ISSN 0006-2979, Biochemistry (Moscow), 2025, Vol. 90, No. 6, pp. 700-724 © Pleiades Publishing, Ltd., 2025.
Published in Russian in Biokhimiya, 2025, Vol. 90, No. 6, pp. 752-780.
700
REVIEW
Telomere Length and Telomerase Activity
as Biomarkers in the Diagnostics and Prognostics
of Pathological Conditions
Elizaveta Yu. Moskaleva
1,a
*, Alexander I. Glukhov
2,3,b
, Alexander S. Zhirnik
1,c
,
Olga V. Vysotskaya
1,d
, and Svetlana A. Vorobiova
2,e
1
Kurchatov complex for NBICS Sciences and Nature-like Technologies,
National Research Center “Kurchatov Institute”, 123182 Moscow, Russia
2
Department of Biological Chemistry,
I.M. Sechenov First Moscow State Medical University (Sechenov University),
119048 Moscow, Russia
3
Faculty of Biology, Lomonosov Moscow State University,
119991 Moscow, Russia
a
e-mail: Moskaleva_EY@nrcki.ru; moskalevaey@mail.ru 
b
e-mail: aiglukhov1958@gmail.com
c
e-mail: as.zhirnik@mail.ru 
d
e-mail: ovvysotskaya@mail.ru 
e
e-mail: vorobeva_s_a@staff.sechenov.ru
Received March 20, 2025
Revised June 16, 2025
Accepted June 16, 2025
AbstractTelomere biology still remains a topic of interest in life sciences. Analysis of several thousand
clinical samples from healthy individuals performed in recent years has shown that the telomere length (TL)
in peripheral blood leukocytes correlates with the TL in cells of internal organ and reflects their condition.
TLdecreases under the influence of damaging factors and can serve as an indicator of health status. Thetelo-
mere shortening leads to the cell proliferation arrest and is considered as a marker of replicative aging of
proliferating cells. A decrease in the TL in peripheral blood leukocytes is viewed as an indicator of organism
aging. Recent studies have allowed to formulate the concept on the role of the CST–polymerase α/primase
in the C-strand fill in after completion of 3′G overhang synthesis by telomerase during telomere replication.
The discovery of the telomeric RNA (TERRA) and its role in the regulation of telomerase activity (TA) and
alternative lengthening of telomeres, as well as the possibility of TERRA translation, has provided evidence of
the complex epigenetic regulation of the TL maintenance. Analysis of the published data indicates that telo-
meres are dynamic structures, whose length undergoes significant changes under the influence of damaging
factors. TL is determined not only by the chronological age, but also by the exposure to the exogenous and
endogenous deleterious factors during the lifetime. A decrease in the TL due to inherited mutations in the
genes coding for proteins involved in the telomere structure formation and telomere replication (primarily,
proteins of the shelterin and CST complexes and telomerase) has been found in a number of hereditary
diseases – telomeropathies. The assessment of TL and TA is of great importance for the diagnostics of telo-
meropathies and can be useful in the diagnostics of cancer. Analysis of TL can be used for monitoring the
health status (e.g., in the case of exposure to ionizing radiation and space flight factors), as well as predicting
individual’s sensitivity to the action of various damaging agents. The application of modern advancement in
genetic technologies in the analysis of TL and TA makes it available for the use in clinical and epidemiological
studies, diagnostics of telomeropathies, and monitoring of astronauts’ health.
DOI: 10.1134/S0006297925600814
Keywords: telomeres, telomerase, telomere length, CST–polymerase α/primase, shelterin complex, CST complex,
TERRA, ionizing radiation, radiosensitivity, oxidative stress, occupational irradiation, telomeropathies
* To whom correspondence should be addressed.
TELOMERE LENGTH AS PROGNOSTIC MARKER 701
BIOCHEMISTRY (Moscow) Vol. 90 No. 6 2025
INTRODUCTION
About 50 years ago, the Russian journal Proceed-
ing of the USSR Academy of Sciences (Doklady Aka-
demii Nauk SSSR) in 1971 and then the Journal of
Theoretical Biology in 1973 published the visionary
articles of the research scientist and theoretician Alex-
ey Matveevich Olovnikov (1936-2022), the author of
the terminal underreplication theory that explained
the shortening of the chromosomal ends with each
cell division [1, 2]. The ideas presented in these arti-
cles had been developed long before the discovery of
telomere shortening and its molecular mechanisms.
The studies of telomeres and mechanisms preserving
their length with the involvement of special poly-
merase have been initiated due to the publication of
the famous work by Leonard Hayflick, who stated that
normal somatic cells (fibroblasts) cultured in vitro are
unable to proliferate indefinitely and stop dividing af-
ter approximately 50 divisions [3].
To explain this phenomenon, Olovnikov suggest-
ed that during preparation for the cell division, the
chromosomal DNA cannot not be fully replicated and
becomes shorter at the ends after each doubling. DNA
shortening to a certain critical length causes the mal-
functioning of genes located close to the telomeres,
eventually leading to the cell death. According to the
theory of telomere aging, the shortening of chromo-
somal ends is a timer mechanism that determines the
number of cell divisions and explains the Hayflick limit.
These ideas have been confirmed by subsequent
discoveries. First, Greider and Blackburn found a new
enzyme, telomere terminal transferase (later named
telomerase), in the extracts of Tetrahymena cells [4].
The same authors demonstrated that telomerase con-
tains RNA essential for the synthesis of telomeric re-
peats [5]. Inspired by the work of Olovnikov, CarolW.
Greider conducted similar studies in human fibro-
blasts [6], resulting in the discovery of telomerase in
human cells and cells of other eukaryotic organisms.
It was demonstrated that the aging of cultured hu-
man fibroblasts was indeed accompanied by telomere
shortening [7, 8]. The results obtained by Greider and
Blackburn, who were awarded the Nobel Prize in
Physiology or Medicine in 2004, have fully proven the
Olovnikov’s hypothesis on the mechanisms of somatic
cell aging. Moreover, these studies have opened a new
era in the research of the role and mechanisms of telo-
mere length (TL) maintenance in normal cells and in
various pathological processes, including those caused
by deleterious factors, such as ionizing radiation (IR),
and space flight factors. Biochemistry (Moscow) [9]
and Biogerontology [10] have dedicated special issues
to the memory of A.  M.  Olovnikov and modern devel-
opments in the concepts of telomere structure he had
predicted.
The structure of chromosomal telomeric regions
is characterized by a number of features that make
them particularly sensitive to the effects of genotoxic
factors. A decrease in the TL can lead to irreversible
disturbances in cell functioning and can serve as a
marker for the individual’s increased sensitivity to the
effects of endogenous and exogenous damaging fac-
tors. Current achievements in biochemistry and mo-
lecular genetics and introduction of methods for the
analysis of large amounts of data have significantly
expanded the possibilities of medical and biological
studies of the state of telomeres.
The aim of this review was to analyze the results
of studies on the mechanisms of telomere mainte-
nance in human cells under the influence of damag-
ing factors and in diseases caused by disturbances in
the telomere biology. Such analysis can be helpful in
selection of prognostic markers of the increased sen-
sitivity of patients to radiotherapy and chemotherapy,
which would allow to evaluate the risk of developing
unwanted late complications. Here, we addressed cur-
rent concepts on the telomere structure, mechanisms
of TL maintenance, regulation of telomerase activi-
ty (TA), telomere sensitivity to irradiation, oxidative
stress, and space flight factors, state of the TA-TL sys-
tem, and technologies for the TA and TL assessments
that can be used in clinical and epidemiological stud-
ies.
TELOMERE STRUCTURE AND MECHANISMS
OF TELOMERE MAINTENANCE
Telomere structure. Telomeres are specialized
DNA sequences at the ends of eukaryotic chromo-
somes. In humans, they consist of the repeating
non-coding six-nucleotide sequence TTAGGG. The un-
derreplication of chromosomal DNA due to the prop-
erties of DNA polymerase functioning results in the
generation of free single-stranded GGG overhangs at
the telomere 3′-ends in the course of DNA replication.
During DNA replication in the S phase of the cell cy-
cle, telomeres exist in the open linear form, while in
other phases of the cell cycle, telomeres form loops
that close the ends of the chromosomes (Fig. 1).
Electron microscopy studies of the in situ con-
figuration of human and mouse telomeric DNA [11]
showed the presence of large loops formed by fold-
ing back the telomere ends. The circular segment
of the loops consisted of the telomeric DNA duplex
called the T-loop. The invasion of the 3′ telomeric
overhang into the duplex telomeric repeat array re-
sults in the displacement of the TTAGGG repeat strand
at the loop-tail junction. Based on the binding with
a single-stranded DNA, this displacement loop of
TTAGGG repeats (D-loop, Fig.  1) was deduced to be
MOSKALEVA et al.702
BIOCHEMISTRY (Moscow) Vol. 90 No. 6 2025
Fig. 1. Open telomere and telomere as the T-loop with D-loop at the site of the single-stranded telomere overhang inser-
tion (a); the binding of shelterins to the double-stranded (TRF1 and TRF2) and single-stranded (POT1) telomeric DNA (b).
Shelterin complex proteins cover the entire telomere. According to [11, 15].
approximately several hundred nucleotides long in
many T-loops. Although the exact site of the 3′-end
invasion has not been identified, in most cases the
loop was very large (many kilobases) and sometimes
encompassed the entire telomere. A close correlation
was found between the length of the telomeric repeats
and the T-loop size [11]. Figure1 shows the structure
of the T-loop and provides a general overview of how
the loops can form to protect the telomeric ends, al-
though the possibility of a different organization of
the loops cannot be ruled out [12, 13].
Human telomeres contain a large number of
G-quadruplexes (G4s). G4s are non-canonical four-
stranded structures that can form in guanine-rich
DNA and RNA sequences. The core structure of G4s
consists of stacked guanine tetrads (G-tetrads), square
planar platforms of four guanine bases held togeth-
er by Hoogsteen hydrogen bonds. Formation of G4s
requires cations, in particular, K
+
or Na
+
, to stabilize
the stacked guanine tetrads through coordination with
the O6 atoms in guanines. G4s can promote the T-loop
formation and provide intermolecular DNA–RNA in-
teractions [14].
An important role in the formation and stabili-
zation of T-loops and, thus, in the protection of telo-
meric ends, belongs to the proteins of the shelterin
complex (Fig. 1b) [15-18]. These proteins bind to the
telomeric DNA and determine the shape of the telo-
meric ends. They protect telomeres from the action of
DNA repair enzymes and perform a number of other
functions resulting in telomere protection [15, 17, 19].
The shelterin complex consists of six proteins listed
in Table 1.
The POT1–TPP1 complex is a structural compo-
nent of the telomere that increases the activity and
processivity of telomerase core enzyme and provides
the TL increase during DNA replication [21, 22].
Telomeres are also associated with a number
of proteins (Mre11/Rad50/Nbs1, ERCC1/XPF, DNA-PK,
PARP2) involved in recombination and DNA repair,
including homologous recombination repair. These
proteins interact with the shelterin complex proteins
and participate in the formation and maintenance of
T-loops [15]. Beside the formation of T-loops to prevent
the recognition of chromosomal ends as double-strand
DNA breaks (DSBs), other cellular mechanisms have
been discovered that suppress the development of the
DNA damage response (DDR) at the telomeres. These
mechanisms involve proteins of the shelterin complex
and have been described in detail in [12, 17, 23].
In his discussion of the connection between the
telomere shortening, decreased cell survival, and
aging, Yegorov [24] pointed out the strained confor-
mation of DNA in the T-loop. Gradual shortening of
telomeric DNA may come to a point when the TL
becomes insufficient for the telomeric DNA to form
a loop. The telomere acquires a free open conforma-
tion and is perceived by the cell as a DSB, which trig-
gers the DDR and results in telomere damage [24].
This hypothesis explains the connection between the
TELOMERE LENGTH AS PROGNOSTIC MARKER 703
BIOCHEMISTRY (Moscow) Vol. 90 No. 6 2025
Table 1. Structural proteins of the shelterin complex and their functions [15, 20]
Abbreviation Full name Protein function
TRF1 telomere repeat factor 1
TRF1 homodimer binds to the double-stranded
TTAGGG telomere region and inhibits telomere
elongation by telomerase
TRF2 telomere repeat factor 2
TRF2 homodimer binds to the same region as TRF1,
but prevents recognition of double-strand DNA breaks
(DSBs) as a type of DNA damage that requires repair
POT1 protection of telomere 1
exhibits affinity for single-stranded guanine-rich
telomere regions, protects them from nucleases
RAP1 repressor/activator protein 1 stabilizes TRF2
TIN2
TRF1- and TRF2-interacting
nuclear protein 2
stabilizes the complex of TPP1-POT1 with TRF1 and TRF2
TPP1 (TINT1,
PTOP, PIP1)
telomere-protecting protein 1 binds POT1 to TIN2 and is required for POT1 functioning
decrease in the TL, arrest of cell proliferation, and
cell aging and death.
During each cell cycle, telomeres shorten by 50-
150bp at the DNA replication stage [25]. Critical short-
ening of telomeres impairs their function, leading to
the cell aging or apoptotic death. Chromosomes lack-
ing telomeres may undergo further shortening, result-
ing in the loss of coding genes or fusion with other
chromosomes, which causes genetic instability and in-
creases the risk of cell malignant transformation [26].
A decrease in the TL or prolonged DDR lead to the
cessation of proliferation and activation of cell senes-
cence. Senescent cells actively secrete proinflammato-
ry cytokines (IL-6, IL-8, TNFα, IL-1, and CCL), growth
factors (HGF and IGFBP), metalloproteinases (enzymes
that degrade the extracellular matrix), reactive oxy-
gen species (ROS), and nitric oxide, i.e., acquire the
so-called senescence-associated secretory phenotype
(SASP). The cytokines secreted by senescent cells can
induce senescence of neighboring cells and provoke
body aging [27-29]. Therefore, telomere shortening
can be considered as a marker of replicative aging of
proliferating cells, while stable TL is an indicator of
a healthy state.
The TL as a biomarker of aging or some pathol-
ogies is typically assessed in leukocytes or peripheral
blood lymphocytes. Analysis of human tissues from
the Genotype-Tissue Expression (GTEx) Project collec-
tion [30,  31] allowed to characterize the TL in more
than 6300 samples from over 20 tissue types obtained
from 952 individuals. It was shown that variations in
the TL are determined by the tissue type, donor, do-
nor age, and to a lesser extent by race or ethnicity,
smoking, and hereditary variants. Generally, the TL in
the whole blood cells correlated with the TL in most
tissues. The TL was the shortest in the whole blood
cells and the longest in the testis. In most tissues, the
TL was inversely associated with age, and this associ-
ation was the strongest in the tissues with a shorter
average TL. The results of this study demonstrated
that changes in the TL in the blood cells reflected the
TL features characteristic of all other tissues of an
examined individual [31].
Similar results were obtained for the compari-
son of the TL in human brain and peripheral tissues
[32]. DNA was isolated from saliva, buccal epitheli-
um, blood, and brain tissue from the patients under-
going neurosurgery for the intractable epilepsy, and
average TL was assessed by qPCR. The highest cor-
relation was detected between the TL values in the
brain and buccal samples. This study was unique, be-
cause the authors were able to directly compare the
TL in the brain and peripheral tissues from living
subjects. The correlation between the TL values in si-
multaneously collected buccal samples and leukocytes
was sufficient to suggest the use of buccal DNA as a
reliable non-invasive source for determining the TL
in humans [33].
Currently, the TL is commonly considered as an
indicator of human health. The maximal TL is ob-
served in children at the age of 18 months. Then, the
TL rapidly decreases, gets stabilized by the age of 5,
and remains at ~12  kb until the age of 25. After this,
the TL steadily decreases and could reach 5  kb by the
age of 80 [34, 35]. The TL depends not only on the
age, but also on the individual’s genetic features and
the action of various damaging factors experienced by
a person throughout the lifetime.
MOSKALEVA et al.704
BIOCHEMISTRY (Moscow) Vol. 90 No. 6 2025
Fig. 2. The role of CST–Polα/Prim in the synthesis of telomere ends. Telomerase elongates the G-overhang of the leading DNA
strand, while CST–Polα/Prim fills in the complementary C-strands of the lagging and leading DNA strands at the telomere end.
When the replisome reaches the telomere end, the synthesis of the last Okazaki fragment begins >40nucleotides (nt) from
the site of the double-stranded DNA unwinding (according to [46]). Polδ and Polε are DNA polymerases δ and ε, respectively.
Telomere maintenance mechanisms. As men-
tioned above [4, 7], the TL in proliferating cells is
maintained by telomerase and CST–Polymerase  α/pri-
mase (CST–Polα/Prim) [36,  37]. In some normal and tu-
mor cells, the TL is maintained by the homologous re-
combination of telomeric DNA. During this process, the
3′ overhang of a short telomere of one chromosome is
inserted into the telomere of another “long” chromo-
some and uses it as a template for DNA synthesis with
the participation of enzymes and factors required for
homologous recombination. This mechanism is called
alternative lengthening of telomeres (ALT) [26,  38,  39].
Under certain conditions (e.g., when DNA is dam-
aged), the elongation of telomeres may switch from
the telomerase-catalyzed process to the ALT [40]. In
most proliferating cells, the main mechanism of the
TL maintenance is the synthesis of telomeric DNA by
telomerase and CST–Polα/Prim, but under stress con-
ditions, even these cells can simultaneously use the
ALT mechanism.
Telomerase and CSTpolymerase α/primase. Telo-
merase is a ribonucleoprotein complex consisting of
the catalytic subunit (telomerase reverse transcriptase,
TERT) and the RNA component (TR, or TERC) that acts
as a template for the synthesis of telomeric repeats
at the ends of eukaryotic chromosomes [4, 7]. The
template for the synthesis of telomeric repeats in the
hTR is the 5′-CUAACCCU-3′ sequence. The template
binds complementarily to the protruding 3′ end of
the telomere and is used to synthesize new telomeric
5′-TTAGGG-3′ repeats in vertebrates [41-43]. Telomer-
ase adds G-rich telomeric repeats (TTAGGG)
n
to the
3′ ends of telomeres, thereby counteracting telomere
shortening caused by the loss of telomeric 3′ over-
hangs during the synthesis of the DNA leading strand.
However, telomerase does not compensate for the loss
of DNA sequence at the 5′ ends of chromosomal DNA,
which occurs during the synthesis of the lagging DNA
strand, as well as by partial degradation of the termi-
nal fragments of telomeric DNA by nucleases.
Until recently, the mechanism of synthesis of
the (CCCTAA)
n
ends of the telomere complementary
C-strand had remained poorly understood [44]. In re-
cent years, several excellent experimental papers and
reviews on this problem have been published [36, 37,
45, 46].
The replication of the C-strand end region of telo-
meres in vivo requires the CST protein complex that
acts as an auxiliary factor for the activity of CST–Polα/
Prim received its name from the first letters of the
names of proteins in its content: CTC1 (conserved telo-
mere component  1), STN1 (suppressor of cdc thirteen),
and TEN1 (telomeric pathways in association with
STN1, number  1) [44, 47]. The elongation of telomeres
TELOMERE LENGTH AS PROGNOSTIC MARKER 705
BIOCHEMISTRY (Moscow) Vol. 90 No. 6 2025
begins at the end of the S phase. First, the 3′  G-end is
elongated by telomerase, and then the complementary
C-strand is synthesized by CST–Polα/Prim (Fig. 2).
Regulation of telomerase activity. The catalytic
core of human telomerase exists invivo as a function-
ally cooperative dimer of two reverse transcriptase
subunits (hTERT and hTR) or as a multimer [48]. In
addition to the proteins of the shelterin and CST com-
plexes, hTERT can bind p23/p90 (a chaperone respon-
sible for the complex assembly and conformation),
nuclear localization protein 14-3-3, and TP1 protein
with an unknown function [49]. hTR can bind hGAR1,
Dyskerin/NAP57, hNHP2, and C1/C2 (proteins responsi-
ble for the RNA stability, maturation, and localization),
proteins providing binding to the telomeres, and some
other proteins [50].
The gene coding for hTERT consists of 16 exons.
Its length is 37  kb, of which ~33  kb are introns and
the rest ~4  kb encodes the transcript [51]. The TA can
be regulated through different mechanisms, including
gene expression, phosphorylation/dephosphorylation,
alternative splicing, etc. [52,  53]. Humans express
more than 20 telomerase isoforms. The N-terminal
fragment of telomerase contains the nuclear localiza-
tion signal, RNA interaction domains, and a sequence
that directs it to the mitochondria. The C-terminal
part contains regions that determine the reverse
transcriptase activity of hTERT. In most telomerase
isoforms, these critical elements of protein structure
are disrupted to various degrees. Only the full-length
enzyme, whose mRNA contains all 16 exons, displays
a high TA. All known hTERT splice variants in hu-
man and mouse cells are inactive and, moreover, in-
hibit the TA. This suggests that the hTERT isoforms
formed by alternative splicing, play an important role
in the TA regulation and implementation of telomer-
ase non-canonical functions unrelated to its catalytic
activity [54].
The recruitment of telomerase to telomeres is me-
diated by the shelterin complex proteins POT1–TPP1
[42], whereas timely termination of telomere elonga-
tion occurs with involvement of the CST heterotri-
meric complex composed of the CTC1, STN1, and TEN
proteins [55, 56]. The CST complex competes with the
telomerase processivity factors POT1–TPP1 for binding
to the single-stranded region of telomeric DNA and
inhibits the TA by preventing enzyme binding to the
single-stranded region of DNA and its physical inter-
action with POT1–TPP1. The binding of the CST com-
plex to the telomere increases during the late S/G2
stages only when the telomerase is active and results
in the inhibition of its activity. Removal of the CST
complex leads to the excessive TA and promotes ex-
cessive telomere elongation. By binding to the elon-
gated telomere, the CST complex interrupts the action
of telomerase. Therefore, the elongation of the telo-
mere 3′ G-end is determined by the sequence of three
events (Fig. 2) which first turn on and then stop the
telomerase-mediated telomere elongation: (i)telomer-
ase recruitment to telomeres in the region of the sin-
gle-stranded 3′ G-end with participation of the POT1–
TPP1 proteins; (ii)elongation of the telomere 3′ G-end
by telomerase; (iii) displacement of the POT1–TPP1
proteins from the complex with the single-stranded
3′ G-end by the CST complex and blockade of telo-
merase binding to the 3′ G-overhang, resulting in the
cessation of telomere elongation [37, 45, 47, 49]. The
synthesis of the complementary C-strand by CST–Polα/
Prim begins after completion of the G-strand elon-
gation [46, 57].
Hence, the CST complex plays a key role in the
regulation of two steps of telomere replication: termi-
nation of the G-strand elongation by telomerase and
recruitment of CST–Polα/Prim to the newly synthe-
sized G-overhang and its activation for the synthesis
of the complementary C-strand.
A long non-coding RNA with the telomeric repeats
at the 3′ end, called TERRA (telomeric repeat-contain-
ing RNA), may play an important role in the TA reg-
ulation at the epigenetic level [58] (see reviews [59,
60] for the TERRA formation and properties). It was
shown that telomeres can be actively transcribed by
RNA polymerase II from promoters located in the
subtelomeric regions in the direction from the cen-
tromere toward the end of telomeres with the forma-
tion of the TERRA. The length of this RNA can vary
from 100nt to 9 kb [58]. The 5′-UUAGGG-3′ repeats in
TERRA interact with the hTR sequence. In addition,
TERRA binds to the TERT protein independently of its
interaction with hTR. TERRA is not used as a telomer-
ase substrate. Instead, it acts as a potent competitive
inhibitor of telomerase interaction with the telomer-
ic DNA [61]. TERRA colocalizes with the telomerase
hTR in the nucleoplasm and at telomeres at different
phases of cell cycle.
Most TERRA molecules are diffusely located in
the nucleoplasm. One of the functions of this soluble
pool is telomerase binding and regulation of its activ-
ity. TERRA controls the TL homeostasis by regulating
the TA at the telomeres. High TERRA expression was
found to correlate with the content of short telomeres
in various cell lines, which supports the model of
TERRA as a negative regulator of TA [62].
TERRA may be regarded as a structural compo-
nent of the telomere binding to the telomere through
the shelterin complex proteins TRF1 and TRF2, het-
erochromatin protein  1, histone H3trimethylK9, and
some other proteins [63]. Telomeric DNA exists in the
cell as a component of heterochromatin. TERRA is
currently considered as a key regulator of telomere
maintenance and heterochromatin formation at telo-
meres [59, 63].
MOSKALEVA et al.706
BIOCHEMISTRY (Moscow) Vol. 90 No. 6 2025
Xu and Komiyama [64] summarized the data
on the structure and conformation of G4s in DNA,
RNA, and DNA–RNA hybrids in human telomeres
and showed that TERRA plays a crucial role in the
telomere capping through the formation of telomer-
ic G4s [64]. Notably, TERRA exhibits a preference for
the formation of G4s in the DNA–RNA hybrids at the
3′end of telomeric DNA. Intramolecular G4s with dif-
ferent folding structures and loop conformations and
experimentally determined molecular structures have
been shown for the parallel, hybrid, and basket G4s
in [14, 64] and for intermolecular hybrid DNA–RNA
G4s in [64]. It should be noted that G4s are mobile
structures capable of unfolding, in particular, with
the participation of POT1 protein  [65]. Some small
G4 ligands stabilize G4s, resulting in telomerase in-
hibition [66]. Telomere damage triggers TERRA tran-
scription, so that the upregulation of TERRA can be
considered as a molecular marker of telomere desta-
bilization [67].
TERRA can form RNA–DNA hybrids with the cyto-
sine-rich telomeric DNA strands. The telomeric strand
containing the TTAGGG repeats is displaced, resulting
in the formation of the R-loop structure  [68] crucial
for the telomere maintenance  [69]. Excessive TERRA
R-loops can cause replicative stress, chromosome dam-
age, and loss of telomeres [69,  70]. TERRA and TERRA
R-loops are highly abundant in human cancer cells, in
which telomere elongation occurs via the ALT mech-
anism [71].
It has been recently shown that translation of
TERRA yields two proteins consisting of repeating
dipeptides: a highly charged protein formed by va-
line-arginine (VR) repeats and a hydrophobic pro-
tein formed by the glycine–leucine (GL) repeats [72].
The VR protein binds nucleic acids and localizes at
the replication forks. Both VR and GL proteins form
long 8-nm filaments with the amyloid properties. The
authors showed that cells with the elevated levels of
TERRA in the nuclei contained three to four times
more VR protein than the original cell line. Deletion
of TRF2 caused the dysfunction of telomere and in-
creased the content of VR protein, while manipulation
of TERRA levels using specific approaches resulted in
the appearance of large VR aggregates in the nucle-
us. These data suggest that telomeres, particularly in
cells with dysfunctional telomeres, can express two
unusual dipeptide-repeat proteins with potentially
important biological properties [72]. These new find-
ings suggest that despite extensive research into the
role of TERRA in the regulation of telomere function,
many unanswered questions still remain.
Alternative functions of TERT and TR. The exten-
sive studies of telomerase have provided new data
on the functions of holoenzyme components that are
not associated with the telomere elongation per  se,
but play an important role in ensuring cell survival
under the influence of damaging factors. Both TERT
and TR may be involved in the regulation of various
intracellular processes, including gene expression and
stress response.
Alternative functions of TERT have been dis-
cussed in [43, 73-76]. An important property of TERT is
its ability to protect cells from the damage caused by
oxidative stress. Alternative functions of TERT include
not only the antiapoptotic and antioxidant effects,
but also the protection against specific DNA-damag-
ing agents. TERT can translocate between the nucleus,
cytoplasm, and mitochondria due to the nuclear ex-
port signal identified at the protein N-terminus. In the
case of oxidative stress, TERT is phosphorylated at the
tyrosine residue 707 by the Src kinase, which triggers
its nuclear export. In addition, TERT contains a spe-
cific N-terminal 20-amino acid sequence that serves
as a mitochondrial localization signal. The transport
of TERT into the mitochondrial matrix involves trans-
locases of the outer and inner mitochondrial mem-
branes.
The role of TERT in the mitochondria was dis-
cussed in [75]. hTERT-overexpressing non-prolifer-
ating human fibroblasts were characterized by the
reduced ROS generation and upregulated expression
of antioxidant defense proteins. In these cells, the ox-
idative stress induced by X-rays or H
2
O
2
caused less
damage to the mitochondrial membrane potential and
mitochondrial morphology than in normal fibroblasts.
Both genotoxic factors (X-rays and H
2
O
2
) significant-
ly increased the number of γH2AX foci (phosphory-
lated form of H2AX histone, DNA damage marker).
The DNA damage induced by X-ray irradiation in the
control cells persisted for many days, while in the
hTERT-overexpressing fibroblasts, this damage was
considerably less and DNA was fully repaired within
the following days.
It should be noted that the early studies on the
telomerase content in different tissues have shown the
presence of hTERT in proliferating cells, while hTR
has been found in almost all tissues [41], suggesting
that hTR may have the functions unrelated to the
TA regulation. It was later discovered that the hTR
knockdown reproducibly induced apoptosis in the ab-
sence of any detectable telomere shortening or DDR
activation. In contrast, the knockdown of hTERT did
not induce apoptosis [76]. The authors suggested that
hTR can function as a non-coding RNA that protects
cells from apoptosis independently of its involvement
in telomere lengthening by telomerase in normal hu-
man cells [76].
Possible non-telomeric functions of hTR have
been discussed in reviews [77, 78]. Numerous studies
have provided evidence on the implication of hTR in
protective mechanisms. Thus, hTR contains an open
TELOMERE LENGTH AS PROGNOSTIC MARKER 707
BIOCHEMISTRY (Moscow) Vol. 90 No. 6 2025
Fig. 3. Telomerase functions in the cell. The TL is maintained through the synthesis of telomeric DNA by telomerase or
via the ALT pathway, which is necessary for the T-loop formation and chromosome preservation. Telomerase components
TERT and TR possess the protective functions. TERT can migrate to the mitochondria and increase the level of antioxidant
defense and anti-apoptotic activity in these organelles. TR can serve as a template for the synthesis of TERP protein, which
exhibits the anti-apoptotic activity. TERRA reduces the TA and stimulates the ALT pathway to maintain the TL.
reading frame that starts at position 176 and codes
for a 121-amino acid protein named hTERP. hTERP is
encoded as an immature transcript. The existence of
hTERP has been confirmed by several experimental
approaches including mass spectrometry and immu-
nodetection [79]. The overexpression of the wild-type
hTR (but not its mutant version incapable of directing
hTERP synthesis due to mutation in the start codon)
protected HEK293T cells from the doxorubicin-induced
apoptosis. hTERP may be involved in cell protection
from stress and promotion of cell survival under the
action of damaging factors and during adaptation to
unfavorable conditions. This alternative activity of
telomerase components may play an important role
in ensuring cell survival under the influence of dele-
terious factors.
The known functions of telomerase ensuring cell
stability are shown in Fig. 3.
Therefore, the maintenance of TL in proliferating
cells by telomerase and CST–Polα/Prim or via the ALT
mechanism, the antiapoptotic activity of telomerase
components TERT and TR, and the antioxidant activity
of TERT in the mitochondria provide cell resistance to
the action of damaging factors.
Telomerase and telomere maintenance in tumors.
Telomerase expression is suppressed in resting cells,
but is reactivated upon their malignant transforma-
tion. High expression of telomerase gene and high TA
have been detected in various types of malignant tu-
mors [26,  52,  80]. Therefore, the presence of TA in a
tissue extract can be used as a marker in the diagnos-
tics of malignant tumors. Unlimited cell proliferation
facilitated by telomerase reactivation is a hallmark of
cancer, as it enables long-term proliferation of cells
with short telomeres.
In 5-15% tumors, the TL is maintained by the ALT
mechanism [26,  38,  81]. ALT is frequently found in
tumors of mesenchymal origin: in 60% of soft tissue
sarcomas, 100% of chondrosarcomas, 63% of undiffer-
entiated pleomorphic sarcomas, 53% of undifferenti-
ated leiomyosarcomas, and 14% of fibrosarcomas. In
CNS tumors, ALT was found in 63% of diffuse and
anaplastic astrocytomas and in 44% of glioblastoma
multiforme in children (but only in 11% tumors in
adults).
No association between the TL in peripheral
blood leukocytes and risk of cancer development has
been established for solid cancers [82]. At the same
time, there is a correlation between the reduced TL
in leukocytes and various hematological malignancies
[83]. Therefore, the assessment of TL and TA for the
diagnosis and prognostics of cancer should be per-
formed only in tumor biopsies or surgically removed
tumor tissues.
MOSKALEVA et al.708
BIOCHEMISTRY (Moscow) Vol. 90 No. 6 2025
The high level of TA in malignant tumors may
be due to mutations in the TERT gene promoter or
genes coding for the shelterin and CST complex pro-
teins; itcan also be determined by specific expression
patterns of numerous transcription factors that regu-
late the TA [84].
The use of TA analysis in the early diagnostics of
bladder cancer seems particularly promising due to
the possibility of non-invasive TA measurement in the
urine. Therefore, the TA assay might become a gold
standard for the non-invasive diagnostics of bladder
cancer [85, 86].
A pilot study demonstrated a high value of the
urine and tissue TA levels (measured by RT-TRAP-
2PCR) in detection and monitoring of bladder cancer.
An association was found between the tumor size
(≥3  cm) and elevated TA levels in tissues and urine.
However, no such correlation was observed between
the higher TA levels and tumor grade/size at the ad-
vanced cancer stages [87,  88]. It should be noted that
TA assays are available, but they have not yet been
introduced into clinical practice because of the high
cost and problems with the standardization of pro-
cedures for the sample collection and preparation.
At the same time, preparation of samples for the cur-
rently used histological examination is less compli-
cated and can be automated. A more promising ap-
proach for the diagnostics of malignant tumors and
monitoring of the therapy efficacy is a rapidly devel-
oping analysis of specific marker mutations in liquid
biopsies. For example, the C228T and C250T mutations
in the TERT gene promoter play an important role
in the malignant transformation of cells and can be
used as biomarkers in many types of cancer, including
glioblastoma multiforme, as they are detected in 80%
of glioblastoma multiforme cases [89, 90].
To summarize the above, we should emphasize,
in most proliferating cells the TL is maintained by
telomerase and CST–Polα/Prim with the participation
of DNA polymerases δ and ε. The TA is regulated at
the transcriptional level, by post-transcriptional mod-
ifications, and epigenetically. Proteins of the shelterin
and CST complexes, as well as numerous other pro-
teins and regulatory factors involved in the TL main-
tenance, have been intensively explored. At the same
time, the telomerase components hTERT and hTR
perform a number of functions aimed at increasing
cell resistance to the stress factors. In some cells and
tumors, the TL is maintained through the ALT mech-
anism. Reactivation of telomerase during malignant
cell transformation ensures the long-term prolifera-
tion of tumor cells with short telomeres. The pres-
ence of TA and mutations in the hTERT gene promoter
(C228T and C250T) may be the hallmarks of malignant
tumors, with the exception of sarcomas and some
brain tumors.
TELOMERE SENSITIVITY
TO IONIZING RADIATION
DNA damage and telomere DNA repair. Among
the types of molecular damage caused by IR, forma-
tion of DSBs in DNA has the most severe consequences
for the cells, because persistence of unrepaired DSBs
leads to cell death. DSB repair occurs through the two
main mechanisms: homologous recombination, which
occurs only during the S and G2 phases of cell cycle,
and classical non-homologous end joining (c-NHEJ)
that takes place during all phases of cell cycle [91-93].
In mammalian cells, DSBs are repaired by both c-NHEJ
and homologous recombination mechanisms. The cells
can also use the alternative end-joining mechanism,
which is associated with the loss of nucleotides, re-
quires the presence of homologous sequences in the
same chromosome, and leads to the appearance of er-
rors in the DNA structure. Alternative end-joining is
a less studied mechanism; however, it is known to in-
volve PARP1, DNA polymerase θ, Lig1 ligase, and Lig3–
XRCC1 complex [91-95]. It is employed when c-NHEJ or
homologous recombination cannot function properly.
In addition to the above-mentioned mechanisms,
DSBs can be repaired via the RAD51-independent ho-
mology-dependent single-strand sequence annealing,
i.e., joining of the ends of two homologous 3′  single-
stranded DNA fragments in the region of tandem
sequences, which leads to the obligatory removal of
DNA fragment between the repeats. This mechanism
functions only during the S and G2 phases of cell cy-
cle and is associated with the appearance of errors in
DNA and increased frequency of mutations [93].
The development of DDR in the telomere region
can lead to the disturbances in the telomere structure;
however, reactions that are dangerous for the cell
can be suppressed by the shelterin complex proteins
(Table1). Thus, TRF2 binds the ATM kinase and inhibits
the ATM-dependent DDR [96]. It was found that TRF2
interacts with DDR factors not only at the telomeres,
but also in other chromosomal regions. Phosphoryla-
tion of TRF2 at serine 20 reduced its binding affini-
ty to the telomeres and promoted TRF2 relocation to
non-telomeric DSBs, thus facilitating their repair [97].
Another protein of the shelterin complex, POT1,
prevents activation of the ATR kinase involved in the
single-stranded DNA repair, thus arresting DDR in the
telomeres [98]. TRF2 and POT1 act independently to
suppress the two DDR pathways. It was found that
X-ray irradiation of fibroblasts was followed by a
long-term presence of DDR markers at the telomeres,
while in other chromosomal regions, such damage
was quickly eliminated by DNA repair. No telomere
shortening was observed [28, 99]. The prolonged
presence of unrepaired DSBs in the telomeric DNA
leads to the DDR persistence, arrest of proliferation,
TELOMERE LENGTH AS PROGNOSTIC MARKER 709
BIOCHEMISTRY (Moscow) Vol. 90 No. 6 2025
and activation of cell senescence [27, 29]. There is ev-
idence that TERRA is also involved in the regulation
of DSB repair in DNA [100, 101].
The features of the telomere response to DNA
damage have been discussed in detail in comprehen-
sive reviews [12, 13, 15, 17]. As telomeres constitute
only a small fraction of the human genome, the proba-
bility that photons or accelerated particles will direct-
ly cross the telomeric sequence is very low. Therefore,
the IR-induced telomere damage likely occurs due to
disruptions in the telomere maintenance mechanism.
Telomere damage as a result of ionizing radia-
tion-induced oxidative stress. IR causes both direct
damage to the cell macromolecules and indirect dam-
age through the water radiolysis products [28, 102].
Irradiation of water results in the formation of ROS,
reactive nitrogen species and other free radicals ca-
pable of damaging DNA and other macromolecules.
The effects of irradiation can be manifested within
several minutes to several hours after exposure. The
most reactive species are superoxide anions, hydroxyl
radicals, nitric oxide radicals, lipid radicals, and some
other molecules with a high oxidizing potential. ROS
production can also be a result of activation of some
cellular metabolic processes, as well as inflammation,
ischemia, and stress [103].
One of the most prevalent forms of oxidative
DNA damage is 8-oxo-7,8-dihydro-2′-deoxyguanosine
(8-OxodG), a product of guanine oxidation. Its accu-
mulation in DNA is considered a hallmark of chron-
ic oxidative stress [104]. Mitochondrial DNA is more
susceptible to the oxidative damage than nuclear DNA
[105, 106]. Thus, an exposure to IR induced a multi-
fold increase in the 8-OxodG content in mitochondrial
DNA [107].
Densely IR is a more potent inducer of the
long-lasting oxidative stress than sparsely IR (gam-
ma- and X-rays) [104, 108, 109]. The oxidative damage
to telomeric DNA is thought to cause premature se-
nescence by accelerating telomere shortening. Barnes
et al. [110] tested this model directly using a preci-
sion chemoptogenetic tool that generated 8-OxodG
exclusively in the telomeres. It has been shown that
a one-time induction of the 8-OxodG formation in the
telomeres was sufficient to trigger the p53-dependent
senescence. Formation of 8-OxodG activated the ATM
and ATR signaling and resulted in telomere dysfunc-
tion in replicating cells [110]. Chronic selective induc-
tion of telomeric 8-OxodG shortened the telomeres and
impaired cell proliferation over time. Accumulation
of telomeric 8-OxodG in chronically exposed 7,8-di-
hydro-8-oxoguanine-DNA glycosylase-deficient cells
triggered the replication stress, significantly increased
the loss of telomeres, and generated chromosome fu-
sions, leading to the formation of chromatin bridges
and micronuclei during cell division [111]. Targeted
oxidative stress directed toward either telomeres or
mitochondria increased the ROS production, extent of
telomere damage, mitochondrial dysfunction, and cell
apoptosis [112]. Figure 4 illustrates the relationship
between the development of disorders caused by the
elevated levels of ROS and other free radicals and de-
crease in the TA and TL.
The damage and shortening of telomeres as a re-
sult of oxidative stress development occur throughout
the lifetime of an organism against the background
of normal metabolism and influence of various dam-
aging factors and inflammation, which explains the
decrease in the TL in human peripheral blood leuko-
cytes with age [113-115]. The damaging factors causing
telomere shortening include chemotherapeutic drugs
and IR [116].
The effect of irradiation on the telomere–telo-
merase system. The IR-induced changes in the TL
depend on the type of radiation, its duration, dose
received, and time after exposure.
Analysis of TL and chromosomal instability in
the prostate cancer patients subjected to the intensi-
ty-modulated radiotherapy (IMRT) showed individual
response to the therapy: 11 patients with a relative-
ly short telomeres at baseline showed a dramatic
increase in the TL immediately post IMRT that was
sustained for at least 3 months. In patients with ini-
tially longer telomeres, the TL dramatically decreased
post IMRT [117].
The telomere–telomerase system was found to be
highly sensitive to IR. Proteins of the shelterin com-
plex play an important role in protecting telomeric
DNA from the IR-induced damage. Thus, it was shown
that the siRNA-mediated inactivation of TRF1, TRF2,
or POT1 led to the impairment of telomere protective
functions and the increase in the frequency of spon-
taneous and radiation-induced mutations in the het-
erozygous thymidine kinase locus in human lympho-
blastoid WTK1 cells after exposure to γ-radiation and
accelerated
56
Fe ions with an energy of 1 GeV/n at the
doses of 1 and 2 Gy. The highest increase in the muta-
tion rate was observed upon inactivation of POT1 [118].
The effect of IR on the TA varies depending on
the cell type, dose, type of radiation, and TL [118]. For
example, after exposure of human breast adenocarci-
noma MCF-7 cells to IR, the TA increased within the
first 48 h, but then decreased. These changes were
accompanied by changes in the TL. Irradiation of cul-
tured cells at a dose of 10Gy caused a decrease in the
average TL 5 days after the exposure, followed by the
partial TL recovery after 10 days [118], indicating dis-
appearance of short telomeres that had appeared af-
ter irradiation. However, it remained unclear whether
this phenomenon was caused by the actual recovery
of telomeres by telomerase or via the ALT mechanism
or by the death of cells with short telomeres.
MOSKALEVA et al.710
BIOCHEMISTRY (Moscow) Vol. 90 No. 6 2025
Along with the decrease in the TL, the post-irra-
diation disturbances in the TA-TL system included an
increase in the proportion of senescent (β-galactosi-
dase-positive) cells to 45% (MCF-10A cells) and 70%
(MCF-7 cells) on day 10 after irradiation [118]. The
results of this detailed study suggest a high sensitivity
of the TA-TL system to the effects of IR in both normal
and tumor cells.
An increase in the TA after irradiation has been
shown in various tumor cells, such as glioma [119]
and nasopharyngeal carcinoma cells [120], colon carci-
noma and colorectal carcinoma cell lines [121], Ewing
sarcoma SK-N-MC cells, breast cancer MCF-7 cells,
chronic myelogenous leukemia K562 cells [122], etc.
The upregulation of the TA after irradiation can be
considered as a protective response leading to the in-
creased radioresistance of tumor cells. In this regard,
there is currently an active search for the efficient
and selective TA inhibitors to increase the sensitivity
of tumors to the radiation therapy.
Radiation damage to regional stem cells may be
an important mechanism in radiation-induced car-
cinogenesis and some delayed post-irradiation effects.
Stem cells are found in almost all tissues of an adult
organism. They play an important role in the main-
tenance of cellular homeostasis and post-damage tis-
sue regeneration. Since stem cells are slowly renewed
throughout the lifetime, they can accumulate genetic
damage, which can cause their malignant transforma-
tion, formation of cancer stem cells, and cancer devel-
opment, in particular, emergence of secondary tumors
after radiation therapy. Therefore, many studies of the
mechanisms of radiation carcinogenesis are currently
focused on the effects of different types of radiation
on stem cells. In addition, a decrease in the stem cells
pool due to the whole-body or regional exposure to
radiation at the doses that cause the death of some
stem cells can lead to the accelerated aging of organs
and tissues and slow down regeneration processes.
γ-Irradiation at a dose of 0.1  Gy increased the TA
in cultured mouse bone marrow mesenchymal stro-
mal cells (MSCs) 2 months after irradiation, while γ-ir-
radiation at a dose of 6  Gy and exposure to neutrons
at the doses of 0.05, 0.5, and 2  Gy decreased the TA.
At the same time, only γ-irradiation, but not exposure
to neutrons decreased the TL in the irradiated vs. con-
trol MSCs [123]. The absence of telomere shortening in
MSCs with a low TA suggests that either the existing
TA was sufficient to maintain the TL or that the cells
utilized both the TA and ALT to restore the TL.
In subsequent experiments, irradiated MSCs were
cultured for 2 months and then transplanted to mice
to induce tumors (sarcomas). Next, fibrosarcoma cell
lines were prepared from these formed tumors. In
cells of the fibrosarcoma lines, the TA was either ab-
sent (after γ-irradiation at a dose of 1  Gy and after
neutron irradiation at a dose of 0.5  Gy) or remained
at a very low level. Hence, malignant transformation
of MSCs was accompanied by the decrease in the TA
(up to its complete disappearance), which distinguish-
es sarcomas from other types of tumors, which are
characterized, on the contrary, by telomerase reacti-
vation [123].
TL was investigated as a marker of replicative
aging of bone marrow and thymus cells after pro-
longed irradiation of mice with neutrons at the doses
of 0.05-0.5  Gy  [124]. It was found that the TL in the
bone marrow cells and the thymus in control animals
aged 4 and 16 months was similar and did not change
during mice aging. In the irradiated animals, the TL
in the bone marrow and thymus did not differ from
the TL in the corresponding organs of the control
mice 2 months after exposure; however, 14 months
after the irradiation, the decrease in the TL was pro-
portional to the radiation dose in both bone marrow
and thymus cells [124]. Analysis of internal organs in
the euthanized animals found that 2 out of 10 C57Bl/6
mice had uterine tumors (squamous cell keratinizing
carcinomas) and 1 out of 10 CBA mice had uterine
carcinosarcoma. One of the CBA mice irradiated at
a dose of 50 mGy had invasive bronchoalveolar ad-
enocarcinoma in the left lung. These data attest that
neutron irradiation caused a delayed decrease in the
TL in the bone marrow and thymus and increased
the probability of tumor development. The appear-
ance of cytogenetic abnormalities in bone marrow
cells was observed at the irradiation dose of 10 mGy
and above [125].
Telomeres are more sensitive to IR than chromo-
somal DNA. Analysis of the association between the
TL and sensitivity of TK6 lymphoblasts to IR revealed
no correlation between the radiosensitivity and mean
TL; however, there was a positive correlation between
the radiosensitivity and loss of telomeres, suggesting
it is the irradiation-induced telomere loss, rather
than changes in the TL itself, that plays an important
role in the formation of radiosensitivity or radiore-
sistance [126].
Experiments in telomerase-deficient mice generat-
ed by the deletion of the telomeric RNA gene (Terc),
have shown the role of telomerase and telomeres in
the response of cells and entire organism to IR. It was
found that the telomere dysfunction in the late gen-
erations of Terc
–/–
mice has led to the development
of the increased radiosensitivity syndrome associated
with accelerated mortality, intensification of apopto-
sis in intestinal cells and thymocytes, and elevated
radiosensitivity of embryonic fibroblasts. The radio-
sensitivity of these cells correlated with a decrease
in the DDR rate, persistent chromosomal breaks, and
cytogenetic profiles characterized by complex chromo-
somal aberrations [127].
TELOMERE LENGTH AS PROGNOSTIC MARKER 711
BIOCHEMISTRY (Moscow) Vol. 90 No. 6 2025
Fig. 4. Mechanisms of TA suppression and TL shortening in cells subjected to IR and oxidative stress. 8-OxodG, 8-oxo-7,8-di-
hydro-2′-deoxyguanosine; R-loops, RNA–DNA hybrids formed by TERRA with the cytosine-rich sequences of telomere DNA
that can cause displacement of DNA strand containing TTAGGG repeats.
DNA damage triggers the DDR response, which
causes cell cycle arrest until the damage is repaired.
The presence of unrepaired DNA damage induces per-
sistent DDR activation, leading to the cell apoptosis
and/or senescence. It is important to note that the re-
pair of DNA damage in the telomere region is limit-
ed [28,  128], which determines a higher sensitivity of
telomeres to IR compared to the rest of the genome.
The mechanisms underlying this phenomenon require
further study. Figure 4 shows the sequence of events
leading to the TA decrease and TL shortening as a
result of direct damage by IR and the action of ROS,
reactive nitrogen species, and free radicals, as well
as by the oxidative stress caused by ROS and reactive
nitrogen species generated in pathological processes
not associated with radiation.
Delayed effects of the radiation exposure on
the telomere length. The biological effects of the
long-term exposure to a low-dose IR are attributed to
the direct DNA damage and indirect effect mediated
by the oxidative changes in DNA. As specific nucleo-
protein structures with a high guanine content, telo-
meres are more sensitive to oxidative damage and
can be damaged and shortened more easily under
the combined effect of radiation and oxidative stress
caused by it.
In the study of Hiroshima atomic bomb survi-
vors, Lustig et al. [129] showed that the TL in leu-
kocytes 50 and 68 years after the exposure inversely
correlated with the radiation dose. The results indi-
cate a long-term negative effect of IR on the TL in
leukocytes, which depended on both the irradiation
dose and the age of subjects at the time of exposure
[129]. A nonlinear relationship between the TL in T
lymphocytes and radiation dose was also demonstrat-
ed in a study of 620 individuals who had survived
the atomic explosion, which discovered the presence
of longer telomeres in individuals who had received
a lower radiation dose and a trend to the TL de-
crease in subjects who had received a dose higher
than 0.5 Gy [130].
Scherthan et al. [131] examined 100 workers of
the plutonium production plant at the Mayak Produc-
tion Association, who had been chronically exposed to
plutonium-239 α-radiation and/or γ-radiation, for the
relative TL in leukocytes in comparison to the con-
trol group (51 local residents). The mean age of all
participants was about 80 years. The authors found
that the TL decreased with age. The workers irradi-
ated at the lowest dose demonstrated a significant
decrease in the TL (by ~20%) after being exposed to
external γ-radiation (≤1  Gy) or internal α-radiation
(≤0.05-0.1  Gy to red bone marrow). The relative TL
in the workers exposed to a high-dose irradiation
(>0.1  Gy for α-radiation and >1-1.5  Gy for γ-radia-
tion) did not differ from that in the control group.
MOSKALEVA et al.712
BIOCHEMISTRY (Moscow) Vol. 90 No. 6 2025
Stratification by sex revealed a significant (~30%) de-
crease in the TL in the males exposed to the low dos-
es, but not in the females. The authors concluded that
chronic systemic exposure to different radiation doses
leads to different changes in the TL, which may be
due to the selection against cells with a low TL [131].
The TL and telomere damage were also studied
in the individuals working at the diagnostic radiolo-
gy departments and occupationally exposing to low
doses of X-ray radiation. These workers demonstrat-
ed increased levels of chromosomal aberrations and
damaged telomeres in peripheral blood lymphocytes,
as well as increased levels of lipid peroxidation prod-
ucts and 8-OxodG in the blood plasma. The latter two
parameters correlated with the extent of telomere
damage, suggesting that the chronic exposure to a
low-dose radiation causes oxidative modification of
guanine, leading to telomere damage [132].
These data confirm a delayed decrease in the TL
after exposure to a low-dose (including chronic) irra-
diation, as well as significant contribution of oxidative
stress to the telomere damage in individuals subjected
to the chronic low-dose irradiation.
Telomere length and radiosensitivity. The TL
is closely related to the cell radiosensitivity. For in-
stance, it was demonstrated that the radiosensitivi-
ty of human larynx squamous cell carcinoma Hep-2
cells negatively correlated with the TL and positively
correlated with the TA, which allowed the authors to
conclude that both TL and TA can serve as markers
for predicting the radiosensitivity of patients’ cells
[133]. These data have been confirmed in different
types of tumor cells (eight hepatocellular carcino-
ma cell lines and five breast cancer cell lines [134])
and mice [135].
A similar dependence was found for the mouse
cells defective in the telomerase gene. At the same
time, telomerase-deficient mouse fibroblasts immor-
talized by transfection with the telomerase gene dis-
played no correlation between the TL and radiosensi-
tivity (as demonstrated for three cell clones with the
TL of 13, 10, and 4  kb, respectively) [136]. The authors
concluded that the presence of TA causes stabilization
of short telomeres, which prevents the increase in the
radiosensitivity of cells.
A relationship between the TL and radiosensitiv-
ity has been well established. It was shown that the
shortening of telomeres enhances cell sensitivity to
IR, but the molecular mechanisms of this phenome-
non remain unclear. To understand the relationship
between the telomere shortening and radiation sensi-
tivity, Drissi et al. [137] showed that the late-passage
cells with short telomeres are characterized by an
increased sensitivity to IR compared to the early-pas-
sage cells with longer telomeres. Before exposure to
IR, late-passage cells had a higher baseline level of
γH2AX, which reduced after irradiation. The rates of
the appearance and disappearance of γH2AX foci in
irradiated cells decreased, indicating a reduced level
of DNA repair. Ectopic telomerase expression in the
late-passage cells completely restored the kinetics of
formation and resolution of γH2AX foci to the levels
observed in the early-passage cells. These data demon-
strate the role of TL in the DSB repair suppression in
cells with short telomeres [137].
The phosphorylation kinetics of ATM kinase and
p53 was similar in the early- and late-passage cells,
but phosphorylation of the chromatin-bound SMC1
and NBS1 proteins (ATM targets) was reduced in the
late-passage cells. The authors investigated the chro-
matin structure and showed that the late-passage chro-
matin was resistant to digestion with the micrococcal
nuclease and had a reduced level of H3K9 acetylation
and a higher level of H3K9 methylation, indicating its
transition to heterochromatin. Such chromatin con-
version to a more compact state may explain why
phosphorylation and activation of the chromatin-as-
sociated DDR proteins (H2AX, SMC1, and NBS1) were
reduced in the late-passage cells, while activation of
the non-chromatin-associated proteins (p53 and ATM)
remained unchanged. The authors concluded that
short telomeres are associated with changes in the
chromatin structure that limit the access of the acti-
vated ATM kinase to its targets present in the chroma-
tin content and restrict the DDR, which might explain
an increased radiosensitivity of cells with shortened
telomeres [137].
To study the mechanisms underlying the radiore-
sistance of tumors, Zhou etal. [138] generated a radio-
resistant cell line from the human larynx squamous
cell carcinoma Hep-2 cells. The authors showed that
the expression of some genes associated with the DNA
repair, cell cycle, apoptosis, etc. (for example, genes
for telomere proteins, such as POT1) was significant-
ly altered in the radioresistant cells, which also had
a higher TA and longer telomeres than the parental
cells. The authors concluded that both TA and TL may
be indicators of cell radioresistance [138].
Long telomeres and high TA have been commonly
associated with the radioresistance of different can-
cers. The protection of telomeres, telomere function,
and TL depend on the TA and proteins of the shel-
terin and CST complexes. Ferrandon et al. [139] eval-
uated the telomeric status of glioblastoma cells after
the photon and carbon ion irradiation and found a
significant correlation between the TL, basal POT1
expression, and photon radioresistance in vitro. The
authors also observed a significant increase in the sur-
vival of patients with long telomeres or a high POT1
level. Expression of POT1 was a predictive indicator of
the patients’ response irrespectively of the TL. How-
ever, the observed correlations were absent in  vitro
TELOMERE LENGTH AS PROGNOSTIC MARKER 713
BIOCHEMISTRY (Moscow) Vol. 90 No. 6 2025
after the carbon ion irradiation. To identify radiore-
sistant tumors in patients who would benefit from the
carbon ion hadron therapy the authors proposed the
assessment of the TL and POT1 expression in tumor
biopsies [139].
As shown above, both TL and TA can be used as
markers for monitoring the patients’ condition during
radiation therapy. Indeed, it has been shown that the
stability of telomeres in peripheral blood lymphocytes
correlates with the life expectancy of patients receiv-
ing radiation therapy, and that the telomere instabili-
ty correlates with the development of toxic response
long after the radiation therapy [140]. The implemen-
tation of this approach into clinical studies will help
to analyze in more details the dynamics of this indi-
cator in patients subjected to the radiation therapy.
The increase in the radiosensitivity associated
with the TL decrease is not the only problem of cells
and organisms with short telomeres. Another conse-
quence of telomere shortening is an increased risk of
chromosome aberrations and malignant transforma-
tion of cells [82, 128, 141].
The TL stability is currently considered as a
promising marker of normal tissue radiosensitivity
that can be used for predicting the development of
complications in patients after radiation therapy [142].
The telomere–telomerase system in astronauts.
Exposure to IR is the major health risk during future
deep space missions. The keynote paper co-authored
by 38 researchers representing 33 leading aerospace
research institutes and agencies identified six process-
es that shape our current understanding of molecular
changes occurring during space missions: oxidative
stress, DNA damage, dysregulation of mitochondrial
function, epigenetic changes (including regulation of
gene activity), changes in the TL, and microbiome
modification [143].
The TL in blood cells appears to be a relevant in-
tegrative biomarker for studying the impact of space-
flight factors on astronauts, because changes in this
indicator reflect the combined effect of factors that
astronauts encounter in extreme space conditions. The
most striking discovery was the lengthening of telo-
meres during the spaceflight found in all crew mem-
bers. The TL reduced rapidly upon return to Earth, so
that eventually the crew members had significantly
shorter telomeres after the spaceflight than before it.
Chronic exposure to the space radiation and asso-
ciated development of DDR and other stress responses
may promote activation of the TL maintenance path-
ways. Although the exact mechanisms and health ef-
fects of spaceflight-associated shifts in the TL dynam-
ics remain to be elucidated, the existing findings have
already highlighted the importance of monitoring the
TL and genome stability in the cells of spacecraft crew
members for the assessment of the overall health and
risks of disease development and aging. These data
should be taken into account in the creation of per-
sonalized aerospace medicine and protective mea-
sures for future astronauts.
More detailed results of the studies of the TL in
astronauts were presented in the reports from the lab-
oratory of S.  M.  Bailey [144,  145]. The authors found
that the identical twins Scott and Mark Kelly had rela-
tively similar telomeres before the spaceflight. The telo-
meres of Mark Kelly, who stayed on Earth, remained
stable throughout the study, while the telomeres of
Scott Kelly increased in length during the spaceflight
and then shortened rapidly upon his return to Earth.
Overall, the content of short telomeres increased after
the spaceflight. The level of chromosome aberrations
also increased during the spaceflight, and the elevated
frequency of inversions persisted after the flight, indi-
cating long-term genome instability in the astronaut’s
cells after returning to Earth.
During the spaceflight, all crew members experi-
enced oxidative stress that correlated positively with
the TL dynamics. The frequency of chromosomal in-
versions increased significantly during and after the
spaceflight. The authors believed that it was an adap-
tive response of telomeres to chronic oxidative stress
under extreme conditions, because the studied cells
also demonstrated simultaneous ALT activation, as
evidenced by the increase in the chromatid exchang-
es in the telomeres. The radiation doses received by
the astronauts and calculated based on the level of
chromosome inversions were 50-350  mGy, and the
average effective dose over 6  months on the Inter-
national Space Station was 80  mSv. The authors sug-
gested that the observed damage was related to the
oxidative stress (Fig. 4) rather than to the radiation
exposure [145]. This may be true, since no changes
in the TL were found in the residents of regions with
a high natural background radiation (mean dose, 0.5
to 15  mSv/year) despite the presence of cytogenetic
abnormalities [146].
During the long-term flight, the astronauts showed
the signs of inflammation [144], which is always ac-
companied by the increase in the ROS content (oxi-
dative stress).
It should be noted that similar changes in the
TL were registered in the participants of the short-
term “Inspiration 4” civilian spaceflight, namely, an
increase in the TL during the flight and telomere
shortening after returning to Earth [147]. Other de-
tected changes had an adaptive nature.
The above data indicate the possibility of devel-
oping an adaptive response to various factors en-
countered during the spaceflight, including oxidative
stress. The detected changes in the telomeres probably
reflect a universal adaptive response to the damaging
factors.
MOSKALEVA et al.714
BIOCHEMISTRY (Moscow) Vol. 90 No. 6 2025
TELOMERE LENGTH AND TELOMERASE
IN TELOMEROPATHIES
There are several pathological conditions asso-
ciated with mutations in the genes involved in the
mechanisms ensuring the telomere stability. They are
accompanied by disturbances in hematopoiesis, devel-
opment of pulmonary and liver fibrosis, worsening of
nail condition, and changes in the TL. Such conditions
are called telomeropathies, or telomere biology disor-
ders [148, 149]. The TL analysis is of great importance
for the diagnostics and monitoring of telomeropathies,
while clarification of the mechanism of telomere dys-
function in such diseases is necessary for their diag-
nostics, genetic counseling, clinical management of
patients, and choice of therapy.
Dyskeratosis congenita (DC) is a congenital syn-
drome of bone marrow failure and dysplasia of oral
mucosa and skin, which is characterized by the classic
triad of nail dystrophy, spotted hyperpigmentation of
skin, and oral leukoplakia, as well as predisposition to
cancer. Genetic analysis has shown that DC is associat-
ed with a defect in the gene located in the Xq28 locus.
The DKC1 gene encodes dyskerin [150], which is one
of the proteins forming a complex with telomerase.
It is found in the nucleolus, where it binds to RNA
(including ribosomal RNA and hTERC) and influences
many cellular functions.
Laboratory diagnostics of DC is difficult. Detection
of very short telomeres in lymphocytes by flow cy-
tometry and fluorescence insitu hybridization (FISH)
in commercial laboratories can differentiate DC from
other syndromes involving bone marrow failure. Ge-
netic screening of DC patients has also revealed het-
erozygous mutations in the TERC gene, homozygous
mutations in the NOP10 and NHP2 genes coding for
proteins associated with the telomerase complex, and
mutations in the TERT gene. The autosomal dominant
form of DC is characterized by mutations in the TINF2
gene. The loss of functionally active TIN2 in the shel-
terin complex leads to the cells with extremely short
telomeres [151]. Another cause of DC is thymidylate
synthase deficiency [152]. Mutations in the CTC1,
RTEL1, ACD, PARN, NAF1, ZCCHC8, NPM1, MDM4,
RPA1, DCLRE1B, POT1, and TYMS-ENOSF1 genes were
also found in DC and DC-like diseases. These muta-
tions disrupt the functioning of telomerase and/or
proteins involved in the telomere maintenance and
lead to the appearance of short telomeres [153, 154].
Genetic testing of a large cohort of clinically di-
agnosed patients with DC and DC-like diseases has
identified several new mutations in the known genet-
ic loci, X-linked POLA1 gene, and POT1 and ZCCHC8
genes. Functional characterization of the new POLA1
and POT1 variants revealed abnormal protein–protein
interactions between primase and CST and shelterin
complex proteins, which are critical for the TL main-
tenance [155]. Assessment of the TL allows to conduct
differential diagnosis between the acquired aplastic
anemia and DC [156] and control sickle cell anemia
[157] and some other diseases of the hematopoietic
system [158]. Modern genetic technologies make it
possible to conduct such studies.
TECHNOLOGIES FOR THE ASSESSMENT
OFTELOMERASE ACTIVITY
AND TELOMERE LENGTH IN CLINICAL
AND EPIDEMIOLOGICAL STUDIES
Analysis of published data on the TA-TL system
dysfunction in the pathogenesis of diseases, including
cancer and genetic disorders associated with telo-
meropathies, has demonstrated the relevance of TA
assessment in the identification of the malignant na-
ture of a tumor. The presence of short telomeres can
be an important disease (telomeropathy) marker, an
indicator of individual sensitivity to the damaging fac-
tors, and a prognostic marker for the development of
complications during chemo- and radiation therapy of
oncological diseases. The significance of disease diag-
nostics and monitoring has promoted the development
of methods for the assessment of TA and TL in clinical
and epidemiological studies.
The methods for measuring TA in cell and tissue
extracts were described in detail in the review [159].
The authors examined the sensitivity, advantages, dis-
advantages, and implementation of existing methods
for TA analysis, which they divided into two types:
direct assessment of telomerase-synthesized DNA and
methods using various signal amplification schemes to
increase the sensitivity. There are numerous ongoing
studies on the improvement of TA analysis methods
[87, 160, 161].
In terms of sensitivity, cost, complexity of imple-
mentation, and equipment availability, the most avail-
able method for the TA assessment in tumor tissue
in clinical trials is amplification of telomeric repeats
followed by their fluorescent detection, called TRAP
(telomeric repeat amplification protocol). The method
is based on the elongation of oligonucleotide substrate
by telomerase, PCR amplification of the synthesized
telomeric repeats, separation of the amplified TRAP
products by electrophoresis, their visualization by
staining with the SYBR Gold dye, and gel documen-
tation for subsequent processing of the results and
evaluation of the relative TA vs. positive control sam-
ple [162]. An optimized protocol for this method was
described in [163] that included a procedure for pre-
paring the lysate from surgical specimens, as well as
the protocol for the result analysis and quantification
suitable for the use in diagnostic studies.
TELOMERE LENGTH AS PROGNOSTIC MARKER 715
BIOCHEMISTRY (Moscow) Vol. 90 No. 6 2025
Given the above-mentioned difficulties associated
with the standardization of sample preparation, it is
possible that the TA assessment in biological materials
will be eventually replaced by the analysis of C228T
and C250T mutations in the hTERT gene promoter or
other mutations that lead to the telomerase activation
and malignant transformation [89]. A combination of
modern DNA sequencing technologies used for the
mutation identification and liquid or conventional bi-
opsy might be a more promising approach compared
to the routine TA assessment.
A wide range of methods have been developed
for the TL assessment, including analysis of terminal
restriction fragments (TRFs), quantitative PCR (qPCR),
STELA (single telomere length analysis), MMQPCR
(monochrome multiplex qPCR), Q-FISH (quantitative
FISH of telomeres in metaphase chromosomes), and
FlowFISH (determination of relative TL using flow cy-
tofluorimetry). Until recently, FlowFISH had been the
only method acceptable for clinical use, as it allows
highly reproducible, simple, and rapid measurement
of absolute TL [164]. The authors of [164] developed
an algorithm for converting the flow cytometry data
into the absolute TL expressed in kb, which makes
it possible to compare results obtained in different
laboratories, as well as to perform the retrospective
conversion of previously obtained flow cytometry
data. Flow-FISH and qPCR offer specific and highly
sensitive measurement of normal-length telomeres.
However, the sensitivity and specificity of qPCR in the
detection of short telomeres are lower [165], although
this method is suitable for TL measurement in epide-
miological studies. The STELA method has the high-
est sensitivity for analyzing short telomeres, based on
the fact that all telomeres have the GGG repeat at the
3′ end. This method has also been adapted for epide-
miological studies, but it requires special equipment
and reagents [166].
Hence, this brief review of methods used for TA
and TL analysis allows us to conclude that the current
array of molecular biological and genetic technologies
allows to conduct research on the TA and TL in clinical
and epidemiological studies, although these methods
and software for data processing continue to improve.
CONCLUSION
Analysis of published data on the telomere biol-
ogy and mechanisms of TL maintenance in human
cells demonstrate that these topics still remain in the
research focus. In recent years, new data have been
obtained on the important role of CST–Polα/Prim in
the TL maintenance. The primase forms a complex
with CST, which ensures the high specificity of repli-
cation initiation for the complementary C-strand after
completion of the G-chain synthesis by telomerase. In
the case of low TA or its absence, the TL is maintained
by the ALT mechanism based on homologous recom-
bination. The regulation of TA by different mecha-
nisms has been well studied, including the control of
TA at the epigenetic level. TERRA, a long non-coding
telomeric repeat-containing RNA transcribed by RNA
polymerase II from the intrachromosomal telomeric
repeats, likely plays an important role in telomerase
regulation and heterochromatin organization in the
telomere region.
TL is an important marker of cells well-being.
The state of telomeres (length, structure, presence of
oxidized guanine and/or G4s, quality of shelterin and
CST complex proteins) is currently considered as an
indicator of health. The accumulation of short telo-
meres indicates replicative aging of proliferating cells,
transition to the senescent state, and increased sen-
sitivity to damaging factors. Proteins of the shelterin
complex are crucial for the telomere protection. The
inactivation of shelterin binding to the telomere DNA
leads to the appearance of unprotected telomeres,
activation of different DDR pathways, and genomic
instability. The alternative functions of telomerase
associated with the anti-apoptotic activity of its com-
ponents after their translocation to the nucleus (TR)
or mitochondria (TERT) may be of great importance
in the maintenance of cell stability.
A high TA can serve as a marker of neoplasm
malignancy. Mutations in genes coding for proteins
involved in the maintenance of telomere structure
and length lead to the development of genetic dis-
eases (telomeropathies) that can be diagnosed by the
analysis of TL and TA.
IR damages telomeres via direct DNA damage and
through the action of ROS generated by the radiolysis
of intracellular water. A high guanine content makes
telomeres highly sensitive to ROS, while formation
of 8-OxodG in the telomere DNA inhibits telomerase.
Similar lesions were detected after the long-term γ-
and X-irradiation at low doses (mainly after a certain
period of time) or after the chronic low-dose irradia-
tion of occupationally exposed individuals.
According to the National Aeronautics and Space
Administration, changes in the TL in astronauts rep-
resent one of the important biological responses to
the effects of spaceflight factors. Both TL and TA are
among the parameters that should be analyzed in as-
tronauts before, during, and after completion of space
mission.
Telomeres are dynamic structures that undergo
significant changes under the influence of deleterious
factors. The TL is determined by the cumulative im-
pact of all endogenous and exogenous damaging fac-
tors acting throughout the lifetime, rather than by the
chronological age.
MOSKALEVA et al.716
BIOCHEMISTRY (Moscow) Vol. 90 No. 6 2025
The current state of biochemical and molecular
genetic methods has made it possible to develop tech-
nologies for the TA assay and identification of muta-
tions affecting TA that can be used for the diagnostics
of malignant neoplasms, as well as the methods for
TL assessment in order to monitor telomeropathies
and health status in clinical practice. The introduction
of these technologies into clinical practice and epide-
miological studies will significantly expand the pos-
sibilities of medical and biological diagnostic studies,
allow prediction of individual sensitivity of patients
to the planned radiation and chemotherapeutic treat-
ment, and facilitate the assessment of risks of devel-
oping unwanted late complications.
Abbreviations. 8-OxodG, 8-oxo-7,8-dihydro-2′-de-
oxyguanosine; ALT, alternative lengthening of telo-
meres; CST–Polα/Prim, CST–Polymerase α/primase;
DC, dyskeratosis congenita; DDR, DNA damage re-
sponse; DSB, DNA double-strand break; G4s, G-qua-
druplexes; TERT, telomerase reverse transcriptase; TR
(TERC), telomerase RNA component; IR, ionizing radi-
ation; MSC, mesenchymal stromal cell; ROS, reactive
oxygen species; TA, telomerase activity; TL, telomere
length.
Contributions. E.Yu.M. and A.I.G developed the
review concept and wrote the “Introduction” and
“Conclusion” sections; A.S.Zh., O.V.V., and S.A.V. re-
trieved the published information; E.Yu.M., A.I.G.,
and S.A.V. wrote the section “Telomere structure
and mechanisms of telomere maintenance”; E.Yu.M.,
A.S.Zh., and O.V.V. wrote the section “Telomere sensi-
tivity to ionizing radiation”; E.Yu.M. wrote the section
“Telomere length and telomerase in telomeropathies”;
A.I.G. and O.V.V wrote the section “Technologies for the
assessment of telomerase activity and telomere length
in clinical and epidemiological studies”. All authors re-
viewed and edited the manuscript.
Funding. The work was carried out within the
State Assignment for the National Research Center
“Kurchatov Institute”.
Ethics approval and consent to participate.
This work does not contain any studies involving hu-
man and animal subjects.
Conflict of interest. The authors of this work de-
clare that they have no conflicts of interest.
REFERENCES
1. Olovnikov, A. M. (1971) Principle of marginotomy in
template synthesis of polynucleotides [In Russian],
Dokl. Akad. Nauk SSSR, 201, 1496-1499.
2. Olovnikov, A.M. (1973) A theory of marginotomy. The
incomplete copying of template margin in enzymic
synthesis of polynucleotides and biological signifi-
cance of the phenomenon, J.Theor. Biol., 41, 181-190,
https://doi.org/10.1016/0022-5193(73)90198-7.
3. Hayflick,L. (1965) The limited invitro lifetime of hu-
man diploid cell strains, Exp. Cell Res., 37, 614-636,
https://doi.org/10.1016/0014-4827(65)90211-9.
4. Greider, C.W., and Blackburn, E.H. (1985) Identifica-
tion of a specific telomere terminal transferase activ-
ity in Tetrahymena extracts, Cell, 43, 405-413, https://
doi.org/10.1016/0092-8674(85)90170-9.
5. Greider, C.W., and Blackburn, E.H. (1989) A telomer-
ic sequence in the RNA of Tetrahymena telomerase
required for telomere repeat synthesis, Nature, 337,
331-337, https://doi.org/10.1038/337331a0.
6. Greider, C. W. (1998) Telomeres and senescence:
the history, the experiment, the future, Curr.
Biol., 8, R178-R181, https://doi.org/10.1016/s0960-
9822(98)70105-8.
7. Harley, C. B., Futcher, A. B., and Greider, C. W. (1990)
Telomeres shorten during ageing of human fibro-
blasts, Nature, 345, 458-460, https://doi.org/10.1038/
345458a0.
8. Hastie, N.D., Dempster,M., Dunlop, M.G., Thompson,
A. M., Green, D. K., and Allshire, R. C. (1990) Telo-
mere reduction in human colorectal carcinoma
and with ageing, Nature, 346, 866-868, https://
doi.org/10.1038/346866a0.
9. Editorial (2023) [In Russian] Biokhimiya, 88, 2037,
https://doi.org/10.31857/S0320972523110015.
10. Olovnikov, I. A. (2024) Telomeres in health and lon-
gevity: special issue in memory of Alexey Olovnikov,
Biogerontology, 25, 191-193, https://doi.org/10.1007/
s10522-023-10090-7.
11. Griffith, J. D., Comeau, L., Rosenfield, S., Stansel,
R. M., Bianchi, A., Moss, H., and de Lange, T. (1999)
Mammalian telomeres end in a large duplex loop,
Cell, 97, 503-514, https://doi.org/10.1016/s0092-8674
(00)80760-6.
12. Doksani, Y., and de Lange, T. (2014) The role of dou-
ble-strand break repair pathways at functional and
dysfunctional telomeres, Cold Spring Harb. Perspect.
Biol., 6, a016576, https://doi.org/10.1101/cshperspect.
a016576.
13. Ruis, P., and Boulton, S. J. (2021) The end protection
problem-an unexpected twist in the tail, Genes Dev.,
35, 1-21, https://doi.org/10.1101/gad.344044.120.
14. Yang, D. (2019) G-Quadruplex DNA and RNA, Meth-
ods Mol. Biol., 2035, 1-24, https://doi.org/10.1007/978-
1-4939-9666-7_1.
15. De Lange,T. (2005) Shelterin: the protein complex that
shapes and safeguards human telomeres, Genes Dev.,
19, 2100-2110, https://doi.org/10.1101/gad.1346005.
16. O’Connor, M. S., Safari, A., Xin, H., Liu, D., and
Songyang,Z. (2006) A critical role for TPP1 and TIN2
interaction in high-order telomeric complex assembly,
Proc. Natl. Acad. Sci. USA, 103, 11874-11879, https://
doi.org/10.1073/pnas.0605303103.
TELOMERE LENGTH AS PROGNOSTIC MARKER 717
BIOCHEMISTRY (Moscow) Vol. 90 No. 6 2025
17. De Lange, T. (2018) Shelterin-mediated telomere
protection, Annu. Rev. Genet., 52, 223-247, https://
doi.org/10.1146/annurev-genet-032918-021921.
18. Lim, C.J., and Cech, T.R. (2021) Shaping human telo-
meres: from shelterin and CST complexes to telomeric
chromatin organization, Nat. Rev. Mol. Cell Biol., 22,
283-298, https://doi.org/10.1038/s41580-021-00328-y.
19. Smith, E. M., Pendlebury, D. F., and Nandakumar, J.
(2020) Structural biology of telomeres and telomerase,
Cell. Mol. Life Sci., 77, 61-79, https://doi.org/10.1007/
s00018-019-03369-x.
20. Palm, W., and de Lange, T. (2008) How shelterin
protects mammalian telomeres, Annu. Rev. Genet.,
42, 301-334, https://doi.org/10.1146/annurev.genet.
41.110306.130350.
21. Wang, F., and Lei, M. (2011) Human telomere POT1-
TPP1 complex and its role in telomerase activity
regulation, Methods Mol. Biol., 735, 173-187, https://
doi.org/10.1007/978-1-61779-092-8_17.
22. Wang, F., Podell, E. R., Zaug, A. J., Yang, Y., Baciu, P.,
Cech, T.R., and Lei,M. (2007) The POT1-TPP1 telomere
complex is a telomerase processivity factor, Nature,
445, 506-510, https://doi.org/10.1038/nature05454.
23. Doksani, Y. (2019) The response to DNA damage at
telomeric repeats and its consequences for telomere
function, Genes (Basel), 10, 318, https://doi.org/10.3390/
genes10040318.
24. Yegorov, Y. E. (2023) Olovnikov, telomeres, and
telomerase. Is it possible to prolong a healthy life?
Biochemistry (Moscow), 88, 1704-1718, https://
doi.org/10.1134/S0006297923110032.
25. Martens, U.M., Chavez, E.A., Poon, S.S., Schmoor,C.,
and Lansdorp, P. M. (2000) Accumulation of short
telomeres in human fibroblasts prior to replica-
tive senescence, Exp. Cell Res., 256, 291-299, https://
doi.org/10.1006/excr.2000.4823.
26. Gaspar, T.B., Sa,A., Lopes, J.M., Sobrinho-Simoes,M.,
Soares, P., and Vinagre, J. (2018) Telomere mainte-
nance mechanisms in cancer, Genes, 9, 241, https://
doi.org/10.3390/genes9050241.
27. Campisi, J., and d’Adda di Fagagna, F. (2007) Cellu-
lar senescence: when bad things happen to good
cells, Nat. Rev. Mol. Cell Biol., 8, 729-740, https://
doi.org/10.1038/nrm2233.
28. Fumagalli, M., Rossiello, F., Clerici, M., Barozzi, S.,
Cittaro, D., Kaplunov, J. M., Bucci, G., Dobreva, M.,
Matti, V., Beausejour, C. M., Herbig, U., Longhese,
M. P., and d’Adda di Fagagna, F. (2012) Telomeric
DNA damage is irreparable and causes persistent
DNA-damage-response activation, Nat. Cell Biol., 14,
355-365, https://doi.org/10.1038/ncb2466.
29. Slawinska,N., and Krupa,R. (2021) Molecular aspects
of senescence and organismal ageing-DNA damage
response, telomeres, inflammation and chroma-
tin, Int.J. Mol. Sci., 22, 590, https://doi.org/10.3390/
ijms22020590.
30. GTEx Consortium (2020) The GTEx Consortium atlas
of genetic regulatory effects across human tissues,
Science, 369, 1318-1330, https://doi.org/10.1126/sci-
ence.aaz1776.
31. Demanelis, K., Jasmine, F., Chen, L. S., Chernoff, M.,
Tong, L., Delgado, D., Zhang, C., Shinkle, J., Sabar-
inathan, M., Lin, H., Ramirez, E., Oliva, M., Kim-
Hellmuth,S., Stranger, B.E., Lai, T.P., Aviv,A., Ardlie,
K.G., Aguet,F., Ahsan,H., GTEx Consortium, Doherty,
J. A., Kibriyaand, M. G., and Pierce, B. L. (2020) De-
terminants of telomere length across human tissues,
Science, 369, eaaz6876, https://doi.org/10.1126/science.
aaz6876.
32. Carver, A. J., Hing, B., Elser, B. A., Lussier, S. J.,
Yamanashi, T., Howard, M. A., 3rd, Kawasaki, H.,
Shinozaki, G., and Stevens, H. E. (2024) Correlation
of telomere length in brain tissue with peripheral
tissues in living human subjects, Front. Mol. Neu-
rosci., 17, 1303974, https://doi.org/10.3389/fnmol.
2024.1303974.
33. Finnicum, C. T., Dolan, C. V., Willemsen, G., Weber,
Z. M., Petersen, J. L., Beck, J. J., Codd, V., Boomsma,
D.I., Davies, G.E., and Ehli, E.A. (2017) Relative telo-
mere repeat mass in buccal and leukocyte-derived
DNA, PLoS One, 12, e0170765, https://doi.org/10.1371/
journal.pone.0170765.
34. Frenck, R. W., Jr., Blackburn, E. H., and Shannon,
K. M. (1998) The rate of telomere sequence loss in
human leukocytes varies with age, Proc. Natl. Acad.
Sci. USA, 95, 5607-5610, https://doi.org/10.1073/pnas.
95.10.5607.
35. Kagirova, Z. R., Demina, I. A., Blokhin, B. M., and
Rumyantsev, A. G. (2017) Telomere length and chil-
dren’s health [in Russian], Pediatric Hematol. Oncol.
Immunopathol., 16, 107-112, https://doi.org/10.24287/
1726-1708-2017-16-4-107-112.
36. Takai, H., Aria, V., Borges, P., Yeeles, J. T. P., and de
Lange, T. (2024) CST-polymerase alpha-primase
solves a second telomere end-replication problem,
Nature, 627, 664-670, https://doi.org/10.1038/s41586-
024-07137-1.
37. Zaug, A. J., Goodrich, K. J., Song, J. J., Sullivan, A. E.,
and Cech, T. R. (2022) Reconstitution of a telomer-
ic replicon organized by CST, Nature, 608, 819-825,
https://doi.org/10.1038/s41586-022-04930-8.
38. Cesare, A. J., and Reddel, R. R. (2010) Alternative
lengthening of telomeres: models, mechanisms and
implications, Nat. Rev. Genet., 11, 319-330, https://
doi.org/10.1038/nrg2763.
39. Draskovic, I., and Londono Vallejo, A. (2013) Telo-
mere recombination and alternative telomere length-
ening mechanisms, Front. Biosci., 18, 1-20, https://
doi.org/10.2741/4084.
40. Hu, Y., Shi, G., Zhang, L., Li, F., Jiang, Y., Jiang, S.,
Ma, W., Zhao, Y., Songyang, Z., and Huang, J. (2016)
Switch telomerase to ALT mechanism by inducing
MOSKALEVA et al.718
BIOCHEMISTRY (Moscow) Vol. 90 No. 6 2025
telomeric DNA damages and dysfunction of ATRX
and DAXX, Sci. Rep., 6, 32280, https://doi.org/10.1038/
srep32280.
41. Feng, J., Funk, W. D., Wang, S. S., Weinrich, S. L.,
Avilion, A. A., Chiu, C. P., Adams, R. R., Chang, E.,
Allsopp, R. C., Yu, J., Le, S., West, M. D., Harley, C. B.,
Andrews, W. H., Greider, C. W., and Villeponteau, B.
(1995) The RNA component of human telomer-
ase, Science, 269, 1236-1241, https://doi.org/10.1126/
science.7544491.
42. Roake, C. M., and Artandi, S. E. (2020) Regulation of
human telomerase in homeostasis and disease, Nat.
Rev. Mol. Cell Biol., 21, 384-397, https://doi.org/10.1038/
s41580-020-0234-z.
43. Rubtsova, M. P., Vasilkova, D. P., Malyavko, A. N.,
Naraikina, Y. V., Zvereva, M. I., and Dontsova,
O. A. (2012) Telomere lengthening and other func-
tions of telomerase, Acta Naturae, 4, 44-61, https://
doi.org/10.32607/20758251-2012-4-2-44-61.
44. Wang, F., Stewart, J. A., Kasbek, C., Zhao, Y., Wright,
W. E., and Price, C. M. (2012) Human CST has inde-
pendent functions during telomere duplex replication
and C-strand fill-in, Cell Rep., 2, 1096-1103, https://
doi.org/10.1016/j.celrep.2012.10.007.
45. He, Q., and Lim, C. J. (2023) Models for human telo-
mere C-strand fill-in by CST-Polalpha-primase, Trends
Biochem. Sci., 48, 860-872, https://doi.org/10.1016/
j.tibs.2023.07.008.
46. Cai, S. W., and de Lange, T. (2023) CST-Polalpha/
Primase: the second telomere maintenance ma-
chine, Genes Dev, 37, 555-569, https://doi.org/10.1101/
gad.350479.123.
47. Zaug, A. J., Lim, C. J., Olson, C. L., Carilli, M. T.,
Goodrich, K. J., Wuttke, D. S., and Cech, T. R. (2021)
CST does not evict elongating telomerase but prevents
initiation by ssDNA binding, Nucleic Acids Res., 49,
11653-11665, https://doi.org/10.1093/nar/gkab942.
48. Beattie, T. L., Zhou, W., Robinson, M. O., and
Harrington, L. (2001) Functional multimerization of
the human telomerase reverse transcriptase, Mol.
Cell. Biol., 21, 6151-6160, https://doi.org/10.1128/
MCB.21.18.6151-6160.2001.
49. Harrington, L., McPhail, T., Mar, V., Zhou, W.,
Oulton, R., Bass, M. B., Arruda, I., and Robinson,
M. O. (1997) A mammalian telomerase-associated
protein, Science, 275, 973-977, https://doi.org/10.1126/
science.275.5302.973.
50. Cong, Y. S., Wright, W. E., and Shay, J. W. (2002)
Human telomerase and its regulation, Microbiol.
Mol. Biol. Rev., 66, 407-425, https://doi.org/10.1128/
MMBR.66.3.407-425.2002.
51. Wick, M., Zubov, D., and Hagen, G. (1999) Genomic
organization and promoter characterization of the
gene encoding the human telomerase reverse tran-
scriptase (hTERT), Gene, 232, 97-106, https://doi.org/
10.1016/s0378-1119(99)00108-0.
52. Skvortzov, D. A., Rubzova, M. P., Zvereva, M. E.,
Kiselev, F.L., and Donzova, O.A. (2009) The regulation
of telomerase in oncogenesis, Acta Naturae, 1, 51-67,
https://doi.org/10.32607/20758251-2009-1-1-51-67.
53. Nalobin, D.S., Galiakberova, A.A., Alipkina, S.I., and
Glukhov, A. I. (2018) Regulation of telomerase activi-
ty, Biol. Bull. Rev., 8, 142-154, https://doi.org/10.1134/
S2079086418020068.
54. Teichroeb, J. H., Kim, J., and Betts, D. H. (2016) The
role of telomeres and telomerase reverse transcrip-
tase isoforms in pluripotency induction and mainte-
nance, RNA Biol., 13, 707-719, https://doi.org/10.1080/
15476286.2015.1134413.
55. Surovtseva, Y. V., Churikov, D., Boltz, K. A., Song, X.,
Lamb, J. C., Warrington, R., Leehy, K., Heacock, M.,
Price, C.M., and Shippen, D.E. (2009) Conserved telo-
mere maintenance component 1 interacts with STN1
and maintains chromosome ends in higher eukary-
otes, Mol. Cell, 36, 207-218, https://doi.org/10.1016/
j.molcel.2009.09.017.
56. Chen, L. Y., Redon, S., and Lingner, J. (2012) The hu-
man CST complex is a terminator of telomerase ac-
tivity, Nature, 488, 540-544, https://doi.org/10.1038/
nature11269.
57. Feng,X., Hsu, S.J., Kasbek,C., Chaiken,M., and Price,
C. M. (2017) CTC1-mediated C-strand fill-in is an es-
sential step in telomere length maintenance, Nucle-
ic Acids Res., 45, 4281-4293, https://doi.org/10.1093/
nar/gkx125.
58. Azzalin, C. M., Reichenbach, P., Khoriauli, L.,
Giulotto, E., and Lingner, J. (2007) Telomeric repeat
containing RNA and RNA surveillance factors at
mammalian chromosome ends, Science, 318, 798-801,
https://doi.org/10.1126/science.1147182.
59. Lalonde,M., and Chartrand,P. (2020) TERRA, a multi-
faceted regulator of telomerase activity at telomeres,
J. Mol. Biol., 432, 4232-4243, https://doi.org/10.1016/
j.jmb.2020.02.004.
60. Cusanelli, E., and Chartrand, P. (2015) Telomeric
repeat-containing RNA TERRA: a noncoding RNA
connecting telomere biology to genome integrity,
Front. Genet., 6, 143, https://doi.org/10.3389/fgene.
2015.00143.
61. Redon,S., Reichenbach, P., and Lingner,J. (2010) The
non-coding RNA TERRA is a natural ligand and direct
inhibitor of human telomerase, Nucleic Acids Res., 38,
5797-5806, https://doi.org/10.1093/nar/gkq296.
62. Venteicher, A.S., Abreu, E.B., Meng,Z., McCann, K.E.,
Terns, R.M., Veenstra, T.D., Terns, M.P., and Artandi,
S.E. (2009) A human telomerase holoenzyme protein
required for Cajal body localization and telomere syn-
thesis, Science, 323, 644-648, https://doi.org/10.1126/
science.1165357.
63. Deng, Z., Norseen, J., Wiedmer, A., Riethman, H.,
and Lieberman, P. M. (2009) TERRA RNA binding
to TRF2 facilitates heterochromatin formation and
TELOMERE LENGTH AS PROGNOSTIC MARKER 719
BIOCHEMISTRY (Moscow) Vol. 90 No. 6 2025
ORC recruitment at telomeres, Mol. Cell, 35, 403-413,
https://doi.org/10.1016/j.molcel.2009.06.025.
64. Xu, Y., and Komiyama, M. (2023) G-Quadruplexes in
Human Telomere: Structures, Properties, and Appli-
cations, Molecules, 29, 174, https://doi.org/10.3390/
molecules29010174.
65. Wang, H., Nora, G. J., Ghodke, H., and Opresko, P. L.
(2011) Single molecule studies of physiologically rele-
vant telomeric tails reveal POT1 mechanism for pro-
moting G-quadruplex unfolding, J.Biol. Chem., 286,
7479-7489, https://doi.org/10.1074/jbc.M110.205641.
66. Cuesta, J., Read, M. A., and Neidle, S. (2003) The
design of G-quadruplex ligands as telomerase in-
hibitors, Mini Rev. Med. Chem., 3, 11-21, https://
doi.org/10.2174/1389557033405502.
67. Sadhukhan,R., Chowdhury,P., Ghosh,S., and Ghosh,U.
(2018) Expression of telomere-associated proteins is
interdependent to stabilize native telomere struc-
ture and telomere dysfunction by G-quadruplex
ligand causes TERRA upregulation, Cell Biochem.
Biophys., 76, 311-319, https://doi.org/10.1007/s12013-
017-0835-0.
68. Fernandes, R.V., Feretzaki, M., and Lingner, J. (2021)
The makings of TERRA R-loops at chromosome ends,
Cell Cycle, 20, 1745-1759, https://doi.org/10.1080/15384
101.2021.1962638.
69. Graf,M., Bonetti,D., Lockhart,A., Serhal,K., Kellner,V.,
Maicher, A., Jolivet, P., Teixeira, M. T., and Luke, B.
(2017) Telomere length determines TERRA and R-loop
regulation through the cell cycle, Cell, 170, 72-85,
https://doi.org/10.1016/j.cell.2017.06.006.
70. Brickner, J.R., Garzon, J.L., and Cimprich, K.A. (2022)
Walking a tightrope: the complex balancing act of
R-loops in genome stability, Mol. Cell, 82, 2267-2297,
https://doi.org/10.1016/j.molcel.2022.04.014.
71. Arora,R., and Azzalin, C.M. (2015) Telomere elonga-
tion chooses TERRA ALTernatives, RNA Biol., 12, 938-
941, https://doi.org/10.1080/15476286.2015.1065374.
72. Al-Turki, T. M., and Griffith, J. D. (2023) Mammalian
telomeric RNA (TERRA) can be translated to produce
valine-arginine and glycine-leucine dipeptide repeat
proteins, Proc. Natl. Acad. Sci. USA, 120, e2221529120,
https://doi.org/10.1073/pnas.2221529120.
73. Rosen, J., Jakobs, P., Ale-Agha, N., Altschmied, J.,
and Haendeler, J. (2020) Non-canonical functions
of telomerase reverse transcriptase – impact on re-
dox homeostasis, Redox Biol., 34, 101543, https://
doi.org/10.1016/j.redox.2020.101543.
74. Segal-Bendirdjian, E., and Geli, V. (2019) Non-canon-
ical roles of telomerase: unraveling the Imbroglio,
Front. Cell Dev. Biol., 7, 332, https://doi.org/10.3389/
fcell.2019.00332.
75. Marinaccio, J., Micheli, E., Udroiu, I., Di Nottia, M.,
Carrozzo, R., Baranzini, N., Grimaldi, A., Leone, S.,
Moreno, S., Muzzi, M., and Sgura, A. (2023) TERT
extra-telomeric roles: antioxidant activity and mito-
chondrial protection, Int.J. Mol. Sci., 24, 4450, https://
doi.org/10.3390/ijms24054450.
76. Gazzaniga, F.S., and Blackburn, E.H. (2014) An anti-
apoptotic role for telomerase RNA in human immune
cells independent of telomere integrity or telomerase
enzymatic activity, Blood, 124, 3675-3684, https://
doi.org/10.1182/blood-2014-06-582254.
77. Rubtsova, M., and Dontsova, O. (2020) Human telo-
merase RNA: telomerase component or more? Biomol-
ecules, 10, 873, https://doi.org/10.3390/biom10060873.
78. Rubtsova, M., and Dontsova, O. (2022) How structur-
al features define biogenesis and function of human
telomerase RNA primary transcript, Biomedicines, 10,
1650, https://doi.org/10.3390/biomedicines10071650.
79. Rubtsova,M., Naraykina,Y., Vasilkova,D., Meerson,M.,
Zvereva, M., Prassolov, V., Lazarev, V., Manuvera, V.,
Kovalchuk,S., Anikanov,N., Butenko,I., Pobeguts,O.,
Govorun, V., and Dontsova, O. (2018) Protein encod-
ed in human telomerase RNA is involved in cell pro-
tective pathways, Nucleic Acids Res., 46, 8966-8977,
https://doi.org/10.1093/nar/gky705.
80. Artandi, S. E., and DePinho, R. A. (2010) Telomeres
and telomerase in cancer, Carcinogenesis, 31, 9-18,
https://doi.org/10.1093/carcin/bgp268.
81. Durant, S. T. (2012) Telomerase-independent paths to
immortality in predictable cancer subtypes, J.Cancer,
3, 67-82, https://doi.org/10.7150/jca.3965.
82. Tsatsakis, A., Oikonomopoulou, T., Nikolouzakis,
T. K., Vakonaki, E., Tzatzarakis, M., Flamourakis, M.,
Renieri,E., Fragkiadaki,P., Iliaki,E., Bachlitzanaki,M.,
Karzi,V., Katsikantami,I., Kakridonis,F., Hatzidaki,E.,
Tolia, M., Svistunov, A. A., Spandidos, D. A., Nikito-
vic, D., Tsiaoussis, J., and Berdiaki, A. (2023) Role
of telomere length in human carcinogenesis (re-
view), Int. J. Oncol., 63, 78, https://doi.org/10.3892/ijo.
2023.5526.
83. Fang,T., Zhang,Z., Ren,K., and Zou,L. (2024) Geneti-
cally determined telomere length as a risk factor for
hematological malignancies: evidence from Mende-
lian randomization analysis, Aging (Albany NY), 16,
4684-4698, https://doi.org/10.18632/aging.205625.
84. Fernandes, S. G., Dsouza, R., Pandya, G., Kirtonia, A.,
Tergaonkar, V., Lee, S. Y., Garg, M., and Khattar, E.
(2020) Role of telomeres and telomeric proteins in
human malignancies and their therapeutic poten-
tial, Cancers (Basel), 12, 1901, https://doi.org/10.3390/
cancers12071901.
85. Glybochko, P. V., Alyaev, J. G., Potoldykova, N. V.,
Polyakovsky, K. A., Vinarov, A. Z., Glukhov, A. I., and
Gordeev, S. A. (2016) The role of telomerase activi-
ty in non-invasive diagnostics of bladder cancer [in
Russian], Urologiia, 4, 76-81.
86. Glukhov, A. I., Polotdykova, N. V., Gordeev, S. A.,
Vinarov, A. Z., Polyakovsky, K. A., Rapoport, L. M.,
Tsarichenko, D. G., Enikeev, D. V., and Glybochko,
P. V. (2018) Current trends in bladder cancer
MOSKALEVA et al.720
BIOCHEMISTRY (Moscow) Vol. 90 No. 6 2025
diagnosis [in Russian], Urologiia, 5, 100-105, https://
doi.org/10.18565/urology.2018.5.100-105.
87. Glukhov,A., Potoldykova,N., Taratkin,M., Gordeev,S.,
Polyakovsky, K., Laukhtina, E., Moschini, M.,
Abufaraj,M., Shariat, S.F., Sekacheva,M., Enikeev,D.,
and Glybochko,P. (2021) Detection of urothelial blad-
der cancer based on urine and tissue telomerase
activity measured by novel RT-TRAP-2PCR meth-
od, J.Clin. Med., 10, 1055, https://doi.org/10.3390/
jcm10051055.
88. Casadio, V., and Bravaccini, S. (2021) Telomerase
activity analysis in urine sediment for bladder
cancer, Methods Mol. Biol., 2292, 133-141, https://
doi.org/10.1007/978-1-0716-1354-2_12.
89. Powter, B., Jeffreys, S. A., Sareen, H., Cooper, A.,
Brungs, D., Po, J., Roberts, T., Koh, E. S., Scott, K. F.,
Sajinovic, M., Vessey, J. Y., de Souza, P., and Becker,
T. M. (2021) Human TERT promoter mutations as a
prognostic biomarker in glioma, J.Cancer Res. Clin.
Oncol., 147, 1007-1017, https://doi.org/10.1007/s00432-
021-03536-3.
90. Shou,S., Maolan,A., Zhang,D., Jiang,X., Liu,F., Li,Y.,
Zhang,X., Geer,E., Pu,Z., Hua,B., Guo, Q., Zhang,X.,
and Pang, B. (2025) Telomeres, telomerase, and can-
cer: mechanisms, biomarkers, and therapeutics, Exp.
Hematol. Oncol., 14, 8, https://doi.org/10.1186/s40164-
025-00597-9.
91. Penninckx, S., Pariset, E., Cekanaviciute, E., and
Costes, S. V. (2021) Quantification of radiation-in-
duced DNA double strand break repair foci to evalu-
ate and predict biological responses to ionizing radi-
ation, NAR Cancer, 3, zcab046, https://doi.org/10.1093/
narcan/zcab046.
92. Nickoloff, J. A., Sharma, N., Allen, C. P., Taylor, L.,
Allen, S.J., Jaiswal, A.S., and Hromas,R. (2023) Roles
of homologous recombination in response to ioniz-
ing radiation-induced DNA damage, Int. J. Radiat.
Biol., 99, 903-914, https://doi.org/10.1080/09553002.
2021.1956001.
93. Mladenov, E., Mladenova, V., Stuschke, M., and
Iliakis, G. (2023) New facets of DNA double strand
break repair: radiation dose as key determinant of
HR versus c-NHEJ Engagement, Int.J. Mol. Sci., 24,
14956, https://doi.org/10.3390/ijms241914956.
94. Iliakis,G., Murmann,T., and Soni,A. (2015) Alternative
end-joining repair pathways are the ultimate backup
for abrogated classical non-homologous end-joining
and homologous recombination repair: implications
for the formation of chromosome translocations, Mu-
tat. Res. Genet. Toxicol. Environ. Mutagen., 793, 166-
175, https://doi.org/10.1016/j.mrgentox.2015.07.001.
95. Rogakou, E. P., Pilch, D. R., Orr, A. H., Ivanova, V. S.,
and Bonner, W.M. (1998) DNA double-stranded breaks
induce histone H2AX phosphorylation on serine 139,
J.Biol. Chem., 273, 5858-5868, https://doi.org/10.1074/
jbc.273.10.5858.
96. Karlseder, J., Hoke, K., Mirzoeva, O. K., Bakkenist, C.,
Kastan, M. B., Petrini, J. H., and de Lange, T. (2004)
The telomeric protein TRF2 binds the ATM kinase
and can inhibit the ATM-dependent DNA damage
response, PLoS Biol., 2, e240, https://doi.org/10.1371/
journal.pbio.0020240.
97. Imran, S.A.M., Yazid, M.D., Cui,W., and Lokanathan,Y.
(2021) The intra- and extra-telomeric role of TRF2 in
the DNA damage response, Int.J. Mol. Sci., 22, 9900,
https://doi.org/10.3390/ijms22189900.
98. Denchi, E. L., and de Lange, T. (2007) Protection of
telomeres through independent control of ATM and
ATR by TRF2 and POT1, Nature, 448, 1068-1071,
https://doi.org/10.1038/nature06065.
99. Hewitt, G., Jurk, D., Marques, F. D., Correia-Melo, C.,
Hardy, T., Gackowska, A., Anderson, R., Taschuk, M.,
Mann, J., and Passos, J. F. (2012) Telomeres are fa-
voured targets of a persistent DNA damage response
in ageing and stress-induced senescence, Nat. Com-
mun., 3, 708, https://doi.org/10.1038/ncomms1708.
100. Thapar, R. (2018) Regulation of DNA double-strand
break repair by non-coding RNAs, Molecules, 23,
2789, https://doi.org/10.3390/molecules23112789.
101. Porro, A., Feuerhahn, S., and Lingner, J. (2014)
TERRA-reinforced association of LSD1 with MRE11
promotes processing of uncapped telomeres, Cell
Rep., 6, 765-776, https://doi.org/10.1016/j.celrep.
2014.01.022.
102. Shim,G., Ricoul,M., Hempel, W.M., Azzam, E.I., and
Sabatier, L. (2014) Crosstalk between telomere main-
tenance and radiation effects: a key player in the
process of radiation-induced carcinogenesis, Mutat.
Res. Rev. Mutat. Res., 760, 1-17, https://doi.org/10.1016/
j.mrrev.2014.01.001.
103. Azzam, E.I., Jay-Gerin, J.P., and Pain,D. (2012) Ioniz-
ing radiation-induced metabolic oxidative stress and
prolonged cell injury, Cancer Lett., 327, 48-60, https://
doi.org/10.1016/j.canlet.2011.12.012.
104. Kumar, K., Fornace, A. J., Jr., and Suman, S. (2024)
8-OxodG: a potential biomarker for chronic oxidative
stress induced by high-LET radiation, DNA (Basel), 4,
221-238, https://doi.org/10.3390/dna4030015.
105. Santos, R. X., Correia, S. C., Zhu, X., Smith, M. A.,
Moreira, P. I., Castellani, R. J., Nunomura, A., and
Perry,G. (2013) Mitochondrial DNA oxidative damage
and repair in aging and Alzheimers disease, Antioxid
Redox Signal., 18, 2444-2457, https://doi.org/10.1089/
ars.2012.5039.
106. Shokolenko, I. N., Wilson, G. L., and Alexeyev, M. F.
(2014) Aging: a mitochondrial DNA perspective, crit-
ical analysis and an update, World J. Exp. Med., 4,
46-57, https://doi.org/10.5493/wjem.v4.i4.46.
107. Richter,C., Park, J.W., and Ames, B.N. (1988) Normal
oxidative damage to mitochondrial and nuclear DNA
is extensive, Proc. Natl. Acad. Sci. USA, 85, 6465-6467,
https://doi.org/10.1073/pnas.85.17.6465.
TELOMERE LENGTH AS PROGNOSTIC MARKER 721
BIOCHEMISTRY (Moscow) Vol. 90 No. 6 2025
108. Suman, S., Rodriguez, O. C., Winters, T. A., Fornace,
A. J., Jr., Albanese, C., and Datta, K. (2013) Therapeu-
tic and space radiation exposure of mouse brain
causes impaired DNA repair response and prema-
ture senescence by chronic oxidant production, Ag-
ing (Albany NY), 5, 607-622, https://doi.org/10.18632/
aging.100587.
109. Tseng, B. P., Giedzinski, E., Izadi, A., Suarez, T., Lan,
M. L., Tran, K. K., Acharya, M. M., Nelson, G. A.,
Raber, J., Parihar, V.K., and Limoli, C.L. (2014) Func-
tional consequences of radiation-induced oxidative
stress in cultured neural stem cells and the brain
exposed to charged particle irradiation, Antioxid
Redox Signal., 20, 1410-1422, https://doi.org/10.1089/
ars.2012.5134.
110. Barnes, R. P., de Rosa, M., Thosar, S. A., Detwiler,
A. C., Roginskaya, V., Van Houten, B., Bruchez, M. P.,
Stewart-Ornstein, J., and Opresko, P. L. (2022) Telo-
meric 8-oxo-guanine drives rapid premature senes-
cence in the absence of telomere shortening, Nat.
Struct. Mol. Biol., 29, 639-652, https://doi.org/10.1038/
s41594-022-00790-y.
111. Fouquerel, E., Barnes, R. P., Uttam, S., Watkins, S. C.,
Bruchez, M.P., and Opresko, P.L. (2019) Targeted and
persistent 8-oxoguanine base damage at telomeres
promotes telomere Loss and crisis, Mol. Cell., 75, 117-
130.e116, https://doi.org/10.1016/j.molcel.2019.04.024.
112. Wang,L., Lu,Z., Zhao,J., Schank,M., Cao,D., Dang,X.,
Nguyen, L. N., Nguyen, L. N. T., Khanal, S., Zhang, J.,
Wu, X.Y., El Gazzar,M., Ning,S., Moorman, J.P., and
Yao, Z.Q. (2021) Selective oxidative stress induces dual
damage to telomeres and mitochondria in human T
cells, Aging Cell, 20, e13513, https://doi.org/10.1111/
acel.13513.
113. Honig, L. S., Kang, M. S., Cheng, R., Eckfeldt, J. H.,
Thyagarajan,B., Leiendecker-Foster,C., Province, M.A.,
Sanders, J. L., Perls, T., Christensen, K., Lee, J. H.,
Mayeux,R., and Schupf,N. (2015) Heritability of telo-
mere length in a study of long-lived families, Neu-
robiol. Aging, 36, 2785-2790, https://doi.org/10.1016/
j.neurobiolaging.2015.06.017.
114. Krivoshchapova, Y. V., and Vozilova, A. V. (2022) The
study of the telomere length of the chromosomes in
T-lymphocytes of the exposed individuals [in Rus-
sian], J.Radiat. Safety Issues, 107, 71-79.
115. Smirnova, T. Y., Runov, A. L., Vonsky, M. S., Spivak,
D.L., Zakharchuk, A.G., Mikhelson, V.M., and Spivak,
I. M. (2012) Telomere length in a population of long-
lived people of the northwestern region of Russia,
Cell Tissue Biol., 6, 465-471, https://doi.org/10.1134/
S1990519X12050112.
116. Li, P., Hou, M., Lou, F., Bjorkholm, M., and Xu, D.
(2012) Telomere dysfunction induced by chemo-
therapeutic agents and radiation in normal human
cells, Int.J. Biochem. Cell Biol., 44, 1531-1540, https://
doi.org/10.1016/j.biocel.2012.06.020.
117. Luxton, J. J., McKenna, M. J., Lewis, A. M., Taylor,
L. E., Jhavar, S. G., Swanson, G. P., and Bailey, S. M.
(2021) Telomere length dynamics and chromosomal
instability for predicting individual radiosensitivi-
ty and risk via machine learning, J.Personal. Med.,
11, 188, https://doi.org/10.3390/jpm11030188.
118. Sishc, B. J., Nelson, C. B., McKenna, M. J., Battaglia,
C. L., Herndon, A., Idate, R., Liber, H. L., and Bailey,
S. M. (2015) Telomeres and telomerase in the radia-
tion response: implications for instability, reprogram-
ing, and carcinogenesis, Front. Oncol., 5, 257, https://
doi.org/10.3389/fonc.2015.00257.
119. Millet,P., Granotier,C., Etienne,O., and Boussin, F.D.
(2013) Radiation-induced upregulation of telomerase
activity escapes PI3-kinase inhibition in two ma-
lignant glioma cell lines, Int. J. Oncol., 43, 375-382,
https://doi.org/10.3892/ijo.2013.1970.
120. Wang, X., Liu, Y., Chow, L.S., Wong, S. C., Tsao, G. S.,
Kwong, D. L., Sham, J. S., and Nicholls, J. M. (2000)
Regulation of telomerase activity by gamma-radiation
in nasopharyngeal carcinoma cells, Anticancer Res.,
20, 433-437.
121. Hyeon Joo, O., Hande, M. P., Lansdorp, P. M., and
Natarajan, A.T. (1998) Induction of telomerase activ-
ity and chromosome aberrations in human tumour
cell lines following X-irradiation, Mutat. Res., 401, 121-
131, https://doi.org/10.1016/s0027-5107(97)00321-7.
122. Ram, R., Uziel, O., Eldan, O., Fenig, E., Beery, E.,
Lichtenberg,S., Nordenberg,Y., and Lahav, M. (2009)
Ionizing radiation up-regulates telomerase activity
in cancer cell lines by post-translational mechanism
via ras/phosphatidylinositol 3-kinase/Akt pathway,
Clin. Cancer Res., 15, 914-923, https://doi.org/10.1158/
1078-0432.CCR-08-0792.
123. Vysotskaya, O. V., Glukhov, A. I., Semochkina, Y. P.,
Gordeev, S. A., and Moskaleva, E. Y. (2020) Telomer-
ase activity, mTert gene expression and the telomere
length in mouse mesenchymal stem cells in the late
period after γ- and γ,n-irradiation and in tumors de-
veloped from these cells [in Russian], Biomed. Khim.,
66, 265-273, https://doi.org/10.18097/PBMC20206603265.
124. Moskaleva, E. Y., Vysotskaya, O. V., Zhorova, E. S.,
Shaposhnikova, D. A., Saprykin, V. P., Cheshigin, I. V.,
Smirnova, O.D., and Zhirnik, A.S. (2023) Late effects
of γ, n-irradiation of mice: shortening of telomeres
and tumors development [in Russian], Med. Radi-
ol. Radiat. Safety, 68, 11-18, https://doi.org/10.33266/
1024-6177-2023-68-5-11-18.
125. Moskaleva, E. Y., Romantsova, A. N., Semochkina,
Y. P., Rodina, A. D., Cheshigin, I. V., Degtyarev, A.,
and Zhirnik, A. (2021) Analysis of the appearance
of micronuclei in the erythrocytes and activity of
bone marrow cells proliferation after the prolonged
low dose Fast neutrons irradiation of mice [in Rus-
sian], Med. Radiol. Radiat. Safety, 66, 26-33, https://
doi.org/10.12737/1024-6177-2021-66-6-26-33.
MOSKALEVA et al.722
BIOCHEMISTRY (Moscow) Vol. 90 No. 6 2025
126. Berardinelli,F., Nieri,D., Sgura,A., Tanzarella,C., and
Antoccia, A. (2012) Telomere loss, not average telo-
mere length, confers radiosensitivity to TK6-irradiat-
ed cells, Mutat. Res., 740, 13-20, https://doi.org/10.1016/
j.mrfmmm.2012.11.004.
127. Wong, K. K., Chang, S., Weiler, S. R., Ganesan, S.,
Chaudhuri, J., Zhu, C., Artandi, S. E., Rudolph, K. L.,
Gottlieb, G. J., Chin, L., Alt, F. W., and DePinho, R. A.
(2000) Telomere dysfunction impairs DNA repair and
enhances sensitivity to ionizing radiation, Nat. Genet.,
26, 85-88, https://doi.org/10.1038/79232.
128. Ayouaz, A., Raynaud, C., Heride, C., Revaud, D., and
Sabatier, L. (2008) Telomeres: hallmarks of radiosen-
sitivity, Biochimie, 90, 60-72, https://doi.org/10.1016/
j.biochi.2007.09.011.
129. Lustig, A., Shterev, I., Geyer, S., Shi, A., Hu, Y.,
Morishita, Y., Nagamura, H., Sasaki, K., Maki, M.,
Hayashi, I., Furukawa, K., Yoshida, K., Kajimura, J.,
Kyoizumi, S., Kusunoki, Y., Ohishi, W., Nakachi, K.,
Weng, N. P., and Hayashi,T. (2016) Long term effects
of radiation exposure on telomere lengths of leuko-
cytes and its associated biomarkers among atom-
ic-bomb survivors, Oncotarget, 7, 38988-38998, https://
doi.org/10.18632/oncotarget.8801.
130. Yoshida, K., Misumi, M., Kubo, Y., Yamaoka, M.,
Kyoizumi,S., Ohishi,W., Hayashi,T., and Kusunoki,Y.
(2016) Long-term effects of radiation exposure and
metabolic status on telomere length of peripheral
blood T cells in atomic bomb survivors, Radiat. Res.,
186, 367-376, https://doi.org/10.1667/RR14389.1.
131. Scherthan, H., Sotnik, N., Peper, M., Schrock, G.,
Azizova, T., and Abend, M. (2016) Telomere length
in aged mayak PA nuclear workers chronically ex-
posed to internal alpha and external gamma radia-
tion, Radiat. Res., 185, 658-667, https://doi.org/10.1667/
RR14271.1.
132. Trickovic, J. F., Sobot, A. V., Joksic, I., and Joksic, G.
(2022) Telomere fragility in radiology workers oc-
cupationally exposed to low doses of ionising radi-
ation, Arh. Hig. Rada Toksikol., 73, 23-30, https://
doi.org/10.2478/aiht-2022-73-3609.
133. Xiao, C. Y., Zhou, F. X., Liu, S. Q., Xie, C. H., Dai, J.,
and Zhou, Y.F. (2005) Correlations of telomere length
and telomerase activity to radiosensitivity of human
laryngeal squamous carcinoma cells [in Chinese],
Chin. J. Cancer, 24, 653-656.
134. Zhong, Y. H., Liao, Z. K., Zhou, F. X., Xie, C. H., Xiao,
C. Y., Pan, D. F., Luo, Z. G., Liu, S. Q., and Zhou, Y. F.
(2008) Telomere length inversely correlates with ra-
diosensitivity in human carcinoma cells with the
same tissue background, Biochem. Biophys. Res.
Commun., 367, 84-89, https://doi.org/10.1016/j.bbrc.
2007.12.078.
135. Goytisolo, F. A., Samper, E., Martin-Caballero, J.,
Finnon, P., Herrera, E., Flores, J. M., Bouffler, S. D.,
and Blasco, M. A. (2000) Short telomeres result in
organismal hypersensitivity to ionizing radiation
in mammals, J. Exp. Med., 192, 1625-1636, https://
doi.org/10.1084/jem.192.11.1625.
136. Zongaro, S., Verri, A., Giulotto, E., and Mondello, C.
(2008) Telomere length and radiosensitivity in hu-
man fibroblast clones immortalized by ectopic telo-
merase expression, Oncol. Rep., 19, 1605-1609, https://
doi.org/10.3892/or.19.6.1605.
137. Drissi,R., Wu,J., Hu,Y., Bockhold, C., and Dome, J. S.
(2011) Telomere shortening alters the kinetics of the
DNA damage response after ionizing radiation in
human cells, Cancer Prev. Res. (Phila), 4, 1973-1981,
https://doi.org/10.1158/1940-6207.CAPR-11-0069.
138. Zhou, F.X., Xiong,J., Luo, Z.G., Dai,J., Yu, H.J., Liao,
Z. K., Lei, H., Xie, C. H., and Zhou, Y. F. (2010) cDNA
expression analysis of a human radiosensitive-radio-
resistant cell line model identifies telomere function
as a hallmark of radioresistance, Radiat. Res., 174,
550-557, https://doi.org/10.1667/RR1657.1.
139. Ferrandon, S., Saultier, P., Carras, J., Battiston-
Montagne, P., Alphonse, G., Beuve, M., Malleval, C.,
Honnorat,J., Slatter,T., Hung,N., Royds,J., Rodriguez-
Lafrasse,C., and Poncet,D. (2013) Telomere profiling:
toward glioblastoma personalized medicine, Mol.
Neurobiol., 47, 64-76, https://doi.org/10.1007/s12035-
012-8363-9.
140. Sharma, G. G., Hall, E. J., Dhar, S., Gupta, A., Rao,
P.H., and Pandita, T.K. (2003) Telomere stability cor-
relates with longevity of human beings exposed to
ionizing radiations, Oncol. Rep., 10, 1733-1736, https://
doi.org/10.3892/or.10.6.1733.
141. Bernal, A., and Tusell, L. (2018) Telomeres: implica-
tions for cancer development, Int.J. Mol. Sci., 19, 294,
https://doi.org/10.3390/ijms19010294.
142. Mirjolet, C., Boidot, R., Saliques, S., Ghiringhelli, F.,
Maingon, P., and Crehange, G. (2015) The role of
telomeres in predicting individual radiosensitivity of
patients with cancer in the era of personalized ra-
diotherapy, Cancer Treat. Rev., 41, 354-360, https://
doi.org/10.1016/j.ctrv.2015.02.005.
143. Afshinnekoo,E., Scott, R.T., MacKay, M.J., Pariset,E.,
Cekanaviciute, E., Barker, R., Gilroy, S., Hassane, D.,
Smith, S. M., Zwart, S. R., Nelman- Gonzalez, M.,
Crucian, B. E., Ponomarev, S. A., Orlov, O. I.,
Shiba,D., Muratani,M., Yamamoto,M., Richards, S.E.,
Vaishampayan, P.A., Meydan, C., et al. (2020) Funda-
mental biological features of spaceflight: advancing
the field to enable deep-space exploration, Cell, 183,
1162-1184, https://doi.org/10.1016/j.cell.2020.10.050.
144. Garrett-Bakelman, F. E., Darshi, M., Green, S. J., Gur,
R.C., Lin,L., Macias, B.R., McKenna, M.J., Meydan,C.,
Mishra, T., Nasrini, J., Piening, B. D., Rizzardi, L. F.,
Sharma,K., Siamwala, J.H., Taylor,L., Vitaterna, M.H.,
Afkarian, M., Afshinnekoo, E., Ahadi, S., Ambati, A.,
et al. (2019) The NASA twins study: a multidimen-
sional analysis of a year-long human spaceflight,
TELOMERE LENGTH AS PROGNOSTIC MARKER 723
BIOCHEMISTRY (Moscow) Vol. 90 No. 6 2025
Science, 364, eaau8650, https://doi.org/10.1126/science.
aau8650.
145. Luxton, J. J., McKenna, M. J., Taylor, L. E., George,
K. A., Zwart, S. R., Crucian, B. E., Drel, V. R., Garrett-
Bakelman, F. E., Mackay, M. J., Butler, D., Foox, J.,
Grigorev, K., Bezdan, D., Meydan, C., Smith, S. M.,
Sharma, K., Mason, C. E., and Bailey, S. M. (2020)
Temporal telomere and DNA damage responses in the
space radiation environment, Cell Rep., 33, 108435,
https://doi.org/10.1016/j.celrep.2020.108435.
146. Welsh, J., Bevelacqua, J. J., Keshavarz, M., Mortazavi,
S. A. R., and Mortazavi, S. M. J. (2019) Is telomere
length a biomarker of adaptive response in space?
Curious findings from NASA and residents of high
background radiation areas, J. Biomed. Phys. Eng., 9,
381-388, https://doi.org/10.31661/jbpe.v9i3Jun.1151.
147. Garcia-Medina, J.S., Sienkiewicz,K., Narayanan, S.A.,
Overbey, E. G., Grigorev, K., Ryon, K. A., Burke, M.,
Proszynski, J., Tierney, B., Schmidt, C. M., Mencia-
Trinchant, N., Klotz, R., Ortiz, V., Foox, J., Chin, C.,
Najjar, D., Matei, I., Chan, I., Cruchaga, C., Klein-
man, A., et al. (2024) Genome and clonal hematopoi-
esis stability contrasts with immune, cfDNA, mito-
chondrial, and telomere length changes during short
duration spaceflight, Precis. Clin. Med., 7, pbae007,
https://doi.org/10.1093/pcmedi/pbae007.
148. Rolles, B., Tometten, M., Meyer, R., Kirschner, M.,
Beier, F., and Brummendorf, T. H. (2024) Inherited
telomere biology disorders: pathophysiology, clini-
cal presentation, diagnostics, and treatment, Trans-
fus. Med. Hemother., 51, 292-309, https://doi.org/
10.1159/000540109.
149. Savage, S. A. (2022) Dyskeratosis congenita and telo-
mere biology disorders, Hematol. Am. Soc. Hematol.
Educ. Program, 2022, 637-648, https://doi.org/10.1182/
hematology.2022000394.
150. Heiss, N.S., Knight, S.W., Vulliamy, T.J., Klauck, S.M.,
Wiemann, S., Mason, P. J., Poustka, A., and Dokal, I.
(1998) X-linked dyskeratosis congenita is caused by
mutations in a highly conserved gene with putative
nucleolar functions, Nat. Genet., 19, 32-38, https://
doi.org/10.1038/ng0598-32.
151. Calado, R. T., and Young, N. S. (2009) Telomere dis-
eases, N. Engl. J. Med., 361, 2353-2365, https://
doi.org/10.1056/NEJMra0903373.
152. Tummala, H., Walne, A., Buccafusca, R., Alnajar, J.,
Szabo, A., Robinson, P., McConkie-Rosell, A.,
Wilson, M., Crowley, S., Kinsler, V., Ewins, A. M.,
Madapura, P. M., Patel, M., Pontikos, N., Codd, V.,
Vulliamy, T., and Dokal, I. (2022) Germline thymi-
dylate synthase deficiency impacts nucleotide metab-
olism and causes dyskeratosis congenita, Am. J. Hum.
Genet., 109, 1472-1483, https://doi.org/10.1016/j.ajhg.
2022.06.014.
153. Tummala, H., Walne, A., and Dokal, I. (2022) The bi-
ology and management of dyskeratosis congenita
and related disorders of telomeres, Exp. Rev. He-
matol., 15, 685-696, https://doi.org/10.1080/17474086.
2022.2108784.
154. Niewisch, M. R., Beier, F., and Savage, S. A. (2023)
Clinical manifestations of telomere biology disor-
ders in adults, Hematol. Am. Soc. Hematol. Educ.
Program, 2023, 563-572, https://doi.org/10.1182/
hematology.2023000490.
155. Tummala,H., Walne, A.J., Badat,M., Patel,M., Walne,
A. M., Alnajar, J., Chow, C. C., Albursan, I., Frost,
J. M., Ballard, D., Killick, S., Szitanyi, P., Kelly, A. M.,
Raghavan, M., Powell, C., Raymakers, R., Todd, T.,
Mantadakis, E., Polychronopoulou, S., Pontikos, N.,
et al. (2024) The evolving genetic landscape of telo-
mere biology disorder dyskeratosis congenita, EMBO
Mol. Med., 16, 2560-2582, https://doi.org/10.1038/
s44321-024-00118-x.
156. Luchkin, A. V., Mikhailova, E. A., Galtseva, I. V.,
Fidarova, Z. T., Abramova, A. V., Davydova, Yu. O.,
Kapranov, N. M., Nikiforova, K. A., Kulikov, S. M.,
and Parovichnikova, E. N. (2023) Telomere length
of various blood and bone marrow cells in patients
with aplastic anemia [in Russian], Oncohematol-
ogy, 18, 57-64, https://doi.org/10.17650/1818-8346-
2023-18-3-57-64.
157. Suliman, M.E., Ansari, M.G.A., Rayis, M.A., Hamza,
M.A., Saeed, A.A., Mohammed, A. K., and Al-Daghri,
N. M. (2022) Telomere length and telomere re-
peat-binding protein in children with sickle cell dis-
ease, Pediatr. Res., 91, 539-544, https://doi.org/10.1038/
s41390-021-01495-6.
158. Roka,K., Solomou, E.E., and Kattamis,A. (2023) Telo-
mere biology: from disorders to hematological diseas-
es, Front. Oncol., 13, 1167848, https://doi.org/10.3389/
fonc.2023.1167848.
159. Skvortsov, D. A., Zvereva, M. E., Shpanchenko, O. V.,
and Dontsova, O. A. (2011) Assays for detection of
telomerase activity, Acta naturae, 3, 48-68, https://
doi.org/10.32607/20758251-2011-3-1-48-68.
160. Wang, R., Li, J., Jin, R., Ye, Q., Cheng, L., and Liu, R.
(2021) Nonradioactive direct telomerase activi-
ty detection using biotin-labeled primers, J. Clin.
Lab. Anal., 35, e23800, https://doi.org/10.1002/
jcla.23800.
161. Zhang, X., Lou, X., and Xia, F. (2017) Advances in
the detection of telomerase activity using isother-
mal amplification, Theranostics, 7, 1847-1862, https://
doi.org/10.7150/thno.18930.
162. Kim, N. W., Piatyszek, M. A., Prowse, K. R., Harley,
C. B., West, M. D., Ho, P. L., Coviello, G. M., Wright,
W. E., Weinrich, S. L., and Shay, J. W. (1994) Specific
association of human telomerase activity with immor-
tal cells and cancer, Science, 266, 2011-2015, https://
doi.org/10.1126/science.7605428.
163. Skvortsov, D. A., Zvereva, M. E., Rubtsova, M. P.,
Pavlova, L. S., Petrenko, A. A., Kiselev, F. L., and
MOSKALEVA et al.724
BIOCHEMISTRY (Moscow) Vol. 90 No. 6 2025
Dontsova, O. A. (2010) Optimized detection method
of telomerase activity in cancer diagnostics, Moscow
Univ. Chem. Bull., 65, 165-169, https://doi.org/10.3103/
S0027131410030119.
164. Demina, I.A., Semchenkova, A.A., Kagirova, Z.R., and
Popov, A. M. (2018) Flow cytometric measurement of
absolute telomere length, Pediatr. Hematol. Oncol.
Immunopathol., 17, 66-72, https://doi.org/10.24287/
1726-1708-2018-17-4-66-72.
165. Gutierrez-Rodrigues, F., Santana-Lemos, B. A.,
Scheucher, P. S., Alves-Paiva, R. M., and Calado, R. T.
(2014) Direct comparison of flow-FISH and qPCR as
diagnostic tests for telomere length measurement in
humans, PLoS One, 9, e113747, https://doi.org/10.1371/
journal.pone.0113747.
166. Zheng, Y. L., Wu, X., Williams, M., Verhulst, S.,
Lin, J., Takahashi, Y., Ma, J. X., and Wang, Y. (2024)
High-throughput single telomere analysis using DNA
microarray and fluorescent in situ hybridization,
Nucleic Acids Res., 52, e96, https://doi.org/10.1093/
nar/gkae812.
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.