2018-BBA-KH26 Labeling of Extracellular Vesicles

You might also like

You are on page 1of 12

BBA - Biomembranes 1860 (2018) 1350–1361

Contents lists available at ScienceDirect

BBA - Biomembranes
journal homepage: www.elsevier.com/locate/bbamem

PKH26 labeling of extracellular vesicles: Characterization and cellular T


internalization of contaminating PKH26 nanoparticles
Pia Pužar Dominkuša,1, Matjaž Stenovecb,c,1, Simona Sitard, Eva Lasičc, Robert Zorecb,c,

Ana Plemenitaša, Ema Žagard, Marko Kreftb,c,e, Metka Lenassia,
a
Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Vrazov trg 2, Ljubljana, Slovenia
b
Celica BIOMEDICAL, Tehnološki park 24, Ljubljana, Slovenia
c
Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Zaloška 4, Ljubljana, Slovenia
d
Department of Polymer Chemistry and Technology, National Institute of Chemistry, Hajdrihova 19, Ljubljana, Slovenia
e
Department of Biology, Biotechnical Faculty, University of Ljubljana, Večna pot, 111, Ljubljana, Slovenia

A R T I C LE I N FO A B S T R A C T

Keywords: PKH lipophilic dyes are highly fluorescent and stain membranes by intercalating their aliphatic portion into the
Exosomes exposed lipid bilayer. They have established use in labeling and tracking of cells in vivo and in vitro. Despite
PKH26 wide use of PKH-labeled extracellular vesicles (EVs) in cell targeting and functional studies, nonEV-associated
Fluorescent dye fluorescent structures have never been examined systematically, nor was their internalization by cells. Here, we
Nanoparticles
have characterized PKH26-positive particles in lymphoblastoid B exosome samples and exosome-free controls
Confocal microscopy
Asymmetrical-flow field-flow fractionation
stained by ultracentrifugation, filtration, and sucrose-cushion-based and sucrose-gradient-based procedures,
using confocal imaging and asymmetric-flow field-flow fractionation coupled to multi-angle light-scattering
detector analysis. We show for the first time that numerous PKH26 nanoparticles (nine out of ten PKH26-positive
particles) are formed during ultracentrifugation-based exosome staining, which are almost indistinguishable
from PKH26-labeled exosomes in terms of size, surface area, and fluorescence intensity. When PKH26-labeled
exosomes were purified through sucrose, PKH26 nanoparticles were differentiated from PKH26-labeled exo-
somes based on their reduced size. However, PKH26 nanoparticles were only physically removed from PKH26-
labeled exosomes when separated on a sucrose gradient, and at the expense of low PKH26-labeled exosome
recovery. Overall, low PKH26-positive particle recovery is characteristic of filtration-based exosome staining.
Importantly, PKH26 nanoparticles are internalized by primary astrocytes into similar subcellular compartments
as PKH26-labeled exosomes. Altogether, PKH26 nanoparticles can result in false-positive signals for stained EVs
that can compromise the interpretation of EV internalization. Thus, for use in EV uptake and functional studies,
sucrose-gradient-based isolation should be the method of choice to obtain PKH26-labeled exosomes devoid of
PKH26 nanoparticles.

1. Introduction through lateral diffusion of these dyes, which also spread to in-
tracellular membrane organelles due to membrane turnover [2]. Cell
PKH dyes, such as PKH67 and PKH26, are highly fluorescent, lipo- growth, viability, and proliferation are generally not affected, and PKH
philic long-chain carbocyanine dyes that are used to stain artificial and dyes have been used extensively for labeling cells [3,4], in in vivo and
biological membranes. The aliphatic tail of these dyes rapidly inter- in vitro cell-tracking studies [5–7], and for visualizing homing and
calates into the exposed lipid bilayer and forms strong noncovalent proliferation of recirculating hematopoietic stem and progenitor cells
interactions, thereby promoting long-term dye retention and stable [8]. PKH dyes generally have an easy and rapid staining procedure,
fluorescence [1]. In cells, the whole plasma membrane becomes stained with simple dye detection by flow cytometry and fluorescent

Abbreviations: AF4-MALS, asymmetrical-flow field-flow fractionation coupled to multi-angle light-scattering detector; DPBS, Dulbecco's phosphate-buffered saline; EVs, extracellular
vesicles; Rgeom, geometric radius; Rrms, root-mean-square radius

Corresponding author at: Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Vrazov Trg 2, SI-1000 Ljubljana, Slovenia.
E-mail addresses: pia.puzar-dominkus@mf.uni-lj.si (P. Pužar Dominkuš), Matjaz.Stenovec@mf.uni-lj.si (M. Stenovec), Simona.Sitar@ki.si (S. Sitar), eva.lasic@mf.uni-lj.si (E. Lasič),
Robert.Zorec@mf.uni-lj.si (R. Zorec), ana.plemenitas@mf.uni-lj.si (A. Plemenitaš), ema.zagar@ki.si (E. Žagar), marko.kreft@mf.uni-lj.si (M. Kreft),
metka.lenassi@mf.uni-lj.si (M. Lenassi).
1
These authors contributed equally to this work.

https://doi.org/10.1016/j.bbamem.2018.03.013
Received 26 July 2017; Received in revised form 14 February 2018; Accepted 13 March 2018
Available online 16 March 2018
0005-2736/ © 2018 Elsevier B.V. All rights reserved.
P. Pužar Dominkuš et al. BBA - Biomembranes 1860 (2018) 1350–1361

microscopy. However, there are some concerns that remain when using and PKH26 dye resuspended in particle-free buffer as the control. The
these and other fluorescent probes, including noncell-associated signals size, number density, surface area, and fluorescence intensity of the
[2,4], dye transfer to adjacent or dead cells [9,10], dye cytotoxicity due particles in these PHK26-stained samples were determined using con-
to cell over-labeling [11], and dye phototoxicity due to over-exposure focal microscopy and asymmetric-flow field-flow fractionation coupled
[12]. to a multi-angle light-scattering detector (AF4-MALS). These data were
Recently, PKH membrane dyes have also been applied to studies of used to critically evaluate the suitability of the staining procedures.
extracellular vesicles (EVs) and their functions [13,14]. EVs are a het- Internalization of these PHK26-stained samples was also investigated
erogeneous population of membrane vesicles that are shed from cells in using live-cell confocal microscopy, to examine the relevance of the
vitro or are released into various body fluids in vivo [15]. They have nonEV-associated fluorescent structures in terms of EV internalization
been detected at relevant concentrations in blood [16], cerebrospinal and functional studies.
fluid [17], urine [18,19], semen [20], breast milk, saliva [21], amniotic
fluid [22], bronchoalveolar lavage [23,24], and synovial fluid [24].
2. Materials and methods
Numerous studies support the role of EVs in various physiological
processes, such as embryonic development [25], synaptic plasticity
2.1. Exosome staining
[26], liver metabolism [27,28], and antigen presentation [29]. They
have also been shown to be involved in promoting pathologies through
Lyophilized exosome standard that was purified from the lympho-
modulation of immune responses [30], tumor invasiveness and metas-
blastoid B cell-line culture media (HansaBioMed, Estonia) was re-
tasis [31,32], resistance to drugs [33], and transfer of toxic proteins
suspended in ultra-pure water to a protein concentration of 1.0 μg/μL,
[34–37]. This functional diversity of EVs is reflected in their distinctive
following the manufacturer instructions. The resuspended exosomes
protein, lipid, and nucleic acid cargoes, which mirror those of the cell of
were then stained with PKH26 using PKH26 Red Fluorescent Cell Linker
origin [38]. According to their size and site of formation, EVs can be
Kits for General Cell Membrane Labeling (Sigma-Aldrich). Prior to
divided into two main groups: exosomes, which range from 30 nm to
staining, PKH26 in diluent C was incubated in an ultrasonic water bath
100 nm in diameter and are formed as intraluminal vesicles in multi-
at 37 °C for 15 min. Alternatively, for the control sample, particle-free
vesicular bodies, which fuse with the plasma membrane for their re-
Dulbecco's phosphate-buffered saline (DPBS; Sigma-Aldrich) was used
lease; and microvesicles, which are larger structures from 100 nm to
as the input instead of the exosome standard. Exosome and control
1000 nm in diameter that are formed directly at the plasma membrane
samples were stained according to the following procedures.
through outward budding [39].
Functionally, EVs can interact with their target cells through one of
a number of processes, including: fusion or hemi-fusion with the plasma 2.1.1. Procedure A
membrane, followed by the “injection” of their intraluminal contents PKH26 dye was diluted in 100 μL diluent C to a final concentration
into the cytosol; ligand-receptor binding, to modulate intracellular of 8 μM (dye solution). Then 10 μg of exosomes (AF4-MALS analysis,
signaling pathways; internalization through clathrin-dependent en- 20 μg) in 20 μL DPBS were diluted with 80 μL diluent C, added to the
docytosis and clathrin-independent pathways, such as caveolin-medi- dye solution, and incubated for 5 min while mixed with gentle pipet-
ated uptake, macropinocytosis, phagocytosis, and lipid-raft-mediated ting. Excess dye was bound with 100 μL 10% exosome-depleted fetal
endocytosis [40–42]. As inhibitors of specific internalization pathways bovine serum (Sigma-Aldrich) in Dulbecco's modified Eagle's medium
fail to completely abrogate EV uptake by cells, it appears that EVs can (Sigma-Aldrich). Then the exosomes were diluted to 1 mL with DPBS
be internalized via more than one route [42]. EV uptake by cells can be and pelleted by ultracentrifugation at 100000 ×g for 1 h 10 min at 4 °C
visualized directly by use of fluorescent lipid membrane dyes that stain (TLA-55; Beckman Coulter). The pellet was gently resuspended in 50 μL
the EV membranes. By labeling EVs with PKH26 or PKH67, the inter- DPBS.
actions with and uptake of EVs by epithelial and breast cancer cells
[13], macrophages [43], dendritic cells [29], and endothelial and 2.1.2. Procedure B
myocardial cells [44] have been followed using fluorescence micro- Exosomes were stained as described in procedure A. Then they were
scopy and flow cytometry. Similarly, EV uptake by bladder cancer cells diluted to 2 mL with DPBS, transferred to Vivaspin20 300-kDa MWCO
has been studied using imaging flow cytometry [14]. PKH67 pre-la- filters (Sartorius Stedim Biotech GmbH), and centrifuged at 4000 ×g
beled EVs isolated from dendritic cells have also been used in quanti- for 3 min at 4 °C. The exosomes were washed three times with 2 mL
tative and qualitative analyses using high-resolution flow cytometry DBPS before being concentrated to a final volume of 50 μL.
[45].
Despite the wide use of PKH-labeled EVs in cell targeting and
2.1.3. Procedure C
functional studies, nonEV-associated fluorescent structures have never
Exosomes were stained as described in procedure A. Then they were
been systematically characterized, and their internalization by cells has
diluted to 10 mL with DPBS and placed on top of 2 mL 20% sucrose
never been evaluated. The need for such studies was supported by Lai
(Millipore) in DPBS. The exosomes were pelleted by ultracentrifugation
et al. [46], who showed that staining with PKH67 resulted in control
at 100000 ×g for 1 h 10 min at 4 °C (MLA-50; Beckman Coulter). The
samples (i.e., without EVs) that had greater fluorescent signals than
pellet was gently resuspended in 50 μL DPBS.
PKH67-stained EVs. Additionally, tissue macrophages have been shown
to actively acquire various labels in vivo [47], and endocytosis of mi-
celles has been exploited for cellular uptake of nanomedicines [48]. 2.1.4. Procedure D
Thus, the nonEV-associated fluorescent structures present in PKH-la- Exosomes were stained as described in procedure A. Then they were
beled EV samples can compromise the interpretation of EV inter- diluted to 400 μL with DPBS and placed onto a 20% to 60% dis-
nalization and functional studies, and especially when the appropriate continuous sucrose gradient. The gradient was ultracentrifuged at
EV-free controls are not included. 100000 ×g for 18 h at 4 °C (MLS-50; Beckman Coulter). Fractions 3 to 6
Here, the aim was thus to characterize the nonEV-associated fluor- (density range 1.08–1.15 g/mL as measured by refractometer, data not
escent structures in PKH26-labeled EV samples that were stained using shown) were combined, and fractions 7 to 10 (1.17–1.23 g/mL) were
procedures based on ultracentrifugation [46], filtration through Vi- combined, and each diluted to 30 mL with DPBS. The exosomes were
vaspin concentrators [21,49], and sucrose cushion [50] and sucrose pelleted by ultracentrifugation at 100000 ×g for 1 h 10 min at 4 °C
gradient [45] purification. The input samples were exosomes that were (MLA-50; Beckman Coulter). The pellets were gently resuspended in
isolated from culture supernatants from a lymphoblastoid B cell line, 50 μL DPBS.

1351
P. Pužar Dominkuš et al. BBA - Biomembranes 1860 (2018) 1350–1361

2.2. Characterization of PKH26-positive particles and the geometric radius (Rgeom). These radii were obtained from the
MALS detector without the need for the solute concentration and the
2.2.1. Confocal microscopy, image analysis, and statistical analysis increment in the sample refractive index. The Rrms of the fractionated
Small volumes (6 μL) of gently premixed PKH26-labeled control exosomes were calculated using the data from 15 angles from the MALS
(i.e., particle-free DPBS) and exosome samples were transferred to detector, applying a Debye second-order model. The Rgeom were de-
coverslips coated with 10% poly-L-lysine (to favor strong adhesion), termined by the Astra Particle template assuming spherical exosome
sealed to the objective glass, and transferred to a confocal microscope shape. The number density of particles (particles/mL) was evaluated by
for observation (LSM 780; Zeiss). Manual focusing was performed to the Astra Number density template, using an exosome refractive index
obtain the sharpest images of the bright fluorescent particles, which of 1.39 [51]. The recovery of exosomes processed by staining proce-
were monitored within a rectangular field of 2030 μm2 by a plane- dures A and C was calculated by dividing the experimental yield
apochromatic oil-immersion objective (63×/NA 1.4). PKH26 was ex- (number density of unstained exosome output) by the theoretical yield
cited using a 561-nm diode-pumped solid state laser line (2%), and (number density of unstained exosome input) and was expressed as
emission fluorescence was filtered with a 565-nm to 605-nm band-pass percentage.
filter. The settings for the detection of PKH26 emission fluorescence
were kept constant for the given sample pair of the PKH26-labeled
exosome samples and the corresponding controls (procedures A–D). 2.3. Cellular internalization of PKH26-positive particles
Confocal images were exported to tiff files that were analyzed using
the ImageJ software (National Institutes of Health, Bethesda, MD, USA). 2.3.1. Cell culture
Each PKH26-positive particle of several adjacent pixels with fluores- Primary astrocyte cultures were prepared from cortices of 3-day-old
cence intensity ≥51 gray-scale levels (intensity scale, 0–255) was ex- female Wistar rats (obtained from Medical Experimental Centre at the
tracted from the image using the 20% intensity threshold level. The Institute of Pathology, University of Ljubljana, Slovenia) as described
minimum size taken to identify PKH26-positive particles was three previously [52]. The experimental animals were cared for in ac-
adjacent pixels (x = 0.088 μm), and thus the minimum surface area cordance with European and Slovenian legislation (Official Gazette of
covered by a fluorescent particle was 0.023 μm2. The total number of the RS 38/13; UVHVVR, No·U34401-47/2014/7). The cells were sub-
PKH26-positive particles per image, the surface area, and the fluores- cultured onto 22-mm-diameter poly-L-lysine-coated coverslips and used
cence intensity were determined for the PKH26-labeled exosome sam- within 4 days of plating. The cells were maintained in high glucose
ples and the corresponding controls (procedures A–D). All measured Dulbecco's modified Eagle's medium (Thermo Fisher Scientific, Wal-
parameters are reported as means ± standard error of the means tham, MA, USA) containing 10% fetal bovine serum, 1 mM pyruvate,
(SEM). Statistical significance was determined using Mann-Whitney U 2 mM glutamine, 5 U/mL penicillin and 5 μg/mL streptomycin, at 37 °C
tests, using SigmaPlot 11.0 (Systat Software Inc., USA). in a 95% air/5% CO2 atmosphere. Unless stated otherwise, all chemi-
cals were obtained from Sigma-Aldrich (Munich, Germany) and were of
2.2.2. Asymmetric-flow field-flow fractionation coupled to a multi-angle the highest purity grade available.
light-scattering detector and data analysis
Asymmetric-flow field-flow fractionation separations were per-
formed on a system (Eclipse3+; Wyatt Technology Europe GmbH, 2.3.2. Live-cell confocal imaging
Dernbach, Germany) connected to an isocratic pump, an on-line va- Cultured astrocytes were co-incubated with either PKH26-labeled
cuum degasser, and an autosampler (all 1260 series; Agilent control (i.e., particle-free DPBS; con-procedure C) or PKH26-labeled
Technologies, USA). The samples were separated in a trapezoidal- exosome (exo-procedure C) samples for 3 h at 37 °C, in serum-free
shaped channel with a 350-μm spacer and a Nadir Cellulose medium. In a subset of experiments, 20 μM fluorescent dextran (Alexa
Regenerated Cellulose membrane with a 10-kDa cut-off. The fractio- Fluor 488, 3000 MW, D34682, Thermo Fisher Scientific) was also added
nated particles were detected using a MALS detector (Dawn Heleos, to the cells together with the samples. After labeling, the cells were
Wyatt Technology, USA) at 658 nm, which was calibrated using toluene washed with extracellular solution (131.8 mM NaCl, 1.8 mM CaCl2,
and normalized with bovine serum albumin protein as an isotropic 2 mM MgCl2, 5 mM KCl, 10 mM D-glucose, 10 mM HEPES/NaOH,
scatterer standard. Phosphate-buffered saline (pH 7.4) was the running pH 7.2), mounted into the recording chamber, supplied with extra-
eluent, and was composed of 137 mM NaCl, 2.68 mM KCl, 10.14 mM cellular solution, and transferred to the microscope for observation. Z-
Na2HPO4, 1.84 mM KH2PO4, supplemented with 0.02% (w/v) sodium stacked confocal images of fluorescent compartments in live cells were
azide as a bactericide, and filtered through 0.45-μm pore-size mem- acquired as stated above. Alexa Fluor 488 was excited using a 488-nm
brane (Nylon 66; Supelco Analytical, USA). An additional filter with argon laser line (2%) and emission fluorescence was filtered with the
0.1-μm pore size was placed between the HPLC pump and the AF4 500-nm to 550-nm band-pass filter. The settings for the detection of the
channel (in a PEEK Inline Filter Holder). PKH26 emission fluorescence were constant in cells loaded with
Thirty-five microliters of gently premixed samples (PKH26 solution, PKH26-labeled control and PKH26-labeled exosome samples.
PKH26-labeled controls [i.e., particle-free DPBS], exosome solution,
PKH26-labeled exosome samples) were injected during a focusing/in-
jection step of 5 min, with a focus flow rate of 1.5 mL/min and an in- 2.3.3. Analysis of the PKH26-positive intracellular compartments after co-
jection flow rate of 0.2 mL/min, followed by a focusing/relaxation step incubation of primary astrocytes with PKH26-positive particles
for an additional 7 min. The elution step was performed with two linear The total number of PKH26-positive compartments per cell, the
cross-flow gradients, a high initial gradient of 0.275 mL/min2 (i.e., volume and the fluorescence intensity were analyzed (3D Object
cross-flow from 3.0 mL/min to 0.25 mL/min in 10 min) followed by a Counter plugin for ImageJ; National Institutes of Health, Bethesda, MD,
lower gradient of 0.0036 mL/min2 (cross-flow from 0.25 mL/min to USA) in z-stacked confocal images on the basis that individual com-
0.09 mL/min in 45 min). Here, 0.09 mL/min was the lowest cross-flow partment consisted of three to 400 adjacent voxels
limit of the instrument. Once this limit was reached, the cross-flow was (0.132 × 0.132 × 0.601 μm); thus a broad span of PKH26-positive
turned off. The detector flow rate was 1 mL/min. The last step was compartments with different apparent sizes and intensities imaged at
washing the channel for 10 min in elution mode without any cross-flow. different z positions were analyzed. All measured parameters are re-
The Astra 5.3.4.20 software (Wyatt Technology; Santa Barbara, ported as means ± SEM. Statistical significance was determined using
USA) was used to analyze the data. The size of the exosomes was ex- Mann-Whitney U tests, using SigmaPlot 11.0 (Systat Software Inc.,
pressed as two different radii; i.e., the root mean square radius (Rrms) USA).

1352
P. Pužar Dominkuš et al. BBA - Biomembranes 1860 (2018) 1350–1361

Fig. 1. Schematic representation of staining procedures used to label exosomes with


PKH26 dye. Fig. 2. PKH26-labeled exosome-free controls and exosome samples prepared using pro-
Exosomes (exo) were labeled using staining procedures based on ultracentrifugation cedure A.
(procedure A), filtration through Vivaspin concentrators (procedure B), and sucrose (a–d) Representative confocal images of PKH26-positive particles in a PKH26-labeled
cushion (procedure C) and sucrose gradient (procedure D) purification. Key differences exosome-free control (i.e., particle-free DPBS; con) (a) and in a PKH26-labeled exosome
between the procedures are highlighted by the green text. ∂, centrifugation. For details, (exo) (b) sample prepared following procedure A. Inset: Magnified selected PKH26-po-
see Section 2.1. sitive particle. Scale bars: 2 μm; inset, 0.4 μm. (c, d) Black and white image masks from
(a), (b) after image segmentation for individual PKH26-positive particles, as exosome-free
control (c; 72 particles) and exosome (d; 577 particles) samples, respectively. Scale bars:
3. Results 2 μm. (e–g) Quantification of the PKH26-positive particles per image field of 2030 μm2, as
the numbers (e; N°), surface areas (f; S), and fluorescence intensities (g; Int.). Data are
3.1. Ultracentrifugation-based exosome staining generates numerous means ± SEM. Numbers (bottom, e) indicate confocal images analyzed, whereas num-
PKH26 nanoparticles with dimensions similar to PKH26-labeled exosomes bers (top, f) indicate PKH26-positive particles analyzed in f and g. ***, p < 0.001 versus
control (con; Mann-Whitney U tests).
PKH membrane dyes are being used increasingly in studies of EVs
and their functions, although no comprehensive study of nonEV-asso- microscopy, AF4-MALS, dynamic light scattering, and nanoparticle
ciated fluorescent structures present in PKH-stained EV samples has tracking analysis [53]. The PKH26-positive particles in the confocal
been carried out. The existence of nonEV-associated PKH-positive images were analyzed for the number of particles per image, their
structures is supported by Lai et al. [46], who showed that staining with surface area, and their fluorescence intensity (Fig. 2).
PKH67 resulted in controls (PKH67-stained medium depleted of EVs) The confocal images revealed fluorescent particles of heterogeneous
with greater fluorescence signals than the PKH67-stained EV samples. sizes and fluorescence intensities (Fig. 2a, b) in the PKH26-labeled
To characterize these nonEV-associated PKH-positive structures here, exosome-free controls and PKH26-labeled exosome samples prepared
exosomes and particle-free DPBS (control) were labeled with PKH26 using procedure A. More fluorescent particles were observed in the
following the ultracentrifugation-based staining procedure (Fig. 1, exosome-containing samples than in the corresponding exosome-free
procedure A), with the PKH26-labeled samples later analyzed using controls (Fig. 2a, b), as also indicated by the black and white mask
confocal microscopy. For exosomes, exosome standards purified from images obtained after segmentation of the confocal images (Fig. 2c, d).
the culture supernatant of a lymphoblastoid B cell line were used There were 579 ± 8 (n = 20) versus 72 ± 3 (n = 20) PKH26-positive
(provided by HansaBioMed), which we previously characterized for particles per image in the exosome-containing samples and exosome-
purity, morphology, size and number density using electron free controls, respectively (Fig. 2e). The particle surface areas were

1353
P. Pužar Dominkuš et al. BBA - Biomembranes 1860 (2018) 1350–1361

similar, whereas the particle fluorescence intensities were significantly Table 1


smaller in the exosome-containing samples than the exosome-free AF4-MALS measurements of the PKH26-labeled exosome-free (Control) and exosome
samples prepared using procedures A and C.
controls (Fig. 2f, g).
Of note, the measured areas of the fluorescent particles did not re- Sample Preparation Radius (nm) Number densityb
flect their actual areas, as the point-spread function contributes to some
degree of spreading (i.e., blurring) in the images of the particles. To test Procedure Rrms Rgeom (particles/mL × 1010)
this empirically, sub-diffraction fluorescent beads of known diameters a
Exosome standard 85 101 3.85
were imaged, and their diameters were calculated from the confocal Control A 77 84 0.75
images under improved contrast and assuming spherical bead geo- Exosomes A 79 94 10.70
metry. With fluorescent beads of 200-nm diameter, which is close to the Control C 65 70 0.79
theoretical lateral resolution of the fluorescent microscope Exosomes C 81 93 1.13

(d = 202.5 nm; d = λem/2NA; λem = 567 nm; NA = 1.4), the calcu- a


Unstained and unprocessed exosome sample.
lated bead diameter of 206 ± 8 nm (n = 287) corresponded to the b
Number of particles per mL in the final volume of 50 μL DPBS for 20 μg of exosome
actual diameter given. With fluorescent beads of 48-nm diameter, the input (Fig. 1).
calculated bead diameter of 155 ± 3 nm (n = 413) exceeded the given
diameter by 3.2-fold. Therefore, the images of the fluorescent particles by the mean particle Rrms (Rgeom), of 77 (84) nm and 79 (94) nm for the
probably represented both larger individual and smaller clustered ob- PKH26-labeled exosome-free control and PKH26-labeled exosome
jects with diameters below the diffraction limit. Thus, it is possible that samples, respectively, which were slightly higher in the unstained
the largest PKH26-positive particles imaged in the exosome-containing exosome sample, at 85 (101) nm (Table 1). Importantly, as seen from
samples (Fig. 2b, d) represent aggregated particles. Although there Fig. 3a, the PKH26 solution used for the labeling did not give any MALS
were ~8-fold more PKH26-positive particles in the exosome-containing signal, which indicates that the concentration of the PKH26-positive
samples compared to the exosome-free controls, the examined char- particles before processing (Fig. 1) was below the detection limits.
acteristics of the PKH26-particles were similar in both samples. In addition, the number densities were determined (Fig. 3b,
To further characterize the PKH26-positive particles obtained after Table 1). Within the experimental error, the total number density of the
staining of the exosome-free controls and exosome-containing samples particles in the PKH26-labeled exosome samples
using procedure A (Fig. 1), they were analyzed using AF4-MALS. The (10.70 × 1010 particles/mL) was 14.3-fold higher than that for the
AF4 technique has a broad separation range, from a few nanometers up control samples (0.75 × 1010 particles/mL). This observed higher
to a micrometer, and in combination with a MALS detector it is suitable number density in the PKH26-labeled exosome samples was not due to
for separation and characterization of a wide range of biological (e.g., enrichment of any specific particle subpopulation, as a similar increase
virus-like particles, exosomes) and nonbiological (e.g., proteins, plas- in particle numbers was observed over the whole size interval (Fig. 3b).
mids, polysaccharides) analytes [53]. Importantly, unlike confocal mi- As also seen by imaging, a small population of larger particles was
croscopy, in addition to the fluorescent particles, AF4-MALS also de- detected in the PKH26-labeled exosome samples that was not seen in
tects nonfluorescent particles. From the fractograms obtained, the size the unstained exosomes, which might have represented aggregated
distributions, mean sizes, and number densities of the particles were particles (Fig. 3b). To estimate the proportions of exosomes in the
determined (Fig. 3, Table 1). The sizes were expressed according to two PKH26-labeled exosome samples, we first determined the percent re-
different radii: root mean square radius (Rrms), and geometric radius covery of unstained exosomes processed as in procedure A, by mea-
(Rgeom). In addition to the labeled samples, the AF4-MALS analysis was suring the number densities of exosomes before (theoretical yield;
also performed for the PKH26-staining solution and unstained exo- 3.85 × 1010 particles/mL, Table 1) and after (experimental yield;
somes. 1.19 × 1010 particles/mL, data not shown) the processing, using AF4-
As shown in Fig. 3, the elution profiles of the PKH26-labeled exo- MALS. The recovery of the exosomes exposed to the ultracentrifugation-
some-free control (con-procedure A) and PKH26-labeled exosome (exo- based staining of procedure A was approximately 31%. Thus when
procedure A) samples recorded by MALS showed relatively broad size staining 20 μg of exosomes (corresponding to 19.25 × 1012 exosomes)
distributions (Rrms, 20–100 nm). Interestingly, the elution profile of the using procedure A (Fig. 1), we expect to obtain approximately 6 × 1012
unstained exosomes (exo), which were used as the input sample for the exosomes in the PKH26-labeled exosome samples. As we detected
PKH26-labeling procedures (Fig. 1), mostly coincided with the profiles 53.5 × 1012 particles (10.70 × 1010 particles/mL in 50 μL; Table 1), the
of both of the PKH26-labeled samples, so even the exosome-free con- exosomes appear to represent only 11% of all of the particles, with the
trol. The presence of similar particles in all three samples was supported rest likely representing PKH26 dye particles (PKH26 nanoparticles;

Fig. 3. AF4-MALS analysis of PKH26-labeled exosome-free controls and exosome samples prepared using procedure A.
Normalized AF4-MALS fractograms of the PKH26-labeling solution (PKH26; grey), PKH26-labeled exosome-free control (con-procedure A; red), unstained exosomes (exo; black) and
PKH26-labeled exosome sample (exo-procedure A; blue) for Rrms (a) and number density (b). Samples were stained following procedure A (Fig. 1). Continuous curves represent
normalized light scattering signal, while empty circles represent Rrms (a) and number density (b).

1354
P. Pužar Dominkuš et al. BBA - Biomembranes 1860 (2018) 1350–1361

47.5 × 1012).
Similar to the confocal imaging, the AF4-MALS analysis allowed the
detection of ~14-fold more PKH26-positive particles in the exosome-
containing samples compared to the exosome-free controls, with par-
ticles from both samples having similar characteristics. Importantly, in
procedure A, only one out of every 10 particles in the PKH26-labeled
exosome samples appears to represent exosomes. Overall, the com-
monly used ultracentrifugation-based exosome staining of procedure A
generates numerous PKH26 nanoparticles, which are almost indis-
tinguishable from the PKH26-labeled exosomes in terms of their size,
surface area, and fluorescence intensity.

3.2. Low PKH26-positive particle recovery is characteristic of filtration-


based exosome staining

As an alternative, Lässer et al. [21] used a filtration-based exosome


staining procedure, with the concentration and washing of the exo-
somes through 300-kDa filters. To evaluate the efficiency of this
staining procedure and its propensity to promote PKH26 nanoparticles
formation, exosomes and particle-free DPBS (control) were labeled with
PKH26 following procedure B (Fig. 1). The PKH26-positive particles in
the suspension were then evaluated using confocal microscopy, as
above (Fig. 4a–d). Procedure B resulted in the preparation of samples
with similarly low numbers of fluorescent particles per image for both
the PKH26-labeled exosome-free control and PKH26-labeled exosome-
containing samples: 2 ± 2 (n = 10) versus 1 ± 0.1 (n = 20), respec-
tively (Fig. 4e). The particle surface areas and fluorescence intensities
were lower in the PKH26-labeled exosome-free controls than in the
PKH26-labeled exosome-containing samples (Fig. 4f, g). These samples
prepared using procedure B were further analyzed by AF4-MALS, al-
though the sample concentrations were below the detection limit of the
instrument, so no further analyses were performed. Overall, low
PKH26-positive particle recovery was characteristic of the filtration-
based exosome staining of both the exosome-free controls and exosome-
containing samples.

3.3. PKH26 nanoparticles and PKH26-labeled exosomes purified through a


sucrose cushion can be differentiated on the basis of size

Lentiviruses, e.g. human immunodeficiency virus (HIV), and lenti- Fig. 4. PKH26-labeled exosome-free controls and exosome samples prepared using pro-
viral vectors are generally purified by ultracentrifugation through a cedure B.
(a–d) Representative confocal images of PKH26-positive particles in a PKH26-labeled
sucrose cushion to remove any impurities of lower buoyant density than
exosome-free control (i.e., particle-free DPBS; con) (a) and in a PKH26-labeled exosome
that of virions [54]. Exosomes are similar to HIV in terms of many of (exo) (b) sample prepared following procedure B. Inset: Magnified selected PKH26-po-
their physical properties and have similar buoyant density in a sucrose sitive particle. Scale bars: 2 μm; inset, 0.4 μm. (c, d) Black and white image masks from
gradient [55,56]. Therefore, we next analyzed whether PKH26 nano- (a), (b) after image segmentation for individual PKH26-positive particles (encircled), as
particles can be removed from PKH26-labeled exosomes using pur- exosome-free control (c; 1 particle) and exosome (d; 1 particle) samples, respectively.
ification through a sucrose cushion (Fig. 1, procedure C). The PKH26- Scale bars: 2 μm. (e–g) Quantification of the PKH26-positive particles per image field of
2030 μm2, as the numbers (e; N°), surface areas (f; S), and fluorescence intensities (g; Int.).
labeled exosome-free controls and PKH26-labeled exosome-containing
Data are means ± SEM. Numbers (bottom, e) indicate confocal images analyzed,
samples were first analyzed for the numbers of particles per image, whereas numbers (bottom, f) indicate PKH26-positive particles analyzed in f and g. **,
surface areas, and fluorescence intensities using confocal microscopy p < 0.01; ***, p < 0.001 versus control (con; Mann-Whitney U tests).
(Fig. 5). When compared to the ultracentrifugation-based staining
procedure (procedure A), sucrose cushion ultracentrifugation resulted
exosome-free control (con-procedure C) and PKH26-labeled exosome-
in fewer PKH26-positive particles in both the exosome-free controls and
containing (exo-procedure C) samples were further analyzed by AF4-
exosome-containing samples, with the large PKH26-labeled fluorescent
MALS for their particle sizes and number densities (Table 1). The
aggregates no longer seen in the PKH26-labeled exosome samples
fractograms for the unstained and PKH26-labeled exosome samples
(compare Fig. 5a–d with Fig. 2a–d). The number of PKH26-positive
showed relatively broad size distributions (Rrms, 25–100 nm), whereas
particles per image for the exosome-free controls was 46 ± 3 (n = 20)
the size distribution in the exosome-free control samples was a little
versus 340 ± 6 (n = 20) PKH26-positive particles per image for the
narrower (Rrms, 25–80 nm) (Fig. 6a). The presence of smaller particles
exosome-containing samples (Fig. 5e). Similarly, the PKH26-positive
in the PKH26-labeled exosome-free control samples compared to the
particle surface area (Fig. 5f) was smaller in the control than in exo-
PKH26-labeled exosome samples was supported by the mean particle
some-containing samples, whereas the PKH26-positive particle fluor-
Rrms (Rgeom), which were 65 (70) nm and 81 (93) nm, respectively
escence intensity (Fig. 5g) was similar in both samples prepared using
(Table 1). Interestingly, the mean particle size in the PKH26-labeled
procedure C. Importantly, there were ~7-fold more PKH26-positive
exosome samples was similar to the mean size of the unstained exo-
particles in the exosome-containing samples compared to the exosome-
somes, as 85 (101) nm. Although the total number densities of the
free controls, and these also differed in their surface areas.
particles in the PKH26-labeled exosome-free controls and PKH26-
As shown in Fig. 6, sucrose-cushion-purified PKH26-labeled

1355
P. Pužar Dominkuš et al. BBA - Biomembranes 1860 (2018) 1350–1361

of all of the particles, with the rest likely representing PKH26 nano-
particles (3.73 × 1012).
Analysis by AF4-MALS supported the confocal imaging observation
that the PKH26 nanoparticles were smaller in size when compared to
the PKH26-positive particles in the exosome samples. Conversely, only
1.4-fold more PKH26-positive particles were detected in the exosome-
containing samples, compared to the exosome-free control samples.
Three to four out of 10 PKH26-positive particles in the PKH26-labeled
exosome samples appeared to represent exosomes. Importantly, similar
observations were made when using the PKH67 dye instead of the
PKH26 dye for sucrose-cushion-based staining of exosomes (for details
see Appendix A, Table S1, Fig. S1).
In conclusion, although the purification through the sucrose cushion
is not very efficient in removing the PKH26 nanoparticles from the
PKH26-labeled exosome samples, it affects the PKH26 nanoparticle
size, and thereby helps in their differentiation from the PKH26-labeled
exosomes, especially when analyzed using confocal microscopy.

3.4. PKH26-labeled exosomes separate from PKH26 nanoparticles by


passing into denser sucrose gradient fractions

As none of the above procedures provided pure PKH26-labeled


exosomes devoid of PKH26 nanoparticles, the PKH26-labeling protocol
optimized for flow cytometry analysis by Van der Vlist et al. [45] was
then followed. In this procedure, the particles in the PKH26-labeled
exosome samples were separated on a sucrose gradient based on their
buoyant densities, with the PKH26-labeled exosomes generally separ-
ating into fractions with densities between 1.08 g/mL and 1.22 g/mL
[15,57,58]. After following the PKH26-labeling protocol of procedure D
(Fig. 1), fractions 3–5 of the gradient were visibly stained in the PKH26-
labeled exosome-free control (con) and fraction 9 in the PKH26-labeled
exosome-containing sample (exo; data not shown). To isolate the
PKH26-particles from the stained fractions for analysis by confocal
microscopy (Fig. 7), fractions 3–6 (1.08–1.15 g/mL, as measured using
a refractometer) and 7–10 (1.17–1.23 g/mL) were collected for both
samples. These were diluted in a large volume of DPBS and the particles
were pelleted by ultracentrifugation. In the lighter fractions of the su-
crose gradient (i.e., fractions 3–6), the PKH26-labeled exosome-free
Fig. 5. PKH26-labeled exosome-free controls and exosome samples prepared using pro- controls (Fig. 7a–e, con 1) and PKH26-labeled exosome samples
cedure C. (Fig. 7a–e, exo 1) contained low and similar numbers of PKH26-positive
(a–d) Representative confocal images of PKH26-positive particles in a PKH26-labeled particles with similar surface areas and fluorescence intensities (Fig. 7f,
exosome-free control (i.e., particle-free DPBS; con) (a) and in a PKH26-labeled exosome g). However, in the denser fractions of the sucrose gradient (i.e., frac-
(exo) (b) sample prepared following procedure C. Inset: Magnified selected PKH26-po-
tions 7–10), many PKH26-positive particles were observed in the
sitive particle. Scale bars: 2 μm; inset, 0.4 μm. (c, d) Black and white image masks from
(a), (b) after image segmentation for individual PKH26-positive particles, as exosome-free
PKH26-labeled exosome samples (Fig. 7h-l, exo 2), while only a few
control (c; 40 particles) and exosome (d; 337 particles) samples, respectively. Scale bars: PKH26-positive particles were seen in the exosome-free controls
2 μm. (e–g) Quantification of the PKH26-positive particles per image field of 2030 μm2, as (Fig. 7h-l, con 2). In the PKH26-labeled exosome-free control, the
the numbers (e; N°), surface areas (f; S), and fluorescence intensities (g; Int.). Data are number of PKH26-positive particles per image was one-tenth than of
means ± SEM. Numbers (bottom, e) indicate confocal images analyzed, whereas num- the PKH26-labeled exosome sample, as 1 ± 0.3 (n = 19) versus
bers (top, f) indicate PKH26-positive particles analyzed in f and g. ***, p < 0.001 versus
10 ± 2 (n = 20), respectively (Fig. 7l). Again, the PKH26-positive
control (con; Mann-Whitney U tests).
particles in both samples had comparable surface areas and fluores-
cence intensities (Fig. 7m, n), which were similar to those reported for
labeled exosome samples were similar (0.79 × 1010 particles/mL vs. procedure C (Fig. 5f, g). The PKH26-positive particles that were sepa-
1.13 × 1010 particles/mL, respectively; Table 1), the population of rated into the lighter fractions thus appear to represent PKH26 nano-
larger particles was enriched in the PKH26-labeled exosome samples particles, while those separated into the denser fractions appear to re-
compared to the PKH26-labeled exosome-free controls (Fig. 6b). Ad- present pure PKH26-labeled exosomes. Samples prepared using
ditionally, the recovery of unstained exosomes processed using sucrose- procedure D were further analyzed by AF4-MALS, although as the
cushion-based staining of procedure C was calculated from the AF4- concentration of PKH26-positive particles was below the detection limit
MALS measured number densities of exosomes before (theoretical yield; of the instrument, no further analyses were performed.
3.85 × 1010 particles/mL, Table 1) and after (experimental yield; Altogether, these data indicate that procedure D, which includes
0.39 × 1010 particles/mL, data not shown) the processing, to be ~10%. separation on a sucrose gradient and pelleting of the PKH26-positive
Thus when staining 20 μg of exosomes (corresponding to 19.25 × 1012 particles by ultracentrifugation, can be used to efficiently separate
exosomes) using procedure C (Fig. 1), we expect to obtain approxi- PKH26-labeled exosomes from PKH26 nanoparticles, although this is
mately 1.9 × 1012 exosomes in the PKH26-labeled exosome samples. As achieved at the expense of low PKH26-labeled exosome recovery.
we detected 5.65 × 1012 particles (1.13 × 1010 particles/mL in 50 μL;
Table 1) in those samples, the exosomes appear to represent only 34%

1356
P. Pužar Dominkuš et al. BBA - Biomembranes 1860 (2018) 1350–1361

Fig. 6. AF4-MALS analysis of PKH26-labeled exosome-free and exosome samples prepared using procedure C.
Normalized AF4-MALS fractograms of the PKH26-labeling solution (PKH26), PKH26-labeled exosome-free control (con-procedure C; red), unstained exosomes (exo; black) and PKH26-
labeled exosome sample (exo-procedure C; blue) for Rrms (a) and number density (b). Samples were stained following procedure C (Fig. 1). Continuous curves represent normalized light
scattering signal, while empty circles represent Rrms (a) and number density (b).

3.5. PHK26 nanoparticles are internalized into cultured astrocytes fluorescent puncta in the cell cytoplasm. After 3 h co-incubation of the
cells with PKH26 nanoparticles (con) and PKH26-positive particles of
Characterization of PKH26-positive particles in PKH26-labeled the exosome-containing samples (exo) at 37 °C, the fluorescent com-
samples revealed that PKH26 nanoparticles are present in the exosome- partments per cell were 127 ± 24 (n = 12) and 662 ± 78 (n = 12),
free controls, whereas a mix of PKH26 nanoparticles and PKH26-la- respectively (Fig. 8c). This 5.2-fold difference in the number of in-
beled exosomes are present in the PKH26-labeled exosome-containing tracellular fluorescent compartments across these two sets of labeled
samples. To investigate the physiological relevance of contaminating cells closely mirrors the ~7-fold difference in the number of PKH26-
PKH26 nanoparticles in exosome cell targeting, we tested whether positive particles in the PKH26-labeled exosome-free controls and the
cultured astrocytes can pick-up and internalize PKH26 nanoparticles PKH26-labeled exosome samples loaded on the astrocytes. Interest-
obtained using procedure C (con) in a way similar to that seen for ingly, the mean volume of these fluorescent compartments that picked-
PKH26-positive particles present in the PKH26-labeled exosome-con- up the PKH26 nanoparticles (con) was ~76% that of the compartments
taining samples (exo). Confocal microscopy of live cells (z-stacked) that picked-up the PKH26-positive particles of the exosome-containing
revealed that both PKH26 nanoparticles (Fig. 8a) and PKH26-positive sample (exo; Fig. 8d), whereas the mean compartment fluorescence
particles of the exosome-containing samples (Fig. 8b) localized to si- intensity was similar in these cells (Fig. 8e).
milar subcellular compartments, and were visible as numerous These intracellular fluorescent compartments are likely endocytotic

Fig. 7. Purified PKH26-labeled exosome-free


controls and exosome samples prepared using
procedure D.
(a–d, h–k) Representative confocal images of
PKH26-positive particles isolated from the lighter
(fractions 3–6) (a, b) and denser (fractions 7–10)
(h, i) sucrose gradient fractions after separation of
PKH26-labeled exosome-free control (a, con 1; h,
con 2) and PKH26-labeled exosome (b, exo 1; i,
exo 2) samples. Inset: Magnified selected PKH26-
positive particle. Scale bars: 2 μm; inset, 0.4 μm.
(c, d, j, k) Black and white image masks from (a),
(b), (h), (i) after image segmentation for in-
dividual PKH26-positive particles (encircled) as
exosome-free control (c, j) and exosome (d, k)
samples. Scale bars: 2 μm. (e–g, l–n)
Quantification of the PKH26-positive particles per
image field of 2030 μm2, as the numbers (e, l; N°),
surface areas (f, m; S), and fluorescence in-
tensities (g, n; Int.). Data are means ± SEM.
Numbers (bottom, e and l) indicate confocal
images analyzed, whereas numbers (bottom, f
and m) indicate PKH26-positive particles ana-
lyzed in f and g or m and n, respectively. ***,
p < 0.001 versus control (con; Mann-Whitney U
tests).

1357
P. Pužar Dominkuš et al. BBA - Biomembranes 1860 (2018) 1350–1361

localization was observed between the PKH26-labeled fluorescent


compartments and the dextran-laden vesicles (Fig. 8f, g). These data
indicate that the PKH26 nanoparticles present in the PKH26-labeled
exosome-free controls internalized into similar astrocytic compartments
as the mix of PKH26 nanoparticles and PKH26-labeled exosomes pre-
sent in PKH26-labeled exosome-containing samples.

4. Discussion

Despite the wide use of PKH-labeled EVs in cell targeting and


functional studies [13,14], the nonEV-associated fluorescent structures
detected by Lai et al. [46] have never been systematically character-
ized, and their internalization by cells has never been evaluated. Here,
we have shown for the first time that numerous PKH26 nanoparticles
are formed during ultracentrifugation-based exosome staining, and that
these are almost indistinguishable from PKH26-labeled exosomes in
terms of their size, surface area, and fluorescence intensity, as shown by
confocal imaging and AF4-MALS analysis. When PKH26-labeled exo-
some samples were purified through sucrose, the PKH26 nanoparticles
were differentiated from the PKH26-labeled exosomes based on their
reduced size. However, these PKH26 nanoparticles can only be physi-
cally removed from the PKH26-labeled exosome samples when they are
separated on a sucrose gradient. Of note, this is achieved at the expense
of low PKH26-labeled exosome recovery. Importantly, PKH26 nano-
particles can also be internalized into primary astrocytes into similar
subcellular compartments as seen for PKH26-positive particles of the
PKH26-labeled exosome-containing samples.
Regardless of the staining procedure used, PKH26-positive particles
were detected in the PKH26-labeled exosome-free controls, although to
different extents. Around 0.75 × 1010 PKH26 particles/mL were de-
tected in the PKH26-labeled exosome-free controls after ultra-
centrifugation-based staining of procedure A, as determined using AF4-
MALS. Similar levels were detected even after purification through a
sucrose cushion (0.79 × 1010 PKH26 particles/mL) when following
procedure C. In comparison, the particle numbers were below the de-
tection limit of the AF4-MALS analysis following purification of the
PKH26-positive particles of the exosome-free control on a sucrose
gradient according to procedure D, as they were greatly decreased (by
~97% as assessed by confocal imaging). Importantly, the sizes (i.e.,
Rrms, Rgeom), surface areas, and fluorescence intensities of the PKH26-
positive particles in the exosome-free control samples were mostly si-
milar to the characteristics of the PKH26-positive particles in the exo-
some-containing samples. Of interest, the PKH26-positive particles in
the ultracentrifugation-based PKH26-labeled exosome-free controls of
procedure A showed a broad size distribution (Rrms, 20–100 nm; mean
Fig. 8. Internalization of PKH26 nanoparticles and PKH26-positive particles of the exo-
Rrms [Rgeom], 77 [84] nm), which mirrored the particle sizes in the
some-containing samples into subcellular compartments of cultured astrocytes.
(a, b) Representative three-dimensional shaded display of individual live cultured as- PKH26-labeled exosome samples. Only when the PKH26-positive par-
trocytes that internalized PKH26 nanoparticles (a, con) and PKH26-positive particles ticles were purified through the hyperosmotic sucrose cushion of pro-
present in the PKH26-labeled exosome-containing samples (b, exo) into intracellular cedure C did those in the exosome-free control samples become more
compartments, observed as numerous bright fluorescent puncta. Scale bars, 10 μm. (c–e) homogenous in size, with a smaller mean Rrms (Rgeom) of 65 (70) nm.
Quantification of the number (c; N°), volume (d; V), and fluorescence intensities (e; Int.) This will probably be due to their loss of water [59]. All of these details
of compartments (Comp.) that internalized PKH26 nanoparticles (con) and PKH26-posi-
indicate that the PKH26 nanoparticles in the exosome-free control
tive particles of the exosome-containing samples (exo) per cell. Data are means ± SEM.
Numbers (bottom, c) indicate cells analyzed, whereas numbers (top, d) indicate PKH26- samples could represent PKH26 micelles, which in general cannot be
positive compartments analyzed in d and e. ***, p < 0.001 versus control (con; Mann- differentiated from the PKH26-labeled exosomes in terms of the char-
Whitney U tests). (f-g) Confocal micrographs of double-labeled live astrocytes that in- acteristics that were measured here.
ternalized PKH26 nanoparticles (f, con, left) or PKH26-positive particles present in the PKH26 nanoparticles were also detected in the PKH26-labeled
PKH26-labeled exosome-containing samples (g, exo, left) and fluorescent dextran (Dex, exosome samples regardless of the staining procedure used. For the
middle) into the common subcellular compartments as revealed by white mask images
ultracentrifugation-based staining of procedure A, the PKH26-labeled
displaying co-localized pixels (Mask, right). Scale bars: 10 μm.
exosomes represented only 11% of all of the PKH26-positive particles
detected in the PKH26-labeled exosome samples, with the rest re-
vesicles, as we observed a pattern of internalization that is typical for presenting PKH26 nanoparticles. Thus, there were 13-fold more PKH26
normal membrane recycling. To test this, we microscopically examined nanoparticles in the PKH26-labeled exosome samples compared to the
cells that were simultaneously exposed either to PKH26 nanoparticles corresponding exosome-free control for the ultracentrifugation based
or to PKH26-positive particles present in the PKH26-labeled exosome- procedure A. This implies that the PKH26 nanoparticle numbers in the
containing sample, and to 20 μM fluorescent dextran. After 3 h co-in- PKH26-labeled exosome samples cannot simply be inferred from their
cubation of cells with aforementioned probes, ample fluorescence co- numbers in the corresponding PKH26-labeled exosome-free control

1358
P. Pužar Dominkuš et al. BBA - Biomembranes 1860 (2018) 1350–1361

samples. Similarly we showed that for the sucrose-cushion-based procedure A and C, respectively. Thus PKH26-labeled exosome samples
staining of procedure C, exosomes appear to represent 34% of all of the are mostly composed of PKH26 nanoparticles. Consequently, it is cru-
PKH26-positive particles in the PKH26-labeled exosome samples, with cial to remove PKH26 nanoparticles from PKH26-labeled exosomes
the rest of these representing PKH26 nanoparticles. Thus comparable before performing cell targeting and internalization studies.
numbers of PKH26 nanoparticles are present in the PKH26-labeled At present, we can only speculate on the mechanisms of the cellular
exosome samples and exosome-free controls for the sucrose-cushion- uptake of PKH26 nanoparticles. EVs can be taken up by cells through a
based procedure C. Contrary to previously published data [60,61], low variety of receptor-dependent pathways, or they can fuse with or just
PKH26-positive particle recovery was characteristic of the filtration- bind to the plasma membrane [40–42]. Importantly, EVs can also be
based exosome staining of procedure B for both the exosome-free internalized through receptor-independent mechanisms, like macro-
controls and exosome-containing samples. This discrepancy might be pinocytosis, where extracellular fluid and its components are enveloped
due to the nature of the filtration membranes, as a concentrating device by an invagination and there is fusion of the membrane ruffles, to form
that contains a polyethersulfone membrane can reduce EV yield by at a vesicular bud that pinches off into the cytosol [66]. This was sup-
least two-thirds compared to one that contains a cellulose membrane ported by Fitzner et al. [67], who showed that sucrose-gradient-purified
[21,61]. The lower propensity of the PKH26 dye to form particles when PKH67-labeled exosomes from oligodendrocytes are selectively trans-
using non-sedimentation processes for staining might also contribute ferred to microglia by macropinocytosis. Macropinocytosis can be
here. Importantly, by using the sucrose gradient of procedure D it was constitutive, as is found in microglia [67,68], macrophages [69], and
possible to separate PKH26 nanoparticles from PKH26-labeled exo- dendritic [70] and cancer cells [68,71], or it can be growth-factor-ac-
somes in the PKH26-labeled exosome samples. However, the separation tivated. Another study postulated that this process is present in most
is achieved at the expense of < 2% recovery of the PKH26-labeled cells, with the exception of the brain microvessel endothelial cells, and
exosomes, as determined here by confocal imaging. Similarly, a study potentially some other [72]. Importantly, macropinocytosis is also one
by Van Deun et al. [62] showed that about two-thirds fewer exosomes of the pathways that can be exploited for cellular uptake of nanome-
were recovered by an iodixanol-gradient-based isolation procedure dicines, including polymeric micelles [72]. Thus we can assume that
compared to ultracentrifugation. PKH26 nanoparticles can be taken up by cells through macro-
The present data support the sucrose-gradient-based staining of pinocytosis, a process that can be promoted further by the strong
procedure D as the method of choice to obtain PKH26-labeled exosomes electrostatic interaction between the positively charged carbocyanine
that are devoid of PKH26 nanoparticles for use in EV uptake and (PKH) dye and the negatively charged plasma membrane, as was shown
functional studies. As PKH26 nanoparticles only contain buffer, they to be the case for cationic nanoparticles [72,73].
have lower buoyance density in comparison to PKH26-labeled exo- Interestingly, a recent study showed that Lipofectamine, which is a
somes, and therefore they can be separated into lighter fractions of a reagent composed of lipid subunits that can form cationic liposomes,
sucrose gradient. The PKH26-labeled exosomes separated most promi- can increase internalization of exosomes containing tau seeds through
nently into fraction 9, with a density of 1.21 g/mL, which is at the edge induction of the binding of EVs and formation of EV clusters [74]. In
of the density interval given previously as 1.08 g/mL to 1.22 g/mL another study, exosomes released from HIV-infected cells were shown
[15,57,58], which is typical for unstained exosomes. In other studies, to render resting CD4+ T lymphocytes permissive to virus internaliza-
stained EVs floated at sucrose densities of 1.11 g/mL to 1.19 g/mL tion and infection [36,75]. Similarly, the PKH26 nanoparticles in
[15,63]; however, smaller EV populations might have been missed PKH26-labeled exosome samples might promote cellular uptake of
here, as the detection limit of flow cytometry is 100 nm in diameter. PKH26-labeled exosomes, through changing the exosome internaliza-
Interestingly, there was no severe effect of sucrose on exosome size, tion characteristics. In addition to internalization of PKH26-positive
with the mean Rrms (Rgeom) and surface areas of the PKH26-positive particles into astrocytic subcellular compartments, the cellular fluor-
particles in the PKH26-labeled exosome samples remaining at 81 (93) escent patterns observed in the present study might also be the result of
nm and 0.14 μm2, respectively. Although some concerns have been nonspecific leakage of the dye molecules from PKH26-positive particles
expressed about the effects of sucrose on the biological functions of EVs, to the plasma membrane, and subsequently to cellular membranes, as
Zech et al. [64] did not observe impairment of leukocyte effector this would lead to a pattern of internalization similar to that of normal
functions for sucrose-gradient-enriched compared to 0.2-μm-filtered membrane recycling [42]. However, several studies have shown that
exosomes from a pancreatic adenocarcinoma. As determined by several passive dye transfer is probably a minor process, as fixed microglia did
biochemical and biophysical techniques, use of sucrose and iodixanol not stain with PKH67 [67] and low temperature and/or excess un-
gradients also represents an efficient way to obtain highly pure exo- labeled exosomes reduced the uptake of glioblastoma cells [76], en-
somes from serum, and these showed higher biological activity in terms dothelial cells [44], and bladder cancer cells [14].
of NFκB nuclear translocation compared to ultracentrifugation-based Altogether, PKH26 staining of EVs can be an informative tool, but
and PEG-based exosome preparations [44]. Importantly, lentiviruses, only when an appropriate isolation method is used to remove PKH26
like HIV, and lentiviral vectors, can also be purified by ultra- nanoparticles, and rigorous controls are used to evaluate EV uptake by
centrifugation through a sucrose cushion for further use in in vitro and target cells. Our observations about the characteristics of the PKH26
in vivo infection studies [54]. dye can be extended to the PKH67 dye, as shown here by us (Appendix
The presence of PKH26 nanoparticles in PKH26-labeled EV samples A), and likely to the other lipophilic long-chain carbocyanines like DiO
has never been accounted for in previous EV functional studies. and Dil, as PKH26 is structurally identical to DilC18 [77]. The char-
Although they do not carry any functional molecules, PKH26 nano- acteristics of alternative membrane dyes used for staining of EVs, like
particles might contribute to the labeling of target cells and affect es- phospholipid dye rhodamine-DHPE [78] and styryl dye FM 1–43 [79],
timation of the cellular uptake of PKH26-labeled exosomes. Here, we would need to be further studied.
have shown that PKH26 nanoparticles are indeed internalized into
primary astrocytes in similar compartments as seen for PKH26-positive Competing interests
particles of the PKH26-labeled exosome-containing samples. Ample
fluorescence co-localization between the PKH26-laden fluorescent The authors declare no conflict of interest and funding.
compartments and the dextran-laden vesicles revealed that PKH26- Author contributions
positive particles likely internalized into astrocytic endo-/lysosomes Conceived and designed the experiments: PPD, MS, RZ, AP, EŽ, MK,
[65]. This observation is of great importance, as only one or three out of ML.
10 PKH26-positive particles actually represent PKH26-labeled exo- Performed the experiments: PPD, MS, SS, EL.
somes in the PKH26-labeled exosome samples stained following Analyzed the data: MS, SS, EL, ML.

1359
P. Pužar Dominkuš et al. BBA - Biomembranes 1860 (2018) 1350–1361

Wrote the paper: PPD, MS, SS, EL, RZ, AP, EŽ, MK, ML. exosomes contain RNA: uptake by macrophages, J. Transl. Med. 9 (2011) 9.
[22] A. Asea, C. Jean-Pierre, P. Kaur, P. Rao, I.M. Linhares, D. Skupski, S.S. Witkin, Heat
shock protein-containing exosomes in mid-trimester amniotic fluids, J. Reprod.
Transparency document Immunol. 79 (2008) 12–17.
[23] C. Admyre, J. Grunewald, J. Thyberg, S. Gripenback, G. Tornling, A. Eklund,
The http://dx.doi.org/10.1016/j.bbamem.2018.03.013 associated A. Scheynius, S. Gabrielsson, Exosomes with major histocompatibility complex class
II and co-stimulatory molecules are present in human BAL fluid, Eur. Respir. J. 22
this article can be found, in online version. (2003) 578–583.
[24] K. Skriner, K. Adolph, P.R. Jungblut, G.R. Burmester, Association of citrullinated
Acknowledgments proteins with synovial exosomes, Arthritis Rheum. 54 (2006) 3809–3814.
[25] J.C. Gross, V. Chaudhary, K. Bartscherer, M. Boutros, Active Wnt proteins are se-
creted on exosomes, Nat. Cell Biol. 14 (2012) 1036–1045.
This work was supported by the Slovenian Research Agency (ARRS) [26] G. Lachenal, K. Pernet-Gallay, M. Chivet, F.J. Hemming, A. Belly, G. Bodon, B. Blot,
[research grants J3-5499, P1-170, P3-310, and P2-0145]. G. Haase, Y. Goldberg, R. Sadoul, Release of exosomes from differentiated neurons
and its regulation by synaptic glutamatergic activity, Mol. Cell. Neurosci. 46 (2011)
409–418.
Appendix A. Supplementary data [27] J. Conde-Vancells, E. Rodriguez-Suarez, N. Embade, D. Gil, R. Matthiesen, M. Valle,
F. Elortza, S.C. Lu, J.M. Mato, J.M. Falcon-Perez, Characterization and compre-
Supplementary data to this article can be found online at https:// hensive proteome profiling of exosomes secreted by hepatocytes, J. Proteome Res. 7
(2008) 5157–5166.
doi.org/10.1016/j.bbamem.2018.03.013. [28] J. Conde-Vancells, E. Gonzalez, S.C. Lu, J.M. Mato, J.M. Falcon-Perez, Overview of
extracellular microvesicles in drug metabolism, Expert Opin. Drug Metab. Toxicol.
References 6 (2010) 543–554.
[29] A.E. Morelli, A.T. Larregina, W.J. Shufesky, M.L. Sullivan, D.B. Stolz,
G.D. Papworth, A.F. Zahorchak, A.J. Logar, Z. Wang, S.C. Watkins, L.D. Falo Jr.,
[1] P.K. Wallace, J.D. Tario Jr., J.L. Fisher, S.S. Wallace, M.S. Ernstoff, K.A. Muirhead, A.W. Thomson, Endocytosis, intracellular sorting, and processing of exosomes by
Tracking antigen-driven responses by flow cytometry: monitoring proliferation by dendritic cells, Blood 104 (2004) 3257–3266.
dye dilution, Cytometry A 73 (2008) 1019–1034. [30] P.D. Robbins, A.E. Morelli, Regulation of immune responses by extracellular ve-
[2] F. Progatzky, M.J. Dallman, C. Lo Celso, From seeing to believing: labelling stra- sicles, Nat. Rev. Immunol. 14 (2014) 195–208.
tegies for in vivo cell-tracking experiments, Interface Focus 3 (2013) 20130001. [31] A. Janowska-Wieczorek, M. Wysoczynski, J. Kijowski, L. Marquez-Curtis,
[3] H.E. Ghoneim, P.G. Thomas, J.A. McCullers, Depletion of alveolar macrophages B. Machalinski, J. Ratajczak, M.Z. Ratajczak, Microvesicles derived from activated
during influenza infection facilitates bacterial superinfections, J. Immunol. 191 platelets induce metastasis and angiogenesis in lung cancer, Int. J. Cancer 113
(2013) 1250–1259. (2005) 752–760.
[4] M.E. Sheehy, A.B. McDermott, S.N. Furlan, P. Klenerman, D.F. Nixon, A novel [32] J. Skog, T. Wurdinger, S. van Rijn, D.H. Meijer, L. Gainche, M. Sena-Esteves,
technique for the fluorometric assessment of T lymphocyte antigen specific lysis, J. W.T. Curry Jr., B.S. Carter, A.M. Krichevsky, X.O. Breakefield, Glioblastoma mi-
Immunol. Methods 249 (2001) 99–110. crovesicles transport RNA and proteins that promote tumour growth and provide
[5] C.R. Parish, Fluorescent dyes for lymphocyte migration and proliferation studies, diagnostic biomarkers, Nat. Cell Biol. 10 (2008) 1470–1476.
Immunol. Cell Biol. 77 (1999) 499–508. [33] R. Ji, B. Zhang, X. Zhang, J. Xue, X. Yuan, Y. Yan, M. Wang, W. Zhu, H. Qian, W. Xu,
[6] S.A. Bapat, A. Krishnan, A.D. Ghanate, A.P. Kusumbe, R.S. Kalra, Gene expression: Exosomes derived from human mesenchymal stem cells confer drug resistance in
protein interaction systems network modeling identifies transformation-associated gastric cancer, Cell Cycle 14 (2015) 2473–2483.
molecules and pathways in ovarian cancer, Cancer Res. 70 (2010) 4809–4819. [34] E. Emmanouilidou, K. Melachroinou, T. Roumeliotis, S.D. Garbis, M. Ntzouni,
[7] M. Schubert, N. Herbert, I. Taubert, D. Ran, R. Singh, V. Eckstein, M. Vitacolonna, L.H. Margaritis, L. Stefanis, K. Vekrellis, Cell-produced alpha-synuclein is secreted
A.D. Ho, M. Zoller, Differential survival of AML subpopulations in NOD/SCID mice, in a calcium-dependent manner by exosomes and impacts neuronal survival, J.
Exp. Hematol. 39 (2011) 250–263 (e254). Neurosci. 30 (2010) 6838–6851.
[8] P.J. Hendrikx, C.M. Martens, A. Hagenbeek, J.F. Keij, J.W. Visser, Homing of [35] B. Fevrier, D. Vilette, F. Archer, D. Loew, W. Faigle, M. Vidal, H. Laude, G. Raposo,
fluorescently labeled murine hematopoietic stem cells, Exp. Hematol. 24 (1996) Cells release prions in association with exosomes, Proc. Natl. Acad. Sci. U. S. A. 101
129–140. (2004) 9683–9688.
[9] F. Lassailly, E. Griessinger, D. Bonnet, “Microenvironmental contaminations”s in- [36] M. Lenassi, G. Cagney, M. Liao, T. Vaupotic, K. Bartholomeeusen, Y. Cheng,
duced by fluorescent lipophilic dyes used for noninvasive in vitro and in vivo cell N.J. Krogan, A. Plemenitas, B.M. Peterlin, HIV Nef is secreted in exosomes and
tracking, Blood 115 (2010) 5347–5354. triggers apoptosis in bystander CD4+ T cells, Traffic 11 (2010) 110–122.
[10] P. Li, R. Zhang, H. Sun, L. Chen, F. Liu, C. Yao, M. Du, X. Jiang, PKH26 can transfer [37] L. Rajendran, M. Honsho, T.R. Zahn, P. Keller, K.D. Geiger, P. Verkade, K. Simons,
to host cells in vitro and vivo, Stem Cells Dev. 22 (2013) 340–344. Alzheimer's disease beta-amyloid peptides are released in association with exo-
[11] W.E. Samlowski, B.A. Robertson, B.K. Draper, E. Prystas, J.R. McGregor, Effects of somes, Proc. Natl. Acad. Sci. U. S. A. 103 (2006) 11172–11177.
supravital fluorochromes used to analyze the in vivo homing of murine lympho- [38] M.P. Zaborowski, L. Balaj, X.O. Breakefield, C.P. Lai, Extracellular vesicles: com-
cytes on cellular function, J. Immunol. Methods 144 (1991) 101–115. position, biological relevance, and methods of study, Bioscience 65 (2015)
[12] D.J. Oh, G.M. Lee, K. Francis, B.O. Palsson, Phototoxicity of the fluorescent mem- 783–797.
brane dyes PKH2 and PKH26 on the human hematopoietic KG1a progenitor cell [39] E. van der Pol, F.A. Coumans, A.E. Grootemaat, C. Gardiner, I.L. Sargent,
line, Cytometry 36 (1999) 312–318. P. Harrison, A. Sturk, T.G. van Leeuwen, R. Nieuwland, Particle size distribution of
[13] A. Riches, E. Campbell, E. Borger, S. Powis, Regulation of exosome release from exosomes and microvesicles determined by transmission electron microscopy, flow
mammary epithelial and breast cancer cells - a new regulatory pathway, Eur. J. cytometry, nanoparticle tracking analysis, and resistive pulse sensing, J. Thromb.
Cancer 50 (2014) 1025–1034. Haemost. 12 (2014) 1182–1192.
[14] C.A. Franzen, P.E. Simms, A.F. Van Huis, K.E. Foreman, P.C. Kuo, G.N. Gupta, [40] D.D. Taylor, C. Gercel-Taylor, The origin, function, and diagnostic potential of RNA
Characterization of uptake and internalization of exosomes by bladder cancer cells, within extracellular vesicles present in human biological fluids, Front. Genet. 4
Biomed. Res. Int. 2014 (2014) 619829. (2013) 142.
[15] C. Thery, S. Amigorena, G. Raposo, A. Clayton, Isolation and characterization of [41] A. Montecalvo, A.T. Larregina, W.J. Shufesky, D.B. Stolz, M.L. Sullivan,
exosomes from cell culture supernatants and biological fluids, Current Protocols in J.M. Karlsson, C.J. Baty, G.A. Gibson, G. Erdos, Z. Wang, J. Milosevic,
Cell Biology/Editorial Board (2006) Juan S. Bonifacino ..., 3 22 (Chapter 3 (2006) O.A. Tkacheva, S.J. Divito, R. Jordan, J. Lyons-Weiler, S.C. Watkins, A.E. Morelli,
Unit). Mechanism of transfer of functional microRNAs between mouse dendritic cells via
[16] N. Arraud, R. Linares, S. Tan, C. Gounou, J.M. Pasquet, S. Mornet, A.R. Brisson, exosomes, Blood 119 (2012) 756–766.
Extracellular vesicles from blood plasma: determination of their morphology, size, [42] L.A. Mulcahy, R.C. Pink, D.R. Carter, Routes and mechanisms of extracellular ve-
phenotype and concentration, J. Thromb. Haemost. 12 (2014) 614–627. sicle uptake, J Extracell Vesicles 3 (2014).
[17] M.G. Harrington, A.N. Fonteh, E. Oborina, P. Liao, R.P. Cowan, G. McComb, [43] C. Barres, L. Blanc, P. Bette-Bobillo, S. Andre, R. Mamoun, H.J. Gabius, M. Vidal,
J.N. Chavez, J. Rush, R.G. Biringer, A.F. Huhmer, The morphology and biochem- Galectin-5 is bound onto the surface of rat reticulocyte exosomes and modulates
istry of nanostructures provide evidence for synthesis and signaling functions in vesicle uptake by macrophages, Blood 115 (2010) 696–705.
human cerebrospinal fluid, Cerebrospinal Fluid Res. 6 (2009) 10. [44] G. Di Noto, M. Chiarini, L. Paolini, E.L. Mazzoldi, V. Giustini, A. Radeghieri,
[18] T. Pisitkun, R.F. Shen, M.A. Knepper, Identification and proteomic profiling of L. Caimi, D. Ricotta, Immunoglobulin free light chains and GAGs mediate multiple
exosomes in human urine, Proc. Natl. Acad. Sci. U. S. A. 101 (2004) 13368–13373. myeloma extracellular vesicles uptake and secondary NfkappaB nuclear transloca-
[19] I. Lozano-Ramos, I. Bancu, A. Oliveira-Tercero, M.P. Armengol, A. Menezes-Neto, tion, Front. Immunol. 5 (2014) 517.
H.A. Del Portillo, R. Lauzurica-Valdemoros, F.E. Borras, Size-exclusion chromato- [45] E.J. van der Vlist, E.N. Nolte-'t Hoen, W. Stoorvogel, G.J. Arkesteijn, M.H. Wauben,
graphy-based enrichment of extracellular vesicles from urine samples, J Extracell Fluorescent labeling of nano-sized vesicles released by cells and subsequent quan-
Vesicles 4 (2015) 27369. titative and qualitative analysis by high-resolution flow cytometry, Nat. Protoc. 7
[20] A. Poliakov, M. Spilman, T. Dokland, C.L. Amling, J.A. Mobley, Structural hetero- (2012) 1311–1326.
geneity and protein composition of exosome-like vesicles (prostasomes) in human [46] C.P. Lai, E.Y. Kim, C.E. Badr, R. Weissleder, T.R. Mempel, B.A. Tannous,
semen, Prostate 69 (2009) 159–167. X.O. Breakefield, Visualization and tracking of tumour extracellular vesicle delivery
[21] C. Lasser, V.S. Alikhani, K. Ekstrom, M. Eldh, P.T. Paredes, A. Bossios, M. Sjostrand, and RNA translation using multiplexed reporters, Nat. Commun. 6 (2015) 7029.
S. Gabrielsson, J. Lotvall, H. Valadi, Human saliva, plasma and breast milk [47] E. Pawelczyk, E.K. Jordan, A. Balakumaran, A. Chaudhry, N. Gormley, M. Smith,

1360
P. Pužar Dominkuš et al. BBA - Biomembranes 1860 (2018) 1350–1361

B.K. Lewis, R. Childs, P.G. Robey, J.A. Frank, In vivo transfer of intracellular labels M. Faiz, M. Pekny, R. Zorec, IFN-gamma-induced increase in the mobility of MHC
from locally implanted bone marrow stromal cells to resident tissue macrophages, class II compartments in astrocytes depends on intermediate filaments, J.
PLoS One 4 (2009) e6712. Neuroinflammation 9 (2012) 144.
[48] B. Sumer, J. Gao, Theranostic nanomedicine for cancer, Nanomedicine (London) 3 [66] G.J. Doherty, H.T. McMahon, Mechanisms of endocytosis, Annu. Rev. Biochem. 78
(2008) 137–140. (2009) 857–902.
[49] T. Katsuda, R. Tsuchiya, N. Kosaka, Y. Yoshioka, K. Takagaki, K. Oki, F. Takeshita, [67] D. Fitzner, M. Schnaars, D. van Rossum, G. Krishnamoorthy, P. Dibaj, M. Bakhti,
Y. Sakai, M. Kuroda, T. Ochiya, Human adipose tissue-derived mesenchymal stem T. Regen, U.K. Hanisch, M. Simons, Selective transfer of exosomes from oligoden-
cells secrete functional neprilysin-bound exosomes, Sci. Rep. 3 (2013) 1197. drocytes to microglia by macropinocytosis, J. Cell Sci. 124 (2011) 447–458.
[50] N. Kol, M. Tsvitov, L. Hevroni, S.G. Wolf, H.B. Pang, M.S. Kay, I. Rousso, The effect [68] K. Yuyama, H. Sun, S. Mitsutake, Y. Igarashi, Sphingolipid-modulated exosome
of purification method on the completeness of the immature HIV-1 gag shell, J. secretion promotes clearance of amyloid-beta by microglia, J. Biol. Chem. 287
Virol. Methods 169 (2010) 244–247. (2012) 10977–10989.
[51] C. Gardiner, Y.J. Ferreira, R.A. Dragovic, C.W. Redman, I.L. Sargent, Extracellular [69] D. Feng, W.L. Zhao, Y.Y. Ye, X.C. Bai, R.Q. Liu, L.F. Chang, Q. Zhou, S.F. Sui,
vesicle sizing and enumeration by nanoparticle tracking analysis, J. Extracellular Cellular internalization of exosomes occurs through phagocytosis, Traffic 11 (2010)
Vesicles 2 (2013). 675–687.
[52] J.P. Schwartz, D.J. Wilson, Preparation and characterization of type 1 astrocytes [70] Z. Liu, P.A. Roche, Macropinocytosis in phagocytes: regulation of MHC class-II-re-
cultured from adult rat cortex, cerebellum, and striatum, Glia 5 (1992) 75–80. stricted antigen presentation in dendritic cells, Front. Physiol. 6 (2015) 1.
[53] S. Sitar, A. Kejzar, D. Pahovnik, K. Kogej, M. Tusek-Znidaric, M. Lenassi, E. Zagar, [71] C. Escrevente, S. Keller, P. Altevogt, J. Costa, Interaction and uptake of exosomes by
Size characterization and quantification of exosomes by asymmetrical-flow field- ovarian cancer cells, BMC Cancer 11 (2011) 108.
flow fractionation, Anal. Chem. 87 (2015) 9225–9233. [72] L. Kou, J. Sun, Y. Zhai, Z. He, The endocytosis and intracellular fate of nanome-
[54] G. Tiscornia, O. Singer, I.M. Verma, Production and purification of lentiviral vec- dicines: implication for rational design, Asian J. Pharm. Sci. 8 (2013) 1–10.
tors, Nat. Protoc. 1 (2006) 241–245. [73] O. Harush-Frenkel, E. Rozentur, S. Benita, Y. Altschuler, Surface charge of nano-
[55] R. Cantin, J. Diou, D. Belanger, A.M. Tremblay, C. Gilbert, Discrimination between particles determines their endocytic and transcytotic pathway in polarized MDCK
exosomes and HIV-1: purification of both vesicles from cell-free supernatants, J. cells, Biomacromolecules 9 (2008) 435–443.
Immunol. Methods 338 (2008) 21–30. [74] J.C. Polanco, B.J. Scicluna, A.F. Hill, J. Gotz, Extracellular vesicles isolated from the
[56] M. Dettenhofer, X.F. Yu, Highly purified human immunodeficiency virus type 1 brains of rTg4510 mice seed tau protein aggregation in a threshold-dependent
reveals a virtual absence of vif in virions, J. Virol. 73 (1999) 1460–1467. manner, J. Biol. Chem. 291 (2016) 12445–12466.
[57] G. Raposo, H.W. Nijman, W. Stoorvogel, R. Liejendekker, C.V. Harding, C.J. Melief, [75] C. Arenaccio, C. Chiozzini, S. Columba-Cabezas, F. Manfredi, E. Affabris, A. Baur,
H.J. Geuze, B lymphocytes secrete antigen-presenting vesicles, J. Exp. Med. 183 M. Federico, Exosomes from human immunodeficiency virus type 1 (HIV-1)-in-
(1996) 1161–1172. fected cells license quiescent CD4+ T lymphocytes to replicate HIV-1 through a
[58] C. Thery, L. Zitvogel, S. Amigorena, Exosomes: composition, biogenesis and func- Nef- and ADAM17-dependent mechanism, J. Virol. 88 (2014) 11529–11539.
tion, Nat. Rev. Immunol. 2 (2002) 569–579. [76] H.C. Christianson, K.J. Svensson, T.H. van Kuppevelt, J.P. Li, M. Belting, Cancer cell
[59] S. Hupfeld, H.H. Moen, D. Ausbacher, H. Haas, M. Brandl, Liposome fractionation exosomes depend on cell-surface heparan sulfate proteoglycans for their inter-
and size analysis by asymmetrical flow field-flow fractionation/multi-angle light nalization and functional activity, Proc. Natl. Acad. Sci. U. S. A. 110 (2013)
scattering: influence of ionic strength and osmotic pressure of the carrier liquid, 17380–17385.
Chem. Phys. Lipids 163 (2010) 141–147. [77] S. Tajbakhsh, E. Vivarelli, G. Cuselladeangelis, D. Rocancourt, M. Buckingham,
[60] H.G. Lamparski, A. Metha-Damani, J.Y. Yao, S. Patel, D.H. Hsu, C. Ruegg, J.B. Le G. Cossu, A population of myogenic cells derived from the mouse neural-tube,
Pecq, Production and characterization of clinical grade exosomes derived from Neuron 13 (1994) 813–821.
dendritic cells, J. Immunol. Methods 270 (2002) 211–226. [78] T.J. Smyth, J.S. Redzic, M.W. Graner, T.J. Anchordoquy, Examination of the spe-
[61] R.J. Lobb, M. Becker, S.W. Wen, C.S.F. Wong, A.P. Wiegmans, A. Leimgruber, cificity of tumor cell derived exosomes with tumor cells in vitro, Biochim. Biophys.
A. Moller, Optimized exosome isolation protocol for cell culture supernatant and Acta 1838 (2014) 2954–2965.
human plasma, J Extracell Vesicles 4 (2015). [79] A. Hoshino, B. Costa-Silva, T.L. Shen, G. Rodrigues, A. Hashimoto, M.T. Mark,
[62] J. Van Deun, P. Mestdagh, R. Sormunen, V. Cocquyt, K. Vermaelen, H. Molina, S. Kohsaka, A. Di Giannatale, S. Ceder, S. Singh, C. Williams, N. Soplop,
J. Vandesompele, M. Bracke, O. De Wever, A. Hendrix, The impact of disparate K. Uryu, L. Pharmer, T. King, L. Bojmar, A.E. Davies, Y. Ararso, T. Zhang, H. Zhang,
isolation methods for extracellular vesicles on downstream RNA profiling, J J. Hernandez, J.M. Weiss, V.D. Dumont-Cole, K. Kramer, L.H. Wexler,
Extracell Vesicles 3 (2014). A. Narendran, G.K. Schwartz, J.H. Healey, P. Sandstrom, K.J. Labori, E.H. Kure,
[63] I. Nazarenko, A.K. Rupp, P. Altevogt, Exosomes as a potential tool for a specific P.M. Grandgenett, M.A. Hollingsworth, M. de Sousa, S. Kaur, M. Jain, K. Mallya,
delivery of functional molecules, Methods Mol. Biol. 1049 (2013) 495–511. S.K. Batra, W.R. Jarnagin, M.S. Brady, O. Fodstad, V. Muller, K. Pantel, A.J. Minn,
[64] D. Zech, S. Rana, M.W. Buchler, M. Zoller, Tumor-exosomes and leukocyte acti- M.J. Bissell, B.A. Garcia, Y. Kang, V.K. Rajasekhar, C.M. Ghajar, I. Matei,
vation: an ambivalent crosstalk, Cell Commun. Signal. 10 (2012) 37. H. Peinado, J. Bromberg, D. Lyden, Tumour exosome integrins determine organo-
[65] N. Vardjan, M. Gabrijel, M. Potokar, U. Svajger, M. Kreft, M. Jeras, Y. de Pablo, tropic metastasis, Nature 527 (2015) 329.

1361

You might also like