Skip to main content
Erschienen in: Cancer Cell International 1/2019

Open Access 01.12.2019 | Primary research

RETRACTED ARTICLE: Mst1 overexpression combined with Yap knockdown augments thyroid carcinoma apoptosis via promoting MIEF1-related mitochondrial fission and activating the JNK pathway

verfasst von: Xiaoli Zhang, Fei Li, Yeqing Cui, Shuang Liu, Haichen Sun

Erschienen in: Cancer Cell International | Ausgabe 1/2019

Abstract

Background

Cancer cell viability is strongly modulated by the Hippo pathway, which includes mammalian STE20-like protein kinase 1 (Mst1) and yes-associated protein (Yap). Although the roles of Mst1 and Yap in thyroid carcinoma cell death have been fully addressed, no study has determined whether differential modification of Mst1 and Yap could further suppress thyroid carcinoma progression. The aim of our study was to explore the antiapoptotic effects exerted by combined Mst1 overexpression and Yap knockdown in thyroid carcinoma MDA-T32 cells in vitro.

Methods

Mst1 adenovirus and Yap shRNA were transfected into MDA-T32 cells to overexpress Mst1 and inhibit Yap, respectively. Cell viability and death were determined via an MTT assay, a TUNEL assay and western blotting. Mitochondrial function, mitochondrial fission and pathway studies were performed via western blotting and immunofluorescence.

Results

The results of our study showed that combined Mst1 overexpression and Yap knockdown further augmented MDA-T32 cell death by mediating mitochondrial damage. In addition, cancer cell migration and proliferation were suppressed by combined Mst1 overexpression and Yap knockdown. At the molecular level, mitochondrial membrane potential, ATP production, respiratory function, and caspase-9-related apoptosis were activated by combined Mst1 overexpression and Yap knockdown. Further, we found that fatal mitochondrial fission was augmented by combined Mst1 overexpression and Yap knockdown in a manner dependent on the JNK-MIEF1 pathway. Inhibition of JNK-MIEF1 pathway activity abolished the proapoptotic effects exerted by Mst1/Yap on MDA-T32 cells.

Conclusions

Taken together, our data suggest that Mst1 activation and Yap inhibition coordinate to augment thyroid cancer cell death by controlling the JNK-MIEF1-mitochondria pathway, suggesting that differential regulation of the core Hippo pathway components is potentially a novel therapeutic tool for the treatment of thyroid cancer.
Hinweise

Electronic supplementary material

The online version of this article (https://​doi.​org/​10.​1186/​s12935-019-0860-8) contains supplementary material, which is available to authorized users.
A Retraction Note to this article is available online at https://​doi.​org/​10.​1186/​s12935-023-02921-x.
This article has been retracted. Please see the retraction notice for more detail:https://​doi.​org/​10.​1186/​s12935-023-02921-x

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
Mst1
mammalian Ste20-like kinase 1
YAP
yes-associated protein
mROS
mitochondrial reactive oxygen species
MIEF1
mitochondrial elongation factor 1
mPTP
mitochondrial permeability transition pore

Background

The incidence of thyroid carcinoma, the most common endocrine malignancy, has significantly increased over the past decades. More than 50,000 new cases of thyroid carcinoma are currently diagnosed annually in the United States. Several risk factors have been introduced to explain the development of thyroid cancer, including sex, age, genetics, radiation exposure, a low-iodine diet, and race. Although many advances have been made in the early diagnosis and treatment of thyroid carcinoma, the pathogenesis of thyroid carcinoma has not been fully addressed.
Recently, studies have found a close interaction between the Hippo pathway and cancer progression. The Hippo pathway was originally identified as a novel antitumor signaling pathway that modulates tissue growth. The core Hippo pathway components include mammalian STE20-like protein kinase 1, yes-associated protein (YAP), and large tumor suppressor 1 (LATS1). Interestingly, these three Hippo kinases have various functions on cancer fate. For example, Mst1 has been found to promote cell death in gastric cancer, colorectal cancer, lung cancer, pancreatic cancer, and breast cancer [15]. In contrast, Yap has emerged as a growth promoter in cancer by modulating tumor aggressive behaviors, chemotherapy resistance, cancer stem cell differentiation, and tumor epithelial–mesenchymal transition [68]. There is little evidence to explain the exact role of LATS1 in cancer progression. Notably, several reports have indicated the impacts of Yap [9] and Mst1 [10] in controlling the viability of thyroid cancer cells. Loss of Yap sensitizes thyroid cancer to chemotherapy [11], whereas Mst1 overexpression augments papillary thyroid carcinoma apoptosis [10]. Considering the different roles played by Mst1 and Yap in the cancer biological phenotype, we asked whether Mst1 overexpression in combination with Yap knockdown could further promote the death of thyroid cancer cells.
Mitochondria extensively control various critical pathophysiological processes involving cancer metabolism, growth, proliferation, movement, differentiation, survival and metastasis [1215]. As the major consumers of oxygen and glucose, mitochondria produce sufficient ATP, which is required for cancer behaviors [16, 17]. However, damaged mitochondria impair cancer metabolism and even initiate mitochondria-related apoptotic pathway activity [18, 19]. For example, damaged mitochondria produce excessive ROS, which induces oxidative stress to mediate cellular senescence [20]. Moreover, injured mitochondria cannot generate enough energy, which is associated with the inability of cancer cells to adhere and invade [21]. More seriously, poorly structured mitochondria release proapoptotic factors such as cyt-c and HtrA2/Omi to initiate caspase-mediated apoptotic signals [22, 23]. Accordingly, mitochondria play a main role in both the survival and death of cancer cells. Notably, mitochondrial elongation factor 1 (MIEF1) has been found to be a novel mitochondrial homeostasis mediator [24]. Increased MIEF1 expression impairs mitochondrial dynamics, leading to mitochondrial fragmentation, which has been acknowledged as an early event in mitochondrial apoptosis initiation. For example, in lung cancer, MIEF1-dependent activation of mitochondria promotes mitochondrial stress and augments mitochondrial apoptosis in A549 lung cancer cells [25]. In addition, reperfusion-mediated cardiomyocyte death and endothelial damage are also tightly controlled by MIEF1 in a manner dependent on mitochondrial fission [26]. However, there is no evidence to indicate the influence of MIEF1-related mitochondrial fission on thyroid cancer cell viability. Considering that the Yap/Hippo pathway has been reported to be an upstream mediator of MIEF1-related mitochondrial fission, we asked whether differential regulation of Mst1 and Yap could further activate MIEF1-related mitochondrial fission and thus promote the death of thyroid cancer cells.
At the molecular levels, mitochondrial fission is primarily modulated by JNK pathway. For example, in tongue cancer [27], mitochondrial fission is highly modulated by the JNK-Fis1 pathway. In non-small cell lung cancer, mitochondrial fission is signaled by Hippo pathway in a manner dependent on the activity of JNK [28]. Similar results were also noted in liver cancer in response to cytokine-based therapy [29, 30]. This finding is also validated in thyroid cancer that activation of JNK pathway enhances mitochondrial fission and promotes cancer cell death [31, 32]. Based on the above information, we wanted to know whether JNK pathway was involved in mitochondrial fission in Mst1/Yap-modified cell viability in MDA-T32 cell in vitro. Altogether, the goal of our study is to figure out the synergistic effects of Mst1 overexpression and Yap knockdown on thyroid cancer death via modulating MIEF1-related mitochondrial fission and the JNK pathway.

Materials and methods

Cell culture and transfection

Human thyroid carcinoma MDA-T32 (ATCC® CRL-3351™) and MDA-T68 (ATCC® CRL-3353™) cell lines was purchased from American Type Culture Collection (ATCC) (Manassas, VA, USA). To overexpress the Mst1, adenovirus-Mst1 (ad-Mst1) was transfected into MDA-T32 cells. In brief, pDC315-Mst1 vector was obtained from Shanghai Gene-Pharma Co. (Shanghai, China) and then HEK293 cells (ATCC® CRL-3216™, American Type Culture Collection, Manassas, VA, USA) were infected with pDC315-Mst1 vector to obtain the Ad-Mst1 according to a previous study [33]. In brief, a total of 1 × 105 cells/well were infected with 50 multiplicity of infection (MOI) adenovirus (Ad)-Mst1 in serum-free RPMI-1640 for 12 h at 37 °C. To silence the expression of Yap, shRNA against Yap (sh-Yap) was used to transfect into MDA-T32 cells. The sh-Yap was purchased from Shanghai Gene-Pharma Co. (Shanghai, China) and transfection was performed with the help of Opti-MEM medium and Lipofectamine 2000 (Thermo Fisher Scientific, Inc.) according to the manufacturer’s protocol [34]. GFP-labelled adenovirus was used as a preliminary study to observe the infection efficiency (Additional file 1: Figure S1A). To prevent the activation of JNK pathway, SP600125 (25 μM, Selleck Chemicals, Houston, TX, USA) was added into the medium of MDA-T32 cells [35].

Cell proliferation assay and MTT assay

Cellular proliferation was evaluated via EdU assay. Cells were seeded onto a 6-well plate, and the Cell-Light™ EdU Apollo®567 In Vitro Imaging Kit (Thermo Fisher Scientific Inc., Waltham, MA, USA; Catalogue No. A10044) was used to observe the EdU-positive cells according to the manufacturer’s instructions [36]. MTT assay was used to observe the cellular viability. Cells were seeded onto a 96-well plate, and the MTT was then added to the medium (2 mg/mL; Sigma-Aldrich) [37]. Subsequently, the cells were cultured in the dark for 4 h, and DMSO was added to the medium. The OD of each well was observed at A490 nm via a spectrophotometer (Epoch 2; BioTek Instruments, Inc., Winooski, VT, USA) [38].

Immunofluorescence analysis and confocal microscopy

Cells were plated on glass slides in a 6-well plate at a density of 1 × 106 cells per well. Subsequently, cells were fixed in ice-cold 4% paraformaldehyde for 30 min, permeabilized with 0.1% Triton X-100, and blocked with 2% gelatine in PBS at room temperature. The cells were then incubated with the primary antibodies: [cyt-c (1:1000; Abcam; #ab90529), Tom20 (mitochondrial marker, 1:1000, Abcam, #ab186735), LAMP1 (lysosome marker, 1:1000, Abcam, #ab24170), MIEF1 (1:1000, Abcam, #ab89944)] [39].

Mitochondrial membrane potential measurement and ATP detection

Mitochondrial membrane potential was measured with JC-1 assays (Thermo Fisher Scientific Inc., Waltham, MA, USA; Catalogue No. M34152). Cells were treated with 5 mM JC-1 and then cultured in the dark for 30 min at 37 °C [40]. Subsequently, cold PBS was used to remove the free JC-1, and DAPI was used to stain the nucleus in the dark for 3 min at 37 °C. The mitochondrial membrane potential was observed under a digital microscope (IX81, Olympus) [41]. Cellular ATP content was measured according to a previous report via ELISA assay. Cells were washed with PBS and then collected at room temperature. Subsequently, a luciferase-based ATP assay kit (Celltiter-Glo Luminescent Cell Viability assay; Promega, Madison, WI, USA; Catalogue No. A22066) was used according to the instructions.

Western blot

Total protein was extracted by RIPA (R0010, Solarbio Science and Technology, Beijing, China), and the protein concentration of each sample was detected with a bicinchoninic acid (BCA) kit (20201ES76, Yeasen Biotech Co., Ltd, Shanghai, China) [42]. Deionized water was added to generate 30-µg protein samples for each lane. A 10% sodium dodecyl sulphate (SDS) separation gel and concentration gel were prepared [43]. The following diluted primary antibodies were added to the membrane and incubated overnight: JNK (1:1000; Cell Signaling Technology, #4672), p-JNK (1:1000; Cell Signaling Technology, #9251), Mst1 (1:1000, Cell Signaling Technology, #3682), Yap (1:1000; Cell Signaling Technology, #14,074), Mfn2 (1:1000, Abcam, #ab56889), Bcl2 (1:1000, Cell Signaling Technology, #3498), Bax (1:1000, Cell Signaling Technology, #2772), Cyclin D (1:1000, Abcam, #ab134175), CDK4 (1:1000, Abcam, #ab137675), Drp1 (1:1000, Abcam, #ab56788), Fis1 (1:1000, Abcam, #ab71498), Opa1 (1:1000, Abcam, #ab42364), Mff (1:1000, Cell Signaling Technology, #86668).

RNA isolation and qPCR

TRIzol reagent (Invitrogen; Thermo Fisher Scientific, Inc.) was used to isolate total RNA from cells. Subsequently, the Reverse Transcription kit (Kaneka Eurogentec S.A., Seraing, Belgium) was applied to transcribe RNA (1 μg in each group) into cDNA at room temperature (~ 25 °C) for 30 min. The qPCR was performed with primers using SYBR™ Green PCR Master Mix (Thermo Fisher Scientific, Inc. Cat. No. 4309155). The following were primers used in the present study: ROCK-1 (Forward: 5′-ACCTGTAACCCAAGGAGATGTG-3′, Reverse 5′-CACAATTGGCAGGAAAGTGG-3′), and Rac1 (Forward 5′-ATGCAGGCCATCAAGTGTGTGG-3′, Reverse: 5′-TTACAACAGCAGGCATTTTCTC-3′), and GAPDH, (Forward: 5′-AAGTTGTGFATTAGTCA-3′, Reverse 5′-AGAATAGTCCTATAATCA-3′).

ELISA

The Caspase 9 Activity Assay Kit (Beyotime, China, Cat. No: C1158) was used to measure the activity of caspase-9 according to the manufacturer’s instructions. The concentrations of GSH, SOD and GPX were evaluated using commercial kits (Cellular Glutathione Peroxidase Assay Kit, Beyotime, China, Cat. No: S0056; Glutathione Reductase Assay Kit, Beyotime, China, Cat. No: S0055; Total Superoxide Dismutase Assay Kit, Beyotime, China, Cat. No: S0101, respectively). ATP production was measured using a luciferase-based ATP assay kit (Beyotime Institute of Biotechnology) with a microplate reader [44].

Cellular proliferation evaluation and LDH release assay

Cellular proliferation was evaluated via EdU assay. Cells were seeded onto a 6-well plate, and the Cell-Light™ EdU Apollo®567 In Vitro Imaging Kit (Thermo Fisher Scientific Inc., Waltham, MA, USA; Catalogue No. A10044) was used to observe the EdU-positive cells according to the manufacturer’s instructions. Cellular lactate production in the medium was measured via a lactate assay kit (#K607-100; BioVision, Milpitas, CA, USA) according to a previous study [45].

Transwell assay

For Transwell migration assays, the upper chambers of 24-well Transwell assay plates were seeded with 2 × 103 cells in 200 μL serum-free medium per well. The lower chambers were filled with 600 μL medium containing 0.5% FBS. After a 24-h incubation in a humidified incubator at 37 °C, 5% CO2, cells that had migrated to the underside of the membranes were fixed and stained with 0.1% crystal violet. After washing with distilled water, pictures of each chamber were randomly taken using a 200× microscope field, and these images were used to quantify the total number of migrated cells.

Statistical analysis

SPSS 21.0 software (IBM Corp., Armonk, New York, USA) was applied for data analysis. All experiments were repeated 3 times in each group. The mean value of the measurement data was expressed as the mean and SEM. Comparisons among groups were by one-way analysis of variance (ANOVA), and multiple comparisons between the average number of samples were performed by LSD analysis. p < 0.05 indicated that the difference was statistically significant.

Results

Mst1 overexpression combined with Yap knockdown further promotes thyroid carcinoma cell apoptosis

In the present study, Mst1 adenovirus and/or Yap shRNA were transfected into MDA-T32 cells. Then, the knockdown and overexpression efficiencies were confirmed via western blotting. As shown in Fig. 1a, c, compared to the control group, Ms1 adenovirus (ad-Mst1) enhanced Mst1 expression, whereas Yap shRNA transfection decreased the Yap level in MDAT-32 cells. To observe the influence of Mst1 overexpression and Yap knockdown on cell viability, an MTT assay was used. Both Mst1 overexpression and Yap knockdown reduced cell viability compared to that of the control group (Fig. 1d). Interestingly, Mst1 overexpression in combination with Yap knockdown further suppressed cell viability in MDA-T32 cells. This finding was consistent with the result of the LDH release assay. As shown in Fig. 1e, both Mst1 overexpression and Yap knockdown significantly increased the LDH content in the medium. However, LDH release was further enhanced by Mst1 overexpression in combination with Yap knockdown. The above data indicate that Mst1 overexpression cooperates with Yap knockdown to further promote thyroid cancer cell death in vitro. This finding was further validated by counting the TUNEL-positive cells. As shown in Fig. 1f, g, the number of TUNEL+ MDA-T32 cells was rapidly elevated after Yap was silenced and/or Mst1 was overexpressed compared to that in the control group. However, in cells with simultaneous Yap knockdown and Mst1 overexpression, the number of TUNEL+ cells was further increased compared to that in cells with Yap inhibition or Mst1 overexpression alone. At the molecular level, caspase activation is a marker of cell apoptosis; therefore, ELISA was used to observe the alteration in caspase-3 activation in response to Mst1 overexpression and/or Yap knockdown. As shown in Fig. 1h, the activity of caspase-3 was rapidly increased in Mst1-overexpressing cells and/or Yap-silenced cells compared to that in the control group. However, Mst1 overexpression in combination with Yap knockdown further elevated caspase-3 activity in MDA-T32 cells. Similar results were noted in MDA-T68 cells (Additional file 1: Figure S1B). Altogether, our data illustrate that Mst1 overexpression and Yap knockdown have synergistic effects that further reduce cell death in MDA-T32 cells.

Mst1 overexpression enhances Yap silencing-inhibited cancer proliferation and migration

Next, experiments were performed to analyze the influence of Mst1 overexpression and Yap knockdown on cell proliferation and migration. Western blot analysis demonstrated that the levels of cell cycle proteins such as Cyclin D1 and CDK4 were markedly reduced in cells transfected with Mst1 adenovirus and Yap shRNA (Fig. 2a–c), indicating the inhibitory effects of Mst1 activation and Yap inhibition on cell proliferation. This finding was further quantified via EdU staining. The number of EdU-positive cells indicated the ratio of proliferating cells in the cell cycle. As shown in Fig. 2d, e, both Mst1 overexpression and Yap knockdown repressed the ratio of proliferating MDA-T32 cells compared to that in the control group. However, cell proliferation was further suppressed by Mst1 overexpression in combination with Yap knockdown. In addition to cell proliferation assays, experiments were conducted to assess cell migration. Transwell assays showed that the number of migrated cells was rapidly reduced in response to either Mst1 overexpression or Yap knockdown (Fig. 2g, h). Interestingly, combined treatment with Mst1 adenovirus and Yap shRNA further suppressed MDA-T32 cell motility, indicating the essential roles played by Mst1 and Yap in modulating MDA-T32 cell motility. This finding was further supported by analyzing the transcription of prometastatic genes. As shown in Fig. 2i–g, the transcription levels of prometastatic genes such as Rac1 and ROCK1 were markedly reduced in response to either Mst1 overexpression or Yap knockdown. Interestingly, in cells with combined Mst1 overexpression and Yap knockdown, the transcription levels of Rac1 and ROCK1 were further reduced. These results were also noted in MDA-T68 cells (Additional file 1: Figure S1C, D). Therefore, the above data indicate that Mst1 and Yap differentially regulate cancer cell proliferation and migration.

Mitochondrial dysfunction is further enhanced by Mst1 overexpression combined with Yap knockdown

Based on the results of previous studies, the mitochondrion is the potential target of Mst1/Yap, and mitochondrial dysfunction has been found to be associated with cancer cell death. In the present study, we sought to determine whether Mst1 overexpression cooperated with Yap knockdown to further affect mitochondrial function. First, a JC-1 probe was used to stain mitochondria to assess the mitochondrial membrane potential. Red fluorescence indicated a normal mitochondrial membrane potential, whereas green fluorescence reflected a decreased mitochondrial membrane potential. As shown in Fig. 3a, b, either Yap deletion or Mst1 overexpression significantly reduced the mitochondrial membrane potential compared to that in the control group, as evidenced by the decreased ratio of red to green fluorescence. Interestingly, Mst1 overexpression combined with Yap knockdown further decreased the mitochondrial membrane potential. At the molecular level, a reduction in the mitochondrial membrane potential is an early feature of mitochondrial apoptosis, and the consequence of mitochondrial membrane potential dissipation is the opening of the mitochondrial permeability transition pore (mPTP). Via ELISA, we found that the mPTP opening rate was rapidly increased in cells treated with either Mst1 adenovirus or Yap shRNA (Fig. 3c). Interestingly, Mst1 overexpression in combination with Yap knockdown further promoted mPTP opening (Fig. 3c).
Excessive mPTP opening increases the likelihood of nuclear translocation of the mitochondrial proapoptotic protein cyt-c. As shown in Fig. 3d, e, the nuclear expression of cyt-c was significantly elevated in treated cells compared to that in control cells, indicating the release of mitochondrial cyt-c to the nucleus. Interestingly, this effect was further enhanced via the combination of Mst1 overexpression and Yap knockdown. In addition to cyt-c leakage, the production of mitochondrial ROS was markedly increased in cells transfected with either Mst1 adenovirus or Yap shRNA (Fig. 3f, g). Interestingly, mitochondrial oxidative stress was further augmented via combined Mst1 overexpression and Yap knockdown (Fig. 3f, g). Finally, western blot analysis demonstrated that the levels of mitochondrial proapoptotic proteins were significantly elevated by Mst1 overexpression in combination with Yap deletion (Fig. 3h–l). In contrast, the levels of antiapoptotic factors were markedly reduced by combined Mst1 activation and Yap inhibition (Fig. 3h–l). These results were also observed in MDA-T68 cells (Additional file 1: Figure S1E, F). Taken together, the above data indicate that mitochondrial function was highly modulated by Yap and Mst1. Combined treatment via Mst1 upregulation and Yap downregulation further augmented mitochondrial dysfunction in MDA-T32 cells.

Mitochondrial dynamics are impaired by Mst1 overexpression and Yap knockdown

In addition to identifying mitochondrial malfunction, we also sought to determine whether mitochondrial structure was affected by Mst1/Yap. Recent studies reported that mitochondrial function is modulated by mitochondrial structural alterations and especially mitochondrial dynamics. Based on this knowledge, we explored whether mitochondrial dynamics were controlled by Mst1 and Yap in MDA-T32 cells. Immunofluorescence assays showed that mitochondria in normal cells exhibited an intact shuttle structure. However, loss of Yap or overexpression of Mst1 promoted mitochondrial division into several fragments (Fig. 4a, b). This alteration was more prominent in cells with both Yap knockdown and Mst1 overexpression. Subsequently, mitochondrial length was measured to indicate mitochondrial dynamics. As shown in Fig. 4b, the mitochondrial length was ~ 9.3 μm in normal cells and was reduced to ~ 4.2 μm after either knockdown of Yap or overexpression of Mst1. Notably, combined treatment via Mst1 upregulation and Yap downregulation further decreased the mitochondrial length to ~ 2.6 μm. Accordingly, the above results confirmed that mitochondrial dynamics were affected by Mst1 and Yap.
Subsequently, western blotting was performed to analyze the expression of proteins related to mitochondrial dynamics. Drp1/Mff are the factors involved in mitochondrial division. However, Mfn2/Opa1 are the proteins that positively contribute to the fusion of mitochondrial fragments. As shown in Fig. 4c–g, the levels of Drp1 and Mff were rapidly increased in response to Mst1 overexpression compared to those in the control group, and a similar effect was observed in Yap-silenced cells. Notably, Mst1 overexpression combined with Yap knockdown further promoted the upregulation of Mff/Drp1. In contrast, the expression of Mfn2 and Opa1 was significantly downregulated in response to Mst1 overexpression (Fig. 4c–g), consistent with the effect of Yap silencing. Notably, the combination of Mst1 overexpression and Yap knockdown further inhibited Mfn2/Opa1 expression. Taken together, the above data indicate that mitochondrial structural homeostasis was highly affected by Mst1 and Yap.

Mst1 upregulation and Yap downregulation are associated with activation of the JNK-MIEF1 pathway

Next, experiments were performed to determine the downstream effectors of Mst1/Yap in MDA-T32 cells. Previous studies have reported that the JNK pathway could be affected by Mst1 and Yap in different disease states, such as postinfarction myocardial injury and renal ischemia reperfusion stress [46]. Regarding the central role played by JNK in initiating mitochondrial damage, we asked whether JNK was simultaneously modulated by Mst1 and Yap in MDA-T32 cells. Western blot analysis showed that the level of p-JNK was increased ~ 2.5-fold in both Mst1-overexpressing cells and Yap-silenced cells (Fig. 5a–c). Interestingly, Mst1 overexpression in combination with Yap knockdown further elevated the level of p-JNK. This finding indicated that the combination of Mst1 activation and Yap inhibition could further upregulate the activity of the JNK pathway.
Recently, MIEF1 was found to be a novel mediator of mitochondrial function via the modulation of mitochondrial fission and mitochondrial apoptosis [47, 48]. Via western blot analysis, we found that the expression of MIEF1 was slightly increased in response to either Mst1 overexpression or Yap deletion (Fig. 5a–c). Interestingly, cotreatment with Mst1 adenovirus and Yap shRNA further increased the expression of MIEF1 in MDA-T32 cells (Fig. 5a–c). Subsequently, to verify whether MIEF1 was the downstream effector of the JNK pathway, a pathway blocker was used to prevent JNK activation, and the expression of MIEF1 was monitored. As shown in Fig. 5a–c, after treatment with SP600125, the level of p-JNK was reversed to normal levels despite cotreatment with Mst1 adenovirus and Yap shRNA. Interestingly, JNK inhibition also abolished the upregulation of MIEF1 expression induced by Mst1 overexpression and Yap knockdown. Therefore, the above data confirmed that the JNK-MIEF1 pathway was activated by Mst1/Yap in MDA-T31 cells.
This result was further supported via immunofluorescence. As shown in Fig. 5d, e, the fluorescence intensity of MIEF1 was rapidly increased compared to that in the control group after Mst1 overexpression and Yap inhibition. However, blockade of JNK suppressed MIEF1 expression despite cotreatment with Mst1 adenovirus and Yap shRNA. In addition, a coimmunofluorescence assay demonstrated that the average mitochondrial length was reduced in response to Mst1 overexpression and Yap knockdown. However, SP600125 treatment reversed the mitochondrial length in MDA-T32 cells (Fig. 5d, e). Taken together, the above data indicated that the combination of Mst1 upregulation and Yap deletion was connected with the activation of the JNK-MIEF1 pathway.

Inhibition of the JNK pathway abolishes the regulatory effects of Mst1 overexpression and Yap knockdown on cell viability and mitochondrial stress

To demonstrate whether the JNK pathway was also involved in mitochondrial stress and MDA-T32 cell death, we assessed mitochondrial function and cell viability using SP600125 in the presence of Mst1 overexpression/Yap knockdown. First, ATP production was downregulated in response to combined Mst1 overexpression and Yap knockdown (Fig. 6a). Interestingly, this effect was reversed by SP600125, indicating that the JNK pathway was involved in Mst1/Yap-mediated mitochondrial energy metabolism. In addition, an immunofluorescence assay showed that the Mst1/Yap modification-mediated release of cyt-c to the nucleus was reversed by SP600125 (Fig. 6b, c). In addition to cyt-c translocation, mitochondrial oxidative stress was evaluated by measuring the levels of cellular antioxidants. As shown in Fig. 6d–f, the SOD, GPX and GSH levels were significantly decreased in cells cotreated with Mst1 adenovirus and Yap shRNA. However, after treatment with SP600125, the levels of SOD, GSH and GPX were significantly increased to near-normal levels, indicating that mitochondrial oxidative stress was also affected by Mst1/Yap via the JNK pathway. Next, cell viability was evaluated in response to JNK inhibition in the presence of Mst1/Yap modification. As shown in Fig. 6g, cell viability was markedly repressed by Mst1 overexpression in combination with Yap deletion; this effect was reversed by SP600125. Similar results were also obtained in the LDH release assay, which showed that Mst1/Yap modification-mediated LDH release was inhibited by SP600125, highlighting the essential role played by the JNK pathway in mediating cell viability. Taken together, our results indicated that the JNK pathway was required for Mst1/Yap modification-mediated mitochondrial stress and cell death in MDA-T32 cells.

Discussion

During the last two decades, most studies have focused on the pathogenesis of thyroid cancer development and progression. In the present study, we found that the viability of thyroid cancer cells was associated with mitochondrial function, Mst1 expression, Yap levels and JNK-MIEF1 pathway activity. Molecular studies indicated that Mst1 overexpression or Yap knockdown reduced the viability of thyroid cancer MDA-T32 cells in vitro and that this process was closely associated with mitochondrial stress, as evidenced by the observed mitochondrial malfunction and mitochondrial structural disorder. Further, we found that the combination of Mst1 upregulation and Yap inhibition further increased the apoptotic rate of thyroid cancer MDA-T32 cells in vitro by augmenting mitochondrial damage and activating the JNK-MIEF1 pathway. However, blockade of the JNK pathway abolished the regulatory effects of Mst1/Yap modification in MDA-T32 cells. To our knowledge, this study is the first to investigate the synergistic effects of Mst1 overexpression and Yap knockdown on the viability of MDA-T32 cells. In this report, we show how to differentially modify the components of the Hippo pathway in order to further enhance cancer cell death, and the findings described in this manuscript are particularly applicable for designing new drugs to treat thyroid carcinoma by targeting the Hippo pathway.
Many experiments have been conducted to understand the biological significance of the Hippo pathway in tumorigenesis. Dysregulation of the Hippo pathway has been found to be an effective way to limit tumor progression. For example, dephosphorylation of Yap interrupts glucose uptake through the Bcl-XL/GLUT1 pathway in human gastric cancer [49]. In addition, the levels of Yas could be used as an early marker to evaluate breast cancer progression [50]. Moreover, modulation of Yap via ailanthone inhibits bladder cancer in a manner dependent on Nfr2 downregulation and c-Myc inhibition [51]. In liver cancer, loss of Yap attenuates cancer metastasis and mobilization through impairing lamellipodium formation and inactivating the JNK–Bnip3–SERCA–CaMII pathway [52]. With respect to Mst1, overexpression of Mst1 via tanshinone IIA increases the therapeutic sensitivity of colorectal cancer to IL-2-mediated cytokine therapy. In lung cancer, Mst1 upregulation impairs mitochondrial energy metabolism and ultimately impedes cancer migration and movement via the ROCK1/F-actin pathway [53]. In addition, the antitumor effect of marine-origin compounds could be abolished by Mst1 inhibition in liver cancer [54]. Therefore, the above data indicate that Yap and Mst1 seem to play different roles in regulating the cancer phenotype. However, no study has explored the synergistic or antagonistic molecular effects mediated by Yap and Mst1 in thyroid cancer. In the present study, we found that Mst1 overexpression induced cancer cell death, an effect that was similar to that of Yap knockdown. Interestingly, Mst1 overexpression in combination with Yap knockdown further promoted cancer cell death by exacerbating mitochondrial stress. This result indicates that differential regulation of the core components in the Hippo pathway is potentially a novel therapeutic tool for the treatment of thyroid cancer.
At the molecular level, we found that mitochondrial dysfunction, activated by Mst1/Yap modification, was implicated in cancer cell death. After Yap loss and Mst1 overexpression, mitochondrial membrane potential was reduced, an effect that was followed by mitochondrial ROS overproduction. In addition, mitochondrial dynamics were disturbed by Mst1/Yap modification, as evidenced by mitochondrial fragmentation. This result indicates that mitochondria could be considered a potential target of the Hippo pathway. Our data were in accordance with those of previous studies. In glioblastoma [55], gastric cancer, and rectal cancer, Yap dysfunction is associated with mitochondrial damage, including mitochondrial apoptosis activation, mitochondrial fission initiation and mitochondrial oxidative stress [56]. Similarly, in lung cancer and liver cancer, mitochondrial injury is triggered by Mst1 activation. This study showed that mitochondria are a potential target for thyroid cancer therapy, and further research should be undertaken to facilitate this therapeutic application.
Finally, we reported that the JNK-MIEF1 pathway was activated by Mst1/Yap modification. At the molecular level, MIEF1 is a novel mitochondrial damage mediator [57]. In cardiac ischemia reperfusion stress, MIEF1 is upregulated, and the levels of MIEF1 are tightly correlated with the degree of myocardial injury [58]. In addition, after exposure to UV radiation, MIEF1 expression is deregulated, and this alteration has been demonstrated to play a decisive role in initiating epidermal cell death [59]. In the present study, we provide evidence to support the influence of MIEF1 on mitochondrial damage in thyroid cancer [60]. MIEF1 expression was increased in response to Mst1 overexpression and/or Yap knockdown via the JNK pathway. However, the detailed role of MIEF1 in cancer cell death and mitochondrial damage has not been fully explained. More studies are required to determine the detailed role played by MIEF1 in biological functions in cancer [61].

Conclusions

Altogether, our data suggest that Mst1 activation and Yap inhibition coordinate to augment thyroid cancer cell death by controlling the JNK-MIEF1-mitochondria pathway. Based on this finding, we gain further insight into the interactive mechanism between Mst1, Yap, mitochondrial function and thyroid cancer cell death.

Acknowledgements

Not applicable.
Not applicable.
Not applicable.

Competing interests

The authors declare that they have no competing interests.
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.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Reddy KRK, Dasari C, Duscharla D, Supriya B, Ram NS, Surekha MV, Kumar JM, Ummanni R. Dimethylarginine dimethylaminohydrolase-1 (DDAH1) is frequently upregulated in prostate cancer, and its overexpression conveys tumor growth and angiogenesis by metabolizing asymmetric dimethylarginine (ADMA). Angiogenesis. 2018;21(1):79–94.PubMedCrossRef Reddy KRK, Dasari C, Duscharla D, Supriya B, Ram NS, Surekha MV, Kumar JM, Ummanni R. Dimethylarginine dimethylaminohydrolase-1 (DDAH1) is frequently upregulated in prostate cancer, and its overexpression conveys tumor growth and angiogenesis by metabolizing asymmetric dimethylarginine (ADMA). Angiogenesis. 2018;21(1):79–94.PubMedCrossRef
2.
Zurück zum Zitat Kalyanaraman B, Cheng G, Hardy M, Ouari O, Lopez M, Joseph J, Zielonka J, Dwinell MB. A review of the basics of mitochondrial bioenergetics, metabolism, and related signaling pathways in cancer cells: therapeutic targeting of tumor mitochondria with lipophilic cationic compounds. Redox Biol. 2018;14:316–27.PubMedCrossRef Kalyanaraman B, Cheng G, Hardy M, Ouari O, Lopez M, Joseph J, Zielonka J, Dwinell MB. A review of the basics of mitochondrial bioenergetics, metabolism, and related signaling pathways in cancer cells: therapeutic targeting of tumor mitochondria with lipophilic cationic compounds. Redox Biol. 2018;14:316–27.PubMedCrossRef
3.
Zurück zum Zitat Tabish TA, Zhang S, Winyard PG. Developing the next generation of graphene-based platforms for cancer therapeutics: the potential role of reactive oxygen species. Redox Biol. 2018;15:34–40.PubMedCrossRef Tabish TA, Zhang S, Winyard PG. Developing the next generation of graphene-based platforms for cancer therapeutics: the potential role of reactive oxygen species. Redox Biol. 2018;15:34–40.PubMedCrossRef
4.
Zurück zum Zitat Proietti S, Catizone A, Masiello MG, Dinicola S, Fabrizi G, Minini M, Ricci G, Verna R, Reiter RJ, Cucina A, et al. Increase in motility and invasiveness of MCF7 cancer cells induced by nicotine is abolished by melatonin through inhibition of ERK phosphorylation. J Pineal Res. 2018;64(4):e12467.PubMedCrossRef Proietti S, Catizone A, Masiello MG, Dinicola S, Fabrizi G, Minini M, Ricci G, Verna R, Reiter RJ, Cucina A, et al. Increase in motility and invasiveness of MCF7 cancer cells induced by nicotine is abolished by melatonin through inhibition of ERK phosphorylation. J Pineal Res. 2018;64(4):e12467.PubMedCrossRef
5.
Zurück zum Zitat Shen YQ, Guerra-Librero A, Fernandez-Gil BI, Florido J, Garcia-Lopez S, Martinez-Ruiz L, Mendivil-Perez M, Soto-Mercado V, Acuna-Castroviejo D, Ortega-Arellano H, et al. Combination of melatonin and rapamycin for head and neck cancer therapy: suppression of AKT/mTOR pathway activation, and activation of mitophagy and apoptosis via mitochondrial function regulation. J Pineal Res. 2018;64(3):e12461.CrossRef Shen YQ, Guerra-Librero A, Fernandez-Gil BI, Florido J, Garcia-Lopez S, Martinez-Ruiz L, Mendivil-Perez M, Soto-Mercado V, Acuna-Castroviejo D, Ortega-Arellano H, et al. Combination of melatonin and rapamycin for head and neck cancer therapy: suppression of AKT/mTOR pathway activation, and activation of mitophagy and apoptosis via mitochondrial function regulation. J Pineal Res. 2018;64(3):e12461.CrossRef
6.
Zurück zum Zitat Fukumoto M, Kondo K, Uni K, Ishiguro T, Hayashi M, Ueda S, Mori I, Niimi K, Tashiro F, Miyazaki S, et al. Tip-cell behavior is regulated by transcription factor FoxO1 under hypoxic conditions in developing mouse retinas. Angiogenesis. 2018;21(2):203–14.PubMedCrossRef Fukumoto M, Kondo K, Uni K, Ishiguro T, Hayashi M, Ueda S, Mori I, Niimi K, Tashiro F, Miyazaki S, et al. Tip-cell behavior is regulated by transcription factor FoxO1 under hypoxic conditions in developing mouse retinas. Angiogenesis. 2018;21(2):203–14.PubMedCrossRef
7.
Zurück zum Zitat Abukar Y, Ramchandra R, Hood SG, McKinley MJ, Booth LC, Yao ST, May CN. Increased cardiac sympathetic nerve activity in ovine heart failure is reduced by lesion of the area postrema, but not lamina terminalis. Basic Res Cardiol. 2018;113(5):35.PubMedCrossRef Abukar Y, Ramchandra R, Hood SG, McKinley MJ, Booth LC, Yao ST, May CN. Increased cardiac sympathetic nerve activity in ovine heart failure is reduced by lesion of the area postrema, but not lamina terminalis. Basic Res Cardiol. 2018;113(5):35.PubMedCrossRef
8.
Zurück zum Zitat Abeysuriya RG, Lockley SW, Robinson PA, Postnova S. A unified model of melatonin, 6-sulfatoxymelatonin, and sleep dynamics. J Pineal Res. 2018;64(4):e12474.PubMedCrossRef Abeysuriya RG, Lockley SW, Robinson PA, Postnova S. A unified model of melatonin, 6-sulfatoxymelatonin, and sleep dynamics. J Pineal Res. 2018;64(4):e12474.PubMedCrossRef
9.
Zurück zum Zitat Garcia-Rendueles ME, Ricarte-Filho JC, Untch BR, Landa I, Knauf JA, Voza F, Smith VE, Ganly I, Taylor BS, Persaud Y, et al. NF2 loss promotes oncogenic RAS-induced thyroid cancers via YAP-dependent transactivation of RAS proteins and sensitizes them to MEK inhibition. Cancer Discov. 2015;5(11):1178–93.PubMedPubMedCentralCrossRef Garcia-Rendueles ME, Ricarte-Filho JC, Untch BR, Landa I, Knauf JA, Voza F, Smith VE, Ganly I, Taylor BS, Persaud Y, et al. NF2 loss promotes oncogenic RAS-induced thyroid cancers via YAP-dependent transactivation of RAS proteins and sensitizes them to MEK inhibition. Cancer Discov. 2015;5(11):1178–93.PubMedPubMedCentralCrossRef
10.
Zurück zum Zitat Lee SJ, Lee MH, Kim DW, Lee S, Huang S, Ryu MJ, Kim YK, Kim SJ, Kim SJ, Hwang JH, et al. Cross-regulation between oncogenic BRAF(V600E) kinase and the MST1 pathway in papillary thyroid carcinoma. PLoS ONE. 2011;6(1):e16180.PubMedPubMedCentralCrossRef Lee SJ, Lee MH, Kim DW, Lee S, Huang S, Ryu MJ, Kim YK, Kim SJ, Kim SJ, Hwang JH, et al. Cross-regulation between oncogenic BRAF(V600E) kinase and the MST1 pathway in papillary thyroid carcinoma. PLoS ONE. 2011;6(1):e16180.PubMedPubMedCentralCrossRef
11.
Zurück zum Zitat Lowe NM, Loughran S, Slevin NJ, Yap BK. Anaplastic thyroid cancer: the addition of systemic chemotherapy to radiotherapy led to an observed improvement in survival—a single centre experience and review of the literature. Sci World J. 2014;2014:674583.CrossRef Lowe NM, Loughran S, Slevin NJ, Yap BK. Anaplastic thyroid cancer: the addition of systemic chemotherapy to radiotherapy led to an observed improvement in survival—a single centre experience and review of the literature. Sci World J. 2014;2014:674583.CrossRef
12.
Zurück zum Zitat Gianni-Barrera R, Butschkau A, Uccelli A, Certelli A, Valente P, Bartolomeo M, Groppa E, Burger MG, Hlushchuk R, Heberer M, et al. PDGF-BB regulates splitting angiogenesis in skeletal muscle by limiting VEGF-induced endothelial proliferation. Angiogenesis. 2018;21(4):883–900.PubMedPubMedCentralCrossRef Gianni-Barrera R, Butschkau A, Uccelli A, Certelli A, Valente P, Bartolomeo M, Groppa E, Burger MG, Hlushchuk R, Heberer M, et al. PDGF-BB regulates splitting angiogenesis in skeletal muscle by limiting VEGF-induced endothelial proliferation. Angiogenesis. 2018;21(4):883–900.PubMedPubMedCentralCrossRef
13.
Zurück zum Zitat Coverstone ED, Bach RG, Chen L, Bierut LJ, Li AY, Lenzini PA, O’Neill HC, Spertus JA, Sucharov CC, Stitzel JA, et al. A novel genetic marker of decreased inflammation and improved survival after acute myocardial infarction. Basic Res Cardiol. 2018;113(5):38.PubMedPubMedCentralCrossRef Coverstone ED, Bach RG, Chen L, Bierut LJ, Li AY, Lenzini PA, O’Neill HC, Spertus JA, Sucharov CC, Stitzel JA, et al. A novel genetic marker of decreased inflammation and improved survival after acute myocardial infarction. Basic Res Cardiol. 2018;113(5):38.PubMedPubMedCentralCrossRef
14.
Zurück zum Zitat Brazao V, Colato RP, Santello FH, Vale GTD, Gonzaga NA, Tirapelli CR, Prado JCD Jr. Effects of melatonin on thymic and oxidative stress dysfunctions during Trypanosoma cruzi infection. J Pineal Res. 2018;65(3):e12510.PubMedCrossRef Brazao V, Colato RP, Santello FH, Vale GTD, Gonzaga NA, Tirapelli CR, Prado JCD Jr. Effects of melatonin on thymic and oxidative stress dysfunctions during Trypanosoma cruzi infection. J Pineal Res. 2018;65(3):e12510.PubMedCrossRef
15.
Zurück zum Zitat Ba X, Boldogh I. 8-Oxoguanine DNA glycosylase 1: beyond repair of the oxidatively modified base lesions. Redox Biol. 2018;14:669–78.PubMedCrossRef Ba X, Boldogh I. 8-Oxoguanine DNA glycosylase 1: beyond repair of the oxidatively modified base lesions. Redox Biol. 2018;14:669–78.PubMedCrossRef
16.
Zurück zum Zitat Biernacki M, Ambrozewicz E, Gegotek A, Toczek M, Bielawska K, Skrzydlewska E. Redox system and phospholipid metabolism in the kidney of hypertensive rats after FAAH inhibitor URB597 administration. Redox Biol. 2018;15:41–50.PubMedCrossRef Biernacki M, Ambrozewicz E, Gegotek A, Toczek M, Bielawska K, Skrzydlewska E. Redox system and phospholipid metabolism in the kidney of hypertensive rats after FAAH inhibitor URB597 administration. Redox Biol. 2018;15:41–50.PubMedCrossRef
17.
Zurück zum Zitat Davidson SM, Arjun S, Basalay MV, Bell RM, Bromage DI, Botker HE, Carr RD, Cunningham J, Ghosh AK, Heusch G, et al. The 10th Biennial Hatter Cardiovascular Institute workshop: cellular protection-evaluating new directions in the setting of myocardial infarction, ischaemic stroke, and cardio-oncology. Basic Res Cardiol. 2018;113(6):43.PubMedPubMedCentralCrossRef Davidson SM, Arjun S, Basalay MV, Bell RM, Bromage DI, Botker HE, Carr RD, Cunningham J, Ghosh AK, Heusch G, et al. The 10th Biennial Hatter Cardiovascular Institute workshop: cellular protection-evaluating new directions in the setting of myocardial infarction, ischaemic stroke, and cardio-oncology. Basic Res Cardiol. 2018;113(6):43.PubMedPubMedCentralCrossRef
18.
Zurück zum Zitat Gonzalez NR, Liou R, Kurth F, Jiang H, Saver J. Antiangiogenesis and medical therapy failure in intracranial atherosclerosis. Angiogenesis. 2018;21(1):23–35.PubMedCrossRef Gonzalez NR, Liou R, Kurth F, Jiang H, Saver J. Antiangiogenesis and medical therapy failure in intracranial atherosclerosis. Angiogenesis. 2018;21(1):23–35.PubMedCrossRef
19.
Zurück zum Zitat DeLeon-Pennell KY, Mouton AJ, Ero OK, Ma Y, Padmanabhan Iyer R, Flynn ER, Espinoza I, Musani SK, Vasan RS, Hall ME, et al. LXR/RXR signaling and neutrophil phenotype following myocardial infarction classify sex differences in remodeling. Basic Res Cardiol. 2018;113(5):40.PubMedPubMedCentralCrossRef DeLeon-Pennell KY, Mouton AJ, Ero OK, Ma Y, Padmanabhan Iyer R, Flynn ER, Espinoza I, Musani SK, Vasan RS, Hall ME, et al. LXR/RXR signaling and neutrophil phenotype following myocardial infarction classify sex differences in remodeling. Basic Res Cardiol. 2018;113(5):40.PubMedPubMedCentralCrossRef
20.
Zurück zum Zitat Yao S, Yan W. Overexpression of Mst1 reduces gastric cancer cell viability by repressing the AMPK-Sirt3 pathway and activating mitochondrial fission. Onco Targets Ther. 2018;11:8465–79.PubMedPubMedCentralCrossRef Yao S, Yan W. Overexpression of Mst1 reduces gastric cancer cell viability by repressing the AMPK-Sirt3 pathway and activating mitochondrial fission. Onco Targets Ther. 2018;11:8465–79.PubMedPubMedCentralCrossRef
21.
Zurück zum Zitat Chandra M, Escalante-Alcalde D, Bhuiyan MS, Orr AW, Kevil C, Morris AJ, Nam H, Dominic P, McCarthy KJ, Miriyala S, et al. Cardiac-specific inactivation of LPP3 in mice leads to myocardial dysfunction and heart failure. Redox Biol. 2018;14:261–71.PubMedCrossRef Chandra M, Escalante-Alcalde D, Bhuiyan MS, Orr AW, Kevil C, Morris AJ, Nam H, Dominic P, McCarthy KJ, Miriyala S, et al. Cardiac-specific inactivation of LPP3 in mice leads to myocardial dysfunction and heart failure. Redox Biol. 2018;14:261–71.PubMedCrossRef
22.
Zurück zum Zitat Korbel C, Gerstner MD, Menger MD, Laschke MW. Notch signaling controls sprouting angiogenesis of endometriotic lesions. Angiogenesis. 2018;21(1):37–46.PubMedCrossRef Korbel C, Gerstner MD, Menger MD, Laschke MW. Notch signaling controls sprouting angiogenesis of endometriotic lesions. Angiogenesis. 2018;21(1):37–46.PubMedCrossRef
23.
Zurück zum Zitat Edwards KS, Ashraf S, Lomax TM, Wiseman JM, Hall ME, Gava FN, Hall JE, Hosler JP, Harmancey R. Uncoupling protein 3 deficiency impairs myocardial fatty acid oxidation and contractile recovery following ischemia/reperfusion. Basic Res Cardiol. 2018;113(6):47.PubMedPubMedCentralCrossRef Edwards KS, Ashraf S, Lomax TM, Wiseman JM, Hall ME, Gava FN, Hall JE, Hosler JP, Harmancey R. Uncoupling protein 3 deficiency impairs myocardial fatty acid oxidation and contractile recovery following ischemia/reperfusion. Basic Res Cardiol. 2018;113(6):47.PubMedPubMedCentralCrossRef
24.
Zurück zum Zitat Yu R, Liu T, Jin SB, Ning C, Lendahl U, Nister M, Zhao J. MIEF1/2 function as adaptors to recruit Drp1 to mitochondria and regulate the association of Drp1 with Mff. Sci Rep. 2017;7(1):880.PubMedPubMedCentralCrossRef Yu R, Liu T, Jin SB, Ning C, Lendahl U, Nister M, Zhao J. MIEF1/2 function as adaptors to recruit Drp1 to mitochondria and regulate the association of Drp1 with Mff. Sci Rep. 2017;7(1):880.PubMedPubMedCentralCrossRef
25.
Zurück zum Zitat Zhou J, Zhang S, Li Z, Chen Z, Xu Y, Ye W, He Z. Yap-Hippo promotes A549 lung cancer cell death via modulating MIEF1-related mitochondrial stress and activating JNK pathway. Biomed Pharmacother. 2019;113:108754.PubMedCrossRef Zhou J, Zhang S, Li Z, Chen Z, Xu Y, Ye W, He Z. Yap-Hippo promotes A549 lung cancer cell death via modulating MIEF1-related mitochondrial stress and activating JNK pathway. Biomed Pharmacother. 2019;113:108754.PubMedCrossRef
26.
Zurück zum Zitat Fan T, Pi H, Li M, Ren Z, He Z, Zhu F, Tian L, Tu M, Xie J, Liu M, et al. Inhibiting MT2-TFE3-dependent autophagy enhances melatonin-induced apoptosis in tongue squamous cell carcinoma. J Pineal Res. 2018;64(2):e12457.CrossRef Fan T, Pi H, Li M, Ren Z, He Z, Zhu F, Tian L, Tu M, Xie J, Liu M, et al. Inhibiting MT2-TFE3-dependent autophagy enhances melatonin-induced apoptosis in tongue squamous cell carcinoma. J Pineal Res. 2018;64(2):e12457.CrossRef
27.
Zurück zum Zitat Fernandez Vazquez G, Reiter RJ, Agil A. Melatonin increases brown adipose tissue mass and function in Zucker diabetic fatty rats: implications for obesity control. J Pineal Res. 2018;64(4):e12472.PubMedCrossRef Fernandez Vazquez G, Reiter RJ, Agil A. Melatonin increases brown adipose tissue mass and function in Zucker diabetic fatty rats: implications for obesity control. J Pineal Res. 2018;64(4):e12472.PubMedCrossRef
28.
Zurück zum Zitat Chen T, Dai SH, Li X, Luo P, Zhu J, Wang YH, Fei Z, Jiang XF. Sirt1–Sirt3 axis regulates human blood–brain barrier permeability in response to ischemia. Redox Biol. 2018;14:229–36.PubMedCrossRef Chen T, Dai SH, Li X, Luo P, Zhu J, Wang YH, Fei Z, Jiang XF. Sirt1–Sirt3 axis regulates human blood–brain barrier permeability in response to ischemia. Redox Biol. 2018;14:229–36.PubMedCrossRef
29.
Zurück zum Zitat Li J, Cai SX, He Q, Zhang H, Friedberg D, Wang F, Redington AN. Intravenous miR-144 reduces left ventricular remodeling after myocardial infarction. Basic Res Cardiol. 2018;113(5):36.PubMedCrossRef Li J, Cai SX, He Q, Zhang H, Friedberg D, Wang F, Redington AN. Intravenous miR-144 reduces left ventricular remodeling after myocardial infarction. Basic Res Cardiol. 2018;113(5):36.PubMedCrossRef
30.
Zurück zum Zitat Sajib S, Zahra FT, Lionakis MS, German NA, Mikelis CM. Mechanisms of angiogenesis in microbe-regulated inflammatory and neoplastic conditions. Angiogenesis. 2018;21(1):1–14.PubMedCrossRef Sajib S, Zahra FT, Lionakis MS, German NA, Mikelis CM. Mechanisms of angiogenesis in microbe-regulated inflammatory and neoplastic conditions. Angiogenesis. 2018;21(1):1–14.PubMedCrossRef
32.
Zurück zum Zitat Souza LEB, Beckenkamp LR, Sobral LM, Fantacini DMC, Melo FUF, Borges JS, Leopoldino AM, Kashima S, Covas DT. Pre-culture in endothelial growth medium enhances the angiogenic properties of adipose-derived stem/stromal cells. Angiogenesis. 2018;21(1):15–22.PubMedCrossRef Souza LEB, Beckenkamp LR, Sobral LM, Fantacini DMC, Melo FUF, Borges JS, Leopoldino AM, Kashima S, Covas DT. Pre-culture in endothelial growth medium enhances the angiogenic properties of adipose-derived stem/stromal cells. Angiogenesis. 2018;21(1):15–22.PubMedCrossRef
33.
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.CrossRef 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.CrossRef
34.
Zurück zum Zitat Nawaz IM, Chiodelli P, Rezzola S, Paganini G, Corsini M, Lodola A, Di Ianni A, Mor M, Presta M. N-tert-butyloxycarbonyl-Phe-Leu-Phe-Leu-Phe (BOC2) inhibits the angiogenic activity of heparin-binding growth factors. Angiogenesis. 2018;21(1):47–59.PubMedCrossRef Nawaz IM, Chiodelli P, Rezzola S, Paganini G, Corsini M, Lodola A, Di Ianni A, Mor M, Presta M. N-tert-butyloxycarbonyl-Phe-Leu-Phe-Leu-Phe (BOC2) inhibits the angiogenic activity of heparin-binding growth factors. Angiogenesis. 2018;21(1):47–59.PubMedCrossRef
35.
Zurück zum Zitat Kiel AM, Goodwill AG, Baker HE, Dick GM, Tune JD. Local metabolic hypothesis is not sufficient to explain coronary autoregulatory behavior. Basic Res Cardiol. 2018;113(5):33.PubMedPubMedCentralCrossRef Kiel AM, Goodwill AG, Baker HE, Dick GM, Tune JD. Local metabolic hypothesis is not sufficient to explain coronary autoregulatory behavior. Basic Res Cardiol. 2018;113(5):33.PubMedPubMedCentralCrossRef
36.
Zurück zum Zitat Krause J, Loser A, Lemoine MD, Christ T, Scherschel K, Meyer C, Blankenberg S, Zeller T, Eschenhagen T, Stenzig J. Rat atrial engineered heart tissue: a new in vitro model to study atrial biology. Basic Res Cardiol. 2018;113(5):41.PubMedCrossRef Krause J, Loser A, Lemoine MD, Christ T, Scherschel K, Meyer C, Blankenberg S, Zeller T, Eschenhagen T, Stenzig J. Rat atrial engineered heart tissue: a new in vitro model to study atrial biology. Basic Res Cardiol. 2018;113(5):41.PubMedCrossRef
37.
Zurück zum Zitat Giatsidis G, Cheng L, Haddad A, Ji K, Succar J, Lancerotto L, Lujan-Hernandez J, Fiorina P, Matsumine H, Orgill DP. Noninvasive induction of angiogenesis in tissues by external suction: sequential optimization for use in reconstructive surgery. Angiogenesis. 2018;21(1):61–78.PubMedCrossRef Giatsidis G, Cheng L, Haddad A, Ji K, Succar J, Lancerotto L, Lujan-Hernandez J, Fiorina P, Matsumine H, Orgill DP. Noninvasive induction of angiogenesis in tissues by external suction: sequential optimization for use in reconstructive surgery. Angiogenesis. 2018;21(1):61–78.PubMedCrossRef
38.
Zurück zum Zitat Hardeland R. Melatonin and inflammation-story of a double-edged blade. J Pineal Res. 2018;65(4):e12525.PubMedCrossRef Hardeland R. Melatonin and inflammation-story of a double-edged blade. J Pineal Res. 2018;65(4):e12525.PubMedCrossRef
39.
Zurück zum Zitat Kanwar MK, Yu J, Zhou J. Phytomelatonin: recent advances and future prospects. J Pineal Res. 2018;65(4):e12526.PubMedCrossRef Kanwar MK, Yu J, Zhou J. Phytomelatonin: recent advances and future prospects. J Pineal Res. 2018;65(4):e12526.PubMedCrossRef
40.
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.PubMedCrossRef 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.PubMedCrossRef
41.
Zurück zum Zitat Jung M, Dodsworth M, Thum T. Inflammatory cells and their non-coding RNAs as targets for treating myocardial infarction. Basic Res Cardiol. 2018;114(1):4.PubMedPubMedCentralCrossRef Jung M, Dodsworth M, Thum T. Inflammatory cells and their non-coding RNAs as targets for treating myocardial infarction. Basic Res Cardiol. 2018;114(1):4.PubMedPubMedCentralCrossRef
42.
Zurück zum Zitat Hobson SR, Gurusinghe S, Lim R, Alers NO, Miller SL, Kingdom JC, Wallace EM. Melatonin improves endothelial function in vitro and prolongs pregnancy in women with early-onset preeclampsia. J Pineal Res. 2018;65(3):e12508.PubMedCrossRef Hobson SR, Gurusinghe S, Lim R, Alers NO, Miller SL, Kingdom JC, Wallace EM. Melatonin improves endothelial function in vitro and prolongs pregnancy in women with early-onset preeclampsia. J Pineal Res. 2018;65(3):e12508.PubMedCrossRef
43.
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.PubMedPubMedCentralCrossRef 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.PubMedPubMedCentralCrossRef
44.
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.PubMedPubMedCentralCrossRef 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.PubMedPubMedCentralCrossRef
45.
Zurück zum Zitat Kazakov A, Hall RA, Werner C, Meier T, Trouvain A, Rodionycheva S, Nickel A, Lammert F, Maack C, Bohm M, et al. Raf kinase inhibitor protein mediates myocardial fibrosis under conditions of enhanced myocardial oxidative stress. Basic Res Cardiol. 2018;113(6):42.PubMedPubMedCentralCrossRef Kazakov A, Hall RA, Werner C, Meier T, Trouvain A, Rodionycheva S, Nickel A, Lammert F, Maack C, Bohm M, et al. Raf kinase inhibitor protein mediates myocardial fibrosis under conditions of enhanced myocardial oxidative stress. Basic Res Cardiol. 2018;113(6):42.PubMedPubMedCentralCrossRef
46.
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.PubMedPubMedCentralCrossRef 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.PubMedPubMedCentralCrossRef
47.
Zurück zum Zitat Wang B, Yee Aw T, Stokes KY. N-acetylcysteine attenuates systemic platelet activation and cerebral vessel thrombosis in diabetes. Redox Biol. 2018;14:218–28.PubMedCrossRef Wang B, Yee Aw T, Stokes KY. N-acetylcysteine attenuates systemic platelet activation and cerebral vessel thrombosis in diabetes. Redox Biol. 2018;14:218–28.PubMedCrossRef
48.
Zurück zum Zitat Zhou H, Li N, Yuan Y, Jin YG, Guo H, Deng W, Tang QZ. Activating transcription factor 3 in cardiovascular diseases: a potential therapeutic target. Basic Res Cardiol. 2018;113(5):37.PubMedCrossRef Zhou H, Li N, Yuan Y, Jin YG, Guo H, Deng W, Tang QZ. Activating transcription factor 3 in cardiovascular diseases: a potential therapeutic target. Basic Res Cardiol. 2018;113(5):37.PubMedCrossRef
49.
Zurück zum Zitat Li H, Fu L, Liu B, Lin X, Dong Q, Wang E. Ajuba overexpression regulates mitochondrial potential and glucose uptake through YAP/Bcl-xL/GLUT1 in human gastric cancer. Gene. 2019;693:16–24.PubMedCrossRef Li H, Fu L, Liu B, Lin X, Dong Q, Wang E. Ajuba overexpression regulates mitochondrial potential and glucose uptake through YAP/Bcl-xL/GLUT1 in human gastric cancer. Gene. 2019;693:16–24.PubMedCrossRef
51.
Zurück zum Zitat Merz J, Albrecht P, von Garlen S, Ahmed I, Dimanski D, Wolf D, Hilgendorf I, Hardtner C, Grotius K, Willecke F, et al. Purinergic receptor Y2 (P2Y2)-dependent VCAM-1 expression promotes immune cell infiltration in metabolic syndrome. Basic Res Cardiol. 2018;113(6):45.PubMedCrossRef Merz J, Albrecht P, von Garlen S, Ahmed I, Dimanski D, Wolf D, Hilgendorf I, Hardtner C, Grotius K, Willecke F, et al. Purinergic receptor Y2 (P2Y2)-dependent VCAM-1 expression promotes immune cell infiltration in metabolic syndrome. Basic Res Cardiol. 2018;113(6):45.PubMedCrossRef
52.
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.PubMedCrossRef 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.PubMedCrossRef
53.
Zurück zum Zitat Li X, Brestic M, Tan DX, Zivcak M, Zhu X, Liu S, Song F, Reiter RJ, Liu F. Melatonin alleviates low PS I-limited carbon assimilation under elevated CO2 and enhances the cold tolerance of offspring in chlorophyll b-deficient mutant wheat. J Pineal Res. 2018;64(1):e12453.CrossRef Li X, Brestic M, Tan DX, Zivcak M, Zhu X, Liu S, Song F, Reiter RJ, Liu F. Melatonin alleviates low PS I-limited carbon assimilation under elevated CO2 and enhances the cold tolerance of offspring in chlorophyll b-deficient mutant wheat. J Pineal Res. 2018;64(1):e12453.CrossRef
54.
Zurück zum Zitat Olson KR, Gao Y, Arif F, Arora K, Patel S, DeLeon ER, Sutton TR, Feelisch M, Cortese-Krott MM, Straub KD. Metabolism of hydrogen sulfide (H2S) and production of reactive sulfur species (RSS) by superoxide dismutase. Redox Biol. 2018;15:74–85.PubMedCrossRef Olson KR, Gao Y, Arif F, Arora K, Patel S, DeLeon ER, Sutton TR, Feelisch M, Cortese-Krott MM, Straub KD. Metabolism of hydrogen sulfide (H2S) and production of reactive sulfur species (RSS) by superoxide dismutase. Redox Biol. 2018;15:74–85.PubMedCrossRef
55.
Zurück zum Zitat Lu C, Chen X, Wang Q, Xu X, Xu B. TNFalpha promotes glioblastoma A172 cell mitochondrial apoptosis via augmenting mitochondrial fission and repression of MAPK-ERK-YAP signaling pathways. Onco Targets Ther. 2018;11:7213–27.PubMedPubMedCentralCrossRef Lu C, Chen X, Wang Q, Xu X, Xu B. TNFalpha promotes glioblastoma A172 cell mitochondrial apoptosis via augmenting mitochondrial fission and repression of MAPK-ERK-YAP signaling pathways. Onco Targets Ther. 2018;11:7213–27.PubMedPubMedCentralCrossRef
56.
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.PubMedPubMedCentralCrossRef 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.PubMedPubMedCentralCrossRef
57.
Zurück zum Zitat Shi C, Zhou H, Li X, Cai Y. A retrospective analysis on two-week short-course pre-operative radiotherapy in elderly patients with resectable locally advanced rectal cancer. Sci Rep. 2016;6:37866.PubMedPubMedCentralCrossRef Shi C, Zhou H, Li X, Cai Y. A retrospective analysis on two-week short-course pre-operative radiotherapy in elderly patients with resectable locally advanced rectal cancer. Sci Rep. 2016;6:37866.PubMedPubMedCentralCrossRef
58.
Zurück zum Zitat Oh SE, Mouradian MM. Cytoprotective mechanisms of DJ-1 against oxidative stress through modulating ERK1/2 and ASK1 signal transduction. Redox Biol. 2018;14:211–7.PubMedCrossRef Oh SE, Mouradian MM. Cytoprotective mechanisms of DJ-1 against oxidative stress through modulating ERK1/2 and ASK1 signal transduction. Redox Biol. 2018;14:211–7.PubMedCrossRef
59.
Zurück zum Zitat Ding M, Ning J, Feng N, Li Z, Liu Z, Wang Y, Wang Y, Li X, Huo C, Jia X, et al. Dynamin-related protein 1-mediated mitochondrial fission contributes to post-traumatic cardiac dysfunction in rats and the protective effect of melatonin. J Pineal Res. 2018;64(1):e12447.CrossRef Ding M, Ning J, Feng N, Li Z, Liu Z, Wang Y, Wang Y, Li X, Huo C, Jia X, et al. Dynamin-related protein 1-mediated mitochondrial fission contributes to post-traumatic cardiac dysfunction in rats and the protective effect of melatonin. J Pineal Res. 2018;64(1):e12447.CrossRef
60.
Zurück zum Zitat Park HJ, Park JY, Kim JW, Yang SG, Jung JM, Kim MJ, Kang MJ, Cho YH, Wee G, Yang HY, et al. Melatonin improves the meiotic maturation of porcine oocytes by reducing endoplasmic reticulum stress during in vitro maturation. J Pineal Res. 2018;64(2):e12458.PubMedCrossRef Park HJ, Park JY, Kim JW, Yang SG, Jung JM, Kim MJ, Kang MJ, Cho YH, Wee G, Yang HY, et al. Melatonin improves the meiotic maturation of porcine oocytes by reducing endoplasmic reticulum stress during in vitro maturation. J Pineal Res. 2018;64(2):e12458.PubMedCrossRef
61.
Zurück zum Zitat Alessio N, Del Gaudio S, Capasso S, Di Bernardo G, Cappabianca S, Cipollaro M, Peluso G, Galderisi U. Low dose radiation induced senescence of human mesenchymal stromal cells and impaired the autophagy process. Oncotarget. 2015;6(10):8155–66.PubMedCrossRef Alessio N, Del Gaudio S, Capasso S, Di Bernardo G, Cappabianca S, Cipollaro M, Peluso G, Galderisi U. Low dose radiation induced senescence of human mesenchymal stromal cells and impaired the autophagy process. Oncotarget. 2015;6(10):8155–66.PubMedCrossRef
Metadaten
Titel
RETRACTED ARTICLE: Mst1 overexpression combined with Yap knockdown augments thyroid carcinoma apoptosis via promoting MIEF1-related mitochondrial fission and activating the JNK pathway
verfasst von
Xiaoli Zhang
Fei Li
Yeqing Cui
Shuang Liu
Haichen Sun
Publikationsdatum
01.12.2019
Verlag
BioMed Central
Erschienen in
Cancer Cell International / Ausgabe 1/2019
Elektronische ISSN: 1475-2867
DOI
https://doi.org/10.1186/s12935-019-0860-8

Weitere Artikel der Ausgabe 1/2019

Cancer Cell International 1/2019 Zur Ausgabe

Update Onkologie

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