Blood collection and processing
Blood samples were provided by the Institutional Transfusion Center of Ospedale Policlinico San Martino, Genova. Blood was collected from healthy donors in K2E EDTA tubes. Plasma was isolated from blood by serial centrifugation steps, performed at 4°C: 120xg 20 min, 120xg 5 min, 360xg 20 min and 3000xg 10 min. The resulting plasma was stored at –80 °C until EV separation.
EV isolation
Three different EV isolation protocols were tested.
Size exclusion chromatography (SEC) was performed using IZON smart columns qEV/70. According to manufacturer’s instructions, 500 µL of plasma was loaded on the column and eluted with PBS containing 2mM EDTA (PBS-EDTA) to overcome EV aggregation. Twenty eluted fractions (F) of 500 μL were collected in protein low binding tubes, and EVs were collected in fractions 7-9. The elution profile of both EVs and contaminating proteins has been assessed measuring the absorbance at 280nm. Samples were further diluted depending on subsequent analysis. The EV-enriched fractions have been rinsed in dPBS and centrifuged at 100 000×g for 2h with a SN985 swinging rotor (Optima TL, Beckman).
Regarding the combination of iodixanol density gradient and SEC (IDC+SEC) method, we followed a previously described protocol [15]. Briefly, 6 mL of plasma was layered on top of a 2 mL 50%, 2 mL 30%, and 2 mL 10% OptiPrep layer. Cushion and sample were centrifuged at 178.000×g for 2 h at 4 °C in a SW41It swinging rotor. The visible band between the 10% and 30% layers was collected (high-density band, 1.06-1.16g/cm³; EV enriched band) and loaded onto a SEC column. The low-density band (< 1,025g/cm³) on the top of the tube, enriched in lipoproteins, was discarded. The EV enriched fractions have been further centrifuged at 100.000×g for 2h with a SN985 swinging rotor (Optima TL, Beckman).
The sucrose cushion ultracentrifugation (sUC) was performed as previously reported [16]. Either 1 mL or 2.3 mL (the latter amount has been modified from the original protocol) of plasma were diluted in 0.22 mm filtered dPBS to a final volume of 9.5mL, carefully loaded on the top of 2mL 20% sucrose and centrifuged at 100 000×g for 135 minutes in a SW41Ti swinging rotor. Then, the supernatant was discarded and the EV pellet was resuspended in 1ml of PBS-EDTA, centrifuged at 3000×g for 10 minutes, and the supernatant has been further centrifuged at 100 000×g for 2h with a SN985 swinging rotor (Optima TL, Beckman).
EV quantification by Nanoparticle Tracking Analysis (NTA)
EV size distribution and concentration were analyzed by ZetaView® TWIN-NTA PMX-220 (Particle Metrix GmbH, Inning am Ammersee, Germany), equipped with a sample cell and two lasers (488 nm and 640 nm) and Zetaview 8.05.14_SP7 software. Briefly, after calibration with 100 nm polystyrene beads, samples were diluted in filtered dPBS and injected into the sample cell using a 1 mL syringe. For both size and concentration measurements, the 488 nm laser in scatter mode was used; for the fluorescent NTA, 640 nm laser with a 660 nm filter wavelength was used. Size distribution analyses of 11 different positions were performed for each sample and the reading parameters were set as 80% sensitivity, and shutter speed of 100, minimum brightness of 20. For fluorescent NTA, sensitivity was increased to 90%, minimum brightness of 30.
ELISA
EV-enriched samples were diluted in dPBS (100,000-times for measurements of ApoA1 in all samples, 3000-times for measurements of ApoB100 in samples after SEC and 80-times for measurements of ApoB100 in samples after sUC) and the concentration (ng/mL) of
ApoA1 and ApoB100 measured in duplicates by specific ELISAs (ELISA-ApoA1: #3710-1HP-2, ELISA-ApoB100: #3715-1HP-2, Mabtech, Sweden), according to the manufacturer’s instructions. Measured concentrations were normalized to 1 mL of starting plasma and any potential dilution of the samples was accounted for.
EV characterization by flow cytometry
EVs were characterized by non-conventional flow cytometry, as previously described[17]. Briefly, 1 ×108 EVs in 100µL of PBS-EDTA were stained with 1mM CFDA-SE (Vybrant™ CFDA-SE Cell Tracer Kit, Thermo Fisher Scientific, Waltham, MA, USA) at either 4°C or room temperature (RT). The expression of CD9 (APC Mouse Anti-Human CD9, Clone HI9a, 312108; BioLegend, San Diego, CA, USA), CD63 (PE-Cy7 Mouse Anti-Human CD63, Clone H5C6, 561982; BD Biosciences, San Jose, CA, USA ), and CD81 (BV421 Mouse Anti-Human CD81, Clone JS-81, 740079; BD Biosciences ) was evaluated within the CFDA-SE-positive events and compared to the corresponding isotype controls using CytoFLEX S (Beckman Coulter). FlowJo software was used to analyze the data.
Western blot
EVs were resuspended in RIPA buffer (1% NONIDET p-40, 0.1% SDS, 0.1% Sodium deoxycholate, protease inhibitor cocktail 1x, in PBS pH7.5) and protein content was quantified by Bicinchoninic acid (BCA) assay (Thermo Fisher Scientific). Five μg of proteins for each sample were loaded on 4%–12% NuPAGE Bis–Tris gel (Thermo Fisher Scientific). Electrophoresis was performed at 160 V and proteins were blotted on a polyvinylidene fluoride membrane. After blocking nonspecific sites with 5% non-fat dry milk (EuroClone, Italy) in Tris Buffered Saline with Tween 20 (TTBS, 20 mM Tris pH 7.5, 500 mM NaCl, 0.05% Tween 20), the membrane was incubated overnight at 4 °C with specific primary antibodies for: Flotillin-1 (Recombinant Anti-Flotillin 1 antibody [EPR6041] (ab133497), 1:10 000, Abcam), Syntenin (Recombinant Anti-Syntenin antibody [EPR8102] (ab133267), 1:1000, Abcam), Alix (Recombinant Anti-ALIX antibody [EPR15314] (ab186429), 1:1000, Abcam), TSG-101 (Anti-TSG101 antibody produced in rabbit, 1:1000 dilution, T5701, SIgma) diluted in 2.5% non-fat dry milk/TTBS. After washing three times with TTBS, membranes were incubated with specific HRP-conjugated secondary antibodies anti-mouse (Mouse IgG HRP Linked Whole Ab, GENA931) or anti-rabbit (Rabbit IgG HRP Linked Whole Ab, GENA934). Positivity was highlighted by providing the substrates for the chemiluminescence reaction of HRP (Amersham ECL Prime Western Blotting Detection Reagent, GE Healthcare, Chicago, Illinois, USA) and impressing a photographic sheet by autoradiography (GE Healthcare).
Super resolution microscopy
Super-resolution microscopy analysis of plasma-EVs was performed using Nanoimager S Mark II microscope from ONI (Oxford Nanoimaging, Oxford, UK) equipped with a 100x, 1.4NA oil immersion objective, an XYZ closed-loop piezo 736 stage, and triple emission channels split at 640, 488 and 555 nm. The EV profiler Kit (ONI) was utilized for the experiments following manufacturer’s protocol. The Kit is composed by fluorescent antibodies anti CD9-488, CD63-568 and CD81-647 and all the buffer and reagent necessary for the experiment. dSTORM mode acquired sequentially in total reflection fluorescence (TIRF) mode was used for the acquisition of images. Single-molecule data was filtered using NimOS software (v.1.18.3, ONI). Data has been processed with the Collaborative Discovery (CODI) online analysis platform www.alto.codi.bio from ONI and the drift correction pipeline version 0.2.3 was used [46].
Transmission Electron Microscopy (TEM)
EVs were resuspended in 20 μl dPBS and fixed by adding an equal volume of 2% paraformaldehyde in 0.1 mol/l phosphate buffer (pH 7.4), as previously described [47]. EVs were then adsorbed for 10 min to formvar-carbon coated copper grids by floating the grids on 5 μl drops on parafilm. Subsequently, grids with adhered vesicles were rinsed in PBS and negatively stained with 2% uranyl acetate for 5 min at room temperature. Stained grids were embedded in 2.5% methylcellulose for improved preservation and air dried before examination. Electron micrographs were taken at Hitachi TEM microscope (HT7800 series, Tokyo, Japan) equipped with Megaview 3 digital camera and Radius software (EMSIS, Germany). To visualize EV size distribution, the results were plotted as colorblind safe scatter dot plot in which each size measured is represented as a point along with lines for the median value and the range.
EV surface functionalization by copper-free click chemistry
EVs (from 106 to 5x109 for the titration experiments, and in a range between 109 to 1010 for all the other experiments) were resuspended in 200µL of PBS-EDTA and mixed for 1h at RT with 1μg of DBCO-NHS ester (Dibenzocyclooctyne-N-hydroxysuccinimidyl ester for copper free click chemistry, 761524, Sigma). After the first incubation, 1.9μl of a 5.9mM AlexaFluor647- azide (AZ-647) solution (Alexa Fluor™ 647 Azide, Triethylammonium Salt, A10277, ThermoFisher) was added to the reaction and incubated for 4h at RT (Click-EVs). The fluorescent azide mixed with the EV suspension (without DBCO-NHS ester) was considered as negative control (EV + AZ-647). After the last incubation, samples were washed by SEC to remove unlabeled chemicals, stained with CFDA-SE, and analyzed by flow cytometry to evaluate, within the CFDA-SE positive events, the AF647 fluorescence signal associated with the azide. For uptake experiments, Click-EVs and the negative control were concentrated by ultracentrifugation (3h, 100 000×g, 4°C) and resuspended in 100µL of phenol red-free Dulbecco's modified eagle medium (DMEM).
EV internalization
The triple negative human breast cancer cell line MDA-MB 231 has been selected as responder cell line to evaluate the internalization capacity of functionalized EVs. MDA MB 231 were seeded in a 24-multiwell plate (25.000 cells/well) and cultured in DMEM supplemented with 10% Fetal Bovine Serum (FBS) (Gibco, Milan, Italy), 2 mM L-glutamine, and 50 mg/ml penicillin/streptomycin (complete medium) for 1 day, to let them adhere to the plastic support. Cells were then stimulated as follows: i) EVs+AZ647 (negative control, 100µL of sample + 100µL of phenol red-free 2X complete medium); ii) EVs+DBCO+AZ647 (Click-EVs, test group, 100µL of sample + 100µL of red-phenol red-free 2X complete medium). Untreated cells were used as additional negative control.
After 18h of incubation with either EVs+AZ647 or Click-EVs, cells were detached by Trypsin-EDTA (Gibco), rinsed in sterile PBS, centrifuged, resuspended in 300µL of PBS and analyzed by flow cytometry to evaluate the percentage of AF647 positive cells.
EV internalization was evaluated also by confocal microscopy. Cells were seeded on a glass slide fitting in a 24-multiwell plate (40.000 cells/well) and cultured in a complete medium for 1 day, to let them adhere to the glass support. Cells were then stimulated as above described for different time points (3h, 6h, 10h, 18h), rinsed 3 times with PBS, fixed with PFA 4% for 5 minutes at RT and permeabilized with 0,1% (v/v) Triton X-100 for 5 minutes at RT. Anti-HLA-I (supernatant of the clone W6.32, IgG2a) and anti-Rab5 (Abcam, Recombinant Anti-Rab5 antibody [EPR21801] - Early Endosome Marker (ab218624), 1:500) antibodies were incubated for 1h at RT to stain cell plasma membrane and early endosomes, respectively. Cells were then rinsed 3 times with PBS and incubated with the respective secondary antibodies (Goat anti-Mouse Alexa Fluor 546 and Goat anti-Rabbit Alexa Fluor 488) for 1h at RT and counterstained with DAPI for 20 minutes at RT. Samples were mounted on a glass slide with a water-based mounting medium (Permafluor, Thermo Fisher Scientific) and fluorescence imaged with the TCS SP2-AOBS laser scanning confocal microscope (Leica, Mannheim, Germany) through a plan apochromatic oil immersion objective 40x (1.4 NA). The 488 nm argon laser line was used for Rab5 fluorescence excitation, while the 546 nm and the 633 nm He-Ne lasers were utilized for excitation of HLA class I and Click-EVs fluorescence, respectively. DAPI was excited by a 405 nm diode laser. Leica software was used for image acquisition and analysis.
Statistical Analysis
The data were analyzed using unpaired t-test (to compare the percentage of AlexaFluor-647 positive EVs in negative control and Click-EVs), Ordinary one-way ANOVA (to evaluate the lipoprotein contaminants, to evaluate EV characterization by flow cytometry and super resolution microscopy, to compare the EV concentration measured by NTA, and to evaluate the EV internalization by flow cytometry) and Mann-Whitney Test (to evaluate the differences in terms of EV size by TEM and EV internalization at different time points by confocal microscopy). Data are presented as mean ± SD considering at least three independent replicates for each assay and analyzed by GraphPad Prism (Graph Pad Software, Inc.). For all analyses p < 0.05 was considered statistically significant. In all cases: ****p < 0.0001, ***p < 0.001, **p < 0.01, *p < 0.05.