ISSN 0006-2979, Biochemistry (Moscow), 2025, Vol. 90, No. 5, pp. 582-589 © Pleiades Publishing, Ltd., 2025.
Published in Russian in Biokhimiya, 2025, Vol. 90, No. 5, pp. 627-635.
582
FOS Promoter is Overactive Outside of Genome Context
and Weakly Regulated by Changes in the Na
+
i
/K
+
i
Ratio
Andrey M. Gorbunov
1
, Dmitrii A. Fedorov
1
, Olga E.Kvitko
1
,
Olga D. Lopina
1
, and Elizaveta A. Klimanova
1,a
*
1
Faculty of Biology, Lomonosov Moscow State University, 119234 Moscow, Russia
a
e-mail: klimanova.ea@yandex.ru
Received February 10, 2025
Revised March 31, 2025
Accepted May 13, 2025
AbstractChanges in the Na
+
and K
+
intracellular concentrations affect expression of the FOS gene. Here,
we obtained a genetic construct coding for the TurboGFP-dest1 protein under control of the human FOS
promoter (−549 to +155) and studied its expression in HEK293T cells exposed to monovalent metal cations.
Amplification of the FOS promoter sequence from genomic DNA was efficient only in the presence of Li
+
ions.
Incubation of cells with ouabain or in a medium containing Li
+
ions instead of Na
+
ions caused intracellular
accumulation of Na
+
and Li
+
ions, respectively. In addition, both stimuli increased the levels of endogenous
FOS mRNA and the average fluorescence intensity of TurboGFP-dest1 in transfected cells. The mRNA levels of
TurboGFP-dest1 were significantly higher than the FOS mRNA levels and were little affected by the stimuli.
DOI: 10.1134/S0006297925600371
Keywords: lithium, sodium, ouabain, FOS, transcription
* To whom correspondence should be addressed.
INTRODUCTION
The gradient of the Na
+
and K
+
ions between the
extracellular medium and cytoplasm of animal cells
is essential for the regulation of various cellular func-
tions and is strictly maintained mainly by the activity
of Na,K-ATPase [1]. However, some physiological and
pathological conditions might cause changes in the
intracellular concentrations of these cations, which
could affect expression of certain genes [2], in par-
ticular, the early response gene FOS coding for the
c-Fos transcription factor [3]. The baseline FOS expres-
sion is extremely low, but can increase dramatically
in response to external stimuli. Transcription of FOS
is controlled by multiple regulatory elements, includ-
ing serum-response element (SRE), cAMP-responsive
element (CRE), and activator protein 1 (AP-1) binding
site [4].
As demonstrated in various types of mammalian
cells, the effect of changes in the Na
+
i
/K
+
i
ratio on the
gene expression is not mediated by the Ca
2+
-depen-
dent signaling or alterations in the membrane po-
tential, intracellular pH, or cell volume [4-6]. Most of
these genes, including FOS, are early response genes
encoding transcription and translation factors [7].
The product of the FOS gene is the AP-1 family pio-
neer transcription factor c-FOS involved in the chro-
matin remodeling [8]. The study aimed at establishing
the nature of the Na
+
i
/K
+
i
-sensitive regulation of FOS
expression have shown that the promoter regulatory
elements, such as SRE, CRE, and AP-1 binding site, are
not involved in the transcriptional activation of gene
expression driven by the Fos promoter in response to
ouabain (specific inhibitor of Na,K-ATPase) [5]. By us-
ing genetic constructs carrying the human FOS gene
with deletions in the promoter, Nakagawa et al. [9]
were able to identify two sequences responsible for
the ouabain-induced activation of transcription: the
SRE element and a region between −222 and −123 bp
upstream of the transcription start site (TSS) [9]. How-
ever, the authors did not investigate the mechanism
of such regulation. It is important to note that ac-
cording to the bioinformatic analysis, the identified
region (−222 to −123) contained a sequence potentially
capable of forming a G-quadruplex. G-quadruplexes
are non-canonical secondary DNA structures formed
FOS PROMOTER IS HYPERACTIVATED OUTSIDE OF GENOME CONTENT 583
BIOCHEMISTRY (Moscow) Vol. 90 No. 5 2025
by guanine-rich sequences [10] that can bind monova-
lent cations. The ability of monovalent metal ions to
stabilize G-quadruplexes decreases in the following or-
der: K
+
>Rb
+
>Na
+
>Cs
+
>Li
+
[11]. The presence of G-qua-
druplexes in the coding sequences and promoters of
some Na
+
i
/K
+
i
-sensitive genes, including FOS, was pre-
dicted [12]. We hypothesize that these structures may
be involved in the Na
+
i
/K
+
i
-dependent regulation of
transcription.
To further investigate the role of G-quadruplexes
in the regulation of FOS transcription, we obtained a
genetic construct encoding TurboGFP-dest1 (a desta-
bilized variant of TurboGFP) under control of the hu-
man FOS promoter and studied the effects of ouabain
and Li
+
ions on the TurboGFP-dest1 expression in
transfected HEK293T cells. Introduction of plasmid
DNA encoding TurboGFP-dest1 into the cells caused
Na
+
i
accumulation, resulting in the hyperactivation of
the human FOS promoter (−549 to +155  bp relatively
to the TSS) and reduction of its sensitivity to monova-
lent metal cations.
MATERIALS AND METHODS
Genetic constructs. pFOS-TurboGFP and pR-
PLP0-TurboGFP reporter vectors were constructed
that encoded TurboGFP-dest1 under control of the FOS
or RPLP0 promoters, respectively. The RPLP0 promot-
er was chosen as a control because RPLP0 belongs to
housekeeping genes and its promoter is not sensitive
to changes in the Na
+
i
/K
+
i
ratio, as well as lacks pre-
dicted G-quadruplexes. The promoter regions of FOS
(−549 to +155 bp) and RPLP0 (−700 to +295 bp) were
amplified by PCR with an Encyclo Plus PCR kit (Euro-
gen, Russia) using genomic DNA from HeLa cells as a
template and primers carrying HindIII and AsiGI re-
striction sites (Online Resource 1). The FOS promoter
was amplified in a buffer proposed by Chashchina [13]
in which KCl was replaced by LiCl (60 mM Tris-HCl,
pH 9.5, 20 mM LiCl, 2.5 mM MgCl
2
, 5% DMSO). PCR
products were analyzed by sequencing. The FOS and
RPLP0 promoters were cloned into the promoterless
peTurboGFP-PRL-dest1 vector (Eurogen) using HindIII
and AsiGI restriction endonucleases (SibEnzyme, Rus-
sia). Full sequences and maps of the used plasmids
are presented in the Online Resource 1.
Culturing and transfection of HEK293T cells.
HEK293T cells were cultured in Dulbecco’s Modi-
fied Eagle Medium (DMEM; PanEco, Russia) con-
taining 4.5  g/L glucose, 0.58  g/L glutamine (PanEco,
Russia), 10% fetal bovine serum (FBS) (Cytiva, USA),
100 unit/mL streptomycin and penicillin (Life Tech-
nologies, USA) at 37°C in a humidified atmosphere
with 5%  CO
2
. Cell viability was determined using
alamarBlue reagent (Thermo Fisher Scientific, USA)
according to the manufacturers recommendations.
One day before the transfection, the cells were seeded
in 24-well plates (10
5
cells in 500μl of culture medium
per well) pre-coated with poly-D-lysine (MP Biomed-
icals, USA) and cultured for 24 h. The next day, 2 h
before the transfection, the cells were placed in the
culture medium of the same composition but without
antibiotics (500μl per well), and then transfected with
a mixture of 0.8 μg of plasmid DNA (pFOS-TurboGFP
or pRPLP0-TurboGFP) and 2 μl of Lipofectamine 2000
(Invitrogen, USA) in 50 μl of Opti-MEM (Gibco, USA)
per well and incubated for 24 h at 37°C in a hu-
midified atmosphere and 5%  CO
2
. The medium was
then replaced with low-serum DMEM (4.5  g/L glucose,
0.58  g/L glutamine, 0.1% FBS) and incubated for 24 h.
Cell treatment. For studying the effect of ouabain
on the Na
+
i
/K
+
i
ratio, the cells were incubated in the
presence of 1μM ouabain for 3h. For the intracellular
accumulation of Li
+
ions, the cells were incubated for
5 h in the Li-medium containing 10 mM HEPES-LiOH
(pH  7.4), 130  mM  LiCl, 5.2  mM  KCl, 1  mM  CaCl
2
,
0.5  mM  MgCl
2
, 0.4  mM  MgSO
4
, 0.3  mM KH
2
PO
4
, and
6 mM glucose; Na-medium of the same composition,
except containing 130 mM NaCl instead of 130 mM
LiCl, was used as a control (the composition of the
Na-medium mimicked the inorganic salt composition
of DMEM).
Assessment of the intracellular content of
Na
+
, K
+
, and Li
+
by atomic absorption spectrome-
try. The intracellular ion content was determined ac-
cording to the previously described method [14] with
some modifications. Briefly, at the end of incubation,
the plates were placed on ice. The medium was dis-
carded, after which the cells were washed three times
with 3 ml of ice-cold 0.1  M  MgCl
2
. Next, 1.5 ml of 5%
trichloroacetic acid (TCA) was added to the wells and
incubated for several hours at 4°C. The contents of
the wells were scraped off, transferred into microcen-
trifuge tubes, and centrifuged for 10 min at 18,000g.
The supernatant was collected into separate tubes,
the residual TCA was aspirated, and the precipitate
was dissolved in 0.1 M NaOH containing 0.1% sodi-
um deoxycholate. The amount of protein was deter-
mined by the Lowry method [15]. The intracellular
content of Na
+
, Li
+
, and K
+
ions was measured by
atomic absorption spectrometry using a Kvant-2m1
spectrometer (Kortek, Russia) in a propane-air mix-
ture at 589.6, 670.8, and 766.5 nm, respectively. NaCl
(0.05-2  mg/L  Na
+
), LiCl  (0.05-2  mg/L  Li
+
), and KCl
(0.5-2  mg/L  K
+
) solutions were used as standards.
The contents of Na
+
, Li
+
, and K
+
in each sample were
normalized to the protein content.
Laser scanning confocal microscopy and im-
age analysis. HEK293T cells were transfected with
pFOS-TurboGFP or pRPLP0-TurboGFP and subject-
ed to experimental treatments as described above.
GORBUNOV et al.584
BIOCHEMISTRY (Moscow) Vol. 90 No. 5 2025
Cell microscopy was performed with an Olympus
IX83P2ZF microscope (Japan) in the confocal imag-
ing mode (diameter of confocal membrane aperture,
130 μm) at 37°C in 5% CO
2
in a live-cell incubation
chamber (Tokai Hit, Japan) using a 10× objective with
a numerical aperture of 0.4. Fluorescence emission
was excited with a 488-nm laser (absorption max-
imum of turboGFP-dest1, 482 nm) and registered at
490-540nm. The images acquired were processed with
the Fiji/ImageJ 1.54f program. Individual cells were se-
lected using “masks” by setting a critical intensity lev-
el to visually distinguish them from the background,
and the average fluorescence intensity within the se-
lected cells was calculated.
Analysis of pFOS-TurboGFP and pRPLP0-
TurboGFP transcription by qPCR. The plates with
the cells were placed on ice, the cells were washed
with 1 ml of ice-cold Ca
2+
- and Mg
2+
-free Hank’s salt
solution, and lysed by adding 400μl of Trizol reagent
(Invitrogen) to each well. The lysates were treated
with a chloroform-mixture; the aqueous phase was
collected and used for RNA isolation with a Qiagen
RNeasy kit (Netherlands). Reverse transcription was
performed with an ImProm-II™ Reverse Transcription
System kit (Promega, USA) according to the manufac-
turers recommendations (0.5 μg of isolated RNA per
reaction). qPCR was performed with a Bio-Rad Real-
Time PCR System (Bio-Rad, USA) in the following re-
gime: denaturation at 95°C for 5 min followed by 40
cycles of denaturation at 95°C for 10s, primer anneal-
ing at 58°C for 10 s, and elongation at 72°C for 20 s.
The primers were selected using the BLAST NCBI da-
tabase and synthesized by Syntol (Russia). The primer
sequences are given in the Online Resource 1. Gene
expression levels were calculated by the ∆∆Ct method
using the RPLP0 gene as a reference [16]. The average
level of FOS expression in control samples was taken
as 100%. PCR products were analyzed by sequencing.
Statistical analysis. The Origin software pack-
age (OriginLab Corporation, USA) was used for statis-
tical data processing. The samples were tested for the
normality of distribution using the Shapiro–Wilk test.
To compare two independent groups, the Student’s
t-test for normally distributed samples and the Mann–
Whitney U-test for non-normally distributed samples
were used. Multiple comparisons for normally distrib-
uted data were performed using the one-factor analy-
sis of variance (ANOVA) followed by the Tukey’s test.
The differences were considered statistically signifi-
cant at p < 0.05.
RESULTS AND DISSCUSSION
To investigate the mechanism of Na
+
i
/K
+
i
-depen-
dent regulation of transcription, we constructed the
pFOS-TurboGFP and pRPLP0-TurboGFP reporter vec-
tors encoding TurboGFP-dest1 under control of the
human FOS promoter (−549 and +155) and RPLP0 pro-
moter (−700 and +295bp, no predicted G-quadruplex-
es), respectively (Fig. 1a). The sequences of the FOS
and RPLP0 promoters were amplified using genom-
ic DNA from HeLa cells as a template. Importantly,
we were able to amplify the RPLP0 promoter using
commercial buffer solution for Encyclo polymerase,
while amplification of the FOS promoter under these
conditions was unsuccessful. For this reason, we used
a buffer for GC-rich sequences for the FOS promoter
amplification; however, the length of the resulting PCR
product was shorter than the expected one by 78 bp
(Fig.1b), presumably, due to the presence of G-quadru-
plexes in the FOS promoter sequence. G-quadruplexes
are known to disrupt PCR in the presence of K
+
ions,
and most commercial buffers for DNA polymerases
contain K
+
ions. Hence, we used a modified buffer
solution suggested by Chashchina et al. [13] contain-
ing Li
+
ions instead of K
+
ions, to successfully amplify
the FOS promoter sequence.
Next, we evaluated the effect of transfection itself
on the cell intracellular ion composition. Figure 2a
shows that lipofection with pFOS-TurboGFP or
pRPLP0-TurboGFP (but not the treatment with Lipo-
fectamine 200 alone) was accompanied by a 40-60%
increase in the Na
+
i
concentration even 48 h after
transfection. There are at least two factors that may be
responsible for this effect. First, ion transport could be
affected by the induction of interferon expression by
foreign AT-rich DNA via an RNA polymerase III-depen-
dent pathway [17]. On the other hand, expression of
TurboGFP-dest1 could influence various intracellular
functions. For example, TurboGFP-dest1 was found to
affect the NF-κB signaling pathway in T cells, HEK293
cells, and HeLa cells [18]. It is possible that TurboG-
FP-dest1 expression can have an impact on the ion
transport as well. However, the precise mechanisms
of this phenomenon remain unclear.
To compare the effects of Li
+
and Na
+
ions on
the FOS promoter activity, the cells were incubated in
DMEM containing 0.1% FBS in the presence of 1 μM
ouabain or in the Li-medium containing 135 mM Li
+
.
Figure 2b shows that incubation in the presence of
1 μM ouabain for 3 h caused an 11.5-fold decrease
in the K
+
i
concentration and an 11.8-fold increase in
the Na
+
i
concentration in the cells. Incubation in the
Li-medium for 5  h led to the intracellular accumula-
tion of Li
+
(to 740  nmol/mg protein), while the con-
centration of Na
+
i
and K
+
i
decreased by 85 and 90%,
respectively, compared to the cells incubated in the
Na-medium (Fig.2c). Exposure to ouabain or Li-medi-
um did not affect the cell viability (data not shown),
although the morphology of cells in the Li-medium
was slightly altered (Fig. 2d).
FOS PROMOTER IS HYPERACTIVATED OUTSIDE OF GENOME CONTENT 585
BIOCHEMISTRY (Moscow) Vol. 90 No. 5 2025
Fig. 1. Reporter vectors used to study the FOS promoter activity. a)pFOS-TurboGFP and pRPLP0-TurboGFP vectors en-
coding TurboGFP-dest1 under control of the FOS promoter (−549 to +155bp) and RPLP0 promoter (−700 to +295bp),
respectively. b) Electrophoresis of the FOS promoter amplification products obtained by PCR with different buffer
solutions in 1.5% agarose gel stained with ethidium bromide: 1,commercial buffer solution for Encyclo polymerase;
2,commercial buffer solution for GC-rich sequences; 3,modified buffer solution with K
+
ions substituted by Li
+
ions.
The amplified DNA fragments contained HindIII and AsiGI restriction nuclease sites for subsequent cloning; expected
length of PCR product, 728 bp; TSS, transcription start site.
To assess the activity of the FOS promoter in trans-
fected cells, we measured the fluorescence intensity
of TurboGFP-dest1 using confocal microscopy, as well
as determined the levels of TurboGFP-dest1 mRNA by
qPCR. Figure 3 shows that the median fluorescence
intensity in the cells transfected with pFOS-TurboGFP
and incubated in the presence of 1 μM ouabain or
Li
+
(Li-medium) increased by 21 and 11%, respective-
ly, while no significant changes in the fluorescence
intensity were observed for the cells transfected with
pRPLP0-TurboGFP. However, contrary to our expecta-
tions, the fluorescence intensity of cells transfected
with pFOS-TurboGFP and pRPLP0-TurboGFP was very
similar, despite the fact that the levels of endoge-
nous RPLP0 mRNA (Ct  =  18.4  ±  0.12) were orders of
magnitude higher compared to the levels of endog-
enous FOS mRNA (Ct  =  28.3  ±  0.07). No significant
differences were found between the TurboGFP-dest1
gene transcription levels (control vs. ouabain-treated
cells and Na-medium vs. Li-medium samples) in case
of both vectors. Incubation of cells transfected with
pFOS-TurboGFP in the presence of ouabain and in
the Li-medium increased the content of endogenous
FOS mRNA 17 and 10 times, respectively (Fig.  4).
It is important to note that introduction of plas-
mid DNA into the cells was accompanied by the in-
crease in the content of Na
+
i
ions, but had no effect
on the amount of endogenous FOS mRNA (data not
shown). This indicates that the elevated expression
of TurboGFP-dest1 under the FOS promoter was not
related to the experimental conditions, but rather re-
sulted from the dysregulation of the FOS promoter
activity and/or the presence of the TurboGFP-dest1
coding sequence in the used constructs. The levels of
TurboGFP-dest1 mRNA were 1000-3000 times higher
than the levels of endogenous FOS mRNA in all ex-
perimental and control samples. We believe that these
results indicate overactivation of the FOS promoter
in the pFOS-TurboGFP vector, so that its activity was
comparable to that of promoters of the housekeep-
ing genes (e.g., RPLP0). This might be the reason why
the observed effect of the experimental treatments
on the activity of the FOS promoter in the content
of the reporter vector was insignificant.
Taking into account the above data, we can con-
clude that the plasmid carrying the coding sequence
for TurboGFP-dest1 under control of the FOS promoter
is not suitable for studying the role of G-quadruplexes
GORBUNOV et al.586
BIOCHEMISTRY (Moscow) Vol. 90 No. 5 2025
Fig.  2. Effect of different experimental treatments on the intracellular content of monovalent metal cations in
HEK293T cells. a)Transfection of cells with the pFOS-TurboGFP and pRPLP0-TurboGFP vectors increased the content
of Na
+
i
by 60 and 40%, respectively, but had no significant effect on the content of K
+
i
. Transfection with Lipofect-
amine 2000 alone did not affect the Na
+
i
and K
+
i
content. b)Incubation of cells in the presence of 1μM ouabain for
3 h resulted in ~12-fold increase in the Na
+
i
content, but significantly decreased the content of K
+
i
. c) Incubated of
cells in the Li-medium (135 mM Li
+
, 5.5 mM K
+
) for 5 h led to a considerable intracellular accumulation of Li
+
(up
to 740 nmol/mg protein) and reduction in the Na
+
i
and K
+
i
content compared to cells incubated in the Na-medium
(135 mM Na
+
and 5.5 mM K
+
). d) Phase-contrast images of cells incubated in the presence of 1 μM ouabain for 3 h,
in Na-medium for 5 h, or in Li-medium for 5 h. The data are presented as individual experimental values (n =6-8)
and box plots with whiskers equal to 1.5 interquartile range. Statistically significant differences were identified
using the t-test or ANOVA followed by the Tukey’s test (*  p < 0.05).
FOS PROMOTER IS HYPERACTIVATED OUTSIDE OF GENOME CONTENT 587
BIOCHEMISTRY (Moscow) Vol. 90 No. 5 2025
Fig. 3. Expression of the TurboGFP-dest1 reporter protein under control of the FOS or RPLP0 promoters. a)Confocal
images of cells transfected with pFOS-TurboGFP and pRPLP0-TurboGFP. Two days after transfection, the cells were
incubated in the presence of 1 μM ouabain for 3 h, in Li-medium for 5 h, or in Na-medium for 5 h. Scale bars:
100 µm. b) Distribution of TurboGFP-dest1 mean fluorescence intensity in individual cells (n = 500-1000). Statisti-
cal analysis revealed a small but significant increase in the reporter protein fluorescence in response to ouabain
compared to the control (DMEM). A similar effect was observed in the cells incubated in the Li-medium compared
to the Na-medium. Statistically significant differences were detected using the Mann–Whitney U-test (* p < 0.05).
in the regulation of FOS expression by monovalent
cations. Apparently, it is necessary to create a genetic
construct that, in addition to the FOS promoter and
5′-UTR, would include the FOS-coding sequence and
the 3′-UTR rather than the reporter gene.
The effect of Na
+
i
/K
+
i
ratio on the FOS promot-
er activity has been studied before. Using a vector
containing the firefly luciferase gene under control
of the FOS 5′-UTR (−1264 to +103 bp), Haloui et al.
found that ouabain did not affect the expression of
luciferase [4], which is consistent with our results. In
another study, ouabain caused a significant increase
in the FOS transcription level (in this case, the full-
length human FOS gene was used) [9]. Based on the
results of these studies and our findings, we hypoth-
esize that regulatory elements in the FOS coding se-
quence may deactivate transcription during the cell
resting state. The first intron of the FOS gene con-
tains a strong transcription elongation block (+363 to
+387 bp) [19-22], which suppressed the basal expres-
sion of not only the FOS promoter, but also of the
“strong” metallothionein-2 gene promoter placed in
the vector together with the FOS coding sequence [21].
It is likely that the high activity of the FOS promot-
er in plasmid DNA can be explained by the absence
of regulatory elements normally located at a distance
from the FOS gene in the genome or by the differenc-
es in the modification of plasmid and genomic DNAs.
GORBUNOV et al.588
BIOCHEMISTRY (Moscow) Vol. 90 No. 5 2025
Fig.  4. Expression of endogenous FOS mRNA and
TurboGFP-dest1 mRNA under control of the FOS and
RPLP0 promoters in cells transfected with the pFOS-
TurboGFP or pRPLP0-TurboGFP vectors. Two days after
transfection, the cells were incubated in the presence
of 1 μM ouabain for 3 h, in Li-medium for 5 h, or in
Na-medium for 5 h. The levels of endogenous FOS and
TurboGFP-dest1 mRNAs were measured in cell lysates by
qPCR. The data are presented as geometric mean (GM)
with whiskers from GM×SD to GM/SD and individual ex-
perimental values (overlapping; n = 4). Statistical analy-
sis revealed a significant increase in the levels of endog-
enous FOS mRNA in response to ouabain and Li-medium
compared to the control and Na-medium, respective-
ly. Statistically significant differences were detected
using the t-test (* p < 0.05).
It is also important to take into account possible
differences in the post-transcriptional regulation of
the TurboGFP-dest1 and FOS expression, for exam-
ple, different stability of the reporter protein mRNA.
However, these assumptions need further investigation.
CONCLUSIONS
Using a genetic construct encoding TurboGFP-dest1
under control of the FOS promoter, we demonstrated
that ouabain increased the activity of the FOS promot-
er in HEK293T cells and caused intracellular accumu-
lation of Li
+
ions. The levels of the TurboGFP-dest1
gene expression in the cells transfected with the ob-
tained construct were extremely high and insensitive
to changes in the intracellular ion composition. It is
likely that repression of the FOS expression during
the cell resting state requires regulatory elements
located outside of the FOS promoter region. We also
found that lipofection of HEK293T cells with the ob-
tained genetic constructs resulted in the Na
+
i
accumu-
lation, which may be due to the activation of cellular
response to foreign DNA or a nonspecific effect of
TurboGFP-dest1 on the ion transport.
Supplementary information. The online version
contains supplementary material available at https://
doi.org/10.1134/S0006297925600371.
Contributions. E.A.K. developed the concept, su-
pervised the study, and edited the manuscript; A.M.G.,
D.A.F., and O.E.K. planned and performed experiments
and prepared the manuscript; O.D.L. developed the
study conception and edited the manuscript.
Funding. The study was conducted under the
State Assignment to the Lomonosov Moscow State Uni-
versity.
Ethics approval and consent to participate.
This work does not contain any studies involving hu-
man and animal subjects.
Conflict of interest. The authors of this work de-
clare that they have no conflicts of interest.
REFERENCES
1. Skou, J. C., and Esmann, M. (1992) The Na,K-
ATPase, J. Bioenerg. Biomembr., 24, 249-261, https://
doi.org/10.1007/BF00768846.
2. Pollack, L. R., Tate, E. H., and Cook, J. S. (1981) Turn-
over and regulation of Na-K-ATPase in HeLa cells,
Am. J. Physiol. Cell Physiol., 241, C173-C183, https://
doi.org/10.1152/ajpcell.1981.241.5.C173.
3. Koltsova, S. V., Trushina, Y., Haloui, M., Akimova,
O. A., Tremblay, J., Hamet, P., and Orlov, S. N. (2012)
Ubiquitous Na
+
i/K
+
i-sensitive transcriptome in mam-
malian cells: evidence for Ca
2+
i-independent exci-
tation-transcription coupling, PLoS One, 7, e38032,
https://doi.org/10.1371/journal.pone.0038032.
4. Haloui, M., Taurin, S., Akimova, O. A., Guo, D.-F.,
Tremblay, J., Dulin, N. O., Hamet,P., and Orlov, S. N.
(2007) [Na+]i-induced c-Fos expression is not mediat-
ed by activation of the 5′-promoter containing known
transcriptional elements, FEBS J., 274, 3557-3567,
https://doi.org/10.1111/j.1742-4658.2007.05885.x.
5. Taurin,S., Dulin, N. O., Pchejetski,D., Grygorczyk,R.,
Tremblay,J., Hamet,P., and Orlov, S.N. (2002) c-Fos ex-
pression in ouabain-treated vascular smooth muscle
cells from rat aorta: evidence for an intracellular-so-
dium-mediated, calcium-independent mechanism,
FOS PROMOTER IS HYPERACTIVATED OUTSIDE OF GENOME CONTENT 589
BIOCHEMISTRY (Moscow) Vol. 90 No. 5 2025
J.Physiol., 543, 835-847, https://doi.org/10.1113/jphysi-
ol.2002.023259.
6. Shiyan, A.A., Sidorenko, S.V., Fedorov,D., Klimanova,
E. A., Smolyaninova, L. V., Kapilevich, L. V., Gry-
gorczyk, R., and Orlov, S. N. (2019) Elevation of in-
tracellular Na
+
contributes to expression of early
response genes triggered by endothelial cell shrink-
age, Cell. Physiol. Biochem., 53, 638-647, https://
doi.org/10.33594/000000162.
7. Lopina, O.D., Fedorov, D.A., Sidorenko, S.V., Bukach,
O.V., and Klimanova, E.A. (2022) Sodium ions as reg-
ulators of transcription in mammalian cells, Biochem-
istry (Moscow), 87, 789-799, https://doi.org/10.1134/
S0006297922080107.
8. Wolf, B. K., Zhao, Y., McCray, A., Hawk, W. H., Deary,
L. T., Sugiarto, N. W., LaCroix, I. S., Gerber, S. A.,
Cheng, C., and Wang, X. (2023) Cooperation of chro-
matin remodeling SWI/SNF complex and pioneer fac-
tor AP-1 shapes 3D enhancer landscapes, Nat. Struct.
Mol. Biol., 30, 10-21, https://doi.org/10.1038/s41594-
022-00880-x.
9. Nakagawa, Y., Rivera, V., and Larner, A. C. (1992) A
role for the Na/K-ATPase in the control of human c-fos
and c-jun transcription, J.Biol. Chem., 267, 8785-8788,
https://doi.org/10.1016/S0021-9258(19)50347-7.
10. Papp,C., Mukundan, V.T., Jenjaroenpun,P., Winnerdy,
F.R., Ow, G.S., Phan, A.T., and Kuznetsov, V.A. (2023)
Stable bulged G-quadruplexes in the human genome:
identification, experimental validation and function-
alization, Nucleic Acids Res., 51, 4148-4177, https://
doi.org/10.1093/nar/gkad252.
11. Sen, D., and Gilbert, W. (1990) A sodium-potassium
switch in the formation of four-stranded G4-DNA, Na-
ture, 344, 410-414, https://doi.org/10.1038/344410a0.
12. Klimanova, E.A., Sidorenko, S.V., Abramicheva, P.A.,
Tverskoi, A.M., Orlov, S. N., and Lopina, O.D. (2020)
Transcriptomic changes in endothelial cells triggered
by Na,K-ATPase inhibition: a search for upstream
Na
+
i/K
+
i sensitive genes, Int. J. Mol. Sci., 21, 7992,
https://doi.org/10.3390/ijms21217992.
13. Chashchina, G. V., Beniaminov, A. D., and Kaluzhny,
D. N. (2019) Stable G-quadruplex structures of on-
cogene promoters induce potassium-dependent
stops of thermostable DNA polymerase, Biochem-
istry (Moscow), 84, 562-569, https://doi.org/10.1134/
S0006297919050109.
14. Fedorov, D. A., Sidorenko, S. V., Yusipovich, A. I.,
Bukach, O. V., Gorbunov, A. M., Lopina, O. D., and
Klimanova, E. A. (2022) Increased extracellular so-
dium concentration as a factor regulating gene ex-
pression in endothelium, Biochemistry (Moscow), 87,
489-499, https://doi.org/10.1134/S0006297922060013.
15. Lowry, O. H., Rosebrough, N. J., Farr, A. L., and
Randall, R.J. (1951) Protein measurement with the Fo-
lin phenol reagent, J. Biol. Chem., 193, 265-275, https://
doi.org/10.1016/S0021-9258(19)52451-6.
16. Livak, K. J., and Schmittgen, T. D. (2001) Analysis of
relative gene expression data using real-time quanti-
tative PCR and the 2
−ΔΔC
T
method, Methods, 25, 402-
408, https://doi.org/10.1006/meth.2001.1262.
17. Ablasser, A., Bauernfeind, F., Hartmann, G., Latz, E.,
Fitzgerald, K.A., and Hornung,V. (2009) RIG-I-depen-
dent sensing of poly(dA:dT) through the induction of
an RNA polymerase III-transcribed RNA intermediate,
Nat. Immunol., 10, 1065-1072, https://doi.org/10.1038/
ni.1779.
18. Koelsch, K. A., Wang, Y., Maier-Moore, J. S., Sawalha,
A. H., and Wren, J. D. (2013) GFP affects human T
cell activation and cytokine production following
in vitro stimulation, PLoS One, 8, e50068, https://
doi.org/10.1371/journal.pone.0050068.
19. Collart, M. A., Tourkine, N., Belin, D., Vassalli, P.,
Jeanteur, P., and Blanchard, J. M. (1991) c-fos gene
transcription in murine macrophages is modulated by
a calcium-dependent block to elongation in intron 1,
Mol. Cell. Biol., 11, 2826-2831, https://doi.org/10.1128/
MCB.11.5.2826.
20. Mechti, N., Piechaczyk, M., Blanchard, J. M.,
Jeanteur, P., and Lebleu, B. (1991) Sequence require-
ments for premature transcription arrest within the
first intron of the mouse c-fos gene, Mol. Cell. Biol.,
11, 2832-2841, https://doi.org/10.1128/mcb.11.5.2832-
2841.1991.
21. Coulon, V., Veyrune, J. L., Tourkine, N., Vié, A.,
Hipskind, R. A., and Blanchard, J. M. (1999) A novel
calcium signaling pathway targets the c-fos intragenic
transcriptional pausing site, J. Biol. Chem., 274, 30439-
30446, https://doi.org/10.1074/jbc.274.43.30439.
22. Yamada, T., Yamaguchi, Y., Inukai, N., Okamoto, S.,
Mura,T., and Handa,H. (2006) P-TEFb-mediated phos-
phorylation of hSpt5 C-terminal repeats is critical for
processive transcription elongation, Mol. Cell, 21, 227-
237, https://doi.org/10.1016/j.molcel.2005.11.024.
Publishers Note. Pleiades Publishing remains
neutral with regard to jurisdictional claims in published
maps and institutional affiliations. AI tools may have
been used in the translation or editing of this article.