Skip to main content
Erschienen in: Journal of Experimental & Clinical Cancer Research 1/2018

Open Access 01.12.2018 | Research

Yes-associated protein (YAP) binds to HIF-1α and sustains HIF-1α protein stability to promote hepatocellular carcinoma cell glycolysis under hypoxic stress

verfasst von: Xiaodong Zhang, Yan Li, Yingbo Ma, Liang Yang, Tao Wang, Xin Meng, Zhihong Zong, Xun Sun, Xiangdong Hua, Hangyu Li

Erschienen in: Journal of Experimental & Clinical Cancer Research | Ausgabe 1/2018

Abstract

Background

Hypoxia-inducible factor 1α (HIF-1α) is essential in hepatocellular carcinoma (HCC) glycolysis and progression. Yes-associated protein (YAP) is a powerful regulator and is overexpressed in many cancers, including HCC. The regulatory mechanism of YAP and HIF-1α in HCC glycolysis is unknown.

Methods

We detected YAP expression in 54 matched HCC tissues and the adjacent noncancerous tissues. The relationship between YAP mRNA expression and that of HIF-1α was analyzed using The Cancer Genome Atlas HCC tissue data. We cultured HepG2 and Huh7 HCC cells under normoxic (20% O2) and hypoxic (1% O2) conditions, and measured the lactate and glucose levels, migration and invasive capability, and the molecular mechanism of HCC cell glycolysis and progression.

Results

In this study, we detected YAP expression in 54 matched HCC tissues and the adjacent noncancerous tissues. We observed that hypoxia-induced YAP activation is crucial for accelerating HCC cell glycolysis. Hypoxia inhibited the Hippo signaling pathway and promoted YAP nuclear localization, and decreased phosphorylated YAP expression in HCC cells. YAP knockdown inhibited HCC cell glycolysis under hypoxic. Mechanistically, hypoxic stress in the HCC cells promoted YAP binding to HIF-1α in the nucleus and sustained HIF-1α protein stability to bind to PKM2 gene and directly activates PKM2 transcription to accelerate glycolysis.

Conclusions

Our findings describe a new regulatory mechanism of hypoxia-mediated HCC metabolism, and YAP might be a promising therapeutic target in HCC.
Hinweise

Electronic supplementary material

The online version of this article (https://​doi.​org/​10.​1186/​s13046-018-0892-2) contains supplementary material, which is available to authorized users.
Abkürzungen
CHX
Cycloheximide
Glut1
Glucose transporter 1
HCC
Hepatocellular carcinoma
HIF-1α
Hypoxia-inducible factor-1α
LATS1
Large tumor suppressor kinase 1
LDHA
Lactate dehydrogenase A
MMP2
Matrix metallopeptidase 2
MMP9
Matrix metallopeptidase 9
PGK1
Phosphoglycerate kinase 1
PKM2
Pyruvate kinase M 2
YAP
Yes-Associated Protein

Background

One of the commonest cancers is hepatocellular carcinoma (HCC); in 2015, it was the primary cause of cancer death in men under 60 years of age [1]. Although surgical resection and other advanced treatment techniques have improved the survival of patients with HCC, their prognosis is still poor [2]. Therefore, identifying novel genes and elucidating the molecular mechanism of progression and glycolysis in HCC is important.
HCC, and other solid tumors, has the common feature of fast growth. However, the growth of HCC cells often exceeds growth of functional blood vessels, and there is frequently insufficient O2 supplement in the regions of HCC [36]. Therefore, tumor cells exist in a hypoxic environment, which is a fundamental solid tumor microenvironment characteristic. Such cells adapt to hypoxic stress by altering their glucose metabolism from oxidation to glycolysis, which provides sufficient energy and materials for cancer cell anabolic growth. In hypoxia, there is an essential role for hypoxia-inducible factor 1α (HIF-1α) in promoting cell glycolysis. HIF-1α directly up-regulates the expression of the glycolytic genes, e.g., PKM2 (pyruvate kinase M2) [7], HK2 (hexokinase 2), and LDHA (lactate dehydrogenase A) [8, 9], to promote glycolysis in tumors. On the other hand, some oncogenes cooperate with HIF-1α to increase HIF-1α stabilization and transcriptional activity, subsequently promoting hypoxic tumor cell glycolysis [10, 11]. Therefore, investigating the regulatory mechanism between HIF-1α and HCC cell glycolysis under hypoxic conditions is worthwhile.
Yes-associated protein (YAP) is a transcriptional activator in the Hippo signaling, while Hippo signaling is a highly conserved tumor suppressor pathway, and it decreases YAP stability and promotes YAP cytoplasmic localization to decrease YAP activity [12]. In many human cancers, YAP is a possible oncogene; it modulates tumor size and tumorigenesis. In HCC, YAP expression is high and it acts as an independent prognostic marker [1315]. The phosphorylation status and localization of YAP determines its activity; YAP activation via nuclear localization is the most important regulatory mechanism. YAP is highly expressed in various cancers: activated YAP promotes cancer cell proliferation, chemoresistance, and migration [1618]. Recent studies have demonstrated a connection between glycolysis and YAP activity [1921]. YAP up-regulates the expression of glucose transporter 3 (GLUT3) at transcriptional level to promote cell glycolysis [19]. Interestingly, YAP also suppresses glyconeogenesis by inhibiting the ability of PGC1α (PPARG coactivator 1α) to activate the transcription of its gluconeogenic targets by binding to their promoters [22]. Therefore, YAP appears to reprogram cellular metabolism. However, the potential regulatory mechanism of YAP and HIF-1α in HCC glycolysis is unknown.
In this study, we observed that hypoxia-induced YAP activation is crucial for accelerating HCC cell glycolysis. Hypoxia inhibited the Hippo signaling pathway and promoted YAP nuclear localization, and decreased phosphorylated YAP expression in HCC cells. YAP knockdown inhibited HCC cell glycolysis under hypoxia. Mechanistically, hypoxic stress in the HCC cells promoted YAP binding to HIF-1α in the nucleus and sustained HIF-1α protein stability to promote glycolysis. Collectively, our findings describe a new regulatory mechanism of hypoxia-mediated HCC metabolism, and YAP might be a promising therapeutic target in HCC.

Methods

Clinical samples and cell culture

Between July 2012 and November 2014, we obtained 54 tumor tissues and the paired adjacent noncancerous tissues from patients who had undergone surgical hepatectomy at the Liaoning Cancer Hospital and Institute (Shenyang, China) and who has been diagnosed with HCC by pathological examination. The patients had not undergone preoperative chemotherapy or radiotherapy. The China Medical University ethics committee approved this study (NO. 2014PS132K). All patients granted informed consent. The RNA sequencing (RNA-Seq) data from TCGA were obtained from Gene Expression Profiling Interactive Analysis (GEPIA) [23], which is based on the University of California Santa Cruz (UCSC) Xena project (http://xena.ucsc.edu).
The HepG2 and Huh7 human HCC cell lines were purchased from Shanghai Institute of Cell Bank (Shanghai, China). HepG2 cells were grown in Eagle’s Minimum Essential Medium and Huh7 cells were grown in Dulbecco’s Modified Eagle Medium. All medium containing 10% fetal bovine serum (FBS), penicillin (100 units/ml), streptomycin (100 μg/ml), 2 mM glutamine, and 10 mM HEPES buffer. Cells were cultured at 37 °C under 20% O2 (normoxia) or 1% O2 (hypoxia), balanced with N2 in a 3-gas incubator.

RNA extraction, real-time PCR and RNA interference

We extracted RNA with TRIzol (Invitrogen, Carlsbad, CA, USA) and performed real-time PCR as has been described previously [24]. The primers used are listed in Additional file 1: Table S1.
All HCC cell transfections were performed in 6-well plates using Invitrogen Lipofectamine 3000 (Thermo Fisher Scientific, Shanghai, China) as per instructions from the manufacturer. We harvested the cells after 48 h to perform real-time PCR or western blotting. Expression plasmid for YAP5SA was obtained from Dr. Dawang Zhou and Dr. Lanfen Zhen [25]. The sense sequences of the YAP small interfering RNAs (siRNAs) (siYAPs, Sigma, Shanghai, China) were as follows:
  • YAP siRNA#1: 5′-GACAUCUUCUGGUCAGAGATT-3′,
  • YAP siRNA#2: 5′-GGUGAUACUAUCAACCAAATT-3′.

Immunohistochemistry and western blotting

We performed immunohistochemical staining and western blotting using methods that have been described previously [24]. Anti-YAP (1:100, immunohistochemistry), Anti–phosphorylated-YAP (p-YAP) (Ser127) (1:1000, western blot), and anti–p-LATS1 (Ser909) (1:1000, western blot) were from Cell Signaling Technology (Shanghai, China). Anti-YAP (1:1000, western blot), anti-LDHA (1:1000, western blot), anti-PKM2 (1:1000, western blot), anti-Glut1 (1:1000, western blotting), anti-PGK1 (1:1000, western blot), anti–large tumor suppressor kinase 1 (LATS1, 1:1000, western blotting), and anti–HIF-1α (1:1000, western blot) rabbit monoclonal antibodies were from Proteintech (Wuhan, China). Anti–matrix metalloproteinase 2 (MMP2, 1:1000, western blot), anti-MMP9 (1:1000, western blot), horseradish peroxidase–linked secondary antibody (1:5000, western blot), and anti–β-actin (1:3000, western blot) were from Abbkine (Wuhan, China). The protein bands were analyzed with Image J software (National Institutes of Health, Bethesda, MD, USA).

Immunofluorescence

The HCC cells were cultured, plated in 6-well plates, washed with phosphate-buffered saline (PBS), and fixed in 4% polyformaldehyde. Primary antibody against YAP (1:100) or HIF-1α (1:100) was added on the plates of cells or frozen sections of HCC tissues, and the plates were incubated at 4 °C overnight. After washing, we added fluorescein isothiocyanate (FITC)-labeled (1:100) secondary antibody (Beyotime, Shanghai, China) and incubated the samples for 2 h. We counterstained the cells using diaminophenylindole (DAPI, Beyotime, Shanghai, China) and visualized them under a confocal microscope.

In vitro migration and invasion

We assessed HCC cell migration and invasive capability using 24-well Transwell plates with or without Matrigel (BD), respectively. Then cells (1 × 105) in serum-free culture medium were plated in the top chamber. High-glucose Dulbecco’s modified Eagle’s medium (DMEM) with 10% FBS was added to the bottom chamber. After 24-h incubation, the membrane was washed with PBS, fixed using 4% paraformaldehyde, and stained using 0.1% crystal violet solution. The cells were counted in five random fields under microscopy. We performed the experiment in triplicate and repeated it three times.

Extracellular acidification rate (ECAR)

The Seahorse XFe 96 Extracellular Flux Analyzer (Seahorse Bioscience) was used to determine the extracellular acidification rate (ECAR). ECAR was examined with a Seahorse XF glycolysis stress test kit according to the manufacturer’s protocols. In brief, cells (1 × 104 cells / well) were seeded into a Seahorse XF 96 cell culture plate. After baseline measurements, glucose, oligomycin, and 2-DG were sequentially injected into each well at the time points specified. ECAR data were assessed by Seahorse XF-96 Wave software and shown in mpH/ minute.

Lactate and glucose level measurement

Lactate Assay Kit II (Sigma, Shanghai, China) and High Sensitivity Glucose Assay Kit (Sigma, Shanghai, China) were used according to the manufacturer’s instructions to detect HCC cell lactate and glucose levels, respectively.

Co-immunoprecipitation

After 24-h culture under hypoxia (1% O2), the HCC cells were collected and incubated with 300 μl lysis buffer with protease inhibitors for 40 min on ice. Then, the supernatant was collected, and 2 μg HIF-1α, YAP, or immunoglobulin G (IgG) (Proteintech) antibody was added and incubated at 4 °C overnight. Next, 20 μl protein A/G-agarose beads (Santa Cruz Biotechnology, Shanghai, China) was added and rocked for 3 h at 4 °C. The pelleted cells were collected and washed three times with lysis buffer. Finally, the precipitate was boiled with 40 μl loading buffer for 5 min and analyzed by western blotting.

Subcellular fractionation

HCC tissues or HCC cells cytoplasmic and nuclear extracts were separated using a Nuclear and Cytoplasmic Protein Extraction Kit (Beyotime, Shanghai, China) according to the manufacturer instructions. Then, YAP expression was analyzed by western blotting.

Cycloheximide (CHX) chase assay

CHX chase assay was used to determine the half-life of HIF-1α. YAP knockdown or control HCC cells were individually seeded in 60-mm dishes for 24 h. Then, the cells were cultured under hypoxia (1% O2) for 24 h and treated with CHX (100 μg/ml) for 0 h, 2 h, and 4 h. The cells were collected at the indicated time points, and HIF-1α expression was analyzed by western blotting.

Chromatin immunoprecipitation (ChIP) assay

Cells or cells transfected with YAP siRNA were cultured under hypoxic condition (1%O2) for 24 h. Then ChIP assay was performed using the EZ-ChIP kit (17–371, Millipore, Billerica, MA, USA) according to the manufacturer’s instructions. The immunoprecipitated DNA was purified and analyzed by real-time PCR. Primer information used in ChIP assays was listed as follows:
  • PKM2: Forward: 5’-TTCCTGCCTCTTGGTATGAC-3′,
  • Reverse: 5’-CGGCTTGTTCCCTCCTAC-3’

Luciferase reporter assay

The PKM2 promoter reporter plasmids were amplified from a human genomic DNA template and inserted into pGL3-Promoter Vector (Promega, Madison, WI). Cell were seeded in a 96-well plate and co-transfected with PKM2 promoter reporter plasmids (wt) or mutation of PKM2 promoter reporter plasmids and the 8xGTIIC-luciferase plasmid using Invitrogen Lipofectamine 3000 (Thermo Fisher Scientific, Shanghai, China) as per instructions from the manufacturer. Then cells were cultured under hypoxic condition (1%O2) or nomoxic condition (20%O2) for 24 h. Luciferase activities were analyzed using the Dual-luciferase reporter assay (Promega, Madison, WI, USA) according to the manufacturer’s instructions.

Gene set enrichment analysis (GSEA)

The HCC tissues data from TCGA databases were grouped into two groups based on the expression of HIF-1α: HIF-1α high expression and HIF-1α low expression. The gene expression values of the two groups samples were then put through GSEA v3.0 to analyze Hippo signaling genes signatures. Hippo signaling gene sets were obtained from the MSigDB database v6.1 [26].

Statistical analysis

We analyzed the data with SPSS 17.0 (SPSS Inc., Chicago, IL, USA) and GraphPad Prism 6 (GraphPad Software, San Diego, California, USA). The data are reported as the mean ± SD; Student’s t-test or analysis of variance was used for the statistical analyses. The correlation between YAP mRNA and HIF-1α mRNA was analyzed by Pearson’s correlation coefficient using GEPIA. p-values < 0.05 indicates statistical significance. We repeated the experiments three times at minimum.

Results

YAP expression was high in HCC tissues

We firstly investigated YAP expression in 54 cases of HCC tissues and their paired adjacent nontumor tissues by real-time PCR and immunohistochemistry. The results obtained from real-time PCR showed significantly increased YAP mRMA levels in the tumor tissues as compared with the adjacent noncancerous tissues (p = 0.006, Fig. 1a); Immunohistochemical staining revealed 53.7% (29/54) HCC tissues were positive for YAP expression, whereas only 18.5% (10/54) adjacent normal tissues were positive for YAP expression (Fig. 1b). Then immunofluorescence staining and cell fractionation assays were used to investigate YAP localization, among the 29 cases of HCC tissues overexpression YAP, 68.9% (20/29) HCC tissues showed stronger nuclear YAP staining as opposed to cytoplasmic staining (Fig. 1c and d). In addition, we also analyzed the mRNA levels of two canonical YAP transcription target genes (CTGF, CYR61) in the same HCCs displaying higher YAP mRNA levels. The results revealed that expression of CTGF mRNA and CYR61 mRNA were all increased in the HCC, along with the up-regulation of YAP mRNA (Additional file 2: Figure S1A). These results suggest that, in HCC tissues, YAP expression is high and that YAP is localized to the nucleus.

YAP correlated strongly with HIF-1α

As a solid tumor, the abnormal new vasculature of the tumor and the increased consumption of oxygen in the cell proliferation in HCC are imbalanced, so the inner of HCC tissues is always hypoxic. Hypoxia promotes HCC invasion and migration, and hypoxia inducible factor-1α (HIF-1α) is also up-regulated in HCC. So in order to evaluate whether HIF-1α correlated with YAP, we firstly performed gene set enrichment analysis and found enriched expression of Hippo signaling genes in HCC tissues from The Cancer Genome Atlas (TCGA) database (Fig. 2a). Since YAP is a transcriptional activator in the Hippo signaling, and can also be inhibited by Hippo signaling [8], we then retrieved data on 369 HCC cases from The Cancer Genome Atlas (TCGA) to analyze the relationship between YAP mRNA and HIF-1α mRNA expression, and found a positive correlation between the two (R = 0.5, P = 1.7E-24) (Fig. 2b). Furthermore, we analyzed the relationship between the expression of YAP mRNA and HIF-1α mRNA in 54 cases of HCC tissues. It showed that YAP mRNA expression correlated positively with that of HIF-1α mRNA (R = 0.64, p < 0.01) (Fig. 2c). And western blotting yielded similar results both in the HCC tissues (Fig. 2d) and in the nuclear fraction of HCC (Additional file 2: Figure S1B). Our results suggest that YAP overexpression correlated strongly with HIF-1α in HCC tissues under hypoxia.

Hypoxia activated YAP and induced YAP nuclear translocation in HCC cells

To determine whether hypoxia activates YAP, we incubated HCC cells under hypoxic and normoxic conditions for 24 h. The expression of p-YAP (Ser127) was significantly decreased under hypoxic conditions, while total YAP was not increased, and the protein levels of LATS1 and p-LATS1 (Ser909), the upstream regulators of YAP in the Hippo pathway, were decreased (Fig. 3a and b). It implied that Hippo signaling might be inhibited by hypoxia. As YAP nuclear localization is the key regulatory mechanism for activating YAP, we performed immunofluorescence staining to examine the cellular localization of YAP. Hypoxia triggered significant YAP nuclear translocation in the HCC cells (Fig. 3c and d). Consistent with that, cell fractionation showed more YAP protein accumulation in the nuclear fraction and less YAP protein in the cytoplasmic fraction of hypoxic cells (Fig. 3e and f). In addition, hypoxia also increased the mRNA expression of four canonical YAP transcription target genes (CTGF, CYR61, AREG and EDN1) (Fig. 3g and h). The results suggest that hypoxia activates YAP and induces YAP translocation to the nucleus by inhibiting the Hippo pathway in HCC cells.

Hypoxia promoted HCC cell glycolysis in vitro

Recent studies have reported that hypoxia promotes cancer cell glycolysis [8, 9]. To confirm this, the glucose and lactate assays were performed and the results showed that hypoxia significantly increased HCC cell glucose uptake and lactate production rates, respectively, compared with normoxia (Fig. 4a and b). Hypoxia also showed increased extracellular acidification rate (ECAR), which reflects overall glycolytic flux (Fig. 4c and d). Usually, hypoxia promotes cancer cell glycolysis by up-regulating the expression of key glycolysis enzymes [27, 28]. So we then performed real-time PCR to detect the expression of key glycolysis enzymes in HepG2 and Huh7 cells under 24 h normoxia or hypoxia. LDHA mRNA, GLUT1 mRNA, PGK1 mRNA, and PKM2 mRNA expression levels were up-regulated in the hypoxic cells than in the normoxic cells, and PKM2 mRNA had a significantly increased among the key glycolysis enzymes (Fig. 4e), western blotting yielded similar results (Fig. 4f). Besides that, as a potent micro-environmental factor, hypoxia also promotes tumor migration via up-regulation the expression of matrix metallo proteinases 2 and 9 (MMP2 and MMP9) [2932]. We then examined HCC cell invasion and migration under normoxia and hypoxia using Transwell assays. Hypoxia significantly promoted HCC cell migration and invasion when compared with normoxia (Fig. 4g and h). Moreover, the hypoxic cells had higher MMP2 and MMP9 expression than the normoxic cells (Fig. 4i and j). These results suggest that hypoxia contributes to HCC cell glycolysis, invasion, and migration.

Silencing YAP inhibited HCC cell glycolysis under hypoxia

To investigate the role of YAP in hypoxic HCC cells, we knocked down YAP expression in HCC cells using YAP siRNA. The knockdown efficiency was examined using real-time PCR and western blotting (Fig. 5a and b). Silencing YAP under hypoxia significantly decreased the HCC cell glucose uptake and lactate production rates compared with control cells (Fig. 5c and d). In addition, silencing YAP also decreased extracellular acidification rate (ECAR) (Fig. 5e and f) in HCC cells under hypoxic condition. These results indicated that YAP knockdown might inhibit HCC cell glycolysis under hypoxia. To further confirm the hypothesis, we examined the expression of PKM2 and activated YAP in YAP knockdown cells under hypoxia. It showed that YAP knockdown also decreased the expression of PKM2 and HIF-1α protein (Fig. 5g and h). However, it did not significant change the level of HIF-1α mRNA, although it decreased the PKM2 mRNA level (Additional file 3: Figure S2A and B). Moreover, activating YAP restored the expression of HIF-1α protein and PKM2 mRNA (Additional file 3: Figure S2C and E) and HCC cell glycolysis under hypoxia (Additional file 3: Figure S2F and G). Furthermore, silencing YAP significantly inhibited the invasion and migration of hypoxic HCC cells (Fig. 5i and j) and decreased MMP2 and MMP9 protein expression in hypoxic cells (Fig. 5k and l). Our findings indicate that YAP can mediate hypoxia-induced glycolysis in HCC cells.

YAP bound and sustained HIF-1α stability to promote glycolysis in HCC cells under hypoxia

To investigate the molecular mechanism of YAP in contributing to hypoxia-induced glycolysis in HCC cells, the cellular localization of YAP and HIF-1α was examined using immunofluorescence staining. Hypoxia greatly promoted YAP and HIF-1α accumulation in the nucleus (Fig. 6a and b). As hypoxia triggered both YAP and HIF-1α nuclear translocation, we believe that YAP may complex with HIF-1α in the nucleus under hypoxia. Co-immunoprecipitation confirmed that YAP could bind HIF-1α under hypoxic conditions (Fig. 6c). However, silencing YAP significantly decreased HIF-1α protein expression under hypoxia and did not change the level of HIF-1α mRNA, so we wondered whether YAP could sustain HIF-1α stabilization. We used CHX chase assays to explore the effect of YAP on HIF-1α stability, and found that YAP knockdown cells had significantly shortened HIF-1α half-life compared with the control cells (Fig. 6d). The results indicated that YAP bound HIF-1α to form a complex and sustained HIF-1α stability. As shown in Figs. 4 and 5, we found the expression of PKM2 was significant changed, so we further wondered whether YAP/HIF-1a complexes could bind target PKM2 gene to promote HCC cells glycolysis in hypoxia. Since YAP has no DNA-binding domains, and binds to its target genes promoters by interacting with DNA-binding transcription factors [33, 34], so we focused on HIF-1a and analyzed HIF-1a and PKM2 gene sequence. Finally, we found a candidate HRE (HIF-1a binding site 5’-ACGTG-3′) in PKM2 promoter. To determine whether YAP/HIF-1a complexes bind at this site, chromatin immunoprecipitation (ChIP) assay was performed in HCC cells under hypoxia. The results showed that ChIP with HIF-1a or YAP antibody enriched the putative HRE sequence > 5 fold compared to IP with IgG (Fig. 6e). In contrast, after silencing YAP, the enrichment of PKM2 with HIF-1a or YAP antibody was significantly decreased compared with IgG (Fig. 6f). These data indicated that YAP/HIF-1a complexes colud bind to the promoter of PKM2 gene. In addition, we also used luciferase reporter assay to test whether this putative HRE in the PKM2 gene is functional. As shown in Fig. 6g, this putative HRE in the PKM2 gene significantly increased luciferase activity in hypoxic HCC cells, however, mutation HRE significantly decreased hypoxia-induced luciferase activity. Taken together, these data indicated that YAP/HIF-1a complexes bound to PKM2 gene promoter and directly activated its transcription to accelerate glycolysis in hypoxia.

Discussion

YAP is a powerful regulator and is overexpressed in many cancers, including HCC [35, 36]. It plays important roles in cell proliferation, glycolysis, and survival [37, 38]. In our study, we found that YAP was up-regulated in HCC tissues compared to the adjacent normal tissues, and high YAP expression always accumulated in HCC cell nuclei. Analysis of TCGA public data sets using GSEA and GEPIA revealed that YAP correlated positively with HIF-1α levels in HCC tissues, and the results was further confirmed in our 54 cases of HCC tissues. In addition, YAP knockdown significantly decreased PKM2 expression and inhibited glucose uptake and lactate production rates in hypoxic HCC cells. As hypoxia regulates cell glycolysis via HIF-1α [3941], we speculated that YAP might be involved in promoting HCC cell glycolysis via hypoxia.
YAP has been identified as a downstream effector of multiple inputs, such as the Hippo [42], WNT/β-catenin [43], and Janus kinase (JNK) [44] pathways. Recently, novel mechanisms between hypoxia and YAP have garnered increasing attention. Ma et al. reported that SIAH2 (Siah 2E3 ubiquitin ligase 2), an essential hypoxia pathway component, inhibits the Hippo pathway and activates YAP by destabilizing LATS2 under hypoxia [45]. Zhou et al. found that hypoxia promoted cell survival and inhibited apoptosis by inducing YAP nuclear translocation, and YAP activation led to Sorafenib resistance in HCC cells [46]. In the present study, hypoxia promoted HCC cell glycolysis by decreasing p-YAP expression and triggering YAP nuclear translocation. The decreased p-YAP expression under hypoxia is consistent with the results of Yan et al. [47]. Furthermore, YAP knockdown significantly decreased PKM2 expression and inhibited glucose uptake and lactate production rates in hypoxic HCC cells. These results indicate that YAP is essential for hypoxia-triggered glycolysis in HCC cells. Subsequently, we studied the potential molecular mechanism involved.
Ma et al. showed that, under hypoxia, HIF-2α up-regulation of YAP occurred via neither kinase signaling nor protein–protein interaction [48]. Dai et al. also showed that HIF-1α did not mediate hypoxia-triggered YAP nuclear translocation [17]. However, in the present study, LATS1 and p-LATS1 (Ser909) expression levels were decreased under hypoxia. LATS1 is the core protein in the Hippo pathway and is also the upstream regulator of YAP. Upon Hippo pathway activation, p-LATS1 phosphorylates YAP, which is subsequently confined to the cytoplasm or is degraded [49]. Our data show that hypoxia decreased LATS1 and p-LATS1 (Ser909) expression to inhibit the Hippo pathway and activate YAP. Moreover, hypoxia triggered both YAP and HIF-1α nuclear translocation; co-immunoprecipitation confirmed that hypoxia enhanced YAP and HIF-1α interaction. As silencing YAP decreased HIF-1α protein expression, it is possible that YAP is important for HIF-1α stability under hypoxia. To demonstrate this, the CHX chase assays showed significantly shortened half-life of HIF-1α in the YAP knockdown cells compared with the control cells. YAP is a transcriptional co-activator and has no DNA-binding domains [50]. So YAP does not directly bind to the promoter of downstream target genes to activate them, such as PKM2. YAP is mainly dependent on multiple domains to interact with transcription factors and form a complex to promote the expression and activation of downstream target genes in nucleus [51]. Therefore, we revealed that YAP/HIF-1a complexes binds at 5’-ACGTG-3′ in PKM2 gene promoter and directly activates its transcription. Taken together, our findings indicate that YAP forms a complex with HIF-1α in the nucleus and sustains HIF-1α stability to bind to PKM2 gene and directly activates its transcription to accelerate glycolysis in HCC cells under hypoxia.

Conclusions

In summary, our study demonstrated that YAP expression was high in HCC tissues and correlated positively with the expression of HIF-1α and PKM2. Hypoxia promotes HCC cell glycolysis via YAP activation by decreasing p-YAP expression and triggering YAP nuclear translocation. YAP activation interacts directly with HIF-1α in the nucleus and sustains HIF-1α stability to activate PKM2 transcription. The HIF-1α–YAP regulatory axis may provide a better understanding of the molecular mechanism of HCC glycolysis and progression, and YAP may be a potential therapeutic target in HCC.

Acknowledgements

We are particularly grateful to Dr. Dawang Zhou and Dr. Lanfen Zheng for providing the plasmid of YAP5SA.

Funding

This work was supported by the National Natural Science Foundation of China [grant number 81472302 to Hangyu Li].

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.
The research protocol was approved by the China Medical University ethics committee (NO. 2014PS132K).
Not applicable.

Competing interests

The authors declare that they have no competing interests.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Literatur
27.
Metadaten
Titel
Yes-associated protein (YAP) binds to HIF-1α and sustains HIF-1α protein stability to promote hepatocellular carcinoma cell glycolysis under hypoxic stress
verfasst von
Xiaodong Zhang
Yan Li
Yingbo Ma
Liang Yang
Tao Wang
Xin Meng
Zhihong Zong
Xun Sun
Xiangdong Hua
Hangyu Li
Publikationsdatum
01.12.2018
Verlag
BioMed Central
Erschienen in
Journal of Experimental & Clinical Cancer Research / Ausgabe 1/2018
Elektronische ISSN: 1756-9966
DOI
https://doi.org/10.1186/s13046-018-0892-2

Weitere Artikel der Ausgabe 1/2018

Journal of Experimental & Clinical Cancer Research 1/2018 Zur Ausgabe

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.

Bei Senioren mit Prostatakarzinom auf Anämie achten!

24.04.2024 DGIM 2024 Nachrichten

Patienten, die zur Behandlung ihres Prostatakarzinoms eine Androgendeprivationstherapie erhalten, entwickeln nicht selten eine Anämie. Wer ältere Patienten internistisch mitbetreut, sollte auf diese Nebenwirkung achten.

ICI-Therapie in der Schwangerschaft wird gut toleriert

Müssen sich Schwangere einer Krebstherapie unterziehen, rufen Immuncheckpointinhibitoren offenbar nicht mehr unerwünschte Wirkungen hervor als andere Mittel gegen Krebs.

Update Onkologie

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