ISSN 0006-2979, Biochemistry (Moscow), 2024, Vol. 89, Nos. 12-13, pp. 2083-2106 The Author(s) 2024. This article is an open access publication.
2083
REVIEW
Troponins and Skeletal Muscle Pathologies
Agnessa P. Bogomolova
1,2,a
* and Ivan A. Katrukha
1,2,b
1
Faculty of Biology, Lomonosov Moscow State University, 119234 Moscow, Russia
2
Hytest Ltd., Turku, Finland
a
e-mail: bogomolova.agnessa@yandex.ru 
b
e-mail: katrukhai@mail.ru
Received May 2, 2024
Revised November 19, 2024
Accepted December 1, 2024
AbstractSkeletal muscles account for ~30-40% of the total weight of human body and are responsible for
its most important functions, including movement, respiration, thermogenesis, and glucose and protein me-
tabolism. Skeletal muscle damage negatively impacts the whole-body functioning, leading to deterioration of
the quality of life and, in severe cases, death. Therefore, timely diagnosis and therapy for skeletal muscle
dysfunction are important goals of modern medicine. In this review, we focused on the skeletal troponins
that are proteins in the thin filaments of muscle fibers. Skeletal troponins play a key role in regulation of
muscle contraction. Biochemical properties of these proteins and their use as biomarkers of skeletal muscle
damage are described in this review. One of the most convenient and sensitive methods of protein biomarker
measurement in biological liquids is immunochemical analysis; hence, we examined the factors that influence
immunochemical detection of skeletal troponins and should be taken into account when developing diagnostic
test systems. Also, we reviewed the available data on the skeletal troponin mutations that are considered
tobe associated with pathologies leading to the development of diseases and discussed utilization of troponins
as drug targets for treatment of the skeletal muscle disorders.
DOI: 10.1134/S0006297924120010
Keywords: skeletal muscle, troponin, ageing, myopathy, biomarker, monoclonal antibodies
Abbreviations: ALS, amyotrophic lateral sclerosis; cTnI, cardiac troponin I; cTnT, cardiac troponin T; fsTnC, fast skeletal
troponin C; fsTnI, fast skeletal troponinI; fsTnT, fast skeletal troponinT; ss/cTnC, slow skeletal/cardiac troponin C; ssTnI,
slow skeletal troponin I; ssTnT, slow skeletal troponin T.
* To whom correspondence should be addressed.
INTRODUCTION
In mammals, striated muscles are represented by
two tissue types: skeletal and cardiac. Skeletal mus-
cles accounts for ~30-40% of the total body weight and
perform important functions such as movement, res-
piration, and thermogenesis [1, 2]. They are involved
in the major share of glucose, lipid, and protein me-
tabolism. Thus, skeletal muscle damage or pathology
could have a significant impact on various aspects of
whole-body functioning, leading to impaired quality of
life and, in severe cases, disability or death.
Skeletal muscle dysfunction can be caused by
mechanical injuries, myopathies, and various diseases
that are accompanied by muscle atrophy and rhab-
domyolysis. The most common causes of mechanical
damage to skeletal muscles include vigorous physical
activity, surgery, trauma, and prolonged compression
syndrome  [3]. The term “myopathy” describes any
skeletal muscle disease of various etiologies. These pa-
thologies are frequently accompanied by abnormalities
of the skeletal muscle structure and development of
metabolic disorders in the tissue [4]. Myopathies are
classified as either congenital (inherited) or acquired
(secondary). Inherited myopathies include: muscular
dystrophies (associated with mutations in the genes
of certain cytoskeletal proteins of skeletal muscle, e.g.,
Duchenne muscular dystrophy or Becker muscular
dystrophy); congenital myopathies (e.g., nemaline my-
opathy, core myopathy); metabolic myopathies (associ-
ated with mutations in the genes of various enzymes
involved in carbohydrate and lipid metabolism);
BOGOMOLOVA, KATRUKHA2084
BIOCHEMISTRY (Moscow) Vol. 89 Nos. 12-13 2024
mitochondrial myopathies (associated with mutations
in mitochondrial oxidative phosphorylation proteins);
and channelopathies [5-8]. Muscular dystrophies are
considered as one of the most common and severe in-
herited myopathies, some of which, such as Duchenne
muscular dystrophy, are characterized by progressive
muscle weakness and atrophy, eventually leading
to death from cardiopulmonary arrest [9]. Acquired
myopathies include: inflammatory myopathies (most
commonly of autoimmune nature, such as derma-
tomyositis and polymyositis), myopathies associated
with various infections (caused by bacteria, fungi, or
viruses), toxic myopathies (e.g., caused by drug or al-
cohol poisoning or myotoxic drugs such as statins and
fibrates), myopathies associated with various diseases
(rheumatic diseases, malignant tumors, and endocrine
diseases), idiopathic myopathies, and endocrine myop-
athies [10-13].
Another serious pathology of skeletal muscle is
rhabdomyolysis. It is the process of skeletal muscle
breakdown accompanied by the release of intracel-
lular components into the bloodstream, which can
be life-threatening in severe cases. When more than
100  g of muscle tissue is simultaneously damaged,
concentration of myoglobin in the blood reaches a
critical value, and a significant amount of protein is
deposited in renal tubules. This process could provoke
acute renal failure, which is the main cause of fatal
outcomes in rhabdomyolysis [12, 14]. In children, the
major factors leading to rhabdomyolysis are viral my-
ositis, trauma, rheumatic diseases, high-intensity exer-
cise, and medications  [12]. In adults, rhabdomyolysis
most often occurs due to alcohol or drug abuse, med-
ications, trauma, malignant neuroleptic syndrome, or
immobility [5, 12].
Myopathies are accompanied by muscle atrophy,
a process of loss of muscle mass and strength when
catabolism of muscle proteins predominates over
anabolism. In addition to myopathies, other diseases
and pathologies such as chronic heart failure, chronic
obstructive pulmonary disease, cancer, chronic kidney
disease, Alzheimers disease, and infectious diseases
could also lead to the development of muscle atro-
phy [15, 16]. Muscle atrophy could also be caused by
immobility and muscle disuse due to paralysis or pro-
longed bed rest [3].
A loss of muscle mass also occurs during the
ageing process – sarcopenia [from Greek words sarx
(flesh) and penia (deficiency)]. This condition is man-
ifested by the decrease in the cross-sectional area and
number of muscle fibers, accumulation of adipose or
connective tissue in the skeletal muscle, and progres-
sive loss of the muscle mass and strength. All of this
entails poor physical performance and, if prolonged,
geriatric frailty [17]. Sarcopenia is characterized by
the increased risk of soreness, falls, limitations in daily
activities, poor prognosis after surgery and, ultimately,
high mortality [16]. Prevalence of sarcopenia reaches
a level up to 13% in the individuals aged 60-70 years
and up to 50% in the individuals aged over 80 [17].
Presence of diabetes increases the risk of sarcopenia
by 2-fold, and of geriatric frailty – by 1.5-4-fold [18].
Thus, importance of these problems grows with the
increase in life expectancy. To date, nutritional adjust-
ment and exercise have been used to stop progression
of sarcopenia, but also therapeutic strategies are being
developed to prevent muscle damage [16-18].
Skeletal muscle is the primary tissue responsible
for insulin-dependent glucose uptake; hence, loss of
muscle mass in sarcopenia could lead to the develop-
ment of insulin resistance, and ultimately type  2 dia-
betes. In addition, the skeletal muscle adiposis, which
accompanies sarcopenia, could also contribute to dys-
function [17, 19]. In sarcopenia and type  2 diabetes,
functional impairments could develop necessitating
diagnosis and timely treatment. So, monitoring the de-
crease in skeletal muscle mass is of high importance,
especially in the patients with diabetes.
Three types of skeletal muscle fibers have been
identified in humans: slow-twitch (type  1) and fast-
twitch (types 2A and 2X; other mammals also have
type  2B fast-twitch fibers) [20, 21]. The type  2A fast-
twitch muscle fibers are more resistant to fatigue than
the type  2X and contain more enzymes of oxidative
metabolism  [22]. Fiber composition of muscles de-
pends on the function they perform: muscles respon-
sible for maintaining body posture are mainly com-
posed of slow-twitch fibers, while muscles required
for movement are predominantly composed of fast-
twitch fibers [21].
Development of some pathologies could result in
the damage to muscle fibers of only certain type. For
example, predominantly fast-twitch muscle fibers are
affected, especially type 2X fibers, in the Duchenne
muscular dystrophy [23]. Muscle damage induced by ad-
ministration of statins primarily influences fast-twitch
fibers, while fibrates intake impacts slow-twitch fibers
[24,  25]. Intense eccentric muscle contractions also
primarily damage fast-twitch fibers  [26]. Many patho-
logical conditions entailing muscle atrophy are char-
acterized not only by deterioration of certain muscle
fiber types, but also by a fiber-shift from one type to
another. For instance, denervation or immobility of a
limb, spinal cord injury, or prolonged bed rest may
lead to slow-to-fast fiber type shift  [27]. The reverse
process, fast-to-slow fiber type shift, occurs as a result
of starvation, glucocorticoid administration, cachexia,
and sarcopenia [27]. In addition, fast-twitch and slow-
twitch muscle fibers have been shown to have differ-
ent abilities to regenerate after injury. Studies in rats
have shown that while muscles consisting mainly of
fast-twitch fibers (e.g., extensor digitorum) regenerate
TROPONINS AND SKELETAL MUSCLE PATHOLOGIES 2085
BIOCHEMISTRY (Moscow) Vol. 89 Nos. 12-13 2024
efficiently, muscles composed predominantly of slow-
twitch fibers (e.g., soleus) do not regenerate complete-
ly, developing fibrosis [28].
Various instrumental methods have been used
to diagnose skeletal muscle injuries. These include
magnetic resonance imaging, computed tomography,
and dual-energy X-ray absorptiometry [29]. These ap-
proaches allow non-invasive detection of changes in
the skeletal muscles (edema, replacement with adipose
and connective tissue, muscle atrophy), identification
of specific areas of damage, and even quantification
of changes in the structure of skeletal muscles [30-34].
However, disadvantage of using instrumental methods
is the need for expensive equipment and specialized
locations for manipulation. An alternative is determi-
nation of blood concentrations of biomarkers – most
commonly skeletal muscle proteins– that are released
into the bloodstream when muscle fibers are dam-
aged. Currently, the most widely used biomarkers of
skeletal muscle damage are creatine kinase, aspartate
aminotransferase, lactate dehydrogenase, and myo-
globin  [35]. However, it should be noted that all the
abovementioned proteins lack specificity: in addition
to skeletal muscle, they are expressed in other tissues,
that may reduce accuracy of the diagnosis. Therefore,
special studies have been conducted to seek and iden-
tify new and more specific biomarkers. These includ-
ed fatty acid binding protein  3, myosin light chain 3,
MM isoform of creatine kinase, and skeletal troponin I
(TnI) isoforms  [36]. Although all the above biomarkers
are comparable or superior in sensitivity to conven-
tional biomarkers, only the skeletal TnI isoforms are
specifically expressed in skeletal muscle [9, 36, 37].
TnI, together with troponin  C (TnC) and troponin  T
(TnT), constitutes the troponin complex that regulates
muscle contraction [38, 39]. TnT isoforms are also spe-
cific to skeletal muscle, but we were unable to find
information on their use as biomarkers of skeletal
muscle damage. The utilization of skeletal muscle TnT
as a biomarker requires further investigation.
For a long time, skeletal troponins have attracted
attention of the scientists, primarily due to the studies
reporting their role in regulation of muscle contrac-
tion. In this review, we describe role of these proteins
in the development of certain skeletal muscle diseas-
es and discuss applications of troponins for diagnosis
and therapy of skeletal muscle disorders.
BIOCHEMICAL PROPERTIES OF THE TROPONINS
Troponin I. In humans, TnI is represented by
three isoforms: cardiac TnI (cTnI) and two skeletal
isoforms, namely fast skeletal (fsTnI) and slow skele-
tal (ssTnI). fsTnI and ssTnI are expressed in fast-twitch
and slow-twitch muscle fibers, respectively (Table 1)
[23,  40]
1
. ssTnI is also expressed in cardiac muscle
during embryonic development and is replaced by
cTnI in the postnatal period [41]. cTnI is only ex-
pressed in cardiac muscle after birth [42, 43].
TnI inhibits interaction between myosin and actin
in the absence of Ca
2+
. In the presence of Ca
2+
, the in-
hibitory region of TnI dissociates from actin and this
facilitates actin-myosin interaction [44, 45]. The major
difference between the skeletal TnI isoforms and cTnI
is the presence of a unique 31 amino acid N-terminal
sequence (residues 2-32) in cTnI (Fig.  1). Phosphory-
lation of the Ser23 and Ser24 residues is considered
as a mechanism regulating muscle contraction [48].
For the skeletal TnI isoforms, the three-dimensional
(3D) structure has been resolved for fsTnI only. The
highest resolution 3D structure was obtained using
X-ray crystallography for the chicken fsTnI in complex
with TnC and for the portion of TnT in both Ca
2+
-ac-
tivated state (3.00  Å resolution) and Ca
2+
-free state
(7.00  Å resolution)  [46]. The 3D structure has been
resolved for the full-length fsTnI with the exception
of C-terminal part of the molecule. Several function-
al sites could be distinguished in the TnI structure:
the IT arm, inhibitory, switch, and C-terminal regions.
Similar sites for ssTnI have been denoted based on
their sequence similarity with fsTnI and cTnI [46,  47].
1
Numbering in all proteins of this review includes N-terminal Met.
Table 1. Some properties of human skeletal TnI isoforms
Isoforms
Fast skeletal TnI
(UniProt P48788)
Slow skeletal TnI
(UniProt P19237)
Gene TNNI2 TNNI1
Chromosomal
locus
11p15.5 1q32.1
Number
of exons
89
Number
of amino acid
residues
182 187
Molecular
mass (kDa)
21.3 21.7
Isoelectric
point
8.9 9.6
Tissue
specificity
fast-twitch
muscle fibers
slow-twitch
muscle fibers,
embryonic
cardiomyocytes
BOGOMOLOVA, KATRUKHA2086
BIOCHEMISTRY (Moscow) Vol. 89 Nos. 12-13 2024
Fig. 1. Protein sequence alignment of three TnI isoforms: fsTnI, ssTnI and cTnI. Amino acid sequences are derived from
the Uniprot database: fsTnI (TNNI2_HUMAN, P48788), ssTnI (TNNI1_HUMAN, P19237), cTnI (TNNI3_HUMAN, P19429). Clustal
Omega online software was used to make protein sequence alignments. Identical amino acid residues in the aligned se-
quences are colored in blue. Boxes show the borders of α-helices (numbering is for fsTnI sequence). Arrows indicate regions
and TnC- and TnT-binding sites, putative fsTnI and ssTnI phosphorylation (p) and fsTnI glutathionylation (glut) sites are
marked above the sequence [44-47].
The IT arm of TnI (~2-106 amino acid residue of
fsTnI; ~2-107 amino acid positions of ssTnI; number-
ing varies in different animal species) has a structur-
al function: it consists of the N-terminal region that
binds the C-terminal domain of TnC (~2-40 residues
of fsTnI; ~2-40 residues of ssTnI) and the TnT-bind-
ing site (~50-106 amino acid residues of fsTnI; ~50-107
residues of ssTnI). The IT-arm is formed by two op-
positely directed α-helices, H1 (~12-48 amino acids of
fsTnI; ~12-48 residues of ssTnI) and H2 (~58-103 res-
idues of fsTnI; ~59-104 residues of ssTnI), connected
by a linker (~49-57 residues of fsTnI; ~49-58 residues
of ssTnI). The inhibitory region (~107-115 residues of
fsTnI; ~108-116 residues of ssTnI) interacts with ac-
tin in the absence of Ca
2+
ions, moving tropomyosin
molecule to the position where it blocks the inter-
action of myosin with actin thus preventing muscle
contraction. The switch region (~116-131 residues of
fsTnI; ~117-132 residues of ssTnI) includes the H3
α-helix (~118-127 residues of fsTnI; ~119-128 residues
of ssTnI), it binds to the N-terminal domain of TnC
when Ca
2+
concentration increases, which leads to dis-
sociation of the inhibitory region of TnI from actin, dis-
placement of tropomyosin, and interaction of myosin
with actin.
The C-terminal portion of fsTnI could not be re-
solved by X-ray crystallography due to its mobility.
The nuclear magnetic resonance (NMR) data for the
chicken fast skeletal troponin complex shows that the
C-terminal region lacks secondary structure and is in
a disordered state, both in the absence and presence
of Ca
2+
[49, 50]. According to the results of small-angle
neutron scattering of the chicken fast skeletal tropo-
nin complex, the C-terminal part of fsTnI is an elon-
gated structure at low Ca
2+
concentrations that may
represent a supercoiled helix or β-layers, while in the
presence of Ca
2+
the site adopts a compact structure
[51]. Other NMR data suggest that the C-terminal re-
gion of the chicken fsTnI has a secondary structure
consisting of an α-helix, two β-layers, and two more
α-helices [52-54]. The C-terminal region interacts with
actin in the absence of Ca
2+
and thus participates in
inhibition of myosin binding to actin [52, 55, 56].
Troponin T. Like TnI, TnT is represented in the
human body by three isoforms: a cardiac (cTnT) and
two TnT skeletal isoforms, namely fast skeletal (fsTnT)
and slow skeletal (ssTnT) variants (Table 2). fsTnT and
ssTnT are only expressed in skeletal muscle, while cTnT
is expressed in both heart and skeletal muscle during
embryonic development and neonatal period [57, 58].
TROPONINS AND SKELETAL MUSCLE PATHOLOGIES 2087
BIOCHEMISTRY (Moscow) Vol. 89 Nos. 12-13 2024
Table 2. Some properties of human TnT isoforms
Isoforms
Fast skeletal TnT
(UniProt P45378)
Slow skeletal TnT
(UniProt P13805)
Gene TNNT3 TNNT1
Chromosomal
locus
11p15.5 19q13.42
Number
of exons
19 14
Number
of amino acid
residue
245-269 251-278
Molecular
mass (kDa)
29.1-31.8 30-33
Isoelectric
point
5.7-9 5.6-6.1
Tissue
specificity
skeletal muscles skeletal muscles
The function of TnT is to attach the troponin
complex to the actin and regulate the interaction of
the complex with thin filament proteins [59]. The TnT
molecule includes an N-terminal variable region and
conserved central and C-terminal regions (Fig. 2). The
N-terminal domain of TnT is variable not only in ami-
no acid composition but also in length, and diversi-
ty of the forms is the result of alternative splicing.
The conserved portion has been shown to contain
two α-helices, H1 (~162-188 residues of fsTnT) and
H2 (~196-240 residues of fsTnT). The H2 TnT α-helix
forms a supercoiled helix with H2 of TnI and interacts
with TnC via its C-terminus [46, 47, 60-62]. The con-
served portion also includes two tropomyosin bind-
ing sites, Tm1 and Tm2. While localization of Tm1
is determined with sufficient accuracy (~61-99 resi-
dues of fsTnT; ~65-103 residues of ssTnT), the data
on localization of the Tm2 are controversial. Jin and
Chong [60] presumed that Tm2 is located within the
~161-200 residues of fsTnT and ~165-204 residues of
ssTnT, whereas it was suggested in the other works
that Tm2 is formed by the last C-terminal residues of
TnT [63-66]. Studies describing structure of the cardiac
thin filament obtained with cryo-electron microscopy
(cryo-EM) and cryo-electron tomography (cryo-ET)
have suggested that cTnT is capable of binding two
Tm strands simultaneously. With the Tm2 site TnT in-
teracts with the same strand as TnI does, while with
the Tm1 region it binds to the neighboring thread of
tropomyosin [67, 68]. Investigation of the structure
of cardiac [67,  68] and skeletal muscle thin filaments
[69] by cryo-EM and cryo-ET also suggested that fsTnT
is likely to interact with the skeletal muscle protein
nebulin, which, like tropomyosin, binds to actin along
the entire thin filament. Although the exact structure
of skeletal TnT and nebulin has still not been resolved,
it is assumed that the R134-R179 portion of the mouse
fsTnT (R123-R169 in human fsTnT) has two nebulin
binding sites [69].
The TNNT3 gene encoding fsTnT consists of 19 ex-
ons. Eight exons among them can undergo alternative
splicing: namely exons 4, 5, 6, 7, 8, and the fetal exon
in the N-terminal portion of the molecule, as well as
exons 16 and 17 (or exons α and β) encoding a part
of the C-terminus [71]. The fetal exon, located between
the exons 8 and 9, is expressed in embryonic skeletal
muscle only [70,  72]. Expression of the exons 16 and
17 is mutually exclusive: exon 16 is expressed pre-
dominantly in adulthood, while exon 17 is expressed
primarily in embryonic and neonatal muscles [73,  74].
During embryonic development and in postnatal peri-
od, expression of the fsTnT splice variants is changing:
high molecular weight variants are replaced with low
molecular weight splice forms, low isoelectric point
(acidic) variants are replaced with high isoelectric
point (basic) splice forms (see below). The differences
in size and charge of these forms are due to alter-
native splicing of the N-terminal exons that encode
predominantly acidic residues [75].
In the adult rabbit muscle, expression of five
splice variants of fsTnT has been demonstrated by
Western blotting: dominant TnT1f (the longest splice
form, containing all exons except the fetal one), TnT2f,
TnT3f, and to a lesser extent TnT2fa and TnT4f [72,
76, 77]. It is important to note, however, that the dif-
ferences in the splice variant composition have been
observed in various muscle types. In the adult rat, six
splice variants of fsTnT have been identified by mass
spectrometry, and their composition also differed in
various muscle types  [78]. The same method detect-
ed six splice variants of fsTnT in the rhesus macaque
[79]. In the adult humans, only three fsTnT splice vari-
ants (fsTnT III, VI, and VII) have been identified by
the muscle top-down mass spectrometry to date, but
in that study, the authors analyzed only two types of
muscles tibialis anterior and vastus lateralis – the
former of which contains more slow-twitch fibers, and
so some of the fsTnT splice variants may not have
been detected [80].
The TNNT1 gene encoding ssTnT consists of 14
exons. Unlike fsTnT, diversity of the splice forms
resulting from alternative splicing of ssTnT is low.
Presence of high molecular weight and low molecu-
lar weight splice variants of ssTnT has been shown by
Western blotting for mouse and sheep  [81]. Cloning of
the mouse cDNA demonstrated that the low molecular
weight ssTnT form appears due to alternative splic-
ing deletion of 11 residues of the N-terminal exon  5.
BOGOMOLOVA, KATRUKHA2088
BIOCHEMISTRY (Moscow) Vol. 89 Nos. 12-13 2024
Fig. 2. Protein sequence alignment of three TnT isoforms: fsTnT, ssTnT and cTnT. Amino acid sequences are derived from
the Uniprot database: fsTnT (TNNT3_HUMAN, P45378-3), ssTnT (TNNT1_HUMAN, P13805-1), cTnT (TNNT2_HUMAN, P45379-6).
The longest adult human splice variants were chosen; for fsTnT and cTnT, we selected those variants that do not contain
the fetal exon. Clustal Omega online software was used to make sequence alignments. The identically positioned amino acid
residues are marked with green. fsTnT and ssTnT exon structures are represented above. Boxes show borders of α-helices
(residues numeration is for fsTnT sequence), TnI and TnC binding site is marked above. Solid line oval indicates tropomy-
osin Tm1 binding site, dashed line ovals indicate putative tropomyosin Tm2 binding sites and potential nebulin binding
site. Possible fsTnT phosphorylation sites are marked above the sequence [46, 47, 60, 63-66, 69, 70].
Two high molecular weight ssTnT splice forms are
distinguished by a single residue deletion in the
exon  6 [81,  82]. Cloning of the human cDNA also re-
vealed three splice variants of ssTnT: a full-length
splice form, a splice form with a C-terminal deletion
of 16 residues derived from alternative splicing of the
exon 12, and a splice form with two deletions, the
aforementioned C-terminal deletion of the exon 12
and an N-terminal deletion of 11 residues (exon  5),
as in the mouse ssTnT [82-84]. At the protein level,
two splice variants of ssTnT (ssTnT II and III) with
deletions were identified in the human skeletal muscle
(vastus lateralis and tibialis anterior) by mass spec-
trometry [80].
Troponin  C. In contrast to TnI and TnT, TnC is
represented in humans by two isoforms: the slow
skeletal/cardiac isoform ss/cTnC and the fast skeletal
isoform fsTnC, which are expressed in the cardiac/
slow-twitch and fast-twitch muscle fibers, respective-
ly (Table 3) [85].
TnC provides the sensitivity of thin filament to
Ca
2+
. It changes conformation of the troponin complex
after binding of Ca
2+
, whose concentration increases
due to propagation of the nerve impulse along the
muscle fiber. TnC consists of a short N-terminal region
and four EF-hands (Ca
2+
-binding domains) (Fig.  3).
These four EF-hands are joined in pairs to form N-ter-
minal and C-terminal domains connected by a linker.
TROPONINS AND SKELETAL MUSCLE PATHOLOGIES 2089
BIOCHEMISTRY (Moscow) Vol. 89 Nos. 12-13 2024
Table 3. Some properties of human TnC isoforms
Isoforms
Fast skeletal TnC
(UniProt P02585)
Slow skeletal/
cardiac TnC
(UniProt P63316)
Gene TNNC2 TNNC1
Chromosomal
locus
20q13.12 3p21.1
Number
of exons
66
Number
of amino acid
residue
160 161
Molecular
mass (kDa)
18.1 18.4
Isoelectric
point
4.1 4.0
Tissue
specificity
fast-twitch
muscle fibers
slow-twitch
muscle fibers,
cardiomyocytes
The N-terminal EF-hands (EF-hands I and II) have
low affinity, while the C-terminal hands (EF-hands
III and IV) have high affinity for Ca
2+
. TheC-terminal
EF-hands are constantly saturated with Ca
2+
or Mg
2+
ions, whereas the N-terminal ones bind Ca
2+
with
high selectivity only when its concentration increases
during propagation of action potential along the length
of the muscle fiber. In the fsTnC, all four EF-hands can
bind ions whereas in the ss/cTnC, the EF-hand  I has
lost this ability due to a single residue insertion and
substitution of two residues [47, 62, 89].
The 3D protein structure of fsTnC had been de-
ciphered by X-ray crystallography for rabbit fsTnC
[86], for chicken fsTnC [46] alone and as a part of
the troponin complex for four Ca
2+
-binding and two
Ca
2+
-binding states. Although the ss/cTnC isoform is
expressed in slow-twitch muscle fibers, most of the
studies on its 3D structure have been focused on its
place and role in the human cardiac troponin com-
plex [47].
The C-terminal globular domain of TnC is a part
of the IT arm that interacts with other troponins in
the supercoiled helix region (H2 TnI and H2 TnT)
and H1 helix of TnI. The N-terminal and C-terminal
Fig. 3. Protein sequence alignment of two TnC isoforms: fsTnC and ss/cTnC. Amino acid sequences are derived from the
Uniprot database: fsTnC (TNNC2_HUMAN, P02585), ss/cTnC (TNNC1_HUMAN, P63316). The identically positioned amino acid
residues are colored yellow. Amino acid residues that participate in binding of Ca
2+
and Mg
2+
ions are marked with bold.
Boxes show the borders of α-helices (N, A-H). Arrows indicate N-terminal and C-terminal domains and EF-hands. Grey
circles indicate the sites of Ca
2+
and Mg
2+
binding, solid line circles stand for a site that is specific to binding Ca
2+
ions
in both TnC isoforms, dashed circle stands for the site of Ca
2+
binding in fsTnC isoform (the first EF hand in ss/cTnC does
not bind ions) [46, 47, 62, 86-89].
BOGOMOLOVA, KATRUKHA2090
BIOCHEMISTRY (Moscow) Vol. 89 Nos. 12-13 2024
globular domains are connected by the central linker
[46, 47]. Although the TnC protein C-domain is always
occupied by ions, its N-domain can switch between
two states: closed conformation in the absence of Ca
2+
or open conformation initiated by Ca
2+
binding to its
EF-hand(s). Binding of Ca
2+
ions induces formation of
a hydrophobic cleft that binds to the switch region of
TnI and moves the adjacent inhibitory region of TnI
away from actin [87, 88].
FUNCTIONAL DIFFERENCES
BETWEEN THE TROPONIN ISOFORMS
Different fibers of striated muscle – fast-twitch,
slow-twitch, and cardiac – vary in their response to
elevated Ca
2+
concentrations. The slow-twitch fibers
have the highest Ca
2+
sensitivity (threshold of Ca
2+
concentration for activation is lower), while the fast-
twitch fibers have the lowest Ca
2+
sensitivity [21, 90,
91]. The highest cooperativity of contraction (Hill co-
efficient) is observed in the fast-twitch fibers and the
lowest in the slow-twitch fibers. Evidence suggests
that these differences could be mediated by the tro-
ponin isoforms expressed in different fiber types. Re-
placement of ss/cTnC with fsTnC in the cardiac fibers
decreases Ca
2+
sensitivity, while substitution of fsTnC
with ss/cTnC in the fast skeletal fibers increases it [90].
Replacement of the whole troponin complex in the
fast-twitch skeletal fibers with cardiac troponins re-
sults in the increased sensitivity to Ca
2+
and decreased
cooperativity of contraction [92].
Decrease of intracellular pH (acidosis) leads to
contractile dysfunction in all types of striated mus-
cle and involves reduced Ca
2+
sensitivity. In the heart
muscle, reduction in intracellular pH to 6.5 and below
is observed during prolonged ischemia and is caused
by accumulation of metabolic products, including lac-
tate [93, 94]. In the skeletal muscle, decrease of pH is
observed during exhaustive exercise, and although it
does affect skeletal muscle, the effect is not as signif-
icant as in the heart muscle. In the human skeletal
muscle, pH drops down to ~6.5 after high-intensity ex-
ercise, with typical average values of ~6.8-6.9 [95-97].
As with the response to Ca
2+
, the extent of pH decrease
varies between the muscle fiber types, being most pro-
nounced in the cardiac muscle and least prominent in
the slow-twitch fibers [90, 98]. Experiments with iso-
lated muscle fibers have shown that when pH drops
from 7.0 to 6.5, the smallest decrease in Ca
2+
sensi-
tivity is observed in the slow-twitch muscle fibers:
change in Ca
2+
sensitivity is greater in the fast-twitch
and cardiac fibers, and it is approximately equal in
amplitude [90]. Moreover, further reduction in pH to
6.2 results in the strongest drop in Ca
2+
sensitivity in
the cardiac fibers, but not in the skeletal muscle.
Sensitivity of the contractile apparatus to acidosis
is largely determined by the troponin complex, and
magnitude of this effect is determined, among other
things, by different troponin isoforms [99]. In par-
ticular, experiments performed with isolated mus-
cle fibers have shown that replacing of fsTnC with
ss/cTnC in the fast-twitch fibers leads to an increase
in pH sensitivity, and, conversely, replacing of ss/cTnC
with fsTnC in the slow-twitch fibers leads to a slight
decrease in pH sensitivity [100]. The latter effect is
even more pronounced when ss/cTnC is replaced with
fsTnC in the cardiac fibers  [99]. Thus, the ss/cTnC in-
creases pH-dependence and fsTnC decreasesit. Itwas
shown in another study that combinations of TnC
and TnI affect pH sensitivity  [90]. The authors isolat-
ed cardiac fibers and made substitutions of TnI and
TnC together and separately. In the fibers with cTnI
or fsTnI, ss/cTnC accounted for the increased pH sen-
sitivity compared to fsTnC. Moreover, replacement
of cTnI with ssTnI resulted in the reduced pH sensi-
tivity regardless of which TnC isoform was present.
Thus, the authors concluded that TnC is responsible
for pH sensitivity in the fast-twitch or cardiac fibers,
while TnI performs that function in the slow-twitch
fibers [90]. These findings are in good agreement with
the fact that the neonatal cardiomyocytes expressing
ssTnI are less sensitive to acidosis than the adult car-
diomyocytes expressing cTnI [101]. This effect has
been reproduced when cTnI was replaced with ssTnI
in the adult transgenic mice: sensitivity to acidosis
was reduced [102]. More recent studies have shown
that the effect could be associated with the residue
His131 (His131 in fsTnI, His132 in ssTnI) that is pres-
ent in ssTnI and fsTnI, but is replaced with Ala163
in cTnI. This residue is localized in the switch region
of TnI and participates in the interaction with TnC.
It has been shown that at lower pH, electrostatic in-
teraction between the His131 and Glu15 and Glu19
of TnC stabilizes conformation of the troponin com-
plex, leading to activation of the muscle contraction
[103-106].
Three isoforms of TnT differ in their isoelectric
points, with the adult fsTnT being the most basic iso-
form. Moreover, there is an acidic-to-basic fsTnT splice
variant shift that ensures expression of the basic splice
forms in the adult muscle. These differences are due
to the sequence variation in the TnT N-terminal re-
gion. There is a unique acidic fsTnT splice variant that
is synthesized in some of the adult chicken muscle
types  [107]. These muscles show lower sensitivity to
Ca
2+
as well as higher tolerance to acidosis compared
to the other muscle types that express basic fsTnT
splice forms [108]. These findings are consistent with
the fact that expression of fsTnT in the transgenic
mouse hearts lowers tolerance of the cardiac muscle
to acidosis [109].
TROPONINS AND SKELETAL MUSCLE PATHOLOGIES 2091
BIOCHEMISTRY (Moscow) Vol. 89 Nos. 12-13 2024
SKELETAL TROPONINS AS BIOMARKERS
OF SKELETAL MUSCLE DAMAGE
One of the most sensitive and specific approaches
to detect and measure concentration of a biomarker is
the sandwich immunoassay [110]. In most cases, this
method is based on the use of two antibodies that rec-
ognize epitopes on the antigen surface, one of which
is an immobilized capture antibody and the other is
a detection antibody, conjugated with a label. To our
knowledge, there have been no registered immuno-
assays to detect skeletal TnI isoforms and all existing
studies have employed “research use only” assays.
Concentrations of the skeletal TnI isoforms in the
healthy individuals have been determined in a num-
ber of studies. The mean values of basal TnI concen-
trations have ranged between 1.74  ±  0.27  ng/mL and
2.5  ±  0.9  ng/mL when measured by the same method
in different studies [111-114]. In another study, con-
centrations of the skeletal TnI isoforms in the healthy
individuals were below the limit of detection (LOD)
(LOD  =  1.2  ng/mL)  [115]. In the studies where fsTnI
concentration was determined, its basal concentra-
tion was below the LOD of the detection method
(2.4  ng/mL) [9, 116]. Although further clarification is
needed, these data suggest that the median blood lev-
els of skeletal TnI isoforms in the healthy cohort are
below 1-2 ng/mL.
In cases of skeletal muscle damage, integrity of
the muscle fiber membrane is compromised, and in-
tracellular proteins are released into the bloodstream.
It has been shown that the circulating levels of skel-
etal TnI isoforms increase during skeletal muscle im-
pairment of various etiologies. In particular, the skele-
tal TnI isoform concentrations rise in the patients with
rhabdomyolysis [117, 118]; as well as after intensive
physical exercises: for example, after triathlon [113],
running [119], eccentric contractions [26, 120-122];
and in orthopedic and soft tissue injuries, including
those resulting from surgical operations [114, 119].
Elevated concentrations of the skeletal TnI isoforms
have also been observed in the patients with inflam-
matory myopathies such as polymyositis and dermato-
myositis [115, 119, 123]. Concentration of the skeletal
TnI isoforms correlates with the clinical picture in
muscular dystrophies (Duchenne, Becker, and limb-
girdle muscular dystrophy), and therefore it is pos-
sible to use this parameter to assess severity of the
disease and monitor response to the therapy [9].
One of the potential applications of skeletal TnI
isoforms as a biomarker is in determination of myo-
toxicity of drugs, where muscle fibers are destroyed
and troponin is released into the bloodstream [36,
37, 124, 125]. Feasibility of this application has been
demonstrated in rats as well as using 3D human mus-
cle tissue in  vitro [126, 127].
Presence of the TnI isoforms specific for different
types of skeletal fibers provides additional diagnostic
possibilities. As described in the previous section, a
number of skeletal muscle injuries/diseases could af-
fect a particular type of muscle fiber. For example,
eccentric muscle contractions damage predominant-
ly fast-twitch type fibers, and it is fsTnI rather than
ssTnI that is released into the bloodstream [26, 121,
122]. The increased level of fsTnI, but not the level of
ssTnI concentrations have also been shown for some
muscular dystrophies [128].
Maximum concentration of the skeletal TnI iso-
forms reached values of 500  ng/mL 6  h after complet-
ing a triathlon (4  km of swimming, 120  km bike-rid-
ing, 30  km running), with a median concentration of
62.2  ±  139  ng/mL [113]. Also, high skeletal TnI isoform
concentrations were reached 6 h after downhill run-
ning: the median value was 27.3  ng/mL (8.5-43  ng/mL
interquartile range), while lower concentrations were
found 6  h after level running: 6.6  ng/ml (3.7-9  ng/mL)
and after eccentric contractions of the quadriceps fem-
oris muscle: 6.8  ng/ml (3.1-14.9  ng/mL) [26]. Mean con-
centration levels of the skeletal TnI isoforms within
24  h of injury were: 15.3  ±  2.4  ng/mL after orthopedic
injury and 10.4  ±  1.8  ng/mL after soft tissue injury
[114]. Maximum concentration of the skeletal TnI iso-
forms in the patients with inflammatory myopathies
was 516  ng/mL and the median value was 8.6  ng/mL
(3.2-33.5  ng/mL interquartile range) [115].
We were not able to find any studies devoted to
the utilization of skeletal TnT isoforms as possible bio-
markers of muscle damage. Nevertheless, we assume
that, like cardiac troponinsI and T, which are markers
of cardiac muscle damage, the skeletal TnT isoforms,
along with the skeletal TnI isoforms, may also be a
potential marker for diagnosing skeletal muscle injury.
fsTnC has also been proposed as a biomarker
of skeletal muscle deterioration [129]. However, TnC
recognition would have less specificity than detection
of the skeletal TnI or TnT isoforms because fsTnC is
expressed in the fast-twitch muscle fibers only, while
c/ssTnC is expressed in the slow-twitch fibers and car-
diomyocytes.
FACTORS INFLUENCING IMMUNOCHEMICAL
DETECTION OF SKELETAL TROPONINS
Various factors could affect interaction of anti-
bodies with skeletal troponins. These include homol-
ogy with other troponin isoforms, presence of splice
variants, post-translational modifications, proteolysis,
autoantibodies binding, and shielding of epitopes by
proteins of the troponin complex. Such effects could
significantly distort the results of the measurements
and should be taken into account when studying
BOGOMOLOVA, KATRUKHA2092
BIOCHEMISTRY (Moscow) Vol. 89 Nos. 12-13 2024
the biochemical properties of the troponins and de-
veloping immunochemical systems.
Homology of isoforms. The degree of similar-
ity between the isoforms of human TnI and TnT is
~60% for the both TnI and TnT isoforms (when com-
paring the longest splice variants expressed in the
adults: P45378-3 for fsTnT, P13805-1 for ssTnT, and
P45379-6 for cTnT) (Figs. 1 and 2). Therefore, one of
the intricacies of immunochemical detection of skele-
tal troponins is cross-reactivity of antibodies with the
cardiac isoform of the protein or with the second skel-
etal isoform (in the case of differential detection of
slow- or fast-twitch skeletal troponin), and this could
reduce specificity of detection of the target biomarker.
High degree of troponin homology significantly reduc-
es the number of epitopes to which antibodies could
be generated for the specific detection of individual
protein isoforms. The most conserved sites for both
TnI and TnT are located in the C-terminal parts of
these proteins, while the most variable sites are in
their N-terminal portions.
Alternative splicing of TnT. As described above,
the skeletal TnT isoforms, especially fsTnT, are repre-
sented in the human muscle by several splice variants.
To develop immunodetection test systems that would
recognize all possible splice forms of fsTnT, antibod-
ies specific to the domains encoded by exons 1-3 and
9-15 should be utilized. For the fsTnT, antibodies spe-
cific to the N-terminal residues 11-38 as well as the
C-terminal region (residues 231-244 that may differ
between splice variants) should be avoided (residues
numbering is indicated for the sequence accession
number P45378-3). In the case of ssTnT, antibodies
recognizing residues 25-35 and 205-220 (numbering
is for the longest splice variant; sequence accession
number P13805-1), should be avoided because these
sequences may differ between the splice variants.
TnI and TnT phosphorylation. Phosphorylation
is a modification that changes charge of a protein and
could have significant effect on the antibody-antigen
interaction.
TnI phosphorylation. The rabbit fsTnI could be
phosphorylated by phosphorylase kinase, 3′,5′-cAMP-de-
pendent protein kinase, Ca
2+
-phospholipid-dependent
protein kinase, and the human fsTnI by AMP-acti-
vated protein kinase in vitro [130-134]. The residues
that undergo phosphorylation have been identified as
Thr12 and Ser118 for the rabbit fsTnI and Ser118 for
the human protein [131, 132, 134]. Presence of the
Thr12 residue in the human fsTnI sequence as well
as in the rabbit fsTnI provides the possibility of its
phosphorylation in the human protein as well. It has
been shown that phosphorylation in  vitro is inhibited
by interaction with TnC, and this is consistent with
the data on the structure of troponin complex: both
of these residues are located in the sites of interac-
tion with TnC [46, 130]. Phosphorylation of ssTnI has
not been described in vitro, but there is one residue
homologous to the fsTnI phosphorylation site in the
ssTnI sequence, namely Ser118 (ssTnI) (Fig. 1).
The fsTnI isolated from the rabbit skeletal mus-
cle is present in the partially phosphorylated form,
and the degree of phosphorylation could depend on
the method of protein isolation [130, 131, 135]. The
fsTnI from the human muscle extract (vastus lateralis
and tibialis anterior) and rat muscle extract (7 muscle
types) analyzed by mass spectrometry were found to
be in the non-phosphorylated form, whereas the ssTnI
was present in both phosphorylated and non-phos-
phorylated forms [78, 80]. No phosphorylated forms of
the skeletal TnI isoforms were detected by mass spec-
trometry of the rhesus macaque muscle extract [79].
Based on the above data, it could be expected that
the proportion of phosphorylated forms of skeletal TnI
isoforms in blood would be negligible and, therefore,
would not affect their recognition by antibodies.
TnT phosphorylation. Using in vitro experimen-
tation, the fsTnT isolated from the rabbit skeletal
muscle has been shown to be a substrate for various
kinases including phosphorylase kinase, 3′,5′-cAMP-de-
pendent protein kinase, casein kinase2, and Ca
2+
-phos-
pholipid-dependent protein kinase [59, 130, 132, 133,
136-140]. Three phosphorylation sites have been iden-
tified for the rabbit fsTnT: Ser2, Ser152, and Ser159
(numbers of residues are indicated for Uniprot acces-
sion number P45372-3). Ser2 is also represented in
the human fsTnT, while the Ser152 and Ser159 resi-
dues are in composition of the Ala-Leu-Ser-Ser-SerP-
Met-Gly-Ala-Asn-Tyr-Ser-SerP-Tyr sequence, which in
the rabbit fsTnT is completely consistent with the
human protein. This suggests the possibility of phos-
phorylation of these residues in the human fsTnT
as well [132].
Meanwhile, in a series of in  vivo studies, fsTnT
isolated from the rabbit skeletal muscle has been
shown to be present in a solely monophosphorylated
form [130, 132]. Moreover, phosphorylation occurred
at only one of the three residues identified in  vitro:
Ser2. These results are consistent with the mass spec-
trometry data for the proteins extracted from the
muscle tissue of various species. For human, rat, and
rhesus macaque fsTnT samples isolated from several
muscle types, unphosphorylated and monophosphor-
ylated forms (present in different splice variants) of
fsTnT were identified by mass spectrometry [78-80]
and Ser2 was the only site of phosphorylation deter-
mined in vivo.
There are no data on phosphorylation of the ssTnT
in  vitro. However, ssTnT contains Ser2 homologous to
the Ser2 in the fsTnT and cTnT, for which phosphor-
ylation has been shown both in  vitro and in  vivo [141,
142]. High probability of the ssTnT phosphorylation
TROPONINS AND SKELETAL MUSCLE PATHOLOGIES 2093
BIOCHEMISTRY (Moscow) Vol. 89 Nos. 12-13 2024
at Ser2 in the human muscle tissue was confirmed
by mass spectrometry [80]. Using this method, it was
found that in the human and rat skeletal muscle,
ssTnT was presented in a predominantly phosphory-
lated form, and in the rhesus macaque muscle in both
phosphorylated and non-phosphorylated forms [78-80].
It could be assumed that fsTnT and ssTnT are re-
leased into the bloodstream in a partially phosphory-
lated form, but phosphorylation site is located in the
terminal part of the protein and, therefore, could be
presumed not to affect immunochemical detection by
antibodies.
TnI glutathionylation. Lamb et al. showed in a
series of studies on isolated rat and human muscle
fibers that fsTnI, but not ssTnI, could undergo gluta-
thionylation [143-145]. This modification occurred at
one of the cysteine residues, Cys134. This is consistent
with the mass spectrometry data for the protein: two
proteoforms of fsTnI, with and without glutathione,
were detected in the skeletal muscles of rhesus ma-
caque and rat, although, abundance of the glutathio-
nylated form was quite low [78, 79].
TnI and TnT proteolysis. N-terminal and C-ter-
minal proteolysis has been demonstrated for cTnI:
various proteolyzed forms of cTnI were observed in
both necrotic cardiac tissue and in blood [146, 147].
cTnI was found to be a substrate of a number of pro-
teases: metalloproteinase-2, calpain 1 (µ-calpain) and
calpain  2 (m-calpain) [148-151]. Studies of the cTnI
proteolysis in blood of the patients with myocardial
infarction showed presence of at least 11 proteolyt-
ic fragments of different lengths [147, 152]. Due to
the sequence homology to cTnI, proteolysis of skeletal
TnI isoforms is also possible. The proteolyzed fsTnI
and ssTnI and their fragments have been identified
in the blood of patients, and amount of these frag-
ments in the blood varies depending on the type of
injury/disease [118]. Based on these results, the au-
thors suggested that proteolysis occurs directly in the
tissue and that such proteolyzed forms are released
into the bloodstream. Further studies on proteolysis of
the skeletal TnI isoforms and localization of the sites
of proteolysis are needed.
cTnT also undergoes proteolysis. In the apoptot-
ic cardiomyocytes, N-terminal cleavage of cTnT by
caspase-3 with formation of a 25-kDa fragment has
been demonstrated [153]. cTnT is also prone to limited
N-terminal proteolysis by calpain 1 (µ-calpain) during
myocardial ischemia-reperfusion [148, 154]. In the
blood of patients with myocardial infarction, at least
23 cTnT fragments of different lengths and carrying
several sites of proteolysis in the N-terminal, central,
and C-terminal parts of the molecule were found [155].
Similar studies have not yet been performed for the
skeletal TnT isoforms. For fsTnT, proteolysis has been
demonstrated only in the muscles after vigorous exer-
cise [156]. Further investigations are required to iden-
tify the forms of skeletal TnT isoforms present in the
bloodstream and to localize their sites of proteolysis.
Shielding of epitopes in the troponin complex.
cTnI and cTnT are present in the blood of patients
with myocardial infarction as part of a ternary tropo-
nin complex (TnI‒TnT‒TnC), a binary troponin com-
plex (TnI‒TnC), and as proteolytic fragments of TnT
[152, 157, 158]. Although there is no experimental data
describing the form in which skeletal troponins are
released into the blood, it can be assumed that they
would also be present in the samples as complexes.
For immunochemical detection, it is important to use
antibodies that recognize troponin epitopes that are
present in all forms that exist in blood. The skeletal
TnT isoforms could be covered and shielded from de-
tection by TnI and TnC at their binding sites, limited
to approximately 197-241 residues for fsTnT. In the
case of the skeletal TnI isoforms, a significant portion
of the molecules could be shielded from detection at
binding sites located at approximately 2-40, 50-106,
and 116-131 residues for fsTnI, and 2-40, 51-107, and
117-132 residues for ssTnI. Thus, small regions in the
central and C-terminal part of TnI molecule could
remain accessible to antibodies. In order to uncov-
er the epitopes shielded by other troponins in the
central part of TnI, an alternative method could be
to dissociate the troponin complex using EDTA [115,
116, 157].
Autoantibodies binding. One of the factors in-
fluencing the immunochemical detection of cardiac
troponins is the presence of autoantibodies that in-
teract with the analyzed protein in the blood. They
might interfere with the antibodies in composition of
the immunochemical test systems for detection of the
protein and lead to false negative results. It has been
demonstrated that approximately 10% of the healthy
individuals have autoantibodies to cTnI in their blood
[159]. These immunoglobulins have been shown to be
primarily specific for the conformational epitopes that
are formed when TnI and TnT combine into a com-
plex [160]. There are no studies on the presence of
autoantibodies interacting with the skeletal isoforms
of TnI; however, considering high degree of homolo-
gy, it can be assumed that they could also be present
in the blood of some patients.
Finally, we examined the sites that could be most
susceptible to various factors interfering with recogni-
tion of troponins by antibodies. The C-terminal regions
of TnI could be associated with the cross-reactivity
displayed by antibodies to other TnI isoforms, while
the central parts of the molecule could be shielded by
TnC or TnT. The effect of posttranslational modifica-
tions such as phosphorylation and glutathionylation is
considered to be negligible, while the role and extent
of proteolysis and influence of autoantibodies needs
BOGOMOLOVA, KATRUKHA2094
BIOCHEMISTRY (Moscow) Vol. 89 Nos. 12-13 2024
further investigation. Taking into account the above
factors, the most promising sites for the detection of
skeletal TnI isoforms are central parts of the mole-
cule, especially those that are not shielded by other
troponins.
In the case of TnT, the N-terminal (11-38 residues
for fsTnT, 25-35 residues for ssTnT) and C-terminal
(231-244 residues for fsTnT and 205-220 residues for
ssTnT) parts of the molecule are alternatively spliced,
while the C-terminal parts (~197-241 residues for
fsTnT) could be screened by TnI and TnC. The effect of
phosphorylation is considered to be negligible, while
the role and extent of N- and C-terminal proteolysis
needs further investigation. Therefore, central parts
of the skeletal TnT isoforms could be considered as
the most promising sites for their immunochemical
detection.
MUTATIONS IN TROPONINS AS CAUSES
OF SKELETAL MUSCLE PATHOLOGIES
The troponin complex is of high importance for
regulation of muscle contraction. Mutations could lead
to the changes in protein structure and functionality.
To date, a number of mutations have been identified
that are associated with the development of such skel-
etal muscle diseases as arthrogryposis and congenital
myopathies.
Mutations in the skeletal TnI genes. Mutations
in the TNNI2 gene (fsTnI) have been demonstrat-
ed to be one of the causes of distal arthrogryposis.
This is an autosomal dominant disease, in which
joints are affected and mobility of distal limbs is re-
duced. The TNNI2 mutations associated with distal
arthrogryposis are present in the following locations
within the C-terminal region of the molecule: R156X
[leading to expression of a truncated form of fsTnI
(1-156)], R162G, R162K, I165F, E167X, K168E, R174Q,
R174W, K175N, K175X, K176X, F178C, and F178L
[161-172].
For ssTnI (TNNI1), two pathogenic mutations have
been discovered to date [170,  173]. One is located in
the same region as mutations in the fsTnI and is as-
sociated with the development of proximal arthrogry-
posis, an autosomal dominant disease in which prox-
imal joints are affected. This mutation, K175X, results
in the expression of a truncated form of ssTnI [170].
Mutation in the N-terminal region of ssTnI, R37C, is
associated with expression of ssTnI in the heart for up
to two years after birth [173] that increases the risk
of sudden infant death.
Mutations in the skeletal TnT genes. Mutations
in the TNNT3 gene (fsTnT) lead to the development
of distal arthrogryposis. For TNNT3, several substi-
tutions affecting the same residue have been found:
R66C, R66H, and R66S [171, 174-177]
2
. Itis also worth
highlighting the TNNT3 (fsTnT) mutation, which caus-
es two diseases simultaneously: the aforementioned
distal arthrogryposis and the nemaline myopathy that
affects fast-twitch muscle fibers [178]. It is a muta-
tion in an intron that leads to the impaired splicing
and decreased expression of fsTnT, with compensa-
tory hypertrophy of slow-twitch muscle fibers being
observed.
Deletions in the TNNT1 gene (ssTnT) are associ-
ated with nemaline myopathy inherited in an autoso-
mal recessive pattern. This disease is manifested in
early childhood as a respiratory failure, slow-twitch
muscle fiber atrophy, and compensatory hypertrophy
of fast-twitch muscle fibers. Ultimately, nemaline my-
opathy leads to death in childhood from the respira-
tory failure. E180X (expression of the truncated form
of ssTnT 1-179), S108X (expression of the truncated
form of ssTnT 1-107), deletion of the exons 8 and 14,
and expression of the truncated form of ssTnT (1-203)
have been found [179-182]. These deletions affect tro-
pomyosin binding sites as well as TnI and TnC binding
sites.
Absence of the troponin and tropomyosin bind-
ing sites in ssTnT after E180X deletion impedes for-
mation of the troponin complex and its incorporation
into thin filaments [183]. While the ssTnT (1-179)
mRNA is still detectable in muscle fibers, the ssTnT
(1-179) protein molecule degrades rapidly and can no
longer be detected [184]. This fact explains recessive
nature of the mutation: one copy of the gene is suf-
ficient for expression of the full-length ssTnT and its
incorporation into the troponin complex, while ex-
pression of the mutant ssTnT (1-179) protein is not
cytotoxic, as the truncated ssTnT is immediately de-
graded [185].
Mutations in the TnC genes. Mutations in the
TNNC2 gene (fsTnC) are associated with congenital
myopathy inherited in an autosomal dominant pat-
tern. These mutations comprise the D34Y and M79Y
substitutions, which are located in the EF-hand  I and
near EF-hand  II, respectively. They affect Ca
2+
bind-
ing and interaction with TnI switch region, so there is
reduced Ca
2+
sensitivity in the presence of these mu-
tations [186, 187].
Since ss/cTnC is expressed in the slow-twitch skel-
etal muscle as well as in the cardiac muscle, mutations
in TNNC1 (ss/cTnS) are associated with cardiomy-
2
To specify this mutation, the authors of the studies used the fsTnT sequence corresponding to UniProt accession number
P45378-2, which is three residues shorter than the P45378-3 sequence used in this review. Therefore, this substitution
appears in the articles as R63.
TROPONINS AND SKELETAL MUSCLE PATHOLOGIES 2095
BIOCHEMISTRY (Moscow) Vol. 89 Nos. 12-13 2024
opathies and cardiac dysfunction. These genetic alter-
ations include missense mutations A8V, L29Q, A31S,
C84Y, E134D, and D145E, and frameshift mutations
Q122AfsX30 [188-191]. Along with cardiomyopathies,
some of the patients carrying these mutations also
exhibit skeletal muscle abnormalities, but more re-
search is needed to understand which mutations af-
fect skeletal muscle and exact mechanism involved in
the process [187, 192]. It was shown in the in vitro
experiments that all of the abovementioned mutations
except for C84Y had no effect on phenotype and func-
tion of the slow-twitch skeletal muscle fibers, while
the C84Y mutation increased Ca
2+
sensitivity, as it does
in the cardiac muscle fibers. These differences in the
effects of the ss/cTnC mutations on slow-twitch and
cardiac muscle fibers could be due to the fact that in
the slow-twitch muscle fibers ss/cTnC forms complex
with the slow-twitch skeletal but not cardiac isoforms
of TnT and TnI [193].
TROPONINS AS DRUG TARGETS
In addition to being the source of pathologies,
skeletal troponins could also serve as drug targets
used in treatment of skeletal muscle diseases. These
include peripheral motor neuropathies– amyotrophic
lateral sclerosis (ALS), spinal muscle atrophy, Charcot–
Marie–Tooth disease – in which the motor neurons
are damaged, and nerve impulse transmission is
impaired, and also myasthenia gravis, in which the
neuromuscular junction is impaired. Disruption of
the skeletal muscle innervation ultimately leads to
muscle weakness, disability, and high mortality. One
of the therapeutic approaches for these pathologies is
to increase sensitivity of the muscle fibers to Ca
2+
, so
that sufficient muscle activation is achieved even at
low Ca
2+
concentrations. A molecule from the group
of first fast skeletal troponin activators, CK-2017357
(tirasemtiv), selectively interacts with the fast skeletal
troponin complex: binding occurs in the hydrophobic
pocket formed by the N-terminal domain of fsTnC
and the switch region of fsTnI [194]. Tirasemtiv bind-
ing has been shown to increase the degree of affin-
ity of low-affinity Ca
2+
binding sites on fsTnC, there-
by increasing sensitivity of the fsTnC to Ca
2+
[195].
Tirasemtiv has shown promising results in the patients
with ALS, patients with myasthenia gravis, and studies
even reached phase III clinical trials, but were halted
due to the discovery of side effects [196-202]. Today,
other drugs from the group of fast skeletal muscle
troponin activators, namely CK-2127107, CK-2066260,
and reldesemtiv are under development [203-206].
Reldesemtiv, a second-generation molecule that was
derived by optimization of tirasemtiv and has similar
mechanism of action, has been most successful [207,
208]. Reldesemtiv has reached phase II clinical trials
in spinal muscular atrophy and phaseIII in ALS, but,
unfortunately, trials on the patients with ALS were
stopped due to the lack of therapeutic success [206,
209, 210]. Although the ss/cTnC is expressed in both
slow-twitch skeletal and cardiac muscles, regulators of
the ss/cTnC Ca
2+
sensitivity have been studied primar-
ily in the context of modulating heart function. These
drug molecules regulate Ca
2+
sensitivity of muscle con-
traction by adjusting the Ca
2+
-binding affinity of the
N-terminal domain of ss/cTnC. One of the most wide-
ly studied calcium sensitizers, levosimendan, binds
to the hydrophobic pocket formed by the N-terminal
domain of ss/cTnC and the switch region of cTnI– in
a manner similar to that of tirasemtiv [211]. Levosi-
mendan is approved for use in some countries, and
its analogs are intended to be used for treatment of
the heart diseases associated with depressed cardiac
function.
In contrast to the above drug molecules, the tro-
ponin inhibitor W7 acts by decreasing Ca
2+
sensitivity,
and it could be used to treat contractile dysfunction
arising from the alkalosis-induced and inherited car-
diomyopathies [212, 213]. W7 also binds to the hy-
drophobic pocket between the N-terminal domain of
ss/cTnC and the cTnI switch region, but, unlike levosi-
mendan, it shifts the cTnI switch region from its bind-
ing site, which leads to the reduced Ca
2+
sensitivity
[213, 214].
CONCLUSION
Skeletal muscle dysfunction has a negative impact
on the whole-body functioning and could be caused
by mechanical muscle injury, myopathies, and other
diseases accompanied by muscle atrophy, as well as by
rhabdomyolysis. Determination of the concentration
of protein biomarkers released into the bloodstream
when muscle fibers are damaged is a convenient
method of diagnosing and monitoring such pathol-
ogies. Skeletal troponins are potential candidates to
be used as biomarkers of skeletal muscle dysfunction
including various myopathies, traumas, and injuries
caused by vigorous physical exercise, as well as for
assessing severity of muscular dystrophies and deter-
mining myotoxicity of the drugs.
Structure and properties of troponins could af-
fect accuracy of their antibody-based detection in hu-
man blood. Main factors influencing immunochem-
ical detection of these proteins in clinical samples
are isoform homology, alternative splicing of TnT,
post-translational modifications (phosphorylation and
glutathionylation), proteolysis, shielding of TnI and
TnT epitopes within the complex, and autoantibodies
binding. Therefore, a careful selection of monoclonal
BOGOMOLOVA, KATRUKHA2096
BIOCHEMISTRY (Moscow) Vol. 89 Nos. 12-13 2024
antibodies is required for the development of immu-
noassays capable of reliable quantitative detection of
these proteins.
Certain mutations in skeletal troponins are associ-
ated with the development of arthrogryposis and con-
genital myopathies. Furthermore, skeletal troponins
could be used as drug targets for treatment of muscle
diseases such as peripheral motor neuropathies and
myasthenia gravis.
Open access. This article is licensed under a Cre-
ative Commons Attribution4.0 International License,
which permits use, sharing, adaptation, distribution,
and reproduction in any medium or format, as long
as you give appropriate credit to the original au-
thor(s) and the source, provide a link to the Creative
Commons license, and indicate if changes were made.
Theimages or other third party material in this article
are included in the article’s Creative Commons license,
unless indicated otherwise in a credit line to the mate-
rial. If material is not included in the article’s Creative
Commons license and your intended use is not permit-
ted by statutory regulation or exceeds the permitted
use, you will need to obtain permission directly from
the copyright holder. To view a copy of this license,
visit https://creativecommons.org/licenses/by/4.0/.
Contributions. A.P.B. and I.A.K. collected and an-
alyzed the data, prepared illustrations, wrote the man-
uscript, edited the manuscript; I.A.K. conceived and
supervised the study.
Funding. This work was supported by ongoing in-
stitutional funding. No additional grants to carry out
or direct this particular research were obtained.
Ethics declarations. This work does not con-
tain any studies involving human and animal sub-
jects. Theauthors of this work declare that they have
noconflicts of interest.
REFERENCES
1. Yin,L., Li,N., Jia,W., Wang,N., Liang,M., Yang,X., and
Du, G. (2021) Skeletal muscle atrophy: from mecha-
nisms to treatments, Pharmacol. Res., 172, 105807,
https://doi.org/10.1016/j.phrs.2021.105807.
2. Janssen,I., Heymsfield, S.B., Wang, Z.M., and Ross,R.
(2000) Skeletal muscle mass and distribution in 468
men and women aged 18-88 yr, J. Appl. Physiol., 89,
81-88, https://doi.org/10.1152/jappl.2000.89.1.81.
3. Kasper, C. E., Talbot, L. A., and Gaines, J. M. (2002)
Skeletal muscle damage and recovery, AACN Clin.
Issues, 13, 237-247, https://doi.org/10.1097/00044067-
200205000-00009.
4. Nagy, H., and Veerapaneni, K. D. (2023) Myopathy, in
StatPearls, StatPearls Publishing LLC., Treasure Is-
land(FL).
5. Nance, J. R., and Mammen, A. L. (2015) Diagnostic
evaluation of rhabdomyolysis, Muscle Nerve, 51, 793-
810, https://doi.org/10.1002/mus.24606.
6. Cassandrini, D., Trovato, R., Rubegni, A., Lenzi, S.,
Fiorillo, C., Baldacci, J., Minetti, C., Astrea, G.,
Bruno, C., and Santorelli, F. M. (2017) Congenital
myopathies: clinical phenotypes and new diagnostic
tools, Ital. J. Pediatr., 43, 101, https://doi.org/10.1186/
s13052-017-0419-z.
7. Ahmed, S.T., Craven,L., Russell, O.M., Turnbull, D.M.,
and Vincent, A.E. (2018) Diagnosis and treatment of
mitochondrial myopathies, Neurotherapeutics, 15,
943-953, https://doi.org/10.1007/s13311-018-00674-4.
8. Cohen, B. H. (2019) Mitochondrial and metabol-
ic myopathies, Continuum, 25, 1732-1766, https://
doi.org/10.1212/con.0000000000000805.
9. Burch, P.M., Pogoryelova,O., Goldstein,R., Bennett,D.,
Guglieri, M., Straub, V., Bushby, K., Lochmüller, H.,
and Morris,C. (2015) Muscle-derived proteins as se-
rum biomarkers for monitoring disease progression
in three forms of muscular dystrophy, J.Neuromuscul.
Dis., 2, 241-255, https://doi.org/10.3233/jnd-140066.
10. Dalakas, M.C. (2015) Inflammatory muscle diseases,
N.Engl. J. Med., 372, 1734-1747, https://doi.org/10.1056/
NEJMra1402225.
11. Crum-Cianflone, N.F. (2008) Bacterial, fungal, parasit-
ic, and viral myositis, Clin. Microbiol. Rev., 21, 473-494,
https://doi.org/10.1128/cmr.00001-08.
12. Torres, P.A., Helmstetter, J.A., Kaye, A.M., and Kaye,
A.D. (2015) Rhabdomyolysis: pathogenesis, diagnosis,
and treatment, Ochsner.J., 15, 58-69.
13. du Souich, P., Roederer, G., and Dufour, R. (2017)
Myotoxicity of statins: Mechanism of action, Phar-
macol Ther, 175, 1-16, https://doi.org/10.1016/
j.pharmthera.2017.02.029.
14. Khan, F.Y. (2009) Rhabdomyolysis: a review of the lit-
erature, Neth.J. Med., 67, 272-283.
15. Fanzani, A., Conraads, V. M., Penna, F., and Marti-
net,W. (2012) Molecular and cellular mechanisms of
skeletal muscle atrophy: an update, J. Cachexia Sar-
copenia Muscle, 3, 163-179, https://doi.org/10.1007/
s13539-012-0074-6.
16. Jasuja, R., and LeBrasseur, N. K. (2014) Regener-
ating skeletal muscle in the face of aging and dis-
ease, Am. J. Phys. Med. Rehabil., 93, S88-S96, https://
doi.org/10.1097/phm.0000000000000118.
17. Sinclair, A. J., Abdelhafiz, A. H., and Rodríguez-
Mañas, L. (2017) Frailty and sarcopenia - newly
emerging and high impact complications of diabe-
tes, J. Diabetes Complications, 31, 1465-1473, https://
doi.org/10.1016/j.jdiacomp.2017.05.003.
18. Jang, H. C. (2019) Diabetes and muscle dysfunction
in older adults, Ann. Geriatr. Med. Res., 23, 160-164,
https://doi.org/10.4235/agmr.19.0038.
19. Cleasby, M. E., Jamieson, P. M., and Atherton, P. J.
(2016) Insulin resistance and sarcopenia: mechanistic
TROPONINS AND SKELETAL MUSCLE PATHOLOGIES 2097
BIOCHEMISTRY (Moscow) Vol. 89 Nos. 12-13 2024
links between common co-morbidities, J.Endocrinol.,
229, R67-R81, https://doi.org/10.1530/joe-15-0533.
20. Schiaffino,S., and Reggiani,C. (1994) Myosin isoforms
in mammalian skeletal muscle, J. Appl. Physiol., 77,
493-501, https://doi.org/10.1152/jappl.1994.77.2.493.
21. Schiaffino, S., and Reggiani, C. (2011) Fiber types in
mammalian skeletal muscles, Physiol. Rev., 91, 1447-
1531, https://doi.org/10.1152/physrev.00031.2010.
22. Murgia, M., Toniolo, L., Nagaraj, N., Ciciliot, S.,
Vindigni,V., Schiaffino,S., Reggiani,C., and Mann,M.
(2017) Single muscle fiber proteomics reveals fi-
ber-type-specific features of human muscle aging,
Cell Rep., 19, 2396-2409, https://doi.org/10.1016/
j.celrep.2017.05.054.
23. Murgia, M., Nogara, L., Baraldo, M., Reggiani, C.,
Mann, M., and Schiaffino, S. (2021) Protein profile
of fiber types in human skeletal muscle: a single-fi-
ber proteomics study, Skelet Muscle, 11, 24, https://
doi.org/10.1186/s13395-021-00279-0.
24. Westwood, F. R., Bigley, A., Randall, K., Marsden,
A. M., and Scott, R. C. (2005) Statin-induced muscle
necrosis in the rat: distribution, development, and
fibre selectivity, Toxicol. Pathol., 33, 246-257, https://
doi.org/10.1080/01926230590908213.
25. Bodié, K., Buck, W. R., Pieh, J., Liguori, M. J., and
Popp, A. (2016) Biomarker evaluation of skeletal
muscle toxicity following clofibrate administration
in rats, Exp. Toxicol. Pathol., 68, 289-299, https://
doi.org/10.1016/j.etp.2016.03.001.
26. Sorichter,S., Mair, J., Koller, A., Gebert,W., Rama,D.,
Calzolari,C., Artner-Dworzak,E., and Puschendorf,B.
(1997) Skeletal troponin I as a marker of exercise-in-
duced muscle damage, J.Appl. Physiol., 83, 1076-1082,
https://doi.org/10.1152/jappl.1997.83.4.1076.
27. Ciciliot, S., Rossi, A. C., Dyar, K. A., Blaauw, B., and
Schiaffino,S. (2013) Muscle type and fiber type speci-
ficity in muscle wasting, Int.J. Biochem. Cell Biol., 45,
2191-2199, https://doi.org/10.1016/j.biocel.2013.05.016.
28. Zimowska,M., Kasprzycka,P., Bocian,K., Delaney,K.,
Jung,P., Kuchcinska,K., Kaczmarska, K., Gladysz,D.,
Streminska,W., and Ciemerych, M.A. (2017) Inflam-
matory response during slow- and fast-twitch mus-
cle regeneration, Muscle Nerve, 55, 400-409, https://
doi.org/10.1002/mus.25246.
29. Tosato,M., Marzetti,E., Cesari,M., Savera,G., Miller,
R. R., Bernabei, R., Landi, F., and Calvani, R. (2017)
Measurement of muscle mass in sarcopenia: from im-
aging to biochemical markers, Aging Clin. Exp. Res.,
29, 19-27, https://doi.org/10.1007/s40520-016-0717-0.
30. Mercuri, E., Clements, E., Offiah, A., Pichiecchio, A.,
Vasco, G., Bianco, F., Berardinelli, A., Manzur, A.,
Pane, M., Messina,S., Gualandi,F., Ricci, E., Ruther-
ford,M., and Muntoni,F. (2010) Muscle magnetic res-
onance imaging involvement in muscular dystrophies
with rigidity of the spine, Ann. Neurol., 67, 201-208,
https://doi.org/10.1002/ana.21846.
31. Van De Vlekkert, J., Maas, M., Hoogendijk, J. E., De
Visser, M., and Van Schaik, I. N. (2015) Combining
MRI and muscle biopsy improves diagnostic accuracy
in subacute-onset idiopathic inflammatory myopa-
thy, Muscle Nerve, 51, 253-258, https://doi.org/10.1002/
mus.24307.
32. Willcocks, R. J., Rooney, W.D., Triplett, W.T., Forbes,
S. C., Lott, D.J., Senesac, C. R., Daniels, M. J., Wang,
D. J., Harrington, A. T., Tennekoon, G. I., Russman,
B.S., Finanger, E.L., Byrne, B.J., Finkel, R.S., Walter,
G.A., Sweeney, H.L., and Vandenborne,K. (2016) Mul-
ticenter prospective longitudinal study of magnetic
resonance biomarkers in a large duchenne muscular
dystrophy cohort, Ann. Neurol., 79, 535-547, https://
doi.org/10.1002/ana.24599.
33. Burakiewicz, J., Sinclair, C. D. J., Fischer, D., Walter,
G. A., Kan, H. E., and Hollingsworth, K. G. (2017)
Quantifying fat replacement of muscle by quantita-
tive MRI in muscular dystrophy, J.Neurol., 264, 2053-
2067, https://doi.org/10.1007/s00415-017-8547-3.
34. Krššák,M., Lindeboom,L., Schrauwen-Hinderling,V.,
Szczepaniak, L.S., Derave,W., Lundbom,J., Befroy,D.,
Schick,F., Machann,J., Kreis,R., and Boesch,C. (2021)
Proton magnetic resonance spectroscopy in skeletal
muscle: experts’ consensus recommendations, NMR
Biomed., 34, e4266, https://doi.org/10.1002/nbm.4266.
35. Brancaccio,P., Lippi, G., and Maffulli,N. (2010) Bio-
chemical markers of muscular damage, Clin. Chem.
Lab. Med., 48, 757-767, https://doi.org/10.1515/
cclm.2010.179.
36. Goldstein, R. A. (2017) Skeletal muscle injury bio-
markers: assay qualification efforts and translation
to the clinic, Toxicol. Pathol., 45, 943-951, https://
doi.org/10.1177/0192623317738927.
37. Burch, P.M., Greg Hall,D., Walker, E.G., Bracken,W.,
Giovanelli,R., Goldstein, R., Higgs, R.E., King, N. M.,
Lane,P., Sauer, J.M., Michna,L., Muniappa,N., Pritt,
M.L., Vlasakova,K., Watson, D.E., Wescott,D., Zabka,
T.S., and Glaab, W.E. (2016) Evaluation of the relative
performance of drug-induced skeletal muscle injury
biomarkers in rats, Toxicol. Sci., 150, 247-256, https://
doi.org/10.1093/toxsci/kfv328.
38. Katrukha, I. A. (2013) Human cardiac troponin
complex. Structure and functions, Biochemistry
(Moscow), 78, 1447-1465, https://doi.org/10.1134/
S0006297913130063.
39. Sheng, J. J., and Jin, J. P. (2016) TNNI1, TNNI2 and
TNNI3: evolution, regulation, and protein struc-
ture-function relationships, Gene, 576, 385-394, https://
doi.org/10.1016/j.gene.2015.10.052.
40. Dhoot, G. K., and Perry, S. V. (1979) Distribution of
polymorphic forms of troponin components and tro-
pomyosin in skeletal muscle, Nature, 278, 714-718,
https://doi.org/10.1038/278714a0.
41. Sasse, S., Brand, N. J., Kyprianou, P., Dhoot, G. K.,
Wade, R., Arai, M., Periasamy, M., Yacoub, M. H.,
BOGOMOLOVA, KATRUKHA2098
BIOCHEMISTRY (Moscow) Vol. 89 Nos. 12-13 2024
and Barton, P. J. (1993) Troponin I gene expression
during human cardiac development and in end-
stage heart failure, Circ. Res., 72, 932-938, https://
doi.org/10.1161/01.RES.72.5.932.
42. Bodor, G.S., Porterfield,D., Voss, E.M., Smith,S., and
Apple, F.S. (1995) Cardiac troponin-I is not expressed
in fetal and healthy or diseased adult human skele-
tal muscle tissue, Clin. Chem., 41, 1710-1715, https://
doi.org/10.1093/clinchem/41.12.1710.
43. Dellow, K.A., Bhavsar, P.K., Brand, N.J., and Barton,
P.J. (2001) Identification of novel, cardiac-restricted
transcription factors binding to a CACC-box with-
in the human cardiac troponin I promoter, Cardio-
vasc. Res., 50, 24-33, https://doi.org/10.1016/S0008-
6363(01)00204-8.
44. Filatov, V. L., Katrukha, A. G., Bulargina, T. V., and
Gusev, N. B. (1999) Troponin: structure, properties,
and mechanism of functioning, Biochemistry (Mos-
cow), 64, 969-985.
45. Gomes, A. V., Potter, J. D., and Szczesna-Cordary, D.
(2002) The role of troponins in muscle contrac-
tion, IUBMB Life, 54, 323-333, https://doi.org/
10.1080/15216540216037.
46. Vinogradova, M. V., Stone, D. B., Malanina, G. G.,
Karatzaferi, C., Cooke, R., Mendelson, R. A., and
Fletterick, R.J. (2005) Ca
2+
-regulated structural chang-
es in troponin, Proc. Natl. Acad. Sci. USA, 102, 5038-
5043, https://doi.org/10.1073/pnas.0408882102.
47. Takeda,S., Yamashita, A., Maeda, K., and Maéda, Y.
(2003) Structure of the core domain of human cardi-
ac troponin in the Ca
2+
-saturated form, Nature, 424,
35-41, https://doi.org/10.1038/nature01780.
48. Howarth, J. W., Meller, J., Solaro, R. J., Trewhella,J.,
and Rosevear, P. R. (2007) Phosphorylation-depen-
dent conformational transition of the cardiac spe-
cific N-extension of troponin I in cardiac troponin,
J.Mol. Biol., 373, 706-722, https://doi.org/10.1016/j.jmb.
2007.08.035.
49. Blumenschein, T. M., Stone, D. B., Fletterick, R. J.,
Mendelson, R. A., and Sykes, B. D. (2006) Dynamics
of the C-terminal region of TnI in the troponin com-
plex in solution, Biophys. J., 90, 2436-2444, https://
doi.org/10.1529/biophysj.105.076216.
50. Julien,O., Mercier,P., Allen, C. N., Fisette,O., Ramos,
C.H., Lague,P., Blumenschein, T.M., and Sykes, B.D.
(2011) Is there nascent structure in the intrinsically
disordered region of troponin I? Proteins, 79, 1240-
1250, https://doi.org/10.1002/prot.22959.
51. King, W.A., Stone, D.B., Timmins, P.A., Narayanan,T.,
von Brasch, A.A., Mendelson, R.A., and Curmi, P.M.
(2005) Solution structure of the chicken skeletal mus-
cle troponin complex via small-angle neutron and
X-ray scattering, J. Mol. Biol., 45, 797-815, https://
doi.org/10.1016/j.jmb.2004.10.090.
52. Murakami, K., Yumoto, F., Ohki, S. Y., Yasunaga, T.,
Tanokura,M., and Wakabayashi,T. (2005) Structural
basis for Ca
2+
-regulated muscle relaxation at inter-
action sites of troponin with actin and tropomyo-
sin, J. Mol. Biol., 352, 178-201, https://doi.org/10.1016/
j.jmb.2005.06.067.
53. Wang, H., Chalovich, J. M., and Marriott, G. (2012)
Structural dynamics of troponin I during Ca
2+
-acti-
vation of cardiac thin filaments: a multi-site Förster
resonance energy transfer study, PLoS One, 7, e50420,
https://doi.org/10.1371/journal.pone.0050420.
54. Zhou, Z., Li, K.L., Rieck,D., Ouyang,Y., Chandra,M.,
and Dong, W. J. (2012) Structural dynamics of C-do-
main of cardiac troponin I protein in reconstituted
thin filament, J. Biol. Chem., 87, 7661-7674, https://
doi.org/10.1074/jbc.M111.281600.
55. Galinska-Rakoczy, A., Engel, P., Xu, C., Jung, H.,
Craig, R., Tobacman, L. S., and Lehman, W. (2008)
Structural basis for the regulation of muscle contrac-
tion by troponin and tropomyosin, J.Mol. Biol., 379,
929-935, https://doi.org/10.1016/j.jmb.2008.04.062.
56. Galinska,A., Hatch,V., Craig,R., Murphy, A.M., Van
Eyk, J.E., Wang, C.L., Lehman, W., and Foster, D.B.
(2010) The C terminus of cardiac troponin I stabilizes
the Ca
2+
-activated state of tropomyosin on actin fila-
ments, Circ. Res., 106, 705-711, https://doi.org/10.1161/
CIRCRESAHA.109.210047.
57. Saggin, L., Gorza, L., Ausoni, S., and Schiaffino, S.
(1990) Cardiac troponinT in developing, regenerating
and denervated rat skeletal muscle, Development, 110,
547-554, https://doi.org/10.1242/dev.110.2.547.
58. Anderson, P. A., Malouf, N.N., Oakeley, A. E., Pagani,
E.D., and Allen, P.D. (1991) Troponin T isoform ex-
pression in humans. A comparison among normal
and failing adult heart, fetal heart, and adult and fe-
tal skeletal muscle, Circ. Res., 69, 1226-1233, https://
doi.org/10.1161/01.RES.69.5.1226.
59. Perry, S.V. (1998) Troponin T: genetics, properties and
function, J.Muscle Res. Cell Motil., 19, 575-602, https://
doi.org/10.1023/A:1005397501968.
60. Jin, J.P., and Chong, S. M. (2010) Localization of the
two tropomyosin-binding sites of troponin T, Arch.
Biochem. Biophys., 500, 144-150, https://doi.org/
10.1016/j.abb.2010.06.001.
61. Wei,B., and Jin, J.P. (2016) TNNT1, TNNT2, and TNNT3:
isoform genes, regulation, and structure-function re-
lationships, Gene, 582, 1-13, https://doi.org/10.1016/
j.gene.2016.01.006.
62. Marston, S., and Zamora, J.E. (2020) Troponin struc-
ture and function: a view of recent progress, J.Mus-
cle Res. Cell Motil., 41, 71-89, https://doi.org/10.1007/
s10974-019-09513-1.
63. Morris, E.P., and Lehrer, S.S. (1984) Troponin-tropo-
myosin interactions. Fluorescence studies of the bind-
ing of troponin, troponin T, and chymotryptic tropo-
ninT fragments to specifically labeled tropomyosin,
Biochemistry, 23, 2214-2220, https://doi.org/10.1021/
bi00305a018.
TROPONINS AND SKELETAL MUSCLE PATHOLOGIES 2099
BIOCHEMISTRY (Moscow) Vol. 89 Nos. 12-13 2024
64. Tanokura, M., Tawada, Y., Ono, A., and Ohtsuki, I.
(1983) Chymotryptic subfragments of troponin T from
rabbit skeletal muscle. Interaction with tropomyosin,
troponin I and troponin C, J. Biochem., 93, 331-337,
https://doi.org/10.1093/oxfordjournals.jbchem.a134185.
65. Franklin, A.J., Baxley,T., Kobayashi,T., and Chalovich,
J.M. (2012) The C-terminus of troponinT is essential
for maintaining the inactive state of regulated actin,
Biophys. J., 102, 2536-2544, https://doi.org/10.1016/
j.bpj.2012.04.037.
66. Oda, T., Yanagisawa, H., and Wakabayashi,T. (2020)
Cryo-EM structures of cardiac thin filaments reveal
the 3D architecture of troponin, J. Struct. Biol., 209,
107450, https://doi.org/10.1016/j.jsb.2020.107450.
67. Yamada, Y., Namba, K., and Fujii, T. (2020) Cardiac
muscle thin filament structures reveal calcium reg-
ulatory mechanism, Nat. Commun., 11, 153, https://
doi.org/10.1038/s41467-019-14008-1.
68. Risi, C.M., Pepper,I., Belknap,B., Landim-Vieira,M.,
White, H.D., Dryden,K., Pinto, J.R., Chase, P.B., and
Galkin, V.E. (2021) The structure of the native cardiac
thin filament at systolic Ca
2+
levels, Proc. Natl. Acad.
Sci. USA, 118, e2024288118, https://doi.org/10.1073/
pnas.2024288118.
69. Wang, Z., Grange, M., Pospich, S., Wagner, T., Kho,
A. L., Gautel, M., and Raunser, S. (2022) Structures
from intact myofibrils reveal mechanism of thin
filament regulation through nebulin, Science, 375,
eabn1934, https://doi.org/10.1126/science.abn1934.
70. Briggs, M. M., Maready, M., Schmidt, J. M., and
Schachat,F. (1994) Identification of a fetal exon in the
human fast troponin T gene, FEBS Lett., 350, 37-40,
https://doi.org/10.1016/0014-5793(94)00729-2.
71. Wilkinson, J. M., Moir, A. J., and Waterfield, M. D.
(1984) The expression of multiple forms of troponin
T in chicken-fast-skeletal muscle may result from
differential splicing of a single gene, Eur. J. Bio-
chem., 143, 47-56, https://doi.org/10.1111/j.1432-1033.
1984.tb08337.x.
72. Briggs, M. M., and Schachat,F. (1993) Origin of fetal
troponin T: developmentally regulated splicing of
a new exon in the fast troponin T gene, Dev. Biol.,
158, 503-509, https://doi.org/10.1006/dbio.1993.1208.
73. Medford, R. M., Nguyen, H. T., Destree, A. T., Sum-
mers,E., and Nadal-Ginard, B. (1984) A novel mech-
anism of alternative RNA splicing for the develop-
mentally regulated generation of troponin T iso-
forms from a single gene, Cell, 38, 409-421, https://
doi.org/10.1016/0092-8674(84)90496-3.
74. Wang, J., and Jin, J. P. (1997) Primary structure and
developmental acidic to basic transition of 13 alter-
natively spliced mouse fast skeletal muscle troponin
T isoforms, Gene, 193, 105-114, https://doi.org/10.1016/
S0378-1119(97)00100-5.
75. Wang,J., and Jin, J.P. (1998) Conformational modula-
tion of troponin T by configuration of the NH2-termi-
nal variable region and functional effects, Biochemis-
try, 37, 14519-14528, https://doi.org/10.1021/bi9812322.
76. Moore, G.E., Briggs, M.M., and Schachat, F.H. (1987)
Patterns of troponin T expression in mammalian
fast, slow and promiscuous muscle fibres, J. Mus-
cle Res. Cell Motil., 8, 13-22, https://doi.org/10.1007/
BF01767260.
77. Briggs, M. M., McGinnis, H. D., and Schachat, F.
(1990) Transitions from fetal to fast troponin T iso-
forms are coordinated with changes in tropomyo-
sin and alpha-actinin isoforms in developing rab-
bit skeletal muscle, Dev. Biol., 140, 253-260, https://
doi.org/10.1016/0012-1606(90)90075-T.
78. Melby, J. A., Jin, Y., Lin, Z., Tucholski, T., Wu, Z.,
Gregorich, Z.R., Diffee, G. M., and Ge,Y. (2020) Top-
down proteomics reveals myofilament proteoform
heterogeneity among various rat skeletal muscle
tissues, J. Proteome Res., 19, 446-454, https://doi.org/
10.1021/acs.jproteome.9b00623.
79. Jin, Y., Diffee, G. M., Colman, R. J., Anderson, R. M.,
and Ge, Y. (2019) Top-down mass spectrometry of
sarcomeric protein post-translational modifications
from non-human primate skeletal muscle, J. Am.
Soc. Mass Spectrom., 30, 2460-2469, https://doi.org/
10.1007/s13361-019-02139-0.
80. Chen, Y. C., Sumandea, M.P., Larsson,L., Moss, R.L.,
and Ge, Y. (2015) Dissecting human skeletal muscle
troponin proteoforms by top-down mass spectrom-
etry, J. Muscle Res. Cell Motil., 36, 169-181, https://
doi.org/10.1007/s10974-015-9404-6.
81. Jin, J. P., Chen, A., and Huang, Q. Q. (1998) Three
alternatively spliced mouse slow skeletal muscle
troponin T isoforms: conserved primary structure
and regulated expression during postnatal devel-
opment, Gene, 214, 121-129, https://doi.org/10.1016/
S0378-1119(98)00214-5.
82. Huang, Q.Q., Chen, A., and Jin, J.P. (1999) Genomic
sequence and structural organization of mouse slow
skeletal muscle troponin T gene, Gene, 229, 1-10,
https://doi.org/10.1016/S0378-1119(99)00051-7.
83. Gahlmann,R., Troutt, A. B., Wade, R.P., Gunning, P.,
and Kedes, L. (1987) Alternative splicing generates
variants in important functional domains of human
slow skeletal troponin T, J. Biol. Chem., 262, 16122-
16126, https://doi.org/10.1016/S0021-9258(18)47705-8.
84. Samson,F., Mesnard,L., Mihovilovic,M., Potter, T.G.,
Mercadier, J. J., Roses, A.D., and Gilbert, J.R. (1994)
A new human slow skeletal troponin T (TnTs) mRNA
isoform derived from alternative splicing of a single
gene, Biochem. Biophys. Res. Commun., 199, 841-847,
https://doi.org/10.1006/bbrc.1994.
85. Gahlmann, R., Wade, R., Gunning, P., and Kedes, L.
(1988) Differential expression of slow and fast skele-
tal muscle troponin C. Slow skeletal muscle troponin
C is expressed in human fibroblasts, J.Mol. Biol., 201,
379-391, https://doi.org/10.1016/0022-2836(88)90145-3.
BOGOMOLOVA, KATRUKHA2100
BIOCHEMISTRY (Moscow) Vol. 89 Nos. 12-13 2024
86. Houdusse,A., Love, M.L., Dominguez,R., Grabarek,Z.,
and Cohen, C. (1997) Structures of four Ca
2+
-bound
troponin C at 2.0 A resolution: further insights into
the Ca
2+
-switch in the calmodulin superfamily, Struc-
ture, 5, 1695-1711, https://doi.org/10.1016/s0969-2126
(97)00315-8.
87. Mercier, P., Ferguson, R. E., Irving, M., Corrie, J. E.,
Trentham, D.R., and Sykes, B.D. (2003) NMR struc-
ture of a bifunctional rhodamine labeled N-domain
of troponin C complexed with the regulatory “switch”
peptide from troponinI: implications for in situ flu-
orescence studies in muscle fibers, Biochemistry,
42, 4333-4348, https://doi.org/10.1021/bi027041n.
88. Potter, J. D., and Gergely, J. (1974) Troponin, tropo-
myosin, and actin interactions in the Ca
2+
regulation
of muscle contraction, Biochemistry, 13, 2697-2703,
https://doi.org/10.1021/bi00710a007.
89. Sia, S. K., Li, M. X., Spyracopoulos,L., Gagné, S. M.,
Liu,W., Putkey, J.A., and Sykes, B.D. (1997) Structure
of cardiac muscle troponin C unexpectedly reveals a
closed regulatory domain, J.Biol. Chem., 272, 18216-
18221, https://doi.org/10.1074/jbc.272.29.18216.
90. Morimoto,S., Harada,K., and Ohtsuki,I. (1999) Roles
of troponin isoforms in pH dependence of contrac-
tion in rabbit fast and slow skeletal and cardiac mus-
cles, J. Biochem., 126, 121-129, https://doi.org/10.1093/
oxfordjournals.jbchem.a022412.
91. Brotto, M. A., Biesiadecki, B. J., Brotto, L. S., Nosek,
T.M., and Jin, J.P. (2006) Coupled expression of tropo-
nin T and troponin I isoforms in single skeletal mus-
cle fibers correlates with contractility, Am. J. Physiol.
Cell Physiol., 290, C567-C576, https://doi.org/10.1152/
ajpcell.00422.2005.
92. Piroddi, N., Tesi, C., Pellegrino, M. A., Tobacman,
L. S., Homsher, E., and Poggesi, C. (2003) Contrac-
tile effects of the exchange of cardiac troponin for
fast skeletal troponin in rabbit psoas single myofi-
brils, J. Physiol., 552, 917-931, https://doi.org/10.1113/
jphysiol.2003.051615.
93. Yan, G. X., and Kleber, A. G. (1992) Changes in ex-
tracellular and intracellular pH in ischemic rab-
bit papillary muscle, Circ. Res., 71, 460-470, https://
doi.org/10.1161/01.RES.71.2.460.
94. Jennings, R. B., and Reimer, K. A. (1991) The cell bi-
ology of acute myocardial ischemia, Annu. Rev.
Med., 42, 225-246, https://doi.org/10.1146/annurev.
me.42.020191.001301.
95. Sahlin, K., Harris, R. C., Nylind, B., and Hultman,E.
(1976) Lactate content and pH in muscle obtained
after dynamic exercise, Pflugers Arch., 367, 143-149,
https://doi.org/10.1007/bf00585150.
96. Bangsbo, J., Madsen, K., Kiens, B., and Richter, E. A.
(1996) Effect of muscle acidity on muscle metabolism
and fatigue during intense exercise in man, J.Physi-
ol., 495, 587-596, https://doi.org/10.1113/jphysiol.1996.
sp021618.
97. Allen, D.G., Lamb, G.D., and Westerblad, H. (2008)
Skeletal muscle fatigue: cellular mechanisms, Physi-
ol. Rev., 88, 287-332, https://doi.org/10.1152/physrev.
00015.2007.
98. Donaldson, S.K., Hermansen,L., and Bolles,L. (1978)
Differential, direct effects of H
+
on Ca
2+
-activated
force of skinned fibers from the soleus, cardiac and
adductor magnus muscles of rabbits, Pflugers Arch.,
376, 55-65, https://doi.org/10.1007/bf00585248.
99. Metzger, J.M., Parmacek, M.S., Barr,E., Pasyk,K., Lin,
W. I., Cochrane, K. L., Field, L. J., and Leiden, J. M.
(1993) Skeletal troponin C reduces contractile sensi-
tivity to acidosis in cardiac myocytes from transgenic
mice, Proc. Natl. Acad. Sci. USA, 90, 9036-9040, https://
doi.org/10.1073/pnas.90.19.9036.
100. Metzger, J.M. (1996) Effects of troponin C isoforms on
pH sensitivity of contraction in mammalian fast and
slow skeletal muscle fibres, J. Physiol., 492, 163-172,
https://doi.org/10.1113/jphysiol.1996.sp021298.
101. Solaro, R.J., Kumar,P., Blanchard, E.M., and Martin,
A.F. (1986) Differential effects of pH on calcium ac-
tivation of myofilaments of adult and perinatal dog
hearts. Evidence for developmental differences in
thin filament regulation, Circ. Res., 58, 721-729, https://
doi.org/10.1161/01.res.58.5.721.
102. Wolska, B. M., Vijayan, K., Arteaga, G. M., Konhilas,
J. P., Phillips, R. M., Kim, R., Naya, T., Leiden, J. M.,
Martin, A.F., de Tombe, P.P., and Solaro, R. J. (2001)
Expression of slow skeletal troponin I in adult trans-
genic mouse heart muscle reduces the force decline
observed during acidic conditions, J.Physiol., 536, 863-
870, https://doi.org/10.1111/j.1469-7793.2001.00863.x.
103. Robertson, I. M., Holmes, P. C., Li, M. X., Pineda-
Sanabria, S.E., Baryshnikova, O.K., and Sykes, B.D.
(2012) Elucidation of isoform-dependent pH sen-
sitivity of troponin i by NMR spectroscopy, J. Biol.
Chem., 287, 4996-5007, https://doi.org/10.1074/jbc.
M111.301499.
104. Pineda-Sanabria, S.E., Robertson, I.M., Li, M.X., and
Sykes, B.D. (2013) Interaction between the regulatory
domain of cardiac troponinC and the acidosis-resis-
tant cardiac troponin I A162H, Cardiovasc. Res., 97,
481-489, https://doi.org/10.1093/cvr/cvs348.
105. Robertson, I.M., Pineda-Sanabria, S.E., Holmes, P.C.,
and Sykes, B.D. (2014) Conformation of the critical pH
sensitive region of troponin depends upon a single
residue in troponin I, Arch. Biochem. Biophys., 552-
553, 40-49, https://doi.org/10.1016/j.abb.2013.12.003.
106. Pineda-Sanabria, S. E., Robertson, I. M., and Sykes,
B.D. (2015) Structure and dynamics of the acidosis-
resistant A162H mutant of the switch region of tro-
ponin I bound to the regulatory domain of troponin
C, Biochemistry, 54, 3583-3593, https://doi.org/10.1021/
acs.biochem.5b00178.
107. Ogut,O., and Jin, J. P. (1998) Developmentally regu-
lated, alternative RNA splicing-generated pectoral
TROPONINS AND SKELETAL MUSCLE PATHOLOGIES 2101
BIOCHEMISTRY (Moscow) Vol. 89 Nos. 12-13 2024
muscle-specific troponin T isoforms and role of the
NH2-terminal hypervariable region in the tolerance
to acidosis, J. Biol. Chem., 273, 27858-27866, https://
doi.org/10.1074/jbc.273.43.27858.
108. Ogut, O., Granzier, H., and Jin, J. P. (1999) Acidic
and basic troponin T isoforms in mature fast-twitch
skeletal muscle and effect on contractility, Am. J.
Physiol., 276, C1162-C1170, https://doi.org/10.1152/
ajpcell.1999.276.5.C1162.
109. Nosek, T.M., Brotto, M.A., and Jin, J.P. (2004) Tropo-
nin T isoforms alter the tolerance of transgenic mouse
cardiac muscle to acidosis, Arch. Biochem. Biophys.,
430, 178-184, https://doi.org/10.1016/j.abb.2004.07.014.
110. Shen,J., Li,Y., Gu,H., Xia,F., and Zuo,X. (2014) Recent
development of sandwich assay based on the nano-
biotechnologies for proteins, nucleic acids, small mol-
ecules, and ions, Chem. Rev., 114, 7631-7677, https://
doi.org/10.1021/cr300248x.
111. Larue, C., Defacque-Lacquement, H., Calzolari, C.,
Le Nguyen, D., and Pau, B. (1992) New monoclonal
antibodies as probes for human cardiac troponin
I: epitopic analysis with synthetic peptides, Mol.
Immunol., 29, 271-278, https://doi.org/10.1016/0161-
5890(92)90109-B.
112. Larue,C., Calzolari,C., Bertinchant, J. P., Leclercq, F.,
Grolleau, R., and Pau, B. (1993) Cardiac-specific im-
munoenzymometric assay of troponin I in the early
phase of acute myocardial infarction, Clin. Chem., 39,
972-979, https://doi.org/10.1093/clinchem/39.6.972.
113. Rama, D., Margaritis, I., Orsetti, A., Marcon-
net, P., Gros, P., Larue, C., Trinquier, S., Pau, B., and
Calzolari, C. (1996) Troponin I immunoenzymomet-
ric assays for detection of muscle damage applied
to monitoring a triathlon, Clin. Chem., 42, 2033-2035,
https://doi.org/10.1093/clinchem/42.12.2033.
114. Onuoha, G. N., Alpar, E. K., Dean, B., Tidman, J.,
Rama, D., Laprade, M., and Pau, B. (2001) Skeletal
troponin-I release in orthopedic and soft tissue in-
juries, J. Orthop. Sci., 6, 11-15, https://doi.org/10.1007/
s007760170018.
115. Bamberg, K., Mehtälä, L., Arola, O., Laitinen, S.,
Nordling,P., Strandberg,M., Strandberg,N., Paltta,J.,
Mali,M., Espinosa-Ortega,F., Pirilä,L., Lundberg, I.E.,
Savukoski,T., and Pettersson,K. (2020) Evaluation of
a new skeletal troponin I assay in patients with idio-
pathic inflammatory myopathies, J. Appl. Lab. Med.,
5, 320-331, https://doi.org/10.1093/jalm/jfz016.
116. Sun,D., Hamlin,D., Butterfield,A., Watson, D.E., and
Smith, H.W. (2010) Electrochemiluminescent immu-
noassay for rat skeletal troponin I (Tnni2) in serum,
J. Pharmacol. Toxicol. Methods, 61, 52-58, https://
doi.org/10.1016/j.vascn.2009.09.002.
117. Simpson, J.A., Labugger,R., Hesketh, G.G., D’Arsigny,C.,
O’Donnell, D., Matsumoto, N., Collier, C. P., Iscoe, S.,
and Van Eyk, J. E. (2002) Differential detection of
skeletal troponin I isoforms in serum of a patient
with rhabdomyolysis: markers of muscle injury?
Clin. Chem., 48, 1112-1114, https://doi.org/10.1093/
clinchem/48.7.1112.
118. Simpson, J. A., Labugger, R., Collier, C., Brison, R. J.,
Iscoe,S., and Van Eyk, J.E. (2005) Fast and slow skel-
etal troponin I in serum from patients with various
skeletal muscle disorders: a pilot study, Clin. Chem., 51,
966-972, https://doi.org/10.1373/clinchem.2004.042671.
119. Takahashi, M., Lee, L., Shi, Q., Gawad, Y., and
Jackowski,G. (1996) Use of enzyme immunoassay for
measurement of skeletal troponin-I utilizing isoform-
specific monoclonal antibodies, Clin. Biochem., 29,
301-308, https://doi.org/10.1016/0009-9120(96)00016-1.
120. Sorichter, S., Mair, J., Koller, A., Calzolari,C., Huonk-
er, M., Pau,B., and Puschendorf,B. (2001) Release of
muscle proteins after downhill running in male and
female subjects, Scand. J. Med. Sci. Sports, 11, 28-32,
https://doi.org/10.1034/j.1600-0838.2001.011001028.x.
121. Chapman, D.W., Simpson, J. A., Iscoe, S., Robins, T.,
and Nosaka,K. (2013) Changes in serum fast and slow
skeletal troponin I concentration following maximal
eccentric contractions, J. Sci. Med. Sport, 16, 82-85,
https://doi.org/10.1016/j.jsams.2012.05.006.
122. Chen, T. C., Liu, H. W., Russell, A., Barthel, B. L.,
Tseng, K.W., Huang, M.J., Chou, T.Y., and Nosaka,K.
(2020) Large increases in plasma fast skeletal mus-
cle troponin I after whole-body eccentric exercises,
J.Sci. Med. Sport, 23, 776-781, https://doi.org/10.1016/
j.jsams.2020.01.011.
123. Kiely, P.D., Bruckner, F.E., Nisbet, J.A., and Daghir,A.
(2000) Serum skeletal troponin I in inflammatory
muscle disease: relation to creatine kinase, CKMB
and cardiac troponin I, Ann. Rheum. Dis., 59, 750-751,
https://doi.org/10.1136/ard.59.9.750.
124. Vassallo, J. D., Janovitz, E. B., Wescott, D. M.,
Chadwick, C., Lowe-Krentz, L. J., and Lehman-
McKeeman, L.D. (2009) Biomarkers of drug-induced
skeletal muscle injury in the rat: troponin I and
myoglobin, Toxicol. Sci., 111, 402-412, https://doi.org/
10.1093/toxsci/kfp166.
125. Tonomura, Y., Matsushima, S., Kashiwagi, E., Fuji-
sawa, K., Takagi, S., Nishimura, Y., Fukushima, R.,
Torii,M., and Matsubara,M. (2012) Biomarker panel
of cardiac and skeletal muscle troponins, fatty acid
binding protein 3 and myosin light chain 3 for the
accurate diagnosis of cardiotoxicity and musculoskel-
etal toxicity in rats, Toxicology, 302, 179-189, https://
doi.org/10.1016/j.tox.2012.07.012.
126. Madden,L., Juhas,M., Kraus, W.E., Truskey, G.A., and
Bursac,N. (2015) Bioengineered human myobundles
mimic clinical responses of skeletal muscle to drugs,
Elife, 4, e04885, https://doi.org/10.7554/eLife.04885.
127. Khodabukus,A., Kaza,A., Wang,J., Prabhu,N., Gold-
stein, R., Vaidya, V. S., and Bursac, N. (2020) Tissue-
engineered human myobundle system as a platform
for evaluation of skeletal muscle injury biomarkers,
BOGOMOLOVA, KATRUKHA2102
BIOCHEMISTRY (Moscow) Vol. 89 Nos. 12-13 2024
Toxicol. Sci., 176, 124-136, https://doi.org/10.1093/
toxsci/kfaa049.
128. Barthel, B. L., Cox, D., Barbieri, M., Ziemba, M.,
Straub, V., Hoffman, E. P., and Russell, A. J. (2021)
Elevation of fast but not slow troponin I in the cir-
culation of patients with Becker and Duchenne mus-
cular dystrophy, Muscle Nerve, 64, 43-49, https://
doi.org/10.1002/mus.27222.
129. Traa, W.A., Strijkers, G.J., Bader, D.L., and Oomens,
C. W. J. (2019) Myoglobin and troponin concentra-
tions are increased in early stage deep tissue inju-
ry, J. Mech. Behav. Biomed. Mater., 92, 50-57, https://
doi.org/10.1016/j.jmbbm.2018.12.026.
130. Perry, S.V., and Cole, H.A. (1974) Phosphorylation of
troponin and the effects of interactions between the
components of the complex, Biochem.J., 141, 733-743,
https://doi.org/10.1042/bj1410733.
131. Moir, A. J., Wilkinson, J. M., and Perry, S. V. (1974)
The phosphorylation sites of troponin I from white
skeletal muscle of the rabbit, FEBS Lett., 42, 253-256,
https://doi.org/10.1016/0014-5793(74)80739-8.
132. Moir, A. J., Cole, H. A., and Perry, S. V. (1977) The
phosphorylation sites of troponin T from white skel-
etal muscle and the effects of interaction with tropo-
ninC on their phosphorylation by phosphorylase ki-
nase, Biochem.J., 161, 371-382, https://doi.org/10.1042/
bj1610371.
133. Mazzei, G. J., and Kuo, J. F. (1984) Phosphorylation
of skeletal-muscle troponin I and troponin T by
phospholipid-sensitive Ca
2+
-dependent protein ki-
nase and its inhibition by troponin C and tropomy-
osin, Biochem.J., 218, 361-369, https://doi.org/10.1042/
bj2180361.
134. Sancho Solis,R., Ge,Y., and Walker, J.W. (2011) Apre-
ferred AMPK phosphorylation site adjacent to the in-
hibitory loop of cardiac and skeletal troponin I, Pro-
tein Sci., 20, 894-907, https://doi.org/10.1002/pro.623.
135. Cole, H.A., and Perry, S. V. (1975) The phosphoryla-
tion of troponin I from cardiac muscle, Biochem. J.,
149, 525-533, https://doi.org/10.1042/bj1490525.
136. Gusev, N. B., Dobrovol’skii, A. B., and Severin, S. E.
(1978) Skeletal muscle troponin and phosphorylation:
a site of troponinT, that is phosphorylated by specific
protein kinase [in Russian], Biokhimiia, 43, 365-372.
137. Risnik, V. V., Dobrovolskii, A. B., Gusev, N. B., and
Severin, S.E. (1980) Phosphorylase kinase phosphor-
ylation of skeletal-muscle troponin T, Biochem.J., 191,
851-854, https://doi.org/10.1042/bj1910851.
138. Gusev, N. B., Dobrovolskii, A. B., and Severin, S. E.
(1980) Isolation and some properties of troponin T
kinase from rabbit skeletal muscle, Biochem J, 189,
219-226, https://doi.org/10.1042/bj1890219.
139. Zemskova, M. A., Shur, S. A., Skolysheva, L. K., and
Vul’fson, P. L. (1991) Interaction of phosphorylase
kinase with thin filament proteins of rabbit skeletal
muscles [in Russian], Biokhimiia, 56, 100-108.
140. Katrukha, I.A., and Gusev, N.B. (2013) Enigmas of car-
diac troponin T phosphorylation, J.Mol. Cell Cardiol.,
65, 156-158, https://doi.org/10.1016/j.yjmcc.2013.09.017.
141. Gusev, N.B., Barskaya, N.V., Verin, A.D., Duzhenkova,
I.V., Khuchua, Z. A., and Zheltova, A.O. (1983) Some
properties of cardiac troponin T structure, Biochem.J.,
213, 123-129, https://doi.org/10.1042/bj2130123.
142. Zhang,J., Guy, M.J., Norman, H.S., Chen, Y.C., Xu,Q.,
Dong, X., Guner, H., Wang, S., Kohmoto, T., Young,
K.H., Moss, R. L., and Ge,Y. (2011) Top-down quan-
titative proteomics identified phosphorylation of car-
diac troponin I as a candidate biomarker for chronic
heart failure, J.Proteome Res., 10, 4054-4065, https://
doi.org/10.1021/pr200258m.
143. Lamb, G.D., and Westerblad,H. (2011) Acute effects of
reactive oxygen and nitrogen species on the contrac-
tile function of skeletal muscle, J. Physiol., 589, 2119-
2127, https://doi.org/10.1113/jphysiol.2010.199059.
144. Mollica, J.P., Dutka, T.L., Merry, T.L., Lamboley, C.R.,
McConell, G.K., McKenna, M. J., Murphy, R.M., and
Lamb, G. D. (2012) S-glutathionylation of troponin I
(fast) increases contractile apparatus Ca
2+
sensitiv-
ity in fast-twitch muscle fibres of rats and humans,
J. Physiol., 590, 1443-1463, https://doi.org/10.1113/
jphysiol.2011.224535.
145. Dutka, T.L., Mollica, J.P., Lamboley, C.R., Weerakkody,
V.C., Greening, D.W., Posterino, G.S., Murphy, R.M.,
and Lamb, G. D. (2017) S-nitrosylation and S-glu-
tathionylation of Cys134 on troponin I have op-
posing competitive actions on Ca
2+
sensitivity in
rat fast-twitch muscle fibers, Am. J. Physiol. Cell
Physiol., 312, C316-C327, https://doi.org/10.1152/
ajpcell.00334.2016.
146. Katrukha, A. G., Bereznikova, A. V., Filatov, V. L.,
Esakova, T. V., Kolosova, O. V., Pettersson, K.,
Lovgren,T., Bulargina, T.V., Trifonov, I.R., Gratsiansky,
N.A., Pulkki,K., Voipio-Pulkki, L.M., and Gusev, N.B.
(1998) Degradation of cardiac troponin I: implication
for reliable immunodetection, Clin. Chem., 44, 2433-
2440, https://doi.org/10.1093/clinchem/44.12.2433.
147. Katrukha, I. A., Kogan, A. E., Vylegzhanina, A. V.,
Kharitonov, A. V., Tamm, N. N., Filatov, V. L.,
Bereznikova, A.V., Koshkina, E.V., and Katrukha, A.G.
(2018) Full-size cardiac troponin I and its proteolytic
fragments in blood of patients with acute myocardial
infarction: antibody selection for assay development,
Clin. Chem., 64, 1104-1112, https://doi.org/10.1373/
clinchem.2017.286211.
148. Di Lisa, F., De Tullio, R., Salamino, F., Barbato, R.,
Melloni, E., Siliprandi, N., Schiaffino, S., and
Pontremoli, S. (1995) Specific degradation of tropo-
nin T and I by mu-calpain and its modulation by sub-
strate phosphorylation, Biochem.J., 308, 57-61, https://
doi.org/10.1042/bj3080057.
149. Wang, W., Schulze, C. J., Suarez-Pinzon, W.L., Dyck,
J. R., Sawicki, G., and Schulz, R. (2002) Intracellular
TROPONINS AND SKELETAL MUSCLE PATHOLOGIES 2103
BIOCHEMISTRY (Moscow) Vol. 89 Nos. 12-13 2024
action of matrix metalloproteinase-2 accounts for
acute myocardial ischemia and reperfusion injury,
Circulation, 106, 1543-1549, https://doi.org/10.1161/01.
cir.0000028818.33488.7b.
150. Gao, W. D., Atar, D., Liu, Y., Perez, N. G., Murphy,
A.M., and Marban, E. (1997) Role of troponin I pro-
teolysis in the pathogenesis of stunned myocardi-
um, Circ. Res., 80, 393-399, https://doi.org/10.1161/01.
res.0000435855.49359.47.
151. Mahmud, Z., Zahran, S., Liu, P. B., Reiz, B., Chan,
B.Y.H., Roczkowsky,A., McCartney, C.E., Davies, P.L.,
Li, L., Schulz,R., and Hwang, P. M. (2019) Structure
and proteolytic susceptibility of the inhibitory C-ter-
minal tail of cardiac troponin I, Biochim. Biophys.
Acta Gen. Subj., 1863, 661-671, https://doi.org/10.1016/
j.bbagen.2019.01.008.
152. Katrukha, I.A., and Katrukha, A.G. (2021) Myocardi-
al injury and the release of troponins I and T in the
blood of patients, Clin. Chem., 67, 124-130, https://
doi.org/10.1093/clinchem/hvaa281.
153. Communal,C., Sumandea,M., de Tombe,P., Narula,J.,
Solaro, R. J., and Hajjar, R. J. (2002) Functional con-
sequences of caspase activation in cardiac myocytes,
Proc. Natl. Acad. Sci. USA, 99, 6252-6256, https://
doi.org/10.1073/pnas.092022999.
154. Zhang,Z., Biesiadecki, B.J., and Jin, J.P. (2006) Selec-
tive deletion of the NH2-terminal variable region of
cardiac troponin T in ischemia reperfusion by myofi-
bril-associated mu-calpain cleavage, Biochemistry, 45,
11681-11694, https://doi.org/10.1021/bi060273s.
155. Katrukha, I. A., Riabkova, N. S., Kogan, A. E.,
Vylegzhanina, A.V., Mukharyamova, K.S., Bogomolo-
va, A.P., Zabolotskii, A.I., Koshkina, E.V., Berezniko-
va, A. V., and Katrukha, A. G. (2023) Fragmentation
of human cardiac troponin T after acute myocardi-
al infarction, Clin. Chim. Acta, 542, 117281, https://
doi.org/10.1016/j.cca.2023.117281.
156. Feng, H. Z., Wang,Q., Reiter, R.S., Lin, J. L., Lin, J.J.,
and Jin, J. P. (2013) Localization and function of
Xinα in mouse skeletal muscle, Am. J. Physiol. Cell
Physiol., 304, C1002-C1012, https://doi.org/10.1152/
ajpcell.00005.2013.
157. Katrukha, A. G., Bereznikova, A. V., Esakova, T. V.,
Pettersson,K., Lovgren,T., Severina, M.E., Pulkki,K.,
Vuopio-Pulkki, L. M., and Gusev, N. B. (1997) Tro-
ponin I is released in bloodstream of patients with
acute myocardial infarction not in free form but
as complex, Clin. Chem., 43, 1379-1385, https://
doi.org/10.1093/clinchem/43.8.1379.
158. Vylegzhanina, A. V., Kogan, A. E., Katrukha, I. A.,
Koshkina, E. V., Bereznikova, A. V., Filatov, V. L.,
Bloshchitsyna, M.N., Bogomolova, A.P., and Katrukha,
A. G. (2019) Full-size and partially truncated cardi-
ac troponin complexes in the blood of patients with
acute myocardial infarction, Clin. Chem., 65, 882-892,
https://doi.org/10.1373/clinchem.2018.301127.
159. Adamczyk, M., Brashear, R. J., and Mattingly, P. G.
(2009) Circulating cardiac troponin-I autoantibodies
in human plasma and serum, Ann. NY Acad. Sci., 1173,
67-74, https://doi.org/10.1111/j.1749-6632.2009.04617.x.
160. Vylegzhanina, A. V., Kogan, A. E., Katrukha, I. A.,
Antipova, O.V., Kara, A.N., Bereznikova, A.V., Koshki-
na, E.V., and Katrukha, A.G. (2017) Anti-cardiac tropo-
nin autoantibodies are specific to the conformational
epitopes formed by cardiac troponin I and troponinT
in the ternary troponin complex, Clin. Chem., 63,
343-350, https://doi.org/10.1373/clinchem.2016.261602.
161. Sung, S. S., Brassington, A.M., Grannatt,K., Ruther-
ford,A., Whitby, F. G., Krakowiak, P.A., Jorde, L.B.,
Carey, J. C., and Bamshad, M. (2003) Mutations in
genes encoding fast-twitch contractile proteins cause
distal arthrogryposis syndromes, Am. J. Hum. Genet.,
72, 681-690, https://doi.org/10.1086/368294.
162. Kimber, E., Tajsharghi, H., Kroksmark, A. K., Old-
fors,A., and Tulinius,M. (2006) A mutation in the fast
skeletal muscle troponin I gene causes myopathy and
distal arthrogryposis, Neurology, 67, 597-601, https://
doi.org/10.1212/01.wnl.0000230168.05328.f4.
163. Jiang,M., Zhao,X., Han,W., Bian,C., Li,X., Wang,G.,
Ao,Y., Li,Y., Yi, D., Zhe,Y., Lo, W.H., Zhang, X., and
Li,J. (2006) A novel deletion in TNNI2 causes distal
arthrogryposis in a large Chinese family with marked
variability of expression, Hum. Genet., 120, 238-242,
https://doi.org/10.1007/s00439-006-0183-4.
164. Shrimpton, A. E., and Hoo, J. J. (2006) A TNNI2 mu-
tation in a family with distal arthrogryposis type 2B,
Eur.J. Med. Genet., 49, 201-206, https://doi.org/10.1016/
j.ejmg.2005.06.003.
165. Li, X., Jiang, M., Han, W., Zhao, N., Liu, W., Sui, Y.,
Lu, Y., and Li, J. (2013) A novel TNNI2 mutation
causes Freeman-Sheldon syndrome in a Chinese
family with an affected adult with only facial con-
tractures, Gene, 527, 630-635, https://doi.org/10.1016/
j.gene.2013.06.082.
166. Li,Y., Nong,T., Li,X., Li,Z., Lv, H., Xu,H., Li, J., and
Zhu,M. (2022) A TNNI2 variant c.525G>T causes dis-
tal arthrogryposis in a Chinese family, Mol. Genet.
Genomic Med., 10, e2042, https://doi.org/10.1002/
mgg3.2042.
167. Čulić, V., Miyake, N., Janković, S., Petrović, D.,
Šimunović, M., Đapić, T., Shiina, M., Ogata, K., and
Matsumoto,N. (2016) Distal arthrogryposis with vari-
able clinical expression caused by TNNI2 mutation,
Hum. Genome Var., 3, 16035, https://doi.org/10.1038/
hgv.2016.35.
168. Wang, B., Zheng, Z., Wang, Z., Zhang, X., Yang, H.,
Cai,H., and Fu,Q. (2016) A novel missense mutation
of TNNI2 in a Chinese family cause distal arthrogryp-
osis type1, Am.J. Med. Genet.A, 170, 135-141, https://
doi.org/10.1002/ajmg.a.37391.
169. Seyama,R., Uchiyama,Y., Kaneshi, Y., Hamanaka,K.,
Fujita, A., Tsuchida, N., Koshimizu, E., Misawa, K.,
BOGOMOLOVA, KATRUKHA2104
BIOCHEMISTRY (Moscow) Vol. 89 Nos. 12-13 2024
Miyatake, S., Mizuguchi, T., Makino, S., Itakura, A.,
Okamoto,N., and Matsumoto,N. (2023) Distal arthro-
gryposis in a girl arising from a novel TNNI2 variant
inherited from paternal somatic mosaicism, J.Hum.
Genet., 68, 363-367, https://doi.org/10.1038/s10038-
022-01117-x.
170. Nishimori, Y., Iida, A., Ogasawara, M., Okubo, M.,
Yonenobu,Y., Kinoshita,M., Sugie,K., Noguchi,S., and
Nishino,I. (2022) TNNI1 mutated in autosomal domi-
nant proximal arthrogryposis, Neurol. Genet., 8, e649,
https://doi.org/10.1212/nxg.0000000000000649.
171. Beck, A.E., McMillin, M.J., Gildersleeve, H.I., Kezele,
P.R., Shively, K.M., Carey, J.C., Regnier,M., and Bam-
shad, M.J. (2013) Spectrum of mutations that cause
distal arthrogryposis types 1 and 2B, Am J Med Genet
A, 161, 550-555, https://doi.org/10.1002/ajmg.a.35809.
172. Desai, D., Stiene, D., Song, T., and Sadayappan, S.
(2020) Distal arthrogryposis and lethal congenital
contracture syndrome– an overview, Front. Physiol.,
11, 689, https://doi.org/10.3389/fphys.2020.00689.
173. Shafaattalab,S., Li, A.Y., Lin,E., Stevens, C.M., Dewar,
L.J., Lynn, F.C., Sanatani,S., Laksman,Z., Morin, R.D.,
van Petegem, F., Hove-Madsen, L., Tieleman, D. P.,
Davis, J.P., and Tibbits, G.F. (2019) Invitro analyses of
suspected arrhythmogenic thin filament variants as a
cause of sudden cardiac death in infants, Proc. Natl.
Acad. Sci. USA, 116, 6969-6974, https://doi.org/10.1073/
pnas.1819023116.
174. Sung, S.S., Brassington, A.M., Krakowiak, P.A., Carey,
J.C., Jorde, L.B., and Bamshad,M. (2003) Mutations in
TNNT3 cause multiple congenital contractures: a sec-
ond locus for distal arthrogryposis type 2B, Am.J. Hum.
Genet., 73, 212-214, https://doi.org/10.1086/376418.
175. Daly, S. B., Shah, H., O’Sullivan, J., Anderson, B.,
Bhaskar, S., Williams, S., Al-Sheqaih, N., Mueed
Bidchol, A., Banka, S., Newman, W. G., and Girisha,
K. M. (2014) Exome sequencing identifies a dom-
inant TNNT3 mutation in a large family with distal
arthrogryposis, Mol. Syndromol., 5, 218-228, https://
doi.org/10.1159/000365057.
176. Zhao,N., Jiang,M., Han,W., Bian,C., Li,X., Huang,F.,
Kong,Q., and Li,J. (2011) A novel mutation in TNNT3
associated with Sheldon-Hall syndrome in a Chinese
family with vertical talus, Eur.J. Med. Genet., 54, 351-
353, https://doi.org/10.1016/j.ejmg.2011.03.002.
177. Lu,J., Li,H., Zhang, H., Lin,Z., Xu, C., Xu,X., Hu,L.,
Luan,Z., Lou,Y., and Tang,S. (2021) The distal arthro-
gryposis-linked p.R63C variant promotes the stabili-
ty and nuclear accumulation of TNNT3, J.Clin. Lab.
Anal., 35, e24089, https://doi.org/10.1002/jcla.24089.
178. Sandaradura, S. A., Bournazos, A., Mallawaarach-
chi, A., Cummings, B. B., Waddell, L. B., Jones, K. J.,
Troedson, C., Sudarsanam, A., Nash, B. M., Peters,
G. B., Algar, E. M., MacArthur, D. G., North, K. N.,
Brammah,S., Charlton,A., Laing, N.G., Wilson, M.J.,
Davis, M. R., and Cooper, S. T. (2018) Nemaline my-
opathy and distal arthrogryposis associated with an
autosomal recessive TNNT3 splice variant, Hum. Mu-
tat., 39, 383-388, https://doi.org/10.1002/humu.23385.
179. Johnston, J. J., Kelley, R. I., Crawford, T. O., Morton,
D.H., Agarwala,R., Koch,T., Schaffer, A.A., Franco-
mano, C.A., and Biesecker, L.G. (2000) A novel nema-
line myopathy in the Amish caused by a mutation in
troponin T1, Am. J. Hum. Genet., 67, 814-821, https://
doi.org/10.1086/303089.
180. Marra, J. D., Engelstad, K. E., Ankala, A., Tanji, K.,
Dastgir, J., De Vivo, D. C., Coffee, B., and Chiriboga,
C. A. (2015) Identification of a novel nemaline my-
opathy-causing mutation in the troponin T1 (TNNT1)
gene: a case outside of the old order Amish, Muscle
Nerve, 51, 767-772, https://doi.org/10.1002/mus.24528.
181. van der Pol, W. L., Leijenaar, J. F., Spliet, W. G.,
Lavrijsen, S.W., Jansen, N.J., Braun, K.P., Mulder,M.,
Timmers-Raaijmakers,B., Ratsma,K., Dooijes,D., and
van Haelst, M.M. (2014) Nemaline myopathy caused
byTNNT1 mutations in a Dutch pedigree, Mol. Gen-
et. Genomic Med., 2, 134-137, https://doi.org/10.1002/
mgg3.52.
182. Abdulhaq, U.N., Daana,M., Dor,T., Fellig,Y., Eylon,S.,
Schuelke,M., Shaag,A., Elpeleg,O., and Edvardson,S.
(2016) Nemaline body myopathy caused by a novel
mutation in troponin T1 (TNNT1), Muscle Nerve, 53,
564-569, https://doi.org/10.1002/mus.24885.
183. Jin, J.P., Brotto, M. A., Hossain, M.M., Huang, Q.Q.,
Brotto, L. S., Nosek, T. M., Morton, D. H., and Craw-
ford, T.O. (2003) Truncation by Glu180 nonsense mu-
tation results in complete loss of slow skeletal mus-
cle troponin T in a lethal nemaline myopathy, J.Biol.
Chem., 278, 26159-26165, https://doi.org/10.1074/
jbc.M303469200.
184. Wang,X., Huang, Q.Q., Breckenridge, M.T., Chen,A.,
Crawford, T. O., Morton, D. H., and Jin, J. P. (2005)
Cellular fate of truncated slow skeletal muscle tro-
ponin T produced by Glu180 nonsense mutation in
amish nemaline myopathy, J.Biol. Chem., 280, 13241-
13249, https://doi.org/10.1074/jbc.M413696200.
185. Mondal,A., and Jin, J.P. (2016) Protein structure-func-
tion relationship at work: learning from myopathy
mutations of the slow skeletal muscle isoform of tro-
poninT, Front. Physiol., 7, 449, https://doi.org/10.3389/
fphys.2016.00449.
186. Van de Locht, M., Donkervoort, S., de Winter, J. M.,
Conijn,S., Begthel,L., Kusters,B., Mohassel,P., Hu,Y.,
Medne,L., Quinn,C., Moore, S.A., Foley, A.R., Seo,G.,
Hwee, D.T., Malik, F. I., Irving, T., Ma, W., Granzier,
H. L., Kamsteeg, E. J., Immadisetty, K., et al. (2021)
Pathogenic variants in TNNC2 cause congenital my-
opathy due to an impaired force response to calcium,
J. Clin. Invest., 131, e145700, https://doi.org/10.1172/
jci145700.
187. Van de Locht, M., Borsboom, T. C., Winter, J. M.,
and Ottenheijm, C. A. C. (2021) Troponin variants
TROPONINS AND SKELETAL MUSCLE PATHOLOGIES 2105
BIOCHEMISTRY (Moscow) Vol. 89 Nos. 12-13 2024
in congenital myopathies: how they affect skeletal
muscle mechanics, Int. J. Mol. Sci., 22, 9187, https://
doi.org/10.3390/ijms22179187.
188. Hoffmann,B., Schmidt-Traub,H., Perrot,A., Osterziel,
K. J., and Gessner, R. (2001) First mutation in car-
diac troponin C, L29Q, in a patient with hypertro-
phic cardiomyopathy, Hum. Mutat., 17, 524, https://
doi.org/10.1002/humu.1143.
189. Landstrom, A. P., Parvatiyar, M. S., Pinto, J. R., Mar-
quardt, M. L., Bos, J. M., Tester, D.J., Ommen, S. R.,
Potter, J.D., and Ackerman, M.J. (2008) Molecular and
functional characterization of novel hypertrophic
cardiomyopathy susceptibility mutations in TNNC1-
encoded troponin C, J.Mol. Cell Cardiol., 45, 281-288,
https://doi.org/10.1016/j.yjmcc.2008.05.003.
190. Chung, W. K., Kitner, C., and Maron, B. J. (2011)
Novel frameshift mutation in Troponin C (TNNC1)
associated with hypertrophic cardiomyopathy and
sudden death, Cardiol. Young, 21, 345-348, https://
doi.org/10.1017/S1047951110001927.
191. Parvatiyar, M. S., Landstrom, A. P., Figueiredo-
Freitas, C., Potter, J. D., Ackerman, M. J., and Pinto,
J. R. (2012) A mutation in TNNC1-encoded cardiac
troponin C, TNNC1-A31S, predisposes to hypertrophic
cardiomyopathy and ventricular fibrillation, J. Biol.
Chem., 287, 31845-31855, https://doi.org/10.1074/
jbc.M112.377713.
192. Caforio, A. L., Rossi, B., Risaliti, R., Siciliano, G.,
Marchetti,A., Angelini, C., Crea,F., Mariani, M., and
Muratorio, A. (1989) Type 1 fiber abnormalities in
skeletal muscle of patients with hypertrophic and
dilated cardiomyopathy: evidence of subclinical myo-
genic myopathy, J. Am. Coll. Cardiol., 14, 1464-1473,
https://doi.org/10.1016/0735-1097(89)90383-5.
193. Veltri, T., Landim-Vieira, M., Parvatiyar, M. S., Gon-
zalez-Martinez, D., Dieseldorff Jones, K.M., Michell,
C. A., Dweck, D., Landstrom, A. P., Chase, P. B., and
Pinto, J.R. (2017) Hypertrophic cardiomyopathy car-
diac troponin C mutations differentially affect slow
skeletal and cardiac muscle regulation, Front. Physi-
ol., 8, 221, https://doi.org/10.3389/fphys.2017.00221.
194. Li, M.X., Mercier, P., Hartman, J. J., and Sykes, B.D.
(2021) Structural basis of tirasemtiv activation of fast
skeletal muscle, J. Med. Chem., 64, 3026-3034, https://
doi.org/10.1021/acs.jmedchem.0c01412.
195. Russell, A. J., Hartman, J. J., Hinken, A. C., Muci,
A. R., Kawas,R., Driscoll,L., Godinez, G., Lee, K. H.,
Marquez,D., Browne, W.F.t., Chen, M.M., Clarke,D.,
Collibee, S. E., Garard, M., Hansen, R., Jia, Z., Lu,
P.P., Rodriguez, H., Saikali, K.G., Schaletzky,J., et al.
(2012) Activation of fast skeletal muscle troponin as
a potential therapeutic approach for treating neu-
romuscular diseases, Nat. Med., 18, 452-455, https://
doi.org/10.1038/nm.2618.
196. Shefner, J., Cedarbaum, J. M., Cudkowicz, M. E.,
Maragakis, N., Lee, J., Jones, D., Watson, M. L.,
Mahoney, K., Chen, M., Saikali, K., Mao, J., Russell,
A.J., Hansen, R. L., Malik,F., and Wolff, A.A. (2012)
Safety, tolerability and pharmacodynamics of a skel-
etal muscle activator in amyotrophic lateral sclero-
sis, Amyotroph. Lateral. Scler., 13, 430-438, https://
doi.org/10.3109/17482968.2012.684214.
197. Shefner, J. M., Watson, M. L., Meng, L., and Wolff,
A.A. (2013) A study to evaluate safety and tolerabil-
ity of repeated doses of tirasemtiv in patients with
amyotrophic lateral sclerosis, Amyotroph. Lateral
Scler. Frontotemporal Degener., 14, 574-581, https://
doi.org/10.3109/21678421.2013.822517.
198. Shefner, J. M., Wolff, A. A., and Meng, L. (2013) The
relationship between tirasemtiv serum concentration
and functional outcomes in patients with ALS, Amyo-
troph. Lateral Scler. Frontotemporal Degener., 14, 582-
585, https://doi.org/10.3109/21678421.2013.817587.
199. Shefner, J.M., Wolff, A.A., Meng,L., Bian,A., Lee, J.,
Barragan,D., and Andrews, J.A. (2016) A randomized,
placebo-controlled, double-blind phase IIb trial evalu-
ating the safety and efficacy of tirasemtiv in patients
with amyotrophic lateral sclerosis, Amyotroph. Later-
al Scler. Frontotemporal Degener., 17, 426-435, https://
doi.org/10.3109/21678421.2016.1148169.
200. Sanders, D. B., Rosenfeld, J., Dimachkie, M. M.,
Meng,L., and Malik, F.I. (2015) A double-blinded, ran-
domized, placebo-controlled trial to evaluate efficacy,
safety, and tolerability of single doses of tirasemtiv
in patients with acetylcholine receptor-binding anti-
body-positive myasthenia gravis, Neurotherapeutics,
12, 455-460, https://doi.org/10.1007/s13311-015-0345-y.
201. Andrews, J. A., Cudkowicz, M. E., Hardiman, O.,
Meng,L., Bian,A., Lee,J., Wolff, A.A., Malik, F.I., and
Shefner, J. M. (2018) VITALITY-ALS, a phase III trial
of tirasemtiv, a selective fast skeletal muscle tropo-
nin activator, as a potential treatment for patients
with amyotrophic lateral sclerosis: study design and
baseline characteristics, Amyotroph. Lateral Scler.
Frontotemporal Degener., 19, 259-266, https://doi.org/
10.1080/21678421.2018.1426770.
202. Shefner, J.M., Cudkowicz, M.E., Hardiman, O., Cock-
croft, B.M., Lee, J.H., Malik, F.I., Meng,L., Rudnicki,
S.A., Wolff, A.A., and Andrews, J.A. (2019) A phase
III trial of tirasemtiv as a potential treatment for
amyotrophic lateral sclerosis, Amyotroph. Lateral
Scler. Frontotemporal Degener., 20, 584-594, https://
doi.org/10.1080/21678421.2019.1612922.
203. Hwee, D.T., Kennedy, A.R., Hartman, J.J., Ryans,J.,
Durham, N., Malik, F. I., and Jasper, J. R. (2015) The
small-molecule fast skeletal troponin activator,
CK-2127107, improves exercise tolerance in a rat
model of heart failure, J.Pharmacol. Exp. Ther., 353,
159-168, https://doi.org/10.1124/jpet.114.222224.
204. Andrews, J.A., Miller, T.M., Vijayakumar,V., Stoltz,R.,
James, J. K., Meng, L., Wolff, A. A., and Malik, F. I.
(2018) CK-2127107 amplifies skeletal muscle response
BOGOMOLOVA, KATRUKHA2106
BIOCHEMISTRY (Moscow) Vol. 89 Nos. 12-13 2024
to nerve activation in humans, Muscle Nerve, 57, 729-
734, https://doi.org/10.1002/mus.26017.
205. Cheng, A.J., Hwee, D.T., Kim, L.H., Durham,N., Yang,
H.T., Hinken, A.C., Kennedy, A.R., Terjung, R.L., Jas-
per, J. R., Malik, F. I., and Westerblad, H. (2019) Fast
skeletal muscle troponin activator CK-2066260 in-
creases fatigue resistance by reducing the energetic
cost of muscle contraction, J.Physiol., 597, 4615-4625,
https://doi.org/10.1113/jp278235.
206. Shefner, J. M., Andrews, J. A., Genge,A., Jackson,C.,
Lechtzin, N., Miller, T. M., Cockroft, B. M., Meng, L.,
Wei, J., Wolff, A. A., Malik, F. I., Bodkin, C., Brooks,
B. R., Caress, J., Dionne, A., Fee, D., Goutman, S. A.,
Goyal, N. A., Hardiman, O., Hayat, G., et al. (2021)
A phase 2, double-blind, randomized, dose-ranging
trial of reldesemtiv in patients with ALS, Amyotroph.
Lateral Scler. Frontotemporal Degener., 22, 287-299,
https://doi.org/10.1080/21678421.2020.1822410.
207. Collibee, S. E., Bergnes, G., Chuang, C., Ashcraft, L.,
Gardina, J., Garard, M., Jamison, C. R., Lu, K., Lu,
P. P., Muci, A., Romero, A., Valkevich, E., Wang, W.,
Warrington, J., Yao, B., Durham, N., Hartman, J.,
Marquez, A., Hinken, A., Schaletzky, J., et al. (2021)
Discovery of reldesemtiv, a fast skeletal muscle tro-
ponin activator for the treatment of impaired mus-
cle function, J. Med. Chem., 64, 14930-14941, https://
doi.org/10.1021/acs.jmedchem.1c01067.
208. Issahaku, A. R., Ibrahim, M. A. A., Mukelabai, N.,
and Soliman, M.E. S. (2023) Intermolecular and dy-
namic investigation of the mechanism of action of
reldesemtiv on fast skeletal muscle troponin com-
plex toward the treatment of impaired muscle func-
tion, Protein J., 42, 263-275, https://doi.org/10.1007/
s10930-023-10091-y.
209. Rudnicki, S. A., Andrews, J. A., Duong, T., Cock-
roft, B.M., Malik, F. I., Meng,L., Wei,J., Wolff, A.A.,
Genge, A., Johnson, N. E., Tesi-Rocha, C., Connolly,
A.M., Darras, B.T., Felice,K., Finkel, R.S., Shieh, P.B.,
Mah, J.K., Statland,J., Campbell,C., Habib, A.A., etal.
(2021) Reldesemtiv in patients with spinal muscular
atrophy: a phase 2 hypothesis-generating study, Neu-
rotherapeutics, 18, 1127-1136, https://doi.org/10.1007/
s13311-020-01004-3.
210. Shefner, J.M., Al-Chalabi, A., Andrews, J.A., Chio, A.,
DeCarvalho,M., Cockroft, B.M., Corcia,P., Couratier,P.,
Cudkowicz, M.E., Genge,A., Hardiman, O., Heiman-
Patterson,T., Henderson, R.D., Ingre,C., Jackson, C.E.,
Johnston, W., Lechtzin, N., Ludolph, A., Maragakis,
N.J., Miller, T.M., et al. (2023) COURAGE-ALS: a ran-
domized, double-blind phase 3 study designed to im-
prove participant experience and increase the prob-
ability of success, Amyotroph. Lateral Scler. Fronto-
temporal Degener., 24, 523-534, https://doi.org/10.1080/
21678421.2023.2216223.
211. Robertson, I. M., Baryshnikova, O. K., Li, M. X., and
Sykes, B.D. (2008) Defining the binding site of levo-
simendan and its analogues in a regulatory cardi-
ac troponin C-troponin I complex, Biochemistry, 47,
7485-7495, https://doi.org/10.1021/bi800438k.
212. Thompson, B. R., Martindale, J., and Metzger, J. M.
(2016) Sarcomere neutralization in inherited car-
diomyopathy: small-molecule proof-of-concept to
correct hyper-Ca
2+
-sensitive myofilaments, Am. J.
Physiol. Heart Circ. Physiol., 311, H36-H43, https://doi.
org/10.1152/ajpheart.00981.2015.
213. Cai,F., Hwang, P.M., and Sykes, B.D. (2018) Structural
changes induced by the binding of the calcium desen-
sitizer w7 to cardiac troponin, Biochemistry, 57, 6461-
6469, https://doi.org/10.1021/acs.biochem.8b00882.
214. Adhikari, B.B., and Wang,K. (2004) Interplay of tro-
ponin- and myosin-based pathways of calcium ac-
tivation in skeletal and cardiac muscle: the use of
W7 as an inhibitor of thin filament activation, Bio-
phys. J., 86, 359-370, https://doi.org/10.1016/s0006-
3495(04)74112-0.
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.