Caffeic acid methyl ester inhibits LPS‑induced inflammatory response through Nrf2 activation and NF‑κB inhibition in human umbilical vein endothelial cells

  • Authors:
    • Jin-Young Park
    • Muhammad Yasir
    • Hee Jae Lee
    • Eun-Taek Han
    • Jin-Hee Han
    • Won Sun Park
    • Yong-Soo Kwon
    • Wanjoo Chun
  • View Affiliations

  • Published online on: October 17, 2023     https://doi.org/10.3892/etm.2023.12257
  • Article Number: 559
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Caffeic acid (CA) derivatives have been reported to exert anti‑inflammatory activities in various inflammatory conditions. However, the impact of CA methyl ester (CAME) on the inflammatory response in vascular endothelial cells has not been thoroughly elucidated. In the present study, the aim was to understand how CAME can reduce inflammation in human umbilical vein endothelial cells (HUVECs), which were challenged with lipopolysaccharide (LPS), and elucidate its mechanisms. CAME significantly attenuated LPS‑induced TNF‑α and IL‑1β release. Furthermore, CAME inhibited cyclooxygenase 2 expression and consequent secretion of prostaglandin E2. CAME also suppressed LPS‑stimulated inducible nitric oxide synthase expression. In addition, CAME significantly enhanced the expression of heme oxygenase‑1 (HO‑1) and nuclear factor erythroid‑derived 2‑related factor 2 (Nrf2) phosphorylation in the absence or presence of LPS stimulation in HUVECs. CAME also significantly suppressed LPS‑induced NF‑κB phosphorylation and inhibitor of κB phosphorylation and degradation. In conclusion, the present results provide clear evidence that CAME exerts its anti‑inflammatory activities by increasing HO‑1/Nrf2‑mediated cytoprotection and inhibiting NF‑κB‑mediated pro‑inflammatory pathways in HUVECs.

Introduction

Endothelial cells produce a variety of inflammatory agents that intensify endothelial dysfunction in inflammatory conditions (1). Inflammation-induced endothelial dysfunction has a crucial impact on the development of multiple vascular diseases, such as hypertension, atherosclerosis and vascular complications associated with diabetes (2,3). Previous research provides substantial evidence to suggest that endothelial cells may have a crucial role in the pathophysiology of sepsis, which is a serious inflammatory response throughout the body to infection triggered by an improper stimulation of the host's immune system by pathogenic elements (4). The pathogenic impact of endothelial activation and dysfunction in sepsis was clearly demonstrated by the unusual increase in cell adhesion molecules, which are involved in various processes related to the adhesion and coagulation of endothelial cells (5,6). This led to a surge in leukocyte migration, coagulation, vascular permeability and inflammation, all of which are typical characteristics of sepsis (7). Studies suggest that cell adhesion molecules and pro-inflammatory agents are enhanced through the activation of the NF-κB signaling pathway (8,9). Thus, mitigating abnormal vascular endothelial activation and the expression of pro-inflammatory mediators could offer substantial therapeutic value in treating inflammation-induced endothelial dysfunction.

Nuclear factor erythroid-derived 2-related factor 2 (Nrf2) is a crucial transcription factor necessary for the cellular response against a wide range of oxidative stressors. A study suggested that Nrf2 demonstrates cytoprotective effects in a sepsis model in mice (10). Caffeic acid (CA) phenethyl ester (CAPE) has been noticed for its role in modulating peripheral immune functions, which includes the expression of heme oxygenase (HO)-1 via the control of Nrf2, a primary transcription factor of HO-1(11). HO-1, induced by various oxidative stresses, performs a significant cytoprotective function against oxidative cell damage. The ability of lipopolysaccharide (LPS)-challenged macrophages to defend themselves from the aberrant overproduction of inflammatory mediators such as cyclooxygenase 2 (COX-2) by amplifying HO-1 expression has been reported (12). Furthermore, it has been observed that HO-1 attenuates the overproduction of cytokines such as TNF-α in LPS-challenged RAW264.7 cells (13). In addition, induction of HO-1 has been associated with increased survival rates in fatal endotoxemia (10). An animal study indicated that enhanced expression of HO-1 correlates with significant activation of the Nrf2 pathway (14).

Ester derivatives of CA are found abundantly in numerous natural plants. These include derivatives such as CA methyl ester (CAME), CAPE, CA isopropenyl ester and CA benzyl ester (15-17). Research indicates that they display an array of biological effects, including anti-oxidant, anti-inflammatory, anti-microbial, anti-tumor and anti-acetylcholinesterase activities (17-20). CAPE, for instance, is known to restrain cytokine-induced NF-κB signaling in macrophage cells (21), and plays a critical part in regulating the host's immune response (22). CAPE can also counteract allergic reactions by interfering with MAPK and NF-κB signaling in HMC-1 human mast cells that had been activated (23). CAME has been found to exhibit a variety of pharmacological benefits, such as anti-inflammatory and neuroprotective activities (24). Since CAME demonstrates extensive anti-inflammatory effects and CAPE exhibits anti-allergic characteristics, it is plausible to hypothesize that CAME could also display anti-inflammatory activity in endothelial cells. Therefore, the present study aims to explore the potential anti-inflammatory capabilities of CAME and the underlying mechanism in LPS-stimulated HUVECs, with the goal of identifying a possible therapeutic agent capable of alleviating diverse inflammatory vascular conditions.

Materials and methods

Reagents and cell culture

Escherichia coli serotype 055:B5's bacterial LPS was obtained from Sigma-Aldrich (Merck KGaA). CAME and CA were isolated and identified from the bark of Lonicera maackii as described previously (25). To determine whether Lonicera maackii is classified as an endangered species, a thorough search was conducted on Convention of International Trade in Endangered Species of Wild Fauna and Flora (CITES; https://cites.org/eng). The search confirmed that Lonicera maackii is not categorized as an endangered species. Furthermore, Lonicera maackii is commonly found in various Asian countries including Korea, Japan and China. To briefly explain the separation and identification process of CAME and CA, the dried bark of Lonicera maackii, which were collected in August 2014 from Samaksan (Korea) and identified by Dr Yong-Soo Kwon (College of Pharmacy, Kangwon National University, Chuncheon, Korea). The confirmed sample (no. KNUPH-S-14-01) is currently stored in the medicinal plant laboratory of the College of Pharmacy, Kangwon National University (Chuncheon, Korea). The plant material was dried (2.4 kg), cut into small pieces for use and extracted with 15 l methanol at room temperature. The methanolic extract (230 g) was then partitioned sequentially using hexane, chloroform and butanol. The obtained fractions were subjected to structural determination using NMR and mass spectrometry techniques. In addition, their purity was confirmed to be >95% through high-performance liquid chromatography analysis, which was performed using the Waters e2695 system, and the detector used was the Waters 2489 UV-vis detector (Waters Corp.), measuring at 260 nm. The isolated compounds were identified as CAME and CA by comparing their characteristics with those reported in the compound literature library. Lonicera maackii is a shrub of the Caprifoliaceae family, found in Korea, China, Japan and other regions. Its flower buds, leaves and bark are used in traditional medicine for colds and flu. It contains flavonoids, iridoids, caffeoyl quinic acids and phytosterols (25). CAME and CA were dissolved in DMSO and added to the culture media at the required concentrations, with 1 ppm of DMSO in the culture media. Human umbilical vein endothelial cells (HUVECs) were purchased from the American Type Culture Collection (cat. no. CRL-1730) and were cultured on a 2% gelatin-coated plate in M199 medium (Hyclone; Cytiva) (26) supplemented with 20% fetal bovine serum (Gibco; Thermo Fisher Scientific, Inc.), 3 ng/ml basic fibroblast growth factor (Gibco; Thermo Fisher Scientific, Inc.), 100 U/ml penicillin-streptomycin (Gibco; Thermo Fisher Scientific, Inc.) and 5 U/ml heparin (Sigma-Aldrich; Merck KGaA) (culture medium) in an incubator maintained at ~95% humidity, at a temperature of 37˚C with 5% CO2. Cells between passages two and six were used in the experiments. The cells were grown on 6-well plates (Corning; Merck KGaA) coated with gelatin and incubated in M199 medium containing 1% fetal bovine serum and 100 U/ml penicillin-streptomycin for 6 h, which is classified as the starvation medium (26). The cells were then incubated with designated concentrations of CAME and CA before being treated with LPS (1 µg/ml). In this study, the micromolar concentration range of CAME was used, as no significant cell toxicity was observed in previous studies (27).

Cytokine and prostaglandin (PG)E2 assays

HUVECs were incubated with CAME (1-100 µM) for 24 h and then stimulated in the absence or presence of LPS (1 µg/ml) for 24 h. ELISA kits were used to detect TNF-α (cat. no. MTA00B; R&D systems), IL-1β (cat. no. MLB00C; R&D systems) and PGE2 (cat. no. ADI-900-001; Enzo Life Sciences, Inc.) secreted into the culture media of HUVECs according to the manufacturer's instructions.

Western blot analysis

HUVECs were pretreated with CAME for 3 h before LPS stimulation. HUVECs were rinsed with ice-cold PBS and lysed in PRO-PREP lysis buffer (iNtRON Biotechnology, Inc.). Protein lysates (10 µg per lane) were separated by 10% SDS-PAGE. These proteins were then transferred to Hypond PVDF membranes (Amersham; Cytiva) and blocked for 1 h at room temperature in Tris-buffered saline containing Tween-20 (TBST) with 5% skimmed milk. Specific antibodies against inducible nitric oxide synthase (iNOS; 1:1,000 dilution; cat. no. 610329; BD Pharmingen; BD Biosciences), COX-2 (1:1,000 dilution; cat. no. 12282; Cell Signaling Technology, Inc.), HO-1 (1:1,000 dilution; cat. no. ab13243; abchem), Nrf2 (1:1,000 dilution; cat. no. bs-2013R; Bioss Inc.), phosphorylated (p)-Nrf2 (1:1,000 dilution; cat. no. ab76026; abchem), p65 (1:1,000 dilution; cat. no. 8242; Cell Signaling Technology, Inc.), p-p65 (1:1,000 dilution; cat. no. 3033; Cell Signaling Technology, Inc.), inhibitor of kB (IkB; 1:1,000 dilution; cat. no. 9242; Cell Signaling Technology, Inc.), p-IkB (1:1,000 dilution; cat. no. 2589; Cell Signaling Technology, Inc.) and β-actin (1:2,500 dilution; cat. no. A5441; Sigma-Aldrich; Merck KGaA) were diluted in TBST containing 5% milk. After being washed intensely with TBST, the following HRP-conjugated secondary antibodies were added and incubated overnight at room temperature: Anti-mouse IgG (1:1,000 dilution; cat. no. 7076; Cell Signaling Technology, Inc.) or Peroxidase AffiniPure goat anti-rabbit IgG (1:1,000 dilution; cat. no. 111-035-144; Jackson ImmunoResearch, Inc.). The blots were then developed and detected with the use of an enhanced chemiluminescence agent (cat. no. RPN3004; Amersham; Cytiva).

Statistical analysis

Values are expressed as the mean ± SD from three independent experiments. Results were statistically analyzed using SPSS 20.0 (IBM Corp.). To assess the differences between multiple groups, one-way analysis of variance was used, and Dunnett's multiple-comparison test was implemented. P<0.05 was considered to indicate a statistically significant difference.

Results

CAME inhibits TNF-α and IL-1β secretion in LPS-challenged HUVECs

Research studies have reported that inflammatory cytokines such as TNF-α and IL-1β have a vital influence on the progression of inflammation (28,29). Hence, the impact of CAME on the extracellular secretion of TNF-α and IL-1β was examined in LPS-challenged HUVECs. The cells were treated with CAME for 24 h prior to LPS treatment (1 µg/ml). LPS increased the secretion of TNF-α and IL-1β in HUVECs and CAME significantly suppressed the extracellular release of TNF-α and IL-1β in LPS-stimulated HUVECs in a concentration-dependent manner (Fig. 1). Of note, there was no evident cytotoxicity of CAME within the concentration ranges used (data not shown).

CAME inhibits LPS-induced PGE2 release and expression of COX-2 and iNOS

Given the previous report that increased levels of COX-2 are associated with inflammatory response leading to the production of pro-inflammatory mediator PGE2 (22), PGE2 secretion and COX-2 expression were examined in LPS-challenged HUVECs. LPS treatment resulted in increased secretion of PGE2 (Fig. 2). To compare the potency of CAME, CA was utilized as a reference compound. Of note, at the concentrations used in the present study, CA exerted no noticeable attenuation on the LPS-induced PGE2 secretion, whereas CAME showed a significant inhibition of LPS-induced PGE2 production (Fig. 2), suggesting that CAME may be more potent than CA. Furthermore, the expression level of COX-2 was examined. In accordance with PGE2 secretion, CAME significantly suppressed LPS-induced COX-2 expression in HUVECs (Fig. 3A). Quantitative analysis of COX-2 expression revealed considerable inhibition of COX-2 expression in a concentration-dependent manner (Fig. 3B). In addition, effects on iNOS, a pro-inflammatory protein related to endothelial inflammation, were examined. CAME significantly inhibited LPS-induced iNOS expression depending on the concentration of the compound in LPS-challenged HUVECs (Fig. 3), suggesting that CAME suppresses endothelial inflammation through inhibition of the expression of pro-inflammatory proteins such as COX-2 and iNOS in HUVECs.

CAME activates the HO-1/Nrf2 pathway in LPS-challenged HUVECs

HO-1, which is expressed through the transcription of Nrf2, has been reported to exert cytoprotective effects against a wide range of cellular stresses (30,31). Thus, in the present study, the effect of CAME on the expression of HO-1 was determined. In the absence of LPS challenge, CAME produced an increase in the expression of HO-1 depending on the concentration of the compound in HUVECs (Fig. 4). CAME resulted in increased expression of HO-1 in the presence of LPS challenge, suggesting that CAME exerts cytoprotective effects irrespective of the absence or presence of cellular stresses. Furthermore, the phosphorylation level of Nrf2, the transcription factor of HO-1, was also examined. In accordance with the level of HO-1, Nrf2 phosphorylation was increased with CAME in the absence or presence of LPS treatment (Fig. 5). These results strongly indicate that CAME may exert its cytoprotective activity through activation of the HO-1/Nrf2 pathway.

CAME inhibits NF-κB phosphorylation, and IκB phosphorylation and degradation

NF-κB is a pivotal transcription factor of pro-inflammatory genes in various inflammatory conditions (29). In the present study, the impact of CAME on LPS-induced activation of NF-κB was analyzed in LPS-challenged HUVECs. LPS treatment produced significantly increased phosphorylation of NF-κB a concentration-dependent manner in HUVECs (Fig. 6). Representative immunoblots showed increased phosphorylation of the p65 subunit and a gradual decrease with CAME (Fig. 6A), and quantitative analysis showed a significant decrease of p65 phosphorylation in a concentration-dependent manner (Fig. 6B). LPS-induced phosphorylation of IκB was significantly attenuated by CAME in a concentration-dependent manner (Fig. 6C). As IκB prevents the translocation of the phosphorylated p65 subunit of NF-κB into the nucleus (29), the total level of IκB was also examined. IκB levels were significantly attenuated with the LPS treatment in HUVECs and CAME suppressed IκB degradation in a dose-dependent manner (Fig. 6A and C). Presumably, sustained levels of IκB by CAME sequester NF-κB in the cytosol, preventing the NF-κB-mediated transcription of pro-inflammatory genes. Thus, CAME inhibited LPS-induced NF-κB activation through suppression of IκB degradation.

Discussion

The findings of the present study demonstrated that CAME is effective in reducing inflammatory responses triggered by LPS in vascular endothelial cells. CAME significantly reduced the levels of cytokines secreted, as well as the expression of COX-2 and iNOS in HUVECs challenged with LPS. CAME significantly activated the HO-1/Nrf2 pathway and suppressed the aberrantly activated NF-κB pathway in HUVECs.

CA and their derivatives are known to possess a diverse range of biological properties, such as antitumor, anti-inflammatory, antimicrobial, immunosuppressive and neuroprotective activities (32-34). In a previous study by our group, trihydroxycinnamic acid (THC), a derivative of CA, was shown to exert anti-inflammatory effects in LPS-challenged BV2 microglial cells (35). Our group also reported that THC leads to a reduction of LPS-induced inflammatory reactions in RAW264.7 macrophage cells and enhanced survival rates in mice in an LPS-induced endotoxemia model (9).

Pathologic endothelial activation has been observed in sepsis and numerous cardiovascular conditions, including atherosclerosis, hypertension and hemolysis (36,37). During the inflammatory response, vascular endothelial cells excessively produce a range of cytokines and mediators, thereby exacerbating the inflammatory injury (38). In the present study, the LPS-induced overproduction of cytokines, such as TNF-α and IL-1β, was significantly reduced with CAME treatment. The vascular endothelium acts as a barrier that selectively regulates the transfer of plasma and cells between the bloodstream and adjacent tissues (39). In a previous study by our group, it was observed that LPS-induced vascular leakage in multiple organs, encompassing the spleen, kidney and liver, was substantially curtailed by THC, a CA derivative, in a mouse model of sepsis (9). Furthermore, THC significantly reduced the infiltration of macrophages in the kidneys in an LPS-induced mouse model of sepsis (9). These results suggest that ester derivatives including CAME may also possess protective properties against vascular damage.

Nrf2 has been shown to have an essential role in maintaining cellular homeostasis and suppressing inflammatory conditions through the expression of cytoprotective enzymes and stress-responsive proteins (40,41). In experiments involving mice treated with sulforaphane, activation of Nrf2 and an enhancement of HO-1 were observed (42), while the absence of Nrf2 signaling led to increased vulnerability to inflammation (43). In addition, the upregulation of HO-1 contributed to a decrease in oxidative stress by eliminating free heme and simultaneously increasing the level of anti-inflammatory substances (44). Studies have indicated that the activation of the HO-1/Nrf2 signaling pathway may be facilitated via a variety of signaling pathways (9,10,35). CAPE has been reported to activate the Nrf2 pathway in colonic inflammation (45) and osteoarthritis progression (46). In the present study, treatment with CAME resulted in increased levels of phosphorylated Nrf2, which in turn induced the increased expression of HO-1 in HUVECs in the absence or presence of LPS stimulation. These results imply that the HO-1/Nrf2 signaling pathway may be induced with activators such as CAME, irrespective of the presence of cellular stressors. Furthermore, the HO-1/Nrf2 pathway may also be activated as a defense mechanism against cellular stressors. This suggests that activating the HO-1/Nrf2 pathway may prepare cells for the potential stressor under normal conditions and also provide a defense against damage under stress conditions. Given the cytoprotective role of HO-1, the pharmacological induction of HO-1 expression with CAME may serve as a promising therapeutic approach in combating sepsis and inflammatory vascular conditions. However, further studies CA derivatives activate Nrf2 phosphorylation.

NF-κB is the key transcription factor responsible for inflammatory responses (35). It is widely recognized that NF-κB is implicated in the generation of pro-inflammatory cytokines such as IL-1β and TNF-α, as well as proteins such as COX-2 and iNOS (47,48). LPS has been known to activate the IκB kinase (IKK) complex, composed of two catalytic subunits (IKKα and IKKβ) and a regulatory subunit (NEMO/IKKγ). Once activated, IKK phosphorylates the IκB proteins, targeting ubiquitination and subsequent proteasomal degradation. This allows the NF-κB dimers (p50/p65) to translocate to the nucleus and induce the transcription of target genes. Given the fact that inhibition of IKK can prevent the activation and nuclear translocation of NF-κB, IKK inhibitors are potential drugs to suppress the production of cytokines and other inflammatory mediates, ultimately reducing inflammation in tissue damage in various disorders, such as rheumatoid arthritis, inflammatory bowel disease and psoriasis (49,50). Previously, it was reported that CA regulates the NF-κB signaling pathway by acting on the IKK pathway (51). In the present study, CAME significantly suppressed LPS-induced IκB degradation, suggesting a possibility that CAME may directly interfere with the metabolizing action of IKK. Therefore, further studies assessing the involvement of IKK by determining its phosphorylation level and investigating nuclear translocation of NF-κB through a nuclear fractionation assay may offer a more comprehensive understanding of the complete role of the NF-κB pathway.

Of note, the present study had a limitation. CAME, used in the present study, was derived from Lonicera maackii and had a purity of >95%. However, it contained certain impurities. There is a chance that these impurities may have influenced the study's anti-inflammatory results. To address this limitation, future studies should compare the effects with CAME commercially purified to 100% to validate the findings of the present study. In addition to CAME, various other compounds, such as 5-caffeoylquinic acid n-butyl ester, methyl 3,4-dicaffeoyl quinate, 3,5-dicaffeoyl quinic acid n-butyl ester, loganin and CA have been isolated from Lonicera maackii. While the current study investigated the anti-inflammatory properties of CAME, it does not offer a comprehensive understanding of the biological effects of Lonicera maackii. Consequently, in-depth investigations into each individual compound, as well as the whole extract, are crucial for an extensive understanding of the biological properties of Lonicera maackii.

In conclusion, the findings of the present study provide clear evidence that CAME effectively suppresses LPS-triggered inflammatory responses by activating the cytoprotective HO-1/Nrf2 pathway and inhibiting pro-inflammatory NF-κB signaling in LPS-challenged HUVECs. CAME can activate HO-1/Nrf2 pathway under normal conditions to preemptively prepare cells for potential stressors and also provide protection against stressors in inflammatory conditions. The results clearly suggest that CAME is a promising therapeutic agent having dual beneficial properties against inflammatory vascular disorders.

Acknowledgements

Not applicable.

Funding

Funding: This work was supported by the National Research Foundation of Korea grant funded by the Korea government (grant no. 2021-R1A4A1031574) and the Korea Basic Science Institute (National Research Facilities and Equipment Center) grant funded by the Ministry of Education (grant no. 2022R1A6C101A739).

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

JYP and MY conducted the experiments and analyzed the data. ETH, WSP and JHH performed data analysis for the study. YSK isolated and identified CAME and CA from Lonicera maackii. HJL performed the statistical analysis of the results. WC conceptualized the study and wrote, reviewed and edited the manuscript. YSK and WC confirm the authenticity of all the raw data. All authors have read and approved the final manuscript.

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.

References

1 

Yisireyili M, Hayashi M, Wu H, Uchida Y, Yamamoto K, Kikuchi R, Shoaib Hamrah M, Nakayama T, Wu Cheng X, Matsushita T, et al: Xanthine oxidase inhibition by febuxostat attenuates stress-induced hyperuricemia, glucose dysmetabolism, and prothrombotic state in mice. Sci Rep. 7(1266)2017.PubMed/NCBI View Article : Google Scholar

2 

Gray SP and Jandeleit-Dahm KAM: The role of NADPH oxidase in vascular disease-hypertension, atherosclerosis & stroke. Curr Pharm Des. 21:5933–5944. 2015.PubMed/NCBI View Article : Google Scholar

3 

Han JM, Li H, Cho MH, Baek SH, Lee CH, Park HY and Jeong TS: Soy-leaf extract exerts atheroprotective effects via modulation of Krüppel-like factor 2 and adhesion molecules. Int J Mol Sci. 18(373)2017.PubMed/NCBI View Article : Google Scholar

4 

Kang JS, Jeon YJ, Park SK, Yang KH and Kim HM: Protection against lipopolysaccharide-induced sepsis and inhibition of interleukin-1beta and prostaglandin E2 synthesis by silymarin. Biochem Pharmacol. 67:175–181. 2004.PubMed/NCBI View Article : Google Scholar

5 

Reinhart K, Bayer O, Brunkhorst F and Meisner M: Markers of endothelial damage in organ dysfunction and sepsis. Crit Care Med. 30 (5 Suppl):S302–S312. 2002.PubMed/NCBI View Article : Google Scholar

6 

Yano K, Liaw PC, Mullington JM, Shih SC, Okada H, Bodyak N, Kang PM, Toltl L, Belikoff B, Buras J, et al: Vascular endothelial growth factor is an important determinant of sepsis morbidity and mortality. J Exp Med. 203:1447–1458. 2006.PubMed/NCBI View Article : Google Scholar

7 

Shapiro NI, Khankin EV, Van Meurs M, Shih SC, Lu S, Yano M, Castro PR, Maratos-Flier E, Parikh SM, Karumanchi SA and Yano K: Leptin exacerbates sepsis-mediated morbidity and mortality. J Immunol. 185:517–524. 2010.PubMed/NCBI View Article : Google Scholar

8 

Lee HJ, Lim HJ, Lee DY, Jung H, Kim MR, Moon DC, Kim KI, Lee MS and Ryu JH: Carabrol suppresses LPS-induced nitric oxide synthase expression by inactivation of p38 and JNK via inhibition of I-kappaBalpha degradation in RAW 264.7 cells. Biochem Biophys Res Commun. 391:1400–1404. 2010.PubMed/NCBI View Article : Google Scholar

9 

Lee JW, Kwon JH, Lim MS, Lee HJ, Kim SS, Lim SY and Chun W: 3,4,5-Trihydroxycinnamic acid increases heme-oxygenase-1 (HO-1) and decreases macrophage infiltration in LPS-induced septic kidney. Mol Cell Biochem. 397:109–116. 2014.PubMed/NCBI View Article : Google Scholar

10 

Ha YM, Ham SA, Kim YM, Lee YS, Kim HJ, Seo HG, Lee JH, Park MK and Chang KC: β1-adrenergic receptor-mediated HO-1 induction, via PI3K and p38 MAPK, by isoproterenol in RAW 264.7 cells leads to inhibition of HMGB1 release in LPS-activated RAW 264.7 cells and increases in survival rate of CLP-induced septic mice. Biochem Pharmacol. 82:769–777. 2011.PubMed/NCBI View Article : Google Scholar

11 

Fidan H, Sahin O, Yavuz Y, Kilbas A, Cetinkaya Z, Ela Y, Ozen OA and Altuntas I: Caffeic acid phenethyl ester reduces mortality and sepsis-induced lung injury in rats. Crit Care Med. 35:2822–2829. 2007.PubMed/NCBI View Article : Google Scholar

12 

Park SY, Seetharaman R, Ko MJ, Kim DY, Kim TH, Yoon MK, Kwak JH, Lee SJ, Bae YS and Choi YW: Ethyl linoleate from garlic attenuates lipopolysaccharide-induced pro-inflammatory cytokine production by inducing heme oxygenase-1 in RAW264.7 cells. Int Immunopharmacol. 19:253–261. 2014.PubMed/NCBI View Article : Google Scholar

13 

Tsoyi K, Lee TY, Lee YS, Kim HJ, Seo HG, Lee JH and Chang KC: Heme-oxygenase-1 induction and carbon monoxide-releasing molecule inhibit lipopolysaccharide (LPS)-induced high-mobility group box 1 release in vitro and improve survival of mice in LPS- and cecal ligation and puncture-induced sepsis model in vivo. Mol Pharmacol. 76:173–182. 2009.PubMed/NCBI View Article : Google Scholar

14 

Park EJ, Lim JH, Nam SI, Park JW and Kwon TK: Rottlerin induces heme oxygenase-1 (HO-1) up-regulation through reactive oxygen species (ROS) dependent and PKC delta-independent pathway in human colon cancer HT29 cells. Biochim. 92:110–115. 2010.PubMed/NCBI View Article : Google Scholar

15 

Clifford MN: Chlorogenic acids and other cinnamates-nature, occurrence and dietary burden†. J Sci Food Agric. 79:362–372. 1999.

16 

Macheix JJ, Fleuriet A and Billot J: Fruit phenolics. CRC Press, FL,. 113:pp41–43. 1990.

17 

Murtaza G, Karim S, Akram MR, Khan SA, Azhar S, Mumtaz A and Bin Asad MH: Caffeic acid phenethyl ester and therapeutic potentials. Biomed Res Int. 2014(145342)2014.PubMed/NCBI View Article : Google Scholar

18 

Bose JS, Gangan V, Jain SK and Manna SK: Novel caffeic acid ester derivative induces apoptosis by expressing FasL and downregulating NF-KappaB: Potentiation of cell death mediated by chemotherapeutic agents. J Cell Physiol. 218:653–662. 2009.PubMed/NCBI View Article : Google Scholar

19 

Collins W, Lowen N and Blake DJ: Caffeic acid esters are effective bactericidal compounds against paenibacilluslarvae by altering intracellular oxidant and antioxidant levels. Biomolecules. 9(312)2019.PubMed/NCBI View Article : Google Scholar

20 

Gießel JM, Loesche A, Csuk R and Serbian I: Caffeic acid phenethyl ester (CAPE)-derivatives act as selective inhibitors of acetylcholinesterase. Eur J Med Chem. 177:259–268. 2019.PubMed/NCBI View Article : Google Scholar

21 

Schoonbroodt S, Legrand-Poels S, Best-Belpomme M and Piette J: Activation of the NF-kappaB transcription factor in a T-lymphocytic cell line by hypochlorous acid. Biochem J. 321:777–785. 1997.PubMed/NCBI View Article : Google Scholar

22 

Zhou Y, Yang Q, Xu H, Zhang J, Deng H, Gao H, Yang J, Zhao D and Liu F: miRNA-221-3p enhances the secretion of interleukin-4 in mast cells through the phosphatase and tensin homolog/p38/nuclear factor-kappaB pathway. PLoS One. 11(e0148821)2016.PubMed/NCBI View Article : Google Scholar

23 

Cho MS, Park WS, Jung WK, Qian ZJ, Lee DS, Choi JS, Lee DY, Park SG, Seo SK, Kim HJ, et al: Caffeic acid phenethyl ester promotes anti-inflammatory effects by inhibiting MAPK and NF-κB signaling in activated HMC-1 human mast cells. Pharm Biol. 52:926–932. 2014.PubMed/NCBI View Article : Google Scholar

24 

Shin KM, Kim IT, Park YM, Ha J, Choi JW, Park HJ, Lee YS and Lee KT: Anti-inflammatory effect of caffeic acid methyl ester and its mode of action through the inhibition of prostaglandin E2, nitric oxide and tumor necrosis factor-alpha production. Biochem Pharmacol. 68:2327–2336. 2004.PubMed/NCBI View Article : Google Scholar

25 

An GY, Chung SW, Cho HC, Park JR, Kim MJ, Kim HP, Yang HJ, Chun W and Kwon Y: Phytochemical constituents of Lonicera maackii stems. Korean J Pharmacogn. 49:103–107. 2018.

26 

Kim DY, Park JA, Kim Y, Noh M, Park S, Lie E, Kim E, Kim YM and Kwon YG: SALM4 regulates angiogenic functions in endothelial cells through VEGFR2 phosphorylation at Tyr1175. FASEB J. 33:9842–9857. 2019.PubMed/NCBI View Article : Google Scholar

27 

Park JY, Lee HJ, Han ET, Han JH, Park WS, Kwon YS and Chun W: Caffeic acid methyl ester inhibits mast cell activation through the suppresion of MAPKs and NF-κB signaling in RBL-2H3 cells. Heliyon. 9(e16529)2023.PubMed/NCBI View Article : Google Scholar

28 

Hu Frisk JM, Kjellén L, Melo FR, Öhrvik H and Pejler G: Mitogen-activated protein kinase signaling regulates proteoglycan composition of mast cell secretory granules. Front Immunol. 9(1670)2018.PubMed/NCBI View Article : Google Scholar

29 

Rehman MU, Yoshihisa Y, Miyamoto Y and Shimizu T: The anti-inflammatory effects of platinum nanoparticles on the lipopolysaccharide-induced inflammatory response in RAW 264.7 macrophages. Inflamm Res. 61:1177–1185. 2012.PubMed/NCBI View Article : Google Scholar

30 

Bhatt NP, Park JY, Lee HJ, Kim SS, Kwon YS and Chun W: Apocynin protects mesangial cells from lipopolysaccharide-induced inflammation by exerting heme oxygenase 1-mediated monocyte chemoattractant protein-1 suppression. Int J Mol Med. 40:1294–1301. 2017.PubMed/NCBI View Article : Google Scholar

31 

Subedi L, Lee JH, Yumnam S, Ji E and Kim SY: Anti-inflammatory effect of sulforaphane on LPS-activated microglia potentially through JNK/AP-1/NF-κB inhibition and Nrf2/HO-1 activation. Cells. 8(194)2019.PubMed/NCBI View Article : Google Scholar

32 

Nagasaka R, Chotimarkorn C, Shafiqul IM, Hori M, Ozaki H and Ushio H: Anti-inflammatory effects of hydroxycinnamic acid derivatives. Biochem Biophys Res Commun. 358:615–619. 2007.PubMed/NCBI View Article : Google Scholar

33 

Kim YC: Neuroprotective phenolics in medicinal plants. Arch Pharm Res. 33:1611–1632. 2010.PubMed/NCBI View Article : Google Scholar

34 

Lee JW, Cheong IY, Kim HS, Lee JJ, Lee YS, Kwon YS, Kim MJ, Lee HJ, Kim SS and Chun W: Anti-inflammatory activity of 1-docosanoyl cafferate isolated from rhus verniciflua in LPS-stimulated BV2 microglial cells. Korean J Physiol Pharmacol. 15:9–15. 2011.PubMed/NCBI View Article : Google Scholar

35 

Lee JW, Bae CJ, Choi YJ, Kim SI, Kim NH, Lee HJ, Kim SS, Kwon YS and Chun W: 3,4,5-Trihydroxycinnamic acid inhibits LPS-induced iNOS expression by suppressing NF-κB activation in BV2 microglial cells. Korean J Physiol Pharmacol. 16:107–112. 2012.PubMed/NCBI View Article : Google Scholar

36 

Ross R: Atherosclerosis-an inflammatory disease. N Engl J Med. 340:115–126. 1999.PubMed/NCBI View Article : Google Scholar

37 

Bains SK, Foresti R, Howard J, Atwal S, Green CJ and Motterlini R: Human sickle cell blood modulates endothelial heme oxygenase activity: Effects on vascular adhesion and reactivity. Arterioscler Thromb Vasc Biol. 30:305–312. 2010.PubMed/NCBI View Article : Google Scholar

38 

Liu XH, Pan LL, Yang HB, Gong QH and Zhu YZ: Leonurine attenuates lipopolysaccharide-induced inflammatory responses in human endothelial cells: Involvement of reactive oxygen species and NF-κB pathways. Eur J Pharmacol. 680:108–114. 2012.PubMed/NCBI View Article : Google Scholar

39 

Fisher M: Injuries to the vascular endothelium: Vascular wall and endothelial dysfunction. Rev Neurol Dis. 5 (Suppl 1):S4–S11. 2008.PubMed/NCBI

40 

Lee IS, Lim J, Gal J, Kang JC, Kim HJ, Kang BY and Choi HJ: Anti-inflammatory activity of xanthohumol involves heme oxygenase-1 induction via NRF2-ARE signaling in microglial BV2 cells. Neurochem Int. 58:153–160. 2011.PubMed/NCBI View Article : Google Scholar

41 

Surh YJ, Kundu JK and Na HK: Nrf2 as a master redox switch in turning on the cellular signaling involved in the induction of cytoprotective genes by some chemopreventive phytochemicals. Planta Med. 74:1526–1539. 2008.PubMed/NCBI View Article : Google Scholar

42 

Lin W, Wu RT, Wu T, Khor TO, Wang H and Kong AN: Sulforaphane suppressed LPS-induced inflammation in mouse peritoneal macrophages through Nrf2 dependent pathway. Biochem Pharmacol. 76:967–973. 2008.PubMed/NCBI View Article : Google Scholar

43 

Yates MS, Tran QT, Dolan PM, Osburn WO, Shin S, McCulloch CC, Silkworth JB, Taguchi K, Yamamoto M, Williams CR, et al: Genetic versus chemoprotective activation of Nrf2 signaling: Overlapping yet distinct gene expression profiles between Keap1 knockout and triterpenoid-treated mice. Carcinogenesis. 30:1024–1031. 2009.PubMed/NCBI View Article : Google Scholar

44 

Kawakami T, Takahashi T, Shimizu H, Nakahira K, Takeuchi M, Katayama H, Yokoyama M, Morita K, Akagi R and Sassa S: Highly liver-specific heme oxygenase-1 induction by interleukin-11 prevents carbon tetrachloride-induced hepatotoxicity. Int J Mol Med. 18:537–546. 2006.PubMed/NCBI

45 

Kim H, Kim W, Yum S, Hong S, Oh JE, Lee JW, Kwak MK, Park EJ, Na DH and Jung Y: Caffeic acid phenethyl ester activation of Nrf2 pathway is enhanced under oxidative state: Structural analysis and potential as a pathologically targeted therapeutic agent in treatment of colonic inflammation. Free Radic Biol Med. 65:552–562. 2013.PubMed/NCBI View Article : Google Scholar

46 

Sun W, Xie W, Huang D, Cui Y, Yue J, He Q, Jiang L, Xiong J, Sun W and Yi Q: Caffeic acid phenethyl ester attenuates osteoarthritis progression by activating NRF2/HO-1 and inhibiting the NF-κB signaling pathway. Int J Mol Med. 50(134)2022.PubMed/NCBI View Article : Google Scholar

47 

Wan M, Liu J and Ouyang X: Nucleotide-binding oligomerization domain 1 regulates Porphyromonas gingivalis-induced vascular cell adhesion molecule 1 and intercellular adhesion molecule 1 expression in endothelial cells through NF-κB pathway. J Periodontal Res. 50:189–196. 2015.PubMed/NCBI View Article : Google Scholar

48 

Wang L, Xu Y, Yu Q, Sun Q, Xu Y, Gu Q and Xu X: H-RN, a novel antiangiogenic peptide derived from hepatocyte growth factor inhibits inflammation in vitro and in vivo through PI3K/AKT/IKK/NF-κB signal pathway. Biochem Pharmacol. 89:255–265. 2014.PubMed/NCBI View Article : Google Scholar

49 

Antonia RJ, Hagan RS and Baldwin AS: Expanding the view of IKK: New substrates and new biology. Trends Cell Biol. 31:166–178. 2021.PubMed/NCBI View Article : Google Scholar

50 

Mulero MC, Huxford T and Ghosh G: NF-κB, IκB, and IKK: Integral components of immune system signaling. Adv Exp Med Biol. 1172:207–226. 2019.PubMed/NCBI View Article : Google Scholar

51 

Kim SR, Jung YR, Kim DH, An HJ, Kim MK, Kim ND and Chung HY: Caffeic acid regulates LPS-induced NF-κB activation through NIK/IKK and c-Src/ERK signaling pathways in endothelial cells. Arch Pharm Res. 37:539–547. 2014.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

December-2023
Volume 26 Issue 6

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Park J, Yasir M, Lee HJ, Han E, Han J, Park WS, Kwon Y and Chun W: Caffeic acid methyl ester inhibits LPS‑induced inflammatory response through Nrf2 activation and NF‑κB inhibition in human umbilical vein endothelial cells. Exp Ther Med 26: 559, 2023
APA
Park, J., Yasir, M., Lee, H.J., Han, E., Han, J., Park, W.S. ... Chun, W. (2023). Caffeic acid methyl ester inhibits LPS‑induced inflammatory response through Nrf2 activation and NF‑κB inhibition in human umbilical vein endothelial cells. Experimental and Therapeutic Medicine, 26, 559. https://doi.org/10.3892/etm.2023.12257
MLA
Park, J., Yasir, M., Lee, H. J., Han, E., Han, J., Park, W. S., Kwon, Y., Chun, W."Caffeic acid methyl ester inhibits LPS‑induced inflammatory response through Nrf2 activation and NF‑κB inhibition in human umbilical vein endothelial cells". Experimental and Therapeutic Medicine 26.6 (2023): 559.
Chicago
Park, J., Yasir, M., Lee, H. J., Han, E., Han, J., Park, W. S., Kwon, Y., Chun, W."Caffeic acid methyl ester inhibits LPS‑induced inflammatory response through Nrf2 activation and NF‑κB inhibition in human umbilical vein endothelial cells". Experimental and Therapeutic Medicine 26, no. 6 (2023): 559. https://doi.org/10.3892/etm.2023.12257