ISSN 0006-2979, Biochemistry (Moscow), 2025, Vol. 90, No. 12, pp. 1883-1896 © The Author(s) 2025. This article is an open access publication.
Published in Russian in Biokhimiya, 2025, Vol. 90, No. 12, pp. 1981-1996.
1883
REVIEW
Antibiotics and Cellular Senescence:
An Unexplored Territory
Roman A. Zinovkin
1,a
* and Nataliya D. Kondratenko
1,2
1
A. N. Belozersky Institute of Physico-Сhemical Biology, Lomonosov Moscow State University,
119992 Moscow, Russia
2
Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University,
119234 Moscow, Russia
a
e-mail: roman.zinovkin@gmail.com
Received August 26, 2025
Revised November 17, 2025
Accepted November 27, 2025
AbstractAntibiotics are certainly the most important agents in the fight against human and animal bac-
terial infections. Widespread use of antibiotics has a positive impact on the treatment of infectious diseases
but may be accompanied by serious side effects. Clinical aspects of these side effects are well understood,
but nonspecific molecular targets are not fully recognized. It is generally known that many antibiotics can
damage mitochondria, intracellular organelles responsible for aerobic metabolism as well as regulating a
number of important processes, including cellular redox balance and inflammatory responses. Mitochondrial
dysfunction commonly leads to the development of oxidative stress and inflammation, which are known
stimuli of cellular senescence. On the other hand, the same stimuli could induce death of senescent cells.
Thus, mitotoxic antibiotics could influence both the cellular senescence process and elimination of senescent
cells. The effect of antitumor antibiotics on the induction of cell aging has been studied in detail, but the effect
of antibacterial antibiotics on this process is still essentially unknown. This review aims to draw attention of
the researchers to the possibility of accelerated cellular aging induced by common antibacterial antibiotics
and to discuss potential mechanisms of this process.
DOI: 10.1134/S0006297925602758
Keywords: antibiotics, side effects, cellular senescence, mitochondria, reactive oxygen species
* To whom correspondence should be addressed.
INTRODUCTION
Since their discovery, antibiotics have remained
indispensable tools in the fight against bacterial in-
fections, dramatically reducing infection-related mor-
tality and contributing substantially to the increased
life expectancy worldwide. However, their use is
accompanied by various non-specific effects, physi-
ological features of which are well documented [1].
At least part of these side effects could be attribut-
ed to the evolutionary relationship between bacteria
and mitochondria, which originated from the ancient
alphaproteobacteria  [2]. Consequently, many antibi-
otics that target bacterial replication or translation
also induce mitochondrial dysfunction [3]. The down-
stream consequences of mitochondrial dysfunction,
such as oxidative stress and inflammation, could, in
turn, trigger cell cycle arrest and drive cells into a
state of cellular senescence (CS).
Although numerous individual studies and re-
views have addressed antibiotic side effects and the
phenomenon of CS, the relationship between these
two events remains largely unexplored. The primary
exception is antitumor antibiotics, which are known
to induce CS through activation of the cellular DNA
damage response. This review briefly outlines current
understanding of the mechanisms of CS development,
primary and non-specific targets of antibacterial an-
tibiotics, and analyzes the limited available evidence
regarding their ability to promote CS or eliminate se-
nescent cells in human and animal systems.
ZINOVKIN, KONDRATENKO1884
BIOCHEMISTRY (Moscow) Vol. 90 No. 12 2025
CELLULAR SENESCENCE
Cellular aging (senescence) is defined as a sta-
ble cell cycle arrest accompanied by characteristic
phenotypic changes  [4] (Fig.  1). CS is intimately in-
volved in the processes such as embryogenesis, tis-
sue regeneration, suppression of carcinogenesis, and
aging. As an antitumor mechanism, CS prevents pro-
liferation of the potentially cancerous cells. Activation
of the tumor suppressor pathways p53/p21CIP1 and
p16INK4A/pRB plays a central role in the development
of CS [5, 6].
Aging cells remain viable, but their metabolic
and transcriptomic activities change, and they devel-
op a complex secretory phenotype (senescence-asso-
ciated secretory phenotype, SASP). This phenotype is
characterized by the synthesis of cytokines and in-
flammatory mediators, proteases, and growth factors
(such as IL-1α, IL-1β, IL-6, IL-8, and MMP) [7]. Senes-
cent cells can be eliminated by immune cells, this
process contributes to the tissue remodeling and re-
generation. However, under certain conditions, aging
cells are not completely removed, which contributes
to the development of pathology [8]. Factors secreted
by the senescent cells can affect neighboring cells in
a paracrine manner and disturb normal tissue func-
tions.
Cellular aging occurs in response to various en-
dogenous and exogenous stimuli. There are two main
types of CS: replicative and stress-induced (Fig.  1).
In the replicative aging, a cell that has divided many
times with shortened telomeres loses its ability to
proliferate, leading to the complete halt in the cell
cycle [9]. The stress-induced aging is caused by a wide
range of factors, such as mitogenic signals, oncogene
activation, radiation, oxidative and genotoxic stress,
epigenetic changes, chromatin disorganization, proteo-
stasis disruption, mitochondrial dysfunction, inflam-
matory responses, tissue damage signals, chemothera-
peutic agents, and nutrient deprivation [10,  11]. Aging
caused by DNA damage is triggered by a wide range of
chemical compounds, as well as ionizing or UV radia-
tion. Depending on the intensity of DNA damage, the
cell may die by apoptosis or progress to CS [12]. More
than fifty compounds have been identified [13] that
induce cellular aging, with the specific mechanism
of cellular aging development varying for different
groups of substances.
The most important physiologically significant
signs of CS are increase in the cell size, increase in
the activity of senescence-associated β-galactosidase
(SA-β-gal), accumulation of autofluorescent granules,
and SASP [13] (Fig.1). The increased autofluorescence
and the SA-β-gal activity result from accumulation
Fig. 1. Main pathways of cellular senescence (CS) induction and characteristics of senescent cells. ROS, reactive oxygen
species; p53, p16Ink4a, p21CIP1, senescence protein markers; SA-β-Gal, senescence-associated beta-galactosidase; IL-1α,
interleukin-1α; IL-1β, interleukin-1β; IL-6, interleukin-6; IL-8, interleukin-8; MMPs, matrix metalloproteinases. Details are
provided in the text.
ANTIBIOTICS AND CELLULAR SENESCENCE 1885
BIOCHEMISTRY (Moscow) Vol. 90 No. 12 2025
ofthe excessive amounts of lysosomal components in
cells. To date, dozens of phenotypes of stress-induced
CS have been described, which partially overlap with
each other [14]. These aging states can differ signifi-
cantly from each other and from the phenotype of
replicative aging. At the same time, it is still unclear
whether the cell cycle arrest constitutes the “true”
CS. Essential characteristics of this “true” CS are also
undefined.
ANTIBIOTICS: CLASSIFICATION
AND MECHANISM OF ACTION
Antibiotics constitute a heterogeneous group of
compounds belonging to various classes of chemical
substances, each characterized by a unique structure
and mechanism of action. Functionally, they are gen-
erally categorized into two types: bacteriostatic an-
tibiotics, which inhibit growth and reproduction of
microorganisms, and bactericidal antibiotics, which
kill bacterial cells. Although the term antibiotics
originally referred exclusively to the substances of
natural origin, it is now used more broadly to in-
clude semi-synthetic and fully synthetic antimicrobi-
al agents [15]. Based on the spectrum of their activi-
ty, antibiotics may be classified as narrow-spectrum,
targeting specific groups of bacteria (e.g., vancomycin
against Gram-positive organisms), or broad-spectrum,
acting against a wide range of bacterial species (e.g.,
tetracyclines, cephalosporins). Furthermore, antibi-
otics can be categorized according to their chemical
structure into classes such as β-lactams, aminoglyco-
sides, macrolides, tetracyclines, phenicols, glycopep-
tides, polymyxins, lincosamides, fluoroquinolones,
and rifamycins, among others [16].
The primary targets of antibiotic action in bacte-
ria include synthesis of the cell wall, proteins, nucleic
acids, mycolic acids, and folic acid (Table  1) [15,  17].
The table also lists ionophores, such as monensin, la-
salocid, and salinomycin, which disrupt intracellular
ion homeostasis [18]. These ionophores are used in
veterinary medicine.
Several bacterial-derived compounds that are
also classified as antibiotics are widely used in can-
cer therapy. These include the anthracyclines (doxo-
rubicin, daunorubicin, epirubicin, and idarubicin),
as well as bleomycin, dactinomycin, and mitomycin.
Their anticancer activity is mediated through multi-
ple mechanisms, including: (i)  DNA alkylation (e.g.,
certain anthracyclines); (ii)  DNA intercalation (doxo-
rubicin, daunorubicin, actinomycin  D); (iii)  inhibition
of topoisomerase  II (doxorubicin); (iv)  induction of
DNA strand breaks (bleomycin)  [19]. The resulting
DNA damage leads to cell cycle arrest and ultimately
to tumor cell death [20].
Table 1. Mechanism of action of the main groups of
antibiotics
Classification of antibiotics by mechanism of action
Cell wall synthesis
inhibitors
• penicillins
• cephalosporins
• glycopeptides
• β-lactamase inhibitors
• carbapenems
• β-lactams
• polypeptides
Protein synthesis
inhibitors
30S Subunit inhibitors
• aminoglycosides
• tetracyclines
50S Subunit inhibitors
• macrolides
• phenicols
• lincosamides
• oxazolidinone
• streptogramins
DNA synthesis
inhibitors
• fluoroquinolones
• 5-nitroimidazoles
Folic acid synthesis
inhibitors
• sulfonamides
Ionophores
that disrupt ion
conductivity
• carboxylic polyethers
• linear and cyclic peptides
NON-SPECIFIC TARGETS OF ANTIBIOTICS
Being small molecules (400-1200  Da), antibiotics
have good bioavailability and effectively block vital
bacterial functions in the tissues of the infected or-
ganism. On average, the low-molecular-weight com-
pounds can bind to 6-11 off-target molecules inside
the cell in addition to their primary target [21]. An-
tibiotics can cause side effects of varying severity,
which, according to the current data, are partly due
to their effect on the human microbiota and partly
to their interaction with non-specific cellular targets
[22]. Clinical aspects of antibiotic side effects have
been studied in great detail [1, 23], but nonspecific
intracellular interactions are only partially under-
stood.
As noted above, many antibiotic-associated side
effects arise from the evolutionary relationship be-
tween mitochondria and bacteria; mitochondria are
currently believed to have originated from the ancient
Alphaproteobacteria [2]. Consequently, numerous an-
tibiotics designed to target bacterial replication or
translation also exhibit varying degrees of mitochon-
drial toxicity (Table  2), although not all have been
experimentally shown to interact with nonspecific
ZINOVKIN, KONDRATENKO1886
BIOCHEMISTRY (Moscow) Vol. 90 No. 12 2025
Table 2. Nonspecific mitochondrial targets of selected antibiotics and their associated side effects
Antibiotics
Potential antibiotic
target in mitochondria
Effect at the cellular level
Side effects at the
organismal level
References
Oxazolidinones 50S ribosomal subunit inhibition of megakaryocyte
maturation
thrombocytopenia,
anemia
[28]
reduced COX-II production
in PBMCs
hyperlactatemia [29]
mitochondrial dysfunction
in optic nerve cells
optic neuropathy [30, 31]
Lincosamides 50S ribosomal subunit neuronal apoptosis neurotoxicity [32]
Phenicols 50S ribosomal subunit decreased transferrin receptor
expression and ferritin synthesis
sideroblastic
anemia
[33]
disruption of mitochondrial
protein synthesis and
subsequent impairment
of erythroid cell development
aplastic anemia [34]
Macrolides 50S ribosomal subunit neuronal apoptosis neurotoxicity [32]
cardiomyocyte apoptosis cardiotoxicity [35]
hepatocyte oxidative stress hepatotoxicity [35, 36]
Aminoglycosides 30S ribosomal subunit disruption of mitochondrial
protein synthesis in cochlear
cells
ototoxicity [36, 37]
renal tubular cell mitochondrial
dysfunction
nephrotoxicity [38, 39]
Tetracyclines 30S ribosomal subunit mitochondrial dysfunction
and nerve cell death
neurotoxicity [32]
Fluoroquinolones gyrase/topoisomerase mitochondrial dysfunction
and oxidative stress in tenocytes
tendinopathy [40, 41]
oxidative stress in chondrocytes chondrotoxicity [42]
mitochondrial dysfunction
and oxidative stress
in Müller cells
retinopathy [43]
decreased GLUT1 expression dysglycemia [44]
Nitroimidazoles gyrase/topoisomerase ROS-independent neuronal death neurotoxicity [32]
mitochondrial targets [3]. Principal mechanisms un-
derlying this toxicity include inhibition of respiratory
chain complexes, uncoupling of oxidative phosphory-
lation, disruption of mitochondrial protein transport,
and suppression of key reactions within the tricar-
boxylic acid cycle [24]. Comprehensive discussions of
these nonspecific mitochondrial effects can be found
in several recent review articles [2, 25-27].
In particular, proteins of the mitochondrial 50S
ribosomal subunit display high degree of homology to
their bacterial counterparts, which explains why an-
tibiotics targeting the prokaryotic 50S subunit could
also impair mitochondrial translation [45]. Indeed, all
classes of antibiotics listed in Table  1 that act on the
50S subunit (except clindamycin) have been shown
to inhibit not only bacterial, but also mitochondrial
ANTIBIOTICS AND CELLULAR SENESCENCE 1887
BIOCHEMISTRY (Moscow) Vol. 90 No. 12 2025
protein synthesis [46-51]. For example, XL2, a mem-
ber of the oxazolidinone class, suppresses mitochon-
drial translation by binding to the same A-site on the
ribosome as it does in bacterial cells [52].
In addition to targeting the 50S subunit, some an-
tibiotics also act on the mitochondrial 30S ribosomal
subunit, thereby inhibiting mitochondrial translation.
Adverse effects of certain antibiotics appear to de-
pend on structural features of the mitochondrial 30S
subunit. For example, mutations in the mitochondrial
12S rRNA gene (1555A>G and 1494C>T) are associated
with the significantly increased risk of aminoglyco-
side-induced ototoxicity [53, 54]. These mutations are
thought to render the secondary structure of mito-
chondrial 12S rRNA, a component of the 30S subunit,
more similar to the corresponding region of the bac-
terial 16S rRNA, which constitutes a therapeutic tar-
get of aminoglycosides [37, 55, 56].
Doxycycline, a member of the tetracycline class,
induces a “mitonuclear imbalance” characterized by
disruption of the stoichiometric ratio between mito-
chondrial and nuclear genomes that encode electron
transport chain (ETC) proteins [57]. This imbalance
subsequently leads to mitochondrial fragmentation
and impaired respiratory function [57].
In addition to the impairing mitochondrial rep-
lication, fluoroquinolone antibiotics also disrupt mi-
tochondrial protein synthesis and decrease relative
amount of mitochondrial DNA (mtDNA) [58,  59]. Pro-
teomic analyses in the eukaryotic cell line HEK-293
have identified several additional intracellular tar-
gets of fluoroquinolones. Among these is NUDT1, an
enzyme involved in protecting cells from oxidative
stress by hydrolyzing oxidized nucleotides. Treatment
with ciprofloxacin or levofloxacin reduces NUDT1
levels, thereby lowering cellular resistance to the
fluoroquinolone-induced oxidative stress [60]. Sev-
eral mitochondrial proteins were also identified as
targets, including AIFM1 – a regulator of cell death
and a mediator of metabolite transport across the in-
ner mitochondrial membrane. Ciprofloxacin has been
shown to bind AIFM1, resulting in dysfunction of re-
spiratory chain complexes  I and IV. Moreover, both
ciprofloxacin and levofloxacin inhibit IDH2 (isocitrate
dehydrogenase2), further contributing to ETC impair-
ment [60].
Antibiotics that do not target bacterial replication
or translation, such as vancomycin and ceftriaxone
(inhibitors of bacterial cell wall synthesis) also ex-
hibit nonspecific interactions within host tissues. For
example, vancomycin can bind to elastin, a structur-
al protein of the vessel wall, promoting formation of
vancomycin aggregates that exert toxic effects on the
endothelial cells [61]. Ceftriaxone, a cephalosporin
antibiotic, has been shown to interact with Aurora B
kinase, a key regulator of cell cycle and tumor pro-
gression. This unexpected interaction highlights the
potential for exploring anticancer properties of cef-
triaxones [62].
ANTIBIOTICS AND CELLULAR SENESCENCE
Antibiotics used in cancer therapy. As noted
above, several antibiotics are employed as antitumor
agents and, by definition, have the capacity to halt
the cell cycle and induce cellular senescence. These
compounds damage cellular DNA, activate the DNA
damage response, and generate oxidative stress, all
of which contribute to the onset and progression of
cellular senescence.
Anthracyclines form cleavable DNA complexes,
inhibit topoisomerase II activity, and induce oxidative
stress, collectively disrupting both transcription and
DNA replication [63]. They also trigger mitochondrial
dysfunction by inhibiting components of the respira-
tory chain, promoting mitochondrial iron accumula-
tion, and increasing production of reactive oxygen
species (ROS) [64]. Capacity of doxorubicin and other
anthracyclines to induce cellular senescence is well
documented; both their mitochondrial and genotoxic
effects, accompanied by oxidative stress, contribute
to this outcome [65, 66]. Notably, the doxorubicin-in-
duced senescence could proceed through mechanisms
that are either p53-dependent or p53-independent
[67]. Importantly, anthracyclines have been shown to
induce cellular senescence not only in  vitro but also
in animal models [68, 69].
Bleomycin induces the genomic DNA strand
breaks [70] and promotes mitochondrial dysfunction,
both of which contribute to the development of cellu-
lar senescence [71]. It triggers a senescent phenotype
characterized by the increased numbers of SA-β-gal
positive cells, morphological alterations, elevated ly-
sosomal content, and reduced proliferative capacity
in the A549 lung adenocarcinoma cells, as well as in
the primary alveolar epithelial cells isolated from the
rats with bleomycin-induced pulmonary fibrosis [72].
In the lung tissue of these animals, elevated levels
of γH2AX-positive cells, activation of p21, and emer-
gence of SASP were also observed [73]. Furthermore,
two bleomycin derivatives, boanmycin and boningmy-
cin, have likewise been reported to induce cellular
senescence [74, 75].
Dactinomycin (actinomycin  D), an inhibitor of
RNA synthesis in both prokaryotic and eukaryotic
cells, has been shown to induce cellular senescence
[76, 77]. In the human mesenchymal stem cells, dacti-
nomycin treatment leads to the increased SA-β-gal ac-
tivity and SASP development [77]. Senescence induc-
tion has also been observed in the OCI-AML3 acute
myeloid leukemia cells carrying the mutant form of
ZINOVKIN, KONDRATENKO1888
BIOCHEMISTRY (Moscow) Vol. 90 No. 12 2025
NPM1 gene (NPM1c), where it is accompanied by mi-
tochondrial stress, including mitochondrial fragmen-
tation and elevated ROS production [76]. Moreover,
the conditioned medium from the dactinomycin-treat-
ed cells was found to reduce mitochondrial inner
membrane potential (ΔΨm) and increase ROS levels
in the recipient cells [78].
Mitomycin C induces formation of inter- and
intrastrand DNA crosslinks between guanine resi-
dues, thereby inhibiting both DNA replication and
transcription [79]. It also damages mtDNA [80], thus
contributing to mitochondrial dysfunction [81]. In
the human dermal fibroblasts, mitomycin C triggers
cellular senescence, characterized by the increased
SA-β-gal activity, cell-cycle arrest, development of the
senescence-associated secretory phenotype (SASP),
and elevated ROS levels [82]. In  vivo, mitomycin  C has
likewise been shown to induce cellular senescence,
as demonstrated in the rabbit trabeculectomy mod-
el [83].
The promising antitumor ionophore antibiotic sa-
linomycin, which also possesses antibacterial proper-
ties, not only induces DNA damage but also promotes
lysosomal iron accumulation and oxidative stress.
These effects lead to the cell-cycle arrest and devel-
opment of cellular senescence in the MDA-MB-231
breast cancer cells [84, 85]. Interestingly, despite its
pro-senescence activity, salinomycin is also capable
of inducing apoptosis in various human tumor cell
types, including MDA-MB-231 cells [84, 86, 87].
The relatively rare antitumor antibiotic lidamy-
cin also appears to induce cellular senescence. Treat-
ment of the BEL-7402 and MCF-7 cells, with lidamycin
leads to the increased cell size and higher propor-
tion of the SA-β-gal-positive cells [88]. Studies with
the BEL-7402 cells further demonstrate that lidamy-
cin could trigger senescence and mitotic catastrophe
either in parallel or sequentially [89]. Reduction in
the telomerase activity and decreased expression of
EZH2 are thought to play key roles in the lidamy-
cin-induced senescence [89, 90].
Antibacterial antibiotics. Influence of antibiot-
ics not traditionally used in cancer therapy on the
development of cellular senescence remains poorly
understood. Chloramphenicol, an inhibitor of both
bacterial and mitochondrial protein synthesis, has
been shown to suppress the mitomycin C-induced
apoptosis while increasing the p21 expression and
the number of SA-β-gal-positive cells [50]. Similar se-
nescence-promoting effects have been observed with
other protein synthesis inhibitors, including doxycy-
cline, clindamycin, and minocycline [50]. Cephalexin,
a cephalosporin antibiotic, does not induce senes-
cence on its own; however, it enhances senescence
in the cells exposed to ionizing radiation, indicating
potential radiosensitizing properties [91].
It can be hypothesized that antibiotics inducing
mitochondrial dysfunction could, under certain con-
ditions, promote the development of cellular senes-
cence (Fig.  2). Mitochondrial dysfunction is known to
trigger inflammatory responses through the release of
mitochondrial damage-associated molecular patterns
(DAMPs), including mitochondrial DNA, mitochondri-
al RNA, and N-formylmethionine-containing proteins
[92,  93]. Inflammation, in turn, is a well-established
driver of cellular senescence (Fig.  1). Moreover, mito-
chondrial dysfunction is almost invariably accompa-
nied by oxidative stress resulting from the impaired
redox processes [94, 95], which further contributes to
the senescence induction [96] (Figs.  1 and 2).
Antibiotics capable of inducing cellular senescence
include oxazolidinones, which impair mitochondrial
function, cause cell cycle arrest, and increase pro-
portion of the SA-β-gal-positive cells [97, 98]. Similar
to salinomycin, oxazolidinones could simultaneously
trigger both senescence and apoptosis, as demonstrat-
ed in the DU145 prostate cancer cells [98]. A recent
review [99] summarizes evidence supporting poten-
tial use of oxazolidinones and their derivatives as
antitumor agents. In addition, the bacteriostatic vet-
erinary antibiotics amoxicillin and chlortetracycline
have been shown to inhibit cell proliferation [100].
Incubation of glioblastoma cells with the fluoro-
quinolone ciprofloxacin induced CS. When exposure
was limited to 10 days, this senescent state was revers-
ible: after ciprofloxacin was removed from the medi-
um, the cells resumed proliferation after a 2-3-day lag
[101]. In contrast, treatment for 15 days or longer re-
sulted in irreversible senescence. Reversibility of the
ciprofloxacin-induced senescence was shown to de-
pend on RelA (p65), a subunit of the NF-κB complex
[101]. However, it remains unclear how ciprofloxacin
and other fluoroquinolones influence senescence in
non-tumor cell lines or in animal models.
Antibiotics as senolytics. Accumulation of se-
nescent cells is known to impair functions of organs
and tissues, in part due to the chronic inflammation
driven by SASP. The use of senolytics, agents that se-
lectively eliminate senescent cells, has been shown
to restore lost or compromised functions [102]. Se-
nolytics can be broadly divided into two major cate-
gories: the first targets pro-survival pathways in the
senescent cells, thereby promoting their apoptosis;
the second amplifies existing cellular stresses with-
in senescent cells, triggering in addition to apoptosis
alternative forms of cell death such as necrosis or
ferroptosis [102].
In addition to the well-established senolytics
such as combination of quercetin with dasatinib [103,
104], senolytic properties have also been found in
several antibiotics. Although the mechanisms under-
lying the senolytic activity of most antibiotics remain
ANTIBIOTICS AND CELLULAR SENESCENCE 1889
BIOCHEMISTRY (Moscow) Vol. 90 No. 12 2025
Fig. 2. Possible pathways of cell senescence induction by antibiotics. Abbreviations: ↓ΔΨm, decrease in mitochondrial trans-
membrane potential; ↓ATP, decrease in ATP level; ↑Ca
2+
, increase in calcium ion concentration; ROS, reactive oxygen species.
Details are given in the text.
largely unexplored, available evidence indicates that
these compounds fall into the second major catego-
ry of senolytics: they enhance pre-existing stresses
in the senescent cells, primarily through mitochon-
drial disruption and/or interference with autophagy
pathways.
The ionophore antibiotic nigericin has been
shown to exert a multifaceted senolytic effect by
depolarizing both the plasma membrane and the
inner mitochondrial membrane, promoting cytoplas-
mic acidification, and inhibiting autophagy. Together,
these disruptions destabilize cellular homeostasis and
ultimately lead to the death of senescent cells [105].
Notably, targeting any one of these processes individ-
ually was insufficient to produce a senolytic effect.
In the study using the A549 cell-based aging
model induced by alisertib and CFI-400945, addi-
tion of the ionophore salinomycin triggered mito-
chondrial dysfunction and oxidative stress, leading
to elimination of the SA-β-gal positive cells through
PANoptosis – a coordinated activation of pyroptosis,
apoptosis, and necroptosis [106]. Effects of salinomy-
cin resembled those observed upon knockdown of
the SLC25A23 gene encoding a mitochondrial carrier
protein that facilitates Ca
2+
uptake [106].
Senolytic activity has also been identified among
the macrolide antibiotics. Azithromycin and roxithro-
mycin were shown to exert senolytic effects in the
MRC5 and BJ fibroblasts in the bromodeoxyuridine-in-
duced senescence model, with azithromycin demon-
strating greater selectivity toward the senescent cells
[107]. This senolytic action appears to be mediated, at
least in part, by the induction of autophagy, although
azithromycin exhibited a concentration-dependent,
bidirectional influence on mitochondrial respiration.
Azithromycin also displayed senolytic properties in
the “aged” endometrial stromal cells isolated from
the patients with ovarian endometriosis [108]. Inter-
estingly, erythromycin showed no senolytic effect in
one study [107], yet demonstrated senolytic activity
in another, that used a hydrogen peroxide-induced se-
nescence model in the BEAS-2B epithelial cells [109].
Roxithromycin also exhibited senolytic effects in the
WI-38 lung fibroblasts in the bleomycin-induced se-
nescence model [110].
Several antibiotics that inhibit bacterial protein
synthesis have also demonstrated senolytic activity.
The veterinary antibiotic valnemulin, for example,
reduced proportion of the senescent cells in the in-
testinal tissue of mice with experimentally induced
ulcerative colitis [111]. Doxycycline, a member of
the tetracycline class, exhibited senolytic effects
in the mouse embryonic fibroblasts derived from
the Hutchinson–Gilford progeria mice model [112].
ZINOVKIN, KONDRATENKO1890
BIOCHEMISTRY (Moscow) Vol. 90 No. 12 2025
Chloramphenicol was shown to prevent the 5-fluo-
rouracil-induced senescence by activating autophagy
[113]. Interestingly, however, another study reported
that both chloramphenicol and doxycycline increased
the number of SA-β-gal positive cells and elevated the
p21 protein levels, suggesting context-dependent ef-
fects on cellular senescence [50].
CONCLUSION
The ability of antitumor antibiotics to induce
stable cell-cycle arrest and cellular senescence both
in vitro and in vivo has long been recognized and is
supported by numerous experimental studies. A clas-
sic example is the anthracycline doxorubicin, which
induces persistent DNA damage, activates the p53/p21
signaling pathway, and triggers hallmark senescence
phenotypes such as SA-β-gal activity and SASP. The
doxorubicin-induced senescent cells have also been
shown to contribute to the late adverse effects of
chemotherapy [114].
Interestingly, the same or structurally similar
agents can exhibit opposing activities – promoting
senescence under some conditions while acting as
senolytics under others. A notable example is the
antibiotic salinomycin: several studies have shown
that it induces hallmark features of CS in tumor
cells (e.g., increased SA-β-gal activity and elevated
p21), whereas more recent findings demonstrate that
it can also function as a senolytic, triggering PANop-
tosis in the pre-existing senescent cells [84, 106]. This
context- dependent duality underscores complexity of
the antibiotic-mediated stress responses and suggests
potential for the two-stage cancer therapeutic strat-
egies in which the chemotherapy-induced tumor se-
nescent cells are subsequently cleared using senolyt-
ics [106].
Mechanistically, many antibiotics, particularly
bactericidal ones such as fluoroquinolones, amino-
glycosides, and certain β-lactams, exert mitotoxic ef-
fects on eukaryotic cells, leading to mtDNA damage,
impaired mitochondrial energy metabolism, and in-
creased ROS production [115]. These disturbances are
closely linked to activation of the cellular senescence
programs, suggesting that the antibiotic-induced mi-
totoxicity may underlie their pro-senescence side ef-
fects. However, prevalence and biological significance
of these effects in vivo remain poorly understood.
To date, there is virtually no direct evidence demon-
strating causal relationship between the antibiotic
exposure and accumulation of the senescent cells in
human tissues.
These knowledge gaps carry important clinical
implications. Long-term side effects of antibiotic use
(persistent changes in the microbiota, metabolic and
immune shifts, musculoskeletal risks, etc.) have been
well documented in both epidemiological and exper-
imental studies. It is plausible that some of these
lasting consequences may, in part, stem from the
antibiotic-induced cellular senescence in critical cell
populations, including fibroblasts, endothelial cells,
and other mesenchymal-derived cells. This hypothesis
warrants further focused investigation.
Taken together, the evidence suggests that al-
most all antibiotics exhibiting pronounced mitotox-
icity or capacity to induce DNA stress in eukaryotic
cells may, under certain conditions, such as specific
dosages, exposure durations, cellular targets, and mi-
croenvironmental contexts, influence cellular aging
processes. This highlights several important direc-
tions for systematic investigation: (i)  comprehensive
screening of antibiotic molecules for pro-senescence
and senolytic activities across the diverse primary
and tissue-specific cellular models; (ii)  determina-
tion of threshold concentrations and temporal win-
dows at which the pro-senescence effects transition
into overt cytotoxicity; (iii)  in  vivo studies aimed at
detecting and mapping senescent cell accumulation
following clinically relevant antibiotic regimens; and
(iv)  assessment of the contribution of antibiotic-in-
duced cellular senescence to the long-term clinical
outcomes, as well as exploration of whether these
effects could be mitigated using senomorphics or
senolytics.
Convergence of the experimental evidence on mi-
totoxic and pro-senescence effects of multiple antibi-
otics with clinical observations of long-term adverse
outcomes provides a strong rationale for the targeted
investigations into the role of cellular senescence in
antibiotic toxicity. Systematic examination of this is-
sue would not only deepen our understanding of the
fundamental mechanisms underlying antibiotic-asso-
ciated side effects but could also open new strategies
for their prevention and treatment. Such efforts may
ultimately support the design of combination ap-
proaches “antibiotic + senolytics/senomorphics”, when
it is safe and supported by evidence.
Abbreviations
CS cellular senescence
IL interleukin
ROS reactive oxygen species
SA-β-gal senescence-associated beta-galactosi-
dase
SASP senescence-associated secretory phe-
notype
Acknowledgments
The authors would like to thank Evgeniy Sergeevich
Egorov for inspiration, supply of information, and in-
valuable help in the work.
ANTIBIOTICS AND CELLULAR SENESCENCE 1891
BIOCHEMISTRY (Moscow) Vol. 90 No. 12 2025
Contributions
R. A. Zinovkin – concept of the paper and writing the
text. N. D. Kondratenko – writing and editing of the
text.
Funding
The work was financially supported by Russian Sci-
ence Foundation (project no.25-24-00307).
Ethics approval and consent to participate
This work does not contain any studies involving hu-
man and animal subjects.
Conflict of interest
The authors of this work declare that they have
noconflicts of interest.
Open access
This article is licensed under a Creative Commons At-
tribution 4.0 International License, which permits use,
sharing, adaptation, distribution, and reproduction
in any medium or format, as long as you give appro-
priate credit to the original author(s) and the source,
provide a link to the Creative Commons license, and
indicate if changes were made. The images or other
third party material in this article are included in the
article’s Creative Commons license, unless indicated
otherwise in a credit line to the material. If materi-
al is not included in the article’s Creative Commons
license and your intended use is not permitted by
statutory regulation or exceeds the permitted use, you
will need to obtain permission directly from the copy-
right holder. To view a copy of this license, visit http://
creativecommons.org/licenses/by/4.0/.
REFERENCES
1. Cunha, B. A. (2001) Antibiotic side effects, Med. Clin.
North Am., 85, 149-185, https://doi.org/10.1016/s0025-
7125(05)70309-6.
2. Suárez-Rivero, J. M., Pastor-Maldonado, C. J., Povea-
Cabello, S., Álvarez-Córdoba, M., Villalón-García, I.,
Talaverón-Rey, M., Suárez-Carrillo, A., Munuera-
Cabeza, M., and Sánchez-Alcázar, J. A. (2021) Mito-
chondria and antibiotics: for good or for evil? Biomol-
ecules, 11, 1050, https://doi.org/10.3390/biom11071050.
3. Singh, R., Sripada, L., and Singh, R. (2014) Side ef-
fects of antibiotics during bacterial infection: mi-
tochondria, the main target in host cell, Mito-
chondrion, 16, 50-54, https://doi.org/10.1016/j.mito.
2013.10.005.
4. Campisi,J., and d’Adda di Fagagna, F. (2007) Cellular
senescence: when bad things happen to good cells,
Nat. Rev. Mol. Cell Biol., 8, 729-740, https://doi.org/
10.1038/nrm2233.
5. Aliouat-Denis, C.-M., Dendouga, N., Van den
Wyngaert,I., Goehlmann,H., Steller,U., van de Weyer,I.,
van Slycken, N., Andries, L., Kass, S., Luyten, W.,
Janicot,M., and Vialard, J.E. (2005) p53-independent
regulation of p21Waf1/Cip1 expression and senes-
cence by Chk2, Mol. Cancer Res., 3, 627-634, https://
doi.org/10.1158/1541-7786.MCR-05-0121.
6. Mijit,M., Caracciolo,V., Melillo,A., Amicarelli,F., and
Giordano, A. (2020) Role of p53 in the regulation of
cellular senescence, Biomolecules, 10, 420, https://
doi.org/10.3390/biom10030420.
7. Borodkina, A. V., Deryabin, P. I., Giukova, A. A.,
and Nikolsky, N. N. (2018) “Social life” of senescent
cells: what is SASP and why study it? Acta Natu-
rae, 10, 4-14, https://doi.org/10.32607/20758251-2018-
10-1-4-14.
8. Muñoz-Espín, D., and Serrano, M. (2014) Cellular
senescence: from physiology to pathology, Nat. Rev.
Mol. Cell Biol., 15, 482-496, https://doi.org/10.1038/
nrm3823.
9. Campisi, J. (1997) The biology of replicative senes-
cence, Eur. J. Cancer, 33, 703-709, https://doi.org/
10.1016/S0959-8049(96)00058-5.
10. Kumari,R., and Jat,P. (2021) Mechanisms of cellular
senescence: Cell cycle arrest and senescence associ-
ated secretory phenotype, Front. Cell Dev. Biol., 9,
645593, https://doi.org/10.3389/fcell.2021.645593.
11. Di Micco, R., Krizhanovsky, V., Baker, D., and d’Adda
di Fagagna, F. (2021) Cellular senescence in age-
ing: from mechanisms to therapeutic opportunities,
Nat. Rev. Mol. Cell Biol., 22, 75-95, https://doi.org/
10.1038/s41580-020-00314-w.
12. Shmulevich, R., and Krizhanovsky, V. (2021) Cell se-
nescence, DNA damage, and metabolism, Antioxid.
Redox Signal., 34, 324-334, https://doi.org/10.1089/
ars.2020.8043.
13. Petrova, N. V., Velichko, A. K., Razin, S. V., and
Kantidze, O.L. (2016) Small molecule compounds that
induce cellular senescence, Aging Cell, 15, 999-1017,
https://doi.org/10.1111/acel.12518.
14. Hernandez-Segura, A., Nehme, J., and Demaria, M.
(2018) Hallmarks of cellular senescence, Trends
Cell Biol., 28, 436-453, https://doi.org/10.1016/j.
tcb.2018.02.001.
15. Halawa, E.M., Fadel,M., Al-Rabia, M.W., Behairy,A.,
Nouh, N. A., Abdo, M., Olga, R., Fericean, L., Atwa,
A. M., El-Nablaway, M., and Abdeen, A. (2023) An-
tibiotic action and resistance: updated review of
mechanisms, spread, influencing factors, and alter-
native approaches for combating resistance, Front.
Pharmacol., 14, 1305294, https://doi.org/10.3389/
fphar.2023.1305294.
16. Hutchings, M. I., Truman, A. W., and Wilkinson, B.
(2019) Antibiotics: past, present and future, Curr.
Opin. Microbiol., 51, 72-80, https://doi.org/10.1016/
j.mib.2019.10.008.
ZINOVKIN, KONDRATENKO1892
BIOCHEMISTRY (Moscow) Vol. 90 No. 12 2025
17. Kapoor,G., Saigal,S., and Elongavan,A. (2017) Action
and resistance mechanisms of antibiotics: a guide for
clinicians, J. Anaesthesiol. Clin. Pharmacol., 33, 300-
305, https://doi.org/10.4103/joacp.JOACP_349_15.
18. Ekinci, İ. B., Chłodowska, A., and Olejnik, M. (2023)
Ionophore toxicity in animals: a review of clinical
and molecular aspects, Int. J. Mol. Sci., 24, 1696,
https://doi.org/10.3390/ijms24021696.
19. Karamanolis, N. N., Kounatidis, D., Vallianou, N. G.,
Dimitriou,K., Tsaroucha,E., Tsioulos,G., Anastasiou,
I.A., Mavrothalassitis,E., Karampela,I., and Dalama-
ga,M. (2024) Unraveling the anti-cancer mechanisms
of antibiotics: current insights, controversies, and
future perspectives, Antibiotics (Basel), 14, 9, https://
doi.org/10.3390/antibiotics14010009.
20. Riley,A. (2024) Anti-tumor antibiotics: a guide to tar-
geted cancer therapy, J. Canc. Sci. Res., 9, 1-2, https://
doi.org/10.35248/2576-1447.24.9.569.
21. Rao, M.S., Gupta,R., Liguori, M.J., Hu,M., Huang,X.,
Mantena, S. R., Mittelstadt, S. W., Blomme, E. A. G.,
and van Vleet, T. R. (2019) Novel computational ap-
proach to predict off-target interactions for small
molecules, Front. Big Data, 2, 25, https://doi.org/
10.3389/fdata.2019.00025.
22. Patangia, D. V., Anthony Ryan, C., Dempsey, E., Paul
Ross, R., and Stanton, C. (2022) Impact of antibiot-
ics on the human microbiome and consequences
for host health, Microbiologyopen, 11, e1260, https://
doi.org/10.1002/mbo3.1260.
23. Gallagher, J. C., and MacDougall, C. (2022) Anti-
biotics Simplified, 5th Edn, Jones and Bartlett,
Sudbury, MA.
24. Nadanaciva, S., and Will, Y. (2011) Investigating mi-
tochondrial dysfunction to increase drug safety in
the pharmaceutical industry, Curr. Drug Targets, 12,
774-782, https://doi.org/10.2174/138945011795528985.
25. D’Achille, G., and Morroni, G. (2023) Side effects of
antibiotics and perturbations of mitochondria func-
tions, Int. Rev. Cell Mol. Biol., 377, 121-139, https://
doi.org/10.1016/bs.ircmb.2023.03.009.
26. Theodosiou, A. A., Jones, C. E., Read, R. C., and
Bogaert, D. (2023) Microbiotoxicity: antibiotic usage
and its unintended harm to the microbiome, Curr.
Opin. Infect. Dis., 36, 371-378, https://doi.org/10.1097/
QCO.0000000000000945.
27. Miller, M., and Singer, M. (2022) Do antibiotics cause
mitochondrial and immune cell dysfunction? A litera-
ture review, J. Antimicrob. Chemother., 77, 1218-1227,
https://doi.org/10.1093/jac/dkac025.
28. Milosevic, T. V., Vertenoeil, G., Vainchenker, W.,
Tulkens, P. M., Constantinescu, S. N., and Van
Bambeke, F. (2024) Oxazolidinone antibiotics im-
pair ex vivo megakaryocyte differentiation from
hematopoietic progenitor cells and their matura-
tion into platelets, Antimicrob. Agents Chemother.,
68, e0053324, https://doi.org/10.1128/aac.00533-24.
29. Garrabou, G., Soriano, A., López, S., Guallar, J. P.,
Giralt, M., Villarroya, F., Martí nez, J. A., Casade-
mont, J., Cardellach, F., Mensa, J., and Miró , O.
(2007) Reversible inhibition of mitochondrial pro-
tein synthesis during linezolid-related hyperlactate-
mia, Antimicrob. Agents Chemother., 51, 962-967,
https://doi.org/10.1128/AAC.01190-06.
30. Rucker, J. C., Hamilton, S. R., Bardenstein, D., Isada,
C. M., and Lee, M. S. (2006) Linezolid-associated tox-
ic optic neuropathy, Neurology, 66, 595-598, https://
doi.org/10.1212/01.wnl.0000201313.24970.b8.
31. Brandariz-Núñez,D., Hernández-Corredoira,V., Guarc-
Prades, E., and García-Navarro, B. (2019) Optic neu-
ropathy associated with linezolid: systematic review
of cases, Farm. Hosp., 43, 61-65, https://doi.org/10.7399/
fh.11133.
32. Xiao, Y., Xiong, T., Meng, X., Yu, D., Xiao, Z., and
Song, L. (2019) Different influences on mitochondri-
al function, oxidative stress and cytotoxicity of an-
tibiotics on primary human neuron and cell lines,
J. Biochem. Mol. Toxicol., 33, e22277, https://doi.org/
10.1002/jbt.22277.
33. Leiter, L. M., Thatte, H. S., Okafor, C., Marks, P. W.,
Golan, D. E., and Bridges, K. R. (1999) Chloram-
phenicol-induced mitochondrial dysfunction is as-
sociated with decreased transferrin receptor ex-
pression and ferritin synthesis in K562 cells and
is unrelated to IRE-IRP interactions, J. Cell Physi-
ol., 180, 334-344, https://doi.org/10.1002/(SICI)1097-
4652(199909)180:3<334::AID-JCP4>3.0.CO;2-Q.
34. Nyamagoud, S.B., Swamy, A.H.V., Stanly,N., Tom,C.,
Hegadal, V., and Bhavya, D. B. (2025) Chloramphen-
icol-associated aplastic anemia: a review, BLDE
Univ. J. Health Sci., 10, 16-23, https://doi.org/10.4103/
bjhs.bjhs_112_23.
35. Salimi, A., Eybagi, S., Seydi, E., Naserzadeh, P.,
Kazerouni, N. P., and Pourahmad, J. (2016) Toxicity
of macrolide antibiotics on isolated heart mitochon-
dria: a justification for their cardiotoxic adverse ef-
fect, Xenobiotica, 46, 82-93, https://doi.org/10.3109/
00498254.2015.1046975.
36. Woodhead, J. L., Yang, K., Oldach, D., MacLauch-
lin, C., Fernandes, P., Watkins, P. B., Siler, S. Q., and
Howell, B. A. (2019) Analyzing the mechanisms
behind macrolide antibiotic-induced liver injury
using quantitative systems toxicology modeling,
Pharm. Res., 36, 48, https://doi.org/10.1007/s11095-
019-2582-y.
37. Guan, M.-X. (2011) Mitochondrial 12S rRNA muta-
tions associated with aminoglycoside ototoxicity,
Mitochondrion, 11, 237-245, https://doi.org/10.1016/
j.mito.2010.10.006.
38. Chen,Q., Cui,Y., Ding,G., Jia,Z., Zhang,Y., Zhang,A.,
and Huang,S. (2017) PEA3 protects against gentami-
cin nephrotoxicity: role of mitochondrial dysfunction,
Am. J. Transl. Res., 9, 2153-2162.
ANTIBIOTICS AND CELLULAR SENESCENCE 1893
BIOCHEMISTRY (Moscow) Vol. 90 No. 12 2025
39. Morales, A. I., Detaille, D., Prieto, M., Puente, A.,
Briones, E., Arévalo, M., Leverve, X., López-Novoa,
J. M., and El-Mir, M.-Y. (2010) Metformin prevents
experimental gentamicin-induced nephropathy by
a mitochondria-dependent pathway, Kidney Int., 77,
861-869, https://doi.org/10.1038/ki.2010.11.
40. Pouzaud,F., Dutot,M., Martin,C., Debray,M., Warnet,
J.M., and Rat, P. (2006) Age-dependent effects on re-
dox status, oxidative stress, mitochondrial activity
and toxicity induced by fluoroquinolones on prima-
ry cultures of rabbit tendon cells, Comp. Biochem.
Physiol. C Toxicol. Pharmacol., 143, 232-241, https://
doi.org/10.1016/j.cbpc.2006.02.006.
41. Lowes, D.A., Wallace,C., Murphy, M.P., Webster, N.R.,
and Galley, H.F. (2009) The mitochondria targeted an-
tioxidant MitoQ protects against fluoroquinolone-in-
duced oxidative stress and mitochondrial membrane
damage in human Achilles tendon cells, Free Radic. Res.,
43, 323-328, https://doi.org/10.1080/10715760902736275.
42. Li,Q., Peng,S., Sheng,Z., and Wang,Y. (2010) Ofloxa-
cin induces oxidative damage to joint chondrocytes of
juvenile rabbits: excessive production of reactive oxy-
gen species, lipid peroxidation and DNA damage, Eur.
J. Pharmacol., 626, 146-153, https://doi.org/10.1016/
j.ejphar.2009.09.044.
43. Salimiaghdam,N., Singh, L., Schneider, K., Chwa, M.,
Atilano, S.R., Nalbandian,A., Limb, G.A., and Kenney,
M. C. (2022) Effects of fluoroquinolones and tetracy-
clines on mitochondria of human retinal MIO-M1
cells, Exp. Eye Res., 214, 108857, https://doi.org/
10.1016/j.exer.2021.108857.
44. Ge, T.-F., Law, P.Y.P., Wong, H.Y., and Ho, Y.-Y. (2007)
Gatifloxacin affects GLUT1 gene expression and dis-
turbs glucose homeostasis invitro, Eur. J. Pharmacol.,
573, 70-74, https://doi.org/10.1016/j.ejphar.2007.07.038.
45. Koc, E. C., Burkhart, W., Blackburn, K., Moyer, M. B.,
Schlatzer, D. M., Moseley, A., and Spremulli, L. L.
(2001) The large subunit of the mammalian mito-
chondrial ribosome. Analysis of the complement of
ribosomal proteins present, J.Biol. Chem., 276, 43958-
43969, https://doi.org/10.1074/jbc.M106510200.
46. De Vriese, A. S., Van Coster, R., Smet, J., Seneca, S.,
Lovering, A., van Haute, L. L., Vanopdenbosch, L. J.,
Martin, J.-J., Ceuterick-de Groote,C., Vandecasteele,S.,
and Boelaert, J.R. (2006) Linezolid-induced inhibition
of mitochondrial protein synthesis, Clin. Infect. Dis.,
42, 1111-1117, https://doi.org/10.1086/501356.
47. Milosevic, T. V., Payen, V. L., Sonveaux, P., Muccioli,
G. G., Tulkens, P. M., and Van Bambeke, F. (2018)
Mitochondrial alterations (inhibition of mitochon-
drial protein expression, oxidative metabolism, and
ultrastructure) induced by linezolid and tedizolid
at clinically relevant concentrations in cultured hu-
man HL-60 promyelocytes and THP-1 monocytes,
Antimicrob. Agents Chemother., 62, https://doi.org/
10.1128/AAC.01599-17.
48. Marks,J., Young,E., and Hafner,M. (2024) Determin-
ing the off-target activity of antibiotics and novel
translation initiation sites in mitochondria, Biochem.
Chem. Biol., 13, RP103699, https://doi.org/10.7554/
eLife.103699.1.
49. Li, C.-H., Cheng, Y.-W., Liao, P.-L., Yang, Y.-T., and
Kang, J.-J. (2010) Chloramphenicol causes mitochon-
drial stress, decreases ATP biosynthesis, induces ma-
trix metalloproteinase-13 expression, and solid-tu-
mor cell invasion, Toxicol. Sci., 116, 140-150, https://
doi.org/10.1093/toxsci/kfq085.
50. Li, C.-H., Tzeng, S.-L., Cheng, Y.-W., and Kang, J.-J.
(2005) Chloramphenicol-induced mitochondrial
stress increases p21 expression and prevents cell
apoptosis through a p21-dependent pathway, J. Biol.
Chem., 280, 26193-26199, https://doi.org/10.1074/jbc.
M501371200.
51. Denslow, N. D., and O’Brien, T. W. (1978) Antibiot-
ic susceptibility of the peptidyl transferase locus of
bovine mitochondrial ribosomes, Eur. J. Biochem.,
91, 441-448, https://doi.org/10.1111/j.1432-1033.1978.
tb12696.x.
52. Leach, K. L., Swaney, S. M., Colca, J. R., McDonald,
W. G., Blinn, J. R., Thomasco, L. M., Gadwood, R. C.,
Shinabarger, D., Xiong, L., and Mankin, A. S. (2007)
The site of action of oxazolidinone antibiotics in living
bacteria and in human mitochondria, Mol. Cell, 26,
393-402, https://doi.org/10.1016/j.molcel.2007.04.005.
53. Zhao,H., Li,R., Wang,Q., Yan,Q., Deng, J.-H., Han,D.,
Bai, Y., Young, W.-Y., and Guan, M.-X. (2004) Mater-
nally inherited aminoglycoside-induced and nonsyn-
dromic deafness is associated with the novel C1494T
mutation in the mitochondrial 12S rRNA gene in a
large Chinese family, Am. J. Hum. Genet., 74, 139-152,
https://doi.org/10.1086/381133.
54. Prezant, T. R., Agapian, J. V., Bohlman, M. C., Bu, X.,
Oztas, S., Qiu, W. Q., Arnos, K. S., Cortopassi, G. A.,
Jaber, L., Rotter, J. I., Shohat, M., and Fischel-Ghod-
sian, N. (1993) Mitochondrial ribosomal RNA mu-
tation associated with both antibiotic-induced and
non-syndromic deafness, Nat. Genet., 4, 289-294,
https://doi.org/10.1038/ng0793-289.
55. Kokotas,H., Petersen, M.B., and Willems, P.J. (2007)
Mitochondrial deafness, Clin. Genet., 71, 379-391,
https://doi.org/10.1111/j.1399-0004.2007.00800.x.
56. Davies, J., and Davis, B. D. (1968) Misreading of ri-
bonucleic acid code words induced by aminoglyco-
side antibiotics, J.Biol. Chem., 243, 3312-3316, https://
doi.org/10.1016/s0021-9258(18)93308-9.
57. Moullan, N., Mouchiroud, L., Wang, X., Ryu, D.,
Williams, E. G., Mottis, A., Jovaisaite, V., Frochaux,
M.V., Quiros, P.M., Deplancke,B., Houtkooper, R.H.,
and Auwerx,J. (2015) Tetracyclines disturb mitochon-
drial function across eukaryotic models: a call for
caution in biomedical research, Cell Rep., 10, 1681-
1691, https://doi.org/10.1016/j.celrep.2015.02.034.
ZINOVKIN, KONDRATENKO1894
BIOCHEMISTRY (Moscow) Vol. 90 No. 12 2025
58. Hangas, A., Aasumets, K., Kekäläinen, N. J.,
Paloheinä, M., Pohjoismäki, J. L., Gerhold, J. M., and
Goffart,S. (2018) Ciprofloxacin impairs mitochondri-
al DNA replication initiation through inhibition of
topoisomerase 2, Nucleic Acids Res, 46, 9625-9636,
https://doi.org/10.1093/nar/gky793.
59. Jiang, T., Kustermann, S., Wu, X., Zihlmann, C.,
Zhang, M., Mao, Y., Wu, W., and Xie, J. (2023) Mi-
tochondrial dysfunction is underlying fluoroquino-
lone toxicity: an integrated mitochondrial toxicity
assessment, Mol. Cell Toxicol., 19, 333-342, https://
doi.org/10.1007/s13273-022-00263-9.
60. Reinhardt, T., El Harraoui, Y., Rothemann, A., Jauch,
A.T., Müller-Deubert,S., Köllen, M.F., Risch,T., Jacobs,
L. J., Müller,R., Traube, F. R., Docheva, D., Zahler, S.,
Riemer,J., Bach, N.C., and Sieber, S. A. (2025) Chem-
ical proteomics reveals human off-targets of fluoro-
quinolone induced mitochondrial toxicity, Angew.
Chem. Int. Ed., 64, e202421424, https://doi.org/10.1002/
anie.202421424.
61. Hou, P., Wang, S., Shao, Z., Tang, Y., Wang, W.,
Fang, L., Lin, B., Zhu, Y., Xu, R.-H., and Li, J. (2025)
Off-target interactions of vancomycin with vascular
wall involving elastin-induced self-assembly, Anal.
Chem., 97, 7107-7117, https://doi.org/10.1021/acs.anal-
chem.4c06259.
62. Li, X., Li, H., Li, S., Zhu, F., Kim, D. J., Xie, H., Li, Y.,
Nadas, J., Oi, N., Zykova, T. A., Yu, D. H., Lee, M.-H.,
Kim, M.O., Wang,L., Ma,W., Lubet, R.A., Bode, A.M.,
Dong, Z., and Dong, Z. (2012) Ceftriaxone, an FDA-
approved cephalosporin antibiotic, suppresses lung
cancer growth by targeting Aurora B, Carcinogenesis,
33, 2548-2557, https://doi.org/10.1093/carcin/bgs283.
63. McGowan, J.V., Chung,R., Maulik, A., Piotrowska, I.,
Walker, J. M., and Yellon, D. M. (2017) Anthracy-
cline chemotherapy and cardiotoxicity, Cardiovasc.
Drugs Ther., 31, 63-75, https://doi.org/10.1007/s10557-
016-6711-0.
64. Wallace, K.B., Sardão, V. A., and Oliveira, P. J. (2020)
Mitochondrial determinants of doxorubicin-induced
cardiomyopathy, Circ. Res., 126, 926-941, https://
doi.org/10.1161/CIRCRESAHA.119.314681.
65. Cappetta, D., De Angelis, A., Sapio, L., Prezioso, L.,
Illiano,M., Quaini,F., Rossi,F., Berrino,L., Naviglio,S.,
and Urbanek, K. (2017) Oxidative stress and cellu-
lar response to doxorubicin: a common factor in
the complex milieu of anthracycline cardiotoxici-
ty, Oxid. Med. Cell Longev., 2017, 1521020, https://
doi.org/10.1155/2017/1521020.
66. Chelombitko, M.A., Morgunova, G.V., Strochkova, N.Y.,
Zinovkin, R.A., Pavlyuchenkova, A.N., Kondratenko,
N.D., and Lyamzaev, K.G. (2023) Comparative analy-
sis of cell senescence induced by the chemotherapeu-
tic agents doxorubicin, cisplatin and arsenic trioxide
in human myoblasts MB135, Adv. Gerontol., 13, 16-25,
https://doi.org/10.1134/s2079057024600010.
67. Bientinesi, E., Lulli, M., Becatti, M., Ristori, S.,
Margheri, F., and Monti, D. (2022) Doxorubicin-
induced senescence in normal fibroblasts promotes
in vitro tumour cell growth and invasiveness: the role
of quercetin in modulating these processes, Mech.
Ageing Dev., 206, 111689, https://doi.org/10.1016/
j.mad.2022.111689.
68. Sun,T., Zhang, L., Feng,J., Bao, L., Wang,J., Song,Z.,
Mao,Z., Li,J., and Hu,Z. (2022) Characterization of cel-
lular senescence in doxorubicin-induced aging mice,
Exp. Gerontol., 163, 111800, https://doi.org/10.1016/
j.exger.2022.111800.
69. Clayton, Z. S., Hutton, D. A., Mahoney, S. A., and
Seals, D. R. (2021) Anthracycline chemotherapy-
mediated vascular dysfunction as a model of accel-
erated vascular aging, Aging Cancer, 2, 45-69, https://
doi.org/10.1002/aac2.12033.
70. Hecht, S. M. (2000) Bleomycin: new perspectives on
the mechanism of action, J. Nat. Prod., 63, 158-168,
https://doi.org/10.1021/np990549f.
71. Yao, Y.-X., Lu,X., Li,Z., Liao, H.-Y., Liu, Z.-B., Zhao,H.,
Wang, H., Xu, D.-X., and Tan, Z.-X. (2025) Mitochon-
drial dysfunction-associated cellular senescence is
partially involved in bleomycin-induced pulmonary
fibrosis in mice, Toxicol. Appl. Pharmacol., 504,
117524, https://doi.org/10.1016/j.taap.2025.117524.
72. Aoshiba, K., Tsuji, T., and Nagai, A. (2003) Bleomy-
cin induces cellular senescence in alveolar epithe-
lial cells, Eur. Respir. J., 22, 436-443, https://doi.org/
10.1183/09031936.03.00011903.
73. Aoshiba, K., Tsuji, T., Kameyama, S., Itoh, M.,
Semba, S., Yamaguchi, K., and Nakamura, H. (2013)
Senescence-associated secretory phenotype in a
mouse model of bleomycin-induced lung injury, Exp.
Toxicol. Pathol., 65, 1053-1062, https://doi.org/10.1016/
j.etp.2013.04.001.
74. Chen, P., Guo,H., Chen,J., and Fu,Y. (2016) The che-
motherapeutic drug boanmycin induces cell senes-
cence and senescence-associated secretory phenotype
factors, thus acquiring the potential to remodel the
tumor microenvironment, Anticancer Drugs, 27, 84-
88, https://doi.org/10.1097/cad.0000000000000304.
75. Gao, N., Shang, B., Zhang, X., Shen, C., Xu, R., Xu, H.,
Chen, R., and He, Q. (2011) Potent antitumor actions
of the new antibiotic boningmycin through induc-
tion of apoptosis and cellular senescence, Antican-
cer Drugs, 22, 166-175, https://doi.org/10.1097/cad.
0b013e3283409bee.
76. Wu, H.-C., Rérolle, D., Berthier, C., Hleihel, R.,
Sakamoto,T., Quentin,S., Benhenda,S., Morganti,C.,
Wu, C., Conte, L., Rimsky, S., Sebert, M., Clappier, E.,
Souquere, S., Gachet, S., Soulier, J., Durand, S.,
Trowbridge, J. J., et al. (2021) Actinomycin D targets
NPM1c-primed mitochondria to restore PML-driven
senescence in AML therapy, Cancer Discov., 11, 3198-
3213, https://doi.org/10.1158/2159-8290.CD-21-0177.
ANTIBIOTICS AND CELLULAR SENESCENCE 1895
BIOCHEMISTRY (Moscow) Vol. 90 No. 12 2025
77. Minieri,V., Saviozzi,S., Gambarotta,G., Lo Iacono,M.,
Accomasso, L., Cibrario Rocchietti, E., Gallina, C.,
Turinetto, V., and Giachino, C. (2015) Persistent DNA
damage-induced premature senescence alters the
functional features of human bone marrow mes-
enchymal stem cells, J. Cell. Mol. Med., 19, 734-743,
https://doi.org/10.1111/jcmm.12387.
78. Jin, C., Wu, S., Lu, X., Liu, Q., Zhang, L., Yang, J.,
Xi, Q., and Cai, Y. (2012) Conditioned medium from
actinomycin D-treated apoptotic cells induces mi-
tochondria-dependent apoptosis in bystander cells,
Toxicol. Lett., 211, 45-53, https://doi.org/10.1016/
j.toxlet.2012.02.020.
79. Bizanek, R., McGuinness, B. F., Nakanishi, K., and
Tomasz, M. (1992) Isolation and structure of an in-
trastrand cross-link adduct of mitomycin C and DNA,
Biochemistry, 31, 3084-3091, https://doi.org/10.1021/
bi00127a008.
80. Pritsos, C.A., Briggs, L.A., and Gustafson, D.L. (1997)
A new cellular target for mitomycin C: a case for mi-
tochondrial DNA, Oncol. Res., 9, 333-337.
81. Yan, C., Kong, D., Ge, D., Zhang, Y., Zhang, X., Su, C.,
and Cao, X. (2015) Mitomycin C induces apoptosis
in rheumatoid arthritis fibroblast-like synoviocytes
via a mitochondrial-mediated pathway, Cell Physi-
ol. Biochem., 35, 1125-1136, https://doi.org/10.1159/
000373938.
82. Alili, L., Diekmann, J., Giesen, M., Holtkötter, O., and
Brenneisen, P. (2014) A drug-induced accelerated se-
nescence (DIAS) is a possibility to study aging in time
lapse, Age (Dordr), 36, 9658, https://doi.org/10.1007/
s11357-014-9658-8.
83. Lin, L.-T., Chen, J.-T., Lu, D.-W., Tai, M.-C., Liang, C.-M.,
Chen, C.-L., Pao, S.-I., Hsu, C.-K., and Chen, Y.-H. (2020)
Antifibrotic role of low-dose mitomycin-c-induced
cellular senescence in trabeculectomy models, PLoS
One, 15, e0234706, https://doi.org/10.1371/journal.
pone.0234706.
84. Al Dhaheri, Y., Attoub, S., Arafat, K., Abuqamar, S.,
Eid,A., Al Faresi,N., and Iratni,R. (2013) Salinomycin
induces apoptosis and senescence in breast cancer:
upregulation of p21, downregulation of survivin and
histone H3 and H4 hyperacetylation, Biochim. Bio-
phys. Acta, 1830, 3121-3135, https://doi.org/10.1016/
j.bbagen.2013.01.010.
85. Miyazaki,Y., Shibuya,M., Sugawara,H., Kawaguchi,O.,
and Hirsoe, C. (1974) Salinomycin, a new poly-
ether antibiotic, J.Antibiot. (Tokyo), 27, 814-821,
https://doi.org/10.7164/antibiotics.27.814.
86. Huczynski,A. (2012) Salinomycin: a new cancer drug
candidate, Chem. Biol. Drug Des., 79, 235-238, https://
doi.org/10.1111/j.1747-0285.2011.01287.x.
87. Managò, A., Leanza, L., Carraretto, L., Sassi, N.,
Grancara, S., Quintana-Cabrera, R., Trimarco, V.,
Toninello,A., Scorrano,L., Trentin,L., Semenzato,G.,
Gulbins,E., Zoratti,M., and Szabò, I. (2015) Early ef-
fects of the antineoplastic agent salinomycin on mi-
tochondrial function, Cell Death Dis., 6, e1930, https://
doi.org/10.1038/cddis.2015.263.
88. He, Q. Y., Liang, Y. Y., Wang, D. S., and Li, D. D.
(2002) Characteristics of mitotic cell death induced
by enediyne antibiotic lidamycin in human epithe-
lial tumor cells, Int. J. Oncol., 20, 261-266, https://
doi.org/10.3892/ijo.20.2.261.
89. Gao, R.-J., Liang, Y.-X., Li, D.-D., Zhang, H.-Y., and
Zhen, Y.-S. (2007) Effect of lidamycin on telomerase
activity in human hepatoma BEL-7402 cells, Biomed.
Environ. Sci., 20, 189-197.
90. Sha, M.-Q., Zhao, X.-L., Li, L., Li, L.-H., Li, Y., Dong,
T.-G., Niu, W.-X., Jia, L.-J., Shao, R.-G., Zhen, Y.-S.,
and Wang, Z. (2016) EZH2 mediates lidamycin-in-
duced cellular senescence through regulating
p21 expression in human colon cancer cells, Cell
Death Dis., 7, e2486, https://doi.org/10.1038/cddis.
2016.383.
91. Labay, E., Mauceri, H. J., Efimova, E. V., Flor, A. C.,
Sutton, H. G., Kron, S. J., and Weichselbaum, R. R.
(2016) Repurposing cephalosporin antibiotics as
pro-senescent radiosensitizers, Oncotarget, 7, 33919-
33933, https://doi.org/10.18632/oncotarget.8984.
92. Lyamzaev, K. G., Zinovkin, R. A., and Chernyak,
B. V. (2022) Extrusion of mitochondria: Garbage
clearance or cell-cell communication signals? J. Cell
Physiol., 237, 2345-2356, https://doi.org/10.1002/
jcp.30711.
93. Zinovkin, R. A., and Zamyatnin, A. A. (2019) Mito-
chondria-targeted drugs, Curr. Mol. Pharmacol., 12,
202-214, https://doi.org/10.2174/1874467212666181127
151059.
94. Zinovkin, R. A., Romaschenko, V. P., Galkin, I. I.,
Zakharova, V. V., Pletjushkina, O. Y., Chernyak, B. V.,
and Popova, E. N. (2014) Role of mitochondrial re-
active oxygen species in age-related inflammatory
activation of endothelium, Aging, 6, 661-674, https://
doi.org/10.18632/aging.100685.
95. Zorov, D. B., Juhaszova, M., and Sollott, S. J. (2014)
Mitochondrial reactive oxygen species (ROS) and
ROS-induced ROS release, Physiol. Rev., 94, 909-950,
https://doi.org/10.1152/physrev.00026.2013.
96. Martini, H., and Passos, J. F. (2023) Cellular senes-
cence: all roads lead to mitochondria, FEBS J., 290,
1186-1202, https://doi.org/10.1111/febs.16361.
97. Nagiec, E.E., Wu,L., Swaney, S.M., Chosay, J.G., Ross,
D. E., Brieland, J. K., and Leach, K. L. (2005) Oxaz-
olidinones inhibit cellular proliferation via inhibi-
tion of mitochondrial protein synthesis, Antimicrob.
Agents Chemother., 49, 3896-3902, https://doi.org/
10.1128/AAC.49.9.3896-3902.2005.
98. Naresh, A., Venkateswara Rao, M., Kotapalli, S. S.,
Ummanni, R., and Venkateswara Rao, B. (2014) Ox-
azolidinone derivatives: cytoxazone-linezolid hybrids
induces apoptosis and senescence in DU145 prostate
ZINOVKIN, KONDRATENKO1896
BIOCHEMISTRY (Moscow) Vol. 90 No. 12 2025
cancer cells, Eur. J. Med. Chem., 80, 295-307, https://
doi.org/10.1016/j.ejmech.2014.04.062.
99. Ramalho, E.A.V.F., Pitta, M.G.R., Neto, H.B.S., and
Pitta, I. R. (2020) Does the oxazolidinone derivatives
constitute a functional approach for cancer therapy?
Clin. Cancer Drugs, 7, 95-106, https://doi.org/10.2174/
2212697x07999200807210936.
100. Kim, H. Y., Kim, K.-T., and Kim, S. D. (2012) Bio-
chemical effects of veterinary antibiotics on prolif-
eration and cell cycle arrest of human HEK293 cells,
Bull. Environ. Contam. Toxicol., 89, 234-239, https://
doi.org/10.1007/s00128-012-0695-7.
101. Salunkhe,S., Mishra, S.V., Nair,J., Shah,S., Gardi,N.,
Thorat,R., Sarkar,D., Rajendra,J., Kaur,E., and Dutt,S.
(2021) Nuclear localization of p65 reverses thera-
py-induced senescence, J. Cell Sci., 134, jcs253203,
https://doi.org/10.1242/jcs.253203.
102. Rad, A. N., and Grillari, J. (2024) Current senolyt-
ics: Mode of action, efficacy and limitations, and
their future, Mech. Ageing Dev., 217, 111888, https://
doi.org/10.1016/j.mad.2023.111888.
103. Novais, E.J., Tran, V.A., Johnston, S.N., Darris, K.R.,
Roupas, A.J., Sessions, G.A., Shapiro, I.M., Diekman,
B. O., and Risbud, M. V. (2021) Long-term treatment
with senolytic drugs Dasatinib and Quercetin ame-
liorates age-dependent intervertebral disc degen-
eration in mice, Nat. Commun., 12, 5213, https://
doi.org/10.1038/s41467-021-25453-2.
104. Kirkland, J. L., and Tchkonia, T. (2020) Senolytic
drugs: from discovery to translation, J.Intern. Med.,
288, 518-536, https://doi.org/10.1111/joim.13141.
105. Deryabin, P. I., Shatrova, A. N., and Borodkina, A. V.
(2022) Targeting multiple homeostasis-maintaining
systems by ionophore nigericin is a novel approach
for senolysis, Int. J. Mol. Sci., 23, 14251, https://
doi.org/10.3390/ijms232214251.
106. Casagrande Raffi, G., Chen, J., Feng, X., Chen, Z.,
Lieftink, C., Deng, S., Mo, J., Zeng, C., Steur, M.,
Wang,J., Bleijerveld, O.B., Hoekman,L., et al. (2024)
An antibiotic that mediates immune destruction of
senescent cancer cells, Proc. Natl. Acad. Sci. USA, 121,
e2417724121, https://doi.org/10.1073/pnas.2417724121.
107. Ozsvari,B., Nuttall, J.R., Sotgia, F., and Lisanti, M.P.
(2018) Azithromycin and Roxithromycin define a new
family of “senolytic” drugs that target senescent hu-
man fibroblasts, Aging (Albany NY), 10, 3294-3307,
https://doi.org/10.18632/aging.101633.
108. Sonehara, R., Nakamura, T., Takeda, T., Kaseki, S.,
Seki, T., Tanaka, H., Yabuki, A., Miyake, N.,
Muraoka, A., Osuka, S., Iwase, A., and Kajiyama, H.
(2025) A novel senotherapeutic strategy with azith-
romycin for preventing endometriosis progression,
Reprod. Biol. Endocrinol., 23, 47, https://doi.org/
10.1186/s12958-025-01381-4.
109. Xiaofei, Y., Tingting, L., Xuan, W., and Zhiyi, H.
(2022) Erythromycin attenuates oxidative stress-in-
duced cellular senescence via the PI3K-mTOR signal-
ing pathway in chronic obstructive pulmonary dis-
ease, Front. Pharmacol., 13, 1043474, https://doi.org/
10.3389/fphar.2022.1043474.
110. Zhang, X., Dong, Y., Li, W.-C., Tang, B.-X., Li, J., and
Zang, Y. (2021) Roxithromycin attenuates bleomy-
cin-induced pulmonary fibrosis by targeting se-
nescent cells, Acta Pharmacol. Sin., 42, 2058-2068,
https://doi.org/10.1038/s41401-021-00618-3.
111. Wang, Z., Lian, W., Chen, C., Dai, Q., Liu, Z., Liu, J.,
Zhang, Y., Zhou, M., and Wang, X. (2024) Network
pharmacology and experimental verification reveal-
ing valnemulin alleviates DSS-induced ulcerative
colitis by inhibiting intestinal senescence, Int. Immu-
nopharmacol., 141, 112810, https://doi.org/10.1016/
j.intimp.2024.112810.
112. Wang, M., Zhang, J., Qiu, J., Ma, X., Xu, C., Wu, Q.,
Xing, S., Chen, X., and Liu, B. (2024) Doxycycline
decelerates aging in progeria mice, Aging Cell, 23,
e14188, https://doi.org/10.1111/acel.14188.
113. Bai, S.-R., Zhao, Q., Jia, H.-J., He, F., and Wang, X.-B.
(2024) Chloramphenicol alleviates 5-fluorouracil-in-
duced cellular senescence through activation of au-
tophagy, Can. J. Physiol. Pharmacol., 102, 661-671,
https://doi.org/10.1139/cjpp-2023-0432.
114. Karabicici, M., Alptekin, S., Fırtına Karagonlar, Z.,
and Erdal,E. (2021) Doxorubicin-induced senescence
promotes stemness and tumorigenicity in EpCAM-/
CD133- nonstem cell population in hepatocellular car-
cinoma cell line, HuH-7, Mol. Oncol., 15, 2185-2202,
https://doi.org/10.1002/1878-0261.12916.
115. Kalghatgi, S., Spina, C. S., Costello, J. C., Liesa, M.,
Morones-Ramirez, J. R., Slomovic, S., Molina, A.,
Shirihai, O.S., and Collins, J.J. (2013) Bactericidal an-
tibiotics induce mitochondrial dysfunction and oxida-
tive damage in mammalian cells, Sci. Transl. Med., 5,
192ra85, https://doi.org/10.1126/scitranslmed.3006055.
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.