Citation: Xu B, Sui YL, Fan TJ. Gatifloxacin inducing
apoptosis of stromal fibroblasts through cross-talk between caspase-dependent
extrinsic and intrinsic pathways. Int J Ophthalmol
2019;12(10):1524-1530. DOI:10.18240/ijo.2019.10.02
·Basic
Research·
Gatifloxacin
inducing apoptosis of stromal fibroblasts through cross-talk between
caspase-dependent extrinsic and intrinsic pathways
Bin Xu, Yun-Long Sui, Ting-Jun
Fan
Laboratory for Corneal Tissue
Engineering, College of Marine Life Sciences, Ocean University of China,
Qingdao 266003, Shandong Province, China
Correspondence to: Ting-Jun Fan. Laboratory for
Corneal Tissue Engineering, College of Marine Life Sciences, Ocean University
of China, Qingdao 266003, Shandong Province, China. tjfan@ouc.edu.cn
Received:
Abstract
AIM: To reveal the cytotoxicity and related mechanisms of gatifloxacin (GFX)
to stromal fibroblasts (SFs) in vitro.
METHODS: SFs were treated with GFX at different
concentrations (0.009375%-0.3%), and their viability was detected by MTT
method. The cell morphology was observed using light/transmission electron
microscope. The plasma membrane permeability was measured by AO/EB
double-staining. Then cell cycle, phosphatidylserine (PS) externalization, and
mitochondrial transmembrane potential (MTP) were analyzed by flow cytometry.
DNA damage was analyzed by electrophoresis and immunostaining. ELISA was used
to evaluate the caspase-3/-8/-9 activation. Finally, Western blotting was applied
for detecting the expressions of apoptosis-related proteins.
RESULTS: Morphological changes and reduced viability of
GFX-treated SFs demonstrated that GFX above 0.009375% had cytotoxicity to SFs
with dependence of concentration and time. GFX-treating cells also showed G1
phase arrest, increased membrane permeability, PS externalization and DNA
damage, which indicated that GFX induced apoptosis of SFs. Additionally, GFX
could activate the caspase-8, caspase-9, and caspase-3, induce MTP disruption,
downregulate B-cell leukemia-2 (Bcl-2) and B-cell leukemia-XL (Bcl-XL), and
upregulate Bcl-2 assaciated X protein (Bax), Bcl-2-associated death promoter
(Bad), Bcl-2 interacting domain (Bid) and cytoplasmic cytochrome C in SFs,
suggesting that caspase-dependent extrinsic and intrinsic pathways were related
to GFX-contributed apoptosis of SFs.
CONCLUSION: The cytotoxicity of GFX induces apoptosis of SFs
through triggering the caspase-dependent extrinsic and intrinsic pathways.
KEYWORDS: gatifloxacin; stromal fibroblasts;
cytotoxicity; apoptosis; caspase; extrinsic pathway; intrinsic pathway
DOI:10.18240/ijo.2019.10.02
Citation: Xu
B, Sui YL, Fan TJ. Gatifloxacin inducing apoptosis of stromal
fibroblasts through cross-talk between caspase-dependent extrinsic and intrinsic
pathways. Int J Ophthalmol 2019;12(10):1524-1530
INTRODUCTION
Bacterial keratitis is a very common
ocular infection and the main reason causing ocular morbidity and blindness. Whereas,
approximately 72% of infections can be successfully cured topically with
appropriate antibiotics[1]. Fluoroquinolones, as
bactericidal antibiotics, are broadly applied for bacterial keratitis because
of their wide spectrum of activity and their effectiveness against multidrug
resistant organisms[2]. Fourth-generation products
of ophthalmic fluoroquinolones have been developed. Gatifloxacin (GFX; 2003),
one of newer fourth-generation fluoroquinolones, has been approved in eye
disease treatments in clinical. The most important feature of the
fourth-generation fluoroquinolones is their improved Gram-positive activity by
targeting both DNA gyrase and topoisomerase IV of bacterial cells in a balanced
fashion, different from the early-generation agents[3],
and other potentially beneficial characteristics include improved drug delivery
into the anterior segment of the eye, enhanced activity against certain strains
of atypical mycobacteria and lowered likelihood of selecting for resistant
bacterial strains[4].
For treatment of bacterial
keratitis, both efficacy and side-effect of antibiotics should be taken into
account[5]. Recently, GFX has been reported that
it affects the viability of corneal epithelial cells and corneal epithelial
wound healing[6-8]. Due to
ocular well penetration for fluoroquinolones[9-10], GFX might have negative effects on stromal
keratocytes[11-12]. GFX
displays the potential cytotoxic effects to keratocytes, depending on drug
concentrations and duration of exposure[13].
However, the cytotoxic mechanisms of GFX to keratocytes has not been fully
elucidated.
Moreover, keratocytes are quiescent and
can transform into repair-phenotype of activated stromal fibroblasts (SFs)
following injury and keratoplasty[14]. The
activated SFs become more susceptible to drug treatment. Unfortunately, few
attentions have been paid to the cytotoxic effects of GFX to SFs of the exposed
corneal stroma, especially after cornea surgery. Therefore, we established an
SF cell model in vitro using activated human corneal stromal (HCS) cells
cultured with fetal bovine serum (FBS)[15], to
explore the cytotoxicity of GFX and its potential mechanisms for prospective
therapeutic interventions in eye clinics[16-17].
MATERIALS AND METHODS
Cell Culture and Reagents SFs, an established non-transfected
HCS cell line[18], were cultured in DMEM/F12
medium (Gibco, Rockville, MD, USA) containing 10% FBS (Gibco) at
GFX Treatment SFs were seeded into various
specifications of culture plates (Nunc, Copenhagen, Denmark). After cells grew
about 70% confluence, the medium was replaced entirely with fresh medium
containing GFX at concentrations ranging from 0.009375% to 0.3%, respectively.
SFs were as blank controls in all experiments that were cultured in fresh 10%
FBS-DMEM/F12 medium without GFX. Cell morphology and growth status were
observed under an Eclipse TS100 inverted light microscope (Nikon, Tokyo, Japan)
per 4h.
MTT Assay MTT assay was performed to assess
cell viability of SFs as previously described[19].
Briefly, SFs were cultured in 96-well plates (2×104 cells per well)
and treated with GFX as described above. Then, 20 μL of 5 mg/mL MTT was added
into each well and incubated at
Transmission Electron Microscopy The ultrastructure of SFs was
obtained by transmission electron microscopy (TEM) as previously described[20]. In brief, the cells cultured in 6-well plates
(approximately 1.5×106 cells per well) were treated with 0.15% GFX
and harvested at 4h intervals. After successive fixation with 4% glutaraldehyde
and 1% osmium tetroxide, SFs were dehydrated and embedded in epoxy resin. After
staining with 2% uranyl acetate and lead citrate, ultrathin sections were
assessed by an H700 TEM (Hitachi, Tokyo, Japan).
AO/EB Double Staining AO/EB double-staining was conducted
to measure the plasma membrane permeability of SFs as previously reported[20]. In brief, SFs were seeded into 24-well plates
(approximately 1×105 cells per well), and treated with GFX as
described above. Cells were harvested after digestion with 0.25% trypsin and
centrifugation (
DNA Damage Detection The DNA fragmentation of SFs was
analyzed by Agarose gel electrophoresis as previously described[19] and modified. Briefly, SFs cultured in
6-well plates (approximately 1.5×106 cells per well) were treated
with GFX and collected as described above. After washing with ice-cold PBS and
centrifugation (
Flow Cytometry Analysis We further performed flow cytometry
(FCM) assay to analyze cell cycle, phosphatidylserine (PS) orientation, and
mitochondrial transmembrane potential (MTP), as previously reported[19]. Briefly, SFs cultured in 6-well plates
(approximately 1.5×106 cells per well) were treated and harvested
per 4h as described above, and fixed with cold 70% alcohol overnight at
ELISA Detection ELISA was used to measure caspase
activation in SFs as previously described[20] and
modified. Briefly, SFs inoculated in 6-well plates (approximately 1.5×106
cells per well) were treated and collected every 2h as described above.
Whole-cell protein were extracted using 500 µL of RIPA lysis buffer containing
PMSF following manufacturer’s instructions. The 100 µL protein extract was
coated onto a 96-wells ELISA plate (Nunc), followed by blocking with 5% non-fat
milk, incubated with 100 µL of rabbit anti-human caspase-3, -8, and -9 (active
forms) monoclonal antibodies (1:1000), respectively, at
Western Blot Analysis Western blotting was performed to
quantify expression levels of apoptosis-related proteins in GFX-treated SFs, as
previously described[21]. Briefly,
cells cultured in 6-well plate (approximately 1.5×106 cells per
well) were treated with GFX and harvested per 4h as described above. Total
proteins were extracted using RIPA lysis buffer as described above; cytoplasmic
proteins were extracted to analyze the mitochondrion-released
apoptosis-triggering proteins using the mitochondrial/cytoplasmic protein
extraction kit (Sangon biological engineering, Shanghai, China) following
manufacturer’s instructions. Equal amounts of protein were separated by 10%
SDS-PAGE, and transferred onto PVDF membranes. After blocking with 5% non-fat
milk, the membranes were incubated with rabbit anti-human IgG monoclonal
antibody targeting β-actin (1:5000), B-cell leukemia-2 (Bcl-2, 1:1000), B-cell
leukemia-XL (Bcl-XL, 1:500), Bcl-2 assaciated X protein (Bax, 1:2000),
Bcl-2-associated death promoter (Bad, 1:500), Bcl-2 interacting domain (Bid,
1:500) and cytochrome C (1:5000), respectively for 120min at
Statistical Analysis Each experiment was repeated three
times independently. Measurement data were expressed as mean±standard deviation
(SD) and were analyzed by one-way ANOVA and unpaired Dunnett’s t-test
assuming equal variance, with the SPSS 22.0 software (SPSS Inc., Chicago, IL,
USA). P values <0.05 was considered statistically significant.
RESULTS
GFX Exhibits Cytotoxicity to Stromal
Fibroblasts To investigate the cytotoxicity of
GFX to SF cell viability, firstly, we conducted MTT assay. The results showed
that the viability of SFs significantly declined with concentration and time
after treatment with GFX at concentrations of 0.009375% and above (P<0.01
or 0.05), while SFs exposed to 0.0046875% GFX had no significant difference
compared with that of blank control (Figure 1).
Figure 1 Reduced viability of
GFX-treated SFs MTT assay for SF cell viability
after exposure to GFX at different concentrations and different times. Cell
viability was presented as percentage (mean±SD) compared with that of the
corresponding blank control according to absorbance value at 490 nm (n=3).
aP<0.05 and bP<0.01 vs blank
control.
Then we observed the abnormal
alterations of growth status and morphology of SFs in different groups. The
growth inhibition and cytopathic morphological alterations (such as cellular
atrophy) were observed with dependence of concentration and time after
treatment with GFX at concentrations varying from 0.3% to 0.0375%, while there
was no significant difference between the 0.01875% GFX and blank control groups
(Figure
Figure 2 Morphological changes of
GFX-exposed SFs A: Light microscopy results showed GFX-induced
cellular atrophy and growth retardation. Scale bar, 50 μm; B: TEM showed
cytoplasmic vacuolation (v), swollen mitochondrion (m), condensed chromatin
(c), and suspected apoptotic bodies (asterisk) of 0.15% GFX-treated SFs. N:
Nucleus. Concentrations and exposure time points of GFX were showed at the
top-left of each panel. Scale bar, 1 μm.
GFX Induced G1 Phase
Arrest of Stromal Fibroblasts To investigate whether the GFX could
influence the cell cycle, we used FCM analysis. The results showed that the
cell numbers in G1 phase increased by approximately 28% (P<0.01)
at 4h, approximately 40% (P<0.01) at 8h, and approximately 52% (P<0.01)
at 12h, respectively, while obviously decreased in S and G2/M phases
(P<0.01 or 0.05) over time in 0.15% GFX-treated group, compared with
the corresponding blank control (Table 1).
Table 1 Cell cycle parameters of
0.15% GFX-treated SFs
%
Groups |
No. of
cells |
||
G1
phase |
S phase |
G2/M
phase |
|
0h |
50.75±1.04 |
26.06±0.46 |
23.19±0.61 |
Blank control |
|
|
|
4h |
40.93±0.23 |
27.93±1.12 |
31.14±1.12 |
8h |
33.01±0.74 |
30.93±1.18 |
36.07±0.90 |
12h |
29.52±0.20 |
31.62±0.76 |
38.87±0.88 |
0.15% GFX |
|
|
|
4h |
68.67±1.85b |
23.28± |
8.06±0.48b |
8h |
73.17±2.47b |
17.26±0.83b |
9.57±0.37b |
12h |
81.74±2.79b |
8.92±0.91b |
9.34±1.35b |
Assayed by flow cytometry by PI
staining. The cell numbers of each cell cycle phase were presented as
percentage (mean±SD) of the total number of cells (n=3). aP<0.05
and bP<0.01 vs blank control.
GFX Had Pro-apoptotic Effects on
Stromal Fibroblasts AO/EB double staining was conducted
to explore the toxicology mechanism of GFX to the SFs. And our results
indicated that 0.0375% GFX and above induced the increase of membrane permeability
with dependence of concentration and time (P<0.01 or 0.05) compared
with blank control. The apoptotic rates of SFs in all groups are indicated in
Figure
Figure 3 Membrane permeability, PS externalization
and DNA damage of GFX-treated SFs A: AO/EB
double staining. The apoptotic ratio of SFs was presented as percentage
(mean±SD) of total cell number according to the membrane permeability elevation
(n=3). B: Flow cytometry assay with Annexin V/PI staining. The number of
Annexin V-positive cells, i.e. PS externalized cells, was presented as
percentage (mean±SD) of total cell number (n=3). C: DNA electrophoresis.
Genomic DNA of cells treated with or without GFX was electrophoresed, and DNA
bands were shown; D: 0.15% GFX-treated SFs showed immunostaining patterns for
γ-H
GFX Induced Caspase-dependent
Apoptosis of Stromal Fibroblasts To investigate the involved apoptosis pathway inducing
by GFX, we carried out the ELISA assay. As shown in Figure 4, caspase-8 was
significantly activated from 6 to 14h (P<0.01 or 0.05), caspase-9
from 2 to 14h (P<0.01 or 0.05), and caspase-3 from 4 to 14h (P<0.01
or 0.05) in SFs treated with 0.15% GFX, compared with that of blank control,
respectively, in which caspase-8 activation rate peaked at 10h (P<0.01),
caspase-9 at 6h (P<0.01) and caspase-3 at 10h (P<0.01).
Figure 4 Caspase activation of 0.15%
GFX-treated SFs The activation ratios of caspase-3, -8
and -9 were presented as percentage (mean±SD) compared with the corresponding
blank control based on 490 nm absorbance (n=3). aP<0.05
and bP<0.01 vs blank control.
GFX Induced MTP Disruption and Altered
Expressions of Apoptosis-related Proteins
FCM results suggested that GFX caused a time-dependent MTP
disruption of SFs, and the amounts of JC-1 positive cells increased by
approximately 17% (P<0.01), approximately 27% (P<0.01), and
approximately 53% (P<0.01) after exposure to 0.15% GFX for 4, 8, and
12h, compared with that of blank control, respectively (Figure
Figure 5 MTP disruption and
expression alteration of apoptosis-triggering proteins of 0.15% GFX-treated SFs A: Flow cytometry assay with JC-1 staining.
The amounts of JC-1 positive cells, i.e. MTP disrupted cells, were
presented as percentage (mean±SD) of total cell number (n=3). B: Western
blot images. Alterations of expression levels of Bcl-2 family proteins along
with the cytoplasmic amounts of mitochondria-released cytochrome C are shown.
C: Relative densitometric analysis of protein bands by comparing with the
expression of β-actin. The relative amounts of each protein were presented as
the percentage (mean±SD) of protein band density compared to the corresponding
internal control (n=3). Cyt. c: Cytochrome C. bP<0.01
vs blank control.
DISCUSSION
GFX eye drop is one of most common
drugs for treating bacterial keratitis, pre- and postoperative control of
infection during keratoplasty[9]. Unfortunately,
there are some reports that claimed it showed toxicity to corneal cells[7-8,13,22],
and its cytotoxic effects remains unclear. So, in this study, we investigated
the cytotoxicity of GFX to SFs and its possible mechanisms in vitro to
provide a therapeutic intervention for bacterial keratitis.
The cell viability and morphology are
main indices for cytotoxicity evaluation of chemicals and drugs[15,20]. GFX obviously decreased the
viability of SFs with dependence of concentration and time, and also caused
dramatic morphological changes of SFs, including cell shrinkage, mitochondrion
swelling, chromatin condensation and suspected apoptotic bodies, basically in
accordance with the results of cell viability. The results demonstrate that GFX
has significant cytotoxicity to SFs at concentrations above 0.009375%.
Moreover, cell cycle analysis implies that GFX might induce the growth
inhibition of SFs by causing G1 phase arrest. Based on the
cytotoxicity and G1 phase arrest, we consider that GFX may have
pro-apoptotic effects on SFs, which is accordance with previous studies[23-24].
Then the GFX-induced apoptosis was
assessed in SFs. As shown, GFX-treated SFs exhibited the typical
characteristics of cell apoptosis, including membrane permeability, PS
externalization, and DNA ladder[21,25].
Our findings imply that GFX contributes to cytotoxicity most probably by
inducing apoptosis[23,26].
Based on above findings, subsequent experiments were conducted to further
confirm underlying apoptotic pathways including extrinsic and intrinsic
pathways.
The extrinsic pathway is mediated by
death receptors that cause the autocleavage of pro-caspase-8[27].
The cleaved caspase-8 as a major initiator caspase can activate downstream
effector caspase-3 that is known as a key executor caspase in the apoptosis pathways.
After activation of caspase-3, PARP, as the substrate of caspase-3, is cleaved
into two fragments, p89 and p24, contributing to DNA fragmentation, resulting
in the execution of extrinsic apoptosis[28-29]. Our findings showed that GFX could activate
caspase-8 and caspase-3, suggesting that the extrinsic pathway was involved in
the apoptosis of SFs. However, further research is needed to confirm which
death receptor mediates the GFX-induced apoptosis of SFs.
Bcl-2 family, classified as
anti-apoptotic and pro-apoptotic members, plays an important role in the
regulation of the initiation of intrinsic (mitochondrial-mediated) apoptosis[30-31]. The anti-apoptotic subfamily,
including Bcl-2, Bcl-XL, etc., prevents the release of
mitochondria-sequestered pro-apoptotic regulators into cytoplasm, while the
pro-apoptotic subfamily, such as Bax, Bad, etc. induces pro-apoptotic
regulators release into the cytoplasm[32-33].
Moreover, due to MTP disrupting, cytochrome C is released from mitochondria
into cytoplasm, which activates key initiator caspase-9 and subsequent
downstream effector caspases-3, leading to intrinsic apoptosis[34]. Our results demonstrated that GFX could induce the
upregulation of Bax and Bad as well as the downregulation of Bcl-2 and Bcl-XL
of SFs. The rising ratio of pro-apoptotic proteins induced MTP disruption. And
GFX also increased the cytochrome C amounts in cytoplasm, which activated
caspase-9 and caspase-3, resulting in executing intrinsic apoptosis.
Additionally, active caspase-8 can
also induce activation of Bid, a member of Bcl-2 family. The truncated Bid (tBid,
active Bid) binds to the pro-apoptotic protein Bax, causing the MTP distruption
and the release of cytochrome C[35]. This result
was also found in this study, suggesting that the extrinsic pathway
interconnects with the intrinsic pathway, ultimately resulting in caspase-3
activation in the process of apoptosis induction.
GFX shows cytotoxicity to SFs with
dependence of concentration and time, and induces apoptosis through cross-talk
between the caspase-dependent extrinsic and intrinsic pathways. Our findings
shine a light on the cytotoxicity and mechanisms of GFX, and provide relevant
references for prospective therapeutic interventions in eye clinics.
ACKNOWLEDGEMENTS
The authors would like to thank Prof.
Ming-Zhuang Zhu of Key Laboratory of Mariculture, Ocean University of China,
for help and guidance in flow cytometry analysis.
Foundation: Supported by the National High
Technology R&D Program of China (No.2006AA
Conflicts of Interest: Xu B, None; Sui YL, None; Fan
TJ, None.
REFERENCES