ArticlePDF AvailableLiterature Review

Intracellular Monovalent Ions as Second Messengers

Authors:

Abstract

It is generally accepted that electrochemical gradients of monovalent ions across the plasma membrane, created by the coupled function of pumps, carriers and channels, are involved in the maintenance of resting and action membrane potential, cell volume adjustment, intracellular Ca2+ handling and accumulation of glucose, amino acids, nucleotides and other precursors of macromolecular synthesis. In the present review, we summarize data showing that side-by-side with these classic functions, modulation of the intracellular concentration of monovalent ions in a physiologically reasonable range is sufficient to trigger numerous cellular responses, including changes in enzyme activity, gene expression, protein synthesis, cell proliferation and death. Importantly, the engagement of monovalent ions in regulation of the above-listed cellular responses occurs at steps upstream of Ca2+i and other key intermediates of intracellular signaling, which allows them to be considered as second messengers. With the exception of HCO 3−-sensitive soluble adenylyl cyclase, the molecular origin of sensors involved in the function of monovalent ions as second messengers remains unknown.
Intracellular Monovalent Ions as Second Messengers
S.N. Orlov
1,2
, P. Hamet
1
1
Centre de recherche, Centre hospitalier de lÕUniversite
´de Montre
´al, (CHUM)-Hoˆ tel-Dieu, Montreal, Quebec, Canada
2
Laboratory of Pathophysiology of Ion Transport Disorders, Centre de recherche, CHUM - Hoˆ tel-Dieu, 3850 rue St-Urbain, Montreal,
Quebec H2W 1T7, Canada
Received: 13 November 2005/Revised: 8 February 2006
Abstract. It is generally accepted that electrochemical
gradients of monovalent ions across the plasma
membrane, created by the coupled function of
pumps, carriers and channels, are involved in the
maintenance of resting and action membrane poten-
tial, cell volume adjustment, intracellular Ca
2+
han-
dling and accumulation of glucose, amino acids,
nucleotides and other precursors of macromolecular
synthesis. In the present review, we summarize data
showing that side-by-side with these classic functions,
modulation of the intracellular concentration of
monovalent ions in a physiologically reasonable
range is sufficient to trigger numerous cellular re-
sponses, including changes in enzyme activity, gene
expression, protein synthesis, cell proliferation and
death. Importantly, the engagement of monovalent
ions in regulation of the above-listed cellular re-
sponses occurs at steps upstream of Ca
2+
i
and other
key intermediates of intracellular signaling, which
allows them to be considered as second messengers.
With the exception of HCO
3
)
-sensitive soluble ade-
nylyl cyclase, the molecular origin of sensors involved
in the function of monovalent ions as second
messengers remains unknown.
Key words: Sodium Potassium Proton
Bicarbonate Chloride Intracellular signalling
Introduction
In accordance with Earl SutherlandÕs signal trans-
duction hypothesis [113], any intracellular molecule
can be considered a potential second messenger in the
signal transduction pathway if it satisfies 3 major
criteria. i) Modulation of intracellular concentration
of the potential second messenger triggered by
external stimuli precedes cellular responses and nor-
malizes after removal of these stimuli. ii) The tran-
sient modulation of intracellular second messenger
concentration per se is sufficient to trigger cellular
responses in the absence of investigated external
stimuli. iii) Cellular responses triggered by external
stimuli are mediated by the interaction of second
messengers with their intracellular sensors.
In pioneering studies performed more than
40 years ago, it was shown that cAMP satisfies the
above-listed criteria and provides coupling between
excitation of plasma membrane receptors by cate-
cholamines and peptide hormones with gluconeo-
genesis and lipolysis in hepatocytes and adipocytes,
respectively [105]. Later on, the list of second mes-
sengers was broadened in experiments demonstrating
a key role for cGMP, Ca
2+
and lipid molecules, such
as diacyl glycerol, inositol 1,4,5-triphosphate etc., in
the regulation of other cellular functions, such as
myocyte contraction and relaxation, hormone and
neurotransmitter release, light sensing, cell
proliferation and apoptosis, etc. [10, 18, 63, 119].
Similarly to the above-listed second messengers,
the intracellular concentration of monovalent cations
is transiently affected by diverse extracellular stimuli
and normalized to baseline values via feedback acti-
vation of the pumps, carriers and channels shown in
Fig 1. Thus, transient activation of Na
+
/H
+
ex-
change and [Na
+
]
i
elevation appear to be a universal
response of quiescent cells to growth-promoting
stimuli [15, 39, 44, 70, 90, 101, 125]. In neurons, short
periods of synaptic activity produce large increases of
[Na
+
]
i
, from 10 to 30 and 100 mMin apical den-
drites and dendritic spines, respectively, mainly due
to Na
+
influx via N-methyl-D-aspartate (NMDA)
receptor channels [106]. In erythrocytes and other
cells with low resting potential, transient activation of
Correspondence to: S.N. Orlov; email: sergei.n.orlov@umontreal.
ca
J. Membrane Biol. 210: 161–172 (2006)
DOI: 10.1007/s00232-006-0857-9
Topical Review
K
+
channels results in 5- to 8-fold attenuation of
[K
+
]
i
[9]. The regulation of these and other mono-
valent ion transporters by extracellular stimuli is
considered in detail in several comprehensive reviews
[1, 31, 33, 36, 45, 108, 125]. Here, we just summarize
data showing that modulation of the intracellular
concentration of monovalent ions triggered by
external stimuli is sufficient to affect cellular re-
sponses by acting upstream or even independently of
the signaling pathways evoked by well-defined second
messengers. We would like also to underline that the
goal of our mini-review is to support the concept of
monovalent ions as second messengers, to consider
the possible pathophysiological implications of these
signaling cascades, and to provoke the search for
intracellular monovalent ion sensors rather than to
provide a complete list of monovalent ion-dependent
cellular functions documented so far.
Sodium
In cells abundant with Na
+
/Ca
2+
exchanger, eleva-
tion of [Na
+
]
i
is sufficient to activate this carrier and
to elicit diverse Ca
2+i
-mediated responses, including
positive inotropic effect in cardiomyocytes and neu-
rotransmitter release in nerve terminals treated with
low doses of Na
+
/K
+
-ATPase inhibitors, such as
ouabain and other cardiotonic steroids (CTS) [11].
Data obtained in these studies should probably be
considered as first evidence for the involvement of
Na
+i
in the regulation of cellular function as a sec-
ond messenger. This section is focused on data
showing that Na
+i
can modulate cellular function
independently of [Ca
2+
]
i
elevation and of activation
of other Na
+
-coupled ion carriers.
In the late 1990s, we observed that almost com-
plete Na
+
,K
+
pump inhibition with ouabain pro-
tects rat vascular smooth muscle cells (VSMC) from
apoptosis triggered by growth factor withdrawal,
staurosporin or inhibitors of serine-threonine phos-
phatases and potentiated by transfection with c-myc
or its functional analogue E1A adenoviral protein
[86]. Suppression of apoptosis in CTS-treated VSMC
can be mediated by membrane depolarization, accu-
mulation of Na
+
or loss of K
+
. Conformational
transition of the Na
+
/K
+
-ATPase a-subunit trig-
gered by interaction with CTS may be sufficient per se
to generate the antiapoptotic signal. In addition, CTS
interaction with target(s) distinct from the Na
+
/K
+
-
ATPase a-subunit can not be excluded. We found
that apoptosis in VSMC is sharply suppressed by
incubation of VSMC in K
+
-free medium. Keeping in
mind an obligatory role of K
+o
in the activation of
Na
+
/K
+
-ATPase, this observation strongly suggests
the antiapoptotic action of ouabain is mediated by
inhibition of the Na
+
/K
+
pump. To further examine
the role of monovalent cations, we treated VSMC
with ouabain in high-K
+
medium. Sustained depo-
larization in high-K
+
, low-Na
+
medium did not
affect apoptosis. In contrast, dissipation of the
transmembrane gradient of monovalent cations
occurring in this medium sharply diminished the
effect of ouabain on Na
+i
and K
+i
content and
completely abolished its antiapoptotic action [86].
These data led us to conclude that Na
+
/K
+
pump
inhibition protects VSMC against apoptosis via ele-
vation of the [Na
+
]
i
/[K
+
]
i
ratio.
Later on, the antiapoptotic action of ouabain and
K
+
-depleted medium was detected in a cultured renal
proximal tubule cell line [131], in freshly-isolated rat
cerebellar granule cells [49] and in human umbilicial
vein endothelial cells [120]. It should be underlined
that relatively low concentrations of ouabain were
used in these studies, and its action on the [Na
+
]
i
/
[K
+
]
i
ratio was not examined. More recently, we
[Na+]o~140 mM; [K+]o~4 mM; [Cl-]o~100 mM; [HCO3-]o~20 mM; [H+]o~60 nM
out
in 123 567 8 9 104
3Na+
ATP ADP
2K+
K+K+
Cl-
Cl-
Cl-
Na+
H+HCO3-
Na+K+
H+
ATP ADP
2(1)HCO3-
CO2+H2O
CA
Na+Na+
2Cl-
K+
[Na+]i~10 mM; [K+]i~150 mM; [Cl-]o~20 mM; [HCO3-]i~30 mM; [H+]i~80 nM
a) b) c)
Fig. 1. Major plasma membrane ion transporters involved in intracellular Na
+
/K
+
(a), Cl
)
(b) and H
+
/HCO
3
)
(c) handling. 1: Na
+
/K
+
-
ATPase; 2:Na
+
channels; 3: K
+
channels; 4:Na
+
,K
+
, 2Cl
)
cotransport; 5:K
+
,Cl
)
cotransport; 6:Cl
)
channels; 7: Na
+
/H
+
exchange;
8: anion exchanger; 9:Na
+
,NaHCO
3
)
cotransport; 10: H
+
/K
+
-ATPase; CA: carbonic anhydrase. The representative values of extra- and
intracellular concentrations of monovalent ions are shown.
162 S.N. Orlov and P. Hamet: Monovalent Ions as Second Messengers
noted that K
+
-free, Na
+
-containing medium rescues
vascular endothelial cells from apoptosis triggered by
[
3
H]-decay-induced DNA damage. Because this pro-
tection was absent in K
+
-free, low-Na
+
medium, we
concluded that the antiapoptotic signal triggered by
Na
+
/K
+
pump inhibition is mediated by [Na
+
]
i
elevation rather than by loss of K
+i
[87].
To further explore the novel Na
+i
-mediated
antiapoptotic pathway, we treated cells with actino-
mycin D or cycloheximide. Both inhibitors of mac-
romolecular synthesis abolished protection against
apoptosis documented in VSMC pretreated with
ouabain [84]. Deploying a rat multi-probe template
set, we failed to detect, in ouabain-treated VSMC,
elevation of mRNA species encoding major pro- and
antiapoptotic proteins such as Bcl-2, Bcl-xL, Bcl-xS,
Bax, and caspases 1-3 [82]. With these negative data
in mind, we took a proteomics approach to identify a
set of Na
+i
-sensitive genes. Twelve soluble proteins,
including mortalin, whose expression is triggered by
ouabain, were identified by mass spectrometry [117].
Previous studies demonstrated the pancytosolic and
mitochondrial/juxtanuclear localization of mortalin
in mortal and immortal cells, respectively [114, 122–
124]. Northern and Western blotting confirmed the
induction of mortalin expression in ouabain-treated
VSMC and documented its mitochondrial localiza-
tion. We established that, similarly to ouabain,
transfection with mortalin delayed the development
of apoptosis in serum-deprived VSMC-E1A. We also
found that transfection with mortalin inhibits
p53 translocation to the nucleus [117]. Viewed
collectively, these data suggest that elevated [Na
+
]
i
suppresses programmed cell death via augmented
mortalin expression that, in turn, blocks p53
nuclear translocation triggered by apoptotic stimuli
(Fig. 2).
In the last decade, it was found that, in several
types of cells, sustained inhibition of the Na
+
/K
+
pump triggers the expression of the a1- and b1-su-
bunits of the Na
+
/K
+
-ATPase, myosin light chain,
skeletal muscle actin, atrial natriuretic factor and
tumor growth factor-b(for recent review, see [116,
128]). These data together with augmented RNA
synthesis [85] and the appearance of numerous
protein spots, detected in ouabain-treated VSMC by
2-D electrophoresis [82, 117], suggest that this action
of Na
+
,K
+
pump inhibitors is at least partially
mediated by early response genes. Indeed, in VSMC,
we observed 10- and 4-fold elevations of immuno-
reactive c-Fos and c-Jun after 2- and 12-h treatment
with ouabain, respectively [115]. A 4-fold increment
of c-Fos mRNA content was detected in 30 min of
ouabain addition. Importantly, within this time
interval, [Na
+
]
i
was increased by 5-fold whereas
[K
+
]
i
was decreased by only 10–15%. This result
shows that [Na
+
]
i
augmentation rather than [K
+
]
i
attenuation generates a signal that leads to c-Fos
expression.
In accordance with known signaling pathways
triggered by CTS, gene expression, seen under
elevated [Na
+
]
i
, can be mediated by cell
volume modulation or/and activation of Na
+
/H
+
and Na
+
/Ca
2+
exchangers. The latter hypothesis is
also consistent with the presence of (Ca
2+
+ cAMP)
response element (CRE) within the c-Fos promoter
[107]. Indeed, we have demonstrated that K
+o
-in-
duced depolarization leads to c-Fos expression that is
completely abolished by the selective L-type Ca
2+
channel blocker nicardipine [115]. However, the data
listed below strongly indicate that c-Fos expression in
ouabain-treated VSMC is a Ca
2+
-independent phe-
nomenon. First, c-Fos expression in ouabain-treated
cells is not sensitive to nicardipine. Second, neither
ATP
3Na+
PROTEIN SYNTHESIS
[Na+]i
ADP
[K+]i
2K+
12
CTS
eEF-2 eEF-2~P
eEF-2 kinase
SK
?
pHRE 3
Apoptotic
stimuli
AP Gαβγ GαGβγ
+
ONCOSIS
NaRE mortalin
APOPTOSIS
p53
SNa
[Na+]i
[K+]i
?
SpH
caspase-3
Gp-mediated
responses
[H+]i
Fig. 2. Intracellular monovalent cations as second messengers. 1and 2: active and inactive conformation of the Na
+
/K
+
-ATPase a-subunit
triggered by its interaction with CTS; 3: H
+i
-sensitive gene(s) involved in the inhibition of oncosis; S
K
,S
Na
and S
pH
hypothetical sensors
of intracellular K
+
,Na
+
and H
+
, respectively; AP: adapter protein(s) whose interaction with Na
+
,K
+
-ATPase a-subunit is affected by
CTS; pHRE and NaRE:H
+
- and Na
+
-sensitive elements implicated in the regulation of gene expression; eEF-2: elongation factor-2; Gabc::
a,band csubunits of GTP-binding proteins; ?: unknown intermediates; - and --| : activatory and inhibitory signals, respectively. For
more details, see text.
S.N. Orlov and P. Hamet: Monovalent Ions as Second Messengers 163
[Ca
2+
]
i
nor total exchangeable Ca content in VSMC is
affected by ouabain [115]. This observation is consis-
tent with negligible Na
+
/Ca
2+
exchanger activity
detected in VSMC [83]. Third, neither extracellular
(EGTA) nor intracellular (BAPTA-AM) Ca
2+
che-
lators abolish ouabain-induced c-Fos expression
[115]. These data as well as the lack of a significant
effect of ouabain on pH
i
and cell shrinkage on c-Fos
content allow us to hypothesize that gene expression
in ouabain-treated VSMC is mediated by a novel
Na
+i
-dependent, Ca
2+i
-insensitive mechanism of
excitation-transcription coupling.
Gene expression is probably not the only cellular
function controlled by the Na
+i
-sensor. Indeed, it has
been shown in neuronal cells that elevation of [Na
+
]
i
is sufficient to activate heterotrimeric G-proteins via
the GTP-independent mechanism of dissociation of
their a- and bc-subunits [104]. Moreover, in these
cells, [Na
+
]
i
elevation modulates the activity
of NMDA receptors, K
+
and Ca
2+
channels by
G-protein-dependent and -independent mechanisms
[12, 104, 130]. Are these cellular responses and Na
+i
-
dependent gene expression, demonstrated in our
studies [115], mediated by the same Na
+i
-sensor? We
will address this question in future studies.
Potassium
More than 40 years ago, it was demonstrated that
protein synthesis in prokaryotes is sharply inhibited
in the absence of K
+
[65]. Later on, the requirement
of K
+
for protein synthesis was detected in animal
cells of different origins [56, 59, 64, 73, 100]. Using
human fibroblasts subjected to sustained inhibition of
Na
+
/K
+
-ATPase with ouabain, it was shown that
inversion of the [Na
+
]
i
/[K
+
]
i
ratio inhibits protein
synthesis without any impact on mRNA function,
ATP content and amino acid transport [59], thus
suggesting direct influence of [K
+
]
i
on the protein
synthesis machinery.
In reticulocytes, globin contributes to more than
90%of total protein synthesis. In these cells, it was
found that K
+i
depletion inhibits the elongation step
of globin synthesis without any impact on ribosome
subunit assembly [16]. The half-maximal activation of
globin synthesis by reticulocyte lysate in medium
containing 60, 90 and 125 mMNa
+
was observed at
[K
+
] of 15, 25 and 40 mM, respectively [16]. These
data indicate that elevation of [Na
+
]
i
diminishes the
efficacy of protein synthesis regulation by K
+i
via
attenuation of K
+
interaction with its hypothetical
sensor (Fig. 2). Intermediates of the protein synthesis
machinery involved in K
+i
sensing remain unknown.
It should be underlined that the effect of K
+i
loss
on protein synthesis is cell type-specific. Thus, in
contrast to the above-mentioned cells showing 2- to
4-fold attenuation of protein synthesis after sustained
inhibition of Na
+
/K
+
-ATPase in K
+
-free medium
or in the presence of CTS, we did not see any sig-
nificant effect on [
3
H]-leucine protein labelling after
24-h ouabain treatment of cultured VSMC from the
rat aorta [85]. Two hypotheses could explain these
data. First, the K
+i
-sensitive element of the protein
synthesis machinery is absent in VSMC. Second,
attenuation of protein synthesis in K
+
-depleted
VSMC is masked by augmented mRNA synthesis.
Indeed, we discerned a 6-fold elevation of total RNA
synthesis in VSMC treated with ouabain for 10 h [85]
that could be attributed to Na
+i
-mediated expression
of c-Fos and other early response genes detected in
VSMC after 1–2 h of ouabain addition [115].
Proton
The functioning of numerous proteins is affected by
cellular acidification. Thus, for example, acidification
inhibits TASK-3 K
+
channels [102] but activates
TREK-1 K
+
channels [67] and Ca
2+
-permeable acid-
sensitive ion channels [129]. It should be underlined,
however, that modulated activity of these ion trans-
porters was detected in pH ranges from 7.4 to 5.0,
which corresponds to 200-fold elevation of [H
+
]
i
concentration, thus suggesting the involvement of
these proteins in pH sensing under severe hypoxia
and/or HCO
3
)
depletion. A system with much higher
pH sensitivity has been detected in our studies of
oncosis in CTS-treated epithelial and endothelial cells.
In contrast to rat VSMC [85, 86], NIH 3T3
mouse fibroblasts, HEK-293, HeLa, human renal
carcinoma Caki cells [81], renal epithelial cells from
the Rhesus monkey [22, 23], human lymphocytes [32]
and rat astrocytes (unpublished data), 24-h exposure
to ouabain results in massive death of renal epithelial
cells from Madin-Darby canine kidney (MDCK) and
endothelial cells from the porcine aorta (PAEC).
Both types of ouabain-treated cells possess combined
markers of necrosis (cell swelling, negligible labelling
with nucleotides in the presence of terminal trans-
ferase, nuclei staining with cell-impermeable dyes,
such as propidium iodide) and apoptosis (nuclear
condensation, chromatin cleavage, caspase-3 activa-
tion) [23, 87, 96]. We also demonstrated that in
contrast to classical cell culture models of apoptosis,
death of ouabain-treated MDCK cells was insensitive
to the pan-caspase inhibitor z-VAD.fmk [96]. To
underline the striking difference in cell volume
behavior (swelling vs shrinkage detected in cells
undergoing apoptosis), revised terminology has been
proposed, claiming that necrosis was originally of-
fered as a concept to characterize any post-mortem
changes in cell morphology. In accordance with this
nomenclature, the label ‘‘oncosis’’, derived from the
Greek word for swelling, describes cell death that is
distinct from apoptotic shrinkage [68].
164 S.N. Orlov and P. Hamet: Monovalent Ions as Second Messengers
Surprisingly, we found that more than 500-fold
inhibition of the Na
+
/K
+
pump in K
+
-free medium
does not affect the survival of C7-MDCK cells [96].
As predicted, 6-h incubation of C7-MDCK cells in
K
+
-free medium led to a sharp [Na
+
]
i
elevation, and
the addition of ouabain only slightly altered this
parameter, whereas incubation in high-K
+
/low-Na
+
medium did not impact the baseline values of [Na
+
]
i
and [K
+
]
i
, but completely abolished the K
+i
loss
triggered by ouabain. However, similarly to control
medium, ouabain killed cells to the same extent in
K
+
-free and high-K
+
/low-Na
+
media [96]. More-
over, the same left-hand shift was noted in compari-
son to the dose-dependent action of ouabain on Na
+
/
K
+
pump activity and death of C7-MDCK and
PAEC [4, 87, 96]. These results strongly indicate that
in both cell types, CTS trigger Na
+i
,K
+i
-indepen-
dent oncosis via interaction with the Na
+
/K
+
-AT-
Pase a-subunit rather than with other potential K
+o
-
insensitive receptors. Considering these data, we
proposed that CTS-induced conformational transi-
tion of the Na
+
/K
+
-ATPase a-subunit is sufficient to
trigger its interaction with an unidentified adapter
protein(s), resulting in Na
+i
,K
+i
-independent
oncosis of renal epithelial and vascular endothelial
cells (Fig. 2). This adapter protein or downstream
intermediates of the Na
+i
,K
+i
-independent death
machinery are absent in CTS-resistant cells, including
VSMC.
Under analysis of the role of extracellular ions
in CTS-induced oncosis, we observed that decreases
of NaHCO
3
concentration from 44 to 11 mMshar-
ply attenuated the death of C7-MDCK cells trig-
gered by ouabain. Keeping in mind that total Na
+
concentration in control and NaHCO
3
-depleted
medium was the same, 2 hypotheses can explain this
finding. First, medium acidification caused by a
decreased HCO
3
)
/CO
2
ratio is sufficient to inhibit the
cell death machinery. Second, a decreased HCO
3
)
/
CO
2
ratio suppresses cell death independently of
medium acidification. Data obtained in additional
experiments do not support the latter hypothesis.
Indeed, cell death inhibition was also detected in
medium with high NaHCO
3
concentration and
acidified by HEPES, whereas alkalinization with
Tris abolished the protective action of NaHCO
3
depletion [5]. Finally, we used NaHCO
3
-free, HE-
PES-Tris-buffered medium and observed that the
death of ouabain-treated PAEC and C7-MDCK
cells is suppressed by acidification of the medium
from pH 7.4 to 7.0, i.e., under conditions when pH
i
was decreased from 7.2 to 6.9. The rescue of
ouabain-treated C7-MDCK cells was also detected
under selective intracellular acidification caused by
inhibition of the Na
+
/H
+
exchanger with ethyliso-
propyl amiloride [5].
Neither [
3
H]-ouabain binding nor ouabain-sen-
sitive
86
Rb influx was significantly affected by modest
acidification [5], showing that the H
+i
-sensitive ele-
ment of the cell death machinery is located down-
stream of Na
+
/K
+
-ATPase. It should be noted that
acidification from 7.2 to 5.0 activates rather than
inhibits caspases [71] and nucleases [97], excluding
these downstream intermediates as a potential H
+i
-
sensor involved in the suppression of death signaling
triggered by CTS.
Elongation factor-2 (eEF-2) is the most promi-
nently phosphorylated protein detected in mamma-
lian tissue extracts, and its phosphorylation by eEF-2
kinase leads to inactivation and inhibition of protein
synthesis [110]. By comparing liver homogenates
from wild-type and eEF-2 kinase knockout mice, it
was shown that eEF-2 phosphorylation is completely
blocked by pH elevation from 6.6 to 7.4 [30, 109], i.e.,
in the range where switch off/on regulation of the
CTS-induced cell death machinery is detected. Con-
sidering this, it may be proposed that acidosis sup-
presses the death signal via eEF-2 phosphorylation
that in turn abolishes eEF-2-mediated activation of
protein synthesis. However, the death of ouabain-
treated MDCK cells was noted in the presence of
RNA and protein synthesis inhibitors, whereas the
protective effect of acidification was sharply dimin-
ished by these compounds at modest non-toxic con-
centrations [5]. These results strongly suggest that the
rescue by modest intracellular acidification of renal
epithelial and vascular endothelial cells from Na
+i
,
K
+i
-independent oncosis triggered by CTS is medi-
ated by the de novo expression of gene(s) containing
pH
i
-response element.
Bicarbonate
Mammalian spermatozoa undergo activation pro-
cesses induced by bicarbonate and mediated by ele-
vation of intracellular cAMP content. It has been
assumed that this action of HCO
3
)
is caused by
alkalinization of the cytoplasm. However, several
laboratories have reported that spermatozoa are
highly abundant in soluble adenylyl cyclase (sAC)
[19, 37, 79] stimulated by HCO
3
)
in a pH
i
-independent
manner [19]. It was also shown that bicarbonate
activates purified and recombinant sAC with an EC
50
of 20 mM[19] relevant to the physiological range of
[HCO
3
)
]
i
(Fig. 1c). Amino acid residues involved in
the organization of the HCO
3
)
-binding site of sAC
and the role of these sites in the conformational
transition of this enzyme remain unknown.
Side-by-side with spermatozoa, sAC was also
detected in the kidney and choroid plexus [19, 95],
indicating involvement of HCO
3
)
as a second mes-
senger in the regulation of cAMP-dependent func-
tions of these tissues. Based on the identification of
sAC within nuclei, it has been proposed that this
enzyme contributes to [H
+
]
i
/[HCO
3
)
]
i
-dependent
S.N. Orlov and P. Hamet: Monovalent Ions as Second Messengers 165
modulation of gene expression via activation of CRE-
binding protein [132]. It was also suggested that pH
i
-
coupled modulation of sAC activity is responsible for
pH-dependent recycling of vacuolar H
+
-ATPase [95].
Chloride
The first data suggesting the role of Cl
)
i
in cellu-
lar signaling were probably obtained in the study
of regulation of the ubiquitous isoform of Na
+
,
K
+
, 2Cl
)
cotransporter (NKCC1). Several research
groups demonstrated that inwardly-directed
NKCC1 contributes to the accumulation of Cl
)
above the Nernst equilibrium potential. For exam-
ple, in secretory epithelial, renal epithelial, vascular
endothelial, mesangial and neuronal cells, inhibition
of NKCC1 with bumetanide decreases [Cl
)
]
i
by 2- to
6-fold [52, 69, 77]. In resting neuronal cells with high
permeability for K
+
(P
K
>> P
Cl
), NKCC1-medi-
ated elevation of [Cl
)
]
i
does not affect membrane
potential (E
m
) but leads to transient depolarization
under activation of GABA-sensitive anion channels
[29]. In VSMC, P
K
and P
Cl
values are somewhat
similar [21], indicating NKCC1 involvement in the
regulation of resting E
m
. Indeed, bumetanide de-
creases [Cl
)
]
i
[89], hyperpolarizes [28] and abolishes
differences in [Cl
)
]
i
and E
m
between VSMC from
normotensive and deoxycorticosterone-salt-hyper-
tensive rats [14]. In recent studies, we demonstrated
that in HCO
3
)
-depleted medium, NKCC1 inhibition
sharply suppresses smooth muscle cell contrac-
tion triggered by modest depolarization or by
a-adrenergic stimulation [7, 55].
Since the seminal studies of dialyzed squid axons
[13], it was shown that in all types of cells studied so
far NKCC1 activity is decreased by 5- to 10-fold
under elevation of [Cl
)
]
i
from 20 to 150 mM(108),
thus providing feedback regulation of this carrier. In
Cl
)
-depleted tracheal epithelial cells [42] and shark
rectal glands [66], NKCC1 activation is accompanied
by phosphorylation of the carrier, indicating the
presence of protein kinases whose activity is nega-
tively regulated by [Cl
)
]
i
. The presence of Cl
)
i
-sensi-
tive intermediates of signal transduction, including
protein kinases and phosphatases, was also proposed
in the study of activation of permeabilized neu-
trophils [38], GABA receptors [58], and phosphory-
lation of membrane-bound proteins in airway
epithelium, including nucleoside diphosphate kinase
[75, 118]. In contrast, we failed to detect any impact
of Cl
)
i
depletion on baseline phosphorylation of
proteins in MDCK cells or on protein phosphoryla-
tion triggered by activators of protein kinases A, C
and mitogen-activated protein kinases (unpublished
data).
Several laboratories have reported that growth
factors and other mitogens transiently activate
NKCC1 [91, 92], whereas chronic inhibition of the
carrier with bumetanide or furosemide suppresses the
growth of fibroblasts [91], VSMC [17], lymphocytes
[92], vascular endothelial cells [93] and airway smooth
muscle cells [51]. More recently, Panet and co-work-
ers reported that NKCC1 overexpression sharply
accelerates the proliferation of mouse Balb/c 3T3
cells in serum-free medium [94]. Since there is no
conclusive data on the involvement of NKCC in the
regulation of intracellular Na
+
and K
+
content un-
der baseline conditions [61, 89], elevation of [Cl
)
]
i
seen under NKCC activation in the above-listed cells
[52, 69, 77] seems to be crucial for proliferative
responses (Fig. 3).
Pathophysiological implications
Augmented activity of NKCC1 has been detected in
blood cells and VSMC from spontaneously hyper-
tensive rats (SHR) and in some patients with essential
hypertension [80]. Two hypotheses explain the pos-
sible involvement of abnormal activities of this
carrier in the pathogenesis of hypertension. First,
NKCC1 activation leads to augmented contractile
responses of VSMC, as demonstrated by our group
[7, 55] and by OÕNeill and co-workers [2, 3]. Sec-
ond, NKCC1-mediated elevation of [Cl
)
]
i
causes
heightened proliferation of VSMC (Fig. 3), i.e., a
well-documented hallmark of vascular remodeling
(increased wall-to-lumen ratio) detected in hyperten-
sion and implicated in the development of cardio-
vascular complications of this disease [34, 88]. The
latter hypothesis is consistent with data showing
shortening of the G
0
/G
1
phase in cultured VSMC
[HCO3-]i
sAC
cAMP
ATP
cAMP-mediated responses
2Cl-
NKCC1
[Cl-]i
?
proliferation
K+
Na+
SCl-
ClRE COX-2
?
Fig. 3. Intracellular monovalent anions as second messengers. S
Cl
:
hypothetical sensor of intracellular Cl
)
;ClRE:Cl
)
-sensitive ele-
ment implicated in the regulation of gene expression; ?: unknown
intermediates; - and --| : activatory and inhibitory signals,
respectively. For more details, see text.
166 S.N. Orlov and P. Hamet: Monovalent Ions as Second Messengers
from SHR [43, 121] and lengthening of the same cell-
cycle phase in bumetanide-treated cells [51]. Impor-
tantly, vascular remodeling might be further en-
hanced by elevation of [Na
+
]
i
caused by augmented
production of endogenous CTS documented in
hypertension and several other extracellular fluid
volume-expanded disorders [81]. This hypothesis is
consistent with data on Na
+i
-dependent inhibition of
apoptosis in VSMC considered in the Sodium section.
So-called ‘‘kidney resetting’’, i.e., normal salt
and water excretion under elevated blood pressure,
is the most powerful servomechanism of the long-
term maintenance of severe hypertension indepen-
dently of the origin of this disease, including
hypertension caused by enhanced salt consumption
[40, 41]. Several lines of evidence strongly suggest
that elevated intake of Cl
)
rather than Na
+
triggers
the development of NaCl-induced hypertension [53,
111]. It may be assumed that similar to VSMC
(Chloride section) elevation of [Cl
)
] in the macula
densa, caused by augmented NaCl intake via renal
specific NKCC2 isoform [57, 78] leads to the in-
creased contraction of mesangial cells, thus provid-
ing an explanation for the altered tubuloglomerular
feedback regulation of kidney function. In addition,
luminal [Cl
)
] negatively correlates with renin pro-
duction in juxtaglomerular preparations [46, 54]. In
the macula densa, renin secretion is under the con-
trol of cyclooxygenase (COX) activity and prosta-
glandin production [35] (Fig. 3). More recent studies
show that COX-2 expression is augmented in Cl
)
-
depleted medium and in the presence of NKCC
inhibitors [20]. The mechanism of the involvement
of [Cl
)
] in the regulation of COX-2 expression
remains unknown.
As for the physiological significance of H
+i
-
sensing, it must be noted that acidosis with pH
i
< 6.5
is considered a hallmark of hypoxia and ischemia
[112]. In several tissues, including the heart [126],
brain [8] and kidney [50], short ischemic precondi-
tioning protects cells from death caused by a sub-
sequent severe ischemic event. Importantly, both
ouabain-treated cells (Proton section) and cells sub-
jected to severe ischemia [60] possessed combined
markers of apoptosis and necrosis. Moreover, simi-
larly to ouabain-treated cells, the protective action of
ischemic preconditioning on severe ischemia-induced
tissue damage was transient [62] and diminished
sharply in the presence of cycloheximide [8]. The
protective action of acidification was also demon-
strated in mouse macrophages subjected to UV-irra-
diation [98], serum-deprived bovine and human
umbilical vein endothelial cells [26, 27] and endothe-
lial cells from human pulmonary arteries treated with
staurosporin [25]. Viewed collectively, these data
suggest that intracellular signaling triggered by H
+i
sensor plays a universal role in modulation of efficacy
of the cell death machinery.
Search for Intracellular Monovalent Ion Sensors
The ability of proteins to sense the modulation of
monovalent ion concentration is strongly supported
by cell physiology and molecular biology data ob-
tained in studies of superfamilies of monovalent ion
pumps, carriers and channels (Fig. 1). However, with
the exception of HCO
3
)
-sensitive sAC, the molecular
origin of monovalent ion sensors distinct from ion
transporters and involved in intracellular signaling is
still a mystery. This statement is in contrast with
rapid progress in the identification of Ca
2+i
sensors
and may be explained by several features of these
molecules. First, the high thermostability of several
Ca
2+
-binding proteins, such as calmodulin, and the
well-defined molecular origin of their targets, such as
phosphodiesterase and plasma membrane Ca
2+
-AT-
Pase, allowed researchers to purify them and to
identify their amino acid sequence, even before the
molecular biology era. As shown above, the down-
stream targets of monovalent cations and Cl
)
sensors
are still unknown. Second, high-affinity Ca
2+i
sensors
are almost completely saturated at [Ca
2+
]
i
of 1 lM,
and their Ca
2+
-binding sites are slightly affected by
the presence of monovalent cations and Mg
2+
. This
feature led to the identification of amino acid residues
by
45
Ca binding assay. In contrast to Ca
2+
,Na
+
,
K
+
, HCO
3
)
and Cl
)
affect cellular function in the
millimolar range. If monovalent ions bind with low-
affinity sensors, these interactions may be affected
by numerous factors, which complicates their
identification by screening with radioisotopes. Third,
high-affinity binding sites, initially detected in parv-
albumins and calmodulin, are formed by a highly
conservative linear amino acid sequence consisting of
14 amino acid residues (the so-called ‘‘EF-hand’’
domain). This knowledge led to the rapid identifica-
tion of more than 30 other Ca
2+i
sensors by the
screening of cDNA libraries [47]. In contrast,
monovalent ion sensors are probably formed by 3-D
protein structures and recruit space-separated amino
acid residues. This hypothesis is consistent with data
obtained by the identification of amino acid residues
in monovalent ion transporters performed with sin-
gle-point mutated constructs. Thus, it was shown that
Ala
330
, Glu
786
, Glu
796
, Asn
783
and Asp
815
contribute
to Na
+
binding within one of the 3 Na
+
-binding sites
of the Na
+
/K
+
-ATPase a-subunit [103].
Keeping these data in mind, we tried to identify
aNa
+
response element (NaRE) involved in c-Fos
expression triggered by the sustained inhibition of
Na
+
/K
+
-ATPase (Sodium section). In case of
positive results, this approach can lead to the iden-
tification of an upstream Na
+i
-sensor in a 2-hybrid
yeast system, i.e., by mating yeast transformed with
NaRE of the c-Fos promoter as a bait, with yeast
expressing an activation domain fusion cDNA li-
brary. This approach was supported by previous
S.N. Orlov and P. Hamet: Monovalent Ions as Second Messengers 167
data on mouse NIH 3T3 cells transfected with
human c-Fos showing the lack of an activatory ac-
tion of ouabain on human c-Fos mRNA content
after deletion of the )222 to )70 promoter region
[76]. To achieve our goal, we transfected HeLa cells
with the vector encoding the luciferase reporter gene
under the control of the human c-Fos )650 to +103
region, which contains all known transcription ele-
ments of c-Fos promoter. With this construct, we
failed to detect any significant elevation of luciferase
expression in HeLa cells subjected to 6-h inhibition
of Na
+
/K
+
-ATPase with ouabain or in K
+
-free
medium, which contrasted with massive accumula-
tion of endogenous c-Fos mRNA and immunore-
active protein in ouabain-treated HeLa cells
(unpublished data). We also did not observe any
positive impact of Na
+
/K
+
-ATPase inhibition on
luciferase expression driven by Elk-1, SRF, CREB
and AP1 transcription factors [115]. At least two
hypotheses could explain our negative results. (i)
Chromatin architecture involved in the formation of
NaRE is different in c-Fos and luciferase driven by
c-Fos promoter. Indeed, nuclease digestion assays
documented that chromatin transcription-sensitive c-
Fos sites are located in a region centered to )350
base pair as well as at a position )1900, i.e., up-
stream of the classical promoter [48]. (ii) Side-by-
side with 5¢-untranslated region, introns contribute
to organization of c-Fos NaRE. The role of introns
in transcriptional regulation has been proved in the
study of several genes, including c-Fos (24;72) and
WNK kinase [127].
In conclusion, we would like to stress that the
proposed models of monovalent ion sensors (Figs. 2
and 3) are based on the assumption that their
function occurs in a fixed environment. This is not a
case of the cytoplasm containing up to 0.4 g of
protein and 0.1 g of other macromolecules, such as
carbohydrates, lipids and nucleic acids, per ml of
intracellular water [6, 74]. Because of extreme mac-
romolecular crowding, cytoplasm functions as a gel
undergoing gel-sol phase transitions in response to
diverse stimuli, including modulation of the content
of monovalent ions [99]. We speculate that these
transitions per se can affect the functions of target
proteins, thus contributing to the mechanism sensing
monovalent ion concentrations. Further investiga-
tions should be performed to confirm or reject this
hypothesis and to design new approaches for iden-
tification of the molecular nature of monovalent ion
sensors.
This work was supported by grants from the Canadian Institutes of
Health Research, the Heart and Stroke Foundation of Canada and
the Kidney Foundation of Canada. The editorial help of Ovid Da
SilvaÕResearch Support Office, CHUM, is appreciated.
References
1. Adragna, N., Di Fulvio, M., Lauf, P.K. 2004. Regulation of
K-Cl cotransport: from function to genes. J. Membrane. Biol.
201:109–137
2. Akar, F., Jiang, G., Paul, R.J., OÕNeill, W.C. 2001. Con-
tractile regulation of the Na
+
-K
+
-2Cl
)
cotransporter in
vascular smooth muscle. Am. J. Physiol. 281:C579–C584
3. Akar, F., Skinner, E., Klein, J.D., Jena, M., Paul, R.J.,
OÕNeill, W.C. 1999. Vasoconstrictors and nitrovasodilators
reciprocally regulate the Na
+
-K
+
-2Cl
)
cotransporter in rat
aorta. Am. J. Physiol. 276:C1383–C1390
4. Akimova, O.A., Bagrov, A.Y., Lopina, O.D., Kamernitsky,
A.V., Tremblay, J., Hamet, P., Orlov, S.N. 2005. Cardiotonic
steroids differentially affect intracellular Na
+
and [Na
+
]
i
/
[K
+
]
i
-independent signaling in C7-MDCK cells. J. Biol.
Chem. 280:832–839
5. Akimova, O.A., Pchejetski, D., Hamet, P., Orlov, S.N. 2006.
Modest intracellular acidification suppresses death signaling
in ouabain-treated cells. Pfluegers Archiv 451:569–578
6. Al-Habori, M. 2001. Macromolecular crowding and its role
as intracellular signalling of cell volume regulation. Int. J.
Biochem. Cell Biol. 33:844–864
7. Anfinogenova, Y.J., Baskakov, M.B., Kovalev, I.V., Kilin,
A.A., Dulin, N.O., Orlov, S.N. 2004. Cell-volume-dependent
vascular smooth muscle contraction: role of Na
+
,K
+
, 2Cl
)
cotransport, intracellular Cl
)
and L-type Ca
2+
channels.
Pfluegers Archiv 449:42–55
8. Barone, F.C., White, R.F., Spera, P.A., Ellison, J., Currie, R.
W., Wang, X., Feuerstein, G. Z. 1998. Ischemic precondi-
tioning and brain tolerance. Temporal histological and func-
tional outcomes, protein synthesis requirement, and
interleukin-1 receptor antagonists and early gene expression.
Stroke 29:1937–1951
9. Bennekou P., Christophersen. P. 2003. Ion channels. In:.
Bernhardt I., Ellory J. C., eds. Red Cell Membrane Transport
in Health and Disease. Springer, Berlin pp139–152.
10. Berridge, M.J. 1993. Inositol triphosphate and calcium sig-
nalling. Nature 361:315–325
11. Blaustein, M.P., Lederer, W.J. 1999. Sodium/calcium ex-
change: its physiological implications. Physiol. Rev. 79:763–
854
12. Blumenstein, Y., Maximyuk, O.P., Lozovaya, N., Yatsenko,
N.M., Kanevsky, N., Kristal, O., Dascal, N. 2004. Intracel-
lular Na
+
inhibits voltage-dependent N-type Ca
2+
channels
by a G protein bc subunit-dependent mechanism. J. Physiol.
556:121–134
13. Breitwieser, G.E., Altamirano, A.A., Russell, J.M. 1990. Os-
motic stimulation of Na
+
-K
+
-Cl
)
cotransport in squid giant
axon is [Cl
)
]
i
dependent. Am. J. Physiol. 258:C749–C753
14. Brown, R.A., Chipperfield, A.R., Davis, J.P.L., Harper, A.A.
1999. Increased (Na
+
K
+
Cl
)
) cotransport in rat arterial
smooth muscle in deoxycorticosterone (DOCA)/salt-induced
hypertension. J. Vasc. Res. 36:492–501
15. Burns, C.P., Rozengurt, E. 1984. Extracellular Na
+
and ini-
tiation of DNA synthesis: role of intracellular pH and K
+
.J.
Cell Biol. 98:1082–1089
16. Cahn, F., Lubin, M. 1978. Inhibition of elongation steps of
protein synthesis at reduced potassium concentrations in
reticulocytes, reticulocyte lysate. J. Biol. Chem. 253:7798–
7803
17. Canessa, M., Salazar, G., Werner, E., Vallega, G., Gonzalez,
A. 1994. Cell growth and Na-K-Cl cotransport responses of
vascular smooth muscle cells of Milan rats. Hypertension
23:1022–1026
168 S.N. Orlov and P. Hamet: Monovalent Ions as Second Messengers
18. Carafoli, E. 2002. Calcium signaling: a tale for all seasons.
Proc. Natl. Acad. Sci. USA 99:1115–1122
19. Chen, Y., Cann, M.J., Litvin, T.N., Iourgenko, V., Sinclair,
M.L., Levin, L.R., Buck, J. 2000. Soluble adenylyl cyclase as
an evolutionarily conserved bicarbonate sensor. Science
289:625–628
20. Cheng, H.F., Wang, J.L., Zhang, M.Z., McKanna, J.A.M.,
Harris., R.C. 2000. Role of p38 in the regulation of renal
cortical cyclooxygenase-2 expression by extracellular chloride.
J. Clin. Invest. 106:681–688
21. Chipperfield, A.R., Harper, A.A. 2001. Chloride in smooth
muscle. Prog. Biophys. Mol. Biol. 74:175–221
22. Contreras, R.G., Lazaro, A., Mujica, A., Gonzalez-Mariscal,
L., Valdes, J., Garcia-Villegas, M. R., Cereijido, M. 1995.
Ouabain resistance of the epithelial cell line (Ma104) is not
due to lack of affinity of its pumps for the drug. J. Membrane
Biol. 145:295–300
23. Contreras, R. G., Shoshani, L., Flores-Maldonado, C.,
Lazaro, A., Cereijido, M. 1999. Relationship between Na
+
,
K
+
-ATPase and cell attachment. J. Cell Sci. 112:4223–4232
24. Coulon, V., Veyrune, J.-L., Tourkine, N., Vie
´, A., Hipskind,
R.A., Blanchard, J.-M. 1999. A novel calcium signaling
pathway targets the c-fos intragenic transcriptional pausing
site. J. Biol. Chem. 274:30439–30446
25. Cutaia, M., Tollefson, K., Kroczynski, J., Parks, N., Rounds,
S. 2000. Role of Na/H antiport in pH-dependent cell death in
pulmonary artery endothelial cells. Am. J. Physiol. 278:L536–
L544
26. DÕArcangelo, D., Facchiano, F., Barlucchi, L.M., Melilo, G.,
Illi, B., Testolin, L., Gaetano, C., et al. 2000. Acidosis inhibits
endothelial cell apoptosis and function and induces basic
fibroblast growth factor and vascular endothelial growth
factor expression. Circ. Res. 86:312–318
27. DÕArcangelo, D., Gaetano, C., Capogrossi, M.C. 2002.
Acidification prevents endothelial cell apoptosis by Axl acti-
vation. Circ. Res. 91:e4–e12
28. Davis, J.P.L., Chipperfield, A.R., Harper, A.A. 1993. Accu-
mulation of intracellular chloride by (Na-K-Cl) cotransport
in rat arterial smooth muscle is enhanced in deoxycorticos-
terone acetate (DOCA)/salt hypertension. J. Mol. Cell.
Cardiol. 25:233–237
29. Delpire, E. 2000. Cation-chloride cotransporters in neuronal
communication. News Physiol. Sci. 15:309–312
30. Dorovkov, M.V., Pavur, K.S., Petrov, A.N., Ryazanov, A.G.
2002. Regulation of elongation factor-2 kinase by pH. Bio-
chemistry 41:13444–13450
31. Ewart, H.S., Klip, A. 1995. Hormonal regulation of the Na
+
-
K
+
-ATPase: mechanisms underlying rapid and sustained
changes in pump activity. Am. J. Physiol. 269:C295–C311
32. Falciola, J., Volet, B., Anner, R. M., Moosmayer, M.,
Lacotte, D., Anner, B.M. 1994. Role of cell membrane Na, K-
ATPase for survival of human lymphocytes in vivo. Biosci.
Rep. 14:189–204
33. Fe
´raille, E., Doucet, A. 2001. Sodium-potassium-adenosine-
triphosphatase-dependent sodium transport in the kidney:
hormonal control. Physiol. Rev. 81:345–418
34. Folkow, B. 1982. Physiological aspects of primary hyperten-
sion. Physiol. Rev. 62:347–504
35. Francisco, L.J., Osborn, J.L., DiBona, G.F. 1982. Prosta-
glandins in renin release during sodium deprivation. Am. J.
Physiol. 243:F261–F268
36. Gagnon, F., Hamet, P., Orlov, S.N. 1999. Na
+
,K
+
pump
and Na
+
-coupled ion carriers in isolated mammalian kidney
epithelial cells: regulation by protein kinase C. Can. J. Phys-
iol. Pharmacol. 77:305–319
37. Garty, N.B., Salomon, Y. 1987. Stimulation of partially
purified adenylate cyclase from bull sperm by bicarbonate.
FEBS Lett 218:148–152
38. Grinstein, S., Furuya, W., Downey, G.P. 1992. Activation of
permeabilized neutrophils: role of anions. Am. J. Physiol.
263:C78–C85
39. Grinstein, S., Smith, J.D., Benedict, S.H., Gelfand, E.W.
1989. Activation of sodium-hydrogen exchange by mitogens.
Curr. Topics Membr. Transport 34:331–343
40. Guyton, A.C. 1980. Arterial Pressure and Hypertension. WB
Saunders, Philadelphia
41. Guyton A.C., Coleman T.G., Cowley A.W. Jr., Scheel K.W.,
Manning R.D. Jr., Norman R.A. Jr. 1975. Arterial pressure
regulation: overriding dominance of the kidney in long-term
regulation and in hypertension. In: Laragh J.H., ed.
Hypertension Mechanisms. York Medical Books, New York,
pp 1–24
42. Haas, M., McBrayer, D., Lytle, C. 1995. [Cl
)
]
i
-dependent
phosphorylation of the Na-K-Cl cotransport protein of dog
tracheal epithelial cells. J. Biol. Chem. 270:28955–2861
43. Hadrava, V., Tremblay, J., Sekaly, R.-P., Hamet, P. 1992.
Accelerated entry of smooth muscle cells from spontaneously
hypertensive rats into the S phase of the cell cycle. Biochem.
Cell Biol. 70:599–604
44. Hamet P., Orlov S.N., DeBlois D., Sun Y., Kren V.,
Tremblay J. 2004. Angiotensin as a cytokine implicated in
accelerated cellular turnover. In: Unger T., Scholkens B.A.
eds. Handbook of Experimental Pharmacology. Unger,
Springer Verlag, New York, pp 71–98
45. Hayashi, H., Szaszi, K., Grinstein, S. 2002. Multiple modes of
regulation of Na
+
/H
+
exchangers. Ann. N.Y. Acad. Sci.
976:248–258
46. He, X.R., Greenberg, S.C., Briggs, J.P., Schermann, J. 2005.
Effects of furosemide and verapamil on the NaCl dependency
of macula densa-mediated renin secretion. Hypertension
26:137–142
47. Heizmann, C. W., Hunziker, W. 1991. Intracellular calcium-
binding proteins: more sites than insights. TiBS 16:98–103
48. Herrera, R.E., Nordheim, A., Stewart, A.F. 1997. Chromatin
structure analysis of the human c-Fos promoter rveals a
centrally positioned nucleosome. Chromosoma 106:284–292
49. Isaev, N.K., Stelmashook, E.V., Halle, A., Harms, C., Laut-
enschlager, M., Weih, M., Dirnagl, U., et al. 2000. Inhibition
of Na
+
,K
+
-ATPase activity in cultured cerebellar granule
cells prevents the onset of apoptosis induced by low potas-
sium. Neurosci. Lett. 283:41–44
50. Islam, C.F., Mathie, R.T., Dinneen, M.D., Kiely, E.A., Pe-
ters, A.M., Grace, P.A. 1997. Ischemia-reperfusion injury in
the rat kidney: the effect of preconditioning. Br. J. Urol.
79:842–847
51. Iwamoto, L.M., Fujiwara, N., Nakamura, K.T., Wada, R.K.
2004. Na-K-2Cl cotransporter inhibition impairs human lung
cellular proliferation. Am. J. Physiol. 287:L510–L514
52. Jiang, G., Klein, J.D., OÕNeill, W.C. 2001. Growth factors
stimulate the Na-K-2Cl cotransporter NKCC1 through a
novel Cl
)
-dependent mechanism. Am. J. Physiol. 281:C1948–
C1953
53. Kotchen, T.A. 2005. Contribution of sodium and chloride to
NaCl-induced hypertension. Hypertension 45:849–850
54. Kotchen, T.A., Galla, J. H., Luke, R.G. 1976. Failure of
NaHCO
3
and KHCO
3
to inhibit renin in the rat. Am. J.
Physiol. 231:F1050–F1056
55. Kovalev, I.V., Baskakov, M.B., Anfinogenova, Y.J., Borodin,
Y.L., Kilin, A.A., Minochenko, I.L., Popov, A.G., et al. 2003.
Effect of Na
+
,K
+
, 2Cl
)
cotransport inhibitor bumetanide on
S.N. Orlov and P. Hamet: Monovalent Ions as Second Messengers 169
electrical and contractile activity of smooth muscle cells in
guinea pig ureter. Bull. Exp. Biol. Med. 136:145–149
56. Kuchler, R.J. 1967. The role of sodium and potassium in
regulating amino acid accumulation and protein synthesis in
LM-strain mouse fibroblasts. Biochim. Biophys. Acta
136:473–483
57. Laamarti, M.A., Bell, P.D., Lapointe, J.-Y. 1998. Transport
and regulatory properties of the apical Na-K-2Cl cotrans-
porter of macula densa cells. Am. J. Physiol. 275:F703–F709
58. Lanius, R.A., Pasqualotto, B.A., Shaw, C.A. 1993. c-Amin-
obutyric acid A receptor regulation by a chloride-dependent
kinase and a sodium-dependent phosphatase. Brain Res. Mol.
Brain Res. 20:192–198
59. Ledbetter, M.L.S., Lubin, M. 1977. Control of protein syn-
thesis in human fibroblasts by intracellular potassium. Exp.
Cell Res. 105:223–236
60. Lee, J.-M., Grabb, M.C., Zipfel, G.J., Choi, D. W. 2000.
Brain tissue responses to ischemia. J. Clin. Invest. 106:723–
731
61. Lenart, B., Kintner, D.B., Shull, G.E., Sun, D. 2004. Na-K-Cl
cotransporter-mediated intracellular Na
+
accumulation af-
fects Ca
2+
signaling in astrocytes in an in vitro ischemic
model. J. Neurosci. 24:9585–9597
62. Li, Y.W., Whittaker, P., Kloner, R.A. 1992. The transient
nature of the effect of ischemic preconditioning on myocardial
infarct size and ventricular arrhythmia. Am. Heart J. 123:346–
353
63. Lincoln, T.M., Cornwell, T. L. 1993. Intracellular cyclic GMP
receptor proteins. FASEB J. 7:328–338
64. Lubin, M. 1967. Intracellular potassium and macromolecular
synthesis in mammalian cells. Nature 213:451–453
65. Lubin, M., Ennis, H.L. 1964. On the role of intracellular
potassium in protein synthesis. Biochim. Biophys. Acta 80:614–
631
66. Lytle, C., Forbush, B. 1996. Regulatory phosphorylation of
the secretory Na-K-Cl cotransporter: modulation by cyto-
plasmic Cl. Am. J. Physiol. 270:C437–C448
67. Maingret, F., Patel, A., Lesage, J.F., Lazdunski, M., Honore
´,
E. 1999. Mechano- and acid stimulation, two interactive
modes of activation of the TREK-1 potassium channels. J.
Biol. Chem. 274:26691–26696
68. Majno, G., Joris., I. 1995. Apoptosis, oncosis, and necrosis.
An overview of cell death. Am. J. Pathol. 146:3–15
69. Mallis, L., Guber, H., Adler, S.G., Palant, C.E. 1991. Intra-
cellular chloride activity in cultured mesangial cells. Renal
Physiol. Biochem. 14:12–18
70. Marakhova, I.I., Vereninov, A.A., Toropova, F.V.,
Vinogradova, T.A. 1998. Na,K,-ATPase pump in activated
human lymphocytes: on the mechanisms of rapid and long-
term increase in K influxes during the initiation of phytohe-
magglutinin-induced proliferation. Biochim. Biophys. Acta.
1368:61–72
71. Matsuyama, S., Reed, J.C. 2000. Mitochondria-dependent
apoptosis and cellular pH regulation. Cell Death Different.
7:1155–1165
72. Mechti, N., Piechaczyk, M., Blanchard, J.M., Jeanteur, P.,
Lebleu, B. 1991. Sequence requirements for premature tran-
scription arrest within the first intron of the mouse c-fos gene.
Mol. Cell. Biol. 11:2832–2841
73. Meeker, G.L. 1970. Intracellular potassium requirement for
protein synthesis and mitotic apparatus in sea urchin eggs.
Exp. Cell Res. 63:165–170
74. Minton, A.P. 2001. The influence of macromolecular crowd-
ing and macromolecular confinement on biochemical reac-
tions in physiological media. J. Biol. Chem. 276:10577–10580
75. Muimo, R., Banner, S.J., Marshall, L.J., Mehta, A. 1998.
Nucleoside diphosphate kinase and Cl
)
-sensitive protein
phosphorylation in apical membranes from ovine airway
epithelium. Am. J. Respir. Cell Mol.Biol. 18:270–278
76. Nakagawa, Y., Rivera, V., Larner, A.C. 1992. A role for Na/
K-ATPase in the control of human c-fos and c-jun tran-
scription. J. Biol. Chem. 267:8785–8788
77. OÕNeill, W.C., Steinberg, D.F. 1995. Functional coupling of
Na
+
-K
+
-2Cl
)
cotransport and Ca
2+
-dependent K
+
chan-
nels in vascular endothelial cells. Am. J. Physiol 269:C267–
C274
78. Obermuller, N., Kunchaparfy, S., Ellison, D.H., Bachmann,
S. 1996. Expression of the Na-K-2Cl cotransporter by macula
densa and thick ascending limb cells of rabbit and rat neph-
ron. J. Clin. Invest. 98:635–640
79. Okamura, N., Tajima, Y., Onoe, S., Sugita, Y. 1991.
Purification of bicarbonate-sensitive sperm adenylyl cyclase
by 4-acetamido-4¢-isothiocyanostilbene-2,2¢-disulfonic acid-
affinity chromatography. J. Biol. Chem. 266:17754–17759
80. Orlov, S.N., Adragna, N., Adarichev, V.A., Hamet, P. 1999.
Genetic and biochemical determinants of abnormal mono-
valent ion transport in primary hypertension. Am. J. Physiol.
276:C511–C536
81. Orlov, S.N., Akimova, O.A., Hamet, P. 2005. Cardiotonic
steroids: novel mechanisms of Na
+i
-mediated and -inde-
pendent signaling involved in the regulation of gene
expression, proliferation and cell death. Curr. Hypertens.
Rev. 1:143–257
82. Orlov, S.N., Hamet, P. 2004. Apoptosis vs oncosis: role of cell
volume and intracellular monovalent cations. Adv. Exp. Med.
Biol. 559:219–233
83. Orlov, S.N., Resink, T.J., Bernhardt, J., Ferracin, F., Buhler,
F.R. 1993. Vascular smooth muscle cell calcium transport.
Regulation by angiotensin II and lipoproteins. Hypertension
21:195–203
84. Orlov, S.N., Taurin, S., Thorin-Trescases, N., Dulin, N.O.,
Tremblay, J., Hamet, P. 2000. Inversion of the intracellular
Na
+
/K
+
ratio blocks apoptosis in vascular smooth muscle
cells by induction of RNA synthesis. Hypertension 35:1062–
1068
85. Orlov, S.N., Taurin, S., Tremblay, J., Hamet, P. 2001. Inhi-
bition of Na
+
,K
+
pump affects nucleic acid synthesis and
smooth muscle cell proliferation via elevation of the [Na
+
]
i
/
[K
+
]
i
ratio: possible implication in vascular remodeling. J.
Hypertens. 19:1559–1565
86. Orlov, S.N., Thorin-Trescases, N., Kotelevtsev, S.V.,
Tremblay, J., Hamet, P. 1999. Inversion of the intracellular
Na
+
/K
+
ratio blocks apoptosis in vascular smooth muscle at
a site upstream of caspase-3. J. Biol. Chem. 274:16545–16552
87. Orlov, S.N., Thorin-Trescases, N., Pchejetski, D., Taurin, S.,
Farhat, N., Tremblay, J., Thorin, E., et al. 2004. Na
+
/K
+
pump and endothelial cell survival: [Na
+
]
i
/[K
+
]
i
-indepen-
dent necrosis triggered by ouabain, and protection against
apoptosis mediated by elevation of [Na
+
]
i
.Pfluegers Arch.
448:335–345
88. Orlov, S.N., Tremblay, J., DeBlois, D., Hamet, P. 2002.
Genetics in programmed cell death and proliferation. Semin.
Nephrol. 22:161–171
89. Orlov, S.N., Tremblay, J., Hamet, P. 1996. Bumetanide-sen-
sitive ion fluxes in vascular smooth muscle cells: lack of
functional Na
+
,K
+
, 2Cl
)
cotransport. J. Membrane Biol.
153:125–135
90. Orlowski, J., Grinstein, S. 2004. Diversity of the mammalian
sodium/proton exchanger SLC9 gene family. Pfluegers Arch.
447:549–565
170 S.N. Orlov and P. Hamet: Monovalent Ions as Second Messengers
91. Panet, R., Atlan, H. 1991. Stimulation of bumetanide-sensi-
tive Na
+
/K
+
/Cl
)
cotransport by different mitogens in syn-
chronized human skin fibroblasts is essential for cell
proliferation. J. Cell Biol. 114:337–342
92. Panet, R., Ellash, M., Pick, M., Atlan, H. 2002. Na
+
/K
+
/Cl
)
cotransporter activates mitogen-activated protein kinase in
fibroblasts and lymphocytes. J. Cell. Physiol. 190:227–237
93. Panet, R., Markus, M., Atlan, H. 1994. Bumetanide and
furosemide inhibited vascular endothelial cell proliferation. J.
Cell. Physiol. 158:121–127
94. Panet, R., Markus, M., Atlan, H. 2000. Overexpression of the
Na
+
/K
+
/Cl
)
cotransporter gene induces cell proliferation
and phenotypic transformation in mouse fibroblasts. J. Cell.
Physiol. 182:109–118
95. Pastor-Soler, N., Beaulieu, V., Litvin, T.N., Da Silva, N.,
Chen, Y., Brown, D., Buck, J., et al. 2003. Bicarbonate-reg-
ulated adenylyl cyclase (sAC) is a sensor that regulates pH-
dependent V-ATPase recycling. J. Biol. Chem. 278:49523–
49529
96. Pchejetski, D., Taurin, S., der Sarkissian, S., Lopina, O.D.,
Pshezhetsky, A.V., Tremblay, J., DeBlois, D., et al. 2003.
Inhibition of Na
+
,K
+
-ATPase by ouabain triggers epithelial
cell death independently of inversion of the [Na
+
]
i
/[K
+
]
i
ratio. Biochem. Biophys. Res. Commun. 301:735–744
97. Perez-Sala, D., Collado-Escobar, D., Mollinedo, F.
1995. Intracellular alkalinization suppresses lovastatin-
induced apoptosis in HL-60 cells through the inacti-
vation of a pH-dependent endonuclease. J. Biol. Chem.
270: 6235–6242
98. Pirutin, S.K., Turovetsky, V.B., Kudryashov, Y.B., Rubin,
A.B. 2002. Modification of damaging effect of ultraviolet
radiation on mice peritoneal macrophage membranes. Radiat.
Biol. Radioecol. 42:151–154
99. Pollack, G. H. 2004. Cells, Gels and the Engines of Life.
Ebner & Sons, Seattle, WA
100. Pollack, M., Fisher, H.W. 1976. Dissociation of ribonucleic
acid and protein synthesis in mammalian cells deprived of
potassium. Arch. Biochem. Biophys. 172:188–190
101. Prasad, K.V.S., Severini, A., Kaplan, J.G. 1987. Sodium ion
fluxes in proliferating lymphocytes: an early component of
mitogenic signal. Arch. Biochem. Biophys. 252:515–525
102. Rajan, S., Wischmeter, E., Liu, G.X., Preisig-Muller, R.,
Daut, J., Karschin, A., Derst, C. 2000. TASK-3, a novel
tandem pore domain acid-sensitive K
+
channel. An extra-
cellular histidine as pH sensor. J. Biol. Chem. 275:16650–
16657
103. Rakowski, R.F., Sagar, S. 2003. Found: Na
+
and K
+
binding sites of the sodium pump. News Physiol. Sci. 18:164–
168
104. Rishal, I., Keren-Raifman, T., Yakubovich, D., Ivanina, T.,
Dessauer, C.W., Slepak, V. Z., Dascal, N. 2003. Na
+
promotes the dissociation between GaGDP and Gbc, acti-
vating G-protein-gated K
+
channels. J. Biol. Chem. 278:
3840–3845
105. Robison, G.A., Butcher, R.W., Sutherland, E.W. 1971. Cyclic
AMP. Academic Press, New York
106. Rose, C.R., Konnerth, A. 2001. NMDA-receptor-mediated
Na
+
signals in spines and dendrites. J. Neurosci. 21:4207–4214
107. Runkel, L., Shaw, P.E., Herrera, R.E., Hipskind, R.A.,
Norheim, A. 1991. Multiple basal promoter elements deter-
mine the level of human c-fos transcription. Mol. Cell. Biol.
11: 1270–1280
108. Russell, J.M. 2000. Sodium-potassium-chloride cotransport.
Physiol. Rev. 80:212–276
109. Ryazanov, A.G. 2002. Elongation factor-2 kinase and its
newly discovered relatives. FEBS Lett. 514:26–29
110. Ryazanov, A.G., Shestakova, E.A., Natapov, P.G. 1988.
Phosphorylation of elongation factor 2 by EF-2 kinase affects
rate of translation. Nature 334:170–173
111. Schmidlin, O., Tanaka, M., Bollen, A.W., Yi, S.L., Morris,
R.B. 2005. Chloride-dominant salt sensitivity in the stroke-
prone spontaneously hypertensive rat. Hypertension 45:867–
873
112. Siesjo, B.K., Katsura, K., Kristian, T. 1996. Acidosis-related
damage. Adv. Neurol. 71:209–223
113. Sutherland, E.W. 1972. Studies on the mechanism of hor-
mone action. Science 177:401–408
114. Takano, S., Wadhwa, R., Yoshii, Y., Nose, T., Kaul, S.C.,
Mitsui, Y. 1997. Elevated level of mortalin expression in hu-
man brain tumors. Exp. Cell Res. 237:38–45
115. Taurin, S., Dulin, N.O., Pchejetski, D., Grygorczyk, R.,
Tremblay, J., Hamet, P., Orlov, S. N. 2002. c-Fos expression
in ouabain-treated vascular smooth muscle cells from rat
aorta: evidence for an intracellular-sodium-mediated, cal-
cium-independent mechanism. J. Physiol. 543:835–847
116. Taurin, S., Hamet, P., Orlov, S.N. 2003. Na/K pump and
intracellular monovalent cations: novel mechanism of exci-
tation-transcription coupling involved in inhibition of apop-
tosis. Mol. Biol. 37:371–381
117. Taurin, S., Seyrantepe, V., Orlov, S.N., Tremblay, T.-L.,
Thibaut, P., Bennett, M. R., Hamet, P. ,et al. 2002. Proteome
analysis and functional expression identify mortalin as an
anti-apoptotic gene induced by elevation of [Na
+
]
i
/[K
+
]
i
ratio
in cultured vascular smooth muscle cells. Circ. Res. 91:915–
922
118. Treharne, K.J., Riemen, C.E., Marshall, L.J., Muimo, R.,
Mehta, A. 2001. Nucleoside diphosphate kinase a compo-
nent of the [Na
+
]- and [Cl
)
]-sensitive phosphorylation cas-
cade in human and murine airway epithelium. Pfluegers Arch.
443:S97–S102
119. Tremblay, J., Gerzer, R., Vinay, P., Pang, S.C., Beliveau, R.,
Hamet, P. 1985. The increase of cGMP by atrial natriuretic
factor correlates with the distribution of particulate guanylate
cyclase. FEBS Letters 181:17–22
120. Trevisi, L., Visentin, B., Cusinato, F., Pighin, I., Luciani, S.
2004. Antiapoptotic effect of ouabain on human umbilical
endothelial cells. Biochem. Biophys. Res. Commun. 321:716–
721
121. Uehara, Y., Numabe, A., Kawabata, Y., Nagata, T.,
Hirawa, N., Ishimutsu, T., Matsuoka, H., et al. 1991. Rapid
smooth muscle cell growth and endogenous prostaglandin
system in spontaneously hypertensive rats. Am. J. Hypertens.
4:806–814
122. Wadhwa, R., Pereira-Smith, O.M., Reddel, R.R., Sugimoto,
Y., Mitsui, Y., Kaul, S.C. 1995. Correlation between com-
plementation group for immortality and the cellular distri-
bution of mortalin. Exp. Cell Res. 216:101–106
123. Wadhwa, R., Takano, S., Mitsui, Y., Kaul, S. C. 1999. NIH
3T3 cells malignantly transformed by mot-2 show inactivation
and cytoplasmic sequestration of the p53 proteins. Cell Res.
9:261–269
124. Wadhwa, R., Takano, S., Robert, M., Yoshida, A., Nomura,
H., Reddel, R.R., Mitsui, Y., et al. 1998. Inactivation of
tumor suppressor p53 by Mot-2, a hsp70 family member. J.
Biol. Chem. 273:29586–29591
125. Wakabayashi, S., Shigekawa, M., Poyssegur, J. 1997.
Molecular physiology of vertebrate Na
+
/H
+
exchanger.
Physiol. Rev. 77:51–74
126. Williams, R.S., Benjamin, I.J. 2000. Protective responses in
the ischemic myocardium. J. Clin. Invest. 106:813–818
127. Wilson, F.H., Disse-Nicodeme, S., Choate, K.A., Ishikawa,
K., Nelson-Williams, C., Desitter, I., Gunel, M., et al. 2001.
S.N. Orlov and P. Hamet: Monovalent Ions as Second Messengers 171
Human hypertension caused by mutations in WNK kinases.
Science 293:1107–1112
128. Xie, Z., Askari, A. 2002. Na
+
/K
+
-ATPase as a signal
transducer. Eur. J. Biochem. 269:2434–2439
129. Xiong, Z.-G., Zhu, X.-M., Chu, X.-P., Minami, M., Hey, J.,
Wei, W.-L., MacDonald, J.F., et al. 2004. Neuroprotection in
ishemia: blocking calcium-permeable acid-sensitive ion chan-
nels. Cell 118:687–698
130. Yu, X.-M., Salter, M. B. 1998. Gain control of NMDA-
receptor currents by intracellular sodium. Nature 396:469–474
131. Zhou, X., Jiang, G., Zhao, A., Bondeva, T., Hirzel, P., Balla,
T. 2001. Inhibition of Na,K-ATPase activates PI3 kinase and
inhibits apoptosis in LLC-PK1 cells. Biochem. Biophys. Res.
Commun. 285:46–51
132. Zippin, J. H., Farrell, J., Huron, D., Kamenetsy, M., Hess,
K.C., Fischman, D.A., Levin, L.R., et al. 2004. Bicarbonate-
responsive ‘‘soluble’’ adenylyl cyclase defines a nuclear cAMP
microdomain. J. Cell Biol. 164:527–534
172 S.N. Orlov and P. Hamet: Monovalent Ions as Second Messengers
... Conceptually, changes in the cytosolic concentration of any ion, di-or monovalent, can and does contribute to the regulation of cellular events, and hence any ion can and does act as a second messenger. 723 Despite being electrically non-excitable, glial cells express multiple channels, receptors and transporters, which upon physiological stimulation generate transmembrane ionic fluxes. These fluxes translate into spatiotemporally organised changes in the intracellular ion concentration and local non-regenerative astrocyte depolarisation. ...
... Each cell has its own resting intracellular ion-pattern determined by these ion concentrations together. "Intracellular ions together" is an entity that is one of the basic intracellular signalling systems [7][8][9]. The intracellular ion milieu represents the "software of the cell" compared to the hardware of the genome, as its current composition largely determines the biochemical processes currently taking place in the cell. ...
Article
Full-text available
The author presents a new model for age-associated diseases through a hypothesized pathogenesis of hypertension. Preservation or restoration of the constancy of each original cell-specific intracellular ion-pattern is essential for upholding cellular identity and integrity. The ageing background is intracellular acidosis: low-grade respiratory acidosis elevates the intracellular HCO3-/Cl- gradient and induces metabolic syndrome, while low-grade metabolic acidosis causes exhausting buffer syndrome (EBS) with a decreased intracellular HCO3-/Cl- rate. The former elicits higher aldosterone levels to restore the original ion-pattern by NHE-1 (natrium-hydrogen exchange) mechanism located in the membrane of vascular smooth muscle cells (VSMCs) and by retaining NaCl. The fault of ion-status restoration leads to Salt-sensitive hypertension through cascades of events. The ion-pattern changes in EBS elicit angiotensin II levels elevation through the renin-angiotensin system, which tries to eliminate intracellular changes via NBCn1 (natrium-bicarbonate cotransporter). However, it leads to a dead-end, and Salt-resistant hypertension develops. Hypertension occurs because restoring the original intracellular ion-pattern in VSMCs by aldosterone and angiotensin II fails. These hormonal counter-regulations lead to Na+ retention and alkaline overcompensation in the VSMCs. There may be several explanations for the failure, but hypertension would not develop if the intracellular ion-milieu were initially neutral or slightly alkaline. Regulated breathing could overcome hypertension.
... They can influence cellular processes in two ways. First, there is direct low-affinity binding of monovalent ions to proteins [81][82][83][84]. It is difficult to imagine, however, how such a mechanism could act as the first-response volume sensor because the relative magnitude of the concentration change can be small. ...
Article
High density of intracellular macromolecules creates a special condition known as macromolecular crowding (MC). One well-established consequence of MC is that only a slight change in the concentration of macromolecules (e.g., proteins) results in a shift of chemical equilibria towards the formation of macromolecular complexes and oligomers. This suggests a physiological mechanism of converting cell density changes into cellular responses. In this review, we start by providing a general overview of MC; then we examine the available experimental evidence that MC may act as a direct signaling factor in several types of cellular activities: mechano- and osmosensing, cell volume recovery in anisosmotic solutions, and apoptotic shrinkage. The latter phenomenon is analyzed in particular detail, as persistent shrinkage is known both to cause apoptosis and to occur during apoptosis resulting from other stimuli. We point to specific apoptotic reactions that involve formation of macromolecular complexes and, therefore, may provide a link between shrinkage and downstream responses.
... It is well known that monovalent ions intracellular concentrations change temporarily in response to various stimuli. Orlov and Hamet have formulated a hypothesis which proposes monovalent ions as secondary messengers [43]. Data on monovalent cation intracellular sensors has been reviewed in recent publication [44]. ...
Article
Full-text available
Stimulus-dependent elevation of intracellular Ca 2+ affects gene expression via well-documented calmodulin-mediated signaling pathways. Recently, we found that the addition of extra-and intracellular Ca 2+ chelators increased, rather than decreased, the number of genes expressed, and that this is affected by the elevation of [Na + ] i /[K + ] i-ratio. This assumes the existence of a novel Na + i /K + i-mediated Ca 2+ i-independent mechanism of excitation-transcription coupling. To identify upstream Na + i /K + i-sensitive genes, we examined the kinetics of transcriptomic changes in human umbilical vein endothelial cells (HUVEC) subjected to Na,K-ATPase inhibition by ouabain or K +-free medium. According to our data, microRNAs, transcription factors, and proteins involved in immune response and inflammation might be considered as key components of Na + i /K + i-mediated excitation-transcription coupling. Special attention was focused on the FOS gene and the possible mechanism of transcription regulation via G-quadruplexes, non-canonical secondary structures of nucleic acids, whose stability depends on [Na + ] i /[K + ] i-ratio. Verification of the [Na + ] i /[K + ] i-sensitive transcription regulation mechanism should be continued in forthcoming studies.
... Beside its importance for homeostatic processes, the transport of ions across the plasma membrane may change the membrane potential, thereby affecting cellular excitability. Changes in the ionic concentration can also modulate cellular responses, and ions themselves can act as a second messenger (Orlov and Hamet, 2006). Astrocytes, which essentially lack electrical excitability, use such ionic signaling as a main means for intracellular and intercellular communication (Verkhratsky and Nedergaard, 2018). ...
Article
Full-text available
Astrocytes are the main cell type responsible for the regulation of brain homeostasis, including the maintenance of ion gradients and neurotransmitter clearance. These processes are tightly coupled to changes in the intracellular sodium (Na⁺) concentration. While activation of the sodium-potassium-ATPase (NKA) in response to an elevation of extracellular K⁺ may decrease intracellular Na⁺, the cotransport of transmitters, such as glutamate, together with Na⁺ results in an increase in astrocytic Na⁺. This increase in intracellular Na⁺ can modulate, for instance, metabolic downstream pathways. Thereby, astrocytes are capable to react on a fast time scale to surrounding neuronal activity via intracellular Na⁺ fluctuations and adjust energy production to the demand of their environment. Beside the well-documented conventional roles of Na⁺ signaling mainly mediated through changes in its electrochemical gradient, several recent studies have identified more atypical roles for Na⁺, including protein interactions leading to changes in their biochemical activity or Na⁺-dependent regulation of gene expression. In this review, we will address both the conventional as well as the atypical functions of astrocytic Na⁺ signaling, presenting the role of transporters and channels involved and their implications for physiological processes in the central nervous system (CNS). We will also discuss how these important functions are affected under pathological conditions, including stroke and migraine. We postulate that Na⁺ is an essential player not only in the maintenance of homeostatic processes but also as a messenger for the fast communication between neurons and astrocytes, adjusting the functional properties of various cellular interaction partners to the needs of the surrounding network.
Article
NBCn1 (SLC4A7) is one of the two major Na ⁺ -HCO 3 ⁻ cotransporters in the human colonic epithelium, expressed predominantly in the highly proliferating colonocytes at the cryptal base. Increased NBCn1 expression levels are reported in tumours, including colorectal cancer. The study explores its importance for maintenance of the intracellular pH (pH i ), as well as the proliferative, adhesive, and migratory behavior of the self-differentiating Caco2bbe colonic tumour cell line. In the self-differentiating Caco2BBe cells, NBCn1 mRNA was highly expressed from the proliferative stage until full differentiation. The downregulation of NBCn1 expression by RNA interference affected proliferation and differentiation, and decreased intracellular pH (pH i ) of the cells in correlation with the degree of knockdown. In addition, a disturbed cell adhesion and reduced migratory speed were associated with NBCn1 knockdown. Murine colonic Nbcn1 -/- enteroids also displayed reduced proliferative activity. In the migrating Caco2BBe cells, NBCn1 was found at the leading edge and in colocalization with the focal adhesion markers vinculin and paxillin, which suggests that NBCn1 is involved in the establishment of cell-matrix adhesion. Our data highlight the physiological significance of NBCn1 in modulating epithelial pH-homeostasis and cell-matrix interactions in the proliferative region of the colonic epithelium, and unravel the molecular mechanism behind pathological overexpression of this transporter in human colorectal cancers.
Chapter
DNA replication, the process of copying one double stranded DNA molecule to form two identical copies, is highly conserved at the mechanistic level across evolution. Interesting in its own right as a fascinating feat of biochemical regulation and coordination, DNA replication is at the heart of modern advances in molecular biology. An understanding of the process at both the biological and chemical level is essential to developing new techniques in molecular biology. Insights into the process at the molecular level provide opportunities to modulate and intervene in replication. Rapidly dividing cells need to replicate their DNA prior to division, and targeting components of the replication process is a potentially powerful strategy in cancer treatment. Conversely, ageing may be associated with loss of replication activity and restoring it to cells may moderate some of the diseases associated with old age. Replication is, therefore, fundamental to a huge range of molecular biological and biochemical applications, and provides many potential targets for drug design. The fast pace of replication research, particularly in providing new structural insights, has outdated the majority of available texts. This learned, yet accessible, book contains the latest research written by those conducting it. It examines conserved themes providing a biological background for biochemical, chemical and pharmaceutical studies of this huge and exciting field. Rather than simply "itemising" the replication steps and the proteins involved, replication is tackled from a novel perspective. The book provides logical groupings of processes based upon biochemical similarities. The emphasis on mechanisms and the relationship between structure and function targets the chapters towards biochemists and biological chemists as well as molecular and cell biologists. The book highlights new insights into the replication process, from the assembly of pre-replication complexes, through polymerisation mechanisms, to considering replication in the context of chromatin and chromosomes. It also covers mitochondrial DNA replication, and includes archaeal paradigms, which are proving increasingly relevant to the study of replication in higher eukaryotes. Exciting potential drug targets in DNA replication are discussed, particularly in the context of treating malaria and cancer.
Article
The presented material raises the most important question in practical nephrology – how to correctly assess kidney function to understand the prognosis and duration of the predialysis period in a particular patient with chronic kidney disease (CKD)? From the standpoint of onto- and phylogenesis, the hierarchy of kidney functions was assessed. It is noted that the existing approaches to such an assessment are convenient for practice, but do not sufficiently take into account individual characteristics and are devoid of a load component that can show kidney reserves. The authors focused on the functional renal reserve (FRR) and the possibility of its detection. The above technique with 0.45 % saline allows revealing the true functional capabilities of the kidneys and understanding the patient's capabilities in the pre-dialysis period. Conclusions are drawn about the convenient use of the KDIGO scale, a more individual approach when using the QxMD calculator, and the possibility of a personalized approach when assessing the FRR.
Article
Water confined within hydrophobic spaces can undergo cooperative dewetting transitions due to slight changes in water density and pressure that push water toward the vapor phase. Many transmembrane protein ion channels contain nanoscale hydrophobic pores that could undergo dewetting transitions, sometimes blocking the flow of ions without physical blockages. Standard molecular dynamics simulations have been extensively applied to study the behavior of water in nanoscale pores, but the large free energy barriers of dewetting often prevent direct sampling of both wet and dry states and quantitative studies of the hydration thermodynamics of biologically relevant pores. Here, we describe a metadynamics protocol that uses the number of waters within the pore as the collective variable to drive many reversible transitions between relevant hydration states and calculate well-converged free energy profiles of pore hydration. By creating model nanopore systems and changing their radius and morphology and including various cosolvents, we quantify how these pore properties and cosolvents affect the dewetting transition. The results reveal that the dewetting free energy of nanoscale pores is determined by two key thermodynamic parameters, namely, the effective surface tension coefficients of water-air and water-pore interfaces. Importantly, while the effect of salt can be fully captured in the water activity dependence, amphipathic cosolvents such as alcohols modify both dry and wet states of the pore and dramatically shift the wet-dry equilibrium. The metadynamics approach could be applied to studies of dewetting transitions within nanoscale pores of proteins and provide new insights into why different pore properties evolved in biological systems.
Chapter
Proper use of fluids requires consideration of the basic physiology of fluids in the body. This includes consideration of the following questions. What does water do? What is the importance of elements such as Na⁺, Cl⁻, and K⁺ in bodily solutions? What determines how water and electrolytes distribute between the plasma space, interstitial space, and intracellular space? What is the role of colloids, and in particular, albumin? What happens when different types of fluids are infused intravenously? This chapter emphasizes the physiological role of fluids, principles behind the movement and distribution of water and it solutes, and the characteristics of different kinds of commonly infused fluids.
Article
Obligatory, coupled cotransport of Na ⁺ , K ⁺ , and Cl ⁻ by cell membranes has been reported in nearly every animal cell type. This review examines the current status of our knowledge about this ion transport mechanism. Two isoforms of the Na ⁺ -K ⁺ -Cl ⁻ cotransporter (NKCC) protein (∼120–130 kDa, unglycosylated) are currently known. One isoform (NKCC2) has at least three alternatively spliced variants and is found exclusively in the kidney. The other (NKCC1) is found in nearly all cell types. The NKCC maintains intracellular Cl ⁻ concentration ([Cl ⁻ ] i ) at levels above the predicted electrochemical equilibrium. The high [Cl ⁻ ] i is used by epithelial tissues to promote net salt transport and by neural cells to set synaptic potentials; its function in other cells is unknown. There is substantial evidence in some cells that the NKCC functions to offset osmotically induced cell shrinkage by mediating the net influx of osmotically active ions. Whether it serves to maintain cell volume under euvolemic conditons is less clear. The NKCC may play an important role in the cell cycle. Evidence that each cotransport cycle of the NKCC is electrically silent is discussed along with evidence for the electrically neutral stoichiometries of 1 Na ⁺ :1 K ⁺ :2 Cl− (for most cells) and 2 Na ⁺ :1 K ⁺ :3 Cl ⁻ (in squid axon). Evidence that the absolute dependence on ATP of the NKCC is the result of regulatory phosphorylation/dephosphorylation mechanisms is decribed. Interestingly, the presumed protein kinase(s) responsible has not been identified. An unusual form of NKCC regulation is by [Cl ⁻ ] i . [Cl ⁻ ] i in the physiological range and above strongly inhibits the NKCC. This effect may be mediated by a decrease of protein phosphorylation. Although the NKCC has been studied for ∼20 years, we are only beginning to frame the broad outlines of the structure, function, and regulation of this ubiquitous ion transport mechanism.
Book
This book describes our current understanding of the transport of ions, amino acids, nucleosides, sugars, water and gases across the red blood cell membrane. It also outlines the necessary theoretical background to understand the dynamics of membrane constituents together with the mechanisms of transport pathways (pumps, channels, carriers/cotransporters, residual passive permeability). Separate chapters describe our present ideas about membrane and metabolic disorders as well as red blood cell diseases like malaria, sickle cell disease, and hypertension. The latest findings are explained on the basis of a historical review and well-established principles. The book and its chapters are thus structured in a manner that makes the material accessible to beginners in the field of red blood cell physiology and biophysics. Active researchers will also benefit from this carefully organized compilation.
Article
Amiloride-sensitive Na+-H+ exchange activity has been detected in the plasma membrane of virtually all mammalian cell types studied to date. The exchanger, or antiport, catalyzes the electroneutral counter transport of Na+ for H+. Though the mechanism is reversible, entry of Na+ in exchange for intracellular H+ is the predominant transport mode under physiological conditions. This net extrusion of acid equivalents tends to offset the spontaneous tendency of the cells to become acidic, which results from metabolic acid production and from the electrodiffusive accumulation of H+ (or depletion of OH-) driven by the internally negative plasma membrane potential. The Na+-H+ antiport is virtually quiescent at physiological cytosolic pH (pHi), but is markedly activated when the cytoplasm becomes acidic. Such behavior, together with the direction of net acid transport, suggests that the antiport plays an important role in the regulation of pHi in resting (nonproliferating) cells. In addition to being activated by cytosolic acidification, the antiport can also be stimulated without prior pHi change by the addition of mitogenic or comitogenic agents such as growth factors and phorbol esters, respectively.
Chapter
Several models of essential hypertension have revealed abnormalities in pathways regulating cell proliferation and programmed cell death (apoptosis). The increased proliferative phenotype found as early as birth in hypertensives is accompanied by age-dependent alterations in apoptosis, contributing to neonatal hyperplasia of the heart, aorta, and kidneys. During the course of life, accelerated cell turnover occurs and is modifiable by antihypertensive therapy, notably by inhibitors of the renin-angiotensin system. We consider the hypothesis that hypertension may be a case of accelerated aging. Part of this process may involve the defective regulation of cell proliferation in cardiovascular target organs via a putative specific senescence pathway. Candidates include abnormalities in cell cycle control genes, the renin-angiotensin pathway, and regulation of the telomerase pathway. Abnormal activity of angiotensin II-regulated Na+ transporters and augmented production of endogenous ouabain-like substances have been detected in experimental models of primary hypertension. Recent data show that both ouabain and intracellular Na+ are involved in the regulation of gene expression and apoptosis. The relevance of neonatal and early life development as a predictor of cardiovascular disease outcomes later in life is an intriguing issue that remains to be better defined. In this regard, understanding the complex genetic and epigenetic influences contributing to aging and age-related diseases will be a major goal. Because phenotype development can be analyzed longitudinally during the course of life in recombinant inbred rat strains, these models will allow a systematic approach to the molecular analysis of senescence pathway regulation, their determinants early in life, and their control by hereditary and epigenetic factors, including pharmacotherapy.
Chapter
Erythrocyte ion channels have been described from several animal species and have been more or less extensively characterized. However, the most comprehensive picture has been established for mammalian erythrocytes, especially human, which will be the focus of this chapter. Among the pathways for passive, conductive transport of inorganic ions, two different cation channels have been extensively characterized at present, an intermediate conductance Ca2+-activated K+ channel, also known as the Gárdos channel, and a voltage-dependent non-selective cation channel. It should be remembered, however, that transport mediated by these channels was observed decades before ion channels were established facts of life.
Article
This review updates our current knowledge on the regulation of Na+/H+ exchanger, Na+,K+,Cl- cotransporter, Na+,P-i cotransporter, and Na+,K+ pump in isolated epithelial cells from mammalian kidney by protein kinase C (PKC). In cells derived from different tubule segments, an activator of PKC, 4 beta-phorbol 12-myristate 13-acetate (PMA), inhibits apical Na+/H+ exchanger (NHE3), Na+,P-i cotransport, and basolateral Na+,K+ cotransport (NKCCl) and augments Na+,K+ pump. In PMA-treated proximal tubules, activation of Na+,K+ pump probably plays a major role in increased reabsorption of salt and osmotically obliged water. in Madin-Darby canine kidney (MDCK) cells, which are highly abundant with intercalated cells from the collecting duct, PMA completely blocks Na+,K+,Cl- cotransport and decreases the activity of Na+,P-i cotransport by 30-40%. In these cells, agonists of P-2 purinoceptors inhibit Na+,K+,Cl- and Na+,P-i cotransport by 50-70% via a PKC-independent pathway. In contrast with MDCK cells, in epithelial cells derived from proximal and distal tubules of the rabbit kidney, Na+,K+,Cl- cotransport is inhibited by PMA but is insensitive to P-2 receptor activation. In proximal tubules, PKC-induced inhibition of NHE3 and Na+,P-i cotransporter can be triggered by parathyroid hormone. Both PKC and cAMP signaling contribute to dopaminergic inhibition of NHE3 and Na+,K+ pump. The receptors triggering PKC-mediated activation of Na+,K+ pump remain unknown. Recent data suggest that the PKC signaling system is involved in abnormalities of dopaminergic regulation of renal ion transport in hypertension and in the development of diabetic complications. The physiological and pathophysiological implications of PKC-independent regulation of renal ion transporters by P-2 purinoceptors has not yet been examined.
Article
The historical development of the cell death concept is reviewed, with special attention to the origin of the terms necrosis, coagulation necrosis, autolysis, physiological cell death, programmed cell death, chromatolysis (the first name of apoptosis in 1914), karyorhexis, karyolysis, and cell suicide, of which there are three forms: by lysosomes, by free radicals, and by a genetic mechanism (apoptosis). Some of the typical features of apoptosis are discussed, such as budding (as opposed to blebbing and zeiosis) and the inflammatory response. For cell death not by apoptosis the most satisfactory term is accidental cell death. Necrosis is commonly used but it is not appropriate, because it does not indicate a form of cell death but refers to changes secondary to cell death by any mechanism, including apoptosis. Abundant data are available on one form of accidental cell death, namely ischemic cell death, which can be considered an entity of its own, caused by failure of the ionic pumps of the plasma membrane. Because ischemic cell death (in known models) is accompanied by swelling, the name oncosis is proposed for this condition. The term oncosis (derived from onkos, meaning swelling) was proposed in 1910 by von Reckling-hausen precisely to mean cell death with swelling. Oncosis leads to necrosis with karyolysis and stands in contrast to apoptosis, which leads to necrosis with karyorhexis and cell shrinkage.