Europe PMC

This website requires cookies, and the limited processing of your personal data in order to function. By using the site you are agreeing to this as outlined in our privacy notice and cookie policy.

Abstract 


The integrity of microvascular endothelium is an important regulator of myocardial contractility. Microvascular barrier integrity could be altered by increased reactive oxygen species (ROS) stress seen within minutes after cardiac arrest resuscitation. Akt and its downstream target nitric oxide (NO) synthase (NOS)3 can protect barrier integrity during ROS stress, but little work has studied these oxidant stress responses in human cardiac microvascular endothelial cells (HCMVEC). We, therefore, studied how ROS affects barrier function and NO generation via Akt and its downstream target NOS3 in HCMVEC. HCMVEC exposed to 500 microM H2O2 had increased Akt phosphorylation within 10 min at both Ser-473 and Thr-308 sites, an effect blocked by the phosphatidylinositol 3-kinase inhibitor LY-294002. H2O2 also induced NO generation that was associated with NOS3 Ser-1177 site phosphorylation and Thr-495 dephosphorylation, with Ser-1177 effects attenuated by LY-294002 and an Akt inhibitor, Akt/PKB signaling inhibitor-2 (API-2). H2O2 induced significant barrier disruption in HCMVEC within minutes, but recovery started within 30 min and normalized over hours. The NOS inhibitor Nomega-nitro-L-arginine methyl ester (200 microM) blocked NO generation but had no effect on H2O2-induced barrier permeability or the recovery of barrier integrity. By contrast, the Akt inhibitor API-2 abrogated HCMVEC barrier restoration. These results suggest that oxidant stress in HCMVEC activates NOS3 via Akt. NOS3/NO are not involved in the regulation of H2O2-affected barrier function in HCMVEC. Independent of NOS3 regulation, Akt proves to be critical for the restoration of barrier integrity in HCMVEC.

Free full text 


Logo of ajpheartLink to Publisher's site
Am J Physiol Heart Circ Physiol. 2008 Dec; 295(6): H2417–H2426.
Published online 2008 Oct 17. https://doi.org/10.1152/ajpheart.00501.2008
PMCID: PMC2614535
PMID: 18931031

Akt activates NOS3 and separately restores barrier integrity in H2O2-stressed human cardiac microvascular endothelium

Abstract

The integrity of microvascular endothelium is an important regulator of myocardial contractility. Microvascular barrier integrity could be altered by increased reactive oxygen species (ROS) stress seen within minutes after cardiac arrest resuscitation. Akt and its downstream target nitric oxide (NO) synthase (NOS)3 can protect barrier integrity during ROS stress, but little work has studied these oxidant stress responses in human cardiac microvascular endothelial cells (HCMVEC). We, therefore, studied how ROS affects barrier function and NO generation via Akt and its downstream target NOS3 in HCMVEC. HCMVEC exposed to 500 μM H2O2 had increased Akt phosphorylation within 10 min at both Ser-473 and Thr-308 sites, an effect blocked by the phosphatidylinositol 3-kinase inhibitor LY-294002. H2O2 also induced NO generation that was associated with NOS3 Ser-1177 site phosphorylation and Thr-495 dephosphorylation, with Ser-1177 effects attenuated by LY-294002 and an Akt inhibitor, Akt/PKB signaling inhibitor-2 (API-2). H2O2 induced significant barrier disruption in HCMVEC within minutes, but recovery started within 30 min and normalized over hours. The NOS inhibitor Nω-nitro-l-arginine methyl ester (200 μM) blocked NO generation but had no effect on H2O2-induced barrier permeability or the recovery of barrier integrity. By contrast, the Akt inhibitor API-2 abrogated HCMVEC barrier restoration. These results suggest that oxidant stress in HCMVEC activates NOS3 via Akt. NOS3/NO are not involved in the regulation of H2O2-affected barrier function in HCMVEC. Independent of NOS3 regulation, Akt proves to be critical for the restoration of barrier integrity in HCMVEC.

Keywords: oxidative stress, nitric oxide synthase 3, Akt/protein kinase B, endothelial cell barrier function, hydrogen peroxide

microcirculatory dysfunction is an important etiological component of ischemia-reperfusion injury. A significant percentage of victims of cardiac arrest, despite being initially resuscitated, die of heart-related causes within hours due to recurrent arrhythmias and cardiovascular collapse (4, 23, 28, 47). Recent work suggests that these cardiac arrest events are temporally associated with interruptions in microvascular blood flow to the heart that begin within minutes of cardiopulmonary resuscitation (CPR) and may affect the return of spontaneous circulation (ROSC) (17). Furthermore, serious microvascular dysfunction in critical organs such as the heart and brain is seen within hours of ROSC and is associated with critical organ dysfunction (1, 8).

The mechanism of microvascular endothelial dysfunction in the post-cardiac arrest heart is not well known. Our studies in a swine model of cardiac arrest suggest that oxidant stress increases significantly within minutes of CPR and peaks 5–10 min after ROSC (20). Oxidant stress caused by a surge in reactive oxygen species (ROS) generation during reoxygenation (54) can disrupt microvascular integrity and interrupt blood flow (25, 38, 56). Although responses to ROS in several endothelial cell types have been reported, how human cardiac microvascular endothelial cells (HCMVEC) respond to ROS has not been well characterized. Other endothelial cell types lose monolayer integrity when exposed to hydrogen peroxide (H2O2) with variable ability to recover over time (9, 24, 25, 31, 36, 44, 51).

HCMVEC response to ROS could impact heart function via changes in endothelial barrier function that then disrupt tissue blood flow. In addition, these cells serve as an important source of nitric oxide (NO), which regulates blood flow (22, 27, 40). This NO could also affect surrounding cardiomyocyte function. Our work in heart cell models of ischemia-reperfusion injury suggests that NO generation, possibly via survival kinase signaling, can play a highly cardioprotective role (21, 29, 45). In endothelial cells, the survival kinase Akt has been recognized for some time as an upstream regulator of such cardioprotective NO generation (18), but only recently has it been implicated as a positive regulator of endothelial chemotaxis and barrier integrity (31).

Given that ROS can activate Akt (11, 39), and Akt is upstream of both cardioprotective NO signaling and tight junction regulation, we tested the ability of H2O2 to affect HCMVEC monolayer integrity via Akt and NO synthase (NOS)3 signaling. Here we show that H2O2-induced NOS3 activation is, in part, phosphatidylinositol 3-kinase (PI3K)-Akt dependent. We also show that Akt is critical for the restoration of H2O2-impaired HCMVEC monolayer integrity apart from NO signaling. Our findings support the notion that Akt-dependent signaling is pivotal for the functional protection of the heart during conditions of increased oxidant stress.

MATERIALS AND METHODS

Materials.

Cell culture reagents including complete microvascular endothelial cell growth medium-2 (EGM-2MV) and phenol red-free endothelial basal medium-2 (EBM-2) were purchased from Lonza (Walkersville, MD). The selective Akt inhibitor Akt/PKB signaling inhibitor-2 (API-2) was obtained from EMD Chemicals (Gibbstown, NJ). Propidium iodide (PI), Nω-nitro-l-arginine methyl ester (l-NAME), H2O2, LY-294002, and β-actin antibody were obtained from Sigma-Aldrich (St. Louis, MO). 4,5-Diaminofluorescein diacetate (DAF-2 DA) was from EMD Biosciences (San Diego, CA). The monoclonal antibody for endothelial NOS (eNOS; NOS3) was purchased from BD Transduction Laboratories (BD Biosciences, Franklin Lakes, NJ). Vascular endothelial (VE)-cadherin polyclonal antibodies were purchased from Cayman Chemical (Ann Arbor, MI), and zona occludens-1 (ZO-1) polyclonal antibodies were purchased from Invitrogen (Carlsbad, CA). All other antibodies and Akt kinase assay kit were from Cell Signaling Technology (Beverly, MA). The ApoAlert caspase-3 assay plate was obtained from Clontech (Mountain View, CA).

Cell culture.

HCMVEC were purchased from Lonza and cultured in complete EGM-2MV medium containing 5% FBS. Passages between 5 and 8 were used for all experiments. Cells were grown at 37°C in a humidified atmosphere containing 5% CO2. For all signaling and transendothelial electrical resistance measurement experiments, cells were starved for 1 h in serum-reduced EBM-2 medium containing either 0.02% FBS or 0.1% BSA and then treated in the same medium.

Perfusion system and media composition.

HCMVEC grown on 25-mm glass coverslips were placed inside a Sykes-Moore chamber (1.2 ml; Bellco Glass, Vineland, NJ) as described previously (53). The chamber and inflow tubing were maintained at 37°C. Phenol red-free EBM-2 medium with 0.02% FBS was prebubbled with 5% CO2, 21% O2, and 74% N2 gas for 30 min before each experiment. The medium was continuously channeled into the chamber at a flow rate of 0.25 ml/min. Oxygen tension (PO2), pH, and the temperature of the perfusate were controlled as previously described (23, 25). Tubing supplying medium to the chamber was made of stainless steel and PharMed polymer (Cole-Parmer Instrument, Chicago, IL).

Video/fluorescent microscopy.

A Nikon TE 2000-U inverted phase/epifluorescent microscope was used for cell imaging. Phase contrast Hoffman modulation optics and a charge-coupled device (CCD) camera were used to monitor morphologic changes over time in the same field of cells (~70 × 90 μm). Fluorescent images were acquired from a cooled Hamamatsu slow-scanning PC-controlled camera (Coolsnap; Photometrics), and changes in fluorescent intensity were quantified with MetaMorph software (Universal Imaging, Downington, PA).

Cell viability.

Cell viability was assessed with the fluorescent exclusion dye PI (5 μM) with excitation of 540 nm and emission of 590 nm. PI fluorescence was used to quantify cell death throughout the entire experiment in a selected field of endothelial cells. At the end of the experiment, all cells were permeabilized with digitonin (100 μM). Cell death was expressed as the PI fluorescence at any given time point relative to the maximal fluorescence observed after cell permeabilization with digitonin.

Measurement of intracellular NO production.

NO generation by HCMVEC was evaluated in the perfusion system by monitoring fluorescence of the NO-selective probe DAF-2 DA (1 μM) following cell treatment with 500 μM H2O2 for 10 min. Images were taken at 30 min following H2O2 washout with excitation of 480 nm and emission of 520 nm. Fluorescence intensities of cells were measured using MetaMorph software and expressed as arbitrary units.

Measurement of HCMVEC transendothelial electrical resistance.

HCMVEC were grown to confluence in polycarbonate wells containing gold microelectrodes, and transendothelial cell electrical resistance (TEER) measurements were performed using an electrical cell substrate impedance sensing system (Applied Biophysics, Troy, NY) as described previously (27). These measurements provide a highly sensitive biophysical assay that indicates the state of cell shape and focal adhesion (19). TEER values from each microelectrode were pooled at discrete time points and plotted versus time as means ± SE. Each experiment was initiated with HCMVEC monolayer resistance of no less than 1,200 ohm. H2O2 (100, 250, or 500 μM, as indicated) was present all course during these experiments.

Caspase activity assay.

HCMVEC treated with or without H2O2 were harvested at selected times, washed twice with cold phosphate-buffered saline (PBS), lysed, and stored at −80°C until analysis. Cells stimulated with 10 μM staurosporine for 3 h were used as a positive control. Caspase-3 activity was measured by the ApoAlert caspase profiling assay, as described previously (41).

Western blotting.

After H2O2 or control treatments, HCMVEC were washed twice with cold PBS and incubated in lysis buffer (Cell Signaling, Danvers, MA) on ice for 5 min followed by brief sonication. Protein concentrations were determined with a Bradford protein assay. Proteins (50 μg) were separated on 7.5% or 10% gels by SDS-PAGE, transferred to a nitrocellulose membrane, and subjected to immunoblotting with appropriate primary antibody (1:1,000 dilution) overnight at 4°C. After washing and incubation with appropriate secondary antibody, bands were detected with enhanced chemiluminescence kit (Amersham Pharmacia). Densitometric analysis of Western blots was carried out using Quantity One Software (Bio-Rad, Richmond, CA).

Akt activity assay.

The activity of Akt was assessed using a nonradioactive Akt kinase assay kit (Cell Signaling), according to the manufacturer's instructions. Briefly, immunoprecipitation of endogenous Akt was performed overnight on 250 μg of endothelial cell extract with immobilized anti-Akt antibody at 4°C. Following precipitation, the immunoprecipitate was centrifuged, washed two times with lysis and kinase buffers, and then incubated with 1 μg of GSK fusion protein and ATP for 30 min at 30°C in a final volume of 50 μl. The reaction was stopped by adding loading buffer, and the proteins were separated on 10% gels by SDS-PAGE, transferred to nitrocellulose membrane, and probed with anti-phospho-GSK3-α/-β Ser-21/9 antibody. Western blots were developed with the enhanced chemiluminescence kit (Amersham Pharmacia).

Statistical analysis.

Treatment and control groups were used in sets containing cells cultured on the same day to eliminate variability due to cell batch. Each experiment was performed at least three times. Data are presented as means ± SE, and two-tailed unpaired t-tests were performed as post hoc tests of significance, with P < 0.05 considered to be significant.

RESULTS

High concentrations of H2O2 are required to activate Akt in HCMVEC.

H2O2 is known to activate NOS3 in multiple endothelial cell types via Akt signaling pathways (34, 48, 49). To test whether H2O2 can activate Akt in HCMVEC, we used increasing concentrations of H2O2 in an attempt to induce the phosphorylation of Akt at major regulatory sites including Ser-473 and Thr-308 (10, 12, 18). We found that high micromolar concentrations of H2O2 were needed to stimulate Akt phosphorylation at both sites. A detectable increase in phosphorylation occurred after 250 μM H2O2 with substantially more phosphorylation seen after exposure to a 500 μM H2O2 concentration for 10 min (Fig. 1, A and B). Such levels of H2O2 are reasonable for modeling HCMVEC exposure to oxidant stress in the heart. For example, concentrations of 80–480 μM of H2O2 can be electrochemically detected from stimulated leukocytes at normal plasma concentrations (32).

An external file that holds a picture, illustration, etc.
Object name is zh40120886190001.jpg

H2O2 stimulates Akt phosphorylation and activity in human cardiac microvascular endothelial cells (HCMVEC). HCMVEC were treated with increasing concentrations of H2O2 for 10 min, and phosphorylation status of Akt at Ser-473 (A) and Thr-308 (B) were determined by Western blot analysis. C and D: to further determine the time course of Akt activation, HCMVEC were stimulated with 500 μM H2O2 for 1.5, 10, and 30 min and Akt Ser-473 and Thr-308 phosphorylation was determined by Western blotting. In some cells, H2O2-containing medium was replaced with a fresh medium 10 min after stimulation. E: HCMVEC were treated with 500 μM H2O2 and Akt-related phosphorylation of GSK3 was measured. Blots are representative of 3 independent experiments. Densitometric analyses represent means ± SE of 3 independent experiments. *P < 0.05; **P < 0.01; ***P < 0.001. au, Arbitrary units; p, phospho.

We further studied the time dependency of Akt Ser-473 and Thr-308 phosphorylation with 500 μM H2O2. Akt phosphorylation occurred at both sites by 1.5 min, reached a maximum at 10 min, and sustained this level of phosphorylation throughout 30 min of H2O2 stimulation (Fig. 1, C and D). However, Akt activity, as measured by phosphorylation of GSK fusion protein, peaked at 10 min and was only half of the maximum by 30 min of stimulation (Fig. 1E). Given that significant activation occurred by 10 min H2O2 exposure, we also tested whether the removal of H2O2 for 20 min (versus prolonged stimulation for 30 min) had different effects. Despite the removal of H2O2 for 20 min after the initial 10-min stimulation, a significant level of Akt phosphorylation and Akt activity at 30 min was maintained. This level of activation was not significantly different from the level observed with continuous 30-min H2O2 exposure (Fig. 1, CE).

High concentrations of H2O2 do not induce HCMVEC caspase-3 activation or cell death.

To evaluate whether 500 μM H2O2 induced cell death in HCMVEC, we measured caspase-3 activation and cell viability following 30 min H2O2 exposure. We found that 500 μM H2O2 does not increase but rather decreases the background level of caspase-3 activity in HCMVEC (Fig. 2A). Cell death, as determined by PI exclusion, did not exceed 5% during 30 min of exposure to 500 μM H2O2 or during the subsequent 60 min in replacement medium (Fig. 2B). Taken together, these data indicate that the concentrations of H2O2 used did not increase cell death in HCMVEC.

An external file that holds a picture, illustration, etc.
Object name is zh40120886190002.jpg

H2O2 does not induce cell death in HCMVEC. A: HCMVEC were treated with 500 μM H2O2 for 30 min and then with basal medium for 2.5 h or with 10 μM staurosporine for 3 h, and caspase-3 activity was assessed in cell lysates using caspase-3-selective fluorigenic substrate (means ± SE; n = 3). B: HCMVEC were treated with 500 μM H2O2, and cell death was monitored by propidium iodide (PI) exclusion. Data are presented as means ± SE of 6 independent experiments. *P < 0.05; ***P < 0.001.

H2O2-induced activation of NOS3 via Ser-1177 phosphorylation and Thr-495 dephosphorylation.

Maximal activation of eNOS (NOS3) requires phosphorylation at Ser-1177, which is downstream of Akt signaling, and concomitant dephosphorylation of Thr-495 via an Akt-independent process (34, 48, 49). As shown in Fig. 3, A and B, H2O2 induces maximal NOS3 phosphorylation in HCMVEC at Ser-1177 and dephosphorylation at Thr-495 sites by 1.5 min after treatment. The phosphorylation-to-dephosphorylation ratio between these two sites peaked at 1.5 min relative to the ratio seen at 10 min after H2O2 cell stimulation (Fig. 3C). We also measured H2O2-induced NO production using the NO-specific fluorescent probe DAF-2 DA. As shown in Fig. 4, NO generation was substantially upregulated by H2O2 and blocked by the NOS inhibitor l-NAME. The selective Akt inhibitor API-2 also attenuated H2O2-induced NO production. These results implicated Akt as a regulator of NOS3 in ROS-stressed HCMVEC. To further confirm the involvement of the PI3K-Akt pathway in the regulation of NOS3, we tested the effect of the PI3K inhibitor LY-294002 and the Akt inhibitor API-2 on H2O2-induced Akt phosphorylation. Both inhibitors blocked Akt phosphorylation at its Ser-473 and Thr-308 sites (Fig. 5, A and B). In addition, both PI3K and Akt inhibitors blocked H2O2-induced NOS3 phosphorylation at Ser-1177 (Fig. 5C), the site regulated by Akt (13, 33). PI3K and Akt inhibitors, however, did not significantly affect the Thr-495 dephosphorylation of NOS3 caused by H2O2 (Fig. 5D). This confirms that, as in other endothelial cell types, the PI3K-Akt pathway is not involved in the regulation of the NOS3 Thr-495 site. This site has been found to be phosphorylated by protein kinase C and dephosphorylated by protein phosphatase 1 and 2A and calcineurin (5, 15, 16).

An external file that holds a picture, illustration, etc.
Object name is zh40120886190003.jpg

H2O2 affects phosphorylation and activation of nitric oxide (NO) synthase (NOS)3 in HCMVEC. HCMVEC were treated with 500 μM H2O2 for the indicated time period with or without intermittent medium replacement 10 min after stimulation, and the status of NOS3 Ser-1177 (A) and Thr-495 (B) phosphorylation was determined by Western blotting. C: experimental data from A and B are presented as the ratio between NOS3 p-Ser-1177 and p-Thr-495. Blots represent 1 out of 3 separate experiments. Results are means ± SE from 3 independent experiments. *P < 0.05; **P < 0.01; ***P < 0.001 vs. untreated control cells. eNOS, Endothelial NOS.

An external file that holds a picture, illustration, etc.
Object name is zh40120886190004.jpg

Effect of NOS and Akt inhibitors on H2O2-induced NO generation. HCMVEC on coverslips were pretreated with the NOS inhibitor Nω-nitro-l-arginine methyl ester (l-NAME; 200 μM) or the Akt inhibitor Akt/PKB signaling inhibitor-2 (API-2; 10 μM) for 2 h and then treated with 500 μM H2O2 for 10 min. Cells were washed with endothelial basal medium-2 (EBM-2), and coverslips were mounted in the perfusion chamber and perfused with EBM-2 containing 1 μM 4,5-diaminofluorescein diacetate (DAF-2 DA). DAF-2 fluorescence was recorded at 30 min with excitation of 480 nm and emission of 520 nm. Fluorescence intensity was quantified using MetaMorph software.

An external file that holds a picture, illustration, etc.
Object name is zh40120886190005.jpg

Effect of phosphatidylinositol 3-kinase and Akt inhibitors on Akt and NOS3 phosphorylation. HCMVEC were pretreated with 10 μM API-2 or 10 μM LY-294002 for 2 h before exposure to 500 μM H2O2 for 10 min. Lysates were probed for Akt Ser-473 (A) and Thr-308 (B) phosphorylation. Membranes were also probed with anti-Akt antibody to determine total Akt. The same lysates were also analyzed for eNOS phosphorylation at Ser-1177 (C) and Thr-495 (D) and total eNOS. Densitometric analyses represent means ± SE of 3 independent experiments. NS, not significant.

Akt is critical for restoration of H2O2-induced barrier permeability.

Oxidative stress in other endothelial cell types results in the loss of tight junction function and increased barrier permeability (24–26, 42, 50, 51). Rat lung microvascular endothelial cells quickly restore monolayer integrity within 10–15 min of treatment with a low concentration of H2O2 (100 μM) (42), whereas bovine pulmonary artery endothelial cells fail to do so even several hours after initial H2O2 exposure (50). We tested whether H2O2 exposure causes a similar loss of monolayer integrity in HCMVEC. As measured by TEER in HCMVEC, we found that H2O2 quickly disrupted monolayer integrity in a dose-response fashion from 100 to 500 μM concentrations (Fig. 6A). HCMVEC began to restore TEER within 30–60 min, reaching baseline TEER levels within hours, despite continuous exposure to H2O2 concentrations of up to 500 μM. Consistent with this, we found that H2O2 exposure was associated with loss of both VE-cadherin-mediated adherence and ZO-1-mediated tight junctions (Fig. 7). Complementary to the TEER data, both VE-cadherin- and ZO-1-mediated junctions were restored in HCMVEC 3 h after stimulation with 250 μM H2O2 (Fig. 7). Given that Akt and NOS3 are activated within 1.5–30 min after H2O2 exposure, we further tested whether they regulate these changes in HCMVEC barrier integrity. As seen in Fig. 6B, the Akt inhibitor API-2 (10 μM), effective in blocking Akt and NOS activation during ROS exposure, blocked the restoration of endothelial cell TEER. By itself, API-2 had no effect during baseline conditions on the integrity of cell-cell interactions. On the contrary, NOS inhibition with l-NAME at a concentration previously shown to inhibit NO generation (200 μM) had no effect on either baseline TEER, early loss of monolayer integrity, or its subsequent recovery after H2O2 exposure (Fig. 6C). These data suggest that Akt plays an important role in restoring HCMVEC barrier integrity after oxidant stress, apart from its downstream NO signaling.

An external file that holds a picture, illustration, etc.
Object name is zh40120886190006.jpg

Akt but not NOS3 is critical for restoration of H2O2-perturbed HCMVEC monolayer integrity. HCMVEC were grown on gold electrical cell substrate impedance sensing electrodes, and transendothelial cell electrical resistance (TEER) was recorded as described in materials and methods. A: dose dependency of H2O2-induced permeability. B: Akt is critical for restoration of H2O2-elicited permeability. HCMVEC were pretreated with 10 μM Akt inhibitor API-2 for 2 h and then stimulated with 250 μM H2O2. Note that API-2 alone has no effect on HCMVEC TEER measures during baseline conditions. C: NO synthases downstream of Akt do not mediate the H2O2-induced permeability dynamic in HCMVEC. HCMVEC were pretreated with 200 μM l-NAME for 2 h and then stimulated with 250 or 500 μM H2O2. Note that l-NAME alone has no effect of HCMVEC TEER dynamic. Data represent means ± SE of 4 independent experiments.

An external file that holds a picture, illustration, etc.
Object name is zh40120886190007.jpg

Dynamic changes in the adherence and tight junctions in HCMVEC upon treatment with H2O2. HCMVEC grown on coverslips were treated with 250 μM H2O2 for 20 min or 3 h, and adherence junction [vascular endothelial (VE)-cadherin] and tight junction [zona occludens-1 (ZO-1)] proteins were identified by immunostaining. Note that both VE-cadherin and ZO-1 peripheral continuous localization is disrupted by H2O2 treatment (residual contacts between cells 20 min after the treatment are marked by arrows) and then restored back to near normal appearance. Scale bar, 5 μm.

DISCUSSION

Ischemia-reperfusion of cardiac tissue in vivo and cardiomyocytes in vitro results in a massive generation of different forms of ROS including H2O2 (2, 52, 54). This heart cell ROS peaks within 10 min of reperfusion (52). Similarly, even minutes of hypoxia, as occurs during cardiac arrest, can initiate cardiomyocyte ROS generation and release by mitochondria that initiate important stress responses (30, 53). In addition to exposure to neighboring heart cell ROS, endothelial cells are exposed to oxidants from the vascular space. Leukocyte activation occurs early during cardiac arrest and can generate H2O2 in concentrations reaching 500 μM (6, 32). Consistent with this, we have reported in a swine model of cardiac arrest that serum and tissue isoprostane levels increase almost threefold within 10 min after ROSC (20). Such rapid ROS generation during minutes of reoxygenation could disrupt microvascular integrity and interrupt blood flow (25, 38, 56) and is likely related to the microvascular dysfunction that appears within the heart minutes after resuscitation (17). In the present study, the substantial loss of HCMVEC barrier integrity after oxidant stress is also consistent with the microvascular dysfunction seen in critical organs such as the heart and brain within hours of resuscitation (1, 8, 55).

This study was conducted to better understand important oxidant stress responses within a newly available endothelial cell type: the HCMVEC. Different endothelial cell types appear to respond uniquely to ROS, and understanding how the heart responds to the oxidant stress of cardiac arrest is important if we are to develop better treatments for a disease with 95% mortality (35, 47). Despite initially successful resuscitation in many cardiac arrest patients, a significant number die within minutes to hours from heart-related complications, including severe cardiovascular dysfunction and hemodynamic collapse (28, 43). In addition, there are no treatments currently given during CPR designed to specifically modulate oxidant stress or restore microvascular function. This study builds on prior work by focusing on heart-specific microvascular endothelial cells. Our finding that HCMVEC restore barrier function within hours of significant oxidant stress stands in contrast with other endothelial cell types, which are unable to restore barrier integrity after similar intensity of oxidant stress and within the same time frame (50, 51). In the setting of cardiac arrest and increased cardiovascular oxidant stress during CPR and after ROSC, the timing and extent of HCMVEC barrier function failure could impact the early cardiovascular collapse seen in many post-cardiac arrest patients (47). The current work suggests that Akt may be an important modulator of barrier integrity restoration in the heart during conditions of oxidant stress.

ROS and endothelial cell barrier integrity.

The effect of H2O2 on vascular permeability and signaling in endothelial cells of different origin in vitro has been studied, but HCMVEC have only recently become available. In other cell types it is known that H2O2 can cause endothelial cell monolayer permeability due to the disruption of cell tight junctions (9, 31, 44) with ERK (14, 24) and p38 (25, 36, 51) MAP kinases mediating these effects of H2O2. The current data suggest that HCMVEC adapt after oxidant stress and restore TEER. In our experiments, HCMVEC could restore TEER within 30 min after treatment with 100 μM H2O2 and within 3 h after stimulation with 500 μM H2O2. Importantly, HCMVEC restored both adherence (VE-cadherin based) and tight (ZO-1 based) junctions, confirming a strong adaptive potential of these cells (Fig. 7). Similar to HCMVEC, rat lung microvascular endothelial cells could restore TEER within 30 min after stimulation with 100 μM H2O2 (26). In contrast, bovine pulmonary artery endothelial cells (50) and bovine lung microvascular cells (51) do not restore monolayer integrity for several hours after exposure to as low as 100 μM H2O2. Collectively, this indicates relatively high adaptive potential of HCMVEC during exposure to relatively high oxidant stress.

Role of NO in endothelial cell integrity.

H2O2 has been shown to activate NOS3 and NO production in the endothelial cell, and NO appears to play some role in both permeability and restoration of barrier integrity in some cell types (34, 48, 49). A direct role for NOS3 in preserving the normal structure of endothelial cell junctions in multiple vascular beds, including cardiac vasculature, was shown using electron microscopy in eNOS−/− mice (40). Conversely, NO has been demonstrated to be involved in ischemia-reperfusion, platelet-activating factor, histamine, and VEGF-induced microvascular permeability (7, 37). However, despite evidence of NOS3 activation and NO generation, NO in this HCMVEC model appeared to play no major role in either the initial permeability or recovery phases. In HCMVEC, NOS3 seems not to be involved in the regulation of H2O2-induced permeability. l-NAME, at concentrations that inhibit NO production (Fig. 4), failed to affect the H2O2-elicited changes in TEER (see Fig. 6D). It is likely that the NO release seen in these cells could play other important roles in the heart. The release of NO by endothelial cells results in vasorelaxation and in overall improvement of circulation through the effect of NO on vascular wall smooth muscle cells (3) and NO modulation of cardiomyocyte contractility (22). In our own heart cell models, increased cardioprotective NO generated early during reperfusion can modulate ROS generation by the mitochondria and attenuate cardiomyocyte injury (29, 45). In addition, exogenous NO (as could be provided by neighboring endothelial cells) is highly protective against ischemia-reperfusion injury (21). Thus the NO generation seen in HCMVEC could serve many cardioprotective roles apart from the regulation of microvascular barrier integrity.

Role of Akt in endothelial cell integrity.

The present study examined the role of Akt because it has recently been implicated in endothelial cell barrier function regulation (31, 44). Since it can be simultaneously activated by levels of ROS that activate p38 (39) and induce barrier disruption (51), it is reasonable to think that Akt plays a role in regulating barrier dysfunction in the setting of oxidant stress. In addition, Akt is upstream of NOS3 activation (13) and cardioprotective NO generation (18, 22). Others have shown that H2O2 can activate PI3K-Akt signaling pathways and activate NOS3 via downstream NOS3 Ser-1177 phosphorylation (9, 34, 48). Our results suggesting that 250–500 μM H2O2 activates PI3K-Akt signaling in HCMVEC (see Fig. 1) are consistent with this previously reported work. The maximal stimulation of NOS3 Ser-1177 phosphorylation in porcine artery endothelial cells was achieved by 600 μM H2O2 (49). In other reports, NOS3 phosphorylation in the same cell type was achieved by 100 μM H2O2 but not with 500 μM H2O2 (48). In both reports, Akt was shown to be critical for eNOS Ser-1177 phosphorylation. Our finding that ROS-mediated Akt Ser-473 and Thr-308 phosphorylation in HCMVEC is mediated by the upstream activation of PI3K (see Fig. 5) is also consistent with work by others using different cell types (9, 49).

In human umbilical vein endothelial cells, Akt has recently been described as a regulator of endothelial chemotaxis that can increase barrier integrity (31). Other work in human pulmonary artery endothelial cells suggests that Akt can improve barrier integrity by phosphorylating the barrier-promoting sphingosine-1-phosphate receptor 1, inducing downstream RhoA/Rac1 signaling to the cytoskeleton (46). Conversely, Akt may mediate ROS stress-induced permeability changes in rat brain endothelial cells (44). In contrast to the current work, this study exposed cells to prolonged superoxide generation using xanthine/xanthine oxidase exposure. Our own results suggest for the first time that Akt in HCMVEC helps restore H2O2-induced permeability, given that the Akt inhibitor API-2 completely blocked HCMVEC ability to regain monolayer integrity (see Fig. 6B). Interestingly, API-2 had no effect on the basal TEER, indicating that Akt is not required to maintain the integrity of established cell contacts.

Conclusion

Our study suggests that H2O2-induced oxidant stress in HCMVEC activates NOS3 via Akt. NOS3/NO are not involved in the regulation of H2O2-affected barrier function in HCMVEC. Akt, independent of NOS3 regulation, proves to be critical for the restoration of barrier integrity in HCMVEC.

GRANTS

Funding support for this study was received from the National Heart, Lung, and Blood Institute Grants HL-68951, HL-65558, HL-79641, and HL-58064; the Office of Naval Research Grant N00014-04-1-0796; and the Emergency Resuscitation Center.

Acknowledgments

We thank our colleagues Kimberly Wojcik and Michael Retzer for expert medical writing and editorial assistance. We also acknowledge Lynne Harnish for substantial administrative help.

Notes

The costs of publication of this article were defrayed in part by the payment of page charges. The article must therefore be hereby marked “advertisement” in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

REFERENCES

1. Abella BSMDM, Zhao DMDP, Alvarado JBA, Hamann KP, Vanden Hoek TLMD, Becker LBMD. Intra-arrest cooling improves outcomes in a murine cardiac arrest model. Circulation 109: 2786–2791, 2004. [Abstract] [Google Scholar]
2. Anderson TC, Li CQ, Shao ZH, Hoang T, Chan KC, Hamann KJ, Becker LB, Vanden Hoek TL. Transient and partial mitochondrial inhibition for the treatment of postresuscitation injury: getting it just right. Crit Care Med 34: S474–S482, 2006. [Abstract] [Google Scholar]
3. Arnal JF, Dinh-Xuan AT, Pueyo M, Darblade B, Rami J. Endothelium-derived nitric oxide and vascular physiology and pathology. Cell Mol Life Sci 55: 1078–1087, 1999. [Abstract] [Google Scholar]
4. Berg RA, Sanders AB, Kern KB, Hilwig RW, Heidenreich JW, Porter ME, Ewy GA. Adverse hemodynamic effects of interrupting chest compressions for rescue breathing during cardiopulmonary resuscitation for ventricular fibrillation cardiac arrest. Circulation 104: 2465–2470, 2001. [Abstract] [Google Scholar]
5. Boo YC, Jo H. Flow-dependent regulation of endothelial nitric oxide synthase: role of protein kinases. Am J Physiol Cell Physiol 285: C499–C508, 2003. [Abstract] [Google Scholar]
6. Bottiger BWMD, Motsch JMD, Braun VMD, Martin EMD, Kirschfink MDVMP. Marked activation of complement and leukocytes and an increase in the concentrations of soluble endothelial adhesion molecules during cardiopulmonary resuscitation and early reperfusion after cardiac arrest in humans. Critical Care Medicine 30: 2473–2480, 2002. [Abstract] [Google Scholar]
7. Brkovic A, Sirois MG. Vascular permeability induced by VEGF family members in vivo: role of endogenous PAF and NO synthesis. J Cell Biochem 100: 727–737, 2007. [Abstract] [Google Scholar]
8. Caceres MJ, Schleien CL, Kuluz JW, Gelman B, Dietrich WD. Early endothelial damage and leukocyte accumulation in piglet brains following cardiac arrest. Acta Neuropathol (Berl) 90: 582–591, 1995. [Abstract] [Google Scholar]
9. Cai H, Li Z, Davis ME, Kanner W, Harrison DG, Dudley SC Jr. Akt-dependent phosphorylation of serine 1179 and mitogen-activated protein kinase kinase/extracellular signal-regulated kinase 1/2 cooperatively mediate activation of the endothelial nitric-oxide synthase by hydrogen peroxide. Mol Pharmacol 63: 325–331, 2003. [Abstract] [Google Scholar]
10. Chan TO, Rittenhouse SE, Tsichlis PN. AKT/PKB and other D3 phosphoinositide-regulated kinases: kinase activation by phosphoinositide-dependent phosphorylation. Annu Rev Biochem 68: 965–1014, 1999. [Abstract] [Google Scholar]
11. Connor KM, Subbaram S, Regan KJ, Nelson KK, Mazurkiewicz JE, Bartholomew PJ, Aplin AE, Tai YT, Aguirre-Ghiso J, Flores SC, Melendez JA. Mitochondrial H2O2 regulates the angiogenic phenotype via PTEN oxidation. J Biol Chem 280: 16916–16924, 2005. [Abstract] [Google Scholar]
12. Datta SR, Brunet A, Greenberg ME. Cellular survival: a play in three Akts. Genes Dev 13: 2905–2927, 1999. [Abstract] [Google Scholar]
13. Dimmeler S, Fleming I, Fisslthaler B, Hermann C, Busse R, Zeiher AM. Activation of nitric oxide synthase in endothelial cells by Akt-dependent phosphorylation. Nature 399: 601–605, 1999. [Abstract] [Google Scholar]
14. Fischer S, Wiesnet M, Renz D, Schaper W. H2O2 induces paracellular permeability of porcine brain-derived microvascular endothelial cells by activation of the p44/42 MAP kinase pathway. Eur J Cell Biol 84: 687–697, 2005. [Abstract] [Google Scholar]
15. Fleming I, Busse R. Molecular mechanisms involved in the regulation of the endothelial nitric oxide synthase. Am J Physiol Regul Integr Comp Physiol 284: R1–R12, 2003. [Abstract] [Google Scholar]
16. Fleming I, Fisslthaler B, Dimmeler S, Kemp BE, Busse R. Phosphorylation of Thr495 regulates Ca2+/calmodulin-dependent endothelial nitric oxide synthase activity. Circ Res 88: E68–E75, 2001. [Abstract] [Google Scholar]
17. Fries M, Tang W, Chang YT, Wang J, Castillo C, Weil MH. Microvascular blood flow during cardiopulmonary resuscitation is predictive of outcome. Resuscitation 71: 248–253, 2006. [Abstract] [Google Scholar]
18. Fulton D, Gratton JP, McCabe TJ, Fontana J, Fujio Y, Walsh K, Franke TF, Papapetropoulos A, Sessa WC. Regulation of endothelium-derived nitric oxide production by the protein kinase Akt. Nature 399: 597–601, 1999. [Abstract] [Google Scholar]
19. Giaever I, Keese CR. A morphological biosensor for mammalian cells. Nature 366: 591–592, 1993. [Abstract] [Google Scholar]
20. Idris AH, Roberts LJ 2nd, Caruso L, Showstark M, Layon AJ, Becker LB, Vanden Hoek T, Gabrielli A. Oxidant injury occurs rapidly after cardiac arrest, cardiopulmonary resuscitation, reperfusion. Crit Care Med 33: 2043–2048, 2005. [Abstract] [Google Scholar]
21. Iwase H, Robin E, Guzy RD, Mungai PT, Vanden Hoek TL, Chandel NS, Levraut J, Schumacker PT. Nitric oxide during ischemia attenuates oxidant stress and cell death during ischemia and reperfusion in cardiomyocytes. Free Radic Biol Med 43: 590–599, 2007. [Abstract] [Google Scholar]
22. Jones SP, Bolli R. The ubiquitous role of nitric oxide in cardioprotection. J Mol Cell Cardiol 40: 16–23, 2006. [Abstract] [Google Scholar]
23. Kern KB, Hilwig RW, Rhee KH, Berg RA. Myocardial dysfunction after resuscitation from cardiac arrest: an example of global myocardial stunning. J Am Coll Cardiol 28: 232–240, 1996. [Abstract] [Google Scholar]
24. Kevil CG, Oshima T, Alexander B, Coe LL, Alexander JS. H2O2-mediated permeability: role of MAPK and occludin. Am J Physiol Cell Physiol 279: C21–C30, 2000. [Abstract] [Google Scholar]
25. Kevil CG, Oshima T, Alexander JS. The role of p38 MAP kinase in hydrogen peroxide mediated endothelial solute permeability. Endothelium 8: 107–116, 2001. [Abstract] [Google Scholar]
26. Kilic E, Kilic U, Wang Y, Bassetti CL, Marti HH, Hermann DM. The phosphatidylinositol-3 kinase/Akt pathway mediates VEGF's neuroprotective activity and induces blood brain barrier permeability after focal cerebral ischemia. FASEB J 20: 1185–1187, 2006. [Abstract] [Google Scholar]
27. Kuruvilla L, Kartha CC. Molecular mechanisms in endothelial regulation of cardiac function. Mol Cell Biochem 253: 113–123, 2003. [Abstract] [Google Scholar]
28. Laurent I, Monchi M, Chiche JD, Joly LM, Spaulding C, Bourgeois B, Cariou A, Rozenberg A, Carli P, Weber S, Dhainaut JF. Reversible myocardial dysfunction in survivors of out-of-hospital cardiac arrest. J Am Coll Cardiol 40: 2110–2116, 2002. [Abstract] [Google Scholar]
29. Lavani R, Chang WT, Anderson T, Shao ZH, Wojcik KR, Li CQ, Pietrowski R, Beiser DG, Idris AH, Hamann KJ, Becker LB, Vanden Hoek TL. Altering CO2 during reperfusion of ischemic cardiomyocytes modifies mitochondrial oxidant injury. Crit Care Med 35: 1709–1716, 2007. [Abstract] [Google Scholar]
30. Lebuffe G, Schumacker PT, Shao ZH, Anderson T, Iwase H, Vanden Hoek TL. ROS and NO trigger early preconditioning: relationship to mitochondrial KATP channel. Am J Physiol Heart Circ Physiol 284: H299–H308, 2003. [Abstract] [Google Scholar]
31. Lee JF, Zeng Q, Ozaki H, Wang L, Hand AR, Hla T, Wang E, Lee MJ. Dual roles of tight junction-associated protein, zonula occludens-1, in sphingosine 1-phosphate-mediated endothelial chemotaxis and barrier integrity. J Biol Chem 281: 29190–29200, 2006. [Abstract] [Google Scholar]
32. Liu X, Zweier JL. A real-time electrochemical technique for measurement of cellular hydrogen peroxide generation and consumption: evaluation in human polymorphonuclear leukocytes. Free Radic Biol Med 31: 894–901, 2001. [Abstract] [Google Scholar]
33. Michell BJ, Griffiths JE, Mitchelhill KI, Rodriguez-Crespo I, Tiganis T, Bozinovski S, de Montellano PR, Kemp BE, Pearson RB. The Akt kinase signals directly to endothelial nitric oxide synthase. Curr Biol 9: 845–848, 1999. [Abstract] [Google Scholar]
34. Mount PF, Kemp BE, Power DA. Regulation of endothelial and myocardial NO synthesis by multi-site eNOS phosphorylation. J Mol Cell Cardiol 42: 271–279, 2007. [Abstract] [Google Scholar]
35. Nichol G, Stiell IG, Laupacis A, Pham B, De Maio VJ, Wells GA. A cumulative meta-analysis of the effectiveness of defibrillator-capable emergency medical services for victims of out-of-hospital cardiac arrest. Ann Emerg Med 34: 517–525, 1999. [Abstract] [Google Scholar]
36. Niwa K, Inanami O, Ohta T, Ito S, Karino T, Kuwabara M. p38 MAPK and Ca2+ contribute to hydrogen peroxide-induced increase of permeability in vascular endothelial cells but ERK does not. Free Radic Res 35: 519–527, 2001. [Abstract] [Google Scholar]
37. Noel AA, Hobson RW, 2nd Duran WN. Platelet-activating factor and nitric oxide mediate microvascular permeability in ischemia-reperfusion injury. Microvasc Res 52: 210–220, 1996. [Abstract] [Google Scholar]
38. Oliver MG, Specian RD, Perry MA, Granger DN. Morphologic assessment of leukocyte-endothelial cell interactions in mesenteric venules subjected to ischemia and reperfusion. Inflammation 15: 331–346, 1991. [Abstract] [Google Scholar]
39. Posen Y, Kalchenko V, Seger R, Brandis A, Scherz A, Salomon Y. Manipulation of redox signaling in mammalian cells enabled by controlled photogeneration of reactive oxygen species. J Cell Sci 118: 1957–1969, 2005. [Abstract] [Google Scholar]
40. Predescu D, Predescu S, Shimizu J, Miyawaki-Shimizu K, Malik AB. Constitutive eNOS-derived nitric oxide is a determinant of endothelial junctional integrity. Am J Physiol Lung Cell Mol Physiol 289: L371–L381, 2005. [Abstract] [Google Scholar]
41. Qin Y, Vanden Hoek TL, Wojcik K, Anderson T, Li CQ, Shao ZH, Becker LB, Hamann KJ. Caspase-dependent cytochrome c release and cell death in chick cardiomyocytes after simulated ischemia-reperfusion. Am J Physiol Heart Circ Physiol 286: H2280–H2286, 2004. [Abstract] [Google Scholar]
42. Quadri SK, Bhattacharya J. Resealing of endothelial junctions by focal adhesion kinase. Am J Physiol Lung Cell Mol Physiol 292: L334–L342, 2007. [Abstract] [Google Scholar]
43. Schoenenberger RA, von Planta M, von Planta I. Survival after failed out-of-hospital resuscitation. Are further therapeutic efforts in the emergency department futile? Arch Intern Med 154: 2433–2437, 1994. [Abstract] [Google Scholar]
44. Schreibelt G, Kooij G, Reijerkerk A, van Doorn R, Gringhuis SI, van der Pol S, Weksler BB, Romero IA, Couraud PO, Piontek J, Blasig IE, Dijkstra CD, Ronken E, de Vries HE. Reactive oxygen species alter brain endothelial tight junction dynamics via RhoA, PI3 kinase, PKB signaling. FASEB J 21: 3666–3676, 2007. [Abstract] [Google Scholar]
45. Shao ZH, Chang WT, Chan KC, Wojcik KR, Hsu CW, Li CQ, Li J, Anderson T, Qin Y, Becker LB, Hamann KJ, Vanden Hoek TL. Hypothermia-induced cardioprotection using extended ischemia and early reperfusion cooling. Am J Physiol Heart Circ Physiol 292: H1995–H2003, 2007. [Abstract] [Google Scholar]
46. Singleton PA, Dudek SM, Ma SF, Garcia JG. Transactivation of sphingosine 1-phosphate receptors is essential for vascular barrier regulation. Novel role for hyaluronan and CD44 receptor family. J Biol Chem 281: 34381–34393, 2006. [Abstract] [Google Scholar]
47. Spaulding CM, Joly LM, Rosenberg A, Monchi M, Weber SN, Dhainaut JF, Carli P. Immediate coronary angiography in survivors of out-of-hospital cardiac arrest. N Engl J Med 336: 1629–1633, 1997. [Abstract] [Google Scholar]
48. Tanaka T, Nakamura H, Yodoi J, Bloom ET. Redox regulation of the signaling pathways leading to eNOS phosphorylation. Free Radic Biol Med 38: 1231–1242, 2005. [Abstract] [Google Scholar]
49. Thomas SR, Chen K, Keaney JF Jr. Hydrogen peroxide activates endothelial nitric-oxide synthase through coordinated phosphorylation and dephosphorylation via a phosphoinositide 3-kinase-dependent signaling pathway. J Biol Chem 277: 6017–6024, 2002. [Abstract] [Google Scholar]
50. Usatyuk PV, Natarajan V. Regulation of reactive oxygen species-induced endothelial cell-cell and cell-matrix contacts by focal adhesion kinase and adherens junction proteins. Am J Physiol Lung Cell Mol Physiol 289: L999–L1010, 2005. [Abstract] [Google Scholar]
51. Usatyuk PV, Vepa S, Watkins T, He D, Parinandi NL, Natarajan V. Redox regulation of reactive oxygen species-induced p38 MAP kinase activation and barrier dysfunction in lung microvascular endothelial cells. Antioxid Redox Signal 5: 723–730, 2003. [Abstract] [Google Scholar]
52. Vanden Hoek T, Becker LB, Shao ZH, Li CQ, Schumacker PT. Preconditioning in cardiomyocytes protects by attenuating oxidant stress at reperfusion. Circ Res 86: 541–548, 2000. [Abstract] [Google Scholar]
53. Vanden Hoek TL, Becker LB, Shao Z, Li C, Schumacker PT. Reactive oxygen species released from mitochondria during brief hypoxia induce preconditioning in cardiomyocytes. J Biol Chem 273: 18092–18098, 1998. [Abstract] [Google Scholar]
54. Venardos KM, Kaye DM. Myocardial ischemia-reperfusion injury, antioxidant enzyme systems, selenium: a review. Curr Med Chem 14: 1539–1549, 2007. [Abstract] [Google Scholar]
55. Zhao D, Abella BS, Beiser DG, Alvarado JP, Wang H, Hamann KJ, Vanden Hoek TL, Becker LB. Intra-arrest cooling with delayed reperfusion yields higher survival than earlier normothermic resuscitation in a mouse model of cardiac arrest. Resuscitation 77: 242–249, 2008. [Europe PMC free article] [Abstract] [Google Scholar]
56. Zimmerman BJ, Granger DN. Reperfusion injury. Surg Clin North Am 72: 65–83, 1992. [Abstract] [Google Scholar]

Articles from American Journal of Physiology - Heart and Circulatory Physiology are provided here courtesy of American Physiological Society

Citations & impact 


Impact metrics

Jump to Citations
Jump to Data

Citations of article over time

Smart citations by scite.ai
Smart citations by scite.ai include citation statements extracted from the full text of the citing article. The number of the statements may be higher than the number of citations provided by EuropePMC if one paper cites another multiple times or lower if scite has not yet processed some of the citing articles.
Explore citation contexts and check if this article has been supported or disputed.
https://scite.ai/reports/10.1152/ajpheart.00501.2008

Supporting
Mentioning
Contrasting
6
18
0

Article citations


Go to all (24) article citations

Data 


Funding 


Funders who supported this work.

NHLBI NIH HHS (4)