1. Introduction
Complex interactions between vascular smooth muscle cells (VSMCs), endothelial cells (ECs), and immune cells contribute to vascular health and its physiological function [1,2]. Endothelial dysfunction (ED), an early indicator of vascular dysfunction (VD), is characterized by the loss of the endothelium’s anti-inflammatory, anticoagulant, and vasodilatory properties, marking one of the initial stages in cardiovascular disease (CVDs) [3,4,5,6], such as hypertension, atherosclerosis, stroke, obesity, diabetes, and thrombosis [7,8,9,10,11].
A powerful stimulator of vascular inflammation and oxidative stress is a high intake of sodium chloride (NaCl) [12,13,14], which leads to ED by promoting vascular low-grade inflammation and impaired vasodilation even in healthy, normotensive individuals [13,14,15,16]. For example, in young, healthy individuals, a seven-day high salt (HS) intake significantly impairs endothelium-dependent vasodilation in macro- and microcirculation without affecting arterial blood pressure (BP), fluid retention, or body composition [17]. The HS diet is known to increase the risk of heart failure and hypertension [18,19,20,21,22]. Excessive salt intake is a significant, yet preventable, contributor to CVD-related deaths globally. Implementing appropriate dietary restrictions, lifestyle modifications, and effective pharmacological interventions, all of which emphasize sodium reduction to help regulate blood pressure, improve endothelial function [23], and mitigate these negative trends [19,24,25,26,27,28]. Pharmacological treatments for hypertension include antihypertensive medications, such as angiotensin-converting enzyme (ACE) inhibitors [29,30,31], angiotensin II (AngII) receptor blockers (ARBs) [32,33,34,35,36], calcium channel blockers (CCBs) [37,38,39], and diuretics.
The effects of an HS diet are associated with alterations in the redox equilibrium between reactive oxygen species (ROS) generation and antioxidant mechanisms [40]. The bioavailability and production of endothelium-derived vasodilator nitric oxide (NO), along with other endothelium-derived vasodilator factors, are heavily influenced by increased ROS and overall vascular oxidative stress [13,41,42].
Notably, HS intake suppresses the renin–angiotensin system and AngII levels, which is the main regulatory system for the maintenance of blood volume and arterial blood pressure by vasoconstriction, increased aldosterone synthesis, and stimulation of the sympathetic nervous system [43,44,45]. In the vasculature, under normal conditions, higher levels of AngII induce ROS generation in vascular smooth muscle and ECs [46]. However, paradoxically, low levels of AngII also lead to increased ROS generation. For example, Ćosić et al. (2016) reported a significant increase in intracellular ROS levels and down-regulation of antioxidant enzyme expression, accompanied by impaired flow-induced dilation (FID) of middle cerebral arteries (MCAs) in Sprague-Dawley (SD) rats after a seven-day HS dietary intake [42]. On the other hand, subpressor low-dose exposure to AngII has shown beneficial effects on redox balance and restoration of flow-induced dilation after salt-induced impairment of endothelium-dependent vasodilation [47,48]. Interestingly, we have demonstrated that increased NaCl dietary intake induced low-grade systemic inflammation, which could be related to suppressed AngII levels and involved changes in Th17 and Treg cell distribution, a shift in lipid and arachidonic acid metabolism, and vascular wall remodeling in animal models [49] and in humans on an HS diet [13]. Since HS interferes with cellular response through the alteration of key molecules involved in the inflammatory response [13,50,51], using supraphysiological doses of sodium in vitro helps to model pathological conditions, amplify cellular responses, and reveal mechanisms that are not observable under normal baseline conditions. Specifically, it was shown that excess salt leads to cellular senescence, promoting inflammatory/fibrotic response and reducing NO generation in human and animal cells [52,53,54]. A previous study by Dmitrieva and Burg (2015) [55] showed that elevated extracellular NaCl affects adhesion molecules’ gene expression in human umbilical vein endothelial cells (HUVECs), directly involving it in the facilitation of vascular changes. Nevertheless, existing research does not appear to address the effects of AngII supplementation in a cell culture model of high NaCl intake, nor its implications for regulating redox balance in ECs. Thus, the present study hypothesizes that increasing concentrations of NaCl in the cell medium will affect the viability and ROS generation in ECs and will alter the activation state of the cells. At the same time, AngII addition in physiological doses will prevent these changes and exhibit a protective effect in the EC culture model.
The aim of this study was to assess the NaCl-dose response accompanied by AngII exposure in vitro on human aortic endothelial cells (HAECs) viability, intracellular ROS production, and cell adhesion molecules’ (CAMs’) expression (intercellular adhesion molecule 1, ICAM-1; vascular cell adhesion molecule 1, VCAM-1; E-selectin; and endoglin) following treatment, to further elucidate underlying mechanisms and effectors in salt-induced endothelial damage.
2. Materials and Methods
2.1. Materials and Chemical Reagents
HAECs were purchased from Innoprot (Barcelona, Spain). Cell culture flasks and well plates were purchased from TPP Techno Plastic Products AG (Trasagiden, Switzerland). Human Large Vessel Endothelial Cell Basal Medium, low serum growth supplement (LSGS), and Trypsin-EDTA (0.25%) were purchased from Gibco (Thermo Fisher Scientific, Waltham, MA, USA). AngII was purchased from Merck (Darmstadt, Germany). NaCl was purchased from Gram-Mol d.o.o. (Zagreb, Croatia). The 3-(4, 5-dimethylthiazolyl-2)-2 and 5-diphenyltetrazolium bromide (MTT) were purchased from Invitrogen (Thermo Fisher Scientific, Waltham, MA, USA).
2.2. HAECs
The HAECs were carefully thawed, seeded in tissue-appropriate cell culture flasks for adherent cells (T-25 cm2), and placed in an incubator (Shel Lab, CO2 Series, Sheldon Manufacturing Inc., Cornelius, OR, USA) under the following conditions: ~37 °C, 5% CO2, and >80% humidity level. Human Large Vessel Endothelial Cell Basal Medium supplemented with LSGS was used throughout our experiment. HAECs were monitored daily, and basal media was changed every other day until reaching confluence. For experimental purposes, the fifth passage (P5) of cells was used at approximately 80% confluence. Trypsinization was used as a method for the detachment of the HAECs from the flask/plate surface.
2.3. Cell Culture Treatment: NaCl and AngII
Cell culture treatment was performed in 24 and 96-well plates, depending on the following protocols. The osmolality of the control medium was 270 mOsmol/kg (133 mmol/L) (CTRL group). The HS medium was prepared by adding NaCl to the total osmolality of (1) 320 mOsmol/kg (158 mmol/L; HS320 group) and (2) 350 mOsmol/kg (173 mmol/L; HS350 group) [55,56,57,58]. During the experiment, the control medium was replaced by the high NaCl medium for 24, 48, and 72 h. The cell medium used for experimental purposes was not supplemented with LSGS, or in other words, it was serum-free.
Different concentrations of AngII were administered at half of the NaCl incubation (for 24 h after 12 h, for 48 h after 24 h, and for 73 h after 36 h). AngII was added in the following concentrations: 10−4, 10−5, 10−6, and 10−7 mol/L.
For experiments including only HS exposure and flow cytometry analysis of CAMs’ expression, 5 biological replicates (n = 5) were performed, with each replicate carried out over a 6-month period, using newly thawed vials of cells and conducted at the appropriate passage. However, for oxidative stress analysis, which included AngII exposure, 3 biological replicates (n = 3) were performed due to technical challenges encountered during the study period. All experiments were performed in 5 technical replicates.
2.4. Cell Viability and Metabolic Activity
MTT assay was performed as described by Shiwakoti et al. (2020) [59]. Cells were seeded in 96-well plates and treated with appropriate NaCl concentrations and 10−6 mol/L of AngII, as described above. After 24, 48, and 72 h, 10 µL of MTT stock solution was added to each well, and the plate was placed in an incubator for 4 h to produce formazan crystals. After incubation, 100 µL of MTT solvent was added to dissolve the created formazan crystals. The intensity of staining was proportional to the number of living cells. Plates were read spectrophotometrically at 595 nm using a microplate reader (BioRad PR 3100 TSC, Bio-Rad Laboratories, Hercules, CA, USA).
Cell viability was calculated as a percentage of the untreated control by subtracting the absorbance of treated cells from the absorbance of untreated cells, dividing by the absorbance of untreated cells, and multiplying by 100.
2.5. Flow Cytometry
The samples were collected after the HAECs were exposed for 72 h to different concentrations of NaCl and AngII (36 h), which were administered to the cell cultures in 24-well plates. The cells were washed in 1× phosphate-buffered saline (PBS) and prepared for the appropriate staining protocol (approximately 105 cells/mL of medium).
The FACS Canto II flow cytometer (BD Bioscience, Franklin Lakes, NJ, USA; 488 excitation laser and 530/30 BP analysis filter) was used for the assessment of intracellular ROS production, as well as the CAMs’ expression. Data analysis and visualization were performed using Flow Logic software v.8 (Inivai Technologies, Mentone, Australia).
2.5.1. Intracellular ROS Production
A dichlorofluorescein diacetate (DCF-DA) assay was used to determine the levels of hydrogen peroxide (H2O2) and peroxynitrite (ONOO−). DCF-DA is a non-fluorescent, cell-permeable dye that is deacetylated inside the cell by cellular esterases to DCFH (2′,7′-dichlorodihydrofluorescein). DCFH is oxidized in the presence of ROS in a fluorescent DCF (2′,7′-dichlorofluorescein), which can be detected as an indicator of oxidative stress. The dihydroethidium (DHE) assay was used to determine the level of superoxide anion (O2.−) in the HAECs. This is another cell-permeable dye, which, upon entering the cell, is oxidized by superoxide to form ethidium, which emits red fluorescence that can be quantified to assess the levels of superoxide production within the cell.
Cells were resuspended in 100 µL of 1× PBS and incubated with DCF-DA or DHE (10 µM final concentration) for 30 min at +4 °C. Following the incubation period, the samples stained with DCF-DA were re-suspended in PBS and immediately analyzed, while samples stained with DHE required additional rinsing with PBS before resuspension in PBS and cytometer reading. Following initial readings, phorbol 12-myristate 13-acetate (PMA) was added to each sample to stimulate ROS production. Data are expressed as geomean fluorescence intensity (GMFI) in FLH-1 and FLH-2 channels. Assays were performed according to previously reported protocols that were optimized for this purpose [60,61].
2.5.2. CAMs’ Expression
Cells were resuspended and washed twice before staining with appropriate antibodies. The following antibodies were used for staining the HAECs: ICAM-1 (CD54, clone: 15.2, Proteintech Group, Inc., Rosemont, IL, USA), VCAM-1 (CD106, clone: 51-10C9; BD Pharmigen Inc., Franklin Lakes, NJ, USA), E-selectin (CD62E, clone: 68-5H11; BD Pharmigen Inc., Franklin Lakes, NJ, USA), and endoglin (CD105, clone: 266; BD Pharmigen Inc., Franklin Lakes, NJ, USA).
2.6. Statistical Analysis
The normality of the residuals, where appropriate, was assessed using the Shapiro–Wilk test, while the homogeneity of variances was determined using the Brown–Forsythe test. Significant differences between treatment groups were determined using one-way ANOVA and the following post hoc analyses: Dunnett’s test, which compared each treatment group to the control, and Tukey’s HSD test, which compared all treatment groups against each other (p < 0.05).
3. Results
3.1. Assessment of Cell Metabolic Activity (MTT Assay)
Changes in cellular metabolic activity (i.e., an indicator of cell viability) of HAECs following incubation with different NaCl concentrations before and after the addition of AngII in the cell media are shown in Figure 1. Cell metabolic activity was significantly reduced after 24, 48, and 72 h at 350 mOsmol/kg NaCl concentration compared to the CTRL. The addition of AngII to the HS320 incubated cells did not alter cell metabolic activity at 24 and 72 h time points, except for 48 h, where AngII significantly decreased metabolic activity compared to the CTRL and HS320 conditions. The addition of AngII to the HS350 incubated cells resulted in preserved metabolic activity of cells after 24 and 72 h, while it significantly decreased metabolic activity after 48 h of incubation.
3.2. Assessment of Intracellular ROS Production: Analysis of DCF-DA and DHE Fluorescence Signals
In the HS320 group, treatment with NaCl did not have any significant effect on the formation of H2O2 and ONOO− in the HAECs. Following 10−4 and 10−5 mol/L of AngII exposure, H2O2 and ONOO− formation was not different from the levels observed in the HS320 condition without AngII but increased significantly with AngII 10−6 and 10−7 mol/L, suggesting that higher doses of AngII suppressed H2O2 and ONOO− production. Formation of H2O2 and ONOO− was significantly increased in the HS350 group compared to the CTRL group (Figure 2). In the HS350 group, a significant decrease was detected after administering AngII in concentrations from 10−4 to 10−6 mol/L.
Production of O2.− in HAECs was significantly decreased in HS320 compared to CTRL, while there were no significant changes in the formation of O2.− in the HS350 group compared to the CTRL group (Figure 3). O2.− level was significantly decreased in the HS320 group supplemented with AngII compared to the CTRL but increased compared to the HS320 group without AngII. In the HS350 group, O2.− production was significantly decreased after the addition of AngII compared to the CTRL and HS350 group without AngII.
Additionally, PMA stimulation that was performed following the initial baseline readings, specifically under HS conditions (HS320 and HS350), is presented as Supplementary Figures for DCF-DA (Figure S1) and DHE assays (Figure S2).
3.3. Quantitative Analysis of CAM Expression: Flow Cytometry Analysis
Changes in CAM expressions following treatment with NaCl and different concentrations of AngII are shown in Figure 4. HS did not affect VCAM-1 expression (Figure 4A). However, VCAM-1 was significantly increased in the HS320 group following the addition of AngII at a 10−4 mol/L dose compared to the CTRL and HS groups, and in the HS350 group following AngII at 10−5 mol/L compared to the HS group. ICAM-1 expression was significantly decreased in the HS320 and HS350 groups following the addition of 10−4 mol/L of AngII compared to the CTRL (Figure 4B).
A significant decrease in endoglin expression occurred in both HS groups supplemented with 10−5 mol/L of AngII compared to the HS and CTRL groups (Figure 4C). There were no significant changes in the expression of E-selectin in all study groups, with or without AngII exposure (Figure 4D).
3.4. Protein Expression of AT1 and AT2 Receptors
Results that are presented in the Supplementary Materials (Figure S3) showed that protein expression of both AT receptors was significantly decreased in animals on the HS diet compared to control animals. AngII partially restored the AT1 receptor expression compared to the HS group but was still lower than in the CTRL group. The AT2 receptor was not affected by the AngII treatment.
4. Discussion
Other than regulating vascular tone, blood flow, and coagulation, ECs also secrete various cytokines, chemokines, growth factors, and adhesion molecules. This makes them important players in inflammation and leukocyte-EC adhesion, which underlies all major cardio-metabolic diseases [4,7,62,63].
Some of the noteworthy findings in the present study are the following: (1) higher doses and prolonged exposure to NaCl decrease the ECs’ metabolic activity and increase the ECs’ ROS production, thus making ECs an important source of ROS leading to endothelial dysfunction; (2) AngII exposure significantly reduces ROS production of ECs and can protect their metabolic activity to some extent; (3) AngII can modulate the expression of CAMs (as demonstrated by a decrease in ICAM-1 and endoglin expression with certain utilized doses of AngII). Taken together, the present study demonstrates the direct effects of NaCl on ECs’ activation status and redox balance, which could be altered by AngII.
As previously discussed, HS intake is a significant source of ROS, leading to systemic oxidative stress, decreased antioxidant activity, and impaired endothelial function [64,65]. NaCl, being the universal stressor, provokes an adaptive response in cells to mitigate the damage and maintain viability under hyperosmotic conditions. Generally, the elevation of osmolality above 400 mOsmol/kg H2O progressively impairs cell proliferation, and while cell cycle arrest provides short-term adaptation, it ultimately leads to cell death (500 mOsmol/kg), while lower concentration for shorter periods induces reversible changes in cells [66]. In our current study, we aimed to evaluate the NaCl-dose and time-dependent response in conjunction with AngII exposure in vitro, focusing on the HAECs’ metabolic activity, intracellular ROS production, and the expression of CAMs (ICAM-1, VCAM-1, E-selectin, and endoglin) following treatment. The results suggest that a NaCl concentration of 320 mOsmol/kg in the HAECs culture has a hormetic effect (bi-phasic dose–response curve), yielding a lower level of ROS, especially superoxide anion [67,68,69,70]. There are several potential explanations for this occurrence. HS alters the redox state of cells. A low dose of this stressor might produce mild stress-altering gene expression and protein synthesis. As demonstrated earlier, this further affects enzymes involved in ROS production, such as NADPH oxidase and mitochondrial respiratory complex, activating protective mechanisms like antioxidant pathways and consequently leading to reduced ROS [64,66,71,72], suggesting that cellular tolerance occurs when cells are exposed to NaCl concentrations close to physiological levels, such as 320 mOsmol/kg. However, Dmitrieva et al. (2004) and others warned that these surviving, adapted cells may be damaged beyond repair at the DNA level despite rapid proliferation and minimal apoptosis [66,72,73,74]. Furthermore, Xu et al. (2009) co-cultured ECs with VSMCs and reported that ECs upregulate antioxidants, more accurately, thioredoxin, consequently decreasing ROS production by the VSMCs in a state of stress [75]. These findings are supportive of our present results but also present a challenging perspective for future investigations.
On the other hand, a concentration of 350 mOsmol/kg appears to be a critical point for NaCl-induced oxidative stress in our cultured HAECs, significantly increasing H2O2 and ONOO− production (DCF-DA assay). A significant increase in O2.− was observed at a NaCl concentration of 350 mOsmol/kg following PMA stimulation (DHE assay), presented in Figure S1 (Supplementary Materials). These differences in ROS production at different NaCl concentrations might result from different metabolic pathways activated by NaCl, ranging from oxidase activation to structural/mitochondrial damage [76,77,78].
The endothelium, a metabolically active layer of cells, acts as a selective barrier separating the vascular wall and circulating blood [79]. Its intricate role includes functioning as an active endocrine, paracrine, and autocrine organ necessary for maintaining vascular homeostasis [80,81,82]. It is well documented that higher supraphysiological concentrations of AngII increase ROS production via NADPH-oxidase [83,84]. Even at low concentrations, such as 10−7 mol/L, AngII induces protein nitration in endothelial cells, with the extent of nitration increasing in a concentration-dependent manner as the concentration of AngII rises [85]. However, too-low levels of AngII have been shown to increase ROS in the vasculature, too [48,86,87], rendering the conclusion that a physiological range of AngII is needed to balance oxidative stress. Previously, we have shown that three days of a subpressor dose of AngII supplementation in HS-fed rats restored the FID of MCAs and significantly increased GPx4 and extracellular SOD antioxidative enzyme expression [48]. Furthermore, the HS diet significantly reduced the expression of Cu/Zn SOD and Mn SOD in the cerebral resistance arteries of the HS-fed rats, while an infusion of AngII restored protein Cu/Zn SOD (but not Mn SOD) expression [88]. A study involving the two-week infusion of AngII in sham rats and rats with myocardial infarction suggested the activation of counter-regulatory mechanisms by AngII, as blood pressure changes and vascular remodeling were minimal in the animals with myocardial infarction leading to compensation of hypertensive and growth stimulatory effects of AngII observed in sham rats. Namely, rats with myocardial infarction exhibited concomitant increases in plasma ANP and NO synthase activity following AngII infusion, demonstrating AngII-activated protective mechanisms related to blood vessel structure and function [89]. In the present study, AngII exposure in our cell cultures significantly altered metabolic activity following HS treatment after 48 h. Viability was reduced after 48 h but seemingly restored after 72 h of incubation. It has been reported that AngII exposure increases the viability of human mammary epithelial cells (184A1) through mitochondrial metabolism, as well as changes in cell behavior via AT1R overexpression [90]. Interestingly, AngII stimulates the Na/K pump in the proximal tubular cell culture and acutely stimulates the transcellular sodium transport [91], thus preventing an increase in cellular osmolality. If such an effect of AngII is present in the ECs, it remains to be investigated and demonstrates that it could also contribute a piece of the puzzle to the beneficial effect of AngII in the endothelium.
One possible explanation for an observed beneficial effect of AngII on the metabolic activity of the NaCl-stressed HAECs is the upregulation of antioxidative systems. This has been demonstrated in animal models of rats on an HS diet and in Dahl salt-sensitive rats [46,48]. In the vasculature, subpressor doses of AngII have led to restored microvascular reactivity and increased expression of antioxidative enzymes, such as SOD. Interestingly, it was previously reported that AngII, as a stress inducer, increases the expression of Nrf2-related genes in rat and mice neuronal cell lines, consequently leading to an increase in cell viability [92]. Consequently, future research should not only focus on the functions and activation pathways of Nrf2 but also explore its broader implications and applications.
The results showed that elevated levels of NaCl lead to an increase in oxidative stress and cause changes in the expression of certain CAMs. However, these changes do not occur uniformly in the same direction or at the same concentration of NaCl. This variability is likely due to the involvement of different signaling pathways, which may respond differently to NaCl, resulting in diverse effects on oxidative stress and the regulation of CAMs [13,93]. Endothelial pro-inflammatory phenotype is generated by increased pro-inflammatory cytokines production (IL-1β, IL-6, TNF-α) alongside C-reactive protein (CRP). This phenotype is characterized by an increase in CAMs, such as E-selectin, ICAM-1, and VCAM-1 [94,95,96]. Animal studies have shown upregulation in CAM expression following the HS diet [97,98,99]. Leukocyte migration and vascular adhesion mediated by ICAM-1 and VCAM-1 regulate inflammation and homeostasis in various diseases [98,100]. HS intake also stimulates the expression of E-selectin, monocyte chemotactic protein-1 (MCP-1), and endothelin 1 (ET1) [101,102]. ICAM-1 plays an important role in vascular inflammation and represents an attractive target for future treatment due to its increased expression in response to stressors [103,104]. The results of the present study showed decreased ICAM-1 expression following AngII exposure in a moderate NaCl concentration. Furthermore, endoglin is an angiogenesis/vascular remodeling participator with its soluble form that tends to be increased in inflammatory/vascular pathological conditions (atherosclerosis, hypertension, type 2 diabetes, and dyslipidaemia) and endothelial injury events [105,106,107]. In our study, endoglin expression was significantly decreased following AngII exposure in all three HS groups. This is in line with the notion that oxidative stress upregulated endoglin, e.g., in the placenta [108]. Since AngII can decrease oxidative stress, as demonstrated in this and previous studies, the reduction in endoglin levels is warranted.
While our study lays a foundation for future research on the mechanisms of AngII in relation to cell viability and activation, it has certain limitations. Subsequent studies should incorporate pharmacological experiments and explore the effects of angiotensin receptor blockers on ROS production and the expression of adhesion molecules in HAECs following AngII treatment. Indeed, in our previous work on rat animal models, we have demonstrated that the inhibition of AT1R leads to impaired endothelium-mediated vasodilation in response to changes in flow in isolated, pressurized, middle cerebral arteries and increases oxidative stress [93]. Further, results on protein expression of the AT1 and AT2 receptors in brain blood vessels (BBV) provided in Supplementary Figure (Figure S3) suggest that permissive effects of AngII on vasodilation and oxidative balance include and are mediated mainly via AT1 receptors.
One limitation of this study that may arise is the osmolarity control of the experiments that could have been performed by, e.g., mannitol or sorbitol. However, in this study, we opted not to perform this experiment due to growing evidence that increasing the osmolarity of the culture medium by the addition of sorbitol or mannitol (100 mM) did not show alteration in cellular response, as shown in neutrophils [56] or rat glial cell culture [109], but results suggested the sole effect of high sodium and not of increased osmolarity. Furthermore, the mechanisms of action and consequences of mannitol vs. NaCl are different [110], and mannitol also induces apoptosis in ECs [111]. Importantly, the effect of increased osmolarity is reversible [112], as shown in the experiments on mIMCD3 mouse renal collecting duct cells, where, with up to 600 mosmol/kg, the effect was only transient, and by 12 h at 550 mosmol/kg, the effect was reversible to normal. Thus, our control condition is the physiological concentration of NaCl (i.e., 270 mOsmol/L).
While our study focused on measuring intracellular ROS (baseline and following PMA stimulation), we acknowledge that calcium signaling, which can modulate PKC activity, was not directly assessed. This represents a limitation of our study, particularly in the context of HS conditions where calcium-dependent PKC-signaling pathways are relevant [113]. Future research should explore this aspect, including the measurement of intracellular calcium levels and their relationship to PKC activation and ROS generation.
5. Conclusions
The present study demonstrated that a longer duration and higher concentration of NaCl have detrimental effects on ECs’ metabolic activity and induce them as a source of ROS. Importantly, the present study demonstrated that AngII can decrease oxidative stress and alter the activation state of ECs, thus providing beneficial conditions for cell survival. The potential common denominator of observed effects of AngII is the upregulation of antioxidative systems. Thus, future research should focus on further elucidating the mechanisms underlying AngII-induced antioxidative defense modulation and its potential therapeutic implications.
Conceptualization, N.K. (Nikolina Kolobarić), N.K. (Nataša Kozina) and I.D.; Data curation, N.K. (Nikolina Kolobarić), N.K. (Nataša Kozina), Z.M. and I.D.; Formal analysis, N.K. (Nikolina Kolobarić), N.K. (Nataša Kozina), Z.M. and I.D.; Funding acquisition, I.D.; Investigation, N.K. (Nikolina Kolobarić), N.K. (Nataša Kozina), Z.M. and I.D.; Methodology, N.K. (Nikolina Kolobarić), N.K. (Nataša Kozina), Z.M. and I.D.; Project administration, I.D.; Resources, I.D.; Software, N.K. (Nikolina Kolobarić), N.K. (Nataša Kozina) and I.D.; Supervision, I.D.; Validation, I.D.; Visualization, N.K. (Nikolina Kolobarić), N.K. (Nataša Kozina), Z.M. and I.D.; Writing—original draft, N.K. (Nikolina Kolobarić), N.K. (Nataša Kozina), Z.M. and I.D.; Writing—review and editing, N.K. (Nikolina Kolobarić), N.K. (Nataša Kozina), Z.M. and I.D. All authors have read and agreed to the published version of the manuscript.
Not applicable.
Not applicable.
The original contributions presented in this study are included in the article/
The authors declare no conflicts of interest.
ACE | angiotensin-converting enzyme |
AngII | angiotensin II |
ANOVA | analysis of variance |
ARBs | angiotensin II receptor blockers |
AT1R | angiotensin type 1 receptor |
CAMs | cellular adhesion molecules |
CCBs | calcium channel blockers |
CO2 | carbon dioxide |
CRP | C-reactive protein |
Cu | copper |
DCFDA | dichlorofluorescein diacetate |
DCFH | 2′,7′-dichlorofluorescein |
DHE | dihydroethidium |
ET-1 | endothelin 1 |
FID | flow-induced dilation |
FLH | fluorescence height |
GMFI | geometric mean fluorescence intensity |
GPx | glutathione peroxidase |
H2O2 | hydrogen peroxide |
HAECs | human aortic endothelial cells |
HS | high salt |
HUVECs | human umbilical vein endothelial cells |
ICAM-1 | intercellular adhesion molecule 1 |
IL | interleukin |
MCAs | middle cerebral arteries |
MCP-1 | monocyte chemotactic protein-1 |
Mn | manganese |
MTT | 3-(4, 5-dimethylthiazolyl-2)-2, 5-diphenyltetrazolium bromide |
NaCl | sodium chloride |
NADPH | nicotinamide adenine dinucleotide phosphate |
Na/K | sodium/potassium pump |
NO | nitric oxide |
O2.− | superoxide anion |
ONOO− | peroxynitrite |
PBS | phosphate-buffered saline |
PKC | protein kinase C |
PMA | phorbol 12-myristate 13-acetate |
Prx | peroxiredoxin |
ROS | reactive oxygen species |
SD | Sprague-Dawley |
SOD | superoxide dismutase |
Th17 | T helper 17 cells |
TNF-α | tumor necrosis factor alpha |
Treg | T regulatory cells |
VCAM-1 | vascular adhesion molecule 1 |
Zn | zinc |
Footnotes
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.
Figure 1. Cellular metabolic activity of HAECs after treatment with different concentrations of NaCl and AngII assessed via MTT assay. A—absorbance; nm—nanometers; CTRL—control; HS—high salt; AngII—angiotensin II; One-way ANOVA; *,+ significance level p [less than] 0.05 (* compared to control group; + compared to HS group before AngII).
Figure 2. Formation of hydrogen peroxide and peroxynitrite in HAECs following high-salt treatment accompanied by AngII exposure (A). Representative histogram overlay (B) and stacked histograms (C). Grey color representing CTRL group, blue color representing HS320 group, red color representing HS350 group. Results are expressed as geometric mean fluorescence intensity (GMFI). DCF-DA—dichlorofluorescein diacetate; CTRL—control; HS—high salt; One-way ANOVA; *,+ significance level p [less than] 0.05 (* compared to HS group before AngII; + compared to control group).
Figure 3. Formation of superoxide in HAECs following high-salt treatment accompanied by AngII exposure (A). Representative histogram overlay (B) and stacked histograms (C). Grey color representing CTRL group, blue color representing HS320 group, red color representing HS350 group. Results are expressed as geometric mean fluorescence intensity (GMFI). DHE—dihydroethidium; CTRL—control; HS—high salt; One-way ANOVA; *,+ significance level p [less than] 0.05 (* compared to HS group before AngII; + compared to control group).
Figure 4. Changes in CAMs’ expression: VCAM-1 (A), ICAM-1 (B), Endoglin (C), and E-selectin (D) in HAECs following high-salt treatment accompanied by AngII exposure. Grey color representing CTRL group, blue color representing HS320 group, red color represent-ing HS350 group. Results are expressed as geometric mean fluorescence intensity (GMFI). CAMs—cell adhesion molecules; HAECs—human aortic endothelial cells; VCAM-1—vascular cell adhesion molecule 1; ICAM-1—intracellular adhesion molecule 1; HS—high salt; CTRL—control; AngII—angiotensin II; One-way ANOVA; *,+ significance level p [less than] 0.05 (* compared to HS group before AngII; + compared to control group).
Supplementary Materials
The following supporting information can be downloaded at:
References
1. Méndez-Barbero, N.; Gutiérrez-Muñoz, C.; Colio, L.M.B. Cellular Crosstalk between Endothelial and Smooth Muscle Cells in Vascular Wall Remodeling. Int. J. Mol. Sci.; 2021; 22, 7284. [DOI: https://dx.doi.org/10.3390/ijms22147284] [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/34298897]
2. Beck-Joseph, J.; Lehoux, S. Molecular Interactions Between Vascular Smooth Muscle Cells and Macrophages in Atherosclerosis. Front. Cardiovasc. Med.; 2021; 8, 737934. [DOI: https://dx.doi.org/10.3389/fcvm.2021.737934]
3. Versari, D.; Daghini, E.; Virdis, A.; Ghiadoni, L.; Taddei, S. Endothelial dysfunction as a target for prevention of cardiovascular disease. Diabetes Care; 2009; 32, (Suppl. S2), pp. S314-S321. [DOI: https://dx.doi.org/10.2337/dc09-S330]
4. Widlansky, M.E.; Gokce, N.; Keaney, J.F.; Vita, J.A. The clinical implications of endothelial dysfunction. J. Am. Coll. Cardiol.; 2003; 42, pp. 1149-1160. [DOI: https://dx.doi.org/10.1016/S0735-1097(03)00994-X]
5. Fisher, M. Injuries to the vascular endothelium: Vascular wall and endothelial dysfunction. Rev. Neurol. Dis.; 2008; 5, (Suppl. S1), pp. S4-S11. [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/18645570]
6. Huang, Y.; Song, C.; He, J.; Li, M. Research progress in endothelial cell injury and repair. Front. Pharmacol.; 2022; 13, 997272. [DOI: https://dx.doi.org/10.3389/fphar.2022.997272]
7. Sun, H.-J.; Wu, Z.-Y.; Nie, X.-W.; Bian, J.-S. Role of Endothelial Dysfunction in Cardiovascular Diseases: The Link Between Inflammation and Hydrogen Sulfide. Front. Pharmacol.; 2020; 10, 1568. [DOI: https://dx.doi.org/10.3389/fphar.2019.01568] [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/32038245]
8. Widmer, R.J.; Lerman, A. Endothelial dysfunction and cardiovascular disease. Glob. Cardiol. Sci. Pract.; 2014; 2014, pp. 291-308. [DOI: https://dx.doi.org/10.5339/gcsp.2014.43]
9. Little, P.J.; Askew, C.D.; Xu, S.; Kamato, D. Endothelial Dysfunction and Cardiovascular Disease: History and Analysis of the Clinical Utility of the Relationship. Biomedicines; 2021; 9, 699. [DOI: https://dx.doi.org/10.3390/biomedicines9060699]
10. Cai, H.; Harrison, D.G. Endothelial Dysfunction in Cardiovascular Diseases: The Role of Oxidant Stress. Circ. Res.; 2000; 87, pp. 840-844. [DOI: https://dx.doi.org/10.1161/01.RES.87.10.840]
11. Calila, H.; Bălășescu, E.; Nedelcu, R.I.; Ion, D.A. Endothelial Dysfunction as a Key Link between Cardiovascular Disease and Frailty: A Systematic Review. J. Clin. Med.; 2024; 13, 2686. [DOI: https://dx.doi.org/10.3390/jcm13092686] [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/38731215]
12. Ertuglu, L.A.; Mutchler, A.P.; Yu, J.; Kirabo, A. Inflammation and oxidative stress in salt sensitive hypertension; The role of the NLRP3 inflammasome. Front. Physiol.; 2022; 13, 1096296. [DOI: https://dx.doi.org/10.3389/fphys.2022.1096296] [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/36620210]
13. Knezović, A.; Kolobarić, N.; Drenjančević, I.; Mihaljević, Z.; Šušnjara, P.; Jukić, I.; Stupin, M.; Kibel, A.; Marczi, S.; Mihalj, M. et al. Role of Oxidative Stress in Vascular Low-Grade Inflammation Initiation Due to Acute Salt Loading in Young Healthy Individuals. Antioxidants; 2022; 11, 444. [DOI: https://dx.doi.org/10.3390/antiox11030444] [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/35326095]
14. Mihalj, M.; Matić, A.; Mihaljević, Z.; Barić, L.; Stupin, A.; Drenjančević, I. Short-Term High-NaCl Dietary Intake Changes Leukocyte Expression of VLA-4, LFA-1, and Mac-1 Integrins in Both Healthy Humans and Sprague-Dawley Rats: A Comparative Study. Mediat. Inflamm.; 2019; 2019, e6715275. [DOI: https://dx.doi.org/10.1155/2019/6715275]
15. Tolj, I.; Stupin, A.; Drenjančević, I.; Šušnjara, P.; Perić, L.; Stupin, M. The Role of Nitric Oxide in the Micro- and Macrovascular Response to a 7-Day High-Salt Diet in Healthy Individuals. Int. J. Mol. Sci.; 2023; 24, 7157. [DOI: https://dx.doi.org/10.3390/ijms24087157]
16. Wu, Q.; Burley, G.; Li, L.; Lin, S.; Shi, Y. The role of dietary salt in metabolism and energy balance: Insights beyond cardiovascular disease. Diabetes Obes. Metab.; 2023; 25, pp. 1147-1161. [DOI: https://dx.doi.org/10.1111/dom.14980]
17. Barić, L.; Drenjančević, I.; Matić, A.; Stupin, M.; Kolar, L.; Mihaljević, Z.; Lenasi, H.; Šerić, V.; Stupin, A. Seven-Day Salt Loading Impairs Microvascular Endothelium-Dependent Vasodilation without Changes in Blood Pressure, Body Composition and Fluid Status in Healthy Young Humans. Kidney Blood Press. Res.; 2019; 44, pp. 835-847. [DOI: https://dx.doi.org/10.1159/000501747] [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/31430746]
18. Yuan, M.; Yan, D.; Wang, Y.; Qi, M.; Li, K.; Lv, Z.; Gao, D.; Ning, N. Sodium intake and the risk of heart failure and hypertension: Epidemiological and Mendelian randomization analysis. Front. Nutr.; 2024; 10, 1263554. [DOI: https://dx.doi.org/10.3389/fnut.2023.1263554]
19. Stamler, J. The INTERSALT Study: Background, methods, findings, and implications. Am. J. Clin. Nutr.; 1997; 65, pp. 626S-642S. [DOI: https://dx.doi.org/10.1093/ajcn/65.2.626S]
20. He, J.; Whelton, P.K. Commentary: Salt intake, hypertension and risk of cardiovascular disease: An important public health challenge. Int. J. Epidemiol.; 2002; 31, pp. 327-331. [DOI: https://dx.doi.org/10.1093/ije/31.2.327]
21. Strazzullo, P.; D’Elia, L.; Kandala, N.-B.; Cappuccio, F.P. Salt intake, stroke, and cardiovascular disease: Meta-analysis of prospective studies. Br. Med. J. BMJ; 2009; 339, b4567. [DOI: https://dx.doi.org/10.1136/bmj.b4567]
22. Wang, Y.-J.; Yeh, T.-L.; Shih, M.-C.; Tu, Y.-K.; Chien, K.-L. Dietary Sodium Intake and Risk of Cardiovascular Disease: A Systematic Review and Dose-Response Meta-Analysis. Nutrients; 2020; 12, 2934. [DOI: https://dx.doi.org/10.3390/nu12102934] [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/32992705]
23. Qi, H.; Liu, Z.; Cao, H.; Sun, W.-P.; Peng, W.-J.; Liu, B.; Dong, S.-J.; Xiang, Y.-T.; Zhang, L. Comparative Efficacy of Antihypertensive Agents in Salt-Sensitive Hypertensive Patients: A Network Meta-Analysis. Am. J. Hypertens.; 2018; 31, pp. 835-846. [DOI: https://dx.doi.org/10.1093/ajh/hpy027]
24. Vargas-Meza, J.; Nilson, E.A.F.; Nieto, C.; Khandpur, N.; Denova-Gutiérrez, E.; Valero-Morales, I.; Barquera, S.; Campos-Nonato, I. Modelling the impact of sodium intake on cardiovascular disease mortality in Mexico. BMC Public Health; 2023; 23, 983. [DOI: https://dx.doi.org/10.1186/s12889-023-15827-0] [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/37237296]
25. Williams, B.; Mancia, G.; Spiering, W.; Agabiti Rosei, E.; Azizi, M.; Burnier, M.; Clement, D.L.; Coca, A.; de Simone, G.; Dominiczak, A. et al. 2018 ESC/ESH Guidelines for the management of arterial hypertension: The Task Force for the management of arterial hypertension of the European Society of Cardiology and the European Society of Hypertension. J. Hypertens.; 2018; 36, 1953. [DOI: https://dx.doi.org/10.1097/HJH.0000000000001940] [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/30234752]
26. Riegel, B.; Lee, S.; Hill, J.; Daus, M.; Baah, F.O.; Wald, J.W.; Knafl, G.J. Patterns of adherence to diuretics, dietary sodium and fluid intake recommendations in adults with heart failure. Heart Lung J. Crit. Care; 2019; 48, pp. 179-185. [DOI: https://dx.doi.org/10.1016/j.hrtlng.2018.12.008]
27. He, F.J.; Li, J.; Macgregor, G.A. Effect of longer term modest salt reduction on blood pressure: Cochrane systematic review and meta-analysis of randomised trials. BMJ Clin. Res. Ed.; 2013; 346, f1325. [DOI: https://dx.doi.org/10.1136/bmj.f1325]
28. Jablonski, K.L.; Racine, M.L.; Geolfos, C.J.; Gates, P.E.; Chonchol, M.; McQueen, M.B.; Seals, D.R. Dietary sodium restriction reverses vascular endothelial dysfunction in middle-aged/older adults with moderately elevated systolic blood pressure. J. Am. Coll. Cardiol.; 2013; 61, pp. 335-343. [DOI: https://dx.doi.org/10.1016/j.jacc.2012.09.010]
29. Heart Outcomes Prevention Evaluation Study Investigators Yusuf, S.; Sleight, P.; Pogue, J.; Bosch, J.; Davies, R.; Dagenais, G. Effects of an angiotensin-converting-enzyme inhibitor, ramipril, on cardiovascular events in high-risk patients. N. Engl. J. Med.; 2000; 342, pp. 145-153. [DOI: https://dx.doi.org/10.1056/NEJM200001203420301]
30. Danchin, N.; Cucherat, M.; Thuillez, C.; Durand, E.; Kadri, Z.; Steg, P.G. Angiotensin-Converting Enzyme Inhibitors in Patients With Coronary Artery Disease and Absence of Heart Failure or Left Ventricular Systolic Dysfunction: An Overview of Long-term Randomized Controlled Trials. Arch. Intern. Med.; 2006; 166, pp. 787-796. [DOI: https://dx.doi.org/10.1001/archinte.166.7.787]
31. Solomon, S.D.; Rice, M.M.; Jablonski, A.K.; Jose, P.; Domanski, M.; Sabatine, M.; Gersh, B.J.; Rouleau, J.; Pfeffer, M.A.; Braunwald, E. Renal Function and Effectiveness of Angiotensin-Converting Enzyme Inhibitor Therapy in Patients With Chronic Stable Coronary Disease in the Prevention of Events with ACE inhibition (PEACE) Trial. Circulation; 2006; 114, pp. 26-31. [DOI: https://dx.doi.org/10.1161/CIRCULATIONAHA.105.592733] [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/16801465]
32. Yusuf, S.; Pfeffer, M.A.; Swedberg, K.; Granger, C.B.; Held, P.; McMurray, J.J.V.; Michelson, E.L.; Olofsson, B.; Ostergren, J. CHARM Investigators and Committees. Effects of candesartan in patients with chronic heart failure and preserved left-ventricular ejection fraction: The CHARM-Preserved Trial. Lancet; 2003; 362, pp. 777-781. [DOI: https://dx.doi.org/10.1016/S0140-6736(03)14285-7] [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/13678871]
33. Lithell, H.; Hansson, L.; Skoog, I.; Elmfeldt, D.; Hofman, A.; Olofsson, B.; Trenkwalder, P.; Zanchetti, A. SCOPE Study Group The Study on Cognition and Prognosis in the Elderly (SCOPE): Principal results of a randomized double-blind intervention trial. J. Hypertens.; 2003; 21, pp. 875-886. [DOI: https://dx.doi.org/10.1097/00004872-200305000-00011]
34. Dézsi, C.A. A review of clinical studies on angiotensin II receptor blockers and risk of cancer. Int. J. Cardiol.; 2014; 177, pp. 748-753. [DOI: https://dx.doi.org/10.1016/j.ijcard.2014.11.031]
35. Munger, M.A. Use of Angiotensin Receptor Blockers In Cardiovascular Protection: Current Evidence and Future Directions. Pharm. Ther.; 2011; 36, 22.
36. Suzuki, H.; Kanno, Y.; Sugahara, S.; Ikeda, N.; Shoda, J.; Takenaka, T.; Inoue, T.; Araki, R. Effect of angiotensin receptor blockers on cardiovascular events in patients undergoing hemodialysis: An open-label randomized controlled trial. Am. J. Kidney Dis. Off. J. Natl. Kidney Found.; 2008; 52, pp. 501-506. [DOI: https://dx.doi.org/10.1053/j.ajkd.2008.04.031]
37. Cummings, D.M.; Amadio, P.; Nelson, L.; Fitzgerald, J.M. The role of calcium channel blockers in the treatment of essential hypertension. Arch. Intern. Med.; 1991; 151, pp. 250-259. [DOI: https://dx.doi.org/10.1001/archinte.1991.00400020026007]
38. Jones, K.E.; Hayden, S.L.; Meyer, H.R.; Sandoz, J.L.; Arata, W.H.; Dufrene, K.; Ballaera, C.; Lopez Torres, Y.; Griffin, P.; Kaye, A.M. et al. The Evolving Role of Calcium Channel Blockers in Hypertension Management: Pharmacological and Clinical Considerations. Curr. Issues Mol. Biol.; 2024; 46, pp. 6315-6327. [DOI: https://dx.doi.org/10.3390/cimb46070377]
39. Lee, E.M. Calcium channel blockers for hypertension: Old, but still useful. Cardiovasc. Prev. Pharmacother.; 2023; 5, pp. 113-125. [DOI: https://dx.doi.org/10.36011/cpp.2023.5.e16]
40. Ogita, H.; Liao, J.K. Endothelial Function and Oxidative Stress. Endothel. J. Endothel. Cell Res.; 2004; 11, pp. 123-132. [DOI: https://dx.doi.org/10.1080/10623320490482664]
41. Barić, L.; Drenjančević, I.; Mihalj, M.; Matić, A.; Stupin, M.; Kolar, L.; Mihaljević, Z.; Mrakovčić-Šutić, I.; Šerić, V.; Stupin, A. Enhanced Antioxidative Defense by Vitamins C and E Consumption Prevents 7-Day High-Salt Diet-Induced Microvascular Endothelial Function Impairment in Young Healthy Individuals. J. Clin. Med.; 2020; 9, 843. [DOI: https://dx.doi.org/10.3390/jcm9030843] [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/32244956]
42. Cosic, A.; Jukic, I.; Stupin, A.; Mihalj, M.; Mihaljevic, Z.; Novak, S.; Vukovic, R.; Drenjancevic, I. Attenuated flow-induced dilatation of middle cerebral arteries is related to increased vascular oxidative stress in rats on a short-term high salt diet. J. Physiol.; 2016; 594, pp. 4917-4931. [DOI: https://dx.doi.org/10.1113/JP272297]
43. Fyhrquist, F.; Metsärinne, K.; Tikkanen, I. Role of angiotensin II in blood pressure regulation and in the pathophysiology of cardiovascular disorders. J. Hum. Hypertens.; 1995; 9, (Suppl. S5), pp. S19-S24.
44. Patel, P.; Sanghavi, D.K.; Morris, D.L.; Kahwaji, C.I. Angiotensin II. StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2024.
45. Timmermans, P.B.M.W.M.; Benfield, P.; Chiu, A.T.; Herblin, W.F.; Wong, P.C.; Smith, R.D. Angiotensin II Receptors and Functional Correlates. Am. J. Hypertens.; 1992; 5, pp. 221S-235S. [DOI: https://dx.doi.org/10.1093/ajh/5.12.221S]
46. Zhu, J.; Drenjancevic-Peric, I.; McEwen, S.; Friesema, J.; Schulta, D.; Yu, M.; Roman, R.J.; Lombard, J.H. Role of superoxide and angiotensin II suppression in salt-induced changes in endothelial Ca2+ signaling and NO production in rat aorta. Am. J. Physiol. Heart Circ. Physiol.; 2006; 291, pp. H929-H938. [DOI: https://dx.doi.org/10.1152/ajpheart.00692.2005]
47. Drenjančević-Perić, I.; Jelaković, B.; Lombard, J.H.; Kunert, M.P.; Kibel, A.; Gros, M. High-Salt Diet and Hypertension: Focus on the Renin-Angiotensin System. Kidney Blood Press. Res.; 2011; 34, pp. 1-11. [DOI: https://dx.doi.org/10.1159/000320387] [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/21071956]
48. Matic, A.; Jukic, I.; Mihaljevic, Z.; Kolobaric, N.; Stupin, A.; Kozina, N.; Bujak, I.T.; Kibel, A.; Lombard, J.H.; Drenjancevic, I. Low-dose angiotensin II supplementation restores flow-induced dilation mechanisms in cerebral arteries of Sprague-Dawley rats on a high salt diet. J. Hypertens.; 2022; 40, 441. [DOI: https://dx.doi.org/10.1097/HJH.0000000000003030] [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/34845157]
49. Mihalj, M.; Štefanić, M.; Mihaljević, Z.; Kolobarić, N.; Jukić, I.; Stupin, A.; Matić, A.; Frkanec, R.; Tavčar, B.; Horvatić, A. et al. Early Low-Grade Inflammation Induced by High-Salt Diet in Sprague Dawley Rats Involves Th17/Treg Axis Dysregulation, Vascular Wall Remodeling, and a Shift in the Fatty Acid Profile. Cell. Physiol. Biochem.; 2024; 58, pp. 83-103. [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/38459804]
50. Miyauchi, H.; Geisberger, S.; Luft, F.C.; Wilck, N.; Stegbauer, J.; Wiig, H.; Dechend, R.; Jantsch, J.; Kleinewietfeld, M.; Kempa, S. et al. Sodium as an Important Regulator of Immunometabolism. Hypertens. Dallas Tex 1979; 2024; 81, pp. 426-435. [DOI: https://dx.doi.org/10.1161/HYPERTENSIONAHA.123.19489]
51. Hunter, R.W.; Dhaun, N.; Bailey, M.A. The impact of excessive salt intake on human health. Nat. Rev. Nephrol.; 2022; 18, pp. 321-335. [DOI: https://dx.doi.org/10.1038/s41581-021-00533-0]
52. Li, J.; White, J.; Guo, L.; Zhao, X.; Wang, J.; Smart, E.J.; Li, X.-A. Salt inactivates endothelial nitric oxide synthase in endothelial cells. J. Nutr.; 2009; 139, pp. 447-451. [DOI: https://dx.doi.org/10.3945/jn.108.097451] [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/19176751]
53. Pap, D.; Pajtók, C.; Veres-Székely, A.; Szebeni, B.; Szász, C.; Bokrossy, P.; Zrufkó, R.; Vannay, Á.; Tulassay, T.; Szabó, A.J. High Salt Promotes Inflammatory and Fibrotic Response in Peritoneal Cells. Int. J. Mol. Sci.; 2023; 24, 13765. [DOI: https://dx.doi.org/10.3390/ijms241813765] [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/37762068]
54. Yamakami, Y.; Yonekura, R.; Matsumoto, Y.; Takauji, Y.; Miki, K.; Fujii, M.; Ayusawa, D. High concentrations of NaCl induce cell swelling leading to senescence in human cells. Mol. Cell. Biochem.; 2016; 411, pp. 117-125. [DOI: https://dx.doi.org/10.1007/s11010-015-2573-1] [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/26463993]
55. Dmitrieva, N.I.; Burg, M.B. Elevated Sodium and Dehydration Stimulate Inflammatory Signaling in Endothelial Cells and Promote Atherosclerosis. PLoS ONE; 2015; 10, e0128870. [DOI: https://dx.doi.org/10.1371/journal.pone.0128870]
56. Mazzitelli, I.; Bleichmar, L.; Melucci, C.; Gerber, P.P.; Toscanini, A.; Cuestas, M.L.; Diaz, F.E.; Geffner, J. High Salt Induces a Delayed Activation of Human Neutrophils. Front. Immunol.; 2022; 13, 831844. [DOI: https://dx.doi.org/10.3389/fimmu.2022.831844]
57. Ju, H.K.; Hwang, S.-J.; Jeon, C.-J.; Lee, G.M.; Yoon, S.K. Use of NaCl prevents aggregation of recombinant COMP–Angiopoietin-1 in Chinese hamster ovary cells. J. Biotechnol.; 2009; 143, pp. 145-150. [DOI: https://dx.doi.org/10.1016/j.jbiotec.2009.06.017]
58. Jakic, B.; Buszko, M.; Cappellano, G.; Wick, G. Elevated sodium leads to the increased expression of HSP60 and induces apoptosis in HUVECs. PLoS ONE; 2017; 12, e0179383. [DOI: https://dx.doi.org/10.1371/journal.pone.0179383]
59. Shiwakoti, S.; Adhikari, D.; Lee, J.P.; Kang, K.-W.; Lee, I.-S.; Kim, H.J.; Oak, M.-H. Prevention of Fine Dust-Induced Vascular Senescence by Humulus lupulus Extract and Its Major Bioactive Compounds. Antioxidants; 2020; 9, 1243. [DOI: https://dx.doi.org/10.3390/antiox9121243]
60. Kolar, L.; Šušnjara, P.; Stupin, M.; Stupin, A.; Jukić, I.; Mihaljević, Z.; Kolobarić, N.; Bebek, I.; Nejašmić, D.; Lovrić, M. et al. Enhanced Microvascular Adaptation to Acute Physical Stress and Reduced Oxidative Stress in Male Athletes Who Consumed Chicken Eggs Enriched with n-3 Polyunsaturated Fatty Acids and Antioxidants-Randomized Clinical Trial. Life; 2023; 13, 2140. [DOI: https://dx.doi.org/10.3390/life13112140]
61. Kong, J.; Hu, X.-M.; Cai, W.-W.; Wang, Y.-M.; Chi, C.-F.; Wang, B. Bioactive Peptides from Skipjack Tuna Cardiac Arterial Bulbs (II): Protective Function on UVB-Irradiated HaCaT Cells through Antioxidant and Anti-Apoptotic Mechanisms. Mar. Drugs; 2023; 21, 105. [DOI: https://dx.doi.org/10.3390/md21020105]
62. Barthelmes, J.; Nägele, M.P.; Ludovici, V.; Ruschitzka, F.; Sudano, I.; Flammer, A.J. Endothelial dysfunction in cardiovascular disease and Flammer syndrome—Similarities and differences. EPMA J.; 2017; 8, pp. 99-109. [DOI: https://dx.doi.org/10.1007/s13167-017-0099-1] [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/28824736]
63. Lippi, M.; Stadiotti, I.; Pompilio, G.; Sommariva, E. Human Cell Modeling for Cardiovascular Diseases. Int. J. Mol. Sci.; 2020; 21, 6388. [DOI: https://dx.doi.org/10.3390/ijms21176388] [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/32887493]
64. Ristow, M.; Schmeisser, K. Mitohormesis: Promoting Health and Lifespan by Increased Levels of Reactive Oxygen Species (ROS). Dose-Response; 2014; 12, pp. 288-341. [DOI: https://dx.doi.org/10.2203/dose-response.13-035.Ristow] [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/24910588]
65. Schulz, E.; Gori, T.; Münzel, T. Oxidative stress and endothelial dysfunction in hypertension. Hypertens. Res.; 2011; 34, pp. 665-673. [DOI: https://dx.doi.org/10.1038/hr.2011.39]
66. Burg, M.B.; Ferraris, J.D.; Dmitrieva, N.I. Cellular Response to Hyperosmotic Stresses. Physiol. Rev.; 2007; 87, pp. 1441-1474. [DOI: https://dx.doi.org/10.1152/physrev.00056.2006]
67. Calabrese, E.J. Hormesis: A fundamental concept in biology. Microb. Cell; 2014; 1, pp. 145-149. [DOI: https://dx.doi.org/10.15698/mic2014.05.145]
68. Calabrese, E.J.; Baldwin, L.A. The Hormetic Dose-Response Model Is More Common than the Threshold Model in Toxicology. Toxicol. Sci.; 2003; 71, pp. 246-250. [DOI: https://dx.doi.org/10.1093/toxsci/71.2.246] [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/12563110]
69. Calabrese, E.J.; Mattson, M.P. How does hormesis impact biology, toxicology, and medicine?. NPJ Aging Mech. Dis.; 2017; 3, 13. [DOI: https://dx.doi.org/10.1038/s41514-017-0013-z]
70. Calabrese, V.; Cornelius, C.; Dinkova-Kostova, A.T.; Calabrese, E.J.; Mattson, M.P. Cellular Stress Responses, The Hormesis Paradigm, and Vitagenes: Novel Targets for Therapeutic Intervention in Neurodegenerative Disorders. Antioxid. Redox Signal.; 2010; 13, pp. 1763-1811. [DOI: https://dx.doi.org/10.1089/ars.2009.3074]
71. Kunsch, C.; Medford, R.M. Oxidative Stress as a Regulator of Gene Expression in the Vasculature. Circ. Res.; 1999; 85, pp. 753-766. [DOI: https://dx.doi.org/10.1161/01.RES.85.8.753]
72. Kültz, D.; Chakravarty, D. Hyperosmolality in the form of elevated NaCl but not urea causes DNA damage in murine kidney cells. Proc. Natl. Acad. Sci. USA; 2001; 98, pp. 1999-2004. [DOI: https://dx.doi.org/10.1073/pnas.98.4.1999] [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/11172065]
73. Dmitrieva, N.I.; Cai, Q.; Burg, M.B. Cells adapted to high NaCl have many DNA breaks and impaired DNA repair both in cell culture and in vivo. Proc. Natl. Acad. Sci. USA; 2004; 101, pp. 2317-2322. [DOI: https://dx.doi.org/10.1073/pnas.0308463100] [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/14983007]
74. Mukherjee, S.; Dutta, A.; Chakraborty, A. External modulators and redox homeostasis: Scenario in radiation-induced bystander cells. Mutat. Res. Mutat. Res.; 2021; 787, 108368. [DOI: https://dx.doi.org/10.1016/j.mrrev.2021.108368] [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/34083032]
75. Xu, S.; He, Y.; Vokurkova, M.; Touyz, R.M. Endothelial Cells Negatively Modulate Reactive Oxygen Species Generation in Vascular Smooth Muscle Cells. Hypertension; 2009; 54, pp. 427-433. [DOI: https://dx.doi.org/10.1161/HYPERTENSIONAHA.109.133983]
76. Ali, T.; Li, D.; Ponnamperumage, T.N.F.; Peterson, A.K.; Pandey, J.; Fatima, K.; Brzezinski, J.; Jakusz, J.A.R.; Gao, H.; Koelsch, G.E. et al. Generation of Hydrogen Peroxide in Cancer Cells: Advancing Therapeutic Approaches for Cancer Treatment. Cancers; 2024; 16, 2171. [DOI: https://dx.doi.org/10.3390/cancers16122171]
77. Andrés, C.M.C.; Pérez de la Lastra, J.M.; Juan, C.A.; Plou, F.J.; Pérez-Lebeña, E. Chemistry of Hydrogen Peroxide Formation and Elimination in Mammalian Cells, and Its Role in Various Pathologies. Stresses; 2022; 2, pp. 256-274. [DOI: https://dx.doi.org/10.3390/stresses2030019]
78. Nauseef, W. Detection of superoxide anion and hydrogen peroxide production by cellular NADPH oxidases. Biochim. Biophys. Acta; 2013; 1840, pp. 757-767. [DOI: https://dx.doi.org/10.1016/j.bbagen.2013.04.040]
79. Pahwa, R.; Goyal, A.; Bansal, P.; Jialal, I. Chronic Inflammation. StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2020.
80. Bonetti, P.O.; Lerman, L.O.; Lerman, A. Endothelial Dysfunction. Arterioscler. Thromb. Vasc. Biol.; 2003; 23, pp. 168-175. [DOI: https://dx.doi.org/10.1161/01.ATV.0000051384.43104.FC] [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/12588755]
81. Hadi, H.A.; Carr, C.S.; Al Suwaidi, J. Endothelial Dysfunction: Cardiovascular Risk Factors, Therapy, and Outcome. Vasc. Health Risk Manag.; 2005; 1, pp. 183-198.
82. Park, K.-H.; Park, W.J. Endothelial Dysfunction: Clinical Implications in Cardiovascular Disease and Therapeutic Approaches. J. Korean Med. Sci.; 2015; 30, pp. 1213-1225. [DOI: https://dx.doi.org/10.3346/jkms.2015.30.9.1213]
83. Nguyen Dinh Cat, A.; Montezano, A.C.; Burger, D.; Touyz, R.M. Angiotensin II, NADPH Oxidase, and Redox Signaling in the Vasculature. Antioxid. Redox Signal.; 2013; 19, pp. 1110-1120. [DOI: https://dx.doi.org/10.1089/ars.2012.4641] [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/22530599]
84. Frey, R.S.; Ushio-Fukai, M.; Malik, A.B. NADPH Oxidase-Dependent Signaling in Endothelial Cells: Role in Physiology and Pathophysiology. Antioxid. Redox Signal.; 2009; 11, pp. 791-810. [DOI: https://dx.doi.org/10.1089/ars.2008.2220] [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/18783313]
85. Mihm, M.J.; Wattanapitayakul, S.K.; Piao, S.-F.; Hoyt, D.G.; Bauer, J.A. Effects of angiotensin II on vascular endothelial cells: Formation of receptor-mediated reactive nitrogen species. Biochem. Pharmacol.; 2003; 65, pp. 1189-1197. [DOI: https://dx.doi.org/10.1016/S0006-2952(03)00012-1]
86. Drenjancevic-Peric, I.; Lombard, J.H. Reduced Angiotensin II and Oxidative Stress Contribute to Impaired Vasodilation in Dahl Salt-Sensitive Rats on Low-Salt Diet. Hypertension; 2005; 45, pp. 687-691. [DOI: https://dx.doi.org/10.1161/01.HYP.0000154684.40599.03]
87. Drenjancevic-Peric, I.; Frisbee, J.C.; Lombard, J.H. Skeletal Muscle Arteriolar Reactivity in SS.BN13 Consomic Rats and Dahl Salt-Sensitive Rats. Hypertension; 2003; 41, pp. 1012-1015. [DOI: https://dx.doi.org/10.1161/01.HYP.0000067061.26899.3E] [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/12682080]
88. Durand, M.J.; Lombard, J.H. Low-Dose Angiotensin II Infusion Restores Vascular Function in Cerebral Arteries of High Salt–Fed Rats by Increasing Copper/Zinc Superoxide Dimutase Expression. Am. J. Hypertens.; 2013; 26, 739. [DOI: https://dx.doi.org/10.1093/ajh/hpt015]
89. Heeneman, S.; Smits, J.F.M.; Leenders, P.J.A.; Schiffers, P.M.H.; Daemen, M.J.A.P. Effects of Angiotensin II on Cardiac Function and Peripheral Vascular Structure During Compensated Heart Failure in the Rat. Arterioscler. Thromb. Vasc. Biol.; 1997; 17, pp. 1985-1994. [DOI: https://dx.doi.org/10.1161/01.ATV.17.10.1985]
90. Piastowska-Ciesielska, A.W.; Domińska, K.; Nowakowska, M.; Gajewska, M.; Gajos-Michniewicz, A.; Ochędalski, T. Angiotensin modulates human mammary epithelial cell motility. J. Renin Angiotensin Aldosterone Syst.; 2014; 15, pp. 419-429. [DOI: https://dx.doi.org/10.1177/1470320313475904]
91. Hanna, F.S.; Alkhouri, S.; Rajagopalan, C.; Ji, K.; Mattingly, R.R.; Yingst, D.R. Ang II acutely stimulates Na,K-pump in cells from proximal tubules by increasing its phosphorylation at S938 via a PI3K/AKT pathway. Physiol. Rep.; 2022; 10, e15508. [DOI: https://dx.doi.org/10.14814/phy2.15508]
92. Parga, J.A.; Rodriguez-Perez, A.I.; Garcia-Garrote, M.; Rodriguez-Pallares, J.; Labandeira-Garcia, J.L. Angiotensin II induces oxidative stress and upregulates neuroprotective signaling from the NRF2 and KLF9 pathway in dopaminergic cells. Free Radic. Biol. Med.; 2018; 129, pp. 394-406. [DOI: https://dx.doi.org/10.1016/j.freeradbiomed.2018.10.409]
93. Jukic, I.; Mihaljevic, Z.; Matic, A.; Mihalj, M.; Kozina, N.; Selthofer-Relatic, K.; Mihaljevic, D.; Koller, A.; Tartaro Bujak, I.; Drenjancevic, I. Angiotensin II type 1 receptor is involved in flow-induced vasomotor responses of isolated middle cerebral arteries: Role of oxidative stress. Am. J. Physiol. Heart Circ. Physiol.; 2021; 320, pp. H1609-H1624. [DOI: https://dx.doi.org/10.1152/ajpheart.00620.2020] [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/33666506]
94. Medina-Leyte, D.J.; Zepeda-García, O.; Domínguez-Pérez, M.; González-Garrido, A.; Villarreal-Molina, T.; Jacobo-Albavera, L. Endothelial Dysfunction, Inflammation and Coronary Artery Disease: Potential Biomarkers and Promising Therapeutical Approaches. Int. J. Mol. Sci.; 2021; 22, 3850. [DOI: https://dx.doi.org/10.3390/ijms22083850]
95. Theofilis, P.; Sagris, M.; Oikonomou, E.; Antonopoulos, A.S.; Siasos, G.; Tsioufis, C.; Tousoulis, D. Inflammatory Mechanisms Contributing to Endothelial Dysfunction. Biomedicines; 2021; 9, 781. [DOI: https://dx.doi.org/10.3390/biomedicines9070781] [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/34356845]
96. Nafisa, A.; Gray, S.G.; Cao, Y.; Wang, T.; Xu, S.; Wattoo, F.H.; Barras, M.; Cohen, N.; Kamato, D.; Little, P.J. Endothelial function and dysfunction: Impact of metformin. Pharmacol. Ther.; 2018; 192, pp. 150-162. [DOI: https://dx.doi.org/10.1016/j.pharmthera.2018.07.007] [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/30056057]
97. Takahashi, H.; Nakagawa, S.; Wu, Y.; Kawabata, Y.; Numabe, A.; Yanagi, Y.; Tamaki, Y.; Uehara, Y.; Araie, M. A high-salt diet enhances leukocyte adhesion in association with kidney injury in young Dahl salt-sensitive rats. Hypertens. Res. Off. J. Jpn. Soc. Hypertens.; 2017; 40, pp. 912-920. [DOI: https://dx.doi.org/10.1038/hr.2017.31]
98. Kaur, R.; Singh, V.; Kumari, P.; Singh, R.; Chopra, H.; Emran, T.B. Novel insights on the role of VCAM-1 and ICAM-1: Potential biomarkers for cardiovascular diseases. Ann. Med. Surg.; 2022; 84, 104802. [DOI: https://dx.doi.org/10.1016/j.amsu.2022.104802]
99. Granger, D.N.; Senchenkova, E. Leukocyte–Endothelial Cell Adhesion. Inflammation and the Microcirculation; Morgan & Claypool Life Sciences: Willston, VT, USA, 2010.
100. Singh, V.; Kaur, R.; Kumari, P.; Pasricha, C.; Singh, R. ICAM-1 and VCAM-1: Gatekeepers in various inflammatory and cardiovascular disorders. Clin. Chim. Acta Int. J. Clin. Chem.; 2023; 548, 117487. [DOI: https://dx.doi.org/10.1016/j.cca.2023.117487]
101. Afsar, B.; Kuwabara, M.; Ortiz, A.; Yerlikaya, A.; Siriopol, D.; Covic, A.; Rodriguez-Iturbe, B.; Johnson, R.J.; Kanbay, M. Salt Intake and Immunity. Hypertension; 2018; 72, pp. 19-23. [DOI: https://dx.doi.org/10.1161/HYPERTENSIONAHA.118.11128]
102. Ma, P.; Li, G.; Jiang, X.; Shen, X.; Li, H.; Yang, L.; Liu, W. NFAT5 directs hyperosmotic stress-induced fibrin deposition and macrophage infiltration via PAI-1 in endothelium. Aging; 2020; 13, pp. 3661-3679. [DOI: https://dx.doi.org/10.18632/aging.202330]
103. Bui, T.M.; Wiesolek, H.L.; Sumagin, R. ICAM-1: A master regulator of cellular responses in inflammation, injury resolution, and tumorigenesis. J. Leukoc. Biol.; 2020; 108, pp. 787-799. [DOI: https://dx.doi.org/10.1002/JLB.2MR0220-549R]
104. Muro, S.; Gajewski, C.; Koval, M.; Muzykantov, V.R. ICAM-1 recycling in endothelial cells: A novel pathway for sustained intracellular delivery and prolonged effects of drugs. Blood; 2005; 105, pp. 650-658. [DOI: https://dx.doi.org/10.1182/blood-2004-05-1714] [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/15367437]
105. Gallardo-Vara, E.; Gamella-Pozuelo, L.; Perez-Roque, L.; Bartha, J.L.; Garcia-Palmero, I.; Casal, J.I.; López-Novoa, J.M.; Pericacho, M.; Bernabeu, C. Potential Role of Circulating Endoglin in Hypertension via the Upregulated Expression of BMP4. Cells; 2020; 9, 988. [DOI: https://dx.doi.org/10.3390/cells9040988] [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/32316263]
106. Rossi, E.; Bernabeu, C.; Smadja, D.M. Endoglin as an Adhesion Molecule in Mature and Progenitor Endothelial Cells: A Function Beyond TGF-β. Front. Med.; 2019; 6, 10. [DOI: https://dx.doi.org/10.3389/fmed.2019.00010] [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/30761306]
107. Schoonderwoerd, M.J.A.; Goumans, M.-J.T.H.; Hawinkels, L.J.A.C. Endoglin: Beyond the Endothelium. Biomolecules; 2020; 10, 289. [DOI: https://dx.doi.org/10.3390/biom10020289]
108. Jena, M.K.; Sharma, N.R.; Petitt, M.; Maulik, D.; Nayak, N.R. Pathogenesis of Preeclampsia and Therapeutic Approaches Targeting the Placenta. Biomolecules; 2020; 10, 953. [DOI: https://dx.doi.org/10.3390/biom10060953]
109. Arimochi, H.; Morita, K. High salt culture conditions suppress proliferation of rat C6 glioma cell by arresting cell-cycle progression at S-phase. J. Mol. Neurosci.; 2005; 27, pp. 293-301. [DOI: https://dx.doi.org/10.1385/JMN:27:3:293]
110. Kinsman, B.J.; Browning, K.N.; Stocker, S.D. NaCl and osmolarity produce different responses in organum vasculosum of the lamina terminalis neurons, sympathetic nerve activity and blood pressure. J. Physiol.; 2017; 595, pp. 6187-6201. [DOI: https://dx.doi.org/10.1113/JP274537] [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/28678348]
111. Malek, A.M.; Goss, G.G.; Jiang, L.; Izumo, S.; Alper, S.L. Mannitol at Clinical Concentrations Activates Multiple Signaling Pathways and Induces Apoptosis in Endothelial Cells. Stroke; 1998; 29, pp. 2631-2640. [DOI: https://dx.doi.org/10.1161/01.STR.29.12.2631]
112. Michea, L.; Ferguson, D.R.; Peters, E.M.; Andrews, P.M.; Kirby, M.R.; Burg, M.B. Cell cycle delay and apoptosis are induced by high salt and urea in renal medullary cells. Am. J. Physiol. Ren. Physiol.; 2000; 278, pp. F209-F218. [DOI: https://dx.doi.org/10.1152/ajprenal.2000.278.2.F209]
113. Li, W.; Lv, J.; Wu, J.; Zhou, X.; Jiang, L.; Zhu, X.; Tu, Q.; Tang, J.; Liu, Y.; He, A. et al. Maternal high-salt diet altered PKC/MLC20 pathway and increased ANG II receptor-mediated vasoconstriction in adult male rat offspring. Mol. Nutr. Food Res.; 2016; 60, pp. 1684-1694. [DOI: https://dx.doi.org/10.1002/mnfr.201500998]
You have requested "on-the-fly" machine translation of selected content from our databases. This functionality is provided solely for your convenience and is in no way intended to replace human translation. Show full disclaimer
Neither ProQuest nor its licensors make any representations or warranties with respect to the translations. The translations are automatically generated "AS IS" and "AS AVAILABLE" and are not retained in our systems. PROQUEST AND ITS LICENSORS SPECIFICALLY DISCLAIM ANY AND ALL EXPRESS OR IMPLIED WARRANTIES, INCLUDING WITHOUT LIMITATION, ANY WARRANTIES FOR AVAILABILITY, ACCURACY, TIMELINESS, COMPLETENESS, NON-INFRINGMENT, MERCHANTABILITY OR FITNESS FOR A PARTICULAR PURPOSE. Your use of the translations is subject to all use restrictions contained in your Electronic Products License Agreement and by using the translation functionality you agree to forgo any and all claims against ProQuest or its licensors for your use of the translation functionality and any output derived there from. Hide full disclaimer
© 2024 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/). Notwithstanding the ProQuest Terms and Conditions, you may use this content in accordance with the terms of the License.
Abstract
Background/Objectives: Increased sodium chloride (NaCl) intake led to leukocyte activation and impaired vasodilatation via increased oxidative stress in human/animal models. Interestingly, subpressor doses of angiotensin II (AngII) restored endothelium-dependent vascular reactivity, which was impaired in a high-salt (HS) diet in animal models. Therefore, the present study aimed to assess the effects of AngII exposure following high salt (HS) loading on endothelial cells’ (ECs’) viability, activation, and reactive oxygen species (ROS) production. Methods: The fifth passage of human aortic endothelial cells (HAECs) was cultured for 24, 48, and 72 h with NaCl, namely, the control (270 mOsmol/kg), HS320 (320 mOsmol/kg), and HS350 (350 mOsmol/kg). AngII was administered at the half-time of the NaCl incubation (10−4–10−7 mol/L). Results: The cell viability was significantly reduced after 24 h in the HS350 group and in all groups after longer incubation. AngII partly preserved the viability in the HAECs with shorter exposure and lower concentrations of NaCl. Intracellular hydrogen peroxide (H2O2) and peroxynitrite (ONOO−) significantly increased in the HS320 group following AngII exposure compared to the control, while it decreased in the HS350 group compared to the HS control. A significant decrease in superoxide anion (O2.−) formation was observed following AngII exposure at 10−5, 10−6, and 10−7 mol/L for both HS groups. There was a significant decrease in intracellular adhesion molecule 1 (ICAM-1) and endoglin expression in both groups following treatment with 10−4 and 10−5 mol/L of AngII. Conclusions: The results demonstrated that AngII significantly reduced ROS production at HS350 concentrations and modulated the viability, proliferation, and activation states in ECs.
You have requested "on-the-fly" machine translation of selected content from our databases. This functionality is provided solely for your convenience and is in no way intended to replace human translation. Show full disclaimer
Neither ProQuest nor its licensors make any representations or warranties with respect to the translations. The translations are automatically generated "AS IS" and "AS AVAILABLE" and are not retained in our systems. PROQUEST AND ITS LICENSORS SPECIFICALLY DISCLAIM ANY AND ALL EXPRESS OR IMPLIED WARRANTIES, INCLUDING WITHOUT LIMITATION, ANY WARRANTIES FOR AVAILABILITY, ACCURACY, TIMELINESS, COMPLETENESS, NON-INFRINGMENT, MERCHANTABILITY OR FITNESS FOR A PARTICULAR PURPOSE. Your use of the translations is subject to all use restrictions contained in your Electronic Products License Agreement and by using the translation functionality you agree to forgo any and all claims against ProQuest or its licensors for your use of the translation functionality and any output derived there from. Hide full disclaimer