2,3,5,4'‑Tetrahydroxystilbene‑2‑O‑β‑D‑glucoside inhibits septic serum‑induced inflammatory injury via interfering with the ROS‑MAPK‑NF‑κB signaling pathway in pulmonary aortic endothelial cells

  • Authors:
    • Wenqiang Li
    • Ruifang Sun
    • Sumei Zhou
    • Jinluan Ma
    • Yingguang Xie
    • Bingcan Xu
    • Huibao Long
    • Keqin Luo
    • Kuaifa Fang
  • View Affiliations

  • Published online on: December 19, 2017     https://doi.org/10.3892/ijmm.2017.3329
  • Pages: 1643-1650
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Sepsis is characterized by injury to the microvasculature and the microvascular endothelial cells, leading to barrier dysfunction. However, the specific role of injury in septic endothelial barrier dysfunction remains to be elucidated. In the present study, it was hypothesized that endothelial cell inflammatory injury is likely required for barrier dysfunction under septic conditions in vitro. 2,3,5,4'‑Tetrahydroxystilbene‑2‑O‑β‑D‑glucoside (TSG), a compound extracted from Chinese herbs, is able to inhibit the inflammatory injury of septic‑serum in endothelial cells. In the present study, cell viability was assayed by CCK‑8 method; mRNA and protein expression was identified by RT‑qPCR, western blot or Elisa, respectively and the production of reactive oxygen species was observed by a fluorescence microscope. The present study indicated that septic serum significantly decreased the cell viability of pulmonary aortic endothelial cells (PAECs) following co‑cultivation for 6 h, which occurred in a time‑dependent manner. TSG notably increased the viability of PAECs in a time‑ and concentration‑dependent manner. Further investigations revealed that septic serum increased the secretion of interleukin (IL)‑1β, IL‑6 and C‑reactive protein in PAECs, whereas pretreatment with TSG significantly decreased the secretion of these inflammatory factors. These data indicated that septic serum increased inflammatory injury to the PAECs, and TSG decreased this injury via the reactive oxygen species‑mitogen‑activated protein kinase‑nuclear factor‑κB signaling pathway.

Introduction

The organ dysfunction induced by sepsis is largely due to systemic inflammation, which results in dysfunction of the microvasculature, particularly microvascular endothelial cells (1,2). Impaired barrier function in the vascular wall, neutrophil influx into organs, impaired distribution of blood flow in microvascular beds and microvascular thromboses are the most common forms of dysfunction of microvasculature (3). The dysfunction of microvasculature is important clinically, as it has been documented early in the course of sepsis in humans and is associated with increased mortality rates, particularly when it persists over time (4,5).

Endothelial dysfunction is an early stage in several vascular diseases and inflammation is involved in the pathological process of organ dysfunction, primarily by motivating endothelial dysfunction. One of the most common results of inflammation is the activation of endothelial cell apoptosis (6-9).

The lungs are important organs and the systemic inflammatory response caused by sepsis is an important risk factor for pulmonary microvascular dysfunction (1012). Multiple mechanisms promote septic pulmonary microvascular dysfunction, including activation by mechanical interaction with activated leukocytes, inflammatory cytokines, and exposure to harmful leukocyte-derived molecules, including oxidants (13). These factors result in pulmonary microvascular endothelial cell abnormalities, including disruption of inter-pulmonary micro-vascular endothelial cell junctions and cytoskeleton-driven retraction. In addition, the locally produced inflammatory factors in target organs may have a direct and important effect in the pathological process.

2,3,5,4′-Tetrahydroxystilbene-2-O-β-D-glucoside (TSG; Fig. 1) is one of the major bioactive constituents extracted from Polygonum multiflorum Thunb, and exhibits various pharmacologic activities, including anti-inflammatory, antioxidant, and anti-atherosclerotic effects, improvement of memory and learning ability, neuroprotection, anti-aging, attenuation of human platelet aggregation and promotion of hair growth (1419). However, the protective effect of TSG on sepsis-induced inflammatory injury remains to be fully elucidated.

Sepsis can induce the expression of several inflammatory factors (20,21) and the anti-inflammatory effect of TSG is well-documented (2225). However, the pro-inflammatory mechanisms in sepsis and the mechanism underlying the anti-inflammatory effect of TSG induced by septic serum remain to be elucidated. The present study provided evidence that inflammatory factors were produced by activated endothelial cells via the reactive oxygen species (ROS)-mitogen-activated protein kinase (MAPK)-nuclear factor (NF)-κB signaling pathway. TSG exerted a protective effect via interfering with this signaling pathway.

Materials and methods

Reagents

TSG was obtained from the National Food and Drug Testing Institute (Beijing, China). Tissue culture medium 199 (M199) was purchased from GE Healthcare Life Sciences (South Logan, UT, USA) and fetal bovine serum (FBS) were acquired from Gibco; Thermo Fisher Scientific, Inc. (Waltham, MA, USA). Endothelial cell growth factor (ECGF) was purchased from Roche Diagnostics (Basel, Switzerland). TRIzol and a One-Step RT-PCR kit were from Invitrogen; Thermo Fisher Scientific, Inc. Extracellular signal-regulated kinase (ERK) 1/2 inhibitor PD98059, p38 inhibitor SB203580, c-Jun N-terminal kinase (JNK) inhibitor SP600125, antioxidant N-acetylcysteine (NAC), and NF-κB inhibitor pyrrolidine dithiocarbamate (PDTC) were from Sigma-Aldrich; Merck Millipore (Darmstadt, Germany). Rabbit NF-κB antibody (cat. no. ab207297) and rabbit CD31 antibody (cat. no. ab228364) were provided by Abcam (Cambridge, MA, USA). Rabbit monoclonal β-actin antibody (cat. no. NC011) was obtained from Zhuangzhi Biotech (Xi'an, China). ERK1/2 (cat. no. AF1051) and phosphorylated (phospho)-ERK1/2 (cat. no. AM071) antibodies, and 2′,7′-dichlorodihydrofluoro-rescein diacetate (H2DCF-DA) were obtained from Beyotime Institute of Biotechnology (Jiangsu, China). Phospho-p38 (cat. no. 8632S) and p38 (cat. no. 8690T) antibodies were from Cell Signaling Technology, Inc. (Danvers, MA, USA). Goat anti-rabbit IgG, HRP conjugated (cat. no. CW0103) or Goat anti-mouse IgG, HRP conjugated (cat. no. CW0110S) secondary antibodies were provided by CW Biotech Co., Ltd. (Beijing, China). Enzyme-linked immunosorbent assay (ELISA) kits for detecting IL-1β, IL-6 and C-reactive protein (CRP) were from West Tang Biotechnology (Shanghai, China). All other materials, except where indicated were from Sigma-Aldrich; Merck KGaA, and were of analytical grade.

Primary culture of pulmonary aortic endothelial cells (PAECs)

The rat PAECs were isolated from the pulmonary aortae of male Sprague-Dawley (SD) rats (6 weeks old) based on previously described methods (26) and identified using immunofluorescence staining with CD31 antibody (Fig. 2). Before the experiment, all rats had free access to food and water, and were maintained in a constant environment with a conventional 12/12 h light/dark cycle. The cells were cultured in M199 media supplemented with 20% FBS, 100 U/ml streptomycin, 100 U/ml penicillin, 95 µg/ml heparin and 20 µg/ml ECGF. The cells were maintained in a 5% humidified air CO2 atmosphere at 37°C. Cells at passages 3–8 at 80–90% confluence were used in the present study. All experimental procedures were performed in strict accordance with the international, national and institutional rules, and approved by the Institutional Animal Care Committee of Sun Yat-Sen University (Guangzhou, China).

Preparation of septic serum

The male SD rats (n=16, weight: 160–180 g) were randomly assigned into two groups. Septic model rats were established according to the experiment procedure described by Niwa et al (27) as follows: A laparotomy was performed through a midline incision of each animal under ether anesthesia. The cecum was filled with feces by milking stools back from the ascending colon, following which the distal one third of the cecum was tied off. The ligated region of the cecum was punctured twice with an 18-gauge needle, following which the bowel was replaced in the peritoneal cavity and the abdomen closed. In the control group, the rats were treated in the same manner as the septic rat models, but without ligation of the cecum and puncturing. At 7 h post-surgery, serum samples were collected via the abdominal aorta.

Cell viability assay

The CCK-8 method was used to detect the cell viability of PAECs. Approximately 1-1.5×106 cells were incubated with septic serum for 12 h, incubated with different concentrations of TSG for 12 h or subjected to septic serum treatment in the absence/presence of TSG in 96-well plates at 37°C. The experimental protocol was as follows: 10 µl CCK-8 solution was added to each well, followed by incubation for 4 h at 37°C. The optical density was measured at 450 nm using a microplate reader (Molecular Devices LLC, Sunnyvale, CA, USA). The mean optical density of six wells was used to calculate the cell viability percentage.

Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) analysis

The PAECs were cultured in 6-well plates at a density of 1×105 cells per well. Following the indicated treatments, the cells were washed twice with ice-cold PBS and the total mRNA was extracted using TRIzol reagent (Invitrogen; Thermo Fisher Scientific, Inc.), according to the manufacturer's protocol. The concentration of RNA was determined by measuring the absorbance at 260 nm. The total RNA was then reverse transcribed to cDNA using high capacity Reverse Transcriptase and 1 µM oligo(dT) from Takara Bio, Inc. (Tokyo, Japan). The primer pair sequences were specific to rat IL-1β, forward, 5′-CCC AAC TGG TAC ATC AGC ACC TCT C-3′ and reverse, 5′-CTA TGT CCC GAC CAT TGC TG-3′); IL-6, forward, 5′-GAT TGT ATG AAC AGC GAT GAT GC-3′ and reverse, 5′-AGA AAC GGA ACT CCA GAA GAC C-3′); CRP, forward, 5′-TTG GTG GGA GAC ATT GGA GA-3′ and reverse, 5′-AAC ATT GGG GCT GAA TAC CCT AC-3′; and GAPDH, forward, 5′-TGG AGT CTA CTG GCG TCT T-3′ and reverse, 5′-TGT CAT ATT TCT CGT GGT TCA-3′. cDNA (100 ng) was amplified using the above primer pairs (0.25 mM for forward and reverse primers) and normalized to the level of GAPDH. SYBR premix Ex taq (cat. no. DRR081A; Takara, Bio, Inc.) was used in the present study and the reaction was performed with the Mx3000P quantitative PCR system (Stratagene, La Jolla, CA, Inc.). The basic protocol for the qPCR was as follows: Initial incubation at 94°C for 55 sec, followed by 40 cycles of 94°C for 15 sec, 60°C for 30 sec, and 72°C for 30 sec. All samples were run in triplicate, and analyzed using the 2−ΔΔCq method as previously described (28)

ELISA analysis

The PAECs (1-1.5×106) were cultured in a 96-well plate and stimulated with septic serum for 12 h or pretreated with TSG for 2 h. The supernatant was then collected, and the levels of IL-1β, IL-6 and CRP in the supernatant were assayed using ELISA kits specific for rat IL-1β, IL-6 and CRP.

Immunofluorescence staining

The PAECs were incubated with the primary rabbit anti-cd31 antibody (1:300 dilution) overnight at 4°C and were then incubated with the secondary Alexa fluor 488-conjugated antibody (1:500 dilution) for 3 h at room temperature. Subsequently, the cells were stained in 2-(4-amidinophenyl)-6-indolecarbamidine dihydrochloride solution. Each of these steps was followed by washing with ice-cold PBS three times. Finally, the cells on coverslips were preserved in anti-fade mounting medium and the expression of CD31 was observed under a fluorescent microscope.

Western blot analysis

Following the indicated treatments, the cells were washed twice with ice-cold PBS (pH 7.4) and lysed in lysis buffer supplemented with protease inhibitor cocktail and phosphatase inhibitors (Roche Diagnostics; 100 µl per well of a 6-well plate). The concentration of protein was measured using a BCA protein assay kit (Bio-Rad Laboratories, Inc., Hercules, CA, USA). Equal quantities of the protein (30 µg) were loaded and separated by 10% SDS-PAGE, and blotted onto a PVDF membrane (0.45 µm; GE Healthcare Life Sciences). The membranes were incubated with anti-β-actin (1:2,000), anti-p38 (1:1,000) or anti-phospho-p38 (1:500), anti-ERK1/2 (1:1,000) or anti-phospho-ERK1/2 (1:800) antibodies at 4°C overnight. Following washing with PBS three times, the membranes were incubated with the goat anti-rabbit IgG HRP conjugated or goat anti-mouse IgG HRP conjugated secondary antibodies (1:2,500) for 3 h. The immune complexes were enhanced using chemiluminescence and band intensities were measured and analyzed by Image-Pro Plus software (Version 10; Media Cybernetics, Inc., Rockville, MD, USA).

Statistical analysis

The results are represented as the mean ± standard error of the mean. Statistical analysis was performed using the Steer-Dwass or Mann-Whitney multiple comparison test. Analysis was performed using GraphPad prism 5 (GraphPad Software, Inc., La Jolla, CA, USA). P<0.05 was considered to indicate a statistically significant difference.

Results

TSG increases the cell viability of septic serum-induced PAECs

As shown in Fig. 3A, the septic serum significantly decreased the cell viability of the PAECs following incubation for 12, 24 or 48 h. Treatment with TSG alone did not affect the viability of the PAECs (Fig. 3B). However, pretreatment of the cells with TSG at a concentration of 80 µmol/l for 2 h, followed by co-incubation with septic serum for another 12 h increased the viability of the PAECs, in a concentration-dependent manner, compared with the viability in the septic serum group (Fig. 3C).

TSG decreases septic serum-induced inflammatory cytokine expression in PAECs

The mRNA levels of IL-1β, IL-6 and CRP were determined using RT-qPCR analysis. As shown in Fig. 4A–C, the mRNA levels of IL-1β, IL-6 and CRP in the PAECs were significantly increased following exposure to septic serum for 12 h (P<0.01 vs. control). By contrast, pretreatment with TSG at a concentration 40 µmol/l for 2 h, followed by co-incubation with septic serum significantly decreased the expression of mRNA in a concentration-dependent manner.

The results from the ELISA analysis indicated that the protein expression levels of IL-1β, IL-6 and CRP in the PAECs were significantly increased following exposure to septic serum for 12 h (P<0.01 vs. septic serum only). However, pretreatment with TSG at a concentration of 40 µmol/l for 2 h, followed by co-incubation with septic serum, decreased the levels of IL-1β and IL-6 in the supernatant in a concentration-dependent manner (Fig. 4D and E). Pretreatment with TSG at a concentration of 80 µmol/l for 2 h, followed by co-incubation with septic serum notably decreased the level of CRP in the supernatant in a concentration-dependent manner (Fig. 4F).

TSG decreases septic serum-induced ROS generation in PAECs

As shown in Fig. 5 septic serum increased the generation of intracellular ROS in PAECs, whereas pretreatment with TSG for 2 h significantly eliminated the septic serum-induced intracellular expression of ROS (Fig. 5).

TSG decreases septic serum-induced protein expression levels of phosphorylated ERK1/2, p38 and JNK

As it is reported that MAPK is important in the expression of several inflammatory cytokines, the upregulated expression of inflammatory cytokines by septic serum in PAECs may be associated with MAPK signaling. The results of the present study showed that the protein levels of phospho-ERK1/2, phospho-P38 and phospho-JNK in the PAECs were increased following stimulation with septic serum (Fig. 6A–C). Pretreatment with TSG for 2 h significantly reduced the septic serum-induced protein expression of phospho-ERK1/2, phospho-P38 and phospho-JNK in the PAECs. Subsequent experiments indicated that pretreatment with the antioxidant NAC for 1.5 h, alone or in combination with TSG decreased the protein expression levels of phospho-ERK1/2, phospho-P38 and phospho-JNK. These results indicated that the MAPK pathway may be involved in septic serum-induced inflammatory cytokine expression, whereas TSG eliminated the septic serum-induced expression of inflammatory cytokines in the PAECs. Taken together, these results showed that TSG inhibited septic serum-induced inflammatory injury via interfering with the ROS-MAPK signaling pathway in PAECs.

TSG decreases septic serum-induced levels of NF-κB in PAECs

MAPK and NF-κB are involved in the expression of several inflammatory cytokines, and septic serum-induced inf lammation in PAECs may be associated with ROS-MAPK-NF-κB signaling. The results of the present study showed that the nuclear expression of NF-κB in PAECs was increased following stimulation with septic serum for 12 h. Pretreatment of the cells with NAC (antioxidant), PD98059 (ERK1/2 inhibitor), SB203580 (p38 MAPK inhibitor), SP600125 (JNK inhibitor) or PDTC (NF-κB inhibitor) for 1.5 h significantly reduced the septic serum-induced nuclear expression of NF-κB (Fig. 7). These results also indicated that ROS was involved in the septic serum-induced nuclear expression of NF-κB in PAECs.

TSG decreases septic serum induced inflammation via the ROS-MAPK-NF-κB signaling pathway in PAECs

As shown in Fig. 4, the protein levels of IL-1β, IL-6 and CRP in the cells were significantly decreased following pretreatment with TSG for 1 h (P<0.05 or P<0.01 vs. control). As a positive control, the cells pretreated with PD98059 (ERK1/2 inhibitor), SB203580 (p38 MAPK inhibitor), SP600125 (JNK inhibitor) or antioxidant NAC for 1 h showed similar effects (Fig. 8). These results indicated that the ROS-MAPK-NF-κB signaling pathway was involved in septic serum-induced inflammation in the PAECs.

Discussion

In the present report, in vitro conditions of septicemia were established to examine the protective effect of TSG on septic serum-induced inflammatory injury in PAECs. The results confirmed that the stimulation of PAECs with septic serum led to upregulation in the protein expression levels of IL-1β, IL-6 and CRP, whereas pretreatment with TSG significantly reduced the expression of these inflammatory cytokines in PAECs, in a concentration-dependent manner. Subsequent experiments indicated that the protective effect of TSG was predominantly through interfering with the ROS-MAPK-NF-κB signaling pathway.

In sepsis, multiple organ dysfunction is mainly due to systemic inflammatory injury of the microvasculature, particularly microvascular endothelial cells (29-32). Numerous clinical and laboratory investigations have indicated that, for microvascular and microvascular endothelial cells, dysfunction or injury are the initial stages of organ dysfunction (33). In addition, increased numbers of circulating endothelial cells and soluble markers of endothelial cell damage correlate with increased severity of sepsis and higher mortality rates (34,35). In addition, this septic microvascular dysfunction is clinically relevant, as the presence of microvascular dysfunction in human sepsis is associated with more severe sepsis, organ dysfunction, and increased mortality rates (36,37). The clinical outcomes, including survival rates, have been reported to be particularly poor if septic microvascular dysfunction persists over time despite usual clinical management (13,27,3840). In the present study, a rat model of sepsis was established, and rat septic serum was used to investigate the pro-inflammatory effect of septic serum and the protective effect of TSG. The results showed that septic serum significantly induced the expression levels of IL-1β, IL-6 and CRP, whereas TSG eliminated the pro-inflammatory effects of septic serum in the endothelial cells.

The activation of an inflammatory effect occurs via multiple pathways, and data from clinical and experimental investigations have shown that oxidative damage to endothelial cells is severe in sepsis (4143). ROS are important secondary messengers and are directly involved in oxidative stress. The present study showed that ROS were important in the septic serum-induced expression of inflammatory factors. Pre-treatment with antioxidant NAC 10−2 M significantly inhibited the protein expression levels of IL-1β, IL-6 and CRP in PAECs, whereas cells co-cultivated with TSG reduced septic serum-induced superoxide anion generation in PAECs.

MAPK and NF-κ B signaling are pivotal in inflammation (4449). The activation of NF-κB is responsible for the expression of several inflammatory cytokines. The results of the present study showed that p38 MAPK and NF-κB were involved in the protein expression of IL-1β, IL-6 and CRP induced by septic serum; the selective p38 MAPK and NF-κB inhibitor, PDTC, significantly inhibited the protein expression of IL-1β, IL-6 and CRP in PAECs, and TSG had a similar effect to the specific inhibitor.

In conclusion, the present study demonstrated that septic serum induced the protein expression of IL-1β, IL-6 and CRP in PAECs, and that TSG inhibited the septic serum-induced inflammatory injury via interfering with the ROS-p38MAPK-NF-κB signaling pathway. These results provide novel evidence supporting the potential inflammatory effect of septic serum and the anti-inflammatory effect of TSG.

References

1 

Churpek MM, Snyder A, Han X, Sokol S, Pettit N, Howell MD and Edelson DP: Quick sepsis-related organ failure assessment, systemic inflammatory response syndrome, and early warning scores for detecting clinical deterioration in infected patients outside the intensive care unit. Am J Respir Crit Care Med. 195:906–911. 2017. View Article : Google Scholar

2 

Colbert JF, Schmidt EP, Faubel S and Ginde AA: Severe sepsis outcomes among hospitalizations with inflammatory bowel disease. Shock. 47:128–131. 2017. View Article : Google Scholar

3 

Kanashiro A, Sônego F, Ferreira RG, Castanheira FV, Leite CA, Borges VF, Nascimento DC, Cólon DF, Alves-Filho JC, Ulloa L and Cunha FQ: Therapeutic potential and limitations of cholinergic anti-inflammatory pathway in sepsis. Pharmacol Res. 117:1–8. 2017. View Article : Google Scholar

4 

Martin JB and Badeaux JE: Interpreting laboratory tests in infection: Making sense of biomarkers in sepsis and systemic inflammatory response syndrome for intensive care unit patients. Crit Care Nurs Clin North Am. 29:119–130. 2017. View Article : Google Scholar : PubMed/NCBI

5 

Md Ralib A, Mat Nor MB and Pickering JW: Plasma neutrophil gelatinase-associated lipocalin diagnosed acute kidney injury in patients with systemic inflammatory disease and sepsis. Nephrology (Carlton). 22:412–419. 2017. View Article : Google Scholar

6 

Arcêncio L and Evora PR: The lack of clinical applications would be the cause of low interest in an endothelial dysfunction classification. Arq Bras Cardiol. 108:97–99. 2017.In English, Portuguese. PubMed/NCBI

7 

Downey RM, Liao P, Millson EC, Quyyumi AA, Sher S and Park J: Endothelial dysfunction correlates with exaggerated exercise pressor response during whole body maximal exercise in chronic kidney disease. Am J Physiol Renal Physiol. 312:F917–F924. 2017. View Article : Google Scholar : PubMed/NCBI

8 

Graw JA, Yu B, Rezoagli E, Warren HS, Buys ES, Bloch DB and Zapol WM: Endothelial dysfunction inhibits the ability of haptoglobin to prevent hemoglobin-induced hypertension. Am J Physiol Heart Circ Physiol. 312:H1120–H1127. 2017. View Article : Google Scholar : PubMed/NCBI

9 

Hashemi M, Heshmat-Ghahdarijani K, Zarean E, Baktash F and Mortazavi ZS: Evaluation of the effect of high-dose folic acid on endothelial dysfunction in pre-eclamptic patients: A randomized clinical trial. J Res Med Sci. 21:1142016. View Article : Google Scholar

10 

Rothenbach PA, Dahl B, Schwartz JJ, O'Keefe GE, Yamamoto M, Lee WM, Horton JW, Yin HL and Turnage RH: Recombinant plasma gelsolin infusion attenuates burn-induced pulmonary microvascular dysfunction. J Appl Physiol (1985). 96:25–31. 2004. View Article : Google Scholar

11 

Tsukamoto M, Tampo Y, Sawada M and Yonaha M: Paraquat-induced membrane dysfunction in pulmonary microvascular endothelial cells. Pharmacol Toxicol. 86:102–109. 2000. View Article : Google Scholar

12 

Tsukamoto M, Tampo Y, Sawada M and Yonaha M: Paraquat-induced oxidative stress and dysfunction of the glutathione redox cycle in pulmonary microvascular endothelial cells. Toxicol Appl Pharmacol. 178:82–92. 2002. View Article : Google Scholar : PubMed/NCBI

13 

Farley KS, Wang LF, Law C and Mehta S: Alveolar macrophage inducible nitric oxide synthase-dependent pulmonary micro-vascular endothelial cell septic barrier dysfunction. Microvasc Res. 76:208–216. 2008. View Article : Google Scholar : PubMed/NCBI

14 

Chang MJ, Xiao JH, Wang Y, Yan YL, Yang J and Wang JL: 2,3,5,4′-Tetrahydroxystilbene-2-O-beta-D-glucoside improves gastrointestinal motility disorders in STZ-induced diabetic mice. PLoS One. 7:e502912012. View Article : Google Scholar

15 

He H, Wang S, Tian J, Chen L, Zhang W, Zhao J, Tang H, Zhang X and Chen J: Protective effects of 2,3,5,4′-tetrahydro xystilbene-2-O-β-D-glucoside in the MPTP-induced mouse model of Parkinson's disease: Involvement of reactive oxygen species-mediated JNK, P38 and mitochondrial pathways. Eur J Pharmacol. 767:175–182. 2015. View Article : Google Scholar : PubMed/NCBI

16 

Lin CL, Hsieh SL, Leung W, Jeng JH, Huang GC, Lee CT and Wu CC: 2,3,5,4′-tetrahydroxystilbene-2-O-β-D-gluc oside suppresses human colorectal cancer cell metastasis through inhibiting NF-κB activation. Int J Oncol. 49:629–638. 2016. View Article : Google Scholar : PubMed/NCBI

17 

Ling S, Duan J, Ni R and Xu JW: 2,3,5,4′-Tetrahydroxystilbene -2-O-β-D-glucoside promotes expression of the longevity gene klotho. Oxid Med Cell Longev. 2016:31282352016. View Article : Google Scholar

18 

Ling S and Xu JW: Biological Activities of 2,3,5,4′-tetrah ydroxystilbene-2-O-β-D-glucoside in antiaging and anti-aging-related disease treatments. Oxid Med Cell Longev. 2016:49732392016. View Article : Google Scholar

19 

Peng Y, Zeng Y, Xu J, Huang XL, Zhang W and Xu XL: PPAR-gamma is involved in the protective effect of 2,3,4′,5-tetra-hydroxystilbene-2-O-beta-D-glucoside against cardiac fibrosis in pressure-overloaded rats. Eur J Pharmacol. 791:105–114. 2016. View Article : Google Scholar : PubMed/NCBI

20 

Lambertucci F, Motiño O, Villar S, Rigalli JP, de Luján Alvarez M, Catania VA, Martín-Sanz P, Carnovale CE, Quiroga AD, Francés DE and Ronco MT: Benznidazole, the trypanocidal drug used for chagas disease, induces hepatic NRF2 activation and attenuates the inflammatory response in a murine model of sepsis. Toxicol Appl Pharmacol. 315:12–22. 2017. View Article : Google Scholar

21 

Li HR, Liu J, Zhang SL, Luo T, Wu F, Dong JH, Guo YJ and Zhao L: Corilagin ameliorates the extreme inflammatory status in sepsis through TLR4 signaling pathways. BMC Complement Altern Med. 17:182017. View Article : Google Scholar : PubMed/NCBI

22 

Park SY, Jin ML, Wang Z, Park G and Choi YW: 2,3,4′,5-tetrahyd roxystilbene-2-O-β-d-glucoside exerts anti-inflammatory effects on lipopolysaccharide-stimulated microglia by inhibiting NF-κB and activating AMPK/Nrf2 pathways. Food Chem Toxicol. 97:159–167. 2016. View Article : Google Scholar : PubMed/NCBI

23 

Zhang M, Yu LM, Zhao H, Zhou XX, Yang Q, Song F, Yan L, Zhai ME, Li BY, Zhang B, et al: 2,3,5,4′-Tetrahydroxystilbe ne-2-O-β-D-glucoside protects murine hearts against ischemia/reperfusion injury by activating Notch1/Hes1 signaling and attenuating endoplasmic reticulum stress. Acta Pharmacol Sin. 38:317–330. 2017. View Article : Google Scholar : PubMed/NCBI

24 

Zhang W, Chen XF, Huang YJ, Chen QQ, Bao YJ and Zhu W: 2,3,4′,5-Tetrahydroxystilbene-2-O-β-D-glucoside inhibits angiotensin II-induced cardiac fibroblast proliferation via suppression of the reactive oxygen species-extracellular signal-regulated kinase 1/2 pathway. Clin Exp Pharmacol Physiol. 39:429–437. 2012. View Article : Google Scholar : PubMed/NCBI

25 

Zhao J, Xu S, Song F, Nian L, Zhou X and Wang S: 2,3,5,4′-tetr ahydroxystilbene-2-O-β-D-glucoside protects human umbilical vein endothelial cells against lysophosphatidylcholine-induced apoptosis by upregulating superoxide dismutase and glutathione peroxidase. IUBMB Life. 66:711–722. 2014. View Article : Google Scholar : PubMed/NCBI

26 

Peng G, Wen X, Shi Y, Jiang Y, Hu G, Zhou Y and Ran P: Development of a new method for the isolation and culture of pulmonary arterial endothelial cells from rat pulmonary arteries. J Vasc Res. 50:468–477. 2013. View Article : Google Scholar : PubMed/NCBI

27 

Niwa H, Ogawa Y, Kido Y, Abe Y, Kobayashi M, Mori T and Tanaka T: The rate of lipid oxidation in septic rat models. Jpn J Surg. 19:439–445. 1989. View Article : Google Scholar : PubMed/NCBI

28 

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

29 

Cepinskas G and Wilson JX: Inflammatory response in micro-vascular endothelium in sepsis: Role of oxidants. J Clin Biochem Nutr. 42:175–184. 2008. View Article : Google Scholar : PubMed/NCBI

30 

Garrean S, Gao XP, Brovkovych V, Shimizu J, Zhao YY and Vogel SM: Caveolin-1 regulates NF-kappaB activation and lung inflammatory response to sepsis induced by lipopolysaccharide. J Immunol. 177:4853–4860. 2006. View Article : Google Scholar : PubMed/NCBI

31 

McCuskey RS, Nishida J, McDonnell D, Baker GL, Urbaschek R and Urbaschek B: Effect of immunoglobulin G on the hepatic microvascular inflammatory response during sepsis. Shock. 5:28–33. 1996. View Article : Google Scholar : PubMed/NCBI

32 

Orfanos SE, Kotanidou A, Glynos C, Athanasiou C, Tsigkos S, Dimopoulou I, Sotiropoulou C, Zakynthinos S, Armaganidis A, Papapetropoulos A and Roussos C: Angiopoietin-2 is increased in severe sepsis: Correlation with inflammatory mediators. Crit Care Med. 35:199–206. 2007. View Article : Google Scholar

33 

Biegańska-Hensoldt S and Rosolowska-Huszcz D: Polyphenols in preventing endothelial dysfunction. Postepy Hig Med Dosw (Online). 71:227–235. 2017. View Article : Google Scholar

34 

Moussa MD, Santonocito C, Fagnoul D, Donadello K, Pradier O, Gaussem P, De Backer D and Vincent JL: Evaluation of endothelial damage in sepsis-related ARDS using circulating endothelial cells. Intensive Care Med. 41:231–238. 2015. View Article : Google Scholar

35 

Schlichting DE, Waxman AB, O'Brien LA, Wang T, Naum CC, Rubeiz GJ, Um SL, Williams M and Yan SC: Circulating endothelial and endothelial progenitor cells in patients with severe sepsis. Microvasc Res. 81:216–221. 2011. View Article : Google Scholar

36 

Lush CW and Kvietys PR: Microvascular dysfunction in sepsis. Microcirculation. 7:83–101. 2000. View Article : Google Scholar : PubMed/NCBI

37 

Vincent JL and De Backer D: Microvascular dysfunction as a cause of organ dysfunction in severe sepsis. Crit Care. 9(Suppl 4): S9–S12. 2005. View Article : Google Scholar : PubMed/NCBI

38 

Nishida J, Ekataksin W, McDonnell D, Urbaschek R, Urbaschek B and McCuskey RS: Ethanol exacerbates hepatic microvascular dysfunction, endotoxemia, and lethality in septic mice. Shock. 1:413–418. 1994. View Article : Google Scholar : PubMed/NCBI

39 

Armour J, Tyml K, Lidington D and Wilson JX: Ascorbate prevents microvascular dysfunction in the skeletal muscle of the septic rat. J Appl Physiol (1985). 90:795–803. 2001. View Article : Google Scholar

40 

De Blasi RA, Palmisani S, Alampi D, Mercieri M, Romano R, Collini S and Pinto G: Microvascular dysfunction and skeletal muscle oxygenation assessed by phase-modulation near-infrared spectroscopy in patients with septic shock. Intensive Care Med. 31:1661–1668. 2005. View Article : Google Scholar : PubMed/NCBI

41 

Constantino L, Goncalves RC, Giombelli VR, Tomasi CD, Vuolo F, Kist LW, de Oliveira GM, Pasquali MA, Bogo MR, Mauad T, et al: Regulation of lung oxidative damage by endogenous superoxide dismutase in sepsis. Intensive Care Med Exp. 2:172014. View Article : Google Scholar

42 

Schwalm MT, Pasquali M, Miguel SP, Dos Santos JP, Vuolo F, Comim CM, Petronilho F, Quevedo J, Gelain DP, Moreira JC, et al: Acute brain inflammation and oxidative damage are related to long-term cognitive deficits and markers of neurodegeneration in sepsis-survivor rats. Mol Neurobiol. 49:380–385. 2014. View Article : Google Scholar

43 

Taner G, Aydin S, Bacanli M, Sarıgöl Z, Sahin T, Başaran AA and Başaran N: Modulating effects of pycnogenol® on oxidative stress and DNA damage induced by sepsis in rats. Phytother Res. 28:1692–1700. 2014. View Article : Google Scholar : PubMed/NCBI

44 

O'Sullivan AW, Wang JH and Redmond HP: NF-kappaB and p38 MAPK inhibition improve survival in endotoxin shock and in a cecal ligation and puncture model of sepsis in combination with antibiotic therapy. J Surg Res. 152:46–53. 2009. View Article : Google Scholar

45 

Ronco MT, Manarin R, Francés D, Serra E, Revelli S and Carnovale C: Benznidazole treatment attenuates liver NF-κB activity and MAPK in a cecal ligation and puncture model of sepsis. Mol Immunol. 48:867–873. 2011. View Article : Google Scholar : PubMed/NCBI

46 

Song GY, Chung CS, Chaudry IH and Ayala A: Immune suppression in polymicrobial sepsis: differential regulation of Th1 and Th2 responses by p38 MAPK. J Surg Res. 91:141–146. 2000. View Article : Google Scholar : PubMed/NCBI

47 

Song GY, Chung CS, Jarrar D, Chaudry IH and Ayala A: Evolution of an immune suppressive macrophage phenotype as a product of P38 MAPK activation in polymicrobial sepsis. Shock. 15:42–48. 2001. View Article : Google Scholar : PubMed/NCBI

48 

Song GY, Chung CS, Jarrar D, Cioffi WG and Ayala A: Mechanism of immune dysfunction in sepsis: Inducible nitric oxide-meditated alterations in p38 MAPK activation. J Trauma. 53:276–2832. 2002. View Article : Google Scholar : PubMed/NCBI

49 

Sun Y, Li YH, Wu XX, Zheng W, Guo ZH, Li Y, Chen T, Hua ZC and Xu Q: Ethanol extract from Artemisia vestita, a traditional Tibetan medicine, exerts anti-sepsis action through down-regulating the MAPK and NF-kappaB pathways. Int J Mol Med. 17:957–962. 2006.PubMed/NCBI

Related Articles

Journal Cover

March-2018
Volume 41 Issue 3

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Li W, Sun R, Zhou S, Ma J, Xie Y, Xu B, Long H, Luo K and Fang K: 2,3,5,4'‑Tetrahydroxystilbene‑2‑O‑β‑D‑glucoside inhibits septic serum‑induced inflammatory injury via interfering with the ROS‑MAPK‑NF‑κB signaling pathway in pulmonary aortic endothelial cells. Int J Mol Med 41: 1643-1650, 2018
APA
Li, W., Sun, R., Zhou, S., Ma, J., Xie, Y., Xu, B. ... Fang, K. (2018). 2,3,5,4'‑Tetrahydroxystilbene‑2‑O‑β‑D‑glucoside inhibits septic serum‑induced inflammatory injury via interfering with the ROS‑MAPK‑NF‑κB signaling pathway in pulmonary aortic endothelial cells. International Journal of Molecular Medicine, 41, 1643-1650. https://doi.org/10.3892/ijmm.2017.3329
MLA
Li, W., Sun, R., Zhou, S., Ma, J., Xie, Y., Xu, B., Long, H., Luo, K., Fang, K."2,3,5,4'‑Tetrahydroxystilbene‑2‑O‑β‑D‑glucoside inhibits septic serum‑induced inflammatory injury via interfering with the ROS‑MAPK‑NF‑κB signaling pathway in pulmonary aortic endothelial cells". International Journal of Molecular Medicine 41.3 (2018): 1643-1650.
Chicago
Li, W., Sun, R., Zhou, S., Ma, J., Xie, Y., Xu, B., Long, H., Luo, K., Fang, K."2,3,5,4'‑Tetrahydroxystilbene‑2‑O‑β‑D‑glucoside inhibits septic serum‑induced inflammatory injury via interfering with the ROS‑MAPK‑NF‑κB signaling pathway in pulmonary aortic endothelial cells". International Journal of Molecular Medicine 41, no. 3 (2018): 1643-1650. https://doi.org/10.3892/ijmm.2017.3329