Accepted Manuscript Titanium dioxide nanoparticles induce proteostasis disruption and autophagy in human trophoblast cells Yuqing Zhang, Bo Xu, Mengmeng Yao, Tianyu Dong, Zhilei Mao, Bo Hang, Xiumei Han, Zhongning Lin, Bian Qian, Min Li, Yankai Xia PII:
S0009-2797(18)30744-0
DOI:
10.1016/j.cbi.2018.09.015
Reference:
CBI 8416
To appear in:
Chemico-Biological Interactions
Received Date: 7 June 2018 Revised Date:
20 July 2018
Accepted Date: 24 September 2018
Please cite this article as: Y. Zhang, B. Xu, M. Yao, T. Dong, Z. Mao, B. Hang, X. Han, Z. Lin, B. Qian, M. Li, Y. Xia, Titanium dioxide nanoparticles induce proteostasis disruption and autophagy in human trophoblast cells, Chemico-Biological Interactions (2018), doi: https://doi.org/10.1016/j.cbi.2018.09.015. This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
ACCEPTED MANUSCRIPT Titanium dioxide nanoparticles induce proteostasis disruption and autophagy in
2
human trophoblast cells
3
Yuqing Zhanga,b1, Bo Xua,b1, Mengmeng Yaoc1, Tianyu Donga,b, Zhilei Maod, Bo
4
Hange, Xiumei Hana,b, Zhongning Lin f, Bian Qiang, Min Lih*,Yankai Xiaa,b*
5
a
6
Public Health, Nanjing Medical University, Nanjing, 211166, China.
7
b
8
Health, Nanjing Medical University, Nanjing, 211166, China.
9
c
RI PT
1
SC
State Key Laboratory of Reproductive Medicine, Institute of Toxicology, School of
M AN U
Key Laboratory of Modern Toxicology of Ministry of Education, School of Public
Healthcare Management, International Business Center of Nanjing University,
10
Nanjing, 211166, China.
11
d
12
Medical University, Changzhou, 213003, China.
13
e
14
Laboratory, Berkeley, CA 94720, USA.
15
f
16
of Public Health, Xiamen University, Xiamen, P. R. China
17
g
18
Disease Control and Prevention, Nanjing, 210009, China
19
h
TE D
The Affiliated Changzhou Maternity and Child Health Care Hospital, Nanjing
EP
Biological Systems and Engineering Division, Lawrence Berkeley National
AC C
State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School
Department of Toxicology and Function Assessment, Jiangsu Provincial Center for
Department of Anatomy, Nanjing Medical University, Nanjing, 211166, China.
20 21
1
22
* To whom correspondence should be addressed:
These authors contributed equally to this work.
ACCEPTED MANUSCRIPT Dr. Yankai Xia and Dr. Min Li.
24
State Key Laboratory of Reproductive Medicine, Institute of Toxicology, School of
25
Public Health, Nanjing Medical University, No.101 Longmian Road, Nanjing, 211166,
26
China.
27
Phone: +86-25-86868425; E-mail:
[email protected]
28
Department of Anatomy, Nanjing Medical University, Nanjing, 211166, China.
29
Phone: +86-25-86869106; E-mail:
[email protected]
SC
M AN U
30 31 32 33
EP AC C
36
TE D
34 35
RI PT
23
ACCEPTED MANUSCRIPT Abstract
38
Titanium dioxide nanoparticles (TiO2 NPs) exist in many nano-products and concerns
39
have been raised about their potential toxicity on human beings. One such issue is
40
their potential effects on placental function, and the studies on this topic are limited
41
and the mechanism remains unclear. Here we employed human trophoblast
42
HTR-8/SVneo cells to investigate the effects of TiO2 NPs on trophoblast. Results
43
showed that TiO2 NPs could enter cells and were mostly distributed in lysosomes,
44
with some in the cytoplasm. TiO2 NPs and protein aggregation were found in both
45
fetal bovine serum (FBS) in culture medium and cytoplasm of HTR-8/SVneo cells. In
46
consistence with that, proteostasis of HTR-8/SVneo cells was significantly disrupted
47
and endoplasmic reticulum (ER) stress related markers including PERK, IRE1-α were
48
increased. After high speed centrifugation, the proteins PERK and IRE1-α were
49
dramatically decreased in the highest TiO2 NPs treatment group, which indicated
50
interactions between TiO2 NPs and these two proteins. Meanwhile, the protein
51
expressions of LC3-II/LC3-I and P62, the autophagy biomarkers, were increased and
52
the autophagy flux was not blocked. Cellular ROS stress increased and mitophagy
53
related genes including PINK and Parkin increased along with the increased
54
co-localization of LC3 and mitochondria. Taken together, these results indicated that
55
TiO2 NPs interacted with intracellular proteins and activated ER stress and mitophagy
56
in HTR-8/SVneo cells, which might do damage to placental function.
57 58
AC C
EP
TE D
M AN U
SC
RI PT
37
ACCEPTED MANUSCRIPT 59
Keywords:
60
Titanium dioxide nanoparticles (TiO2 NPs); Trophoblast; Proteostasis; Autophagy;
61
Mitophagy
RI PT
62
AC C
EP
TE D
M AN U
SC
63
ACCEPTED MANUSCRIPT 1. Introduction
65
Titanium dioxide nanoparticles (TiO2 NPs) are one type of mostly used nanoparticles,
66
with foods, consumer products and household products being the most important
67
sources of exposure [1, 2]. Previous research has shown that titanium materials are in
68
many personal care products such as sunscreens and toothpastes by weight of 1
69
to >10%, at least 36% of which are in the form of nanoparticles [1]. Due to their small
70
size and biological activity, TiO2 NPs can get into human body and pass through
71
biological barriers [3, 4].
SC
M AN U
72
RI PT
64
Previous studies have confirmed that TiO2 NPs could pass through placental barrier and do damage to the developing fetuses [5, 6]. Studies have shown that
74
prenatal exposure to TiO2 NPs could impair lung development, decrease neurogenesis
75
and damage memory of offspring [7-9]. Exposure to TiO2 NPs has adverse effects on
76
mothers as well. Maternal exposure to TiO2 NPs could induce cytotoxicity in human
77
amniotic fluid-derived cells as well as structural and functional abnormalities in the
78
placenta on the maternal side [6, 10]. However, how TiO2 NPs impact placenta and
79
the exact mechanisms of inducing placental dysfunction are not clear.
EP
AC C
80
TE D
73
Efforts have been made to explore the effects of nanomaterials on organisms.
81
Interaction of nanoparticles with proteins in cell uptake and drug delivery [11-13] is
82
one of the research foci. However, almost all the studies have focused on the
83
nano-protein interactions in blood circulation and extracellular environment [11, 14].
84
The effect of nanoparticles on proteins inside cells is rarely studied so far [15, 16].
85
Trophoblast is a crucial tissue of highly active protein synthesis and secretion [17].
ACCEPTED MANUSCRIPT TiO2 NPs, which can enter trophoblast cells, have the ability of adsorbing proteins
87
and forming “protein corona” on particle surface [18-21] and then might induce
88
trophoblast dysfunction. In this study, we employed HTR-8/SVneo cells as a
89
representative trophoblast cell model to investigate nanoparticle-protein interactions
90
inside these cells and the potential mechanisms to induce trophoblast dysfunction
91
upon exposure to TiO2 NPs.
RI PT
86
SC
92
2. Materials and methods
94
2.1. Characterization of TiO2 NPs
95
TiO2 NPs (Sigma, USA) were suspended in either the same medium applied to cell
96
culture, which was Roswell Park Memorial Institute (RPMI) -1640 medium (Gibco,
97
USA) supplemented with 1% penicillin/streptomycin and 10% fetal bovine serum
98
(FBS) (Gibco, USA), or in distilled and deionized water. Before detection,
99
suspensions were sonicated in ice water for 30 min (100 W). The morphology was
100
determined using transmission electron microscope (TEM, JEOL JEM 2011, Japan).
101
Hydrodynamic diameter and surface zeta potential of TiO2 NPs were measured on
102
Malvern Zetasizer Nano-ZS90 (Malvern, UK).
103
2.2. Cell culture
104
The trophoblast HTR-8/SVneo cell line was purchased from American Type Culture
105
Collection (ATCC, Mamassas VA, USA) and incubated in complete RPMI-1640
106
medium at 37oC, 5% CO2. After ultraviolet sterilization for 30 min, TiO2 NPs were
107
suspended with complete RPMI-1640 medium as stock solution at the concentration
AC C
EP
TE D
M AN U
93
ACCEPTED MANUSCRIPT of 1 mg/ml and sonicated as described above. The suspensions were diluted to
109
different concentrations with complete culture medium (1, 10 and 100 µg/ml) before
110
applied to cell culture. Another group without TiO2 NPs treatment was served as
111
control group. 200 nM Rapamycin (Beyotime, China) or 10 µM chloroquine (Sigma,
112
USA) was added to the culture medium with TiO2 NPs when detecting the autophagy
113
flow.
114
2.3. Cell viability
115
HTR-8/SVneo cells were seeded 1.5x104 per well one day prior to the experiment in
116
96-multiwell plates and cultured with 100 µl TiO2 NPs suspensions at the
117
concentrations of 1, 10 and 100 µg/ml respectively. The results were compared with
118
control group having no TiO2 NPs. After treatment for 24 h or 48 h, we used Cell
119
Counting Kit 8 (CCK-8) (Dojindo, Japan) to determine cell viability following the
120
reference manual. In brief, after incubated with CCK-8-solution for 1 h at 37oC, 100
121
µl supernatant of was transferred to another 96-multiwell plate and absorbance was
122
detected at 450 nm on UV/vis spectrometer (Ocean Optics, HR4000). This
123
experiment was performed three times independently.
124
2.4. Cellular uptake of TiO2 NPs
125
After incubation with TiO2 NPs for 24 h, samples were pretreated as described before
126
[22]. Briefly, through fixation, post-fixation and a series of dehydration,
127
HTR-8/SVneo cells were embedded in Araldite and stained with lead citrate and
128
uranyl acetate before TEM examination.
129
AC C
EP
TE D
M AN U
SC
RI PT
108
HTR-8/SVneo cells were seeded in plates (Corning Costar, USA) with TiO2 NPs
ACCEPTED MANUSCRIPT at different concentrations. After overnight incubation, cell morphology was
131
determined using a light microscope (ECLIPSE, TS100, Japan).
132
2.5. Nano-protein negative staining assay
133
TiO2 NPs were suspended with complete RPMI-1640 culture medium and sonicated
134
in ice water for 30 min (100 W). Afterwards the suspension was negatively stained
135
with phosphotungstic acid (1% in PBS) and then examined using TEM (JEOL JEM
136
2011, Japan).
SC
RI PT
130
After treatment with TiO2 NPs for 24 h, HTR-8/SVneo cells were washed with
138
PBS to remove nanoparticles on the surface as far as possible to decrease potential
139
disturbance from the extracellular nanoparticles. Cells were harvested and lysed for
140
30 min on ice. Then cell lysate was differential-speed centrifuged with 1000 rpm for
141
10 min to remove cell debris then the supernatant was centrifuged at 12000 rpm for
142
30 min to collect nano-protein composite. Deposition was re-suspended in PBS and
143
negatively stained with phosphotungstic acid (1% in PBS) and examined using TEM.
144
2.6. Protein preparation and Western blotting assay
145
After incubated with different concentrations of TiO2 NPs for 24 h, HTR-8/SVneo
146
cells were lysed for 30 min in RIPA buffer (Beyotime, China) with 1%
147
phenylmethanesulfonyl fluoride (PMSF) (Beyotime, China) as protease inhibitor.
148
Lysates were centrifugalized at 1,000 rpm for 10 min at 4oC to collect supernatant.
149
BCA Kit (Beyotime, China) was used to quantitatively analyze protein concentrations.
150
80 µg of total protein of each group was electrophoresed in sodium dodecyl sulfate
151
(SDS)-polyacrylamide gel electrophoresis (PAGE) gel and blotted in polyvinylidene
AC C
EP
TE D
M AN U
137
ACCEPTED MANUSCRIPT fluoride (PVDF) membrane (Bio-Rad, USA). Membranes were blocked with 5% BSA
153
(Sigma, USA) buffer for 1 h and respectively incubated with the following primary
154
antibodies overnight at 4oC: rabbit anti-LC3 (1:1000), rabbit anti-PERK (1:1000),
155
rabbit anti-phospho PERK (1:1000), rabbit anti-IRE1-α (1:1000), rabbit
156
anti-SAPK/JNK (1:1000) and mouse anti-phospho SAPK/JNK (1:1000)(CST, USA),
157
rabbit anti-P62 (Abcam, USA, 1:1000), mouse anti- GAPDH (Beyotime, China,
158
1:1000). PVDF membranes were washed with TBST buffer (20 mM tris (pH7.6), 137
159
mM NaCl, and 0.1% Tween 20) for 1 h, and then incubated with secondary antibodies
160
including horseradish peroxidase (HRP)-conjugated goat anti-rabbit secondary
161
antibody (Beyotime, China, 1:2000) and HRP-conjugated goat anti-mouse secondary
162
antibody (Beyotime, China, 1:2000). Chemiluminescence was developed by the ECL
163
Western blot detection kit (Millipore, USA) and bands were scanned in a Bio-Rad
164
Imaging System. These blots were repeated three times independently.
165
2.7. Quantitative real-time PCR assay
166
Total RNAs were isolated with TRIZOL regent (Invitrogen, Carlsbad, CA) and the
167
concentration was assessed with Nanodrop 2000 (Thermo Fisher Scientific, USA).
168
Reverse transcription was carried out according to instructions of PrimerScript RT
169
reagent Kit (TaKaRa, Japan). The products were stored at -200C until used. The
170
reaction system was conducted following the instruction of SYBR Green mix kit
171
(TaKaRa, Japan). Real-time PCR was performed on ABI7900 Fast Real-Time System
172
(Applied Biosystems, USA) and the housekeeping gene gapdh was used as the
173
internal reference. Expression levels of mRNAs were calculated using 2^-∆Ct
AC C
EP
TE D
M AN U
SC
RI PT
152
ACCEPTED MANUSCRIPT (threshold cycle) method. All the primers were synthesized by Invitrogen (Invitrogen,
175
Shanghai) and the detailed information was shown in Table S1. At least three
176
separate experiments were carried out to assess expression levels of mRNAs.
177
2.8. Immunofluorescence analysis
178
After treated with TiO2 NPs on Confocal Dish (Cellvis, China) for 24 or 48 h, culture
179
medium was replaced with fresh one containing 100 nM MitoTracker® Red CMXRos
180
(Invitrogen, USA) to stain mitochondria for 30 min at 37oC. Cells were fixed with 4%
181
paraformaldehyde for 30 min at room temperature. Following membrane permeation
182
with 0.1% Triton X-100 (Beyotime, China) and blocking with 1% BSA, cells were
183
incubated overnight at 4oC with rabbit anti-LC3 antibodies (CST, USA, 1:200) and
184
followed by fluorescein isothiocyanate (FITC)-conjugated goat anti-rabbit secondary
185
antibody (Beyotime, China, 1:200) for 4 h at room temperature. The nuclei were
186
stained with 4', 6-diamidino-2-phenylindole (DAPI) (Beyotime, China, 1:1000) for 5
187
min. Immunofluorescence photos were obtained by confocal microscope (Nikon,
188
Japan).
189
2.9. Derivative-quenched bovine serum albumin (DQ-Red BSA) assay
190
DQ-Red BSA (Molecular Probes, USA) is a self-quenched lysosome degradation
191
biomarker used to monitor enzyme activity of lysosomes. Cells were seeded onto
192
Confocal Dish (Cellvis, China) and incubated with TiO2 NPs for 24 h. Then cells were
193
further incubated with fresh RPMI-1640 culture medium containing 10 mg/mL
194
DQ-Red BSA for 3 h at 37oC. Then nuclei were stained with Hoechst (Beyotime,
195
China) and fluorescence was visualized by confocal microscopy (Nikon, Japan).
AC C
EP
TE D
M AN U
SC
RI PT
174
ACCEPTED MANUSCRIPT 2.10. Reactive oxygen species (ROS) assay
197
HTR cells were exposed to different concentrations of TiO2 NPs or control medium
198
for 24 h. Cells were collected with trypsin (Beyotime, China) and incubated with
199
fresh culture medium containing 5 µM dichlorofluorescin diacetate (DCFH-DA) for
200
30 min at 37oC. Afterwards, cellular fluorescence intensity was measured by FACS
201
Calibur Flow Cytometry (BD Biosciences, USA) immediately. This assay was carried
202
out three times separately.
203
2.11. Statistical analysis
204
Results were expressed as mean ± standard error (S.E.) from at least three separate
205
experiments for all analyses. Statistical significance was defined as two-tailed p<0.05.
206
Comparisons between groups were conducted by ANOVA. Further comparison
207
between control group and different treated groups were performed by the Dunnett’s
208
multiple comparison test.
TE D
M AN U
SC
RI PT
196
209
3.
211
3.1. Characteristics of TiO2 NPs
212
The characteristics of TiO2 NPs were represented in Fig. 1. TEM images
213
demonstrated that TiO2 NPs suspended in distilled and deionized water were nearly
214
spherical, with the average diameter of 36.90 nm. The particle size distribution
215
showed that TiO2 NPs slightly aggregated in complete culture medium and water,
216
with hydrodynamic diameters of 73.29±5.75 nm and 65.48±1.63 nm, respectively.
217
Zeta potential of TiO2 NPs in water was -8.59±0.37 mV, while in complete culture
AC C
Results
EP
210
ACCEPTED MANUSCRIPT medium was -9.56±0.92 mV.
SC
RI PT
218
M AN U
219 220
Fig. 1. (A) TEM image of TiO2 NPs suspended in distilled and deionized water.
221
Bar=100 nm. (B) Size distribution of TiO2 NPs in RPMI-1640 complete culture
222
medium. (C) Characteristics of TiO2 NPs in water or complete culture medium.
TE D
223
3.2. Effects of TiO2 NPs on cell viability and morphology
225
Treatment with TiO2 NPs did not change cell viability even at a relatively high
226
concentration of 100 µg/ml at both 24 h (Fig. 2A) and 48 h (Fig. 2B). The following
227
analysis of this study were all conducted after treatment for 24 h. The treated cells
228
colored yellowish-brown because of uptake of non-transparent nanoparticles. In the
229
same time, some vacuoles were formed inside the exposed cells (Fig. 2C).
AC C
230
EP
224
Under TEM, we found TiO2 NPs inside the cells, and most of them were
231
distributed in lysosomes, with some in the cytoplasm (Fig. 2D). We also observed that
232
the autophagosomes were increased in the TiO2 NPs-treated group, which indicates
233
autophagy activation (Fig. 2D).
SC
RI PT
ACCEPTED MANUSCRIPT
234
Fig. 2. (A)Viability of HTR-8/SVneo cells treated with various concentrations of TiO2
236
NPs for 24 h. (B) Viability of HTR-8/SVneo cells treated with various concentrations
237
of TiO2 NPs for 48 h. Results of A and B were expressed as mean ± standard error
238
(S.E.) from three separate experiments. (C) Light microscope images with 100x
239
magnification of control cells and TiO2 NPs treated cells (100 µg/ml) after treating for
240
24 h. (D) TEM images of control and nanoparticle-treated groups after 24 h treatment.
241
TiO2 NPs were mostly distributed in the lysosome, with some in the cytoplasm.
242
Autophagosomes were indicated by red arrows.
TE D
EP
AC C
243
M AN U
235
244
3.3. Nano-protein aggregation outside and inside HTR-8/SVneo cells after 24 h
245
exposure
246
To determine if TiO2 NPs adsorb proteins in culture medium, using TEM, we found
247
protein corona around nanoparticles suspended in complete culture medium (Fig. 3A),
248
while no protein corona was found around the ones in distilled and deionized water
249
(Fig. 1A). We further negatively stained nanoparticles suspended in complete culture
ACCEPTED MANUSCRIPT 250
medium and found nano-protein aggregation (Fig. 3B). It is known that nanoparticles enter cells by endocytosis [23], and that enzymes
252
in the endosome degrade them [24]. DQ-Red BSA is a fluorogenic substrate for
253
proteases and bright red fluorescent fragments will be released after proteolysis in the
254
lysosome. As TEM images showed, the brightness of red fluorescent fragments was
255
weakened after TiO2 NPs treatment (Fig. 3C), indicating that the lysosome protease
256
activity was decreased. This also suggests the structural disruption of lysosomes to
257
certain extent and the release of nanoparticles in the cytoplasm.
SC
We used phosphotungstic acid to negatively stain TiO2 NPs in cell lysis precipitate after differential-speed centrifugation and found nano-protein aggregation (Fig. 3D).
AC C
EP
TE D
259
M AN U
258
RI PT
251
260 261
Fig. 3. (A) TEM image of TiO2 NPs suspended in complete culture medium. Bar=500
262
nm. (B) Phosphotungstic acid-stained TEM image of nano-protein aggregation in
ACCEPTED MANUSCRIPT 263
complete culture medium. (C) DQ-Red BSA (bright red fluorescent fragments) was
264
weakened after treated with TiO2 NPs (100 µg/ml) for 24 h, which indicated
265
decreased proteolytic capacity in treated group. Bar=20 µm. (D) Phosphotungstic
266
acid-stained
267
centrifugation.
precipitant after differential-speed
268
RI PT
TEM image of cell lysis
3.4. TiO2 NPs exposure impairs proteostasis after 24 h
270
Since TiO2 NPs could disrupt the structure of lysosomes and adsorb proteins in the
271
cytoplasm, the protein homeostasis could also be disrupted after TiO2 NPs uptaken by
272
the cells. We investigated whether there was endoplasmic reticulum stress (ER stress)
273
after TiO2 NPs treatment, which is a consequence of proteostasis impairment. We
274
measured mRNA expression of ER stress related-sensors, inositol-requiring enzyme
275
1–alpha (IRE1-α) and protein kinase (PKR)-like ER kinase (PERK), and found that
276
they were increased as the concentrations of TiO2 NPs increased (Fig. 4A). After
277
treatment with different concentrations of TiO2 NPs, we collected and lysed cells to
278
detect ER stress-related proteins PERK, IRE1-α and c-Jun N-terminal kinase (JNK).
279
The results showed increased expression of several sensor proteins as shown in Fig.
280
4B, indicating proteostasis impairment. To explore whether TiO2 NPs could absorb
281
these proteins, we separated cell lysate with high-speed centrifugation and found the
282
levels of PERK, IRE1-α were dramatically decreased at the highest concentration
283
group while the reference protein GAPDH remained unchanged (Fig. 4C).
AC C
EP
TE D
M AN U
SC
269
SC
RI PT
ACCEPTED MANUSCRIPT
M AN U
284
Fig. 4. (A) ER stress-related sensors (IRE1-α and PERK) were detected by RT-PCR
286
using gapdh as internal control. Values were expressed as mean ± standard error (S.E.)
287
from three separate experiments. (B) Cell lysis was separated with low-speed
288
centrifugation to obtain total protein. Protein expressions of stress-activated protein
289
kinase (JNK) and ER stress-related proteins (PERK, IRE1-α) were detected by
290
Western blot with GAPDH as internal control. (C) Two ER stress-related proteins
291
(PERK, IRE1-α) were detected with Western blot after high-speed centrifugation,
292
which showed dramatically decreased protein levels in the highest treatment group.
293
Asterisk indicates significant difference when the values were compared to that of the
294
control (*p < 0.05,**p<0.01, ***p<0.001).
AC C
EP
TE D
285
295 296
3.5. TiO2 NPs treatment for 24 h induces autophagy
297
Autophagy is known to be a pro-survival pathway activated by certain stressors [25].
298
As shown in Fig. 2C, our TEM images indicated that the number of autophagosomes
ACCEPTED MANUSCRIPT was increased after TiO2 NPs treatment. We then further explored expression of key
300
components of autophagy. The expression of Beclin 1 and P62 were increased after
301
TiO2 NPs treatment (Fig. 5A). After treating HTR-8/SVneo cells with different
302
concentrations of TiO2 NPs for 24 h, the protein expressions of LC3-II/LC3-I and P62
303
were increased significantly (Fig. 5B). Next, rapamycin (MTORC1 inhibitor to
304
stimulate autophagy) and chloroquine (lysosomal fusion inhibitor to block autophagy)
305
were added to culture medium with 100 µg/ml of TiO2 NPs to test whether the
306
autophagic flux was altered. We found that the LC3-II/LC3-I protein ratios were all
307
increased compared to the controls in TiO2 NP-treated groups, indicating that the
308
autophagic flux was not blocked and autophagosomes were increased after TiO2 NPs
309
treatment (Fig. 5C).
310 311
AC C
EP
TE D
M AN U
SC
RI PT
299
Fig. 5. (A) mRNA expressions of two autophagy-related genes (Beclin1 and 312
SQSTM1/P62) were detected by RT-PCR after treated with TiO2 NPs for 24 h. Values 313
were expressed as mean ± standard error (S.E.) from three separate experiments. (B) 314
Western blot analysis of the expression of microtubule-associated light chain 3 (LC3)
ACCEPTED MANUSCRIPT 315
and Ubiquitin-Binding Protein P62 of different concentration groups with GAPDH as 316
loading control. (C) Western blot analysis of autophagic flux activation with 317
rapamycin (a MTORC1 inhibitor to stimulate autophagy) and chloroquine (a 318 319
RI PT
lysosomal fusion inhibitor) added to culture medium with TiO2 NPs. Asterisk indicates significant difference when the values were compared to that of the control 320
(*p < 0.05,**p<0.01, ***p<0.001).
SC
321
3.6. Clearance of damaged mitochondria after TiO2 NPs treatment for 24 h
323
Proteostasis disruption may disturb the function of mitochondria. When mitochondria
324
are impaired, PTEN induced putative kinase 1 (PINK1) accumulates in mitochondria,
325
which recruits and activates proteins such as Parkin (PARK2). The latter is an
326
ubiquitin ligase for the degradation of damaged mitochondria [26, 27]. We firstly
327
investigated the oxidative stress the cells may undertake and found that the levels
328
were higher as the concentrations of TiO2 NPs increased (Fig. 6A). The mRNA levels
329
of PINK1 and Parkin were increased after TiO2 NPs treatment as well (Fig. 6B). We
330
then used immunofluorescence and confocal microscope to confirm the
331
co-localization of autophagosomes and mitochondria. After treating HTR cells with
332
TiO2 NPs for 24 h, we found the activation of autophagy was increased and
333
membrane potential of mitochondria was decreased (Fig. 6C). The overlap of these
334
two kinds of fluorescent fragments was reduced compared to the control group (Fig.
335
6C), suggesting that the damaged mitochondria were cleared up by the process of
336
autophagy.
AC C
EP
TE D
M AN U
322
M AN U
SC
RI PT
ACCEPTED MANUSCRIPT
337 338
Fig. 6. (A) Intracellular reactive oxygen species (ROS) formation measured by FCM. 339
TE D
(B) Mitophagy-related genes (PINK1 and Parkin) were detected with RT-PCR. Both 340
results in A and B were expressed as mean ± standard error (S.E.) from three separate 341
experiments. (C) Confocal microscopy to show co-localization of LC3 and 342
EP
mitochondria in HTR cells. LC3 was stained green and mitochondria were stained red. 343 344
AC C
Asterisk indicates significant difference when the values were compared to that of the control (*p < 0.05,**p<0.01, ***p<0.001). 345 346
4.
Discussion
347
In this study, we explored the potential interaction between TiO2 NPs and intracellular
348
proteins as well as the possible mechanisms on trophoblast cells. We treated HTR
349
cells at a relative high concentration of TiO2 NPs at which concentration cell viability
ACCEPTED MANUSCRIPT was not decreased. We demonstrated that TiO2 NPs could enter HTR cells and mostly
351
distributed in the lysosomes. The lysosomal degradation function was impaired,
352
leading to some of the TiO2 NPs leaking into the cytoplasm and adsorbing the proteins
353
in the cytoplasm, thus leading to proteostasis disturbance and ER stress. In the
354
meantime, the mitochondria were impaired, leading to autophagy towards the
355
damaged mitochondria.
Increasing evidence has shown that engineered nanoparticles could pass through
SC
356
RI PT
350
placental barrier and damage growing fetuses [28-30]. Although lacking of human
358
studies, TiO2 NPs has been reported to penetrate placenta and present in placental
359
trophoblast and fetus in rodent models [6, 8, 31]. TiO2 NPs used in our study was
360
spherical with hydrodynamic diameter of 73.29±5.75 nm, smaller than the
361
nanoparticles mentioned in above studies, which suggests trophoblast exposure during
362
pregnancy. In our study, cell viability after exposure for 24 h was not changed at the
363
relative high concentration. Pervious studies have demonstrated nanoparticle toxicity
364
without causing cell viability decrease [32, 33].
EP
TE D
M AN U
357
In our study, nano-protein aggregation was found when particles were suspended
366
in complete culture medium, which was in agreement with a previous study [34]. TiO2
367
NPs are not the only nanoparticles exhibiting the ability of protein absorption.
368
Nanoparticles like SiO2, carbon black, CdS and CeO have been reported to absorb
369
proteins in culture medium or plasm [35-38]. In a study comparing protein absorption
370
by several types of materials, TiO2, CeO2 and ZnO showed strong abilities [34]. The
371
potential mechanism of strong protein absorption ability of TiO2 NPs might due to
AC C
365
ACCEPTED MANUSCRIPT 372
negative charges on surface [14] and positive charge on proteins. All of these could
373
lead to nano-protein aggregation [18-21]. Nanoparticle-protein interaction might persist during all the process of
375
bio-distribution. After taken by cells, whether there is direct interaction between
376
nanoparticles and intracellular proteins is not clear. Nanoparticles often get into cells
377
by endocytosis. Endosomes containing the ingested materials then fuse with
378
lysosomes and form lysosome-endosome hybrids, which are rich of enzymes [39].
379
Acid condition and enzyme in lysosomes could degrade nanoparticles and protein
380
aggregation formed by FBS[24], whereas nanoparticle could interact with enzymes
381
inside lysosomes and lead to dysfunction. In our study, we found decreased
382
proteolytic capacity and structural disruption of lysosomes after TiO2 NPs treatment,
383
which phenomenon has been reported present in some other nanoparticles as well [22,
384
40, 41]. As the TEM images showed in this study, some of the nanoparticles were
385
present in the cytoplasm where is full of proteins. The destiny of nanoparticles
386
escaped to cytoplasm is not well studied. A previous study found that SiO2
387
nanoparticles could enter HEp-2 cells and aggregate with proteins in cell nuclei,
388
which impairs nuclear function [15]. By contrast, TiO2 NPs in our study did not
389
appear in cell nuclei. Instead, they were present in cytoplasm after escaped from
390
lysosomes and nano-protein aggregation was still existent. However, the composition
391
of aggregated proteins was not clear, which might include proteins in FBS, enzymes
392
in lysosomes as well as proteins in cytoplasm.
AC C
EP
TE D
M AN U
SC
RI PT
374
ACCEPTED MANUSCRIPT 393
To make out if TiO2 NPs absorbed proteins in cytoplasm and led to proteostasis disruption. We assessed the expression of ER stress related markers. Soluble secretory
395
and transmembrane proteins are synthesized in membrane-bound ribosomes in the
396
form of peptides and then folded in the endoplasmic reticulum (ER) and modified in
397
Golgi [42]. Protein folding in ER is very sensitive to changes in the intracellular and
398
extracellular environments [43], especially in cells like trophoblast cells with a high
399
rate of protein synthesis. Many studies have established that nanoparticle exposure
400
could lead to ER stress [33, 44, 45]. In this study, the expression levels of ER
401
stress-related markers increased after TiO2 NPs treatment, which indicated
402
proteostasis disruption inside cells. The potential possibility might be absorption of
403
endoplasmic reticulum related proteins onto TiO2 NPs, which disturbed the function
404
of endoplasmic reticulum. One of our previous studies has shown that TiO2 NPs could
405
absorb Tau protein inside the HY5Y cells [22]. In this study, we found that the
406
expression of IRE1-α and PERK showed a dramatic decrease in the HTR cells treated
407
with highest concentration of TiO2 NPs. These two proteins are highly expressed in
408
ER, where plenty of peptides are folded. TiO2 NPs might adhere to the surface of ER
409
and absorb these two proteins. Proteins absorbed on TiO2 NPs maybe not specific and
410
may lead to various disruptions inside cells. Although some of the protein
411
identifications were determined, many of them are not.
AC C
EP
TE D
M AN U
SC
RI PT
394
412
At the early stage of TiO2 NPs exposure, some pre-survival reaction might be
413
activated to conserve energy and maintain cellular homeostasis, of which autophagy is
414
an important one [46-48]. In autophagy process, Beclin1 encodes a protein mediating
ACCEPTED MANUSCRIPT vesicle-trafficking processes and regulating autophagy [49]. SQSTM1/P62 binds
416
directly to LC3 to facilitate degradation of ubiquitinated proteins aggregated by
417
autophagy [50]. LC3 is a widely used biomarker of autophagy and the ratio of
418
LC3-II/LC3-I corresponds to the amount of autophagosomes [51]. In our study, gene
419
expression of Beclin1 and P62 were increased as well as ratio of LC3-II/LC3-I protein
420
expression increased after TiO2 NPs treatment. The accumulation of autophagosomes
421
may be due to autophagy activation or blockage of autophagy flux [52, 53].
422
Rapamycin (a MTORC1 inhibitor to stimulate autophagy) and chloroquine (a
423
lysosomal fusion inhibitor) are commonly used to detect whether there is blockage in
424
autophagy flux [51]. Our results showed that the ratio of LC3-II/LC3-I was both
425
increased after adding rapamycin or chloroquine to culture medium with TiO2 NPs
426
when compared to the control, indicating that there was no autophagic flux blockage
427
after TiO2 NPs treatment. Previous studies have indicated that autophagy in
428
trophoblast might lead to miscarriage, preeclampsia, intrauterine growth restriction
429
(IUGR) and other disorders in both mothers and fetuses [54-56]. Although cell
430
viability was not changed by comparing with the control group in our study,
431
autophagy might induce cell dysfunction after long time treatment.
SC
M AN U
TE D
EP
AC C
432
RI PT
415
Most of the proteins in mitochondria are synthesized in the free ribosomes in
433
cytoplasm and imported through the double membrane of the mitochondria in their
434
unfolded state [57]. Inadequate protein transportation into mitochondria may lead to
435
mitochondrial dysfunction. In addition, studies have shown that ions or molecules
436
released after lysosomal degradation of nanoparticles could damage mitochondria and
ACCEPTED MANUSCRIPT induce high levels of ROS [24]. In our study, TiO2 NPs in the cell might absorb
438
proteins critical to mitochondrial function and cause molecules to be released from
439
lysosomes, either of which could damage the function of mitochondria. In the current
440
study, ROS levels were increased in the treated groups and the increased level of ROS
441
might in turn damage mitochondria and generate more ROS by the defective ones[58].
442
Besides, mitochondrial membrane potential was decreased in the treatment group,
443
which suggested imbalance between mitochondrial outer membrane and inter
444
membrane [59]. All these suggests the damage of mitochondria. However, the exact
445
mechanism of mitochondrial damage was not clear in our study. It has been reported
446
that, under stress, mitophagy is initiated after accumulation of PINK and
447
phosphorylated Parkin on the outer mitochondrial membranes [60]. The expression
448
levels of PINK1 and Parkin were indeed increased in our study, suggesting that there
449
was a mitophagy activation. In addition, immunofluorescence and confocal
450
microscope analysis confirmed that mitochondria were damaged and autophagy was
451
activated to clean up the damaged ones.
EP
TE D
M AN U
SC
RI PT
437
In conclusion, we revealed that exposure to TiO2 NPs could disrupt the structure
453
of lysosomes and led to the leakage of these particles into the cytoplasm. As a result,
454
they absorbed cellular proteins in the cytoplasm which may account for the
455
proteostasis disruption. In addition, we observed that, mitochondria were damaged
456
and autophagy was activated upon exposure to TiO2 NPs. These findings provide
457
insight into the toxicological mechanisms of TiO2 NPs effects on human health. A
458
pictorial presentation of the potential mechanisms is outlined in Fig. 7. Certainly more
AC C
452
ACCEPTED MANUSCRIPT studies are warranted to further identify which proteins may interact with TiO2 NPs
460
and the more mechanisms of cellular dysfunction induced by these nanoparticles.
M AN U
SC
RI PT
459
461
Fig. 7. Potential mechanisms of proteostasis disruption and autophagy in human
463
trophoblast cells caused by TiO2 NPs treatment. TiO2 NPs and proteins in FBS are
464
aggregated in culture medium. Then particles are uptaken by endocytosis. Endosomes
465
containing the ingested materials fuse with lysosomes and form lysosome-endosome
466
hybrids. The lysosomal degradation function is impaired, leading to some of the TiO2
467
NPs leaking into the cytoplasm and adsorbing the proteins in the cytoplasm, thus
468
leading to ER stress. Ions or molecules released after lysosomal disruption damage
469
mitochondria. Proteostasis disturbance inside cells might lead to mitochondrial
470
damage as well. All of these activate mitophagy to clear the impaired mitochondria.
472
EP
AC C
471
TE D
462
473 474
References
475 476 477
[1]
A. Weir, P. Westerhoff, L. Fabricius, K. Hristovski, N. von Goetz, Titanium dioxide nanoparticles in food and personal care products, Environ Sci Technol 46 (2012) 2242-2250.
ACCEPTED MANUSCRIPT
[6]
[7]
[8]
[9]
RI PT
SC
[5]
M AN U
[4]
TE D
[3]
L. Sang, Y. Zhao, C. Burda, TiO2 nanoparticles as functional building blocks, Chem Rev 114 (2014) 9283-9318. P.J. Borm, D. Robbins, S. Haubold, T. Kuhlbusch, H. Fissan, K. Donaldson, R. Schins, V. Stone, W. Kreyling, J. Lademann, J. Krutmann, D. Warheit, E. Oberdorster, The potential risks of nanomaterials: a review carried out for ECETOC, Part Fibre Toxicol 3 (2006) 11. A. Peters, B. Veronesi, L. Calderon-Garciduenas, P. Gehr, L.C. Chen, M. Geiser, W. Reed, B. Rothen-Rutishauser, S. Schurch, H. Schulz, Translocation and potential neurological effects of fine and ultrafine particles a critical update, Part Fibre Toxicol 3 (2006) 13. M. Umezawa, H. Tainaka, N. Kawashima, M. Shimizu, K. Takeda, Effect of fetal exposure to titanium dioxide nanoparticle on brain development - brain region information, J Toxicol Sci 37 (2012) 1247-1252. K. Yamashita, Y. Yoshioka, K. Higashisaka, K. Mimura, Y. Morishita, M. Nozaki, T. Yoshida, T. Ogura, H. Nabeshi, K. Nagano, Y. Abe, H. Kamada, Y. Monobe, T. Imazawa, H. Aoshima, K. Shishido, Y. Kawai, T. Mayumi, S. Tsunoda, N. Itoh, T. Yoshikawa, I. Yanagihara, S. Saito, Y. Tsutsumi, Silica and titanium dioxide nanoparticles cause pregnancy complications in mice, Nat Nanotechnol 6 (2011) 321-328. A. Ebrahimzadeh Bideskan, A. Mohammadipour, A. Fazel, H. Haghir, H. Rafatpanah, M. Hosseini, A. Rajabzadeh, Maternal exposure to titanium dioxide nanoparticles during pregnancy and lactation alters offspring hippocampal mRNA BAX and Bcl-2 levels, induces apoptosis and decreases neurogenesis, Exp Toxicol Pathol 69 (2017) 329-337. A. Mohammadipour, A. Fazel, H. Haghir, F. Motejaded, H. Rafatpanah, H. Zabihi, M. Hosseini, A.E. Bideskan, Maternal exposure to titanium dioxide nanoparticles during pregnancy; impaired memory and decreased hippocampal cell proliferation in rat offspring, Environ Toxicol Pharmacol 37 (2014) 617-625. E. Paul, M.L. Franco-Montoya, E. Paineau, B. Angeletti, S. Vibhushan, A. Ridoux, A. Tiendrebeogo, M. Salome, B. Hesse, D. Vantelon, J. Rose, F. Canoui-Poitrine, J. Boczkowski, S. Lanone, C. Delacourt, J.C. Pairon, Pulmonary exposure to metallic nanomaterials during pregnancy irreversibly impairs lung development of the offspring, Nanotoxicology 11 (2017) 484-495. M.S. Acar, Z.B. Bulut, A. Ates, B. Nami, N. Kocak, B. Yildiz, Titanium dioxide nanoparticles induce cytotoxicity and reduce mitotic index in human amniotic fluid-derived cells, Hum Exp Toxicol 34 (2015) 74-82. C.D. Walkey, J.B. Olsen, H. Guo, A. Emili, W.C. Chan, Nanoparticle size and surface chemistry determine serum protein adsorption and macrophage uptake, J Am Chem Soc 134 (2012) 2139-2147. D. Brambilla, R. Verpillot, B. Le Droumaguet, J. Nicolas, M. Taverna, J. Kona, B. Lettiero, S.H. Hashemi, L. De Kimpe, M. Canovi, M. Gobbi, V. Nicolas, W. Scheper, S.M. Moghimi, I. Tvaroska, P. Couvreur, K. Andrieux, PEGylated
EP
[2]
AC C
478 479 480 481 482 483 484 485 486 487 488 489 490 491 492 493 494 495 496 497 498 499 500 501 502 503 504 505 506 507 508 509 510 511 512 513 514 515 516 517 518 519 520 521
[10]
[11]
[12]
ACCEPTED MANUSCRIPT
[16]
[17] [18] [19]
[20]
RI PT
EP
[21]
SC
[15]
M AN U
[14]
TE D
[13]
nanoparticles bind to and alter amyloid-beta peptide conformation: toward engineering of functional nanomedicines for Alzheimer's disease, ACS Nano 6 (2012) 5897-5908. K. Ulbrich, K. Hola, V. Subr, A. Bakandritsos, J. Tucek, R. Zboril, Targeted Drug Delivery with Polymers and Magnetic Nanoparticles: Covalent and Noncovalent Approaches, Release Control, and Clinical Studies, Chem Rev 116 (2016) 5338-5431. Z. Lin, N.A. Monteiro-Riviere, J.E. Riviere, Pharmacokinetics of metallic nanoparticles, Wiley Interdiscip Rev Nanomed Nanobiotechnol 7 (2015) 189-217. M. Chen, A. von Mikecz, Formation of nucleoplasmic protein aggregates impairs nuclear function in response to SiO2 nanoparticles, Exp Cell Res 305 (2005) 51-62. X. Yan, J. Li, Q. Liu, H. Peng, A. Popowich, Z. Wang, X.F. Li, p-Azidophenylarsenoxide: An Arsenical "Bait" for the In Situ Capture and Identification of Cellular Arsenic-Binding Proteins, 55 (2016) 14051-14056. G. Riquelme, Review: Placental syncytiotrophoblast membranes--domains, subdomains and microdomains, Placenta 32 Suppl 2 (2011) S196-202. N. Duran, C.P. Silveira, M. Duran, D.S. Martinez, Silver nanoparticle protein corona and toxicity: a mini-review, J Nanobiotechnology 13 (2015) 55. Z. Ma, J. Bai, X. Jiang, Monitoring of the Enzymatic Degradation of Protein Corona and Evaluating the Accompanying Cytotoxicity of Nanoparticles, ACS Appl Mater Interfaces 7 (2015) 17614-17622. M. Lundqvist, J. Stigler, G. Elia, I. Lynch, T. Cedervall, K.A. Dawson, Nanoparticle size and surface properties determine the protein corona with possible implications for biological impacts, Proc Natl Acad Sci U S A 105 (2008) 14265-14270. S. Tenzer, D. Docter, J. Kuharev, A. Musyanovych, V. Fetz, R. Hecht, F. Schlenk, D. Fischer, K. Kiouptsi, C. Reinhardt, K. Landfester, H. Schild, M. Maskos, S.K. Knauer, R.H. Stauber, Rapid formation of plasma protein corona critically affects nanoparticle pathophysiology, Nat Nanotechnol 8 (2013) 772-781. Z. Mao, B. Xu, X. Ji, K. Zhou, X. Zhang, M. Chen, X. Han, Q. Tang, X. Wang, Y. Xia, Titanium dioxide nanoparticles alter cellular morphology via disturbing the microtubule dynamics, Nanoscale 7 (2015) 8466-8475. S. Zhang, H. Gao, G. Bao, Physical Principles of Nanoparticle Cellular Endocytosis, ACS Nano 9 (2015) 8655-8671. J. Deng, C. Gao, Recent advances in interactions of designed nanoparticles and cells with respect to cellular uptake, intracellular fate, degradation and cytotoxicity, Nanotechnology 27 (2016) 412002. P. Ravanan, I.F. Srikumar, P. Talwar, Autophagy: The spotlight for cellular stress responses, Life Sci (2017). J.J. Lemasters, Variants of mitochondrial autophagy: Types 1 and 2 mitophagy and micromitophagy (Type 3), Redox Biol 2 (2014) 749-754.
AC C
522 523 524 525 526 527 528 529 530 531 532 533 534 535 536 537 538 539 540 541 542 543 544 545 546 547 548 549 550 551 552 553 554 555 556 557 558 559 560 561 562 563 564 565
[22]
[23] [24]
[25] [26]
ACCEPTED MANUSCRIPT
[32]
[33]
[34]
[35]
[36]
[37]
RI PT
[31]
SC
[30]
M AN U
[29]
TE D
[28]
A. Eiyama, K. Okamoto, PINK1/Parkin-mediated mitophagy in mammalian cells, Curr Opin Cell Biol 33 (2015) 95-101. S. Sweeney, A. Adamcakova-Dodd, P.S. Thorne, J.G. Assouline, Multifunctional nanoparticles for real-time evaluation of toxicity during fetal development, PLoS One 13 (2018) e0192474. S.J. Hawkins, L.A. Crompton, A. Sood, M. Saunders, Nanoparticle-induced neuronal toxicity across placental barriers is mediated by autophagy and dependent on astrocytes, 13 (2018) 427-433. B. Zhang, L. Tan, Y. Yu, B. Wang, Z. Chen, J. Han, M. Li, J. Chen, T. Xiao, B.K. Ambati, L. Cai, Q. Yang, N.R. Nayak, J. Zhang, X. Fan, Placenta-specific drug delivery by trophoblast-targeted nanoparticles in mice, Theranostics 8 (2018) 2765-2781. F. Hong, Y. Zhou, X. Zhao, L. Sheng, L. Wang, Maternal exposure to nanosized titanium dioxide suppresses embryonic development in mice, Int J Nanomedicine 12 (2017) 6197-6204. X. Ji, B. Xu, M. Yao, Z. Mao, Y. Zhang, G. Xu, Q. Tang, X. Wang, Y. Xia, Graphene oxide quantum dots disrupt autophagic flux by inhibiting lysosome activity in GC-2 and TM4 cell lines, Toxicology 374 (2016) 10-17. R. Chen, L. Huo, X. Shi, R. Bai, Z. Zhang, Y. Zhao, Y. Chang, C. Chen, Endoplasmic reticulum stress induced by zinc oxide nanoparticles is an earlier biomarker for nanotoxicological evaluation, ACS Nano 8 (2014) 2562-2574. M. Horie, K. Nishio, K. Fujita, S. Endoh, A. Miyauchi, Y. Saito, H. Iwahashi, K. Yamamoto, H. Murayama, H. Nakano, N. Nanashima, E. Niki, Y. Yoshida, Protein adsorption of ultrafine metal oxide and its influence on cytotoxicity toward cultured cells, Chem Res Toxicol 22 (2009) 543-553. H. Ruh, B. Kuhl, G. Brenner-Weiss, C. Hopf, S. Diabate, C. Weiss, Identification of serum proteins bound to industrial nanomaterials, Toxicol Lett 208 (2012) 41-50. E. Casals, T. Pfaller, A. Duschl, G.J. Oostingh, V.F. Puntes, Hardening of the nanoparticle-protein corona in metal (Au, Ag) and oxide (Fe3O4, CoO, and CeO2) nanoparticles, Small 7 (2011) 3479-3486. E. Casals, T. Pfaller, A. Duschl, G.J. Oostingh, V. Puntes, Time evolution of the nanoparticle protein corona, ACS Nano 4 (2010) 3623-3632. P. Roach, D. Farrar, C.C. Perry, Surface tailoring for controlled protein adsorption: effect of topography at the nanometer scale and chemistry, J Am Chem Soc 128 (2006) 3939-3945. S.T. Stern, P.P. Adiseshaiah, R.M. Crist, Autophagy and lysosomal dysfunction as emerging mechanisms of nanomaterial toxicity, Part Fibre Toxicol 9 (2012) 20. Y. Zhu, J.W. Eaton, C. Li, Titanium dioxide (TiO2) nanoparticles preferentially induce cell death in transformed cells in a Bak/Bax-independent fashion, PLoS One 7 (2012) e50607. S. Behzadi, V. Serpooshan, W. Tao, M.A. Hamaly, M.Y. Alkawareek, E.C. Dreaden, D. Brown, A.M. Alkilany, O.C. Farokhzad, M. Mahmoudi, Cellular
EP
[27]
AC C
566 567 568 569 570 571 572 573 574 575 576 577 578 579 580 581 582 583 584 585 586 587 588 589 590 591 592 593 594 595 596 597 598 599 600 601 602 603 604 605 606 607 608 609
[38]
[39]
[40]
[41]
ACCEPTED MANUSCRIPT
[46]
[47] [48]
[49] [50]
[51]
RI PT
SC
[45]
M AN U
[44]
TE D
[43]
EP
[42]
uptake of nanoparticles: journey inside the cell, Chem Soc Rev 46 (2017) 4218-4244. E.M. Sontag, R.S. Samant, J. Frydman, Mechanisms and Functions of Spatial Protein Quality Control, Annu Rev Biochem 86 (2017) 97-122. P. Walter, D. Ron, The unfolded protein response: from stress pathway to homeostatic regulation, Science 334 (2011) 1081-1086. Q. Yang, Y. Wang, Q. Yang, Y. Gao, X. Duan, Q. Fu, C. Chu, X. Pan, X. Cui, Y. Sun, Cuprous oxide nanoparticles trigger ER stress-induced apoptosis by regulating copper trafficking and overcoming resistance to sunitinib therapy in renal cancer, Biomaterials 146 (2017) 72-85. L. Huo, R. Chen, L. Zhao, X. Shi, R. Bai, D. Long, F. Chen, Y. Zhao, Y.Z. Chang, C. Chen, Silver nanoparticles activate endoplasmic reticulum stress signaling pathway in cell and mouse models: The role in toxicity evaluation, Biomaterials 61 (2015) 307-315. J.S. Nowak, D. Mehn, P. Nativo, C.P. Garcia, S. Gioria, I. Ojea-Jimenez, D. Gilliland, F. Rossi, Silica nanoparticle uptake induces survival mechanism in A549 cells by the activation of autophagy but not apoptosis, Toxicol Lett 224 (2014) 84-92. M.N. Moore, J.I. Allen, P.J. Somerfield, Autophagy: role in surviving environmental stress, Mar Environ Res 62 Suppl (2006) S420-425. M.N. Moore, Autophagy as a second level protective process in conferring resistance to environmentally-induced oxidative stress, Autophagy 4 (2008) 254-256. Y. Cao, D.J. Klionsky, Physiological functions of Atg6/Beclin 1: a unique autophagy-related protein, Cell Res 17 (2007) 839-849. S. Pankiv, T.H. Clausen, T. Lamark, A. Brech, J.A. Bruun, H. Outzen, A. Overvatn, G. Bjorkoy, T. Johansen, p62/SQSTM1 binds directly to Atg8/LC3 to facilitate degradation of ubiquitinated protein aggregates by autophagy, J Biol Chem 282 (2007) 24131-24145. K. Peynshaert, B.B. Manshian, F. Joris, K. Braeckmans, S.C. De Smedt, J. Demeester, S.J. Soenen, Exploiting intrinsic nanoparticle toxicity: the pros and cons of nanoparticle-induced autophagy in biomedical research, Chem Rev 114 (2014) 7581-7609. A. Lau, Y. Zheng, S. Tao, H. Wang, S.A. Whitman, E. White, D.D. Zhang, Arsenic inhibits autophagic flux, activating the Nrf2-Keap1 pathway in a p62-dependent manner, Mol Cell Biol 33 (2013) 2436-2446. X.J. Zhang, S. Chen, K.X. Huang, W.D. Le, Why should autophagic flux be assessed?, Acta Pharmacol Sin 34 (2013) 595-599. L. Avagliano, L. Terraneo, E. Virgili, C. Martinelli, P. Doi, M. Samaja, G.P. Bulfamante, A.M. Marconi, Autophagy in Normal and Abnormal Early Human Pregnancies, Reprod Sci 22 (2015) 838-844. T.H. Hung, S.F. Chen, L.M. Lo, M.J. Li, Y.L. Yeh, T.T. Hsieh, Increased autophagy in placentas of intrauterine growth-restricted pregnancies, PLoS One 7 (2012) e40957.
AC C
610 611 612 613 614 615 616 617 618 619 620 621 622 623 624 625 626 627 628 629 630 631 632 633 634 635 636 637 638 639 640 641 642 643 644 645 646 647 648 649 650 651 652 653
[52]
[53] [54]
[55]
ACCEPTED MANUSCRIPT
[57] [58] [59]
[60]
S. Saito, A. Nakashima, A review of the mechanism for poor placentation in early-onset preeclampsia: the role of autophagy in trophoblast invasion and vascular remodeling, J Reprod Immunol 101-102 (2014) 80-88. L. Ellenrieder, H. Rampelt, T. Becker, Connection of Protein Transport and Organelle Contact Sites in Mitochondria, J Mol Biol 429 (2017) 2148-2160. R.J. Youle, A.M. van der Bliek, Mitochondrial fission, fusion, and stress, Science 337 (2012) 1062-1065. D.P. Evenson, Z. Darzynkiewicz, M.R. Melamed, Simultaneous measurement by flow cytometry of sperm cell viability and mitochondrial membrane potential related to cell motility, J Histochem Cytochem 30 (1982) 279-280. S.M. Jin, R.J. Youle, The accumulation of misfolded proteins in the mitochondrial matrix is sensed by PINK1 to induce PARK2/Parkin-mediated mitophagy of polarized mitochondria, Autophagy 9 (2013) 1750-1757.
RI PT
[56]
SC
654 655 656 657 658 659 660 661 662 663 664 665 666 667
AC C
EP
TE D
M AN U
668
ACCEPTED MANUSCRIPT Acknowledgements
670
This work was supported by the National Key R&D Program of China
671
(2016YFC1000207), the National Natural Science Foundation of China (81630085,
672
81602884, 81502832), Qing Lan Project of Jiangsu Province, Six Talent Peaks Project
673
of Jiangsu Province (JY-052), Second Level of Training Object of Jiangsu Province
674
"333"
675
(BK20161585).
676
Competing interests
677
The authors declare that they have no competing interests.
and
the Natural Science Foundation of Jiangsu Province
AC C
EP
TE D
M AN U
SC
Project
RI PT
669
ACCEPTED MANUSCRIPT Highlights: - TiO2 NPs and intracellular protein aggregation was found in HTR-8/SVneo cells - ER stress and mitophagy was activated after TiO2 NPs treatment
AC C
EP
TE D
M AN U
SC
RI PT
- Interaction between TiO2 NPs and proteins like PERK and IRE1-α was suggested