Next Article in Journal
Sex-Inclined Piwi-Interacting RNAs in Serum Exosomes for Sex Determination in the Greater Amberjack (Seriola dumerili)
Next Article in Special Issue
Roles of RNA Methylations in Cancer Progression, Autophagy, and Anticancer Drug Resistance
Previous Article in Journal
Gene Structural Specificity and Expression of MADS-Box Gene Family in Camellia chekiangoleosa
Previous Article in Special Issue
Re-Sensitizing Cancer Stem Cells to Conventional Chemotherapy Agents
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Potential of Senescence as a Target for Developing Anticancer Therapy

Department of Biochemistry, College of Natural Sciences, Kangwon National University, Chuncheon 24341, Republic of Korea
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(4), 3436; https://doi.org/10.3390/ijms24043436
Submission received: 1 December 2022 / Revised: 6 February 2023 / Accepted: 7 February 2023 / Published: 8 February 2023

Abstract

:
Senescence occurs in response to various stimuli. Senescence has attracted attention because of its potential use in anticancer therapy as it plays a tumor-suppressive role. It also promotes tumorigeneses and therapeutic resistance. Since senescence can induce therapeutic resistance, targeting senescence may help to overcome therapeutic resistance. This review provides the mechanisms of senescence induction and the roles of the senescence-associated secretory phenotype (SASP) in various life processes, including therapeutic resistance and tumorigenesis. The SASP exerts pro-tumorigenic or antitumorigenic effects in a context-dependent manner. This review also discusses the roles of autophagy, histone deacetylases (HDACs), and microRNAs in senescence. Many reports have suggested that targeting HDACs or miRNAs could induce senescence, which, in turn, could enhance the effects of current anticancer drugs. This review presents the view that senescence induction is a powerful method of inhibiting cancer cell proliferation.

1. Characteristics of Senescence

Cellular senescence is a state of permanent growth arrest and is thought to be an essential anticarcinogenic barrier in normal cells. Cellular senescence protects tissues from developing cancer by arresting the cell cycle [1,2,3,4,5,6,7,8]. Senescence can also create an inflammatory microenvironment favoring the initiation and progression of various age-related diseases including cancer [9]. Thus, senescence affects tumorigenesis in a context-dependent manner.
Senescent cells (SnCs) are enlarged and flattened and display increased cytoplasmic granularity. These cells remain viable and metabolically active and are different from quiescent cells, which are in a temporary state of growth arrest [10]. SnCs induced by different stimuli share the common characteristics of stable growth arrest, anti-apoptosis, persistent DNA damage response (DDR) signaling, changes in chromatin structures (senescence-associated heterochromatin foci), decreased nuclear lamin-B1 levels, and an increased expression of cyclin-dependent kinase (CDK) inhibitors (p16INK4a, p21Cip1/Waf1) [11,12,13], p53 [14], and senescence-associated β-galactosidase activity (SA-β-gal) [15,16]. Senescence is also characterized by changes in mitochondria dynamics, structure, and function [17]. Impaired mitochondrial dynamics lead to senescence [17]. Figure 1 shows the general characteristics of senescent cells. Senescent cells are unique in that they eventually stop multiplying but do not die off when they should. Senescent cells can remain active and spread senescence. Figure 1 also shows that senescent cells can spread senescence in paracrine and juxtacrine fashions.

2. Factors Inducing Senescence

Radiation has been shown to induced senescence [18,19,20,21,22]. Radiation induced senescence by increasing the expression of STAT3 and autophagic flux, such as Beclin1 and LC-3 II in lung adenocarcinoma cells [18]. This implies a close relationship between senescence and autophagy. Senescence occurs in response to chemotherapy, known as therapy-induced senescence (TIS) [14,16]. Low doses of chemotherapy trigger senescence, while higher doses trigger apoptosis. Paclitaxel (PTX)-induced senescence involved the increased expression of p53 in bladder cancer cells [14]. The accumulation of cryptochrome 1 (CRY1) in cisplatin-resistant cells prevented PTX-induced senescence by promoting p53 degradation [14]. Doxorubicin-induced senescence in cardiac fibroblasts was accompanied by increases in p53 and SA-β-gal activity in an IL-1β-dependent manner [16]. Etoposide, an inhibitor of topoisomerase, induced senescence by increasing the expression of p53 and p21 via DNA protein kinase (DNA-PK)-checkpoint kinase 2 (Chk2 pathway) in adrenocortical carcinoma cells [23].
The combination of a mouse double minute 2 (MDM2) antagonist with an inhibitor of the aurora kinase A (AURKA) activated p53, and induced growth arrest and the clearance of melanoma cells by antitumor leukocytes [24]. P53 displays diverse roles, including apoptosis and senescence. P53 activation in response to DNA damage causes growth arrest, allowing for DNA repair, or directs senescence or apoptosis [25]. For DNA repair, senescence, but not apoptosis, is induced by p53. P53 increases the expression of p21 and p16, which, in turn, causes cell cycle arrest to induce senescence [25]. These findings suggest a role of p53 in chemotherapy-induced senescence. It is probable that low doses of chemotherapy drugs cause senescence via p53, while higher doses of chemotherapy drugs cause apoptosis via p53. Platinum-based drugs, such as cisplatin, induce senescence by the upregulation of senescence-related genes (p53, p21, and p16) in seminoma cells [26]. Cisplatin induced DNA damage and increased ROS level [26].
The irreversible senescence is mediated by INK4a/Rb and p53 pathways, and the reversible senescent phenotype is mediated by p53. This suggests that the p53 pathway could be effectively harnessed as a therapeutic intervention to trigger senescence and ultimately suppress tumorigenesis.
Punicalagin (PUN), an active component from pomegranate, induced the senescence of human papillary thyroid carcinoma cells by increasing the expression of p21, interleukin-6 (IL-6), and IL-1β via nuclear factor NF-κB [27]. Chemotherapy-induced senescence involved the increased expression of anti-apoptotic proteins, such as BCL-2 and the induction of p53-mediated DNA damage response in T-cell lymphoma cell lines [28]. Senescent cells could persist without cell death owing to the upregulation of anti-apoptotic proteins such as BCL-2, and the downregulation of BAX and p53 in human dermal papilla cells [29]. This implies the role of senescence in anticancer drug resistance. Doxorubicin (DOX) induced endothelial cell (EC) senescence based on SA-β-gal activity, the upregulation of senescence markers (p53, p21, and cyclinD1), and the elevated expression of SASP [30].
Senescence can also be induced by inhibitors of CDK4/6 [31], polo and aurora kinases [32]. The CDK4/6 inhibitor (palbociclib) enhanced the anticancer effect of the I kappa B kinase (IKK)β inhibitor Bay 11-7082 in hepatocellular carcinoma [31]. The inhibition of aurora kinases induced polyploidy and the ATM/Chk2 DNA damage response, which mediated senescence and an NF-κB-related, senescence-associated secretory phenotype (SASP) [32]. This implies that AURK inhibitors can also induce senescence in a p53-independent way. The inhibition of HDAC2 or HDAC7 induced senescence by increasing the expression of p16 and p21 in dermal fibroblasts [33]. The downregulation of DNA methyl transferase 2 causes telomere shortening and senescence [34].
Senescence can also be induced by damage to mitochondria [35,36,37,38,39], which in turn, leads to the increased production of reactive oxygen species (ROS) [40] and DNA damage [41,42]. Mitochondrial dysfunction was shown to induce senescence via AMPK-p53 activation, and modified SASP without IL-1 in human fibroblasts [36]. Cathepsin F mediated the effect of DNA damage on the induction of senescence in human skin fibroblasts and keratinocytes [42]. Cellular senescence can also be induced by oxidative stress [43] and telomeric shortening [44]. Oxidative stress induced senescence by increasing SASP in human dental pulp cells [43]. Bortezomib, an anticancer drug, induced senescence by promoting telomeric shortening and increasing the expression of p53 and p21 in lung cancer cells [44]. Oxidative stress induced senescence and epithelial-to-mesenchymal transition (EMT) in amnionic epithelial cells (AEC) via p38MAPK activation [45]. Since therapeutic resistance eventually develops, it is probable that senescence induced by therapy may induce therapeutic resistance.
Figure 2A shows various factors inducing senescence and that senescence can promote both tumor suppression and tumor progression via SASP. Figure 2B shows therapy-induced senescence (TIS) and the mechanisms involved.

3. Oncogene-Induced Senescence

Oncogenes are known to induce senescence [45,46]. A combination of oncogenes (CTNNB1, TERT, and MYC) induced senescence in human fibroblasts and primary hepatocytes [47]. Transformed cells after senescence became more aggressive and display high invasion potential and anticancer drug resistance [46]. Continuous functional changes in senescent cells, and the permanent need to actively maintain senescence-supporting transcription puts the stability of cell cycle arrest at risk. Oncogene-induced senescence (OIS) can induce permanent growth arrest. OIS can also enable senescent cells to evade the potential toxicity of therapeutics, allowing for the eventual re-emergence or escape from senescence that could lead to disease recurrence. Senescence is potentially reversible through the inactivation of p53, p16(INK4A) and/or Rb, overexpression of Cdc2/cdk1, and the survival of cancer stem cells.
MYC is critical for the immune evasion of triple-negative breast cancer cells (TNBCs) [48]. TNBCs with high MYC are resistant to immune checkpoint inhibitor (ICI) therapy [48]. IL-6, a component of SASP, mediates the effect of MYC on somatic cell reprogramming (transformation) [49]. MYC is necessary for the SASP-induced somatic cell programming, such as the generation of induced pluripotent stem cells [49].
Oncogene-induced senescence (OIS) increased the production of ROS, which, in turn, caused DNA damage and DNA double-strand breaks (DSB) [5,50]. OIS increased cytoplasmic release of cytochrome c, the expression of BAX, and the cleavage of poly(ADP-ribose) polymerase (PARP) in human keratinocytes [50]. During OIS, Toll-like receptor 2 (TLR2) was shown to mediate senescence in vitro and in murine models [3]. TLR2 impairs early lung cancer progression by activating cell cycle arrest and the pro-inflammatory phenotype [3]. TLR2 promoted cell cycle arrest by increasing the expression of tumor suppressors (p53-p21CIP1, p16INK4a, and p15INK4b) by inducing the acute-phase serum amyloids A1 and A2 (A-SAAs) [51]. These reports suggest that OIS involves the initial activation of tumor-suppressive responses. It is also noteworthy that oncogenes that induce senescence can also promote tumor initiation by inducing immune evasion [48].
Oncogenic Ras transformation resulted in the upregulation of the cell cycle inhibitors p15Ink4b, p16Ink4a, and p19Arf; Rho-associated protein kinase (ROCK); SASP; and the downregulation of p-AKT in skin keratinocytes [52]. The upregulation of ROCK activity initiated a senescence response characterized by cell enlargement, growth inhibition, the upregulation of SA-β-gal activity, and the release of multiple pro-inflammatory factors, such as colony-stimulating factors (CSFs) and matrix metalloproteinase 9 (MMP-9) [52]. p53 and retinoblastoma (RB) act as master regulators of OIS [53,54]. The inactivation of p53 or p16 blocked the ability of oncogenic Ras to induce senescence in mouse fibroblasts [53]. Ras induced cellular senescence by promoting caspase-4 inflammasomes in a p53-dependent manner [54]. p53 was shown to be necessary for mitochondrial elongation in H-RAS-induced cellular senescence and in the replicative senescence of normal human cells [55]. Ras (mutant HRAS and NRAS)-induced senescence involved ROS and EGFR-argonaute 2 (AGO2) signaling in various human cancer cells, including HeLa, melanoma cells, and bladder cancer cells [56]. AGO2 bound to Ras and the loss of AGO2 inhibited cancer cell proliferation, senescence, and Ras signaling [56]. Thus, targeting AGO2 could eliminate senescent cancer cells. These reports suggest that inhibiting senescence induced during oncogenic transformation is necessary for overcoming tumorigenesis.
The immediate early response gene 2 (IER2) has been associated with the progression of several types of cancer. The sustained expression of IER2 induced senescence in a subset of melanoma cells in a p53/mitogen-activated protein kinase (MAPK)/AKT-dependent manner [57]. The senescent cells produced extracellular factors, such as osteopontin (OPN). OPN secreted by IER2-expressing senescent cells stimulated the migration and invasion of non-senescent melanoma cells [57]. The downregulation of OPN enhanced sensitivity to 5-fluorouracil (5-FU) in HepG2 cells [58]. This suggests that OIS leads to the secretion of extracellular factors that can promote cancer cell proliferation and anticancer drug resistance. Figure 3 shows the effects of oncogenes on senescence and the mechanisms of OIS.

4. Roles of SASP in Cancer Cell Proliferation and Therapeutic Resistance

Senescence inhibits the replication of damaged and precancerous cells in the short-term but is implicated in various diseases owing to SASP secretions. Senescent cells can induce other non-senescent cells to undergo senescence (secondary senescence) via SASP. Secondary senescence occurs via paracrine mechanisms and cell-to-cell contacts (juxtacrine) (Figure 1). The SASP includes pro-inflammatory cytokines, chemokines, MMPs, bioactive lipids, and growth factors (Figure 4) [59,60,61,62], as well as vascular endothelial growth factor (VEGF), hepatocyte growth factor (HGF), interleukins (IL-1a, IL-6, IL-8, IL-10, IL-13, IL-15), and CC/CXC chemokines (CXCL1, CXCL2, CXCL5, CXCL11, CXCL12, CCL2, CCL20) (Figure 4).
The SASP also includes extracellular vesicles (sEVs), which mediate cell-to-cell communication through various types of cargos, including proteins, lipids, and miRNAs. sEVs activate macrophages by increasing cellular levels of pro-inflammatory cytokines, such as IL-1β and IL-6, via NF-κB signaling [63]. sEVs released by senescent vascular smooth muscle cells (VSMCs) induced the secretion of IL-17, interferon γ (INFγ), and IL-10 by T cells, and tumor necrosis factor α (TNFα) by monocytes. sEVs released by senescent VSMCs promoted M1 macrophage polarization favoring the pro-inflammatory phenotype [64]. Thus, SASP could modulate immune responses.
Ras and tumor protein 53 (TP53) mutations, and cellular senescence are frequently detected in zebrafish imaging [63]. Additional TP53 mutation changes OIS from a tumor suppressor to a driver [65]. Newly transformed cells with an RASG12V mutation became senescent and were eliminated from the epithelia. This was prevented by adding a TP53 gain-of-function mutation (TP53R175H). The surviving RASG12V-TP53R175H double-mutant cells secreted SASP-related inflammatory molecules that converted neighboring normal cells into SASP factor-secreting senescent cells, generating a heterogeneous tumor-like cell mass [65]. Thus, OIS involves the secretion of the SASP, which further spreads senescence through cellular interactions.
Senescence and the associated SASP can affect cells within the tumor microenvironment (TME). The SASP was shown to promote tumorigenesis [66], EMT [67,68,69], cancer stemness [70,71], anticancer drug resistance [72,73,74], angiogenesis [75,76,77,78], immune suppression to promote tumorigenesis [79,80], and tumor suppression by the immune clearance of senescent cells [81]. Figure 4 shows the roles of the SASP, including tumor suppression and tumor promotion.
The SASP increased the expression of lipocalin-2 (LCN2) to enhance breast cancer cell proliferation and migration [66]. High levels of LCN2 could predict the poor prognosis of patients with breast cancer [66]. The inactivation of LCN2 enhanced the response to chemotherapeutic drugs in mouse models of breast cancer [64]. This implies the roles of the SASP in cancer cell proliferation and anticancer drug resistance. Senescent lung fibroblasts (SLF) can promote the production of IL-6 and IL-8, which are effectively inhibited by overexpressing Klotho (Figure 4) [68]. Conditioned medium (CM) of SLFs enhanced the migration of lung cancer cells and increased the expression of the phospho-signal transducer and activator of T cell 3 (p-STAT3) but decreased the expression of P53 and E-cadherin [68]. Klotho inhibited the extracellular release of IL-6 and IL-8, which influenced STAT3 activation, P53 expression, and EMT in lung cancer cells, finally inhibiting lung cancer cell growth and migration [68]. CM from senescent MPM (malignant pleural mesothelioma) cells trigger, via STAT3 activation, the emergence of EMT-like, clonogenic, and chemo-resistant cell subpopulations, expressing high levels of ALDH (aldehyde dehydrogenase) activity (ALDH(bright) cells) [73]. Thus, SASP can enhance EMT, tumorigenic potential, and the anticancer drug resistance of senescent cancer cells.
Senescent fibroblasts promote angiogenesis by secreting VEGF, a component of the SASP [76]. The increased expression of VEGF resulted from senescence induced by Ras expression or p16 overexpression [76]. This suggests that senescence can also promote tumorigenesis. CM of IL-1β (a component of SASP)-treated chondrocytes enhanced migration and the tube-forming potential of human umbilical vein endothelial cells (HUVEC) in a growth differentiation factor 15 (GDF15)-dependent manner [78]. CDK4/6 inhibitors induced senescence and increased the production of pro-angiogenic SASP factors such as VEGF, basic FGF (bFGF), PDGF, as well as multiple MMPs, resulting in vascular remodeling in preclinical models of pancreatic ductal adenocarcinoma (PDAC) [82]. This vascular remodeling enhanced the efficacy of anti-PD-1 by improving gemcitabine delivery to the tumors and promoting cytotoxic T-cell infiltration [73]. This implies that TIS could enhance the efficacy of current anticancer therapy by inducing the SASP-promoted angiogenesis.
Cancer cells emerging from senescence display higher anticancer drug resistance than cancer cells without senescence [46]. Senescent cells display resistance to apoptosis owing to high level of BCL-2 [83], suggesting the role of senescence in therapeutic resistance. Lapatinib and fulvestrant (EGFR and HER2 inhibitors) treatment induced apoptosis in chemotherapy-induced senescent breast cancer cells [84]. Senescence induced by radiation, oncogenes, and chemicals increased the expression of BCL-2 in non-transformed retinal pigment endothelial cells and fibroblasts [83]. Thus, targeting BCL-2 or other anti-apoptotic proteins can eliminate senescent cancer cells (senolysis or senolytic therapy).
IL-8, a component of the SASP, was necessary for the increased expression of multi-drug resistance 1 (MDR1)/ABCB1 by chemotherapy in urothelial carcinoma endothelial cells [85]. Targeting ABCB1 and/or IL-8 in tumor endothelial cells may overcome chemotherapeutic resistance and improve clinical outcomes. Low levels of IL-8 are correlated with longer overall patient survival [86]. The IL-8/STAT3 axis is necessary for hepatocellular carcinoma metastasis mediated by Snail and Twist 1 [86]. Anticancer drug resistance is closely related to the enhanced metastatic potential of cancer cells [87]. Since IL-8 is a component of the SASP, it is probable that the SASP may confer anticancer drug resistance.
Cancer stem cells (CSCs) play a critical role in resistance to chemotherapy. Doxorubicin increased the expression of stemness-related genes, such as epithelial cell adhesion molecule (EpCAM), cytoskeletal 19 (CK19), annexin A3 (ANXA3), and the multidrug-resistance-related gene ABCG2 in hepatocellular carcinoma cells (HCCs) [71]. Platinum induced senescence and cancer stemness in ovarian cancer cells by enhancing aldehyde dehydrogenase 1A1 (ALDH1A1) activity [88]. These reports suggest that doxorubicin and platinum induced the SASP to promote cancer stemness and therapeutic resistance.
CD44 conferred doxorubicin resistance by upregulating MDR1 expression in osteosarcoma cells [89]. CD44, a cell surface marker of CSCs [90], conferred 5-FU resistance by increasing intracellular glutathione and suppressing the drug-induced production of ROS in gastric cancer cells [91]. Thus, the acquisition of cancer-stem-cell-like properties by senescence could induce anticancer drug resistance. The IL-6/STAT3 axis was necessary for chemo resistance in glioblastoma stem cells [92]. IL-6/IL-8-JAK2 signaling activated by cancer-associated fibroblasts (CAFs) conferred resistance to a bromodomain and extra-terminal (BET) inhibitor in colorectal cancer cells [93]. The NF-κB-IL-6-STAT3 signaling pathway increased cancer stemness and the tumorigenic potential of glioma cells [94]. Since IL-6/IL-8 is a component of the SASP, it is probable that the SASP may confer chemotherapeutic resistance by enhancing cancer-stem-cell-like properties. Erlotinib increased the expression of p21, p53, and IL-6 in keratinocytes [95]. Thus, erlotinib resistance could be induced by senescence. Erlotinib resistance involved the activation of STAT3 by EGFR signaling in osteosarcoma cells [96]. Since STAT3 can enhance cancer-stem-cell-like properties, erlotinib resistance may be associated with the enhanced cancer-stem-cell-like properties. These reports suggest the role of the SASP in anticancer drug resistance in relation to cancer stemness.
A persistent inflammatory state evokes compensatory immunosuppression, which impairs the functions of effector immune cells, such as macrophages, T cells, and natural killer (NK) cells. DNA damage induced the production of amphiregulin (AREG), a component of the SASP, in stromal cells, which increased the expression of immune checkpoint molecule programmed death-ligand 1 (PD-L1) in cancer cells and induced immune suppression in the TME to promote tumor formation [72]. PD-L1 is overexpressed in cancer cells and confers resistance to cancer therapy. Thus, the SASP can induce immune suppression to promote tumor progression.
The SASP of senescent cancer cells enhanced immunosenescence (immune suppression) in the TME of peritoneal carcinomatosis (PC) to promote tumor progression [97]. Doxorubicin-induced senescence promoted the recruitment of Treg cells and myeloid-derived suppressor cells to mediate apoptotic resistance in diffuse large B-cell lymphoma (DLBCL) via pro-inflammatory cytokines, such as IL-2, IL-6, IL-8, IL-10, IL-35, TGFβ, and VEGF [77]. Senescent cancer cells were shown to mediate pro-tumorigenic effect by inhibiting the infiltration of CD8+ T cells via CXCL12/CXCR4 signaling [98]. Thus, the SASP of senescent cancer cells could promote cancer recurrence by exerting immune suppression.
IL-8 secreted by hypoxic macrophages enhanced esophageal cancer cell proliferation and metastasis by increasing the expression of PD-L1 in esophageal cancer cells [99]. PD-L1 expression was positively correlated with high levels of the SASP in a mouse model [100]. This implies that IL-8, a SASP component, could induce immune evasion by inhibiting CTL activity against PD-L1 expressing cancer cells. These reports imply the role of PD-L1 in senescence. PD-L1 depletion induced senescence by increasing the expression of stimulator of interferon genes (STING) in human melanoma cells and lung cancer cells [101]. This suggests that the blockade of PD-L1 could enhance the sensitivity of cancer cells to cancer therapeutics by enhancing senescence. Thus, the SASP could induce immune suppression by increasing the expression of PD-L1, which leads to tumor promotion.
Senescent cancer cells in tumors are metabolically active and can promote tumor relapse, metastasis, and resistance to therapy through the SASP [46,102,103,104,105,106,107]. The pro-inflammatory SASP promoted the secretion of pro-tumorigenic factors, such as IL-1α and IL-6, and a less inflammatory SASP promoted the clearance of senescent cells (senescence surveillance) by the immune system [108]. This implies the role of the SASP in tumor suppression. Senescent cells can undergo conversion to an immunogenic phenotype that enables them to be eliminated by the immune system. If senescent cancer cells are not cleared, they acquire the potential to recapture the proliferative state. The impaired surveillance of pre-malignant senescent cells resulted in the development of murine hepatocellular carcinoma (HCC) [109]. Senescent cells were reported to secrete various inflammatory cytokines, such as IL-1β, IL-6, IL-8, TGF-β, granulocyte-macrophage colony-stimulating factor (GM-CSF), and MCP-1, to induce senescence surveillance in p16-positive myeloid cells [110]. During Ras-induced senescence, Cyclooxygenase-2 (COX-2) promoted senescence surveillance and tumor suppression by regulating multiple components of the SASP, such as prostaglandin E2 (PGE2) and IL-6 [81]. Thus, the SASP suppressed tumor progression by inducing senescence surveillance.

5. Role of Autophagy in Senescence

Radiation-induced senescence involves increased autophagy in lung cancer cells [18]. Senescent cells appear large, flattened, and irregularly shaped, which is attributed to increased mammalian target of rapamycin (mTOR) signaling [111,112]. Derivatives of indolo [2,3-a]pyrrolo [3,4-c]carbazole induce senescence by modulating AKT/mTOR/S6K signaling in breast cancer cells, lung cancer cells, and colon cancer cells [112]. These reports suggest a close relationship between autophagy and senescence. Autophagy is a self-digesting cellular process that allows cells to sequester cytoplasmic contents through the formation of double membrane vesicles (autophagosomes). As with senescence, autophagy is stimulated by oxidative stress, chemotherapy, oncogene activation, and various other conditions [113]. Cellular senescence and autophagy commonly induce oxidative stress, DNA damage, telomere shortening, and oncogenic stress. Thus, senescence and autophagy are interrelated. Complete autophagy can lead to cell death. Defective autophagy is linked to various human diseases such as cancer, neurodegeneration, microbial infection, and aging [114]. SIRT3 promotes autophagy to suppress doxorubicin-induced senescence by inactivating the PI3K/Akt/mTOR pathway in lung cancer cells [115]. However, it is still unclear whether autophagy has a positive or negative effect on senescence.
Receptor tyrosine kinase inhibitors (RTKi) induced protective autophagy for cell survival and anticancer drug resistance in neuroblastoma cells [116]. Autophagy promoted the acquisition of cancer-stem-cell-like properties [117]. The inhibition of autophagy by siATG5 or 3-MA enhanced the sensitivity of liver cancer stem cells to doxorubicin [117]. It is interesting to examine whether autophagy can induce senescence in liver cancer cells. Resistance to clinically used anticancer drugs developed in prostate cancer cells and breast cancer cells due to autophagy in [118]. Thus, the inhibition of autophagy could overcome resistance to these clinically used anticancer drugs. Cancer/testis antigen CAGE promotes anticancer drug resistance by promoting autophagy in gastric cancer cells [119]. Icotinib resistance resulted from the increased expression of autophagic flux (ATG3, ATG5, and ATG7) via STAT3-FOXM signaling in lung cancer cells [120]. Chloroquine (CQ), an inhibitor of autophagy, enhanced the sensitivity of lung cancer patients to icotinib [120]. CAGE was shown to bind to Beclin1 (a mediator of autophagy) and confer resistance to various anticancer drugs in non-small-cell lung cancer cells (NSCLCs) [121]. This suggest that autophagy promotes anticancer drug resistance.
Erlotinib induced autophagy to promote erlotinib resistance via 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 (PFKFB3) in NSCLCs [122]. Wingless-related integration site 5A (Wnt5A)-induced autophagy mediated doxorubicin resistance in osteosarcoma cells [123]. The inhibition of autophagy sensitized glioblastoma cells to Src tyrosine kinase inhibitors such as Si306 and pro-Si306 [124]. These reports suggest that targeting autophagy may enhance the sensitivity of cancer cells to anticancer drugs. These reports also imply that anticancer drugs induce therapy resistance by promoting autophagy in association with senescence.
The inhibition of STAT3 restored sensitivity to icotinib by reducing autophagy [120]. Since STAT3 regulates the expression of IL-6 and IL-8, components of the SASP, it is probable that the SASP may regulate resistance to icotinib in association with senescence. Autophagy induction during mitotic slippage involved the autophagy activator AMP-dependent kinase (AMPK) and the endoplasmic reticulum stress response protein kinase R-like ER kinase (PERK) [125]. The pharmacologic inhibition of autophagy or downregulation of ATG5 led to a bypass of G1 arrest senescence, reduced SASP-associated paracrine tumorigenic effects, and increased DNA damage after S-phase entry with a concomitant increase in apoptosis in various cancer cells, including osteosarcoma cells, colon cancer cells, pancreatic cancer cells, and breast cancer cells [125]. These reports suggest that autophagy can promote senescence.
Oncogene HRAS-induced senescence involved the upregulation of genes involved in autophagy, such as unc51-like autophagy activating kinase 3 (ULK3), in human diploid fibroblast IMR90 cells [126]. HRAS-induced senescence was negatively regulated by the PI3K-mTOR signaling pathway [124]. Hyperglycemia (HG) (25 mM) increased the expression of Beclin-1, ATG 5, 7, and 12, the generation of microtubule-associated proteins 1A/1B light chain 3B (LC3-II), and autophagosome formation that was correlated with the development of cellular senescence [127]. N-acetyl-L-cystein (NAC) inhibited the effect of HG on senescence by decreasing ROS formation (Figure 5) [127]. X-ray radiation induced senescence and increased the expression of STAT3, Beclin1, and the LC3-II/LC3-I ratio in lung adenocarcinoma cells [18]. These reports suggest a crosstalk between autophagy and senescence. Figure 5 shows that complete autophagy may induce cell death, while impaired autophagy induces senescence. Figure 5 also shows the mechanisms of autophagy-induced senescence.

6. MicroRNAs in Senescence

MiRNAs, noncoding RNAs, are families of 21–25 nucleotides and small RNAs. These RNAs negatively control gene expression by binding to the 3′ untranslated region (UTR) of mRNA. Since miRNA targets multiple genes, miRNAs are involved in various diseases. Senescence increases the secretion of EVs, components of the SASP, and EVs can transport proteins and miRNAs. Since the SASP mediates cellular interactions, miRNAs may play critical roles in the spread of senescence. miR-34a targets Wnt and Notch pathways, as well as BCL2, and was found in EVs secreted from subjects with senescence [128]. Thus, targeting miR-34a may improve senescence. miR-485-5p targeted CDK16 to decrease the expression of C-MYC and PD-L1, which, in turn, induced the senescence-associated phenotypes in lung cancer cell lines [129]. Therefore, CDK16 can be a target for the induction of senescence in cancer cells. Thus, an miR-485-5p mimic could be employed for inducing senescence in cancer cells.
Doxorubicin increased the number of senescence-associated secretory phenotypes but decreased the expression of miR-199a-3p in cardiomyocytes [130]. The overexpression of miR-199a-3p inhibited SASP production and the spread of senescence by decreasing the expression of GATA-4 in cardiomyocytes [130]. This suggests that an miR-199a-3p mimic could be developed as an anticancer drug owing to its role in regulating SASP production.
Levels of miR-200 family members are decreased in the late stage of tumorigenesis [67]. miR-200 full knock out (FKO)-associated senescence in cancer epithelial cells recruited stromal cells in the TME [65]. The genetic deletion of all miR-200s in human gastric cancer cell lines induced potent morphological alterations and G1/S cell cycle arrest, increased SA-β-gal activity, and activated TGF-β/TNF-α signaling and aberrant metabolism, collectively resembling the senescent phenotype [67]. MiR-200b is negatively regulated by CAGE in anticancer drug-resistant melanoma cells [131]. Thus, CAGE can confer anticancer drug resistance by inducing senescence in melanoma cells by decreasing miR-200b expression. Thus, inhibitors of the miR-200 family can enhance the effects of anticancer drugs by inducing senescence.
The overexpression of miR-335 and miR-34a induced the premature senescence of young mesangial cells via the direct suppression of superoxide dismutase 2 (SOD2) and thioredoxin reductase 2 (Txnrd2), with a concomitant increase in ROS in mesangial cells [132]. ROS induced DNA damage in osteosarcoma cells, which, in turn, increased miR-335 expression in a p53-dependent manner [133]. miR-335 and p53 cooperated in a positive feedback loop to drive cell cycle arrest in osteosarcoma cells [133]. Thus, miR-335 activation plays an important role in the induction of p53-dependent senescence after DNA damage [133] and an miR-335 mimic could enhance the effects of anticancer drugs by inducing senescence in cancer cells.
The overexpression of miR-217 promoted vascular endothelial cell senescence by targeting the NAD-dependent deacetylase sirtuin 1 (SIRT1)/p53 signaling pathway [134]. MiR-21-5p/miR-217 carried by sEV from senescent cells spread pro-senescence signals by targeting DNA methyl transferase 1 (DNMT1) and NAD-dependent deacetylase sirtuin 1 (SIRT1) and inhibiting endothelial cell proliferation [135]. Thus, it is probable that an miR-217 mimic could prevent endothelial cells from promoting cancer cell proliferation.
MiR-335, upregulated in senescent normal fibroblasts and CAFs, modulated the secretion of SASP factors and enhanced the invasion potential of squamous cell carcinoma in co-cultures by suppressing the expression of phosphatase and tensin homolog (PTEN) [136]. Additionally, elevated levels of COX-2 and PGE2 were observed in senescent fibroblasts, and the inhibition of COX-2 by celecoxib reduced the expression of miR-335, restored PTEN expression, and decreased the pro-tumorigenic effects of the SASP [136]. miR-335 mediated the pro-tumorigenic effect of the SASP by decreasing the expression of PTEN and SASP [136]. It is probable that an miR-335 inhibitor could be developed as an anticancer drug that could enhance current anticancer therapy. Table 1 shows the roles of miRNAs in senescence and the mechanisms.

7. Role of HDACs in Senescence

Histone modifications regulate gene expression. Tet methylcytosine dioxygenase 2 (TET2) inhibited PD-L1 gene expression by recruiting histone deacetyalses (HDACs) (HDAC1 and -2) in MDA-MB-231 cells [137]. This implies that HDACs can regulate the immune evasion of cancer cells. Since the SASP regulates immune evasion [79], it is probable that HDACs may regulate senescence. Histone methylation (H3K27me3) in the promoter region of cell-cycle-dependent kinase N2a (CDKN2a) by enhancer of zeste homolog 2 (Ezh2), a histone methyl transferase, inhibited replicative senescence in atrial fibroblasts [138]. These reports suggest the roles of histone modifications in senescence.
4-Phenylbutyric acid (4-PBA), an HDAC inhibitor (HDACi), induced EMT in gastric cancer cell lines by upregulating IL-8 [139]. IL-8, a component of the SASP, is known to mediate immune suppression and the recruitment of MDSCs (Figure 4). This further suggests the role of HDACs in senescence. SAHA (vorinostat), an HDACi, or the specific downregulation of HDAC2 or HDAC7 by siRNA, induced senescence in dermal fibroblasts by increasing the expression of IL-6, IL-8, MMP-1/-3, and IL-1β [33]. The ectopic overexpression of HDAC7 by lentiviral transduction in pre-senescent dermal fibroblasts extended their proliferative lifespan [33]. These reports suggest that HDACs could inhibit senescence.
HDAC1/2-deficient epidermis displayed elevated acetylated p53 and the increased expression of the senescence gene p16 in pre-basal cell carcinoma [140]. This suggests the roles of HDAC1/2 as negative regulators of senescence. CAGE was shown to bind to HDAC2 and directly decreases the expression of p53 and confers resistance to anticancer drugs in melanoma cells [141]. This suggests that HDAC2 may confer resistance to anticancer drugs by suppressing p53-mediated senescence. HDAC2-specific inhibitors may enhance the effects of anticancer drugs by inducing senescence. The overexpression of HDAC2 inhibited cigarette smoke (CSE)-induced senescence by decreasing muscle ring finger protein 1 (MURF1), ATROGIN1, P53, and P21 expression in C2C12 cells [142]. These reports suggest the role of HDAC2 in senescence, cancer cell proliferation, and anticancer drug resistance.
HDAC6 differs from the other HDAC family members not only in terms of its cytoplasmic localization but also in terms of its substrate repertoire and hence cellular functions. HDAC6 mostly deacetylates cytoplasmic proteins [143]. An HDAC6 knockout mice study showed the role of HDAC6 in cell survival under stress conditions [144]. The downregulation of HDAC6 inhibited the proliferation and migration of glioblastoma cell lines and the sonic hedgehog (shh) pathway, and decreased autophagy [145]. ACY-1215 (ricolinostat, a specific HDAC6 inhibitor) suppresses glioblastoma growth by inhibiting TGF-β/SMAD2 signaling to decrease the expression of p21 [146]. This suggests the role of HDAC6 in promoting senescence. Given the fact that HDAC6 regulated autophagy [145], HDAC6 could induce senescence by promoting autophagy.
HDAC6 inhibitors decreased the expression of PD-L1 in urothelial cancer cell lines [147]. Since SASP regulates the expression of PD-L1 [72,99,100], HDAC6 inhibition can suppress the immune evasion of cancer cells by decreasing autophagy and senescence. High levels of HDAC6 are observed in colon cancer patients with KRAS activation mutation [148]. Oncogenic KRAS contributes to SAHA resistance by upregulating HDAC6 and C-MYC expression [148]. Oncogenes can induce senescence (Figure 2). It is probable that K-RAS enhances SAHA resistance by promoting autophagy and senescence. The acetylation of lysine 104 by HDAC6 inhibition attenuated KRAS-transforming activity by interfering with guanine nucleotide exchange factor (GEF)-induced nucleotide exchange in various cancer cells [149]. Thus, targeting HDAC6 may overcome the oncogenic effects of KRAS by regulating senescence. These reports implicate the role of HDAC6 in autophagy and senescence.
Azaindolyl sulfonamide, a potent HDAC6 inhibitor, induced G2/M arrest and senescence in temozolomide-resistant glioblastoma cells [150]. Microtubule stability was increased during senescence by enhancing α-tubulin acetylation through the depletion of HDAC6 [151]. These reports imply that HDAC6 may act as a negative regulator of senescence. The natural product 2,4-di-tert-butylphenol (DTBP), an HDAC6 inhibitor, induced senescence (mitotic catastrophe) in human gastric adenocarcinoma AGS cells by increasing the expression of p21 and Rb [152]. Thus, DTBP targets HDAC6 to induce senescence. HDAC6 was increased by NF-κB and promoted hepatocellular carcinoma cell proliferation by inducing the degradation of p53 in HCCs [153] and promoted the invasion of breast cancer cells by decreasing E-cadherin while increasing STAT3 levels [154]. These reports suggest that HDAC6 promotes cancer cell proliferation by inhibiting senescence. It is probable that HDAC6 helps cancer cells escape senescence surveillance to display aggressive phenotypes. Thus, the inhibition of HDAC6 could enhance the effects of anticancer drugs by inducing senescence.
Ras promotes cisplatin resistance through HDAC4, which activates transcription factor CREB3 regulatory factor (CREBZF) to increase ATG3 expression [110]. This suggests the positive regulatory role of HDAC4 in autophagy and anticancer drug resistance. HDAC4 was highly expressed in CRC tissues, while p53 was poorly expressed in CRC tissues [155]. The downregulation of HDAC4 increased the expression of p53 in CRC [155]. This implies the negative regulatory role of HDAC4 in senescence. High HDAC4 levels predicted the poor overall survival (OS) of ovarian cancer patients [156]. The downregulation of HDAC4 was seen in aged or UV-irradiated skin [157]. The overexpression of HDAC4 overcame senescence by increasing the expression of DNA-damage-inducible transcript 4 (DDI4) [157]. HDAC4 was degraded during senescence via ubiquitination, and the deletion of HDAC4 in transformed mouse embryonic fibroblasts (MEF) was responsible for the reappearance of senescence by enhancing H3K27 acetylation levels [158]. These reports suggest that HDAC4 may act as a negative regulator of senescence.
An HDAC4 triple mutant mediated OIS in a p53-dependent manner in human primary fibroblasts [159]. HDAC4 induced growth arrest and DNA damage was seen from an unusual pattern of γH2AX-positive foci [159]. Thus, HDAC4 can act as a positive regulator of senescence. Since HDAC4 regulates senescence and autophagy, it is reasonable to employ HDAC4 as a target for developing anticancer drugs. Taken together, HDACs regulate senescence in a context-dependent manner in association with autophagy. Table 2 shows the roles of HDACs in senescence.

8. Conclusions and Perspectives

Since senescent cancer cells can evade immune surveillance (senescence surveillance) and acquire aggressive phenotypes, it is necessary to eliminate those senescent cancer cells. For this, a one-two punch strategy has been tried. The first step involves the induction of senescence with chemotherapy or other treatment modalities. The therapeutic induction of senescence is a potential means to treat cancer, primarily acting through the induction of a persistent growth-arrested state in tumors. Defensin-like peptide CopA3 increased SA-β-gal activity and ROS levels and DNA damage and G1 cell cycle arrest in colon cancer cells [160]. Thus, CopA3 is useful for the induction of senescence in cancer cells. CopA3 can be used with current chemotherapeutic drugs to eliminate senescent cancer cells. The peptides that can increase the expression of p53 or other inducers of senescence may enhance the sensitivity of cancer cells to current anticancer drugs.
The inflammatory cytokines released by senescent cells can upregulate PD-L1 in non-senescent cells via the JAK-STAT pathway [161]. This suggests that the SASP can confer an immune checkpoint response in non-senescent cells. Chemotherapeutic drugs that can increase the expression of PD-L1 induce senescence and can sensitize cancer cells to anticancer drugs. Doxorubicin-induced senescence promoted the recruitment of PD-L1-expressing T cells [162]. Thus, the induction of senescence by chemotherapy may enhance the immunotherapeutic efficacy of anti-PD-L1 antibodies. Therefore, senescence can serve as a target for developing anticancer drugs employing anti-PD-L1 antibodies. Chemotherapy induces cellular senescence in lung cancer cells and the evasion of senescence surveillance occurs via the activation of Cdc2/Cdk1 [163]. Thus, senescence-inducing chemotherapeutic drugs can be used with inhibitors of Cdc2/Cdk1.
The second step consists of follow-up treatment with senolytic drugs. Senolytic drugs selectively eliminate senescent cells to attenuate senescence-mediated pathologies. The report on the one-two punch approach to selectively eliminate chemotherapy-induced senescent lymphoma cells in mice was achieved by using a metabolic senolytic to block glucose utilization or autophagy [164]. The elimination of senescent lymphoma occurs through caspase-12- and caspase-3-mediated endoplasmic-reticulum-related apoptosis [164]. Nano-drugs, such as lapatinib- poly(amidoamine) and fulvestrant- poly(amidoamine), promoted the elimination of doxorubicin-induced senescent breast cancer cells by apoptosis-mediated senolysis [84]. Thus, the induction of senescence can sensitize cancer cells to senolytic drugs.
The main task of senolytic drugs is the elimination of senescence cells by targeting anti-apoptotic pathways. Panobinostat, a Food and Drug Administration (FDA)-approved histone deacetylase inhibitor, eliminated TIS cancer cells (NSCLCs and head and neck squamous cell carcinoma) by decreasing the expression of BCL-XL [165]. Panobinostat has shown an antitumor effect by inhibiting Wnt/β-catenin signaling in breast cancer [166]. Since TIS involves the increased expression of BCL-2, targeting BCL-2 may exert anticancer effects. BCL2-targeting drugs, including ABT-737 and ABT-263 (navitoclax), natural substances, such as artesunate, fisetin, and curcumin, showed senolytic effects in glioblastoma cells and non-transformed cells [83,167]. ABT-263 eliminated senescent cells and enhanced the efficacy of olaparib, a PARP inhibitor, in both breast and ovarian cancer cell lines [168]. Olaparib increased the expression of Bcl-XL, inflammatory cytokines, check point kinase 2 (Chk2), and p21 [168]. Thus, PARPi and senolytic drugs can induce efficient clinical outcomes. Oridonin, an active ingredient of Chinese herbal medicine, targeted glutathione S-transferases to activate ROS-p38 signaling and eliminate senescent cell via apoptosis [169]. Doxorubicin induced senescence in lung cancer cells by increasing cytoplasmic p21 and the senolytic effect of ABT-263 was enhanced in p21 knockout lung cancer cells [170].
Senolytic drugs, such as dasatinib, quercetin, and fisetin, eliminated doxorubicin-induced senescent cells by inducing apoptosis [30]. Doxorubicin increased the expression of the SASP to induce senescence in endothelial cells [30]. Dasatinib and quercetin attenuated cisplatin-induced ovarian injury by targeting senescent cells and reducing DNA damage in primary granulosa cells [171]. Fisetin induced G2/M arrest and apoptosis in NSCLCs by inhibiting EGFR/ERK1/2/STAT3 signaling [172]. Thus, TIS can sensitize cancer cells to senolytic drugs. The identification of the targets of these senolytic drugs may help us to better understand the mechanisms of senescence. Although senolytic drugs have shown some promising anticancer effects, current drugs have some limitations. Since senolytic drugs target aging and aging-related diseases, they might cause undesirable side effects. Senolytic drugs cause tissue atrophy through the massive removal of senescent cells. The complete removal of senescent cells wipes out the beneficial effects of senescent cells, including tissue regeneration, metabolic reprograming, and wound healing. It will be necessary to target another aspect of senescence and increase the specificity of senolytic drugs.
OIS causes G1 cell cycle arrest and limits the generation of cancer stem cells [173]. Since oncogenes can induce senescence, targeting oncogenes can be employed for overcoming OIS and anticancer drug resistance. Wogonin, a natural flavonoid compound, induced cellular senescence in T-cell malignancies and activated the p53-mediated DNA damage response [26]. The upregulated expression of BCL-2 was seen in senescent malignant T cells via the anti-apoptotic properties of senescent cells [28]. ABT-263 induced apoptotic cell death in wogonin-induced senescent cells [28]. Thus, the induction of senescence by wogonin increased the sensitivity of malignant T cells with a low expression of BCL-2 to ABT-263. Although oncogenes can induce senescence, they can also promote pathways that can inhibit senescence. Thus, it is necessary to identify oncogene-activated pathways that can promote or inhibit senescence.
Selectively inhibiting the SASP in normal cells comprising the TME by senostatics may enhance the therapeutic efficacy of current therapies by eliminating pro-tumorigenic factors. Silybum marianum flower extract (SMFE) suppressed the expression of SASP factors such as IL-6 and MMP-1 in senescent human diploid fibroblasts (HDFs) [174]. The CM of HDFs treated with SMFE decreased the expression of p16 and SASP in keratinocytes [174]. Rapamycin, an inducer of autophagy, inhibits senescence and SASP by decreasing SA-β-Gal activity and p16 expression in human coronary artery endothelial cells [175]. Senostatics target the SASP regulatory network (NF-κB, p38 MAPK, mTOR, and others) or a specific component of the SASP. Components of the SASP regulatory network have other functions. Since the SASP shares many overlapping factors, it would be necessary to identify the SASP in a precise way. For this, a large-scale RNA interference screening may identify targets for senostatic drugs. An analysis of senescent human fibroblast cells revealed that individual senescent cells showed heterogeneity in their gene expression signatures [176]. This heterogeneity led to a weak correlation among genes encoding the SASP [176]. The composition of the SASP shows variation depending on the stimuli, the cell type, and the stage of senescence [176]. Therefore, it will be necessary to categorize SASP factors according to their functions in a context-dependent manner.
Since HDACs play a role in senescence induction, it is necessary to examine whether HDAC inhibitors or activators can synergistically enhance the anticancer effects of senolytic drugs. The structural identification of these HDACs may help to identify novel drugs that enhance the anticancer effects of senolytic drugs. miRNAs have also been shown to regulate senescence. Therefore, miR mimics or miR inhibitors can be used in combination with senolytic drugs for the elimination of senescence-induced cancer cells.
The elimination of SASP-producing senescent cells or modification of the SASP could be the main areas of cancer therapy. However, it is necessary to further identify the factors responsible for the induction of senescence. This will make it possible to screen senescence-targeting anticancer drugs. Since the SASP is dependent on cell type, tissue, and the type of stress, it is necessary to define the SASP. Senescence-targeting drugs can synergistically enhance the anticancer effects of current cancer therapeutics. Overall, research on senotherapies is an extremely important area of cancer research that can reduce the adverse effects of senescence and improve current cancer therapies.

Author Contributions

D.J. conceived the idea and wrote the manuscript. H.J. and K.S. critically reviewed the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by National Research Foundation Grants (2020R1A2C1006996, 2022R1F1A1060031, and 2017M3A9G7072417) and a grant from the BK21 Four Program.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Huang, M.L.; Luo, W.L. Engrailed homeobox 1 transcriptional regulation of COL22A1 inhibits nasopharyngeal carcinoma cell senescence through the G1/S phase arrest. J. Cell. Mol. Med. 2022, 26, 5473–5485. [Google Scholar] [CrossRef] [PubMed]
  2. Schmitt, C.A.; Fridman, J.S.; Yang, M.; Lee, S.; Baranov, E.; Hoffman, R.M.; Lowe, S.W. A senescence program controlled by p53 and p16INK4a contributes to the outcome of cancer therapy. Cell 2002, 109, 335–346. [Google Scholar] [CrossRef] [PubMed]
  3. Millar, F.R.; Pennycuick, A.; Muir, M.; Quintanilla, A.; Hari, P.; Freyer, E.; Gautier, P.; Meynert, A.; Grimes, G.; Coll, C.S.; et al. Toll-like receptor 2 orchestrates a tumor suppressor response in non-small cell lung cancer. Cell Rep. 2022, 41, 111596. [Google Scholar] [CrossRef] [PubMed]
  4. Yu, J.B.; Lee, D.S.; Padanilam, B.J.; Kim, J. Repeated Administration of Cisplatin Transforms Kidney Fibroblasts through G2/M Arrest and Cellular Senescence. Cells 2022, 11, 3472. [Google Scholar] [CrossRef] [PubMed]
  5. Vickridge, E.; Faraco, C.C.F.; Tehrani, P.S.; Ramdzan, Z.M.; Rahimian, H.; Leduy, L.; Gingras, A.C.; Nepveu, A. The DNA repair function of BCL11A suppresses senescence and promotes continued proliferation of triple-negative breast cancer cells. NAR Cancer 2022, 4, zcac028. [Google Scholar] [CrossRef]
  6. He, S.; Sharpless, N.E. Senescence in Health and Disease. Cell 2017, 169, 1000–1011. [Google Scholar] [CrossRef]
  7. Campisi, J. Aging, cellular senescence, and cancer. Annu. Rev. Physiol. 2013, 75, 685–705. [Google Scholar] [CrossRef]
  8. Braumüller, H.; Wieder, T.; Brenner, E.; Aßmann, S.; Hahn, M.; Alkhaled, M.; Schilbach, K.; Essmann, F.; Kneilling, M.; Griessinger, C.; et al. T-helper-1-cell cytokines drive cancer into senescence. Nature 2013, 494, 361–365. [Google Scholar] [CrossRef]
  9. Bousset, L.; Gil, J. Targeting senescence as an anticancer therapy. Mol. Oncol. 2022, 16, 3855–3880. [Google Scholar] [CrossRef]
  10. Shelton, D.N.; Chang, E.; Whittier, P.S.; Choi, D.; Funk, W.D. Microarray analysis of replicative senescence. Curr. Biol. 1999, 9, 939–945. [Google Scholar] [CrossRef] [Green Version]
  11. Baker, D.J.; Childs, B.G.; Durik, M.; Wijers, M.E.; Sieben, C.J.; Zhong, J.; Saltness, R.A.; Jeganathan, K.B.; Verzosa, G.C.; Pezeshki, A.; et al. Naturally occurring p16(Ink4a)-positive cells shorten healthy lifespan. Nature 2016, 530, 184–189. [Google Scholar] [CrossRef] [PubMed]
  12. Lim, J.H.; Kim, H.Y.; Kang, H.G.; Jeong, H.J.; Kim, H.M. RANKL down-regulates the mast cell proliferation through inducing senescence. Cytokine 2022, 159, 156018. [Google Scholar] [CrossRef]
  13. Liu, X.; Shi, Q.; Choudhry, N.; Zhang, T.; Liu, H.; Zhang, S.; Zhang, J.; Yang, D. The Effect of Circumscribed Exposure to the Pan-Aurora Kinase Inhibitor VX-680 on Proliferating Euploid Cells. Int. J. Mol. Sci. 2022, 23, 12104. [Google Scholar] [CrossRef] [PubMed]
  14. Jia, M.; Su, B.; Mo, L.; Qiu, W.; Ying, J.; Lin, P.; Yang, B.; Li, D.; Wang, D.; Xu, L.; et al. Circadian clock protein CRY1 prevents paclitaxel induced senescence of bladder cancer cells by promoting p53 degradation. Oncol. Rep. 2021, 45, 1033–1043. [Google Scholar] [CrossRef]
  15. Dimri, G.P.; Lee, X.; Basile, G.; Acosta, M.; Scott, G.; Roskelley, C.; Medrano, E.E.; 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. USA 1995, 92, 9363–9367. [Google Scholar] [CrossRef] [PubMed]
  16. Espitia-Corredor, J.A.; Shamoon, L.; Olivares-Silva, F.; Rimassa-Tare, C.; Munoz-Rodriguez, C.; Espinoza-Perez, C.; Sanchez-Ferrer, C.F.; Peiro, C.; Diaz-Araya, G. Resolvin E1 attenuates doxorubicin-induced cardiac fibroblast senescence: A key role for IL-1beta. Biochim. Biophys. Acta. Mol. Basis Dis. 2022, 1868, 166525. [Google Scholar] [CrossRef]
  17. Martini, H.; Passos, J.F. Cellular senescence: All roads lead to mitochondria. FEBS J. 2022. [Google Scholar] [CrossRef]
  18. Tian, Y.T.; Ma, L.P.; Ding, C.Y.; Liu, M.M.; Wang, S.N.; Tian, M.; Gao, L.; Liu, Q.J. Autophagy regulates X-ray radiation-induced premature senescence through STAT3-Beclin1-p62 pathway in lung adenocarcinoma cells. Int. J. Radiat. Biol. 2022, 98, 1432–1441. [Google Scholar] [CrossRef]
  19. Ewald, J.A.; Desotelle, J.A.; Wilding, G.; Jarrard, D.F. Therapy-induced senescence in cancer. J. Natl. Cancer Inst. 2010, 102, 1536–1546. [Google Scholar] [CrossRef]
  20. Shao, L.; Feng, W.; Li, H.; Gardner, D.; Luo, Y.; Wang, Y.; Liu, L.; Meng, A.; Sharpless, N.E.; Zhou, D. Total body irradiation causes long-term mouse BM injury via induction of HSC premature senescence in an Ink4a- and Arf-independent man-ner. Blood 2014, 123, 3105–3115. [Google Scholar] [CrossRef] [Green Version]
  21. 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. [Google Scholar] [CrossRef]
  22. White, R.R.; Vijg, J. Do DNA Double-Strand Breaks Drive Aging? Mol. Cell 2016, 63, 729–738. [Google Scholar] [CrossRef] [PubMed]
  23. Teng, Y.N.; Chang, H.C.; Chao, Y.Y.; Cheng, H.L.; Lien, W.C.; Wang, C.Y. Etoposide Triggers Cellular Senescence by Inducing Multiple Centrosomes and Primary Cilia in Adrenocortical Tumor Cells. Cells 2021, 10, 1466. [Google Scholar] [CrossRef] [PubMed]
  24. Vilgelm, A.E.; Pawlikowski, J.S.; Liu, Y.; Hawkins, O.E.; Davis, T.A.; Smith, J.; Weller, K.P.; Horton, L.W.; McClain, C.M.; Ayers, G.D.; et al. Mdm2 and aurora kinase a inhibitors synergize to block melanoma growth by driving apoptosis and immune clearance of tumor cells. Cancer Res. 2015, 75, 181–193. [Google Scholar] [CrossRef] [PubMed]
  25. Eleftheriadis, T.; Pissas, G.; Filippidis, G.; Efthymiadi, M.; Liakopoulos, V.; Stefanidis, I. Dapagliflozin Prevents High-Glucose-Induced Cellular Senescence in Renal Tubular Epithelial Cells. Int. J. Mol. Sci. 2022, 23, 16107. [Google Scholar] [CrossRef] [PubMed]
  26. Yin, Y.; Peng, J.; Zheng, X.; Zhou, J.; Wang, Y.; Dai, Y.; Yin, G.; Tang, Y. Extrinsic apoptosis and senescence involved in growth kinetics of seminoma to cisplatin. Clin. Exp. Pharmacol. Physiol. 2023, 50, 140–148. [Google Scholar] [CrossRef]
  27. Cheng, X.; Yao, X.; Xu, S.; Pan, J.; Yu, H.; Bao, J.; Guan, H.; Lu, R.; Zhang, L. Punicalagin induces senescent growth arrest in human papillary thyroid carcinoma BCPAP cells via NF-kappaB signaling pathway. Biomed. Pharmacother. 2018, 103, 490–498. [Google Scholar] [CrossRef]
  28. Qing, Y.; Li, H.; Zhao, Y.; Hu, P.; Wang, X.; Yu, X.; Zhu, M.; Wang, H.; Wang, Z.; Guo, Q.; et al. One-Two Punch Therapy for the Treatment of T-Cell Malignancies Involving p53-Dependent Cellular Senescence. Oxid. Med. Cell. Longev. 2021, 2021, 5529518. [Google Scholar] [CrossRef]
  29. Ma, L.; Shen, H.; Fang, C.; Chen, T.; Wang, J. Camellia Seed Cake Extract Supports Hair Growth by Abrogating the Effect of Dihydrotestosterone in Cultured Human Dermal Papilla Cells. Molecules 2022, 27, 6443. [Google Scholar] [CrossRef]
  30. Abdelgawad, I.Y.; Agostinucci, K.; Ismail, S.G.; Grant, M.K.O.; Zordoky, B.N. EA. hy926 Cells and HUVECs Share Similar Se-nescence Phenotypes but Respond Differently to the Senolytic Drug ABT-263. Cells 2022, 11, 1992. [Google Scholar] [CrossRef]
  31. Sheng, J.; Kohno, S.; Okada, N.; Okahashi, N.; Teranishi, K.; Matsuda, F.; Shimizu, H.; Linn, P.; Nagatani, N.; Yamamura, M.; et al. Treatment of Retinoblastoma 1-Intact Hepatocellular Carcinoma with Cyclin-Dependent Kinase 4/6 Inhibitor Combination Therapy. Hepatology 2021, 74, 1971–1993. [Google Scholar] [CrossRef] [PubMed]
  32. Liu, Y.; Hawkins, O.E.; Su, Y.; Vilgelm, A.E.; Sobolik, T.; Thu, Y.M.; Kantrow, S.; Splittgerber, R.C.; Short, S.; Amiri, K.I.; et al. Targeting aurora kinases limits tumour growth through DNA damage-mediated senescence and blockade of NF-κB impairs this drug-induced senescence. EMBO Mol. Med. 2013, 5, 149–166. [Google Scholar] [CrossRef] [PubMed]
  33. Warnon, C.; Bouhjar, K.; Ninane, N.; Verhoyen, M.; Fattaccioli, A.; Fransolet, M.; Lambert de Rouvroit, C.; Poumay, Y.; Piel, G.; Mottet, D.; et al. HDAC2 and 7 down-regulation induces senescence in dermal fibroblasts. Aging 2021, 13, 17978–18005. [Google Scholar] [CrossRef] [PubMed]
  34. Lewinska, A.; Adamczyk-Grochala, J.; Kwasniewicz, E.; Wnuk, M. Downregulation of methyltransferase Dnmt2 results in condition-dependent telomere shortening and senescence or apoptosis in mouse fibroblasts. J. Cell. Physiol. 2017, 232, 3714–3726. [Google Scholar] [CrossRef] [PubMed]
  35. Chapman, J.; Fielder, E.; Passos, J.F. Mitochondrial dysfunction and cell senescence: Deciphering a complex relationship. FEBS Lett. 2019, 593, 1566–1579. [Google Scholar] [CrossRef] [PubMed]
  36. Wiley, C.D.; Velarde, M.C.; Lecot, P.; Liu, S.; Sarnoski, E.A.; Freund, A.; Shirakawa, K.; Lim, H.W.; Davis, S.S.; Ramanathan, A.; et al. Mitochondrial Dysfunction Induces Senescence with a Distinct Secretory Phenotype. Cell Metab. 2016, 23, 303–314. [Google Scholar] [CrossRef] [PubMed]
  37. Correia-Melo, C.; Marques, F.D.; Anderson, R.; Hewitt, G.; Hewitt, R.; Cole, J.; Carroll, B.M.; Miwa, S.; Birch, J.; Merz, A.; et al. Mitochondria are required for pro-ageing features of the senescent phenotype. EMBO J. 2016, 35, 724–742. [Google Scholar] [CrossRef] [PubMed]
  38. Lafargue, A.; Degorre, C.; Corre, I.; Alves-Guerra, M.C.; Gaugler, M.H.; Vallette, F.; Pecqueur, C.; Paris, F. Ionizing radiation induces long-term senescence in endothelial cells through mitochondrial respiratory complex II dysfunction and superoxide generation. Free Radic. Biol. Med. 2017, 108, 750–759. [Google Scholar] [CrossRef]
  39. Hong, G.L.; Kim, K.H.; Kim, Y.J.; Lee, H.J.; Kim, H.T.; Jung, J.Y. Decreased mitophagy aggravates benign prostatic hyperplasia in aged mice through DRP1 and estrogen receptor alpha. Life Sci. 2022, 309, 120980. [Google Scholar] [CrossRef]
  40. Wolf, D.A. Is reliance on mitochondrial respiration a “chink in the armor” of therapy-resistant cancer? Cancer Cell 2014, 26, 788–795. [Google Scholar] [CrossRef] [Green Version]
  41. Vizioli, M.G.; Liu, T.; Miller, K.N.; Robertson, N.A.; Gilroy, K.; Lagnado, A.B.; Perez-Garcia, A.; Kiourtis, C.; Dasgupta, N.; Lei, X.; et al. Mitochondria-to-nucleus retrograde signaling drives formation of cytoplasmic chromatin and inflammation in senes-cence. Genes Dev. 2020, 34, 428–445. [Google Scholar] [CrossRef] [PubMed]
  42. Takaya, K.; Asou, T.; Kishi, K. Cathepsin F is a potential marker for senescent human skin fibroblasts and keratinocytes asso-ciated with skin aging. Geroscience 2022, 45, 427–437. [Google Scholar] [CrossRef] [PubMed]
  43. Ok, C.Y.; Park, S.; Jang, H.O.; Takata, T.; Lee, O.H.; Bae, M.K.; Bae, S.K. FK866 Protects Human Dental Pulp Cells against Oxi-dative Stress-Induced Cellular Senescence. Antioxidants 2021, 10, 271. [Google Scholar] [CrossRef] [PubMed]
  44. Wang, L.; Yin, H.; Huang, S.; Huang, S.; Huang, C.; Zhang, Z.; Liu, H. Bortezomib induces cellular senescence in A549 lung cancer cells by stimulating telomere shortening. Hum. Exp. Toxicol. 2022, 41, 9603271221124094. [Google Scholar] [CrossRef]
  45. Radnaa, E.; Richardson, L.; Goldman, B.; Burks, J.K.; Baljinnyam, T.; Vora, N.; Zhang, H.J.; Bonney, E.A.; Han, A.; Menon, R. Stress signaler p38 mitogen-activated kinase activation: A cause for concern? Clin. Sci. 2022, 136, 1591–1614. [Google Scholar] [CrossRef]
  46. Palazzo, A.; Hernandez-Vargas, H.; Goehrig, D.; Medard, J.J.; Vindrieux, D.; Flaman, J.M.; Bernard, D. Transformed cells after senescence give rise to more severe tumor phenotypes than transformed non-senescent cells. Cancer Lett. 2022, 546, 215850. [Google Scholar] [CrossRef]
  47. Sahu, B.; Pihlajamaa, P.; Zhang, K.; Palin, K.; Ahonen, S.; Cervera, A.; Ristimaki, A.; Aaltonen, L.A.; Hautaniemi, S.; Taipale, J. Human cell transformation by combined lineage conversion and oncogene expression. Oncogene 2021, 40, 5533–5547. [Google Scholar] [CrossRef]
  48. Lee, J.V.; Housley, F.; Yau, C.; Nakagawa, R.; Winkler, J.; Anttila, J.M.; Munne, P.M.; Savelius, M.; Houlahan, K.E.; Van de Mark, D.; et al. Combinatorial immunotherapies overcome MYC-driven immune evasion in triple negative breast cancer. Nat. Commun. 2022, 13, 3671. [Google Scholar] [CrossRef]
  49. Xi, C.; Sun, J.; Xu, X.; Wu, Y.; Kou, X.; Zhao, Y.; Shen, J.; Dong, Y.; Chen, K.; Su, Z.; et al. Mettl14-driven senescence-associated secretory phenotype facilitates somatic cell reprogramming. Stem Cell Rep. 2022, 17, 1799–1809. [Google Scholar] [CrossRef]
  50. Park, C.; Lee, H.; Jin, S.; Park, J.H.; Han, M.H.; Jeong, J.W.; Kwon, H.J.; Kim, B.W.; Park, S.H.; Hong, S.H.; et al. The preventive effect of loganin on oxidative stress-induced cellular damage in human keratinocyte HaCaT cells. Biosci. Trends 2022, 16, 291–300. [Google Scholar] [CrossRef]
  51. Hari, P.; Millar, F.R.; Tarrats, N.; Birch, J.; Quintanilla, A.; Rink, C.J.; Fernandez-Duran, I.; Muir, M.; Finch, A.J.; Brunton, V.G.; et al. The innate immune sensor Toll-like receptor 2 controls the senescence-associated secretory phenotype. Sci. Adv. 2019, 5, eaaw0254. [Google Scholar] [CrossRef]
  52. Lee, A.J.; Fraser, E.; Flowers, B.; Kim, J.; Wong, K.; Cataisson, C.; Liu, H.; Yang, H.; Lee, M.P.; Yuspa, S.H.; et al. RAS induced senescence of skin keratinocytes is mediated through Rho-associated protein kinase (ROCK). Mol. Carcinog. 2021, 60, 799–812. [Google Scholar] [CrossRef]
  53. Serrano, M.; Lin, A.W.; McCurrach, M.E.; Beach, D.; Lowe, S.W. Oncogenic ras provokes premature cell senescence associated with accumulation of p53 and p16INK4a. Cell 1997, 88, 593–602. [Google Scholar] [CrossRef] [PubMed]
  54. Fernandez-Duran, I.; Quintanilla, A.; Tarrats, N.; Birch, J.; Hari, P.; Millar, F.R.; Lagnado, A.B.; Smer-Barreto, V.; Muir, M.; Brunton, V.G.; et al. Cytoplasmic innate immune sensing by the caspase-4 non-canonical inflammasome promotes cellular se-nescence. Cell Death Differ. 2022, 29, 1267–1282. [Google Scholar] [CrossRef] [PubMed]
  55. Kim, Y.Y.; Um, J.H.; Shin, D.J.; Jeong, D.J.; Hong, Y.B.; Yun, J. p53-mediated regulation of mitochondrial dynamics plays a pivotal role in the senescence of various normal cells as well as cancer cells. FASEB J. 2021, 35, e21319. [Google Scholar] [CrossRef] [PubMed]
  56. Siebenaler, R.F.; Chugh, S.; Waninger, J.J.; Dommeti, V.L.; Kenum, C.; Mody, M.; Gautam, A.; Patel, N.; Chu, A.; Bawa, P.; et al. Argonaute 2 modulates EGFR-RAS signaling to promote mutant HRAS and NRAS-driven malignancies. PNAS Nexus 2022, 1, pgac084. [Google Scholar] [CrossRef]
  57. Kyjacova, L.; Saup, R.; Ronsch, K.; Wallbaum, S.; Dukowic-Schulze, S.; Foss, A.; Scherer, S.D.; Rothley, M.; Neeb, A.; Grau, N.; et al. IER2-induced senescence drives melanoma invasion through osteopontin. Oncogene 2021, 40, 6494–6512. [Google Scholar] [CrossRef]
  58. Grbcic, P.; Tomljanovic, I.; Klobucar, M.; Kraljevic Pavelic, S.; Lucin, K.; Sedic, M. Dual sphingosine kinase inhibitor SKI-II enhances sensitivity to 5-fluorouracil in hepatocellular carcinoma cells via suppression of osteopontin and FAK/IGF-1R signal-ling. Biochem. Biophys. Res. Commun. 2017, 487, 782–788. [Google Scholar] [CrossRef]
  59. Borghesan, M.; Fafian-Labora, J.; Eleftheriadou, O.; Carpintero-Fernandez, P.; Paez-Ribes, M.; Vizcay-Barrena, G.; Swisa, A.; Kolodkin-Gal, D.; Ximenez-Embun, P.; Lowe, R.; et al. Small Extracellular Vesicles Are Key Regulators of Non-cell Autonomous Intercellular Communication in Senescence via the Interferon Protein IFITM3. Cell Rep. 2019, 27, 3956–3971.e3956. [Google Scholar] [CrossRef]
  60. Basisty, N.; Kale, A.; Jeon, O.H.; Kuehnemann, C.; Payne, T.; Rao, C.; Holtz, A.; Shah, S.; Sharma, V.; Ferrucci, L.; et al. A pro-teomic atlas of senescence-associated secretomes for aging biomarker development. PLoS Biol. 2020, 18, e3000599. [Google Scholar] [CrossRef] [Green Version]
  61. Iske, J.; Seyda, M.; Heinbokel, T.; Maenosono, R.; Minami, K.; Nian, Y.; Quante, M.; Falk, C.S.; Azuma, H.; Martin, F.; et al. Senolytics prevent mt-DNA-induced inflammation and promote the survival of aged organs following transplantation. Nat. Commun. 2020, 11, 4289. [Google Scholar] [CrossRef]
  62. Colpani, O.; Spinetti, G. MicroRNAs orchestrating senescence of endothelial and vascular smooth muscle cells. Vasc. Biol. 2019, 1, H75–H81. [Google Scholar] [CrossRef]
  63. Xiao, Y.; Liang, J.; Witwer, K.W.; Zhang, Y.; Wang, Q.; Yin, H. Extracellular vesicle-associated microRNA-30b-5p activates macrophages through the SIRT1/NF-kappaB pathway in cell senescence. Front. Immunol. 2022, 13, 955175. [Google Scholar] [CrossRef]
  64. Gluchowska, A.; Cysewski, D.; Baj-Krzyworzeka, M.; Szatanek, R.; Weglarczyk, K.; Podszywalow-Bartnicka, P.; Sunderland, P.; Kozlowska, E.; Sliwinska, M.A.; Dabrowski, M.; et al. Unbiased proteomic analysis of extracellular vesicles secreted by senescent human vascular smooth muscle cells reveals their ability to modulate immune cell functions. Geroscience 2022, 44, 2863–2884. [Google Scholar] [CrossRef]
  65. Haraoka, Y.; Akieda, Y.; Nagai, Y.; Mogi, C.; Ishitani, T. Zebrafish imaging reveals TP53 mutation switching onco-gene-induced senescence from suppressor to driver in primary tumorigenesis. Nat. Commun. 2022, 13, 1417. [Google Scholar] [CrossRef]
  66. Morales-Valencia, J.; Lau, L.; Marti-Nin, T.; Ozerdem, U.; David, G. Therapy-induced senescence promotes breast cancer cells plasticity by inducing Lipocalin-2 expression. Oncogene 2022, 41, 4361–4370. [Google Scholar] [CrossRef]
  67. Yu, L.; Cao, C.; Li, X.; Zhang, M.; Gu, Q.; Gao, H.; Balic, J.J.; Xu, D.; Zhang, L.; Ying, L.; et al. Complete loss of miR-200 family induces EMT associated cellular senescence in gastric cancer. Oncogene 2022, 41, 26–36. [Google Scholar] [CrossRef]
  68. Chen, B.; Liang, Y.; Chen, L.; Wei, Y.; Li, Y.; Zhao, W.; Wu, J. Overexpression of Klotho Inhibits HELF Fibroblasts SASP-related Protumoral Effects on Non-small Cell Lung Cancer Cells. J. Cancer 2018, 9, 1248–1258. [Google Scholar] [CrossRef]
  69. Faheem, M.M.; Seligson, N.D.; Ahmad, S.M.; Rasool, R.U.; Gandhi, S.G.; Bhagat, M.; Goswami, A. Convergence of thera-py-induced senescence (TIS) and EMT in multistep carcinogenesis: Current opinions and emerging perspectives. Cell Death Discov. 2020, 6, 51. [Google Scholar] [CrossRef]
  70. Chaturvedi, P.; George, V.; Shrestha, N.; Wang, M.; Dee, M.J.; Zhu, X.; Liu, B.; Egan, J.; D’Eramo, F.; Spanoudis, C.; et al. Im-munotherapeutic HCW9218 augments anti-tumor activity of chemotherapy via NK cell-mediated reduction of therapy-induced senescent cells. Mol. Ther. 2022, 30, 1171–1187. [Google Scholar] [CrossRef]
  71. Karabicici, M.; Alptekin, S.; Firtina Karagonlar, Z.; Erdal, E. Doxorubicin-induced senescence promotes stemness and tumor-igenicity in EpCAM-/CD133- nonstem cell population in hepatocellular carcinoma cell line, HuH-7. Mol. Oncol. 2021, 15, 2185–2202. [Google Scholar] [CrossRef]
  72. Xu, Q.; Long, Q.; Zhu, D.; Fu, D.; Zhang, B.; Han, L.; Qian, M.; Guo, J.; Xu, J.; Cao, L.; et al. Targeting amphiregulin (AREG) de-rived from senescent stromal cells diminishes cancer resistance and averts programmed cell death 1 ligand (PD-L1)-mediated immunosuppression. Aging Cell 2019, 18, e13027. [Google Scholar] [CrossRef]
  73. Canino, C.; Mori, F.; Cambria, A.; Diamantini, A.; Germoni, S.; Alessandrini, G.; Borsellino, G.; Galati, R.; Battistini, L.; Blan-dino, R.; et al. SASP mediates chemoresistance and tumor-initiating-activity of mesothelioma cells. Oncogene 2012, 31, 3148–3163. [Google Scholar] [CrossRef]
  74. Chakrabarty, A.; Chakraborty, S.; Bhattacharya, R.; Chowdhury, G. Senescence-Induced Chemoresistance in Triple Negative Breast Cancer and Evolution-Based Treatment Strategies. Front. Oncol. 2021, 11, 674354. [Google Scholar] [CrossRef]
  75. Parikh, P.; Wicher, S.; Khandalavala, K.; Pabelick, C.M.; Britt, R.D., Jr.; Prakash, Y.S. Cellular senescence in the lung across the age spectrum. Am. J. Physiol. Lung Cell. Mol. Physiol. 2019, 316, L826–L842. [Google Scholar] [CrossRef]
  76. Coppe, J.P.; Kauser, K.; Campisi, J.; Beausejour, C.M. Secretion of vascular endothelial growth factor by primary human fi-broblasts at senescence. J. Biol. Chem. 2006, 281, 29568–29574. [Google Scholar] [CrossRef]
  77. Pauty, J.; Nakano, S.; Usuba, R.; Nakajima, T.; Johmura, Y.; Omori, S.; Sakamoto, N.; Kikuchi, A.; Nakanishi, M.; Matsunaga, Y.T. A 3D tissue model-on-a-chip for studying the effects of human senescent fibroblasts on blood vessels. Biomater. Sci. 2021, 9, 199–211. [Google Scholar] [CrossRef]
  78. Weng, P.W.; Pikatan, N.W.; Setiawan, S.A.; Yadav, V.K.; Fong, I.H.; Hsu, C.H.; Yeh, C.T.; Lee, W.H. Role of GDF15/MAPK14 Axis in Chondrocyte Senescence as a Novel Senomorphic Agent in Osteoarthritis. Int. J. Mol. Sci. 2022, 23, 7043. [Google Scholar] [CrossRef]
  79. Wang, J.; Tao, Q.; Pan, Y.; Wanyan, Z.; Zhu, F.; Xu, X.; Wang, H.; Yi, L.; Zhou, M.; Zhai, Z. Stress-induced premature senescence activated by the SENEX gene mediates apoptosis resistance of diffuse large B-cell lymphoma via promoting immunosuppressive cells and cytokines. Immun. Inflamm. Dis. 2020, 8, 672–683. [Google Scholar] [CrossRef]
  80. Ruhland, M.K.; Loza, A.J.; Capietto, A.H.; Luo, X.; Knolhoff, B.L.; Flanagan, K.C.; Belt, B.A.; Alspach, E.; Leahy, K.; Luo, J.; et al. Stromal senescence establishes an immunosuppressive microenvironment that drives tumorigenesis. Nat. Commun. 2016, 7, 11762. [Google Scholar] [CrossRef] [Green Version]
  81. Goncalves, S.; Yin, K.; Ito, Y.; Chan, A.; Olan, I.; Gough, S.; Cassidy, L.; Serrao, E.; Smith, S.; Young, A.; et al. COX2 regulates senescence secretome composition and senescence surveillance through PGE(2). Cell Rep. 2021, 34, 108860. [Google Scholar] [CrossRef]
  82. Ruscetti, M.; Morris, J.P.T.; Mezzadra, R.; Russell, J.; Leibold, J.; Romesser, P.B.; Simon, J.; Kulick, A.; Ho, Y.J.; Fennell, M.; et al. Senescence-Induced Vascular Remodeling Creates Therapeutic Vulnerabilities in Pancreas Cancer. Cell 2020, 181, 424–441.e421. [Google Scholar] [CrossRef]
  83. Rysanek, D.; Vasicova, P.; Kolla, J.N.; Sedlak, D.; Andera, L.; Bartek, J.; Hodny, Z. Synergism of BCL-2 family inhibitors facili-tates selective elimination of senescent cells. Aging 2022, 14, 6381–6414. [Google Scholar] [CrossRef]
  84. Lewinska, A.; Wrobel, K.; Bloniarz, D.; Adamczyk-Grochala, J.; Wolowiec, S.; Wnuk, M. Lapatinib- and fulvestrant-PAMAM dendrimer conjugates promote apoptosis in chemotherapy-induced senescent breast cancer cells with different receptor status. Biomater. Adv. 2022, 140, 213047. [Google Scholar] [CrossRef]
  85. Kikuchi, H.; Maishi, N.; Annan, D.A.; Alam, M.T.; Dawood, R.I.H.; Sato, M.; Morimoto, M.; Takeda, R.; Ishizuka, K.; Matsumo-to, R.; et al. Chemotherapy-Induced IL8 Upregulates MDR1/ABCB1 in Tumor Blood Vessels and Results in Unfavorable Out-come. Cancer Res. 2020, 80, 2996–3008. [Google Scholar] [CrossRef]
  86. Peng, S.; Chen, Y.; Li, T.; Mao, J.; Yang, P.; Zou, B.; Luo, L.; Zhang, W.; Wang, W.; Xie, R.; et al. Hsa-microRNA-370-3p targeting Snail and Twist1 suppresses IL-8/STAT3-driven hepatocellular carcinoma metastasis. Cancer Sci. 2022, 113, 4120–4134. [Google Scholar] [CrossRef]
  87. Kim, Y.; Kim, H.; Park, D.; Lee, H.; Lee, Y.S.; Choe, J.; Kim, Y.M.; Jeon, D.; Jeoung, D. The pentapeptide Gly-Thr-Gly-Lys-Thr confers sensitivity to anti-cancer drugs by inhibition of CAGE binding to GSK3beta and decreasing the expression of cy-clinD1. Oncotarget 2017, 8, 13632–13651. [Google Scholar] [CrossRef]
  88. Muralikrishnan, V.; Fang, F.; Given, T.C.; Podicheti, R.; Chtcherbinine, M.; Metcalfe, T.X.; Sriramkumar, S.; O’Hagan, H.M.; Hurley, T.D.; Nephew, K.P. A Novel ALDH1A1 Inhibitor Blocks Platinum-Induced Senescence and Stemness in Ovarian Can-cer. Cancers 2022, 14, 3437. [Google Scholar] [CrossRef]
  89. Gerardo-Ramirez, M.; Keggenhoff, F.L.; Giam, V.; Becker, D.; Groth, M.; Hartmann, N.; Straub, B.K.; Morrison, H.; Galle, P.R.; Marquardt, J.U.; et al. CD44 Contributes to the Regulation of MDR1 Protein and Doxorubicin Chemoresistance in Osteosar-coma. Int. J. Mol. Sci. 2022, 23, 8616. [Google Scholar] [CrossRef]
  90. Guo, L.; Ke, H.; Zhang, H.; Zou, L.; Yang, Q.; Lu, X.; Zhao, L.; Jiao, B. TDP43 promotes stemness of breast cancer stem cells through CD44 variant splicing isoforms. Cell Death Dis. 2022, 13, 428. [Google Scholar] [CrossRef]
  91. Miyoshi, S.; Tsugawa, H.; Matsuzaki, J.; Hirata, K.; Mori, H.; Saya, H.; Kanai, T.; Suzuki, H. Inhibiting xCT Improves 5-Fluorouracil Resistance of Gastric Cancer Induced by CD44 Variant 9 Expression. Anticancer Res. 2018, 38, 6163–6170. [Google Scholar] [CrossRef]
  92. Filppu, P.; Tanjore Ramanathan, J.; Granberg, K.J.; Gucciardo, E.; Haapasalo, H.; Lehti, K.; Nykter, M.; Le Joncour, V.; Laakko-nen, P. CD109-GP130 interaction drives glioblastoma stem cell plasticity and chemoresistance through STAT3 activity. JCI In-sight 2021, 6, e141486. [Google Scholar] [CrossRef]
  93. Wang, W.; Tang, Y.A.; Xiao, Q.; Lee, W.C.; Cheng, B.; Niu, Z.; Oguz, G.; Feng, M.; Lee, P.L.; Li, B.; et al. Stromal induction of BRD4 phosphorylation Results in Chromatin Remodeling and BET inhibitor Resistance in Colorectal Cancer. Nat. Commun. 2021, 12, 4441. [Google Scholar] [CrossRef]
  94. Liu, S.; Zhang, C.; Wang, B.; Zhang, H.; Qin, G.; Li, C.; Cao, L.; Gao, Q.; Ping, Y.; Zhang, K.; et al. Regulatory T cells promote glioma cell stemness through TGF-beta-NF-kappaB-IL6-STAT3 signaling. Cancer Immunol. Immunother. 2021, 70, 2601–2616. [Google Scholar] [CrossRef]
  95. Gerber, P.A.; Buhren, B.A.; Schrumpf, H.; Hevezi, P.; Bolke, E.; Sohn, D.; Janicke, R.U.; Belum, V.R.; Robert, C.; Lacouture, M.E.; et al. Mechanisms of skin aging induced by EGFR inhibitors. Support. Care Cancer 2016, 24, 4241–4248. [Google Scholar] [CrossRef]
  96. Ji, X.L.; He, M. Sodium cantharidate targets STAT3 and abrogates EGFR inhibitor resistance in osteosarcoma. Aging 2019, 11, 5848–5863. [Google Scholar] [CrossRef]
  97. Braumuller, H.; Mauerer, B.; Berlin, C.; Plundrich, D.; Marbach, P.; Cauchy, P.; Laessle, C.; Biesel, E.; Holzner, P.A.; Kesselring, R. Senescent Tumor Cells in the Peritoneal Carcinomatosis Drive Immunosenescence in the Tumor Microenvironment. Front. Immunol. 2022, 13, 908449. [Google Scholar] [CrossRef]
  98. Choi, Y.W.; Kim, Y.H.; Oh, S.Y.; Suh, K.W.; Kim, Y.S.; Lee, G.Y.; Yoon, J.E.; Park, S.S.; Lee, Y.K.; Park, Y.J.; et al. Senescent Tu-mor Cells Build a Cytokine Shield in Colorectal Cancer. Adv. Sci. 2021, 8, 2002497. [Google Scholar] [CrossRef]
  99. Song, S.; Zhang, Y.; Duan, X.; Liu, C.; Du, Y.; Wang, X.; Luo, Y.; Cui, Y. HIF-1alpha/IL-8 axis in hypoxic macrophages promotes esophageal cancer progression by enhancing PD-L1 expression. Cancer Gene Ther. 2022. [Google Scholar] [CrossRef]
  100. Wang, T.W.; Johmura, Y.; Suzuki, N.; Omori, S.; Migita, T.; Yamaguchi, K.; Hatakeyama, S.; Yamazaki, S.; Shimizu, E.; Imoto, S.; et al. Blocking PD-L1-PD-1 improves senescence surveillance and ageing phenotypes. Nature 2022, 611, 358–364. [Google Scholar] [CrossRef]
  101. Lee, J.J.; Kim, S.Y.; Kim, S.H.; Choi, S.; Lee, B.; Shin, J.S. STING mediates nuclear PD-L1 targeting-induced senescence in cancer cells. Cell Death Dis. 2022, 13, 791. [Google Scholar] [CrossRef]
  102. Saleh, T.; Tyutyunyk-Massey, L.; Gewirtz, D.A. Tumor Cell Escape from Therapy-Induced Senescence as a Model of Disease Recurrence after Dormancy. Cancer Res. 2019, 79, 1044–1046. [Google Scholar] [CrossRef]
  103. Liu, H.; Zhao, H.; Sun, Y. Tumor microenvironment and cellular senescence: Understanding therapeutic resistance and har-nessing strategies. Semin. Cancer Biol. 2022, 86, 769–781. [Google Scholar] [CrossRef]
  104. Cahu, J.; Bustany, S.; Sola, B. Senescence-associated secretory phenotype favors the emergence of cancer stem-like cells. Cell Death Dis. 2012, 3, e446. [Google Scholar] [CrossRef]
  105. Milanovic, M.; Fan, D.N.Y.; Belenki, D.; Dabritz, J.H.M.; Zhao, Z.; Yu, Y.; Dorr, J.R.; Dimitrova, L.; Lenze, D.; Monteiro Barbosa, I.A.; et al. Senescence-associated reprogramming promotes cancer stemness. Nature 2018, 553, 96–100. [Google Scholar] [CrossRef]
  106. Guillon, J.; Petit, C.; Moreau, M.; Toutain, B.; Henry, C.; Roche, H.; Bonichon-Lamichhane, N.; Salmon, J.P.; Lemonnier, J.; Campone, M.; et al. Regulation of senescence escape by TSP1 and CD47 following chemotherapy treatment. Cell Death Dis. 2019, 10, 199. [Google Scholar] [CrossRef]
  107. Faget, D.V.; Ren, Q.; Stewart, S.A. Unmasking senescence: Context-dependent effects of SASP in cancer. Nat. Rev. Cancer 2019, 19, 439–453. [Google Scholar] [CrossRef]
  108. Rodier, F.; Coppe, J.P.; Patil, C.K.; Hoeijmakers, W.A.; Munoz, D.P.; Raza, S.R.; Freund, A.; Campeau, E.; Davalos, A.R.; Campisi, J. Persistent DNA damage signalling triggers senescence-associated inflammatory cytokine secretion. Nat. Cell Biol. 2009, 11, 973–979. [Google Scholar] [CrossRef]
  109. Kang, T.W.; Yevsa, T.; Woller, N.; Hoenicke, L.; Wuestefeld, T.; Dauch, D.; Hohmeyer, A.; Gereke, M.; Rudalska, R.; Potapova, A.; et al. Senescence surveillance of pre-malignant hepatocytes limits liver cancer development. Nature 2011, 479, 547–551. [Google Scholar] [CrossRef]
  110. Zhang, X.; Pearsall, V.M.; Carver, C.M.; Atkinson, E.J.; Clarkson, B.D.S.; Grund, E.M.; Baez-Faria, M.; Pavelko, K.D.; Kacher-gus, J.M.; White, T.A.; et al. Rejuvenation of the aged brain immune cell landscape in mice through p16-positive senescent cell clearance. Nat. Commun. 2022, 13, 5671. [Google Scholar] [CrossRef]
  111. Cui, X.; Hao, X.; Wen, J.; Zhang, S.; Zhao, B.; Miao, J. Grp94 Inhibitor HCP1 Inhibits Human Dermal Fibroblast Senescence. Genes 2022, 13, 1651. [Google Scholar] [CrossRef] [PubMed]
  112. Kalitin, N.N.; Ektova, L.V.; Kostritsa, N.S.; Sivirinova, A.S.; Kostarev, A.V.; Smirnova, G.B.; Borisova, Y.A.; Golubeva, I.S.; Ermolaeva, E.V.; Vergun, M.A.; et al. A novel glycosylated indolocarbazole derivative LCS1269 effectively inhibits growth of human cancer cells in vitro and in vivo through driving of both apoptosis and senescence by inducing of DNA damage and modulating of AKT/mTOR/S6K and ERK pathways. Chem. Biol. Interact. 2022, 364, 110056. [Google Scholar] [CrossRef] [PubMed]
  113. Li, Q.; Chu, Y.; Li, S.; Yu, L.; Deng, H.; Liao, C.; Liao, X.; Yang, C.; Qi, M.; Cheng, J.; et al. The oncoprotein MUC1 facilitates breast cancer progression by promoting Pink1-dependent mitophagy via ATAD3A destabilization. Cell Death Dis. 2022, 13, 899. [Google Scholar] [CrossRef] [PubMed]
  114. Mizushima, N.; Levine, B.; Cuervo, A.M.; Klionsky, D.J. Autophagy fights disease through cellular self-digestion. Nature 2008, 451, 1069–1075. [Google Scholar] [CrossRef]
  115. Fan, X.; He, Y.; Wu, G.; Chen, H.; Cheng, X.; Zhan, Y.; An, C.; Chen, T.; Wang, X. Sirt3 activates autophagy to prevent DOX-induced senescence by inactivating PI3K/AKT/mTOR pathway in A549 cells. Biochim. Biophys. Acta. Mol. Cell Res. 2023, 1870, 119411. [Google Scholar] [CrossRef]
  116. Belounis, A.; Nyalendo, C.; Le Gall, R.; Imbriglio, T.V.; Mahma, M.; Teira, P.; Beaunoyer, M.; Cournoyer, S.; Haddad, E.; Vassal, G.; et al. Autophagy is associated with chemoresistance in neuroblastoma. BMC Cancer 2016, 16, 891. [Google Scholar] [CrossRef]
  117. Yin, W.; Pham, C.V.; Wang, T.; Al Shamaileh, H.; Chowdhury, R.; Patel, S.; Li, Y.; Kong, L.; Hou, Y.; Zhu, Y.; et al. Inhibition of Autophagy Promotes the Elimination of Liver Cancer Stem Cells by CD133 Aptamer-Targeted Delivery of Doxorubicin. Biomolecules 2022, 12, 1623. [Google Scholar] [CrossRef]
  118. Tan, Q.; Joshua, A.M.; Wang, M.; Bristow, R.G.; Wouters, B.G.; Allen, C.J.; Tannock, I.F. Up-regulation of autophagy is a mechanism of resistance to chemotherapy and can be inhibited by pantoprazole to increase drug sensitivity. Cancer Chemother. Pharmacol. 2017, 79, 959–969. [Google Scholar] [CrossRef]
  119. Yeon, M.; Kim, Y.; Pathak, D.; Kwon, E.; Kim, D.Y.; Jeong, M.S.; Jung, H.S.; Jeoung, D. The CAGE-MiR-181b-5p-S1PR1 Axis Regulates Anticancer Drug Resistance and Autophagy in Gastric Cancer Cells. Front. Cell Dev. Biol. 2021, 9, 666387. [Google Scholar] [CrossRef]
  120. Lyu, X.; Zeng, L.; Shi, J.; Ming, Z.; Li, W.; Liu, B.; Chen, Y.; Yuan, B.; Sun, R.; Yuan, J.; et al. Essential role for STAT3/FOXM1/ATG7 signaling-dependent autophagy in resistance to Icotinib. J. Exp. Clin. Cancer Res. 2022, 41, 200. [Google Scholar] [CrossRef]
  121. Yeon, M.; Byun, J.; Kim, H.; Kim, M.; Jung, H.S.; Jeon, D.; Kim, Y.; Jeoung, D. CAGE Binds to Beclin1, Regulates Autophagic Flux and CAGE-Derived Peptide Confers Sensitivity to Anti-cancer Drugs in Non-small Cell Lung Cancer Cells. Front. Oncol. 2018, 8, 599. [Google Scholar] [CrossRef] [PubMed]
  122. Lypova, N.; Dougherty, S.M.; Lanceta, L.; Chesney, J.; Imbert-Fernandez, Y. PFKFB3 Inhibition Impairs Erlotinib-Induced Autophagy in NSCLCs. Cells 2021, 10, 1679. [Google Scholar] [CrossRef] [PubMed]
  123. Fei, D.; Yuan, H.; Zhao, M.; Zhao, D. LncRNA FGD5-AS1 potentiates autophagy-associated doxorubicin resistance by regulating the miR-154-5p/WNT5A axis in osteosarcoma. Cell Biol. Int. 2022, 46, 1937–1946. [Google Scholar] [CrossRef] [PubMed]
  124. Jovanovic Stojanov, S.; Kostic, A.; Ljujic, M.; Lupsic, E.; Schenone, S.; Pesic, M.; Dinic, J. Autophagy Inhibition Enhances Anti-Glioblastoma Effects of Pyrazolo[3,4-d]pyrimidine Tyrosine Kinase Inhibitors. Life 2022, 12, 1503. [Google Scholar] [CrossRef]
  125. Jakhar, R.; Luijten, M.N.H.; Wong, A.X.F.; Cheng, B.; Guo, K.; Neo, S.P.; Au, B.; Kulkarni, M.; Lim, K.J.; Maimaiti, J.; et al. Autophagy Governs Protumorigenic Effects of Mitotic Slippage-induced Senescence. Mol. Cancer Res. 2018, 16, 1625–1640. [Google Scholar] [CrossRef]
  126. Young, A.R.; Narita, M.; Ferreira, M.; Kirschner, K.; Sadaie, M.; Darot, J.F.; Tavare, S.; Arakawa, S.; Shimizu, S.; Watt, F.M.; et al. Autophagy mediates the mitotic senescence transition. Genes Dev. 2009, 23, 798–803. [Google Scholar] [CrossRef]
  127. Chang, T.C.; Hsu, M.F.; Wu, K.K. High glucose induces bone marrow-derived mesenchymal stem cell senescence by upregulating autophagy. PLoS ONE 2015, 10, e0126537. [Google Scholar] [CrossRef]
  128. Alfonzo, M.C.; Al Saedi, A.; Fulzele, S.; Hamrick, M.W. Extracellular Vesicles as Communicators of Senescence in Musculoskeletal Aging. JBMR Plus 2022, 6, e10686. [Google Scholar] [CrossRef]
  129. Jia, Q.; Xie, B.; Zhao, Z.; Huang, L.; Wei, G.; Ni, T. Lung cancer cells expressing a shortened CDK16 3′UTR escape senescence through impaired miR-485-5p targeting. Mol. Oncol. 2022, 16, 1347–1364. [Google Scholar] [CrossRef]
  130. Xia, W.; Chang, B.; Li, L.; Hu, T.; Ye, J.; Chen, H.; Li, W.; Zan, T.; Hou, M. MicroRNA therapy confers anti-senescent effects on doxorubicin-related cardiotoxicity by intracellular and paracrine signaling. Aging 2021, 13, 25256–25270. [Google Scholar] [CrossRef]
  131. Kim, Y.; Park, D.; Kim, H.; Choi, M.; Lee, H.; Lee, Y.S.; Choe, J.; Kim, Y.M.; Jeoung, D. miR-200b and cancer/testis antigen CAGE form a feedback loop to regulate the invasion and tumorigenic and angiogenic responses of a cancer cell line to microtubule-targeting drugs. J. Biol. Chem. 2013, 288, 36502–36518. [Google Scholar] [CrossRef] [PubMed]
  132. Bai, X.Y.; Ma, Y.; Ding, R.; Fu, B.; Shi, S.; Chen, X.M. miR-335 and miR-34a Promote renal senescence by suppressing mitochondrial antioxidative enzymes. J. Am. Soc. Nephrol. 2011, 22, 1252–1261. [Google Scholar] [CrossRef] [PubMed]
  133. Scarola, M.; Schoeftner, S.; Schneider, C.; Benetti, R. miR-335 directly targets Rb1 (pRb/p105) in a proximal connection to p53-dependent stress response. Cancer Res. 2010, 70, 6925–6933. [Google Scholar] [CrossRef] [PubMed]
  134. Wang, Z.; Shi, D.; Zhang, N.; Yuan, T.; Tao, H. MiR-217 promotes endothelial cell senescence through the SIRT1/p53 signaling pathway. J. Mol. Histol. 2021, 52, 257–267. [Google Scholar] [CrossRef]
  135. Mensa, E.; Guescini, M.; Giuliani, A.; Bacalini, M.G.; Ramini, D.; Corleone, G.; Ferracin, M.; Fulgenzi, G.; Graciotti, L.; Prattichizzo, F.; et al. Small extracellular vesicles deliver miR-21 and miR-217 as pro-senescence effectors to endothelial cells. J. Extracell. Vesicles 2020, 9, 1725285. [Google Scholar] [CrossRef]
  136. Kabir, T.D.; Leigh, R.J.; Tasena, H.; Mellone, M.; Coletta, R.D.; Parkinson, E.K.; Prime, S.S.; Thomas, G.J.; Paterson, I.C.; Zhou, D.; et al. A miR-335/COX-2/PTEN axis regulates the secretory phenotype of senescent cancer-associated fibroblasts. Aging 2016, 8, 1608–1635. [Google Scholar] [CrossRef]
  137. Shen, Y.; Liu, L.; Wang, M.; Xu, B.; Lyu, R.; Shi, Y.G.; Tan, L. TET2 Inhibits PD-L1 Gene Expression in Breast Cancer Cells through Histone Deacetylation. Cancers 2021, 13, 2207. [Google Scholar] [CrossRef]
  138. Li, Y.; Fang, G.; Cao, W.; Yuan, J.; Song, S.; Peng, H.; Wang, Y.; Wang, Q. Ezh2 Inhibits Replicative Senescence of Atrial Fibroblasts Through Promotion of H3K27me3 in the Promoter Regions of CDKN2a and Timp4 Genes. J. Inflamm. Res. 2022, 15, 4693–4708. [Google Scholar] [CrossRef]
  139. Shi, X.; Gong, L.; Liu, Y.; Hou, K.; Fan, Y.; Li, C.; Wen, T.; Qu, X.; Che, X. 4-phenylbutyric acid promotes migration of gastric cancer cells by histone deacetylase inhibition-mediated IL-8 upregulation. Epigenetics 2020, 15, 632–645. [Google Scholar] [CrossRef]
  140. Zhu, X.; Leboeuf, M.; Liu, F.; Grachtchouk, M.; Seykora, J.T.; Morrisey, E.E.; Dlugosz, A.A.; Millar, S.E. HDAC1/2 Control Proliferation and Survival in Adult Epidermis and Pre-Basal Cell Carcinoma through p16 and p53. J. Investig. Dermatol. 2022, 142, 77–87.e10. [Google Scholar] [CrossRef]
  141. Kim, Y.; Park, H.; Park, D.; Lee, Y.S.; Choe, J.; Hahn, J.H.; Lee, H.; Kim, Y.M.; Jeoung, D. Cancer/testis antigen CAGE exerts negative regulation on p53 expression through HDAC2 and confers resistance to anti-cancer drugs. J. Biol. Chem. 2010, 285, 25957–25968. [Google Scholar] [CrossRef] [PubMed]
  142. Li, C.; Deng, Z.; Zheng, G.; Xie, T.; Wei, X.; Huo, Z.; Bai, J. Histone Deacetylase 2 Suppresses Skeletal Muscle Atrophy and Senescence via NF-kappaB Signaling Pathway in Cigarette Smoke-Induced Mice with Emphysema. Int. J. Chron. Obstruct. Pulmon. Dis. 2021, 16, 1661–1675. [Google Scholar] [CrossRef] [PubMed]
  143. Kim, J.Y.; Hwang, H.G.; Lee, J.Y.; Kim, M.; Kim, J.Y. Cortactin deacetylation by HDAC6 and SIRT2 regulates neuronal migration and dendrite morphogenesis during cerebral cortex development. Mol. Brain 2020, 13, 105. [Google Scholar] [CrossRef]
  144. Kaur, S.; Rajoria, P.; Chopra, M. HDAC6: A unique HDAC family member as a cancer target. Cell. Oncol. 2022, 45, 779–829. [Google Scholar] [CrossRef]
  145. Urdiciain, A.; Erausquin, E.; Zelaya, M.V.; Zazpe, I.; Lanciego, J.L.; Melendez, B.; Rey, J.A.; Idoate, M.A.; Riobo-Del Galdo, N.A.; Castresana, J.S. Silencing of Histone Deacetylase 6 Decreases Cellular Malignancy and Contributes to Primary Cilium Restoration, Epithelial-to-Mesenchymal Transition Reversion, and Autophagy Inhibition in Glioblastoma Cell Lines. Biology 2021, 10, 467. [Google Scholar] [CrossRef] [PubMed]
  146. Li, S.; Liu, X.; Chen, X.; Zhang, L.; Wang, X. Histone deacetylase 6 promotes growth of glioblastoma through inhibition of SMAD2 signaling. Tumour. Biol. 2015, 36, 9661–9665. [Google Scholar] [CrossRef] [PubMed]
  147. Kuroki, H.; Anraku, T.; Kazama, A.; Shirono, Y.; Bilim, V.; Tomita, Y. Histone deacetylase 6 inhibition in urothelial cancer as a potential new strategy for cancer treatment. Oncol. Lett. 2021, 21, 64. [Google Scholar] [CrossRef] [PubMed]
  148. Wang, Q.; Tan, R.; Zhu, X.; Zhang, Y.; Tan, Z.; Su, B.; Li, Y. Oncogenic K-ras confers SAHA resistance by up-regulating HDAC6 and c-myc expression. Oncotarget 2016, 7, 10064–10072. [Google Scholar] [CrossRef]
  149. Yang, M.H.; Laurent, G.; Bause, A.S.; Spang, R.; German, N.; Haigis, M.C.; Haigis, K.M. HDAC6 and SIRT2 regulate the acetylation state and oncogenic activity of mutant K-RAS. Mol. Cancer Res. 2013, 11, 1072–1077. [Google Scholar] [CrossRef]
  150. Yang, W.B.; Hsu, C.C.; Hsu, T.I.; Liou, J.P.; Chang, K.Y.; Chen, P.Y.; Liu, J.J.; Yang, S.T.; Wang, J.Y.; Yeh, S.H.; et al. Increased activation of HDAC1/2/6 and Sp1 underlies therapeutic resistance and tumor growth in glioblastoma. Neuro Oncol. 2020, 22, 1439–1451. [Google Scholar] [CrossRef]
  151. Moujaber, O.; Fishbein, F.; Omran, N.; Liang, Y.; Colmegna, I.; Presley, J.F.; Stochaj, U. Cellular senescence is associated with reorganization of the microtubule cytoskeleton. Cell. Mol. Life Sci. 2019, 76, 1169–1183. [Google Scholar] [CrossRef] [PubMed]
  152. Song, Y.W.; Lim, Y.; Cho, S.K. 2,4 Di tert butylphenol, a potential HDAC6 inhibitor, induces senescence and mitotic catastrophe in human gastric adenocarcinoma AGS cells. Biochim. Biophys. Acta. Mol. Cell Res. 2018, 1865, 675–683. [Google Scholar] [CrossRef]
  153. Ding, G.; Liu, H.D.; Huang, Q.; Liang, H.X.; Ding, Z.H.; Liao, Z.J.; Huang, G. HDAC6 promotes hepatocellular carcinoma progression by inhibiting P53 transcriptional activity. FEBS Lett. 2013, 587, 880–886. [Google Scholar] [CrossRef] [PubMed]
  154. Banik, D.; Noonepalle, S.; Hadley, M.; Palmer, E.; Gracia-Hernandez, M.; Zevallos-Delgado, C.; Manhas, N.; Simonyan, H.; Young, C.N.; Popratiloff, A.; et al. HDAC6 Plays a Noncanonical Role in the Regulation of Antitumor Immune Responses, Dissemination, and Invasiveness of Breast Cancer. Cancer Res. 2020, 80, 3649–3662. [Google Scholar] [CrossRef] [PubMed]
  155. Zhao, L.; Chen, H.; Zhang, Q.; Ma, J.; Hu, H.; Xu, L. ATF4-mediated microRNA-145/HDAC4/p53 axis affects resistance of colorectal cancer cells to 5-fluorouracil by regulating autophagy. Cancer Chemother. Pharmacol. 2022, 89, 595–607. [Google Scholar] [CrossRef]
  156. Zhang, X.; Qi, Z.; Yin, H.; Yang, G. Interaction between p53 and Ras signaling controls cisplatin resistance via HDAC4- and HIF-1alpha-mediated regulation of apoptosis and autophagy. Theranostics 2019, 9, 1096–1114. [Google Scholar] [CrossRef]
  157. Lee, Y.; Song, M.J.; Park, J.H.; Shin, M.H.; Kim, M.K.; Hwang, D.; Lee, D.H.; Chung, J.H. Histone deacetylase 4 reverses cellular senescence via DDIT4 in dermal fibroblasts. Aging 2022, 14, 4653–4672. [Google Scholar] [CrossRef]
  158. Di Giorgio, E.; Paluvai, H.; Dalla, E.; Ranzino, L.; Renzini, A.; Moresi, V.; Minisini, M.; Picco, R.; Brancolini, C. HDAC4 degradation during senescence unleashes an epigenetic program driven by AP-1/p300 at selected enhancers and super-enhancers. Genome Biol. 2021, 22, 129. [Google Scholar] [CrossRef]
  159. Paluvai, H.; Di Giorgio, E.; Brancolini, C. Unscheduled HDAC4 repressive activity in human fibroblasts triggers TP53-dependent senescence and favors cell transformation. Mol. Oncol. 2018, 12, 2165–2181. [Google Scholar] [CrossRef]
  160. Dey, D.K.; Kang, S.C. CopA3 peptide induces permanent cell-cycle arrest in colorectal cancer cells. Mech. Ageing Dev. 2021, 196, 111497. [Google Scholar] [CrossRef]
  161. Onorati, A.; Havas, A.P.; Lin, B.; Rajagopal, J.; Sen, P.; Adams, P.D.; Dou, Z. Upregulation of PD-L1 in Senescence and Aging. Mol. Cell. Biol. 2022, 42, e0017122. [Google Scholar] [CrossRef] [PubMed]
  162. Uceda-Castro, R.; Margarido, A.S.; Cornet, L.; Vegna, S.; Hahn, K.; Song, J.Y.; Putavet, D.A.; van Geldorp, M.; Citirikkaya, C.H.; de Keizer, P.L.J.; et al. Re-purposing the pro-senescence properties of doxorubicin to introduce immunotherapy in breast cancer brain metastasis. Cell Rep. Med. 2022, 3, 100821. [Google Scholar] [CrossRef] [PubMed]
  163. Roberson, R.S.; Kussick, S.J.; Vallieres, E.; Chen, S.Y.; Wu, D.Y. Escape from therapy-induced accelerated cellular senescence in p53-null lung cancer cells and in human lung cancers. Cancer Res. 2005, 65, 2795–2803. [Google Scholar] [CrossRef] [PubMed]
  164. Dorr, J.R.; Yu, Y.; Milanovic, M.; Beuster, G.; Zasada, C.; Dabritz, J.H.; Lisec, J.; Lenze, D.; Gerhardt, A.; Schleicher, K.; et al. Synthetic lethal metabolic targeting of cellular senescence in cancer therapy. Nature 2013, 501, 421–425. [Google Scholar] [CrossRef] [PubMed]
  165. Samaraweera, L.; Adomako, A.; Rodriguez-Gabin, A.; McDaid, H.M. A Novel Indication for Panobinostat as a Senolytic Drug in NSCLC and HNSCC. Sci. Rep. 2017, 7, 1900. [Google Scholar] [CrossRef]
  166. Qin, G.; Li, Y.; Xu, X.; Wang, X.; Zhang, K.; Tang, Y.; Qiu, H.; Shi, D.; Zhang, C.; Long, Q.; et al. Panobinostat (LBH589) inhibits Wnt/β-catenin signaling pathway via upregulating APCL expression in breast cancer. Cell. Signal. 2019, 59, 62–75. [Google Scholar] [CrossRef]
  167. Beltzig, L.; Christmann, M.; Kaina, B. Abrogation of Cellular Senescence Induced by Temozolomide in Glioblastoma Cells: Search for Senolytics. Cells 2022, 11, 2588. [Google Scholar] [CrossRef]
  168. Fleury, H.; Malaquin, N.; Tu, V.; Gilbert, S.; Martinez, A.; Olivier, M.A.; Sauriol, A.; Communal, L.; Leclerc-Desaulniers, K.; Carmona, E.; et al. Exploiting interconnected synthetic lethal interactions between PARP inhibition and cancer cell reversible senescence. Nat. Commun. 2019, 10, 2556. [Google Scholar] [CrossRef]
  169. Zhang, Y.; Zhang, Q.; Chu, Z.; Chen, L.; Chen, J.; Yang, Y.; Tang, H.; Cheng, G.; Ma, A.; Zhang, Y.; et al. Oridonin acts as a novel senolytic by targeting glutathione S-transferases to activate the ROS-p38 signaling axis in senescent cells. Chem. Commun. 2022, 58, 13250–13253. [Google Scholar] [CrossRef]
  170. Koyanagi, A.; Kotani, H.; Iida, Y.; Tanino, R.; Kartika, I.D.; Kishimoto, K.; Harada, M. Protective roles of cytoplasmic p21Cip1/Waf1 in senolysis and ferroptosis of lung cancer cells. Cell Prolif. 2022, 55, e13326. [Google Scholar] [CrossRef]
  171. Du, D.; Tang, X.; Li, Y.; Gao, Y.; Chen, R.; Chen, Q.; Wen, J.; Wu, T.; Zhang, Y.; Lu, H.; et al. Senotherapy Protects against Cisplatin-Induced Ovarian Injury by Removing Senescent Cells and Alleviating DNA Damage. Oxid. Med. Cell. Longev. 2022, 2022, 9144644. [Google Scholar] [CrossRef] [PubMed]
  172. Sabarwal, A.; van Rooyen, J.C.; Caburet, J.; Avgenikos, M.; Dheeraj, A.; Ali, M.; Mishra, D.; de Meester, J.S.B.; Stander, S.; van Otterlo, W.A.L.; et al. A novel 4′-brominated derivative of fisetin induces cell cycle arrest and apoptosis and inhibits EGFR/ERK1/2/STAT3 pathways in non-small-cell lung cancer without any adverse effects in mice. FASEB J. 2022, 36, e22654. [Google Scholar] [CrossRef] [PubMed]
  173. Banito, A.; Rashid, S.T.; Acosta, J.C.; Li, S.; Pereira, C.F.; Geti, I.; Pinho, S.; Silva, J.C.; Azuara, V.; Walsh, M.; et al. Senescence impairs successful reprogramming to pluripotent stem cells. Genes Dev. 2009, 23, 2134–2139. [Google Scholar] [CrossRef]
  174. Woo, J.; Shin, S.; Cho, E.; Ryu, D.; Garandeau, D.; Chajra, H.; Frechet, M.; Park, D.; Jung, E. Senotherapeutic-like effect of Silybum marianum flower extract revealed on human skin cells. PLoS ONE 2021, 16, e0260545. [Google Scholar] [CrossRef]
  175. Sasaki, N.; Itakura, Y.; Toyoda, M. Rapamycin promotes endothelial-mesenchymal transition during stress-induced premature senescence through the activation of autophagy. Cell Commun. Signal. 2020, 18, 43. [Google Scholar] [CrossRef] [PubMed]
  176. Wiley, C.D.; Flynn, J.M.; Morrissey, C.; Lebofsky, R.; Shuga, J.; Dong, X.; Unger, M.A.; Vijg, J.; Melov, S.; Campisi, J. Analysis of individual cells identifies cell-to-cell variability following induction of cellular senescence. Aging Cell 2017, 16, 1043–1050. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Characteristics of senescence. (A) SAHF is senescence-associated heterochromatin foci. Senescence is associated with SAHF in which expression of growth-related genes, such as targets of E2F, is suppressed. SASP refers to the senescence-associated secretory phenotype. Increased SA-β-gal activity leads to increased lysosomal mass. ↑ indicates an increase in expression/activity. The hollow arrows indicate the direction of the reaction. (B) Senescence is transmitted in paracrine and juxtacrine fashions. Senescent cells spread senescence via the SASP.
Figure 1. Characteristics of senescence. (A) SAHF is senescence-associated heterochromatin foci. Senescence is associated with SAHF in which expression of growth-related genes, such as targets of E2F, is suppressed. SASP refers to the senescence-associated secretory phenotype. Increased SA-β-gal activity leads to increased lysosomal mass. ↑ indicates an increase in expression/activity. The hollow arrows indicate the direction of the reaction. (B) Senescence is transmitted in paracrine and juxtacrine fashions. Senescent cells spread senescence via the SASP.
Ijms 24 03436 g001
Figure 2. Factors inducing senescence and the mechanisms of senescence are shown. (A) Various factors induce senescence. Senescence initially leads to tumor suppression. Senescent cells can also promote tumor progression by recruiting immune suppressor cells, such as Treg cells and myeloid-derived suppressor cells (MDSCs). SAHA, an inhibitor of HDACs, is suberohydroxamic acid. ↑ indicates an increase in expression/activity. ↓ indicates a decrease in expression/activity. The hollow arrows indicate the direction of the reaction. (B) The mechanisms of therapy-induced senescence are also shown.
Figure 2. Factors inducing senescence and the mechanisms of senescence are shown. (A) Various factors induce senescence. Senescence initially leads to tumor suppression. Senescent cells can also promote tumor progression by recruiting immune suppressor cells, such as Treg cells and myeloid-derived suppressor cells (MDSCs). SAHA, an inhibitor of HDACs, is suberohydroxamic acid. ↑ indicates an increase in expression/activity. ↓ indicates a decrease in expression/activity. The hollow arrows indicate the direction of the reaction. (B) The mechanisms of therapy-induced senescence are also shown.
Ijms 24 03436 g002
Figure 3. Mechanism of OIS. ↑ indicates an increase in expression/activity. ↓ indicates a decrease in expression/activity. The hollow arrows denote the direction of the reaction. OPN is osteopontin. TLR2 is toll-like receptor 2, and IER2 is immediate early response.
Figure 3. Mechanism of OIS. ↑ indicates an increase in expression/activity. ↓ indicates a decrease in expression/activity. The hollow arrows denote the direction of the reaction. OPN is osteopontin. TLR2 is toll-like receptor 2, and IER2 is immediate early response.
Ijms 24 03436 g003
Figure 4. Roles of the SASP in tumor promotion and tumor suppression. The SASP of senescent cells regulates various life processes in a paracrine fashion. The SASP eliminates senescent cancer cells (senescence surveillance). The SASP exerts immune suppression to promote tumor progression. The SASP contains various cytokines/chemokines, MMPs, and soluble extracellular vesicles (EVs) to promote these activities. The hollow arrows and black arrows indicate the direction of reaction. → indicates the direction of the reaction. ↑ indicates an increase in expression/activity. ↓ indicates a decrease in expression/activity. The T bar indicates a negative regulation.
Figure 4. Roles of the SASP in tumor promotion and tumor suppression. The SASP of senescent cells regulates various life processes in a paracrine fashion. The SASP eliminates senescent cancer cells (senescence surveillance). The SASP exerts immune suppression to promote tumor progression. The SASP contains various cytokines/chemokines, MMPs, and soluble extracellular vesicles (EVs) to promote these activities. The hollow arrows and black arrows indicate the direction of reaction. → indicates the direction of the reaction. ↑ indicates an increase in expression/activity. ↓ indicates a decrease in expression/activity. The T bar indicates a negative regulation.
Ijms 24 03436 g004
Figure 5. Autophagy leads to the induction of senescence. Autophagy is induced in cancer cells under various stresses. Complete autophagy usually results in cell death. Incomplete autophagy leads to senescence. ↑ indicates an increase in expression/activity. ↓ indicates a decrease in expression/activity. Hollow arrows and black arrows denote the direction of the reaction. The T bar indicates negative regulation. ULK3 is unc51-like autophagy activating kinase 3 and Ub is ubiquitin.
Figure 5. Autophagy leads to the induction of senescence. Autophagy is induced in cancer cells under various stresses. Complete autophagy usually results in cell death. Incomplete autophagy leads to senescence. ↑ indicates an increase in expression/activity. ↓ indicates a decrease in expression/activity. Hollow arrows and black arrows denote the direction of the reaction. The T bar indicates negative regulation. ULK3 is unc51-like autophagy activating kinase 3 and Ub is ubiquitin.
Ijms 24 03436 g005
Table 1. Roles of miRNAs in senescence and the mechanisms.
Table 1. Roles of miRNAs in senescence and the mechanisms.
miRNAsEffect on
Senescence
MechanismReference
miR-34aPromotes senescenceTargets Wnt/Notch/BCL-2[128]
miR-485-5pPromotes senescence (tumor-suppressive functions)Targets CDK16
C-MYC ↑
PD-L1 ↑
[129]
miR-199a-3pInhibits senescenceSASP ↓[130]
miR-200 familyInhibits senescenceEMT ↓[67]
miR-335/-34aPromotes premature senescenceSOD ↓ Txnrd2 ↓
ROS ↑
[132]
miR-335Promotes DNA-damage-induced senescenceRb1 ↓
Forms a positive feedback loop with p53
[133]
miR-217Promotes vascular endothelial cell senescenceTargets SIRT1/p53 signaling[134]
miR-21-5p/miR-217Promotes endothelial cell senescenceTargets DNMT1 and SIRT1[135]
miR-335Promotes senescence, cancer cell motilityPTEN ↓
SASP ↑
[136]
↓ indicates a decrease in expression/activity. ↑ indicates an increase in expression/activity. Rb1 is retinoblastoma 1.
Table 2. Roles of HDACs in senescence and the mechanisms.
Table 2. Roles of HDACs in senescence and the mechanisms.
HDACEffect on SenescenceMechanismCellsReference
HDAC2/
HDAC7
Inhibits senescenceRNAi of HDAC7 increases levels of IL-6, IL-8, and MMP-3Dermal fibroblasts[33]
HDAC2Inhibits senescenceMURF1 ↓ ATROGIN1 ↓
P53 ↓, P21 ↓
C2C12 cells[142]
HDAC6Inhibits senescenceMicrotubule stability ↑ ROCK ↓Kidney epithelial cells[150]
HDAC4Inhibits senescenceDDI4 ↑Dermal fibroblasts[157]
HDAC4Promotes senescenceTP53 ↑Primary fibroblasts[159]
↓ denotes a decrease in expression/activity; ↑ denotes an increased expression/activity. MMP-3, matrix metalloproteinase-3; RNAi, RNA interference; ROCK, Rho-associated protein kinase; TP53, tumor protein 539.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Jo, H.; Shim, K.; Jeoung, D. The Potential of Senescence as a Target for Developing Anticancer Therapy. Int. J. Mol. Sci. 2023, 24, 3436. https://doi.org/10.3390/ijms24043436

AMA Style

Jo H, Shim K, Jeoung D. The Potential of Senescence as a Target for Developing Anticancer Therapy. International Journal of Molecular Sciences. 2023; 24(4):3436. https://doi.org/10.3390/ijms24043436

Chicago/Turabian Style

Jo, Hyein, Kyeonghee Shim, and Dooil Jeoung. 2023. "The Potential of Senescence as a Target for Developing Anticancer Therapy" International Journal of Molecular Sciences 24, no. 4: 3436. https://doi.org/10.3390/ijms24043436

APA Style

Jo, H., Shim, K., & Jeoung, D. (2023). The Potential of Senescence as a Target for Developing Anticancer Therapy. International Journal of Molecular Sciences, 24(4), 3436. https://doi.org/10.3390/ijms24043436

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop