Skip to main content
Erschienen in: BMC Complementary Medicine and Therapies 1/2017

Open Access 01.12.2017 | Research article

Anti-inflammatory evaluation of the methanolic extract of Taraxacum officinale in LPS-stimulated human umbilical vein endothelial cells

verfasst von: Daun Jeon, Seok Joong Kim, Hong Seok Kim

Erschienen in: BMC Complementary Medicine and Therapies | Ausgabe 1/2017

Abstract

Background

Atherosclerosis is a chronic vascular inflammatory disease. Since even low-level endotoxemia constitutes a powerful and independent risk factor for the development of atherosclerosis, it is important to find therapies directed against the vascular effects of endotoxin to prevent atherosclerosis. Taraxacum officinale (TO) is used for medicinal purposes because of its choleretic, diuretic, antioxidative, anti-inflammatory, and anti-carcinogenic properties, but its anti-inflammatory effect on endothelial cells has not been established.

Methods

We evaluated the anti-inflammatory activity of TO filtered methanol extracts in LPS-stimulated human umbilical vein endothelial cells (HUVECs) by monocyte adhesion and western blot assays. HUVECs were pretreated with 100 μg/ml TO for 1 h and then incubated with 1 μg/ml LPS for 24 h. The mRNA and protein expression levels of the targets (pro-inflammatory cytokines and adhesion molecules) were analyzed by real-time PCR and western blot assays. We also preformed HPLC analysis to identify the components of the TO methanol extract.

Results

The TO filtered methanol extracts dramatically inhibited LPS-induced endothelial cell–monocyte interactions by reducing vascular cell adhesion molecule-1 and monocyte chemoattractant protein-1, and pro-inflammatory cytokine expression. TO suppressed the LPS-induced nuclear translocation of NF-κB, whereas it did not affect MAPK activation.

Conclusions

Our findings demonstrated that methanol extracts of TO could attenuate LPS-induced endothelial cell activation by inhibiting the NF-κB pathway. These results indicate the potential clinical benefits and applications of TO for the prevention of vascular inflammation and atherosclerosis.
Hinweise

Electronic supplementary material

The online version of this article (https://​doi.​org/​10.​1186/​s12906-017-2022-7) contains supplementary material, which is available to authorized users.
Abkürzungen
DMSO
Dimethyl sulfoxide
HPLC
High-performance liquid chromatography
HUVEC
Human umbilical vein endothelial cell
IκBα
Inhibitor of NF-κB alpha
ICAM-1
Intercellular adhesion molecule-1
IL-1
Interleukin-1
LPS
Lipopolysaccharide
MAPK
Mitogen-activated protein kinase
MCP-1
Monocyte chemoattractant protein-1
NF-κB
Nuclear factor-kappa B
TNF-α
Tumor necrosis factor-alpha
TO
Taraxacum officinale
VCAM-1
Vascular cell adhesion molecule-1

Background

Insults to the vasculature can cause a wide range of life-threatening diseases, including stroke, myocardial infarction, hypertension, and chronic kidney disease. Inflammation is emerging as a key contributor to many vascular diseases, and furthermore plays a major role in autoimmune diseases, arthritis, allergic reactions, and cancer.
Besides autoimmune disorders related to vascular inflammation, a more common chronic vascular inflammatory disease is atherosclerosis [1, 2]. Clinical evidence indicates that atherosclerosis is accelerated in patients suffering from large and medium-sized vessel vasculitis [3].
Endotoxemia, the presence of endotoxin in the blood, constitutes a strong risk factor for early atherogenesis in subjects with chronic or recurrent bacterial infections [4]. Even low-level endotoxemia (as low as 50 pg/ml) can be a powerful and independent risk factor for the development of atherosclerosis [5] through multiple mechanisms, including increases in reactive oxygen species, chemotactic and pro-inflammatory cytokines, and adhesion molecules. Therefore, it is important to find therapies directed against the vascular effects of endotoxin to prevent atherosclerosis in humans.
Most therapy regimens for treating atherosclerosis aim at modulating hypertension and hyperlipidemia or controlling hemostasis in order to avoid thrombotic complications [6]. However, many of these modalities often neglect the role of inflammation in atherosclerosis [6]. As these therapies remain insufficient in reducing the burden of atherosclerosis-related mortality, other approaches like complementary and alternative medicine have recently started to gain more attention [7, 8].
Taraxacum officinale (TO), commonly known as dandelion, is used for medicinal purposes because of its choleretic, diuretic, antioxidative, anti-inflammatory, and anti-carcinogenic properties [9, 10]. The anti-inflammatory effects of extracts of TO or its single components have been reported in both in vitro and animal models. TO extracts (100 and 1000 μg/ml) were demonstrated to inhibit lipopolysaccharide (LPS)-induced tumor necrosis factor-alpha (TNF-α) production in rat astrocytes by inhibiting interleukin-1 (IL-1) production [11]. Luteolin and luteolin-7-O-glucoside, two active components from TO flower extracts, significantly suppressed the production of both inducible nitric oxide synthase and cyclooxygenase-2 in LPS-activated mouse macrophage RAW264.7 cells [12]. Pretreatment with TO extracts also protected against LPS-induced acute lung injury in mice [13]. However, the effect of TO on endothelial activation has not been established.
Our study shows that TO reduces the expression of vascular cell adhesion molecule-1 (VCAM-1) and pro-inflammatory cytokines in endothelial cells, and decreases mononuclear cell adhesion by suppressing nuclear factor-kappa B (NF-κB) signaling.

Methods

Reagents

LPS, dimethyl sulfoxide (DMSO), and Hoechst 33,258 were purchased from Sigma-Aldrich (St. Louis, MO, USA). Calcein AM was obtained from Thermo Fisher Scientific (Waltham, MA, USA). Antibodies against VCAM-1, intercellular adhesion molecule-1 (ICAM-1), β-actin, NF-κB p65, inhibitor of NF-κB alpha (IκBα), and phospho-IκBα were obtained from Cell Signaling Technology (Danvers, MA, USA).

Methanol extracts of Taraxacum officinale

Methanol extracts (code numbers: PB5027.2) from whole plants of TO were purchased from the Plant Extract Bank at the Korea Research Institute of Bioscience and Biotechnology (Daejeon, Republic of Korea; http://​extract.​kribb.​re.​kr; E-mail: plantext@kribb.re.kr) [14, 15]. The extracts were dissolved in DMSO at a concentration of 100 mg/ml.

Cell culture

Human umbilical vein endothelial cells (HUVECs) were obtained from ScienCell Research Laboratories (San Diego, CA, USA) [16]. The cells were cultured in endothelial cell medium (ScienCell Research Laboratories) containing 5% (v/v) fetal bovine serum, at 37 °C under an atmosphere with 5% (v/v) CO2 and 95% humidity.

Cytotoxicity assay

Cell viability was assessed by using the CellTilter 96 Aqueous One Solution Cell Proliferation Assay (Promega Corporation, Madison, WI, USA), according to the manufacturer’s instructions. Cells were seeded at a density of 1 × 104 cells/well into 96-well plates. After their subjection to different treatments, the cells were incubated with 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H–tetrazolium, inner salt (MTS) solution at a final concentration of 0.4 mg/ml for 4 h at 37 °C. One Solution Reagent was then added directly to the culture wells, and the plates were incubated for 4 h, following which the absorbance at 490 nm was recorded with a 96-well plate reader [17, 18]. The absorbance was measured at 490 nm with a Multiskan GO microplate spectrophotometer (Thermo Fisher Scientific).

Western blot analysis

Cells were washed with ice-cold PBS and lysed on ice in RIPA lysis buffer (50 mM Tris-HCl (pH 7.5), 150 mM NaCl, 1% Nonidet P-40, 0.1% sodium dodecyl sulfate, and 0.5% sodium deoxycholate) supplemented with protease and phosphatase inhibitors. Aliquots with equal amounts of protein were loaded and separated on a SDS-PAGE gel. The proteins on the gel were then transferred to a nitrocellulose membrane (Bio-Rad, Hercules, CA, USA) and probed using specific antibodies as indicated. The bands were detected by chemiluminescence on a ChemiDoc imaging system (Bio-Rad). To control for sample loading, the blots were subsequently stripped and re-probed for total IκBα or β-actin.

RNA extraction and real-time reverse-transcription polymerase chain reaction

Total RNA was isolated from the cells, using the NucleoSpin RNA Plus Kit (MACHEREY-NAGEL, Düren, Germany), and 1 μg was then used for cDNA synthesis with the iScript cDNA Synthesis Kit (Bio-Rad). The resulting cDNA was PCR amplified with the appropriate primer pairs: MCP-1, 5′-AGAATCACCAGCAGCAAGTGTCC-3′ (forward) and 5′-TCCTGAACCCACTTCTGCTTGG-3′ (reverse); TNF-α, 5′-CTCTTCTGCCTGCTGCACTTTG-3′ (forward) and 5′-ATGGGCTACAGGCTTGTCACTC-3′ (reverse); IL-1β, 5′- CCACAGACCTTCCAGGAGAATG-3′ (forward) and 5′-GTGCAGTTCAGTGATCGTACAGG-3′ (reverse); IL-6, 5′-AGACAGCCACTCACCTCTTCAG-3′ (forward) and 5′- TTCTGCCAGTGCCTCTTTGCTG-3′ (reverse); and 18S rRNA, 5′-AACCCGTTGAACCCCATT-3′ (forward) and 5′-CCATCCAATCGGTAGTAGCG-3′ (reverse) (Bioneer, Daejeon, Republic of Korea). The RT-qPCR was performed and analyzed with a CFX Connect Real-Time PCR detection system (Bio-Rad), and the gene expression levels were normalized to that of 18S rRNA as the housekeeping gene [19].

Analysis of monocyte adhesion to HUVECs

HUVECs were seeded and incubated in 6-well plates until they reached >85% confluence. Subsequently, the cells were pre-incubated with the TO extract (100 μg/ml) for 1 h prior to stimulation with LPS (1 μg/ml) for 24 h. Human monocytic THP-1 cells were labeled with 5 μM Calcein AM for 30 min in RPMI-1640 medium and then added to the HUVEC-containing 6-well plates and incubated for 1 h. Subsequently, unbound monocytes were removed by 3 washes with warm phosphate-buffered saline (PBS) [20]. Bound monocytes were determined using a fluorescence microscope (EVOS FL Cell Imaging System, Thermo Fisher Scientific).

Immunofluorescence analysis of NF-κB p65 nuclear translocation

HUVECs were pre-incubated with the TO extract (100 μg/ml) for 1 h, followed by LPS (1 μg/ml) for 1 h. The cells were then fixed with 4% paraformaldehyde in PBS (pH 7.4) for 20 min at room temperature, and permeabilized with 0.1% Triton X-100. Nonspecific binding was blocked with 5% normal goat serum for 1 h. Then, the cells were incubated overnight with NF-κB p65 antibody at 4 °C, followed by incubation with Alexa Fluor 594-conjugated goat anti-rabbit IgG (Thermo Fisher Scientific) for 1 h at room temperature. Nuclei were stained with Hoechst 33,258. The locations of NF-κB p65 and nuclei were determined using a fluorescence microscope (EVOS FL Cell Imaging System). Results were expressed as the percentage of NF-κB p65-positive cells in the total cells counted from 4 randomly chosen high-power (×20) fields in each well. Each assay was performed in triplicate.

High-performance liquid chromatography analysis

The TO methanol extracts were filtered (Millipore 0.45 μm) and 5 μl was injected into a high-performance liquid chromatography (HPLC) column (Supelcosil LC-18, 25 cm × 4.6 mm × 5 μm), at 25 °C. The extract separation was carried out on an HPLC device composed of Waters 510 pumps, 2489 UV/Vis detector, gradient controller, and Rheodyne injector, with a solvent A and B mixture as follows: methanol:acetic acid:water (10:2:88; solvent A) and methanol:acetic acid:water (90:3:7; solvent B) at a flow of 1 ml/min, with detection at 280 nm. The solvent A/B gradient applied was as follows: A from 100% to 85% (minutes 0–10), A from 85% to 50% (minutes 10–30), A from 50% to 15% (minutes 30–45), and A from 15% to 100% (minutes 45–55) [21]. The identification of peaks was made by comparison with HPLC chromatograms of individual pure phenolic acid (gallic, protocatechuic, chlorogenic, caffeic, p-coumaric, and ferulic acids) standards procured from Sigma-Aldrich.

Statistics

Data were analyzed by analysis of variance (Sigma Stat 12.0) and tested for use of parametric or nonparametric post hoc analysis. Multiple comparisons were performed using the least significant difference method. All data are presented as the mean ± standard error of at least 3 independent experiments. Results were considered statistically significant at the p < 0.05 level.

Results

Effect of TO on HUVEC viability

The cytotoxicity of the TO extract was examined by MTS assay. The viability of HUVECs treated with TO for 24 h was not affected at any of the extract concentrations (50, 100, and 200 μg/ml) tested (Fig. 1).

Effect of TO on monocyte adhesion to LPS-stimulated endothelial cells

To determine the effect of TO on endothelial cell–monocyte interactions, we performed adhesion assays. We used human monocytic THP-1 cells to mimic the interaction of monocytes with the endothelium. LPS treatment increased THP-1 cell adherence to the endothelial surface. Pretreatment with the TO extract (100 μg/ml, 1 h) decreased the LPS-induced THP-1 cell adhesion to a near-baseline level, indicating that TO can significantly decrease LPS-induced monocyte adhesion to endothelial cells (Fig. 2).

Effect of TO on adhesion molecule and inflammatory cytokine expression in LPS-stimulated endothelial cells

Endothelial activation involves the coordinated induction of genes encoding for leukocyte adhesion molecules (e.g., VCAM-1 and ICAM-1), chemotactic factors (e.g., monocyte chemoattractant protein-1 [MCP-1]), and growth factors (e.g., macrophage colony-stimulating factor). Endothelial VCAM-1 is an important mediator of mononuclear cell adhesion, given that its cognate ligand, the integrin very late antigen-4, is selectively expressed on monocytes (and on some T lymphocytes) but not on neutrophils [22]. We therefore examined whether TO treatment decreases VCAM-1 induction in response to LPS stimulation in HUVECs. Pretreatment of TO decreased the LPS-induced VCAM-1 expression by 53% (Fig. 3a). The potency of 100 μg/ml TO was quite compatible with that of 1 mM acetylsalicylic acid (Additional file 1: Figure S1). We also measured the ICAM-1 protein levels but did not observe any changes with TO treatment (Additional file 1: Figure S2).
Since MCP-1 is one of the key chemokines that regulate leukocyte trafficking and modulate interactions between leukocytes and endothelial cells [23, 24], we next examined whether TO inhibits MCP-1 expression in LPS-stimulated HUVECs. To this end, we used RT-qPCR to examine MCP-1 mRNA expression in HUVECs in response to LPS stimulation. MCP-1 mRNA expression was induced by LPS (58.5-fold), but this effect was significantly decreased (16.4-fold) in HUVECs pretreated with the TO extract (Fig. 3b).
We also measured pro-inflammatory cytokine mRNA expression levels by RT-qPCR. TNF-α, IL-1β, and IL-6 were induced by 9-, 157-, and 30-fold, respectively (Fig. 4). However, these pro-inflammatory cytokine mRNA expression levels were markedly reduced by TO pretreatment (Fig. 4).

Effect of TO on NF-κB activation in LPS-stimulated endothelial cells

LPS stimulation can lead to the activation of several intracellular signaling molecules. It has been reported that LPS can activate the mitogen-activated protein kinase (MAPK) [25, 26] and NF-κB pathways in endothelial cells [27]. These pathways are involved in controlling the expression of adhesion molecules and pro-inflammatory cytokines. Therefore, we first examined whether the TO extract inhibits MAPK (Erk1/2, p38MAPK, and JNK) activation in response to LPS stimulation. Western blot analysis indicated that TO did not markedly affect LPS-induced MAPK activation (Additional file 1: Figure S3).
To determine whether TO regulates VCAM-1 and pro-inflammatory cytokine expression by inhibiting NF-κB, we performed immunofluorescence staining of the NF-κB p65 subunit and showed that TO markedly suppresses LPS-induced NF-κB nuclear translocation (Fig. 5a and b). Since NF-κB nuclear translocation is regulated by IκBα, the degradation of which is triggered by phosphorylation, we studied whether TO treatment was associated with the inhibition of IκBα phosphorylation. As shown in Fig. 5c, TO pretreatment inhibited IκBα phosphorylation by 65%, and the degradation of IκBα induced by LPS in 30 min was also partially recovered by TO treatment. These results suggest that TO reduces the LPS-induced endothelial expression of VCAM-1 and pro-inflammatory cytokines by suppressing activation of the NF-κB pathways.

HPLC analysis of the TO methanol extract

Several peaks were monitored in the HPLC profile of the TO extract (Fig. 6). By comparing the chromatographic peaks of TO with those of the reference standards, protocatechuic acid, chlorogenic acid, caffeic acid, p-coumaric acid, and ferulic acid were qualitatively identified in the extract. The other peaks of the extract could not be identified owing to a lack of authentic references to compare with.

Discussion

Inflammation is a set of interrelated processes in response to injuries caused by a variety of biological, chemical, and physical stimuli [28]. Vascular endothelial cells form an interface between blood flow and the vessel wall, and execute a number of important functions in the maintenance of the body’s homeostasis [29]. Not only does the endothelium provide a nonadhesive and highly selective physical barrier to control the vascular permeability, but it also secretes a large number of vasoactive substances to regulate the vascular tone and remodeling of the vessel wall [30]. Most importantly, as the key regulators and major targets of the inflammatory process, endothelial cells are indispensable components of inflammation. Endothelial cells are constantly exposed to various biological, chemical, and mechanical milieus, and maintain a quiescent state with antithrombotic, anti-inflammatory, and antiproliferative properties [31]. During inflammatory responses, endothelial cells are phenotypically converted into an “activated” state that is characterized by increased permeability, induced leukocyte adhesion, and gene expression of a variety of pro-inflammatory cytokines [32]. Endothelial cell activation leads to endothelial dysfunction, which can be caused by several conditions, including various infections, diabetes or the metabolic syndrome, hypertension, smoking, and physical inactivity [33, 34].
Atherosclerosis is a chronic inflammatory disease characterized by monocyte infiltration and macrophage accumulation in the vessel wall [35]. One potentially important source of inflammation is endotoxin (LPS), a unique glycolipid that comprises most of the outer leaflet of the outer wall of gram-negative bacteria [36, 37]. The Bruneck study provided the first epidemiological evidence that subclinical endotoxemia constitutes a strong risk factor for the development of carotid atherosclerosis, particularly among smokers [5]. A 5-year prospective study showed that in subjects without atherosclerosis at baseline, ~40% of newly developed carotid atherosclerosis was attributable to chronic infection, making it a leading atherogenic risk predictor [4]. Moreover, chronic infections caused by gram-negative bacteria conferred an increased risk of atherosclerosis development, even in low-risk subjects who lacked conventional vascular risk factors [4]. Even if there is no apparent infection source, a high-fat diet augments plasma LPS (“metabolic endotoxemia”) to a concentration sufficient to trigger inflammation and metabolic diseases, such as obesity and diabetes [38, 39], by increased intestinal permeability, favoring translocation of microbiome-derived LPS to the bloodstream [38]. In addition, the plasma LPS level is also markedly increased in diabetic patients compared with that in non-diabetic subjects [40, 41]. These observations support the hypothesis that chronic exposure to endotoxins may be pathogenically linked to atherosclerosis.
TO has long been used as a herbal remedy to treat medical problems, including inflammatory disease [10, 4244]. The anti-inflammatory effects of TO extracts or its single components have been reported in both in vitro and animal models [11, 13, 4547]. Among the identified components in our HPLC analysis, protocatechuic acid [48, 49], chlorogenic acid [50], caffeic acid [51, 52], and ferulic acid [53, 54] have showed anti-inflammatory activity in the endothelial system. Recently, Hu et al. reported that aqueous extracts of TO inhibited both TNF-α and ICAM-1 expression in LPS-stimulated rat mammary microvascular endothelial cells [55]. However, the authors did not identify any underlying molecular mechanisms of the TO extracts.
In this study, we have shown that the anti-inflammatory effect of the TO methanol extract on human endothelial cells is mediated through its reduction of VCAM-1 and pro-inflammatory cytokine expression. Since endothelial VCAM-1 is an important mediator of mononuclear cell (monocytes and some T lymphocytes) adhesion, our finding of the reduced VCAM-1 expression explains the significantly inhibited monocyte adhesion to LPS-stimulated endothelial cells. We also examined whether TO reduces ICAM-1 induction in LPS-stimulated endothelial cells, but could not repeat the findings of Hu et al. [55] in this study. This might be due to the difference in solvents used for the TO extraction, because the solvent type is one of the most common factors affecting bioactive compounds in extraction processes [56, 57]. Since acetylsalicylic acid, also known as Aspirin, is reported to suppress endothelial VCAM-1 induction [58], we compared the potency of TO with acetylsalicylic acid on VCAM-1 induction in LPS-stimulated HUVECs (Additional file 1: Figure S1). At 1 mM, acetylsalicylic acid was significantly more potent than 100 μg/ml of TO at suppressing LPS-stimulated VCAM-1 induction, although 500 μM of acetylsalicylic acid only inhibited VCAM-1 induction by 18% (data not shown).
To elucidate the underlying molecular mechanism of the TO effect, components of the MAPK signaling pathway and NF-κB and its upstream effectors were examined, because this pathway and transcription factor play an essential role in the modulation of LPS-induced inflammation and transcriptional regulation. As shown in Fig. 5, the phosphorylation of IκBα and the nuclear translocation of p65 were suppressed by TO pretreatment. However, TO had no apparent effect on the phosphorylation of Erk1/2, p38MAPK, and JNK (Additional file 1: Figure S3). These results suggest that inhibition of the LPS-induced transactivation of p65 consequently reduces the expression of inflammatory mediators in TO-pretreated HUVECs.

Conclusion

Our findings demonstrated that the methanol extract of TO could attenuate LPS-induced endothelial activation by inhibiting the NF-κB pathway. Although TO has been used in the treatment of various disease, recent studies suggest it can be used effectively in the treatment of endothelial inflammation and atherosclerosis. However, to confirm these claims, more systematic, well-designed animal and randomized clinical studies with sufficient sample sizes are essential to investigate the exact action mechanisms, safety, and pharmacokinetics of this plant.

Acknowledgements

The authors would like to thank Dr. Heon Joo Park for the valuable comments and suggestions to improve the quality of the paper.

Funding

This work was supported by grants to H.S.K.; the National Research Foundation of Korea (NRF) grant (NRF-2014R1A5A2009392 and NRF-2015R1D1A1A01058340) and Inha University Research Grant (INHA-51361).

Availability of data and materials

The datasets supporting the conclusions of this article are included within the article.
Not applicable.
Not applicable.

Competing interests

The authors declare no competing financial interests.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Literatur
1.
3.
Zurück zum Zitat Cohen Tervaert JW. Cardiovascular disease due to accelerated atherosclerosis in systemic vasculitides. Best Pract Res Clin Rheumatol. 2013;27(1):33–44.CrossRefPubMed Cohen Tervaert JW. Cardiovascular disease due to accelerated atherosclerosis in systemic vasculitides. Best Pract Res Clin Rheumatol. 2013;27(1):33–44.CrossRefPubMed
4.
Zurück zum Zitat Kiechl S, Egger G, Mayr M, Wiedermann CJ, Bonora E, Oberhollenzer F, Muggeo M, Xu Q, Wick G, Poewe W, et al. Chronic infections and the risk of carotid atherosclerosis: prospective results from a large population study. Circulation. 2001;103(8):1064–70.CrossRefPubMed Kiechl S, Egger G, Mayr M, Wiedermann CJ, Bonora E, Oberhollenzer F, Muggeo M, Xu Q, Wick G, Poewe W, et al. Chronic infections and the risk of carotid atherosclerosis: prospective results from a large population study. Circulation. 2001;103(8):1064–70.CrossRefPubMed
5.
Zurück zum Zitat Wiedermann CJ, Kiechl S, Dunzendorfer S, Schratzberger P, Egger G, Oberhollenzer F, Willeit J. Association of endotoxemia with carotid atherosclerosis and cardiovascular disease: prospective results from the Bruneck study. J Am Coll Cardiol. 1999;34(7):1975–81.CrossRefPubMed Wiedermann CJ, Kiechl S, Dunzendorfer S, Schratzberger P, Egger G, Oberhollenzer F, Willeit J. Association of endotoxemia with carotid atherosclerosis and cardiovascular disease: prospective results from the Bruneck study. J Am Coll Cardiol. 1999;34(7):1975–81.CrossRefPubMed
6.
Zurück zum Zitat Weber C, Noels H. Atherosclerosis: current pathogenesis and therapeutic options. Nat Med. 2011;17(11):1410–22.CrossRefPubMed Weber C, Noels H. Atherosclerosis: current pathogenesis and therapeutic options. Nat Med. 2011;17(11):1410–22.CrossRefPubMed
7.
Zurück zum Zitat Frishman WH, Beravol P, Carosella C. Alternative and complementary medicine for preventing and treating cardiovascular disease. Dis Mon. 2009;55(3):121–92.CrossRefPubMed Frishman WH, Beravol P, Carosella C. Alternative and complementary medicine for preventing and treating cardiovascular disease. Dis Mon. 2009;55(3):121–92.CrossRefPubMed
8.
Zurück zum Zitat Orekhov AN, Sobenin IA, Korneev NV, Kirichenko TV, Myasoedova VA, Melnichenko AA, Balcells M, Edelman ER, Bobryshev YV. Anti-atherosclerotic therapy based on botanicals. Recent Pat Cardiovasc Drug Discov. 2013;8(1):56–66.CrossRefPubMedPubMedCentral Orekhov AN, Sobenin IA, Korneev NV, Kirichenko TV, Myasoedova VA, Melnichenko AA, Balcells M, Edelman ER, Bobryshev YV. Anti-atherosclerotic therapy based on botanicals. Recent Pat Cardiovasc Drug Discov. 2013;8(1):56–66.CrossRefPubMedPubMedCentral
9.
Zurück zum Zitat Guarrera PM, Savo V. Perceived health properties of wild and cultivated food plants in local and popular traditions of Italy: a review. J Ethnopharmacol. 2013;146(3):659–80.CrossRefPubMed Guarrera PM, Savo V. Perceived health properties of wild and cultivated food plants in local and popular traditions of Italy: a review. J Ethnopharmacol. 2013;146(3):659–80.CrossRefPubMed
10.
Zurück zum Zitat Schutz K, Carle R, Schieber A. Taraxacum--a review on its phytochemical and pharmacological profile. J Ethnopharmacol. 2006;107(3):313–23.CrossRefPubMed Schutz K, Carle R, Schieber A. Taraxacum--a review on its phytochemical and pharmacological profile. J Ethnopharmacol. 2006;107(3):313–23.CrossRefPubMed
11.
Zurück zum Zitat Kim HM, Shin HY, Lim KH, Ryu ST, Shin TY, Chae HJ, Kim HR, Lyu YS, An NH, Lim KS. Taraxacum Officinale inhibits tumor necrosis factor-alpha production from rat astrocytes. Immunopharmacol Immunotoxicol. 2000;22(3):519–30.CrossRefPubMed Kim HM, Shin HY, Lim KH, Ryu ST, Shin TY, Chae HJ, Kim HR, Lyu YS, An NH, Lim KS. Taraxacum Officinale inhibits tumor necrosis factor-alpha production from rat astrocytes. Immunopharmacol Immunotoxicol. 2000;22(3):519–30.CrossRefPubMed
12.
Zurück zum Zitat Hu C, Kitts DD. Luteolin and luteolin-7-O-glucoside from dandelion flower suppress iNOS and COX-2 in RAW264.7 cells. Mol Cell Biochem. 2004;265(1–2):107–13.CrossRefPubMed Hu C, Kitts DD. Luteolin and luteolin-7-O-glucoside from dandelion flower suppress iNOS and COX-2 in RAW264.7 cells. Mol Cell Biochem. 2004;265(1–2):107–13.CrossRefPubMed
13.
Zurück zum Zitat Liu L, Xiong H, Ping J, Ju Y, Zhang X. Taraxacum Officinale protects against lipopolysaccharide-induced acute lung injury in mice. J Ethnopharmacol. 2010;130(2):392–7.CrossRefPubMed Liu L, Xiong H, Ping J, Ju Y, Zhang X. Taraxacum Officinale protects against lipopolysaccharide-induced acute lung injury in mice. J Ethnopharmacol. 2010;130(2):392–7.CrossRefPubMed
14.
Zurück zum Zitat Kim IG, Kang SC, Kim KC, Choung ES, Zee OP. Screening of estrogenic and antiestrogenic activities from medicinal plants. Environ Toxicol Pharmacol. 2008;25(1):75–82.CrossRefPubMed Kim IG, Kang SC, Kim KC, Choung ES, Zee OP. Screening of estrogenic and antiestrogenic activities from medicinal plants. Environ Toxicol Pharmacol. 2008;25(1):75–82.CrossRefPubMed
15.
Zurück zum Zitat Hossen MJ, Jeon SH, Kim SC, Kim JH, Jeong D, Sung NY, Yang S, Baek KS, Kim JH, Yoon DH, et al. Vitro and in vivo anti-inflammatory activity of Phyllanthus Acidus methanolic extract. J Ethnopharmacol. 2015;168:217–28.CrossRefPubMed Hossen MJ, Jeon SH, Kim SC, Kim JH, Jeong D, Sung NY, Yang S, Baek KS, Kim JH, Yoon DH, et al. Vitro and in vivo anti-inflammatory activity of Phyllanthus Acidus methanolic extract. J Ethnopharmacol. 2015;168:217–28.CrossRefPubMed
16.
Zurück zum Zitat Wang Y, Wang B, Guerram M, Sun L, Shi W, Tian C, Zhu X, Jiang Z, Zhang L. Deoxypodophyllotoxin suppresses tumor vasculature in HUVECs by promoting cytoskeleton remodeling through LKB1-AMPK dependent rho a activatio. Oncotarget. 2015;6(30):29497–512.CrossRefPubMedPubMedCentral Wang Y, Wang B, Guerram M, Sun L, Shi W, Tian C, Zhu X, Jiang Z, Zhang L. Deoxypodophyllotoxin suppresses tumor vasculature in HUVECs by promoting cytoskeleton remodeling through LKB1-AMPK dependent rho a activatio. Oncotarget. 2015;6(30):29497–512.CrossRefPubMedPubMedCentral
17.
Zurück zum Zitat Goodwin CJ, Holt SJ, Downes S, Marshall NJ. Microculture tetrazolium assays: a comparison between two new tetrazolium salts, XTT and MTS. J Immunol Methods. 1995;179(1):95–103.CrossRefPubMed Goodwin CJ, Holt SJ, Downes S, Marshall NJ. Microculture tetrazolium assays: a comparison between two new tetrazolium salts, XTT and MTS. J Immunol Methods. 1995;179(1):95–103.CrossRefPubMed
18.
Zurück zum Zitat Cho YY, Tang F, Yao K, Lu C, Zhu F, Zheng D, Pugliese A, Bode AM, Dong Z. Cyclin-dependent kinase-3-mediated c-Jun phosphorylation at Ser63 and Ser73 enhances cell transformation. Cancer Res. 2009;69(1):272–81.CrossRefPubMedPubMedCentral Cho YY, Tang F, Yao K, Lu C, Zhu F, Zheng D, Pugliese A, Bode AM, Dong Z. Cyclin-dependent kinase-3-mediated c-Jun phosphorylation at Ser63 and Ser73 enhances cell transformation. Cancer Res. 2009;69(1):272–81.CrossRefPubMedPubMedCentral
19.
Zurück zum Zitat ET O, Kim JW, Kim JM, Kim SJ, Lee JS, Hong SS, Goodwin J, Ruthenborg RJ, Jung MG, Lee HJ, et al. NQO1 inhibits proteasome-mediated degradation of HIF-1alpha. Nat Commun. 2016;7:13593.CrossRef ET O, Kim JW, Kim JM, Kim SJ, Lee JS, Hong SS, Goodwin J, Ruthenborg RJ, Jung MG, Lee HJ, et al. NQO1 inhibits proteasome-mediated degradation of HIF-1alpha. Nat Commun. 2016;7:13593.CrossRef
20.
Zurück zum Zitat Wang Y, Cao J, Fan Y, Xie Y, Xu Z, Yin Z, Gao L, Wang C. Artemisinin inhibits monocyte adhesion to HUVECs through the NF-kappaB and MAPK pathways in vitro. Int J Mol Med. 2016;37(6):1567–75.CrossRefPubMedPubMedCentral Wang Y, Cao J, Fan Y, Xie Y, Xu Z, Yin Z, Gao L, Wang C. Artemisinin inhibits monocyte adhesion to HUVECs through the NF-kappaB and MAPK pathways in vitro. Int J Mol Med. 2016;37(6):1567–75.CrossRefPubMedPubMedCentral
21.
Zurück zum Zitat Pop R, Csernatoni F, Ranga F, Fetea F, Socaciu C: HPLC-UV Analysis Coupled with Chemometry to Identify Phenolic Biomarkers from Medicinal Plants, used as Ingredients in Two Food Supplement Formulas 2013, v. 70. Pop R, Csernatoni F, Ranga F, Fetea F, Socaciu C: HPLC-UV Analysis Coupled with Chemometry to Identify Phenolic Biomarkers from Medicinal Plants, used as Ingredients in Two Food Supplement Formulas 2013, v. 70.
22.
Zurück zum Zitat Elices MJ, Osborn L, Takada Y, Crouse C, Luhowskyj S, Hemler ME, Lobb RR. VCAM-1 on activated endothelium interacts with the leukocyte integrin VLA-4 at a site distinct from the VLA-4/fibronectin binding site. Cell. 1990;60(4):577–84.CrossRefPubMed Elices MJ, Osborn L, Takada Y, Crouse C, Luhowskyj S, Hemler ME, Lobb RR. VCAM-1 on activated endothelium interacts with the leukocyte integrin VLA-4 at a site distinct from the VLA-4/fibronectin binding site. Cell. 1990;60(4):577–84.CrossRefPubMed
23.
Zurück zum Zitat Deshmane SL, Kremlev S, Amini S, Sawaya BE. Monocyte chemoattractant protein-1 (MCP-1): an overview. J Interf Cytokine Res. 2009;29(6):313–26.CrossRef Deshmane SL, Kremlev S, Amini S, Sawaya BE. Monocyte chemoattractant protein-1 (MCP-1): an overview. J Interf Cytokine Res. 2009;29(6):313–26.CrossRef
24.
Zurück zum Zitat Olson TS, Ley K. Chemokines and chemokine receptors in leukocyte trafficking. Am J Physiol Regul Integr Comp Physiol. 2002;283(1):R7–28.CrossRefPubMed Olson TS, Ley K. Chemokines and chemokine receptors in leukocyte trafficking. Am J Physiol Regul Integr Comp Physiol. 2002;283(1):R7–28.CrossRefPubMed
25.
Zurück zum Zitat Yan W, Zhao K, Jiang Y, Huang Q, Wang J, Kan W, Wang S. Role of p38 MAPK in ICAM-1 expression of vascular endothelial cells induced by lipopolysaccharide. Shock. 2002;17(5):433–8.CrossRefPubMed Yan W, Zhao K, Jiang Y, Huang Q, Wang J, Kan W, Wang S. Role of p38 MAPK in ICAM-1 expression of vascular endothelial cells induced by lipopolysaccharide. Shock. 2002;17(5):433–8.CrossRefPubMed
26.
Zurück zum Zitat Anand AR, Bradley R, Ganju RK. LPS-induced MCP-1 expression in human microvascular endothelial cells is mediated by the tyrosine kinase, Pyk2 via the p38 MAPK/NF-kappaB-dependent pathway. Mol Immunol. 2009;46(5):962–8.CrossRefPubMed Anand AR, Bradley R, Ganju RK. LPS-induced MCP-1 expression in human microvascular endothelial cells is mediated by the tyrosine kinase, Pyk2 via the p38 MAPK/NF-kappaB-dependent pathway. Mol Immunol. 2009;46(5):962–8.CrossRefPubMed
27.
Zurück zum Zitat Ghosh S, Hayden MS. New regulators of NF-kappaB in inflammation. Nat Rev Immunol. 2008;8(11):837–48.CrossRefPubMed Ghosh S, Hayden MS. New regulators of NF-kappaB in inflammation. Nat Rev Immunol. 2008;8(11):837–48.CrossRefPubMed
28.
Zurück zum Zitat Coggins M, Rosenzweig A. The fire within: cardiac inflammatory signaling in health and disease. Circ Res. 2012;110(1):116–25.CrossRefPubMed Coggins M, Rosenzweig A. The fire within: cardiac inflammatory signaling in health and disease. Circ Res. 2012;110(1):116–25.CrossRefPubMed
29.
Zurück zum Zitat Aird WC. Phenotypic heterogeneity of the endothelium: I. Structure, function, and mechanisms. Circ Res. 2007;100(2):158–73.CrossRefPubMed Aird WC. Phenotypic heterogeneity of the endothelium: I. Structure, function, and mechanisms. Circ Res. 2007;100(2):158–73.CrossRefPubMed
30.
Zurück zum Zitat Tousoulis D, Kampoli AM, Papageorgiou N, Androulakis E, Antoniades C, Toutouzas K, Stefanadis C. Pathophysiology of atherosclerosis: the role of inflammation. Curr Pharm Des. 2011;17(37):4089–110.CrossRefPubMed Tousoulis D, Kampoli AM, Papageorgiou N, Androulakis E, Antoniades C, Toutouzas K, Stefanadis C. Pathophysiology of atherosclerosis: the role of inflammation. Curr Pharm Des. 2011;17(37):4089–110.CrossRefPubMed
31.
Zurück zum Zitat Chiu JJ, Chien S. Effects of disturbed flow on vascular endothelium: pathophysiological basis and clinical perspectives. Physiol Rev. 2011;91(1):327–87.CrossRefPubMed Chiu JJ, Chien S. Effects of disturbed flow on vascular endothelium: pathophysiological basis and clinical perspectives. Physiol Rev. 2011;91(1):327–87.CrossRefPubMed
32.
Zurück zum Zitat Pober JS, Cotran RS. Cytokines and endothelial cell biology. Physiol Rev. 1990;70(2):427–51.PubMed Pober JS, Cotran RS. Cytokines and endothelial cell biology. Physiol Rev. 1990;70(2):427–51.PubMed
33.
Zurück zum Zitat Stenvinkel P. Endothelial dysfunction and inflammation-is there a link? Nephrol Dial Transplant. 2001;16(10):1968–71.CrossRefPubMed Stenvinkel P. Endothelial dysfunction and inflammation-is there a link? Nephrol Dial Transplant. 2001;16(10):1968–71.CrossRefPubMed
34.
Zurück zum Zitat Hadi HA, Carr CS, Al Suwaidi J. Endothelial dysfunction: cardiovascular risk factors, therapy, and outcome. Vasc Health Risk Manag. 2005;1(3):183–98.PubMedPubMedCentral Hadi HA, Carr CS, Al Suwaidi J. Endothelial dysfunction: cardiovascular risk factors, therapy, and outcome. Vasc Health Risk Manag. 2005;1(3):183–98.PubMedPubMedCentral
36.
Zurück zum Zitat Rietschel ET, Kirikae T, Schade FU, Mamat U, Schmidt G, Loppnow H, Ulmer AJ, Zahringer U, Seydel U, Di Padova F, et al. Bacterial endotoxin: molecular relationships of structure to activity and function. FASEB J. 1994;8(2):217–25.PubMed Rietschel ET, Kirikae T, Schade FU, Mamat U, Schmidt G, Loppnow H, Ulmer AJ, Zahringer U, Seydel U, Di Padova F, et al. Bacterial endotoxin: molecular relationships of structure to activity and function. FASEB J. 1994;8(2):217–25.PubMed
37.
38.
Zurück zum Zitat Saito T, Hayashida H, Furugen R. Comment on: Cani et al. (2007) Metabolic endotoxemia initiates obesity and insulin resistance: Diabetes 56:1761–1772. Diabetes. 2007;56(12):e20. author reply e21CrossRefPubMed Saito T, Hayashida H, Furugen R. Comment on: Cani et al. (2007) Metabolic endotoxemia initiates obesity and insulin resistance: Diabetes 56:1761–1772. Diabetes. 2007;56(12):e20. author reply e21CrossRefPubMed
39.
Zurück zum Zitat Cani PD, Bibiloni R, Knauf C, Waget A, Neyrinck AM, Delzenne NM, Burcelin R. Changes in gut microbiota control metabolic endotoxemia-induced inflammation in high-fat diet-induced obesity and diabetes in mice. Diabetes. 2008;57(6):1470–81.CrossRefPubMed Cani PD, Bibiloni R, Knauf C, Waget A, Neyrinck AM, Delzenne NM, Burcelin R. Changes in gut microbiota control metabolic endotoxemia-induced inflammation in high-fat diet-induced obesity and diabetes in mice. Diabetes. 2008;57(6):1470–81.CrossRefPubMed
40.
Zurück zum Zitat Jayashree B, Bibin YS, Prabhu D, Shanthirani CS, Gokulakrishnan K, Lakshmi BS, Mohan V, Balasubramanyam M. Increased circulatory levels of lipopolysaccharide (LPS) and zonulin signify novel biomarkers of proinflammation in patients with type 2 diabetes. Mol Cell Biochem. 2014;388(1–2):203–10.CrossRefPubMed Jayashree B, Bibin YS, Prabhu D, Shanthirani CS, Gokulakrishnan K, Lakshmi BS, Mohan V, Balasubramanyam M. Increased circulatory levels of lipopolysaccharide (LPS) and zonulin signify novel biomarkers of proinflammation in patients with type 2 diabetes. Mol Cell Biochem. 2014;388(1–2):203–10.CrossRefPubMed
41.
Zurück zum Zitat Gomes JM, Costa JA, Alfenas RC. Metabolic endotoxemia and diabetes mellitus: a systematic review. Metabolism. 2017;68:133–44.CrossRefPubMed Gomes JM, Costa JA, Alfenas RC. Metabolic endotoxemia and diabetes mellitus: a systematic review. Metabolism. 2017;68:133–44.CrossRefPubMed
42.
Zurück zum Zitat Ahmad VU, Yasmeen S, Ali Z, Khan MA, Choudhary MI, Akhtar F, Miana GA, Zahid M. Taraxacin, a new guaianolide from Taraxacum Wallichii. J Nat Prod. 2000;63(7):1010–1.CrossRefPubMed Ahmad VU, Yasmeen S, Ali Z, Khan MA, Choudhary MI, Akhtar F, Miana GA, Zahid M. Taraxacin, a new guaianolide from Taraxacum Wallichii. J Nat Prod. 2000;63(7):1010–1.CrossRefPubMed
43.
Zurück zum Zitat Sweeney B, Vora M, Ulbricht C, Basch E. Evidence-based systematic review of dandelion (Taraxacum Officinale) by natural standard research collaboration. J Herb Pharmacother. 2005;5(1):79–93.CrossRefPubMed Sweeney B, Vora M, Ulbricht C, Basch E. Evidence-based systematic review of dandelion (Taraxacum Officinale) by natural standard research collaboration. J Herb Pharmacother. 2005;5(1):79–93.CrossRefPubMed
44.
Zurück zum Zitat Jeon HJ, Kang HJ, Jung HJ, Kang YS, Lim CJ, Kim YM, Park EH. Anti-inflammatory activity of Taraxacum Officinale. J Ethnopharmacol. 2008;115(1):82–8.CrossRefPubMed Jeon HJ, Kang HJ, Jung HJ, Kang YS, Lim CJ, Kim YM, Park EH. Anti-inflammatory activity of Taraxacum Officinale. J Ethnopharmacol. 2008;115(1):82–8.CrossRefPubMed
45.
Zurück zum Zitat Koh YJ, Cha DS, Ko JS, Park HJ, Choi HD. Anti-inflammatory effect of Taraxacum Officinale leaves on lipopolysaccharide-induced inflammatory responses in RAW 264.7 cells. J Med Food. 2010;13(4):870–8.CrossRefPubMed Koh YJ, Cha DS, Ko JS, Park HJ, Choi HD. Anti-inflammatory effect of Taraxacum Officinale leaves on lipopolysaccharide-induced inflammatory responses in RAW 264.7 cells. J Med Food. 2010;13(4):870–8.CrossRefPubMed
46.
Zurück zum Zitat Park CM, Jin KS, Lee YW, Song YS. Luteolin and chicoric acid synergistically inhibited inflammatory responses via inactivation of PI3K-Akt pathway and impairment of NF-kappaB translocation in LPS stimulated RAW 264.7 cells. Eur J Pharmacol. 2011;660(2–3):454–9.CrossRefPubMed Park CM, Jin KS, Lee YW, Song YS. Luteolin and chicoric acid synergistically inhibited inflammatory responses via inactivation of PI3K-Akt pathway and impairment of NF-kappaB translocation in LPS stimulated RAW 264.7 cells. Eur J Pharmacol. 2011;660(2–3):454–9.CrossRefPubMed
47.
Zurück zum Zitat Zhang X, Xiong H, Liu L. Effects of taraxasterol on inflammatory responses in lipopolysaccharide-induced RAW 264.7 macrophages. J Ethnopharmacol. 2012;141(1):206–11.CrossRefPubMed Zhang X, Xiong H, Liu L. Effects of taraxasterol on inflammatory responses in lipopolysaccharide-induced RAW 264.7 macrophages. J Ethnopharmacol. 2012;141(1):206–11.CrossRefPubMed
48.
Zurück zum Zitat Stumpf C, Fan Q, Hintermann C, Raaz D, Kurfurst I, Losert S, Pflederer W, Achenbach S, Daniel WG, Garlichs CD. Anti-inflammatory effects of danshen on human vascular endothelial cells in culture. Am J Chin Med. 2013;41(5):1065–77.CrossRefPubMed Stumpf C, Fan Q, Hintermann C, Raaz D, Kurfurst I, Losert S, Pflederer W, Achenbach S, Daniel WG, Garlichs CD. Anti-inflammatory effects of danshen on human vascular endothelial cells in culture. Am J Chin Med. 2013;41(5):1065–77.CrossRefPubMed
49.
Zurück zum Zitat Jiang X, Lv B, Li P, Ma X, Wang T, Zhou Q, Wang X, Gao X. Bioactivity-integrated UPLC/Q-TOF-MS of Danhong injection to identify NF-kappaB inhibitors and anti-inflammatory targets based on endothelial cell culture and network pharmacology. J Ethnopharmacol. 2015;174:270–6.CrossRefPubMed Jiang X, Lv B, Li P, Ma X, Wang T, Zhou Q, Wang X, Gao X. Bioactivity-integrated UPLC/Q-TOF-MS of Danhong injection to identify NF-kappaB inhibitors and anti-inflammatory targets based on endothelial cell culture and network pharmacology. J Ethnopharmacol. 2015;174:270–6.CrossRefPubMed
50.
Zurück zum Zitat Hebeda CB, Bolonheis SM, Nakasato A, Belinati K, Souza PD, Gouvea DR, Lopes NP, Farsky SH. Effects of chlorogenic acid on neutrophil locomotion functions in response to inflammatory stimulus. J Ethnopharmacol. 2011;135(2):261–9.CrossRefPubMed Hebeda CB, Bolonheis SM, Nakasato A, Belinati K, Souza PD, Gouvea DR, Lopes NP, Farsky SH. Effects of chlorogenic acid on neutrophil locomotion functions in response to inflammatory stimulus. J Ethnopharmacol. 2011;135(2):261–9.CrossRefPubMed
51.
Zurück zum Zitat Kim SR, Jung YR, Kim DH, An HJ, Kim MK, Kim ND, Chung HY. Caffeic acid regulates LPS-induced NF-kappaB activation through NIK/IKK and c-Src/ERK signaling pathways in endothelial cells. Arch Pharm Res. 2014;37(4):539–47.CrossRefPubMed Kim SR, Jung YR, Kim DH, An HJ, Kim MK, Kim ND, Chung HY. Caffeic acid regulates LPS-induced NF-kappaB activation through NIK/IKK and c-Src/ERK signaling pathways in endothelial cells. Arch Pharm Res. 2014;37(4):539–47.CrossRefPubMed
52.
Zurück zum Zitat Moon MK, Lee YJ, Kim JS, Kang DG, Lee HS. Effect of caffeic acid on tumor necrosis factor-alpha-induced vascular inflammation in human umbilical vein endothelial cells. Biol Pharm Bull. 2009;32(8):1371–7.CrossRefPubMed Moon MK, Lee YJ, Kim JS, Kang DG, Lee HS. Effect of caffeic acid on tumor necrosis factor-alpha-induced vascular inflammation in human umbilical vein endothelial cells. Biol Pharm Bull. 2009;32(8):1371–7.CrossRefPubMed
53.
Zurück zum Zitat Zhao J, Suyama A, Tanaka M, Matsui T. Ferulic acid enhances the vasorelaxant effect of epigallocatechin gallate in tumor necrosis factor-alpha-induced inflammatory rat aorta. J Nutr Biochem. 2014;25(7):807–14.CrossRefPubMed Zhao J, Suyama A, Tanaka M, Matsui T. Ferulic acid enhances the vasorelaxant effect of epigallocatechin gallate in tumor necrosis factor-alpha-induced inflammatory rat aorta. J Nutr Biochem. 2014;25(7):807–14.CrossRefPubMed
54.
Zurück zum Zitat Ma ZC, Hong Q, Wang YG, Tan HL, Xiao CR, Liang QD, Cai SH, Gao Y. Ferulic acid attenuates adhesion molecule expression in gamma-radiated human umbilical vascular endothelial cells. Biol Pharm Bull. 2010;33(5):752–8.CrossRefPubMed Ma ZC, Hong Q, Wang YG, Tan HL, Xiao CR, Liang QD, Cai SH, Gao Y. Ferulic acid attenuates adhesion molecule expression in gamma-radiated human umbilical vascular endothelial cells. Biol Pharm Bull. 2010;33(5):752–8.CrossRefPubMed
55.
Zurück zum Zitat Hu G, Wang J, Hong D, Zhang T, Duan H, Mu X, Yang Z. Effects of aqueous extracts of Taraxacum Officinale on expression of tumor necrosis factor-alpha and intracellular adhesion molecule 1 in LPS-stimulated RMMVECs. BMC Complement Altern Med. 2017;17(1):38.CrossRefPubMedPubMedCentral Hu G, Wang J, Hong D, Zhang T, Duan H, Mu X, Yang Z. Effects of aqueous extracts of Taraxacum Officinale on expression of tumor necrosis factor-alpha and intracellular adhesion molecule 1 in LPS-stimulated RMMVECs. BMC Complement Altern Med. 2017;17(1):38.CrossRefPubMedPubMedCentral
56.
Zurück zum Zitat Basri AM, Taha H, Ahmad N, Review A. On the pharmacological activities and phytochemicals of Alpinia Officinarum (galangal) extracts derived from bioassay-guided fractionation and isolation. Pharmacogn Rev. 2017;11(21):43–56.CrossRefPubMedPubMedCentral Basri AM, Taha H, Ahmad N, Review A. On the pharmacological activities and phytochemicals of Alpinia Officinarum (galangal) extracts derived from bioassay-guided fractionation and isolation. Pharmacogn Rev. 2017;11(21):43–56.CrossRefPubMedPubMedCentral
57.
Zurück zum Zitat DP X, Li Y, Meng X, Zhou T, Zhou Y, Zheng J, Zhang JJ, Li HB. Natural antioxidants in foods and medicinal plants: extraction, assessment and resources. Int J Mol Sci. 2017;18(1):E96.CrossRef DP X, Li Y, Meng X, Zhou T, Zhou Y, Zheng J, Zhang JJ, Li HB. Natural antioxidants in foods and medicinal plants: extraction, assessment and resources. Int J Mol Sci. 2017;18(1):E96.CrossRef
58.
Zurück zum Zitat Iademarco MF, McQuillan JJ, Rosen GD, Dean DC. Characterization of the promoter for vascular cell adhesion molecule-1 (VCAM-1). J Biol Chem. 1992;267(23):16323–9.PubMed Iademarco MF, McQuillan JJ, Rosen GD, Dean DC. Characterization of the promoter for vascular cell adhesion molecule-1 (VCAM-1). J Biol Chem. 1992;267(23):16323–9.PubMed
Metadaten
Titel
Anti-inflammatory evaluation of the methanolic extract of Taraxacum officinale in LPS-stimulated human umbilical vein endothelial cells
verfasst von
Daun Jeon
Seok Joong Kim
Hong Seok Kim
Publikationsdatum
01.12.2017
Verlag
BioMed Central
Erschienen in
BMC Complementary Medicine and Therapies / Ausgabe 1/2017
Elektronische ISSN: 2662-7671
DOI
https://doi.org/10.1186/s12906-017-2022-7

Weitere Artikel der Ausgabe 1/2017

BMC Complementary Medicine and Therapies 1/2017 Zur Ausgabe