ISSN 0006-2979, Biochemistry (Moscow), 2025, Vol. 90, No. 12, pp. 1811-1848 © Pleiades Publishing, Ltd., 2025.
Russian Text © The Author(s), 2025, published in Biokhimiya, 2025, Vol. 90, No. 12, pp. 1941-1980.
1811
REVIEW
Classification of Mitochondrial Protonophoric
Uncouplers and their Modifications
in Biological Environment
Yuri N. Antonenko
1,a
*, Elena A. Kotova
1
, Vladimir S. Krasnov
1,2
,
and Roman S. Kirsanov
1
1
Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University,
119991 Moscow, Russia
2
Faculty of Chemistry, Lomonosov Moscow State University, 119991 Moscow, Russia
a
e-mail: antonen@belozersky.msu.ru
Received July 14, 2025
Revised October 7, 2025
Accepted October 8, 2025
AbstractIn this review, we analyze diversity of mitochondrial uncouplers, a class of compounds, which was
in the focus of Vladimir Skulachev’s attention throughout his scientific career, starting from the basics of bio-
energetics and validation of Mitchell’s chemiosmotic theory to the development of concepts of mild uncoupling
and its therapeutic role. The review is the first to put forward the idea of classifying uncouplers by the type
of a functional group that provides their protonophoric activity, i.e., the ability to transfer protons across
the membrane, causing its depolarization and thereby uncoupling the ATP synthesis from the operation of
proton pumps in the electron transport chain. In particular, it is shown that anionic and zwitterionic uncou-
plers can be divided into groups of OH-, NH-, SH-, and CH-acids. Of importance, here we consider metabolic
transformations of mitochondrial uncouplers determining tissue-specificity of their action.
DOI: 10.1134/S000629792560245X
Keywords: mitochondria, uncouplers, protonophores, oxidative phosphorylation, lipid membranes
* To whom correspondence should be addressed.
INTRODUCTION
Protonophores comprise a subgroup in a gener-
al group of ionophores. Ionophores are low molecu-
lar weight compounds of diverse chemical structure,
which are capable of forming complexes with ions
and transfer them across natural and artificial mem-
branes. In particular, the 12-membered cyclic peptide
valinomycin is capable of transporting potassium
cations across membranes with high potassium-sodi-
um selectivity [1]. In addition to electrogenic iono-
phores that transport ions together with their charge,
there are non-electrogenic ionophores that exchange
cations for protons or anions for hydroxide ions.
The best known among them is the polyether antibi-
otic nigericin [2] that exchanges potassium ions for
protons, and the dye of bacterial origin prodigiosin
[3] that exchanges chloride ions for hydroxide ions.
In this review we focus on the special type of
ionophores – electrogenic transporters of hydrogen
ions, which were coined protonophores. It was found
out that these compounds are able to uncouple both
oxidative and photosynthetic phosphorylation, i.e. to
disrupt coupling of proton pump functioning in elec-
tron transport chains of mitochondria, chloroplasts,
and bacteria with ATP synthesis on the energy-trans-
forming membranes. Under normal conditions, oxida-
tion of respiratory substrates in the inner mitochon-
drial membrane is coupled with the process of ADP
phosphorylation to ATP, which is a universal energy
“currency” in cells. Uncouplers disconnect these two
processes, so that mitochondrial respiration is not ac-
companied with ATP synthesis.
According to Mitchell’s theory, uncouplers per-
form their functions via transporting protons across
lipid parts of membranes, hence, they are protono-
phores [4, 5]. The simplest electric analogy of this
system is a battery (respiratory chain) and a lantern
ANTONENKO et al.1812
BIOCHEMISTRY (Moscow) Vol. 90 No. 12 2025
(ATP-synthase). If a shunt resistor is inserted between
them, the lantern is switched off. Opening of proton
channels formed by the special proton-translocating
proteins of the UCP family (Uncoupling Proteins) or
addition of special protonophoric uncouplers (com-
pounds that selectively transport hydrogen ions
across the membrane) could serve as such shunt re-
sistors. Among those, the uncouplers 2,4-dinitrophe-
nol (DNP) and carbonyl cyanide m-chlorophenylhy-
drazone (CCCP) are best known. They are weak acids
containing aromatic moieties, which help the mole-
cule to effectively cross the membrane even when
carrying negative charge despite the high energy bar-
rier for the anion to enter the hydrophobic region of
the membrane.
Due to disruptions in the cell bioenergetics
caused by protonophores they are considered as im-
portant compounds for biochemistry, cell biology, and
physiology. In addition to fundamental studies, they
are used in agriculture and forestry industry due to
their antimicrobial, insecticide, and herbicide (pesti-
cide) properties. In addition to the widely used DNP,
pentachlorophenol [6], 2-(1-methylpropyl)-4,6-dinitro-
phenol (dinoseb)[7,8], as well as diarylamine fluazi-
nam [9], and other compounds are used in industry.
However, not the industrial use of protonophores, but
their potential application in pharmacology is import-
ant for us. It is known that protonophores exhibit
protection against muliple significant pathologies in
animal models. They are cardio- [10], neuro- [11,  12],
nephro-  [13], and radio-  [14] protectors, and exert
antidiabetic effects [15-17]; and this list can be ex-
tended further. Potential of uncouplers as anticancer
preparations has been mentioned in many studies
[18]. Moreover, low doses of DNP were shown to sig-
nificantly extend lifespan of rats  [19], mice  [20], yeast
[21], and Drosophila flies [22].
Classic protonophores are organic acids with pK
a
close to physiological pH values, which have an ex-
tended system of π-electrons assumed to be capable of
delocalizing negative charge. This facilitates penetra-
tion of the anionic form of a protonophore (P
) across
the membrane in response to the applied voltage. Pro-
tonophores differ fundamentally from the penetrating
acids or bases, such as salicylic acid or 9-aminoacri-
dine, which are capable of crossing the membrane in
the neutral form, but cannot penetrate the membrane
in the charged form. As a result, they only can change
pH gradient on the membrane due to their concentra-
tion gradient, but cannot dissipate membrane potential
in mitochondria. Measurement of external concentra-
tion of salicylic acid could help to assess pH gradient
on mitochondrial and bacterial membranes [23, 24],
which is usually around 0.5-1 pH units [25].
In the recent study by Bertholet et al. [26], an
electric current through an individual mitoplast at-
tached to a glass micropipette was measured using
the patch-clamp technique. The addition of 50  µM
DNP resulted in a significant increase in the current
through a fragment of the inner mitochondrial mem-
brane, while in the case of plasma membrane of the
same surface area no increase in the current was ob-
served at 50  µM DNP. However, higher concentration
of DNP (500  µM and 1  mM) increased the current
through the plasma membrane. The DNP-induced
current through mitoplast was suppressed by the ad-
dition of the specific inhibitor of adenine nucleotide
translocator  1 (ANT1) carboxyatractyloside (CATR).
Similar results were obtained by another method in
the earlier studies conducted in Skulachev’s laborato-
ry demonstrating that the uncoupling effect of DNP
on mitochondria is partially mediated by interaction
with ANT1 [27]. In addition to the effect of CATR, Ber-
tholet et al. investigated [26] mitoplasts derived from
the mice with knockout of the ANT1 gene. It was
shown that the effect of other well-known uncou-
plers, CCCP and carbonyl cyanide p-trifluoromethoxy-
phenylhydrazone (FCCP), also is partially mediated by
the interaction with ANT1 [26, 28]. These results are
in agreement with the results of studies conducted
in 1960s-1970s reporting ability of uncouplers to bind
to proteins [29] and the existence of specific binding
sites for uncouplers on the membranes [30, 31].
CHEMICAL STRUCTURES
OF PROTONOPHORES, THEIR DIVERSITY
AND CLASSIFICATION
Several reviews have been published in recent
years that summarize the data of numerous studies
on various uncouplers [32-36]. However, no classifi-
cation of uncouplers has been discussed in these re-
views. According to the type of the charged form of
the molecules, which participate in proton transfer
across membranes, protonophores could be divided
into three main classes: anionic, cationic, and zwit-
terionic (Fig. 1). We suggest this classification in the
present review.
In the earlier studies, protonophores were often
divided into two types: type  1 included those com-
pounds that transport protons as monomers, and
type  2– transport as dimers [37,38]. The dimer-based
mechanism was postulated for DNP [39], tetrachlo-
rotrifluoromethyl benzimidazole (TTFB) [40], 5,6-di-
chloro-2-trifluoromethylbenzimidazole (DTFB) [41],
and pentachlorophenol  [42] based on measuring
electric current through planar bilayer lipid mem-
brane (BLM). The current across BLMs induced by
these protonophores increased quadratically with the
increase in their concentration under certain con-
ditions. On the other hand, the uncoupling activity
MITOCHONDRIAL UNCOUPLERS 1813
BIOCHEMISTRY (Moscow) Vol. 90 No. 12 2025
Fig. 1. Operation cycles of anionic protonophore (a), cationic protonophore (b), zwitterionic protonophore in cationic
charged form (c), and zwitterionic protonophore in anionic charged form (d).
in isolated mitochondria depended linearly on the
concentration of these compounds [43, 44]. Of note,
the formation of dimers in the case of pentachloro-
phenol depended significantly on the solvent and was
suppressed upon the addition of water to organic sol-
vent [45]. The formation of dimers of uncouplers was
also reduced in the case of BLM formed from the lip-
ids dissolved in chlorodecane but not from the lipids
dissolved in the commonly used decane [44]. It could
be suggested that unlike the case of planar BLM, the
uncoupling activity of these compounds in mitochon-
dria proceeds via the monomeric mechanism due to
the presence of a large number of proteins in these
membranes, which affect physicochemical properties
of the membrane. Hence, the previously suggested
classification of protonophores into two types could
not correspond to their properties in biological en-
vironment.
Anionic protonophores. At present, anionic pro-
tonophores comprise the most numerous group of
compounds that includes the well-known uncouplers
such as DNP and CCCP. Such protonophores could
exhibit very high rate of operation, because anions
penetrate through membranes much more effectively
than cations due to the presence of the dipole poten-
tial on lipid membranes [46, 47]. The dipole poten-
tial is formed by the oriented dipoles of phospholipid
heads and a layer of tightly bound water, with large
contribution provided by carbonyls of ester bonds be-
tween fatty acids and glycerol [48]. In turn, anionic
protonophores could be divided into classes according
to the type of a group from which proton is cleaved:
OH-acids, NH-acids, SH-acids, and CH-acids.
Based on the results of investigation of uncou-
plers on BLM, a classic scheme of operation of an-
ionic protonophores has been suggested: an anionic
form of a protonophore (P
) crosses the membrane in
response to the applied potential, then it is protonat-
ed (transforming into the PH-form), diffuses in a neu-
tral form to the opposite side along the concentration
gradient, and, finally, is deprotonated, thus complet-
ing the full cycle of protonophore operation (Fig.  1a).
Such a carousel-like proton cycling may occur with
proton release into water and without it. The latter
variant was termed ‘small carousel’ by Markin and
Chizmadzhev  [49]. In this model, transport of the
anionic form of a protonophore P
is determined by
electrogenicity and voltage dependence. Proton se-
lectivity of protonophore functioning is usually eval-
uated from the potential of the open circuit in the
presence of pH gradient, while comparison with the
Nernst equation allows one to quantitatively estimate
the selectivity. High permeability of the neutral (PH)
form of a protonophore, resulting in its equal con-
centrations at the opposite sides of the membrane
[PH]
1
 = [PH]
2
, determines the proton selectivity of a
protonophore. Provided that protonation/deprotona-
tion reactions at the membrane are at equilibrium,
we have got [PH]  =  [P
]∙[H
+
]/K
a
at both sides of the
membrane. It follows from this equation that the ra-
tio of [P
]
1
to[P
]
2
is equal to the ratio [H
+
]
2
to [H
+
]
1
.
It means that the pH gradient leads to formation
ANTONENKO et al.1814
BIOCHEMISTRY (Moscow) Vol. 90 No. 12 2025
of the gradient of anionic forms of the protonophore
P
at two sides of the membrane. In this case, in-
variability of the near-membrane pH, which is pro-
vided by high buffer capacity of aqueous solution, is
an important issue. The question arises: at what pH
value the highest permeability of protonophore will
be achieved? Theoretical estimates indicate that this
pH value corresponds to the value of pK
a
plus half
of the logarithm of the ratio of permeabilities of the
neutral and anionic forms of the protonophore (see
equation 3.8 in the book by Markin and Chizmadzhev
[49]) [50]. Correspondingly, in the case of equal per-
meabilities of these two forms, we would obtain max-
imum permeability of the protonophore at the pH
value equal to pK
a
.
Above, we presented a kinetic model of protono-
phore operation. To have a possibility of predicting
the protonophoric activity of new potential drugs,
several authors have made attempts to create theo-
retical models based on the structure of compounds
[51-56]. Most of these models suggest that the key
factors determining the uncoupling activity are hy-
drophobicity of the compound and its pK
a
. In addi-
tion, the importance of the permeability not only of
the neutral form, but also of the deprotonated anion
has been noted. It has been reported in the review
by Kotova and Antonenko [34] that validity of such
models is limited due to contribution of the proteins
in the inner mitochondrial membrane to the uncou-
pling activity of protonophores.
The question arises: how closely associated are
the protonophore properties on BLM and their un-
coupling capacity in mitochondria? Correlation be-
tween these parameters has been shown to be rather
strong [57-59], but there are striking exceptions for
certain compounds [34, 60]. Correlation between pro-
tonophoric activity in mitochondria and in artificial
membranes is significantly better in the case of using
liposomes instead of BLMs [58]. Even better correla-
tion has been observed if the protonophoric activity
in mitochondria is assessed from their swelling under
certain conditions [61] or from the pH change in the
suspension of mitochondria in the presence of vali-
nomycin [38].
Uncouplers based on OH-acids. All synthetic phe-
nols uncouple mitochondria to varying degrees. Var-
ious substituted phenols have been analyzed previ-
ously in a number of reviews [30, 37, 38], and it has
been concluded that their protonophoric activity is
determined by their lipophilicity and the pK
a
value.
In addition to these two parameters, the activity is
affected by certain other factors. In particular, 3,5-di-
bromo-4-hydroxybenzonitrile has almost the same
lipophilicity and pK
a
, as DNP, but is 6-9-fold more
active in mitochondria [30, 62]. How could we ex-
plain this? Two main scenarios could be considered.
(1)  Translocation across the membrane is a complex
process associated with adaptation of the molecule
to the local environment. (2)  Another possible cause
could be interaction of substituted phenols with pro-
teins as described in the case of DNP [63].
The uncoupling action of halogen-substituted
phenols has been thoroughly investigated in the ear-
lier studies [51, 59, 64]. Pentachlorophenol, which
uncouples mitochondria at submicromolar concentra-
tions, has been found to be the most active among
them [43]. Its strong protonophoric activity has been
demonstrated on BLM [42, 58, 60, 65]. However, pen-
tachlorophenol has been found to inhibit respiratory
complex II with high efficiency [66]. Pentachloro-
phenol is widely used in the forestry industry as a
wood preservative, as well as pesticide in agriculture
[6]. The compound itself and its esters are also used
in peptide synthesis as reagents activating carboxyl
group. Extensive use of pentachlorophenol caused
significant contamination of the environment with
this compound, which is toxic to humans and, in par-
ticular, is a carcinogen.
The problem of environmental contamination is
even more significant in the case of using another
phenol – triclosan (5-chloro-2-(2′,4′-dichlorophenoxy)
phenol (Fig.  2). This compound exerts strong pro-
tonophoric action on the model lipid membranes,
while its uncoupling effect in mitochondria is rather
weak [67]. Actually, triclosan induces proton current
through BLM more effectively than CCCP, while in
mitochondria it should be added at a concentration
100-fold higher to induce the same effect as CCCP.
The reason for such discrepancy has not been eluci-
dated yet. The weak uncoupling activity of triclosan
could be associated with its inability to interact with
mitochondrial proteins that facilitate proton transfer
by uncouplers  [34]. Triclosan is well known as an
antibacterial agent, which has been widely used in
personal hygiene products. Currently, its use is legally
limited due to harmful effects on water ecosystems
via contaminated wastewater.
The best-known uncoupling phenol is DNP
(Fig. 2), which for some time was used as a drug
for stimulating metabolism in obesity treatment.
High interest in this compound arose during and
after World War I, when the toxic effect of DNP to
the workers, which participated in its production for
the war needs, was reported. Studies of the effects
of low doses of DNP revealed its ability to reduce
the amount of excessive fat both in animals and hu-
mans. This led to the use of DNP, as well as dinitro-
cresol, for obesity treatment. In the period from 1931
to 1934 this over-the-counter medicine was used by
~100,000 individuals [68]. Its effectiveness in terms of
weight loss was confirmed in more than 80% of the
cases. However, significant side effects were observed.
MITOCHONDRIAL UNCOUPLERS 1815
BIOCHEMISTRY (Moscow) Vol. 90 No. 12 2025
Fig.  2. Chemical structures of DNP, triclosan, SF6847, HU6, niclosamide, nitazoxanide, tizoxanide, fluorescein, 7-hydroxycou-
marin, 1799, phloretin, NPA, chlorfenapyr and its metabolite AC-303268.
ANTONENKO et al.1816
BIOCHEMISTRY (Moscow) Vol. 90 No. 12 2025
Some individuals had problems with liver, skin ulcers
were formed, and cases of cataract development were
reported. These side effects were observed even with
some patients taking recommended doses, not due to
the use of higher doses. Unfortunately, the cause of
enhanced toxicity of DNP for some individuals had not
been established. Monitoring the workers at chemical
factories producing DNP revealed that high toxicity
is exhibited in the individuals with high urine con-
centration of 2-amino-4-nitrophenol, a product of DNP
degradation [68]. Development of cataract was report-
ed in approximately 1% of the patients taking DNP,
and this occurred months after completion of the obe-
sity treatment. Search for therapeutics to treat such
cataract was unsuccessful, however, surgical interven-
tion helped in the majority of cases. Finally, in 1934
the use of DNP as a drug was prohibited. However, up
until now the use of DNP as an effective weight-loss
medicine has been reported in some fitness communi-
ties [69-71]. Interestingly enough, labels on the pack-
ages of DNP in pharmacies in 1930s stated that the
excessive effect of DNP could be suppressed by taking
baking soda [70]. It was indeed shown in the recent
study by Khailova et al. [72] that uncoupling of rat
liver mitochondria (RLM) by DNP could be effectively
inhibited by millimolar concentrations of bicarbonate.
Unfortunately, the mechanism of this effect has not
been elucidated. Further, we will consider in detail
modern approaches for decreasing side effects of DNP
and development of therapeutics on its basis.
After DNP was banned for use as a medicine due
to its high toxicity for humans, researchers started
to search for its analogues that would not cause side
effects and would be less toxic. In particular, it was
suggested to use methyl- or ethyl ethers of DNP, which
could be hydrolyzed in an organism producing DNP
[73]. This idea was developed in later studies [11,
15, 74]. At present, the preparation containing ethyl
ether of DNP is being examined in clinical trails as
a drug for treatment of Alzheimers and Parkinson’s
diseases. The fact is that DNP has manifested itself
as a neuroprotector in mouse models [12, 75,  76], as
well as cardioprotector [77,  78] and nephroprotec-
tor [13]. Another example of DNP ethers is methyl
ether of DNP (DNPME, 1-methoxy-2,4-dinitroben-
zene), which was investigated in Shulman’s laborato-
ry [74] in the USA. The authors demonstrated that
DNPME effectively protected mice from development
of non-alcoholic fatty liver disease (NAFLD), as well
as from insulin tolerance development. The authors
mentioned that DNPME hydrolysis and release of DNP
occurs predominantly in liver, hence, this preparation
is more suitable for treatment of liver disorders than
DNP itself. However, particular biochemical path-
ways mediating DNPME hydrolysis in liver cells were
not identified in this study.
Etherification of DNP with (1-methyl-2-nitro-
1H-imidazol-5-yl)methanol results in formation of
the compound HU6 (5-[(2′,4′-dinitrophenoxy)methyl]-
1-methyl-2-nitroimidazole) shown in Fig.  2. HU6
was tested in humans as an anti-obesity agent, as
well as in the patients with developed NAFLD [79].
It was concluded that HU6 could become a promising
pharmaceutical for treating patients with obesity and
NAFLD, as well as with their metabolic complications
[79]. According to the results of the study by Kitz-
manetal.[80], this agent could be also promising for
treating heart failure associated with obesity. It was
suggested in these studies that HU6 is a precursor
of DNP in an organism, which, in turn, mediates its
pharmacological effect. However, no details of HU6
metabolism were actually presented.
At present, DNP together with CCCP (Fig.  3) re-
main the most used uncouplers in the laboratory
practice, especially working with isolated mitochon-
dria. Itis the opinion of a number of researchers that
there is still significant therapeutic potential in using
DNP, which could be further increased by improv-
ing pharmacokinetics of DNP delivery to target cells.
C8-alkyloxy-substituted derivatives of FCCP and DNP
were used in the study by Ng et al. [81] for targeted
delivery of uncouplers to adipose tissues. The obtained
compounds containing ester bond (carbonyl cyanide
4-octyloxyphenylhydrazone and 2,6-dinitro-4-octyloxy-
phenol) accumulated in adipose tissues and were
more effective in acceleration of cellular metabolism
than FCCP and DNP; however, the esters were shown
to be unstable in the organism of mice, which was
considered as a drawback in this study. Several lab-
oratories have synthesized compounds that could
accumulate in mitochondria and release DNP upon
illumination or addition of hydrogen sulfide. Thus,
the light- and H
2
S-activated protonophores based on
4-hydroxybenzylidenepropanedinitrile (AG10) [82]
and DNP [83-86] were obtained. Scientists hope that
mitochondrial targeting of such forms of uncouplers
could ensure reduction of their acting concentrations
and initiate local uncoupling upon illumination or
in the presence of H
2
S. It was shown in our recent
study that 2-azido-4-nitrophenol, on the contrary, ex-
hibits properties of a protonophore that could be in-
activated by light [87], which creates a possibility of
local control of mitochondrial uncoupling.
One of the strongest mitochondrial uncouplers
is SF6847 ([(3,5-di-tert-butyl-4-hydroxyphenyl)methyl-
idene]propanedinitrile) (Fig.  2). This substituted phe-
nol uncouples mitochondria at nanomolar concen-
trations [88, 89]. The authors explain the enhanced
activity by high stability of the anionic form of
SF6847 in the membrane. According to calculations,
the anion has a planar structure, which facilitates
effective delocalization of the charge involving the
MITOCHONDRIAL UNCOUPLERS 1817
BIOCHEMISTRY (Moscow) Vol. 90 No. 12 2025
Fig. 3. Chemical structures of CCCP, FCCP, fluazinam, BAM15, pentachlorothiophenol (PCBT), SR4, and nonyl 3-picolinoyl-
carbazate (PDTC-9).
malononitrile group [90]. Low acting concentration of
SF6847  [89] could indicate that the uncoupler itself
rather than its complex with a protein operates as a
protonophore. However, it has been suggested in the
study by Grivennikova et al. [91] that the uncoupling
action of SF6847 could be partially mediated by its
binding to respiratory Complex I. It has been discov-
ered by Starkov et al. [92,  93] that the uncoupling
activity of SF6847 and other strong protonophores is
suppressed by 6-ketocholestanol, known as a modifi-
er of the membrane dipole potential [94]. Therefore,
the association of the recoupling with the action of
6-ketocholestanol on the lipid part of the membrane
cannot be ruled out [93,  95]. SF6847 is also known
as an inhibitor of tyrosine kinase [96,  97] and a po-
tential anticancer agent, usually named tyrphostin or
malonoben [96,  97].
Another strong uncoupler, niclosamide, belongs
to the group of salicylanilides and is the best-known
representative of this numerous group of com-
pounds (Fig.  2). Despite its high uncoupling activity,
this agent exhibits low cytotoxicity and is used as an
anthelmintic drug. Based on the approval of the Food
and Drug Administration of USA (FDA), at present,
niclosamide is tested for fighting many other diseases
including COVID-19 [98]. The fact is that one of the
critical stages of virus entry into cytoplasm is acidi-
fication of endosomes, which could be suppressed by
uncouplers [99,  100]. That is why many uncouplers
exhibit antiviral activity. With regards to the mech-
anism of action, it is known that niclosamide and
other salicylanilides induce proton current through
BLM [101,  102], however, more detailed investiga-
tion of the proton-transporting properties of salic-
ylanilides were conducted using the compound S13
(3-tert-butyl-4′-nitro-2′,5-dichlorosalicylanilide) as an
example. In the experiments with BLM it was shown
that classical monomeric model of proton transport
described above for CCCP is fully applicable for S13
[103]. The only difference consisted in the fact that
permeabilities of the protonated and deprotonat-
ed forms of S13 were approximately 10-fold higher
ANTONENKO et al.1818
BIOCHEMISTRY (Moscow) Vol. 90 No. 12 2025
than those of CCCP. Due to poor solubility, it is dif-
ficult to measure pK
a
of S13, but in the presence of
liposomes the pK
a
value was 7.0-7.3 [103]. S13 is a
strong uncoupler of oxidative phosphorylation be-
ing second only to SF6847 [38]. Hydroxyl serves as
a proton-donor group in salicylanilides; for example,
its methylation in niclosamide results in the loss of
the uncoupling activity [104]. Of note, salicylanilides
contain amino group in the form of CO–NH-linker
between two aryl groups, so that the amino group
proton could participate in formation of a hydrogen
bond with the hydroxyl oxygen atom in the salicylic
residue, which could facilitate delocalization of the
negative charge with formation of additional 6-mem-
bered cycle, thus increasing permeability of the com-
pounds through lipid membranes [105, 106].
Nitazoxanide has been also approved by FDA
as an anthelmintic drug. In a human organism it
is rapidly deacetylated and transformed into tizox-
anide (Fig.  2), an analogue of the above-described
niclosamide, and exhibits uncoupling properties both
in mitochondria and whole cells [107,  108]. Unlike
niclosamide, nitazoxanide has good bioavailability
and can easily penetrate into blood circulation in
the case of peroral administration. Based on the data
that nitazoxanide inhibits formation of atheroscle-
rotic plaques in the ApoE
−/−
mice with liver steatosis
fed according to the so-called ‘Western diet’ [108],
its use as a new antiatherosclerosis agent with high
clinical potential was suggested. As shown in another
study [109], administration of nitazoxanide through
a tube caused therapeutic effect in liver steatosis
induced by high-fat-diet in the C57BL/6J mice. This
study also confirmed that nitazoxanide is a promising
therapeutic agent against liver pathologies.
It is known that many natural phenols uncou-
ple oxidative phosphorylation in mitochondria [110].
Among those phloretin [111] (Fig.  2), as well as quer-
cetin [112] and hyperforin [113] should be men-
tioned. Many plant-derived metabolites of phenolic
nature exert mitochondrial uncoupling [112] and in-
crease electric current through BLM [110]. At micro-
molar concentrations, phloretin is known as a strong
modifier of dipole potential in BLM [114,115] (Fig.2).
Phloretin molecule has a large dipole moment and is
adsorbed at the membrane-water interface in an ori-
ented manner, which leads to suppression of proton
current mediated by anionic uncouplers and stimu-
lation of current mediated by cationic protonophores
[116]. Interestingly, natural phenols could be bromi-
nated, as, for example, (4,5,6-tribromo-2-(2′,4′-dibro-
mophenoxy)phenol also known as P01F08 [117]. This
compound exhibits high uncoupling capacity and
hasbeen investigated as a potential anticancer agent.
It is well-known that not only substituted phe-
nols could be uncouplers and anionic protonophores
of the OH-series. Fluorescein also has a suitable hy-
droxyl with pK
a
(~7) in its more complex aromatic
core; however, fluorescein itself does not exhibit un-
coupling properties (Fig. 2). This could be explained
by the presence of a free carboxyl group in the fluo-
rescein molecule. Nevertheless, butyl ester of fluores-
cein is also inactive, while the octyl ester of fluores-
cein exhibits uncoupling properties at submicromolar
concentrations [118]. Thus, it could be concluded that
fluorescein does not possess enough lipophilicity for
protonophoric activity and acquires it upon pending
a lipophilic tail. Lipophilic fluorescein derivatives
could be of interest as antibacterial agents [119].
Similar phenomenon, namely, the emergence
of protonophoric activity based on proton-donating
property of a hydroxyl substituent in the aromatic
system, caused by the addition of optimal-length alkyl
substituents to this system, has been demonstrated in
our study of 7-hydroxycoumarin derivatives (Fig.  2).
Coumarins (derivatives of 2H-1-benzopyran-2-one)
comprise a large class of natural heterocyclic com-
pounds exhibiting a wide range of therapeutic effects
[120]. Uncoupling activity of ostruthin [121] and some
other 7-hydroxycoumarin (umbelliferone) derivatives
[122] with hydroxyl pK
a
of 7.5 [123] has been re-
ported previously. It was shown in our recent study
that the introduction of hydrophobic substituents to
7-hydroxycoumarin results in creation of effective un-
couplers acting at micromolar or even submicromo-
lar concentrations [124, 125]. Detailed investigation
of the uncoupling activity of octyl and decyl esters
of 7-hydroxycoumarin-3-carboxylic acid on RLM re-
vealed a very interesting feature, namely, sponta-
neous decay of the activity occurring due to the loss
of alkyl substituents [125] as a result of enzymatic
hydrolysis of the esters. Surprisingly, the uncoupling
remains constant in the case of fluorescein octyl ester
[118]. The phenomenon of time-limited uncoupling
and metabolism of uncouplers will be discussed in
more detail further in the review.
All examples of anionic protonophores of the OH-
type described above are compounds with aromatic
hydroxyl. However, it is known that uncouplers could
be found among the compounds with aliphatic hy-
droxyl, such as, for example compound 1799 (2,6-di-
hydroxy-2,6-bis(trifluoromethyl)-1,1,1,7,7,7-hexafluoro-
heptane-4-one) (Fig. 2), which is a strong uncoupler
and protonophore [126]. It is known that the com-
pound 1799 increases permeability of BLM [58,  60].
However, the mechanism of its action is unknown, and
can be only assumed. Protonophoric action of 1799
indicates that aromaticity is not needed for effective
penetration of anionic form of protonophore across
the membrane. The absence of the effect of aroma-
ticity on the membrane permeability has been also
noted for tricyclohexylphosphonium derivatives[127].
MITOCHONDRIAL UNCOUPLERS 1819
BIOCHEMISTRY (Moscow) Vol. 90 No. 12 2025
Another variant of non-aromatic hydroxyls with
suitable pK
a
are fatty acids. It has been known for
a long time that fatty acids uncouple mitochondria
at concentrations of tens of micromoles. At the same
time, they exhibit very weak ability to increase con-
ductance of planar BLMs: noticeable currents have
been observed only for the membranes formed from
liposomes [128] according to the method of Montal
and Mueller [129]. Moreover, it was shown that the
uncoupling effect of fatty acids in mitochondria is
partially mediated by the proteins – ADP/ATP-anti-
porter and UCP [27, 63, 128, 130-137]. Interestingly
enough, dicarboxylic fatty acids are also capable of
uncoupling mitochondria; more precisely they stim-
ulate respiration, but not dissipate mitochondrial
membrane potential [138]. This rather complicated
phenomenon was explained by the interaction of
such compounds with respiratory complex II in mi-
tochondria. Dicarboxylic fatty acids do not increase
current in BLM, but penetrate across the BLM in a
neutral form [139]. It should be noted that carbox-
yl groups in the vicinity of aromatic ring usually do
not mediate uncoupling as can be demonstrated with
salicylic acid as an example [140]. But in some cases,
the compounds with such structure could function as
weak uncouplers, as in the case of BT2 (3,6-dichlo-
robenzo[b]thiophene-2-carboxylic acid), which uncou-
ples mitochondria at hundreds of micromoles and
exhibits a cardioprotective effect [141]. Azido-aryl-de-
rivatives of various physiologically active compounds
are often used for searching target proteins [142].
Upon illumination with near ultraviolet light, the
azido group loses N
2
and is transformed into the
highly reactive nitrene, which is capable of reacting
with various groups in proteins, in particular, amino
groups. 2-Azido-4-nitrophenol (NPA; Fig.  2) was syn-
thesized as the closest analogue of DNP and was used
for investigation of interactions of uncouplers with
mitochondrial proteins [143]. NPA was shown to be
even more active as an uncoupler than DNP (active
concentration of NPA is approximately 3-4-fold lower
than that of DNP) [87, 144]. Based on the studies of
isolated mitochondria, NPA has specific binding sites
on unidentified mitochondrial proteins; moreover,
this affinity depends significantly on the presence
of other known uncouplers such as DNP and CCCP
[30, 143]. Among these proteins, a band, correspond-
ing to the molecular mass ~30,000  Da, was found.
In the case of azido-derivatives of fatty acids ex-
posed to UV illumination, researchers observed cova-
lent binding to the protein with molecular mass of
~30  kDa, which was identified as ANT1 [145].
It has been shown recently that the antibiotic
pyrrolomycin is also, most likely, an anionic proto-
nophore of OH-acid type [146, 147]. The pH-depen-
dence of pyrrolomycin  D activity in BLM exhibited
an optimum at pH 9 [147]. Unlike synthetic protono-
phores, pyrrolomycins are produced by the bacteria
Streptomyces vitaminophilus, Streptomyces sp., and
Streptomyces fumanus. These compounds, having a
large number of chlorine atoms in the molecule, un-
couple mitochondria and submitochondrial particles
(SMPs) at subnanomolar concentrations [147]. These
concentrations are extraordinary low, because usual-
ly acting concentrations of uncouplers in SMPs are
significantly higher than those in mitochondria. The
question on the site of deprotonation in the pyrro-
lomycin molecule remains open, because in addition
to phenolic hydroxyl, deprotonation may also occur
at the nitrogen atom in the pyrrole ring. However,
antibacterial and protonophoric activities of pyrrolo-
mycinsI andJ, having methylated phenolic hydroxyl,
are significantly reduced [146].
Based on the natural antibiotic – derivative of
halogenated pyrrole dioxapyrrolomycin [7], the in-
secticide chlorfenapyr was developed. Its toxicity is
too high for crop treatment, but it is used for treat-
ment of home gardens and forests where there are
no edible plants. Chlorfenapyr itself does not exert
the uncoupling effect on RLM; however, in insect
cells N-dealkylation occurs resulting in the forma-
tion of free pyrrole, presumably with participation
of cytochromes P450 (CYP450) [148]. Structures of
chlorfenapyr and the product of its metabolism with
uncoupling properties are presented in Fig.  2. In fact,
the chlorfenapyr metabolite, although related to pyr-
rolomycins, performs the mitochondrial uncoupling
as NH-acid.
Uncouplers based on NH-acids. This numerous
group includes compounds that contain nitrogen
both within and outside the aromatic ring. The best
known uncoupler CCCP, as well as its close analogue
FCCP (Fig.  3), both introduced by Heytler at the be-
ginning of 1960s[126, 149,150], belong to this group.
Despite the fact that these compounds are unstable
in the presence of thiol-reagents [150-155], CCCP
became very popular, especially in the tests involv-
ing mitochondrial uncoupling in cells. Most likely,
this is associated with its low acting concentrations
(hundreds of nanomoles for isolated mitochondria
and several micromoles for intact cells), as well as
with its rather low toxicity [14, 156, 157]. Another
advantage of CCCP is its weak inhibiting effect on
mitochondrial respiratory pumps, which is observed
only at concentrations tens-fold higher than the un-
coupling concentrations. With regard to the mecha-
nism of the protonophoric action, CCCP is a typical
anionic protonophore with pK
a
≈  6 and maximum
protonophoric activity at pH around 8 [158-160]. The
difference between pK
a
andpH-optimum of the activ-
ity is explained by a large difference in permeability
for the anionic and neutral forms of the uncoupler.
ANTONENKO et al.1820
BIOCHEMISTRY (Moscow) Vol. 90 No. 12 2025
Similar to DNP, CCCP was tested in cells and animal
physiological models of various pathologies, where it
was proved to have properties of neuroprotector [161]
and cardioprotector [78]. Similar to the case of DNP,
azido-CCCP was synthesized (N
3
CCP, carbonyl cyanide
2-nitro-4-azidophenyl hydrazone) and its interaction
with mitochondria in the dark and under illumina-
tion with UV light was investigated; conjugation with
some mitochondrial proteins was observed under
illumination [31, 162]. Unfortunately, these proteins
have not been yet identified, the label was observed
in the band corresponding to the protein with mo-
lecular weight of 10,000-15,000  Da. Azido-CCCP was
poorly investigated as an uncoupler: there are even
no data on comparison of its acting concentrations
with those for CCCP in mitochondria and no data at
all on its protonophoric activity in BLM.
It was mentioned above that CCCP and its more
active analogue FCCP could react with thiols in a
chemical system in the absence of mitochondria. How-
ever, the issue of the possibility of interaction of CCCP
and FCCP with natural thiols in cells, and primarily
with glutathione (GSH), remains unsolved. This prob-
lem becomes especially important because, according
to our data, CCCP and FCCP can interact with N-acetyl
cysteine (NAC) in a chemical system [155]. The fact
is that NAC as an antioxidant is often used in the
experiments with cells (as well as in  vivo) together
with CCCP and FCCP [163-169]. In particular, it was
shown that cardioprotective effect of FCCP could be
completely eliminated by the addition of NAC [163],
which was explained as an effect of FCCP on the level
of reactive oxygen species (ROS) in heart cells. How-
ever, another possibility exists, that involves simple
chemical modification of FCCP upon the addition of
NAC to this system, which could block the uncoupling
action.
Another synthetic anionic protonophore of the
NH-acid series – BAM15 (N
5
,N
6
-bis(2-fluorophenyl)
[2,1,3]oxadiazolo[4,5-b]pyrazine-5,6-diamine) (Fig.  3),
introduced by Santos’s research group in 2013 [170],
could be considered as currently the most investigat-
ed from the physiological point of view. BAM15 ex-
hibits strong hepatoprotective properties [171] and is
effective in murine obesity models [172]. The chem-
ical structure of BAM15 is more complex than that
of CCCP, hence, the issue of its protonation-depro-
tonation is not so trivial. By using the capillary elec-
trophoresis method, it was shown that BAM15 could
be deprotonated in two steps with pK
a
6.44 and 7.99
forming mono- and di- anions, respectively [50]. The
proton transfer across BLM mediated by BAM15
could be suppressed by the addition of phloretin,
which confirms the anionic nature of this protono-
phore [50]. It has been stated previously that BAM15
has significant advantage compared to CCCP, because
it does not change permeability and electrical activity
of the plasma membrane [170], however, this conclu-
sion was challenged in the study by Firsovet al. [50].
In addition to the direct protonophoric action, BAM15
is capable of inducing proton transport across the in-
ner mitochondrial membrane via its interaction with
the ANT1 protein [26, 28, 50].
Fluazinam is also a very strong uncoupler of the
NH-type, which belongs to the group of diarylamines
(Fig. 3). Noteworthy, diarylamines comprise a large
group of compounds synthesized with the primary
goal to create effective pesticides [7]. The protono-
phoric action of fluazinam on BLM, demonstrated by
Khailova et al. [155], appeared to be close to that of
FCCP. The uncoupling concentrations of fluazinam
were shown to be subnanomolar, i.e., even less than
those of SF6847 [173]. However, fluazinam is rapid-
ly metabolized in RLM with formation of conjugates
with glutathione facilitated by glutathione-S-transfer-
ase (GST) [155, 173-175].
Similar metabolic transformation was observed
in our recent study [176] on another group of NH-
type uncouplers – derivatives of N-phenylthiophen-
2- amine [177]. Details of the transformation will be
discussed further in the section devoted to metab-
olism of uncouplers. The protonophore, discovered
almost 10 year ago – endosidin-9 (ES9, 5-bromo- N-
(4-nitrophenyl)thiophene-2-sulfonamide), which is
N-aryl thiophenesulfonamide – compound related to
anilinothiophenes, also belongs to the NH-acid type
of protonophores [178].
Uncouplers of the NH-type also include deriva-
tives of the well-known fluorescent dye NBD (7-nitro-
benz-2-oxa-1,3-diazole), modified by introduction of
amino group at position 4 of the benzene ring, which
could be deprotonated with pK
a
~10 [179]. A series of
4-alkylamino-7-nitrobenz-2-oxa-1,3-diazoles was syn-
thesized, for which it was shown that starting with
octyl the uncoupling and protonophoric properties
have been observed [180]. The maximum uncoupling
effect on mitochondria was observed precisely for
the N-octyl derivative with acting concentration be-
ing tens of micromoles. The N-dodecyl-derivative was
shown to be a significantly stronger protonophore
than the octyl analogue in experiments on BLM [180].
Such difference between the activities in mitochon-
dria and BLM could be associated with the ability of
7-nitro-4-octylaminobenz-2-oxa-1,3-diazole to interact
with ANT1, thereby promoting additional proton flow
with participation of this protein [180, 181].
All the above-mentioned uncouplers of the
NH-acid type are synthetic compounds. However, it
was shown recently that the natural toxin aetok-
thonotoxin, which is produced by the cyanobacteria
Aetokthonos hydrillicola that causes poisoning in
fishes and birds, is a strong uncoupler of oxidative
MITOCHONDRIAL UNCOUPLERS 1821
BIOCHEMISTRY (Moscow) Vol. 90 No. 12 2025
phosphorylation, and this uncoupling is likely the
main mechanism of the toxicity [182]. This toxin
contains an unusual bi-indole group and has five
bromine atoms as substituents. Methylation of the ni-
trogen atom in the indole ring results in the loss of
the uncoupling activity in animal cells at micromolar
concentrations of this toxin. It was shown that at the
same concentrations aetokthonotoxin induces electric
current through BLM at neutral pH.
Compounds based on diphenylurea such as SR4
(N,N′-bis(3,5-dichlorophenyl)urea) (Fig.  3) and ana-
logues could be also assigned with a certain caution
to protonophores of the NH-acid type [183]. Some au-
thors, on the contrary, associate protonophoric prop-
erties of SR4 with its ability to bind hydroxide ions
as well as other inorganic anions [184]. N,N′-diphenyl
urea is a natural compound – a plant hormone, which
controls, in particular, the formation of flowers. SR4
was shown to dissipate membrane potential and stim-
ulate respiration at submicromolar concentrations
[183]. Furthermore, derivatives of diphenylurea in-
creased proton permeability of liposome membranes
[185], which indicates their ability to act as classical
protonophores. These compounds were investigated
during the search for effective anticancer agents
[186]. For example, Sorafenib, which is a deriva-
tive of diphenylurea with a picolinamide fragment,
is used as an anticancer agent, and its main mech-
anism is inhibition of kinases [187]. On the other
hand, it was shown that Sorafenib uncouples mito-
chondria, and this property could be significant for
the anticancer effect [188, 189]. Interestingly enough,
similar compounds also could transport chloride ions
and other anions across model and natural mem-
branes [184].
Uncouplers based on CH-acids. C–H bond has, as
a rule, a negligibly weak capacity for dissociation, es-
pecially in an aqueous environment. However, in the
case of carboranes it was possible to observe disso-
ciation in different protic and aprotic solvents [190].
Moreover, decachloro-o-carborane has pK
a
~  7, and its
uncoupling action was demonstrated in the study by
Libermanetal.[191] already in 1970. The uncoupling
activity of ortho-carborane, unlike meta- and pa-
ra-carborane, in line with higher acidity of ortho-iso-
mers as compared to others [190], was confirmed in
the study by Rokitskaya et al. [192]. The uncoupling
effect on mitochondria correlated with the protono-
phoric activity in BLM and liposomes [192]. To the
best of our knowledge, these compounds have not
been tested in physiological experiments with ani-
mals. However, there are many studies on application
of carboranes as pharmacophores; a large number of
compounds – conjugates with carboranes were syn-
thesized [193]. Part of them exhibited the uncoupling
activity and was tested in tissue cultures [194].
Uncouplers based on SH-acids. Uncoupling action
of pentachlorothiophenol (PCBT; Fig. 3) in mitochon-
dria stimulating their respiration in the state 4 was
investigated in the early study by Wilson et  al. [43].
In particular, it was shown that PCBT uncouples mi-
tochondria at submicromolar concentrations, while it
practically does not induce current through BLM [43].
In the later (1983) study by Smejtek et al. [195] it
was clarified that in the study by Wilson et al. [43]
in 1971, photosensitivity and poor water solubility of
PCBT was not taken into account, and in fact perme-
ability induced by PCBT in BLM was approximately
10-fold higher, than the one at the same concentra-
tion of pentachlorophenol [195]. The pH-dependence
of BLM permeability in induced by PCBT had max-
imum at pH  ~  6; and pK
a
of PCBT was estimated
as 4.3 [43], while pK
a
of pentachlorophenol was ~4.7.
All these data imply that uncouplers based on SH-
acids have high activity and their application is lim-
ited only by their low chemical stability in biological
environment.
This conclusion has been supported by the results
of another study on the uncoupling activity of alkyl
acyldithiocarbazates, and, in particular, of nonyl 3-pi-
colinoyldithiocarbazate (PDTC-9) [196], which uncou-
ples mitochondria and induces proton permeability of
liposomes at micromolar concentrations. In aqueous
solutions PDTC-9 exists as two tautomers, with one
of them being thione, and another – thiol (Fig.  3).
Methylation of the nitrogen atom N2 in thione or the
sulfur atom in thiol results in complete loss of the
uncoupling activity. Considering that one of the tau-
tomers is a NH-acid, and another – SH-acid, PDTC-9
could be assigned both to NH- and SH-protonophores.
According to the data reported by Teradaet al. [196],
PDTC-9 exhibits weak reactivity with such natural thi-
ols as cysteine and GSH. Several structural analogues
of PDTC-9 were tested, and it was shown, in partic-
ular, that their activity increases with an increase
in the length of alkyl substituent reaching maximum
in the case of nonyl; further increase in the alkyl
substituent length leads to a decrease in the uncou-
pling activity [196].
Cationic protonophores. This group of uncou-
plers is not numerous in comparison with the group
of anionic protonophores. These compounds exist in
protonated cationic and deprotonated neutral forms.
It is essential that the protonated form in this group
of compounds is a charged molecule and not a zwit-
terion. Schematic representation of functioning of
these protonophores is shown in Fig.  1b. The cation-
ic charged form penetrates across the membranes
significantly less effective than the anionic one due
to the membrane dipole potential, hence, cationic
protonophores usually are effective at higher con-
centrations than the anionic ones. Another feature
ANTONENKO et al.1822
BIOCHEMISTRY (Moscow) Vol. 90 No. 12 2025
Fig. 4. Chemical structures of ellipticine, C4R1, Nile Blue, bupivacaine, and F16.
of cationic protonophores is their ability to accu-
mulate in the mitochondrial matrix, because mito-
chondrial membrane potential is negative inside.
Ellipticine is the best-known uncoupler of this type
[197] (Fig. 4). These compounds are practically not
used in the laboratory practice as uncouplers, because
they produce effect at very high concentrations and
the mechanism of their action is poorly understood.
However, ellipticine and related compounds exhibit
anticancer activity, and in this relation were investi-
gated in great detail [198]. The rhodamine derivative
C4R1 [199, 200] and the Nile Blue dye [201] could also
be assigned to cationic uncouplers.
Ellipticine is an anticancer alkaloid isolated from
the plants Ochrosia elliptica. It was shown that it
uncouples mitochondria at concentrations of tens of
micromoles with maximum at ~100  µM [197]. Ellipti-
cine contains a pyridine ring (Fig.  4), in which meth-
ylation of nitrogen atom results in almost complete
loss of the uncoupling activity. This observation indi-
cates that protonation-deprotonation of the nitrogen
atom is responsible for the protonophoric action of
ellipticine. The measured value of pK
a
of this nitro-
gen atom is close to 7.0 [202]. It was shown that in
cancer cells ellipticine is accumulated in nuclei and
mitochondria, furthermore, fluorescence of ellipticine
in mitochondria depends on the pH gradient on the
membrane [203].
As reported in 1984, rhodamine 6G, rhodamine
19 ethyl ester, uncouples mitochondria at micromo-
lar concentrations [204]. It was shown in the fol-
lowing studies that the butyl ester (C4R1; Fig. 4) ex-
hibits the highest uncoupling activity in the series
of rhodamine derivatives with alkyl substituents of
varying length [199]. Surprisingly, attempts to detect
the process of deprotonation in rhodamine 6G failed.
It seems that there was an error in the study by
Duvvurietal.[205]: in fact, there is no pH dependence
of the rhodamine 6G spectrum. It was shown with the
help of capillary electrophoresis that rhodamine 6G
does not exhibit pK
a
in the physiological range of
pH [206], while this method was successful in reli-
able determination of pK
a
for many other uncouplers.
It was also shown that the derivatives of rhodamine
19 increase current across BLM and behave as proto-
nophores [207]. This current is stimulated by phlore-
tin, i.e., it increases with a decrease in membrane
dipole potential [116]. At the same time, the ATPase
enzyme is apparently involved in the uncoupling ef-
fect of C4R1 on mitochondria [200]. Weak toxicity
of C4R1 was demonstrated in the experiments with
mice; in addition, this rhodamine stimulated weight
loss in rats maintained on a high fat diet [208].
Alkyl-rhodamines also exhibited antibacterial proper-
ties [209, 210].
Other dyes, such as Nile Blue (Fig. 4), pyronin Y,
and acridine orange also show properties of cation-
ic uncouplers [201]. However, Nile Blue is an inhibi-
tor of ATP synthase, and it has been suggested that,
similar to C4R1, it operates as an uncoupler in mito-
chondria with participation of this protein. This dye
was not investigated on BLM, however, its structur-
ally close derivative increased proton current across
a lipid membrane [211].
Bupivacaine (Fig. 4) is a local anesthetic with
piperidine heterocycle, which can be protonated at
MITOCHONDRIAL UNCOUPLERS 1823
BIOCHEMISTRY (Moscow) Vol. 90 No. 12 2025
neutral pH. The use of bupivacaine was approved
by FDA. It was shown that, similarly to some other
anesthetics [212], it decreases mitochondrial mem-
brane potential and stimulates their respiration at
millimolar concentrations, i.e. it functions as an un-
coupler [213, 214]. Evaluation of pK
a
of protonation
of the piperidine ring in bupivacaine gave the value
of 8.2[215]. Investigation of bupivacaine in liposomes
showed its protonophoric properties, however, the
authors suggest that the mechanism of the uncou-
pling effect of this anesthetic involves the formation
of selective proton channels by its aggregates [216],
which is in agreement with the data on the ability of
bupivacaine to form defects in the lipid membrane,
obtained by Terada et al. [214] with planar BLMs.
Screening a wide range of synthetic compounds
for anticancer effects revealed the cationic compound
F16 ((E)-4-(3-indolylvinyl)-N-methylpyridinium iodide)
with high antitumor activity [217] (Fig.  4). The au-
thors suggested that this anticancer effect is associat-
ed with accumulation of this cation in mitochondria
of tumor cells, causing induction of a mitochondrial
pore, release of cytochrome  c, and finally the apop-
tosis. It was shown both in the first study by Fan-
tin et al. [217], and in the later study [218] that F16
stimulates respiration of mitochondria and behaves
as an uncoupler. The authors of the later study by
Wang et al. [218] concluded that the uncoupling ac-
tivity of F16 is precisely responsible for its antican-
cer effects. Isomers of F16 were synthesized (o-F16,
(E)-2-(3-indolylvinyl)-N-methylpyridinium iodide and
m-F16, (E)-3-(3-indolylvinyl)-N-methylpyridinium io-
dide), which differ only by orientation of indole and
pyridine rings of the molecule [219]. It was found
that o-F16 exhibits properties of a strong uncoupler,
while m-F16 is practically inactive [219]. The authors
associate this property with lower acidity of the me-
ta-isomer. It should be mentioned that the mechanism
of deprotonation of F-16 and o-F16 is poorly under-
stood. Dissociation of proton from these compounds
could result in the formation of both neutral mole-
cules without charged groups, and zwitterions, as
was postulated in the study by Xu et  al. [219]. The
completely neutral deprotonated forms of F16 and
o-F16 were previously isolated and characterized
with the help of IR- and mass-spectroscopy [220]
(scheme in Fig. 4). We believe that formation of the
less hydrophobic zwitterion is more probable in the
case of deprotonation of m-F16. This fact could also
explain the reduced activity of m-F16. Of note, if in-
deed the deprotonated form of F16 is a zwitterion,
this uncoupler should be assigned to the next group
in our classification, namely the group of zwitteri-
onic uncouplers. To the best of our knowledge, the
protonophoric action of F16 on BLM has not been
investigated. In recent years, works on synthesis of
the conjugates of F16 with other anticancer agents
were published. In particular, the conjugate of F16
with betulinic acid exhibited significantly higher cyto-
toxicity towards tumor cells than individual betulinic
acid or F16 [221]. In later studies, the list of anti-
cancer preparations based on F16 was significantly
expanded [222-224].
Zwitterionic protonophores. Relatively recently
zwitterionic protonophores have been discovered, i.e.,
compounds with deprotonated form carrying simulta-
neously positive and negative charges comprising a
zwitterion [225,226]. Mechanism of their functioning
is presented in Fig. 1c. It is essential that zwitterion-
ic and cationic forms of such protonophores should
exhibit high permeability across the membrane. One
could imagine the process, when the zwitterionic
form of protonophores could be combined with an-
ionic form as a pair for proton transport (Fig.  1d),
however, such compounds have not been observed
experimentally. The majority of the confirmed zwit-
terionic protonophores are conjugates of triphenyl-
phosphonium with anionic protonophores (Fig.  5).
It was shown that these protonophores initiate pro-
ton current through BLM, which is stimulated by de-
creasing dipole potential upon the addition of phlore-
tin or including certain lipids in the composition of
BLMs [116], i.e. in this regard they behave as cation-
ic protonophores. Furthermore, these compounds are
accumulated in the cell mitochondria, which allows
considering them as mitochondria-targeted protono-
phores [225]. It must be noted that similar to anion-
ic protonophores, zwitterionic protonophores could
be classified according to the type of proton-donor
group: OH-, NH-, and CH-acids.
Conjugates of anionic uncouplers with triphenyl-
phosphonium cations. The first attempt to synthesize
a mitochondria-targeted protonophore was made in
2006 in the research group of Murphy and Smith
[227] by conjugating triphenylphosphonium with
DNP. However, the obtained compound termed mi-
toDNP (3-(4′-hydroxy-3′,5′-dinitrophenyl)propyltriph-
enylphosphonium methanesulfonate) (Fig.  5) did not
exhibit uncoupling properties in mitochondria, which
made questionable the initial assumption about the
existence of zwitterionic protonophores. However, in
2014 a conjugate of triphenylphosphonium and fluo-
rescein was synthesized, which was named mitoFluo
(10-[(2-(6-hydroxy-3-oxo-3H-xanthen-9-yl)benzoyl)oxy]
decyltriphenylphosphonium bromide) (Fig.  5); this
compound appeared to be more advantageous [225].
It was shown that mitoFluo uncouples mitochondria
at submicromolar concentrations, and is accumu-
lated in mitochondria, which is easily detected due
to the bright fluorescence of this compound. The
weak uncoupling effect of mitoDNP could be ex-
plained by: (1) the weaker uncoupling effect of DNP
ANTONENKO et al.1824
BIOCHEMISTRY (Moscow) Vol. 90 No. 12 2025
Fig. 5. Chemical structures of mitoDNP, mitoFluo, mitoCCCP, SF-C5-TPP, PP6, CMTPP-Cn (precursors of phosphonium ylides).
in comparison with alkyl-fluorescein and (2) by the
too short trimethylene linker in mitoDNP. The mito-
Fluo compound contained the decyl linker, and the
uncoupling effect was significantly reduced in the
case of a similar compound with the butyl linker
[228]. MitoFluo induced electric current through BLM,
which was selective for protons [225]. It was shown
that in vivo mitoFluo exerts neuro- and nephro-pro-
tective effects in the models of brain trauma and
rat kidney ischemia-reperfusion, respectively [228].
MitoFluo, as well as mitoNBD – the conjugate of
decyltriphenylphosphonium with NBD (7-nitrobenz-
2-oxa-1,3-diazole) also exhibited strong antibacterial
properties especially towards Gram-positive bacteria
[119, 229].
The success with mitoFluo stimulated research
in this direction: conjugates of triphenylphosphoni-
um with CCCP (mitoCCCP, 10-([4′-(dicyanomethylene)
hydrazinyl-2′-chlorophenyl]oxy)decyltriphenylphos-
phonium bromide) [230] and with the strongest
uncoupler SF6847 (mitoSF, covalent conjugates of
[(3,5-di-tert-butyl-4-hydroxyphenyl)methylidene]pro-
panedinitrile with alkyltriphenylphosphonium) were
synthesized [231]. It should be noted that attachment
MITOCHONDRIAL UNCOUPLERS 1825
BIOCHEMISTRY (Moscow) Vol. 90 No. 12 2025
of triphenylphosphonium to an anionic protono-
phore does not change its proton-donor group, hence,
mitoDNP, mitoFluo, and mitoSF could be considered
as OH-acids, similar to corresponding anionic pro-
tonophores, while mitoNBD and mitoCCCP – as NH-
acids. MitoCCCP exhibited very weak uncoupling
effect at submicromolar concentrations, while the
conjugate with SF6847 uncoupled mitochondria at
submicromolar concentrations. Among the deriva-
tives of SF6847, the conjugate with the pentyl linker
(SF-C5-TPP, (6-[3′,5′-di-tert-butyl-4′-hydroxyphenyl]-
7,7-dicyanohept-6-en-1-yl)triphenylphosphonium bro-
mide) exhibited the highest activity, while the uncou-
pling activity was significantly reduced in the case of
butyl- and decyl- linkers. It was also shown that the
compound with butyl linker is accumulated in mito-
chondria in response to their energization. Inisolated
RLM, SF-C5-TPP exhibited significantly lower uncou-
pling activity as compared to SF6847 itself (approxi-
mately by 2 orders of magnitude), however, it effec-
tively decreased membrane potential in mitochondria
in cell culture, which could be associated with the
ability of SF-C5-TPP to accumulate in cell mitochon-
dria. Furthermore, it induced proton current across
BLM, which was stimulated by the addition of phlore-
tin. This indicates that transmembrane diffusion of
the protonated cation is the rate-limiting step in the
protonophoric cycle of SF-C5-TPP. Of note, mitoCCCP
was found to be a strong uncoupler in the system
of inverted submitochondrial particles, as well as in
subbacterial particles, while this compound was a
weak uncoupler in isolated mitochondria [230].
In another series of studies, derivatives of [(E)-
2-(5-chloro-2-hydroxyphenyl)-2-phenylethenyl]phos-
phonium with substituents of varying length at the
phosphorus atom, which could be considered as
conjugates of phosphonium with para-chlorophenol
(PP6; Fig. 5) were investigated [226, 232, 233]. The
most active compounds in this series exhibited the
uncoupling effect in mitochondria at micromolar con-
centrations, suppressed growth of Bacillus subtilis
bacteria, and also induced proton current in BLMs.
Compounds with higher hydrophobicity were able to
create nonspecific defects in lipid membranes and
even cause hemolysis of erythrocytes.
Precursors of phosphonium ylides. Phosphonium
ylides are often used in the Wittig reaction. They com-
prise zwitterions with the positive charge at the phos-
phorus atom and the negative charge at the closest
carbon atom [234]. It was shown in our recent study
[235] that the cations of (alkyloxycarbonylmethyl)
triphenylphosphonium (Fig.  5) could release protons
of the methylene group and, in this way, to partici-
pate in the proton transfer across lipid membranes.
It was found out that the precursor of the phospho-
nium ylide CMTPP-C8 ((octyloxycarbonylmethyl)triph-
enylphosphonium bromide) stabilized by the ester
bond, is able to uncouple mitochondria at micromo-
lar concentrations and induce proton current across
BLM. Hence, the precursors of stabilized ylides, as
well as the carboranes described above, are protono-
phoric uncoulplers based on CH-acids. CMTPP-C8 and
its analogues exhibited moderate cytotoxicity in cell
culture and caused a decrease in the mitochondrial
membrane potential in cells at concentrations that
did not cause a significant decrease in cell survival.
CMTPP-C12 dissipated pH gradient in thylakoid mem-
branes of chloroplasts and exhibited antimicrobial
activity. It was also shown that methylation of phe-
nyl residues significantly increased the protonophoric
activity of these compounds, so the uncoupling con-
centrations in mitochondria decreased to the level of
tens of nanomoles [236].
Uncouplers not characterized in model sys-
tems. At present not all described uncouplers can be
classified. The data available in the literature demon-
strate uncoupling activity of certain compounds,
which were investigated only in biological systems
(cell cultures and mice), while their physicochemi-
cal properties and mechanisms of uncoupling were
not explored. In particular, the compound CZ5 (ethyl
(2E)-5-(4-chlorophenyl)-2-[(3,5-dibromo-4-hydroxyphe-
nyl)methylidene]-7-methyl-3-oxo-2,3-dihydro-5H-[1,3]
thiazolo[3,2-a]pyrimidine-6-carboxylate), described
in the study by Fu et  al. [237], most likely operates
as an anionic uncoupler, because it is a derivative
of 2,6-dibromophenol. The same could be said about
OPC-163493, a triazole-containing compound (4-(5-
methyl-2-(4-(trifluoromethyl)phenyl)-1,3-thiazol-4-yl)-
1H-1,2,3-triazole-5-carbonitrile), which most likely
could be deprotonated at alkaline pH [17]. However,
classification of these and some other protonophores
described in the literature requires further investi-
gation.
Cation-dependent protonophores. It has been
noted above in this review that the transporter of po-
tassium ions nigericin (Fig. 6a) is a non-electrogenic
ionophore and cannot uncouple mitochondria at low
concentrations; its main function is the induction of
potassium/H
+
-exchange. However, it was shown that
at high concentrations nigericin is capable of mito-
chondrial uncoupling, i.e. accelerating mitochondri-
al respiration [238]. Moreover, such well-known un-
coupler of oxidative phosphorylation as usnic acid
(UA) (Fig.  6a) [239,  240] is also an ionophore trans-
porting divalent cations [241]. Below the mechanism
of their protonophoric function will be discussed
in detail.
It was shown previously that nigericin at con-
centrations of tens of micromoles is able to stimulate
mitochondrial respiration and reduce the efficiency
of oxidative phosphorylation [238]. Acceleration of
ANTONENKO et al.1826
BIOCHEMISTRY (Moscow) Vol. 90 No. 12 2025
Fig. 6. Chemical structure of nigericin and usnic acid (UA)(a). Scheme of functioning of cation-dependent protonophores(b).
Scheme of functioning of complexes of fatty acids with lipophilic cations (c). BLM, bilayer lipid membrane.
respiration was clearly pronounced in the case of us-
ing succinate as a substrate of respiratory complexII,
while in the case of using substrates of respiratory
complex I (glutamate and malate) nigericin inhibit-
ed respiration of mitochondria [242]. Both stimula-
tion of respiration and its inhibition were observed
only in the presence of potassium ions in the me-
dium, while no effect was observed in presence of
other cations. This is in accordance with the cation
selectivity of nigericin as an ionophore. Inhibition of
respiration in the case of using glutamate and ma-
late could be possibly associated with inhibition of
transporters of these compounds in the mitochondrial
membrane.
Nigericin at micromolar concentrations increased
permeability of BLM proportionally to the square of
concentration [243]. The authors hypothesized that
this pattern of membrane permeability could be ex-
plained by formation of nigericin dimers in complex
with potassium ions and proton, in particular, pro-
ton is transported across BLM in the form of a neu-
tral complex with nigericin, while potassium cation
is transported in the charged form by the dimer of
protonated nigericin molecules.
Selectivity of ion transport mediated by nigeri-
cin was examined in the study by Markin and Soko-
lov[244] based on potential of the open circuit in the
presence of ion concentration gradient on the mem-
brane. This parameter usually varies in the range
from zero to the value calculated from the Nernst
equation, i.e. approximately 59  mV per 10-fold con-
centration gradient of monovalent ions. It was un-
expectedly observed by Markin and Sokolov that
in the case of nigericin a potential of opposite sign
is generated in the presence of the pH gradient,
while in the presence of the potassium ion gradient
MITOCHONDRIAL UNCOUPLERS 1827
BIOCHEMISTRY (Moscow) Vol. 90 No. 12 2025
a ‘super-Nernst’ potential is generated, i.e., potential
higher than 59 mV. The authors developed a theoret-
ical model, which described adequately the obtained
data [244]. According to their suggestion, the process
does not involve establishing of interfacial equilibri-
um, but equilibrium between the penetrating compo-
nents. In the case of complex co-transport of several
ions, as in the case of nigericin, its cessation could
occur without equalization of electrochemical poten-
tials of these ions, hence, the potential could differ
from the Nernst value. The obtained experimental
data are consistent with the model, implying that
transport stoichiometry of the ions is 2K
+
/1H
+
.
Usnic acid (Fig.  6a) is a secondary metabolite of
certain lichens, used as a supplement in the weight-
loss diets. It has been known for a long time that
UA uncouples mitochondria at micromolar concentra-
tions [239,245]. According to our data [241], the pres-
ence of calcium or magnesium ions in the medium
is necessary for induction of electric current across
BLM by UA; in the presence of the chelating agent
EDTA the current becomes zero. The BLM current is
proportional to the square of usnic acid concentra-
tion, which indicates the formation of UA dimers in
the course of the ion transport across BLM. Investi-
gation of selectivity of the UA-mediated ion transport
revealed that potentials in the case of pH gradient
were approximately 2-fold higher than the Nernst po-
tentials [246]. Use of the model suggested by Markin
and Sokolov for description of the UA-mediated ion
transport led to the conclusion that the main process
of this transport is electrogenic exchange of three
protons for calcium or magnesium ion (see scheme
in Fig. 6b).
The pentadecapeptide gramicidin A produced by
Bacillus brevis has the following amino acid sequence:
HCO-L-Val
1
-Gly
2
-L-Ala
3
-D-Leu
4
-L-Ala
5
-D-Val
6
-L-Val
7
-D-
Val
8
-L-Trp
9
-D-Leu
10
-L-Trp
11
-D-Leu
12
-L-Trp
13
-D-Leu
14
-L-
Trp
15
-NHCH
2
CH
2
OH; it is known as a channel-former
selective for monovalent cations [247-249]. It is often
used for dissipation of membrane potential in vari-
ous bioenergetic systems such as mitochondria [250],
bacteria [251], submitochondrial particles [252], and
chloroplasts [253]. As was shown, gramicidin  A also
can transport protons; and proton transport is 2 or-
ders of magnitude more effective than transport of
potassium ions [254]. However, in biological systems
gramicidin  A primarily functions as a potassium and
sodium ionophore, because amount of protons in
cells is approximately 7 orders of magnitude lower
than that of potassium ions. From this point of view,
it formally could be assigned to the group of cat-
ion-dependent protonophores. However, in reality,
the mechanism is fundamentally different involving
the formation of an ionic channel and transport of
protons along the hydrophilic wall of the channel
inside the membrane. Wide use of gramicidin  A as
an uncoupler is limited due to its high cytotoxicity
associated with equalization of potassium and sodi-
um ion gradients on the plasma membrane of cells
[255, 256], as well as by its low permeability across
cellular and subcellular membranes [253, 257]. Some
derivatives of gramicidin  A exhibit significantly lower
cytotoxicity, but, at the same time, maintain high pro-
tonophoric activity [255, 258]. It was shown that one
of such analogues with replacement of valine at the
first position with glutamic acid exhibits neuroprotec-
tive and nephroprotective properties, similar toother
uncouplers [259].
Complexes of fatty acids with hydrophobic
cations (and other transporters) as protonophores.
Certain amounts of fatty acids are always present in
biological membranes, which themselves cannot be
considered as protonophores due to low efficiency
of penetration of fatty acid anionic form across lipid
membranes. However, there are compounds that form
complexes with anions of fatty acids and to certain
extent facilitate their translocation across the mem-
brane. Among those relatively popular compounds
there are conjugates of decyltriphenylphosphonium
with plastoquinone or ubiquinone, which were named
SkQ1 (plastoquinoyldecyltriphenylphosphonium bro-
mide) and mitoQ (ubiquinonyldecyltriphenylphospho-
nium methanesulfonate), respectively. It was shown
that at micromolar concentrations SkQ1 (or dodecyl-
triphenylphosphonium C
12
TPP) uncouples mitochon-
dria due to interaction with endogenous fatty acids
[260]. In the review by Childressetal.[32] mitoQ and
C
12
TPP are described as usual protonophores, which
is not precisely correct. Below the mechanism of ac-
tion of these and similar compounds exhibiting the
uncoupling effect due to interaction with fatty acids
will be considered in more detail.
Complexes of fatty acids with lipophilic cations.
It was shown previously that fatty acids are capable
of forming complexes with lipophilic cations in lip-
id membranes [260, 261]. These complexes facilitate
diffusion of fatty acid anions across the membranes,
which results in protonophoric activity due to high
membrane permeability of the neutral form of fatty
acid. Scheme of this process is presented in Fig.  6c. De-
rivatives of alkyltriphenylphosphonium, rhodamine19,
rhodamine  B, and berberine could serve as lipophilic
cations [260-263], as well as some local anesthetics of
a cationic nature [264]. In general, it has been known
for a long time that hydrophobic cations form com-
plexes with hydrophobic anions in lipid membranes
[46, 264,265]. Interestingly enough, anions of classical
anionic uncouplers can play a role of such hydropho-
bic anions, and the addition of lipophilic cations re-
sults in significant acceleration of proton transfer by
CCCP and DNP [266-268].
ANTONENKO et al.1828
BIOCHEMISTRY (Moscow) Vol. 90 No. 12 2025
Derivatives of urea and thiourea. It was shown
using liposomes loaded with the pH indicator pyra-
nine that lipophilic cations in combination with
thiourea-containing compounds could transport hy-
drogen ions [269]. Previously, it was reported that
such compounds are transporters of inorganic anions,
including chloride ions [184]. Complexes of fatty acid
carboxyls with a thiourea group are assembled via
formation of several hydrogen bonds between them.
The uncoupling effect of one of the thiourea deriva-
tives, NT-1505 (N-allyl-N′,N′-dibenzyl-S-ethylthioisourea
hydroiodide) was demonstrated in the study by An-
tonenkoet al. [270] both in isolated mitochondria and
in the neuron culture. It was also shown that NT-1505
increases proton current across BLM in the presence
of palmitate. According to the studies by York et  al.
[186, 271], the ability of transporting fatty acid an-
ions is characteristic not only for the derivatives of
thiourea, but also for other urea derivatives such
as, for example, N,N′-bis(3,5-dichlorophenyl)urea also
known as compound SR4. Protonophoric properties of
SR4 have been described above in the section devot-
ed to NH-acids, i.e. these compounds could serve as
protonophores even without fatty acids.
Uncoupling via formation of defects in the
membrane. The process of uncoupling in mitochon-
dria implies preservation of membrane integrity
and formation of selective pathway for hydrogen
ions in the presence of the added proton transport-
er. However, the main signs of uncoupling, namely
stimulation of respiration and depolarization of the
mitochondrial membrane, could be also caused by
compounds inducing formation of non-selective pores
in lipid membranes [272-276], or even through di-
rect membrane disruption [277-279]. In some cases,
the true uncoupling and formation of non-selective
leakage is difficult to distinguish. For example, in
the case of anesthetic bupivacaine described above
it is not clear whether this compound is a cationic
protonophore, or it induces defects in the membrane.
Below this quite contradictory issue will be discussed
in more detail.
Detergent action and uncoupling. Classical action
of detergents occurs at concentrations above the crit-
ical micelle concentration (CMC) and results in com-
plete solubilization of membrane components (pro-
teins and lipids). However, the uncoupling effect of
classical detergents could be observed at concentra-
tions significantly lower than CMC [277-279]. This is
in line with the fact that detergents induce formation
of non-specific channels in BLMs at concentrations,
which are several orders of magnitude lower than
the concentration of micelle formation [280]. Under
these conditions, BLMs maintain their integrity and
are not disrupted when regular potentials are ap-
plied. Changes in spontaneous curvature of monolay-
ers in the bilayer due to non-lamellar structure of the
majority of detergents could play a significant role
in formation of such transient defects by lowering
the energy of formation of hydrophilic cavities and
transient channels in the membrane, which defines
its ionic permeability  [281]. As examples, the action
of minocycline  [282] and SkQ1  [283] could be consid-
ered, which cause leakage in both mitochondrial and
artificial membranes. Terada and colleagues [284] de-
scribed the induction of ionic transport through the
mitochondrial membrane and BLM by the dicationic
cyanine dye triS-C4(5) (2,2′-[5-(3-butyl-4-methyl-1,3-
thiazol-2-ylidene)penta-1,3-diene-1,3-diyl]bis[3-butyl-
4-methyl-1,3-thiazolium] diiodide), which depended
on the presence of phosphate in the medium. The
authors suggested that it is exactly the complex of
triS-C4(5) with phosphate that is capable of induc-
ing transient defects in the lipid membrane, which
manifested themselves in mitochondria as uncoupling
of oxidative phosphorylation.
Interaction of lauryl sulfate with UCP1 and ANT1
proteins. It was noted earlier in this review that an-
ionic uncouplers could interact with ANT1 and ac-
tivate proton leakage through this protein. It was
shown that this protein could also interact with sodi-
um lauryl (dodecyl) sulfate (SDS) causing uncoupling
in mitochondria, which is inhibited by the specific
inhibitor of ANT1 carboxyatractyloside [27]. Further-
more, it was shown that SDS stimulates proton leak-
age via the UCP1 protein [133]. In this experiment
the used concentrations of SDS (micromoles) were
significantly lower than CMC (~8  mM).
Nonprotonophoric uncoupling. Quite a few
agents are known that provide the pattern of un-
coupling (stimulation of respiration, cessation of ATP
synthesis, and drop of membrane potential) without
relation to the described above proton uncoupling.
A classic example is an effect of arsenate [285, 286]
Arsenate is a substrate of ATP synthase; ADP-arse-
nate, formed as a result of this reaction, is unstable
in water and decomposes rapidly to initial arsenate
and ADP. This causes stimulation of respiration in mi-
tochondria in the presence of ADP, which, however,
is not accompanied by ATP synthesis. Phenomeno-
logically, in the presence of arsenate, mitochondria
are transformed into the continuous ‘third’ state.
The Ca
2+
/H
+
-exchanger A23187 (calcimycin) could be
another example, which causes the uncoupling be-
cause the electrogenic calcium uniporter is present
in the mitochondrial membrane [287]. The list of
such examples could be continued, but they are not
the subject of this review. It should be also men-
tioned that freezing-thawing cycles transform mito-
chondria into the completely uncoupled state due to
the formation of multiple defects in its inner mem-
brane [288].
MITOCHONDRIAL UNCOUPLERS 1829
BIOCHEMISTRY (Moscow) Vol. 90 No. 12 2025
METABOLIC TRANSFORMATIONS
OF PROTONOPHORES IN CELLS
It is obvious that protonophores, similar to the
majority of xenobiotics, are subjected to metabolic
transformations in organisms. Chemical modifica-
tions of xenobiotics, including drugs, environmental
contaminants, or food supplements, which are com-
pounds foreign for cells, predominantly occur via en-
zymatic processes classified as phase  I and phase  II
reactions. These metabolic pathways are directed
towards increasing hydrophilicity of xenobiotics,
which facilitates their excretion from an organism by
phase  III transporters. Phase  I reactions include func-
tionalization of a compound via oxidation, reduction,
or hydrolysis, which makes the molecule more reac-
tive in the subsequent conjugation during phase  II.
The key player in oxidation is the family of cyto-
chrome P450 enzymes, which introduce polar groups
such as hydroxyl. For example, oxidation of benzene
derivatives to phenol mediated by CYP450 increases
their solubility. The reactions of reduction are less
common; they involve transformation of nitro groups
into amines. Hydrolytic enzymes, such as esterases,
cleave ester or amide bonds, as occurs with aspirin,
which is transformed into salicylic acid [289]. It is
remarkable that in phase  I super-reactive intermedi-
ate products, such as epoxides, sometimes could be
formed, which could exert toxic effects, if they are
not detoxified effectively.
Phase II reactions include conjugation of wa-
ter-soluble fragments with initial compounds or with
compounds modified during phase I. Glucuronization
catalyzed by uridine diphosphate (UDP)-glucurono-
syltransferase (UGT) attaches glucuronic acid from
UDP-glucuronic acid to substrates [290] such as mor-
phine forming highly soluble, excretable glucuronides
[291]. Sulfotransferases (SULT) transfer sulfate groups
from 3′-phosphoadenosine-5′-phosphosulfate (PAPS) to
phenols and alcohols [292], as in the case of sulfa-
tion of paracetamol. Glutathione-S-transferases (GST)
link glutathione with electrophilic centers [293], thus
neutralizing metabolites formed, for example, in the
case of acetaminophen overdose. N-acetyltransfer-
ases (NAT) catalyze the transfer of an acetyl group
from acetyl-CoA to aromatic amines [294], while
methyltransferases, such as catechol-O-methyltrans-
ferase (COMT), catalyze the transfer of a methyl
group from S-adenosylmethionine (SAM) to substrates
such as catecholamines [295]. Conjugation with ami-
no acids, although less common, links xenobiotics
with glycine or taurine; one of the examples of such
processes is excretion of benzoic acid with formation
of hippuric acid.
Concerning metabolic transformations of proto-
nophoric uncouplers, the following reactions could
be of interest: phase  I reactions involving hydrolysis
of ester bonds catalyzed, in particular, by esterases;
phase II reactions involving glutathionylation. Among
the enzymes with esterase activity, in this review we
will consider only enzymes that are primarily in-
volved in detoxification and metabolism of xenobi-
otics via hydrolysis of ester bonds. Carboxylesterases
(CES), members of the superfamily of serine hydro-
lases, are representatives of this group. CES1 and
CES2, main isoforms in humans, hydrolyze substances
containing esters (such as clopidogrel, irinotecan) and
toxic compounds from the environment. They belong
to the α/β-hydrolase-fold family with the catalytic
triad (Ser-His-Glu/Asp) and conserved oxyanion hole
(for stabilization of negative charge at the deprotonat-
ed oxygen in the intermediate state), which provides
broad substrate specificity. CES enzymes are localized
in endoplasmic reticulum and plasma, where they
process lipophilic esters into hydrophilic metabolites
for excretion. There are also data on CES activity as-
sociated with mitochondria [296, 297].
Glutathionylation involved in xenobiotic metabo-
lism is associated primarily with conjugation of GSH
with electrophilic xenobiotics, which represents a
critical mechanism of detoxification mediated by glu-
tatione-S-transferases. This process, a part of the mer-
capturate pathway, neutralizes harmful compounds
by increasing their solubility in water, facilitating
excretion through urine or bile [298]. The reaction
involves a nucleophilic attack of the thiole group in
GSH on electrophilic centers of xenobiotics forming
glutathione-S-conjugates. These conjugates are further
processed by enzymes such as γ-glutamyl transferase
and dipeptidase for the production of cysteine S-con-
jugates, which are further acetylated with formation
of mercapturic acids (conjugates of N-acetylcysteine)
for elimination [298]. Glutathione-S-transferases com-
prise a superfamily of enzymes playing a central role
in metabolism of phaseII via conjugation of the GSH
tripeptide (γ-glutamyl-cysteinyl-glycine) with electro-
philic xenobiotics and endogenous toxins. The conju-
gation increases solubility of these compounds facili-
tating their excretion via phase III transporters, such
as proteins associated with multidrug resistance.
Ester derivatives of 7-hydroxycoumarin and
esterases. Structure and uncoupling properties of
7-hydroxycoumarin ester derivatives have been de-
scribed above. In particular, rapid hydrolysis of the
compounds of this series in RLM and the observed
tissue specificity of the inactivation was mentioned
before. 7-Hydroxycoumarin is a natural pH-dependent
fluorophore, which is derived from medicinal plants
of the Umbelliferae family (hence, the second name –
umbelliferone) and some others. We have synthesized
two series of esters: with umbelliferone-3-carbox-
ylic acid and umbelliferone-4-acetic acid [124, 125].
ANTONENKO et al.1830
BIOCHEMISTRY (Moscow) Vol. 90 No. 12 2025
Compounds of both series exhibited pronounced un-
coupling activity in mitochondria. Surprisingly, this
activity disappeared within minutes, unlike the case
of classical uncouplers DNP and CCCP. Taking into
account that protonophoric activity of the esters in
model membranes (BLMs and liposomes) did not
change with time, it was suggested that the uncou-
pling activity in mitochondria disappeared due to
enzymatic hydrolysis of the esters. Indeed, thin lay-
er chromatography (TLC) showed that the umbellif-
erone-containing esters, incubated in the presence
of isolated RLM, were almost completely converted
into initial acids within minutes, while no hydrolysis
was observed in the case of fluorescein octyl ester
[118]. We hypothesized that the mitochondrial alde-
hyde dehydrogenase ALDH2 could be a good candi-
date for the role of an enzyme catalyzing hydrolysis
of the umbelliferone-containing esters, because this
enzyme exhibits high esterase activity [299]. More-
over, this activity is sensitive to the same inhibitors
as the main activity of this enzyme – oxidation of
acetaldehyde. The known inhibitor of aldehyde de-
hydrogenases disulfiram suppressed the drop in the
uncoupling activity of the umbelliferone esters [124].
An alternative candidate of such enzymatic activity is
the carboxyl esterase CES1, which plays a significant
role in metabolism of xenobiotics. Its activity is also
inhibited by disulfiram [300, 301]. Certain isoforms
of carboxyl esterases are located predominantly in
liver, while only low amounts of these enzymes are
detected in heart and kidney, which could explain
the tissue specificity of the mitochondrial uncoupling
by the umbelliferone derivatives, namely: the sponta-
neous disappearance of the uncoupling activity of the
umbelliferone esters found in RLM was not observed
in rat heart and kidney mitochondria [125]. Hence,
the umbelliferone esters could be considered as tis-
sue-specific uncouplers.
Diarylamines and glutathione-S-transferases.
Above we have already discussed diarylamines, a
group of compounds with some representatives exhib-
iting properties of anionic uncouplers due to an abili-
ty of their NH group to lose a proton. The best-known
compound among them is fluazinam (Fig.  3), which
uncouples mitochondria at nanomolar concentrations
[155, 174, 175]. This compound is a broad-spectrum
fungicide widely used in agriculture. In the pioneer
study published in 1991 it was shown that the time
course of oxygen consumption by isolated RLM after
the addition of fluazinam is biphasic, namely: stim-
ulation of mitochondrial respiration by fluazinam is
followed byreversal of the respiration rate to the ini-
tial level on the minute time scale [174]. Efficiency
of the reversal depended significantly on the level of
glutathione (GSH) in mitochondria, and in the pres-
ence of GSH-depleting agents the reversal practically
disappeared. The author suggested that the resto-
ration of the respiration rate is associated with conju-
gation of fluazinam with GSH, which is catalyzed by
mitochondrial glutathione-S-transferase. The analog of
fluazinam B-3 (N-[2,4-dinitro-6-trifluoromethyl-3-chlo-
rophenyl]-5-(trifluoromethyl)pyridine-2-amine), dis-
covered in the study by Brandt et  al.  [175], also ex-
hibited very high uncoupling activity, but, unlike
fluazinam itself, was not metabolized by RLM due to
the absence of electrophilic substituent in the third
position of the benzene ring. According to the data
reported by Clarke et  al.  [9], fluazinam was found to
be the most reactive substrate of glutathione-S-trans-
ferase among the very wide range of agrochemical
compounds. In our study [173], the addition of GSH
to RLM enhanced the fluazinam inactivation. The
formation of the conjugates of fluazinam and GSH
was detected experimentally using methods of TLC
and chromato-mass spectrometry (LC-MS): catalytic
replacement of chlorine and one of the nitro groups
with glutathione occurred in the process. It is worth
noting that the phase of fluazinam inactivation is ab-
sent in the case of rat heart mitochondria, i.e. fluazi-
nam exhibits the properties of a tissue-specific un-
coupler [173].
N-phenylthiophenamines and glutathione-S-
transferases. N-phenylthiophenamines comprise a
class of organic compounds having a thiophene ring
(5-membered aromatic heterocycle containing sulfur
atom) attached through an amino group to a phenyl
ring. Interestingly enough, in 1970 the paper was pub-
lished by Buechel and Schaefer [302] describing ef-
fects of a large series of anilinothiophene derivatives
on isolated RLM. The study mainly concentrated on
three groups of compounds: 2-anilino-3,5-dinitrothio-
phenes, 2-anilino-5-halo-3,4-dinitrothiophenes and de-
rivatives of 2-hydroxy-3,5-dinitrothiophene. Buechel
and Schaefer[302] revealed the relationship between
the structure and activity, demonstrating the manda-
tory requirement of NH group for the uncoupling ac-
tivity: the absence of this group resulted in the com-
plete loss of activity. Electron acceptor substituents in
the phenyl ring, such as nitro (-NO
2
), trifluoromethyl
(-CF
3
), and chlorine, enhanced the activity likely due
to lowering of pK
a
. It is worth mentioning that some
of these compounds exhibited properties similar to
those described later for fluazinam [174, 177], name-
ly, the ability to stimulate mitochondrial respiration,
which disappeared within minutes. However, the au-
thors explained this phenomenon not by chemical
modification of anilinothiophenes, but by their inter-
action with non-identified components of the mito-
chondrial respiratory chain. Noteworthy, the study by
Buechel and Schaefer [302] (1970) was conducted in
the era, when the modern ideas of bioenergetics just
started to emerge. The biphasic uncoupling action of
MITOCHONDRIAL UNCOUPLERS 1831
BIOCHEMISTRY (Moscow) Vol. 90 No. 12 2025
Fig. 7. Chemical structures of BDNT and its adduct with glutathione.
5-bromo-3,4-dinitro-N-(4-nitrophenyl)thiophen-2-amine
(BDNT) in RLM was investigated in great detail in our
recent study [176]. The restoration of the membrane
potential level after the BDNT-induced decrease be-
came more pronounced upon the addition of GSH
to incubation medium and practically disappeared
upon complete depletion of intramitochondrial GSH
pool. In the case of rat heart mitochondria (RHM),
the restoration of the decreased membrane poten-
tial did not occur. By using capillary electrophoresis
and LC-MS, the formation of BDNT conjugates with
glutathione (Fig. 7) was found upon incubation with
RLM, and these conjugates were absent in the case
of incubation with RHM. In was concluded that BDNT
is a substrate of the glutathione-S-transferase (GST)
enzyme, which catalyzed the formation of the BDNT
conjugate with GSH: similar to the case of fluazinam,
the nitro group of thiophene is replaced with gluta-
thione. Higher expression of GST or predominance of
another isoform in liver as compared to heart could
be responsible for the tissue-specific action of BDNT.
It should be mentioned that BDNT caused depolariza-
tion of mitochondria in the culture of fibroblasts, but
not in liver cells (HepG2). BDNT induced proton-selec-
tive current through planar BLM. The current inten-
sity decreased upon the addition of phloretin, which
indicated the anionic nature of the protonophore. The
pK
a
value of BDNT was found to be 7.38.
CONCLUSIONS
By classifying mitochondrial uncouplers, we
were able for the first time to systematize numerous
examples of uncouplers present in the literature. This
allowed us to review the chemical diversity of uncou-
plers, as well as to identify gaps in the table of chem-
ical structures. In particular, among the known zwit-
terionic uncouplers there are many examples with
a proton cycle performed by the pair zwitterion-cat-
ion, while there are yet no examples with a proton
cycle performed by the pair zwitterion- anion in the
literature (Fig.  1). Such a compound could consist of
triphenylborate and a fragment containing amino
group capable of protonating-deprotonating under
physiological pH. It should be examined in future,
how effective this molecule (or the similar one) is in
transporting hydrogen ions across lipid membranes.
Such ‘anionic’ zwitterionic protonophores could be
much more active than ‘cationic’ zwitterionic proto-
nophores, because anions are transported significant-
ly more effectively across lipid membranes as com-
pared to cations of the similar structure due to the
presence of membrane dipole potential.
A goal in the search for the most active uncou-
plers is usually their selective action in the uncou-
pling of oxidative phosphorylation without affecting
other processes vital for the cell functioning. Pene-
tration of these compounds across the plasma mem-
brane and their predominant accumulation in mito-
chondria are essential issues. In this regard, cationic
uncouplers as well as zwitterionic uncouplers that
accumulate in mitochondria due to the inner mem-
brane potential of approximately −180  mV should
have a certain advantage. However, this advantage is
offset by the weak protonophoric activity of cationic
uncouplers; likely, this is the reason why most of the
commonly used uncouplers belong to the class of an-
ionic protonophores.
On the other hand, as has been mentioned pre-
viously [33], a therapeutic effect of uncouplers could
be significantly improved by using the compounds
that have advantages in certain tissues. In particu-
lar, the use of DNP ethers for treating non-alcohol-
ic steatohepatitis (NASH) was demonstrated to be
successful [15]. Ethyl and methyl ethers of DNP are
easily absorbed from intestine to blood and are de-
livered to liver, where they are rapidly transformed
ANTONENKO et al.1832
BIOCHEMISTRY (Moscow) Vol. 90 No. 12 2025
into DNP by hydrolases. Hence, in this case the un-
coupling agents emerge locally, which should reduce
side effects of DNP. This means that application of
tissue-specific uncouplers is of great interest in med-
icine. Ideally, such uncouplers could actively work in
one tissue type, while exhibiting low activity (ideally
no activity at all) in other tissues. In the series of
studies on the uncoupling activity of fluazinam and
anilinothiophenes, it was demonstrated that these
compounds are subjected to rapid glutathionylation
in liver, while in heart and kidney their activity is
preserved [173,  176]. In another series of studies,
the tissue-specific activity was demonstrated for the
derivatives of 7-hydroxycoumarin [125]. Hence, tis-
sue-specific inactivation together with formation of
an active uncoupler in the specific tissue could be
also used for achieving partial or complete tissue
specificity of the corresponding uncouplers.
Another approach in the search for tissue-spe-
cific uncouplers is the use of ability of certain com-
pounds to mediate proton transfer across membranes
via interaction with the proteins of the inner mi-
tochondrial membrane, primarily with the protein
transporters of the SLC25 family. In particular, it was
shown that DNP uncouples mitochondria partially via
interaction with the adenine nucleotide translocator
ANT1 [26, 27, 63]. Considering that the patterns of
ANT1 expression differ significantly in different tis-
sues, the ability of DNP to uncouple mitochondria
could vary significantly: it is shown that the uncou-
pling activity of DNP is significantly higher in heart
mitochondria than in liver mitochondria [63]. Hence,
even such widely known and used uncoupler as DNP
could be considered as a tissue-specific uncoupler. In
this regard, fatty acids could be also considered as
uncouplers specific for brown adipose tissue due to
participation of the UCP1 protein in their uncoupling
activity. Further studies should reveal uncouplers
with higher tissue specificity than DNP, moreover,
the protein partner of such an uncoupler should not
be necessary ANT1. In particular, it was shown that
the uncoupling effect of the cationic uncoupler C4R1
involves the proton ATPase of mitochondria [200].
Vladimir Petrovich Skulachev has strongly boost-
ed studies of uncouplers by suggesting the concept
of ‘mild uncoupling’ [286, 303]. This concept is based
on the steep dependence of the ROS formation rate
on mitochondrial membrane potential. Considering
that organism aging is often associated with oxida-
tive stress and accumulation of oxidation products,
uncouplers could cause an increase in life span.
Indeed, it was shown that low doses of DNP reliably
increase life span of rats [19], mice [20], yeast [21],
and Drosophila flies [22]. Investigation of life span of
animals is a rather complex experimental task; how-
ever, fortunately, uncouplers exhibit therapeutic prop-
erties, which are easier to investigate, with many of
them mentioned above. One could hope that all the
facts taken together would make this review useful
for readers and inspire researchers to further inves-
tigate new uncouplers and elucidate mechanisms of
their action.
Abbreviations
ANT1 adenine nucleotide translocator 1 (ATP/
ADP-antiporter)
BDNT 5-bromo-3,4-dinitro-N-(4-nitrophenyl)
thiophen-2-amine
C4R1 butyl ester of Rhodamine 19
CCCP carbonyl cyanide 3 chlorophenylhydra-
zone
CES carboxylesterases
DNP 2,4-dinitrophenol
DNPME 1-methoxy-2,4-dinitrobenzene
o-F16 F16, and m-F16, (E)-2-, (E)-3-, and
(E)-4-(3-indolylvinyl)-N-methylpyridini-
um iodide, respectively
FCCP carbonyl cyanide p-trifluoromethoxy-
phenylhydrazone
GSH glutathione
HU6 5-[(2′,4′-dinitrophenoxy)methyl]-1-meth-
yl-2-nitroimidazole
BLM bilayer lipid membrane
mitoCCCP 10-([4′-(dicyanomethylene)hydraz-
inyl-2′-chlorophenyl]oxy)decyltriphen-
ylphosphonium bromide
mitoDNP 3-(4′-hydroxy-3′,5′-dinitrophenyl)prop-
yltriphenylphosphonium methanesul-
fonate
mitoFluo 10-[(2-(6-hydroxy-3-oxo-3H-xanthen-
9-yl)benzoyl)oxy]decyltriphenylphos-
phonium bromide
NAC N-acetyl cysteine
PCBT pentachlorothiophenol
PDTC-9 nonyl 3-picolinoyldithiocarbazate
S13 3-tert-butyl-4′-nitro-2′,5-dichlorosalicy-
lanilide
SF6847 [(3,5-di-tert-butyl-4-hydroxyphenyl)
methylidene]propanedinitrile
SF-C5-TPP (6-[3′,5′-di-tert-butyl-4′-hydroxyphenyl]-
7,7-dicyanohept-6-en-1-yl)triphenyl-
phosphonium bromide
SkQ1 10-(6′-plastoquinonyl)decyltriphenyl-
phosphonium
SR4 N,N′-bis(3,5-dichlorophenyl)urea
UA usnic acid
UCP uncoupling proteins
Contributions
Y. N. Antonenko – concept and supervision of the
work; Y. N.Antonenko, E. A.Kotova, V. S.Krasnov, and
R. S.Kirsanov – writing and editing of the paper.
MITOCHONDRIAL UNCOUPLERS 1833
BIOCHEMISTRY (Moscow) Vol. 90 No. 12 2025
Funding
The study was conducted under the state assign-
ment of Lomonosov Moscow State University (no.
AAAA-A19-119031390114-5).
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 declare that they have
noconflicts of interest.
REFERENCES
1. Ovchinnikov, Yu. A., Ivanov, V. T., and Shkrob, A. M.,
Membrane active complexons [in Russian], Moscow:
Nauka, 1974.
2. Henderson, P. J. F., McGivan, J. D., and Chappell,
J. B. (1969) The action of certain antibiotics on mi-
tochondrial, erythrocyte and artificial phospholipid
membranes. The role of induced proton permeabil-
ity, Biochem. J., 111, 521-535, https://doi.org/10.1042/
bj1110521.
3. Sato,T., Konno,H., Tanaka,Y., Kataoka, T., Nagai, K.,
Wasserman, H. H., and Ohkuma, S. (1998) Prodigios-
ins as a new group of H
+
/Cl
symporters that uncouple
proton translocators, J.Biol. Chem., 273, 21455-21462,
https://doi.org/10.1074/jbc.273.34.21455.
4. Mitchell, P., and Moyle, J. (1967) Acid-base titration
across the membrane system of rat-liver mitochon-
dria. Catalysis by uncouplers, Biochem. J., 104, 588-
600, https://doi.org/10.1042/bj1040588.
5. Skulachev, V. P. (1970) Electric fields in coupling
membranes, FEBS Lett., 11, 301-308, https://doi.org/
10.1016/0014-5793(70)80554-3.
6. Ranga Rao, K. (1978) Pentachlorophenol: Chemistry,
pharmacology, and environmental toxicology, Plenum
Press, New York.
7. Hollingworth, R.M., and Gadelhak, G.G. (1998) Mech-
anisms of action and toxicity of new pesticides that
disrupt oxidative phosphorylation, Rev. Toxicol., 2,
253-266.
8. Palmeira, C. M., Moreno, A. J., and Madeira, V. M.
(1994) Interactions of herbicides 2,4-D and dinoseb
with liver mitochondrial bioenergetics, Toxicol.
Appl. Pharmacol., 127, 50-57, https://doi.org/10.1006/
taap.1994.1138.
9. Clarke, E. D., Greenhow, D. T., and Adams, D.
(1998) Metabolism-related assays and their appli-
cation to agrochemical research: reactivity of pes-
ticides with glutathione and glutathione transferas-
es, Pestic. Sci., 54, 385-393, https://doi.org/10.1002/
(SICI)1096-9063(199812)54:4<385::AID-PS842>
3.0.CO;2-C.
10. Cadenas, S. (2018) Mitochondrial uncoupling, ROS
generation and cardioprotection, Biochim. Biophys.
Acta Bioenerg., 1859, 940-950, https://doi.org/10.1016/j.
bbabio.2018.05.019.
11. Geisler, J. G. (2019) 2,4 Dinitrophenol as medicine,
Cells, 8, E280, https://doi.org/10.3390/cells8030280.
12. Zorov, D. B., Andrianova, N. V., Babenko, V. A.,
Pevzner, I.B., Popkov, V.A., Zorov, S.D., Zorova, L.D.,
Plotnikov, E. Y., Sukhikh, G. T., and Silachev, D. N.
(2021) Neuroprotective potential of mild uncoupling
in mitochondria. Pros and Cons, Brain Sci., 11, 1050,
https://doi.org/10.3390/brainsci11081050.
13. Plotnikov, E. Y., Silachev, D. N., Jankauskas, S. S.,
Rokitskaya, T. I., Chupyrkina, A. A., Pevzner, I. B.,
Zorova, L.D., Isaev, N.K., Antonenko, Y.N., Skulachev,
V.P., and Zorov, D.B. (2012) Mild uncoupling of respi-
ration and phosphorylation as a mechanism provid-
ing nephro- and neuroprotective effects of penetrating
cations of the SkQ family, Biochemistry (Moscow), 77,
1029-1037, https://doi.org/10.1134/S0006297912090106.
14. Rai,Y., Anita, Kumari,N., Singh,S., Kalra,N., Soni,R.,
and Bhatt, A. N. (2021) Mild mitochondrial uncou-
pling protects from ionizing radiation induced cell
death by attenuating oxidative stress and mitochon-
drial damage, Biochim. Biophys. Acta Bioenerg., 1862,
148325, https://doi.org/10.1016/j.bbabio.2020.148325.
15. Perry, R. J., Zhang, D., Zhang, X. M., Boyer, J. L., and
Shulman, G.I. (2015) Controlled-release mitochondrial
protonophore reverses diabetes and steatohepatitis in
rats, Science, 347, 1253-1256, https://doi.org/10.1126/
science.aaa0672.
16. Tao,H., Zhang,Y., Zeng,X., Shulman, G.I., and Jin,S.
(2014) Niclosamide ethanolamine-induced mild mito-
chondrial uncoupling improves diabetic symptoms
in mice, Nat. Med., 20, 1263-1269, https://doi.org/
10.1038/nm.3699.
17. Kanemoto, N., Okamoto, T., Tanabe, K., Shimada, T.,
Minoshima, H., Hidoh, Y., Aoyama, M., Ban, T.,
Kobayashi, Y., Ando, H., Inoue, Y., Itotani, M., and
Sato,S. (2019) Antidiabetic and cardiovascular bene-
ficial effects of a liver-localized mitochondrial uncou-
pler, Nat. Commun., 10, 2172, https://doi.org/10.1038/
s41467-019-09911-6.
18. Shrestha, R., Johnson, E., and Byrne, F. L. (2021) Ex-
ploring the therapeutic potential of mitochondrial
uncouplers in cancer, Mol. Metab., 51, 101222, https://
doi.org/10.1016/j.molmet.2021.101222.
19. Tainter, M. L. (1938) Growth, life-span and food
intake of white rats fed dinitrophenol through-
out life, J.Pharmacol. Exp. Ther., 63, 51-57, https://
doi.org/10.1016/S0022-3565(25)08958-X.
20. Caldeira da Silva, C.C., Cerqueira, F.M., Barbosa, L.F.,
Medeiros, M.H., and Kowaltowski, A.J. (2008) Mild mi-
tochondrial uncoupling in mice affects energy metab-
olism, redox balance and longevity, Aging Cell, 7, 552-
560, https://doi.org/10.1111/j.1474-9726.2008.00407.x.
ANTONENKO et al.1834
BIOCHEMISTRY (Moscow) Vol. 90 No. 12 2025
21. Barros, M. H., Bandy, B., Tahara, E. B., and Kowal-
towski, A. J. (2004) Higher respiratory activity de-
creases mitochondrial reactive oxygen release and in-
creases life span in Saccharomyces cerevisiae, J.Biol.
Chem., 279, 49883-49888, https://doi.org/10.1074/
jbc.M408918200.
22. Padalko, V.I. (2005) Uncoupler of oxidative phosphor-
ylation prolongs the lifespan of Drosophila, Biochem-
istry (Moscow), 70, 986-989, https://doi.org/10.1007/
s10541-005-0213-1.
23. Setty, O. H., Hendler, R. W., and Shrager, R. I. (1983)
Simultaneous measurements of proton motive force,
delta pH, membrane potential, and H
+
/O ratios in in-
tact Escherichia coli, Biophys. J., 43, 371-381, https://
doi.org/10.1016/S0006-3495(83)84360-4.
24. Westerhoff, H. V., Juretic, D., Hendler, R. W., and
Zasloff, M. (1989) Magainins and the disruption of
membrane-linkedfree-energy transduction, Proc.
Natl. Acad. Sci. USA, 86, 6597-6601, https://doi.org/
10.1073/pnas.86.17.6597.
25. Jung, D. W., Davis, M. H., and Brierley, G. P. (1989)
Estimation of matrix pH in isolated heart mitochon-
dria using a fluorescent probe, Anal. Biochem., 178,
348-354, https://doi.org/10.1016/0003-2697(89)90651-9.
26. Bertholet, A.M., Natale, A.M., Bisignano,P., Suzuki,J.,
Fedorenko,A., Hamilton,J., Brustovetsky,T., Kazak,L.,
Garrity, R., Chouchani, E. T., Brustovetsky, N.,
Grabe,M., and Kirichok, Y. (2022) Mitochondrial un-
couplers induce proton leak by activating AAC and
UCP1, Nature, 606, 180-187, https://doi.org/10.1038/
s41586-022-04747-5.
27. Andreyev, A. Y., Bondareva, T. O., Dedukhova, V. I.,
Mokhova, E. N., Skulachev, V. P., Tsofina, L. M.,
Volkov, N. I., and Vygodina, T. V. (1989) The ATP/
ADP-antiporter is involved in the uncoupling effect
of fatty acids on mitochondria, Eur. J. Biochem.,
182, 585-592, https://doi.org/10.1111/j.1432-1033.1989.
tb14867.x.
28. Khailova, L. S., Kirsanov, R. S., Rokitskaya, T. I.,
Krasnov, V. S., Korshunova, G. A., Kotova, E. A., and
Antonenko, Y. N. (2024) Mitochondrial uncoupling
caused by a wide variety of protonophores is differ-
ently sensitive to carboxyatractyloside in rat heart
and liver mitochondria, Biochim. Biophys. Acta Bio-
energ., 1865, 149506, https://doi.org/10.1016/j.bbabio.
2024.149506.
29. Weinbach, E. C., and Garbus, J. (1969) Mechanism
of action of reagents that uncouple oxidative phos-
phorylation, Nature, 221, 1016-1018, https://doi.org/
10.1038/2211016a0.
30. Hanstein, W.G. (1976) Uncoupling of oxidative phos-
phorylation, Biochim. Biophys. Acta, 456, 129-148,
https://doi.org/10.1016/0304-4173(76)90010-0.
31. Katre, N. V., and Wilson, D. F. (1978) Interaction of
uncouplers with the mitochondrial membrane: iden-
tification of the high affinity binding site, Arch. Bio-
chem. Biophys., 191, 647-656, https://doi.org/10.1016/
0003-9861(78)90403-4.
32. Childress, E. S., Alexopoulos, S. J., Hoehn, K. L., and
Santos, W. L. (2018) Small molecule mitochondrial
uncouplers and their therapeutic potential, J. Med.
Chem., 61, 4641-4655, https://doi.org/10.1021/acs.jmed-
chem.7b01182.
33. Goedeke, L., and Shulman, G. I. (2021) Therapeutic
potential of mitochondrial uncouplers for the treat-
ment of metabolic associated fatty liver disease and
NASH, Mol. Metab., 46, 101178, https://doi.org/10.1016/
j.molmet.2021.101178.
34. Kotova, E.A., and Antonenko, Y.N. (2022) Fifty years
of research on protonophores: mitochondrial uncou-
pling as a basis for therapeutic action, Acta Naturae,
14, 4-13, https://doi.org/10.32607/actanaturae.11610.
35. Dos Santos, B. G., Brisnovali, N. F., and Goedeke, L.
(2024) Biochemical basis and therapeutic poten-
tial of mitochondrial uncoupling in cardiometabol-
ic syndrome, Biochem. J., 481, 1831-1854, https://
doi.org/10.1042/BCJ20240005.
36. Samartsev, V.N., Semenova, A.A., Belosludtsev, K.N.,
and Dubinin, M. V. (2023) Modulators reducing the
efficiency of oxidative ATP synthesis in mitochon-
dria: protonophore uncouplers, cyclic redox agents,
and decouplers, Biophys. Rev., 15, 851-857, https://
doi.org/10.1007/s12551-023-01160-8.
37. McLaughlin, S. G., and Dilger, J. P. (1980) Transport
of protons across membranes by weak acids, Physi-
ol. Rev., 60, 825-863, https://doi.org/10.1152/phys-
rev.1980.60.3.825.
38. Terada, H. (1981) The interaction of highly active
uncouplers with mitochondria, Biochim. Biophys.
Acta, 639, 225-242, https://doi.org/10.1016/0304-4173
(81)90011-2.
39. McLaughlin, S. (1972) The mechanism of action of
DNP on phospholipid bilayer membranes, J. Membr.
Biol., 9, 361-372, https://doi.org/10.1007/BF01868062.
40. Borisova, M. P., Ermishkin, L. N., Liberman, E. A.,
Silberstein, A. Y., and Trofimov, E. M. (1974) Mech-
anism of conductivity of bimolecular lipid mem-
branes in the presence of tetrachlorotrifluorometh-
ylbenzimidazole, J. Membr. Biol., 18, 243-261, https://
doi.org/10.1007/BF01870115.
41. Foster, M., and McLaughlin, S. (1974) Complexes
between uncouplers of oxidative phosphorylation,
J. Membr. Biol., 17, 155-180, https://doi.org/10.1007/
BF01870177.
42. Smejtek,P., Hsu,K., and Perman, W.H. (1976) Electri-
cal conductivity in lipid bilayer membranes induced
by pentachlorphenol, Biophys.J., 16, 319-336, https://
doi.org/10.1016/S0006-3495(76)85691-3.
43. Wilson, D.F., Ting, H.P., and Koppelman, M.S. (1971)
Mechanism of action of uncouplers of oxidative
phosphorylation, Biochemistry, 10, 2897-2902, https://
doi.org/10.1021/bi00791a016.
MITOCHONDRIAL UNCOUPLERS 1835
BIOCHEMISTRY (Moscow) Vol. 90 No. 12 2025
44. Dilger,J., and McLaughlin,S. (1979) Proton transport
through membranes induced by weak acids: a study
of two substituted benzimidazoles, J.Membr. Biol., 46,
359-384, https://doi.org/10.1007/BF01868755.
45. Barstad, A. W., Peyton, D. H., and Smejtek, P. (1993)
AHA – heterodimer of a class-2 uncoupler – penta-
chlorophenol, Biochim. Biophys. Acta, 1140, 262-270,
https://doi.org/10.1016/0005-2728(93)90065-N.
46. Liberman, E.A., and Topaly, V.P. (1969) Permeability
of biomolecular phospholipid membranes for fat-sol-
uble ions [in Russian], Biofizika, 14, 452-461.
47. Brockman, H. (1994) Dipole potential of lipid mem-
branes, Chem. Phys. Lipids, 73, 57-79, https://
doi.org/10.1016/0009-3084(94)90174-0.
48. Gawrisch, K., Ruston, D., Zimmerberg, J., Parsegian,
V.A., Rand, R. P., and Fuller,N. (1992) Membrane di-
pole potentials, hydration forces, and the ordering of
water at membrane surfaces, Biophys. J., 61, 1213-
1223, https://doi.org/10.1016/S0006-3495(92)81931-8.
49. Markin, V. S., and Chizmadzhev, Yu. A. (1974) Induced
ion transport [in Russian], Nauka, Moscow.
50. Firsov, A. M., Popova, L. B., Khailova, L. S., Nazarov,
P.A., Kotova, E.A., and Antonenko, Y. N. (2021) Pro-
tonophoric action of BAM15 on planar bilayers, li-
posomes, mitochondria, bacteria and neurons, Bio-
electrochemistry, 137, 107673, https://doi.org/10.1016/
j.bioelechem.2020.107673.
51. Miyoshi,H., Tsujishita,H., Tokutake,N., and Fujita,T.
(1990) Quantitative analysis of uncoupling activi-
ty of substituted phenols with a physicochemical
substituent and molecular parameters, Biochim.
Biophys. Acta, 1016, 99-106, https://doi.org/10.1016/
0005-2728(90)90011-R.
52. Spycher, S., Escher, B. I., and Gasteiger, J. (2005)
A quantitative structure–activity relationship model
for the intrinsic activity of uncouplers of oxidative
phosphorylation, Chem. Res. Toxicol., 18, 1858-1867,
https://doi.org/10.1021/tx050166j.
53. Spycher, S., Smejtek, P., Netzeva, T. I., and Escher,
B. I. (2008) Toward a class-independent quantitative
structure–activity relationship model for uncouplers
of oxidative phosphorylation, Chem Res. Toxicol.,
21, 911-927, https://doi.org/10.1021/tx700391f.
54. Martineau, L. C. (2012) Simple thermodynamic mod-
el of unassisted proton shuttle uncoupling and pre-
diction of activity from calculated speciation, lipo-
philicity, and molecular geometry, J.Theor. Biol., 303,
33-61, https://doi.org/10.1016/j.jtbi.2012.02.032.
55. Naven, R. T., Swiss, R., Klug-McLeod, J., Will, Y., and
Greene,N. (2013) Thedevelopment of structure-activ-
ity relationships for mitochondrial dysfunction: un-
coupling of oxidative phosphorylation, Toxicol. Sci.,
131, 271-278, https://doi.org/10.1093/toxsci/kfs279.
56. Ebert,A., and Goss, K.-U. (2020) Predicting uncoupling
toxicity of organic acids based on their molecular
structure using a biophysical model, Chem. Res. Tox-
icol., 33, 1835-1844, https://doi.org/10.1021/acs.chem-
restox.0c00063.
57. Liberman, E. A., Topaly, V. P., Tsofina, L. M., Jasaitis,
A. A., and Skulachev, V. P. (1969) Mechanism of cou-
pling of oxidative phosphorylation and the mem-
brane potential of mitochondria, Nature, 222, 1076-
1078, https://doi.org/10.1038/2221076a0.
58. Bakker, E.P., van den Heuvel, E.J., Wiechmann, A.H.,
and van Dam, K. (1973) A comparison between the
effectiveness of uncouplers of oxidative phosphoryla-
tion in mitochondria and in different artificial mem-
brane systems, Biochim. Biophys. Acta, 292, 78-87,
https://doi.org/10.1016/0005-2728(73)90252-1.
59. Miyoshi, H., Nishioka, T., and Fujita, T. (1987) Quan-
titative relationship between protonophoric and un-
coupling activities of substituted phenols, Biochim.
Biophys. Acta, 891, 194-204, https://doi.org/10.1016/
0005-2728(87)90011-9.
60. Ting, H. P., Wilson, D. F., and Chance, B. (1970) Ef-
fects of uncouplers of oxidative phosphorylation on
the specific conductance of bimolecular lipid mem-
branes, Arch. Biochem. Biophys., 141, 141-146, https://
doi.org/10.1016/0003-9861(70)90116-5.
61. Cunarro, J., and Weiner, M. W. (1975) Mechanism of
action of agents which uncouple oxidative phosphor-
ylation: direct correlation between proton-carrying
and respiratory-releasing properties using rat liver
mitochondria, Biochim. Biophys. Acta, 387, 234-240,
https://doi.org/10.1016/0005-2728(75)90106-1.
62. Parker, V.H. (1965) Uncouplers of rat-liver mitochon-
drial oxidative phosphorylation, Biochem.J., 97, 658-
662, https://doi.org/10.1042/bj0970658.
63. Zuna, K., Jovanovic, O., Khailova, L. S., Skulj, S.,
Brkljaca, Z., Kreiter, J., Kotova, E. A., Vazdar, M.,
Antonenko, Y. N., and Pohl, E. E. (2021) Mitochon-
drial uncoupling proteins (UCP1-UCP3) and adenine
nucleotide translocase (ANT1) enhance the protono-
phoric action of 2,4-dinitrophenol in mitochondria
and planar bilayer membranes, Biomolecules, 11,
1178, https://doi.org/10.3390/biom11081178.
64. Yaguzhinskii, L. S., Ratnikova, L. A., and Kolesova,
G.M. (1973) Quantitative relation between the value
of the dissociation constant of uncoupling agents and
their efficiency in mitochondrial membranes [in Rus-
sian], Biofizika, 18, 460-465.
65. Liberman, E. A., and Topaly, V. P. (1968) Selective
transport of ions through bimolecular phospholipid
membranes, Biochim. Biophys. Acta, 163, 125-136,
https://doi.org/10.1016/0005-2736(68)90089-8.
66. Afanas’eva, E. V., and Kostyrko, V. A. (1986) Penta-
chlorophenol inhibition of succinate oxidation by
the respiratory chain in submitochondrial parti-
cles from the bovine heart [in Russian], Biokhimiia,
51, 823-829.
67. Popova, L.B., Nosikova, E.S., Kotova, E.A., Tarasova,
E. O., Nazarov, P. A., Khailova, L. S., Balezina, O. P.,
ANTONENKO et al.1836
BIOCHEMISTRY (Moscow) Vol. 90 No. 12 2025
and Antonenko, Y. N. (2018) Protonophoric action of
triclosan causes calcium efflux from mitochondria,
plasma membrane depolarization and bursts of min-
iature end-plate potentials, Biochim. Biophys. Acta
Biomembr., 1860, 1000-1007, https://doi.org/10.1016/
j.bbamem.2018.01.008.
68. Horner, W.D. (1941) A study of dinitrophenol and its
relation to cataract formation, Trans. Am. Ophthal-
mol. Soc., 39, 405-437.
69. Miranda, E. J., McIntyre, I. M., Parker, D. R., Gary,
R. D., and Logan, B. K. (2006) Two deaths attributed
to the use of 2,4-dinitrophenol, J. Anal. Toxicol., 30,
219-222, https://doi.org/10.1093/jat/30.3.219.
70. Colman,E. (2007) Dinitrophenol and obesity: an ear-
ly twentieth-century regulatory dilemma, Regul. Tox-
icol. Pharmacol., 48, 115-117, https://doi.org/10.1016/
j.yrtph.2007.03.006.
71. Grundlingh, J., Dargan, P. I., El-Zanfaly, M., and
Wood, D.M. (2011) 2,4-dinitrophenol (DNP): a weight
loss agent with significant acute toxicity and risk
of death, J. Med. Toxicol., 7, 205-212, https://doi.org/
10.1007/s13181-011-0162-6.
72. Khailova, L. S., Vygodina, T. V., Lomakina, G. Y.,
Kotova, E. A., and Antonenko, Y. N. (2020) Bicarbon-
ate suppresses mitochondrial membrane depolariza-
tion induced by conventional uncouplers, Biochem.
Biophys. Res. Commun., 530, 29-34, https://doi.org/
10.1016/j.bbrc.2020.06.131.
73. Wesson, L.G. (1940) Alkyl ethers of 2,4-dinitrophenol
as stimulants of the metabolic rate, J.Am. Chem. Soc.,
62, 3466-3468, https://doi.org/10.1021/ja01869a049.
74. Perry, R.J., Kim,T., Zhang, X.M., Lee, H.Y., Pesta,D.,
Popov, V.B., Zhang,D., Rahimi,Y., Jurczak, M.J., Cline,
G. W., Spiegel, D. A., and Shulman, G. I. (2013) Re-
versal of hypertriglyceridemia, fatty liver disease,
and insulin resistance by a liver-targeted mitochon-
drial uncoupler, Cell Metab., 18, 740-748, https://
doi.org/10.1016/j.cmet.2013.10.004.
75. De Felice, F. G., and Ferreira, S. T. (2006) Novel neu-
roprotective, neuritogenic and anti-amyloidogen-
ic properties of 2,4-dinitrophenol: the gentle face
of Janus, IUBMB Life, 58, 185-191, https://doi.org/
10.1080/15216540600702198.
76. Sebollela,A., Freitas-Correa,L., Oliveira, F.F., Mendes,
C.T., Wasilewska-Sampaio, A. P., Camacho-Pereira, J.,
Galina, A., Brentani, H., Passetti, F., De Felice, F. G.,
Dias-Neto, E., and Ferreira, S. T. (2010) Expression
profile of rat hippocampal neurons treated with the
neuroprotective compound 2,4-dinitrophenol: up-reg-
ulation of cAMP signaling genes, Neurotox. Res., 18,
112-123, https://doi.org/10.1007/s12640-009-9133-y.
77. Holmuhamedov, E. L., Jahangir, A., Oberlin, A.,
Komarov,A., Colombini,M., and Terzic,A. (2004) Potas-
sium channel openers are uncoupling protonophores:
implication in cardioprotection, FEBS Lett., 568,
167-170, https://doi.org/10.1016/j.febslet.2004.05.031.
78. Modriansky, M., and Gabrielova, E. (2009) Uncou-
ple my heart: the benefits of inefficiency, J. Bioen-
erg. Biomembr., 41, 133-136, https://doi.org/10.1007/
s10863-009-9212-z.
79. Noureddin, M., Khan, S., Portell, F., Jorkasky, D.,
Dennis, J., Khanb, O., Johansson, L., Johansson, E.,
and Sanyal, A. J. (2023) Safety and efficacy of once-
daily HU6 versus placebo in people with non-alco-
holic fatty liver disease and high BMI: a randomised,
double-blind, placebo-controlled, phase 2a trial,
Lancet Gastroenterol. Hepatol., 8, 1094-1105, https://
doi.org/10.1016/S2468-1253(23)00198-X.
80. Kitzman, D.W., Lewis, G.D., Pandey,A., Borlaug, B.A.,
Sauer, A. J., Litwin, S. E., Sharma, K., Jorkasky, D. K.,
Khan, S., and Shah, S. J. (2024) A novel controlled
metabolic accelerator for the treatment of obesity-re-
lated heart failure with preserved ejection fraction:
rationale and design of the Phase 2a HuMAIN tri-
al, Eur. J. Heart Fail., 26, 2013-2024, https://doi.org/
10.1002/ejhf.3305.
81. Ng, M. Y., Song, Z. J., Venkatesan, G., Rodriguez-
Cuenca,S., West, J.A., Yang,S., Tan, C.H., Ho, P.C.L.,
Griffin, J.L., Vidal-Puig,A., Bassetto,M., and Hagen,T.
(2024) Conjugating uncoupler compounds with hy-
drophobic hydrocarbon chains to achieve adipose
tissue selective drug accumulation, Sci. Rep., 14,
4932, https://doi.org/10.1038/s41598-024-54466-2.
82. Avlonitis, N., Chalmers, S., McDougall, C., Stanton-
Humphreys, M.N., Brown, C.T., McCarron, J.G., and
Conway, S. J. (2009) Caged AG10: new tools for spa-
tially predefined mitochondrial uncoupling, Mol. Bio-
syst., 5, 450-457, https://doi.org/10.1039/b820415m.
83. Chalmers, S., Caldwell, S. T., Quin, C., Prime, T. A.,
James, A.M., Cairns, A. G., Murphy, M. P., McCarron,
J. G., and Hartley, R. C. (2012) Selective uncoupling
of individual mitochondria within a cell using a mi-
tochondria-targeted photoactivated protonophore,
J.Am. Chem. Soc., 134, 758-761, https://doi.org/10.1021/
ja2077922.
84. Bobba, K. N., Binoy, A., Koo, S., Nedungadi, D.,
Podder, A., Sharma, A., Mishra, N., Kim, J. S., and
Bhuniya, S. (2019) Direct readout protonophore in-
duced selective uncoupling and dysfunction of in-
dividual mitochondria within cancer cells, Chem.
Commun., 55, 6429-6432, https://doi.org/10.1039/
C9CC01483G.
85. Kand, D., Pizarro, L., Angel, I., Avni, A., Friedmann-
Morvinski, D., and Weinstain, R. (2019) Organelle-
targeted BODIPY photocages: visible-light-mediated
subcellular photorelease, Angew. Chem. Int. Ed. Engl.,
58, 4659-4663, https://doi.org/10.1002/anie.201900850.
86. Lopez-Corrales,M., Rovira,A., Gandioso,A., Nonell,S.,
Bosch, M., and Marchan, V. (2023) Mitochondria-
targeted COUPY photocages: synthesis and visi-
ble-light photoactivation in living cells, J.Org. Chem.,
88, 7128-7140, https://doi.org/10.1021/acs.joc.3c00387.
MITOCHONDRIAL UNCOUPLERS 1837
BIOCHEMISTRY (Moscow) Vol. 90 No. 12 2025
87. Firsov, A. M., Khailova, L. S., Nazarov, P. A., Ukrain-
tsev, M., Kirsanov, R. S., Kovalchuk, S. I., Lyamzaev,
K. G., Panteleeva, A. A., Korshunova, G. A., Kotova,
E. A., and Antonenko, Y. N. (2025) 2-Azido-4-nitro-
phenol is a light-sensitive antibacterial protono-
phore, Bioelectrochemistry, 167, 109055, https://
doi.org/10.1016/j.bioelechem.2025.109055.
88. Muraoka, S., and Terada, H. (1972) 3,5-Di-tert-butyl-
4-hydroxybenzylidenemalononitrile, a new powerful
uncoupler of respiratory-chain phosphorylation, Bio-
chim. Biophys. Acta Bioenerg., 275, 271-275, https://
doi.org/10.1016/0005-2728(72)90047-3.
89. Terada, H., and van Dam, K. (1975) On the stoichi-
ometry between uncouplers of oxidative phosphor-
ylation and respiratory chains. The catalytic action
of SF 6847 (3,5-di-tert-butyl-4-hydroxybenzylidene-
malononitrile), Biochim. Biophys. Acta Bioenerg., 387,
507-518, https://doi.org/10.1016/0005-2728(75)90089-4.
90. Terada,H. (1990) Uncouplers of oxidative phosphory-
lation, Environ. Health Perspect., 87, 213-218, https://
doi.org/10.1289/ehp.9087213.
91. Grivennikova, V. G., Khailova, L. S., Zharova, T. V.,
Kotova, E. A., and Antonenko, Y. N. (2022) Inhibi-
tion of respiratory complex I by 6-ketocholestanol:
relevance to recoupling action in mitochondria, Bio-
chim. Biophys. Acta Bioenerg., 1863, 148594, https://
doi.org/10.1016/j.bbabio.2022.148594.
92. Starkov, A. A., Dedukhova, V. I., and Skulachev, V. P.
(1994) 6-ketocholestanol abolishes the effect of the
most potent uncouplers of oxidative phosphorylation
in mitochondria, FEBS Lett., 355, 305-308, https://
doi.org/10.1016/0014-5793(94)01211-3.
93. Starkov, A.A., Bloch, D.A., Chernyak, B.V., Dedukhova,
V.I., Mansurova, S.E., Severina, I.I., Simonyan, R.A.,
Vygodina, T. V., and Skulachev, V. P. (1997) 6-Keto-
cholestanol is a recoupler for mitochondria, chro-
matophores and cytochrome oxidase proteolipo-
somes, Biochim. Biophys. Acta, 1318, 159-172, https://
doi.org/10.1016/S0005-2728(96)00134-X.
94. Franklin, J.C., and Cafiso, D.S. (1993) Internal electro-
static potentials in bilayers - measuring and controlling
dipole potentials in lipid vesicles, Biophys.J., 65, 289-
299, https://doi.org/10.1016/S0006-3495(93)81051-8.
95. Cuellar, A., Ramirez,J., Infante, V. M., and Chavez,E.
(1997) Further studies on the recoupling effect of
6-ketocholestanol upon oxidative phosphorylation in
uncoupled liver mitochondria, FEBS Lett., 411, 365-
368, https://doi.org/10.1016/S0014-5793(97)00741-2.
96. Gazit, A., Yaish, P., Gilon, C., and Levitzki, A. (1989)
Tyrphostins I: synthesis and biological activity of
protein tyrosine kinase inhibitors, J. Med. Chem., 32,
2344-2352, https://doi.org/10.1021/jm00130a020.
97. Levitzki, A., and Gilon, C. (1991) Tyrphostins as
molecular tools and potential antiproliferative
drugs, Trends Pharmacol. Sci., 12, 171-174, https://
doi.org/10.1016/0165-6147(91)90538-4.
98. Needham, D. (2024) Niclosamide: a career build-
er, J.Control Release, 369, 786-856, https://doi.org/
10.1016/j.jconrel.2023.07.016.
99. Jurgeit, A., Mcdowell, R., Moese, S., Meldrum, E.,
Schwendener,R., and Greber, U.F. (2012) Niclosamide
is a proton carrier and targets acidic endosomes with
broad antiviral effects, PLoS Pathog., 8, e1002976,
https://doi.org/10.1371/journal.ppat.1002976.
100. Jung, E., Nam, S., Oh, H., Jun, S., Ro, H. J., Kim, B.,
Kim, M., and Go, Y. Y. (2019) Neutralization of acid-
ic intracellular vesicles by niclosamide inhibits mul-
tiple steps of the dengue virus life cycle in vitro,
Sci. Rep., 9, 8682, https://doi.org/10.1038/s41598-019-
45095-1.
101. Kozhokaru, A. F., Liberman, E. A., Topaly, V. P., and
Topaly, E. E. (1976) Study of the mechanism of ac-
tion of fasciolocides using bimolecular phospholipid
membranes [in Russian], Biofizika, 21, 766-767.
102. Kozhokaru, A.F., G’bev, E. E., Vladimirov, Iu.A., and
Akoev, I. G. (1981) Action of new class of substitut-
ed salicylanilides on ion conductivity of lipid mem-
branes [in Russian], Biofizika, 26, 995-998.
103. Kasianowicz, J., Benz, R., and Mclaughlin, S. (1987)
How do protons cross the membrane solution in-
terface? kinetic studies on bilayer membranes ex-
posed to the protonophore s-13 (5-chloro-3-tert-bu-
tyl-2-chloro-4nitrosalicylanilide), J. Membrane Biol.,
95, 73-89, https://doi.org/10.1007/BF01869632.
104. Ng, M.Y., Song, Z.J., Tan, C.H., Bassetto,M., and Ha-
gen, T. (2024) Structural investigations on the mito-
chondrial uncouplers niclosamide and FCCP, FEBS
Open Bio., 14, 1057-1071, https://doi.org/10.1002/
2211-5463.13817.
105. Terada, H., Goto, S., Yamamoto, K., Takeuchi, I.,
Hamada, Y., and Miyake, K. (1988) Structural re-
quirements of salicylanilides for uncoupling activ-
ity in mitochondria: quantitative analysis of struc-
ture-uncoupling relationships, Biochim. Biophys.
Acta, 936, 504-512, https://doi.org/10.1016/0005-2728
(88)90027-8.
106. Storey, B. T., Wilson, D. F., Bracey, A., Rosen, S. L.,
and Stephenson, S. (1975) Steric and electronic ef-
fects on the uncoupling activity of substituted 3,5 di-
chlorosalicylanilides, FEBS Lett., 49, 338-341, https://
doi.org/10.1016/0014-5793(75)80780-0.
107. Amireddy, N., Puttapaka, S. N., Vinnakota, R. L.,
Ravuri, H. G., Thonda, S., and Kalivendi, S. V. (2017)
The unintended mitochondrial uncoupling effects
of the FDA-approved anti-helminth drug nitazoxa-
nide mitigates experimental parkinsonism in mice,
J.Biol. Chem., 292, 15731-15743, https://doi.org/
10.1074/jbc.M117.791863.
108. Ma, M.-H., Li, F.-F., Li, W.-F., Zhao,H., Jiang,M., Yu, Y.-
Y., Dong, Y.-C., Zhang, Y.-X., Li, P., Bu, W.-J., Sun, Z.-J.,
and Dong, D.-L. (2023) Repurposing nitazoxanide as
a novel anti-atherosclerotic drug based on mitochon-
ANTONENKO et al.1838
BIOCHEMISTRY (Moscow) Vol. 90 No. 12 2025
drial uncoupling mechanisms, Br.J. Pharmacol., 180,
62-79, https://doi.org/10.1111/bph.15949.
109. Li,F., Jiang,M., Ma,M., Chen,X., Zhang,Y., Zhang,Y.,
Yu,Y., Cui,Y., Chen,J., Zhao,H., Sun,Z., and Dong,D.
(2022) Anthelmintics nitazoxanide protects against
experimental hyperlipidemia and hepatic steato-
sis in hamsters and mice, Acta Pharm. Sin. B, 12,
1322-1338, https://doi.org/10.1016/j.apsb.2021.09.009.
110. Kozhokaru, A. F., Ruzieva, R. Kh., Topaly, V. P., and
Topaly, E. E. (1978) Protonophoric and uncoupling
activity of water-soluble pea flavonols [in Russian],
Biofizika, 23, 716-717.
111. De Jonge, P. C., Wieringa, T., Van Putten, J. P., Krans,
H. M., and van Dam, K. (1983) Phloretin – an un-
coupler and an inhibitor of mitochondrial oxi-
dative phosphorylation, Biochim. Biophys. Acta,
722, 219-225, https://doi.org/10.1016/0005-2728(83)
90177-9.
112. Trumbeckaite, S., Bernatoniene, J., Majiene, D.,
Jakstas,V., Savickas,A., and Toleikis,A. (2006) Theef-
fect of flavonoids on rat heart mitochondrial func-
tion, Biomed. Pharmacother., 60, 245-248, https://
doi.org/10.1016/j.biopha.2006.04.003.
113. Sell, T. S., Belkacemi, T., Flockerzi, V., and Beck, A.
(2014) Protonophore properties of hyperforin are es-
sential for its pharmacological activity, Sci. Rep., 4,
7500, https://doi.org/10.1038/srep07500.
114. Andersen, O.S., Finkelstein, A., Katz, I., and Cass, A.
(1976) Effect of phloretin on the permeability of thin
lipid membranes, J.Gen. Physiol., 67, 749-771, https://
doi.org/10.1085/jgp.67.6.749.
115. Pohl, P., Rokitskaya, T. I., Pohl, E. E., and Saparov,
S. M. (1997) Permeation of phloretin across bilayer
lipid membranes monitored by dipole potential and
microelectrode measurements, Biochim. Biophys.
Acta, 1323, 163-172, https://doi.org/10.1016/S0005-
2736(96)00185-X.
116. Rokitskaya, T. I., Ilyasova, T. M., Severina, I. I.,
Antonenko, Y. N., and Skulachev, V. P. (2013) Elec-
trogenic proton transport across lipid bilayer mem-
branes mediated by cationic derivatives of rhodamine
19: comparison with anionic protonophores, Eur. Bio-
phys. J., 42, 477-485, https://doi.org/10.1007/s00249-
013-0898-9.
117. Schmitt, L., Krings, K. S., Wolsing, A., Buque, X.,
Zimmermann, M., Flores-Romero, H., Lenz, T.,
Lechtenberg, I., Peter, C., Stork, B., Teusch, N.,
Proksch, P., Stuehler, K., Garcia-Saez, A. J., Reichert,
A. S., Aspichueta, P., Bhatia, S., and Wesselborg, S.
(2024) Targeting mitochondrial metabolism by the
mitotoxin bromoxib in leukemia and lymphoma
cells, Cell Commun. Signal., 22, 541, https://doi.org/
10.1186/s12964-024-01913-2.
118. Shchepinova, M. M., Denisov, S. S., Kotova, E. A.,
Khailova, L. S., Knorre, D. A., Korshunova, G. A.,
Tashlitsky, V. N., Severin, F. F., and Antonenko, Y. N.
(2014) Dodecyl and octyl esters of fluorescein as pro-
tonophores and uncouplers of oxidative phosphory-
lation in mitochondria at submicromolar concentra-
tions, Biochim. Biophys. Acta Bioenerg., 1837, 149-158,
https://doi.org/10.1016/j.bbabio.2013.09.011.
119. Nazarov, P.A., Kirsanov, R.S., Denisov, S.S., Khailova,
L.S., Karakozova, M.V., Lyamzaev, K.G., Korshunova,
G.A., Lukyanov, K.A., Kotova, E.A., and Antonenko,
Y. N. (2020) Fluorescein derivatives as antibacterial
agents acting via membrane depolarization, Biomole-
cules, 10, 309, https://doi.org/10.3390/biom10020309.
120. Venugopala, K. N., Rashmi, V., and Odhav, B. (2013)
Review on natural coumarin lead compounds for
their pharmacological activity, Biomed. Res. Int.,
2013, 963248, https://doi.org/10.1155/2013/963248.
121. Dadak,V., Zboril,P., Sanek,J., and Svoboda,A. (1980)
Mode of effect of ostruthin, a phenolic coumarin on
respiration and oxidative phosphorylation of rat liver
mitochondria, Collect. Czechoslovak Chem. Commun.,
45, 653-666, https://doi.org/10.1135/cccc19800653.
122. Du, L., Mahdi, F., Jekabsons, M. B., Nagle, D. G., and
Zhou, Y. D. (2010) Mammea E/BB, an isoprenylated
dihydroxycoumarin protonophore that potently un-
couples mitochondrial electron transport, disrupts
hypoxic signaling in tumor cells, J. Nat. Prod., 73,
1868-1872, https://doi.org/10.1021/np100501n.
123. Nowak, P. M., Sagan, F., and Mitoraj, M. P. (2017)
Origin of remarkably different acidity of hydroxy-
coumarins – joint experimental and theoretical
studies, J. Phys. Chem. B, 121, 4554-4561, https://
doi.org/10.1021/acs.jpcb.7b01849.
124. Krasnov, V. S., Kirsanov, R.S., Khailova, L. S., Firsov,
A. M., Nazarov, P. A., Tashlitsky, V. N., Korshunova,
G.A., Kotova, E.A., and Antonenko, Y.N. (2022) Alkyl
esters of umbelliferone-4-acetic acid as protonophores
in bilayer lipid membranes and ALDH2-dependent
soft uncouplers in rat liver mitochondria, Bioelec-
trochemistry, 145, 108081, https://doi.org/10.1016/
j.bioelechem.2022.108081.
125. Krasnov, V.S., Kirsanov, R.S., Khailova, L.S., Popova,
L. B., Lyamzaev, K. G., Firsov, A. M., Korshunova,
G. A., Kotova, E. A., and Antonenko, Y. N. (2022) Al-
kyl esters of 7-hydroxycoumarin-3-carboxylic acid as
potent tissue-specific uncouplers of oxidative phos-
phorylation: involvement of ATP/ADP translocase
in mitochondrial uncoupling, Arch. Biochem. Bio-
phys., 728, 109366, https://doi.org/10.1016/j.abb.2022.
109366.
126. Heytler, P. G. (1979) Uncouplers of oxidative phos-
phorylation, Methods Enzymol., 55, 462-472, https://
doi.org/10.1016/0076-6879(79)55060-5.
127. Rokitskaya, T.I., Kotova, E.A., Luzhkov, V.B., Kirsanov,
R.S., Aleksandrova, E.V., Korshunova, G.A., Tashlitsky,
V.N., and Antonenko, Y.N. (2021) Lipophilic ion aro-
maticity is not important for permeability across
lipid membranes, Biochim. Biophys. Acta Biomem-
MITOCHONDRIAL UNCOUPLERS 1839
BIOCHEMISTRY (Moscow) Vol. 90 No. 12 2025
br., 1863, 183483, https://doi.org/10.1016/j.bbamem.
2020.183483.
128. Rupprecht,A., Sokolenko, E.A., Beck,V., Ninnemann,O.,
Jaburek, M., Trimbuch, T., Klishin, S. S., Jezek, P.,
Skulachev, V. P., and Pohl, E. E. (2010) Role of the
transmembrane potential in the membrane pro-
ton leak, Biophys. J., 98, 1503-1511, https://doi.org/
10.1016/j.bpj.2009.12.4301.
129. Montal, M., and Mueller, P. (1972) Formation of bi-
molecular membranes from lipid monolayers and
a study of their electrical properties, Proc. Natl.
Acad. Sci. USA, 69, 3561-3566, https://doi.org/10.1073/
pnas.69.12.3561.
130. Andreyev, A. Y., Bondareva, T. O., Dedukhova, V. I.,
Mokhova, E. N., Skulachev, V. P., and Volkov, N. I.
(1988) Carboxyatractylate inhibits the uncoupling
effect of free fatty acids, FEBS Lett., 226, 265-269,
https://doi.org/10.1016/0014-5793(88)81436-4.
131. Urbankova,E., Voltchenko, A., Pohl, P., Jezek, P., and
Pohl, E. E. (2003) Transport kinetics of uncoupling
proteins. Analysis of UCP1 reconstituted in planar
lipid bilayers, J.Biol. Chem., 278, 32497-32500, https://
doi.org/10.1074/jbc.M303721200.
132. Beck, V., Jaburek, M., Demina, T., Rupprecht, A.,
Porter, R. K., Jezek, P., and Pohl, E. E. (2007) Poly-
unsaturated fatty acids activate human uncou-
pling proteins 1 and 2 in planar lipid bilayers,
FASEB J., 21, 1137-1144, https://doi.org/10.1096/
fj.06-7489com.
133. Fedorenko, A., Lishko, P. V., and Kirichok, Y. (2012)
Mechanism of fatty-acid-dependent UCP1 uncoupling
in brown fat mitochondria, Cell, 151, 400-413, https://
doi.org/10.1016/j.cell.2012.09.010.
134. Bertholet, A.M., Chouchani, E.T., Kazak,L., Angelin,A.,
Fedorenko, A., Long, J. Z., Vidoni, S., Garrity, R.,
Cho, J., Terada, N., Wallace, D. C., Spiegelman, B. M.,
and Kirichok, Y. (2019) H
+
transport is an integral
function of the mitochondrial ADP/ATP carrier,
Nature, 571, 515-520, https://doi.org/10.1038/s41586-
019-1400-3.
135. Kreiter, J., Rupprecht, A., Skulj, S., Brkljaca, Z.,
Žuna,K., Knyazev, D.G., Bardakji,S., Vazdar,M., and
Pohl, E.E. (2021) ANT1 Activation and inhibition pat-
terns support the fatty acid cycling mechanism for
proton transport, Int.J. Mol. Sci., 22, 2490, https://
doi.org/10.3390/ijms22052490.
136. Kreiter,J., Skulj,S., Brkljaca,Z., Bardakji,S., Vazdar,M.,
and Pohl, E. E. (2023) FA sliding as the mechanism
for the ANT1-mediated fatty acid anion transport
in lipid bilayers, Int.J. Mol. Sci., 24, 13701, https://
doi.org/10.3390/ijms241813701.
137. Pohl, E. E., Vazdar, M., and Kreiter, J. (2025) Explor-
ing the proton transport mechanism of the mito-
chondrial ADP/ATP carrier: FA-cycling hypothesis
and beyond, Prot. Sci., 34, e70047, https://doi.org/
10.1002/pro.70047.
138. Semenova, A.A., Samartsev, V.N., and Dubinin, M.V.
(2021) The stimulation of succinate-fueled respira-
tion of rat liver mitochondria in state 4 by α,ω-hexa-
decanedioic acid without induction of proton con-
ductivity of the inner membrane. Intrinsic uncou-
pling of the bc1 complex, Biochimie, 181, 215-225,
https://doi.org/10.1016/j.biochi.2020.12.021.
139. Evtodienko, V. Y., Bondarenko, D. I., and Antonenko,
Y. N. (1999) Permeation of dicarboxylic acids with
different terminal position of two carboxylic groups
through planar bilayer lipid membranes, Biochim.
Biophys. Acta, 1420, 95-103, https://doi.org/10.1016/
S0005-2736(99)00095-4.
140. Radde, B. N., Alizadeh-Rad, N., Price, S. M., Schultz,
D. J., and Klinge, C. M. (2016) Anacardic acid, sal-
icylic acid, and oleic acid differentially alter cel-
lular bioenergetic function in breast cancer cells,
J. Cell Biochem., 117, 2521-2532, https://doi.org/
10.1002/jcb.25544.
141. Acevedo, A., Jones, A. E., Danna, B. T., Turner, R.,
Montales, K. P., Beninca, C., Reue, K., Shirihai, O. S.,
Stiles,L., Wallace, M., Wang, Y., Bertholet, A. M., and
Divakaruni, A. S. (2024) The BCKDK inhibitor BT2
is a chemical uncoupler that lowers mitochondrial
ROS production and de novo lipogenesis, J. Biol.
Chem., 300, 105702, https://doi.org/10.1016/j.jbc.
2024.105702.
142. Zhang, Y., Tan, J., and Chen, Y. (2023) Visible-light-
induced protein labeling in live cells with aryl
azides, Chem. Commun., 59, 2413-2420, https://doi.org/
10.1039/D2CC06987C.
143. Hanstein, W. G., and Hatefi, Y. (1974) Characteriza-
tion and localization of mitochondrial uncoupler
binding sites with an uncoupler capable of photoaf-
finity labeling, J.Biol. Chem., 249, 1356-1362, https://
doi.org/10.1016/S0021-9258(19)42889-5.
144. Hatefi,Y. (1975) Energy conservation and uncoupling
in mitochondria, J.Supramol. Struct., 3, 201-213,
https://doi.org/10.1002/jss.400030302.
145. Schonfeld, P., Jezek, P., Belyaeva, E. A., Borecky, J.,
Slyshenkov, V.S., Wieckowski, M.R., and Wojtczak,L.
(1996) Photomodification of mitochondrial proteins
by azido fatty acids and its effect on mitochondri-
al energetics. Further evidence for the role of the
ADP/ATP carrier in fatty-acid-mediated uncoupling,
Eur. J. Biochem., 240, 387-393, https://doi.org/10.1111/
j.1432-1033.1996.0387h.x.
146. Valderrama,K., Pradel,E., Firsov, A.M., Drobecq,H.,
Bauderlique-le Roy, H., Villemagne, B., Antonenko,
Y. N., and Hartkoorn, R. C. (2019) Pyrrolomycins are
potent natural protonophores, Antimicrob. Agents
Chemother., 63, e01450-19, https://doi.org/10.1128/
AAC.01450-19.
147. Firsov, A.M., Khailova, L.S., Rokitskaya, T.I., Kotova,
E. A., and Antonenko, Y. N. (2022) Antibiotic pyrro-
lomycin as an efficient mitochondrial uncoupler,
ANTONENKO et al.1840
BIOCHEMISTRY (Moscow) Vol. 90 No. 12 2025
Biochemistry (Moscow), 87, 812-822, https://doi.org/
10.1134/S0006297922080120.
148. Treacy, M., Miller, T., Black, B., Gard, I., Hunt, D.,
and Hollingworth, R. M. (1994) Uncoupling activity
and pesticidal properties of pyrroles, Biochem. Soc.
Trans., 22, 244-247, https://doi.org/10.1042/bst0220244.
149. Heytler, P.G., and Prichard, W.W. (1962) Anew class
of uncoupling agents – carbonyl cyanide phenylhy-
drazones, Biochem. Biophys. Res. Commun., 7, 272-
275, https://doi.org/10.1016/0006-291X(62)90189-4.
150. Heytler, P. G. (1963) Uncoupling of oxidative phos-
phorylation by carbonyl cyanide phenylhydrazones.
I. Some characteristics of m-Cl-CCP action on mito-
chondria and chloroplasts, Biochemistry, 2, 357-361,
https://doi.org/10.1021/bi00902a031.
151. Blum, J.J., and Sanadi, D.R. (1964) Activation of my-
osin adenosine triphosphatase by carbonyl cyanide
p-chlorophenylhydrazone, J.Biol. Chem., 239, 452-454,
https://doi.org/10.1016/S0021-9258(18)51700-2.
152. Drobnica, L., and Sturdik, E. (1979) The reaction of
carbonyl cyanide phenylhydrazones with thiols, Bio-
chim. Biophys. Acta, 585, 462-476, https://doi.org/
10.1016/0304-4165(79)90091-6.
153. Toninello, A., and Siliprandi, N. (1982) Restoration
of membrane potential in mitochondria deener-
gized with carbonyl cyanide p-trifluoromethoxy-
phenylhydrazone (FCCP), Biochim. Biophys. Acta,
682, 289-292, https://doi.org/10.1016/0005-2728(82)
90110-4.
154. Mlejnek, P., and Dolezel, P. (2015) Loss of mitochon-
drial transmembrane potential and glutathione de-
pletion are not sufficient to account for induction of
apoptosis by carbonyl cyanide 4-(trifluoromethoxy)
phenylhydrazone in human leukemia K562 cells,
Chem. Biol. Interact., 239, 110, https://doi.org/10.1016/
j.cbi.2015.06.033.
155. Khailova, L. S., Firsov, A. M., Kotova, E. A., and
Antonenko, Y. N. (2019) Interaction of potent mito-
chondrial uncouplers with thiol-containing antiox-
idants, Antioxidants, 8, 194, https://doi.org/10.3390/
antiox8060194.
156. Doczi, J., Karnok, N., Bui, D., Azarov, V., Pallag, G.,
Nazarian, S., Czumbel, B., Seyfried, T. N., and
Chinopoulos, C. (2023) Viability of MCF cells is not
correlated with mitochondrial bioenergetics, Sci.
Rep., 13, 10822, https://doi.org/10.1038/s41598-023-
37677-x.
157. Demine, S., Renard, P., and Arnould, T. (2019) Mito-
chondrial uncoupling: a key controller of biological
processes in physiology and diseases, Cells, 8, E795,
https://doi.org/10.3390/cells8080795.
158. LeBlanc, O. H. (1971) The effect of uncouplers of
oxidative phosphorylation on lipid bilayer mem-
branes: carbonylcyanide m-chloropenylhydrazone,
J. Membr. Biol., 4, 227-251, https://doi.org/10.1007/
BF02431973.
159. O’Shaughnessy, K., and Hladky, S. B. (1983) Tran-
sient currents carried by the uncoupler, carbonyl
cyanide m-chlorophenylhydrazone, Biochim. Bio-
phys. Acta, 724, 381-387, https://doi.org/10.1016/
0005-2728(83)90097-X.
160. Benz, R., and McLaughlin, S. (1983) The molecu-
lar mechanism of action of the proton ionophore
FCCP (carbonylcyanide p-trifluoromethoxyphenyl-
hydrazone), Biophys. J., 41, 381-398, https://doi.org/
10.1016/S0006-3495(83)84449-X.
161. Cho, I., Song, H.-O., and Cho, J. H. (2017) Mitochon-
drial uncoupling attenuates age-dependent neuro-
degeneration in C. elegans, Mol. Cells, 40, 864-870,
https://doi.org/10.14348/molcells.2017.0172.
162. Katre, N. V., and Wilson, D. F. (1977) Interaction of
uncouplers with the mitochondrial membrane: a
high-affinity binding site, Arch. Biochem. Biophys.,
184, 578-585, https://doi.org/10.1016/0003-9861(77)
90468-4.
163. Brennan, J.P., Southworth,R., Medina, R.A., Davidson,
S. M., Duchen, M. R., and Shattock, M. J. (2006) Mi-
tochondrial uncoupling, with low concentration
FCCP, induces ROS-dependent cardioprotection in-
dependent of KATP channel activation, Cardiovasc.
Res., 72, 313-321, https://doi.org/10.1016/j.cardiores.
2006.07.019.
164. Chaudhari, A.A., Seol, J.W., Kim, S.J., Lee, Y.J., Kang,
H.S., Kim, I. S., Kim, N. S., and Park, S.Y. (2007) Re-
active oxygen species regulate Bax translocation and
mitochondrial transmembrane potential, a possible
mechanism for enhanced TRAIL-induced apoptosis
by CCCP, Oncol. Rep., 18, 71-76, https://doi.org/10.3892/
or.18.1.71.
165. Han, Y.H., and Park, W.H. (2011) Intracellular gluta-
thione levels are involved in carbonyl cyanide p-(tri-
fluoromethoxy) phenylhydrazone-induced apoptosis
in As4. 1 juxtaglomerular cells, Int. J. Mol. Med., 27,
575-581, https://doi.org/10.3892/ijmm.2011.604.
166. Lee, J.E., Kang, J.S., Shin, I.C., Lee, S.J., Hyun, D.H.,
Lee, K. S., and Koh, H. C. (2011) Fluazinam-induced
apoptosis of SH-SY5Y cells is mediated by p53 and
Bcl-2 family proteins, NeuroToxicol., 32, 702-710,
https://doi.org/10.1016/j.neuro.2011.08.004.
167. Jing, K., Shin, S., Jeong, S., Kim, S., Song, K.-S., Park,
J.-H., Heo, J.-Y., Seo, K.-S., Park, S.-K., Kweon, G.-R.,
Wu,T., Park, J.-I., and Lim,K. (2014) Docosahexaeno-
ic acid induces the degradation of HPV E6/E7 oncop-
roteins by activating the ubiquitin–proteasome sys-
tem, Cell Death Dis., 5, e1524, https://doi.org/10.1038/
cddis.2014.477.
168. Zhang, X., Cheng, X., Yu, L., Yang, J., Calvo, R.,
Patnaik, S., Hu, X., Gao, Q., Yang, M., Lawas, M.,
Delling,M., Marugan,J., Ferrer,M., and Xu,H. (2016)
MCOLN1 is a ROS sensor in lysosomes that regulates
autophagy, Nat. Commun., 7, 12109, https://doi.org/
10.1038/ncomms12109.
MITOCHONDRIAL UNCOUPLERS 1841
BIOCHEMISTRY (Moscow) Vol. 90 No. 12 2025
169. Xiao, B., Goh, J.-Y., Xiao, L., Xian, H., Lim, K.-L., and
Liou, Y.-C. (2017) Reactive oxygen species trigger
Parkin/PINK1 pathway-dependent mitophagy by in-
ducing mitochondrial recruitment of Parkin, J. Biol.
Chem., 292, 16697-16708, https://doi.org/10.1074/
jbc.M117.787739.
170. Kenwood, B. M., Weaver, J. L., Bajwa, A., Poon, I. K.,
Byrne, F.L., Murrow, B.A., Calderone, J.A., Huang,L.,
Divakaruni, A. S., Tomsig, J. L., Okabe, K., Lo, R. H.,
Coleman, G. C., Columbus, L., Yan, Z., Saucerman,
J.J., Smith, J.S., Holmes, J.W., Lynch, K.R., Ravichan-
dran, K.S., Uchiyama,S., Santos, W.L., Rogers, G.W.,
Okusa, M. D., Bayliss, D. A., and Hoehn, K. L. (2013)
Identification of a novel mitochondrial uncoupler
that does not depolarize the plasma membrane, Mol.
Metab., 3, 114-123, https://doi.org/10.1016/j.molmet.
2013.11.005.
171. Childress, E. S., Salamoun, J. M., Hargett, S. R.,
Alexopoulos, S. J., Chen, S.-Y., Shah, D. P., Santiago-
Rivera, J., Garcia, C. J., Dai, Y., Tucker, S. P., Hoehn,
K.L., and Santos, W.L. (2020) [1,2,5]Oxadiazolo[3,4-b]
pyrazine-5,6-diamine derivatives as mitochondrial
uncouplers for the potential treatment of nonalco-
holic steatohepatitis, J. Med. Chem., 63, 2511-2526,
https://doi.org/10.1021/acs.jmedchem.9b01440.
172. Alexopoulos, S. J., Chen, S.-Y., Brandon, A. E.,
Salamoun, J.M., Byrne, F.L., Garcia, C.J., Beretta,M.,
Olzomer, E.M., Shah, D.P., Philp, A.M., Hargett, S.R.,
Lawrence, R. T., Lee, B., Sligar, J., Carrive, P., Tucker,
S. P., Philp, A., Lackner, C., Turner, N., Cooney, G. J.,
Santos, W. L., and Hoehn, K. L. (2020) Mitochondri-
al uncoupler BAM15 reverses diet-induced obesity
and insulin resistance in mice, Nat. Commun., 11,
2397, https://doi.org/10.1038/s41467-020-16298-2.
173. Khailova, L. S., Krasnov, V. S., Kirsanov, R. S., Pop-
ova, L. B., Tashlitsky, V. N., Kotova, E. A., and An-
tonenko, Y. N. (2023) The transient character of mi-
tochondrial uncoupling by the popular fungicide
fluazinam is specific for liver, Arch. Biochem. Bio-
phys., 746, 109735, https://doi.org/10.1016/j.abb.2023.
109735.
174. Guo,Z., Miyoshi,H., Komyoji,T., Haga,T., and Fujita,T.
(1991) Uncoupling activity of a newly developed
fungicide, fluazinam [3-chloro-N-(3-chloro-2,6-dini-
tro-4-trifluoromethylphenyl)-5-trifluoromethyl-2-pyr-
idinamine], Biochim. Biophys. Acta, 1056, 89-92,
https://doi.org/10.1016/S0005-2728(05)80077-5.
175. Brandt, U., Schubert, J., Geck, P., and von Jagow, G.
(1992) Uncoupling activity and physicochemical
properties of derivatives of fluazinam, Biochim.
Biophys. Acta, 1101, 41-47, https://doi.org/10.1016/
0167-4838(92)90464-O.
176. Kirsanov, R. S., Khailova, L. S., Krasnov, V. S., Firsov,
A.M., Lyamzaev, K.G., Panteleeva, A.A., Popova, L.B.,
Nazarov, P. A., Tashlitsky, V. N., Korshunova, G. A.,
Kotova, E.A., and Antonenko, Y.N. (2024) Spontaneous
reversal of small molecule-induced mitochondrial
uncoupling: the case of anilinothiophenes, FEBS J.,
291, 5523-5539, https://doi.org/10.1111/febs.17329.
177. Schaefer, G., and Buechel, K. H. (1970) Cyclic re-
sponse of energy linked functions of liver mito-
chondria to uncoupling thiophene derivatives, FEBS
Lett., 6, 217-220, https://doi.org/10.1016/0014-5793(70)
80061-8.
178. Dejonghe, W., Kuenen, S., Mylle, E., Vasileva, M.,
Keech, O., Viotti, C., Swerts, J., Fendrych, M., Ortiz-
Morea, F.A., Mishev,K., Delang,S., Scholl,S., Zarza,X.,
Heilmann, M., Kourelis, J., Kasprowicz, J., Nguyen,
L. S. L., Drozdzecki, A., Van Houtte, I., Szatmari,
A.-M., Majda,M., Baisa,G., Bednarek, S.Y., Robert,S.,
Audenaert, D., Testerink, C., Munnik, T., Van
Damme, D., Schumacher, K., Winne, J., Friml, J.,
Verstreken, P., and Russinova, E. (2016) Mitochon-
drial uncouplers inhibit clathrin-mediated endo-
cytosis largely through cytoplasmic acidification,
Nat. Commun., 7, 11710, https://doi.org/10.1038/
ncomms11710.
179. Cardoso, R.M.S., Filipe, H.A.L., Gomes,F., Moreira,
N. D., Vaz, W. L. C., and Moreno, M. J. (2010) Chain
length effect on the binding of amphiphiles to se-
rum albumin and to POPC bilayers, J.Phys. Chem. B,
114, 16337-16346, https://doi.org/10.1021/jp105163k.
180. Denisov, S. S., Kotova, E. A., Khailova, L. S.,
Korshunova, G. A., and Antonenko, Y. N. (2014)
Tuning the hydrophobicity overcomes unfavorable
deprotonation making octylamino-substituted 7-ni-
trobenz-2-oxa-1,3-diazole (n-octylamino-NBD) a pro-
tonophore and uncoupler of oxidative phosphoryla-
tion in mitochondria, Bioelectrochemistry, 98, 30-38,
https://doi.org/10.1016/j.bioelechem.2014.02.002.
181. Antonenko, Y. N., Denisov, S. S., Khailova, L. S.,
Nazarov, P. A., Rokitskaya, T. I., Tashlitsky, V. N.,
Firsov, A. M., Korshunova, G. A., and Kotova, E. A.
(2017) Alkyl-substituted phenylamino derivatives of
7-nitrobenz-2-oxa-1,3-diazole as uncouplers of ox-
idative phosphorylation and antibacterial agents:
involvement of membrane proteins in the uncou-
pling action, Biochim. Biophys. Acta Biomembr.,
1859, 377-387, https://doi.org/10.1016/j.bbamem.2016.
12.014.
182. Rebhahn, V. I. C., Saoud, M., Winterhalter, M.,
Schanbacher, F., Jobst, M., Rulz, R., Sonntag, A.,
Kollatz,J., Sprengel,R., Donovan, S.F., Del Favero,G.,
Rennert, R., and Niedermeyer, T. H. J. (2025) Aetok-
thonotoxin, the causative agent of vacuolar my-
elinopathy, uncouples oxidative phosphorylation
due to protonophore activity, Chem. Res. Toxicol.,
38, 1495-1508, https://doi.org/10.1021/acs.chemrestox.
5c00147.
183. Figarola, J. L., Singhal, J., Tompkins, J. D., Rogers,
G. W., Warden, C., Horne, D., Riggs, A. D., Awas-
thi, S., and Singhal, S. S. (2015) SR4 uncouples mito-
ANTONENKO et al.1842
BIOCHEMISTRY (Moscow) Vol. 90 No. 12 2025
chondrial oxidative phosphorylation, modulates
AMP- dependent kinase (AMPK)-mammalian target of
rapamycin (mTOR) signaling, and inhibits prolifera-
tion of HepG2 hepatocarcinoma cells, J.Biol. Chem.,
290, 30321-30341, https://doi.org/10.1074/jbc.M115.
686352.
184. Wu,X., Judd, L.W., Howe, E.N.W., Withecombe, A.M.,
Soto-Cerrato, V., Li, H., Busschaert, N., Valkenier, H.,
Perez-Tomas, R., Sheppard, D. N., Jiang, Y.-B., Davis,
A. P., and Gale, P. A. (2016) Nonprotonophoric elec-
trogenic Cl transport mediated by valinomycin-
like carriers, Chem., 1, 127-146, https://doi.org/
10.1016/j.chempr.2016.04.002.
185. Routaboul, J.-M., Taillandier, G., Ravanel, P., and
Tissut, M. (1994) Potent non-protonophore uncou-
plers acting on natural and artificial membranes,
Phytochemistry, 35, 553-559, https://doi.org/10.1016/
S0031-9422(00)90560-0.
186. York,E., McNaughton, D.A., Roseblade,A., Cranfield,
C. G., Gale, P. A., and Rawling, T. (2022) Structure-
activity relationship and mechanistic studies of bis-
aryl urea anticancer agents indicate mitochondrial
uncoupling by a fatty acid-activated mechanism, ACS
Chem. Biol., 17, 2065-2073, https://doi.org/10.1021/
acschembio.1c00807.
187. Wilhelm, S.M., Adnane,L., Newell,P., Villanueva,A.,
Llovet, J. M., and Lynch, M. (2008) Preclinical over-
view of sorafenib, a multikinase inhibitor that tar-
gets both Raf and VEGF and PDGF receptor tyrosine
kinase signaling, Mol. Cancer Ther., 7, 3129-3140,
https://doi.org/10.1158/1535-7163.MCT-08-0013.
188. Jian,C., Fu,J., Cheng,X., Shen, L.-J., Ji, Y.-X., Wang,X.,
Pan, S., Tian, H., Tian, S., Liao, R., Song, K., Wang,
H.-P., Zhang, X., Wang, Y., Huang, Z., She, Z.-G.,
Zhang, X.-J., Zhu, L., and Hongliang, L. (2020) Low-
dose sorafenib acts as a mitochondrial uncoupler
and ameliorates nonalcoholic steatohepatitis, Cell
Metab., 31, 892-908, https://doi.org/10.1016/j.cmet.
2020.04.011.
189. Ek,F., Blom,K., Selvin,T., Rudfeldt, J., Andersson,C.,
Senkowski, W., Brechot, C., Nygren, P., Larsson, R.,
Jarvius, M., and Fryknaes, M. (2022) Sorafenib and
nitazoxanide disrupt mitochondrial function and in-
hibit regrowth capacity in three-dimensional mod-
els of hepatocellular and colorectal carcinoma, Sci.
Rep., 12, 8943, https://doi.org/10.1038/s41598-022-
12519-4.
190. Fox, M. A., and Hughes, A. K. (2004) Cage C–H⋯X
interactions in solid-state structures of icosahedral
carboranes, Coord. Chem. Rev., 248, 457-476, https://
doi.org/10.1016/j.ccr.2003.10.002.
191. Liberman, E. A., Topaly, V. P., and Silberstein, A. Y.
(1970) Charged and neutral ion carriers through
bimolecular phospholipid membranes, Biochim.
Biophys. Acta, 196, 221-234, https://doi.org/10.1016/
0005-2736(70)90010-6.
192. Rokitskaya, T. I., Khailova, L. S., Makarenkov, A. V.,
Ol’shevskaya, V. A., Kalinin, V. N., and Antonenko,
Y. N. (2016) Weak C–H acids as protonophores can
carry hydrogen ions through lipid membranes and
mitochondria: a case of o-carborane, Phys. Chem.
Chem. Phys., 18, 16476-16482, https://doi.org/10.1039/
C6CP02581A.
193. Bregadze, V. I., Sivaev, I. B., and Glazun, S. A.
(2006) Polyhedral boron compounds as poten-
tial diagnostic and therapeutic antitumor agents,
Anticancer Agents Med. Chem., 6, 75-109, https://
doi.org/10.2174/187152006776119180.
194. Rokitskaya, T. I., Khailova, L. S., Makarenkov, A. V.,
Shunaev, A. V., Tatarskiy, V. V., Shtil, A. A., Ol'shev-
skaya, V. A., and Antonenko, Y. N. (2019) Carborane
derivatives of 1,2,3-triazole depolarize mitochon-
dria by transferring protons through the lipid part
of membranes, Biochim. Biophys. Acta Biomem-
br., 1861, 573-583, https://doi.org/10.1016/j.bbamem.
2018.12.009.
195. Smejtek, P., Jayaweera, A. R., and Hsu, K. (1983)
Electrical conductivity, transfer of hydrogen ions
in lipid bilayer membranes and uncoupling effect
induced by pentachlorobenzenethiol (pentachlo-
rothiophenol), J. Membr. Biol., 76, 227-234, https://
doi.org/10.1007/BF01870365.
196. Terada, H., Uda, M., Kametani, F., and Kubota, S.
(1978) Structural requirements of alkyl acyldithio-
carbazates for the uncoupling of oxidative phos-
phorylation in mitochondria, Biochim. Biophys. Acta,
504, 237-247, https://doi.org/10.1016/0005-2728(78)
90172-X.
197. Schwaller, M.A., Allard,B., Lescot,E., and Moreau,F.
(1995) Protonophoric activity of ellipticine and iso-
mers across the energy-transducing membrane of
mitochondria, J.Biol. Chem., 270, 22709-22713, https://
doi.org/10.1074/jbc.270.39.22709.
198. Auclair, C. (1987) Multimodal action of antitumor
agents on DNA: the ellipticine series, Arch. Bio-
chem. Biophys., 259, 1-14, https://doi.org/10.1016/
0003-9861(87)90463-2.
199. Khailova, L. S., Silachev, D. N., Rokitskaya, T. I.,
Avetisyan, A. V., Lyamzaev, K. G., Severina, I. I.,
Il’yasova, T. M., Gulyaev, M. V., Dedukhova, V. I.,
Trendeleva, T.A., Plotnikov, E.Y., Zvyagilskaya, R.A.,
Chernyak, B. V., Zorov, D. B., Antonenko, Y. N., and
Skulachev, V. P. (2014) A short-chain alkyl deriva-
tive of Rhodamine 19 acts as a mild uncoupler of
mitochondria and a neuroprotector, Biochim. Bio-
phys. Acta, 1837, 1739-1747, https://doi.org/10.1016/
j.bbabio.2014.07.006.
200. Zorova, L. D., Pevzner, I. B., Khailova, L. S.,
Korshunova, G. A., Kovaleva, M. A., Kovalev, L. I.,
Serebryakova, M. V., Silachev, D. N., Sudakov, R. V.,
Zorov, S. D., Rokitskaya, T. I., Popkov, V. A., Plotnikov,
E. Y., Antonenko, Y. N., and Zorov, D. B. (2023) Mito-
MITOCHONDRIAL UNCOUPLERS 1843
BIOCHEMISTRY (Moscow) Vol. 90 No. 12 2025
chondrial ATP synthase and mild uncoupling by butyl
ester of rhodamine 19, C4R1, Antioxidants, 12, 646,
https://doi.org/10.3390/antiox12030646.
201. Mai, M. S., and Allison, W. S. (1983) Inhibition of an
oligomycin-sensitive ATPase by cationic dyes, some
of which are atypical uncouplers of intact mitochon-
dria, Arch. Biochem. Biophys., 221, 467-476, https://
doi.org/10.1016/0003-9861(83)90165-0.
202. Schwaller, M.A., Allard,B., Sureau,F., and Moreau,F.
(1994) Protonation equilibrium of ellipticine bound
to the energy-transducing membrane of mitochon-
dria, J.Phys. Chem., 98, 4209-4211, https://doi.org/
10.1021/j100067a002.
203. Sureau, F., Moreau, F., Millot, J. M., Manfait, M.,
Allard, B., Aubard, J., and Schwaller, M. A. (1993)
Microspectrofluorometry of the protonation state
of ellipticine, an antitumor alkaloid, in single cells,
Biophys. J., 65, 1767-1774, https://doi.org/10.1016/
S0006-3495(93)81273-6.
204. Gear, A. R. (1974) Rhodamine 6G. A potent inhib-
itor of mitochondrial oxidative phosphorylation,
J.Biol. Chem., 249, 3628-3637, https://doi.org/10.1016/
S0021-9258(19)42620-3.
205. Duvvuri, M., Gong, Y., Chatterji, D., and Krise, J. P.
(2004) Weak base permeability characteristics influ-
ence the intracellular sequestration site in the multi-
drug-resistant human leukemic cell line HL-60, J.Biol.
Chem., 279, 32367-32372, https://doi.org/10.1074/
jbc.M400735200.
206. Milanova, D., Chambers, R. D., Bahga, S. S., and
Santiago, J. G. (2011) Electrophoretic mobility mea-
surements of fluorescent dyes using on-chip capil-
lary electrophoresis, Electrophoresis, 32, 3286-3294,
https://doi.org/10.1002/elps.201100210.
207. Antonenko, Y.N., Avetisyan, A. V., Cherepanov, D.A.,
Knorre, D. A., Korshunova, G. A., Markova, O. V.,
Ojovan, S. M., Perevoshchikova, I. V., Pustovidko,
A. V., Rokitskaya, T. I., Severina, I. I., Simonyan,
R.A., Smirnova, E.A., Sobko, A.A., Sumbatyan, N.V.,
Severin, F. F., and Skulachev, V. P. (2011) Derivatives
of rhodamine 19 as mild mitochondria-targeted cat-
ionic uncouplers, J.Biol. Chem., 286, 17831-17840,
https://doi.org/10.1074/jbc.M110.212837.
208. Kalinovich, A.V., and Shabalina, I.G. (2015) Novel mi-
tochondrial cationic uncoupler C4R1 is an effective
treatment for combating obesity in mice, Biochem-
istry (Moscow), 80, 620-628, https://doi.org/10.1134/
S0006297915050156.
209. Luo,X., Qian,L., Xiao,Y., Tang,Y., Zhao,Y., Wang,X.,
Gu,L., Lei, Z., Bao, J., Wu,J., He, T., Hu,F., Zheng, J.,
Li, H., Zhu, W., Shao, L., Dong, X., Chen, D., Qian, X.,
and Yang, Y. (2019) A diversity-oriented rhodamine
library for wide-spectrum bactericidal agents with
low inducible resistance against resistant patho-
gens, Nat. Commun., 10, 258, https://doi.org/10.1038/
s41467-018-08241-3.
210. Nazarov, P.A., Maximov, V.S., Firsov, A.M., Karakozova,
M. V., Panfilova, V., Kotova, E. A., Skulachev, M. V.,
and Antonenko, Y. N. (2024) Rhodamine 19 alkyl es-
ters as effective antibacterial agents, Int.J. Mol. Sci.,
25, 6137, https://doi.org/10.3390/ijms25116137.
211. Pfeffermann, J., Yadav, R., Glasnov, T., Thorn-
Seshold,O., and Pohl,P. (2025) Phototriggered proton-
selective carrier currents through photoswitch-
able membranes, bioRxiv, https://doi.org/10.1101/
2025.01.13.632814.
212. Rottenberg, H. (1983) Uncoupling of oxidative phos-
phorylation in rat liver mitochondria by general an-
esthetics, Proc. Natl. Acad. Sci. USA, 80, 3313-3317,
https://doi.org/10.1073/pnas.80.11.3313.
213. Dabadie, P., Bendriss, P., Erny, P., and Mazat, J.-P.
(1987) Uncoupling effects of local anesthetics on
rat liver mitochondria, FEBS Lett., 226, 77-82,
https://doi.org/10.1016/0014-5793(87)80554-9.
214. Terada, H., Shima, O., Yoshida, K., and Shinohara, Y.
(1990) Effects of the local anesthetic bupivacaine on
oxidative phosphorylation in mitochondria. Change
from decoupling to uncoupling by formation of a
leakage type ion pathway specific for H
+
in coop-
eration with hydrophobic anions, J.Biol. Chem.,
265, 7837-7842, https://doi.org/10.1016/S0021-9258
(19)39006-4.
215. Strichartz, G.R., Sanchez,V., Arthur, G.R., Chafetz,R.,
and Martin, D. (1990) Fundamental properties of lo-
cal anesthetics. II. Measured octanol:buffer parti-
tion coefficients and pKa values of clinically used
drugs, Anesth. Analg., 71, 158-170, https://doi.org/
10.1213/00000539-199008000-00008.
216. Sun, X., and Garlid, K. D. (1992) On the mechanism
by which bupivacaine conducts protons across the
membranes of mitochondria and liposomes, J. Biol.
Chem., 267, 19147-19154, https://doi.org/10.1016/
S0021-9258(18)41754-1.
217. Fantin, V. R., Berardi, M. J., Scorrano, L., Korsmeyer,
S. J., and Leder, P. (2002) A novel mitochondriotox-
ic small molecule that selectively inhibits tumor
cell growth, Cancer Cell, 2, 29-42, https://doi.org/
10.1016/S1535-6108(02)00082-X.
218. Wang, J., He, H., Xiang, C., Fan, X.-Y., Yang, L.-Y.,
Yuan,L., Jiang, F.-L., and Liu,Y. (2018) Uncoupling ef-
fect of F16 is responsible for its mitochondrial toxic-
ity and anticancer activity, Toxicol. Sci., 161, 431-442,
https://doi.org/10.1093/toxsci/kfx218.
219. Xu, J., He, H., Zhou, L. J., Liu, Y. Z., Li, D. W., Jiang,
F. L., and Liu, Y. (2018) Pyridinium and indole ori-
entation determines the mitochondrial uncou-
pling and anti-cancer efficiency of F16, Eur. J. Med.
Chem., 154, 305-313, https://doi.org/10.1016/j.ejmech.
2018.05.036.
220. Stupnikova, T. V., Kalafat, V. N., Klyuev, N. A., Mar-
shtupa, V. P., and Sagitullin, R. S. (1980) Vinylogous
anhydro bases of pyridylindoles, Chem. Heterocycl.
ANTONENKO et al.1844
BIOCHEMISTRY (Moscow) Vol. 90 No. 12 2025
Compd., 16, 1031-1035, https://doi.org/10.1007/
BF00496604.
221. Dubinin, M. V., Nedopekina, D. A., Ilzorkina, A. I.,
Semenova, A. A., Sharapov, V. A., Davletshin, E. V.,
Mikina, N. V., Belsky, Y. P., Spivak, A. Yu., Akatov,
V. S., Belosludtseva, N. V., Liu, J., and Belosludtsev,
K. N. (2023) Conjugation of triterpenic acids of ur-
sane and oleanane types with mitochondria-target-
ing cation F16 synergistically enhanced their cyto-
toxicity against tumor cells, Membranes, 13, 563,
https://doi.org/10.3390/membranes13060563.
222. Heise, N. V., Heisig, J., Meier, K., Csuk, R., and
Mueller, T. (2024) F16 Hybrids derived from steviol
or isosteviol are accumulated in the mitochondria of
tumor cells and overcome drug resistance, Molecules,
29, 381, https://doi.org/10.3390/molecules29020381.
223. Xing, W., Liu, G., Zhang, Y., Zhang, T., Lou, H., and
Fan, P. (2024) Selective antitumor effect and lower
toxicity of mitochondrion-targeting derivatization
of triptolide, J. Med. Chem., 67, 1093-1114, https://
doi.org/10.1021/acs.jmedchem.3c01508.
224. Miao, H., Cui, W., Zhang, T., Zhang, Y., Zhang, J.,
Lou, H., and Fan, P. (2024) Mitochondrial targeting
derivatives of honokiol enhanced selective antitu-
mor activity in NCI-H446 cells and decreased invivo
toxicity in Caenorhabditis elegans, Eur. J. Med.
Chem., 264, 115996, https://doi.org/10.1016/j.ejmech.
2023.115996.
225. Denisov, S.S., Kotova, E.A., Plotnikov, E.Y., Tikhonov,
A.A., Zorov, D.B., Korshunova, G.A., and Antonenko,
Y. N. (2014) A mitochondria-targeted protono-
phoric uncoupler derived from fluorescein, Chem.
Commun., 50, 15366-15369, https://doi.org/10.1039/
C4CC04996A.
226. Rokitskaya, T. I., Terekhova, N. V., Khailova, L. S.,
Kotova, E.A., Plotnikov, E.Y., Zorov, D.B., Tatarinov,
D. A., and Antonenko, Y. N. (2019) Zwitterionic pro-
tonophore derived from 2-(2-hydroxyaryl)alkenyl-
phosphonium as an uncoupler of oxidative phos-
phorylation, Bioconjug. Chem., 30, 2435-2443,
https://doi.org/10.1021/acs.bioconjchem.9b00516.
227. Blaikie, F.H., Brown, S.E., Samuelsson, L.M., Brand,
M. D., Smith, R. A., and Murphy, M. P. (2006) Target-
ing dinitrophenol to mitochondria: limitations to the
development of a self-limiting mitochondrial pro-
tonophore, Biosci. Rep., 26, 231-243, https://doi.org/
10.1007/s10540-006-9018-8.
228. Antonenko, Y. N., Denisov, S. S., Silachev, D. N.,
Khailova, L. S., Jankauskas, S. S., Rokitskaya, T. I.,
Danilina, T. I., Kotova, E. A., Korshunova, G. A.,
Plotnikov, E. Y., and Zorov, D.B. (2016) A long-linker
conjugate of fluorescein and triphenylphosphoni-
um as mitochondria-targeted uncoupler and fluo-
rescent neuro- and nephroprotector, Biochim. Bio-
phys. Acta, 1860, 2463-2473, https://doi.org/10.1016/
j.bbagen.2016.07.014.
229. Iaubasarova, I. R., Khailova, L. S., Nazarov, P. A.,
Rokitskaya, T. I., Silachev, D. N., Danilina, T. I., Plot-
nikov, E.Y., Denisov, S.S., Kirsanov, R.S., Korshunova,
G.A., Kotova, E.A., Zorov, D.B., and Antonenko, Y.N.
(2020) Linking 7-nitrobenzo-2-oxa-1,3-diazole (NBD)
to triphenylphosphonium yields mitochondria-target-
ed protonophore and antibacterial agent, Biochemis-
try (Moscow), 85, 1578-1590, https://doi.org/10.1134/
S000629792012010X.
230. Iaubasarova, I. R., Khailova, L. S., Firsov, A. M.,
Grivennikova, V. G., Kirsanov, R. S., Korshunova,
G. A., Kotova, E. A., and Antonenko, Y. N. (2020)
Themitochondria-targeted derivative of the classical
uncoupler of oxidative phosphorylation carbonyl cy-
anide m-chlorophenylhydrazone is an effective mito-
chondrial recoupler, PLoS One, 15, e0244499, https://
doi.org/10.1371/journal.pone.0244499.
231. Kirsanov, R. S., Khailova, L. S., Rokitskaya, T. I.,
Lyamzaev, K. G., Panteleeva, A. A., Nazarov, P. A.,
Firsov, A. M., Iaubasarova, I. R., Korshunova, G. A.,
Kotova, E. A., and Antonenko, Y. N. (2024) Synthe-
sis of triphenylphosphonium-linked derivative of
3,5-ditert-butyl-4-hydroxybenzylidene-malononitrile
(SF6847) via knoevenagel reaction yields an effec-
tive mitochondria-targeted protonophoric uncoupler,
ACS Omega, 9, 11551-11561, https://doi.org/10.1021/
acsomega.3c08621.
232. Terekhova, N. V., Khailova, L. S., Rokitskaya, T. I.,
Nazarov, P.A., Islamov, D.R., Usachev, K.S., Tatarinov,
D. A., Mironov, V. F., Kotova, E. A., and Antonenko,
Y. N. (2021) Trialkyl(vinyl)phosphonium chlorophe-
nol derivatives as potent mitochondrial uncouplers
and antibacterial agents, ACS Omega, 6, 20676-20685,
https://doi.org/10.1021/acsomega.1c02909.
233. Rokitskaya, T.I., Khailova, L.S., Strelnik, A.G., Kotova,
E.A., Mironov, V.F., Tatarinov, D.A., and Antonenko,
Y. N. (2025) Replacement of phenyl substituents at
phosphorus by hexyl ones in 2-(2-hydroxyaryl)vin-
ylphosphonium salts can tune the nature of the in-
duced proton transport through lipid membranes,
Biochim. Biophys. Acta Biomembr., 1867, 184417,
https://doi.org/10.1016/j.bbamem.2025.184417.
234. Listvan, V. N., and Listvan, V. V. (2003) Reactions of
phosphorus ylides with acyl chlorides: pathways and
preparative potential, Russ. Chem. Rev., 72, 705-713,
https://doi.org/10.1070/RC2003v072n08ABEH000800.
235. Kirsanov, R. S., Khailova, L. S., Rokitskaya, T. I.,
Iaubasarova, I. R., Nazarov, P. A., Panteleeva, A. A.,
Lyamzaev, K. G., Popova, L. B., Korshunova, G. A.,
Kotova, E. A., and Antonenko, Y. N. (2023) Ester-sta-
bilized phosphorus ylides as protonophores on bilay-
er lipid membranes, mitochondria and chloroplasts,
Bioelectrochemistry, 150, 108369, https://doi.org/
10.1016/j.bioelechem.2023.108369.
236. Rokitskaya, T. I., Kirsanov, R. S., Khailova, L. S.,
Panteleeva, A.A., Lyamzaev, K.G., Korshunova, G.A.,
MITOCHONDRIAL UNCOUPLERS 1845
BIOCHEMISTRY (Moscow) Vol. 90 No. 12 2025
Kotova, E. A., and Antonenko, Y. N. (2024) Methyla-
tion of phenyl rings in ester-stabilized phosphorus
ylides vastly enhances their protonophoric activi-
ty, Chembiochem., 25, e202300848, https://doi.org/
10.1002/cbic.202300848.
237. Fu, Y.Y., Zhang,M., Turner,N., Zhang, L.N., Dong, T.C.,
Gu,M., Leslie, S.J., Li, J.Y., Nan, F.J., and Li,J. (2013)
Anovel chemical uncoupler ameliorates obesity and
related phenotypes in mice with diet-induced obesity
by modulating energy expenditure and food intake,
Diabetologia, 56, 2297-2307, https://doi.org/10.1007/
s00125-013-2987-9.
238. Ferguson, S. M. F., Estrada-O, S., and Lardy, H. A.
(1971) Potassium-specific uncoupling by nigericin,
J.Biol. Chem., 246, 5645-5652, https://doi.org/10.1016/
S0021-9258(18)61855-1.
239. Johnson, R. B., Feldott, G., and Lardy, H. A. (1950)
Themode of action of the antibiotic, usnic acid, Arch.
Biochem., 28, 317-323.
240. Abo-Khatwa, A. N., Al-Robai, A. A., and Al-Jawhari,
D. A. (1996) Lichen acids as uncouplers of oxidative
phosphorylation of mouse-liver mitochondria, Nat.
Toxins, 4, 96-102, https://doi.org/10.1002/19960402NT7.
241. Antonenko, Y. N., Khailova, L. S., Rokitskaya,
T. I., Nosikova, E. S., Nazarov, P. A., Luzina, O. A.,
Salakhutdinov, N. F., and Kotova, E. A. (2019) Mech-
anism of action of an old antibiotic revisited: role
of calcium ions in protonophoric activity of usnic
acid, Biochim. Biophys. Acta Bioenerg., 1860, 310-316,
https://doi.org/10.1016/j.bbabio.2019.01.005.
242. Graven, S. N., Estrada-O, S., and Lardy, H. A. (1966)
Alkali metal cation release and respiratory inhi-
bition induced by nigericin in rat liver mitochon-
dria, Proc. Natl. Acad. Sci. USA, 56, 654-658, https://
doi.org/10.1073/pnas.56.2.654.
243. Toro, M., Gomez-Lojero, C., Montal, M., and
Estrada-O, S. (1976) Charge transfer mediated by
nigericin in black lipid membranes, J. Bioenerg., 8,
19-26, https://doi.org/10.1007/BF01559387.
244. Markin, V. S., and Sokolov, V. S. (1990) A new con-
cept of electrochemical membrane equlibrium cou-
pled transport and membrane potential, Bioelec-
trochem. Bioenerg., 23, 1-16, https://doi.org/10.1016/
0302-4598(90)80001-Y.
245. Whitehouse, M.W., and Dean, P.D.G. (1965) Biochem-
ical properties of anti-inflammatory drugs-V: uncou-
pling of oxidative phosphorylation by some γ-resor-
cyl and other dihydroxybenzol compounds, Biochem.
Pharmacol., 14, 557-567, https://doi.org/10.1016/
0006-2952(65)90228-5.
246. Rokitskaya, T. I., Kotova, E. A., and Antonenko,
Y. N. (2021) Anomalous potentials on bilayer lipid
membranes in the presence of usnic acid: Markin–
Sokolov versus Nernst–Donnan equilibrium, Bioelec-
trochemistry, 141, 107825, https://doi.org/10.1016/
j.bioelechem.2021.107825.
247. Andersen, O. S. (1984) Gramicidin channels, Annu.
Rev. Physiol., 46, 531-548, https://doi.org/10.1146/an-
nurev.ph.46.030184.002531.
248. Woolley, G. A., and Wallace, B. A. (1992) Model ion
channels– gramicidin and alamethicin, J.Membrane
Biol., 129, 109-136, https://doi.org/10.1007/BF00219508.
249. Andersen, O. S., and Koeppe, R. E. (1992) Molecu-
lar determinants of channel function, Physiol Rev.,
72, S89-S158, https://doi.org/10.1152/physrev.1992.72.
suppl_4.S89.
250. Mitchell, P., and Moyle, J. (1967) Respiration-driv-
en proton translocation in rat liver mitochondria,
Biochem. J., 105, 1147-1162, https://doi.org/10.1042/
bj1051147.
251. Grinius, L. L., Daugelabichius, R. I., and Al’kimavi-
chius, G. A. (1980) Study of membrane potential of
Bacillus subtilis and Escherichia coli cells by the pen-
etration ions methods [in Russian], Biokhimiia, 45,
1609-1618.
252. Matsuno-Yagi, A., and Hatefi, Y. (1989) Uncoupling
of oxidative phosphorylation: different effects of li-
pophilic weak acids and electrogenic ionophores
on the kinetics of ATP synthesis, Biochemistry, 28,
4367-4374, https://doi.org/10.1021/bi00436a037.
253. Nishio, J. N., and Whitmarsh, J. (1991) Dissipation
of the proton electrochemical potential in intact
and lysed chloroplasts: I. The electrical potential,
Plant Physiol., 95, 522-528, https://doi.org/10.1104/
pp.95.2.522.
254. Myers, V. B., and Haydon, D. A. (1972) Ion transfer
across lipid membranes in the presence of gram-
icidin A. II. The ion selectivity, Biochim. Biophys.
Acta, 274, 313-322, https://doi.org/10.1016/0005-
2736(72)90179-4.
255. Sorochkina, A. I., Plotnikov, E. Y., Rokitskaya, T. I.,
Kovalchuk, S. I., Kotova, E. A., Sychev, S. V., Zorov,
D. B., and Antonenko, Y. N. (2012) N-terminally glu-
tamate-substituted analogue of gramicidin A as pro-
tonophore and selective mitochondrial uncoupler,
PLoS One, 7, e41919, https://doi.org/10.1371/journal.
pone.0041919.
256. David, J. M., and Rajasekaran, A. K. (2015) Grami-
cidin A: a new mission for an old antibiotic, J. Kid-
ney Cancer VHL, 2, 15-24, https://doi.org/10.15586/
jkcvhl.2015.21.
257. Khailova, L. S., Rokitskaya, T. I., Kovalchuk, S. I.,
Kotova, E.A., Sorochkina, A. I., and Antonenko, Y.N.
(2019) Role of mitochondrial outer membrane in the
uncoupling activity of N-terminally glutamate-substi-
tuted gramicidin A, Biochim. Biophys. Acta Biomem-
br., 1861, 281-287, https://doi.org/10.1016/j.bbamem.
2018.06.013.
258. Haoyang, W.-W., Xiao, Q., Ye, Z., Fu, Y., Zhang, D.-W.,
Li, J., Xiao, L., Li, Z.-T., and Hou, J.-L. (2021) Grami-
cidin A-based unimolecular channel: cancer cell-tar-
geting behavior and ion transport-induced apoptosis,
ANTONENKO et al.1846
BIOCHEMISTRY (Moscow) Vol. 90 No. 12 2025
Chem. Commun., 57, 1097-1100, https://doi.org/10.1039/
D0CC08073J.
259. Silachev, D.N., Khailova, L.S., Babenko, V.A., Gulyaev,
M. V., Kovalchuk, S. I., Zorova, L. D., Plotnikov, E. Y.,
Antonenko, Y.N., and Zorov, D.B. (2014) Neuroprotec-
tive effect of glutamate-substituted analog of gramici-
dinA is mediated by the uncoupling of mitochondria,
Biochim. Biophys. Acta Gen. Subject, 1840, 3434-3442,
https://doi.org/10.1016/j.bbagen.2014.09.002.
260. Severin, F. F., Severina, I. I., Antonenko, Y. N.,
Rokitskaya, T. I., Cherepanov, D. A., Mokhova, E. N.,
Vyssokikh, M. Y., Pustovidko, A. V., Markova, O. V.,
Yaguzhinsky, L. S., Korshunova, G. A., Sumbatyan,
N.V., Skulachev, M.V., and Skulachev, V.P. (2010) Pene-
trating cation/fatty acid anion pair as a mitochondria-
targeted protonophore, Proc. Natl. Acad. Sci.
USA, 107, 663-668, https://doi.org/10.1073/pnas.
0910216107.
261. Rokitskaya, T. I., Sumbatyan, N. V., Tashlitsky, V. N.,
Korshunova, G. A., Antonenko, Y. N., and Skulachev,
V. P. (2010) Mitochondria-targeted penetrating cat-
ions as carriers of hydrophobic anions through lipid
membranes, Biochim. Biophys. Acta, 1798, 1698-1706,
https://doi.org/10.1016/j.bbamem.2010.05.018.
262. Lyamzaev, K. G., Pustovidko, A. V., Simonyan, R. A.,
Rokitskaya, T. I., Domnina, L. V., Ivanova, O. Yu.,
Severina, I. I., Sumbatyan, N. V., Korshunova, G. A.,
Tashlitsky, V. N., Roginsky, V. A., Antonenko, Y. N.,
Skulachev, M. V., Chernyak, B. V., and Skulachev,
V. P. (2011) Novel mitochondria-targeted antioxi-
dants: plastoquinone conjugated with cationic plant
alkaloids berberine and palmatine, Pharm. Res., 28,
2883-2895, https://doi.org/10.1007/s11095-011-0504-8.
263. Chernyak, B. V., Antonenko, Y. N., Domnina, L. V.,
Ivanova, O. Yu., Lyamzaev, K. G., Pustovidko, A. V.,
Rokitskaya, T.I., Severina, I.I., Simonyan, R.A., Tren-
deleva, T. A., and Zvyagilskaya, R. A. (2013) Novel
penetrating cations for targeting mitochondria, Curr.
Pharm. Des., 19, 2795-2806, https://doi.org/10.2174/
1381612811319150015.
264. Garlid, K.D., and Nakashima, R.A. (1983) Studies on
the mechanism of uncoupling by amine local anes-
thetics. Evidence for mitochondrial proton trans-
port mediated by lipophilic ion pairs, J.Biol. Chem.,
258, 7974-7980, https://doi.org/10.1016/S0021-9258
(20)82015-8.
265. Stark, G. (1980) Negative hydrophobic ions as trans-
port-mediators for positive ions. Evidence for a car-
rier mechanism, Biochim. Biophys. Acta Biomembr.,
600, 233-237, https://doi.org/10.1016/0005-2736(80)
90428-9.
266. Ahmed, I., and Krishnamoorthy, G. (1990) En-
hancement of transmembrane proton conductivi-
ty of protonophores by membrane-permeant cat-
ions, Biochim. Biophys. Acta, 1024, 298-306, https://
doi.org/10.1016/0005-2736(90)90358-U.
267. Kolajova,M., Antalik,M., and Sturdik,E. (1993) Study
of the complex formation between amine local anes-
thetics and uncouplers of oxidative phosphorylation
carbonyl cyanide phenylhydrazones, Gen. Physiol.
Biophys., 12, 213-229.
268. Antonenko, Y. N., Khailova, L. S., Knorre, D. A., Mar-
kova, O.V., Rokitskaya, T.I., Ilyasova, T.M., Severina,
I. I., Kotova, E.A., Karavaeva, Y. E., Prikhodko, A.S.,
Severin, F.F., and Skulachev, V. P. (2013) Penetrating
cations enhance uncoupling activity of anionic pro-
tonophores in mitochondria, PLoS One, 8, e61902,
https://doi.org/10.1371/journal.pone.0061902.
269. Wu, X., and Gale, P. A. (2016) Small-molecule un-
coupling protein mimics: synthetic anion receptors
as fatty acid-activated proton transporters, J. Am.
Chem. Soc., 138, 16508-16514, https://doi.org/10.1021/
jacs.6b10615.
270. Antonenko, Y. N., Veselov, I. M., Rokitskaya, T. I.,
Vinogradova, D. V., Khailova, L. S., Kotova, E. A.,
Maltsev, A. V., Bachurin, S. O., and Shevtsova, E. F.
(2024) Neuroprotective thiourea derivative uncou-
ples mitochondria and exerts weak protonophoric
action on lipid membranes, Chem. Biol. Interact., 402,
111190, https://doi.org/10.1016/j.cbi.2024.111190.
271. York,E., McNaughton, D.A., Duman, M.-N., Gale, P.A.,
and Rawling, T. (2023) Fatty acid-activated proton
transport by bisaryl anion transporters depolaris-
es mitochondria and reduces the viability of MDA-
MB-231 breast cancer cells, Biomolecules, 13, 1202,
https://doi.org/10.3390/biom13081202.
272. Shkinev, A. V., Gagelgans, A. I., Tashmukhamedov,
B.A., and Miroshnikov, A.I. (1978) Effect of melittin
from bee venom on mitochondrial functions [in Rus-
sian], Biokhimiia, 43, 1452-1457.
273. Sholtz, K.F., Aliverdieva, D.A., and Kotelnikova, A.V.
(1985) Action of alamethicin on the coupling mem-
brane of mitochondria [in Russian], Dokl. Akad. Nauk
SSSR, 283, 723-727.
274. Hugosson,M., Andreu,D., Boman, H.G., and Glaser,E.
(1994) Antibacterial peptides and mitochondrial
presequences affect mitochondrial coupling, res-
piration and protein import, Eur. J. Biochem., 223,
1027-1033, https://doi.org/10.1111/j.1432-1033.1994.
tb19081.x.
275. Diaz-Achirica, P., Prieto, S., Ubach, J., Andreu, D.,
Rial, E., and Rivas, L. (1994) Permeabilization of
the mitochondrial inner membrane by short ce-
cropin-A-melittin hybrid peptides, Eur. J. Biochem.,
224, 257-263, https://doi.org/10.1111/j.1432-1033.
1994.tb20019.x.
276. Aliverdieva, D., Mamaev, D., Snezhkova, L., and
Sholtz, C. (2012) Evaluation of molecularity of
rate-limiting step of pore formation by antimicro-
bial peptides studied using mitochondria as a bio-
sensor, Toxicol In Vitro, 26, 939-949, https://doi.org/
10.1016/j.tiv.2012.04.006.
MITOCHONDRIAL UNCOUPLERS 1847
BIOCHEMISTRY (Moscow) Vol. 90 No. 12 2025
277. Lees, H. (1967) The effects of laurylamine on res-
piration and related reactions of liver mitochon-
dria in vitro, Biochim. Biophys. Acta, 131, 310-316,
https://doi.org/10.1016/0005-2728(67)90144-2.
278. Bragadin,M., and Dell’Antone,P. (1996) Mitochondri-
al bioenergetics as affected by cationic detergents,
Arch. Environ. Contam. Toxicol., 30, 280-284, https://
doi.org/10.1007/BF00215809.
279. Bragadin, M., Moret, I., Piazza, R., Grasso, M., and
Manente, S. (2002) The interactions of cetyltrimeth-
ylammonium with mitochondria: an uncoupler or a
detergent? Chemosphere, 46, 219-223, https://doi.org/
10.1016/S0045-6535(01)00095-9.
280. Bangham, J. A., and Lea, E. J. (1978) The interaction
of detergents with bilayer lipid membranes, Biochim.
Biophys. Acta, 511, 388-396, https://doi.org/10.1016/
0005-2736(78)90275-4.
281. Kondrashov, O.V., and Akimov, S.A. (2023) Alteration
of average thickness of lipid bilayer by membrane-
deforming inclusions, Biomolecules, 13, 1731, https://
doi.org/10.3390/biom13121731.
282. Antonenko, Y. N., Rokitskaya, T. I., Cooper, A. J. L.,
and Krasnikov, B.F. (2010) Minocycline chelatesCa
2+
,
binds to membranes, and depolarizes mitochondria
by formation of Ca
2+
-dependent ion channels, J. Bio-
energ. Biomembr., 42, 151-163, https://doi.org/10.1007/
s10863-010-9271-1.
283. Sokolov, S., Zyrina, A., Akimov, S., Knorre, D., and
Severin, F. (2023) Toxic effects of penetrating cat-
ions, Membranes, 13, 841, https://doi.org/10.3390/
membranes13100841.
284. Terada,H., Nagamune,H., Osaki,Y., and Yoshikawa,K.
(1981) Specific requirement for inorganic phosphate
for induction of bilayer membrane conductance by
the cationic uncoupler carbocyanine dye, Biochim.
Biophys. Acta, 646, 488-490, https://doi.org/10.1016/
0005-2736(81)90320-5.
285. Ter Welle, H. F., and Slater, E. C. (1967) Uncoupling
of respiratory-chain phosphorylation by arsenate,
Biochim. Biophys. Acta, 143, 1-17, https://doi.org/
10.1016/0005-2728(67)90104-1.
286. Skulachev, V. P. (1998) Uncoupling: new approaches
to an old problem of bioenergetics, Biochim. Bio-
phys. Acta, 1363, 100-124, https://doi.org/10.1016/
S0005-2728(97)00091-1.
287. Wallace, K. B., and Starkov, A. A. (2000) Mitochon-
drial targets of drug toxicity, Annu. Rev. Pharmacol.
Toxicol., 40, 353-388, https://doi.org/10.1146/annurev.
pharmtox.40.1.353.
288. Sherman, J. K. (2025) Correlation in ultrastructural
cryoinjury of mitochondria with aspects of their re-
spiratory function, Exp. Cell Res., 66, 378-384, https://
doi.org/10.1016/0014-4827(71)90691-4.
289. Guengerich, F. P. (2008) Cytochrome p450 and chem-
ical toxicology, Chem. Res. Toxicol., 21, 70-83, https://
doi.org/10.1021/tx700079z.
290. Rowland, A., Miners, J. O., and Mackenzie, P. I.
(2013) The UDP-glucuronosyltransferases: their role
in drug metabolism and detoxification, Int. J. Bio-
chem. Cell Biol., 45, 1121-1132, https://doi.org/10.1016/
j.biocel.2013.02.019.
291. Bock, K. W. (2014) Homeostatic control of xeno- and
endobiotics in the drug-metabolizing enzyme sys-
tem, Biochem. Pharmacol., 90, 1-6, https://doi.org/
10.1016/j.bcp.2014.04.009.
292. Gamage, N., Barnett, A., Hempel, N., Duggleby, R. G.,
Windmill, K. F., Martin, J. L., and McManus, M. E.
(2006) Human sulfotransferases and their role in
chemical metabolism, Toxicol. Sci., 90, 5-22, https://
doi.org/10.1093/toxsci/kfj061.
293. Josephy, P.D. (2010) Genetic variations in human glu-
tathione transferase enzymes: significance for phar-
macology and toxicology, Hum. Genomics Proteomics,
2010, 876940, https://doi.org/10.4061/2010/876940.
294. Sim, E., Westwood, I., and Fullam, E. (2007) Aryl-
amine N-acetyltransferases, Expert. Opin. Drug
Metab. Toxicol., 3, 169-184, https://doi.org/10.1517/
17425255.3.2.169.
295. Mato, J.M., Martinez-Chantar, M.L., and Lu, S.C. (2013)
S-adenosylmethionine metabolism and liver disease,
Ann. Hepatol., 12, 183-189, https://doi.org/10.1016/
S1665-2681(19)31355-9.
296. Jiang, A., Chen, G., Xu, J., Liu, Y., Zhao, G., Liu, Z.,
Chen, T., Li, Y., and James, T. D. (2019) Ratiometric
two-photon fluorescent probe for in situ imaging of
carboxylesterase (CE)-mediated mitochondrial acid-
ification during medication, Chem. Commun., 55,
11358-11361, https://doi.org/10.1039/C9CC05759E.
297. Miwa,S., Treumann,A., Bell,A., Vistoli,G., Nelson,G.,
Hay, S., and von Zglinicki, T. (2016) Carboxylester-
ase converts Amplex red to resorufin: implications
for mitochondrial H
2
O
2
release assays, Free Rad-
ic. Biol. Med., 90, 173-183, https://doi.org/10.1016/
j.freeradbiomed.2015.11.011.
298. Cooper, A. J. L., and Hanigan, M. H. (2018) Metab-
olism of glutathione S-conjugates: multiple path-
ways, Comprehens. Toxicol., 10, 406, https://doi.org/
10.1016/B978-0-12-801238-3.01973-5.
299. Sidhu, R.S., and Blair, A.H. (1975) Human liver alde-
hyde dehydrogenase. Esterase activity, J.Biol. Chem.,
250, 7894-7898, https://doi.org/10.1016/S0021-9258
(19)40899-5.
300. Zemaitis, M. A., and Greene, F. E. (1976) Impairment
of hepatic microsomal and plasma esterases of the
rat by disulfiram and diethyldithiocarbamate, Bio-
chem. Pharmacol., 25, 453-459, https://doi.org/10.1016/
0006-2952(76)90349-X.
301. Nousiainen, U., and Torronen, R. (1984) Differentia-
tion of microsomal and cytosolic carboxylesterases
in the rat liver by in vivo and in vitro inhibition,
Gen. Pharmacol., 15, 223-227, https://doi.org/10.1016/
0306-3623(84)90163-0.
ANTONENKO et al.1848
BIOCHEMISTRY (Moscow) Vol. 90 No. 12 2025
302. Buechel, K. H., and Schaefer, G. (1970) 2-Anilinothio-
phenes as uncouplers of oxidative phosphorylation
in mitochondria [in German], Z. Naturforsch., 25 b,
1465-1474, https://doi.org/10.1515/znb-1970-1232.
303. Skulachev, V. P., Vyssokikh, M. Y., Chernyak, B. V.,
Mulkidjanian, A.Y., Skulachev, M.V., Shilovsky, G.A.,
Lyamzaev, K. G., Borisov, V. B., Severin, F. F., and
Sadovnichii, V.A. (2023) Six functions of respiration:
isn’t it time to take control over ROS production in
mitochondria, and aging along with it? Int. J. Mol.
Sci., 24, 12540, https://doi.org/10.3390/ijms241612540.
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.