Extracellular vesicle in vivo biodistribution is determined by cell source, route of administration and targeting

Oscar P B Wiklander, Joel Z Nordin, Aisling O'Loughlin, Ylva Gustafsson, Giulia Corso, Imre Mäger, Pieter Vader, Yi Lee, Helena Sork, Yiqi Seow, Nina Heldring, Lydia Alvarez-Erviti, C I Edvard Smith, Katarina Le Blanc, Paolo Macchiarini, Philipp Jungebluth, Matthew J A Wood, Samir El Andaloussi, Oscar P B Wiklander, Joel Z Nordin, Aisling O'Loughlin, Ylva Gustafsson, Giulia Corso, Imre Mäger, Pieter Vader, Yi Lee, Helena Sork, Yiqi Seow, Nina Heldring, Lydia Alvarez-Erviti, C I Edvard Smith, Katarina Le Blanc, Paolo Macchiarini, Philipp Jungebluth, Matthew J A Wood, Samir El Andaloussi

Abstract

Extracellular vesicles (EVs) have emerged as important mediators of intercellular communication in a diverse range of biological processes. For future therapeutic applications and for EV biology research in general, understanding the in vivo fate of EVs is of utmost importance. Here we studied biodistribution of EVs in mice after systemic delivery. EVs were isolated from 3 different mouse cell sources, including dendritic cells (DCs) derived from bone marrow, and labelled with a near-infrared lipophilic dye. Xenotransplantation of EVs was further carried out for cross-species comparison. The reliability of the labelling technique was confirmed by sucrose gradient fractionation, organ perfusion and further supported by immunohistochemical staining using CD63-EGFP probed vesicles. While vesicles accumulated mainly in liver, spleen, gastrointestinal tract and lungs, differences related to EV cell origin were detected. EVs accumulated in the tumour tissue of tumour-bearing mice and, after introduction of the rabies virus glycoprotein-targeting moiety, they were found more readily in acetylcholine-receptor-rich organs. In addition, the route of administration and the dose of injected EVs influenced the biodistribution pattern. This is the first extensive biodistribution investigation of EVs comparing the impact of several different variables, the results of which have implications for the design and feasibility of therapeutic studies using EVs.

Keywords: biodistribution; drug delivery; exosomes; extracellular vesicles; microvesicles; nanotechnology; tissue targeting.

Figures

Fig. 1
Fig. 1
DiR labelling does not affect EV morphology and co-localizes with EVs. (A) Western Blot (WB) stained for Alix and Tsg101 on HEK293T purified, DiR-labelled EVs and cell lysate (CL) (whole WB-membrane shown in Supplementary Fig. 1A). (B) NTA profile of DiR-labelled HEK293T EVs showing an EV-like distribution with a mode size around 100 nm. The curve has been normalized to dilution factors. (C) Electron microscopy picture of DiR-labelled HEK-293T EVs. Scale bar represents 100 nm. (D) Image depicting DiR-EVs and free DiR sucrose gradients in UC tubes and subsequent 12-well plates containing the different fractions. (E) Fluorescent values (normalized to background) of the DiR-EVs and free DiR gradients (assayed from the 12-well plates depicted in 1D). (F) WB stained for Alix for DiR-EVs from fractions 2–10 (whole WB-membrane shown in Supplementary Fig. 1B).
Fig. 2
Fig. 2
Systemically delivered EVs display a dose-dependent increase in tissue uptake in mice. (A) Representative IVIS image 24 hours post-injection of a live mouse injected i.v. with 1.0×1010 p/g DiR-EVs (left) and PBS-treated control (right). (B) Representative co-registration of FLIT with CT-scan 24 hours post-injection of a mouse injected i.v. with 1.0×1010 p/g DiR-EVs. (C) Fluorescent signal in each organ 24 hours post-iv injection of HEK293T-EVs (1.0×1010 p/g) with (HEK293T-p) and without (HEK293T) perfusion and free OptiMEM-DiR. Misc.=brain, heart, kidneys and quadriceps muscle. N=4. The results represent mean+SEM. (D) Representative IVIS images of organs (24 hours post-injection) from mice injected i.v. with 1.5×1010 p/g, 1.0×1010 p/g and 0.25×1010 p/g of HEK293T EVs. (E) The percentage of fluorescent signal in each organ for the same experiment as in Fig. 2D (Absolute fluorescent signals are plotted in Supplementary Fig. 3). N=4, in E *represents p=0.05, **p=0.001 and ****p=0.0001. The results represent mean+SEM.
Fig. 3
Fig. 3
Distribution kinetics and effect of different injection routes of DiR-labelled EVs on tissue distribution. (A) Percentage fluorescent signal of each organ ex vivo at different time points after injection of 1.0×1010 p/g DiR-labelled HEK293T EVs (fluorescent signal in absolute numbers in Supplementary Fig. 4). (B) Percentage fluorescent signal in each organ following i.p., s.c. and i.v. injection of 1.0×1010 p/g DiR-labelled HEK293T EVs (Absolute fluorescent signals are plotted in Supplementary Fig. 5b). Misc.=brain, heart, kidneys and quadriceps muscle. N=3–4, **represents p<0.01, ***p<0.001 and ****p<0.0001. The results represent mean+SEM.
Fig. 4
Fig. 4
The biodistribution of EVs differs between cell sources. (A) Particle distribution of DiR-labelled EVs from mouse C2C12-, B16F10- and DC-cells measured by NTA. (B) Western Blot stained against Alix and Tsg101 of EVs from C2C12-, B16F10- and DC-cells (whole WB-membranes in Supplementary Fig. 7). (C) The percentage of fluorescent signal in each organ after injection of 1.0×1010 p/g DiR-labelled EVs from the 3 different mouse cell types (Absolute fluorescent signals are plotted in Supplementary Fig. 8). Misc.=brain, heart, kidneys and quadriceps muscle. (D) Representative spleen, liver and lung IVIS images from mice injected with EVs from the 3 different mouse cell sources and PBS-treated control. N=4, in C *represents p=0.05, ***p=0.001 and ****p=0.0001. The results represent mean+SEM.
Fig. 5
Fig. 5
EVs distribute to tumour tissue and can be targeted to the brain. (A) The percentage of fluorescent signal in each organ of tumour-bearing mice after injection of 1.0×1010 p/g HEK293T DiR-labelled EVs (Absolute fluorescent signals are plotted in Supplementary Fig. 9). (B) Fluorescent signal from tumour, kidneys and brain shown from the same mice described in Fig. 5A. (C) NTA of RVG-positive (+RVG) and RVG-negative (-RVG) DC-EVs. (D) The percentage of fluorescent signal in each organ after injection of 1.0×1010 p/g of +RVG and –RVG DC-EVs (Absolute fluorescent signals are plotted in Supplementary Fig. 10). (E) Representative image of mouse brains, harvested 24 hour post-injection of 1.0×1010 p/g of +RVG and –RVG DC-EVs. (F) The percentage of fluorescence signal in the brain, heart and muscle from the same mice as shown in Fig. 5D (Absolute fluorescent signals are plotted in Supplementary Fig. 11). Misc.=brain, heart, kidneys and quadriceps muscle. N=4, **represents p<0.01 and ***represents p<0.001, the results represent mean+SEM.

References

    1. EL Andaloussi S, Mager I, Breakefield XO, Wood MJ. Extracellular vesicles: biology and emerging therapeutic opportunities. Nat Rev Drug Discov. 2013;12:347–57.
    1. Ratajczak J, Miekus K, Kucia M, Zhang J, Reca R, Dvorak P, et al. Embryonic stem cell-derived microvesicles reprogram hematopoietic progenitors: evidence for horizontal transfer of mRNA and protein delivery. Leukemia. 2006;20:847–56.
    1. Peinado H, Aleckovic M, Lavotshkin S, Matei I, Costa-Silva B, Moreno-Bueno G, et al. Melanoma exosomes educate bone marrow progenitor cells toward a pro-metastatic phenotype through MET. Nat Med. 2012;18:883–91.
    1. Al-Nedawi K, Meehan B, Micallef J, Lhotak V, May L, Guha A, et al. Intercellular transfer of the oncogenic receptor EGFRvIII by microvesicles derived from tumour cells. Nat Cell Biol. 2008;10:619–24.
    1. Valadi H, Ekstrom K, Bossios A, Sjostrand M, Lee JJ, Lotvall JO. Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat Cell Biol. 2007;9:654–9.
    1. Keller S, Ridinger J, Rupp AK, Janssen JW, Altevogt P. Body fluid derived exosomes as a novel template for clinical diagnostics. J Transl Med. 2011;9:86.
    1. Street JM, Barran PE, Mackay CL, Weidt S, Balmforth C, Walsh TS, et al. Identification and proteomic profiling of exosomes in human cerebrospinal fluid. J Transl Med. 2012;10:5.
    1. Lasser C, Alikhani VS, Ekstrom K, Eldh M, Paredes PT, Bossios A, et al. Human saliva, plasma and breast milk exosomes contain RNA: uptake by macrophages. J Transl Med. 2011;9:9.
    1. Pisitkun T, Shen RF, Knepper MA. Identification and proteomic profiling of exosomes in human urine. Proc Natl Acad Sci USA. 2004;101:13368–73.
    1. Yamada T, Inoshima Y, Matsuda T, Ishiguro N. Comparison of methods for isolating exosomes from bovine milk. J Vet Med Sci. 2012;74:1523–5.
    1. Bellingham SA, Guo BB, Coleman BM, Hill AF. Exosomes: vehicles for the transfer of toxic proteins associated with neurodegenerative diseases? Front Physiol. 2012;3:124.
    1. Emmanouilidou E, Melachroinou K, Roumeliotis T, Garbis SD, Ntzouni M, Margaritis LH, et al. Cell-produced alpha-synuclein is secreted in a calcium-dependent manner by exosomes and impacts neuronal survival. J Neurosci. 2010;30:6838–51.
    1. Lai RC, Chen TS, Lim SK. Mesenchymal stem cell exosome: a novel stem cell-based therapy for cardiovascular disease. Regen Med. 2011;6:481–92.
    1. Timmers L, Lim SK, Hoefer IE, Arslan F, Lai RC, Van Oorschot AA, et al. Human mesenchymal stem cell-conditioned medium improves cardiac function following myocardial infarction. Stem Cell Res. 2011;6:206–14.
    1. Gatti S, Bruno S, Deregibus MC, Sordi A, Cantaluppi V, Tetta C, et al. Microvesicles derived from human adult mesenchymal stem cells protect against ischaemia-reperfusion-induced acute and chronic kidney injury. Nephrol Dial Transplant. 2011;26:1474–83.
    1. Zitvogel L, Regnault A, Lozier A, Wolfers J, Flament C, Tenza D, et al. Eradication of established murine tumors using a novel cell-free vaccine: dendritic cell-derived exosomes. Nat Med. 1998;4:594–600.
    1. Alvarez-Erviti L, Seow Y, Yin H, Betts C, Lakhal S, Wood MJ. Delivery of siRNA to the mouse brain by systemic injection of targeted exosomes. Nat Biotechnol. 2011;29:341–5.
    1. Kordelas L, Rebmann V, Ludwig AK, Radtke S, Ruesing J, Doeppner TR, et al. MSC-derived exosomes: a novel tool to treat therapy-refractory graft-versus-host disease. Leukemia. 2014;28:970–3.
    1. Grange C, Tapparo M, Bruno S, Chatterjee D, Quesenberry PJ, Tetta C, et al. Biodistribution of mesenchymal stem cell-derived extracellular vesicles in a model of acute kidney injury monitored by optical imaging. Int J Mol Med. 2014;33:1055–63.
    1. Jang SC, Kim SR, Yoon YJ, Park KS, Kim JH, Lee J, et al. In vivo kinetic biodistribution of nano-sized outer membrane vesicles derived from bacteria. Small. 2014;11:456–61.
    1. Lai CP, Mardini O, Ericsson M, Prabhakar S, Maguire CA, Chen JW, et al. Dynamic biodistribution of extracellular vesicles in vivo using a multimodal imaging reporter. ACS Nano. 2014;8:483–94.
    1. Takahashi Y, Nishikawa M, Shinotsuka H, Matsui Y, Ohara S, Imai T, et al. Visualization and in vivo tracking of the exosomes of murine melanoma B16-BL6 cells in mice after intravenous injection. J Biotechnol. 2013;165:77–84.
    1. Ignowski JM, Schaffer DV. Kinetic analysis and modeling of firefly luciferase as a quantitative reporter gene in live mammalian cells. Biotechnol Bioeng. 2004;86:827–34.
    1. Smyth T, Kullberg M, Malik N, Smith-Jones P, Graner MW, Anchordoquy TJ. Biodistribution and delivery efficiency of unmodified tumor-derived exosomes. J Control Release. 2014;199C:145–55.
    1. Ohno S, Takanashi M, Sudo K, Ueda S, Ishikawa A, Matsuyama N, et al. Systemically injected exosomes targeted to EGFR deliver antitumor microRNA to breast cancer cells. Mol Ther. 2013;21:185–91.
    1. Tian Y, Li S, Song J, Ji T, Zhu M, Anderson GJ, et al. A doxorubicin delivery platform using engineered natural membrane vesicle exosomes for targeted tumor therapy. Biomaterials. 2014;35:2383–90.
    1. Le Blanc K, Frassoni F, Ball L, Locatelli F, Roelofs H, Lewis I, et al. Mesenchymal stem cells for treatment of steroid-resistant, severe, acute graft-versus-host disease: a phase II study. Lancet. 2008;371:1579–86.
    1. Le Blanc K, Samuelsson H, Gustafsson B, Remberger M, Sundberg B, Arvidson J, et al. Transplantation of mesenchymal stem cells to enhance engraftment of hematopoietic stem cells. Leukemia. 2007;21:1733–8.
    1. Dragovic RA, Gardiner C, Brooks AS, Tannetta DS, Ferguson DJ, Hole P, et al. Sizing and phenotyping of cellular vesicles using Nanoparticle tracking analysis. Nanomedicine. 2011;7:780–8.
    1. Niska JA, Meganck JA, Pribaz JR, Shahbazian JH, Lim E, Zhang N, et al. Monitoring bacterial burden, inflammation and bone damage longitudinally using optical and muCT imaging in an orthopaedic implant infection in mice. PLoS One. 2012;7:e47397.
    1. Patel AR, Lim E, Francis KP, Singh M. Opening up the optical imaging window using nano-luciferin. Pharm Res. 2014;31:3073–84.
    1. Herschman HR. Molecular imaging: looking at problems, seeing solutions. Science. 2003;302:605–8.
    1. Hilderbrand SA, Weissleder R. Near-infrared fluorescence: application to in vivo molecular imaging. Curr Opin Chem Biol. 2010;14:71–9.
    1. Luo S, Zhang E, Su Y, Cheng T, Shi C. A review of NIR dyes in cancer targeting and imaging. Biomaterials. 2011;32:7127–38.
    1. Ntziachristos V, Bremer C, Weissleder R. Fluorescence imaging with near-infrared light: new technological advances that enable in vivo molecular imaging. Eur Radiol. 2003;13:195–208.
    1. Kalchenko V, Shivtiel S, Malina V, Lapid K, Haramati S, Lapidot T, et al. Use of lipophilic near-infrared dye in whole-body optical imaging of hematopoietic cell homing. J Biomed Optics. 2006;11:050507.
    1. Mathivanan S, Simpson RJ. ExoCarta: a compendium of exosomal proteins and RNA. Proteomics. 2009;9:4997–5000.
    1. Lopez-Berestein G, Kasi L, Rosenblum MG, Haynie T, Jahns M, Glenn H, et al. Clinical pharmacology of 99mTc-labeled liposomes in patients with cancer. Cancer Res. 1984;44:375–8.
    1. Rank A, Nieuwland R, Crispin A, Grutzner S, Iberer M, Toth B, et al. Clearance of platelet microparticles in vivo. Platelets. 2011;22:111–16.
    1. Ostro MJ, Cullis PR. Use of liposomes as injectable-drug delivery systems. Am J Hosp Pharm. 1989;46:1576–87.
    1. Liu D, Mori A, Huang L. Role of liposome size and RES blockade in controlling biodistribution and tumor uptake of GM1-containing liposomes. Biochim Biophys Acta. 1992;1104:95–101.
    1. Moghimi SM, Hunter AC, Murray JC. Long-circulating and target-specific nanoparticles: theory to practice. Pharmacol Rev. 2001;53:283–318.
    1. Sarin H. Physiologic upper limits of pore size of different blood capillary types and another perspective on the dual pore theory of microvascular permeability. J Angiogenesis Res. 2010;2:14.
    1. Garnett MC, Kallinteri P. Nanomedicines and nanotoxicology: some physiological principles. Occup Med. 2006;56:307–11.
    1. Kupiec-Weglinski JW, Austyn JM, Morris PJ. Migration patterns of dendritic cells in the mouse. Traffic from the blood, and T cell-dependent and -independent entry to lymphoid tissues. J Exp Med. 1988;167:632–45.
    1. Rana S, Yue S, Stadel D, Zoller M. Toward tailored exosomes: the exosomal tetraspanin web contributes to target cell selection. Int J Biochem Cell Biol. 2012;44:1574–84.
    1. Rana S, Zoller M. Exosome target cell selection and the importance of exosomal tetraspanins: a hypothesis. Biochem Soc Trans. 2011;39:559–62.
    1. Bae YH, Park K. Targeted drug delivery to tumors: myths, reality and possibility. J Control Release. 2011;153:198–205.
    1. Kumar P, Wu H, McBride JL, Jung KE, Kim MH, Davidson BL, et al. Transvascular delivery of small interfering RNA to the central nervous system. Nature. 2007;448:39–43.
    1. Mazarakis ND, Azzouz M, Rohll JB, Ellard FM, Wilkes FJ, Olsen AL, et al. Rabies virus glycoprotein pseudotyping of lentiviral vectors enables retrograde axonal transport and access to the nervous system after peripheral delivery. Hum Mol Genet. 2001;10:2109–21.
    1. Pulford B, Reim N, Bell A, Veatch J, Forster G, Bender H, et al. Liposome-siRNA-peptide complexes cross the blood–brain barrier and significantly decrease PrP on neuronal cells and PrP in infected cell cultures. PLoS One. 2010;5:e11085.
    1. Boksa P, Quirion R. [3H]N-methyl-carbamylcholine, a new radioligand specific for nicotinic acetylcholine receptors in brain. Eur J Pharmacol. 1987;139:323–33.
    1. Lentz TL, Burrage TG, Smith AL, Crick J, Tignor GH. Is the acetylcholine receptor a rabies virus receptor? Science. 1982;215:182–4.
    1. Lentz TL, Burrage TG, Smith AL, Tignor GH. The acetylcholine receptor as a cellular receptor for rabies virus. Yale J Biol Med. 1983;56:315–22.
    1. Peralta EG, Ashkenazi A, Winslow JW, Smith DH, Ramachandran J, Capon DJ. Distinct primary structures, ligand-binding properties and tissue-specific expression of four human muscarinic acetylcholine receptors. EMBO J. 1987;6:3923–9.

Source: PubMed

3
Suscribir