ISSN 0006-2979, Biochemistry (Moscow), 2023, Vol. 88, No. 11, pp. 1763-1777 © Pleiades Publishing, Ltd., 2023.
Russian Text © The Author(s), 2023, published in Biokhimiya, 2023, Vol. 88, No. 11, pp. 2138-2155.
1763
REVIEW
Microgravity Effects and Aging Physiology:
Similar Changes or Common Mechanisms?
Andrey Yu. Ratushnyy
1
and Ludmila B. Buravkova
1,a
*
1
Institute of Biomedical Problems, Russian Academy of Sciences, 123007 Moscow, Russia
e-mail: buravkova@imbp.ru
Received July 18, 2023
Revised October 13, 2023
Accepted October 14, 2023
AbstractDespite the use of countermeasures (including intense physical activity), cosmonauts and astronauts develop
muscle atony and atrophy, cardiovascular system failure, osteopenia, etc. All these changes, reminiscent of age-related
physiological changes, occur in a healthy person in microgravity quite quickly– within a few months. Adaptation to the
lost of gravity leads to the symptoms of aging, which are compensated after returning to Earth. The prospect of interplan-
etary flights raises the question of gravity thresholds, below which the main physiological systems will decrease their func-
tional potential, similar to aging, and affect life expectancy. An important role in the aging process belongs to the body’s
cellular reserve– progenitor cells, which are involved in physiological remodeling and regenerative/reparative processes
of all physiological systems. With age, progenitor cell count and their regenerative potential decreases. Moreover, their
paracrine profile becomes pro-inflammatory during replicative senescence, disrupting tissue homeostasis. Mesenchymal
stem/stromal cells (MSCs) are mechanosensitive, and therefore deprivation of gravitational stimulus causes serious changes
in their functional status. The review compares the cellular effects of microgravity and changes developing in senescent
cells, including stromal precursors.
DOI: 10.1134/S0006297923110081
Keywords: microgravity, aging, cell senescence, mesenchymal stem/stromal cells (MSCs)
Abbreviations: ECM, extracellular matrix; MSCs,mesenchymal stem/stromal cells; Rb,retinoblastoma protein; ROS,reactive
oxygen species; RPM,Random Positioning Machine; SMG,simulated microgravity
* To whom correspondence should be addressed.
INTRODUCTION
Despite the careful pre-flight selection, factors re-
lated to space flight, primarily weightlessness/micrograv-
ity, increase the risk of deterioration of the space travel-
er’s health [1-3]. In this regard, pinpointing fundamental
mechanisms of adaptation to microgravity-related effects
at tissue, cellular, and molecular levels is one of the most
essential scientific problems for space medicine. Resem-
blance of the physiological changes occurring during
space flight to the processes developing during human
aging is of special interest that raises a crucial question
about the similarity of underlying molecular and cellular
mechanisms. At the same time, restoration and normal-
ization of physiological processes in organs and tissues
is observed after returning to Earth, but aging leads to
unidirectional, progressive pathological changes [1,4].
It is recognized that in many physiological systems
the long-term space flights could result in emergence of
signs typical to aging [4-7]. Studies carried out on the
MIR orbital station and the International Space Station
revealed that the long-term stay in space results in de-
creased bone density [8-11], dysfunction of the immune
system [12], issues related to cardiovascular system func-
tioning [13, 14] as well as lowered skeletal muscle mass
and strength [15]. Furthermore, cartilage tissue in the
musculoskeletal system is also noted to be affected.
In addition, the size of chondrogenic pellets, synthe-
sis of proteoglycans, and dynamic stiffness of three-
dimensional cartilage constructs are reduced [7]. Final-
ly, moderate hypothyroidism, increased stress hormone
(mainly catecholamines) and decreased sex steroid lev-
els, insulin resistance, as well as systemic pro-inflam-
matory state could be observed as well [6, 16]. Similar
RATUSHNYY, BURAVKOVA176 4
BIOCHEMISTRY (Moscow) Vol. 88 No. 11 2023
changes occur upon aging under Earth’s gravity, which
develop, however, much faster in spaceflights.
Degenerative changes in the musculoskeletal system
pose one of the most important risks upon exposure to
microgravity. In this regard, a recent meta-review sum-
marized the data from 25 experimental papers that an-
alyzed post-flight bone density level as well as in-flight
and post-flight bone biochemical markers in 148 and
124 subjects, respectively. It turned out that compared
with the pre-flight level the cosmonauts who spent more
than 28 days in space were found to have lower extremity
bone density decreased by 5.4% (n = 96). After landing,
level of the bone resorption markers decreased, and the
balance shifted toward prevailing bone formation [11].
Some of the mechanisms underlying decline in
bone density during spaceflight could highly resemble
those related to osteoporosis developed in aging and/or
due to physical inactivity. The reduced bone mass and
osteoporosis has been observed both in older people as
well as in immobilized or bedridden patients of any age
[6] primarily highlighting a reduced mechanical load on
the musculoskeletal system. In addition, the decreased
level of sex hormones such as testosterone and estro-
gen should be noted. Interestingly, the male astronauts
demonstrate both decreased workload and lowered tes-
tosterone level (comparable to the older men) [6, 17].
It should be noted that neither daily physical tasks nor
mechanical loads can fully prevent osteopenia devel-
opment.
Similar data were also observed in the animal study
with 16-week-old female C57BL/6J mice exposed to mi-
crogravity for 15 days during the STS-131 mission. It was
shown that the bone volume fraction and bone thickness
decreased by 6.2% and 11.9%, respectively. A more de-
tailed analysis revealed elevated number of osteoclasts
along with higher expression of the bone matrix metal-
loproteinases (MMP-1, -3, and -10). At the same time,
osteoblast expression of the CDKN1a gene encoding p21
(one of the cell cycle inhibitors and crucial markers of
cellular senescence) was upregulated by 3.3-fold. Hence,
it could indicate that alterations in bone homeostasis
under microgravity conditions underlie osteocyte oste-
olysis and p21-mediated arrest in osteoblast cell cycle
[18]. Thus, phenomenology of the long-term impact of
space flight factors on bone tissue physiology allows to
draw some parallels with the age-related changes sug-
gesting existence of similar cellular mechanisms.
Recent studies with astronauts identify more links
between the alterations related to spaceflight and aging.
Modern molecular biological methods allow to assess
the state of genetic apparatus upon different damages.
Thus, American researchers analyzed temporal changes
in the telomere (the ends of chromosomes) length and
DNA damage response (DDR) in the peripheral blood
mononuclear cells from 11 astronauts on board of the
ISS before, during, and after the long-term spaceflights
(up to a year). In this regard, shortened telomeric regions
is considered as one of the classic signs of aging, which
will be discussed in more detail below. Despite that all
space travelers underwent strict medical selection having
no health complaints, they had shorter telomere length
and lower level of telomerase activity compared to the
control age-matched healthy subjects. Interestingly, the
telomere length increased slightly during space flights,
but rapidly decreased upon return to Earth [19]. Hence,
it was unveiled that despite of the individual differences
the post-flight telomere length was decreased in almost
all astronauts in comparison with the pre-flight length.
Furthermore, a positive correlation between the oxida-
tive stress and dynamically fluctuating telomere length
was established. Aside from this, increased prevalence
of the chromosomal inversions was observed during and
after the space flights. It was proposed that regardless of
telomerase activity in somatic cells the in-flight chronic
oxidative stress transiently activates an alternative path-
way for telomere length regulation [19].
Discussing the data on the telomere length in-
creased during space flights, it should be noted that the
cosmonauts and astronauts use physical activity (about
an hour daily) as a countermeasure of adverse effects
related to weightlessness. Analyzing the effects of run-
ning revealed its positive effect on peripheral blood mono-
nuclear cell telomere length [20], the telomere length
positively correlated with the performance level of ath-
letes [21].
Considering telomere length (or any other sign of
aging) as one of an “integrative markers” for cumulative
effect of genetic and external cues (environment and
lifestyle) on human aging, it is necessary to remember
that together with microgravity, a healthy person be-
comes affected by the elevated level of radiation, tran-
sient changes in spacecraft normoxic atmosphere (in-
creased CO
2
level, airborne organic impurities), altered
microbiome, as well as stressful situations (e.g., extra-
vehicular activities, failures of life support system ele-
ments, etc.), which should be taken into consideration
while analyzing impact of space flights on physiological
processes.
Interestingly, the level of blood plasma pro-inflam-
matory interleukins and chemokines in astronauts sig-
nificantly correlated with the telomere length and in-
creased during the long-term space flights. For instance,
the astronauts’ plasma samples contained higher level of
various cytokines including pro-inflammatory cues such
as TNFα, IL-8, IL-1ra, Tpo, VEGF, MCP-1, CCL4,
CXCL5 [12] that could be considered as a sign of chron-
ic (sterile) inflammation, one of the signs of aging.
Microgravity is a major element affecting health of
astronauts during space flight [22]. Because physiologi-
cal changes at an organismal level result from the mod-
ified cellular functioning, cellular changes related to ag-
ing and microgravity were compared.
MICROGRAVITY AND AGING-RELATED EFFECTS 1765
BIOCHEMISTRY (Moscow) Vol. 88 No. 11 2023
It is necessary to remember that the central regula-
tory processes such as fluctuated biorhythms, hormonal
status etc. affecting the cell state (and aging) in the con-
text of an integral unified system also greatly impact the
gravity-related physiological effects. These effects are the
subject of separate investigation. In particular, they being
considered in the Dilman’s neuroendocrine theory of ag-
ing and in the hypothesis suggested by A. M. Olovnikov
about potential molecular mechanisms for the develop-
ment and aging of multicellular organisms, including in-
volvement of neuroendocrine cells and gravitational ef-
fects. Moreover, perception of the gravitational infradian
rhythms by the animals, which could become disrupted
during space flight and, probably, result in accelerated
aging, has been also mentioned [23-27].
CELL SENESCENCE
Nowadays, aging is often envisioned as the loss of
body physiological integrity that proceeds over time re-
sulting in its impaired functions and elevated risk of
death. Hypotheses, theories, definitions of aging at var-
ious levels of life organization have been extensively dis-
cussed in the Biochemistry (Moscow) journal including the
Issue12-13, 2022. In this review we will solely refer to the
hallmark issues. It is also worth noting about an epigen-
etic “clock” of aging proposed in 2013 by Horvath [28]
and in 2017 by the scientific team headed by Dr.Gladyshev
[29]. However, a rather broad application of the “clock”
concept with regard to space biology needs to be further
assessed.
Currently, at the cellular level, altered intercellular
interaction, depleted stem cell pool, cellular aging, mi-
tochondrial dysfunction, metabolic disorders, impaired
proteostasis, genome instability, telomere shortening,
and epigenetic changes are referred to as signs of ag-
ing at the cellular level [30]. Cell aging (senescence) is
thought to be one of the essential arms in the host ag-
ing [31]. Such phenomenon is characterized by the irre-
versibly arrested cell cycle and could be accompanied by
the prominent phenotypic changes including increased
autophagic events, modulated metabolism, chromatin
remodeling, production of proinflammatory cytokines,
etc. [32-36]. Morphological changes including larger
cell size and flattening should be highlighted among
the most recognized markers of cell senescence [37].
In addition, the senescence-associated β-galactosidase
(SA-β-gal) activity also increases [38] along with the
frequency of emerging of heterochromatic γ-H2AX
foci[39]. To some extent, formation of a senescent cell
may be considered as a consequence and as a cause of
aging. On the one hand, it results from alterations at the
molecular level, translated into the senescence program
at the cell level, which is an elementary unit of life or-
ganization. On the other hand, the senescent cell phys-
iology transforms dramatically leading to the impaired
tissue and organ functioning in the long term.
Cell senescence is generally referred to as an an-
tagonistic trait that has both negative and positive sides.
It is generally accepted that activation of the senescence
state is the most crucial barrier to tumorigenesis. Uncon-
trolled tumor cell division and inability of the senescent
cell to divide seem to be opposite consequences resulting
from the same causes primarily represented by accumu-
lation of damages in the cell genetic material [40, 41].
Cell senescence also plays an essential role in wound
healing, where, e.g., upon skin wound healing, the ma-
tricellular protein CCN1 can induce senescence of fibro-
blasts or myofibroblasts and thereby reduce fibrosis[42].
Another aspect of the positive senescence-related effects
may also be presented as the changed intercellular cross-
talk. A number of paracrine mediators typical to senes-
cent secretome are even used for the stromal progenitor
cell priming in regenerative medicine. In recent years,
various approaches have been explored to extend capa-
bilities of the stromal progenitor cells that resulted in the
development of new cell products with improved poten-
tial for diverse clinical applications [43]. These studies
clearly illustrate ambiguity of negative and positive bi-
ological effects, which is also applicable to senescent
cells.
Today, replicative and stress-induced cellular se-
nescence are distinguished: the former is considered as
a cell condition in which proliferative activity becomes
irreversibly lost following a series of mitoses. The cell
cycle arrest (in this case) largely results from the short-
ening of telomeric regions, which can be considered as
a special case of genome instability. In 1961, cellular se-
nescence was first described as the progressive and irre-
versible loss of proliferative potential in human somatic
cells. It was shown that even under ideal culture condi-
tions, human embryonic fibroblasts are capable of divid-
ing only a limited number of times (50 ± 10) [44], the
phenomenon named after the author, who discovered
it– the “Hayflick limit”. In 1971 A. M. Olovnikov pro-
posed a hypothesis to explain this phenomenon based
on the data regarding the principles of DNA synthesis
in cells [45], this hypothesis was later confirmed ex-
perimentally. It states that with each cell division chro-
mosomes become slightly shortened due to underrepli-
cation of telomeric DNA region. Human telomeres are
the terminal regions of chromosomes that contain from
4 to 15 thousand base pairs and consist of TTAGGG
repetitive sequences. It is known that DNA polymerase
is unable to synthesize a daughter DNA copy from the
end of its chain – it can only add nucleotides to the
pre-existing 3′-hydroxyl group, i.e., requires an RNA
primer. Upon removal of the last primer at the 3′end,
the daughter strand will inevitably be shorter leading to
gradual loss of telomeric nucleotides during successive
cycles of DNA synthesis [46].
RATUSHNYY, BURAVKOVA1766
BIOCHEMISTRY (Moscow) Vol. 88 No. 11 2023
The stress-induced cellular senescence is also char-
acterized by irreversible cell cycle arrest. Unlike in the
case of replicative cellular senescence, it is not associat-
ed with the number of cell divisions. The stress-induced
senescence is triggered by the sublethal damages or on-
cogene activation [36, 47]. Most often, this condition
is related to oxidative stress, i.e., an imbalance between
the oxidants [usually reactive oxygen species (ROS)]
and antioxidant systems. An effect of ROS on DNA in-
cluding mtDNA induces formation of the products of
oxidative DNA base damage such as 8-hydroxy-2′-deox-
yguanosine (8OHdG) and could also lead to the strand
breaks [48]. Apart from this, mitochondrial ROS could
activate the c-Jun N-terminal kinase (JNK), which, in
turn, facilitates release of the chromatin fragments into
cytoplasm and activation of the pro-inflammatory se-
cretome [49]. Interestingly, oxidative stress could also
accelerate telomere shortening [50] probably owing to
high content of guanine (G), which is most vulnerable
to ROS [51].
The programs of replicative and stress-induced cel-
lular senescence are executed via similar mechanisms.
Relatively small DNA damage results in the temporary
arrest of the cell cycle. After successful DNA repair, a
cell can begin to divide again. By definition, this con-
dition cannot be called cell senescence. More profound
damage that cannot be repaired for a long time triggers
a chronically activated DDR signaling cascade, cell re-
sponse to the genetic material damage. The chronically
activated DDR usually occurs upon multiple DNA dam-
ages and represents a major hallmark of the senescent
state– stable cell cycle arrest. The latter is achieved via
activation of the p16INK4a/Rb and p53/p21CIP1 tu-
mor suppressor signaling cascades [47, 51] so that the
cell would never be able to start division again. Sup-
pression of activity of the relevant cyclin-dependent ki-
nases by both inhibitors, p21 and p16 (encoded by the
CDKN1A and CDKN2A genes, respectively), results in
hypophosphorylation of the retinoblastoma protein(Rb).
The hypophosphorylated Rb is able to bind E2F family
transcription factors regulating cell cycle [52,53]. By re-
versibly binding and, consequently, functionally inacti-
vating E2F proteins, Rb controls expression of the genes,
products of which are essential for regulating cell cycle
as well as blocking the G1-to-S phase transition. In this
scenario, the p53/p21 pathway is predominantly activat-
ed first by preventing proliferation of the cells with se-
rious DNA damage, whereas the p16/Rb-axis becomes
involved somewhat later [32]. However, based on the
cellular context, one or another may become preferable.
Cellular senescence could be affected by various
mechanical forces including shear stress, tension, and
pressure [54,55]. This raises the question whether mi-
crogravity could be considered as a factor causing cellu-
lar senescence or other similar changes in the cell phys-
iology?
SENESCENCE-ASSOCIATED
ALTERATIONS UNDER
SIMULATED MICROGRAVITY
During the space flight, human body is exposed to
multiple unfavorable stress factors noted above that can
aggravate development of the aging-associated signs.
At the cellular level, they may be presented by the
changed radiation background able to disrupt DNA in-
tegrity and promote oxidative stress. In this review, the
microgravity-related effects will be discussed in detail.
In this review, the senescence-associated chang-
es occurring in the diverse cell types will be assessed
in the simulated microgravity (SMG) settings. Due to
the technical limitations posed by experiments in space
missions, various ground-based models are common-
ly used. Usually, such models are aimed at gravitation-
al “unloading” to simulate some of the microgravity-
related effects. Ground-based experiments also allow to
avoid an impact from the increased background radia-
tion and other space flight-associated factors. To sim-
ulate microgravity effects on cell cultures, rotating wall
vessels (RWV) and 2D/3D clinostats such as a device
for randomizing object position relative to the gravity
vector (Random Positioning Machine, RPM) are used
most often. It is believed that the computer-controlled
RPM consisting of two frames rotating in two perpen-
dicular planes represents the most suitable approach for
simulating microgravity effects on the adherent cell cul-
tures. Using the RPM allows to randomize object’s po-
sition relative to the gravity vector. In standard operating
modes, the device simulates gravitational acceleration
equivalent to 10
–2
g [56-58].
Various cell cultures including immortalized lines,
endothelium, stromal precursors, etc. have been used in
the studies [59-63]. A wide range of changes demon-
strating direct gravity-related effects on the cellular
structures were revealed in the studies investigating
invitro cell morphofunctional state. A change in posi-
tion of the heavy organelles such as nucleus results in
redistributed load on the cytoskeleton followed by its
reorganization; modifications in the adhesion molecule
physical interaction between the cell and extracellular
matrix also occur. Altogether, it results in the altered
gene expression, changes in functioning of multiple
proteins, and overall modification of the cell functional
state [64-68].
Some studies using SMG attempted to identify
ac tivated senescence in the pheochromocytoma cells
(PC12), erythrocytes, skeletal muscle myoblasts, and
cardiomyocytes [59, 69-71]. Wang et al. [59] used SMG
to analyze early (6-96 h) effects on the rat neuro-
nal PC12 cell line. It was shown that the cell cycle
was arrested in the G1 phase along with the increased
SA-β-gal activity as well as activated p53 and p16 signal-
ing cascades associated with senescence. More detailed
MICROGRAVITY AND AGING-RELATED EFFECTS 1767
BIOCHEMISTRY (Moscow) Vol. 88 No. 11 2023
analysis revealed elevated level of ROS, which could
induce cellular senescence. Activity of the intracellu-
lar antioxidant enzymes such as superoxide dismutase
(SOD), glutathione peroxidase (GSH-Px), and catalase
(CAT) were markedly enhanced 12h later but decreased
96 h after onset of the experiment. Moreover, the an-
tioxidant N-acetylcysteine was able to profoundly block
the ROS-associated effects that prominently abrogated
the microgravity-related increase in the SA-β-gal activ-
ity. These data allowed to suggest that SMG exposure
elicits cellular senescence in the PC12 cells via enhanced
oxidative stress [59].
Modifications in the human erythrocyte structure
and functions were assessed by using a 3D clinostat.
For this, the erythrocyte structural parameters were
analyzed together with the cellular senescence-specific
metabolic parameters. The obtained data show that the
long-term microgravity exposure results in emergence of
the senescence-specific morphological patterns [69].
It was shown that SMG with the Zeromo 3D cli-
nostat accelerates senescence in the human skeletal
muscle myoblast culture. A markedly decreased prolifer-
ation, typical cytoskeletal remodeling, and hypertrophy
of the cell nuclei as well as upregulated SA-β-gal expres-
sion were demonstrated. Similar changes are observed
in the senescent myoblasts after several passages. It was
noted that such effects were sustained even after return-
ing to the normal gravitational conditions. Moreover,
the myoblasts exposed to SMG demonstrated a reduced
ability to differentiate into myotubes [70].
More recently, studies were conducted with the
human iPSC (induced pluripotent stem cell)-derived
cardiomyocytes. It was revealed that exposure to SMG
resulted in chromosomal reorganizations, downregulat-
ed mitochondrial function, elevated ROS level, as well
as other signs indicative of cellular senescence. It is
worth noting that the following study limitations were
specifically highlighted, which are difficult to disagree
with: (i) short-term SMG exposure (48 h), (ii) further
experiments assessing durability of the detected changes
are required [71].
Other researchers tend to believe that ROS are
involved in the DNA damage during SMG exposure.
Forinstance, it has been noted that SMG induces DNA
damage and mitochondria-mediated apoptosis via in-
creased ROS production in the human promyelocytic
leukemia cells [72]. Earlier studies already revealed that
clinorotation affects mitochondria, one of the main cel-
lular free radical producers, thereby inducing apopto-
sis in the thyroid cells. In particular, it was found out
that 24 h after the onset of experiment 10% of the thy-
roid carcinoma cells (ONCO-DG1 cell line) entered
the Fas-dependent apoptotic pathway. Mitochondrial
destruction and redistribution, disrupted microtubules,
and activated effector caspase-3 were detected. Clinoro-
tation was also elicited apoptosis in the normal thyroid
cells (HTU-5) as evidenced by the caspase-3 activation
and elevated Fas and Bax protein levels [73].
Investigation of the mouse embryonic stem cells
demonstrated enhancement of the ROS-associated ef-
fects under SMG conditions. The authors added hy-
drogen peroxide to the cells and the number of DNA
double-strand breaks was analyzed. It turned out that
24-hour-exposure of the treated cells to microgravity
substantially elevated the degree of DNA damage [74].
In 2003, similar data were reported by Greco et al. [75]
demonstrating that the frequency of chromosomal ab-
errations increased by around 1.2-2.8-fold in the post-
flight blood samples exposed to X-ray radiation in
comparison with the pre-flight blood samples. At the
same time, another work shows that the early response
to bleomycin-induced DNA damage (the number of
γ-H2AX foci) was similar in both cells exposed to mi-
crogravity and those under control static ground con-
ditions [76].
Thus, a set of studies indicate that exposure to
SMG could result in the development of some signs of
cell senescence. Moreover, a mechanism involving ox-
idative stress has been even suggested for this phenom-
enon, which, to some extent, is associated with mito-
chondrial dysfunction. Nonetheless, not a single study
is available to confirm permanent cell cycle arrest, i.e.,
there is no clear evidence that after SMG exposure cells
can no longer proliferate. Therefore, it is too early to
conclude unequivocally that cellular senescence is initi-
ated under exposure to SMG. We believe that the effect
of SMG is hardly strong enough to cause such a pro-
found damage at the cellular level.
SENESCENCE-ASSOCIATED STROMAL
PROGENITOR FUNCTIONAL STATE
IN SMG SETTINGS
Mesenchymal stromal/stem cells (MSCs) or stro-
mal progenitors, which play a major role in renewal and
regeneration, have been found in almost all body tissues.
MSCs are involved in maintaining bone tissue homeo-
stasis, hematopoiesis, regulated immunomodulation, and
angiogenesis, etc. MSCs are of considerable interest
both for basic science and applied regenerative medicine
including age-related pathologies. To date, a consensus
has been reached allowing to refer the MSC-associated
beneficial effects to production of various secreted fac-
tors including extracellular matrix components and cy-
tokines [77-80].
Some studies suggest that pathological changes in
astronauts could be associated with the stromal precur-
sor cell senescence. Further assessment of the micro-
gravity-related effect on MSC senescence contributes
to understanding of the role of senescent cells in the
development of physiological and pathological changes
RATUSHNYY, BURAVKOVA1768
BIOCHEMISTRY (Moscow) Vol. 88 No. 11 2023
under spaceflight conditions [81]. Aging of an organism
correlates with the decreased MSC functional activity.
It decreases the rate of tissue repair typical to aging.
For instance, osteoporotic bone fractures in older peo-
ple heal more slowly due to the decreased MSC func-
tions and their count [82].
As noted above, oxidative stress may be the main
cause underlying cell damage. Microgravity is a stressor
potentially able to induce ROS production, ultimately
resulting in various damages to subcellular compart-
ments [83]. Increased level of free radicals and mito-
chondrial dysfunction in MSCs have been noted in
the recent study. However, addition of the antioxidant
restored mitochondrial functions and reversed cell se-
nescence. Moreover, SMG promoted expression of YAP
(Yes-associated protein) and its translocation into the
nucleus. YAP is an important effector in the Hippo sig-
naling pathway that regulates development, homeosta-
sis, and regeneration [81, 84, 85]. It is known that YAP
could regulate cell senescence by acting on ATM (atax-
ia-telangiectasia mutated kinase), p53/p21, p16/CDK/
Rb, autophagy, AMPK, mTOR, and SIRT1 signaling
pathways [86-88]. Verteporfin (VP), YAP inhibitor, re-
stored the SMG-induced MSC mitochondrial dysfunc-
tion and senescence [81].
Some other studies also suggested that SMG causes
stromal progenitor cell senescence. Molecular changes
associated with stemness (OCT-4, SOX2, NANOG) and
cellular senescence (p19, p21, p53) in the MSCs isolat-
ed from the Wharton’s jelly were analyzed. The authors
supposed that the results of the study indicate cellular
adaptation occurring within the first hours after expo-
sure followed by loss of stemness and emergence of the
signs of molecular senescence program [89]. Bellow the
main physiological parameters of MSCs exposed to mi-
crogravity will be discuss in more detail.
Proliferation. Proliferation is one of the major cell
characteristics particularly in the context of senescence.
Firstly, MSC senescence is typically characterized by
permanent cell cycle arrest in the G1 phase, i.e., senes-
cent MSCs cannot proliferate and form colonies [36,
90, 91]. Several studies evidence, at least slightly decreas-
ed MSC proliferative potential upon SMG conditions.
For instance, over time (from 1h to 10 days) clinostating
resulted in lowered proliferation rate and changed mor-
phology of the cells, which became flatter and reached
confluency at lower density. When exposure period pro-
longed up to 20 days, proliferation rate also decreased,
while the number of large flat culture cells increased
[92, 93]. Such changes can also be the signs of senes-
cence. Study with the rat bone marrow MSCs confirms
the findings published previously [94]. It is noted that
SMG inhibits MSC growth at the G0/G1 phase of the
cell cycle.
Other studies not only failed to obtain similar re-
sults, but, on the contrary, observed the opposite effect.
Yuge et al. [95] showed that proliferation rate of the
human MSC in the 3D clinostat increased by almost
3-fold under SMG in comparison with the control
group. Hence, it was noted that SMG can be used to
enhance stem cell expansion in vitro. We investigated
functional status of the human adipose tissue-derived
MSCs under SMG conditions (96h) using RPM. It was
found that the cell count increased 1.5-2-fold, whereas
activity of the lysosomal compartment, cell size, and
granularity decreased. No change in the ROS levels or
mitochondrial transmembrane potential was observed.
Therefore, this study suggests lack of the signs associ-
ated with cell stress during MSCs culturing under SMG
settings [96].
Investigation of the more committed MSCs osteo-
blasts, showed that SMG did not affect cell growth or
viability. Cells were incubated in the 3D clinostat for
12-96 h. Twenty-four hours after the onset of the ex-
periment, the Bax/Bcl-2 mRNA level ratio (parameter
of apoptosis) increased up to 136% relative to the static
control. However, it was accompanied by the increase of
XIAP (anti-apoptotic molecule) mRNA level to 138% of
the static control. No DNA fragmentation was observed
and the level of the effector caspase-3 mRNA remained
unaltered [97].
Thus, it is at least premature to draw conclusions
about the lowered MSC proliferation or enhanced apop-
tosis under SMG conditions, which requires further
detailed investigated.
Differentiation. Reduced multipotency is consid-
ered as an important feature of senescent MSCs [90, 91],
which may weaken their reparative potential in all tissue
types. Shift in the balance between adipocyte and osteo-
cyte lineages has been observed, although a specific vec-
tor for it remains debatable. Some studies noted decline
in the osteogenic properties of MSC with increased du-
ration of the cells culturing or upon senescence [98-100].
Other studies revealed no such changes or even reported
elevated osteogenic potential [101, 102]. Such data ob-
tained in different studies are usually accounted for by
different methodological approaches and experimental
models, heterogeneity of the MSC populations, as well as
lack of assays accurately assessing osteogenic differentia-
tion [98, 103]. The most informative invitro marker of os-
teogenic potential is presented by relevant differentiation
pathway followed by detection of matrix mineralization.
At the same time, elevated level of cell death may result
in the false-positive data due to the large calcium ion re-
lease from the dying cells and its binding to extracellular
matrix [36, 90, 91]. Regarding the adipocyte potential,
we are getting closer to a consensus. A somewhat wide
range of data has been accumulated, however, majority of
the researchers come to the conclusion that adipogenic
potential wanes upon senescence [91].
Our own data clearly and unambiguously indicate
reduced adipogenic potential of the MSCs isolated from
MICROGRAVITY AND AGING-RELATED EFFECTS 1769
BIOCHEMISTRY (Moscow) Vol. 88 No. 11 2023
adipose tissue during replicative senescence. This is
manifested by lack of pronounced formation of lipid in-
clusions during differentiation. A markedly downregu-
lated expression of the gene encoding the key transcrip-
tional regulator PPARγ was detected, which probably
underlies this phenomenon. On the other hand, signs
pointing at the reciprocally elevated osteogenic poten-
tial, despite the downmodulated expression of the genes
coding for some positive regulators of the osteoblastic
pathway (BMP2, BMP6, IGF1, IL1B) were additionally
observed. In the process, intensity of the matrix calci-
fication and osteoprotegerin concentration during dif-
ferentiation increased, which may be essential in the
context of atherosclerotic plaque calcification in elderly
people. Transcriptional activity of the key osteogene-
sis regulator (RUNX2) and some analyzed marker genes
(SPARC, SPP1, COL1A1, BGLAP) remained stable dur-
ing the replicative senescence of MSCs [104]. Despite
potentiated calcification of the matrix, its morphology
differed in the “young” and senescent cell cultures that
may indicate false positive data due to calcium release
[36, 90, 91]. At the same time, increased osteoprotegerin
production observed in the study may indicate a pro-os-
teogenic paracrine activity in the senescent MSCs [104].
A greater consensus was achieved on the issue regard-
ing osteogenic differentiation under SMG setting results,
with vast majority of the studies agrees that microgravity
contributes to the reduced osteogenic potential in MSC
[105]. In particular, such effects were demonstrated in the
rat [94, 106] and human [107, 108] bone marrow MSCs,
which was confirmed by Saxena etal. [109] revealing that
SMG inhibits the MSC-related osteoblastogenesis and
enhances adipocytogenesis under osteogenic conditions.
The authors believed that this process involves decreased
RhoA activity and cofilin phosphorylation, disruption of
F-actin stress fibers, as well as decreased focal adhesion
kinase-mediated integrin signaling. Other studies reported
that the lowered osteoblastogenesis under SMG settings
is, at least in part, caused by downmodulated integrin/
MAPK signaling [110].
Transcriptome analysis using a genome-wide mi-
croarray showed that 882 genes were downregulated, and
505 genes were upregulated after 24-hour SMG expo-
sure. In particular, a significantly reduced expression of
the osteocytic and chondrocytic genes, as well as higher
expression level of the adipocyte genes was noted [111].
More recently, attention was attracted to non-
canonical pathways of cell differentiation. For instance,
SMG turned out to promote MSC differentiation into
neurons evidenced by the upregulated expression of sev-
eral relevant markers. Moreover, secretion of neurotro-
phins such as nerve growth factor (NGF), brain-derived
neurotrophic factor (BDNF), or ciliary neurotrophic
factor (CNTF) was elevated [112]. In another study, the
rat MSCs were cultured for 72 h or 10 days in a clinos-
tat followed by expansion in various differential culture
media. It was found that the short-term exposure (72h)
promoted endothelial, neuronal, and adipogenic dif-
ferentiation, whereas the long-term exposure (10 days),
unexpectedly, facilitated MSC differentiation into osteo-
blasts. In addition, the short-term SMG exposure pro-
foundly downregulated RhoA activity, which, however,
increased with prolonging SMG exposure. Hence, such
data demonstrated that the duration of SMG exposure
regulates MSC differentiation via the RhoA-associated
pathway [113], which corroborated the above-mentioned
data obtained by Saxena etal. [109].
Thus, SMG can largely affect MSC differentiation
potential. Such effect is of inhibitory nature, at least
with regard to osteogenic differentiation, which may ex-
plain similar physiological consequences related to the
impaired bone metabolism.
Secretory activity. It is assumed that the senescent
cells could contribute to maintenance of chronic inflam-
mation and development of aging-associated diseases,
hence, investigating the issue of paracrine regulation is
of great importance. Senescent cells continue to interact
with the surrounding environment and exert local and
systemic effects primarily through paracrine regulation.
The secretory activity changes dramatically, and this
phenotype was even named accordingly SASP (senes-
cence-associated secretory phenotype). First of all, the
SASP is characterized by increased proinflammatory se-
cretome, although individual cues vary widely depend-
ing on the cell type and the way to induce senescence
[36, 90, 91, 114].
It was discovered that the level of one of the main
pro-inflammatory cytokines, IL-8, in the human bone
marrow-derived MSCs increased in the culture medium
during culturing in 2D clinostat by 1.4-3.2-fold (20-d
exposure), as well as 1.5-6-fold (10-d exposure) and
1.6-2.1-fold (20-d exposure) on average in the culture
subjected to RPM [115]. Later, studies with the human
adipose tissue-derived MSCs complemented these find-
ings. Assessing the changes in paracrine activity upon
96-hour exposure to RPM showed elevated production
of IL-8 accompanied by the decreased IL-6 level. At the
same time, higher production of VEGF, a key positive
angiogenesis regulator, was noted [96].
Our study demonstrated that the post-RPM MSC-
conditioned medium stimulated formation of the vas-
cular network in ovo, of the capillary-like network of
endothelial cells (ECs) in Matrigel, and the EC non-di-
rectional migration invitro. Such effects were explained
by the changed expression of the genes and proteins as-
sociated with angiogenesis, primarily by the higher level
of expression of the angiogenesis regulators SerpinE1,
SerpinF1, IGFBP, VEGF, and IL-8 along with upreg-
ulated transcription of the genes encoding proangiogen-
ic growth factors such as VEGF-c and VEGF-a. Hence,
such data demonstrate that microgravity may exert an
MSC-mediated effect on the EC functional state [63].
RATUSHNYY, BURAVKOVA1770
BIOCHEMISTRY (Moscow) Vol. 88 No. 11 2023
In addition, we found higher transcriptional activi-
ty of the brain-derived neurotrophic factor, BDNF [63],
corroborating the findings reported by Chen et al. [112]
about potentiated MSC differentiation into neuron-like
cells under SMG. Additionally, therapeutic efficacy of
the microgravity-cultured MSCs after spinal cord isch-
emia-reperfusion injury was assessed revealing high-
er count of the BDNF-positive and lower count of the
caspase-3-positive apoptotic astrocytes, as well as re-
stored cell motility, which implies existence of positive ef-
fect of the post-SMG MSCs on regenerative process [116].
From the viewpoint of assessing the senescent state
in progenitor cells, it is important to study the proin-
flammatory secretome. While analyzing the SMG-
related effect of the adipose tissue-derived MSCs on
the TNFα-mediated priming, it was shown that SMG
perse results in no changes in the surface expression of
ICAM-1 and HLA-ABC, which can be considered as
markers of pro-inflammatory cell activation. A weak-
ened MSC response to the TNFα priming upon SMG
exposure was documented in the process that was man-
ifested as downmodulated production of the TNFα-de-
pendent pleiotropic cytokines (IL-8 and MCP-1), ma-
trix remodeling proteases, and suppression of some
genes encoding growth factors and cytokines [117].
Along with the proinflammatory cytokines, a stim-
ulatory effect related to the 10-day exposure to micro-
gravity on paracrine activity of the osteocommitted and
intact MSCs was evaluated. It was found that the MSC
response to SMG depended on the degree of cell com-
mitment so that for the osteocommitted MSCs it was
less pronounced and manifested by higher production
of sclerostin, negative regulator of osteoblastogenesis.
In contrast, the intact MSCs were distinguished by low
osteoprotegerin production. Such SMG-related changes
may underlie shift of the bone homeostasis towards bone
resorption [118]. In this regard, it is worth reminding that
the osteoprotegerin level becomes increased during se-
nescence [104].
Thus, it may be noted that exposure to SMG leads
to elevated secretion of the pro-inflammatory cytokine
IL-8, which, in turn, could promote production of the
downstream cues such as VEGF. Experiments show that
the changes in the secretome could be considered for
application in regenerative medicine to enhance neuro-
and angiogenesis. It is known that the pro-inflammatory
SASP in the senescent cells could also positively affect
tissue regeneration, whereas potential negative conse-
quences could be realized only upon chronic exposure.
Nonetheless, it is still premature to suggest the existence
of similarities between the SMG exposure and senes-
cence in the context of secretome.
Extracellular matrix. In addition to cells perse and
relevant paracrine cues, extracellular matrix (ECM)
plays an important role in the body tissue functioning.
It varies greatly depending on localization and medi-
ates intercellular interactions. The cell-ECM crosstalk is
necessary for normal cell functioning including cell pro-
liferation and differentiation [119, 120]. Various ECM
components perform specific functions. Proteoglycans
retain water, deposit metabolites and growth factors
due to their molecular structure and large number of
charged groups [121]. On the other hand, collagen and
fibronectin as protein constituents ensure tissue me-
chanical properties that cells rely on to maintain their
own shape as well as to migrate. Together with other
ECM proteins such as elastin and laminin, they ensure
matrix elasticity.
Available publications point at changes of ECM in
the senescent cells associated with their catabolic phe-
notype. In particular, upregulated expression of proteo-
lytic enzymes (matrix metalloproteinases, adamalysins
(ADAMs), urokinases and cathepsins) and lowered
production of ECM structural components (collagens,
glycoproteins, proteoglycans) was demonstrated. Even-
tually, it results in the reduced tissue elasticity, basement
membrane damage, and increased ECM stiffness [122].
At the moment, studies are mainly focused on as-
sessing effects of the ECM produced by young and se-
nescent cells on functional activity of the cellular ele-
ments in the tissue niche. The study by Choi etal. [123]
showed that the senescent fibroblasts seeded on the
ECM derived from the early passage fibroblasts had
reduced SA-β-gal expression along with the decreased
free radical levels, restored mitochondrial potential, as
well as telomere elongation. And vice versa, decline in
proliferation of the “young” fibroblasts cultured on the
senescent cell-derived ECM was observed [123].
The study of bone marrow MSCs obtained from the
young (3 weeks) and old (18 weeks) animals described
altered ECM properties. For this, the MSCs from young
and old mice were seeded onto decellularized matrix
from the relevant cell groups. It turned out that the
ECM released from the young MSCs produced lowered
ROS level that decreased by 30-50% in both young and
senescent MSCs compared to the ECM derived from
the old MSCs or cells grown on plastic [124]. It was
also shown that cultivation of the synovial fluid-derived
senescent MSCs on the decellularized fetal matrix en-
hances the MSC potential to undergo chondro- and adi-
podifferentiation [125].
From the viewpoint of gravireception, the ECM–
integrin–cytoskeleton complex represents a mechano-
sensitive platform that coordinates cell and tissue func-
tional state in the gravitational field [68]. The Myoui’s
group assessed cell differentiation [106] while the rat
bone marrow MSCs were cultured for 2 weeks inside
the pores of calcium hydroxyapatite on a 3D clinostat.
It was discovered that compared with the control group,
alkaline phosphatase activity (a marker of osteoblastic
differentiation) reduced by 40%. The MSC-containing
composites implanted into the syngeneic rats revealed
MICROGRAVITY AND AGING-RELATED EFFECTS 1771
BIOCHEMISTRY (Moscow) Vol. 88 No. 11 2023
that bone formation was markedly lowered upon SMG
exposure. It is important to note that the lower amount
extracellular matrix was observed in the culture culti-
vated in clinostat, so these two events might be relat-
ed[106]. More recent work noted that exposure to SMG
resulted in elevated expression of the adhesion mole-
cules (ITGB1, CD44), MMP1 protease, as well as one
of the collagen types (ColIII) in the MSCs. MMP1 is
known to degrade interstitial collagens including ColIII.
It is likely that the authors observed a compensatory
reaction in this case. Expression of the FBN1 and VIM
genes was downregulated. FBN1 is an extracellular ma-
trix glycoprotein required for elastic fiber formation,
whereas VIM is the major intermediate filament in the
stromal progenitor cell cytoskeleton [126].
Recent studies in our laboratory complemented the
results obtained previously. It was shown that the 10-day
exposure to RPM results in the decreased level of col-
lagen components in ECM likely due to the decreased
collagen synthesis and protease activation. The present-
ed data demonstrate that the ECM-associated mole-
cules from both native and osteocommitted MSCs could
be involved in the bone matrix reorganization during
spaceflight [127].
Flight experiments on the Foton 10 spacecraft
demonstrated downregulated expression of the major
structural protein COL1A in the MG-63 osteoblast cell
line. In addition, the SJ-10 experiment showed that the
2-day-spaceflight was associated with the lowered level of
several genes encoding matrisome structural proteins and
elevated MMP1 expression in the bone marrow MSCs.
An inhibitory effect on the COL1A2 level was observed
additionally after 5 days of spaceflight. Based on the re-
sults of onboard experiments, it can be concluded that
the matrisome structural proteins are negatively affected
by microgravity at the transcriptional level [128,129].
It is easy to recognize certain similarities between
the changes occurring during activation of cellular se-
nescence and upon exposure to SMG. In both cases,
less amounts of the ECM constituents are produced,
which is accompanied by the higher protease activity.
Itis likely that the negative link between the matrix deg-
radation and osteogenic differentiation really exists.
CONCLUSION
Development of physiological/pathophysiological
alterations in humans during long-term space flights
may be a sign of the “atrophic syndrome” described re-
peatedly by G. Libertini [130]. This syndrome is char-
acterized by the lowered cell duplication capacity, de-
creased number of cells, substitution of specific cells
with nonspecific cells, hypertrophy of the remaining
specific cells, altered functioning of the cells with short-
ened telomeres, and altered cellular microenvironment
depending on the state of senescent cells [130]. The atro-
phic syndrome resulting from the unloading is consid-
ered reversible in the spaceflights by duration no more
than a year under condition when a fairly wide range of
preventive measures is performed on board of the or-
bital space stations. In our opinion, it is impossible to
determine whether there are some threshold values for
decline in gravity or a maximum permissible time spent
in the unloading environment without countermeasures,
below which the essential physiological systems would
lose own functional potential due to the processes simi-
lar to those occurring in aging. Moreover, almost noth-
ing is known about the microgravity-related effect on
the lifespan of different organisms including mammals.
Summarizing some experimental data of the micro-
gravity effects on the cells and comparing the identified
effects to the senescence-associated changes, it should be
noted that microgravity may probably trigger initial stag-
es of the stress-induced reactions. Some of the studies
discussed here directly suggest similar mechanisms un-
derlying the analogy response to microgravity and senes-
cence. Even upon the short-term exposure, such shifts
can cause lowered proliferation, skewed differentiation
pathway, altered secretory profile including paracrine
mediators and ECM-associated molecules. At the same
time, the question regarding reversibility of the chang-
es discovered during SMG remains open, because such
reversibility does not allow to infer senescence directly.
And, secondly, it should be noted that the vast majority
of experimental studies proving that the cellular senes-
cent state could be activated invitro rely on the short-
term microgravity exposures (24-72h), which is clearly
insufficient to enable the cell senescence program.
Contributions. L.B.B. proposed initial concept of
the study; L.B.B. and A.Yu.R. wrote the manuscript,
comparatively analyzed microgravity- and aging-related
effects discussed in the review.
Funding. The study was financially supported
equally by the program of fundamental research of
the State Scientific Center of the Russian Federation
Institute of Biomedical Problems of the Russian Acade-
my of Sciences (Topic65.3) and by the Russian Science
Foundation (grant no.21-75-10117).
Ethics declarations. The authors declare no conflict
of interest in financial or any other sphere. The article
contains no description of studies with human subjects
or animals performed by any of the authors.
REFERENCES
1. Demontis, G. C., Germani, M. M., Caiani, E. G., Barra-
vecchia, I., Passino, C., and Angeloni, D. (2017) Human
pathophysiological adaptations to the space environment,
Front. Physiol., 8, 547, doi:10.3389/fphys.2017.00547.
RATUSHNYY, BURAVKOVA17 72
BIOCHEMISTRY (Moscow) Vol. 88 No. 11 2023
2. Tran, K. N., and Choi, J. I. (2022) Mimic microgravi-
ty effect on muscle transcriptome under ionizing radia-
tion, Life Sci. Space Res., 32, 96-104, doi: 10.1016/j.lssr.
2021.12.002.
3. Patel, S. (2020) The effects of microgravity and space
radiation on cardiovascular health: from low-Earth or-
bit and beyond, Int.J. Cardiol. Heart Vasc., 30, 100595,
doi:10.1016/j.ijcha.2020.100595.
4. Vernikos, J., and Schneider, V. S. (2010) Space, grav-
ity and the physiology of aging: parallel or convergent
disciplines? A mini-review, Gerontology, 56, 157-166,
doi:10.1159/000252852.
5. Wang, E. (1999) Age-dependent atrophy and micrograv-
ity travel: what do they have in common? FASEBJ., 13,
S167-S174, doi:10.1096/fasebj.13.9001.s167.
6. Biolo, G., Heer, M., Narici, M., and Strollo, F. (2003)
Microgravity as a model of ageing, Curr. Opin. Clin.
Nutr. Metab. Care, 6, 31-40, doi: 10.1097/00075197-
200301000-00006.
7. Strollo, F., Gentile, S., Strollo, G., Mambro, A., and
Vernikos, J. (2018) Recent progress in space physiol-
ogy and aging, Front. Physiol., 9, 1551, doi: 10.3389/
fphys.2018.01551.
8. Burger, E. H., and Klein-Nulend, J. (1998) Microgravity
and bone cell mechanosensitivity, Bone, 22, 127S-130S,
doi:10.1016/s8756-3282(98)00010-6.
9. Keune, J. A., Branscum, A. J., Iwaniec, U. T., and Turn-
er, R. T. (2015) Effects of spaceflight on bone microarchi-
tecture in the axial and appendicular skeleton in growing
ovariectomized rats, Sci. Rep., 5, 18671, doi: 10.1038/
srep18671.
10. Gerbaix, M., Gnyubkin, V., Farlay, D., Olivier, C.,
Ammann, P., Courbon, G., Laroche, N., Genthial, R.,
Follet, H., Peyrin, F., Shenkman, B., Gauquelin-Koch, G.,
and Vico, L. (2017) One-month spaceflight compromises
the bone microstructure, tissue-level mechanical prop-
erties, osteocyte survival and lacunae volume in mature
mice skeletons, Sci. Rep., 7, 2659, doi: 10.1038/s41598-
017-03014-2.
11. Stavnichuk, M., Mikolajewicz, N., Corlett, T., Morris, M.,
and Komarova, S. V. (2020) A systematic review and
meta-analysis of bone loss in space travelers, NPJ Micro-
gravity, 6, 13, doi:10.1038/s41526-020-0103-2.
12. Crucian, B., Simpson, R. J., Mehta, S., Stowe, R.,
Chouker, A., Hwang, S. A., Actor, J. K., Salam, A. P.,
Pierson, D., and Sams, C. (2014) Terrestrial stress an-
alogs for spaceflight associated immune system dysreg-
ulation, Brain Behav. Immun., 39, 23-32, doi: 10.1016/
j.bbi.2014.01.011.
13. Sofronova, S. I., Tarasova, O. S., Gaynullina, D., Borzykh,
A. A., Behnke, B. J., Stabley, J. N., McCullough, D.J.,
Maraj, J. J., Hanna, M., Muller-Delp, J. M., Vinogra-
dova, O.L., and Delp, M.D. (2015) Spaceflight on the
Bion-M1 biosatellite alters cerebral artery vasomotor and
mechanical properties in mice, J. Appl. Physiol., 118,
830-838, doi:10.1152/japplphysiol.00976.2014.
14. Hughson, R. L., Yee, N. J., and Greaves, D. K. (2016)
Elevated end-tidal Pco
2
during long-duration spaceflight,
Aerospace Med. Hum. Perform., 87, 894-897, doi:10.3357/
AMHP.4598.2016.
15. Shen, H., Lim, C., Schwartz, A. G., Andreev-Andrievskiy, A.,
Deymier, A. C., and Thomopoulos, S. (2017) Effects
of spaceflight on the muscles of the murine shoulder,
FASEBJ., 31, 5466-5477, doi:10.1096/fj.201700320R.
16. Novikov, V. E., and Ilyin, E. A. (1981) Age-related reac-
tions of rat bones to their unloading, Aviat. Space Environ.
Med., 52, 551-553.
17. Strollo, F., Riondino, G., Harris, B., Strollo, G., Casaro-
sa, E., Mangrossa, N., Ferretti, C., and Luisi, M. (1998)
The effect of microgravity on testicular androgen secre-
tion, Aviat. Space Environ. Med., 69, 133-136.
18. Blaber, E. A., Dvorochkin, N., Lee, C., Alwood, J. S.,
Yousuf, R., Pianetta, P., Globus, R.K., Burns, B.P., and
Almeida, E.A. (2013) Microgravity induces pelvic bone
loss through osteoclastic activity, osteocytic osteolysis, and
osteoblastic cell cycle inhibition by CDKN1a/p21, PLoS
One, 8, e61372, doi:10.1371/journal.pone.0061372.
19. Luxton, J. J., McKenna, M. J., Lewis, A., Taylor, L. E.,
George, K. A., Dixit, S. M., Moniz, M., Benegas, W.,
Mackay, M. J., Mozsary, C., Butler, D., Bezdan, D.,
Meydan, C., Crucian, B. E., Zwart, S. R., Smith, S. M.,
Mason, C. E., and Bailey, S. M. (2020) Telomere
Length dynamics and DNA damage responses associat-
ed with long-duration spaceflight, Cell Rep., 33, 108457,
doi:10.1016/j.celrep.2020.108457.
20. Zhuikova, S. E., and Nagovitsyn, R. S. (2021) Influence
of running on some physiological and molecular biologi-
cal markers of human aging, Hum. Physiol., 47, 587-594,
doi:10.1134/S0362119721050133.
21. Simoes, H. G., Sousa, C. V., Dos Santos Rosa, T., da
Silva Aguiar, S., Deus, L.A., Rosa, E. C. C. C., Amato,
A. A., and Andrade, R. V. (2017) Longer telomere length
in elite master sprinters: relationship to performance and
body composition, Int. J. Sports Med., 38, 1111-1116,
doi:10.1055/s-0043-120345.
22. Prasad, B., Grimm, D., Strauch, S. M., Erzinger, G. S., Co-
rydon, T.J., Lebert, M., Magnusson, N.E., Infanger, M.,
Richter, P., and Krüger, M. (2020) Influence of micro-
gravity on apoptosis in cells, tissues, and other systems
invivo and invitro, Int.J. Mol. Sci., 21, 9373, doi:10.3390/
ijms21249373.
23. Fernando, H. J., and Bowers, D. (2021) Neuroendocrine
Theory of Aging, in Encyclopedia of Gerontology and Pop-
ulation Aging (Gu, D., and Dupre, M.E., eds) Springer,
Cham, doi:10.1007/978-3-030-22009-9_673.
24. Dilman, V. M., Revskoy, S. Y., and Golubev, A.G. (1986)
Neuroendocrine-ontogenetic mechanism of aging: toward
an integrated theory of aging, Int. Rev. Neurobiol., 28,
89-156, doi:10.1016/s0074-7742(08)60107-5.
25. Olovnikov, A. (2005) Lunasensor, infradian rhythms, telo-
meres, and the chronomere program of aging, Ann. NY
Acad. Sci., 1057, 112-132, doi:10.1196/annals.1356.006.
MICROGRAVITY AND AGING-RELATED EFFECTS 1773
BIOCHEMISTRY (Moscow) Vol. 88 No. 11 2023
26. Olovnikov, A. M. (2015) Chronographic theory of devel-
opment, aging, and origin of cancer: role of chronomeres
and printomeres, Curr. Aging Sci., 8, 76-88, doi:10.2174/
1874609808666150422114916.
27. Olovnikov, A. M. (2022) Planetary metronome as a regu-
lator of lifespan and aging rate: the metronomic hypoth-
esis, Biochemistry (Moscow), 87, 1640-1650, doi:10.1134/
S0006297922120197.
28. Horvath, S. (2013) DNA methylation age of human tis-
sues and cell types, Genome Biol., 14, R115, doi:10.1186/
gb-2013-14-10-r115.
29. Petkovich, D. A., Podolskiy, D. I., Lobanov, A. V., Lee,
S.G., Miller, R.A., and Gladyshev, V.N. (2017) Using
DNA methylation profiling to evaluate biological age
and longevity interventions, Cell Metab., 25, 954-960.e6,
doi:10.1016/j.cmet.2017.03.016.
30. López-Otín, C., Blasco, M. A., Partridge, L., Serrano, M.,
and Kroemer, G. (2013) Thehallmarks of aging, Cell,153,
1194-1217, doi:10.1016/j.cell.2013.05.039.
31. McHugh, D., and Gil, J. (2018) Senescence and aging:
causes, consequences, and therapeutic avenues, J. Cell
Biol., 217, 65-77, doi:10.1083/jcb.201708092.
32. Campisi, J., and d’Adda di Fagagna, F. (2007) Cellular se-
nescence: when bad things happen to good cells, Nat. Rev.
Mol. Cell Biol., 8, 729-740, doi:10.1038/nrm2233.
33. Ratushnyy, A. Y., Rudimova, Y. V., and Buravkova, L.B.
(2020) Replicative senescence and expression of auto-
phagy genes in mesenchymal stromal cells, Biochemistry
(Moscow), 85, 1169-1177, doi:10.1134/S0006297920100053.
34. Kuilman, T., Michaloglou, C., Mooi, W. J., and Peeper,
D.S. (2010) Theessence of senescence, Genes Dev., 24,
2463-2479, doi:10.1101/gad.1971610.
35. Salama, R., Sadaie, M., Hoare, M., and Narita, M. (2014)
Cellular senescence and its effector programs, Genes Dev.,
28, 99-114, doi:10.1101/gad.235184.113.
36. Ratushnyy, A. Y., and Buravkova, L. B. (2020) Cell senes-
cence and mesenchymal stromal cells, Hum. Physiol., 46,
85-93, doi:10.1134/S0362119720010132.
37. Imai, S., and Guarente, L. (2014) NAD
+
and sirtu-
ins in aging and disease, Trends Cell Biol., 24, 464-471,
doi:10.1016/j.tcb.2014.04.002.
38. Dimri, G. P., Lee, X., Basile, G., Acosta, M., Scott, G.,
Roskelley, C., Medrano, E.E., Linskens, M., Rubelj, I.,
and Pereira-Smith, O. (1995) Abiomarker that identifies
senescent human cells in culture and in aging skin invivo,
Proc. Natl. Acad. Sci. USA, 92, 9363-9367, doi:10.1073/
pnas.92.20.9363.
39. Narita, M., Nũnez, S., Heard, E., Narita, M., Lin,
A.W., Hearn, S.A., Spector, D.L., Hannon, G.J., and
Lowe, S. W. (2003) Rb-mediated heterochromatin for-
mation and silencing of E2F target genes during cellular
senescence, Cell, 113, 703-716, doi: 10.1016/s0092-8674
(03)00401-x.
40. Collado, M., Blasco, M. A., and Serrano, M. (2007) Cel-
lular senescence in cancer and aging, Cell, 130, 223-233,
doi:10.1016/j.cell.2007.07.003.
41. Hanahan, D., and Weinberg, R. A. (2011) Hallmarks
of cancer: the next generation, Cell, 144, 646-674,
doi:10.1016/j.cell.2011.02.013.
42. Jun, J. I., and Lau, L. F. (2010) Thematricellular protein
CCN1 induces fibroblast senescence and restricts fibrosis
in cutaneous wound healing, Nat. Cell Biol., 12, 676-685,
doi:10.1038/ncb2070.
43. Noronha, N. C., Mizukami, A., Caliári-Oliveira, C.,
Cominal, J. G., Rocha, J. L. M., Covas, D.T., Swiech, K.,
and Malmegrim, K. C. R. (2019) Priming approaches to
improve the efficacy of mesenchymal stromal cell-based
therapies, Stem Cell Res. Ther., 10, 131, doi: 10.1186/
s13287-019-1224-y.
44. Hayflick, L., and Moorhead, P. S. (1961) Theserial cul-
tivation of human diploid cell strains, Exp. Cell Res.,
25, 585-621, doi:10.1016/0014-4827(61)90192-6.
45. Olovnikov, A. M. (1971) Principle of marginotomy in
template synthesis of polynucleotides, Dokl. Akad. Nauk
SSSR, 201, 1496-1499.
46. Victorelli, S., and Passos, J. F. (2017) Telomeres and cell
senescence– size matters not, EBioMedicine, 21, 14-20,
doi:10.1016/j.ebiom.2017.03.027.
47. De Magalhães, J. P., and Passos, J. F. (2018) Stress, cell
senescence and organismal ageing, Mech. Ageing Dev.,
170, 2-9, doi:10.1016/j.mad.2017.07.001.
48. Gruber, J., Schaffer, S., and Halliwell, B. (2008) Themi-
tochondrial free radical theory of ageing– where do we
stand? Front. Biosci., 13, 6554-6579, doi:10.2741/3174.
49. Vizioli, M. G., Liu, T., Miller, K. N., Robertson, N. A.,
Gilroy, K., Lagnado, A. B., Perez-Garcia, A., Kiourtis, C.,
Dasgupta, N., Lei, X., Kruger, P. J., Nixon, C., Clark, W.,
Jurk, D., Bird, T. G., Passos, J. F., Berger, S. L., Dou, Z.,
and Adams, P. D. (2020) Mitochondria-to-nucleus retro-
grade signaling drives formation of cytoplasmic chromatin
and inflammation in senescence, Genes Dev., 34, 428-445,
doi:10.1101/gad.331272.119.
50. Von Zglinicki, T. (2002) Oxidative stress shortens telo-
meres, Trends Biochem. Sci., 27, 339-344, doi: 10.1016/
s0968-0004(02)02110-2.
51. Campisi, J. (2013) Aging, cellular senescence, and can-
cer, Annu. Rev. Physiol., 75, 685-705, doi: 10.1146/
annurev-physiol-030212-183653.
52. Sherr, C. J., and Roberts, J. M. (1999) CDK inhibitors:
positive and negative regulators of G1-phase progression,
Genes Dev., 13, 1501-1512, doi:10.1101/gad.13.12.1501.
53. Sherr, C. J., and McCormick, F. (2002) TheRB and p53
pathways in cancer, Cancer Cell, 2, 103-112, doi:10.1016/
s1535-6108(02)00102-2.
54. Tharp, K. M., Higuchi-Sanabria, R., Timblin, G. A.,
Ford, B., Garzon-Coral, C., Schneider, C., Muncie, J. M.,
Stashko, C., Daniele, J. R., Moore, A. S., Frankino, P. A.,
Homentcovschi, S., Manoli, S. S., Shao, H., Richards,
A.L., Chen, K. H., Hoeve, J. T., Ku, G. M., Hellerstein, M.,
Nomura, D. K., etal. (2021) Adhesion-mediated mech-
anosignaling forces mitohormesis, Cell Metab., 33, 1322-
1341.e13, doi:10.1016/j.cmet.2021.04.017.
RATUSHNYY, BURAVKOVA17 74
BIOCHEMISTRY (Moscow) Vol. 88 No. 11 2023
55. Duan, J. L., Ruan, B., Song, P., Fang, Z. Q., Yue, Z.S.,
Liu, J. J., Dou, G. R., Han, H., and Wang, L. (2022)
Shear stress-induced cellular senescence blunts liver re-
generation through Notch-sirtuin 1-P21/P16 axis, Hepa-
tology, 75, 584-599, doi:10.1002/hep.32209.
56. Van Loon, J. W. A. (2007) Some history and use of the
random positioning machine, RPM, in gravity related
research, Adv. Space Res., 39, 1161-1165, doi: 10.1016/
j.asr.2007.02.016.
57. Kopp, S., Warnke, E., Wehland, M., Aleshcheva, G.,
Magnusson, N. E., Hemmersbach, R., Corydon, T. J.,
Bauer, J., Infanger, M., and Grimm, D. (2015) Mecha-
nisms of three-dimensional growth of thyroid cells during
long-term simulated microgravity, Sci. Rep., 5, 16691,
doi:10.1038/srep16691.
58. Wuest, S. L., Richard, S., Kopp, S., Grimm, D., and
Egli, M. (2015) Simulated microgravity: critical review
on the use of random positioning machines for mam-
malian cell culture, BioMed Res. Int., 2015, 971474,
doi:10.1155/2015/971474.
59. Wang, J., Zhang, J., Bai, S., Wang, G., Mu, L., Sun, B.,
Wang, D., Kong, Q., Liu, Y., Yao, X., Xu, Y., and Li, H.
(2009) Simulated microgravity promotes cellular senes-
cence via oxidant stress in rat PC12 cells, Neurochem. Int.,
55, 710-716, doi:10.1016/j.neuint.2009.07.002.
60. Kapitonova, M. Y., Muid, S., Froemming, G. R., Yusoff,
W. N., Othman, S., Ali, A. M., and Nawawi, H. M. (2012)
Real space flight travel is associated with ultrastructural
changes, cytoskeletal disruption and premature senescence
of HUVEC, Malays.J. Pathol., 34, 103-113.
61. Ulbrich, C., Wehland, M., Pietsch, J., Aleshcheva, G.,
Wise, P., van Loon, J., Magnusson, N., Infanger, M.,
Grosse, J., Eilles, C., Sundaresan, A., and Grimm, D.
(2014) Theimpact of simulated and real microgravity on
bone cells and mesenchymal stem cells, BioMed Res. Int.,
2014, 928507, doi:10.1155/2014/928507.
62. Winkelmaier, G., Jabbari, K., Chien, L. C., Grabham, P.,
Parvin, B., and Pluth, J. (2023) Influence of simulated
microgravity on mammary epithelial cells grown as 2D
and 3Dcultures, Int.J. Mol. Sci., 24, 7615, doi:10.3390/
ijms24087615.
63. Ratushnyy, A., Ezdakova, M., Yakubets, D., and Bu-
ravkova, L. (2018) Angiogenic activity of human adi-
pose-derived mesenchymal stem cells under simulated
microgravity, Stem Cells Dev., 27, 831-837, doi:10.1089/
scd.2017.0262.
64. Ingber, D. E. (2003) Mechanobiology and diseas-
es of mechanotransduction, Ann. Med., 35, 564-577,
doi:10.1080/07853890310016333.
65. Louis, F., Deroanne, C., Nusgens, B., Vico, L., and Guig-
nandon, A. (2015) RhoGTPases as key players in mam-
malian cell adaptation to microgravity, BioMed Res. Int.,
2015, 747693, doi:10.1155/2015/747693.
66. Mao, X., Chen, Z., Luo, Q., Zhang, B., and Song, G.
(2016) Simulated microgravity inhibits the migration of
mesenchymal stem cells by remodeling actin cytoskeleton
and increasing cell stiffness, Cytotechnology, 68, 2235-
2243, doi:10.1007/s10616-016-0007-x.
67. Ratushnyy, A. Y., and Buravkova, L. B. (2017) Expression
of focal adhesion genes in mesenchymal stem cells under
simulated microgravity, Dokl. Biochem. Biophys., 477,
354-356, doi:10.1134/S1607672917060035.
68. Buravkova, L., Larina, I., Andreeva, E., and Grigoriev, A.
(2021) Microgravity effects on the matrisome, Cells,10,
2226, doi:10.3390/cells10092226.
69. Dinarelli, S., Longo, G., Dietler, G., Francioso, A.,
Mosca, L., Pannitteri, G., Boumis, G., Bellelli, A., and
Girasole, M. (2018) Erythrocyte’s aging in microgravity
highlights how environmental stimuli shape metabolism
and morphology, Sci. Rep., 8, 5277, doi:10.1038/s41598-
018-22870-0.
70. Takahashi, H., Nakamura, A., and Shimizu, T. (2021)
Simulated microgravity accelerates aging of human skel-
etal muscle myoblasts at the single cell level, Biochem.
Biophys. Res. Commun., 578, 115-121, doi:10.1016/j.bbrc.
2021.09.037.
71. Acharya, A., Nemade, H., Papadopoulos, S., Heschel-
er, J., Neumaier, F., Schneider, T., Rajendra Prasad, K.,
Khan, K., Hemmersbach, R., Gusmao, E.G., Mizi, A.,
Papantonis, A., and Sachinidis, A. (2022) Microgravi-
ty-induced stress mechanisms in human stem cell-de-
rived cardiomyocytes, iScience, 25, 104577, doi:10.1016/
j.isci.2022.104577.
72. Singh, R., Rajput, M., and Singh, R. P. (2021) Simulated
microgravity triggers DNA damage and mitochondria-me-
diated apoptosis through ROS generation in human pro-
myelocytic leukemic cells, Mitochondrion, 61, 114-124,
doi:10.1016/j.mito.2021.09.006.
73. Kossmehl, P., Shakibaei, M., Cogoli, A., Infanger, M.,
Curcio, F., Schönberger, J., Eilles, C., Bauer, J., Picken-
hahn, H., Schulze-Tanzil, G., Paul, M., and Grimm, D.
(2003) Weightlessness induced apoptosis in normal thy-
roid cells and papillary thyroid carcinoma cells via extrin-
sic and intrinsic pathways, Endocrinology, 144, 4172-4179,
doi:10.1210/en.2002-0171.
74. Ran, F., An, L., Fan, Y., Hang, H., and Wang, S. (2016)
Simulated microgravity potentiates generation of reac-
tive oxygen species in cells, Biophys. Rep., 2, 100-105,
doi:10.1007/s41048-016-0029-0.
75. Greco, O., Durante, M., Gialanella, G., Grossi, G.,
Pugliese, M., Scampoli, P., Snigiryova, G., and Obe, G.
(2003) Biological dosimetry in Russian and Italian as-
tronauts, Adv. Space Res., 31, 1495-1503, doi: 10.1016/
s0273-1177(03)00087-5.
76. Lu, T., Zhang, Y., Kidane, Y., Feiveson, A., Stodieck, L.,
Karouia, F., Ramesh, G., Rohde, L., and Wu, H. (2017)
Cellular responses and gene expression profile changes
due to bleomycin-induced DNA damage in human fi-
broblasts in space, PLoS One, 12, e0170358, doi:10.1371/
journal.pone.0170358.
77. Murphy, M. B., Moncivais, K., and Caplan, A. I. (2013)
Mesenchymal stem cells: environmentally responsive
MICROGRAVITY AND AGING-RELATED EFFECTS 1775
BIOCHEMISTRY (Moscow) Vol. 88 No. 11 2023
therapeutics for regenerative medicine, Exp. Mol. Med.,
45, e54, doi:10.1038/emm.2013.94.
78. Spees, J. L., Lee, R. H., and Gregory, C.A. (2016) Mech-
anisms of mesenchymal stem/stromal cell function, Stem
Cell Res. Ther., 7, 125, doi:10.1186/s13287-016-0363-7.
79. Andreeva, E., Bobyleva, P., Gornostaeva, A., and Bu-
ravkova, L. (2017) Interaction of multipotent mesenchy-
mal stromal and immune cells: bidirectional effects, Cyto-
therapy, 19, 1152-1166, doi:10.1016/j.jcyt.2017.07.001.
80. Fujii, S., and Miura, Y. (2022) Immunomodulatory and
regenerative effects of MSC-derived extracellular ves-
icles to treat acute GVHD, Stem Cells, 40, 977-990,
doi:10.1093/stmcls/sxac057.
81. Lv, W., Peng, X., Tu, Y., Shi, Y., Song, G., and Luo, Q.
(2023) YAP inhibition alleviates simulated microgravi-
ty-induced mesenchymal stem cell senescence via tar-
geting mitochondrial dysfunction, Antioxidants, 12, 990,
doi:10.3390/antiox12050990.
82. Moretta, L., Uccelli, A., and Pistoia, V. (2015) Mesen-
chymal stromal cells and immunity: Introductory over-
view, Immunol. Lett., 168, 127-128, doi: 10.1016/j.imlet.
2015.08.010.
83. Yatagai, F., Honma, M., Dohmae, N., and Ishioka, N.
(2019) Biological effects of space environmental factors:
A possible interaction between space radiation and mi-
crogravity, Life Sci. Space Res., 20, 113-123, doi:10.1016/
j.lssr.2018.10.004.
84. Moya, I. M., and Halder, G. (2019) Hippo-YAP/TAZ
signalling in organ regeneration and regenerative medi-
cine, Nat. Rev. Mol. Cell Biol., 20, 211-226, doi:10.1038/
s41580-018-0086-y.
85. Pobbati, A. V., and Hong, W. (2020) Acombat with the
YAP/TAZ-TEAD oncoproteins for cancer therapy, Thera-
nostics, 10, 3622-3635, doi:10.7150/thno.40889.
86. Childs, B. G., Gluscevic, M., Baker, D. J., Laberge,
R.M., Marquess, D., Dananberg, J., and van Deursen,
J.M. (2017) Senescent cells: an emerging target for dis-
eases of ageing, Nat. Rev. Drug Discov., 16, 718-735,
doi:10.1038/nrd.2017.116.
87. Xie, Q., Chen, J., Feng, H., Peng, S., Adams, U., Bai,
Y., Huang, L., Li, J., Huang, J., Meng, S., and Yuan, Z.
(2013) YAP/TEAD-mediated transcription controls cellu-
lar senescence, Cancer Res., 73, 3615-3624, doi:10.1158/
0008-5472.CAN-12-3793.
88. Yeung, Y. T., Guerrero-Castilla, A., Cano, M., Muñoz,
M.F., Ayala, A., and Argüelles, S. (2019) Dysregulation of
the hippo pathway signaling in aging and cancer, Pharma-
col. Res., 143, 151-165, doi:10.1016/j.phrs.2019.03.018.
89. Pala, R., Cruciani, S., Manca, A., Garroni, G., El Faqir,
M. A., Lentini, V., Capobianco, G., Pantaleo, A., and
Maioli, M. (2023) Mesenchymal stem cell behavior un-
der microgravity: from stress response to a premature
senescence, Int. J. Mol. Sci., 24, 7753, doi: 10.3390/
ijms24097753.
90. Turinetto, V., Vitale, E., and Giachino, C. (2016) Se-
nescence in human mesenchymal stem cells: functional
changes and implications in stem cell-based therapy, Int.J.
Mol. Sci., 17, 1164, doi:10.3390/ijms17071164.
91. Li, Y., Wu, Q., Wang, Y., Li, L., Bu, H., and Bao, J. (2017)
Senescence of mesenchymal stem cells (Review), Int. J.
Mol. Sci., 39, 775-782, doi:10.3892/ijmm.2017.2912.
92. Merzlikina, N. V., Buravkova, L. B., and Romanov, Y. A.
(2004) The primary effects of clinorotation on cultured
human mesenchymal stem cells, J.Gravitat. Physiol., 11,
P193-P194.
93. Gershovich, J. G., and Buravkova, L. B. (2007) Morpho-
functional status and osteogenic differentiation potential
of human mesenchymal stromal precursor cells during
in vitro modeling of microgravity effects, Bull. Exp. Biol.
Med., 144, 608-613, doi:10.1007/s10517-007-0387-1.
94. Dai, Z. Q., Wang, R., Ling, S. K., Wan, Y. M., and Li,
Y. H. (2007) Simulated microgravity inhibits the pro-
liferation and osteogenesis of rat bone marrow mesen-
chymal stem cells, Cell Prolif., 40, 671-684, doi:10.1111 /
j.1365-2184.2007.00461.x.
95. Yuge, L., Kajiume, T., Tahara, H., Kawahara, Y., Ume-
da, C., Yoshimoto, R., Wu, S. L., Yamaoka, K., Asashi-
ma, M., Kataoka, K., and Ide, T. (2006) Microgravity
potentiates stem cell proliferation while sustaining the
capability of differentiation, Stem Cells Dev., 15, 921-929,
doi:10.1089/scd.2006.15.921.
96. Ratushnyy, A. Yu., and Buravkova, L. B. (2016) Func-
tional state of multipotent mesenchymal stromal cells
during modeling the effects of microgravity [in Russian],
Aviakosm. Ekol. Med., 50, 24-29.
97. Nakamura, H., Kumei, Y., Morita, S., Shimokawa, H.,
Ohya, K., and Shinomiya, K. (2003) Antagonism be-
tween apoptotic (Bax/Bcl-2) and anti-apoptotic (IAP)
signals in human osteoblastic cells under vector-averaged
gravity condition, Ann. NY Acad. Sci., 1010, 143-147,
doi:10.1196/annals.1299.023.
98. Kim, M., Kim, C., Choi, Y. S., Kim, M., Park, C., and
Suh, Y. (2012) Age-related alterations in mesenchymal
stem cells related to shift in differentiation from osteo-
genic to adipogenic potential: implication to age-associ-
ated bone diseases and defects, Mech. Ageing Dev., 133,
215-225, doi:10.1016/j.mad.2012.03.014.
99. Despars, G., Carbonneau, C. L., Bardeau, P., Coutu,
D.L., and Beauséjour, C.M. (2013) Loss of the osteogen-
ic differentiation potential during senescence is limited to
bone progenitor cells and is dependent on p53, PLoS One,
8, e73206, doi:10.1371/journal.pone.0073206.
100. Yang, Y. K., Ogando, C. R., Wang See, C., Chang, T.Y.,
and Barabino, G.A. (2018) Changes in phenotype and dif-
ferentiation potential of human mesenchymal stem cells
aging in vitro, Stem Cell Res. Ther., 9, 131, doi: 10.1186/
s13287-018-0876-3.
101. Wagner, W., Horn, P., Castoldi, M., Diehlmann, A.,
Bork,S., Saffrich, R., Benes, V., Blake, J., Pfister, S., Eck-
stein, V., and Ho, A.D. (2008) Replicative senescence of
mesenchymal stem cells: a continuous and organized pro-
cess, PLoS One, 3, e2213, doi:10.1371/journal.pone.0002213.
RATUSHNYY, BURAVKOVA1776
BIOCHEMISTRY (Moscow) Vol. 88 No. 11 2023
102. Digirolamo, C. M., Stokes, D., Colter, D., Phinney, D. G.,
Class, R., and Prockop, D. J. (1999) Propagation and
senescence of human marrow stromal cells in culture:
a simple colony-forming assay identifies samples with
the greatest potential to propagate and differentiate,
Br. J. Haematol., 107, 275-281, doi:10.1046/j.1365-2141.
1999.01715.x.
103. Cheng, H., Qiu, L., Ma, J., Zhang, H., Cheng, M., Li, W.,
Zhao, X., and Liu, K. (2011) Replicative senescence of hu-
man bone marrow and umbilical cord derived mesenchy-
mal stem cells and their differentiation to adipocytes and
osteoblasts, Mol. Biol. Rep., 38, 5161-5168, doi:10.1007/
s11033-010-0665-2.
104. Ratushnyy, A. Yu., and Buravkova, L. B. (2022) Differen-
tiation potential of the mesenchymal stromal cells during
replicative senescence [in Russian], Aviakosm. Ekol. Med.,
56, 64-69.
105. Buravkova, L. B., Gershovich, P. M., Gershovich, J.G.,
and Grigor’ev, A. I. (2010) Mechanisms of gravitation-
al sensitivity of osteogenic precursor cells, Acta Naturae,
2, 28-36, doi:10.32607/actanaturae.10734.
106. Nishikawa, M., Ohgushi, H., Tamai, N., Osuga, K.,
Uemura, M., Yoshikawa, H., and Myoui, A. (2005) Theef-
fect of simulated microgravity by three-dimensional clinos-
tat on bone tissue engineering, Cell Transplant., 14, 829-
835, doi:10.3727/000000005783982477.
107. Zayzafoon, M., Gathings, W. E., and McDonald, J.M.
(2004) Modeled microgravity inhibits osteogenic differen-
tiation of human mesenchymal stem cells and increases ad-
ipogenesis, Endocrinology, 145, 2421-2432, doi:10.1210/
en.2003-1156.
108. Buravkova, L. B., Gershovich, P. M., Gershovich, J.G.,
and Grigoriev, A.I. (2013) Microgravity and mesenchy-
mal stem cell response, Curr. Biotechnol., 2, 217-225,
doi:10.2174/22115501113029990016.
109. Saxena, R., Pan, G., and McDonald, J. M. (2007) Os-
teoblast and osteoclast differentiation in modeled micro-
gravity, Ann. NY Acad. Sci., 1116, 494-498, doi:10.1196/
annals.1402.033.
110. Meyers, V. E., Zayzafoon, M., Gonda, S. R., Gathings,
W. E., and McDonald, J. M. (2004) Modeled microgravity
disrupts collagen I/integrin signaling during osteoblastic
differentiation of human mesenchymal stem cells, J.Cell.
Biochem., 93, 697-707, doi:10.1002/jcb.20229.
111. Sheyn, D., Pelled, G., Netanely, D., Domany, E., and
Gazit, D. (2010) Theeffect of simulated microgravity on
human mesenchymal stem cells cultured in an osteogenic
differentiation system: a bioinformatics study, Tissue Eng.
PartA, 16, 3403-3412, doi:10.1089/ten.tea.2009.0834.
112. Chen, J., Liu, R., Yang, Y., Li, J., Zhang, X., Li, J.,
Wang, Z., and Ma, J. (2011) Thesimulated microgravity
enhances the differentiation of mesenchymal stem cells
into neurons, Neurosci. Lett., 505, 171-175, doi:10.1016/
j.neulet.2011.10.014.
113. Xue, L., Li, Y., and Chen, J. (2017) Duration of simulat-
ed microgravity affects the differentiation of mesenchymal
stem cells, Mol. Med. Rep., 15, 3011-3018, doi:10.3892/
mmr.2017.6357.
114. Coppé, J. P., Desprez, P. Y., Krtolica, A., and Campisi,J.
(2010) The senescence-associated secretory phenotype:
the dark side of tumor suppression, Annu. Rev. Pathol., 5,
99-118, doi:10.1146/annurev-pathol-121808-102144.
115. Gershovich, Iu. G., and Buravkova, L. B. (2009) Interleu-
kine production in culture of mesenchymal stromal cells
of humans during simulation of the microgravity effects
[in Russian], Aviakosm. Ecolog. Med., 43, 44-50.
116. Kurose, T., Takahashi, S., Otsuka, T., Nakagawa, K.,
Imura, T., Sueda, T., and Yuge, L. (2019) Simulated mi-
crogravity-cultured mesenchymal stem cells improve re-
covery following spinal cord ischemia in rats, Stem Cell
Res.,
41, 101601, doi:10.1016/j.scr.2019.101601.
117. Ratushnyy, A., Yakubets, D., Andreeva, E., and Bu-
ravkova, L. (2019) Simulated microgravity modulates
the mesenchymal stromal cell response to inflammatory
stimulation, Sci. Rep., 9, 9279, doi:10.1038/s41598-019-
45741-8.
118. Zhivodernikov, I. V., Ratushnyy, A. Y., and Buravkova,
L. B. (2021) Secretory activity of mesenchymal stromal
cells with different degree of commitment under condi-
tions of simulated microgravity, Bull. Exp. Biol. Med., 170,
560-564, doi:10.1007/s10517-021-05106-6.
119. Aamodt, J. M., and Grainger, D. W. (2016) Extra-
cellular matrix-based biomaterial scaffolds and the
host response, Biomaterials, 86, 68-82, doi: 10.1016/
j.biomaterials.2016.02.003.
120. Choudhury, D., Tun, H. W., Wang, T., and Naing, M.W.
(2018) Organ-derived decellularized extracellular matrix: a
game changer for bioink manufacturing? Trends Biotech-
nol., 36, 787-805, doi:10.1016/j.tibtech.2018.03.003.
121. Hospodiuk, M., Dey, M., Sosnoski, D., and Ozbolat, I. T.
(2017) Thebioink: Acomprehensive review on bioprint-
able materials, Biotechnol. Adv., 35, 217-239, doi:10.1016/
j.biotechadv.2016.12.006.
122. Mavrogonatou, E., Pratsinis, H., Papadopoulou, A.,
Karamanos, N. K., and Kletsas, D. (2019) Extracellu-
lar matrix alterations in senescent cells and their signifi-
cance in tissue homeostasis, Matrix Biol., 75-76, 27-42,
doi:10.1016/j.matbio.2017.10.004.
123. Choi, H. R., Cho, K. A., Kang, H. T., Lee, J. B., Kaeber-
lein, M., Suh, Y., Chung, I. K., and Park, S. C. (2011)
Restoration of senescent human diploid fibroblasts by
modulation of the extracellular matrix, Aging Cell, 10,
148-157, doi:10.1111/j.1474-9726.2010.00654.x.
124. Sun, Y., Li, W., Lu, Z., Chen, R., Ling, J., Ran, Q., Jilka,
R.L., and Chen, X.D. (2011) Rescuing replication and
osteogenesis of aged mesenchymal stem cells by exposure
to a young extracellular matrix, FASEBJ., 25, 1474-1485,
doi:10.1096/fj.10-161497.
125. Lynch, K., and Pei, M. (2014) Age associated communica-
tion between cells and matrix: a potential impact on stem
cell-based tissue regeneration strategies, Organogenesis,
10, 289-298, doi:10.4161/15476278.2014.970089.
MICROGRAVITY AND AGING-RELATED EFFECTS 1777
BIOCHEMISTRY (Moscow) Vol. 88 No. 11 2023
126. Ebnerasuly, F., Hajebrahimi, Z., Tabaie, S.M., and Dar-
bouy, M. (2018) Simulated microgravity condition alters
the gene expression of some ECM and adhesion mole-
cules in adipose derived stem cells, Int.J. Mol. Cell. Med.,
7, 146-157, doi:10.22088/IJMCM.BUMS.7.3.146.
127. Zhivodernikov, I., Ratushnyy, A., and Buravkova, L.
(2021) Simulated microgravity remodels extracellular ma-
trix of osteocommitted mesenchymal stromal cells, Int.J.
Mol. Sci., 22, 5428, doi:10.3390/ijms22115428.
128. Carmeliet, G., Nys, G., and Bouillon, R. (1997) Micro-
gravity reduces the differentiation of human osteoblastic
MG-63 cells, J.Bone Min. Res., 12, 786-794, doi:10.1359/
jbmr.1997.12.5.786.
129. Zhang, C., Li, L., Jiang, Y., Wang, C., Geng, B., Wang,Y.,
Chen, J., Liu, F., Qiu, P., Zhai, G., Chen, P., Quan, R.,
and Wang, J. (2018) Space microgravity drives transdiffer-
entiation of human bone marrow-derived mesenchymal
stem cells from osteogenesis to adipogenesis, FASEB J.,
32, 4444-4458, doi:10.1096/fj.201700208RR.
130. Libertini, G. (2014) The programmed aging paradigm:
how we get old, Biochemistry (Moscow), 79, 1004-1016,
doi:10.1134/S0006297914100034.