ISSN 0006-2979, Biochemistry (Moscow), 2025, Vol. 90, No. 12, pp. 1789-1810 © Pleiades Publishing, Ltd., 2025.
Russian Text © The Author(s), 2025, published in Biokhimiya, 2025, Vol. 90, No. 12, pp. 1917-1940.
1789
REVIEW
Ca
2+
-Dependent Mitochondrial Permeability
Transition Pore: Structure, Properties,
and Role in Cellular Pathophysiology
Konstantin N. Belosludtsev
1,2,a
*, Mikhail V. Dubinin
1
, and Natalia V. Belosludtseva
2
1
Mari State University, 424000 Yoshkar-Ola, Mari El Republic, Russia
2
Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences,
142290 Pushchino, Moscow Region, Russia
a
e-mail: bekonik@gmail.com
Received July 30, 2025
Revised September 9, 2025
Accepted September 22, 2025
AbstractThe Mitochondrial Permeability Transition pore(MPT pore) activated by Ca
2+
ions is a phenomenon
that has long been the subject of intense study. Cyclophilin D-dependent opening of the MPT pore in mito-
chondria in response to calcium overload and oxidative stress leads to swelling of the mitochondrial matrix,
depolarization of the inner membrane and dysregulation of ion homeostasis. These processes are accompanied
by damage to mitochondrial membranes and, ultimately, to cell death. Despite decades of research, the molecular
identity of the MPT pore remains unclear. Currently, the inner membrane proteins– ATP synthase and adenine
nucleotide translocator(ANT)– are considered to be its key structural components, along with the regulatory
protein cyclophilinD. The involvement of the MPT pore in the progression of various pathological conditions
and diseases, as well as in a number of physiological processes, such as the regulation of cellular bioenergetics
and rapid release of Ca
2+
, is widely discussed. This review summarizes modern molecular genetic data on the
putative structure of the MPT pore, traces the evolution of views on its functioning– from interpreting it as
a simple experimental artifact to its recognition as a putative key regulator of energy metabolism– and also
considers the mechanisms of its regulation and its multifaceted pathophysiological role.
DOI: 10.1134/S0006297925602369
Keywords: MPT pore, mitochondria, Ca
2+
, cyclophilinD, adenine nucleotide translocator, F
0
F
1
–ATP synthase, cell
death, neurodegenerative diseases, neuromuscular diseases, diabetes mellitus
* To whom correspondence should be addressed.
INTRODUCTION
Mitochondria are organelles that perform a wide
range of functions in eukaryotic cells. They are the
primary energy generators in most types of cells, they
participate in the regulation of ion homeostasis and
thermogenesis, and their production of reactive oxy-
gen species (ROS) is closely linked to aging and cell
death. However, one of the most intriguing and enig-
matic phenomena associated with these organelles is
the formation and functioning of the Mitochondrial
Permeability Transition pore (MPT pore), which is
formed by a complex of mitochondrial proteins under
a wide range of pathophysiological conditions.
The MPT pore is a megachannel formed in the
outer and inner mitochondrial membranes as a re-
sult of exposure to high concentrations of intracel-
lular Ca
2+
, oxidative stress and other inducers [1].
Despite more than 70years of studying this phenom-
enon, the exact molecular structure of the MPT pore
has not been established yet. The main difficulty lies
in identifying the protein components that form the
pore in the inner mitochondrial membrane. Current-
ly, the most abundant proteins of the inner mitochon-
drial membrane are traditionally considered as such
components, specifically the F
0
F
1
–ATP synthase (com-
plex  V) and adenine nucleotide translocator (ANT)
[2, 3]. At the same time, cyclophilin  D, the key reg-
ulator of the MPT pore, as well as the proteins and
protein subunits involved in the formation of the
BELOSLUDTSEV et al.1790
BIOCHEMISTRY (Moscow) Vol. 90 No. 12 2025
pore complex in the intermembrane space and the
outer membrane of organelles, have been discussed
quite rarely in recent years.
The pathophysiological significance of MPT pore
formation in mitochondria is currently undisputed
and highlights the dual role of these organelles in
cell life and death. It is believed that a prolonged
open state of the MPT pore leads to mitochondrial
and cell death, whereas its short-term “flickering”
opening facilitates the removal of excess Ca
2+
ions
from the mitochondria into the cytoplasm, protecting
cells from the damaging effects of calcium overload
on organelles [3-7].
Based on existing observations, new natural and
synthetic modulators of the MPT pore are being ac-
tively investigated, and genetic manipulations of
known components of the pore complex are being
conducted in model systems. This allows to establish
of a link between the functioning of the pore in mi-
tochondria and the development of various cellular
pathologies. This review analyzes the current state
of research regarding the mechanisms of MPT pore
formation, regulation and functioning, as well as the
prerequisites that have led to the current under-
standing of the nature of this phenomenon in cells,
its physiological and pathological role, and key unre-
solved issues.
THE HISTORY OF PORE STRUCTURE STUDIES
The history of studying the mitochondrial pore
had started long before the term “mitochondrial per-
meability transition” (Fig.  1) was coined. As early as
the 1950s, it was discovered that mitochondria have
the ability to swell, which leads to impaired ATP syn-
thesis [8, 9]. However, the observed mitochondrial
permeabilization was thought to be either an in  vitro
artifact or a consequence of nonspecific damage
to their inner membrane by phospholipase  A
2
[10].
Ca
2+
was found to activate phospholipase  A
2
, whose
hydrolytic activity results in the accumulation of “de-
fects” – lysophospholipids and free fatty acids – in
mitochondrial membranes. These were believed to be
the primary cause of changes in mitochondrial mem-
brane permeability[11]. However, this hypothesis did
not explain why rapid and dramatic swelling of mi-
tochondria occurs if the accumulation of membrane
defects associated with phospholipase  A
2
activity oc-
curs gradually. However, at the turn of the 21st centu-
ry, studies based on this hypothesis were conducted,
resulting in the discovery of a new type of mitochon-
drial pore – a lipid pore inducible by saturated fatty
acids and Ca
2+
[12, 13].
In the late 1970s, permeabilization of the inner
mitochondrial membrane was described in detail[14-
16]. It was found that when Ca
2+
ions accumulate in
mitochondria, their inner membrane becomes per-
meable to hydrophilic compounds with a molecular
weight of up to 1.5  kDa, resulting in swelling of the
organelles. The influence of the major metabolites
on this process was studied, and regulation of inner
membrane permeabilization was demonstrated. Based
on these studies, it was suggested that a non-selec-
tive Ca
2+
-dependent channel is formed in the mito-
chondrial membrane in the presence of Ca
2+
ions,
Fig. 1. Timeline of MPT pore research. ANT – adenine nucleotide translocator.
MPT PORE: STRUCTURE AND FUNCTIONS 1791
BIOCHEMISTRY (Moscow) Vol. 90 No. 12 2025
which can be inhibited by chelating agents. Since
the molecular nature of the channel remained un-
known, the authors proposed the term “permeability
transition” to describe this phenomenon (meaning a
sharp non-specific change in permeability). Since the
change occurred abruptly according to the “all or
nothing” principle[1,14], it was discussed specifically
as a “transition” (and, subsequently, the mitochondri-
al pore) and not a change in mitochondrial permea-
bility, in order to emphasize the transitive nature of
the phenomenon.
In the late 1980s, a high-affinity inhibitor of the
mitochondrial pore, the cyclic undecapeptide cyclo-
sporin  A(CsA) was discovered. It completely inhibited
the MPT pore opening at submicromolar concentra-
tions [17, 18]. It was established that CsA binds to
the mitochondrial matrix protein, peptidyl prolyl cis/
trans isomerase cyclophilin  D [19]. This discovery fi-
nally confirmed the concept of the protein nature of
the mitochondrial pore.
It was initially believed that the target of cyclo-
philin D and the channel component of the pore is
the adenine nucleotide translocator, the predominant
protein of the inner mitochondrial membrane respon-
sible for the antiport of ADP and ATP. The prerequi-
sites for this were discovered back in the 1970s, when
it became clear that the ANT modulators carboxya-
tractylate and bongkrekic acid activate and inhibit
Ca
2+
-dependent mitochondrial swelling, respective-
ly [14]. Later, Halestrap and Davidson [20] proposed
a mechanism of action for these inhibitors, according
to which carboxyatractylate stabilized ANT in the cy-
tosolic(c) conformation, and bongkrekic acid– in the
matrix(m) conformation[20]. The hypothesis of a key
role for ANT existed until the mid-2000s, when it was
discovered that ANT is not an essential component of
the MPT pore. In mitochondria of mice with genetic
knockout of the first and second ANT isoforms (ANT1
and ANT2), the MPT pore opened and remained sen-
sitive to cyclosporin A [21].
The search for new channel components of the
MPT pore led to suggestions that such a component
could be a phosphate transporter, and, later, a mito-
chondrial ATP synthase [22, 23]. These proteins have
a common feature of being able to interact with cy-
clophilin  D. However, interest in the phosphate trans-
porter quickly became irrelevant, since its knockdown
or overexpression did not affect pore opening.
Mitochondrial ATP synthase is currently con-
sidered a key component of the MPT pore. Its in-
volvement (or the involvement of individual ATP
synthase subunits) in membrane permeabilization
has been demonstrated by various methods. Sever-
al models for the transformation of ATP synthase
into the MPT pore have been proposed [3]. How-
ever, ANT has recently come to be regarded as an
important component of the MPT pore once more,
responsible for its low-conductance mode of oper-
ation [3].
However, even today we can only discuss a hy-
pothetical structure of the MPT pore. This is primar-
ily due to the limitations of current methodological
approaches.
MPT PORE STRUCTURE
According to current concepts, the mitochondrial
pore is a large protein megachannel formed in the
outer and inner mitochondrial membranes. Depending
on the conditions, the size of the MPT pore can vary
from a small non-selective ion channel to a large pore
with high conductivity (up to 1000pS), through which
ions and hydrophilic molecules with a mass of up to
1.5  kDa can penetrate. Prolonged MPT pore opening
leads to suppression of mitochondrial energy metab-
olism, disruption of ion homeostasis and a drop in
mitochondrial membrane potential. Furthermore, the
entry of osmotically active metabolites into the mito-
chondrial matrix due to MPT pore opening leads to
swelling, followed by rupture of the outer mitochon-
drial membrane. All this, along with the release of
proapoptotic proteins from mitochondria, can lead to
organelle destruction and cell death [1, 3, 7, 12, 24].
These mechanisms of cell death, mediated by the
opening of the mitochondrial pore, play a key role
in the pathogenesis of numerous diseases associated
with ischemic damage to organs and tissues, neuro-
degenerative diseases, aging, etc. At the same time,
the temporary opening of the MPT pore, character-
ized by a low-conductivity state, is of physiological
significance for the cell. This type of pore is involved
in the unloading of Ca
2+
ions from mitochondria, a
decrease in ROS production and regulation of bio-
energetic metabolism [6,  24,  25]. Activation of the
pore is believed to be associated primarily with an
increase in Ca
2+
concentration in the mitochondrial
matrix, development of oxidative stress, an increase
in cyclophilin D levels and a decrease in the pool of
adenine nucleotides. At the same time, di- and triva-
lent cations, adenine nucleotides, SH-reducing agents
and cyclophilinD inhibitors suppress MPT pore open-
ing. More details on this can be found in the review
by Zoratti and Szabo [1].
The abovementioned facts make it clear that
MPT pore formation in mitochondria is a complexly
regulated process that may involve a large number
of proteins. Indeed, current models suggest a multi-
protein nature of the pore, with ANT, ATP synthase,
cyclophilin  D and other proteins forming a dynamic
“sensory cluster” that adapts to particular conditions
in a cell (Fig.  2) [2, 3].
BELOSLUDTSEV et al.1792
BIOCHEMISTRY (Moscow) Vol. 90 No. 12 2025
Fig. 2. The proteins forming the MPT pore in mitochondria and their transformation into pore channels under the influence
of Ca
2+
ions and/or oxidative stress. Fluxes of osmotically active molecules through the pore channels in the inner and
outer membranes upon pore induction are shown. PDB IDs used: ANT– 1OKC; ATP synthase (both) – 6RD4; VDAC – 2JK4;
CypD – 5CBV. Descriptions are given in the text. ANT – adenine nucleotide translocator; VDAC – voltage-dependent anion
channel of the outer mitochondrial membrane; CypD– cyclophilin D.
MPT PORE: STRUCTURE AND FUNCTIONS 1793
BIOCHEMISTRY (Moscow) Vol. 90 No. 12 2025
When describing the mitochondrial pore model,
three groups of proteins can be distinguished: the
obligatory regulator – cyclophilin  D; hypothetical
channels of the inner membrane pore – ANT and
ATP synthase; auxiliary proteins, which include
VDAC (voltage-dependent anion channels of the out-
er mitochondrial membrane), TSPO(translocator pro-
tein, also known as the peripheral benzodiazepine
receptor), mitochondrial creatine kinase, mitochon-
drial hexokinase and a number of other proteins.
Cyclophilin  D is a key regulator of the MPT pore.
Cyclophilin  D is a protein belonging to the CsA-sensi-
tive peptidyl prolyl cis/trans isomerase family capable
of catalyzing the isomerization of proline residues, a
rate-limiting step in protein folding [26]. All proteins
of this family contain a highly conserved cyclophi-
lin-like domain consisting of 8 antiparallel β-sheets
that form a hydrophobic core with the catalytic cen-
ter and other functional regions [19]. Cyclophilin  D
is encoded by the Ppif gene, the mature protein is
localized in the mitochondrial matrix and contains
178 amino acid residues (19  kDa) [19].
Cyclophilin  D is able to interact with a large
number of various transporters which are associat-
ed with membrane proteins, including ATP synthase,
ANT, respiratory chain complex  III, the anti-apoptotic
Bcl2 protein and the key metabolic protein kinases
GSK-3β and Akt2 [19, 27-29]. Due to this, cyclophil-
in D is able to regulate the formation of various mi-
tochondrial supercomplexes. However, its most well-
described function is regulation of MPT pore forma-
tion. Cyclophilin D modulates pore formation by re-
ducing the threshold level of calcium load required
for pore opening in mitochondria; this requires the
interaction of cyclophilin  D with ATP synthase or ANT,
which form a channel in the pore complex[2,3]. This
effect is suppressed in the presence of cyclosporin  A,
which is believed to cause dissociation of cyclophilin
D from channel proteins[20]. Mitochondria from var-
ious tissues of mice with the knocked out Ppif gene
(and, consequentially, lacking the interaction of pore
proteins with cyclophilin  D) were more resistant to
Ca
2+
overload, and the addition of CsA had no inhib-
itory effect in this case. At the same time, overex-
pression of the Ppif gene caused an increase in Ca
2+
-
dependent mitochondrial swelling and cell death[30].
The exact mechanism of modulation of MPT pore
formation involving cyclophilin  D has not been ful-
ly established. Early studies demonstrated that MPT
pore formation requires interaction of cyclophilin  D
with ANT and VDAC proteins [29]. However, it is
now recognized that cyclophilin D binds primarily
to ATP synthase [3, 28] (Fig.  2). In the ATP synthase
complex, cyclophilin  D interacts with OSCP (oligomy-
cin sensitivity conferring protein), which appears to
be involved in peripheral stalk formation [23, 28].
The issue of whether the enzymatic activity of cyclo-
philin  D is required also remains debatable. It was
initially assumed that cyclophilin  D induces MPT
pore formation independently of its peptidyl prolyl
cis/trans isomerase activity  [31]. However, fibroblasts
with inactivated cyclophilin  D (Arg96Gly substitution)
were insensitive to oxidative stress induction, simi-
lar to what was observed in cells with a knockout
of this protein  [30]. This suggests that the enzymatic
activity of the protein does facilitate MPT pore acti-
vation. Furthermore, peptidyl prolyl cis/trans isomer-
ase activity correlates with increased self-assembly of
ATP synthasomes (a supercomplex consisting of ATP
synthase, ANT and a phosphate transporter) in vari-
ous tissues and the formation of highly ordered ATP
synthase oligomeric structures, thereby reducing the
likelihood of MPT pore formation [32].
The ability of cyclophilin  D to modulate MPT
pore opening is provided in the cell by mechanisms
of post-translational protein modification [19]. For
example, acetylation of Lys138 stimulates MPT pore
formation [33]. It is believed that Lys138 is localized
in the catalytic site of the protein and is involved in
the interaction with OSCP [23]. GSK-3β increases cy-
clophilin  D binding to OSCP and stimulates MPT pore
opening by phosphorylating Ser162 [34, 35]. Cleavage
of the N-terminal region of cyclophilin  D by calpain  1
can also contribute to an increase in the probability
of MPT pore opening by enhancing the interaction
with OSCP [36]. Oxidative modification of cyclophil-
in  D at cysteine residues (Cys174(C203) in particular)
is involved in the process of activation of MPT pore
formation in the presence of oxidative stress induc-
ers [37].
ANT and ATP synthase as channel proteins of
the pore complex. The adenine nucleotide transloca-
tor is a protein of the SLC25 family of mitochondri-
al transporters that transport metabolites, inorganic
ions and cofactors [38]. ANT exchanges matrix ATP
for cytoplasmic ADP across the inner mitochondrial
membrane [39]. It is also believed that one of the
alternative functions of ANT is fatty acid-catalyzed
proton transfer, which ensures uncoupling of oxida-
tive phosphorylation. In humans, 4  ANT isoforms are
present (while mice lack the Ant3 gene) [40], with a
molecular weight of ~30  kDa. The protein structure
contains 6 transmembrane α-helices [41] (Fig. 2).
As mentioned above, the involvement of ANT
in MPT pore formation was suggested in the 1970s,
and in the 1990s, understanding of the mechanism
of ANT transformation into the mitochondrial pore
was developed [14, 20, 42]. Indeed, ANT inhibitors
and substrates significantly affected Ca
2+
capacity
and swelling of organelles [14]. When ANT was in-
corporated into giant vesicles, channel conductance
(up to 600  pS) was observed [43]. It was shown that
BELOSLUDTSEV et al.1794
BIOCHEMISTRY (Moscow) Vol. 90 No. 12 2025
oxidative modification of cysteine residues (Cys57 and
Cys160) of ANT caused formation of disulfide bridg-
es in the protein, increased mitochondrial sensitivity
to MPT pore induction through enhanced cyclophil-
in  D binding and suppressed the inhibitory effect of
ADP [44]. All this convincingly indicated the involve-
ment of ANT in MPT pore formation.
However, the results of certain experiments in
the mid-2000s almost made mitochondriologists com-
pletely abandon the idea of ANT as a part of the MPT
pore. It was shown that liver mitochondria from mice
with knockouts of the Ant1 and Ant2 genes were still
sensitive to Ca
2+
-dependent mitochondrial swelling,
which was prevented by cyclosporin  A. Moreover,
such mitochondria had increased Ca
2+
capacity (com-
pared to liver mitochondria from control animals),
which was reduced by the addition of oxidizing
agents, but not atractyloside (an ANT inhibitor and
MPT pore inducer) [21]. Only recently it was shown
that knockouts of all three ANT genes (Ant1, Ant2,
Ant4) in mice resulted in liver mitochondria becom-
ing virtually insensitive to Ca
2+
ions, while the ad-
dition of cyclosporin  A or knockout of the Ppif gene
resulted in complete inhibition of the pore. Patch
clamp analysis of mitoplasts isolated from mouse em-
bryonic fibroblasts with knockouts of all three ANT
genes showed virtually no channel conductance. This
allowed to conclude that ANT is an essential compo-
nent of the MPT pore, which induces the formation
of a low-conductance pore [45].
Interestingly, increased expression of different
ANT isoforms is observed in various pathologies. For
example, in Duchenne muscular dystrophy, a decrease
in ANT1 expression and an increase in ANT2 levels
are observed in skeletal muscle cells [46]. In he-
patocytes of diabetic mice (type  1 diabetes mellitus),
a decrease in ANT1 levels was also observed [47].
On the other hand, suppression of ANT2 expression
in HEK293T cells (a cell line derived from human em-
bryonic kidneys) even enhanced the progression of
mitochondrial dysfunction under hyperlipidemic con-
ditions [48]. All this may indicate tissue-specific reg-
ulation of MPT pore activity in various pathologies.
It is believed that ANT is capable of transforming
into a channel under the influence of Ca
2+
ions [42].
However, ANT does not have a Ca
2+
binding site,
which suggests the effects of additional factors in
the regulation of the MPT pore (possibly cardiolip-
in) or post-translational modification [2,  49]. There
are doubts about the necessity of ANT interaction
with cyclophilin D for MPT pore formation [2,  3].
All this allows to assume that another channel
that ensures the opening of high-conductance MPT
pore can be formed in mitochondria. Surprising-
ly, a slightly incorrect interpretation of the experi-
ment conducted in the mid-2000s led to significant
progress in the search for new structural components
of the MPT pore. As a result, ATP synthase is cur-
rently considered as the key channel component of
the MPT pore.
Mitochondrial ATP synthase is one of the key mi-
tochondrial supercomplexes (~600  kDa), consisting of
a F1 complex (ATP synthesizing) and a membrane-
embedded F0. The complexes interact via a central
and a peripheral stalk. The F1 complex consists of
three pairs of αβsubunits arranged around the central
stalk (γ, δ, and γsubunits). The central stalk is linked
to the F0 complex, which consists of a lipid-filled
c-ring (integral csubunits) and an α subunit. The pe-
ripheral stalk consists of OSCP, F6, b and d subunits.
ATP synthase forms dimer rows in the inner mito-
chondrial membrane, with the key proteins being the
e, f, g, A6L, j and k subunits. These dimer rows form
the cristae structure of the inner membrane, giving
it a positive curvature [50, 51].
The possibility of ATP synthase involvement in
the formation of the mitochondrial pore was first
suggested with the discovery of a CsA-sensitive inter-
action between cyclophilin  D and OSCP (Fig.  2)  [28].
This interaction resulted in inhibition of mitochon-
drial ATP synthase activity and stimulation of MPT
pore opening. Benzodiazepine-423, a classic inhibi-
tor of ATP synthase activity, acted in a similar man-
ner, binding to OSCP at the same site as cyclophil-
in  D [23, 28].
There are currently two hypotheses on ATP syn-
thase involvement in MPT pore formation: the pore is
formed either between ATP synthase dimers or with-
in the ring of c subunits [3].
When ATP synthase was embedded in an artifi-
cial lipid membrane, a high Ca
2+
-dependent channel
conductance (1-1.3  nS) was observed, similar to the
conductance of the MPT pore in mitoplasts [23,  52].
The conductance was observed only in the case of
the dimeric or oligomeric form of ATP synthase, but
not its monomeric form [53]. Suppression of the ex-
pression of proteins involved in the dimer formation
(subunits e, g, f) led to complete inhibition of MPT
pore opening [54-56]. Dimerization of ATP synthase
was enhanced by the ATPase inhibitor, the IF1 pro-
tein, which inhibits MPT pore opening and protects
the cell from ischemic damage [57]. All this allowed
us to assume that the MPT pore is formed in the di-
mers of ATP synthases between the e, g and f sub-
units due to their destabilization or structural rear-
rangements in each monomer [58].
On the other hand, it was shown that ATP syn-
thase monomers are also capable of forming high-con-
ductance channels [59]. When purified c  subunits
were embedded in a lipid membrane, voltage-gated
channels of varying conductance appeared in the
membrane. The conductance of most channels was
MPT PORE: STRUCTURE AND FUNCTIONS 1795
BIOCHEMISTRY (Moscow) Vol. 90 No. 12 2025
at a level of 100  pS, but in some cases, high conduc-
tance at a level of 1.5-2  nS was also observed [60],
which is also similar to the conductance of the MPT
pore [52]. Knockdown of the c subunit of ATP syn-
thase resulted in inhibition of MPT pore opening in
mitochondria [61].
However, the possibility of the c-ring involve-
ment in MPT pore formation remains debatable. This
is due to the fact that the inner space of the ring is
filled with lipid molecules and is hydrophobic [62].
The “death fingers” hypothesis attempts to circumvent
this limitation and combine the two mechanisms de-
scribed above [63]. According to this hypothesis, the
binding of Ca
2+
to the β  subunit of the F1 complex
of ATP synthase and the binding of cyclophilin  D to
OSCP causes a chain of conformational changes in the
F
1
complex, which are transmitted through the pe-
ripheral stalk to the inner mitochondrial membrane
in the region of the esubunit. Simultaneous displace-
ment of the central stalk subunits and the F
1
complex
away from the c-ring can also occur. The e subunit
expels lipids from the c-ring lumen, which may lead
to its expansion and the emergence of channel activ-
ity. Indirect evidence for such a mechanism may be
provided by the data showing that mutations weak-
ening the c-ring packing led to an increase in the
internal diameter of the ring, an increase in chan-
nel conductance and an increase in the sensitivity of
various cells to cell death inducers [60,  64]. This hy-
pothesis describes both the reversible opening of the
pore, which can be observed under physiological and
sublethal pathological conditions, and the irreversible
opening of the pore. In the latter case, dissociation
of the F
1
and F
0
subunits may occur, which will lead
to mitochondrial swelling and cell death. However,
it should be noted that displacement of lipids from
the c-ring lumen into the hydrophilic region of the
intermembrane space is a thermodynamically unfa-
vorable process [3].
J.  Walker’s group demonstrated that knocking out
various ATP synthase proteins involved in MPT pore
formation does not suppress its activity. For exam-
ple, genetic knockout of each of the e, f, g, ksubunits
and of the 6.8  kDa proteolipid (6.8PL) disrupted the
dimerization of the complex, but the probability of
MPT pore opening did not decrease[58]. Knockout of
three nuclear genes, ATP5G1, ATP5G2, and ATP5G3,
encoding the c subunits, rendered mitochondria in
HAP1-A12 cells sensitive to MPT pore induction  [65].
Knocking down peripheral stalk proteins (β  subunits
and OSCP) either did not affect the induction of the
mitochondrial pore in cells, or ionic currents were
observed that were insensitive to cyclosporin  A, but
underwent inhibition by bongkrekic acid[54, 55]. All
this allowed us to consider ANT as a channel of the
MPT pore once again.
Recently, a consensus has emerged that adding
Ca
2+
to mitochondria can activate the formation of
the MPT pore, whose channel in the inner membrane
can be either ANT or ATP synthase [3, 66, 67]. It has
been suggested that the opening of the pore associated
with ATP synthase occurs at physiological pH values,
while the action of ANT regulators (and, consequent-
ly, ANT-mediated pore opening) is enhanced by de-
creasing the pH from 7.4 to 6.5 [68]. In this case, the
two structures can function as an ATP synthasome.
ANT and ATP synthase embedded in an artificial lipid
membrane are able to induce channel conductivity
upon the addition of high concentrations of Ca
2+
ions.
However, the precise molecular structure of the
mitochondrial pore remains far from being resolved.
This is due to the numerous contradictions and phe-
nomena which are difficult to explain observed in
experiments with mitochondria. This has been men-
tioned above in regards to the studies when suppres-
sion of the activity of the same genes encoding ATP
synthase proteins led to different effects in different
experiments. Meanwhile, the removal of ANT or var-
ious ATP synthase subunits leads to a decrease in
energy metabolism, suppression of respiratory activ-
ity and membrane potential generation [55, 65]. This
may be an additional factor influencing pore for-
mation in mitochondria even in the absence of cer-
tain subunits. In addition to these discrepancies, no
precise answers to questions posed several decades
ago are present yet. For example, it is still unclear
why pore induction in mitoplasts and artificial mem-
branes requires Ca
2+
ion amounts 2-3 orders of mag-
nitude higher than in the case of intact mitochondria.
Moreover, it is still not entirely clear what the bind-
ing site for Ca
2+
ions is [3]. Another mystery is the
ratio of the number of opening pores (up to 10) to
the number of ANT and ATP synthases (several thou-
sand copies) per mitochondrion [69, 70]. Which par-
ticular events lead to a very small number of ANT
or ATP synthases undergoing transformation and
changing their physiological function to pathological?
The answers to these questions may be one of the
cornerstones which will allow us to understand the
structure of the pore in the future.
The accessory proteins required for MPT pore
induction in mitochondria. The ability of mitochon-
drial outer membrane proteins to regulate MPT pore
opening has been known for a long time. VDAC is
a family of β-barrel proteins of the mitochondrial
outer membrane (~30-35  kDa). These are the most
abundant outer membrane proteins that regulate the
transport of metabolites and ions from cytoplasm to
mitochondria. VDAC has long been considered one of
the main components of the MPT pore, since the con-
ductance of VDAC channels is similar to that of the
MPT pore (Fig.  2) [71]. It was believed that the pore
BELOSLUDTSEV et al.1796
BIOCHEMISTRY (Moscow) Vol. 90 No. 12 2025
is formed in the contact site region where the
connection of outer and inner mitochondrial mem-
branes occurs. However, in isolated mitochondria,
VDAC is not required for MPT pore formation [5].
Thus, mitochondrial pore opening can be induced in
mitoplasts and submitochondrial particles [72,  73].
Furthermore, pore opening occurs in mitochondria
isolated from mice in which all three VDAC isoforms
were knocked out [74]. However, the role of VDAC
in MPT pore formation in a living cell should not
be underestimated, since these channels provide the
main pathway for Ca
2+
ions influx into mitochon-
dria. Various cytoplasmic protein kinases (e.g., GSK3β,
PKA (protein kinase  A), etc.) lower the threshold for
MPT pore opening to various inducers by phosphor-
ylating VDAC [71].
TSPO is another protein (18  kDa) localized in the
outer mitochondrial membrane[75], for which partic-
ipation in MPT pore formation has been described.
TSPO is involved in the regulation of many meta-
bolic processes, the key one of which is cholesterol
transport into mitochondria [76,  77]. It was believed
that this protein facilitates the formation of the con-
tact site between VDAC and ANT [77]. Meanwhile, in
rat brain mitochondria, cholesterol binding to TSPO
resulted in suppression of cyclosporin A-insensitive
membrane potential decrease and Ca
2+
release from
mitochondria [78]. Antibodies against TSPO caused
an increase in Ca
2+
capacity of mitochondria [78,  79].
A number of TSPO ligands caused MPT pore activa-
tion [76]. At the same time, there is evidence that
suppression of TSPO expression in animal tissues did
not cause a significant change in MPT pore induc-
tion [79]. Therefore, the role of TSPO in MPT pore
formation cannot be considered proven yet, and the
stimulating effects of ligands may be associated with
the presence of other mitochondrial targets in the
latter [23].
The involvement of other proteins of the out-
er mitochondrial membrane (in particular, proteins
of the Bcl2 family) in MPT pore regulation has also
been shown [5]. It has been suggested that the Bax
and Bid proteins stimulate the pore opening in mi-
tochondria, since their genetic knockout leads to in-
hibition of MPT-induced mitochondrial swelling and
cell death[80]. However, it should be noted that these
proteins induce the formation of large pores in the
outer mitochondrial membrane primarily as a result
of oligomerization with each other or with VDAC[71].
This can surely promote MPT pore opening, but
it rather appears to be a parallel process.
As mentioned above, in the mid-2000s, a phos-
phate transporter capable of interacting with cyclo-
philin D was considered as a possible structural unit
of the MPT pore in the inner membrane [22]. Today,
it is believed that even if the phosphate transporter is
involved in pore formation, its expression level does
not limit the process of MPT pore opening[81]. It can
be assumed that, being part of the ATP synthasome,
this transporter may be indirectly involved in pore
induction mediated by ANT or ATP synthase[2]. Other
proteins (aldehyde dehydrogenase (Aldh6a1 gene)
and prohibitin 2) associated with ATP synthase can
also modulate ATP synthase-mediated MPT pore [82].
A 2015 study showed that, in addition to cyclo-
philin  D, spastic paraplegia 7 (SPG7) is also an es-
sential component [83]. This is a protein of the AAA
protease family, localized in the inner mitochondri-
al membrane. Genetic knockout of SPG7 resulted in
suppression of MPT pore opening in mitochondria
induced by Ca
2+
or oxidative stress and also prevent-
ed cell death. There are several possible explanations
for SPG7 involvement in MPT pore induction, rang-
ing from its effect on mitochondrial Ca
2+
transport
to suppression of SIRT3 (sirtuin  3) expression and, a
consequently, suppression of cyclophilin  D deacetyla-
tion [84, 85]. Other studies found that increasing or
decreasing SPG7 expression by using siRNA did not
affect MPT pore induction in mitochondria [86]. All
these facts allow us to say that SPG7 may indirectly
participate in the induction of the MPT pore, but it is
not an essential component of the pore.
Another group of enzymes involved in MPT pore
regulation are various kinases, including mitochon-
drial creatine kinase, hexokinase, and GSK-3β[87-89].
Their ability to regulate the pore is believed to be
associated primarily with their interaction with VDAC
and ANT. Thus, increased expression of creatine ki-
nase and hexokinase suppresses MPT pore opening
and prevents the development of cell death  [49].
Due to this, one of the cancer cell markers is over-
expression of mitochondria-associated hexokinases  I
and II [90]. Kinases can interact with VDAC either
directly (the hydrophobic N-terminus of hexokinase
interacts with Glu73 of VDAC, preventing its oligo-
merization) or through Ser and Thr residue phos-
phorylation (inhibition or phosphorylation of GSK-3β
prevents mitochondrial ROS production, MPT pore
induction and cell death) [71, 89].
Alternative mechanisms of mitochondrial pore
induction which are insensitive to cyclosporin  A.
The possibility of inhibiting mitochondrial pore for-
mation by cyclosporinA allowed researchers to focus
their attention on the MPT pore phenomenon. Thus,
studies of the role of the mitochondrial pore in var-
ious diseases are often determined by the effect of
CsA or other MPT inhibitors on the process. Howev-
er, it is possible to induce non-specific mitochondrial
permeability that is insensitive to cyclosporin  A. This
suggests that regulation of such permeability is not
associated with cyclophilin  D. Exogenous amphipathic
peptides (e.g., signaling proteins), certain prooxidants,
MPT PORE: STRUCTURE AND FUNCTIONS 1797
BIOCHEMISTRY (Moscow) Vol. 90 No. 12 2025
hormones (thyroxine) and fatty acids have the ability
to induce Ca
2+
-dependent permeability of the inner
mitochondrial membrane that is insensitive to cyclo-
sporin [12, 91].
The possibility of inducing a Ca
2+
-dependent pore
mediated by fatty acids and phospholipase A
2
activ-
ity was established quite a long time ago [11-13, 92,
93]. The insensitivity of this phenomenon to CsA was
demonstrated in the early 2000s: saturated palmitic
acid in the presence of Ca
2+
ions induced swelling
of mitochondria isolated from various tissues. At the
same time, it was shown that palmitic acid similar-
ly induces Ca
2+
-dependent permeabilization of both
natural (mitochondrial and erythrocyte plasma mem-
branes) and artificial lipid membranes (unilamellar li-
posomes and bilayer lipid membranes)[13]. Based on
these and a number of other characteristics, we con-
cluded that the pore induced by saturated fatty acids
and Ca
2+
is of lipid origin[93]. It was shown that the
mechanism of palmitate/Ca
2+
-induced pore formation
is based on the ability of saturated fatty acid anions
to form strong complexes with Ca
2+
in the lipid bilay-
er, followed by their separation into solid crystalline
membrane domains and the appearance of hydro-
philic lipid pores[12,93]. Importantly, lipid pores are
characterized by their self-sealing ability, which may
be of physiological significance for the mechanism of
mitochondrial Ca
2+
unloading [13]. The formation of
such pores in a living cell can occur due to phos-
pholipase  A
2
activation [13,  94]. More details on the
lipid pore induced by palmitic acid and Ca
2+
can be
found in the review by Mironova and Pavlov [13].
MPT PORE INHIBITORS
IN MITOCHONDRIA
Understanding the mechanisms of MPT pore ac-
tivation and its involvement in pathological processes
naturally leads us to the question of the possibilities
of modulating this pore’s function. One of the most
detailed analyses of MPT pore modulators was giv-
en in the review by Zoratti and Szabo [1] in 1995.
Today, the development of MPT pore inhibitors has
entered the phase of targeted design: using structural
modeling, high-throughput screening and analysis of
interactions with both the regulators of its opening
(cyclophilin  D, TSPO) and the discussed direct com-
ponents of the pore (such as ANT and ATP synthase).
The most studied MPT pore inhibitor is cyclo-
sporin  A [17-19]. Its discovery not only confirmed
the role of the MPT pore in cell death but also pro-
vided a direction for further research. However, the
use of CsA as a cardio- or neuroprotector is limit-
ed by its side effects, including immunosuppression,
nephrotoxicity and its lack of selectivity regarding
other cyclophilins. Furthermore, its low permeabili-
ty across the blood-brain barrier limits its potential
use against neurological diseases [95]. Other natural
cyclosporins B, C, D, and E have also been studied
for their ability to modulate MPT pore opening. They
differ by minor variations in amino acid sequences,
which, however, have a significant impact on their
activity. For example, cyclosporins A, B, C, and D ex-
hibit significant conformational dynamics, whereas
cyclosporin  E, being more rigid due to the absence
of a methyl group at Val11 position, loses the ability
to inhibit MPT pore opening [96].
One of the ways to enhance the selective prop-
erties of cyclosporin A was elimination of immu-
nosuppressive activity. Thus, in 1994, a cyclosporin
derivative, N-methyl-4-isoleucine-cyclosporin (NIM811
or GNX-4975)  [1,  97], was obtained. It retained high
affinity for cyclophilin  D but did not suppress cal-
cineurin, which deprived it of immunomodulatory
activity and could potentially expand its therapeutic
window. This allowed NIM811 to reach the stage of
preclinical and early clinical trials against myocar-
dial infarction and stroke. Another promising com-
pound lacking any immunosuppressive properties
is alisporivir (Debio 025), which has shown higher
selectivity for cyclophilin  D, having successfully per-
formed in models of diabetes, Duchenne muscular
dystrophy [98,  99], collagenopathy [100], Alzheimers
disease [101] and having passed clinical trials for
hepatitis  C. However, like cyclosporin  A, alisporivir
exhibited non-selective effects on mitochondrial bio-
energetics (already at micromolar concentrations,
which are classical for in  vitro experiments), which
is due to its hydrophobic nature and the ability to
accumulate in the lipid phase of membranes and af-
fect their physical properties [102].
Cyclophilin  D is not the only regulatory molecule
through which MPT pore status is modulated. The
involvement of TSPO in pore opening and its modu-
lation by the synthetic agent TRO40303, which is ca-
pable of preventing pathological opening of the MPT
pore, is being discussed [103].
In addition to cyclosporins, the natural macrolide
compound sanglifehrin A, isolated from actinomy-
cetes, is among the classic MPT pore inhibitors. San-
glifehrin  A is capable of binding to cyclophilin  D, but
at a different site than cyclosporin  A, and does not
inhibit calcineurin. Furthermore, unlike CsA, sangli-
fehrin  A does not affect the binding of cyclophilin  D
to ANT [104]. In experiments on cardiac tissue, san-
glifehrinA reduced myocardial damage during reper-
fusion, decreasing infarction size and protecting cells
from oxidative stress if the compound was adminis-
tered during the first minutes of reperfusion [105].
However, like CsA, sanglifehrin  A also has immuno-
suppressive properties, which limits its use. Another
BELOSLUDTSEV et al.1798
BIOCHEMISTRY (Moscow) Vol. 90 No. 12 2025
example is bongkrekic acid, produced by Burkholde-
ria gladioli pathovar cocovenenans bacteria. It blocks
MPT pore opening binding to ANT. However, its high
toxicity limits its clinical applications.
In addition to well-known ANT modulators ca-
pable of inhibiting MPT pore opening, agents capa-
ble of influencing the pore-forming activity of ATP
synthase are currently being developed. For exam-
ple, a new class of MPT pore inhibitors based on
1,4,8-triazaspiro[4.5]decan-2-one has recently been
presented [106]. The most promising compound, 14e,
demonstrated high inhibitory activity at micromolar
concentrations and a pronounced cytoprotective ef-
fect in a model of cardiomyocyte hypoxia/reoxygen-
ation. The effect is apparently due to the influence of
the compounds on ATP synthase which is a structural
component of the MPT pore. Molecular modeling re-
vealed a potential binding site for the compounds at
the junction of the c-ring and the a subunit of ATP
synthase, and in this case, the structural features of
14e provided the greatest affinity for this site [106].
High-throughput screening also identified efficient
MPT pore inhibitors among cinnamyl anilides, isoxaz-
oles and benzamides [107-110]. These compounds are
more potent than cyclosporin  A and exhibit marked
specificity, acting independently of cyclophilin  D.
However, these compounds are characterized by low
metabolic stability (e.g., isoxazoles) and, in some cas-
es (benzamides), toxicity, meaning that further opti-
mization of their structure is required.
It should be noted that pore inhibitors are be-
ing actively developed for at least two main rea-
sons: the emergence of new scientific hypotheses
about the pore structure and the identification of the
role of the mitochondrial pore in the development
of pathologies.
THE ROLE OF MPT PORES
IN THE DEVELOPMENT
OF CELLULAR PATHOLOGIES
The first papers suggesting the involvement of
the MPT pore in the development of cellular pathol-
ogies appeared in the late 1970s, when it was discov-
ered that a sharp increase in the permeability of the
inner mitochondrial membrane can be triggered by
Ca
2+
overload and oxidative stress and is closely as-
sociated with cell death. At that time, it was already
shown that MPT pore opening leads to mitochondrial
swelling, rupture of outer mitochondrial membrane
and activation of apoptosis or necrosis, which was
believed to be associated with tissue damage during
cardiac ischemia/reperfusion and other acute condi-
tions. In the following decades, research confirmed
the key role of the MPT pore in the pathogenesis of
awide range of diseases, including neurodegenerative
and neuromuscular diseases, cardiovascular diseases,
diabetes, bone remodeling pathologies and cancer
(Fig. 3). Although the molecular structure of the pore
is still under debate, its involvement in pathologies is
widely accepted, and modulation of its activity is con-
sidered a promising therapeutic strategy [111]. More-
over, accumulated data suggest that the occurrence
of pathologies associated with the process of aging,
primarily neurodegenerative disorders, metabolic and
cardiovascular pathologies, is also due to an age-relat-
ed increase in cell sensitivity to MPT pore induction.
Indeed, indirect measurements show that the MPT
pore is activated more efficiently in tissues of old
organisms, especially when exposed to high concen-
trations of Ca
2+
and inorganic phosphate ions [112].
In addition, experiments on model organisms such as
Caenorhabditis elegans demonstrate that stimulation
of MPT pore induction shortens lifespan, while its in-
hibition can have a protective effect [113]. However,
the mechanisms that determine the transition from
the physiological to the pathological role of the MPT
pore remain not fully understood.
Participation of the MPT pore in the pathogen-
esis of neurodegenerative diseases. The MPT pore
plays a key role in the development of neurodegen-
erative diseases such as Alzheimer’s and Parkinson’s
diseases, amyotrophic lateral sclerosis (ALS), as well
as in the processes of axonal degeneration and neu-
ronal death. It is believed that MPT pore induction in
Alzheimers and Parkinson’s diseases is caused by ac-
cumulation of aggregates of pathological proteins in
nerve cells, such as β-amyloid, tau protein and α-sy-
nuclein, which are able to interact with MPT pore
components (e.g., cyclophilin D, VDAC, ANT, and ATP
synthase) [7, 114-116]. Experimental models of ALS
(G93A-mSOD1 mice) revealed structural rearrange-
ments of motor neuron mitochondria, accompanied
by an increase in the number of contacts between the
inner and outer membranes, which is believed to con-
tribute to the induction of the MPT pore [116]. Since
such mitochondria cease to produce ATP efficiently,
cells with high energy demands (e.g., dopaminergic
neurons in Parkinson’s disease or motor neurons in
ALS) begin to experience energy deficiency, which
leads to their degeneration. Furthermore, mitochon-
drial dysfunction promotes increased ROS formation
and development of oxidative stress. Opening of the
MPT pore promotes the release of cytochrome  c and
other proinflammatory molecules, such as mitochon-
drial DNA, which increases neuroinflammation and
further contributes to nerve tissue damage [7]. More-
over, mitochondrial sensitivity to MPT pore opening
increases with age. This is associated with impaired
calcium homeostasis, increased oxidative stress and
changes in the composition of mitochondrial proteins,
MPT PORE: STRUCTURE AND FUNCTIONS 1799
BIOCHEMISTRY (Moscow) Vol. 90 No. 12 2025
Fig. 3. A scheme illustrating MPT pore participation in the development of various pathological processes. Explanations are
given in the text. ANT, adenine nucleotide translocator; VDAC, voltage-dependent anion channel of the outer mitochondrial
membrane. The image was created by using BioRender.
which may explain the age-related vulnerability of the
brain to neurodegeneration [7]. These effects can be
eliminated by pharmacological inhibition of the MPT
pore with cyclosporin A [117] and olesoxime [118].
Participation of the MPT pore in the devel-
opment of pathologies caused by tissue ischemia/
reperfusion. The role of the MPT pore in damage
caused in tissues highly sensitive to hypoxia and subse-
quent reoxygenation (heart, brain, kidneys) is current-
ly being actively discussed [3,  4]. Specifically, during
myocardial infarction, which occurs due to ischemic
damage to the heart muscle, Ca
2+
ions and inorgan-
ic phosphate accumulate in cardiomyocytes, creating
preconditions for MPT pore induction in mitochon-
dria. However, in the initial period of pathology de-
velopment, low intracellular pH and high ADP levels
temporarily suppress pore opening, delaying the on-
set of consequences. The situation changes drastical-
ly under reperfusion conditions, when restoration of
blood flow leads to normalization of pH values and
the “washout” of inhibitory factors, creating favor-
able conditions for MPT pore formation. It is also
important to note that, in addition to the classical
Ca
2+
-dependent mechanism, oxidative stress plays a
significant role in regulating MPT pore formation in
mitochondria in cardiovascular pathologies. For ex-
ample, accumulation of intracellular succinate during
ischemia can lead to increased ROS formation as a
result of reverse electron transfer in the mitochondri-
al respiratory chain during subsequent reperfusion,
which further facilitates MPT pore opening in organ-
elles [4]. Subsequently, pore induction under such
conditions causes a collapse of mitochondrial mem-
brane potential, swelling and destruction of mito-
chondria, which triggers a cascade of events leading
to necrosis of cardiomyocytes and other types of cells
in the myocardium. This process is directly related to
the localization and volume of the infarction zone:
the more mitochondria undergo MPT pore induction,
the larger the area of necrotic tissue. Experimental
models convincingly demonstrate that preventing
MPT pore formation with cyclosporin  A, using cyclo-
philin  D gene knockout methods or initiating the isch-
emic preconditioning effect significantly reduce tis-
sue damage during ischemia/reperfusion [119, 120].
However, clinical studies based on pharmacological
BELOSLUDTSEV et al.1800
BIOCHEMISTRY (Moscow) Vol. 90 No. 12 2025
inhibition of the MPT pore in patients with myocardi-
al infarction have shown conflicting results [121], in-
dicating the need to optimize therapeutic approaches.
MPT pore involvement in neuromuscular dis-
eases. The idea of mitochondrial dysfunction being
involved in the pathogenesis of neuromuscular dis-
eases was proposed for the first time almost 50years
ago [122]. Currently, the main focus is on how MPT
pore induction caused by Ca
2+
overload and oxida-
tive stress in dysfunctional mitochondria leads to
muscle fiber damage and death. It has been found
that the MPT pore may play an important role in
the development of muscular dystrophies associated
with deficiency of collagen  VI (e.g., Bethlem myopa-
thy, Ullrich congenital muscular dystrophy, myoscle-
rosis), dystrophin (Duchenne and Becker muscular
dystrophy), δ-sarcoglycan (limb-girdle muscular dys-
trophy), and laminin [123, 124]. In all these cases, the
absence of structural proteins leads to a disruption of
the stability of muscle fiber sarcolemma (both its in-
tegrity and the function of ion channels and signaling
molecules) and an excessive influx of calcium ions
from the extracellular space [125]. Excessive calcium
uptake by mitochondria under these conditions com-
bined with oxidative stress makes them extremely
sensitive to MPT pore opening and triggers a cascade
of pathological events, from the loss of mitochondri-
al membrane potential to the release of proapoptot-
ic factors and, ultimately, muscle fiber death [46].
In addition, mitochondria overloaded with calci-
um phosphate can act as foci of tissue calcification,
thereby promoting tissue degeneration [126]. It is in-
teresting to note that mitochondria in the hearts of
dystrophic mdx mice (a model of Duchenne muscular
dystrophy) exhibit resistance to MPT pore induction,
which may be linked to tissue-specific expression of
proteins associated with its formation and regulation
(cyclophilin  D, ANT, ATP synthase subunits)[127]. This
is supposed to contribute to the delayed development
of cardiac pathologies, which is characteristic of both
model animals and patients. Meanwhile, MPT pore in-
hibitors (CsA, alisporivir, NIM) are able to slow the
development of muscle pathology in model animals
and also improve the condition of skeletal muscles in
patients[98, 99, 110,124]. A similar effect is achieved
by blocking mitochondrial calcium overload with in-
hibitors of Ca
2+
-transporting proteins[125]. A number
of pharmacological inhibitors are currently in early
clinical trials. Recent studies involving inactivation of
genes of cyclophilin  D and ANT, the presumed pro-
tein components of the pore, also confirm the role of
the MPT pore in the development of muscle pathol-
ogies [124, 128].
It has been shown that the MPT pore may be
involved in pathogenesis of multisystem diseases, in-
cluding those affecting muscle tissue. Early stages of
ALS are characterized by disrupted calcium homeosta-
sis and oxidative stress in the brain, spinal cord, and
skeletal muscles, which contributes to increased sus-
ceptibility of mitochondria in the cells of these tissues
to MPT pore opening [129]. The involvement of the
MPT pore formation in age-related muscle degenera-
tion has also been described [130]. The lack of effec-
tive therapy for these pathologies, which are in most
cases sporadic and multifactorial, allows us to consid-
er the MPT pore as one of the promising pharmaco-
logical targets. Further research in this area may also
contribute to improving the diagnostic approaches to
most neuromuscular pathologies accompanied by mi-
tochondrial dysfunction already in the early stages.
The role of the MPT pore in the development
of diabetes mellitus. In type  1 and type  2 diabetes
mellitus, hyperglycemia, hyperlipidemia, and oxida-
tive stress increase mitochondrial sensitivity to MPT
pore opening in most tissues, which leads to a loss
of membrane potential, increased ROS production
and the release of proapoptotic factors [131]. Patho-
logical induction of the MPT pore in diabetes has
been described for pancreatic β-cells [132], cardio-
myocytes [133] and neurons [134]. The involvement
of this phenomenon in diabetic complications, such
as cardiomyopathy, is also being discussed. Mean-
while, sensitivity to MPT pore opening in diabetes
varies depending on the tissue type. For example,
mitochondria of the heart and skeletal muscles be-
come more susceptible to the opening of the pore in
diabetes, which increases oxidative stress and cell
damage [135, 136]. At the same time, an increase in
resistance to MPT pore induction is observed in the
liver, which may also be associated with tissue-spe-
cific expression of pore proteins and is considered
an adaptive mechanism which prevents acute toxic
organ damage [137]. Interestingly, this significantly
distinguishes the pathology of diabetes from other
metabolic diseases, such as non-alcoholic fatty liv-
er disease, which is characterized by accumulation
of free fatty acids in hepatocytes and the develop-
ment of lipotoxicity, which contributes to the disrup-
tion of the respiratory chain, increased ROS forma-
tion and a significant increase in the sensitivity of
liver cells to MPT pore induction [138]. It has been
shown that both genetic and pharmacological inhibi-
tion of the MPT pore by using cyclophilinD blockers
(CsA and alisporivir) can protect against mitochon-
drial damage caused by diabetes or other metabol-
ic pathologies, improve glucose utilization and re-
duce oxidative stress, thereby alleviating cardiac
and skeletal muscle dysfunction [131, 133, 139-141].
We have shown that blockers of VDAC (VBIT-4) and
ANT(bongkrekic acid), potential channel components
of the MPT pore, exhibit a cytoprotective effect under
these conditions [48, 142].
MPT PORE: STRUCTURE AND FUNCTIONS 1801
BIOCHEMISTRY (Moscow) Vol. 90 No. 12 2025
Fig. 4. Number of publications by year in the PubMed database in response to the query “Mitochondrial Permeability
Transition”. https://pubmed.ncbi.nlm.nih.gov/?term=mitochondrial+permeability+transition&sort=date.
It should be noted that diabetes mellitus is a
chronic disease. In this regard, mitochondrial pore in-
duction in a high-conductivity state may be a feature
of such extremely pathological manifestations as dia-
betic foot syndrome. In all other cases, however, MPT
pore activation occurs in a low-conductivity “flicker-
ing” state which rather represents a mechanism of
adaptation to mitochondrial calcium overload.
MPT pore participation in bone remodeling
pathologies. Recent data indicate that the MPT pore
is involved in the regulation of bone cell metabolism,
including mesenchymal stem cells, osteoblasts and os-
teoclasts [143]. In mesenchymal stem cells, decreased
MPT pore opening activity promotes osteogenic dif-
ferentiation, which is associated with the suppression
of cyclophilin  D expression via the BMP/Smad signal-
ing pathway [144].
Pathological opening of the MPT pore is observed
in age-related osteoporosis, which is accompanied by
a decrease in bone mass and impaired mitochondri-
al function. Knockdown of cyclophilin  D or its phar-
macological inhibition (e.g., by using NIM811) im-
proves bone health in aged mice, preventing loss of
bone mass [145]. Osteoporosis induced by excessive
glucocorticoid (e.g., dexamethasone) intake has also
been shown to be associated with MPT pore activa-
tion, which promotes osteoblast apoptosis. This effect,
observed in gum tissue, is believed to be due to an
increase in cyclophilin  D levels in tissues and is re-
moved by cyclosporin  A or knockdown of this pore
protein [146].
In models of periodontitis and osteoarthritis, ex-
cessive induction of the MPT pore facilitates activa-
tion of inflammatory pathways and bone resorption.
Meanwhile, MPT pore inhibition reduces the intensity
of inflammation and bone tissue loss [147].
MPT pore and oncological diseases. Unlike
other pathologies discussed above, in which the MPT
pore acts as an unambiguous driver of cell, tissue,
and organ damage, its activity in carcinogenesis is
subject to complex regulation, reflecting tumor cell
adaptation to stress. The Warburg effect, which is
characteristic of many tumors and involves increased
glycolysis in cells even in the presence of oxygen,
creates a unique metabolic background (cytoplasmic
acidosis and decreased inorganic phosphate levels)
that helps to block MPT pore induction [148]. Fur-
thermore, oncogenic signaling pathways (e.g., Bcl-2
family proteins, Akt or mutant p53) actively suppress
the pore’s sensitivity to calcium and oxidative stress.
This allows malignant cells to maintain viability even
under conditions that would inevitably lead to mito-
chondria-mediated death in normal tissues [4].
Expression of many MPT pore components is sig-
nificantly increased in tumor cells[71]. It is suggested
that this may provide cancer cells with the capabil-
ity of fine regulation, ranging from complete sup-
pression of MPT pore formation (for cell survival) to
its controlled activation (for adaptation to changing
microenvironmental conditions). This may underlie
the heterogeneity of many tumor types and explain
their resistance to therapy. Nevertheless, a number
of approaches involving the use of oxidative stress
inducers and calcium homeostasis modulators may
be used to combat tumors, provided that the issue of
their selectivity is addressed [4].
Is it possible to accurately decipher the structure
of the MPT pore, determine the molecular mechanism
of its formation and visualize this phenomenon in a
living cell? It should be acknowledged that the pros-
pects, given current capabilities and experimental ap-
proaches, as well as the known limitations (including
BELOSLUDTSEV et al.1802
BIOCHEMISTRY (Moscow) Vol. 90 No. 12 2025
the rarity of this event in mitochondria), are not very
promising. This is indirectly evidenced by the trend in
publications: despite the recognized role of the MPT
pore in the development of cellular pathologies and
socially significant diseases, as well as active research
in this area, the number of articles has been grad-
ually decreasing since 2015 (Fig.  4). The emergence
of new methodological approaches and selective in-
hibitors could not only allow to establish the precise
structure of the pore complex but also to provide a
new direction for therapy and diagnosing various cel-
lular pathologies associated with MPT pore induction
and associated organelle dysfunction.
Abbreviations
ANT adenine nucleotide translocator
CsA cyclosporinA
MPT
mitochondrial permeability transition
pore
OSCP
oligomycin sensitivity conferring pro-
tein
ROS reactive oxygen species
SPG7 spastic paraplegia 7
TSPO peripheral benzodiazepine receptor
VDAC voltage-dependent anion channel
Contributions
K. N. Belosludtsev – concept; N. V. Belosludtseva,
M. V. Dubinin, and K. N. Belosludtsev – writing the
manuscript; N. V. Belosludtseva and K. N. Beloslud-
tsev – editing the manuscript; M. V. Dubinin and
K. N. Belosludtsev– creating the figures.
Funding
The work was carried out with the support of the Rus-
sian Science Foundation (grant no.25-65-00005).
Ethics approval and consent to participate
This work does not contain any studies involving
human and animal subjects.
Conflict of interest
The authors of this work declare that they have
noconflicts of interest.
REFERENCES
1. Zoratti,M., and Szabo,I. (1995) Mitochondrial perme-
ability transition, Biochim. Biophys. Acta, 1241, 139-
176, https://doi.org/10.1016/0304-4157(95)00003-A.
2. Frigo, E., Tommasin, L., Lippe, G., Carraro, M., and
Bernardi, P. (2023) The haves and have-nots: the
mitochondrial permeability transition pore across
species, Cells, 12, 1409, https://doi.org/10.3390/
cells12101409.
3. Bernardi, P., Gerle, C., Halestrap, A. P., Jonas, E. A.,
Karch,J., Mnatsakanyan,N., Pavlov,E., Sheu, S.S., and
Soukas, A. A. (2023) Identity, structure, and function
of the mitochondrial permeability transition pore:
controversies, consensus, recent advances, and future
directions, Cell Death Differ., 30, 1869-1885, https://
doi.org/10.1038/s41418-023-01187-0.
4. Bonora,M., Patergnani,S., Ramaccini,D., Morciano,G.,
Pedriali, G., Kahsay, A. E., Bouhamida, E., Giorgi, C.,
Wieckowski, M.R., and Pinton,P. (2020) Physiopathol-
ogy of the permeability transition pore: molecular
mechanisms in human pathology, Biomolecules, 10,
998, https://doi.org/10.3390/biom10070998.
5. Halestrap, A.P., and Richardson, A.P. (2015) Themi-
tochondrial permeability transition: a current per-
spective on its identity and role in ischaemia/reper-
fusion injury, J.Mol. Cell. Cardiol., 78, 129-141, https://
doi.org/10.1016/j.yjmcc.2014.08.018.
6. Carraro, M., and Bernardi, P. (2023) The mitochon-
drial permeability transition pore in Ca
2+
homeosta-
sis, Cell Calcium, 111, 102719, https://doi.org/10.1016/
j.ceca.2023.102719.
7. Baev, A.Y., Vinokurov, A.Y., Potapova, E. V., Dunaev,
A.V., Angelova, P.R., and Abramov, A.Y. (2024) Mito-
chondrial permeability transition, cell death and neu-
rodegeneration, Cells, 13, 648, https://doi.org/10.3390/
cells13070648.
8. Raaflaub, J. (1953) Swelling of isolated mitochondria
of the liver and their susceptibility to physicochem-
ical influences, Helv. Physiol. Pharmacol. Acta, 11,
142-156.
9. Lehninger, A. L. (1959) Reversal of various types of
mitochondrial swelling by adenosine triphosphate,
J.Biol. Chem., 234, 2465-2471, https://doi.org/10.1016/
S0021-9258(18)69836-9.
10. Waite,M., Van Deenen, L., Ruigrok,T., and Elbers, P.
(1969) Relation of mitochondrial phospholipase A
activity to mitochondrial swelling, J.Lipid Res.,
10, 599-608, https://doi.org/10.1016/S0022-2275(20)
43055-X.
11. Broekemeier, K.M., Schmid, P. C., Schmid, H.H., and
Pfeiffer, D. R. (1985) Effects of phospholipase A2 in-
hibitors on ruthenium red-induced Ca
2+
release from
mitochondria, J. Biol. Chem., 260, 105-113, https://
doi.org/10.1016/S0021-9258(18)89700-9.
12. Belosludtsev, K. N., Dubinin, M. V., Belosludtseva,
N. V., and Mironova, G. D. (2019) Mitochondrial Ca
2+
transport: mechanisms, molecular structures, and
role in cells, Biochemistry (Moscow), 84, 593-607,
https://doi.org/10.1134/S0006297919060026.
13. Mironova, G.D., and Pavlov, E.V. (2021) Mitochondri-
al cyclosporine A-independent palmitate/Ca
2+
-induced
permeability transition pore (PA-mPT Pore) and its
role in mitochondrial function and protection against
calcium overload and glutamate toxicity, Cells, 10,
125, https://doi.org/10.3390/cells10010125.
MPT PORE: STRUCTURE AND FUNCTIONS 1803
BIOCHEMISTRY (Moscow) Vol. 90 No. 12 2025
14. Hunter, D.R., and Haworth, R. A. (1979) TheCa
2+
-in-
duced membrane transition in mitochondria.
I. The protective mechanisms, Arch. Biochem. Bio-
phys., 195, 453-459, https://doi.org/10.1016/0003-
9861(79)90371-0.
15. Haworth, R. A., and Hunter, D. R. (1979) TheCa
2+
-in-
duced membrane transition in mitochondria. II. Na-
ture of the Ca
2+
trigger site, Arch Biochem Biophys.,
195, 460-467, https://doi.org/10.1016/0003-9861(79)
90372-2.
16. Hunter, D.R., and Haworth, R. A. (1979) TheCa
2+
-in-
duced membrane transition in mitochondria. III.Tran-
sitional Ca
2+
release, Arch Biochem Biophys., 195,
468-477, https://doi.org/10.1016/0003-9861(79)90373-4.
17. Crompton, M., Ellinger, H., and Costi, A. (1988) Inhi-
bition by cyclosporin A of a Ca
2+
-dependent pore in
heart mitochondria activated by inorganic phosphate
and oxidative stress, Biochem.J., 255, 357-360.
18. Fournier,N., Ducet,G., and Crevat,A. (1987) Action of
cyclosporine on mitochondrial calcium fluxes, J.Bio-
energ. Biomembr., 19, 297-303, https://doi.org/10.1007/
BF00762419.
19. Coluccino,G., Muraca, V.P., Corazza,A., and Lippe,G.
(2023) Cyclophilin D in mitochondrial dysfunction:
a key player in neurodegeneration? Biomolecules,
13, 1265, https://doi.org/10.3390/biom13081265.
20. Halestrap, A. P., and Davidson, A. M. (1990) Inhibi-
tion of Ca
2+
-induced large-amplitude swelling of liver
and heart mitochondria by cyclosporin is probably
caused by the inhibitor binding to mitochondrial-ma-
trix peptidyl-prolyl cis-trans isomerase and prevent-
ing it interacting with the adenine nucleotide translo-
case, Biochem.J., 268, 153-160, https://doi.org/10.1042/
bj2680153.
21. Kokoszka, J., Waymire, K., Levy, S., Sligh, J., Gai, J.,
Jones, D., MacGregor, G., and Wallace, D. (2004)
The ADP/ATP translocator is not essential for the
mitochondrial permeability transition pore, Nature,
427, 461-465, https://doi.org/10.1038/nature02229.
22. Leung, A.W., Varanyuwatana,P., and Halestrap, A.P.
(2008) Themitochondrial phosphate carrier interacts
with cyclophilinD and may play a key role in the per-
meability transition, J.Biol. Chem., 283, 26312-26323,
https://doi.org/10.1074/jbc.M805235200.
23. Giorgio,V., von Stockum,S., Antoniel,M., Fabbro,A.,
Fogolari, F., Forte, M., Glick, G. D., Petronilli, V.,
Zoratti, M., Szabo, I., Lippe, G., and Bernardi, P.
(2013) Dimers of mitochondrial ATP synthase form
the permeability transition pore, Proc. Natl. Acad.
Sci. USA, 110, 5887-5892, https://doi.org/10.1073/pnas.
1217823110.
24. Mnatsakanyan,N., Beutner, G., Porter, G. A., Alavian,
K. N., and Jonas, E. A. (2017) Physiological roles of
the mitochondrial permeability transition pore, J.Bio-
energ. Biomembr., 49, 13-25, https://doi.org/10.1007/
s10863-016-9652-1.
25. Mithal, D. S., and Chandel, N. S. (2020) Mitochon-
drial dysfunction in fragile-X syndrome: plugging
the leak may save the ship, Mol Cell., 80, 381-383,
https://doi.org/10.1016/j.molcel.2020.10.002.
26. Galat, A. (1993) Peptidylproline cis-trans-isomeras-
es: immunophilins, Eur. J. Biochem., 216, 689-707,
https://doi.org/10.1111/j.1432-1033.1993.tb18189.x.
27. Porter, G. A., Jr., and Beutner, G. (2018) Cyclophil-
in D, somehow a master regulator of mitochondrial
function, Biomolecules, 8, 176, https://doi.org/10.3390/
biom8040176.
28. Giorgio,V., Bisetto,E., Soriano, M.E., Dabbeni-Sala,F.,
Basso,E., Petronilli,V., Forte, M.A., Bernardi, P., and
Lippe, G. (2009) Cyclophilin D modulates mitochon-
drial F
0
F
1
-ATP synthase by interacting with the lateral
stalk of the complex, J.Biol. Chem., 284, 33982-33988,
https://doi.org/10.1074/jbc.M109.020115.
29. Crompton, M., Virji, S., and Ward, J. M. (1988) Cy-
clophilin-D binds strongly to complexes of the volt-
age-dependent anion channel and the adenine nucle-
otide translocase to form the permeability transition
pore, Eur. J. Biochem., 258, 729-735, https://doi.org/
10.1046/j.1432-1327.1998.2580729.x.
30. Baines, C. P., Kaiser, R. A., Purcell, N. H., Blair, N. S.,
Osinska, H., Hambleton, M. A., Brunskill, E. W.,
Sayen, M.R., Gottlieb, R. A., Dorn, G. W., Robbins, J.,
and Molkentin, J. D. (2005) Loss of cyclophilin D
reveals a critical role for mitochondrial permea-
bility transition in cell death, Nature, 434, 658-662,
https://doi.org/10.1038/nature03434.
31. Scorrano, L., Nicolli, A., Basso, E., Petronilli, V., and
Bernardi, P. (1997) Two modes of activation of the
permeability transition pore: the role of mitochon-
drial cyclophilin, Mol. Cell Biochem., 174, 181-184,
https://doi.org/10.1007/978-1-4615-6111-8_27.
32. Beutner, G., Alanzalon, R. E., and Porter, G. A., Jr.
(2017) Cyclophilin D regulates the dynamic assem-
bly of mitochondrial ATP synthase into synthasomes,
Sci. Rep., 7, 14488, https://doi.org/10.1038/s41598-017-
14795-x.
33. Castillo, E. C., Morales, J. A., Chapoy-Villanueva, H.,
Silva-Platas,C., Treviño-Saldaña,N., Guerrero- Beltrán,
C. E., Bernal-Ramírez, J., Torres-Quintanilla, A.,
García, N., Youker, K., Torre-Amione, G., and García-
Rivas, G. (2019) Mitochondrial hyperacetylation in
the failing hearts of obese patients mediated partly
by a reduction in SIRT3: the involvement of the mi-
tochondrial permeability transition pore, Cell Physi-
ol. Biochem., 53, 465-479, https://doi.org/10.33594/
000000151.
34. Hurst, S., Gonnot, F., Dia, M., Crola Da Silva, C.,
Gomez, L., and Sheu, S. S. (2020) Phosphorylation
of cyclophilin D at serine 191 regulates mitochon-
drial permeability transition pore opening and cell
death after ischemia-reperfusion, Cell Death Dis.,
11, 661, https://doi.org/10.1038/s41419-020-02864-5.
BELOSLUDTSEV et al.1804
BIOCHEMISTRY (Moscow) Vol. 90 No. 12 2025
35. Rasola, A., Sciacovelli, M., Chiara, F., Pantic, B.,
Brusilow, W. S., and Bernardi, P. (2010) Activation
of mitochondrial ERK protects cancer cells from
death through inhibition of the permeability transi-
tion, Proc. Natl. Acad. Sci. USA, 107, 726-731, https://
doi.org/10.1073/pnas.0912742107.
36. Coluccino,G., Negro, A., Filippi,A., Bean,C., Muraca,
V.P., Gissi,C., Canetti,D., Mimmi, M.C., Zamprogno,E.,
Ciscato, F., Acquasaliente, L., De Filippis, V., Comel-
li, M., Carraro, M., Rasola, A., Gerle, C., Bernardi, P.,
Corazza,A., and Lippe,G. (2024) N-terminal cleavage
of cyclophilin D boosts its ability to bind F-ATP syn-
thase, Commun. Biol., 7, 1486, https://doi.org/10.1038/
s42003-024-07172-8.
37. Amanakis, G., Sun, J., Fergusson, M. M., McGinty, S.,
Liu, C., Molkentin, J. D., and Murphy, E. (2021) Cys-
teine 202 of cyclophilin D is a site of multiple
post-translational modifications and plays a role
in cardioprotection, Cardiovasc. Res., 117, 212-223,
https://doi.org/10.1093/cvr/cvaa053.
38. Monné, M., and Palmieri, F. (2014) Antiporters of
the mitochondrial carrier family, Curr. Top. Membr.,
73, 289-320, https://doi.org/10.1016/B978-0-12-800223-
0.00008-6.
39. Klingenberg, M. (2008) The ADP and ATP trans-
port in mitochondria and its carrier, Biochim. Bio-
phys. Acta, 1778, 1978-2021, https://doi.org/10.1016/
j.bbamem.2008.04.011.
40. Lim, C. H., Hamazaki, T., Braun, E. L., Wade, J., and
Terada, N. (2011) Evolutionary genomics implies a
specific function of Ant4 in mammalian and anole
lizard male germ cells, PLoS One, 6, e23122, https://
doi.org/10.1371/journal.pone.0023122.
41. Clémençon, B., Babot, M., and Trézéguet, V. (2013)
The mitochondrial ADP/ATP carrier (SLC25 family):
pathological implications of its dysfunction, Mol.
Aspects Med., 34, 485-493, https://doi.org/10.1016/
j.mam.2012.05.006.
42. Brustovetsky, N. (2020) The role of adenine nucle-
otide translocase in the mitochondrial permeabili-
ty transition, Cells, 9, 2686, https://doi.org/10.3390/
cells9122686.
43. Brustovetsky, N., Tropschug, M., Heimpel, S., Heid-
kämper,D., and Klingenberg,M. (2002) Alarge Ca
2+
-de-
pendent channel formed by recombinant ADP/ATP
carrier from Neurospora crassa resembles the mito-
chondrial permeability transition pore, Biochemistry,
41, 11804-11811, https://doi.org/10.1021/bi0200110.
44. McStay, G. P., Clarke, S.J., and Halestrap, A.P. (2002)
Role of critical thiol groups on the matrix surface
of the adenine nucleotide translocase in the mech-
anism of the mitochondrial permeability transition
pore, Biochem.J., 367, 541-548, https://doi.org/10.1042/
BJ20011672.
45. Karch, J., Bround, M. J., Khalil, H., Sargent, M. A.,
Latchman,N., Terada,N., Peixoto, P.M., and Molken-
tin, J. D. (2019) Inhibition of mitochondrial permea-
bility transition by deletion of the ANT family and
CypD, Sci Adv., 5, eaaw4597, https://doi.org/10.1126/
sciadv.aaw4597.
46. Dubinin, M.V., Talanov, E.Y., Tenkov, K.S., Starinets,
V. S., Mikheeva, I. B., Sharapov, M. G., and Beloslud-
tsev, K. N. (2020) Duchenne muscular dystrophy is
associated with the inhibition of calcium uniport
in mitochondria and an increased sensitivity of the
organelles to the calcium-induced permeability tran-
sition, Biochim. Biophys. Acta Mol. Basis Dis., 1866,
165674, https://doi.org/10.1016/j.bbadis.2020.165674.
47. Belosludtsev, K. N., Talanov, E. Y., Starinets, V. S.,
Agafonov, A. V., Dubinin, M. V., and Belosludtseva,
N. V. (2019) Transport of Ca
2+
and Ca
2+
-dependent
permeability transition in rat liver mitochondria un-
der the streptozotocin-induced type I diabetes, Cells,
8, 1014, https://doi.org/10.3390/cells8091014.
48. Belosludtseva, N. V., Ilzorkina, A. I., Serov, D. A.,
Dubinin, M. V., Talanov, E. Y., Karagyaur, M. N.,
Primak, A. L., Liu, J., and Belosludtsev, K. N. (2024)
ANT-mediated inhibition of the permeability tran-
sition pore alleviates palmitate-induced mitochon-
drial dysfunction and lipotoxicity, Biomolecules, 14,
1159, https://doi.org/10.3390/biom14091159.
49. Alves-Figueiredo, H., Silva-Platas, C., Lozano, O.,
Vázquez-Garza, E., Guerrero-Beltrán, C. E., Zarain-
Herzberg,A., and García-Rivas,G. (2021) Asystematic
review of post-translational modifications in the mi-
tochondrial permeability transition pore complex as-
sociated with cardiac diseases, Biochim. Biophys. Acta
Mol. Basis Dis., 1867, 165992, https://doi.org/10.1016/
j.bbadis.2020.165992.
50. Macdonald, J.E., and Ashby, P.D. (2025) Themolecu-
lar mechanism of ATP synthase constrains the evolu-
tionary landscape of chemiosmosis, Biophys. J., 124,
2103-2119, https://doi.org/10.1016/j.bpj.2025.05.017.
51. Pinke,G., Zhou,L., and Sazanov, L.A. (2020) Cryo-EM
structure of the entire mammalian F-type ATP syn-
thase, Nat. Struct. Mol. Biol., 27, 1077-1085, https://
doi.org/10.1038/s41594-020-0503-8.
52. Kinnally, K. W., Campo, M. L., and Tedeschi, H.
(1989) Mitochondrial channel activity studied by
patch-clamping mitoplasts, J.Bioenerg. Biomembr., 21,
497-506, https://doi.org/10.1007/BF00762521.
53. Urbani, A., Giorgio, V., Carrer, A., Franchin, C.,
Arrigoni, G., Jiko, C., Abe, K., Maeda, S., Shinzawa-
Itoh,K., Bogers, J.F. M., McMillan, D.G. G., Gerle,C.,
Szabò, I., and Bernardi, P. (2019) Purified F-ATP
synthase forms a Ca
2+
-dependent high-conductance
channel matching the mitochondrial permeabili-
ty transition pore, Nat. Commun., 10, 4341, https://
doi.org/10.1038/s41467-019-12331-1.
54. Carrer, A., Tommasin, L., Šileikytė, J., Ciscato, F.,
Filadi, R., Urbani, A., Forte, M., Rasola, A., Szabò, I.,
Carraro, M., and Bernardi, P. (2021) Defining the
MPT PORE: STRUCTURE AND FUNCTIONS 1805
BIOCHEMISTRY (Moscow) Vol. 90 No. 12 2025
molecular mechanisms of the mitochondrial per-
meability transition through genetic manipulation
of F-ATP synthase, Nat. Commun., 12, 4835, https://
doi.org/10.1038/s41467-021-25161-x.
55. He,J., Carroll,J., Ding,S., Fearnley, I.M., and Walker,
J. E. (2017) Permeability transition in human mito-
chondria persists in the absence of peripheral stalk
subunits of ATP synthase, Proc. Natl. Acad. Sci.
USA, 114, 9086-9091, https://doi.org/10.1073/pnas.
1711201114.
56. Carraro, M., Giorgio, V., Šileikytė, J., Sartori, G.,
Forte, M., Lippe, G., Zoratti, M., Szabò, I., and Ber-
nardi, P. (2014) Channel formation by yeast F-ATP
synthase and the role of dimerization in the mi-
tochondrial permeability transition, J.Biol. Chem.,
289, 15980-15985, https://doi.org/10.1074/jbc.C114.
559633.
57. Campanella, M., Casswell, E., Chong, S., Farah, Z.,
Wieckowski, M. R., Abramov, A. Y., Tinker, A., and
Duchen, M. R. (2008) Regulation of mitochondri-
al structure and function by the F
1
Fo-ATPase in-
hibitor protein, IF1, Cell Metab., 8, 13-25, https://
doi.org/10.1016/j.cmet.2008.06.001.
58. Carroll,J., He,J., Ding,S., Fearnley, I.M., and Walker,
J.E. (2019) Persistence of the permeability transition
pore in human mitochondria devoid of an assembled
ATP synthase, Proc. Natl. Acad. Sci. USA, 116, 12816-
12821, https://doi.org/10.1073/pnas.1904005116.
59. Mnatsakanyan, N., Llaguno, M. C., Yang, Y., Yan, Y.,
Weber, J., Sigworth, F. J., and Jonas, E. A. (2019)
Amitochondrial megachannel resides in monomeric
F
1
F
O
ATP synthase, Nat. Commun., 10, 5823, https://
doi.org/10.1038/s41467-019-13766-2.
60. Alavian, K.N., Beutner,G., Lazrove,E., Sacchetti,S., Park,
H.A., Licznerski,P., Li, H., Nabili,P., Hockensmith,K.,
Graham, M., Porter, G. A., Jr., and Jonas, E. A. (2014)
An uncoupling channel within the c-subunit ring
of the F
1
F
O
ATP synthase is the mitochondrial
permeability transition pore, Proc. Natl. Acad. Sci.
USA, 111, 10580-10585, https://doi.org/10.1073/pnas.
1401591111.
61. Bonora, M., Bononi, A., De Marchi, E., Giorgi, C.,
Lebiedzinska, M., Marchi, S., Patergnani, S., Rimes-
si, A., Suski, J. M., Wojtala, A., Wieckowski, M. R.,
Kroemer, G., Galluzzi, L., and Pinton, P. (2013) Role
of the c subunit of the FO ATP synthase in mitochon-
drial permeability transition, Cell Cycle, 12, 674-683,
https://doi.org/10.4161/cc.23599.
62. Zhou, W., Marinelli, F., Nief, C., and Faraldo- Gómez,
J. D. (2017) Atomistic simulations indicate the c-sub-
unit ring of the F
1
F
O
ATP synthase is not the mi-
tochondrial permeability transition pore, Elife, 6,
e23781, https://doi.org/10.7554/eLife.23781.
63. Gerle, C. (2020) Mitochondrial F-ATP synthase as
the permeability transition pore, Pharmacol Res.,
160, 105081, https://doi.org/10.1016/j.phrs.2020.105081.
64. Morciano, G., Pedriali, G., Bonora, M., Pavasini, R.,
Mikus, E., Calvi, S., Bovolenta, M., Lebiedzinska-
Arciszewska,M., Pinotti,M., Albertini,A., Wieckowski,
M. R., Giorgi, C., Ferrari, R., Galluzzi, L., Campo, G.,
and Pinton, P. (2021) A naturally occurring muta-
tion in ATP synthase subunit c is associated with in-
creased damage following hypoxia/reoxygenation in
STEMI patients, Cell Rep., 35, 108983, https://doi.org/
10.1016/j.celrep.2021.108983.
65. He,J., Ford, H.C., Carroll,J., Ding,S., Fearnley, I.M., and
Walker, J. E. (2017) Persistence of the mitochondrial
permeability transition in the absence of subunit c of
human ATP synthase, Proc. Natl. Acad. Sci. USA, 114,
3409-3414, https://doi.org/10.1073/pnas.1702357114.
66. Neginskaya, M. A., Morris, S. E., and Pavlov, E. V.
(2022) Both ANT and ATPase are essential for mito-
chondrial permeability transition but not depolar-
ization, iScience, 25, 105447, https://doi.org/10.1016/
j.isci.2022.105447.
67. Neginskaya, M. A., Morris, S. E., and Pavlov, E. V.
(2023) Refractive index imaging reveals that elimina-
tion of the ATP synthase Csubunit does not prevent
the adenine nucleotide translocase-dependent mi-
tochondrial permeability transition, Cells, 12, 1950,
https://doi.org/10.3390/cells12151950.
68. Tommasin, L., Carrer, A., Nata, F. B., Frigo, E.,
Fogolari, F., Lippe, G., Carraro, M., and Bernardi, P.
(2025) Adenine nucleotide translocator and ATP syn-
thase cooperate in mediating the mitochondrial per-
meability transition, J.Physiol., https://doi.org/10.1113/
JP287147.
69. Richardson, A. P., and Halestrap, A. P. (2016) Quan-
tification of active mitochondrial permeability
transition pores using GNX-4975 inhibitor titra-
tions provides insights into molecular identity, Bio-
chem. J., 473, 1129-1140, https://doi.org/10.1042/
BCJ20160070.
70. Neginskaya, M.A., Strubbe, J.O., Amodeo, G.F., West,
B. A., Yakar, S., Bazil, J. N., and Pavlov, E. V. (2020)
Thevery low number of calcium-induced permeabili-
ty transition pores in the single mitochondrion, J.Gen.
Physiol., 152, e202012631, https://doi.org/10.1085/
jgp.202012631.
71. Belosludtseva, N.V., Dubinin, M.V., and Belosludtsev,
K.N. (2024) Pore-forming VDAC proteins of the outer
mitochondrial membrane: regulation and pathophys-
iological role, Biochemistry (Moscow), 89, 1061-1078,
https://doi.org/10.1134/S0006297924060075.
72. De Macedo, D.V., da Costa,C., and Pereira-Da-Silva,L.
(1997) The permeability transition pore opening in
intact mitochondria and submitochondrial parti-
cles, Comp. Biochem. Physiol. B Biochem. Mol. Biol.,
118, 209-216, https://doi.org/10.1016/s0305-0491(97)
00007-2.
73. Belosludtsev, K. N., Belosludtseva, N. V., Agafonov,
A.V., Astashev, M.E., Kazakov, A.S., Saris, N.E., and
BELOSLUDTSEV et al.1806
BIOCHEMISTRY (Moscow) Vol. 90 No. 12 2025
Mironova, G. D. (2014) Ca
2+
-dependent permeabili-
zation of mitochondria and liposomes by palmitic
and oleic acids: a comparative study, Biochim. Bio-
phys. Acta, 1838, 2600-2606, https://doi.org/10.1016/
j.bbamem.2014.06.017.
74. Baines, C. P., Kaiser, R. A., Sheiko, T., Craigen, W. J.,
and Molkentin, J. D. (2007) Voltage-dependent anion
channels are dispensable for mitochondrial-depen-
dent cell death, Nat. Cell Biol., 9, 550-555, https://
doi.org/10.1038/ncb1575.
75. Jaremko, L., Jaremko, M., Giller, K., Becker, S., and
Zweckstetter,M. (2014) Structure of the mitochondri-
al translocator protein in complex with a diagnostic
ligand, Science, 343, 1363-1366, https://doi.org/10.1126/
science.1248725.
76. Morin, D., Musman, J., Pons, S., Berdeaux, A., and
Ghaleh, B. (2016) Mitochondrial translocator protein
(TSPO): from physiology to cardioprotection, Bio-
chem. Pharmacol., 105, 1-13, https://doi.org/10.1016/
j.bcp.2015.12.003.
77. Shoshan-Barmatz, V., Pittala, S., and Mizrachi, D.
(2019) VDAC1 and the TSPO: expression, interac-
tions, and associated functions in health and disease
states, Int.J. Mol. Sci., 20, 3348, https://doi.org/10.3390/
ijms20133348.
78. Azarashvili, T., Grachev, D., Krestinina, O.,
Evtodienko, Y., Yurkov, I., Papadopoulos, V., and
Reiser, G. (2007) The peripheral-type benzodiazepine
receptor is involved in control of Ca
2+
-induced per-
meability transition pore opening in rat brain mi-
tochondria, Cell Calcium, 42, 27-39, https://doi.org/
10.1016/j.ceca.2006.11.004.
79. Šileikytė, J., Blachly-Dyson, E., Sewell, R., Carpi, A.,
Menabò,R., Di Lisa, F., Ricchelli, F., Bernardi, P., and
Forte,M. (2014) Regulation of the mitochondrial per-
meability transition pore by the outer membrane
does not involve the peripheral benzodiazepine re-
ceptor (Translocator Protein of 18kDa (TSPO)), J.Biol.
Chem., 289, 13769-13781, https://doi.org/10.1074/
jbc.M114.549634.
80. Whelan, R.S., Konstantinidis,K., Wei, A.C., Chen, Y.,
Reyna, D.E., Jha,S., Yang,Y., Calvert, J.W., Lindsten,T.,
Thompson, C. B., Crow, M. T., Gavathiotis, E., Dorn,
G. W., 2nd, O’Rourke,B., and Kitsis, R. N. (2012) Bax
regulates primary necrosis through mitochondrial
dynamics, Proc. Natl. Acad. Sci. USA, 109, 6566-6571,
https://doi.org/10.1073/pnas.1201608109.
81. Gutiérrez-Aguilar, M., Douglas, D. L., Gibson, A. K.,
Domeier, T. L., Molkentin, J. D., and Baines, C. P.
(2014) Genetic manipulation of the cardiac mito-
chondrial phosphate carrier does not affect perme-
ability transition, J. Mol. Cell Cardiol., 72, 316-325,
https://doi.org/10.1016/j.yjmcc.2014.04.008.
82. Nikiforova, A.B., Baburina, Y.L., Borisova, M.P., Surin,
A. K., Kharechkina, E. S., Krestinina, O. V., Suvorina,
M.Y., Kruglova, S.A., and Kruglov, A.G. (2023) Mito-
chondrial F-ATP synthase co-migrating proteins and
Ca
2+
-dependent formation of large channels, Cells, 12,
414, https://doi.org/10.3390/cells12192414.
83. Shanmughapriya,S., Rajan,S., Hoffman, N.E., Higgins,
A.M., Tomar,D., Nemani,N., Hines, K.J., Smith, D.J.,
Eguchi,A., Vallem,S., Shaikh,F., Cheung,M., Leonard,
N.J., Stolakis, R.S., Wolfers, M.P., Ibetti,J., Chuprun,
J.K., Jog, N.R., Houser, S.R., Koch, W.J., Elrod, J.W.,
and Madesh, M. (2015) SPG7 is an essential and
conserved component of the mitochondrial perme-
ability transition pore, Mol. Cell, 60, 47-62, https://
doi.org/10.1016/j.molcel.2015.08.009.
84. Hurst, S., Baggett, A., Csordas, G., and Sheu, S. S.
(2019) SPG7 targets the m-AAA protease complex to
process MCU for uniporter assembly, Ca
2+
influx, and
regulation of mitochondrial permeability transition
pore opening, J.Biol. Chem., 294, 10807-10818, https://
doi.org/10.1074/jbc.RA118.006443.
85. Sambri, I., Massa, F., Gullo, F., Meneghini, S.,
Cassina, L., Carraro, M., Dina, G., Quattrini, A.,
Patanella, L., Carissimo, A., Iuliano, A., Santorelli, F.,
Codazzi, F., Grohovaz, F., Bernardi, P., Becchetti, A.,
and Casari, G. (2020) Impaired flickering of the per-
meability transition pore causes SPG7 spastic para-
plegia, EBioMedicine, 61, 103050, https://doi.org/
10.1016/j.ebiom.2020.103050.
86. Klutho, P. J., Dashek, R. J., Song, L., and Baines, C. P.
(2020) Genetic manipulation of SPG7 or NipSnap2
does not affect mitochondrial permeability transi-
tion, Cell Death Discov., 6, 5, https://doi.org/10.1038/
s41420-020-0239-6.
87. Dolder,M., Wendt,S., and Wallimann,T. (2001) Mito-
chondrial creatine kinase in contact sites: interaction
with porin and adenine nucleotide translocase, role
in permeability transition and sensitivity to oxida-
tive damage, Biol. Signals Recept., 10, 93-111, https://
doi.org/10.1159/000046878.
88. Smeele, K. M., Southworth, R., Wu, R., Xie, C.,
Nederlof,R., Warley,A., Nelson, J.K., van Horssen,P.,
van den Wijngaard, J. P., Heikkinen, S., Laakso, M.,
Koeman, A., Siebes, M., Eerbeek, O., Akar, F. G.,
Ardehali, H., Hollmann, M. W., and Zuurbier, C. J.
(2011) Disruption of hexokinase II-mitochondrial
binding blocks ischemic preconditioning and caus-
es rapid cardiac necrosis, Circ. Res., 108, 1165-1169,
https://doi.org/10.1161/CIRCRESAHA.111.244962.
89. Juhaszova, M., Wang, S., Zorov, D. B., Nuss, H. B.,
Gleichmann,M., Mattson, M.P., and Sollott, S.J. (2008)
Theidentity and regulation of the mitochondrial per-
meability transition pore: where the known meets
the unknown, Ann. N. Y. Acad. Sci., 1123, 197-212,
https://doi.org/10.1196/annals.1420.023.
90. Guo, D., Meng, Y., Jiang, X., and Lu, Z. (2023) Hex-
okinases in cancer and other pathologies, Cell In-
sight., 2, 100077, https://doi.org/10.1016/j.cellin.2023.
100077.
MPT PORE: STRUCTURE AND FUNCTIONS 1807
BIOCHEMISTRY (Moscow) Vol. 90 No. 12 2025
91. Zoratti, M., Szabò, I., and De Marchi, U. (2005) Mito-
chondrial permeability transitions: how many doors
to the house? Biochim. Biophys. Acta, 1706, 40-52,
https://doi.org/10.1016/j.bbabio.2004.10.006.
92. Sultan,A., and Sokolove,P. (2001) Palmitic acid opens
a novel cyclosporin A-insensitive pore in the inner
mitochondrial membrane, Arch. Biochem. Biophys.,
386, 31-51, https://doi.org/10.1006/abbi.2000.2194.
93. Agafonov, A., Gritsenko, E., Belosludtsev, K.,
Kovalev, A., Gateau-Roesch, O., Saris, N.-E. L., and
Mironova, G. D. (2003) A permeability transition in
liposomes induced by the formation of Ca
2+
/palmitic
acid complexes, Biochim. Biophys. Acta, 1609, 153-
160, https://doi.org/10.1016/S0005-2736(02)00666-1.
94. Belosludtseva, N. V., Pavlik, L. L., Belosludtsev, K.N.,
Saris, N.L., Shigaeva, M.I., and Mironova, G.D. (2022)
Theshort-term opening of cyclosporin A-independent
palmitate/Sr
2+
-induced pore can underlie ion efflux
in the oscillatory mode of functioning of rat liver
mitochondria, Membranes (Basel), 12, 667, https://
doi.org/10.3390/membranes12070667.
95. Šileikytė, J., and Forte, M. (2016) Shutting down the
pore: the search for small molecule inhibitors of the
mitochondrial permeability transition, Biochim. Bio-
phys. Acta, 1857, 1197-1202, https://doi.org/10.1016/
j.bbabio.2016.02.016.
96. Efimov, S.V., Dubinin, M.V., Kobchikova, P.P., Zgadzay,
Y.O., Khodov, I.A., Belosludtsev, K.N., and Klochkov,
V. V. (2020) Comparison of cyclosporin variants B-E
based on their structural properties and activity in
mitochondrial membranes, Biochem. Biophys. Res.
Commun., 526, 1054-1060, https://doi.org/10.1016/
j.bbrc.2020.03.184.
97. Rosenwirth, B., Billich, A., Datema, R., Donatsch, P.,
Hammerschmid, F., Harrison, R., Hiestand, P.,
Jaksche,H., Mayer,P., and Peichl,P. (1994) Inhibition
of human immunodeficiency virus type1 replication
by SDZ NIM 811, a nonimmunosuppressive cyclospo-
rine analog, Antimicrob. Agents Chemother., 38, 1763-
1772, https://doi.org/10.1128/AAC.38.8.1763.
98. Reutenauer, J., Dorchies, O. M., Patthey-Vuadens, O.,
Vuagniaux, G., and Ruegg, U. T. (2008) Investigation
of Debio 025, a cyclophilin inhibitor, in the dystro-
phic mdx mouse, a model for Duchenne muscular
dystrophy, Br. J. Pharmacol., 155, 574-584, https://
doi.org/10.1038/bjp.2008.285.
99. Dubinin, M.V., Starinets, V.S., Talanov, E.Y., Mikheeva,
I. B., Belosludtseva, N. V., and Belosludtsev, K. N.
(2021) Alisporivir improves mitochondrial function
in skeletal muscle of mdx mice but suppresses mito-
chondrial dynamics and biogenesis, Int.J. Mol. Sci.,
22, 9780, https://doi.org/10.3390/ijms22189780.
100. Tiepolo, T., Angelin, A., Palma, E., Sabatelli, P.,
Merlini, L., Nicolosi, L., Finetti, F., Braghetta, P.,
Vuagniaux,G., Dumont, J.M., Baldari, C.T., Bonaldo,P.,
and Bernardi, P. (2009) The cyclophilin inhibitor
Debio 025 normalizes mitochondrial function, mus-
cle apoptosis and ultrastructural defects in Col6a1
–/–
myopathic mice, Br. J. Pharmacol., 157, 1045-1052,
https://doi.org/10.1111/j.1476-5381.2009.00316.x.
101. Montagne, A., Nikolakopoulou, A. M., Huuskonen,
M.T., Sagare, A.P., Lawson, E.J., Lazic,D., Rege, S.V.,
Grond,A., Zuniga,E., Barnes, S.R., Prince,J., Sagare,M.,
Hsu, C.J., LaDu, M.J., Jacobs, R.E., and Zlokovic, B.V.
(2021) APOE4 accelerates advanced-stage vascular
and neurodegenerative disorder in old Alzheimer’s
mice via cyclophilin A independently of amyloid-β,
Nat. Aging, 1, 506-520, https://doi.org/10.1038/s43587-
021-00073-z.
102. Dubinin, M.V., Sharapov, V.A., Ilzorkina, A.I., Efimov,
S.V., Klochkov, V. V., Gudkov, S. V., and Belosludtsev,
K.N. (2022) Comparison of structural properties of cy-
closporinA and its analogue alisporivir and their ef-
fects on mitochondrial bioenergetics and membrane
behavior, Biochim. Biophys. Acta Biomembr., 1864,
183972, https://doi.org/10.1016/j.bbamem.2022.183972.
103. Schaller, S., Paradis, S., Ngoh, G. A., Assaly, R.,
Buisson, B., Drouot, C., Ostuni, M. A., Lacapere, J. J.,
Bassissi, F., Bordet, T., Berdeaux, A., Jones, S. P.,
Morin, D., and Pruss, R. M. (2010) TRO40303, a new
cardioprotective compound, inhibits mitochondrial
permeability transition, J.Pharmacol. Exp. Ther., 333,
696-706, https://doi.org/10.1124/jpet.110.167486.
104. Clarke, S.J., McStay, G.P., and Halestrap, A.P. (2002)
Sanglifehrin A acts as a potent inhibitor of the mi-
tochondrial permeability transition and reperfusion
injury of the heart by binding to cyclophilin-D at a
different site from cyclosporinA, J.Biol. Chem., 277,
34793-34799, https://doi.org/10.1074/jbc.M202191200.
105. Lim, S.Y., Davidson, S.M., Hausenloy, D.J., and Yellon,
D. M. (2007) Preconditioning and postconditioning:
the essential role of the mitochondrial permeability
transition pore, Cardiovasc. Res., 75, 530-535, https://
doi.org/10.1016/j.cardiores.2007.04.022.
106. Albanese, V., Pedriali, G., Fabbri, M., Ciancetta, A.,
Ravagli, S., Roccatello, C., Guerrini, R., Morciano, G.,
Preti, D., Pinton, P., and Pacifico, S. (2025) Design
and synthesis of 1,4,8-triazaspiro[4.5]decan-2-one de-
rivatives as novel mitochondrial permeability tran-
sition pore inhibitors, J.Enzyme Inhib. Med. Chem.,
40, 2505907, https://doi.org/10.1080/14756366.2025.
2505907.
107. Fancelli, D., Abate, A., Amici, R., Bernardi, P.,
Ballarini, M., Cappa, A., Carenzi, G., Colombo, A.,
Contursi, C., Di Lisa, F., Dondio, G., Gagliardi, S.,
Milanesi, E., Minucci, S., Pain, G., Pelicci, P. G.,
Saccani,A., Storto,M., Thaler,F., Varasi,M., Villa,M.,
and Plyte, S. (2014) Cinnamic anilides as new mito-
chondrial permeability transition pore inhibitors en-
dowed with ischemia-reperfusion injury protective
effect in vivo, J. Med. Chem., 57, 5333-5347, https://
doi.org/10.1021/jm500547c.
BELOSLUDTSEV et al.1808
BIOCHEMISTRY (Moscow) Vol. 90 No. 12 2025
108. Šileikytė, J., Devereaux, J., de Jong, J., Schiavone, M.,
Jones, K., Nilsen, A., Bernardi, P., Forte, M., and
Cohen, M. S. (2019) Second-generation inhibitors
of the mitochondrial permeability transition pore
with improved plasma stability, ChemMedChem, 14,
1771-1782, https://doi.org/10.1002/cmdc.201900376.
109. Roy,S., Šileikytė,J., Schiavone,M., Neuenswander,B.,
Argenton, F., Aubé, J., Hedrick, M. P., Chung, T. D.,
Forte, M. A., Bernardi, P., and Schoenen, F. J. (2015)
Discovery, synthesis, and optimization of diaryli-
soxazole-3-carboxamides as potent inhibitors of
the mitochondrial permeability transition pore,
ChemMedChem, 10, 1655-1671, https://doi.org/10.1002/
cmdc.201500284.
110. Stocco, A., Smolina, N., Sabatelli, P., Šileikytė, J.,
Artusi, E., Mouly, V., Cohen, M., Forte, M.,
Schiavone, M., and Bernardi, P. (2021) Treatment
with a triazole inhibitor of the mitochondrial per-
meability transition pore fully corrects the patholo-
gy of sapje zebrafish lacking dystrophin, Pharmacol
Res., 165, 105421, https://doi.org/10.1016/j.phrs.2021.
105421.
111. Bernardi, P., Krauskopf, A., Basso, E., Petronilli, V.,
Blachly-Dyson, E., Di Lisa, F., and Forte, M. A. (2006)
Themitochondrial permeability transition from in vi-
tro artifact to disease target, FEBSJ., 273, 2077-2099,
https://doi.org/10.1111/j.1742-4658.2006.05213.x.
112. Panel,M., Ghaleh,B., and Morin,D. (2018) Mitochon-
dria and aging: Arole for the mitochondrial transition
pore? Aging Cell, 17, e12793, https://doi.org/10.1111/
acel.12793.
113. Angeli, S., Foulger, A., Chamoli, M., Peiris, T. H.,
Gerencser, A., Shahmirzadi, A. A., Andersen, J., and
Lithgow, G. (2021) The mitochondrial permeability
transition pore activates the mitochondrial unfold-
ed protein response and promotes aging, Elife, 10,
e63453, https://doi.org/10.7554/eLife.63453.
114. Du, H., Guo, L., Fang, F., Chen, D., Sosunov, A. A.,
McKhann, G. M., Yan, Y., Wang, C., Zhang, H.,
Molkentin, J. D., Gunn-Moore, F. J., Vonsattel, J. P.,
Arancio, O., Chen, J. X., and Yan, S. D. (2008) Cyclo-
philin D deficiency attenuates mitochondrial and
neuronal perturbation and ameliorates learning and
memory in Alzheimers disease, Nat. Med., 14, 1097-
1105, https://doi.org/10.1038/nm.1868.
115. Zhu, Y., Duan, C., Lü, L., Gao, H., Zhao, C., Yu, S.,
Uéda, K., Chan, P., and Yang, H. (2011) α-Synuclein
overexpression impairs mitochondrial function by
associating with adenylate translocator, Int. J. Bio-
chem. Cell Biol., 43, 732-741, https://doi.org/10.1016/
j.biocel.2011.01.014.
116. Belosludtseva, N.V., Matveeva, L.A., and Belosludtsev,
K. N. (2023) Mitochondrial dyshomeostasis as an
early hallmark and a therapeutic target in amyo-
trophic lateral sclerosis, Int.J. Mol. Sci., 24, 16833,
https://doi.org/10.3390/ijms242316833.
117. Singh,S., Ganguly,U., Pal,S., Chandan,G., Thakur,R.,
Saini, R. V., Chakrabarti, S. S., Agrawal, B. K., and
Chakrabarti, S. (2022) Protective effects of cyclospo-
rineA on neurodegeneration and motor impairment
in rotenone-induced experimental models of Parkin-
son’s disease, Eur.J. Pharmacol., 929, 175129, https://
doi.org/10.1016/j.ejphar.2022.175129.
118. Bordet, T., Berna, P., Abitbol, J. L., and Pruss, R. M.
(2010) Olesoxime (TRO19622): a novel mitochon-
drial-targeted neuroprotective compound, Pharma-
ceuticals (Basel), 3, 345-368, https://doi.org/10.3390/
ph3020345.
119. Argaud, L., Gateau-Roesch, O., Muntean, D.,
Chalabreysse, L., Loufouat, J., Robert, D., and
Ovize, M. (2005) Specific inhibition of the mito-
chondrial permeability transition prevents lethal
reperfusion injury, J. Mol. Cell Cardiol., 38, 367-374,
https://doi.org/10.1016/j.yjmcc.2004.12.001.
120. Nakagawa,T., Shimizu,S., Watanabe,T., Yamaguchi,O.,
Otsu, K., Yamagata, H., Inohara, H., Kubo, T., and
Tsujimoto, Y. (2005) Cyclophilin D-dependent mito-
chondrial permeability transition regulates some ne-
crotic but not apoptotic cell death, Nature, 434, 652-
658, https://doi.org/10.1038/nature03317.
121. Cung, T. T., Morel, O., Cayla, G., Rioufol, G., Garcia-
Dorado, D., Angoulvant, D., Bonnefoy-Cudraz, E.,
Guérin,P., Elbaz,M., Delarche,N., Coste,P., Vanzetto,G.,
Metge,M., Aupetit, J.F., Jouve,B., Motreff,P., Tron,C.,
Labeque, J. N., Steg, P. G., et al. (2015) Cyclosporine
before PCI in patients with acute myocardial infarc-
tion, N.Engl.J. Med., 373, 1021-1031, https://doi.org/
10.1056/NEJMoa1505489.
122. Wrogemann,K., and Pena, S.D. (1976) Mitochondrial
calcium overload: a general mechanism for cell-
necrosis in muscle diseases, Lancet, 1, 672-674, https://
doi.org/10.1016/s0140-6736(76)92781-1.
123. Zulian,A., Schiavone,M., Giorgio,V., and Bernardi,P.
(2016) Forty years later: mitochondria as therapeu-
tic targets in muscle diseases, Pharmacol. Res., 113,
563-573, https://doi.org/10.1016/j.phrs.2016.09.043.
124. Millay, D. P., Sargent, M. A., Osinska, H., Baines,
C. P., Barton, E. R., Vuagniaux, G., Sweeney, H. L.,
Robbins, J., and Molkentin, J. D. (2008) Genetic and
pharmacologic inhibition of mitochondrial-dependent
necrosis attenuates muscular dystrophy, Nat. Med.,
14, 442-447, https://doi.org/10.1038/nm1736.
125. Dubinin, M. V., and Belosludtsev, K. N. (2023) Ion
channels of the sarcolemma and intracellular or-
ganelles in duchenne muscular dystrophy: a role in
the dysregulation of ion homeostasis and a possible
target for therapy, Int.J. Mol. Sci., 24, 2229, https://
doi.org/10.3390/ijms24032229.
126. Wang, P., Zhang, N., Wu, B., Wu, S., Zhang, Y., and
Sun, Y. (2020) The role of mitochondria in vascular
calcification, J.Transl. Int. Med., 8, 80-90, https://
doi.org/10.2478/jtim-2020-0013.
MPT PORE: STRUCTURE AND FUNCTIONS 1809
BIOCHEMISTRY (Moscow) Vol. 90 No. 12 2025
127. Dubinin, M.V., Talanov, E.Y., Tenkov, K.S., Starinets,
V. S., Mikheeva, I. B., and Belosludtsev, K. N. (2020)
Transport of Ca
2+
and Ca
2+
-dependent permeability
transition in heart mitochondria in the early stages
of Duchenne muscular dystrophy, Biochim. Biophys.
Acta Bioenerg., 1861, 148250, https://doi.org/10.1016/
j.bbabio.2020.148250.
128. Bround, M. J., Havens, J. R., York, A. J., Sargent,
M. A., Karch, J., and Molkentin, J. D. (2023) ANT-
dependent MPTP underlies necrotic myofiber death
in muscular dystrophy, Sci. Adv., 9, eadi2767, https://
doi.org/10.1126/sciadv.adi2767.
129. Belosludtseva, N. V., Ilzorkina, A. I., Dubinin, M. V.,
Mikheeva, I. B., and Belosludtsev, K. N. (2025) Com-
parative study of structural and functional rearrange-
ments in skeletal muscle mitochondria of SOD1-G93A
transgenic mice at pre-, early-, and late-symptomatic
stages of ALS progression, Front. Biosci. (Landmark
Ed), 30, 28260, https://doi.org/10.31083/FBL28260.
130. Hepple, R. T. (2016) Impact of aging on mitochon-
drial function in cardiac and skeletal muscle, Free
Radic. Biol. Med., 98, 177-186, https://doi.org/10.1016/
j.freeradbiomed.2016.03.017.
131. Belosludtsev, K.N., Belosludtseva, N.V., and Dubinin,
M. V. (2020) Diabetes mellitus, mitochondrial dys-
function and Ca
2+
-dependent permeability transition
pore, Int.J. Mol. Sci., 21, 6559, https://doi.org/10.3390/
ijms21186559.
132. Lablanche, S., Cottet-Rousselle, C., Lamarche, F.,
Benhamou, P. Y., Halimi, S., Leverve, X., and Fon-
taine,E. (2011) Protection of pancreatic INS-1 β-cells
from glucose- and fructose-induced cell death by in-
hibiting mitochondrial permeability transition with
cyclosporinA or metformin, Cell Death Dis., 2, e134,
https://doi.org/10.1038/cddis.2011.15.
133. Belosludtseva, N. V., Starinets, V. S., Mikheeva, I. B.,
Serov, D. A., Astashev, M. E., Belosludtsev, M. N.,
Dubinin, M. V., and Belosludtsev, K. N. (2021) Effect
of the MPT pore inhibitor alisporivir on the devel-
opment of mitochondrial dysfunction in the heart
tissue of diabetic mice, Biology (Basel), 10, 839,
https://doi.org/10.3390/biology10090839.
134. Couturier,K., Hininger,I., Poulet, L., Anderson, R. A.,
Roussel, A. M., Canini, F., and Batandier, C. (2016)
Cinnamon intake alleviates the combined effects of
dietary-induced insulin resistance and acute stress
on brain mitochondria, J.Nutr. Biochem., 28, 183-190,
https://doi.org/10.1016/j.jnutbio.2015.10.016.
135. Oliveira, P. J., Seiça, R., Coxito, P. M., Rolo, A. P.,
Palmeira, C.M., Santos, M.S., and Moreno, A.J. (2003)
Enhanced permeability transition explains the re-
duced calcium uptake in cardiac mitochondria from
streptozotocin-induced diabetic rats, FEBS Lett., 554,
511-514, https://doi.org/10.1016/s0014-5793(03)01233-x.
136. Monaco, C. M. F., Hughes, M. C., Ramos, S. V., Varah,
N. E., Lamberz, C., Rahman, F. A., McGlory, C.,
Tarnopolsky, M. A., Krause, M.P., Laham, R., Hawke,
T.J., and Perry, C.G. R. (2018) Altered mitochondrial
bioenergetics and ultrastructure in the skeletal mus-
cle of young adults with type 1 diabetes, Diabetolo-
gia, 61, 1411-1423, https://doi.org/10.1007/s00125-018-
4602-6.
137. Ferreira, F. M., Seiça, R., Oliveira, P. J., Coxito, P. M.,
Moreno, A.J., Palmeira, C.M., and Santos, M.S. (2003)
Diabetes induces metabolic adaptations in rat liver
mitochondria: role of coenzyme Q and cardiolip-
in contents, Biochim. Biophys. Acta, 1639, 113-120,
https://doi.org/10.1016/j.bbadis.2003.08.001.
138. Teodoro, J. S., Rolo, A.P., Duarte, F.V., Simões, A.M.,
and Palmeira, C.M. (2008) Differential alterations in
mitochondrial function induced by a choline-deficient
diet: understanding fatty liver disease progression,
Mitochondrion, 8, 367-376, https://doi.org/10.1016/
j.mito.2008.07.008.
139. Taddeo, E. P., Laker, R. C., Breen, D.S., Akhtar, Y. N.,
Kenwood, B. M., Liao, J. A., Zhang, M., Fazakerley,
D.J., Tomsig, J.L., Harris, T.E., Keller, S.R., Chow, J.D.,
Lynch, K. R., Chokki, M., Molkentin, J. D., Turner, N.,
James, D. E., Yan, Z., and Hoehn, K. L. (2013) Open-
ing of the mitochondrial permeability transition pore
links mitochondrial dysfunction to insulin resistance
in skeletal muscle, Mol. Metab., 3, 124-134, https://
doi.org/10.1016/j.molmet.2013.11.003.
140. Belosludtsev, K. N., Starinets, V. S., Talanov, E. Y.,
Mikheeva, I. B., Dubinin, M. V., and Belosludtseva,
N.V. (2021) Alisporivir treatment alleviates mitochon-
drial dysfunction in the skeletal muscles of C57BL/
6NCrl mice with high-fat diet/streptozotocin-induced
diabetes mellitus, Int.J. Mol. Sci., 22, 9524, https://
doi.org/10.3390/ijms22179524.
141. Starinets, V. S., Serov, D. A., Penkov, N. V., Beloslud-
tseva, N. V., Dubinin, M. V., and Belosludtsev, K. N.
(2022) Alisporivir normalizes mitochondrial function
of primary mouse lung endothelial cells under con-
ditions of hyperglycemia, Biochemistry (Moscow), 87,
605-616, https://doi.org/10.1134/S0006297922070033.
142. Belosludtsev, K.N., Serov, D.A., Ilzorkina, A.I., Stari-
nets, V. S., Dubinin, M. V., Talanov, E. Y., Karagyaur,
M. N., Primak, A. L., and Belosludtseva, N. V. (2023)
Pharmacological and genetic suppression of VDAC1
alleviates the development of mitochondrial dys-
function in endothelial and fibroblast cell cultures
upon hyperglycemic conditions, Antioxidants (Basel),
12, 1459, https://doi.org/10.3390/antiox12071459.
143. Zhu, X., Qin, Z., Zhou, M., Li, C., Jing, J., Ye, W., and
Gan, X. (2024) The role of mitochondrial permea-
bility transition in bone metabolism, bone healing,
and bone diseases, Biomolecules, 14, 1318, https://
doi.org/10.3390/biom14101318.
144. Sautchuk, R., Kalicharan, B. H., Escalera-Rivera, K.,
Jonason, J.H., Porter, G.A., Awad, H.A., and Eliseev,
R.A. (2022) Transcriptional regulation of cyclophilinD
BELOSLUDTSEV et al.1810
BIOCHEMISTRY (Moscow) Vol. 90 No. 12 2025
by BMP/Smad signaling and its role in osteogenic dif-
ferentiation, Elife, 11, e75023, https://doi.org/10.7554/
eLife.75023.
145. Shares, B.H., Smith, C.O., Sheu, T.J., Sautchuk,R.,Jr.,
Schilling,K., Shum, L.C., Paine,A., Huber,A., Gira,E.,
Brown, E., Awad, H., and Eliseev, R. A. (2020) Inhi-
bition of the mitochondrial permeability transition
improves bone fracture repair, Bone, 137, 115391,
https://doi.org/10.1016/j.bone.2020.115391.
146. He,Y., Zhang,L., Zhu,Z., Xiao,A., Yu,H., and Gan,X.
(2017) Blockade of cyclophilinD rescues dexametha-
sone-induced oxidative stress in gingival tissue, PLoS
One, 12, e0173270, https://doi.org/10.1371/journal.
pone.0173270.
147. He, P., Liu, F., Li, M., Ren, M., Wang, X., Deng, Y.,
Wu, X., Li, Y., Yang, S., and Song, J. (2023) Mitochon-
drial calcium ion nanogluttons alleviate periodon-
titis via controlling mPTPs, Adv. Health. Mater., 12,
e2203106, https://doi.org/10.1002/adhm.202203106.
148. Bonora, M., and Pinton, P. (2014) The mitochondrial
permeability transition pore and cancer: molecular
mechanisms involved in cell death, Front. Oncol., 4,
302, https://doi.org/10.3389/fonc.2014.00302.
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.