Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Unique structural configuration of EV-DNA primes Kupffer cell-mediated antitumor immunity to prevent metastatic progression

Abstract

Extracellular vesicles (EVs) transport biomolecules that mediate intercellular communication. We previously showed that EVs contain DNA (EV-DNA) representing the entire genome. However, the mechanism of genomic EV-DNA packaging and its role in cancer remain elusive. We now demonstrate that EV-DNA is predominantly localized on the vesicle surface and associated with uniquely modified and cleaved histones. Moreover, a genome-wide clustered regularly interspaced short palindromic repeats knockout screen revealed that immune developmental pathways and genes, including apoptotic peptidase activating factor 1 (APAF1) and neutrophil cytosolic factor 1 (NCF1), regulate EV-DNA packaging. Furthermore, in colorectal cancer models, uptake of EV-DNA by pre-metastatic liver Kupffer cells (KCs) activated DNA damage responses. This activation rewired KC cytokine production and promoted the formation of tertiary lymphoid structures, thereby suppressing liver metastasis. Conversely, loss of APAF1 decreased EV-DNA packaging and promoted liver metastasis. Importantly, colorectal cancer biopsy EV-DNA secretion could serve as a predictive biomarker for postoperative metastasis. Taken together, our findings indicate that uniquely chromatinized EV-DNA induces antitumor immunity.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: EV-DNA is uniquely chromatinized and presented on the vesicle surface.
Fig. 2: Genome-wide CRISPR–Cas9 KO screen identifies immune development genes as regulators of EV-DNA packaging.
Fig. 3: APAF1 regulates EV-DNA packaging.
Fig. 4: Loss of EV-DNA secretion promotes liver metastasis in CRC and PDAC models.
Fig. 5: Education with EV-DNA is sufficient to attenuate metastasis in a CRC model.
Fig. 6: KCs internalize CRC-derived EVs irrespective of their DNA content.
Fig. 7: EV-DNA induces DDR in KCs and TLS formation in the liver.
Fig. 8: EV-DNA as a potential prognostic biomarker for predicting metastasis in persons with CRC.

Similar content being viewed by others

Data availability

Raw sequencing data can be found on the Sequence Read Archive (PRJNA1072813), including whole-genome sequencing, DNA sequencing for the CRISPR screen and RNA sequencing. The proteomics data were deposited to the ProteomeXchange Consortium through the PRIDE partner repository under dataset identifiers PXD056104 (PTM MS proteomics data of histones from cells and EVs) and PXD056026 (MS proteomics data for HCT116 and MDA-MB-231 EVs). Uncropped western blot images and raw data for all main and extended data figures have been provided in the Source Data files. All other data supporting the findings of this study are available from the corresponding authors on reasonable request. Source data are provided with this paper.

Code availability

No custom code was generated in the course of this study.

References

  1. Zijlstra, A. & Di Vizio, D. Size matters in nanoscale communication. Nat. Cell Biol. 20, 228–230 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Zhang, H. et al. Identification of distinct nanoparticles and subsets of extracellular vesicles by asymmetric flow field-flow fractionation. Nat. Cell Biol. 20, 332–343 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Zhang, Q. et al. Transfer of functional cargo in exomeres. Cell Rep 27, 940–954 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Jeppesen, D. K. et al. Reassessment of exosome composition. Cell 177, 428–445 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Lucotti, S., Kenific, C. M., Zhang, H. & Lyden, D. Extracellular vesicles and particles impact the systemic landscape of cancer. EMBO J. 41, e109288 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Costa-Silva, B. et al. Pancreatic cancer exosomes initiate pre-metastatic niche formation in the liver. Nat. Cell Biol. 17, 816–826 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Rodrigues, G. et al. Tumour exosomal CEMIP protein promotes cancer cell colonization in brain metastasis. Nat. Cell Biol. 21, 1403–1412 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Peinado, H. et al. Melanoma exosomes educate bone marrow progenitor cells toward a pro-metastatic phenotype through MET. Nat. Med. 18, 883–891 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Hoshino, A. et al. Tumour exosome integrins determine organotropic metastasis. Nature 527, 329–335 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Abdouh, M. et al. Exosomes isolated from cancer patients’ sera transfer malignant traits and confer the same phenotype of primary tumors to oncosuppressor-mutated cells. J. Exp. Clin. Cancer Res. 36, 113 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  11. Clancy, J. W., Sheehan, C. S., Boomgarden, A. C. & D’Souza-Schorey, C. Recruitment of DNA to tumor-derived microvesicles. Cell Rep. 38, 110443 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Kahlert, C. et al. Identification of double-stranded genomic DNA spanning all chromosomes with mutated KRAS and P53 DNA in the serum exosomes of patients with pancreatic cancer. J. Biol. Chem. 289, 3869–3875 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Thakur, B. K. et al. Double-stranded DNA in exosomes: a novel biomarker in cancer detection. Cell Res. 24, 766–769 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Fischer, S. et al. Indication of horizontal DNA gene transfer by extracellular vesicles. PLoS ONE 11, e0163665 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  15. Lazaro-Ibanez, E. et al. DNA analysis of low- and high-density fractions defines heterogeneous subpopulations of small extracellular vesicles based on their DNA cargo and topology. J. Extracell. Vesicles 8, 1656993 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Maire, C. L. et al. Genome-wide methylation profiling of glioblastoma cell-derived extracellular vesicle DNA allows tumor classification. Neuro Oncol. 23, 1087–1099 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Nemeth, A. et al. Antibiotic-induced release of small extracellular vesicles (exosomes) with surface-associated DNA. Sci. Rep. 7, 8202 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  18. Choi, D. et al. Mapping subpopulations of cancer cell-derived extracellular vesicles and particles by nano-flow cytometry. ACS Nano 13, 10499–10511 (2019).

    Article  CAS  PubMed  Google Scholar 

  19. Fernando, M. R., Jiang, C., Krzyzanowski, G. D. & Ryan, W. L. New evidence that a large proportion of human blood plasma cell-free DNA is localized in exosomes. PLoS ONE 12, e0183915 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  20. Lazaro-Ibanez, E. et al. Different gDNA content in the subpopulations of prostate cancer extracellular vesicles: apoptotic bodies, microvesicles, and exosomes. Prostate 74, 1379–1390 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Torralba, D. et al. Priming of dendritic cells by DNA-containing extracellular vesicles from activated T cells through antigen-driven contacts. Nat. Commun. 9, 2658 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  22. Vagner, T. et al. Large extracellular vesicles carry most of the tumour DNA circulating in prostate cancer patient plasma. J. Extracell. Vesicles 7, 1505403 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Bernard, V. et al. Circulating nucleic acids are associated with outcomes of patients with pancreatic cancer. Gastroenterology 156, 108–118 (2019).

    Article  CAS  PubMed  Google Scholar 

  24. Yang, S. et al. Detection of mutant KRAS and TP53 DNA in circulating exosomes from healthy individuals and patients with pancreatic cancer. Cancer Biol. Ther. 18, 158–165 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Choi, J. et al. Detection of circulating KRAS mutant DNA in extracellular vesicles using droplet digital PCR in patients with colon cancer. Front. Oncol. 12, 1067210 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Cai, J. et al. SRY gene transferred by extracellular vesicles accelerates atherosclerosis by promotion of leucocyte adherence to endothelial cells. Clin. Sci. (Lond.) 129, 259–269 (2015).

    Article  CAS  PubMed  Google Scholar 

  27. Sansone, P. et al. Packaging and transfer of mitochondrial DNA via exosomes regulate escape from dormancy in hormonal therapy-resistant breast cancer. Proc. Natl Acad. Sci. USA 114, E9066–E9075 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Ghanam, J. et al. Extracellular vesicles transfer chromatin-like structures that induce non-mutational dysfunction of p53 in bone marrow stem cells. Cell Discov. 9, 12 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Lian, Q. et al. Chemotherapy-induced intestinal inflammatory responses are mediated by exosome secretion of double-strand DNA via AIM2 inflammasome activation. Cell Res. 27, 784–800 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Kitai, Y. et al. DNA-containing exosomes derived from cancer cells treated with topotecan activate a STING-dependent pathway and reinforce antitumor immunity. J. Immunol. 198, 1649–1659 (2017).

    Article  CAS  PubMed  Google Scholar 

  31. Diamond, J. M. et al. Exosomes shuttle TREX1-sensitive IFN-stimulatory dsDNA from irradiated cancer cells to DCs. Cancer Immunol. Res. 6, 910–920 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Sisquella, X. et al. Malaria parasite DNA-harbouring vesicles activate cytosolic immune sensors. Nat. Commun. 8, 1985 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  33. Tsilioni, I. & Theoharides, T. C. Extracellular vesicles are increased in the serum of children with autism spectrum disorder, contain mitochondrial DNA, and stimulate human microglia to secrete IL-1β. J. Neuroinflammation 15, 239 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  34. Takahashi, A. et al. Exosomes maintain cellular homeostasis by excreting harmful DNA from cells. Nat. Commun. 8, 15287 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Yokoi, A. et al. Mechanisms of nuclear content loading to exosomes. Sci. Adv. 5, eaax8849 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Simpson, R. T. Structure of chromatin containing extensively acetylated H3 and H4. Cell 13, 691–699 (1978).

    Article  CAS  PubMed  Google Scholar 

  37. Santos-Rosa, H. et al. Histone H3 tail clipping regulates gene expression. Nat. Struct. Mol. Biol. 16, 17–22 (2009).

    Article  CAS  PubMed  Google Scholar 

  38. Yi, S. J. & Kim, K. Histone tail cleavage as a novel epigenetic regulatory mechanism for gene expression. BMB Rep. 51, 211–218 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Zhou, P., Wu, E., Alam, H. B. & Li, Y. Histone cleavage as a mechanism for epigenetic regulation: current insights and perspectives. Curr. Mol. Med. 14, 1164–1172 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Ahn, B. K., Kim, S. H., Paik, S. S. & Lee, K. H. Loss of APAF-1 expression is associated with early recurrence in stage I, II, and III colorectal cancer. Langenbecks Arch. Surg. 401, 1203–1210 (2016).

    Article  PubMed  Google Scholar 

  41. Pelissier Vatter, F. A. et al. Extracellular vesicle- and particle-mediated communication shapes innate and adaptive immune responses. J. Exp. Med. 218, e20202579 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  42. Li, J. et al. Tumor cell-intrinsic factors underlie heterogeneity of immune cell infiltration and response to immunotherapy. Immunity 49, 178–193 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Wang, G. et al. Tumour extracellular vesicles and particles induce liver metabolic dysfunction. Nature 618, 374–382 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Kasapi, A. & Triantafyllopoulou, A. Genotoxic stress signalling as a driver of macrophage diversity. Cell Stress 6, 30–44 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Braumüller, H. et al. The cytokine network in colorectal cancer: implications for new treatment strategies. Cells 12, 138 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  46. Chang, H. W. et al. Mechanism of FACT removal from transcribed genes by anticancer drugs curaxins. Sci. Adv. 4, eaav2131 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Petitprez, F. et al. B cells are associated with survival and immunotherapy response in sarcoma. Nature 577, 556–560 (2020).

    Article  CAS  PubMed  Google Scholar 

  48. Helmink, B. A. et al. B cells and tertiary lymphoid structures promote immunotherapy response. Nature 577, 549–555 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Cabrita, R. et al. Tertiary lymphoid structures improve immunotherapy and survival in melanoma. Nature 577, 561–565 (2020).

    Article  CAS  PubMed  Google Scholar 

  50. Biller, L. H. & Schrag, D. Diagnosis and treatment of metastatic colorectal cancer: a review. JAMA 325, 669–685 (2021).

    Article  CAS  PubMed  Google Scholar 

  51. Hugen, N., van de Velde, C. J. H., de Wilt, J. H. W. & Nagtegaal, I. D. Metastatic pattern in colorectal cancer is strongly influenced by histological subtype. Ann. Oncol. 25, 651–657 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Ponten, F., Jirstrom, K. & Uhlen, M. The Human Protein Atlas—a tool for pathology. J. Pathol. 216, 387–393 (2008).

    Article  CAS  PubMed  Google Scholar 

  53. Xiao, W. et al. Roles and clinical significances of ATF6, EMC6, and APAF1 in prognosis of pancreatic cancer. Front. Genet. 12, 730847 (2021).

    Article  CAS  PubMed  Google Scholar 

  54. Zou, H., Henzel, W. J., Liu, X., Lutschg, A. & Wang, X. APAF-1, a human protein homologous to C. elegans CED-4, participates in cytochrome c-dependent activation of caspase-3. Cell 90, 405–413 (1997).

    Article  CAS  PubMed  Google Scholar 

  55. Jagot-Lacoussiere, L. et al. DNA damage-induced nuclear translocation of APAF-1 is mediated by nucleoporin Nup107. Cell Cycle 14, 1242–1251 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Ferraro, E. et al. APAF1 plays a pro-survival role by regulating centrosome morphology and function. J. Cell Sci. 124, 3450–3463 (2011).

    Article  CAS  PubMed  Google Scholar 

  57. Dillekas, H., Rogers, M. S. & Straume, O. Are 90% of deaths from cancer caused by metastases? Cancer Med. 8, 5574–5576 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  58. Zeng, D. et al. Immunosuppressive microenvironment revealed by immune cell landscape in pre-metastatic liver of colorectal cancer. Front. Oncol. 11, 620688 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Kaczanowska, S. et al. Genetically engineered myeloid cells rebalance the core immune suppression program in metastasis. Cell 184, 2033–2052 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Awad, R. M., De Vlaeminck, Y., Maebe, J., Goyvaerts, C. & Breckpot, K. Turn back the TIMe: targeting tumor infiltrating myeloid cells to revert cancer progression. Front. Immunol. 9, 1977 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  61. Labani-Motlagh, A., Ashja-Mahdavi, M. & Loskog, A. The tumor microenvironment: a milieu hindering and obstructing antitumor immune responses. Front. Immunol. 11, 940 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Zhang, Y. & Zhang, Z. The history and advances in cancer immunotherapy: understanding the characteristics of tumor-infiltrating immune cells and their therapeutic implications. Cell. Mol. Immunol. 17, 807–821 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Bojmar, L. et al. Extracellular vesicle and particle isolation from human and murine cell lines, tissues, and bodily fluids. STAR Protoc. 2, 100225 (2021).

    Article  CAS  PubMed  Google Scholar 

  64. Livak, K. J. & Schmittgen, T. D. Analysis of relative gene expression data using real-time quantitative PCR and the 2ΔΔCT method. Methods 25, 402–408 (2001).

    Article  CAS  PubMed  Google Scholar 

  65. Bojmar, L. et al. Protocol for cross-platform characterization of human and murine extracellular vesicles and particles. STAR Protoc. 5, 102754 (2024).

    Article  CAS  PubMed  Google Scholar 

  66. Qian, B. Z. et al. CCL2 recruits inflammatory monocytes to facilitate breast-tumour metastasis. Nature 475, 222–225 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Kang, W. et al. Multiplex spatial protein detection by combining immunofluorescence with immunohistochemistry. Methods Mol. Biol. 2593, 233–244 (2023).

    Article  CAS  PubMed  Google Scholar 

  68. Liu, Z., Gu, Y., Shin, A., Zhang, S. & Ginhoux, F. Analysis of myeloid cells in mouse tissues with flow cytometry. STAR Protoc. 1, 100029 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  69. Joung, J. et al. Genome-scale CRISPR–Cas9 knockout and transcriptional activation screening. Nat. Protoc. 12, 828–863 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Shechter, D., Dormann, H. L., Allis, C. D. & Hake, S. B. Extraction, purification and analysis of histones. Nat. Protoc. 2, 1445–1457 (2007).

    Article  CAS  PubMed  Google Scholar 

  71. Sidoli, S. & Garcia, B. A. Characterization of individual histone posttranslational modifications and their combinatorial patterns by mass spectrometry-based proteomics strategies. Methods Mol. Biol. 1528, 121–148 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Sidoli, S., Bhanu, N. V., Karch, K. R., Wang, X. & Garcia, B. A.Complete workflow for analysis of histone post-translational modifications using bottom-up mass spectrometry: from histone extraction to data analysis. J. Vis. Exp. 111, e54112 (2016).

    Google Scholar 

  73. Sidoli, S., Simithy, J., Karch, K. R., Kulej, K. & Garcia, B. A. Low resolution data-independent acquisition in an LTQ-Orbitrap allows for simplified and fully untargeted analysis of histone modifications. Anal. Chem. 87, 11448–11454 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Zhou, Y. et al. Metascape provides a biologist-oriented resource for the analysis of systems-level datasets. Nat. Commun. 10, 1523 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  75. Bu, D. et al. KOBAS-i: intelligent prioritization and exploratory visualization of biological functions for gene enrichment analysis. Nucleic Acids Res. 49, W317–W325 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We acknowledge the Genomics Resource Core Facility (Weill Cornell Medicine), Electron Microscopy and Histology Core Facility (Weill Cornell Medicine) and Molecular Cytology Core Facility (Memorial Sloan Kettering Cancer Center, MSKCC) for their high-quality service. We also acknowledge support from T. Miller and M. Lima Silva Santos with flow cytometry and cell sorting. We thank members of the Lyden laboratory for insightful discussions. We gratefully acknowledge support from the National Cancer Institute (CA224175 (D.L.), CA210240 (D.L.), CA232093 (D.L.), CA207983 (D.L.), CA169538 (D.L.) and CA218513 (D.L. and H.Z.)), the Malcolm Hewitt Weiner Foundation, the Manning Foundation, the Hartwell Foundation and the Children’s Cancer and Blood Foundation (all to D.L.), the Beth C. Tortolani Foundation, Lerner Foundation, Gitty Leiner Fund, Sussman Family Fund and MSKCC core grant P30 CA008748 (all to J.B.), a Research Assistance for Primary Parents grant from the Mastercard Diversity–Mentorship Collaborative at Weill Cornell Medicine and Worldwide Cancer Research WRC 23-0105 (I.W.), a National Research Foundation of Korea grant funded by the Korean government (MSIT) (2022R1A2C4001879 to H.S.K.), a grant of the Korea Health Technology R&D Project through the Korea Health Industry Development Institute (KHIDI), funded by the Ministry of Health & Welfare, Republic of Korea (HI22C0353 and RS-2023-00304686 to H.S.K. and HI19C1330 to D.K.), the Bio & Medical Technology Development Program of the National Research Foundation (NRF) & funded by the Korean government (Ministry of Science and ICT) (2022M3A9F3016364 to H.S.K.), the Hankookilbo Myung-Ho Seung Faculty Research Assistance Program of Yonsei University College of Medicine (6-2023-0172 to H.S.K.), Institute for Project-Y funded by Yonsei University (H.S.K.), a research grant (H.S.K.) funded by the CKDBiO-YONSEI Microbiome Research Center (CYMRC) and the Alchemist Project through the Korean Ministry of Trade, Industry and Energy (20012443 to H.S.K.). The Y.D. Lab is supported by the Josie Robertson Foundation, the Pershing Square Sohn Cancer Research Alliance, the National Institutes of Health (CCSG core Grant No. P30 CA008748, MSK SPORE P50 CA192937 and R35 GM138386), the Parker Institute for Cancer Immunotherapy, the STARR Cancer Alliance award and the Anna Fuller Trust. In addition, the Y.D. lab is supported by W. H. Goodwin, A. Goodwin, the Commonwealth Foundation for Cancer Research and the Center for Experimental Therapeutics at MSKCC. Several illustrations for this manuscript were created with BioRender.com.

Author information

Authors and Affiliations

Authors

Contributions

I.W. designed the experimental approach, performed the experimental work, analyzed the data, coordinated the project and wrote the manuscript. Y.S., I.A., L.S., G.C.T., J.H., K.-A.K., D.K., S.D., Y.L., G.A., M.C., T.H., M.Z.T., A.K. and R.M. performed experimental work. K.E.J. performed and analyzed the whole-genome sequencing. Y.D.H., D.H.H., S.P., J.-S.S., M.J., J.Y.P. and M.G.L. provided patient samples. M.V.R., D.Z., J.B. and S.M. generated APAF1 KO in murine cells. S.S. performed and analyzed the PTM MS for histones. K.M. and B.K.T. gave feedback on the project. R.E.S., N.B., I.M. and H.Z. read the manuscript and gave feedback on the project. J.B. and Y.D. designed the experimental approach, read the manuscript and gave feedback on the project. H.S.K. designed the experimental approach, analyzed the data and wrote the manuscript. D.L. coordinated and designed the experimental approach, coordinated the project, interpreted the data and wrote the manuscript.

Corresponding authors

Correspondence to Jacqueline Bromberg, Yael David, Han Sang Kim or David Lyden.

Ethics declarations

Competing interests

D.L. is on the scientific advisory board of Aufbau Holdings. R.E.S. is on the scientific advisory board of Miromatrix and is a speaker and consultant for Alnylam. The remaining authors declare no competing interests.

Peer review

Peer review information

Nature Cancer thanks Lei Qi, Janusz Rak and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Inclusion and diversity statement We support inclusive, diverse, and equitable conduct of research.

Extended data

Extended Data Fig. 1 EV-DNA originates from genomic DNA.

(a,b) A549 and K562 EVs were treated with or without DNase 0, and EV-DNA was extracted. (a) EV-DNA was quantified using Qubit3 and normalized to EV protein content. Data are mean ± s.e.m., analyzed using a two-tailed, unpaired Student’s t-test, with n = 4 independent experiments per group. (b) EV-DNA was analyzed using chip-based capillary electrophoresis (Bioanalyzer). The data are representative of n = 3 independent experiments. (c) HCT116 cells were treated with 1 ug/ml puromycin for 24 h to induce apoptosis, with untreated cells as a control. Lysates were prepared from cells and the indicated EVs and analyzed by Western blot using the indicated antibodies. (d) Fragment coverage for paired-end whole genome sequencing (WGS) of EV-DNA (with or without DNase 0) and gDNA in HCT116 and MDA-MB-231 cells. Each box represents one chromosome, and the y-axis shows relative read count abundance along the genome.

Source data

Extended Data Fig. 2 APAF1 and NCF1 regulate EV-DNA packaging.

(a-b) EVs were isolated from control and APAF1 KO cells. (a) Mode diameter of EVs by nanoparticle tracking analysis. Data are mean ± s.e.m., analyzed using a two-tailed, unpaired Student’s t-test, with n = 4 independent experiments for HCT116 and n = 3 independent experiments for MDA-MB-231. (b) EVs protein amount (μg) was measured by BCA analysis and normalized to cell number at the time of collection. Data are mean ± s.e.m., analyzed using a two-tailed, unpaired Student’s t-test, with n = 6 independent experiments for HCT116 and n = 5 independent experiments for MDA-MB-231. (c) Venn diagram showing unique and overlapping proteins identified by MS/MS between control and APAF1 KO EVs in each cell line. (d-g) EVs were isolated from control and NCF1 KO cells. (d) Transmission electron microscopy for EVs, representative of n = 3 independent experiments. Scale bar: 200 nm. (e) Mode diameter of EVs by nanoparticle tracking analysis. Data are mean ± s.e.m., analyzed using a two-tailed, unpaired Student’s t-test, with n = 4 independent experiments for HCT116 and n = 3 independent experiments for MDA-MB-231 from independent experiments. (f) EVs protein amount (μg) was measured by BCA analysis and normalized to cell number at the time of collection. Data are mean ± s.e.m., analyzed using a two-tailed, unpaired Student’s t-test, with n = 6 independent experiments for HCT116 and n = 5 independent experiments for MDA-MB-231 from independent experiments. (g) EV-DNA was extracted and quantified using Qubit3, normalized to EV protein content. Data are mean ± s.e.m., analyzed using a two-tailed, unpaired Student’s t-test, with n = 5 independent experiments per group. (h) Genomic DNA and EV-DNA were isolated from HCT116 or MDA-MB-231 cells expressing control or NCF1 KO sgRNAs. qPCR was used to detect sgRNA levels in gDNA and EV-DNA and normalized to control sgRNA sequence in gDNA. Data are mean ± s.e.m., analyzed using a two-tailed, unpaired Student’s t-test, n = 5 independent experiments per group from independent experiments. (i) Validation of APAF1 knockdown in NCF1 KO cell lines, the data represent two independent experiments. (j) EV-DNA was extracted from EVs derived from the indicated cell lines. Data are mean ± s.e.m., analyzed using a two-tailed, unpaired Student’s t-test, with n = 4 independent experiments per group.

Source data

Extended Data Fig. 3 Education of NSG mice with HCT116 EVs has no effect on metastasis formation.

(a) Gating strategy for GFP-positive cells. (b-g) NOD.Cg mice were educated 3 times a week for 3 weeks with 10 μg of EVs (Control or APAF1 KO) from HCT116 cells, or PBS, followed by intra-splenically injection of control GFP-Luc-labeled cells (5×104/mouse). Tumors were allowed to grow for 3-4 weeks with continued education. Experiments were performed twice in male and female mice. (b) Schematics of the education experiment. (c) Representative image of tumors in the spleen. (d) Tumor weight in grams. Data are mean ± s.e.m., analyzed by one-way ANOVA (n = 8 mice per Con group, n = 8 mice per PBS group, and n = 9 mice per KO group from n = 2 independent experiments). (e) Representative IVIS images. (f) Quantification of IVIS data. Data are mean ± s.e.m., analyzed by one-way ANOVA (n = 9 mice per Con group, n = 10 mice per KO group, and n = 10 mice per PBS group, from n = 2 independent experiments). (g) One liver lobe per mouse was analyzed by flow cytometry for GFP-positive cells. Data are mean ± s.e.m., one-way ANOVA (n = 9 mice per Con group, n = 11 mice per KO group, and n = 9 mice per PBS group from n = 2 independent experiments). (h-l) NSG mice educated 3 times a week for 3 weeks with 10 µg of EVs (Control or KO) from HCT116 cells, or PBS, were then intra-splenically injected with APAF1 KO cells (5×104/mouse) labeled with GFP-Luc. Tumors were allowed to grow for 3-4 weeks with continued education. The experiment was performed with 5 mice per group from one experiment in female mice. (h) Representative image of tumors in the spleen. (i) Tumor weight in grams. Data are mean ± s.e.m., analyzed by one-way ANOVA, with n = 5 mice per group from one experiment. (j) Representative IVIS images. (k) Quantification of IVIS data. Data are mean ± s.e.m., analyzed by one-way ANOVA, with n = 5 mice per group from one experiment. (l) One liver lobe per mouse was digested and analyzed by flow cytometry for GFP-positive cells. Data are mean ± s.e.m., analyzed by one-way ANOVA, with n = 5 mice per group from one experiment.

Source data

Extended Data Fig. 4 Generation of APAF1 KO in murine cancer cells.

(a-f) APAF1 was knocked out in CT26 cells, and EVs were isolated from control and APAF1 KO CT26 cells. (a) Total cell extracts were prepared from WT and APAF1 KO cells, and the lysates were analyzed by Western blot using the indicated antibodies. (b) EVs were analyzed by transition electron microscopy (TEM). Scale bar: 200 nm. (c) Nanoparticle tracking analysis; the histogram represents 3 independent readings per EV sample and cell type. (d) EV protein content was measured using BCA analysis and normalized to the number of cells at the time of collection. Data are mean ± s.e.m., analyzed using a two-tailed, unpaired Student’s t-test, with n = 3 independent experiments per group. (e) EV-DNA was extracted and quantified using Qubit3, normalized to EV protein content. Data are mean ± s.e.m., analyzed by two-tailed, unpaired Student’s t-test, with n = 3 independent experiments per group. (f) EV-DNA was analyzed using chip-based capillary electrophoresis (Bioanalyzer). (g-l) APAF1 was knocked out in KPC cells, and EVs were isolated from control and APAF1 KO KPC cells. (g) Total cell extracts were prepared from WT and APAF1 KO cells, and the lysates were analyzed by Western blot using the indicated antibodies. (h) EVs were visualized by transition electron microscopy. Scale bar: 200 nm. (i) Nanoparticle tracking analysis; the histogram represents 3 independent readings per EV sample and cell type. (j) EV protein content (mg) was measured by BCA analysis and normalized to the number of cells at the time of collection. Data are mean ± s.e.m., analyzed by a two-tailed, unpaired Student’s t-test, with n = 10 per cell type from 5 independent experiments. (k) EV-DNA was extracted and quantified using Qubit3, normalized to protein content. Data are mean ± s.e.m., analyzed by two-tailed, unpaired Student’s t-test, with n = 7 per cell type from 4 independent experiments. (l) EV-DNA was analyzed using chip-based capillary electrophoresis (Bioanalyzer).

Source data

Extended Data Fig. 5 Reduced EV-DNA induces pro-tumor programming in KCs.

(a) Gating strategy for immune cell populations in the liver. (b) Percentages of EV+CD31+ cells. Data are mean ± s.e.m., analyzed by one-way ANOVA, with n = 6 mice per group from two independent experiments. (c-d) RNA sequencing analysis of Kupffer cells (KCs) isolated from n = 4 mice per group from two independent experiments after treatment with CT26 control or APAF1 KO EVs. (c) Gene Set Enrichment Analysis (GSEA) of pathways enriched in KCs treated with control or APAF1 KO EVs. (d) Pathway analysis highlighting those pathways upregulated in KCs from mice treated with APAF1 KO EVs compared to control EV-treated mice.

Source data

Extended Data Fig. 6 EV-DNA is damaged and induces cytokines secretion from KCs.

(a) Total protein was extracted from HCT116 and CT26 cells and EVs. The extracts (5 μg) were analyzed by Western blot for the indicated proteins. Data are representative of n = 2 independent experiments. (b) Primary KCs were isolated from the livers of naïve mice, plated in 48-well plates, and allowed to adhere for 16-24 hr. The KCs were then treated with the LPS (1 mg/ml) or control EVs (10 µg) for 24 h, after which cytokines secreted in the media were quantified (LegendPlex assay). Data summarize n = 2 independent repeats. (c) Balb/c mice were educated 3 times per week for 3 weeks with 10 µg of EVs (control or APAF1 KO) derived from CT26 cells or PBS. One liver lobe from each mouse was used for flow cytometric analysis of immune cell populations in the liver. Data are mean ± s.e.m., analyzed by one-way ANOVA, with n = 3 mice per group from one experiment. (d) Balb/c mice were educated 3 times per week for 3 weeks with 10 µg of EVs derived from CT26 cells. The liver was extracted and stained for analysis. Left panel: A representative liver section stained with multiplex immunofluorescence, showing B cells (B220, green), T cells (CD3, white), dendritic cells (CD11c, red), and DAPI (blue). Right panel: Following IF imaging, coverslips were removed, and immunohistochemistry staining for vWF was performed. Scale bar: 250 µm. Data represent n = 3 independent experiments.

Source data

Supplementary information

Reporting Summary

Supplementary Tables

Supplementary Table 1: sgRNA sequences and corresponding read counts from the genome-wide CRISPR KO screen. Supplementary Table 2: List of genes ranked by the top 10% of sgRNAs identified from genome-wide CRISPR KO screening in MDA-MB-231, HCT116, A549 and K562 cells. Supplementary Table 3: Pathway analysis of the top candidate genes associated with the regulation or inhibition of EV-DNA packaging, identified from the genome-wide CRISPR screen. Supplementary Table 4: List of 612 genes, 6,410 sgRNA sequences and corresponding read counts obtained from next-generation sequencing of EV-DNA and gDNA. Supplementary Table 5: Gene list ranked by secondary screening in MDA-MB-231 and HCT116 cells. Supplementary Table 6: Top enriched pathways of 612 genes ranked by secondary screening. Supplementary Table 7: Participant characteristics. Supplementary Table 8: Univariate and multivariate Cox regression analyses for disease-free survival. Supplementary Table 9: List of plasmids and primers used in the study. Supplementary Table 10: List of antibodies used in this study.

Source data

Source Data

Supporting blots for main and extended data figures.

Source Data

Raw data for all figures and extended data figures.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wortzel, I., Seo, Y., Akano, I. et al. Unique structural configuration of EV-DNA primes Kupffer cell-mediated antitumor immunity to prevent metastatic progression. Nat Cancer 5, 1815–1833 (2024). https://doi.org/10.1038/s43018-024-00862-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s43018-024-00862-6

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer