Download as pdf or txt
Download as pdf or txt
You are on page 1of 12

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/268877830

Cationic polystyrene nanospheres induce autophagic cell death through the


induction of endoplasmic reticulum stress

Article  in  Nanoscale · November 2014


DOI: 10.1039/c4nr05509h · Source: PubMed

CITATIONS READS

136 580

6 authors, including:

Tian Xia Yu Hsuan Lee


University of California, Los Angeles National Cheng Kung University
265 PUBLICATIONS   44,866 CITATIONS    37 PUBLICATIONS   1,280 CITATIONS   

SEE PROFILE SEE PROFILE

Chun-Wan Chen
Institute of Labor, Occupational Safety And Health, Taiwan
42 PUBLICATIONS   1,818 CITATIONS   

SEE PROFILE

Some of the authors of this publication are also working on these related projects:

PLGA nanoparticles for immune tolerance View project

Filter Quality of Pleated Filter Cartridges View project

All content following this page was uploaded by Tian Xia on 18 August 2015.

The user has requested enhancement of the downloaded file.


Nanoscale
View Article Online
PAPER View Journal | View Issue
Published on 17 November 2014. Downloaded by University of California - Los Angeles on 18/08/2015 22:17:24.

Cationic polystyrene nanospheres induce


Cite this: Nanoscale, 2015, 7, 736
autophagic cell death through the induction of
endoplasmic reticulum stress†
Hui-Wen Chiu,a,e,f Tian Xia,b Yu-Hsuan Lee,a Chun-Wan Chen,c Jui-Chen Tsai*d and
Ying-Jan Wang*a,g,h

Nanoparticles (NPs) have been used to produce a wide range of products that have applications in
imaging and drug delivery in medicine. Due to their chemical stability, well-controlled sizes and surface
charges, polystyrene (PS) NPs have been developed as biosensors and drug delivery carriers. However, the
possible adverse biological effects and underlying mechanisms are still unclear. Recently, autophagy has
been implicated in the regulation of cell death. In this study, we evaluated a library of PS NPs with
different surface charges. We found that NH2-labeled polystyrene (NH2-PS) nanospheres were highly
toxic with enhanced uptake in macrophage (RAW 264.7) and lung epithelial (BEAS-2B) cells. Furthermore,
NH2-PS could induce autophagic cell death. NH2-PS increased autophagic flux due to reactive oxygen
species (ROS) generation and endoplasmic reticulum (ER) stress caused by misfolded protein aggregation.
Received 21st September 2014, The inhibition of ER stress decreased cytotoxicity and autophagy in the NH2-PS-treated cells. In addition,
Accepted 9th November 2014
the Akt/mTOR and AMPK signaling pathways were involved in the regulation of NH2-PS-triggered auto-
DOI: 10.1039/c4nr05509h phagic cell death. These results suggest an important role of autophagy in cationic NP-induced cell death
www.rsc.org/nanoscale and provide mechanistic insights into the inhibition of the toxicity and safe material design.

1. Introduction systems. However, the understanding of the interactions of


NPs with biological systems is still rudimentary.2 Polystyrene
Applications of nanoparticles (NPs) are prevalent and continue (PS) NPs can be easily synthesized in a wide range of sizes,
to grow rapidly. Industrial and commercial applications of which facilitates their application as biosensors as well as
NPs include catalysis, sensors, environmental remediation, in photonics and self-assembling nanostructures.3,4 Specific-
personal care products and cosmetics, and NPs show great surface modification, high drug-loading capacity and colloidal
promise in the field of medicine, including imaging and drug stability in biological media also contribute to their appli-
delivery.1 The development of nanotechnology calls for a com- cation as experimental drug carrier systems.5 Our group has
prehensive understanding of the impact of NPs on biological shown that NH2-labeled PS (NH2-PS) nanospheres can induce
cell death in macrophage and lung epithelial cells with apop-
totic and necrotic features, respectively.6 Furthermore, we
a
Department of Environmental and Occupational Health, National Cheng Kung found that macrophage cells used an endosomal–lysosomal
University, Tainan, Taiwan. E-mail: yjwang@mail.ncku.edu.tw; route of uptake, while lung epithelial cells used a caveolar
Fax: +886-6-2752484; Tel: +886-6-235-3535 ext. 5804
b
uptake mechanism.6 Considering the proposed nanomedical
Division of NanoMedicine, Department of Medicine, University of California,
Los Angeles, California, USA
applications of PS NPs, an evaluation of these processes
c
Institute of Labor, Occupational Safety and Health, Ministry of Labor, Executive becomes even more important as these processes may result in
Yuan, Taiwan potentially unwanted consequences.
d
Institute of Clinical Pharmacy and Pharmaceutical Sciences, National Cheng Kung It is now known that different modalities of cell death
University, Tainan, Taiwan. E-mail: jctsai@mail.ncku.edu.tw; Fax: +886-6-237-3149;
(apoptosis, necrosis, autophagy) contribute to the patho-
Tel: +886-6-235-3535 ext. 5689
e
Division of Nephrology, Department of Internal Medicine, Shuang Ho Hospital,
physiology of different human disorders.7 Autophagy is a protein
Taipei Medical University, Taiwan degradation system in which cellular proteins and organelles
f
Graduate Institute of Clinical Medicine, Taipei Medical University, Taipei, Taiwan are sequestered, delivered to lysosomes, and digested by lyso-
g
Department of Biomedical Informatics, Asia University, Taichung, Taiwan somal hydrolases. In normal cells, autophagy functions to
h
Department of Medical Research, China Medical University Hospital, China
maintain homeostasis by eliminating excessive or unnecessary
Medical University, Taichung, Taiwan
† Electronic supplementary information (ESI) available. See DOI: 10.1039/ proteins.8 In recent years, the role of autophagy as an alterna-
c4nr05509h tive cell death mechanism has been a topic of debate. A clearer

736 | Nanoscale, 2015, 7, 736–746 This journal is © The Royal Society of Chemistry 2015
View Article Online

Nanoscale Paper

understanding of the signaling pathways involved in auto- bovine serum (FBS) (HyClone, South Logan, UT). BEAS-2B cells
phagy as well as its role in inducing cell death is highly desir- were cultured in LHC-9 serum-free medium (Gibco BRL, Grand
able. Recently, treatment with NPs from various sources has Island, NY). The cells were incubated under a humidified
been shown to induce autophagy in cell lines.9–12 However, the atmosphere containing 5% CO2 at 37 °C. Exponentially
roles of NP-induced autophagy in cell survival or cell death are growing cells were detached with 0.05% trypsin-EDTA (Gibco
still unclear. Thus, a better understanding of the implication BRL, Grand Island, NY) in DMEM or LHC-9 medium. PS nano-
Published on 17 November 2014. Downloaded by University of California - Los Angeles on 18/08/2015 22:17:24.

and biological significance of PS NP-induced cell death will spheres were obtained from Bangs Laboratory (Fishers, IN).
help us understand the risks associated with its uses and These include 60 nm unmodified (PS), 60 nm carboxylated
develop safer nanotechnology. (COOH-PS) and 60 nm amino-modified (NH2-PS) polystyrene
Among the factors contributing to autophagy, there is particles. Fluorescent PS and COOH-PS were obtained from
increasing evidence that endoplasmic reticulum (ER) stress Bangs Laboratory and fluorescent NH2-PS was obtained from
plays an important role.13 The ER, which functions in protein Nanocs Inc. (New York, NY). For all experiments and analyses,
folding and assembly, lipid biosynthesis, vesicular traffic and water was deionized and filtered with 0.45 μm pore size Acro-
cellular calcium storage, is sensitive to alterations in homeo- disc Syringe Filters (Pall Corporation, NY). All of the NP solu-
stasis.14 Proper functioning of the ER is essential to autophagy tions were freshly prepared from stock solutions (5 mg mL−1)
and cell survival.15 Reactive oxygen species (ROS), misfolded and sonicated for 30 s before addition to cell cultures.
protein aggregates, DNA damage, hypoxia and hypocalcemia
can induce ER stress.16,17 If the cells are exposed to prolonged 2.2 Physicochemical characterization
or robust ER stress, they die by apoptosis.18 Previous studies The average hydrodynamic size, zeta potential and polydisper-
have demonstrated that the ER stress response in combination sity index (PDI) of all NPs were determined by dynamic laser
with autophagy represents an adaptive mechanism for sup- scattering (Delsa™ Nano C, Beckman Coulter, Inc., USA). This
porting cell survival in response to a great variety of detrimen- instrument is capable of measuring particles in the size range
tal conditions.19 Recently, silver NP-induced apoptosis has of 0.6 nm to 7 μm. The size measurements were performed on
been reported to be mediated by the ER stress-signaling dilute NP suspensions in aqueous solution and DMEM
pathway.20 Christen and Fent indicated that silica NPs and (including 10% FBS).
silver-doped silica NPs induced the ER stress response, as
demonstrated in induced expression of BiP and splicing of 2.3 Cell viability assay
XBP1 mRNA, two selective markers of ER stress in human liver The cellular viability was measured by the MTS assay, which
cells.21 However, whether ER stress is induced by PS NPs is looks at the reduction of 3-(4,5-dimethylthiazol-2-yl)-5-(3-carb-
still unknown. oxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium (MTS) to
Our previous study has demonstrated that NH2-PS nano- formazan in viable cells. Briefly, cells were plated onto 96-well
spheres were highly toxic to RAW 264.7 cells (a mouse macro- plates (Thermo, MA, USA). After incubation with the indicated
phage cell line) and BEAS-2B cells (a human bronchial dose of NPs for various lengths of time at 37 °C, the formazan
epithelial cell line).6 In this study, RAW 264.7 and BEAS-2B cells absorbance was measured at 490 nm. The mean absorbance of
were used to investigate the autophagic effects and ER stress non-exposed cells was the reference value for calculating 100%
induced by NH2-PS nanospheres. We examined whether NH2- cellular viability.
PS-induced autophagy could serve as a pro-survival or cell death
mechanism. Furthermore, we also examined the signaling path- 2.4 PS NP uptake using fluorescence confocal microscopy
ways associated with the process of autophagy induced by NH2- and flow cytometry
PS in RAW 264.7 and BEAS-2B cells. Our data demonstrated We plated cells onto 6-well plates with a glass coverslip per
clearly the involvement of ER stress and the importance of the well. After PS NP exposure, cells were fixed with 4% para-
Akt/mTOR and AMPK signaling pathways in NH2-PS-induced formaldehyde. After three washes in PBS, the cells were stained
autophagy, which served primarily as a pro-death pathway. with 4′-6-diamidino-2-phenylindole (DAPI) (Sigma, MO, USA).
Fluorescence confocal images were obtained using a confocal
microscope (Carl Zeiess LSM780, Instrument Development
2. Experimental Center, NCKU). The uptake of particles by cells was also ana-
lyzed by flow cytometry (Becton Dickinson, San Jose, Califor-
2.1 Cell culture and co-incubation with NPs nia). The side scatter (SSC) data were analyzed using
The mouse macrophage cell line RAW 264.7 (ATCC TIB-71) and CELLQuest™ software (Becton Dickinson). Ten thousand cells
the human bronchial epithelial cell line BEAS-2B (ATCC were acquired for each measurement.
CRL-9609) were obtained from the American Type Culture Col-
lection (ATCC). RAW 264.7 cells were cultured in Dulbecco’s 2.5 ROS production measurement
modified essential medium (DMEM) (Gibco BRL, Grand To measure ROS generation, a fluorometric assay using the
Island, NY), containing 4.5 g l−1 D-glucose, supplemented with intracellular oxidation of 2,7-dichlorofluorescein diacetate
antibiotics containing 100 U ml−1 penicillin, 100 μg ml−1 (DCFH-DA, Sigma, MO, USA) was performed.22 The cells were
streptomycin (Gibco BRL, Grand Island, NY) and 10% fetal treated with different concentrations of the NH2-PS nano-

This journal is © The Royal Society of Chemistry 2015 Nanoscale, 2015, 7, 736–746 | 737
View Article Online

Paper Nanoscale

spheres for 2, 4 or 6 hours and were then incubated with was obtained from Abcam (Cambridge, MA, USA); anti-
10 μM DCFH-DA for 30 min. After washing with PBS, the LC3 was obtained from Abgent (San Diego, CA, USA); anti-p62/
DCFH fluorescence of the cells from each well was measured SQSTM1 was obtained from MBL (Nagoya, Japan); anti-mTOR
on a fluorescence microplate reader (Thermo, MA, USA) at an and anti-p70S6 K were obtained from Epitomics (Burlingame,
excitation wavelength of 485 nm and emission at 530 nm. The CA, USA).
intensity of fluorescence reflects the extent of oxidative stress.
Published on 17 November 2014. Downloaded by University of California - Los Angeles on 18/08/2015 22:17:24.

2.10 RNA interference (RNAi)


2.6 Staining of misfolded proteins
We used the Arrest-In Transfection Reagent (Thermo, MA,
The misfolded proteins were measured using a ProteoStat USA) to transfect cells according to the manufacturer’s proto-
Aggresome Detection Kit (Enzo Life Sciences, NY, USA) accord- col. RNAi reagents were obtained from the National RNAi Core
ing to the manufacturer’s protocol. Briefly, the cells were fixed Facility located at the Institute of Molecular Biology/Genomic
with 4% paraformaldehyde and then the permeabilizing solu- Research Center, Academia Sinica, supported by the National
tion was added over 30 min. The cells were washed twice and Core Facility Program for Biotechnology Grants of NSC
the substrate solution was added to each sample over 30 min. (NSC 100-2319-B-001-002). The human library should be
Finally, the cells were washed twice and the Dual Detection referred to as TRC-Hs 1.0. Individual clones are identified as
Reagent was added over 30 min. After staining, the cells were shRNA TRCN0000010171, shRNA TRCN0000000859, shRNA
analyzed under a fluorescence microscope (Olympus, Japan). TRCN0000002168 and shRNA TRCN0000072178.
2.7 Staining of ER
The cells were treated with NH2-PS nanospheres. Sixteen hours
later, the ER-Tracker Blue-White DPX (Molecular Probes,
3. Results and discussion
Eugene, OR) probe was added to the cells and incubated for 3.1 PS NP characterization
30 min under the same growth conditions. The loading solu- Non-labeled (PS), NH2-PS and carboxy-labeled (COOH-PS) with
tion was removed, and the cells were then washed with PBS. a primary particle size of 60 nm were purchased from Bangs
Microscopic images were collected using the fluorescence Laboratory. The detailed physicochemical characteristics of PS
microscope. Cells were pretreated with the ER stress inhibitor, NPs after suspension in cell culture medium or water are pre-
tauroursodeoxycholic acid (TUDCA) (Merck KGaA, Darmstadt, sented in Table 1. Non-labeled (PS), NH2-PS and carboxy-
Germany), for 1 h before NH2-PS treatment. labeled (COOH-PS) NPs have similar hydrodynamic sizes of
approximately 60 nm in water, confirming their primary par-
2.8 Immunofluorescence microscopy
ticle size as provided by the manufacturer. The zeta potential
The cells were cultured on coverslips. After NH2-PS nanosphere of NH2-PS in water is positive (+34.97), whereas plain PS and
treatment, the cells were fixed in 4% paraformaldehyde and COOH-PS have negative charges. As previously reported, all of
blocked with 1% BSA for 30 min. This was followed by incu- the NPs have negatively charged surfaces in DMEM due to the
bation with a specific antibody against LC3 (MBL, Japan) for formation of a corona of negatively charged proteins.6 NPs
1 h. After washing, the cells were labeled with a DyLight™ 488- with a zeta potential above (±)30 mV have been shown to be
conjugated affinipure goat anti-rabbit IgG (Jackson Immuno- stable in suspension because the surface charge prevents
Research Laboratories, PA, USA) for 1 h and with DAPI. Finally, aggregation of the particles.24 Thus, the zeta potential of
the cells were washed in PBS, covered with a coverslip, and NPs in water indicates that they have surface properties
examined with a fluorescence microscope or confocal micro- that provide good stability in suspension. All NPs showed a
scope (Carl Zeiess LSM780, Instrument Development Center, relatively low polydispersity index (PDI): the values for PS,
NCKU). To quantify LC3-positive cells, a minimum of 50 cells NH2-PS and COOH-PS in water were 0.049, 0.113 and 0.112,
per sample was counted, and the number of LC3 aggregates respectively. Previous studies have indicated that a PDI lower
was enumerated. The data are presented as a percentage of than 0.2 is associated with a high homogeneity in the particle
LC3-positive cells within the total of the cells examined. population.25 In summary, these particles formed a stable
suspension in aqueous media.
2.9 Western blot analysis
Total cellular protein lysates were prepared by harvesting cells
in protein extraction buffer for 1 h at 4 °C as described pre- Table 1 Physical characteristics of PS NPs

viously.23 The densities of the bands were quantified with a Hydrodynamic Zeta potential
computer densitometer (AlphaImager™ 2200 System Alpha diameter (nm) PDIa (mV)
Innotech Corporation, San Leandro, CA, USA). The expression
Water DMEM Water DMEM Water DMEM
of GAPDH was used as the protein loading control. The anti-
bodies for detecting Akt, phospho-Akt, phospho-p70S6 K, PS 61.3 86.7 0.049 0.080 −33.99 −14.01
phospho-AMPK, AMPK and Beclin 1 were obtained from Cell NH2-PS 62.1 206.1 0.113 0.240 +34.97 −12.33
COOH-PS 62.0 97.1 0.112 0.143 −40.06 −14.01
Signaling Technology (Ipswich, MA, USA); anti-phospho-mTOR
a
was obtained from Millipore (Billerica, MA, USA); anti-GAPDH PDI is the polydispersity index.

738 | Nanoscale, 2015, 7, 736–746 This journal is © The Royal Society of Chemistry 2015
View Article Online

Nanoscale Paper

3.2 Cytotoxic effects and cellular uptake of PS NPs in RAW PS NPs into the cells, we used fluorescent PS NPs (Fig. 1D).
264.7 and BEAS-2B cells Confocal microscopic analysis showed that exposure to NH2-PS
increased the fluorescence intensity of both RAW 264.7 and
To confirm that the observed toxic effects are specifically BEAS-2B cells. However, only weak intracellular fluorescence
related to the NH2 substitution on the particle surface, we was observed when the cells were treated with PS or COOH-PS.
compared NH2-PS with COOH-PS and PS nanospheres Furthermore, the side scatter (SSC) intensity analyzed by flow
Published on 17 November 2014. Downloaded by University of California - Los Angeles on 18/08/2015 22:17:24.

(Fig. 1A). The results demonstrated that only NH2-PS but cytometry also confirmed that the NH2-PS was apparently
not PS or COOH-PS particles decreased the cell viability. engulfed by the BEAS-2B cells, whereas low levels of cellular
In addition, we further examined the viability of the cells uptake of PS and COOH-PS were observed (Fig. 1E and F). Con-
treated with different doses and time points for NH2-PS and sistent with our findings, numerous reports in the literature
found that cell death was dependent on both the concen- show that the rate of uptake is significantly higher for the posi-
tration and time in both RAW 264.7 and BEAS-2B cells (Fig. 1B tively charged NPs than for their negatively charged counter-
and C). The uptake of NPs by cells is an important factor in parts.27,28 Previous studies have demonstrated that surface
the assessment of their toxicity.26 To evaluate the entry of the functionalization of NPs is crucial for particle durability, sus-

Fig. 1 Cell viability detection by the MTS assay and cellular uptake of PS NPs. (A) After stimulation of RAW 264.7 and BEAS-2B cells with 20 μg ml−1
of the different types of PS nanospheres for 16 hours, cell viability was determined using the MTS assay. *, p < 0.05, PS versus control. (B and C) Con-
centration- and time-dependent effects of NH2-PS on the viability of RAW and BEAS-2B cells. The cells were treated with 1, 5, 10, 20 or 40 μg ml−1
NH2-PS for 4, 8, 16 or 24 hours. After stimulation with NH2-PS particles, the cells were incubated with the MTS reagent for 30 min, and the absor-
bance was measured at 490 nm. All of the MTS values of different doses were normalized according to the control values (no particle exposure),
which were regarded as 100% cell viability. (D) Uptake of PS NPs detected by fluorescence confocal microscopy. The different types of PS NPs are
shown in red and DAPI (blue) is a nuclei-specific marker. The cells were treated with 20 μg ml−1 PS NPs for 8 hours. (E) The results of SSC light of
flow cytometry demonstrated that PS NPs were apparently engulfed by BEAS-2B cells. The cells were treated with PS NPs at 10, 20 or 40 μg ml−1 for
8 hours. (F) Quantification of the scatter intensity in BEAS-2B cells with PS NP treatment. The data are presented as the mean ± standard deviation
of three independent experiments.

This journal is © The Royal Society of Chemistry 2015 Nanoscale, 2015, 7, 736–746 | 739
View Article Online

Paper Nanoscale

pensibility in biological media, biocompatibility, and biodistri- dye, suggesting a remarkable induction of ER stress. However,
bution.29 Recent findings, including ours, have indicated that the fluorescence staining intensity did not increase after treat-
cationic particles have more adverse effects than anionic ment with PS or COOH-PS (Fig. S2B†). Furthermore, ER stress
particles.6,30–32 Moreover, it is known that the cellular toxicity is initiated by three ER transmembrane proteins: RNA-depen-
and apoptosis induced by many of these non-viral transfection dent protein kinase (PKR)-like ER kinase (PERK), inositol-
systems can be attributed to their cationic charge and inter- requiring protein 1 (IRE1) and activating transcription factor 6
Published on 17 November 2014. Downloaded by University of California - Los Angeles on 18/08/2015 22:17:24.

ference in proton pump activities.33,34 The toxicity of cationic (ATF6). These three ER stress sensors trigger divergent and
NPs has been reported to be related to their enhanced inter- convergent signaling cascades that lead to adaptation or cell
actions with the cell membrane, a feature initially mediated by death.46 We therefore measured the expression of ER stress-
their strong electrostatic attraction to the negatively charged related proteins in RAW 264.7 and BEAS-2B cells and found
bilayer.31,35 The currently available evidence indicated that dose-dependent increases in the expression of IRE1α in the
the protein corona plays an important role in the interactions cells treated with NH2-PS compared with the control cells
between NPs and biological systems.36,37 Living systems usually (Fig. 2D). However, the expression of IRE1α did not change
interact with protein-coated NPs rather than bare NPs, and the after treatment with PS or COOH-PS (Fig. S2C & S2D†). In
structure, dynamics and stability of the corona can be decisive addition, to determine whether ROS generated by NH2-PS
factors governing the biological response of cells and organisms contributes directly to ER stress, we applied the anti-oxidant
to NP exposure.38,39 Furthermore, the cellular uptake of NPs is N-acetylcysteine (NAC). Cells pretreated with NAC and then sub-
strongly affected by the presence and the physicochemical pro- jected to NH2-PS treatment were analyzed to determine ROS
perties of the protein corona around these NPs.37 NPs that carry levels. We observed that pretreatment with NAC decreased
a positive surface charge attract different protein components of NH2-PS-induced ROS and misfolded protein aggregates (Fig. 2E
the medium from those proteins carrying a negative surface and F). Additionally, NAC pretreatment significantly attenuated
charge. Nevertheless, the underlying mechanisms of how cat- the NH2-PS-induced upregulation of IRE1α expression (Fig. 2G).
ionic NPs disrupt cellular signaling and the role of the protein The observation confirms the induction of misfolded protein
corona are still unclear and require further investigation. aggregation and ER stress via an oxidative stress mechanism.

3.3 ROS generation, misfolded protein aggregation and ER 3.4 Measurement of autophagy in RAW 264.7 and BEAS-2B
stress in RAW 264.7 and BEAS-2B cells treated with NH2-PS cells treated with NH2-PS
Induction of ROS-mediated cell death has been reported in a Various NPs have been well demonstrated to induce autophagic
variety of cells treated with different NPs.8,40,41 Our current events in cultured cells.47,48 Microtubule-associated protein
findings showed higher ROS levels in the RAW 264.7 and light chain 3 (LC3) is widely used to monitor autophagy.49
BEAS-2B cells exposed to NH2-PS than in the controls (Fig. 2A). Thus, we applied fluorescence microscopy to determine the
The generation of ROS in RAW 264.7 cells was significantly percentage of cells with punctate LC3 staining (Fig. 3) and
enhanced by 116%, 127%, 144% and 185% after 6 hours of found that NH2-PS increased the LC3 signals in RAW 264.7
treatment with 5, 10, 20 and 40 μg ml−1 of NH2-PS, respecti- and BEAS-2B cells in a time- and concentration-dependent
vely. Accumulating evidence suggests that ROS directly or manner. To detect the expression of the autophagic-related
indirectly affects ER homeostasis and protein folding.17 There- proteins (LC3, Beclin 1 and p62), we performed western blot-
fore, we further analyzed the misfolded protein aggregation. ting with lysates from RAW 264.7 and BEAS-2B cells treated
Hoechst 33342 staining and ProteoStat Aggresome Detection with different concentrations of NH2-PS (Fig. 4A). The
Kit were applied to measure the aggregated protein using a expression levels of the LC3-II, p62 and Beclin 1 proteins
fluorescence microscope, through detection of denatured increased with NH2-PS treatment. However, the levels of LC3-II
and/or misfolded protein cargo in fixed and permeabilized did not change when the cells were exposed to PS or COOH-PS
cells.40,42 We found an increased red signal representing (Fig. S2C & S2D†). In addition, we applied bafilomycin A1
higher levels of misfolded protein aggregates, in the NH2-PS- (BAF) to inhibit autophagic flux and found elevated levels of
treated cells (Fig. 2B). However, the red signal did not increase LC3-II in cells treated with NH2-PS but not in those treated
after treatment with PS or COOH-PS (Fig. S2A†). ER stress can with PS or COOH-PS (Fig. 4B & S3†), implying that NH2-PS-
activate signaling pathways involved in apoptosis and auto- induced autophagosome synthesis is regulated at a point
phagy.13 In mammalian cells, ER stress has been shown to upstream of the autophagosome–lysosome fusion. Previous
facilitate the formation of autophagosomes, and induction of studies have demonstrated that poly(amidoamine) (PAMAM)
autophagy enables the removal of toxic misfolded proteins.43 dendrimers induced both cytotoxicity and autophagic flux in a
Because autophagy has been invoked as a means of cell death panel of human glioma cell lines.50 Interestingly, our previous
under higher ER stress,44 we investigated whether the NH2-PS study also found that NH2-PS increased lysosomal permeabili-
could induce ER stress and subsequent autophagic cell death. zation, permitting escape by lysosomal rupture in RAW 264.7
Detection of ER stress can be achieved using ER-Tracker Blue- cells.6 The dual roles of NH2-PS in both autophagosome
White DPX, an ER-specific dye.45 The results shown in Fig. 2C synthesis and damage to the lysosomes, leading to blockage of
demonstrated that the treatment of the cells with NH2-PS sig- autophagosome–lysosome fusion, may explain in part the
nificantly increased the fluorescence staining intensity of this complex role of autophagy in cell death.51 In addition, it is

740 | Nanoscale, 2015, 7, 736–746 This journal is © The Royal Society of Chemistry 2015
View Article Online

Nanoscale Paper
Published on 17 November 2014. Downloaded by University of California - Los Angeles on 18/08/2015 22:17:24.

Fig. 2 ROS generation, misfolded protein aggregation and ER stress induced by NH2-PS in RAW 264.7 and BEAS-2B cells. (A) Concentration- and
time-dependent quantification of ROS generation in cells that were treated with NH2-PS and were then incubated with DCFH-DA for 30 min. The
fluorescence of the cells was immediately assayed using a fluorescence microplate reader. (B) The cells were treated with NH2-PS at 20 μg ml−1 for
16 hours and stained with Hoechst 33342 and then with a ProteoStat Aggresome Detection Kit. The red color and the blue color indicate the fluore-
scence of the detected aggregates and stained nuclei, respectively. (C) ER staining enhanced by NH2-PS. The cells were treated with NH2-PS at
20 μg ml−1 for 16 hours and were treated with the ER Tracker Blue-White DPX probe for ER staining. (D) Western blotting for IRE1α in RAW 264.7 and
BEAS-2B cells. The cells were treated with NH2-PS for 16 hours. (E) Effects of NAC on ROS generation induced by NH2-PS in RAW 264.7 cells. The
cells were pretreated with NAC for 1 h before NH2-PS treatment (20 μg ml−1) for 6 hours. *, p < 0.05, NH2-PS versus NH2-PS + NAC. (F) The cells
were pretreated with NAC (5 mM) for 1 h before NH2-PS at 20 μg ml−1 for 16 hours and stained with Hoechst 33342 and then with a ProteoStat
Aggresome Detection Kit in RAW 264.7 cells. (G) Western blotting for IRE1α. The cells were pretreated with NAC (5 mM) for 1 h before NH2-PS treat-
ment (20 μg ml−1) for 16 hours. The data are presented as the mean ± standard deviation of three independent experiments.

This journal is © The Royal Society of Chemistry 2015 Nanoscale, 2015, 7, 736–746 | 741
View Article Online

Paper Nanoscale
Published on 17 November 2014. Downloaded by University of California - Los Angeles on 18/08/2015 22:17:24.

Fig. 3 Immunofluorescence staining of the LC3 protein in RAW 264.7 and BEAS-2B cells treated with NH2-PS. (A and B) Representative cell images
showing punctate LC3 distribution after NH2-PS treatment. The cells were treated with 20 μg ml−1 of NH2-PS for 16 hours and were then examined
using a confocal microscope. (C and D) Concentration dependence of NH2-PS-induced punctate LC3 staining in RAW and BEAS-2B cells. The cells
were incubated with 0–40 μg ml−1 NH2-PS for 16 hours. (E and F) The time course of NH2-PS-induced punctate LC3 staining in RAW and BEAS-2B
cells. The cells were incubated with 20 μg ml−1 NH2-PS for 0–24 hours. *, p < 0.05, NH2-PS versus control. The data are presented as the mean ±
standard deviation of three independent experiments.

worth mentioning that gold nanorods (AuNRs) can escape alters NH2-PS-induced cytotoxicity. The results revealed a sig-
from the lysosomes occasionally and the escaped AuNRs are nificant decrease in LC3-II expression and cytotoxicity in cells
recycled back into the lysosomal system through autophagy.52 when the combined treatment was compared with NH2-PS
Recent evidence shows that NPs not only enter cells but also treatment alone (Fig. 4C and D). Our observations paralleled
trigger a disturbance of intracellular component. It has been that of other investigators.50 On the one hand, autophagy plays
reported that NPs caused massive disruption of the intracellu- a role as a cell survival mechanism that allows cells to remove
lar microtubule assembly and thereby led to limited cell moti- damaged cytoplasmic proteins and organelles through lyso-
lity.53 Furthermore, NPs can damage mitochondria and cause somal degradation and thus to survive metabolic stress.56 On the
lysosomal dysfunction, and then induce toxicity.54,55 There- other hand, autophagy has also been found to contribute to
fore, PS NPs may induce autophagy through the disruption of programmed cell death in response to various stimuli.57 It is
the intracellular organelles. In addition, apoptosis and necro- now known that autophagy can promote cell death by the
sis were examined using Annexin V-FITC/propidium iodide selective removal of survival factors and/or prolonged removal
staining. Using this technique, we found that NH2-PS not only of cellular constituents, resulting in the demise of cells.58 Pre-
induced autophagy but also apoptosis and necrosis (Fig. S1†). vious studies have indicated that the autophagy triggered by a
Pretreatment with a pan-caspase inhibitor, Z-VAD, significantly variety of NPs might be due in part to the early adaptive
prevented cell death (Fig. S1E†). responses to stress. However, these responses might sub-
Next, we used 3-methyladenine (3-MA), an inhibitor of sequently lead to cytotoxicity.8,9,48,59,60 In our current study,
autophagy, to determine whether inhibition of autophagy the role of autophagy as a contributor to cell death was further

742 | Nanoscale, 2015, 7, 736–746 This journal is © The Royal Society of Chemistry 2015
View Article Online

Nanoscale Paper

phate-activated protein kinase (AMPK) pathway are involved in


regulating autophagy.61 Therefore, to investigate whether the
Akt/mTOR and AMPK signaling pathways were involved in the
NH2-PS-induced autophagy, we performed western blotting to
evaluate the protein phosphorylation status (Fig. 5). The
results indicated that phosphorylation of Akt, mTOR and
Published on 17 November 2014. Downloaded by University of California - Los Angeles on 18/08/2015 22:17:24.

p70S6 K decreased and phosphorylation of AMPK increased in


cells treated with NH2-PS compared with the control in both
RAW 264.7 and BEAS-2B cells. However, the expression of the
proteins of the Akt/mTOR and AMPK signaling pathways did
not change in the RAW 264.7 or BEAS-2B cells treated with PS
or COOH-PS (Fig. S4A & S4B†). In our previous study, we
observed a steep decline in the cellular ATP levels in cells
treated with NH2-PS.6 Activation of AMPK has been observed
to increase with ROS generation and ATP depletion.8,62 Under
various stresses, such as ATP depletion, AMPK is activated by
increased catabolism. Our current findings further revealed
that ATP depletion by NH2-PS could enhance the phosphoryl-
ation of AMPK in RAW 264.7 and BEAS-2B cells, which trig-
gered autophagy via the suppression of mTOR.62
Next, we investigated whether the inhibition of Akt and
AMPK could change the levels of autophagic cell death
induced by NH2-PS. We applied the shRNA technology to
inhibit either Akt or AMPK expression. As shown in Fig. 6A
and B, the expression of the Akt and AMPK proteins was mark-
edly decreased by Akt and AMPK shRNA, respectively, com-
pared with those treated with control shRNA. We then
examined whether the reduced expression of Akt or AMPK
altered the NH2-PS-induced cytotoxicity (Fig. 6C). Transfection
with Akt shRNA significantly enhanced the cytotoxicity
induced by the NH2-PS treatment. Transfection with AMPK
shRNA significantly reduced the toxic effect of the NH2-PS
treatment in BEAS-2B cells. Furthermore, BEAS-2B cells trans-
Fig. 4 Autophagic cell death and autophagic flux induced by NH2-PS in
RAW 264.7 and BEAS-2B cells. (A) Western blotting for LC3-I, LC3-II,
p62/SQSTM1 and Beclin 1. The cells were treated with 0–40 μg ml−1
NH2-PS for 16 hours. (B) Autophagic flux was determined by western
blotting with anti-LC3 antibody. The cells were pretreated with bafilo-
mycin A1 (BAF) (10 nM) for 1 h before NH2-PS treatment (20 μg ml−1) for
16 hours. (C) Western blotting of LC3-I and LC3-II expression in the
absence or presence of 3-methyladenine (3-MA). (D) Cytotoxic effects
in the absence or presence of 3-MA. The cells were pretreated with
3-MA (3 mM) for 1 h before NH2-PS treatment (20 μg ml−1) for 16 hours.
*, p < 0.05, NH2-PS versus NH2-PS + 3-MA. The data are presented as
the mean ± standard deviation of three independent experiments.

confirmed by the autophagy inhibitor 3-MA. The results


demonstrated that 3-MA has a protective effect on NH2-PS-
induced cell death (Fig. 4D), suggesting that autophagy plays a
pro-death role in this case.

3.5 The Akt/mTOR and AMPK signaling pathways are


involved in the NH2-PS-induced autophagy in RAW 264.7 and
BEAS-2B cells
Fig. 5 Akt/mTOR (A) and AMPK (B) signaling pathway protein
Previous studies have demonstrated that the Akt/mammalian expression in RAW 264.7 and BEAS-2B cells treated with NH2-PS. The
target of rapamycin (mTOR) and the adenosine monophos- cells were treated with 0–40 μg ml−1 NH2-PS for 16 hours.

This journal is © The Royal Society of Chemistry 2015 Nanoscale, 2015, 7, 736–746 | 743
View Article Online

Paper Nanoscale
Published on 17 November 2014. Downloaded by University of California - Los Angeles on 18/08/2015 22:17:24.

Fig. 6 The Akt/mTOR and AMPK signaling pathways are involved in


NH2-PS-induced autophagy in BEAS-2B cells. (A and B) Western blotting
for Akt or AMPK. The cells were transfected with control, Akt or AMPK
shRNA for 48 hours. (C) Measurement of LC3-punctate cells in the
Fig. 7 Measurement of autophagy and cytotoxic effects in BEAS-2B
absence or presence of Akt or AMPK shRNA. (D) Cytotoxic effects in the
cells pretreated with TUDCA. (A) Effects of TUDCA on cytotoxicity
absence or presence of Akt shRNA or AMPK shRNA. The cells were
induced by NH2-PS. The cells were pretreated with TUDCA for 2 hours
transfected with control, Akt or AMPK shRNA for 48 hours and were
before NH2-PS treatment (20 μg ml−1) for 16 hours. *, p < 0.05, NH2-PS
then incubated with 20 μg ml−1 of NH2-PS for 16 hours. *, p < 0.05
versus NH2-PS + TUDCA. (B) Measurement of LC3-punctate cells in the
versus control shRNA + NH2-PS.
absence or presence of TUDCA. The cells were pretreated with TUDCA
(1 mM) for 2 hours before NH2-PS treatment (20 μg ml−1) for 16 hours.
*, p < 0.05, NH2-PS versus NH2-PS + TUDCA. The data are presented as
fected with AMPK shRNA showed a significant decrease in the
the mean ± standard deviation of three independent experiments. (C)
percentage of LC3 punctate cells compared with the NH2-PS Western blotting for phosphorylation of Akt, phosphorylation of AMPK,
treatment, whereas cells transfected with Akt shRNA showed a Akt and AMPK. The cells were pretreated with TUDCA (1 mM) for 2 hours
significant increase in the percentage of LC3 punctate cells before NH2-PS treatment (20 μg ml−1) for 16 hours.
(Fig. 6D). These results suggest that one of the mechanisms of
NH2-PS-induced autophagy could be mediated by inhibition of
Akt/mTOR and activation of AMPK signaling pathways in RAW of the Akt/mTOR and the activation of the AMPK signaling
264.7 and BEAS-2B cells. Recent studies have found that the pathway. We found that pretreatment with TUDCA retarded
Akt/mTOR and AMPK pathways play crucial roles in the regu- the NH2-PS-decreased expression of Akt phosphorylation
lation of both apoptosis and autophagy.63,64 Akt can activate and NH2-PS-increased expression of AMPK phosphorylation
mTOR and lead to inhibition of autophagy.65 By contrast, (Fig. 7C). Inhibition of ER stress has recently been suggested
AMPK is a universal autophagy activator.56 It has recently been as a novel therapeutic strategy in various diseases.66,67 For
reported that functionalized single-walled carbon nanotubes example, TUDCA may inhibit apoptosis by ameliorating ER
induced autophagic cell death in human lung cells through stress through the modulation of intracellular calcium and
Akt-TSC2-mTOR signaling.11 However, the role of the AMPK thus attenuate liver cell death.68 In addition, TUDCA treatment
pathway in the activation of autophagy in cells treated with was reported to attenuate tunicamycin-induced ER stress,
NPs has not previously been reported. To our knowledge, this autophagy and cell death in rat hepatocytes,69 implying that
is the first study demonstrating that NH2-PS exposure leads to autophagy was involved, at least in part, in ER stress-induced
autophagy induction through inhibition of the Akt/mTOR and cell death. Our current results showed that TUDCA increased
activation of the AMPK signaling pathways. Akt activation in NH2-PS-treated macrophage and lung epi-
thelial cells. Consistent with these results, TUDCA was found
3.6 Inhibition of ER stress decreases cytotoxicity and to activate Akt in skeletal muscles and myocardium.70,71 The
autophagy mechanisms through which TUDCA increases Akt activation
To test whether NH2-PS-induced ER stress contributes directly are still unclear. One of the possible mechanisms may be a
to cell death, we treated BEAS-2B cells with the chemical G-protein coupled signal cascade.72 Moreover, ER stress leads
chaperone tauroursodeoxycholic acid (TUDCA), which is to a release of calcium and subsequent activation of AMPK
known to selectively inhibit ER stress.66 The results indicated that inhibits mTOR, thereby promoting autophagy.73 Thus,
that TUDCA treatment decreased NH2-PS-induced cytotoxicity NH2-PS-induced ER stress and AMPK activation found in our
and autophagy (Fig. 7A and B). Next, we examined whether current study might play important roles in autophagy induc-
inhibition of ER stress altered the NH2-PS-induced inhibition tion and cell death.

744 | Nanoscale, 2015, 7, 736–746 This journal is © The Royal Society of Chemistry 2015
View Article Online

Nanoscale Paper

4. Conclusions 11 H. L. Liu, Y. L. Zhang, N. Yang, Y. X. Zhang, X. Q. Liu,


C. G. Li, Y. Zhao, Y. G. Wang, G. G. Zhang, P. Yang,
Taken together, our results indicate that NH2-PS increased F. Guo, Y. Sun and C. Y. Jiang, Cell Death Dis., 2011, 2,
ROS generation and produced misfolded protein aggregates, e159.
which lead to ER stress that results in autophagic cell death in 12 Y. Zhao, J. L. Howe, Z. Yu, D. T. Leong, J. J. Chu, J. S. Loo
RAW 264.7 and BEAS-2B cells. TUDCA, an ER stress inhibitor, and K. W. Ng, Small, 2013, 9, 387–392.
Published on 17 November 2014. Downloaded by University of California - Los Angeles on 18/08/2015 22:17:24.

decreased cytotoxicity and autophagy in the NH2-PS-treated 13 Y. Cheng and J. M. Yang, World J. Biol. Chem., 2011, 2, 226–
cells. Furthermore, NH2-PS-induced autophagic cell death may 231.
be mediated primarily through the inhibition of the Akt/ 14 R. J. Kaufman, Genes Dev., 1999, 13, 1211–1233.
mTOR signaling pathway and the activation of the AMPK sig- 15 M. K. Kim, H. S. Kim, I. K. Lee and K. G. Park, Exp. Diabetes
naling pathway. Specifically, cell viability in the NH2-PS treat- Res., 2012, 2012, 509437.
ment group was significantly increased when autophagy was 16 S. M. Schleicher, L. Moretti, V. Varki and B. Lu, Drug Resist.
inhibited by the autophagy inhibitor 3-MA compared with Updates, 2010, 13, 79–86.
cells treated with the vehicle, suggesting that autophagy 17 J. D. Malhotra and R. J. Kaufman, Antioxid. Redox Signaling,
may serve a pro-death role in NH2-PS-induced cell death. 2007, 9, 2277–2293.
In addition, NH2-PS increased the autophagic flux. Thus, in 18 X. Li, H. Zhu, H. Huang, R. Jiang, W. Zhao, Y. Liu, J. Zhou
addition to apoptosis and necrosis, autophagy should be con- and F. J. Guo, Mol. Cell. Biochem., 2012, 365, 99–108.
sidered as a potential cell death mechanism, providing intra- 19 A. H. Schonthal, Front. Biosci., Scholar Ed., 2012, 4, 412–
cellular selectivity for exposure to NH2-PS nanospheres. 431.
20 R. Zhang, M. J. Piao, K. C. Kim, A. D. Kim, J. Y. Choi,
J. Choi and J. W. Hyun, Int. J. Biochem. Cell Biol., 2012, 44,
Acknowledgements 224–232.
21 V. Christen and K. Fent, Chemosphere, 2012, 87, 423–434.
This study was supported by the Food and Drug Adminis-
22 G. Fotakis, E. Cemeli, D. Anderson and J. A. Timbrell,
tration, Ministry of Health and Welfare, Executive Yuan
Toxicol. in Vitro, 2005, 19, 481–489.
(DOH101-FDA-41301, DOH102-FDA-41702 and MOHW103-
23 H. W. Chiu, S. Y. Ho, H. R. Guo and Y. J. Wang, Autophagy,
FDA-41407) and Ministry of Labor, Executive Yuan, Taiwan
2009, 5, 472–483.
(1013080, 1023038 and 1033059). T.X. was supported by US
24 V. J. Mohanraj and Y. Chen, Trop. J. Pharm. Res., 2006, 5,
Public Health Service Grant U19 ES019528 and NSF/EPA DBI
561–573.
0830117 and 1266377. The Instrument Development Center of
25 S. Saremi, F. Atyabi, S. P. Akhlaghi, S. N. Ostad and
the National Cheng Kung University (NCKU) provided techni-
R. Dinarvand, Int. J. Nanomed., 2011, 6, 119–128.
cal support with the Carl Zeiess LSM780 laser-scanning
26 C. Y. Tay, Y. Yu, M. I. Setyawati, J. Xie and D. T. Leong,
microscope.
Nano Res., 2014, 7, 805–815.
27 O. Harush-Frenkel, N. Debotton, S. Benita and
References Y. Altschuler, Biochem. Biophys. Res. Commun., 2007, 353,
26–32.
1 A. Nel, T. Xia, L. Madler and N. Li, Science, 2006, 311, 622– 28 K. T. Thurn, E. Brown, A. Wu, S. Vogt, B. Lai, J. Maser,
627. T. Paunesku and G. E. Woloschak, Nanoscale Res. Lett.,
2 S. Hussain and S. Garantziotis, Autophagy, 2013, 9, 101– 2007, 2, 430–441.
103. 29 A. E. Nel, L. Madler, D. Velegol, T. Xia, E. M. Hoek,
3 O. D. Velev and E. W. Kaler, Langmuir, 1999, 15, 3693–3698. P. Somasundaran, F. Klaessig, V. Castranova and
4 A. K. Boal, F. Ilhan, J. E. DeRouchey, T. Thurn-Albrecht, M. Thompson, Nat. Mater., 2009, 8, 543–557.
T. P. Russell and V. M. Rotello, Nature, 2000, 404, 746–748. 30 N. Lewinski, V. Colvin and R. Drezek, Small, 2008, 4,
5 A. T. Florence, J. Drug Targeting, 2004, 12, 65–70. 26–49.
6 T. Xia, M. Kovochich, M. Liong, J. I. Zink and A. E. Nel, ACS 31 C. M. Goodman, C. D. McCusker, T. Yilmaz and
Nano, 2008, 2, 85–96. V. M. Rotello, Bioconjugate Chem., 2004, 15, 897–900.
7 S. W. Lowe and A. W. Lin, Carcinogenesis, 2000, 21, 485– 32 F. Wang, M. G. Bexiga, S. Anguissola, P. Boya,
495. J. C. Simpson, A. Salvati and K. A. Dawson, Nanoscale,
8 M. I. Khan, A. Mohammad, G. Patil, S. A. Naqvi, 2013, 5, 10868–10876.
L. K. Chauhan and I. Ahmad, Biomaterials, 2012, 33, 1477– 33 A. Akinc, M. Thomas, A. M. Klibanov and R. Langer, J. Gene
1488. Med., 2005, 7, 657–663.
9 O. Zabirnyk, M. Yezhelyev and O. Seleverstov, Autophagy, 34 A. Asokan and M. J. Cho, J. Pharm. Sci., 2002, 91, 903–913.
2007, 3, 278–281. 35 S. Hong, P. R. Leroueil, E. K. Janus, J. L. Peters,
10 S. Hussain, F. Al-Nsour, A. B. Rice, J. Marshburn, M. M. Kober, M. T. Islam, B. G. Orr, J. R. Baker Jr. and
B. Yingling, Z. Ji, J. I. Zink, N. J. Walker and S. Garantziotis, M. M. Banaszak Holl, Bioconjugate Chem., 2006, 17, 728–
ACS Nano, 2012, 6, 5820–5829. 734.

This journal is © The Royal Society of Chemistry 2015 Nanoscale, 2015, 7, 736–746 | 745
View Article Online

Paper Nanoscale

36 P. Pino, B. Pelaz, Q. Zhang, P. Maffre, G. U. Nienhausbc 57 K. Degenhardt, R. Mathew, B. Beaudoin, K. Bray,


and W. J. Parak, Mater. Horiz., 2014, 1, 301–313. D. Anderson, G. Chen, C. Mukherjee, Y. Shi, C. Gelinas,
37 L. Treuel, S. Brandholt, P. Maffre, S. Wiegele, L. Shang and Y. Fan, D. A. Nelson, S. Jin and E. White, Cancer Cell, 2006,
G. U. Nienhaus, ACS Nano, 2014, 8, 503–513. 10, 51–64.
38 T. Cedervall, I. Lynch, S. Lindman, T. Berggard, E. Thulin, 58 D. Gozuacik and A. Kimchi, Oncogene, 2004, 23, 2891–2906.
H. Nilsson, K. A. Dawson and S. Linse, Proc. Natl. Acad. 59 O. Seleverstov, O. Zabirnyk, M. Zscharnack, L. Bulavina,
Published on 17 November 2014. Downloaded by University of California - Los Angeles on 18/08/2015 22:17:24.

Sci. U. S. A., 2007, 104, 2050–2055. M. Nowicki, J. M. Heinrich, M. Yezhelyev, F. Emmrich,


39 J. Leszczynski, Nat. Nanotechnol., 2010, 5, 633–634. R. O’Regan and A. Bader, Nano Lett., 2006, 6, 2826–
40 A. Kothawala, K. Kilpatrick, J. A. Novoa and L. Segatori, 2832.
PLoS One, 2012, 7, e43505. 60 Y. Zhang, C. Yu, G. Huang, C. Wang and L. Wen,
41 S. R. Panikkanvalappil, M. A. Mahmoud, M. A. Mackey and Int. J. Nanomed., 2010, 5, 601–609.
M. A. El-Sayed, ACS Nano, 2013, 7, 7524–7533. 61 S. Chen, S. K. Rehman, W. Zhang, A. Wen, L. Yao and
42 K. Ohnishi, E. Nakahata, K. Irie and A. Murakami, Biochem. J. Zhang, Biochim. Biophys. Acta, 2010, 1806, 220–229.
Biophys. Res. Commun., 2013, 430, 616–622. 62 D. G. Hardie, EMBO J., 2011, 30, 634–635.
43 W. X. Ding, H. M. Ni, W. Gao, T. Yoshimori, D. B. Stolz, 63 H. Takeuchi, Y. Kondo, K. Fujiwara, T. Kanzawa, H. Aoki,
D. Ron and X. M. Yin, Am. J. Pathol., 2007, 171, 513–524. G. B. Mills and S. Kondo, Cancer Res., 2005, 65, 3336–3346.
44 E. Ullman, Y. Fan, M. Stawowczyk, H. M. Chen, Z. Yue and 64 Q. Y. Zheng, F. S. Jin, C. Yao, T. Zhang, G. H. Zhang and
W. X. Zong, Cell Death Differ., 2008, 15, 422–425. X. Ai, Biochem. Biophys. Res. Commun., 2012, 419, 741–747.
45 M. Abdelrahim, K. Newman, K. Vanderlaag, I. Samudio 65 N. Shinojima, T. Yokoyama, Y. Kondo and S. Kondo, Auto-
and S. Safe, Carcinogenesis, 2006, 27, 717–728. phagy, 2007, 3, 635–637.
46 J. Wu and R. J. Kaufman, Cell Death Differ., 2006, 13, 374– 66 U. Ozcan, E. Yilmaz, L. Ozcan, M. Furuhashi,
384. E. Vaillancourt, R. O. Smith, C. Z. Gorgun and
47 Q. Zhang, W. Yang, N. Man, F. Zheng, Y. Shen, K. Sun, Y. Li G. S. Hotamisligil, Science, 2006, 313, 1137–1140.
and L. P. Wen, Autophagy, 2009, 5, 1107–1117. 67 P. Dromparis, R. Paulin, T. H. Stenson, A. Haromy,
48 S. T. Stern, B. S. Zolnik, C. B. McLeland, J. Clogston, G. Sutendra and E. D. Michelakis, Circulation, 2013, 127,
J. Zheng and S. E. McNeil, Toxicol. Sci., 2008, 106, 140–152. 115–125.
49 N. Mizushima and T. Yoshimori, Autophagy, 2007, 3, 542– 68 Q. Xie, V. I. Khaoustov, C. C. Chung, J. Sohn, B. Krishnan,
545. D. E. Lewis and B. Yoffe, Hepatology, 2002, 36, 592–
50 S. Wang, Y. Li, J. Fan, Z. Wang, X. Zeng, Y. Sun, P. Song and 601.
D. Ju, Biomaterials, 2014, 35, 7588–7597. 69 J. Zhang, M. W. Morris Jr., W. A. Dorsett-Martin,
51 H. M. Shen and P. Codogno, Autophagy, 2011, 7, 457–465. L. C. Drake and C. D. Anderson, J. Surg. Res., 2013, 183,
52 W. Zhang, Y. Ji, X. Wu and H. Xu, ACS Appl. Mater. Inter- 929–935.
faces, 2013, 5, 9856–9865. 70 M. Kars, L. Yang, M. F. Gregor, B. S. Mohammed,
53 C. Y. Tay, P. Cai, M. I. Setyawati, W. Fang, L. P. Tan, T. A. Pietka, B. N. Finck, B. W. Patterson, J. D. Horton,
C. H. Hong, X. Chen and D. T. Leong, Nano Lett., 2014, 14, B. Mittendorfer, G. S. Hotamisligil and S. Klein, Diabetes,
83–88. 2010, 59, 1899–1905.
54 Y. Qiu, Y. Liu, L. Wang, L. Xu, R. Bai, Y. Ji, X. Wu, Y. Zhao, 71 S. Turdi, N. Hu and J. Ren, PLoS One, 2013, 8, e63615.
Y. Li and C. Chen, Biomaterials, 2010, 31, 7606–7619. 72 Y. Fang, E. Studer, C. Mitchell, S. Grant, W. M. Pandak,
55 S. T. Stern, P. P. Adiseshaiah and R. M. Crist, Part. Fibre P. B. Hylemon and P. Dent, Mol. Pharmacol., 2007, 71,
Toxicol., 2012, 9, 20. 1122–1128.
56 E. A. Corcelle, P. Puustinen and M. Jaattela, FEBS J., 2009, 73 M. Hoyer-Hansen and M. Jaattela, Cell Death Differ., 2007,
276, 6084–6096. 14, 1576–1582.

746 | Nanoscale, 2015, 7, 736–746 This journal is © The Royal Society of Chemistry 2015

View publication stats

You might also like