Isolation, purification and characterization of extracellular vesicles
Mesenchymal stem cells (MSCs), human embryonic kidney cells (HEK293T), human ovarian cancer cells (SKOVE-3), B-lymphoma cells (B16-F10) and HeLa cells were used to isolate EVs. All the cells were purchased from ATCC. Cells were grown in DMEM GlutaMax medium (ThermoFisher Scientific, UK), containing 10% fetal bovine serum and 1 × penicillin-streptomycin (PS, 100 × contains 5000 U/ml of penicillin and 5000 µg/ml streptomycin; Sigma). MSCs were grown in RPMI GlutaMax media (ThermoFisher Scientific) with 10% FBS and 1 × PS (Sigma). Cells were maintained at 37 °C with 5% CO2. For isolation of EVs, cells were seeded in 150 × 20 mm dishes (Star Labs, UK) at 106 cells per dish in DMEM + 10% FBS media. Twenty four hours after seeding, the media of the cells was replaced with Opti-MEM reduced serum media (ThermoFisher Scientific, UK). Cells were incubated further for 48 h followed by collection of media in 50 ml falcon tubes (Sigma). To remove dead and floating cells, the media was centrifuged at 500 × g for 5 min. The supernatant was gently transferred into a fresh tube and centrifuged at 3000 × g for 20 min at 4 °C to pellet cell fragments and remaining cell debris. Purified media was concentrated by tangential ultrafiltration (TFF), using 100 kDa Vivaflow 50 R cartridges (Sartorius UK limited). Concentrated media was centrifuged at 10,000 × g for 30 min to remove aggregates and larger particles. The volume of media was further reduced to 2 ml, using Amicon ultra-15 centrifugal filter units of 100 kDa MWCO (Millipore). Finally, the media was passed through a Sepharose 4 fast flow gel-filtration column (GE Healthcare, 170149001), using the ÄKTA pure chromatography system (GE Healthcare). Purified EVs eluted from the column were used for the various biological assays. Typically, 500 ml of cell-culture media was used at the very beginning of the process and concentrated to 2 ml before loading to the column.
For animal experiments, large quantities of EVs were isolated using a hollow fibre bioreactor (FibreCell System, UK). 5 × 108 MSCs or HEK293T cells were seeded into the extra-capillary space (ECS) of a hollow fibre bioreactor cartridge as per the guidelines of the manufacturer. The level of glucose in the media was measured on a daily basis to monitor cell growth. After 1 week of growth, extracellular material was harvested by gently flushing 25 ml of Opti-MEM media into the ECS from the left end port to collect extracellular material on right end port of the cartridge. Withdrawn medium was flushed back and forth between the syringes, through the ECS, four times to dislodge material collected within the fibre bundles. The resulting harvest was used for isolation of EVs using the differential centrifugation, ultrafiltration and size-exclusion chromatography methodology as described above.
Nanoparticle tracking analysis
To measure size distribution and number of EVs, nanoparticle tracking analysis (NTA) was performed using the NS500 nanoparticles analyser (NanoSight, Malvern, Worcestershire, UK)71. Dilutions of EVs ranging from 1:500 to 1:1000 in PBS were used to achieve a particle count of between 2 × 108 and 2 × 109 ml−1. During the recordings, a camera level of 12–14 was used. Using the script control function, three 30 s videos of each sample were recorded. After each video, a fresh sample was injected into the stage followed by a delay of 7 s to reduce turbulence of the flow. NTA2.3 software provided by the manufacturer was used to analyse and measure size and concentration of the EVs. All NTA measurements were carried out in triplicate.
EV protease digestion assay
HEK293T and HeLa cells were seeded at a density of 8 × 106 cells in 15 cm cell-culture dishes. After 24 h, cells were co-transfected with CD81-GFP and DC-LAMP-lactoferrin encoding plasmids and EVs were isolated 48 h later. Purified EVs were concentrated in Amicon ultra 2 ml centrifugal filters (Merck, UK) at 4 °C in table-top centrifuge at 3200 × g. Concentrated EVs were analysed by NTA to determine number of EVs. 1 × 1012 EVs in 30 µl of PBS buffer were added to PCR tubes. The tubes were divided into three groups as:
Group I: Normal EVs. In this group neither proteases nor denaturants were added.
Group II: Protease treated EVs. Here, 0.2 µg/µl of pronase (mixture of proteases, Sigma UK) was added to EVs.
Group III: Denatured EVs treated with proteases. In this group, EVs were denatured by adding 0.1% triton X-100 followed by heating EVs at 100 °C for 5 min. After EVs were cooled down to room temperature, 0.2 µg/µl of pronase was added.
Samples treated with Pronase were incubated at 37 °C at different timepoints, varying from 5 to 60 min. After the incubation, proteases were inactivated by adding a protease inhibitor (final concentration 10× from 100× stock, Cambridge Bioscience UK). The samples were further incubated at room temperature for 5 min to allow inactivation of proteases followed by addition of reducing lithium dodecyl sulfate sample loading buffer (0.5× final concentration, ThermoFisher Scientific). The samples were immediately transferred into PCR machine maintained at 70 °C for 10 min. Treated and non-treated EV samples were loaded into SDS-PAGE (Bolt 4–12%, Bis-Tris gels, ThermoFisher Scientific UK) for western blotting. For B16-F10, SKOV-3 and MSCs cells, alix protein was used as a positive control for proteins residing in the lumen of the vesicles.
Electron microscopy
For electron microscopy (EM) analysis of EVs, samples were prepared as per the protocol described previously72. Briefly, 20 µl drops of purified EVs were added to parafilm and formvar-carbon-coated EM grids (Agar Scientific, Stansted, UK), which were floated on the top of EV droplets for 10 min with the coated side of the grid facing the drop. After the incubation, the grids were carefully transferred to PBS droplets for washing. Finally, the grids were placed briefly on water droplets to remove excess salt. For contrasting, grids were initially transferred to a 50 µl drop of uranyl oxalate solution, pH 7, for 5 min, followed by embedding in 2% uranyl acetate for 5 min. Next, the grids were carefully removed and excess of fluid was removed by gently pushing the grid sideways on Whatman no. 1 filter paper. The grids were air dried and visualized under a JEOL 1010 transmission electron microscope at 120 kV on an EFI Tecnai 12 TEM (JEOL, Peabody, MA, USA).
Western blot
For western blotting, EV lysates corresponding to 5–10 µg of total protein were separated on 4–12% Bis-Tris plus gels (Invitrogen, ThermoFisher Scientific). Prior to loading, EV proteins were denatured by adding NuPAGE LDS Sample loading dye and reducing agent (ThermoFisher Scientific) to the EVs and heated at 70 °C for 10 min. Samples loaded on the gel were run at 150 V for 1 h in ice-cold NuPAGE MES running buffer. Proteins resolved on the gel were transferred onto 0.45 µm Immobilon-P, PVDF Membrane (Millipore), using a mini transfer blot cell (Bio-Rad). Membranes were blocked with SuperBlock T20 (TBS) Blocking Buffer (ThermoFisher Scientific) for 1 h at room temperature with gentle shaking. After blocking, primary antibodies diluted in the blocking buffer were added to the membrane and incubated overnight at 4 °C on a shaker. Details of the antibodies used for western blotting of EVs are given in Table S1. After the incubation, the membranes were washed three times for 5 min each, using 1× tris-buffered saline, pH 7.6 (TBST, 20 mm Tris base, 150 mM of NaCl and 0.1% Tween). After the final wash, membranes were incubated with horseradish peroxidase (HRP)-conjugated secondary antibodies for 1 h at room temperature with gentle shaking. Post-incubation, membranes were washed three times for 5 min each with 1× TBST buffer and developed for by chemiluminescence detection (GE healthcare, RPN2106). The Odyssey FC imaging system (LI-COR) was used to visualize the bands on the membrane. Image studio software provided with the system was used to analyse the data.
For western blotting of proteins from cells, radioimmunoprecipitation assay (RIPA) buffer (ThermoFisher Scientific) containing 1× protease inhibitor (Halt protease inhibitor, ThermoFisher Scientific) was added to cells and incubated on ice for 10 min. Lysates of the cells were passed through 26 G needle syringes several times and centrifuged at 12,000 × g for 20 min to remove genomic DNA aggregates and insoluble lipids. Cell supernatants corresponding to 5–10 µg of the total proteins were used for the western blotting.
Purification of GAPDH from bacterial cells
The cDNA sequence of human GAPDH (OriGene, UK) was inserted into the pET-28b(+) vector (Novagen). Six residues of histidine were inserted at the N-terminus for purification by Ni-NTA chromatography and a FLAG-tag inserted at the C-terminus. The vector was transformed into BL21(DE3) competent E. coli cells (New
England Biolabs, UK) for expression. Sequences of GAPDH gene and primers used for cloning are given in Supplementary Table 6. 1 mM isopropyl β-D-1-thiogalactopyranoside (IPTG, Sigma) was used to induce expression of the gene in cells cultured at 37 °C. GAPDH protein was purified by Ni-NTA chromatography (Qiagen) under native conditions, using standard protocols recommended by the manufacturer. Ice-cold sodium-phosphate buffer, pH 8.0 (50 mM Na2HPO4/NaH2PO4, 10 mM Tris-Cl, 300 mM NaCl, 5 mM imidazole) was used during resuspension and lysis of the BL21(DE3) cells. Purified GAPDH protein was eluted with 250 mM imidazole in sodium-phosphate buffer, pH 6.0. Protein samples were dialyzed and passed through a Sephacryl s-200 HR column (GE Health Care) for further purification. The protein was analysed by SDS-PAGE, and the size of the protein was confirmed by mass spectrometry, using matrix-assisted laser desorption/ionization-time of flight-mass spectrometry (MALDI-TOF-MS).
G58T, G58T(TAT)2 and G58TF chimeric protein
The DNA sequence of GAPDH gene, which encodes the PS-binding domain (designated as G58), was fused with the second domain of the human TAR RNA-binding protein (TARBP2, domain sequence was from UniProtKB; Sequence ID: Q15633). The DNA sequence corresponding to 58–100 amino acid of GAPDH protein (PS-binding domain lies between 70–90 amino acids) were selected during fusion with TARBP2 second domain and recombinant DNA was synthesized commercially (Integrated DNA technology, Belgium). Additional amino acid sequence of G58 peptide (amino acid sequence from 58 to 69) were chosen for stability of G58 domain while fusing with other domains of the chimeric protein the chimeric protein. The DNA sequence of TAT and arginine-rich peptide of flock house virus alpha capsid protein was custom synthesized (Integrated DNA Technology) and fused to the TARBP2 second domain by PCR cloning. The sequence of G58 peptide and arrangement of domains in different G58 chimeric proteins is described in Table S2 and Table S7. The DNA sequences of these chimeric proteins were cloned into the pET-28b(+) vector. G58T was expressed in BL2(DE3) cells, using similar conditions to those mentioned above. G58T(TAT)2 and G58TF were expressed in BL2(DE3) Rosetta (Novagen, Merk, USA). The cells were grown normally at 37 °C until OD600 reached between 0.5 and 0.6. At this point, 0.2 mM IPTG was added to the culture, and bacteria were grown at 21 °C for 10–12 h. All versions of G58 chimeric proteins were purified by Ni-NTA chromatography under denaturing conditions, using 6 M urea. Purified proteins were analysed by SDS-PAGE and MALDI-TOF-MS, as described above. Proteins were refolded by the dilution method. Briefly, proteins bound to Ni-NTA resin were eluted by 250 mM imidazole in sodium-phosphate buffer, containing 2 M urea. Eluted proteins were further diluted with an equal volume of ice-cold PBS containing 5% glycerol and 0.1 mM DTT. After 5 min of incubation, the protein was added dropwise to PBS (with constant stirring) to reduce the urea concentration to 0.5 M. After the dilution, aggregated proteins were removed by centrifugation at 30,000 × g for 20 min (Beckman Coulter, USA). Protein was concentrated using centrifugal spin filters (Millipore) and passed through a PD10 column (GE Healthcare, USA) to remove final traces of urea.
Generation of GAPDH mutants and their interaction with EVs
Using the human GAPDH cDNA sequence as a template different types of GAPDH mutant were developed. For the G150T clone, the NAD+ binding domain of GAPDH was amplified and fused with the second domain of the TARBP2 protein. The DNA sequence was inserted into pET-28b(+) for expression in BL21(DE3) E.coli cells as described above. The protein was purified under denatured conditions using a Ni-NTA matrix followed by refolding of the protein by gradual dilution of urea concentration. Purified protein was isolated by gel-filtration column chromatography. To insert specific single amino acid mutant in wtGAPDH, two codons specifying Arginine 80 and Lysine 84 were replaced with alanine codons. The whole gene was synthesized as gBlock (integrated DNA technologies, Belgium) and inserted into pET-28b vector, using standard restriction digestion and ligation protocols. After confirming the sequence, mtGAPDH was expressed and purified from BL21(DE3) E. coli cells using Ni-NTA matrix under native conditions. Both an enzymatic assay and SDS-PAGE were used to characterize the protein.
For incubation of mtGAPDH with EVs, HEK293T EVs isolated either from a Bioreactor or by culturing HEK293T cells in optiMEM media were purified using TFF and gel-exclusion chromatography. 1.4 × 1011 EVs in 700 ul of PBS buffer were incubated with either wtGAPDH or mtGAPDH for 2 h at 4 °C. Excess of unbound protein was removed by size-exclusion chromatography. After elution, EVs were counted by Nanoparticle tracking analysis (NTA) using Nanosight NS500 (Malvern, UK). 3.0 × 108 particles in 10 µl of PBS were taken from each sample for GAPDH enzymatic kinetic assay (KDalert GAPDH assay, ThermoFisher Scientific). For immunoblotting, 7.5 × 109 EV particles were heated at 70 °C for 10 min after adding reducing SDS loading dye. Samples were loaded into precast 4–12% Bis-Tris gels (Invitrogen, ThermoFisher Scientific). Proteins resolved on SDS-PAGE were transferred to a PVD membrane that was incubated with monoclonal antibodies against the desired proteins. The membrane was developed with chemiluminescence (GE Health Care, US). Images and quantification of the gels were carried out with Odyssey FC imaging system (LI-COR).
Binding of GAPDH and G58T chimeric proteins to extracellular vesicles
For exogenous binding of GAPDH to EVs, purified EVs ranging from 7.5 × 1011 particles/ml to 1 × 1012 particles/ml were used for incubation with GAPDH and G58 chimeric proteins. Proteins (5–20 nmol as required) were added to 1 pmol of EVs and incubated for 2 h at 4 °C on a rotor top. Unbound proteins were removed by gel filtration. Proteins were also added directly to concentrated cell-culture medium, containing fetal calf bovine serum for binding followed by gel filtration to remove the unbound proteins from the EVs. Binding of GAPDH proteins to the EV surface was monitored by absorbance at 280 nm (Akta pure, GE Healthcare), western blot, GAPDH activity assay and gel-shift assay.
GAPDH activity assay
A fluorescence-based KDalert GAPDH assay (Invitrogen, Life Technologies) was used to measure enzymatic activity of GAPDH protein of extracellular vesicles. The assay measures the conversion of NAD+ to NADH in the presence of phosphate and glyceraldehyde-3-phosphate (G-3-P). Under the recommended assay conditions, the rate of NADH production is proportional to the amount of GAPDH enzyme present. A fixed number of EVs were added to the substrate in a 96-well plate. A fluorescent microplate reader (Clariostar Plus, BMG Labtech) was used to acquire data using the kinetic mode setting, with λex = 560 nm and λem = 590 nm. Data were analyzed using MARS Data Analysis software (Clariostar Plus, BMG Labtech).
Gel-shift assay
Binding of G58T, G58T(TAT)2 and G58TF EVs to siRNA was assessed by gel-shift assay. 20 pmol of siRNA was added to a fixed number of EVs in PBS buffer and incubated for 10 min at room temperature. After the incubation, complexes were loaded onto a 2% agarose gel stained with 0.5 µg/ml ethidium bromide for visualization under a UV-transilluminator. Binding of siRNA to EVs was determined by analyzing the shift of siRNA on the gel. siRNA bound to EVs remained trapped near the wells while free siRNA migrated freely in the agarose gel. Using NTA data and the gel-binding assay, the number of siRNAs bound to EVs was determined.
RNase A protection assay
For the RNase A protection assay, 0.2 mg/ml of RNase A (Qiagen) was added to EV-siRNA complexes and incubated for 6 h at 37 °C. The enzyme was inactivated by adding an equal volume of hot SDS lysis solution (2% SDS, 16 mM EDTA) and incubated at 100 °C for 5 min. siRNA was isolated using TRIzol reagent (Invitrogen, Life Technologies) and analysed by agarose-gel electrophoresis.
High-resolution single EV analysis by imaging flow cytometry
For single EV analysis experiments by Imaging Flow Cytometry (IFCM), EVs were isolated from HEK293 Freestyle suspension cells (ThermoFisher) and immortalized human cord blood-derived mesenchymal stromal cells (MSCs; Inscreenex) through ultrafiltration and bind-elute size-exclusion chromatography (BE-SEC). In brief, conditioned medium was pre-cleared by low-speed centrifugation (5 min at 700 × g, then 15 min at 2000 × g) and by filtration through 0.22 µm filters (Corning, cellulose acetate, low protein binding) before it was concentrated and diafiltrated with two volumes of PBS by tangential flow filtration (300 kDa MidiKros columns, 370 cm2 surface area, Spectrum Labs). EVs were further purified with BE-SEC columns (HiScreen Capto Core 700 column, GE Healthcare) and concentrated using Amicon Ultra-15 10 kDa molecular weight cut-off spin filters (Millipore). Particle concentrations were assessed by nanoparticle tracking analysis with a NanoSight NS500 instrument. Samples were diluted in PBS 0.2% human albumin (albunorm, Octapharma AB) to a final concentration of 1 × 1012 particles/ml before use. For control purposes, EVs tagged with mNeonGreen (neon GFP) fused to CD63 were prepared from HEK293 freestyle cells stably expressing CD63-mNeonGreen73. GAPDH expression and G58 peptide binding were studied on the single EV level by high-resolution IFCM (Amnis Cellstream, Luminex; equipped with 405, 488, 561 and 642 nm lasers) based on previously optimized settings and protocols with an Amnis ImagestreamX MkII instrument. In brief, 25 µl of EVs at a concentration of 1 × 1012 particles/ml were incubated overnight at 4 °C with 400 pmol AlexaFluor488 labelled G58 peptide and/or with AlexaFluor647-labelled rabbit anti human GAPDH antibodies (abcam, ab204480, clone EPR16884) or APC-labelled mouse anti human CD63 antibodies (Miltenyi Biotec, 130–100–182; clone H5C6) at a final concentration of 8 nM in v-bottom 96 wells (ThermoFisher Scientific). Samples were diluted 1:10,000 in PBS before acquisition using the plate reader of the Cellstream instrument with FSC turned off, SSC laser set to 40%, and all other lasers set to 100% of the maximum power. Small EVs were defined as SSC (low) by using neon GFP-tagged EVs as biological reference material and regions to quantify fluorescence-positive populations were set according to unstained samples and single-stained controls. Samples were acquired for 5 min at a flow rate of 3.66 µl/min (setting: slow) with CellStream software version 1.2.3 and analyzed with FlowJo Software version 10.5.0 (FlowJo, LLC). Dulbecco’s PBS pH 7.4 (Gibco) was used as sheath fluid.
Confocal microscopy
To determine uptake of siRNA-loaded EVs into cells, N2a cells were seeded onto coverslips in 6-well tissue culture plates (Sigma). Surface proteins of EVs were labelled with Alexa Fluor-633 (Invitrogen, Life Technologies). siRNACy3 was custom synthesized (Integrated DNA Technologies). After 24 h of seeding cells, complexes of Alexa Fluor-633-labelled EVs and siRNACy3 (20 pmol) were added to the cells and incubated for 6 h. After the incubation, cells were washed with PBS containing 20 U/ml heparin sulphate (Sigma), stained with Hoechst 33258 (Molecular probes, Life technologies) and fixed with 4% paraformaldehyde (ThermoFisher scientific, UK). The coverslips were mounted on glass slides using Ibidi mounting medium (Ibidi, Germany) and visualized using a fluorescence laser confocal microscope, ×60 oil immersion objective lens (FV 1000, Olympus). The data were processed using FV100 software.
In vitro gene silencing by G58-modified extracellular vesicles
For silencing of genes in N2a cells and HeLa cells, G58T, G58T(TAT)2 and G58TF proteins were incubated with either MSC- or HEK293T-derived EVs. After binding, purified EVs were analyzed for siRNA binding by gel-shift assay, to determine the amount of EVs needed to completely bind a given amount of siRNA. Using this ratio, EVs with bound siRNAs were added to the cells in complete DMEM GlutaMax medium. Details of the siRNA used for gene silencing experiment are given in Table S3. After 24 h of incubation, medium was changed and cells were further incubated. For mRNA analysis, cells were harvested after 48 h of treatment, using TRIzol (Invitrogen, Life Technologies). Two hundred and fifty nanograms of total RNA was used for reverse transcription PCR, using the PrimeScript reverse transcriptase kit (Takara, Japan). Probe-based reverse transcriptase-quantitative PCR (RT-qPCR) was used to assess quantities of GAPDH and Htt mRNA. 18 S rRNA and Hypoxanthine-guanine phosphoribosyl transferase (HPRT) mRNA was used as reference genes for normalization. PCR efficiencies were determined using LinRegPCR. Data were analyzed using the Pfaffl method74. Details of RT-qPCR assays are provided in Table S4. For protein analysis, cells were harvested 72 h post treatment and western blotting performed as described above.
Animal experiments
Biodistribution of extracellular vesicles
All animal experiments were performed in compliance with the Animals (Scientific Procedures) Act 1986, revised 2012 (ASPA, UK). Six-week-old female C57BL/6 mice were randomly assigned into four groups (n = 3). HEK293T cells stably transfected with RVG-LAMP-2B protein were seeded into 15 × 1.2 cm tissue culture plates for isolation of EVs. Immunoblotting of EVs and HEK293T cell lysates was carried out to assess expression of RVG peptide on surface of EVs. RVG-expressing EVs were labeled with sulpho-Cy5.5-NHS ester dyes (Abcam, ab235032). Briefly, 2 × 1012 EVs in 1 ml PBS buffer were incubated with 100 µmol of sulpho-Cy5.5-NHS ester, which was prepared in DMSO as a 10 mM stock. EVs were incubated at room temperature for 1 h with gentle shaking. After 1 h of incubation, G58TF protein was added to EVs and incubated for 2 h at 4 °C. Excess dye and protein was removed by passing the EVs through a gel-filtration column. After concentrating EVs, doses of EVs were given to animals as described in Table S5. After 4 h of administration, animals were sacrificed, organs harvested and Cy5.5 fluorescence visualized using the in vivo animal imaging system (IVIS, Perkin Elmer). Data were analyzed using the IVIS software.
Silencing of huntingtin gene Htt gene in Q140 Huntington disease model animals
Q140 mice were used to assess mutant and wild Htt gene silencing by intravenous administration of siRNA-loaded EVs. In the Q140 mouse model, exon 1 of mouse Htt is humanized and contains 140 CAG repeats. The mice have a slow progression of disease phenotype, which starts to appear at the age of 6 months. One-year-old Q140 mice were assigned to saline, negative siRNA control, and Htt siRNA treatment groups (n = 6). Saline group was used as a control to assess levels of Htt mRNA after administering EVs loaded with either negative siRNA (Negative group) or a mixture of Htt siRNA (treatment group). RVG-EVs bound to G58TF protein were used for the Htt silencing experiment. EV doses were calculated based on 0.5 mg/kg siRNA dosage regimen. The number of EVs needed to bind a given amount of siRNA were calculated by gel-shift assay. One hundred fifty to two hundred microlitres of EVs were administered intravenously. A total of four doses were given to animals at weekly intervals. Seventy two hours after the last dose, animals were euthanized and the various sections of the brain were analyzed for Htt mRNA and protein expression. For determining the level of HTT protein in the brain tissues, Agarose-gel electrophoresis for resolving aggregates (AGERA) was carried out to detect mutant HTT protein aggregates. However, we could not analyze the immunoblot due to high background. Immunohistochemistry of the cortex regions of the brain was carried out to determine the level of mutant HTT protein aggregates and p62 inclusion bodies.
Drosophila stocks and genetics
Flies were reared at 25 °C in vials containing standard cornmeal agar medium (12.5 g agar, 75 g cornmeal, 93 g glucose, 31.5 g inactivated yeast, 8.6 g potassium sodium tartrate, 0.7 g calcium, and 2.5 g Nipagen (dissolved in 12 ml ethanol) per litre. They were transferred onto fresh food every 2 days. No additional dried yeast was added to the vials. Temperature-controlled, SC-specific expression of UAS-CD63-GFP and UAS-Btl-GFP was achieved by combining these transgenes with the specific driver, dsx-GAL4 and the temperature-sensitive, ubiquitously expressed repressor tubulin-GAL80ts. Newly enclosed virgin adult males were transferred to 29 °C to induce post-developmental SC-specific expression. For overexpression and knockdown experiments, the same strategy was employed, but in the presence of a UAS transgene or the YFP-Rab11 gene trap. Six-day-old adult virgin males were used throughout this study to ensure that age- and mating-dependent effects on SC biology were mitigated44,45.
Preparation of accessory glands for live imaging and exosome secretion analysis
Adult male flies were anaesthetized using CO2. The abdomens were removed from anaesthetized flies and submerged in PBS (Gibco). The whole male reproductive tract was carefully pulled out of the body cavity. The accessory glands were isolated by separation of the ejaculatory bulb from the external genitalia, fat tissues and gut. Finally, the testes were removed by scission close to the seminal vesicles, as they often fold over the accessory glands, obscuring imaging.
The isolated accessory glands were transferred to a 9-spot depression glass plate (Corning PYREX) containing ice-cold PBS and kept on ice until sufficient numbers had been obtained. They were then stained with ice-cold 500 nM LysoTracker Red DND-99 (Invitrogen) (1:1000) in 1 × PBS for 5 min. Finally, the glands were rinsed in ice-cold PBS before being mounted onto high precision microscope cover glasses. A custom-built holder secured the cover glasses in place during imaging by wide-field microscopy. To avoid dehydration and hypoxia, the glands were carefully maintained in a small drop of PBS, surrounded by 10 S Voltalef (VWR Chemicals), an oxygen-diffusible hydrocarbon oil, and kept stably in place by the application of a small cover glass (VWR).
Imaging of Drosophila secondary cells
For wide-field imaging, living SCs were imaged using a DeltaVision Elite wide-field fluorescence deconvolution microscope (GE Healthcare Life Sciences) equipped with a ×100, NA 1.4 UPlanSApo oil objective lens (Olympus), and a Cool SNAP HQ2 CCD camera (Photometrics). The images acquired were typically z-stacks spanning 8–12 µm depth with a z-distance of 0.2 µm. Images were subsequently deconvolved using the Resolve 3D-constrained iterative deconvolution algorithm within the softWoRx 5.5 software package (GE Healthcare Life Sciences).
Confocal images of accessory glands were acquired by using LSM 880 laser scanning confocal microscope equipped with a ×63, NA 1.4 Plan APO oil DIC objective (Carl Zeiss). RI 1.514 objective immersion oil (Carl Zeiss) was employed.
Drosophila exosome secretion assay
Virgin six-day-old males of each genotype were dissected in 4% paraformaldehyde (Sigma–Aldrich) dissolved in PBS. The glands were left in 4% paraformaldehyde for 15 min to preserve the luminal contents before being washed in PBT (PBS containing 0.3% Triton X-100, Sigma–Aldrich) for 5 min. Glands were rinsed with PBS and then mounted onto SuperFrost Plus glass slides (VWR), removing excess liquid using a Gilson pipette, and finally immersed in a drop of Vectashield with DAPI (Vector Laboratories) for imaging by confocal microscopy.
Exosome secretion was measured by sampling within the central third of each gland. Identical microscope settings and equipment were used throughout. At each sampling location, ten different z-planes, spaced by 1 μm and at a distance from any SCs, were analysed.
The automated analysis of exosome secretion by SCs was performed using ImageJ2, distributed by Fiji. The ‘Noise>Despeckle’ function was first used to remove background noise, followed by conversion to a binary image. The number of fluorescent particles was assessed using the ‘Analyse Particles’ function. The average number of particles for all stacks was then quantified and compared to controls.
Analysis of ILV content in dense-core granule compartments
Living SCs from 6-day-old males of each genotype were imaged using identical settings by wide-field microscopy. The total number of DCG compartments that contained clustered fluorescently labelled puncta was counted in each cell, using a full z-stack of the epithelium. Three individual SCs were analysed from each of ten to fifteen glands.
Dense-core granule compartment analysis
Dense-core granule compartment numbers were manually quantified in ImageJ2 by using complete z-stacks of individual cells acquired with differential interference contrast (DIC) microscopy on the wide-field microscope. The morphology and position of DCGs were also analysed, and each compartment was scored for the presence of a single spherical DCG, an abnormally shaped DCG or multiple small fragmented DCGs. Three individual SCs per gland from each of ten to fifteen glands were used.
Statistical analysis
All data are representative from at least two to three independent experiments. The distribution of residuals was tested for normality using Q–Q plots and the appropriate statistical test was applied. For quantification of western blots, Image Studio Lite (Li-Cor) software was used. Adobe Photoshop CS4 software was used to crop and arrange the western blotting, confocal microscopy and electron microscopy figures. Nonparametric one-way ANOVA, using Bartlett’s and Brown-Forsythe test, was used to calculate F and P-values. For mouse experiments, data were plotted and analysed, using multivariant (two-tail) ANOVA. For SC analysis and exosome biogenesis, the Kruskal–Wallis test was employed followed by Dunn’s test to compare individual control and experimental datasets. All data analyses and statistics were conducted using GraphPad Prism v8.0 (GraphPad Software Inc., La Jolla, CA, USA).
Reporting summary
Further information on research design is available in the Nature Research Reporting Summary linked to this article.

