Uric acid induces the expression of TNF‑α via the ROS‑MAPK‑NF‑κB signaling pathway in rat vascular smooth muscle cells

  • Authors:
    • Lirong Tang
    • Yu'e Xu
    • Ying Wei
    • Xu He
  • View Affiliations

  • Published online on: August 31, 2017     https://doi.org/10.3892/mmr.2017.7405
  • Pages: 6928-6933
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Hyperuricemia and artery atherosclerosis are closely associated and, as a classic inflammatory biomarker, tumor necrosis factor‑α (TNF‑α) has a direct role in atherogenesis. In the present study, it was demonstrated that uric acid was capable of inducing the generation of TNF‑α in vascular smooth muscle cells (VSMCs). The expression levels of proteins were detected using enzyme‑linked immunosorbent assays and western blot analysis. The expression levels of mRNAs were determined using reverse transcription‑quantitative polymerase chain reaction analysis, and superoxide anion levels were detected using a fluorescence microscope. From the results, it was concluded that uric acid induced the expression of TNF‑α in the VSMCs. The antioxidant, N‑acetylcysteine, eliminated the uric acid‑induced expression of TNF‑α. In addition, uric acid increased the level of reactive oxygen species (ROS) and activated the phosphorylation of p38. Subsequent experiments confirmed that the p38 mitogen‑activated protein kinase (MAPK) inhibitor, SB203580, and nuclear factor (NF)‑κB inhibitor, pyrrolidine dithiocarbamate, eliminated the uric acid‑induced expression of TNF‑α. It was demonstrated that uric acid induced the expression of TNF‑α via the ROS‑MAPK‑NF‑κB signaling pathway in VSMCs, providing novel evidence supporting the pro‑inflammatory and pro‑atherosclerotic effects of uric acid.

Introduction

Inflammation is involved in the pathological process of several cardiovascular diseases, including atherosclerosis. The results of clinical and basic studies have shown that hyperlipidemia can promote progressive narrowing of the artery lumen (1,2), and increasing epidemiological analyses and experimental data from previous studies support atherosclerosis as a chronic inflammatory disease (37). Inflammation is important in the whole process of atherosclerosis.

As an inflammatory biomarker, tumor necrosis factor-α (TNF-α) is an established pro-atherosclerotic factor (8). TNF-α promotes atherosclerosis by increasing the transcytosis of low density lipoprotein (LDL) across endothelial cells and facilitating the retention of LDL in vascular walls (9). TNF-α is capable of inducing the expression of ephrin A1 in a nuclear factor (NF)-κB-dependent manner (10) and activating endothelial cells, which facilitates the promotion of monocyte adhesion to endothelial cells (1113).

The role of vascular smooth muscle cells (VSMCs) requires consideration in the process of plaque formation and the progression of atherosclerosis. The local inflammatory factors produced by VSMCs may have a direct and essential effect on the process of atherosclerosis and the development of cardiovascular complications.

The association between increased serum uric acid and cardiovascular risk, including hypertension or coronary artery disease, in the general population has been demonstrated (1416). It has been reported that uric acid is able to stimulate VSMC proliferation and oxidative stress via the vascular renin-angiotensin system (17) in addition to increasing the expression of platelet-derived growth factor A-chain (18). Uric acid is also a marker of chronic inflammation, and can cause the secretion of inflammatory factors in vitro and in vivo (1921); it can induce the expression of C-reactive protein in VSMCs (22). This evidence suggests that uric acid is capable of accelerating the generation and progression of atherosclerosis via the pro-inflammatory response in the vessel wall.

Although uric acid is known to evoke responses of inflammation in vitro and in vivo, there is no direct evidence to indicate that the pro-inflammatory effect of uric acid on VSMCs is through TNF-α. Therefore, the present study examined the effect of uric acid on the expression of TNF-α and its mechanism in VSMCs, with a particular focus on the reactive oxygen species (ROS), mitogen-activated protein kinase (MAPK) and NF-κB signaling pathways.

Materials and methods

Reagents

Dulbecco's high-glucose modified Eagle's medium (DMEM) and fetal bovine serum (FBS) were purchased from Gibco; Thermo Fisher Scientific, Inc. (Waltham, MA, USA). Uric acid, PD98059, SB203580, SP600125, N-acetylcysteine (NAC), pyrrolidine dithiocarbamate (PDTC), thenoyltrifluoroacetone (TTFA) and diphenyleneiodonium (DPI) were produced by Sigma-Aldrich; Merck KGaA (Darmstadt, Germany). Rabbit polyclonal TNF-α antibody was provided by Invitrogen; Thermo Fisher Scientific, Inc. Mouse GAPDH antibody was ordered from CoWin Biotech Co., Ltd. (Beijing, China). Phosphorylated-p38 antibody, p38 antibody and 2′,7′-dichlorodihydrofluororescein diacetate (H2DCF-DA) were obtained from Beyotime Institute of Biotechnology (Jiangsu, China). Relative secondary antibodies were provided by CoWin Biotech Co., Ltd. (Beijing, China), and the enzyme-linked immunosorbent assay (ELISA) kit for detecting TNF-α was from West Tang Biotechnology (Shanghai, China).

Culture of rat VSMCs

VSMCs were isolated from the thoracic aorta of male Sprague-Dawley rats according to a previous report (23). Prior to the experiment, rats (n=8, 6 weeks old, 150–180 g, were purchased from the Institutional Animal Care Committee of Xi'an Jiaotong University, Xi'An, China) had free access to food and water, and were maintained in a constant environment with a conventional 12/12 h light/dark cycle. Isolated VSMCs were cultured in DMEM supplemented with 10% FBS, 100 U/ml penicillin and 100 U/ml streptomycin in a humidified atmosphere of 5% CO2, 95% air at 37°C. After 7 days, the cells exhibited a typical ‘hill and valley℉ growth morphology. The cells between passages 3 and 10 were used for all experiments. Prior to the experiments, the cells were incubated in serum-free medium for an additional 24 h. All experimental procedures were performed in accordance with the international, national and institutional guidelines, and approved by the Central Hospital of Hanzhong (Hanzhong, China).

MTT assay of VSMC viability

The viability of the VSMCs was assessed using the MTT method. VSMCs (1–1.5×106) were incubated with uric acid (10–160 mg/l) for 12 h, or with 80 mg/l of uric acid for 0, 3, 6, 12 and 24 h at 37°C. Subsequently, 20 µl of MTT (5 mg/ml; Invitrogen; Thermo Fisher Scientific, Inc.) was added to each well. Following incubation at 37°C for 4 h, the culture medium was removed and the formazan crystals were dissolved by adding 150 µl DMSO (Sigma-Aldrich; Merck KGaA). Following vigorous agitation for 10 min, the absorbance was measured at 490 nm on a microplate reader (Bio-Rad Laboratories, Inc., Hercules, CA, USA).

ELISA analysis

The VSMCs were cultured in a 96-well plate and co-cultivated with the indicated agents for the indicated times. Subsequently, the supernatant was collected via centrifugation (1,080 × g, 5 min) at 4°C and detected for TNF-α using an ELISA kit (Cat: F16960) specific for rat TNF-α.

Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) analysis of the mRNA expression of TNF-e

Total RNA was purified from the VSMCs using a RNA fast 200 purification kit (Xianfeng Biotech, Shaanxi, China). cDNA was synthesized using a Revert Aid™ First Strand cDNA synthesis kit (Takara Bio, Inc. Tokyo, Japan) according to the manufacturer's protocol. The cDNA (200 ng) was amplified using the following primer pairs (0.25 mM for forward and reverse primers) specific for rat, with GAPDH amplified as an internal control for normalization: GAPDH, sense 5′-GCAAGTTCAACGGCACAGTCAAG-3′ and antisense 5′-ACATACTCAGCACCAGCATCACC-3′); TNF-α, sense 5′-ATGGGCTCCCTCTCATCAGT-3′ and antisense 5′-GCTTGGTGGTTTGCTACGAC-3′. The expression of mRNA was expressed as relative to the internal control. The RT-qPCR analysis was performed using the ABI PRISM® 7300 Fast Real-Time PCR system (Applied Biosystems; Thermo Fisher Scientific, Inc.). The reaction products were detected by measuring the binding of SYBR Green I to DNA using the SYBR Green PCR Master Mix (Cat: 04913850001, Roche Diagnostics, Basel, Switzerland). The optimization of the amplification reaction was assured by dissociation curve analysis. The basic protocol for the qPCR was as follows: Initial incubation at 95°C for 1 min, followed by 40 cycles of 95°C for 15 sec, 60°C for 30 sec, and 72°C for 30 sec. All samples were run in triplicate, and were analyzed using the 2−ΔΔCq method, as previously described (24).

Measurement of ROS in VSMCs

VSMCs (1–1.5×106) were pretreated with NAC (10−2 M), TTFA (10−5 M) or DPI (10−5 M) for 1 h incubation at 37°C, followed by cultivation with uric acid (80 mg/l) for 3 h. For the positive ROS up control group, cells were pretreated with ROS up (Beyotime Institute of Biotechnology) for 20 min. H2DCF-DA (10 µM) was then added to each well for 1 h at 37°C and the cells were subsequently washed with PBS three times. Fluorescence images were captured at the excitation wavelength of 488 nm and the emission wavelength of 525 nm under a fluorescence microscope (Olympus, Tokyo, Japan). The fluorescence intensity of the experimental field was measured and analyzed from the fluorescence images using Image-Pro Plus software (Version 10; Media Cybernetics, Inc., Silver Springs, MD, USA). The relative fluorescence intensity was calculated as the average value from six repeated experiments.

Western blot analysis

Following the indicated treatments, the VSMCs were washed with ice-cold PBS three times, and the whole cell lysates were acquired by adding lysis buffer supplemented with protease inhibitor cocktail and phosphatase inhibitors (Roche Diagnostics, Basel, Switzerland). The concentration of protein was measured using a BCA protein assay kit (Bio-Rad Laboratories, Inc.). Equal quantities of protein extract (40 µg) were loaded and separated on a 10% SDS-PAGE gel, and then transferred onto PVDF membranes (0.22-µm; EMD Millipore, Billerica, MA, USA). The membranes were then incubated with anti-TNF-α (Cat: 25-7423-82, 1:1,500 dilution), anti-GAPDH (Cat: CW0101 M, 1:2,000 dilution), anti-p38 (Cat: AM076, 1:1,000 dilution) or anti-phosphorylated-p38 (Cat: AM071, 1:500 dilution) antibodies overnight at 4°C. Following washing, the membranes were incubated with the appropriate horseradish peroxidase-conjugated secondary antibody (Goat anti-rabbit, Cat: CW0103S; Goat anti-mouse, Cat: CW0102S) diluted 1:6,000, for 2.5 h at 25°C, followed by enhanced chemiluminescence (Cat: WBKLS0050, Merck KGaA).

Statistical analysis

All values are presented as the mean ± standard error of the mean. Statistical significance between groups was assessed using one-way analysis of variance, followed by multiple comparisons using GraphPad Prism software (version 6; GraphPad Software, Inc., La Jolla, CA, USA). P<0.05 was considered to indicate a statistically significant difference.

Results

Effect of uric acid on VSMC viability

As shown in Fig. 1, treatment with concentrations of uric acid between 10 and 80 mg/l had no significant effect on the viability of the VSMCs. In addition, no significant effect on the viability of the VSMCs was observed when the cells were exposed to the uric acid concentration of 80 mg/l for 3, 6, 12 and 24 h.

Uric acid induces the expression of TNF-α in VSMCs

The protein expression levels of TNF-α were detected using ELISA and the mRNA expression levels were detected using RT-qPCR analysis. As shown in Fig. 2A and B, the protein and mRNA expression levels of TNF-α in the VSMCs were significantly increased following exposure to 40 and 80 mg/l uric acid for 12 h (P<0.05 and P<0.01, vs. control) in a concentration-dependent manner. In addition, the results, as shown in Fig. 2C and D, showed that uric acid at a concentration of 80 mg/l increased the protein and mRNA levels of TNF-α in a time-dependent manner.

Uric acid stimulates the generation of ROS in VSMCs

To investigate whether ROS are involved in the uric acid-induced expression of TNF-α, the protein expression levels of TNF-α were determined using western blot analysis. As shown in Fig. 3A, the VSMCs co-cultivated with the antioxidant, NAC, eliminated the uric acid-induced expression of TNF-α. The levels of intracellular ROS were further analyzed using the ROS fluorescent probe, H2DCF-DA. As shown in Fig. 3B, minor DCF fluorescence was observed in the control VSMCs, which represented basal ROS generation, whereas uric acid (80 mg/l) increased the generation of ROS in the VSMCs (P<0.01, vs. control). However, pre-incubation of the cells with NAC (10−2 M), TTFA (10−5 M) and DPI (10−5 M) reduced the uric acid-stimulated generation of ROS in the VSMCs (P<0.001, vs. uric acid alone).

Uric acid induces the expression of TNF-α via the ROS-MAPK-NF-κB signaling pathway

As it is reported that MAPK and NF-κB are important in the expression of several inflammatory cytokines (25,26), the upregulation of TNF-α by uric acid in VSMCs may be associated with MAPK-NF-κB signaling. The results of the present study showed that the protein expression of TNF-α in VSMCs was increased following stimulation with 80 mg/l for 24 h. Pretreatment of the cells with SB203580 (p38 MAPK inhibitor) or PDTC (NF-κB inhibitor) for 1 h significantly reduced the uric acid-induced expression of TNF-α, however, PD98059 (extracellular signal-regulated kinase 1/2 inhibitor) and SP600125 (Janus kinase inhibitor) did not have this effect (Fig. 4A and B). As the results described above indicated that ROS are involved in the uric acid-induced expression of TNF-α in VSMCs, the present study investigated whether the activation of p38 was mediated by ROS in the VSMCs by determining the levels of phosphorylated p38. The results showed that phosphorylated p38 was markedly increased following stimulation of the cells with uric acid for 1 h. However, NAC and SB203580 significantly inhibited the uric acid-induced phosphorylation of p38 (Fig. 4C).

Discussion

Uric acid is an end product generated by the metabolism of endogenous and exogenous purines in humans. Several findings have led to the ongoing reappraisal of the role of uric acid in cardiovascular disease (2729). The findings of previous basic and clinical studies have indicated that uric acid may be an independent risk factor for cardiovascular disease and kidney disease (30). Other studies have reported that an elevated level of uric acid predicts the development of hypertension, obesity, kidney disease and diabetes (3134). Studies using animal models and cell cultures have identified mechanisms by which uric acid may induce cardiovascular and renal disease. There is increasing evidence that serum uric acid may be crucial in inflammatory responses. Atherosclerosis is a type of cardiovascular disease, and is also a chronic inflammatory disease. Inflammation is important in the progression of atherosclerosis. As a classic inflammatory biomarker, TNF-α is directly involved in atherogenesis. In the present study, it was found that uric acid induced the expression of TNF-α in VSMCs in a time-dependent and concentration-dependent manner.

ROS are important secondary messengers and are directly involved in oxidative stress. A low concentration of uric acid can exert a protective effect against the effect of ROS in vivo, whereas a high concentration of uric acid has the opposite effect (35,36). The results of the present study showed that ROS are important in the uric acid-induced expression of TNF-α. Pre-treatment with the antioxidant, NAC (10−2 M), significantly inhibited the protein expression of TNF-α in the VSMCs; whereas cells co-cultivated with TTFA (complex II inhibitor) and DPI (NADPH oxidase inhibitor) reduced uric acid-induced superoxide anion generation in the VSMCs.

MAPK and NF-κB signaling are important in inflammation (3739). The activation of NF-κB is responsible for the expression of several inflammatory cytokines. Soluble uric acid has been found to induce the expression of monocyte chemoattractant protein-1 from VSMCs through the activation of NF-κB and p38 MAPK (17). In the production of pro-inflammatory cytokines (40), a previous in vivo experiment demonstrated that uric acid is a potent pro-inflammatory molecule derived from dying cells. The results of the present study showed that p38 MAPK and NF-κB were involved in the expression of TNF-α induced by uric acid; the selective p38 MAPK and NF-κB inhibitor, PDTC, significantly inhibited the expression of TNF-α in the VSMCs.

In conclusion, the present study demonstrated that uric acid induced the expression of TNF-α in VSMCs through the ROS-p38MAPK-NF-κB signaling pathway. These findings provide novel evidence supporting the potential inflammatory effect and pro-atherosclerotic effect of uric acid.

References

1 

Besler C, Lüscher TF and Landmesser U: Molecular mechanisms of vascular effects of High-density lipoprotein: Alterations in cardiovascular disease. EMBO Mol Med. 4:251–268. 2012. View Article : Google Scholar : PubMed/NCBI

2 

Botham KM and Wheeler-Jones CP: Postprandial lipoproteins and the molecular regulation of vascular homeostasis. Prog Lipid Res. 52:446–464. 2013. View Article : Google Scholar : PubMed/NCBI

3 

Libby P: Inflammation in atherosclerosis. Nature. 420:868–874. 2002. View Article : Google Scholar : PubMed/NCBI

4 

Tall AR and Yvan-Charvet L: Cholesterol, inflammation and innate immunity. Nat Rev Immunol. 15:104–116. 2015. View Article : Google Scholar : PubMed/NCBI

5 

Liao KP and Solomon DH: Traditional cardiovascular risk factors, inflammation and cardiovascular risk in rheumatoid arthritis. Rheumatology (Oxford). 52:45–52. 2012. View Article : Google Scholar : PubMed/NCBI

6 

Krishnan E: Inflammation, oxidative stress and lipids: The risk triad for atherosclerosis in gout. Rheumatology (Oxford). 49:1229–1238. 2010. View Article : Google Scholar : PubMed/NCBI

7 

Manduteanu I and Simionescu M: Inflammation in atherosclerosis: A cause or a result of vascular disorders? J Cell Mol Med. 16:1978–1990. 2012. View Article : Google Scholar : PubMed/NCBI

8 

Zelová H and Hošek J: TNF-α signalling and inflammation: Interactions between old acquaintances. Inflamm Res. 62:641–651. 2013. View Article : Google Scholar : PubMed/NCBI

9 

Zhang Y, Yang X, Bian F, Wu P, Xing S, Xu G, Li W, Chi J, Ouyang C, Zheng T, et al: TNF-α promotes early atherosclerosis by increasing transcytosis of LDL across endothelial cells: Crosstalk between NF-κB and PPAR-γ. J Mol Cell Cardiol. 72:85–94. 2014. View Article : Google Scholar : PubMed/NCBI

10 

Ende G, Poitz DM, Augstein A, Wiedemann E, Barthel P, Maennel A, Friedrichs J, Werner C, Strasser RH and Jellinghaus S: TNF-α mediated monocyte adhesion: Role of ephrinA1 as potential link to atherosclerosis. Cardiovascular Res. 103 Suppl:S3–S5. 2014. View Article : Google Scholar

11 

Chanet A, Milenkovic D, Claude S, Maier JA, Khan M Kamran, Rakotomanomana N, Shinkaruk S, Bérard AM, Bennetau-Pelissero C, Mazur A and Morand C: Flavanone metabolites decrease monocyte adhesion to TNF-α-activated endothelial cells by modulating expression of atherosclerosis-related genes. Br J Nutr. 110:587–598. 2013. View Article : Google Scholar : PubMed/NCBI

12 

Chen HW, Lin AH, Chu HC, Li CC, Tsai CW, Chao CY, Wang CJ, Lii CK and Liu KL: Inhibition of TNF-α-Induced Inflammation by andrographolide via down-regulation of the PI3K/Akt signaling pathway. J Nat Prod. 74:2408–2413. 2011. View Article : Google Scholar : PubMed/NCBI

13 

Umar S, Hedaya O, Singh AK and Ahmed S: Thymoquinone inhibits TNF-α-induced inflammation and cell adhesion in rheumatoid arthritis synovial fibroblasts by ASK1 regulation. Toxicol Appl Pharmacol. 287:299–305. 2015. View Article : Google Scholar : PubMed/NCBI

14 

Feig DI, Kang D and Johnson RJ: Uric Acid and Cardiovascular Risk. N Engl J Med. 359:1811–1821. 2008. View Article : Google Scholar : PubMed/NCBI

15 

Cicero AF, Salvi P, D'Addato S, Rosticci M and Borghi C; Brisighella Heart Study group, : Association between serum uric acid, hypertension, vascular stiffness and subclinical atherosclerosis. J Hypertens. 32:57–64. 2014. View Article : Google Scholar : PubMed/NCBI

16 

Shimizu Y, Sato S, Koyamatsu J, Yamanashi H, Tamai M, Kadota K, Arima K, Yamasaki H, Takamura N, Aoyagi K and Maeda T: Subclinical carotid atherosclerosis and hyperuricemia in relation to renal impairment in a rural Japanese population: The Nagasaki Islands study. Atherosclerosis. 233:525–529. 2014. View Article : Google Scholar : PubMed/NCBI

17 

Corry DB, Eslami P, Yamamoto K, Nyby MD, Makino H and Tuck ML: Uric acid stimulates vascular smooth muscle cell proliferation and oxidative stress via the vascular renin-angiotensin system. J Hypertens. 26:269–275. 2008. View Article : Google Scholar : PubMed/NCBI

18 

Rao GN, Corson MA and Berk BC: Uric acid stimulates vascular smooth muscle cell proliferation by increasing platelet-derived growth factor A-chain expression. J Biol Chem. 266:8604–8608. 1991.PubMed/NCBI

19 

Gasse P, Riteau N, Charron S, Girre S, Fick L, Pétrilli V, Tschopp J, Lagente V, Quesniaux VF, Ryffel B and Couillin I: Uric acid is a danger signal activating NALP3 inflammasome in lung injury inflammation and fibrosis. Am J Respir Crit Care Med. 179:903–913. 2009. View Article : Google Scholar : PubMed/NCBI

20 

Neumann K, Castiñeiras-Vilariño M, Höckendorf U, Hannesschlager N, Lemeer S, Kupka D, Meyermann S, Lech M, Anders HJ, Kuster B, et al: Clec12a is an inhibitory receptor for uric acid crystals that regulates inflammation in response to cell death. Immunity. 40:389–399. 2014. View Article : Google Scholar : PubMed/NCBI

21 

Wijnands JM, Boonen A, Dagnelie PC, van Greevenbroek MM, Van Der Kallen CJ, Ferreira I, Schalkwijk CG, Feskens EJ, Stehouwer CD, van der Linden S and Arts IC: The cross-sectional association between uric acid and atherosclerosis and the role of low-grade inflammation: The CODAM study. Rheumatology (Oxford). 53:2053–2062. 2014. View Article : Google Scholar : PubMed/NCBI

22 

Kang DH, Park SK, Lee IK and Johnson RJ: Uric acid-induced C-reactive protein expression: Implication on cell proliferation and nitric oxide production of human vascular cells. J Am Soc Nephrol. 16:3553–3562. 2005. View Article : Google Scholar : PubMed/NCBI

23 

Ramos K and Cox LR: Primary cultures of rat aortic endothelial and smooth muscle cells: I. An in vitro model to study xenobiotic-induced vascular cytotoxicity. In Vitro Cell Dev Biol. 23:288–296. 1987. View Article : Google Scholar : PubMed/NCBI

24 

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

25 

Xu ZR, Li JY, Dong XW, Tan ZJ, Wu WZ, Xie QM and Yang YM: Apple polyphenols decrease atherosclerosis and hepatic steatosis in apoE-/- mice through the ROS/MAPK/NF-κB pathway. Nutrients. 7:7085–7105. 2015. View Article : Google Scholar : PubMed/NCBI

26 

Pamukcu B, Lip GY and Shantsila E: The nuclear factor-kappa B pathway in atherosclerosis: A potential therapeutic target for atherothrombotic vascular disease. Thromb Res. 128:117–123. 2011. View Article : Google Scholar : PubMed/NCBI

27 

Rodrigues TC, Maahs DM, Johnson RJ, Jalal DI, Kinney GL, Rivard C, Rewers M and Snell-Bergeon JK: Serum Uric acid predicts progression of subclinical coronary atherosclerosis in individuals without renal disease. Diabetes Care. 33:2471–2473. 2010. View Article : Google Scholar : PubMed/NCBI

28 

Kim SY, Guevara JP, Kim KM, Choi HK, Heitjan DF and Albert DA: Hyperuricemia and risk of stroke: A systematic review and meta-analysis. Arthritis Rheum. 61:885–892. 2009. View Article : Google Scholar : PubMed/NCBI

29 

Chen H, Mosley TH, Alonso A and Huang X: Plasma Urate and Parkinson's Disease in the Atherosclerosis Risk in Communities (ARIC) Study. Am J Epidemiol. 169:1064–1069. 2009. View Article : Google Scholar : PubMed/NCBI

30 

Wang H, Jacobs DR Jr, Gaffo AL, Gross MD, Goff DC Jr and Carr JJ: Longitudinal association between serum urate and subclinical atherosclerosis: The Coronary Artery Risk Development in Young Adults (CARDIA) study. J Intern Med. 274:594–609. 2013. View Article : Google Scholar : PubMed/NCBI

31 

Viazzi F, Parodi D, Leoncini G, Parodi A, Falqui V, Ratto E, Vettoretti S, Bezante GP, Del Sette M, Deferrari G and Pontremoli R: Serum uric acid and target organ damage in primary hypertension. Hypertension. 45:991–996. 2005. View Article : Google Scholar : PubMed/NCBI

32 

Soletsky B and Feig DI: Uric acid reduction rectifies prehypertension in obese adolescents. Hypertension. 60:1148–1156. 2012. View Article : Google Scholar : PubMed/NCBI

33 

Mellen PB, Bleyer AJ, Erlinger TP, Evans GW, Nieto FJ, Wagenknecht LE, Wofford MR and Herrington DM: Serum uric acid predicts incident hypertension in a biethnic cohort: The atherosclerosis risk in communities study. Hypertension. 48:1037–1042. 2006. View Article : Google Scholar : PubMed/NCBI

34 

Waring WS, McKnight JA, Webb DJ and Maxwell SR: Lowering serum urate does not improve endothelial function in patients with type 2 diabetes. Diabetologia. 50:2572–2579. 2007. View Article : Google Scholar : PubMed/NCBI

35 

Bagheri B, Zargari M, Meshkini F, Dinarvand K, Mokhberi V, Azizi S and Rasouli M: Uric acid and coronary artery disease, two sides of a single coin: A determinant of antioxidant system or a factor in metabolic syndrome. J Clin Diagn Res. 10:OC27–OC31. 2016.PubMed/NCBI

36 

Zhang JX, Zhang YP, Wu QN and Chen B: Uric acid induces oxidative stress via an activation of the renin-angiotensin system in 3T3-L1 adipocytes. Endocrine. 48:135–142. 2015. View Article : Google Scholar : PubMed/NCBI

37 

Zhang X, Liu J, Pang X, Zhao J, Wang S and Wu D: Aldosterone induces C-reactive protein expression via MR-ROS-MAPK-NF-κB signal pathway in rat vascular smooth muscle cells. Mol Cell Endocrinol. 395:61–68. 2014. View Article : Google Scholar : PubMed/NCBI

38 

Han C, Liu J, Liu X and Li M: Angiotensin II induces C-reactive protein expression through ERK1/2 and JNK signaling in human aortic endothelial cells. Atherosclerosis. 212:206–212. 2010. View Article : Google Scholar : PubMed/NCBI

39 

Pang X, Liu J, Zhao J, Mao J, Zhang X, Feng L, Han C, Li M, Wang S and Wu D: Homocysteine induces the expression of C-reactive protein via NMDAr-ROS-MAPK-NF-κB signal pathway in rat vascular smooth muscle cells. Atherosclerosis. 236:73–81. 2014. View Article : Google Scholar : PubMed/NCBI

40 

Leyva F, Anker SD, Godsland IF, Teixeira M, Hellewell PG, Kox WJ, Poole-Wilson PA and Coats AJ: Uric acid in chronic heart failure: A marker of chronic inflammation. Eur Heart J. 19:1814–1822. 1998. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2017
Volume 16 Issue 5

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Tang L, Xu Y, Wei Y and He X: Uric acid induces the expression of TNF‑α via the ROS‑MAPK‑NF‑κB signaling pathway in rat vascular smooth muscle cells. Mol Med Rep 16: 6928-6933, 2017
APA
Tang, L., Xu, Y., Wei, Y., & He, X. (2017). Uric acid induces the expression of TNF‑α via the ROS‑MAPK‑NF‑κB signaling pathway in rat vascular smooth muscle cells. Molecular Medicine Reports, 16, 6928-6933. https://doi.org/10.3892/mmr.2017.7405
MLA
Tang, L., Xu, Y., Wei, Y., He, X."Uric acid induces the expression of TNF‑α via the ROS‑MAPK‑NF‑κB signaling pathway in rat vascular smooth muscle cells". Molecular Medicine Reports 16.5 (2017): 6928-6933.
Chicago
Tang, L., Xu, Y., Wei, Y., He, X."Uric acid induces the expression of TNF‑α via the ROS‑MAPK‑NF‑κB signaling pathway in rat vascular smooth muscle cells". Molecular Medicine Reports 16, no. 5 (2017): 6928-6933. https://doi.org/10.3892/mmr.2017.7405