Skip main navigation

Tetrahydrobiopterin, but Not l-Arginine, Decreases NO Synthase Uncoupling in Cells Expressing High Levels of Endothelial NO Synthase

Originally publishedhttps://doi.org/10.1161/01.HYP.0000196735.85398.0eHypertension. 2006;47:87–94

Abstract

Endothelial NO synthase (eNOS) produces superoxide when depleted of (6R)-5,6,7,8-tetrahydro-l-biopterin (BH4) and l-arginine by uncoupling the electron flow from NO production. High expression of eNOS has been reported to have beneficial effects in atherosclerotic arteries after relatively short periods of time. However, sustained high expression of eNOS may have disadvantageous vascular effects because of uncoupling. We investigated NO and reactive oxygen species (ROS) production in a microvascular endothelial cell line (bEnd.3) with sustained high eNOS expression and absent inducible NOS and neuronal NOS expression using 4,5-diaminofluorescein diacetate and diacetyldichlorofluorescein as probes, respectively. Unstimulated cells produced both NO and ROS. After stimulation with vascular endothelial growth factor (VEGF), NO and ROS production increased. VEGF-induced ROS production was even further increased by the addition of extra l-arginine. Nω-nitro-l-arginine methyl ester decreased ROS production. These findings strongly suggest that eNOS is a source of ROS in these cells. Although BH4 levels were increased as compared with another endothelial cell line, eNOS levels were >2 orders of magnitude higher. The addition of BH4 resulted in increased NO production and decreased generation of ROS, indicating that bEnd.3 cells produce ROS through eNOS uncoupling because of relative BH4 deficiency. Nevertheless, eNOS-dependent ROS production was not completely abolished by the addition of BH4, suggesting intrinsic superoxide production by eNOS. This study indicates that potentially beneficial sustained increases in eNOS expression and activity could lead to eNOS uncoupling and superoxide production as a consequence. Therefore, sustained increases of eNOS or VEGF activity should be accompanied by concomitant supplementation of BH4.

In the vasculature, NO is generated by endothelial NO synthase (eNOS), where it regulates vascular tone (reviewed in Reference1) and affects endothelial transcription.2 Reactive oxygen species (ROS) play a role in signal transduction and are involved in the regulation of the biologically effective concentration of NO.3 In vascular disease states, excessive production of ROS may overwhelm the antioxidant defense mechanisms of cells, resulting in oxidative stress.4 Interestingly, eNOS itself can produce superoxide, a process referred to as “eNOS uncoupling.”5 Reduced levels of BH4 or l-arginine lead to uncoupling of reduced nicotinamide-adenine dinucleotide phosphate (NADPH) oxidation and NO synthesis, with oxygen as terminal electron acceptor instead of l-arginine, resulting in the generation of superoxide (O2·−) by eNOS.1,6,7

The antiatherogenic actions of NO on the vessel wall suggest that increasing eNOS expression may inhibit the development of atherosclerosis. Indeed, short-term studies on upregulation of eNOS by gene transfer of recombinant eNOS demonstrated beneficial effects in atherosclerotic arteries.8–10 However, longer-term high expression of eNOS may not be as beneficial. In prehypertensive spontaneously hypertensive rats (SHR), increased eNOS expression has been observed,11 but this was associated with decreased NO release and increased superoxide production in aortic tissue, suggesting the presence of eNOS uncoupling, which may contribute to the development of hypertension and its vascular complications in the SHR.12 Moreover, studies in apolipoprotein (apoE)-deficient mice showed that chronic overexpression of eNOS accelerated atherosclerosis, which was associated with lower NO production relative to eNOS expression and enhanced superoxide production in the endothelium.13 Supplementation of BH4 in these mice reduced atherosclerotic lesion size, suggesting that in these hypercholesterolemic mice, reduced BH4 availability is involved in eNOS dysfunction during chronic eNOS overexpression.

We hypothesized that under conditions of sustained high expression of eNOS, acute stimulation of eNOS induces uncoupling of the eNOS enzyme because of a relative shortage of substrate and/or cofactors with superoxide production as a consequence. bEnd.3 cells14 chronically express high levels of eNOS protein and produce large amounts of NO in comparison with primary endothelial cells [eg, human umbilical vein endothelial cells (HUVECs)] or a human microvascular endothelial cell line (CDC.HMEC-1) while retaining the functional properties of endothelial cells. Use of bEnd.3 cells facilitates the detection of subtle differences in NO production as a consequence of treatment with agonists or antagonists. In this study, we addressed the following questions: (1) Does eNOS uncoupling occur in bEnd.3 cells?; (2) Does stimulation of bEnd.3 cells with vascular endothelial growth factor (VEGF) enhance eNOS uncoupling?; and (3) If this uncoupling occurs, is the uncoupling of eNOS because of a shortage of l-arginine, BH4, or both?

Materials and Methods

Materials

All of the drugs were purchased from Sigma, except recombinant human VEGF165 (Peprotech), diethylenetriamine NONOate (DETA/NO; Cayman Chemical), H2O2 (Merck), and BH4 (Schircks Laboratories).

Cell Culture

An immortalized bEnd.3 cell line14 was generously provided by Dr Alan Schwartz (University of Washington, St Louis, MO). Cells were cultured at 37°C in humidified 95% air-5% CO2 in DMEM supplemented with 10% FCS, 2 mmol/L glutamine, 100 IU/mL penicillin, and 100 IU/mL streptomycin (Life Technologies).

HUVECs were harvested from freshly obtained umbilical cords by use of the method described by Jaffe et al.15 The cells were cultured in fibronectin-coated T-flasks with EBM-2 (Bio-Whittaker) supplemented with 0.4% human fibroblast growth factor B, 0.1% human endothelial growth factor, 0.1% ascorbic acid, 0.1% gentamicin sulfate-amphotericin-B, 0.1% VEGF, 0.1% recombinant long R insulin–like growth factor, 0.1% heparin, 0.04% hydrocortisone (all supplements from Bio-Whittaker), 2% FCS (Life Technologies), 100 IU/mL penicillin, and 100 IU/mL streptomycin.

A human microvascular endothelial cell line (CDC.HMEC-1)16 was generously provided by Dr Edwin Ades, Francisco J. Candal (Centers for Disease Control and Prevention/National Center for infectious Diseases, Atlanta, GA), and Dr Thomas Lawley (Emory University, Atlanta, GA). CDC.HMEC-1 were cultured at 37°C in humidified 95% air-5% CO2 in MCDB 131 (Life Technologies) supplemented with 10% FCS, 10 ng/mL human endothelial growth factor, 0.05 μmol/L hydrocortisone, 10 mmol/L glutamine, 100 IU/mL penicillin, and 100 IU/mL streptomycin.

Measurement of NO: 4,5-Diaminofluorescein Diacetate Assay

To measure intracellular NO production, the cell-permeable fluorescent NO indicator 4,5-diaminofluorescein diacetate (DAF-2DA; Calbiochem) was used. bEnd.3 cells were grown to confluence in a black clear-bottom 96-well plate and serum deprived for 16 hours in DMEM containing penicillin/streptomycin and 0.1% BSA. BH4, tetrahydroneopterin (NH4), and apocynin were incubated for 16 hours during starvation of the cells. All of the solutions were prepared in Tris buffer [200 mmol/L Tris-HCl, 10 mmol/L CaCl2, 10 mmol/L MgCl2, 1.33 mmol/L NaCl, 65 mmol/L KCl, 1% (wt/vol) d-glucose, 0.1% (wt/vol) BSA, and 50 μmol/L l-arginine (pH 7.4)]. Cells were washed and incubated with 5 μmol/L DAF-2DA for 40 minutes at room temperature in the dark. After incubation, cells were washed twice and incubated for 20 minutes at 37°C in the presence or absence of inhibitors or scavengers. VEGF was added, after which fluorescence was measured every 2 minutes for 70 minutes (excitation wavelength, 485 nm; emission wavelength, 538 nm; Fluoroskan Ascent, Labsystems).

To determine the reactivity of DAF-2DA toward NO, DAF-loaded bEnd.3 cells were exposed to the exogenous NO donor DETA/NO (1 to 10 μmol/L), and fluorescence was measured. At the highest concentrations of DETA/NO, the cells were still viable as determined by Trypan blue staining (data not shown). The fluorescent signal increased linearly over time (Figure 1A). DETA/NO concentration dependently increased the DAF signal as compared with basal NO production in bEnd.3 cells (Figure 1b), demonstrating time- and concentration-dependent linearity for the DAF assay of NO production.

Figure 1. Effect of NO and H2O2 on DAF and DCF. bEnd.3 cells were loaded with DAF-2DA or CM-H2DCFDA and exposed to DETA/NO or H2O2. (A) Time-dependent effects of DETA/NO on the DAF signal. □, 0 μmol/L DETA/NO; ▵, 1 μmol/L DETA/NO; ⋄, 3 μmol/L DETA/NO; ○, 10 μmol/L DETA/NO. (B) Concentration-dependent effects of DETA/NO on the DAF signal. (C) Concentration-dependent effects of H2O2 on the DAF signal. □, H2O2; ▪, H2O2+l-NAME (30 μmol/L). (D) Concentration-dependent effects of H2O2 on the DCF signal. (E) Effect of DETA/NO on the DCF signal. *P<0.05 vs 0 μmol/L; **P<0.01 vs 0 μmol/L.

Treating bEnd.3 cells with H2O2 (1 to 10 μmol/L) increased fluorescence (Figure 1C); however, Nω-nitro-l-arginine methyl ester (l-NAME) attenuated the increase in DAF signal, implying H2O2-induced eNOS activation rather than direct oxidation of DAF-2 by H2O2.

Measurement of ROS: Diacetyldichlorofluorescein Diacetate Assay

Intracellular ROS were measured using diacetyldichlorofluorescein diacetate (CM-H2DCFDA; Molecular Probes), a nonfluorescent cell–permeable indicator for ROS. Cells were seeded in a clear 96-well plate and treated as in the DAF-2DA assay. All of the solutions were made in PBS with additions [1 mmol/L CaCl2, 0.5 mmol/L MgCl2, 0.1% (wt/vol) d-glucose, and 50 μmol/L l-arginine]. Cells were washed and incubated with 10 μmol/L CM-H2DCFDA for 30 minutes at 37°C in the dark. Interventions and measurements were done in a fashion comparable to the DAF-2DA assay.

Reactivity of CM-H2DCFDA toward H2O2 was determined by exposing bEnd.3 cells to 1 to 10 μmol/L H2O2. H2O2 induced a time- and concentration-dependent increase in the CM-H2DCFDA assay (Figure 1D). Treating bEnd.3 cells with DETA/NO (0.1 to 10 μmol/L) did not affect the DCF signal (Figure 1E), indicating that CM-H2DCFDA is not reactive toward NO.

Measurement of Biopterin Levels in Cell Lysates

Biopterin levels in lysates of bEnd.3 cells or CDC.HMEC-1 were determined as described previously.17 Briefly, cell pellets from T-flasks were lysed in cold extract buffer [50 mmol/L Tris-HCl, 1 mmol/L DTT, 1 mmol/L EDTA, and 0.4 μmol/L 6,7-dimethylpterine (pH7.4)]. Protein concentration was measured using the Pierce BCA protein assay. The whole procedure was performed in the dark. Proteins were removed by adding 10 μL of a 1:1 mixture of 1.5 mol/L HClO4 and 2 mol/L H3PO4 to 90 μL of extracts followed by centrifugation. Total biopterins [BH4, 7,8-dihydro-l-biopterin (BH2), and biopterin] were determined by acid oxidation. Therefore, 10 μL of 1% iodine in 2% KI solution was added to 90 μL protein-free supernatant. BH2 and biopterin were determined by alkali oxidation by adding 10 μL of 1 mol/L NaOH to 80 μL of extract followed by 10 μL of iodine/KI solution. Samples were incubated at room temperature for 1 hour. Alkaline-oxidation samples were acidified with 20 μL of 1 mol/L H3PO4. Iodine was reduced by adding 5 μL of fresh ascorbic acid (20 mg/mL).

Pterines were measured by high-performance liquid chromatography (HPLC) on a Waters 600E HPLC (Etten-Leur). The HPLC was equipped with a Pontisil ODS 10 μm column (Alltech Associates Inc). A linear gradient was used for elution of the pterines [80% A [50% MeOH in H2O (v/v)]] and 20% B (H2O) in 10 minutes. After 10 minutes the column was washed with H2O for 20 minutes. Fluorescence detection (360 nm excitation and 435 nm emission) was performed using a Waters 2475 Multi Lambda Fluorescence Detector. BH4 concentration, expressed as picomoles per milligram of protein, was calculated by subtracting BH2+biopterin from total biopterins.

Protein Measurements

To confirm high expression of eNOS and rule out expression of inducible NO synthase (iNOS) or neuronal NOS, bEnd.3 cells, HUVECs, and CDC.HMEC-1 were lysed [20 mmol/L Tris-HCl, 150 mmol/L NaCl, 1 mmol/L EDTA, 1 mmol/L EGTA, 1% Triton-X100, and protease inhibitors (MiniComplete, Roche Diagnostics Corporation; pH 7.4)]. Cell extracts and positive control protein supplied with antibodies were subjected to SDS-PAGE and transferred to polyvinylidene fluoride membrane. Blots were incubated with monoclonal antibodies against eNOS, nNOS (Transduction Laboratories), and iNOS (Alexis) and subsequently with a horseradish peroxidase–conjugated rabbit anti-mouse antibody (Jackson Immunochemicals). Membrane-bound antibodies were visualized using chemiluminescence reagent (Roche).

Expression of eNOS was also determined using an ELISA. A polyclonal rabbit antibody raised against a recombinant fragment spanning residues 1 to 67 of bovine eNOS (unpublished data) was bound to microtiter plates (NUNC-Immuno Plate Maxisorb Surface, NUNC) by incubation at 4°C. Plates were washed 3 times in Tris buffer [50 mmol/L Tris-HCl and 150 mmol/L NaCl (pH7.4)] with 0.1% Tween 20 and then incubated with 3% BSA in Tris buffer with 0.05% Tween 20 for 2 hours at room temperature. Samples were incubated in a total volume of 50 μL for 2 hours at room temperature. Plates were thoroughly washed 3 times and incubated for 2 hours at room temperature with 100 μL of 125 ng/mL eNOS antibody (Transduction Laboratories) in Tris buffer with 0.1% BSA and 0.05% Tween 20. Subsequently, plates were incubated with 650 μg/mL horseradish peroxidase–conjugated rabbit anti-mouse antibody (Jackson Immunochemicals) in Tris buffer with 0.1% BSA and 0.05% Tween 20 for 2 hours at room temperature. A color reaction was observed by incorporating 3,3′,5,5′-tetramethyl-benzidine into the reaction, which was stopped by adding 50 μL of 2 mol/L H2SO4. The optical density was measured at 450 nm in a microplate reader (Multiskan Ascent, Labsystems). The assay was validated using a dilution series of bEnd.3 cells. The interassay variability was 5±2%, whereas the intraassay variability was 2.0±0.8%.

Calculations and Statistical Analysis

NO and ROS production was calculated by determining the slopes of each line with linear regression. Results were expressed relative to control. Statistical comparisons were made by 1-way or 2-way ANOVA, as required. Subsequent post hoc testing was done with the Student–Newman–Keuls test. P values <0.05 were considered statistically significant.

Results

eNOS in bEnd.3 Cells Is, in Part, in an Uncoupled State

To identify the source of NO production in bEnd.3 cells, expression levels of eNOS, nNOS, and iNOS were determined. High expression of eNOS and the absence of iNOS or nNOS protein were confirmed by Western blot (Figure 2A). Nonstimulated bEnd.3 cells produced detectable levels of NO (Figure 2B). Incubation with l-NAME showed that 30 μmol/L was the minimal concentration to inhibit NO production (data not shown). Similar results were obtained with NG-methyl-l-arginine acetate (minimal concentration, 100 μmol/L) and Nω-nitro-l-arginine (minimal concentration 30 μmol/L; data not shown).

Figure 2. Effect of VEGF on eNOS-dependent NO production. (A) eNOS, iNOS, and nNOS expression levels in bEnd.3 cells. Lanes 1, 4, 6: bEnd.3 (15 μg protein/lane); lane 2: HUVEC (150 μg protein/lane); lane 3: human aortic endothelial cell (50 μg protein/lane); lane 5: macrophage + IFN-γ/lipopolysaccharide (LPS; 15 μg protein/lane); lane 7: rat pituitary (15 μg protein/lane). (B) bEnd.3 cells loaded with DAF-2DA were stimulated with VEGF (1 to 50 ng/mL) in the presence and absence of l-NAME. Open bars: control; closed bars: 30 μmol/L l-NAME. *P<0.01 vs 0 ng/mL VEGF.

Incubation of bEnd.3 cells with the DCF probe resulted in a fluorescent signal, which was decreased 18% (P<0.05) by the addition of l-NAME. Because CM-H2DCFDA is not reactive toward NO (Figure 1E), these data show that, other than NO production, there is constitutive eNOS-dependent formation of ROS, that is, uncoupling, in the basal state.

VEGF Enhances eNOS-Dependent NO Production but Also ROS Production in bEnd.3 Cells

VEGF induced a concentration-dependent increase in eNOS-dependent NO production (Figure 2B). VEGF also significantly increased eNOS-dependent ROS production (Figure 3). In VEGF-stimulated cells, the addition of l-NAME resulted in a 35% (P<0.05) decrease in the DCF signal versus an 18% (P<0.05) decrease in DCF signal in nonstimulated cells.

Figure 3. Effect of VEGF and l-arginine on eNOS-dependent ROS production. bEnd.3 cells were loaded with CM-H2DCFDA and stimulated with VEGF in the presence and absence of l-NAME. Results plotted as eNOS-dependent ROS production (difference between total ROS production and eNOS-independent ROS production).

eNOS Uncoupling Is Because of Relative Shortage of BH4 but Not Because of Shortage of l-Arginine

Shortage of BH4 can lead to eNOS uncoupling. Determination of biopterin levels in bEnd.3 cells and CDC.HMEC-1 revealed that BH4 levels are &8 times increased in bEnd.3 cells as compared with CDC.HMEC-1 (Figure 4A). The BH4/BH2+biopterin ratio in bEnd.3 cells and HMECs was not significantly different. In addition, eNOS expression in bEnd.3 cells was 250-fold increased in comparison with CDC.HMEC-1 as determined with Western blot (Figure 4B) and ELISA (OD450 bEnd.3 cells, 2.162±0.3; CDC.HMEC-1, 0.007±0.001). The addition of BH4 (1 to 10 μmol/L) to bEnd.3 cells resulted in a significant (P<0.01) increase in NO production at 3 and 10 μmol/L (Figure 5A). A similar result was obtained with VEGF. ROS levels showed a tendency to decrease in the presence of BH4 (1 to 10 μmol/L; Figure 5B). A similar result was found in the presence of VEGF. To exclude any antioxidant effects of BH4, NH4 was used as a negative control. The addition of 10 μmol/L NH4 had no effects on NO production and decreased ROS production in bEnd.3 cells (Figure 6).

Figure 4. Biopterin and eNOS content in bEnd.3 cells and CDC.HMEC-1. (A) BH4 concentration was calculated by subtracting BH2+biopterin from total biopterins. Open bars: bEnd.3 cells; closed bars: CDC.HMEC-1. (B) Representative Western blot and densitometric analysis of the bands (n=6) on eNOS expression in bEnd.3 cells and CDC.HMEC-1. Lanes 1 to 5: bEnd.3 cells (0.25, 0.5, 1, 2.5, and 5 μg protein/lane, respectively); lane 6: CDC.HMEC-1 (5 μg protein/lane).

Figure 5. Effect of BH4 on NO and ROS production. bEnd.3 cells were incubated for 16 hours with BH4. Results were plotted as eNOS-dependent NO or ROS production (as in Figure 3). (A) Effect of BH4 on NO production. All VEGF-stimulated bEnd.3 cells had significantly higher NO production than nonstimulated cells. (B) Effect of BH4 on ROS production. *P<0.05 vs 0 μmol/L BH4; #P<0.05 vs 50 μmol/L l-arginine; $P<0.05 vs 0 ng/mL VEGF.

Figure 6. Effect of NH4 on NO and ROS production. bEnd.3 cells were incubated for 16 hours with 10 μmol/L BH4 or 10 μmol/L NH4. Results were plotted as eNOS-dependent NO or ROS production (as in Figure 3). (A) Effect of BH4 and NH4 on NO production. (B) Effect of BH4 and NH4 on ROS production. *P<0.01 vs 0 ng/mL VEGF.

Because eNOS uncoupling can also be caused by a shortage of the substrate l-arginine, the effect of additional l-arginine on NO and ROS production was determined. In the presence of high l-arginine levels (500 μmol/L), the addition of BH4 did not significantly increase basal NO production and also had no effect on VEGF-induced NO production (Figure 5A). Increasing l-arginine concentrations up to 500 μmol/L did not affect the basal ROS production but did lead to a significant increase in eNOS-dependent ROS production in the presence of VEGF (Figure 3). In the presence of high l-arginine levels, ROS production was significantly decreased after the addition of 3 and 10 μmol/L BH4, both in the absence and presence of VEGF (Figure 5B).

Other Potential ROS Sources

Diphenyleneiodonium (DPI) was used to identify flavin-containing enzymes as possible ROS sources. Incubation of bEnd.3 cells with DPI lead to a significant 20% (P<0.05) decrease in basal ROS production (Figure 7) and a 25% (P<0.05) decrease in the presence of VEGF (data not shown). To determine whether NADPH oxidase, xanthine oxidase, and mitochondria are possible ROS sources, bEnd.3 cells were incubated with apocynin, oxypurinol, and thenoyltrifluoroacetone (TTFA), respective inhibitors of these oxidase systems. Apocynin and TTFA had no effect on the DCF signal (Figure 7). Oxypurinol decreased ROS production slightly in the absence (Figure 7) but not in the presence of VEGF (data not shown). DPI and oxypurinol were also combined with l-NAME, resulting in an additional decrease in ROS production as compared with the effect of the separate inhibitors (Figure 7).

Figure 7. Sources of ROS in bEnd.3 cells. ROS production was measured with the CM-H2DCFDA assay in the presence or absence of 10 μmol/L DPI, 100 μmol/L apocynin (apo), 100 μmol/L oxypurinol (oxy), 1 μmol/L TTFA, or a combination of 30 μmol/L l-NAME and 100 μmol/L oxypurinol or 10 μmol/L DPI. *P<0.01 vs control.

To exclude the involvement of peroxynitrite (ONOO) in eNOS uncoupling, cells were treated with the ONOO scavenger Ebselen. Incubation with 10 μmol/L Ebselen did not significantly affect the DCF signal (control, 1.00±0.02; 10 μmol/L Ebselen, 1.05±0.05; 50 ng/mL VEGF, 1.12±0.06; 50 ng/mL VEGF +10 μmol/L Ebselen, 0.96±0.05).

Discussion

It has been proposed that long-term elevation of eNOS expression may require adjustments in tetrahydrobiopterin metabolism,6 which could explain the controversial results between studies on prolonged12,13 and short-term eNOS overexpression.8–10 The present study demonstrates that bEnd.3 cells chronically express high eNOS levels but do not express the other NOS isoforms. With respect to eNOS, bEnd.3 cells are relatively deficient in BH4 as compared with other endothelial cells. As a consequence, eNOS is partially in the uncoupled state. Stimulation of eNOS activity in these cells by VEGF enhanced ROS production, particularly at high l-arginine concentrations, which was corrected to a substantial degree by BH4. Our data suggest that under conditions of chronically increased eNOS expression and particularly if eNOS activity is stimulated, a relative shortage of BH4 may lead to eNOS uncoupling, resulting in superoxide production. Our results are in line with observations by Ozaki et al,13 who demonstrated that overexpression of eNOS in the endothelium promoted atherosclerosis in apoE-knockout mice. Dysfunctional eNOS and subsequent increased superoxide production seems to be responsible for the progression of atherosclerosis in these animals, mainly caused by a deficiency of the cofactor BH4. In agreement with this study, Cosentino et al12 showed that increased eNOS expression in SHR is also associated with dysfunctional eNOS, which may contribute to the development of hypertension and its vascular complications in these rats.

bEnd.3 cells are particularly useful for studying eNOS uncoupling, because NADPH oxidase, which is a major source of ROS in endothelial cells,18 does not contribute to ROS production in these cells. The combined results of this study show that the ROS production in bEnd.3 cells is eNOS dependent, implying that eNOS in bEnd.3 cells is partly in an uncoupled state.

We hypothesized that activation of the enzyme would enhance ROS production. Indeed, stimulation with VEGF increased eNOS-dependent ROS production. This is in line with studies in other endothelial cells that show an eNOS-dependent increase in ROS production after VEGF stimulation.19,20

We and others found a decrease in eNOS-dependent ROS production after addition of l-NAME, whereas Colavitti et al19 reported the opposite. The latter observation is consistent with the common perspective that inhibition of functional NOS and, thus, blockage of NO production, results in the loss of the NO-scavenging effect and, thus, increased O2·− levels. On the other hand, inhibition of dysfunctional NOS will lead to a decrease in ROS production because of the coupling state of eNOS.21–24

eNOS dysfunction and subsequent O2·− production is commonly thought to result from a shortage of BH4.6 In the present study, the addition of BH4 resulted in a significant increase in NO production in bEnd.3 cells, whereas ROS levels showed a tendency to decrease. NH4 was used as a negative control, because it exerts antioxidant effects but has no influence on eNOS uncoupling. The addition of NH4 had no effect on NO production and decreased ROS production in bEnd.3 cells. From these results, it can be concluded that the decreased ROS production in the presence of BH4 is because of a decrease in eNOS uncoupling and not caused by antioxidant effects of BH4. In line with these results, it has been shown that overexpression of GTP-cyclohydrolase I, the rate-limiting enzyme in BH4 synthesis, reduced endothelial dysfunction in apoE-knockout mice.25 In addition, GTP-cyclohydrolase I–knockout mice, which are deficient in BH4, showed hypertension and increased vascular superoxide production. The latter was inhibited by NG-methyl-l-arginine acetate, indicating that ROS was generated by uncoupled eNOS.21

Other than BH4 deficiency, shortage of the substrate l-arginine could theoretically lead to uncoupling of eNOS.1 However, there is limited evidence on the role of l-arginine in eNOS dysfunction.26,27 Moreover, there is no consensus on whether supplementation of l-arginine is beneficial under conditions of endothelial dysfunction. Some studies in animals and humans show restored endothelial function,28,29 whereas others fail to show a beneficial effect of l-arginine.30–32 Chen et al33 found that chronic treatment with l-arginine negated the positive effect of iNOS deficiency in apoE/iNOS double-knockout mice. Furthermore, diabetic rats showed increased ROS production after the addition of l-arginine, which was partly reduced by the addition of l-NAME and DPI and the addition of BH4.24 Our results indicate that a shortage of l-arginine is not causing eNOS-dependent ROS production in bEnd.3 cells. In fact, increasing the l-arginine concentration enhanced VEGF-induced ROS production. There are 2 possible explanations for these findings. First, increased l-arginine levels have been associated with an increase in total biopterin levels, but unaltered BH4 levels, suggesting that l-arginine supplementation leads to oxidation of biopterin.33 Second, increased O2·− production in the presence of l;arginine might be because of increased calmodulin binding and a resulting increased electron flow through the enzyme,34,35 leading to a relatively augmented shortage of BH4. The mechanism through which increasing levels of l-arginine enhances ROS production by increasing l-arginine levels in bEnd.3 cells is unclear.

Although the addition of BH4 led to a reduction of ROS production in bEnd.3 cells, it did not completely abolish ROS production. These results correspond with the in vitro study with the purified enzyme,7 in which BH4 only partly inhibited uncoupling of the purified eNOS enzyme. We identified xanthine oxidase as an additional source of ROS in bEnd.3 cells under basal conditions. ONOO, NADPH oxidase, and mitochondria were excluded as contributors to ROS production in bEnd.3 cells.

Several reports have been published on intrinsic superoxide production by NO synthases. For nNOS, it has been reported that ROS are produced by the haem group.36 For iNOS and eNOS, however, the source of ROS production is unclear. Some reports suggest that both the haem and the flavin domain in eNOS produce superoxide.27,37 In bEnd.3 cells, the addition of DPI led to a decrease in but not complete inhibition of ROS production, indicating that flavins are not involved in the intrinsic ROS production by eNOS in these cells. These data suggest that the source of the remaining eNOS-dependent ROS production in bEnd.3 cells, even in the presence of additional BH4, is probably the haem group in eNOS.27,7,37

Perspectives

Although eNOS gene transfer has been proposed to be useful in the treatment of endothelial dysfunction,8–10 we now show that sustained high expression of eNOS protein results in ROS production because of uncoupling. eNOS-dependent ROS production in bEnd.3 cells is partly corrected by BH4 supplementation. These findings suggest that attempts to improve endothelial function, by sustained increases of eNOS expression and activity, for instance by gene transfer of eNOS or VEGF, should be accompanied by concomitant increases of BH4 to prevent eNOS uncoupling.

This study was supported by the Dutch Heart Foundation (99.041). Branko Braam is a fellow of the Royal Dutch Academy of Arts and Sciences. Marianne C. Verhaar is supported by the Netherlands Science Organization (NWO; VENI grant 016.036.041). We thank Monique de Sain for the biopterin measurements. The technical support of Petra M. de Bree and Martin W. Roeleveld is highly appreciated.

Footnotes

Correspondence to Jaap A. Joles, Department of Nephrology and Hypertension F03.223, Heidelberglaan 100, 3584 CX Utrecht, the Netherlands. E-mail

References

  • 1 Govers R, Rabelink TJ. Cellular regulation of endothelial nitric oxide synthase. Am J Physiol Renal Physiol. 2001; 280: F193–F206.CrossrefMedlineGoogle Scholar
  • 2 Braam B, de Roos R, Dijk A, Boer P, Post JA, Kemmeren PP, Holstege FC, Bluysen HA, Koomans HA. Nitric oxide donor induces temporal and dose-dependent reduction of gene expression in human endothelial cells. Am J Physiol Heart Circ Physiol. 2004; 287: H1977–H1986.CrossrefMedlineGoogle Scholar
  • 3 Katusic ZS. Superoxide anion and endothelial regulation of arterial tone. Free Radic Biol Med. 1996; 20: 443–448.CrossrefMedlineGoogle Scholar
  • 4 Chen K, Keaney J. Reactive oxygen species-mediated signal transduction in the endothelium. Endothelium. 2004; 11: 109–121.CrossrefMedlineGoogle Scholar
  • 5 Xia Y, Tsai AL, Berka V, Zweier JL. Superoxide generation from endothelial nitric-oxide synthase. A Ca2+/calmodulin-dependent and tetrahydrobiopterin regulatory process. J Biol Chem. 1998; 273: 25804–25808.CrossrefMedlineGoogle Scholar
  • 6 Katusic ZS. Vascular endothelial dysfunction: does tetrahydrobiopterin play a role? Am J Physiol Heart Circ Physiol. 2001; 281: H981–H986.CrossrefMedlineGoogle Scholar
  • 7 Stroes E, Hijmering M, van Zandvoort M, Wever R, Rabelink TJ, van Faassen EE. Origin of superoxide production by endothelial nitric oxide synthase. FEBS Lett. 1998; 438: 161–164.CrossrefMedlineGoogle Scholar
  • 8 Kullo IJ, Mozes G, Schwartz RS, Gloviczki P, Tsutsui M, Katusic ZS, O’Brien T. Enhanced endothelium-dependent relaxations after gene transfer of recombinant endothelial nitric oxide synthase to rabbit carotid arteries. Hypertension. 1997; 30: 314–320.CrossrefMedlineGoogle Scholar
  • 9 Mujynya-Ludunge K, Viswambharan H, Driscoll R, Ming XF, von Segesser LK, Kappenberger L, Yang Z, Vassalli G. Endothelial nitric oxide synthase gene transfer restores endothelium-dependent relaxations and attenuates lesion formation in carotid arteries in apolipoprotein E-deficient mice. Basic Res Cardiol. 2005; 100: 102–111.CrossrefMedlineGoogle Scholar
  • 10 Ooboshi H, Chu Y, Rios CD, Faraci FM, Davidson BL, Heistad DD. Altered vascular function after adenovirus-mediated overexpression of endothelial nitric oxide synthase. Am J Physiol. 1997; 273: H265–H270.CrossrefMedlineGoogle Scholar
  • 11 Vaziri ND, Ni Z, Oveisi F. Upregulation of renal and vascular nitric oxide synthase in young spontaneously hypertensive rats. Hypertension. 1998; 31: 1248–1254.CrossrefMedlineGoogle Scholar
  • 12 Cosentino F, Patton S, d’Uscio LV, Werner ER, Werner-Felmayer G, Moreau P, Malinski T, Luscher TF. Tetrahydrobiopterin alters superoxide and nitric oxide release in prehypertensive rats. J Clin Invest. 1998; 101: 1530–1537.CrossrefMedlineGoogle Scholar
  • 13 Ozaki M, Kawashima S, Yamashita T, Hirase T, Namiki M, Inoue N, Hirata K, Yasui H, Sakurai H, Yoshida Y, Masada M, Yokoyama M. Overexpression of endothelial nitric oxide synthase accelerates atherosclerotic lesion formation in apoE-deficient mice. J Clin Invest. 2002; 110: 331–340.CrossrefMedlineGoogle Scholar
  • 14 Montesano R, Pepper MS, Mohle-Steinlein U, Risau W, Wagner EF, Orci L. Increased proteolytic activity is responsible for the aberrant morphogenetic behavior of endothelial cells expressing the middle T oncogene. Cell. 1990; 62: 435–445.CrossrefMedlineGoogle Scholar
  • 15 Jaffe EA, Nachman RL, Becker CG, Minick CR. Culture of human endothelial cells derived from umbilical veins. Identification by morphologic and immunologic criteria. J Clin Invest. 1973; 52: 2745–2756.CrossrefMedlineGoogle Scholar
  • 16 Ades EW, Candal FJ, Swerlick RA, George VG, Summers S, Bosse DC, Lawley TJ. HMEC-1: establishment of an immortalized human microvascular endothelial cell line. J Invest Dermatol. 1992; 99: 683–690.CrossrefMedlineGoogle Scholar
  • 17 Cai S, Khoo J, Channon KM. Augmented BH4 by gene transfer restores nitric oxide synthase function in hyperglycemic human endothelial cells. Cardiovasc Res. 2005; 65: 823–831.CrossrefMedlineGoogle Scholar
  • 18 Ushio-Fukai M, Alexander RW. Reactive oxygen species as mediators of angiogenesis signaling: role of NAD(P)H oxidase. Mol Cell Biochem. 2004; 264: 85–97.CrossrefMedlineGoogle Scholar
  • 19 Colavitti R, Pani G, Bedogni B, Anzevino R, Borrello S, Waltenberger J, Galeotti T. Reactive oxygen species as downstream mediators of angiogenic signaling by vascular endothelial growth factor receptor-2/KDR. J Biol Chem. 2002; 277: 3101–3108.CrossrefMedlineGoogle Scholar
  • 20 Marumo T, Noll T, Schini-Kerth VB, Harley EA, Duhault J, Piper HM, Busse R. Significance of nitric oxide and peroxynitrite in permeability changes of the retinal microvascular endothelial cell monolayer induced by vascular endothelial growth factor. J Vasc Res. 1999; 36: 510–515.CrossrefMedlineGoogle Scholar
  • 21 Cosentino F, Barker JE, Brand MP, Heales SJ, Werner ER, Tippins JR, West N, Channon KM, Volpe M, Luscher TF. Reactive oxygen species mediate endothelium-dependent relaxations in tetrahydrobiopterin-deficient mice. Arterioscler Thromb Vasc Biol. 2001; 21: 496–502.CrossrefMedlineGoogle Scholar
  • 22 Guzik TJ, Mussa S, Gastaldi D, Sadowski J, Ratnatunga C, Pillai R, Channon KM. Mechanisms of increased vascular superoxide production in human diabetes mellitus: role of NAD(P)H oxidase and endothelial nitric oxide synthase. Circulation. 2002; 105: 1656–1662.LinkGoogle Scholar
  • 23 Hink U, Li H, Mollnau H, Oelze M, Matheis E, Hartmann M, Skatchkov M, Thaiss F, Stahl RAK, Warnholtz A, Meinertz T, Griendling K, Harrison DG, Forstermann U, Munzel T. Mechanisms underlying endothelial dysfunction in diabetes mellitus. Circ Res. 2001; 88: 14e–22.CrossrefMedlineGoogle Scholar
  • 24 Satoh M, Fujimoto S, Haruna Y, Arakawa S, Horike H, Komai N, Sasaki T, Tsujioka K, Makino H, Kashihara N. NAD(P)H oxidase and uncoupled nitric oxide synthase are major sources of glomerular superoxide in rats with experimental diabetic nephropathy. Am J Physiol Renal Physiol. 2005; 288: F1144–F1152.CrossrefMedlineGoogle Scholar
  • 25 Alp NJ, McAteer MA, Khoo J, Choudhury RP, Channon KM. Increased endothelial tetrahydrobiopterin synthesis by targeted transgenic GTP-cyclohydrolase I overexpression reduces endothelial dysfunction and atherosclerosis in ApoE-knockout mice. Arterioscler Thromb Vasc Biol. 2004; 24: 445–450.LinkGoogle Scholar
  • 26 Mayer B, John M, Heinzel B, Werner ER, Wachter H, Schultz G, Bohme E. Brain nitric oxide synthase is a biopterin- and flavin-containing multi-functional oxido-reductase. FEBS Lett. 1991; 288: 187–191.CrossrefMedlineGoogle Scholar
  • 27 Berka V, Yeh HC, Gao D, Kiran F, Tsai AL. Redox function of tetrahydrobiopterin and effect of L-arginine on oxygen binding in endothelial nitric oxide synthase. Biochemistry. 2004; 43: 13137–13148.CrossrefMedlineGoogle Scholar
  • 28 Boger RH, Bode-Boger SM, Mugge A, Kienke S, Brandes R, Dwenger A, Frolich JC. Supplementation of hypercholesterolaemic rabbits with L-arginine reduces the vascular release of superoxide anions and restores NO production. Atherosclerosis. 1995; 117: 273–284.CrossrefMedlineGoogle Scholar
  • 29 Creager MA, Gallagher SJ, Girerd XJ, Coleman SM, Dzau VJ, Cooke JP. L-arginine improves endothelium-dependent vasodilation in hypercholesterolemic humans. J Clin Invest. 1992; 90: 1248–1253.CrossrefMedlineGoogle Scholar
  • 30 Cross JM, Donald AE, Kharbanda R, Deanfield JE, Woolfson RG, MacAllister RJ. Acute administration of L-arginine does not improve arterial endothelial function in chronic renal failure. Kidney Int. 2001; 60: 2318–2323.CrossrefMedlineGoogle Scholar
  • 31 Jeremy RW, McCarron H, Sullivan D. Effects of dietary L-arginine on atherosclerosis and endothelium-dependent vasodilatation in the hypercholesterolemic rabbit. Response according to treatment duration, anatomic site, and sex. Circulation. 1996; 94: 498–506.CrossrefMedlineGoogle Scholar
  • 32 Oomen CM, van Erk MJ, Feskens EJ, Kok FJ, Kromhout D. Arginine intake and risk of coronary heart disease mortality in elderly men. Arterioscler Thromb Vasc Biol. 2000; 20: 2134–2139.CrossrefMedlineGoogle Scholar
  • 33 Chen J, Kuhlencordt P, Urano F, Ichinose H, Astern J, Huang PL. Effects of chronic treatment with L-arginine on atherosclerosis in apoE knockout and apoE/inducible NO synthase double-knockout mice. Arterioscler Thromb Vasc Biol. 2003; 23: 97–103.LinkGoogle Scholar
  • 34 Abu-Soud HM, Feldman PL, Clark P, Stuehr DJ. Electron transfer in the nitric-oxide synthases. Characterization of L-arginine analogs that block heme iron reduction. J Biol Chem. 1994; 269: 32318–32326.CrossrefMedlineGoogle Scholar
  • 35 Abu-Soud HM, Ichimori K, Presta A, Stuehr DJ. Electron transfer, oxygen binding, and nitric oxide feedback inhibition in endothelial nitric-oxide synthase. J Biol Chem. 2000; 275: 17349–17357.CrossrefMedlineGoogle Scholar
  • 36 Porasuphatana S, Tsai P, Rosen GM. The generation of free radicals by nitric oxide synthase. Comp Biochem Physiol C Toxicol Pharmacol. 2003; 134: 281–289.CrossrefMedlineGoogle Scholar
  • 37 Vasquez-Vivar J, Kalyanaraman B, Martasek P, Hogg N, Masters BS, Karoui H, Tordo P, Pritchard KA. Superoxide generation by endothelial nitric oxide synthase: the influence of cofactors. Proc Natl Acad Sci U S A. 1998; 95: 9220–9225.CrossrefMedlineGoogle Scholar

eLetters(0)

eLetters should relate to an article recently published in the journal and are not a forum for providing unpublished data. Comments are reviewed for appropriate use of tone and language. Comments are not peer-reviewed. Acceptable comments are posted to the journal website only. Comments are not published in an issue and are not indexed in PubMed. Comments should be no longer than 500 words and will only be posted online. References are limited to 10. Authors of the article cited in the comment will be invited to reply, as appropriate.

Comments and feedback on AHA/ASA Scientific Statements and Guidelines should be directed to the AHA/ASA Manuscript Oversight Committee via its Correspondence page.