nature.com

Genetic tools for investigating the life cycle of extracellular vesicles

Abstract

Mammalian cells ubiquitously release membrane-enclosed vesicles, known as extracellular vesicles. These particles carry a variety of molecules that reflect the status of their cells of origin, making them valuable sources for biomarker discovery. Furthermore, extracellular vesicles deliver their cargo locally and systemically to regulate biological processes, piquing interest in modulating extracellular vesicle biogenesis and developing extracellular vesicle-based therapies. Therefore, a thorough understanding of the extracellular vesicle life cycle, from biogenesis and trafficking to degradation, is essential for unlocking their full potential in biomarker identification and for the design of extracellular vesicle-based therapies. In this Review, we start by outlining the key steps in the extracellular vesicle life cycle and highlight remaining open questions. We then discuss the design and application of genetically encoded systems that can be applied to study extracellular vesicle biogenesis and fate. Finally, we highlight technical challenges that remain to be addressed in the engineering and application of genetically encoded systems to extracellular vesicle research.

Key points

Extracellular vesicles are shed by cells and implicated in biological processes through diverse mechanisms.

Specific and sensitive characterization of extracellular vesicles is fundamental for identifying extracellular vesicle-based biomarkers and developing therapies. However, their small size and close resemblance to other non-vesicular extracellular particles make their characterization challenging.

Genetically encoded systems allow the high-specificity and high-sensitivity characterization of the extracellular vesicle life cycle, from biogenesis and trafficking to degradation.

Genetic circuits can be incorporated for the long-term tracking of extracellular vesicle biogenesis and biodistribution in vivo.

Caution must be applied when extrapolating knowledge of genetically engineered extracellular vesicles to their native counterparts, which requires complementary approaches.

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

Access options

Access through your institution

Change institution

Buy or subscribe

Subscribe to this journal

Receive 12 digital issues and online access to articles

$119.00 per year

only $9.92 per issue

Learn more

Buy this article

Purchase on SpringerLink

Instant access to full article PDF

Buy now

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

Additional access options:

Log in

Learn about institutional subscriptions

Read our FAQs

Contact customer support

Fig. 1: Biogenesis, trafficking and degradation of extracellular vesicles.

Fig. 2: Genetic methods to capture extracellular vesicle-inherent and extracellular vesicle-associated molecules.

Fig. 3: Reporter systems to identify EV-recipient cells.

Fig. 4: Genetic methods to investigate endocytic and non-endocytic routes of EV–cell interaction.

References

Welsh, J. A. et al. Minimal information for studies of extracellular vesicles (MISEV2023): from basic to advanced approaches. J. Extracell. Vesicles 13, e12404 (2024).

ArticleMATHGoogle Scholar

Poupardin, R., Wolf, M. & Strunk, D. Adherence to minimal experimental requirements for defining extracellular vesicles and their functions. Adv. Drug Deliv. Rev. 176, 113872 (2021).

ArticleMATHGoogle Scholar

Lian, M. Q. et al. Plant-derived extracellular vesicles: recent advancements and current challenges on their use for biomedical applications. J. Extracell. Vesicles 11, e12283 (2022).

ArticleGoogle Scholar

Hosseini-Giv, N. et al. Bacterial extracellular vesicles and their novel therapeutic applications in health and cancer. Front. Cell. Infect. Microbiol. 12, 962216 (2022).

ArticleMATHGoogle Scholar

Rodrigues, M. L. & Nimrichter, L. From fundamental biology to the search for innovation: the story of fungal extracellular vesicles. Eur. J. Cell Biol. 101, 151205 (2022).

ArticleGoogle Scholar

Cheng, L. & Hill, A. F. Therapeutically harnessing extracellular vesicles. Nat. Rev. Drug Discov. 21, 379–399 (2022).

ArticleGoogle Scholar

Herrmann, I. K., Wood, M. J. A. & Fuhrmann, G. Extracellular vesicles as a next-generation drug delivery platform. Nat. Nanotechnol. 16, 748–759 (2021).

ArticleGoogle Scholar

Urabe, F. et al. Extracellular vesicles as biomarkers and therapeutic targets for cancer. Am. J. Physiol. Cell Physiol. 318, C29–C39 (2020).

ArticleMATHGoogle Scholar

Shaba, E. et al. Multi-omics integrative approach of extracellular vesicles: a future challenging milestone. Proteomes 10, 12 (2022).

ArticleMathSciNetMATHGoogle Scholar

Hendrix, A. et al. Extracellular vesicle analysis. Nat. Rev. Methods Primers 3, 56 (2023).

ArticleMATHGoogle Scholar

Rupert, D. L. M., Claudio, V., Lässer, C. & Bally, M. Methods for the physical characterization and quantification of extracellular vesicles in biological samples. Biochim. Biophys. Acta Gen. Subj. 1861, 3164–3179 (2017).

ArticleMATHGoogle Scholar

Hartjes, T., Mytnyk, S., Jenster, G., van Steijn, V. & van Royen, M. E. Extracellular vesicle quantification and characterization: common methods and emerging approaches. Bioengineering 6, 7 (2019).

ArticleGoogle Scholar

Penders, J. et al. Single particle automated Raman trapping analysis. Nat. Commun. 9, 4256 (2018).

ArticleMATHGoogle Scholar

Ter-Ovanesyan, D. et al. Improved isolation of extracellular vesicles by removal of both free proteins and lipoproteins. eLife 12, e86394 (2023).

ArticleGoogle Scholar

Karimi, N. et al. Detailed analysis of the plasma extracellular vesicle proteome after separation from lipoproteins. Cell. Mol. Life Sci. 75, 2873–2886 (2018).

ArticleMATHGoogle Scholar

Wang, T. & Turko, I. V. Proteomic toolbox to standardize the separation of extracellular vesicles and lipoprotein particles. J. Proteome Res. 17, 3104–3113 (2018).

ArticleMATHGoogle Scholar

Serrano-Pertierra, E. et al. Extracellular vesicles: current analytical techniques for detection and quantification. Biomolecules 10, 824 (2020).

ArticleMATHGoogle Scholar

Verweij, F. J. et al. The power of imaging to understand extracellular vesicle biology in vivo. Nat. Methods 18, 1013–1026 (2021).

ArticleMATHGoogle Scholar

Lai, C. P. et al. Dynamic biodistribution of extracellular vesicles in vivo using a multimodal imaging reporter. ACS Nano 8, 483–494 (2014).

ArticleMATHGoogle Scholar

Cheng, A. A. & Lu, T. K. Synthetic biology: an emerging engineering discipline. Annu. Rev. Biomed. Eng. 14, 155–178 (2012).

ArticleMATHGoogle Scholar

Kelwick, R. J. R., Webb, A. J., Heliot, A., Segura, C. T. & Freemont, P. S. Opportunities to accelerate extracellular vesicle research with cell-free synthetic biology. J. Extracell. Biol. 2, e90 (2023).

ArticleGoogle Scholar

Buzas, E. I. Opportunities and challenges in studying the extracellular vesicle corona. Nat. Cell Biol. 24, 1322–1325 (2022).

ArticleMATHGoogle Scholar

van Niel, G., D'Angelo, G. & Raposo, G. Shedding light on the cell biology of extracellular vesicles. Nat. Rev. Mol. Cell Biol. 19, 213–228 (2018).

ArticleGoogle Scholar

Edgar, J. R., Eden, E. R. & Futter, C. E. Hrs- and CD63-dependent competing mechanisms make different sized endosomal intraluminal vesicles. Traffic 15, 197–211 (2014).

ArticleGoogle Scholar

Stuffers, S., Sem Wegner, C., Stenmark, H. & Brech, A. Multivesicular endosome biogenesis in the absence of ESCRTs. Traffic 10, 925–937 (2009).

ArticleGoogle Scholar

Verweij, F. J. et al. ER membrane contact sites support endosomal small GTPase conversion for exosome secretion. J. Cell Biol. 221, e202112032 (2022).

ArticleMATHGoogle Scholar

Miranda, A. M. et al. Neuronal lysosomal dysfunction releases exosomes harboring APP C-terminal fragments and unique lipid signatures. Nat. Commun. 9, 291 (2018).

ArticleGoogle Scholar

Eitan, E., Suire, C., Zhang, S. & Mattson, M. P. Impact of lysosome status on extracellular vesicle content and release. Ageing Res. Rev. 32, 65–74 (2016).

ArticleGoogle Scholar

Strauss, K. et al. Exosome secretion ameliorates lysosomal storage of cholesterol in Niemann–Pick type C disease. J. Biol. Chem. 285, 26279–26288 (2010).

ArticleGoogle Scholar

Teng, F. & Fussenegger, M. Shedding light on extracellular vesicle biogenesis and bioengineering. Adv. Sci. 8, 2003505 (2021).

ArticleGoogle Scholar

Margolis, L. & Sadovsky, Y. The biology of extracellular vesicles: the known unknowns. PLoS Biol. 17, e3000363 (2019).

ArticleMATHGoogle Scholar

Hurwitz, S. N., Conlon, M. M., Rider, M. A., Brownstein, N. C. & Meckes, D. G. Jr Nanoparticle analysis sheds budding insights into genetic drivers of extracellular vesicle biogenesis. J. Extracell. Vesicles 5, 31295 (2016).

ArticleGoogle Scholar

Bost, J. P. et al. Growth media conditions influence the secretion route and release levels of engineered extracellular vesicles. Adv. Healthc. Mater. 11, e2101658 (2021).

ArticleMATHGoogle Scholar

Shpigelman, J. et al. Generation and application of a reporter cell line for the quantitative screen of extracellular vesicle release. Front. Pharmacol. 12, 668609 (2021).

ArticleMATHGoogle Scholar

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

ArticleMATHGoogle Scholar

Dixson, A. C., Dawson, T. R., Di Vizio, D. & Weaver, A. M. Context-specific regulation of extracellular vesicle biogenesis and cargo selection. Nat. Rev. Mol. Cell Biol. 24, 454–476 (2023).

ArticleMATHGoogle Scholar

Lenzini, S., Bargi, R., Chung, G. & Shin, J. W. Matrix mechanics and water permeation regulate extracellular vesicle transport. Nat. Nanotechnol. 15, 217–223 (2020).

ArticleGoogle Scholar

Gupta, D., Wiklander, O. P. B., Wood, M. J. A. & El-Andaloussi, S. Biodistribution of therapeutic extracellular vesicles. Extracell. Vesicles Circ. Nucl. Acids 4, 170–190 (2023).

ArticleMATHGoogle Scholar

Sariano, P. A. et al. Convection and extracellular matrix binding control interstitial transport of extracellular vesicles. J. Extracell. Vesicles 12, e12323 (2023).

ArticleMATHGoogle Scholar

Debnath, K., Las Heras, K., Rivera, A., Lenzini, S. & Shin, J.-W. Extracellular vesicle–matrix interactions. Nat. Rev. Mater. 8, 390–402 (2023).

ArticleGoogle Scholar

Hallal, S., Tűzesi, Á., Grau, G. E., Buckland, M. E. & Alexander, K. L. Understanding the extracellular vesicle surface for clinical molecular biology. J. Extracell. Vesicles 11, e12260 (2022).

ArticleGoogle Scholar

Tóth, E. Á. et al. Formation of a protein corona on the surface of extracellular vesicles in blood plasma. J. Extracell. Vesicles 10, e12140 (2021).

ArticleMATHGoogle Scholar

Ghosh, P., Liu, Q.-R., Chen, Q., Zhu, M. & Egan, J. M. Pancreatic β cell derived extracellular vesicles containing surface preproinsulin are involved in glucose stimulated insulin secretion. Life Sci. 340, 122460 (2024).

ArticleGoogle Scholar

Gupta, D. et al. Quantification of extracellular vesicles in vitro and in vivo using sensitive bioluminescence imaging. J. Extracell. Vesicles 9, 1800222 (2020).

ArticleGoogle Scholar

Vidal, M. Exosomes: revisiting their role as “garbage bags”. Traffic 20, 815–828 (2019).

ArticleMATHGoogle Scholar

Keller, M. D. et al. Decoy exosomes provide protection against bacterial toxins. Nature 579, 260–264 (2020).

ArticleGoogle Scholar

Buzás, E. I., Tóth, E. Á., Sódar, B. W. & Szabó-Taylor, K. É. Molecular interactions at the surface of extracellular vesicles. Semin. Immunopathol. 40, 453–464 (2018).

ArticleGoogle Scholar

Roy, S., Hochberg, F. H. & Jones, P. S. Extracellular vesicles: the growth as diagnostics and therapeutics; a survey. J. Extracell. Vesicles 7, 1438720 (2018).

ArticleGoogle Scholar

Edelmann, M. J. & Kima, P. E. Current understanding of extracellular vesicle homing/tropism. Zoonoses 2, 14 (2022).

ArticleMATHGoogle Scholar

Liam-Or, R. et al. Cellular uptake and in vivo distribution of mesenchymal-stem-cell-derived extracellular vesicles are protein corona dependent. Nat. Nanotechnol. 19, 846–855 (2024).

ArticleMATHGoogle Scholar

Heidarzadeh, M., Zarebkohan, A., Rahbarghazi, R. & Sokullu, E. Protein corona and exosomes: new challenges and prospects. Cell Commun. Signal. 21, 64 (2023).

ArticleGoogle Scholar

Nishida-Aoki, N., Tominaga, N., Kosaka, N. & Ochiya, T. Altered biodistribution of deglycosylated extracellular vesicles through enhanced cellular uptake. J. Extracell. Vesicles 9, 1713527 (2020).

ArticleGoogle Scholar

Gonda, A., Kabagwira, J., Senthil, G. N. & Wall, N. R. Internalization of exosomes through receptor-mediated endocytosis. Mol. Cancer Res. 17, 337–347 (2019).

ArticleGoogle Scholar

Qian, F. et al. Analysis and biomedical applications of functional cargo in extracellular vesicles. ACS Nano 16, 19980–20001 (2022).

ArticleGoogle Scholar

Bonsergent, E. et al. Quantitative characterization of extracellular vesicle uptake and content delivery within mammalian cells. Nat. Commun. 12, 1864 (2021).

ArticleMATHGoogle Scholar

Somiya, M. & Kuroda, S. Real-time luminescence assay for cytoplasmic cargo delivery of extracellular vesicles. Anal. Chem. 93, 5612–5620 (2021).

ArticleMATHGoogle Scholar

Vargas, A. et al. Syncytin proteins incorporated in placenta exosomes are important for cell uptake and show variation in abundance in serum exosomes from patients with preeclampsia. FASEB J. 28, 3703–3719 (2014).

ArticleMATHGoogle Scholar

Zhang, X. et al. Programmable extracellular vesicles for macromolecule delivery and genome modifications. Dev. Cell 55, 784–801.e9 (2020).

ArticleMATHGoogle Scholar

Somiya, M. & Kuroda, S. Reporter gene assay for membrane fusion of extracellular vesicles. J. Extracell. Vesicles 10, e12171 (2021).

ArticleGoogle Scholar

Nambiar, D., Le, Q.-T. & Pucci, F. A case for the study of native extracellular vesicles. Front. Oncol. 14, 1430971 (2024).

ArticleMATHGoogle Scholar

Kowal, J. et al. Proteomic comparison defines novel markers to characterize heterogeneous populations of extracellular vesicle subtypes. Proc. Natl Acad. Sci. USA 113, E968–E977 (2016).

ArticleMATHGoogle Scholar

Mathieu, M. et al. Specificities of exosome versus small ectosome secretion revealed by live intracellular tracking of CD63 and CD9. Nat. Commun. 12, 4389 (2021).

ArticleGoogle Scholar

Liu, D.-A. et al. A phosphoinositide switch mediates exocyst recruitment to multivesicular endosomes for exosome secretion. Nat. Commun. 14, 6883 (2023).

ArticleMATHGoogle Scholar

Bebelman, M. P. et al. Real-time imaging of multivesicular body–plasma membrane fusion to quantify exosome release from single cells. Nat. Protoc. 15, 102–121 (2020).

ArticleGoogle Scholar

Sung, B. H. et al. A live cell reporter of exosome secretion and uptake reveals pathfinding behavior of migrating cells. Nat. Commun. 11, 2092 (2020).

ArticleMATHGoogle Scholar

Verweij, F. J. et al. Quantifying exosome secretion from single cells reveals a modulatory role for GPCR signaling. J. Cell Biol. 217, 1129–1142 (2018).

ArticleMATHGoogle Scholar

Morimoto, Y. V., Kojima, S., Namba, K. & Minamino, T. M153R mutation in a pH-sensitive green fluorescent protein stabilizes its fusion proteins. PLoS ONE 6, e19598 (2011).

ArticleGoogle Scholar

Liu, A. et al. pHmScarlet is a pH-sensitive red fluorescent protein to monitor exocytosis docking and fusion steps. Nat. Commun. 12, 1413 (2021).

ArticleMATHGoogle Scholar

Bebelman, M. P. et al. Exosomal release of the virus-encoded chemokine receptor US28 contributes to chemokine scavenging. iScience 26, 107412 (2023).

ArticleGoogle Scholar

Beer, K. B. et al. Degron-tagged reporters probe membrane topology and enable the specific labelling of membrane-wrapped structures. Nat. Commun. 10, 3490 (2019).

ArticleMATHGoogle Scholar

Datta, A. et al. High-throughput screening identified selective inhibitors of exosome biogenesis and secretion: a drug repurposing strategy for advanced cancer. Sci. Rep. 8, 8161 (2018).

ArticleMATHGoogle Scholar

Ruan, Z. et al. Functional genome-wide short hairpin RNA library screening identifies key molecules for extracellular vesicle secretion from microglia. Cell Rep. 39, 110791 (2022).

ArticleMATHGoogle Scholar

Welsh, J. A. et al. A compendium of single extracellular vesicle flow cytometry. J. Extracell. Vesicles 12, e12299 (2023).

ArticleMATHGoogle Scholar

Corso, G. et al. Systematic characterization of extracellular vesicles sorting domains and quantification at the single molecule–single vesicle level by fluorescence correlation spectroscopy and single particle imaging. J. Extracell. Vesicles 8, 1663043 (2019).

ArticleMATHGoogle Scholar

Bebelman, M. P. et al. Luminescence-based screening for extracellular vesicle release modulators reveals a role for PI4KIIIβ in exosome biogenesis upon lysosome inhibition. Preprint at bioRxivhttps://doi.org/10.1101/2023.02.23.529257 (2023).

Chiao, J. J. C., Roberts, J. P., Illner, H. P. & Shires, G. T. Permeability of red-cell membrane to adenosine triphosphate (ATP) molecules during hemorrhagic shock. Surgery 102, 528–533 (1987).

Google Scholar

Zheng, W. et al. Identification of scaffold proteins for improved endogenous engineering of extracellular vesicles. Nat. Commun. 14, 4734 (2023).

ArticleMATHGoogle Scholar

Dixon, A. S. et al. NanoLuc complementation reporter optimized for accurate measurement of protein interactions in cells. ACS Chem. Biol. 11, 400–408 (2016).

ArticleMATHGoogle Scholar

Leidal, A. M. et al. The LC3-conjugation machinery specifies the loading of RNA-binding proteins into extracellular vesicles. Nat. Cell Biol. 22, 187–199 (2020).

ArticleGoogle Scholar

Lu, A. et al. Genome-wide interrogation of extracellular vesicle biology using barcoded miRNAs. eLife 7, e41460 (2018).

ArticleGoogle Scholar

Zeng, Y., Wagner, E. J. & Cullen, B. R. Both natural and designed micro RNAs can inhibit the expression of cognate mRNAs when expressed in human cells. Mol. Cell 9, 1327–1333 (2002).

ArticleGoogle Scholar

Imai, T. et al. Macrophage-dependent clearance of systemically administered B16BL6-derived exosomes from the blood circulation in mice. J. Extracell. Vesicles 4, 26238 (2015).

ArticleMATHGoogle Scholar

Wu, A. Y.-T. et al. Multiresolution imaging using bioluminescence resonance energy transfer identifies distinct biodistribution profiles of extracellular vesicles and exomeres with redirected tropism. Adv. Sci. 7, 2001467 (2020).

ArticleGoogle Scholar

Driedonks, T. et al. Pharmacokinetics and biodistribution of extracellular vesicles administered intravenously and intranasally to Macaca nemestrina. J. Extracell. Biol. 1, e59 (2022).

ArticleMATHGoogle Scholar

Paget, D. et al. Comparative and integrated analysis of plasma extracellular vesicle isolation methods in healthy volunteers and patients following myocardial infarction. J. Extracell. Biol. 1, e66 (2022).

ArticleMATHGoogle Scholar

Visan, K. S. et al. Comparative analysis of tangential flow filtration and ultracentrifugation, both combined with subsequent size exclusion chromatography, for the isolation of small extracellular vesicles. J. Extracell. Vesicles 11, e12266 (2022).

ArticleMATHGoogle Scholar

Hsia, T. et al. Rigorous comparison of extracellular vesicle processing to enhance downstream analysis for glioblastoma characterization. Adv. Biol. 8, e2300233 (2024).

ArticleMATHGoogle Scholar

Zhang, Z. et al. Comprehensive characterization of human brain-derived extracellular vesicles using multiple isolation methods: implications for diagnostic and therapeutic applications. J. Extracell. Vesicles 12, e12358 (2023).

ArticleMATHGoogle Scholar

Nieuwland, R., Siljander, P. R.-M., Falcón-Pérez, J. M. & Witwer, K. W. Reproducibility of extracellular vesicle research. Eur. J. Cell Biol. 101, 151226 (2022).

ArticleGoogle Scholar

Poupardin, R. et al. Advances in extracellular vesicle research over the past decade: source and isolation method are connected with cargo and function. Adv. Healthc. Mater. 13, e2303941 (2024).

ArticleMATHGoogle Scholar

Abyadeh, M. et al. Proteomic profiling of mesenchymal stem cell-derived extracellular vesicles: impact of isolation methods on protein cargo. J. Extracell. Biol. 3, e159 (2024).

ArticleGoogle Scholar

Han, C. et al. CD63-snorkel tagging for isolation of exosomes. Extracell. Vesicle 2, 100031 (2023).

ArticleMATHGoogle Scholar

Rufino-Ramos, D. et al. Using genetically modified extracellular vesicles as a non-invasive strategy to evaluate brain-specific cargo. Biomaterials 281, 121366 (2022).

ArticleGoogle Scholar

Li, W. et al. Construction of a mouse model that can be used for tissue-specific EV screening and tracing in vivo. Front. Cell Dev. Biol. 10, 1015841 (2022).

ArticleGoogle Scholar

Zheng, W. et al. Cell‐specific targeting of extracellular vesicles though engineering the glycocalyx. J. Extracell. Vesicles 11, e12290 (2022).

ArticleGoogle Scholar

Liang, X. et al. Extracellular vesicles engineered to bind albumin demonstrate extended circulation time and lymph node accumulation in mouse models. J. Extracell. Vesicles 11, e12248 (2022).

ArticleGoogle Scholar

Vogt, S. et al. An engineered CD81-based combinatorial library for selecting recombinant binders to cell surface proteins: laminin binding CD81 enhances cellular uptake of extracellular vesicles. J. Extracell. Vesicles 10, e12139 (2021).

ArticleMATHGoogle Scholar

Dooley, K. et al. A versatile platform for generating engineered extracellular vesicles with defined therapeutic properties. Mol. Ther. 29, 1729–1743 (2021).

ArticleMATHGoogle Scholar

Alvarez-Erviti, L. et al. Delivery of siRNA to the mouse brain by systemic injection of targeted exosomes. Nat. Biotechnol. 29, 341–345 (2011).

ArticleGoogle Scholar

Martin Perez, C. et al. An extracellular vesicle delivery platform based on the PTTG1IP protein. Extracell. Vesicle 4, 100054 (2024).

ArticleGoogle Scholar

Ohno, S. et al. Systemically injected exosomes targeted to EGFR deliver antitumor microRNA to breast cancer cells. Mol. Ther. 21, 185–191 (2013).

ArticleGoogle Scholar

Kehrloesser, S. et al. Cell-of-origin–specific proteomics of extracellular vesicles. PNAS Nexus 2, pgad107 (2023).

ArticleGoogle Scholar

Kokkinopoulou, M., Simon, J., Landfester, K., Mailänder, V. & Lieberwirth, I. Visualization of the protein corona: towards a biomolecular understanding of nanoparticle-cell-interactions. Nanoscale 9, 8858–8870 (2017).

ArticleGoogle Scholar

Wolf, M. et al. A functional corona around extracellular vesicles enhances angiogenesis, skin regeneration and immunomodulation. J. Extracell. Vesicles 11, e12207 (2022).

ArticleMATHGoogle Scholar

Choi, D. et al. Quantitative proteomic analysis of trypsin-treated extracellular vesicles to identify the real-vesicular proteins. J. Extracell. Vesicles 9, 1757209 (2020).

ArticleGoogle Scholar

Xu, R. et al. Surfaceome of exosomes secreted from the colorectal cancer cell line SW480: peripheral and integral membrane proteins analyzed by proteolysis and TX114. Proteomics 19, e1700453 (2019).

ArticleGoogle Scholar

Rai, A., Fang, H., Claridge, B., Simpson, R. J. & Greening, D. W. Proteomic dissection of large extracellular vesicle surfaceome unravels interactive surface platform. J. Extracell. Vesicles 10, e12164 (2021).

ArticleGoogle Scholar

Santucci, L. et al. Biological surface properties in extracellular vesicles and their effect on cargo proteins. Sci. Rep. 9, 13048 (2019).

ArticleMATHGoogle Scholar

Kirkemo, L. L. et al. Cell-surface tethered promiscuous biotinylators enable comparative small-scale surface proteomic analysis of human extracellular vesicles and cells. eLife 11, e73982 (2022).

ArticleGoogle Scholar

Chauhan, S. et al. Surface glycoproteins of exosomes shed by myeloid-derived suppressor cells contribute to function. J. Proteome Res. 16, 238–246 (2017).

ArticleMATHGoogle Scholar

Li, Y., Kanao, E., Yamano, T., Ishihama, Y. & Imami, K. TurboID-EV: proteomic mapping of recipient cellular proteins proximal to small extracellular vesicles. Anal. Chem. 95, 14159–14164 (2023).

ArticleGoogle Scholar

Skotland, T., Sandvig, K. & Llorente, A. Lipids in exosomes: current knowledge and the way forward. Prog. Lipid Res. 66, 30–41 (2017).

ArticleGoogle Scholar

Lee, B. R. et al. Ascorbate peroxidase-mediated in situ labelling of proteins in secreted exosomes. J. Extracell. Vesicles 11, e12239 (2022).

ArticleMATHGoogle Scholar

Hagey, D. W. et al. The cellular response to extracellular vesicles is dependent on their cell source and dose. Sci. Adv. 9, eadh1168 (2024).

ArticleGoogle Scholar

Shipunova, V. O., Shilova, O. N., Shramova, E. I., Deyev, S. M. & Proshkina, G. M. A highly specific substrate for nanoLUC luciferase furimazine is toxic in vitro and in vivo. Russ. J. Bioorg. Chem. 44, 225–228 (2018).

ArticleGoogle Scholar

Gaspar, N. et al. Evaluation of NanoLuc substrates for bioluminescence imaging of transferred cells in mice. J. Photochem. Photobiol. B 216, 112128 (2021).

ArticleMATHGoogle Scholar

van Solinge, T. S. et al. Illuminating cellular and extracellular vesicle-mediated communication via a split-Nanoluc reporter in vitro and in vivo. Cell Rep. Methods 3, 100412 (2023).

ArticleGoogle Scholar

Leary, N. et al. Melanoma-derived extracellular vesicles mediate lymphatic remodelling and impair tumour immunity in draining lymph nodes. J. Extracell. Vesicles 11, e12197 (2022).

ArticleMATHGoogle Scholar

Men, Y. et al. Exosome reporter mice reveal the involvement of exosomes in mediating neuron to astroglia communication in the CNS. Nat. Commun. 10, 4136 (2019).

ArticleMATHGoogle Scholar

Hu, Y. et al. Cancer-cell-secreted miR-204-5p induces leptin signalling pathway in white adipose tissue to promote cancer-associated cachexia. Nat. Commun. 14, 5179 (2023).

ArticleGoogle Scholar

Ma, S. et al. Skeletal muscle-derived extracellular vesicles transport glycolytic enzymes to mediate muscle-to-bone crosstalk. Cell Metab. 35, 2028–2043.e7 (2023).

ArticleMATHGoogle Scholar

Hyenne, V. et al. Studying the fate of tumor extracellular vesicles at high spatiotemporal resolution using the zebrafish embryo. Dev. Cell 48, 554–572.e7 (2019).

ArticleGoogle Scholar

Verweij, F. J. et al. Live tracking of inter-organ communication by endogenous exosomes in vivo. Dev. Cell 48, 573–589.e4 (2019).

ArticleMATHGoogle Scholar

Chu, J. et al. A bright cyan-excitable orange fluorescent protein facilitates dual-emission microscopy and enhances bioluminescence imaging in vivo. Nat. Biotechnol. 34, 760–767 (2016).

ArticleMATHGoogle Scholar

Hikita, T., Miyata, M., Watanabe, R. & Oneyama, C. In vivo imaging of long-term accumulation of cancer-derived exosomes using a BRET-based reporter. Sci. Rep. 10, 16616 (2020).

ArticleGoogle Scholar

Schaub, F. X. et al. Fluorophore-NanoLuc BRET reporters enable sensitive in vivo optical imaging and flow cytometry for monitoring tumorigenesis. Cancer Res. 75, 5023–5033 (2015).

ArticleMATHGoogle Scholar

Suzuki, K. et al. Five colour variants of bright luminescent protein for real-time multicolour bioimaging. Nat. Commun. 7, 13718 (2016).

ArticleMATHGoogle Scholar

Zhang, H. et al. Quantitative assessment of near-infrared fluorescent proteins. Nat. Methods 20, 1605–1616 (2023).

ArticleMATHGoogle Scholar

Liang, X. et al. Multimodal engineering of extracellular vesicles for efficient intracellular protein delivery. Preprint at bioRxivhttps://doi.org/10.1101/2023.04.30.535834 (2023).

Zickler, A. M. et al. Novel endogenous engineering platform for robust loading and delivery of functional mRNA by extracellular vesicles. Adv. Sci. 11, e2407619 (2024).

ArticleMATHGoogle Scholar

Zomer, A. et al. In vivo imaging reveals extracellular vesicle-mediated phenocopying of metastatic behavior. Cell 161, 1046–1057 (2015).

ArticleMATHGoogle Scholar

Casella, G. et al. Oligodendrocyte-derived extracellular vesicles as antigen-specific therapy for autoimmune neuroinflammation in mice. Sci. Transl Med. 12, eaba0599 (2020).

ArticleGoogle Scholar

Kang, M., Jordan, V., Blenkiron, C. & Chamley, L. W. Biodistribution of extracellular vesicles following administration into animals: a systematic review. J. Extracell. Vesicles 10, e12085 (2021).

ArticleGoogle Scholar

Kur, I.-M. et al. Neuronal activity triggers uptake of hematopoietic extracellular vesicles in vivo. PLoS Biol. 18, e3000643 (2020).

ArticleMATHGoogle Scholar

Ridder, K. et al. Extracellular vesicle-mediated transfer of genetic information between the hematopoietic system and the brain in response to inflammation. PLoS Biol. 12, e1001874 (2014).

ArticleMATHGoogle Scholar

Valkov, N. et al. SnRNA sequencing defines signaling by RBC-derived extracellular vesicles in the murine heart. Life Sci. Alliance 4, e202101048 (2021).

ArticleMATHGoogle Scholar

Nikolic, J. et al. Structural basis for the recognition of LDL-receptor family members by VSV glycoprotein. Nat. Commun. 9, 1029 (2018).

ArticleMATHGoogle Scholar

de Jong, O. G. et al. A CRISPR-Cas9-based reporter system for single-cell detection of extracellular vesicle-mediated functional transfer of RNA. Nat. Commun. 11, 1113 (2020).

ArticleMATHGoogle Scholar

Platt, R. J. et al. CRISPR-Cas9 knockin mice for genome editing and cancer modeling. Cell 159, 440–455 (2014).

ArticleMATHGoogle Scholar

Zhang, L. et al. sgRNA knock-in mouse provides an alternative approach for in vivo genetic modification. Front. Cell Dev. Biol. 9, 769673 (2022).

ArticleGoogle Scholar

Kranich, J. et al. In vivo identification of apoptotic and extracellular vesicle-bound live cells using image-based deep learning. J. Extracell. Vesicles 9, 1792683 (2020).

ArticleMATHGoogle Scholar

Joshi, B. S., de Beer, M. A., Giepmans, B. N. G. & Zuhorn, I. S. Endocytosis of extracellular vesicles and release of their cargo from endosomes. ACS Nano 14, 4444–4455 (2020).

ArticleGoogle Scholar

Votteler, J. et al. Designed proteins induce the formation of nanocage-containing extracellular vesicles. Nature 540, 292–295 (2016).

ArticleGoogle Scholar

Qin, W. et al. Dynamic mapping of proteome trafficking within and between living cells by TransitID. Cell 186, 3307–3324.e30 (2023).

ArticleMATHGoogle Scholar

Rennick, J. J., Johnston, A. P. R. & Parton, R. G. Key principles and methods for studying the endocytosis of biological and nanoparticle therapeutics. Nat. Nanotechnol. 16, 266–276 (2021).

ArticleMATHGoogle Scholar

Atai, N. A. et al. Heparin blocks transfer of extracellular vesicles between donor and recipient cells. J. Neurooncol. 115, 343–351 (2013).

ArticleMATHGoogle Scholar

Zhang, W. et al. ICAM-1-mediated adhesion is a prerequisite for exosome-induced T cell suppression. Dev. Cell 57, 329–343.e7 (2022).

ArticleMATHGoogle Scholar

Franciszkiewicz, K. et al. CD103 or LFA-1 engagement at the immune synapse between cytotoxic T cells and tumor cells promotes maturation and regulates T-cell effector functions. Cancer Res. 73, 617–628 (2013).

ArticleMATHGoogle Scholar

Nolte-‘t Hoen, E. N. M., Buschow, S. I., Anderton, S. M., Stoorvogel, W. & Wauben, M. H. M. Activated T cells recruit exosomes secreted by dendritic cells via LFA-1. Blood 113, 1977–1981 (2009).

ArticleGoogle Scholar

Zapatero-Belinchón, F. J., Carriquí-Madroñal, B. & Gerold, G. in Advances in Virus Research Vol. 109 (ed. Gerold, G.) 63–104 (Academic, 2021).

Lu, S. et al. Native and engineered extracellular vesicles for wound healing. Front. Bioeng. Biotechnol. 10, 1053217 (2022).

ArticleGoogle Scholar

de Abreu, R. C. et al. Native and bioengineered extracellular vesicles for cardiovascular therapeutics. Nat. Rev. Cardiol. 17, 685–697 (2020).

ArticleMATHGoogle Scholar

Zaborowski, M. P. et al. Membrane-bound Gaussia luciferase as a tool to track shedding of membrane proteins from the surface of extracellular vesicles. Sci. Rep. 9, 17387 (2019).

ArticleMATHGoogle Scholar

Ai, Y. et al. Endocytosis blocks the vesicular secretion of exosome marker proteins. Sci. Adv. 10, eadi9156 (2024).

ArticleGoogle Scholar

Thompson, A. G. et al. Extracellular vesicles in neurodegenerative disease — pathogenesis to biomarkers. Nat. Rev. Neurol. 12, 346–357 (2016).

ArticleMATHGoogle Scholar

Boulanger, C. M., Loyer, X., Rautou, P.-E. & Amabile, N. Extracellular vesicles in coronary artery disease. Nat. Rev. Cardiol. 14, 259–272 (2017).

ArticleGoogle Scholar

Grange, C. & Bussolati, B. Extracellular vesicles in kidney disease. Nat. Rev. Nephrol. 18, 499–513 (2022).

ArticleMATHGoogle Scholar

Buzas, E. I. The roles of extracellular vesicles in the immune system. Nat. Rev. Immunol. 23, 236–250 (2023).

ArticleMATHGoogle Scholar

O’Brien, K., Breyne, K., Ughetto, S., Laurent, L. C. & Breakefield, X. O. RNA delivery by extracellular vesicles in mammalian cells and its applications. Nat. Rev. Mol. Cell Biol. 21, 585–606 (2020).

ArticleGoogle Scholar

Wu, J. & Jaffrey, S. R. Imaging mRNA trafficking in living cells using fluorogenic proteins. Curr. Opin. Chem. Biol. 57, 177–183 (2020).

ArticleGoogle Scholar

Lu, X., Kong, K. Y. S. & Unrau, P. J. Harmonizing the growing fluorogenic RNA aptamer toolbox for RNA detection and imaging. Chem. Soc. Rev. 52, 4071–4098 (2023).

ArticleMATHGoogle Scholar

Bonacquisti, E. E. et al. Fluorogenic RNA-based biomaterials for imaging and tracking the cargo of extracellular vesicles. J. Control. Release 374, 349–368 (2024).

ArticleMATHGoogle Scholar

Lindenbergh, M. F. S. & Stoorvogel, W. Antigen presentation by extracellular vesicles from professional antigen-presenting cells. Annu. Rev. Immunol. 36, 435–459 (2018).

ArticleMATHGoogle Scholar

Shedden, K., Xie, X. T., Chandaroy, P., Chang, Y. T. & Rosania, G. R. Expulsion of small molecules in vesicles shed by cancer cells: association with gene expression and chemosensitivity profiles. Cancer Res. 63, 4331–4337 (2003).

MATHGoogle Scholar

Maugeri, M. et al. Linkage between endosomal escape of LNP-mRNA and loading into EVs for transport to other cells. Nat. Commun. 10, 4333 (2019).

ArticleMATHGoogle Scholar

Nawaz, M. et al. Lipid nanoparticles deliver the therapeutic VEGFA mRNA in vitro and in vivo and transform extracellular vesicles for their functional extensions. Adv. Sci. 10, e2206187 (2023).

ArticleMATHGoogle Scholar

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

ArticleMATHGoogle Scholar

Troyer, Z. et al. Extracellular vesicles carry SARS-CoV-2 spike protein and serve as decoys for neutralizing antibodies. J. Extracell. Vesicles 10, e12112 (2021).

ArticleGoogle Scholar

Zhao, F. et al. Extracellular vesicles from Zika virus-infected cells display viral E protein that binds ZIKV-neutralizing antibodies to prevent infection enhancement. EMBO J. 42, e112096 (2023).

ArticleGoogle Scholar

Gupta, D. et al. Amelioration of systemic inflammation via the display of two different decoy protein receptors on extracellular vesicles. Nat. Biomed. Eng. 5, 1084–1098 (2021).

ArticleMATHGoogle Scholar

Carotti, V., Rigalli, J. P., van Asbeck-van der Wijst, J. & Hoenderop, J. G. J. Interplay between purinergic signalling and extracellular vesicles in health and disease. Biochem. Pharmacol. 203, 115192 (2022).

ArticleGoogle Scholar

Angioni, R. et al. CD73+ extracellular vesicles inhibit angiogenesis through adenosine A2B receptor signalling. J. Extracell. Vesicles 9, 1757900 (2020).

ArticleGoogle Scholar

Sanderson, R. D., Bandari, S. K. & Vlodavsky, I. Proteases and glycosidases on the surface of exosomes: newly discovered mechanisms for extracellular remodeling. Matrix Biol. 75–76, 160–169 (2019).

ArticleGoogle Scholar

An, Y. et al. Tumor exosomal ENPP1 hydrolyzes cGAMP to inhibit cGAS-STING signaling. Adv. Sci. 11, e230811 (2024).

ArticleGoogle Scholar

Hansen, A. S. et al. T-cell derived extracellular vesicles prime macrophages for improved STING based cancer immunotherapy. J. Extracell. Vesicles 12, e12350 (2023).

ArticleGoogle Scholar

Roefs, M. T. et al. Cardiac progenitor cell-derived extracellular vesicles promote angiogenesis through both associated- and co-isolated proteins. Commun. Biol. 6, 800 (2023).

ArticleGoogle Scholar

Chen, G. et al. Exosomal PD-L1 contributes to immunosuppression and is associated with anti-PD-1 response. Nature 560, 382–386 (2018).

ArticleMATHGoogle Scholar

Poggio, M. et al. Suppression of exosomal PD-L1 induces systemic anti-tumor immunity and memory. Cell 177, 414–427.e13 (2019).

ArticleMATHGoogle Scholar

Zhong, W. et al. Tumor-derived small extracellular vesicles inhibit the efficacy of CAR T cells against solid tumors. Cancer Res. 83, 2790–2806 (2023).

ArticleMATHGoogle Scholar

Rausch, L. et al. Phosphatidylserine-positive extracellular vesicles boost effector CD8+ T cell responses during viral infection. Proc. Natl Acad. Sci. USA 120, e2210047120 (2023).

ArticleGoogle Scholar

Raposo, G. et al. B lymphocytes secrete antigen-presenting vesicles. J. Exp. Med. 183, 1161–1172 (1996).

ArticleGoogle Scholar

Blandin, A. et al. Extracellular vesicles are carriers of adiponectin with insulin-sensitizing and anti-inflammatory properties. Cell Rep. 42, 112866 (2023).

ArticleMATHGoogle Scholar

Jin, S. et al. Astroglial exosome HepaCAM signaling and ApoE antagonization coordinates early postnatal cortical pyramidal neuronal axon growth and dendritic spine formation. Nat. Commun. 14, 5150 (2023).

ArticleGoogle Scholar

Seras-Franzoso, J. et al. Extracellular vesicles from recombinant cell factories improve the activity and efficacy of enzymes defective in lysosomal storage disorders. J. Extracell. Vesicles 10, e12058 (2021).

ArticleGoogle Scholar

Harmati, M., Bukva, M., Böröczky, T., Buzás, K. & Gyukity-Sebestyén, E. The role of the metabolite cargo of extracellular vesicles in tumor progression. Cancer Metastasis Rev. 40, 1203–1221 (2021).

ArticleGoogle Scholar

Zhang, Y., Liang, F., Zhang, D., Qi, S. & Liu, Y. Metabolites as extracellular vesicle cargo in health, cancer, pleural effusion, and cardiovascular diseases: an emerging field of study to diagnostic and therapeutic purposes. Biomed. Pharmacother. 157, 114046 (2023).

ArticleGoogle Scholar

Pham, T. T. et al. Endocytosis of red blood cell extracellular vesicles by macrophages leads to cytoplasmic heme release and prevents foam cell formation in atherosclerosis. J. Extracell. Vesicles 12, e12354 (2023).

ArticleMATHGoogle Scholar

Liang, Y. et al. Cell-derived extracellular vesicles for CRISPR/Cas9 delivery: engineering strategies for cargo packaging and loading. Biomater. Sci. 10, 4095–4106 (2022).

ArticleMATHGoogle Scholar

Kostyushev, D. et al. Gene editing by extracellular vesicles. Int. J. Mol. Sci. 21, 7362 (2020).

ArticleGoogle Scholar

Valadi, H. et al. Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat. Cell Biol. 9, 654–659 (2007).

ArticleMATHGoogle Scholar

Zhang, C. et al. Intercellular mitochondrial component transfer triggers ischemic cardiac fibrosis. Sci. Bull. 68, 1784–1799 (2023).

ArticleMATHGoogle Scholar

Crewe, C. et al. Extracellular vesicle-based interorgan transport of mitochondria from energetically stressed adipocytes. Cell Metab. 33, 1853–1868.e11 (2021).

ArticleMATHGoogle Scholar

Liang, W. et al. Mitochondria are secreted in extracellular vesicles when lysosomal function is impaired. Nat. Commun. 14, 5031 (2023).

ArticleMATHGoogle Scholar

Borcherding, N. & Brestoff, J. R. The power and potential of mitochondria transfer. Nature 623, 283–291 (2023).

ArticleMATHGoogle Scholar

Li, X. et al. Extracellular vesicle–encapsulated adeno-associated viruses for therapeutic gene delivery to the heart. Circulation 148, 405–425 (2023).

ArticleMATHGoogle Scholar

Hirose, H., Hirai, Y., Sasaki, M., Sawa, H. & Futaki, S. Quantitative analysis of extracellular vesicle uptake and fusion with recipient cells. Bioconjug. Chem. 33, 1852–1859 (2022).

ArticleGoogle Scholar

Bui, S., Dancourt, J. & Lavieu, G. Virus-free method to control and enhance extracellular vesicle cargo loading and delivery. ACS Appl. Bio Mater. 6, 1081–1091 (2023).

ArticleMATHGoogle Scholar

Hamilton, J. R. et al. In vivo human T cell engineering with enveloped delivery vehicles. Nat. Biotechnol. 42, 1684–1692 (2024).

ArticleMATHGoogle Scholar

Hamilton, J. R. et al. Targeted delivery of CRISPR-Cas9 and transgenes enables complex immune cell engineering. Cell Rep. 35, 109207 (2021).

ArticleMATHGoogle Scholar

Segel, M. et al. Mammalian retrovirus-like protein PEG10 packages its own mRNA and can be pseudotyped for mRNA delivery. Science 373, 882–889 (2021).

ArticleGoogle Scholar

Download references

Acknowledgements

S.E.A. was funded by the European Research Council under the European Union’s Horizon 2020 research and innovation programme (EXPERT, grant agreement No. 825828), the European Research Council Consolidator Grant (DELIVER, grant agreement No. 101001374), the Swedish Foundation of Strategic Research (FormulaEx, grant agreement No. SM19-0007), the Swedish Cancer Society (project agreement No. 21-1762-Pj-01-H) and the Swedish Research Council (project agreement No. 4-258/2021). The authors acknowledge M. Mowoe for linguistic editing.

Author information

Authors and Affiliations

Division of Biomolecular and Cellular Medicine, Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Stockholm, Sweden

Wenyi Zheng, Samantha Roudi, Houze Zhou, Joel Z. Nordin & Samir EL Andaloussi

Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital Huddinge, Huddinge, Stockholm, Sweden

Wenyi Zheng, Samantha Roudi, Houze Zhou, Joel Z. Nordin & Samir EL Andaloussi

Karolinska ATMP Center, ANA Futura, Huddinge, Stockholm, Sweden

Wenyi Zheng, Samantha Roudi, Houze Zhou, Joel Z. Nordin & Samir EL Andaloussi

Angers Cancer and Immunology Research Center, Nantes University, Nantes, France

Maribel Lara Corona & Guillaume van Niel

Authors

Wenyi Zheng

View author publications

You can also search for this author in PubMedGoogle Scholar

2. Samantha Roudi

View author publications

You can also search for this author in PubMedGoogle Scholar

3. Houze Zhou

View author publications

You can also search for this author in PubMedGoogle Scholar

4. Maribel Lara Corona

View author publications

You can also search for this author in PubMedGoogle Scholar

5. Guillaume van Niel

View author publications

You can also search for this author in PubMedGoogle Scholar

6. Joel Z. Nordin

View author publications

You can also search for this author in PubMedGoogle Scholar

7. Samir EL Andaloussi

View author publications

You can also search for this author in PubMedGoogle Scholar

Contributions

W.Z. researched data for the draft and contributed to the writing and editing of this manuscript. S.R. and H.Z. contributed to the discussion of content, writing and editing of this manuscript. M.L.C. and G.v.N. contributed to reviewing and editing of the manuscript before submission. J.Z.N. made a substantial contribution to the discussion of content. S.E.A. contributed to the discussion and reviewed the manuscript before submission.

Corresponding author

Correspondence to Samir EL Andaloussi.

Ethics declarations

Competing interests

J.Z.N. and S.E.A. are consultants for and have equity interests in EVOX Therapeutics, Oxford, UK. The other authors declare no competing interests.

Peer review

Peer review information

Nature Reviews Bioengineering thanks Yongjie Yang, Dima Ter-Ovanesyan, who co-reviewed with David Walt, 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.

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

Zheng, W., Roudi, S., Zhou, H. et al. Genetic tools for investigating the life cycle of extracellular vesicles. Nat Rev Bioeng (2025). https://doi.org/10.1038/s44222-025-00286-6

Download citation

Accepted:03 February 2025

Published:17 March 2025

DOI:https://doi.org/10.1038/s44222-025-00286-6

Share this article

Anyone you share the following link with will be able to read this content:

Get shareable link

Sorry, a shareable link is not currently available for this article.

Copy to clipboard

Provided by the Springer Nature SharedIt content-sharing initiative

Read full news in source page