DOI QR코드

DOI QR Code

Ginsenoside compound K inhibits nuclear factor-kappa B by targeting Annexin A2

  • Wang, Yu-Shi (Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, College of Life Science, Jilin University) ;
  • Zhu, Hongyan (Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, College of Life Science, Jilin University) ;
  • Li, He (Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, College of Life Science, Jilin University) ;
  • Li, Yang (Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, College of Life Science, Jilin University) ;
  • Zhao, Bing (State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University) ;
  • Jin, Ying-Hua (Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, College of Life Science, Jilin University)
  • Received : 2017.10.19
  • Accepted : 2018.04.16
  • Published : 2019.07.15

Abstract

Background: Ginsenoside compound K(C-K), a major metabolite of ginsenoside, exhibits anticancer activity in various cancer cells and animal models. A cell signaling study has shown that C-K inhibited nuclear factor-kappa B ($NF-{\kappa}B$) pathway in human astroglial cells and liver cancer cells. However, the molecular targets of C-K and the initiating events were not elucidated. Methods: Interaction between C-K and Annexin A2 was determined by molecular docking and thermal shift assay. HepG2 cells were treated with C-K, followed by a luciferase reporter assay for $NF-{\kappa}B$, immunofluorescence imaging for the subcellular localization of Annexin A2 and $NF-{\kappa}B$ p50 subunit, coimmunoprecipitation of Annexin A2 and $NF-{\kappa}B$ p50 subunit, and both cell viability assay and plate clone formation assay to determine the cell viability. Results: Both molecular docking and thermal shift assay positively confirmed the interaction between Annexin A2 and C-K. This interaction prevented the interaction between Annexin A2 and $NF-{\kappa}B$ p50 subunit and their nuclear colocalization, which attenuated the activation of $NF-{\kappa}B$ and the expression of its downstream genes, followed by the activation of caspase 9 and 3. In addition, the overexpression of Annexin A2-K320A, a C-K binding-deficient mutant of Annexin A2, rendered cells to resist C-K treatment, indicating that C-K exerts its cytotoxic activity mainly by targeting Annexin A2. Conclusion: This study for the first time revealed a cellular target of C-K and the molecular mechanism for its anticancer activity.

Keywords

1. Introduction

Ginseng is regarded as a most famous herb in eastern Asia as both pharmaceutical and food resource for over a thousand years, and ginsenosides are the main bioactive compounds in ginseng. Ginsenoside compound K (C-K) is a major metabolite when high-molecular weight ginsenosides were degraded in the intestinal tract with a remarkable human availability compared with other ginsenosides [113]. Recent reports have shown that C-K inhibits both proliferation and angiogenesis of cancer cells in vitro [417]. C-K inhibits growth of cancer cells primarily via apoptosis, and both mitochondrial-and death receptoremediated apoptosis pathway were triggered by C-K [4,5,8110]. Moreover, despite the extraor dinary direct damage to cancer cells, C-K appears to sensitize cancer cells to anticancer reagents such as fluorouracil and cisplatin and presents a synergistic effect under cotreatment with them, indicating that C-K probably functions via removing drug resistance in cancer cells [11,12]. A further study shows that C-K inhibits nuclear factor-kappa B (NF-κB) pathway and acts as an inhibitor of nuclear factor kappa-B kinase (IKK) in inflammatory response [13]. However, its exact cellular target and molecular acting mechanism remain elusive.

NF-κB is a highly conserved transcription factor involved in immune and inflammatory responses in mammals. Recent re ports suggest the constitutive activation of NF-κB is necessarily required in multiple types of cancer [14], particularly contributing to the acquisition of chemical resistance by inducing inhibition of apoptosis proteins (IAPs) [15,16]. In liver tissue, NF-κB functions as a key regulator in the development of liver diseases and even cancer by promoting survival and inflammation of injuredhepatocytes, leading to spontaneous liver damage, fibrosis, and hepatocellular carcinoma [17e20]. Thus, targeting NF-κB may provide a new strategy for liver cancer prevention and treatment.

A couple of studies show that C-K inhibits NF-κB activation and cell proliferation in cancer cells [21e23]. To explore the molecular mechanisms involved in NF-κB suppression by C-K, we performed a thermal shift assay with proteins regulating NF-κB. In the present study, we testified that C-K positively acted as an inhibitor to Annexin A2, a cancer-related protein, promoting NF-κB activation by binding to the NF-κB p50 subunit (p50) [24]. C-K prevented Annexin A2 binding to p50 and inhibited NF-κB activation, followed by a downregulation of its downstream genes, IAPs. Moreover, overexpression of Annexin A2-K302A, a single-site mutant version of Annexin A2 failing to interact with C-K, rendered cells to resist C-Keinduced NF-κB deactivation and apoptosis. These results suggested that C-K exerts its anti-NF-κB and subsequent pro-apoptosis effect mainly by inhibiting Annexin A2 in HepG2 cells.

2. Materials and method

2.1. Cell lines, reagents, and plasmid vectors

HepG2 cells were obtained from the American Type Culture Collection (ATCC, Rockville, MA, USA). Dulbecco’s Modified Eagle’s Medium and fetal bovine serum were obtained from Gibco BRL (Grand Island, NE, USA). Chemical reagents including C-K, phorbol myristate acetate (PMA), and etoposide were obtained from Sigma Aldrich (St. Louis, MO, USA). C-K was dissolved to a final concen tration of 12 mM in 75% alcohol. PMA (10 μg/mL) and etoposide (25 mg/mL) were dissolved in dimethyl sulfoxide (DMSO). Primary antibodies, including mouse anti-Annexin A2 (sc-47696), mouse anti-p50 (sc-8414), rabbit anti-p50 (sc-7178), mouse anti-C myc (sc-49), rabbit anti-C myc (sc-789), mouse anti-X-linked inhibitor of apoptosis (X-IAP) (sc-55551), rabbit anti-cellular inhibitor of apoptosis (c-IAP1) (sc-7943), rabbit anti-c-IAP2 (sc-7944), mouse anti-survivin (sc-17779), and mouse anti-β-actin (sc-47778), were obtained from Santa Cruz Biotechnology (Dallas, Texas, USA). Sec ondary antibodies conjugated with horse radish peroxidase (HRP) for immune blotting were obtained from Pierce (Thermo Fisher Scientific, Rockford, IL, USA). Cy3 affinipure donkey anti-mouse IgG and Alexa Fluor 488 affinipure donkey anti-Rabbit IgG were ob tained from Jackson (Jackson ImmunoResearch Inc., PA, USA). Vectors were constructed as previously described [25] for Annexin A2 knockdown, luciferase reporter assay, prokaryotic expression of Annexin A2 and p50, and eukaryotic expression of Annexin A2 and K302A mutant of Annexin A2.

2.2. Molecular docking

Molecular docking was performed with C-K and Annexin A2 with structure resource of PubChem CID: 9852086 from the NCBI PubChem Compound database (http://www.ncbi.nlm.nih.gov/ pccompound) and PDB ID: 2HYU from the RCSB Protein Data Bank (http://www.rcsb.org/pdb). Molecular docking was per formed with AutoDock tools (version 4.2.6) and visualized with the Discovery Studio 4.0 Visualizer (BIOVIA, CA, USA).

2.3. Thermal shift assay

Cellular thermal shift assay, CTSA (in vivo), was performed with HepG2 cells and 12 μM of C-K with procedure described previously [25].

In vitro thermal shift assay: Purified Annexin A2 protein with final concentration of 0.2 mM (~7 μg/mL) was mixed with C-K with a dose gradient from 0.2 μM to 10 μM with a final volume of 200 μL then heated at 55°C for 3 min, with the control tube on ice for 3 min. Then, 10 μL from each tube was used for sodium dodecyl sulfate-polyacrylamide gel electropheresis (SDS-PAGE) and immunoblot.

2.4. Immunoprecipitation

In vivo immunoprecipitation was carried out with HepG2 cells pretreated with C-K (6 μM), PMA (100 ng/mL), and etoposide (25 μg/mL) for 12 h. Cell lysate was precipitated by anti-Annexin A2, anti-p50, or anti-C-myc antibody and used for immunoblot.

In vitro immunoprecipitation was carried out with prokaryotic expressed Annexin A2 and p50. Protein lysate was precipitated by anti-Annexin A2 or anti-p50 antibody and used for immunoblot.

2.5. Immunofluorescence

HepG2 cells were seeded onto glass cover slips and treated with C-K, PMA, and etoposide for 2 h. Then, cells were fixed,permeabilized, immunolabeled, and stained as described [25]. Images were gained by fluorescence microscopy.

2.6. Quantitative real-time polymerase chain reaction

Total RNA was isolated using TRIzol (Invitrogen, Thermo Fisher Scientific, Rockford, IL, USA) and reverse transcripted with the EasyScript Reverse Transcriptase kit (Transgen, Beijing, China). Quantitative real-time polymerase chain reaction (qRT-PCR) was generated on Applied Biosystem 7500 Real-time PCR system (Applied Biosystems, Thermo Fisher Scientific, Rockford, IL, USA). The amplification was performed with a three-step program, one cycle at 95°C for 30 s, followed by 45 cycles at 95°C for 5 s, 50°C for 15 s, and 72°C for 10 s, with signal-collecting steps after annealing and extending for 34 s.

2.7. Statistical analysis

Data are repeated thrice and presented with the mean± standard deviation. Statistical significance was determined by the Student t test.

3. Results

3.1. C-K interacted with Annexin A2

Molecular docking was performed with C-K- and NF-κB- associated proteins, among which Annexin A2 interacted with C-K, and Asp182, Gln226, and Lys302 appeared to be responsible for their interaction (Fig. 1A). A following CTSA in HepG2 cells showed evident increase of the thermal stability of Annexin A2 by C-K, indicating that C-K penetrated cells and bound to Annexin A2 (Fig. 1B). A further CTSA with overexpressed wild-type version of Annexin A2 (Annexin A2-WT) also showed a similar thermal sta bility shift under treatment of C-K, whereas that of the K302A mutant of Annexin A2 (Annexin A2-K302A) was only slightly affected by C-K, indicating Lys302 was responsible for the inter action between C-K and Annexin A2 (Fig. 1B). To eliminate the in fluence of other cellular component, a thermal shift In vitro was then performed. C-K enhanced the thermal stability of Annexin A2-WT, but not Annexin A2-K302A, in a dose-dependent manner, indicating that C-K upregulated Annexin A2 thermal stability by direct interaction and Lys302 was a dominant amino acid for their interaction (Fig. 1C).

Fig. 1. C-K bound to Annexin A2. (A) Results of molecular docking shows the interaction between Annexin A2 and C-K. (B) Cellular thermal shift assay presented the thermal stability of Annexin A2 under treatment with C-K (12 μM). (C) Thermal shift in vitro showed the thermal stability of purified Annexin A2 (0.2 μM) under treatment with C-K.

3.2. C-K inhibited the interaction and nuclear colocalization of Annexin A2 and p50

To assess whether C-K can exert its activity by targeting Annexin A2, the effect of C-K on the association between Annexin A2 and p50 was examined. Immunoprecipitation was performed with whole-cell lysate of HepG2 cells under treatment of 6 μM of C-K, with or without NF-κB activator, 100 ng/mL of PMA, or 25 μg/mL of etoposide. C-K significantly reduced the interaction between Annexin A2 and p50 both in resting cells and NF-κB activatore treated cells (Fig. 2A). Immunoprecipitation with purified proteins also showed significant inhibition of C-K on this interaction, indi cating that C-K inhibits the assembly of Annexin A2-p50 complex by directly targeting Annexin A2 (Fig. 2B). Then, immunofluores cence was engaged to examine the subcellular distribution of Annexin A2 and p50 under C-K treatment. The colocalization of Annexin A2 and p50 in the nucleus was significantly reduced under C-K treatment in NF-κB activator-stimulating and resting states (Fig. 2C). Taken together, these data suggested that C-K prevented Annexin A2 binding to p50 and inhibited their nuclear colocaliza tion by targeting Annexin A2.

Fig. 2. C-K inhibited interaction between Annexin A2 and NF-κB p50 subunit and their nuclear colocalization. (A) Immunoprecipitation was performed with whole-cell lysate under treatment with C-K (6 μM), etoposide (25 μg/mL), and PMA (100 ng/mL), and the interaction was analyzed by an immunoblot. (B) Immunoprecipitation was performed with prokaryotic cells-expressed Annexin A2 and NF-κB p50 subunit under treatment with C-K (6 μM), and the interaction was analyzed by an immunoblot. (C) The subcellular distribution of Annexin A2 and NF-κB p50 subunit was examined by immunofluorescence under treatment with C-K (6 mM), etoposide (25 mg/mL), and PMA (100 ng/mL), and DAPI showed the nuclear region. C-K, compound K; DAPI, 4',6-diamidino-2-phenylindole; NF-κB, nuclear factor-kappa B; PMA, phorbol myristate acetate.

3.3. C-K suppressed NF-κB activation and downregulated the expression of downstream antiapoptosis gene

HepG2 cells were cultured with C-K (6 μM) and etoposide (25 μg/mL) or PMA (100 ng/mL) followed by a dual luciferase re porter assay to determine the activity of NF-κB. The results showed that NF-κB activation was narrowed by C-K both in resting state and under treatment with etoposide or PMA, which almost tripled NF-κB activation individually (Fig. 3A). Interestingly, the well-known NF-κB-regulated genes, interleukin-6 (Fig. 2B), and antiapoptosis genes from IAPs superfamily including X-IAP, c-IAP1, c-IAP2, and survivin were transcriptionally downregulated under C-K treat ment (Figs. 3C-3F). As expected, the protein levels of these genes were also downregulated by C-K in both resting and NF-κB acti vator-stimulated cells (Fig. 3G), indicating that the pro-apoptosis effect of C-K might be mediated by inhibiting the expression of. IAPs

Fig. 3. C-K inhibited NF-κB activation and downregulated downstream antiapoptosis genes. (A) NF-κB activity was examined by luciferase reporter assay under treatment with C-K (6 μM), etoposide (25 μg/mL), and PMA (100 ng/mL). (B-F) Relative gene expression levels of (B) IL-6, (C) X-IAP, (D) c-IAP1, (E) c-IAP2, and (F) survivin were examined by qRTPCR under treatment with C-K (6 μM), etoposide (25 μg/mL), and PMA (100 ng/mL). E represented etoposide and P represented PMA, with E&C-K and P&C-K representing etoposide and PMA under cotreatment with C-K, respectively. (G) Protein levels of X-IAP, c-IAP1, c-IAP2, and survivin were examined by immunoblot under treatment with C-K (6 μM), etoposide (25 μg/mL), and PMA (100 ng/mL), and β-actin was shown as a loading control. All data are shown as the mean ± SD, and the experimental points show the average of at least triplicates. All experiments were repeated at least three times. c-IAP, cellular inhibitor of apoptosis; C-K, compound K; IAP, inhibition of apoptosis protein; IL, interleukin; NF-κB, nuclear factor-kappa B; PMA, phorbol myristate acetate; qRT-PCR, quantitative real-time polymerase chain reaction; SD, standard deviation; X-IAP, X-linked inhibitor of apoptosis.

3.4. Annexin A2 knockdown enhanced anticancer activity of C-K

To examine the role of Annexin A2 in NF-κB activation, NF-κB activity was determined in HepG2 cells with Annexin A2 knock down via shRNA. Both Annexin A2 knockdown and C-K treatment effectively reduced NF-κB activity respectively and presented a synergetic effect when combined treatment (Fig. 4A). In addition, caspase 3 and 9 were activated under both C-K or Annexin A2 knockdown treatment, and the activity was also enhanced under cotreatment with C-K and Annexin A2 knockdown (Figs. 4B, 4C). MTT assay also showed a significant reduction of cell viability under C-K treatment, which was further enhanced by Annexin A2 knockdown (Fig. 4D). Then, a plate clone formation assay also showed that the clone number was markedly decreased by either C-K or Annexin A2 knockdown. As expected, the clone-forming activity was much more significantly impaired by cotreatment with these two steps (Fig. 4E).

Fig. 4. Knockdown of Annexin A2 enhanced the cytotoxicity of C-K. (A) NF-κB activity was examined by luciferase reporter assay under treatment of C-K with (sh-Annexin A2) or without (sh-NC) knockdown of Annexin A2. Activity of (B) caspase 9 and (C) caspase 3 was examined under treatment of C-K with (sh-Annexin A2) or without (sh-NC) knockdown of Annexin A2. (D) Cell viability was examined by MTT for 48 h under treatment of C-K with (sh-Annexin A2) or without (sh-NC) knockdown of Annexin A2. (E) Plate clone formation assay was examined under treatment of C-K (6 μM) with (sh-Annexin A2) or without (sh-NC) knockdown of Annexin A2. (F) Protein level of Annexin A2 was examined by immunoblot with (sh-Annexin A2) or without (sh-NC) knockdown of Annexin A2, and β-actin was shown as a loading control. All data are shown as the mean ± SD. *p < 0.05, **p < 0.01, and ***p < 0.001, and the experimental points show the average of at least triplicates. All experiments were repeated at least three times. Statistical analyses were performed using Student t test. C-K, compound K; NF-κB, nuclear factor-kappa B; SD, standard deviation.

3.5. K302A mutant of Annexin A2 protected cells from anticancer effect induced by C-K

As C-K failed to bind to Annexin A2-K302A (Figs. 1B, 1C), func tional studies were carried out with Annexin A2-K302A-or Annexin A2-WT-overexpressed HepG2 cells. Both Annexin A2-K302A and Annexin A2-WT interacted with p50 in vivo, and only the interaction between p50 and Annexin A2-WT (both endoge nous and ectopically expressed) was decreased under C-K treat ment (Fig. 5A). Interestingly, C-K failed to dissociate Annexin A2-K302A and p50 (Fig. 5A), and further immunoprecipitation In vitro demonstrated similar results with prokaryotic cells-expressed Annexin A2-K302A and p50 (Fig. 5B). Then, the subcel lular distribution was detected by immunofluorescence. The nu clear colocalization of Annexin A2-WT and p50 disappeared under C-K treatment, whereas p50 colocalized with Annexin A2-K302A in the nucleus under C-K treatment (Fig. 5C). The NF-κB activity was enhanced when Annexin A2-WT was overexpressed and down regulated by C-K in a dose-dependent manner. Interestingly, C-K could not inhibit the NF-κB activation in Annexin A2-K302A-overexpressed cells (Fig. 6A). A cell viability assay showed that overexpression of both Annexin A2-WT and Annexin A2-K302A desensitized HepG2 cells to cytotoxicity of C-K and Annexin A2-K302A-expressing cells presented a better resistance to C-K (Fig. 6B). Activation of caspase 9 and 3 was less evident in Annexin A2-WT-overexpressing cells than in mock transfectant cells, and only slight activation was detected in Annexin-K302A-expressing cells (Figs. 6C, 6D). A following plate clone formation showed that C-K presented weaker cytotoxicity in Annexin A2-WT-over expressing cells and little effect was detected in Annexin A2-K302A-eoverexpressing cells (Fig. 6E). Taken together, over expression of Annexin A2-WT and Annexin A2-K302A rendered cells resistance to C-K treatment, and Annexin A2-K302A showed more potent resistance to the inhibition of NF-κB and subsequent caspase activation.

Fig. 5. Annexin A2-K302A blocked the inhibitory effect of C-K on the interaction between Annexin A2 and NF-κB p50 subunit and their nuclear colocalization. (A) HepG2 cells were transfected with Annexin A2-WT-myc and Annexin A2-K302A-myc, and immunoprecipitation was performed with whole-cell lysate under treatment with C-K (6 μM) and examined by an immunoblot. (B) Immunoprecipitation was performed with prokaryotic cells-expressed Annexin A2-K302A and NF-κB p50 subunit under treatment with C-K (6 μM), and the interaction was analyzed by an immunoblot. (C) HepG2 cells were transfected with Annexin A2-WT-myc and Annexin A2-K302A-myc, and the subcellular distribution of Annexin A2 and NF-κB p50 subunit was examined by immunofluorescence under treatment with C-K (6 μM), and DAPI showed the nuclear region. C-K, compound K; IP, immunoprecipitation; NF-κB, nuclear factor-kappa B; OE, overexpression.

Fig. 6. Annexin A2-K302A protected cells from the cytotoxicity of C-K. (A) NF-κB activity was examined by luciferase reporter assay under treatment of C-K with Annexin A2-WTmyc and Annexin A2-K302A-myc overexpressed in HepG2 cells. (B) Cell viability was examined by MTT for 48 h under treatment of C-K with Annexin A2-WT-myc and Annexin A2- K302A-myc overexpressed in HepG2 cells. Activity of (C) caspase 9 and (D) caspase 3 was examined under treatment of C-K with Annexin A2-WT-myc and Annexin A2-K302A-myc overexpressed in HepG2 cells. (E) Plate clone formation assay was examined under treatment C-K (6 μM) with Annexin A2-WT-myc and Annexin A2-K302A-myc overexpressed in HepG2 cells. All data are shown as the mean ± SD, and the experimental points show the average of at least triplicates. All experiments were repeated at least three times. C-K, compound K; NF-κB, nuclear factor-kappa B; SD, standard deviation.

4. Discussion

Antiproliferation activity is one of the major effects of C-K [4-13]. A number of study showed that C-K inhibits NF-κB activation [13,21-23]; however, the underlying mechanism was not fully revealed. Previous reports have shown that Annexin family proteins including Annexin A1 [26], A2 [24], A4 [27], and A6 [28] interact with NF-κB members and regulate NF-κB pathway. Mo lecular docking analysis and CTSA displayed that Annexin A2, but not the other members, interacted with C-K (Figs. 1A-1C). To confirm the interaction, single-site mutants of D182A, Q226A, and K302A were constructed, with only K032A mutant successfully expressed. As Lys302 was shown responsible for the conforma tional stability of the N-terminus of Annexin A2 and interacted with p50 [29], a single-point mutant was generated for further research, and Annexin A2-K302A was demonstrated as the binding-deficient mutant of Annexin A2 (Figs. 1B,1C).

Annexin A2 interacts with p50 and facilitates their nuclear colocalization, which promotes NF-κB transcriptional activity [24]. The treatment of C-K broke the interaction between Annexin A2 and p50 (Figs. 2A,2B) and their colocalization in the nucleus (Fig. 2C). As a consequence, NF-κB failed to get activated; accord ingly, the expression of downstream antiapoptosis proteins was reduced under C-K treatment (Fig. 3), resulting in the inhibition of cell viability (Fig. 4B-4E).

Knockdown of Annexin A2 induced the loss of NF-κB activation and cell viability, indicating the essential role of Annexin A2 on NF-κB activation and cell survival in HepG2 cells. Gene knockdown of Annexin A2 showed a synergistic effect with C-K in NF-κB activa tion (Fig. 4A).On the other hand, the overexpression of Annexin A2-WT reduced the cytotoxicity of C-K but did not change the pattern as C-K inhibited NF-κB (Figs. 5A-5C, 6A). However, the over expression of Annexin A2-K302A totally relieved NF-κB suppres sion by C-K and transformed C-K to an activator of NF-κB pathway (Figs. 5Ae5C, 6A).

The following series of analysis on the proliferation capability under modulation of Annexin A2 well coincided with those of NF-κB activity. Knockdown of Annexin A2 presented a synergetic effect coupled with C-K cytotoxicity (Fig. 4), and the overexpression of Annexin A2 markedly attenuated the cytotoxicity of C-K (Fig. 6). The overexpression of Annexin A2-K302A further protected HepG2 cells from C-K-induced cell death (Fig. 6). Thus, it was possible that C-K-induced downregulation of NF-κB activity was mainly medi ated by Annexin A2. We have previously indicated that (20S)G-Rh2, G-Rg5, and G-Rk1 inhibited NF-κB activation targeting Annexin A2; however, a similar mechanism was not obtained to other ginse nosides such as (20S/R)G-Rg3, (20R)G-Rh2, protopanoxadiol (PPD), and protopanaxatriol (PPT) (data not shown) [25,30]. These re searches suggested that C-K acted as a specific binding molecule to Annexin A2.

Upregulation of NF-κB is a frequent and origin event during hepatocarcinogenesis, which may trigger the acquisition of trans formed phenotype [31], and may also contribute to the spreading of liver tumor cells via proliferative and antiapoptosis effect [32,33]. Targeting NF-κB may provide a new strategy for liver cancer pre vention and treatment. The antieliver cancer function of C-K will be examined in future studies.

In summary, we demonstrated that C-K bound to Annexin A2 and inhibited the interaction between Annexin A2 and p50, which resulted in the impairment of NF-κB activity and downstream antiapoptosis genes expression. On the contrary, Annexin A2-K302A, a binding-deficient mutant of Annexin A2, relieved the inactivation of NF-κB driven by C-K and protected cells from cyto toxicity of C-K. In this study, we described the cellular target of C-K for the first time and explained how C-K inhibited NF-κB activation and cancer cell proliferation in human hepatoma cell lines.

Conflicts of interest

The authors declare that there are no conflicts of interest.

Acknowledgments

This study was funded by the Provincial Project for Industrial Innovation Special Fund of Jilin Province (2017) and Special Project for Province &amp; University Construction Plan of Jilin Province (SXGJXX2017-13).

Yu-Shi Wang contributed to thermal shift assay in Fig. 1, immunoprecipitation in Figs. 2 and 5, NF-κB activity analysis in Figs. 3, 4, and 6, and caspase activity analysis in Figs. 4 and 6, as well as the writing of the article.

Hongyan Zhu contributed to the immunofluorescence in Figs. 2 and 5.

He Li contributed to the construction of the vectors and purifi cation of Annexin A2.

Yang Li contributed to the cell viability assay in Figs. 4 and 6.

Bing Zhao contributed to molecular docking analysis in Fig. 1.

Ying-Hua Jin was responsible for the arrangement of the project and contributed to the writing of the article.

References

  1. Liu CY, Zhou RX, Sun CK, Jin YH, Yu HS, Zhang TY, Xu LQ, Jin FX. Preparation of minor ginsenosides C-Mc, C-Y, F2, and C-K from American ginseng PPDginsenoside using special ginsenosidase type-I from Aspergillus niger g.848. J Ginseng Res 2015;39(3):221-9. https://doi.org/10.1016/j.jgr.2014.12.003
  2. Yayeh T, Yun K, Jang S, Oh S. Morphine dependence is attenuated by red ginseng extract and ginsenosides Rh2, Rg3, and compound K. J Ginseng Res 2016;40(4):445-52. https://doi.org/10.1016/j.jgr.2016.08.006
  3. Upadhyaya J, Kim MJ, Kim YH, Kim YH, Ko SR, Park HW, Kim MK. Enzymatic formation of compound-K from ginsenoside Rb1 by enzyme preparation from cultured mycelia of Armillaria mellea. J Ginseng Res 2016;40(2):105-12. https://doi.org/10.1016/j.jgr.2015.05.007
  4. Lei C, Yue M, Qi S, Zhongyu Z, Xiaoqing G, Xiaotong S, Guihua T, Hairong C, Yifa Z. Ginsenoside compound K sensitizes human colon cancer cells to TRAILinduced apoptosis via autophagy-dependent and -independent DR5 upregulation. Cell Death Dis 2016;7(8):e2334. https://doi.org/10.1038/cddis.2016.234
  5. Sunghee C, Kyungsook C, Junghye C, Donghyun K, Kyungtae L, Compound K. A metabolite of ginseng saponin, induces apoptosis via caspase-8-dependent pathway in HL-60 human leukemia cells. BMC Cancer 2009;9(1):449. https://doi.org/10.1186/1471-2407-9-449
  6. Shin KO, Seo CH, Cho HH, Oh S, Hong SP, Yoo HS, Hong JT, Oh KW, Lee YM. Ginsenoside compound K inhibits angiogenesis via regulation of sphingosine kinase-1 in human umbilical vein endothelial cells. Arch Pharm Res 2014;37(9):1183. https://doi.org/10.1007/s12272-014-0340-6
  7. Jeong A, Lee HJ, Jeong SJ, Lee HJ, Lee EO, Bae H, Kim SH. Compound K inhibits basic fibroblast growth factor-induced angiogenesis via regulation of p38 mitogen activated protein kinase and AKT in human umbilical vein endothelial cells. Biol Pharm Bull 2010;33(6):945-50. https://doi.org/10.1248/bpb.33.945
  8. Shin DH, Leem DG, Shin JS, Kim JI, Kim KY, Choi SY, Lee MH, Choi JH, Lee KT. Compound K induced apoptosis via endoplasmic reticulum Ca2+ release through ryanodine receptor in human lung cancer cells. J Ginseng Res 2017.
  9. Hu C, Song G, Zhang B, Liu Z, Chen R, Zhang H, Hu T. Intestinal metabolite compound K of panaxoside inhibits the growth of gastric carcinoma by augmenting apoptosis via Bid-mediated mitochondrial pathway. J Cell Mol Med 2012;16(1):96. https://doi.org/10.1111/j.1582-4934.2011.01278.x
  10. Lee IK, Kang KA, Lim CM, Kim KC, Kin HS, Kim DH, Kim BJ, Chang WY, Choi JH, Hyun JW. Compound K, a metabolite of ginseng saponin, induces mitochondria-dependent and caspase-dependent apoptosis via the generation of reactive oxygen species in human colon cancer cells. Int J Mol Sci 2010;11(12):4916-31. https://doi.org/10.3390/ijms11124916
  11. Wang CZ, Zhang Z, Wan JY, Zhang CF, Anderson S, He X, Yu C, He TC, Qi LW, Yuan CS. Protopanaxadiol, an active ginseng metabolite, significantly enhances the effects of fluorouracil on colon cancer. Nutrients 2015;7(2):799. https://doi.org/10.3390/nu7020799
  12. Zhang K, Li Y. Effects of ginsenoside compound K combined with cisplatin on the proliferation, apoptosis and epithelial mesenchymal transition in MCF-7 cells of human breast cancer. Pharm Biol 2015;54(4):1.
  13. Kang S, Siddiqi MH, Yoon SJ, Ahn S, Noh HY, Kumar NS, Kin YJ, Yang DC. Therapeutic potential of compound K as an IKK inhibitor with implications for osteoarthritis prevention: an in silico and in vitro study. Vitro Cell Develop Biol Anim 2016:1-11.
  14. Luedde T, Schwabe RF. $NF-{\kappa}B$ in the liver-linking injury, fibrosis and hepatocellular carcinoma. Nat Rev Gastroenterol Hepatol 2011;8(2):108-18. https://doi.org/10.1038/nrgastro.2010.213
  15. Bonavida B. RKIP-mediated chemo-immunosensitization of resistant cancer cells via disruption of the $NF-{\kappa}B$/snail/YY1/RKIP resistance-driver loop. Crit Rev Oncog 2014;19(6):431. https://doi.org/10.1615/CritRevOncog.2014011929
  16. Arlt A, Gehrz A, Muerkoster S, Vorndamn J, Kruse ML, Folsch UR, Schafer H. Role of NF-kappaB and Akt/PI3K in the resistance of pancreatic carcinoma cell lines against gemcitabine-induced cell death. Oncogene 2003;22(21):3243. https://doi.org/10.1038/sj.onc.1206390
  17. Karin M. NF-kappaB as a critical link between inflammation and cancer. Cold Spring Harb Perspect Biol 2009;1(5):a000141. https://doi.org/10.1101/cshperspect.a000141
  18. Wasmuth HE, Tacke F, Trautwein C. Chemokines in liver inflammation and fibrosis. Front Biosci J Virtual Lib 2002;7(03):d1899. https://doi.org/10.2741/A887
  19. Okuda K. Hepatocellular carcinoma. Curr Opin Oncol 2000;32(4):357.
  20. Luedde T, Trautwein C. Intracellular survival pathways in the liver. Liver Int 2006;26(10):1163-74. https://doi.org/10.1111/j.1478-3231.2006.01366.x
  21. Yang WS, Yi YS, Kim D, Kim MH, Park JG, Kim E, Lee SY, Yoon K, Kim JH, Park J, et al. Nuclear factor kappa-B- and activator protein-1-mediated immunostimulatory activity of compound K in monocytes and macrophages. J Ginseng Res 2017;41(3):298-306. https://doi.org/10.1016/j.jgr.2016.06.004
  22. Choi K, Kim M, Ryu J, Choi C. Ginsenosides compound K and Rh 2, inhibit tumor necrosis factor-${\alpha}$-induced activation of the $NF-{\kappa}B$ and JNK pathways in human astroglial cells. Neurosci Lett 2007;421(1):37. https://doi.org/10.1016/j.neulet.2007.05.017
  23. Ming Y, Chen Z, Chen L, Lin D, Tong Q, Zheng Z, Song G. Ginsenoside compound K attenuates metastatic growth of hepatocellular carcinoma, which is associated with the translocation of nuclear factor-kB p65 and reduction of matrix metalloproteinase-2/9. Planta Med 2010;77(5):428-33. https://doi.org/10.1055/s-0030-1250454
  24. Jung H, Kim JS, Kim WK, Oh KJ, Kim JM, Lee HJ, Han BS, Kim DS, Seo YS, Lee SC, et al. Intracellular annexin A2 regulates $NF-{\kappa}B$ signaling by binding to the p50 subunit: implications for gemcitabine resistance in pancreatic cancer. Cell Death Dis 2015;6(1):e1606. https://doi.org/10.1038/cddis.2014.558
  25. Wang YS, Lin Y, Li H, Li Y, Song ZG, Jin YH. The identification of molecular target of (20S) ginsenoside Rh2 for its anti-cancer activity. Sci Rep 2017.
  26. Wang ZM, Zhu SG, Wu ZW, Lu Y, Fu HZ, Qian RQ. Kirenol upregulates nuclear annexin-1 which interacts with $NF-{\kappa}B$ to attenuate synovial inflammation of collagen-induced arthritis in rats. J Ethnopharmacol 2011;137(1):774-82. https://doi.org/10.1016/j.jep.2011.06.037
  27. Jeon YJ, Kim DH, Jung H, Chung SJ, Chi SW, Cho S, Lee SC, Park BC, Park SG, Bae KH. Annexin A4 interacts with the NF-kappaB p50 subunit and modulates NF-kappaB transcriptional activity in a Ca2+-dependent manner. Cell Mol Life Sci 2010;67(13):2271. https://doi.org/10.1007/s00018-010-0331-9
  28. Campbell KA, Minashima T, Zhang Y, Hadley S, Lee YJ, Giovinazzo J, Quirno M, Jirsch T. Annexin A6 interacts with p65 and stimulates $NF-{\kappa}B$ activity and catabolic events in articular chondrocytes. Arthr Rheumatol 2013;65(12):3120. https://doi.org/10.1002/art.38182
  29. Ecsedi P, Kiss B, Gogl G, Radnai L, Buday L, Koprivanacz K, Liliom K, Leveles I, Vertessy B, Jeszen}o N, et al. Regulation of the equilibrium between closed and open conformations of annexin A2 by N-terminal phosphorylation and S100A4-binding. Structure 2017;25(8). 1195-1207.e5. https://doi.org/10.1016/j.str.2017.06.001
  30. Wang YS, Li H, Li Y, Zhu H, Jin YH. Identification of natural compounds targeting Annexin A2 with an anti-cancer effect. Protein Cell 2017.
  31. Liu P, Kimmoun E, Legrand A, Sauvanet A, Degott C, Lardeux B, Bernuau D. Activation of $NF-{\kappa}B$, AP-1, and STAT transcription factors is a frequent and early event in human hepatocellular carcinomas. J Hepatol 2002;37(1):63-71. https://doi.org/10.1016/S0168-8278(02)00064-8
  32. Lu B, Guo H, Zhao J, Wang C, Wu G, Pang M, Tong X, Bu F, Liang A, Hou S, et al. Increased expression of iASPP, regulated by hepatitis B virus X proteinmediated $NF-{\kappa}B$ activation, in hepatocellular carcinoma. Gastroenterology 2010;139(6):2183-94. e5. https://doi.org/10.1053/j.gastro.2010.06.049
  33. Zhang F, Wang Q, Ye L, Feng Y, Zhang X. Hepatitis B virus X protein upregulates expression of calpain small subunit 1 via nuclear factor-kappaB/p65 in hepatoma cells. J Med Virol 2010;82(6):920-8. https://doi.org/10.1002/jmv.21753
  34. Xiao C, Ghosh S. NF-kappaB, an evolutionarily conserved mediator of immune and inflammatory responses. Adv Exp Med Biol 2005;560:41. https://doi.org/10.1007/0-387-24180-9_5

Cited by

  1. Amino acid sequences characterization and anti-inflammatory potency evaluation of Portulaca oleracea L. oligopeptides in macrophages vol.10, pp.12, 2019, https://doi.org/10.1039/c9ra10465h
  2. Ginsenoside CK Inhibits Hypoxia-Induced Epithelial-Mesenchymal Transformation through the HIF-1α/NF-κB Feedback Pathway in Hepatocellular Carcinoma vol.10, pp.6, 2019, https://doi.org/10.3390/foods10061195
  3. The Anti-Tumor Effect and Underlying Apoptotic Mechanism of Ginsenoside Rk1 and Rg5 in Human Liver Cancer Cells vol.26, pp.13, 2019, https://doi.org/10.3390/molecules26133926
  4. Anti-Gastritis and Anti-Lung Injury Effects of Pine Tree Ethanol Extract Targeting Both NF-κB and AP-1 Pathways vol.26, pp.20, 2019, https://doi.org/10.3390/molecules26206275