calculate KD via nonlinear regression (R). h–i, Thermal stability. DSF analysis of 1D3 antibody and Crunch (mean of triplicates shown in h); aggregation onset temperatures (Tm) shown in i (n = 3, triplicate using the same sample). j, Vitamin K effect. GFPNb-Crunch produced with or without vitamin K was PNGaseF-treated and analyzed by by immunoblotting with anti-FLAG. All graphs show mean ± S.D.Source dataExtended Data Fig. 2 MerTK dimerization and phosphorylation.a–b, Split-GFP assay for MerTK-sfGFP. NIH3T3 cells expressing MerTK-sfGFP and tagRFP were co-cultured with BDKO or GFPm⁺ BDKO cells in conditioned medium containing mock or GFPNb-Crunch (from HEK293T) at 37 °C for 2 hr. After removing target cells, RFP⁺ MerTK-sfGFP⁺ NIH3T3 cells were analyzed by flow cytometry. Dot plots show % GFP⁺ cells among RFP⁺ NIH3T3 cells (a); bar plot shows % GFP⁺ cells among MerTK-sfGFP⁺ NIH3T3 cells with BDKO (b) (n = 4, independent biological samples). GFPm⁺ BDKO data in Fig. 2e. Mean ± S.D.; Student’s t test; N.S.P > 0.05 c, MerTK phosphorylation by apoptotic thymocytes and ProS. Mouse thymocytes were treated with PBS (–) or FasL (+) for 3 hr at 37 °C, incubated with 10% FBS medium for ProS loading, washed, and co-cultured with NIH3T3 or MerTK⁺ NIH3T3 cells in serum-free medium. After centrifugation (300 × g, 2 min, 37 °C) and 15 min incubation, NIH3T3 cells were washed, lysed, and analyzed by immunoblotting with anti–phospho-MerTK and anti-MerTK antibodies. d, MerTK phosphorylation by PtdSer-exposed living cells and ProS. Ba/F3 or aXkr4⁺ Ba/F3 cells were incubated with 10% FBS medium, washed, and added to MerTK⁺ NIH3T3 cells. After centrifugation and 15 min incubation, MerTK⁺ cells were lysed and analyzed by immunoblotting as in (c). e, Time-dependent MerTK phosphorylation by GFPNb-Crunch. GFPm⁺ BDKO cells were added to MerTK⁺ NIH3T3 cells with (+) or without (-) 1 µg/ml GFPNb-Crunch in serum-free medium with 5 mg/ml BSA. After centrifugation and incubation (0–30 min, 37 °C), MerTK⁺ cells were lysed and analyzed by immunoblotting as in (c).Source dataExtended Data Fig. 3 Cell survival assay.a, Experimental design. Ba/F3 cells were engineered to express MerTK, aXkr4 (to expose PtdSer), or both. 2.5 × 105 cells were cultured in 500 µl IL-3(–) RPMI1640 with FBS. MerTK activation supports survival and proliferation. b–e, Cell survival by PtdSer and MerTK. Cell number and viability were measured by Trypan blue exclusion every 24 hr. Line graphs show mean cell number (b) and viability (c); bar plots show both parameters at 48 and 72 hr (d, e) (n = 3, independent biological samples). f–i, Cell survival by Crunch. MerTK⁺ Ba/F3 cells were cultured with (+) or without (-) 10 µg/ml GFPNb-Crunch in IL-3(–) medium. Cell number (f) and viability (g) were measured every 24 hr. Bar plots show both at 48 and 72 hr (h, i) (n = 3, independent biological samples). Data are mean ± S.D. Statistical analysis: one-way ANOVA with Tukey-Kramer t-test (d–e, g); Student’s t test (f–i). N.S., P > 0.05; *P