Download as pdf or txt
Download as pdf or txt
You are on page 1of 11

Anal Bioanal Chem (2011) 400:535–545

DOI 10.1007/s00216-011-4770-4

ORIGINAL PAPER

PEGylated polyethyleneimine grafted silica nanoparticles:


enhanced cellular uptake and efficient siRNA delivery
Haisung Lee & Dongkyung Sung &
Murugan Veerapandian & Kyusik Yun & Soo-Won Seo

Received: 12 November 2010 / Revised: 24 January 2011 / Accepted: 3 February 2011 / Published online: 22 February 2011
# Springer-Verlag 2011

Abstract The present paper reports the utilization of hybrid groups of SiO2NPs; resonance shifts and bending vibrations of
nanocomposite particles consisting of PEI25k-PEG5k copoly- PEI25k, –CH2–CH2–NH–; and PEG5k, –CH2–CH2–O–) from
mer grafted silica nanoparticles (SiO2NPs) for enhanced cellular copolymer nanoparticle. Stable complexation of siRNA and
uptake and siRNA delivery. High-resolution transmission nanocomposite particle (wt.%:wt.%) was achieved from 1:5 to
electron microscopy and dynamic light scattering measure- 1:15 ratio. Nanocomposite particle (N/P) ratio and siRNA
ments ensured the average particle size of the final hybrid concentration determine the stability and knockdown efficiency
component as 45 nm (core SiO2, 28–30 nm and shell PEI25k- of the PEI25k-PEG5k-graft-SiO2NPs–siRNA complexes. It
PEG5k, 12–15 nm). Surface morphology from atomic force was shown that highly positively charged (zeta potential,
microscopy analysis showed the significant relationship +66 mV) PEI25k-PEG5k-graft-SiO2NPs result in strong
between the particle size and shape. 29Si and 13C cross- affinity with negatively charged siRNA. Confocal microscopy
polarization–magic angle spinning solid state nuclear magnetic showed intensified cellular uptake of siRNA into cytoplasm of
resonance (NMR), 1H-NMR, and Fourier transform infrared A549 cancer cell utilized for in vitro study. In conclusion, the
spectroscopy were used to obtain the relevant structural coherence, graft density of copolymer-SiO2NPs, and siRNA
information (such as Q3, silanol; Q4, siloxane functional concentration were found to strongly influence the stability,
and hence determine the knockdown efficiency, of PEI25k-
PEG5k-graft-SiO2NPs–siRNA complexes.
Electronic supplementary material The online version of this article
(doi:10.1007/s00216-011-4770-4) contains supplementary material,
which is available to authorized users. Keywords siRNA . PEI25k-PEG5k-graft-SiO2NPs .
H. Lee and D. Sung contributed equally to this work. Cellular transfection . Low cytotoxicity

H. Lee : S.-W. Seo (*)


Interdisciplinary Program of Biomedical Engineering,
School of Medicine, Sungkyunkwan University, Introduction
50 Irwon-Dong, Gangnam-Gu,
Seoul 135-710, Republic of Korea The novel design of a nanoparticle delivery system for
e-mail: sswbme@skku.edu
nucleic acids such as small interference RNAs (siRNAs)
D. Sung has been studied due to their potential applications for in
Department of Life Science, vitro and in vivo experiments [1–4]. The protected and
The Graduate School of Korea University, successful intracellular delivery of siRNAs is the final goal
Anam-dong, Seongbuk-Gu,
to an extensive application, using siRNAs as therapeutic
Seoul 136-701, Republic of Korea
agents [5, 6]. A variety of nonviral delivery systems such as
M. Veerapandian : K. Yun (*) liposomes, polymers, cationic charged peptides, and gold
College of Bionanotechnology, nanoparticles has been utilized for enhanced intracellular
Gachon BioNano Research Institute, Kyungwon University,
siRNAs delivery [7, 8]. There are many different accessible
San 65 Bokjeong-dong, Sujeong-gu,
Seongnam City 460-701, Republic of Korea methods for making the complexes, using cationic carriers
e-mail: ykyusik@kyungwon.ac.kr such as polymers, liposomes, cationic peptides, and hard-
536 H. Lee et al.

sphere nanoparticles such as gold nanoparticles, quantum 97%, ethyl acetate (EA), hexane, and branched polyethylenei-
dots, and silica nanoparticles [9–15]. Functionalized nano- mine 25,000 (PEI25k) were obtained from Sigma. Mono-N-
particles can be placed in the core of the complex, and hydroxysuccinyl-polyethyleneglycol 5,000 (NHS-PEG5k) was
siRNAs make a position on the surface of cationic nano- purchased from Sun Bio Chemical company (South Korea).
particles. This method has been used for multifunctional All chemicals were purchased from commercial sources.
nanoparticles such as siRNA delivery and other diagnostics
methods: magnetic resonance (MR) imaging and fluores- Synthesis of derivative 1 (NHS-PEG5k and PEI25k)
cent imaging analysis [16–27]. Core-shell nanoparticles
with optimized size tuning and maintenance of charge A solution of NHS-PEG5k (50 mg, 10 mM) in anhydrous
intensity are the key factors to achieve successful delivery DCM (80 ml) was added to a solution of PEI25k (250 mg,
of siRNAs [28–30]. Recently, the functionalized silica 10 mM) in anhydrous DCM (80 ml) under 0 °C using ice-
nanoparticles via surface modification were released upon bath for cooling down. The mixture was stirred at room
invention of multi-modal and smart nanoparticles capable temperature then naturally increased from 0 °C for 12 h.
of searching for specific cancer cells or receptor and For checking of reaction progress, the TLC was used with
delivery to targeted location [31–33]. Even though, there eluent condition MeOH: DCM=1:1 (Rf value of NHS-
are several promising results that were obtained from the PEG5k (0.3∼0.4), PEI25k (0.5∼0.6), PEI25k-PEG5k mix-
functionalized nanoparticles (such as SiO2NPs) for drug ture (0.0∼0.1), and co-mixture of three (0.5∼0.6) samples
delivery and other biological application. The choice of were spotted and stained with iodine after 12 h of reaction
better delivery system with more specific stabilizing agent period). All the compounds showed a tailed spot because of
and biocompatible modification procedures is expected to their polar nature. It was finally washed with DCM three
acquire the complete pharmaceutical and pharmacological times. The solvent was evaporated with Speed-Vac and
value with minimal/no toxic effects. To do so, currently, the freeze-dried, and the crude compound was kept in −20 °C.
pharmaceutical and bioengineering scientists were involved The crude solid material was obtained after the evaporation
in finding new and innovative research schemes. Here, we of the solvent. It was finally purified by Sephadex LH20.
present a simple and fast method to fabricate core-shell
silica nanoparticles for siRNA delivery system. This Synthesis of derivative 2 (PEI25k-PEG5k and 3-GPTMS)
research is based on block-copolymer grafting method
using polyethyleneimine (PEI), polyethyleneglycol (PEG), A solution of PEI25k-PEG5k (100 mg) in DCM (80 mL) was
and epoxy functionalized silane to conjugate onto the added to a solution of 3-GPTMS (0.80 mL, 4.6 mM) in the
surface of silica nanoparticles. This hydrophilic and highly same solvent (20 mL) under 0 °C using ice-bath for cooling
positively charged surface modified silica nanoparticle can be down. The mixture was stirred at room temperature for 12 h.
used as a drug delivery system for anti-cancer drug, anti-
biotics, and therapeutic siRNAs. Techniques such as cross- Synthesis of derivative 3 (PEI25k-PEG5k-graft-SiO2NPs)
polarization–magic angle spinning (CP-MAS) solid nuclear
magnetic resonance (NMR), 1H-NMR, high-resolution trans- Fifty-three milligrams of derivative 2 was placed in a round
mission electron microscopy (HR-TEM), Bio-atomic force bottom flask and then dissolved with 1.25 mL of ethanol,
microscopy (AFM), and dynamic light scattering (DLS) have 1.25 mL of water, and 1.25 mL of acetic acid. One hundred
been used to investigate the different properties of the twenty-five microliters of an aqueous suspension of
nanostructures. Furthermore, the significance of enhanced SiO2NPs was added. The reaction mixture was warmed at
cellular uptake, effective siRNA delivery with low cytotox- 80 °C under stirring for 48 h. After this time, ethanol was
icity, and higher knockdown efficiency of core-shell silica evaporated under reduced pressure, and solid NaHCO3 was
nanoparticles were compared with the PEI cationic polymer. added to the suspension to reach a pH value between 7 and
8. The precipitate was filtered and washed with borate
buffer (pH 9.5, 5×2 mL) and with water (5×2 mL). The
Materials and methods solid was dried under vacuum and redissolved with 100 mL
of DCM. The organic solution was washed with water
General (100 mL) and dried over Na2SO4.

Dichloromethane (DCM), NaHCO3, trimethoxy (3-(oxiran-2- Synthesis of derivative 4 (RITC-labeled


ylmethoxy)propyl)silane (3-GPTMS), silica nanoparticles PEI25k-PEG5k-graft-SiO2NPs)
(LUDOX AS-40 Colloidal Silica), sodium sulfate (Na2SO4),
dimethylsulfoxide (DMSO), rhodamine isothiocyanate (RITC), One hundred micrograms per milliliter colloidal solution of
DSS [3-(trimethylsilyl)-1-propane sulfonic acid], sodium salt PEI25k-PEG5k-graft-SiO2NPs was placed in a V-shaped
PEGylated polyethyleneimine grafted silica nanoparticles 537

small volume vial and then added with 5 mM RITC rate of 7 kHz. The spectrum width was 23.8 kHz; delay or
solution in DMSO under pH 7.4∼8.0. The reaction mixture repetition time was 5.0 s. Contact time was performed for
was stirred at room temperature for 12 h under dark room 5 ms. The 29Si spectrum was calibrated with the signal from
condition. After this time, the mixture was washed with using external standard DSS 0.00 ppm. In case of 13C solid-
DCM twice, and aqueous colloidal solution was separated state NMR experiments, it has been done with the same
by dialysis for 2 days. NMR instrument such as 29Si CP-MAS NMR analysis
operating at 100.690 MHz; 4-mm ZrO2 rotors were used,
Characterization of size and zeta potential of silica and magic angle spinning was carried out at a spinning rate
nanoparticles of 7 kHz. Two milliseconds contact time, 30.3 kHz spectrum
width, and 5.0 s delay or repetition time were used.
Size and zeta potential were analyzed using Malvern
Zetasizer Nano ZS series with a laser at 633 nm with a Cytotoxicity of silica nanoparticles
90° scattering angle. The concentration of analytical
samples was 0.1 mg/mL. Human alveolar basal epithelial cell (A549 cells) was
provided by the Korea Cell Line Bank. The A549 cells
AFM analysis were used for the in vitro study of the nanoparticles
toxicology. At day 1, 1.0×104 cells were placed in each
The AFM images were obtained by Nanowizard II (JPK; well of a 96-well plate in 100 μL of Dulbecco's modified
Germany). It was equipped with atomic lattice resolution on Eagle's medium containing 10% fetal bovine serum (FBS,
inverted microscope (<0.055 nm RMS z noise level). The purchased from GIBCO) and cultured for 24 h at 37 °C.
cantilever used in our experiment was Soft Tapping Mode Then, 100 μL of the colloidal solution of nanoparticles in
probes. pH 7.0 PBS was added to the cell culture plate for
incubation. The final concentrations of nanoparticles ranged
Solid-state NMR analysis from 25×10−3 to 0.10 mg/mL. The nanoparticles were pure
PEI25k-PEG5k-graft-SiO2NPs containing no fluorescent
Solid-state 29Si CP-MAS NMR analysis was performed dye molecules to stay away from interferences between
using a Bruker Avance 400 MHz spectrometer operating at the nanoparticles and the tetrazolium salt of a detection
79.545 MHz. The samples were located in 4-mm ZrO2 reagent. A prepared dye XTT solution was added to the
rotors. Magic angle spinning was carried out at a spinning plate wells, and then, the cells were incubated at 37 °C for

H O
N
NH2 N CH3
H O HO OH
n
N N
H
N HO SiO2 OH
N N HO OH
H n

N O
O Si CH3
EtOH, H 2O
H2N N
H
CH3
OH O O AcOH @ 80 Celcuis for 48hrs

CH3

PEG5K-PEI 25K-silane

O H
N
H3C N H2N
O H
n
H
N N N
N N
n H

O N
2 Si
O N NH2 PEG5K-PEI 25K layer
H
O
OH

PEG5K-PEI 25K-silica nanoparticle 2

Fig. 1 Synthetic scheme of PEG5k-PEI25k-graft-SiO2NPs


538 H. Lee et al.

4 h. In this period, the living cells converted the tetrazolium Agarose gel electrophoresis
component of the dye solution into a formazan product that
showed absorption at 470 nm in a UV–Vis spectrum. The The complexes of siRNA and PEI25k-PEG5k-graft-
formazan product was solubilized, and the absorbance at SiO2NPs were formulated with applicable N/P ratios.
470 nm was measured using a 96-well plate reader. The Afterwards, the complexes were applied to 1% (w/v)
experiment was repeated three times to get the average results. agarose gel contacting 0.5 μg/mL ethidium bromide (EtBr).

Fig. 2 a, b High-resolution
TEM images of PEI25k-
PEG5k-graft-SiO2NPs core-
shell nanoparticles. c DLS size
analysis and d DLS zeta poten-
tial analysis
PEGylated polyethyleneimine grafted silica nanoparticles 539

Table 1 Energy-dispersive X-ray spectroscopy (EDS) analysis of PEI25k-PEG5k-graft-SiO2NPs

Element Peak area Area sigma K factor Abs corrn. wt.% wt.% sigma Atomic %

Ck 355 54 2.208 1.000 15.28 2.07 23.11


Nk 177 54 2.965 1.000 10.23 2.83 13.27
Ok 893 59 1.810 1.000 31.54 1.95 35.83
Sik 2,202 82 1.000 1.000 42.95 2.11 27.79
Total 100
14
Si and 8 O atomic composition contributed more than half of the total atomic composition of PEI25k-PEG5k-graft-SiO2NPs. Then, 6 C and 7 N
contribution in total atomic composition was about half a portion. 6 C fraction in EDS was bigger than 7 N for the reason that hydrocarbon was included in
PEI as well as PEG. Furthermore, 8 O atomic composition was similar with 14 Si atomic composition because 8 O atomic composition was located in
SiO2NPs and PEG5k as well

RNase protection assay Cells were routinely cultivated at 37 °C in a humidified


atmosphere of 5% CO2. A549 cells were seeded in a 12-
siRNA (2 μg) and its complexes with PEI25k-PEG5k-graft- well culture plate at a density of 2×105 cells per well and
SiO2NPs were prepared just before the experiments. After allowed to attach overnight. Then, incubation medium was
the preparation of the complexes, each sample was incubated replaced with fresh media. The complex was prepared with
with 10 U of RNase ONE ribonuclease (Promega) for 1 h at PEI25k-PEG5k-graft-SiO2NPs and FITC-labeled siRNA
37 °C. Nucleic acid degradation was detected by gel via different conditions. The nucleus was stained by DAPI
electrophoresis. The levels of the remaining siRNAs were (blue), and the complex was represented with green through
quantified by Quantity One software (Bio-Rad, USA). FAM-labeled RNA-PEI25k-PEG5k-graft-SiO2NPs, and
then the membrane was colored with wheat germ aggluti-
Confocal laser scanning microscopic observation nin; transfection was carried out by adding the prepared
complex to the wells in a drop-wise manner. After 4 h
Human alveolar basal epithelial cell (A549 cells) was incubation, the medium was exchanged again with fresh
provided by the Korea Cell Line Bank. A549 cells were medium, and transfected cells were incubated for 24 h. A
cultured in RPMI 1640 medium (Invitrogen, USA) supple- series of images were obtained by using a confocal laser
mented with 10% heat-inactivated FBS and 1% antibiotics. scanning microscope (Bio-Rad, USA).

Fig. 3 29Si CP MAS NMR


spectrum of bare SiO2NPs, ami-
nopropyl functionalized
SiO2NPs, and PEI25k-PEG5k-
graft-SiO2NPs (a); 13C CP MAS
NMR spectrum of aminopropyl
functionalized SiO2NPs and
PEI25k-PEG5k-graft-SiO2NPs,
as well as SiO2NPs (b); and
600-MHz 1H-NMR NMR spec-
trum of PEI25k, PEG5k, as well
as PEI25k-PEG5k
copolymer (c)
540 H. Lee et al.

2000 analyzer (Bio-Rad, USA). The level of each gene


expression was semi-quantified by densitometric analysis
using the software. Relative expression levels were estimated
by the density ratio of human GAPDH to actin.

Results

The general procedure to fabricate core-shell SiO2NPs has


been performed via three simple steps. Initially, we
synthesized the block-copolymer consisting of PEI25k
and NHS-PEG5k (see Electronic Supplementary Material
Fig. S1); later, this copolymer is modified with the 3-
GPTMS (see Electronic Supplementary Material Fig. S2).
Then, the final PEI25k-PEG5k-graft-SiO2NPs have been
fabricated by conjugation of the copolymer (PEI25k-
Fig. 4 Cytotoxicity of PEI25k-PEG5k-graft-SiO2NPs in A549 lung PEG5k), which has been modified with a silane from 3-
cancer cell with XTT assay GPTMS, and this silane reacts with silica nanoparticles (see
Fig. 1). The synthesized PEI25k-PEG5k-graft-SiO2NPs
Reverse transcription-PCR method showed an average particle size of 45 nm by TEM and
DLS (Fig. 2a–c). The core part of the nanoparticles showed
Twenty-four-hour post-transfection with GAPDH, total RNA an average particle size of 28–30 nm, and the shell part
in the sample was extracted by using a RNA Trizol according (PEI25k-PEG5k block-copolymer) of the particle was 12–
to the manufacturer's protocol (Invitrogen). Total RNA 15-nm thick as measured from TEM analysis (Fig. 2b).
concentration was measured by spectrophotometer (Nano- PEGylated-PEI-SiO2NPs were analyzed with Nano ZS zeta
drop) at 260 nm. RNA was used to produce cDNA with an potential analyzer in neutral pH (7.0). The zeta potential
iScriptTM cDNA synthesis kit (Bio-Rad). Polymerase chain became more positive such as +66 mV in pH 7 (Fig. 2d). For
reaction (PCR) primers human glyceraldehyde-3-phosphate the detailed atomic analysis of the PEGylated-PEI polymer
dehydrogenase (GAPDH) and actin were synthesized from shell layer and silica core particle, energy-dispersive X-ray
Bioneer Inc. The sequence of primers used was as follows: spectroscopy atomic analysis has been done. Table 1
human GAPDH (sense, GGTCGGAGTCAACGGATTTG; demonstrated the atomic composition (wt.%) of 14Si, 8O,
antisense, CCTCCGACGCCTGCTTCACC) and actin (sense, and 6C in the part of core-shell silica nanoparticles. The
AGGGAAATCGTGCGTGACAT; antisense, CATCTGCTG- morphology of PEI25k-PEG5k-graft-SiO2NPs was further
GAAGGTGGACA). For each reaction, 1 μL of cDNA was observed from AFM. Figure S3 in the Electronic Supple-
placed in a 19-μL reaction mixture PCR Master Mix mentary Material shows the two- and three-dimensional
(Bioneer, Korea) and 0.5 μM of each primer. All amplification topography of PEI25k-PEG5k-graft-SiO2NPs. From the
reactions were performed in T1 thermocycler. PCR products AFM study, the size of the particles was measured to be
were run on a 1.0% agarose gel electrophoresis and were around 40–50 nm; this is in good relation with the other
visualized by ethidium bromide and scanned by a Gel Doc measurements.

Fig. 5 Gel retardation assay of the different N/P ratio complexes and incubation with RNase and RNA-PEI25k-PEG5k-graft-SiO2NPs, low
RNase protection assay. a Agarose gel electrophoresis of PEI25k- N/P ratio siRNA complex was degraded by RNase, but high N/P ratio
PEG5k-graft-SiO2NPs–RNA complex at different N/P ratio. Com- siRNA was protected by RNase
plexes were prepared at N/P ratios range from 1 to 30. b After 1 h
PEGylated polyethyleneimine grafted silica nanoparticles 541

c 100 d
110
90
100
80

Intensity of GAPDH
Intensity of GAPDH

90
70
80
60
70

50 60

40 50

30 40

0 50 100 150 200 250 300 NC RNA 1 5 7 10


Dose of siRNA (pmol) N/P ratio

Fig. 6 Semiquantitative RT-PCR analysis for the GAPDH mRNA knockdown. a, c A549 cells transfected naked RNA and complex prepared with
different siRNAs concentrations (25, 50, 100, 200, and 300 pmol). b, d Complexes were prepared at N/P ratios range from 1 to 10

Figure 3 shows relevant structural information with grafted onto the surface of SiO2NPs is evaluated to that of
solid-state NMR spectrum analysis in 29Si, 13C with CP/ the unmodified SiO2NPs and 3-aminopropyltriethoxysilane
MAS NMR (Fig. 3a, b), and 1H-NMR (Fig. 3c). Solid- (3-APTES)-modified SiO2NPs. The 29Si CP-MAS NMR
state NMR has been used as one of the key analytical spectrum of the unmodified silica nanoparticles shows three
methods to investigate the molecular structures and peaks at −100.2 and −108.9 ppm, which are assigned to be
dynamics of the surface-bound and intercalated organic Q3, free silanols, and Q4, siloxane functional groups. In
species [34, 35]. Owing to its easy interpretation of the 29Si CP-MAS NMR spectra of the modified silica
spectra, 29Si CP-MAS NMR is widely used to study the nanoparticles, two functional groups of peaks are detected
surface analysis of structural information from silica. The at all times. Comparing to the spectrum of unmodified
relation between Si chemical shifts and corresponding SiO2NPs, Q3 peak is enhanced, and Q4 peaks are reduced.
assigned structures onto the surface of silica is simple to The reaction of SiO2NPs surface with a triethoxysilyl
analyze. Figure 3a shows that 29Si CP-MAS NMR functional group is able to show the way to three available
spectrum of the different organosilanes and organic types of new chemical structures onto the SiO2NPs. The
functional groups in PEI25k-PEG5k copolymer materials T3 and T4 structures are the product of each reaction
Fig. 7 Confocal laser microsco-
py images of A549 cells trans-
fected with PEI25k-PEG5k-
graft-SiO2NPs. a Transfection
after 2 h incubation. b Trans-
fection after 4 h incubation
542 H. Lee et al.

individually, two and three ethoxy groups of the same XTT assay with different concentrations from 25 to 100 μg/
silane molecule with the silanols at the surface of mL. As we can see in Fig. 4, the PEI25k-PEG5k-graft-
SiO2NPs. It was noticed that a nice correspondence is SiO2NPs showed a significant low cytotoxicity in XTT
located between 29Si CP-MAS NMR analysis and 13C CP- assay. Thus, the SiO2NPs could be used as a possible
MAS NMR analysis (Fig. 3a, b). delivery system for siRNAs. In order exploit the cellular
1
H-NMR spectroscopy was used to elucidate the uptake and its efficiency towards the siRNA delivery, an in
structural information from the PEI25k, PEG5k, and vitro bioanalysis is done. The cell transfection efficiency has
PEGylated-PEI. The sharp peak at around 3.6 ppm is been tested through conjugated RITC red fluorescence dye
assigned to be PEG (–CH2–CH2–O–), and proton shift on the PEI25k-PEG5k-graft-SiO2NPs with further purifica-
located from 2.0 to 2.6 ppm is found to be associated with tion process: dialysis (MWCO, 50 K) for 2 days. For
PEI (–CH2–CH2–NH2–). From the proton signals identi- confirmation of the efficient transfection and delivery, we
fied from the current experiment, it is clear that the tested the gel retardation assay with different siRNA/particle
successful copolymerization exists between the ratios in weight percentage. The ratio of complex was
PEGylated-PEI groups (Fig. 3c). The analysis of Fourier increased gradually from 1 to 30 (Fig. 5a). The ratio 1:1=
transform infrared spectrum about unmodified SiO2NPs, siRNA:particles (wt.%/wt.%) seems to be an unstable
amine functionalized SiO2NPs, and PEI25k-PEG5k-graft- complex in solution and has no significant protection of
SiO2NPs was performed to obtain the stretching and siRNA. In the case of 1:3 ratio complexes, it was not clearly
bending vibrations before and after surface modification produced, which was characterized with tailed band in
of SiO 2NPs (see Electronic Supplementary Material agarose gel. The complex was formulated strongly from
Fig. S4). Our PEI25k-PEG5k-graft-SiO2NPs have been the 1:5 to 1:15 ratio. Figure 5b shows the low ratio complex
represented with the competitive low cytotoxicity in XTT which seems to be not protecting the RNase. In parallel, 1:5
assay with A549 lung cancer cell. We have performed the ratio complexes could protect from RNase.

Fig. 8 Confocal laser microsco-


py images of A549 cells trans-
fected with PEI25k-PEG5k-
graft-SiO2NPs-siRNA complex.
Complexes were prepared with
200 nM FAM-oligo at N/P ratio
of 1:10 and transfected into the
A549 cells for 4 h. Blue nucleus
(DAPI), green FAM-labeled
siRNA-PEI25k-PEG5k-graft-
SiO2NPs complexes, red mem-
brane (wheat germ agglutinin,
Alexa Fluor 594 conjugate).
Magnification, ×1,000
PEGylated polyethyleneimine grafted silica nanoparticles 543

In semiquantitative RT-PCR analysis for the GAPDH


mRNA knockdown, A549 cells were transfected with
naked RNA, and the complex was prepared with different
siRNA concentrations (25, 50, 100, 200, and 300 pM;
Fig. 6a, c). The knockdown efficiency was increased
through the change of siRNAs concentrations. In Fig. 6a, c,
there is not much difference between 0- and 50-pM
ranges. It was the high knockdown efficiency in the
300 pM of siRNA. Moreover, Fig. 6b, d showed that the
1:10 ratio complex worked properly to knockdown
GAPDH comparing beta-actin. Therefore, we optimized
the suitable N/P ratio (wt.%/wt.%) and siRNA concentra-
tion. Figure 7 ensures that the transfection of the particle
itself with red fluorescence and the nucleus was stained
with DAPI (blue). The silica particles during the 4-h incubation
were transfected better than that of the 2-h incubation. We
confirmed that our PEI25k-PEG5k-graft-SiO2NPs did have the
ability of transfection into A549 lung cancer cell without
difficulty.
Figure 8 describes that the cell transfection experiment
was performed via confocal laser scanning microscopy. The
cell transfection experiment has been performed with A549
cell by DAPI staining in nucleus into blue color and the
complex between FAM-labeled siRNA and PEI25k-
PEG5k-graft-SiO2NPs into green color; the membrane with
wheat germ agglutinin, Alexa Fluor 594 conjugate, was
colored red. The complex of FAM-labeled siRNA and
PEI25k-PEG5k-graft-SiO2NPs was transfected suitably in
the A549 lung cancer cell. The PEI25k-PEG5k block-
copolymer had been present that provided a significant
protection from RNase and knockdown efficiency in a few
research papers [36–40]. In relation to that, our PEI25k-
PEG5k-graft-SiO2NPs had shown a considerable efficiency
of protection and knockdown. From Fig. 9, the cytotoxicity
of PEI25k and PEI25k-PEG5k-graft-SiO2NPs provided the
merit of our core-shell SiO2NPs for siRNA delivery system Fig. 9 Cell cytotoxicity and knockdown efficiency between PEI25k
(Fig. 9a). In knockdown efficiency, our PEI25k-PEG5k- polymer and PEI25k-PEG5k-graft-SiO2NPs. a Cytotoxicity experi-
ment has been performed in fixed concentration (100 μg/mL of each
graft-SiO2NPs could have a priority to deliver siRNA under
delivery system). b GAPDH knockdown experiment has been done in
serum condition comparing PEI cationic polymer with 200 pM siRNA concentration, 1:10 N/P ratio condition. Each symbol
itself. The PEI25k-PEG5k-graft-SiO2NPs did knock down (triangle, rectangle, and circle, and so on) indicates each experiment.
GAPDH, about half of them; PEI25k only did knock down Experiment A (cytotoxicity) has been repeated seven times, and
experiment B (comparison of GADPH knockdown efficiency) has
GAPDH with about 20% in same condition (Fig. 9b). been repeated three times

Discussion MR imaging through paramagnetic oxide nanoparticles for


diagnostics applications under in vitro or in vivo conditions
We show that the SiO2NPs can be modified to deliver as reported before individually [41].
siRNA into cancer cell line with a significant low The highly positive-charged PEGylated-PEI copolymer
cytotoxicity. The functionalization of SiO2NP surface has grafted SiO2NPs can provide a chance for enhancing the
been demonstrated using a covalent bonding between interaction with negative charge surface of cell membrane.
PEGlyated-PEI copolymer and epoxy group. This inexpen- The advantages of using cationic PEI polymer for gene
sive, simple fabrication method can be utilized for multi- delivery stay on the simple experimental condition which
functionality, which may conjugate with a fluorescence dye, PEI can be combined with siRNA or DNA using charge–
544 H. Lee et al.

charge interaction [42–45]. There were many mechanisms Optimal nanoparticle size (∼45 nm), highly positive-charged
explained on the release of siRNA from delivery materials. (+66 mV) surface, and RNAs protection ability directed the
In the present study, it is probably released from the fabricated nano-hybrid for enhanced cellular uptake and
nanocomposite based on the phenomena of ligand exchange effective siRNA delivery in A549 cancer cell, as studied
effect/charge effect [45]. under in vitro condition. In the complexation between
The high molecular weight and charged polymer PEI25k-PEG5k-graft-SiO2NPs and siRNAs, it was shown
normally have a high cytotoxicity. The researches of that N/P ratio could verify the complexation efficiency of
reducing the cytotoxicity of PEI polymer have been studied PEI25k-PEG5k-graft-SiO2NPs. The complete complexation
using anhydride, aldehyde moieties to neutralize primary was reached at N/P ratio (1:10). Moreover, N/P ratio 1:10
amine groups, and other conjugation of low molecular complex did protect RNase well than other N/P ratio
weight polymers. For example, partial acetylation of PEI complexes. In conclusion, the optimized PEI25k-PEG5k-
amino groups was shown to enhance gene transfer activity graft-SiO2NPs can have the ability to modify and enhance
[46]. N-Acetylation of PEI lowered its toxicity [47]. Also, the stability of siRNA. Furthermore, our PEI25k-PEG5k-
conjugation of acetate, butanoate, and hexanoate to graft-SiO2NPs promise to apply as an effective siRNA
branched PEI at low degree of substitution (below 25%) delivery system with low cytotoxicity compared with
resulted in moderate improvement of transfection activity PEI25k cationic polymer. The additional optimization is also
as demonstrated by Putnam et al. [48]. Petersen et al. promising to enhance the biocompatibility of modified-
reported that the larger molecular weight of PEG polymers PEI25k-PEG5k-graft-SiO2NPs for in vivo applications. A
(>5 kDa) can influence the surface charge, cytotoxicity, and detailed study is in progress to unravel the underlying
aggregation of PEI polymers [49]. Similar to other study, mechanism of dual function of this hybrid nanoparticle for
we have used simple PEGylation onto PEI polymer using both MR imaging detection of single cell and siRNA
NHS-PEG 5 kDa, and then PEGylated PEI copolymer has transfection using synthesized T1 weight contrast agent.
been grafted on the SiO2NPs using epoxy silane compound.
The two combinations significantly reduced the cytotox- Acknowledgements This study was supported by a grant of the
Ministry of Health and Welfare (A040041) and Samsung Biomedical
icity of PEI25k polymer, which is observed from the XTT
Research Institute, Republic of Korea (PB00021). We thank Ms.
cytotoxicity assay comparing to only PEI polymer. For Yunhee Kim for solid NMR spectroscopic analysis in NICEM, SNU,
instance, it is reported that the hypothetical “proton sponge” and Ms. Youngshin Yoo for HR-TEM analysis in SKKU.
mechanism of PEI on modification with PEG synergisti-
cally improves the biodistribution and pharmacokinetics of
their complexes [50]. Neu et al. demonstrated that usage of References
endosome-disrupting polymers such as PEG grafted and
hyperbranched polyethyleneimine could allow moving 1. Ulbrich W, Lamprecht A (2010) J R Soc Interface 7:55–66
across cell membranes via endocytosis and disrupting 2. Green JJ, Zhou BY, Mitalipova MM, Beard C, Langer R, Jaenisch
intracellular organelles using a proton-sponge effect [51]. R (2008) Nano Lett 8:3126–3130
3. Allouche J, Boissiere M, Helary C, Livage J, Coradin T (2006) J
Additionally, this endosomolytic effect is believed to arise
Mater Chem 16:3120–3125
from the large number of amine groups on each PEI 4. Yu YY, Chen CY, Chen WC (2003) Polymer 44:593–601
polymer, leading to proton absorption in acid organelles 5. Borrego T, Andrade M, Pinto ML, Silva AR, Carvalho AP, Rocha
and an osmotic pressure buildup across the organelle J, Cristina F, Joao P (2010) J Colloid Interf Sci 344:603–610
6. Kleemann E, Neu M, Jekel N, Fink L, Schmehl T, Gessler T,
membrane. This osmotic pressure causes a disruption of
Seeger W, Kissel T (2005) J Control Release 109:299–316
the acidic endosomes and a release of the trapped materials 7. Burke RS, Pun SH (2008) Bioconjugate Chem 19:693–704
into the cytoplasm [52]. Finally, the PEI-PEG-graft- 8. Ashtari P, He X, Wang K, Gong P (2005) Talanta 67:548–554
SiO2NPs could allow the high cellular transfection with a 9. Jere D, Jiang HL, Arote R, Kim YK, Choi YJ, Cho MH, Akaike
T, Cho CS (2009) Expert Opin Drug Deliv 6:827–834
low cytotoxicity, safe protection, and delivery of siRNA
10. Fuller JE, Zugates GT, Ferreira LS, Ow HS, Nguyen NN, Wiesner
using a proton-sponge effect, steric interaction, electrostatic UB, Robert SL (2008) Biomaterials 29:1526–1532
interaction, and polymer bridging interactions. 11. Choi SJ, Oh JM, Choy JH (2009) J Inorg Biochem 103:463–471
12. Wang H, Yang R, Yang L, Tan W (2009) ACS Nano 3:2451–2460
13. Elbakry A, Zaky A, Liebl R, Rachel R, Goepferich A, Breunig M
(2009) Nano Lett 9:2059–2064
Conclusion 14. Bartlett DW, Su H, Hildebrandt IJ, Weber WA, Davis ME (2007)
Proc Natl Acad Sci USA 104:15549–15554
From the current study, we have examined the simple and 15. Merkel OM, Livrizzi D, Pfestroff A, Schurrat T, Behe M, Kissel T
(2009) Bioconjugate Chem 20:174–182
facile approach to hybridize the inorganic SiO2NPs and
16. Christensen LV, Chang CW, Yockman JW, Conners R, Jackson H,
organic copolymer PEI25k-PEG5k. As obtained, nano- Zhong Z, Jan F, David AB, Sung WK (2007) J Control Release
hybrid was successfully evaluated for siRNA delivery. 118:254–261
PEGylated polyethyleneimine grafted silica nanoparticles 545

17. Meade BR, Dowdy SF (2008) Adv Drug Deliv Rev 60:530–536 37. Petersen H, Petra MF, Dagmar F, Thomas K (2002) Macro-
18. Yoon TJ, Kim JS, Kim BG, Yu KN, Cho MH, Lee JK (2005) molecules 35:6867–6874
Angew Chem Int Ed 44:1068–1071 38. Nguyen HK, Lemieux P, Vinogradov SV, Gebhart CL, Guérin L,
19. Bartlett DW, Davis ME (2008) Biotechnol Bioeng 99:975–985 Paradis G, Bronich TK, Alakhov VY, Kabanov AV (2000) Gene
20. Akhtar S, Benter IF (2007) J Clin Invest 117:3623–3632 Ther 7:126–138
21. Davis ME (2009) Mol Pharmaceut 6:659–668 39. Thomas M, Lu JJ, Ge Q, Zhang C, Chen J, Klibanov AM (2005)
22. Lipski AM, Pino CJ, Haselton FR, Chen IW, Shastri VP (2008) Proc Natl Acad Sci U S A 102:5679–5684
Biomaterials 29:3836–3846 40. Urban-Klein B, Werth S, Abuharbeid S, Czubayko F, Aigner A
23. Xu Y, Li Q (2007) Clin Chem 53:1503–1510 (2005) Gene Ther 12:461–466
24. Feng L, Wang Y, Wang N, Ma Y (2009) Polym Bull 63:313–327 41. Liong M, Lu J, Kovochich M, Xia T, Ruehm SG, Nel AE,
25. Joubert M, Delaite C, Bourgeat-Lami B, Dumas P (2005) Tamanoi F, Zink JI (2008) ACS Nano 2:889–896
Macromol Rapid Commun 26:602–607 42. Brus C, Petersen H, Aigner A, Czubayko F, Kissel T (2004)
26. Estévez MC, O’donoghue MB, Chen X, Tan W (2009) Nano Res Bioconjugate Chem 15:677–684
2:448–461 43. Merdan T, Callahan J, Petersen H, Kunath K, Bakowsky U,
27. Graf C, Vossen DLJ, Imhof A, Blaaderen AV (2003) Langmuir Kopecková P, Thomas K, Jindřich K (2003) Bioconjugate Chem
19:6693–6700 14:989–996
28. An Y, Chen M, Xue Q, Liu W (2007) J Colloid Interf Sci 44. Brus C, Petersen H, Aigner A, Czubayko F, Kissel T (2004) Eur J
311:507–513 Pharm Biopharm 57:427–430
29. Park KM, Kang HC, Cho JK, Chung IJ, Cho SH, Bae YH, Kun N 45. Glodde M, Sirsi SR, Lutz GJ (2006) Biomacromolecules 7:347–
(2009) Biomaterials 30:2642–2652 356
30. Salon MC, Gerbaud G, Abdelmouleh M, Bruzzese C, Boufi S, 46. Forrest ML, Meister GE, Koerber JT, Pack DW (2004) Pharm Res
Belgacem MN (2007) Magn Reson Chem 45:473–483 21:365–371
31. Brannon-Peppas L, Blanchette JO (2004) Adv Drug Deliv Rev 47. Thomas M, Klibanov AM (2002) Proc Natl Acad Sci U S A
56:1649–1659 99:14640–14645
32. Kobayashi S, Nakase I, Kawabata N, Yu HH, Pujals S, Imanishi 48. Doody AM, Korley JN, Dang KP, Zawaneh PN, Putnam D (2006)
M, Ernest G, Shiroh F (2009) Bioconjugate Chem 20:953–959 J Control Release 116:227–237
33. Lessard-Viger M, Rioux M, Rainville L, Boudreau D (2009) Nano 49. Petersen H, Fechner PM, Martin AL, Kunath K, Stolnik S,
Lett 9(8):3066–3071 Roberts CJ, Dagmar F, Martyn CD, Thomas K (2002) Bioconjugate
34. Mijatovic WH, Binder HG (2000) Mikrochim Acta 133:175–181 Chem 13:845–854
35. Takayama K, Tadokoro A, Pujals S, Nakase I, Giralt E, Futaki S 50. Kunath K, von Harpe A, Petersen H, Fischer D, Voigt K, Kissel T,
(2009) Bioconjugate Chem 20:249–257 Ulrich B (2002) Pharm Res 19:810–817
36. Mao S, Neu M, Germershaus O, Merkel O, Sitterberg J, Bakowsky 51. Neu M, Fischer D, Kissel T (2005) J Gene Med 7:992–1009
U, Thomas K (2006) Bioconjugate Chem 17:1209–1218 52. Duan H, Nie S (2007) J Am Chem Soc 129:3333–3338

You might also like