Salidroside prevents tumor necrosis factor‑α‑induced vascular inflammation by blocking mitogen‑activated protein kinase and NF‑κB signaling activation

  • Authors:
    • Ruoshui Li
    • Zhen Dong
    • Xinyu Zhuang
    • Rongchen Liu
    • Fangying Yan
    • Yufei Chen
    • Xiufang Gao
    • Haiming Shi
  • View Affiliations

  • Published online on: September 27, 2019     https://doi.org/10.3892/etm.2019.8064
  • Pages: 4137-4143
  • 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

Vascular inflammation is a key factor in the pathogenesis of atherosclerosis. Salidroside is an important active ingredient extracted from the root of the Rhodiola rosea plant, which has been reported to have antioxidative, anti‑cancer, neuroprotective and cardioprotective effects. However, the effects of salidroside on vascular inflammation have not been clarified. The purpose of the present study was to investigate the protective effects of salidroside against tumor necrosis factor (TNF)‑α‑induced vascular inflammation in cardiac microvascular endothelial cells (CMECs), a specific cell type derived from coronary micro‑vessels. Over a 24‑h period, salidroside did not exert any significant cytotoxicity up to a dose of 100 µM. Additionally, salidroside decreased the expression levels of the cell adhesion molecule vascular cell adhesion molecule‑1 (VCAM‑1) in TNF‑α‑stimulated CMECs, thus suppressing monocyte‑to‑CMEC adhesion. Salidroside also decreased the production of inflammatory cytokines such as interleukin (IL)‑1β, IL‑6 and monocyte chemotactic protein 1 (MCP‑1) in TNF‑α‑induced CMECs, as well as suppressing TNF‑α‑activated mitogen‑activated protein kinase (MAPK) and NF‑κB activation. Since MAPKs and NF‑κB both serve notable roles in regulating the expression of VCAM‑1, IL‑1β, IL‑6 and MCP‑1, the present study provided a preliminary understanding of the mechanism underlying the protective effects of salidroside. Overall, salidroside alleviated vascular inflammation by mediating MAPK and NF‑κB activation in TNF‑α‑induced CMECs. These results indicated that salidroside may have potential applications as a therapeutic agent against vascular inflammation and atherosclerosis.

Introduction

As a barrier between the vessel lumen and the surrounding tissue, the vascular endothelium is the primary participant in, and regulator of, vascular inflammatory reactions (1). Vascular inflammation has been shown to be involved in the progression of various cardiovascular diseases, including atherosclerosis (2). Tumor necrosis factor (TNF)-α is a pleiotropic, pro-inflammatory cytokine. It serves a critical role in the disruption of vascular function and subsequent inflammatory responses by triggering several intracellular signaling pathways; this may ultimately stimulate the expression of adhesion molecules, particularly vascular cell adhesion molecule-1 (VCAM-1) and cytokines such as interleukin (IL)-1β, IL-6 and monocyte chemotactic protein 1 (MCP-1) (35). These molecules recruit monocytes to the endothelial cell surface, resulting in inflammation and, ultimately, atherosclerosis (6). NF-κB and mitogen-activated protein kinases (MAPKs) serve pivotal roles in the progression of vascular inflammation by mediating the expression of adhesion molecules and chemokines in vascular endothelial cells (7,8). Both are essential for the transcriptional regulation of factors induced by TNF-α (9); therefore, compounds that can suppress TNF-α-induced MAPK and NF-κB activation represent promising candidates for the treatment and prevention of vascular endothelial dysfunction and inflammation.

Salidroside, a principal active ingredient isolated from the root of the Rhodiola rosea plant, has been reported to exert various physiological and pharmacological effects. A review summarized that salidroside was of benefit to patients with diabetes mellitus, as it was able to regulate 5′-AMP-activated protein kinase pathway-mediated glycolipid metabolism, oxidative stress and inflammatory responses (10). A previous study also demonstrated that salidroside retarded the proliferation of breast cancer cells by inhibiting MAPK pathway activation (11). In terms of cardiac protection, one report confirmed that salidroside alleviates lipopolysaccharide (LPS)-induced cardiac injury by regulating the PI3K/AKT/mTOR pathway (12). Notably, salidroside has been shown to possess strong anti-inflammatory properties. A previous study proposed that salidroside exerts anti-inflammatory effects on macrophages by blocking MAPK and NF-κB activation, and subsequently reducing the secretion of inflammatory cytokines (13). However, the effect of salidroside against TNF-α-stimulated vascular inflammation in CMECs remains to be elucidated. The present study details an investigation into the protective effects of salidroside on CMECs by regulating the production of pro-inflammatory cytokines and adhesion molecules. The results may provide novel ideas and intervention targets for the recognition and treatment of pathogenesis associated with vascular inflammation.

Materials and methods

Reagents

Salidroside was purchased from Yuanye Biotechnology Co., Ltd. Recombinant rabbit TNF-α was purchased from Novoprotein. High-glucose DMEM, RPMI 1640 medium, FBS and trypsin were purchased from Thermo Fisher Scientific, Inc. The primary antibody against factor VIII (cat. no. sc-14014) was purchased from Santa Cruz Biotechnology, Inc., and anti-VCAM-1 (cat. no. ab134047) was obtained from Abcam. Antibodies against p38 (cat. no. 8690), phospho-p38 (cat. no. 4511), p44/42 (cat. no. 4695), phospho-p44/42 (cat. no. 4370), stress-activated protein kinase (SAPK)/JNK (cat. no. 9252), phospho-SAPK/JNK (cat. no. 9255), inhibitor of NF-κB (IκB)α (cat. no. 4814), phospho-IκBα (cat. no. 2589), NF-κB p65 (cat. no. 8242) and phospho-NF-κB p65 (cat. no. 3033) were obtained from Cell Signaling Technology, Inc. The PrimeScript™ RT Reagent kit and SYBR® Premix Ex Taq™ kit were purchased from Takara Bio, Inc. The ELISA kits were purchased from MultiSciences Biotech Co., Ltd. All other chemicals were of analytical grade.

Isolation and culture of CMECs

The isolation and primary culture of CMECs were performed as previously described (14). All experiments related to animals were conducted in accordance with the Guidelines to Laboratory Animal Research of Fudan University and were approved by the Institutional Animal Care and Use Committee of Fudan University. A total of 50 2-week-old Sprague Dawley rats were purchased from Shanghai Jiesijie Experimental Animal Co., Ltd., and were housed under a 12:12 h light: Dark cycle with controlled temperature (21–25°C), humidity (50±5%) and free access to food and water in the Department of Laboratory Animal Science at Fudan University. Animal health and behavior were monitored every day. The 2-week-old male Sprague Dawley rats (30–40 g) were sacrificed by cervical dislocation after anesthesia with 5% isoflurane, and the heart was immediately excised and rinsed with PBS pre-cooled to 4°C. After removal of the atrial tissues, great vessels, epicardium and endocardium, the remaining ventricular tissues were cut into 1-mm3 pieces and plated in a 10-cm culture dish pre-coated with 1 ml FBS. The tissues were then incubated at 37°C (5% CO2) for 4 h, and for an additional 48 h in high-glucose DMEM (10% FBS). After a further 48 h, when the cells had reached a confluence of 80%, the tissue pieces were removed and the cells were passaged using trypsin (0.25%). The second generation of cells was used for subsequent experimentation. THP-1 monocytic cells (American Type Culture Collection) were cultured in RPMI-1640 containing 10% FBS, and maintained in conditions identical to those of the CMECs.

Cell viability assay

The Cell Counting Kit-8 (CCK-8; Beyotime Institute of Biotechnology) was used to determine cell viability. Briefly, CMECs (1×104 cells/well) were seeded into a 96-well plate and incubated with 10, 50 or 100 µM salidroside for 24 h. Following salidroside treatment, CCK-8 reagent was added to each well and the plate was incubated for 2 h at 37°C. The optical density was measured at 450 nm using a microplate reader (Synergy™ H4; BioTek Instruments, Inc.).

Monocyte adhesion assay

To determine the degree of monocyte THP-1 cell adhesion to CMECs, CMECs (1×105 cells/well) were seeded into 6-well plates, treated with salidroside (according to the aforementioned protocol) and 10 ng/ml TNF-α for 12 h, and incubated until 100% confluence is achieved. THP-1 cells were labeled with 5 µM 1,1′-dioctadecyl-3,3,3′,3′-tetramethylindocarbo-cyanine perchlorate for 10 min and resuspended in high-glucose DMEM (1% FBS); the cells were then added to the confluent CMECs and incubated for 1 h. The cells were washed in PBS to remove non-adherent THP-1 cells, and the adherent monocytes were visualized using a fluorescence microscope at ×200 magnification (Olympus Corporation).

Western blot analysis

Total protein was extracted from the CMECs using RIPA buffer (Beyotime Institute of Biotechnology), and the protein concentration was determined using the Bradford method. Equal amounts (30 µg) of total protein were separated by SDS-PAGE using a 10% gel, and transferred onto PVDF membranes (EMD Millipore). After being blocked with 5% non-fat dry milk for 1 h at room temperature, the membranes were incubated with the following primary antibodies at a 1:1,000 dilution, overnight at 4°C: VCAM-1, p38, phospho-p38, p44/42, phospho-P44/42, SAPK/JNK, phospho-SAPK/JNK, IκBα, phospho-IκBα, NF-κB p65 and phospho-NF-κB p65. A further incubation with a horseradish peroxidase-conjugated secondary antibody (cat. no. 31460; Invitrogen; Thermo Fisher Scientific, Inc.; dilution, 1:5,000) was then conducted at room temperature for 2 h. The bands were visualized using the Gel Doc™ XR+System (Bio-Rad Laboratories, Inc.), and the band intensity was quantified using ImageJ 1.6.0 software (National Institutes of Health).

Reverse transcription-quantitative PCR (RT-qPCR)

Total RNA was extracted from the CMECs using TRIzol® reagent (Invitrogen; Thermo Fisher Scientific, Inc.) and 1 µg/sample of total RNA was reverse transcribed using the PrimeScript™ RT Reagent kit. The reverse transcribed conditions were as follows: 37°C for 15 min, 85°C for 5 sec and 4°C for 5 min. The resulting cDNA was then amplified using the SYBR® Premix Ex Taq™ kit on a CFX Connect™ Real-Time System (Bio-Rad Laboratories, Inc.). The thermal cycler conditions were as follows: Initial denaturation at 95°C for 30 sec, followed by 40 cycles at 95°C for 5 sec and 60°C for 30 sec. The relative quantification of the mRNA expression levels was normalized to that of the β-actin endogenous control, and calculated using the PCR system software via the 2−ΔΔCq relative quantification method (15). The primers used for PCR were as follows: Rat VCAM-1 forward, 5′-GCTGCTGTTGGCTGTvGACTCTC-3′ and reverse, 5′-GCTCAGCGTCAGTGTGGATGTAG-3′; rat IL-6 forward, 5′-AGACTTCCATCCAGTTGCCTTCTTG-3′ and reverse, 5′-CATGTGTAATTAAGCCTCCGACTTGTG-3′; rat IL-1β forward, 5′-AACTGTGAAATAGCAGCTTTCG-3′ and reverse, 5′-CTGTGAGATTTGAAGCTGGATG-3′; rat MCP-1 forward, 5′-GCAGGTCTCTGTCACGCTTCTG-3′ and reverse, 5′-GAATGAGTAGCAGCAGGTGAGTGG-3′; rat β-actin forward, 5′-TACAACCTTCTTGCAGCTCC-3′ and reverse, 5′-ATCTTCATGAGGTAGTCTGTC-3′.

Immunocytochemistry

CMECs were cultured on glass coverslips and exposed to TNF-α (10 ng/ml) and salidroside (10, 50 and 100 µM), before being washed with PBS and fixed in 4% paraformaldehyde for 10 min at room temperature. The cells were incubated in 3% bovine serum albumin (Beyotime Institute of Biotechnology)-PBS blocking solution for 30 min at room temperature, with anti-factor VIII (1:250), anti-VCAM-1 (1:700) and anti-p65 (1:800) antibodies, overnight at 4°C, and then with a secondary antibody labeled with Alexa Fluor 488 (dilution, 1:200; cat. no. A11034; Invitrogen; Thermo Fisher Scientific, Inc.) for 2 h at room temperature. Cell nuclei were stained with DAPI (Sigma-Aldrich; Merck KGaA) for 10 min at room temperature, and all samples were observed under a fluorescence microscope at ×400 magnification (Olympus Corporation).

ELISA

CMECs, seeded into 6-well plates at 1×105 cells/well, were pre-treated with 10, 50 or 100 µM salidroside for 12 h, followed by 10 ng/ml TNF-α for a further 12 h in a 37°C thermostatic cell incubator. The cell supernatants were collected, and IL-1β (cat. no. KGERC007-1), IL-6 (cat. no. KGERC003-1) and MCP-1 (cat. no. KGERC113-1) secretion was quantified using the corresponding ELISA kits (Nanjing KeyGen Biotech Co., Ltd.), according to the manufacturer's instructions.

Statistical analysis

All data are expressed as the mean ± standard deviation. Each experiment was repeated at least 3 times. The data were analyzed using SPSS 19.0 software (SPSS, Inc.). The differences between groups were determined using one-way ANOVA and Tukey's test, and P<0.05 was considered to indicate a statistically significant difference.

Results

Isolation and identification of CMECs

After incubation in vitro for 24–48 h, primary rat CMECs of the ventricular tissue were spindle-like or polygonal in shape. After 72–96 h incubation, when the CMECs had reached 95–100% confluence, a ‘cobblestone’ appearance was observed. Most of the CMECs were positively stained with the microvascular endothelial cell specific anti-factor VIII antibody (Fig. 1).

Salidroside inhibits TNF-α-induced binding of monocytes to CMECs

To ensure that a non-cytotoxic concentration of salidroside was used, a CCK-8 assay was performed at three predetermined concentrations of salidroside (10, 50 and 100 µM) in TNF-α-stimulated CMECs. Since cell viability was not affected by up to 100 µM salidroside (Fig. 2A), CMECs were treated with 10, 50 and 100 µM in the subsequent experiments. Considering that monocyte-to-endothelial cell adhesion is an essential step in the development of atherosclerosis (6), the inhibitory effects of salidroside on the adhesion of THP-1 monocytes to TNF-α-stimulated CMECs were further investigated. As indicated, 100 µM salidroside did not alter the number of THP-1 cells adhering to the CMECs, and 10 ng/ml TNF-α significantly increased these numbers; however, salidroside pretreatment prior to TNF-α administration decreased the degree of monocyte adhesion to TNF-α-treated CMECs in a concentration-dependent manner (Fig. 2B and C).

Salidroside suppresses TNF-α-induced VCAM-1 protein expression

To determine whether VCAM-1 was involved in the salidroside-associated inhibition of TNF-α-induced monocyte-to-CMEC binding, the VCAM-1 mRNA expression level was quantified, and protein abundance and immunofluorescence intensity were determined. According to the observed results, treatment of CMECs with 10 ng/ml TNF-α strongly induced VCAM-1 gene expression (Fig. 3A). By contrast, pretreatment with salidroside inhibited the induction of VCAM-1 by TNF-α. In accordance with these results, western blot analysis and immunocytochemistry revealed that salidroside treatment also decreased the protein expression levels of VCAM-1 in CMECs, compared with a single dose of TNF-α (Fig. 3B and C).

Salidroside reduces the production of pro-inflammatory cytokines in CMECs

In order to identify the effect of salidroside on the production of TNF-α-induced pro-inflammatory cytokines, the expression levels of IL-1β, IL-6 and MCP-1 in CMECs were measured. After a 12-h incubation with 10, 50 or 100 µM salidroside, CMECs were treated with 10 ng/ml TNF-α for a further 12 h. RT-qPCR analysis and ELISA revealed that the TNF-α-induced mRNA expression levels and secretion of IL-1β, IL-6 and MCP-1 were reduced by salidroside treatment in CMECs (Fig. 4).

Salidroside regulates MAPK and NF-κB signaling in CMECs

In order to verify the possible downstream changes stimulated by salidroside, CMECs were pretreated with salidroside for 12 h and then incubated with TNF-α for an additional 30 min. The salidroside concentration was regulated, and MAPK and NF-κB activation were investigated. The levels of p38, Jnk and Erk phosphorylation were markedly upregulated following TNF-α treatment. However, salidroside effectively suppressed the TNF-α-induced activation of p38, Jnk and Erk, as indicated by a reduction in the phosphorylation of these proteins (Fig. 5A). Also, the effect of salidroside on NF-κB nuclear accumulation was detected by p65 immunostaining. Salidroside was demonstrated to strongly suppress the degradation and phosphorylation of IκBα, and the phosphorylation of NF-κB p65, in a concentration-dependent manner (Fig. 5B and C).

Discussion

Endothelial dysfunction is the cause of a number of cardiovascular diseases (16). Oxidative and endoplasmic reticulum stress, unfolded endothelial cell protein responses and various other factors may lead to endothelial dysfunction (17), and vascular inflammation serves a key role in the pathogenesis of endothelial dysfunction and associated cardiovascular diseases (18,19). Attenuating endothelial inflammatory responses alleviates endothelial dysfunction and reduces the incidence of cardiovascular disease. Previous studies have demonstrated that activation of the endothelium at sites of inflammation results in the expression of a series of adhesion molecules, including VCAM-1, and cytokines such as IL-1β, IL-6 and MCP-1 (20,21). The present study demonstrated that salidroside significantly reduced TNF-α-induced monocyte adherence to CMECs by downregulating the expression of VCAM-1, as well as IL-1β, IL-6 and MCP-1. Furthermore, it was indicated that these pharmacological properties of salidroside were associated with its inhibitory effects on MAPK and NF-κB activation.

VCAM-1 is an immunoglobulin-like adhesion molecule expressed on activated endothelial cells. It is able to recruit leukocytes and promote their infiltration into injured arteries; this initiates atherosclerotic plaque development via its interaction with α4β1 integrin, which is constitutively expressed on lymphocytes, monocytes and eosinophils (6). Under normal physiological conditions, VCAM-1 is not usually expressed, but is rapidly induced by stimuli such as cytokine secretion and TLR activation (22). TNF-α is a pleiotropic pro-inflammatory cytokines (5) and was used to induce vascular inflammation in the present study. The results demonstrated that salidroside attenuated the TNF-α-induced expression of VCAM-1 in a concentration-dependent manner at both the mRNA and protein levels. Additionally, the secretion of pro-inflammatory cytokines (IL-1β, IL-6 and MCP-1) by CMECs was decreased in a concentration-dependent manner following salidroside treatment. Furthermore, salidroside significantly inhibited THP-1-to-CMEC adhesion. Previous studies have demonstrated that a reduction in the overexpression of VCAM-1 and other inflammatory cytokines serves a preventative role in the development of inflammatory diseases, resulting in improved patient prognosis (2326). Thus, the present study revealed that salidroside modulates vascular inflammation by suppressing inflammatory responses in TNF-α-activated CMECs.

The MAPK and NF-κB pathways are indispensable for the activation of TNF-α-stimulated endothelial cells (27). Specifically, NF-κB is a major transcription factor which participates in the inflammatory regulation of endothelial cells by responding to pro-inflammatory stimuli (28). During endothelial inflammation, NF-κB regulates the expression of VCAM-1 and numerous inflammatory cytokines (2931). In the cytoplasm, NF-κB exists in its inactive form associated with the inhibitory protein IκB via its IκBα subunit. Once stimulated by cytokines, IκBα becomes phosphorylated and degraded, which creates the optimal conditions for NF-κB translocation into the nucleus, triggering gene transcription (32). Also, the MAPK signaling pathway is known to crucially regulate endothelial inflammation, and serves an important role in the induction of pro-inflammatory mediators (p38, Jnk and Erk) in endothelial cells after stimulation with TNF-α. The MAPKs primarily constitute a highly-conserved serine/threonine protein kinase family, which is important for signal transduction from the cell surface to the nucleus. MAPKs regulate cell growth, differentiation, environmental stress adaptation, inflammatory responses, and other important physiological and pathological processes within the cell (33). It has been shown that MAPKs also regulate the expression of VCAM-1 and inflammatory cytokines (8,34,35). A previous study indicated that salidroside reduces cell mobility by regulating NF-κB and MAPK signaling in LPS-treated-microglial cells (36). The present study revealed that salidroside significantly suppressed the TNF-α-induced phosphorylation and degradation of IκBα, and the subsequent nuclear translocation of NF-κB in CMECs. Simultaneously, salidroside was observed to inhibit TNF-α-induced p38, Erk and Jnk1/2 phosphorylation in CMECs. These data suggest that salidroside may suppress TNF-α-induced endothelial cell inflammation by inhibiting NF-κB and MAPK activation.

In conclusion, the present study revealed the suppressive effects of salidroside on TNF-α-induced CMEC activation, which effectively inhibited TNF-α-induced monocyte/CMEC interactions and the release of proinflammatory mediators, including VCAM-1, IL-1β IL-6 and MCP-1. Such suppressive effects are likely to have resulted from MAPK and NF-κB restriction. These results provide novel insights into the therapeutic potential of salidroside in preventing vascular inflammatory diseases, including atherosclerosis.

Acknowledgements

Not applicable.

Funding

The present study was supported by the National Natural Science Foundation of China (grant nos. 81573710 and 81573711).

Availability of data and materials

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

Authors' contributions

HS and XG contributed to the conception of the study. RSL, XZ and RCL contributed significantly to analysis and manuscript preparation. RSL and ZD also performed data analyses and wrote the manuscript, and FY and YC helped perform the analysis with constructive discussions. All authors read and approved the final manuscript.

Ethics approval and consent to participate

The present study was approved by the Ethics committee of Fudan University, Shanghai, China.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Pober JS and Sessa WC: Evolving functions of endothelial cells in inflammation. Nat Rev Immunol. 7:803–815. 2007. View Article : Google Scholar : PubMed/NCBI

2 

Hansson GK: Inflammation, atherosclerosis, and coronary artery disease. N Engl J Med. 352:1685–1695. 2005. View Article : Google Scholar : PubMed/NCBI

3 

Wu S, Xu H, Peng J, Wang C, Jin Y, Liu K, Sun H and Qin J: Potent anti-inflammatory effect of dioscin mediated by suppression of TNF-α-induced VCAM-1, ICAM-1and EL expression via the NF-κB pathway. Biochimie. 110:62–72. 2015. View Article : Google Scholar : PubMed/NCBI

4 

Huang W, Huang M, Ouyang H, Peng J and Liang J: Oridonin inhibits vascular inflammation by blocking NF-κB and MAPK activation. Eur J Pharmacol. 826:133–139. 2018. View Article : Google Scholar : PubMed/NCBI

5 

Zhang H, Park Y, Wu J, Chen Xp, Lee S, Yang J, Dellsperger KC and Zhang C: Role of TNF-alpha in vascular dysfunction. Clin Sci (Lond). 116:219–230. 2009. View Article : Google Scholar : PubMed/NCBI

6 

Cook-Mills JM, Marchese ME and Abdala-Valencia H: Vascular cell adhesion molecule-1 expression and signaling during disease: Regulation by reactive oxygen species and antioxidants. Antioxid Redox Signal. 15:1607–1638. 2011. View Article : Google Scholar : PubMed/NCBI

7 

Collins T, Read MA, Neish AS, Whitley MZ, Thanos D and Maniatis T: Transcriptional regulation of endothelial cell adhesion molecules: NF-kappa B and cytokine-inducible enhancers. FASEB J. 9:899–909. 1995. View Article : Google Scholar : PubMed/NCBI

8 

Pan LL and Dai M: Paeonol from Paeonia suffruticosa prevents TNF-alpha-induced monocytic cell adhesion to rat aortic endothelial cells by suppression of VCAM-1 expression. Phytomedicine. 16:1027–1032. 2009. View Article : Google Scholar : PubMed/NCBI

9 

Lee CW, Lin WN, Lin CC, Luo SF, Wang JS, Pouyssegur J and Yang CM: Transcriptional regulation of VCAM-1 expression by tumor necrosis factor-alpha in human tracheal smooth muscle cells: Involvement of MAPKs, NF-kappaB, p300, and histone acetylation. J Cell Physiol. 207:174–186. 2006. View Article : Google Scholar : PubMed/NCBI

10 

Zheng T, Bian F, Chen L, Wang Q and Jin S: beneficial effects of rhodiola and salidroside in diabetes: Potential role of AMP-activated protein kinase. Mol Diagn Ther. 23:489–498. 2019. View Article : Google Scholar : PubMed/NCBI

11 

Zhao G, Shi A, Fan Z and Du Y: Salidroside inhibits the growth of human breast cancer in vitro and in vivo. Oncol Rep. 33:2553–2560. 2015. View Article : Google Scholar : PubMed/NCBI

12 

Chen L, Liu P, Feng X and Ma C: Salidroside suppressing LPS-induced myocardial injury by inhibiting ROS-mediated PI3K/Akt/mTOR pathway in vitro and in vivo. J Cell Mol Med. 21:3178–3189. 2017. View Article : Google Scholar : PubMed/NCBI

13 

Guan S, Feng H, Song B, Guo W, Xiong Y, Huang G, Zhong W, Huo M, Chen N, Lu J and Deng X: Salidroside attenuates LPS-induced pro-inflammatory cytokine responses and improves survival in murine endotoxemia. Int Immunopharmacol. 11:2194–2199. 2011. View Article : Google Scholar : PubMed/NCBI

14 

Wang Y, Han X, Fu M, Wang J, Song Y, Liu Y, Zhang J, Zhou J and Ge J: Qiliqiangxin attenuates hypoxia-induced injury in primary rat cardiac microvascular endothelial cells via promoting HIF-1α-dependent glycolysis. J Cell Mol Med. 22:2791–2803. 2018. View Article : Google Scholar : PubMed/NCBI

15 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

16 

Widlansky ME, Gokce N, Keaney JF Jr and Vita JA: The clinical implications of endothelial dysfunction. J Am Coll Cardiol. 42:1149–1160. 2003. View Article : Google Scholar : PubMed/NCBI

17 

Amodio G, Moltedo O, Faraonio R and Remondelli P: Targeting the Endoplasmic Reticulum Unfolded Protein Response to Counteract the Oxidative Stress-Induced endothelial dysfunction. Oxid Med Cell Longev. 2018:49462892018. View Article : Google Scholar : PubMed/NCBI

18 

Gareus R, Kotsaki E, Xanthoulea S, van der Made I, Gijbels MJ, Kardakaris R, Polykratis A, Kollias G, de Winther MP and Pasparakis M: Endothelial cell-specific NF-kappaB inhibition protects mice from atherosclerosis. Cell Metab. 8:372–383. 2008. View Article : Google Scholar : PubMed/NCBI

19 

Libby P: Inflammation in atherosclerosis. Arterioscler Thromb Vasc Biol. 32:2045–2051. 2012. View Article : Google Scholar : PubMed/NCBI

20 

Robinson A: Dimethyl fumarate (Tecfidera) for multiple sclerosis. Nurse Pract. 39:10–11. 2014. View Article : Google Scholar : PubMed/NCBI

21 

Tian Y, Jain S, Kelemen SE and Autieri MV: AIF-1 expression regulates endothelial cell activation, signal transduction, and vasculogenesis. Am J Physiol Cell Physiol. 296:C256–C266. 2009. View Article : Google Scholar : PubMed/NCBI

22 

Cybulsky MI, Iiyama K, Li H, Zhu S, Chen M, Iiyama M, Davis V, Gutierrez-Ramos JC, Connelly PW and Milstone DS: A major role for VCAM-1, but not ICAM-1, in early atherosclerosis. J Clin Invest. 107:1255–1262. 2001. View Article : Google Scholar : PubMed/NCBI

23 

Dansky HM, Barlow CB, Lominska C, Sikes JL, Kao C, Weinsaft J, Cybulsky MI and Smith JD: Adhesion of monocytes to arterial endothelium and initiation of atherosclerosis are critically dependent on vascular cell adhesion molecule-1 gene dosage. Arterioscler Thromb Vasc Biol. 21:1662–1667. 2001. View Article : Google Scholar : PubMed/NCBI

24 

Kirii H, Niwa T, Yamada Y, Wada H, Saito K, Iwakura Y, Asano M, Moriwaki H and Seishima M: Lack of interleukin-1beta decreases the severity of atherosclerosis in ApoE-deficient mice. Arterioscler Thromb Vasc Biol. 23:656–660. 2003. View Article : Google Scholar : PubMed/NCBI

25 

Atreya R and Neurath MF: Involvement of IL-6 in the pathogenesis of inflammatory bowel disease and colon cancer. Clin Rev Allergy Immunol. 28:187–196. 2005. View Article : Google Scholar : PubMed/NCBI

26 

Gu L, Okada Y, Clinton SK, Gerard C, Sukhova GK, Libby P and Rollins BJ: Absence of monocyte chemoattractant protein-1 reduces atherosclerosis in low density lipoprotein receptor-deficient mice. Mol Cell. 2:275–281. 1998. View Article : Google Scholar : PubMed/NCBI

27 

Pober JS: Endothelial activation: Intracellular signaling pathways. Arthritis Res. 4 (Suppl 3):S109–S116. 2002. View Article : Google Scholar : PubMed/NCBI

28 

Csiszar A, Wang M, Lakatta EG and Ungvari Z: Inflammation and endothelial dysfunction during aging: role of NF-kappaB. J Appl Physiol (1985). 105:1333–1341. 2008. View Article : Google Scholar : PubMed/NCBI

29 

Zhou Z, Connell MC and MacEwan DJ: TNFR1-induced NF-kappaB, but not ERK, p38MAPK or JNK activation, mediates TNF-induced ICAM-1 and VCAM-1 expression on endothelial cells. Cell Signal. 19:1238–1248. 2007. View Article : Google Scholar : PubMed/NCBI

30 

Wei G, Zhang X, Su Z and Li X: Glatiramer acetate (GA) prevents TNF-α-induced monocyte adhesion to primary endothelial cells through interfering with the NF-κB pathway. Biochem Biophys Res Commun. 457:101–105. 2015. View Article : Google Scholar : PubMed/NCBI

31 

Li X, Tang Y, Ma B, Wang Z, Jiang J, Hou S, Wang S, Zhang J, Deng M, Duan Z, et al: The peptide lycosin-I attenuates TNF-α-induced inflammation in human umbilical vein endothelial cells via IκB/NF-κB signaling pathway. Inflamm Res. 67:455–466. 2018. View Article : Google Scholar : PubMed/NCBI

32 

Collins T and Cybulsky MI: NF-kappaB: Pivotal mediator or innocent bystander in atherogenesis? J Clin Invest. 107:255–264. 2001. View Article : Google Scholar : PubMed/NCBI

33 

Johnson GL and Lapadat R: Mitogen-activated protein kinase pathways mediated by ERK, JNK, and p38 protein kinases. Science. 298:1911–1912. 2002. View Article : Google Scholar : PubMed/NCBI

34 

Chen YH, Lin SJ, Ku HH, Shiao MS, Lin FY, Chen JW and Chen YL: Salvianolic acid B attenuates VCAM-1 and ICAM-1 expression in TNF-alpha-treated human aortic endothelial cells. J Cell Biochem. 82:512–521. 2001. View Article : Google Scholar : PubMed/NCBI

35 

Kim KH, Lee EN, Park JK, Lee JR, Kim JH, Choi HJ, Kim BS, Lee HW, Lee KS and Yoon S: Curcumin attenuates TNF-α-induced expression of intercellular adhesion molecule-1, vascular cell adhesion molecule-1 and proinflammatory cytokines in human endometriotic stromal cells. Phytother Res. 26:1037–1047. 2012. View Article : Google Scholar : PubMed/NCBI

36 

Hu H, Li Z, Zhu X, Lin R and Chen L: Salidroside reduces cell mobility via NF-κ B and MAPK signaling in LPS-induced BV2 microglial cells. Evid Based Complement Alternat Med. 2014:3838212014. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2019
Volume 18 Issue 5

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
Li R, Dong Z, Zhuang X, Liu R, Yan F, Chen Y, Gao X and Shi H: Salidroside prevents tumor necrosis factor‑α‑induced vascular inflammation by blocking mitogen‑activated protein kinase and NF‑κB signaling activation. Exp Ther Med 18: 4137-4143, 2019
APA
Li, R., Dong, Z., Zhuang, X., Liu, R., Yan, F., Chen, Y. ... Shi, H. (2019). Salidroside prevents tumor necrosis factor‑α‑induced vascular inflammation by blocking mitogen‑activated protein kinase and NF‑κB signaling activation. Experimental and Therapeutic Medicine, 18, 4137-4143. https://doi.org/10.3892/etm.2019.8064
MLA
Li, R., Dong, Z., Zhuang, X., Liu, R., Yan, F., Chen, Y., Gao, X., Shi, H."Salidroside prevents tumor necrosis factor‑α‑induced vascular inflammation by blocking mitogen‑activated protein kinase and NF‑κB signaling activation". Experimental and Therapeutic Medicine 18.5 (2019): 4137-4143.
Chicago
Li, R., Dong, Z., Zhuang, X., Liu, R., Yan, F., Chen, Y., Gao, X., Shi, H."Salidroside prevents tumor necrosis factor‑α‑induced vascular inflammation by blocking mitogen‑activated protein kinase and NF‑κB signaling activation". Experimental and Therapeutic Medicine 18, no. 5 (2019): 4137-4143. https://doi.org/10.3892/etm.2019.8064