Open Access

TNF‑α‑mediated epithelial‑to‑mesenchymal transition regulates expression of immune checkpoint molecules in hepatocellular carcinoma

  • Authors:
    • Ritu Shrestha
    • Kim R. Bridle
    • Darrell H.G. Crawford
    • Aparna Jayachandran
  • View Affiliations

  • Published online on: February 19, 2020     https://doi.org/10.3892/mmr.2020.10991
  • Pages: 1849-1860
  • Copyright: © Shrestha et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Hepatocellular carcinoma (HCC) is the fastest growing cause of cancer‑related deaths globally. Epithelial‑to‑mesenchymal transition (EMT) is a cellular process that confers HCC tumor cells with the ability to evade the immune system. Immune escape in most tumors, including HCC, is controlled by immune checkpoint molecules. The aim of the present study was to investigate the association between EMT and immune checkpoint in HCC, and identify novel therapeutic targets for HCC. An in vitro model of reversible EMT was utilized based on cytokine tumor necrosis factor (TNF)‑α treatment of HCC cell lines Hep3B and PLC/PRF/5. Hep3B and PLC/PRF/5 cells were treated with TNF‑α, and the EMT status and the expression of immune checkpoint molecules was assessed by reverse transcription‑quantitative PCR, western blotting and immunofluorescence. To confirm an association between EMT and immune modulators, cells were exposed to culture medium with TNF‑α for 3 days to induce EMT, following which a reversal assay was performed. The expression of immune modulators and mesenchymal‑to‑epithelial transition (MET) status was investigated upon reversal of EMT. Furthermore, SurvExpress, a web‑based platform was utilized to analyze survival and recurrence in a dataset of patients with HCC. TNF‑α treatment for 3 days induced EMT in Hep3B and PLC/PRF/5 cells, as demonstrated by the downregulation of epithelial markers along with upregulation in mesenchymal markers. An EMT reversal assay was able to induce MET by increasing epithelial markers and decreasing mesenchymal markers. TNF‑α‑induced EMT led to the upregulation of immune modulators, including programmed death receptor ligand (PD‑L)1, PD‑L2, CD73 and B7‑H3. In contrast, reversal of EMT suppressed the expression of PD‑L1, PD‑L2, CD73 and B7‑H3. In addition, high expression of TNF‑α and PD‑L1 in 422 patients with HCC was associated with poor overall survival. The coordinate expression of TNF‑α with PD‑L2 in this patient cohort was associated with increased HCC recurrence. In conclusion, the present study demonstrated a close association between immune modulator expression and EMT induction/reversal driven by TNF‑α.

Introduction

Hepatocellular carcinoma (HCC) is a primary liver malignancy accounting for 80–90% of all liver cancer (1). According to the latest global cancer data, the incidence of HCC has rapidly risen with ~782,000 deaths in 2018 (2). The majority of HCC is resistant to treatment as it is often diagnosed at advanced stages of disease, so the overall median survival is <1 year (3). To the best of the authors' knowledge, sorafenib and levatinib are the only first-line systemic therapeutic agents available for the treatment of advanced HCC. Several second-line drugs, including regorafenib, ramucirumab and cabozantinib, have been approved for patients with HCC who have previously been treated with sorafenib, but the overall survival (OS) rate remains poor (4,5).

Immune checkpoint blockade therapies have emerged as potential treatments in cancer immunotherapy (6). The cytotoxic T-cell mediated immune response is accompanied by co-stimulatory signals and co-inhibitory signals or immune checkpoints (7,8). The immune checkpoint molecules are often overexpressed or activated in the tumor microenvironment, thus resulting in immune evasion by tumor cells (79). Immune checkpoint inhibitors (ICIs) that block programmed cell death protein-1 (PD-1) or its ligand, programmed death receptor ligand (PD-L)1, have shown therapeutic potential in several cancer types, including HCC (10,11). Anti-PD-1 ICIs, nivolumab and pembrolizumab, have been approved by The Food and Drug Administration for the treatment of HCC in patients where treatment using sorafenib has failed (12). There are a number of ongoing clinical trials on ICIs targeting PD-1 (nivolumab and pembrolizumab) and PD-L1 (atezolizumab) in HCC (7,13). Due to the success of anti-PD-1/PD-L1 immunotherapy clinical studies, understanding the underlying mechanisms regulating PD-L1 expression has attracted increasing attention. Previous research by the authors and other researchers demonstrated an increased expression of other immune checkpoint molecules in HCC (8,14). However, the mechanisms regulating the expression of these and other immune checkpoint molecules in HCC remains unclear (8).

A key process in HCC progression is epithelial-to-mesenchymal transition (EMT). EMT is a complex molecular and cellular process that allows epithelial cells to gain mesenchymal features, including migration, invasiveness and increased resistance to immune evasion (15,16). Mesenchymal-to-epithelial transition (MET) is the reverse process of EMT that offers phenotypic plasticity for conversion of mesenchymal cells to epithelial derivatives (15). EMT status is closely associated with cancer metastasis, stemness, immune escape and drug resistance in HCC (17,18). It was previously identified that EMT is involved in the regulation of PD-L1 in several cancer types, including breast cancer (19,20), lung cancer (17,21), pancreatic cancer (22), esophageal cancer (23) and salivary adenoid cystic carcinoma (24). The authors previously demonstrated that EMT is associated with PD-L1 expression in patients with HCC (8).

In the present study, it was hypothesized that EMT is associated with the ability of HCC tumor cells to escape detection and destruction by the immune response, by regulating the expression of immune checkpoint molecules, such as PD-L1. In the present study, the changes in immune checkpoint expression during the EMT process were examined by incorporating a reversible model of EMT based on tumor necrosis factor (TNF)-α treatment of the HCC cell lines Hep3B and PLC/PRF/5. TNF-α was selected as an inducer of EMT, as it has been previously demonstrated to play an important role in promoting EMT in HCC (25). This in vitro system may provide an improved understanding of the modulation of immune checkpoints during both EMT and MET.

Materials and methods

Cell culture and reagents

The human HCC cell line Hep3B was provided by Professor V. Nathan Subramaniam, The Queensland University of Technology. The human HCC cell line PLC/PRF/5 was purchased from CellBank Australia (cat. no. 85061113). Both cell lines were mycoplasma-tested using the MycoAlert™ mycoplasma detection kit (Lonza Group, Ltd.) and cultured in DMEM (Thermo Fisher Scientific, Inc.) with 10% FBS (Gibco; Thermo Fisher Scientific, Inc.) and 1% penicillin/streptomycin (Thermo Fisher Scientific, Inc.), and incubated at 37°C under a humidified atmosphere with 5% CO2 in air (26). The cytokine TNF-α was purchased from PeproTech, Inc.

EMT reversal assay

An EMT reversal assay was performed to determine the association between EMT and immune checkpoint expression. Firstly, EMT was induced by using culture medium with 20 ng/ml TNF-α for 3 days at 37°C and then reversal of EMT was induced by changing the culture medium without TNF-α for the next 3 days.

RNA extraction and cDNA synthesis

RNA was isolated from Hep3B and PLC/PRF/5 cells, as previously described, using Isolate II Bioline RNA synthesis kit (Bioline), according to the manufacturer's protocol (26). The quantity and purity of RNA was confirmed using the NanoDrop™ 2000c spectrophotometer (Thermo Fisher Scientific, Inc.). cDNA was synthesized by reverse transcribing 1 µg RNA into cDNA using the Bioline SensiFAST cDNA synthesis kit (Bioline) with the following thermocycling conditions: Primer annealing at 25°C for 10 min, reverse transcription at 42°C for 15 min, inactivation at 85°C for 5 min and a final extension 4°C for 30 min.

Reverse transcription-quantitative (RT-q)PCR

RT-qPCR was performed using SensiFast™ SYBR® Lo-ROX kit (Bioline) on a ViiA7 Real-Time PCR system (Applied Biosystems; Thermo Fisher Scientific, Inc.), as previously described (26). A three-step cycle procedure was applied with 40 cycles of the following conditions: 95°C for 5 sec, 63°C for 20 sec and 75°C for 20 sec. β-actin was used as an internal control. The primer sequences used are listed in Table I. Data were analyzed using the 2−ΔΔCq method where β-actin was assigned as the housekeeping gene. The results are expressed as relative mRNA expression to the control (27).

Table I.

List of primers for reverse transcription-quantitative PCR.

Table I.

List of primers for reverse transcription-quantitative PCR.

GenePrimer sequence, 5′-3′
β-actinF: CCAACCGCGAGAAGATGA
R: CCAGAGGCGTACAGGGATAG
E-cadherinF: AGGCCAAGCAGCAGTACATT
R: ATTCACATCCAGCACATCCA
N-cadherinF: TCCTTGCTTCTGACAATGGA
R: TTCGCAAGTCTCTGCCTCTT
OccludinF: TAGTCAGATGGGGGTGAAGG
R: CATTTATGATGAGCAGCCCC
VimentinF: CTTCAGAGAGAGGAAGCCGA
R: ATTCCACTTTGCGTTCAAGG
Snai2F: TGGTTGCTTCAAGGACACAT
R: GTTGCAGTGAGGGCAAGAA
FibronectinF: CAGTGGGAGACCTCGAGAAG
R: TCCCTCGGAACATCAGAAAC
PD-L1F: TGCCGACTACAAGCGAATTACTG
R: CTGCTTGTCCAGATGACTTCGG
PD-L2F: ACCGTGAAAGAGCCACTTTG
R: GCGACCCCATAGATGATTATGC
CD73F: TTGGAAATTTGGCCTCTTTG
R: ACTTCATGAACGCCCTGC
B7-H3F: CTGGCTTTCGTGTGCTGGAGAA
R: GCTGTCAGAGTGTTTCAGAGGC
VISTAF: AGATGCACCATCCAACTGTGTGG
R: AGGCAGAGGATTCCTACGATGC
VTCN1F: CTCACAGATGCTGGCACCTACA
R: GCAAGGTCTCTGAGCTGGCATT

[i] PD-L, programmed death receptor ligand; Snai2, snail family transcriptional repressor 2; F, forward; R, reverse; VTCN1, V-set domain-containing T-cell activation inhibitor 1; VISTA, V-domain immunoglobulin suppressor of T-cell activation.

Western blot analysis

Cells were seeded (2×105 cells/well) in 6-well plates and subsequently lysed using RIPA buffer (Thermo Fisher Scientific, Inc.) with complete protease inhibitors and PhosSTOP™ phosphatase inhibitors (both purchased from Roche Diagnostics) on ice to extract the total protein. The total protein was measured with a Pierce™ BCA protein assay kit (Thermo Fisher Scientific, Inc.). A total of 10 µg protein was used for 4% SDS-PAGE, following which proteins were transferred to a PVDF membrane. After blocking with 5% skimmed milk in Tris-buffered saline containing 0.1% Tween-20 at room temperature for 1 h, the membranes were incubated with primary antibodies at 4°C overnight. An enhanced chemiluminescence reagent, SuperSignal™ West Femto Maximum Sensitivity Substrate (Thermo Fisher Scientific, Inc.), was used to detect the protein adhering to the membranes following incubation with horseradish peroxidase-conjugated secondary antibodies at room temperature for 1 h. All antibodies used are listed in Table II, including the dilutions, cat. nos. and suppliers. GAPDH was selected as the housekeeping control in each group. The images were captured with ImageQuant™ LAS 500 and quantified with Image Studio™ Lite v5.2 software (LI-COR Biosciences).

Table II.

List of antibodies.

Table II.

List of antibodies.

A, Primary antibodies

Dilution

NameCat. no.ManufacturerWestern blotting Immuno-fluorescence
E-cadherinab76055Abcam1:1,0001:100
N-cadherinSC-53488Santa Cruz Biotechnology, Inc.1:200
OccludinSC-133255Santa Cruz Biotechnology, Inc.1:2001:50
Vimentinab92547Abcam1:1,0001:600
CD73ab133582Abcam1:250
CD73ab175396Abcam1:100
PD-L1ab238697Abcam2 µg/ml2 µg/ml
B7-H314058SCell Signaling Technology, Inc.1:1,000
PD-L282723SCell Signaling Technology, Inc.1:1,000
GAPDHMAB376EMD Millipore   1:100,000

B, Secondary antibody

Dilution

NameCat. no. ManufacturerWestern blotting Immuno-fluorescence

Goat anti-mouse HRP6421040Invitrogen; Thermo Fisher Scientific, Inc.1:50,000
Goat anti-rabbit HRP656120Invitrogen; Thermo Fisher Scientific, Inc.1:50,000
Alexafluor 488 anti-mouseA21202Thermo Fisher Scientific, Inc.1:200
Alexafluor 594 anti-rabbitA21207Thermo Fisher Scientific, Inc.1:200

[i] PD-L, programmed death receptor ligand; HRP, horseradish peroxidase; -, not applicable.

Immunofluorescence

Cells (5×103 cells/well) were cultured in 8-well tissue culture treated chamber slides. Cells were washed with 1X PBS and then fixed with 4% paraformaldehyde (Thermo Fisher Scientific, Inc.) at room temperature for 20 min followed by treatment with 0.1% Triton X-100 (Sigma-Aldrich; Merck KGaA). The cells were then blocked with 5% FBS at room temperature for 1 h, followed by overnight incubation with primary antibodies at 4°C. The cells were incubated with secondary antibodies at room temperature for 1 h, followed by 10 min incubation with DAPI (Thermo Fisher Scientific, Inc.) at room temperature. All antibodies used are listed in Table II, including the dilutions, cat. nos. and suppliers. The slides were then mounted in ProLong® Diamond (Thermo Fisher Scientific, Inc.) for observation with a Nikon C2 confocal microscope (magnification, ×40), and captured and analyzed with NIS-Elements-AR software (version 5.01.00; Nikon Corporation).

SurvExpress bioinformatics tool

An online tool, SurvExpress, was used to generate survival analysis using datasets of patients with HCC, as previously described (8,28) A total of six publically available datasets of patients with HCC were used: Hoshida Golub Liver GSE10143 (162 patients), Tsuchiya Rusyn Liver GSE17856 (95 patients), Hoshida Golub Liver GSE10186 (118 patients), Liver Hepatocellular Carcinoma The Cancer Genome Atlas (TCGA; 12 patients), TCGA-Liver-Cancer (422 patients) and LIHC-TCGA-Liver hepatocellular carcinoma June 2016 (361 patients) (8,2831). The gene expression of immune checkpoint molecules in combination with TNF-α were examined and their association with the survival of patients was analyzed (Cox regression analyses) using the patient database with patient survival information (8). The survival times were estimated using Kaplan-Meier curves (8).

Statistical analysis

All experiments were repeated at least three times and representative results are presented. Statistical analyses were performed using Prism software version 8.00 (GraphPad Software, Inc.). Data for dose concentration and time course experiments were analyzed using one-way ANOVA followed by Dunnet's multiple comparisons test. Comparisons of TNF-α-induced EMT and MET following reversal assay were performed using ANOVA followed by Sidak's multiple comparisons test. Gene expression differences between control and TNF-α treated cells were analyzed using Student's t-test. The results are presented as the mean ± SEM. Error bars indicate SEM. P<0.05 was considered to indicate a statistically significant difference.

For survival analyses using SurvExpress, a log-rank test was used for testing the P-value of survival curves, and deviance residuals were applied for the correlation coefficient (8,32). Moreover, the hazard ratio (HR) between the groups were estimated using another Cox model (8).

Results

TNF-α induces EMT in human HCC cells

Several cytokines are known to induce EMT in HCC cells, including TNF-α. In order to induce EMT, Hep3B and PLC/PRF/5 cells were treated with various concentrations of TNF-α for 72 h to assess EMT induction. EMT induced by TNF-α in Hep3B cells was determined by downregulation of epithelial markers (E-cadherin and Occludin) and upregulation of mesenchymal markers [N-cadherin, Vimentin, snail family transcriptional repressor 2 (Snai2) and Fibronectin]. The optimal EMT effects of TNF-α were observed at 20 ng/ml (Fig. 1). Thus, the concentration of 20 ng/ml was selected for all further studies. Furthermore, Hep3B cells were treated with 20 ng/ml TNF-α at various time-points to observe optimal EMT induction. The qPCR results showed that TNF-α treatment at various time-points was able to induce EMT in Hep3B cells (Fig. 2). At 72 h, robust EMT marker changes were observed; therefore, this time-point was selected for further study. Similarly, robust EMT in PLC/PRF/5 cells treated with 20 ng/ml of TNF-α at 72 h was observed. EMT induction was demonstrated by the downregulation of epithelial markers (E-cadherin and Occludin) and an upregulation of mesenchymal markers (N-cadherin and Vimentin; Fig. S1). No expression of Snai2 and Fibronectin was detected in this cell line. The ability of TNF-α to induce EMT in HCC was further validated at the protein level by western blot analysis in Hep3B (Fig. 3A) and PLC/PRF/5 (Fig. 3B) cells.

TNF-α-mediated EMT upregulates immune checkpoint expression

To examine whether TNF-α-mediated EMT influenced the expression of immune modulators, Hep3B and PLC/PRF/5 cells were treated with 20 ng/ml TNF-α for 72 h. The cells were then evaluated for expression of immune modulators PD-L1, PD-L2, CD73, B7-H3, V-set domain-containing T-cell activation inhibitor 1 (VTCN1) and V-domain immunoglobulin suppressor of T-cell activation (VISTA). These immune regulators were assessed as our group previously demonstrated that these immune checkpoint molecules are associated with a poor prognosis in patients with HCC (8). The qPCR results revealed upregulation of four immune modulators (PD-L1, PD-L2, CD73 and B7-H3) and downregulation of two immune modulators (VTCN1 and VISTA) upon treatment with TNF-α in Hep3B cells (Fig. 4A and B). The upregulation of immune checkpoints by TNF-α-mediated EMT in Hep3B cells was validated at the protein level by western blot analysis (Fig. 4C).

Similarly, upregulation of immune modulators PD-L1, CD73 and B7-H3 was observed upon treatment with TNF-α in PLC/PRF/5 cells (Fig. 5A). PD-L2, VTCN1 and VISTA were not detected in PLC/PRF/5 cells. This upregulation of immune modulators by TNF-α in PLC/PRF/5 cells was further confirmed by western blot analysis (Fig. 5B). This demonstrated that TNF-α has a potentially important role in modulating immune checkpoints and EMT in HCC.

Reversal of TNF-α-mediated EMT reverses immune checkpoint expression

In order to assess the association between TNF-α-mediated EMT and immune modulator expression, an EMT reversal assay was performed. The induction of MET was evidenced by the increase in epithelial markers (E-cadherin and Occludin) and a decrease in mesenchymal markers (N-cadherin, Vimentin, Snai2 and Fibronectin) in Hep3B cells (Fig. 6A and B). Similar MET was observed in PLC/PRF/5 cells, with an increase in epithelial markers (E-cadherin and Occludin) and a decrease in mesenchymal markers (N-cadherin and Vimentin; Fig. 6C and D). The changes in the EMT status observed by the reversal assay in Hep3B cells was further confirmed by immunofluorescence and western blot analysis (Fig. 7). The reversal of EMT status in the reversal assay in PLC/PRF/5 cells was also demonstrated by immunofluorescence and western blot analysis (Fig. S2).

The expression of immune checkpoints was also detected following the reversal assay. Downregulation of PD-L1, PD-L2, CD73 and B7-H3 was observed in Hep3B cells upon removal of TNF-α (Fig. 8A). Downregulation of PD-L1, CD73 and B7-H3 was additionally observed in PLC/PRF/5 cells upon removal of TNF-α (Fig. 8B). The reversal in immune checkpoint expression upon reversal of TNF-α-mediated EMT suggested that TNF-α is involved in regulation of immune checkpoint expression.

To further investigate the association between TNF-α-mediated EMT and PD-L1 expression, immunofluorescence and western blot analysis were performed upon TNF-α treatment followed by the reversal assay. The immunofluorescence staining demonstrated upregulated expression of PD-L1 and CD73 during TNF-α-induced EMT and decreased expression upon reversal of EMT in Hep3B cells (Fig. 9A and B). In addition, western blot analysis showed increased expression of PD-L1, CD73, B7-H3 and PD-L2 during TNF-α-induced EMT and decreased expression upon reversal of EMT in Hep3B cells (Fig. 9C).

Furthermore, the immunofluorescence staining showed elevated expression of PD-L1 and CD73 during TNF-α-induced EMT and decreased expression upon reversal of EMT in PLC/PRF/5 cells (Fig. 10A and B). Moreover, western blot analysis showed increased expression of PD-L1, CD73 and B7-H3 during TNF-α-induced EMT and decreased expression upon reversal of EMT in PLC/PRF/5 cells (Fig. 10C). Overall, the present data demonstrated that TNF-α simultaneously induced EMT and immune checkpoint expression in HCC cells.

Coordinate expression of TNF-α and immune modulators in HCC

A total of six different HCC datasets within SurvExpress were used to analyze the recurrence free survival and OS of immune checkpoint molecules in combination with TNF-α in HCC patients. It was observed that coordinate expression of PD-L1 in combination with TNF-α resulted in a significantly worse OS in 422 patients [HR: 1.45; confidence interval (CI): 1.03–2.02; Log-Rank Equal Curves P=0.02948; Fig. 11A]. Similarly, PD-L2 when combined with TNF-α showed a significantly worse recurrence free survival (HR: 1.44; CI: 1.00–2.06; Log-Rank Equal Curves P=0.04606; Fig. 11B). There was no significant difference in recurrence free survival and OS in patients with HCC who showed coordinate expression of TNF-α with CD73 or B7-H3 (data not shown). The present data suggested a significant association between TNF-α and immune checkpoint expression in patients with HCC, as they showed an adverse prognosis with this profile.

Discussion

In the present study, an association between EMT and immune checkpoint molecules in HCC was identified. It was demonstrated that TNF-α simultaneously induced EMT and the expression of immune checkpoint molecules PD-L1, PD-L2, CD73 and B7-H3 in Hep3B cells, along with increased expression of PD-L1, CD73 and B7-H3 in PLC/PRF/5 cells. Moreover, it was demonstrated that EMT status is closely associated with immune checkpoint upregulation as MET induced by the reversal of TNF-α-mediated EMT attenuated the expression of immune checkpoints. Notably, a significant association between TNF-α and immune checkpoint levels was identified in patients with HCC. Patients with increased expression of both TNF-α and PD-L1 showed poor OS, whereas patients with high expression of TNF-α and PD-L2 showed high rates of recurrence.

Various systems have been utilized to induce EMT in human HCC cell lines (33,34). Stimulation of HCC cells with TNF-α leads to the induction of EMT (35). TNF-α is known to induce EMT alone or in combination with other cytokines such as transforming growth factor (TGF)-β in several cancer types, such as breast cancer, lung cancer and HCC (19,21,25). In the present study, a TNF-α based in vitro model was used to induce EMT in a reversible manner. To the best of the authors' knowledge, the present study is the first study to evaluate the role of EMT in the regulation of immune checkpoint expression in HCC.

Despite the promising results of ICIs from clinical trials, these therapies have failed in several instances due to mutations that alter immunogenicity, expression of alternative immune checkpoint molecules and dysregulated T-cell infiltration (36). Understanding the underlying molecular biological mechanisms regulating immune checkpoints may result in developing new and effective treatment strategies. In the present study it was noted that the expression of immune checkpoint molecules PD-L1, PD-L2, CD73 and B7-H3 was upregulated following TNF-α-induced EMT. PD-L1 is an essential and widely studied immunomodulatory ligand that is aberrantly upregulated in several cancer types, has roles in promoting tumor escape and is associated with poor prognosis (3741). Our group and other researchers have reported the association between upregulation of PD-L1 and poor survival in patients with HCC (8,42). Several previous studies have reported different mechanisms involved in the regulation of PD-L1 expression in cancer cells (19,4345). However, the mechanism of PD-L1 expression in HCC still remains unclear (46).

In previous years, previous studies have demonstrated cytokine-induced EMT, in particular TNF-α, TGF-β and interferon-γ are responsible for elevating the expression of PD-L1 in cancer, such as non-small cell lung carcinoma and breast cancer (17,19,21). The present finding that TNF-α is involved in the upregulation of PD-L1 is consistent with these studies. Another previous study demonstrated that PD-L1 was upregulated in EMT positive human esophageal tumor samples compared with the EMT negative samples (23). The relationship between EMT and PD-L1 was further examined in human breast cancer cells by Noman et al (20). This previous study identified increased expression of PD-L1 in cells having undergone EMT by EMT-transcription factors (zinc finger E-box binding homeobox 1, microRNA 200 or Snai1) and PD-L1 rendered EMT-activated cells resistant to cytotoxic T-lymphocytes-mediated lysis (20). In patients with HCC, our group previously identified an association between PD-L1 expression and EMT status (8). High PD-L1 expression was closely associated with high expression of the mesenchymal maker Vimentin and low expression of epithelial marker E-cadherin (8). However, to the best of the authors' knowledge, no studies have reported the association between EMT and other immune checkpoint in HCC.

PD-L2 is a second ligand that binds to PD-1 to prevent cytokine production, cell adhesion and T-cell proliferation (47). Our group and other researchers have previously reported that PD-L2 expression is associated with poor survival and recurrence of HCC in patients (8,48). A previous meta-analysis study by Yang et al (49) identified that upregulation of PD-L2 predicted poor OS in HCC.

CD73 is also reported to be a novel prognostic biomarker for HCC (8,50,51). However, to the best of the authors' knowledge, the regulation of CD73 expression has not been studied in the context of EMT, and the present study is the first to report that TNF-α-induced EMT regulates CD73 expression. In a recent triple-negative breast cancer study by Qiao et al (52), an anti-CD73 antibody was demonstrated to inhibit lung metastasis in vivo.

B7-H3 is mostly upregulated in several cancer types, including liver, breast, bladder, colorectal, cervical, glioma, esophageal and gastric cancer (53). Previous studies have also demonstrated that dysregulation of B7-H3 in HCC results in impaired T-lymphocyte function, thus leading to poor prognosis and recurrence (54,55). In muscle-invasive bladder cancer, high expression of B7-H3 was associated with a poor clinicopathological status and poor prognosis (56). Most notably, B7-H3 is known to promote EMT by repressing E-cadherin expression, and upregulating Vimentin and N-cadherin expression in colorectal cancer (57). To the best of the authors' knowledge, the present study is the first to report the regulation of B7-H3 expression by EMT in HCC.

Further mechanistic and functional studies are necessary to examine the molecular biology of immune checkpoints and its precise role in EMT in HCC. However, given the association between EMT, TNF-α and immune checkpoint in HCC, it is conceivable that combining EMT or TNF-α inhibitors with ICIs in patients with HCC may be a future therapeutic approach for the management of HCC.

Supplementary Material

Supporting Data

Acknowledgements

The authors would like to thank Mrs Lesley-Anne Jaskowski, Gallipoli Medical Research Institute, Greenslopes Private Hospital, for her technical assistance during the revision of the manuscript.

Funding

The present study was funded by Gallipoli Medical Research Foundation (project no. 017401).

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

RS, KRB, DHGC and AJ designed the study, performed data acquisition, analysis and interpretation, and wrote the manuscript. RS, AJ, KRB and DHGC critically revised the manuscript. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Chacko S and Samanta S: ‘Hepatocellular carcinoma: A life-threatening disease’. Biomed Pharmacother. 84:1679–1688. 2016. View Article : Google Scholar : PubMed/NCBI

2 

Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA and Jemal A: Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 68:394–424. 2018. View Article : Google Scholar : PubMed/NCBI

3 

Sayiner M, Golabi P and Younossi ZM: Disease burden of hepatocellular carcinoma: A global perspective. Dig Dis Sci. 64:910–917. 2019. View Article : Google Scholar : PubMed/NCBI

4 

Kudo M: A new era of systemic therapy for hepatocellular carcinoma with regorafenib and lenvatinib. Liver Cancer. 6:177–184. 2017. View Article : Google Scholar : PubMed/NCBI

5 

Mir N, Jayachandran A, Dhungel B, Shrestha R and Steel JC: Epithelial-to-mesenchymal transition: A Mediator of sorafenib resistance in advanced hepatocellular carcinoma. Curr Cancer Drug Targets. 17:698–706. 2017. View Article : Google Scholar : PubMed/NCBI

6 

Sharma P and Allison JP: The future of immune checkpoint therapy. Science. 348:56–61. 2015. View Article : Google Scholar : PubMed/NCBI

7 

Pardoll DM: The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer. 12:252–264. 2012. View Article : Google Scholar : PubMed/NCBI

8 

Shrestha R, Prithviraj P, Anaka M, Bridle KR, Crawford DH, Dhungel B, Steel JC and Jayachandran A: Monitoring immune checkpoint regulators as predictive biomarkers in hepatocellular carcinoma. Front Oncol. 8:2692018. View Article : Google Scholar : PubMed/NCBI

9 

Hato T, Goyal L, Greten TF, Duda DG and Zhu AX: Immune checkpoint blockade in hepatocellular carcinoma: Current progress and future directions. Hepatology. 60:1776–1782. 2014. View Article : Google Scholar : PubMed/NCBI

10 

Brahmer J, Reckamp KL, Baas P, Crinò L, Eberhardt WE, Poddubskaya E, Antonia S, Pluzanski A, Vokes EE, Holgado E, et al: Nivolumab versus docetaxel in advanced squamous-cell non-small-cell lung cancer. N Engl J Med. 373:123–135. 2015. View Article : Google Scholar : PubMed/NCBI

11 

Weber JS, D'Angelo SP, Minor D, Hodi FS, Gutzmer R, Neyns B, Hoeller C, Khushalani NI, Miller WH Jr, Lao CD, et al: Nivolumab versus chemotherapy in patients with advanced melanoma who progressed after anti-CTLA-4 treatment (CheckMate 037): A randomised, controlled, open-label, phase 3 trial. Lancet Oncol. 16:375–384. 2015. View Article : Google Scholar : PubMed/NCBI

12 

Jindal A, Thadi A and Shailubhai K: Hepatocellular carcinoma: Etiology and current and future drugs. J Clin Exp Hepatol. 9:221–232. 2019. View Article : Google Scholar : PubMed/NCBI

13 

Kudo M: Immune checkpoint inhibition in hepatocellular carcinoma: Basics and ongoing clinical trials. Oncology. 92 (Suppl 1):50–62. 2017. View Article : Google Scholar : PubMed/NCBI

14 

Li Z, Li N, Li F, Zhou Z, Sang J, Chen Y, Han Q, Lv Y and Liu Z: Immune checkpoint proteins PD-1 and TIM-3 are both highly expressed in liver tissues and correlate with their gene polymorphisms in patients with HBV-related hepatocellular carcinoma. Medicine (Baltimore). 95:e57492016. View Article : Google Scholar : PubMed/NCBI

15 

Kalluri R and Weinberg RA: The basics of epithelial-mesenchymal transition. J Clin Invest. 119:1420–1428. 2009. View Article : Google Scholar : PubMed/NCBI

16 

Jayachandran A, Dhungel B and Steel JC: Epithelial-to-mesenchymal plasticity of cancer stem cells: Therapeutic targets in hepatocellular carcinoma. J Hematol Oncol. 9:742016. View Article : Google Scholar : PubMed/NCBI

17 

David JM, Dominguez C, McCampbell KK, Gulley JL, Schlom J and Palena C: A novel bifunctional anti-PD-L1/TGF-β Trap fusion protein (M7824) efficiently reverts mesenchymalization of human lung cancer cells. OncoImmunology. 6:e13495892017. View Article : Google Scholar : PubMed/NCBI

18 

David JM, Dominguez C and Palena C: Pharmacological and immunological targeting of tumor mesenchymalization. Pharmacol Ther. 170:212–225. 2017. View Article : Google Scholar : PubMed/NCBI

19 

Alsuliman A, Colak D, Al-Harazi O, Fitwi H, Tulbah A, Al-Tweigeri T, Al-Alwan M and Ghebeh H: Bidirectional crosstalk between PD-L1 expression and epithelial to mesenchymal transition: Significance in claudin-low breast cancer cells. Mol Cancer. 14:1492015. View Article : Google Scholar : PubMed/NCBI

20 

Noman MZ, Janji B, Abdou A, Hasmim M, Terry S, Tan TZ, Mami-Chouaib F, Thiery JP and Chouaib S: The immune checkpoint ligand PD-L1 is upregulated in EMT-activated human breast cancer cells by a mechanism involving ZEB-1 and miR-200. Oncoimmunology. 6:e12634122017. View Article : Google Scholar : PubMed/NCBI

21 

Asgarova A, Asgarov K, Godet Y, Peixoto P, Nadaradjane A, Boyer-Guittaut M, Galaine J, Guenat D, Mougey V, Perrard J, et al: PD-L1 expression is regulated by both DNA methylation and NF-κB during EMT signaling in non-small cell lung carcinoma. Oncoimmunology. 7:e14231702018. View Article : Google Scholar : PubMed/NCBI

22 

Imai D, Yoshizumi T, Okano S, Itoh S, Ikegami T, Harada N, Aishima S, Oda Y and Maehara Y: IFN-γ promotes epithelial-mesenchymal transition and the expression of PD-L1 in pancreatic cancer. J Surg Res. 240:115–123. 2019. View Article : Google Scholar : PubMed/NCBI

23 

Chen L, Xiong Y, Li J, Zheng X, Zhou Q, Turner A, Wu C, Lu B and Jiang J: PD-L1 expression promotes epithelial to mesenchymal transition in human esophageal cancer. Cell Physiol Biochem. 42:2267–2280. 2017. View Article : Google Scholar : PubMed/NCBI

24 

Wang Y, Hu J, Wang Y, Ye W, Zhang X, Ju H, Xu D, Liu L, Ye D, Zhang L, et al: EGFR activation induced Snail-dependent EMT and myc-dependent PD-L1 in human salivary adenoid cystic carcinoma cells. Cell Cycle. 17:1457–1470. 2018. View Article : Google Scholar : PubMed/NCBI

25 

Chen Y, Wen H, Zhou C, Su Q, Lin Y, Xie Y, Huang Y, Qiu Q, Lin J, Huang X, et al: TNF-α derived from M2 tumor-associated macrophages promotes epithelial-mesenchymal transition and cancer stemness through the Wnt/β-catenin pathway in SMMC-7721 hepatocellular carcinoma cells. Exp Cell Res. 378:41–50. 2019. View Article : Google Scholar : PubMed/NCBI

26 

Jayachandran A, Shrestha R, Dhungel B, Huang IT, Vasconcelos MY, Morrison BJ, Ramlogan-Steel CA and Steel JC: Murine hepatocellular carcinoma derived stem cells reveal epithelial-to-mesenchymal plasticity. World J Stem Cells. 9:159–168. 2017. View Article : Google Scholar : PubMed/NCBI

27 

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

28 

Aguirre-Gamboa R, Gomez-Rueda H, Martínez-Ledesma E, Martínez-Torteya A, Chacolla-Huaringa R, Rodriguez-Barrientos A, Tamez-Peña JG and Treviño V: SurvExpress: An online biomarker validation tool and database for cancer gene expression data using survival analysis. PLoS One. 8:e742502013. View Article : Google Scholar : PubMed/NCBI

29 

Hoshida Y, Nijman SM, Kobayashi M, Chan JA, Brunet JP, Chiang DY, Villanueva A, Newell P, Ikeda K, Hashimoto M, et al: Integrative transcriptome analysis reveals common molecular subclasses of human hepatocellular carcinoma. Cancer Res. 69:7385–7392. 2009. View Article : Google Scholar : PubMed/NCBI

30 

Hoshida Y, Villanueva A, Kobayashi M, Peix J, Chiang DY, Camargo A, Gupta S, Moore J, Wrobel MJ, Lerner J, et al: Gene expression in fixed tissues and outcome in hepatocellular carcinoma. N Engl J Med. 359:1995–2004. 2008. View Article : Google Scholar : PubMed/NCBI

31 

Tsuchiya M, Parker JS, Kono H, Matsuda M, Fujii H and Rusyn I: Gene expression in nontumoral liver tissue and recurrence-free survival in hepatitis C virus-positive hepatocellular carcinoma. Mol Cancer. 9:742010. View Article : Google Scholar : PubMed/NCBI

32 

Awan FM, Naz A, Obaid A, Ali A, Ahmad J, Anjum S and Janjua HA: Identification of Circulating Biomarker Candidates for Hepatocellular Carcinoma (HCC): An Integrated Prioritization Approach. PLoS One. 10:e01389132015. View Article : Google Scholar : PubMed/NCBI

33 

Lee TK, Poon RT, Yuen AP, Ling MT, Kwok WK, Wang XH, Wong YC, Guan XY, Man K, Chau KL, et al: Twist overexpression correlates with hepatocellular carcinoma metastasis through induction of epithelial-mesenchymal transition. Clin Cancer Res. 12:5369–5376. 2006. View Article : Google Scholar : PubMed/NCBI

34 

Xu Z, Shen MX, Ma DZ, Wang LY and Zha XL: TGF-beta1-promoted epithelial-to-mesenchymal transformation and cell adhesion contribute to TGF-beta1-enhanced cell migration in SMMC-7721 cells. Cell Res. 13:343–350. 2003. View Article : Google Scholar : PubMed/NCBI

35 

Zhu Y, Cheng Y, Guo Y, Chen J, Chen F, Luo R and Li A: Protein kinase D2 contributes to TNF-α-induced epithelial mesenchymal transition and invasion via the PI3K/GSK-3β/β-catenin pathway in hepatocellular carcinoma. Oncotarget. 7:5327–5341. 2016.PubMed/NCBI

36 

Xu F, Jin T, Zhu Y and Dai C: Immune checkpoint therapy in liver cancer. J Exp Clin Cancer Res. 37:1102018. View Article : Google Scholar : PubMed/NCBI

37 

Afreen S and Dermime S: The immunoinhibitory B7-H1 molecule as a potential target in cancer: Killing many birds with one stone. Hematol Oncol Stem Cell Ther. 7:1–17. 2014. View Article : Google Scholar : PubMed/NCBI

38 

Mu CY, Huang JA, Chen Y, Chen C and Zhang XG: High expression of PD-L1 in lung cancer may contribute to poor prognosis and tumor cells immune escape through suppressing tumor infiltrating dendritic cells maturation. Med Oncol. 28:682–688. 2011. View Article : Google Scholar : PubMed/NCBI

39 

Muenst S, Schaerli AR, Gao F, Däster S, Trella E, Droeser RA, Muraro MG, Zajac P, Zanetti R, Gillanders WE, et al: Expression of programmed death ligand 1 (PD-L1) is associated with poor prognosis in human breast cancer. Breast Cancer Res Treat. 146:15–24. 2014. View Article : Google Scholar : PubMed/NCBI

40 

Shi SJ, Wang LJ, Wang GD, Guo ZY, Wei M, Meng YL, Yang AG and Wen WH: B7-H1 expression is associated with poor prognosis in colorectal carcinoma and regulates the proliferation and invasion of HCT116 colorectal cancer cells. PLoS One. 8:e760122013. View Article : Google Scholar : PubMed/NCBI

41 

Thompson RH, Gillett MD, Cheville JC, Lohse CM, Dong H, Webster WS, Krejci KG, Lobo JR, Sengupta S, Chen L, et al: Costimulatory B7-H1 in renal cell carcinoma patients: Indicator of tumor aggressiveness and potential therapeutic target. Proc Natl Acad Sci USA. 101:17174–17179. 2004. View Article : Google Scholar : PubMed/NCBI

42 

Calderaro J, Rousseau B, Amaddeo G, Mercey M, Charpy C, Costentin C, Luciani A, Zafrani ES, Laurent A, Azoulay D, et al: Programmed death ligand 1 expression in hepatocellular carcinoma: Relationship with clinical and pathological features. Hepatology. 64:2038–2046. 2016. View Article : Google Scholar : PubMed/NCBI

43 

Crane CA, Panner A, Murray JC, Wilson SP, Xu H, Chen L, Simko JP, Waldman FM, Pieper RO and Parsa AT: PI(3) kinase is associated with a mechanism of immunoresistance in breast and prostate cancer. Oncogene. 28:306–312. 2009. View Article : Google Scholar : PubMed/NCBI

44 

Dey N, Crosswell HE, De P, Parsons R, Peng Q, Su JD and Durden DL: The protein phosphatase activity of PTEN regulates SRC family kinases and controls glioma migration. Cancer Res. 68:1862–1871. 2008. View Article : Google Scholar : PubMed/NCBI

45 

Ghebeh H, Tulbah A, Mohammed S, Elkum N, Bin Amer SM, Al-Tweigeri T and Dermime S: Expression of B7-H1 in breast cancer patients is strongly associated with high proliferative Ki-67-expressing tumor cells. Int J Cancer. 121:751–758. 2007. View Article : Google Scholar : PubMed/NCBI

46 

Funaki S, Shintani Y, Kawamura T, Kanzaki R, Minami M and Okumura M: Chemotherapy enhances programmed cell death 1/ligand 1 expression via TGF-β induced epithelial mesenchymal transition in non-small cell lung cancer. Oncol Rep. 38:2277–2284. 2017. View Article : Google Scholar : PubMed/NCBI

47 

Latchman Y, Wood CR, Chernova T, Chaudhary D, Borde M, Chernova I, Iwai Y, Long AJ, Brown JA, Nunes R, et al: PD-L2 is a second ligand for PD-1 and inhibits T cell activation. Nat Immunol. 2:261–268. 2001. View Article : Google Scholar : PubMed/NCBI

48 

Jung HI, Jeong D, Ji S, Ahn TS, Bae SH, Chin S, Chung JC, Kim HC, Lee MS and Baek MJ: Overexpression of PD-L1 and PD-L2 is associated with poor prognosis in patients with hepatocellular carcinoma. Cancer Res Treat. 49:246–254. 2017. View Article : Google Scholar : PubMed/NCBI

49 

Yang H, Zhou X, Sun L and Mao Y: Correlation Between PD-L2 expression and clinical outcome in solid cancer patients: A meta-analysis. Front Oncol. 9:472019. View Article : Google Scholar : PubMed/NCBI

50 

Ma XL, Shen MN, Hu B, Wang BL, Yang WJ, Lv LH, Wang H, Zhou Y, Jin AL, Sun YF, et al: CD73 promotes hepatocellular carcinoma progression and metastasis via activating PI3K/AKT signaling by inducing Rap1-mediated membrane localization of P110β and predicts poor prognosis. J Hematol Oncol. 12:372019. View Article : Google Scholar : PubMed/NCBI

51 

Sciarra A, Monteiro I, Ménétrier-Caux C, Caux C, Gilbert B, Halkic N, La Rosa S, Romero P, Sempoux C and de Leval L: CD73 expression in normal and pathological human hepatobiliopancreatic tissues. Cancer Immunol Immunother. 68:467–478. 2019. View Article : Google Scholar : PubMed/NCBI

52 

Qiao Z, Li X, Kang N, Yang Y, Chen C, Wu T, Zhao M, Liu Y and Ji X: A Novel specific Anti-CD73 antibody inhibits triple-negative breast cancer cell motility by regulating autophagy. Int J Mol Sci. 20:E10572019. View Article : Google Scholar : PubMed/NCBI

53 

Dong P, Xiong Y, Yue J, Hanley SJB and Watari H: B7H3 as a promoter of metastasis and promising therapeutic target. Front Oncol. 8:2642018. View Article : Google Scholar : PubMed/NCBI

54 

Kang FB, Wang L, Jia HC, Li D, Li HJ, Zhang YG and Sun DX: B7-H3 promotes aggression and invasion of hepatocellular carcinoma by targeting epithelial-to-mesenchymal transition via JAK2/STAT3/Slug signaling pathway. Cancer Cell Int. 15:452015. View Article : Google Scholar : PubMed/NCBI

55 

Zheng Y, Liao N, Wu Y, Gao J, Li Z, Liu W, Wang Y, Li M, Li X, Chen L, et al: High expression of B7 H2 or B7 H3 is associated with poor prognosis in hepatocellular carcinoma. Mol Med Rep. 19:4315–4325. 2019.PubMed/NCBI

56 

Xu ZL, Zhang Y, Wang L, Li F, Man HW, Li PF and Shan BE: B7 H3 promotes malignant progression of muscle invasive bladder cancer. Oncol Rep. 40:2722–2733. 2018.PubMed/NCBI

57 

Jiang B, Zhang T, Liu F, Sun Z, Shi H, Hua D and Yang C: The co-stimulatory molecule B7-H3 promotes the epithelial-mesenchymal transition in colorectal cancer. Oncotarget. 7:31755–31771. 2016.PubMed/NCBI

Related Articles

Journal Cover

April-2020
Volume 21 Issue 4

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Shrestha R, Bridle KR, Crawford DH and Jayachandran A: TNF‑α‑mediated epithelial‑to‑mesenchymal transition regulates expression of immune checkpoint molecules in hepatocellular carcinoma. Mol Med Rep 21: 1849-1860, 2020
APA
Shrestha, R., Bridle, K.R., Crawford, D.H., & Jayachandran, A. (2020). TNF‑α‑mediated epithelial‑to‑mesenchymal transition regulates expression of immune checkpoint molecules in hepatocellular carcinoma. Molecular Medicine Reports, 21, 1849-1860. https://doi.org/10.3892/mmr.2020.10991
MLA
Shrestha, R., Bridle, K. R., Crawford, D. H., Jayachandran, A."TNF‑α‑mediated epithelial‑to‑mesenchymal transition regulates expression of immune checkpoint molecules in hepatocellular carcinoma". Molecular Medicine Reports 21.4 (2020): 1849-1860.
Chicago
Shrestha, R., Bridle, K. R., Crawford, D. H., Jayachandran, A."TNF‑α‑mediated epithelial‑to‑mesenchymal transition regulates expression of immune checkpoint molecules in hepatocellular carcinoma". Molecular Medicine Reports 21, no. 4 (2020): 1849-1860. https://doi.org/10.3892/mmr.2020.10991