You are on page 1of 9

Article

Cite This: ACS Sens. 2019, 4, 1433−1441 pubs.acs.org/acssensors

Ultrasensitive and Reversible Nanoplatform of Urinary Exosomes for


Prostate Cancer Diagnosis
Ping Li,† Xiyuan Yu,† Wujuan Han,† Ying Kong,‡ Weiyang Bao,† Jiaqi Zhang,† Wancun Zhang,†
and Yueqing Gu*,†,‡

Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, No. 639 Longmian Avenue,
Jiangning District, Nanjing 211198, China

State Key Laboratory of Natural Medicine, China Pharmaceutical University, No. 639 Longmian Avenue, Jiangning District,
Nanjing 211198, China
See https://pubs.acs.org/sharingguidelines for options on how to legitimately share published articles.

*
S Supporting Information
Downloaded via CARLETON UNIV on July 17, 2020 at 12:10:09 (UTC).

ABSTRACT: Prostate cancer cell-derived exosomes in urine have been extensively studied recently and regarded as novel
biomarkers for cancer diagnosis and prognosis, which presents wide prospects in clinical applications. Sensitive detection and
specific capture methods are essential for exosomes analysis. Herein, a dual functional platform composed of superparamagnetic
conjunctions and molecular beacons (SMC-MB) is reported. The SMC-MB platform is designed based on aptamer
immunoaffinity with ultrasensitive detection efficiency and reversible isolation capacity, which, respectively, profit from
nonenzymatic amplification methods and magnetic separation along with restriction cleavage. It is noteworthy that exosomes
quantification was exactly amplified and transformed into single strand DNA detection. Correlated measurements evidence that
the limit of detection of SMC-MB is as low as ∼100 particles/μL in urine, and a linear relationship meets between the
logarithmic concentration of exosomes and fluorescence intensity of the molecular beacon. Furthermore, employing prostate
specific membrane antigen (PSMA) aptamer, the platform adapted to detect and capture PMSA-positive exosomes from urine
samples provides excellent diagnostic efficiency for prostate cancer (PCa). The expression of typical biomarkers of PCa, i.e.,
PSA and PCA3 mRNA, is significantly higher in PSMA-positive exosomes. Altogether, the platform and strategy described in
this paper are promising in urinary exosomes analysis and prostate cancer detection.
KEYWORDS: urinary exosomes, cancer biomarker, prostate cancer diagnosis, prostate specific membrane antigen, aptamer

E xosomes are nanoscale extracellular vesicles generated by


inward budding of intercellular endosomes which have
diameters of 30−150 nm, and have gained much attention due
delivered and exchanged through the double lipid membrane
vesicles, thus contributing to the recruitment and reprogram-
ming of the tumor microenvironment. 7,8 Accordingly,
to their abundance of bioinformation alongside the important exosomes are of great significance in cancer diagnosis, as the
roles in intracellular communication.1−3 Cancer-derived amount of secreted exosomes together with cancerous nucleic
exosomes have been identified as important in cancer acids and proteins they contain allow the prediction of cancer
development, progression, as well as drug resistance. Cancer
related pathogenic components, including mRNA (mRNA), Received: April 1, 2019
microRNA (miRNA), noncoding RNA (ncRNA), DNA, and Accepted: April 24, 2019
membrane and cytosolic proteins and lipids4−6 can be Published: April 24, 2019

© 2019 American Chemical Society 1433 DOI: 10.1021/acssensors.9b00621


ACS Sens. 2019, 4, 1433−1441
ACS Sensors Article

Figure 1. Schematic illustration of SMC-MB platform. (a) Construction of SMC-MB platform. (b) Procedure of SMC-MB platform in exosomes
analysis. (c) Principle of nonenzymatic amplification cycle. (d) Exosomes purification by restriction enzyme.

risk.9,10 Circulating exosomes in urine, blood, and saliva can be along with electrochemical24,25 and microfluidic methods,26
noninvasive or minimally invasive biomarkers.11,12 Compared surface enhanced Raman scattering (SERS)27 probes, and
with serum or plasma, urine yields large volumes and permits fluorescent probes28 to detect exosomes simultaneously.
routine collection.13 Urine exosomes have been proposed as However, the limitation of low concentration as well as the
treasure chests of biomarkers especially for some tumors complex background of urine are cannot be ignored in
located anatomically proximate to the urethra.14 Notably, exosomes collection and detection. As a result, signal
recent studies have proved that prostate cancer (PCa) derived amplification and reversible strategy should be further
exosomes are enriched in urine after a slight prostate massage, developed.
which assists the discovery and detection of prostate cancer.15 Herein, a dual functional platform composed of a super-
Even though prostate specific antigen (PSA) has shown paramagnetic conjunction and molecular beacon (SMC-MB)
reasonable sensitivity for prostate cancer screening, diagnosis, was reported to separate and quantify PCa-related exosomes,
as well as prognosis, one of the drawbacks is low specificity, and convert exosome detection to single strand DNA (ssDNA)
such that benign hyperplasic conditions can also lead to PSA detection. Typically, PSMA (prostate specific membrane
increment.16 Thus, tumor-derived exosomes detection is a antigen) aptamer, a recognized PSMA target ligand with
novel approach to find predictive markers for PCa according to high affinity and low immunogenicity was applied to capture
numerous studies. Relatively, prostate specific membrane PSMA-positive exosomes.29 And ssDNAs complementary with
antigen (PSMA) positive exosomes demonstrate obvious aptamers were divided into two parts in order to decrease the
merits in PCa diagnosis, and offer essential biomarkers for hybridization energy with aptamers, and further increase the
the development of biosensor-based PCa detection methods.17 competition capacity of exosomes to replace ssDNAs from
Although increasing studies have focused on urinary aptamers, as well as double the exosomes signal (single
exosomes, it is still challenging to isolate and detect exosomes exosome produces two ssDNA signal) which could be helpful
from the complex matrix. To date, conventional methods for when exosomes concentration was low. According to previous
exosomes detection include transmission electron microscopy studies,30 two hairpin DNA probes (HP1, HP2) were designed
(TEM), nanoparticle tracking analysis (NTA), Western blot, to initiate amplification cycle and detection of released ssDNAs
and ELISA which are either time-consuming, unwieldy, with low concentration. Particularly, HP2 was designed as a
inconvenient, or expensive.18,19 The main techniques for molecular beacon, an “on−off” fluorescent probe which has
highly efficient separation of exosomes are ultracentrifugation, been widely used in various biomarkers detections.31,32 The
immunoaffinity capture, exosome precipitation and micro- high sensitivity and efficiency of this platform were verified in
fluidics-based methods.20−22 In addition, ultracentrifugation both cell medium and urine samples. In addition, different
serves as the gold standard for total exosomes isolation, while from other irreversible immunoaffinity isolation methods,
immunoaffinity method can accurately capture exosomes with restriction sites were introduced to purify target exosomes
specific surface proteins, but both of them are expensive, from the superparamagnetic platform. Finally, exosomes were
irreversible and monofunctional in clinical separation.23 further studied for cancerous properties. Above all, this
Recently, convenient and quick exosomes analysis methods platform can be used to isolate and detect PCa related
with high sensitivity and specificity have been reported. exosomes, revealing application potential in prostate cancer
Researchers prefer to isolate exosomes by magnetic separation diagnosis and prognosis.
1434 DOI: 10.1021/acssensors.9b00621
ACS Sens. 2019, 4, 1433−1441
ACS Sensors Article

Figure 2. Characterization of SMC-MB. (a) FTIR spectrum of Fe3O4 cores and conjunctions. (b) Absorbance intensity in supernatant before and
after aptamers reaction with Fe3O4 cores. (c) PAGE gel of different samples: Lane 1, standard DNA markers; lane 2, 0.5 mM ssDNA1; lane 3, 0.5
mM ssDNA2; lane 4, supernatant after ssDNA1 incubation; lane 5, supernatant after ssDNA2 incubation. (d) Absorbance intensity in supernatant
before and after ssDNA incubation. (e) Zeta potential of superparamagnetic Fe3O4 cores, Fe3O4-EDC, and Fe3O4 cores-aptamer and SMCs. (f)
Hydrodynamic diameters of superparamagnetic Fe3O4 cores and SMCs. (g) TEM image of superparamagnetic Fe3O4 cores. (h) TEM image of
superparamagnetic conjunctions (SMCs).

■ RESULTS AND DISCUSSION


Principle of SMC-MB Platform. The capture and
to form the HP1−HP2 complex thereby turning on the
fluorescence signal. Exact target ssDNAs allowed the initiation
detection mechanism of the SMC-MB platform for PCa of the amplification process and produced detection signals.
related exosomes is exhibited in Figure 1. SMCs were Exosomes separated by magnet were further purified by
composed of superparamagnetic Fe3O4 cores and specific Endonuclease EcoRI corresponding to restriction sites
aptamer-ssDNA complex (Figure 1a). All sequences applied in designed in aptamers, avoiding the drawbacks of irreversible
this paper are listed in Supporting Information Table S1. After binding of immunoaffinity capture (Figure 1d).
incubation with exosomes samples, two steps were carried out Characterization of Superparamagnetic Conjunc-
to quantify and purify exosomes, respectively. Due to higher tions. To validate the formulation of SMCs, FTIR absorbance
affinity between exosomes and aptamers, ssDNAs were was detected to prove the formation of amido bonds between
replaced and disassociated into supernatant and subsequently nanoparticles and aptamers. At the same time, the absorbance
separated from SMCs by magnet (Figure 1b). In such a intensity at 260 nm was used to quantify the aptamers before
pattern, the quantity of released ssDNAs could represent that and after amide reaction in the supernatant, an indirect
of exosomes. Particularly, ssDNA was composed of a quantification method of aptamers combination. Polyacryla-
combination and recognition part which is complementary to mide gel electrophoresis (PAGE) and absorbance detection
aptamer and HP1, respectively. HP2 is a molecular beacon were utilized to identify ssDNAs hybridization efficiency. As
designed based on the Förster resonance energy transfer displayed in Figure 2a, the peak shift from 1690 to 1630 cm−1
(FRET) effect between the quencher (BHQ) and fluorescent was attributed to the formation vibration of the carbonyl (C
dye (FITC) at separate ends of the hairpin structure (Figure O) of the amido bond. And the peaks at 1380 and 3100 cm−1
1c). Owing to the stronger affinity, HP2 took place of ssDNAs were ascribed to the bending vibrations of C−N bonds and
1435 DOI: 10.1021/acssensors.9b00621
ACS Sens. 2019, 4, 1433−1441
ACS Sensors Article

Figure 3. Validation of amplification platform for ssDNA detection. (a) PAGE gel of different samples in the amplification system: Lane 1, standard
DNA markers; lane 2, 1 μM of HP1; lane 3, 1 μM of HP2; lane 4, 1 μM HP1 incubated with 1 μM HP2 for 1 h. lane 5, 1 μM HP1 incubated with 1
μM of random DNA and 1 μM HP2 for 1 h. Lane 6, 1 μM of HP1 incubated with 1 μM of target DNA and 1 μM of HP2 for 1 h. (b) Fluorescence
intensity of molecular beacon in different samples corresponding to (a). (c) Relationship between target DNA concentration and fluorescence
intensity of molecular beacon. (d) Linear relationship between logarithmic target DNA concentrations and fluorescence intensity.

stretching vibrations of N−H bonds in the conjunction. All the particle-aptamers and ssDNAs conjunctions exhibited average
specific groups in the FTIR spectrum indicated the covalent diameters around 20 nm, as evidenced by dynamic light
conjunction of aptamers and Fe3O4 cores. The intensity of scattering (DLS) and TEM in Figure 2f and g,h, which indicate
aptamers in the supernatant decreased after 12 h reaction, that SMCs had ultrasmall diameters and the binding of
indicating the binding of aptamers to superparamagnetic Fe3O4 aptamers and ssDNAs led to a slight increase to Fe3O4 cores,
cores, and the binding efficiency was about 80%, calculated which are 10 nm in diameter. Superparamagnetic particles with
from absorbance peak value (Figure 2b). On the other hand, small size were more easily redispersed and exhibited better
Fe3O4 cores directly incubated with aptamers without solubility in aqueous solution, as well as stronger magnetism.
condensing agent caused no decrease in supernatant, which Instead, some commercial magnetic beads can easily
suggested ignorable nonspecific attachment in the reaction precipitate or form large complexes, which display disadvan-
system (Figure S1). Figure 2c and d describes the hybrid- tages on purification and separation operations.
ization of ssDNA1 and ssDNA2 with PAGE as well as a UV− Feasibility of Amplification Method for ssDNA
vis spectrograph. The bands of lane 2 and lane 3 refer to two Detection. The quantity of exosomes in real samples might
ssDNA standards before reaction, while lane 4 and lane 5 are result in extremely low concentration of replaced ssDNAs.
the supernatant after two ssDNAs react with aptamers. The Thus, the DNA amplification method played a vital role in
weakened signal suggested successful conjugation of ssDNAs exosome detection. Taking the advantages of base pairing
and aptamers. The hybridization efficiency calculated by gray principle and stem-loop structure of DNA realized the
value and absorbance peak are shown in Figure S2, which nonenzymatic amplification of target ssDNAs. First of all,
achieved similar efficiency results as approximately 70%. The hairpin DNA HP1 and HP2, as well as HP1-HP2 complex
zeta potential of the superparamagnetic Fe3O4 cores was about were determined by PAGE assay, which confirmed the
−9.8 mV due to carboxyl groups functionalized on the surface feasibility of the amplification strategy. In the absence of
of Fe3O4 particles. After modification with aptamers and target ssDNAs, no HP1-HP2 complexes were formed, and only
ssDNAs, the zeta potential decreased gradually to −16 and target ssDNAs could trigger the amplification circle. Random
−23 mV (Figure 2e), which may result from the negative DNAs were set as contrast group (Figure 3a). Oligo Analyzer
charge of ssDNAs. On the other hand, superparamagnetic 3.1 was used to calculate the relevant parameters T m
1436 DOI: 10.1021/acssensors.9b00621
ACS Sens. 2019, 4, 1433−1441
ACS Sensors Article

Figure 4. Detection of PCa related exosomes. (a) Dilution and calibration curve of exosomes from LNCaP cells diluted by exosome-free urine and
PBS. (b) Linear relationship between fluorescence intensity and logarithmic exosomes count. (c) Fluorescence intensity of exosomes from different
cell cultures medium. (d) Western blot of PSMA expression in exosomes from different cell cultures medium. (e) TEM image of SMC-MB
captured exosomes.

(annealing temperature), ΔG (Gibbs free energy), ΔH linear relationship between logarithmic target ssDNA concen-
(enthalpy change), and ΔS (entropy change) of hairpin probes tration and fluorescence intensity (Figure 3c, d), demonstrat-
HP1 and HP2 (Table S2), and predicted the stability of ing the effectiveness and practicability of the amplification
oligonucleotides. As displayed in Figure S3, the hairpin method in ssDNA detection.
structures HP1 and HP2 were stable enough at 37 °C with PCa Related Exosomes Detection Efficiency by SMC-
hybridization of 14 and 11 pairs of bases in the stem parts. The MB. Based on the optimized conditions, we developed and
Tm values of HP1, HP2, and HP1−HP2 duplex calculated by characterized an analysis platform for PCa related exosomes
RNA structure were 73.3, 69.0, and 71.6 °C (CNa+ = 25 mM, with the PSMA aptamer. To investigate detection efficiency of
CMg2+ = 4.5 mM), respectively, indicating the stability of HP1 the novel platform, LNCaP cell derived exosomes were
and HP2. Furthermore, the hybridization between bases in enriched by ultracentrifugation and gradient diluted to exact
single DNA was superior to that of bases from different DNAs. concentrations with exosome-free urine and PBS separately. As
Additionally, the mixture of HP1 and HP2 could regain the shown in Figure 4a, the fluorescent signal elevated along with
fluorescence signal as temperature reached 50 °C, revealing the the increasing exosome concentrations, which resulted from
temperature should be carefully controlled (Figure S4). The increasing ssDNAs released from the SMCs. Similar to the
optimized incubation time and ratio are further discussed in relationship between the concentration of ssDNAs and the
Figures S5 and S6, which was 1 h for incubation with a ratio of fluorescence signal of HP1−HP2 complexes (Figure 4b), a
1.5:1 for HP1 and HP2. To simplify the detection method of linear relationship was obtained between fluorescent signal and
amplified ssDNAs, HP2 was designed as a molecular beacon, the logarithmic exosomes counts ranging from 103 to 108 /μL,
which had no fluorescence signal in the stem-loop structure with a correlation coefficient (R2) of 0.989. Owing to the
owing to the FRET between the fluorescent group FITC and interference of urine itself, the limit of detection (LOD) was
the quenching group BHQ, while it regained fluorescence in estimated and calculated to be ∼100 particles/μL, much higher
the HP1−HP2 complex because of the remoteness of the two than the detection limit in PBS solution (∼50 particles/μL)
groups. Similarly, the relevant fluorescence at the wavelength due to the complex urine background. However, in accordance
of 525 nm indicated the quantity of the HP1−HP2 complex with the results of several studies and our research, the
and was presented by target DNAs (equimolar mixture of concentration of urine exosomes is far beyond the calculated
ssDNA1 and ssDNA2). The fluorescence responses of different LOD in this experiment, revealing the applicability in urine
samples are described in Figure 3b. Obviously, there was a exosomes detection. Furthermore, it should be noted that the
1437 DOI: 10.1021/acssensors.9b00621
ACS Sens. 2019, 4, 1433−1441
ACS Sensors Article

Figure 5. Comparison between ultracentrifugation and SMC-MB platform. (a) TEM image of exosomes purified by SMC and restriction enzyme.
(b) TEM image of exosomes isolated by UC. (c) Particle diameter of SMC-MB and UC separated exosomes determined by DLS. (d) Total protein
amount and particle concentration of exosomes separated by SMC-MB and UC. (e) Specific protein expression of exosomes separated by SMC and
UC.

Figure 6. Analysis of urine exosomes in PCa patients and healthy donors. (a) TEM images of immunogold labeled exosomes from PCa patients
and (b) healthy donors. (c) Specific protein expression of exosomes separated by SMC-MB in urine of PCa patients and healthy donors. (d)
Histogram and (e) box plots for fluorescence intensity of exosomes from PCa patients and healthy donors detected by SMC-MB platform. (f) PSA
and (g) PCA3 mRNA expression quantified by qRT-PCR in PSMA-positive exosomes. (*p < 0.05; **p < 0.01; ***p < 0.001).

dual function platform described here is simple and highly and detected by MB amplification cycle. The fluorescence
efficient without complicated instruments and expensive intensity of exosomes from human prostate cancer cell LNCaP
reagents. To further explore the specificity of the platform, was significantly higher than that of other cancer cells A549,
five cancer cell derived exosomes were incubated with SMC T24, HCT116, and HepG2 (Figure 4c), consistent with the
1438 DOI: 10.1021/acssensors.9b00621
ACS Sens. 2019, 4, 1433−1441
ACS Sensors Article

protein expression levels of PSMA (Figure 4d), which also and 50 healthy donors. The fluorescent signal of the PCa
proved the aptamer applicability in this platform. Figure 4e group obtained from the SMC-MB detection platform was
displays typical binding morphology of SMCs and exosomes, significantly higher than that of the healthy group (P < 0.05),
indicating successful capture of exosomes by SMCs. indicating the quantity of PSMA-positive exosomes in urine of
Exosome Isolation Efficiency Compared to Ultra- PCa patients discriminated from that of cancer-free donors
centrifugation. With the benefit of the restriction site of (Figure 6d, e). In addition, the total exosomes concentrations
aptamers, exosomes captured by SMCs could be easily showed no significant difference between healthy donors and
separated from particles. And the morphology and size PCa patients detected by NTA (particle diameter: 30−150 nm,
distribution of exosomes isolated by SMCs followed by Figure S7). However, the PSMA-positive exosomes concen-
enzyme restriction were determined by TEM and DLS. tration isolated by SMC-MB was much higher in the PCa
Considering that ultracentrifugation (UC) is the gold standard group than in the healthy group. Therefore, PSMA-positive
isolation method in current exosome studies, which is exosomes reveal their potential as PCa biomarkers. In
employed by over 50% of researchers, an ultracentrifugation consideration of the accepted biomarkers in clinical prediction
group was utilized to compare with SMC-MB platform to of prostate cancer including PSA and PCA3, qRT-PCR was
further illustrate capture capacity. Thus, LNCaP cell derived applied to detect PSA and PCA3 mRNA levels in exosomes
exosomes were isolated by UC, and further resuspended in isolated by our platform. As a result, PSA mRNA signal
exosomes-free urine followed by SMC-MB treatment. Gen- presented in 18 PCa cases and PCA3 mRNA signals presented
erally, UC and SMCs isolated exosomes were both cap-shaped in 19 PCa cases (Figure 6f, g), indicating the exosomes
with average diameters of 30−150 nm (Figure 5a, b). captured by SMC-MB were prostate cancer related. These
However, exosomes in UC precipitate inevitably existed with findings proved that higher specificity was achieved in the
protein aggregations and extracellular vesicles (EVs, more than newly developed platform, and the molecular signatures of
200 nm in diameter), leading to a complex background and individual exosomes were also beneficial in PCa diagnosis,
larger diameter determined by DLS (Figure 5c). On the which further confirmed the prospective diagnosis value of
contrary, exosomes obtained by the SMCs isolation method PSMA-positive exosomes for prostate cancer.


were observed to have fewer impurities and smaller size. Also,
the total protein and particle concentration of exosomes were CONCLUSION
analyzed by BCA protein determination and NTA in both UC
and SMC-MB groups. As a result, more protein was acquired A dual functional platform for exosomes detection and
by UC (Figure 5d). It may be induced by protein bundles and isolation is presented. In this paper, we introduce an
EVs resulting from a nonspecific isolation method. Meanwhile, aptamer-based strategy to separate exosomes from urine
there was no big difference of particle concentration between samples and convert exosomes detection into ssDNA detection
two groups. Exosome-specific proteins PSMA, CD63, and CD9 by virtue of a signal amplification method. In the amplification
detected by Western blot further confirmed the successful cycle, two hairpin DNAs were successfully designed to produce
exosomes isolation (Figure 5e) ,displaying the exosomes a stronger signal. Consequently, the superparamagnetic
isolation capability of the SMC-MB platform. conjunction-molecular beacon (SMC-MB) platform embodied
PCa Detection Efficiency via SMC-MB Platform. profound capture capacity and detection efficiency, thereby
Considering the platform exhibits excellent properties of achieving ideal effects in urine exosome analysis. The LOD of
exosome isolation and detection, SMC-MB was further applied SMC-MB in urine samples was as low as 100 particles/μL,
to capture and detect exosomes related to prostate cancer which was competitive in current urine exosomes detection
(PCa) in urine samples. Prostate specific membrane antigen methods.33 Compared to ultracentrifugation, SMC-MB iso-
(PSMA) was regarded as a specific PCa biomarker in a number lated exosomes were purer, containing fewer proteins and EVs.
of research studies, with higher expression in prostate cancer. Most importantly, the PSMA aptamer based SMC-MB
First, immunogold labeling of PSMA in exosomes from urine platform was useful to separate PSMA positive exosomes
samples of healthy donors and prostate cancer subjects is from urine, which is highly expressed in prostate cancer
demonstrated by TEM in Figure 6a and b, respectively. patients, and possesses great value in prostate cancer detection.
Apparently, PSMA antibody labeled gold nanoparticles In summary, the SMC-MB platform is a promising method for
anchored on some of the exosomes from PCa patients while exosome studies and cancer diagnosis.
being irregularly distributed in cancer-free samples. Accord-
ingly, the SMC-MB platform mentioned above was directly
applied to analyze PSMA-positive exosomes in urine samples
■ EXPERIMENTAL SECTION
Preparation of Superparamagnetic Conjunctions (SMCs).
of PCa patients and healthy donors, and the diagnosis Carboxyl-group-functionalized superparamagnetic Fe3O4 nanopar-
efficiency of PCa was evaluated. Exosomes separated by the ticles (50 μL, 4 mg/mL) were incubated with 3-ethylcarbodiimide
SMC platform also matched the established exosomal hydrochloride (EDC) and N-hydroxysulfosuccinimide sodium (NHS)
morphology and size. The PSMA, CD63, and CD9 protein at a molar ratio of 1:2:3 at room temperature for 1 h. Next, the
expressions were determined to explain the validity of the activated superparamagnetic Fe3O4 nanoparticles were purified via
SMC-MB platform. It was obvious that PSMA-positive magnetic separation and resuspended in 200 μL of diethyl
exosomes also harbored exosomes specific proteins. However, pyrocarbonate (DEPC) treated water. Then, 10 μL of 0.5 mM
amino-modified aptamer was added in the solution and incubated for
since our platform was designed for PSMA positive exosomes,
12 h with gentle shaking. After that, the mixture was purified by
which was extremely low in healthy donors, specific protein magnetic separation and washed three times. Afterward, 10 μL of 0.5
expressions of exosomes captured by SMC in healthy urine mM complementary ssDNA1 and ssDNA2 was added successively
were hard to detect as shown in Figure 6c. To further explore after magnetic purification and incubated at room temperature for 2 h.
the efficiency of PSMA-positive exosomes in urine for PCa Finally, the resuspended solution (200 μL) was stored at 4 °C after
diagnosis, we collected urine samples from 20 PCa patients magnetic separation.

1439 DOI: 10.1021/acssensors.9b00621


ACS Sens. 2019, 4, 1433−1441
ACS Sensors Article

Cell Culture and Exosomes Extraction. Exosome-free fetal calf were stored at −20 °C. The qRT-PCR process was conducted by the
serum (FBS) was prepared by ultracentrifugation at 120 000g at 4 °C following method: 5 μL of 2× AceTaq Master Mix, 0.2 μL of universal
overnight. LNCaP cells were cultured in RPMI-1640 medium forward primer (10 μM), 0.2 μL of reverse primer (10 μM), 1 μL of
(Invitrogen) and A549, T24, HCT116, and HepG2 cells were RT product, 0.5 μL of U-MB (10 μM), and DEPC-treated water were
grown in DMEM medium. Both media were supplemented with 10% mixed to a final volume of 10 μL. The qRT-PCR process was
exosome-free FBS, (Gibco), penicillin (100 μg/mL), and streptomy- conducted under the following conditions: 95 °C for 5 min, 35 cycles
cin (100 μg/mL). All cells were maintained in an incubator with 5% of 95 °C for 10 s, 45 °C for 30 s, and 60 °C for 30 s. The fluorescence
CO2 at 37 °C. The supernatant was first centrifuged at 300g for 10 signal was detected at 45 °C.
min, 2000g for 20 min, and 10 000g for 30 min sequentially, in order
to remove cells, cellular debris, and large extracellular vesicles.
Enriched exosome samples were diluted to different concentrations
with PBS and exosome-free urine.

*
ASSOCIATED CONTENT
S Supporting Information
The Supporting Information is available free of charge on the
Urine Samples Collection. Exosome-free urine was prepared by
ultracentrifugation at 120 000g at 4 °C overnight. According to the ACS Publications website at DOI: 10.1021/acssen-
experimental implementation plan, and the norms and reasonable sors.9b00621.
operating procedures of collection, the whole process strictly followed Sequence information on hairpin DNA probes and other
the relevant ethical requirements and all volunteers signed the oligonucleotides, structure and relevant parameters of
informed consent before sample collection. Twenty patients with hairpin DNA probes, fluorescence intensity of amplifi-
locally advanced PCa provided 10 mL urine in mid-morning or late cation products (HP1−HP2 complex) with different
morning. Fifteen healthy male volunteers samples were collected from
the China Pharmaceutical University students and teaching staff
incubation time and ratio, quantification methods for
whose physical examination and biochemical test results showed no incubation efficiency and fluorescence intensity of
exception. different substrates at different temperature, comparison
Isolation and Detection of Exosomes. 100 μL of SMCs was of total exosome concentration from healthy donors and
added into 10 mL of exosome samples at room temperature for 2 h PCa patients (PDF)


with gentle shaking to release ssDNAs. The SMCs captured exosomes
on their surfaces were separated by magnets and redispersed in 100 AUTHOR INFORMATION
μL of PBS. Then 10 μL of HP1 (100 μM), 15 μL of HP2 (100 μM),
equivalent 0.5× Nt.BstNBI buffer, and 1× ThermoPol buffer were Corresponding Author
introduced to 50 μL of supernatant and incubated at 37 °C for 1 h at *E-mail: guengineering@cpu.edu.cn.
37 °C for 60 min. The fluorescence of molecular beacon was ORCID
measured by using a fluorescence spectrophotometer. The SMC Yueqing Gu: 0000-0003-1315-4618
suspension with exosomes was mixed with 0.03 U/μL EcoRI
endonuclease in Nt.BstNBI buffer and incubated at 37 °C for 30 Author Contributions
min. Finally, extra superparamagnetic Fe3O4 nanoparticles were P.L. and X.Y. contributed equally. All authors contributed to
removed by magnets while purified exosomes were left in the the conception of the experiments and discussion of the results
supernatant and kept at −80 °C for further study. Ultracentrifugation and contributed to writing the manuscript. All authors have
(UC) treated samples were centrifuged at 120 000g for 2 h and given approval to the final version of the manuscript.
resuspended with 100 μL of PBS. Notes
Western Blot. Purified exosomes were mixed with loading buffer
The authors declare no competing financial interest.


and heated to 100 °C for 15 min with loading buffer, and Western
blot analysis was carried out on 8% SDS−PAGE followed by
electrotransfer onto nitrocellulose filters. After blocking for 1 h in TBS ACKNOWLEDGMENTS
with 0.1% Tween 20 (TBST) and 5% skim milk, the membranes were The authors sincerely acknowledge the financial support from
incubated overnight with specific primary antibody in TBST Natural Science Foundation of China (NSFC 81729002,
containing 5% BSA. Detection was carried out by the use of a 91859204, 81727804), the 973 Key Project (2015CB755504),
HRP-labeled secondary antibody (Bioworld Technology) and State key laboratory of Natural Medicines
developing agent (Thermo). Western blot was scanned by Quantity (SKLNMZZCX201819) and the Priority Academic Program
One Imaging Software from Bio-Rad.
Development of Jiangsu Higher Education.


Measurement of Exosome Size and Concentration. The
particle size and concentration were determined by dynamic light
scattering (Litesizer 500, Anton Paar) and with a NanoSight REFERENCES
instrument (TM LM10-HS, Malvern). The exosomes samples were (1) Trajkovic, K.; Hsu, C.; Chiantia, S.; Rajendran, L.; Wenzel, D.;
diluted with PBS to comply with the best working range of the Wieland, F.; Schwille, P.; Brugger, B.; Simons, M. Ceramide triggers
analysis software. Next, the diluted samples were injected into the budding of exosome vesicles into multivesicular endosomes. Science
laser pool. The mode size, mean size, and particulate concentration in 2008, 319 (5867), 1244−7.
each 1 mL sample were analyzed by NTA 3.0 analysis software. All (2) Janowska-Wieczorek, A.; Wysoczynski, M.; Kijowski, J.;
the measurements were performed for 60 s and repeated in triplicate. Marquez-Curtis, L.; Machalinski, B.; Ratajczak, J.; Ratajczak, M. Z.
Total RNA Isolation, cDNA Synthesis, and qRT-PCR. Total Microvesicles derived from activated platelets induce metastasis and
RNA was extracted from the exosomes using the Trizol Plus RNA angiogenesis in lung cancer. Int. J. Cancer 2005, 113 (5), 752−760.
purification kit (Life Technologies) according to manufacturer’s (3) Lötvall, J.; Hill, A. F.; Hochberg, F.; Buzás, E. I.; Vizio, D. D.;
protocol. The detection included reverse a transcription (RT) process Gardiner, C.; Yong, S. G.; Kurochkin, I. V.; Mathivanan, S.;
and quantitative real time polymerase chain reaction (qRT-PCR) Quesenberry, P. Minimal experimental requirements for definition
process. The RT process was conducted by the following method: of extracellular vesicles and their functions: a position statement from
4 μL of 5× RT buffer, 1 μL of dNTP Mix (10 nM each), 1 μL of RNA the International Society for Extracellular Vesicles. J. Extracell Vesicles
inhibitor (40 U/μL), 1 μL of RT primer (20 nM), 1 μL of M-MLV 2014, 3, 26913.
(H−) reverse transcriptase (100 U/μL), total RNA extracted from (4) Puhka, M.; Takatalo, M.; Nordberg, M. E.; Valkonen, S.;
real samples, and DEPC-treated water were mixed to a final volume of Nandania, J.; Aatonen, M.; Yliperttula, M.; Laitinen, S.; Velagapudi,
20 μL. The RT process was conducted under the following V.; Mirtti, T.; et al. Metabolomic Profiling of Extracellular Vesicles
conditions: 45 min at 42 °C and 15 min at 70 °C. The RT products and Alternative Normalization Methods Reveal Enriched Metabolites

1440 DOI: 10.1021/acssensors.9b00621


ACS Sens. 2019, 4, 1433−1441
ACS Sensors Article

and Strategies to Study Prostate Cancer-Related Changes. Thera- (23) Shao, H.; Im, H.; Castro, C. M.; Breakefield, X.; Weissleder, R.;
nostics 2017, 7 (16), 3824−3841. Lee, H. New Technologies for Analysis of Extracellular Vesicles.
(5) Zaborowski, M. P.; Balaj, L.; Breakefield, X. O.; Lai, C. P. Chem. Rev. 2018, 118 (4), 1917−1950.
Extracellular Vesicles: Composition, Biological Relevance, and (24) Jeong, S.; Park, J.; Pathania, D.; Castro, C. M.; Weissleder, R.;
Methods of Study. BioScience 2015, 65 (8), 783−797. Lee, H. Integrated Magneto-Electrochemical Sensor for Exosome
(6) Kahlert, C.; Kalluri, R. Exosomes in tumor microenvironment Analysis. ACS Nano 2016, 10 (2), 1802−1809.
influence cancer progression and metastasis. J. Mol. Med. 2013, 91 (25) Lewis, J. M.; Vyas, A. D.; Qiu, Y.; Messer, K. S.; White, R.;
(4), 431−437. Heller, M. J. Integrated Analysis of Exosomal Protein Biomarkers on
(7) Tao, X.; Liao, S.; Wang, S.; Wu, D.; Wang, Y. Body Compatible Alternating Current Electrokinetic Chips Enables Rapid Detection of
Thermometer Based on Green Electrolytes. ACS Sensors 2018, 3 (7), Pancreatic Cancer in Patient Blood. ACS Nano 2018, 12 (4), 3311−
1338−1346. 3320.
(8) Mathivanan, S.; Fahner, C. J.; Reid, G. E.; Simpson, R. J. (26) Zhang, P.; He, M.; Zeng, Y. Ultrasensitive microfluidic analysis
ExoCarta 2012: database of exosomal proteins, RNA and lipids. of circulating exosomes using a nanostructured graphene oxide/
Nucleic Acids Res. 2012, 40, D1241−D1224. polydopamine coating. Lab Chip 2016, 16 (16), 3033−3042.
(9) Valadi, H.; Ekstrom, K.; Bossios, A.; Sjostrand, M.; Lee, J. J.; (27) Wang, Z.; Zong, S.; Wang, Y.; Li, N.; Li, L.; Lu, J.; Wang, Z.;
Lotvall, J. O. Exosome-mediated transfer of mRNAs and microRNAs Chen, B.; Cui, Y. Screening and multiple detection of cancer
is a novel mechanism of genetic exchange between cells. Nat. Cell Biol. exosomes using an SERS-based method. Nanoscale 2018, 10 (19),
2007, 9 (6), 654−659. 9053−9062.
(10) Balaj, L.; Lessard, R.; Dai, L.; Cho, Y. J.; Pomeroy, S. L.; (28) Chen, X.; Lan, J.; Liu, Y.; Li, L.; Yan, L.; Xia, Y.; Wu, F.; Li, C.;
Breakefield, X. O.; Skog, J. Tumour microvesicles contain Li, S.; Chen, J. A paper-supported aptasensor based on upconversion
retrotransposon elements and amplified oncogene sequences. Nat. luminescence resonance energy transfer for the accessible determi-
Commun. 2011, 2 (2), 180. nation of exosomes. Biosens. Bioelectron. 2018, 102, 582−588.
(11) Fais, S.; O’Driscoll, L.; Borras, F. E.; Buzas, E.; Camussi, G.; (29) Dong, H.; Chen, H.; Jiang, J.; Zhang, H.; Cai, C.; Shen, Q.
Cappello, F.; Carvalho, J.; Cordeiro da Silva, A.; Del Portillo, H.; El Highly Sensitive Electrochemical Detection of Tumor Exosomes
Andaloussi, S.; et al. Evidence-Based Clinical Use of Nanoscale Based on Aptamer Recognition-induced Multi-DNA Release and
Extracellular Vesicles in Nanomedicine. ACS Nano 2016, 10 (4), Cyclic Enzymatic Amplification. Anal. Chem. 2018, 90 (7), 4507−
3886−3899. 4513.
(12) Junker, K.; Heinzelmann, J.; Beckham, C.; Ochiya, T.; Jenster, (30) Zheng, A.-X.; Li, J.; Wang, J.-R.; Song, X.-R.; Chen, G.-N.;
G. Extracellular Vesicles and Their Role in Urologic Malignancies. Yang, H.-H. Enzyme-free signal amplification in the DNAzyme sensor
Eur. Urol. 2016, 70 (2), 323−331. via target-catalyzed hairpin assembly. Chem. Commun. 2012, 48 (25),
(13) Rigau, M.; Olivan, M.; Garcia, M.; Sequeiros, T.; Montes, M.; 3112−3114.
Colas, E.; Llaurado, M.; Planas, J.; de Torres, I.; Morote, J.; Cooper, (31) Lee, J. H.; Kim, J. A.; Kwon, M. H.; Kang, J. Y.; Rhee, W. J. In
C.; Reventos, J.; Clark, J.; Doll, A. The present and future of prostate situ single step detection of exosome microRNA using molecular
cancer urine biomarkers. Int. J. Mol. Sci. 2013, 14 (6), 12620−12649. beacon. Biomaterials 2015, 54, 116−125.
(14) Urquidi, V.; Rosser, C. J.; Goodison, S. Molecular diagnostic (32) Ge, Z.; Lin, M.; Wang, P.; Pei, H.; Yan, J.; Shi, J.; Huang, Q.;
trends in urological cancer: biomarkers for non-invasive diagnosis. He, D.; Fan, C.; Zuo, X. Hybridization Chain Reaction Amplification
Curr. Med. Chem. 2012, 19 (22), 3653−3663. of MicroRNA Detection with a Tetrahedral DNA Nanostructure-
(15) Logozzi, M.; De Milito, A.; Lugini, L.; Borghi, M.; Calabrò, L.; Based Electrochemical Biosensor. Anal. Chem. 2014, 86 (4), 2124−
Spada, M.; Perdicchio, M.; Marino, M. L.; Federici, C.; Iessi, E.; et al. 2130.
High Levels of Exosomes Expressing CD63 and Caveolin-1 in Plasma (33) Merchant, M. L.; Rood, I. M.; Deegens, J. K. J.; Klein, J. B.
of Melanoma Patients. PLoS One 2009, 4 (4), e5219−e5219. Isolation and characterization of urinary extracellular vesicles:
(16) Flanigan, R. C.; Catalona, W. J.; Richie, J. P.; Ahmann, F. R.; Implications for biomarker discovery. Nat. Rev. Nephrol. 2017, 13
Hudson, M. A.; Scardino, P. T.; Dekernion, J. B.; Ratliff, T. L.; (12), 731−749.
Kavoussi, L. R.; Dalkin, B. L.; et al. Accuracy of digital rectal
examination and transrectal ultrasonography in localizing prostate
cancer. J. Urol. 1994, 152 (5), 1506−1509.
(17) Mohan, K.; Donavan, K. C.; Arter, J. A.; Penner, R. M.; Weiss,
G. A. Sub-nanomolar detection of prostate-specific membrane antigen
in synthetic urine by synergistic, dual-ligand phage. J. Am. Chem. Soc.
2013, 135 (20), 7761−7767.
(18) Nawaz, M.; Camussi, G.; Valadi, H.; Nazarenko, I.; Ekström,
K.; Wang, X.; Principe, S.; Shah, N.; Ashraf, N. M.; Fatima, F.; et al.
The emerging role of extracellular vesicles as biomarkers for
urogenital cancers. Nat. Rev. Urol. 2014, 11 (12), 688−701.
(19) Im, H.; Shao, H.; Park, Y. I.; Peterson, V. M.; Castro, C. M.;
Weissleder, R.; Lee, H. Label-free detection and molecular profiling of
exosomes with a nano-plasmonic sensor. Nat. Biotechnol. 2014, 32
(5), 490−495.
(20) Liu, F.; Vermesh, O.; Mani, V.; Ge, T. J.; Madsen, S. J.; Sabour,
A.; Hsu, E. C.; Gowrishankar, G.; Kanada, M.; Jokerst, J. V.; et al. The
Exosome Total Isolation Chip. ACS Nano 2017, 11 (11), 10712−
10723.
(21) Shehadul Islam, M.; Aryasomayajula, A.; Selvaganapathy, P. A
Review on Macroscale and Microscale Cell Lysis Methods. Micro-
machines 2017, 8 (3), 83−110.
(22) Chiriacò, M.; Bianco, M.; Nigro, A.; Primiceri, E.; Ferrara, F.;
Romano, A.; Quattrini, A.; Furlan, R.; Arima, V.; Maruccio, G. Lab-
on-Chip for Exosomes and Microvesicles Detection and Character-
ization. Sensors 2018, 18 (10), 3175−3216.

1441 DOI: 10.1021/acssensors.9b00621


ACS Sens. 2019, 4, 1433−1441

You might also like