Open Access

4‑Methoxydalbergione inhibits esophageal carcinoma cell proliferation and migration by inactivating NF‑κB

  • Authors:
    • Ming Li
    • Yubo Xiao
    • Pinyue Liu
    • Le Wei
    • Ti Zhang
    • Ziye Xiang
    • Xiaoyan Liu
    • Keyun Zhang
    • Qiaoqing Zhong
    • Fangzhi Chen
  • View Affiliations

  • Published online on: January 12, 2023     https://doi.org/10.3892/or.2023.8479
  • Article Number: 42
  • Copyright: © Li et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

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


Abstract

4‑Methoxydalbergione (4‑MD) can inhibit the progression of certain types of cancer; however, its effects on esophageal cancer (EC) remain unclear. The present study aimed to investigate the inhibitory effect of 4‑MD on EC and its molecular mechanism. ECA‑109 and KYSE‑105 cells were treated with or without lipopolysaccharide (LPS) and 4‑MD. Cell Counting Kit‑8 and colony formation assays were used to analyze cell proliferation. Wound healing assay was performed to evaluate cell migration. ELISA and western blotting were performed to measure the expression levels of NF‑κB and inflammatory cytokines. In cells treated with 4‑MD, proliferation and migration were significantly inhibited, the levels of inflammatory cytokines were downregulated and the NF‑κB signaling pathway was inactivated. Notably, proliferation, migration, inflammation and NF‑κB were promoted by LPS, whereas 4‑MD reversed the increases induced by LPS in EC cells. In conclusion, 4‑MD may attenuate the proliferation and migration of EC cells by inactivating the NF‑κB signaling pathway.

Introduction

It has previously been reported that the incidence of esophageal cancer (EC) is ~604,100 per year, thus accounting for 3.1% of new cancer cases diagnosed and ranking 7th in cancer incidence worldwide. In addition, it has been reported that there are 544,000 cases of EC-associated mortality per year, accounting for 5.5% of cancer-associated deaths and ranking 6th in cancer deaths worldwide (1). EC is divided into squamous cell carcinoma and adenocarcinoma according to histological subtype (2). The highest incidence of esophageal squamous cell carcinoma has been reported to occur in Eastern Asia, especially in China (1). EC is difficult to diagnose at an early stage due to the lack of specific biomarkers; therefore, patients are nearly always diagnosed with advanced EC (3). The overall 5-year survival of patients with EC is 15–20% worldwide (4); therefore, a novel therapeutic strategy to improve the 5-year survival rate of patients with EC is required.

During the COVID-19 pandemic, it was revealed that traditional Chinese medicine (TCM), such as Jinhua Qinggan granules and Lianhua Qingwen capsules, had a decisive role in the clinical treatment of COVID-19 by targeting ACE (5). Furthermore, a large number of studies have demonstrated that TCM serves an important role in the treatment of cancer, including colorectal cancer (6), breast cancer (7), hepatocellular carcinoma (8), lung cancer (9) and EC (1012). Notably, the active ingredients of TCM are thought to inhibit cancer progression by regulating certain genes or signaling pathways.

4-Methoxydalbergione (4-MD) is a flavonoid with methoxy groups, which is isolated and purified from Dalbergia sissoo Roxb. 4-MD has been indicated to have anti-inflammatory effects via inhibition of the NF-κB signaling pathway (13) and antitumor effects due to its high cytotoxicity in tumor cells (14). In addition, it has been demonstrated that 4-MD can inhibit osteosarcoma (15), human astroglioma (16) and bladder cancer (17) by regulating multiple signaling pathways, including JAK2/STAT3 and Akt/ERK, but not NF-κB. However, the inhibitory effect of 4-MD on EC remains to be determined.

It has been reported that the development of EC is a multistep pathogenic process from inflammation to cancer (18,19). The malignant degree of EC is positively associated with inflammatory factors, such as tumor necrosis factor α (TNF-α) (20), and the survival time of patients with EC has been confirmed to be negatively correlated with inflammation (21,22). Furthermore, EC is accelerated though EC angiogenesis via activation of the NF-κB signaling pathway (23). Therefore, inflammation may accelerate the progression of EC. Whether 4-MD regulates the NF-κB signaling pathway to inhibit EC remains unclear. Previous studies have shown that NF-κB can promote the release of TNF-α (24,25), the synthesis of prostaglandin E2 (PGE2) (2628), and the expression of cyclin-dependent kinase 1 (CDK1) (29,30), cyclin D1 (31,32) and proliferating cell nuclear antigen (PCNA) (33,34). However, whether 4-MD inhibits EC by suppressing the release of PGE2 and TNF-α, and the expression of proliferation-associated proteins through the NF-κB signaling pathway requires further investigation. Therefore, the present study aimed to explore the effects of 4-MD on cell proliferation and invasion, and to determine the underlying mechanism by which 4-MD regulates the malignant characteristics of EC cells.

Materials and methods

Cell culture

ECA-109 esophageal squamous carcinoma cells were purchased from the American Type Culture Collection and KYSE-150 esophageal squamous carcinoma cells were purchased from The Cell Bank of Type Culture Collection of The Chinese Academy of Sciences. ECA-109 cells were cultured in Dulbecco's modified Eagle's medium (cat. no. SH30021; Hyclone; Cytiva) and KYSE-150 cells were cultured in RPMI 1640 (cat. no. 11875093; Gibco; Thermo Fisher Scientific, Inc.). The media were supplemented with 10% fetal bovine serum (cat. no. SH30396; Hyclone; Cytiva) and 1% penicillin-streptomycin (cat. no. V900929; MilliporeSigma). Cells were incubated in a humidified incubator containing 5% CO2 at 37°C.

Drug preparation

4-MD (cat. no. CB31393122; Chemical Book) was dissolved in DMSO to form a 10 mmol/l solution, which was stored at −20°C. The storage solution was not diluted to the corresponding concentration of the working solution until it was used.

Cell Counting Kit-8 (CCK-8) assay

The proliferation of cells was analyzed using the CCK-8 assay. ECA-109 cells were seeded into 96-well plates at 1,000 cells/well, and were treated with different concentrations of 4-MD (0, 3.125, 6.25, 12.5, 25, 50 and 100 µmol/l) for 24 h. The optical density (OD) of the cells was evaluated using a spectrophotometer at a wavelength of 450 nm. Using the OD of cells, the half maximal inhibitory concentration (IC50) and IC50 95% confidence interval (CI) were calculated using GraphPad Prism 8.3 (GraphPad Software, Inc.). In addition, ECA-109 and KYSE-150 cells were seeded into 96-well plates at 1,000 cells/well, and were pre-incubated with or without 1 µg/ml lipopolysaccharide (LPS; cat. no. ST1470; Beyotime Institute of Biotechnology) for 30 min at 37°C in an incubator with 5% CO2 to activate NF-κB (35), followed by treatment with or without 20 µmol/l 4-MD for 12, 24, 36 or 48 h at 37°C in an incubator with 5% CO2. Subsequently,10 µl CCK-8 (cat. no. C0038; Beyotime Institute of Biotechnology) solution was added to each well. After culture for 1 h in an incubator, the OD value of the cells was evaluated.

Colony formation assay

ECA-109 and KYSE-150 cells were re-suspended in culture medium. Subsequently, 1,000 cells/well were seeded into 24-well plates, and were treated with or without 20 µmol/l 4-MD and 1 µg/ml LPS for 10 days in a humidified incubator containing 5% CO2 at 37°C. ECA-109 and KYSE-150 cells were then fixed with 4% paraformaldehyde (cat. no. P0099; Beyotime Institute of Biotechnology) for 30 min at room temperature (RT) and were stained with crystal violet (cat. no. C0121; Beyotime Institute of Biotechnology) for 5 min at RT. Finally, the cell colonies containing ≥50 cells were observed, images were captured under a light microscope and colonies were analyzed using ImageJ software (version 1.8.0; National Institutes of Health).

Wound-healing assay

ECA-109 and KYSE-150 cells were used for wound-healing assay. A total of 1ⅹ105 cells/well were seeded into 12-well plates. Once cells reached 100% confluence, a scratch was evenly drawn using a 10-µl pipette tip. Subsequently, the cells were incubated in FBS-free medium in a 5% CO2 incubator at 37°C, and treated with or without 20 µmol/l 4-MD and 1 µg/ml LPS for a further 36 or 48 h. Finally, the images of wound healing were captured under a light microscope. The wound area was analyzed using ImageJ software. The rate of migration was calculated using the following formula: Rate of migration (%)=(scratch area at 0 h-scratch area at 48 h)/scratch area at 0 h ⅹ100.

ELISA

The levels of TNF-α and PGE2 in the culture supernatant of ECA-109 cells treated with or without LPS and 4-MD were analyzed using TNF-α (cat. no. PT518; Beyotime Institute of Biotechnology) and PGE2 (cat. no. HB833-Hu; Shanghai Hengyuan Biotechnology Co., Ltd.) ELISA kits according to the manufacturers' protocols. OD value was measure at 450 nm using a spectrophotometer.

Antibodies

All antibodies were purchased from Beyotime Institute of Biotechnology. The antibodies used for western blotting (WB) were as follows: Cyclin D1 rabbit monoclonal antibody (1:1,000; cat. no. AF1183), CDK1 rabbit polyclonal antibody (1:500; cat. no. AF0111), PCNA rabbit monoclonal antibody (1:1,000; cat. no. AF1363), phosphorylated (p)-IKBα (Ser32) rabbit monoclonal antibody (1:1,000; cat. no. AF1870), IKBα rabbit monoclonal antibody (1:1,000; cat. no. AF1282), P65 rabbit monoclonal antibody (1:1,000; cat. no. AF1234), p-NF-κB P65 (Ser536) rabbit polyclonal antibody (1:500; cat. no. AF5881), β-actin rabbit monoclonal antibody (1:2,000; cat. no. AF5003) and HRP-labeled goat anti-rabbit IgG (1:1,000; cat. no. A0208).

WB

ECA-109 cells were harvested and lysed in pre-chilled RIPA buffer (cat. no. P0013B; Beyotime Institute of Biotechnology) on ice for 30 min. The supernatants were collected after centrifugation of lysates at 13,300 × g for 30 min at 4°C. The concentrations of proteins were quantified using a NanoDrop ND-2000 spectrophotometer (NanoDrop; Thermo Fisher Scientific, Inc.). Subsequently, ~20 µg proteins were separated by SDS-PAGE on 10% gels. The proteins were then transferred onto PVDF membranes (cat. no. IPVH00010; MilliporeSigma) and blocked with 5% skim milk (cat. no. P0216; Beyotime Institute of Biotechnology) for 2 h at room temperature (RT). The membranes were incubated with primary antibodies overnight at 4°C, then incubated with the secondary antibody for 2 h at RT. Finally, the bands were observed by chemiluminescence using SuperSignal™ West Pico PLUS (cat. no. 34577; Thermo Fisher Scientific, Inc.), and images were captured using a gel imaging system (Chemidoc MP; Bio-Rad Laboratories, Inc.) and blots were analyzed using ImageJ software (version 1.8.0; National Institutes of Health).

Statistical analysis

All data from three or five independent experiments are presented as the mean ± SD, and were analyzed by SPSS 23.0 (IBM Corp.) or GraphPad Prism 8.3 software (GraphPad Software, Inc.). Unpaired Student's t-test was performed to analyze the differences between two groups. One-way ANOVA followed by Sidak's multiple comparisons test was used to analyze the differences among multiple groups. P<0.05 was considered to indicate a statistically significant difference.

Results

4-MD alleviates the proliferation and migration of ECA-109 cells

To explore the inhibitory effect of 4-MD on EC cells, ECA-109 cells were treated with different concentrations of 4-MD for 24 h. As shown in Fig. 1A, the IC50 of 4-MD was 15.79 µmol/l and the 95% CI of IC50 was 10.03-24.97 µmol/l. Subsequently, 20 µmol/l 4-MD was used for further studies. ECA-109 cells were treated with 4-MD, and the proliferation of cells was analyzed by CCK-8 and colony formation assays. Compared with those in the untreated control group, the OD values of cells treated with 4-MD were significantly lower at 36 and 48 h (Fig. 1B). In addition, the colonies of cells treated with 4-MD were fewer and smaller than those in the control group (Fig. 1C). A wound-healing assay was performed to evaluate the migration of ECA-109 cells treated with or without 4-MD for 48 h. The migration rate of control cells was ~80%, whereas the migration rate of cells treated with 4-MD was ~60%, thus suggesting that the migration of ECA-109 cells was suppressed by 4-MD (Fig. 1D). The expression levels of proliferation-associated proteins were determined by WB. The results revealed that the protein expression levels of cyclin D1, CDK1 and PCNA were significantly downregulated in cells treated with 4-MD compared with those in the control group (Fig. 1E). These results indicated that 4-MD inhibited proliferation and migration, and downregulated the expression levels of cyclin D1, CDK1 and PCNA in ECA-109 cells.

4-MD reduces the production of inflammatory cytokines in ECA-109 cells

To investigate the inhibitory effects of 4-MD on inflammation, the levels of inflammatory cytokines, including TNF-α and PGE2, were measured in ECA-109 cells by ELISA. As shown in Fig. 2A and B, the production of TNF-α and PGE2 was significantly decreased in ECA-109 cells treated with 4-MD. In addition, WB was performed to measure the expression levels of IKBα and P65. Compared with those in the control groups, the expression levels of p-IKBα and p-P65 were decreased in cells treated with 4-MD, thus suggesting that 4-MD could inactivate NF-κB in ECA-109 cells (Fig. 2C).

4-MD reverses the LPS-induced proliferation and migration of EC cells

To further assess whether 4-MD inhibits the proliferation and migration of EC cells by inactivating NF-κB, ECA-109 cells were treated with LPS to activate NF-κB, and subsequently treated with 4-MD. Results of the CCK-8 assay revealed that the proliferation of cells pre-incubated with LPS was significantly increased compared with that in the control group, whereas cell proliferation was inhibited following treatment with LPS and 4-MD, thus indicating that 4-MD partly inhibited the LPS-induced increase in cell proliferation (Fig. 3A). Furthermore, the size and number of cell colonies was increased in the LPS group compared with that in the control group, whereas the colonies in the group treated with LPS and 4-MD were fewer and smaller compared with in the group treated with LPS alone, indicating that 4-MD suppressed the acceleration of cell proliferation induced by LPS in ECA-109 cells (Fig. 3B). Cell migration was analyzed by wound healing. The migration rate of the control cells was ~80% 48 h after scratching; however, the migration rate was ~100% in the group of cells treated with LPS, and was ~90% in the group of cells treated with LPS and 4-MD, which suggested that 4-MD could partly abolish the LPS-induced increase in cell migration (Fig. 3C). To fully demonstrate the inhibitory effect of 4-MD on another EC cell line, KYSE-105 cells were treated with LPS and 4-MD, and proliferation and migration were measured. Notably, the results of CCK-8, colony formation and wound-healing assays in KYSE-105 cells were consistent with the results in ECA-109 cells, which indicated that 4-MD could inhibit the LPS-induced increase in the proliferation and migration of KYSE-105 cells (Fig. 3D-F).

4-MD inhibits the production of TNF-α and PGE2, and the expression levels of proliferation-related proteins by inactivating NF-κB in ECA-109 cells

To elucidate the potential mechanism underlying the suppressive effects of 4-MD on the proliferation and migration of EC cells, ECA-109 cells were incubated with LPS and 4-MD. As shown in Fig. 4A, the expression levels of cyclin D1, CDK1 and PCNA were increased in cells treated with LPS compared with those in the control group; meanwhile, the expression levels of these proteins were downregulated in cells treated with LPS and 4-MD compared with those in cells treated with LPS alone.

The production of TNF-α and PGE2 was analyzed by ELISA. The levels of TNF-α (Fig. 4B) and PGE2 (Fig. 4C) were increased in cells treated with LPS compared with those in the control group. By contrast, compared with in cells treated with LPS alone, the levels of TNF-α and PGE2 were decreased in cells treated with LPS and 4-MD. Compared with in the control cells, the protein expression levels of p-IKBα and p-P65 were increased in cells treated with LPS, whereas p-IKBα and p-P65 expression levels were significantly decreased in cells treated with LPS and 4-MD compared with those in cells treated with LPS alone (Fig, 4D), which indicated that 4-MD could inhibit LPS-induced activation of NF-κB.

Taken together, these results suggested that 4-MD inhibited the proliferation and migration of EC cells by decreasing TNF-α and PGE2 production, and downregulated cyclin D1, CDK1 and PCNA expression through inactivating the NF-κB pathway.

Discussion

Approximately 25% of cancer is caused by inflammation, including EC (36). EC is an aggressive malignant tumor of the digestive system, which is seriously life-threatening and has a complex pathogenesis. Studies have shown that microbial infections serve a catalytic role in EC development (3739). Furthermore, it has been suggested that inflammation triggered by infections is a notable cause of cancer (40,41). Increasing evidence has shown that inflammation is the main cause of EC induction (19). In the present study, the inflammatory inducer LPS (42,43) was used to promote inflammation in EC cells, which simulated the inflammatory response of EC cells.

Esophagectomy is one of the main treatments for advanced EC but is likely to impair quality of life (44). In a previous study, the single-cell transcriptomic analysis of EC indicated that genes related to macrophages and neutrophils were activated in early esophageal tissue, which created a chronic inflammatory environment that accelerated the progression of EC (18). During the follow-up of patients with EC, it has been revealed that the levels of inflammatory cytokines in recurrent EC are significantly higher than those in patients without recurrence, and inflammation is positively associated with the recurrence of EC, thus suggesting that inflammation could accelerate EC recurrence (45). Under inflammatory stimulation, the expression levels of microRNA-302b have been reported to be decreased in EC cells, resulting in increased expression levels of ERBB4, IRF2 and CXCR4, which may promote tumor cell growth (46). These findings suggested that inflammation could promote the development and recurrence of EC. Therefore, inhibiting inflammation may be a therapeutic strategy for the treatment of EC. Patients with EC undergoing continuous oral administration of vitamin C exhibited reduced drug resistance via the inactivation of NF-κB (47). Furthermore, curcumin has been shown to enhance the sensitivity of EC to chemotherapy by reducing the expression levels of cyclooxygenase-2 and lipoxygenase through inhibiting the inflammatory response (48). Natural astaxanthin (49) and lycopene (50) may also significantly inhibit the occurrence of EC by suppressing NF-κB activity. These results indicated that NF-κB may be an effective molecular target for the treatment of EC.

Notably, the active ingredients of TCM have attracted attention due to their inhibitory effects on cancer. A previous study showed that sulforaphene activated the GADD45B/MAP2K3/p38/p53 feedback loop, and downregulated the expression levels of SCD and CDH3 to alleviate the progression of EC (51). Echinatin has also been shown to inhibit the proliferation and invasion of EC cells by inducing AKT/mTOR signaling pathway-dependent autophagy and apoptosis (52). These findings indicated that the active ingredients of TCM may act on different genes/signaling pathways to inhibit the progression of EC. 4-MD was isolated and purified from D. sissoo Roxb. It has been shown that 4-MD can suppress the proliferation and induce apoptosis of human osteosarcoma cells by downregulating the JAK2/STAT3 pathway in vitro and in vivo (15). 4-MD can also effectively arrest the cell cycle of astroglioma cells in G2 phase, and regulate multiple genes that enrich the cell cycle and the p53, TNF and MAPK signaling pathways (16). 4-MD has been reported to attenuate the proliferation of bladder cancer cells by inducing autophagy and suppressing the Akt/ERK signaling pathway in vitro (17). The present study revealed that the OD value and the number of cell colonies were reduced, the size of colonies was smaller, and wound healing was significantly inhibited in EC cells treated with 4-MD. Furthermore, WB showed that the expression levels of cyclin D1, CDK1 and PCNA were decreased in EC cells treated with 4-MD. These results indicated that 4-MD may be a potential drug for the treatment of EC that functions by downregulating cyclin D1, CDK1 and PCNA.

It has previously been reported that 4-MD can suppress inflammatory responses in addition to being a potent inhibitor of cancer. 4-MD has been demonstrated to exert cytoprotection by inducing anti-inflammatory effects, specifically via inhibiting the LPS-induced production of nitric oxide and PGE2 in microglia (53) and reducing LPS-induced inflammation in RAW264.7 cells (54,55). In the present study, the levels of TNF-α and PGE2 were decreased in EC cells treated with 4-MD, as determined by ELISA. Subsequently, the related proteins in the NF-κB signaling pathway were analyzed by WB, and the results revealed that p-IKBα and p-P65 expression levels were significantly reduced, thus suggesting that 4-MD reduced inflammatory cytokines by inactivating NF-κB in EC cells. To confirm that 4-MD inhibited the proliferation and migration of EC cells by suppressing inflammation, EC cells were treated with the NF-κB agonist LPS. Proliferation, migration, and TNF-α and PGE2 levels were markedly increased, and the NF-κB signaling pathway was activated in response to LPS. These results were consistent with the finding that LPS can promote the proliferation of EC cells (35,56). Following activation of NF-κB, EC cells were treated with 4-MD. Notably, 4-MD partially reversed the LPS-induced increases in proliferation, migration and inflammatory cytokines (TNF-α and PGE2), and activation of the NF-κB signaling pathway.

In conclusion, the results of the present study demonstrated that 4-MD significantly inhibited proliferation and migration by inactivating the NF-κB signaling pathway in EC cells, thus providing a novel strategy for 4-MD-induced inhibition of NF-κB in the treatment of EC. However, the present study has the following limitations: First, esophageal inflammation is one of the pathogeneses that accelerate the occurrence of EC. 4-MD can inhibit the malignant characteristics of EC cells by reducing the release of inflammatory factors; however, the present study does not show that the antitumor effect of 4-MD is superior to other antitumor drugs. Second, the present study revealed that 4-MD inhibited EC by inactivating the NF-κB signaling pathway; however, whether the inactivation of NF-κB is the dominant role of 4-MD in tumor suppression requires further investigation. Third, 4-MD has only been demonstrated to inhibit the proliferation of EC cells by suppressing NF-κB in vitro; therefore, further studies are needed to explore the inhibitory effect of 4-MD on EC in vivo.

Acknowledgements

Not applicable.

Funding

This work is supported by the Scientific Research Fund of Hunan Provincial Education Department (grant no. 21A0612), the Hunan Provincial Natural Science Foundation of China (grant no. 2021JJ30482), the Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair (grant no. GXLIRMMKL-K202006) and the Rehabilitation project of Hunan Disabled Persons' Federation (grant no. 2022XK0223).

Availability of data and materials

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

Authors' contributions

QZ and FC designed the study and revised the manuscript. ML wrote the manuscript, performed cell culture and migration assays, and analyzed the data. YB, PY, LW, TZ, ZX and XL performed the experiments, including the proliferation assay and WB. KZ performed the WB experiment shown in Fig. 4A, analyzed these data, described the relevant results and financially sponsored this study. All authors read and approved the final manuscript. ML and FC confirm the authenticity of all the raw data.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

4-MD

4-methoxydalbergione

EC

esophageal cancer

LPS

lipopolysaccharide

CCK-8

Cell Counting Kit-8

OD

optical density

TCM

traditional Chinese medicine

WB

western blotting

RT

room temperature

References

1 

Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A and Bray F: Global Cancer Statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 71:209–249. 2021. View Article : Google Scholar : PubMed/NCBI

2 

Rogers JE, Sewastjanow-Silva M, Waters RE and Ajani JA: Esophageal cancer: Emerging therapeutics. Expert Opin Ther Targets. 26:107–117. 2022. View Article : Google Scholar : PubMed/NCBI

3 

Fan J, Liu Z, Mao X, Tong X, Zhang T, Suo C and Chen X: Global trends in the incidence and mortality of esophageal cancer from 1990 to 2017. Cancer Med. 9:6875–6887. 2020. View Article : Google Scholar : PubMed/NCBI

4 

Uhlenhopp DJ, Then EO, Sunkara T and Gaduputi V: Epidemiology of esophageal cancer: Update in global trends, etiology and risk factors. Clin J Gastroenterol. 13:1010–1021. 2020. View Article : Google Scholar : PubMed/NCBI

5 

Huang K, Zhang P, Zhang Z, Youn JY, Wang C, Zhang H and Cai H: Traditional Chinese Medicine (TCM) in the treatment of COVID-19 and other viral infections: Efficacies and mechanisms. Pharmacol Ther. 225:1078432021. View Article : Google Scholar : PubMed/NCBI

6 

Huang S, Zhang Z, Li W, Kong F, Yi P, Huang J, Mao D, Peng W and Zhang S: Network Pharmacology-Based prediction and verification of the active ingredients and potential targets of Zuojinwan for treating colorectal cancer. Drug Des Devel Ther. 14:2725–2740. 2020. View Article : Google Scholar : PubMed/NCBI

7 

Yang Z, Zhang Q, Yu L, Zhu J, Cao Y and Gao X: The signaling pathways and targets of traditional Chinese medicine and natural medicine in triple-negative breast cancer. J Ethnopharmacol. 264:1132492021. View Article : Google Scholar : PubMed/NCBI

8 

Jingjing H, Hongna H, Xiaojiao W, Yan G, Yuexue Z and Yueqiang H: Bie Jia Jian pill enhances the amelioration of bone mesenchymal stem cells on hepatocellular carcinoma progression. J Nat Med. 76:49–58. 2022. View Article : Google Scholar : PubMed/NCBI

9 

Kan Y, Song M, Cui X, Yang Q, Zang Y, Li Q, Li Y, Cai W, Chen Y, Weng X, et al: Muyin extract inhibits non-small-cell lung cancer growth by inducing autophagy and apoptosis in vitro and in vivo. Phytomedicine. 96:1538342022. View Article : Google Scholar : PubMed/NCBI

10 

Kong L, Lu X, Chen X, Wu Y, Zhang Y, Shi H and Li J: Qigesan inhibits esophageal cancer cell invasion and migration by inhibiting Gas6/Axl-induced epithelial-mesenchymal transition. Aging (Albany NY). 12:9714–9725. 2020. View Article : Google Scholar : PubMed/NCBI

11 

Wu Z, Zhao Y, Yu F, Shi H and Li J: Qigefang inhibits migration, invasion, and metastasis of ESCC by Inhibiting Gas6/Axl signaling pathway. Recent Pat Anticancer Drug Discov. 16:285–294. 2021. View Article : Google Scholar : PubMed/NCBI

12 

Li H, Ma C, Chang S, Xi X, Shao S, Chen M, Ren J, Sun M and Dong L: Traditional Chinese Medicine decoctions improve longevity following diagnosis with stage IV esophageal squamous cell carcinoma: A retrospective analysis. Int J Gen Med. 15:1665–1675. 2022. View Article : Google Scholar : PubMed/NCBI

13 

Funakoshi-Tago M, Okamoto K, Izumi R, Tago K, Yanagisawa K, Narukawa Y, Kiuchi F, Kasahara T and Tamura H: Anti-inflammatory activity of flavonoids in Nepalese propolis is attributed to inhibition of the IL-33 signaling pathway. Int Immunopharmacol. 25:189–198. 2015. View Article : Google Scholar : PubMed/NCBI

14 

Hirano T, Abe K, Gotoh M and Oka K: Citrus flavone tangeretin inhibits leukaemic HL-60 cell growth partially through induction of apoptosis with less cytotoxicity on normal lymphocytes. Br J Cancer. 72:1380–1388. 1995. View Article : Google Scholar : PubMed/NCBI

15 

Park KR, Yun HM, Quang TH, Oh H, Lee DS, Auh QS and Kim EC: 4-Methoxydalbergione suppresses growth and induces apoptosis in human osteosarcoma cells in vitro and in vivo xenograft model through down-regulation of the JAK2/STAT3 pathway. Oncotarget. 7:6960–6971. 2016. View Article : Google Scholar : PubMed/NCBI

16 

Li R, Xu CQ, Shen JX, Ren QY, Chen DL, Lin MJ, Huang RN, Li CH, Zhong RT, Luo ZH, et al: 4-Methoxydalbergione is a potent inhibitor of human astroglioma U87 cells in vitro and in vivo. Acta Pharmacol Sin. 42:1507–1515. 2021. View Article : Google Scholar : PubMed/NCBI

17 

Du H, Tao T, Xu S, Xu C, Li S, Su Q, Yan J, Liu B and Li R: 4-Methoxydalbergione inhibits bladder cancer cell growth via inducing autophagy and inhibiting Akt/ERK signaling pathway. Front Mol Biosci. 8:7896582022. View Article : Google Scholar : PubMed/NCBI

18 

Yao J, Cui Q, Fan W, Ma Y, Chen Y, Liu T, Zhang X, Xi Y, Wang C, Peng L, et al: Single-cell transcriptomic analysis in a mouse model deciphers cell transition states in the multistep development of esophageal cancer. Nat Commun. 11:37152020. View Article : Google Scholar : PubMed/NCBI

19 

Ekheden I, Ludvigsson JF, Yin L, Elbe P and Ye W: Esophageal abnormalities and the risk for gastroesophageal cancers-a histopathology-register-based study in Sweden. Eur J Epidemiol. 37:401–411. 2022. View Article : Google Scholar : PubMed/NCBI

20 

Keeley BR, Islami F, Pourshams A, Poustchi H, Pak JS, Brennan P, Khademi H, Genden EM, Abnet CC, Dawsey SM, et al: Prediagnostic serum levels of inflammatory biomarkers are correlated with future development of lung and esophageal cancer. Cancer Sci. 105:1205–1211. 2014. View Article : Google Scholar : PubMed/NCBI

21 

Wu C, Wang M, Zhou Q and Shi H: Associations of changes in intestinal flora and inflammatory factors with prognosis of patients with esophageal cancer. J Healthc Eng. 2022:24263012022. View Article : Google Scholar : PubMed/NCBI

22 

Sugawara K, Yagi K, Okumura Y, Aikou S, Yamashita H and Seto Y: Survival prediction capabilities of preoperative inflammatory and nutritional status in esophageal squamous cell carcinoma patients. World J Surg. 46:639–647. 2022. View Article : Google Scholar : PubMed/NCBI

23 

Chen Y, Wang D, Peng H, Chen X, Han X, Yu J, Wang W, Liang L, Liu Z, Zheng Y, et al: Epigenetically upregulated oncoprotein PLCE1 drives esophageal carcinoma angiogenesis and proliferation via activating the PI-PLCℇ-NF-κB signaling pathway and VEGF-C/Bcl-2 expression. Mol Cancer. 18:12019. View Article : Google Scholar : PubMed/NCBI

24 

Sojoodi M, Erstad DJ, Barrett SC, Salloum S, Zhu S, Qian T, Colon S, Gale EM, Jordan VC, Wang Y, et al: Peroxidasin deficiency re-programs macrophages toward pro-fibrolysis function and promotes collagen resolution in liver. Cell Mol Gastroenterol Hepatol. 13:1483–1509. 2022. View Article : Google Scholar : PubMed/NCBI

25 

Balzano T, Arenas YM, Dadsetan S, Forteza J, Gil-Perotin S, Cubas-Nuñez L, Casanova B, Gracià F, Varela-Andrés N, Montoliu C, et al: Sustained hyperammonemia induces TNF-a IN Purkinje neurons by activating the TNFR1-NF-κB pathway. J Neuroinflammation. 17:702020. View Article : Google Scholar : PubMed/NCBI

26 

Yamada T, Osawa S, Ikuma M, Kajimura M, Sugimoto M, Furuta T, Iwaizumi M and Sugimoto K: Guggulsterone, a plant-derived inhibitor of NF-TB, suppresses CDX2 and COX-2 expression and reduces the viability of esophageal adenocarcinoma cells. Digestion. 90:208–217. 2014. View Article : Google Scholar : PubMed/NCBI

27 

Zhao J, Bi W, Xiao S, Lan X, Cheng X, Zhang J, Lu D, Wei W, Wang Y, Li H, et al: Neuroinflammation induced by lipopolysaccharide causes cognitive impairment in mice. Sci Rep. 9:57902019. View Article : Google Scholar : PubMed/NCBI

28 

Sousa FBM, Pacheco G, Oliveira AP, Nicolau LAD, Lopes ALF, Ferreira-Fernandes H, Pinto GR and Medeiros JVR: Mechanism of preservation of the intestinal mucosa architecture and NF-κB/PGE2 reduction by hydrogen sulfide on cholera toxin-induced diarrhea in mice. Life Sci. 284:1198692021. View Article : Google Scholar : PubMed/NCBI

29 

Voce DJ, Bernal GM, Cahill KE, Wu L, Mansour N, Crawley CD, Campbell PS, Arina A, Weichselbaum RR and Yamini B: CDK1 is up-regulated by temozolomide in an NF-κB dependent manner in glioblastoma. Sci Rep. 11:56652021. View Article : Google Scholar : PubMed/NCBI

30 

Han D, Zhu S, Li X, Li Z, Huang H, Gao W, Liu Y, Zhu H and Yu X: The NF-κB/miR-488/ERBB2 axis modulates pancreatic cancer cell malignancy and tumor growth through cell cycle signaling. Cancer Biol Ther. 23:294–309. 2022. View Article : Google Scholar : PubMed/NCBI

31 

Gu P, Zhang M, Zhu J, He X and Yang D: Suppression of CDCA3 inhibits prostate cancer progression via NF-κB/cyclin D1 signaling inactivation and p21 accumulation. Oncol Rep. 47:422022. View Article : Google Scholar : PubMed/NCBI

32 

Wang X, Liu X, Yang Y and Yang D: Cyclin D1 mediated by the nuclear translocation of nuclear factor kappa B exerts an oncogenic role in lung cancer. Bioengineered. 13:6866–6879. 2022. View Article : Google Scholar : PubMed/NCBI

33 

Zhao Y, Liu C, Gao Z, Shao D, Zhao X, Wei Q and Ma B: G protein-coupled estrogen receptor 1 mediates proliferation and adipogenic differentiation of goat adipose-derived stem cells through ERK1/2-NF-κB signaling pathway. Acta Biochim Biophys Sin (Shanghai). 54:494–503. 2022. View Article : Google Scholar : PubMed/NCBI

34 

Zhang W and Wang Y: Activation of RIPK2-mediated NOD1 signaling promotes proliferation and invasion of ovarian cancer cells via NF-κB pathway. Histochem Cell Biol. 157:173–182. 2022. View Article : Google Scholar : PubMed/NCBI

35 

Zu Y, Ping W, Deng T, Zhang N, Fu X and Sun W: Lipopolysaccharide-induced toll-like receptor 4 signaling in esophageal squamous cell carcinoma promotes tumor proliferation and regulates inflammatory cytokines expression. Dis Esophagus. 30:1–8. 2017.PubMed/NCBI

36 

Kawanishi S, Ohnishi S, Ma N, Hiraku Y and Murata M: Crosstalk between DNA Damage and inflammation in the multiple steps of carcinogenesis. Int J Sci. 18:18082017.

37 

Holleczek B, Schottker B and Brenner H: Helicobacter pylori infection, chronic atrophic gastritis and risk of stomach and esophagus cancer: Results from the prospective population-based ESTHER cohort study. Int J Cancer. 146:2773–2783. 2020. View Article : Google Scholar : PubMed/NCBI

38 

Zhou Q, Wei Y, Zhai H, Li S, Xu R and Li P: Comorbid early esophageal cancer and Gongylonema pulchrum infection: A case report. BMC Gastroenterol. 21:3052021. View Article : Google Scholar : PubMed/NCBI

39 

Kawasaki M, Ikeda Y, Ikeda E, Takahashi M, Tanaka D, Nakajima Y, Arakawa S, Izumi Y and Miyake S: Oral infectious bacteria in dental plaque and saliva as risk factors in patients with esophageal cancer. Cancer. 127:512–519. 2021. View Article : Google Scholar : PubMed/NCBI

40 

Fujiwara N, Kitamura N, Yoshida K, Yamamoto T, Ozaki K and Kudo Y: Involvement of fusobacterium species in oral cancer progression: A literature review including other types of cancer. Int J Mol Sci. 21:62072020. View Article : Google Scholar : PubMed/NCBI

41 

Tian C, Chen K, Gong W, Yoshimura T, Huang J and Wang JM: The G-Protein coupled formyl peptide receptors and their role in the progression of digestive tract cancer. Technol Cancer Res Treat. 19:15330338209732802020. View Article : Google Scholar : PubMed/NCBI

42 

Liu CH, Chen Z, Chen K, Liao FT, Chung CE, Liu X, Lin YC, Keohavong P, Leikauf GD and Di YP: Lipopolysaccharide-Mediated chronic inflammation promotes tobacco carcinogen-induced lung cancer and determines the efficacy of immunotherapy. Cancer Res. 81:144–157. 2021. View Article : Google Scholar : PubMed/NCBI

43 

Li M, Liu H, Shao H, Zhang P, Gao M, Huang L, Shang P, Zhang Q, Wang W and Feng F: Glyburide attenuates B(a)p and LPS-induced inflammation-related lung tumorigenesis in mice. Environ Toxicol. 36:1713–1722. 2021. View Article : Google Scholar : PubMed/NCBI

44 

Watanabe M, Otake R, Kozuki R, Toihata T, Takahashi K, Okamura A and Mamura Y: Recent progress in multidisciplinary treatment for patients with esophageal cancer. Surg Today. 50:12–20. 2020. View Article : Google Scholar : PubMed/NCBI

45 

Zheng L, Jiang J, Liu Y, Zheng X and Wu C: Correlations of recurrence after radical surgery for esophageal cancer with glucose-lipid metabolism, insulin resistance, inflammation, stress and serum p53 expression. J BUON. 24:1666–1672. 2019.PubMed/NCBI

46 

Zhang M, Zhang L, Cui M, Ye W, Zhang P, Zhou S and Wang J: miR-302b inhibits cancer-related inflammation by targeting ERBB4, IRF2 and CXCR4 in esophageal cancer. Oncotarget. 8:49053–49063. 2017. View Article : Google Scholar : PubMed/NCBI

47 

Abdel-Latif MMM, Babar M, Kelleher D and Reynolds JV: A pilot study of the impact of Vitamin C supplementation with neoadjuvant chemoradiation on regulators of inflammation and carcinogenesis in esophageal cancer patients. J Cancer Res Ther. 15:185–191. 2019.PubMed/NCBI

48 

Komal K, Chaudhary S, Yadav P, Parmanik R and Singh M: The therapeutic and preventive efficacy of curcumin and its derivatives in esophageal cancer. Asian Pac J Cancer Prev. 20:1329–1337. 2019. View Article : Google Scholar : PubMed/NCBI

49 

Cui L, Xu F, Wang M, Li L, Qiao T, Cui H, Li Z and Sun C: Dietary natural astaxanthin at an early stage inhibits N-nitrosomethylbenzylamine-induced esophageal cancer oxidative stress and inflammation via downregulation of NFκB and COX2 in F344 rats. Onco Targets Ther. 12:5087–5096. 2019. View Article : Google Scholar : PubMed/NCBI

50 

Cui L, Xu F, Wu K, Li L, Qiao T, Li Z, Chen T and Sun C: Anticancer effects and possible mechanisms of lycopene intervention on N-methylbenzylnitrosamine induced esophageal cancer in F344 rats based on PPARү1. Eur J Pharmacol. 881:1732302020. View Article : Google Scholar : PubMed/NCBI

51 

Han S, Wang Y, Ma J, Wang Z, Wang HD and Yuan Q: Sulforaphene inhibits esophageal cancer progression via suppressing SCD and CDH3 expression, and activating the GADD45B-MAP2K3-p38-p53 feedback loop. Cell Death Dis. 11:7132020. View Article : Google Scholar : PubMed/NCBI

52 

Hong P, Liu QW, Xie Y, Zhang QH, Liao L, He QY, Li B and Xu WW: Echinatin suppresses esophageal cancer tumor growth and invasion through inducing AKT/mTOR-dependent autophagy and apoptosis. Cell Death Dis. 11:5242020. View Article : Google Scholar : PubMed/NCBI

53 

Kim DC, Lee DS, Ko W, Kim KW, Kim HJ, Yoon CS, Oh H and Kim YC: Heme Oxygenase-1-Inducing Activity of 4-Methoxydalbergione and 4′-Hydroxy-4-methoxydalbergione from Dalbergia odorifera and their anti-inflammatory and cytoprotective effects in murine hippocampal and BV2 microglial cell line and primary rat microglial cells. Neurotox Res. 33:337–352. 2018. View Article : Google Scholar : PubMed/NCBI

54 

Sun K, Su C, Li W, Gong Z, Sha C and Liu R: Quality markers based on phytochemical analysis and anti-inflammatory screening: An integrated strategy for the quality control of Dalbergia odorifera by UHPLC-Q-Orbitrap HRMS. Phytomedicine. 84:1535112021. View Article : Google Scholar : PubMed/NCBI

55 

Funakoshi-Tago M, Ohsawa K, Ishikawa T, Nakamura F, Ueda F, Narukawa Y, Kiuchi F, Tamura H, Tago K and Kasahara T: Inhibitory effects of flavonoids extracted from Nepalese propolis on the LPS signaling pathway. Int Immunopharmacol. 40:550–560. 2016. View Article : Google Scholar : PubMed/NCBI

56 

Gergen AK, Kohtz PD, Halpern AL, White AM, Meng X, Fullerton DA and Weyant MJ: Statins Inhibit Toll-Like Receptor 4-Mediated growth of human esophageal adenocarcinoma cells. J Surg Res. 260:436–447. 2021. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

February-2023
Volume 49 Issue 2

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Li M, Xiao Y, Liu P, Wei L, Zhang T, Xiang Z, Liu X, Zhang K, Zhong Q, Chen F, Chen F, et al: 4‑Methoxydalbergione inhibits esophageal carcinoma cell proliferation and migration by inactivating NF‑κB. Oncol Rep 49: 42, 2023
APA
Li, M., Xiao, Y., Liu, P., Wei, L., Zhang, T., Xiang, Z. ... Chen, F. (2023). 4‑Methoxydalbergione inhibits esophageal carcinoma cell proliferation and migration by inactivating NF‑κB. Oncology Reports, 49, 42. https://doi.org/10.3892/or.2023.8479
MLA
Li, M., Xiao, Y., Liu, P., Wei, L., Zhang, T., Xiang, Z., Liu, X., Zhang, K., Zhong, Q., Chen, F."4‑Methoxydalbergione inhibits esophageal carcinoma cell proliferation and migration by inactivating NF‑κB". Oncology Reports 49.2 (2023): 42.
Chicago
Li, M., Xiao, Y., Liu, P., Wei, L., Zhang, T., Xiang, Z., Liu, X., Zhang, K., Zhong, Q., Chen, F."4‑Methoxydalbergione inhibits esophageal carcinoma cell proliferation and migration by inactivating NF‑κB". Oncology Reports 49, no. 2 (2023): 42. https://doi.org/10.3892/or.2023.8479