Exogenous hydrogen sulfide exerts proliferation/anti-apoptosis/angiogenesis/migration effects via amplifying the activation of NF-κB pathway in PLC/PRF/5 hepatoma cells

  • Authors:
    • Yulan Zhen
    • Wanying Pan
    • Fen Hu
    • Hongfu Wu
    • Jianqiang Feng
    • Ying Zhang
    • Jingfu Chen
  • View Affiliations

  • Published online on: March 3, 2015     https://doi.org/10.3892/ijo.2015.2914
  • Pages: 2194-2204
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Hydrogen sulfide (H2S) takes part in a diverse range of intracellular pathways and hss physical and pathological properties in vitro and in vivo. However, the effects of H2S on cancer are controversial and remain unclear. The present study investigates the effects of H2S on liver cancer progression via activating NF-κB pathway in PLC/PRF/5 hepatoma cells. PLC/PRF/5 hepatoma cells were pretreated with 500 µmol/l NaHS (a donor of H2S) for 24 h. The expression levels of CSE, CBS, phosphosphorylate (p)-NF-κB p65, caspase-3, COX-2, p-IκB and MMP-2 were measured by western blot assay. Cell viability was detected by cell counter kit 8 (CCK-8). Apoptotic cells were observed by Hoechst 33258 staining assay. The production level of H2S in cell culture medium was measured by using the sulfur-sensitive electrode method. The production of vascular endothelial growth factor (VEGF) was tested by enzyme-linked immunosorbent assay (ELISA). Our results showed that the production of H2S was dramatically increased in the PLC/PRF/5 hepatoma cells, compared with human LO2 hepatocyte cells group, along with the overexpression levels of CSE and CBS. Treatment of PLC/PRF/5 hepatoma cells with 500 µmol/l NaHS (a donor of H2S) for 24 h markedly increased the expression levels of CSE, CBS, p-IκB and NF-κB activation, leading to COX-2 and MMP-2 overexpression, and decreased caspase-3 production, as well as increased cell viability and decreased number of apoptotic cells. Otherwise, the production level of H2S and VEGF were also significantly increased. Furthermore, co-treatment of PLC/PRF/5 hepatoma cells with 500 µmol/l NaHS and 200 µmol/l PDTC for 24 h significantly overturned these indexes. The findings of the present study provide evidence that the NF-κB is involved in the NaHS-induced cell proliferation, anti-apoptisis, angiogenesis, and migration in PLC/PRF/5 hepatoma cells, and that the PDTC against the NaHS-induced effects were by inhibition of the NF-κB pathway.

Introduction

The incidence and mortality of primary liver cancer (PLC) are increasing year by year (1). PLC is one of the most common malignant tumors in China, the occurrence of liver cancer is approximately one million people all around the world each year and most of them occur in mainland of China, accounting for 54% of all cases worldwide (2). PLC contains mainly three subcategories: hepatocellular carcinoma (HCC), intrahepatic bile duct carcinoma (IHBD) and hybrid liver cancer. HCC is the major form of PLC and is the sixth most common neoplasm in the world (3). Over the past several decades, a series of risk factors for HCC have been established (47), including hepatitis B virus (HBV) or hepatitis C virus (HCV) infection, aflatoxin exposure, and tobacco smoking. However, the mechanisms of the oncogenesis of HCC have not been completely described. Recently, several studies have strongly suggested that nuclear factor-κB (NF-κB) may play a pivotal role in this pathophysiological process (811).

NF-κB, which contains five subunits [RelA (p65), C-Rel, NF-κB1 (P50/P105), RelB and NF-κB2 (P52/P100)], is a major transcription regulator of the cell functions, including adhesion, immune response, cell invasion, cell differentiation, cell proliferation and apoptosis (12). In the resting cells, NF-κB is a dimer and is inactivated retentively in the cytoplasm via binding to specific inhibitor of NF-κB: IκB family. However, NF-κB can be activated by various stimuli (1318). First, IκB is phosphorylated, and NF-κB is activated and released from its IκB-bound complex. In addition, NF-κB translocates into the nucleus from cytoplasm. The activated NF-κB (p65) binds to κB sequences and alters the expression of various target genes (13). The above physiologic process also occurred in different tumor tissues or in cancer cells (1418), such as breast cells, and especially in liver cancer cells (19), indicating that NF-κB is involved in oncogenesis process of liver cancer.

Hydrogen sulfide (H2S), is an unusual virulent gas, and has been qualified as the third gasotransmitter following nitric oxide (NO) and carbon monoxide (CO) (2022). H2S can be endogenously produced mainly by 3-mercaptopyruvate sulfurtransferase (3-MST), cystathionineb-synthase (CBS) or cystathionine-γ-lyase (CSE) (23,24). The expression of CSE and CBS is distinctly tissue-specific. CBS is mainly found in the central nervous system (CNS), and CSE mostly in the cardiovascular system. Interestingly, both CBS and CSE have been simultaneously identified in some systems, including kidney, liver, intestine and brain (25). In recent years, more attention is paid to H2S for its extensive physiological and pathophysiological properties. Accumulating studies have demonstrated that H2S can exert cardioprotection (2631), angiogenesis (3234), antioxidant (35), pro- and anti-inflammatory (36,37) and other wide range of physiological functions (3841). Furthermore, increasing evidence has shown that H2S is involved in the pathophysiological process of tumors (4252). Notably, the effects of H2S on cancer are still controversial. On the one hand, some findings from in vivo and in vitro studies showed that H2S is beneficial for cancer cell growth, proliferation, migration, and invasion (4248), owing to its angiogenesis and vascular relaxant effects, H2S promotes the supply of nutrients and blood to the tumor cells and tissues (42). Nevertheless, on the other hand it was found that H2S exerted its potential anticancer effects on SGC-7901 gastric cancer cells (49), oral cancer cell lines (50), colon cancer cell (51) and several different human cancer cell lines (HeLa, HCT-116, Hep G2, HL-60, MCF-7, MV4-11 and U2OS) (52). Thus, the effects of H2S on cancer are complicated and still unclear. Furthermore, to our knowledge, no study has been focused on the effect of exogenous H2S on liver cancer cells and its mechanisms. Based on recent studies (4248), we investigated whether exogenous H2S can contribute to cancer progress and explored these potential effects via amplification of NF-κB pathway in PLC/PRF/5 hepatoma cells.

Materials and methods

Materials

NaHS, a donor of H2S, was obtained from Sigma Chemical Co. (St. Louis, MO, USA), stored at 2–4°C and protected from sunlight. Hoechst 33258 and PDTC were also purchased from Sigma Chemical Co. The cell counter kit-8 (CCK-8) was supplied by Dojindo Lab (Kumamoto, Japan). Fetal bovine serum (FBS) and RPMI-1640 medium were obtained from Gibco BRL (Grand Island, NY, USA). Anti-MMP2 antibody, anti-COX-2 antibody, anti-p-IκB antibody, anti-NF-κB p65 antibody and anti-p-NF-κB p65 antibody were supplied by Cell Signaling Technology (Boston, MA, USA). Horseradish peroxidase (HRP)-conjugated secondary antibody and BCA protein assay kit were obtained from KangChen Bio-tech, Inc. (Shanghai, China). Enhanced chemiluminescence (ECL) solution was purchased from KeyGen Biotech (Nanjing, China). Enzyme-linked immunosorbent assay (ELISA) was supplied by ExCell Bio Co. (Shanghai, China). A sulfur-sensitive electrode was obtained from Fuji Electric (ELIT 8225, ISEIonometer, EA Instruments Ltd., UK).

Cell culture and treatments

The human hepatoma cells PLC (PLC) and the Lo2 cells (LO2) were supplied by Sun Yat-sen University Experimental Animal Center (Guangzhou, Guangdong, China). The PLC cells and LO2 cells were grown in RPMI-1640 medium supplemented with 10% fetal bovine serum under an atmosphere of 5% CO2 and at 37°C with 95% air. The PLC cells were treatment with 500 μmol/l NaHS for 24 h or co-treatment with 500 μmol/l NaHS and 200 μmol/l PDTC for 24 h.

Western blot analysis

After the indicated treatments, the cells were harvested and lysed with cell lysis solution at 4°C for 30 min. The total proteins were quantified through using the BCA protein assay kit. Loading buffer was added to cytosolic extracts, then boiled for 6 min, the same amounts of supernatant from each sample were fractionated by 10% sodium dodecyl sulphate-polyacrylamide gel electrophoresis (SDS-PAGE), and the total proteins were transferred into polyvinylidene difluoride (PVDF) membranes. The membranes were blocked with 5% fat-free milk for 60 min in fresh blocking buffer [0.1% Tween-20 in Tris-buffered saline (TBS-T)] at room temperature, and incubated with either anti-MMP2 antibody (1:1,000 dilution), anti-COX-2 antibody (1:1,000 dilution), anti-p-IκB antibody (1:1,000 dilution), anti-NF-κB p65 antibody (1:1,000 dilution), and anti-p-NF-κB p65 antibody (1:1,000 dilution) in freshly prepared TBS-T with 3% free-fat milk overnight with gentle agitation at 4°C. Membranes were washed for 5 min with TBS-T three times and incubated with HRP-conjugated goat anti-rabbit secondary antibody at a concentration of 1:3,000 dilution (KangChen Bio-tech), in TBS-T with 3% fat-free milk for 1.5 h at room temperature. Then membranes were washed three times with TBS-T for 5 min. The immunoreactive signals were visualized via using the ECL (enhanced chemiluminescence) detection. In order to quantify the protein expression, the X-ray film was scanned and analyzed with ImageJ 1.47i software. The experiment was carried out three times.

Measurement of cell viability

The PLC cells were seeded in 96-well plates at concentration of 1×104/ml, and incubated at 37°C, the CCK-8 assay was employed to assess the cell viability of PLC cells. After the indicated treatments, 10 μl CCK-8 solution at a 1/10 dilution was added to each well and then the plate was incubated for 1.5 h in the incubator. Absorbance at 450 nm was assayed using a microplate reader (Molecular Devices, Sunnyvale, CA, USA). The means of the optical density (OD) of three wells in the indicated groups were used to calculate the percentage of cell viability according to the formula: Cell viability (%) = (OD treatment group/OD control group) × 100%. The experiment was carried out in three times.

Hoechst 33258 nuclear staining for evaluation of apoptosis

Apoptotic cell death was tested by the Hoechst 33258 staining followed by photofluorography. PLC cells were plated in 35-mm dishes at a density of 1×106 cells/well. After the above indicated treatments, the PLC cells were fixed with 4% paraformaldehyde in 0.1 mol/l phosphate-buffered saline (PBS, pH 7.4) for 10 min at 4°C. In addition, the slides were washed three times with PBS. After staining followed by 5 mg/ml Hoechst 33258 for 15 min, the PLC cells were washed three times with PBS. The PLC cells were visualized under a fluorescence microscope (Bx50-FLA; Olympus, Tokyo, Japan). Viable PLC cells displayed a uniform blue fluorescence throughout the nucleus and normal nuclear size. However, apoptotic PLC cells showed condensed, distorted or fractured nuclei. The experiment was carried out three times.

Measurement of cell culture medium H2S levels

A sulfur-sensitive electrode (ELIT 8225, ISEIonometer, EA Instruments Ltd.) was used to measure the production levels of H2S in the cell culture medium. Cell culture medium samples were obtained from all subjects and mixed with an equal volume of antioxidant solution. The total volume covered the electrode; usually >0.8 ml. The electrode was activated in the deionized water for ≥2 h. The modified sulfide electrode and a reference electrode were dipped into the above mixture. The electrode was flushed with the deionized water after sample determination, and the activity state was maintained through dipping the electrode in deionized water. A standard curve was generated using a standard S2-solution, and then the H2S concentration was calculated according to this standard curve.

ELISA for detection of VEGF in culture supernatant

PLC/PRF/5 hepatoma cells were cultured in 96-well plates. After the different indicated treatments, the level of VEGF in the culture media was tested by enzyme-linked immunosorbent assay (ELISA) according to the manufacturer’s instruction. The experiment was performed at least five times.

Statistical analysis

All data are presented as the mean ± SEM. Differences between groups were analyzed by one-way analysis of variance (ANOVA) by using SPSS 13.0 (SPSS, Chicago, IL, USA) software, and followed by LSD post hoc comparison test. Statistical significance was set at P<0.05.

Results

The expression levels of CBS and CSE, and endogenous H2S production in PLC/PRF/5 hepatoma cells

To determine the role of hydrogen sulfide (H2S) in the oncogenesis process of liver cancer, the expression levels of CSE and CBS were detected by western blot assay both in PLC/PRF/5 hepatoma cells (PLC) and in the human hepatic cell lines LO2 (LO2). As shown in Fig. 1A–C, both the expression of CSE (Fig. 1A and B) and CBS (Fig. 1A and C) were significantly upregulated, compared with the LO2 group. In addition, the sulfur-sensitive electrode was used to explore the production level of H2S in the cell culture medium. We found that the production of hydrogen sulfide in PLC/PRF/5 hepatoma cells was distinctly increased, compared with the LO2 group (Fig. 1D). So we hypothesized that endogenous H2S might take part in the development of liver cancer.

NaHS promotes cells proliferation in PLC/PRF/5 hepatoma cells

In order to test the effect of exogenous H2S in human liver cancer cell proliferation, the dose-response study with varying doses (10, 50, 100, 200, 300, 400, 500, 600, 700 and 1,000 μmol/l) of NaHS (a donor of H2S) for 24 h was performed to calculate the effective doses of NaHS. As shown in Fig. 2A, low concentrations of NaHS (10, 50 and 100 μmol/l) did not alter cell viability. Whereas, the doses of NaHS from 200 to 1,000 μmol/l markedly promoted cell proliferation, leading to an increase in cell viability and reaching a peaking at 500 μmol/l. Therefore, 500 μmol/l NaHS was used in the subsequent time-response study with different treatment times (12, 24, 36, 48, 60 and 72 h). As shown in Fig. 2B, treatment of PLC cells with 500 μmol/l NaHS for the indicated times all dramatically promoted cell proliferation, reaching the maximal proliferative effect at 24 h. Based on the above results, PLC/PRF/5 hepatoma cells were treated with 500 μmol/l NaHS for 24 h in all subsequent experiments.

NaHS increases the expression levels of CSE and CBS and production of H2S in PLC/PRF/5 hepatoma cells

We observed the effects of NaHS on the expression levels of CSE and CBS. As shown in Fig. 3A–C, exposure of PLC/PRF/5 hepatoma cells for the indicated time (1, 3, 6, 9, 12 and 24 h) to 500 μmol/l NaHS markedly enhanced the expression levels of CSE and CBS, reaching a peak at 24 h. In addition, at the same time, we found that the production of H2S was significantly increased in the cell culture medium, compared with the control group.

NaHS alleviates the expression level of caspase-3 and upregulates the expression levels of COX-2 and MMP-2 in PLC/PRF/5 hepatoma cells

In order to observe the effects of NaHS on the expression levels of caspase-3, COX-2 and MMP-2 in PLC/PRF/5 hepatoma cells, exposure of PLC/PRF/5 hepatoma cells to 500 μmol/l NaHS for different times (1, 3, 6, 9, 12 and 24 h). As shown in Fig. 4, NaHS significantly enhanced the expression levels of COX-2 and MMP-2, and reaching a peak at 12 h, whereas, the expression level of caspase-3 was markedly decreased.

NaHS amplifies the activation of NF-κB and p-IκBα in PLC/PRF/5 hepatoma cells

We observed the effects of NaHS on NF-κB p65 phosphorylation and IκBα phosphorylation in PLC/PRF/5 hepatoma cells. PLC/PRF/5 hepatoma cells were exposed to 500 μmol/l NaHS for the indicated times (3, 6, 9, 12 and 24 h), the expression levels of p-NF-κB p65 were significantly upregulated, reaching a peak at 12 h (Fig. 5A and B), and the t-NF-κB p65 expression was unchanged. Interestingly, the expression levels of p-IκBα were also markedly increased from 3 h, reaching the maximum at 24 h. Subsequently, we explored the effect of NaHS on the nuclear translocation of NF-κB p65 subunit. NaHS treatment significantly increased the nuclear translocation (Fig. 5E and F), with ameliorating amounts of NF-κB p65 in the cytosol (Fig. 5G and H). These results suggested that NaHS amplifies the activation and nuclear translocation of NF-κB in PLC/PRF/5 hepatoma cells.

PDTC alleviates NaHS-induced increased cell viability in PLC/PRF/5 hepatoma cells

As shown in Fig. 6, exposure of PLC/PRF/5 hepatoma cells to 500 μmol/l NaHS for 24 h obviously induced cell proliferation, leading to an increase in cell viability. However, the increased cell viability was repressed by co-treatment with different doses PDTC (a specific inhibitor of NF-κB pathway) for 24 h.

As shown in Fig. 6, at the dose of PDTC from 5 to 100 μmol/l did not change in the cell viability. On the contrary, the dose of PDTC from 150 to 250 μmol/l significantly suppressed the cells proliferation, leading to a decrease in cell viability and reaching the minimum at 200 μmol/l. According to the above results, PLC/PRF/5 hepatoma cells were co-treated with 500 μmol/l NaHS and 200 μmol/l PDTC for 24 h in all following experiments.

PDTC increases NaHS-induced decreased cell apoptosis in PLC/PRF/5 hepatoma cells

We observed the effect of PDTC against NaHS-induced decreased cell apoptosis in PLC/PRF/5 hepatoma cells. It was showed that exposure of cells to 500 μmol/l NaHS for 24 h markedly enhanced proliferation, as evidenced by a decrease in apoptotic cells (Fig. 7B). In addition, the above proliferation was partly inhibited by co-treating PLC/PRF/5 hepatoma cells with 500 μmol/l NaHS and 100 μmol/l PDTC for 24 h or almost completely inhibited by co-treating PLC/PRF/5 hepatoma cells with 500 μmol/l NaHS and 200 μmol/l PDTC for 24 h.

PDTC inhibits NaHS-induced increased expression levels of MMP-2 and COX-2 and upregulates NaHS-induced decreased caspase-3 expression in PLC/PRF/5 hepatoma cells

As shown in Fig. 8, PLC/PRF/5 hepatoma cells were exposed to 500 μmol/l NaHS for 24 h, the expression levels of MMP-2 and COX-2 were significantly increased, on the contrary, the expression level of caspase-3 was markedly decreased. Notably, co-treatment of PLC/PRF/5 hepatoma cells with 500 μmol/l NaHS and 200 μmol/l PDTC for 24 h considerably depressed NaHS-induced increased expression levels of MMP-2 and COX-2, however, caspase-3 expression was obviously downregulated. Treatment of cells with 200 μmol/l PDTC for 24 h did not alter the basal expression levels of MMP-2, COX-2 and caspase-3.

PDTC suppresses NaHS-induced upregulated production of VEGF in PLC/PRF/5 hepatoma cells

As shown in Fig. 9, the level of VEGF was markedly increased in NaHS-induced PLC/PRF/5 hepatoma cells, compared with the control group (P<0.01). However, the increased level of VEGF was significantly suppressed by co-treatment cells with PDTC and NaHS.

H2S demonstrates pro-proliferation, anti-apoptosis, angiogenesis, invasion and migration effects on PLC/PRF/5 hepatoma cells via amplifying the activation of NF-κB pathway

We found that NaHS upregulated both CSE and CBS activity resulting in an elevated rate of H2S production level, which in turn modulates protein expressions of caspase-3, MMP-2 and VEGF. The downregulated caspase-3 directly induced anti-apoptosis, led to decreased apoptosis and increased cell viability of PLC/PRF/5 hepatoma cells. MMP-2 contributes to cancer cell invasion and migration. The increased production of VEGF stimulates angiogenesis, promoting the supply of nutrients and blood to the tumor. Conversely, the above properties of H2S were significantly inhibited by the co-condition of 500 μmol/l NaHS and 200 μmol/l PDTC for 24 h.

Discussion

In this study, we demonstrated a novel finding in tumor development of H2S on PLC/PRF/5 hepatoma cells and also provide data to revel its potential mechanisms. Herein we report that: i) the production of H2S was dramatically increased in the PLC/PRF/5 hepatoma cells compared with the human LO2 hepatocyte cells group, along with overexpression levels of CSE and CBS, ii) NaHS upregulated the expression levels of CBS and CSE, as well as the production of H2S, iii) NaHS caused an increase in cell viability, iv) NaHS induced a decrease in cell apoptosis, due to the increased expression level of caspase-3, v) NaHS activated the NF-κB pathway and promoted NF-κB nuclear translocation, vi) NaHS induced increased expression levels of COX-2, MMP-2 and VEGF, vii) co-treatment of PLC/PRF/5 hepatoma cells with NaHS and PDTC (an inhibitor of NF-κB) was able to largely suppress the above NaHS-induced effects.

H2S, as the third gaseous transmitter following NO and CO, modulates an array of cellular and molecular mechanism, physiological and pathophysiological processes. It can be endogenously catalyzed by CBS or CSE or both and contributes to cardioprotection (2631,53), angiogenesis (3234), antioxidant (35), pro- and anti-inflammatory (36,37) and other wide range of physiological functions (3841) in a variety of animal or human non-tumor cells. However, the effects of H2S on the cancer cells are comparatively complicated and extremely controversial. Accumulating evidence has demonstrated that H2S may possess anticancer functions by reason of its anti-inflammatory effect (54), activation of MAPKs pathway (p38 MAPK, ERK1/2 and JNK) (49), and pro-apoptosis performance (49). Significantly, it has been reported that H2S can exert totally opposite properties, in the main mechanisms related to AKT/ERK pathways (47), and angiogenesis (42,49). NaHS, a donor of H2S, is actively being investigated on account of the above effects of H2S. In the present study, our results suggested that both CSE and CBS were expressed in PLC/PRF/5 hepatoma cells and significantly increased compared with the human hepatic cell line LO2, suggesting H2S might be closely linked to liver cancer progression. Previous studies have demonstrated that H2S can exert anti-apoptosis and pro-proliferation on various diversified cells (2631,53,55,56). In this study, we treated PLC/PRF/5 hepatoma cells with NaHS and found interesting results. Our findings demonstrated that NaHS improved PLC/PRF/5 hepatoma cells proliferation at the concentrations ranging from 100 to 1,000 μmol/l and the optimal concentration of NaHS that induced maximal effect of proliferation was 500 μmol/l, leading to increased cell viability, which indicates that H2S might participate in the PLC cancer growth. Treatment of PLC/PRF/5 hepatoma cells with 500 μmol/l NaHS for 24 h markedly diminished cell apoptosis, and decreased the expression level of caspase-3, an apoptotic factor. The above result was consistent with previous reports (1319). These results demonstrate that H2S induces cell proliferation via exerting its dual cytoprotective and anti-apoptosis effects. A large number of experiments have shown that H2S can contribute to VEGF production (42,5761). In agreement we found that H2S notably increased the production of VEGF in PLC/PRF/5 hepatoma cells, compared with the control group. VEGF is one of the most potent and pivotal angiogenic factors and is crucial for the persistent proliferation and metastasis of tumor cells (62). Therefore, we hypothesized that H2S promotes the supply of blood and nutrients to the tumor via angiogenesis effect. Further studies are needed to explore our hypothesis in vivo. Fourthly, we studied the MMP-2 and found for the first time that treatment with 500 μmol/l NaHS significantly upregulated the expression level of MMP-2, and reached a peak at 12 h. The upregulated expression of MMPs, particularly the gelatinase (MMP-2 and MMP-9), is high associated with metastasis potential in several types of carcinomas (6366). It indicated that H2S was involved in PLC/PRF/5 hepatoma cell invasion and migration. In our observational study, treatment with 500 μmol/l NaHS for 24 h obviously promoted protein COX-2 secretion in PLC/PRF/5 hepatoma cells. Importantly, accumulating evidence has demonstrated that cyclooxygenase (COX)-2 is overexpressed in several types of cancers cells (7375) including hepatocellular carcinoma (HCC) (76). COX-2 expression in cells and animal models is closely associated with tumor cell growth and is a crucial molecule in the development of malignant tumors, including promotion of angiogenesis (77), anti-apoptotic effects (78), invasiveness of tumor cells (74), and tumor cell proliferation (79), demonstrating that COX-2 pathway is implicated in NaHS-induced PLC/PRF/5 hepatoma cell proliferation, anti-apoptosis, angiogenesis and migration.

To investigate the complicated mechanism for NaHS-induced pro-proliferative effect, anti-apoptosis, angiogenesis and migration in PLC/PRF/5 hepatoma cells, we studied the NF-κB pathway, which has been demonstrated previously linked to cancer progression by mean of cell invasion, cell differentiation, cell proliferation, and apoptosis (12). It has been reported that NF-κB can be activated by various stimuli both in normal cells (13) and in cancer cells (1319). Herein, we found that NaHS not only activated NF-κB in PLC/PRF/5 hepatoma cells, leading to increased phosphorylation of NF-κB p65 and IκBα, but also increased NF-κB nuclear translocation. Interestingly, PDTC, an inhibitor of NF-κB, blocked NaHS-induced NF-κB activation, along with NaHS-induced pro-proliferative effect, anti-apoptosis, angiogenesis and migration in PLC/PRF/5 hepatoma cells, because of decreased expression levels of MMP-2, VEGF, and COX-2, and increased caspase-3 expression. These results suggest that NF-κB activation is necessary in NaHS-induced PLC/PRF/5 hepatoma cell progression.

NO, which is another important gaseous transmitter, can also exert a wide variety of biological properties. It is hard to prove that treatment of PLC/PRF/5 hepatoma cells with 500 μmol/l NaHS for 24 h significantly reduced NO production in culture medium (data not shown). Thefore, the interaction of H2S and NO in PLC/PRF/5 hepatoma cells is still unclear and need to be further investigated.

A novel finding of our present study is the interaction between H2S and its catalyzing enzyme (CSE and CBS) in PLC/PRF/5 hepatoma cells. On the one hand, naturally, H2S can be catalyzed by CBS or CSE and treatment of PLC/PRF/5 hepatoma cells with NaHS significantly increased its production compared with the control group. Unexpectedly, treatment of PLC/PRF/5 hepatoma cells with NaHS significantly upregulated both CSE and CBS expression. This finding means that there may be positive regulation mechanism between H2S and its catalyzing enzyme (CSE and CBS) in PLC/PRF/5 hepatoma cells. The positive regulation mechanism might play a crucial role in NaHS-induced liver cancer cell progression. However, this mechanism remains to be further elucidated.

In conclusion, H2S induced cells proliferation, anti-apoptosis, angiogenesis, and migration in PLC/PRF/5 hepatoma cells. These effects might be mediated by the activation of NF-κB pathway, leading to overexpression levels of MMP-2, COX-2 and VEGF, down-expression of caspase-3, increased cell viability, and decreased number of apoptotic cells. In PLC, the findings provide novel insight into a unified concept and identify CBS- and CSE-derived H2S as an endogenous tumor-promoting factor and anticancer drug target. The interaction of H2S and NO in PLC/PRF/5 hepatoma cells is still unclear and needs to be further investigated.

References

1 

Nordenstedt H, White DL and El-Serag HB: The changing pattern of epidemiology in hepatocellular carcinoma. Dig Liver Dis. 42(Suppl 3): S206–S214. 2010. View Article : Google Scholar : PubMed/NCBI

2 

Ferlay J, Shin HR, Bray F, Forman D, Mathers C and Parkin DM: Estimates of worldwide burden of cancer in 2008: GLOBOCAN 2008. Int J Cancer. 127:2893–2917. 2010. View Article : Google Scholar

3 

Parkin DM, Bray F, Ferlay J and Pisani P: Global cancer statistics, 2002. CA Cancer J Clin. 55:74–108. 2005. View Article : Google Scholar : PubMed/NCBI

4 

Yu MC and Yuan JM: Environmental factors and risk for hepatocellular carcinoma. Gastroenterology. 127(Suppl 1): S72–S78. 2004. View Article : Google Scholar : PubMed/NCBI

5 

Chuang SC, La Vecchia C and Boffetta P: Liver cancer: Descriptive epidemiology and risk factors other than HBV and HCV infection. Cancer Lett. 286:9–14. 2009. View Article : Google Scholar

6 

Liang X, Bi S, Yang W, et al: Epidemiological serosurvey of hepatitis B in China - declining HBV prevalence due to hepatitis B vaccination. Vaccine. 27:6550–6557. 2009. View Article : Google Scholar : PubMed/NCBI

7 

Liang H, Wang J, Xiao H, Wang D, Wei W, Qiao Y and Boffetta P: Estimation of cancer incidence and mortality attributable to alcohol drinking in China. BMC Public Health. 10:7302010. View Article : Google Scholar : PubMed/NCBI

8 

Arsura M and Cavin LG: Nuclear factor-kappaB and liver carcinogenesis. Cancer Lett. 229:157–169. 2005. View Article : Google Scholar : PubMed/NCBI

9 

Pikarsky E, Porat RM, Stein I, Abramovitch R, Amit S, Kasem S, Gutkovich-Pyest E, Urieli-Shoval S, Galun E and Ben-Neriah Y: NF-kappaB functions as a tumour promoter in inflammation-associated cancer. Nature. 431:461–466. 2004. View Article : Google Scholar : PubMed/NCBI

10 

He G and Karin M: NF-κB and STAT3 - key players in liver inflammation and cancer. Cell Res. 21:159–168. 2011. View Article : Google Scholar

11 

Berasain C, Castillo J, Perugorria MJ, Latasa MU, Prieto J and Avila MA: Inflammation and liver cancer: New molecular links. Ann NY Acad Sci. 1155:206–221. 2009. View Article : Google Scholar

12 

Baldwin AS Jr: Series introduction: The transcription factor NF-kappaB and human disease. J Clin Invest. 107:3–6. 2001. View Article : Google Scholar : PubMed/NCBI

13 

Werner SL, Barken D and Hoffmann A: Stimulus specificity of gene expression programs determined by temporal control of IKK activity. Science. 309:1857–1861. 2005. View Article : Google Scholar : PubMed/NCBI

14 

Curran JE, Weinstein SR and Griffiths LR: Polymorphic variants of NFKB1 and its inhibitory protein NFKBIA, and their involvement in sporadic breast cancer. Cancer Lett. 188:103–107. 2002. View Article : Google Scholar : PubMed/NCBI

15 

Zhang P, Wei Q, Li X, Wang K, Zeng H, Bu H and Li H: A functional insertion/deletion polymorphism in the promoter region of the NFKB1 gene increases susceptibility for prostate cancer. Cancer Genet Cytogenet. 191:73–77. 2009. View Article : Google Scholar : PubMed/NCBI

16 

Lo SS, Chen JH, Wu CW and Lui WY: Functional polymorphism of NFKB1 promoter may correlate to the susceptibility of gastric cancer in aged patients. Surgery. 145:280–285. 2009. View Article : Google Scholar : PubMed/NCBI

17 

Yu Y, Liu H, Jin M, Zhang M, Pan Y, Zhang S, Li Q and Chen K: The joint association of REST and NFKB1 polymorphisms on the risk of colorectal cancer. Ann Hum Genet. 76:269–276. 2012. View Article : Google Scholar : PubMed/NCBI

18 

Lin CW, Hsieh YS, Hsin CH, Su CW, Lin CH, Wei LH, Yang SF and Chien MH: Effects of NFKB1 and NFKBIA gene polymorphisms on susceptibility to environmental factors and the clinicopathologic development of oral cancer. PLoS One. 7:e350782012. View Article : Google Scholar : PubMed/NCBI

19 

Gao J, Xu HL, Gao S, et al: Genetic polymorphism of NFKB1 and NFKBIA genes and liver cancer risk: A nested case-control study in Shanghai, China. BMJ Open. 4:e0044272014. View Article : Google Scholar : PubMed/NCBI

20 

Wang R: Two’s company, three’s a crowd: Can H2S be the third endogenous gaseous transmitter? FASEB J. 16:1792–1798. 2002. View Article : Google Scholar : PubMed/NCBI

21 

Guidotti TL: Hydrogen sulfide: Advances in understanding human toxicity. Int J Toxicol. 29:569–581. 2010. View Article : Google Scholar : PubMed/NCBI

22 

Kilburn KH, Thrasher JD and Gray MR: Low-level hydrogen sulfide and central nervous system dysfunction. Toxicol Ind Health. 26:387–405. 2010. View Article : Google Scholar : PubMed/NCBI

23 

Tan BH, Wong PT and Bian JS: Hydrogen sulfide: A novel signaling molecule in the central nervous system. Neurochem Int. 56:3–10. 2010. View Article : Google Scholar

24 

Rong W, Kimura H and Grundy D: The neurophysiology of hydrogen sulfide. Inflamm Allergy Drug Targets. 10:109–117. 2011. View Article : Google Scholar : PubMed/NCBI

25 

Kamoun P: Endogenous production of hydrogen sulfide in mammals. Amino Acids. 26:243–254. 2004. View Article : Google Scholar : PubMed/NCBI

26 

Guo R, Wu K, Chen J, Mo L, Hua X, Zheng D, Chen P, Chen G, Xu W and Feng J: Exogenous hydrogen sulfide protects against doxorubicin-induced inflammation and cytotoxicity by inhibiting p38MAPK/NFκB pathway in H9c2 cardiac cells. Cell Physiol Biochem. 32:1668–1680. 2013.

27 

Guo RM, Xu WM, Lin JC, Mo LQ, Hua XX, Chen PX, Wu K, Zheng DD and Feng JQ: Activation of the p38 MAPK/NF-κB pathway contributes to doxorubicin-induced inflammation and cytotoxicity in H9c2 cardiac cells. Mol Med Rep. 8:603–608. 2013.PubMed/NCBI

28 

El-Seweidy MM, Sadik NA and Shaker OG: Role of sulfurous mineral water and sodium hydrosulfide as potent inhibitors of fibrosis in the heart of diabetic rats. Arch Biochem Biophys. 506:48–57. 2011. View Article : Google Scholar

29 

Dong XB, Yang CT, Zheng DD, et al: Inhibition of ROS-activated ERK1/2 pathway contributes to the protection of H2S against chemical hypoxia-induced injury in H9c2 cells. Mol Cell Biochem. 362:149–157. 2012. View Article : Google Scholar

30 

Yang Z, Yang C, Xiao L, Liao X, Lan A, Wang X, Guo R, Chen P, Hu C and Feng J: Novel insights into the role of HSP90 in cyto-protection of H2S against chemical hypoxia-induced injury in H9c2 cardiac myocytes. Int J Mol Med. 28:397–403. 2011.PubMed/NCBI

31 

Chen SL, Yang CT, Yang ZL, Guo RX, Meng JL, Cui Y, Lan AP, Chen PX and Feng JQ: Hydrogen sulphide protects H9c2 cells against chemical hypoxia-induced injury. Clin Exp Pharmacol Physiol. 37:316–321. 2010. View Article : Google Scholar

32 

Coletta C, Papapetropoulos A, Erdelyi K, et al: Hydrogen sulfide and nitric oxide are mutually dependent in the regulation of angiogenesis and endothelium-dependent vasorelaxation. Proc Natl Acad Sci USA. 109:9161–9166. 2012. View Article : Google Scholar : PubMed/NCBI

33 

Cai WJ, Wang MJ, Moore PK, Jin HM, Yao T and Zhu YC: The novel proangiogenic effect of hydrogen sulfide is dependent on Akt phosphorylation. Cardiovasc Res. 76:29–40. 2007. View Article : Google Scholar : PubMed/NCBI

34 

Papapetropoulos A, Pyriochou A, Altaany Z, et al: Hydrogen sulfide is an endogenous stimulator of angiogenesis. Proc Natl Acad Sci USA. 106:21972–21977. 2009. View Article : Google Scholar : PubMed/NCBI

35 

Kimura H: Hydrogen sulfide: From brain to gut. Antioxid Redox Signal. 12:1111–1123. 2010. View Article : Google Scholar

36 

Fiorucci S: Hydrogen sulfide: From physiology to pharmacology. Inflamm Allergy Drug Targets. 10:77–84. 2011. View Article : Google Scholar : PubMed/NCBI

37 

Hegde A and Bhatia M: Hydrogen sulfide in inflammation: Friend or foe? Inflamm Allergy Drug Targets. 10:118–122. 2011. View Article : Google Scholar : PubMed/NCBI

38 

Zhang H and Bhatia M: Hydrogen sulfide: A novel mediator of leukocyte activation. Immunopharmacol Immunotoxicol. 30:631–645. 2008. View Article : Google Scholar : PubMed/NCBI

39 

Mok YY and Moore PK: Hydrogen sulphide is pro-inflammatory in haemorrhagic shock. Inflamm Res. 57:512–518. 2008. View Article : Google Scholar : PubMed/NCBI

40 

Gong QH, Wang Q, Pan LL, Liu XH, Huang H and Zhu YZ: Hydrogen sulfide attenuates lipopolysaccharide-induced cognitive impairment: A pro-inflammatory pathway in rats. Pharmacol Biochem Behav. 96:52–58. 2010. View Article : Google Scholar : PubMed/NCBI

41 

Zhang J, Sio SW, Moochhala S and Bhatia M: Role of hydrogen sulfide in severe burn injury-induced inflammation in mice. Mol Med. 16:417–424. 2010.PubMed/NCBI

42 

Szabo C, Coletta C, Chao C, Módis K, Szczesny B, Papapetropoulos A and Hellmich MR: Tumor-derived hydrogen sulfide, produced by cystathionine-β-synthase, stimulates bioenergetics, cell proliferation, and angiogenesis in colon cancer. Proc Natl Acad Sci USA. 110:12474–12479. 2013. View Article : Google Scholar

43 

Pupo E, Pla AF, Avanzato D, Moccia F, Cruz JE, Tanzi F, Merlino A, Mancardi D and Munaron L: Hydrogen sulfide promotes calcium signals and migration in tumor-derived endothelial cells. Free Radic Biol Med. 51:1765–1773. 2011. View Article : Google Scholar : PubMed/NCBI

44 

Du SX, Xiao J, Guan F, Sun LM, Wu WS, Tang H, Du JB, Tang CS and Jin HF: Predictive role of cerebrospinal fluid hydrogen sulfide in central nervous system leukemia. Chin Med J (Engl). 124:3450–3454. 2011.

45 

Levine J, Ellis CJ, Furne JK, Springfield J and Levitt MD: Fecal hydrogen sulfide production in ulcerative colitis. Am J Gastroenterol. 93:83–87. 1998. View Article : Google Scholar : PubMed/NCBI

46 

Rose P, Moore PK, Ming SH, Nam OC, Armstrong JS and Whiteman M: Hydrogen sulfide protects colon cancer cells from chemopreventative agent beta-phenylethyl isothiocyanate induced apoptosis. World J Gastroenterol. 11:3990–3997. 2005.PubMed/NCBI

47 

Cai WJ, Wang MJ, Ju LH, Wang C and Zhu YC: Hydrogen sulfide induces human colon cancer cell proliferation: Role of Akt, ERK and p21. Cell Biol Int. 34:565–572. 2010. View Article : Google Scholar : PubMed/NCBI

48 

Cao Q, Zhang L, Yang G, Xu C and Wang R: Butyrate-stimulated H2S production in colon cancer cells. Antioxid Redox Signal. 12:1101–1109. 2010. View Article : Google Scholar

49 

Ma K, Liu Y, Zhu Q, Liu CH, Duan JL, Tan BK and Zhu YZ: H2S donor, S-propargyl-cysteine, increases CSE in SGC-7901 and cancer-induced mice: Evidence for a novel anti-cancer effect of endogenous H2S? PLoS One. 6:e205252011. View Article : Google Scholar

50 

Murata T, Sato T, Kamoda T, Moriyama H, Kumazawa Y and Hanada N: Differential susceptibility to hydrogen sulfide-induced apoptosis between PHLDA1-overexpressing oral cancer cell lines and oral keratinocytes: Role of PHLDA1 as an apoptosis suppressor. Exp Cell Res. 320:247–257. 2014. View Article : Google Scholar

51 

Chattopadhyay M, Kodela R, Olson KR and Kashfi K: NOSH-aspirin (NBS-1120), a novel nitric oxide- and hydrogen sulfide-releasing hybrid is a potent inhibitor of colon cancer cell growth in vitro and in a xenograft mouse model. Biochem Biophys Res Commun. 419:523–528. 2012. View Article : Google Scholar : PubMed/NCBI

52 

Lee ZW, Zhou J, Chen CS, Zhao Y, Tan CH, Li L, Moore PK and Deng LW: The slow-releasing hydrogen sulfide donor, GYY4137, exhibits novel anti-cancer effects in vitro and in vivo. PLoS One. 6:e210772011. View Article : Google Scholar : PubMed/NCBI

53 

Xu W, Wu W, Chen J, Guo R, Lin J, Liao X and Feng J: Exogenous hydrogen sulfide protects H9c2 cardiac cells against high glucose-induced injury by inhibiting the activities of the p38 MAPK and ERK1/2 pathways. Int J Mol Med. 32:917–925. 2013.PubMed/NCBI

54 

Kashfi K: Anti-cancer activity of new designer hydrogen sulfide-donating hybrids. Antioxid Redox Signal. 20:831–846. 2014. View Article : Google Scholar :

55 

Shi S, Li QS, Li H, Zhang L, Xu M, Cheng JL, Peng CH, Xu CQ and Tian Y: Anti-apoptotic action of hydrogen sulfide is associated with early JNK inhibition. Cell Biol Int. 33:1095–1101. 2009. View Article : Google Scholar : PubMed/NCBI

56 

Rinaldi L, Gobbi G, Pambianco M, Micheloni C, Mirandola P and Vitale M: Hydrogen sulfide prevents apoptosis of human PMN via inhibition of p38 and caspase 3. Lab Invest. 86:391–397. 2006. View Article : Google Scholar : PubMed/NCBI

57 

Holwerda KM, Burke SD, Faas MM, et al: Hydrogen sulfide attenuates sFlt1-induced hypertension and renal damage by upregulating vascular endothelial growth factor. J Am Soc Nephrol. 25:717–725. 2014. View Article : Google Scholar :

58 

Polhemus DJ, Kondo K, Bhushan S, Bir SC, Kevil CG, Murohara T, Lefer DJ and Calvert JW: Hydrogen sulfide attenuates cardiac dysfunction after heart failure via induction of angiogenesis. Circ Heart Fail. 6:1077–1086. 2013. View Article : Google Scholar : PubMed/NCBI

59 

Köhn C, Dubrovska G, Huang Y and Gollasch M: Hydrogen sulfide: Potent regulator of vascular tone and stimulator of angiogenesis. Int J Biomed Sci. 8:81–86. 2012.

60 

Bir SC, Kolluru GK, McCarthy P, Shen X, Pardue S, Pattillo CB and Kevil CG: Hydrogen sulfide stimulates ischemic vascular remodeling through nitric oxide synthase and nitrite reduction activity regulating hypoxia-inducible factor-1α and vascular endothelial growth factor-dependent angiogenesis. J Am Heart Assoc. 1:e0040932012. View Article : Google Scholar

61 

Tao BB, Liu SY, Zhang CC, et al: VEGFR2 functions as an H2S-targeting receptor protein kinase with its novel Cys1045-Cys1024 disulfide bond serving as a specific molecular switch for hydrogen sulfide actions in vascular endothelial cells. Antioxid Redox Signal. 19:448–464. 2013. View Article : Google Scholar :

62 

Leung WK, To KF, Go MY, Chan KK, Chan FK, Ng EK, Chung SC and Sung JJ: Cyclooxygenase-2 upregulates vascular endothelial growth factor expression and angiogenesis in human gastric carcinoma. Int J Oncol. 23:1317–1322. 2003.PubMed/NCBI

63 

Jones JL, Shaw JA, Pringle JH and Walker RA: Primary breast myoepithelial cells exert an invasion-suppressor effect on breast cancer cells via paracrine down-regulation of MMP expression in fibroblasts and tumour cells. J Pathol. 201:562–572. 2003. View Article : Google Scholar : PubMed/NCBI

64 

Li C, Li F, Zhao K, Yao J, Cheng Y, Zhao L, Li Z, Lu N and Guo Q: LFG-500 inhibits the invasion of cancer cells via downregulation of PI3K/AKT/NF-κB signaling pathway. PLoS One. 9:e913322014. View Article : Google Scholar

65 

Puzovic V, Brcic I, Ranogajec I and Jakic-Razumovic J: Prognostic values of ETS-1, MMP-2 and MMP-9 expression and co-expression in breast cancer patients. Neoplasma. 61:439–446. 2014. View Article : Google Scholar : PubMed/NCBI

66 

Ruan M, Zhang Z, Li S, Yan M, Liu S, Yang W, Wang L and Zhang C: Activation of Toll-like receptor-9 promotes cellular migration via up-regulating MMP-2 expression in oral squamous cell carcinoma. PLoS One. 9:e927482014. View Article : Google Scholar : PubMed/NCBI

67 

Li Y, Liu G, Cai D, Pan B, Lin Y, Li X, Li S, Zhu L, Liao X and Wang H: H2S inhibition of chemical hypoxia-induced proliferation of HPASMCs is mediated by the upregulation of COX-2/PGI2. Int J Mol Med. 33:359–366. 2014.

68 

Ang SF, Sio SW, Moochhala SM, MacAry PA and Bhatia M: Hydrogen sulfide upregulates cyclooxygenase-2 and prostaglandin E metabolite in sepsis-evoked acute lung injury via transient receptor potential vanilloid type 1 channel activation. J Immunol. 187:4778–4787. 2011. View Article : Google Scholar : PubMed/NCBI

69 

Terzuoli E, Meini S, Cucchi P, Catalani C, Cialdai C, Maggi CA, Giachetti A, Ziche M and Donnini S: Antagonism of bradykinin B2 receptor prevents inflammatory responses in human endothelial cells by quenching the NF-κB pathway activation. PLoS One. 9:e843582014. View Article : Google Scholar

70 

Tsagaraki I, Phenekos C, Tsilibary E and Tzinia A: Calcitonin-induced NF-κB activation up-regulates fibronectin expression in MG63 osteosarcoma cells. Anticancer Res. 33:4901–4906. 2013.PubMed/NCBI

71 

Hsu CJ, Wu MH, Chen CY, Tsai CH, Hsu HC and Tang CH: AMP-activated protein kinase activation mediates CCL3-induced cell migration and matrix metalloproteinase-2 expression in human chondrosarcoma. Cell Commun Signal. 11:682013. View Article : Google Scholar : PubMed/NCBI

72 

Son SW, Kim HG, Han JM, Lee JS, Choi MK, Lee JS and Son CG: Anti-melanoma activity of Cynanchi atrati Radix is mediated by regulation of NF-kappa B activity and pro-apoptotic proteins. J Ethnopharmacol. 153:250–257. 2014. View Article : Google Scholar : PubMed/NCBI

73 

Jana D, Sarkar DK, Ganguly S, Saha S, Sa G, Manna AK, Banerjee A and Mandal S: Role of cyclooxygenase 2 (COX-2) in prognosis of breast cancer. Indian J Surg Oncol. 5:59–65. 2014. View Article : Google Scholar : PubMed/NCBI

74 

Wu X, Cai M, Ji F and Lou LM: The impact of COX-2 on invasion of osteosarcoma cell and its mechanism of regulation. Cancer Cell Int. 14:272014. View Article : Google Scholar : PubMed/NCBI

75 

Xu K, Wang L and Shu HK: COX-2 overexpression increases malignant potential of human glioma cells through Id1. Oncotarget. 5:1241–1252. 2014.PubMed/NCBI

76 

Dong X, Li R, Xiu P, et al: Meloxicam executes its antitumor effects against hepatocellular carcinoma in COX-2-dependent and -independent pathways. PLoS One. 9:e928642014. View Article : Google Scholar

77 

Tegeder I, Niederberger E, Israr E, Gühring H, Brune K, Euchenhofer C, Grösch S and Geisslinger G: Inhibition of NF-kappaB and AP-1 activation by R- and S-flurbiprofen. FASEB J. 15:2–4. 2001.

78 

Seo KW, Coh YR, Rebhun RB, et al: Antitumor effects of celecoxib in COX-2 expressing and non-expressing canine melanoma cell lines. Res Vet Sci. pii: S0034–5288. 00051–00054. 2014.

79 

Sui W, Zhang Y, Wang Z, Wang Z, Jia Q, Wu L and Zhang W: Antitumor effect of a selective COX-2 inhibitor, celecoxib, may be attributed to angiogenesis inhibition through modulating the PTEN/PI3K/Akt/HIF-1 pathway in an H22 murine hepatocarcinoma model. Oncol Rep. 31:2252–2260. 2014.PubMed/NCBI

Related Articles

Journal Cover

May-2015
Volume 46 Issue 5

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhen Y, Pan W, Hu F, Wu H, Feng J, Zhang Y and Chen J: Exogenous hydrogen sulfide exerts proliferation/anti-apoptosis/angiogenesis/migration effects via amplifying the activation of NF-κB pathway in PLC/PRF/5 hepatoma cells. Int J Oncol 46: 2194-2204, 2015
APA
Zhen, Y., Pan, W., Hu, F., Wu, H., Feng, J., Zhang, Y., & Chen, J. (2015). Exogenous hydrogen sulfide exerts proliferation/anti-apoptosis/angiogenesis/migration effects via amplifying the activation of NF-κB pathway in PLC/PRF/5 hepatoma cells. International Journal of Oncology, 46, 2194-2204. https://doi.org/10.3892/ijo.2015.2914
MLA
Zhen, Y., Pan, W., Hu, F., Wu, H., Feng, J., Zhang, Y., Chen, J."Exogenous hydrogen sulfide exerts proliferation/anti-apoptosis/angiogenesis/migration effects via amplifying the activation of NF-κB pathway in PLC/PRF/5 hepatoma cells". International Journal of Oncology 46.5 (2015): 2194-2204.
Chicago
Zhen, Y., Pan, W., Hu, F., Wu, H., Feng, J., Zhang, Y., Chen, J."Exogenous hydrogen sulfide exerts proliferation/anti-apoptosis/angiogenesis/migration effects via amplifying the activation of NF-κB pathway in PLC/PRF/5 hepatoma cells". International Journal of Oncology 46, no. 5 (2015): 2194-2204. https://doi.org/10.3892/ijo.2015.2914