Department of Surgery, University of Medicine and Dentistry-New Jersey Medical School, Newark, New Jersey 07103
![]() |
ABSTRACT |
---|
![]() ![]() ![]() ![]() ![]() ![]() ![]() |
---|
Na+/H+
exchanger (NHE) activation has been documented to contribute to
endothelial cell injury caused by inflammatory states. However, the
role of NHEs in regulation of the endothelial cell inflammatory
response has not been investigated. The present study tested the
hypothesis that NHEs contribute to endothelial cell inflammation
induced by endotoxin or interleukin (IL)-1. NHE inhibition using
amiloride, 5-(N-ethyl-N-isopropyl)-amiloride, and
5-(N-methyl-N-isobutyl)amiloride as well as the
non-amiloride NHE inhibitors cimetidine, clonidine, and harmaline
suppressed endotoxin-induced IL-8 and monocyte chemoattractant protein
(MCP)-1 production by human umbilical endothelial vein cells (HUVECs). The suppressive effect of amiloride on endotoxin-induced IL-8 production was associated with a decreased accumulation of IL-8 mRNA.
NHE inhibitors suppressed both inhibitory (I)
B degradation and
nuclear factor (NF)-
B DNA binding, suggesting that a decrease in
activation of the I
B-NF-
B system contributed to the suppression of HUVEC inflammatory response by NHE blockade. NHE inhibition decreased also the IL-1
-induced HUVEC inflammatory response, because
amiloride suppressed IL-1
-induced E-selectin expression on HUVECs.
These results demonstrate that maximal activation of the HUVEC
inflammatory response requires a functional NHE.
Na+/H+ exchanger; transcription factors; inflammation; cytokines; sepsis; adhesion molecule
![]() |
INTRODUCTION |
---|
![]() ![]() ![]() ![]() ![]() ![]() ![]() |
---|
NA+/H+ EXCHANGERS (NHEs) are a group of integral transmembrane proteins found in all mammalian cells and play a role in the regulation of intracellular pH, cell volume, vectorial ion transport, and cell proliferation (31). There is a large body of evidence demonstrating the presence of NHE activity in endothelial cells (2, 7, 12, 18, 19). NHEs in endothelial cells have been shown to be activated by a variety of stimuli, including acidosis (2, 19), hypoxia-reoxygenation (18), extracellular nucleosides (12), and nucleotides (7). Pharmacological inhibition of the increased activity of NHEs prevents endothelial cell injury and swelling induced by acidosis (2, 19) and decreases the detrimental elevation of intracellular Ca2+ caused by hypoxia-reoxygenation (18), suggesting that NHE activation contributes to endothelial cell damage during acidosis and hypoxia-reoxygenation. In agreement with these in vitro observations, there is accumulating in vivo evidence that the activation of NHEs contributes to endothelial cell injury during a variety of pathophysiological processes associated with hypoxia and acidosis. For example, Mazzoni et al. (23) reported that the inhibition of NHEs by amiloride prevents capillary swelling and the consequent luminal narrowing during hemorrhagic shock. Furthermore, pharmacological blockade of NHEs prevents the increase in lung endothelial permeability induced by either septic shock (32) or hemorrhagic shock (24).
Pathophysiological states associated with hypoxia-reoxygenation and
acidosis, such as shock and sepsis, are also characterized by an
enhanced endothelial cell inflammatory response (20). During shock and sepsis, endothelial cells become chemoattractant, facilitating leukocyte adhesion, activation, and transmigration. There
are numerous chemoattractant agents produced at the site of injury that
participate in the recruitment of leukocytes, which include chemokines,
platelet-activating factor, leukotriene B4, the complement
products C5a and C3a, and granulocyte-monocyte colony-stimulating
factor. Chemokines are small cytokines, with molecular masses in the
range of 8-12 kDa (39). Chemokine domains are defined
by the presence of four cysteines in highly conserved position. One
major chemokine subfamily is called "CXC" because the two amino
acids nearest the NH2 termini of these proteins are
separated by a single amino acid. This is in contrast to the other
major subfamily, which is called "CC" because these two cysteines
are adjacent. The prototypic CXC chemokine is interleukin (IL)-8, which
has been purified by several groups as a monocyte-derived factor that
attracts neutrophils but not monocytes. IL-8 is also the major CXC
chemokine secreted by endothelial cells. On the other hand, the CC
chemokine monocyte chemoattractant protein (MCP)-1 is the most abundant
chemokine released by endothelial cells. The most important inducers of
chemokines in endothelial cells are bacterial products, such as
bacterial lipopolysaccharide (LPS, endotoxin), as well as the
monocyte/macrophage-derived cytokines tumor necrosis factor (TNF)-
and IL-1 (40).
Recently, NHEs have been implicated in the regulation of chemokine
production. Inhibition of NHEs suppresses IL-8 production in
respiratory epithelium infected with respiratory syncytial virus as
well as in immunostimulated monocytes (21, 36). In addition, we have recently demonstrated that NHE inhibition suppresses macrophage inflammatory protein (MIP)-1 and MIP-2 production by
LPS-stimulated macrophages (26). These
observations suggest that NHE activation may be an important
contributory factor to chemokine production during inflammatory cell
activation. Thus we hypothesized that NHEs may be involved in the
regulation of the endothelial cell chemokine response. The results of
this study present evidence that, similar to other cell types, NHE
activation is involved in the promotion of chemokine production in
endothelial cells. Furthermore, our results demonstrate that one of the
major intracellular targets of the proinflammatory effect of NHE
activation is the inhibitory (I)
B-nuclear factor (NF)-
B system.
![]() |
MATERIALS AND METHODS |
---|
![]() ![]() ![]() ![]() ![]() ![]() ![]() |
---|
Cell culture. Human umbilical vein endothelial cells (HUVECs) were obtained from BioWhittaker (Walkersville, MD) and cultured in endothelial growth medium. The HUVECs used for all experiments were from a Caucasian female umbilical cord. The cells were grown in a humidified atmosphere of 95% air-5% CO2 at 37 °C. Second- to fourth-passage HUVECs were used in all experiments.
Drugs and reagents.
The selective NHE inhibitors amiloride HCl,
5-(N-ethyl-N-isopropyl)-amiloride (EIPA), and
5-(N-methyl-N-isobutyl)amiloride (MIA) were
obtained from Research Biochemicals (Natick, MA). The nonselective NHE
inhibitors cimetidine, harmaline, and clonidine were purchased from
Sigma (St. Louis, MO). All NHE inhibitors were dissolved in 0.5% DMSO.
Human IL-1 was obtained from R&D Systems (Minneapolis, MN). LPS
(Escherichia coli 055:B5) was purchased from Sigma.
3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) was
obtained from Fisher Scientific (Pittsburgh, PA).
Chemokine measurement. To study the effect of NHE inhibitors, cells in 96-well plates were pretreated with these agents 30 min before stimulation with LPS (10 µg/ml). IL-8 and MCP-1 production were measured from supernatants taken 6, 12, 18, and 24 h after the LPS challenge. However, because both MIA and EIPA were toxic to the cells when the incubation lasted for 24 h, the effect of these agents on IL-8 and MCP-1 production was tested 4 h after LPS stimulation. Cimetidine, harmaline, and clonidine were administered to the cells 30 min before addition of LPS. The effect of these agents on IL-8 production was tested from supernatants obtained 8 h after LPS stimulation. Human IL-8 and MCP-1 levels in the supernatants were determined by using commercially available ELISA kits (R&D Systems) according to the manufacturer's instructions.
Western blot analysis for the assessment of IB
degradation.
HUVECs in six-well plates were pretreated with amiloride (300 µM) or
vehicle (0.5% DMSO), and 30 min later the cells were stimulated with
LPS (10 µg/ml) for 45 min. After being washed with PBS, the cells
were lysed by the addition of modified radioimmunoprecipitation buffer
[50 mM Tris · HCl, 150 mM NaCl, 1 mM EDTA, 0.25%
Na-deoxycholate, 1% NP-40, 1 µg/ml pepstatin, 1 µg/ml leupeptin, 1 mM phenylmethylsulfonyl fluoride (PMSF), 1 mM
Na3VO4]. The lysates were transferred to Eppendorf tubes and centrifuged at 15,000 g, and the
supernatant was recovered. Protein concentrations were determined by
using the Bio-Rad protein assay (Bio-Rad, Hercules, CA). Sample (10 µg) was separated on a 8-16% Tris-glycine gel (Invitrogen,
Carlsbad, CA) and transferred to a nitrocellulose membrane. The
membranes were probed with anti-I
B
antibody (Cell Signaling,
Beverly, MA) and subsequently incubated with a secondary horseradish
peroxidase-conjugated donkey anti-rabbit antibody (Boehringer,
Indianapolis, IN). Bands were detected by using enhanced
chemiluminescence Western blotting detection reagent (ECL; Amersham
Life Science, Arlington Heights, IL).
RNA isolation and RT-PCR. HUVECs in six-well plates were pretreated with amiloride (300 µM) or vehicle (0.5% DMSO), and 30 min later the cells were stimulated with LPS (10 µg/ml) for 4 h. Total RNA was isolated from HUVECs by using Trizol reagent (Invitrogen). RT of the RNA was performed by using MuLV reverse transcriptase (50 U/µl) from Perkin Elmer (Foster City, CA). RNA (5 µg) was transcribed in a 20-µl reaction containing 10.7 µl of RNA, 2 µl of 10× PCR buffer, 2 µl of 10 mM dNTP mix, 2 µl of 25 mM MgCl2, 2 µl of 100 mM dithiothreitol (DTT), 0.5 µl of RNase inhibitor (20 U/µl; Perkin Elmer), 0.5 µl of 50 mM oligo d(T)16, and 0.3 µl of reverse transcriptase. The reaction mix was incubated at 42°C for 15 min for reverse transcription. Thereafter, the reverse transcriptase was inactivated at 99°C for 5 min. RT-generated DNA was amplified by using the Expand high-fidelity PCR system (Boehringer Mannheim). The reaction buffer (25 µl) contained 1-5 µl of cDNA, water, 2.5 µl of 10× PCR buffer, 1.5 µl of 25 mM MgCl2, 1 µl of 10 mM dNTP mix, 0.5 µl of 10 mM oligonucleotide primer (each), and 0.2 µl of enzyme. cDNA was amplified by using the following primers and conditions: IL-8 (27), 5'-ATGACTTCCAAGCTGGCCGTGGCT-3' (sense) and 5'-TCTCAGCCCTCTTCAAAAACTTCTC-3' (antisense); and glyceraldehyde-3-phosphate dehydrogenase (GAPDH), 5'-CGGAGTCAACGGATTTGGTCGTAT-3' (sense) and 5'-AGCCTTCTCCATGGTGGTGAAGAC-3' (anti-sense). An initial denaturation occurred at 94°C for 5 min, with 27 and 23 cycles of 94°C for 30 s for IL-8 and GAPDH, respectively, 58°C for 45 s, 72°C for 45 s, and a final dwell at 72°C for 7 min. The expected PCR products were IL-8 (289 bp) and GAPDH (306 bp). PCR products were resolved on a 1.5% agarose gel and stained with ethidium bromide.
NF-B electrophoretic mobility shift assay and supershift
assay.
After pretreatment with amiloride or its vehicle for 30 min, HUVECs in
75-ml tissue culture flasks were stimulated with LPS (10 µg/ml) for
45 min, and nuclear protein extracts were prepared as described
previously (13, 25). All nuclear extraction procedures were performed on ice with ice-cold reagents. Cells were washed twice
with PBS and harvested by scraping into 1 ml of PBS and pelleted at
6,000 g for 5 min. The pellet was resuspended in one packed
cell volume of lysis buffer (10 mM HEPES, pH 7.9, 10 mM KCl, 0.1 mM
EDTA, 1.5 mM MgCl2, 0.2% vol/vol Nonidet P-40, 1 mM DTT,
and 0.1 mM PMSF) and incubated for 5 min with occasional vortexing.
After centrifugation at 6,000 g, one cell pellet volume of
extraction buffer (20 mM HEPES, pH 7.9, 10 mM KCl, 0.1 mM EDTA, 1.5 mM
MgCl2, 25% vol/vol glycerol, 1 mM DTT, and 0.5 mM PMSF) was added to the nuclear pellet and incubated on ice for 15 min with
occasional vortexing. Nuclear proteins were isolated by centrifugation at 14,000 g for 15 min. Protein concentrations were
determined with the Bio-Rad protein assay. Nuclear extracts were stored
at
70°C until used for electrophoretic mobility shift assay (EMSA). The oligonucleotide probe used for the EMSA was purchased from Promega.
Oligonucleotide probes were labeled with [
-32P]ATP by
using T4 polynucleotide kinase (Invitrogen) and then purified in Bio
Spin chromatography columns (Bio-Rad). For the EMSA analysis, 10 µg
of nuclear protein were preincubated with EMSA buffer [12 mM HEPES, pH
7.9, 4 mM Tris · HCl, pH 7.9, 25 mM KCl, 5 mM
MgCl2, 1 mM EDTA, 1 mM DTT, 50 ng/ml poly(dI-dC), 12%
glycerol vol/vol, and 0.2 mM PMSF] on ice for 10 min before addition
of the radiolabeled oligonucleotide for an additional 25 min. The
specificities of the binding reactions were tested by incubating
duplicate samples with 100-fold molar excess of the unlabeled
oligonucleotide probe. Protein-nucleic acid complexes were resolved by
using a nondenaturing polyacrylamide gel consisting of 5% acrylamide
(29:1 ratio of acrylamide:bisacrylamide) and run in 0.5× TBE (45 mM Tris · HCl, 45 mM boric aid, and 1 mM EDTA) for 1 h at
constant current (30 mA). Gels were transferred to Whatman 3M paper,
dried under vacuum at 80°C for 1 h, and exposed to photographic
film at
70°C with an intensifying screen. For supershift studies,
after addition of the radiolabeled probe, samples were incubated for
1 h with control, p65, or p50 antibody (Santa Cruz, CA) and then
loaded on the gel.
Cell surface ELISA for the determination of E-selectin.
HUVECs were seeded at 20,000 cells/well onto Biocoat Matrigel 96-well
plates (Becton Dickinson, Bedford, MA) and used for the enzyme
immunoassay protocols. To test the effect of NHE inhibition on HUVEC
E-selectin expression, cells were pretreated with amiloride or its
vehicle for 30 min. Subsequently, the cells were stimulated with 20 ng/ml IL-1 for 4 h. At the end of the incubation period, the
supernatant was removed and the cells were washed twice with PBS and
fixed with 1.5% paraformaldehyde for 30 min. Subsequently, the cells
were blocked with 2% bovine serum albumin and then incubated for
1 h at 37°C with a monoclonal antibody to E-selectin (BD
PharMingen, San Diego, CA). The cells were then incubated with an
alkaline phosphatase-conjugated secondary antibody for 1 h. The
cells were again washed three times, after which
p-nitrophenyl disodium phosphate (1 mg/ml; Sigma) was added,
and incubated at room temperature for 30 min. The colorimetric reaction
was then read in a plate reader at 410 nm.
Measurement of mitochondrial respiration. Mitochondrial respiration, an indicator of cell viability, was assessed by the mitochondrion-dependent reduction of MTT to formazan. Cells in 96-well plates were incubated with MTT (0.5 mg/ml) for 60 min at 37°C. Culture medium was removed by aspiration, and the cells were solubilized in DMSO (100 µl). The extent of reduction of MTT to formazan within cells was quantitated by measurement of optical density at 550 nm using a Spectramax 250 microplate reader (17).
![]() |
RESULTS |
---|
![]() ![]() ![]() ![]() ![]() ![]() ![]() |
---|
NHE inhibition suppresses IL-8 and MCP-1 production by HUVECs.
First, we examined the effect of NHE inhibition on the production of
IL-8 by HUVECs. Stimulation of HUVECs with LPS for 24 h increased
IL-8 production by these cells from 1.26 ± 0.18 to 6.65 ± 0.26 ng/ml (n = 6, P < 0.01).
Amiloride pretreatment of the HUVECs caused a dose-dependent,
significant blunting of the IL-8 response to LPS measured at the 24-h
time point (Fig. 1A). This
inhibitory effect was not associated with any decrease in cell
viability, as determined by the MTT assay (Fig. 1B). In the next set of experiments, we examined the effect of amiloride
(300 µM) pretreatment on LPS-induced IL-8 production various
time periods after LPS stimulation. Similar to results at the 24-h time
point, amiloride also attenuated LPS-induced IL-8 production 12 and
18 h after stimulation with LPS (Fig. 1C). Importantly,
the effect of amiloride was more pronounced at these early time points
than at 24 h, because amiloride suppressed IL-8 production at 12 and 18 h by 61 and 54%, respectively, whereas the suppressive
effect of amiloride amounted only to 30% at 24 h after the LPS
challenge (Fig. 1C). Also similar to results at the 24-h
point, amiloride failed to affect cell viability at any of the earlier
time points tested (data not shown).
|
|
|
|
Amiloride suppresses LPS-induced IL-8 mRNA accumulation in HUVECs.
To determine whether the suppressive effect of amiloride on IL-8
protein production was associated with an effect on the accumulation of
IL-8 mRNA, we measured IL-8 mRNA levels from amiloride-pretreated HUVECs and controls. As shown in Fig. 5,
LPS treatment for 4 h induced the accumulation of IL-8 mRNA.
Amiloride pretreatment of the cells 30 min before the LPS challenge
decreased the LPS-induced accumulation of IL-8 mRNA. GAPDH mRNA levels
were not affected by amiloride treatment (Fig. 5), demonstrating that
amiloride did not cause a general suppression of mRNA accumulation in
HUVECs.
|
NHE inhibition inhibits LPS-induced NF-B activation in HUVECs.
Because the effect of amiloride was pretranslational, we hypothesized
that amiloride may have decreased the rate of transcription due to an
effect on transcription factor activation. Because NF-
B is one of
the most important transcription factors that mediates transcription of
the IL-8 gene in response to LPS in HUVECs (6, 34, 35,
50), we first examined whether amiloride altered the binding of
NF-
B to its consensus site by using the gel mobility shift assay. As
shown in Fig. 6, using nuclear extracts
from HUVECs treated with LPS, we observed a NF-
B DNA-binding complex
that was not seen in LPS-nontreated cells (Fig. 6A).
Supershift studies confirmed that the band induced by LPS contains both
p65 and p50, because both the p50 and p65 antibodies shifted this
complex (Fig. 6A). The increase in NF-
B binding caused by
LPS was partially prevented when the cells were pretreated with
amiloride 30 min before LPS treatment. This observation suggests that
the mechanism of action of amiloride in suppressing IL-8 production
involves an interference with the NF-
B pathway.
|
|
Amiloride suppresses IL-1-induced E-selectin expression.
We next determined whether blockade of NHEs by amiloride attenuated the
expression of E-selectin. Stimulation of HUVECs with IL-1
caused a
substantial upregulation of E-selectin expression (Fig.
8). Amiloride pretreatment of the cells
30 min before the IL-1
challenge attenuated the IL-1
-induced
E-selectin response in a concentration-dependent manner (Fig. 8). This
observation demonstrates that NHE inhibition decreases the
IL-1
-induced HUVEC inflammatory response.
|
![]() |
DISCUSSION |
---|
![]() ![]() ![]() ![]() ![]() ![]() ![]() |
---|
NHEs are a family of ubiquitous plasma membrane transport proteins
that catalyze the exchange of extracellular Na+ for
intracellular H+ (31). However, NHEs have
additionally been implicated in other cellular functions. For example,
because they are stimulated by a variety of growth factors, a role in
the initiation and/or control of mitogenesis has been proposed
(16). In addition, NHEs are believed to be central to the
maintenance and regulation of cell volume (49). In
epithelial cells of the intestine and kidney, the apical NHE plays an
important role in Na+ reabsorption and acid secretion
(9). Recent evidence indicates that NHEs regulate
inflammatory processes. NHEs are rapidly activated in response to a
variety of inflammatory signals, such as IL-1 (3, 5),
TNF- (43), interferon-
(33),
colony-stimulating factor 1 (42), and bacterial LPS
(30, 43). This activation of NHEs by inflammatory stimuli
crucially contributes to inflammatory responses of "professional"
inflammatory cells, such as monocytes (30) and macrophages
(33). These include TNF-
and IL-8 production (29,
30, 36), prostaglandin release (11), upregulation of Ia expression (33), Fc
receptor expression
(4), and colony-stimulating factor-induced proliferation
(42). There is also evidence that the activation of NHEs
is involved in promoting neutrophil migration (37) as well
as myeloperoxidase activity and release (45). We recently
demonstrated that inhibition of NHEs suppressed IL-12, MIP-1
, and
MIP-2 production by LPS-stimulated macrophages (26). Furthermore, in the same study, we showed that inhibition of the Na+/H + antiporter reduced plasma levels of
IL-12 in an endotoxemic mouse model. Finally, inhibition of NHEs by
amiloride suppresses IL-8 and IL-6 production in respiratory epithelium
infected with respiratory syncytial virus (21).
Recent molecular cloning studies have confirmed that NHEs constitute a
gene family from which seven mammalian isoforms (NHE1-7) have been
cloned and sequenced (28, 31, 41). Characterization of the
individual NHE isoforms has revealed differences in their primary
structure, mode of regulation, cellular localization, and tissue
expression. Despite the recent large expansion in our understanding of
the molecular biology of NHEs, the most important tool to characterize
the function of these proteins remains their pharmacological inhibition
by the antidiuretic drug amiloride and its analogs as well as several
structurally unrelated molecules, including cimetidine, clonidine, and
harmaline (41, 47). Although amiloride at low, nanomolar
concentrations inhibits epithelial Na+ channels (ENaCs)
(22), effects that only appear at high amiloride concentrations in the micromolar range are thought to be exclusively selective for NHEs (31). The data presented here
demonstrate, for the first time, that NHE activation contributes to the
endothelial cell inflammatory response induced by LPS or IL-1. The
fact that amiloride and its analogs suppressed IL-8 and MCP-1
production in HUVECs with an IC50 value of 30-100 µM
excludes a role for ENaCs in the amiloride suppression of endothelial
cell inflammatory response. The idea that NHE inhibition suppresses the
endothelial cell inflammatory response was confirmed by using
three additional non-amiloride NHE inhibitors, clonidine, harmaline,
and cimetidine, since all three of these inhibitors suppressed
LPS-induced IL-8 production.
Cutaia et al. (8) have recently demonstrated that human pulmonary artery endothelial cells possess NHE1 but do not express NHE2-4. Furthermore, HUVECs have also been shown to express NHE1 (51); however, the expression of other subtypes has not been investigated. Nevertheless, it appears unlikely that NHE1 or NHE2 is the target of the suppressive effect of NHE inhibitors on inflammatory function in HUVECs. Both NHE1 and NHE2 are inhibited by amiloride concentrations not exceeding 1 µM (41, 47), and the IC50 values for NHE1 and NHE2 inhibition by EIPA and MIA are in the nanomolar range (41, 47). However, in our study, neither amiloride nor its analogs suppressed chemokine production at concentrations <10 µM. Thus it is improbable that either NHE1 or NHE2 is responsible for the attenuation of chemokine production by amiloride or its analogs. Interestingly, both amiloride and EIPA blocked thrombin-induced platelet-activating factor production by HUVECs at concentrations comparable to those that suppressed IL-8 production in our study (14). These results confirm the notion that HUVECs express amiloride-insensitive NHEs that regulate inflammatory processes. This view is further supported by the observation that harmaline, cimetidine, and clonidine decreased IL-8 production with IC50 values between 0.3 and 3 mM, which are higher than would have been expected if the NHE1 or NHE2 was involved (41, 47). Because NHE5 is also sensitive to nanomolar concentrations of EIPA (41, 47), it appears unlikely that this isoform plays a role in the regulation of IL-8 production. On the other hand, because both NHE3 and NHE4 have been documented to be inhibited by amiloride or non-amiloride compounds with a similar potency (41, 47) to that observed for the suppression of IL-8 production by HUVECs, NHE3 or NHE4 could be the subtype mediating the inhibitory effect of NHE inhibitors on IL-8 production. Because the NHE6 and NHE7 isoforms have not been characterized pharmacologically, their role in the regulation of endothelial cell IL-8 production remains to be elucidated.
Another important finding of the present study is that the suppressive effect of NHE inhibition on IL-8 production appears to have a pretranslational component, because amiloride suppressed LPS-induced IL-8 mRNA levels. This idea is supported by the observation that the effect of amiloride on IL-8 production was more pronounced early after LPS stimulation than at the 24-h time point. However, it is unlikely that the Golgi-related early secretion of IL-8 from the Weibel-Palade bodies (46) could be the target of NHE inhibition, because NHE inhibition failed to suppress IL-8 production 6 h after LPS stimulation.
We believe that the demonstration of the decreasing effect of NHE
inhibition on the IB-NF-
B system is a major step toward answering
the question of how NHEs regulate endothelial cell or other
inflammatory responses. The importance of NF-
B as a central mediator
of the immunoregulatory effects of NHEs is highlighted by the fact that
NF-
B activation is also under the control of NHEs in other cell
types, such as the intestinal epithelial cell line HT-29
(25) and respiratory epithelial cells (15).
These findings raise the important question of how a signal provided by
the membrane protein NHE is transmitted to the cytosolic protein NF-
B. One of the most intriguing possibilities is that it is the
alteration of cytoskeletal organization of actin filaments that links
NHEs to NF-
B activation. This possibility is supported by the fact
that NHEs are important regulators of actin filament assembly
(10, 44, 48) and that changes in the actin microfilament system are involved in activation of the NF-
B system (1,
52). In this respect, it is important to note that NHE3
(38, 44, 48) has been reported to regulate cytoskeletal
organization. The tethering of actin filaments to the plasma membrane
NHE3 is mediated in part by the ezrin, radixin, and moesin (ERM) family of actin-binding proteins (38, 44, 48). Clearly, further studies are necessary to delineate the mechanisms whereby NHEs couple
extracellular inflammatory signals to activation of the intracellular
inflammatory cascade and NF-
B.
In summary, we have demonstrated that NHE activation contributes to the endothelial cell inflammatory response to inflammatory stimuli. We speculate that the mechanism of NHE promotion of inflammatory processes may have evolved as a positive feedback signal during endothelial cell activation.
![]() |
FOOTNOTES |
---|
Address for reprint requests and other correspondence: G. Haskó, Dept. of Surgery, UMD-New Jersey Medical School, 185 South Orange Ave., Univ. Heights, Newark, NJ 07103 (E-mail: haskoge{at}umdnj.edu).
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.
March 13, 2002;10.1152/ajpcell.00491.2001
Received 12 October 2001; accepted in final form 10 March 2002.
![]() |
REFERENCES |
---|
![]() ![]() ![]() ![]() ![]() ![]() ![]() |
---|
1.
Are, AF,
Galkin VE,
Pospelova TV,
and
Pinaev GP.
The p65/RelA subunit of NF-B interacts with actin-containing structures.
Exp Cell Res
256:
533-544,
2000[ISI][Medline].
2.
Behmanesh, S,
and
Kempski O.
Mechanisms of EC swelling from lactacidosis studied in vitro.
Am J Physiol Heart Circ Physiol
279:
H1512-H1517,
2000
3.
Benos, DJ,
McPherson S,
Hahn BH,
Chaikin MA,
and
Benveniste EN.
Cytokines and HIV envelope glycoprotein gp120 stimulate Na+/H+ exchange in astrocytes.
J Biol Chem
269:
13811-13816,
1994
4.
Cassatella, MA,
Flynn RM,
Amezaga MA,
Bazzoni F,
Vicentini F,
and
Trinchieri G.
Interferon gamma induces in human neutrophils and macrophages expression of the mRNA for the high affinity receptor for monomeric IgG (FcR-I or CD64).
Biochem Biophys Res Commun
170:
582-588,
1990[ISI][Medline].
5.
Civitelli, R,
Teitelbaum SL,
Hruska KA,
and
Lacey DL.
IL-1 activates the Na+/H+ antiport in a murine T cell.
J Immunol
143:
4000-4008,
1989
6.
Cooper, JT,
Stroka DM,
Brostjan C,
Palmetshofer A,
Bach FH,
and
Ferran C.
A20 blocks EC activation through a NF-B-dependent mechanism.
J Biol Chem
271:
18068-18073,
1996
7.
Cutaia, M,
Dawicki DD,
Papazian LM,
Parks N,
Clarke E,
and
Rounds S.
Differences in nucleotide effects on intracellular pH, Na+/H+ antiport activity, and ATP-binding proteins in ECs.
In Vitro Cell Dev Biol Anim
33:
608-614,
1997[ISI][Medline].
8.
Cutaia, MV,
Parks N,
Centracchio J,
Rounds S,
Yip KP,
and
Sun AM.
Effect of hypoxic exposure on Na+/H+ antiport activity, isoform expression, and localization in ECs.
Am J Physiol Lung Cell Mol Physiol
275:
L442-L451,
1998
9.
Daniel, H.
Function and molecular structure of brush border membrane peptide/H+ symporters.
J Membr Biol
154:
197-203,
1996[ISI][Medline].
10.
Denker, SP,
Huang DC,
Orlowski J,
Furthmayr H,
and
Barber DL.
Direct binding of the Na-H exchanger NHE1 to ERM proteins regulates the cortical cytoskeleton and cell shape independently of H+ translocation.
Mol Cell
6:
1425-1436,
2000[ISI][Medline].
11.
Dieter, P,
Schulze-Specking A,
Karck U,
and
Decker K.
Prostaglandin release but not superoxide production by rat Kupffer cells stimulated in vitro depends on Na+/H+ exchange.
Eur J Biochem
170:
201-206,
1987[Abstract].
12.
Ethier, MF,
and
Dobson JG, Jr.
Adenosine stimulation of DNA synthesis in human ECs.
Am J Physiol Heart Circ Physiol
272:
H1470-H1479,
1997
13.
Garcia Soriano, F,
Virag L,
Jagtap P,
Szabo E,
Mabley JG,
Liaudet L,
Marton A,
Hoyt DG,
Murthy KG,
Salzman AL,
Southan GJ,
and
Szabo C.
Diabetic endothelial dysfunction: the role of poly(ADP-ribose) polymerase activation.
Nat Med
7:
108-113,
2001[ISI][Medline].
14.
Ghigo, D,
Bussolino F,
Garbarino G,
Heller R,
Turrini F,
Pescarmona G,
Cragoe EJ, Jr,
Pegoraro L,
and
Bosia A.
Role of Na+/H+ exchange in thrombin-induced platelet-activating factor production by human endothelial cells.
J Biol Chem
263:
19437-19446,
1988
15.
Haddad, JJ,
and
Land SC.
Amiloride blockades lipopolysaccharide-induced proinflammatory cytokine biosynthesis in an IB-
/NF-
B-dependent mechanism. Evidence for the amplification of an anti-inflammatory pathway in the alveolar epithelium.
Am J Respir Cell Mol Biol
26:
114-126,
2002
16.
Harguindey, S,
and
Cragoe EJ, Jr.
The Na+/H+ antiporter in oncology in the light of the spontaneous regression of cancer and cell metabolism.
Med Hypotheses
39:
229-237,
1992[ISI][Medline].
17.
Haskó, G,
Szabó C,
Németh ZH,
Kvetan V,
Pastores SM,
and
Vizi ES.
Adenosine receptor agonists differentially regulate IL-10, TNF- and nitric oxide production in RAW 264.7 macrophages and in endotoxemic mice.
J Immunol
157:
4634-4640,
1996[Abstract].
18.
Hattori, R,
Otani H,
Moriguchi Y,
Matsubara H,
Yamamura T,
Nakao Y,
Omiya H,
Osako M,
and
Imamura H.
NHE and ICAM-1 expression in hypoxic/reoxygenated coronary microvascular ECs.
Am J Physiol Heart Circ Physiol
280:
H2796-H2803,
2001
19.
Kempski, O,
and
Behmanesh S.
EC swelling and brain perfusion.
J Trauma
42:
S38-S40,
1997[ISI][Medline].
20.
Maier, RV,
and
Bulger EM.
Endothelial changes after shock and injury.
New Horiz
4:
211-223,
1996[Medline].
21.
Mastronarde, JG,
Monick MM,
Gross TJ,
and
Hunninghake GW.
Amiloride inhibits cytokine production in epithelium infected with respiratory syncytial virus.
Am J Physiol Lung Cell Mol Physiol
271:
L201-L207,
1996
22.
Matalon, S,
and
O'Brodovich H.
Sodium channels in alveolar epithelial cells: molecular characterization, biophysical properties, and physiological significance.
Annu Rev Physiol
61:
627-661,
1999[ISI][Medline].
23.
Mazzoni, MC,
Intaglietta M,
Cragoe EJ, Jr,
and
Arfors KE.
Amiloride-sensitive Na+ pathways in capillary EC swelling during hemorrhagic shock.
J Appl Physiol
73:
1467-1473,
1992
24.
Modelska, K,
Matthay MA,
McElroy MC,
and
Pittet JF.
Upregulation of alveolar liquid clearance after fluid resuscitation for hemorrhagic shock in rats.
Am J Physiol Lung Cell Mol Physiol
273:
L305-L314,
1997
25.
Németh, ZH,
Deitch EA,
Szabó C,
Fekete Z,
Hauser CJ,
and
Haskó G.
Lithium induces NF-B activation and interleukin-8 production in human intestinal epithelial cells.
J Biol Chem
277:
7713-7719,
2002
26.
Németh, ZH,
Deitch EA,
Szabó C,
and
Haskó G.
Inhibition of the Na+/H+ antiporter suppresses IL-12 production by mouse macrophages.
Biochim Biophys Acta
1539:
233-242,
2001[ISI][Medline].
27.
Nilsen, EM,
Johansen FE,
Jahnsen FL,
Lundin KEA,
Scholz T,
Brandtzaeg P,
and
Haraldsen G.
Cytokine profiles of cultured microvascular endothelial cells from the human intestine.
Gut
42:
635-642,
1998
28.
Numata, M,
and
Orlowski J.
Molecular cloning and characterization of a novel (Na+,K+)/H+ exchanger localized to the trans-Golgi network.
J Biol Chem
276:
17387-17394,
2001
29.
Ohmori, Y,
Reynolds E,
and
Hamilton TA.
Modulation of Na+/K+ exchange potentiates lipopolysaccharide-induced gene expression in murine peritoneal macrophages.
J Cell Physiol
148:
96-105,
1991[ISI][Medline].
30.
Orlinska, U,
and
Newton RC.
Modification of tumor necrosis factor- (TNF-
) production by the Na+-dependent HCO
31.
Orlowski, J,
and
Grinstein S.
NHEs of mammalian cells.
J Biol Chem
272:
22373-22376,
1997
32.
Pittet, JF,
Wiener-Kronish JP,
McElroy MC,
Folkesson HG,
and
Matthay MA.
Stimulation of lung epithelial liquid clearance by endogenous release of catecholamines in septic shock in anesthetized rats.
J Clin Invest
94:
663-671,
1994[ISI][Medline].
33.
Prpic, V,
Yu SF,
Figueiredo F,
Hollenbach PW,
Gawdi G,
Herman B,
Uhing RJ,
and
Adams DO.
Role of Na+/H+ exchange by interferon-gamma in enhanced expression of JE and I-A beta genes.
Science
244:
469-471,
1989[ISI][Medline].
34.
Rahman, A,
Anwar KN,
True AL,
and
Malik AB.
Thrombin-induced p65 homodimer binding to downstream NF-B site of the promoter mediates endothelial ICAM-1 expression and neutrophil adhesion.
J Immunol
162:
5466-5476,
1999
35.
Read, MA,
Whitley MZ,
Williams AJ,
and
Collins T.
NF-B and I
B
: an inducible regulatory system in endothelial activation.
J Exp Med
179:
503-512,
1994[Abstract].
36.
Rolfe, MW,
Kunkel SL,
Rowens B,
Standiford TJ,
Cragoe EJ, Jr,
and
Strieter RM.
Suppression of human alveolar macrophage-derived cytokines by amiloride.
Am J Respir Cell Mol Biol
6:
576-582,
1992[ISI][Medline].
37.
Rosengren, S,
Henson PM,
and
Worthen GS.
Migration-associated volume changes in neutrophils facilitate the migratory process in vitro.
Am J Physiol Cell Physiol
267:
C1623-C1632,
1994
38.
Shenolikar, S,
and
Weinman EJ.
NHERF: targeting and trafficking membrane proteins.
Am J Physiol Renal Physiol
280:
F389-F395,
2001
39.
Strieter, RM,
Kunkel SL,
Keane MP,
and
Standiford TJ.
Chemokines in lung injury: Thomas A. Neff Lecture.
Chest
116:
103S-110S,
1999
40.
Strieter, RM,
Kunkel SL,
Showell HJ,
Remick DG,
Phan SH,
Ward PA,
and
Marks RM.
Endothelial cell gene expression of a neutrophil chemotactic factor by TNF-, LPS, and IL-1
.
Science
243:
1467-1469,
1989[ISI][Medline].
41.
Szabo, EZ,
Numata M,
Shull GE,
and
Orlowski J.
Kinetic and pharmacological properties of human brain Na+/H+ exchanger isoform 5 stably expressed in Chinese hamster ovary cells.
J Biol Chem
275:
6302-6307,
2000
42.
Vairo, G,
Argyriou S,
Bordun AM,
Gonda TJ,
Cragoe EJ, Jr,
and
Hamilton JA.
Na+/H+ exchange involvement in colony-stimulating factor-1-stimulated macrophage proliferation. Evidence for a requirement during late G1 of the cell cycle but not for early growth factor responses.
J Biol Chem
265:
16929-16939,
1990
43.
Vairo, G,
Royston AK,
and
Hamilton JA.
Biochemical events accompanying macrophage activation and the inhibition of colony-stimulating factor-1-induced macrophage proliferation by tumor necrosis factor-, interferon-
, and lipopolysaccharide.
J Cell Physiol
151:
630-641,
1992[ISI][Medline].
44.
Weinman, EJ,
Minkoff C,
and
Shenolikar S.
Signal complex regulation of renal transport proteins: NHERF and regulation of NHE3 by PKA.
Am J Physiol Renal Physiol
279:
F393-F399,
2000
45.
Witko-Sarsat, V,
Allen RC,
Paulais M,
Nguyen AT,
Bessou G,
Lenoir G,
and
Descamps-Latscha B.
Disturbed myeloperoxidase-dependent activity of neutrophils in cystic fibrosis homozygotes and heterozygotes, and its correction by amiloride.
J Immunol
157:
2728-2735,
1996[Abstract].
46.
Wolff, B,
Burns AR,
Middleton J,
and
Rot A.
Endothelial cell "memory" of inflammatory stimulation: human venular endothelial cells store interleukin 8 in Weibel-Palade bodies.
J Exp Med
188:
1757-1762,
1998
47.
Yu, FH,
Shull GE,
and
Orlowski J.
Functional properties of the rat Na/H exchanger NHE-2 isoform expressed in Na/H exchanger-deficient Chinese hamster ovary cells.
J Biol Chem
268:
25536-25541,
1993
48.
Yun, CH,
Oh S,
Zizak M,
Steplock D,
Tsao S,
Tse CM,
Weinman EJ,
and
Donowitz M.
cAMP-mediated inhibition of the epithelial brush border Na+/H+ exchanger, NHE3, requires an associated regulatory protein.
Proc Natl Acad Sci USA
94:
3010-3015,
1997
49.
Yun, CH,
Tse CM,
Nath SK,
Levine SA,
Brant SR,
and
Donowitz M.
Mammalian NHE gene family: structure and function studies.
Am J Physiol Gastrointest Liver Physiol
269:
G1-G11,
1995
50.
Zen, K,
Karsan A,
Eunson T,
Yee E,
and
Harlan JM.
Lipopolysaccharide-induced NF-B activation in human ECs involves degradation of I
B
but not I
B
.
Exp Cell Res
243:
425-433,
1998[ISI][Medline].
51.
Zerbini, G,
Roth T,
Podesta F,
Cagliero E,
Doria A,
Canessa M,
and
Lorenzi M.
Activity and expression of the Na+/H+ exchanger in human endothelial cells cultured in high glucose.
Diabetologia
38:
785-791,
1995[ISI][Medline].
52.
Zhu, P,
Xiong W,
Rodgers G,
and
Qwarnstrom EE.
Regulation of interleukin 1 signalling through integrin binding and actin reorganization: disparate effects on NF-B and stress kinase pathways.
Biochem J
330:
975-981,
1998[ISI][Medline].