Expression of hypoxia‑inducible factor 3α in hepatocellular carcinoma and its association with other hypoxia‑inducible factors

  • Authors:
    • Ping Liu
    • Xiefan Fang
    • Yang Song
    • Jian‑Xin Jiang
    • Qian‑Jin He
    • Xiang‑Jie Liu
  • View Affiliations

  • Published online on: March 24, 2016     https://doi.org/10.3892/etm.2016.3193
  • Pages: 2470-2476
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The functional role of hypoxia-inducible factor (HIF)-3α in the development of hepatocellular carcinoma (HCC) is not yet fully understood. The aim of the present study was to elucidate the association between HIF‑3α expression and the clinicopathological features as well as prognosis of HCC patients. In addition, we investigated the association between HIF‑3α expression and the expression of HIF‑1α and HIF‑2α in tumor tissues. The protein levels of HIF‑3α were determined using immunohistochemical analysis of paraffin sections of 126 paired HCC and peritumoral tissues. PLC/PRF/5 cells, a human HCC cell line, were transfected with HIF‑1α and HIF‑2α vectors and HIF‑3α mRNA and protein expression was detected using quantitative polymerase chain reaction and western blot analysis, respectively. The expression of HIF‑3α was upregulated in 46.0% (58/126) and downregulated in 42.9% (54/126) of tumor tissues, respectively, when compared to peritumoral tissues. HIF‑3α protein expression was not associated with peripheral blood vessel invasion, overall survival, or disease‑free survival in HCC patients (P>0.05). In HCC tissues, the levels of HIF‑3α protein were positively correlated with HIF‑2α, but not with HIF‑1α expression in HCC tissues. HIF‑3α was upregulated in PLC/PRF/5 and Hep3B cells overexpressed with HIF‑1α or HIF‑2α. The hypoxic microenvironment of liver cancer did not lead to elevated HIF‑3α protein expression, indicating that HIF‑3α is regulated differently from HIF‑1α in vivo. The correlation between HIF‑3α and HIF‑2α expression at the cellular and tissue levels indicated that HIF‑3α may be a target gene of HIF‑2α. The hypoxic microenvironment did not lead to elevation of HIF‑3α protein expression in liver cancer; thus, HIF‑3α may be a target gene of HIF‑2α.

Introduction

Tumor hypoxia was first described in the 1950s, and there is currently increasing evidence to indicate that it is a common feature in numerous types of cancer, including hepatocellular carcinoma (HCC) (1,2). Although HCC is among the most hypervascular tumor types, it contains hypoxic regions due to rapid cell proliferation and aberrant formation of blood vessels (3). The effects of hypoxia on cells are predominantly mediated by hypoxia-inducible factors (HIF), which consist of an oxygen-regulated subunit (HIF-1α, HIF-2α or HIF-3α) and a constitutively expressed HIF-1β subunit (4).

Under normal oxygen pressure and in the presence of Fe2+ and acetone dicarboxylic acid, prolyl hydroxylase domain (PHD) catalyzes the hydroxylation of key amino acid residues in the HIF-α oxygen-dependent degradation domain (5). Hydroxylated HIF-α binds to the Von Hippel-Lindau tumor suppressor and is rapidly degraded via a ubiquitin-proteasome pathway (6). Under hypoxic conditions, HIFs are not modified by PHDs, but dimerize with the aryl-hydrocarbon receptor nuclear translator (ARNT)/HIF-1β via interactions with helix-loop-helix and Per/Arnt/Sim domains (5). The HIF heterodimers are translocated to the nucleus, and co-activators such as CBP/p300 are recruited (5). The HIF heterodimers recognize and bind hypoxia response elements (HREs) that contain a consensus sequence (G/A) CGTG within the promoter regions of target genes to drive adaptive gene transcription (7,8). It has been reported that HIF-1 and HIF-2 regulate the expression of hundreds of genes that are involved in numerous processes associated with cancer biology, including cell survival, tumor angiogenesis, metastasis and resistance to radiation and chemotherapy (8,9).

HIF-3α is a member of the HIF family and was initially discovered by Gu et al (10) in 1998. HIF-3α has relatively low sequence identities with HIF-1α and HIF-2α (10). HIF-1α and HIF-2α have two transactivation domains (TADs) (11), while HIF-3a has only one TAD (12). HIF-3α has a unique leucine zipper domain and an LXXLL (L is Leucine and X is any amino acid) motif (10). These unique structural features are evolutionarily conserved. Compared with HIF-1α and HIF-2α, which have been studied extensively (5), little is known about the regulation and function of HIF-3α. Recent studies have indicated that hypoxia induces HIF-3α expression, and that HIF-3α may be a target gene of HIF-1 and HIF-2 (1315). HIF-3 may suppress the expression of genes that are typically inducible by HIF-1α and HIF-2α in tumor cells, and therefore, it may be a negative regulator of gene expression in response to hypoxia (12,14).

The expression pattern of HIF-3α in HCC tissues is currently unknown, and only a few studies have investigated the association between HIF-3α expression and the expression of HIF-1α and HIF-2α (1315); however, the results are inconsistent or even conflicting. To determine the role of HIF-3α in hepatocarcinogenesis, immunostaining was used herein to compare HIF-3α expression in HCC and paired peritumoral tissues obtained from 126 clinical samples. Furthermore, the association between the expression of HIF-3α and HIF-1α/HIF-2α was assessed in HCC clinical tissues and the human cell lines PLC/PRF/5 and Hep3B.

Materials and methods

Patients and specimens

Tissue samples from a total of 126 patients with HCC that underwent a surgical liver resection were obtained between October 2005 and June 2009. Tissue samples for 76 patients were obtained from the Department of Hepatobiliary Surgery of the Affiliated Hospital of Guiyang Medical College (Guiyang, China) and the remaining samples were sourced from 50 patients at the Department of General Surgery of Center Hospital of Huanggang (Huanggang, China). Informed consent was obtained from all patients. The research protocol was approved by the Human Ethics Committees of the Guiyang Medical College and the Center Hospital of Huanggang. All tissue specimens were obtained from the patients prior to any medical treatments. Peritumoral tissues were obtained from at least 2 cm away from the primary tumor site. All patients tested positively for the hepatitis B antigen HBsAg and negatively for hepatitis C virus and human immunodeficiency virus. The cohort had 110 males and 16 females, with an average age of 48.8 years and an age range of 19–66 years. The maximum diameter of HCC tissue was <5.0 cm in 60 patients and was ≥5.0 cm in 66 patients. Data from follow-up examinations following liver resection were collected for all patients. The clinical pathological features of the 126 HCC patients are listed in Table I.

Table I.

Correlations between HIF-3α protein expression in surgical specimens of HCC and clinicopathological characteristics.

Table I.

Correlations between HIF-3α protein expression in surgical specimens of HCC and clinicopathological characteristics.

HIF-3α

ParameterLowHighP-value
Age (years) 0.200
  <501321
  ≥504745
Gender 0.202
  Male5060
  Female106
Cirrhosis 0.595
  Absent4041
  Present2025
Tumor size (cm) 0.114
  <53327
  ≥52739
AFP (µg/l) 0.377
  <4003230
  ≥4002836
Histological grade 0.676
  Well98
  Moderate4346
  Poor812
Capsular infiltration 0.526
  Absent4445
  Present1621
Vascular invasion 0.098
  Absent4259
  Present1510

[i] Data were analyzed using the two-tailed Mann-Whitney U-test. HIF-3α, hypoxia-inducible factor 3α; HCC, hepatocellular carcinoma; AFP, α-fetoprotein.

Cell culture and transfection

The human HCC cell lines PLC/PRF/5 and Hep3B were purchased from the Institute of Biochemistry & Cell Biology of the Shanghai Institutes for Biological Sciences (Chinese Academy of Sciences, Shanghai, China). HIF-1α and HIF-2α expression plasmids and the control plasmid pcDNA3.1 were purchased from Shanghai Gene Chem Co., Ltd. (Shanghai, China). Cells were grown in six-well plates with Dulbecco's modified Eagle's medium (DMEM; GE Healthcare Life Sciences, Logan, UT, USA) containing 10% fetal bovine serum (FBS), 2 mmol/l L-glutamine, 50 units/ml penicillin and 50 g/ml streptomycin (all GE Healthcare Life Sciences) at 37°C and in 5% CO2. PLC/PRF/5 and Hep3B cells at 70–80% confluence were transfected with 1.2 µg plasmids using Lipofectamine® 2000 (Thermo Fisher Scientific, Inc., Waltham, MA, USA), following the manufacturer's protocol. Transfected cells were incubated at 37°C for 6 h, and were then cultured for a further 16 h with fresh DMEM medium containing 10% FBS. The mRNA and protein expression levels of the target genes in the transfected cells were analyzed using quantitative reverse transcription polymerase chain reaction (RT-qPCR) and western blot analysis, respectively.

RT-qPCR

Total RNA was isolated from the PLC/PRF/5 and Hep3B cells using TRIzol reagent (Thermo Fisher Scientific, Inc.), as previously described (16,17). To avoid genomic DNA contamination, RNA samples were treated with RNase-free DNase I ((Takara Bio, Inc., Shiga, Japan) for 20 min at 37°C. The quantity and quality of total RNA were determined using a NanoDrop 2000 spectrophotometer (Thermo Fisher Scientific, Inc.). cDNA synthesis was performed at 42°C for 60 min in a total volume of 25 µl containing 2 µg RNA, 1.6 µM oligo(dT)18, 0.6 µM dNTPs, 200 U M-MLV reverse transcriptase, and 10X Reaction buffer (all Promega Corporation, Madison, WI, USA). All researchers received biosafety training and were screened and vaccinated against hepatitis B virus. qPCR analysis was performed as described previously (16,18), using a reaction mixture consisting of 10 µl 2X SYBR Green mix (Invitrogen; Thermo Fisher Scientific, Inc.), 2 µl cDNA template, 0.6 µl each of forward and reverse primers (10 µM) and double distilled H2O, to a final volume of 20 µl. qPCR was performed on a ABI 7500 Real-Time PCR system (Applied Biosystems; Thermo Fisher Scientific, Inc.) with the following cycling program: Denaturation at 94°C for 1 min, followed by 40 cycles at 94°C for 1 min, 55°C for 1 min and 72°C for 1 min. The primer sequences were as follows: HIF-1α forward, 5′-ACTTCTGGATGCTGGTGATTTG-3′ and reverse, 5′-GCTTCGCTGTGTGTTTTGTTCT-3′; HIF-2α forward, 5′-TCATGCGACTGGCAATCAGC-3′ and reverse, 5′-GTCACCACGGCAATGAAACC-3′; HIF-3α forward, 5′-CCTGGACATGAAGTTCACCTACTG-3′ and reverse, 5′-GGAAGCGATACTGCCCTGTTA-3′; and β-actin forward, 5′-AGTTGCGTTACACCCTTTCTTGAC-3′ and reverse, 5′-GCTCGCTCCAACCGACTGC-3′. The number of replications was three for each sample. Reactions without template cDNA were used as negative control. The cycle quantification (Cq) values were determined and the data were analyzed using the 2−ΔΔCq method (19), following normalization to of β-actin.

Immunohistochemical (IHC) analysis and scoring of protein expression

IHC analysis of the tissue samples was performed as described previously (18). Briefly, the tissue samples were fixed using 10% formaldehyde, embedded in paraffin (both Boster Biological Technology, Ltd., Wuhan, China) and rehydrated using ethanol, after which endogenous peroxidase activity was blocked using 3% H2O2 in methanol solution (Boster Biological Technology, Ltd.). Subsequently, the tissue samples were cut into 5 µm sections using a microtome (Leica RM2155; Leica Microsystems GmbH, Wetzlar, Germany). The sections were then immersed in citrate buffer (pH 6.0; Boster Biological Technology, Ltd.) and heated in a microwave oven for 15 min in order to unmask the antigens, after which the sections were incubated with mouse anti-HIF-3α (1:250; cat. no. NBP2-45735; Novus Biologicals LLC, Littleton, CO, USA) at 37°C for 1 h and then overnight at 48°C. After washing three times with phosphate-buffered saline, the sections were incubated with biotin-conjugated goat anti-mouse immunoglobulin G (IgG) secondary antibody (1:200; cat. no. sc-2075; Santa Cruz Biotechnology, Inc., Dallas, Texas, USA), followed by incubation with horseradish peroxidase (HRP)-conjugated streptavidin (Boster Biological Technology, Ltd.) for 20 min. Detection of immunoreactivity was performed using 3,3′-diaminobenzidine (Boster Biological Technology, Ltd.) under a FV300 fluorescent microscope (Olympus Corporation, Tokyo, Japan).

The protein expression level of HIF-3α was assessed using the following classification system based on the number of cells with cytoplasmic and nuclear staining: I) No staining; II) nuclear staining in <10% of cells and/or with weak cytoplasmic staining; III) nuclear staining in 10–50% of cells and/or with distinct cytoplasmic staining; and IV) nuclear staining in >50% of cells and/or with strong cytoplasmic staining (2023). The staining scores I and II were considered to be low expression, while the scores III and IV were considered as high expression (2023). The protein level of HIF-1α and HIF-2α in HCC and paired peritumoral tissues has been described in our previous study (18).

Protein preparation and western blot analysis

Cells were lysed in lysis buffer (50 mM Tris, pH 7.2, 1% Triton X-100, 0.5% sodium deoxycholate, 0.1% SDS, 500 mM NaCl, 10 mM MgCl2, 10 µg/ml leupeptin, 10 µg/ml aprotinin, 10 ml/l NP-40, 0.2 g/l NaN3 and 1 mM PMSF; Bio-Rad Laboratories, Inc., Hercules, CA, USA) and the protein concentration was quantified using a Bradford Protein Assay (Bio-Rad Laboratories, Inc.). A total of 30–50 µg protein from each sample was separated using 10% SDS-PAGE, transferred to polyvinylidene fluoride membranes, and blocked with 5% nonfat milk in Tris-buffered saline and Tween 20 (Boster Biological Technology, Ltd.) for 2 h (24). The membranes were then incubated with mouse anti-HIF-3α (1:250; cat. no. NBP2-45735; Novus Biologicals LLC), HIF-1α (1:1,000; cat. no. sc-53546; Santa Cruz Biotechnology, Inc.), HIF-2α (1:250; cat. no. sc-13596; Santa Cruz Biotechnology, Inc.) and β-actin (1:3,000; cat. no. sc-47778; Santa Cruz Biotechnology, Inc.) monoclonal antibodies for 1 h at 37°C and at 4°C overnight. The membranes were washed and then incubated with HRP-conjugated goat anti-mouse IgG monoclonal antibody (1:2,000, cat. no. sc-2005; Santa Cruz Biotechnology, Inc.) for 1 h at room temperature. The bands were visualized using an enhanced chemiluminescence detection system (SuperSignal West Pico Chemiluminescent Substrate; cat. no. 34080; Thermo Fisher Scientific, Inc.) and band intensities were calculated using ImageJ software, version 1.41 (https://imagej.nih.gov/ij/).

Statistical analysis

Statistical analysis was performed using SPSS software, version 16.0 (SPSS, Inc., Chicago, IL, USA). Continuous variables were expressed as the mean ± standard deviation and analyzed using a two-tailed Student's t-test. The correlation analyses between HCC clinicopathological parameters and HIF-3α expression were conducted using a two-tailed Mann-Whitney U-test. Survival curves were computed by Kaplan-Meier analysis. Linear associations were evaluated using Spearman's Rank or Pearson's correlation coefficients. Prognostic significance was analyzed using a log-rank test. P<0.05 was considered to indicate a statistically significant difference.

Results

Expression of HIF-3α in HCC tissues

The protein level of HIF-3α was measured using IHC analysis on paraffin-embedded sections of 126 human HCC samples and paired peritumor tissues. Positive staining of HIF-3α was located in cytoplasm and/or nuclei of HCC tissues, with representative staining shown in Fig. 1A. High expression of HIF-3α was found in 66/126 tumor tissues (52.3%) and 63/126 peritumoral tissues (50.0%). The expression of HIF-3α was upregulated in 46.0% (58/126) and downregulated in 42.9% (54/126) of tumor tissues, when compared with the peritumoral tissues. No obvious difference in HIF-3α expression was identified between the remaining 11.1% (14/126) of tumor and peritumoral tissues. Our previous study showed that HIF-1α was higher in HCC tissues compared with peritumoral tissues (18). Therefore, as opposed to HIF-1α, the hypoxic microenvironment in liver cancer did not increase HIF-3α protein expression.

Next, we analyzed the association between HIF-3α expression and pathological features of HCC. As shown in Table I, no significant correlation was found between HIF-3α expression and the clinicopathological features of HCC, including age, gender, presence of liver cirrhosis, tumor size, serum α-fetoprotein level, tumor differentiation grade, capsular infiltration and portal vein invasion.

Disease-free survival (DFS) and overall survival (OS) of HCC patients

The correlation between HIF-3α expression and long-term patient survival following hepatectomy was analyzed. No significant correlation was found between HIF-3α expression levels in HCC tissues and the OS or DFS times of HCC patients. The mean OS periods for patients with high and low HIF-3α expression levels in their tumor tissues were 36.5±2.7 and 39.0±3.3 months (P=0.457), respectively. The mean DFS period of the patients with high and low HIF-3α expression levels in their tumor tissues were 25.9±2.6 and 29.9±3.3 months (P=0.344, Fig. 2), respectively.

Association between HIF-3α expression and the expression of HIF-1α and HIF-2α

A few studies have reported an association between HIF-3α expression and the expression of HIF-1α and HIF-2α (1315); however, the results are inconsistent or even conflicting. Therefore, we investigated their relationship both in vivo and in vitro. In HCC tissues, Spearman correlation analysis revealed that the expression of HIF-3α was significantly correlated with the expression of HIF-2α (rs=0.198, P=0.030), but not with HIF-1α expression (rs=0.045, P=0.855) (data not shown). In addition, PLC/PRF/5 and Hep3B cells transfected with either HIF-1α or HIF-2α plasmids expressed a higher level of HIF-3α compared with the pcDNA2.1 control cells. The elevation of HIF-3α was higher in the HIF-2α overexpressing cells compared with the HIF-1α overexpressing cells, indicating that HIF-3α may be a target gene of HIF-1α and HIF-2α in PLC/PRF/5 and Hep3B cells, but HIF-3α is regulated more effectively by HIF-2α (Fig. 3).

Discussion

In general, hypoxia is the most important factor involved in the regulation of the expression of HIF-α members. It has been reported that the mRNA and protein levels of HIF-α members are increased in hypoxic environments, and are often overexpressed in hypoxic solid tumors (25). Unlike the observation of consistent increase of HIF-2α in the majority of tumors, our previous data have shown that the expression patterns of HIF-1α and HIF-2α are opposite in HCC and paired peritumoral tissues (18). The level of HIF-1α in HCC tissues is significantly higher compared with that in peritumoral tissues, whereas the level of HIF-2α is markedly lower in tumor tissues compared with peritumoral tissues (18). Notably, we did not identify any obvious differences in HIF-3α expression between HCC and peritumoral tissues in this study. The expression of HIF-3α protein was increased in ~50% of the HCC specimens compared with peritumoral tissues, but was decreased or unaltered in the other ~50%. This discrepancy may be attributed to different sensitivities of HIF-1α, HIF-2α and HIF-3α in response to hypoxia. Furthermore, although all HIF-α members are predominantly regulated by oxygen pressure, they are additionally regulated by other factors in the tumor microenvironment. These factors include glucose metabolism and mutations in proto-oncogenes and tumor suppressor genes (2629). These factors are likely to exert different effects on the expression of HIF-α factors, leading to the upregulation of HIF-1α, the downregulation of HIF-2α and the inconsistent expression of HIF-3α in HCC tissues. Therefore, the regulation of HIF-α expression is complicated, and further investigations are required to determine the underling mechanisms that control HIF-3a expression in HCC tissues.

The varying expression patterns of HIF-α factors in HCC tissues indicate that they may serve different functions in response to hypoxia. The transactivation activity of HIF-3α is different from those of HIF-1α and HIF-2α (30). Previous studies have suggested that HIF-3α may suppress HIF-1α and HIF-2α mediated gene expression when the expression of ARNT is limited (13,31,32). Hara et al (12) transfected expression vectors containing HIF-1α, HIF-2α or HIF-3α genes into COS-7 cells and found that HIF-1α and HIF-2α upregulated the transcription of HRE-driven genes, whereas HIF-3α inhibited their expression. Recent research has indicated that HIF-3α is an oxygen-dependent transcription activator, and serves a crucial function in the transcriptional response to hypoxia by binding to target gene promoters, including LC3C, REDD1 and SQRDL, thus stimulating their expression (33). Therefore, further studies are required to investigate the role of HIF-3α in response to hypoxia.

To reveal the role of HIF-3α in the development of HCC, we divided the human samples into two groups based on the score of HIF-3α expression. However, no significant correlation was identified between HIF-3α expression and the clinicopathological characteristics of HCC samples. Furthermore, no significant correlation was detected between the expression of HIF-3α in HCC tissues and the OS or DFS of HCC patients. No statistically significant correlation was detected between HIF-3α expression and the prognosis of HCC patients. However, larger population-based studies are required to confirm the inconsistent expression patterns of HIF-3α in HCC and to identify the underlying causes.

Previous studies have reported associations among the expression levels of HIF-1α, HIF-2α, and HIF-3α; however, their results are inconsistent or conflicting (1315,34). Tanaka et al (13) have found that the siRNA-mediated knockdown of HIF-1α in human renal cell carcinoma notably reduced the 2,2-dipyridyl-induced expression of HIF-3α protein. In addition, an IHC study revealed an overlapping region with positive HIF-1α and HIF-3α expression within the cells (13). These results indicate that HIF-3α is a target gene of HIF-1α. However, the expression of a stabilized form of HIF-1α did not alter HIF-3α mRNA levels in either zebrafish embryos (34) or 3T3-L1 cells (15). Hatanaka et al (15) have observed that HIF-2α specifically binds to the sequence between −251 and −228 bp upstream of the transcription start site of mouse HIF-3α, which is essential in the response to HIF-2α stimulation. In human umbilical venous endothelial cells, HIF-3α expression is promoted by HIF-1 and HIF-2 (14). All these inconsistent results regarding the HIF-1α- and HIF-2α-mediated regulation of HIF-3α induction may be due to the different cell types used in the experiments. In HCC cell lines, HIF-1α and HIF-2α increased the expression of HIF-3α in PLC/PRF/5 and Hep3B cells. The correlation between HIF-2α and HIF-3α expression is more marked, indicating that HIF-3α may be a target gene of HIF-2α in both PLC/PRF/5 and Hep3B cells.

In conclusion, despite the structural similarities between HIF-1α, HIF-2α and HIF-3α, their expression patterns notably differ, indicating that they may play different roles in the development of HCC. The expression of HIF-3α protein was not associated with histopathological features, OS or DFS in HCC patients. However, HIF-3α is a potential target gene of HIF-2α in PLC/PRF/5 cells. Further studies are required to confirm the direct regulation of HIF-3α by HIF-2α.

Acknowledgements

This study was supported by the National Natural Science Foundation of China (grant no. 81160311).

References

1 

Mucaj V, Shay JE and Simon MC: Effects of hypoxia and HIFs on cancer metabolism. Int J Hematol. 95:464–470. 2012. View Article : Google Scholar : PubMed/NCBI

2 

Cao S, Yang S, Wu C, Wang Y, Jiang J and Lu Z: Protein expression of hypoxia-inducible factor-1 alpha and hepatocellular carcinoma: A systematic review with meta-analysis. Clin Res Hepatol Gastroenterol. 38:598–603. 2014. View Article : Google Scholar : PubMed/NCBI

3 

Aravalli RN, Cressman EN and Steer CJ: Cellular and molecular mechanisms of hepatocellular carcinoma: An update. Arch Toxicol. 87:227–247. 2013. View Article : Google Scholar : PubMed/NCBI

4 

Semenza GL: Oxygen sensing, hypoxia-inducible factors and disease pathophysiology. Ann Rev Pathol. 9:47–71. 2014. View Article : Google Scholar

5 

Yang SL, Wu C, Xiong ZF and Fang X: Progress on hypoxia-inducible factor-3: Its structure, gene regulation and biological function (Review). Mol Med Rep. 12:2411–2416. 2015.PubMed/NCBI

6 

Haase VH: The VHL tumor suppressor: Master regulator of HIF. Curr Pharm Des. 15:3895–3903. 2009. View Article : Google Scholar : PubMed/NCBI

7 

Majmundar AJ, Wong WJ and Simon MC: Hypoxia-inducible factors and the response to hypoxic stress. Mol Cell. 40:294–309. 2010. View Article : Google Scholar : PubMed/NCBI

8 

Semenza GL: Hypoxia-inducible factors: Mediators of cancer progression and targets for cancer therapy. Trends in pharmacological sciences. 33:207–214. 2012. View Article : Google Scholar : PubMed/NCBI

9 

Yang Y, Sun M, Wang L and Jiao B: HIFs, angiogenesis and cancer. J Cell Biochem. 114:967–974. 2013. View Article : Google Scholar : PubMed/NCBI

10 

Gu YZ, Moran SM, Hogenesch JB, Wartman L and Bradfield CA: Molecular characterization and chromosomal localization of a third alpha-class hypoxia inducible factor subunit, HIF3alpha. Gene Expr. 7:205–213. 1998.PubMed/NCBI

11 

Tian H, McKnight SL and Russell DW: Endothelial PAS domain protein 1 (EPAS1), a transcription factor selectively expressed in endothelial cells. Genes Dev. 11:72–82. 1997. View Article : Google Scholar : PubMed/NCBI

12 

Hara S, Hamada J, Kobayashi C, Kondo Y and Imura N: Expression and characterization of hypoxia-inducible factor (HIF)-3alpha in human kidney: Suppression of HIF-mediated gene expression by HIF-3alpha. Biochem Biophys Res Commun. 287:808–813. 2001. View Article : Google Scholar : PubMed/NCBI

13 

Tanaka T, Wiesener M, Bernhardt W, Eckardt KU and Warnecke C: The human HIF (hypoxia-inducible factor)-3alpha gene is a HIF-1 target gene and may modulate hypoxic gene induction. Biochem J. 424:143–151. 2009. View Article : Google Scholar : PubMed/NCBI

14 

Augstein A, Poitz DM, Braun-Dullaeus RC, Strasser RH and Schmeisser A: Cell-specific and hypoxia-dependent regulation of human HIF-3α: Inhibition of the expression of HIF target genes in vascular cells. Cell Mol Life Sci. 68:2627–2642. 2011. View Article : Google Scholar : PubMed/NCBI

15 

Hatanaka M, Shimba S, Sakaue M, Kondo Y, Kagechika H, Kokame K, Miyata T and Hara S: Hypoxia-inducible factor-3alpha functions as an accelerator of 3T3-L1 adipose differentiation. Biol Pharm Bull. 32:1166–1172. 2009. View Article : Google Scholar : PubMed/NCBI

16 

Fang X, Dong W, Thornton C, Scheffler B and Willett KL: Benzo(a)pyrene induced glycine N-methyltransferase messenger RNA expression in fundulus heteroclitus embryos. Mar Environ Res. 69(Suppl): S74–S76. 2010. View Article : Google Scholar : PubMed/NCBI

17 

Fang X, Thornton C, Scheffler BE and Willett KL: Benzo[a]pyrene decreases global and gene specific DNA methylation during zebrafish development. Environ Toxicol Pharmacol. 36:40–50. 2013. View Article : Google Scholar : PubMed/NCBI

18 

Yang SL, Liu LP, Jiang JX, Xiong ZF, He QJ and Wu C: The correlation of expression levels of HIF-1α and HIF-2α in hepatocellular carcinoma with capsular invasion, portal vein tumor thrombi and patients' clinical outcome. Jpn J Clin Oncol. 44:159–167. 2014. View Article : Google Scholar : PubMed/NCBI

19 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(−Delta Delta C(T)) Method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

20 

Dai CX, Gao Q, Qiu SJ, Ju MJ, Cai MY, Xu YF, Zhou J, Zhang BH and Fan J: Hypoxia-inducible factor-1 alpha, in association with inflammation, angiogenesis and MYC, is a critical prognostic factor in patients with HCC after surgery. BMC Cancer. 9:4182009. View Article : Google Scholar : PubMed/NCBI

21 

Xiang ZL, Zeng ZC, Fan J, Tang ZY, He J, Zeng HY and Chang JY: The expression of HIF-1α in primary hepatocellular carcinoma and its correlation with radiotherapy response and clinical outcome. Mol Biol Rep. 39:2021–2029. 2012. View Article : Google Scholar : PubMed/NCBI

22 

Xiang ZL, Zeng ZC, Fan J, Tang ZY, Zeng HY and Gao DM: Gene expression profiling of fixed tissues identified hypoxia-inducible factor-1α, VEGF and matrix metalloproteinase-2 as biomarkers of lymph node metastasis in hepatocellular carcinoma. Clin Cancer Res. 17:5463–5472. 2011. View Article : Google Scholar : PubMed/NCBI

23 

Xie H, Song J, Liu K, Ji H, Shen H, Hu S, Yang G, Du Y, Zou X, Jin H, et al: The expression of hypoxia-inducible factor-1alpha in hepatitis B virus-related hepatocellular carcinoma: Correlation with patients' prognosis and hepatitis B virus X protein. Dig Dis Sci. 53:3225–3233. 2008. View Article : Google Scholar : PubMed/NCBI

24 

Yang SL, Yu C, Jiang JX, Liu LP, Fang X and Wu C: Hepatitis B virus X protein disrupts the balance of the expression of circadian rhythm genes in hepatocellular carcinoma. Oncol Lett. 8:2715–2720. 2014.PubMed/NCBI

25 

Semenza GL: HIF-1 mediates metabolic responses to intratumoral hypoxia and oncogenic mutations. J Clin Invest. 123:3664–3671. 2013. View Article : Google Scholar : PubMed/NCBI

26 

Tsapournioti S, Mylonis I, Hatziefthimiou A, Ioannou MG, Stamatiou R, Koukoulis GK, Simos G, Molyvdas PA and Paraskeva E: TNFα induces expression of HIF-1α mRNA and protein but inhibits hypoxic stimulation of HIF-1 transcriptional activity in airway smooth muscle cells. J Cell Physiol. 228:1745–1753. 2013. View Article : Google Scholar : PubMed/NCBI

27 

Kilic-Eren M, Boylu T and Tabor V: Targeting PI3K/Akt represses hypoxia inducible factor-1α activation and sensitizes rhabdomyosarcoma and Ewing's sarcoma cells for apoptosis. Cancer Cell Int. 13:362013. View Article : Google Scholar : PubMed/NCBI

28 

Chen C, Cai S, Wang G, Cao X, Yang X, Luo X, Feng Y and Hu J: c-Myc enhances colon cancer cell-mediated angiogenesis through the regulation of HIF-1α. Biochem Biophys Res Commun. 430:505–511. 2013. View Article : Google Scholar : PubMed/NCBI

29 

Agani F and Jiang BH: Oxygen-independent Regulation of HIF-1: Novel Involvement of PI3K/AKT/mTOR pathway in cancer. Curr Cancer Drug Targets. 13:245–251. 2013. View Article : Google Scholar : PubMed/NCBI

30 

Li QF, Wang XR, Yang YW and Lin H: Hypoxia upregulates hypoxia inducible factor (HIF)-3alpha expression in lung epithelial cells: Characterization and comparison with HIF-1alpha. Cell Res. 16:548–558. 2006. View Article : Google Scholar : PubMed/NCBI

31 

Maynard MA, Evans AJ, Hosomi T, Hara S, Jewett MA and Ohh M: Human HIF-3alpha4 is a dominant-negative regulator of HIF-1 and is down-regulated in renal cell carcinoma. FASEB J. 19:1396–1406. 2005. View Article : Google Scholar : PubMed/NCBI

32 

Maynard MA, Evans AJ, Shi W, Kim WY, Liu FF and Ohh M: Dominant-negative HIF-3 alpha 4 suppresses VHL-null renal cell carcinoma progression. Cell Cycle. 6:2810–2816. 2007. View Article : Google Scholar : PubMed/NCBI

33 

Zhang P, Yao Q, Lu L, Li Y, Chen PJ and Duan C: Hypoxia-inducible factor 3 is an oxygen-dependent transcription activator and regulates a distinct transcriptional response to hypoxia. Cell Rep. 6:1110–1121. 2014. View Article : Google Scholar : PubMed/NCBI

34 

Zhang P, Lu L, Yao Q, Li Y, Zhou J, Liu Y and Duan C: Molecular, functional and gene expression analysis of zebrafish hypoxia-inducible factor-3α. Am J Physiol Regul, Integr Comp Physiol. 303:R1165–R1174. 2012. View Article : Google Scholar

Related Articles

Journal Cover

June-2016
Volume 11 Issue 6

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Liu P, Fang X, Song Y, Jiang JX, He QJ and Liu XJ: Expression of hypoxia‑inducible factor 3α in hepatocellular carcinoma and its association with other hypoxia‑inducible factors. Exp Ther Med 11: 2470-2476, 2016
APA
Liu, P., Fang, X., Song, Y., Jiang, J., He, Q., & Liu, X. (2016). Expression of hypoxia‑inducible factor 3α in hepatocellular carcinoma and its association with other hypoxia‑inducible factors. Experimental and Therapeutic Medicine, 11, 2470-2476. https://doi.org/10.3892/etm.2016.3193
MLA
Liu, P., Fang, X., Song, Y., Jiang, J., He, Q., Liu, X."Expression of hypoxia‑inducible factor 3α in hepatocellular carcinoma and its association with other hypoxia‑inducible factors". Experimental and Therapeutic Medicine 11.6 (2016): 2470-2476.
Chicago
Liu, P., Fang, X., Song, Y., Jiang, J., He, Q., Liu, X."Expression of hypoxia‑inducible factor 3α in hepatocellular carcinoma and its association with other hypoxia‑inducible factors". Experimental and Therapeutic Medicine 11, no. 6 (2016): 2470-2476. https://doi.org/10.3892/etm.2016.3193