Skip to main content
Erschienen in: Journal of Experimental & Clinical Cancer Research 1/2020

Open Access 01.12.2020 | Research

Simvastatin re-sensitizes hepatocellular carcinoma cells to sorafenib by inhibiting HIF-1α/PPAR-γ/PKM2-mediated glycolysis

verfasst von: Jiao Feng, Weiqi Dai, Yuqing Mao, Liwei Wu, Jingjing Li, Kan Chen, Qiang Yu, Rui Kong, Sainan Li, Jie Zhang, Jie Ji, Jianye Wu, Wenhui Mo, Xuanfu Xu, Chuanyong Guo

Erschienen in: Journal of Experimental & Clinical Cancer Research | Ausgabe 1/2020

Abstract

Background

Hepatocellular carcinoma (HCC) is a common primary malignant tumor which usually progresses to an advanced stage because of late diagnosis. Sorafenib (Sora) is a first line medicine for advanced stage HCC; however, it has been faced with enormous resistance. Simvastatin (Sim) is a cholesterol-lowering drug and has been reported to inhibit tumor growth. The present study aims to determine whether Sora and Sim co-treatment can improve Sora resistance in HCC.

Methods

The HCC cell line LM3 and an established Sora-resistant LM3 cell line (LM3-SR) were used to study the relationship between Sora resistance and aerobic glycolysis. Cell proliferation, apoptosis and glycolysis levels were analyzed by western blotting, flow cytometry analysis and biomedical tests. A xenograft model was also used to examine the effect of Sim in vivo. Detailed mechanistic studies were also undertaken by the use of activators and inhibitors, and lentivirus transfections.

Results

Our results demonstrated that the resistance to Sora was associated with enhanced aerobic glycolysis levels. Furthermore, LM3-SR cells were more sensitive to Sim than LM3 cells, suggesting that combined treatment with both Sora and Sim could enhance the sensitivity of LM3-SR cells to Sora. This finding may be due to the suppression of the HIF-1α/PPAR-γ/PKM2 axis.

Conclusions

Simvastatin can inhibit the HIF-1α/PPAR-γ/PKM2 axis, by suppressing PKM2-mediated glycolysis, resulting in decreased proliferation and increased apoptosis in HCC cells, and re-sensitizing HCC cells to Sora.
Hinweise

Supplementary information

Supplementary information accompanies this paper at https://​doi.​org/​10.​1186/​s13046-020-1528-x.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
2-DG
2-Deoxyglucose
CCK-8
Cell counting kit
CI
Combination index
Co-IP
Co-immunoprecipitation
CTRL
Control
DRI
Dose reduction index
EGFR
Epidermal growth factor receptor
EMT
Epithelial-mesenchymal transition
Fa-CI
Fraction affected-combination index
GLUT
Glucose transporter
H&E
Hematoxylin and eosin
HCC
Hepatocellular carcinoma
HDL-C
High-density lipoprotein cholesterol
HIF-1α
Hypoxia inducible factor-1α
HK2
Hexokinase 2
HMG CoA
Hydroxymethylglutaryl coenzyme A
IHC
Immunohistochemistry
LDL-C
Low-density lipoprotein cholesterol
LM3-SR
Sora-resistant LM3 cell line
OXPHOS
Oxidative phosphorylation
PFK1
Phosphofructokinase 1
PKM2
Pyruvate kinase, type M2
PPAR-γ
Peroxisome proliferator–activated receptor γ
qRT-PCR
Quantitative reverse transcription-polymerase chain reaction
SD
Standard deviation
Sim
Simvastatin
Sora
Sorafenib
TCHO
Total cholesterol
TG
Triglyceride
TUNEL
Terminal deoxynucleotidyl transferase dUTP nick end labeling

Background

Hepatocellular carcinoma (HCC) is one of the most common primary malignant tumors worldwide and according to global cancer statistics (2018), liver cancer is the fourth leading cause of cancer deaths [13]. The pathogenesis of HCC is associated with chronic viral hepatitis infection, alcohol abuse, and aflatoxin B1 intake [4]. The standard therapeutic methods for the treatment of HCC include surgical resection, trans-arterial embolization, radiotherapy and chemotherapy. However, these treatments are often inadequate because of delays in diagnosis and metastasis, resulting in advanced HCC [5].
Sorafenib (Sora) is a multikinase inhibitor and has been approved as the first-line targeted therapy for advanced HCC [2, 6]. In two phase III trials, results showed that Sora could prolong the overall survival of HCC patients by 2–3 months. However, only 30% of patients benefitted from Sora because of acquired resistance which occurred within 6 months [7, 8]. The mechanisms of Sora resistant are complex and undefined, but include increased epidermal growth factor receptor (EGFR) expression, c-Jun and Akt activation of HCC cells, epithelial-mesenchymal transition (EMT), increased cancer stem cells, and an increase in the hypoxic environment [2, 5]. Recently, researchers have found that Sora can impair oxidative phosphorylation (OXPHOS) and promote aerobic glycolysis in HCC [9, 10]. Although aerobic glycolysis is a hallmark of cancer, few studies have attempted to elucidate the relationship between aerobic glycolysis and Sora resistance.
Currently, strategies to prevent Sora resistance include co-treatment with other medicines in clinical use, including agents that target specific molecules such as anti-EGFR antibodies, (Cetuximab), cytotoxic chemotherapeutic drugs (Epirubicin, Cisplatin, 5-FU and Capecitabine), and immunotherapeutic drugs (anti-PD-1 antibodies) [2, 11, 12]. However, this combinational therapy is usually limited due to severe adverse side-effects, such as diarrhea, and organ damage [11]. Therefore, a safer agent is needed to overcome or improve Sora resistance in HCC.
Simvastatin is a cholesterol-lowering statin, which can inhibit the activity of hydroxymethylglutaryl coenzyme A (HMG CoA) reductase and prevent the synthesis of cholesterol. Recently, many studies have demonstrated that statins also have additional benefits, including antioxidant, anti-proliferative, and anti-inflammatory and can function to protect the vascular endothelium [1315]. Statins also play a role in the prevention of liver diseases, including non-alcoholic fatty liver disease, cholestatic liver disease, and liver cirrhosis [16, 17]. Moreover, statins always exhibit synergistic effects when combined with other chemotherapeutic drugs [18]. For example, fluvastatin has been reported to increase the cytotoxicity of Sora in melanoma cells [19]. Kim et al. found that co-treatment with lovastatin and enzastaurin, both PKC inhibitors, synergistically inhibited HCC cell growth in vitro and in vivo [20]. Some researchers have reported that the combination of celecoxib or NS 398 and statins enhanced the inhibition of HCC growth [21, 22]. However, there are few studies investigating the combined treatment of Sim and Sora to treat Sora-resistant-HCC, and the effect on aerobic glycolysis.
Therefore, in the present study, we combined Sora with Sim to determine a role for Sim in the treatment of Sora resistance in HCC, and to explore the potential mechanisms.

Materials and methods

Reagents

Sorafenib tosylate, FG-4592, BAY87–2243, Rogislitazone, GW9662, Compound 3 k and DASA 58 were purchased from Selleck Chemicals (Shanghai, China). Sim was purchased from Yuanye Biotechnology (Shanghai, China). The cell counting kit (CCK-8) and the nuclear and cytoplasmic protein extraction kit were obtained from Yeasen Biotechnology (Shanghai, China), the Hoechst 33342 fluorescence staining kit was from Servicebio (Wuhan, China), the Annexin V-FITC apoptosis assay kit was from BD Biosciences (San Jose, CA, USA), and the primary antibodies used in the study are listed in Table 1.
Table 1
The primary antibodies used in the study
Antibody
Species
Targeted species
Dilution ratio
Supplier
Catalogue number
β-actin
M
H, M, R
1:1000
CST
3700
PCNA
Rbt
H, M, R
1:2000
PT
10,205–2-AP
Bax
M
H, M, R
1:1000
PT
60,267–1-Ig
Bcl-2
Rbt
H
1:1000
CST
15,071
Caspase 3
Rbt
H, M, R
1:1000
PT
19,677–1-AP
HK2
Rbt
H, M, R
1:1000
PT
22,029–1-AP
PFKL (PFK1)
Rbt
H, M
1:1000
CST
8175
PKM2
M
H, M, R
1:1000
PT
60,268–1-Ig
OXPHOS
M
H, M, R
1:250
MT
MS604
HIF-1α
Rbt
H
1:1000
PT
20,960–1-AP
PPAR-γ
Rbt
H, M, R
1:1000
ABclonal
60,318–1-Ig
Cleaved PARP
Rbt
H
1:1000
CST
5625S
Cleaved PARP
M
M
1:1000
CST
9548S
Ki-67
Rbt
M, R
1:1000
Servicebio
GB111141
LaminA/C
Rbt
H, M, R
1:1000
PT
10,298–1-AP
β-Tubulin
Rbt
H, M, R
1:1000
PT
10,094–1-AP
Abbreviations for the table: H human; M mouse; Rbt rabbit; R rat; CST Cell Signaling Technology (Danvers, MA, USA). PT Proteintech (Chicago, IL, USA). ABclonal Biotechnology (Wuhan, China). MS Mitoscience (St. Louis Park, MN, USA)

Cell culture

Four different HCC cell lines, including HCC-LM3, SMMC-7721, Bel-7402, and Huh-, a hepatoblastoma cell line HepG2 [23], and the LO2 normal human liver cell line were purchased from the Cell Bank of Type Culture Collection of the Chinese Academy of Sciences (Shanghai, China), and maintained in high glucose Dulbecco’s Modified Eagle Medium (DMEM HyClone, GE Healthcare, Logan, UT, USA) supplemented with 10% fetal bovine serum, 100 U/mL of penicillin, and 100 g/mL of streptomycin (all from Gibco, Thermo Fisher Scientific, Waltham, MA, USA).

Establishment of SORA-resistant LM3 cells

The establishment of SORA-resistant LM3 cells (LM3-SR) was conducted according to previous studies [24, 25]. Briefly, LM3 cells were cultured in a step-wise increase in Sora concentration (4–10 μM), by 10% every two weeks until the maximum tolerated dose (10 μM) had been reached. LM3-SR cells were cultured in the presence of 1 μM Sora, which was withdrawal for three days before analysis.

CCK8 assay, quantitative reverse transcription-polymerase chain reaction (qRT-PCR) and western blotting

The primers used in the study were synthesized by Generay Biotech (Shanghai, China), and their sequences listed in Table 2. The PrimeScript RT Reagent kit and SYBR Premix Ex Taq were purchased from TaKaRa Biotechnology (Dalian, China). CCK8 assay, quantitative RT-PCR (qRT-PCR), and western blotting were conducted as described previously [2628]. The effects of different drugs were determined using CCK8 assay. Therefore, Sora at a concentration of 15 μM and Sim at 10 μM or 50 μM were used in the following studies where treatment was given for 24 h.
Table 2
Primers used for qPCR
Gene name
Forward (5′-3′)
Reverse (5′-3′)
PKM2
ATGTCGAAGCCCCATAGTGAA
TGGGTGGTGAATCAATGTCCA
HK2
GAGCCACCACTCACCCTACT
CCAGGCATTCGGCAATGTG
PFKFB1
AGAAGGGGCTCATCCATACCC
CTCTCGTCGATACTGGCCTAA
PFKFB2
TGGGCCTCCTACATGACCAA
CAGTTGAGGTAGCGTGTTAGTTT
PFKFB3
TTGGCGTCCCCACAAAAGT
AGTTGTAGGAGCTGTACTGCTT
PFKFB4
TCCCCACGGGAATTGACAC
GGGCACACCAATCCAGTTCA
LDH-A
ATGGCAACTCTAAAGGATCAGC
CCAACCCCAACAACTGTAATCT
LDH-B
TGGTATGGCGTGTGCTATCAG
TTGGCGGTCACAGAATAATCTTT
LDH-C
AGAACATGGTGATTCTAGTGTGC
ACAGTCCAATAGCCCAAGAGG
HIF-1α
GAACGTCGAAAAGAAAAGTCTCG
CCTTATCAAGATGCGAACTCACA
AMPK-α1
TTGAAACCTGAAAATGTCCTGCT
GGTGAGCCACAACTTGTTCTT
AMPK-α2
GTGAAGATCGGACACTACGTG
CTGCCACTTTATGGCCTGTTA
AMPK-β1
CCACTCCGAGGAAATCAAGGC
CTGGGCGGGAGCTTTATCA
GLUT1
GGCCAAGAGTGTGCTAAAGAA
ACAGCGTTGATGCCAGACAG
β-actin
CATGTACGTTGCTATCCAGGC
CTCCTTAATGTCACGCACGAT
PGC1
TCTGAGTCTGTATGGAGTGACAT
CCAAGTCGTTCACATCTAGTTCA
PPRC1
CAAGCGCCGTATGGGACTTT
GGAGGCATCCATGTAGCTCT
PPAR-α
ATGGTGGACACGGAAAGCC
CGATGGATTGCGAAATCTCTTGG
PPAR-γ
GGGATCAGCTCCGTGGATCT
TGCACTTTGGTACTCTTGAAGTT

Standard colony formation, Hoechst 33342 staining, immunofluorescence staining and flow cytometry analysis for apoptosis

Standard colony formation, Hoechst 33342 staining, immunofluorescence staining and flow cytometry analysis for apoptosis were conducted as described previously [29]. The flow cytometry used in the study was FACSCalibur (Becton, Dickinson, Franklin Lakes, NJ, USA), and analyzed by FlowJo software (version 10; FlowJo LLC, Ashland, OR, USA). All the images were captured using Leica inverted fluorescence microscope DMI6000B (Leica Microsystems, Wetzlar, Germany).

Biomedical analysis

Glycolysis levels were determined using the detection of lactate production and glucose uptake levels in LM3 or LM3-SR cells. The lactate assay kit was obtained from Nanjing Jiancheng Bioengineering Institute (Nanjing, China). Glucose uptake levels were calculated using a glucose detection kit from Rongsheng Biotechnology (Shanghai, China), and the values were normalized to the protein concentrations of the cell lysates [10, 30]. The triglyceride (TG), total cholesterol (TCHO) low-density lipoprotein cholesterol (LDL-C) and the high-density lipoprotein cholesterol (HDL-C) assay kits were all purchased from Nanjing Jiancheng Bioengineering Institute (Nanjing, China). All the experiments were carried out according to the manufacturer’s instructions.

Plasmid construction, lentivirus packaging, and cell transfection

The lentivirus overexpression or knock down construct for PKM2 was synthesized by BioLink Biotechnology (Shanghai, China). All plasmids used in this study were verified by sequencing. Lentiviral transfection was conducted following the manufacturer’s instructions. The positive cells were selected by puromycin resistance and transfection efficiency was determined by qPCR and western blotting.

Co-immunoprecipitation (co-IP) assay

The Co-IP assay was carried out using the Pierce Co-immunoprecipitation Kit (Thermo Scientific, Waltham, MA, USA) and according to manufacturer’s protocol, 1 mg of total protein lysate was mixed with 10 μg of primary antibody, or IgG. The results were analyzed by western blotting [29].

Subcutaneous xenograft tumor model

Four-week-old nude mice were obtained from Shanghai Slack Laboratory Animal Co. LTD (Shanghai, China), and housed in a standard laboratory environment with free access to water and food. The study was approved by the Animal Care and Use Committee of Shanghai Tongji University, and conducted following the ARRIVE guidelines.
For the establishment of the subcutaneous xenograft tumor model, LM3 or LM3-SR cells were resuspended to a density of 3 × 106/mL in serum-free DMEM and each mouse injected with 200 μL of cells in the upper flank region. Tumor volume was calculated as: volume (mm3) = 0.5 × (major axis) × (minor axis)2, and when it reached 100 mm3, mice received Sora (10 mg/kg), Sim (10 mg/kg) or a co-treatment with Sora (10 mg/kg) + Sim (10 mg/kg) orally once daily until the end of the study. At day 28 post cell injection, mice were anesthetized with 1.25% pentobarbital, and blood samples were collected by removing the eyes, and the tumors, as well as heart, kidney and lung were resected and immersed in 4% paraformaldehyde.
The following study groups were used: (1) for the analysis of the LM3-SR cell-induced xenograft, 12 mice were randomly divided into four groups (n = 3): 1) LM3-SR-Control (CTRL) group, mice were injected with LM3-SR cells in the absence of Sora; 2) LM3-SR-Sora group, mice were injected with LM3-SR cells and received Sora (10 mg/kg) administration; 3) LM3-Control (CTRL) group, mice were injected with LM3 cells but with no Sora administration; 4) LM3-Sora group, mice were injected with LM3 cells and received Sora (10 mg/kg) administration. (2) For the analysis of Sora and Sim co-treatment, 16 mice were randomly divided into four groups (n = 4): 1) CTRL group, mice were injected with LM3-SR cells alone; 2) Sora group, mice were injected with LM3-SR cells and received Sora (10 mg/kg) administration; 3) Sim group, mice were injected with LM3-SR cells and received Sim (10 mg/kg) administration; 4) Sora + Sim group, mice were injected with LM3-SR cells and received both Sora (10 mg/kg) + Sim (10 mg/kg).

Hematoxylin and eosin (H&E) staining, immunohistochemistry (IHC) staining, and terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL)

Animal tissues were immersed in 4% paraformaldehyde for at least 24 h, then embedded in paraffin and cut into 3 μm thick sections. The H&E, IHC and TUNEL staining was carried out as described previously [31, 32].

Statistical analysis

Data were presented as mean ± standard deviation (SD) from three independent experiments. After the statistical analysis was carried out the results were imaged using GraphPad Prism 6 software (GraphPad Software, San Diego, CA, USA). The comparisons between two groups were analyzed by Student’s t-tests (unpaired, two-tailed) or the one-way analysis of variance (ANOVA) (followed by Tukey’s post-hoc tests). P < 0.05 was considered statistically significant. All data generated and analyzed during this study are available on request from the corresponding author.

Results

The establishment and characteristics of Sora-resistant cells

Both the primary and the secondary resistance to Sora have restricted the application and treatment effects of this drug in the clinic. Therefore, four types of HCC cell lines, including HCC-LM3 (shorted to LM3), SMMC-7721, Bel-7402 and Huh-7, and a hepatoblastoma cell line HepG2 were used to detect the half inhibitory concentration (IC50) of Sora in this study. As the results show in Fig. 1a, different concentrations of Sora were given for 24 h, and the cell viability was calculated using the CCK8 assay. The IC50s for Sora in these cell lines were 4.47 μM (LM3), 8.79 μM (SMMC-7721), 8.98 μM (Bel-7402), 4.65 μM (HepG2), and 7.26 μM (Huh-7), respectively. Since the IC50 of LM3 was the lowest among the five liver cancer cell lines, which meant that LM3 was the most sensitive to Sora. Therefore, LM3 cells were chosen as Sora-sensitive cells, and we then established the Sora-resistant LM3 cells line (LM3-SR) as control to explore the underlying mechanism in Sora resistance [9, 33, 34]. The IC50 of LM3-SR was 16.33 μM (Fig. 1a), which is approximately four-fold higher than LM3 cells. This implied the successful establishment of the Sora-resistant LM3 cells. Therefore, the LM3 and LM3-SR cells were used in the following study to represent the Sora-sensitive and Sora-resistant cells, at a Sora concentration of 15 μM.
The different characteristics seen between LM3 and LM3-SR cells were detected using proliferation assays and flow cytometry. As shown in Fig. 1b, 15 μM Sora clearly prevented colony formation in LM3 cells and this effect was reduced in LM3-SR cells, indicating that the ability to proliferate in LM3-SR cells was higher than in LM3 cells. Hoechst 33342 staining revealed that there were more positive cells (representing apoptotic cells) in the Sora treated group of LM3 cells, when compared to LM3-SR cells (Fig. 1c). Furthermore, flow cytometry (Fig. 1d) demonstrated that the apoptosis rate in LM3-SR cells when exposed to Sora was much lower than that seen in LM3 cells (32.65 ± 2.37% vs. 8.63 ± 2.62%). These findings suggest that it was difficult for Sora (15 μM) to induce apoptosis in LM3-SR cells. These results suggest that LM3-SR cells were resistant to Sora and were less likely to experience inhibition of proliferation or apoptosis.

Resistance to Sora is associated with enhanced aerobic glycolysis

Aerobic glycolysis is a hallmark of tumor cell metabolism, and enhanced aerobic glycolysis is characterized by high glucose uptake and high lactate production. Some studies have reported that long-term use of Sora could lead to increased aerobic glycolysis, and this may be associated with Sora resistance [9]. Therefore, in our study, glycolysis levels of LM3-SR cells were measured. The results presented in Fig. 2a show that both glucose uptake and lactate production were higher in LM3-SR cells when compared to LM3 cells, suggesting that LM3-SR cells produced higher glycolysis levels. Moreover, Sora (15 μM) was effective at inhibiting both glucose uptake and lactate production in LM3 cells; whereas Sora (15 μM) produced a slight decrease in LM3-SR cells. The three key enzymes involved in glycolysis, including hexokinase 2 (HK2), phosphofructokinase 1(PFK1) and pyruvate kinase, type M2 (PKM2), and OXPHOS were determined by western blotting (Fig. 2b). The results were similar to those seen in Fig. 2a, demonstrating that LM3-SR cells had higher expression levels of glycolysis-associated proteins and impaired OXPHOS associated proteins. The proliferation indicator PCNA, as wells as the apoptosis markers Bcl-2, Bax, caspase 3 and cleaved PARP were also detected, and the results demonstrated that Sora (15 μM) had a limited effect on inhibiting proliferation, glycolysis, and induction of apoptosis in LM3-SR cells. These findings indicate that Sora resistance was associated with enhanced glycolysis. Since Bel-7402 maybe a naïve Sora-resistant HCC cell line when compared to LM3, the mechanism underlying Sora resistance was also explored in Bel-7402 cells (Additional file 1: Figure S1).
To further explore the Sora-resistant effect in HCC cells in vivo, both LM3 and LM3-SR cells were seeded into nude mice to produce a xenografted tumor model (Fig. 2c). The results showed that LM3-SR cells could induce a larger tumor volume than LM3 cells, and Sora (10 mg/kg) was unable to suppress tumor volume in the LM3-SR groups as well as in the LM3 groups. The H&E, TUNEL and IHC staining of Ki-67 results showed that there were less necrotic (pink areas) and apoptotic areas (the nucleus was stained dark-brown), but more tumor parenchyma and higher Ki-67 staining in the LM3-SR groups (Fig. 2d), as well as a reduced treatment effect of Sora (10 mg/kg). In conclusion, these results show that the resistance to Sora in LM3-SR cells is associated with enhanced aerobic glycolysis levels, and this may result in the decreased effect of treatment seen in vivo.

LM3-SR cells have a greater sensitivity to Sim when compared to LM3 cells

Sim is a commonly used drug to lower blood lipid levels. Recently, anti-fibrotic and anti-cancer effects of Sim have been found. In our study, the effect of Sim on LM3, LM3-SR and normal liver cells LO2 were determined using CCK8 assay (Fig. 3a). Surprisingly, the IC50 of Sim in these cells were 55.99 μM (LM3), 14.93 μM (LM3-SR), and 74.92 μM (LO2) respectively, indicating that LM3-SR cells were 3.75 fold more sensitive to Sim than LM3 and LO2. Furthermore, Sim (10 μM) was able to kill LM3-SR cells without influencing the viability of normal liver cells (LO2). Therefore, two concentrations of Sim (10 μM and 50 μM) were used to explore their effects on apoptosis and glycolysis in LM3 and LM3-SR cells. Consistently, Sim (10 μM) did not significantly induced LM3 cell apoptosis, but apoptosis was higher in LM3-SR cells and the high dose of Sim (50 μM) was able to induce apoptosis in both LM3 and LM3-SR cells (Fig. 3b-c). The results in Fig. 3d show that Sim (10 μM) failed to decrease lactate production or glucose uptake in LM3 cells, whereas Sim (50 μM) was able to produce both affects. The two concentrations of Sim (10 μM and 50 μM) were able to reduce lactate production and glucose uptake in LM3-SR cells. Western blotting for PCNA, Bcl-2, Bax, Caspase 3, cleaved PARP, glycolysis-related enzymes and OXPOHS further verified the effect of Sim (Fig. 3e). Therefore, these results demonstrated that LM3-SR cells are more sensitive to Sim than LM3 cells, and Sim (10 μM) was effective at reducing proliferation, glycolysis and inducing apoptosis in LM3-SR cells.

Sim enhanced the sensitivity of LM3-SR cells to Sora when used in combination

Since LM3-SR cells were more sensitive to Sim, we combined Sora (15 μM) with Sim (10 μM, IC50 1:1 combination) to determine whether Sim can enhance the sensitivity of LM3-SR cells to Sora. In Fig. 4a, different doses of Sora and Sim were combined at a constant ratio, and the effects analyzed by CKK8 assay. The fraction affected-combination index (Fa-CI) plot shows that the CI calculated for Sora (15 μM) and Sim (10 μM) was 0.722 (CI < 1), indicating a synergistic effect. The dose reduction index (DRI) was also calculated, and we found that Sim (10 μM) was able to reduce the dose of Sora by 2.843-fold. The apoptosis assays also reflected that both Sora + Sim treatment enhanced the apoptosis rate of LM3-SR cells (Fig. 4b-c). Lactate production and glucose uptake levels with both Sora + Sim was also lower than Sora (15 μM) or Sim (10 μM) treatment alone (Fig. 4d), again inferring a synergic effect on the suppression of glycolysis by Sora + Sim co-treatment. Western blotting analysis of PCNA, Bcl-2, Bax, Caspase 3, cleaved PARP, glycolysis-related enzymes and OXPOHS further confirmed a synergistic effect of Sora + Sim co-treatment (Fig. 4e). In addition, we also explored the co-treatment of Sora (5 μM) and Sim (10 μM) in LM3 cells, and the synergic effects were also observed in LM3 cell (Additional file 2: Figure S2).
We also explored the co-treatment of Sora + Sim on HCC cells in vivo. As show in Fig. 4f, the tumor volume in the Sora + Sim treated group was much smaller than when Sora or Sim were treated alone. H&E and TUNEL staining demonstrated that there were more necrotic and apoptotic areas in the Sora + Sim group. Furthermore, we also examined the pathological manifestations of important organs to determine whether Sim was detrimental to organ function. The H&E staining seen in Fig. 4h demonstrated that Sim did not harm the liver, lungs, kidneys or heart. Since Sim was used as a lipid-lowering drug, serum lipid levels were also detected. The results in Fig. 4i revealed that Sim could lower TG, TCHO and LDL-C levels, but increased HDL-L levels in nude mice; whereas Sora + Sim co-treatment could also lower serum levels, indicating that Sim was beneficial to liver function and blood lipid levels in HCC. In conclusion, these results show that Sora + Sim co-treatment could enhance the effect (sensitivity) of Sora on HCC by promoting apoptosis and suppressing glycolysis.

Sim enhances Sora sensitivity by inhibiting PKM2, HIF-1α and PPAR-γ

We next examined glycolysis, glucose and fatty acid metabolism-related genes by qPCR (Fig. 5a). Among 18 potential genes, we found that Sora + Sim co-treatment decreased the transcription of PKM2, hypoxia inducible factor-1α (HIF-1α) and peroxisome proliferator–activated receptor γ (PPAR-γ) the most. Western blotting analysis of PKM2, HIF-1α and PPAR-γ also showed a trend towards decreased protein levels in the Sora + Sim group. As seen in Fig. 5a, Sim activated the transcription of PPAR-α, the protein expression of PPAR-α was also detected by western blotting. However, the results seen in Fig. 5b show that the co-treatment with Sora and Sim did not inhibit PPAR-α expression to the same extent as Sora alone. Immunofluorescence staining further showed that the fluorescence intensity of PKM2, HIF-1α and PPAR-γ were reduced by Sora + Sim co-treatment (Fig. 5c). Moreover, from the localization of PKM2, we see that Sora + Sim co-treatment does not only inhibit the total expression of PKM2 (Fig. 5b-c), but also specifically inhibits its nuclear expression (Fig. 5c). These results were verified using a cytoplasm-nuclear protein extraction kit (Fig. 5d). Besides, the results in Fig. 5d showed that most of HIF-1α and PPAR-γ were localized in the nucleus, and Sim treatment can inhibit the expression of both HIF-1α and PPAR-γ in the nucleus. Given that PKM2, HIF-1α and PPAR-γ can localize to the nucleus, we conducted a Co-IP assay to determine their interaction. The results presented in Fig. 5e demonstrated that both HIF-1α and PPAR-γ can be pulled-down by PKM2, indicating that both HIF-1α and PPAR-γ can interact with PKM2 in the nucleus. Based on this finding, we concluded that Sim enhanced the sensitivity of Sora by inhibiting the expression and interaction of PKM2, HIF-1α and PPAR-γ.

Sim enhances the sensitivity of Sora by down-regulating the HIF-1α/PPAR-γ/PKM2 axis

Activators and inhibitors of the HIF-1α/PPAR-γ/PKM2 axis were used to explore the involvement of HIF-1α, PPAR-γ and PKM2 on the effects seen with Sim. As shown in Fig. 6a, after FG-4592 treatment (50 μM) [35, 36], the expression of HIF-1α, PPAR-γ and PKM2 were all up-regulated; however, in the Sora + Sim group, the up-regulation after FG-4592 treatment was reversed. However, after application of the HIF-1α inhibitor BAY87–2243 (10 mM) [37], the expression of HIF-1α, PPAR-γ and PKM2 were all down-regulated. Furthermore, after co-treated with Sora + Sim, their inhibitory effect were overlaid. The nuclear expression of HIF-1α, PPAR-γ and PKM2 was also detected using a nuclear and cytoplasmic protein extraction kit again, and the results reflected that the nuclear expression of HIF-1α, PPAR-γ and PKM2 can be all inhibited by Sora + Sim co-treatment, and the changes of them after FG-4592 or BAY87–2243 treatment were similar to the total protein (Fig. 6b). These effects imply that HIF-1α is the up-stream regulator of PPAR-γ and PKM2.
Next, we used an activator and inhibitor of PPAR-γ for further confirmation. In Fig. 6c, after treatment with the PPAR-γ activator Rosiglitazone (10 μM) [38, 39], the expression of both PPAR-γ and PKM2 were increased, whereas the expression of HIF-1α remained similar to the CTRL group. If however, Rosiglitazone was co-treated with Sora + Sim, the increases seen in PPAR-γ and PKM2 were abolished, and the expression of HIF-1α remained the same as in the Sora + Sim group. Moreover, after treatment with the PPAR-γ inhibitor GW9662 (2 μM) [38, 39], the expression of PPAR-γ and PKM2 were decreased, whereas the expression of HIF-1α remained unchanged. Furthermore, when GW9662 was co-treated with Sora + Sim, the decrease in PPAR-γ and PKM2 was enhanced. As for the nuclear fractions, the changes of HIF-1α, PPAR-γ and PKM2 were similar to the total protein when treated with Rosiglitazone or GW9662 (Fig. 6d). These results suggest that HIF-1α was the up-stream regulator of PPAR-γ, and PKM2 was possibly the down-stream regulator of PPAR-γ.
Based on the above findings, we went on to use both an activator and inhibitor of PKM2 (Fig. 6e). Since PKM2 is a pyruvate kinase, the activator DASA 58 was able to elevate its enzymatic activity but inhibit its expression, and the inhibitor compound 3 K was able to reduce the enzymatic activity of PKM2 but promote its expression. Therefore after treatment with compound 3 K (3 μM) [40], the expression of PKM2 was up-regulated, whereas the expression of HIF-1α and PPAR-γ were found to be the same as the CTRL group. However, if compound 3 K was co-treated with Sora + Sim, the up-regulation of PKM2 was decreased slightly, and the expression of HIF-1α and PPAR-γ remained the same as the Sora + Sim group. The effect of co-treatment with DASA 58 (40 μM) was however, opposite to that seen with compound 3 K [29]. The analysis of nuclear fractions also showed that the changes of HIF-1α, PPAR-γ and PKM2 were similar to the total protein when treated with compound 3 K or DASA 58 (Fig. 6f). In conclusion, these results provide further evidence for an up-stream regulatory role for HIF-1α and PPAR-γ on PKM2. The HIF-1α/PPAR-γ/PKM2 axis was suppressed in Sim treatment.
PKM2 is not only a rate-limiting enzyme during glycolysis, but also a critical transcription factor in the nucleus. We next looked for a role for PKM2 in Sora resistance using lentiviral transfection. Figure 6g shows the over expression of PKM2 in LM3 cells, and Fig. 6h the knock down of PKM2 in LM2-SR cells. As revealed in Fig. 6i and j, if LM3 cells were overexpressed with PKM2, the expression of PCNA was elevated in LM3 cells, and the expression of Bax, Caspase 3 and OXPHOS were inhibited. Moreover, lactate production levels and glucose uptake levels were enhanced in the PKM2-OE group in LM3 cells. This implied that the over expression of PKM2 in LM3 cells may lead to resistance to Sora. This hypothesis was then verified by using the CCK8 assay, as seen in Fig. 6k. The results showed that the IC50 of LM3-PKM2-OE cells to Sora was 8.68 μM (LM3 cells were 4.47 μM) at 24 h. However, if PKM2 was knocked down in LM3-SR cells, the proliferation, inhibition of apoptosis and glycolysis levels were all suppressed, suggesting that the resistance to Sora in LM3-SR cells was recovered (Fig. 6i and j). The effect of Sora + Sim co-treatment in LM3-SR-PKM2-KD cells was also detected by western blotting (Fig. 6l). The results demonstrated that if PKM2 was knocked down in LM3-SR cells, Sim failed to inhibit the expression of PKM2, PCNA, Bcl-2, or increase Bax, Caspase 3 and OXPHOS in the Sora + Sim group when compared to the Sora group alone. Therefore, in summary, these results confirmed the involvement of the HIF-1α/PPAR-γ/PKM2 axis in Sim treatment, and proved that the over expression of PKM2 may lead to resistance to Sora in LM3 cells, whereas the knock down of PKM2 may improve Sora resistance in LM3-SR cells.

Discussion

Sora resistance is the main limiting factor in the effective treatment of advanced HCC. In humans, the average time for Sora resistance to occur is approximately 12.2 moths but can vary from months to years [6]. The establishment of a Sora-resistant HCC cell line usually takes about 12 weeks. In our study, we successfully established LM3-SR cells, which could tolerate 10 μM Sora (with an IC50 of 16.33 μM) to study the relationship between Sora resistance and aerobic glycolysis in vivo and in vitro.
The mechanisms of Sora resistance remain obscure, but new insights include a higher EGFR, c-Jun and Akt activation in HCC cells, as well as increased EMT, cancer stem cells, hypoxic environment, autophagy, and exosomes [2, 5, 34, 41, 42]. Recently, several studies reported that aerobic glycolysis may also contribute to Sora resistance. In the 1920s, Otto Warburg found that even in conditions of sufficient oxygen levels, tumor cells prefer to metabolize glucose via glycolysis to lactate, rather than OXPHOS to generate metabolic intermediates rapidly, and this phenomenon is now termed the Warburg effect [43, 44]. Fiume et al. found that Sora treatment could damage OXPHOS and promote aerobic glycolysis in cells grown in a glucose rich environment [45]. Reyes et al. found that Sora and 2-Deoxyglucose (2-DG) co-treatment could synergistically inhibit the proliferation of Sora resistant HCC cells by inhibiting ATP production [34]. It was also shown by Wong et al. that 2-DG can reverse Sora resistance in HCC [44]. Therefore, in our study, we used the original LM3 and LM3-SR cells to detect glycolysis levels. Our results show that LM3-SR cells exhibited a higher lactate production and glucose uptake levels when compared to LM3 cells, which can support the enhanced aerobic glycolysis seen during Sora resistance in HCC cells.
Additionally, Sim, is a cholesterol-lowering agent, and recently has also been reported to participate in the suppression of glycolysis and Sora resistance. Christie et al. found that statins can partially block the utilization of glycolytic ATP [46]. Huang et al. found that the combination of pitavastatin and paclitaxel can significantly decrease the glycolytic rate in renal carcinoma [47]. Nowis et al. reported that statins impaired glucose uptake in human cells derived from the liver [48]. These findings suggest that Sim may also be effective in inhibiting glycolysis and may be a potential agent to help improve glycolysis-mediated Sora resistance. In our study therefore, based on the pluripotent nature of statins, we wanted to combine Sim with Sora to explore whether Sim can improve the sensitivity of LM3-SR cells to Sora. Firstly, we detected the effect of Sim on LM3 and LM3-SR cells alone. We found that LM3-SR cells were more sensitive to Sim (10 μM) than LM3 cells, which was reflected by a higher inhibition of proliferation rate and greater apoptosis. In our study, Sim (10 μM) was also effective at inhibiting aerobic levels by reducing glycolytic lactate and glucose production, and inhibiting the expression of glycolysis related protein expression. Secondly, combined treatments using both Sora and Sim were performed, and the results showed that Sim synergistically enhanced the sensitivity of LM3-SR cells to Sora in a combined treatment, as reflected by a CI value less than 1. Increased inhibition of proliferation and higher apoptosis were also seen, both in vivo and in vitro.
The mechanism underlying the re-sensitizing effect of Sim on Sora was explored next. There are three rate-limiting enzymes during aerobic glycolysis: HK2, PFK1 and PKM2. Among them, PKM2, can catalyze the last step of glycolysis, and is up-regulated in many tumors [49, 50]. PKM2 has three functions in cancer cells: (1) cytoplasmic PKM2 is a tetramer with high enzyme activity, and takes part in glycolysis to provide increased metabolic intermediates for cancer cell biosynthesis, (2) the dimeric isoform of PKM2 can translocate to nucleus and act as a transcriptional co-activator, thereby facilitating the transcription of genes beneficial to growth, such as GLUTs, HIF-1α, VEGF-A, Bax, Bcl-2, and PCNA. (3) PKM2 can also translocate to the mitochondria under oxidative stress to interact with Bcl-2/Bcl-xl, causing the inhibition of cancer cell apoptosis [5153]. Based on the critical role of PKM2, Zhang et al. reported that silencing PKM2 could re-sensitize Hep3B-SR and LM3-SR cells to Sora [33]. Wong et al. also found that the PRMT6-ERK-PKM2 regulatory axis takes part in Sora resistance and glucose metabolism in HCC [44]. In our study, we firstly found that Sora + Sim co-treatment can not only inhibit the expression of PKM2, but also inhibit the nuclear translocation of PKM2 by IF staining. Secondly, we revealed that the over expression of PKM2 in LM3 cells led to Sora resistance. However, knock down of PKM2 in LM3-SR cells effectively restored the sensitivity of LM3-SR cells to Sora, and Sim + Sora co-treatment failed to inhibit the proliferation or increase apoptosis in LM3-SR-PKM2-KD cells. These finding provide further proof for a critical role of PKM2 in the synergic co-treatment of Sora and Sim.
The expression of the PKM2 gene can also be driven by various factors, including HIF-1α, STAT3, β-Catenin, and NF-κB [5456]. In this study we found that both HIF-1α and PPAR-γ may be involved in the re-sensitization effect of Sim on Sora resistance. HIF-1α, which is a regulatory factor involved in the cellular response to hypoxia, can promote glycolysis in cancer cells via the direct transcriptional activation of glycolysis related genes, including glucose transporters (GLUTs) and PKM2 [57]. PPAR-γ plays an important role in maintaining energy homeostasis through the modulation of glucose and lipid metabolism, and PPAR-γ is usually overexpressed in cancer cells causing accelerated tumor growth [58, 59]. However, the role of PPAR-γ agonists and antagonists in tumor treatment is complex, as both have been found to inhibit the growth of tumor cells [58, 6062]. PPAR-γ has been found to promote glucose uptake during lipid metabolism, and can induce the expression of glycolytic proteins, including GLUT-4 [63]. It has been seen that atorvastatin can inhibit the HIF-1α/PPAR-γ pathway and inhibit the survival of induced pluripotent stem cells [15]. In addition, Panasyuk et al. also reported that PPAR-γ can promote the expression of PKM2 and HK2 in fatty liver [59]. In our study, we found that PKM2 can interact with both HIF-1α and PPAR-γ using CO-IP assay. We subsequently used activators and inhibitors of HIF-1α, PPAR-γ and PKM2 in LM3-SR cells, and the results confirmed a role for the HIF-1α/PAR-γ/PKM2 axis in LM3-SR cells. In addition, our rescue experiments showed that the co-treatment effects of Sora + Sim can be reversed by the HIF-1α, and PPAR-γ activators FG4592 and Rosiglitazone and the PKM2 inhibitor compound 3 k. These findings provide convincing evidence for the HIF-1α/PAR-γ/PKM2 axis as the target for Sim during re-sensitization of HCC cells to Sora (Fig. 7).
The mechanism underlying Sora resistance is complex. Except for the HIF-1α/PPAR-γ/PKM2 axis investigated in the present study, the PKM2 isoform, PKM1, is also reported to increase glycolysis through autophagy and cause cancer chemoresistance [64, 65]. Moreover, the oncogene Myc, which includes c-Myc, N-Myc, and L-Myc, plays an important role in aerobic glycolysis in HCC as well. C-Myc is reported to overexpressed in HCC, and can promote the Warburg effect by increasing the expression of glycolytic related markers, such as GLUT1, LDH and PKM2 [6669]. Conversely, c-Myc can also be regulated by PKM2 in the nucleus, for PKM2 can translocate into nucleus and act as a coactivator of β-catenin to induce c-Myc expression, leading to the expression of c-Myc targeted genes [30, 67]. In addition, high levels of c-Myc activity will enhance the PKM2/PKM1 ratios [49, 70]. c-Myc also increases the glutaminolysis in cancer cells and then promotes the progression of cancer [71]. Based on these, we proposed that c-Myc may be a possible for the treatment of Sora resistance in HCC, and this can be investigated in the future.

Conclusions

In conclusion, our study investigated the promising role of Sim in improving HCC resistance to Sora, and we found that: (1) Sim was safe for co-treatment with Sora in vivo and did not aggravate liver function or organ damage. (2) Sim can inhibit the HIF-1α/PAR-γ/PKM2 axis, causing the suppression of PKM2-mediated glycolysis, decrease proliferation and increased apoptosis in HCC cells, thereby re-sensitize HCC cells to Sora.

Supplementary information

Supplementary information accompanies this paper at https://​doi.​org/​10.​1186/​s13046-020-1528-x.

Acknowledgements

We appreciate International Science Editing (http://www.internationalscienceediting.com) for language editing for us.
The study was approved by the Animal Care and Use Committee of Shanghai Tongji University, and conducted following the ARRIVE guidelines.
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 Lonardo A, Loria P. Potential for statins in the chemoprevention and management of hepatocellular carcinoma. J Gastroenterol Hepatol. 2012;27:1654–64.PubMedCrossRef Lonardo A, Loria P. Potential for statins in the chemoprevention and management of hepatocellular carcinoma. J Gastroenterol Hepatol. 2012;27:1654–64.PubMedCrossRef
2.
Zurück zum Zitat Chen J, Jin R, Zhao J, Liu J, Ying H, Yan H, et al. Potential molecular, cellular and microenvironmental mechanism of sorafenib resistance in hepatocellular carcinoma. Cancer Lett. 2015;367:1–11.PubMedCrossRef Chen J, Jin R, Zhao J, Liu J, Ying H, Yan H, et al. Potential molecular, cellular and microenvironmental mechanism of sorafenib resistance in hepatocellular carcinoma. Cancer Lett. 2015;367:1–11.PubMedCrossRef
3.
Zurück zum Zitat Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018;68:394–424.PubMedCrossRef Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018;68:394–424.PubMedCrossRef
4.
Zurück zum Zitat El-Serag HB, Rudolph KL. Hepatocellular carcinoma: epidemiology and molecular carcinogenesis. Gastroenterol. 2007;132:2557–76.CrossRef El-Serag HB, Rudolph KL. Hepatocellular carcinoma: epidemiology and molecular carcinogenesis. Gastroenterol. 2007;132:2557–76.CrossRef
5.
Zurück zum Zitat Niu L, Liu L, Yang S, Ren J, Lai PBS, Chen GG. New insights into sorafenib resistance in hepatocellular carcinoma: responsible mechanisms and promising strategies. Biochim Biophys Acta Rev Cancer. 2017;1868:564–70.PubMedCrossRef Niu L, Liu L, Yang S, Ren J, Lai PBS, Chen GG. New insights into sorafenib resistance in hepatocellular carcinoma: responsible mechanisms and promising strategies. Biochim Biophys Acta Rev Cancer. 2017;1868:564–70.PubMedCrossRef
6.
Zurück zum Zitat Hatooka M, Kawaoka T, Aikata H, Morio K, Kobayashi T, Hiramatsu A, et al. Comparison of outcome of hepatic arterial infusion chemotherapy and Sorafenib in patients with hepatocellular carcinoma refractory to Transcatheter arterial chemoembolization. Anticancer Res. 2016;36:3523–9.PubMed Hatooka M, Kawaoka T, Aikata H, Morio K, Kobayashi T, Hiramatsu A, et al. Comparison of outcome of hepatic arterial infusion chemotherapy and Sorafenib in patients with hepatocellular carcinoma refractory to Transcatheter arterial chemoembolization. Anticancer Res. 2016;36:3523–9.PubMed
7.
Zurück zum Zitat Llovet JM, Ricci S, Mazzaferro V, Hilgard P, Gane E, Blanc JF, et al. Sorafenib in advanced hepatocellular carcinoma. N Engl J Med. 2008;359:378–90.PubMedCrossRef Llovet JM, Ricci S, Mazzaferro V, Hilgard P, Gane E, Blanc JF, et al. Sorafenib in advanced hepatocellular carcinoma. N Engl J Med. 2008;359:378–90.PubMedCrossRef
8.
Zurück zum Zitat Cheng AL, Kang YK, Chen Z, Tsao CJ, Qin S, Kim JS, et al. Efficacy and safety of sorafenib in patients in the Asia-Pacific region with advanced hepatocellular carcinoma: a phase III randomised, double-blind, placebo-controlled trial. Lancet Oncol. 2009;10:25–34.PubMedCrossRef Cheng AL, Kang YK, Chen Z, Tsao CJ, Qin S, Kim JS, et al. Efficacy and safety of sorafenib in patients in the Asia-Pacific region with advanced hepatocellular carcinoma: a phase III randomised, double-blind, placebo-controlled trial. Lancet Oncol. 2009;10:25–34.PubMedCrossRef
9.
Zurück zum Zitat Li S, Dai W, Mo W, Li J, Feng J, Wu L, et al. By inhibiting PFKFB3, aspirin overcomes sorafenib resistance in hepatocellular carcinoma. Int J Cancer. 2017;141:2571–84.PubMedCrossRef Li S, Dai W, Mo W, Li J, Feng J, Wu L, et al. By inhibiting PFKFB3, aspirin overcomes sorafenib resistance in hepatocellular carcinoma. Int J Cancer. 2017;141:2571–84.PubMedCrossRef
10.
Zurück zum Zitat Li S, Li J, Dai W, Zhang Q, Feng J, Wu L, et al. Genistein suppresses aerobic glycolysis and induces hepatocellular carcinoma cell death. Br J Cancer. 2017;117:1518–28.PubMedPubMedCentralCrossRef Li S, Li J, Dai W, Zhang Q, Feng J, Wu L, et al. Genistein suppresses aerobic glycolysis and induces hepatocellular carcinoma cell death. Br J Cancer. 2017;117:1518–28.PubMedPubMedCentralCrossRef
11.
Zurück zum Zitat Takimoto CH, Awada A. Safety and anti-tumor activity of sorafenib (Nexavar) in combination with other anti-cancer agents: a review of clinical trials. Cancer Chemother Pharmacol. 2008;61:535–48.PubMedCrossRef Takimoto CH, Awada A. Safety and anti-tumor activity of sorafenib (Nexavar) in combination with other anti-cancer agents: a review of clinical trials. Cancer Chemother Pharmacol. 2008;61:535–48.PubMedCrossRef
12.
Zurück zum Zitat Abou-Alfa GK, Johnson P, Knox JJ, Capanu M, Davidenko I, Lacava J, et al. Doxorubicin plus sorafenib vs doxorubicin alone in patients with advanced hepatocellular carcinoma: a randomized trial. JAMA. 2010;304:2154–60.PubMedCrossRef Abou-Alfa GK, Johnson P, Knox JJ, Capanu M, Davidenko I, Lacava J, et al. Doxorubicin plus sorafenib vs doxorubicin alone in patients with advanced hepatocellular carcinoma: a randomized trial. JAMA. 2010;304:2154–60.PubMedCrossRef
13.
Zurück zum Zitat Pose E, Trebicka J, Mookerjee RP, Angeli P, Gines P. Statins: old drugs as new therapy for liver diseases? J Hepatol. 2019;70:194–202.PubMedCrossRef Pose E, Trebicka J, Mookerjee RP, Angeli P, Gines P. Statins: old drugs as new therapy for liver diseases? J Hepatol. 2019;70:194–202.PubMedCrossRef
14.
Zurück zum Zitat Rzouq F, Alahdab F, Olyaee M. Statins and hepatitis C virus infection: an old therapy with new scope. Am J Med Sci. 2014;348:426–30.PubMedCrossRef Rzouq F, Alahdab F, Olyaee M. Statins and hepatitis C virus infection: an old therapy with new scope. Am J Med Sci. 2014;348:426–30.PubMedCrossRef
15.
Zurück zum Zitat Nakashima Y, Miyagi-Shiohira C, Noguchi H, Omasa T. Atorvastatin inhibits the HIF1alpha-PPAR Axis, which is essential for maintaining the function of human induced pluripotent stem cells. Mol Ther. 2018;26:1715–34.PubMedPubMedCentralCrossRef Nakashima Y, Miyagi-Shiohira C, Noguchi H, Omasa T. Atorvastatin inhibits the HIF1alpha-PPAR Axis, which is essential for maintaining the function of human induced pluripotent stem cells. Mol Ther. 2018;26:1715–34.PubMedPubMedCentralCrossRef
16.
Zurück zum Zitat Goh MJ, Sinn DH, Kim S, Woo SY, Cho H, Kang W, et al. Statin Use and the Risk of Hepatocellular Carcinoma in Patients with Chronic Hepatitis B. Hepatol. 2019. Goh MJ, Sinn DH, Kim S, Woo SY, Cho H, Kang W, et al. Statin Use and the Risk of Hepatocellular Carcinoma in Patients with Chronic Hepatitis B. Hepatol. 2019.
17.
Zurück zum Zitat Miao Q, Zeng X, Ma G, Li N, Liu Y, Luo T, et al. Simvastatin suppresses the proangiogenic microenvironment of human hepatic stellate cells via the Kruppel-like factor 2 pathway. Rev Esp Enferm Dig. 2015;107:63–71.PubMed Miao Q, Zeng X, Ma G, Li N, Liu Y, Luo T, et al. Simvastatin suppresses the proangiogenic microenvironment of human hepatic stellate cells via the Kruppel-like factor 2 pathway. Rev Esp Enferm Dig. 2015;107:63–71.PubMed
18.
Zurück zum Zitat Huang X, Ma J, Xu J, Su Q, Zhao J. Simvastatin induces growth inhibition and apoptosis in HepG2 and Huh7 hepatocellular carcinoma cells via upregulation of Notch1 expression. Mol Med Rep. 2015;11:2334–40.PubMedCrossRef Huang X, Ma J, Xu J, Su Q, Zhao J. Simvastatin induces growth inhibition and apoptosis in HepG2 and Huh7 hepatocellular carcinoma cells via upregulation of Notch1 expression. Mol Med Rep. 2015;11:2334–40.PubMedCrossRef
19.
Zurück zum Zitat Zhang S, Doudican NA, Quay E, Orlow SJ. Fluvastatin enhances sorafenib cytotoxicity in melanoma cells via modulation of AKT and JNK signaling pathways. Anticancer Res. 2011;31:3259–65.PubMed Zhang S, Doudican NA, Quay E, Orlow SJ. Fluvastatin enhances sorafenib cytotoxicity in melanoma cells via modulation of AKT and JNK signaling pathways. Anticancer Res. 2011;31:3259–65.PubMed
20.
Zurück zum Zitat Kim W, Yoon JH, Kim JR, Jang IJ, Bang YJ, Kim YJ, et al. Synergistic anti-tumor efficacy of lovastatin and protein kinase C-beta inhibitor in hepatocellular carcinoma. Cancer Chemother Pharmacol. 2009;64:497–507.PubMedCrossRef Kim W, Yoon JH, Kim JR, Jang IJ, Bang YJ, Kim YJ, et al. Synergistic anti-tumor efficacy of lovastatin and protein kinase C-beta inhibitor in hepatocellular carcinoma. Cancer Chemother Pharmacol. 2009;64:497–507.PubMedCrossRef
21.
Zurück zum Zitat Lee SJ, Hwang JW, Yim H, Yim HJ, Woo SU, Suh SJ, et al. Synergistic effect of simvastatin plus NS398 on inhibition of proliferation and survival in hepatocellular carcinoma cell line. J Gastroenterol Hepatol. 2014;29:1299–307.PubMedCrossRef Lee SJ, Hwang JW, Yim H, Yim HJ, Woo SU, Suh SJ, et al. Synergistic effect of simvastatin plus NS398 on inhibition of proliferation and survival in hepatocellular carcinoma cell line. J Gastroenterol Hepatol. 2014;29:1299–307.PubMedCrossRef
22.
Zurück zum Zitat Gao J, Jia WD, Li JS, Wang W, Xu GL, Ma JL, et al. Combined inhibitory effects of celecoxib and fluvastatin on the growth of human hepatocellular carcinoma xenografts in nude mice. J Int Med Res. 2010;38:1413–27.PubMedCrossRef Gao J, Jia WD, Li JS, Wang W, Xu GL, Ma JL, et al. Combined inhibitory effects of celecoxib and fluvastatin on the growth of human hepatocellular carcinoma xenografts in nude mice. J Int Med Res. 2010;38:1413–27.PubMedCrossRef
23.
Zurück zum Zitat Lopez-Terrada D, Cheung SW, Finegold MJ, Knowles BB. Hep G2 is a hepatoblastoma-derived cell line. Hum Pathol. 2009;40:1512–5.PubMedCrossRef Lopez-Terrada D, Cheung SW, Finegold MJ, Knowles BB. Hep G2 is a hepatoblastoma-derived cell line. Hum Pathol. 2009;40:1512–5.PubMedCrossRef
24.
Zurück zum Zitat Liu J, Liu Y, Meng L, Liu K, Ji B. Targeting the PD-L1/DNMT1 axis in acquired resistance to sorafenib in human hepatocellular carcinoma. Oncol Rep. 2017;38:899–907.PubMedPubMedCentralCrossRef Liu J, Liu Y, Meng L, Liu K, Ji B. Targeting the PD-L1/DNMT1 axis in acquired resistance to sorafenib in human hepatocellular carcinoma. Oncol Rep. 2017;38:899–907.PubMedPubMedCentralCrossRef
25.
Zurück zum Zitat Gao L, Wang X, Tang Y, Huang S, Hu CA, Teng Y. FGF19/FGFR4 signaling contributes to the resistance of hepatocellular carcinoma to sorafenib. J Exp Clin Cancer Res. 2017;36:8.PubMedPubMedCentralCrossRef Gao L, Wang X, Tang Y, Huang S, Hu CA, Teng Y. FGF19/FGFR4 signaling contributes to the resistance of hepatocellular carcinoma to sorafenib. J Exp Clin Cancer Res. 2017;36:8.PubMedPubMedCentralCrossRef
26.
Zurück zum Zitat Feng J, Chen K, Xia Y, Wu L, Li J, Li S, et al. Salidroside ameliorates autophagy and activation of hepatic stellate cells in mice via NF-kappaB and TGF-beta1/Smad3 pathways. Drug Des Devel Ther. 2018;12:1837–53.PubMedPubMedCentralCrossRef Feng J, Chen K, Xia Y, Wu L, Li J, Li S, et al. Salidroside ameliorates autophagy and activation of hepatic stellate cells in mice via NF-kappaB and TGF-beta1/Smad3 pathways. Drug Des Devel Ther. 2018;12:1837–53.PubMedPubMedCentralCrossRef
27.
Zurück zum Zitat Feng J, Zhang Q, Mo W, Wu L, Li S, Li J, et al. Salidroside pretreatment attenuates apoptosis and autophagy during hepatic ischemia-reperfusion injury by inhibiting the mitogen-activated protein kinase pathway in mice. Drug Des Devel Ther. 2017;11:1989–2006.PubMedPubMedCentralCrossRef Feng J, Zhang Q, Mo W, Wu L, Li S, Li J, et al. Salidroside pretreatment attenuates apoptosis and autophagy during hepatic ischemia-reperfusion injury by inhibiting the mitogen-activated protein kinase pathway in mice. Drug Des Devel Ther. 2017;11:1989–2006.PubMedPubMedCentralCrossRef
28.
Zurück zum Zitat Xu S, Wu L, Zhang Q, Feng J, Li S, Li J, et al. Pretreatment with propylene glycol alginate sodium sulfate ameliorated concanavalin A-induced liver injury by regulating the PI3K/Akt pathway in mice. Life Sci. 2017;185:103–13.PubMedCrossRef Xu S, Wu L, Zhang Q, Feng J, Li S, Li J, et al. Pretreatment with propylene glycol alginate sodium sulfate ameliorated concanavalin A-induced liver injury by regulating the PI3K/Akt pathway in mice. Life Sci. 2017;185:103–13.PubMedCrossRef
29.
Zurück zum Zitat Feng J, Wu L, Ji J, Chen K, Yu Q, Zhang J, et al. PKM2 is the target of proanthocyanidin B2 during the inhibition of hepatocellular carcinoma. J Exp Clin Cancer Res. 2019;38:204.PubMedPubMedCentralCrossRef Feng J, Wu L, Ji J, Chen K, Yu Q, Zhang J, et al. PKM2 is the target of proanthocyanidin B2 during the inhibition of hepatocellular carcinoma. J Exp Clin Cancer Res. 2019;38:204.PubMedPubMedCentralCrossRef
30.
Zurück zum Zitat Liu R, Li Y, Tian L, Shi H, Wang J, Liang Y, et al. Gankyrin drives metabolic reprogramming to promote tumorigenesis, metastasis and drug resistance through activating beta-catenin/c-Myc signaling in human hepatocellular carcinoma. Cancer Lett. 2019;443:34–46.PubMedCrossRef Liu R, Li Y, Tian L, Shi H, Wang J, Liang Y, et al. Gankyrin drives metabolic reprogramming to promote tumorigenesis, metastasis and drug resistance through activating beta-catenin/c-Myc signaling in human hepatocellular carcinoma. Cancer Lett. 2019;443:34–46.PubMedCrossRef
31.
Zurück zum Zitat Lu X, Liu T, Chen K, Xia Y, Dai W, Xu S, et al. Isorhamnetin: a hepatoprotective flavonoid inhibits apoptosis and autophagy via P38/PPAR-alpha pathway in mice. Biomed Pharmacother. 2018;103:800–11.PubMedCrossRef Lu X, Liu T, Chen K, Xia Y, Dai W, Xu S, et al. Isorhamnetin: a hepatoprotective flavonoid inhibits apoptosis and autophagy via P38/PPAR-alpha pathway in mice. Biomed Pharmacother. 2018;103:800–11.PubMedCrossRef
32.
Zurück zum Zitat Yu Q, Liu T, Li S, Feng J, Wu L, Wang W, et al. The protective effects of Levo-Tetrahydropalmatine on ConA-induced liver injury are via TRAF6/JNK signaling. Mediat Inflamm. 2018;2018:4032484. Yu Q, Liu T, Li S, Feng J, Wu L, Wang W, et al. The protective effects of Levo-Tetrahydropalmatine on ConA-induced liver injury are via TRAF6/JNK signaling. Mediat Inflamm. 2018;2018:4032484.
33.
Zurück zum Zitat Zhang M, Zhang H, Hong H, Zhang Z. MiR-374b re-sensitizes hepatocellular carcinoma cells to sorafenib therapy by antagonizing PKM2-mediated glycolysis pathway. Am J Cancer Res. 2019;9:765–78.PubMedPubMedCentral Zhang M, Zhang H, Hong H, Zhang Z. MiR-374b re-sensitizes hepatocellular carcinoma cells to sorafenib therapy by antagonizing PKM2-mediated glycolysis pathway. Am J Cancer Res. 2019;9:765–78.PubMedPubMedCentral
34.
Zurück zum Zitat Reyes R, Wani NA, Ghoshal K, Jacob ST, Motiwala T. Sorafenib and 2-Deoxyglucose synergistically inhibit proliferation of both Sorafenib-sensitive and -resistant HCC cells by inhibiting ATP production. Gene Expr. 2017;17:129–40.PubMedCrossRef Reyes R, Wani NA, Ghoshal K, Jacob ST, Motiwala T. Sorafenib and 2-Deoxyglucose synergistically inhibit proliferation of both Sorafenib-sensitive and -resistant HCC cells by inhibiting ATP production. Gene Expr. 2017;17:129–40.PubMedCrossRef
35.
Zurück zum Zitat Bhadange Y, Lautert J, Li S, Lawando E, Kim ET, Soper MC, et al. Hypoxia and the Prolyl hydroxylase inhibitor FG-4592 protect corneal endothelial cells from mechanical and perioperative surgical stress. Cornea. 2018;37:501–7.PubMedPubMedCentralCrossRef Bhadange Y, Lautert J, Li S, Lawando E, Kim ET, Soper MC, et al. Hypoxia and the Prolyl hydroxylase inhibitor FG-4592 protect corneal endothelial cells from mechanical and perioperative surgical stress. Cornea. 2018;37:501–7.PubMedPubMedCentralCrossRef
36.
Zurück zum Zitat Xie RY, Fang XL, Zheng XB, Lv WZ, Li YJ, Ibrahim Rage H, et al. Salidroside and FG-4592 ameliorate high glucose-induced glomerular endothelial cells injury via HIF upregulation. Biomed Pharmacother. 2019;118:109175.PubMedCrossRef Xie RY, Fang XL, Zheng XB, Lv WZ, Li YJ, Ibrahim Rage H, et al. Salidroside and FG-4592 ameliorate high glucose-induced glomerular endothelial cells injury via HIF upregulation. Biomed Pharmacother. 2019;118:109175.PubMedCrossRef
37.
Zurück zum Zitat Ellinghaus P, Heisler I, Unterschemmann K, Haerter M, Beck H, Greschat S, et al. BAY 87-2243, a highly potent and selective inhibitor of hypoxia-induced gene activation has antitumor activities by inhibition of mitochondrial complex I. Cancer Med. 2013;2:611–24.PubMedPubMedCentral Ellinghaus P, Heisler I, Unterschemmann K, Haerter M, Beck H, Greschat S, et al. BAY 87-2243, a highly potent and selective inhibitor of hypoxia-induced gene activation has antitumor activities by inhibition of mitochondrial complex I. Cancer Med. 2013;2:611–24.PubMedPubMedCentral
38.
Zurück zum Zitat Jiang C, Liu S, Cao Y, Shan H. High glucose induces autophagy through PPARgamma-dependent pathway in human nucleus Pulposus cells. PPAR Res. 2018;2018:8512745.PubMedPubMedCentralCrossRef Jiang C, Liu S, Cao Y, Shan H. High glucose induces autophagy through PPARgamma-dependent pathway in human nucleus Pulposus cells. PPAR Res. 2018;2018:8512745.PubMedPubMedCentralCrossRef
39.
Zurück zum Zitat Zhang M, Qian C, Zheng ZG, Qian F, Wang Y, Thu PM, et al. Jujuboside a promotes Abeta clearance and ameliorates cognitive deficiency in Alzheimer's disease through activating Axl/HSP90/PPARgamma pathway. Theranostics. 2018;8:4262–78.PubMedPubMedCentralCrossRef Zhang M, Qian C, Zheng ZG, Qian F, Wang Y, Thu PM, et al. Jujuboside a promotes Abeta clearance and ameliorates cognitive deficiency in Alzheimer's disease through activating Axl/HSP90/PPARgamma pathway. Theranostics. 2018;8:4262–78.PubMedPubMedCentralCrossRef
40.
Zurück zum Zitat Ning X, Qi H, Li R, Li Y, Jin Y, McNutt MA, et al. Discovery of novel naphthoquinone derivatives as inhibitors of the tumor cell specific M2 isoform of pyruvate kinase. Eur J Med Chem. 2017;138:343–52.PubMedCrossRef Ning X, Qi H, Li R, Li Y, Jin Y, McNutt MA, et al. Discovery of novel naphthoquinone derivatives as inhibitors of the tumor cell specific M2 isoform of pyruvate kinase. Eur J Med Chem. 2017;138:343–52.PubMedCrossRef
41.
Zurück zum Zitat Qu Z, Wu J, Wu J, Luo D, Jiang C, Ding Y. Exosomes derived from HCC cells induce sorafenib resistance in hepatocellular carcinoma both in vivo and in vitro. J Exp Clin Cancer Res. 2016;35:159.PubMedPubMedCentralCrossRef Qu Z, Wu J, Wu J, Luo D, Jiang C, Ding Y. Exosomes derived from HCC cells induce sorafenib resistance in hepatocellular carcinoma both in vivo and in vitro. J Exp Clin Cancer Res. 2016;35:159.PubMedPubMedCentralCrossRef
42.
44.
Zurück zum Zitat Wong TL, Ng KY, Tan KV, Chan LH, Zhou L, Che N, et al. CRAF methylation by PRMT6 regulates aerobic glycolysis driven hepatocarcinogenesis via ERK-dependent PKM2 nuclear relocalization and activation. Hepatol. 2019. Wong TL, Ng KY, Tan KV, Chan LH, Zhou L, Che N, et al. CRAF methylation by PRMT6 regulates aerobic glycolysis driven hepatocarcinogenesis via ERK-dependent PKM2 nuclear relocalization and activation. Hepatol. 2019.
45.
Zurück zum Zitat Fiume L, Manerba M, Vettraino M, Di Stefano G. Effect of sorafenib on the energy metabolism of hepatocellular carcinoma cells. Eur J Pharmacol. 2011;670:39–43.PubMedCrossRef Fiume L, Manerba M, Vettraino M, Di Stefano G. Effect of sorafenib on the energy metabolism of hepatocellular carcinoma cells. Eur J Pharmacol. 2011;670:39–43.PubMedCrossRef
46.
Zurück zum Zitat Christie CF, Fang D, Hunt EG, Morris ME, Rovini A, Heslop KA, et al. Statin-dependent modulation of mitochondrial metabolism in cancer cells is independent of cholesterol content. FASEB J. 2019;33:8186–201.PubMedCrossRefPubMedCentral Christie CF, Fang D, Hunt EG, Morris ME, Rovini A, Heslop KA, et al. Statin-dependent modulation of mitochondrial metabolism in cancer cells is independent of cholesterol content. FASEB J. 2019;33:8186–201.PubMedCrossRefPubMedCentral
47.
Zurück zum Zitat Huang J, Yang X, Peng X, Huang W. Inhibiting prenylation augments chemotherapy efficacy in renal cell carcinoma through dual inhibition on mitochondrial respiration and glycolysis. Biochem Biophys Res Commun. 2017;493:921–7.PubMedCrossRef Huang J, Yang X, Peng X, Huang W. Inhibiting prenylation augments chemotherapy efficacy in renal cell carcinoma through dual inhibition on mitochondrial respiration and glycolysis. Biochem Biophys Res Commun. 2017;493:921–7.PubMedCrossRef
48.
Zurück zum Zitat Nowis D, Malenda A, Furs K, Oleszczak B, Sadowski R, Chlebowska J, et al. Statins impair glucose uptake in human cells. BMJ Open Diabetes Res Care. 2014;2:e000017.PubMedPubMedCentralCrossRef Nowis D, Malenda A, Furs K, Oleszczak B, Sadowski R, Chlebowska J, et al. Statins impair glucose uptake in human cells. BMJ Open Diabetes Res Care. 2014;2:e000017.PubMedPubMedCentralCrossRef
49.
50.
Zurück zum Zitat Wong N, Ojo D, Yan J, Tang D. PKM2 contributes to cancer metabolism. Cancer Lett. 2015;356:184–91.PubMedCrossRef Wong N, Ojo D, Yan J, Tang D. PKM2 contributes to cancer metabolism. Cancer Lett. 2015;356:184–91.PubMedCrossRef
51.
Zurück zum Zitat Dong T, Yan Y, Chai H, Chen S, Xiong X, Sun D, et al. Pyruvate kinase M2 affects liver cancer cell behavior through up-regulation of HIF-1alpha and Bcl-xL in culture. Biomed Pharmacother. 2015;69:277–84.PubMedCrossRef Dong T, Yan Y, Chai H, Chen S, Xiong X, Sun D, et al. Pyruvate kinase M2 affects liver cancer cell behavior through up-regulation of HIF-1alpha and Bcl-xL in culture. Biomed Pharmacother. 2015;69:277–84.PubMedCrossRef
52.
Zurück zum Zitat Palsson-McDermott EM, Curtis AM, Goel G, Lauterbach MA, Sheedy FJ, Gleeson LE, et al. Pyruvate kinase M2 regulates Hif-1alpha activity and IL-1beta induction and is a critical determinant of the Warburg effect in LPS-activated macrophages. Cell Metab. 2015;21:65–80.PubMedPubMedCentralCrossRef Palsson-McDermott EM, Curtis AM, Goel G, Lauterbach MA, Sheedy FJ, Gleeson LE, et al. Pyruvate kinase M2 regulates Hif-1alpha activity and IL-1beta induction and is a critical determinant of the Warburg effect in LPS-activated macrophages. Cell Metab. 2015;21:65–80.PubMedPubMedCentralCrossRef
53.
Zurück zum Zitat Zhao H, Han L, Jian Y, Ma Y, Yan W, Chen X, et al. Resveratrol induces apoptosis in human melanoma cell through negatively regulating Erk/PKM2/Bcl-2 axis. Onco Targets Ther. 2018;11:8995–9006.PubMedPubMedCentralCrossRef Zhao H, Han L, Jian Y, Ma Y, Yan W, Chen X, et al. Resveratrol induces apoptosis in human melanoma cell through negatively regulating Erk/PKM2/Bcl-2 axis. Onco Targets Ther. 2018;11:8995–9006.PubMedPubMedCentralCrossRef
54.
Zurück zum Zitat Sun R, Meng X, Pu Y, Sun F, Man Z, Zhang J, et al. Overexpression of HIF-1a could partially protect K562 cells from 1,4-benzoquinone induced toxicity by inhibiting ROS, apoptosis and enhancing glycolysis. Toxicol in Vitro. 2019;55:18–23.PubMedCrossRef Sun R, Meng X, Pu Y, Sun F, Man Z, Zhang J, et al. Overexpression of HIF-1a could partially protect K562 cells from 1,4-benzoquinone induced toxicity by inhibiting ROS, apoptosis and enhancing glycolysis. Toxicol in Vitro. 2019;55:18–23.PubMedCrossRef
55.
Zurück zum Zitat Liang J, Cao R, Wang X, Zhang Y, Wang P, Gao H, et al. Mitochondrial PKM2 regulates oxidative stress-induced apoptosis by stabilizing Bcl2. Cell Res. 2017;27:329–51.PubMedCrossRef Liang J, Cao R, Wang X, Zhang Y, Wang P, Gao H, et al. Mitochondrial PKM2 regulates oxidative stress-induced apoptosis by stabilizing Bcl2. Cell Res. 2017;27:329–51.PubMedCrossRef
56.
Zurück zum Zitat Azoitei N, Becher A, Steinestel K, Rouhi A, Diepold K, Genze F, et al. PKM2 promotes tumor angiogenesis by regulating HIF-1alpha through NF-kappaB activation. Mol Cancer. 2016;15:3.PubMedPubMedCentralCrossRef Azoitei N, Becher A, Steinestel K, Rouhi A, Diepold K, Genze F, et al. PKM2 promotes tumor angiogenesis by regulating HIF-1alpha through NF-kappaB activation. Mol Cancer. 2016;15:3.PubMedPubMedCentralCrossRef
57.
Zurück zum Zitat Luo W, Hu H, Chang R, Zhong J, Knabel M, O'Meally R, et al. Pyruvate kinase M2 is a PHD3-stimulated coactivator for hypoxia-inducible factor 1. Cell. 2011;145:732–44.PubMedPubMedCentralCrossRef Luo W, Hu H, Chang R, Zhong J, Knabel M, O'Meally R, et al. Pyruvate kinase M2 is a PHD3-stimulated coactivator for hypoxia-inducible factor 1. Cell. 2011;145:732–44.PubMedPubMedCentralCrossRef
59.
Zurück zum Zitat Panasyuk G, Espeillac C, Chauvin C, Pradelli LA, Horie Y, Suzuki A, et al. PPARgamma contributes to PKM2 and HK2 expression in fatty liver. Nat Commun. 2012;3:672.PubMedCrossRef Panasyuk G, Espeillac C, Chauvin C, Pradelli LA, Horie Y, Suzuki A, et al. PPARgamma contributes to PKM2 and HK2 expression in fatty liver. Nat Commun. 2012;3:672.PubMedCrossRef
60.
Zurück zum Zitat Li J, Guo C, Wu J. 15-Deoxy--(12,14)-prostaglandin J2 (15d-PGJ2), an endogenous ligand of PPAR-gamma: function and mechanism. PPAR Res. 2019;2019:7242030.PubMedPubMedCentralCrossRef Li J, Guo C, Wu J. 15-Deoxy--(12,14)-prostaglandin J2 (15d-PGJ2), an endogenous ligand of PPAR-gamma: function and mechanism. PPAR Res. 2019;2019:7242030.PubMedPubMedCentralCrossRef
61.
Zurück zum Zitat Heudobler D, Rechenmacher M, Luke F, Vogelhuber M, Pukrop T, Herr W, et al. Peroxisome proliferator-activated receptors (PPAR) gamma agonists as master modulators of tumor tissue. Int J Mol Sci. 2018;19. Heudobler D, Rechenmacher M, Luke F, Vogelhuber M, Pukrop T, Herr W, et al. Peroxisome proliferator-activated receptors (PPAR) gamma agonists as master modulators of tumor tissue. Int J Mol Sci. 2018;19.
62.
Zurück zum Zitat Yun SH, Han SH, Park JI. Peroxisome proliferator-activated receptor gamma and PGC-1alpha in Cancer: dual actions as tumor promoter and suppressor. PPAR Res. 2018;2018:6727421.PubMedPubMedCentralCrossRef Yun SH, Han SH, Park JI. Peroxisome proliferator-activated receptor gamma and PGC-1alpha in Cancer: dual actions as tumor promoter and suppressor. PPAR Res. 2018;2018:6727421.PubMedPubMedCentralCrossRef
64.
Zurück zum Zitat Morita M, Sato T, Nomura M, Sakamoto Y, Inoue Y, Tanaka R, et al. PKM1 confers metabolic advantages and promotes cell-autonomous tumor cell growth. Cancer Cell. 2018;33:355–67 e7.PubMedCrossRef Morita M, Sato T, Nomura M, Sakamoto Y, Inoue Y, Tanaka R, et al. PKM1 confers metabolic advantages and promotes cell-autonomous tumor cell growth. Cancer Cell. 2018;33:355–67 e7.PubMedCrossRef
65.
Zurück zum Zitat Choi AM, Ryter SW, Levine B. Autophagy in human health and disease. N Engl J Med. 2013;368:651–62.PubMedCrossRef Choi AM, Ryter SW, Levine B. Autophagy in human health and disease. N Engl J Med. 2013;368:651–62.PubMedCrossRef
67.
Zurück zum Zitat Yang W, Zheng Y, Xia Y, Ji H, Chen X, Guo F, et al. ERK1/2-dependent phosphorylation and nuclear translocation of PKM2 promotes the Warburg effect. Nat Cell Biol. 2012;14:1295–304.PubMedPubMedCentralCrossRef Yang W, Zheng Y, Xia Y, Ji H, Chen X, Guo F, et al. ERK1/2-dependent phosphorylation and nuclear translocation of PKM2 promotes the Warburg effect. Nat Cell Biol. 2012;14:1295–304.PubMedPubMedCentralCrossRef
68.
Zurück zum Zitat Gupta A, Ajith A, Singh S, Panday RK, Samaiya A, Shukla S. PAK2-c-Myc-PKM2 axis plays an essential role in head and neck oncogenesis via regulating Warburg effect. Cell Death Dis. 2018;9:825.PubMedPubMedCentralCrossRef Gupta A, Ajith A, Singh S, Panday RK, Samaiya A, Shukla S. PAK2-c-Myc-PKM2 axis plays an essential role in head and neck oncogenesis via regulating Warburg effect. Cell Death Dis. 2018;9:825.PubMedPubMedCentralCrossRef
69.
Zurück zum Zitat Kim JW, Gao P, Liu YC, Semenza GL, Dang CV. Hypoxia-inducible factor 1 and dysregulated c-Myc cooperatively induce vascular endothelial growth factor and metabolic switches hexokinase 2 and pyruvate dehydrogenase kinase 1. Mol Cell Biol. 2007;27:7381–93.PubMedPubMedCentralCrossRef Kim JW, Gao P, Liu YC, Semenza GL, Dang CV. Hypoxia-inducible factor 1 and dysregulated c-Myc cooperatively induce vascular endothelial growth factor and metabolic switches hexokinase 2 and pyruvate dehydrogenase kinase 1. Mol Cell Biol. 2007;27:7381–93.PubMedPubMedCentralCrossRef
70.
Zurück zum Zitat Mendez-Lucas A, Li X, Hu J, Che L, Song X, Jia J, et al. Glucose catabolism in liver tumors induced by c-MYC can be sustained by various PKM1/PKM2 ratios and pyruvate kinase activities. Cancer Res. 2017;77:4355–64.PubMedPubMedCentralCrossRef Mendez-Lucas A, Li X, Hu J, Che L, Song X, Jia J, et al. Glucose catabolism in liver tumors induced by c-MYC can be sustained by various PKM1/PKM2 ratios and pyruvate kinase activities. Cancer Res. 2017;77:4355–64.PubMedPubMedCentralCrossRef
Metadaten
Titel
Simvastatin re-sensitizes hepatocellular carcinoma cells to sorafenib by inhibiting HIF-1α/PPAR-γ/PKM2-mediated glycolysis
verfasst von
Jiao Feng
Weiqi Dai
Yuqing Mao
Liwei Wu
Jingjing Li
Kan Chen
Qiang Yu
Rui Kong
Sainan Li
Jie Zhang
Jie Ji
Jianye Wu
Wenhui Mo
Xuanfu Xu
Chuanyong Guo
Publikationsdatum
01.12.2020
Verlag
BioMed Central
Erschienen in
Journal of Experimental & Clinical Cancer Research / Ausgabe 1/2020
Elektronische ISSN: 1756-9966
DOI
https://doi.org/10.1186/s13046-020-1528-x

Weitere Artikel der Ausgabe 1/2020

Journal of Experimental & Clinical Cancer Research 1/2020 Zur Ausgabe

Umsetzung der POMGAT-Leitlinie läuft

03.05.2024 DCK 2024 Kongressbericht

Seit November 2023 gibt es evidenzbasierte Empfehlungen zum perioperativen Management bei gastrointestinalen Tumoren (POMGAT) auf S3-Niveau. Vieles wird schon entsprechend der Empfehlungen durchgeführt. Wo es im Alltag noch hapert, zeigt eine Umfrage in einem Klinikverbund.

CUP-Syndrom: Künstliche Intelligenz kann Primärtumor finden

30.04.2024 Künstliche Intelligenz Nachrichten

Krebserkrankungen unbekannten Ursprungs (CUP) sind eine diagnostische Herausforderung. KI-Systeme können Pathologen dabei unterstützen, zytologische Bilder zu interpretieren, um den Primärtumor zu lokalisieren.

Sind Frauen die fähigeren Ärzte?

30.04.2024 Gendermedizin Nachrichten

Patienten, die von Ärztinnen behandelt werden, dürfen offenbar auf bessere Therapieergebnisse hoffen als Patienten von Ärzten. Besonders gilt das offenbar für weibliche Kranke, wie eine Studie zeigt.

Adjuvante Immuntherapie verlängert Leben bei RCC

25.04.2024 Nierenkarzinom Nachrichten

Nun gibt es auch Resultate zum Gesamtüberleben: Eine adjuvante Pembrolizumab-Therapie konnte in einer Phase-3-Studie das Leben von Menschen mit Nierenzellkarzinom deutlich verlängern. Die Sterberate war im Vergleich zu Placebo um 38% geringer.

Update Onkologie

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