Download as pdf or txt
Download as pdf or txt
You are on page 1of 10

Environmental Pollution 255 (2019) 113359

Contents lists available at ScienceDirect

Environmental Pollution
journal homepage: www.elsevier.com/locate/envpol

Mitochondrial respiratory chain dysfunction mediated by ROS is a


primary point of fluoride-induced damage in Hepa1-6 cells*
Hong-wei Wang, Yan Zhang, Pan-pan Tan, Liu-shu Jia, Yu Chen, Bian-hua Zhou*
Henan Key Laboratory of Environmental and Animal Product Safety, Henan University of Science and Technology, Luoyang 471003, Henan, China

a r t i c l e i n f o a b s t r a c t

Article history: To evaluate the mechanism of fluoride (F) mitochondrial toxicity, we cultured Hepa1-6 cells with
Received 21 July 2019 different F concentrations (0, 1 and 2 mmoL/L) and determined cell pathological morphology, mito-
Received in revised form chondrial respiratory chain damage and cell cycle change. Results showed that the activities and mRNA
12 September 2019
expression levels of antioxidant enzymes considerably decreased, whereas the contents of reactive ox-
Accepted 6 October 2019
ygen species (ROS), malondialdehyde (MDA) and nitric oxide (NO) markedly increased. Breakage of
Available online 7 October 2019
mitochondrial cristae and substantial vacuolated mitochondria were observed by transmission electron
microscopy. These results indicate the F-induced oxidative damage in Hepa1-6 cells. The enzyme ac-
Keywords:
Fluoride
tivities of mitochondrial complexes I, II, III and IV were disordered in Hepa1-6 cells treated by excessive F,
Mitochondrion thereby indicating a remarkable down-regulation. Further research showed that complex subunits also
Membrane potential demonstrated the development of disorder, in which the protein expressions levels of NDUFV2 and SDHA
ATP were substantially down-regulated, whereas those of CYC1 and COX Ⅳ were markedly up-regulated.
Apoptosis Reductions in ATP and mitochondrial membrane potential were detected with the dysfunction of the
Hepa1-6 mitochondrial respiratory chain. The G2/M phase arrest of the cell cycle in Hepa1-6 cells was measured
via flow cytometry, and the up-regulated protein expressions of Cyt c, caspase 9, caspase 3 and sub-
stantial apoptotic cells were determined. In summary, this study demonstrated that ROS-mediated
mitochondrial respiratory chain dysfunction causes F-induced Hepa1-6 cell damage.
© 2019 Elsevier Ltd. All rights reserved.

1. Introduction provides approximately 40% proton power of ATP synthesis and


mediates oxidative phosphorylation (OXPHOS) (Telford et al.,
The mitochondrial respiratory chain is an electron transport 2009); complex I also is the leading site for reactive oxygen spe-
system composed of four main enzyme complexes (I, II, III and IV) cies (ROS) production (Ait-Aissa et al., 2019) similar to complex III
located in the inner membrane of mitochondria (Sander and (France et al., 2017). The inhibition of mitochondrial complex I can
Garaude, 2018). Electrons are transferred from mitochondrial cause the decrease in ATP synthesis (Kilbride et al., 2008), oxidative
complex I (NADH dehydrogenase) and II (succinate dehydrogenase) damage and cell death (Urrutia et al., 2017). The inhibition of the
to complex III (ubiquinol cytochrome c oxidoreductase) via the mitochondrial respiratory chain complex activity can lead to severe
hydrophobic electron carrier ubiquinone and then from complex III mitochondrial dysfunction (Lo pez de Figueroa et al., 2015), thereby
to complex IV (cytochrome c oxidase) via cytochrome c (Cyt c) resulting in membrane potential loss (Wang et al., 2016), OXPHOS
(Davies et al., 2018). The electron transfer via the mitochondrial disturbance, decreased ATP production (Sharaf et al., 2017) and
respiratory chain is coupled with an electrochemical proton mitochondrial respiration inhibition (Teplova et al., 2017). Mito-
gradient on the mitochondrial intima, which is then utilised by ATP chondrial function and cell ATP content have been considered
synthase to generate ATP (Pe rez-Pe
rez et al., 2016). Complex I important factors affecting cell survival, and their changes may
account for apoptosis (Bergamini et al., 2016; Song et al., 2016).
Mitochondrial respiratory chain damage can induce a sharp
increase in intracellular ROS (Sun et al., 2015). When the ROS level
*
This paper has been recommended for acceptance by Dr. Sarah Harmon. exceeds the physiological threshold of the body, it will aggravate
* Corresponding author. Henan Key Laboratory of Environmental and Animal
mitochondrial damage and depress the antioxidant capacity of
Product Safety, Henan University of Science and Technology, Kaiyuan Avenue 263,
Luoyang 471003, Henan, China. cells, thus leading to a vicious cycle (Stefanatos and Sanz, 2018).
E-mail address: zhoubh@haust.edu.cn (B.-h. Zhou). ROS can destroy organelles and suborganelles (Fan et al., 2017) and

https://doi.org/10.1016/j.envpol.2019.113359
0269-7491/© 2019 Elsevier Ltd. All rights reserved.
2 H.-w. Wang et al. / Environmental Pollution 255 (2019) 113359

affect lipid metabolism, nucleic acid transcription and protein added for 4 h. The absorbance was read at 450 nm by using a
synthesis. Oxidative damages, including increased malondialde- microplate reader (Multiskan FC, Thermo Scientific, USA), and the F
hyde (MDA) generation (Zhou et al., 2015), decreased mitochon- IC50 of Hepa1-6 cells was calculated using GraphPad Prism 5. The
drial membrane potential decline (Li et al., 2017a) and DNA damage trials were confirmed by replicating the independent experiment
(Suzuki et al., 2015), are induced. Studies have shown that thrice.
mitochondria-mediated apoptosis arises from oxidative damage
(Wang et al., 2017c), and cell cycle arrest at G0/G1 phase can be 2.3. Transmission electron microscopy (TEM) observation
triggered by the increase in intracellular ROS, thereby aggravating
cell death (Vijayalakshmi and Sindhu, 2017). Based on the calculated concentration of IC50, the F doses of 0, 1
Fluorine exists widely in nature and is a kind of polar and active and 2 mmoL/L were used in subsequent experiments. The Hepa1-6
small molecular element, which can cause damage to various hard cells were collected and fixed overnight at 4  C in 2.5% glutaral-
tissues and soft tissues of the whole body (Zhou et al., 2015). Latest dehyde solution. The stationary solution was emptied. The samples
researches have shown that fluoride (F) can even cross the blood- were washed thrice in a phosphate buffer (PB, pH ¼ 7.0) and fixed in
brain barrier to damage the central nervous system (Goschorska 1% osmic acid solution for 1.5 h. The samples were washed thrice in
et al., 2018); can pass through the placenta to accumulate in the PB again, dehydrated with gradient ethanol, treated with pure
fetal brain, resulting in mental retardation in children (Green et al., acetone for 20 min and embedded in Araldite resin. After staining
2019), which has caused a strong response. At present, it is gener- 50 nm sections with uranyl acetate and lead citrate, the ultra-
ally believed that mitochondrion is one of the main targets of F structure of Hepa1-6 cells was observed using TEM (H-7500).
toxicity. F exposure can destroy mitochondrial membrane poten-
tial, interfere with the ATP production and also cause mitochondrial 2.4. Analysis of the biochemical markers of oxidative damage
fission and fusion disorder, bringing about mitochondrial defects
and cell death (Yan et al., 2017; Sun et al., 2016; Zhao et al., 2019b). The Hepa1-6 cells were homogenised in cold phosphate-
Our previous study has also demonstrated that F induces mito- buffered saline (PBS) for 60 s using an High-Speed Tissue Homog-
chondrial dysfunction, causing increase in the positive expression eniser (Servicebio, China) with vibration frequency of 60 Hz and
of ATP5J and ATP5H (Wang et al., 2017b). Mitochondrial respiratory centrifuged at 3,000g for 5 min. Supernatant was used to detect
chain is a necessary pathway for OXPHOS and ATP synthesis. the activities of superoxide dismutase (SOD, A001-1-1), glutathione
However, the relationship between F-induced mitochondrial peroxidase (GSH-Px, A005-1-1) and catalase (Cat, A007-1-1), and
dysfunction and mitochondrial respiratory chain as well as the the concentrations of MDA (A003-1-1) and total nitric oxide (T-NO,
mechanism of F damage to mitochondrial respiratory chain remain A014-1-1). These biochemical markers were determined via spec-
unclear. In this study, therefore, a model of Hepa1-6 cell exposed to trophotometry, following the standard procedures of commercially
F was established to explore the role of mitochondrial respiratory available diagnostic kits (Nanjing Jiancheng, China). For ROS
chain in F-induced cell impairment. determination, the Hepa1-6 cells were collected and incubated
with 10 mmoL/L dichloro-dihydro-fluorescein diacetate at 37  C for
2. Materials and methods 20 min, washed thrice with serum-free media and detected via flow
cytometry (BD Accuri C6, Becton Dickinson and Company, USA).
2.1. Cell culture
2.5. Real-time PCR
Hepa1-6 cells were obtained from Procell Life & Technology Co.,
Ltd., Wuhan, China. Hepa1-6 cells were cultured in Dulbecco’s Total cellular RNA was extracted from Hepa1-6 cells by using
Modified Eagle Medium (PM150210, Procell, China) supplemented TRIzol reagent (15596026, Invitrogen, USA), and RNase-free DNase I
with 10% foetal bovine serum (164210-500, Procell, China) and 1% was applied to remove the contaminated DNA. A small amount of
penicillin-streptomycin solution (P1400, Solarbio, Beijing), grown RNA solution was quantified on a nucleic acid analyser (Eppendorf,
at 37  C in a humidifying incubator (Thermo Scientific, USA) con- Germany). The extracted RNA was transcribed to cDNA by using a
taining 5% CO2 and sub-cultured every 48 h. SuperScript™ II Reverse Transcriptase kit (18064014, Invitrogen,
USA) in accordance with the manufacturer’s instructions. The
2.2. Determination of F half maximal inhibitory concentration specific primers of SOD1, GSH-Px1, Cat and nitric oxide synthase-2
(IC50) in Hepa1-6 cells (NOS2) were designed using Primer 5.0 software (Table 1). The
mRNA relative expression levels of SOD1, GSH-Px1, Cat and NOS2
Hepa1-6 cells were seeded in 96-well plates for 24 h. After F (0, were determined from the cDNA preparation by using the Bio-Rad
0.05, 0.1, 0.5, 1, 2.5, 5, 10 and 20 mmoL/L) was administered to the CFX96 Touch (Bio-Rad, USA) and the Premix Ex Taq™ (Perfect Real
medium for 48 h, cell counting kit-8 (DJ657, Beyotime, China) was Time) kit (DRR041A, Takara, Japan).

Table 1
Sequence of primers used in qRT-PCR.

Gene symbol Accession number Primer Primer Sequence (50 -30 ) Product size (bp)

GAPDH GU214026.1 Forward ACCCAGAAGACTGTGGATGG 171


Reverse CACATTGGGGGTAGGAACAC
Cat NM_009804 Forward ACATGGTCTGGGACTTCTGG 197
Reverse CAAGTTTTTGATGCCCTGGT
SOD1 NM_011434.1 Forward CCAGTGCAGGACCTCATTTT 197
Reverse TTGTTTCTCATGGACCACCA
GSH-PX1 NM_008160.6 Forward GAGGGTAGAGGCCGGATAAG 213
Reverse AGAAGGCATACACGGTGGAC
NOS2 AF065922.2 Forward CACCTTGGAGTTCACCCAGT 170
Reverse ACCACTCGTACTTGGGATGC
H.-w. Wang et al. / Environmental Pollution 255 (2019) 113359 3

2.6. Determination of enzyme activities of the mitochondrial After centrifugation, 5 mL of precooled 80% ethanol was added to
respiratory chain complex the cells, and the vortex-mixed cells were kept in the dark over-
night (>18 h) at 4  C. The cells were centrifuged at 1,200g for
The Hepa1-6 cells in logarithmic growth phase were seeded in a 10 min, and the supernatant was discarded. Then, the cells were
culture flask and cultured in an incubator containing 5% CO2 at washed with PBS and stain buffer to remove ethanol. The cells were
37  C for 24 h. Culture DMEM containing 0, 1 and 2 mmoL/L of F was resuspended in 0.5 mL PI/RNase A staining buffer, incubated for
added. After 48 h of F exposure, F medium was removed. Then, the 15 min at room temperature and detected using a flow cytometer
sample was washed with PBS, and the cells were collected. The cells (BD Accuri C6, Becton Dickinson and Company, USA).
were ruptured using a mitochondrial extraction kit, and the total
protein was determined using Coomassie brilliant blue. The protein 2.11. Immunofluorescence staining
content of each group was adjusted, and the enzyme activities of
mitochondrial respiratory chain complexes I, II, III and IV were Hepa1-6 cells were seeded on slides in a 6-well plate with a
measured using a kit for the determination of respiratory chain concentration of 2  104 cells/mL. The slides were fixed with 4%
complex enzyme activity in mitochondria (135897573, GENMED, paraformaldehyde for 15 min. After washing with PBS thrice, the
China). slides were added with 0.5% Triton X-100 (T8200, Solarbio, China)
to permeabilise the cells for 20 min at room temperature, and
2.7. Western blotting assay bovine serum albumin (Servicebio, China) was used for blocking for
30 min. Caspase 3 (GB13009-1, Servicebio, China), caspase 9 (9502,
Hepa1-6 cells were collected and homogenised in RIPA lysis CST, USA) and Cyt c (PB0291, Boster, China) were added and incu-
buffer (G2002, Servicebio, China) containing 1% PMSF. The ho- bated with the cells overnight at 4  C. After rinsing the unbound
mogenisation solution was incubated on ice for 30 min and monoclonal antibody, the slides were incubated with a fluorescent
centrifuged at 12,000g for 5 min at 4  C, and the supernatant was second antibody (GB21301, Servicebio, China) in a dark room for
collected. The protein concentration was determined using the BCA 1 h. DAPI (G1012, Servicebio, China) was added to stain the nuclei,
protein assay (G2026, Servicebio, China). Afterward, 25 mg of pro- and the slides were sealed with a mounting medium containing an
tein per sample was ran in 10% SDS-PAGE, and proteins were anti-fluorescence quenching agent. The images were observed
transferred onto PVDF membranes. The PVDF membranes were under a fluorescence microscope (Olympus, Japan).
blocked with 5% non-fat milk for 60 min. The membranes were
then incubated overnight at 4  C with primary antibodies against
2.12. TUNEL assay
NDUFV2 (15301-1-AP, Proteintech, China), SDHA (14865-1-AP,
Proteintech, China), CYC1 (10242-1-AP, Proteintech, China), COX Ⅳ
The degree of damage in Hepa1-6 cells was determined via
(11242-1-AP, Proteintech, China) and ACTIN (GB12001, Servicebio,
TUNEL assay. Frozen slices were fixed with cold acetone for 10 min.
China). The membranes were washed thrice with 0.5% TBS-Tween
After complete drying, the frozen slices were washed thrice on a
20 buffer (TBST) for 10 min each, followed by incubation with
decolorising shaking bed in PBS. The tissues were covered with
secondary antibodies (074-1506, KPL, USA) for 30 min at room
protease k solution and incubated at 37  C for 25 min. The slides
temperature. The protein bands on membranes were detected via
were also washed thrice in PBS on a decolorising shaking bed. The
enhanced chemiluminescence (G2014, Servicebio, China).
tissues were covered with broken membrane solution and incu-
bated at room temperature for 20 min. After cleaning with PBS,
2.8. Determination of ATP content
proper amounts of reagents 1 (TdT) and 2 (dUTP) in the TUNEL kit
(11684817910, Roche, Shanghai) were mixed at 1:9 ratio and added
The ATP content of Hepa1-6 cells was determined using a
to the slices, which were then incubated at 37  C for 2 h. Afterward,
luciferase-based luminescence enhanced ATP assay kit (S0027,
3% BSA was added for blocking at 37  C for 30 min. After removing
Beyotime, China). In accordance with the manufacturer’s in-
the sealing solution, a primary antibody was added, and the slices
structions, the cells in a 6-well plate were washed twice with ice-
were incubated overnight in a wet box at 4  C. Next, a second
cold PBS and homogenised in an ice-cold ATP lysis buffer. ATP
antibody was added and incubated at 37  C for 50 min. The nuclei
concentrations were then measured using an ATP standard with
were retained with DAPI solution and incubated at 37  C for 10 min.
the SpectraMax Paradigm multi-mode microplate reader (Molec-
The slices were sealed with anti-fluorescence quenching agents.
ular Devices, CA, USA).
Finally, the images were observed under a fluorescence microscope
and then collected. Green fluorescent nuclei were verified as pos-
2.9. Determination of mitochondrial membrane potential
itive for impaired Hepa1-6 cells.
The change in mitochondrial membrane potential was deter-
mined in accordance with the standard procedures by using a 2.13. Statistical analysis
mitochondrial membrane potential assay kit with JC-1 (C2006,
Beyotime, China). After exposing the Hepa1-6 cells to F, the cell Experimental data were expressed as the mean ± standard de-
culture medium was removed. Next, 1 mL cell culture medium and viation. Statistical analyses were performed via t-test. Significance
1 mL JC-1 staining working solution were added. The cells were was considered at P < 0.05.
incubated for 20 min at 37  C. The suspension was centrifuged at
600g for 3 min, and the supernatant was discarded. After washing 3. Results
twice with JC-1 staining buffer (1  ), 2 mL cell culture medium was
added, and the Hepa1-6 cells were observed under a fluorescence 3.1. IC50 of F-induced Hepa1-6 cells
microscope (Olympus, Japan).
As shown in Fig. 1, the survival rate of Hepa1-6 cells decreased
2.10. Determination of cell cycle with the increase in F dosage, and the F IC50 of 2.074 mmoL/L was
obtained in Hepa1-6. Thus, F concentrations of 0 (control group), 1
The cells were harvested in a flow tube and cleaned with PBS. and 2 mmoL/L were selected in this study.
4 H.-w. Wang et al. / Environmental Pollution 255 (2019) 113359

3.5. F-induced changes in the mitochondrial membrane potential


and cell cycle in Hepa1-6 cells

Fig. 5 displays the changes in mitochondrial membrane poten-


tial. With the increase in F doses, JC-aggregates decreased, and JC-
monomers simultaneously increased compared with the control
group, which illustrated that mitochondrial membrane potential
collapsed.
In comparison with the control group, the number of G2/M
phase Hepa1-6 cells in the F groups distinctly increased (blue peak;
Fig. 6a). The proportion of G2/M phase Hepa1-6 cells significantly
increased given excessive F, particularly at 2 mmoL/L F (P < 0.01),
and the proliferation index (PI) of Hepa1-6 cells was significantly
decreased at 2 mmoL/L F (P < 0.05; Fig. 6b and c).

3.6. F-induced changes in Cyt c, caspase 9 and caspase 3 protein


expression levels in Hepa1-6 cells
Fig. 1. F IC50 of Hepa1-6 cells. After 48 h of F (0, 0.05, 0.1, 0.5, 1, 2.5, 5, 10 and 20 mmoL/
L) exposure, the survival rate of Hepa1-6 cells decreased with the increase in F doses,
and the IC50 of this was showed to be 2.074 mmoL/L. Fig. 7a, b and 7c show the higher fluorescence intensities of Cyt
c, caspase 9 and caspase 3, respectively, in the F 1 and 2 mmoL/L
groups than those in the control group. These results indicated the
3.2. Ultrastructure of the Hepa1-6 cells protein expression levels of Cyt c, caspase 9 and caspase 3 increased
compared with the control group.
Fig. 2 depicts the changes in the ultrastructure of the Hepa1-6
cells. Clear nuclear membrane and mitochondrial cristae and a 3.7. TUNEL assay
normal structure of endoplasmic reticulum (ER) were observed in
the control group (Fig. 2a, a1, a2 and a3). By contrast, vague nuclear Numerous Hepa1-6 cells that lacked apparent signs of apoptosis
membrane, dilated ER and distinctly increased lysosomes were were observed in the control group (Fig. 7dd1 and dd2). The pos-
observed in F-treated Hepa1-6 cells (Fig. 2b, b1, c and c1), especially itive expressions became increasingly evident with the increase in F
changes in mitochondria with the breakage of mitochondrial doses (Fig. 7dd3, dd4, dd5 and dd6). Especially in the F 2 mmoL/L
cristae and generation of substantial vacuolated mitochondria. group, a large area of green fluorescence Hepa1-6 cells was
(Fig. 2b2, b3, c2 and c3). observed (Fig. 7dd5 and dd6).

4. Discussion
3.3. Effect of F on the biochemical markers of oxidative damage in
Hepa1-6 cells Hepa1-6 cells, which are representatives of a mouse hepatoma
cell line, are frequently used to evaluate the toxicity of substances
In comparison with the control group, the ROS contents of toxic to cells. In this study, Hepa1-6 cells were selected to investi-
Hepa1-6 cells were significantly increased in F groups (P < 0.01; gate F cytotoxicity. Accumulating evidences have demonstrated
Fig. 3a and b). The activities of SOD, GSH-Px, and total antioxidant that F can cause oxidative stress in the liver (Lu et al., 2017), kidney
capacity (T-AOC) decreased significantly in the cells exposed to F, (Adedara et al., 2017), brain (Sarkar et al., 2014; Wang et al., 2018),
especially at 2 mmoL/L F (P < 0.01; Fig. 3c). The concentrations of thyroid (Banji et al., 2013) and embryo (Zhao et al., 2017). We
MDA and T-NO significantly increased in the F groups (P < 0.01; observed that F has caused severe damage to the ultrastructure of
Fig. 3d). Hepa1-6 cells; the cell injuries include vague nuclear membrane,
As shown in Fig. 3e, in comparison with the control group, the dilated ER, cracked mitochondrial cristae, vacuolated mitochondria
mRNA expression levels of SOD1, GSH-Px1 and Cat were signifi- in large quantities and increased lysosomes. The main cause of
cantly decreased in the F groups (P < 0.05 or P < 0.01). NOS2 damage in the ultrastructure induced by F may be the significant
exhibited significantly increased mRNA expression levels with the increase in ROS in Hepa1-6 cells. Excessive ROS can destroy the
increase in F doses (P < 0.05 or P < 0.01). These results indicated activities of SOD, GSH-Px and Cat (Mohamed, 2019) and engender
that F induces oxidative stress, thereby leading to the damage in the lipid peroxidation (LPO). After exposure to F, the antioxidant ca-
ultrastructure of Hepa1-6 cells. pacity decreased, and oxidative stress was triggered by the inability
of cells to convert superoxide (Li et al., 2017b). In the present study,
F remarkably inhibited the protein expression levels and activities
3.4. Effect of F on ATP concentration, mitochondrial complexes, and of SOD, GSH-Px and Cat and promoted the generation of MDA and
relative subunits in Hepa1-6 cells NO in Hepa1-6 cells. These results confirm that excessive ROS in-
duces the oxidative damage in the ultrastructure of Hepa1-6 cells,
The ATP concentration of Hepa1-6 cells was significantly especially in the mitochondria.
decreased (Fig. 4a). The enzyme activities of mitochondrial com- Mitochondrial cristae represent a specialised chamber that
plexes I, II, III and IV, as displayed in Fig. 4b, were markedly down- harbours respiratory chain and ATP synthase, and the integrity of
regulated after exposure to F, especially at 2 mmoL/L F (P < 0.01). In the crista membrane is a prerequisite to drive OXPHOS (Ikon and
comparison with those in the control groups, the protein expres- Ryan, 2017; Rampelt et al., 2017). The breakage and loss of mito-
sion levels of NDUFV2 and SDHA were markedly down-regulated in chondrial cristae in Hepa1-6 cells, inevitably affected the respira-
the F groups (P < 0.05 or P < 0.01). CYC1 and COX Ⅳ protein tory chain function and destroyed the OXPHOS course. Complexes I,
expression levels showed remarkable up-regulation in Hepa1-6 II, III and IV constitute the respiratory chain with ubiquinone and
cells with the increase in F concentrations (P < 0.01; Fig. 4c and d). Cyt c; their inhibition can decrease electron transfer kinetics and
H.-w. Wang et al. / Environmental Pollution 255 (2019) 113359 5

Fig. 2. Effect of F on the ultrastructure of Hepa1-6 cells. a, a1, a2 and a3: Hepa1-6 cells from the control group. b, b1, b2 and b3: Hepa1-6 cells from the F 1 mmoL/L group. c, c1, c2
and c3: Hepa1-6 cells from the F 2 mmoL/L group. Nu, NM, Mi, ER and Ly represent nucleus, nucleus membrane, mitochondrion, endoplasmic reticulum and lysosome, respectively.

impair cell respiratory function (Sanderson et al., 2013; Luo et al., protein expression levels of complex subunits NDUFV2 and SDHA
2019). In addition, complexes I, III and IV feature a proton- and promoting the generation of MDA also provide considerable
pumping function. When their activities decline, the function of evidence. In addition, an evident sign of mitochondrial respiratory
the proton pump is impaired, thereby causing decreased mito- chain damage is the remarkable increase in ROS (Matsuhashi et al.,
chondrial membrane potential, power-deficient ADP phosphory- 2017; Chauhan et al., 2013). Complexes I and III are the main sites of
lation and reduced ATP synthesis (Connolly et al., 2018). Swollen electron leakage in the mitochondria, and leaked electron can react
mitochondria and increased outer membrane permeability touch with O2 to form superoxides (Sun et al., 2015). Under abnormal
off the release of pro-apoptotic factor Cyt c in the mitochondria conditions, other respiratory chain complexes can also cause an
(Poo r et al., 2019; Wang et al., 2017a). Here, the decreased ATP increase in ROS. The dysfunction of complex II mainly caused the
content and the release of Cyt c were detected; these events are excessive generation of H2O2 in the brain of Rett syndrome mice
important indicators of mitochondrial dysfunction. The mito- (De Filippis et al., 2015). The blockage of complex IV can lead to a
chondrial function disorder of Hepa1-6 cells might be attributed to reverse electron transfer in the respiratory chain, that is, ubiqui-
the damage of respiratory chain complex and LPO impairment of none transfers electrons to complex I, resulting in the leakage of
membrane as the site of the complex. The results showing that F numerous electrons and a high level of ROS (Scialo  et al., 2017). ROS
remarkably inhibited the activities of complexes I, II, III and IV and bursts further aggravate the oxidative damage of crista membrane
6 H.-w. Wang et al. / Environmental Pollution 255 (2019) 113359

Fig. 3. Analysis of the biochemical markers of oxidative damage and real-time PCR of different gene levels in Hepa1-6 cells. (a) ROS fluorescence intensity in Hepa1-6 cells. (b) The
analysis of ROS generation in Hepa1-6 cells. (c) SOD, GSH-Px and T-AOC activities in Hepa1-6 cells. (d) MDA and T-NO concentrations in Hepa1-6 cells. (e) mRNA relative expression
levels of Cat, SOD1, GSH-Px1 and NOS2 in Hepa1-6 cells. *P < 0.05, **P < 0.01.

Fig. 4. ELISA, Western blotting assay and ATP concentration analysis. (a) The change in ATP concentration in Hepa1-6 cells. (b) The activities of mitochondrial complex I, II, III and IV.
“COX” is the abbreviation of “complex”. (c) Western blotting electrophoretic pattern of complex subunits. (d) Relative protein expression levels of complex subunits.
H.-w. Wang et al. / Environmental Pollution 255 (2019) 113359 7

Fig. 5. Determination of Mitochondrial membrane potential in Hepa1-6 cells. Red fluorescence and green fluorescence are represent JC-aggregates and JC-monomers, respectively.
After F exposure for 48 h, the red fluorescence intensity of Hepa1-6 cells in F 1 and 2 mmoL/L groups was down-regulated, while green fluorescence intensity was up-regulated
compared with the control group. These changes indicate the decreased membrane potential.

Fig. 6. Cell cycle determination. (a) The changes in number of different phase Hepa1-6 cells. Red, gray and blue peak represent G0/G1, S and G2/M phase, respectively. (b) The
proportion of cell cycle of Hepa1-6 cells. (c) The proliferation index (PI) of Hepa1-6 cells.

and destroy the mitochondrial respiratory chain, thereby causing a considering the elevated ROS-mediated dysfunction of the respi-
vicious cycle that substantially affects the mitochondrial function ratory chain. We detected a remarkable decrease in the mito-
(Lin et al., 2018). chondrial membrane potential of Hepa1-6 cells. This decrease led
The alteration of mitochondrial function is a strong indicator of to the release of intracellular pro-apoptotic factor Cyt c. Cyt c has
apoptosis (Meng et al., 2018; Song et al., 2019). ROS can trigger the been combined with apoptosis protease-activating factor-1 (Apaf-
apoptosis of caspase-9-dependent endogenous mitochondrial 1) to induce conformational change and oligomerisation of Apaf-1
pathway (Zhang et al., 2015; Liu et al., 2016; Gong et al., 2019) (Su et al., 2016), which further recruits and activates pro-caspase 9
8 H.-w. Wang et al. / Environmental Pollution 255 (2019) 113359

Fig. 7. Immunofluorescence assay and TUNEL assay. (a), (b) and (c) respectively showed the fluorescence intensities of Cyt c, caspase 9 and caspase 3 protein expressions levels in
Hepa1-6 cells. (d) Showed the result of TUNEL assay. d1 and d2 are control groups, d3 and d4 represent F 1 mmoL/L groups, d5 and d6 mean F 2 mmoL/L groups. The green
fluorescence part showed the apoptotic cells.

via the caspase 9 recruitment domain at the N-terminal. Activated arrested at the G2/M phase, thereby indicating the development of
caspase 9 continuously cleaves and induces caspase 3 to initiate apoptosis in the Hepa1-6 cells. This conclusion is consistent with
caspase cascade reaction, thus touching off cell apoptosis and DNA previous findings. The decrease in the ATP content is an important
degradation (Lee et al., 2016; Zhang et al., 2016). This process was factor for cell cycle delay and cell death (Du et al., 2017). Accord-
demonstrated by the increased expression levels of caspase 9, ingly, in this study, the decline in the ATP content caused by
caspase 3 and Cyt c and the increased apoptosis of Hepa1-6 cells in mitochondrial respiratory chain damage may be another method
the TUNEL assay after F exposure. Moreover, caspase activation and for inducing apoptosis in Hepa1-6 cells.
the increased intracellular ROS, which are accompanied by cell
cycle S-phase arrest, result in cell apoptosis (Tania et al., 2019; Zhao 5. Conclusions
et al., 2019a). Increased ROS level leads to the cell cycle arrest at the
G2/M phase, thereby causing cell apoptosis, which can eliminate As illustrated in Fig. 8, this study demonstrated that ROS-
cancer cells (Hirata et al., 2019). The present study determined that mediated mitochondrial respiratory chain dysfunction and
the content of ROS increased, and the cell cycle of Hepa1-6 cells was apoptotic mechanism cause F-induced Hepa1-6 cell damage.
H.-w. Wang et al. / Environmental Pollution 255 (2019) 113359 9

complex II/III respiratory chain deficiency. Fetal Pediatr. Pathol. 36 (3),


263e264.
Gong, Z.G., Wang, X.Y., Wang, J.H., Fan, R.F., Wang, L., 2019. Trehalose prevents
cadmium-induced hepatotoxicity by blocking Nrf2 pathway, restoring auto-
phagy and inhibiting apoptosis. J. Inorg. Biochem. 192, 62e71.
Goschorska, M., Baranowska-Bosiacka, I., Gutowska, I., Metryka, E., Sko rka-
Majewicz, M., Chlubek, D., 2018. Potential role of fluoride in the etiopatho-
genesis of alzheimer’s disease. Int. J. Mol. Sci. 19 (12), 3965.
Green, R., Lanphear, B., Hornung, R., et al., 2019. Association between maternal
fluoride exposure during pregnancy and IQ scores in offspring in Canada. JAMA
Pediatr.
Hirata, T., Cho, Y.M., Suzuki, I., et al., 2019. 4-Methylthio-3-butenyl isothiocyanate
(MTBITC) induced apoptotic cell death and G2/M cell cycle arrest via ROS
production in human esophageal epithelial cancer cells. J. Toxicol. Sci. 44 (2),
73e81.
Ikon, N., Ryan, R.O., 2017. Cardiolipin and mitochondrial cristae organization. Bio-
chim. Biophys. Acta Biomembr. 1859 (6), 1156e1163.
Kilbride, S.M., Telford, J.E., Tipton, K.F., Davey, G.P., 2008. Partial inhibition of
complex I activity increases Ca2þ-independent glutamate release rates from
depolarized synaptosomes. J. Neurochem. 106 (2), 826e834.
Lee, M.H., Hong, S.H., Park, C., et al., 2016. Hwang-Heuk-San induces apoptosis in
HCT116 human colorectal cancer cells through the ROS-mediated activation of
caspases and the inactivation of the PI3K/Akt signaling pathway. Oncol. Rep. 36
(1), 205e214.
Li, A.X., Sun, M., Li, X., 2017a. Withaferin-A induces apoptosis in osteosarcoma U2OS
cell line via generation of ROS and disruption of mitochondrial membrane
Fig. 8. The schema indicates that fluoride induces mitochondrial respiratory chain potential. Eur. Rev. Med. Pharmacol. Sci. 21 (6), 1368e1374.
damage and thus triggers apoptosis. After F exposure, ROS increased and mitochon- Li, W.S., Jiang, B.H., Cao, X.L., Xie, Y.J., Huang, T., 2017b. Protective effect of lycopene
drial respiratory chain was impaired in Hepa1-6 cells, thus resulting in remarkably on fluoride-induced ameloblasts apoptosis and dental fluorosis through
decreased enzyme activities of complex I, II, III and IV. The protein expression levels of oxidative stress-mediated caspase pathways. Chem. Biol. Interact. 261, 27e34.
NDUFV2 and SDHA were markedly down-regulated, whereas CYC1 and COX Ⅳ protein Lin, J., Xia, J., Zhao, H.S., et al., 2018. Lycopene triggers Nrf2-AMPK cross talk to
expression levels were remarkably up-regulated. Simultaneously, reductions in ATP alleviate atrazine-induced nephrotoxicity in mice. J. Agric. Food Chem. 66 (46),
synthesis and mitochondrial membrane potential caused Cyt c release, which further 12385e12394.
triggered caspase cascade reaction, thereby touching off cell apoptosis. Liu, G., Wang, Z.K., Wang, Z.Y., Yang, D.B., Liu, Z.P., Wang, L., 2016. Mitochondrial
permeability transition and its regulatory components are implicated in
apoptosis of primary cultures of rat proximal tubular cells exposed to lead.
Arch. Toxicol. 90 (5), 1193e1209.
Declaration of competing interest pez de Figueroa, P., Lotz, M.K., Blanco, F.J., Carame
Lo s, B., 2015. Autophagy activation
and protection from mitochondrial dysfunction in human chondrocytes.
Arthritis Rheumatol. 67 (4), 966e976.
The authors declare that there is no conflict of interest. Lu, Y.J., Luo, Q., Cui, H.M., et al., 2017. Sodium fluoride causes oxidative stress and
apoptosis in the mouse liver. Aging (Albany NY) 9 (6), 1623.
Luo, Y., Li, X.N., Zhao, Y., Du, Z.H., Li, J.L., 2019. DEHP triggers cerebral mitochondrial
Acknowledgements
dysfunction and oxidative stress in quail (Coturnix japonica) via modulating
mitochondrial dynamics and biogenesis and activating Nrf2-mediated defense
This work is sponsored by the National Natural Science Foun- response. Chemosphere 224, 626e633.
dation of China (grant no. 31201963) and Young Backbone Teachers Matsuhashi, T., Sato, T., Kanno, S.I., et al., 2017. Mitochonic Acid 5 (MA-5) facilitates
ATP synthase oligomerization and cell survival in various mitochondrial dis-
Training Project of Colleges and Universities in Henan Province, eases. EBioMedicine 20, 27e38.
China (grant no. 2016GGJS-061). Meng, L., Xin, G., Li, B., et al., 2018. Anthocyanins extracted from aronia melanocarpa
protect SH-SY5Y cells against amyloid-beta (1e42)-induced apoptosis by
regulating Ca2þ homeostasis and inhibiting mitochondrial dysfunction. J. Agric.
References Food Chem. 66 (49), 12967e12977.
Mohamed, H.R.H., 2019. Estimation of genomic instability and mitochondrial DNA
Adedara, I.A., Ojuade, T.J.D., Olabiyi, B.F., et al., 2017. Taurine ameliorates renal damage induction by acute oral administration of calcium hydroxide normal-
oxidative damage and thyroid dysfunction in rats chronically exposed to fluo- and nano-particles in mice. Toxicol. Lett. 304, 1e12.
ride. Biol. Trace Elem. Res. 175 (2), 388e395. rez-Pe
Pe rez, R., Lobo-Jarne, T., Milenkovic, D., et al., 2016. COX7A2L is a mito-
Ait-Aissa, K., Blaszak, S.C., Beutner, G., et al., 2019. Mitochondrial oxidative phos- chondrial complex III binding protein that stabilizes the III2þIV supercomplex
phorylation defect in the heart of subjects with coronary artery disease. Sci. without affecting respirasome formation. Cell Rep. 16 (9), 2387e2398.
Rep. 9 (1), 7623. Poo r, P., Patyi, G., Taka
cs, Z., et al., 2019. Salicylic acid-induced ROS production by
Banji, D., Banji, O.J., Pratusha, N.G., Annamalai, A.R., 2013. Investigation on the role mitochondrial electron transport chain depends on the activity of mitochon-
of spirulina platensis in ameliorating behavioural changes, thyroid dysfunction drial hexokinases in tomato (Solanum lycopersicum L.). J. Plant Res. 132 (2),
and oxidative stress in offspring of pregnant rats exposed to fluoride. Food 273e283.
Chem. 140 (1‒2), 321e331. Rampelt, H., Zerbes, R.M., van der Laan, M., Pfanner, N., 2017. Role of the mito-
Bergamini, C., Moruzzi, N., Volta, F., et al., 2016. Role of mitochondrial complex I and chondrial contact site and cristae organizing system in membrane architecture
protective effect of CoQ 10 supplementation in propofol induced cytotoxicity. and dynamics. Biochim. Biophys. Acta Mol. Cell Res. 1864 (4), 737e746.
J. Bioenerg. Biomembr. 48 (4), 413e423. Sander, L.E., Garaude, J., 2018. The mitochondrial respiratory chain: a metabolic
Chauhan, S.S., Mahmood, A., Ojha, S., 2013. Ethanol and age enhances fluoride rheostat of innate immune cell-mediated antibacterial responses. Mitochon-
toxicity through oxidative stress and mitochondrial dysfunctions in rat intes- drion 41, 28e36.
tine. Mol. Cell. Biochem. 384 (1‒2), 251e262. Sanderson, T.H., Reynolds, C.A., Kumar, R., Przyklenk, K., Hüttemann, M., 2013.
Connolly, N.M.C., Theurey, P., Adam-Vizi, V., et al., 2018. Guidelines on experimental Molecular mechanisms of ischemia-reperfusion injury in brain: pivotal role of
methods to assess mitochondrial dysfunction in cellular models of neurode- the mitochondrial membrane potential in reactive oxygen species generation.
generative diseases. Cell Death Differ. 25 (3), 542. Mol. Neurobiol. 47 (1), 9e23.
Davies, K.M., Blum, T.B., Kühlbrandt, W., 2018. Conserved in situ arrangement of Sarkar, C., Pal, S., Das, N., Dinda, B., 2014. Ameliorative effects of oleanolic acid on
complex I and III2 in mitochondrial respiratory chain supercomplexes of fluoride induced metabolic and oxidative dysfunctions in rat brain: experi-
mammals, yeast, and plants. Proc. Natl. Acad. Sci. 115 (12), 3024e3029. mental and biochemical studies. Food Chem. Toxicol. 66, 224e236.
De Filippis, B., Valenti, D., de Bari, L., et al., 2015. Mitochondrial free radical over- Scialo, F., Fernandez-Ayala, D.J., Sanz, A., 2017. Role of mitochondrial reverse elec-
production due to respiratory chain impairment in the brain of a mouse model tron transport in ROS signaling: potential roles in health and disease. Front.
of Rett syndrome: protective effect of CNF1. Free Radic. Biol. Med. 83, 167e177. Physiol. 8, 428.
Du, X., Fu, X.F., Yao, K., et al., 2017. Bcl-2 delays cell cycle through mitochondrial ATP Sharaf, M.S., Stevens, D., Kamunde, C., 2017. Mitochondrial transition ROS spike
and ROS. Cell Cycle 16 (7), 707e713. (mTRS) results from coordinated activities of complex I and nicotinamide
Fan, W.T., Shen, T.T., Ding, Q.Q., et al., 2017. Zearalenone induces ROS-mediated nucleotide transhydrogenase. Biochim. Biophys. Acta Bioenerg. 1858 (12),
mitochondrial damage in porcine IPEC-J2 cells. J. Biochem. Mol. Toxicol. 31 955e965.
(10), e21944. Song, B., Aoki, S., Liu, C., Ito, K., 2019. A toll-like receptor 9 agonist sensitizes mice to
France, J., Ashoor, I., Craver, R., 2017. Renal tubular mitochondrial abnormalities in mitochondrial dysfunction-induced hepatic apoptosis via the Fas/FasL pathway.
10 H.-w. Wang et al. / Environmental Pollution 255 (2019) 113359

Arch. Toxicol. 1e12. Wang, C., Liang, C., Ma, J., et al., 2018. Co-exposure to fluoride and sulfur dioxide on
Song, X.B., Liu, G., Wang, Z.Y., Wang, L., 2016. Puerarin protects against cadmium- histological alteration and DNA damage in rat brain. J. Biochem. Mol. Toxicol. 32
induced proximal tubular cell apoptosis by restoring mitochondrial function. (2), e22023.
Chem. Biol. Interact. 260, 219e231. Wang, H., Chen, Y., Zhai, N., et al., 2017a. Ochratoxin A-induced apoptosis of IPEC-J2
Stefanatos, R., Sanz, A., 2018. The role of mitochondrial ROS in the aging brain. FEBS cells through ROS-mediated mitochondrial permeability transition pore open-
Lett. 592 (5), 743e758. ing pathway. J. Agric. Food Chem. 65 (48), 10630e10637.
Su, K., Wang, C.F., Zhang, Y., Ca, Y.J., Zhang, Y.Y., Zhao, Q., 2016. The inhibitory effects Wang, H.W., Zhao, W.P., Liu, J., Tan, P.P., Tian, W.S., Zhou, B.H., 2017b. ATP5J and
of carnosic acid on cervical cancer cells growth by promoting apoptosis via ATP5H proactive expression correlates with cardiomyocyte mitochondrial
ROS-regulated signaling pathway. Biomed. Pharmacother. 82, 180e191. dysfunction induced by fluoride. Biol. Trace Elem. Res. 180 (1), 63e69.
Sun, Q., Zhong, W., Zhang, W.L., Zhou, Z.X., 2015. Defect of mitochondrial respiratory Wang, H.W., Zhao, W.P., Liu, J., Tan, P.P., Zhang, C., Zhou, B.H., 2017c. Fluoride-
chain is a mechanism of ROS overproduction in a rat model of alcoholic liver induced oxidative stress and apoptosis are involved in the reducing of oocytes
disease: role of zinc deficiency. Am. J. Physiol. Gastrointest. Liver Physiol. 310 development potential in mice. Chemosphere 186, 911e918.
(3), G205eG214. Wang, L., Zhang, X.J., Cui, G.Z., et al., 2016. A novel agent exerts antitumor activity in
Sun, Z.L., Zhang, W., Xue, X.C., et al., 2016. Fluoride decreased the sperm ATP of mice breast cancer cells by targeting mitochondrial complex II. Oncotarget 7 (22),
through inhabiting mitochondrial respiration. Chemosphere 144, 1012e1017. 32054.
Suzuki, M., Bandoski, C., Bartlett, J.D., 2015. Fluoride induces oxidative damage and Yan, X.Y., Wang, L., Yang, X., et al., 2017. Fluoride induces apoptosis in H9c2 car-
SIRT1/autophagy through ROS-mediated JNK signaling. Free Radic. Biol. Med. diomyocytes via the mitochondrial pathway. Chemosphere 182, 159e165.
89, 369e378. Zhang, J.H., Xia, Y.F., Xu, Z.F., Deng, X.M., 2016. Propofol suppressed hypoxia/
Tania, M., Shawon, J., Saif, K., et al., 2019. Cordycepin downregulates Cdk-2 to reoxygenation-induced apoptosis in HBVSMC by regulation of the expression
interfere with cell cycle and increases apoptosis by generating ROS in cervical of Bcl-2, Bax, Caspase3, Kir6.1, and p-JNK. Oxid. Med. Cell. Longev. 2016.
cancer cells: in vitro and in silico study. Curr. Cancer Drug Targets 19 (2), Zhang, M., Harashima, N., Moritani, T., Huang, W., Harada, M., 2015. The roles of ROS
152e159. and caspases in TRAIL-induced apoptosis and necroptosis in human pancreatic
Telford, J.E., Kilbride, S.M., Davey, G.P., 2009. Complex I is rate-limiting for oxygen cancer cells. PLoS One 10 (5), e0127386.
consumption in the nerve terminal. J. Biol. Chem. 284 (14), 9109e9114. Zhao, C.K., Wang, M.S., Cheng, A.C., et al., 2019a. Duck plague virus promotes DEF
Teplova, V.V., Belosludtsev, K.N., Kruglov, A.G., 2017. Mechanism of triclosan cell apoptosis by activating caspases, increasing intracellular ROS levels and
toxicity: mitochondrial dysfunction including complex II inhibition, superoxide inducing cell cycle S-phase arrest. Viruses 11 (2), 196.
release and uncoupling of oxidative phosphorylation. Toxicol. Lett. 275, Zhao, J.M., Fu, B.B., Peng, W., Mao, T.C., Wu, H.B., Zhang, Y., 2017. Melatonin protect
108e117. the development of preimplantation mouse embryos from sodium fluoride-
Urrutia, P.J., Aguirre, P., Tapia, V., Carrasco, C.M., Mena, N.P., Nún ~ ez, M.T., 2017. Cell induced oxidative injury. Environ. Toxicol. Pharmacol. 54, 133e141.
death induced by mitochondrial complex I inhibition is mediated by Iron Zhao, Q., Niu, Q., Chen, J.W., et al., 2019b. Roles of mitochondrial fission inhibition in
Regulatory Protein 1. Biochim. Biophys. Acta (BBA) - Mol. Basis Dis. 1863 (9), developmental fluoride neurotoxicity: mechanisms of action in vitro and as-
2202e2209. sociations with cognition in rats and children. Arch. Toxicol. 93 (3), 709e726.
Vijayalakshmi, A., Sindhu, G., 2017. Umbelliferone arrest cell cycle at G0/G1 phase Zhou, B.H., Zhao, J., Liu, J., Zhang, J.L., Li, J., Wang, H.W., 2015. Fluoride-induced
and induces apoptosis in human oral carcinoma (KB) cells possibly via oxidative oxidative stress is involved in the morphological damage and dysfunction of
DNA damage. Biomed. Pharmacother. 92, 661e671. liver in female mice. Chemosphere 139, 504e511.

You might also like