Vol.1, No.3, 152-158 (2009)
doi:10.4236/health.2009.13025
SciRes Copyright © 2009 Openly accessible at http://www.scirp.org/journal/HEALTH/
Health
Difference in regulation mechanisms of ENaC by
aldosterone and glucocorticords
Chengchun Tang1, Hao Zhang2, Su Wang3, Juyou Wu3, Yuchun Gu3, Jeng Wei3
1Department of Cardiology, The 1st Affiliated Hospital of Southeast University, Nanjing, China
2General Surgery, Hua Shan Hospital, China
3Department of Physiology, University of Birmingham, Edgbaston, UK; Corresponding author: y.gu@bham.ac.uk
Received 11 October 2009; revised 15 September 2009; accepted 22 September 2009.
ABSTRACT
Na+ transport occurs across many epithelial
surfaces and plays a key role in regulating salt
and water absorption. The molecular pathway
underlying this Na+ transport is the epithelial Na
channel (ENaC), which is strictly determined by a
variety of hormones like aldosterone, ADH and
glucocorticoids. In this study, we found that
stimulation of either aldosterone or dexameth-
asone (Dex) distributed ENaC channel on the
apical membrane of mouse cortical collecting
duct cells (M1). In the single channel recordings
from excised membrane, high density ENaC was
found in the cell with a dome shape by the
treatment of either dex or aldosterone. However,
low active ENaC was revealed in intact cells
treated with dex, when compared to cells treated
with aldosterone. Only 5.84% of cells treated with
dex containing ENaC exhibited ENaC current
transition in the cell-attach recording, whereas
40% of cells treated with aldosterone containing
ENaC exhibited ENaC current transition. ENaC
currents appeared rapid rundown within 5 min-
utes since formation of inside-out configuration
in cells treated with aldosterone but not with dex.
SKF-525A, a general antagonist of CYP, failed to
significantly enhance ENaC activity in intact cells
treated with dex, but EGTA, which deforming the
cells, increased the ENaC activity in the cells
treated with dex. PTX, an antagonist of G-protein,
reversed the effect of aldosterone on number of
active ENaC in intact cells. Based on our obser-
vation, we concluded that there are different
mechanisms in regulation of ENaC activity be-
tween stimulation of aldosterone and glucocor-
ticoids. The activation of G-protein is required to
maintain the activity of ENaC in the collecting
ducts.
Keywords: ENaC; Aldosterone; Glucocorticoids;
Single Channel Recording
1. INTRODUCTION
Active transport of Na+ across epithelial cells plays an
important role in maintenance of homeostasis of electro-
lytes and water in the body. Hormone sensitive Na+ re-
absorption is mainly mediated by epithelial Na+ channels
(ENaC), which are distributed in distal and collecting
nephron ducts, and alveolar epithelial cells. Disorder of
ENaC could lead to Na+ and water retention [1], ‘dry
lung’ and even death due to pulmonary edema [2]. Nev-
ertheless, ENaC activities are strictly under the control of
a variety of hormones like aldosterone, ADH, and glu-
cocorticoids.
Aldosterone is known to enhance the density of ENaC
on apical membrane of epithelial cells by modifying gene
expression. Several aldosterone sensitive genes including
serum and glucocorticoid (GC) inducible kinase (SGK-1)
[3,4], PI3-K [5] have implicated the action mechanism of
GC. Previously in vivo and in vitro studies in a number of
models show that endogenous and exogenous GC could
enhance expression of ENaC and increase the Na+ uptake
in airway [6-13], gut [14] and kidney [6,15]. The regula-
tion of ENaC by GC determines alveolar fluid clearance
and lung development [9]. However, it still remains un-
clear of the mechanism in regulation of ENaC by GC,
even by aldosterone. A slight difference in the expression
of ENaC subunits has been observed in some studies. The
expression of α subunit of ENaC is the most pronounced
in lung, correlating with the high GC level, whereas ex-
pression of α, β and γ subunits of ENaC is gradually in-
creased in kidney and colon [9]. Aldosterone is the major
regulator in kidney, although GC could also exert the
effects on ENaC expression in collecting ducts [6,15]. Till
today it is still less clear whether GC and aldosterone
exert the same effects on ENaC. In this study, we em-
ployed the electrophysiology methods to examine ENaC
activity in cells stimulated with either dexamethasone
(dex) or aldosterone.
C. C. Tang et al. / HEALTH 1 (2009) 152-158
SciRes Copyright © 2009 Openly accessible at http://www.scirp.org/journal/HEALTH/
153
153
2. MATERIALS AND METHODS
2.1. Cell Culture
M1 cells (mouse kidney cortical collecting duct cells)
were purchased from European Collection of Cell Cul-
tures at 21st passages. Cells were grown in the medium
containing DMEM: Ham’s F12 medium (1;1) (Sigma), 2
mM glutamine (Gibco), 5 µM dexamethasone (Sigma),
5% FBS (Sigma) in a 5% CO2 and 37 ºC incubator. 1.5
µM aldosterone (Sigma) was added into the culture me-
dium instead of 5 µM dexamethasone. When cells in the
culture flasks reached 70% confluence, cells were seeded
in low density to either coverslips or culture inserts (BD).
SKF-525A was added in the culture medium containing
dex 8 or 24 hours prior to experiments. Pertussis toxin
(PTX) was prepared according to previous description
[16] and manufacture instruction. Pertussis toxin was
added in the culture medium containing aldosterone in
final concentration 300 ng/ml 18 hours prior to experi-
ments.
2.2. Single Channel Patch Clamp Recording
Cell-attached single channel recording was performed as
previously described [17]. Briefly, M1 cells on the cov-
erslip or insert were transferred into the recording
chamber mounted on a Nikon inverted microscope
(Nikon TE 2000U). Patch pipettes with a tip resistance of
7 M were fabricated from borosilicate glass capillary
(1.5 od, 0.86 id) (Warner) on a Sutter Puller (P97). Bath
solutions contained (in mM): 110 NaCl, 4.5 KCl, 1 MgCl2,
1 CaCl2, 5 HEPEs, 5 Na-HEPEs, pH 7.2. Pipette solution
contained (in mM): 110 NaCl, 4.5 KCl, 0.1 EGTA, 5
HEPEs, 5 Na-HEPEs, pH 7.2. Single channel currents
were recorded with an Axon 1D amplifier and Axon
clampex 9.0. The recording was analyzed by axon
clampfit 9.0. Due to the variance of channel open prob-
ability, the first 2-5 minute single channel recording in
normal bath medium was used as the control and NPo of
ENaC during the period of applying chemicals was di-
rectly compared with NPo of the control. Data is pre-
sented as means ± S.E.M., and statistical differences were
compared using ANOVA, taking P < 0.05 as significant
and represented as *.
2.3. Chemicals
The following chemicals were used: dexamethasone(Sigma),
aldosterone (Sigma), SKF-525A (Biomol), pertussis toxin
(PTX) (Sigma). Most chemicals which dissolved in ethanol
were made up as 1000 to 5000 times stock. Preparation of
PTX was following the previous description and manufac-
ture instruction. All chemical solutions were made as re-
quired on the day of experiments. The solvent, ethanol and
DMSO at the same dilution, was tested alone in controls and
had no effect.
Figure 1. ENaC currents obtained in single channel recordings
from excised membrane. a. In an inside-out recording, small
conductance inward currents were recorded, when pipette
voltage was held at +100 mV and +60 mV, respectively. b. In an
outside-out recording, small conductance inward currents were
monitored, when pipette voltage was held at -60 mV. Bath
application of 5 µM amiloride almost abolished these currents.
The currents were reversed, when bath amiloride was washed
off. c. Currents corresponding to the voltages from inside-out
and outside-out recordings were plotted. The points were fitted
by a linear line.
Figure 2. ENaC currents obtained in cell-attach recordings. a. In
a cell-attach recording, inward currents were recorded, when
pipette voltage was held at 0 mV and +40 mV, respectively. b.
The figure represented multiple step currents recorded in cell-
attach recording. c. The conventional histogram shows the
current events against the current size. d. The I-V curve is ob-
tained from currents in cell-attach recordings.
3. RESULTS
3.1. Identification of ENaC Currents
ENaC currents were identified in either inside-out or
outside-out recording by the unique ENaC channel con-
ductance, sensitivity to amiloride and channel kinetics.
Figure 1a represented the example currents recorded in
C. C. Tang et al. / HEALTH 1 (2009) 152-158
SciRes Copyright © 2009 http://www.scirp.org/journal/HEALTH/
154
an inside-out recording. Currents were elicited when
pipette voltages (Vp) were held at +100 mV and +60 mV,
respectively. In an outside-out recording, the small con-
ductance currents possessed conductance similar to those
obtained in inside-out recording. These currents were
reversely sensitive to amiloride in the range of micro-
molar (Figure 1b). I-V curves constructed by currents
obtained in either inside-out recording (n=50) or out-
side-out recording (n=20) were almost overlapped (Fig-
ure 1c) and possessed the same conductance (5.1 pS,
fitted by linear line, r2=0.99). Figure 2a and Figure 2b
represented the currents obtained in the cell-attach re-
cordings. Currents in Figure 2b contained a multiple
transition status. I-V curve (Figure 2d) constructed by
currents in cell-attach recordings possessed the similar
conductance (5.0 pS, fitted by linear line, r2=0.99).
Therefore, we concluded that currents detected in single
channel recording in M1 cells were mediated by ENaC
channel.
Figure 3. The rundown of ENaC currents in cells treated with
either dex or aldosterone. a represented single channel currents
of ENaC in an inside-out recording within 10 minutes since
formation of inside-out configuration in a cell treated with dex.
b represented single channel currents of ENaC in an inside-out
recording in a cell treated with aldosterone. c. Summary plots of
NPo (+40 mV in a cell-attach recording) showing a sudden
rundown during the period from 3rd to 5th minute in cells treated
with aldosterone and a constant NPo in cells treated with dex.
3.2. Different ENaC Activities in M1 Cells
Treated with Either Dexmethasone (Dex)
or Aldosterone
There was no significant difference in cell morphology of
M1 cells by different treatments with either dex or al-
dosterone. Electrophysiology experiments were per-
formed, when cells formed a monolayer. As a standard
protocol, cell-attach recording was carried out for 5-10
minutes in each cell prior to single channel recording
from the excised membrane including inside-out re-
cording and outside-out recording. In cells treated with
dex, 325 cells exhibited ENaC currents in either in-
side-out or outside-out recording, but only 19 of 325 cells
possessed the detectable current transition of ENaC
channel during 5-minute of cell-attach recording (Table
1). However, in cells treated with aldosterone, 150 cells
exhibited ENaC currents in either inside-out or out-
side-out recording, and 60 of 150 cells possessed de-
tectable current transition of ENaC channel during
5-minute of cell-attach recording. Based on our observa-
tion, a large number of cells contained ENaC channels but
they did not mediate Na+ currents, when cells were in the
intact condition. About 5.84% of cells treated with dex
could mediate Na+ currents, whereas 40% of cells treated
with aldosterone could mediate Na+ currents, although
these cells contained the ENaC in apical side of mem-
brane. However, cells possessed low active ENaC
(p<0.05, in comparing with aldosterone treatment group)
on the apical membrane when cells were incubated with
aldosterone and PTX (300 ng/ml) 18 hours prior to ex-
periments.
Figure 4. The morphology of M1 cells in monolayer selected for
the patch clamping study. a, b, c, d represented the morphology
of M1 cells in monolayer. Numbers 1 to 7 represented the cell to
be studied. e and f represented the cell morphology after 10
minutes of treatment of EGTA.
Figure 5. M1 cells with certain patterns possessed high density of
ENaC currents.
3.3. Rundown of ENaC Currents in Cells
Treated with Either Dex or Aldosterone A6 [18,19] were stable for the initial 4 minutes and ex
hibited a sudden rundown during the period from the 5th
to 10th minute. In cells treated with dex, ENaC currents
Previous studies have demonstrated that ENaC currents in
Openly accessible at
C. C. Tang et al. / HEALTH 1 (2009) 152-158
SciRes Copyright © 2009 Openly accessible at http://www.scirp.org/journal/HEALTH/
155
Table 1. Active ENaC detected in different single channel configurations and cells with different treatment. In the cells treated with dex,
325 cells possessed ENaC activity in inside-out recordings but only 19 out of 325 cells possessed ENaC activity in cell-attach re-
cordings. In cells treated with aldosterone, 150 cells possessed ENaC activity in inside-out recordings and 60 out of 150 cells possessed
ENaC activity in cell-attach recordings. Deforming cells by EGTA enhanced the ENaC activity in cells treated with dex. SKF-525A did
not significantly affect ENaC activity in cells treated with dex. PTX significantly reduced the number of active ENaC in intact cells
treated with aldosterone.
Number of cells detected ENaC Dex Aldosterone Dex+EGTA Dex+SKF
525A
Aldosterone
+PTX
Inside out recording Or outside recording
(EC) 325 150 80 62 75
Cell-attach recording (CA) 19 60 15 4 1
Percentage % (CA/EC) 5.84% 40% 18.8% 6.45% 1.3%
from most cells maintained fairly constant open prob-
ability (Po) for over 10 minutes, since formation of in-
side-out configuration (Figure 3a). However, in cells
treated with aldosterone, ENaC currents were stable for
the initial 3-5 minutes and possessed a sudden rundown
from then on (Figure 3b and Figure 3c).
3.4. Enhancement of Na+ Currents in
Dex Treated Cells
Although the apical side membrane of M1 cells contained
ENaC channels, only 5.84% of cells treated with Dex
could mediate Na+ currents. SKF-525A, a general an-
tagonist of CYP, was then used to test whether the low
activity of ENaC was due to 11,12-EET [20,21]. M1 cells
were incubated with SKF-525A (60 µM) for 8 hours and
even 24 hours. Activity of ENaC was then determined in
the inside-out recording from 62 cells. About 4 out of 62
cells possessed the ENaC currents in the cell-attach re-
cording (Table 1). There was no significant difference,
when compared to cells treated with dex. Bath EGTA was
then used to stretch the membrane and rearrange the cy-
toskeleton (Figure 4e and Figure 4f), which may affect
ENaC activities. After the treatment of EGTA for 10
minutes, the cell morphology displayed change within
monolayer. The change always occurred in a patch. The
patch was in the shape of a circle. Cells in the patch lost
the flat structure and became swollen (Figure 4e) with
dome. Cells in the right side of Figure 4f were still flat
and tight, but cells on the left side were pulled by EGTA.
After the wash off EGTA, single channel recordings were
then performed. ENaC currents were detected in the in-
side-out or outside-out recordings from 80 cells. About 15
out of 80 cells possessed the ENaC currents in the
cell-attach recording.
3.5. The Linkage Between Cell Morphology
and Enac Detection
The common morphology of M1 cells from a monolayer
could be summarized into 7 shapes as shown in Figures
4a-4d. Type 1 was flat and could be found in a very flat
part of the monolayer. Types 2, 3, 5 and 6 had recogniz-
able surface domes. Type 4 also had a reasonable dome
and formed a circle with other similar shape cells. Type 7
was constituted by round cells found on the joint of flat
patches. The percentage to detect ENaC currents in Type
1 was 3% (3 out of 100), as compared to over 60% (90 out
of 150) in Type 4. In addition, before cells formed the
monolayer, there were the patterns as shown in Figure 5a
and Figure 5b with high probability to detect ENaC
currents over 60% (12 out of 20).
4. DISCUSSION
Dex and aldosterone were suggested to stimulate the
activity and expression of ENaC in renal and pulmonary
systems. In this study, we showed that both dex and al-
dosterone could allocate ENaC on the apical membrane.
However, these ENaC channels mediating Na+ entry
depended on the treatment on the cells. Only 5.84% of
them in cells treated with dex could permeate Na+ entry,
as compared to over 40% of them in cells treated with
aldosterone. ENaC in cells treated with aldosterone pos-
sessed a quick rundown. The low percentage of ENaC
permeating Na+ entry in cells treated by dex was not
related to inhibition by 11,12-EET and likely due to ac-
tivation of G-protein and how channels were anchored on
the membrane. Deformation of cytoskeleton of cells
could enhance the percentage of ENaC to permeate Na+
entry in intact cells.
In addition to the regulation on the ENaC transcription,
hormones like dex, aldosterone and epidermal growth
factor [22] exert the acute mediation on ENaC activity.
These acute effects were revealed to link with the cellular
level of PIP2. Anionic phosphilipids such as PIP2
[18,19,23] and PIP3 [24-27], located in the inner leaflet of
plasma membrane, were suggested to regulate the activity
of ENaC [26]. The negatively charged head group of PIP2
or PIP3 could directly interact with the positive charged
cytoplasmic parts in β and/or γ subunits [19,24,26,28,29]
of ENaC to exert the regulation. Evidences further indi-
C. C. Tang et al. / HEALTH 1 (2009) 152-158
SciRes Copyright © 2009 Openly accessible at http://www.scirp.org/journal/HEALTH/
156
cated that aldosterone elevates the cellular concentration
of PIP3 by activating PI-3-kinase to lock the cytoplasmic
termini of ENaC to the inner surface of plasma membrane
[3] and/or pull the channel to activate ENaC [25]. Al-
dosterone-induced protein K-Ras localizes PI3-K near
ENaC. The spatial organization of phosphatidylinositides
and specific phospholipid precursors [30,31] within cel-
lular-membrane might determine the turnover of ENaC to
allow Na+ currents [32]. Therefore, the activation of
PI3-K and spatial organization of phosphatidylinositides
on the apical membrane by aldosterone might explain the
active ENaC in the cells treated with aldosterone, but not
dex, suggesting the presence of another mechanism.
Degeneration of PIP2 by PLC [33] could lead to
unlocking of cytoplasmic termini of ENaC [25,33], re-
sulting in decreased ENaC activity. It was manifested as
the rundown of ENaC in excise membrane recording.
Rundown of ENaC currents in excise membrane re-
cordings reflected the simultaneous/continuous activation
of PLC in a platform interacting with PIP2 and ENaC.
Such platforms were revealed in G protein activated in-
ward rectifying K+ channel [34], trp channel [35] and
suggested to locate in β and γ subunits of ENaC [19].
Moreover, PLC-γ via receptor typrosine kinase [22] and
PLC-β via Gq/11 [33,36] were demonstrated to mediate the
degeneration of PIP2 in epithelial cells. However, the
effects of PIP2 on ENaC activity are suggested to be
permissive rather than regulatory [26]. Declines in PIP2
levels are parallel to the loss of ENaC activity, whereas
addition of exogenous PIP2 rarely causes ENaC open
probability to exceed the control level in excise mem-
brane recording [36]. Increase in ENaC open probability
by addition of PIP2 with GTP in inside-out recordings [19]
implicates that the activation of G-protein plays an im-
portant role in regulating ENaC activity. Decrease in
ENaC open probability by dialysis of intracellular GTP,
which is not dependent on PI-3 K [37], further supported
this hypothesis. We, therefore, proposed that activation of
G-protein in cells treated with aldosterone might be the
key to keep active ENaC on the membrane. Activation of
G-protein could activate PLC, resulting in PIP2 degen-
eration. The rapid rundown of ENaC in cells treated with
aldosterone might reflect activation of G-protein. In intact
cells, the degeneration and resynthesis of phosphatidy-
linositides reach a balance to maintain the lipid compo-
nent of the membrane structure [38,39], but the lack of
resynthesis of PIP2 pathway in excised membrane re-
cording causes the decline of PIP2, resulting in rundown
of ENaC. Like aldosterone, dex too distributes ENaC on
the apical membrane via a similar mechanism as aldos-
terone via SGK-1 [3,4,7,40,41]. It is demonstrated by the
ENaC activity detected in inside-out recordings in cells
treated with dex. The low active ENaC in intact cells
treated with dex and constant ENaC open probability in
inside-out recording might be attributed to the low activ-
ity of G-protein. The linkage between aldosterone and
activation of G-protein could be implicated by experi-
ments that prevention of ATP release due to stimulation of
aldosterone abolishes the aldosterone action [17]. Our
observation that PTX incubation significantly reverses
the effect of aldosterone on active ENaC in intact cells is
consistent to previous report [42] and further supports our
hypothesis.
11,12 - EET, a metabolite of AA via CYP epoxygenase,
was demonstrated to mediate the direct inhibition of
ENaC [20]. The evidence that EET mediates the inhibi-
tion effect of adenosine [21] on ENaC indicates a pro-
found pathway to regulate the cellular EET level. The
other rational explanation to low active ENaC in cells
treated with dex is due to presence of cellular inhibitor of
ENaC. We, therefore, applied SKF-525A, a general an-
tagonist for CYP, to reduce the cellular EET level. There
was no significant difference in the ENaC activity in
intact cells, suggesting that EET is not the case to respond
to low active ENaC.
It is widely accepted that ENaC activity is strongly
regulated by cytoskeleton elements [43-45]. Recent evi-
dence that activation of P2 receptors in epithelial baso-
lateral membrane could deform the cells and eventually
activate ENaC addressed the classic observation that
aldosterone action on Na+ reabsorption relies on ATP
production. Cell cytoskeletons could be deformed by
many methods like ATP stimulation and EGTA. Our pre-
vious work demonstrated that elevation in [Ca2+]i could
reduce ENaC open probability, but there was no signifi-
cant difference in ENaC activity, when 500 nM and 0 nM
Ca2+ were in cytoplasmic medium. EGTA was, therefore,
used to deform the cell. A significant increase in ENaC
activity in cells treated with dex demonstrated that ele-
ments of cytoskeleton regulate ENaC activity. It also
explains that ENaC activity is commonly observed in the
excised membrane recording. However, the mechanism
remains unclear. Consistent to previous observation [11],
ENaC channels are in high density and distributed in the
cells containing splitting pattern, suggesting that ENaC
might involve cell differentiation and proliferation. The
recognized dome in some cells, which contain ENaC on
apical membrane, might be formed due to Na+ entry via
ENaC with subsequent water entry.
In conclusion, aldosterone and glucocorticords could
increase the expression of ENaC and distribute ENaC on
the apical membrane of cortical collecting duct cells. Low
active ENaC is found in cells treated with dex. Activation
of G-protein is required to keep the channel active. Al-
dosterone could lead to activation of G-protein to there-
fore possess high ENaC activity in intact cells.
REFERENCES
[1] Y. Guan, C. Hao, D. R. Cha, R. Rao, W. Lu, D. E. Kohan,
M. A. Magnuson, R. Redha, Y. Zhang, M. D. Breyer,
(2005) Thiazolidinediones expand body fluid volume
C. C. Tang et al. / HEALTH 1 (2009) 152-158
SciRes Copyright © 2009 Openly accessible at http://www.scirp.org/journal/HEALTH/
157
157
through PPARgamma stimulation of ENaC-mediated re-
nal salt absorption. Nat Med, 11, 861-866.
[2] P. Factor, G. M. Mutlu, L. Chen, J. Mohameed, A. T.
Akhmedov, F. J. Meng, T. Jilling, E. R. Lewis, M. D.
Johnson, A. Xu, D. Kass, J. M. Martino, A. Bellmeyer, J.
S. Albazi, C. Emala, H. T. Lee, L. G. Dobbs, S. Matalon,
(2007) Adenosine regulation of alveolar fluid clearance.
Proc Natl Acad Sci U S A, 104, 4083-4088.
[3] J. D. Stockand, (2002) New ideas about aldosterone sig-
naling in epithelia. Am J Physiol Renal Physiol, 282,
F559-576.
[4] C. Boyd, A. Naray-Fejes-Toth, (2005) Gene regulation of
ENaC subunits by serum- and glucocorticoid-inducible
kinase-1. Am J Physiol Renal Physiol, 288, F505-512.
[5] J. Wang, P. Barbry, A. C. Maiyar, D. J. Rozansky, A.
Bhargava, M. Leong, G. L. Firestone, D. Pearce, (2001)
SGK integrates insulin and mineralocorticoid regulation
of epithelial sodium transport. Am J Physiol Renal Physiol,
280, F303-313.
[6] P. MacDonald, S. MacKenzie, L. E. Ramage, J. R. Seckl,
R. W. Brown, (2000) Corticosteroid regulation of amilo-
ride-sensitive sodium-channel subunit mRNA expression
in mouse kidney. J Endocrinol, 165, 25-37.
[7] O. A. Itani, S. D. Auerbach, R. F. Husted, K. A. Volk, S.
Ageloff, M. A. Knepper, J. B. Stokes, C. P. Thomas,
(2002a) Glucocorticoid-stimulated lung epithelial Na(+)
transport is associated with regulated ENaC and sgk1 ex-
pression. Am J Physiol Lung Cell Mol Physiol, 282,
L631-641.
[8] O. A. Itani, K. Z. Liu, K. L. Cornish, J. R. Campbell, C. P.
Thomas, (2002b) Glucocorticoids stimulate human sgk1
gene expression by activation of a GRE in its 5'-flanking
region. Am J Physiol Endocrinol Metab, 283, E971-979.
[9] K. Nakamura, J. B. Stokes, P. B. McCray Jr, (2002) En-
dogenous and exogenous glucocorticoid regulation of
ENaC mRNA expression in developing kidney and lung.
Am J Physiol Cell Physiol, 283, C762-772.
[10] S. R. Pondugula, J. D. Sanneman, P. Wangemann, P. G.
Milhaud, D. C. Marcus, (2004) Glucocorticoids stimulate
cation absorption by semicircular canal duct epithelium
via epithelial sodium channel. Am J Physiol Renal Physiol,
286, F1127-1135.
[11] V. Shlyonsky, A. Goolaerts, R. Van Beneden, S. Sari-
ban-Sohraby, (2005) Differentiation of epithelial Na+
channel function: An in vitro model. J Biol Chem, 280,
24181-24187.
[12] S. R. Pondugula, N. N. Raveendran, Z. Ergonul, Y. Deng, J
Chen, J. D. Sanneman, L. G. Palmer, D. C. Marcus, (2006)
Glucocorticoid regulation of genes in the amiloride-sensitive
sodium transport pathway by semicircular canal duct epithe-
lium of neonatal rat. Physiol Genomics, 24, 114-123.
[13] R. R. Quesnell, X. Han, B. D. Schultz, (2007) Glucocor-
ticoids stimulate ENaC upregulation in bovine mammary
epithelium. Am J Physiol Cell Physiol.
[14] M. Horster, (2000) Embryonic epithelial membrane trans-
porters. Am J Physiol Renal Physiol, 279, F982-996.
[15] M. T. Bens, V. Vallet, F. Cluzeaud, L. Pascual-Letallec, A.
Kahn, M. E. Rafestin-Oblin, B. C. Rossier, A. Vandewalle,
(1999) Corticosteroid-dependent sodium transport in a
novel immortalized mouse collecting duct principal cell
line. J Am Soc Nephrol, 10, 923-934.
[16] H. R. Kaslow, L. K. Lim, J. Moss, D. D. Lesikar, (1987)
Structure-activity analysis of the activation of pertussis
toxin. Biochemistry, 26, 123-127.
[17] J. Gorelik, Y. Zhang, D. Sanchez, A. Shevchuk, G.
Frolenkov, M. Lab, D. Klenerman, C. Edwards, Y. Kor-
chev, (2005) Aldosterone acts via an ATP autocrine/
paracrine system: the Edelman ATP hypothesis revisited.
Proc Natl Acad Sci U S A, 102, 15000-15005.
[18] H. P. Ma, S. Saxena, D. G. Warnock, (2002b) Anionic
phospholipids regulate native and expressed epithelial
sodium channel (ENaC). J Biol Chem, 277, 7641-7644.
[19] G. Yue, B. Malik, G. Yue, D. C. Eaton, (2002) Phosphati-
dylinositol 4,5-bisphosphate (PIP2) stimulates epithelial
sodium channel activity in A6 cells. J. Biol Chem, 277,
11965-11969.
[20] Y. Wei, D. H. Lin, R. Kemp, G. S. Yaddanapudi, A. Nas-
jletti, J. R. Falck, W. H. Wang, (2004) Arachidonic acid
inhibits epithelial Na channel via cytochrome P450 (CYP)
epoxygenase-dependent metabolic pathways. J Gen
Physiol, 124, 719-727.
[21] Y. Wei, P. Sun, Z. Wang, B. Yang, M. A. Carroll, W. H.
Wang, (2006) Adenosine inhibits ENaC via cytochrome
P-450 epoxygenase-dependent metabolites of arachidonic
acid. Am J Physiol Renal Physiol, 290, F1163-1168.
[22] Q. Tong, J. D. Stockand, (2005) Receptor tyrosine kinases
mediate epithelial Na(+) channel inhibition by epidermal
growth factor. Am J Physiol Renal Physiol, 288, F150-
161.
[23] D. W. Hilgemann, S. Feng, C. Nasuhoglu, (2001) The
complex and intriguing lives of PIP2 with ion channels
and transporters. Sci STKE, RE19.
[24] M. N. Helms, L. Liu, Y. Y. Liang, O. Al-Khalili, A.
Vandewalle, S. Saxena, D. C. Eaton, H. P. Ma, (2005)
Phosphatidylinositol 3,4,5-trisphosphate mediates aldos-
terone stimulation of epithelial sodium channel (ENaC)
and interacts with gamma-ENaC. J Biol Chem, 280,
40885-40891.
[25] H. P. Ma, D. C. Eaton, (2005) Acute regulation of epithe-
lial sodium channel by anionic phospholipids. J Am Soc
Nephrol, 16, 3182-3187.
[26] O. Pochynyuk, Q. Tong, J. Medina, A. Vandewalle, A.
Staruschenko, V. Bugaj, J. D. Stockand, (2007) Molecu lar
determinants of PI(4,5)P2 and PI(3,4,5)P3 regulation of
the epithelial Na+ channel. J Gen Physiol, 130, 399-413.
[27] K. M. Weixel, R. S. Edinger, L. Kester, C. J. Guerriero, H.
Wang, L. Fang, T. R. Kleyman, P. A. Welling, O. A. Weisz,
J. P. Johnson, (2007) Phosphatidylinositol 4-phosphate
5-kinase reduces cell surface expression of the epithelial
sodium channel (ENaC) in cultured collecting duct cells. J.
Biol Chem.
[28] Q. Tong, N. Gamper, J. L. Medina, M. S. Shapiro, J. D.
Stockand, (2004b) Direct activation of the epithelial Na(+)
channel by phosphatidylinositol 3,4,5-trisphosphate and
phosphatidylinositol 3,4-bisphosphate produced by phos-
phoinositide 3-OH kinase. J Biol Chem, 279, 22654-
22663.
[29] O. Pochynyuk, Q. Tong, J. Medina, A. Vandewalle, A.
Staruschenko, V. Bugaj, J. D. Stockand, (2007) Molecu lar
determinants of PI(4,5)P2 and PI(3,4,5)P3 regulation of
the epithelial Na+ channel. J Gen Physiol, 130, 399-413.
[30] R. L. Doughman, A. J Firestone, R. A. Anderson, (2003)
Phosphatidylinositol phosphate kinases put PI4,5P(2) in
its place. J Membr Biol, 194, 77-89.
C. C. Tang et al. / HEALTH 1 (2009) 152-158
SciRes Copyright © 2009 http://www.scirp.org/journal/HEALTH/Openly accessible at
158
[31] P. A. Oude Weernink, M. Schmidt, K. H. Jakobs, (2004)
Regulation and cellular roles of phosphoinositide 5-
kinases. Eur J Pharmacol, 500, 87-99.
[32] A. Staruschenko, O. M. Pochynyuk, Q. Tong, J. D.
Stockand, (2005) Ras couples phosphoinositide 3-OH
kinase to the epithelial Na+ channel. Biochim Biophys
Acta, 1669, 108-115.
[33] K. Kunzelmann, T. Bachhuber, R. Regeer, D. Markovich,
J. Sun, R. Schreiber, (2005) Purinergic inhibition of the
epithelial Na+ transport via hydrolysis of PIP2. Faseb J, 19,
142-143.
[34] T. Rohacs, J. Chen, G. D. Prestwich, D. E. Logothetis,
(1999) Distinct specificities of inwardly rectifying K(+)
channels for phosphoinositides. J Biol Chem, 274, 36065-
36072.
[35] R. C. Hardie, (2003a) Regulation of TRP channels via
lipid second messengers. Annu Rev Physiol, 65, 735-759.
[36] H. P. Ma, L. Li, Z. H. Zhou, D. C. Eaton, D. G. Warnock,
(2002a) ATP masks stretch activation of epithelial sodium
channels in A6 distal nephron cells. Am J Physiol Renal
Physiol, 282, F501-505.
[37] O. Pochynyuk, A. Staruschenko, Q. Tong, J. Medina, J. D.
Stockand, (2005) Identification of a functional phos-
phatidylinositol 3,4,5-trisphosphate binding site in the
epithelial Na+ channel. J Biol Chem, 280, 37565-37571.
[38] R. C. Hardie, (2003b) TRP channels in Drosophila photore-
ceptors: the lipid connection. Cell Calcium, 33, 385- 393.
[39] R. C. Hardie, (2004) Regulation of Drosophila TRP
channels by lipid messengers. Novartis Found Symp, 258,
160-167; discussion 167-171, 263-166.
[40] D. Pearce, (2003) SGK1 regulation of epithelial sodium
transport. Cell Physiol Biochem, 13, 13-20.
[41] Q. Tong, R. E. Booth, R. T. Worrell, J. D. Stockand,
(2004a) Regulation of Na+ transport by aldosterone: sig-
naling convergence and cross talk between the PI3-K and
MAPK1/2 cascades. Am J Physiol Renal Physiol, 286,
F1232-1238.
[42] L. Gambling, R. E. Olver, G. K. Fyfe, P. J. Kemp, D. L.
Baines, (1998) Differential regulation of Na+ and Cl-
conductances by PTX-sensitive G proteins in fetal lung
apical membrane vesicles. Biochim Biophys Acta, 1372,
187-197.
[43] B. K. Berdiev, A. G. Prat, H. F. Cantiello, D. A. Ausiello,
C. M. Fuller, B. Jovov, D. J Benos, I. I. Ismailov, (1996)
Regulation of epithelial sodium channels by short actin
filaments. J Biol Chem, 271, 17704-17710.
[44] J. B. Zuckerman, X. Chen, J. D. Jacobs, B. Hu, T. R.
Kleyman, P. R. Smith, (1999) Association of the epithelial
sodium channel with Apx and alpha-spectrin in A6 renal
epithelial cells. J Biol Chem, 274, 23286-23295.
[45] C. Mazzochi, J. K. Bubien, P. R. Smith, D. J. Benos, (2006)
The carboxyl terminus of the alpha-subunit of the amilo-
ride-sensitive epithelial sodium channel binds to F-actin. J
Biol Chem, 281, 6528-6538.