You are on page 1of 9

Article

pubs.acs.org/ac

Colorimetric Nanoplasmonic Assay To Determine Purity and Titrate


Extracellular Vesicles
Daniele Maiolo,*,∥,† Lucia Paolini,‡ Giuseppe Di Noto,‡ Andrea Zendrini,‡ Debora Berti,§ Paolo Bergese,†
and Doris Ricotta‡

Chemistry for Technologies Laboratory and INSTM, Department of Mechanical and Industrial Engineering, University of Brescia,
via Branze 38, 25123 Brescia, Brescia, Italy

Department of Molecular and Translational Medicine, Faculty of Medicine, University of Brescia, 25123 Brescia, Brescia, Italy
§
Department of Chemistry “Ugo Schiff” and CSGI, University of Florence, via della Lastruccia 3, 50019 Sesto Fiorentino, Florence
See https://pubs.acs.org/sharingguidelines for options on how to legitimately share published articles.

Italy
Downloaded via UNIV NACIONAL DE COLOMBIA on March 31, 2019 at 04:58:13 (UTC).

*
S Supporting Information

ABSTRACT: Extracellular Vesicles (EVs) − cell secreted


vesicles that carry rich molecular information of the parental
cell and constitute an important mode of intercellular
communication − are becoming a primary topic in transla-
tional medicine. EVs (that comprise exosomes and micro-
vesicles/microparticles) have a size ranging from 40 nm to 1
μm and share several physicochemical proprieties, including
size, density, surface charge, and light interaction, with other
nano-objects present in body fluids, such as single and
aggregated proteins. This makes separation, titration, and
characterization of EVs challenging and time-consuming. Here
we present a cost-effective and fast colorimetric assay for
probing by eye protein contaminants and determine the concentration of EV preparations, which exploits the synergy between
colloidal gold nanoplasmonics, nanoparticle−protein corona, and nanoparticle−membrane interaction. The assay hits a limit of
detection of protein contaminants of 5 ng/μL and has a dynamic range of EV concentration ranging from 35 fM to 35 pM, which
matches the typical range of EV concentration in body fluids. This work provides the first example of the exploitation of the
nanoparticle−protein corona in analytical chemistry.

E xtracellular Vesicles (EVs) have been identified as


mediators of intercellular communication in many
pathophysiological processes throughout the body; for this
disorders.2,10−12 Therefore, EVs are envisioned as captivating
means for “remote” biopsies13,14 and theranostics.15−18
Unfortunately, understanding the molecular mechanisms of
role they have been the subject of a growing scientific interest EV biogenesis, the precise identification of EV physiological
over the past few years.1 relevance and role, and the implementation of EV based
These cellular secreted vesicles comprise microvesicles and biotechnology are to date hampered by the poor physicochem-
ectosomes, budded from the plasma membrane with a size from ical knowledge of these nanosized objects, due to inherent
40 nm to 1 μm, and exosomes of intracellular origin with a size shortcomings of the current bioanalytical methods used for
from 30 to 120 nm.2,3 EVs contain cell specific cargos such as their separation and characterization.2,3,19,20
lipids, proteins, DNA, mRNA, and miRNA4 and populate EVs have a low refractive index and are heterogeneous both
different biological fluids (plasma, cerebrospinal fluid, urine, in size and composition. In addition, the fact that protein
saliva, etc.), making them ideal candidates for biomarker complexes, especially insoluble immunocomplexes, share some
studies.5−7 critical biophysical properties with EVs − including size, surface
The role that EVs are believed to play in various pathological charge, and interaction with light − compromises their
conditions is linked to the delivery of cargos to surrounding detection and isolation. This, in turn, affects EV quantification
cells actuating a functional manipulation of the microenviron- by conventional bioanalytical methods, such as flow cytometry,
ment, for example tumor derived EVs can impair antitumor and purification by differential centrifugation, especially in
immune response reducing lymphocyte activity.8 In addition diseases where formation of immunocomplexes is common,
several studies have focused on the role of EVs in cancer
biology because their release is significantly altered in cancer Received: September 30, 2014
cells;9 in the same way their activity in intercellular Accepted: February 12, 2015
communication2 seems to be involved in neurodegenerative Published: February 12, 2015

© 2015 American Chemical Society 4168 DOI: 10.1021/ac504861d


Anal. Chem. 2015, 87, 4168−4176
Analytical Chemistry


Article

including autoimmune diseases, as well as hematologic EXPERIMENTAL SECTION


disorders, infections, and cancer.19,21
EV Purification and Characterization. EVs were
Recently, the problem has been tackled from the analytical
separated from human serum by performing differential
side by dedicated resistive pulse sensing22 and multiplex
centrifugation (DC) or DC and sucrose gradient fractionation
systems.13,14 In parallel, unedited biophysical methods,
(SGF) in sequence, adapting recently published protocols20,21
including nanoparticle tracking analysis, Raman microspectro-
(see scheme and methods in the SI). As demonstrated in this
scopy, micronuclear magnetic resonance, and small-angle X-ray
subsection, the preparations obtained by DC contain single and
scattering (SAXS) are under exploration.22 In the last year,
excellent results have been obtained with commercial9,23 and aggregated protein contaminants not associated with the EVs
nanostructured13,14 Surface Plasmon Resonance (SPR) sensors. (hereafter referred as SAP) which can be subsequently removed
The present work is a nanotechnological contribution to by SGF. Therefore, the samples obtained through DC can be
these efforts and reports about a cost-effective and fast considered as the model for SAP contaminated EV preparations
colorimetric assay for probing protein contaminants and (hereafter referred as EVs+SAP), while the samples purified by
determining the concentration of EV preparations. The assay DC and SGF applied in sequence can be considered as the
working principle, sketched in Figure 1, exploits colloidal gold model for pure EV preparations (hereafter referred as EVs).
EVs and EVs+SAP were first verified by routine biochemical
and biophysical analysis21 (materials and methods are given as
SI). Both preparations resulted in containing high concen-
trations of the same EV population, as evidenced by the
characteristic phosphatidyl-choline and sfingomyelin lipid
content, by the acetylcholine-esterase enzymatic activity by
the presence of the typical exosomal and microvesicular
markers (SI, Figure S2). The morphology of the EVs was
checked by Scanning Electron Microscopy (SEM) (SI, Figure
S2) and Atomic Force Microscopy (AFM), which gave
consistent results (SI, Figures S3 and S4). In particular, both
EVs and EVs+SAP resulted in containing vesicles of the
expected36 spherical shape and size distribution, centered at
160 nm. The nature of the considered EV populations was
further confirmed by the ζ-potential value, equal to −17.7 ± 2.3
mV for EVs, while it is lower for EV+SAP (−10.3 + 1.2 mV)
due to a higher presence of proteins in the sample that lower
the “apparent” ζ-potential.37,38 Finally the hydrodynamic sizes
obtained from Photon Correlation Spectroscopy (PCS) (Figure
2A), and the positivity to the TritonX-100 test (SI, Figure S3,
panel C), are all in perfect agreement with the typical
characteristics of purified EVs reported in the literature.19
Figure 1. Nanoplasmonic assay for probing by eye protein The total protein content of EVs and of EVs+SAP derived
contaminants (single and aggregated exogenous proteins, SAP) in from 1 mL of human serum was first determined through a
EV preparations. See the main text for a full explanation.
Bradford quantification39 and resulted in 0.3 μg/μL for EVs and
9 μg/μL for EVs+SAP, respectively (SI, Figure S2). These
nanoplasmonics24−29 and the fact that nanoparticle (NP) values highlight a significant difference in protein content
aggregation at lipid membranes30 is modulated by the presence between the two samples. However, the Bradford assay
of a protein corona (PC31−34) around the NPs. The assay kit routinely used to quantify the total proteins and often used
simply consists of a probe solution of dispersed gold NPs of to quantify EVs is biased by the lipid amount in the sample and
15−20 nm size, which looks red due to the NP characteristic therefore does not allow discriminating between EV associated
LSPR (localized SPR, red absorption spectrum). After the proteins, SAP, and lipids.20 This drawback, recurrent in
addition of a pure EV preparation to the colloidal dispersion, literature,20,40 has been circumvented performing dedicated
the NPs adsorb and cluster at the EV membrane causing a red- experiments of PCS, AFM, and developing an ad hoc native
shift and a broadening of the LSPR (blue absorption spectrum), agarose gel electrophoresis assay.
thus a change of the solution color into blue. Conversely, in the PCS results are reported in Figure 2A, where the scattered
case of an EV preparation containing single and aggregated field autocorrelation curves for EVs (black triangles) and EVs
protein contaminants (hereafter referred as SAP) the NPs +SAP (red circles) are reported. Here EVs+SAP displays a
preferentially interact with those proteins to form a PC (EV faster and steeper decay of the autocorrelation function, G(τ),
+SAP, Extracellular Vesicles + Single and Aggregated with respect to EVs, indicating that the EVs+SAP sample
Proteins).35 The PC both inhibits the interaction between contains a significant fraction of objects with a smaller size with
NPs and EVs and prevents NPs from aggregating at the EV respect to the EV sample (see also SI Figure S4).
membrane, thus keeping unchanged the NP LSPR and in turn To confirm that these smaller particles are SAP, we
the solution color to the initial red. developed and ran an agarose gel electrophoretic assay in
In this contribution we will present and discuss the proof of native conditions for the separation of a mixed colloidal
concept of the assay, as well as its implementation for the solution.41,42 We fluorescently stained EVs derived from 1 mL
titration of pure EV preparations and the mechanism of signal of human serum, EVs+SAP derived from 1 mL of human
generation. serum, 3 ng of synthetic phosphatidylcholine (PCh) liposomes
4169 DOI: 10.1021/ac504861d
Anal. Chem. 2015, 87, 4168−4176
Analytical Chemistry Article

Figure 2. (A) Normalized autocorrelation curves obtained through photon correlation spectroscopy (PCS) for EV and EVs+SAP preparations. The
lower decay time of the EVs+SAP is due to the presence of SAP. (B) Agarose electrophoretic separation of the EVs and of the EVs+SAP
preparations. Left panel and right panel refer to PHK67 fluorescent staining and Comassie staining, respectively. PCh liposomes (PCh ves) and BSA
were loaded as control. (C and D) AFM images of EVs and EVs+SAP adsorbed on mica (topographic mode).

of 150 nm average diameter, and 10 μg of Bovine Serum DC or by DC-SGF share the same integrity and structure and
Albumin (BSA) protein with the marker PKH67, specific for that the preparations just differ for the presence or not of SAP
lipid bilayers (all diluted in 10 mM PBS solution, see the SI for contaminants.
description of the experiment). Synthesis and Characterization of Au NPs. Fifteen nm
Note that PCh liposomes are a convenient synthetic mimic Au NPs were synthesized in Milli-Q water (nanomolar
of EVs, since they have similar lipid composition and size concentration) following the classical Turkevich protocol and
distribution with respect to EVs;23 the PCh liposomes were characterized by UV/vis/NIR spectroscopy, AFM, and PCS.
prepared as described in ref 30. Full details are given in the SI.
The preparations were run on the gel shown in the left panel UV/vis/NIR Spectroscopy. UV−vis spectra were measured
of Figure 2B, where dark bands corresponding to the lipid with a JASCO UV/vis/NIR spectrophotometer. Briefly, NP or
vesicles appear at the same height in the lanes of the 150 nm PC were resuspended in the appropriated solvent (water or
liposomes (PCh ves), EVs, and of the EVs+SAP, while they are PBS) and the absorbance of the solution measured in the
not present in the BSA lane. The smearing of the EVs+SAP presence and absence of the different EVs preparations.
band can be attributed to the SAP contaminants present in the Nanoplasmonic Assay for Detecting SAP in EV
sample. The Coomassie staining, which marks both lipids and Preparations. EVs and EVs+SAP were prepared and
proteins, of the same gel in the right panel of Figure 2B characterized as described in the SI and then resuspended at
confirms this interpretation and evidence that the SAP content the desired concentration using PBS. EVs and EVS+SAP were
is negligible in the EV sample. Therefore, the small amount of then incubated for 20 min, with gold NPs (concentration 6
proteins measured in EVs by the Bradford assay consists of nM), and the spectroscopic features of the solution were then
proteins and lipids associated with EVs. evaluated using a JASCO spectrophotometer. Alternatively gold
These salient characteristics are definitively confirmed by the NPs after their purification from a 10% human serum solution
AFM images of EVs and EVs+SAP deposited onto freshly were incubated with EVs. The same solutions were also
cleaved mica. In fact, while the EVs sample shows isolated and measured using Photon Correlation Spectroscopy using a
well-defined EVs of 100−200 nm size lying onto the Brookhaven Instrument 90 Plus (Brookhaven, Holtsville, NY).
subnanometer smooth mica surface (Figure 2C), the EVs One mL of human serum was processed by DC (see Results
+SAP sample (Figure 2D) consists of a few EVs of the same and Discussion). The supernatant of the solution containing
size buried into a matrix of smaller SAP nanoparticles. All these the soluble SAP was then collected discarding the pelleted
observation confirm, as already shown,20 that EVs retrieved by fraction containing the EVs. SAP were quantified by Bradford
4170 DOI: 10.1021/ac504861d
Anal. Chem. 2015, 87, 4168−4176
Analytical Chemistry Article

Figure 3. (A) 15 μL of the nanoplasmonic assay, that is an aqueous dispersion of 15 nm gold NPs at 6 nM concentration, upon the addition of 10 μL
of a 10 mM PBS solution containing EVs+SAP (left tube) or EVs (right tube) derived from 1 mL of human serum. (B) UV/vis/NIR absorption
spectra of the assay before and after the addition of EVs and pure 10 mM PBS. (C) UV/vis/NIR absorption spectra of the assay before and after the
addition of EVs+SAP. Spectra were acquired with 1 nm step size and a data point every 25 nm highlighted with a symbol to ease comparison. (D)
and (E) SAP progressive inhibition of NP clustering visually probed with a multiwelled plate and quantified by the NP aggregation index (AI),
respectively. In panel (E) the errors bars indicate the standard error of three different replicates.

assay and were spiked in SGF purified EVs at concentrations


ranging from 5 ng/mL to 1 μg/mL. The PBS solutions
■ RESULTS AND DISCUSSION
Probing the Presence of SAP in EV Preparation by the
containing increasing concentrations of SAP and EVS were Nanoplasmonic Colorimetric Assay. The nanoplasmonic
then incubated with gold NPs (concentration 6 nM). After 20 assay consists of 5 μL of a 6 nM water dispersion of 15 nm gold
min of incubation 100 μL of each sample was loaded on a 96 NPs, stabilized by citrate anions (the physicochemical
wells plate, and the absorbances of the solutions were measured characterization has been reported in the SI Figures S5 and
by a plate reader at the wavelengths 540 and 650 nm. S6). Figure 3A shows the assay at work. When 10 μL of a 10
Plasmonic Titration. Increasing concentrations of the mM PBS solution containing EVs derived from 1 mL of human
synthetic PCh vesicle were resuspended in Tris NaCl EDTA serum is added, the solution color turns into blue (right tube).
(the same concentration used for the EV sample), and they Conversely, upon addition of the same amount of EVs+SAP the
were then diluted in PBS (see the main text for the solution keeps the initial red color (left tube) (a thorough
concentration tested) and incubated with gold NP (concen- report about EV and EV+SAP preparations and character-
tration 6 nM). After 20 min of incubation samples were ization can be found in the Experimental Section and in the
evaluated by UV/vis/NIR spectroscopy using a JASCO UV/ Supporting Information). Therefore, the assay allows for
vis/NIR spectrophotometer. probing EV preparation purity by eye.
Other Experimental Information. A fully detailed The assay optical proprieties were quantitatively investigated
Materials and Methods section is given in the SI, which by UV/vis/NIR spectroscopy.43 To ensure optimal conditions
includes the description of chemicals and the standard methods for collecting gold NPs LSPR absorption spectra, all the
and techniques used (e.g., immunoassays, synthesis of PCh analyzed assay solutions were diluted to the same final NP
liposomes, AFM and SEM microscopy, PCS experiments, and molar concentration of 2 nM. From the spectra, reported in
data analysis). Figure 3B, we learn that the addition of the EVs drives a red
4171 DOI: 10.1021/ac504861d
Anal. Chem. 2015, 87, 4168−4176
Analytical Chemistry Article

Figure 4. (A) UV/vis/NIR absorption spectra of PBS solutions containing a fixed concentration of NPs and increasing concentrations of EVs,
obtained after the dilution of the EV stock preparation (EVin is the dilution ratio of the stock solution). Spectra were acquired with 1 nm step size
and a data point every 25 nm highlighted with a symbol to ease comparison. (B) AI obtained from part A plotted versus EVin and linear regression fit
(red line). (C) UV/vis/NIR absorption spectra of PBS solutions containing a fixed amount of NPs and increasing concentrations of PCh liposomes.
Spectra were acquired and represented as in panel A. (D) Calibration line obtained by plotting the NP AI obtained from part C versus the PCh
liposome solution concentration (black circles) and linear regression best fit (red line, R2 = 0.99). Errors bars indicate standard error of three
different replicates. The star point highlights the intercept of the AI value of the EV preparation (obtained from the spectra of Figure 3B) with the
regression line. The star point projection on the abscissa axis allows for the extrapolation of the unknown concentration (x = 4.5 ·109 vesicles/mL).

shift (blue triangles) of the initial LSPR peak (red squares). (10 mM PBS solution), covering the typical SAP concentration
This well agrees with the observed color change of the solution range commonly found in EVs prepared from biological fluids
from red to blue and mirrors the agglomeration and clustering (see the “EV Purification and Characterization” section and refs
of NPs at the EV surface.30 The hypothesis that the red shift is 19 and 21).
not merely due to NP aggregation caused by the addition of 10 The samples were then loaded in a multiwell plate, and the
mM PBS solution (through charge shielding) is ruled out by a probe NP solution was added (again to a final NP
control sample obtained by adding pure 10 mM PBS solution concentration of 2 nM). The most relevant results are
to the NP solution (and by other results presented in the next displayed in Figure 3D. The addition of the NP probe to the
sections of the paper). The spectrum of this control sample EVs with an increasing SAP amount is mirrored by a gradual
(black circles) has a markedly different profile with respect to change of the solution color from blue to red, indicative of
the one of EVs+NP and is characterized by the unspecific gradual NP aggregation inhibition. NP aggregation was
broadening of the LSPR peak (see the SI for replicates of the quantified through the aggregation index (AI), which can be
experiment). Figure 3C shows the typical LSPR spectrum defined for spherical gold NPs as the ratio of the LSPR
obtained upon addition of EVs+SAP to the NP solution. In absorption peak of pure NPs and the LSPR at some significant
contrast to what happens with the addition of EVs, here the red-shift wavelength.19,45 In view of Figure 3B and 3C spectra,
LSPR peak undergoes a slight broadening and red shift with here we chose LSPR absorption at 540 and 650 nm (AI =
respect to bare NPs (black circles and red squares spectra, A540/A650). The obtained AI values are plotted against the
respectively). These features account for the unchanged color SAP concentrations in Figure 3E to yield a calibration curve for
of the solution and are typical of gold NPs cloaked by a protein the SAP detection range of the nanoplasmonic assay, which hits
corona.44 This suggests the NPs are passivated (with respect to a lower limit of 5 ng/μL of SAP, far below the typical SAP
aggregation or clustering) by a preferential interaction with the content in EVs preparations.19,21 Additional tests of the assay
SAP contained in the sample, eventually bringing to the effectiveness against pure EVs, high SAP concentration (4 μg/
formation of isolated NP-protein corona (NP-PC) complexes μL), and human serum are reported in the SI, Figure S7.
(see later in the text for a thorough discussion). Determining EV Concentration in Pure EV Prepara-
We then investigated the dynamic range of the nano- tions. The nanoplasmonic assay can be implemented to titrate
plasmonic assay. Ten mM PBS solutions containing a fixed pure EV preparations (viz. preparations containing negligible
concentration of pure EVs were spiked with increasing amounts amounts of SAP). Titration grounds on the finding that in pure
of SAP varying from 5 ng/μL to 1 μg/μL molar concentrations EV solutions the LSPR red shift, viz. the AI, of the NPs has a
4172 DOI: 10.1021/ac504861d
Anal. Chem. 2015, 87, 4168−4176
Analytical Chemistry Article

Figure 5. (A) Normalized UV/vis/NIR spectra obtained for an aqueous solution containing 15 nm gold NP and a 10 mM PBS solution containing
the same NP coated by a human serum PC in the presence or absence of EVs. Spectra were acquired with 1 nm step size and a data point every 35
nm highlighted with a symbol to ease comparison. (B) AFM phase imaging of 15 nm gold NP and the same NP coated by a human serum protein
corona (C) after their interaction with EVs and their deposition on a mica substrate. Inset reports a magnification of the EV boxed. NP clustering
over an extended EV zone can be detected (dark area on EVs in B), while PC adsorption at the EV surface results in a limited event. (D)
Autocorrelation curves obtained for an aqueous solution of 15 nm gold NPs (NP) at nanomolar concentration, the same NP coated by a human
serum protein corona (PC) and dispersed in 10 mM PBS, 10 mM PBS solution of EVs (EVs), and a 10 mM PBS solution containing EVs+PC or
EVs+NP. The amplitude of the autocorrelation curves is normalized to 1 for better comparison of the characteristic diffusion time, τ.

linear dependence on the NP/EV molar ratio, as shown in containing the lower EV or PCh liposome concentration
Figure 4A and 4B. Since the molar concentration of any EV (yellow triangles up and red circles, presented respectively in
preparation is unknown, we used the 150 nm synthetic Figure 4 A and C) are characterized by the presence of a
phosphatidylcholine (PCh) liposomes introduced in the shoulder in the near IR region. This spectroscopic feature is
Experimental Section (Figure 2B) − which are synthetic very likely ascribable to the presence of nanoparticles
vesicles that mimic EVs in both size and lipid composition − to structuring in dimers, that can couple the LSPR transverse
build a calibration line. PCh liposome molar concentration can mode proper of isolated nanoparticles to a longitudinal mode
be easily calculated from the stoichiometry of the prepared PCh due to the presence of interconnected particles.48,49
solution.30 By applying the calculations presented in our recent study on
In particular, 10 mM PBS solutions of PCh liposomes with the interaction of gold NPs with giant unilamellar vesicles30
concentrations ranging from 2.1 × 107 to 2.1 × 1010 vesicles/ (see the SI), we were able to evaluate the dependence of the
mL, viz. from 35 fM to 35 pM, were prepared. This range was clustering to the NP cross section/EV surface area, finding that
chosen as it includes the pathophysiological EV concentrations at the highest liposome concentration the overall surface of the
reported in the literature.46 Each PCh liposome solution was NPs matches the overall surface of the external bilayer leaflet of
then incubated with the 6 nM solution of 15 nm gold NPs in a the liposomes and that as the liposome amount decreases the
1:1 volume ratio. The collected LSPR spectra are shown in overall surface of the NPs exceeds the liposome one up to a
Figure 4C. Here we see that at the highest liposome factor of 10. This suggests that at the highest liposome
concentration the spectrum (blue squares) is slightly shifted concentration the amount of NPs is too low to saturate the
with respect to one of the isolated gold NPs reported in Figure overall liposome surface and clustering is negligible; clustering
3B and 3C, with the LSPR peak centered at about 540 nm. is then progressively triggered as the liposome/NP ratio
Then, as the liposome concentration decreases, the LSPR peak decreases and gets to plateau for low vesicle concentration.30
undergoes a gradual red shift and a decrease of intensity (red Indeed, as suggested by comparison of Figure 4A and 4C,
circles). This suggests that NP clustering is linearly dependent analogous scenario and reasoning hold if one has EVs in place
to the liposome concentration and becomes more and more of PCh liposomes.
prominent along with the decrease of the vesicle concen- In Figure 4D the AI from Figure 4C is plotted as a function
tration.47 Interestingly the LSPR spectra of the samples of the liposome concentration, and, as evidenced by the linear
4173 DOI: 10.1021/ac504861d
Anal. Chem. 2015, 87, 4168−4176
Analytical Chemistry Article

regression line, in the investigated conditions the parameters spectroscopy.50 Applying this method we were able to measure
are in linear dependence. This calibration line finally allows for the diffusion coefficients of the NP and of the EV+NP sample,
extrapolating the unknown concentration of any pure EV that are related to the hydrodynamic sizes of the diffusing
solution, provided it falls in the calibration range. objects through the Stokes−Einstein relationship. The average
For example, we determined the concentration of the EV of 30 autocorrelation curves for the NPs before and after the
preparation purified from human plasma used throughout this formation of the PC have been evaluated. Figure 5D shows that
work. The AI value was obtained from the UV/vis/NIR the characteristic diffusion time of NPs increased in the
spectrum reported in Figure 3B. In particular the star point in presence of PC. In particular, PC formation drives a shift of the
Figure 4D highlights the intercept of the AI value with the autocorrelation curve to higher decay times τ, while when PC
regression line, namely x = 4.5 ·109 vesicles/mL. Since the are dispersed in the presence of EVs there is a small increase of
original EV sample was diluted to 1:12 vol/vol in a 10 mM PBS the decay time that can be attributed to a partial interaction of
solution for UV/vis/NIR spectrometry, from the value of x and PC with EVs. On the contrary the clustering of NP at the EV
of the dilution proportion we obtain the original concentration surface drove a higher shift of the autocorrelation curve that is
of the sample is equal to 5.4 × 1010 vesicles/mL. similar to the one obtained for EVs. This hints to the presence
Remarkably, this value is consistent with the typical EV in solution of object containing NPs with size similar to the size
concentrations in human plasma.20 of the EVs. The ξ-pot for these hybrid NP-EV objects is
Titration was successfully replicated on another EV different (−11.1 ± 1.3 mV) from the one of the NPs alone in
preparation separated from a different donor plasma and by water (−31.6 ± 3.3 mV) or the NPs dispersed in PBS (−24.8 ±
using a different batch of NPs and PCh liposomes, proofing the 0.5 mV) (see SI Figure S9) suggesting the presence of NP-
robustness and repeatability of the titration method (SI, Figure membrane complexes than simple NP aggregates. Moreover
S8). the fact that NP clustering at EV membrane, that is mirrored by
Further Assessment of the Interaction between NPs, a variation of the AI, has a linear dependence on the EV
SAP, and EVs. The (bio)chemical/(bio)physical mechanism concentration in the studied range (see Figure 4 A-D)
we propose for explaining the signal generation of the represents another genuine control of the ability of the assay
nanoplasmonic assay (sketched in Figure 1) relies on the to sense EVs in pure EV preparations.
extension to EVs of our recently reported findings about the Taken together, all the above data point to the mechanism of
interaction of gold NPs with synthetic Giant Unilamellar signal generation of the assay that is summarized and described
Vesicles.30 In essence, our experimental evidence are in in Figure 1. In the presence of pure EV preps, NPs can freely
agreement with the claim that bare (citrate capped) gold NPs interact with EV membranes clustering at their surface, while in
cluster at the EV surface, while the formation of a protein the presence of SAP the NP preferentially interact with them
corona (PC)31 around the NPs strongly attenuates this forming a SAP corona which inhibits membrane clustering.
phenomenon − this for NP concentrations in excess with Therefore, NP-biomolecule interactions can be exploited as
respect to the full coverage (upon adsorption) of the EV nanoplasmonic transducer of the presence of SAP or as
external bilayer leaflet, see ref 30 for a thorough discussion. nanoplasmonic gauge for the evaluation of the EV concen-
To substantiate this hypothesis, we compared the interaction tration.
of the assay NPs and the same NPs passivated with a protein
corona from human serum (hereafter referred as PCs) with
EVs. Figure 5 shows the UV/vis/NIR spectra of a 2 nM water
■ CONCLUSIONS
We presented a nanoplasmonic colorimetric assay for probing
solution of bare gold NP (red squares), of a 2 nM PBS solution by eye the presence of SAP contaminants in EV preparations
(PBS 10 mM) of PC (blue triangles), and of the PC solution and to determine the concentration of EVs in pure
after incubation with a 10 mM PBS solution of EVs of preparations. The assay applies to the whole multiscale
concentration of 4.2 × 109 vesicles/mL EVs (black circles). population of EVs in body fluids, which comprises vesicles
All three spectra share the same main features, showing a from 40 nm to 1 μm in size.
LSPR peak centered at 540−550 nm, indicating that all the The working principle of the assay is based on the fact that
samples contain isolated NPs. Remarkably, the spectra of the gold NP clustering at the EV membrane and the related LSPR
PC+EV sample (gray triangles) definitively matches the one shift is directly gauged by the amount of SAP contained in the
obtained for the NP+EV+SAP sample reported in Figure 3C. preparation. Bare gold NPs adsorb and aggregate at the EV
This indicates SAP inhibits the interaction between NPs and membrane in pure EV preparations, while this phenomenon is
EVs as a protein corona formed ex situ does. inhibited in SAP contaminated preparations by the SAP-NP
Interaction between EVs and PC was further confirmed interaction that leads to the formation of a passivating protein
through an AFM morphological analysis of EVs+NP and EVs corona that keeps the NPs dispersed, inhibiting NP-EV
+PC deposited on mica. As the AFM phase image of EVs+NP interactions. The two conditions are optically characterized
shows (Figure 5B), the presence of areas of the lipid bilayers by a specific LSPR shift, which therefore acts as a color switch,
with different composition and stiffness, visible as dark clusters giving a naked eye colorimetric read out of the presence of
that decorate the EV membrane (see inset), is consistent with protein contaminants, that is visible down to 5 ng/μL of SAP,
the presence of NPs onto the membrane. far below the typical concentrations of SAP in EVs
Conversely, for the PC+EV sample (AFM phase image, preparations.
Figure 5C) the presence of the PC limits the formation of NP Remarkably, the LSPR red-shift driven by NP clustering at
clusters, as most of the PC can be found on the background of the EV membrane is directly proportional to the EV
the mica surface, and very few NPs can be visualized adsorbed concentration. This discloses the opportunity to exploit the
at the EV surface (inset). assay for titrating EV pure preparations, which is another
To further prove this hypothesis in situ, we performed some fundamental issue in EV sample analysis.20 The titration
equilibrium binding experiments with photon correlation method resulted robust and repeatable, with a dynamic range
4174 DOI: 10.1021/ac504861d
Anal. Chem. 2015, 87, 4168−4176
Analytical Chemistry


Article

from 35 fM to 35 pM, matching the range of EV concentration ABBREVIATIONS


in body fluids. NP nanoparticle
These findings are an effective application of nanotechnology PC protein corona
in biology with immediate impact in EV biology − which strives EVs extracellular vesicles
for effective analytical procedures that fit the requirements of SAP single and aggregated protein contaminants
low volumes, reduced operative costs, fast readout, and DC differential centrifugation
compatibility with common laboratory practices51,52 − and SGF sucrose gradient fractionation
complement the available information on the interaction of SPM scanning probe microscopy


NPs with biological membranes.53 On a wider perspective, the
fact that the assay applies invariably to micro- and nanovesicles REFERENCES
prompts its extensions to other biomembrane objects, such as (1) Théry, C.; Zitvogel, L.; Amigorena, S. Nat. Rev. Immunol. 2002, 2,
membrane coated viruses or organelles isolated from cell 569−579.
materials. (2) Raposo, G.; Stoorvogel, W. J. Cell Biol. 2013, 200, 373−383.
Finally, it is worth noticing that this work widens to clinical (3) Bence, G.; Szabo, G.; Pasztoi, M.; Pal, Z.; Misja, P.; Aradi, B.;
and biological analytical chemistry applications aimed at Laszlo, V.; Pallinger, E.; Pap, E.; Kittel, A.; Ngy, G; Falus, A.; Buzas, E.
exploiting, rather than avoiding, the NP-protein corona, that Cell. Mol. Life Sci. 2011, 68, 2667−2688.
to date, with few exceptions,59 have been focused on drug (4) Yang, J.-M.; Gould, S. J. Biochem. Soc. Trans. 2013, 41, 277−282.
loading and delivery.54−58 (5) Inal, J. M.; Fairbrother, U.; Heugh, S. Biochem. Soc. Trans. 2013,


41, 237−240.
(6) Inal, J. M.; Kosgodage, U.; Azam, S.; Stratton, D.; Antwi-baffour,
ASSOCIATED CONTENT
S.; Lange, S. Biochim. Biophys. Acta, Proteins Proteomics 2013, 1834,
*
S Supporting Information 2317−2325.
Supplementary data, figures, and a fully detailed Materials and (7) Di Noto, G.; Paolini, L.; Zendrini, A.; Radeghieri, A.; Caimi, L.;
Methods section. This material is available free of charge via the Ricotta, D. PLoS One 2013, 8, e70811.
Internet at http://pubs.acs.org. (8) Clayton, A.; Mitchell, J. P.; Court, J.; Mason, M. D.; Tabi, Z.


Cancer Res. 2007, 67, 7458−7466.
(9) Zhu, L.; Wang, K.; Cui, J.; Liu, H.; Bu, X.; Ma, H.; Wang, W.;
AUTHOR INFORMATION Gong, H.; Lausted, C.; Hood, L.; Yang, G.; Hu, Z. Anal. Chem. 2014,
Corresponding Author 86, 8857−8864.
*E-mail: daniele.maiolo@polimi.it. (10) Beninson, L. A.; Fleshner, M. Semin. Immunol. 2014, 1−8.
(11) Katoh, M. Int. J. Mol. Med. 2013, 32, 763−767.
Present Address (12) Van Balkom, B. W. M.; van Doorn, J.; Verhoeven-Duif, N. M.;

European Center for Nanomedicine CEN foundation, c/o Verhaar, M. C. J. Inherited Metab. Dis. 2014, 37, 497−504.
Nanostructured Fluorinated Material laboratory, Department of (13) Yoshioka, Y.; Kosaka, N.; Konishi, Y.; Ohta, H.; Okamoto, H.;
Chemistry, Material and Chemical Engineering, Milan Italian Sonoda, H.; Nonaka, R.; Yamamoto, H.; Ishii, H.; Mori, M.; Furuta,
Polytechnic, Via Mancinelli 7, 20131 Milan, Italy. K.; Nakajima, T.; Hayashi, H.; Sugisaki, H.; Higashimoto, H.; Kato, T.;
Author Contributions Takeshita, F.; Ochiya, T. Nat. Commun. 2014, 5, 3591.
(14) Im, H.; Shao, H.; Park, Y., Il; Peterson, V. M.; Castro, C. M.;
D.R. selected and collected serum for EV purification. D.M., Weissleder, R.; Lee, H. Nat. Biotechnol. 2014, 1−9.
L.P., A.Z., and G.D.N. performed the experiments, with the (15) Alhasan, A. H.; Patel, P. C.; Choi, C. H. J.; Mirkin, C. A. Small
exception of PCS experiments, that were performed by D.B. 2014, 10, 186−192.
D.M., D.R., and P.B. conceived and supervised the work. All (16) El-Andaloussi, S.; Lee, Y.; Lakhal-Littleton, S.; Li, J.; Seow, Y.;
authors contributed to the conception of the experiments and Gardiner, C.; Alvarez-Erviti, L.; Sargent, I. L.; Wood, M. J. A. Nat.
discussion of the results and contributed to writing the Protoc. 2012, 7, 2112−2126.
manuscript. All authors have given approval to the final version (17) Alvarez-Erviti, L.; Seow, Y.; Yin, H.; Betts, C.; Lakhal, S.; Wood,
of the manuscript. M. J. A. Nat. Biotechnol. 2011, 29, 341−345.
(18) Sahay, G.; Querbes, W.; Alabi, C.; Eltoukhy, A.; Sarkar, S.;
Notes Zurenko, C.; Karagiannis, E.; Love, K.; Chen, D.; Zoncu, R.; Buganim,
The authors declare no competing financial interest. Y.; Schroeder, A.; Langer, R.; Anderson, D. G. Nat. Biotechnol. 2013,

■ ACKNOWLEDGMENTS
The authors would like to thank Sara Busatto for technical
31, 653−658.
(19) György, B.; Módos, K.; Pállinger, É.; Pálóczi, K.; Pásztói, M.;
Misják, P.; Mária, A.; Sipos, Á .; Szalai, A.; Voszka, I.; Polgár, A.; Tóth,
K.; Csete, M.; Nagy, G.; Gay, S.; Falus, A.; Kittel, Á .; Buzás, E. I.; Dc,
assistance, Kimberly Hamad Schifferli, Francesca Baldelli W. Blood 2011, 117, e39−e48.
Bombelli, Ciro Chiappini, and Eugenio Monti for fruitful (20) Webber, J.; Clayton, A. J. Extracell. Vesicles 2013, 2, 1−6.
discussions and suggestions. We thank Marco Vitale and (21) Kalra, H.; Adda, C. G.; Liem, M.; Ang, C.; Mechler, A.;
Davide Dallatana for SEM consulting and Roberta Giuliani for Simpson, R. J.; Hulett, M. D.; Mathivanan, S. Proteomics 2013, 3354−
use of a UV−vis spectrometer. Dr. Di Noto received a 3364.
fellowship “Bando Mecenati” from “Fondazione CEUR”. D.M. (22) Van Der Pol, E.; Hoekstra, A.; Sturk, A.; Otto, C.; Van Leeuwen,
is also grateful to the Academia Delle Nanoscienze di Gagliato G.; Nieuwland, R. J. Thromb. Haemostasis 2010, 8, 2596−2607.
(Nanogagliato) for the Salvatore Venuta fellowship. This work (23) Rupert, D. L. M.; Lässer, C.; Eldh, M.; Block, S.; Zhdanov, V. P.;
Lotvall, J. O.; Bally, M.; Höök, F. Anal. Chem. 2014, 86, 5929−5936.
was supported by MIUR (Ministero Italiano dell’Università e (24) D Howes, P.; Rana, S.; M Stevens, M. Chem. Soc. Rev. 2013, 43,
della Ricerca) through the project Soft Matter Nanostrutturata: 3835−3853.
dall’indagine chimico-fisica allo sviluppodi applicazioni innova- (25) Liz-Marzán, L. M.; Murphy, C. J.; Wang, J. Chem. Soc. Rev. 2014,
tive, PRIN 2010−2011 grant 2010BJ23MN to D.M., D.B., and 43, 3820−3822.
P.B. and by University of Brescia Research found (ex 60%) and (26) Sonntag, M. D.; Klingsporn, J. M.; Zrimsek, A. B.; Sharma, B.;
“‘Prima spes onlus”’ foundation to D.R., L.P., A.Z., and G.D.N. Ruvuna, L. K.; Van Duyne, R. P. Chem. Soc. Rev. 2014, 43, 1230−1247.

4175 DOI: 10.1021/ac504861d


Anal. Chem. 2015, 87, 4168−4176
Analytical Chemistry Article

(27) Saha, K.; Agasti, S. S.; Kim, C.; Li, X.; Rotello, V. M. Chem. Rev. (57) Kah, J. C. Y.; Chen, J.; Zubieta, A.; Hamad-Schifferli, K. ACS
2012, 112, 2739−2779. Nano 2012, 6, 6730−6740.
(28) Rodríguez-Lorenzo, L.; de la Rica, R.; Á lvarez-Puebla, R. A.; Liz- (58) Cifuentes-Rius, A.; de Puig, H.; Kah, J. C. Y.; Borros, S.; Hamad-
Marzán, L. M.; Stevens, M. M. Nat. Mater. 2012, 11, 604−607. Schifferli, K. ACS Nano 2013, 7, 10066−10074.
(29) Liu, G. L.; Yin, Y.; Kunchakarra, S.; Mukherjee, B.; Gerion, D.; (59) Hamad-Schifferli, K. Nanomedicine 2013, 8, 1−3.
Jett, S. D.; Bear, D. G.; Gray, J. W.; Alivisatos, A. P.; Lee, L. P.; Chen,
F. F. Nat. Nanotechnol. 2006, 1, 47−52.
(30) Montis, C.; Maiolo, D.; Alessandri, I.; Bergese, P.; Berti, D.
Nanoscale 2014, 6, 6452−6457.
(31) Monopoli, M. P.; Åberg, C.; Salvati, A.; Dawson, K. A. Nat.
Nanotechnol. 2012, 7, 779−786.
(32) Walkey, C. D.; Chan, W. C. W. Chem. Soc. Rev. 2012, 41, 2780−
2799.
(33) Nel, A. E.; Mädler, L.; Velegol, D.; Xia, T.; Hoek, E. M. V;
Somasundaran, P.; Klaessig, F.; Castranova, V.; Thompson, M. Nat.
Mater. 2009, 8, 543−557.
(34) Walczyk, D.; Bombelli, F. B.; Monopoli, M. P.; Lynch, I.;
Dawson, K. A. J. Am. Chem. Soc. 2010, 132, 5761−5768.
(35) Del Pino, P.; Pelaz, B.; Zhang, Q.; Maffre, P.; Nienhaus, G. U.;
Parak, W. J. Mater. Horizons 2014, 1, 301.
(36) Sharma, S.; Rasool, H. I.; Palanisamy, V.; Mathisen, C.; Schmidt,
M.; Wong, D. T.; Gimzewski, J. K. ACS Nano 2010, 4, 1921−1926.
(37) Weiner, B. B. Measuring the Size & Surface Charge of
Exosomes, Microvesicles & Liposomes, 1. http://www.
brookhaveninstruments.com/literature/pd (accessed Feb 19, 2015).
(38) Maiolo, D.; Bergese, P.; Mahon, E.; Dawson, K. A.; Monopoli,
M. P. Anal. Chem. 2014, 86, 12055−12063.
(39) Morton, R. E.; Evans, T. A. Anal. Biochem. 1992, 204, 332−334.
(40) Franquesa, M.; Hoogduijn, M. J.; Ripoll, E.; Luk, F.; Salih, M.;
Betjes, M. G. H.; Torras, J.; Baan, C. C.; Grinyó, J. M.; Merino, A. M.
Front. Immunol. 2014, 5, 525.
(41) Åkesson, A.; Cárdenas, M.; Elia, G.; Monopoli, M. P.; Dawson,
K. A. RSC Adv. 2012, 2, 11245.
(42) Wijaya, A.; Hamad-Schifferli, K. Langmuir 2008, 24, 9966−
9969.
(43) Chanana, M.; Rivera Gil, P.; Correa-Duarte, M. A.; Liz-Marzán,
L. M.; Parak, W. J. Angew. Chem., Int. Ed. Engl. 2013, 52, 4179−4183.
(44) Zhang, H.; Zheng, H.; Long, Y.; Xiao, G.; Zhang, L.; Wang, Q.;
Gao, M.; Bai, W. Talanta 2012, 89, 401−406.
(45) Kah, J. In Nanomaterial Interfaces in Biology SE - 9; Bergese, P.,
Hamad-Schifferli, K., Eds.; Methods in Molecular Biology; Humana
Press: 2013; Vol. 1025, pp 119−126.
(46) Carr, B.; Wright, M.Nanoparticle Tracking Analysis. In
Nanosight Ltd, see: http://www.nanosight.com/uploads/Exosome_
and_Microvesicle_Research_2010-2012.pdf, 2012.
(47) Volodkin, D. V.; Skirtach, A. G.; Möhwald, H. Angew. Chem., Int.
Ed. Engl. 2009, 48, 1807−1809.
(48) Skirtach, A. G.; Karageorgiev, P.; Bédard, M. F.; Sukhorukov, G.
B.; Möhwald, H. J. Am. Chem. Soc. 2008, 130, 11572−11573.
(49) Takei, H.; Bessho, N.; Ishii, A.; Okamoto, T.; Beyer, A.; Vieker,
H.; Gölzhäuser, A. Langmuir 2014, 30, 2297−2305.
(50) Dominguez-Medina, S.; Mcdonough, S.; Swanglap, P.; Landes,
C. F.; Link, S. Langmuir 2012, 28, 9131−9139.
(51) Marchesano, V.; Hernandez, Y.; Salvenmoser, W.; Ambrosone,
A.; Tino, A.; Hobmayer, B.; de la Fuente, J. M.; Tortiglione, C. ACS
Nano 2013, 7, 2431−2442.
(52) Giljohann, D. A.; Mirkin, C. A. Nature 2009, 462, 461−464.
(53) Montis, C.; Castroflorio, B.; Mendozza, M.; Salvatore, A.; Berti,
D.; Baglioni, P. J. Colloid Interface Sci. 2014, DOI: 10.1016/
j.jcis.2014.11.056.
(54) Prapainop, K.; Witter, D. P.; Wentworth, P. J. Am. Chem. Soc.
2012, 134, 4100−4103.
(55) Liu, H.; Moynihan, K. D.; Zheng, Y.; Szeto, G. L.; Li, A. V.;
Huang, B.; Egeren, D. S. Van; Park, C.; Irvine, D. J. Nature 2014, 507,
519−522.
(56) Bertoli, F.; Davies, G.; Monopoli, M. P.; Moloney, M.; Gun, Y.
K.; Salvati, A.; Dawson, K. A. Small 2014, DOI: 10.1002/
smll.201303841.

4176 DOI: 10.1021/ac504861d


Anal. Chem. 2015, 87, 4168−4176

You might also like