You are on page 1of 11

Nanoscale

View Article Online


PAPER View Journal
Published on 05 June 2017. Downloaded by University of California - San Diego on 29/06/2017 06:44:46.

The effect of PEGylated hollow gold nanoparticles


Cite this: DOI: 10.1039/c7nr01853c
on stem cell migration: potential application in
tissue regeneration†
Maria del Mar Encabo-Berzosa,a,b Maria Sancho-Albero,a,b Alejandra Crespo,c
Vanesa Andreu,a Victor Sebastian, a,b Silvia Irusta, a,b Manuel Arruebo, *a,b
Pilar Martín-Duque*c and Jesus Santamariaa,b

Mesenchymal stem cells (MSCs) not only can be differentiated into different cell types but also have
tropism towards injured or inflamed tissues serving as repair cells. Here we have demonstrated that MSCs
containing gold nanoparticles (GNPs) whose surface has been functionalized with PEG show accelerated
cell migration, successful scaffold colonization and regeneration. We report the impact of GNPs on the
migration (by the wound healing assay), and proliferation (by flow cytometry analysis and by the detection
of metabolic mitochondrial activity) on the behaviour of different cell lines including MSCs, HeLa cells,
and human dermal fibroblasts. We conclude that GNPs are easily internalized by MSCs causing an
increase in their migration rate, mediated by actin and tubulin with a 4-fold increased expression level of
Received 15th March 2017, those proteins. We also demonstrate that MSCs containing GNPs are able to successfully colonize fibrin
Accepted 31st May 2017
and PCL-based scaffolds and that an enhanced osteoblastic differentiation is reached when using the
DOI: 10.1039/c7nr01853c nanoparticle-laden cells compared to untreated cells used as a control. These results highlight the poten-
rsc.li/nanoscale tial use of MSCs as therapeutic nanoparticle-carriers in regenerative medicine.

Introduction tissues4–6 (i.e., wounds, tumor sites and inflamed tissues) to


help in their repair. Promising results using these cells have
Every year, trauma together with bone, joint and cartilage- already been reported in wound healing, for example, in
associated diseases usually involve structural damage, result- repairing corneal damage7 or spinal cord injury.8 Skeletal dis-
ing in a severe pain and disability for millions of people world- orders present many opportunities for the use of MSCs, and
wide.1 In regenerative medicine, cellular, tissue and organ- numerous clinical trials have shown encouraging results.9
based approaches are developed to restore biological functions These cells present high potential in regenerative medicine
that have been lost.2,3 Therefore, tissue repair and regenerative where novel therapeutic strategies could be developed, based
medicine have attracted the interest of the scientific commu- on different chemical and biological cues.
nity, providing promising results in preclinical models and Nanotechnological tools have often been leveraged for
clinical pilot studies. tissue regeneration because some nanoscale properties make
Different cells such as mesenchymal stem cells (MSCs) are cell interactions and other cellular functions (cell adhesion,
crucial players in the processes of tissue regeneration. MSCs proliferation and differentiation) far more efficient than tra-
are progenitor cells that can be differentiated into many cell ditional macroscopic materials.10 The capacity of some nano-
types and also have the ability to migrate towards damaged materials to speed up the repair of several tissues (such as
bone, cartilage, bladder, nerves and vessels) by improving
a
their intrinsic biological functions has been reported.11 The
Department of Chemical Engineering, Aragon Institute of Nanoscience (INA),
University of Zaragoza, Campus Río Ebro-Edificio I+D, C/Poeta Mariano Esquillor S/N,
quantification of the migration ability and proliferative
50018-Zaragoza, Spain. E-mail: arruebom@unizar.es capacity of cells on different substrates provide valuable
b
Networking Research Center on Bioengineering, Biomaterials and Nanomedicine, insights for designing new biomaterials.12 Numerous in vitro
CIBER-BBN, 28029-Madrid, Spain and in vivo reports have studied the effect of nanoparticles
c
Universidad Francisco de Vitoria, Facultad de Ciencias Biosanitarias,
(characteristics such as sizes,13 shapes14 and surface coat-
Carretera Pozuelo a Majadahonda, Km 1.800, 28223 Pozuelo de Alarcón, Madrid,
Spain. E-mail: pmartin@ufv.es
ings15) on cell migration, proliferation, differentiation and
† Electronic supplementary information (ESI) available. See DOI: 10.1039/ wound healing. For example, internalization of SiO2 nano-
c7nr01853c particles (NPs) affects essential physiological functions in the

This journal is © The Royal Society of Chemistry 2017 Nanoscale


View Article Online

Paper Nanoscale

cell including adhesion and migration.16 In the same way, it cantly affected by the presence of the nanoparticles at the
has been demonstrated that the human endothelial exposition same doses. Our results provide some insights into the influ-
to carbon nanotubes induces an increase in endothelial mono- ence of GNPs on cellular behaviour (migration, proliferation,
layer permeability and cell migration.17 Also the effects of differentiation and ability to colonize scaffolds) and show the
TiO2, SiO2 and hydroxyapatite NPs on epithelial cell migration potential of these materials for strategic regenerative medicine
have been evaluated, resulting in an increased cell contractility and tissue repair. Incubation of MSCs with GNPs appears as a
with significantly impaired wound healing capability.18 useful tool to modulate their migration capacity to achieve a
Published on 05 June 2017. Downloaded by University of California - San Diego on 29/06/2017 06:44:46.

Finally, the use of PLGA microparticles to promote bone regen- more rapid and effective regenerative therapy.
eration has also been analyzed. In this sense, the use of PLGA
micro- and nanoparticles could represent an advance in the
design of scaffolds, since it allows the encapsulation of growth Results and discussion
factors such as BMP2 inducing bone regeneration. In addition
PLGA is biocompatible and biodegradable, which is advan- Nanoparticle characterization
tageous over other materials.19 Fig. 1 shows the TEM images of the as-prepared nanoparticles.
Gold nanoparticles (GNPs) have attracted the interest of The morphology of the obtained nanoparticles was pseudo-
recent research because of their biomedical potential as drug spherical with a thick shell visualized in the TEM images as a
carriers, gene vectors and therapeutic agents in a wide range dark ring. The functionalization with PEG resulted in an exter-
of promising applications ranging from cancer and neuro- nal polymeric shell that was visualized in a negative-stained
psychiatric disorders to tissue repair and regeneration.20 Their TEM image as a halo of almost 4 nm surrounding the hollow
advantages include the ease of synthesis in a varied range of gold nanoparticles (HGNs) (Fig. 1, top, far right). After pegyla-
monodisperse sizes, the fact that they are essentially inert and tion, no changes in the morphology or in the stability of the
non-toxic at the doses used and their ability to be readily func- nanoparticles were observed. The mean size of HGNs was 38 ±
tionalized with targeting ligands and drugs. Recent studies 5.4 nm, while that of PEG-HGNs was 41.7 ± 4.4 nm. We
indicate that these nanoscale materials have a direct influence included PEG on the surface of the nanoparticles in order to
on the behaviour of MSCs from a cellular to a molecular level. provide them with steric hindrance against agglomeration, to
Thus, GNPs functionalized with different coatings induce decrease their cytotoxicity and to reduce protein adsorption in
differential MSC responses and, when optimized, could a potential future in vivo application. A maximum absorbance
promote MSC proliferation, contributing to the development peak was observed in the NIR region, precisely at 808 nm,
of new strategies for tissue regeneration therapies.21 which corresponds to the geometrical shape and size observed
Here we report the potential impact of poly(ethylene in the TEM images according to the literature.22 At pH = 7 the
glycol)-ether thiol functionalized GNPs on the migration and surface charge of the HGNs and the PEG-HGNs, measured by
proliferation behaviour of different cell lines: MSCs, HeLa cells DLS, was −12.63 ± 0.66 and −7.51 ± 1.53 mV, respectively. TGA
and human dermal fibroblasts. GNPs are easily internalized by analysis (Fig. 1c) showed that the quantity of PEG on the func-
MSCs causing an increase in their migration rate, mediated by tionalized nanoparticles was around 30% of the total nano-
actin and tubulin expression. On the other hand, the particle mass. XPS analysis (Fig. 2) revealed that in addition to
migration capacity of tumoral and epithelial cells is not signifi- gold, oxygen and carbon were also present on the surface of

Fig. 1 Characterization of HGNs and PEG-HGNs. (a) TEM images of HGNs and PEG-HGNs. PEG functionalization is clearly visible around the nano-
spheres. (b) UV-Vis absorption spectra for both types of nanoparticles. (c) TGA results of HGNs (left) and PEG-HGNs (right).

Nanoscale This journal is © The Royal Society of Chemistry 2017


View Article Online

Nanoscale Paper

viability at the studied doses (12, 5, 25, 50 and 100 μg mL−1) in


any of the cell types of interest (MSCs, HeLa and fibroblasts).
In contrast, a clear cell viability decrease was observed for the
non-pegylated HGNs with a marked cytotoxicity at doses above
100 µg mL−1 after 24 h of contact for the three cellular types
(Fig. 3). The dose of 50 µg mL−1 PEG-HGNs was chosen for the
subsequent experiments as the limit of the sub-cytotoxic dose.
Published on 05 June 2017. Downloaded by University of California - San Diego on 29/06/2017 06:44:46.

These results are in agreement with previous studies demon-


strating that the nanoparticle cytotoxicity was reduced after
pegylation.24
Nanoparticle uptake is influenced by factors such as the
physical properties (e.g. size and surface charge) of the nano-
material or variations in cell-membrane characteristics.25,26
Confocal microscopy experiments showed large amounts of

Fig. 2 (a) Elemental composition of HGNs and PEG-HGNs. (b) Binding


energies and peak assignment for S 2p3/2 and Au 4f7/2 core level
PEG-HGNs; (c) Au 4f core level spectrum of PEG-HGNs. The blue line
indicates the binding energies of the elemental gold and the red line
indicates the binding energies of the gold bound to sulfur.

HGNs, which was mainly attributed to the presence of PVP on


the surfaces in agreement with our TGA results (Fig. 1c) where
approx. 20% of the nanoparticle weight corresponds to organic
matter. The PEG used for the functionalization contains
sulphur due to the presence of the thiol group in the molecule.
This sulphur was also observed in PEG-HGNs, and can be used
to trace the presence of PEG on the particles. Also, the chemi-
cal environment of gold and sulphur atoms can be studied to
assess the chemical bonding between PEG and the gold NPs.
A peak displacement to higher binding energies was observed in
the chemical environment of gold, possibly due to the union
between gold and sulphur. This suggests that the interaction
between PEG and HGNs occurs, thanks to a gold–sulphur
bond. We also observed that almost 90% of the gold was
linked to the sulphur. This is consistent with the previously
published results.23

Nanoparticle cytotoxicity and uptake


According to the ISO 10993-5 (Biological evaluation of medical
devices. Part 5: Tests for in vitro cytotoxicity) a reduction in cell
viability higher than 30% compared to the control is con-
sidered as a cytotoxic effect and consequently we set the con-
Fig. 3 Cytotoxicity analysis. AlamarBlue® results for (a) MSCs; (b) HeLa
centration of 100 μg mL−1 as a subcytotoxic dose for our cells; and (c) human dermal fibroblast after incubation with HGNs or
PEGylated HGNPs. In this way, the AlamarBlue® analysis PEG-HGNs for different times (n = 3). *Significant differences with
showed that PEG-HGNPs did not significantly decrease cell untreated cells ( p < 0.05).

This journal is © The Royal Society of Chemistry 2017 Nanoscale


View Article Online

Paper Nanoscale
Published on 05 June 2017. Downloaded by University of California - San Diego on 29/06/2017 06:44:46.

Fig. 4 HGN and PEG-HGN internalization evaluation by confocal microscopy. In order to visualize the cytoplasm of the cells, the actin was labeled
with phalloidin-488, whereas the nuclei were stained with Draq5. PEG-HGNs were directly visualized by reflection. In all cases, a Z-stack was per-
formed to confirm the internalization of the particles. PEG-HGN agglomerates were only observed inside MSCs. The yellow arrows indicate the
location of nanoparticles within the cells.

PEG-HGNs clearly internalized in MSCs forming aggregates readily internalized by MSCs, but not by the other cell types.
inside vesicles (shown as red-colored agglomerates in Fig. 4 In addition, a possible reduction in cell-plate adhesion caused
and in ESI1†), probably following the endosomal route. by the NPs was ruled out as a possible cause of faster
Moreover, these NPs were only distributed in the cytoplasm migration, since the effect was only observed for MSCs and not
rather than in the nuclei of MSCs because of their size.27,28 for other cell types. Also, the presence of non-internalized
Unlike MSCs, a significantly lower accumulation of PEG-HGNs nanoparticles at the interphase between the culture plate and
was observed inside fibroblast or HeLa cells. It is important to the cells is unlikely to be responsible for an enhanced
point out that cells were washed before observation, hence any migration rate since cells were washed to remove non-interna-
non-internalized nanoparticles would be washed away and lized nanoparticles before performing the wound healing
only the internalized ones could be observed. assay. To corroborate this, the level of an adhesion protein
(vinculin) was studied, without any observed effect on its
Nanoparticle influence on cell properties expression. The level of proteins involved in cell adhesion and
Migration evaluation. A variety of physiological and patho- migration such as actin, tubulin and vinculin30 was studied in
logical processes such as embryonic development, blood vessel cells incubated with PEG-HGNs and control cells. The
formation and remodeling, tissue regeneration, immune sur- immunoassay revealed that MSCs incubated with PEG-HGNs
veillance and inflammation are strongly dependent on the cell showed an increased rate of actin and tubulin expression with
migration mechanism.29 In order to study the capacity of respect to control cells, this expression being almost 4 times
PEG-HGNs as modifiers of cellular behavior for either stem- higher (Fig. 6). On the other hand, as we mentioned before,
cell based strategies or for regenerative medicine, some com- the vinculin protein level was not affected. Although in this
parative migration experiments were performed following the case no change in the vinculin levels was observed in the pres-
wound healing assay after incubating three cell types tested ence of PEG-HGNs, a post-translational regulation cannot be
with PEG-HGNs for 24 h. The assay revealed that PEG-HGNs ruled out. In fact, several reports have remarked that the phos-
only significantly influenced MSC migration but not the other phorylation of some tyrosines plays a relevant role in the func-
cell types studied. For MSCs, they caused a clear increase in tion of this protein.30,31 Here, we propose that the increment
the migration rate compared to untreated cells ( p < 0.05) observed in the closure speed was probably due to the
(Fig. 5). The untreated cells reached 42% of wound closure enhancement in the actin and tubulin expression. It is impor-
24 h after the scratch, whereas cells treated with NPs closed tant to remark that the expression level of the two structural
almost 80% of the bare surface. However, migration in HeLa proteins studied (actin and tubulin) in the different cellular
cells and fibroblasts was not clearly affected and their types treated with PEG-HGNs was analyzed by quantifying
migration capacity was significantly smaller than the their fluorescence level using confocal microscopy and appro-
migration rate of MSCs at 24 h (Fig. 5). These results can be priate software to analyze fluorescence. In order to compare
explained at least in part by the fact that PEG-HGNs were the protein expression levels, all the samples were observed

Nanoscale This journal is © The Royal Society of Chemistry 2017


View Article Online

Nanoscale Paper
Published on 05 June 2017. Downloaded by University of California - San Diego on 29/06/2017 06:44:46.

Fig. 5 Migration evaluation. (a) Wound healing assay in MSCs, HeLa cells and fibroblasts incubated without (control) or with PEG-HGNs
(50 μg mL−1) evaluated by inverted optical microscopy (scale bar = 500 µm). (b) Quantitative analysis of relative cell migration for 0, 4 and 24 h. The
differences between the closed surfaces at different intervals of time are shown. Significant differences ( p < 0.05) between the percentage of closed
surface among the control and MSCs incubated with PEG-HGNs were observed.

Fig. 6 Actin and tubulin expression evaluation. (a) Immunoassay study of proteins by confocal microscopy. Actin was labeled with phalloidine-488
and tubulin and vinculin were marked with specific antibodies (scale bar = 47.7 µm). (b) Relative fluorescence intensity analysis of actin, tubulin and
vinculin in the three cellular types. Significant differences between the control and the treated cells were observed for MSCs ( p < 0.05).

under the same optical conditions as the control. In contrast, lished results today regarding the effects of nanoparticles on
both fibroblast and HeLa cells treated with PEG-HGNs showed scenarios relevant to regenerative medicine show limitations
a similar tubulin and actin expression compared to the or inhibition of cell migration and proliferation,16,32,33 the
control. Therefore, only those cellular types capable of signifi- results of this work evidence the potential of HGNs as per-
cantly internalizing PEG-HGNs increased their migration rate formance enhancers in stem-cell based strategies.
via an increment of actin and tubulin expression. These Proliferation evaluation by cell cycle analysis. No significant
results are highly promising because cells involved in regener- differences were found in the cell cycle phase percentages for
ation such as MSCs, were able to internalize the NPs studied, MSCs or HeLa cells when treated with PEG-HGNs compared to
while the tumoral cells studied here (HeLa) did not show any the control (Fig. 7). Consequently, we can conclude that the
increase in their migration capacity. While most of the pub- arrest of the cell cycle or the damage in the DNA was not

This journal is © The Royal Society of Chemistry 2017 Nanoscale


View Article Online

Paper Nanoscale
Published on 05 June 2017. Downloaded by University of California - San Diego on 29/06/2017 06:44:46.

Fig. 7 Cell-cycle analysis. The three cellular types were incubated with
Fig. 8 qPCR results for osteogenic differentiated MSCs. The graph
PEG-HGNs at a concentration of 50 µg mL−1 for 24 h.
shows the gene expression of osteogenic marker genes.

induced by PEG-HGNs at the studied doses in these cellular


types. Only in human dermal fibroblast, the G1 phase was Scaffold colonization. For an initial assessment of the poten-
markedly diminished while the S phase was increased. The tial application of the MSC/PEG-HGN combination in regen-
arrest of the cell cycle is greatly dependent on the cellular and erative medicine, the ability of these cells to colonize different
nanomaterial type. Uz et al.34 have recently described that types of scaffolds was studied. As can be seen in Fig. 9, both
different GNPs with varying degrees of PEG grafting density control MSCs and those that had been cultured in the pres-
may affect the cell cycle in different ways. Also it was reported ence of PEG-HGNs were able to colonize PCL scaffolds efficien-
that titanium nanoparticles might affect the MSC cycle in a tly 24 h after seeding. Qualitatively, it was observed that cells
negative way by increasing the G1 phase and decreasing the G2 pre-incubated with PEG-HGNs colonized the scaffold more
and S phases.33 homogeneously than control cells, as indicated by the greater
MSC differentiation studies. MSC differentiation is a key number and better distribution of nuclei visible in similar
step in various tissue regeneration processes, such as bone for- areas of the scaffold.
mation. However, the reports on the influence of GNPs on the Colonization studies of fibrin scaffolds were also carried
differentiation process are scarce and their mechanism of out. In this case the ability of cells incubated (or not) with NPs
action is still under debate. Some studies have shown that to colonize fibrin scaffolds with or without NPs (Fig. 9) was
GNPs enhance osteogenic differentiation and decrease adipo- analyzed. In all cases an efficient colonization of the different
genic differentiation.21,35 gels was qualitatively observed by confocal microscopy.
Li et al.36 proposed that different gold nanospheres might Moreover, the cellular proliferation inside the scaffolds was
provide mechanical signals that increase YAP activity and con- confirmed by the Alamar Blue® assay (Fig. SI3†). The improve-
sequently induce osteogenic gene expression, such as Runx2. ment in cell viability and proliferation mediated by the
The same research showed that larger spheres, stars or rods addition of GNPs to scaffolds has been recently reported.40
did not affect cell differentiation and 40 nm nanorods even Those authors demonstrated that the attachment of GNPs and
reduced YAP activity. Zhang et al.37 described the stimulation hydroxyapatite NPs to scaffolds improved the proliferation and
of the ERK/MAPK signaling pathway in primary osteoblasts viability of fibroblasts. They proposed that the observed prolifer-
mediated by GNPs. Also Ko et al.13 reported the positive effect ation increase might be due to the high surface energy of NPs
of GNPs of different sizes on osteogenic differentiation, as well that could affect protein adsorption. Protein adsorption influ-
as Yi et al.38 who concluded that the mechanism involved in ences cell response to nanoparticles and may enhance cell pro-
the differentiation promoted by 20 nm GNPs was the modifi- liferation.40 On the other hand, Baranes et al.41 observed the
cation of the p38 MAPK pathway. beneficial effect of GNPs on the differentiation, proliferation
In our study, the real time PCR results showed that Runx2 and maturation of cultured neurons on 3D scaffolds. In this
expression was increased in MSCs incubated with PEG-HGNs case, the addition of GNPs provided additional anchoring sites
and differentiated to osteoblasts, compared to the negative that allowed for greater morphogenesis. According to that study,
control (untreated cells), to a greater extent compared to differ- the role of GNPs needs to be more investigated, in order to
entiated MSCs without NPs (Fig. 8). Since it has been determine whether the NPs promote axon elongation and
described37,39 that Runx2 is regulated by the MAPK pathway, higher expression of neuronal markers due to topographical
we propose that PEG-HGNs might interact with this protein cues or due to their promoted conductivity.
cascade and therefore acting by a mechanical stimulus. Here, we demonstrate that MSCs containing PEG-HGNs are
Specific staining of adipogenic and osteogenic differentiation able to successfully colonize fibrin and PCL-based scaffolds
is shown in Fig. SI2.† may be the NPs responsible for a faster migration rate

Nanoscale This journal is © The Royal Society of Chemistry 2017


View Article Online

Nanoscale Paper
Published on 05 June 2017. Downloaded by University of California - San Diego on 29/06/2017 06:44:46.

Fig. 9 Scaffold colonization. (a) PCL membrane colonization by control cells or cells with internalized PEG-HGNs 24 h after seeding the cells. Scale
bar: 47.6 µm. (b) Fibrin scaffold colonization by MSCs incubated with or without PEG-HGNs. In the first row the photos show control cells seeded
onto control scaffolds. The second row represents control scaffold colonization by cells incubated with PEG-HGNs. The third row shows the coloni-
zation of a fibrin gel containing PEG-HGNs by control cells. In the last row both the cells and the gel contained PEG-HGNs. Scale bar: 254 µm.

compared to the control cells as seen in the wound healing Conclusions


assay. A positive scaffold colonization and infiltration when
using nanoparticle-containing cells would prompt an earlier HGNs have been used in different biomedical settings as heat
integration and a rapid remodeling and healing. In addition, transducers in tumor photothermal treatments. Nevertheless,
an enhanced osteoblastic differentiation also highlights the their use as promoters in regenerative medicine has not been
potential use of nanoparticle-laden cells in regenerative studied in depth. The results here presented are very promis-
medicine. ing because they show that PEG-HGNs are able to accelerate

This journal is © The Royal Society of Chemistry 2017 Nanoscale


View Article Online

Paper Nanoscale

MSC migration whereas this effect is not induced in tumoral software (NIH-RSB). Using UV-VIS spectroscopy (Jasco V670)
cells. Also the enhancement in osteogenic differentiation the characteristic plasmon at a particular wavelength of HGNs
could open up new horizons for the use of PEG-HGNs in the and PEG-HGNs was evaluated, and correlated with their size
design of biomaterials as scaffolds for bone regeneration. and shape.
Although more studies are still necessary, our data provide evi- Thermogravimetric analysis (TGA; Mettler Toledo TGA/
dence for the potential of combining MSCs and HGNs in the STDA 851e) was carried out in a temperature range between 30
development of new materials with their possible application and 850 °C with a heating rate of 20 °C min−1 to assess the
Published on 05 June 2017. Downloaded by University of California - San Diego on 29/06/2017 06:44:46.

in regenerative medicine. PEG/Au weight ratio in the NPs. In order to analyze the stabi-
lity and the surface charge of the particles, a zeta potential
assay at pH = 7 was also performed (Brookhaven 90 plus and
Methods ZetaPALS software). The NPs were resuspended and measured
in a 1 mM KCl solution at pH = 7. Finally, X-ray photoelectron
Synthesis of hollow gold nanoparticles
spectroscopy (XPS) was used to determine the elemental com-
All the materials employed in the synthesis of HGNs were pro- position and the surface grafting of PEG-HGNs. It was per-
vided by Sigma Aldrich and used as received: cobalt(II) chloride formed with an Axis Supra (Kratos Tech.). The spectra were col-
hexahydrate, sodium citrate tribasic dihydrate, poly(vinylpyrrol- lected by using a monochromatized Al Kα source (1486.6 eV)
idone) (PVP) Mw = 55 000 Da, gold(III) chloride hydrate (50% run at 15 kV and 15 mA. For the individual peak regions, a
purity), sodium borohydride, and poly (ethylene glycol)-ether pass energy of 20 eV was used. Analyses of the peaks were per-
thiol (PEG 1000 Da MW). formed with CasaXPS software, using a weighted sum of
The synthesis of HGNs was carried out following the pro- Lorentzian and Gaussian component curves after background
cedure used in previous work.42 Briefly, in a two-necked round- subtraction.
bottom flask 400 mL of distilled water, 400 μL of 0.4 M cobalt
chloride hexahydrate (CoCl2·6H2O) and 1.6 mL of 0.1 M Cell culture conditions
sodium citrate trihydrate (Na3C6H5O7·3H2O) were mixed Murine MSCs, human dermal fibroblasts and HeLa cells were
without magnetic stirring under an inert Ar atmosphere to obtained from Lonza and Cancer Research-UK cell services.
avoid a premature Co oxidation. After 40 minutes, 2 mL of a MSCs were cultured in Dulbecco’s modified Eagle’s
1 wt% solution of poly(vinylpyrrolidone) (PVP) with an average F-12 medium (DMEM F-12, GIBCO) with 10% fetal bovine
Mw of 55 000 Da and 400 µL of 1.0 M sodium borohydride serum (FBS, GIBCO), 1% penicillin/streptomycin and 1%
(NaBH4) were added. The color change from pale pink to amphotericin and maintained at 37 °C under a 5% CO2-
brown was indicative of the cobalt nanoparticle formation. humidified atmosphere under hypoxic conditions (3% O2). For
Subsequently, 120 mL of distilled water and 180 µL of 0.1 M culturing fibroblasts and HeLa cells, Dulbecco’s modified
chloroauric acid trihydrate (HAuCl4·3H2O) were mixed with Eagle’s medium (DMEM, GIBCO) with 10% fetal bovine serum
360 mL of the previous cobalt-based dispersion used as a sacri- (FBS, GIBCO), 1% penicillin/streptomycin and 1% amphoteri-
ficial template to promote the formation of CoCl2 and CoO, cin was used under normoxic conditions.
and the galvanic reduction of Au3+ yielding hollow Au-based
shells. Nanoparticle cellular uptake
The resulting HGNs were functionalized with PEG (a steric Confocal microscopy characterization (Spectral Confocal
polymer) to obtain PEG-HGNs using an excess of monofunc- Microscope Leica TCA SP2) was carried out to evaluate the cel-
tional poly(ethylene glycol)-ether thiol (PEG 1000 Da), taking lular uptake and trafficking of PEG-HGNs in the different cell
advantage of the strong chemical bond between Au and S. types. Cells were seeded at a density of 2 × 104 cells onto
A solution of HGNs was brought into contact with a dilution of 20 mm cover slips (in a 24-well plate) and allowed to grow for
PEG during one hour under magnetic stirring. Any excess of 24 h. The PEG-HGNs (50 μg mL−1) were incubated with the
unbound PEG was removed by dialysis. different cellular types for one day. After that, cells were fixed
The concentration of the final dispersion was adjusted by with para-formaldehyde 4% and marked with phalloidin-
centrifugation at 10 000 rpm for 10 minutes. Alexa488 (Invitrogen) to label the cytoplasmic actin and with
Draq-5 to label the nuclei. PEG-HGN-based agglomerates were
Nanoparticle characterization directly observed by reflection of the incident light in the con-
The size, shape and surface chemistry are all important for focal microscope at 488/490 nm excitation/emission wave-
determining the specific properties and characteristics of NPs. lengths. Z-Stack orthogonal projections were carried out in
The morphology and size distribution of HGNs and PEG-HGNs order to analyze the presence of PEG-HGNs inside the cytosol.
were characterized by transmission electron microscopy (T20-
FEI Tecnai thermoionic transmission electron microscope) Wound healing assay
operated at 200 kV with a LaB6 electron source fitted with a To study the role of PEG-HGNs in cell migration wound
“SuperTwin®” objective lens allowing a point-to-point resolu- healing assays were carried out. First, 2 × 105 cells (MSCs,
tion of 2.4 Å. From the TEM images, an average particle dia- fibroblasts and HeLa) were seeded onto 6 multi-well culture
meter was obtained for HGNs and PEG-HGNs with ImageJ plates and 24 h later 50 µg mL−1 of PEG-HGNs was added to

Nanoscale This journal is © The Royal Society of Chemistry 2017


View Article Online

Nanoscale Paper

the cells. The control wells were maintained without nano- priate confluence and a different induction medium was
particles. When a homogeneous monolayer of cells was added. On the one hand, the induction adipogenic medium
achieved, a 1 mm wide straight-line indentation was made (Stem Cells Technologies) was changed every 2–3 days. Cells
with a 1000 µL pipette tip simulating a wound. Cells were were maintained in that medium for 14 days. The final con-
washed twice with PBS and visualized using an inverted centration of PEG-HGNs in the culture was 50 µg mL−1. At the
optical microscope (Olympus IX81) at 0 h, 4 h and 24 h after end of the experiment, cells were recovered for RNA extrac-
scratching. The data were statistically analyzed taking five tion. On the other hand, the osteogenic induction medium
Published on 05 June 2017. Downloaded by University of California - San Diego on 29/06/2017 06:44:46.

independent measurements in three different zones of the (R&D systems supplements) was also changed every 2–3 days
wound. keeping the same final PEG-HGN concentration (50 µg mL−1).
In this case, cells were cultured with the induction medium
Immunoassay and confocal microscopy for 21 days. After that time the total RNA was purified from
Tubulin and actin expression in MSCs, fibroblasts and HeLa the cells.
cells was evaluated by confocal microscopy. First, cells were
seeded onto 20 mm sterile cover slips (in a 24-well plate) at a Real time PCR
density of 2 × 104 cells per well and allowed to grow for 24 h. The total RNA was isolated from mesenchymal stem cells
After that time, PEG-HGNs were added to the cells at a concen- treated or not with PEG-HGNs during the osteogenic or adipo-
tration of 50 µg mL−1 keeping the incubation for 24 h. genic differentiation induction experiments. Reverse tran-
Subsequently, cells were washed three times with PBS and scription was performed using the PrimeScript™ Reverse
fixed with para-formaldehyde 4% for 20 minutes. Transcriptase kit (Takara). The relative mRNA expression of
The immunoassay was performed as follows: cells were per- the osteogenic differentiation marker genes was normalized to
meabilized with 0.1% PBS–BSA saponin. Afterwards, they were the GADPH gene and expressed as a fold change relative to the
incubated for 1 h at room temperature in a wet chamber with undifferentiated control. Calculations of quantification relative
the specific antibodies. For actin labelling, a phaloidin- to gene expression were carried out using the 2ΔΔCt method.43
Alexa488 (Invitrogen) was used. In the case of tubulin, not only The 2ΔΔCt method expresses the proportion obtained from the
a primary antibody against α-tubulin (Invitrogen) was relationship between the Ct values of the sample of interest
employed, but also an immunofluorescence-conjugated sec- and the Ct values of the control sample as expressed in the fol-
ondary antibody (Zymed Laboratories) was needed. In that lowing equation:
case, a pre-blocking with 5% PBS-BSA was performed. Next, Induction, relative quantity (RQ):
the cover-slips were mounted in the holders with mowiol con-
2½ΔCtðsampleÞ  ΔCtðcalibratorÞ
taining Draq5 and visualized by confocal laser scanning
microscopy (Spectral Confocal Microscope Leica TCA SP2). ΔCt ðsampleÞ ¼ Ct ðHÞm  CtðPÞm
Finally, the fluorescence intensity, corresponding to the
protein expression level, was measured using Confocal Uniovi ΔCt ðcalibratorÞ ¼ Ct ðHÞc  CtðPÞc
ImageJ software (NIH-RSB). The polymerase chain reaction (PCR) conditions were: an
Cell cycle evaluation initial step at 95 °C for 30 s, 40 denaturation cycles of 95 °C for
5 s and annealing at 60 °C for 34 s. Table 1 shows the
To study the effect of HGNs on the proliferation rate of the sequence of the employed primers in order to study genetic
different cell types, the distribution of the cell-cycle phases expression. In order to carry out PCR experiments, Taqman
after NP treatment was assessed by flow cytometry. Firstly, probes were used.
cells were seeded onto 6-well plates at a density of 2.5 × 105
cells per well. After 24 h of maintenance, PEG-HGNs (50
μg mL−1) were added into the treated wells. One day later, cells Table 1 Nucleotidic sequence of the employed primers in the qPCR
were trypsinized and washed twice with PBS (1200 rpm, analysis
5 min). Then, cells were collected in PBS and fixed with 70%
ice-cold ethanol (1 × 106 cells per mL) and maintained at 4 °C Gene Primer sequence
in these solutions almost for 24 h. DNA staining was per- Osteopontin (OPN) 5′-CTTTCACTCCAATCGTCCCAC-3′
formed by adding RNase A and propidium iodide to the cell 5′-CAGAAACCTGGAAACCTGGAAACTCCTAGAC-3′
solution. Finally, samples were analyzed in a FACSARRAY BD Runx2 5′-CGTCCACTGTCACTTTAATAGCTC-3′
5′-GTAGCCAGGTTCAACGATCTG-3′
equipment with the MODIFIT 3.0 Verity software. Control Bmp2 5′-TGCAGATGTGAGAAACTCGTC-3′
samples (not treated cells) were also run to know the standard 5′-CGCAGCTTCCATCACGAA-3′
distribution of cell cycles in the cell lines assayed. PPAR-ɤ 5′-CTGGCCTCCCTGATGAATAAAG-3′
5′-AGGCTCCATAAAGTCACCAAAG-3′
CD44 5′-CAGTCACAGACCTACCCAATTC-3′
Mesenchymal stem cell differentiation 5′-GTGTGTTCTATACTCGCCCTTC-3′
The adipogenic and osteogenic differentiation capacity β-Actine 5′-GAGGTATCCTGACCCTGAAGTA-3′
5′-CACACGCAGCTCATTGTAGA-3′
of MSCs was evaluated in the presence or absence of Gadph 5′-GTGGAGTCATACTGGAACATGTAG-3′
PEG-HGNs. Briefly, cells were allowed to grow until the appro- 5′-AATGGTGAAGGTCGGTGTG-3′

This journal is © The Royal Society of Chemistry 2017 Nanoscale


View Article Online

Paper Nanoscale

Scaffold colonization Acknowledgements


In order to prove that MSCs incubated with PEG-HGNs
(50 µg mL−1) were able to colonize different scaffolds, Financial support from the EU through the ERC Consolidator
they were seeded onto fibrin or polycaprolactone (PCL) Grant (ERC-2013-CoG-614715, NANOHEDONISM) is gratefully
scaffolds. Fibrin scaffolds were prepared following our pre- acknowledged. CIBER-BBN is an initiative funded by the VI
vious report.44 Polycaprolactone scaffolds were prepared by National R&D&i Plan 2008–2011, Iniciativa Ingenio 2010,
electrospinning using an Yflow 2.2.D-500 electrospinner Consolider Program, CIBER Actions and financed by the
Published on 05 June 2017. Downloaded by University of California - San Diego on 29/06/2017 06:44:46.

(Electrospinning Machines/R&D Microencapsulation, Malaga, Instituto de Salud Carlos III (Spain) with assistance from the
Spain). In brief PCL pellets were dissolved in DCM/DMF European Regional Development Fund. We are grateful to the
(1 : 1) and the solution was stirred overnight. The polymer Scientific Services of the Aragon Institute of Health Sciences
solution was then electrospun using a voltage of 10.25 kV. The and to the LMA.
flow rate and the spinning distance were fixed at 0.5 mL h−1
and 19 cm, respectively. The spun fibres were collected on
a static plate (covered with aluminium foil) connected to a
negative voltage power supply, at a voltage of 3.11 kV for Notes and references
18 hours.
Briefly, 4 × 104 control cells or cells incubated with 1 T. G. Koch, L. C. Berg and D. H. Betts, Circ. Vasc. J., 2009,
PEG-HGNs were seeded in the centre of PCL membranes (into 50, 155–165.
MW-24 culture plates). One hour later, 600 µL of culture 2 B. A. Nasseri, K. Ogawa and J. P. Vacanti, Surgery, 2001,
medium were added to each well. Then, cells were incubated 130, 781–784.
for an additional 24 h. Next, PCL membranes were fixed with 3 R. Atala, A. Lanza, R. Thomson and J. A. Nerem,
4% paraformaldehyde for 20 min at room temperature. After Principles of regenerative medicine, Elsevier, Canada, 2nd
two washing cycles with PBS the membranes were placed on a edn, 2010.
microscopy slide and cell nuclei were stained with Draq5 4 I. R. Murray, C. C. West, W. R. Hardy, A. W. James,
diluted in Mowiol. Subsequently, a cover was placed on the T. S. Park, A. Nguyen, T. Tawonsawatruk, L. Lazzari, C. Soo
membranes for facilitating visualization by confocal and B. Peault, Cell. Mol. Life Sci., 2014, 71, 1353–1374.
microscopy. Samples were observed using a Leica TCA SP2 con- 5 M. M. Encabo-Berzosa, M. Gimeno, L. Lujan, M. Sancho-
focal microscope. In all cases, Z acquisitions of different Albero, L. Gomez, V. Sebastian, M. Quintanilla, M. Arruebo,
sample areas were performed in order to visualize different J. Santamaria and P. Martin-Duque, RSC Adv., 2016, 6,
planes of the membrane. 58723–58732.
In parallel, 3.75 × 104 control or pre-incubated with NPs 6 T. Sadhukha, T. D. O’Brien and S. Prabha, J. Controlled
(50 µg mL−1), MSCs were seeded onto fibrin gels. In this case Release, 2014, 196, 243–251.
the MSCs employed were able to express the GFP protein, 7 B. Demirayak, N. Yüksel, O. S. Çelik, C. Subas, G. Duruksu,
which facilitated their visualization by fluorescence Z. S. Unal, D. Kürs and E. Karaöz, Exp. Eye Res, 2016, 151,
microscopy. The GFP positive cells were kindly provided 227–235.
by Rebeca González. In this experiment, different planes 8 F. R. Melo, R. B. Bressan, S. Forner, A. C. Martini, M. Rode,
of the gels were acquired at different times using a multi- P. B. Delben, G. A. Rae, C. P. Figueiredo and A. G. Trentin,
dimensional microscope with a real time control Leica AF6000 Cell. Mol. Neurobiol., 2017, 37, 941–947.
LX, which allowed visualizing the in vivo colonization of 9 H. Saeed, M. Ahsan, Z. Saleem, M. Iqtedar, M. Islam,
the gel. Z. Danish and A. M. Khan, J. Biomed. Sci., 2016, 23, 41.
In order to evaluate the capacity of the cells to proliferate 10 O. V. Salata, J. Nanobiotechnol., 2004, 6, 1–6.
inside the scaffolds, MSCs containing nanoparticles (or 11 K.-B. L. Aniruddh Solanki and J. D. Kim, Futur. Med., 2008,
without them) were seeded onto both types of scaffolds. 3, 567–578.
Subsequently, the proliferation of cells was evaluated by the 12 A. Tremel, A. Cai, N. Tirtaatmadja, B. D. Hughes,
AlamarBlue® assay 4 and 24 h after cell seeding. G. W. Stevens, K. A. Landman and A. J. O. Connor, Chem.
Eng. Sci., 2009, 64, 247–253.
13 W.-K. Ko, D. N. Heo, H.-J. Moon, S. J. Lee, M. S. Bae,
Statistical analysis J. B. Lee, I.-C. Sun, H. B. Jeon, H. K. Park and I. K. Kwon,
All the data were expressed as the mean ± standard deviation J. Colloid Interface Sci., 2015, 438, 68–76.
and all the experiments were performed in triplicate. The stat- 14 X. Huang, X. Teng, D. Chen, F. Tang and J. He,
istical analysis of the data with a normal distribution was per- Biomaterials, 2010, 31, 438–448.
formed using Student’s t-test and one way analysis of variance 15 C. Lambertz, S. Pierrat, M. Tarantola, D. Schneider,
using software Stata/SE 12.0. For the data groups with a non- E. Sunnick, A. Janshoff and C. Sonnichsen, Beilstein
normal distribution, the Wilcoxon Rank Sum test and the J. Nanotechnol., 2014, 5, 2479–2488.
Kruskal–Wallis test were employed. p < 0.05 was considered 16 Y. Zhang, L. Hu, D. Yu and C. Gao, Biomaterials, 2010, 31,
statistically significant. 8465–8474.

Nanoscale This journal is © The Royal Society of Chemistry 2017


View Article Online

Nanoscale Paper

17 M. Pacurari, Y. Qian, W. Fu, D. Schwegler-Berry, M. Ding, 31 X. Peng, E. S. Nelson, J. L. Maiers and K. A. DeMali, Int.
V. Castranova and N. L. Guo, J. Toxicol. Environ. Health, Rev. Cell Mol. Biol., 2011, 287, 191–231.
Part A, 2012, 75, 112–128. 32 S. Hackenberg, A. Scherzed, M. Kessler, S. Hummel,
18 C. Y. Tay, P. Cai, M. I. Setyawati, W. Fang, L. P. Tan, A. Technau, K. Froelich, C. Ginzkey, C. Koehler, R. Hagen
C. H. L. Hong, X. Chen and D. T. Leong, Nano Lett., 2014, and N. Kleinsasser, Toxicol. Lett., 2011, 201, 27–33.
14, 83–88. 33 Y. Hou, K. Cai, J. Li, X. Chen, M. Lai, Y. Hu, Z. Luo, X. Ding
19 I. Ortega-Oller, M. Padial-Molina, P. Galindo-Moreno, and D. Xu, Int. J. Nanomed., 2013, 8, 3619–3630.
Published on 05 June 2017. Downloaded by University of California - San Diego on 29/06/2017 06:44:46.

F. O’Valle, A. B. Jódar-Reyes and J. M. Peula-García, BioMed 34 M. Uz, V. Bulmus and S. Alsoy Altinkaya, Langmuir, 2016,
Res. Int., 2015, 2–18. 32, 5997–6009.
20 S. Song, Y. Hao, X. Yang, P. Patra and J. Chen, J. Nanosci. 35 D. Zhang, D. Liu, J. Zhang, C. Fong and M. Yang, Mater.
Nanotechnol., 2016, 16, 2582–2586. Sci. Eng., C, 2014, 42, 70–77.
21 J. J. Li, N. Kawazoe and G. Chen, Biomaterials, 2015, 54, 36 J. Li, J. J. Li, J. Zhang, X. Wang, N. Kawazoe and G. Chen,
226–236. Nanoscale, 2016, 8, 7992–8007.
22 N. Fairbairn, A. Christofidou, A. G. Kanaras, T. a. Newman 37 D. Zhang, D. Liu, J. Zhang, C. Fong and M. Yang, Mater.
and O. L. Muskens, Phys. Chem. Chem. Phys., 2013, 15, Sci. Eng., C, 2014, 42, 70–77.
4163–4168. 38 M. Y. Changqing Yi, D. Liu, C.-C. Fong and J. Zhang, ACS
23 C. Fernandes, C. Pereira, A. Guedes, S. L. H. Rebelo and Nano, 2010, 4, 6439–6448.
C. Freire, Appl. Catal., A, 2014, 486, 150–158. 39 N. Artigas, C. Ureña, E. Rodríguez-Carballo, J. L. Rosa and
24 B. Díaz, C. Sánchez-Espinel, M. Arruebo, J. Faro, E. de F. Ventura, J. Biol. Chem., 2014, 289, 27105–27117.
Miguel, S. Magadán, C. Yagüe, R. Fernández-Pacheco, 40 S. E. Smith, R. A. White, D. A. Grant and S. A. Grant,
M. R. Ibarra, J. Santamaría and A. González-Fernández, J. Nanosci. Nanotechnol., 2016, 16, 1160–1169.
Small, 2008, 4, 2025–2034. 41 K. Baranes, M. Shevach, O. Shefi and T. Dvir, Nano Lett.,
25 A. M. Alkilany and C. J. Murphy, J. Nanopart. Res., 2010, 12, 2016, 16, 2916–2920.
2313–2333. 42 V. Cebrian, F. Martin-Saavedra, L. Gomez, M. Arruebo,
26 B. Chithrani, A. Ghazani and W. Chan, Nano Lett., 2006, 6, J. Santamaria and N. Vilaboa, Nanomedicine, 2013, 9, 646–
662–668. 656.
27 J. Rejman, M. Nazarenus, D. J. de Aberasturi, A. H. Said, 43 K. J. Livak and T. D. Schmittgen, Methods, 2001, 25, 402–
N. Feliu and W. J. Parak, J. Colloid Interface Sci., 2016, 482, 408.
260–266. 44 F. M. Martin-Saavedra, V. Cebrian, L. Gomez, D. Lopez,
28 A. C. Wong and D. W. Wright, Small, 2016, 1–9. M. Arruebo, C. G. Wilson, R. T. Franceschi, R. Voellmy,
29 D. A. Lauffenburger and A. F. Horwitz, Cell, 1996, 84, 359–369. J. Santamaria and N. Vilaboa, Biomaterials, 2014, 35,
30 W. H. Goldmann, Cell Biol. Int., 2016, 40, 241–256. 8134–8143.

This journal is © The Royal Society of Chemistry 2017 Nanoscale

You might also like