Open Access

Effects of uric acid on oxidative stress in vascular smooth muscle cells

  • Authors:
    • Segun Dogru
    • Ekrem Yasar
    • Akin Yesilkaya
  • View Affiliations

  • Published online on: September 18, 2024     https://doi.org/10.3892/br.2024.1859
  • Article Number: 171
  • Copyright: © Dogru et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Hyperuricemia during hypertension is associated with aberrant vascular functions and increased oxidative stress, which affects endothelial dysfunction. Nevertheless, the molecular mechanisms underlying the effects of uric acid on vascular smooth muscle cells (VSMCs) through oxidative stress remain unclear. The aim of the present study was to investigate the dose‑ and time‑dependent effects of uric acid on oxidative stress and p53 protein expression in VSMCs. VSMCs were incubated with various concentrations of uric acid (0‑50 mg/dl) for different time periods (1‑24 h). Thiobarbituric acid reactive substances (TBARs), protein carbonylation and nitric oxide (NO) levels were determined using appropriate assay kits. Superoxide anion release was detected using the Görlach method. Western blotting was performed to determine the protein expression levels of p53. The findings demonstrated that the application of uric acid led to an increase in protein carbonylation and superoxide anion levels while causing a decrease in NO levels. Conversely, no significant effect was observed on TBARS levels. Additionally, it was observed that high concentrations of uric acid suppressed p53 expression at 6, 12 and 24 h. The present study provided evidence that the influence of uric acid on oxidative stress was more closely associated with time than dose; however, not all effects observed were strictly time‑dependent.

Introduction

Uric acid is the end product of purine metabolism in humans and higher primates, which is mainly synthesized in the liver and is predominantly excreted by the kidneys. Under normal physiological conditions, serum uric acid concentrations range from 3.5 to 7.2 mg/dl in men, and from 2.6 to 6.0 mg/l in women; these levels are maintained through a tightly regulated balance between uric acid production and excretion (1). When this balance is disrupted, leading to elevated levels of uric acid in the blood, hyperuricemia occurs (2,3). Clinically, hyperuricemia is defined as serum uric acid concentrations ≥7 mg/dl (0.42 mmol/l) at physiological temperature and pH (4).

Chronic hyperuricemia is implicated in the development of various pathophysiological conditions, including hypertension, chronic kidney disease, cardiovascular disease, nonalcoholic fatty liver disease, metabolic syndrome and diabetes (5,6). The association between hyperuricemia and these conditions is well known, making it a significant risk factor for cardiovascular and renal diseases. Despite this, the precise serum uric acid concentration at which the risk begins to increase remains unclear. Emerging evidence has suggested that the threshold for increased risk may be lower than previously considered, potentially <6 mg/dl (5). Consequently, this issue continues to be a focal point for clinical research, as understanding the exact uric acid levels that pose a risk is crucial for developing effective treatment and prevention strategies (5-7). Uric acid can interact with reactive oxygen species (ROS), such as hydroxyl radicals and hypochlorous acid, transforming them into less harmful substances such as allantoin, allantoate, glyoxylate, urea and oxalate (8). This antioxidant capability has been demonstrated in various experimental studies, both in vitro and in vivo, including studies on isolated organs and the human lung (9). Moreover, uric acid serves as an oxidizable co-substrate for cyclooxygenase enzymes, thereby contributing to the reduction of oxidative stress and the maintenance of endothelial function, particularly in coronary circulation (10). Despite its beneficial antioxidant properties, elevated uric acid levels, or hyperuricemia, are linked to numerous adverse health conditions.

Epidemiological studies have consistently identified hyperuricemia as a key contributor to hypertension (11-14). Experimental research in a rat model has shown that dietary supplementation with oxonic acid can lead to increased plasma uric acid levels and subsequent increased blood pressure (15-17). Additionally, hyperuricemia has been associated with endothelial dysfunction, impaired vasodilation and vascular injury, which are precursors to cardiovascular diseases, including hypertension and atherosclerosis (18-21). These findings highlight the complex role of uric acid in vascular biology, where its antioxidant functions are unclear due to its potential to contribute to vascular damage under hyperuricemic conditions. The exacerbation of endothelial dysfunction and vascular injury in the presence of elevated uric acid levels suggests a critical need to understand the underlying molecular mechanisms. Further research is needed to identify targeted therapeutic approaches that potentially reduce the deleterious effects of hyperuricemia.

Vascular smooth muscle cells (VSMCs) and endothelial cells (ECs), are integral components of blood vessels, serving crucial roles in vasoconstriction, vascular tone regulation and the development of vasculature (22). Nitric oxide (NO), a key cellular signaling molecule involved in both physiological and pathological processes, mediates vasodilation. Under normal conditions, NO is synthesized by endothelial NO synthase in ECs, diffuses to adjacent VSMCs and induces vasodilation (22-24). However, under oxidative stress, NO reacts with superoxide anions to form peroxynitrite (25,26). Additionally, uric acid leads to a reduction in NO levels because of the formation of peroxynitrite in vascular cells (27,28). Peroxynitrite is a potent oxidant known to cause oxidative damage and nitrosative stress to macromolecules, such as proteins, lipids and DNA (25,29,30). Previous studies have demonstrated that elevated uric acid levels decrease NO production in human umbilical vein ECs (HUVECs) (31-34). Furthermore, it has previously been indicated that uric acid increases oxidative stress, which triggers a phenotypic transition in vascular ECs (21). While the deleterious effects of uric acid-induced NO depletion and increased oxidative stress on EC function have been demonstrated (20,35-37), emerging evidence has suggested that high uric acid conditions may also affect VSMCs (38,39). However, the specific impact of uric acid on NO levels in VSMCs remains elusive. Further studies are required to elucidate the mechanisms through which uric acid influences NO bioavailability in VSMCs, and to understand the implications for vascular health and disease.

The p53 gene is widely recognized as a tumor suppressor gene, playing a pivotal role in regulating key cellular processes, including cell cycle control, DNA repair, cell proliferation, apoptosis, aging and oxidative stress response, which are associated with diseases such as diabetes, cancer and hypertension (40,41). In VSMCs, p53 has been shown to promote senescence and apoptosis, and is actively involved in the pathogenesis of atherosclerotic plaques (42). However, there are conflicting reports in the literature regarding the role of p53 in VSMCs. Recent data have revealed that p53 deficiency reduces VSMCs senescence and calcification, and knockdown of p53 can decrease mitochondrial ROS (43), suggesting that it protects VSMCs against oxidative stress. Moreover, p53 has been shown to protect VSMCs from NO-mediated oxidative stress (44). Previous findings have also revealed that p53 can be directly targeted by uric acid (45). To the best of our knowledge, although numerous aspects of uric acid-induced cell proliferation (38,39,46), inflammation (47,48) and oxidative stress (38,48) have been studied in VSMCs, the effect of uric acid on p53 has not yet been demonstrated. The interplay between uric acid and p53 in VSMCs is of particular interest, given the potential implications for understanding how uric acid influences cellular senescence, oxidative stress and vascular pathophysiology.

In the present study, it was hypothesized that uric acid could elevate oxidative stress in rat VSMCs in a dose- and time-dependent manner. To investigate this hypothesis, the effects of various concentrations of uric acid on oxidative stress markers, including protein carbonylation, thiobarbituric acid reactive substances (TBARs) and superoxide anion levels, were examined. Additionally, the protein expression levels of p53 and NO levels were assessed in rat VSMCs. By comparing these parameters, the present study aimed to understand the association between uric acid exposure and oxidative stress, as well as its impact on p53 expression and NO levels over time and across different doses. The present findings provide preliminary novel insights into oxidative stress responses induced by uric acid and highlight the potential role of uric acid in mediating vascular cell function.

Materials and methods

Reagents and antibodies

Uric acid, fetal bovine serum (FBS), penicillin-streptomycin, HEPES, elastase, collagenase, cytochrome c, superoxide dismutase (SOD) and bovine serum albumin (BSA) were purchased from Sigma-Aldrich (Merck KGaA). DMEM (cat. no. E0500-100) was obtained from Cegrogen Biotech GmbH and Hank's balanced salt solution (HBSS) was obtained from Biochrom, Ltd. Nitrate/Nitrite (NO detection) Colorimetric Assay Kit (cat. no. 780001), TBARS Assay Kit (cat. no. 10009055) and Protein Carbonyl Assay Kit (cat. no. 10005020) were purchased from Cayman Chemical Company. Primary antibodies against p53 (1C12) (cat. no. 2524) and β-actin (cat. no. 4967), as well as secondary antibodies [horseradish peroxidase (HRP)-linked goat anti-rabbit immunoglobulin G (IgG) (cat. no. 7074) and anti-mouse IgG (cat. no. 7076)] were purchased from Cell Signaling Technology, Inc.

Isolation and culture of primary rat vascular smooth muscle cells

All animal experiments were performed according to the Guide for the Care and Use of Laboratory Animals (49) following experimental protocols approved by the Local Committee on Animal Research Ethics at Akdeniz University (approval no. 727/2018.01.024; Antalya, Turkey). For the present study, a total of 4 male Wistar rats (age, 8-10 weeks; weight, 200-300 g) were used. The male Wistar rats were obtained from the Local Committee on Animal Research Ethics at Akdeniz University. The rats were housed in a controlled environment with a temperature of 22±2˚C, a relative humidity of 50±10% and a 12-h light/dark cycle. The rats had ad libitum access to standard chow and water. According to the latest guidelines from the American Veterinary Medical Association (50) and guidelines adopted by institutions such as Boston University (51) and the University of Maryland (52), the rats were anesthetized by intraperitoneal injection of ketamine (80 mg/kg) and xylazine (10 mg/kg), in compliance with approved ethical standards (53,54). Upon confirmation of deep anesthesia, which was verified by the lack of response to painful stimuli and absence of the corneal reflex, the chest cavity was exposed to allow access to the aorta. The aorta was then carefully dissected, the adventitial layer was meticulously removed using forceps, and the tissue was subsequently transferred to cell culture dishes under sterile conditions. VSMCs were isolated from the dissected aorta using enzymatic dissociation solution [HEPES dissolved in HBSS (15 mM, pH: 7.2-7.3), BSA (2 mg/ml), CaCl2 (0.2 mM), Soybean Trypsin Inhibitor (0.25 mg/ml), elastase (0.0625 mg/ml) and collagenase (0.5 mg/ml)] (39). After the isolation of VSMCs, while the rats were still under deep anesthesia, they were sacrificed by cervical dislocation to ensure a humane and painless death. The isolated cells were subsequently transferred to cell culture dishes and cultured as previously described (39,47,55). VSMCs were maintained in DMEM supplemented with 10% FBS and 1% penicillin-streptomycin, and were cultured at 37˚C and 5% CO2. VSMCs in passages 3 to 5 were used in all experiments and the cells were incubated up to 70-80% density. After the control and experimental groups were formed, all cells were incubated with FBS-free DMEM overnight for serum starvation. Uric acid was prepared by filtration through 0.2-mm sterile filters and VSMCs were stimulated at 37˚C with different uric acid doses (0-50 mg/dl) for various durations (1-24 h), excluding the control groups.

NO determination

Once cells reached 80% confluence, NO levels were detected. NO (total nitrate + nitrite) levels were determined using a colorimetric assay kit that included Griess reagents (Griess 1 and 2), according to the manufacturer's instructions. NO content was determined as the total value measured in the presence of cells minus the value determined from the media alone in the absence of cell growth, according to the manufacturer's protocol. Each sample absorbance was measured by spectrophotometry (540 nm) and NO concentrations were calculated using a standard curve. The levels of NO are shown in µM. Experiments were repeated four times and the results are presented as the mean ± standard error of the mean (SEM).

Superoxide anion accumulation levels

For the determination of superoxide anion production, after reaching 80% cell confluence, the Görlach method of spectrophotometric SOD-inhibitable reduction of cytochrome c was performed for each sample, with a blank for each one (negative control), as previously described (47,56). VSMCs were cultured in 12-well plates for 48 h. Superoxide anion related to cytochrome c reduction was calculated for each sample by measuring between cells incubated with SOD and without SOD. Superoxide anion accumulation levels are shown in nmol/µg. Experiments were repeated five times and the results are presented as the mean ± SEM.

Quantification of TBARS levels

Lipid peroxidation was detected by measuring the amount of malondialdehyde (MDA)-TBA adduct in the cell homogenates. The cells were collected (2x107) with 1 ml PBS buffer according to the whole cell lysis procedure indicated in the TBARS assay kit. The cells were homogenized on ice with an ultrasonic homogenizer (UW2070; BANDELIN electronic GmbH & Co. KG), SDS was then added and the cells were mixed with the color reagent (TBA in acetic acid and sodium hydroxide) in a boiling water bath for 1 h, cooled in an ice-water bath, and then incubated on ice. After cooling, the sample was centrifuged at 1,600 x g for 10 min at 4˚C and was maintained at room temperature for 30 min. TBARS was measured by spectrophotometry (530 nm) and each sample concentration was determined using the MDA colorimetric standard curve. Results are expressed as µM. Experiments were repeated three times and the results are presented as the mean ± SEM.

Quantification of protein carbonyl content

The concentration of protein carbonyl was determined spectrophotometrically using a Protein Carbonyl Assay Kit according to the manufacturer's instructions. Briefly, cells at 80% confluence were collected and then homogenized in phosphate buffer (pH 6.7; containing 1 mM EDTA), after which, the sample was centrifuged at 10,000 x g for 15 min at 4˚C and the supernatant was removed. The lysates were incubated with dinitrophenylhydrazine for 1 h at room temperature in the dark. The protein was precipitated twice with trichloroacetic acid (first 20%, second 10%) and was then washed in an ethanol/ethyl acetate mixture. After being washed, the sample was resuspended in guanidine hydrochloride and centrifuged at 10,000 x g for 10 min at 4˚C. The protein carbonyl content was measured at 360 nm using spectrophotometry and was calculated according to the manufacturer's instructions. Results are expressed as nmol/mg protein. The protein content was determined using the BCA Protein Assay Kit (Takara Bio, Inc.), with BSA solution as the standard. This assay had a detection limit of 1-10 mg protein. Experiments were repeated three times and the results are presented the mean ± SEM.

Western blot analysis

Primary rat VSMCs were seeded in 6-well plates. After the cells reached 80% confluence, sample preparation for western blotting was performed using lysis buffer [50 mmol/l HEPES, 50 mmol/l NaCl, 1% (v/v) Triton X-100, 10% (v/v) glycerol, 1.5 mmol/l MgCl2, 1 mmol/l EDTA, 10 mmol/l sodium pyrophosphate, 1 mmol/l Na3VO4, 100 mmol/l NaF, 30 mmol/l 2-(p-nitrophenyl) phosphate, 1 mmol/l phenylmethylsulfonyl fluoride, 10 mg/ml leupeptin and 10 mg/ml aprotinin (pH 7.4)] as previously described (39). After the sample protein concentrations were calculated using the BCA Protein Assay Kit, total proteins (25 µg) were separated by SDS-PAGE on 10% gels and were then transferred onto 0.2-µm nitrocellulose membranes (Whatman plc; Cytiva). The membranes were blocked for 1 h at room temperature with 5% w/v nonfat dry milk solution, followed by incubation with primary antibodies against p53 (1:1,000) and β-actin (1:5,000) at 4˚C overnight. The following day, membranes were washed three times for 5 min in Tris-buffered saline with 0.01% Tween-20 (TBST) and then incubated with anti-mouse and anti-rabbit secondary antibodies for 2 h at room temperature. Subsequently, the membranes were washed three times for 5 min in TBST. The protein bands were detected using an enhanced chemiluminescence reagent-based Super Signal West Pico HRP Substrate System (Thermo Fisher Scientific, Inc.). Semi-quantification of the protein bands was performed using Alpha Digi Doc 1,000 Gel Documentation Unit (AlphaEaseFC™; Alpha Innotech Corporation). After stripping, the membranes were probed with anti-β-actin antibody for the same duration and at the same temperature as with the anti-p53 antibody to confirm equal protein loading. All experiments were performed in triplicate.

Statistical analysis

Statistical analysis was performed with GraphPad Prism (version 8.01; Dotmatics). The results are presented as the mean ± SEM. The differences among the control and experimental groups were evaluated by one-way ANOVA followed by Dunnett's multiple comparisons test. P<0.05 was considered to indicate a statistically significant difference.

Results

Superoxide anion accumulation is affected by uric acid in VSMCs independent of dose

Uric acid stimulation can promote the occurrence of oxidative stress in VSMCs and superoxide anion release is an upstream target mediator of oxidative stress (57-59). It was thus hypothesized that uric acid may promote the occurrence of superoxide anion release by upregulating oxidative stress. To understand whether superoxide anion release is affected by uric acid in VSMCs in a dose- and time-dependent manner, the accumulation of superoxide anion was determined using the Görlach method (56). Uric acid increased superoxide anion release at 1, 6, 12 and 24 h in a dose-independent manner compared with that in the control groups, although there was no significant difference observed at 3 h (Fig. 1). All uric acid doses decreased superoxide anion levels at 1, 3, 6, 12, and 24 h compared with the control; however, the increase observed at 3 h was less pronounced than at the other time points. In addition, in response to 25 and 50 mg/dl uric acid, superoxide anion accumulation was decreased compared with in response to the other uric acid concentrations (1, 2.5, 5, 10 and 12.5 mg/dl uric acid) at 6, 12 and 24 h.

Uric acid promotes protein carbonylation but does not affect lipid peroxidation

High levels of uric acid can be a key regulator for oxidative stress (21,35). In order to evaluate the effects of uric acid on oxidative stress in VSMCs, protein carbonylation and lipid peroxidation assays were performed. VSMCs were incubated with different uric acid concentrations (2.5, 5, 10 and 25 mg/dl) for various durations (1, 3, 12 and 24 h). The control group (0 mg/dl uric acid concentration) was not treated with uric acid. Treatment with all uric acid concentrations caused a significant increase in protein carbonyl levels at 1 h compared with those in the control group, but 10 mg/dl uric acid dose resulted in a reduction compared with the other doses (2.5, 5 and 25 mg/dl uric acid) (Fig. 2A). Protein carbonylation in VSMCs was significantly increased by all concentrations of uric acid at 3, 12 and 24 h compared with that in the control groups in a dose-dependent manner. As shown in Fig. 2B, uric acid stimulation had no effect on TBARS levels in VSMCs compared with those in the control group, thus indicating that no dose of uric acid promoted lipid peroxidation in VSMCs.

Uric acid decreases NO levels independent of dose and time

NO is a vasodilator that modulates important processes, such as vascular tone, inflammation and oxidation-sensitive mechanisms (60,61). To determine if higher uric acid concentrations modify NO levels in VSMCs, NO levels were analyzed in response to different uric acid doses for various durations. As shown in Fig. 3, all uric acid doses significantly diminished NO levels in a dose- and time-independent manner.

Long-term exposure to high uric acid levels suppresses p53 expression in VSMCs

Uric acid affected the protein expression levels of p53 in rat primary VSMCs in a time-dependent manner. As determined by western blotting, it was determined that all uric acid concentrations did not affect the expression levels of p53 in VSMCs at 1 and 3 h compared with those in the control group (0 mg/dl uric acid) (Fig. 4A and B). As shown in Fig. 4C, p53 expression was suppressed in VSMCs stimulated with high uric acid doses (12.5, 25 and 50 mg/dl uric acid concentrations) at 6 h. Long-term uric acid stimulation of VSMCs indicated that high concentrations of uric acid (12.5, 25 and 50 mg/dl uric acid) abolished p53 expression at 12 h (Fig. 4D), but the same doses of uric acid only slightly reduced the protein expression levels of p53 at 24 h (Fig. 4E).

Discussion

Uric acid is known to induce oxidative stress, which can pathologically contribute to hypertension in VSMCs (62). Given its significant role in causing hypertension, the vascular damage and endothelial injury induced by high uric acid levels was further assessed in the present study. VSMCs have a crucial role in maintaining endothelial homeostasis and in the development of blood vessels (22). Although research on the effects of uric acid on VSMCs is insufficient, numerous in vitro and in vivo studies have demonstrated that ECs continuously interact with VSMCs. These studies also showed that large amounts of NO produced by ECs diffuse into VSMCs, where NO is essential for regulating vascular contraction and relaxation (63-65). Despite these findings, the precise molecular mechanisms by which NO operates in uric acid-stimulated VSMCs under oxidative stress conditions remain elusive. Understanding these mechanisms is crucial as NO serves a pivotal role in vascular function and integrity. The interactions between ECs and VSMCs, particularly the transfer and effects of NO, are central to vascular health, and disruptions can lead to pathological states. Further research is needed to elucidate the pathways involved in NO signaling within VSMCs exposed to high levels of uric acid, as this knowledge could contribute to the development of therapeutic strategies aimed at mitigating uric acid-induced vascular damage and hypertension.

Oxidative stress is a pathological condition characterized by the excessive production of ROS, such as superoxide anion, hydroxyl radicals and hydrogen peroxide, along with oxidative damage to macromolecules (66-69). This process has been identified as a significant clinical risk factor in contributing to vascular damage, endothelial injury and the progression of vascular diseases (70-73). Emerging evidence has consistently demonstrated that uric acid is a critical factor in the development of endothelial dysfunction by regulating the oxidative stress of HUVECs (18,21,35,36). Specifically, studies have shown that uric acid stimulation leads to increased oxidative stress. Research has also indicated that uric acid-stimulated VSMCs have a marked increase in ROS production, and uric acid has been shown to affect oxidative stress-related signaling pathways within VSMCs (48,74).

The present study focused on the effect of uric acid on various oxidative stress parameters, including protein carbonylation, lipid peroxidation and superoxide anion levels in VSMCs after 1-24 h of treatment. Uric acid increased protein carbonylation levels in a dose-dependent manner. Specifically, protein carbonyl levels showed a statistically significant increase even at the 1-h time point, and this increase was almost maintained at 3, 6, 12 and 24 h, especially in response to higher concentrations of uric acid. The results at the short-term time point (1 h) suggested that uric acid may induce oxidative stress, leading to protein carbonylation in VSMCs and a high reactivity of uric acid with cellular proteins indicating that it may rapidly cause oxidative modifications. Additionally, the results in response to prolonged exposure (3, 12 and 24 h) provide a more comprehensive understanding of the impact of uric acid on oxidative stress. These findings emphasize the necessity of examining both short-term and long-term exposures to fully elucidate the biochemical pathways and molecular mechanisms. By contrast, the present analysis of lipid peroxidation, as measured by TBARs levels, revealed that uric acid did not significantly affect lipid peroxidation in VSMCs. This indicated that uric acid may preferentially induce oxidative stress in proteins rather than lipids. This selectivity may be attributed to differences in the susceptibility of proteins and lipids to oxidative damage, or it may be related to the specific localization of uric acid within cellular compartments. Moreover, VSMCs may possess robust antioxidant defense mechanisms, such as glutathione peroxidase and catalase, which effectively mitigate lipid peroxidation but are less effective against protein oxidation. This could explain the observed increase in protein carbonylation despite unchanged TBARs levels (75). To the best of our knowledge, the present study is the first to present data on the effects of uric acid stimulation on protein carbonylation and lipid peroxidation in rat primary VSMCs.

The present study indicated that all doses of uric acid increased superoxide anion release compared with that in the control groups at most time points (1-24 h, with the exception of at 3 h). Previous studies showed that a uric acid dose of 5 mg/dl significantly increased superoxide anion accumulation at 1 h in primary rat VSMCs (47). Consistent with previous findings (47), the current results demonstrated that all doses of uric acid caused a transient reduction of superoxide anion production in VSMCs at 3 h compared with at the other time points (1, 6, 12 and 24 h). Although there is no direct evidence to explain the transient decrease in superoxide anion levels at 3 h, it may be that the protective effect of the cells against oxidative damage is related to its ability to reduce oxidative stress in the early time intervals. These findings have the potential to provide evidence for the time-dependent effects of uric acid on superoxide anion production in VSMCs.

p53 is a central mediator of oxidative stress and apoptosis signaling in vascular functions (76), but its role in the pathogenesis of vascular damage remains insufficiently understood. Previous studies have linked p53 to apoptosis in VSMCs (77,78), although these investigations have been largely confined to the process of atherosclerotic plaque formation. Notably, p53 exhibits bidirectional functions in various biological processes, and its paradoxical role in metabolic pathways is attributed to the context-dependent nature of its activity (79,80). Despite this complexity, p53 signaling is recognized as a crucial regulator of oxidative stress, proliferation and inflammation. Furthermore, p53 activity has been implicated in uric acid-induced oxidative stress (45,81,82). Previous studies have shown that in response to mild ROS concentrations, p53 promotes cell survival by exerting an anti-oxidative effect to protect cells from damage. However, when cells are exposed to excessive and/or prolonged ROS levels, which can cause uncontrollable damage, p53 activity is inhibited by ROS, leading to cell death pathways to protect adjacent undamaged cells (83,84). In light of these findings, the present study aimed to evaluate the changes in p53 protein signaling in VSMCs stimulated with uric acid in a dose- and time-dependent manner. The results demonstrated that p53 protein expression was significantly suppressed by high doses of uric acid (12.5, 25 and 50 mg/dl) during prolonged stimulations (6-24 h). These results provided preliminary findings indicating a dose- and time-dependent relationship between uric acid exposure and p53 activity in VSMCs.; with p53 expression remaining stable at early time points (1 and 3 h) but progressively decreasing at 6 and 12 h, especially in response to higher uric acid concentrations. Furthermore, when comparing the results of p53 protein expression with oxidative stress parameters, it was observed that the suppression of p53 protein expression was associated with increased superoxide anion accumulation during long-term stimulation of high uric acid doses. These findings suggested that the accumulation of superoxide anions induced by uric acid may be associated with the suppression of p53 activation in VSMCs. Although the data provide valuable information about the molecular mechanisms by which uric acid affects vascular cell function, further studies are needed to explore p53 and oxidative stress pathways in vascular diseases.

It has been determined that NO-mediated apoptosis is increased by p53 deficiency in VSMCs (85,86). Furthermore, it has been shown that p53 deficiency can be increased by NO-mediated oxidative stress (44). These findings highlight the protective role of p53 in VSMCs; however, the precise relationship between p53 and NO in these cells remains unclear. It was hypothesized that p53 might also modulate the levels of oxidative stress in VSMCs, thereby controlling NO levels, given that one of the main functions of p53 is the regulation of oxidative stress. To explore this hypothesis, VSMCs were stimulated with various doses of uric acid over different time periods and superoxide anion, TBARS and protein carbonylation levels were measured. The results of the present study showed that uric acid concentrations decreased NO levels in VSMCs in a dose-independent manner at all time points. Based on these findings, it was concluded that the protective effect of p53 against uric acid-induced oxidative stress in VSMCs may not be mediated through the regulation of NO levels. This suggests that p53 may exert its protective effects through alternative pathways or mechanisms that do not directly involve NO. Additionally, the study observed increases in protein carbonyl levels and superoxide anion in response to uric acid exposure, which are all indicators of oxidative stress. Furthermore, TBARS was not directly related to the concentration of uric acid, indicating that TBARS levels were not sensitive to changes in uric acid concentration. This conclusion is visually summarized in Fig. 5, which illustrates the complex interactions between uric acid, oxidative stress markers and p53 activity.

To the best of our knowledge, the present study is the first to show that uric acid affected NO levels in VSMCs in a dose- and time-independent manner, while also providing comprehensive evidence that uric acid increased superoxide anion levels at 1, 3, 6, 12 and 24 h, with a less pronounced increase at 3 h compared with the other time points, and its dose-dependent effects on protein carbonyl levels. The results demonstrated that the doses of uric acid that induced accumulation of superoxide anion may also inhibit p53 protein activity in the long term, independent of NO levels. Additionally, a significant finding of the present study was that uric acid reduced NO levels in VSMCs regardless of the exposure time and dose. The effects of uric acid on protein carbonylation and lipid peroxidation in VSMCs are little-known oxidative damage parameters that need further investigation.

The results of the present study indicated that uric acid may significantly increase oxidative stress in VSMCs, suggesting that controlling uric acid levels could represent a potential therapeutic target for preventing hypertension and related diseases. Furthermore, it is clear that additional research is needed to understand the role of the p53 signaling pathway in uric acid-induced oxidative stress-mediated vascular damage and to overcome cardiovascular diseases.

The primary aim of the present study was to provide a detailed analysis of the cellular mechanisms involved. Therefore, in vivo experiments and clinical studies are essential to translate these findings into real-world applications. In future studies, further analyses, including assessing the activities of antioxidant enzymes such as SOD, catalase and glutathione peroxidase, may provide more comprehensive data on the effects of uric acid, and these additional markers will help to elucidate cellular antioxidant defense mechanisms and their response to uric acid-induced oxidative stress. The integration of such data with the current study could provide a more comprehensive understanding of the effects of uric acid on vascular health.

In conclusion, while the current study lays the groundwork by elucidating the cellular effects of uric acid in VSMCs, further in vivo studies and clinical research are required for a complete understanding of these mechanisms. The data presented in the current study may serve as a foundation for future investigations, and the findings could have broader implications when integrated with additional studies. Therefore, future studies will aim to include investigations on ECs and immune cells to provide a comprehensive understanding of the effects of uric acid on vascular health. These future studies are crucial for developing effective therapeutic strategies to mitigate the adverse effects of uric acid on vascular health.

Acknowledgements

Not applicable.

Funding

Funding: This research was supported by Akdeniz University Scientific Research Project Unit (grant no. TSA-2018-3543).

Availability of data and materials

The data generated in the present study may be requested from the corresponding author.

Authors' contributions

SD was involved in data curation, formal analysis, investigation, methodology and the use of software. SD also contributed to the writing of the original draft, and participated in the review and editing process. EY contributed to data curation, formal analysis and methodology, as well as writing the original draft. AY was responsible for conceptualization, funding acquisition, investigation, project administration and providing resources. Additionally, AY provided supervision and validation, and was involved in the review and editing of the manuscript. SD and AY confirm the authenticity of all the raw data. All authors read and approved the final version of the manuscript.

Ethics approval and consent to participate

All animal experiments were performed according to the Guide for the Care and Use of Laboratory Animals following experimental protocols approved by the Local Committee on Animal Research Ethics at Akdeniz University (approval no. 727/2018.01.024).

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Becker BF: Towards the physiological function of uric acid. Free Radic Biol Med. 14:615–631. 1993.PubMed/NCBI View Article : Google Scholar

2 

Bardin T and Richette P: Definition of hyperuricemia and gouty conditions. Curr Opin Rheumatol. 26:186–191. 2014.PubMed/NCBI View Article : Google Scholar

3 

Dalbeth N, Gosling A, Gaffo A and Abhishek A: Gout. Lancet. 397:1843–1855. 2021.PubMed/NCBI View Article : Google Scholar

4 

Johnson RJ, Kang DH, Feig D, Kivlighn S, Kanellis J, Watanabe S, Tuttle KR, Rodriguez-Iturbe B, Herrera-Acosta J and Mazzali M: Is there a pathogenetic role for uric acid in hypertension and cardiovascular and renal disease? Hypertension. 41:1183–1190. 2003.PubMed/NCBI View Article : Google Scholar

5 

Feig DI, Kang DH and Johnson RJ: Uric acid and cardiovascular risk. N Engl J Med. 359:1811–1821. 2008.PubMed/NCBI View Article : Google Scholar

6 

Lanaspa MA, Sanchez-Lozada LG, Choi YJ, Cicerchi C, Kanbay M, Roncal-Jimenez CA, Ishimoto T, Li N, Marek G, Duranay M, et al: Uric acid induces hepatic steatosis by generation of mitochondrial oxidative stress: Potential role in fructose-dependent and-independent fatty liver. J Biol Chem. 287:40732–40744. 2012.PubMed/NCBI View Article : Google Scholar

7 

Sautin YY, Nakagawa T, Zharikov S and Johnson RJ: Adverse effects of the classic antioxidant uric acid in adipocytes: NADPH oxidase-mediated oxidative/nitrosative stress. Am J Physiol Cell Physiol. 293:C584–C596. 2007.PubMed/NCBI View Article : Google Scholar

8 

Ames BN, Cathcart R, Schwiers E and Hochstein P: Uric acid provides an antioxidant defense in humans against oxidant- and radical-caused aging and cancer: A hypothesis. Proc Natl Acad Sci USA. 78:6858–6862. 1981.PubMed/NCBI View Article : Google Scholar

9 

Peden DB, Hohman R, Brown ME, Mason RT, Berkebile C, Fales HM and Kaliner MA: Uric acid is a major antioxidant in human nasal airway secretions. Proc Natl Acad Sci USA. 87:7638–7642. 1990.PubMed/NCBI View Article : Google Scholar

10 

Sautin YY and Johnson RJ: Uric acid: The oxidant-antioxidant paradox. Nucleosides Nucleotides Nucleic Acids. 27:608–619. 2008.PubMed/NCBI View Article : Google Scholar

11 

Zhao G, Huang L, Song M and Song Y: Baseline serum uric acid level as a predictor of cardiovascular disease related mortality and all-cause mortality: A meta-analysis of prospective studies. Atherosclerosis. 231:61–68. 2013.PubMed/NCBI View Article : Google Scholar

12 

Zhang W, Iso H, Murakami Y, Miura K, Nagai M, Sugiyama D, Ueshima H and Okamura T: EPOCH-JAPAN GROUP. Serum uric acid and mortality form cardiovascular disease: EPOCH-JAPAN study. J Atheroscler Thromb. 23:692–703. 2016.PubMed/NCBI View Article : Google Scholar

13 

Lee SW, Kim HC, Nam C, Lee HY, Ahn SV, Oh YA and Suh I: Age-differential association between serum uric acid and incident hypertension. Hypertens Res. 42:428–437. 2019.PubMed/NCBI View Article : Google Scholar

14 

Sakata S, Hata J, Honda T, Hirakawa Y, Oishi E, Shibata M, Yoshida D, Goto K, Kitazono T and Ninomiya T: Serum uric acid levels and cardiovascular mortality in a general Japanese population: The Hisayama study. Hypertens Res. 43:560–568. 2020.PubMed/NCBI View Article : Google Scholar

15 

Kurra V, Vehmas T, Eräranta A, Jokihaara J, Pirttiniemi P, Ruskoaho H, Tokola H, Niemelä O, Mustonen J and Pörsti I: Effects of oxonic acid-induced hyperuricemia on mesenteric artery tone and cardiac load in experimental renal insufficiency. BMC Nephrol. 16(35)2015.PubMed/NCBI View Article : Google Scholar

16 

Garcia-Arroyo FE, Gonzaga G, Munoz-Jimenez I, Blas-Marron MG, Silverio O, Tapia E, Soto V, Ranganathan N, Ranganathan P, Vyas U, et al: Probiotic supplements prevented oxonic acid-induced hyperuricemia and renal damage. PLoS One. 13(e0202901)2018.PubMed/NCBI View Article : Google Scholar

17 

Mazzali M, Hughes J, Kim YG, Jefferson JA, Kang DH, Gordon KL, Lan HY, Kivlighn S and Johnson RJ: Elevated uric acid increases blood pressure in the rat by a novel crystal-independent mechanism. Hypertension. 38:1101–1106. 2001.PubMed/NCBI View Article : Google Scholar

18 

Yu MA, Sánchez-Lozada LG, Johnson RJ and 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. 28:1234–1242. 2010.PubMed/NCBI

19 

Hsu WL, Li SY, Liu JS, Huang PH, Lin SJ, Hsu CC, Lin YP and Tarng DC: High uric acid ameliorates indoxyl sulfate-induced endothelial dysfunction and is associated with lower mortality among hemodialysis patients. Toxins (Basel). 9(20)2017.PubMed/NCBI View Article : Google Scholar

20 

Cai W, Duan XM, Liu Y, Yu J, Tang YL, Liu ZL, Jiang S, Zhang CP, Liu JY and Xu JX: Uric acid induces endothelial dysfunction by activating the HMGB1/RAGE signaling pathway. Biomed Res Int. 2017(4391920)2017.PubMed/NCBI View Article : Google Scholar

21 

Ko J, Kang HJ, Kim DA, Kim MJ, Ryu ES, Lee S, Ryu JH, Roncal C, Johnson RJ and Kang DH: Uric acid induced the phenotype transition of vascular endothelial cells via induction of oxidative stress and glycocalyx shedding. FASEB J. 33:13334–13345. 2019.PubMed/NCBI View Article : Google Scholar

22 

Sandoo A, van Zanten JJ, Metsios GS, Carroll D and Kitas GD: The endothelium and its role in regulating vascular tone. Open Cardiovasc Med J. 4:302–312. 2010.PubMed/NCBI View Article : Google Scholar

23 

Brozovich F, Nicholson C, Degen C, Gao YZ, Aggarwal M and Morgan K: Mechanisms of vascular smooth muscle contraction and the basis for pharmacologic treatment of smooth muscle disorders. Pharmacol Rev. 68:476–532. 2016.PubMed/NCBI View Article : Google Scholar

24 

Touyz RM, Alves-Lopes R, Rios FJ, Camargo LL, Anagnostopoulou A, Arner A and Montezano AC: Vascular smooth muscle contraction in hypertension. Cardiovasc Res. 114:529–539. 2018.PubMed/NCBI View Article : Google Scholar

25 

Pacher P, Beckman JS and Liaudet L: Nitric oxide and peroxynitrite in health and disease. Physiol Rev. 87:315–424. 2007.PubMed/NCBI View Article : Google Scholar

26 

Radi R: Oxygen radicals, nitric oxide, and peroxynitrite: Redox pathways in molecular medicine. Proc Natl Acad Sci USA. 115:5839–5848. 2018.PubMed/NCBI View Article : Google Scholar

27 

Maruhashi T, Hisatome I, Kihara Y and Higashi Y: Hyperuricemia and endothelial function: From molecular background to clinical perspectives. Atherosclerosis. 278:226–231. 2018.PubMed/NCBI View Article : Google Scholar

28 

Yu W and Cheng JD: Uric acid and cardiovascular disease: An update from molecular mechanism to clinical perspective. Front Pharmacol. 11(582680)2020.PubMed/NCBI View Article : Google Scholar

29 

Pacher P, Obrosova IG, Mabley JG and Szabó C: Role of nitrosative stress and peroxynitrite in the pathogenesis of diabetic complications. Emerging new therapeutical strategies. Curr Med Chem. 12:267–275. 2005.PubMed/NCBI View Article : Google Scholar

30 

Wang F, Yuan Q, Chen F, Pang J, Pan C, Xu F and Chen Y: Fundamental mechanisms of the cell death caused by nitrosative stress. Front Cell Dev Biol. 9(742483)2021.PubMed/NCBI View Article : Google Scholar

31 

Choi YJ, Yoon Y, Lee KY, Hien TT, Kang KW, Kim KC, Lee J, Lee MY, Lee SM, Kang DH and Lee BH: Uric acid induces endothelial dysfunction by vascular insulin resistance associated with the impairment of nitric oxide synthesis. FASEB J. 28:3197–3204. 2014.PubMed/NCBI View Article : Google Scholar

32 

Mishima M, Hamada T, Maharani N, Ikeda N, Onohara T, Notsu T, Ninomiya H, Miyazaki S, Mizuta E, Sugihara S, et al: Effects of uric acid on the NO production of HUVECs and its restoration by urate lowering agents. Drug Res (Stuttg). 66:270–274. 2016.PubMed/NCBI View Article : Google Scholar

33 

Lin Y, Xie Y, Hao Z, Bi H, Liu Y, Yang X and Xia Y: Protective effect of uric acid on ox-LDL-induced HUVECs injury via Keap1-Nrf2-ARE pathway. J Immunol Res. 2021(5151168)2021.PubMed/NCBI View Article : Google Scholar

34 

Ouyang R, Zhao X, Zhang R, Yang J, Li S and Deng D: FGF21 attenuates high uric acid-induced endoplasmic reticulum stress, inflammation and vascular endothelial cell dysfunction by activating Sirt1. Mol Med Rep. 25(35)2022.PubMed/NCBI View Article : Google Scholar

35 

Li P, Zhang L, Zhang M, Zhou C and Lin N: Uric acid enhances PKC-dependent eNOS phosphorylation and mediates cellular ER stress: A mechanism for uric acid-induced endothelial dysfunction. Int J Mol Med. 37:989–997. 2016.PubMed/NCBI View Article : Google Scholar

36 

Huang Z, Hong Q, Zhang X, Xiao W, Wang L, Cui S, Feng Z, Lv Y, Cai G, Chen X and Wu D: Aldose reductase mediates endothelial cell dysfunction induced by high uric acid concentrations. Cell Commun Signal. 15(3)2017.PubMed/NCBI View Article : Google Scholar

37 

Lee TS, Lu TM, Chen CH, Guo BC and Hsu CP: Hyperuricemia induces endothelial dysfunction and accelerates atherosclerosis by disturbing the asymmetric dimethylarginine/dimethylarginine dimethylaminotransferase 2 pathway. Redox Biol. 46(102108)2021.PubMed/NCBI View Article : Google Scholar

38 

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.PubMed/NCBI View Article : Google Scholar

39 

Doğru S, Yaşar E and Yeşilkaya A: Uric acid can enhance MAPK pathway-mediated proliferation in rat primary vascular smooth muscle cells via controlling of mitochondria and caspase-dependent cell death. J Recept Signal Transduct Res. 42:293–301. 2022.PubMed/NCBI View Article : Google Scholar

40 

Li T, Kon N, Jiang L, Tan M, Ludwig T, Zhao Y, Baer R and Gu W: Tumor suppression in the absence of p53-mediated cell-cycle arrest, apoptosis, and senescence. Cell. 149:1269–1283. 2012.PubMed/NCBI View Article : Google Scholar

41 

Itahana Y and Itahana K: Emerging roles of p53 family members in glucose metabolism. Int J Mol Sci. 19(776)2018.PubMed/NCBI View Article : Google Scholar

42 

Mercer J and Bennett M: The role of p53 in atherosclerosis. Cell Cycle. 5:1907–1909. 2006.PubMed/NCBI View Article : Google Scholar

43 

Phadwal K, Tang QY, Luijten I, Zhao JF, Corcoran B, Semple RK, Ganley IG and MacRae VE: p53 regulates mitochondrial dynamics in vascular smooth muscle cell calcification. Int J Mol Sci. 24(1643)2023.PubMed/NCBI View Article : Google Scholar

44 

Popowich DA, Vavra AK, Walsh CP, Bhikhapurwala HA, Rossi NB, Jiang Q, Aalami OO and Kibbe MR: Regulation of reactive oxygen species by p53: Implications for nitric oxide-mediated apoptosis. Am J Physiol Heart Circ Physiol. 298:H2192–H2200. 2010.PubMed/NCBI View Article : Google Scholar

45 

Itahana Y, Han R, Barbier S, Lei Z, Rozen S and Itahana K: The uric acid transporter SLC2A9 is a direct target gene of the tumor suppressor p53 contributing to antioxidant defense. Oncogene. 34:1799–1810. 2015.PubMed/NCBI View Article : Google Scholar

46 

Kang DH, Han L, Ouyang X, Kahn AM, Kanellis J, Li P, Feng L, Nakagawa T, Watanabe S, Hosoyamada M, et al: Uric acid causes vascular smooth muscle cell proliferation by entering cells via a functional urate transporter. Am J Nephrol. 25:425–433. 2005.PubMed/NCBI View Article : Google Scholar

47 

Oğuz N, Kırça M, Çetin A and Yeşilkaya A: Effect of uric acid on inflammatory COX-2 and ROS pathways in vascular smooth muscle cells. J Recept Signal Transduct Res. 37:500–505. 2017.PubMed/NCBI View Article : Google Scholar

48 

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.PubMed/NCBI View Article : Google Scholar

49 

National Research Council: Guide for the Care and Use of Laboratory Animals: 8th edition. The National Academies Press, Washington, DC, 2011. https://doi.org/10.17226/12910.

50 

American Veterinary Medical Association: AVMA Guidelines for the Euthanasia of Animals. AVMA, Schaumburg, IL, 2020. https://www.avma.org/resources-tools/avma-policies/avma-guidelines-euthanasia-animals.

51 

Boston University: Institutional animal care and use committee (IACUC) Guidelines. https://www.bu.edu/research/ethics-compliance/.

52 

University of Maryland: Animal care and use Training. https://research.umd.edu/resources/department-laboratory-animal-resources-dlar/animal-care-and-use-training.

53 

Ahmadi-Noorbakhsh S, Abbasi MF, Ghasemi M, Bayat G, Davoodian N, Sharif-Paghaleh E, Poormoosavi SM, Rafizadeh M, Maleki M, Shirzad-Aski H, et al: Anesthesia and analgesia for common research models of adult mice. Lab Anim Res. 38(40)2022.PubMed/NCBI View Article : Google Scholar

54 

Parasuraman S and Christapher PV: Anesthesia and euthanasia of experimental animals. In: Introduction to basics of pharmacology and toxicology: Volume 3: Experimental Pharmacology: Research methodology and biostatistics. Springer, pp65-75, 2022.

55 

Gunther S, Alexander RW, Atkinson WJ and Gimbrone MA Jr: Functional angiotensin II receptors in cultured vascular smooth muscle cells. J Cell Biol. 92:289–298. 1982.PubMed/NCBI View Article : Google Scholar

56 

Görlach A, Brandes RP, Bassus S, Kronemann N, Kirchmaier CM, Busse R and Schini-Kerth VB: Oxidative stress and expression of p22phox are involved in the up-regulation of tissue factor in vascular smooth muscle cells in response to activated platelets. FASEB J. 14:1518–1528. 2000.PubMed/NCBI

57 

Muraoka S and Miura T: Inhibition by uric acid of free radicals that damage biological molecules. Pharmacol Toxicol. 93:284–289. 2003.PubMed/NCBI View Article : Google Scholar

58 

Rodrigo R, González J and Paoletto F: The role of oxidative stress in the pathophysiology of hypertension. Hyperten Res. 34:431–440. 2011.PubMed/NCBI View Article : Google Scholar

59 

Liu N, Xu H, Sun Q, Yu X, Chen W, Wei H, Jiang J, Xu Y and Lu W: The role of oxidative stress in hyperuricemia and xanthine oxidoreductase (XOR) inhibitors. Oxid Med Cell Longev. 2021(1470380)2021.PubMed/NCBI View Article : Google Scholar

60 

Levine AB, Punihaole D and Levine TB: Characterization of the role of nitric oxide and its clinical applications. Cardiology. 122:55–68. 2012.PubMed/NCBI View Article : Google Scholar

61 

Napoli C, Paolisso G, Casamassimi A, Al-Omran M, Barbieri M, Sommese L, Infante T and Ignarro LJ: Effects of nitric oxide on cell proliferation: Novel insights. J Am Coll Cardiol. 62:89–95. 2013.PubMed/NCBI View Article : Google Scholar

62 

Gherghina ME, Peride I, Tiglis M, Neagu TP, Niculae A and Checherita IA: Uric acid and oxidative stress-relationship with cardiovascular, metabolic, and renal impairment. Int J Mol Sci. 23(3188)2022.PubMed/NCBI View Article : Google Scholar

63 

Li M, Qian M, Kyler K and Xu J: Endothelial-vascular smooth muscle cells interactions in atherosclerosis. Front Cardiovasc Med. 5(151)2018.PubMed/NCBI View Article : Google Scholar

64 

Hirase T and Node K: Endothelial dysfunction as a cellular mechanism for vascular failure. Am J Physiol Heart Circ Physiol. 302:H499–H505. 2012.PubMed/NCBI View Article : Google Scholar

65 

Vanhoutte PM, Zhao Y, Xu A and Leung SW: Thirty years of saying NO: Sources, fate, actions, and misfortunes of the endothelium-derived vasodilator mediator. Circ Res. 119:375–396. 2016.PubMed/NCBI View Article : Google Scholar

66 

Tsutsui H, Kinugawa S and Matsushima S: Oxidative stress and heart failure. Am J Physiol Heart Circ Physiol. 30:H2181–H2190. 2011.PubMed/NCBI View Article : Google Scholar

67 

Sugamura K and Keaney JF Jr: Reactive oxygen species in cardiovascular disease. Free Radic Biol Med. 51:978–992. 2011.PubMed/NCBI View Article : Google Scholar

68 

Drummond GR, Selemidis S, Griendling KK and Sobey CG: Combating oxidative stress in vascular disease: NADPH oxidases as therapeutic targets. Nat Rev Drug Discov. 10:453–471. 2011.PubMed/NCBI View Article : Google Scholar

69 

Murray M, Selby-Pham S, Colton BL, Bennett L, Williamson G and Dordevic AL: Does timing of phytonutrient intake influence the suppression of postprandial oxidative stress? A systematic literature review. Redox Biol. 46(102123)2021.PubMed/NCBI View Article : Google Scholar

70 

Baradaran A, Nasri H and Rafieian-Kopaei M: Oxidative stress and hypertension: Possibility of hypertension therapy with antioxidants. J Res Med Sci. 19:358–367. 2014.PubMed/NCBI

71 

Tahhan AS, Sandesara PB, Hayek SS, Alkhoder A, Chivukula K, Hammadah M, Mohamed-Kelli H, O'Neal WT, Topel M, Ghasemzadeh N, et al: Association between oxidative stress and atrial fibrillation. Heart Rhythm. 14:1849–1855. 2017.PubMed/NCBI View Article : Google Scholar

72 

Ahmad KA, Yuan DY, Nawaz W, Ze H, Zhuo CX, Talal B, Taleb A, Mais E and Qilong D: Antioxidant therapy for management of oxidative stress induced hypertension. Free Radic Res. 51:428–438. 2017.PubMed/NCBI View Article : Google Scholar

73 

Kattoor AJ, Pothineni NVK, Palagiri D and Mehta JL: Oxidative stress in atherosclerosis. Curr Atheroscler Rep. 19(42)2017.PubMed/NCBI View Article : Google Scholar

74 

Dai Y, Cao Y, Zhang Z, Vallurupalli S and Mehta JL: Xanthine oxidase induces foam cell formation through LOX-1 and NLRP3 activation. Cardiovasc Drugs Ther. 31:19–27. 2017.PubMed/NCBI View Article : Google Scholar

75 

Coliva G, Lange M, Colombo S, Chervet JP, Domingues MR and Fedorova M: Sphingomyelins prevent propagation of lipid peroxidation-LC-MS/MS evaluation of inhibition mechanisms. Molecules. 25(1925)2020.PubMed/NCBI View Article : Google Scholar

76 

Chan HH, Chan E, Kwok CTK, Leung GPH, Lee SMY and Seto SW: The role of p53 in the alternation of vascular functions. Front Pharmacol. 13(981152)2022.PubMed/NCBI View Article : Google Scholar

77 

Mercer J, Figg N, Stoneman V, Braganza D and Bennett MR: Endogenous p53 protects vascular smooth muscle cells from apoptosis and reduces atherosclerosis in ApoE knockout mice. Circ Res. 96:667–674. 2005.PubMed/NCBI View Article : Google Scholar

78 

Cao RY, Eves R, Jia L, Funk CD, Jia Z and Mak AS: Effects of p53-knockout in vascular smooth muscle cells on atherosclerosis in mice. PLoS One. 12(e0175061)2017.PubMed/NCBI View Article : Google Scholar

79 

Wang M and Attardi LD: A balancing act: p53 activity from tumor suppression to pathology and therapeutic implications. Annu Rev Pathol. 17:205–226. 2022.PubMed/NCBI View Article : Google Scholar

80 

Kastenhuber ER and Lowe SW: Putting p53 in context. Cell. 170:1062–1078. 2017.PubMed/NCBI View Article : Google Scholar

81 

Buizza L, Cenini G, Lanni C, Ferrari-Toninelli G, Prandelli C, Govoni S, Buoso E, Racchi M, Barcikowska M, Styczynska M, et al: Conformational altered p53 as an early marker of oxidative stress in Alzheimer's disease. PLoS One. 7(e29789)2012.PubMed/NCBI View Article : Google Scholar

82 

Yang L, Chang B, Guo Y, Wu X and Liu L: The role of oxidative stress-mediated apoptosis in the pathogenesis of uric acid nephropathy. Ren Fail. 41:616–622. 2019.PubMed/NCBI View Article : Google Scholar

83 

Liu Y and Gu W: The complexity of p53-mediated metabolic regulation in tumor suppression. Semin Cancer Biol. 85:4–32. 2022.PubMed/NCBI View Article : Google Scholar

84 

Kruiswijk F, Labuschagne CF and Vousden KH: p53 in survival, death and metabolic health: A lifeguard with a licence to kill. Nat Rev Mol Cell Biol. 16:393–405. 2015.PubMed/NCBI View Article : Google Scholar

85 

Kibbe MR, Li J, Nie S, Choi BM, Kovesdi I, Lizonova A, Billiar TR and Tzeng E: Potentiation of nitric oxide-induced apoptosis in p53-/- vascular smooth muscle cells. Am J Physiol Cell Physiol. 282:C625–C634. 2002.PubMed/NCBI View Article : Google Scholar

86 

Kim YM, Choi BM, Kim YS, Kwon YG, Kibbe MR, Billiar TR and Tzeng E: Protective effect of p53 in vascular smooth muscle cells against nitric oxide-induced apoptosis is mediated by up-regulation of heme oxygenase-2. BMB Rep. 41:164–169. 2008.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

December-2024
Volume 21 Issue 6

Print ISSN: 2049-9434
Online ISSN:2049-9442

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Dogru S, Yasar E and Yesilkaya A: Effects of uric acid on oxidative stress in vascular smooth muscle cells. Biomed Rep 21: 171, 2024.
APA
Dogru, S., Yasar, E., & Yesilkaya, A. (2024). Effects of uric acid on oxidative stress in vascular smooth muscle cells. Biomedical Reports, 21, 171. https://doi.org/10.3892/br.2024.1859
MLA
Dogru, S., Yasar, E., Yesilkaya, A."Effects of uric acid on oxidative stress in vascular smooth muscle cells". Biomedical Reports 21.6 (2024): 171.
Chicago
Dogru, S., Yasar, E., Yesilkaya, A."Effects of uric acid on oxidative stress in vascular smooth muscle cells". Biomedical Reports 21, no. 6 (2024): 171. https://doi.org/10.3892/br.2024.1859