Reactive Oxygen Species and p53 Mediated Activation of p38 and

Sep 13, 2018 - College of Korean Medicine, Kyung Hee University , Seoul 02447 , ... Department of East West Medical Science, Graduate School of East W...
0 downloads 0 Views 8MB Size
Article pubs.acs.org/JAFC

Cite This: J. Agric. Food Chem. XXXX, XXX, XXX−XXX

Reactive Oxygen Species and p53 Mediated Activation of p38 and Caspases is Critically Involved in Kaempferol Induced Apoptosis in Colorectal Cancer Cells Jhin-Baek Choi,† Ju-Ha Kim,† Hyemin Lee,† Ji-Na Pak,‡ Bum Sang Shim,† and Sung-Hoon Kim*,† †

College of Korean Medicine, Kyung Hee University, Seoul 02447, Korea Department of East West Medical Science, Graduate School of East West Medical Science, Yongin 17104, Korea



J. Agric. Food Chem. Downloaded from pubs.acs.org by UNIV OF SOUTH DAKOTA on 09/13/18. For personal use only.

S Supporting Information *

ABSTRACT: Here the molecular mechanisms of Kaempferol were examined in colorectal cancers (CRCs). Kaempferol significantly exerted antiproliferative and cytotoxic effect in HCT116, HCT15, and SW480 cells. Also, Kaempferol increased sub G1 population, G2/M arrest, and the numbers of TUNEL cells in HCT116 colorectal cancer cells. Also, Kaempferol increased the PARP cleavages and activation of caspase-8, -9, and -3, phospho-p38 MAPK, p53, and p21 in HCT116 and HCT15 cells. Of note, Kaempferol generated reactive oxygen species (ROS) (43.7 ± 0.56 vs 25.8 ± 0.43, P < 0.01) in HCT116 cells and reversely ROS inhibitor NAC obstructed the effects of Kaempferol to cleave PARP and caspase-3 and activate phosphorylation of p38 MAPK in HCT116 colorectal cancer cells. Likewise, pancaspase inhibitor z-vad-fmk, p38 MAPK inhibitor SB203580, and p53 depletion blocked PARP and caspase-3 in Kaempferol treated HCT116 colorectal cancer cells. Therefore, these findings provide novel insight that ROS and p53 signalings mediate p38 phosphorylation and caspase activation in Kaempferol stimulated apoptosis in CRCs. KEYWORDS: Kaempferol, apoptosis, ROS, p53, p38 MAPK



INTRODUCTION Colorectal cancer (CRC) is the third most common cause of cancer-related death occurring in the colon or rectum.1,2 Recently, CRC incidence and mortality rates have been rising rapidly in many countries due to the adoption of Western lifestyles.3,4 CRCs have usually been treated by resection, radiotherapy, and chemotherapy with fluorouracil (5-FU) and oxaliplatin.5 Nevertheless, it has been still a hot issue to overcome chemoresistance, side effects, and recurrence of CRCs. Thus, some natural compounds such as curcumin,6 gallotannin,7 reseveratrol,8 and decursin9 are attractive with little toxicity and potency for combination therapy with classical anticancer agents.10 Mitogen-activated protein kinase (MAPK) pathways consists of JNK, ERK1/2, ERK5, and p38 MAPK pathways.11,12 The p38 MAPK and JNK pathways are often deregulated in cancers and also can be stimulated by various stress and stimuli such as reactive oxygen species (ROS).13 It is well-known that several factors such as increased metabolic activity and dysfunction of mitochondria induce elevation of ROS levels.14 Accumulating evidence reveals that ROS act as a double-edged sword; hydrogen peroxide works for insulin, AP-1, growth factor, cytokine, and NF-kB signaling under physiologic conditions, while ROS induce pathologic processes including neurodegenerative diseases and carcinogenesis by their cytotoxicity.15 Kaempferol with the chemical name 3,5,7-trihydroxy-2-(4hydroxyphenyl)-4H-1-benzopyran-4-one is a flavonoid found in many edible plants (e.g., highbush blueberry, kale, beans, endive, tea, broccoli, cabbage, leek, tomato, grapes, strawberries, Gingko biloba, Moringa oleifera, and Sophora japonica.16,17 © XXXX American Chemical Society

Kaempferol was known to have antioxidant, antiinflammatory, and antitumor activities.18,19 Regarding anticancer effects, Kaempferol stimulates apoptosis via p53 upregulation and the caspase cascade in CRCs.20−22 Also, it was reported that Kaempferol induced apoptosis via elevated generation of ROS16,23 or knockdown of TRX-1 and SOD-116,24 in glioblastoma cells. Likewise, Zhang et al. demonstrated that Kaempferol elevates ROS level via regulation of p53-inducible gene 3 (PIG3) in HepG2 cells.24 Despite previous evidence of Kaempferol activity, its antitumor mechanisms in CRCs are unclear so far. Hence, in this work, the antitumor mechanism of Kaempferol was investigated in p53 wild type (p53WD) HCT116, and p53 mutant (p53mut) type HCT15 cells via ROS production and p38 MAPK upregulation.



MATERIALS AND METHODS

Chemicals. Kaempferol, anti-β-actin, N-acetyl cysteine (NAC), MTT, DAPI, and SB203580 were supplied from Sigma-Aldrich (St Louis, MO, USA). Procaspase-9, cleaved caspase-3, procaspase-8, anticleaved PARP, phospho-p38 MAPK, p38 MAPK, p-JNK, JNK, phospho-p42/44 MAPK, p42/44 MAPK, and p21 were bought from Cell Signaling Technology (Beverly, MA, USA; no. 2947). Anti-p53 was obtained from Santa Cruz (Dallas, TX, USA). Cell Culture. SW480 (ATCC CCL-228), HCT15 (ATCC CCL225), and HCT116 (ATCC CCL-247) were bought from ATCC (Manassas, VA, USA). These cells were maintained in RPMI 1640 medium including 10% FBS, 1% antibiotic-antimycotic solution Received: May 21, 2018 Revised: August 19, 2018 Accepted: August 27, 2018

A

DOI: 10.1021/acs.jafc.8b02656 J. Agric. Food Chem. XXXX, XXX, XXX−XXX

Article

Journal of Agricultural and Food Chemistry

Figure 1. Kaempferol inhibits viability and proliferation in colorectal cancer cells. (A) The chemical name of Kaempferol is C15H10O6, and its molecular weight is 286.24 g/mol (MW). (B) Cytotoxicity of Kaempferol, decursin, and DMSO in HCT116 and HCT15 cells. The cells were treated with various concentrations of Kaempferol and decursin (0, 12.5, 25, 50, and 100 μM) for 24, 48, and 72 h. DMSO was used as a vehicle control. Cell viability was measured by MTT assay. Data represent means ± SD from three independent experiments. (C) Antiproliferative effect of Kaempferol in HCT116 and HCT15 cells by colony formation assay. The cells (5 × 102 cells) were treated with various concentrations (0, 50, and 100 μM) of Kaempferol for 24 h in 12 well plate and culture media were changed every 3 days. Cells were incubated for 2 weeks to grow colonies at 37 °C under the condition of 5% CO2, fixed with methanol, stained with crystal violet solution, and dried overnight. B

DOI: 10.1021/acs.jafc.8b02656 J. Agric. Food Chem. XXXX, XXX, XXX−XXX

Article

Journal of Agricultural and Food Chemistry

Figure 2. Kaempferol increases subG1 population and the number of TUNEL positive cells in HCT116 and HCT15 cells. (A) HCT116 and HCT15 cells were treated with Kaempferol (50 and 100 μM) and decursin (100 μM) for 24, 48, and 72 h, washed with PBS and fixed with 70% ethanol overnight at 4 °C. DMSO was used as a vehicle control. The cells were treated by RNase A for 1 h at 37 °C. These were stained by PI (25 μg/mL). Samples were analyzed by FACS Calibur using CellQuest software. (B) HCT116 cells were treated with Kaempferol and decursin for 24 h and stained with DAPI and TUNEL according to Roche’s protocol. Green fluorescent TUNEL staining was observed in HCT116 cells by using Olympus FLOVIEW FV10i confocal microscope. obtained from WelGENE (Daegu, South Korea). The cells were grown to confluency at 37 °C under CO2 incubator. Growth Inhibition Assay. The growth inhibition assay was conducted for assessing cytotoxicity of Kaempferol (positive control; DMSO and decursin) in HCT116, HCT15, and SW480 cells. The cells were cultured with Kaempferol treatment (0, 25, 50, and 100 μM) for 24 h and exposed to MTT solution (50 μL, 1 mg/mL stock).

Then, optical density (OD) was measured and calculated at 570 nm wavelength using an OD Microplate Reader (Mannedorf, Switzerland). Colony Formation Assay. HCT116, SW480, and HCT15 (5 × 102 cells per well) were cultured in a 12-well culture plate to assess antiproliferation of Kaempferol. CRCs were exposed to Kaempferol (0, 50, and 100 μM) for 24 h and maintained for 2 weeks. Then cells were fixed and stained with Diff-Quik solution kit and dried overnight. C

DOI: 10.1021/acs.jafc.8b02656 J. Agric. Food Chem. XXXX, XXX, XXX−XXX

Article

Journal of Agricultural and Food Chemistry

Figure 4. Kaempferol activates phospho-38 MAPK and p53/21 signalings in HCT116 and HCT15 cells. HCT116 and HCT15 cells were treated with 50 or 100 μM of Kaempferol for 24 h and were subjected to Western blotting for phospho-p38 MAPK, p38 MAPK, phospho-ERK, ERK, phospho-JNK, JNK, p53, and p21. Membranes were probed with a β-actin antibody as a loading control. p21, and β-actin, washed by TBST solution, and attached by the secondary HRP-conjugated antibodies for 2 h. The luminescence was detected with the ECL reagent (GE Healthcare, Buckinghamshire, UK). ROS Measurement. Production of hydrogen peroxide was measured by using dichlorodihydrofluorescein diacetate (H2DCFDA) (Invitrogen). HCT116 cells were equilibrated with 10 μM DCFDA for 3 or 6 h at 37 °C, blocking light and then resuspended with PBS. Then DCFDA level was analyzed by using FACS Calibur. Statistical Analyses. All the statistical data were analyzed with GraphPad Prism software (California, USA), especially ANOVA test (Turkey posthoc test) for control and sample group comparison. For two way comparison, Student’s t-test was carried out. Statistical significant values were considered when p-value ≤ 0.05.

Figure 3. Kaempferol cleaves PARP and activates caspases in HCT116 and HCT15 cells. (A) HCT116 and HCT15 cells were treated with 50 or 100 μM of Kaempferol for 24 h and were subjected to Western blotting for cleaved PARP, cleaved caspase-3, pro caspase-8, and pro caspase-9. Membranes were probed with a β-actin antibody as a loading control. (B) Pan caspase inhibitor (z-vad-fmk) was treated with Kaempferol (50 and 100 μM) and subjected to Western blotting with antibodies of cleaved PARP, cleaved caspase-3, and β-actin as a loading control.



Cell Cycle Analysis. HCT116 and HCT15 cells at density of 2 × 105 cells/well were cultured onto 6-well plate and treated by DMSO, decursin, or Kaempferol for 24, 48, and 72 h. The cells were trypsinized, washed, and fixed by the ethanol solution (75%) at −20 °C. The cells were harvested and treated with RNase solution (1 mg/mL) at 37 °C for 1 h, and 25 μg/mL PI (Sigma-Aldrich) was added to cells at room temperature (RT). Finally, the population of the cell cycle phases was calculated by FACS Calibur flow cytometer (Becton Dickinson, FranklinLakes, USA). TUNEL Assay. HCT116 cells exposed to Kaempferol for 24 h were washed by PBS, fixed with 3.5% paraformaldehyde, and permeabilized in PBS with Triton X-100 (0.1%) for 2 min on ice. A 50 μL TUNEL mixture (Roche, Mannheim, Germany) was distributed and incubated under a CO2 incubator at 37 °C for 1 h on condition of being covered by the film. Thereafter, the cells were carefully stained with DAPI solution, exposed to mounting medium (Vectashield, CA, USA), and then observed by Olympus FLUOVIEW FV10i confocal microscope (Olympus, Tokyo, Japan). Western Blotting. Based on the work of Lee et al.,25 the cell lysates were produced by RIPA buffer with phosphatase and protease inhibitors. It was quantified with DC Protein Assay Kit II and electrophoresed on SDS polyacrylamide gels and moved to nitrocellulose (NC) membranes. The skim milk-blocked membranes were exposed to the antibodies of c-PARP, procaspase-8, procaspase-9, cleaved caspase-3, phospho-p38, p38, phospho-JNK, JNK, phospho-ERK, ERK, p53,

RESULTS Kaempferol Exerts Cytotoxic and Antiproliferative Effects with Morphological Changes in Colorectal Cancer Cells. MTT assay was performed to evaluate the cytotoxicity of Kaempferol and decursin (positive control) (Figure 1A, B, and supplementary Figure 1A) in HCT116, HCT15, and SW480 cell lines for 24, 48, and 72 h. Kaempferol and decursin decreased the viability of these cells in the time and concentration dependent manner with the morphological alterations (Figure 1B and supplementary Figure 1B). Also, it was found that Kaempferol attenuated the numbers of colonies by colony formation assay (Figure 1C and supplementary Figure 1C). Kaempferol Increases subG1 Population and Caspase Dependent Apoptosis in HCT116 and HCT15 Cells. Next, the apoptotic effect of Kaempferol was examined in CRC cells at the cellular level. Kaempferol increased subG1 population and G2/M arrest in HCT116 and HCT15 cells (Figure 2A) and also increased the number of TUNEL positive green fluorescent cells in HCT116 cells (Figure 2B). To confirm the molecular effect of Kaempferol at a protein level, immunoblotting was conducted with cleaved caspase-3, procaspase-9, procaspase-8, D

DOI: 10.1021/acs.jafc.8b02656 J. Agric. Food Chem. XXXX, XXX, XXX−XXX

Article

Journal of Agricultural and Food Chemistry

Figure 5. Pivotal roles of ROS, p53, and p38 in Kaempferol induced apoptosis in HCT116 cells. (A) Effect of Kaempferol on ROS production in HCT116 cells. DCFDA was added to Kaempferol-treated HCT116 cells and ROS production by Kaempferol was analyzed using by FACS Calibur using CellQuest software. Data were obtained from three independent experiments. Data represent means ± SD from three independent experiments. ** p < 0.01 vs control, *** p < 0.001 vs control. (B) Effect of NAC on PARP, caspase-3, and p38 in Kaempferol-treated HCT116 cells. HCT116 were treated with 10 mM NAC and, then, exposed to 50 or 100 μM Kaempferol for 24 h. The samples were subjected to Western blotting for cleaved PARP, cleaved caspase-3, p-p38 MAPK, and p38 MAPK. Membranes were probed with a β-actin antibody as a loading control. (C) Effect of p38 inhibitor SB203580 on PARP, caspase-3, and p38 in Kaempferol-treated HCT116 cells. HCT116 cells were treated with Kaempferol in the presence or absence of 10 μM SB203580 for 24 h, and Western blotting was performed for cleaved PARP, cleaved caspase-3, phospho-p38 MAPK, and p38 MAPK. Membranes were probed with a β-actin antibody as a loading control. (D) Effect of p53 depletion on cleaved caspase-3, phospho-p38, p38, p53, and p21 in Kaempferol-treated HCT116 cells. HCT116 cells transfected with p53 siRNA plasmid were exposed to Kaempferol and subjected to Western blotting for cleaved caspase-3, phospho-p38, p38, p53, and p21. Membranes were probed with a β-actin antibody as a loading control. E

DOI: 10.1021/acs.jafc.8b02656 J. Agric. Food Chem. XXXX, XXX, XXX−XXX

Article

Journal of Agricultural and Food Chemistry

Figure 6. Antitumor mechanisms of Kaempferol in colorectal cancer cells via ROS and p53 mediated activation of phospho-p38 and caspases. Kaempferol-metabolites that bacteria transformed might affect apoptosis signaling.

cleaved PARP, and β-actin antibodies. Kaempferol cleaved the proteins of PARP and caspase-3 and reduced pro-caspase-8/9 expression in HCT116 and HCT15 cells (Figure 3A). Consistently, caspase inhibition using pancaspase inhibitor z-vad-fmk blocked PARP cleavage induced by Kaempferol in HCT116 cells (Figure 3B). Kaempferol Activates Phospho-p38 MAPK and p53/21 Signalings, but Reduces Phospho-ERK and Phospho-JNK in HCT116 and HCT15 Cells. The expression of p53 and p21 and phospho-p38 MAPK were upregulated by Kaempferol treatment in HCT116 and HCT15 cells, while phosphorylation of JNK and ERK was attenuated in HCT116 cells (Figure 4). Kaempferol Increases ROS Production and Activates p38 MAPK Phosphorylation in HCT116 Cells. ROS production was enhanced in Kaempferol-treated HCT116 cells (Figure 5A). Consistently, as shown in Figure 5B, NAC inhibiting ROS blocked phosphorylation of p38 MAPK and cleavages of caspase-3 and PARP by Kaempferol in HCT116 cells. Also, inhibition of p38 MAPK using SB203580 reduced PARP cleavage in HCT116 cells (Figure 5C) and also depletion p53 using p53 siRNA transfection blocked activation of caspase-3, phosphop38, p53, and p21 by Kaempferol in HCT116 cells (Figure 5D).

cytotoxicity by MTT assay and suppressed proliferative effects in HCT116, HCT15, and SW480 colorectal cancer cells, implying the anticancer potential of Kaempferol. The apoptotic pathway is generally known to comprise the intrinsic mitochondrial pathway and the extrinsic death receptor pathway. In the intrinsic pathway, cytochrome C is released into the cytosol from the mitochondria by BAX/BAK multimeric pores.26,27 It forms apoptosome, activating caspase-9 with apoptotic protease activating factor 1 (Apaf-1). Also, in the extrinsic pathway, death ligands binds the death receptors such as CD95 (APO-1/Fas) and TNF-related apoptosis-inducing ligand receptors (TRAIL-R) and leading to activation of procaspase-8 and pro caspase-3 for apoptosis induction.28 Under these kinds of stress, apoptosis is induced with morphological changes of DNA fragmentation and biochemical alterations in the cells.29 Kaempferol induced blebbing debris-like cell particles and anoikis-like features and increased subG1 population, indicating its apoptotic effects in HCT116 and HCT15 cells. Consistently, Kaempferol increased TUNEL + cells in HCT116 cells. Furthermore, immunoblotting revealed that Kaempferol stimulated the caspase cascades (caspase-8, -9, and -3), cleaved PARP in HCT116 and HCT15 cells. Conversely, pancaspase inhibitor z-vad-fmk blocked PARP cleavage induced by Kaempferol in HCT116 cells, demonstrating caspase dependent apoptosis by Kaempferol. Accumulating evidence revealed that some herbal extracts such as Torilis japonica30 and natural compounds including



DISCUSSION The underlying anticancer mechanism of Kaempferol was examined in CRC cells to develop more potent therapeutic agents for CRC patients. Herein, Kaempferol showed significant F

DOI: 10.1021/acs.jafc.8b02656 J. Agric. Food Chem. XXXX, XXX, XXX−XXX

Article

Journal of Agricultural and Food Chemistry carnosic acid31 and miltirone32 induce ROS mediated apoptosis in several cancers. Kaempferol generated ROS in HCT116 cells and eventually induced apoptosis. Conversely, blockade of ROS using N-acetyl cysteine (NAC) attenuated cleavages of caspase-3 and PARP and phosphorylation of p38 MAPK induced by Kaempferol in HCT116 cells, implying ROS mediated apoptosis by Kaempferol. Among three MAPK proteins, p38 MAPK is known to regulate transcription factors and protein kinases such as activating transcription factor 2 (ATF2), C/EBPα, MAPKactivated kinase 2 (MK2), and p53.33 Here, Kaempferol activated phosphorylation of p38 and attenuated phosphorylation of 42/44 MAPK (p-ERK) and JNK and also upregulated tumor suppressor p53 and p21 in HCT116 and HCT15 cells, indicating the important role of p38 MAPK. Conversely, p38 MAPK inhibitor SB203580 reversed cleavages of caspase-3 and PARP and activation of p53 by Kaempferol in HCT116 cells, while depletion of p53 using siRNA transfection blocked phosphorylation of p38 and caspase-3 activation in HCT116 cells, implying crosstalk between p53 and p38, which requires further mechanistic study in the future. Also, it is noteworthy that Kaempferol activated the expression of p53/p21 in p53 mutant HCT15 cells, though the basal level of p53 was weakly expressed in HCT15 cells compared to p53 wild type HCT116 cells, which was supported by Banerjee et al.,34 Okayama et al.,35 and Xavier et al.36 However, it is noteworthy to find that Kaempferol can be transformed into its metabolite kaempferol 4′-O-alpha-Lrhamnopyranoside by Mucor ramannianus (ATCC 9628),37 indicating further PK study and the functions of its metabolite in vivo in the future. Through current study to elucidate the underlying mechanism of Kaempferol, we found that Kaempferol exhibited cytotoxic and antiproliferative effects, increased subG1 population and TUNEL positive cells, activated caspases (-8, -9, and -3) and cleaved PARP, increased ROS production, stimulated phosphorylation of p38 MAPK in CRCs. Conversely, pancaspase inhibitor z-vad-fmk or p38 MAPK inhibitor SB203580 blocked cleavages of caspase-3 and PARP by Kaempferol in HCT116 cells and also ROS inhibitor NAC reversed the cleavages of caspase-3 and PARP and phosphorylation of p38 MAPK by Kaempferol in CRCs (Figure 6). However, the antitumor effect of Kaempferol was more effective in p53 wild type HCT116 cells than in p53 mutant HCT15 cells. Taken together, our findings suggest that Kaempferol induces apoptosis via ROS and p53 dependent activation of p38 and caspases in CRCs as a potent anticancer agent.



Author Contributions

J.-B.C. and J.-H.K. are equally contributing first authors. J.-B.C. and J.-H.K. conceived and designed the experiments; J.-B.C., J.H.K, J.-N.P., and H.L. performed the experiments; J.-B.C., J.H.K., and B.S.S. analyzed the data; J.-H.K. contributed reagents/materials/analysis tools; J.-B.C., J.-H.K., and S.-H.K. wrote the paper. Funding

This study was financially supported by the National Research Foundation of Korea (NRF) (no. 2017R1A2A1A17069297). Notes

The authors declare no competing financial interest.



ABBREVIATIONS CRC, human colorectal carcinoma; FBS, fetal bovine serum; BSA, bovine serum albumin; ROS, reactive oxygen species; JNK, c-Jun N-terminal kinase; ERK1/2, extracellular signalrelated kinases 1 and 2; p38 MAPK, p38 mitogen-activated protein kinase; PARP, poly(ADP-ribose) polymerase; Caspase, cysteine aspartyl-specific protease; HRP, horseradish peroxidase; PBS, phosphate buffered saline; DMSO, dimethyl sulfoxide; RT, room temperature; ECL, enhanced chemoluminescence; SDS, sodium dodecyl sulfate; RNase A, ribonuclease



(1) Torre, L. A.; Bray, F.; Siegel, R. L.; Ferlay, J.; Lortet-Tieulent, J.; Jemal, A. Global cancer statistics, 2012. Ca-Cancer J. Clin. 2015, 65, 87−108. (2) Siegel, R. L.; Miller, K. D.; Fedewa, S. A.; Ahnen, D. J.; Meester, R. G. S.; Barzi, A.; Jemal, A. Colorectal cancer statistics, 2017. CaCancer J. Clin. 2017, 67, 177−193. (3) van Zutphen, M.; Kampman, E.; Giovannucci, E. L.; van Duijnhoven, F. J. B. Lifestyle after Colorectal Cancer Diagnosis in Relation to Survival and Recurrence: A Review of the Literature. Curr. Colorectal Cancer Rep 2017, 13, 370−401. (4) Arnold, M.; Sierra, M. S.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global patterns and trends in colorectal cancer incidence and mortality. Gut 2017, 66, 683−691. (5) Park, Y. S.; Ji, J.; Zalcberg, J. R.; El-Serafi, M.; Buzaid, A.; Ghosn, M. Oxaliplatin/5-fluorouracil-based adjuvant chemotherapy as a standard of care for colon cancer in clinical practice: Outcomes of the ACCElox registry. Asia Pac J. Clin Oncol 2015, 11, 334−42. (6) Mou, S.; Zhou, Z.; He, Y.; Liu, F.; Gong, L. Curcumin inhibits cell proliferation and promotes apoptosis of laryngeal cancer cells through Bcl-2 and PI3K/Akt, and by upregulating miR-15a. Oncol. Lett. 2017, 14, 4937−4942. (7) Kwon, H. Y.; Kim, J. H.; Kim, B.; Srivastava, S. K.; Kim, S. H. Regulation of SIRT1/AMPK axis is critically involved in gallotannininduced senescence and impaired autophagy leading to cell death in hepatocellular carcinoma cells. Arch. Toxicol. 2018, 92, 241. (8) Chai, R.; Fu, H.; Zheng, Z.; Liu, T.; Ji, S.; Li, G. Resveratrol inhibits proliferation and migration through SIRT1 mediated posttranslational modification of PI3K/AKT signaling in hepatocellular carcinoma cells. Mol. Med. Rep. 2017, 16, 8037−8044. (9) Jung, M. H.; Lee, S. H.; Ahn, E. M.; Lee, Y. M. Decursin and decursinol angelate inhibit VEGF-induced angiogenesis via suppression of the VEGFR-2-signaling pathway. Carcinogenesis 2009, 30, 655−61. (10) Yang, N.; Zhao, Y.; Wang, Z.; Liu, Y.; Zhang, Y. Scutellarin suppresses growth and causes apoptosis of human colorectal cancer cells by regulating the p53 pathway. Mol. Med. Rep. 2017, 15, 929− 935. (11) Kim, E. K.; Choi, E. J. Compromised MAPK signaling in human diseases: an update. Arch. Toxicol. 2015, 89, 867−82.

ASSOCIATED CONTENT

* Supporting Information S

The Supporting Information is available free of charge on the ACS Publications website at DOI: 10.1021/acs.jafc.8b02656. Supplementary Figure 1 as described in the text (PDF)



REFERENCES

AUTHOR INFORMATION

Corresponding Author

*Mailing address: Cancer Molecular Targeted Herbal Research Lab, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, South Korea. Tel.: 82-2-961-9233. Fax: 82-2-961-9597. E-mail: [email protected] (S.-H.K.). ORCID

Sung-Hoon Kim: 0000-0003-2423-1973 G

DOI: 10.1021/acs.jafc.8b02656 J. Agric. Food Chem. XXXX, XXX, XXX−XXX

Article

Journal of Agricultural and Food Chemistry (12) Cuenda, A.; Rousseau, S. p38 MAP-kinases pathway regulation, function and role in human diseases. Biochim. Biophys. Acta, Mol. Cell Res. 2007, 1773, 1358−75. (13) Liu, T.; Wu, L.; Wang, D.; Wang, H.; Chen, J.; Yang, C.; Bao, J.; Wu, C. Role of reactive oxygen species-mediated MAPK and NFkappaB activation in polygonatum cyrtonema lectin-induced apoptosis and autophagy in human lung adenocarcinoma A549 cells. J. Biochem. 2016, 160, 315−324. (14) Raza, M. H.; Siraj, S.; Arshad, A.; Waheed, U.; Aldakheel, F.; Alduraywish, S.; Arshad, M. ROS-modulated therapeutic approaches in cancer treatment. J. Cancer Res. Clin. Oncol. 2017, 143, 1789. (15) Jimenez-Del-Rio, M.; Velez-Pardo, C. The bad, the good, and the ugly about oxidative stress. Oxidative medicine and cellular longevity 2012, 2012, 163913. (16) McCreath, S. B.; Delgoda, R. Pharmacognosy: Fundamentals, Applications and Strategies; Academic Press, 2017. (17) Calderon-Montano, J. M.; Burgos-Moron, E.; Perez-Guerrero, C.; Lopez-Lazaro, M. A review on the dietary flavonoid kaempferol. Mini-Rev. Med. Chem. 2011, 11, 298−344. (18) Kang, J. W.; Kim, J. H.; Song, K.; Kim, S. H.; Yoon, J. H.; Kim, K. S. Kaempferol and quercetin, components of Ginkgo biloba extract (EGb 761), induce caspase-3-dependent apoptosis in oral cavity cancer cells. Phytother. Res. 2010, 24 (Suppl 1), S77−82. (19) Sak, K. Cytotoxicity of dietary flavonoids on different human cancer types. Pharmacogn. Rev. 2014, 8, 122−46. (20) Yoshida, T.; Konishi, M.; Horinaka, M.; Yasuda, T.; Goda, A. E.; Taniguchi, H.; Yano, K.; Wakada, M.; Sakai, T. Kaempferol sensitizes colon cancer cells to TRAIL-induced apoptosis. Biochem. Biophys. Res. Commun. 2008, 375, 129−33. (21) Lee, H. S.; Cho, H. J.; Yu, R.; Lee, K. W.; Chun, H. S.; Park, J. H. Mechanisms underlying apoptosis-inducing effects of Kaempferol in HT-29 human colon cancer cells. Int. J. Mol. Sci. 2014, 15, 2722− 37. (22) Li, W.; Du, B.; Wang, T.; Wang, S.; Zhang, J. Kaempferol induces apoptosis in human HCT116 colon cancer cells via the Ataxia-Telangiectasia Mutated-p53 pathway with the involvement of p53 Upregulated Modulator of Apoptosis. Chem.-Biol. Interact. 2009, 177, 121−7. (23) Sharma, V.; Joseph, C.; Ghosh, S.; Agarwal, A.; Mishra, M. K.; Sen, E. Kaempferol induces apoptosis in glioblastoma cells through oxidative stress. Mol. Cancer Ther. 2007, 6, 2544−53. (24) Zhang, Q.; Cheng, G.; Qiu, H.; Zhu, L.; Ren, Z.; Zhao, W.; Zhang, T.; Liu, L. The p53-inducible gene 3 involved in flavonoidinduced cytotoxicity through the reactive oxygen species-mediated mitochondrial apoptotic pathway in human hepatoma cells. Food Funct. 2015, 6, 1518−25. (25) Lee, S. Y.; Lee, J.; Lee, H.; Kim, B.; Lew, J.; Baek, N.; Kim, S. H. MicroRNA134 Mediated Upregulation of JNK and Downregulation of NFkB Signalings Are Critically Involved in Dieckol Induced Antihepatic Fibrosis. J. Agric. Food Chem. 2016, 64, 5508−14. (26) Youle, R. J.; Strasser, A. The BCL-2 protein family: opposing activities that mediate cell death. Nat. Rev. Mol. Cell Biol. 2008, 9, 47− 59. (27) Kim, J. H.; Lee, H.; Shin, E. A.; Kim, D. H.; Choi, J. B.; Kim, S. H. Implications of Bcl-2 and its interplay with other molecules and signaling pathways in prostate cancer progression. Expert Opin. Ther. Targets 2017, 21, 911−920. (28) Weinberg, R. The biology of cancer; Garland science, 2013. (29) Millimouno, F. M.; Dong, J.; Yang, L.; Li, J.; Li, X. Targeting apoptosis pathways in cancer and perspectives with natural compounds from mother nature; Cancer prevention research: Philadelphia, PA, 2014; Vol. 7, pp 1081−107. (30) Kim, G. T.; Lee, S. H.; Kim, Y. M. Torilis japonica extractgenerated intracellular ROS induces apoptosis by reducing the mitochondrial membrane potential via regulation of the AMPK-p38 MAPK signaling pathway in HCT116 colon cancer. Int. J. Oncol. 2016, 49, 1088−98. (31) Kim, D. H.; Park, K. W.; Chae, I. G.; Kundu, J.; Kim, E. H.; Kundu, J. K.; Chun, K. S. Carnosic acid inhibits STAT3 signaling and

induces apoptosis through generation of ROS in human colon cancer HCT116 cells. Mol. Carcinog. 2016, 55, 1096−110. (32) Wang, L.; Hu, T.; Shen, J.; Zhang, L.; Li, L. F.; Chan, R. L.; Li, M. X.; Wu, W. K.; Cho, C. H. Miltirone induced mitochondrial dysfunction and ROS-dependent apoptosis in colon cancer cells. Life Sci. 2016, 151, 224−234. (33) Wagner, E. F.; Nebreda, A. R. Signal integration by JNK and p38 MAPK pathways in cancer development. Nat. Rev. Cancer 2009, 9, 537−49. (34) Banerjee, K.; Mandal, M. Oxidative stress triggered by naturally occurring flavone apigenin results in senescence and chemotherapeutic effect in human colorectal cancer cells. Redox Biol. 2015, 5, 153−62. (35) Okayama, S.; Kopelovich, L.; Balmus, G.; Weiss, R. S.; Herbert, B. S.; Dannenberg, A. J.; Subbaramaiah, K. p53 protein regulates Hsp90 ATPase activity and thereby Wnt signaling by modulating Aha1 expression. J. Biol. Chem. 2014, 289, 6513−25. (36) Xavier, C. P.; Lima, C. F.; Pedro, D. F.; Wilson, J. M.; Kristiansen, K.; Pereira-Wilson, C. Ursolic acid induces cell death and modulates autophagy through JNK pathway in apoptosis-resistant colorectal cancer cells. J. Nutr. Biochem. 2013, 24, 706−12. (37) Ibrahim, A.; Khalifa, S. I.; Khafagi, I.; Youssef, D. T.; Khan, S.; Mesbah, M.; Khan, I. Microbial metabolism of biologically active secondary metabolites from Nerium oleander L. Chem. Pharm. Bull. 2008, 56, 1253−8.

H

DOI: 10.1021/acs.jafc.8b02656 J. Agric. Food Chem. XXXX, XXX, XXX−XXX