ISSN 0006-2979, Biochemistry (Moscow), 2025, Vol. 90, No. 12, pp. 1985-1998 © The Author(s) 2025. This article is an open access publication.
Published in Russian in Biokhimiya, 2025, Vol. 90, No. 12, pp. 2093-2107.
1985
Succinate Confers Stronger Cytoprotection
in Kidney Cells than in Astrocytes Due
to Its More Efficient Involvement in Energy Metabolism
Marina I. Buyan
1,2
, Kseniia S. Cherkesova
1,3
, Anna A. Brezgunova
1
,
Irina B. Pevzner
1
, Nadezda V. Andrianova
1
, and Egor Y. Plotnikov
1,a
*
1
Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia
2
Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 119991 Moscow, Russia
3
Faculty of Biology, Lomonosov Moscow State University, 119991 Moscow, Russia
a
e-mail: plotnikov@belozersky.msu.ru
Received August 18, 2025
Revised October 30, 2025
Accepted November 3, 2025
AbstractBeing among the most metabolically active organs, brain and kidneys critically depend on efficient
energy metabolism, which primarily relies on oxidative phosphorylation. Acute pathological conditions asso-
ciated with a lack of metabolic substrates or their impaired utilization trigger signaling cascades that initiate
cell death and lead to poorly reversible organ dysfunction. One of the therapeutic approaches to correct the
energy deficit is administration of exogenous metabolites of the tricarboxylic acid cycle, such as succinate.
Inthis study, we investigated the effects of exogenous succinate on astrocytes and renal epithelial cells under
normal conditions and in serum deprivation-induced injury. Incubation with succinate increased the viability
of both cell types under normal and pathological conditions, but a more pronounced cytoprotective effect was
observed in renal cells. In injured renal epithelial cells, succinate increased mitochondrial membrane poten-
tial, a critical parameter for the maintenance of mitochondrial function and ATP generation. Comparison of
respiration and oxidative phosphorylation parameters in astrocytes and renal epithelial cells in the presence
of exogenous succinate revealed that epithelial cells exhibited a significantly higher respiratory control and
lower proton leak compared to astrocytes, which correlated with the higher cytoprotective activity of succi-
nate for kidney cells. Therefore, succinate showed a noticeable positive effect in the renal epithelium both
under normal conditions and after serum deprivation; however, in astrocytes, its effect was less pronounced.
This discrepancy might be related to a more efficient succinate utilization by the mitochondria in renal
cells and intrinsic bioenergetic differences between astrocytes and epithelial cells. Despite the clinical use of
succinate-containing drugs, the determination of optimal dosages and development of effective therapeutic
regimens require further investigation. Our results demonstrate cell type-dependent differences in the efficacy
of succinate, suggesting that its therapeutic potential may differ significantly depending on the organ-specific
bioenergetic and metabolic properties.
DOI: 10.1134/S0006297925602539
Keywords: brain, astrocytes, kidneys, apoptosis, energy substrates, succinate, mitochondria, oxidative phos-
phorylation
* To whom correspondence should be addressed.
INTRODUCTION
Severe pathological conditions, such as ischemic
disease (including myocardial infarction and ischemic
stroke), various forms of shock (septic, hypovolemic,
and others), and acute and chronic heart failure in
the decompensation stage provoke systemic and lo-
cal tissue hypoperfusion, resulting in strong depriva-
tion of key energy substrates in organs and tissues.
The ensuing energy crisis triggers pathophysiological
BUYAN et al.1986
BIOCHEMISTRY (Moscow) Vol. 90 No. 12 2025
cascades, the central event being dysregulation of
cellular energy metabolism. This disrupts the balance
between anabolic and catabolic processes and sup-
presses key metabolic and signaling pathways [1, 2],
leading to cell death. The functioning of organs with
a high basal energy demand, such as the brain, heart,
and kidneys, critically depends on a high rate of ad-
enosine triphosphate (ATP) synthesis, which makes
them particularly vulnerable to such disruptions in
metabolism. The kidneys are among the most met-
abolically active human organs [3, 4] due to a high
density of mitochondria in the tubular epithelial cells
[5, 6]. This ultrastructural feature is directly related
to the significant energy expenditure required for
ATP-dependent processes, primarily, active tubular re-
absorption of electrolytes, glucose, amino acids, and
water. The energy expenditure of the brain is also
very high. Despite its relatively small mass, the brain
consumes approximately 20% of all oxygen and 25%
of glucose utilized by the human body at rest [7, 8].
High metabolic demands are characteristic not only
of neurons, but astrocytes as well. Astrocytes play a
key role in the neurometabolic interactions; they ac-
tively capture glucose from the bloodstream, metabo-
lize it glycolytically to lactate, and supply this lactate
to neurons via the astrocyte-neuron lactate shuttle
as an energy substrate for oxidative phosphorylation
in the mitochondria [8-10]. Hence, astrocytes require
metabolic substrates both for their own needs and to
maintain the energy level in neurons.
A decreased supply of nutrients to tissues leads
to impaired mitochondrial function, in particular, in-
efficient oxidative phosphorylation. The damage to
the mitochondria not only reduces ATP production
but also induces oxidative stress, ultimately resulting
in cell death [2,  11]. It is commonly accepted that
the use of additional energy substrates can promote
metabolic adaptation and prevent energy collapse
by enabling rapid metabolism of tricarboxylic acid
(TCA) cycle intermediates and replenishment of re-
ducing equivalents utilized in the electron transport
chain [12]. Since mitochondrial dysfunction caused
by cell damage leads to extensive cell death, resto-
ration of normal mitochondrial function and mainte-
nance of oxidative phosphorylation and ATP synthesis
are considered promising therapeutic approaches in
acute conditions [13]. Succinic acid salts have long
been proposed as therapeutic agents for this purpose.
However, no systematic in  vitro studies of the effects
of succinate salts on the bioenergetic processes in
various cell types under physiological and stress con-
ditions have yet been fully conducted.
Here, we investigated the efficacy of disodium
succinate in preventing the death of astrocytes and
renal epithelial cells in the invitro model of cellular
stress induced by serum deprivation (a classic method
for inducing cellular damage). We also analyzed its
effects on the key parameters of mitochondrial func-
tion, including transmembrane potential and cellular
respiration. The aim of this study was to compare
the molecular mechanisms of succinate-mediated cy-
toprotection in astrocytes and renal tubular epithelial
cells under physiological and stress conditions, with
a focus on the mitochondrial function as a potential
basis for this protective effect.
MATERIALS AND METHODS
Cell cultures. Experiments were performed in
primary astrocyte cultures isolated from the brains
of neonatal rats and in cultured normal rat kidney
epithelial NRK-52E cells (CRL-1571; ATCC, USA). All
animal procedures were carried out in accordance
with the guidelines of the Federation of European
Laboratory Animal Science Associations (FELASA)
and the ARRIVE guidelines. Animal protocols were
approved by the Ethics Committee of the Belozer-
sky Institute of Physico-Chemical Biology, Lomonosov
Moscow State University (protocol 006-1/2/2024). For
the experiments, neonatal rats were humanely euth-
anized with CO
2
. The skull was then opened asepti-
cally, the brain hemispheres were removed, and the
meninges were excised. Brain tissue was transferred
to 0.05% trypsin-EDTA solution (BioinnLabs, Russia),
incubated for 30min, washed with trypsin-EDTA solu-
tion, and resuspended in complete culture medium
containing DMEM with 1  g/L glucose (BioinnLabs)
and F-12 (BioinnLabs) at a 1  :  1 ratio, supplemented
with stable L-glutamine (BioinnLabs) and 10% fetal
bovine serum (FBS) (HiMedia, India). The suspension
was transferred to a culture flask pre-coated with
poly-D-lysine (Sigma-Aldrich, USA). After cells formed
a monolayer, they were washed to remove the mi-
croglia by shaking at 135  rpm for 16  h. The medium
was then removed, and astrocytes were detached
with 0.05% trypsin-EDTA solution to be used in the
experiments. NRK-52E cells were cultured in a medi-
um containing F-12 and DMEM at a 1  :  1 ratio with
1  g/L glucose and stable L-glutamine supplemented
with 10%  FBS (HiMedia). The cells were passaged
using 0.05%  trypsin-EDTA solution. Both astrocytes
and renal epithelial cells were incubated until the
formation of 50% monolayer, after which the control
cells received normal culture medium, while experi-
mental cells were grown in the medium containing
disodium succinate (Sigma-Aldrich). Serum depriva-
tion was performed by incubating the cells in the
medium containing all components except FBS (with
or without succinate) for 48  h, after which the cells
were analyzed. For all treatments, medium pH was
monitored using phenol red.
EFFECTS OF SUCCINATE ON KIDNEY AND BRAIN CELLS 1987
BIOCHEMISTRY (Moscow) Vol. 90 No. 12 2025
Cell viability assay. Astrocytes and renal epithe-
lial cells were seeded in 96-well plates, cultured until
50% confluence, and treated with disodium succinate
(12.5, 25, 50, and 100  mM) under normal and serum
deprivation conditions. Cell viability was assessed us-
ing an MTT assay (Macklin, China). The MTT reagent
was dissolved at 5  mg/mL in DMEM/F-12 medium
without sodium bicarbonate (GE Healthcare, USA) and
incubated with cells for 1  h at 37°C. The cells were
then lysed with dimethyl sulfoxide (Macklin, China),
and the optical density of formazan was measured at
595 nm with a Zenyth 3100 multimodal plate reader
(Anthos Labtec, Austria). Cells incubated with water
for 24 h served as a negative control.
Cell death assessment. To study the effect of
succinate on cell death, the cells were stained with
propidium iodide (PI) according to the manufactur-
ers instructions (Lumiprobe, Russia). To visualize
live cells, the cells were stained with the vital dye
Calcein  AM as recommended by the manufacturer
(Invitrogen, USA) and imaged using an LSM  900 con-
focal microscope (Carl Zeiss, Germany) at the exci-
tation wavelengths of 543  nm for PI and 488  nm for
Calcein  AM, with emission recorded at 560-590  nm
for PI and 505-530  nm for Calcein  AM.
Mitochondrial transmembrane potential mea-
surement. To assess changes in the mitochondrial
membrane potential, astrocytes and kidney epitheli-
al cells were incubated with 50  mM disodium succi-
nate for 48 h and stained with 200  nM tetramethyl-
rhodamine ethyl ester (TMRE, Invitrogen) for 20  min
at 37°C. TMRE-stained cells were visualized with
LSM 900 microscope at the excitation wavelength of
543  nm with emission detected at 616-700  nm. Subse-
quent image processing was performed with the Fiji/
ImageJ software by analyzing the average TMRE flu-
orescence intensity in the cells.
Cellular respiration analysis. The oxygen con-
sumption rate (OCR) of astrocytes and kidney epithe-
lial cells was measured using a Seahorse XFp analyz-
er (Seahorse Bioscience, USA). The cells were seeded
in the wells of a Seahorse XF8 microplate. The fol-
lowing day before the experiment, the medium in the
wells was replaced with a Seahorse XF DMEM base
medium (Agilent, USA) supplemented with 2  mM glu-
tamine (BioinnLabs), and the cells were incubated for
one hour at 37°C. The plate was then placed in the an-
alyzer, and various stressors (all from Sigma-Aldrich)
were added to the indicated final concentrations in
the following order: 1)  disodium succinate, 50  mM;
2)  oligomycin, 4.5  μM; 3)  carbonyl cyanide-p-trifluo-
romethoxyphenylhydrazone (FCCP), 1  μM; 4)  a com-
bination of rotenone (2.5  μM), antimycin  A (4  μM),
and 2-deoxy-D-glucose (2-DG, 50  mM). In some experi-
ments, the substances were added in a different order:
1)  rotenone, 2.5  μM; 2)  disodium succinate, 50  mM;
3)  antimycin  A, 4  μM. The obtained OCR values were
normalized to the total protein concentration in each
well, measured using the bicinchoninic acid assay
(Sigma-Aldrich). After the experiment, the medium
from each well was collected in a separate microtube
and centrifuged for 10  min at 12,000g to precipitate
detached cells. Also, 10  μl of 2x RIPA buffer (Milli-
pore, Germany) was added to the bottom of each
well to lyse the attached cells. The resulting lysate
was combined with the pellet obtained by centrifug-
ing the culture medium from the same well, and the
pooled lysate was assessed for protein content. The
results of OCR measurements were analyzed using
the Seahorse XFp software (Wave 2.6.1, Seahorse Bio-
science), according to the recommendations for the
Seahorse XF Cell Mito Stress Test Kit (103015-100,
Agilent, USA). Basal respiration was calculated as the
difference between the OCR values after succinate
addition and after addition of a mixture of rotenone,
antimycin, and 2-DG. Respiration associated with ATP
production was calculated as the difference between
the OCR values after succinate addition and addition
of oligomycin. Proton leak was calculated as the dif-
ference in OCR values after addition of oligomycin
and addition of rotenone, antimycin, and 2-DG, while
respiratory control was assessed as the ratio of OCR
values after addition of FCCP to those after addition
of oligomycin. The contribution of succinate-based
respiration to the total oxygen consumption was as-
sessed as the ratio of OCR after succinate addition to
the basal OCR (before the addition of rotenone).
Statistical analysis of data was performed us-
ing the GraphPad Prism9 software (version9.5.1). All
values are presented as mean ± standard deviation
(SD). The data were tested for the normal distribu-
tion using the Shapiro–Wilk test. For experiments in-
cluding two experimental groups, the nonparametric
Mann–Whitney U  test or Student’s t-test for normally
distributed data were used. For the experiments in-
cluding four experimental groups, parametric vari-
ables were analyzed using the one-way analysis of
variance (ANOVA) with the Tukey’s multiple compar-
ison test; nonparametric variables were analyzed us-
ing the Kruskal–Wallis test with the Dunn’s multiple
comparison test.
RESULTS
Succinate promotes the viability of renal epi-
thelial cells and astrocytes under both physiologi-
cal and serum deprivation conditions. We evaluat-
ed the effect of a 48  h incubation with succinate on
the viability of renal epithelial cells and astrocytes
under both physiological and serum deprivation
conditions. Under physiological conditions, succinate
BUYAN et al.1988
BIOCHEMISTRY (Moscow) Vol. 90 No. 12 2025
Fig. 1. Effect of succinate on the viability of renal epithelial cells and astrocytes after 48h of incubation under physiological
(a, c) or serum deprivation (b, d) conditions. Cell viability was evaluated using the MTT assay. Data are presented as optical
density (OD) at 595 nm for renal epithelial cells (a, b) and astrocytes (c, d); dotted line (b, d) shows the viability of intact
cells; * p < 0.05; ** p < 0.01; *** p < 0.001; **** p < 0.0001 compared to the control succinate-free group.
significantly increased the viability of renal epithe-
lial cells at all tested concentrations (Fig.  1a). The
maximal effect was observed at the succinate con-
centration of 50 mM, which increased cell viability
more than two-fold compared to the untreated con-
trol. Serum deprivation for 48  h reduced the viability
of renal epithelial cells by 71%. Succinate markedly
enhanced cell viability upon serum deprivation; the
treatment with 50  mM succinate restored cell viabil-
ity to the levels comparable to those observed under
physiological conditions (Fig.  1b). However, 100  mM
succinate exhibited a toxic effect in serum-deprived
renal epithelial cells, whereas under normal condi-
tions, the same concentration produced a positive
effect (although less pronounced compared to other
tested concentrations) (Fig. 1, a, b).
Succinate also improved the viability of astro-
cytes, albeit to a lesser extent than that of renal ep-
ithelial cells. Under physiological conditions, the via-
bility of astrocytes was significantly enhanced at 12.5,
50, and 100  mM succinate (Fig.  1c) (1.25- and 1.5-fold
at 50 and 100  mM, respectively). Serum deprivation
reduced the viability of astrocytes by ~50%, where-
as the treatment with 100  mM succinate completely
abolished this detrimental effect (Fig.  1d). Overall,
succinate exerted a pro-survival effect in both cell
types, but it was more pronounced in renal epithe-
lial cells.
Succinate treatment reduces cell death in re-
nal epithelial cells, but not in astrocytes. The effect
of succinate on cell death in renal tubular epithelial
cells was further assessed using propidium iodide (PI)
and Calcein AM staining (Fig.  2). Serum deprivation
markedly increased the proportion of PI-positive cells
(Fig.  2, b, d-f), whereas 48-h incubation with succi-
nate significantly reduced the percentage of dead
cells (Fig. 2, b, e). In contrast, succinate failed to at-
tenuate the death of serum-starved astrocytes (Fig.  2,
d, f). These results supported the data of MTT as-
say, indicating that reduced viability during serum
deprivation was due to cell death, predominantly via
necrosis, as well as the ability of succinate to sub-
stantially improved the survival of renal tubular epi-
thelial cells under damaging conditions.
Succinate increases mitochondrial transmem-
brane potential in renal epithelial cells under
both physiological and serum deprivation condi-
tions. Mitochondrial transmembrane potential was
measured to evaluate the effect of succinate on the
status of mitochondria (Fig.  3). A significant increase
EFFECTS OF SUCCINATE ON KIDNEY AND BRAIN CELLS 1989
BIOCHEMISTRY (Moscow) Vol. 90 No. 12 2025
Fig. 2. The effect of succinate on the death of renal epithelial cells and astrocytes under physiological and serum depri-
vation conditions. Renal epithelial cells (a, b) and astrocytes (c, d) were stained with Calcein AM and PI after 48 hours of
incubation with succinate under physiological (a, c) and serum deprivation (b, d) conditions. Quantification of PI-positive
renal epithelial cells(e) and astrocytes(f) following succinate treatment; ****p<0.0001 compared to 0mM succinate within
the same experimental group; # p < 0.05 compared to the group exposed to physiological conditions.
BUYAN et al.1990
BIOCHEMISTRY (Moscow) Vol. 90 No. 12 2025
Fig. 3. Changes in the mitochondrial transmembrane potential of renal epithelial cells and astrocytes after succinate treat-
ment under physiological and serum deprivation conditions. Mean TMRE fluorescence in renal epithelial cells (a) and
astrocytes (b). Representative confocal images of TMRE-stained renal epithelial cells (c) and astrocytes (d). ** p < 0.01;
**** p < 0.0001 vs. 0 mM succinate under physiological conditions; # p < 0.05 vs. corresponding groups under normal
conditions.
in the transmembrane potential was observed in re-
nal epithelial cells (Fig.  3, a, c), but not in astrocytes
(Fig.  3, b, d), following succinate treatment under
control conditions. Serum deprivation for 48  h led to
a substantial decrease in the TMRE fluorescence in
both renal epithelial cells and astrocytes (Fig.  3). The
treatment with 50  mM succinate under serum depri-
vation conditions improved mitochondrial potential
in renal epithelial cells (Fig.  3, a, c), but failed to ex-
ert a similar effect in astrocytes (Fig.  3, b, d).
Respiratory response of kidney epithelial cells
and astrocytes to succinate treatment. The effects
of exogenous succinate on the respiration parameters
were studied to identify possible mechanisms under-
lying the differences in the cytoprotective effect of
succinate in kidney epithelial cells and astrocytes
(Fig.  4). To measure the OCR, the culture medium
was replaced with a reaction medium containing
no substrates for the respiration or glycolysis, after
which cells were analyzed using a Seahorse XF an-
alyzer after sequential addition of respiratory sub-
strates and inhibitors (see Materials and Methods).
The basal respiration rate was significantly higher in
astrocytes than in kidney epithelial cells (Fig.  4, a, b).
At the same time, renal epithelial cells showed lower
proton leak (Fig.  4d) and, therefore, higher respira-
tory control compared to astrocytes (Fig.  4e). These
findings suggest that the higher basal respiratory rate
of astrocytes may be due to a greater uncoupling of
respiration and oxidative phosphorylation. Never-
theless, both cell types utilized exogenous succinate
as a respiration substrate, as demonstrated by the
OCR increase following succinate addition after rote-
none-mediated inhibition of complexI (Fig.4f). Nota-
bly, succinate utilization was faster in epithelial cells,
as indicated by the kinetics of OCR response (Fig.  4f),
EFFECTS OF SUCCINATE ON KIDNEY AND BRAIN CELLS 1991
BIOCHEMISTRY (Moscow) Vol. 90 No. 12 2025
Fig. 4. The effect of exogenous disodium succinate on the respiration of astrocytes and renal epithelial cells. a)OCR curves
following addition of disodium succinate to the cells in the absence of complex I inhibition. b-e) Quantitative analysis of
mitochondrial respiration parameters calculated from the OCR changes after addition of respiration inhibitors and uncou-
pler: basal respiration(b), ATP-linked respiration(c), proton leak(d), and respiratory control(e). f)OCR curves showing the
response to disodium succinate following rotenone-induced complex I inhibition. g)The contribution of succinate-dependent
respiration to the total oxygen consumption in astrocytes and renal epithelial cells; *p < 0.05; *** p < 0.001.
BUYAN et al.1992
BIOCHEMISTRY (Moscow) Vol. 90 No. 12 2025
and its contribution to the oxygen consumption was
more significant than in astrocytes (Fig. 4g).
DISCUSSION
The brain and kidneys are among the organs
with the highest basal energy metabolism. Numer-
ous studies have shown that the optimal function-
ing of brain cells depends directly on the activity
of mitochondria, while mitochondrial dysfunction
contributes to or exacerbates brain pathologies [14].
Although neurons are very metabolically active cells,
astrocytes demonstrate a high glycolytic activity in
order to supply neurons with metabolic substrates
[15]. Nevertheless, mitochondria are considered an
important factor in the regulation of astrocyte func-
tions. It has been shown that during ischemic injury,
changes in the mitochondrial function of astrocytes
are closely associated with impaired brain homeosta-
sis, defects in glutamate and fatty acid metabolism,
Ca
2+
regulation, formation of reactive oxygen spe-
cies (ROS), and induction of inflammation [16]. The
kidneys are also among the most energy-consuming
organs, especially renal tubular cells [17, 18]. These
cells are particularly vulnerable to the mitochondrial
dysfunction, which can impair kidney function [19].
Despite the fact that bioenergetic characteristics
of various cell types can differ, mitochondrial dys-
function is a central component of many, if not all,
pathological conditions associated with a deficiency
of nutrient substrates and oxygen. An insufficient
supply of glycolytic and respiratory substrates dis-
rupts oxidative phosphorylation [20], leading to the
destabilization of electron transport chain complexes,
electron leakage, and excessive ROS generation [21].
This creates a vicious cycle, as oxidative stress po-
tentiates further mitochondrial damage, thus increas-
ing ROS production, critically reducing ATP synthesis
[22, 23], and ultimately activating intrinsic apoptotic
pathway, which results in cell death and, finally, or-
gan dysfunction. Therefore, strategies aimed at main-
taining energy homeostasis, such as replenishing me-
tabolite deficiencies with exogenous substrates, are
considered as a promising approach for correcting
mitochondrial dysfunction and preventing cell death.
In this study, serum (FBS) deprivation was used to in-
duce the damage and death of kidney and brain cells
as an in vitro model of cell damage and to examine
the possibility of metabolic correction of the observed
pathological changes with succinate. Low concentra-
tions or absence of FBS in the medium lead to the
activation of signaling molecules of the intrinsic mito-
chondrial apoptotic pathway that involves proteins of
the Bcl-2 family [11]. The absence of FBS also causes
ROS generation, DNA damage, and activation of apop-
totic processes [2]. After serum deprivation, cells con-
sume nutrients in particular, metabolites of the TCA
cycle (e.g., succinate) at a higher rate [24]. In the
acute phase of injury, maintaining a normal amount
of energy substrates is very important, as ROS can
impair the functioning of succinate dehydrogenase
and other respiratory chain complexes, which may
disrupt normal mitochondrial function even after the
nutrient supply restoration [25].
In this study, we investigated the effects of di-
sodium succinate on brain astrocytes and renal ep-
ithelial cells. Disodium succinate was used to avoid
acidification of the culture medium. Although succi-
nate is widely used in clinical practice, its organ-spe-
cific effects, optimal dosage, and effective therapeutic
regimens for various pathologies have not been fully
explored.
Mammalian cells take up exogenous succinate
primarily via sodium-dependent dicarboxylate trans-
porters (NaDCs) of the SLC13A family [26]. Two main
classes of NaDCs have been identified, that differ in
their affinity for succinate: low-affinity transporters
with the Michaelis constant (K
M
) for succinate of
~0.5  mM, and high-affinity transporters with a K
M
of ~25  μM [27]. These transporters are expressed in
various tissues, including astrocytes, neurons, kidney
cells, hepatocytes, and immune cells, and facilitate
the entry of succinate and other dicarboxylates into
cells, where these compounds are incorporated into
energy metabolism [28].
Disodium succinate increased the viability of re-
nal epithelial cells and astrocytes under both stan-
dard conditions and serum deprivation-induced stress.
However, the beneficial effect of succinate was more
pronounced in renal epithelial cells (Fig.  1). These
data are consistent with the previously obtained re-
sults showing that succinate improved mitochondrial
function in glial cells [29] and reduced neuroinflam-
mation in experimental intracerebral hemorrhage
[30]. One of the key biochemical markers of brain
injury is the lactate/pyruvate ratio in brain tissue.
An increase in this ratio reflects a shift towards an-
aerobic glycolysis and oxidative stress development.
Clinical administration of succinate-containing formu-
lations to patients with traumatic brain injury (TBI)
decreased the lactate/pyruvate ratio, indicating im-
proved redox balance and increased glucose utiliza-
tion efficiency [31-33]. This effect is thought to result
from the stimulation of the TCA cycle and oxidative
phosphorylation, leading to the increased pyruvate
levels and decreased pyruvate reduction to lactate
[33]. Furthermore, exogenous succinate improved
glutamatergic synaptic transmission in rat hippocam-
pal vital slices, which may be due to the improved
energy supply to presynaptic terminals [34]. In con-
trast to the nervous system, the effects of succinate
EFFECTS OF SUCCINATE ON KIDNEY AND BRAIN CELLS 1993
BIOCHEMISTRY (Moscow) Vol. 90 No. 12 2025
on the renal tissue have been studied much less.
However, succinate can also improve mitochondrial
function by reducing oxidative stress and replenish-
ing deficient energy substrate in the cases of renal
proximal tubule cell damage [35].
We demonstrated that exogenous succinate main-
tained cellular energy status by significantly increas-
ing the mitochondrial transmembrane potential. How-
ever, this effect was specific to renal epithelial cells
(Fig.  3). To understand why succinate had a more
pronounced positive effect on renal epithelial cells vs.
astrocytes, we compared the respiration parameters
under succinate energization (Fig. 4). The basal bio-
energetic characteristics of astrocytes and renal epi-
thelial cells differed significantly. Astrocytes exhibited
higher basal respiration rates, likely due to a higher
degree of mitochondrial uncoupling rather than to
a more active oxidative phosphorylation. Oligomy-
cin-sensitive respiration was similar between the two
cell types, whereas the proton leak and respiratory
control values (Fig.  4, d, e) indicated the occurrence
of the futile cycle in astrocytes. This might reflect the
reliance of astrocytes on glycolysis to fuel the astro-
cyte–neuron lactate shuttle, with an unchanged rate
of oxidative phosphorylation. Since the cells under the
experimental conditions had only succinate as a respi-
ratory substrate, it can be assumed that it was more
efficiently utilized by renal tubular cells. Oxygen con-
sumption values in the case of complex I inhibition
by rotenone following addition of succinate, showed
that succinate significantly increased the respiration
rate (Fig.  4f). This confirms the functional involve-
ment of exogenous succinate in the electron transport
chain. Moreover, the increase in the oxygen consump-
tion in tubular epithelial cells after succinate addition
occurred more rapidly than in astrocytes (Fig.  4f),
and the overall contribution of succinate-dependent
respiration in renal epithelial cells was significantly
higher than in astrocytes (Fig.  4g). The cell type-spe-
cific efficacy of disodium succinate can be explained
by both differences in the expression of dicarboxyl-
ate transport systems and higher activity of succinate
dehydrogenase in renal mitochondria. These features
make the energy metabolism of tubular epithelial
cells dependent on the availability of succinate [36].
In addition to its direct role in energy metab-
olism, the protective effect of succinate under the
damaging conditions may be mediated by several
other mechanisms, such as activation of the SUCNR1
receptor (GPR91) and inhibition of prolyl hydroxylas-
es (PHDs). The SUCNR1 receptor is widely expressed
in brain cells, in particular, astrocytes, microglia, and
neurons, as well as in renal tubular epithelial cells
[33, 37]. SUCNR1 is thought to be involved in tissue
adaptation to adverse conditions, including stimula-
tion of cell proliferation, migration, and angiogenesis
[38,  39]. Notably, the interaction of extracellular suc-
cinate with SUCNR1 can induce expression of SLC13A
family genes, which ensures succinate transport into
the cells [40]. This creates a positive feedback loop,
where extracellular succinate stimulates its own up-
take, enhancing both its metabolic and signaling ef-
fects. Moreover, after entering the cytosol, succinate
can inhibit 2-oxoglutarate-dependent dioxygenases,
primarily PHDs [41,  42], which are responsible for
the oxygen-dependent degradation of hypoxia-in-
ducible factor 1-alpha (HIF-1α). Inhibition of PHDs
stabilizes HIF-1α even under normoxic conditions,
enabling its nuclear translocation [43] and activa-
tion of genes involved in the adaptation to hypox-
ia and regulation of mitochondrial function [44].
In particular, this triggers the synthesis of cytochrome
oxidase subunit  2 (COX4-2), which replaces the con-
stitutive form COX4-1, thus increasing the efficien-
cy of complex IV at low oxygen concentrations [45].
It is worth noting that, in addition to succinate, oth-
er exogenous metabolites and their derivatives with
a higher bioavailability are actively studied. One ex-
ample is diacetoxymethyl succinate (NV118), which
shows a pronounced cytoprotective effect due to the
improvements in the mitochondrial metabolism in
the models of damage induced by carbon monox-
ide, cyanides, amiodarone, N-methylcarbamate, and
metformin [46-52]. Other alternative substrates (e.g.,
sodium fumarate) reduce cell death during hypoxia,
mostly by activating alternative pathways rather than
restoring mitochondrial ATP synthesis [53]. Dimeth-
yl fumarate has demonstrated the renoprotective ef-
fect against cyclosporine A- and lipopolysaccharide
(LPS)-induced toxicity, mediated by suppression of ox-
idative stress and inflammation, as well as enhance-
ment of the antioxidant defense [54, 55]. Dimethyl
fumarate also inhibited pyroptosis via modulation
of endoplasmic reticulum stress and the JAK2–STAT3
pathway in a model of contrast-induced nephropathy
[56] and improved mitochondrial function in the case
of di(2-ethylhexyl)phthalate toxicity [57]. The effects
of dimethyl fumarate are most likely explained by the
activation of the NRF2 pathway [58], which accounts
for its neuroprotective potential [59,  60], including
attenuation of neuroinflammation and neuronal cell
death after traumatic brain injury [61,  62]. Similarly,
monomethyl fumarate acts through NRF2, as it sup-
presses cell death and oxidative stress, and reduces
brain edema in a model of ischemic stroke [63-65].
Taken together, these data suggest that TCA cycle me-
tabolites and their derivatives have a considerable
therapeutic potential for ameliorating mitochondrial
dysfunction across a broad spectrum of pathologies
that require multiorgan protection. However, despite
their enhanced membrane permeability, ester deriva-
tives of dicarboxylic acids, including succinate, have
BUYAN et al.1994
BIOCHEMISTRY (Moscow) Vol. 90 No. 12 2025
limitations as therapeutic agents. In particular, com-
monly used alkyl esters, such as dimethyl succinate,
are hydrolyzed by cellular esterases with the release
of corresponding alcohols (e.g., methanol), which may
exert toxic effects. This highlights the critical need
for the development of alternative, safe formulations
capable of enhancing succinate bioavailability with-
out compromising the safety.
CONCLUSION
This study demonstrated a prominent cytopro-
tective potential of exogenous disodium succinate in
cells derived from two highly metabolically active or-
gans – the brain and kidneys – especially after injury.
Succinate significantly increased the viability of both
astrocytes and renal epithelial cells under both phys-
iological and stress conditions. However, its protec-
tive effect was more pronounced in kidney epithelial
cells than in astrocytes. This difference likely reflects
a more pronounced ability of succinate to modulate
the mitochondrial transmembrane potential in kidney
epithelial cells and to support oxidative phosphoryla-
tion when other metabolic substrates are limited, sug-
gesting more efficient utilization of succinate in renal
epithelial cells. These findings support the hypothesis
that succinate, a key TCA cycle intermediate, can al-
leviate cellular energy deficit and prevent cell death.
However, its effects appear to be cell type-dependent,
necessitating careful selection of target pathologies.
Therefore, succinate represents a particularly prom-
ising candidate for the nephroprotective strategies
in acute kidney injuries associated with bioenergetic
crisis.
Abbreviations
2-DG 2-deoxyglucose
ATP adenosine triphosphate
FBS fetal bovine serum
PHD prolyl hydroxylases
PI propidium iodide
ROS reactive oxygen species
TMRE tetramethylrhodamine ethyl ester
Contributions
N. V. Andrianova and E. Y. Plotnikov developed
the concept and supervised the study; M. I. Buyan,
K. S. Cherkesova, N. V. Andrianova, A. A. Brezgunova,
and I. B. Pevzner conducted the experiments; E. Y. Plot-
nikov, N. V. Andrianova, M. I. Buyan, K. S. Cherkeso-
va, A. A. Brezgunova, and I. B. Pevzner discussed
the study results; M. I. Buyan, K. S. Cherkesova, and
N. V. Andrianova wrote the original draft; E. Y. Plot-
nikov, A. A. Brezgunova, and I. B. Pevzner edited
themanuscript.
Funding
This work was supported by the Russian Science Foun-
dation (project no.24-75-10013).
Acknowledgments
The study was conducted on a Zeiss LSM900 confocal
microscope with support from the Moscow State Uni-
versity Development Program.
Ethics approval and consent to participate
Animal study protocols were reviewed and approved
by the Ethics Committee of the Belozersky Institute of
Physico-Chemical Biology, Lomonosov Moscow State
University (protocol no. 006-1/2/2024). All animal
manipulations were performed in accordance with
ARRIVE guidelines.
Conflict of interest
The authors of this work declare that they have
noconflicts of interest.
Open access
This article is licensed under a Creative Commons At-
tribution 4.0 International License, which permits use,
sharing, adaptation, distribution, and reproduction
in any medium or format, as long as you give appro-
priate credit to the original author(s) and the source,
provide a link to the Creative Commons license, and
indicate if changes were made. The images or other
third party material in this article are included in the
article’s Creative Commons license, unless indicated
otherwise in a credit line to the material. If materi-
al is not included in the article’s Creative Commons
license and your intended use is not permitted by
statutory regulation or exceeds the permitted use,
you will need to obtain permission directly from the
copyright holder. To view a copy of this license, visit
http://creativecommons.org/licenses/by/4.0/.
REFERENCES
1. Duann, P., and Lin, P.-H. (2017) Mitochondria dam-
age and kidney disease, Adv. Exp. Med. Biol.,
982, 529-551, https://doi.org/10.1007/978-3-319-55330-
6_27.
2. Gong, M., Li, Z., Zhang, X., Liu, B., Luo, J., Qin, X.,
and Wei,Y. (2021) PTEN mediates serum deprivation-
induced cytotoxicity in H9c2 cells via the PI3K/AKT
signaling pathway, Toxicol. In Vitro, 73, 105131,
https://doi.org/10.1016/j.tiv.2021.105131.
3. Forbes, J. M. (2016) Mitochondria-power players
in kidney function? Trends Endocrinol. Metab.,
27, 441-442, https://doi.org/10.1016/j.tem.2016.05.002.
4. Hoenig, M. P., and Zeidel, M. L. (2014) Homeostasis,
the milieu intérieur, and the wisdom of the nephron,
EFFECTS OF SUCCINATE ON KIDNEY AND BRAIN CELLS 1995
BIOCHEMISTRY (Moscow) Vol. 90 No. 12 2025
Clin. J. Am. Soc. Nephrol., 9, 1272-1281, https://doi.org/
10.2215/CJN.08860813.
5. Wang, Z., Ying, Z., Bosy-Westphal, A., Zhang, J.,
Schautz, B., Later, W., Heymsfield, S. B., and Müller,
M. J. (2010) Specific metabolic rates of major organs
and tissues across adulthood: evaluation by mech-
anistic model of resting energy expenditure, Am.
J. Clin. Nutr., 92, 1369-1377, https://doi.org/10.3945/
ajcn.2010.29885.
6. Ho, H.-J., and Shirakawa, H. (2022) Oxidative stress
and mitochondrial dysfunction in chronic kid-
ney disease, Cells, 12, 88, https://doi.org/10.3390/
cells12010088.
7. Cunnane, S.C., Trushina,E., Morland,C., Prigione,A.,
Casadesus, G., Andrews, Z. B., Beal, M. F., Bergersen,
L. H., Brinton, R. D., de la Monte, S., Eckert, A.,
Harvey, J., Jeggo, R., Jhamandas, J. H., Kann, O., la
Cour, C.M., Martin, W.F., Mithieux,G., Moreira, P. I.,
et al. (2020) Brain energy rescue: an emerging ther-
apeutic concept for neurodegenerative disorders of
ageing, Nat. Rev. Drug Discov., 19, 609-633, https://
doi.org/10.1038/s41573-020-0072-x.
8. Song, N., Mei,S., Wang, X., Hu, G., and Lu, M. (2024)
Focusing on mitochondria in the brain: from biology
to therapeutics, Transl. Neurodegener., 13, 23, https://
doi.org/10.1186/s40035-024-00409-w.
9. Pellerin,L., and Magistretti, P.J. (1994) Glutamate up-
take into astrocytes stimulates aerobic glycolysis: a
mechanism coupling neuronal activity to glucose uti-
lization, Proc. Natl. Acad. Sci. USA, 91, 10625-10629,
https://doi.org/10.1073/pnas.91.22.10625.
10. Kasischke, K.A., Vishwasrao, H.D., Fisher, P.J., Zipfel,
W.R., and Webb, W.W. (2004) Neural activity triggers
neuronal oxidative metabolism followed by astrocytic
glycolysis, Science, 305, 99-103, https://doi.org/10.1126/
science.1096485.
11. Qiu, T., He, Y.-Y., Zhang,X., and Ma, X.-L. (2018) Nov-
el role of ER stress and mitochondria stress in se-
rum-deprivation induced apoptosis of rat mesenchy-
mal stem cells, Curr. Med. Sci., 38, 229-235, https://
doi.org/10.1007/s11596-018-1870-9.
12. Andrianova, N. V., Buyan, M. I., Brezgunova, A. A.,
Cherkesova, K. S., Zorov, D. B., and Plotnikov, E. Y.
(2025) Hemorrhagic shock and mitochondria: patho-
physiology and therapeutic approaches, Int. J. Mol.
Sci., 26, 1843, https://doi.org/10.3390/ijms26051843.
13. Nguyen, H., Zerimech, S., and Baltan, S. (2021) As-
trocyte mitochondria in white-matter injury, Neu-
rochem. Res., 46, 2696-2714, https://doi.org/10.1007/
s11064-021-03239-8.
14. Belenguer, P., Duarte, J. M. N., Schuck, P. F., and
Ferreira, G. C. (2019) Mitochondria and the brain:
bioenergetics and beyond, Neurotox. Res., 36, 219-
238, https://doi.org/10.1007/s12640-019-00061-7.
15. Bonvento, G., and Bolaños, J.P. (2021) Astrocyte-neu-
ron metabolic cooperation shapes brain activity,
Cell Metab., 33, 1546-1564, https://doi.org/10.1016/j.
cmet.2021.07.006.
16. Zhang, L.-Y., Hu, Y.-Y., Liu, X.-Y., Wang, X.-Y., Li, S.-C.,
Zhang, J.-G., Xian, X.-H., Li, W.-B., and Zhang, M.
(2024) The role of astrocytic mitochondria in the
pathogenesis of brain ischemia, Mol. Neurobiol., 61,
2270-2282, https://doi.org/10.1007/s12035-023-03714-z.
17. Bhargava, P., and Schnellmann, R. G. (2017) Mito-
chondrial energetics in the kidney, Nat. Rev. Nephrol.,
13, 629-646, https://doi.org/10.1038/nrneph.2017.107.
18. Ma, H., Guo, X., Cui, S., Wu, Y., Zhang, Y., Shen, X.,
Xie, C., and Li, J. (2022) Dephosphorylation of AMP-
activated protein kinase exacerbates ischemia/reper-
fusion-induced acute kidney injury via mitochon-
drial dysfunction, Kidney Int., 101, 315-330, https://
doi.org/10.1016/j.kint.2021.10.028.
19. Huang, R., Zhang, C., Xiang, Z., Lin, T., Ling, J., and
Hu, H. (2024) Role of mitochondria in renal isch-
emia-reperfusion injury, FEBS J., 291, 5365-5378,
https://doi.org/10.1111/febs.17130.
20. Liu,F., Lu,J., Manaenko,A., Tang,J., and Hu,Q. (2018)
Mitochondria in ischemic stroke: new insight and
implications, Aging Dis., 9, 924-937, https://doi.org/
10.14336/AD.2017.1126.
21. Jurcau, A., and Ardelean, A. I. (2022) Oxidative
stress in ischemia/reperfusion injuries following
acute ischemic stroke, Biomedicines, 10, 574, https://
doi.org/10.3390/biomedicines10030574.
22. Zhang, X., Agborbesong, E., and Li, X. (2021) The
role of mitochondria in acute kidney injury and
chronic kidney disease and its therapeutic poten-
tial, Int. J. Mol. Sci., 22, 11253, https://doi.org/10.3390/
ijms222011253.
23. Lan,R., Geng,H., Singha, P.K., Saikumar,P., Bottinger,
E. P., Weinberg, J. M., and Venkatachalam, M. A.
(2016) Mitochondrial pathology and glycolytic shift
during proximal tubule atrophy after ischemic AKI,
J. Am. Soc. Nephrol., 27, 3356-3367, https://doi.org/
10.1681/ASN.2015020177.
24. Lorenz, M., Fritsche-Guenther, R., Bartsch, C.,
Vietzke,A., Eisenberger,A., Stangl,K., Stangl,V., and
Kirwan, J. A. (2023) Serum starvation accelerates in-
tracellular metabolism in endothelial cells, Int. J. Mol.
Sci., 24, 1189, https://doi.org/10.3390/ijms24021189.
25. Chouchani, E.T., Pell, V.R., Gaude,E., Aksentijević,D.,
Sundier, S.Y., Robb, E.L., Logan,A., Nadtochiy, S.M.,
Ord, E. N. J., Smith, A. C., Eyassu, F., Shirley, R., Hu,
C.-H., Dare, A. J., James, A. M., Rogatti, S., Hartley,
R.C., Eaton,S., Costa, A.S.H., etal. (2014) Ischaemic
accumulation of succinate controls reperfusion inju-
ry through mitochondrial ROS, Nature, 515, 431-435,
https://doi.org/10.1038/nature13909.
26. Pajor, A.M. (2014) Sodium-coupled dicarboxylate and
citrate transporters from the SLC13 family, Pflugers
Arch., 466, 119-130, https://doi.org/10.1007/s00424-
013-1369-y.
BUYAN et al.1996
BIOCHEMISTRY (Moscow) Vol. 90 No. 12 2025
27. Pajor, A. M. (2000) Molecular properties of sodium/
dicarboxylate cotransporters, J. Membr. Biol., 175, 1-8,
https://doi.org/10.1007/s002320001049.
28. Yodoya, E., Wada, M., Shimada, A., Katsukawa, H.,
Okada,N., Yamamoto,A., Ganapathy,V., and Fujita,T.
(2006) Functional and molecular identification of sodi-
um-coupled dicarboxylate transporters in rat prima-
ry cultured cerebrocortical astrocytes and neurons,
J. Neurochem., 97, 162-173, https://doi.org/10.1111/
j.1471-4159.2006.03720.x.
29. Giorgi-Coll, S., Amaral, A. I., Hutchinson, P. J. A.,
Kotter, M.R., and Carpenter, K.L.H. (2017) Succinate
supplementation improves metabolic performance
of mixed glial cell cultures with mitochondrial dys-
function, Sci. Rep., 7, 1003, https://doi.org/10.1038/
s41598-017-01149-w.
30. Wang, Y., Huang, C., Wang, X., Cheng, R., Li, X.,
Wang, J., Zhang, L., Li, F., Wang, H., Li, X., Li, Y.,
Xia,Y., Cheng,J., Pan,X., Jia,J., and Xiao, G.-D. (2025)
Succinate activates uncoupling protein 2 to suppress
neuroinflammation and confer protection following
intracerebral hemorrhage, Antioxid. Redox Signal.,
42, 687-710, https://doi.org/10.1089/ars.2024.0573.
31. Jalloh, I., Helmy, A., Howe, D. J., Shannon, R. J.,
Grice, P., Mason, A., Gallagher, C. N., Stovell, M. G.,
van der Heide, S., Murphy, M. P., Pickard, J. D.,
Menon, D. K., Carpenter, T. A., Hutchinson, P. J., and
Carpenter, K.L. (2017) Focally perfused succinate po-
tentiates brain metabolism in head injury patients,
J. Cereb. Blood Flow Metab., 37, 2626-2638, https://
doi.org/10.1177/0271678X16672665.
32. Khellaf,A., Garcia, N.M., Tajsic, T., Alam, A., Stovell,
M. G., Killen, M. J., Howe, D. J., Guilfoyle, M. R.,
Jalloh,I., Timofeev,I., Murphy, M.P., Carpenter, T.A.,
Menon, D.K., Ercole,A., Hutchinson, P. J., Carpenter,
K. L., Thelin, E. P., and Helmy, A. (2022) Focally ad-
ministered succinate improves cerebral metabolism
in traumatic brain injury patients with mitochondrial
dysfunction, J. Cereb. Blood Flow Metab., 42, 39-55,
https://doi.org/10.1177/0271678X211042112.
33. Stovell, M.G., Mada, M.O., Helmy,A., Carpenter, T.A.,
Thelin, E.P., Yan, J.-L., Guilfoyle, M.R., Jalloh,I., Howe,
D. J., Grice, P., Mason, A., Giorgi-Coll, S., Gallagher,
C. N., Murphy, M. P., Menon, D. K., Hutchinson, P. J.,
and Carpenter, K.L.H. (2018) The effect of succinate
on brain NADH/NAD
+
redox state and high energy
phosphate metabolism in acute traumatic brain inju-
ry, Sci. Rep., 8, 11140, https://doi.org/10.1038/s41598-
018-29255-3.
34. Malakhova, V. I., Godukhin, O. V., and Kondrash-
ova, M. N. (1992) Influence of succinate on the ef-
fectiveness of glutamatergic synaptic transmission
in surviving slices of rat hippocampus [in Russian],
Neurofiziologiia, 24, 238-243.
35. Nowak, G., Clifton, G. L., and Bakajsova, D. (2008)
Succinate ameliorates energy deficits and prevents
dysfunction of complex I in injured renal proximal
tubular cells, J. Pharmacol. Exp. Ther., 324, 1155-1162,
https://doi.org/10.1124/jpet.107.130872.
36. Panov,A., Mayorov, V. I., and Dikalov, S. (2022) Met-
abolic syndrome and β-oxidation of long-chain fat-
ty acids in the brain, heart, and kidney mitochon-
dria, Int. J. Mol. Sci., 23, 4047, https://doi.org/10.3390/
ijms23074047.
37. Pu, M., Zhang, J., Hong, F., Wang, Y., Zhang, C.,
Zeng,Y., Fang,Z., Qi,W., Yang,X., Gao,G., and Zhou,T.
(2024) The pathogenic role of succinate-SUCNR1: a
critical function that induces renal fibrosis via M2
macrophage, Cell Commun. Signal., 22, 78, https://
doi.org/10.1186/s12964-024-01481-5.
38. Moyon, A., Garrigue, P., Balasse, L., Fernandez, S.,
Brige, P., Bouhlel, A., Hache, G., Dignat-George, F.,
Taïeb, D., and Guillet, B. (2021) Succinate injection
rescues vasculature and improves functional recov-
ery following acute peripheral ischemia in rodents:
a multimodal imaging study, Cells, 10, 795, https://
doi.org/10.3390/cells10040795.
39. Sapieha, P., Sirinyan, M., Hamel, D., Zaniolo, K.,
Joyal, J.-S., Cho, J.-H., Honoré, J.-C., Kermorvant-
Duchemin, E., Varma, D. R., Tremblay, S., Leduc, M.,
Rihakova,L., Hardy,P., Klein, W.H., Mu,X., Mamer,O.,
Lachapelle,P., di Polo,A., Beauséjour,C., etal. (2008)
The succinate receptor GPR91 in neurons has a major
role in retinal angiogenesis, Nat. Med., 14, 1067-1076,
https://doi.org/10.1038/nm.1873.
40. Wu, K.K. (2023) Extracellular succinate: a physiolog-
ical messenger and a pathological trigger, Int. J. Mol.
Sci., 24, 11165, https://doi.org/10.3390/ijms241311165.
41. Huang, H., Li, G., He, Y., Chen, J., Yan, J., Zhang, Q.,
Li, L., and Cai, X. (2024) Cellular succinate me-
tabolism and signaling in inflammation: implica-
tions for therapeutic intervention, Front. Immu-
nol., 15, 1404441, https://doi.org/10.3389/fimmu.
2024.1404441.
42. Zhang, W., and Lang, R. (2023) Succinate metabo-
lism: a promising therapeutic target for inflamma-
tion, ischemia/reperfusion injury and cancer, Front.
Cell Dev. Biol., 11, 1266973, https://doi.org/10.3389/
fcell.2023.1266973.
43. Koyasu, S., Kobayashi, M., Goto, Y., Hiraoka, M., and
Harada, H. (2018) Regulatory mechanisms of hypox-
ia‐inducible factor 1 activity: two decades of knowl-
edge, Cancer Sci., 109, 560-571, https://doi.org/10.1111/
cas.13483.
44. Semenza, G. L. (2010) Hypoxia-inducible factor 1:
regulator of mitochondrial metabolism and me-
diator of ischemic preconditioning, Biochim. Bio-
phys. Acta, 1813, 1263-1268, https://doi.org/10.1016/
j.bbamcr.2010.08.006.
45. Fukuda,R., Zhang,H., Kim, J.W., Shimoda,L., Dang,
C. V., and Semenza, G. L. (2007) HIF-1 regulates cy-
tochrome oxidase subunits to optimize efficiency of
EFFECTS OF SUCCINATE ON KIDNEY AND BRAIN CELLS 1997
BIOCHEMISTRY (Moscow) Vol. 90 No. 12 2025
respiration in hypoxic cells, Cell, 129, 111-122, https://
doi.org/10.1016/j.cell.2007.01.047.
46. Piel, S., Ehinger, J. K., Chamkha, I., Frostner, E. Å.,
Sjövall, F., Elmér, E., and Hansson, M. J. (2018) Bio-
energetic bypass using cell-permeable succinate, but
not methylene blue, attenuates metformin-induced
lactate production, Intensive Care Med. Exp., 6, 22,
https://doi.org/10.1186/s40635-018-0186-1.
47. Owiredu,S., Ranganathan,A., Eckmann, D.M., Shofer,
F. S., Hardy, K., Lambert, D. S., Kelly, M., and Jang,
D. H. (2020) Ex vivo use of cell-permeable succi-
nate prodrug attenuates mitochondrial dysfunction
in blood cells obtained from carbon monoxide-poi-
soned individuals, Am. J. Physiol. Cell Physiol., 319,
C129-C135, https://doi.org/10.1152/ajpcell.00539.2019.
48. Owiredu, S., Ranganathan, A., Greenwood, J. C.,
Piel, S., Janowska, J. I., Eckmann, D. M., Kelly, M.,
Ehinger, J. K., Kilbaugh, T. J., and Jang, D. H. (2020)
In vitro comparison of hydroxocobalamin (B12a) and
the mitochondrial directed therapy by a succinate
prodrug in a cellular model of cyanide poisoning,
Toxicol. Rep., 7, 1263-1271, https://doi.org/10.1016/
j.toxrep.2020.09.002.
49. Bețiu, A. M., Chamkha, I., Gustafsson, E., Meijer, E.,
Avram, V. F., Åsander Frostner, E., Ehinger, J. K.,
Petrescu, L., Muntean, D. M., and Elmér, E. (2021)
Cell-permeable succinate rescues mitochondrial res-
piration in cellular models of amiodarone toxicity,
Int. J. Mol. Sci., 22, 11786, https://doi.org/10.3390/
ijms222111786.
50. Janowska, J. I., Piel, S., Saliba, N., Kim, C. D., Jang,
D.H., Karlsson, M., Kilbaugh, T. J., and Ehinger, J. K.
(2020) Mitochondrial respiratory chain complex I
dysfunction induced by N-methyl carbamate ex
vivo can be alleviated with a cell-permeable succi-
nate prodrug, Toxicol In Vitro, 65, 104794, https://
doi.org/10.1016/j.tiv.2020.104794.
51. Avram, V. F., Bîna, A. M., Sima, A., Aburel, O. M.,
Sturza, A., Burlacu, O., Timar, R. Z., Muntean, D. M.,
Elmér, E., and Crețu, O. M. (2021) Improvement of
platelet respiration by cell-permeable succinate in
diabetic patients treated with statins, Life (Basel), 11,
288, https://doi.org/10.3390/life11040288.
52. Avram, V. F., Chamkha, I., Åsander-Frostner, E.,
Ehinger, J. K., Timar, R. Z., Hansson, M. J., Muntean,
D. M., and Elmér, E. (2021) Cell-permeable succinate
rescues mitochondrial respiration in cellular mod-
els of statin toxicity, Int. J. Mol. Sci., 22, 424, https://
doi.org/10.3390/ijms22010424.
53. Vinokurov, A. Y., Popov, S. V., Belyakov, D. Y., Popov,
D. Y., Nikulin, A. S., Zakrzhevskaya, V. D., Guseinov,
R. G., Sivak, K.V., Dunaev, A.V., Potapova, E. V., and
Abramov, A. Yu. (2025) Cytoprotective action of so-
dium fumarate in an in vitro model of hypoxia us-
ing sodium dithionite, Sovrem. Tekhnologii Med.,
17, 93-106, https://doi.org/10.17691/stm2025.17.1.09.
54. Takasu, C., Vaziri, N. D., Li, S., Robles, L., Vo, K.,
Takasu, M., Pham, C., Liu, S., Farzaneh, S. H., Foster,
C. E., Stamos, M. J., and Ichii, H. (2015) Treatment
with dimethyl fumarate attenuates calcineurin inhibi-
tor-induced nephrotoxicity, Transplantation, 99, 1144-
1150, https://doi.org/10.1097/TP.0000000000000647.
55. Zhou, K., Xie, M., Yi, S., Tang, Y., Luo, H., Xiao, Q.,
Xiao, J., and Li, Y. (2021) Dimethyl fumarate ame-
liorates endotoxin-induced acute kidney injury
against macrophage oxidative stress, Ren. Fail., 43,
1229-1239, https://doi.org/10.1080/0886022X.2021.
1963774.
56. An, X., Yin, M., Shen, Y., Guo, X., Xu, Y., Cheng, D.,
and Gui, D. (2025) Dimethyl fumarate ameliorated
pyroptosis in contrast-induced acute renal injury by
regulating endoplasmic reticulum stress and JAK2-
STAT3 pathway, Ren. Fail., 47, 2504633, https://doi.org/
10.1080/0886022X.2025.2504633.
57. Ashari, S., Naghsh, N., Salari, Y., Barghi, N. G., and
Bagheri, A. (2023) Dimethyl fumarate attenuates
Di-(2-ethylhexyl) phthalate-induced nephrotoxicity
through the Nrf2/HO-1 and NF-κB signaling pathways,
Inflammation, 46, 453-467, https://doi.org/10.1007/
s10753-022-01746-6.
58. Yang,Y., Cai,F., Zhou, N., Liu,S., Wang, P., Zhang,S.,
Zhang, Y., Zhang,A., Jia,Z., and Huang, S. (2021) Di-
methyl fumarate prevents ferroptosis to attenuate
acute kidney injury by acting on NRF2, Clin. Transl.
Med., 11, e382, https://doi.org/10.1002/ctm2.382.
59. Zhao, X., Sun, G., Zhang, J., Ting, S.-M., Gonzales, N.,
and Aronowski, J. (2015) Dimethyl fumarate pro-
tects brain from damage produced by intracerebral
hemorrhage by mechanism involving Nrf2, Stroke,
46, 1923-1928, https://doi.org/10.1161/STROKEAHA.
115.009398.
60. Iniaghe, L. O., Krafft, P. R., Klebe, D. W., Omogbai,
E. K. I., Zhang, J. H., and Tang, J. (2015) Dimethyl
fumarate confers neuroprotection by casein kinase
2 phosphorylation of Nrf2 in murine intracerebral
hemorrhage, Neurobiol. Dis., 82, 349-358, https://
doi.org/10.1016/j.nbd.2015.07.001.
61. Casili, G., Campolo, M., Paterniti, I., Lanza, M.,
Filippone, A., Cuzzocrea, S., and Esposito, E. (2018)
Dimethyl fumarate attenuates neuroinflammation
and neurobehavioral deficits induced by experimen-
tal traumatic brain injury, J. Neurotrauma, 35, 1437-
1451, https://doi.org/10.1089/neu.2017.5260.
62. Krämer,T., Grob,T., Menzel,L., Hirnet,T., Griemert,E.,
Radyushkin,K., Thal, S.C., Methner,A., and Schaefer,
M. K. E. (2017) Dimethyl fumarate treatment after
traumatic brain injury prevents depletion of antioxi-
dative brain glutathione and confers neuroprotection,
J. Neurochem., 143, 523-533, https://doi.org/10.1111/
jnc.14220.
63. Yao, Y., Miao, W., Liu, Z., Han, W., Shi, K., Shen, Y.,
Li, H., Liu, Q., Fu, Y., Huang, D., and Shi, F.-D. (2016)
BUYAN et al.1998
BIOCHEMISTRY (Moscow) Vol. 90 No. 12 2025
Dimethyl fumarate and monomethyl fumarate promote
post-ischemic recovery in mice, Transl. Stroke Res.,
7, 535-547, https://doi.org/10.1007/s12975-016-0496-0.
64. Singh,D., Reeta, K.H., Sharma,U., Jagannathan, N.R.,
Dinda, A. K., and Gupta, Y. K. (2019) Neuro-protec-
tive effect of monomethyl fumarate on ischemia
reperfusion injury in rats: Role of Nrf2/HO1 pathway
in peri-infarct region, Neurochem. Int., 126, 96-108,
https://doi.org/10.1016/j.neuint.2019.03.010.
65. Clausen, B. H., Lundberg, L., Yli-Karjanmaa, M.,
Martin, N. A., Svensson, M., Alfsen, M. Z., Flæng,
S. B., Lyngsø, K., Boza-Serrano, A., Nielsen, H. H.,
Hansen, P. B., Finsen, B., Deierborg, T., Illes, Z., and
Lambertsen, K. L. (2017) Fumarate decreases ede-
ma volume and improves functional outcome af-
ter experimental stroke, Exp. Neurol., 295, 144-154,
https://doi.org/10.1016/j.expneurol.2017.06.011.
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.