Gallic acid inhibits the growth of calf pulmonary arterial endothelial cells through cell death and glutathione depletion

  • Authors:
    • Woo Hyun Park
  • View Affiliations

  • Published online on: September 21, 2017     https://doi.org/10.3892/mmr.2017.7585
  • Pages: 7805-7812
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Gallic acid (GA) exhibits a number of cellular effects, including apoptosis, which is associated with oxidative stress. The present study investigated the effects of GA on calf pulmonary arterial endothelial cell (CPAEC) growth and death, along with the levels of reactive oxygen species (ROS) and glutathione (GSH). GA treatment inhibited the growth of CPAECs at 24 h, and the half‑maximal inhibitory concentration (IC50) value of GA was ~30 µM. GA treatment also induced cell death, which was accompanied by a loss of mitochondrial membrane potential (ΔѰm). GA treatment in CPAECs resulted in decreased ROS levels, including O2•‑, whereas the number of GSH‑depleted cells increased. Neither a pan‑caspase inhibitor (benzyloxycarbonyl‑Val‑Ala‑Asp‑fluoromethylketone) nor buthionine sulfoximine treatment affected GA‑induced cell growth inhibition, cell death, ROS and GSH levels in CPAECs, whereas co‑treatment with N‑acetyl‑cysteine (NAC) resulted in enhanced cell growth inhibition, cell death and ΔѰm loss in these cells. Although NAC treatment did not significantly influence ROS levels in GA‑treated CPAECs, it significantly enhanced GSH depletion in these cells. In conclusion, GA inhibited the growth of CPAECs via cell death, which was associated with GSH depletion rather than alterations to ROS levels.

Introduction

Gallic acid (GA) is found in a variety of fruits and foods, and is well absorbed in human body (1). GA has been reported to have diverse biological and pharmacological behaviors, such as antibacterial and antiviral effects (2,3). However, the most notable role of GA is associated with its anticancer activity, which has been reported in lung cancer (4,5), leukemia (6) and prostate cancer (7), as well as breast, gastric, colon, cervical and esophageal cancers (8,9). GA-induced apoptosis has been associated with oxidative stress derived from reactive oxygen species (ROS) (46,10). The central components of ROS are hydrogen peroxide (H2O2), hydroxyl radical (OH) and superoxide anion (O2•−), which are generated as by-products of mitochondrial respiration or certain oxidases (11). O2•− is metabolized to H2O2 by superoxide dismutases, and H2O2 is further detoxified to O2 and H2O by catalase or glutathione (GSH) peroxidases (11). Oxidative stress occurs due to the overproduction of ROS and/or the decreased decomposition of them. GA-induced cell death has also been correlated with mitochondrial dysfunction and increased intracellular Ca2+ level (4,5,12); however, GA treatment did not lead to cytotoxicity in normal rat fibroblast and endothelial cells (13). In addition, GA exhibited potential antiapoptotic effects in normal human lymphocytes (14) and protected rat insulinoma RINm5F β-cells from glucolipotoxicity through its antiapoptotic mechanism (15). Controversially, GA was suggested to have pro-oxidative in addition to antioxidative properties, depending on the levels of iron or H2O2 (16,17). Therefore, additional studies are required to reassess the cellular properties of GA under diverse conditions.

Vascular endothelial cells (ECs) are involved in the regulation of inflammation, blood pressure and angiogenesis (18). Vascular ECs experience a broad range of oxidative stress, which may ultimately lead to endothelial dysfunction through the induction of apoptosis (19). Angiogenesis is a crucial step in the transition of a dormant tumor into a malignant state; the proliferation of ECs is an early step during sprouting angiogenesis. Despite important roles of vascular ECs in tumor biogenesis and development, the effects of GA on ECs are relatively poorly understood. In the vasculature, ROS serve physiological and pathophysiological roles through the regulation of numerous cellular processes, including cell proliferation, death and survival (19). Therefore, further research is required to explore the cellular effects of GA on ECs in relation to the levels of ROS and GSH expression. Calf pulmonary arterial ECs (CPAECs) are an established cell model for both cellular and molecular endothelial cell research (20,21).

The present study investigated the effects of GA exposure on CPAEC growth and death in relation to ROS and GSH levels, and examined whether the antioxidant N-acetyl cysteine (NAC) and the GSH synthesis inhibitor L-buthionine sulfoximine (BSO) were able to affect GA-induced CPAEC death.

Materials and methods

Cell culture

CPAECs were obtained from Korean Cell Line Bank (Seoul, Korea) and were cultured in RPMI-1640 medium (GE Healthcare Life Sciences, Little Chalfont, UK) supplemented with 10% fetal bovine serum (FBS; Sigma-Aldrich; Merck KGaA, Darmstadt, Germany) and 1% penicillin-streptomycin (Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA). CPAECs were routinely grown in 100 mm plastic tissue culture dishes (Nalge Nunc International, Penfield, NY, USA) in humidified incubator containing 5% CO2 at 37°C and harvested with a solution of trypsin-EDTA (Gibco; Thermo Fisher Scientific, Inc.) while in the logarithmic phase of growth.

Reagents

GA (Sigma-Aldrich; Merck KGaA) was dissolved in 100% ethanol to 200 mM, and used at the indicated concentrations. The pan-caspase inhibitor benzyloxycarbonyl-Val-Ala-Asp-fluoromethylketone (Z-VAD-FMK; R&D Systems, Inc., Minneapolis, MN, USA) was dissolved in DMSO (Sigma-Aldrich; Merck KGaA) to 10 mM. NAC and BSO were obtained from Sigma-Aldrich (Merck KGaA). NAC 200 mM as a stock solution was dissolved in the buffer of 20 mM 4-(2-hydroxyethyl)-1-piperazineethanesulphonic acid [HEPES; (pH 7.0)], and BSO 100 mM as a stock solution was dissolved in distilled water. Based on a previous study (22), cells were pre-incubated at 37°C for 1 h with Z-VAD-FMK (15 µM), NAC (2 mM) or BSO (10 µM), followed by treatment with the GA (25 µM) at 37°C for 24 h before the assays were performed.

Cell proliferation assay

Viable and dead cell numbers and cell proliferation were determined by trypan blue staining and MTT dye absorbance by living cells, respectively, as previously described (23,24). Briefly, 2×105 cells/well in 24-well plates (Nalge Nunc International) were seeded for cell counting and 5×103 cells/well in 96-well microtiter plates (Nalge Nunc International) were seeded for MTT assays. The cells which were cultured in RPMI-1640 medium (GE Healthcare Life Sciences) supplemented with 10% fetal bovine serum (FBS; Sigma-Aldrich; Merck KGaA) and 1% penicillin-streptomycin (Gibco; Thermo Fisher Scientific, Inc.) were exposed to the indicated amounts of GA (between 0 and 50 µM) at 37°C for 24 h and/or 1 h pre-incubation with Z-VAD-FMK (15 µM), NAC (2 mM) or BSO (10 µM) at 37°C. The plates were incubated for 4 h at 37°C. Medium in plates was removed by pipetting, and 200 µl DMSO was added to each well to solubilize the formazan crystals. The optical density was measured at 570 nm using a microplate reader (Synergy™ 2; BioTek Instruments Inc., Winooski, VT, USA).

Sub-G1 analysis

Cells at the sub-G1 phase were determined by staining with propidium iodide [PI; Sigma-Aldrich; Merck KGaA; excitation (Ex)/emission (Em)=488/617 nm], as previously described (25). Briefly, 1×106 cells cultured in RPMI-1640 medium (GE Healthcare Life Sciences) supplemented with 10% FBS (Sigma-Aldrich; Merck KGaA) and 1% penicillin-streptomycin (Gibco; Thermo Fisher Scientific, Inc.) in 60-mm culture dish (Nalge Nunc International) were exposed to the indicated amounts of GA and/or Z-VAD-FMK, NAC or BSO at 37°C for 24 h. Cells were then washed in PBS and fixed in 70% ethanol at 4°C for 1 h. Cells were again washed with PBS and then incubated with PI (10 µg) with simultaneous treatment with RNase at 37°C for 30 min. Cellular DNA content was measured with a FACStar Flow Cytometer (BD Biosciences, Franklin Lakes, NJ, USA) and analyzed using lysis II and Cellfit software (version 2.0; BD Biosciences).

Annexin V-fluorescein isothiocyanate (FITC)/PI staining for cell death detection

Cell death was measured by staining cells with Annexin V-FITC (Ex/Em=488/519 nm; Molecular Probes; Thermo Fisher Scientific, Inc.) and PI (Ex/Em=488/617 nm; Sigma-Aldrich; Merck KGaA), as previously described (26). Briefly, 1×106 cells in 60 mm culture dish (Nalge Nunc International) were incubated with the indicated amounts of GA with or without Z-VAD-FMK, NAC or BSO at 37°C for 24 h. The prepared cells were washed twice with cold PBS and then resuspended in 500 µl of binding buffer [10 mM HEPES/NaOH (pH 7.4), 140 mM NaCl, 2.5 mM CaCl2] at a concentration of 1×106 cells/ml. Then 5 µl of annexin V-FITC (BD Biosciences) and 10 µl of 20 µg/ml PI were added to these cells. Annexin V-FITC/PI staining was analyzed with a FACStar flow cytometer (BD Biosciences) and CellQuest Pro software (version 5.1; BD Biosciences).

Measurement of mitochondrial membrane potential (ΔѰm)

ΔѰm was measured using the Rhodamine 123 mitochondrial-specific fluorescent dye (Sigma-Aldrich; Merck KGaA; Ex/Em=485/535 nm), as previously described (23). Briefly, 1×106 cells in 60 mm culture dish (Nalge Nunc International) were incubated with the indicated amounts of GA with or without Z-VAD-FMK, NAC or BSO at 37°C for 24 h. Cells were washed twice with PBS and incubated with Rhodamine 123 (0.1 µg/ml) at 37°C for 30 min. Rhodamine 123 staining intensity was determined by FACStar flow cytometry (BD Biosciences) and analyzed using CellQuest Pro software (version 5.1; BD Biosciences). An absence of Rhodamine 123 from cells indicated the loss of ΔѰm in CPAECs.

Detection of intracellular ROS levels

Intracellular ROS levels were assessed with the non-fluorescent probe dye 2′,7′-dichlorodihydrofluorescein diacetate (H2DCFDA; Ex/Em=495/529 nm; Molecular Probes; Thermo Fisher Scientific, Inc.); cleavage of the acetate groups converts H2DCFDA into the highly fluorescent 2′,7′-dichloroflurosceine (DCF). Dihydroethidium (DHE; Ex/Em=518/605 nm; Molecular Probes; Thermo Fisher Scientific, Inc.) is a fluorogenic probe that is highly selective for O2•−. Briefly, 1×106 cells in 60 mm culture dish (Nalge Nunc International) were incubated with the indicated amounts of GA with or without Z-VAD-FMK, NAC or BSO at 37°C for 24 h. Following incubation, cells were washed with PBS and incubated with H2DCFDA (20 µM) or DHE (20 µM) at 37°C for 30 min. DCF and DHE fluorescence was detected with a FACStar flow cytometer (BD Biosciences). ROS and O2•− levels were expressed as the mean fluorescence intensity, which was calculated by CellQuest Pro software (version 5.1; BD Biosciences).

Detection of the intracellular glutathione (GSH)

Cellular GSH expression levels were analyzed using 5-chloromethylfluorescein diacetate (CMFDA; Ex/Em=522/595 nm; Molecular Probes; Thermo Fisher Scientific, Inc.), as previously described (27). Briefly, 1×106 cells in 60 mm culture dish (Nalge Nunc International) were incubated with the indicated amounts of GA with or without Z-VAD-FMK, NAC or BSO at 37°C for 24 h. The cells were then washed in PBS. They were then incubated with 5 µM CMFDA at 37°C for 30 min. Fluorescence intensity of the cleaved CMF was determined using a FACStar flow cytometer (BD Biosciences) and analyzed using CellQuest Pro software (version 5.1; BD Biosciences).

Statistical analysis

The results represent the mean of at least three independent experiments ± standard deviation. Student's t-test or one-way analysis of variance with post hoc analysis using Tukey's multiple comparison test for parametric data. P<0.05 was considered to indicate a statistically significant difference.

Results

Effects of GA on cell growth, cell death and ΔѰm in CPAECs

The effects of GA exposure on the growth of CPAECs were examined by counting the number of trypan blue positive- and negative-stained cells. Cells treated with various concentrations of GA for 24 h exhibited a dose-dependent decrease in the population of viable (trypan blue negative) CPAEC cells (Fig. 1A), whereas the number of dead (trypan blue positive) cells increased. The ratio of dead cells to viable cells rose in a dose-dependent manner. Changes in cell proliferation were assessed by MTT assay. CPAEC cells that were treated with GA for 24 h exhibited a reduction in proliferation, and the half-maximal inhibitory concentration (IC50) value of GA in CPAEC cells was ~30 µM at 24 h (Fig. 1B).

In addition, GA treatment enhanced the number of sub-G1 cells, which indicates cell death, in a dose-dependent manner (Fig. 2A). At a dose of 25 µM GA, the number of sub-G1 cells increased ~13% compared with the untreated control CPAECs (Fig. 2A). Whether GA was able to induce apoptosis in CPAECs was examined further. As shown in Fig. 2B, the number of Annexin V-FITC-stained cells increased in CPAECs as the concentration of GA increased. At a dose of 25 µM GA, the number of Annexin V-FITC-stained cells increased by ~16% compared with the untreated control CPAECs (Fig. 2B). Since apoptosis is closely related to a collapse of ΔѰm, the effects of GA on ΔѰm were assessed using rhodamine 123 dye. Treatment with GA triggered the loss of ΔѰm [that is, (−) rhodamine 123 cells] in CPAECs in a dose-dependent manner (Fig. 2C). CPAEC cells treated with 25 µM GA exhibited ~10% decrease in ΔѰm compared with untreated control cells, whereas 50 µM GA treatment strongly increased the loss by ~70% (Fig. 2C).

Effects of GA on intracellular ROS and GSH levels in CPAECs

To assess the levels of intracellular ROS in GA-treated CPAECs at 24 h, H2DCFDA and DHE fluorescent dyes were used. ROS levels, as measured by mean DCF fluorescence intensity, were not significantly altered in CPAECs treated with 5, 10 or 25 µM GA, whereas the ROS level was significantly decreased by 50 µM GA treatment, compared with untreated control cells (Fig. 2D). The level of red fluorescence derived from DHE, which indicates the level of O2•− in the cell, decreased significantly in CPAECs treated with 5, 10 or 25 µM GA (Fig. 2E); however, the level was not significantly altered when cells were treated with 50 µM GA.

Finally, changes in the levels of GSH expression in CPAECs were analyzed using a CMFDA fluorescence dye. GA treatment led to a dose-dependent increase in the number of GSH-depleted cells [(−) CMF; Fig. 2F]. CPAECs treated with 25 µM GA exhibited ~11% increase in the number of GSH-depleted cells compared with untreated control cells.

Effects of Z-VAD-FMK, NAC and BSO on cell growth, cell death and ΔѰm in GA-treated CPAECs

In these experiments, 25 µM GA was used as a suitable dose to differentiate the levels of proliferation and apoptosis in the presence or absence of Z-VAD-FMK (15 µM), NAC (2 mM) or BSO (10 µM) since the IC50 value of GA in CPAEC cells was ~30 µM. Treatment with Z-VAD-FMK, NAC or BSO alone did not significantly affect the growth of CPAECs at 24 h (Fig. 3A). GA-treated CPAEC cells were unaffected by co-treatment with either Z-VAD-FMK or BSO, whereas the GA-induced growth inhibition was strongly enhance by NAC exposure (Fig. 3A).

In relation to cell death and ΔѰm, the number of sub-G1 cells in GA-treated CPAECs that were also treated with Z-VAD-FMK and BSO did not significantly change, whereas co-treatment with NAC resulted in an increase in the number of CPAEC cells at sub-G1 (Fig. 3B). Similarly, neither Z-VAD-FMK nor BSO treatment affected the number of Annexin V-FITC-stained cells in GA-treated CPAECs, whereas GA and NAC co-treatment significantly increased the number in these apoptotic cells (Fig. 3C and D). Treatment with 25 µM GA did not significantly trigger necrotic cell death (that is, cells staining positive for PI and negative for Annexin V-FITC) in CPAECs (Fig. 3C and E). However, NAC co-treatment strongly induced necrotic cell death in CPAECs treated with 25 µM GA, whereas treatment with Z-VAD-FMK or BSO did not (Fig. 3C and E). Similarly, neither Z-VAD-FMK nor BSO co-treatment was able to affect the level of ΔѰm loss [as measured by (−) rhodamine 123 expression] in GA-treated CPAECs, but co-treatment with NAC significantly augmented the loss of ΔѰm in these cells (Fig. 4A and B).

Effects of Z-VAD-FMK, NAC or BSO on ROS, O2•- and GSH levels in GA-treated CPAECs

ROS and GSH levels in GA-treated CPAECs were examined to assess any changes in their expression levels due to treatment with Z-VAD-FMK, NAC or BSO at 24 h. Neither Z-VAD-FMK nor NAC treatment resulted in a change in ROS levels (as determined by DCF fluorescence intensity) in GA-treated CPAECs (Fig. 5A); although co-treatment with BSO appeared to slightly increase the ROS level, this increase was not significant. Cells treated with NAC alone appeared to exhibit a mild reduction in the basal level of ROS, but this was not significant (Fig. 5A). In relation to O2•− levels (as measured by mean DHE fluorescence intensity), none of the co-treatments with Z-VAD-FMK, NAC or BSO significantly affected the GA-induced reduction in O2•− levels in CPAEC cells (Fig. 5B). Cells treated BSO alone exhibited a significant increase in O2•− level compared with untreated control CPAECs (Fig. 5B). Although co-treatment with Z-VAD-FMK or BSO did not significantly affect the levels of GSH depletion (as measured by absence of CMF fluorescence) in GA-treated CPAECs, cells co-treated with NAC significantly increased the number of GSH-depleted cells (Fig. 5C and D). Notably, cells treated with BSO alone exhibited an increased number of GSH-depleted cells compared with untreated control CPAECs (Fig. 5C and D).

Discussion

A number of previous studies have reported that GA exposure inhibited the growth of HeLa cervical cancer cells and Calu-6 and A549 lung cancer cells, and the IC50 was between 30 and 150 µM in these cell lines (4,5,9). In the present study, GA treatment decreased the growth of CPAECs in a dose-dependent manner, with the IC50 value of ~30 µM at 24 h. Conversely, GA treatment was previously demonstrated to have no relative cytotoxicity in normal fibroblasts and ECs (13,28,29). CPAECs appeared less resistant to GA treatment compared with other normal cells, and instead they were similar to cancer cells in their susceptibility to GA exposure. These differences may be due to the differing activities and antioxidant systems in each cell line.

GA treatment has previously been demonstrated to induce apoptosis in cancer cells through mitochondrial dysfunction (4,5,10). In the present study, GA exposure appeared to induce apoptosis in CPAECs, as evidenced by the increase in the number of sub-G1 cells and Annexin V-FITC-positive staining cells, and it triggered the loss of ΔѰm. The dose-dependent loss of ΔѰm corresponded to the dose-dependent rise in Annexin V-FITC staining cells, supporting the hypothesis that GA-induced cell death may be tightly correlated with the collapse of ΔѰm. In particular, the tested dose of 15 µM Z-VAD-FMK used in the present study did not affect cell death or ΔѰm in GA-treated CPAECs. However, 15 µM Z-VAD-FMK was reported to significantly reduce cell death and ΔѰm loss in CPAECs treated with pyrogallol, a derivative of GA (20). Therefore, this result suggested that the activation of caspases is not closely related to GA-induced apoptosis in CPAECs. It has also been reported that Z-VAD-FMK effectively prevented GA-induced apoptosis in lung cancer cells (30). Therefore, the modes of caspase activation during GA-induced apoptosis may be dependent on cell types (e.g., normal cells vs. cancer cells) and the difference requires further study.

GA exhibits both pro- and antioxidative properties (16,17). Results from a number of previous studies suggested that GA-induced apoptosis may be associated with oxidative stress derived from ROS (46,10). However, treatment with the lower doses of GA (5–25 µM) did not increase ROS levels in CPAECs in the present study, whereas a dose of 50 µM GA induced CPAEC death and decreased ROS levels. Conversely, 5 to 25 µM GA treatment significantly decreased O2•− levels in CPAECs, whereas exposure to 50 µM GA had no effect. Therefore, in the present study, GA treatment appeared to exhibit more antioxidative than pro-oxidative properties in CPAECs. Notably, although NAC exposure did not affect ROS levels in GA-treated CPAECs, NAC co-treatment did result in reduced cell proliferation and increased cell death and ΔѰm loss in these cells; NAC treatment also strongly induced necrotic cell death in CPAECs treated with GA. A previous study reported a similar NAC-induced increase in growth inhibition and apoptotic death in GA-treated lung cancer cells (5). In the present study, neither Z-VAD-FMK nor BSO treatment significantly influenced ROS levels or cell death in GA-treated CPAECs. Cells treated with BSO alone exhibited increased O2•− levels without triggering cell death. Overall, the results from the present study suggest that GA-mediated CPAEC death is not related to oxidative stress. The precise roles of ROS in GA-induced CPAEC death require further investigation.

In general, apoptotic effects have been reported to be inversely proportional to GSH content in cells (22,31,32). The intracellular GSH content may have a decisive effect on GA-induced cell death (4,5,33). Similarly, results from the present study demonstrated that GA treatment dose-dependently decreased the number of GSH-positive cells in CPAECs. Z-VAD-FMK co-treatment did not affect the GA-induced depletion of GSH in CPAECs. In addition, NAC treatment, which showed increases in both necrotic and apoptotic cell deaths by GA, significantly augmented GSH depletion in these cells. Although it is recognized that NAC contains a thiol group and is a GSH precursor, this agent did not seem to be a GSH precursor in GA-treated CPAECs. However, NAC significantly attenuated both GSH depletion and cell death in propyl gallate- and MG132-treated CPAECs (21,34). Thus, NAC may or may not be a GSH precursor depending on other agents used in co-treatments. In addition, when treated with BSO alone, there was a significant increase in the number of GSH-depleted cells; however, co-treatment with GA did not augment GSH depletion CPAECs. By contrast, previous studies have reported that BSO co-treatment enhanced GSH depletion and cell death in GA-treated lung cancer cells, fibroblasts and normal ECs (5,28,33), and decreased GSH levels in MG132-treated CPAECs (35). Therefore, these data suggested that GSH content may serve a vital role in GA-induced cell death, and that BSO exposure may influence GSH levels dependent on cell type and co-incubation drugs.

In conclusion, GA exposure induced growth inhibition and death in CPAECs, which were revealed to be cause by GSH depletion rather than changes in ROS levels. The present data may provide useful information to understand the antiproliferative effects of GA in ECs, particularly CPAECs, in relation to the cellular changes in ROS and GSH levels.

Acknowledgements

The present study was supported by a grant from the National Research Foundation of Korea funded by the Korean government (Ministry of Science ICT and Future Planning; grant no. 2016R1A2B4007773).

Glossary

Abbreviations

Abbreviations:

ΔѰm

mitochondrial membrane potential

BSO

l-buthionine sulfoximine

CMFDA

5-chloromethylfluorescein diacetate

CPAEC

calf pulmonary arterial endothelial cell

DHE

dihydroethidium

EC

endothelial cell

FITC

fluorescein isothiocyanate

GA

gallic acid

GSH

glutathione

H2DCFDA

2′,7′-dichlorodihydrofluorescein diacetate

MTT

3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide

NAC

N-acetyl cysteine

PI

propidium iodine

ROS

reactive oxygen species

Z-VAD-FMK

benzyloxycarbonyl-Val-Ala-Asp-fluoromethylketone.

References

1 

Shahrzad S, Aoyagi K, Winter A, Koyama A and Bitsch I: Pharmacokinetics of gallic acid and its relative bioavailability from tea in healthy humans. J Nutr. 131:1207–1210. 2001.PubMed/NCBI

2 

Kang MS, Oh JS, Kang IC, Hong SJ and Choi CH: Inhibitory effect of methyl gallate and gallic acid on oral bacteria. J Microbiol. 46:744–750. 2008. View Article : Google Scholar : PubMed/NCBI

3 

Kratz JM, Andrighetti-Fröhner CR, Leal PC, Leal PC, Nunes RJ, Yunes RA, Trybala E, Bergström T, Barardi CR and Simões CM: Evaluation of anti-HSV-2 activity of gallic acid and pentyl gallate. Biol Pharm Bull. 31:903–907. 2008. View Article : Google Scholar : PubMed/NCBI

4 

You BR, Kim SZ, Kim SH and Park WH: Gallic acid-induced lung cancer cell death is accompanied by ROS increase and glutathione depletion. Mol Cell Biochem. 357:295–303. 2011. View Article : Google Scholar : PubMed/NCBI

5 

You BR and Park WH: Gallic acid-induced lung cancer cell death is related to glutathione depletion as well as reactive oxygen species increase. Toxicol In Vitro. 24:1356–1362. 2010. View Article : Google Scholar : PubMed/NCBI

6 

Inoue M, Sakaguchi N, Isuzugawa K, Tani H and Ogihara Y: Role of reactive oxygen species in gallic acid-induced apoptosis. Biol Pharm Bull. 23:1153–1157. 2000. View Article : Google Scholar : PubMed/NCBI

7 

Agarwal C, Tyagi A and Agarwal R: Gallic acid causes inactivating phosphorylation of cdc25A/cdc25C-cdc2 via ATM-Chk2 activation, leading to cell cycle arrest, and induces apoptosis in human prostate carcinoma DU145 cells. Mol Cancer Ther. 5:3294–3302. 2006. View Article : Google Scholar : PubMed/NCBI

8 

Faried A, Kurnia D, Faried LS, Usman N, Miyazaki T, Kato H and Kuwano H: Anticancer effects of gallic acid isolated from Indonesian herbal medicine, Phaleria macrocarpa (Scheff.) Boerl, on human cancer cell lines. Int J Oncol. 30:605–613. 2007.PubMed/NCBI

9 

You BR, Moon HJ, Han YH and Park WH: Gallic acid inhibits the growth of HeLa cervical cancer cells via apoptosis and/or necrosis. Food Chem Toxicol. 48:1334–1340. 2010. View Article : Google Scholar : PubMed/NCBI

10 

Chen HM, Wu YC, Chia YC, Chang FR, Hsu HK, Hsieh YC, Chen CC and Yuan SS: Gallic acid, a major component of Toona sinensis leaf extracts, contains a ROS-mediated anti-cancer activity in human prostate cancer cells. Cancer Lett. 286:161–171. 2009. View Article : Google Scholar : PubMed/NCBI

11 

Apak R, Özyürek M, Güçlü K and Çapanoğlu E: Antioxidant activity/capacity measurement. 3. Reactive oxygen and nitrogen species (ROS/RNS) scavenging assays, oxidative stress biomarkers, and chromatographic/chemometric assays. J Agric Food Chem. 64:1046–1070. 2016. View Article : Google Scholar : PubMed/NCBI

12 

Hsieh SC, Wu CH, Wu CC, Yen JH, Liu MC, Hsueh CM and Hsu SL: Gallic acid selectively induces the necrosis of activated hepatic stellate cells via a calcium-dependent calpain I activation pathway. Life Sci. 102:55–64. 2014. View Article : Google Scholar : PubMed/NCBI

13 

Inoue M, Suzuki R, Sakaguchi N, Li Z, Takeda T, Ogihara Y, Jiang BY and Chen Y: Selective induction of cell death in cancer cells by gallic acid. Biol Pharm Bull. 18:1526–1530. 1995. View Article : Google Scholar : PubMed/NCBI

14 

Sohi KK, Mittal N, Hundal MK and Khanduja KL: Gallic acid, an antioxidant, exhibits antiapoptotic potential in normal human lymphocytes: A Bcl-2 independent mechanism. J Nutr Sci Vitaminol (Tokyo). 49:221–227. 2003. View Article : Google Scholar : PubMed/NCBI

15 

Sameermahmood Z, Raji L, Saravanan T, Vaidya A, Mohan V and Balasubramanyam M: Gallic acid protects RINm5F beta-cells from glucolipotoxicity by its antiapoptotic and insulin-secretagogue actions. Phytother Res. 24 Suppl 1:S83–S94. 2010. View Article : Google Scholar : PubMed/NCBI

16 

Strlic M, Radovic T, Kolar J and Pihlar B: Anti- and prooxidative properties of gallic acid in fenton-type systems. J Agric Food Chem. 50:6313–6317. 2002. View Article : Google Scholar : PubMed/NCBI

17 

Sakagami H and Satoh K: Prooxidant action of two antioxidants: Ascorbic acid and gallic acid. Anticancer Res. 17:221–224. 1997.PubMed/NCBI

18 

Bassenge E: Endothelial function in different organs. Prog Cardiovasc Dis. 39:209–228. 1996. View Article : Google Scholar : PubMed/NCBI

19 

Irani K: Oxidant signaling in vascular cell growth, death, and survival: A review of the roles of reactive oxygen species in smooth muscle and endothelial cell mitogenic and apoptotic signaling. Circ Res. 87:179–183. 2000. View Article : Google Scholar : PubMed/NCBI

20 

Han YH and Park WH: Pyrogallol-induced calf pulmonary arterial endothelial cell death via caspase-dependent apoptosis and GSH depletion. Food Chem Toxicol. 48:558–563. 2010. View Article : Google Scholar : PubMed/NCBI

21 

Han YH, Moon HJ, You BR and Park WH: Propyl gallate inhibits the growth of calf pulmonary arterial endothelial cells via glutathione depletion. Toxicol In Vitro. 24:1183–1189. 2010. View Article : Google Scholar : PubMed/NCBI

22 

Park WH: Pyrogallol induces the death of human pulmonary fibroblast cells through ROS increase and GSH depletion. Int J Oncol. 49:785–792. 2016. View Article : Google Scholar : PubMed/NCBI

23 

You BR, Kim SH and Park WH: Reactive oxygen species, glutathione, and thioredoxin influence suberoyl bishydroxamic acid-induced apoptosis in A549 lung cancer cells. Tumour Biol. 36:3429–3439. 2015. View Article : Google Scholar : PubMed/NCBI

24 

Park WH: Anti-apoptotic effect of caspase inhibitors on H2O2-treated HeLa cells through early suppression of its oxidative stress. Oncol Rep. 31:2413–2421. 2014. View Article : Google Scholar : PubMed/NCBI

25 

Han YH, Kim SZ, Kim SH and Park WH: Apoptosis in pyrogallol-treated Calu-6 cells is correlated with the changes of intracellular GSH levels rather than ROS levels. Lung Cancer. 59:301–314. 2008. View Article : Google Scholar : PubMed/NCBI

26 

You BR, Shin HR, Han BR and Park WH: PX-12 induces apoptosis in Calu-6 cells in an oxidative stress-dependent manner. Tumour Biol. 36:2087–2095. 2015. View Article : Google Scholar : PubMed/NCBI

27 

Han YH, Kim SH, Kim SZ and Park WH: Caspase inhibitor decreases apoptosis in pyrogallol-treated lung cancer Calu-6 cells via the prevention of GSH depletion. Int J Oncol. 33:1099–1105. 2008.PubMed/NCBI

28 

Park WH and Kim SH: Involvement of reactive oxygen species and glutathione in gallic acid-induced human umbilical vein endothelial cell death. Oncol Rep. 28:695–700. 2012. View Article : Google Scholar : PubMed/NCBI

29 

You BR and Park WH: Gallic acid-induced human pulmonary fibroblast cell death is accompanied by increases in ROS level and GSH depletion. Drug Chem Toxicol. 34:38–44. 2011. View Article : Google Scholar : PubMed/NCBI

30 

Ohno Y, Fukuda K, Takemura G, Toyota M, Watanabe M, Yasuda N, Xinbin Q, Maruyama R, Akao S, Gotou K, et al: Induction of apoptosis by gallic acid in lung cancer cells. Anticancer Drugs. 10:845–851. 1999. View Article : Google Scholar : PubMed/NCBI

31 

Estrela JM, Ortega A and Obrador E: Glutathione in cancer biology and therapy. Crit Rev Clin Lab Sci. 43:143–181. 2006. View Article : Google Scholar : PubMed/NCBI

32 

Han YH, Kim SZ, Kim SH and Park WH: Intracellular GSH level is a factor in As4.1 juxtaglomerular cell death by arsenic trioxide. J Cell Biochem. 104:995–1009. 2008. View Article : Google Scholar : PubMed/NCBI

33 

You BR and Park WH: Enhancement of gallic acid-induced human pulmonary fibroblast cell death by N-acetyl cysteine and L-buthionine sulfoximine. Hum Exp Toxicol. 30:992–999. 2011. View Article : Google Scholar : PubMed/NCBI

34 

You BR and Park WH: MG132, a proteasome inhibitor-induced calf pulmonary arterial endothelial cell growth and death, are changed by MAPK inhibitors. Drug Chem Toxicol. 34:45–52. 2011. View Article : Google Scholar : PubMed/NCBI

35 

Han YH, Kim SZ, Kim SH and Park WH: Reactive oxygen species and glutathione level changes by a proteasome inhibitor, MG132, partially affect calf pulmonary arterial endothelial cell death. Drug Chem Toxicol. 33:403–409. 2010. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2017
Volume 16 Issue 5

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
Park WH: Gallic acid inhibits the growth of calf pulmonary arterial endothelial cells through cell death and glutathione depletion. Mol Med Rep 16: 7805-7812, 2017
APA
Park, W.H. (2017). Gallic acid inhibits the growth of calf pulmonary arterial endothelial cells through cell death and glutathione depletion. Molecular Medicine Reports, 16, 7805-7812. https://doi.org/10.3892/mmr.2017.7585
MLA
Park, W. H."Gallic acid inhibits the growth of calf pulmonary arterial endothelial cells through cell death and glutathione depletion". Molecular Medicine Reports 16.5 (2017): 7805-7812.
Chicago
Park, W. H."Gallic acid inhibits the growth of calf pulmonary arterial endothelial cells through cell death and glutathione depletion". Molecular Medicine Reports 16, no. 5 (2017): 7805-7812. https://doi.org/10.3892/mmr.2017.7585