Skip to main content
Erschienen in: Cancer Cell International 1/2018

Open Access 01.12.2018 | Primary research

RETRACTED ARTICLE: Excessive mitochondrial fragmentation triggered by erlotinib promotes pancreatic cancer PANC-1 cell apoptosis via activating the mROS-HtrA2/Omi pathways

verfasst von: Jun Wan, Jie Cui, Lei Wang, Kunpeng Wu, Xiaoping Hong, Yulin Zou, Shuang Zhao, Hong Ke

Erschienen in: Cancer Cell International | Ausgabe 1/2018

Abstract

Background

Mitochondrial fragmentation drastically regulates the viability of pancreatic cancer through a poorly understood mechanism. The present study used erlotinib to activate mitochondrial fragmentation and then investigated the downstream events that occurred in response to mitochondrial fragmentation.

Methods

Cell viability and apoptosis were determined via MTT assay, TUNEL staining and ELISA. Mitochondrial fragmentation was measured via an immunofluorescence assay and qPCR. siRNA transfection and pathway blockers were used to perform the loss-of-function assays.

Results

The results of our study demonstrated that erlotinib treatment mediated cell apoptosis in the PANC-1 pancreatic cancer cell line via evoking mitochondrial fragmentation. Mechanistically, erlotinib application increased mitochondrial fission and reduced mitochondrial fusion, triggering mitochondrial fragmentation. Subsequently, mitochondrial fragmentation caused the overproduction of mitochondrial ROS (mROS). Interestingly, excessive mROS induced cardiolipin oxidation and mPTP opening, finally facilitating HtrA2/Omi liberation from the mitochondria into the cytoplasm, where HtrA2/Omi activated caspase-9-dependent cell apoptosis. Notably, neutralization of mROS or knockdown of HtrA2/Omi attenuated erlotinib-mediated mitochondrial fragmentation and favored cancer cell survival.

Conclusions

Together, our results identified the mROS-HtrA2/Omi axis as a novel signaling pathway that is activated by mitochondrial fragmentation and that promotes PANC-1 pancreatic cancer cell mitochondrial apoptosis in the presence of erlotinib.
Hinweise

Electronic supplementary material

The online version of this article (https://​doi.​org/​10.​1186/​s12935-018-0665-1) contains supplementary material, which is available to authorized users.
The authors have retracted this article [1] because Figure 4a has been duplicated from Figure 5d in a previously published article [2]. In addition, the article contains sections that substantially overlap with the following articles (amongst others) [3, 4, 5]. The data reported in this article are therefore unreliable. Author Hong Ke stated on behalf of all co-authors that they agree to this retraction.
1. Wan, J., Cui, J., Wang, L. et al. Excessive mitochondrial fragmentation triggered by erlotinib promotes pancreatic cancer PANC-1 cell apoptosis via activating the mROS-HtrA2/Omi pathways. Cancer Cell Int 18, 165 (2018) https://doi.org/10.1186/s12935-018-0665-1
2. Liu J., Yan, W., Zhao X. et al. Sirt3 attenuates post-infarction cardiac injury via inhibiting mitochondrial fission and normalization of AMPK-Drp1 pathways, Cellular Signalling 53, 1 (2019) https://doi.org/10.1016/j.cellsig.2018.09.009
3. He, M., Xiang, Z., Xu, L. et al. Lipopolysaccharide induces human olfactory ensheathing glial apoptosis by promoting mitochondrial dysfunction and activating the JNK-Bnip3-Bax pathway, Cell Stress and Chaperones 24, 91 (2019) https://doi.org/10.1007/s12192-018-0945-7
4. 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 109, 679 (2019) https://doi.org/10.1016/j.biopha.2018.10.097
5. Chen, Z., Wang, C., Yu, N. et al. INF2 regulates oxidative stress-induced apoptosis in epidermal HaCaT cells by modulating the HIF1 signaling pathway, Biomedicine & Pharmacotherapy 111, 151 (2019) https://doi.org/10.1016/j.biopha.2018.12.046
A correction to this article is available online at https://​doi.​org/​10.​1186/​s12935-019-1019-3.

Background

Pancreatic cancer is the fourth leading cause of cancer-related death worldwide [1]. Although the incidence of pancreatic cancer is relatively low, approximately 3.2% of all new cancer cases in the United States, the 5-year survival rate is 8.5% in patients diagnosed with pancreatic cancer. In addition, the detection rate of early pancreatic cancer remains low due to the lack of specific symptoms. Accordingly, most patients (52%) are diagnosed with distant metastasis [2], and, unfortunately, the 5-year relative survival of patients with metastatic pancreatic cancer is less than 2% [3]. Although smoking and health history can affect the risk of pancreatic cancer, the pathogenesis of pancreatic cancer development is not completely understood. Therefore, exploring the molecular features of pancreatic cancer growth and death is vital to control the disease progression and bring more clinical benefits to patients with pancreatic cancer.
The biological behavior of cancer is closely regulated by mitochondria [4, 5]. Sufficient ATP supply, intracellular calcium homeostasis, metabolic signaling transduction, and cell apoptosis management are affected by mitochondria [68]. In addition, mitochondria are also the key target of several chemotherapeutics and radiotherapies [9]. A recent study has reported that pancreatic cancer death, proliferation and metastasis are modulated by mitochondrial homeostasis, especially mitochondrial fission [10]. Excessive mitochondrial fission induces cancer cell oxidative injury and subsequently mediates mitochondrial ATP depletion; this effect impairs PANC-1 cell proliferation and evokes mitochondrial apoptosis [10]. Notably, this conclusion is also supported by other studies. In colorectal cancer, the activation of mitochondrial fission is associated with SW837 cell apoptosis and migration inhibition [11]. In gastric cancer, abnormal mitochondrial fission contributes to cancer cell oxidative stress and energy undersupply [12]. In breast cancer, Drp1-mediated mitochondrial fission suppresses breast cancer cell invasion [13]. This information indicates that mitochondrial fission has a well-characterized role in the regulation of cancer viability. However, the downstream molecular events of mitochondrial fission activation remain to be discovered.
Based on a previous study in a mouse model of cardiac ischemia reperfusion injury, the activation of mitochondrial fission promotes the formation of mitochondrial fragmentation, and these mitochondrial debris contain a decreased mitochondrial potential [14]. In addition, mitochondrial fragmentation can activate cell death via two mechanisms [15]; one mechanism is driven via HK2/VDAC1 disassociation-mediated mPTP opening, and the other involves mROS-induced cardiolipin oxidation. Notably, mitochondrial ROS (mROS) overloading, as a primary result of mitochondrial fragmentation [16], has been noted in different disease models such as those of gastric cancer [17], breast cancer [18], and leukemia [19]. Subsequently, excessive mitochondrial oxidative injury can activate the HtrA2/Omi-related apoptotic pathway in a manner that is dependent on caspase-9 activity [11]. This evidence indicates that the downstream effectors of mitochondrial fragmentation include mROS overproduction, HtrA2/Omi upregulation, caspase-9 activation and mitochondrial apoptosis augmentation. Given these factors, we want to know whether mitochondrial fragmentation regulates pancreatic cancer viability via mROS-HtrA2/Omi-caspase-9 pathways.
To this end, erlotinib is the first-line anti-tumor drug for the treatment of pancreatic cancer in the clinic [20]. Several human studies have verified the efficacy of erlotinib in improving the 5-year survival rate of patients with pancreatic cancer [21, 22]. Molecular investigations report that several biological processes are modulated by erlotinib, including mTOR inhibition [23], epidermal growth factor receptor downregulation [24], and epidermal interstitial transformation (EMT) suppression [25]. However, no study that explores the role of erlotinib in triggering mitochondrial stress has been conducted. In the present study, erlotinib was applied to activate mitochondrial fragmentation in a human PANC-1 pancreatic cancer cell line. Then, we explored the regulatory mechanism of mitochondrial fragmentation on cell viability in the presence of erlotinib.

Methods and materials

Pancreatic carcinoma cell lines

The PANC-1 (ATCC® CRL-1469™) and MIA PaCa-2 (ATCC® CRL-1420™) pancreatic cell lines were used in the present study. These cells were cultured in Dulbecco’s modified Eagle’s medium (DMEM) (Thermo Fisher Scientific, Waltham, MA) supplemented with 10% fetal bovine serum (FBS) (Thermo Fisher Scientific, Waltham, MA) at 37 °C in a 5% CO2 atmosphere. Different doses of erlotinib (ERL, Sigma. Cat. No. SML2156) were incubated with the cancer cells for 24 h, and these concentrations of ERL were chosen according to a previous study [26]. FCCP (5 μm, Selleck Chemicals, Houston, TX, USA) and mitochondrial division inhibitor 1 (Mdivi1; 10 mM; Sigma-Aldrich; Merck KGaA) were used to activate and inhibit mitochondrial fragmentation, respectively, according to a previous study. To repress mROS overproduction, mitochondrial-targeted antioxidant MitoQ (2 μM, MedKoo Biosciences, Inc.; CAT#: 317102) was used.

Western blotting and antibodies

Cells were scraped in RIPA lysis buffer (Beyotime, Shenzhen, Guangdong, China). The lysates (50–70 µg) were separated by 10% SDS-polyacrylamide gel (10–15%) electrophoresis (SDS-PAGE). Proteins were electrotransferred onto the Pure Nitrocellulose Blotting membrane (Life Sciences) (Millipore, Bedford, MA, USA) and then blocked with 5% nonfat milk for 2 h at room temperature [27]. After washing with TBST three times, the membranes were incubated at 4 °C overnight with the following primary antibodies: HrtA2/Omi (:1000; Abcam; #ab32092), caspase9 (1:1000, Cell Signaling Technology, #9504), Bax (1:1000, Cell Signaling Technology, #2772), Opa1 (1:1000, Abcam, #ab42364), Mfn2 (1:1000, Abcam, #ab56889), Tom20 (1:1000, Abcam, #ab186735), CDK4 (1:1000, Abcam, #ab137675), Cyclin D1 (1:1000, Abcam, #ab134175), Bcl2 (1:1000, Cell Signaling Technology, #3498), Bad (:1000; Abcam; #ab90435), survivin (1:1000, Cell Signaling Technology, #2808), cyt-c (1:1000; Abcam; #ab90529), complex III subunit core (CIII-core2, 1:1000, Invitrogen, #459220), complex II (CII-30, 1:1000, Abcam, #ab110410), complex IV subunit II (CIV-II, 1:1000, Abcam, #ab110268). Next, the membranes were visualized using an enhanced chemiluminescence system (ECL; Pierce Company, USA) [28].

MTT assay, caspase activity detection and LDH release assay

MTT was used to analyze the cellular viability [29]. Cells (1 × 106 cells/well) were cultured on a 96-well plate at 37 °C with 5% CO2. Then, 40 µl of MTT solution (2 mg/ml; Sigma-Aldrich) was added to the medium for 4 h at 37 °C with 5% CO2. Subsequently, the cell medium was discarded, and 80 µl of DMSO was added to the wells for 1 h at 37 °C with 5% CO2 in the dark. The OD of each well was observed at A490 nm via a spectrophotometer (Epoch 2; BioTek Instruments, Inc., Winooski, VT, USA). To analyze changes in caspase-9, caspase-9 activity kits (Beyotime Institute of Biotechnology, China; Catalog No. C1158) were used according to the manufacturer’s protocol [30]. In brief, to measure caspase-9 activity, 5 μl of LEHD-p-NA substrate (4 mM, 200 μM final concentration) was added to the samples for 1 h at 37 °C. Then, the absorbance at 400 nm was recorded via a microplate reader to reflect the caspase-3 and caspase-9 activities. To analyze caspase-3 activity, 5 μl of DEVD-p-NA substrate (4 mM, 200 μM final concentration) was added to the samples for 2 h at 37 °C [31].

ELISA

Glutathione (GSH, Thermo Fisher Scientific Inc., Waltham, MA, USA; Catalog No. T10095), glutathione peroxidase GPX, (Beyotime Institute of Biotechnology, China; Catalog No. S0056) and SOD (Thermo Fisher Scientific Inc., Waltham, MA, USA; Catalog No. BMS222TEN) were measured according to the manufacturer’s instructions using a microplate reader (Epoch 2; BioTek Instruments, Inc.) [32]. Cellular ATP generation was measured to reflect mitochondrial function. Firstly, cells were washed three times with cold PBS at room temperature. Subsequently, a luciferase-based ATP assay kit (CellTiter-Glo® Luminescent Cell Viability Assay; cat. no. G7570; Promega Corporation, Madison, WI, USA) was used to analyze ATP content, according to the manufacturer’s protocols. ATP production was measured using a microplate reader at the wavelength of 570 nm (Epoch 2; BioTek Instruments, Inc., Winooski, VT, USA) [33].

Immunostaining

Cells were washed twice with PBS, permeabilized in 0.1% Triton X-100 overnight at 4 °C. After the fixation procedure, the sections were cryoprotected in a PBS solution supplemented with 0.9 mol/l of sucrose overnight at 4 °C [34]. The primary antibodies used in the present study were as follows: caspase9 (1:1000, Cell Signaling Technology, #9504), Mff (1:1000, Cell Signaling Technology, #86668), Tom20 (1:1000, Abcam, #ab186735), HrtA2/Omi (1:1000; Abcam; #ab32092).

Small interfering RNA transfection

To inhibit HtrA2/Omi expression, two independent siRNAs against HtrA2/Omi were transfected into PANC-1 cells according to a previous study [27]. Briefly, the cells were seeded onto 6-well plates and then incubated with Opti-Minimal Essential Medium (Invitrogen; Thermo Fisher Scientific, Inc.) for 24 h. Then, Lipofectamine® 2000 transfection reagent (Thermo Fisher Scientific, Inc.) was added into the medium of PANC-1 cells and supplemented with 5 nmol/l siRNA solution. Transfection was performed for 48 h, and then the cells were collected. Western blotting was used to verify the transfection efficiency.

Detection of mitochondrial membrane potential and mPTP opening

To observe the mitochondrial potential, JC-1 staining (Thermo Fisher Scientific Inc., Waltham, MA, USA; Catalog No. M34152) was used. Then, 10 mg/ml JC-1 was added to the medium for 10 min at 37 °C in the dark to label the mitochondria. Normal mitochondrial potential showed red fluorescence, and damaged mitochondrial potential showed green fluorescence [35]. The mPTP opening rate was detected using calcein-AM (Sigma, Cat. No. 17783) as described previously [36]. Briefly, cells were incubated with calcein-AM for 30 min at 37 °C in the dark. Next, PBS was used to wash the cells three times. Finally, the optical density (OD) at an absorbance of 579 nm was recorded using a multifunction microplate reader (Epoch 2; BioTek Instruments, Inc., Winooski, VT, USA). The mPTP opening rate was calculated as a ratio to that of the control group [14]. The relative mPTP opening was measured as a ratio to that of the control group.

TUNEL assay and cardiolipin staining

Apoptotic cells were detected with an In Situ Cell Death Detection Kit (Thermo Fisher Scientific Inc., Waltham, MA, USA; Catalog No. C1024) according to the manufacturer’s protocol. Briefly, cells were fixed with 4% paraformaldehyde at 37 °C for 15 min. Blocking buffer (3% H2O2 in CH3OH) was added to the wells, and then cells were permeabilized with 0.1% Triton X-100 in 0.1% sodium citrate for 2 min on ice. The cells were incubated with TUNEL reaction mixture for 1 h at 37 °C. DAPI (Sigma-Aldrich, St. Louis, MO, USA) was used to counterstain the nuclei, and the numbers of TUNEL-positive cells were recorded [37]. Cardiolipin oxidation was stained with 10-N-nonylacridine orange (NAO; 2 mmol/l; Molecular Probes, Eugene, OR, USA). Under normal conditions, NAO interacts with nonoxidized cardiolipin and generates a characteristic green fluorescence. However, upon cardiolipin oxidation, NAO cannot interact with cardiolipin, and this result is accompanied by a drop in green fluorescence. Accordingly, the green fluorescence intensity of NAO was used to quantify the cardiolipin oxidation with the help of Image-Pro Plus 6.0; Media Cybernetics, Rockville, MD, USA) [16].

RNA extraction and qPCR analysis

For mRNA expression analysis, total RNA was isolated using Trizol (Invitrogen, Carlsbad, California, USA) according to a previous study. Then, cDNA was synthesized using 1 mg RNA and the First-Strand Synthesis Kit (Fermentas, Flamborough, Ontario, Canada) according to a previous study [38]. The cycling conditions were as follows: 92 °C for 7 min, 40 cycles of 95 °C for 20 s and 70 °C for 45 s. β-actin was amplified as an internal standard. All the primer sequences are listed below: Drp1 (forward prime 5′-CATGGACGAGCTGGCCTTC-3′, reverse prime 5′-ATCCTGTAGTGATGTATCAGG-3′), Mff (forward prime 5′-TGTCCAGTCCGTAACTGAC-3′, reverse prime 5′-TTCGATACCTGACTTAC-3′), Mfn2 (forward prime 5′-CCTCTTGATCCTGATCTTAACGT-3′, reverse prime 5′-GGACTACCTGATTGTCATTC-3′), OPA1 (forward prime 5′-GCTACTTGTGAGGTCGATTC-3′, reverse prime 5′-GCCGTATACCGTGGTATGTCTG-3′) [14].

EdU staining

EdU staining was performed to analyze the cell proliferation according to a previous study [39]. The EdU incorporation assay was performed using the EdU kit (cat. no. A10044; Thermo Fisher Scientific Inc.). Briefly, EdU (2 nM/well) was diluted in complete culture medium, and the cells were incubated with the dilution for 2 h at 37 °C. Subsequently, the cells were fixed with 4% paraformaldehyde for 15 min at 37 °C and were incubated with Apollo Staining reaction liquid for 30 min. DAPI was used to counterstain the nuclei for 15 min at room temperature under a digital microscope system (IX81; Olympus Corporation).

Flow cytometry assay

Flow cytometry was applied as a quantitative method for evaluating mitochondrial ROS levels according to a previous study [4]. In brief, PANC-1 cells were seeded onto 6-well plates and then treated with erlotinib. Subsequently, the cells were isolated using 0.25% trypsin and then incubated with MitoSOX red mitochondrial superoxide indicator (Molecular Probes, USA) for 30 min in the dark at 37 °C. Subsequently, PBS was used to wash cell two times, and then the cells were analyzed with a FACS Calibur Flow cytometer. Data were analyzed by FACS Diva software. The experiment was repeated three times to improve the accuracy [39]. The number of apoptotic cells was analyzed quantitatively using the Annexin V–FITC/PI Apoptosis Detection Kit (BD Biosciences, USA). Aſter treatment, the cells were harvested, resuspended in 200 µl of binding buffer, and then incubated with 5 µl of Annexin V–FITC/binding buffer mixture (30 min, 37 °C) in the dark. Subsequently, the cells were incubated with 10 µl of propidium iodide for 5 min and immediately analyzed by bivariate flow cytometry using a BD FACSCalibur cytometer [36].

Statistical analysis

Data are expressed as the mean ± SE of triplicate samples. Statistical analysis for multiple comparisons was analyzed by a one-way analysis of variance (ANOVA) followed by Bonferroni’s multiple comparison test. p values below 0.05 were considered statistically significant.

Results

Erlotinib dose-dependently promotes PANC-1 pancreatic cancer cell apoptosis

First, erlotinib was incubated with PANC-1 pancreatic cancer cells. Then, cell viability was observed using the MTT assay and LDH-cytotoxicity assay. Compared to the control group, erlotinib treatment reduced the viability of PANC-1 cells (Fig. 1a, b), and this effect was achieved in a dose-dependent manner. This finding was also found in erlotinib-treated PaCa-2 pancreatic cancer cells (Fig. 1c, d). To explore whether the reduction in cell viability was attributable to excessive cell apoptosis, the TUNEL assay was used. The number of TUNEL-positive cells was counted as the apoptotic index. As shown in Fig. 1e, f, erlotinib dose-dependently increased the apoptotic index in PANC-1 cells. Similarly, the number of TUNEL-positive cells was also elevated in PaCa-2 cells upon exposure to erlotinib (Fig. 1e, g). Furthermore, since cell apoptosis is primarily executed via caspase-3 activation, caspase-3 activity was determined via ELISA. Compared to the control group, caspase-3 activity was relatively increased in response to erlotinib treatment (Fig. 1h), which is suggestive of caspase-3 activation by erlotinib. This alteration was also noted in PaCa-2 cells (Fig. 1I). These data were further supported via quantitative analysis of cell apoptosis with the help of flow cytometry (Additional file 1: Figure S1). Together, our results indicated that erlotinib dose-dependently promoted PANC-1 and PaCa-2 cell apoptosis. Notably, no phenotypic difference was noted in erlotinib-mediated apoptosis in PANC-1 cells or PaCa2 cells, and thus PANC-1 cells were used in the following study. In addition, we have found that the minimum concentration of erlotinib that induces cell death was 10 μM, and thus, 10 μM erlotinib was used to conduct the molecular investigations.

Erlotinib induces mitochondrial fragmentation in PANC-1 pancreatic cancer cells via elevating mitochondrial fission and repressing mitochondrial fusion

Subsequently, the mitochondrial morphology was observed via an immunofluorescence assay using a Tom-20 antibody [40]. Compared to the control group, we found that erlotinib treatment mediated the formation of mitochondrial fragmentation (Fig. 2a). Then, the average length of the mitochondria was measured after erlotinib treatment and was used to quantify mitochondrial fragmentation. As shown in Fig. 2b, the mean length of the mitochondria was ~ 9.1 μm at baseline. However, after treatment with erlotinib, the mean length of mitochondria was reduced to ~ 2.3 μm (Fig. 2b). In addition, the fluorescence intensity of Mff, an activator of mitochondrial fragmentation, was obviously increased in response to erlotinib treatment compared to that in control group (Fig. 2c). Subsequently, to further confirm the promotive effect of erlotinib on mitochondrial fragmentation, Mdivi-1, an antagonist of mitochondrial fragmentation, was added into the medium of erlotinib-treated cells. Meanwhile, FCCP, an agonist of mitochondrial fragmentation, was used to incubate with normal cells, which was used as the positive control group. Then, mitochondrial fission, mitochondrial length and Mff expression were evaluated again. Compared to the control group, FCCP triggered mitochondrial fragmentation and upregulated Mff expression, similar to the results obtained via supplementation with erlotinib (Fig. 2a–c). However, Mdivi-1 treatment abrogated the promotive effect of erlotinib on mitochondrial fragmentation.
Notably, the fragmented mitochondria could be the result of increased mitochondrial fission and decreased mitochondrial fusion. To verify the alterations of mitochondrial fission/fusion, qPCR was performed to analyze the transcription factors that are related to mitochondrial fission/fusion. In response to erlotinib treatment, the transcription of pro-fission factors such as Drp1 and Mff were significantly upregulated (Fig. 2d–g), indicative of mitochondrial fission activation by erlotinib. In contrast, the transcription and expression of pro-fusion factors, such as Mfn2 and Opa1 were obviously downregulated in response to erlotinib treatment (Fig. 2d–j), suggesting that mitochondrial fusion was repressed by erlotinib. Together, our results confirmed that erlotinib promoted mitochondrial fragmentation in PANC-1 cells.

Mitochondrial fragmentation induces oxidative stress via mitochondrial ROS (mROS)

Additional experiments were performed to explore the downstream events of mitochondrial fragmentation. Based on a previous study, mitochondrial fragmentation was associated with cellular oxidative stress via mROS overloading [41]. To confirm this, a mROS probe and flow cytometry were used to quantify mROS levels after erlotinib treatment. As shown in Fig. 3a, b, the level of mROS was significantly elevated in response to erlotinib treatment. To validate whether mitochondrial fragmentation was required for mROS overloading, Mdivi-1 and FCCP were used. FCCP treatment elevated the ROS production in control group, similar to the results obtained vis supplementation of erlotinib, However, Mdivi-1 application attenuated erlotinib-mediated mROS overloading (Fig. 3a, b), indicating the necessary role that is played by mitochondrial fragmentation in mROS generation. Excessive mROS production would induce cellular oxidative injury. To confirm this, an ELISA assay was used to observe alterations in the levels of cellular antioxidants. Compared to the control group, the concentration of Mn-SOD, GSH and GPX were markedly reduced after erlotinib treatment (Fig. 3c–e). In contrast, the level of MDA, an end product of the peroxidation of lipids in the cell membrane, was increased in response to erlotinib treatment (Fig. 3f). Interestingly, blockade of mitochondrial fragmentation via Mdivi-1 could decrease the level of antioxidants and suppress the production of MDA (Fig. 3c–f). Excessive oxidative injury can also disrupt cellular energy metabolism. Accordingly, total ATP production was measured using ELISA. Compared to the control group, erlotinib treatment significantly reduced the ATP production in PANC-1 cells (Fig. 3g), and this effect could be reversed by Mdivi-1. Furthermore, we also found that the expression of proteins related to mitochondrial ATP synthesis were notably downregulated in response to erlotinib (Fig. 3h–k); this effect was abrogated by Mdivi-1. Accordingly, our data indicated that mitochondrial fragmentation evoked mitochondrial ROS overloading and oxidative stress in PANC-1 cells.

Mitochondrial fragmentation-mediated mROS promotes HtrA2/Omi liberation

Next, experiments were performed to observe the consequence of mROS-mediated cell oxidative stress. Based on a previous report [42], excessive mROS could cause mitochondrial membrane permeabilization, which facilitates the translocation of mitochondrial proapoptotic factors to the nucleus/cytoplasm [43]. In the present study, an immunofluorescence analysis demonstrated that erlotinib increased the migration of HtrA2/Omi to nucleus when compared to the control group (Fig. 4a, b). Interestingly, this effect of erlotinib could be abolished via Mdivi-1 (Fig. 4a, b). Subsequently, western blotting was performed to quantify HtrA2/Omi liberation. As shown in Fig. 4c–e, compared to the control group, erlotinib treatment increaased the levels of cytoplasmic HtrA2/Omi (cyto-Htra2/Omi) and reduced the expression of mitochondrial HtrA2/Omi (mito-HtrA2/Omi). Similar results were also observed in cytochrome c (cyt c) liberation from mitochondria into cytoplasm (Fig. 4c–f). However, Mdivi-1 treatment repressed the erlotinib-mediated HtrA2/Omi and cyt c translocation from mitochondria into the cytoplasm. These results indicated that mitochondrial fragmentation accounted for HtrA2/Omi liberation.
At the molecular level, HtrA2/Omi is primarily expressed in the inner membrane of mitochondria. Based on a recent study, the liberation of HtrA2/Omi from mitochondria into the cytoplasm is dependent on cardiolipin oxidation and mPTP opening [15, 44]. First, the oxidation of cardiolipin lowers the affinity of HtrA2/Omi to the mitochondria. Second, the opening of mPTP provides a channel for HtrA2/Omi leakage [45]. Given the role of mitochondrial fragmentation in cellular oxidative stress via mROS overproduction, we asked whether mROS was required for the mitochondrial fragmentation-mediated HtrA2/Omi liberation via modulating cardiolipin oxidation and mPTP opening. To support our hypothesis, cardiolipin oxidation was determined via staining with NAO, which is a cardiolipin probe. Under physiological conditions, NAO could interact with cardiolipin to display a green fluorescence. In response to cardiolipin oxidation, NAO cannot bind to oxidized cardiolipin, and thus the green fluorescence is reduced. As shown in Fig. 4g, h, the fluorescence of cardiolipin was significantly downregulated in response to erlotinib, and this effect was reversed by Mdivi-1. To verify whether mROS was responsible for cardiolipin oxidation, mitoQ was used to neutralize the mitochondrial fragmentation-produced mROS. Interestingly, mitoQ treatment also reversed the green fluorescence intensity of cardiolipin (Fig. 4g, h), similar to the results obtained via supplementation with Mdivi-1. These results verified the role played by mROS in cardiolipin oxidation. In addition, we also found that the mPTP opening rate was significantly increased in response to erlotinib (Fig. 4i), and this effect was inhibited by Mdivi-1 or mitoQ (Fig. 4i). Together, our data demonstrated that the mitochondrial fragmentation-mediated mROS regulated HtrA2/Omi liberation via inducing cardiolipin oxidation and mPTP opening.

Released HtrA2/Omi induces caspase-9-dependent apoptosis

After it is released into the cytoplasm, HtrA2/Omi can interact with and activate mitochondrial apoptosis in a manner that is dependent on caspase-9 activity [11]. Notably, an early feature of caspase-9-related apoptosis is the reduction of mitochondrial potential. In the present study, a JC-1 kit was used to stain for the mitochondrial potential. The results indicated that erlotinib treatment significantly reduced the mitochondrial potential (Fig. 5a, b), and this effect was inhibited by Mdivi-1. To confirm whether HtrA2/Omi accounted for the mitochondrial potential collapse, two independent siRNAs were used. After knockdown of HtrA2/Omi, the mitochondrial potential was analyzed again. Compared to the erlotinib-treated group, the loss of HtrA2/Omi stabilized the mitochondrial potential (Fig. 5a, b), an effect that was similar to the results obtained via treatment with Mdivi-1. Furthermore, the last characteristic of caspase-9-related apoptosis is the activation of caspase-9, an effect that is accompanied by an increase in proapoptotic proteins. In the present study, the protein activity (Fig. 5c) and expression (Fig. 5d, e) of caspase-9 were both upregulated in answer to erlotinib stress and these effects could be repressed by Mdivi-1 or HtrA2/Omi siRNA. As a consequence of caspase-9 activation, the levels of proapoptotic factors such as Bad and Bax were significantly increased in response to erlotinib treatment, and this effect was negated by Mdivi-1 treatment or HtrA2/Omi siRNA transfection (Fig. 5f–j). By comparison, the expression of antiapoptotic proteins, including Bcl-2 and survivin, were obviously downregulated by erlotinib (Fig. 5f–j) and were reversed to near-normal levels with Mdivi-1 treatment or HtrA2/Omi knockdown. Together, our results indicated that mitochondrial fragmentation activated caspase-9-dependent apoptosis via HtrA2/Omi.

Mitochondrial fragmentation also modulated PANC-1 cell proliferation via mROS-HtrA2/Omi pathways

To this end, we asked whether mitochondrial fragmentation was involved in PANC-1 cell proliferation via the mROS-HtrA2/Omi pathways. First, the EdU assay was conducted to observe cellular proliferation. As shown in Fig. 6a, b, compared to the control group, erlotinib treatment significantly reduced the ratio of EdU-positive cells; this effect was repressed by Mdivi-1 (Fig. 6a, b). In addition, the neutralization of mROS via mitoQ and knockdown of HtrA2/Omi via siRNA transfection also reversed the number of EdU-positive cells after erlotinib treatment (Fig. 6a, b). These results indicated that mitochondrial fragmentation affected the cell proliferation in PANC-1 cell via the mROS-HtrA2/Omi axis. Further, the cell proliferation is primarily regulated by CDK4 and Cyclin D1. Cyclin D1 and cyclin E interact with each other and generate cyclin-dependent kinase (Cdk)4/6-cyclin D and/or Cdk2-cyclin E complexes, which accelerate transition from the G0/G1 to S stage, according to the previous study [46]. We have provided the references for this. With the help of a western blotting assay, we found that the expression of CDK4 and Cyclin D1 were both reduced in response to erlotinib treatment, and this effect was negated by Mdivi-1 (Fig. 6c–e). Interestingly, the neutralization of mROS via mitoQ and knockdown of HtrA2/Omi via siRNA transfection also reversed the levels of CDK4 and Cyclin D1. Together, our results confirmed that PANC-1 cell proliferation was modulated by erlotinib via mitochondrial fragmentation in a manner that was dependent on the mROS-HtrA2/Omi pathways.

Discussion

According to the previous findings, mitochondrial fission has been acknowledged as a potential target to reduce the proliferation, migration and survival of PANC-1 pancreatic cancer cells [10]. Excessive mitochondrial fission promotes mitochondrial fragmentation [15]. Fragmented mitochondria induce damage to mitochondrial structure and function, eventually interrupting the cellular ATP supply and activating the apoptosis response [47, 48]. However, the detailed molecular mechanism by which mitochondrial fragmentation triggers mitochondrial damage and cellular apoptosis remains unclear. Our study provides an answer to this question. We used different doses of ERL to screen its proapoptotic effect in two types of cancer cell lines. Then, we used the minimal lethal dose of ERL to investigate its apoptotic mechanism, with a focus on mitochondrial damage. We observed the minimal lethal dose of ERL has an ability to induce the mitochondrial fragmentation and this finding may explain one of the mechanisms by which ERL mediated cancer cell apoptosis. Notably, whether higher dose of ERL could activate other signaling pathway to induce cell apoptosis requires further investigation. Our data illustrated that erlotinib treatment promoted mitochondrial fragmentation that occurred via increased mitochondrial fission and decreased mitochondrial fusion. Subsequently, excessive mitochondrial fragmentation triggered mROS overloading, leading to cellular oxidative stress and disordered energy metabolism. In addition, mROS overproduction was closely associated with cardiolipin oxidation and mPTP opening, favoring HtrA2/Omi liberation from mitochondria into the cytoplasm. As a consequence of HtrA2/Omi leakage, reduction of the mitochondrial potential and caspase-9 activation were noted, and these alterations were accompanied by an upregulation of proapoptotic proteins and a downregulation of antiapoptotic factors. Overall, we demonstrated for the first time that erlotinib-activated mitochondrial fragmentation mediated PANC-1 apoptosis via the mROS-HtrA2/Omi pathways. This finding fills the knowledge gap regarding how mitochondrial fragmentation induces mitochondrial damage and triggers the apoptotic pathway.
Mitochondrial fission and fusion are a part of mitochondrial dynamics. Under physiological conditions, the mitochondrial network undergoes moderate fission and fusion to fill the requirements for cellular metabolism [49, 50]. Mild levels of mitochondrial fission help the mitochondria in generating daughter mitochondria, whereas moderate levels of mitochondrial fusion provides the energy for communication between the mitochondrial network [51, 52]. Interestingly, uncontrolled mitochondrial fission generates massive amounts of fragmented mitochondria and disrupts mitochondrial homeostasis. Previous studies have identified mitochondrial fragmentation, which is produced by mitochondrial fission, as the apoptotic trigger in various disease models. For instance, in fatty liver disease, mitochondrial fragmentation promotes the apoptosis of hepatocytes and the progression of liver fibrosis by decreasing mitophagy [53]. In neurodegenerative illness such as Alzheimer’s disease, excessive mitochondrial fragmentation disturbs mitochondrial energy metabolism and causes neuronal oxidative injury [54]. In addition, in rectal cancer, activated mitochondrial fragmentation limits tumor proliferation and augments cancer apoptosis [11]. In accordance with these findings, our data also illustrated the necessary role played by mitochondrial fragmentation in initiating pancreatic cancer PANC-1 cell death. Thus, mitochondrial fragmentation would be considered as a tumor-suppressor, and strategies to promote mitochondrial fragmentation are of significant importance in the design of anti-cancer drugs.
Although the proapoptotic effect of mitochondrial fragmentation has been well-documented, the detailed mechanisms by which mitochondrial fragmentation induces mitochondrial damage and activates cellular apoptosis are incompletely understood. In the present study, we found that mitochondrial fragmentation modulated mitochondrial homeostasis and cell viability through two mechanisms. One mechanism was driven by the promotion of mROS-mediated cell oxidative injury, and the other involved the HtrA2/Omi liberation-induced caspase-9 activation. First, mitochondrial fragmentation generated superfluous amounts of mROS, and the excess mROS induced cardiolipin oxidation and mPTP opening [55]. Subsequently, oxidized cardiolipin and increased mPTP opening worked together to augment the liberation of HtrA2/Omi from mitochondria into the cytoplasm, where Htra2/Omi reduced the mitochondrial potential and induced caspase-9 activation. This information was also consistent with previous studies. In cardiac ischemia–reperfusion injury, excessive mitochondrial fragmentation-induced mitochondrial DNA damage evokes mROS overproduction and cardiolipin oxidation [14, 15]. Additionally, in oral cancer, mitochondrial fragmentation-related cardiolipin oxidation and mPTP opening eventually contribute to caspase-involved cellular apoptosis [56].
In the present study, we used erlotinib to activate mitochondrial fragmentation and found that erlotinib-mediated PANC-1 cellular apoptosis could be inhibited by Mdivi-1, which is an antagonist of mitochondrial fragmentation. To the best of our knowledge, this is the first study to investigate the role of erlotinib in mitochondrial stress. Although erlotinib has been tested in several human clinical studies [57, 58], its pharmacological mechanism has not been adequately explored. Our study proposed that the anti-cancer property of erlotinib relied on the activation of mitochondrial fragmentation by upregulating mitochondrial fission and downregulating mitochondrial fusion. Notably, the dose selection of ERL was according to a previous study [26] and this selection may be also relied on the types of cancer cell lines. In clinical practice, different doses of ERL have been used according to the tumor staging and pathologic grading. Further insight is required to figure out the appropriate concentration of ERL on different types of pancreatic cancer. Besides, there are several limitations in the present study. Although we used two pancreatic cancer cell lines to screen the role of erlotinib, an animal study is necessary to further support our finding. In addition, human evidence is also required to validate the tumor-suppressive effects of mitochondrial fragmentation in response to erlotinib treatment.

Conclusion

Collectively, our results reported that mitochondrial fragmentation, which was activated by erlotinib, regulated the viability of the PANC-1 pancreatic cancer cell line via the mROS-HtrA2/Omi pathways. This conclusion provides a potential target to modify pancreatic cancer viability via augmenting mitochondrial fragmentation and activating the mROS-HtrA2/Omi pathways.

Authors’ contributions

JW, JC, and LW were involved in the conception and design, performance of experiments, data analysis and interpretation, and manuscript writing. KPW, XPH, YLZ, HK and SZ were involved in data analysis and interpretation. All authors read and approved the final manuscript.

Acknowledgements

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.
Not applicable.
Not applicable.

Funding

This study was supported by grants from the Hubei Province Health and Family Planning Scientific Research Project (WJ2017F085) and the Youth Project of China Three Gorges University (KJ2016A018).

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Literatur
1.
Zurück zum Zitat 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.PubMed 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.PubMed
2.
Zurück zum Zitat Rossi ML, Rehman AA, Gondi CS. Therapeutic options for the management of pancreatic cancer. World J Gastroenterol. 2014;20(32):11142–59.PubMedPubMedCentral Rossi ML, Rehman AA, Gondi CS. Therapeutic options for the management of pancreatic cancer. World J Gastroenterol. 2014;20(32):11142–59.PubMedPubMedCentral
3.
Zurück zum Zitat Zhu H, Jin Q, Li Y, Ma Q, Wang J, Li D, Zhou H, Chen Y. Melatonin protected cardiac microvascular endothelial cells against oxidative stress injury via suppression of IP3R-[Ca(2+)]c/VDAC-[Ca(2+)]m axis by activation of MAPK/ERK signaling pathway. Cell Stress Chaperones. 2018;23(1):101–13.PubMed Zhu H, Jin Q, Li Y, Ma Q, Wang J, Li D, Zhou H, Chen Y. Melatonin protected cardiac microvascular endothelial cells against oxidative stress injury via suppression of IP3R-[Ca(2+)]c/VDAC-[Ca(2+)]m axis by activation of MAPK/ERK signaling pathway. Cell Stress Chaperones. 2018;23(1):101–13.PubMed
4.
Zurück zum Zitat Shi C, Cai Y, Li Y, Li Y, Hu N, Ma S, Hu S, Zhu P, Wang W, Zhou H. Yap promotes hepatocellular carcinoma metastasis and mobilization via governing cofilin/F-actin/lamellipodium axis by regulation of JNK/Bnip3/SERCA/CaMKII pathways. Redox Biol. 2018;14:59–71.PubMed Shi C, Cai Y, Li Y, Li Y, Hu N, Ma S, Hu S, Zhu P, Wang W, Zhou H. Yap promotes hepatocellular carcinoma metastasis and mobilization via governing cofilin/F-actin/lamellipodium axis by regulation of JNK/Bnip3/SERCA/CaMKII pathways. Redox Biol. 2018;14:59–71.PubMed
5.
Zurück zum Zitat Viale A, Pettazzoni P, Lyssiotis CA, Ying H, Sanchez N, Marchesini M, Carugo A, Green T, Seth S, Giuliani V, et al. Oncogene ablation-resistant pancreatic cancer cells depend on mitochondrial function. Nature. 2014;514(7524):628–32.PubMedPubMedCentral Viale A, Pettazzoni P, Lyssiotis CA, Ying H, Sanchez N, Marchesini M, Carugo A, Green T, Seth S, Giuliani V, et al. Oncogene ablation-resistant pancreatic cancer cells depend on mitochondrial function. Nature. 2014;514(7524):628–32.PubMedPubMedCentral
6.
Zurück zum Zitat Li G, Gan Y, Fan Y, Wu Y, Lin H, Song Y, Cai X, Yu X, Pan W, Yao M, et al. Enriched environment inhibits mouse pancreatic cancer growth and down-regulates the expression of mitochondria-related genes in cancer cells. Sci Rep. 2015;5:7856.PubMedPubMedCentral Li G, Gan Y, Fan Y, Wu Y, Lin H, Song Y, Cai X, Yu X, Pan W, Yao M, et al. Enriched environment inhibits mouse pancreatic cancer growth and down-regulates the expression of mitochondria-related genes in cancer cells. Sci Rep. 2015;5:7856.PubMedPubMedCentral
7.
Zurück zum Zitat Zhou H, Li D, Zhu P, Hu S, Hu N, Ma S, Zhang Y, Han T, Ren J, Cao F, et al. Melatonin suppresses platelet activation and function against cardiac ischemia/reperfusion injury via PPARgamma/FUNDC1/mitophagy pathways. J Pineal Res. 2017;63(4):e12438. Zhou H, Li D, Zhu P, Hu S, Hu N, Ma S, Zhang Y, Han T, Ren J, Cao F, et al. Melatonin suppresses platelet activation and function against cardiac ischemia/reperfusion injury via PPARgamma/FUNDC1/mitophagy pathways. J Pineal Res. 2017;63(4):e12438.
8.
Zurück zum Zitat Zhu P, Hu S, Jin Q, Li D, Tian F, Toan S, Li Y, Zhou H, Chen Y. Ripk3 promotes ER stress-induced necroptosis in cardiac IR injury: A mechanism involving calcium overload/XO/ROS/mPTP pathway. Redox Biol. 2018;16:157–68.PubMedPubMedCentral Zhu P, Hu S, Jin Q, Li D, Tian F, Toan S, Li Y, Zhou H, Chen Y. Ripk3 promotes ER stress-induced necroptosis in cardiac IR injury: A mechanism involving calcium overload/XO/ROS/mPTP pathway. Redox Biol. 2018;16:157–68.PubMedPubMedCentral
9.
Zurück zum Zitat Xia J, Inagaki Y, Gao J, Qi F, Song P, Han G, Sawakami T, Gao B, Luo C, Kokudo N, et al. Combination of cinobufacini and doxorubicin increases apoptosis of hepatocellular carcinoma cells through the Fas- and mitochondria-mediated pathways. Am J Chin Med. 2017;45(7):1537–56.PubMed Xia J, Inagaki Y, Gao J, Qi F, Song P, Han G, Sawakami T, Gao B, Luo C, Kokudo N, et al. Combination of cinobufacini and doxorubicin increases apoptosis of hepatocellular carcinoma cells through the Fas- and mitochondria-mediated pathways. Am J Chin Med. 2017;45(7):1537–56.PubMed
10.
Zurück zum Zitat 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):124–36.PubMedPubMedCentral 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):124–36.PubMedPubMedCentral
11.
Zurück zum Zitat Li H, He F, Zhao X, Zhang Y, Chu X, Hua C, Qu Y, Duan Y, Ming L. 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):2073–89.PubMed Li H, He F, Zhao X, Zhang Y, Chu X, Hua C, Qu Y, Duan Y, Ming L. 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):2073–89.PubMed
12.
Zurück zum Zitat Yan H, Xiao F, Zou J, Qiu C, Sun W, Gu M, Zhang L. NR4A1-induced increase in the sensitivity of a human gastric cancer line to TNFalpha-mediated apoptosis is associated with the inhibition of JNK/Parkin-dependent mitophagy. Int J Oncol. 2018;52(2):367–78.PubMed Yan H, Xiao F, Zou J, Qiu C, Sun W, Gu M, Zhang L. NR4A1-induced increase in the sensitivity of a human gastric cancer line to TNFalpha-mediated apoptosis is associated with the inhibition of JNK/Parkin-dependent mitophagy. Int J Oncol. 2018;52(2):367–78.PubMed
13.
Zurück zum Zitat Zhang J, Zhang Y, Wu W, Wang F, Liu X, Shui G, Nie C. Guanylate-binding protein 2 regulates Drp1-mediated mitochondrial fission to suppress breast cancer cell invasion. Cell Death Dis. 2017;8(10):e3151.PubMedPubMedCentral Zhang J, Zhang Y, Wu W, Wang F, Liu X, Shui G, Nie C. Guanylate-binding protein 2 regulates Drp1-mediated mitochondrial fission to suppress breast cancer cell invasion. Cell Death Dis. 2017;8(10):e3151.PubMedPubMedCentral
14.
Zurück zum Zitat Zhou H, Wang J, Zhu P, Zhu H, Toan S, Hu S, Ren J, Chen Y. NR4A1 aggravates the cardiac microvascular ischemia reperfusion injury through suppressing FUNDC1-mediated mitophagy and promoting Mff-required mitochondrial fission by CK2alpha. Basic Res Cardiol. 2018;113(4):23.PubMed Zhou H, Wang J, Zhu P, Zhu H, Toan S, Hu S, Ren J, Chen Y. NR4A1 aggravates the cardiac microvascular ischemia reperfusion injury through suppressing FUNDC1-mediated mitophagy and promoting Mff-required mitochondrial fission by CK2alpha. Basic Res Cardiol. 2018;113(4):23.PubMed
16.
Zurück zum Zitat Zhou H, Shi C, Hu S, Zhu H, Ren J, Chen Y. BI1 is associated with microvascular protection in cardiac ischemia reperfusion injury via repressing Syk-Nox2-Drp1-mitochondrial fission pathways. Angiogenesis. 2018;21(3):599–615.PubMed Zhou H, Shi C, Hu S, Zhu H, Ren J, Chen Y. BI1 is associated with microvascular protection in cardiac ischemia reperfusion injury via repressing Syk-Nox2-Drp1-mitochondrial fission pathways. Angiogenesis. 2018;21(3):599–615.PubMed
17.
Zurück zum Zitat Yuan X, Zhou Y, Wang W, Li J, Xie G, Zhao Y, Xu D, Shen L. Activation of TLR4 signaling promotes gastric cancer progression by inducing mitochondrial ROS production. Cell Death Dis. 2013;4:e794.PubMedPubMedCentral Yuan X, Zhou Y, Wang W, Li J, Xie G, Zhao Y, Xu D, Shen L. Activation of TLR4 signaling promotes gastric cancer progression by inducing mitochondrial ROS production. Cell Death Dis. 2013;4:e794.PubMedPubMedCentral
18.
Zurück zum Zitat Hong H, Tao T, Chen S, Liang C, Qiu Y, Zhou Y, Zhang R. MicroRNA-143 promotes cardiac ischemia-mediated mitochondrial impairment by the inhibition of protein kinase Cepsilon. Basic Res Cardiol. 2017;112(6):60.PubMed Hong H, Tao T, Chen S, Liang C, Qiu Y, Zhou Y, Zhang R. MicroRNA-143 promotes cardiac ischemia-mediated mitochondrial impairment by the inhibition of protein kinase Cepsilon. Basic Res Cardiol. 2017;112(6):60.PubMed
19.
Zurück zum Zitat Dall’Acqua S, Linardi MA, Bortolozzi R, Clauser M, Marzocchini S, Maggi F, Nicoletti M, Innocenti G, Basso G, Viola G. Natural daucane esters induces apoptosis in leukaemic cells through ROS production. Phytochemistry. 2014;108:147–56.PubMed Dall’Acqua S, Linardi MA, Bortolozzi R, Clauser M, Marzocchini S, Maggi F, Nicoletti M, Innocenti G, Basso G, Viola G. Natural daucane esters induces apoptosis in leukaemic cells through ROS production. Phytochemistry. 2014;108:147–56.PubMed
20.
Zurück zum Zitat Salminen A, Kaarniranta K, Kauppinen A. Integrated stress response stimulates FGF21 expression: Systemic enhancer of longevity. Cell Signal. 2017;40:10–21.PubMed Salminen A, Kaarniranta K, Kauppinen A. Integrated stress response stimulates FGF21 expression: Systemic enhancer of longevity. Cell Signal. 2017;40:10–21.PubMed
21.
Zurück zum Zitat Zhou H, Zhang Y, Hu S, Shi C, Zhu P, Ma Q, Jin Q, Cao F, Tian F, Chen Y. Melatonin protects cardiac microvasculature against ischemia/reperfusion injury via suppression of mitochondrial fission-VDAC1-HK2-mPTP-mitophagy axis. J Pineal Res. 2017;63(1):e12413.PubMedCentral Zhou H, Zhang Y, Hu S, Shi C, Zhu P, Ma Q, Jin Q, Cao F, Tian F, Chen Y. Melatonin protects cardiac microvasculature against ischemia/reperfusion injury via suppression of mitochondrial fission-VDAC1-HK2-mPTP-mitophagy axis. J Pineal Res. 2017;63(1):e12413.PubMedCentral
22.
Zurück zum Zitat Xie CG, Sun SL, Wei SM, Xu XM, Shao LM, Chen JM, Cai JT. Downregulation of GEP100 improved the growth inhibition effect of erlotinib through modulating mesenchymal epithelial transition process in pancreatic cancer. Pancreas. 2018;47(6):732–7.PubMed Xie CG, Sun SL, Wei SM, Xu XM, Shao LM, Chen JM, Cai JT. Downregulation of GEP100 improved the growth inhibition effect of erlotinib through modulating mesenchymal epithelial transition process in pancreatic cancer. Pancreas. 2018;47(6):732–7.PubMed
23.
Zurück zum Zitat Nunez-Gomez E, Pericacho M, Ollauri-Ibanez C, Bernabeu C, Lopez-Novoa JM. The role of endoglin in post-ischemic revascularization. Angiogenesis. 2017;20(1):1–24.PubMed Nunez-Gomez E, Pericacho M, Ollauri-Ibanez C, Bernabeu C, Lopez-Novoa JM. The role of endoglin in post-ischemic revascularization. Angiogenesis. 2017;20(1):1–24.PubMed
24.
Zurück zum Zitat Kadera BE, Toste PA, Wu N, Li L, Nguyen AH, Dawson DW, Donahue TR. Low expression of the E3 ubiquitin ligase CBL confers chemoresistance in human pancreatic cancer and is targeted by epidermal growth factor receptor inhibition. Clin Cancer Res. 2015;21(1):157–65.PubMed Kadera BE, Toste PA, Wu N, Li L, Nguyen AH, Dawson DW, Donahue TR. Low expression of the E3 ubiquitin ligase CBL confers chemoresistance in human pancreatic cancer and is targeted by epidermal growth factor receptor inhibition. Clin Cancer Res. 2015;21(1):157–65.PubMed
25.
Zurück zum Zitat Zhou H, Ma Q, Zhu P, Ren J, Reiter RJ, Chen Y. Protective role of melatonin in cardiac ischemia-reperfusion injury: From pathogenesis to targeted therapy. J Pineal Res. 2018;64(3):e12471. Zhou H, Ma Q, Zhu P, Ren J, Reiter RJ, Chen Y. Protective role of melatonin in cardiac ischemia-reperfusion injury: From pathogenesis to targeted therapy. J Pineal Res. 2018;64(3):e12471.
26.
Zurück zum Zitat Shan F, Shao Z, Jiang S, Cheng Z. Erlotinib induces the human non-small-cell lung cancer cells apoptosis via activating ROS-dependent JNK pathways. Cancer Med. 2016;5(11):3166–75.PubMedPubMedCentral Shan F, Shao Z, Jiang S, Cheng Z. Erlotinib induces the human non-small-cell lung cancer cells apoptosis via activating ROS-dependent JNK pathways. Cancer Med. 2016;5(11):3166–75.PubMedPubMedCentral
27.
Zurück zum Zitat Couto JA, Ayturk UM, Konczyk DJ, Goss JA, Huang AY, Hann S, Reeve JL, Liang MG, Bischoff J, Warman ML, et al. A somatic GNA11 mutation is associated with extremity capillary malformation and overgrowth. Angiogenesis. 2017;20(3):303–6.PubMedPubMedCentral Couto JA, Ayturk UM, Konczyk DJ, Goss JA, Huang AY, Hann S, Reeve JL, Liang MG, Bischoff J, Warman ML, et al. A somatic GNA11 mutation is associated with extremity capillary malformation and overgrowth. Angiogenesis. 2017;20(3):303–6.PubMedPubMedCentral
28.
Zurück zum Zitat Zhou H, Li D, Zhu P, Ma Q, Toan S, Wang J, Hu S, Chen Y, Zhang Y. Inhibitory effect of melatonin on necroptosis via repressing the Ripk3-PGAM5-CypD-mPTP pathway attenuates cardiac microvascular ischemia-reperfusion injury. J Pineal Res. 2018;65:e12503.PubMed Zhou H, Li D, Zhu P, Ma Q, Toan S, Wang J, Hu S, Chen Y, Zhang Y. Inhibitory effect of melatonin on necroptosis via repressing the Ripk3-PGAM5-CypD-mPTP pathway attenuates cardiac microvascular ischemia-reperfusion injury. J Pineal Res. 2018;65:e12503.PubMed
29.
Zurück zum Zitat Kingery JR, Hamid T, Lewis RK, Ismahil MA, Bansal SS, Rokosh G, Townes TM, Ildstad ST, Jones SP, Prabhu SD. Leukocyte iNOS is required for inflammation and pathological remodeling in ischemic heart failure. Basic Res Cardiol. 2017;112(2):19.PubMedPubMedCentral Kingery JR, Hamid T, Lewis RK, Ismahil MA, Bansal SS, Rokosh G, Townes TM, Ildstad ST, Jones SP, Prabhu SD. Leukocyte iNOS is required for inflammation and pathological remodeling in ischemic heart failure. Basic Res Cardiol. 2017;112(2):19.PubMedPubMedCentral
30.
Zurück zum Zitat Feng D, Wang B, Wang L, Abraham N, Tao K, Huang L, Shi W, Dong Y, Qu Y. Pre-ischemia melatonin treatment alleviated acute neuronal injury after ischemic stroke by inhibiting endoplasmic reticulum stress-dependent autophagy via PERK and IRE1 signalings. J Pineal Res. 2017. https://doi.org/10.1111/jpi.12395.CrossRefPubMed Feng D, Wang B, Wang L, Abraham N, Tao K, Huang L, Shi W, Dong Y, Qu Y. Pre-ischemia melatonin treatment alleviated acute neuronal injury after ischemic stroke by inhibiting endoplasmic reticulum stress-dependent autophagy via PERK and IRE1 signalings. J Pineal Res. 2017. https://​doi.​org/​10.​1111/​jpi.​12395.CrossRefPubMed
31.
Zurück zum Zitat Brasacchio D, Alsop AE, Noori T, Lufti M, Iyer S, Simpson KJ, Bird PI, Kluck RM, Johnstone RW, Trapani JA. Epigenetic control of mitochondrial cell death through PACS1-mediated regulation of BAX/BAK oligomerization. Cell Death Differ. 2017;24(6):961–70.PubMedPubMedCentral Brasacchio D, Alsop AE, Noori T, Lufti M, Iyer S, Simpson KJ, Bird PI, Kluck RM, Johnstone RW, Trapani JA. Epigenetic control of mitochondrial cell death through PACS1-mediated regulation of BAX/BAK oligomerization. Cell Death Differ. 2017;24(6):961–70.PubMedPubMedCentral
32.
Zurück zum Zitat 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;18:229–43.PubMedPubMedCentral 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;18:229–43.PubMedPubMedCentral
33.
Zurück zum Zitat Ligeza J, Marona P, Gach N, Lipert B, Miekus K, Wilk W, Jaszczynski J, Stelmach A, Loboda A, Dulak J, et al. MCPIP1 contributes to clear cell renal cell carcinomas development. Angiogenesis. 2017;20(3):325–40.PubMedPubMedCentral Ligeza J, Marona P, Gach N, Lipert B, Miekus K, Wilk W, Jaszczynski J, Stelmach A, Loboda A, Dulak J, et al. MCPIP1 contributes to clear cell renal cell carcinomas development. Angiogenesis. 2017;20(3):325–40.PubMedPubMedCentral
34.
Zurück zum Zitat Yang X, Xu Y, Wang T, Shu D, Guo P, Miskimins K, Qian SY. Inhibition of cancer migration and invasion by knocking down delta-5-desaturase in COX-2 overexpressed cancer cells. Redox Biol. 2017;11:653–62.PubMedPubMedCentral Yang X, Xu Y, Wang T, Shu D, Guo P, Miskimins K, Qian SY. Inhibition of cancer migration and invasion by knocking down delta-5-desaturase in COX-2 overexpressed cancer cells. Redox Biol. 2017;11:653–62.PubMedPubMedCentral
36.
Zurück zum Zitat Zhou H, Zhu P, Wang J, Zhu H, Ren J, Chen Y. Pathogenesis of cardiac ischemia reperfusion injury is associated with CK2alpha-disturbed mitochondrial homeostasis via suppression of FUNDC1-related mitophagy. Cell Death Differ. 2018;25(6):1080–93.PubMedPubMedCentral Zhou H, Zhu P, Wang J, Zhu H, Ren J, Chen Y. Pathogenesis of cardiac ischemia reperfusion injury is associated with CK2alpha-disturbed mitochondrial homeostasis via suppression of FUNDC1-related mitophagy. Cell Death Differ. 2018;25(6):1080–93.PubMedPubMedCentral
37.
Zurück zum Zitat Zhou H, Wang J, Zhu P, Hu S, Ren J. Ripk3 regulates cardiac microvascular 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.PubMed Zhou H, Wang J, Zhu P, Hu S, Ren J. Ripk3 regulates cardiac microvascular 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.PubMed
38.
Zurück zum Zitat Sarkar C, Ganju RK, Pompili VJ, Chakroborty D. Enhanced peripheral dopamine impairs post-ischemic healing by suppressing angiotensin receptor type 1 expression in endothelial cells and inhibiting angiogenesis. Angiogenesis. 2017;20(1):97–107.PubMed Sarkar C, Ganju RK, Pompili VJ, Chakroborty D. Enhanced peripheral dopamine impairs post-ischemic healing by suppressing angiotensin receptor type 1 expression in endothelial cells and inhibiting angiogenesis. Angiogenesis. 2017;20(1):97–107.PubMed
40.
Zurück zum Zitat Garcia-Nino WR, Correa F, Rodriguez-Barrena JI, Leon-Contreras JC, Buelna-Chontal M, Soria-Castro E, Hernandez-Pando R, Pedraza-Chaverri J, Zazueta C. Cardioprotective kinase signaling to subsarcolemmal and interfibrillar mitochondria is mediated by caveolar structures. Basic Res Cardiol. 2017;112(2):15.PubMed Garcia-Nino WR, Correa F, Rodriguez-Barrena JI, Leon-Contreras JC, Buelna-Chontal M, Soria-Castro E, Hernandez-Pando R, Pedraza-Chaverri J, Zazueta C. Cardioprotective kinase signaling to subsarcolemmal and interfibrillar mitochondria is mediated by caveolar structures. Basic Res Cardiol. 2017;112(2):15.PubMed
41.
Zurück zum Zitat Gadicherla AK, Wang N, Bulic M, Agullo-Pascual E, Lissoni A, De Smet M, Delmar M, Bultynck G, Krysko DV, Camara A, et al. Mitochondrial Cx43 hemichannels contribute to mitochondrial calcium entry and cell death in the heart. Basic Res Cardiol. 2017;112(3):27.PubMed Gadicherla AK, Wang N, Bulic M, Agullo-Pascual E, Lissoni A, De Smet M, Delmar M, Bultynck G, Krysko DV, Camara A, et al. Mitochondrial Cx43 hemichannels contribute to mitochondrial calcium entry and cell death in the heart. Basic Res Cardiol. 2017;112(3):27.PubMed
42.
Zurück zum Zitat Rossello X, Riquelme JA, He Z, Taferner S, Vanhaesebroeck B, Davidson SM, Yellon DM. The role of PI3 Kalpha isoform in cardioprotection. Basic Res Cardiol. 2017;112(6):66.PubMedPubMedCentral Rossello X, Riquelme JA, He Z, Taferner S, Vanhaesebroeck B, Davidson SM, Yellon DM. The role of PI3 Kalpha isoform in cardioprotection. Basic Res Cardiol. 2017;112(6):66.PubMedPubMedCentral
44.
46.
Zurück zum Zitat Zhou H, Li D, Shi C, Xin T, Yang J, Zhou Y, Hu S, Tian F, Wang J, Chen Y. Effects of Exendin-4 on bone marrow mesenchymal stem cell proliferation, migration and apoptosis in vitro. Sci Rep. 2015;5:12898.PubMedPubMedCentral Zhou H, Li D, Shi C, Xin T, Yang J, Zhou Y, Hu S, Tian F, Wang J, Chen Y. Effects of Exendin-4 on bone marrow mesenchymal stem cell proliferation, migration and apoptosis in vitro. Sci Rep. 2015;5:12898.PubMedPubMedCentral
47.
Zurück zum Zitat Zhou H, Wang S, Zhu P, Hu S, Chen Y, Ren J. Empagliflozin rescues diabetic myocardial microvascular injury via AMPK-mediated inhibition of mitochondrial fission. Redox Biol. 2018;15:335–46.PubMed Zhou H, Wang S, Zhu P, Hu S, Chen Y, Ren J. Empagliflozin rescues diabetic myocardial microvascular injury via AMPK-mediated inhibition of mitochondrial fission. Redox Biol. 2018;15:335–46.PubMed
48.
Zurück zum Zitat Hu Z, Cheng J, Xu J, Ruf W, Lockwood CJ. Tissue factor is an angiogenic-specific receptor for factor VII-targeted immunotherapy and photodynamic therapy. Angiogenesis. 2017;20(1):85–96.PubMed Hu Z, Cheng J, Xu J, Ruf W, Lockwood CJ. Tissue factor is an angiogenic-specific receptor for factor VII-targeted immunotherapy and photodynamic therapy. Angiogenesis. 2017;20(1):85–96.PubMed
49.
Zurück zum Zitat 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.PubMedPubMedCentral 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.PubMedPubMedCentral
50.
Zurück zum Zitat Fuhrmann DC, Brune B. Mitochondrial composition and function under the control of hypoxia. Redox Biol. 2017;12:208–15.PubMedPubMedCentral Fuhrmann DC, Brune B. Mitochondrial composition and function under the control of hypoxia. Redox Biol. 2017;12:208–15.PubMedPubMedCentral
51.
Zurück zum Zitat Kozlov AV, Lancaster JR Jr, Meszaros AT, Weidinger A. Mitochondria-meditated pathways of organ failure upon inflammation. Redox Biol. 2017;13:170–81.PubMedPubMedCentral Kozlov AV, Lancaster JR Jr, Meszaros AT, Weidinger A. Mitochondria-meditated pathways of organ failure upon inflammation. Redox Biol. 2017;13:170–81.PubMedPubMedCentral
52.
Zurück zum Zitat Jin Q, Li R, Hu N, Xin T, Zhu P, Hu S, Ma S, Zhu H, Ren J, Zhou H. DUSP1 alleviates cardiac ischemia/reperfusion injury by suppressing the Mff-required mitochondrial fission and Bnip3-related mitophagy via the JNK pathways. Redox Biol. 2018;14:576–87.PubMed Jin Q, Li R, Hu N, Xin T, Zhu P, Hu S, Ma S, Zhu H, Ren J, Zhou H. DUSP1 alleviates cardiac ischemia/reperfusion injury by suppressing the Mff-required mitochondrial fission and Bnip3-related mitophagy via the JNK pathways. Redox Biol. 2018;14:576–87.PubMed
54.
Zurück zum Zitat Lei Q, Tan J, Yi S, Wu N, Wang Y, Wu H. Mitochonic acid 5 activates the MAPK-ERK-yap signaling pathways to protect mouse microglial BV-2 cells against TNFalpha-induced apoptosis via increased Bnip3-related mitophagy. Cell Mol Biol Lett. 2018;23:14.PubMedPubMedCentral Lei Q, Tan J, Yi S, Wu N, Wang Y, Wu H. Mitochonic acid 5 activates the MAPK-ERK-yap signaling pathways to protect mouse microglial BV-2 cells against TNFalpha-induced apoptosis via increased Bnip3-related mitophagy. Cell Mol Biol Lett. 2018;23:14.PubMedPubMedCentral
55.
Zurück zum Zitat Zhou H, Zhu P, Guo J, Hu N, Wang S, Li D, Hu S, Ren J, Cao F, Chen Y. Ripk3 induces mitochondrial apoptosis via inhibition of FUNDC1 mitophagy in cardiac IR injury. Redox Biol. 2017;13:498–507.PubMedPubMedCentral Zhou H, Zhu P, Guo J, Hu N, Wang S, Li D, Hu S, Ren J, Cao F, Chen Y. Ripk3 induces mitochondrial apoptosis via inhibition of FUNDC1 mitophagy in cardiac IR injury. Redox Biol. 2017;13:498–507.PubMedPubMedCentral
56.
Zurück zum Zitat Lee K, Back K. Overexpression of rice serotonin N-acetyltransferase 1 in transgenic rice plants confers resistance to cadmium and senescence and increases grain yield. J Pineal Res. 2017;62(3):e12392. Lee K, Back K. Overexpression of rice serotonin N-acetyltransferase 1 in transgenic rice plants confers resistance to cadmium and senescence and increases grain yield. J Pineal Res. 2017;62(3):e12392.
57.
Zurück zum Zitat Abravan A, Eide HA, Knudtsen IS, Londalen AM, Helland A, Malinen E. Assessment of pulmonary (18)F-FDG-PET uptake and cytokine profiles in non-small cell lung cancer patients treated with radiotherapy and erlotinib. Clin Transl Radiat Oncol. 2017;4:57–63.PubMedPubMedCentral Abravan A, Eide HA, Knudtsen IS, Londalen AM, Helland A, Malinen E. Assessment of pulmonary (18)F-FDG-PET uptake and cytokine profiles in non-small cell lung cancer patients treated with radiotherapy and erlotinib. Clin Transl Radiat Oncol. 2017;4:57–63.PubMedPubMedCentral
58.
Zurück zum Zitat Liu D, Zeng X, Li X, Mehta JL, Wang X. Role of NLRP3 inflammasome in the pathogenesis of cardiovascular diseases. Basic Res Cardiol. 2017;113(1):5.PubMed Liu D, Zeng X, Li X, Mehta JL, Wang X. Role of NLRP3 inflammasome in the pathogenesis of cardiovascular diseases. Basic Res Cardiol. 2017;113(1):5.PubMed
Metadaten
Titel
RETRACTED ARTICLE: Excessive mitochondrial fragmentation triggered by erlotinib promotes pancreatic cancer PANC-1 cell apoptosis via activating the mROS-HtrA2/Omi pathways
verfasst von
Jun Wan
Jie Cui
Lei Wang
Kunpeng Wu
Xiaoping Hong
Yulin Zou
Shuang Zhao
Hong Ke
Publikationsdatum
01.12.2018
Verlag
BioMed Central
Erschienen in
Cancer Cell International / Ausgabe 1/2018
Elektronische ISSN: 1475-2867
DOI
https://doi.org/10.1186/s12935-018-0665-1

Weitere Artikel der Ausgabe 1/2018

Cancer Cell International 1/2018 Zur Ausgabe

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.