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

Open Access 01.12.2020 | Research

Mitophagy promotes sorafenib resistance through hypoxia-inducible ATAD3A dependent Axis

verfasst von: Hong Wu, Tao Wang, Yiqiang Liu, Xin Li, Senlin Xu, Changtao Wu, Hongbo Zou, Mianfu Cao, Guoxiang Jin, Jinyi Lang, Bin Wang, Baohua Liu, Xiaolin Luo, Chuan Xu

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

Abstract

Background

The identification of novel targets for recovering sorafenib resistance is pivotal for Hepatocellular carcinoma (HCC) patients. Mitophagy is the programmed degradation of mitochondria, and is likely involved in drug resistance of cancer cells. Here, we identified hyperactivated mitophagy is essential for sorafenib resistance, and the mitophagy core regulator gene ATAD3A (ATPase family AAA domain containing 3A) was down regulated in hypoxia induced resistant HCC cells. Blocking mitophagy may restore the sorafenib sensitivity of these cells and provide a new treatment strategy for HCC patients.

Methods

Hypoxia induced sorafenib resistant cancer cells were established by culturing under 1% O2 with increasing drug treatment. RNA sequencing was conducted in transfecting LM3 cells with sh-ATAD3A lentivirus. Subsequent mechanistic studies were performed in HCC cell lines by manipulating ATAD3A expression isogenically where we evaluated drug sensitivity, molecular signaling events. In vivo study, we investigated the combined treatment effect of sorafenib and miR-210-5P antagomir.

Results

We found a hyperactivated mitophagy regulating by ATAD3A-PINK1/PARKIN axis in hypoxia induced sorafenib resistant HCC cells. Gain- and loss- of ATAD3A were related to hypoxia-induced mitophagy and sorafenib resistance. In addition, ATAD3A is a functional target of miR-210-5p and its oncogenic functions are likely mediated by increased miR-210-5P expression. miR-210-5P was upregulated under hypoxia and participated in regulating sorafenib resistance. In vivo xenograft assay showed that miR-210-5P antagomir combined with sorafenib abrogated the tumorigenic effect of ATAD3A down-regulation in mice.

Conclusions

Loss of ATAD3A hyperactivates mitophagy which is a core event in hypoxia induced sorafenib resistance in HCC cells. Targeting miR-210-5P-ATAD3A axis is a novel therapeutic target for sorafenib-resistant HCC.
Begleitmaterial
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1186/​s13046-020-01768-8.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
MIR
Myocardial ischemia reperfusion injury
AS
Ankylosing spondylitis
CRC
Colorectal cancer
PINK1
PTEN–induced putative kinase protein 1
HCC
Hepatocellular carcinoma
BNIP3L/NIX
Bcl-2/adenovirus E1B 19-kDa interacting protein 3 like
FUNDC1
FUN14 domain-containing protein 1
OS
Overall survival
HIF-1α
Hypoxia-inducible factor 1α
TOMM20
Mitochondrial import receptor subunit TOM20 homolog
TOMM70
Mitochondrial import receptor subunit TOM70 homolog
LC3
Microtubule-associated protein1light chain3
ABC20
ABC-type transport system permease protein
ABCB1
ATP-binding cassette subfamily B member 1
ABCC1
Multidrug resistance-associated protein 1
LRP
Lipoprotein Receptor-Related Protein
MRP1
Multidrug resistance-associated protein 1
IHC
Immunohistochemistry
qRT-PCR
Quantitative real-time PCR

Background

Mitophagy is a mitochondrial form of autophagy that is critical for mitochondrial quality control and homeostasis [1, 2]. In response to various stimuli, mitochondria undergo self-depolarization and the damaged organelles are identified by autophagosomes which then fuse with lysosomes to complete the degradation process [3, 4]. Defective or excessive mitophagy is a pathological factor in chronic diseases including parkinson’s disease (PD), diabetes, myocardial ischemia reperfusion (MIR) injury, ankylosing spondylitis (AS) and cancers [5]. Several mitophagic pathways have been identified that counteract therapy-induced mitochondrial damage [5, 6] . Guo et al. reported that reversing mitophagy inhibition by the PIK3CA/AKT1/MTOR /RPS6KB1 pathway reduced the risk of colorectal cancer (CRC) development [7]. Katreddy et al. found that EGFR down-regulation cleared ovarian cancer cells by activating the mitophagy/mTORC2/Akt axis [8]. Furthermore, mitophagy suppression via knocking down key mitophagy receptors such as PINK1, FUNDC1 or AMBRA1 could chemo-sensitize cancer cells [9, 10]. The E3 ubiquitin-ligase ARIH1 and not Parkin mediates PINK1-induced mitophagic responses to cisplatin/etoposide in breast and lung adenocarcinomas [11]. In fact, the predominance of certain mitophagy receptors or mediators in specific cancer subtypes is a decisive factor in therapeutic resistance via mitochondrial clearance [6, 12], and the underlying mechanisms are still unclear.
The ubiquitin-dependent PINK1/Parkin pathway is the most widely studied mammalian mitophagy cascade. PINK1 is an outer mitochondrial membrane (OMM) Ser/Thr kinase that is stabilized upon mitochondrial depolarization, and induces Parkin E3 activity and recruits Parkin by phosphorylating it at Ser-65 [13]. Recent studies has shown that ectopic expression of PINK1 plays a dual role in cancer development and drug treatment [14]. Wang et al. reported that the induction of PINK1/Parkin-mediated mitophagy sensitized tongue cancer cells to ZnO NPs [15]. In contrast, Villa et al. showed that ARIH1/HHARI triggered PINK1-dependent mitophagy in the breast and lung adenocarcinomas and protected them from chemotherapy-induced death [11]. In the recent study, our group showed that the AAA domain containing 3A (ATAD3A) protein could prevent excessive accumulation of PINK1 and therefore inhibited unnecessary mitophagy in stem and progenitor cells [16]. As an up-regulator of PINK1, ATAD3A spans the inner mitochondrial membrane with its two terminal domains in the outer membrane and the matrix [17]. However, little is known regarding its role in cancer development and drug sensitivity under hypoxia microenvironment of solid tumors.
microRNAs are members of a large class of endogenous small noncoding RNAs that control gene expression and regulate a wide array of biological processes by binding to the 3′-untranslated regions (3′-UTR) directly to regulate target mRNA expression, and eventually promote target mRNA degradation or translational inhibition [18]. Several studies have reported miR-210, as one of the highly up-regulated miRNAs in hypoxic cells that is involved in numerous biological processes of the human body involving regulating mitochondrial metabolism, promoting the angiogenesis, proliferation and apoptosis. miR-210 has two versions, miR-210-3P and miR-210-5P. miR-210-3p is the guide-strand that integrates into the RISC (RNA induced silencing complex), whereas miR-210-5p is the passenger-strand that is inactivated through degradation [19, 20]. miR-210-5P is up-regulated in several malignant tumors that are associated with a variety of functionally important targets involved in cancers [21]. However, whether miR-210-5p could regulate mitophagy to participate in drug-sensitivity remains to be examined in depth.
Hepatocellular carcinoma (HCC) is the sixth most common fatal malignancy and the major cause of cancer-related deaths worldwide [22]. Most patients are diagnosed at the advanced stage which is highly recalcitrant to the current therapies [23]. The multi-target tyrosine kinase inhibitor sorafenib is the FDA-approved first-line systematic therapy for advanced HCC patients, which increased median survival from 7.9 to 10.7 months [24]. It blocks tumor cell proliferation and angiogenesis by inhibiting the Rad/Mitogen-activated protein (MAP)/extracellular signal-regulated kinase (ERK)/ MEK signaling cascade, as well as the kinase activity of vascular endothelial growth factor receptor (VEGFR) and platelet-derived growth factor receptor (PDGFR)-β. However, almost all HCC patients have developed sorafenib resistance within a few months [25]. Since the drug targets several signaling pathways, the tumors acquire resistance through different mechanisms, such as activation of compensatory signaling cascades and a hypoxic microenvironment [26]. It is crucial to understand the precise molecular mechanisms underlying sorafenib resistance in order to identify novel therapeutic targets and improve the clinical outcome of HCC patients. Although, sorafenib-induced autophagy contributes to drug resistance, it is unknown whether hyperactivated mitophagy is also involved in sorafenib resistance in HCC [2729]. Regarding hypoxia is a major stimulus of mitophagy and also an important cause of sorafenib resistance [3032], our objective is to determine the potential relationship between mitophagy and hypoxia-induced sorafenib resistance in HCC. We found that hypoxia induced sorafenib resistance in hepatoma cells was accompanied by hyperactivated mitophagy and a downregulation of ATAD3A expression. Thus, ATAD3A is a crucial mediator of hypoxia induced mitophagy signaling in HCCs and a novel therapeutic target for reversing sorafenib resistance.

Materials and methods

Patients and tissue microarray

 Liver cancer tissues and paried normal tissues were collected form the First Affiliated Hospital of Third Military Medical University and Shanghai Biochip Company Ltd (Shanghai, China). The informed consent was obtained from all patients. 

Cell culture

Human hepatoma cell lines Huh7 and LM3 were obtained from the American Type Culture Collection (ATCC) and authenticated by the Cell Bank of Type Culture Collection of Chinese Academy of Science. The cells were maintained under recommended conditions. For hypoxia treatment, the cells were either cultured in a sealed hypoxia chamber (Thermo fisher, Inc.) containing 1% O2, 5% CO2 and 94% N2, or treated with 100 mM CoCl2 for 24 or 48 h. Sorafenib-resistant Huh7 (Huh7-SR) and LM3 (LM3-SR) cells were enriched by steadily increasing the drug dose, while hypoxia-induced sorafenib resistant cell lines (Huh7-H-SR and LM3-H-SR) were established by culturing under 1% O2 with increasing drug doses.

Virus production

The pLVX-CMV-EGFP-3FLAG-PGK-Puro lentiviral vector expressing full-length human ATAD3A was purchased from SunBio (Shanghai. China). The pMAGic2.1-CMV-HygroR-U6 shRNA lentivirus vector purchased from SunBio (Shanghai. China), and the shRNA sequences are shown in Additional file 1. Human HEK293T cells (American Type Culture Collection) were cultured in 6-well plates till ~ 70% confluent, and co-transfected with 2 μg overexpression or knockdown virus vector, 1 μg pMD2.G and 1 μg psPAX2 lentivirus packaging vectors using Lipofectamine 2000 (Invitrogen) according to the manufacturer’s protocol. The cells were maintained in high-glucose DMEM containing10% FBS, 2 mM glutamine, and100 units/ml penicillin and streptomycin. The supernatants with virus were harvested twice at 48 h and 72 h, filtered through a 0.45 μm syringe filter and frozen in liquid nitrogen.

Target cell transduction

HCC cells were cultured in complete DMEM, and 1.5 × 105 cells were mixed with 450 μl virus-containing supernatant in the presence of 4 μg/ml polybrene (Sigma). The cells were seeded into a 6-well plate, and the medium was changed 12 h post-infection. After 48 h, the infected cells were trypsinized and seeded into 10 cm culture dish with 4 μg/ml puromycin (Thermo Fisher Scientific). The stably transduced cells were selected over 48 h, and harvested 6 days post-infection to determine knockdown efficiency.

Western blotting

Total protein was extracted from cancer cells using Mammalian Protein Extraction Buffer (P0013, Beyotime, Beijing, China) supplemented with protease inhibitor cocktail (87786, ThermoFisher, USA). The mitochondrial fractions were separated using Mitochondria Isolation Kit (number: SM0020, Solarbio, Beijing, China) according to the manufacturer’s protocol, and the protein was extracted as above. Equal amounts of protein lysates were separated by SDS-PAGE gel and electro-transferred to PVDF membrane (Millipore, USA). After blocking in 5% milk-PBST for 2 h at 37 °C, the membranes were incubated overnight with primary antibodies (Additional file 2) at 4 °C and with secondary antibodies at 37 °C for 2 h. The positive bands were visualized with Immobilon Western Chemiluminescent HRP Substrate detection reagent (Millipore, USA), and acquired using a ChemiDoc™ imaging System (Bio-Rad, USA).

Quantitative real-time PCR

Total RNA was extracted from cancer cells using Trizol Reagent (Invitrogen, USA), and qRT-PCR was performed using SYBR Prime Script RT-PCR kit (TaKaRa, Japan) on a Rotor-Gene 6000 real-time genetic analyzer (Corbett Life Science, USA). The primer sequences and the product sizes are listed in Additional file 3. Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) was used as the internal control. The PCR conditions were as follows: denaturation at 95 °C for 2 mins, followed by 40 cycles of amplification and quantification (95 °C for 5 s, 55 °C–57 °C for 30 s), and melting curve (55 °C–95 °C, with 0.5 °C increment each cycle). Each sample was tested in triplicates.

Apoptosis assay

For apoptosis assays, the suitably treated cells were washed twice with cold PBS, and resuspended in binding buffer at the density of 1 × 106 cells/ml, and distributed into 500 μl aliquots (1 × 105) in 2 ml tubes. 5 μL of Annexin V-FITC were added to each tube. Cells were incubated at room temperature (25 °C) for 15 min in a dark environment, and then analyzed on a FACS Calibur (BD Biosciences, USA), and results were calculated using Cell Quest software (BD Biosciences, USA).

Immunofluorescence assay

HCC cells were washed, fixed with 5% paraformaldehyde (PFA) and permeabilized in 0.1% Triton X-100. After incubating overnight with monoclonal mouse anti-HIF-1α antibody (Cell signaling technology, #79233) or monoclonal rabbit anti-ATAD3A antibody (Invitrogen, PA5–03671) at 4 °C, the cells were probed with fluorescein isothiocyanate (FITC)-labeled anti-mouse IgG or Cy3-labeled anti-rabbit IgG secondary antibodies (Santa Cruz Biotechnology, Santa Cruz, USA). The nuclei were counterstained with Hochest 33258 and observed under laser confocal scanning microscopy (Leica TCS-SP5, Germany).

Luciferase reporter assay

The 3′-UTR region of human ATAD3A was amplified by PCR from genomic DNA and cloned downstream to the firefly luciferase coding region in the pMIR-REPORTTM plasmid. The 293 T cells were seeded in 96-well plates, and co-transfected with 100 ng/ml reporter plasmid and 50 nM miR-210-5P or NC mimics using Lipofectamine 2000 24h late. Luciferase activity were measured after 72 h using the Dual-Glo Luciferase Assay System (Promega, Madison, WI, USA). All experiments were performed in triplicates.

Cell proliferation assay

Cells were seeded in 96-well plates at the density of 5000 cells/well. After treating with 5.13 μM (Huh7) or 7.92 μM (LM3) sorafenib for varying durations (0 h,12 h,24 h,48 h and 72 h), the cells were incubated for 2 h with 0.5 mg/ml 3-(4, 5-dimethyl-thiazol-2-yl)-2,5-diphenyl-tetrazolium bromide (MTT) (Sigma-Aldrich) in serum free medium. To measure the IC50 of different cell lines, 10− 3,10− 2,10− 1,100,101 and 102μM sorafenib were added in the 96-well plates for 72 h, then treated same as above. The optical densities were measured at 450 nM spectral wavelength using the microplate reader (ELx800; Bio-Tek Instruments, Inc.).

Colony formation assay

For colony formation assay, 5 × 102 cells were seeded in 6-well plates with sorafenib 5.13 μM (Huh7) or 7.92 μM (LM3) as appropriate. The conditioned medium was removed 3 days later and replaced by medium containing 10% serum for 2 weeks. The ensuing colonies were stained by crystal violet and counted. The experiment was performed thrice.

Mitophagy assay

Western blotting was used to test LC3 mobility shift by analyzing the expression levels of LC3 I/II, and mitochondrial marker protein TOMM20 and TOMM70. Subcellular localization of mitophagy bodies was tracked by transmission-electron-microscopy (TEM, JEM-1230, Japan). The cells were fixed in cooling 2% glutaraldehyde and 0.1 M cacodylate buffer at 4 °C overnight, then exposed to phosphate buffer containing 1% osmium tetroxide for 1 h. Following dehydrated in different concentrations of acetone, these cells were infiltrated and embedded into Epon. The embedded cells were sectioned and stained with 3% uranyl acetate and lead citrate. Mitochondrial content was analyzed by Mito-Tracker staining (number: C1049, Beyotime Biotechnology, Shanghai, China) according to the manufacturer’s protocol and then analysed by FACS (BD Biosciences, USA), and results were calculated using Flow Jowo software (BD Biosciences, USA).

Lactate measurement

The suitably treated cells were seeded in 6-well plates in triplicate at the density of 5*104 cells/well. After culturing for 2 days, the number of viable cells was counted, and the culture medium was collected. Lactate levels were detected in the latter using a specific analytical kit (Nanjing Jian cheng Bioengineering Institute, China), and normalized to cell number and calculated as relative units per cell.

Immunohistochemistry (IHC)

IHC was performed on tissue array slides as previously described. The tissues were probed with rabbit anti-human ATAD3A (Invitrogen, PA5–03671 1:200) and rabbit anti-human HIF-1α (abcam, ab51608, 1:200) antibodies, and counterstained with hematoxylin (Sigma). The staining intensity (negative = 0, weak = 1, moderate = 2, or strong = 3) and the percentage of positively stained cells (<5%=0, 5% to < 25% = 1, 25 to 50% = 2, > 50 to < 75% = 3, ≥75% = 4) were scored independently by two pathologists. The staining index was calculated by multiplying the intensity score with percentage score, and the samples were classified as negative/low expression or positive/high expression accordingly.

RNA sequencing and bioinformatic data analysis

LM3-shControl and LM3-shATAD3A cells were generated as before and total RNA was extracted. RNA libraries were constructed using a TruSeq Stranded mRNA LT Sample Prep Kit (Illumina, San Diego, CA, USA) according to the manufacturer’s instructions, and sequenced on an Illumina platform (HiSeqTM 2500 or Illumina HiSeq X Ten) into 125 bp/150 bp paired-end reads by OE Biotech (Shanghai, China). The differentially expressed genes (DEGs) were identified using Cuff-diff with p value < 0.05 and fold change > 2 as the criteria. Gene set enrichment analysis was performed using software.​broadinstitute.​org/​gsea. Heatmaps were generated with the heatmap package of R program.

Tumorigenicity analysis in vivo

Female BALB/c (nu/nu) athymic nude mice, 5 weeks of age, were purchased from hfkbio Inc. Mice were maintained in specific pathogen-free conditions: 20–24 °C, 12/12 h of dark/light cycle, 60 ± 5% of humidity, and plastic cage (3–4 mice/cage). Bedding materials were changed every week, and environmental enrichment was done with sterile materials. All animal experiments were approved by the Institutional Animal Care and Use Committee of the Medicine University of Electronic Science and Technology of China’s. Control and miR-210-5P antagomir-transfected Huh7-shControl and Huh7-shATAD3A cells were inoculated subcutaneously (n = 5 each, 2 × 106 cells per mouse) in the right flank of 4–6 week old BALB/c athymic female nude mice. Once the tumors grew to approximately 100mm3 (around 5 days), the mice were intraperitoneally injected with sorafenib tosylate (10 mg/kg) daily for a week, and monitored every 4 days for the appearance of subcutaneous tumors. We conducted the sacrifice of mice at 28 days, placed the mice in the chamber and introduced 100% carbon dioxide. After we removed each tumor, we maintained the carbon dioxide flow for a minimum of 1 min after respiration ceases. The tumor weight and tumor volume (TV; mm3) were calculated. Tumor volume was calculated as d2 × D/2 (d and D represent the shortest and the longest diameters, respectively). All animal experiments were performed in accordance with the guidelines of the ARRIVE reporting guidelines.

Statistical analysis

Statistical analyses were performed using GraphPad Prism 6.0 (GraphPad, La Jolla, CA, USA). Two tailed unpaired t-test or one-way ANOVA were used to compare two or multiple groups. Survival curves were plotted using the Kaplan–Meier method and compared by the log-rank test. All data was presented as mean ± SD or mean ± SEM. P < 0.05 was considered statistically significant.

Results

Hyperactivated mitophagy plays a vital role in hypoxia-induced sorafenib resistance

As shown in Supplementary Fig. S1a-1d, HCC cells cultured under hypoxia showed significantly lower apoptosis rates and better cell viability response to sorafenib treatment in a time-dependent manner. To further analyze the role of hypoxia in sorafenib resistance, we established sorafenib resistant Huh7 (Huh7-SR) and LM3 (LM3-SR) cell lines, as well as the corresponding hypoxia-induced sorafenib resistant cell lines (Huh7-H-SR and LM3-H-SR) in the presence of increasing doses of sorafenib under 1% O2 culture condition (Fig. 1a-c). Compared to the respective control, Huh7-H-SR and LM3-H-SR cells showed increased levels of HIF-1α and drug resistance-related genes, including ABC20, ABCB1, ABCC1 and LRP (Supplementary Fig. S1e). Interestingly, extensive mitophagy was observed in Huh7-H-SR cells compared to the untreated controls, hypoxia treated Huh7 and Huh7-SR cells (Fig. 1d). Consistent with this, the Huh7-H-SR cells showed highest level of PINK1 expression, along with accumulation of PINK1, Parkin and LC3-II in the mitochondrial fraction (Fig. 1e). We also characterized a higher ubiquitination of multiple mitochondrial proteins via Parkin in Huh7-H-SR cells (Fig. 1e). Hypoxia-induced sorafenib resistant Huh7 and LM3 cells lines significantly upregulated PINK1, LC3-II, HIF-1α, FUNDC1 expression, and downregulated TOMM20 and ATAD3A expression (Fig. 1f). Furthermore, TCGA analysis results showed that PINK1 levels were positively correlated with that of ABCB1 and ABCG2 in HCC patients (Fig. 1g-h), and HIF-1α was positively correlated with ABCC1 and negatively with TOMM20 and TOMM70 (Supplementary Fig. S1f-1 h). These results indicated that activated mitophagy is the likely cause of hypoxia-induced sorafenib resistance. In agreement with this, the mitophagy inhibitor 3MA sensitized Huh7-H-SR cells to sorafenib, resulting in a significant decrease in colony yield and proliferation rates (Fig. 1i-j). Furthermore, PINK1 knockdown in Huh7-H-SR cells also sensitized the cells to sorafenib (Fig. 1k-l). Taken together, hyperactivated mitophagy plays a pro-survival role in the hypoxia-induced sorafenib resistant HCC cells.

Hypoxia-induced sorafenib resistance is dependent on ATAD3A-mediated mitophagy

The mitochondria transmembrane protein ATAD3A was significantly downregulated in the Huh7-H-SR and LM3-H-SR cells compared to the respective controls (Supplementary Fig. S2a). To determine the biological relevance of ATAD3A in HCC, we established stable ATAD3A-overexpressing and knockdown Huh7 and LM3 cell lines (Fig. 2c and Supplementary Fig.S2b-2c). Mitophagy was hyperactivated in the shATAD3A LM3 cells compared to the control (Fig. 2a), which paralleled a marked increase in the polyubiquitination and accumulation of global mitochondrial proteins as seen in the hypoxia-induced sorafenib resistant cells (Fig. 2b). In addition, PINK1 and LC3-II were significantly upregulated, and TOMM20, TOMM70 were downregulated following ATAD3A knockdown (Fig. 2c). Transcriptome sequencing analysis of the control and shATAD3A LM3 cells identified 319 upregulated and 302 downregulated genes in the latter (Supplementary Fig. S2i-2j). Gene set enrichment analysis (GSEA) (Fig. 2f) showed that the shATAD3A cells were commonly enriched with drug resistance genes (NES = -1.939, P = 0.00828 for GEFITINIB resistance; NES = 1.8435; P = 0.02 for TAMOXIFEN resistance) (Fig. 2d, e). The expression levels of these genes were validated by qRT-PCR (Fig. 2g, h). We also confirmed that the expression level of these genes in LM3-H-SR were similar to that in sh-ATAD3A cells compared to shControl (Supplementary Fig. S2f). In addition, shATAD3A and hypoxia-induced sorafenib resistantLM3 cells desensitized the HCC cells to sorafenib, resulting in a significant decrease in apoptotic index (Fig. 2i and Supplementary Fig. S2g-2 h), whereas ATAD3A overexpression augmented the sorafenib-induced cell death (Fig. 2j, k and Supplementary Fig. S2d, 2e). Loss of ATAD3A downregulated the pro-apoptotic PARP, Bax and Cleaved-Caspase 3 expression (Fig. 2l), decreased mitochondrial mass (Fig. 2m), and increased lactate production (Supplementary Fig. S3a-3b) in Huh7 and LM3 cells. Taken together, ATAD3A plays a vital role in mitophagy and sorafenib insensitivity in HCC cells.

ATAD3A participates in hypoxia-induced mitophagy and sorafenib sensitivity

To further determine the biological significance of hypoxia and ATAD3A expression in HCC cells, we introduced ectopic ATAD3A in the hypoxia cultured cells. As shown in Fig. 3a, hypoxia-induced mitophagy was abrogated by ATAD3A overexpression, which also upregulated TOMM20 and TOMM70 in Huh7 and LM3 cells (Fig. 3b). Ectopic ATAD3A also restored the mitochondrial mass (Fig. 3c, d) and decreased lactate concentration (Supplementary Fig. S3c, 3d) upon hypoxia-treated LM3 and Huh7 cells. Under hypoxia condition, cancer cells were insensitive to sorafenib treatment, while, gain of ATAD3A expression mitigated the insensitivity as indicated by the fewer colonies (Fig. 3e, f) and increased apoptotic index in LM3 cells (Fig. 3g, h). Western blot confirmed that PARP, Bax and Cleaved-Caspase3 were downregulated in hypoxia cells which were restored in ATAD3A overexpression cell under sorafenib treatment. Furthermore, we confirmed that ATAD3A expression restored sorafenib sensitivity in hypoxia treated Huh7 and LM3 cells via the apoptotic related proteins (Fig. 3i).

ATAD3A is a direct functional target of hypoxia-induced mir-210-5p

To determine the mechanism underlying hypoxia-mediated regulation of ATAD3A expression, we performed a systematic bioinformatics analysis of gene-gene interaction networks based on mutations, copy number alterations, mRNA expression profiles, and protein expression profiles using the STRING database (http://​string-db.​org/​cgi/​network). ATAD3A and the hypoxia-responsive transcriptional factor HIF-1α are located in a network containing 40 nodes (Fig. 4a), suggesting a potential link between HIF-1α signaling and ATAD3A. Furthermore, in situ HIF-1α protein expression in 85 HCC tissues was negatively correlated with that of ATAD3A (Fig. 4b). As shown in Fig. 4c and Supplementary Fig. S4a, hypoxia treatment significantly down regulated ATAD3A expression in hepatoma cells. In addition, both 1% O2 and CoCl2 induced HIF-1α expression and downregulated ATAD3A by western blot in Huh7 and LM3 cells (Fig. 4d), and decreased ATAD3A expression in Huh7 cells as a time-dependent manner (Supplementary Fig. S4b). In addition, we found a loss of ATAD3A gene expression in 1% O2 and CoCl2 treated Huh7 and LM3 cells (Fig. 4e-f and Supplementary Fig. S4c). Bioinformatics analysis identified hsa-miR-210 and hsa-miR-193b as the most hypoxia-responsive miRNAs (Supplementary Fig. S4d), and ATAD3A as the potential target gene of miR-210-5P (Fig. 4g). Consistent with this, 1% O2 and CoCl2 increased miR-210-5p expression after 24 h (Fig. 4h and Supplementary Fig. S4e). Furthermore, miR-210-5P suppressed the expression of reporter gene carrying the wild-type but not mutant 3′-UTR of human ATAD3A gene (Fig. 4j, k). Hepatoma cells transfected with miR-210-5P mimic significantly inhibited ATAD3A expression, whereas miR-210-5p inhibitor had the opposite effect (Fig. 4i, l and Supplementary Fig. S4f). These findings indicated that ATAD3A is a direct target of the hypoxia induced miR-210-5P. This was confirmed by treating hepatoma cells with CoCl2 and/or the miR-210-5P inhibitor, which showed that hypoxia-induced downregulation of ATAD3A was abrogated by blocking miR-210-5P (Fig. 4m-o). Taken together, inhibition of ATAD3A under hypoxia is dependent on miR-210-5P.
MiR-210 is a classic hypoxia-induced miRNA that can be activated by HIF-1α. Here, we found miR-210-5P mimic induced mitophagy in HCC cells that were similar to CoCl2 treatment. The hyper-activated mitophagy induced by CoCl2 could be reversed by miR-210-5P inhibitor transfection (Fig. 5a). Furthermore, miR-210-5P mimic downregulated TOMM20 and TOMM70 expression (Fig. 5b), decreased mitochondrial mass by flow cytometry (Fig. 5c), upregulated PINK1 expression level (Fig. 5e), and increased lactate production (Supplementary Fig. S3e-h) in the Huh7 and LM3 cells. Nevertheless, these could be neutralized by the miR-210-5P inhibitor in CoCl2-treated cells. As shown in Fig. 5d-h, depletion of miR-210-5P significantly increased the percentage of apoptotic cells in the presence of sorafenib, while the miR-210-5P mimic decreased apoptosis in the sorafenib-treated cells. The expression levels of the pro-apoptotic proteins differed accordingly in the miR-210-5P mimic/inhibitor-transfected cells (Fig. 5i). In conclusion, hypoxia-induced mitophagy is partially mediated by miR-210-5P.

Clinical significance of ATAD3A expression in HCC and the miR-210-5P-ATAD3A axis in exnografts formation in vivo

To investigate the expression level of ATAD3A in HCC tissues and their paired normal tissues. We generated a tumor sample tissue array containing 135 hepatocellular carcinoma samples with survival information and 85 cases with paired normal tissues to determine ATAD3A expression by IHC. Results showed that ATAD3A expression in HCC patients was heterogeneous, ranging from completely absent to strongly expressed and the representative images of hepatocellular carcinoma samples with strong, moderate or low ATAD3A expression were shown in Fig. 6a. ATAD3A expression level was lower in cancerous liver tissues compared to the adjacent normal tissues (Fig. 6b-c), and lower expression of ATAD3A was associated with higher-grade HCC patients (Fig. 6d). 14% of hepatocellular carcinoma samples had strong ATAD3A expression, whereas percentages of moderate and no ATAD3A expression were 21% and 65%, respectively (Fig. 6e). Kaplan–Meier analysis showed that lower ATAD3A expression in HCC was correlated closely with poor overall survival (OS) of the patients (P = 0.0184; Fig. 6f). Western blot analysis also showed that ATAD3A had a significantly lower expression in 6 HCC tissues compared to their paired normal tissues (Fig. 6g). Taken together, these data indicated that ATAD3A is down regulated in HCCs and indicative of poor clinical outcome of HCC patients. To further determine the tumorigenicity of sh-ATAD3A and whether miR-210-5P antagomir could attenuate the tumor progression role of sh-ATAD3A under sorafenib treatment in vivo. Huh7-shcontrol and Huh7-shATAD3A cells that have been transfected with or without antagomir of miR-210-5p construct were subcutaneously implanted into the right flank of nude mice (Fig. 6h). Results showed that sh-ATAD3A in Huh7 cells significantly increased the tumor weight and tumor volume compared to the control group in the presence of sorafenib treatment, and miR-210-5P antagomir reduced the tumor size and tumor volume both in control and sh-ATAD3A groups (Fig. 6i-l). Collectively, these data provide evidence for an important inverse regulating relationship between ATAD3A and miR-210-5P in vitro and in vivo. Taken together, our findings point to an inverse regulatory relationship between ATAD3A and miR-210-5P in mediating mitophagy in the sorafenib-resistant HCC cells under hypoxia (Fig. 6m).

Discussion

Hypoxia is a hallmark of solid tumors and is the consequence of absent or abnormal vasculature in the tumor microenvironment. It is essential for tumor progression, angiogenesis, metastasis, invasion, immune escape and chemoresistance [33]. Various studies showed that sorafenib could stabilize HIF-1α expression under hypoxia and increase the expression of HIF-1α targets like MDR1, GLUT-1, VEGF and miRNAs (e.g. miR-210), and the whole progress played vital roles in cancer biology [3436]. In addition, evidence indicated that hypoxia-induced mitophagy mainly depends on BNIP3/NIX and the FUNDC1 pathway [37, 38]. In the present study, we found that hypoxia induced mitophagy also dependent on the loss of ATAD3A, which could increase PINK1 accumulation in the outer mitochondrial membrane.
ATAD3A located on the mitochondrial membrane which regulated fragmentation, fission, protein transport and other aspects of mitochondrial physiology [3942]. ATAD3A deletion triggered mitophagy through the PINK1/Parkin pathway in the hematopoietic system and induced cisplatin sensitivity by increasing mitochondrial fragmentation. In our present study, hypoxia-induced sorafenib resistance in HCC cells was associated with lower ATAD3A expression and upregulation of BNIP3/NIX, and loss of ATAD3A increased sorafenib insensitivity. Since PINK1 deletion nearly completely abrogated sorafenib resistance of Huh7 cells under hypoxia, we hypothesized that ATAD3A played a vital role in hypoxia-induced sorafenib resistance through the mitophagic pathway. Under both basal conditions and times of abnormal stress, the process of mitophagy was to prevent the accumulation of dysfunctional mitochondria and preserved mitochondrial homeostasis [43]. While, how mitophagy affected drug sensitivity was still confusing. The removal of damaged mitochondria through mitophagy could dampen the toxicity of chemotherapy [44]. Mitophagy is also described as a possible mechanism for preventing ROS production [45]. And cancer cells were likely to be more sensitive to the additional oxidative damage promoted by ROS [4648]. Therefore, we presumed that hypoxia induced hyperactivated mitophagy may decrease ROS production and then result in sorafenib insensitivity.
HIF-1α is a key transcriptional factor that allows cells and tissues to adapt to hypoxia. Bioinformatics analysis predicted ATAD3A as a direct target of miR-210-5P, the predominant miRNA induced under hypoxia in multiple solid tumors and a surrogate marker for tumor hypoxia [49]. It promoted cancer cell proliferation, metabolism, angiogenesis, metastasis, invasion, chemo-resistance, and correlated with patient prognosis [5053]. Mitochondrial homeostasis was crucial for cellular survival during hypoxia [52], and in line with this, miR-210-5P blockade reversed hypoxia-induced down-regulation of ATAD3A and sensitized the HCC cells to sorafenib by promoting mitophagy.

Conclusions

In summary, the lack of reliable and robust predictive biomarkers of sorafenib sensitivity and treatment response has hindered the development of personalized therapy in HCC. Loss of ATAD3A increased HCC tumor growth in the presence of sorafenib treatment. However, miR-210-5P antagomir abolished the tumorigenic effect of ATAD3A knockdown. Thus, ATAD3A is an effective target for overcoming resistance to sorafenib and also a predictive marker for sorafenib response and clinical outcomes. Our findings also point to the role of ATAD3A-PINK1-Parkin signaling pathway in hypoxia-induced sorafenib resistance in HCC. Further investigation is warranted to examine the feasibility of miR-210-5P inhibition or ectopic ATAD3A expression combined with sorafenib as a novel therapeutic strategy against HCC. In addition, additional response predictors need to be assessed in order to identify patient subgroups most likely to benefit from sorafenib, achieve optimal risk-to-benefit ratio and maximize the treatment efficacy for each patient.

Acknowledgements

We thank the flow cytometry core facility at Third Military Medical University for flow cytometry analysis.
This study was approved by the Institute Sichuan Cancer Center School of Medicine University of Electronic Science and Technology of China.
All authors have consented to publication of the results presented in this manuscript.

Competing interests

The authors declare no competing financial interests.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. 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 in a credit line to the data.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Anhänge

Supplementary Information

Literatur
2.
Zurück zum Zitat Sorrentino V, Menzies KJ, Auwerx J. Repairing mitochondrial dysfunction in disease. Annu Rev Pharmacol Toxicol. 2018;58:353–89.PubMedCrossRef Sorrentino V, Menzies KJ, Auwerx J. Repairing mitochondrial dysfunction in disease. Annu Rev Pharmacol Toxicol. 2018;58:353–89.PubMedCrossRef
3.
Zurück zum Zitat Wu Q, Luo CL, Tao LY. Dynamin-related protein 1 (Drp1) mediating mitophagy contributes to the pathophysiology of nervous system diseases and brain injury. Histol Histopathol. 2017;32(6):551–9.PubMed Wu Q, Luo CL, Tao LY. Dynamin-related protein 1 (Drp1) mediating mitophagy contributes to the pathophysiology of nervous system diseases and brain injury. Histol Histopathol. 2017;32(6):551–9.PubMed
4.
5.
Zurück zum Zitat Yang X, Pan W, Xu G, Chen L. Mitophagy: a crucial modulator in the pathogenesis of chronic diseases. Clin Chim Acta. 2020;502:245–54.PubMedCrossRef Yang X, Pan W, Xu G, Chen L. Mitophagy: a crucial modulator in the pathogenesis of chronic diseases. Clin Chim Acta. 2020;502:245–54.PubMedCrossRef
6.
Zurück zum Zitat Vara-Perez M, Felipe-Abrio B, Agostinis P. Mitophagy in Cancer: A Tale of Adaptation. Cells. 2019;8(5):493–531. Vara-Perez M, Felipe-Abrio B, Agostinis P. Mitophagy in Cancer: A Tale of Adaptation. Cells. 2019;8(5):493–531.
7.
Zurück zum Zitat Guo W, Sun Y, Liu W, Wu X, Guo L, Cai P, Wu X, Wu X, Shen Y, Shu Y, et al. Small molecule-driven mitophagy-mediated NLRP3 inflammasome inhibition is responsible for the prevention of colitis-associated cancer. Autophagy. 2014;10(6):972–85.PubMedPubMedCentralCrossRef Guo W, Sun Y, Liu W, Wu X, Guo L, Cai P, Wu X, Wu X, Shen Y, Shu Y, et al. Small molecule-driven mitophagy-mediated NLRP3 inflammasome inhibition is responsible for the prevention of colitis-associated cancer. Autophagy. 2014;10(6):972–85.PubMedPubMedCentralCrossRef
8.
Zurück zum Zitat Katreddy RR, Bollu LR, Su F, Xian N, Srivastava S, Thomas R, Dai Y, Wu B, Xu Y, Rea MA, et al. Targeted reduction of the EGFR protein, but not inhibition of its kinase activity, induces mitophagy and death of cancer cells through activation of mTORC2 and Akt. Oncogenesis. 2018;7(1):5.PubMedPubMedCentralCrossRef Katreddy RR, Bollu LR, Su F, Xian N, Srivastava S, Thomas R, Dai Y, Wu B, Xu Y, Rea MA, et al. Targeted reduction of the EGFR protein, but not inhibition of its kinase activity, induces mitophagy and death of cancer cells through activation of mTORC2 and Akt. Oncogenesis. 2018;7(1):5.PubMedPubMedCentralCrossRef
10.
Zurück zum Zitat MacKeigan JP, Murphy LO, Blenis J. Sensitized RNAi screen of human kinases and phosphatases identifies new regulators of apoptosis and chemoresistance. Nat Cell Biol. 2005;7(6):591–600.PubMedCrossRef MacKeigan JP, Murphy LO, Blenis J. Sensitized RNAi screen of human kinases and phosphatases identifies new regulators of apoptosis and chemoresistance. Nat Cell Biol. 2005;7(6):591–600.PubMedCrossRef
11.
Zurück zum Zitat Villa E, Proics E, Rubio-Patino C, Obba S, Zunino B, Bossowski JP, Rozier RM, Chiche J, Mondragon L, Riley JS, et al. Parkin-independent Mitophagy controls chemotherapeutic response in Cancer cells. Cell Rep. 2017;20(12):2846–59.PubMedCrossRef Villa E, Proics E, Rubio-Patino C, Obba S, Zunino B, Bossowski JP, Rozier RM, Chiche J, Mondragon L, Riley JS, et al. Parkin-independent Mitophagy controls chemotherapeutic response in Cancer cells. Cell Rep. 2017;20(12):2846–59.PubMedCrossRef
12.
Zurück zum Zitat Abdrakhmanov A, Kulikov AV, Luchkina EA, Zhivotovsky B, Gogvadze V. Involvement of mitophagy in cisplatin-induced cell death regulation. Biol Chem. 2019;400(2):161–70.PubMedCrossRef Abdrakhmanov A, Kulikov AV, Luchkina EA, Zhivotovsky B, Gogvadze V. Involvement of mitophagy in cisplatin-induced cell death regulation. Biol Chem. 2019;400(2):161–70.PubMedCrossRef
13.
Zurück zum Zitat McWilliams TG, Muqit MM. PINK1 and Parkin: emerging themes in mitochondrial homeostasis. Curr Opin Cell Biol. 2017;45:83–91.PubMedCrossRef McWilliams TG, Muqit MM. PINK1 and Parkin: emerging themes in mitochondrial homeostasis. Curr Opin Cell Biol. 2017;45:83–91.PubMedCrossRef
14.
Zurück zum Zitat Wei R, Cao J, Yao S. Matrine promotes liver cancer cell apoptosis by inhibiting mitophagy and PINK1/Parkin pathways. Cell Stress Chaperones. 2018;23(6):1295–309.PubMedPubMedCentralCrossRef Wei R, Cao J, Yao S. Matrine promotes liver cancer cell apoptosis by inhibiting mitophagy and PINK1/Parkin pathways. Cell Stress Chaperones. 2018;23(6):1295–309.PubMedPubMedCentralCrossRef
15.
Zurück zum Zitat Wang J, Gao S, Wang S, Xu Z, Wei L. Zinc oxide nanoparticles induce toxicity in CAL 27 oral cancer cell lines by activating PINK1/Parkin-mediated mitophagy. Int J Nanomedicine. 2018;13:3441–50.PubMedPubMedCentralCrossRef Wang J, Gao S, Wang S, Xu Z, Wei L. Zinc oxide nanoparticles induce toxicity in CAL 27 oral cancer cell lines by activating PINK1/Parkin-mediated mitophagy. Int J Nanomedicine. 2018;13:3441–50.PubMedPubMedCentralCrossRef
16.
Zurück zum Zitat Jin G, Xu C, Zhang X, Long J, Rezaeian AH, Liu C, Furth ME, Kridel S, Pasche B, Bian XW, et al. Atad3a suppresses Pink1-dependent mitophagy to maintain homeostasis of hematopoietic progenitor cells. Nat Immunol. 2018;19(1):29–40.PubMedCrossRef Jin G, Xu C, Zhang X, Long J, Rezaeian AH, Liu C, Furth ME, Kridel S, Pasche B, Bian XW, et al. Atad3a suppresses Pink1-dependent mitophagy to maintain homeostasis of hematopoietic progenitor cells. Nat Immunol. 2018;19(1):29–40.PubMedCrossRef
17.
Zurück zum Zitat Teng Y, Ren X, Li H, Shull A, Kim J, Cowell JK. Mitochondrial ATAD3A combines with GRP78 to regulate the WASF3 metastasis-promoting protein. Oncogene. 2016;35(3):333–43.PubMedCrossRef Teng Y, Ren X, Li H, Shull A, Kim J, Cowell JK. Mitochondrial ATAD3A combines with GRP78 to regulate the WASF3 metastasis-promoting protein. Oncogene. 2016;35(3):333–43.PubMedCrossRef
18.
Zurück zum Zitat Bavelloni A, Ramazzotti G, Poli A, Piazzi M, Focaccia E, Blalock W, Faenza I. MiRNA-210: a current overview. Anticancer Res. 2017;37(12):6511–21.PubMed Bavelloni A, Ramazzotti G, Poli A, Piazzi M, Focaccia E, Blalock W, Faenza I. MiRNA-210: a current overview. Anticancer Res. 2017;37(12):6511–21.PubMed
19.
Zurück zum Zitat Ren Z, Yu J, Wu Z, Si W, Li X, Liu Y, Zhou J, Deng R, Chen D. MicroRNA-210-5p contributes to cognitive impairment in early vascular dementia rat model through targeting Snap25. Front Mol Neurosci. 2018;11:388.PubMedPubMedCentralCrossRef Ren Z, Yu J, Wu Z, Si W, Li X, Liu Y, Zhou J, Deng R, Chen D. MicroRNA-210-5p contributes to cognitive impairment in early vascular dementia rat model through targeting Snap25. Front Mol Neurosci. 2018;11:388.PubMedPubMedCentralCrossRef
20.
Zurück zum Zitat Shao MX, Qu AZ, Wang YQ, Zhong YY. Expression level of miRNA-210-3p in cervical cancer and its prognostic potential. Eur Rev Med Pharmacol Sci. 2020;24(12):6583–8.PubMed Shao MX, Qu AZ, Wang YQ, Zhong YY. Expression level of miRNA-210-3p in cervical cancer and its prognostic potential. Eur Rev Med Pharmacol Sci. 2020;24(12):6583–8.PubMed
21.
Zurück zum Zitat Liu W, Jiang D, Gong F, Huang Y, Luo Y, Rong Y, Wang J, Ge X, Ji C, Fan J, et al. miR-210-5p promotes epithelial-mesenchymal transition by inhibiting PIK3R5 thereby activating oncogenic autophagy in osteosarcoma cells. Cell Death Dis. 2020;11(2):93.PubMedPubMedCentralCrossRef Liu W, Jiang D, Gong F, Huang Y, Luo Y, Rong Y, Wang J, Ge X, Ji C, Fan J, et al. miR-210-5p promotes epithelial-mesenchymal transition by inhibiting PIK3R5 thereby activating oncogenic autophagy in osteosarcoma cells. Cell Death Dis. 2020;11(2):93.PubMedPubMedCentralCrossRef
22.
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(6):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(6):394–424.PubMedCrossRef
23.
24.
Zurück zum Zitat Kudo M, Ueshima K, Yokosuka O, Ogasawara S, Obi S, Izumi N, Aikata H, Nagano H, Hatano E, Sasaki Y, et al. Sorafenib plus low-dose cisplatin and fluorouracil hepatic arterial infusion chemotherapy versus sorafenib alone in patients with advanced hepatocellular carcinoma (SILIUS): a randomised, open label, phase 3 trial. Lancet Gastroenterol Hepatol. 2018;3(6):424–32.PubMedCrossRef Kudo M, Ueshima K, Yokosuka O, Ogasawara S, Obi S, Izumi N, Aikata H, Nagano H, Hatano E, Sasaki Y, et al. Sorafenib plus low-dose cisplatin and fluorouracil hepatic arterial infusion chemotherapy versus sorafenib alone in patients with advanced hepatocellular carcinoma (SILIUS): a randomised, open label, phase 3 trial. Lancet Gastroenterol Hepatol. 2018;3(6):424–32.PubMedCrossRef
25.
Zurück zum Zitat Pollutri D, Patrizi C, Marinelli S, Giovannini C, Trombetta E, Giannone FA, Baldassarre M, Quarta S, Vandewynckel YP, Vandierendonck A, et al. The epigenetically regulated miR-494 associates with stem-cell phenotype and induces sorafenib resistance in hepatocellular carcinoma. Cell Death Dis. 2018;9(1):4.PubMedPubMedCentralCrossRef Pollutri D, Patrizi C, Marinelli S, Giovannini C, Trombetta E, Giannone FA, Baldassarre M, Quarta S, Vandewynckel YP, Vandierendonck A, et al. The epigenetically regulated miR-494 associates with stem-cell phenotype and induces sorafenib resistance in hepatocellular carcinoma. Cell Death Dis. 2018;9(1):4.PubMedPubMedCentralCrossRef
26.
Zurück zum Zitat Ardelt MA, Frohlich T, Martini E, Muller M, Kanitz V, Atzberger C, Cantonati P, Messner M, Posselt L, Lehr T, et al. Inhibition of Cyclin-dependent kinase 5: a strategy to improve Sorafenib response in hepatocellular carcinoma therapy. Hepatology. 2019;69(1):376–93.PubMedCrossRef Ardelt MA, Frohlich T, Martini E, Muller M, Kanitz V, Atzberger C, Cantonati P, Messner M, Posselt L, Lehr T, et al. Inhibition of Cyclin-dependent kinase 5: a strategy to improve Sorafenib response in hepatocellular carcinoma therapy. Hepatology. 2019;69(1):376–93.PubMedCrossRef
27.
Zurück zum Zitat Levy JM, Thompson JC, Griesinger AM, Amani V, Donson AM, Birks DK, Morgan MJ, Mirsky DM, Handler MH, Foreman NK, et al. Autophagy inhibition improves chemosensitivity in BRAF(V600E) brain tumors. Cancer Discov. 2014;4(7):773–80.PubMedPubMedCentralCrossRef Levy JM, Thompson JC, Griesinger AM, Amani V, Donson AM, Birks DK, Morgan MJ, Mirsky DM, Handler MH, Foreman NK, et al. Autophagy inhibition improves chemosensitivity in BRAF(V600E) brain tumors. Cancer Discov. 2014;4(7):773–80.PubMedPubMedCentralCrossRef
28.
Zurück zum Zitat Tong M, Che N, Zhou L, Luk ST, Kau PW, Chai S, Ngan ES, Lo CM, Man K, Ding J, et al. Efficacy of annexin A3 blockade in sensitizing hepatocellular carcinoma to sorafenib and regorafenib. J Hepatol. 2018;69(4):826–39.PubMedCrossRef Tong M, Che N, Zhou L, Luk ST, Kau PW, Chai S, Ngan ES, Lo CM, Man K, Ding J, et al. Efficacy of annexin A3 blockade in sensitizing hepatocellular carcinoma to sorafenib and regorafenib. J Hepatol. 2018;69(4):826–39.PubMedCrossRef
29.
Zurück zum Zitat Braggio D, Koller D, Jin F, Siva N, Zewdu A, Lopez G, Batte K, Casadei L, Welliver M, Strohecker AM, et al. Autophagy inhibition overcomes sorafenib resistance in S45F-mutated desmoid tumors. Cancer. 2019;125(15):2693–703.PubMed Braggio D, Koller D, Jin F, Siva N, Zewdu A, Lopez G, Batte K, Casadei L, Welliver M, Strohecker AM, et al. Autophagy inhibition overcomes sorafenib resistance in S45F-mutated desmoid tumors. Cancer. 2019;125(15):2693–703.PubMed
30.
Zurück zum Zitat Drake LE, Springer MZ, Poole LP, Kim CJ, Macleod KF. Expanding perspectives on the significance of mitophagy in cancer. Semin Cancer Biol. 2017;47:110–24.PubMedPubMedCentralCrossRef Drake LE, Springer MZ, Poole LP, Kim CJ, Macleod KF. Expanding perspectives on the significance of mitophagy in cancer. Semin Cancer Biol. 2017;47:110–24.PubMedPubMedCentralCrossRef
31.
Zurück zum Zitat Wu FQ, Fang T, Yu LX, Lv GS, Lv HW, Liang D, Li T, Wang CZ, Tan YX, Ding J, et al. ADRB2 signaling promotes HCC progression and sorafenib resistance by inhibiting autophagic degradation of HIF1alpha. J Hepatol. 2016;65(2):314–24.PubMedCrossRef Wu FQ, Fang T, Yu LX, Lv GS, Lv HW, Liang D, Li T, Wang CZ, Tan YX, Ding J, et al. ADRB2 signaling promotes HCC progression and sorafenib resistance by inhibiting autophagic degradation of HIF1alpha. J Hepatol. 2016;65(2):314–24.PubMedCrossRef
32.
Zurück zum Zitat Dong XF, Liu TQ, Zhi XT, Zou J, Zhong JT, Li T, Mo XL, Zhou W, Guo WW, Liu X, et al. COX-2/PGE2 Axis regulates HIF2alpha activity to promote hepatocellular carcinoma hypoxic response and reduce the sensitivity of Sorafenib treatment. Clin Cancer Res. 2018;24(13):3204–16.PubMedCrossRef Dong XF, Liu TQ, Zhi XT, Zou J, Zhong JT, Li T, Mo XL, Zhou W, Guo WW, Liu X, et al. COX-2/PGE2 Axis regulates HIF2alpha activity to promote hepatocellular carcinoma hypoxic response and reduce the sensitivity of Sorafenib treatment. Clin Cancer Res. 2018;24(13):3204–16.PubMedCrossRef
33.
Zurück zum Zitat Zhang H, Lu H, Xiang L, Bullen JW, Zhang C, Samanta D, Gilkes DM, He J, Semenza GL. HIF-1 regulates CD47 expression in breast cancer cells to promote evasion of phagocytosis and maintenance of cancer stem cells. Proc Natl Acad Sci U S A. 2015;112(45):E6215–23.PubMedPubMedCentralCrossRef Zhang H, Lu H, Xiang L, Bullen JW, Zhang C, Samanta D, Gilkes DM, He J, Semenza GL. HIF-1 regulates CD47 expression in breast cancer cells to promote evasion of phagocytosis and maintenance of cancer stem cells. Proc Natl Acad Sci U S A. 2015;112(45):E6215–23.PubMedPubMedCentralCrossRef
34.
Zurück zum Zitat Liang Y, Zheng T, Song R, Wang J, Yin D, Wang L, Liu H, Tian L, Fang X, Meng X, et al. Hypoxia-mediated sorafenib resistance can be overcome by EF24 through Von Hippel-Lindau tumor suppressor-dependent HIF-1alpha inhibition in hepatocellular carcinoma. Hepatology. 2013;57(5):1847–57.PubMedCrossRef Liang Y, Zheng T, Song R, Wang J, Yin D, Wang L, Liu H, Tian L, Fang X, Meng X, et al. Hypoxia-mediated sorafenib resistance can be overcome by EF24 through Von Hippel-Lindau tumor suppressor-dependent HIF-1alpha inhibition in hepatocellular carcinoma. Hepatology. 2013;57(5):1847–57.PubMedCrossRef
35.
36.
Zurück zum Zitat Liu LP, Ho RL, Chen GG, Lai PB. Sorafenib inhibits hypoxia-inducible factor-1alpha synthesis: implications for antiangiogenic activity in hepatocellular carcinoma. Clin Cancer Res. 2012;18(20):5662–71.PubMedCrossRef Liu LP, Ho RL, Chen GG, Lai PB. Sorafenib inhibits hypoxia-inducible factor-1alpha synthesis: implications for antiangiogenic activity in hepatocellular carcinoma. Clin Cancer Res. 2012;18(20):5662–71.PubMedCrossRef
38.
Zurück zum Zitat Kang R, Zeng L, Xie Y, Yan Z, Zhou B, Cao L, Klionsky DJ, Tracey KJ, Li J, Wang H, et al. A novel PINK1- and PARK2-dependent protective neuroimmune pathway in lethal sepsis. Autophagy. 2016;12(12):2374–85.PubMedPubMedCentralCrossRef Kang R, Zeng L, Xie Y, Yan Z, Zhou B, Cao L, Klionsky DJ, Tracey KJ, Li J, Wang H, et al. A novel PINK1- and PARK2-dependent protective neuroimmune pathway in lethal sepsis. Autophagy. 2016;12(12):2374–85.PubMedPubMedCentralCrossRef
39.
Zurück zum Zitat Baudier J. ATAD3 proteins: brokers of a mitochondria-endoplasmic reticulum connection in mammalian cells. Biol Rev Camb Philos Soc. 2018;93(2):827–44.PubMedCrossRef Baudier J. ATAD3 proteins: brokers of a mitochondria-endoplasmic reticulum connection in mammalian cells. Biol Rev Camb Philos Soc. 2018;93(2):827–44.PubMedCrossRef
40.
Zurück zum Zitat Desai R, Frazier AE, Durigon R, Patel H, Jones AW, Dalla Rosa I, Lake NJ, Compton AG, Mountford HS, Tucker EJ, et al. ATAD3 gene cluster deletions cause cerebellar dysfunction associated with altered mitochondrial DNA and cholesterol metabolism. Brain. 2017;140(6):1595–610.PubMedPubMedCentralCrossRef Desai R, Frazier AE, Durigon R, Patel H, Jones AW, Dalla Rosa I, Lake NJ, Compton AG, Mountford HS, Tucker EJ, et al. ATAD3 gene cluster deletions cause cerebellar dysfunction associated with altered mitochondrial DNA and cholesterol metabolism. Brain. 2017;140(6):1595–610.PubMedPubMedCentralCrossRef
41.
Zurück zum Zitat Goller T, Seibold UK, Kremmer E, Voos W, Kolanus W. Atad3 function is essential for early post-implantation development in the mouse. PLoS One. 2013;8(1):e54799.PubMedPubMedCentralCrossRef Goller T, Seibold UK, Kremmer E, Voos W, Kolanus W. Atad3 function is essential for early post-implantation development in the mouse. PLoS One. 2013;8(1):e54799.PubMedPubMedCentralCrossRef
42.
Zurück zum Zitat Li S, Lamarche F, Charton R, Delphin C, Gires O, Hubstenberger A, Schlattner U, Rousseau D. Expression analysis of ATAD3 isoforms in rodent and human cell lines and tissues. Gene. 2014;535(1):60–9.PubMedCrossRef Li S, Lamarche F, Charton R, Delphin C, Gires O, Hubstenberger A, Schlattner U, Rousseau D. Expression analysis of ATAD3 isoforms in rodent and human cell lines and tissues. Gene. 2014;535(1):60–9.PubMedCrossRef
43.
Zurück zum Zitat Rizza S, Di Leo L, Mandatori S, De Zio D, Filomeni G. Mitophagy contributes to alpha-tocopheryl succinate toxicity in GSNOR-deficient hepatocellular carcinoma. Biochem Pharmacol. 2020;176:113885.PubMedCrossRef Rizza S, Di Leo L, Mandatori S, De Zio D, Filomeni G. Mitophagy contributes to alpha-tocopheryl succinate toxicity in GSNOR-deficient hepatocellular carcinoma. Biochem Pharmacol. 2020;176:113885.PubMedCrossRef
44.
Zurück zum Zitat Prieto-Domínguez N, Ordóñez R, Fernández A, Méndez-Blanco C, Baulies A, Garcia-Ruiz C, Fernández-Checa JC, Mauriz JL, González-Gallego J. Melatonin-induced increase in sensitivity of human hepatocellular carcinoma cells to sorafenib is associated with reactive oxygen species production and mitophagy. J Pineal Res. 2016;61(3):396–407.PubMedPubMedCentralCrossRef Prieto-Domínguez N, Ordóñez R, Fernández A, Méndez-Blanco C, Baulies A, Garcia-Ruiz C, Fernández-Checa JC, Mauriz JL, González-Gallego J. Melatonin-induced increase in sensitivity of human hepatocellular carcinoma cells to sorafenib is associated with reactive oxygen species production and mitophagy. J Pineal Res. 2016;61(3):396–407.PubMedPubMedCentralCrossRef
45.
Zurück zum Zitat Fu ZJ, Wang ZY, Xu L, Chen XH, Li XX, Liao WT, Ma HK, Jiang MD, Xu TT, Xu J, et al. HIF-1α-BNIP3-mediated mitophagy in tubular cells protects against renal ischemia/reperfusion injury. Redox Biol. 2020;36:101671.PubMedPubMedCentralCrossRef Fu ZJ, Wang ZY, Xu L, Chen XH, Li XX, Liao WT, Ma HK, Jiang MD, Xu TT, Xu J, et al. HIF-1α-BNIP3-mediated mitophagy in tubular cells protects against renal ischemia/reperfusion injury. Redox Biol. 2020;36:101671.PubMedPubMedCentralCrossRef
46.
Zurück zum Zitat Yang H, Shu Z, Jiang Y, Mao W, Pang L, Redwood A, Jeter-Jones SL, Jennings NB, Ornelas A, Zhou J, et al. 6-Phosphofructo-2-kinase/Fructose-2,6-Biphosphatase-2 regulates TP53-dependent paclitaxel sensitivity in ovarian and breast cancers. Clin Cancer Res. 2019;25(18):5702–16.PubMedPubMedCentralCrossRef Yang H, Shu Z, Jiang Y, Mao W, Pang L, Redwood A, Jeter-Jones SL, Jennings NB, Ornelas A, Zhou J, et al. 6-Phosphofructo-2-kinase/Fructose-2,6-Biphosphatase-2 regulates TP53-dependent paclitaxel sensitivity in ovarian and breast cancers. Clin Cancer Res. 2019;25(18):5702–16.PubMedPubMedCentralCrossRef
47.
48.
Zurück zum Zitat Abdul-Aziz A, MacEwan DJ, Bowles KM, Rushworth SA. Oxidative stress responses and NRF2 in human leukaemia. Oxidative Med Cell Longev. 2015;2015:454659.CrossRef Abdul-Aziz A, MacEwan DJ, Bowles KM, Rushworth SA. Oxidative stress responses and NRF2 in human leukaemia. Oxidative Med Cell Longev. 2015;2015:454659.CrossRef
49.
Zurück zum Zitat Huang X, Ding L, Bennewith KL, Tong RT, Welford SM, Ang KK, Story M, Le QT, Giaccia AJ. Hypoxia-inducible mir-210 regulates normoxic gene expression involved in tumor initiation. Mol Cell. 2009;35(6):856–67.PubMedPubMedCentralCrossRef Huang X, Ding L, Bennewith KL, Tong RT, Welford SM, Ang KK, Story M, Le QT, Giaccia AJ. Hypoxia-inducible mir-210 regulates normoxic gene expression involved in tumor initiation. Mol Cell. 2009;35(6):856–67.PubMedPubMedCentralCrossRef
50.
Zurück zum Zitat Kai AK, Chan LK, Lo RC, Lee JM, Wong CC, Wong JC, Ng IO. Down-regulation of TIMP2 by HIF-1alpha/miR-210/HIF-3alpha regulatory feedback circuit enhances cancer metastasis in hepatocellular carcinoma. Hepatology. 2016;64(2):473–87.PubMedCrossRef Kai AK, Chan LK, Lo RC, Lee JM, Wong CC, Wong JC, Ng IO. Down-regulation of TIMP2 by HIF-1alpha/miR-210/HIF-3alpha regulatory feedback circuit enhances cancer metastasis in hepatocellular carcinoma. Hepatology. 2016;64(2):473–87.PubMedCrossRef
51.
Zurück zum Zitat Ren D, Yang Q, Dai Y, Guo W, Du H, Song L, Peng X. Oncogenic miR-210-3p promotes prostate cancer cell EMT and bone metastasis via NF-kappaB signaling pathway. Mol Cancer. 2017;16(1):117.PubMedPubMedCentralCrossRef Ren D, Yang Q, Dai Y, Guo W, Du H, Song L, Peng X. Oncogenic miR-210-3p promotes prostate cancer cell EMT and bone metastasis via NF-kappaB signaling pathway. Mol Cancer. 2017;16(1):117.PubMedPubMedCentralCrossRef
52.
Zurück zum Zitat Tohme S, Yazdani HO, Liu Y, Loughran P, van der Windt DJ, Huang H, Simmons RL, Shiva S, Tai S, Tsung A. Hypoxia mediates mitochondrial biogenesis in hepatocellular carcinoma to promote tumor growth through HMGB1 and TLR9 interaction. Hepatology. 2017;66(1):182–97.PubMedCrossRef Tohme S, Yazdani HO, Liu Y, Loughran P, van der Windt DJ, Huang H, Simmons RL, Shiva S, Tai S, Tsung A. Hypoxia mediates mitochondrial biogenesis in hepatocellular carcinoma to promote tumor growth through HMGB1 and TLR9 interaction. Hepatology. 2017;66(1):182–97.PubMedCrossRef
53.
Zurück zum Zitat Tang Y, Zhou X, Ji J, Chen L, Cao J, Luo J, Zhang S. High expression levels of miR-21 and miR-210 predict unfavorable survival in breast cancer: a systemic review and meta-analysis. Int J Biol Markers. 2015;30(4):e347–58.PubMedCrossRef Tang Y, Zhou X, Ji J, Chen L, Cao J, Luo J, Zhang S. High expression levels of miR-21 and miR-210 predict unfavorable survival in breast cancer: a systemic review and meta-analysis. Int J Biol Markers. 2015;30(4):e347–58.PubMedCrossRef
Metadaten
Titel
Mitophagy promotes sorafenib resistance through hypoxia-inducible ATAD3A dependent Axis
verfasst von
Hong Wu
Tao Wang
Yiqiang Liu
Xin Li
Senlin Xu
Changtao Wu
Hongbo Zou
Mianfu Cao
Guoxiang Jin
Jinyi Lang
Bin Wang
Baohua Liu
Xiaolin Luo
Chuan Xu
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/s13046-020-01768-8

Weitere Artikel der Ausgabe 1/2020

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

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.

Alectinib verbessert krankheitsfreies Überleben bei ALK-positivem NSCLC

25.04.2024 NSCLC Nachrichten

Das Risiko für Rezidiv oder Tod von Patienten und Patientinnen mit reseziertem ALK-positivem NSCLC ist unter einer adjuvanten Therapie mit dem Tyrosinkinase-Inhibitor Alectinib signifikant geringer als unter platinbasierter Chemotherapie.

Update Onkologie

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