ArticlePDF Available

RETRACTED ARTICLE: TNFα promotes glioblastoma A172 cell mitochondrial apoptosis via augmenting mitochondrial fission and repression of MAPK–ERK–YAP signaling pathways

Taylor & Francis
OncoTargets and Therapy
Authors:

Abstract and Figures

Lu C, Chen X, Wang Q, Xu X, Xu B. Onco Targets Ther. 2018;11:7213–7227. We, the Editors and Publisher of OncoTargets and Therapy, have retracted the following article. Following publication of the article, concerns were raised about the duplication of images from Figures 1, 2, 3, 4, 5, and 6 with images from other unrelated articles. Specifically, Images for Figure 1E have been duplicated with images for Figure 2a from Li X, Wu M, An D, et al. Suppression of Tafazzin promotes thyroid cancer apoptosis via activating the JNK signaling pathway and enhancing INF2-mediated mitochondrial fission. J Cell Physiol. 2019;234:16238–16251. https://doi.org/10.1002/jcp.28287 and 5I from Jieensinue S, Zhu H, Li G, et al. Tanshinone IIA reduces SW837 colorectal cancer cell viability via the promotion of mitochondrial fission by activating JNK-Mff signaling pathways. BMC Cell Biol. 2018;19:21. https://doi.org/10.1186/s12860-018-0174-z. Images for Figure 2A have been duplicated with images for Figure 5H from Li R, Xin T, Li D, et al. Therapeutic effect of Sirtuin 3 on ameliorating nonalcoholic fatty liver disease: The role of the ERK-CREB pathway and Bnip3-mediated mitophagy. Redox Biology. 2018;18:229–243. https://doi.org/10.1016/j.redox.2018.07.011. Images for Figure 3G have been duplicated with images for Figures 4A and 5C from Yu W, Xu M, Zhang T, et al. Mst1 promotes cardiac ischemia–reperfusion injury by inhibiting the ERK-CREB pathway and repressing FUNDC1-mediated mitophagy. J Physiol Sci. 2019;69:113–127. https://doi.org/10.1007/s12576-018-0627-3. Images for Figure 4A have been duplicated with images for Figure 2C from Zheng Z, Xiang S, Wang Y, et al. NR4A1 promotes TNF‑α‑induced chondrocyte death and migration injury via activating the AMPK/Drp1/mitochondrial fission pathway. International Journal of Molecular Medicine. 2020;45:151–161. https://doi.org/10.3892/ijmm.2019.4398. Images for Figure 4D have been duplicated with images for Figure 4K from Qin R, Zhang L, Lin D, Xiao F, Guo L. Sirt1 inhibits HG-induced endothelial injury: Role of Mff-based mitochondrial fission and F‑actin homeostasis-mediated cellular migration. International Journal of Molecular Medicine. 2019;44:89–102. https://doi.org/10.3892/ijmm.2019.4185. Images for Figure 5D have been duplicated with images for Figure 7d from Wang Q, Xu J, Li X, et al. Sirt3 modulate renal ischemia-reperfusion injury through enhancing mitochondrial fusion and activating the ERK-OPA1 signaling pathway. J Cell Physiol. 2019;234:23495–23506. https://doi.org/10.1002/jcp.28918. Images for Figure 5H have been duplicated with images for Figure 2A from Xie Y, Lv Y, Zhang Y, et al. LATS2 promotes apoptosis in non-small cell lung cancer A549 cells via triggering Mff-dependent mitochondrial fission and activating the JNK signaling pathway. Biomedicine & Pharmacotherapy. 2019;109:679–689. https://doi.org/10.1016/j.biopha.2018.10.097; Figure 3A from Ouyang H, Zhou E, Wang H. RETRACTED ARTICLE: Mst1-Hippo pathway triggers breast cancer apoptosis via inducing mitochondrial fragmentation in a manner dependent on JNK–Drp1 axis. Onco Targets Ther. 2019;12:1147–1159. https://doi.org/10.2147/OTT.S193787 and Figure 5H from Li R, Xin T, Li D, et al (2018). Images for Figure 6A have been duplicated with images for Figure 2A from Li J, Li N, Yan S, et al. Liraglutide protects renal mesangial cells against hyperglycemia‑mediated mitochondrial apoptosis by activating the ERK‑Yap signaling pathway and upregulating Sirt3 expression. Molecular Medicine Reports. 2019;19:2849–2860. https://doi.org/10.3892/mmr.2019.9946. Images for Figure 6C have been duplicated with images for Figure 3I from Zhang W, Liu K, Pei Y, Ma J, Tan J, Zhao J. Mst1 regulates non-small cell lung cancer A549 cell apoptosis by inducing mitochondrial damage via ROCK1/F‑actin pathways. International Journal of Oncology. 2018;53:2409–2422. https://doi.org/10.3892/ijo.2018.4586; Figure 3g from Qin R, Lin D, Zhang L, Xiao F, Guo L. Mst1 deletion reduces hyperglycemia-mediated vascular dysfunction via attenuating mitochondrial fission and modulating the JNK signaling pathway. J Cell Physiol. 2019;235:294–303. https://doi.org/10.1002/jcp.28969; Figure 5D from Xu P, Zhang G, Sha L, Hou S. RETRACTED: DUSP1 alleviates cerebral ischaemia reperfusion injury via inactivating JNKMff pathways and repressing mitochondrial fission. Life Sciences. 2018;210:251–262. https://doi.org/10.1016/j.lfs.2018.08.049 and Figure 5g from Li X, et al (2019). Images for Figure 6F have been duplicated with images for Figure 1d from Zhang Y, Wang M, Xu X, Liu Y, Xiao C. Matrine promotes apoptosis in SW480 colorectal cancer cells via elevating MIEF1-related mitochondrial division in a manner dependent on LATS2-Hippo pathway. J Cell Physiol. 2019;234:22731–22741. https://doi.org/10.1002/jcp.28838 and Figure 1I from Zhang L, Li S, Wang R, et al. Anti-tumor effect of LATS2 on liver cancer death: Role of DRP1-mediated mitochondrial division and the Wnt/β-catenin pathway. Biomedicine & Pharmacotherapy. 2019;114:108825. https://doi.org/10.1016/j.biopha.2019.108825. The authors did not respond to our queries and were unable to provide an explanation for the duplicated images or provide data for the study. As verifying the validity of published work is core to the integrity of the scholarly record, we are therefore retracting the article and the authors were notified of this. We have been informed in our decision-making by our editorial policies and COPE guidelines. The retracted article will remain online to maintain the scholarly record, but it will be digitally watermarked on each page as “Retracted”.
This content is subject to copyright. Terms and conditions apply.
© 2018 Lu et al. This work is published and licensed by Dove Medical Press Limited. The full terms of this license are available at https://www.dovepress.com/terms.php
and incorporate the Creative Commons Attribution – Non Commercial (unported, v3.0) License (http://creativecommons.org/licenses/by-nc/3.0/). By accessing the work you
hereby accept the Terms. Non-commercial uses of the work are permitted without any further permission from Dove Medical Press Limited, provided the work is properly attributed. For permission
for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms (https://www.dovepress.com/terms.php).
OncoTargets and Therapy 2018:11 7213–7227
OncoTargets and erapy Dovepress
submit your manuscript | www.dovepress.com
Dovepress 7213
ORIGINAL RESEARCH
open access to scientific and medical research
Open Access Full Text Article
http://dx.doi.org/10.2147/OTT.S184337
Changyu Lu
Xiaolei Chen
Qun Wang
Xinghua Xu
Bainan Xu
Department of Neurosurgery,
Chinese PLA General Hospital,
Beijing 100853, China
Background and objective: The present study was designed to explore the roles of
mitochondrial fission and MAPK–ERK–YAP signaling pathways and to determine their
mutual relationship in TNFα-mediated glioblastoma mitochondrial apoptosis.
Materials and methods: Cellular viability was measured via TUNEL staining, MTT assays,
and Western blot. Immunofluorescence was performed to observe mitochondrial fission. YAP
overexpression assays were conducted to observe the regulatory mechanisms of MAPK–ERK–
YAP signaling pathways in mitochondrial fission and glioblastoma mitochondrial apoptosis.
Results: The results in our present study indicated that TNFα treatment dose dependently
increased the apoptotic rate of glioblastoma cells. Functional studies confirmed that TNFα-
induced glioblastoma apoptosis was attributable to increased mitochondrial fission. Excessive
mitochondrial fission promoted mitochondrial dysfunction, as evidenced by decreased mito-
chondrial potential, repressed ATP metabolism, elevated ROS synthesis, and downregulated
antioxidant factors. In addition, the fragmented mitochondria liberated cyt-c into the cytoplasm/
nucleus where it activated a caspase-9-involved mitochondrial apoptosis pathway. Furthermore,
our data identified MAPK–ERK–YAP signaling pathways as the primary molecular mechanisms
by which TNFα modulated mitochondrial fission and glioblastoma apoptosis. Reactivation of
MAPK–ERK–YAP signaling pathways via overexpression of YAP neutralized the cytotoxicity
of TNFα, attenuated mitochondrial fission, and favored glioblastoma cell survival.
Conclusion: Overall, our data highlight that TNFα-mediated glioblastoma apoptosis stems
from increased mitochondrial fission and inactive MAPK–ERK–YAP signaling pathways, which
provide potential targets for new therapies against glioblastoma.
Keywords: glioblastoma, apoptosis, mitochondrion, TNFα, mitochondrial fission, MAPK-
ERK-YAP signaling pathways
Introduction
Although glioblastoma multiforme (GBM) is a rare tumor whose incidence is less than
3.19/100,000 in the population globally, its poor prognosis with a median survival
of 15 months and inevitable recurrence after a median survival time of 32–36 weeks
make it a heavy burden on the health care system. Unfortunately, little is known about
the etiology of GBM, although several risk factors have been proposed, such as age,
exposure to radiation, and family history. Notably, excessive hyperplasia of glial cells
is the primary pathogenesis of GBM.1 Accordingly, several approaches have been
attempted to induce the death of glial cells, especially TNFα-based therapy.
Correspondence: Bainan Xu
Department of Neurosurgery,
Chinese PLA General Hospital,
28 Fuxing Road, Beijing 100853, China
Tel +86 138 0127 1711
Email dr_lcy@126.com
Journal name: OncoTargets and Therapy
Article Designation: Original Research
Year: 2018
Volume: 11
Running head verso: Lu et al
Running head recto: TNFα promotes glioblastoma A172 cell
DOI: 184337
RETRACTED ARTICLE: TNFα promotes
glioblastoma A172 cell mitochondrial apoptosis via
augmenting mitochondrial fission and repression of
MAPK–ERK–YAP signaling pathways
RETRACTED
This article was published in the following Dove Press journal:
OncoTargets and Therapy
OncoTargets and Therapy 2018:11
submit your manuscript | www.dovepress.com
Dovepress
Dovepress
7214
Lu et al
A gene delivery strategy to induce TNFα overexpres-
sion has been attempted to increase the apoptotic index of
glioblastoma cells.2 The effectiveness of the TNFα-based
therapy is later validated by several clinical studies.3 Ample
in vivo and in vitro evidence potentially implies that TNFα
considerably augments the apoptosis of glioblastoma cells.4
This information indicates that TNFα-based therapy is a
promising tool for the treatment of glioblastoma. However,
the molecular mechanisms of TNFα involved in glioblastoma
cell death have not been fully described.
Mitochondria control an array of subcellular functions,
such as energy metabolism, ROS production, cell prolif-
eration, calcium balance, and cell death.5,6 Previous studies
have provided molecular insight into the mitochondrial
etiology in GBM and have identified mitochondria as a
potentially therapeutic target to modulate the growth of
gliomas.7 In addition, TNFα-based therapy has been linked
to mitochondrial dysfunction in GBM. For example, TNFα
promotes mitochondrial oxidative stress via the JNK–
NF–κB pathways.8 Some researchers have demonstrated
that TNFα induces mitochondrial apoptosis via increas-
ing tBid stability.9 In addition, other studies suggest that
Bnip3-related mitochondrial necrotic death is activated by
TNFα.10 This information indicates that TNFα potentially
targets mitochondria in glioblastoma cells. Recently, mito-
chondrial fission has been thought to be the early feature
of mitochondrial abnormalities and to promote the death
of several kinds of tumors, such as breast cancer,11 ovarian
cancer,12 pancreatic cancer,13 and bladder cancer.14 TNFα has
been found to be associated with Drp1 activation during the
inflammation-mediated cardiomyocyte injury.15 However, no
studies have investigated the role of mitochondrial fission in
TNFα-treated glioblastoma cells. In the present study, we ask
whether mitochondrial fission is required for TNFα-mediated
mitochondrial apoptosis in glioblastoma cells.
The MAPK–ERK signaling pathway has been found to
be the upstream inhibitor of mitochondrial fission. In liver
cancer, defective ERK signaling upregulates FAK expression
and the latter promotes mitochondrial fission.16 Moreover,
in neuroblastoma N2a cells, increased ERK signaling inhib-
its mitochondrial fission and sustains cellular viability.17
Furthermore, in-depth studies have indicated that ERK
modulates mitochondrial fission via YAP. Increased YAP
suppresses mitochondrial fission in human rectal cancer,18
cerebral ischemia-reperfusion injury,19 and dendritic cells.20
These findings uncover the critical role played by ERK–YAP
signaling in inhibiting mitochondrial fission. Considering
that ERK is also the classical antiapoptotic signal for cancer,21
we ask whether TNFα handles mitochondrial fission via
repressing the MAPK–ERK–YAP signaling pathways. Alto-
gether, the aim of our study was to investigate the therapeutic
effects of TNFα on glioblastoma cells and determine its
influence on mitochondrial fission and the MAPK–ERK–
YAP signaling pathways.
Materials and methods
Cell culture and treatment
Human glioblastoma cell line A172 (ATCC® CRL 1620™)
was purchased from American Type Culture Collection.
These cells were cultured with l-DMEM supplemented
with 10% FBS (Biowest, Mexico City, Mexico, USA) and
1% penicillin/streptomycin in a humidified atmosphere with
5% CO2 at 37°C. Different doses of TNFα were added to the
medium of A172 cells for 12 hours to induce cell damage
(0–20 ng/mL). This concentration of TNFα was chosen
according to a previous study.22 Cells were exposed to
10 mM mitochondrial division inhibitor-1 (Mdivi-1; Sigma-
Aldrich Co., St Louis, MO, USA; EMD Millipore, Billerica,
MA, USA) to inhibit the activity of mitochondrial fission.
In contrast, to activate mitochondrial fission, 5 µm FCCP
(Selleck Chemicals, Houston, TX, USA) was pretreated for
40 minutes at 37°C in a 5% CO2 atmosphere.23
MTT assay, TUNEL staining, and LDH
release assay
The cell viability was determined by MTT assays (Sigma-
Aldrich Co.). Briefly, cells were seeded onto 96-well plates,
and then 20 µL of MTT at a concentration of 5 mg/mL was
added to the medium. The plates were placed for 4 hours in
the dark at 37°C and 5% CO2. After that, the medium was
removed and 100 µL of dimethyl sulfoxide (DMSO) was
added into the medium for 15 minutes in the dark at 37°C and
5% CO2. Then, the samples were observed at a wavelength of
570 nm. The relative cell viability was recorded as a ratio to
that of the control group. Apoptotic cells were quantified using
a one-step TUNEL kit (Beyotime Institute of Biotechnology,
Haimen, China) according to the manufacturer’s instructions.24
Cells were seeded onto the 12-well plates and incubated with
fluorescein–dUTP (Beyotime Institute of Biotechnology) for
30 minutes at 37°C in a 5% CO2 atmosphere. After being
labeled with DAPI, the cells were observed using a laser confo-
cal microscope (TcS SP5; Leica Microsystems, Inc., Buffalo
Grove, IL, USA). LDH was released into the medium when
cellular membranes ruptured. To evaluate the levels of LDH in
the medium, an LDH Release Detection kit (Beyotime Institute
of Biotechnology) was used according to manufacturer’s proto-
col. Cells treated with PBS were used as the control group for
MTT assay, LDH release assay, and TUNEL staining.
RETRACTED
OncoTargets and Therapy 2018:11 submit your manuscript | www.dovepress.com
Dovepress
Dovepress
7215
TNFα promotes glioblastoma A172 cell
Measurement of mitochondrial
membrane potential and mitochondrial
permeability transition pore (mPTP)
opening rate
Mitochondrial potential was evaluated using 5,5,6,6-
tetrachloro-1,1,3,3-tetraethyl-benzimidazolylcarbocyanine
chloride (JC-1) staining. Cells were seeded onto 12-well
plates. After washing with PBS three times, the cells were
treated with the JC-1 probe for 30 minutes in the dark at
37°C and 5% CO2. Then, cells were washed with PBS three
times to remove the free JC-1. After replaced with fresh
DMEM, the cells were observed using a laser confocal
microscope (TcS SP5). At least 30 cells were randomly
chosen.25 To measure the mPTP opening, cells were loaded
with PBS containing 25 nM tetramethylrhodamine, methyl
ester (TMRM, T668; Thermo Fisher Scientific, Waltham,
MA, USA). After 30 minutes, cells were washed with
PBS again to remove the free TMRM. Then, samples were
observed at a wavelength of 480 nm using a microplate
reader (Epoch 2; BioTek Instruments, Inc., Tokyo, Japan).
Cells treated with PBS were used as the control group for
MTT assay and TUNEL staining.
Western blots
Total proteins were extracted using RIPA Lysis Buffer
(Cat. No: P0013E; Beyotime, Beijing, China). After that,
proteins were rapidly centrifuged (20,000 rpm) for 10 min
at 4°C to pellet cell debris. Supernatant was collected and
quantified using an Enhanced BCA Protein Assay Kit
(Beyotime, Cat. No: P0009). Then, proteins (45–60 µg)
were loaded in a 10%–15% SDS-PAGE gel and transferred
to PVDF membranes (Bio-Rad, Hercules, CA, USA). Sub-
sequently, membranes were blocked with 5% skim milk
for 45 minutes at room temperature. After washing with
tris buffered saline with tween 20 (TBST) three times at
room temperature, the membranes were incubated with
the primary antibodies at 4°C overnight.26 After washing,
horseradish peroxidase-conjugated secondary antibodies
were incubated with membranes for 50 minutes at room
temperature. Then, the bands were observed using an ECL
Prime Western Blotting Detection Reagent (GE Healthcare,
Buckinghamshire, UK). The primary antibodies used in the
present study are described as follows: p-ERK (1:1,000,
#ab176660; Abcam, Cambridge, MA, USA), t-ERK
(1:1,000, #ab54230; Abcam), Yap (1:1,000; #14074; Cell
Signaling Technology, Danvers, MA, USA), complex III
subunit core (CIII-core2, 1:1,000, #459220; Invitrogen,
Merck KGaA, Darmstadt, Germany), complex II (CII-30,
1:1,000, #ab110410; Abcam), complex IV subunit II
(CIV-II, 1:1,000, #ab110268; Abcam), Drp1 (1:1,000,
#ab56788; Abcam), Fis1 (1:1,000, #ab71498; Abcam),
Opa1 (1:1,000, #ab42364; Abcam), Mfn2 (1:1,000,
#ab57602; Abcam), Mff (1:1,000, #86668; Cell Signaling
Technology), Tom20 (1:1,000, #ab186735; Abcam), Bcl-2
(1:1,000, #3498; Cell Signaling Technology), Bax (1:1,000,
#2772; Cell Signaling Technology), Bcl-2 (1:1,000, #3498;
Cell Signaling Technology), Bad (1:1,000, #ab90435;
Abcam), and x-IAP (1:1,000, #ab28151; Abcam).
Immunouorescence
Cells were seeded onto poly-d-lysine-coated coverslips.
Then, methanol-free 4% paraformaldehyde was used
to fix cells for 15 minutes at room temperature. Subse-
quently, samples were blocked with 5% goat serum at
room temperature for 45 minutes. After washing with
TBST, samples were incubated with primary antibody at
4°C overnight. The primary antibodies used in the present
study were as follows: p-ERK (1:1,000, #ab176660), Yap
(1:1,000, #14074), Tom20 (1:1,000, #ab186735), and cyt-c
(1:1,000, #ab90529; Abcam). Subsequently, samples were
rinsed three times with TBST for 15 minutes and followed
by incubation with secondary antibody for 45 minutes at
room temperature; after rinsing three times for 5 minutes
using TBST, the samples were labeled with DAPI to tag
the nuclei. Cells were observed using a laser confocal
microscope (TcS SP5).27
Transfection
The pDC315–YAP vector was designed and purchased
from Vigene Biosciences, Inc. (Rockville, MD, USA).
Then, the plasmid was transfected in 293 T cells using
Lipofectamine 2000®. After 48 hours, the supernatant was
collected and amplified to obtain adenovirus-YAP (Ad-YAP).
Subsequently, A172 cells were infected with Ad-YAP using
Lipofectamine 2000® for 6 hours at 37°C and 5% CO2.
Western blot was performed to observe the overexpression
efficiency.28
ROS and antioxidant factors
quantication
ROS generation was quantified using flow cytometry. Cells
were seeded onto the 12-well plates. After washing with
PBS, dihydroethidium (DHE) staining was added into the
medium and the cells were incubated with the DHE probe
for 30 minutes in the dark at 37°C and 5% CO2. Then, PBS
was used to wash cells to remove the free DHE probe. Sub-
sequently, 0.25% trypsin was applied to collect the cell. Flow
RETRACTED
OncoTargets and Therapy 2018:11
submit your manuscript | www.dovepress.com
Dovepress
Dovepress
7216
Lu et al
cytometry analysis was performed using the BD FACSCanto
II cytometer (BD, San Diego, CA, USA). Analysis of the data
was performed using FACSDiva software (BD). Besides, the
ROS production was also observed using a laser confocal
microscope (TcS SP5). The concentration of cellular anti-
oxidant factors such as GSH (Glutathione Reductase Assay
Kit, Cat. No: S0055; Beyotime), SOD (Total Superoxide
Dismutase Assay Kit, Cat. No: S0101; Beyotime), and GPX
(Cellular Glutathione Peroxidase Assay Kit, Cat. No: S0056;
Beyotime) was measured via ELISA according to the manu-
facture’s guidelines.29 Cells treated with PBS were used as
the control group for MTT assay and TUNEL staining.
Caspase-3/9 activities and Trypan Blue
staining
Caspase-3 and caspase-9 activities were measured using the
Caspase-3 Activity Assay Kit (Cat. No: C1115; Beyotime)
and Caspase-9 Activity Assay Kit (Cat. No: C1158; Beyo-
time) following the manufacturer’s instructions.30 Briefly,
cells were seeded onto 96-well plates. Then, 100 µL of
caspase-3 and caspase-9 reagents were added to each sample.
After incubation for 30 minutes in the dark at 37°C and 5%
CO2, the samples were measured at a wavelength of 570 nm
using the microplate reader (Epoch 2). The relative caspase
activity was recorded as the ratio to that of the control group.
Trypan Blue staining was conducted using 0.4% Trypan Blue
probe, which was treated with cells for 2 minutes. Then,
the number of Trypan Blue-positive cells was calculated by
counting at least three random separate fields.
Cellular ATP level detection
Cellular ATP levels were measured using the Enhanced
ATP Assay Kit (Cat. No: S0027; Beyotime) following the
supplier’s specifications.31 Briefly, cells were seeded onto
96-well plates at a density of 1×104/well. Subsequently,
100 µL of staining solution (Enhanced ATP Assay Kit, Cat.
No: S0027) was added to each well and incubated with the
cells for 4 hours in the dark at 37°C and 5% CO2. The rela-
tive ATP production was recorded using a microplate reader
(Epoch 2) at a wavelength of 570 nm.
Statistical data analyses
The results are presented as the mean±standard error (SE)
from at least three independent experiments using SPSS
16.0 software (SPSS Inc., Chicago, IL, USA). One-way
ANOVAs were carried out for comparisons between control
and treated groups. Pairwise comparisons were made by post
hoc Tukey’s test. Differences were considered as significant
at P,0.05.
Results
TNFα dose-dependently promotes
glioblastoma cell apoptosis in vitro
In the present study, glioblastoma cells were incubated
with different doses of TNFα for 12 hours. Then, the cel-
lular viability was measured via MTT assay. As shown in
Figure 1A, with increasing concentrations of TNFα, the
viability of glioblastoma cells decreased progressively.
Reduction in cellular viability may result from cell death.
To analyze the cellular death rate, the LDH release assay
was performed. Compared to the control group, TNFα dose
dependently elevated the content of LDH in the medium
(Figure 1B), indicating that TNFα promoted glioblastoma
cell death. This finding was further supported via Trypan Blue
and TUNEL staining, which exhibited an increased number
of Trypan-positive (Figure 1C and D) and TUNEL-positive
cells (Figure 1E and F) in the presence of TNFα stress.
At the molecular levels, cell death is primarily executed via
caspase-3 activation, which cleaves DNA into fragments.
Accordingly, caspase-3 activity was measured, and the
results shown in Figure 1G illustrate that caspase-3 activity
was drastically increased with the rise in TNFα. Altogether,
our data indicate that TNFα treatment dose dependently pro-
motes glioblastoma cell apoptosis. Notably, no significant
difference was observed between the control group and the
1 ng/mL TNFα group. The minimal proapoptotic dose of
TNFα is 5 ng/mL; accordingly, 5 ng/mL TNFα was used in
the following studies.
Mitochondrial ssion is activated by
TNFα treatment
Several thorough studies from many laboratories have
reported that mitochondrial fission is an early event leading
to cell death.13 In the present study, we explored the functional
role of TNFα in mitochondrial fission. The immunofluores-
cence assay in Figure 2A demonstrated that mitochondria
are highly connected networks. However, after TNFα
treatment, mitochondria become small, roundish fragments
that are characteristic of mitochondrial fission. To quantify
mitochondrial fission, we measured the average length of
mitochondria with or without TNFα treatment. In the control
group, the length of mitochondria was ~8.9 µm. Interest-
ingly, TNFα treatment (5 ng/mL) reduced the mitochondrial
length to ~2.3 µm (Figure 2B). This information indicated
RETRACTED
OncoTargets and Therapy 2018:11 submit your manuscript | www.dovepress.com
Dovepress
Dovepress
7217
TNFα promotes glioblastoma A172 cell
that mitochondrial fission was activated by TNFα treatment
in glioblastoma cells. To provide additional evidence for the
role of TNFα in triggering mitochondrial division, Mdivi-1,
an antagonist of mitochondrial division was used. Mean-
while, a mitochondrial fission agonist was administered to the
normal glioblastoma cells to activate mitochondrial fission,
which was used as the positive control group. Then, Western
blot was performed to analyze alterations in protein levels
related to mitochondrial fission.32,33 When compared to the
control group, TNFα treatment increased the levels of Drp1,
Mff, and Fis1, the key elements in executing mitochondrial
fission (Figure 2C–H). In contrast, inhibitors of mitochondrial
fission such as Mfn2 and Opa1 were significantly downregu-
lated in response to TNFα treatment (Figure 2C–H). This
effect of TNFα was similar to the action of FCCP, which
caused an imbalance between mitochondrial fission factors
(Figure 2C–H). Interestingly, Mdivi-1 could abrogate the
promotive effects of TNFα on mitochondrial fission-related
proteins (Figure 2C–H). Altogether, our data confirm that
TNFα promotes mitochondrial fission activation in glio-
blastoma cells.
TNFα-mediated mitochondrial ssion
promotes mitochondrial dysfunction
Abnormal mitochondrial fission plays a decisive role in medi-
ating mitochondrial dysfunction. To verify whether TNFα
induces mitochondrial damage in glioblastoma cells via
mitochondrial fission, mitochondrial function was measured.
First, cell ROS production was determined via flow cytom-
etry. When compared to the control group, TNFα treatment
significantly increased ROS production in glioblastoma cells
(Figure 3A and B), and this effect was similar to the results
obtained via administering FCCP (Figure 3A and B). Interest-
ingly, TNFα-mediated ROS production was mostly negated
by Mdivi-1 (Figure 3A and B). Because of the cellular ROS
outburst, the concentration of cellular antioxidants such as
Figure 1 TNFα promotes glioblastoma cell apoptosis in a dose-dependent manner.
Notes: (A) Different doses of TNFα were added into the media of glioblastoma cells, and then, the cellular viability was measured via MTT assay. (B) An LDH release assay
was performed to detect cell death. (C and D) Trypan Blue staining for cell death. The number of Trypan Blue-positive cell was recorded. (E and F) A TUNEL assay was
used to determine the rate of apoptosis. The number of TUNEL-positive cells was measured. (G) Caspase-3 activity was measured to determine the activation level of the
caspase-3 protein. *P,0.05 vs the control group. The 0 ng/mL TNFα was used as the control group.
077DVVD\
71)αQJP/
$








 
/'+UHOHDVH
IROGV
71)αQJP/
%








 
7U\SDQ%OXH
SRVLWLYHFHOOV
71)αQJP/
&








 
781(/
SRVLWLYHFHOOV
71)αQJP/
)








 
&DVSDVHDFWLYLW\
IROGV
71)αQJP/
*






 
&RQWURO
71)αQJP/
'
7U\SDQ%OXH
 
P
71)αQJP/
(
781(/
0HUJHG
P
&RQWURO  
RETRACTED
OncoTargets and Therapy 2018:11
submit your manuscript | www.dovepress.com
Dovepress
Dovepress
7218
Lu et al
Figure 2 TNFα activates mitochondrial ssion in glioblastoma cells.
Notes: (A) An immunouorescence assay for mitochondria using mitochondrial specic antibody Tom20. (B) The average length of mitochondria was measured, which was
used to analyze the extent of mitochondrial ssion. (CH) Western blot was performed to analyze protein expression of mitochondrial ssion-related factors. To perform
the loss- and gain-of-function assays for mitochondrial ssion, Mdivi-1, a pharmacological antagonist was used in TNFα-treated cells to inhibit the activation of mitochondrial
ssion. FCCP, an agonist for mitochondrial ssion, was administered to the control group, which was used as the positive control group. Drp1, Fis1, and Mff are mitochondrial
ssion activators whose levels were upregulated in response to TNFα treatment and downregulated by Mdivi-1. By contrast, Mfn2 and Opa1 are the mitochondrial ssion
inhibitors whose expression levels were repressed by TNFα stress and were increased by Mdivi-1. *P,0.05.
Abbreviation: Mdivi-1, mitochondrial division inhibitor-1.
10
*
8
6
4
2
Mitochondrial
length (µm)
0
Control
TNFα (1 ng/mL)
TNFα (5 ng/mL)
AB
C
2.50
**
2.00
1.50
1.00
Drp1 expression
0.50
0.00
Control
TNFα (5 ng/mL)
TNFα (5 ng/mL) +
Mdivi-1
Control FCCP
1.25
**
1.00
0.75
0.50
Mfn2 expression
0.25
0.00
Control
TNFα (5 ng/mL)
TNFα (5 ng/mL) +
Mdivi-1
Control FCCP
1.25
**
1.00
0.75
0.50
Opa1 expression
0.25
0.00
Control
TNFα (5 ng/mL)
TNFα (5 ng/mL) +
Mdivi-1
Control FCCP
3.00
**
2.00
1.00
Mff expression
0.00
Control
TNFα (5 ng/mL)
TNFα (5 ng/mL) +
Mdivi-1
Control FCCP
D
GH
4.00
**
3.00
2.00
1.00
Fis1 expression
0.00
Control
TNFα (5 ng/mL)
TNFα (5 ng/mL) +
Mdivi-1
Control FCCP
EF
β-actin
Drp1
Control
TNFα (5 ng/mL)
TNFα (5 ng/mL) +
Mdivi-1
Control+FCCP
Fis1
Mff
Mfn2
Opa1
Control
MitochondriaMagnification
10 µm
TNFα (1 ng/mL) TNFα (5 ng/mL)
10 µm
RETRACTED
OncoTargets and Therapy 2018:11 submit your manuscript | www.dovepress.com
Dovepress
Dovepress
7219
TNFα promotes glioblastoma A172 cell
GSH, SOD, and GPX was obviously reduced in response to
TNFα treatment (Figure 3C–E). However, Mdivi-1 could
reverse the levels of GSH, SOD, and GPX (Figure 3C–E).
The abovementioned data suggested that TNFα-mediated
mitochondrial oxidative stress via mitochondrial fission.
The core function of mitochondria is to produce ATP,
which is required for cellular metabolism. Interestingly,
the content of ATP was significantly reduced in the pres-
ence of TNFα treatment (Figure 3F), similar to the results
obtained after administering FCCP. However, Mdivi-1
supplementation abrogated the inhibitory effects of TNFα
on ATP production (Figure 3F). At the molecular level,
mitochondria produce ATP via the mitochondrial respiratory
complex. Notably, the protein expression of mitochondrial
respiratory complex was significantly repressed by TNFα
(Figure 3G–J), and this effect was negated by Mdivi-1. This
information indicated that TNFα-mediated mitochondrial
fission reduced the levels of the mitochondrial respiratory
complex. Altogether, our data confirm that TNFα treatment
causes an obvious mitochondrial malfunction that occurs, at
least in part, through mitochondrial fission.
TNFα-mediated mitochondrial
ssion activates a caspase-9-related
mitochondrial apoptotic pathway
Damaged mitochondria initiate cellular apoptosis programs.34
Based on this, we explored whether TNFα-mediated mito-
chondrial fission accounted for glioblastoma cell apoptosis. An
early molecular feature of mitochondrial apoptosis is a drop
in the mitochondrial potential. As shown in Figure 4A and B,
Figure 3 TNFα-initiated mitochondrial ssion contributes to glioblastoma mitochondrial injury.
Notes: (A and B) ROS levels were measured using DHE probe, and ow cytometry was performed to quantify the content of cell ROS. To perform the loss- and gain-of-
function assays for mitochondrial ssion, Mdivi-1, a pharmacological antagonist was used in TNFα-treated cells to inhibit mitochondrial ssion activation. FCCP, an agonist
for mitochondrial ssion was administered to the control group, which was set as the positive control group. (CE) ELISAs for cellular antioxidants such as GSH, SOD,
and GPX. (F) Cellular ATP production was measured using a commercial kit. (GJ) Western blot was conducted to analyze the protein expression of the mitochondrial
respiratory complex. *P,0.05.
Abbreviations: DHE, dihydroethidium; Mdivi-1, mitochondrial division inhibitor-1; CIII-core2, complex III subunit core; CII-30, complex II; CIV-II, complex IV subunit II.





*6+OHYHOV
QPROPJSURWHLQ
&RQWURO
&RQWURO)&&3
71)α
71)α0GLYL
&

62'DFWLYLW\
8PJSURWHLQ
&RQWURO
&RQWURO)&&3
71)α
71)α0GLYL
'




*3;DFWLYLW\
8PJSURWHLQ
&RQWURO
&RQWURO)&&3
71)α
71)α0GLYL
(







$73SURGXFWLRQ
RIFRQWURO
&RQWURO
&RQWURO)&&3
71)α
71)α0GLYL
)
&RQWURO
&RQWURO)&&3
71)α
71)α0GLYL






&,9,,
H[SUHVVLRQ
&RQWURO
&RQWURO)&&3
71)α
71)α0GLYL






&,,
H[SUHVVLRQ







&,,,FRUH
H[SUHVVLRQ
&RQWURO
&RQWURO)&&3
71)α
71)α0GLYL
7RP
&,9,,
&,,
&,,,FRUH
&RQWURO
&RQWURO)&&3
71)α
71)α0GLYL
*+,-






526
&RQWURO)&&3
&RQWURO






526
71)α0GLYL
&RQWURO






526
71)α
&RQWURO
$








526IOXRUHVFHQFH
IROGV
&RQWURO
&RQWURO)&&3
71)α
71)α0GLYL
%
RETRACTED
OncoTargets and Therapy 2018:11
submit your manuscript | www.dovepress.com
Dovepress
Dovepress
7220
Lu et al
compared to the control group, TNFα markedly reduced the
mitochondrial potential as evidenced by decreased red fluo-
rescence and increased green fluorescence. Interestingly, this
alteration could be abrogated by Mdivi-1 (Figure 4A and B),
suggesting that inhibition of mitochondrial fission protected
the mitochondrial potential in the presence of TNFα treat-
ment. The collapse of the mitochondrial potential indicates
hyperpermeability of the mitochondrial outer membrane.35
Accordingly, we evaluated the opening rate of the mPTP.
Compared to the control group, TNFα treatment increased
the opening rate of mPTP (Figure 4C), similar to the results
obtained via administration of FCCP. However, Mdivi-1
supplementation significantly blocked the mPTP opening
(Figure 4C). Excessive opening of mPTP could facilitate
mitochondrial proapoptotic cyt-c translocation into the cyto-
plasm where cyt-c interacts with and activates caspase-9.36
The immunofluorescence assay for cyt-c indicated that TNFα
treatment promoted cyt-c migration to the nucleus (Figure 4D
and E), and this effect was negated by Mdivi-1. In response to
the cyc-c liberation, the activity of caspase-9 was increased
in TNFα-treated cells, whereas Mdivi-1 treatment prevented
caspase-9 activation (Figure 4F).
In addition, we also found that the expression levels
of mitochondrial proapoptotic proteins such as Bax and
Bad were significantly upregulated in TNFα-treated cells
(Figure 4G–K), similar to the results obtained via adding
FCCP. By comparison, the levels of antiapoptotic proteins
such as Bcl-2 and x-IAP were downregulated in response to
TNFα stress (Figure 4G–K). Interestingly, Mdivi-1 treatment
reversed the levels of antiapoptotic factors. These results
indicated that mitochondrial apoptosis was activated by
TNFα via mitochondrial fission.
TNFα modulates mitochondrial ssion
via MAPK–ERK–YAP signaling pathways
Subsequently, we explored the molecular mechanism by
which TNFα controlled mitochondrialssion in glioblastoma
cells. Previous studies have suggested that mitochondrial
fission is negatively regulated by the MAPK–ERK–YAP
signaling pathways.37,38 In the present study, we noted abun-
dant p-ERK expression in the control group via Western blot
(Figure 5A–C). However, TNFα treatment significantly sup-
pressed p-ERK expression (Figure 5A–C), indicative of ERK
inactivation in response to TNFα stimulus. Moreover, the
TNFα-mediated decrease in p-ERK expression was closely
associated with a drop in YAP expression (Figure 5A–C),
suggesting that TNFα inactivated MAPK–ERK–YAP path-
ways in glioblastoma cells. PD98059 was used to inhibit ERK
activity, which was used to mimic the inhibitory effects of
TNFα on ERK pathways. This finding was further supported
Figure 4 (Continued)
&RQWURO)&&371)α0GLYL71)α&RQWURO
5HG
IOXRUHVFHQFH
*UHHQ
IOXRUHVFHQFH0HUJHG
P
&RQWURO
71)α
71)α0GLYL
&RQWURO)&&3

P373RSHQLQJ
UDWHIROGV







&RQWURO
71)α
71)α0GLYL
&RQWURO)&&3

5DWLRRIUHGWR
JUHHQIOXRUHVFHQFH
LQWHQVLW\







$%
&
RETRACTED
OncoTargets and Therapy 2018:11 submit your manuscript | www.dovepress.com
Dovepress
Dovepress
7221
TNFα promotes glioblastoma A172 cell
Figure 4 TNFα-activated caspase-9 apoptosis is regulated by mitochondrial ssion.
Notes: (A and B) Mitochondrial potential was observed via JC-1 staining. Red uorescence of the JC-1 probe indicates the normal mitochondrial potential, whereas green
uorescence of the JC-1 probe means a defective mitochondrial potential. The red-to-green uorescence intensity was recorded to quantify the mitochondrial potential.
To perform the loss- and gain-of-function assays for mitochondrial ssion, Mdivi-1, a pharmacological antagonist was used in TNFα-treated cells to inhibit mitochondrial
ssion activation. FCCP, an agonist for mitochondrial ssion, was administered to the control group, which was set as the positive control group. (C) mPTP opening was
measured in response to TNFα stress and/or mitochondrial ssion inhibition. (D and E) Immunouorescence assay for mitochondrial cyt-c translocation into nucleus. Nuclei
were labeled by DAPI, and the colocalization of cyt-c and DAPI indicates the migration of mitochondrial cyt-c into nucleus. The relative expression of nuclear cyt-c was
monitored. (F) Caspase-9 activity was determined via ELISA. TNFα-mediated caspase-9 activation could be abrogated by Mdivi-1. (GK) Western blot was performed to
analyze the alterations in mitochondrial apoptotic proteins. Bax and Bad are proapoptotic proteins, whereas Bcl-2 and x-IAP are antiapoptotic proteins. TNFα regulated the
balance of proapoptotic and antiapoptotic proteins via mitochondrial ssion. *P,0.05.
Abbreviation: Mdivi-1, mitochondrial division inhibitor-1.
&RQWURO)&&371)α0GLYL71)α&RQWURO
&\WF'$3,0HUJHG
P
&RQWURO
71)α
71)α0GLYL
&RQWURO)&&3

&DVSDVH
DFWLYLW\IROGV







&RQWURO
71)α
71)α0GLYL
&RQWURO)&&3

1XFOHDU
H[SUHVVLRQRIF\WF







&RQWURO
71)α
71)α0GLYL
&RQWURO)&&3
%DG
%D[
%FO
[,$3
*$3'+
&RQWURO
71)α
71)α0GLYL
&RQWURO)&&3

%FOH[SUHVVLRQ






&RQWURO
71)α
71)α0GLYL
&RQWURO)&&3

[,$3H[SUHVVLRQ






&RQWURO
71)α
71)α0GLYL
&RQWURO)&&3

%DGH[SUHVVLRQ






&RQWURO
71)α
71)α0GLYL
&RQWURO)&&3

%D[H[SUHVVLRQ




'(
)
+,
.
-
*
RETRACTED
OncoTargets and Therapy 2018:11
submit your manuscript | www.dovepress.com
Dovepress
Dovepress
7222
Lu et al
Figure 5 TNFα handles mitochondrial ssion via MAPK–ERK–YAP pathways.
Notes: (AC) The expression values of ERK and YAP were determined via Western blot. Phosphorylated ERK and YAP expressions were both downregulated by TNFα.
Subsequently, Ad-YAP was transfected into cells to overexpress YAP in TNFα-treated cells. PD98059 was used to inhibit ERK activity, which was used to mimic the inhibitory
effects of TNFα on ERK pathways. (D and E) Immunouorescence of p-ERK and YAP in cells treated with TNFα or transfected with Ad-YAP or Ad-ctrl. (F and G). The
overexpression efciency of Ad-YAP infection. Western blot was performed to analyze the protein expression of YAP in cells treated with Ad-YAP or Ad-ctrl. (H and I)
Mitochondrial ssion was determined via immunouorescence using mitochondrial-specic Tom20 antibody. The average length of mitochondria was evaluated to quantify
mitochondrial ssion. *P,0.05.
Abbreviation: Ad, adenovirus.
$
&RQWURO
&RQWURO3'
71)αQJP/
71)αQJP/
$GFRQWURO
71)αQJP/
$G<$3
S(5.
W(5.
<$3
βDFWLQ
&RQWURO
&RQWURO3'
71)αQJP/
71)αQJP/
$GFRQWURO
71)αQJP/
$G<$3
S(5.
H[SUHVVLRQ







%
&RQWURO
&RQWURO3'
71)αQJP/
71)αQJP/
$GFRQWURO
71)αQJP/
$G<$3
<$3
H[SUHVVLRQ






&
&RQWURO
&RQWURO
3'
71)α
QJP/
71)αQJP/
$GFRQWURO
71)αQJP/
$G<$3
P
+
0LWRFKRQGULD0DJQLILFDWLRQ
&RQWURO
&RQWURO3'
71)αQJP/
71)αQJP/
$GFRQWURO
71)αQJP/
$G<$3
0LWRFKRQGULDO
OHQJWKP

,
'
*
)
&RQWURO
&RQWURO3'
71)αQJP/
71)αQJP/
$GFRQWURO
71)αQJP/
Y$G<$3
<$3
IOXRUHVFHQFH
LQWHQVLW\






(
&RQWURO
$GFRQWURO
$G<$3
<$3
H[SUHVVLRQ






&RQWURO
$G<$3
$GFRQWURO
<$3
βDFWLQ
&RQWURO
&RQWURO
3'
71)α
QJP/
71)αQJP/
$GFRQWURO
71)αQJP/
$G<$3
S(5.0HUJHG <$3
P
RETRACTED
OncoTargets and Therapy 2018:11 submit your manuscript | www.dovepress.com
Dovepress
Dovepress
7223
TNFα promotes glioblastoma A172 cell
via immunofluorescence (Figure 5D and E). The fluorescence
intensities of p-ERK and YAP in the TNFα-treated cells
decreased by ~65% and ~50% of the control levels, respec-
tively. To demonstrate whether MAPK–ERK–YAP signaling
pathways were required for TNFα-mediated mitochondrial
fission, we overexpressed YAP in TNFα-treated cells. The
transfection efficiency was verified via immunofluorescence
assay (Figure 5D and E) and Western blot (Figure 5F and G).
Then, mitochondrial fission was evaluated again. As shown
in Figure 5H and I, TNFα treatment promoted the forma-
tion of fragmented mitochondria whose length was shorter
when compared to that of the control group. Interestingly,
TNFα-mediated mitochondrial division could be inhibited
by YAP overexpression (Figure 5H and I). Altogether, our
results confirm that MAPK–ERK–YAP signaling pathways
are required for TNFα-controlled mitochondrial fission.
MAPK–ERK–YAP signaling pathways
are also involved in mitochondrial
malfunction and glioblastoma cell death
We explored whether MAPK–ERK–YAP signaling path-
ways are involved in TNFα-mediated mitochondrial injury
and cell death. First, ROS production was measured via
immunofluorescence assay. Compared to the control group,
TNFα treatment elevated the levels of cell ROS (Figure 6A
and B), and this effect was reversed by YAP overexpression.
In addition, cyt-c translocation from the mitochondria into
the cytoplasm/nucleus was exacerbated by TNFα stress
and was repressed by YAP overexpression (Figure 6C
and D). In response to cyt-c leakage, caspase-9 activity was
augmented in TNFα-treated cells and was reduced to
near-normal levels with YAP overexpression (Figure 6E).
Altogether, this information indicated that TNFα-mediated
mitochondrial injury could be interrupted via activation of
the MAPK–ERK–YAP axes.
With respect to cell apoptosis, TUNEL assays were con-
ducted to observe the apoptotic cells. Compared to the control
group, TNFα treatment elevated the number of TUNEL-
positive cells (Figure 6F and G), and this effect was abro-
gated by YAP overexpression. Similarly, the LDH cytotoxic
test also indicated that TNFα-mediated LDH release could
be suppressed by YAP overexpression (Figure 6H). Col-
lectively, the above data demonstrate that TNFα-mediated
mitochondrial damage and cell death are mainly regulated
by the MAPK–ERK–YAP axes.
Discussion
The treatment of glioblastomas currently remains difficult
due to inevitable recurrence and rapid progression.39 Current
treatment options include radiation therapy in addition to
surgery or surgery combined with chemotherapy.40 In the
present study, we found that TNFα treatment significantly
reduced the viability of glioblastoma cells in a dose-dependent
manner. Functional investigations revealed that TNFα
supplementation activated mitochondrial fission and that
mitochondrial fission subsequently mediated mitochondrial
injury and initiated caspase-9-involved mitochondrial apop-
tosis. Inhibition of mitochondrial fission could abrogate the
proapoptotic effects of TNFα on glioblastoma cells. Further-
more, we showed that TNFα-induced mitochondrial fission
was modified by the MAPK–ERK–YAP signaling pathways.
TNFα treatment repressed the activity of MAPK–ERK–YAP
signaling pathways, leading to an increase in the content of
mitochondrial fission factors such as Drp1. Reactivation of
MAPK–ERK–YAP signaling pathways could inhibit TNFα-
mediated mitochondrial fission and provide a prosurvival
advantage for glioblastomas cells. Collectively, this is the
first study to demonstrate that TNFα regulates glioblastoma
cell viability and mitochondrial homeostasis by modulating
mitochondrial fission through MAPK–ERK–YAP-dependent
signaling pathways (Figure 7). Our results lay the foundation
to help us understand the molecular mechanisms of TNFα-
mediated cancer cytotoxicity.
TNFα, an inflammatory cytokine, is of significant
importance in regulating cancer progression in many types
of malignant tumors.41,42 This fact led to several animal
experiments and clinical studies to explore the detailed role
of TNFα in retarding the progression of glioblastomas.
Early studies have demonstrated that gene transduction of
a human TNFα-vector substantially increased the apoptotic
index and reduced the growth rate in human glioblastoma
cells.2 Subsequent studies determined that TNFα supplemen-
tation enhanced the susceptibility of human glioblastoma
cells to natural killer cells.43 In addition, TNFα treatment
also reduced the adhesion capacity, evoked cellular oxidant
stress,44,45 and suppressed tumor angiogenesis46 in primary
or recurrent glioblastomas. In the current study, our results
demonstrated that TNFα stress was closely associated with
mitochondrial damage in glioblastoma cells. In response
to TNFα stimulus, mitochondrial ROS production was
increased, which was accompanied by a drop in the levels
of antioxidant factors. In addition, mitochondrial ATP pro-
duction was also impaired, which may result from TNFα-
mediated downregulation of the mitochondrial respiratory
complex. More importantly, decreased mitochondrial poten-
tial, extended mPTP opening time, and more cyt-c liberation
into the nucleus were noted in TNFα-treated cells. These
alterations worked together to initiate caspase-9-related mito-
RETRACTED
OncoTargets and Therapy 2018:11
submit your manuscript | www.dovepress.com
Dovepress
Dovepress
7224
Lu et al
AB
D
E
H
2.50
**
2.00
1.50
1.00
ROS
production (fold)
0.50
0.00
Control
TNFα (5 ng/mL)
TNFα (5 ng/mL) +
Ad-control
TNFα (5 ng/mL) +
Ad-YAP
4.00
**
3.00
2.00
Nuclear cyt-c
expression (folds)
1.00
0.00
Control
TNFα (5 ng/mL)
TNFα (5 ng/mL) +
Ad-control
TNFα (5 ng/mL) +
Ad-YAP
4.00
**
3.00
2.00
Caspase-9 activity
(folds)
1.00
0.00
Control
TNFα (5 ng/mL)
TNFα (5 ng/mL) +
Ad-control
TNFα (5 ng/mL) +
Ad-YAP
4.00
**
3.00
2.00
LDH release
(folds)
1.00
0.00
Control
TNFα (5 ng/mL)
TNFα (5 ng/mL) +
Ad-control
TNFα (5 ng/mL) +
Ad-YAP
60
* *
50
40
30
Number of
TUNEL + cell (%)
20
10
0
Control
TNFα (5 ng/mL)
TNFα (5 ng/mL) +
Ad-control
TNFα (5 ng/mL) +
Ad-YAP
C
FG
Control
ROS
45 µm
TNFα (5 ng/mL)
TNFα (5 ng/mL) +
Ad-control
TNFα (5 ng/mL) +
Ad-YAP
DAPICyt-cMerged
10 µm
ControlTNFα (5 ng/mL)
TNFα (5 ng/mL) +
Ad-control
TNFα (5 ng/mL) +
Ad-YAP
TUNELDAPIMerged
60 µm
ControlTNFα (5 ng/mL)
TNFα (5 ng/mL) +
Ad-control
TNFα (5 ng/mL) +
Ad-YAP
Figure 6 MAPK–ERK–YAP pathways also participate in the regulation of mitochondrial homeostasis and cell death.
Notes: (A and B) ROS production was measured using immunouorescence. Ad-YAP was transfected into cells to reactivate MAPK–ERK–YAP pathways. (C and D)
Immunouorescence assays for cyt-c. The cellular location of cyt-c was determined, and DAPI was used to label the nucleus. (E) Caspase-9 activity was examined to
determine the role of MAPK–ERK–YAP pathways in caspase-9-mediated mitochondrial apoptosis. (F and G) TUNEL staining for apoptotic cells. The ratio of TUNEL-positive
cells was recorded. (H) An LDH release assay was used to analyze cell death. The ratio of relative LDH release was recorded compared to the control group. *P,0.05.
Abbreviation: Ad, adenovirus.
RETRACTED
OncoTargets and Therapy 2018:11 submit your manuscript | www.dovepress.com
Dovepress
Dovepress
7225
TNFα promotes glioblastoma A172 cell
chondrial apoptotic pathways, accounting for glioblastoma
cell death. Our findings are similar to previous studies that
indicated that TNFα treatment promoted mitochondrial dys-
function in glioblastoma cells.47 This information identifies
mitochondria as a primary target for TNFα-based therapy.
Based on this, the discovery of other drugs principally act-
ing on mitochondria may provide more clinical benefits for
patients with glioblastoma.
The novel finding in our study is that we show that
TNFα induces mitochondrial damage via mitochondrial
fission. Notably, mitochondrial fission has been suggested
as a chief cause of cell death by inducing mitochondrial
damage in several diseases. In cardiac ischemia-reperfusion
injuries, aberrant mitochondrial fission exacerbates cardio-
myocyte death via promoting mPTP opening and cardio-
lipin oxidation.5,6,48 Moreover, uncontrolled mitochondrial
fission also participates in fatty liver disease by disrupting
hepatocyte mitochondrial metabolism.49 In addition, in pan-
creatic cancer,13 breast cancer,11 ovarian cancer,12 and liver
cancer,50 mitochondrial fission exerts negative effects on
mitochondrial homeostasis and has been proposed to be a
primary apoptotic trigger. In the present study, we show for
the first time that mitochondrial fission induces mitochon-
drial damage, which precedes cell apoptosis in a caspase-9-
dependent manner. This is the first study to define the role
of mitochondrial fission in glioblastoma. Considering the
detrimental effects of mitochondrial fission on cell viability,
approaches to activate mitochondrial fission are of utmost
importance when designing antitumor therapies. Notably,
several studies have also found that TNFα treatment also
activated mitochondrial fusion in human kidney-2 cells51 and
cardiomyocytes.52 These results establish the various effects
of TNFα on mitochondrial fission and mitochondrial fusion.
This seems to be dependent on cell types. However, more
researches are required to validate our concept.
At the molecular level, we found that TNFα activated
mitochondrial fission via repression of MAPK–ERK–YAP
signaling pathways. First, more robust data concerning
the inhibitory effects of MAPK–ERK pathways on mito-
chondrial fission have been provided by several in vitro
and in vivo studies.53 More importantly, the MAPK–ERK
pathway, as the classical antiapoptotic pathway, has been
demonstrated to send beneficial signals to cells under
various states of stress.54 As a new downstream effector of
MAPK–ERK pathways, YAP was originally identified as
a proto-oncogene.55 Higher YAP expression is closely cor-
related with cancer progression and tumor metastasis.18 In
addition, increased YAP effectively controls mitochondrial
fission and sustains mitochondrial integrity,18 favoring cell
metabolism and growth. Based on this finding, several
researchers propose that MAPK–ERK–YAP pathways
are the upstream inhibitors of mitochondrial fission. This
conclusion is supported by our results. We found that reac-
tivation of MAPK–ERK–YAP pathways repressed mito-
chondrial fission and abrogated TNFα-mediated cell death.
Accordingly, our results combined with previous findings
highlight the molecular mechanisms by which TNFα regu-
lates mitochondrial fission. At the molecular levels, several
researchers have investigated the mechanism by which YAP
modulated mitochondrial fission. Increased YAP reduces
the transcription and expression of Mff and Drp1, strongly
attenuating mitochondrial fission. Moreover, YAP has an
ability to modify the phosphorylation of Drp1. In addition,
YAP overexpression also reverses mitochondrial fusion via
upregulating the expression of mitochondrial fusion factors
such as OPA1 and Mfn2. These results explain the inhibitory
effect of YAP on mitochondrial fission.
The clinical implication that can be drawn from our
study is multifold. Our data provide a piece of evidence for
the role of mitochondrial fission in glioblastoma viability.
This information indicates that mitochondrial fission would
be considered as a potential target to prevent glioblastoma
progression via promoting mitochondrial fission-mediated
cell apoptosis. On the other hand, our findings identify
MAPK–ERK–YAP pathways as novel regulators for han-
dling mitochondrial function and glioblastoma viability. This
may highlight a new entry point for treating glioblastoma by
targeting the MAPK–ERK–YAP signaling axes.
Figure 7 TNFα treatment elevates the apoptotic rate of glioblastoma in vitro by
initiating fatal mitochondrial ssion and interrupting MAPK–ERK–YAP signaling
pathways.
RETRACTED
OncoTargets and Therapy 2018:11
submit your manuscript | www.dovepress.com
Dovepress
Dovepress
7226
Lu et al
Limitation
The primary limitation of our study is that only one cell line
was used in the present study to explore the roles of TNFα
and mitochondrial fission in cell viability. Animal studies
and clinical researches are required to further verify our
findings.56
Conclusion
Altogether, our results show that TNFα treatment elevates
the apoptotic rate of glioblastoma in vitro by initiating fatal
mitochondrial fission and interrupting MAPK–ERK–YAP
signaling pathways. These findings define mitochondrial
fission as a novel tumor suppressor that acts by inducing
mitochondrial damage, with potential implications for new
approaches to glioblastoma treatment.
Availability of data and materials
The datasets used and/or analyzed during the current study
are available from the corresponding author on reasonable
request.
Acknowledgment
No funding was received.
Author contributions
CL, XC, and QW made substantial contributions to the
concept and design of the present study, QW, XX, BX,
and CL contributed to the performance of experiments,
data analysis and interpretation, and manuscript writing.
All authors contributed to data analysis, drafting and
revising the article, gave final approval of the version to
be published, and agree to be accountable for all aspects
of the work.
Disclosure
The authors report no conflicts of interest in this work.
References
1. Groneberg DA, Addicks AM, Bendels MH, Quarcoo D, Jaque J,
Brüggmann D. Glioblastoma research: US and international network-
ing achievements. Oncotarget. 2017;8(70):115730–115735.
2. Walther W, Stein U, Pfeil D. Gene transfer of human TNF alpha into
glioblastoma cells permits modulation of mdr1 expression and potentia-
tion of chemosensitivity. Int J Cancer. 1995;61(6):832–839.
3. Zhang W, Sun Y, Liu L, Li Z. Prognostic Significance of TNFR-Associated
Factor 1 and 2 (TRAF1 and TRAF2) in Glioblastoma. Med Sci Monit.
2017;23:4506–4512.
4. Ge H, Mu L, Jin L, et al. Tumor associated CD70 expression is involved
in promoting tumor migration and macrophage infiltration in GBM. Int J
Cancer. 2017;141(7):1434–1444.
5. Zhou H, Shi C, Hu S, Zhu H, Ren J, Chen Y. BI1 is associated with micro-
vascular protection in cardiac ischemia reperfusion injury via repress-
ing Syk-Nox2-Drp1-mitochondrial fission pathways. Angiogenesis.
2018;21(3):599–615.
6. Zhou H, Wang J, Zhu P, et al. NR4A1 aggravates the cardiac micro-
vascular ischemia reperfusion injury through suppressing FUNDC1-
mediated mitophagy and promoting Mff-required mitochondrial fission
by CK2α. Basic Res Cardiol. 2018;113(4):23.
7. Cho SY, Kim SH, Yi MH, et al. Expression of PGC1α in glioblastoma
multiforme patients. Oncol Lett. 2017;13(6):4055–4076.
8. Gupta P, Dixit D, Sen E. Oncrasin targets the JNK-NF-κB axis to sen-
sitize glioma cells to TNFα-induced apoptosis. Carcinogenesis. 2013;
34(2):388–396.
9. Unterkircher T, Cristofanon S, Vellanki SH, et al. Bortezomib primes
glioblastoma, including glioblastoma stem cells, for TRAIL by increas-
ing tBid stability and mitochondrial apoptosis. Clin Cancer Res. 2011;
17(12):4019–4030.
10. Kim JY, Kim YJ, Lee S, Park JH. BNip3 is a mediator of TNF-induced
necrotic cell death. Apoptosis. 2011;16(2):114–126.
11. Lucantoni F, Düssmann H, Llorente-Folch I, Prehn JHM. BCL2 and
BCL(X)L selective inhibitors decrease mitochondrial ATP produc-
tion in breast cancer cells and are synthetically lethal when combined
with 2-deoxy-D-glucose. Oncotarget. 2018;9(40):26046–26063.
12. Noack S, Raab M, Matthess Y, et al. Synthetic lethality in CCNE1-
amplified high grade serous ovarian cancer through combined inhibition
of Polo-like kinase 1 and microtubule dynamics. Oncotarget. 2018;
9(40):25842–25859.
13. Pan L, Zhou L, Yin W, Bai J, Liu R. miR-125a induces apoptosis,
metabolism disorder and migration impairment in pancreatic cancer
cells by targeting Mfn2-related mitochondrial fission. Int J Oncol. 2018;
53(1):124–136.
14. Huang L, Luan T, Chen Y, et al. LASS2 regulates invasion and chemore-
sistance via ERK/Drp1 modulated mitochondrial dynamics in bladder
cancer cells. J Cancer. 2018;9(6):1017–1024.
15. Shen YL, Shi YZ, Chen GG, et al. TNF-α induces Drp1-mediated
mitochondrial fragmentation during inflammatory cardiomyocyte
injury. Int J Mol Med. 2018;41(4):2317–2327.
16. Sun X, Cao H, Zhan L, et al. Mitochondrial fission promotes cell migra-
tion by Ca2+/CaMKII/ERK/FAK pathway in hepatocellular carcinoma.
Liver Int. 2018;38(7):1263–1272.
17. Qiao P, Zhao F, Liu M, Gao D, Zhang H, Yan Y. Hydrogen sulfide inhib-
its mitochondrial fission in neuroblastoma N2a cells through the Drp1/
ERK1/2 signaling pathway. Mol Med Rep. 2017;16(1):971–977.
18. Li H, He F, Zhao X, et al. YAP Inhibits the Apoptosis and Migration of
Human Rectal Cancer Cells via Suppression of JNK-Drp1-Mitochondrial
Fission-HtrA2/Omi Pathways. Cell Physiol Biochem. 2017;44(5):
2073–2089.
19. Geng C, Wei J, Wu C. Yap-Hippo pathway regulates cerebral hypoxia-
reoxygenation injury in neuroblastoma N2a cells via inhibiting
ROCK1/F-actin/mitochondrial fission pathways. Acta Neurol Belg.
Epub 2018 May 23.
20. Du X, Wen J, Wang Y, et al. Hippo/Mst signalling couples metabolic
state and immune function of CD8α+ dendritic cells. Nature. 2018;
558(7708):141–145.
21. Zhang S, Shi R, Chen S, Wei X, Zhou Q, Wang Y. All-trans retinoic
acid inhibits the proliferation of SGC7901 cells by regulating caveolin-1
localization via the ERK/MAPK signaling pathway. Oncol Lett.
2018;15(2):1523–1528.
22. Yan H, Xiao F, Zou J, et al. NR4A1-induced increase in the sensitivity
of a human gastric cancer line to TNFα-mediated apoptosis is associated
with the inhibition of JNK/Parkin-dependent mitophagy. Int J Oncol.
2018;52(2):367–378.
23. Ackermann M, Kim YO, Wagner WL, et al. Effects of nintedanib on
the microvascular architecture in a lung fibrosis model. Angiogenesis.
2017;20(3):359–372.
24. du GQ, Shao ZB, Wu J, et al. Targeted myocardial delivery of GDF11 gene
rejuvenates the aged mouse heart and enhances myocardial regeneration
after ischemia-reperfusion injury. Basic Res Cardiol. 2017;112(1):7.
25. Schock SN, Chandra NV, Sun Y, et al. Induction of necroptotic cell
death by viral activation of the RIG-I or STING pathway. Cell Death
Differ. 2017;24(4):615–625.
RETRACTED
OncoTargets and erapy
Publish your work in this journal
Submit your manuscript here: http://www.dovepress.com/oncotargets-and-therapy-journal
OncoTargets and Therapy is an international, peer-reviewed, open
access journal focusing on the pathological basis of all cancers, potential
targets for therapy and treatment protocols employed to improve the
management of cancer patients. The journal also focuses on the impact
of management programs and new therapeutic agents and protocols on
patient perspectives such as quality of life, adherence and satisfaction.
The manuscript management system is completely online and includes
a very quick and fair peer-review system, which is all easy to use. Visit
http://www.dovepress.com/testimonials.php to read real quotes from
published authors.
OncoTargets and Therapy 2018:11 submit your manuscript | www.dovepress.com
Dovepress
Dovepress
Dovepress
7227
TNFα promotes glioblastoma A172 cell
26. Li J, Chen L, Xiong Y, et al. Knockdown of PD-L1 in Human Gastric
Cancer Cells Inhibits Tumor Progression and Improves the Cytotoxic
Sensitivity to CIK Therapy. Cell Physiol Biochem. 2017;41(3):907–920.
27. Zhang Y, Zhou H, Wu W, et al. Liraglutide protects cardiac microvas-
cular endothelial cells against hypoxia/reoxygenation injury through
the suppression of the SR-Ca(2+)-XO-ROS axis via activation of the
GLP-1R/PI3K/Akt/survivin pathways. Free Radic Biol Med. 2016;95:
278–292.
28. Yu S, Wang X, Geng P, et al. Melatonin regulates PARP1 to control
the senescence-associated secretory phenotype (SASP) in human fetal
lung fibroblast cells. J Pineal Res. 2017;63(1):e12405.
29. Hernansanz-Agustín P, Ramos E, Navarro E, et al. Mitochondrial
complex I deactivation is related to superoxide production in acute
hypoxia. Redox Biol. 2017;12:1040–1051.
30. Zhu H, Jin Q, Li Y, et al. Melatonin protected cardiac microvascular
endothelial cells against oxidative stress injury via suppression of
IP3R-[Ca2+]c/VDAC-[Ca2+]m axis by activation of MAPK/ERK signal-
ing pathway. Cell Stress Chaperones. 2018;23(1):101–113.
31. Randriamboavonjy V, Kyselova A, Elgheznawy A, Zukunft S, Wittig I,
Fleming I. Calpain 1 cleaves and inactivates prostacyclin synthase
in mesenteric arteries from diabetic mice. Basic Res Cardiol. 2017;
112(1):10.
32. Zhou H, Hu S, Jin Q, et al. Mff-Dependent Mitochondrial Fission
Contributes to the Pathogenesis of Cardiac Microvasculature Ischemia/
Reperfusion Injury via Induction of mROS-Mediated Cardiolipin
Oxidation and HK2/VDAC1 Disassociation-Involved mPTP Opening.
J Am Heart Assoc. 2017;6(3):e005328.
33. Jin Q, Li R, Hu N, et al. DUSP1 alleviates cardiac ischemia/reperfu-
sion injury by suppressing the Mff-required mitochondrial fission and
Bnip3-related mitophagy via the JNK pathways. Redox Biol. 2018;14:
576–587.
34. Fuhrmann DC, Brüne B. Mitochondrial composition and function under
the control of hypoxia. Redox Biol. 2017;12:208–215.
35. Li R, Xin T, Li D, Wang C, Zhu H, Zhou H. Therapeutic effect of
Sirtuin 3 on ameliorating nonalcoholic fatty liver disease: The role of
the ERK-CREB pathway and Bnip3-mediated mitophagy. Redox Biol.
2018;18:229–243.
36. Zhou H, Yue Y, Wang J, Ma Q, Chen Y. Melatonin therapy for diabetic
cardiomyopathy: A mechanism involving Syk-mitochondrial complex
I-SERCA pathway. Cell Signal. 2018;47:88–100.
37. Zhou H, Li D, Zhu P, et al. Inhibitory effect of melatonin on necroptosis
via repressing the Ripk3-PGAM5-CypD-mPTP pathway attenuates
cardiac microvascular ischemia-reperfusion injury. J Pineal Res. 2018:
e12503.
38. Zhou H, Wang J, Zhu P, Hu S, Ren J. Ripk3 regulates cardiac microvas-
cular reperfusion injury: The role of IP3R-dependent calcium overload,
XO-mediated oxidative stress and F-action/filopodia-based cellular
migration. Cell Signal. 2018;45:12–22.
39. Tamimi AF, Juweid M. Epidemiology and outcome of glioblastoma.
In: De Vleeschouwer S, editor. Glioblastoma. Brisbane, AU: Codon
Publications; 2017.
40. Mann J, Ramakrishna R, Magge R, Wernicke AG. Advances in Radio-
therapy for Glioblastoma. Front Neurol. 2017;8:748.
41. Perez JG, Tran NL, Rosenblum MG, et al. The TWEAK receptor Fn14
is a potential cell surface portal for targeted delivery of glioblastoma
therapeutics. Oncogene. 2016;35(17):2145–2155.
42. Xu Q, Choksi S, Liu Z. Switching from TNF-induced inflammation to
death signaling. Mol Cell Oncol. 2018;5(1):e1392402.
43. Kondo S, Yin D, Takeuchi J, et al. Tumour necrosis factor-alpha induces
an increase in susceptibility of human glioblastoma U87-MG cells to
natural killer cell-mediated lysis. Br J Cancer. 1994;69(4):627–632.
44. Sawada M, Kiyono T, Nakashima S, et al. Molecular mechanisms
of TNF-alpha-induced ceramide formation in human glioma cells:
P53-mediated oxidant stress-dependent and -independent pathways.
Cell Death Differ. 2004;11(9):997–1008.
45. Zhou H, Wang S, Hu S, Chen Y, Ren J. ER-Mitochondria Microdomains
in Cardiac Ischemia-Reperfusion Injury: A Fresh Perspective. Front
Physiol. 2018;9:755.
46. Jazayeri S, Feli A, Bitaraf MA, Solaymani Dodaran M, Alikhani M,
Hosseinzadeh-Attar MJ. Effects of Copper Reduction on Angiogenesis-
Related Factors in Recurrent Glioblastoma Cases. Asian Pac J Cancer
Prev. 2016;17(10):4609–4614.
47. Zhang C, Jiang H, Wang P, Liu H, Sun X. Transcription factor NF-kappa
B represses ANT1 transcription and leads to mitochondrial dysfunc-
tions. Sci Rep. 2017;7:44708.
48. Zhou H, Zhu P, Wang J, Zhu H, Ren J, Chen Y. Pathogenesis of cardiac
ischemia reperfusion injury is associated with CK2α-disturbed mito-
chondrial homeostasis via suppression of FUNDC1-related mitophagy.
Cell Death Differ. 2018;25(6):1080–1093.
49. Zhou H, du W, Li Y, et al. Effects of melatonin on fatty liver disease:
The role of NR4A1/DNA-PKcs/p53 pathway, mitochondrial fission,
and mitophagy. J Pineal Res. 2018;64(1):e12450.
50. Tak H, Kang H, Ji E, Hong Y, Kim W, Lee EK. Potential use of TIA-1,
MFF, microRNA-200a-3p, and microRNA-27 as a novel marker for
hepatocellular carcinoma. Biochem Biophys Res Commun. 2018;497(4):
1117–1122.
51. Liu N, Jiang Z, Liu Y, et al. Human trypsin inhibitor reduces the apop-
tosis of lipopolysaccharide-induced human kidney-2 cells by promot-
ing mitochondrial fusion. Mol Med Rep. 2017;16(3):2899–2906.
52. Nan J, Hu H, Sun Y, et al. TNFR2 Stimulation Promotes Mitochondrial
Fusion via Stat3- and NF-kB-Dependent Activation of OPA1 Expres-
sion. Circ Res. 2017;121(4):392–410.
53. Tomer D, Chippalkatti R, Mitra K, Rikhy R. ERK regulates mitochon-
drial membrane potential in fission deficient Drosophila follicle cells
during differentiation. Dev Biol. 2018;434(1):48–62.
54. Kochetkova EY, Blinova GI, Bystrova OA, Martynova MG, Pospelov VA,
Pospelova TV. Targeted elimination of senescent Ras-transformed
cells by suppression of MEK/ERK pathway. Aging. 2017;9(11):
2352–2375.
55. Yan H, Qiu C, Sun W, et al. Yap regulates gastric cancer survival
and migration via SIRT1/Mfn2/mitophagy. Oncol Rep. 2018;39(4):
1671–1681.
56. Kusaczuk M, Krętowski R, Stypułkowska A, Cechowska-Pasko M.
Molecular and cellular effects of a novel hydroxamate-based HDAC
inhibitor – belinostat – in glioblastoma cell lines: a preliminary report.
Invest New Drugs. 2016;34(5):552–564.
RETRACTED
... Mitochondria contribute significantly to ATP production, ROS generation, metabolic regulation, and additional determinants of cellular health. 30 Mitochondria experience structural variation and functional remodeling in response to external stimuli. 31 The inward folding of the mitochondrial inner membrane forms mitochondrial cristae, which are sites of OXPHOS. ...
Article
Purpose: Epithelial-mesenchymal transition (EMT) of retinal pigment epithelial (RPE) cells contributes to the epiretinal membrane development in proliferative vitreoretinopathy (PVR). This study aimed at investigating changes in mitochondrial function during EMT in PVR. Methods: Transmission electron microscopy (TEM) was utilized to examine the mitochondrial morphology in human PVR epiretinal membranes and retinal pigment epithelium of human donor eyes. Utilizing TGF-β1 induced EMT in ARPE-19 cells as an in vitro model, we assessed mitochondrial morphology using transmission electron microscopy (TEM), evaluated mitochondrial function through various assays including detection and analysis of mitochondrial membrane potential (MMP), mitochondrial deoxyribonucleic acid (mtDNA), reactive oxygen species (ROS), ATP, oxygen consumption rate (OCR), and extracellular acidification rate (ECAR). RNA sequencing was performed to identify differentially expressed genes (DEGs) related to mitochondrial function and PVR pathogenesis. Results: Mitochondrial morphological damage was observed in human PVR epiretinal membranes. TGF-β1 treatment led to morphological changes in mitochondria, increased oxidative stress, mitochondrial membrane depolarization, and reduction in mtDNA, mitochondrial respiration, and ATP production, indicating mitochondrial dysfunction in EMT ARPE-19 cells. Furthermore, RNA sequencing data highlighted the dysfunction, showing downregulation of mitochondria-related pathways and mitochondrial transcription factor A (TFAM), crucial for mtDNA maintenance. Conclusion: Our findings indicated that TGF-β1 treatment induced mitochondrial dysfunction in RPE cells during EMT, providing insights into the molecular mechanisms of PVR development.
... Similarly, studies also found that PD98059 reduced YAP1 expression and promoted mitochondrial fragmentation by reducing p-ERK in glioblastoma. 26 In previous studies, studies on PD-L1 and PD-1 mainly focus on the effect of tumor cells on immune cells, while the PD-1 molecule focuses more on its effect on immune cells. However, recent studies found that PD-1 was also expressed in melanoma, nonsmall cell lung cancer cells, and liver cancer cells. ...
Article
Full-text available
Background Antibodies, which target programmed cell death protein-1 (PD-1) or its ligand programmed death ligand-1 (PD-L1), can rescue T cells from an exhausted state and resume their immune response to cancer cells. Clinically, the purpose of blocking the PD-1/PD-L1 signaling pathway is to induce immune cells to play an anti-tumor role. However, the effect of intertumor PD-1/PD-L1 signal blocking on tumor cells remains unclear. Methods HepG2 cells were treated with DHA, IFN-γ, BSA, DDP, PD-1-Fc (1 μg/ml), IgG-Fc, nivolumab, or human IgG for 24 h, respectively. GEPIA, cBioPortal, and TIMER databases were used to analyze the correlation between YAP1, PD-1, and PD-L1 and ERK, ERK-5, JNK, and p38. Western blot was used to detect the expression of YAP1 and p-ERK. Results GEPIA, cBioPortal, and TIMER databases analysis showed that YAP1 was positively correlated with ERK. After HepG2 cells were treated with PD98059 (ERK inhibitor), the expression of YAP1 was decreased. In this study, we investigated the inhibitory effect of PD98059 on PD-1/PD-L1 signaling. Our study found that PD-1-Fc (PD-1 fusion protein) promoted the expression of p-ERK/ERK and YAP1 in HepG2 cells. In contrast, nivolumab (PD-1 blocking antibody) reduced the expression of p-ERK/ERK and YAP1 in IFN-γ-pretreated HepG2 cells. In addition, the application of DHA also inhibited the expression of p-ERK/ERK to inhibit YAP1. Furthermore, treatment of HepG2 cells with DHA alone or DHA combined with cisplatin (DDP) both inhibited the expression of p-ERK/ERK and YAP1. Conclusions These results suggested that PD-1/PD-L1 interactions between tumor cells could promote the expression of ERK or YAP1 within tumors. Moreover, the conduction of PD-1/PD-L1 could be reversed using ERK inhibitors.
... Mitochondria play critical organelles roles in various aspects of biological processes such as energy generation, immune responses, programmed cell death and metabolism of various bioactive molecules including calcium, iron-sulfur cluster, one-carbon units, nucleotide, amino acid and lipid metabolism (Huertas et al., 2017;Li et al., 2018;Lu et al., 2018;Spinelli & Haigis, 2018;. Various factors such as free radical production, mtDNA mutation, etc., can cause mitochondrial dysfunction. ...
Article
Full-text available
Mitochondrial DNA (mtDNA) encodes proteins and RNAs that are essential for mitochondrial function and cellular homeostasis, and participates in important processes of cellular bioenergetics and metabolism. Alterations in mtDNA are associated with various diseases, especially cancers, and are considered as biomarkers for some types of tumors. Moreover, mtDNA alterations have been found to affect the proliferation, progression and metastasis of cancer cells, as well as their interactions with the immune system and the tumor microenvironment (TME). The important role of mtDNA in cancer development makes it a significant target for cancer treatment. In recent years, many novel therapeutic methods targeting mtDNA have emerged. In this study, we first discussed how cancerogenesis is triggered by mtDNA mutations, including alterations in gene copy number, aberrant gene expression and epigenetic modifications. Then, we described in detail the mechanisms underlying the interactions between mtDNA and the extramitochondrial environment, which are crucial for understanding the efficacy and safety of mtDNA-targeted therapy. Next, we provided a comprehensive overview of the recent progress in cancer therapy strategies that target mtDNA. We classified them into two categories based on their mechanisms of action: indirect and direct targeting strategies. Indirect targeting strategies aimed to induce mtDNA damage and dysfunction by modulating pathways that are involved in mtDNA stability and integrity, while direct targeting strategies utilized molecules that can selectively bind to or cleave mtDNA to achieve the therapeutic efficacy. This study highlights the importance of mtDNA-targeted therapy in cancer treatment, and will provide insights for future research and development of targeted drugs and therapeutic strategies.
... This finding defines that mitochondrial fission may act as a tumor suppressor which implicates for a potential therapeutic approach in GBM. 58 Raddeanin (RA), an active pharmacological component isolated from Anemone raddeana regel, induces apoptosis by an increase in the production of intracellular ROS and activation of Jun N-terminal kinase (JNK) in an in vitro study. 59 It has also shown that RA could induce autophagy in GBM cell lines and inhibits GBM in BALB/C nude mice. ...
Article
Full-text available
Glioblastoma multiforme (GBM) is one of the most common aggressive and fatal forms of adult brain tumors. According to WHO classification, GBM is usually classified as a grade IV form of a brain tumor. Glioblastoma multiforme exhibits high intra- and inter-tumoral heterogeneity. Free radical generation in GBM plays a robust role in promoting and inducing inflammatory processes mediated by various signaling pathways mainly focusing on Janus-kinases (JAK). Phosphatidylinositol-3-kinase (PI3K)/AKT/mammalian target of rapamycin (mTOR) stimulates signal transducer activating transcription factor 3 (STAT3) and JNK to induce proinflammatory cytokines, such as, interleukin (IL)-2, IL-6, and IL-8 to aggravate the inflammatory process. This review summarizes the convergence of inflammation and oxidative stress and examines the potential therapeutic targets aimed at the molecular markers in GBM.
... In addition to the above studies, Hippo pathway further regulates apoptosis by regulating mitochondrial homeostasis (Figure 4). Mitochondrial division mediates oxidative stress, reduces mitochondrial respiratory complex function, increases mitochondrial permeability, and promotes the leakage of pro-apoptotic molecule cyt-c into the nucleus and cytoplasm, leading to caspase-9-related mitochondrial apoptosis [112]. The Hippo pathway interacts with mitochondrial homeostasis to mediate injury and apoptosis of cardiomyocytes, neurons, and so on. ...
Article
Hippo pathway is a chain of kinases consists of a series of protein kinases and transcription factors. Meanwhile, oxidative stress is a condition of elevated concentrations of reactive oxygen species (ROS) that cause molecular damage to vital structures and functions. Both of them are key regulators in cell proliferation, survival, and development. These processes are strictly regulated by highly coordinated mechanisms, including c-Jun n-terminal kinase (JNK) pathway, mTOR pathway and a number of extrinsic and intrinsic factors. Recently, emerging evidence suggests that Hippo pathway is involved in the responses to cellular stresses, including mechanic stress, DNA damage, and oxidative stress, to mediate biological process, such as apoptosis, pyroptosis, and metastasis. But the exact mechanism remains to be further explored. Therefore, the purpose of this review is to summarize recent findings and discuss how Hippo pathway, oxidative stress, and the crosstalk between them regulate some biological process which determines cell fate.
... GAPdH and U6 served as the internal controls for mRNA and miRNA detection, respectively. The relative gene expression was calculated using the 2 -ΔΔCq method (24). The primer sequences used in qPCR were as follows: Stem-loop primer, 5'-cTc AAc TGG TGT cGT GGA GTc GGc AAT TcA GTT GAG TcA TGT Ac-3'; miR-3651 forward, 5'-Gcc GAG cAT AGc ccG GTc Gc-3' , and reverse, 5'-cTc AAc TGG TGT cGT GGA-3'; U6 forward, 5'-cTc GcT TcG GcA GcA cA-3', and reverse, 5'-AAc GcT TcA cGA ATT TGc GT-3'; TBX1 forward, 5'-AcG AcA AcG Gcc AcA TTA TTc-3' , and reverse, 5'-ccT cGG cAT ATT TcT cGc TAT cT-3'; GAPdH forward, 5'-AcA AcT TTG GTA TcG TGG AAG G-3', and reverse, 5'-Gcc ATc AcG ccA cAG TTT c-3'. ...
Article
Colorectal cancer is a commonly diagnosed gastrointestinal malignancy worldwide with a high mortality rate. Accumulating evidence has indicated that the expression of a number of microRNAs (miRNAs) is associated with the development of colorectal cancer. However, the precise molecular mechanism of these miRNAs in regulating cancer progression is yet to be determined. In the present study, miR‑3651 was demonstrated to be overexpressed in colorectal cancer tissues compared with normal tissues, and to be associated with the tumor‑node‑metastasis stage. The downregulation of miR‑3651 was found to induce growth arrest and apoptosis in colorectal cancer cells. In addition, western blot analysis demonstrated that the downregulation of miR‑3651 inactivated PI3K/AKT and MAPK/ERK signaling in colorectal cancer cells. Bioinformatics analysis predicted T‑box transcription factor 1 (TBX1) as a potential target gene of miR‑3651, and a dual‑luciferase reporter assay confirmed that TBX1 was directly repressed by miR‑3651. The results of the current study also indicated that TBX1 was associated with the miR‑3651 mediated activation of oncogenic signaling and colorectal cancer cell proliferation. In conclusion, the results of the current study revealed the oncogenic potential of miR‑3651 in colorectal cancer.
... For example, Lei Q et al found that the MAPK-ERK-YAP axis was involved in MA-5mediated Bnip3 upregulation and mitophagy activation [23]. One study in glioblastoma showed that TNFα stimulation decreased p-ERK expression, led to reduction of YAP1 expression [24]. In primary hPDLF cells, inhibition of ERK decreased total YAP amount in biomechanical strained cells but did not influence distribution of YAP1 in nuclear or cytoplasm [25]. ...
Article
Full-text available
The mitogen-activated protein kinase/extracellular signal-regulated (MAPK/ERK) pathway is a well-characterized signaling pathway during the development of various cancer types. ERK1 and ERK2, the two kinase effectors of MAPK cascade, exhibit high similarity. However, it is still unknown whether these two kinases are functionally different or in contrast functionally redundant during the development of breast cancer. We found that ERK1 expression levels were significantly lower in basal breast cancer compared with luminal breast cancer and normal breast tissues. RNA sequencing data suggested that ERK1 was associated with Hippo signaling pathway and cell proliferation in breast cancer cells. The gene set enrichment analysis (GSEA) further showed enrichment for YAP1 signaling pathway in breast cancer cell lines and tumors with low expression of ERK1. Silencing of ERK1 elevated YAP1 expression and TEAD activity in breast cancer cells. Additionally, ERK1 inhibited breast cancer cell proliferation via regulation of YAP1. The Kaplan-Meier analysis of data in patients with breast cancer suggested that, higher expression of ERK1 was associated with better prognosis, whereas, higher expression of ERK2 predicted poorer prognosis. These findings unveiled the role of ERK1 on regulation of YAP1 signaling pathway, indicating ERK1 as a negative regulator of breast cancer progression.
Article
Scope: Our previous in vivo studies showed Ganoderma atrum polysaccharide (PSG-F2 ) has a protective effect against the acrylamide (AA)-induced intestinal oxidative damage in rats. Now, we aimed to explore the protective mechanism with IEC-6 cell model. Methods and results: Based on RNA Sequencing (RNA-Seq), we screened MAPK signaling pathway as one of the most crucial pathways for pretreatment with PSG-F2 in AA-induced damage in IEC-6 cells. In total, six key MAPK signaling pathway-related proteins (p-P38/P38, p-ERK/ERK, and p-JNK/JNK), and three tight junction key proteins (Zonula Occludens protein-1, Claudin-1, and Occludin) were detected by Western blot and immunofluorescence, which verified the RNA-Seq data. Moreover, PD98059 interference inhibited critical proteins in the MAPK signaling pathway, thus uncovering the precise molecular mechanisms of MAPK/ERK signaling pathway involved in the protective effects of PSG-F2 against AA-induced intestinal barrier damage. Conclusion: These finding confirmed that PSG-F2 could be used as a daily dietary supplement to protect the intestinal cells from damage caused by thermal processing hazards AA. This article is protected by copyright. All rights reserved.
Chapter
Glioblastomas (GBMs) secrete hundreds of molecules that facilitate their growth and invasiveness. GBMs also release numerous pro- and antiinflammatory cytokines, checkpoint molecules and other molecules in order to cause profound, local and systemic, immunosuppression, a mechanism by which they can resist chemotherapy, radiation therapy, vaccines, and other treatments. This is thought to be a major factor responsible for the extremely poor prognosis of patients with GBM. In this chapter, we look closely at some of the most important of these immunosuppressive mechanisms, as well as at potential ways of increasing GBM patient survival by reversing this immunosuppression.
Article
Study design: Animal experimental study with intervention. Objective: To investigate the effect of melatonin on rat radiculopathy model and dorsal root ganglion (DRG) cells, and to elucidate the underlying mechanism. Summary of background data: Melatonin has a well-documented efficacy in intervertebral disc degeneration (IVDD) and low back pain. IVDD can also lead to other complications such as disc herniation which will cause radiculopathy. Herniated nucleus pulposus (NP) induced apoptosis and NLR pyrin domain containing 3 (NLRP3) activation in DRG. However, the effect and underlying mechanism of melatonin on radiculopathy and DRG cells are still unclear. Methods: Rat radiculopathy model was induced by implanting NP tissue from the tail disc of the same rat into the left L4/5 inter-laminar space near the left DRG. Melatonin was injected intraperitoneally in the treated group to test its function. Apoptosis was determined by Tunnel staining and flow cytometry. NLRP3 inflammasome activation was determined by levels of NLRP3, ASC, GSMDM-N, IL-1β, and Caspase-1. Mitophagy was determined by levels of Parkin, Beclin-1, p62, and LCB-II. Mitophagy was blocked by treatment with Parkin-si or cyclosporine A (CsA). Results: NLRP3 was significantly upregulated in DRG of rat radiculopathy model; moreover, melatonin markedly decreased pain behavior in rat radiculopathy model. Furthermore, melatonin treatment decreases the incidence of apoptosis in DRG cells. Melatonin also promotes mitophagy and inhibits NLRP3 inflammasomes in DRG cells. In addition, mitophagy was blocked by treatment with Parkin-si and CsA. Both Parkin-si and CsA attenuated melatonin's inhibitory effect on apoptosis and the NLRP3 inflammasome, indicating that the beneficial effects of melatonin in DRG cells are mediated through the Parkin-mediated mitophagy. Conclusion: Melatonin alleviates radiculopathy against apoptosis and NLRP3 inflammasomes by promoting Parkin-mediated mitophagy, which may help us provide a potential target for the treatment of radiculopathy.Level of Evidence: N/A.
Article
Full-text available
Increased mitochondrial damage is related to the progression of a diet-induced nonalcoholic fatty liver disease. The aim of our study is to investigate the role of Sirtuin 3 (Sirt3) in treating nonalcoholic fatty liver disease with a focus on mitophagy and the ERK-CREB pathway. Our data indicated that Sirt3 was downregulated in liver tissue in response to chronic HFD treatment. Interestingly, re-introduction of Sirt3 protected hepatic function, attenuated liver fibrosis, alleviated the inflammatory response, and prevented hepatocyte apoptosis. Molecular investigations demonstrated that lipotoxicity was associated with an increase in mitochondrial apoptosis as evidenced by reduced mitochondrial potential, augmented ROS production, increased cyt-c leakage into the nucleus, and activated caspase-9 apoptotic signalling. Additionally, Sirt3 overexpression protected hepatocytes against mitochondrial apoptosis via promoting Bnip3-required mitophagy. Functional studies showed that Sirt3 reversed Bnip3 expression and mitophagy activity via the ERK-CREB signalling pathway. Blockade of the ERK-CREB axis repressed the promotive effects of Sirt3 on Bnip3 activation and mitophagy augmentation, finally negating the anti-apoptotic influences of Sirt3 on hepatocytes in the setting of high-fat-stress. Collectively, our data show that high-fat-mediated liver damage is associated with Sirt3 downregulation, which is followed by ERK-CREB pathway inactivation and Bnip3-mediated inhibition of mitophagy, causing hepatocytes to undergo mitochondria-dependent cell death. Based on this, strategies for enhancing Sirt3 activity and activating the ERK-CREB-Bnip3-mitophagy pathways could be used to treat nonalcoholic fatty liver disease.
Article
Full-text available
The mitochondrial and endoplasmic reticulum (ER) homeostasis is pivotal to the maintenance of an array of physiological processes. The physical contact and association between ER and mitochondria, known as the ER–mitochondria microdomains or mitochondria-associated ER membrane (MAM), temporally and spatially regulates the mitochondria/ER structure and function. More evidence suggests a role for MAMs in energy production, cellular contraction and mobility, and normal extracellular signal transmission. In pathological states, such as cardiac ischemia–reperfusion (I/R injury), this ER–mitochondria microdomains may act to participate in the cellular redox imbalance, ER stress, mitochondrial injury, energy deletion, and programmed cell death. From a therapeutic perspective, a better understanding of the cellular and molecular mechanisms of the pathogenic ER–mitochondria contact should help to identify potential therapeutic target for cardiac I/R injury and other cardiovascular diseases and also pave the road to new treatment modalities pertinent for the treatment of reperfusion damage in clinical practice. This review will mainly focus on the possible signaling pathways involved in the regulation of the ER–mitochondria contact. In particular, we will summarize the downstream signaling modalities influenced by ER–mitochondria microdomains, for example, mitochondrial fission, mitophagy, calcium balance, oxidative stress, and programmed cell death in details.
Article
Full-text available
Dendritic cells orchestrate the crosstalk between innate and adaptive immunity. CD8α+ dendritic cells present antigens to CD8+ T cells and elicit cytotoxic T cell responses to viruses, bacteria and tumours 1 . Although lineage-specific transcriptional regulators of CD8α+ dendritic cell development have been identified 2 , the molecular pathways that selectively orchestrate CD8α+ dendritic cell function remain elusive. Moreover, metabolic reprogramming is important for dendritic cell development and activation3,4, but metabolic dependence and regulation of dendritic cell subsets are largely uncharacterized. Here we use a data-driven systems biology algorithm (NetBID) to identify a role of the Hippo pathway kinases Mst1 and Mst2 (Mst1/2) in selectively programming CD8α+ dendritic cell function and metabolism. Our NetBID analysis reveals a marked enrichment of the activities of Hippo pathway kinases in CD8α+ dendritic cells relative to CD8α- dendritic cells. Dendritic cell-specific deletion of Mst1/2-but not Lats1 and Lats2 (Lats1/2) or Yap and Taz (Yap/Taz), which mediate canonical Hippo signalling-disrupts homeostasis and function of CD8+ T cells and anti-tumour immunity. Mst1/2-deficient CD8α+ dendritic cells are impaired in presentation of extracellular proteins and cognate peptides to prime CD8+ T cells, while CD8α- dendritic cells that lack Mst1/2 have largely normal function. Mechanistically, compared to CD8α- dendritic cells, CD8α+ dendritic cells exhibit much stronger oxidative metabolism and critically depend on Mst1/2 signalling to maintain bioenergetic activities and mitochondrial dynamics for their functional capacities. Further, selective expression of IL-12 by CD8α+ dendritic cells depends on Mst1/2 and the crosstalk with non-canonical NF-κB signalling. Our findings identify Mst1/2 as selective drivers of CD8α+ dendritic cell function by integrating metabolic activity and cytokine signalling, and highlight that the interplay between immune signalling and metabolic reprogramming underlies the unique functions of dendritic cell subsets.
Article
Full-text available
Cancer cells display differences regarding their engagement of glycolytic vs. mitochondrial oxidative phosphorylation (OXPHOS) pathway. Triple negative breast cancer, an aggressive form of breast cancer, is characterized by elevated glycolysis, while estrogen receptor positive breast cancer cells rely predominantly on OXPHOS. BCL2 proteins control the process of mitochondrial outer membrane permeabilization during apoptosis, but also regulate cellular bioenergetics. Because BCL2 proteins are overexpressed in breast cancer and targetable by selective antagonists, we here analysed the effect of BCL2 and BCL(X)L selective inhibitors, Venetoclax and WEHI- 539, on mitochondrial bioenergetics and cell death. Employing single cell imaging using a FRET-based mitochondrial ATP sensor, we found that MCF7 breast cancer cells supplied with mitochondrial substrates reduced their mitochondrial ATP production when treated with Venetoclax or WEHI-539 at concentrations that per se did not induce cell death. Treatments with lower concentrations of both inhibitors also reduced the length of the mitochondrial network and the dynamics, as evaluated by quantitative confocal microscopy. We next tested the hypothesis that mitochondrial ATP production inhibition with BCL2 or BCL(X)L antagonists was synthetically lethal when combined with glycolysis inhibition. Treatment with 2-deoxy-D-glucose in combination with Venetoclax or WEHI-539 synergistically reduced the cellular bioenergetics of ER+ and TNBC breast cancer cells and abolished their clonogenic potential. Synthetic lethality was also observed when cultures were grown in 3D spheres. Our findings demonstrate that BCL2 antagonists exert potent effects on cancer metabolism independent of cell death-inducing effects, and demonstrate a synthetic lethality when these are applied in combination with glycolysis inhibitors.
Article
Full-text available
The taxanes are effective microtubule-stabilizing chemotherapy drugs that inhibit mitosis, induce apoptosis, and produce regression in a fraction of cancers that arise at many sites including the ovary. Novel therapeutic targets that augment taxane effects are needed to improve clinical chemotherapy response in CCNE1-amplified high grade serous ovarian cancer (HGSOC) cells. In this study, we conducted an siRNA-based kinome screen to identify modulators of mitotic progression in CCNE1-amplified HGSOC cells that may influence clinical paclitaxel response. PLK1 is overexpressed in many types of cancer, which correlates with poor prognosis. Here, we identified a novel synthetic lethal interaction of the clinical PLK1 inhibitor BI6727 and the microtubuletargeting drug paclitaxel in HGSOC cell lines with CCNE1-amplification and elucidated the underlying molecular mechanisms of this synergism. BI6727 synergistically induces apoptosis together with paclitaxel in different cell lines including a patient-derived primary ovarian cancer culture. Moreover, the inhibition of PLK1 reduced the paclitaxelinduced neurotoxicity in a neurite outgrowth assay. Mechanistically, the combinatorial treatment with BI6727/paclitaxel triggers mitotic arrest, which initiates mitochondrial apoptosis by inactivation of anti-apoptotic BCL-2 family proteins, followed by significant loss of the mitochondrial membrane potential and activation of caspase-dependent effector pathways. This conclusion is supported by data showing that BI6727/ paclitaxel-co-treatment stabilizes FBW7, a component of SCF-type ubiquitin ligases that bind and regulate key modulators of cell division and growth including MCL-1 and Cyclin E. This identification of a novel synthetic lethality of PLK1 inhibitors and a microtubule-stabilizing drug has important implications for developing PLK1 inhibitorbased combination treatments in CCNE1-amplified HGSOC cells.
Article
Full-text available
The molecular features of necroptosis in cardiac ischemia-reperfusion (IR) injury have been extensively explored. However, there have been no studies investigating the physiological regulatory mechanisms of melatonin acting on necroptosis in cardiac IR injury. This study was designed to determine the role of necroptosis in microvascular IR injury, and investigate the contribution of melatonin in repressing necroptosis and preventing IR-mediated endothelial system collapse. Our results demonstrated that Ripk3 was primarily activated by IR injury and consequently aggravated endothelial necroptosis, microvessel barrier dysfunction, capillary hyperpermeability, the inflammation response, microcirculatory vasospasms, and microvascular perfusion defects. However, administration of melatonin prevented Ripk3 activation and provided a pro-survival advantage for the endothelial system in the context of cardiac IR injury, similar to the results obtained via genetic ablation of Ripk3. Functional investigations clearly illustrated that activated Ripk3 upregulated PGAM5 expression, and the latter increased CypD phosphorylation, which obligated endothelial cells to undergo necroptosis via augmenting mPTP (mitochondrial permeability transition pore) opening. Interestingly, melatonin supplementation suppressed mPTP opening and interrupted endothelial necroptosis via blocking the Ripk3-PGAM5-CypD signal pathways. Taken together, our studies identified the Ripk3-PGAM5-CypD-mPTP axis as a new pathway responsible for reperfusion-mediated microvascular damage via initiating endothelial necroptosis. In contrast, melatonin treatment inhibited the Ripk3-PGAM5-CypD-mPTP cascade and thus reduced cellular necroptosis, conferring a protective advantage to the endothelial system in IR stress. These findings establish a new paradigm in microvascular IR injury and update the concept for cell death management handled by melatonin under the burden of reperfusion attack.
Article
Full-text available
Mitochondrial fission and mitophagy are considered key processes involved in the pathogenesis of cardiac microvascular ischemia reperfusion (IR) injury although the upstream regulatory mechanism for fission and mitophagy still remains unclear. Herein, we reported that NR4A1 was significantly upregulated following cardiac microvascular IR injury, and its level was positively correlated with microvascular collapse, endothelial cellular apoptosis and mitochondrial damage. However, NR4A1-knockout mice exhibited resistance against the acute microvascular injury and mitochondrial dysfunction compared with the wild-type mice. Functional studies illustrated that IR injury increased NR4A1 expression, which activated serine/threonine kinase casein kinase2 α (CK2α). CK2α promoted phosphorylation of mitochondrial fission factor (Mff) and FUN14 domain-containing 1 (FUNDC1). Phosphorylated activation of Mff enhanced the cytoplasmic translocation of Drp1 to the mitochondria, leading to fatal mitochondrial fission. Excessive fission disrupted mitochondrial function and structure, ultimately triggering mitochondrial apoptosis. In addition, phosphorylated inactivation of FUNDC1 failed to launch the protective mitophagy process, resulting in the accumulation of damaged mitochondria and endothelial apoptosis. By facilitating Mff-mediated mitochondrial fission and FUNDC1-required mitophagy, NR4A1 disturbed mitochondrial homeostasis, enhanced endothelial apoptosis and provoked microvascular dysfunction. In summary, our data illustrated that NR4A1 serves as a novel culprit factor in cardiac microvascular IR injury that operates through synchronous elevation of fission and suppression of mitophagy. Novel therapeutic strategies targeting the balance among NR4A1, fission and mitophagy might provide survival advantage to microvasculature following IR stress.
Article
Full-text available
Mitochondrial fission is important for the development and progression of pancreatic cancer (PC). However, little is known regarding its role in pancreatic cancer apoptosis, metabolism and migration. In the current study, the mechanism by which mitochondrial fission modifies the biological characteristics of PC was explored. MicroRNA‑125a (miR‑125a) had the ability to inhibit mitochondrial fission and contributed to cellular survival. Suppressed mitochondrial fission led to a reduction in mitochondrial debris, preserved the mitochondrial membrane potential, inhibited mitochondrial permeability transition pore opening, ablated cytochrome c leakage into the cytoplasm and reduced the pro‑apoptotic protein contents, finally blocking mitochondria related apoptosis pathways. Furthermore, defective mitochondrial fission induced by miR‑125a enhanced mitochondria‑dependent energy metabolism by promoting activity of electron transport chain complexes. Furthermore, suppressed mitochondrial fission also contributed to PANC‑1 cell migration by preserving the F‑actin balance. Furthermore, mitofusin 2 (Mfn2), the key defender of mitochondrial fission, is involved in inhibition of miR125a‑mediated mitochondrial fission. Low contents of miR‑125a upregulated Mfn2 transcription and expression, leading to inactivation of mitochondrial fission. Ultimately, the current study determined that miR‑125a and Mfn2 are regulated by hypoxia‑inducible factor 1 (HIF1). Knockdown of HIF1 reversed miR‑125a expression, and therefore, inhibited Mfn2 expression, leading to activation of mitochondrial fission. Collectively, the present study demonstrated mitochondrial fission as a tumor suppression process that is regulated by the HIF/miR‑125a/Mfn2 pathways, acting to restrict PANC‑1 cell survival, energy metabolism and migration, with potential implications for novel approaches for PC therapy.
Article
Full-text available
Background: Mitochondrial fission has been identified as the pathogenesis underlying the development of cardiac microvascular ischemia reperfusion (IR) injury, although the regulatory signaling upstream from fission is far from clear. Bax inhibitor is a novel anti-apoptotic factor, and, however, its role of cardiac microvascular IR injury and mitochondrial homeostasis remains unclear. Methods: The cardiac microvascular IR injury was performed in WT mice and BI1 transgenic (BITG) mice. The alterations of microvascular structure and function were detected via electron microscope, immunohistochemistry and immunofluorescence in vivo. Cardiac microvascular endothelial cells were isolated form WT and BITGmice and underwent hypoxia/reoxygenation injury in vitro. Cellular viability and apoptosis were analyzed via MTT assay and caspase-3 activity. Mitochondrial function, morphology and apoptosis were detected. Signaling pathways were analyzed via inhibitor, siRNA and mutant plasmid. Results: Herein, we demonstrated that Bax inhibitor 1 (BI1) was downregulated following cardiac microvascular IR injury, and its expression correlated negatively with microvascular collapse, endothelial cell apoptosis and mitochondrial damage. However, compared to wild-type mice, BI1 transgenic mice were actually protected from the acute microvascular injury and mitochondrial dysfunction. Functional studies illustrated that reintroduced BI1 directly interacted with and inhibited the Syk pathway, leading to the inactivation of Nox2. Subsequently, less Nox2 was associated with ROS downregulation, inhibiting Drp1 phosphorylated activation. Through repression of the Syk-Nox2-Drp1 signaling axis, BI1 strongly disrupted mitochondrial fission, abolishing mitochondrial apoptosis and thus sustaining endothelial cell viability. Conclusions: In summary, our report illustrates that BI1 functions as a novel microvascular guardian in cardiac IR injury that operates via inhibition of the Syk-Nox2-Drp1-mitochondrial fission signaling axis. Thus, novel therapeutic strategies to regulate the balance between BI1 and mitochondrial fission could provide a survival advantage to microvasculature following IR stress.
Article
Yes-associated protein (Yap), a regulator of cellular apoptosis, has been demonstrated to be involved in cerebral ischemia–reperfusion (IR) injury through poorly defined mechanisms. The present study aimed to explore the role of Yap in regulating cerebral IR injury in vitro, with a focus on mitochondrial fission and ROCK1/F-actin pathways. Our data demonstrated that Yap was actually downregulated in N2a cells after cerebral hypoxia-reoxygenation (HR) injury, and that lower expression of Yap was closely associated with increased cell death. However, the reintroduction of Yap was able to suppress the HR-mediated N2a cells death via blocking the mitochondria-related apoptotic signal. At the molecular levels, Yap overexpression sustained mitochondrial potential, normalized the mitochondrial respiratory function, reduced ROS overproduction, limited HtrA2/Omi release from mitochondria into the nucleus, and suppressed pro-apoptotic proteins activation. Subsequently, functional studies have further illustrated that HR-mediated mitochondrial apoptosis was highly regulated by mitochondrial fission, whereas Yap overexpression was able to attenuate HR-mediated mitochondrial fission and, thus, promote N2a cell survival in the context of HR injury. At last, we demonstrated that Yap handled mitochondrial fission via closing ROCK1/F-actin signaling pathways. Activation of ROCK1/F-actin pathways abrogated the protective role of Yap overexpression on mitochondrial homeostasis and N2a cell survival in the setting of HR injury. Altogether, our data identified Yap as the endogenous defender to relieve HR-mediated nerve damage via antagonizing ROCK1/F-actin/mitochondrial fission pathways.