Acessibilidade / Reportar erro

Resveratrol inhibits the intracellular calcium increase and angiotensin/endothelin system activation induced by soluble uric acid in mesangial cells

Abstract

Resveratrol (Resv) is natural polyphenol found in grapes. This study evaluated the protective effect of Resv against the effects of uric acid (UA) in immortalized human mesangial cells (ihMCs). ihMCs were preincubated with Resv (12.5 µM) for 1 h and treated with UA (10 mg/dL) for 6 or 12 h. The intracellular calcium concentration [Ca2+]i was quantified by fluorescence using flow cytometry. Angiotensinogen (AGT) and pre-pro endothelin-1 (ppET-1) mRNA were assayed by quantitative real-time RT-PCR. Angiotensin II (AII) and endothelin-1 (ET-1) were assayed by ELISA. UA significantly increased [Ca2+]i. Pre-incubation with Resv significantly reduced the change in [Ca2+]i induced by UA. Incubation with UA for 6 or 12 h also increased AGT mRNA expression and AII protein synthesis. Resv blunted these increases in AGT mRNA expression and AII protein. Incubation with UA in the ihMCs increased ppET-1 expression and ET-1 protein synthesis at 6 and 12 h. When ihMCs were pre-incubated with Resv, UA had a significantly diminished effect on ppET-1 mRNA expression and ET-1 protein synthesis at 6 and 12 h, respectively. Our results suggested that UA triggers reactions including AII and ET-1 production in mesangial cells. The renin-angiotensin system may contribute to the pathogenesis of renal function and chronic kidney disease. Resv can minimize the impact of UA on AII, ET-1 and the increase of [Ca2+]i in mesangial cells, suggesting that, at least in part, Resv can prevent the effects of soluble UA in mesangial cells.

Resveratrol; Intracellular calcium; Angiotensin; Endothelin; Mesangial cells; Uric acid


Introduction

Resveratrol (Resv), a phenolic compound with significant biological activity, is of great interest to several research groups worldwide (11. Scott E, Steward WP, Gescher AJ, Brown K. Resveratrol in human cancer chemoprevention - choosing the ‘right’ dose. Mol Nutr Food Res 2012; 56: 7-13, doi: 10.1002/mnfr.201100400.
https://doi.org/10.1002/mnfr.201100400...
,22. Roupe KA, Remsberg CM, Yanez JA, Davies NM. Pharmacometrics of stilbenes: seguing towards the clinic. Curr Clin Pharmacol 2006; 1: 81-101, doi: 10.2174/157488406775268246.
https://doi.org/10.2174/1574884067752682...
). Dietary intake of Resv in grapes, red wine, peanuts, purple grape juice, and berries may have beneficial effects on human heath (11. Scott E, Steward WP, Gescher AJ, Brown K. Resveratrol in human cancer chemoprevention - choosing the ‘right’ dose. Mol Nutr Food Res 2012; 56: 7-13, doi: 10.1002/mnfr.201100400.
https://doi.org/10.1002/mnfr.201100400...

2. Roupe KA, Remsberg CM, Yanez JA, Davies NM. Pharmacometrics of stilbenes: seguing towards the clinic. Curr Clin Pharmacol 2006; 1: 81-101, doi: 10.2174/157488406775268246.
https://doi.org/10.2174/1574884067752682...

3. Smoliga JM, Baur JA, Hausenblas HA. Resveratrol and health - a comprehensive review of human clinical trials. Mol Nutr Food Res 2011; 55: 1129-1141, doi: 10.1002/mnfr.201100143.
https://doi.org/10.1002/mnfr.201100143...

4. Delmas D, Aires V, Limagne E, Dutartre P, Mazue F, Ghiringhelli F, et al. Transport, stability, and biological activity of resveratrol. Ann N Y Acad Sci 2011; 1215: 48-59, doi: 10.1111/j.1749-6632.2010.05871.x.
https://doi.org/10.1111/j.1749-6632.2010...

5. Timmers S, Konings E, Bilet L, Houtkooper RH, van de Weijer T, Goossens GH, et al. Calorie restriction-like effects of 30 days of resveratrol supplementation on energy metabolism and metabolic profile in obese humans. Cell Metab 2011; 14: 612-622, doi: 10.1016/j.cmet.2011.10.002.
https://doi.org/10.1016/j.cmet.2011.10.0...

6. Brasnyo P, Molnar GA, Mohas M, Marko L, Laczy B, Cseh J, et al. Resveratrol improves insulin sensitivity, reduces oxidative stress and activates the Akt pathway in type 2 diabetic patients. Br J Nutr 2011; 106: 383-389, doi: 10.1017/S0007114511000316.
https://doi.org/10.1017/S000711451100031...
-77. Magyar K, Halmosi R, Palfi A, Feher G, Czopf L, Fulop A, et al. Cardioprotection by resveratrol: A human clinical trial in patients with stable coronary artery disease. Clin Hemorheol Microcirc 2012; 50: 179-187.), and may have health-promoting antinephrolithic, antidiabetes, anticancer, antioxidation, anti-inflammation, cardioprotective, chemopreventive, and neuroprotective properties (55. Timmers S, Konings E, Bilet L, Houtkooper RH, van de Weijer T, Goossens GH, et al. Calorie restriction-like effects of 30 days of resveratrol supplementation on energy metabolism and metabolic profile in obese humans. Cell Metab 2011; 14: 612-622, doi: 10.1016/j.cmet.2011.10.002.
https://doi.org/10.1016/j.cmet.2011.10.0...

6. Brasnyo P, Molnar GA, Mohas M, Marko L, Laczy B, Cseh J, et al. Resveratrol improves insulin sensitivity, reduces oxidative stress and activates the Akt pathway in type 2 diabetic patients. Br J Nutr 2011; 106: 383-389, doi: 10.1017/S0007114511000316.
https://doi.org/10.1017/S000711451100031...

7. Magyar K, Halmosi R, Palfi A, Feher G, Czopf L, Fulop A, et al. Cardioprotection by resveratrol: A human clinical trial in patients with stable coronary artery disease. Clin Hemorheol Microcirc 2012; 50: 179-187.

8. Kondratyuk TP, Park EJ, Marler LE, Ahn S, Yuan Y, Choi Y, et al. Resveratrol derivatives as promising chemopreventive agents with improved potency and selectivity. Mol Nutr Food Res 2011; 55: 1249-1265, doi: 10.1002/mnfr.201100122.
https://doi.org/10.1002/mnfr.201100122...
-99. Matsuoka A, Kodama Y, Fukuhara K, Honda S, Hayashi M, Sai K, et al. A pilot study of evaluation of the antioxidative activity of resveratrol and its analogue in a 6-month feeding test in young adult mice. Food Chem Toxicol 2008; 46: 1125-1130, doi: 10.1016/j.fct.2007.11.008.
https://doi.org/10.1016/j.fct.2007.11.00...
). In addition, Resv may prevent renal lipotoxicity and have antihyperuricemic activity (1010. Kim MY, Lim JH, Youn HH, Hong YA, Yang KS, Park HS, et al. Resveratrol prevents renal lipotoxicity and inhibits mesangial cell glucotoxicity in a manner dependent on the AMPK-SIRT1-PGC1alpha axis in db/db mice. Diabetologia 2013; 56: 204-217, doi: 10.1007/s00125-012-2747-2.
https://doi.org/10.1007/s00125-012-2747-...
,1111. Shi YW, Wang CP, Liu L, Liu YL, Wang X, Hong Y, et al. Antihyperuricemic and nephroprotective effects of resveratrol and its analogues in hyperuricemic mice. Mol Nutr Food Res 2012; 56: 1433-1444, doi: 10.1002/mnfr.201100828.
https://doi.org/10.1002/mnfr.201100828...
).

Several pathways are thought to be related to the protective effect of Resv. In a nephrolithic model, about 70% of kidney stones were composed of calcium oxalate (CaOx), which causes renal cell injury through the production of reactive oxygen species (ROS) and nicotinamide adenine dinucleotide phosphate (NADPH) when deposited in kidney tissue (1212. Khan SR. Hyperoxaluria-induced oxidative stress and antioxidants for renal protection. Urol Res 2005; 33: 349-357, doi: 10.1007/s00240-005-0492-4.
https://doi.org/10.1007/s00240-005-0492-...
). Resv exerts its antinephrolithic potential via the inhibition of ROS, monocyte chemoattractant protein-1 (MCP-1), hyaluronan, and osteopontin (OPN) signaling (1313. Hong SH, Lee HJ, Sohn EJ, Ko HS, Shim BS, Ahn KS, et al. Anti-nephrolithic potential of resveratrol via inhibition of ROS, MCP-1, hyaluronan and osteopontin in vitro and in vivo. Pharmacol Rep 2013; 65: 970-979, doi: 10.1016/S1734-1140(13)71078-8.
https://doi.org/10.1016/S1734-1140(13)71...
). Resv has been shown to suppress the migration of oxalate-treated human renal epithelial cells (HRCs), It also can attenuate the expression of NADPH oxidase subunit, MCP-1, and OPN mRNAs, and downregulate the expression of transforming growth factor-β (TGF-β1), TGF-β receptor and hyaluronan proteins in oxalate-treated HRCs (1313. Hong SH, Lee HJ, Sohn EJ, Ko HS, Shim BS, Ahn KS, et al. Anti-nephrolithic potential of resveratrol via inhibition of ROS, MCP-1, hyaluronan and osteopontin in vitro and in vivo. Pharmacol Rep 2013; 65: 970-979, doi: 10.1016/S1734-1140(13)71078-8.
https://doi.org/10.1016/S1734-1140(13)71...
).

Diabetic nephropathy is a serious complication of type 1 and type 2 diabetes. It is characterized by an expansion of the glomerular mesangium caused by mesangial cell proliferation and an excess of extracellular matrix (ECM) proteins synthesized by mesangial cells (1414. Zhang L, Pang S, Deng B, Qian L, Chen J, Zou J, et al. High glucose induces renal mesangial cell proliferation and fibronectin expression through JNK/NF-kappaB/NADPH oxidase/ROS pathway, which is inhibited by resveratrol. Int J Biochem Cell Biol 2012; 44: 629-638, doi: 10.1016/j.biocel.2012.01.001.
https://doi.org/10.1016/j.biocel.2012.01...
). There is increasing evidence that overproduction of ROS is involved in the development of diabetic nephropathy (1515. Ha H, Hwang IA, Park JH, Lee HB. Role of reactive oxygen species in the pathogenesis of diabetic nephropathy. Diabetes Res Clin Pract 2008; 82 (Suppl 1): S42-S45, doi: 10.1016/j.diabres.2008.09.017.
https://doi.org/10.1016/j.diabres.2008.0...
,1616. Forbes JM, Coughlan MT, Cooper ME. Oxidative stress as a major culprit in kidney disease in diabetes. Diabetes 2008; 57: 1446-1454, doi: 10.2337/db08-0057.
https://doi.org/10.2337/db08-0057...
) by activating protein kinase C, mitogen-activated protein (MAP) kinases, and transcription factors nuclear factor kappa beta (NF-κB) and activated protein-1. The resulting altered expression of genes and ECM proteins leads to diabetic nephropathy (1717. Kashihara N, Haruna Y, Kondeti VK, Kanwar YS. Oxidative stress in diabetic nephropathy. Curr Med Chem 2010; 17: 4256-4269, doi: 10.2174/092986710793348581.
https://doi.org/10.2174/0929867107933485...
). Mesangial cell proliferation and fibronectin expression are induced by hyperglycemia through the janus kinase (JNK)/NF-κB/NADPH oxidase/ROS signaling pathway (1414. Zhang L, Pang S, Deng B, Qian L, Chen J, Zou J, et al. High glucose induces renal mesangial cell proliferation and fibronectin expression through JNK/NF-kappaB/NADPH oxidase/ROS pathway, which is inhibited by resveratrol. Int J Biochem Cell Biol 2012; 44: 629-638, doi: 10.1016/j.biocel.2012.01.001.
https://doi.org/10.1016/j.biocel.2012.01...
). Resv has been shown to inhibit hyperglycemia-induced mesangial cell expansion and fibronectin expression by blocking this signaling pathway (1414. Zhang L, Pang S, Deng B, Qian L, Chen J, Zou J, et al. High glucose induces renal mesangial cell proliferation and fibronectin expression through JNK/NF-kappaB/NADPH oxidase/ROS pathway, which is inhibited by resveratrol. Int J Biochem Cell Biol 2012; 44: 629-638, doi: 10.1016/j.biocel.2012.01.001.
https://doi.org/10.1016/j.biocel.2012.01...
).

Hyperuricemia, present as a metabolic disorder, is usually associated with gout, kidney disease, hypertension, cardiovascular diseases, inflammation, diabetes and metabolic syndrome. There is increasing evidence that hyperuricemia is an independent risk factor for the occurrence and development of kidney disease, including damage to mesangial cells. At least one study reported that Resv has antihyperuricemic and nephroprotective activity in oxonate-induced mice (1818. Shi YW, Wang CP, Liu L, Liu YL, Wang X, Hong Y, et al. Antihyperuricemic and nephroprotective effects of resveratrol and its analogues in hyperuricemic mice. Mol Nutr Food Res 2012; 56: 1433-1444, doi: 10.1002/mnfr.201100828.
https://doi.org/10.1002/mnfr.201100828...
). These effects, mediated by changes in renal expression of mGLUT9, mABCG2, mOAT1, and mOCT1, are evidence of the possible preventive efficacy of Resv on hyperuricemia (1111. Shi YW, Wang CP, Liu L, Liu YL, Wang X, Hong Y, et al. Antihyperuricemic and nephroprotective effects of resveratrol and its analogues in hyperuricemic mice. Mol Nutr Food Res 2012; 56: 1433-1444, doi: 10.1002/mnfr.201100828.
https://doi.org/10.1002/mnfr.201100828...
,1818. Shi YW, Wang CP, Liu L, Liu YL, Wang X, Hong Y, et al. Antihyperuricemic and nephroprotective effects of resveratrol and its analogues in hyperuricemic mice. Mol Nutr Food Res 2012; 56: 1433-1444, doi: 10.1002/mnfr.201100828.
https://doi.org/10.1002/mnfr.201100828...
). The present study analyzed the effects of Resv on uric acid (UA)-treated mesangial cells as well as effects on the renin-angiotensin system (RAS) and the endothelin system.

Material and Methods

Cell culture

Immortalized human mesangial cells (ihMCs), kindly provided by Dr. Richard Banas (Munich, Germany), were grown in Dulbecco's modified eagle medium (DMEM; Gibco, USA) with the addition of 10% fetal bovine serum (FBS; Gibco), 24 mM NaHCO3 (Merck, USA), 10 mM HEPES (Sigma, USA) and 10,000 U/L penicillin/streptomycin (Gibco). The cultures were incubated at 37°C in a humidified atmosphere containing 5% carbon dioxide. When semiconfluent, the cells were detached from the plastic flasks with trypsin (0.5%; Cultilab, Brazil), centrifuged for 5 min at 1000 g, and then resuspended in DMEM and subcultured in 22-cm2 plastic culture flasks for further experimental procedures.

Resv preparation

Resv of 99% purity (Sigma) was dissolved in DMSO (Merck), and stored at -20°C in the dark.

Uric acid preparation

UA (Sigma) was sterilized in a steam autoclave and added to culture medium without FBS. The UA crystals were suspended in DMEM containing 1% FBS, incubated at 37°C, and sonicated for 15 min to solubilize the crystals.

Exposure of the ihMCs to uric acid

Prior to the experiments, ihMCs were transferred to a plastic plate (1×106 cells/plate) and maintained under culture conditions for 3 days. At confluence, the cells were exposed for 6 or 12 h to either DMEM, without FBS, for the control or DMEM containing 10 mg/dL UA.

RNA isolation, reverse transcription and quantitative real-time PCR

Total RNA was isolated and purified from ihMCs by a phenol and guanidine isothiocyanate-cesium chloride method, using the appropriate kit (Trizol; Life Technologies, USA). Two micrograms of total RNA were treated with DNase (RQ1 RNase-Free DNase; Promega, USA) to prevent genomic DNA contamination. The RNA pellet was resuspended in RNase-free water and was reverse transcribed into cDNA by the addition of 0.5 mg/mL oligo d(T), 10 mM dithiothreitol (DTT), 0.5 mM dNTPs (GE Healthcare, UK), and 200 U reverse transcriptase enzyme (SuperScript RT, Gibco, USA).

Real-time amplification was carried out with a 7500 Real-Time Sequence Detection System (SDS; ABI Prism 7500; Applied Biosystems, USA). The real-time PCR product was quantified using an intercalating dye (SYBR Green I; Molecular Probes Inc., USA) that exhibits increased fluorescence upon binding double-stranded DNA.

The relative gene expression was calculated using the conditions at the early stages of the PCR. At this point, the amplification is logarithmic and can thus be correlated to the initial copy number of the gene transcripts. The reactions were cycled 40 times under the conditions previously determined by conventional PCR. The fluorescence for each cycle was quantitatively analyzed using an ABI Prism 7500 SDS (Applied Biosystems). At the end of the PCR reaction, the temperature was increased from 60°C to 95°C at a rate of 2°C/min. During that time, fluorescence was measured every 15 s to construct a melting curve. A nontemplate control was run with each assay.

The PCR was performed with primers specific for pre-pro endothelin-1 (ppET-1), angiotensinogen (AGT), and β-actin. The primer sequences were: ppET-1 sense (5′-GAGAAACCCACTCCCAGTCC-3′) and antisense (5′-GATGTCCAGGTGGCAGAAGT-3′); β-actin, sense (5′-TCACCCACACTGTGCCCATCTACGA-3′) and antisense (5′-CAGCGGAACCGCTCATTGCCAATGG-3′); and AGT sense (5′-ACTTCTCGGTGACTCAAGTGCC-3′) and antisense (5′-GAAAGTGAGACCCTCCACCTTGT-3′). The results of these experiments are reported in each group as the relative expression normalized to the β-actin housekeeping gene. This gene was used as an internal control and is expressed here as a percentage of the control reaction.

Real-time data analysis

The cycle threshold (Ct) values were subtracted from the Ct value for each gene to yield the ΔCt values. These values were used for statistical comparisons. For the graphic representations, the fold variation was determined using the 2-(ΔΔCt) method according to published protocols (1919. Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods 2001; 25: 402-408, doi: 10.1006/meth.2001.1262.
https://doi.org/10.1006/meth.2001.1262...
) and manufacturer recommendations. The fold variations were calculated by determining the difference in the ΔCt values between the chosen reference and the test samples (ΔΔCt value), with subsequent application of the 2-(ΔΔCt) formula.

Enzyme-linked immunosorbent assay (ELISA)

The concentrations of angiotensin II (AII) and endothelin (ET)-1 were assayed in cell cultures treated with UA using a commercially available competitive ELISA (Cayman Chemical, USA). All assays were performed according to the manufacturer's protocols. The absorbance of each sample was determined using an Ultra Microplate (Biotek, USA) and reported as ng/mL and pg/mL.

Measurement of intracellular calcium by flow cytometry

For the cytometric analyses, the cells were cultured to a density of 2×106 cells/well, were pretreated with Resv (12.5 uM) and stimulated with UA (10 mg/dL). The cells were then trypsinized, washed twice with PBS, and resuspended in 0.5 mL isotonic solution. The osmolarity of the isotonic solution was 323±6 mOsm and consisted of 80 nM D-mannitol with 120 mM NaCl, 6 mM KCl, 1 mM MgCl2, 2 mM CaCl2, and 5.4 mM HEPES. The cells were loaded with 2 µM fluo-4/AM (Invitrogen, USA) for 30 min and were analyzed by flow cytometry (BD FACSCanto II, BD Biosciences, USA) at an excitation wavelength of 488 nm and an emission wavelength of 525 nm. The fluorescence intensity of approximately 1×106 labeled cells was measured in each assay, and the data are reported as the median fluorescence intensity in arbitrary units (AU) obtained by averaging at least 3 separate experiments (2020. Adebiyi A. RGS2 regulates urotensin II-induced intracellular Ca2+ elevation and contraction in glomerular mesangial cells. J Cell Physiol 2014; 229: 502-511, doi: 10.1002/jcp.24470.
https://doi.org/10.1002/jcp.24470...
).

Statistical analysis

The data are reported as means±SE. The experimental and control groups were compared via the Student t-test. The significance level for the null hypothesis was set at 5% (P<0.05).

Results

Resv inhibited the increase in [Ca2+]i induced by UA

To study the effect of Resv on mesangial cells, the effect of UA was first analyzed. Treatment of ihMCs with 10 mg/dL UA increased [Ca2+]i (80.60±0.20) compared with the fluorescence at baseline, but the increase in AII was greater than that of [Ca2+]i after UA administration. The effect of UA was inhibited by pre-exposure of mesangial cells to Resv. When the ihMCs were preincubated with 12.5 µM Resv for 1 h, [Ca2+]i, as estimated by fluorescence intensity, was 38.34±0.01 compared to 80.60±0.20 in cells that had not been pretreated (P<0.005; Figure 1).

Figure 1
Intracellular calcium concentration in immortalized human mesangial cells (ihMCs) treated with uric acid (UA) and resveratrol (Resv). Briefly, semi-confluent ihMC cells were detached with trypsin and the fluorescence intensity of intracellular calcium was determined by Fluo-4/AM in the presence of 10 mg/dL UA and 12.5 µM Resv. Data are reported as the mean percentage (±SE). *P<0.001 vs control (CTL); **P<0.005 vs UA (Student t-test).

Resv inhibited the RAS system in the mesangial cells

One of the main regulators of contraction in mesangial cells is the peptide AII; therefore, we evaluated the effect of Resv on UA-induced AII synthesis. There was a significant increase in UA-induced AGT mRNA expression in the ihMCs in a time-dependent manner. Incubation of the ihMCs with 10 mg/dL UA significantly increased AGT mRNA expression at 6 h (12.40±0.92 vs 1.24±0.18 AU for the control, P<0.001) and at 12 h (22.04±0.79 vs 1.24±0.18 AU for the control, P<0.001). When ihMCs were pre-incubated with 12.5 µM Resv for 1 h, the increase in AGT mRNA was not as great at 6 h (12.40±0.92 vs 3.03±1.08 AU for the UA condition) and at 12 h (22.04±0.79 vs 5.36±1.06 AU for the UA condition) (both P <0.001, Figure 2A).

Figure 2
Effect of resveratrol (Resv) treatment on the angiotensinogen mRNA expression (A) and AII protein synthesis (B) in immortalized human mesangial cells (ihMCs) stimulated with uric acid (UA). Cells were seeded onto plates and after 6 and 12 h were exposed to DMEM with 1% FBS containing 10 mg/dL UA, and 12.5 µM Resv. Data are reported as means±SE. *P<0.001 vs control (CTL); **P<0.001 vs UA (Student t-test).

UA increased AII protein synthesis at 6 h (0.07±0.01 vs 0.05±0.01 ng/mL for the control, P<0.05) and 12 h (0.10±0.01 vs 0.05±0.01 ng/mL for the control, P<0.05) Pre-incubation with 12.5 µM Resv for 1 h attenuated the increase in AII protein synthesis at 6 h (0.05±0.01 vs 0.07±0.01 ng/mL for the UA group) and at 12 h (0.04±0.003 vs 0.10±0.01 ng/mL for the UA group) (both P<0.05, Figure 2B).

Resv inhibited the endothelin system in mesangial cells

ET-1 is also a peptide involved in smooth muscle cell contraction (2121. Chuang TY, Au LC, Wang LC, Ho LT, Yang DM, Juan CC. Potential effect of resistin on the ET-1-increased reactions of blood pressure in rats and Ca2+ signaling in vascular smooth muscle cells. J Cell Physiol 2012 ; 227: 1610-8, doi: 10.1002/jcp.22878.
https://doi.org/10.1002/jcp.22878...
). Nevertheless, the effect of UA on ET-1 synthesis in mesangial cells is not known. UA significantly increased ppET-1 mRNA expression in cultured ihMCs in a time-dependent manner. Incubation of the ihMCs with UA increased ppET-1 expression at 6 h (1.52±0.23 vs 1.02±0.10 AU for the control group) and 12 h (2.32±0.18 vs 1.02±0.10 AU for the control group) (both P<0.05, Figure 3A). When the ihMCs were pre-incubated with 12.5 µM Resv for 1 h, the increases were significantly lower at both 6 h (0.77±0.18 vs 1.52±0.23 arbitrary units for the UA group) and 12 h (0.41±0.06 vs 2.32±0.18 arbitrary units for the UA group (both P<0.05, Figure 3A).

Figure 3
Effect of resveratrol (Resv) treatment (12.5 µM) on mRNA pre-pro endothelin-1 (ppET-1) expression (A) and ET-1 protein synthesis (B) in immortalized human mesangial cells (ihMCs) stimulated with uric acid (UA). Cells were seeded on plates and after 6 and 12 h were exposed to DMEM with 1% FBS containing 10 mg/dL UA and 12.5 µM Resv. Data are reported as means±SE. A, *P<0.05 vs control (CTL); **P<0.05 vs UA (Student t-test). B, *P<0.05 vs CTL; **P<0.001 vs UA (Student t-test).

UA increased ET-1 protein synthesis at 6 h (32.69±0.22 vs 29.68±0.35 pg/mL for the control group) and 12 h (37.29±1.23 vs 29.68±0.35 pg/mL for the control group (both P<0.05, Figure 3B).

Again, when the ihMCs were pre-incubated with 12.5 µM Resv for 1 h, the increases were significantly lower in ET-1 protein synthesis at 6 h (27.78±1.38 vs 32.69±0.22 pg/mL for the UA group) and at 12 h (29.92±1.02 vs 37.29±1.23 pg/mL for the UA group (all P<0.05, Figure 3B).

Discussion

Mesangial cells play a major role in glomerular contraction and regulate filtration surface area and ultrafiltration coefficient (Kf) through the release of hormones such as AII. Mesangial cells express all components of RAS, and can regulate the Kf via contraction or the release of vasoactive hormones (2222. Sorokin A. Endothelin signaling and actions in the renal mesangium. Contrib Nephrol 2011; 172: 50-62, doi: 10.1159/000328680.
https://doi.org/10.1159/000328680...
). ETs are also powerful vasoconstrictor peptides, and act as modulators of vasomotor tone, cell proliferation, and hormone production. Mesangial cells are the main site of ET production in the kidney (2323. Woodward OM, Kottgen A, Coresh J, Boerwinkle E, Guggino WB, Kottgen M. Identification of a urate transporter, ABCG2, with a common functional polymorphism causing gout. Proc Natl Acad Sci U S A 2009; 106: 10338-10342, doi: 10.1073/pnas.0901249106.
https://doi.org/10.1073/pnas.0901249106...
).

UA has recently been recognized as a key factor in multifactorial renal disorders, including chronic kidney disease, hypertension and acute kidney injury (2424. McElnea EM, Quill B, Docherty NG, Irnaten M, Siah WF, Clark AF, et al. Oxidative stress, mitochondrial dysfunction and calcium overload in human lamina cribrosa cells from glaucoma donors. Mol Vis 2011; 17: 1182-1191.,2525. Pasalic D, Marinkovic N, Feher-Turkovic L. Uric acid as one of the important factors in multifactorial disorders - facts and controversies. Biochem Med 2012; 22: 63-75, doi: 10.11613/BM.2012.007.
https://doi.org/10.11613/BM.2012.007...
). Recent studies have confirmed that RAS is strongly related to elevated serum UA levels in hypertensive patients. We have previously demonstrated that UA stimulates RAS and ET-1 in mesangial cells after 24 h (2626. Albertoni G, Maquigussa E, Pessoa E, Barreto JA, Borges F, Schor N. Soluble uric acid increases intracellular calcium through an angiotensin II-dependent mechanism in immortalized human mesangial cells. Exp Biol Med 2010; 235: 825-832, doi: 10.1258/ebm.2010.010007.
https://doi.org/10.1258/ebm.2010.010007...
). Experimental studies in vitro have also demonstrated that UA has direct effects on rat vascular smooth cell, proliferation (2727. Corry DB, Eslami P, Yamamoto K, Nyby MD, Makino H, Tuck ML. Uric acid stimulates vascular smooth muscle cell proliferation and oxidative stress via the vascular renin-angiotensin system. J Hypertens 2008; 26: 269-275, doi: 10.1097/HJH.0b013e3282f240bf.
https://doi.org/10.1097/HJH.0b013e3282f2...
), human vascular endothelial cell dysfunction (2828. Yu MA, Sanchez-Lozada LG, Johnson RJ, Kang DH. Oxidative stress with an activation of the renin-angiotensin system in human vascular endothelial cells as a novel mechanism of uric acid-induced endothelial dysfunction. J Hypertens 2010; 28: 1234-1242.), and ihMC proliferation (2626. Albertoni G, Maquigussa E, Pessoa E, Barreto JA, Borges F, Schor N. Soluble uric acid increases intracellular calcium through an angiotensin II-dependent mechanism in immortalized human mesangial cells. Exp Biol Med 2010; 235: 825-832, doi: 10.1258/ebm.2010.010007.
https://doi.org/10.1258/ebm.2010.010007...
) via local tissue RAS activation. This evidence indicates that the activation of systemic and local tissue RAS by UA may be partially responsible for the pathogenetic effects of UA in hypertension.

The experimental choice of 10 mg/d UA was determined by the saturation concentration of monosodium urate in human plasma of approximately 7 mg/dL. Thus, the definition of hyperuricemia is usually a plasma UA concentration >7 mg/dL (2929. Baldree LA, Stapleton FB. Uric acid metabolism in children. Pediatr Clin North Am 1990; 37: 391-418.). In addition, Aida et al. (3030. Aida Y, Shibata Y, Osaka D, Abe S, Inoue S, Fukuzaki K, et al. The relationship between serum uric acid and spirometric values in participants in a health check: the Takahata study. Int J Med Sci 2011; 8: 470-478, doi: 10.7150/ijms.8.470.
https://doi.org/10.7150/ijms.8.470...
) reported that the plasma concentration of UA in the general population has a wide range, extending from 1 to 12 mg/dL. Observations in an American cohort were confirmed in a community-based study with participants in Vienna that reported mild hyperuricemia in patients with uric acid concentrations of 7.0-8.9 mg/dL (3131. Obermayr RP, Temml C, Knechtelsdorfer M, Gutjahr G, Kletzmayr J, Heiss S, et al. Predictors of new-onset decline in kidney function in a general middle-european population. Nephrol Dial Transplant 2008; 23: 1265-1273, doi: 10.1093/ndt/gfm790.
https://doi.org/10.1093/ndt/gfm790...
). High UA concentrations (5 or 15 mg/dL) upregulated both RAS mRNA expression and AII II protein secretion in 3T3-L1 adipocytes. In addition, UA caused a significant increase in ROS production in differentiated adipocytes (3232. Zhang JX, Zhang YP, Wu QN, Chen B. Uric acid induces oxidative stress via an activation of the renin-angiotensin system in 3T3-L1 adipocytes. Endocrine 2014.).

Evidence from studies indicates that the generation of ROS is, at least in part, involved in endothelial dysfunction (3333. Doughan AK, Harrison DG, Dikalov SI. Molecular mechanisms of angiotensin II-mediated mitochondrial dysfunction: linking mitochondrial oxidative damage and vascular endothelial dysfunction. Circ Res 2008; 102: 488-496, doi: 10.1161/CIRCRESAHA.107.162800.
https://doi.org/10.1161/CIRCRESAHA.107.1...
,3434. Wassmann S, Stumpf M, Strehlow K, Schmid A, Schieffer B, Bohm M, et al. Interleukin-6 induces oxidative stress and endothelial dysfunction by overexpression of the angiotensin II type 1 receptor. Circ Res 2004; 94: 534-541, doi: 10.1161/01.RES.0000115557.25127.8D.
https://doi.org/10.1161/01.RES.000011555...
), and the production of ROS, particularly the superoxide anion (O2 -), is closely related to calcium concentration (3535. Victor VM, Apostolova N, Herance R, Hernandez-Mijares A, Rocha M. Oxidative stress and mitochondrial dysfunction in atherosclerosis: mitochondria-targeted antioxidants as potential therapy. Curr Med Chem 2009; 16: 4654-4667, doi: 10.2174/092986709789878265.
https://doi.org/10.2174/0929867097898782...
).

The concentration of Resv (12.5 µM) was chosen from among various concentrations that are known to have relevant cardiovascular effects on isolated tissues or organs (3636. Opie LH, Lecour S. The red wine hypothesis: from concepts to protective signalling molecules. Eur Heart J 2007; 28: 1683-1693, doi: 10.1093/eurheartj/ehm149.
https://doi.org/10.1093/eurheartj/ehm149...
). The first evidence of the specific benefits of red wine was shown in 1993 when Frankel et al. (3737. Frankel EN, Kanner J, German JB, Parks E, Kinsella JE. Inhibition of oxidation of human low-density lipoprotein by phenolic substances in red wine. Lancet 1993; 341: 454-457, doi: 10.1016/0140-6736(93)90206-V.
https://doi.org/10.1016/0140-6736(93)902...
) demonstrated that red wine diluted 1000-fold and containing 10 µmol of total phenols inhibited the oxidation of LDL. This is approximately the Resv concentration found in red wines, and suggested that half a bottle of such wine should contain an active concentration of resveratrol (3737. Frankel EN, Kanner J, German JB, Parks E, Kinsella JE. Inhibition of oxidation of human low-density lipoprotein by phenolic substances in red wine. Lancet 1993; 341: 454-457, doi: 10.1016/0140-6736(93)90206-V.
https://doi.org/10.1016/0140-6736(93)902...
).

In a study similar to ours, Chao et al. (3838. Chao HH, Juan SH, Liu JC, Yang HY, Yang E, Cheng TH, et al. Resveratrol inhibits angiotensin II-induced endothelin-1 gene expression and subsequent proliferation in rat aortic smooth muscle cells. Eur J Pharmacol 2005; 515: 1-9, doi: 10.1016/j.ejphar.2005.03.035.
https://doi.org/10.1016/j.ejphar.2005.03...
) found that Resv (10 µM) inhibited the formation of AII-induced ROS, extracellular signal-regulated kinase phosphorylation, ET-1 gene expression, and cell proliferation in smooth muscle cells.

In this study, UA induced an increase in ppET-1 mRNA expression and protein synthesis after 6 and 12 h and increased AGT mRNA expression and AII protein synthesis after 6 and 12 h. In addition, Resv reduced UA-induced pre-proET-1 gene expression and the production of AII and ET-1 in mesangial cells, suggesting that it can minimize the impact of these hormones on glomerular function. In addition, Resv inhibited the increase in [Ca2+]i. These results are the first direct evidence that UA induces an increase in [Ca2+]i that is minimized by Resv. Our results suggested that UA triggers reactions including AII and ET-1 production in mesangial cells. RAS may contribute to the pathogenesis of renal function and chronic kidney disease. Resv can minimize the impact of UA on the increases of AII, ET-1 and [Ca2+]i in mesangial cells, suggesting that it can prevent the effects of soluble UA on mesangial cells.

Lastly, multidisciplinary approaches are recommended for future investigations because of the wide range of polyphenol actions on body fat reduction, mitigation of liver disease, improvements in muscle function, and cardiovascular and renal protection.

Acknowledgments

Research supported by CNPq, FINEP, FAPESP, CAPES, and Fundação Oswaldo Ramos (FOR).

References

  • 1
    Scott E, Steward WP, Gescher AJ, Brown K. Resveratrol in human cancer chemoprevention - choosing the ‘right’ dose. Mol Nutr Food Res 2012; 56: 7-13, doi: 10.1002/mnfr.201100400.
    » https://doi.org/10.1002/mnfr.201100400
  • 2
    Roupe KA, Remsberg CM, Yanez JA, Davies NM. Pharmacometrics of stilbenes: seguing towards the clinic. Curr Clin Pharmacol 2006; 1: 81-101, doi: 10.2174/157488406775268246.
    » https://doi.org/10.2174/157488406775268246
  • 3
    Smoliga JM, Baur JA, Hausenblas HA. Resveratrol and health - a comprehensive review of human clinical trials. Mol Nutr Food Res 2011; 55: 1129-1141, doi: 10.1002/mnfr.201100143.
    » https://doi.org/10.1002/mnfr.201100143
  • 4
    Delmas D, Aires V, Limagne E, Dutartre P, Mazue F, Ghiringhelli F, et al. Transport, stability, and biological activity of resveratrol. Ann N Y Acad Sci 2011; 1215: 48-59, doi: 10.1111/j.1749-6632.2010.05871.x.
    » https://doi.org/10.1111/j.1749-6632.2010.05871.x
  • 5
    Timmers S, Konings E, Bilet L, Houtkooper RH, van de Weijer T, Goossens GH, et al. Calorie restriction-like effects of 30 days of resveratrol supplementation on energy metabolism and metabolic profile in obese humans. Cell Metab 2011; 14: 612-622, doi: 10.1016/j.cmet.2011.10.002.
    » https://doi.org/10.1016/j.cmet.2011.10.002
  • 6
    Brasnyo P, Molnar GA, Mohas M, Marko L, Laczy B, Cseh J, et al. Resveratrol improves insulin sensitivity, reduces oxidative stress and activates the Akt pathway in type 2 diabetic patients. Br J Nutr 2011; 106: 383-389, doi: 10.1017/S0007114511000316.
    » https://doi.org/10.1017/S0007114511000316
  • 7
    Magyar K, Halmosi R, Palfi A, Feher G, Czopf L, Fulop A, et al. Cardioprotection by resveratrol: A human clinical trial in patients with stable coronary artery disease. Clin Hemorheol Microcirc 2012; 50: 179-187.
  • 8
    Kondratyuk TP, Park EJ, Marler LE, Ahn S, Yuan Y, Choi Y, et al. Resveratrol derivatives as promising chemopreventive agents with improved potency and selectivity. Mol Nutr Food Res 2011; 55: 1249-1265, doi: 10.1002/mnfr.201100122.
    » https://doi.org/10.1002/mnfr.201100122
  • 9
    Matsuoka A, Kodama Y, Fukuhara K, Honda S, Hayashi M, Sai K, et al. A pilot study of evaluation of the antioxidative activity of resveratrol and its analogue in a 6-month feeding test in young adult mice. Food Chem Toxicol 2008; 46: 1125-1130, doi: 10.1016/j.fct.2007.11.008.
    » https://doi.org/10.1016/j.fct.2007.11.008
  • 10
    Kim MY, Lim JH, Youn HH, Hong YA, Yang KS, Park HS, et al. Resveratrol prevents renal lipotoxicity and inhibits mesangial cell glucotoxicity in a manner dependent on the AMPK-SIRT1-PGC1alpha axis in db/db mice. Diabetologia 2013; 56: 204-217, doi: 10.1007/s00125-012-2747-2.
    » https://doi.org/10.1007/s00125-012-2747-2
  • 11
    Shi YW, Wang CP, Liu L, Liu YL, Wang X, Hong Y, et al. Antihyperuricemic and nephroprotective effects of resveratrol and its analogues in hyperuricemic mice. Mol Nutr Food Res 2012; 56: 1433-1444, doi: 10.1002/mnfr.201100828.
    » https://doi.org/10.1002/mnfr.201100828
  • 12
    Khan SR. Hyperoxaluria-induced oxidative stress and antioxidants for renal protection. Urol Res 2005; 33: 349-357, doi: 10.1007/s00240-005-0492-4.
    » https://doi.org/10.1007/s00240-005-0492-4
  • 13
    Hong SH, Lee HJ, Sohn EJ, Ko HS, Shim BS, Ahn KS, et al. Anti-nephrolithic potential of resveratrol via inhibition of ROS, MCP-1, hyaluronan and osteopontin in vitro and in vivo Pharmacol Rep 2013; 65: 970-979, doi: 10.1016/S1734-1140(13)71078-8.
    » https://doi.org/10.1016/S1734-1140(13)71078-8
  • 14
    Zhang L, Pang S, Deng B, Qian L, Chen J, Zou J, et al. High glucose induces renal mesangial cell proliferation and fibronectin expression through JNK/NF-kappaB/NADPH oxidase/ROS pathway, which is inhibited by resveratrol. Int J Biochem Cell Biol 2012; 44: 629-638, doi: 10.1016/j.biocel.2012.01.001.
    » https://doi.org/10.1016/j.biocel.2012.01.001
  • 15
    Ha H, Hwang IA, Park JH, Lee HB. Role of reactive oxygen species in the pathogenesis of diabetic nephropathy. Diabetes Res Clin Pract 2008; 82 (Suppl 1): S42-S45, doi: 10.1016/j.diabres.2008.09.017.
    » https://doi.org/10.1016/j.diabres.2008.09.017
  • 16
    Forbes JM, Coughlan MT, Cooper ME. Oxidative stress as a major culprit in kidney disease in diabetes. Diabetes 2008; 57: 1446-1454, doi: 10.2337/db08-0057.
    » https://doi.org/10.2337/db08-0057
  • 17
    Kashihara N, Haruna Y, Kondeti VK, Kanwar YS. Oxidative stress in diabetic nephropathy. Curr Med Chem 2010; 17: 4256-4269, doi: 10.2174/092986710793348581.
    » https://doi.org/10.2174/092986710793348581
  • 18
    Shi YW, Wang CP, Liu L, Liu YL, Wang X, Hong Y, et al. Antihyperuricemic and nephroprotective effects of resveratrol and its analogues in hyperuricemic mice. Mol Nutr Food Res 2012; 56: 1433-1444, doi: 10.1002/mnfr.201100828.
    » https://doi.org/10.1002/mnfr.201100828
  • 19
    Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods 2001; 25: 402-408, doi: 10.1006/meth.2001.1262.
    » https://doi.org/10.1006/meth.2001.1262
  • 20
    Adebiyi A. RGS2 regulates urotensin II-induced intracellular Ca2+ elevation and contraction in glomerular mesangial cells. J Cell Physiol 2014; 229: 502-511, doi: 10.1002/jcp.24470.
    » https://doi.org/10.1002/jcp.24470
  • 21
    Chuang TY, Au LC, Wang LC, Ho LT, Yang DM, Juan CC. Potential effect of resistin on the ET-1-increased reactions of blood pressure in rats and Ca2+ signaling in vascular smooth muscle cells. J Cell Physiol 2012 ; 227: 1610-8, doi: 10.1002/jcp.22878.
    » https://doi.org/10.1002/jcp.22878
  • 22
    Sorokin A. Endothelin signaling and actions in the renal mesangium. Contrib Nephrol 2011; 172: 50-62, doi: 10.1159/000328680.
    » https://doi.org/10.1159/000328680
  • 23
    Woodward OM, Kottgen A, Coresh J, Boerwinkle E, Guggino WB, Kottgen M. Identification of a urate transporter, ABCG2, with a common functional polymorphism causing gout. Proc Natl Acad Sci U S A 2009; 106: 10338-10342, doi: 10.1073/pnas.0901249106.
    » https://doi.org/10.1073/pnas.0901249106
  • 24
    McElnea EM, Quill B, Docherty NG, Irnaten M, Siah WF, Clark AF, et al. Oxidative stress, mitochondrial dysfunction and calcium overload in human lamina cribrosa cells from glaucoma donors. Mol Vis 2011; 17: 1182-1191.
  • 25
    Pasalic D, Marinkovic N, Feher-Turkovic L. Uric acid as one of the important factors in multifactorial disorders - facts and controversies. Biochem Med 2012; 22: 63-75, doi: 10.11613/BM.2012.007.
    » https://doi.org/10.11613/BM.2012.007
  • 26
    Albertoni G, Maquigussa E, Pessoa E, Barreto JA, Borges F, Schor N. Soluble uric acid increases intracellular calcium through an angiotensin II-dependent mechanism in immortalized human mesangial cells. Exp Biol Med 2010; 235: 825-832, doi: 10.1258/ebm.2010.010007.
    » https://doi.org/10.1258/ebm.2010.010007
  • 27
    Corry DB, Eslami P, Yamamoto K, Nyby MD, Makino H, Tuck ML. Uric acid stimulates vascular smooth muscle cell proliferation and oxidative stress via the vascular renin-angiotensin system. J Hypertens 2008; 26: 269-275, doi: 10.1097/HJH.0b013e3282f240bf.
    » https://doi.org/10.1097/HJH.0b013e3282f240bf
  • 28
    Yu MA, Sanchez-Lozada LG, Johnson RJ, Kang DH. Oxidative stress with an activation of the renin-angiotensin system in human vascular endothelial cells as a novel mechanism of uric acid-induced endothelial dysfunction. J Hypertens 2010; 28: 1234-1242.
  • 29
    Baldree LA, Stapleton FB. Uric acid metabolism in children. Pediatr Clin North Am 1990; 37: 391-418.
  • 30
    Aida Y, Shibata Y, Osaka D, Abe S, Inoue S, Fukuzaki K, et al. The relationship between serum uric acid and spirometric values in participants in a health check: the Takahata study. Int J Med Sci 2011; 8: 470-478, doi: 10.7150/ijms.8.470.
    » https://doi.org/10.7150/ijms.8.470
  • 31
    Obermayr RP, Temml C, Knechtelsdorfer M, Gutjahr G, Kletzmayr J, Heiss S, et al. Predictors of new-onset decline in kidney function in a general middle-european population. Nephrol Dial Transplant 2008; 23: 1265-1273, doi: 10.1093/ndt/gfm790.
    » https://doi.org/10.1093/ndt/gfm790
  • 32
    Zhang JX, Zhang YP, Wu QN, Chen B. Uric acid induces oxidative stress via an activation of the renin-angiotensin system in 3T3-L1 adipocytes. Endocrine 2014.
  • 33
    Doughan AK, Harrison DG, Dikalov SI. Molecular mechanisms of angiotensin II-mediated mitochondrial dysfunction: linking mitochondrial oxidative damage and vascular endothelial dysfunction. Circ Res 2008; 102: 488-496, doi: 10.1161/CIRCRESAHA.107.162800.
    » https://doi.org/10.1161/CIRCRESAHA.107.162800
  • 34
    Wassmann S, Stumpf M, Strehlow K, Schmid A, Schieffer B, Bohm M, et al. Interleukin-6 induces oxidative stress and endothelial dysfunction by overexpression of the angiotensin II type 1 receptor. Circ Res 2004; 94: 534-541, doi: 10.1161/01.RES.0000115557.25127.8D.
    » https://doi.org/10.1161/01.RES.0000115557.25127.8D
  • 35
    Victor VM, Apostolova N, Herance R, Hernandez-Mijares A, Rocha M. Oxidative stress and mitochondrial dysfunction in atherosclerosis: mitochondria-targeted antioxidants as potential therapy. Curr Med Chem 2009; 16: 4654-4667, doi: 10.2174/092986709789878265.
    » https://doi.org/10.2174/092986709789878265
  • 36
    Opie LH, Lecour S. The red wine hypothesis: from concepts to protective signalling molecules. Eur Heart J 2007; 28: 1683-1693, doi: 10.1093/eurheartj/ehm149.
    » https://doi.org/10.1093/eurheartj/ehm149
  • 37
    Frankel EN, Kanner J, German JB, Parks E, Kinsella JE. Inhibition of oxidation of human low-density lipoprotein by phenolic substances in red wine. Lancet 1993; 341: 454-457, doi: 10.1016/0140-6736(93)90206-V.
    » https://doi.org/10.1016/0140-6736(93)90206-V
  • 38
    Chao HH, Juan SH, Liu JC, Yang HY, Yang E, Cheng TH, et al. Resveratrol inhibits angiotensin II-induced endothelin-1 gene expression and subsequent proliferation in rat aortic smooth muscle cells. Eur J Pharmacol 2005; 515: 1-9, doi: 10.1016/j.ejphar.2005.03.035.
    » https://doi.org/10.1016/j.ejphar.2005.03.035
  • First published online.

Publication Dates

  • Publication in this collection
    24 Oct 2014
  • Date of issue
    Jan 2015

History

  • Received
    12 Apr 2014
  • Accepted
    10 Sept 2014
Associação Brasileira de Divulgação Científica Av. Bandeirantes, 3900, 14049-900 Ribeirão Preto SP Brazil, Tel. / Fax: +55 16 3315-9120 - Ribeirão Preto - SP - Brazil
E-mail: bjournal@terra.com.br