- Review
- Open access
- Published:
Regulation of cellular senescence in tumor progression and therapeutic targeting: mechanisms and pathways
Molecular Cancer volume 24, Article number: 106 (2025)
Abstract
Cellular senescence, a stable state of cell cycle arrest induced by various stressors or genomic damage, is recognized as a hallmark of cancer. It exerts a context-dependent dual role in cancer initiation and progression, functioning as a tumor suppressor and promoter. The complexity of senescence in cancer arises from its mechanistic diversity, potential reversibility, and heterogeneity. A key mediator of these effects is the senescence-associated secretory phenotype (SASP), a repertoire of bioactive molecules that influence tumor microenvironment (TME) remodeling, modulate cancer cell behavior, and contribute to therapeutic resistance. Given its intricate role in cancer biology, senescence presents both challenges and opportunities for therapeutic intervention. Strategies targeting senescence pathways, including senescence-inducing therapies and senolytic approaches, offer promising avenues for cancer treatment. This review provides a comprehensive analysis of the regulatory mechanisms governing cellular senescence in tumors. We also discuss emerging strategies to modulate senescence, highlighting novel therapeutic opportunities. A deeper understanding of these processes is essential for developing precision therapies and improving clinical outcomes.
Introduction
Cellular senescence is a stable state of cell cycle arrest, characterized by distinct morphological changes, altered metabolism, gene expression patterns, and a secretory phenotype known as the senescence-associated secretory phenotype (SASP). Senescence can be triggered not only by intrinsic mechanisms such as the cellular division limit (replicative senescence), but also by extrinsic factors like culture conditions (premature senescence) [1]. The impact of cellular senescence on the human body extends across various physiological processes, including embryonic development, wound healing, tumor suppression, and immune regulation [2]. Furthermore, senescence is implicated in the development of numerous diseases, such as cancer, cardiovascular disease and neurodegenerative disorders [3,4,5].
In recent decades, cellular senescence has emerged as a focal point in cancer research. Initially, it was discovered that p53-mediated senescence serves as a critical natural barrier against tumorigenesis, as p53 activation inhibits abnormal cell proliferation [6]. As research progresses, more mechanisms of tumor cell senescence have been discovered, including telomere shortening, oxidative stress, and oncogene activation. These mechanisms are precisely regulated through complex and overlapping signaling pathways.
This article aims to review the molecular mechanisms of cellular senescence in tumor progression and provide an overview of senescence-based therapies for tumors. By gaining a comprehensive understanding of the dual role of cellular senescence in tumors, a more rational approach can be developed to enhance therapeutic interventions (such as promoting senescence, eliminating senescence, and combining both therapies).
Milestones in the history of cellular senescence research
In 1961, Hayflick and Moorhead discovered that human fibroblasts cultured in vitro do not divide indefinitely but instead enter an irreversible state of proliferative arrest, a phenomenon now known as the Hayflick limit [7] (Fig. 1). Since then, scientists have referred to this phenomenon as cellular aging or cellular senescence. It is worth noting that the terms "cellular aging" and "cellular senescence" are not interchangeable. "Aging" refers to the gradual decline in bodily functions as an organism ages and accumulates damage. In contrast, "cellular senescence" specifically refers to a state in which cells permanently stop dividing in response to stress or damage, a process that can occur at any point during an organism's life [8]. Therefore, "cellular senescence" is a more precise term. In 1990, Calvin B. Harley demonstrated that the quantity and length of telomeric DNA in human fibroblasts decrease with cell division, promoting cellular senescence and elucidating the Hayflick limit [9]. In 1995, Gerardo Dimri and colleagues introduced senescence-associated β-galactosidase (SA-β-gal), a biomarker that identifies senescent cells [10]. In the same year, a study demonstrated that oxidative stress-induced DNA damage significantly contributes to cellular senescence [11]. In 1997, Manuel Serrano's research indicated that activation of the oncogene RAS can induce senescence in rodent cells [12]. Until 2008, oncogene-induced senescence associated tumor barrier functions were systematically reviewed and published in Science [13, 14]. In the same year, the concept of SASP was expanded, leading to a broader understanding of cellular senescence. It was no longer viewed as a passive state but one where senescent cells actively secrete factors that influence their surrounding tissue environment [15].As the role of cellular senescence in age-related diseases was gradually uncovered, it was included as one of the Hallmarks of Aging in 2013 [16]. In 2015, the team of Mayo Clinic and Scripps Research first found that the combination of dasatinib and quercetin selectively clear senescent cells [17]. A 2016 study reported that a subset of atypical cancerous cells, which strongly expressed p21, exhibited proliferative features after senescence, suggesting that cellular senescence is reversible [18]. In 2018, by studying the genetic lineage tracing of the fate of senescent cells in vivo, it was found that some senescent cells are not eliminated during mouse embryonic development and may re-enter the cell cycle after birth, thereby confirming the reversibility of senescence during embryonic development [19]. In 2019, summarized the drugs that can inhibit SASP such as rapamycin named senostatics, helping to block the adverse effects of senescent cells [20]. In 2022, the importance of senescent cells in cancer development was further recognized and incorporated into the latest Hallmarks of Cancer [21].
Key milestones in the study of cellular senescence. This timeline highlights significant discoveries in cellular senescence research from 1961 to present. In 1961, the Hayflick Limit was established, demonstrating that human fibroblasts have a finite capacity for cell division, marking the discovery of replicative senescence. By 1990, telomere shortening was identified as a key mechanism underlying cellular senescence, providing an explanation for the Hayflick Limit. In 1995, senescence-associated β-galactosidase (SA-β-gal) was identified as a biomarker for cellular senescence, alongside findings that oxidative stress can induce cellular senescence. In 1997, activation of the RAS oncogene was shown to trigger cellular senescence. By 2008, the mechanisms underlying oncogene-induced senescence associated tumor barrier function were summarized. In the same year the focus of senescence research shifted from the phenomenon and mechanisms to exploring the functions and applications, spurred by the concept of the senescence-associated secretory phenotype (SASP). In 2013, cellular senescence was recognized as one of the Hallmarks of Aging. In 2015, the first senolytic agents, dasatinib and quercetin, were identified. In 2016, The potential reversibility of cellular senescence was found in cancer cells. In 2019, the concept of senostatics emerged, aimed at mitigating the adverse effects of senescent cells. Finally, in 2022, the role of senescent cells in cancer development was integrated into the latest Hallmarks of Cancer
Inducing factors of cellular senescence
The occurrence of cellular senescence is driven by multiple factors, which can be categorized into environmental and host factors based on their origin.
Environmental factors
A variety of environmental factors, including biological, physical, chemical, and social factors, have been implicated in the acceleration of cellular senescence, possibly through direct cellular damage or altered cellular metabolism (Fig. 2).
Inducing factors of cellular senescence. Cellular senescence is driven by multiple factors, categorized as environmental and host factors based on their origin. Environmental factors include biological, physical, chemical, and social factors. Biological factors (1) include bacteria, viruses, and fungi. Physical factors (2) encompass mechanical stress, radiation, temperature, etc. Chemical factors (3) involve exposure to inorganic and organic compounds. Social factors (4) are reflected in the poor social relationship and the psychological stress. Host factors include telomere shortening (5), oxidative stress (6), oncogene activation (7) and SASP (8). Together, these factors orchestrate the complex mechanisms leading to cellular senescence
Biological factors
Research studies have demonstrated that many pathogens, including bacteria, viruses, and fungi, can induce cellular senescence [22]. In vitro, the accumulation of bacterial lipopolysaccharide (LPS) in the cytoplasm activates the pattern recognition receptor (PRR) caspase-4, which subsequently triggers the p53-p21 and p16INK4a-CDK4/6 pathways, leading to cellular senescence [23]. Studies on coronavirus disease 2019 (COVID-19) demonstrate that SARS-CoV-2 degrades the DNA damage-responsive kinase CHK1 in host cells, reducing DNA repair and upregulating p16INK4a expression. This process induces cellular senescence, which in turn triggers the secretion of pro-inflammatory cytokines, extracellular matrix remodeling factors, and pro-coagulatory mediators by senescent cells. These events contribute to a cytokine storm, which accelerates senescence in neighboring cells and leads to surrounding tissue damage [24,25,26]. Moreover, studies have shown that HIV can inhibit the PI3K/ATM pathway, leading to DNA damage and telomere attrition, which accelerates the senescence of CD4 + T cells, thus exacerbating immune system dysfunction in HIV infection [27]. In candidiasis, the upregulation of circular RNA circHIPK3 competitively binds miR-148b-3p, leading to increased expression of DNMT1/3a. This enhances methylation of the anti-aging gene Klotho promoter, reducing Klotho expression and ultimately promoting cellular senescence, which exacerbates the inflammatory response to infection [28].
Physical factors
Physical factors include mechanical stress, radiation, temperature, atmospheric pressure, electric currents, and noise (Fig. 2). For example, shear stress from surgical incisions can suppress the transcription of sirtuin 1 (SIRT1) in liver sinusoidal endothelial cells (LSECs), promoting the activity of proteins like p53, p21, and p16INK4a, thereby accelerating LSEC senescence and impairing liver regeneration following partial hepatectomy [29]. Ultraviolet radiation (UVA and UVB), combined with urban particulate matter (UPM), can lead to mitochondrial dysfunction, elevation of reactive oxygen species (ROS), DNA damage, and can contribute to skin aging [30]. Heat exposure accelerates cellular metabolism, increasing ROS production and disrupting DNA replication. This activates the cyclic GMP-AMP synthase (cGAS) and stimulator of interferon genes (STING) pathways, promoting lung tissue aging and fibrosis [31].
Chemical factors
Chemical factors encompass both inorganic and organic compounds. Inorganic factors such as atmospheric particulate matter (PM) have long been recognized as a threat to human health. Research has shown that PM exposure can induce replicative senescence (RS), which is associated with the downregulation of telomerase reverse transcriptase (TERT) and proliferating cell nuclear antigen (PCNA) in human lung epithelial cells [32]. PM exposure also inhibits the SIRT1/PGC-1α/SIRT3 signaling pathway, impairing the antioxidant system, which leads to ROS accumulation and promotes cellular senescence [33]. Zinc oxide (ZnO) nanoparticles (NPs), frequently used in cosmetics, agriculture, biosensors, and drug delivery, have been shown to induce mesenchymal stem cell senescence through oxidative stress and DNA damage [34].
Cannabidiol (CBD), an organic compound used in the treatment of childhood epilepsy, inhibits the expression of key proteins involved in DNA replication, including E2F1, E2F2, cyclin D3, and CDK2, resulting in cell cycle arrest. Additionally, CBD suppresses key enzymes in DNA repair pathways, ultimately activating a p53-dependent senescence pathway [35]. Additionally, certain chemotherapy drugs have been shown to induce senescence (therapy-induced senescence, TIS) in tumor cells. For example, doxorubicin, used in treating chronic myeloid leukemia (CML) in the K562 cell model, upregulates miR-375 and induces autophagy, leading to senescence, independent of the p16INK4a and p53 senescence pathways [36]. Prolonged exposure to bleomycin induces oxidative stress and DNA double-strand breaks in alveolar epithelial cells, inhibiting Rad51 expression, impairing DNA repair, and triggering senescence, which leads to lung toxicity [37].
Social factors
Social factors refer to the functional or prescriptive effects of a person's social relationships, such as emotional support, instrumental help, and information. Systematic reviews indicate that poor social relationships, including negative perceptions of one's neighborhood, contribute to telomere shortening and accelerate physical aging [38]. Additionally, psychological stress has been identified as another factor that induces telomere shortening [39, 40]. For instance, loneliness and emotional distress can increase biological aging by 1.65 years, exceeding the influence of factors such as biological sex, living area, marital status, or smoking [41]. These findings underscore the importance of the biopsychosocial model in the etiology of cellular senescence.
Host factors
In addition to external environmental factors, cellular senescence is regulated by intrinsic stressors and damaging elements, collectively referred to as host factors in this paper. Specifically, host factors include telomere shortening, oxidative stress, oncogene activation, and SASP (Fig. 2).
Telomere shortening
The discovery of telomeres dates back to 1978, when Elizabeth Blackburn first identified a repetitive sequence of nucleotides at the ends of chromosomes in the protozoan Tetrahymena, marking the beginning of telomere research [42]. Telomeres are specialized DNA–protein complexes composed of hexameric TTAGGG nucleotide repeats and associated proteins in mammals, protecting chromosomal integrity [7, 43]. Telomere length is regulated by the activity of telomerase and shelterin complexes.
Telomerase is an enzyme complex consisting of TERT, telomerase RNA component (TERC), and associated cofactors. Telomerase synthesizes new DNA sequences to elongate telomeres, which counteracts the natural shortening of telomeres that occurs during cell division. The tumor suppressor liver kinase B1 (LKB1) enhances the transcription of Sp1, which inhibits TERT expression, leading to telomere shortening, promoting senescence in tumor cells, and inhibiting tumor growth [44].
The shelterin complex consists of six subunits: TRF1, TRF2, TIN2, Rap1, TPP1, and POT1 [45]. It binds to telomeric DNA associated with nucleosomes and inhibits the recognition of cellular DNA repair machinery. Studies have shown that external stimuli, such as radiation, oxidative stress, or bleomycin, can induce ubiquitination of telomere protection protein 1 (TPP1) by the E3 ubiquitin ligase FBW7, leading to TPP1 degradation and triggering telomere uncapping and the DNA damage response (DDR) [46]. Additionally, shelterin senses telomere length, regulates telomerase activity, and recruits telomerase to the telomere, coordinating the conversion of newly synthesized telomeric single-stranded DNA into double-stranded DNA [47].
Oxidative stress
ROS are reactive oxygen-containing molecules, including superoxide (O₂⁻), hydrogen peroxide (H₂O₂), singlet oxygen (1O₂), and hydroxyl radicals (·OH) [48]. Major sources of ROS include mitochondria, NADPH oxidases (NOXs), the endoplasmic reticulum (ER), and peroxisomes. To maintain redox homeostasis, the intracellular antioxidant system, comprising various antioxidant enzymes (e.g., superoxide dismutase, catalase, glutathione peroxidase, and peroxiredoxin) and non-enzymatic molecules (e.g., glutathione, flavonoids, and vitamin C), dynamically regulates ROS levels and repair oxidative damage [49].
Oxidative stress is a condition characterized by an imbalance between ROS production and the antioxidant defense system's ability to detoxify these harmful molecules or repair the resulting damage. This state can lead to oxidative damage to biomolecules, including proteins, lipids, and DNA, and has been linked to cellular senescence [50]. For example, mitochondrial calcium overload can disrupt the electron transport chain (ETC) in cardiomyocytes, leading to excessive ROS production and damaging the DNA structure, ultimately triggering premature cardiac aging [51]. Furthermore, vanadate is found to directly oxidize and increase the efflux of glutathione (GSH) species, causing cellular senescence in lung fibroblast cells independent of ROS intermediates [52].
Oncogene activation
Oncogenic activation is a critical mechanism in tumor initiation and progression, driving cell growth and division. However, studies have shown that activated RAS can induce senescence in fibroblasts. Subsequent studies have confirmed and expanded this finding, demonstrating that other activated oncogenes, such as BRAF and MYC, can also induce replication stress, resulting in stable growth arrest in cultured cells [53,54,55] (Fig. 2).
RAS is a family of small GTPases, including HRAS, KRAS, and NRAS, that play a crucial role in regulating various cellular processes, such as growth, differentiation, and survival [56]. Upon activation, RAS interacts with several downstream effectors, most notably the MAPK/ERK and PI3K-AKT pathways, leading to a cascade of signaling events that promote cell proliferation and survival [57, 58]. In various tumors, mutations or overexpression of RAS are commonly associated with constitutive activation, resulting in uncontrolled cell growth and proliferation. Aberrant proliferative signaling induces DNA replication stress and activates DDR, thereby leading to cellular senescence [59]. Furthermore, RAS-related pathways can also promote the metabolic production of ROS in cells, leading to the accumulation of ROS and exacerbating DNA damage [60, 61]. Other components of the RAS pathway, including RAF, RAC1, and MEK, may also induce cellular senescence [62, 63]. For instance, baicalin has been demonstrated to upregulate the expression of decidual protein induced by progesterone, which interacts with KRAS, activating the MAPK/ERK pathway and the p16INK4a-CDK4/6 pathway, resulting in colon cancer cell senescence with an anti-cancer effect [64]. Similarly, BRAF, a member of the RAF kinase family, is commonly mutated at position V600E, triggering p53 and p16INK4a-dependent senescence [65].
SASP
SASP refers to the secretion of a variety of pro-inflammatory and pro-senescence molecules, including cytokines, chemokines, matrix metalloproteinases (MMPs), and growth factors, by senescent cells. These secreted factors not only mark the senescent state but can also induce senescence in neighboring cells [66]. Pro-inflammatory cytokines like IL-6 and IL-8 sustain and amplify inflammatory responses through autocrine and paracrine signaling, exacerbating tissue damage. This chronic inflammation, known as "inflammaging," can further activate DDR, driving cells into senescence [67]. The long-term presence of inflammatory mediators also leads to the inhibition of antigen presentation and immune cell differentiation, thereby impairing the ability of the immune system to clear senescent or damaged cells, further perpetuating the cycle of inflammation and senescence [68]. Moreover, this chronic inflammatory environment can foster tumor growth by supporting cell proliferation, angiogenesis, and metastasis [69, 70]. Additionally, matrix-degrading enzymes disrupt the extracellular matrix, impair tissue architecture, and induce DNA damage in surrounding cells, ultimately promoting senescence in neighboring cells [71, 72].
In summary, cellular senescence arises from a complex interplay of various factors. Environmental factors can induce telomere shortening, disrupt redox balance, and activate oncogenes, thereby triggering the activation of senescence pathways. Simultaneously, host factors contribute to cellular senescence through gene regulation, metabolic buildup, and DNA damage. These factors interact synergistically, thereby shaping the pathways of senescence.
Mechanisms of senescence
The molecular mechanisms underlying cellular senescence are complex and diverse, which can be categorized into two main classes: DNA damage-induced senescence and non-DNA damage-induced senescence. DNA damage-induced senescence is primarily driven by factors such as telomere shortening, oxidative stress, and oncogene activation, while non-DNA damage mechanisms promote senescence by regulating key molecules including p16INK4a and SASP.
DNA damage-induced cellular senescence
Cellular DNA is frequently subjected to damaging events from both environmental factors and intrinsic factors as mentioned previously. DNA damage can be broadly classified into two categories: (1) lesions affecting a single strand of the double helix, such as modified bases, abasic sites, helix-distorting base lesions, and single-strand breaks (SSBs); (2) lesions involving both strands, including interstrand crosslinks and double-strand breaks (DSBs) [73]. In response, cells activate DNA repair pathways to remove lesions. When damage cannot be effectively repaired, it can lead to genomic instability and trigger signaling cascades, ultimately resulting in cellular senescence or apoptosis [74]. This phenomenon, where DNA damage induces senescence, is termed DNA damage-induced cellular senescence (Fig. 3). This type of senescence can be categorized based on its origin into RS, telomere dysfunction-induced senescence, oxidative stress-induced senescence (OSIS), oncogene-induced senescence (OIS), and TIS. RS refers to the gradual shortening of telomeres in normal cells as the number of divisions increases, leading to DNA damage and cell cycle arrest [75, 76]. Telomere dysfunction-induced senescence, on the other hand, refers to the process in which telomere shortening and cellular senescence are triggered by dysfunction of telomerase and the shelterin, often due to factors other than normal division [77,78,79]. OSIS results from oxidative damage that compromises DNA integrity and repair mechanisms [80]. OIS is triggered by oncogene activation, causing replication stress, oxidative stress and DDR [12, 81]. TIS arises from cancer therapies, including chemotherapy and radiation, which induce DNA damage and promote senescence in tumor cells [82].
Pathways of DNA damage-induced cellular senescence. DNA damage is a critical event that can arise from various sources, including cancer therapies, oxidative stress, telomere shortening, and oncogene activation. In response to DNA damage, cells activate a series of signaling pathways collectively known as the DNA damage response (DDR). Severe types of DNA damage, particularly SSBs and DSBs, are more likely to trigger cellular senescence. SSBs activate the ATR-CHK1 pathway, which inhibits CDC25 and subsequently suppresses the CDK1/Cyclin A and CDK1/Cyclin B complexes, thereby blocking G2/M transitions and inducing cell cycle arrest. In the case of DSBs, the ATM-CHK2 axis activates p53, leading to p21 induction, which suppresses CDK4/6-Cyclin D and CDK2-Cyclin E/A complexes, driving senescence. Phosphorylated CHK2 also inhibits CDC25, blocking the activation of CDK1 and CDK2, inducing cell cycle arrest. Additionally, ATM also stabilizes p53 by inhibiting MDM2, further enhancing p53-mediated senescence. Moreover, Sp1 activated by ATM, Ets activated by ERK as well as the accumulation of E2F activate p16INK4a, which binds to CDK4/6, preventing their association with Cyclin D and further inhibiting Rb phosphorylation, then reinforcing cell cycle arrest
Severe types of DNA damage, particularly SSBs and DSBs, are more likely to trigger cellular senescence. Following the formation of SSBs, ataxia-telangiectasia and Rad3-related protein (ATR) is recruited to the damage site or replication stress by its partner protein ATRIP, which facilitates ATR's binding to replication protein A (RPA)-coated single-stranded DNA. ATR is subsequently activated by topoisomerase-binding protein 1 (TOPBP1), which phosphorylates and activates CHK1 [83]. Phosphorylated CHK1 inhibits the phosphatase CDC25, a group of phosphatases including CDC25A, CDC25B, and CDC25C, thereby preventing the activation of the CDK1-cyclin A/B complexes, obstructing the transition from G2 to M phase, ultimately leading to senescence [84, 85].
After DSBs occur, the Mre11-Rad50-Nbs1 (MRN) complex first reaches the damage site and uses its nuclease activity to recognize the broken ends. Subsequently, the MRN complex interacts with ataxia-telangiectasia mutated kinase (ATM), promoting its dimer dissociation into monomers and triggering ATM autophosphorylation, thereby activating ATM [86]. ATM is a key protein kinase that detects DSBs and initiates the DDR by phosphorylating essential proteins, including CHK2 and p53. Upon DSB formation, the MRN complex (MRE11-RAD50-NBS1) recruits ATM to the damage site and facilitates ATM autophosphorylation, leading to the dissociation of its dimer into active monomers[87]. Activated ATM phosphorylates downstream targets such as CHK2 and p53 [88] (Fig. 3). Phosphorylated CHK2 inhibits CDC25A and CDC25C, blocking the activation of CDK2 and CDK1, which results in cell cycle arrest [89, 90]. Furthermore, activated CHK2 phosphorylates p53 at Ser20, enhancing its stability and transcriptional activity [91]. ATM also directly phosphorylates p53 at Ser15 and inhibits MDM2, preventing it from ubiquitinating p53, which further promotes p53 accumulation [92]. Additionally, ATM enhances p53's transcriptional activity by inhibiting the SIRT1 deacetylase activity, which is regulated by LARP7 [93]. p53 stimulates the expression of the cyclin-dependent kinase inhibitor p21, which binds to CDK2, inhibiting the CDK2-cyclin E/A complex while also blocking the CDK4/6-cyclin D complex. This dual inhibition prevents the cell cycle from progressing from the G1 to S phase, ultimately resulting in cellular senescence [94]. The retinoblastoma protein (RB) is a crucial downstream target of cyclin-dependent kinases (CDKs) in cell cycle regulation. As the cell cycle progresses, RB is phosphorylated by the CDK2-cyclin E/A complex and the CDK4/6-cyclin D complex, facilitating the release of E2F and the transition to the S phase. When CDK complexes are inhibited, hypophosphorylated RB binds to E2F transcription factors, blocking E2F-mediated transcription of genes essential for cell cycle progression [95]. Additionally, ATM directly phosphorylates the transcription factor Specificity Protein 1 (Sp1) at Ser101, enhancing its transcriptional activity. Phosphorylated Sp1 subsequently upregulates DDR genes, including p16INK4a, promoting cellular senescence [96, 97].
Non-DNA damage-induced cellular senescence
In addition to DNA damage, non-DNA damage signals significantly contribute to cellular senescence. A key pathway involved in non-DNA damage-induced senescence is the p16INK4a-CDK4/6 pathway. When cells detect excessive proliferative signals, p16INK4a activity is upregulated, forming a negative feedback mechanism to suppress uncontrolled proliferation. This mechanism is particularly important in response to oncogenic activation. During periods of excessive cell proliferation, hyperphosphorylated Rb releases more E2F transcription factors, which activate p16INK4a transcription. p16INK4a then binds to CDK4 or CDK6, preventing their association with cyclin D. This inhibits the phosphorylation of Rb by the CDK4/6-cyclin D complex, ultimately suppressing the cell cycle [98, 99]. Additionally, transcription factors from the Ets family, such as Ets1 and Ets2, are activated by the MAPK/ERK pathway, and bind to the p16INK4a promoter, further increasing its expression [100].
SASP factors operate upstream in multiple signaling pathways and have broad-reaching effects. Released by primary senescent cells, these factors induce senescence in neighboring cells through a process known as Paracrine Senescence (PS) [101]. The signaling pathways activated by SASP factors are diverse and largely independent of DNA damage. For instance, the secreted cytokine TGF-β binds to receptors on adjacent cells, activating receptor-regulated SMADs (such as SMAD2 and SMAD3), which form complexes with SMAD4 and translocate into the nucleus, enhancing the expression of p21 and p15, thus inducing senescence [102]. TGF-β also activates the PI3K/AKT pathway, upregulating the expression of ubiquitin-specific protease 15 (USP15), which stabilizes p53 through deubiquitination [103]. Similarly, the SASP factor IL-6 can bind to the gp130 subunit of its receptor, activating JAK-STAT3 pathways. This activation upregulates p21 expression and promotes senescence [104].
Characteristics and detection indexes of senescent cells
Cellular senescence is characterized by distinct morphological, molecular, and metabolic changes. Understanding these features facilitates the effective identification of senescent cells. The following are common characteristics of cellular senescence along with their respective detection indicators (Fig. 4).
Characteristics of senescent cells. Senescent cells exhibit distinct morphological, metabolic and functional changes. Main morphological changes (1) include cell enlargement, flattening, lipofuscin (2) and the formation of senescence-associated heterochromatin foci (SAHF) (3). Another defining characteristic of senescent cells is their stable cell cycle arrest (4). The lysosomal enzyme SA-β-gal becomes increasingly active during senescence and serves as a reliable marker for this state (5). Additionally, senescent cells exhibit mitochondrial dysfunction (6), resulting in the accumulation of ROS and oxidative stress. DNA damage (7), indicated by γ-H2AX foci, serves as an additional hallmark of senescent cells. When telomeres reach a critically short length (8), they trigger a DNA damage response that leads to cell cycle arrest. Furthermore, senescent cells secrete pro-inflammatory cytokines, chemokines, and proteases, collectively termed the senescence-associated secretory phenotype (SASP) (9)
Cellular morphological changes
Morphological alterations represent a key characteristic of senescent cells, especially in tumor cells, and are associated with a diminished capacity for proliferation. Senescent cells typically display enlargement, flattening, and increased dispersion [105]. These changes correlate with the accumulation of cofilin-1 and the hyperphosphorylation of microtubule-associated protein tau in senescent cells. These modifications enhance the rigid cytoskeletal structures formed by actin filaments, microtubules, and intermediate filaments [106]. Moreover, the accumulation of cytoplasmic particles, such as lipofuscin, in senescent cells represents a significant morphological change that can be clearly observed in histological sections. Lipofuscin is a pigment composed of oxidatively damaged lipids, proteins, and other metabolites. Its accumulation typically occurs in senescent cells due to a weakened clearance mechanism, particularly the reduced function of lysosomes. Currently, Sudan Black B (SBB) and its analogues are the most widely used methods for detecting lipofuscin. SBB specifically binds to oxidized lipids and lipoproteins in lipofuscin, forming a dark precipitate that can be directly observed under an optical microscope [107,108,109,110,111]. Beyond these cellular changes, nuclear morphology serves as a predictive biomarker of senescence, particularly through chromatin remodeling. These changes are primarily characterized by the formation of heterochromatin regions, alterations in histone modifications, and the suppression of gene transcription [112, 113]. SAHF are unique chromatin structures found in senescent cells and can be detected using immunofluorescence staining techniques [114]. SAHF plays a crucial role in silencing genes related to cell proliferation, such as E2F target genes, thereby preventing further cell division [62]. Researchers observe that cellular senescence in breast cancer leads to the formation of distinct SAHF foci and an upregulation of H3K9me3. Chromatin remodeling inhibits the expression of cell cycle-related genes, prompting cancer cells to enter an irreversible state of arrest [115].
Cell cycle arrest
A hallmark of senescent cells is their inability to continue dividing, often halting at the G1 or G2/M phase of the cell cycle. This arrest is mediated by altered cell cycle regulatory mechanisms that prevent senescent cells from entering the S phase for DNA replication (Fig. 4). As previously discussed, several cell cycle inhibitors, including p53, p21, and p16INK4a, impede cell division by inhibiting cyclin-CDK complexes. For instance, studies show that inhibition of the enhancer of zeste homolog 2 (EZH2) in gastric cancer cells leads to increased expression of p21 and p16INK4a, resulting in irreversible G1 phase arrest and initiation of cellular senescence [116]. Techniques like western blotting and quantitative polymerase chain reaction (qPCR) are commonly used to measure the expression levels of these inhibitors, providing insights into tumor proliferative activity and the onset of senescence [117, 118].
Cellular metabolic changes
With the occurrence of senescence, the metabolism of cells undergoes significant changes, which not only impair the cell's own function but also affect the overall health of the surrounding microenvironment and tissues. Common metabolic characteristics of senescent cells include enhanced glycolysis [119], lipid accumulation [120], alterations in amino acid metabolism [121], and an imbalance between oxidative stress and antioxidant defense due to mitochondrial dysfunction [122]. In addition, senescent cells also have unique protein alterations, among which the change in β-galactosidase activity is an important indicator for detecting senescent cells.
β-galactosidase is a hydrolase enzyme commonly used as a classic biomarker for cellular senescence [123]. The number and size of lysosomes increase in senescent cells, and SA-β-gal, as a lysosomal enzyme, becomes more active during the senescence process. In tumor cells, SA-β-gal activity is frequently used to evaluate whether treatment strategies induce senescence in cancer cells [124]. The most widely used method for detecting SA-β-gal is X-gal staining, a chemical reaction that produces a blue product through hydrolysis. At pH 6.0, SA-β-gal in the cells hydrolyzes X-gal, resulting in blue spots observable under a light microscope. For more sensitive and quantitative detection of SA-β-gal, especially in high-throughput screening, fluorescence-based assays have also been employed [125].
SASP
Once cells enter a senescent state, they secrete various pro-inflammatory cytokines, chemokines, proteases, and other molecules collectively known as the SASP [126]. In senescence induced by genotoxic stress, the p38 mitogen-activated protein kinase (p38MAPK) pathway is activated, regulating the mRNA levels of SASP factors such as GM-CSF, IL-6, IL-8, GROα, MCP-2, and IL-1α [127]. Additionally, the cGAS-STING pathways also play critical roles in this process [128]. The SASP enables senescent cells to transmit damage signals to surrounding cells or tissues, promoting a series of immune responses [129, 130]. However, when senescent cells are not promptly cleared, sustained secretion of the SASP creates a growth-stimulatory and immunosuppressive microenvironment that supports tumor development [5, 131]. For instance, SASP factors like IL-6 and IL-8 are prominently expressed in senescent tumor cells, promoting inflammation and angiogenesis in the microenvironment [132]. Given that the expression of SASP factors is often elevated in senescent cells, monitoring these factors through techniques such as ELISA, qPCR, or western blotting can provide valuable insights into cellular senescence. However, the SASP of senescent cells exhibits diverse and unstable characteristics, complicating their identification with a single, universal, or model-specific biomarker. Thus, a combination of multiple detection indicators is necessary for the accurate identification of senescent cells in tissues [133].
DNA damage
As mentioned earlier, in response to DNA damage, cells initiate a series of protective mechanisms, collectively known as the DDR. When DDR is activated but fails to repair the damage, the stress may drive cells to enter senescence. This process is accompanied by the activation of DNA damage markers such as 53BP1 and γ-H2AX, which can be detected by immunofluorescence to identify senescent cells [134, 135]. Additionally, because telomere shortening can also induce DNA damage and promote cellular senescence, telomere length is commonly used as an auxiliary marker for detecting senescent cells.
It is worth noting that, to date, no single biomarker has been proven sufficient to reliably detect cellular senescence in vivo, and a combination of multiple biomarkers remains necessary [109]. The biomarkers currently widely accepted for indicating the presence or absence of senescent cells are listed below (Table 1).
Reversibility of cellular senescence
Since 1961, cellular senescence has commonly been defined as a state in which a cell undergoes irreversible cell cycle arrest following prolonged division or exposure to various forms of stress, with cyclin dysregulation playing a critical role. However, the view of senescence as a strictly irreversible phenomenon has been increasingly questioned [170]. As noted above, the p53-p21 and p16INK4a-CDK4/6 pathways are critical regulators of cellular senescence, acting to inhibit the activity of CDK-cyclin complexes and thereby arresting the cell cycle. Yet, in cases where p53 is ineffective or inactivated, particularly in certain tumors, the cellular response to DNA damage becomes less stringent [171, 172]. In such cases, p21 may promote genomic instability, leading to the accumulation of DNA damage and impairing the progression of the senescence program. As a result, tumor cells in a senescent state may bypass the senescence barrier, producing more aggressive progeny and facilitating tumor progression [173]. Experimental evidence shows in p53-deficient lung cancer cells, p21-mediated senescence can be reversed. This occurs through the upregulation of replication factors, such as Cdt1 and Cdc6, which suppress the expression of tumor suppressors like p16INK4a and p14. These findings suggest that p21 may play a role in allowing tumor cells to escape senescence, providing a potential mechanism for cancer cell survival and proliferation [18].
In addition to the reversibility of individual cell senescence, the potential reversibility of entire senescent cell populations is also a significant concern. Cellular senescence is not confined to cells under severe stress; it can also affect neighboring cells through the secretion of SASP. Studies have shown that SASP plays a crucial role in maintaining tissue homeostasis by preventing the loss of entire tissue regions when cells are exposed to stressors that induce cell death or senescence [3]. In the context of tumors, senescent cells can contribute to a more aggressive growth phenotype in neighboring cells through SASP signaling [174]. The reversibility of these senescent cell populations is closely linked to tumor growth, recurrence, and drug resistance [175]. Therefore, the development of targeted therapies aimed at eliminating senescent cells represents a promising therapeutic strategy.
Heterogeneity of cellular senescence
The heterogeneity of cellular senescence implies that different cells exhibit distinct features, mechanisms, and outcomes during the process of senescence [176, 177]. Even within the same cell type, variability exists in the onset, progression, manifestation, and response to therapy [178]. As previously mentioned, the factors inducing cellular senescence can be classified into environmental and host factors, and the pathways involved in senescence, such as ATR-CHK1, p53-p21, and p16INK4a-CDK4/6, may also vary. The specific manifestations of heterogeneity of senescence are primarily reflected in the characteristics of the SASP [179]. The types and secretion levels of SASP factors vary between different cells and tissues. The factors can be influenced by the type of senescence and its inducing factors. Additionally, the composition of the SASP can change at different stages of the senescence process [170]. In the tumor microenvironment, this heterogeneity is particularly pronounced. We will describe in detail below that senescent cells have a dual role in tumorigenesis and progression. Senescent tumor cells can secrete both pro-inflammatory cytokines, such as IL-6, IL-8, and TNF-α, as well as immunosuppressive factors like IL-10, TNF-β, and VEGF [66]. They can exert anti-tumor effects by inhibiting tumor growth, but may also contribute to tumor progression by promoting adverse outcomes such as metastasis and drug resistance [44, 178, 180]. Therefore, understanding the role of senescent cells and their SASP factors in tumors, particularly across different tumor types and therapeutic contexts, is crucial for the development of novel and more effective therapeutic strategies [181].
Epigenetic regulation of cellular senescence
Epigenetic modifications to DNA and chromatin, such as DNA methylation, histone modifications, and chromatin remodeling, are key regulators of genome architecture and gene expression [182]. These modifications are crucial in controlling cellular senescence by influencing the expression of senescence-associated genes and the secretion of SASP factors [183].
Cytosine residues in the promoter regions of genes can undergo methylation to form 5-methylcytosine, typically leading to gene silencing. This repression can either promote or inhibit cellular senescence, depending on the specific genes involved. For instance, Oroxylin A has been demonstrated to inhibit cGAS gene methylation by reducing DNMT3A activity, thereby enhancing the cGAS-STING pathway and promoting cellular senescence [184]. Conversely, treatment with telomerase activator compounds (TAC) can stimulate DNMT3B-mediated methylation of the p16INK4a promoter, thus suppressing cellular senescence [141]. Additionally, DNA methylation analysis methods can be used to predict the biological age of individual cells [185].
Recent findings have highlighted that the chemical modifications (e.g., acetylation, methylation, phosphorylation) of histones can alter chromatin structure, thereby influencing gene expression. For example, histone modifications such as H3K9me3 and H3K27me3 contribute to the formation of the senescence-associated heterochromatin foci (SAHF), which silence proliferation-related genes and reinforce the senescent state. Additionally, the transcription factor ZBP-89 and histone deacetylase 3 bind to the p16INK4a promoter, regulating its expression via histone acetylation and consequently influencing cellular senescence [186, 187].
Chromatin remodeling complexes, such as SWI/SNF, ISWI, NuRD, and INO80, can modify chromatin structure by sliding, rearranging, or removing nucleosomes [188]. This modification renders DNA more accessible to transcription factors and other regulatory proteins, thereby promoting gene expression, or alternatively, tightly packaging DNA to suppress gene expression. The long non-coding RNA JPX (just proximal to XIST) interacts with components of chromatin remodeling complexes, such as p65 and BRD4, enhancing the transcription of SASP genes and facilitating cellular senescence [189].
Together, these dynamic epigenetic mechanisms underscore the central role of epigenetic regulation in controlling the cellular senescence program.
Cellular senescence and tumor
Cancer cells are characterized by their ability to proliferate indefinitely, circumvent regulatory controls, and evade programmed cell death, contributing to their prolonged survival. In contrast, senescent cells lose the ability to divide and enter a state of permanent growth arrest. Oncogene-induced cellular senescence is a critical barrier to the malignant progression of human tumors. It induces DNA replication stress and activates DDR, leading to cell cycle arrest during the early stages of tumorigenesis [81, 190]. It has been demonstrated that cellular senescence can limit in vivo tumorigenesis through the p53 and p16INK4a pathways [14, 191]. Additionally, senescent cells are frequently observed in precancerous tumor tissue [192]. Therefore, cellular senescence is typically viewed as a detrimental factor in tumor development and progression. However, emerging evidence indicates that senescent cells can also promote malignant behavior in cancers.
Tumor-associated senescence regulators
Although intracellular oncogenes can induce cellular senescence due to replication stress, research has shown that tumor cells have developed multiple mechanisms to inhibit this process. These adaptations allow tumor cells to evade senescence, enabling them to continue proliferating (Table 2).
A series of studies has demonstrated that specific signaling molecules inhibit cancer cell senescence in various cancer models by directly modulating the p53-p21 and p16INK4a pathways. Sirtuins, a family of NAD⁺-dependent deacetylases, play a crucial role in regulating various biological processes, including senescence, metabolism, oxidative stress, and inflammation [249, 250]. Recent studies show that upregulation of SIRT1, induced by estrogen receptor α (ERα) in breast cancer, leads to the inactivation of p53 and cyclin G2, thereby inhibiting senescence and promoting cell survival [196]. Synaptotagmin-7 (SYT7) is upregulated in lung cancer, which augments the interaction between p53 and MDM2, subsequently downregulating the expression levels of p53, p21, and p16INK4a, thereby shutting down the senescence program [202]. Similarly, the serine/threonine protein kinase NEK6 [201], the deubiquitinase PSMD7 [203], and casein kinase 2 (CK2) [204] all exhibit inhibition of p53 activity, thereby blocking senescence in lung cancer cells. In lung adenocarcinoma cells, the deletion of IKKα leads to decreased levels of NRF2 and NQO1, which inhibits the p53/p21 pathway and subsequently prevents the induction of senescence [206]. In addition to these factors, the GATA family, comprising GATA1, GATA2, GATA3, GATA4, GATA5, and GATA6, plays a crucial role as essential transcription factors that influence cell proliferation, differentiation, and survival. Specifically, GATA4 and GATA6 exhibit tumor-suppressing functions and are often downregulated in lung cancer. GATA4 facilitates cellular senescence by regulating microRNAs (miRNAs), promoting chromatin remodeling, and activating senescence-associated genes. Meanwhile, GATA6 enhances the expression of p53 and p21, which collectively inhibits tumor progression [207, 208]. As a critical node in the senescence pathway, p16INK4a is often inhibited by cancer-promoting factors. For example, the transcription factor ZBP-89 epigenetically represses p16INK4a expression, preventing senescence in lung cancer cells [186]. Similarly, the enzyme aspartate β-hydroxylase (ASPH) is overexpressed in HCC. Inhibition of ASPH expression and activity leads to the inactivation of GSK3β, promoting stabilization of p16INK4a and inducing senescence [223]. In the context of colorectal cancer, ZEB1 promotes tumor growth by activating DKK1, mutant p53, Mdm2, and CtBP, while simultaneously inhibiting p53-mediated senescence and apoptosis. Moreover, ZEB1 suppresses senescence-related genes such as p16INK4a and p21, contributing to the aggressive nature of the tumor [210].
Under normal conditions, oxidative stress can trigger cellular senescence, leading to growth arrest. However, cancer cells boost their tolerance to oxidative damage, enabling them to evade this process and keep proliferating. Compared to normal gastric epithelial cells, the expression of SIRT3 is significantly elevated in gastric cancer cells. SIRT3 enhances the activity of MnSOD, protecting cells from oxidative damage, which confers strong anti-senescence properties [213]. In glioblastoma, VEGFR2 has been found to suppress cell progression by inducing OSIS through the AKT-PGC1α-TFAM mitochondrial biogenesis signaling cascade [239].
RS, one of the important mechanisms underlying cellular senescence, is often suppressed in various cancer cells. Sphingosine kinase 2 (SK2) has been reported to produce sphingosine-1-phosphate (S1P), which binds to TERT, assisting in telomerase stabilization and thus inhibiting senescence while sustaining tumor growth [200]. In infant acute lymphoblastic leukemia (ALL) and acute myeloid leukemia (AML), fusion oncogenes such as MLL/AF4 and AML/MTG8 are frequently activated. These genes are critical for maintaining telomerase activity, and inhibiting their expression can downregulate TERT and induce cellular senescence [236].
Non-coding RNAs (ncRNAs) also play critical roles in regulating cellular senescence in cancers. Compared to normal liver tissues, levels of ncRNAs such as Let-7i-3p, miR-449b-3p, miR-624-5p, miR-885-5p, miR-138, and miR-125b are significantly reduced in tumors. These miRNAs suppress cell proliferation and induce senescence by modulating downstream pathways, including inhibition of Wnt/β-catenin signaling, downregulation of TERT expression, and suppression of SUV39H1 [228,229,230]. miR-449a inhibits E2F3 activity, blocking the G1/S phase transition, and its expression is significantly reduced in lung cancer tissues [228].
Cancer-promoting functions of cellular senescence
While the suppression of cellular senescence acts as a protective mechanism in tumorigenesis and progression, senescent cells may also exert beneficial effects within this context. Premature senescence in normal cells may increase the risk of tumorigenesis [251,252,253]. For instance, the restoration of mammary cell viability by the drug TPCA-1 significantly reduces the incidence of age-related cancers [254]. Similarly, skin cells exposed to ultraviolet radiation age more rapidly and demonstrate a significantly higher risk of developing skin cancer. In this context, the accumulation of senescent cells and SASP may accelerate the progression of cutaneous squamous cell carcinoma (cSCC) [255, 256]. This increased cancer risk may be associated with factors such as telomere shortening, accumulation of genetic and epigenetic alterations, and reduced DNA repair capacity [257].
An increasing body of evidence suggests that SASP plays an important role in mediating both pro-tumorigenic and anti-tumorigenic effects in cancer progression. Components of the SASP contribute to the establishment of a pro-inflammatory and immunosuppressive microenvironment that facilitates the growth of cancer cells. For example, senescent hepatic stellate cells contribute to chronic inflammation in the tumor microenvironment (TME) by secreting IL-1β, thereby facilitating the progression from non-alcoholic steatohepatitis (NASH) to liver cancer [44, 258]. Similarly, senescent cells can secrete IL-33 to stimulate the recruitment of immunosuppressive cells such as Tregs, further promoting tumor growth through immune suppression [259]. Additionally, SASP can drive tumor invasion and metastasis. For example, senescent glioma cells promote invasion through the secretion of cathepsin B [260], while senescent colon cancer cells induce epithelial-mesenchymal transition (EMT) in surrounding cells via IL-6, MMP-3, fibroblast growth factor (FGF), and hepatocyte growth factor (HGF) [261]. Conversely, the SASP exerts antitumor effects. In the early stages of liver carcinogenesis, IL-1β and TNF-α activate a "senescence surveillance" mechanism that recruits immune cells, including natural killer cells and macrophages, to eliminate potential cancerous cells [262].
Harnessing cellular senescence for cancer therapy
Cellular senescence plays a dual role in cancer development, making it a promising target for anticancer therapy [263]. Several treatments—including targeted therapies, chemotherapy, radiotherapy, and traditional Chinese medicine (TCM) —aim to inhibit tumor growth and metastasis by inducing senescence in tumor cells. However, senescence in immune cells within the TME can weaken antitumor immunity and promote cancer progression, underscoring the importance of anti-senescence strategies. This review will summarize the mechanisms involved in targeting cellular senescence in anticancer therapies.
Targeted therapy
Tumor-targeted therapy is an innovative approach to cancer treatment that aims to inhibit the growth and spread of cancer cells while minimizing damage to normal cells and reducing side effects by targeting specific molecular or genetic pathways. The mechanism of senescence inducers aligns with this concept, as they drive cancer cells into a state of senescence, halting their proliferation [264]. This positions senescence inducers as a novel class of tumor-targeted therapies. In the following section, we will explore the mechanisms and efficacy of different senescence inducers (Table 3).
Telomere shortening is a crucial factor in cell senescence. In many cancers, elevated telomerase expression and activity enable cancer cells to maintain telomere length and evade senescence. To counter this, several telomerase-targeted therapies aim to induce senescence in tumor cells by promoting telomere shortening. Atorvastatin, commonly used to lower cholesterol, inhibits the IL-6/STAT3 signaling pathway, thereby reducing TERT expression and inducing G0/G1 phase cell cycle arrest, ultimately suppressing the proliferation of hepatocellular carcinoma (HCC) cells [265]. Additionally, the TTAGGG repeat sequence at the 3' end of telomeric DNA can form G-quadruplex structures through G-G pairing, which prevents telomerase from binding and elongating telomeres. G-quadruplex-stabilizing ligands, such as perylene and phenyl-imidazole-ethylamine-platinum (II) (PIP), promote the formation of these structures, inhibit telomerase activity, and suppress cell proliferation [269, 270]. Bone morphogenetic protein-7 (BMP7) inhibits TERT activity via the BMPRII receptor and SMAD3 pathway, leading to telomere shortening in breast cancer cells [271]. Moreover, shelterin complexes play a crucial role in regulating telomere length. For example, the aflatoxin B derivative FKB04 induces senescence in liver cancer cells by inhibiting the expression of telomeric repeat-binding factor 2 (TRF2), disrupting the T-loop structure, shortening telomeres, and ultimately leading to cellular senescence [266].
ROS accumulation is a key trigger of cellular senescence and is often targeted by senescence inducers. Resveratrol enhances ROS generation through the SIRT1/p38MAPK and NO/dLC1 pathways by increasing the expression of tumor suppressor gene DLC1, triggering ER stress, and depleting intracellular antioxidants. Additionally, it increases the expression of DNA double-strand break marker γ-H2AX and decreases the levels of DNA repair proteins p-BRCA1 and RAD51, ultimately promoting SA-β-gal activity and the expression of senescence markers p53, p21, and LaminB [272,273,274]. Some studies show that leukemia can be effectively combated by inducing tumor cells into senescence. Imidazo[1,2-a]pyridines (IPs) enhance lipid peroxidation and reduce GSH levels [277], while curcumin analog pentachloropropane-1 (PGV-1) competes with GSH for binding to GST-P1, inhibiting ROS scavenger enzyme activity [278]. Anthraquinone lactone AS1041 decreases total thiols and disrupts matrix metalloproteases [275], all of which elevate ROS levels and induce senescence in leukemia cells.
In addition to regulating telomere length and ROS levels, targeting key pathways involved in cellular senescence, such as the p53-p21 axis and p16INK4a-CDK4/6 axis, has proven to be an effective therapeutic strategy. DNA methyltransferase inhibitors (DNMTis), including 5-azacytidine, 5-aza-2'-deoxycytidine, pseudolaric acid B (PAB), β-Asarone, and the platinum-zoledronate complex [Pt(en)]2ZL, as well as combinations like cucurbitacin B with Withanone and Aurora kinase A (AURKA) inhibitors with MDM2 antagonists, have been shown to activate the p53 pathway, induce cellular senescence, and inhibit cancer cell growth, tumor progression, and metastasis [280,281,282,283,284,285]. It is noteworthy that some tumors, such as PD-L1-positive senescent tumor cells, can resist immune clearance and relapse after senescence. However, berberine derivative B68 has been shown to induce p53-dependent cellular senescence, disrupt the immunosuppressive PD-1/PD-L1 interaction, and facilitate the rapid clearance of senescent tumor cells [286]. Some drugs can induce cellular senescence by directly activating p21, independent of the p53 pathway. For example, guanylate cyclase inhibitor LY83583 [287], sodium valproate (a known HDAC inhibitor) [288], Argentatin B [289], 1,25(OH)2D3 [290], adapalene [291] and carvacrol [292] all enhance p21 expression, inhibit CDK and cyclins activity, and induce cellular senescence. The frontline treatment for chronic myeloid leukemia (CML), imatinib, not only induces apoptosis and autophagy but also increases p21 and p27 expression, as well as the population of SA-β-gal-positive cells [293]. Selective CDK4/6 inhibitors have also been proven to be effective senescence inducers [294]. For instance, palbociclib inhibits CDK4/6 activity, while also suppressing proteasome inhibitor EMC29, leading to proteasome activation and preventing Rb phosphorylation and E2F release, blocking the transition of cancer cells from the G1 to the S phase [295, 297]. It is worth noting that the senescence induced by CDK4/6 inhibitors in tumor cells is partially reversible, which may increase the risk of tumor recurrence, warranting further investigation [298].
miRNAs are small, non-coding RNA molecules that play a crucial role in regulating senescence-related gene expression by binding to messenger RNA (mRNA), thereby preventing its translation or promoting its degradation [299]. Some miRNAs mentioned earlier (such as miR-449b-3p, miR-624-5p, and miR-138) have been shown to promote cellular senescence [228]. However, their expression is often suppressed in tumor cells. Enhancing the expression of these miRNAs is considered a therapeutic approach to induce senescence. Avenanthramide A (AVN A) increases miR-129-3p expression, inhibits the expression of the ubiquitin ligase Pirh2, and upregulates p53 and its downstream target p21, thereby inducing senescence and inhibiting colon cancer growth [296].
Gene therapy plays a significant role in cancer treatment by manipulating genetic pathways related to cellular senescence to inhibit tumor growth. For instance, infection of A549 lung cancer cells with a recombinant adenovirus carrying the p16INK4a gene leads to high-level expression of the p16INK4a protein, inhibition of telomerase activity, increased SA-β-gal expression, and significant suppression of cancer cell growth [300]. Furthermore, using CRISPR interference and programmable base editing to correct the −124C > T mutation in the TERT promoter effectively suppresses abnormal TERT overexpression, thereby inhibiting the growth of gliomas harboring this mutation [301]. Additionally, knocking down the gastrin-releasing peptide receptor elevates p53, p21, and p16INK4a levels, activates the epidermal growth factor receptor (EGFR), and reduces p38MAPK levels, leading to increased cell size and cell cycle dynamics consistent with cellular senescence [302].
Chemotherapy and radiotherapy
As early as 2003, studies demonstrated that chemotherapy can induce premature senescence in tumor cells, thereby inhibiting tumor growth [303]. For example, doxorubicin has been shown to upregulate the expression of the growth factor BMP4, which subsequently activates the SMAD pathway to increase the expression of p16INK4a and p21, inducing premature senescence in lung cancer cells [304]. Moreover, combining senescence inducers with chemotherapeutic agents can reduce chemotherapy resistance and increase tumor cell sensitivity to treatment. In colon cancer models, citrate, by promoting excessive lipid biosynthesis in tumor cells and disrupting lipid metabolism, may initiate ATM-mediated senescence pathways, thereby enhancing the inhibitory effect of standard chemotherapy on tumor growth [305]. Norcantharidin (NCTD) has been found to enhance chemotherapy in triple-negative breast cancer (TNBC) by inducing cell senescence and cell cycle arrest through the inhibition of phosphorylated Akt and ERK1/2, as well as the upregulation of p21 and p16INK4a [306]. Inhibitors of DNA-dependent protein kinase (DNA-PK), such as the novel inhibitor M3814, enhance chemotherapy's antitumor effects by reducing DNA repair and inducing a p53-dependent senescence pathway [307].
However, other studies suggest that senescence of tumor cells can contribute to enhance the resistance of tumor tissues to chemotherapy. In colon cancer, senescent cells enhance tumor resistance by upregulating INHBA expression, which negatively regulates the Hippo signaling pathway and inhibits apoptosis [308]. In melanoma, cisplatin has shown limited efficacy, possibly due to senescent melanoma cells activating the ERK1/2-RSK1 pathway through SASP factors, promoting the proliferation of non-senescent cells [309]. In addition, chemotherapy can also lead to immune cell senescence and promote tumor recurrence [310].
As previously mentioned, ionizing radiation is a significant inducer of cellular senescence. Therefore, radiotherapy can induce senescence in tumor cells. Ionizing radiation can upregulate the expression of the E3 ligase TRIM22 in hepatocellular carcinoma cells, triggering senescence by degrading the AKT phosphatase PHLPP2 and activating the AKT-p53-p21 pathway [311]. Senescence inducers have been demonstrated to enhance the antitumor efficacy of radiotherapy. PARP inhibitors, for instance, can further inhibit DSB repair, promoting senescence in breast cancer cells and enhancing the effects of radiotherapy [312]. Lipoic acid has been shown to synergize with radiation to induce death and senescence in breast cancer cells by increasing p53 expression, activating p38MAPK and NF-κB, and causing G2/M cell cycle arrest [313]. BIBR1532 inhibits telomerase and increases radiation-induced telomere dysfunction, leading to chromosomal instability and inhibition of the ATM/CHK1 pathway, impairing DNA damage repair and ultimately increasing radiosensitivity in non-small cell lung cancer (NSCLC) [314].
Traditional Chinese medicine
In recent years, as research into the pharmacological effects of TCM has deepened, an increasing number of TCM components and derivatives have been found to exhibit antitumor activity, demonstrating great potential in cancer prevention, treatment, and adjuvant therapy [315]. Many TCM compounds and their derivatives, such as andrographolide [316], artemisinin [317], oridonin [318, 319], and curcumin [320], can induce cellular senescence in cancer cells by enhancing the p53/p21 signaling pathway and SA-β-gal activity, making them promising candidates for antitumor therapy. The alkaloid matrine, derived from the plant Sophora flavescens, and Ligustrum lucidum fruit extract (LLFE) induce G0/G1 phase arrest and senescence in liver cancer cells by upregulating p21 and downregulating Rb phosphorylation [321, 322]. Additionally, certain compounds extracted from traditional Chinese medicinal herbs can induce senescence through oxidative stress. For example, curcumin analog CCA-1.1 and pentachloropropenone PGV-1 selectively induce G2/M phase arrest and OSIS in colorectal cancer cells [323], while Platycodin D2 (PD2), extracted from Platycodon, promotes mitophagy in liver cancer cells via NIX, leading to ROS production and activation of the p21-CDK2 pathway to induce senescence [324]. Interestingly, curcumin's anti-cellular senescence effect is now widely accepted [325]. Additionally, it has been demonstrated that oridonin suppresses the senescence of normal fibroblasts by inhibiting AKT signaling [326]. These findings suggest that the dual role of these compounds in regulating senescence may vary depending on the dose and the specific cell type involved.
Tumor interventions related to the TME
The TME refers to the surrounding environment in which tumor cells reside, including immune cells, fibroblasts, the vascular system, and the extracellular matrix. In recent years, many studies have revealed that the senescent microenvironment plays a significant role in tumorigenesis, progression, and metastasis, providing new insights and therapeutic targets for cancer treatment.
Current research indicates that immune cell senescence within the TME plays a detrimental role in tumor development and progression by facilitating immune suppression and evasion [327]. In certain tumors, senescent T cells (CD57, lacking CD28) accumulate in the TME, resulting in an impaired immune response [328, 329]. This is partly due to dysregulated lipid metabolism and altered phospholipase A2 IVa activity, which result in lipid droplet accumulation in T cells, further compromising their immune function [330]. Senescent macrophages enhance anaerobic glycolysis and promote tumorigenesis by secreting various SASP factors in a paracrine manner, such as Bmp2, Ccl2, Ccl7, Ccl8, Ccl24, Cxcl13, and Il10. Notably, the chemokines CCL7 and CCL24 are particularly involved in enhancing cancer cell invasion and metastasis [331,332,333]. In response to the adverse effects of immune cell senescence, several studies have proposed new therapeutic targets aimed at mitigating immune dysfunction and enhancing antitumor immunity. Preventing tumor-specific T cell senescence by blocking ATM and MAPK signaling, in combination with anti-PD-L1 checkpoint inhibitors, can synergistically enhance antitumor immunity and improve the efficacy of immunotherapy [328]. Moreover, autologous NK cell infusion has been shown to significantly eliminate senescent T cells and suppress tumor progression [334]. Inhibiting the release of SASP factors from senescent cells can effectively alleviate immunosenescence and enhance tumor resistance [335, 336].
Cancer-associated fibroblasts (CAFs), which are abundant in the TME, play a key role in tumor progression. Substantial evidence suggests that senescent CAFs can enhance treatment resistance in tumor cells. Studies have found that radiotherapy induces CAF senescence, which promotes tumor growth through the secretion of insulin-like growth factor-1 [337]. Additionally, senescent CAFs promote the proliferation of NSCLC cells and enhance their radioresistance via the JAK/STAT pathway [338]. One study observed that pretreatment with quercetin effectively reduced the number of doxorubicin-induced senescent fibroblasts and SASP production, thus reducing their pro-tumorigenic effects on osteosarcoma cells [339].
The role of endothelial cell senescence in the TME is primarily related tumor growth, metastasis and spread. Senescent endothelial cells can loosen intercellular junctions, facilitating the spread of tumor cells through the endothelial barrier [340], while also secreting IL-6, which promotes chemotherapy resistance [341]. Interestingly, the tumor suppressor miR-34a induces endothelial progenitor cell (EPC) senescence by inhibiting SIRT1, thereby reducing EPC-mediated angiogenesis and ultimately suppressing tumor growth [342]. Senescent endothelial cells can also promote immune-mediated senescence surveillance through SASP secretion and NF-κB regulation, potentially preventing tumor formation [343].
Senolytics and senostatics
Notably, therapeutic strategies as mentioned above that induce tumor cells senescence may not always be ideal, as senescent cells can promote tumor progression, metastasis, and drug resistance through the secretion of SASP factors and the reversible nature of senescence, especially if they persist for a prolonged period. As a result, inhibiting or eliminating senescent cells is a promising complementary approach to overcoming these challenges, potentially improving therapeutic outcomes and reducing adverse effects associated with prolonged senescence [344].
Senolytics are a class of compounds designed to selectively eliminate senescent cells. These agents reduce the negative effects of senescent cells by specifically targeting BCL-2 family proteins and the p53 pathway to induce programmed cell death in senescent cells [345, 346]. Representative senolytic drugs, such as dasatinib [347] and quercetin [348], exhibit stronger activity in eliminating senescent cells when used in combination [17]. Additionally, when combined with cisplatin and other chemotherapeutic agents, senolytics can potentiate the anti-tumor effects of these treatments [349]. It is important to note that senolytic therapies may have certain toxic side effects under specific conditions, including gastrointestinal toxicity, thrombocytopenia, and the potential to promote tumorigenesis [350,351,352]. These risks highlight the need for further investigation to better understand their safety profile.
Senostatics are another class of drugs designed to target senescent cells. Unlike senolytics, which directly eliminate senescent cells, senostatics inhibit the activity of senescent cells or their SASP [353]. Senostatics can be classified into three categories based on their mechanisms: inhibitors of SASP, blocking antibodies, and inducers of SASP reprogramming [354]. SASP inhibitors, which are numerous, reduce the secretion of IL-6, IL-8, and TGF-β by inhibiting NF-κB and mTOR, thereby alleviating their negative impact on surrounding tissues and the tumor microenvironment [355, 356]. Blocking antibodies, including various neutralizing antibodies against SASP components or their receptors, such as those targeting IL-11, have been shown to reduce cellular senescence and improve organ function [357]. Inducers of SASP reprogramming can convert pro-inflammatory SASP into a pro-immune phenotype, thereby enhancing antitumor effects. For example, a combination of palbociclib (a CDK4/6 inhibitor) and trametinib (a MEK inhibitor) promotes the secretion of TNF-α and ICAM-1 by senescent cells, which in turn stimulates the NK cell immune response [358].
Despite their promising potential, senotherapies face several challenges, including enhancing specificity, mitigating long-term adverse effects, and overcoming senescent cell heterogeneity [354, 359]. To address these challenges more effectively, emerging evidence suggests that combining senescence inducers, senolytics, and senostatics may enhance therapeutic efficacy compared to using them individually [360]. Recent findings show that ferroptosis inducers or Fe (II)-activated prodrugs can selectively trigger apoptosis in both primary and paracrine senescent cells, reducing cytotoxicity in an Fe (II)-dependent manner [361]. Furthermore, various studies have explored ways to improve the specificity and reduce the side effects of senolytics and senostatics by developing specific delivery systems [362, 363]. For instance, the high lysosomal β-galactosidase activity in senescent cells has been exploited to design delivery systems using galactooligosaccharide-coated drugs [364,365,366]; the lipofuscin accumulation in senescent cells has been utilized to develop micelle nanocarriers that bind to lipofuscin [367], allowing for more precise targeting of senescent tumor cells and enhancing the anti-tumor effect.
Summary and outlook
In conclusion, cellular senescence plays a dual role in tumor development and progression, functioning as both a tumor suppressor and promoter, with its effects shaped by the heterogeneity of senescence. This complexity necessitates a deeper understanding of the mechanisms underlying senescence and its context-dependent effects within the TME. Future research should focus on elucidating these intricate pathways, particularly the specific contributions of SASP components in various tumor contexts. Developing targeted therapies that modulate SASP could enhance efficacy while minimizing off-target effects, especially in metastatic cancers [368]. Additionally, since senescent tumor cells may impact the efficacy of senescence-inducing therapies due to their reversibility, it is crucial to understand the mechanisms underlying this process. At the same time, optimizing combinations of senescence-inducing therapeutic strategies with senolytics and senostatics is crucial for achieving an optimal therapeutic balance that maximizes benefits while minimizing side effects [369]. Finally, identifying reliable senescence biomarkers to predict treatment response and monitor therapeutic efficacy could facilitate more precise and personalized treatment approaches. Addressing these challenges could significantly advance the clinical impact of senescence-targeted therapies in oncology.
Data availability
No datasets were generated or analysed during the current study.
The authors declare no competing interests.
Abbreviations
- ALL:
-
Acute lymphoblastic leukemia
- AML:
-
Acute myeloid leukemia
- ASPH:
-
Aspartate β-hydroxylase
- ATM:
-
Ataxia-telangiectasia mutated kinase
- ATR:
-
Ataxia-telangiectasia and Rad3-related protein
- AURKA:
-
Aurora kinase A
- AVN A:
-
Avenanthramide A
- BMP7:
-
Bone morphogenetic protein-7
- CAFs:
-
Cancer-associated fibroblasts
- CBD:
-
Cannabidiol
- CDKs:
-
Cyclin-dependent kinases
- cGAS:
-
Cyclic GMP-AMP synthase
- CK2:
-
Casein kinase 2
- CML:
-
Chronic myeloid leukemia
- COVID-19:
-
Coronavirus disease 2019
- cSCC:
-
Cutaneous squamous cell carcinoma
- DDR:
-
DNA damage response
- DNA-PK:
-
DNA-dependent protein kinase
- DNMTis:
-
DNA methyltransferase inhibitors
- DSBs:
-
Double-strand breaks
- EGFR:
-
Epidermal growth factor receptor
- EMT:
-
Epithelial-mesenchymal transition
- EPC:
-
Endothelial progenitor cell
- ER:
-
Endoplasmic reticulum
- ERα:
-
Estrogen receptor α
- ETC:
-
Electron transport chain
- EZH2:
-
Enhancer of zeste homolog 2
- FGF:
-
Fibroblast growth factor
- GSH:
-
Glutathione
- HCC:
-
Hepatocellular carcinoma
- HGF:
-
Hepatocyte growth factor
- IPs:
-
Imidazo[1,2-a]pyridines
- LKB1:
-
Liver kinase B1
- LLFE:
-
Ligustrum lucidum fruit extract
- LPS:
-
Lipopolysaccharide
- LSECs:
-
Liver sinusoidal endothelial cells
- miRNAs:
-
MicroRNAs
- MMPs:
-
Matrix metalloproteinases
- MRN:
-
Mre11-Rad50-Nbs1
- mRNA:
-
Messenger RNA
- NASH:
-
Non-alcoholic steatohepatitis
- ncRNAs:
-
Non-coding RNAs
- NCTD:
-
Norcantharidin
- NOXs:
-
NADPH oxidases
- NPs:
-
Nanoparticles
- NSCLC:
-
Non-small cell lung cancer
- OIS:
-
Oncogene-induced senescence
- OSIS:
-
Oxidative stress-induced senescence
- p38MAPK:
-
p38 mitogen-activated protein kinase
- PAB:
-
Pseudolaric acid B
- PCNA:
-
Proliferating cell nuclear antigen
- PD2:
-
Platycodin D2
- PGV-1:
-
Pentachloropropane-1
- PIP:
-
Phenyl-imidazole-ethylamine-platinum
- PM:
-
Particulate matter
- PRR:
-
Pattern recognition receptor
- PS:
-
Paracrine senescence
- qPCR:
-
Quantitative polymerase chain reaction
- RB:
-
Retinoblastoma protein
- ROS:
-
Reactive oxygen species
- RPA:
-
Replication protein A
- RS:
-
Replicative senescence
- S1P:
-
Sphingosine-1-phosphate
- SAHF:
-
Senescence-associated heterochromatin foci
- SASP:
-
Senescence-associated secretory phenotype
- SA-β-gal:
-
Senescence-associated β-galactosidase
- SBB:
-
Sudan Black B
- SIRT1:
-
Sirtuin 1
- SK2:
-
Sphingosine kinase 2
- Sp1:
-
Specificity Protein 1
- SSBs:
-
Single-strand breaks
- STING:
-
Stimulator of interferon genes
- SYT7:
-
Synaptotagmin-7
- TAC:
-
Telomerase activator compound
- TCM:
-
Traditional Chinese medicine
- TERC:
-
Telomerase RNA component
- TERT:
-
Telomerase reverse transcriptase
- TIS:
-
Therapy-induced senescence
- TME:
-
Tumor microenvironment
- TNBC:
-
Triple-negative breast cancer
- TOPBP1:
-
Topoisomerase-binding protein 1
- TPP1:
-
Telomere protection protein 1
- TRF2:
-
Telomeric repeat-binding factor 2
- UPM:
-
Urban particulate matter
- USP15:
-
Ubiquitin-specific protease 15
- ZnO:
-
Zinc oxide
References
Mathon NF, Lloyd AC. Cell senescence and cancer. Nat Rev Cancer. 2001;1(3):203–13.
He S, Sharpless NE. Senescence in Health and Disease. Cell. 2017;169(6):1000–11.
Saurat N, Minotti AP, Rahman MT, Sikder T, Zhang C, Cornacchia D, Jungverdorben J, Ciceri G, Betel D, Studer L. Genome-wide CRISPR screen identifies neddylation as a regulator of neuronal aging and AD neurodegeneration. Cell Stem Cell. 2024;31(8):1162-1174.e8.
Niu F, Li Z, Ren Y, Li Z, Guan H, Li Y, Zhang Y, Li Y, Yang J, Qian L, Shi W, Fan X, Li J, Shi L, Yu Y, Xiong Y. Aberrant hyper-expression of the RNA binding protein GIGYF2 in endothelial cells modulates vascular aging and function. Redox Biol. 2023;65:102824.
Huang W, Hickson LJ, Eirin A, Kirkland JL, Lerman LO. Cellular senescence: the good, the bad and the unknown. Nat Rev Nephrol. 2022;18(10):611–27.
DeLeo AB, Jay G, Appella E, Dubois GC, Law LW, Old LJ. Detection of a transformation-related antigen in chemically induced sarcomas and other transformed cells of the mouse. Proc Natl Acad Sci U S A. 1979;76(5):2420–4.
Hayflick L, Moorhead PS. The serial cultivation of human diploid cell strains. Exp Cell Res. 1961;25:585–621.
Ji S, Xiong M, Chen H, Liu Y, Zhou L, Hong Y, Wang M, Wang C, Fu X, Sun X. Cellular rejuvenation: molecular mechanisms and potential therapeutic interventions for diseases. Signal Transduct Target Ther. 2023;8(1):116.
Harley CB, Futcher AB, Greider CW. Telomeres shorten during ageing of human fibroblasts. Nature. 1990;345(6274):458–60.
Dimri GP, Lee X, Basile G, Acosta M, Scott G, Roskelley C, Medrano EE, Linskens M, Rubelj I, Pereira-Smith O, et al. A biomarker that identifies senescent human cells in culture and in aging skin in vivo. Proc Natl Acad Sci U S A. 1995;92(20):9363–7.
Chen Q, Fischer A, Reagan JD, Yan LJ, Ames BN. Oxidative DNA damage and senescence of human diploid fibroblast cells. Proc Natl Acad Sci U S A. 1995;92(10):4337–41.
Serrano M, Lin AW, McCurrach ME, Beach D, Lowe SW. Oncogenic ras provokes premature cell senescence associated with accumulation of p53 and p16INK4a. Cell. 1997;88(5):593–602.
Halazonetis TD, Gorgoulis VG, Bartek J. An oncogene-induced DNA damage model for cancer development. Science. 2008;319(5868):1352–5.
Chen Z, Trotman LC, Shaffer D, Lin HK, Dotan ZA, Niki M, Koutcher JA, Scher HI, Ludwig T, Gerald W, Cordon-Cardo C, Pandolfi PP. Crucial role of p53-dependent cellular senescence in suppression of Pten-deficient tumorigenesis. Nature. 2005;436(7051):725–30.
Kuilman T, Peeper DS. Senescence-messaging secretome: SMS-ing cellular stress. Nat Rev Cancer. 2009;9(2):81–94.
López-Otín C, Blasco MA, Partridge L, Serrano M, Kroemer G. The hallmarks of aging. Cell. 2013;153(6):1194–217.
Zhu Y, Tchkonia T, Pirtskhalava T, Gower AC, Ding H, Giorgadze N, Palmer AK, Ikeno Y, Hubbard GB, Lenburg M, O’Hara SP, LaRusso NF, Miller JD, Roos CM, Verzosa GC, LeBrasseur NK, Wren JD, Farr JN, Khosla S, Stout MB, McGowan SJ, Fuhrmann-Stroissnigg H, Gurkar AU, Zhao J, Colangelo D, Dorronsoro A, Ling YY, Barghouthy AS, Navarro DC, Sano T, Robbins PD, Niedernhofer LJ, Kirkland JL. The Achilles’ heel of senescent cells: from transcriptome to senolytic drugs. Aging Cell. 2015;14(4):644–58.
Galanos P, Vougas K, Walter D, Polyzos A, Maya-Mendoza A, Haagensen EJ, Kokkalis A, Roumelioti FM, Gagos S, Tzetis M, Canovas B, Igea A, Ahuja AK, Zellweger R, Havaki S, Kanavakis E, Kletsas D, Roninson IB, Garbis SD, Lopes M, Nebreda A, Thanos D, Blow JJ, Townsend P, Sørensen CS, Bartek J, Gorgoulis VG. Chronic p53-independent p21 expression causes genomic instability by deregulating replication licensing. Nat Cell Biol. 2016;18(7):777–89.
Li Y, Zhao H, Huang X, Tang J, Zhang S, Li Y, Liu X, He L, Ju Z, Lui KO, Zhou B. Embryonic senescent cells re-enter cell cycle and contribute to tissues after birth. Cell Res. 2018;28(7):775–8.
Short S, Fielder E, Miwa S, von Zglinicki T. Senolytics and senostatics as adjuvant tumour therapy. EBioMedicine. 2019;41:683–92.
Hanahan D. Hallmarks of Cancer: New Dimensions. Cancer Discov. 2022;12(1):31–46.
Abavisani M, Faraji S, Ebadpour N, Karav S, Sahebkar A. Beyond the Hayflick limit: How microbes influence cellular aging. Ageing Res Rev. 2025;104:102657.
Fernández-Duran I, Quintanilla A, Tarrats N, Birch J, Hari P, Millar FR, Lagnado AB, Smer-Barreto V, Muir M, Brunton VG, Passos JF, Acosta JC. Cytoplasmic innate immune sensing by the caspase-4 non-canonical inflammasome promotes cellular senescence. Cell Death Differ. 2022;29(6):1267–82.
Lee S, Yu Y, Trimpert J, Benthani F, Mairhofer M, Richter-Pechanska P, Wyler E, Belenki D, Kaltenbrunner S, Pammer M, Kausche L, Firsching TC, Dietert K, Schotsaert M, Martínez-Romero C, Singh G, Kunz S, Niemeyer D, Ghanem R, Salzer HJF, Paar C, Mülleder M, Uccellini M, Michaelis EG, Khan A, Lau A, Schönlein M, Habringer A, Tomasits J, Adler JM, Kimeswenger S, Gruber AD, Hoetzenecker W, Steinkellner H, Purfürst B, Motz R, Di Pierro F, Lamprecht B, Osterrieder N, Landthaler M, Drosten C, García-Sastre A, Langer R, Ralser M, Eils R, Reimann M, Fan DNY, Schmitt CA. Virus-induced senescence is a driver and therapeutic target in COVID-19. Nature. 2021;599(7884):283–9.
Evangelou K, Veroutis D, Paschalaki K, Foukas PG, Lagopati N, Dimitriou M, Papaspyropoulos A, Konda B, Hazapis O, Polyzou A, Havaki S, Kotsinas A, Kittas C, Tzioufas AG, de Leval L, Vassilakos D, Tsiodras S, Stripp BR, Papantonis A, Blandino G, Karakasiliotis I, Barnes PJ, Gorgoulis VG. Pulmonary infection by SARS-CoV-2 induces senescence accompanied by an inflammatory phenotype in severe COVID-19: possible implications for viral mutagenesis. Eur Respir J. 2022;60(2):2102951.
Gioia U, Tavella S, Martínez-Orellana P, Cicio G, Colliva A, Ceccon M, Cabrini M, Henriques AC, Fumagalli V, Paldino A, Presot E, Rajasekharan S, Iacomino N, Pisati F, Matti V, Sepe S, Conte MI, Barozzi S, Lavagnino Z, Carletti T, Volpe MC, Cavalcante P, Iannacone M, Rampazzo C, Bussani R, Tripodo C, Zacchigna S, Marcello A, d’Adda di Fagagna F. SARS-CoV-2 infection induces DNA damage, through CHK1 degradation and impaired 53BP1 recruitment, and cellular senescence. Nat Cell Biol. 2023;25(4):550–64.
Khanal S, Tang Q, Cao D, Zhao J, Nguyen LN, Oyedeji OS, Dang X, Nguyen LNT, Schank M, Thakuri BKC, Ogbu C, Morrison ZD, Wu XY, Zhang Z, He Q, El Gazzar M, Li Z, Ning S, Wang L, Moorman JP, Yao ZQ. Telomere and ATM Dynamics in CD4 T-Cell depletion in active and virus-suppressed HIV infections. J Virol. 2020;94(22):e01061-20.
Han J, Li W, Zhang J, Guan Y, Huang Y, Li X. Mechanism of circHIPK3-miRNA-124-3p/miRNA-148b-3p-Mediated Inflammatory Responses and Cell Senescence in Candida albicans-Induced Septic Acute Kidney Injury. Gerontology. 2022;68(10):1145–65.
Duan JL, Ruan B, Song P, Fang ZQ, Yue ZS, Liu JJ, Dou GR, Han H, Wang L. Shear stress-induced cellular senescence blunts liver regeneration through Notch-sirtuin 1–P21/P16 axis. Hepatology. 2022;75(3):584–99.
Guerrero-Navarro L, Jansen-Dürr P, Cavinato M. Synergistic interplay of UV radiation and urban particulate matter induces impairment of autophagy and alters cellular fate in senescence-prone human dermal fibroblasts. Aging Cell. 2024;23(4):e14086.
Hou T, Zhang J, Wang Y, Zhang G, Li S, Fan W, Li R, Sun Q, Liu C. Early pulmonary fibrosis-like changes in the setting of heat exposure: DNA damage and cell senescence. Int J Mol Sci. 2024;25(5):2992.
Chang-Chien J, Huang JL, Tsai HJ, Wang SL, Kuo ML, Yao TC. Particulate matter causes telomere shortening and increase in cellular senescence markers in human lung epithelial cells. Ecotoxicol Environ Saf. 2021;222:112484.
Yan Q, Zheng R, Li Y, Hu J, Gong M, Lin M, Xu X, Wu J, Sun S. PM(2.5)-induced premature senescence in HUVECs through the SIRT1/PGC-1α/SIRT3 pathway. Sci Total Environ. 2024;921:171177.
Deylam M, Alizadeh E, Sarikhani M, Hejazy M, Firouzamandi M. Zinc oxide nanoparticles promote the aging process in a size-dependent manner. J Mater Sci Mater Med. 2021;32(10):128.
Li Y, Li X, Cournoyer P, Choudhuri S, Yu X, Guo L, Chen S. Cannabidiol-induced transcriptomic changes and cellular senescence in human Sertoli cells. Toxicol Sci. 2023;191(2):227–38.
Yang MY, Lin PM, Liu YC, Hsiao HH, Yang WC, Hsu JF, Hsu CM, Lin SF. Induction of cellular senescence by doxorubicin is associated with upregulated miR-375 and induction of autophagy in K562 cells. PLoS ONE. 2012;7(5):e37205.
Chen F, Zhao W, Du C, Chen Z, Du J, Zhou M. Bleomycin induces senescence and repression of DNA repair via downregulation of Rad51. Mol Med. 2024;30(1):54.
Coimbra BM, Carvalho CM, van Zuiden M, Williamson RE, Ota VK, Mello AF, Belangero SI, Olff M, Mello MF. The impact of neighborhood context on telomere length: A systematic review. Health Place. 2022;74:102746.
O’Donovan A, Tomiyama AJ, Lin J, Puterman E, Adler NE, Kemeny M, Wolkowitz OM, Blackburn EH, Epel ES. Stress appraisals and cellular aging: a key role for anticipatory threat in the relationship between psychological stress and telomere length. Brain Behav Immun. 2012;26(4):573–9.
Chang SC, Crous-Bou M, Prescott J, Rosner B, Simon NM, Wang W, De Vivo I, Okereke OI. Relation of long-term patterns in caregiving activity and depressive symptoms to telomere length in older women. Psychoneuroendocrinology. 2018;89:161–7.
Galkin F, Kochetov K, Koldasbayeva D, Faria M, Fung HH, Chen AX, Zhavoronkov A. Psychological factors substantially contribute to biological aging: evidence from the aging rate in Chinese older adults. Aging (Albany NY). 2022;14(18):7206–22.
Blackburn EH, Gall JG. A tandemly repeated sequence at the termini of the extrachromosomal ribosomal RNA genes in Tetrahymena. J Mol Biol. 1978;120(1):33–53.
Allsopp RC, Vaziri H, Patterson C, Goldstein S, Younglai EV, Futcher AB, Greider CW, Harley CB. Telomere length predicts replicative capacity of human fibroblasts. Proc Natl Acad Sci U S A. 1992;89(21):10114–8.
Meng SS, Gu HW, Zhang T, Li YS, Tang HB. Gradual deterioration of fatty liver disease to liver cancer via inhibition of AMPK signaling pathways involved in energy-dependent disorders, cellular aging, and chronic inflammation. Front Oncol. 2023;13:1099624.
Hockemeyer D, Daniels JP, Takai H, de Lange T. Recent expansion of the telomeric complex in rodents: Two distinct POT1 proteins protect mouse telomeres. Cell. 2006;126(1):63–77.
Wang L, Chen R, Li G, Wang Z, Liu J, Liang Y, Liu JP. FBW7 Mediates Senescence and Pulmonary Fibrosis through Telomere Uncapping. Cell Metab. 2020;32(5):860-77.e9.
Lim CJ, Cech TR. Shaping human telomeres: from shelterin and CST complexes to telomeric chromatin organization. Nat Rev Mol Cell Biol. 2021;22(4):283–98.
Yang B, Chen Y, Shi J. Reactive Oxygen Species (ROS)-Based Nanomedicine. Chem Rev. 2019;119(8):4881–985.
Lennicke C, Cochemé HM. Redox metabolism: ROS as specific molecular regulators of cell signaling and function. Mol Cell. 2021;81(18):3691–707.
Martini H, Passos JF. Cellular senescence: all roads lead to mitochondria. Febs j. 2023;290(5):1186–202.
Wang K, Du Y, Li P, Guan C, Zhou M, Wu L, Liu Z, Huang Z. Nanoplastics causes heart aging/myocardial cell senescence through the Ca(2+)/mtDNA/cGAS-STING signaling cascade. J Nanobiotechnology. 2024;22(1):96.
He X, Jarrell ZR, Liang Y, Ryan Smith M, Orr ML, Marts L, Go YM, Jones DP. Vanadium pentoxide induced oxidative stress and cellular senescence in human lung fibroblasts. Redox Biol. 2022;55:102409.
Deng Q, Liao R, Wu BL, Sun P. High intensity ras signaling induces premature senescence by activating p38 pathway in primary human fibroblasts. J Biol Chem. 2004;279(2):1050–9.
Petti C, Molla A, Vegetti C, Ferrone S, Anichini A, Sensi M. Coexpression of NRASQ61R and BRAFV600E in human melanoma cells activates senescence and increases susceptibility to cell-mediated cytotoxicity. Cancer Res. 2006;66(13):6503–11.
Purhonen J, Banerjee R, Wanne V, Sipari N, Mörgelin M, Fellman V, Kallijärvi J. Mitochondrial complex III deficiency drives c-MYC overexpression and illicit cell cycle entry leading to senescence and segmental progeria. Nat Commun. 2023;14(1):2356.
Huang L, Guo Z, Wang F, Fu L. KRAS mutation: from undruggable to druggable in cancer. Signal Transduct Target Ther. 2021;6(1):386.
Yuan J, Dong X, Yap J, Hu J. The MAPK and AMPK signalings: interplay and implication in targeted cancer therapy. J Hematol Oncol. 2020;13(1):113.
Slomovitz BM, Coleman RL. The PI3K/AKT/mTOR pathway as a therapeutic target in endometrial cancer. Clin Cancer Res. 2012;18(21):5856–64.
Di Micco R, Fumagalli M, d’Adda di Fagagna F. Breaking news: high-speed race ends in arrest–how oncogenes induce senescence. Trends Cell Biol. 2007;17(11):529–36.
Bartolacci C, Andreani C, El-Gammal Y, Scaglioni PP. Lipid metabolism regulates oxidative stress and ferroptosis in RAS-Driven cancers: a perspective on cancer progression and therapy. Front Mol Biosci. 2021;8:706650.
Hole PS, Pearn L, Tonks AJ, James PE, Burnett AK, Darley RL, Tonks A. Ras-induced reactive oxygen species promote growth factor-independent proliferation in human CD34+ hematopoietic progenitor cells. Blood. 2010;115(6):1238–46.
Narita M, Nũnez S, Heard E, Narita M, Lin AW, Hearn SA, Spector DL, Hannon GJ, Lowe SW. Rb-mediated heterochromatin formation and silencing of E2F target genes during cellular senescence. Cell. 2003;113(6):703–16.
Tu Z, Zhuang X, Yao YG, Zhang R. BRG1 is required for formation of senescence-associated heterochromatin foci induced by oncogenic RAS or BRCA1 loss. Mol Cell Biol. 2013;33(9):1819–29.
Wang Z, Ma L, Su M, Zhou Y, Mao K, Li C, Peng G, Zhou C, Shen B, Dou J. Baicalin induces cellular senescence in human colon cancer cells via upregulation of DEPP and the activation of Ras/Raf/MEK/ERK signaling. Cell Death Dis. 2018;9(2):217.
Raabe EH, Lim KS, Kim JM, Meeker A, Mao XG, Nikkhah G, Maciaczyk J, Kahlert U, Jain D, Bar E, Cohen KJ, Eberhart CG. BRAF activation induces transformation and then senescence in human neural stem cells: a pilocytic astrocytoma model. Clin Cancer Res. 2011;17(11):3590–9.
Ohtani N. The roles and mechanisms of senescence-associated secretory phenotype (SASP): can it be controlled by senolysis? Inflammation and Regeneration. 2022;42(1):11.
Hao X, Zhao B, Towers M, Liao L, Monteiro EL, Xu X, Freeman C, Peng H, Tang HY, Havas A, Kossenkov AV, Berger SL, Adams PD, Speicher DW, Schultz D, Marmorstein R, Zaret KS, Zhang R. TXNRD1 drives the innate immune response in senescent cells with implications for age-associated inflammation. Nat Aging. 2024;4(2):185–97.
Li L, Yu R, Cai T, Chen Z, Lan M, Zou T, Wang B, Wang Q, Zhao Y, Cai Y. Effects of immune cells and cytokines on inflammation and immunosuppression in the tumor microenvironment. Int Immunopharmacol. 2020;88:106939.
Huynh J, Chand A, Gough D, Ernst M. Therapeutically exploiting STAT3 activity in cancer - using tissue repair as a road map. Nat Rev Cancer. 2019;19(2):82–96.
Cho U, Kim B, Kim S, Han Y, Song YS. Pro-inflammatory M1 macrophage enhances metastatic potential of ovarian cancer cells through NF-κB activation. Mol Carcinog. 2018;57(2):235–42.
Hou J, Chen KX, He C, Li XX, Huang M, Jiang YZ, Jiao YR, Xiao QN, He WZ, Liu L, Zou NY, Huang M, Wei J, Xiao Y, Yang M, Luo XH, Zeng C, Lei GH, Li CJ. Aged bone marrow macrophages drive systemic aging and age-related dysfunction via extracellular vesicle-mediated induction of paracrine senescence. Nat Aging. 2024;4(11):1562–81.
Chou LY, Ho CT, Hung SC. Paracrine senescence of mesenchymal stromal cells involves inflammatory cytokines and the NF-κB pathway. Cells. 2022;11(20):3324.
Li Q, Qian W, Zhang Y, Hu L, Chen S, Xia Y. A new wave of innovations within the DNA damage response. Signal Transduct Target Ther. 2023;8(1):338.
Petr MA, Tulika T, Carmona-Marin LM, Scheibye-Knudsen M. Protecting the Aging Genome. Trends Cell Biol. 2020;30(2):117–32.
Casari E, Gnugnoli M, Rinaldi C, Pizzul P, Colombo CV, Bonetti D, Longhese MP. To fix or not to fix: maintenance of chromosome ends versus repair of DNA double-strand breaks. Cells. 2022;11(20):3224.
Campisi J. Replicative senescence: an old lives’ tale? Cell. 1996;84(4):497–500.
Deng Y, Chang S. Role of telomeres and telomerase in genomic instability, senescence and cancer. Lab Invest. 2007;87(11):1071–6.
Sullivan DI, Bello FM, Silva AG, Redding KM, Giordano L, Hinchie AM, Loughridge KE, Mora AL, Königshoff M, Kaufman BA, Jurczak MJ, Alder JK. Intact mitochondrial function in the setting of telomere-induced senescence. Aging Cell. 2023;22(10):e13941.
Cao X, Fang L, Jiang Y, Zeng T, Bai S, Li S, Liu Y, Zhong W, Lu C, Yang H. Nanoscale octopus guiding telomere entanglement: An innovative strategy for inducing apoptosis in cancer cells. Biomaterials. 2025;313:122777.
Xiao X, Xu M, Yu H, Wang L, Li X, Rak J, Wang S, Zhao RC. Mesenchymal stem cell-derived small extracellular vesicles mitigate oxidative stress-induced senescence in endothelial cells via regulation of miR-146a/Src. Signal Transduct Target Ther. 2021;6(1):354.
Di Micco R, Fumagalli M, Cicalese A, Piccinin S, Gasparini P, Luise C, Schurra C, Garre M, Nuciforo PG, Bensimon A, Maestro R, Pelicci PG, d’Adda di Fagagna F. Oncogene-induced senescence is a DNA damage response triggered by DNA hyper-replication. Nature. 2006;444(7119):638–42.
Roninson IB. Tumor cell senescence in cancer treatment. Cancer Res. 2003;63(11):2705–15.
Cimprich KA, Cortez D. ATR: an essential regulator of genome integrity. Nat Rev Mol Cell Biol. 2008;9(8):616–27.
Furnari B, Rhind N, Russell P. Cdc25 mitotic inducer targeted by chk1 DNA damage checkpoint kinase. Science. 1997;277(5331):1495–7.
Gralewska P, Gajek A, Marczak A, Rogalska A. Participation of the ATR/CHK1 pathway in replicative stress targeted therapy of high-grade ovarian cancer. J Hematol Oncol. 2020;13(1):39.
Maréchal A, Zou L. DNA damage sensing by the ATM and ATR kinases. Cold Spring Harb Perspect Biol. 2013;5(9):a012716.
Tatsukawa K, Sakamoto R, Kawasoe Y, Kubota Y, Tsurimoto T, Takahashi TS, Ohashi E. Resection of DNA double-strand breaks activates Mre11-Rad50-Nbs1- and Rad9-Hus1-Rad1-dependent mechanisms that redundantly promote ATR checkpoint activation and end processing in Xenopus egg extracts. Nucleic Acids Res. 2024;52(6):3146–63.
Lee JH, Paull TT. Cellular functions of the protein kinase ATM and their relevance to human disease. Nat Rev Mol Cell Biol. 2021;22(12):796–814.
Hoffmann I. The role of Cdc25 phosphatases in cell cycle checkpoints. Protoplasma. 2000;211(1):8–11.
Ray D, Kiyokawa H. CDC25A Phosphatase: a Rate-Limiting Oncogene That Determines Genomic Stability. Can Res. 2008;68(5):1251–3.
Stolz A, Ertych N, Bastians H. Tumor suppressor CHK2: regulator of DNA damage response and mediator of chromosomal stability. Clin Cancer Res. 2011;17(3):401–5.
Cheng Q, Chen L, Li Z, Lane WS, Chen J. ATM activates p53 by regulating MDM2 oligomerization and E3 processivity. Embo j. 2009;28(24):3857–67.
Yan P, Li Z, Xiong J, Geng Z, Wei W, Zhang Y, Wu G, Zhuang T, Tian X, Liu Z, Liu J, Sun K, Chen F, Zhang Y, Zeng C, Huang Y, Zhang B. LARP7 ameliorates cellular senescence and aging by allosterically enhancing SIRT1 deacetylase activity. Cell Rep. 2021;37(8):110038.
Fotedar R, Fitzgerald P, Rousselle T, Cannella D, Dorée M, Messier H, Fotedar A. p21 contains independent binding sites for cyclin and cdk2: both sites are required to inhibit cdk2 kinase activity. Oncogene. 1996;12(10):2155–64.
Giacinti C, Giordano A. RB and cell cycle progression. Oncogene. 2006;25(38):5220–7.
Olofsson BA, Kelly CM, Kim J, Hornsby SM, Azizkhan-Clifford J. Phosphorylation of Sp1 in Response to DNA Damage by Ataxia Telangiectasia-Mutated Kinase. Mol Cancer Res. 2007;5(12):1319–30.
Wang X, Feng Y, Pan L, Wang Y, Xu X, Lu J, Huang B. The proximal GC-rich region of p16(INK4a) gene promoter plays a role in its transcriptional regulation. Mol Cell Biochem. 2007;301(1–2):259–66.
Safwan-Zaiter H, Wagner N, Wagner KD. P16INK4A-more than a senescence marker. Life (Basel). 2022;12(9):1332.
Bandyopadhyay D, Medrano EE. Melanin accumulation accelerates melanocyte senescence by a mechanism involving p16INK4a/CDK4/pRB and E2F1. Ann N Y Acad Sci. 2000;908:71–84.
Ohtani N, Zebedee Z, Huot TJ, Stinson JA, Sugimoto M, Ohashi Y, Sharrocks AD, Peters G, Hara E. Opposing effects of Ets and Id proteins on p16INK4a expression during cellular senescence. Nature. 2001;409(6823):1067–70.
Acosta JC, Banito A, Wuestefeld T, Georgilis A, Janich P, Morton JP, Athineos D, Kang TW, Lasitschka F, Andrulis M, Pascual G, Morris KJ, Khan S, Jin H, Dharmalingam G, Snijders AP, Carroll T, Capper D, Pritchard C, Inman GJ, Longerich T, Sansom OJ, Benitah SA, Zender L, Gil J. A complex secretory program orchestrated by the inflammasome controls paracrine senescence. Nat Cell Biol. 2013;15(8):978–90.
Hannon GJ, Beach D. p15INK4B is a potential effector of TGF-beta-induced cell cycle arrest. Nature. 1994;371(6494):257–61.
Liu WT, Huang KY, Lu MC, Huang HL, Chen CY, Cheng YL, Yu HC, Liu SQ, Lai NS, Huang HB. TGF-β upregulates the translation of USP15 via the PI3K/AKT pathway to promote p53 stability. Oncogene. 2017;36(19):2715–23.
Bhunia AK, Piontek K, Boletta A, Liu L, Qian F, Xu PN, Germino FJ, Germino GG. PKD1 induces p21(waf1) and regulation of the cell cycle via direct activation of the JAK-STAT signaling pathway in a process requiring PKD2. Cell. 2002;109(2):157–68.
Joung J, Heo Y, Kim Y, Kim J, Choi H, Jeon T, Jang Y, Kim EJ, Lee SH, Suh JM, Elledge SJ, Kim MS, Kang C. Cell enlargement modulated by GATA4 and YAP instructs the senescence-associated secretory phenotype. Nat Commun. 2025;16(1):1696.
Tsai CH, Chang CY, Lin BZ, Wu YL, Wu MH, Lin LT, Huang WC, Holz JD, Sheu TJ, Lee JS, Kitsis RN, Tai PH, Lee YJ. Up-regulation of cofilin-1 in cell senescence associates with morphological change and p27(kip1) -mediated growth delay. Aging Cell. 2021;20(1):e13288.
Evangelou K, Gorgoulis VG. Sudan Black B, The Specific Histochemical Stain for Lipofuscin: A Novel Method to Detect Senescent Cells. In: Nikiforov MA, editor. Oncogene-Induced Senescence: Methods and Protocols. Springer, New York: New York, NY; 2017. p. 111–9.
Magkouta S, Veroutis D, Pousias A, Papaspyropoulos A, Pippa N, Lougiakis N, Kambas K, Lagopati N, Polyzou A, Georgiou M, Chountoulesi M, Pispas S, Foutadakis S, Pouli N, Marakos P, Kotsinas A, Verginis P, Valakos D, Mizi A, Papantonis A, Vatsellas G, Galanos P, Bartek J, Petty R, Serrano M, Thanos D, Roussos C, Demaria M, Evangelou K, Gorgoulis VG. A fluorophore-conjugated reagent enabling rapid detection, isolation and live tracking of senescent cells. Mol Cell. 2023;83(19):3558-73.e7.
Ogrodnik M, Carlos Acosta J, Adams PD, d’Adda di Fagagna F, Baker DJ, Bishop CL, Chandra T, Collado M, Gil J, Gorgoulis V, Gruber F, Hara E, Jansen-Dürr P, Jurk D, Khosla S, Kirkland JL, Krizhanovsky V, Minamino T, Niedernhofer LJ, Passos JF, Ring NAR, Redl H, Robbins PD, Rodier F, Scharffetter-Kochanek K, Sedivy JM, Sikora E, Witwer K, von Zglinicki T, Yun MH, Grillari J, Demaria M. Guidelines for minimal information on cellular senescence experimentation in vivo. Cell. 2024;187(16):4150–75.
Evangelou K, Lougiakis N, Rizou SV, Kotsinas A, Kletsas D, Muñoz-Espín D, Kastrinakis NG, Pouli N, Marakos P, Townsend P, Serrano M, Bartek J, Gorgoulis VG. Robust, universal biomarker assay to detect senescent cells in biological specimens. Aging Cell. 2017;16(1):192–7.
Gorgoulis V, Adams PD, Alimonti A, Bennett DC, Bischof O, Bishop C, Campisi J, Collado M, Evangelou K, Ferbeyre G, Gil J, Hara E, Krizhanovsky V, Jurk D, Maier AB, Narita M, Niedernhofer L, Passos JF, Robbins PD, Schmitt CA, Sedivy J, Vougas K, von Zglinicki T, Zhou D, Serrano M, Demaria M. Cellular Senescence: Defining a Path Forward. Cell. 2019;179(4):813–27.
Belhadj J, Surina S, Hengstschläger M, Lomakin AJ. Form follows function: Nuclear morphology as a quantifiable predictor of cellular senescence. Aging Cell. 2023;22(12):e14012.
Pathak RU, Soujanya M, Mishra RK. Deterioration of nuclear morphology and architecture: A hallmark of senescence and aging. Ageing Res Rev. 2021;67:101264.
Di Micco R, Sulli G, Dobreva M, Liontos M, Botrugno OA, Gargiulo G, dal Zuffo R, Matti V, d’Ario G, Montani E, Mercurio C, Hahn WC, Gorgoulis V, Minucci S, d’Adda di Fagagna F. Interplay between oncogene-induced DNA damage response and heterochromatin in senescence and cancer. Nat Cell Biol. 2011;13(3):292–302.
Zhao L, Zhang Y, Gao Y, Geng P, Lu Y, Liu X, Yao R, Hou P, Liu D, Lu J, Huang B. JMJD3 promotes SAHF formation in senescent WI38 cells by triggering an interplay between demethylation and phosphorylation of RB protein. Cell Death Differ. 2015;22(10):1630–40.
Bai J, Chen J, Ma M, Cai M, Xu F, Wang G, Tao K, Shuai X. Inhibiting enhancer of zeste homolog 2 promotes cellular senescence in gastric cancer cells SGC-7901 by activation of p21 and p16. DNA Cell Biol. 2014;33(6):337–44.
Abbas T, Dutta A. p21 in cancer: intricate networks and multiple activities. Nat Rev Cancer. 2009;9(6):400–14.
Zhang X, Habiballa L, Aversa Z, Ng YE, Sakamoto AE, Englund DA, Pearsall VM, White TA, Robinson MM, Rivas DA, Dasari S, Hruby AJ, Lagnado AB, Jachim SK, Granic A, Sayer AA, Jurk D, Lanza IR, Khosla S, Fielding RA, Nair KS, Schafer MJ, Passos JF, LeBrasseur NK. Characterization of cellular senescence in aging skeletal muscle. Nat Aging. 2022;2(7):601–15.
Li X, Chen M, Chen X, He X, Li X, Wei H, Tan Y, Min J, Azam T, Xue M, Zhang Y, Dong M, Yin Q, Zheng L, Jiang H, Huo D, Wang X, Chen S, Ji Y, Chen H. TRAP1 drives smooth muscle cell senescence and promotes atherosclerosis via HDAC3-primed histone H4 lysine 12 lactylation. Eur Heart J. 2024;45(39):4219–35.
Byrns CN, Perlegos AE, Miller KN, Jin Z, Carranza FR, Manchandra P, Beveridge CH, Randolph CE, Chaluvadi VS, Zhang SL, Srinivasan AR, Bennett FC, Sehgal A, Adams PD, Chopra G, Bonini NM. Senescent glia link mitochondrial dysfunction and lipid accumulation. Nature. 2024;630(8016):475–83.
Liang Y, Pan C, Yin T, Wang L, Gao X, Wang E, Quang H, Huang D, Tan L, Xiang K, Wang Y, Alexander PB, Li QJ, Yao TP, Zhang Z, Wang XF. Branched-Chain Amino Acid Accumulation Fuels the Senescence-Associated Secretory Phenotype. Adv Sci (Weinh). 2024;11(2):e2303489.
Monroe TB, Hertzel AV, Dickey DM, Hagen T, Santibanez SV, Berdaweel IA, Halley C, Puchalska P, Anderson EJ, Camell CD, Robbins PD, Bernlohr DA. Lipid peroxidation products induce carbonyl stress, mitochondrial dysfunction, and cellular senescence in human and murine cells. Aging Cell. 2025;24(1):e14367.
Juers DH, Matthews BW, Huber RE. LacZ β-galactosidase: structure and function of an enzyme of historical and molecular biological importance. Protein Sci. 2012;21(12):1792–807.
Feng B, Chu F, Bi A, Huang X, Fang Y, Liu M, Chen F, Li Y, Zeng W. Fidelity-oriented fluorescence imaging probes for beta-galactosidase: From accurate diagnosis to precise treatment. Biotechnol Adv. 2023;68:108244.
Lozano-Torres B, Blandez JF, Sancenón F, Martínez-Máñez R. Chromo-fluorogenic probes for β-galactosidase detection. Anal Bioanal Chem. 2021;413(9):2361–88.
Coppé JP, Patil CK, Rodier F, Sun Y, Muñoz DP, Goldstein J, Nelson PS, Desprez PY, Campisi J. Senescence-associated secretory phenotypes reveal cell-nonautonomous functions of oncogenic RAS and the p53 tumor suppressor. PLoS Biol. 2008;6(12):2853–68.
Freund A, Patil CK, Campisi J. p38MAPK is a novel DNA damage response-independent regulator of the senescence-associated secretory phenotype. Embo j. 2011;30(8):1536–48.
Glück S, Guey B, Gulen MF, Wolter K, Kang TW, Schmacke NA, Bridgeman A, Rehwinkel J, Zender L, Ablasser A. Innate immune sensing of cytosolic chromatin fragments through cGAS promotes senescence. Nat Cell Biol. 2017;19(9):1061–70.
Rodier F, Coppé JP, Patil CK, Hoeijmakers WA, Muñoz DP, Raza SR, Freund A, Campeau E, Davalos AR, Campisi J. Persistent DNA damage signalling triggers senescence-associated inflammatory cytokine secretion. Nat Cell Biol. 2009;11(8):973–9.
Ritschka B, Storer M, Mas A, Heinzmann F, Ortells MC, Morton JP, Sansom OJ, Zender L, Keyes WM. The senescence-associated secretory phenotype induces cellular plasticity and tissue regeneration. Genes Dev. 2017;31(2):172–83.
Li X, Li C, Zhang W, Wang Y, Qian P, Huang H. Inflammation and aging: signaling pathways and intervention therapies. Signal Transduct Target Ther. 2023;8(1):239.
Li Y, Kračun D, Dustin CM, El Massry M, Yuan S, Goossen CJ, DeVallance ER, Sahoo S, St Hilaire C, Gurkar AU, Finkel T, Straub AC, Cifuentes-Pagano E, Pagano PJ. Forestalling age-impaired angiogenesis and blood flow by targeting NOX: Interplay of NOX1, IL-6, and SASP in propagating cell senescence. Proc Natl Acad Sci U S A. 2021;118(42):e2015666118.
Suryadevara V, Hudgins AD, Rajesh A, Pappalardo A, Karpova A, Dey AK, Hertzel A, Agudelo A, Rocha A, Soygur B, Schilling B, Carver CM, Aguayo-Mazzucato C, Baker DJ, Bernlohr DA, Jurk D, Mangarova DB, Quardokus EM, Enninga EAL, Schmidt EL, Chen F, Duncan FE, Cambuli F, Kaur G, Kuchel GA, Lee G, Daldrup-Link HE, Martini H, Phatnani H, Al-Naggar IM, Rahman I, Nie J, Passos JF, Silverstein JC, Campisi J, Wang J, Iwasaki K, Barbosa K, Metis K, Nernekli K, Niedernhofer LJ, Ding L, Wang L, Adams LC, Ruiyang L, Doolittle ML, Teneche MG, Schafer MJ, Xu M, Hajipour M, Boroumand M, Basisty N, Sloan N, Slavov N, Kuksenko O, Robson P, Gomez PT, Vasilikos P, Adams PD, Carapeto P, Zhu Q, Ramasamy R, Perez-Lorenzo R, Fan R, Dong R, Montgomery RR, Shaikh S, Vickovic S, Yin S, Kang S, Suvakov S, Khosla S, Garovic VD, Menon V, Xu Y, Song Y, Suh Y, Dou Z, Neretti N. SenNet recommendations for detecting senescent cells in different tissues. Nat Rev Mol Cell Biol. 2024;25(12):1001–23.
Felgentreff K, Baumann U, Klemann C, Schuetz C, Viemann D, Wetzke M, Pannicke U, von Hardenberg S, Auber B, Debatin KM, Jacobsen EM, Hoenig M, Schulz A, Schwarz K. Biomarkers of DNA Damage Response Enable Flow Cytometry-Based Diagnostic to Identify Inborn DNA Repair Defects in Primary Immunodeficiencies. J Clin Immunol. 2022;42(2):286–98.
Furia L, Pelicci S, Scanarini M, Pelicci PG, Faretta M. From double-strand break recognition to cell-cycle checkpoint activation: high content and resolution image cytometry unmasks 53BP1 multiple roles in DNA damage response and p53 action. Int J Mol Sci. 2022;23(17):10193.
Chen J, Crutchley J, Zhang D, Owzar K, Kastan MB. Identification of a DNA Damage-Induced Alternative Splicing Pathway That Regulates p53 and Cellular Senescence Markers. Cancer Discov. 2017;7(7):766–81.
Sun Y, You Y, Wu Q, Hu R, Dai K. Senescence-targeted MicroRNA/Organoid composite hydrogel repair cartilage defect and prevention joint degeneration via improved chondrocyte homeostasis. Bioact Mater. 2024;39:427–42.
Yang K, Fu K, Zhang H, Wang X, To KKW, Yang C, Wang F, Chen ZS, Fu L. PBA2, a novel inhibitor of the β-catenin/CBP pathway, eradicates chronic myeloid leukemia including BCR-ABL T315I mutation. Mol Cancer. 2024;23(1):209.
Chan ASL, Zhu H, Narita M, Cassidy LD, Young ARJ, Bermejo-Rodriguez C, Janowska AT, Chen HC, Gough S, Oshimori N, Zender L, Aitken SJ, Hoare M, Narita M. Titration of RAS alters senescent state and influences tumour initiation. Nature. 2024;633(8030):678–85.
Born E, Lipskaia L, Breau M, Houssaini A, Beaulieu D, Marcos E, Pierre R, Do Cruzeiro M, Lefevre M, Derumeaux G, Bulavin DV, Delcroix M, Quarck R, Reen V, Gil J, Bernard D, Flaman JM, Adnot S, Abid S. Eliminating Senescent Cells Can Promote Pulmonary Hypertension Development and Progression. Circulation. 2023;147(8):650–66.
Shim HS, Iaconelli J, Shang X, Li J, Lan ZD, Jiang S, Nutsch K, Beyer BA, Lairson LL, Boutin AT, Bollong MJ, Schultz PG, DePinho RA. TERT activation targets DNA methylation and multiple aging hallmarks. Cell. 2024;187(15):4030-42.e13.
Majewska J, Agrawal A, Mayo A, Roitman L, Chatterjee R, Sekeresova Kralova J, Landsberger T, Katzenelenbogen Y, Meir-Salame T, Hagai E, Sopher I, Perez-Correa JF, Wagner W, Maimon A, Amit I, Alon U, Krizhanovsky V. p16-dependent increase of PD-L1 stability regulates immunosurveillance of senescent cells. Nat Cell Biol. 2024;26(8):1336–45.
Guan L, Crasta KC, Maier AB. Assessment of cell cycle regulators in human peripheral blood cells as markers of cellular senescence. Ageing Res Rev. 2022;78:101634.
Zhang D, Zhang JW, Xu H, Chen X, Gao Y, Jiang HG, Wang Y, Wu H, Yang L, Wang WB, Dai J, Xia L, Peng J, Zhou FX. Therapy-induced senescent tumor cell-derived extracellular vesicles promote colorectal cancer progression through SERPINE1-mediated NF-κB p65 nuclear translocation. Mol Cancer. 2024;23(1):70.
Van Nguyen T, Puebla-Osorio N, Pang H, Dujka ME, Zhu C. DNA damage-induced cellular senescence is sufficient to suppress tumorigenesis: a mouse model. J Exp Med. 2007;204(6):1453–61.
Wiman KG. Pharmacological reactivation of mutant p53: from protein structure to the cancer patient. Oncogene. 2010;29(30):4245–52.
Murray A, Gough G, Cindrić A, Vučković F, Koschut D, Borelli V, Petrović DJ, Bekavac A, Plećaš A, Hribljan V, Brunmeir R, Jurić J, Pučić-Baković M, Slana A, Deriš H, Frkatović A, Groet J, O’Brien NL, Chen HY, Yeap YJ, Delom F, Havlicek S, Gammon L, Hamburg S, Startin C, D’Souza H, Mitrečić D, Kero M, Odak L, Krušlin B, Krsnik Ž, Kostović I, Foo JN, Loh YH, Dunn NR, de la Luna S, Spector T, Barišić I, Thomas MSC, Strydom A, Franceschi C, Lauc G, Krištić J, Alić I, Nižetić D. Dose imbalance of DYRK1A kinase causes systemic progeroid status in Down syndrome by increasing the un-repaired DNA damage and reducing LaminB1 levels. EBioMedicine. 2023;94:104692.
Ma M, Zeng J, Zhu M, Li H, Lin T, Yang H, Wei X, Song T. Human umbilical cord mesenchymal stem cells derived extracellular vesicles ameliorate kidney ischemia-reperfusion injury by suppression of senescent tubular epithelial cells - experimental study. Int J Surg. 2024;111(1):394–410.
Yue S, Zheng X, Zheng Y. Cell-type-specific role of lamin-B1 in thymus development and its inflammation-driven reduction in thymus aging. Aging Cell. 2019;18(4):e12952.
Indovina P, Marcelli E, Casini N, Rizzo V, Giordano A. Emerging roles of RB family: new defense mechanisms against tumor progression. J Cell Physiol. 2013;228(3):525–35.
Thangavel C, Boopathi E, Liu Y, McNair C, Haber A, Perepelyuk M, Bhardwaj A, Addya S, Ertel A, Shoyele S, Birbe R, Salvino JM, Dicker AP, Knudsen KE, Den RB. Therapeutic Challenge with a CDK 4/6 Inhibitor Induces an RB-Dependent SMAC-Mediated Apoptotic Response in Non-Small Cell Lung Cancer. Clin Cancer Res. 2018;24(6):1402–14.
Zoumpoulidou G, Broceño C, Li H, Bird D, Thomas G, Mittnacht S. Role of the tripartite motif protein 27 in cancer development. J Natl Cancer Inst. 2012;104(12):941–52.
Nader GPF, Agüera-Gonzalez S, Routet F, Gratia M, Maurin M, Cancila V, Cadart C, Palamidessi A, Ramos RN, San Roman M, Gentili M, Yamada A, Williart A, Lodillinsky C, Lagoutte E, Villard C, Viovy JL, Tripodo C, Galon J, Scita G, Manel N, Chavrier P, Piel M. Compromised nuclear envelope integrity drives TREX1-dependent DNA damage and tumor cell invasion. Cell. 2021;184(20):5230-46.e22.
Ghodke I, Remisova M, Furst A, Kilic S, Reina-San-Martin B, Poetsch AR, Altmeyer M, Soutoglou E. AHNAK controls 53BP1-mediated p53 response by restraining 53BP1 oligomerization and phase separation. Mol Cell. 2021;81(12):2596-610.e7.
Manohar S, Estrada ME, Uliana F, Vuina K, Alvarez PM, de Bruin RAM, Neurohr GE. Genome homeostasis defects drive enlarged cells into senescence. Mol Cell. 2023;83(22):4032-46.e6.
Karimian K, Groot A, Huso V, Kahidi R, Tan KT, Sholes S, Keener R, McDyer JF, Alder JK, Li H, Rechtsteiner A, Greider CW. Human telomere length is chromosome end-specific and conserved across individuals. Science. 2024;384(6695):533–9.
Schmidt TT, Tyer C, Rughani P, Haggblom C, Jones JR, Dai X, Frazer KA, Gage FH, Juul S, Hickey S, Karlseder J. High resolution long-read telomere sequencing reveals dynamic mechanisms in aging and cancer. Nat Commun. 2024;15(1):5149.
Sanchez SE, Gu Y, Wang Y, Golla A, Martin A, Shomali W, Hockemeyer D, Savage SA, Artandi SE. Digital telomere measurement by long-read sequencing distinguishes healthy aging from disease. Nat Commun. 2024;15(1):5148.
Muthamil S, Kim HY, Jang HJ, Lyu JH, Shin UC, Go Y, Park SH, Lee HG, Park JH. Biomarkers of Cellular Senescence and Aging: Current State-of-the-Art, Challenges and Future Perspectives. Adv Biol (Weinh). 2024;8(9):e2400079.
Sati S, Bonev B, Szabo Q, Jost D, Bensadoun P, Serra F, Loubiere V, Papadopoulos GL, Rivera-Mulia JC, Fritsch L, Bouret P, Castillo D, Gelpi JL, Orozco M, Vaillant C, Pellestor F, Bantignies F, Marti-Renom MA, Gilbert DM, Lemaitre JM, Cavalli G. 4D Genome Rewiring during Oncogene-Induced and Replicative Senescence. Mol Cell. 2020;78(3):522-38.e9.
Narita M, Narita M, Krizhanovsky V, Nuñez S, Chicas A, Hearn SA, Myers MP, Lowe SW. A novel role for high-mobility group a proteins in cellular senescence and heterochromatin formation. Cell. 2006;126(3):503–14.
Mosteiro L, Pantoja C, Alcazar N, Marión RM, Chondronasiou D, Rovira M, Fernandez-Marcos PJ, Muñoz-Martin M, Blanco-Aparicio C, Pastor J, Gómez-López G, De Martino A, Blasco MA, Abad M, Serrano M. Tissue damage and senescence provide critical signals for cellular reprogramming in vivo. Science. 2016;354(6315):aaf4445.
Fang C, Du L, Gao S, Chen Y, Chen Z, Wu Z, Li L, Li J, Zeng X, Li M, Li Y, Tian X. Association between premature vascular smooth muscle cells senescence and vascular inflammation in Takayasu’s arteritis. Ann Rheum Dis. 2024;83(11):1522–35.
Veroutis D, Argyropoulou OD, Goules AV, Kambas K, Palamidas DA, Evangelou K, Havaki S, Polyzou A, Valakos D, Xingi E, Karatza E, Boki KA, Cavazza A, Kittas C, Thanos D, Ricordi C, Marvisi C, Muratore F, Galli E, Croci S, Salvarani C, Gorgoulis VG, Tzioufas AG. Senescent cells in giant cell arteritis display an inflammatory phenotype participating in tissue injury via IL-6-dependent pathways. Ann Rheum Dis. 2024;83(3):342–50.
Majdinasab M, Lamy de la Chapelle M, Marty JL. Recent Progresses in Optical Biosensors for Interleukin 6 Detection. Biosensors (Basel). 2023;13(9):898.
Fan G, Yu B, Tang L, Zhu R, Chen J, Zhu Y, Huang H, Zhou L, Liu J, Wang W, Tao Z, Zhang F, Yu S, Lu X, Cao Y, Du S, Li H, Li J, Zhang J, Ren H, Gires O, Liu H, Wang X, Qin J, Wang H. TSPAN8(+) myofibroblastic cancer-associated fibroblasts promote chemoresistance in patients with breast cancer. Sci Transl Med. 2024;16(741):eadj5705.
Wei L, Yang X, Wang J, Wang Z, Wang Q, Ding Y, Yu A. H3K18 lactylation of senescent microglia potentiates brain aging and Alzheimer’s disease through the NFκB signaling pathway. J Neuroinflammation. 2023;20(1):208.
Tong M, Kayani T, Jones DM, Salmon JE, Whirledge S, Chamley LW, Abrahams VM. Antiphospholipid Antibodies Increase Endometrial Stromal Cell Decidualization, Senescence, and Inflammation via Toll-like Receptor 4, Reactive Oxygen Species, and p38 MAPK Signaling. Arthritis Rheumatol. 2022;74(6):1001–12.
Carver CM, Rodriguez SL, Atkinson EJ, Dosch AJ, Asmussen NC, Gomez PT, Leitschuh EA, Espindola-Netto JM, Jeganathan KB, Whaley MG, Kamenecka TM, Baker DJ, Haak AJ, LeBrasseur NK, Schafer MJ. IL-23R is a senescence-linked circulating and tissue biomarker of aging. Nat Aging. 2024;5(2):291–305.
Reimann M, Lee S, Schmitt CA. Cellular senescence: Neither irreversible nor reversible. J Exp Med. 2024;221(4):e20232136.
Macedo-Silva C, Miranda-Gonçalves V, Tavares NT, Barros-Silva D, Lencart J, Lobo J, Oliveira Â, Correia MP, Altucci L, Jerónimo C. Epigenetic regulation of TP53 is involved in prostate cancer radioresistance and DNA damage response signaling. Signal Transduct Target Ther. 2023;8(1):395.
Wang YH, Ho TLF, Hariharan A, Goh HC, Wong YL, Verkaik NS, Lee MY, Tam WL, van Gent DC, Venkitaraman AR, Sheetz MP, Lane DP. Rapid recruitment of p53 to DNA damage sites directs DNA repair choice and integrity. Proc Natl Acad Sci U S A. 2022;119(10):e2113233119.
Zampetidis CP, Galanos P, Angelopoulou A, Zhu Y, Polyzou A, Karamitros T, Kotsinas A, Lagopati N, Mourkioti I, Mirzazadeh R, Polyzos A, Garnerone S, Mizi A, Gusmao EG, Sofiadis K, Gál Z, Larsen DH, Pefani DE, Demaria M, Tsirigos A, Crosetto N, Maya-Mendoza A, Papaspyropoulos A, Evangelou K, Bartek J, Papantonis A, Gorgoulis VG. A recurrent chromosomal inversion suffices for driving escape from oncogene-induced senescence via subTAD reorganization. Mol Cell. 2021;81(23):4907-23.e8.
Milanovic M, Fan DNY, Belenki D, Däbritz JHM, Zhao Z, Yu Y, Dörr JR, Dimitrova L, Lenze D, Monteiro Barbosa IA, Mendoza-Parra MA, Kanashova T, Metzner M, Pardon K, Reimann M, Trumpp A, Dörken B, Zuber J, Gronemeyer H, Hummel M, Dittmar G, Lee S, Schmitt CA. Senescence-associated reprogramming promotes cancer stemness. Nature. 2018;553(7686):96–100.
Angelopoulou A, Theocharous G, Valakos D, Polyzou A, Magkouta S, Myrianthopoulos V, Havaki S, Fiorillo M, Tremi I, Vachlas K, Nisotakis T, Thanos DF, Pantazaki A, Kletsas D, Bartek J, Petty R, Thanos D, McCrimmon RJ, Papaspyropoulos A, Gorgoulis VG. Loss of the tumour suppressor LKB1/STK11 uncovers a leptin-mediated sensitivity mechanism to mitochondrial uncouplers for targeted cancer therapy. Mol Cancer. 2024;23(1):147.
Cohn RL, Gasek NS, Kuchel GA, Xu M. The heterogeneity of cellular senescence: insights at the single-cell level. Trends Cell Biol. 2023;33(1):9–17.
Zhang Z, Schaefer C, Jiang W, Lu Z, Lee J, Sziraki A, Abdulraouf A, Wick B, Haeussler M, Li Z, Molla G, Satija R, Zhou W, Cao J. A panoramic view of cell population dynamics in mammalian aging. Science. 2025;387(6731):eadn3949.
Kwiatkowska KM, Mavrogonatou E, Papadopoulou A, Sala C, Calzari L, Gentilini D, Bacalini MG, Dall’Olio D, Castellani G, Ravaioli F, Franceschi C, Garagnani P, Pirazzini C, Kletsas D. Heterogeneity of cellular senescence: cell type-specific and senescence stimulus-dependent epigenetic alterations. Cells. 2023;12(6):927.
Wang B, Han J, Elisseeff JH, Demaria M. The senescence-associated secretory phenotype and its physiological and pathological implications. Nat Rev Mol Cell Biol. 2024;25(12):958–78.
Zingoni A, Antonangeli F, Sozzani S, Santoni A, Cippitelli M, Soriani A. The senescence journey in cancer immunoediting. Mol Cancer. 2024;23(1):68.
Tao W, Yu Z, Han JJ. Single-cell senescence identification reveals senescence heterogeneity, trajectory, and modulators. Cell Metab. 2024;36(5):1126-43.e5.
Allis CD, Jenuwein T. The molecular hallmarks of epigenetic control. Nat Rev Genet. 2016;17(8):487–500.
Zhu X, Chen Z, Shen W, Huang G, Sedivy JM, Wang H, Ju Z. Inflammation, epigenetics, and metabolism converge to cell senescence and ageing: the regulation and intervention. Signal Transduct Target Ther. 2021;6(1):245.
Zhao D, Gao Y, Su Y, Zhou Y, Yang T, Li Y, Wang Y, Sun Y, Chen L, Zhang F, Zhang Z, Wang F, Shao J, Zheng S. Oroxylin A regulates cGAS DNA hypermethylation induced by methionine metabolism to promote HSC senescence. Pharmacol Res. 2023;187:106590.
Bonder MJ, Clark SJ, Krueger F, Luo S, Agostinho de Sousa J, Hashtroud AM, Stubbs TM, Stark AK, Rulands S, Stegle O, Reik W, von Meyenn F. scEpiAge: an age predictor highlighting single-cell ageing heterogeneity in mouse blood. Nat Commun. 2024;15(1):7567.
Feng Y, Wang X, Xu L, Pan H, Zhu S, Liang Q, Huang B, Lu J. The transcription factor ZBP-89 suppresses p16 expression through a histone modification mechanism to affect cell senescence. Febs j. 2009;276(15):4197–206.
Schleich K, Kase J, Dörr JR, Trescher S, Bhattacharya A, Yu Y, Wailes EM, Fan DNY, Lohneis P, Milanovic M, Lau A, Lenze D, Hummel M, Chapuy B, Leser U, Reimann M, Lee S, Schmitt CA. H3K9me3-mediated epigenetic regulation of senescence in mice predicts outcome of lymphoma patients. Nat Commun. 2020;11(1):3651.
Clapier CR, Cairns BR. The biology of chromatin remodeling complexes. Annu Rev Biochem. 2009;78:273–304.
Gu J, Chen J, Yin Q, Dong M, Zhang Y, Chen M, Chen X, Min J, He X, Tan Y, Zheng L, Jiang H, Wang B, Li X, Chen H. lncRNA JPX-Enriched Chromatin Microenvironment Mediates Vascular Smooth Muscle Cell Senescence and Promotes Atherosclerosis. Arterioscler Thromb Vasc Biol. 2024;44(1):156–76.x
Bartkova J, Rezaei N, Liontos M, Karakaidos P, Kletsas D, Issaeva N, Vassiliou LV, Kolettas E, Niforou K, Zoumpourlis VC, Takaoka M, Nakagawa H, Tort F, Fugger K, Johansson F, Sehested M, Andersen CL, Dyrskjot L, Ørntoft T, Lukas J, Kittas C, Helleday T, Halazonetis TD, Bartek J, Gorgoulis VG. Oncogene-induced senescence is part of the tumorigenesis barrier imposed by DNA damage checkpoints. Nature. 2006;444(7119):633–7.
Michaloglou C, Vredeveld LC, Soengas MS, Denoyelle C, Kuilman T, van der Horst CM, Majoor DM, Shay JW, Mooi WJ, Peeper DS. BRAFE600-associated senescence-like cell cycle arrest of human naevi. Nature. 2005;436(7051):720–4.
Collado M, Gil J, Efeyan A, Guerra C, Schuhmacher AJ, Barradas M, Benguría A, Zaballos A, Flores JM, Barbacid M, Beach D, Serrano M. Tumour biology: senescence in premalignant tumours. Nature. 2005;436(7051):642.
Brenner AJ, Stampfer MR, Aldaz CM. Increased p16 expression with first senescence arrest in human mammary epithelial cells and extended growth capacity with p16 inactivation. Oncogene. 1998;17(2):199–205.
Chen Y, Li Y, Peng Y, Zheng X, Fan S, Yi Y, Zeng P, Chen H, Kang H, Zhang Y, Xiao ZX, Li C. ΔNp63α down-regulates c-Myc modulator MM1 via E3 ligase HERC3 in the regulation of cell senescence. Cell Death Differ. 2018;25(12):2118–29.
Dimri GP, Martinez JL, Jacobs JJ, Keblusek P, Itahana K, Van Lohuizen M, Campisi J, Wazer DE, Band V. The Bmi-1 oncogene induces telomerase activity and immortalizes human mammary epithelial cells. Cancer Res. 2002;62(16):4736–45.
Elangovan S, Ramachandran S, Venkatesan N, Ananth S, Gnana-Prakasam JP, Martin PM, Browning DD, Schoenlein PV, Prasad PD, Ganapathy V, Thangaraju M. SIRT1 is essential for oncogenic signaling by estrogen/estrogen receptor α in breast cancer. Cancer Res. 2011;71(21):6654–64.
Yu W, Lin X, Leng S, Hou Y, Dang Z, Xue S, Li N, Zhang F. The PRC2 complex epigenetically silences GATA4 to suppress cellular senescence and promote the progression of breast cancer. Transl Oncol. 2024;46:102014.
Xing L, Tang X, Wu K, Huang X, Yi Y, Huan J. TRIM27 Functions as a Novel Oncogene in Non-Triple-Negative Breast Cancer by Blocking Cellular Senescence through p21 Ubiquitination. Mol Ther Nucleic Acids. 2020;22:910–23.
Sandhu C, Donovan J, Bhattacharya N, Stampfer M, Worland P, Slingerland J. Reduction of Cdc25A contributes to cyclin E1-Cdk2 inhibition at senescence in human mammary epithelial cells. Oncogene. 2000;19(47):5314–23.
Panneer Selvam S, De Palma RM, Oaks JJ, Oleinik N, Peterson YK, Stahelin RV, Skordalakes E, Ponnusamy S, Garrett-Mayer E, Smith CD, Ogretmen B. Binding of the sphingolipid S1P to hTERT stabilizes telomerase at the nuclear periphery by allosterically mimicking protein phosphorylation. Sci Signal. 2015;8(381):ra58.
Jee HJ, Kim AJ, Song N, Kim HJ, Kim M, Koh H, Yun J. Nek6 overexpression antagonizes p53-induced senescence in human cancer cells. Cell Cycle. 2010;9(23):4703–10.
Fei Z, Gao W, Xie R, Feng G, Chen X, Jiang Y. Synaptotagmin-7, a binding protein of P53, inhibits the senescence and promotes the tumorigenicity of lung cancer cells. Biosci Rep. 2019;39(2):BSR20181298.
Xu X, Xuan X, Zhang J, Xu H, Yang X, Zhang L, Zhao Y, Xu H, Li D. PSMD7 downregulation suppresses lung cancer progression by regulating the p53 pathway. J Cancer. 2021;12(16):4945–57.
Scaglioni PP, Yung TM, Choi S, Baldini C, Konstantinidou G, Pandolfi PP. CK2 mediates phosphorylation and ubiquitin-mediated degradation of the PML tumor suppressor. Mol Cell Biochem. 2008;316(1–2):149–54.
Ren XS, Yin MH, Zhang X, Wang Z, Feng SP, Wang GX, Luo YJ, Liang PZ, Yang XQ, He JX, Zhang BL. Tumor-suppressive microRNA-449a induces growth arrest and senescence by targeting E2F3 in human lung cancer cells. Cancer Lett. 2014;344(2):195–203.
Song NY, Zhu F, Wang Z, Willette-Brown J, Xi S, Sun Z, Su L, Wu X, Ma B, Nussinov R, Xia X, Schrump DS, Johnson PF, Karin M, Hu Y. IKKα inactivation promotes Kras-initiated lung adenocarcinoma development through disrupting major redox regulatory pathways. Proc Natl Acad Sci U S A. 2018;115(4):E812–21.
Gao L, Hu Y, Tian Y, Fan Z, Wang K, Li H, Zhou Q, Zeng G, Hu X, Yu L, Zhou S, Tong X, Huang H, Chen H, Liu Q, Liu W, Zhang G, Zeng M, Zhou G, He Q, Ji H, Chen L. Lung cancer deficient in the tumor suppressor GATA4 is sensitive to TGFBR1 inhibition. Nat Commun. 2019;10(1):1665.
Chen W, Chen Z, Zhang M, Tian Y, Liu L, Lan R, Zeng G, Fu X, Ru G, Liu W, Chen L, Fan Z. GATA6 Exerts Potent Lung Cancer Suppressive Function by Inducing Cell Senescence. Front Oncol. 2020;10:824.
Liang J, Lu F, Li B, Liu L, Zeng G, Zhou Q, Chen L. IRF8 induces senescence of lung cancer cells to exert its tumor suppressive function. Cell Cycle. 2019;18(23):3300–12.
de Barrios O, Győrffy B, Fernández-Aceñero MJ, Sánchez-Tilló E, Sánchez-Moral L, Siles L, Esteve-Arenys A, Roué G, Casal JI, Darling DS, Castells A, Postigo A. ZEB1-induced tumourigenesis requires senescence inhibition via activation of DKK1/mutant p53/Mdm2/CtBP and repression of macroH2A1. Gut. 2017;66(4):666–82.
Sharma BK, Mureb D, Murab S, Rosenfeldt L, Francisco B, Cantrell R, Karns R, Romick-Rosendale L, Watanabe-Chailland M, Mast J, Flick MJ, Whitlock PW, Palumbo JS. Fibrinogen activates focal adhesion kinase (FAK) promoting colorectal adenocarcinoma growth. J Thromb Haemost. 2021;19(10):2480–94.
Qin S, Liu Y, Zhang X, Huang P, Xia L, Leng W, Li D. lncRNA FGD5-AS1 is required for gastric cancer proliferation by inhibiting cell senescence and ROS production via stabilizing YBX1. J Exp Clin Cancer Res. 2024;43(1):188.
Cui Y, Qin L, Wu J, Qu X, Hou C, Sun W, Li S, Vaughan AT, Li JJ, Liu J. SIRT3 Enhances Glycolysis and Proliferation in SIRT3-Expressing Gastric Cancer Cells. PLoS ONE. 2015;10(6):e0129834.
Xu MD, Dong L, Qi P, Weng WW, Shen XH, Ni SJ, Huang D, Tan C, Sheng WQ, Zhou XY, Du X. Pituitary tumor-transforming gene-1 serves as an independent prognostic biomarker for gastric cancer. Gastric Cancer. 2016;19(1):107–15.
Li Y, Li D, Zhao M, Huang S, Zhang Q, Lin H, Wang W, Li K, Li Z, Huang W, Che Y, Huang C. Long noncoding RNA SNHG6 regulates p21 expression via activation of the JNK pathway and regulation of EZH2 in gastric cancer cells. Life Sci. 2018;208:295–304.
Ma X, Wang N, Chen K, Zhang C. Oncosuppressive role of MicroRNA-205-3p in gastric cancer through inhibition of proliferation and induction of senescence: Oncosuppressive role of MicroRNA-205 in gastric cancer. Transl Oncol. 2021;14(11):101199.
Wang K, Jiang X, Jiang Y, Liu J, Du Y, Zhang Z, Li Y, Zhao X, Li J, Zhang R. EZH2-H3K27me3-mediated silencing of mir-139-5p inhibits cellular senescence in hepatocellular carcinoma by activating TOP2A. J Exp Clin Cancer Res. 2023;42(1):320.
Wang H, Cui Y, Gong H, Xu J, Huang S, Tang A. Suppression of AGTR1 Induces Cellular Senescence in Hepatocellular Carcinoma Through Inactivating ERK Signaling. Front Bioeng Biotechnol. 2022;10:929979.
Wang Y, Cheng C, Lu Y, Lian Z, Liu Q, Xu Y, Li Y, Li H, Zhang L, Jiang X, Li B, Yu D. β-Catenin Activation Reprograms Ammonia Metabolism to Promote Senescence Resistance in Hepatocellular Carcinoma. Cancer Res. 2024;84(10):1643–58.
Deng Q, Li KY, Chen H, Dai JH, Zhai YY, Wang Q, Li N, Wang YP, Han ZG. RNA interference against cancer/testis genes identifies dual specificity phosphatase 21 as a potential therapeutic target in human hepatocellular carcinoma. Hepatology. 2014;59(2):518–30.
Yang D, Li L, Liu H, Wu L, Luo Z, Li H, Zheng S, Gao H, Chu Y, Sun Y, Liu J, Jia L. Induction of autophagy and senescence by knockdown of ROC1 E3 ubiquitin ligase to suppress the growth of liver cancer cells. Cell Death Differ. 2013;20(2):235–47.
Zhao B, Lv X, Zhao X, Maimaitiaili S, Zhang Y, Su K, Yu H, Liu C, Qiao T. Tumor-promoting actions of HNRNP A1 in HCC are associated with cell cycle, mitochondrial dynamics, and necroptosis. Int J Mol Sci. 2022;23(18):10209.
Iwagami Y, Huang CK, Olsen MJ, Thomas JM, Jang G, Kim M, Lin Q, Carlson RI, Wagner CE, Dong X, Wands JR. Aspartate β-hydroxylase modulates cellular senescence through glycogen synthase kinase 3β in hepatocellular carcinoma. Hepatology. 2016;63(4):1213–26.
Zeng W, Liu F, Liu Y, Zhang Z, Hu H, Ning S, Zhang H, Chen X, Liao Z, Zhang Z. Targeting TM4SF1 promotes tumor senescence enhancing CD8+ T cell cytotoxic function in hepatocellular carcinoma. Clin Mol Hepatol. 2024.
Lao Y, Cui X, Xu Z, Yan H, Zhang Z, Zhang Z, Geng L, Li B, Lu Y, Guan Q, Pu X, Zhao S, Zhu J, Qin X, Sun B. Glutaryl-CoA dehydrogenase suppresses tumor progression and shapes an anti-tumor microenvironment in hepatocellular carcinoma. J Hepatol. 2024;81(5):847–61.
Yuan F, Zhang Y, Ma L, Cheng Q, Li G, Tong T. Enhanced NOLC1 promotes cell senescence and represses hepatocellular carcinoma cell proliferation by disturbing the organization of nucleolus. Aging Cell. 2017;16(4):726–37.
Hu B, Ding GY, Fu PY, Zhu XD, Ji Y, Shi GM, Shen YH, Cai JB, Yang Z, Zhou J, Fan J, Sun HC, Kuang M, Huang C. NOD-like receptor X1 functions as a tumor suppressor by inhibiting epithelial-mesenchymal transition and inducing aging in hepatocellular carcinoma cells. J Hematol Oncol. 2018;11(1):28.
Indersie E, Lesjean S, Hooks KB, Sagliocco F, Ernault T, Cairo S, Merched-Sauvage M, Rullier A, Le Bail B, Taque S, Grotzer M, Branchereau S, Guettier C, Fabre M, Brugières L, Hagedorn M, Buendia MA, Grosset CF. MicroRNA therapy inhibits hepatoblastoma growth in vivo by targeting β-catenin and Wnt signaling. Hepatol Commun. 2017;1(2):168–83.
Shiu TY, Shih YL, Feng AC, Lin HH, Huang SM, Huang TY, Hsieh CB, Chang WK, Hsieh TY. HCV core inhibits hepatocellular carcinoma cell replicative senescence through downregulating microRNA-138 expression. J Mol Med (Berl). 2017;95(6):629–39.
Fan DN, Tsang FH, Tam AH, Au SL, Wong CC, Wei L, Lee JM, He X, Ng IO, Wong CM. Histone lysine methyltransferase, suppressor of variegation 3–9 homolog 1, promotes hepatocellular carcinoma progression and is negatively regulated by microRNA-125b. Hepatology. 2013;57(2):637–47.
Mahajan AS, Arikatla VS, Thyagarajan A, Zhelay T, Sahu RP, Kemp MG, Spandau DF, Travers JB. Creatine and nicotinamide prevent oxidant-induced senescence in human fibroblasts. Nutrients. 2021;13(11):4102.
Ke H, Harris R, Coloff JL, Jin JY, Leshin B, Miliani de Marval P, Tao S, Rathmell JC, Hall RP, Zhang JY. The c-Jun NH2-terminal kinase 2 plays a dominant role in human epidermal neoplasia. Cancer Res. 2010;70(8):3080–8.
Sang B, Zhang YY, Guo ST, Kong LF, Cheng Q, Liu GZ, Thorne RF, Zhang XD, Jin L, Wu M. Dual functions for OVAAL in initiation of RAF/MEK/ERK prosurvival signals and evasion of p27-mediated cellular senescence. Proc Natl Acad Sci U S A. 2018;115(50):E11661–70.
Schultz J, Ibrahim SM, Vera J, Kunz M. 14-3-3sigma gene silencing during melanoma progression and its role in cell cycle control and cellular senescence. Mol Cancer. 2009;8:53.
Krones-Herzig A, Mittal S, Yule K, Liang H, English C, Urcis R, Soni T, Adamson ED, Mercola D. Early growth response 1 acts as a tumor suppressor in vivo and in vitro via regulation of p53. Cancer Res. 2005;65(12):5133–43.
Gessner A, Thomas M, Castro PG, Büchler L, Scholz A, Brümmendorf TH, Soria NM, Vormoor J, Greil J, Heidenreich O. Leukemic fusion genes MLL/AF4 and AML1/MTG8 support leukemic self-renewal by controlling expression of the telomerase subunit TERT. Leukemia. 2010;24(10):1751–9.
Liu L, Xu F, Chang CK, He Q, Wu LY, Zhang Z, Li X. MYCN contributes to the malignant characteristics of erythroleukemia through EZH2-mediated epigenetic repression of p21. Cell Death Dis. 2017;8(10):e3126.
Peng D, Wang H, Li L, Ma X, Chen Y, Zhou H, Luo Y, Xiao Y, Liu L. miR-34c-5p promotes eradication of acute myeloid leukemia stem cells by inducing senescence through selective RAB27B targeting to inhibit exosome shedding. Leukemia. 2018;32(5):1180–8.
Guo M, Zhang J, Han J, Hu Y, Ni H, Yuan J, Sun Y, Liu M, Gao L, Liao W, Ma C, Liu Y, Li S, Li N. VEGFR2 blockade inhibits glioblastoma cell proliferation by enhancing mitochondrial biogenesis. J Transl Med. 2024;22(1):419.
Bernhart E, Damm S, Heffeter P, Wintersperger A, Asslaber M, Frank S, Hammer A, Strohmaier H, DeVaney T, Mrfka M, Eder H, Windpassinger C, Ireson CR, Mischel PS, Berger W, Sattler W. Silencing of protein kinase D2 induces glioma cell senescence via p53-dependent and -independent pathways. Neuro Oncol. 2014;16(7):933–45.
Xiaoping L, Zhibin Y, Wenjuan L, Zeyou W, Gang X, Zhaohui L, Ying Z, Minghua W, Guiyuan L. CPEB1, a histone-modified hypomethylated gene, is regulated by miR-101 and involved in cell senescence in glioma. Cell Death Dis. 2013;4(6):e675.
Stegh AH, Brennan C, Mahoney JA, Forloney KL, Jenq HT, Luciano JP, Protopopov A, Chin L, Depinho RA. Glioma oncoprotein Bcl2L12 inhibits the p53 tumor suppressor. Genes Dev. 2010;24(19):2194–204.
Verginelli F, Perin A, Dali R, Fung KH, Lo R, Longatti P, Guiot MC, Del Maestro RF, Rossi S, di Porzio U, Stechishin O, Weiss S, Stifani S. Transcription factors FOXG1 and Groucho/TLE promote glioblastoma growth. Nat Commun. 2013;4:2956.
Yamashita D, Kondo T, Ohue S, Takahashi H, Ishikawa M, Matoba R, Suehiro S, Kohno S, Harada H, Tanaka J, Ohnishi T. miR340 suppresses the stem-like cell function of glioma-initiating cells by targeting tissue plasminogen activator. Cancer Res. 2015;75(6):1123–33.
Tao YP, Zhu HY, Shi QY, Wang CX, Hua YX, Hu HY, Zhou QY, Zhou ZL, Sun Y, Wang XM, Wang Y, Zhang YL, Guo YJ, Wang ZY, Che X, Xu CW, Zhang XC, Heger M, Tao SP, Zheng X, Xu Y, Ao L, Liu AJ, Liu SB, Cheng SQ, Pan WW. S1PR1 regulates ovarian cancer cell senescence through the PDK1-LATS1/2-YAP pathway. Oncogene. 2023;42(47):3491–502.
Li G, Xu W, Li X, Chen M, Shi Y, Wei M, Peng D. Oncogenic SIRT7 inhibits GATA4 transcriptional activity and activates the Wnt signaling pathway in ovarian cancer. Gynecol Oncol. 2023;171:39–48.
Prathapam T, Aleshin A, Guan Y, Gray JW, Martin GS. p27Kip1 mediates addiction of ovarian cancer cells to MYCC (c-MYC) and their dependence on MYC paralogs. J Biol Chem. 2010;285(42):32529–38.
Shaosheng W, Shaochuang W, Lichun F, Na X, Xiaohong Z. ITPKA induces cell senescence, inhibits ovarian cancer tumorigenesis and can be downregulated by miR-203. Aging (Albany NY). 2021;13(8):11822–32.
Onyiba CI, Scarlett CJ, Weidenhofer J. The mechanistic roles of sirtuins in breast and prostate cancer. Cancers (Basel). 2022;14(20):5118.
Liu R, Fan M, Candas D, Qin L, Zhang X, Eldridge A, Zou JX, Zhang T, Juma S, Jin C, Li RF, Perks J, Sun LQ, Vaughan AT, Hai CX, Gius DR, Li JJ. CDK1-Mediated SIRT3 Activation Enhances Mitochondrial Function and Tumor Radioresistance. Mol Cancer Ther. 2015;14(9):2090–102.
Boulanger CA, Smith GH. Reducing mammary cancer risk through premature stem cell senescence. Oncogene. 2001;20(18):2264–72.
Kruk PA, Godwin AK, Hamilton TC, Auersperg N. Telomeric instability and reduced proliferative potential in ovarian surface epithelial cells from women with a family history of ovarian cancer. Gynecol Oncol. 1999;73(2):229–36.
Mikheev AM, Ramakrishna R, Stoll EA, Mikheeva SA, Beyer RP, Plotnik DA, Schwartz JL, Rockhill JK, Silber JR, Born DE, Kosai Y, Horner PJ, Rostomily RC. Increased age of transformed mouse neural progenitor/stem cells recapitulates age-dependent clinical features of human glioma malignancy. Aging Cell. 2012;11(6):1027–35.
Bai H, Liu X, Lin M, Meng Y, Tang R, Guo Y, Li N, Clarke MF, Cai S. Progressive senescence programs induce intrinsic vulnerability to aging-related female breast cancer. Nat Commun. 2024;15(1):5154.
Toutfaire M, Dumortier E, Fattaccioli A, Van Steenbrugge M, Proby CM, Debacq-Chainiaux F. Unraveling the interplay between senescent dermal fibroblasts and cutaneous squamous cell carcinoma cell lines at different stages of tumorigenesis. Int J Biochem Cell Biol. 2018;98:113–26.
Golomb L, Sagiv A, Pateras IS, Maly A, Krizhanovsky V, Gorgoulis VG, Oren M, Ben-Yehuda A. Age-associated inflammation connects RAS-induced senescence to stem cell dysfunction and epidermal malignancy. Cell Death Differ. 2015;22(11):1764–74.
Takeshima H, Ushijima T. Accumulation of genetic and epigenetic alterations in normal cells and cancer risk. NPJ Precision Oncol. 2019;3(1):7.
Yoshimoto S, Loo TM, Atarashi K, Kanda H, Sato S, Oyadomari S, Iwakura Y, Oshima K, Morita H, Hattori M, Honda K, Ishikawa Y, Hara E, Ohtani N. Obesity-induced gut microbial metabolite promotes liver cancer through senescence secretome. Nature. 2013;499(7456):97–101.
Yamagishi R, Kamachi F, Nakamura M, Yamazaki S, Kamiya T, Takasugi M, Cheng Y, Nonaka Y, Yukawa-Muto Y, Thuy LTT, Harada Y, Arai T, Loo TM, Yoshimoto S, Ando T, Nakajima M, Taguchi H, Ishikawa T, Akiba H, Miyake S, Kubo M, Iwakura Y, Fukuda S, Chen WY, Kawada N, Rudensky A, Nakae S, Hara E, Ohtani N. Gasdermin D-mediated release of IL-33 from senescent hepatic stellate cells promotes obesity-associated hepatocellular carcinoma. Sci Immunol. 2022;7(72):eabl7209.
Wang Y, Zhang H, Wang M, He J, Guo H, Li L, Wang J. CCNB2/SASP/Cathepsin B & PGE2 Axis Induce Cell Senescence Mediated Malignant Transformation. Int J Biol Sci. 2021;17(13):3538–53.
Dalmasso G, Cougnoux A, Faïs T, Bonnin V, Mottet-Auselo B, Nguyen HT, Sauvanet P, Barnich N, Jary M, Pezet D, Delmas J, Bonnet R. Colibactin-producing Escherichia coli enhance resistance to chemotherapeutic drugs by promoting epithelial to mesenchymal transition and cancer stem cell emergence. Gut Microbes. 2024;16(1):2310215.
Kang TW, Yevsa T, Woller N, Hoenicke L, Wuestefeld T, Dauch D, Hohmeyer A, Gereke M, Rudalska R, Potapova A, Iken M, Vucur M, Weiss S, Heikenwalder M, Khan S, Gil J, Bruder D, Manns M, Schirmacher P, Tacke F, Ott M, Luedde T, Longerich T, Kubicka S, Zender L. Senescence surveillance of pre-malignant hepatocytes limits liver cancer development. Nature. 2011;479(7374):547–51.
Dong Z, Luo Y, Yuan Z, Tian Y, Jin T, Xu F. Cellular senescence and SASP in tumor progression and therapeutic opportunities. Mol Cancer. 2024;23(1):181.
Marin I, Boix O, Garcia-Garijo A, Sirois I, Caballe A, Zarzuela E, Ruano I, Attolini CS, Prats N, López-Domínguez JA, Kovatcheva M, Garralda E, Muñoz J, Caron E, Abad M, Gros A, Pietrocola F, Serrano M. Cellular Senescence Is Immunogenic and Promotes Antitumor Immunity. Cancer Discov. 2023;13(2):410–31.
Wang ST, Huang SW, Liu KT, Lee TY, Shieh JJ, Wu CY. Atorvastatin-induced senescence of hepatocellular carcinoma is mediated by downregulation of hTERT through the suppression of the IL-6/STAT3 pathway. Cell Death Discov. 2020;6:17.
Qiu YD, Yan Q, Wang Y, Ye YF, Wang Y, Wang MY, Wang PP, Zhang SY, Wang DL, Yan H, Ruan J, Zhao YJ, Huang LH, Cho N, Wang K, Zheng XH, Liu ZG. Discovery of a selective TRF2 inhibitor FKB04 induced telomere shortening and senescence in liver cancer cells. Acta Pharmacol Sin. 2024;45(6):1276–86.
Kaewtunjai N, Wongpoomchai R, Imsumran A, Pompimon W, Athipornchai A, Suksamrarn A, Lee TR, Tuntiwechapikul W. Ginger Extract Promotes Telomere Shortening and Cellular Senescence in A549 Lung Cancer Cells. ACS Omega. 2018;3(12):18572–81.
Ji HJ, Park KH, Kim TS, Rha SY, Yoo NC, Kim JM, Kim JS, Roh JK, Jang WI, Chung HC. Changes of Telomerase Activity and Proliferation by Inhibition of Reverse Transcriptase Activity in Human Cancer Cell. Cancer Res Treat. 2002;34(3):223–33.
Taka T, Huang L, Wongnoppavich A, Tam-Chang SW, Lee TR, Tuntiwechapikul W. Telomere shortening and cell senescence induced by perylene derivatives in A549 human lung cancer cells. Bioorg Med Chem. 2013;21(4):883–90.
Mancini J, Rousseau P, Castor KJ, Sleiman HF, Autexier C. Platinum(II) phenanthroimidazole G-quadruplex ligand induces selective telomere shortening in A549 cancer cells. Biochimie. 2016;121:287–97.
Cassar L, Nicholls C, Pinto AR, Chen R, Wang L, Li H, Liu JP. TGF-beta receptor mediated telomerase inhibition, telomere shortening and breast cancer cell senescence. Protein Cell. 2017;8(1):39–54.
Ma F, Ma Y, Liu K, Gao J, Li S, Sun X, Li G. Resveratrol induces DNA damage-mediated cancer cell senescence through the DLC1-DYRK1A-EGFR axis. Food Funct. 2023;14(3):1484–97.
Bian Y, Wang X, Zheng Z, Ren G, Zhu H, Qiao M, Li G. Resveratrol drives cancer cell senescence via enhancing p38MAPK and DLC1 expressions. Food Funct. 2022;13(6):3283–93.
Liang C, Yi K, Zhou X, Li X, Zhong C, Cao H, Xie C, Zhu J. Destruction of the cellular antioxidant pool contributes to resveratrol-induced senescence and apoptosis in lung cancer. Phytother Res. 2023;37(7):2995–3008.
Lu X, Yuan F, Qiao L, Liu Y, Gu Q, Qi X, Li J, Li D, Liu M. AS1041, a novel derivative of marine natural compound Aspergiolide A, induces senescence of leukemia cells via oxidative stress-induced DNA damage and BCR-ABL degradation. Biomed Pharmacother. 2024;171:116099.
Rahmani F, Hashemzehi M, Avan A, Barneh F, Asgharzadeh F, Moradi Marjaneh R, Soleimani A, Parizadeh M, Ferns GA, Ghayour Mobarhan M, Ryzhikov M, Afshari AR, Ahmadian MR, Giovannetti E, Jafari M, Khazaei M, Hassanian SM. Rigosertib elicits potent anti-tumor responses in colorectal cancer by inhibiting Ras signaling pathway. Cell Signal. 2021;85:110069.
Burkner GT, Dias DA, Souza KFS, Araújo AJP, Basilio D, Jacobsen FT, Moraes ACR, Silva-Filho SE, Cavalcante MFO, Moraes CAO, Saba S, Macedo MLR, Paredes-Gamero EJ, Rafique J, Parisotto EB. Selenylated Imidazo[1,2-a]pyridine induces cell senescence and oxidative stress in chronic myeloid leukemia cells. Molecules. 2023;28(2):893.
Lestari B, Nakamae I, Yoneda-Kato N, Morimoto T, Kanaya S, Yokoyama T, Shionyu M, Shirai T, Meiyanto E, Kato JY. Pentagamavunon-1 (PGV-1) inhibits ROS metabolic enzymes and suppresses tumor cell growth by inducing M phase (prometaphase) arrest and cell senescence. Sci Rep. 2019;9(1):14867.
Wang LX, Wang JD, Chen JJ, Long B, Liu LL, Tu XX, Luo Y, Hu Y, Lin DJ, Lu G, Long ZJ, Liu Q. Aurora A Kinase Inhibitor AKI603 Induces Cellular Senescence in Chronic Myeloid Leukemia Cells Harboring T315I Mutation. Sci Rep. 2016;6:35533.
Venturelli S, Berger A, Weiland T, Essmann F, Waibel M, Nuebling T, Häcker S, Schenk M, Schulze-Osthoff K, Salih HR, Fulda S, Sipos B, Johnstone RW, Lauer UM, Bitzer M. Differential induction of apoptosis and senescence by the DNA methyltransferase inhibitors 5-azacytidine and 5-aza-2’-deoxycytidine in solid tumor cells. Mol Cancer Ther. 2013;12(10):2226–36.
Yao GD, Yang J, Li Q, Zhang Y, Qi M, Fan SM, Hayashi T, Tashiro S, Onodera S, Ikejima T. Activation of p53 contributes to pseudolaric acid B-induced senescence in human lung cancer cells in vitro. Acta Pharmacol Sin. 2016;37(7):919–29.
Liu L, Wang J, Shi L, Zhang W, Du X, Wang Z, Zhang Y. β-Asarone induces senescence in colorectal cancer cells by inducing lamin B1 expression. Phytomedicine. 2013;20(6):512–20.
Yang H, Qiu L, Zhang L, Lv G, Li K, Yu H, Xie M, Lin J. Platinum-zoledronate complex blocks gastric cancer cell proliferation by inducing cell cycle arrest and apoptosis. Tumour Biol. 2016;37(8):10981–92.
Garg S, Huifu H, Kumari A, Sundar D, Kaul SC, Wadhwa R. Induction of Senescence in Cancer Cells by a Novel Combination of Cucurbitacin B and Withanone: Molecular Mechanism and Therapeutic Potential. J Gerontol A Biol Sci Med Sci. 2020;75(6):1031–41.
Vilgelm AE, Pawlikowski JS, Liu Y, Hawkins OE, Davis TA, Smith J, Weller KP, Horton LW, McClain CM, Ayers GD, Turner DC, Essaka DC, Stewart CF, Sosman JA, Kelley MC, Ecsedy JA, Johnston JN, Richmond A. Mdm2 and aurora kinase a inhibitors synergize to block melanoma growth by driving apoptosis and immune clearance of tumor cells. Cancer Res. 2015;75(1):181–93.
Hu H, Wang Q, Yu D, Tao X, Guo M, Tian S, Zhang Q, Xu M, Geng X, Zhang H, Xu H, Li L, Xie S, Chen K, Zhu W, Li XW, Xu H, Li B, Zhang W, Liu S. Berberine derivative B68 promotes tumor immune clearance by dual-targeting BMI1 for senescence induction and CSN5 for PD-L1 degradation. Adv Sci (Weinh). 2024;12(7):e2413122.
Lodygin D, Menssen A, Hermeking H. Induction of the Cdk inhibitor p21 by LY83583 inhibits tumor cell proliferation in a p53-independent manner. J Clin Invest. 2002;110(11):1717–27.
An HM, Xue YF, Shen YL, Du Q, Hu B. Sodium valproate induces cell senescence in human hepatocarcinoma cells. Molecules. 2013;18(12):14935–47.
Alcántara-Flores E, Brechú-Franco AE, García-López P, Rocha-Zavaleta L, López-Marure R, Martínez-Vázquez M. Argentatin B Inhibits Proliferation of Prostate and Colon Cancer Cells by Inducing Cell Senescence. Molecules. 2015;20(12):21125–37.
Huang J, Yang G, Huang Y, Kong W, Zhang S. 1,25(OH)2D3 inhibits the progression of hepatocellular carcinoma via downregulating HDAC2 and upregulating P21(WAFI/CIP1). Mol Med Rep. 2016;13(2):1373–80.
Colucci M, Zumerle S, Bressan S, Gianfanti F, Troiani M, Valdata A, D’Ambrosio M, Pasquini E, Varesi A, Cogo F, Mosole S, Dongilli C, Desbats MA, Contu L, Revankdar A, Chen J, Kalathur M, Perciato ML, Basilotta R, Endre L, Schauer S, Othman A, Guccini I, Saponaro M, Maraccani L, Bancaro N, Lai P, Liu L, Pernigoni N, Mele F, Merler S, Trotman LC, Guarda G, Calì B, Montopoli M, Alimonti A. Retinoic acid receptor activation reprograms senescence response and enhances anti-tumor activity of natural killer cells. Cancer Cell. 2024;42(4):646-61.e9.
Khan I, Bahuguna A, Bhardwaj M, Pal Khaket T, Kang SC. Carvacrol nanoemulsion evokes cell cycle arrest, apoptosis induction and autophagy inhibition in doxorubicin resistant-A549 cell line. Artif Cells Nanomed Biotechnol. 2018;46(sup1):664–75.
Drullion C, Trégoat C, Lagarde V, Tan S, Gioia R, Priault M, Djavaheri-Mergny M, Brisson A, Auberger P, Mahon FX, Pasquet JM. Apoptosis and autophagy have opposite roles on imatinib-induced K562 leukemia cell senescence. Cell Death Dis. 2012;3(8):e373.
Gleason CE, Dickson MA, Klein Dooley ME, Antonescu CR, Gularte-Mérida R, Benitez M, Delgado JI, Kataru RP, Tan MWY, Bradic M, Adamson TE, Seier K, Richards AL, Palafox M, Chan E, D’Angelo SP, Gounder MM, Keohan ML, Kelly CM, Chi P, Movva S, Landa J, Crago AM, Donoghue MTA, Qin LX, Serra V, Turkekul M, Barlas A, Firester DM, Manova-Todorova K, Mehrara BJ, Kovatcheva M, Tan NS, Singer S, Tap WD, Koff A. Therapy-Induced Senescence Contributes to the Efficacy of Abemaciclib in Patients with Dedifferentiated Liposarcoma. Clin Cancer Res. 2024;30(4):703–18.
Bi H, Shang J, Zou X, Xu J, Han Y. Palbociclib induces cell senescence and apoptosis of gastric cancer cells by inhibiting the Notch pathway. Oncol Lett. 2021;22(2):603.
Fu R, Yang P, Sajid A, Li Z. Avenanthramide A Induces Cellular Senescence via miR-129-3p/Pirh2/p53 Signaling Pathway To Suppress Colon Cancer Growth. J Agric Food Chem. 2019;67(17):4808–16.
Wang H, Yuan S, Zheng Q, Zhang S, Zhang Q, Ji S, Wang W, Cao Y, Guo Y, Yang X, Geng H, Yang F, Xi S, Jin G, Zhang J, Gao Q, Bernards R, Qin W, Wang C. Dual Inhibition of CDK4/6 and XPO1 Induces Senescence With Acquired Vulnerability to CRBN-Based PROTAC Drugs. Gastroenterology. 2024;166(6):1130-44.e8.
Drugs Halt Replication Later than Thought. Cancer Discov. 2023;13(9):1955.
Wagner V, Kern F, Hahn O, Schaum N, Ludwig N, Fehlmann T, Engel A, Henn D, Rishik S, Isakova A, Tan M, Sit R, Neff N, Hart M, Meese E, Quake S, Wyss-Coray T, Keller A. Characterizing expression changes in noncoding RNAs during aging and heterochronic parabiosis across mouse tissues. Nat Biotechnol. 2024;42(1):109–18.
Bai XY, Che FX, Chen XM, Meng LY, Zhou Y. p16INK4a expression mediated by recombinant adenovirus can induce senescence of A549 cells. Zhonghua Shi Yan He Lin Chuang Bing Du Xue Za Zhi. 2004;18(1):54–8.
Li X, Qian X, Wang B, Xia Y, Zheng Y, Du L, Xu D, Xing D, DePinho RA, Lu Z. Programmable base editing of mutated TERT promoter inhibits brain tumour growth. Nat Cell Biol. 2020;22(3):282–8.
Menegotto PR, da Costa Lopez PL, Souza BK, de Farias CB, Filippi-Chiela EC, Vieira IA, Schwartsmann G, Lenz G, Roesler R. Gastrin-Releasing Peptide Receptor Knockdown Induces Senescence in Glioblastoma Cells. Mol Neurobiol. 2017;54(2):888–94.
Christov KT, Shilkaitis AL, Kim ES, Steele VE, Lubet RA. Chemopreventive agents induce a senescence-like phenotype in rat mammary tumours. Eur J Cancer. 2003;39(2):230–9.
Su D, Zhu S, Han X, Feng Y, Huang H, Ren G, Pan L, Zhang Y, Lu J, Huang B. BMP4-Smad signaling pathway mediates adriamycin-induced premature senescence in lung cancer cells. J Biol Chem. 2009;284(18):12153–64.
Zhao Y, Liu X, Si F, Huang L, Gao A, Lin W, Hoft DF, Shao Q, Peng G. Citrate Promotes Excessive Lipid Biosynthesis and Senescence in Tumor Cells for Tumor Therapy. Adv Sci (Weinh). 2022;9(1):e2101553.
He Q, Xue S, Tan Y, Zhang L, Shao Q, Xing L, Li Y, Xiang T, Luo X, Ren G. Dual inhibition of Akt and ERK signaling induces cell senescence in triple-negative breast cancer. Cancer Lett. 2019;448:94–104.
Wang M, Chen S, Wei Y, Wei X. DNA-PK inhibition by M3814 enhances chemosensitivity in non-small cell lung cancer. Acta Pharm Sin B. 2021;11(12):3935–49.
Zhang Z, Chen L, Yang Q, Tang X, Li J, Zhang G, Wang Y, Huang H. INHBA regulates Hippo signaling to confer 5-FU chemoresistance mediated by cellular senescence in colon cancer cells. Int J Biochem Cell Biol. 2024;171:106570.
Sun X, Shi B, Zheng H, Min L, Yang J, Li X, Liao X, Huang W, Zhang M, Xu S, Zhu Z, Cui H, Liu X. Senescence-associated secretory factors induced by cisplatin in melanoma cells promote non-senescent melanoma cell growth through activation of the ERK1/2-RSK1 pathway. Cell Death Dis. 2018;9(3):260.
Dai D, Pei Y, Zhu B, Wang D, Pei S, Huang H, Zhu Q, Deng X, Ye J, Xu J, Chen X, Huang M, Xiao Y. Chemoradiotherapy-induced ACKR2(+) tumor cells drive CD8(+) T cell senescence and cervical cancer recurrence. Cell Rep Med. 2024;5(5):101550.
Kang D, Hwang HJ, Baek Y, Sung JY, Kim K, Park HJ, Ko YG, Kim YN, Lee JS. TRIM22 induces cellular senescence by targeting PHLPP2 in hepatocellular carcinoma. Cell Death Dis. 2024;15(1):26.
Efimova EV, Mauceri HJ, Golden DW, Labay E, Bindokas VP, Darga TE, Chakraborty C, Barreto-Andrade JC, Crawley C, Sutton HG, Kron SJ, Weichselbaum RR. Poly(ADP-ribose) polymerase inhibitor induces accelerated senescence in irradiated breast cancer cells and tumors. Cancer Res. 2010;70(15):6277–82.
Choi HS, Kim JH, Jang SJ, Yun JW, Kang KM, Jeong H, Ha IB, Jeong BK. Synergistic Tumoricidal Effects of Alpha-Lipoic Acid and Radiotherapy on Human Breast Cancer Cells via HMGB1. Cancer Res Treat. 2021;53(3):685–94.
Ding X, Cheng J, Pang Q, Wei X, Zhang X, Wang P, Yuan Z, Qian D. BIBR1532, a Selective Telomerase Inhibitor, Enhances Radiosensitivity of Non-Small Cell Lung Cancer Through Increasing Telomere Dysfunction and ATM/CHK1 Inhibition. Int J Radiat Oncol Biol Phys. 2019;105(4):861–74.
Zou Y, Wang S, Zhang H, Gu Y, Chen H, Huang Z, Yang F, Li W, Chen C, Men L, Tian Q, Xie T. The triangular relationship between traditional Chinese medicines, intestinal flora, and colorectal cancer. Med Res Rev. 2024;44(2):539–67.
Zhang J, Li C, Zhang L, Heng Y, Wang S, Pan Y, Cai L, Zhang Y, Xu T, Chen X, Hoffman RM, Jia L. Andrographolide, a diterpene lactone from the Traditional Chinese Medicine Andrographis paniculate, induces senescence in human lung adenocarcinoma via p53/p21 and Skp2/p27. Phytomedicine. 2022;98:153933.
Wei S, Liu L, Chen Z, Yin W, Liu Y, Ouyang Q, Zeng F, Nie Y, Chen T. Artesunate inhibits the mevalonate pathway and promotes glioma cell senescence. J Cell Mol Med. 2020;24(1):276–84.
Wang Y, Lv H, Dai C, Wang X, Yin Y, Chen Z. Oridonin Dose-Dependently Modulates the Cell Senescence and Apoptosis of Gastric Cancer Cells. Evid Based Complement Alternat Med. 2021;2021:5023536.
Gao FH, Hu XH, Li W, Liu H, Zhang YJ, Guo ZY, Xu MH, Wang ST, Jiang B, Liu F, Zhao YZ, Fang Y, Chen FY, Wu YL. Oridonin induces apoptosis and senescence in colorectal cancer cells by increasing histone hyperacetylation and regulation of p16, p21, p27 and c-myc. BMC Cancer. 2010;10:610.
Mosieniak G, Adamowicz M, Alster O, Jaskowiak H, Szczepankiewicz AA, Wilczynski GM, Ciechomska IA, Sikora E. Curcumin induces permanent growth arrest of human colon cancer cells: link between senescence and autophagy. Mech Ageing Dev. 2012;133(6):444–55.
Bao X, Liu Y, Huang J, Yin S, Sheng H, Han X, Chen Q, Wang T, Chen S, Qiu Y, Zhang C, Yu H. Stachydrine hydrochloride inhibits hepatocellular carcinoma progression via LIF/AMPK axis. Phytomedicine. 2022;100:154066.
Hu B, Du Q, Deng S, An HM, Pan CF, Shen KP, Xu L, Wei MM, Wang SS. Ligustrum lucidum Ait. fruit extract induces apoptosis and cell senescence in human hepatocellular carcinoma cells through upregulation of p21. Oncol Rep. 2014;32(3):1037–42.
Wulandari F, Ikawati M, Widyarini S, Kirihata M, Novitasari D, Kato JY, Meiyanto E. Tumour-suppressive effects of curcumin analogs CCA-1.1 and Pentagamavunone-1 in colon cancer: In vivo and in vitro studies. J Adv Pharm Technol Res. 2023;14(4):317–24.
Sun L, Li Y, Zhao R, Fan Q, Liu F, Zhu Y, Han J, Liu Y, Jin N, Li X, Li Y. Platycodin D2 enhances P21/CyclinA2-mediated senescence of HCC cells by regulating NIX-induced mitophagy. Cancer Cell Int. 2024;24(1):79.
Zia A, Farkhondeh T, Pourbagher-Shahri AM, Samarghandian S. The role of curcumin in aging and senescence: Molecular mechanisms. Biomed Pharmacother. 2021;134:111119.
An Y, Zhu J, Wang X, Sun X, Luo C, Zhang Y, Ye Y, Li X, Abulizi A, Huang Z, Zhang H, Yang B, Xie Z. Oridonin Delays Aging Through the AKT Signaling Pathway. Front Pharmacol. 2022;13:888247.
Magkouta S, Markaki E, Evangelou K, Petty R, Verginis P, Gorgoulis V. Decoding T cell senescence in cancer: Is revisiting required? Semin Cancer Biol. 2025;108:33–47.
Liu X, Si F, Bagley D, Ma F, Zhang Y, Tao Y, Shaw E, Peng G. Blockades of effector T cell senescence and exhaustion synergistically enhance antitumor immunity and immunotherapy. J Immunother Cancer. 2022;10(10):e005020.
Zelle-Rieser C, Thangavadivel S, Biedermann R, Brunner A, Stoitzner P, Willenbacher E, Greil R, Jöhrer K. T cells in multiple myeloma display features of exhaustion and senescence at the tumor site. J Hematol Oncol. 2016;9(1):116.
Liu X, Hartman CL, Li L, Albert CJ, Si F, Gao A, Huang L, Zhao Y, Lin W, Hsueh EC, Shen L, Shao Q, Hoft DF, Ford DA, Peng G. Reprogramming lipid metabolism prevents effector T cell senescence and enhances tumor immunotherapy. Sci Transl Med. 2021;13(587):eaaz6314.
Prieto LI, Sturmlechner I, Graves SI, Zhang C, Goplen NP, Yi ES, Sun J, Li H, Baker DJ. Senescent alveolar macrophages promote early-stage lung tumorigenesis. Cancer Cell. 2023;41(7):1261-75.e6.
Haston S, Gonzalez-Gualda E, Morsli S, Ge J, Reen V, Calderwood A, Moutsopoulos I, Panousopoulos L, Deletic P, Carreno G, Guiho R, Manshaei S, Gonzalez-Meljem JM, Lim HY, Simpson DJ, Birch J, Pallikonda HA, Chandra T, Macias D, Doherty GJ, Rassl DM, Rintoul RC, Signore M, Mohorianu I, Akbar AN, Gil J, Muñoz-Espín D, Martinez-Barbera JP. Clearance of senescent macrophages ameliorates tumorigenesis in KRAS-driven lung cancer. Cancer Cell. 2023;41(7):1242-60.e6.
Roth-Walter F, Adcock IM, Benito-Villalvilla C, Bianchini R, Bjermer L, Caramori G, Cari L, Chung KF, Diamant Z, Eguiluz-Gracia I, Knol EF, Jesenak M, Levi-Schaffer F, Nocentini G, O’Mahony L, Palomares O, Redegeld F, Sokolowska M, Van Esch B, Stellato C. Metabolic pathways in immune senescence and inflammaging: novel therapeutic strategy for chronic inflammatory lung diseases. An EAACI position paper from the Task Force for Immunopharmacology. Allergy. 2024;79(5):1089–122.
Tang X, Deng B, Zang A, He X, Zhou Y, Wang D, Li D, Dai X, Chen J, Zhang X, Liu Y, Xu Y, Chen J, Zheng W, Zhang L, Gao C, Yang H, Li B, Wang X. Characterization of age-related immune features after autologous NK cell infusion: Protocol for an open-label and randomized controlled trial. Front Immunol. 2022;13:940577.
Chibaya L, Murphy KC, DeMarco KD, Gopalan S, Liu H, Parikh CN, Lopez-Diaz Y, Faulkner M, Li J, Morris JP, Ho YJ, Chana SK, Simon J, Luan W, Kulick A, de Stanchina E, Simin K, Zhu LJ, Fazzio TG, Lowe SW, Ruscetti M. EZH2 inhibition remodels the inflammatory senescence-associated secretory phenotype to potentiate pancreatic cancer immune surveillance. Nat Cancer. 2023;4(6):872–92.
Zhou Z, Yao J, Wu D, Huang X, Wang Y, Li X, Lu Q, Qiu Y. Type 2 cytokine signaling in macrophages protects from cellular senescence and organismal aging. Immunity. 2024;57(3):513-27.e6.
Tommelein J, De Vlieghere E, Verset L, Melsens E, Leenders J, Descamps B, Debucquoy A, Vanhove C, Pauwels P, Gespach CP, Vral A, De Boeck A, Haustermans K, de Tullio P, Ceelen W, Demetter P, Boterberg T, Bracke M, De Wever O. Radiotherapy-Activated Cancer-Associated Fibroblasts Promote Tumor Progression through Paracrine IGF1R Activation. Cancer Res. 2018;78(3):659–70.
Meng J, Li Y, Wan C, Sun Y, Dai X, Huang J, Hu Y, Gao Y, Wu B, Zhang Z, Jiang K, Xu S, Lovell JF, Hu Y, Wu G, Jin H, Yang K. Targeting senescence-like fibroblasts radiosensitizes non-small cell lung cancer and reduces radiation-induced pulmonary fibrosis. JCI Insight. 2021;6(23):e146334.
Bientinesi E, Lulli M, Becatti M, Ristori S, Margheri F, Monti D. 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. 2022;206:111689.
Wang D, Xiao F, Feng Z, Li M, Kong L, Huang L, Wei Y, Li H, Liu F, Zhang H, Zhang W. Sunitinib facilitates metastatic breast cancer spreading by inducing endothelial cell senescence. Breast Cancer Res. 2020;22(1):103.
Bent EH, Gilbert LA, Hemann MT. A senescence secretory switch mediated by PI3K/AKT/mTOR activation controls chemoprotective endothelial secretory responses. Genes Dev. 2016;30(16):1811–21.
Zhao T, Li J, Chen AF. MicroRNA-34a induces endothelial progenitor cell senescence and impedes its angiogenesis via suppressing silent information regulator 1. Am J Physiol Endocrinol Metab. 2010;299(1):E110–6.
Yin K, Patten D, Gough S, de Barros GS, Chan A, Olan I, Cassidy L, Poblocka M, Zhu H, Lun A, Schuijs M, Young A, Martinez-Jimenez C, Halim TYF, Shetty S, Narita M, Hoare M. Senescence-induced endothelial phenotypes underpin immune-mediated senescence surveillance. Genes Dev. 2022;36(9–10):533–49.
Healey N. Senolytics target cellular senescence - but can they slow aging? Nat Med. 2024;30(10):2698–9.
Chang J, Wang Y, Shao L, Laberge RM, Demaria M, Campisi J, Janakiraman K, Sharpless NE, Ding S, Feng W, Luo Y, Wang X, Aykin-Burns N, Krager K, Ponnappan U, Hauer-Jensen M, Meng A, Zhou D. Clearance of senescent cells by ABT263 rejuvenates aged hematopoietic stem cells in mice. Nat Med. 2016;22(1):78–83.
Wang L, Lankhorst L, Bernards R. Exploiting senescence for the treatment of cancer. Nat Rev Cancer. 2022;22(6):340–55.
Chang Q, Jorgensen C, Pawson T, Hedley DW. Effects of dasatinib on EphA2 receptor tyrosine kinase activity and downstream signalling in pancreatic cancer. Br J Cancer. 2008;99(7):1074–82.
Shao Z, Wang B, Shi Y, Xie C, Huang C, Chen B, Zhang H, Zeng G, Liang H, Wu Y, Zhou Y, Tian N, Wu A, Gao W, Wang X, Zhang X. Senolytic agent Quercetin ameliorates intervertebral disc degeneration via the Nrf2/NF-κB axis. Osteoarthritis Cartilage. 2021;29(3):413–22.
Wang X, Yik-Lok Chung C, Yoshioka A, Hashimoto S, Jimbo H, Tanizawa H, Ohta S, Fukumoto T, Noma KI. Chemo-Senolytic Therapeutic Potential against Angiosarcoma. J Invest Dermatol. 2024;144(10):2285-97.e13.
Bogdanova DA, Kolosova ED, Pukhalskaia TV, Levchuk KA, Demidov ON, Belotserkovskaya EV. The differential effect of senolytics on SASP cytokine secretion and regulation of EMT by CAFs. Int J Mol Sci. 2024;25(7):4031.
Rajesh RU, Sangeetha D. Therapeutic potentials and targeting strategies of quercetin on cancer cells: Challenges and future prospects. Phytomedicine. 2024;133:155902.
Greenberg EF, Voorbach MJ, Smith A, Reuter DR, Zhuang Y, Wang JQ, Wooten DW, Asque E, Hu M, Hoft C, Duggan R, Townsend M, Orsi K, Dalecki K, Amberg W, Duggan L, Knight H, Spina JS, He Y, Marsh K, Zhao V, Ybarra S, Mollon J, Fang Y, Vasanthakumar A, Westmoreland S, Droescher M, Finnema SJ, Florian H. Navitoclax safety, tolerability, and effect on biomarkers of senescence and neurodegeneration in aged nonhuman primates. Heliyon. 2024;10(16):e36483.
Schmitt CA, Wang B, Demaria M. Senescence and cancer - role and therapeutic opportunities. Nat Rev Clin Oncol. 2022;19(10):619–36.
McHugh D, Durán I, Gil J. Senescence as a therapeutic target in cancer and age-related diseases. Nat Rev Drug Discov. 2024;24(1):57–71.
Lim JS, Lee DY, Kim HS, Park SC, Park JT, Kim HS, Oh WK, Cho KA. Identification of a novel senomorphic agent, avenanthramide C, via the suppression of the senescence-associated secretory phenotype. Mech Ageing Dev. 2020;192:111355.
Hu Q, Peng J, Jiang L, Li W, Su Q, Zhang J, Li H, Song M, Cheng B, Xia J, Wu T. Metformin as a senostatic drug enhances the anticancer efficacy of CDK4/6 inhibitor in head and neck squamous cell carcinoma. Cell Death Dis. 2020;11(10):925.
Widjaja AA, Lim WW, Viswanathan S, Chothani S, Corden B, Dasan CM, Goh JWT, Lim R, Singh BK, Tan J, Pua CJ, Lim SY, Adami E, Schafer S, George BL, Sweeney M, Xie C, Tripathi M, Sims NA, Hübner N, Petretto E, Withers DJ, Ho L, Gil J, Carling D, Cook SA. Inhibition of IL-11 signalling extends mammalian healthspan and lifespan. Nature. 2024;632(8023):157–65.
Ruscetti M, Leibold J, Bott MJ, Fennell M, Kulick A, Salgado NR, Chen CC, Ho YJ, Sanchez-Rivera FJ, Feucht J, Baslan T, Tian S, Chen HA, Romesser PB, Poirier JT, Rudin CM, de Stanchina E, Manchado E, Sherr CJ, Lowe SW. NK cell-mediated cytotoxicity contributes to tumor control by a cytostatic drug combination. Science. 2018;362(6421):1416–22.
Jochems F, Thijssen B, De Conti G, Jansen R, Pogacar Z, Groot K, Wang L, Schepers A, Wang C, Jin H, Beijersbergen RL, Leite de Oliveira R, Wessels LFA, Bernards R. The Cancer SENESCopedia: A delineation of cancer cell senescence. Cell Rep. 2021;36(4):109441.
Zhang Y, Xiao B, Yuan S, Ding L, Pan Y, Jiang Y, Sun S, Ke X, Cai L, Jia L. Tryptanthrin targets GSTP1 to induce senescence and increases the susceptibility to apoptosis by senolytics in liver cancer cells. Redox Biol. 2024;76:103323.
Admasu TD, Kim K, Rae M, Avelar R, Gonciarz RL, Rebbaa A, Pedro de Magalhães J, Renslo AR, Stolzing A, Sharma A. Selective ablation of primary and paracrine senescent cells by targeting iron dyshomeostasis. Cell Rep. 2023;42(2):112058.
Xing X, Tang Q, Zou J, Huang H, Yang J, Gao X, Xu X, Ma S, Li M, Liang C, Tan L, Liao L, Tian W. Bone-targeted delivery of senolytics to eliminate senescent cells increases bone formation in senile osteoporosis. Acta Biomater. 2023;157:352–66.
Zhang L, Wang C, Hu W, Bu T, Sun W, Zhou T, Qiu S, Wei M, Xing H, Li Z, Yang G, Yuan L. Targeted elimination of senescent cells by engineered extracellular vesicles attenuates atherosclerosis in ApoE(-/-) mice with minimal side effects. Theranostics. 2023;13(14):5114–29.
Muñoz-Espín D, Rovira M, Galiana I, Giménez C, Lozano-Torres B, Paez-Ribes M, Llanos S, Chaib S, Muñoz-Martín M, Ucero AC, Garaulet G, Mulero F, Dann SG, VanArsdale T, Shields DJ, Bernardos A, Murguía JR, Martínez-Máñez R, Serrano M. A versatile drug delivery system targeting senescent cells. EMBO Mol Med. 2018;10(9):e9355.
Parshad B, Baker AG, Ahmed I, Estepa-Fernández A, Muñoz-Espín D, Fruk L. Improved therapeutic efficiency of senescent cell-specific, galactose-functionalized micelle nanocarriers. Small. 2024;21(7):e2405732.
Chang M, Dong Y, Xu H, Cruickshank-Taylor AB, Kozora JS, Behpour B, Wang W. Senolysis Enabled by Senescent Cell-Sensitive Bioorthogonal Tetrazine Ligation. Angew Chem Int Ed Engl. 2024;63(9):e202315425.
Magkouta S, Veroutis D, Papaspyropoulos A, Georgiou M, Lougiakis N, Pippa N, Havaki S, Palaiologou A, Thanos DF, Kambas K, Lagopati N, Boukos N, Pouli N, Marakos P, Kotsinas A, Thanos D, Evangelou K, Sampaziotis F, Tamvakopoulos C, Pispas S, Petty R, Kotopoulos N, Gorgoulis VG. Generation of a selective senolytic platform using a micelle-encapsulated Sudan Black B conjugated analog. Nat Aging. 2025;5(1):162–75.
D’Ambrosio M, Gil J. Reshaping of the tumor microenvironment by cellular senescence: An opportunity for senotherapies. Dev Cell. 2023;58(12):1007–21.
Wang B, Kohli J, Demaria M. Senescent Cells in Cancer Therapy: Friends or Foes? Trends Cancer. 2020;6(10):838–57.
Acknowledgements
The figures in this article were supported by BioRender.
Funding
This work was supported by the National Natural Science Foundation of China (NO.82372943,NO.82073893), Hunan Youth Science and Technology Talent Project (NO.2023RC3074), Key R&D Program of Jiangxi Province, China (NO.20243BBI91007) and the Science Foundation of the AMHT Group (NO.2022YK04).
Author information
Authors and Affiliations
Contributions
CQ, ZWX, SRH and PL provided funding support and contributed to conceive, design, revision and oversight of the manuscript sections. BWL and ZGP wrote and revised the manuscript, designed the figures, and created the tables. HZ, NZ, and ZQL revised the manuscript. All authors read and approved the final manuscript.
Corresponding authors
Ethics declarations
Ethics approval and consent to participate
Not applicable.
Consent for publication
All authors consent to publication.
Competing interests
The authors declare no competing interests.
Additional information
Publisher’s Note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.
About this article
Cite this article
Liu, B., Peng, Z., Zhang, H. et al. Regulation of cellular senescence in tumor progression and therapeutic targeting: mechanisms and pathways. Mol Cancer 24, 106 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12943-025-02284-z
Received:
Accepted:
Published:
DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12943-025-02284-z