Next Article in Journal
Analysis of the Type 4 Effectome across the Genus Rickettsia
Previous Article in Journal
Exploring Highly Conserved Regions of SARS-CoV-2 Spike S2 Subunit as Targets for Fusion Inhibition Using Chimeric Proteins
Previous Article in Special Issue
Scientific Discoveries Supporting Theories in Science: From Thinking to Practice
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Senolytic Therapy: A Potential Approach for the Elimination of Oncogene-Induced Senescent HPV-Positive Cells

1
Department of Pharmacology and Public Health, Faculty of Medicine, The Hashemite University, Zarqa 13133, Jordan
2
Department of Microbiology, Pathology, and Forensic Medicine, Faculty of Medicine, The Hashemite University, Zarqa 13133, Jordan
3
Department of Anatomy and Histology, Faculty of Medicine, The University of Jordan, Amman 11942, Jordan
4
Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA
5
Department of Pharmacology and Toxicology, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh 33516, Egypt
6
Massey Cancer Center, Virginia Commonwealth University, Richmond, VA 23298, USA
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(24), 15512; https://doi.org/10.3390/ijms232415512
Submission received: 18 October 2022 / Revised: 29 November 2022 / Accepted: 2 December 2022 / Published: 8 December 2022

Abstract

:
Senescence represents a unique cellular stress response characterized by a stable growth arrest, macromolecular alterations, and wide spectrum changes in gene expression. Classically, senescence is the end-product of progressive telomeric attrition resulting from the repetitive division of somatic cells. In addition, senescent cells accumulate in premalignant lesions, in part, as a product of oncogene hyperactivation, reflecting one element of the tumor suppressive function of senescence. Oncogenic processes that induce senescence include overexpression/hyperactivation of H-Ras, B-Raf, and cyclin E as well as inactivation of PTEN. Oncogenic viruses, such as Human Papilloma Virus (HPV), have also been shown to induce senescence. High-risk strains of HPV drive the immortalization, and hence transformation, of cervical epithelial cells via several mechanisms, but primarily via deregulation of the cell cycle, and possibly, by facilitating escape from senescence. Despite the wide and successful utilization of HPV vaccines in reducing the incidence of cervical cancer, this measure is not effective in preventing cancer development in individuals already positive for HPV. Accordingly, in this commentary, we focus on the potential contribution of oncogene and HPV-induced senescence (OIS) in cervical cancer. We further consider the potential utility of senolytic agents for the elimination of HPV-harboring senescent cells as a strategy for reducing HPV-driven transformation and the risk of cervical cancer development.

1. Introduction

Cellular replicative senescence was first described by Leonard Hayflick and Paul Moorhead more than five decades ago as a stable exit from the cell cycle in non-transformed fibroblasts [1,2]. Senescence is a specialized form of growth arrest that plays a dynamic role in mediating multiple physiological and pathological processes [3]. The senescent growth arrest is stable and durable, in that, senescent cells are unresponsive to mitogenic drivers, but remain viable and metabolically active [4]. Replicative senescence represents the classical response to telomeric dysfunction that occurs due to the “end replication crisis” in dividing eukaryotic cells [5]. In addition to preventing the proliferation of cells containing dysfunctional telomeres, senescence is induced in response to other stimuli that also have the potential to promote malignant transformation, including oxidative and genotoxic stress, the latter often precipitated by exposure to DNA- damaging drugs or ionizing radiation [6]. With reference to the focus of this article, senescence is also a well-established response to oncogene overexpression [7], thereby presenting a fundamental barrier to malignant transformation [8].
In support of senescence as a tumor suppressive mechanism, senescent cells have been shown to accumulate in premalignant lesions [9]. The classical example is the identification of senescence markers in naevi containing B-Raf overexpressing melanocytes [10]. In addition to B-Raf, the activation of several other oncogenes, such as Ras, Akt, and E2F, or conversely, the inactivation of certain tumor suppressor genes, such as PTEN, can also promote senescence in somatic cells [11]. In most cases, senescence is sufficient to block oncogenic transformation by conferring a permanent abrogation of growth in cells harboring an active oncogene; consequently, bypassing oncogene-induced senescence is essential for malignant transformation. Nevertheless, the co-expression of an additional oncogene or inactivation of a tumor suppressor gene, or a “second hit”, is generally accepted as being required for a cell to overcome (or escape) Oncogene-Induced Senescence (OIS) and proceed towards malignancy. However, the function of senescence in transformation is likely to be more complex, as recent evidence has suggested that the accumulation of oncogene-induced senescent cells contributes to disease progression through cell-non-autonomous effects [12]. While OIS is not directly related to telomeric dysfunction, it is also a product of DNA damage which results when cells are undergoing abnormally rapid replication due to oncogene-driven proliferative stimulation [13]. Despite the spectrum of senescence-inducing insults, the senescent phenotype shares several prominent features, as detailed below. Nonetheless, it is essential to note that senescent cells are highly heterogeneous, with extensive diversity in their gene expression profiles and the manifestation of senescence markers [14].
Human papillomavirus (HPV) infection is the leading cause of cervical cancer [15]. It has been established that infection with high-risk strains of HPV, namely, HPV-16 and HPV-18, is associated with senescence induction, a process largely dictated through the actions of the viral oncoproteins E6 and E7 (both suppression of E6 or E7 by E2 in HPV-infected cells or transfection of naïve cells with E7) [16,17,18]. Both E6 and E7 are considered essential factors for carcinogenesis in HPV-positive cells, as the continuous expression of these oncoproteins plays a key role in the induction and maintenance of the cellular malignant features [16]. E6 and E7 expression is associated with the degradation of the tumor suppressors p53 and retinoblastoma (Rb) proteins, respectively [19]. Conversely, E6 suppression induces p53 expression and upregulates the cyclin-dependent kinase inhibitor p21Cip1, leading to cell cycle arrest and senescence induction [20]. Similarly, the suppression of E7 induces Rb expression and the formation of the Rb-E2F complex leading to transcriptional repression and cell cycle arrest [20]. Thus, HPV infection represents a potential pathway to OIS, making the accumulation of HPV-positive senescent cells in cervical lesions a likely precancerous process. Consequently, targeting either E6 and/or E7 oncoproteins in cancerous cells or senescent cells, represents a potential strategy for the suppression of HPV-positive tumor cell growth, thereby reducing the risk of cervical cancer in infected individuals [21]. In this commentary, we provide an overview of OIS, and its complex contribution to cancer development, and, more specifically, discuss HPV-Induced Senescence as a variant of OIS. Moreover, we propose the use of senolytics, a newly emerging drug class that selectively targets senescent cells, as a novel approach for the mitigation of the progression of premalignant lesions toward cancer, including HPV-infected Cervical Intraepithelial Neoplasia (CIN).

2. Oncogene-Induced Senescence

2.1. Hallmarks of Senescence

Aside from the durable growth arrest, which is considered the primary characteristic of senescence, senescent cells exhibit a plethora of features that collectively represent the senescent phenotype [22]. The senescence characteristics are both cell-autonomous and cell-non-autonomous [23]. Intrinsically, senescent cells exhibit an enlarged and flattened morphology, chromatin rearrangement known as senescence-associated heterochromatic foci (SAHFs) [24], which are frequently associated with OIS, and enhanced expression of the senescence-associated β-galactosidase (SA-β-gal) activity [25], which reflects increased lysosomal content [26]. Furthermore, senescent cells are characterized by the accumulation of reactive oxygen species (ROS) coupled with ROS-mediated macromolecular damage [27]. In addition, under persistent activation of the DNA damage repair response (DDR), senescent cells display nuclear foci termed DNA segments with chromatin alterations reinforcing senescence (DNA-SCARS); these are crucial elements that further sustain the senescent state due to the activation of DDR proteins such as p53 [18,28]. Another essential feature defining senescent cells is the production of a diverse range of cytokines, chemokines, extracellular matrix proteases, growth factors, and other signaling molecules, collectively termed the senescence-associated secretory phenotype (SASP), which largely mediates the extrinsic effects of senescence [29,30]. However, none of these features is uniquely representative or specific to senescence as they can also be observed in other forms of cellular stress or cell cycle arrest [22]. Accordingly, senescent cells are usually identified by the examination of a profile of multiple senescence-associated biomarkers [31] (Table 1).

2.1.1. Growth Arrest

Senescence activation is commonly observed at the G1 phase of the cell cycle, but cell growth may also be arrested in the G1, S, or G2/M phases of the cell cycle [47]. The senescent growth arrest is governed by several cell cycle regulators, primarily the p53/p21Cip1 and p16INK4a/pRb pathways, which are activated upon entry into senescence [48,49]. The essential role of p53 in regulating the senescent growth arrest stems from the transcriptional upregulation of the Cyclin-dependent Kinase Inhibitor (CDKI) p21Cip1. In order to prevent the proliferation of damaged cells, the tumor suppressor protein p53 transactivates its target genes, whose gene products promote either apoptosis to remove damaged cells from the body and/or cell cycle arrest and DNA damage repair mechanisms to mitigate genotoxic injury [50,51,52]. p53 binding can repress the expression of several genes including survivin, CDC25C, CDC25B, CHK2, cyclin B, CKS1B, RECQL4, and cdc20 while enhancing the transcription of p21Cip1 [51,53]. p21Cip1 inhibits the activity of various cyclin-dependent kinases (CDKs) by binding and blocking the ATP binding site of CDKs, subsequently preventing CDK phosphorylation and interfering with cell cycle progression [51,54,55]. p21Cip1 inhibition of cyclin D-CDK4/6 complexs activation prevents Rb phosphorylation and enhances Rb-E2F complex sequestration. In proliferating cells, Rb is hyperphosphorylated by CDK/cyclin complexes, allowing the release of the transcription factor, E2F, from the Rb-E2F complex, and transcription of key S-phase genes [56]; hence binding of Rb to E2F represses the transcription of cell cycle genes such as hTERT, EZH2, and CHKI [50,57]. Despite the critical role of p21Cip1 in mediating the senescent growth arrest, the deletion of p21Cip1 is not sufficient to prevent mouse fibroblasts from undergoing a proliferative arrest upon senescence induction, indicating that p21Cip1 is not absolutely required for the senescent growth arrest to occur [58]. This is true, in part, because growth arrest can be mediated through the p16INK4a/Rb pathway [58,59]. p16INK4a, an inhibitor of CDK4 and CDK6, is more closely associated with senescence induction and is considered a significant activator of Rb [33,48,60].
Similarly to p21Cip1, p16INK4a is a CDKI that interferes with cyclin D-CDK4/6 complexes, maintaining Rb-E2F binding, and stabilizing a G1/S cell cycle arrest by activating the pRb checkpoint [48,54,60]. p16INK4a is classically activated in senescent cells and is frequently utilized as a characteristic marker to identify senescent cells in a variety of senescence models [61]. Moreover, it has been demonstrated that cells that are positive for p16INK4a often display other markers of senescence including SA-β-gal, altered morphology, and increased expression of SASP (discussed below) [61]. Accordingly, and since the senescent growth arrest, particularly that associated with OIS, interferes with the progression of cells at risk of malignant transformation, p16INK4a is considered to function as a tumor suppressor gene [62,63]. Moreover, the CDK2A locus encoding for p16INK4a is frequently inactivated through deletions, point mutations, or hypermethylation in many malignancies including melanoma, leukemia, pancreatic and head and neck carcinomas, reflecting its established tumor suppressor function [64]. Importantly, as p16INK4a is expressed in several types of senescent cells, its overexpression is also considered a major hallmark of HPV-associated cancers, including HPV-positive cervical carcinomas and HPV-positive head and neck cancers (see section below) [20,65]. In HPV-positive cancers, the virus-encoded oncoprotein, E7, mediates the inactivation of Rb, resulting in the release of E2F from the inhibitory complex and entry into the S phase [66]. In order to compensate for the loss of Rb function, cells increase expression of p16INK4a in effort to suppress cell cycle progression [66].
Senescent growth arrest is unique in that it is highly stable and durable. Unlike quiescent cells that can resume proliferation when favorable growth conditions are restored, senescent cells are unresponsive to growth stimuli [67]. It should also be emphasized that quiescent cells do not demonstrate the hallmarks of senescence. Furthermore, senescent cells preserve their active metabolic status, albeit dysregulated, as evidenced by a remarkable increase in oxygen consumption rate and mitochondrial oxidative metabolism in OIS cells as opposed to quiescent cells which typically have reduced metabolic rates [41]. Nevertheless, recent evidence has demonstrated that certain forms of senescence can be overcome, and that senescent cells, especially tumor cells induced into senescence by exposure to cancer therapy, can resume proliferation [68]. For example, H1299 non-small cell lung cancer cells exposed to camptothecin can regain proliferative ability upon discontinuation of drug exposure, although recovery from the senescent growth arrest was a rare event (approximately, 1 in 106 cells) [69]. Our own laboratory has also previously demonstrated that several tumor cell types (specifically, H460 lung, HCT116 colon, HN30 head and neck, Myc-CaP prostate, and 4T1 breast cancer cells) induced into senescence by exposure to chemotherapy or ionizing radiation can overcome the senescent growth arrest following short-term drug or radiation treatment [70,71,72].
It is noteworthy that conditional p53 inactivation or interference with SAHF formation can be permissive for the escape from doxorubicin-induced senescent growth arrest in tumor cells [73]. While the likelihood of the escape from the stable senescence-induced growth arrest is largely represented in tumor cell models, the reversal has also been observed in BJ cells induced into senescence by replicative exhaustion [74]. The stability of the senescent growth arrest is partly dependent on the expression of p16INK4a or p53, as their loss of function can be associated with a less stable form of growth arrest. For instance, senescent BJ cells, which have relatively low expression of p16INK4a, can re-enter the cell cycle upon p53 inactivation, in contrast to the more stable senescence in WI38 fibroblasts, where the p16INK4a/Rb pathway tends to be fully active [33,74]. The reversibility of senescence-mediated growth arrest has been also documented in OIS models including Ras-induced senescent pancreatic cells, Akt1-induced senescent pancreatic cells, and oncogene-induced senescent astrocytes [75,76,77]. The potential for escape from OIS is consistent with the premise that oncogene-overexpressing cells are likely to utilize this strategy to progress into a state of malignancy.

2.1.2. Morphological and Macromolecular Changes

Senescent cells are characterized by distinct morphological features. Specifically, senescent cells exhibit an enlarged, flattened appearance and irregularly-shaped nuclei [36]. The increase in cellular size is frequently accompanied by an increase in the size of the nucleus and nucleoli [78]. Senescent cells also develop numerous cytoplasmic vacuoles, increased numbers of cytoplasmic microfilaments, enlarged lysosomal bodies, and prominent Golgi apparatuses [42,79]. Moreover, senescent cells have enhanced lysosomal biogenesis, which is marked by increased expression of SA-β-gal, the most frequently used marker for the identification of senescent cells [80]. The accumulation of lipofuscin aggregates, proteins that accumulate progressively in the lysosomes of aged, post-mitotic cells, was also established as a hallmark of senescent cells [40,81,82,83].

2.1.3. DNA Damage

Persistent DNA damage is the most consistent trigger of senescence. Classically, as cells become replicatively exhausted, their telomeres reach a critical length and fail to bind telomere-capping proteins. In brief, a strip of telomeric nucleotides is excised each time a proliferating cell completes a cycle of DNA synthesis in preparation for mitosis [5]. Upon repetitive loss of the 6-base sequence of telomeric DNA, the cell reaches a point where further division cycles will result in chromosomal injury [84]. In fact, telomere attrition can lead to a number of DNA lesions including end-end chromosomal fusions, breakage of anaphase chromatin bridges, and translocations. However, it is the altered status of shortened telomeres, rather than the mere loss of telomeric DNA, that is directly linked to replicative senescence [37]. These dysfunctional telomeric ends are then recognized as “exposed” ends of DNA [85], subsequently activating the DDR pathways and leading to the prompt activation of cell cycle blockers such as p21Cip1 [86]. Importantly, dysfunctional telomeres continue to possess enough content of telomere-binding proteins to inhibit DNA repair, and thus, maintain a persistent DNA damage drive [87,88]. Similarly, oncogene activation results in hyperproliferation as well as a high degree of replicative stress, subsequently leading to the accumulation of single and double-stranded DNA breaks [89,90]. Double-stranded DNA breaks (DSBs) are a major activator of DDR, initiating autophosphorylation and activation of ATM, which then drives histone H2AX phosphorylation, facilitating DNA repair complex formation [6]. Moreover, ATM activation results in downstream phosphorylation of p53 and subsequent induction of p21Cip1, which mediates the senescent growth arrest [91].
DNA damage responses play an essential role in the activation of oncogene-induced senescence, given that inactivation of key proteins involved in DDR results in evasion of the senescent phenotype, continued proliferation, and malignant transformation [13,92]. Persistent DNA damage in senescent cells can be irreparable and associated with lesions encompassing PML nuclear bodies, lack of activation of the established DNA repair proteins, such as RAD51, but with activated p53 and CHK2, and suppressed DNA synthesis. These lesions are more likely to describe senescent cells in a stable growth arrest, and are collectively termed DNA segments with chromatin alterations reinforcing senescence (DNA-SCARS) [18]. Interestingly, and relevant to this review, artificial expression of the HPV oncoprotein, E7 in HCA2 cells results in the formation of DNA-SCARS, consistent with the ability of HPV to induce senescence [18]. Collectively, DNA damage and the activation of the DDR are major hallmarks of senescence, including OIS [13].

2.1.4. Mitochondrial Dysfunction

Senescent cells often accumulate damaged mitochondria characterized by (i) reduced oxidative potential, (ii) decreased mitochondrial membrane potential, (iii) structural changes, and (iv) build-up in free radicals [93,94]. In addition to being a component of replicative and therapy-induced senescence, mitochondrial dysfunction is frequently observed in models of OIS [95]. For example, upon Ras overexpression in normal human fibroblasts, mitochondria increase in mass, potentially due to reduced mitophagic turnover [96], and accumulate ROS leading to oxidative DNA injury [95]. Moreover, the induction of Ras reduces the energy-generating capacity of mitochondria marked by low ATP production [95]. However, unlike what occurs in aging-associated senescent cells, NAD+, which plays an important role in mitochondrial redox reactions, is increased rather than decreased (thus shifting the NAD+/NADPH balance up) in OIS models [97]. Still, in oncogene-induced senescent cells, the accumulation of ROS due to mitochondrial dysfunction appears to be necessary for gene expression changes leading to the activation of the SASP, which are largely mediated by the nuclear factor kappa B (NFκB) pathway [27]. Lastly, the role of mitochondria in regulating Ca+2 homeostasis is perturbed in senescent cells, and in fact, might facilitate the escape from OIS [98]. Interestingly, components of oncogenic HPV proteins can localize in juxtaposition to the mitochondrial inner membrane causing morphological changes and facilitating increased production of ROS [99]. This provides a possible link between HPV infection, the development of mitochondrial dysfunction, and OIS.

2.1.5. Epigenetic Changes

In addition to their constitutive structural roles, in senescent cells, heterochromatin and euchromatin exhibit distinct post-translational modifications of their histone proteins and associate with different sets of facultative chromatin binding proteins, together termed as Senescence-associated heterochromatin foci (SAHF) [43]. SAHF were first described by Scott Lowe and coworkers after observing that DAPI-stained senescent human cells displayed a relatively diffuse distribution of DNA throughout the nucleus, which appeared as bright, punctate DNA foci, and that the chromatin in these foci appeared more compact than during the normal interphase of growing cells [43]. Each SAHF might consist of a single condensed chromosome that is enriched with histone modifications and proteins that are associated with epigenetically silenced genes such as the E2F family [100]. SAHF mediate epigenetic regulation during OIS; however, SAHF are not considered a common feature of cellular senescence as they are not universally observed in cells undergoing replicative senescence and are rarely observed in vivo [100].
Histone edits involved in SAHF include trimethylation of histone H3 at lysine 9 (H3K9Me3) and its binding partner heterochromatin protein 1 (HP1) [42], and the persistent phosphorylation of the DNA DSBs marker H2AX (γH2AX) [42,100]. Functionally, SAHF repress the expression of proliferation-linked genes, such as cyclin A, thereby contributing to senescence-associated cell cycle arrest. Evidently, interference with the ability of senescent cells to condense their chromatin and to generate some of these classical histone modifications, such as H3K9Me3, can facilitate the escape from the senescent growth arrest [73]. Lastly, a functional pRB/p16INK4a pathway is required for the efficient formation of SAHF [101,102].

2.1.6. Resistance to Apoptosis

Senescent cells tend to be resistant to apoptosis. A potential mechanism for this characteristic of senescent cells is the upregulation of the anti-apoptotic members of the BCL-2 family [103]. More specifically, senescent cells (whether by replicative exhaustion, chemotherapy, radiation, or oncogene overexpression) appear to be largely dependent on BCL-XL for their survival, since the genetic or pharmacological inhibition of BCL-XL (and, to a lesser extent, BCL-W) results in selective and immediate induction of apoptosis in these cells [104]. However, in other studies comparing young cells and senescent cells exposed to H2O2, despite the low levels of BCL-2 in the senescent cells, they were still more resistant to apoptosis compared to their young counterparts [105]. Thus, the decrease of BCL-2 during cellular aging has no apparent impact on induced death in senescent cells. Other studies suggest that senescent cells being in a non-cycling state, along with increasing levels of CDK inhibition, may alternatively explain their resistance to cell death [105]. MCL-1 is another member of the BCL-2 family that plays a role in maintaining the survival of senescent cells since its selective inhibition commits these cells to cellular demise [106]. Another potential explanation of how senescent cells resist cell death is by downregulating the expression of caspase-3, which is responsible for the execution of apoptosis upon the mitochondrial release of cytochrome c and activation of the caspase pathway [107].
Recent evidence has also suggested a role for the Forkhead box O (FOXO4)-p53 interaction in the maintenance of senescent cell survival [108]. FOXO4 is part of a family of transcription factors that are involved in regulating gene expression of several cell survival pathways. Moreover, FOXO4 has been described to regulate senescence-associated pathways, especially in models of OIS. For example, BRAFV600E expression, which routinely triggers OIS, results in ROS build-up, JNK activation, and eventually, phosphorylation and activation of FOXO4 [109]. FOXO4, in turn, is capable of inducing a stable senescent growth arrest via upregulation of p21Cip1 (another p53 downstream target) expression [109]. The interaction between FOXO4 and p53 at sites of DNA damage appears to be responsible for shifting cell fate from apoptosis to senescence. Baar et al. showed that radiated senescent fibroblasts upregulate FOXO4. Once FOXO4 is inhibited prior to radiation exposure, cells instead undergo classical cytochrome c-mediated mitochondrial apoptosis, thus reflecting an important role for FOXO4 in rendering senescent cells resistant to cell death [110]. The importance of FOXO4-p53 interaction in preventing apoptosis in senescent cells has been confirmed in studies by Le et al., which showed that the selective targeting of FOXO4-p53 robustly kills senescent cells [111]. Overall, these mechanisms appear to account for the ability of senescent cells to persist for prolonged periods in vivo, including in premalignant lesions upon oncogene overexpression.

2.1.7. The SASP

In contrast to quiescent cells, senescent cells modulate their microenvironment via the secretion of proinflammatory chemokines, cytokines, growth factors, and proteases, collectively termed the senescence-associated secretory phenotype or SASP [112]. Indeed, the SASP represents a double-edged sword. On one hand, the SASP directly contributes to an insidious inflammatory process that triggers immunosurveillance of senescent cells [29], while on the other hand, the SASP can facilitate the ability of a subpopulation of senescent cells to evade immunorecognition [113,114]. Transcription of the SASP inflammatory genes is regulated primarily by two well-established transcription factors NF-κB and CCAAT-enhancer binding protein β (C/EBPβ) [115]. Analysis of the senescent cell chromatin identified the NF-kB subunit p65 (also known in humans as the RELA gene) as a major component of the transcription machinery that leads to the expression of cytokines and chemokines such as IL-6 and IL-8 [116]. Recent evidence has also demonstrated that the cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) signaling pathway, a critical marker of the innate immune response, stimulates and regulates the SASP as a consequence of the accumulation of cytoplasmic DNA (cytoplasmic chromatin fragments, mtDNA, and cDNA) in senescent cells [117].
The SASP exerts both autocrine and paracrine functions that influence senescent cells, but also neighboring cells, particularly innate and adaptive immune cells, which eventually result in the clearance of senescent cells and contribute to the final tissue hemostasis [118]. In an autocrine fashion, the SASP often reinforces senescence through factors such as CXCR-2 [119] and IL-6 [120] that can bind to self-receptors. In contrast, the SASP’s paracrine effects promote tumor suppression by the activation of either the p53/p21Cip1 or p16INK4a/Rb tumor suppressor pathways; at the same time, the SASP can drive the progression of premalignant cells to develop more aggressive phenotypes through, for example, the stimulation of epithelial-to-mesenchymal transition (EMT) [30]. Evidence also indicates that the SASP of senescent fibroblasts enhances proliferation and facilitates the malignant transformation of pre-malignant epithelial cells along with tumor vascularization; events mediated by the proinflammatory cytokines IL-6 [121], IL-8 [122], vascular endothelial growth factor (VEGF) [123], and the matrix metalloproteases (MMPs) [124], while inducing senescence in adjacent non-senescent cells as a bystander effect in a manner that contributes to the heterogeneity of premalignant lesions. Collectively, the SASP in general opposes the cell-autonomous tumor-suppressing function of senescence and allows senescent cells to contribute to chronic inflammation, aging processes, and cancer.

2.2. Evidence for Oncogene-Induced Senescence (OIS)

Genomic instability confers high tumorigenic potential and increases the risk of malignant transformation. Since eukaryotic cells undergo senescence as a consequence of telomere shortening/dysfunction, as mentioned previously, in essence, senescence reflects a tumor suppressive mechanism, in that it facilitates the suppression of further proliferation of cells with the potential of becoming cancerous [8]. In a similar fashion, OIS serves as a classical example of a form of senescence that is primarily induced in response to tumorigenic events. Specifically, OIS is a robustly brought about by aberrant activation of oncogenic signaling, which is generally driven by activating mutations of oncogenes such as Ras, Akt, E2F1, B-Raf, and cyclins [92,125], or the inactivation of tumor suppressor genes such as PTEN and NF1 [11]. One of the first observations of OIS was reported by Serrano et al., who showed that the insertion of an activated Ras allele (H-Ras V12) into primary diploid fibroblasts resulted in the induction of cellular senescence hallmarks including p53 and p16INK4a accumulation, an enlarged, flattened morphology, as well as suppressed mitotic activity [60]. These in vitro findings were later supported by Sarkisian et al. using in vivo models, further demonstrating that oncogene expression levels appeared to be critical for OIS induction [126]. In these studies, Sarkisian et al. utilized doxycycline-inducible transgenic mice that permit the “titration” of Ras activation, demonstrating that cellular proliferation as well as mammary epithelial hyperplasia, were stimulated by low levels of Ras activation; however, senescence required the establishment of high levels of Ras activation [126].
Several oncogenes have been associated with the induction of senescence (Table 2). In addition to Ras, a GTPase, first established to induce OIS by Serrano et al. [60], and later became the most frequently utilized oncogene to induce senescence experimentally, others have demonstrated that B-Raf, a serine/threonine-protein kinase, can induce a p16INK4a-driven senescent growth arrest in cells found in Spitz nevi (benign melanocytic precursors for melanoma) [10,127]. Hyperactivation of PI3K/Akt pathways have also been demonstrated to result in senescence induction through the promotion of p53 [128]. Overexpression of cyclins, such as cyclin E, has also been shown to promote OIS marked by SA-β-gal expression and p16INK4a upregulation [92]; that is, cells activate a senescence program to delay or interfere with the tumorigenic actions of drivers of the cell cycle. As is the case with oncogene overexpression, the inactivation of some tumor suppressor genes can also force somatic cells into a senescent growth arrest. For example, loss of the PTEN can precipitate p53-dependent OIS that prevents further cellular proliferation, and interferes with initial malignant transformation in vivo [129].
Oncogene-induced senescent cells have been shown to accumulate in premalignant lesions, which by definition, are rich in cells harboring mutations that either result in oncogene hyperactivation or tumor suppressor gene inactivation. For example, senescent cells accumulate in dysplastic skin and oral mucosal lesions [130]. Evidence for OIS has also been established in several other premalignant processes including human preneoplastic gastrointestinal lesions [92,131,132], pre-melanoma nevi [133], prostatic intraepithelial neoplasia [77], oral leukoplakia [134], and premalignant nasopharyngeal epithelium [135]. In premalignant lesions, OIS is thought to act as a “failsafe” protective mechanism, limiting the propagation of cells harboring oncogenic DNA [129]. For instance, high levels of Ras activation in mammary epithelial cells resulted in senescence induction in vivo and promoted the induction of tumor-suppressive pathways [126]. Inactivation of PTEN resulted in the promotion of p53-dependent senescence and prevented the onset of aggressive prostatic cancer [129]. Furthermore, induction of senescence increases the susceptibility to immune clearance, since both components of the adaptive and innate immune cells have the capacity to recognize and remove the deleterious accumulation of senescent cells [136,137,138]. Activation of DDR responses and senescence induction can act as barriers to malignant transformation, and cancer progression may rely on bypassing these checkpoints. Evidently, mutational inactivation of key DDR genes can drive the development of human somatic cancers [139] and frequently alters cellular susceptibility to senescence and apoptosis [140], allowing evasion of regulatory and cell death pathways.
Table 2. Examples of established oncogene/tumor suppressor genes implicated in the induction of Oncogene-Induced Senescence (OIS).
Table 2. Examples of established oncogene/tumor suppressor genes implicated in the induction of Oncogene-Induced Senescence (OIS).
Oncogene/Tumor Suppressor GeneAlterationFunctionModelPremalignant/Malignant LesionReference
c-mosOverexpressionSerine/threonine kinaseHuman fibroblastsLung cancer[92]
PTENLoss of functionTumor suppressor geneMurine embryonic fibroblastProstate cancer[129]
RasActivation Regulation of signal transductionMurine embryonic fibroblastPancreas, colon, and lung cancers[60]
RafActivation Ras signaling Human diploid fibroblast Lung adenomas [141]
AktActivationAkt signaling Murine embryonic fibroblasts
Endothelial cells
-[142]
E2F1OverexpressionPromotes G1 to S-phaseHuman diploid fibroblastsPituitary gland hyperplasia[143]
Cyclin EOverexpressionActivation of cyclin dependent kinase-2-Breast cancer[92]
E7OverexpressionInactivation of RbHCA2 human fibroblasts-[18]
In contrast to the anti-tumorigenic role of OIS, components of the SASP can promote tumorigenicity. SASP factors are highly variable and can enable both a tumor-suppressive and a tumor-promoting environment [144,145,146]. Depending on the spectrum of secreted factors, SASP factors can promote tumorigenesis through the activation of proliferation pathways, immunosuppression, and promotion of a migratory phenotype [147,148,149]. For example, secretion of specific SASP factors, such as IL-6 and IL-8, exert paracrine effects on surrounding cells and promote tumorigenesis in vivo [147,150]. Moreover, studies in a pancreas model showed an OIS-mediated tumor-promoting effect where the elimination of OIS cells decelerated the development of pancreatic cancer in animals [12]. These studies and others delineate the heterogeneous nature of OIS in different models, which may be mediated through different combinations of downstream effectors as well as the interplay between senescent cells and the microenvironment [10,92,129]. Thus, whether senescence induction is beneficial or harmful with regard to tumorigenesis is not fully understood and may be contextual in nature. Importantly, since HPV infection can induce OIS in somatic human cells, it is likely that the accumulation of HPV-positive senescent cells in cervical or head and neck premalignant lesions could contribute directly to tumorigenesis.

3. Human Papilloma Virus (HPV)-Induced Senescence

Human papillomavirus (HPV) infection is the most common sexually-transmitted infection among women globally [151]. Multiple clinical and epidemiological reports have clearly established the role of HPV infection as the primary etiologic factor for cervical cancer [152]. Furthermore, HPV infection is associated with cancerous transformation in the penis, vagina, anus, vulva, and oropharynx [153]. Of the fifteen high-risk (HR) HPV genotypes that can cause cancer at these sites, HPV 16 and 18 are the most common, causing 70–75% of cancer-associated lesions in humans [154]. Low-risk (LR) HPV genotypes 6 and 11 are rarely associated with cancer, and are instead more closely associated with warts and respiratory papillomatoses [155]. The oncogenic HPV is a double-stranded DNA virus that infects epithelial cells in the anogenital region or naturally discontinuous oropharyngeal epithelium. Most HPV infections remain asymptomatic and are cleared by the immune system in the 6–18 months after infection. Only a minority of infected patients fail to eliminate the infectious virus and, after a latency period, develop dysplastic changes such CIN.
HPV replication in target cells leads to the production of the early (E1–E7) and late proteins (L1, L2). The major and minor capsid proteins L1 and L2, respectively, enclose the HPV genome [156,157]. The virus capsid is made up of L1 protein pentamers, while virus-like particles, which are extremely protective and produce large amounts of neutralizing antibodies, are created when the L1 and L2 proteins are self-assembled [158,159]. The HPV E1 is the only viral protein exhibiting enzymatic activity in the Human Papillomavirus family, and its major known function is to control the viral DNA replication process [160]. HPV E2 is essential for viral genome replication, RNA transcription, and viral epigenome partitioning during replication [161,162]. HPV E4 has been demonstrated to cause G2/M arrest and aid in the amplification of E6/E7 viruses; it also contributes to viral genome amplification, and the maintenance of MAPK activity, and may interact with and stabilize E2 [163,164,165]. It has been demonstrated that HPV16 E5 cooperates with E7 in cell transformation, inhibits immunological response, and increases cell motility [166]. Importantly, the oncogenicity of HPV is primarily attributed to the oncogenic proteins E6 and E7, which interfere with select cell signaling pathways and continue to be expressed during tumor formation and progression [16]. E6 and E7 enhance cellular transformation through the inactivation of the tumor-suppressor proteins, p53, and Rb protein, respectively, which results in cell cycle disruption and the accumulation of DNA mutations [19].
While the tumor-suppressive aspect of senescence has been established in response to DNA aberration-associated telomere dysfunction or oncogene hyperactivation, its contribution to the suppression of virus-mediated transformation is not fully understood. A seminal finding by Baz-Martínez et al. indicates that senescence interferes with the replication, and thus the infectivity, of vesicular stomatitis virus (VSV) in several cell models including MEFs, MCF-7 breast and A549 lung tumor cells [167], strongly suggesting that senescence has antiviral properties. However, in these experiments, senescence was induced by classical means, e.g., replicative exhaustion or DNA-damaging agents, followed by viral infection. Thus, there was no examination of whether senescence is directly induced in somatic fibroblasts by VSV infection. Conversely, another report by Kim et al., presents opposing evidence. As in the work of Baz-Martinez, senescence was induced in primary human bronchial epithelial cells (HBE) and human dermal fibroblasts (HDF) through replicative exhaustion, which was followed by viral infection using influenza virus (IFV) and varicella-zoster virus (VZV) [168]. Unexpectedly, viral replication was enhanced in senescent cells in comparison to their non-senescent counterparts. That is, senescent HDF exhibited approximately 1.5-fold higher VZV infectivity titers than non-senescent HDF. Moreover, the expression levels of VZV glycoprotein E were higher in senescent cells, indicative of higher replication [168]. These findings highlight the complexity of considering senescence as an antiviral defense mechanism.
The first report to hint that HPV infection might be associated with senescence induction was provided in studies by Velasco et al., which described p53 upregulation in HPV-positive cells [169]. Subsequently, the Shay laboratory showed that HPV16 E6 and E7, through their modulation of the tumor suppressors p53 and pRb, are necessary for human diploid fibroblast cells to evade cellular senescence [170,171]. Interestingly, replicative senescence was reversed in cultures of human skin fibroblasts by the ability of E6 oncoprotein to inactivate p53 [172]. Accordingly, the hypothesis at the time was that E6 and E7 are key factors for premalignant replicatively exhausted cells to escape from senescence [173]. In later studies, the Howley laboratory demonstrated that the artificial expression of E2 in HPV-positive cancer cells results in a senescent growth arrest, accompanied by robust p21Cip1 upregulation, which can be reversed through the re-expression of exogenous E6 and E7 [174,175]. E2 blocks the transcription of E6 and E7 [176]. Importantly, these studies were performed in HeLa cells which are HPV positive and possibly addicted to E6 and E7 for their immortalization; thus, repression of E6 and E7 resulted in a senescent growth arrest [177,178,179]. As an alternative to using E2, these results were confirmed using shRNA-mediated knockdown of E6 and E7 expression, which also caused senescence-mediated growth arrest in HeLa cells [180]. Moreover, pharmacological inhibition of E6-mediated degradation of p53 results in a reduced proliferative capacity of HPV-positive cancer cells and senescence induction [181,182]. Lastly, CRISPR/Cas9-mediated knockout of E6 or E7 induced senescence in HPV 18-immortalized HeLa cells, marked by cellular hypertrophy, upregulation of SA-β-gal, and Lamin B1 degradation [183].
In marked contrast to the studies described above, transfection of naïve somatic cells (i.e., not previously infected with HPV) with the HPV oncoprotein E7 has been shown to induce senescence (e.g., transfecting WI-38 human fibroblasts with E7) [184]. Moreover, Rodier et al. have confirmed the use of E7 as a senescence inducer in HCA2 human fibroblasts, similar to Ras-induced senescence models, and showed that ectopic E7 expression results in significant SA-β-gal expression and formation of DNA-SCARS [18]. Moreover, cancer-associated fibroblasts (CAF) were found to secrete high levels of IL-6 upon STAT3 activation and exhibit a senescence morphology in vitro or in cervical cancer tissues infected with high-risk HPV, where the overexpression of E6 activates STAT3, increases IL-6 expression and induces senescence [185]. Importantly, it is well-known that clinically-isolated samples of the cervical epithelium (using Pap smears) have high levels of p16INK4a (and p14ARF) expression, suggesting that early during HPV infection, cervical epithelial cells are in a senescent state [186]. More specifically, Feng et al. investigated the protein expression levels of p14ARF, p15INK4b, p16INK4a, p53, and Ki-67 in a tissue microarray of 20 samples of CIN (II-III) [187] and reported a significant upregulation of the senescence-associated markers p14ARF, p15INK4b, p16INK4a in CIN compared to their normal cervical epithelial counterparts, suggesting that cervical premalignant lesions are highly enriched in senescent cells [187]. However, given that Feng et al. did not assess the HPV infection status in the CIN samples, it may prove to be the case that the majority were HPV positive, since HPV is the main oncogenic driver of cervical premalignant lesions. If this were, in fact, the case, accumulation of senescent cells in CIN lesions should provide an avenue whereby pharmacological therapy can be utilized to selectively target cervical senescent cells and prevent or delay malignant progression.
Collectively, our literature review suggests that, while most studies indicate that inactivation of the HPV oncogenic proteins E6 or E7 is the primary inducer of senescence, these studies were conducted predominantly in immortalized cells that had previously been infected by the HPV virus and were likely dependent on E6 or E7-mediated inhibition of p53 and Rb. On the other hand, it appears that when a naïve, virus-free cell is infected with HPV (of course, restricting this hypothesis to oncogenic variants 16 and 18), cells undergo senescence, which is likely to represent one form of OIS. This suggests that cells that have already been immortalized by HPV infection through the expression of E6 or E7 can still undergo senescence upon the suppression of these oncoproteins, and in addition, cells that have not been exposed to HPV can undergo senescence once infected due to the ability of E7 to induce OIS. Consequently, our hypothetical model suggests that cervical epithelial cells infected with HPV undergo one form of senescence in the process of developing clinically detectable premalignant lesions (CIN), or at least, senescence represents a component of those premalignant lesions. However, only those cells that manage to escape OIS or those under constant pro-tumorigenic stimulation by the SASP, are capable of progressing into malignant phenotypes (Figure 1).

4. Should Senolytics Be Considered for the Elimination of HPV-Infected Senescent Cells?

Senolytics are a diverse group of natural and synthetic compounds that have been found to selectively kill senescent cells. Initially, senolytics were identified through high-throughput drug screening designed to identify compounds that can eliminate senescent cells induced by replicative exhaustion [188]. Therefore, the translational goal for the development of these compounds was to cull senescent cells that accumulate during organismal aging to mitigate some of the senescence-associated aging-related pathologies [189]. Preclinical evidence has demonstrated the efficacy of senolytics in eliminating senescent cells in vitro, but more importantly, in eliminating senescent cells in animal models of atherosclerosis [190], osteoarthritis [191,192], neurodegeneration [193,194], neuropathy [195], fibrotic lung disease [196], and chronic kidney disease [197], among many others. Moreover, early evidence from clinical trials investigating some of these compounds in aging-related disorders has generated cautious optimism [198]. Of several senolytics being investigated clinically, the most successful ones that are currently being studied in the clinic include the dasatinib + quercetin cocktail [199,200], fisetin [NCT04815902, NCT04210986, NCT03325322], and inhibitors of members of the BCL-2 family, particularly, BCL-XL [NCT04229225, NCT04129944, NCT04537884, NCT04857996].
Senolytics have also been proposed as adjuvant cancer treatments [201], since therapy-induced senescence represents a major component of tumor biology and an established outcome of cancer therapeutics [68,202]; in this context, senolytics have shown significant efficacy in eliminating senescent tumor cells both in vivo and in vitro [71,106,203,204,205]. In addition to reducing tumor volume, senolytics have also been shown to reduce the ability of tumor cells to metastasize [106], interfere with therapy resistance [71,204,206], and alleviate some of the therapy-associated adverse effects of chemotherapy [207]. Thus, senolytics have a substantial utility in cancer therapy, despite several limitations including their heterogenous effect among different cancer models and some having significant toxicity when employed in vivo [208]. As such, efforts continue to identify more efficacious and safe compounds that exert senolytic potential and can be exploited for cancer treatment [204].
Despite having the promising potential to eliminate senescent cells induced by replicative exhaustion (RS) or exposure to various therapeutics (TIS), there is currently less evidence available relating to the ability of senolytics to eliminate oncogene-induced senescent cells (OIS) as a strategy for treating premalignant lesions where oncogene-induced senescent cells represent a major component [21]. As we propose in this work, HPV can also induce a form of OIS and has a role in the development of cervical premalignant lesions, and senolytics can also be investigated for the treatment of CIN and other premalignant lesions where HPV plays a pathogenetic role.
Several studies support the potential feasibility of senolytics to eliminate OIS. For example, the overexpression of H-Ras in WI-38 human fibroblasts renders these cells susceptible to killing in vitro by the BCL-2 inhibitor, navitoclax (ABT-263) [209]. In a similar fashion, navitoclax (ABT-263) was shown to eliminate senescent cells in a KIAA1549:BRAF fusion-driven, pilocytic astrocytoma DKFZ-BT66 cell model [210,211]. In addition to navitoclax, natural compounds such as piperlongumine [212] have been shown to kill oncogene-induced senescent cells; however, the frequently used dasatinib + quercetin cocktail has not proven to be similarly effective [211]. Ouabain, a cardiac glycoside, that has been shown to eliminate senescent cells induced by replicative exhaustion or therapy, was also reported to kill senescent cells induced by a transposon-mediated transfer of oncogenic N-Ras in vivo [213]. Moreover, ouabain was shown to exert comparable senolytic activity in a mouse model of adamantinomatous craniopharyngioma [214]. However, a profound limitation of ouabain as a senolytic stems from the fact that the investigated concentrations used to demonstrate its senolytic ability are supraclinical, but with the exception of a report by L’Hôte et al. where ouabain in the nanomolar concentration ranges eradicated oncogene-induced senescent cells [215].
To more directly interrogate the overall effect of the senolytic-mediated elimination of oncogene-induced senescent cells, Kolodkin-Gal et al. utilized a model of K-RAS- induced pancreatic adenocarcinoma, where K-Ras overexpression led to the formation of premalignant pancreatic lesions [12]. Treatment of mice harboring these pancreatic lesions with ABT-737, another BCL-2 inhibitor, and an established senolytic, resulted in a dramatic reduction in the burden of senescent cells in the pancreas and was accompanied by a decreased expression of several SASP factors [12]. Senolytic elimination of oncogene-induced senescent cells reduced the chance of the progression of pancreatic premalignant lesions into fully transformed pancreatic adenocarcinoma. [12]. Whether the elimination of HPV-induced premalignant cells in cervical or head and neck tissue would result in a similar outcome remains largely unknown.
There is no direct evidence for senolytics having been tested in HPV-induced senescence models; however, recent studies have indicated the potential utility of senolytics for the treatment of illnesses caused by other viral infections, including SARS.CoV.2 [216,217,218]. Preliminary data by Pham et al. indicated that Merkel cell polyomavirus is capable of inducing senescence in human skin fibroblasts coupled with a robust SASP; here the senolytic, navitoclax, decreased senescence and viral genome levels in these cells [219]. This led to the proposition of utilizing senolytics as antiviral therapy [220]. Moreover, a review of the recent literature has led Giannakoulis et al. to hypothesize that the use of senolytics might be beneficial in interfering with HBV and HCV oncogenic potential, since viral senescence appears to contribute to the development of hepatocellular carcinoma [221]. Furthermore, Szaniawski et al. have proposed the use of senolytics in HIV-1 persistence and HIV-1-associated immune exhaustion driven by the accumulation of senescent cells [222]. Lastly, the most direct evidence in support of this review is on the use of metformin, a senomorphic rather than a senolytic, wherein metformin blocked senescence induction in HPV-positive cancer cells in response to E6/E7 inhibition, allowing HPV-positive cancer cells to escape from therapy-induced senescence [223]. While this escape from senescence might not be a desirable outcome, it provides proof of concept that HPV-induced senescence is amenable to modulation by several of the currently available senolytics and senomorphics. Collectively, the evidence thus far available in the literature supports the need for further studies to test whether exploiting HPV-induced senescence as a target for established or novel senolytics might be valuable for developing pharmacological strategies for the prevention of virus-induced premalignant/malignant transformation.

Author Contributions

Conceptualization, T.S. and A.I.K.; resources, T.S and D.A.G.; writing—original draft preparation, T.S., A.I.K., N.H., J.A.-R., V.E. and A.M.E.; writing—review and editing, T.S. and D.A.G.; supervision, T.S. and D.A.G. All authors have read and agreed to the published version of the manuscript.

Funding

Tareq Saleh is supported for this work, in part, by the internal grant (465/83/2019) provided by the Deanship of Scientific Research, The Hashemite University. Research in David A. Gewirtz’s laboratory is supported by grant numbers CA268819 and CA239706 from the National Cancer Institute/National Institutes of Health and Grant # W81XWH 19-1-0490 from the Department of Defense Congressionally Directed Breast Cancer Research Program.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Hayflick, L.; Moorhead, P.S. The Serial Cultivation of Human Diploid Cell Strains. Exp. Cell Res. 1961, 25, 585–621. [Google Scholar] [CrossRef] [PubMed]
  2. Hayflick, L. The Limited in Vitro Lifetime of Human Diploid Cell Strains. Exp. Cell Res. 1965, 636, 614–636. [Google Scholar] [CrossRef] [PubMed]
  3. He, S.; Sharpless, N.E. Senescence in Health and Disease. Cell 2017, 169, 1000–1011. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Sharpless, N.E.; Sherr, C.J. Forging a Signature of in Vivo Senescence. Nat. Rev. Cancer 2015, 15, 397–408. [Google Scholar] [CrossRef]
  5. Harley, C.B.; Futcher, A.B.; Greider, C.W. Telomeres Shorten during Ageing of Human Fibroblasts. Nature 1990, 345, 458–460. [Google Scholar] [CrossRef]
  6. Poele, R.H.; Okorokov, A.L.; Jardine, L.; Cummings, J.; Joel, S.P.; te Poele, R.H.; Okorokov, A.L.; Jardine, L.; Cummings, J.; Joel, S.P. DNA Damage Is Able to Induce Senescence in Tumor Cells In Vitro and In Vivo. Cancer Res. 2002, 62, 1876–1883. [Google Scholar]
  7. Braig, M.; Schmitt, C.A. Oncogene-Induced Senescence: Putting the Brakes on Tumor Development. Cancer Res. 2006, 66, 2881–2884. [Google Scholar] [CrossRef] [Green Version]
  8. Campisi, J. Cellular Senescence as a Tumor-Suppressor Mechanism. Trends Cell Biol. 2001, 11, 27–31. [Google Scholar] [CrossRef] [Green Version]
  9. Collado, M.; Gil, J.; Efeyan, A.; Guerra, C.; Schuhmacher, A.J.; Barradas, M.; Benguría, A.; Zaballos, A.; Flores, J.M.; Barbacid, M.; et al. Tumour Biology: Senescence in Premalignant Tumours. Nature 2005, 436, 642. [Google Scholar] [CrossRef]
  10. Michaloglou, C.; Vredeveld, L.C.W.; Soengas, M.S.; Denoyelle, C.; Kuilman, T.; Van Der Horst, C.M.A.M.; Majoor, D.M.; Shay, J.W.; Mooi, W.J.; Peeper, D.S. BRAFE600-Associated Senescence-like Cell Cycle Arrest of Human Naevi. Nature 2005, 436, 720–724. [Google Scholar] [CrossRef] [Green Version]
  11. Courtois-Cox, S.; Jones, S.L.; Cichowski, K. Many Roads Lead to Oncogene-Induced Senescence. Oncogene 2008, 27, 2801–2809. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Kolodkin-Gal, D.; Roitman, L.; Ovadya, Y.; Azazmeh, N.; Assouline, B.; Schlesinger, Y.; Kalifa, R.; Horwitz, S.; Khalatnik, Y.; Hochner-Ger, A.; et al. Senolytic Elimination of Cox2-Expressing Senescent Cells Inhibits the Growth of Premalignant Pancreatic Lesions. Gut 2022, 71, 345–355. [Google Scholar] [CrossRef] [PubMed]
  13. Di Micco, R.; Fumagalli, M.; Cicalese, A.; Piccinin, S.; Gasparini, P.; Luise, C.; Schurra, C.; Garré, M.; Giovanni Nuciforo, P.; Bensimon, A.; et al. Oncogene-Induced Senescence Is a DNA Damage Response Triggered by DNA Hyper-Replication. Nature 2006, 444, 638–642. [Google Scholar] [CrossRef]
  14. Hernandez-Segura, A.; de Jong, T.V.; Melov, S.; Guryev, V.; Campisi, J.; Demaria, M. Unmasking Transcriptional Heterogeneity in Senescent Cells. Curr. Biol. 2017, 27, 2652–2660. [Google Scholar] [CrossRef] [Green Version]
  15. Faridi, R.; Zahra, A.; Khan, K.; Idrees, M. Oncogenic Potential of Human Papillomavirus (HPV) and Its Relation with Cervical Cancer. Virol. J. 2011, 8, 269. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Hoppe-seyler, K.; Bossler, F.; Braun, J.A.; Herrmann, A.L.; Hoppe-seyler, F. The HPV E6 / E7 Oncogenes: Key Factors for Viral Carcinogenesis and Therapeutic Targets. Trends Microbiol. 2018, 26, 158–168. [Google Scholar] [CrossRef]
  17. Burkhart, D.L.; Sage, J. Cellular Mechanisms of Tumour Suppression by the Retinoblastoma Gene. Nat. Rev. Cancer 2008, 8, 671–682. [Google Scholar] [CrossRef]
  18. Rodier, F.; Muñoz, D.P.; Teachenor, R.; Chu, V.; Le, O.; Bhaumik, D.; Coppé, J.P.; Campeau, E.; Beauséjour, C.M.; Kim, S.H.; et al. DNA-SCARS: Distinct Nuclear Structures That Sustain Damage-Induced Senescence Growth Arrest and Inflammatory Cytokine Secretion. J. Cell Sci. 2011, 124, 68–81. [Google Scholar] [CrossRef] [Green Version]
  19. Tomaić, V. Functional Roles of E6 and E7 Oncoproteins in HPV-Induced Malignancies at Diverse Anatomical Sites. Cancers 2016, 8, 95. [Google Scholar] [CrossRef] [Green Version]
  20. Fischer, M.; Uxa, S.; Stanko, C.; Magin, T.M.; Engeland, K. Human Papilloma Virus E7 Oncoprotein Abrogates the P53-P21-DREAM Pathway. Sci. Rep. 2017, 7, 2603. [Google Scholar] [CrossRef] [Green Version]
  21. Saleh, T.; Carpenter, V.J. Potential Use of Senolytics for Pharmacological Targeting of Precancerous Lesions. Mol. Pharmacol. 2021, 100, 580–587. [Google Scholar] [CrossRef] [PubMed]
  22. Gorgoulis, V.; Adams, P.D.; Alimonti, A.; Bennett, D.C.; Bischof, O.; Bishop, C.; Campisi, J.; Collado, M.; Evangelou, K.; Ferbeyre, G.; et al. Cellular Senescence: Defining a Path Forward. Cell 2019, 179, 813–827. [Google Scholar] [CrossRef] [PubMed]
  23. Galluzzi, L.; Vitale, I. Oncogene-Induced Senescence and Tumour Control in Complex Biological Systems. Cell Death Differ. 2018, 25, 1005–1006. [Google Scholar] [CrossRef]
  24. Zhang, R.; Poustovoitov, M.V.; Ye, X.; Santos, H.A.; Chen, W.; Daganzo, S.M.; Erzberger, J.P.; Serebriiskii, I.G.; Canutescu, A.A.; Dunbrack, R.L.; et al. Formation of MacroH2A-Containing Senescence-Associated Heterochromatin Foci and Senescence Driven by ASF1a and HIRA. Dev. Cell 2005, 8, 19–30. [Google Scholar] [CrossRef] [Green Version]
  25. Dimri, G.P.; Lee, X.; Basile, G.; Acosta, M.; Scott, G.; Roskelley, C.; Medrano, E.E.; Linskensi, 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] [Green Version]
  26. Kurz, D.J.D.J.; Decary, S.; Hong, Y.; Erusalimsky, J.D.J.D. Senescence-Associated (Beta)-Galactosidase Reflects an Increase in Lysosomal Mass during Replicative Ageing of Human Endothelial Cells. J. Cell Sci. 2000, 113, 3613–3622. [Google Scholar] [CrossRef] [PubMed]
  27. Nelson, G.; Kucheryavenko, O.; Wordsworth, J.; von Zglinicki, T. The Senescent Bystander Effect Is Caused by ROS-Activated NF-ΚB Signalling. Mech. Ageing Dev. 2018, 170, 30–36. [Google Scholar] [CrossRef]
  28. Rodier, F.; Coppé, J.; Patil, C.K.; Hoeijmakers, W.A.M.; Muñoz, 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]
  29. Coppé, J.-P.; Desprez, P.-Y.; Krtolica, A.; Campisi, J. The Senescence-Associated Secretory Phenotype: The Dark Side of Tumor Suppression. Annu. Rev. Pathol. 2010, 5, 99–118. [Google Scholar] [CrossRef] [Green Version]
  30. Coppé, J.P.; Patil, C.K.; Rodier, F.; Sun, Y.Y.; Muñoz, D.P.; Goldstein, J.; Nelson, P.S.; Desprez, P.-Y.Y.; Campisi, J.; Coppe, J.-P.; et al. Senescence-Associated Secretory Phenotypes Reveal Cell-Nonautonomous Functions of Oncogenic RAS and the P53 Tumor Suppressor. Aging Cell 2008, 6, 2853–2868. [Google Scholar] [CrossRef]
  31. González-Gualda, E.; Baker, A.G.; Fruk, L.; Muñoz-Espín, D. A Guide to Assessing Cellular Senescence In Vitro and In Vivo. FEBS J. 2020, 288, 56–80. [Google Scholar] [CrossRef] [PubMed]
  32. Yosef, R.; Pilpel, N.; Papismadov, N.; Gal, H.; Ovadya, Y.; Vadai, E.; Miller, S.; Porat, Z.; Ben-Dor, S.; Krizhanovsky, V. P21 Maintains Senescent Cell Viability under Persistent DNA Damage Response by Restraining JNK and Caspase Signaling. EMBO J. 2017, 36, 2280–2295. [Google Scholar] [CrossRef] [PubMed]
  33. Beausejour, C.M.; Krtolica, A.; Galimi, F.; Narita, M.; Lowe, S.W.; Yaswen, P.; Campisi, J. Reversal of Human Cellular Senescence: Roles of the P53 and P16 Pathways. Eur. Mol. Biol. Organ. J. 2003, 22, 4212–4222. [Google Scholar] [CrossRef] [PubMed]
  34. Lessard, F.; Igelmann, S.; Trahan, C.; Huot, G.; Saint-Germain, E.; Mignacca, L.; Del Toro, N.; Lopes-Paciencia, S.; Le Calvé, B.; Montero, M.; et al. Senescence-Associated Ribosome Biogenesis Defects Contributes to Cell Cycle Arrest through the Rb Pathway. Nat. Cell Biol. 2018, 20, 789–799. [Google Scholar] [CrossRef]
  35. Nishimura, K.; Kumazawa, T.; Kuroda, T.; Katagiri, N.; Tsuchiya, M.; Goto, N.; Furumai, R.; Murayama, A.; Yanagisawa, J.; Kimura, K. Perturbation of Ribosome Biogenesis Drives Cells into Senescence through 5S RNP-Mediated P53 Activation. Cell Rep. 2015, 10, 1310–1323. [Google Scholar] [CrossRef] [Green Version]
  36. Sikora, E.; Mosieniak, G.; Alicja Sliwinska, M. Morphological and Functional Characteristic of Senescent Cancer Cells. Curr. Drug Targets 2016, 17, 377–387. [Google Scholar] [CrossRef]
  37. Karlseder, J.; Smogorzewska, A.; De Lange, T. Senescence Induced by Altered Telomere State, Not Telomere Loss. Science 2002, 295, 2446–2449. [Google Scholar] [CrossRef] [Green Version]
  38. Höhn, A.; Weber, D.; Jung, T.; Ott, C.; Hugo, M.; Kochlik, B.; Kehm, R.; König, J.; Grune, T.; Castro, J.P. Happily (n)Ever after: Aging in the Context of Oxidative Stress, Proteostasis Loss and Cellular Senescence. Redox Biol. 2017, 11, 482–501. [Google Scholar] [CrossRef]
  39. Georgakopoulou, E.A.; Tsimaratou, K.; Evangelou, K.; Fernandez-Marcos, P.J.; Zoumpourlis, V.; Trougakos, I.P.; Kletsas, D.; Bartek, J.; Serrano, M.; Gorgoulis, V.G. Specific Lipofuscin Staining as a Novel Biomarker to Detect Replicative and Stress-Induced Senescence. A Method Applicable in Cryo-Preserved and Archival Tissues. Aging 2013, 5, 37–50. [Google Scholar] [CrossRef] [Green Version]
  40. Sitte, N.; Merker, K.; Grune, T.; Von Zglinicki, T. Lipofuscin Accumulation in Proliferating Fibroblasts in Vitro: An Indicator of Oxidative Stress. Exp. Gerontol. 2001, 36, 475–486. [Google Scholar] [CrossRef]
  41. Kaplon, J.; Zheng, L.; Meissl, K.; Chaneton, B.; Selivanov, V.A.; MacKay, G.; Van Der Burg, S.H.; Verdegaal, E.M.E.; Cascante, M.; Shlomi, T.; et al. A Key Role for Mitochondrial Gatekeeper Pyruvate Dehydrogenase in Oncogene-Induced Senescence. Nature 2013, 498, 109–112. [Google Scholar] [CrossRef] [PubMed]
  42. Zhao, H.; Darzynkiewicz, Z. Biomarkers of Cell Senescence Assessed by Imaging Cytometry. Methods Mol. Biol. 2013, 965, 83–92. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Zhang, R.; Adams, P.D. Heterochromatin and Its Relationship to Cell Senescence and Cancer Therapy. Cell Cycle 2007, 6, 784–789. [Google Scholar] [CrossRef] [PubMed]
  44. Basisty, N.; Kale, A.; Jeon, O.H.; Kuehnemann, C.; Payne, T.; Rao, C.; Holtz, A.; Shah, S.; Sharma, V.; Ferrucci, L.; et al. A Proteomic Atlas of Senescence-Associated Secretomes for Aging Biomarker Development. PLoS Biol. 2020, 18, e3000599. [Google Scholar] [CrossRef] [Green Version]
  45. Ortiz-Montero, P.; Londoño-Vallejo, A.; Vernot, J.P. Senescence-Associated IL-6 and IL-8 Cytokines Induce a Self- and Cross-Reinforced Senescence/Inflammatory Milieu Strengthening Tumorigenic Capabilities in the MCF-7 Breast Cancer Cell Line. Cell Commun. Signal. 2017, 15, 17. [Google Scholar] [CrossRef] [Green Version]
  46. Childs, B.G.; Durik, M.; Baker, D.J.; Deursen, J.M. Van Cellular Senescence in Aging and Age-Related Disease: From Mechanisms to Therapy. Nat. Med. 2016, 21, 1424–1435. [Google Scholar] [CrossRef] [Green Version]
  47. Chen, Q.M.; Bartholomew, J.C.; Campisi, J.; Acosta, M.; Reagan, J.D.; Ames, B.N. Molecular Analysis of H2O2-Induced Senescent-like Growth Arrest in Normal Human Fibroblasts: P53 and Rb Control G1 Arrest but Not Cell Replication. Biochem. J. 1998, 332, 43–50. [Google Scholar] [CrossRef] [Green Version]
  48. Alcorta, D.A.; Xiong, Y.; Phelps, D.; Hannon, G.; Beach, D.; Barrett, J.C. Involvement of the Cyclin-Dependent Kinase Inhibitor P16 (INK4a) in Replicative Senescence of Normal Human Fibroblasts. Proc. Natl. Acad. Sci. USA 1996, 93, 13742–13747. [Google Scholar] [CrossRef] [Green Version]
  49. Stein, G.H.; Drullinger, L.F.; Soulard, A.; Dulić, V. Differential Roles for Cyclin-Dependent Kinase Inhibitors P21 and P16 in the Mechanisms of Senescence and Differentiation in Human Fibroblasts. Mol. Cell. Biol. 1999, 19, 2109–2117. [Google Scholar] [CrossRef] [Green Version]
  50. Riley, T.; Sontag, E.; Chen, P.; Levine, A. Transcriptional Control of Human P53-Regulated Genes. Nat. Rev. Mol. Cell Biol. 2008, 9, 402–412. [Google Scholar] [CrossRef]
  51. Sullivan, K.D.; Galbraith, M.D.; Andrysik, Z.; Espinosa, J.M. Mechanisms of Transcriptional Regulation by P53. Cell Death Differ. 2018, 25, 133–143. [Google Scholar] [CrossRef] [PubMed]
  52. Caspari, T. Checkpoints: How to Activate P53. Curr. Biol. 2000, 10, R315–R317. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Goh, A.M.; Coffill, C.R.; Lane, D.P. The Role of Mutant P53 in Human Cancer. J. Pathol. 2011, 223, 116–126. [Google Scholar] [CrossRef] [PubMed]
  54. Ausserlechner, M.J.; Obexer, P.; Geley, S.; Kofler, R. G1 Arrest by P16INK4A Uncouples Growth from Cell Cycle Progression in Leukemia Cells with Deregulated Cyclin E and C-Myc Expression. Leukemia 2005, 19, 1051–1057. [Google Scholar] [CrossRef] [Green Version]
  55. Al-Khalaf, H.H.; Aboussekhra, A. P16 Controls P53 Protein Expression through Mir-Dependent Destabilization of MDM2. Mol. Cancer Res. 2018, 16, 1299–1308. [Google Scholar] [CrossRef] [Green Version]
  56. Shay, J.W.; Pereira-Smith, O.M.; Wright, W.E. A Role for Both RB and P53 in the Regulation of Human Cellular Senescence. Exp. Cell Res. 1991, 196, 33–39. [Google Scholar] [CrossRef]
  57. Benson, E.K.; Mungamuri, S.K.; Attie, O.; Kracikova, M.; Sachidanandam, R.; Manfredi, J.J.; Aaronson, S.A. P53-Dependent Gene Repression through P21 Is Mediated by Recruitment of E2F4 Repression Complexes. Oncogene 2014, 33, 3959–3969. [Google Scholar] [CrossRef] [Green Version]
  58. Pantoja, C.; Serrano, M. Murine Fibroblasts Lacking P21 Undergo Senescence and Are Resistant to Transformation by Oncogenic Ras. Oncogene 1999, 18, 4974–4982. [Google Scholar] [CrossRef] [Green Version]
  59. Lin, A.W.; Barradas, M.; Stone, J.C.; Aelst, L.V.; Serrano, M.; Lowe, S.W. Premature Senescence Involving P53 and P16 Is Activated in Response to Constitutive MEK/MAPK Mitogenic Signaling. Genes Dev. 1998, 12, 3008–3019. [Google Scholar] [CrossRef] [Green Version]
  60. 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] [Green Version]
  61. Liu, J.Y.; Souroullas, G.P.; Diekman, B.O.; Krishnamurthy, J.; Hall, B.M.; Sorrentino, J.A.; Parker, J.S.; Sessions, G.A.; Gudkov, A.V.; Sharpless, N.E. Cells Exhibiting Strong P16 INK4a Promoter Activation in Vivo Display Features of Senescence. Proc. Natl. Acad. Sci. USA 2019, 116, 2603–2611. [Google Scholar] [CrossRef] [PubMed]
  62. Romagosa, C.; Simonetti, S.; López-Vicente, L.; Mazo, A.; Lleonart, M.E.; Castellvi, J.; Ramon y Cajal, S. P16Ink4a Overexpression in Cancer: A Tumor Suppressor Gene Associated with Senescence and High-Grade Tumors. Oncogene 2011, 30, 2087–2097. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Liggett, W.H.; Sidransky, D. Role of the P16 Tumor Suppressor Gene in Cancer. J. Clin. Oncol. 1998, 16, 1197–1206. [Google Scholar] [CrossRef] [PubMed]
  64. Zhao, R.; Choi, B.Y.; Lee, M.H.; Bode, A.M.; Dong, Z. Implications of Genetic and Epigenetic Alterations of CDKN2A (P16INK4a) in Cancer. EBioMedicine 2016, 8, 30–39. [Google Scholar] [CrossRef] [Green Version]
  65. Garbuglia, A.R. Human Papillomavirus in Head and Neck Cancer. Cancers 2014, 6, 1705–1726. [Google Scholar] [CrossRef] [Green Version]
  66. Parry, D.; Bates, S.; Mann1, D.J.; Peters, G. Lack of Cyclin D-Cdk Complexes in Rb-Negative Cells Correlates with High Levels of P16INK4IMTS1 Tumour Suppressor Gene Product. EMBO J. 1995, 14, 503–511. [Google Scholar] [CrossRef]
  67. Seshadri, T.; Campisi, J. Growth-Factor-Inducible Gene Expression in Senescent Human Fibroblasts. Exp. Gerontol. 1989, 24, 515–522. [Google Scholar] [CrossRef]
  68. Saleh, T.; Bloukh, S.; Carpenter, V.J.; Alwohoush, E.; Bakeer, J.; Darwish, S.; Azab, B.; Gewirtz, D.A. Therapy-Induced Senescence: An “Old” Friend Becomes the Enemy. Cancers 2020, 12, 822. [Google Scholar] [CrossRef] [Green Version]
  69. Roberson, R.S.; Kussick, S.J.; Vallieres, E.; Chen, S.Y.J.; 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] [Green Version]
  70. Saleh, T.; Tyutyunyk-Massey, L.; Murray, G.F.; Alotaibi, M.R.; Kawale, A.S.; Elsayed, Z.; Henderson, S.C.; Yakovlev, V.; Elmore, L.W.; Toor, A.; et al. Tumor Cell Escape from Therapy-Induced Senescence. Biochem. Pharmacol. 2019, 162, 202–212. [Google Scholar] [CrossRef]
  71. Carpenter, V.; Saleh, T.; Min Lee, S.; Murray, G.; Reed, J.; Souers, A.; Faber, A.C.; Harada, H.; Gewirtz, D.A. Androgen-Deprivation Induced Senescence in Prostate Cancer Cells Is Permissive for the Development of Castration-Resistance but Susceptible to Senolytic Therapy. Biochem. Pharmacol. 2021, 193, 114765. [Google Scholar] [CrossRef] [PubMed]
  72. Ahmadinejad, F.; Bos, T.; Hu, B.; Britt, E.; Koblinski, J.; Souers, A.J.; Leverson, J.D.; Faber, A.C.; Gewirtz, D.A.; Harada, H. Senolytic-Mediated Elimination of Head and Neck Tumor Cells Induced Into Senescence by Cisplatin. Mol. Pharmacol. 2022, 101, 168–180. [Google Scholar] [CrossRef]
  73. Milanovic, M.; Fan, D.N.Y.; Belenki, D.; Däbritz, J.H.M.; Zhao, Z.; Yu, Y.; Dörr, 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] [PubMed] [Green Version]
  74. Dirac, A.M.G.; Bernards, R. Reversal of Senescence in Mouse Fibroblasts through Lentiviral Suppression of P53. J. Biol. Chem. 2003, 278, 11731–11734. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Carrire, C.; Gore, A.J.; Norris, A.M.; Gunn, J.R.; Young, A.L.; Longnecker, D.S.; Korc, M. Deletion of Rb Accelerates Pancreatic Carcinogenesis by Oncogenic Kras and Impairs Senescence in Premalignant Lesions. Gastroenterology 2011, 141, 1091–1101. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Seoane, M.; Iglesias, P.; Gonzalez, T.; Dominguez, F.; Fraga, M.; Aliste, C.; Forteza, J.; Costoya, J.A. Retinoblastoma Loss Modulates DNA Damage Response Favoring Tumor Progression. PLoS ONE 2008, 3, e3632. [Google Scholar] [CrossRef]
  77. Majumder, P.K.; Grisanzio, C.; O’Connell, F.; Barry, M.; Brito, J.M.; Xu, Q.; Guney, I.; Berger, R.; Herman, P.; Bikoff, R.; et al. A Prostatic Intraepithelial Neoplasia-Dependent P27 Kip1 Checkpoint Induces Senescence and Inhibits Cell Proliferation and Cancer Progression. Cancer Cell 2008, 14, 146–155. [Google Scholar] [CrossRef] [Green Version]
  78. Funayama, R.; Ishikawa, F. Cellular Senescence and Chromatin Structure. Chromosoma 2007, 116, 431–440. [Google Scholar] [CrossRef]
  79. Zhao, H.; Halicka, H.D.; Traganos, F.; Jorgensen, E.; Darzynkiewicz, Z. New Biomarkers Probing Depth of Cell Senescence Assessed by Laser Scanning Cytometry. Cytom. Part A 2010, 77, 999–1007. [Google Scholar] [CrossRef]
  80. Debacq-Chainiaux, F.; Erusalimsky, J.D.; Campisi, J.; Toussaint, O. Protocols to Detect Senescence-Associated Beta-Galactosidase (SA-Betagal) Activity, a Biomarker of Senescent Cells in Culture and In Vivo. Nat. Protoc. 2009, 4, 1798–1806. [Google Scholar] [CrossRef]
  81. Evangelou, K.; Lougiakis, N.; Rizou, S.V.; Kotsinas, A.; Kletsas, D.; Muñoz-Espín, D.; Kastrinakis, N.G.; Pouli, N.; Marakos, P.; Townsend, P.; et al. Robust, Universal Biomarker Assay to Detect Senescent Cells in Biological Specimens. Aging Cell 2017, 16, 192–197. [Google Scholar] [CrossRef] [PubMed]
  82. Nowotny, K.; Jung, T.; Grune, T.; Höhn, A. Accumulation of Modified Proteins and Aggregate Formation in Aging. Exp. Gerontol. 2014, 57, 122–131. [Google Scholar] [CrossRef] [PubMed]
  83. Ogrodnik, M.; Miwa, S.; Tchkonia, T.; Tiniakos, D.; Wilson, C.L.; Lahat, A.; Day, C.P.; Burt, A.; Palmer, A.; Anstee, Q.M.; et al. Cellular Senescence Drives Age-Dependent Hepatic Steatosis. Nat. Commun. 2017, 8, 15691. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Shay, J.W.; Wright, W.E. Senescence and Immortalization: Role of Telomeres and Telomerase. Carcinogenesis 2005, 26, 867–874. [Google Scholar] [CrossRef] [PubMed]
  85. De Lange, T. How Telomeres Solve the End-Protection Problem. Science 2009, 326, 948–952. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Herbig, U.; Jobling, W.A.; Chen, B.P.C.; Chen, D.J.; Sedivy, J.M. Telomere Shortening Triggers Senescence of Human Cells through a Pathway Involving ATM, P53, and P21CIP1, but Not P16INK4a. Mol. Cell 2004, 14, 501–513. [Google Scholar] [CrossRef] [PubMed]
  87. Fumagalli, M.; Rossiello, F.; Clerici, M.; Barozzi, S.; Cittaro, D.; Kaplunov, J.M.; Bucci, G.; Dobreva, M.; Matti, V.; Beausejour, C.M.; et al. Telomeric DNA Damage Is Irreparable and Causes Persistent DNA-Damage-Response Activation. Nat. Cell Biol. 2012, 14, 355–365. [Google Scholar] [CrossRef] [Green Version]
  88. d’Adda di Fagagna, F.; Reaper, P.M.; Clay-Farrace, L.; Fiegler, H.; Carr, P.; von Zglinicki, T.; Saretzki, G.; Carter, N.P.; Jackson, S.P. A DNA Damage Checkpoint Response in Telomere-Initiated Senescence. Nature 2003, 426, 194–198. [Google Scholar] [CrossRef]
  89. Bartkova, J.; Hořejší, Z.; Koed, K.; Krämer, A.; Tort, F.; Zieger, K.; Guldberg, P.; Sehested, M.; Nesland, J.M.; Lukas, C.; et al. DNA Damage Response as a Candidate Anti-Cancer Barrier in Early Human Tumorigenesis. Nature 2005, 434, 864–870. [Google Scholar] [CrossRef]
  90. Halazonetis, T.D.; Gorgoulis, V.G.; Bartek, J. An Oncogene-Induced DNA Damage Model for Cancer Development. Science 2008, 319, 1352–1355. [Google Scholar] [CrossRef] [Green Version]
  91. Malumbres, M.; Pérez De Castro, I.; Hernández, M.I.; Jiménez, M.; Corral, T.; Pellicer, A. Cellular Response to Oncogenic Ras Involves Induction of the Cdk4 and Cdk6 Inhibitor P15INK4b. Mol. Cell. Biol. 2000, 20, 2915–2925. [Google Scholar] [CrossRef] [PubMed]
  92. Bartkova, J.; Rezaei, N.; Liontos, M.; Karakaidos, P.; Kletsas, D.; Issaeva, N.; Vassiliou, L.V.F.; Kolettas, E.; Niforou, K.; Zoumpourlis, V.C.; et al. Oncogene-Induced Senescence Is Part of the Tumorigenesis Barrier Imposed by DNA Damage Checkpoints. Nature 2006, 444, 633–637. [Google Scholar] [CrossRef] [PubMed]
  93. Miwa, S.; Kashyap, S.; Chini, E.; von Zglinicki, T. Mitochondrial Dysfunction in Cell Senescence and Aging. J. Clin. Investig. 2022, 132, e158447. [Google Scholar] [CrossRef] [PubMed]
  94. 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] [Green Version]
  95. Moiseeva, O.; Bourdeau, V.; Roux, A.; Deschênes-Simard, X.; Ferbeyre, G. Mitochondrial Dysfunction Contributes to Oncogene-Induced Senescence. Mol. Cell. Biol. 2009, 29, 4495–4507. [Google Scholar] [CrossRef] [Green Version]
  96. Korolchuk, V.I.; Miwa, S.; Carroll, B.; von Zglinicki, T. Mitochondria in Cell Senescence: Is Mitophagy the Weakest Link? EBioMedicine 2017, 21, 7–13. [Google Scholar] [CrossRef] [Green Version]
  97. Nacarelli, T.; Lau, L.; Fukumoto, T.; Zundell, J.; Fatkhutdinov, N.; Wu, S.; Aird, K.M.; Iwasaki, O.; Kossenkov, A.V.; Schultz, D.; et al. NAD+ Metabolism Governs the Proinflammatory Senescence-Associated Secretome. Nat. Cell Biol. 2019, 21, 397–407. [Google Scholar] [CrossRef]
  98. Wiel, C.; Lallet-Daher, H.; Gitenay, D.; Gras, B.; Le Calvé, B.; Augert, A.; Ferrand, M.; Prevarskaya, N.; Simonnet, H.; Vindrieux, D.; et al. Endoplasmic Reticulum Calcium Release through ITPR2 Channels Leads to Mitochondrial Calcium Accumulation and Senescence. Nat. Commun. 2014, 5, 3792. [Google Scholar] [CrossRef] [Green Version]
  99. Lai, D.; Tan, C.L.; Gunaratne, J.; Quek, L.S.; Nei, W.; Thierry, F.; Bellanger, S. Localization of HPV-18 E2 at Mitochondrial Membranes Induces ROS Release and Modulates Host Cell Metabolism. PLoS ONE 2013, 8, e75625. [Google Scholar] [CrossRef] [Green Version]
  100. Zhu, H.; Blake, S.; Kusuma, F.K.; Pearson, R.B.; Kang, J.; Chan, K.T. Oncogene-Induced Senescence: From Biology to Therapy. Mech. Ageing Dev. 2020, 187, 111229. [Google Scholar] [CrossRef]
  101. Ye, X.; Zerlanko, B.; Zhang, R.; Somaiah, N.; Lipinski, M.; Salomoni, P.; Adams, P.D. Definition of PRB- and P53-Dependent and -Independent Steps in HIRA/ASF1a-Mediated Formation of Senescence-Associated Heterochromatin Foci. Mol. Cell. Biol. 2007, 27, 2452–2465. [Google Scholar] [CrossRef] [PubMed]
  102. Narita, M.; Nun, S.; Heard, E.; Narita, M.; Lin, A.W.; Hearn, S.A.; Spector, D.L.; Hannon, G.J.; Lowe, S.W. Rb-Mediated Heterochromatin Formation and Silencing of E2F Target Genes during Cellular Senescence. Cell 2003, 113, 703–716. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Nelyudova, A.; Aksenov, N.; Pospelov, V.; Pospelova, T. By Blocking Apoptosis, Bcl-2 in P38-Dependent Manner Promotes Cell Cycle Arrest and Accelerated Senescence after DNA Damage and Serum Withdrawal. Cell Cycle 2007, 6, 2171–2177. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Yosef, R.; Pilpel, N.; Tokarsky-amiel, R.; Biran, A.; Ovadya, Y.; Cohen, S.; Vadai, E.; Dassa, L.; Shahar, E.; Condiotti, R.; et al. Directed Elimination of Senescent Cells by Inhibition of BCL-W and BCL-XL. Nat. Commun. 2016, 7, 11190. [Google Scholar] [CrossRef]
  105. Sasaki, M.; Kumazaki, T.; Takano, H.; Nishiyama, M.; Mitsui, Y. Senescent Cells Are Resistant to Death despite Low Bcl-2 Level. Mech. Ageing Dev. 2001, 122, 1695–1706. [Google Scholar] [CrossRef] [PubMed]
  106. Troiani, M.; Colucci, M.; D’Ambrosio, M.; Guccini, I.; Pasquini, E.; Varesi, A.; Valdata, A.; Mosole, S.; Revandkar, A.; Attanasio, G.; et al. Single-Cell Transcriptomics Identifies Mcl-1 as a Target for Senolytic Therapy in Cancer. Nat. Commun. 2022, 13, 2177. [Google Scholar] [CrossRef]
  107. Marcotte, R.; Lacelle, C.; Wang, E. Senescent Fibroblasts Resist Apoptosis by Downregulating Caspase-3. Mech. Ageing Dev. 2004, 125, 777–783. [Google Scholar] [CrossRef]
  108. Bourgeois, B.; Madl, T. Regulation of Cellular Senescence via the FOXO4-P53 Axis. FEBS Lett. 2018, 592, 2083–2097. [Google Scholar] [CrossRef] [Green Version]
  109. De Keizer, P.L.J.; Packer, L.M.; Szypowska, A.A.; Riedl-Polderman, P.E.; Van Den Broek, N.J.F.; De Bruin, A.; Dansen, T.B.; Marais, R.; Brenkman, A.B.; Burgering, B.M.T. Activation of Forkhead Box O Transcription Factors by Oncogenic BRAF Promotes P21cip1-Dependent Senescence. Cancer Res. 2010, 70, 8526–8536. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  110. Baar, M.P.; Brandt, R.M.C.; Putavet, D.A.; Klein, J.D.D.; Derks, K.W.J.; Bourgeois, B.R.M.; Stryeck, S.; Rijksen, Y.; van Willigenburg, H.; Feijtel, D.A.; et al. Targeted Apoptosis of Senescent Cells Restores Tissue Homeostasis in Response to Chemotoxicity and Aging. Cell 2017, 169, 132–147. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  111. Le, H.H.; Cinaroglu, S.S.; Manalo, E.C.; Ors, A.; Gomes, M.M.; Duan Sahbaz, B.; Bonic, K.; Origel Marmolejo, C.A.; Quentel, A.; Plaut, J.S.; et al. Molecular Modelling of the FOXO4-TP53 Interaction to Design Senolytic Peptides for the Elimination of Senescent Cancer Cells. EBioMedicine 2021, 73, 103646. [Google Scholar] [CrossRef] [PubMed]
  112. Davalos, A.R.; Coppe, J.P.; Campisi, J.; Desprez, P.Y. Senescent Cells as a Source of Inflammatory Factors for Tumor Progression. Cancer Metastasis Rev. 2010, 29, 273–283. [Google Scholar] [CrossRef] [Green Version]
  113. Pereira, B.I.; Devine, O.P.; Vukmanovic-Stejic, M.; Chambers, E.S.; Subramanian, P.; Patel, N.; Virasami, A.; Sebire, N.J.; Kinsler, V.; Valdovinos, A.; et al. Senescent Cells Evade Immune Clearance via HLA-E-Mediated NK and CD8+ T Cell Inhibition. Nat. Commun. 2019, 10, 2387. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Muñoz, D.P.; Yannone, S.M.; Daemen, A.; Sun, Y.; Vakar-Lopez, F.; Kawahara, M.; Freund, A.M.; Rodier, F.; Wu, J.D.; Desprez, P.-Y.; et al. Targetable Mechanisms Driving Immunoevasion of Persistent Senescent Cells Link Chemotherapy-Resistant Cancer to Aging. JCI Insight 2019, 4, e124716. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Barakat, D.J.; Zhang, J.; Barberi, T.; Denmeade, S.R.; Friedman, A.D. CCAAT/Enhancer Binding Protein β Controls Androgen-Deprivation-Induced Senescence in Prostate Cancer Cells. Oncogene 2015, 34, 5912–5922. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Chien, Y.; Scuoppo, C.; Wang, X.; Fang, X.; Balgley, B.; Bolden, J.E.; Premsrirut, P.; Luo, W.; Chicas, A.; Lee, C.S.; et al. Control of the Senescence-Associated Secretory Phenotype by NF- k B Promotes Senescence and Enhances Chemosensitivity. Genes Dev. 2011, 25, 2125–2136. [Google Scholar] [CrossRef] [Green Version]
  117. Loo, T.M.; Miyata, K.; Tanaka, Y.; Takahashi, A. Cellular Senescence and Senescence-Associated Secretory Phenotype via the CGAS-STING Signaling Pathway in Cancer. Cancer Sci. 2020, 111, 304–311. [Google Scholar] [CrossRef] [Green Version]
  118. Xue, W.; Zender, L.; Miething, C.; Dickins, R.A.; Hernando, E.; Krizhanovsky, V.; Cordon-cardo, C.; Lowe, S.W. Senescence and Tumour Clearance Is Triggered by P53 Restoration in Murine Liver Carcinomas. Nature 2007, 445, 656–660. [Google Scholar] [CrossRef] [Green Version]
  119. Acosta, J.C.; Loghlen, A.O.; Banito, A.; Guijarro, M.V.; Augert, A.; Raguz, S.; Fumagalli, M.; Costa, M.D.; Brown, C.; Popov, N.; et al. Chemokine Signaling via the CXCR2 Receptor Reinforces Senescence. Cell 2008, 133, 1006–1018. [Google Scholar] [CrossRef] [Green Version]
  120. Kuilman, T.; Michaloglou, C.; Vredeveld, L.C.W.; Douma, S.; van Doorn, R.; Desmet, C.J.; Aarden, L.A.; Mooi, W.J.; Peeper, D.S. Oncogene-Induced Senescence Relayed by an Interleukin-Dependent Inflammatory Network. Cell 2008, 133, 1019–1031. [Google Scholar] [CrossRef] [Green Version]
  121. Ancrile, B.; Lim, K.H.; Counter, C.M. Oncogenic Ras-Induced Secretion of IL6 Is Required for Tumorigenesis. Genes Dev. 2007, 21, 1714–1719. [Google Scholar] [CrossRef] [PubMed]
  122. Sparmann, A.; Bar-Sagi, D. Ras-Induced Interleukin-8 Expression Plays a Critical Role in Tumor Growth and Angiogenesis. Cancer Cell 2004, 6, 447–458. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Acosta, J.C.; Banito, A.; Wuestefeld, T.; Georgilis, A.; Janich, P.; Morton, J.P.; Athineos, D.; Kang, T.; Lasitschka, F.; Andrulis, M.; et al. A Complex Secretory Program Orchestrated by the Inflammasome Controls Paracrine Senescence. Nat. Cell Biol. 2013, 15, 978–990. [Google Scholar] [CrossRef] [PubMed]
  124. Liu, D.; Hornsby, P.J. Senescent Human Fibroblasts Increase the Early Growth of Xenograft Tumors via Matrix Metalloproteinase Secretion. Cancer Res. 2007, 67, 3117–3127. [Google Scholar] [CrossRef] [Green Version]
  125. Liu, X.L.; Ding, J.; Meng, L.H. Oncogene-Induced Senescence: A Double Edged Sword in Cancer. Acta Pharmacol. Sin. 2018, 39, 1553–1558. [Google Scholar] [CrossRef]
  126. Sarkisian, C.J.; Keister, B.A.; Stairs, D.B.; Boxer, R.B.; Moody, S.E.; Chodosh, L.A. Dose-Dependent Oncogene-Induced Senescence in Vivo and Its Evasion during Mammary Tumorigenesis. Nat. Cell Biol. 2007, 9, 493–505. [Google Scholar] [CrossRef]
  127. Maldonado, J.L.; Timmerman, L.; Fridlyand, J.; Bastian, B.C. Mechanisms of Cell-Cycle Arrest in Spitz Nevi with Constitutive Activation of the MAP-Kinase Pathway. Am. J. Pathol. 2004, 164, 1783–1787. [Google Scholar] [CrossRef] [Green Version]
  128. Astle, M.V.; Hannan, K.M.; Ng, P.Y.; Lee, R.S.; George, A.J.; Hsu, A.K.; Haupt, Y.; Hannan, R.D.; Pearson, R.B. AKT Induces Senescence in Human Cells via MTORC1 and P53 in the Absence of DNA Damage: Implications for Targeting MTOR during Malignancy. Oncogene 2012, 31, 1949–1962. [Google Scholar] [CrossRef] [Green Version]
  129. Chen, Z.; Trotman, L.C.; Shaffer, D.; Lin, H.K.; Dotan, Z.A.; Niki, M.; Koutcher, J.A.; Scher, H.I.; Ludwig, T.; Gerald, W.; et al. Crucial Role of P53-Dependent Cellular Senescence in Suppression of Pten-Deficient Tumorigenesis. Nature 2005, 436, 725–730. [Google Scholar] [CrossRef] [Green Version]
  130. Natarajan, E.; Saeb, M.; Crum, C.P.; Woo, S.B.; McKee, P.H.; Rheinwald, J.G. Co-Expression of P16INK4A and Laminin 5 Γ2 by Microinvasive and Superficial Squamous Cell Carcinomas in Vivo and by Migrating Wound and Senescent Keratinocytes in Culture. Am. J. Pathol. 2003, 163, 477–491. [Google Scholar] [CrossRef]
  131. Tateishi, K.; Ohta, M.; Kanai, F.; Guleng, B.; Tanaka, Y.; Asaoka, Y.; Tada, M.; Seto, M.; Jazag, A.; Lianjie, L.; et al. Dysregulated Expression of Stem Cell Factor Bmi1 in Precancerous Lesions of the Gastrointestinal Tract. Clin. Cancer Res. 2006, 12, 6960–6966. [Google Scholar] [CrossRef]
  132. Miyasaka, Y.; Nagai, E.; Ohuchida, K.; Fujita, H.; Nakata, K.; Hayashi, A.; Mizumoto, K.; Tsuneyoshi, M.; Tanaka, M. Senescence in Intraductal Papillary Mucinous Neoplasm of the Pancreas. Hum. Pathol. 2011, 42, 2010–2017. [Google Scholar] [CrossRef] [PubMed]
  133. Gray-Schopfer, V.C.; Cheong, S.C.; Chong, H.; Chow, J.; Moss, T.; Abdel-Malek, Z.A.; Marais, R.; Wynford-Thomas, D.; Bennett, D.C. Cellular Senescence in Naevi and Immortalisation in Melanoma: A Role for P16? Br. J. Cancer 2006, 95, 496–505. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Bascones-Martínez, A.; López-Durán, M.; Cano-Sánchez, J.; Sánchez-Verde, L.; Díez-Rodríguez, A.; Aguirre-Echebarría, P.; Alvarez-Fernández, E.; González-Moles, M.A.; Bascones-Ilundain, J.; Muzio, L.L.; et al. Differences in the Expression of Five Senescence Markers in Oral Cancer, Oral Leukoplakia and Control Samples in Humans. Oncol. Lett. 2012, 3, 1319–1325. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Tsang, C.M.; Yip, Y.L.; Lo, K.W.; Deng, W.; To, K.F.; Hau, P.M.; Lau, V.M.Y.; Takada, K.; Lui, V.W.Y.; Lung, M.L.; et al. Cyclin D1 Overexpression Supports Stable EBV Infection in Nasopharyngeal Epithelial Cells. Proc. Natl. Acad. Sci. USA 2012, 109, E3473–E3482. [Google Scholar] [CrossRef] [Green Version]
  136. Song, P.; An, J.; Zou, M.-H. Immune Clearance of Senescent Cells to Combat Ageing and Chronic Diseases. Cells 2020, 9, 671. [Google Scholar] [CrossRef] [Green Version]
  137. Kale, A.; Sharma, A.; Stolzing, A.; Stolzing, A.; Desprez, P.Y.; Desprez, P.Y.; Campisi, J.; Campisi, J. Role of Immune Cells in the Removal of Deleterious Senescent Cells. Immun. Ageing 2020, 17, 16. [Google Scholar] [CrossRef]
  138. 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]
  139. Greenman, C.; Stephens, P.; Smith, R.; Dalgliesh, G.L.; Hunter, C.; Bignell, G.; Davies, H.; Teague, J.; Butler, A.; Stevens, C.; et al. Patterns of Somatic Mutation in Human Cancer Genomes. Nature 2007, 446, 153–158. [Google Scholar] [CrossRef] [Green Version]
  140. Tort, F.; Bartkova, J.; Sehested, M.; Ørntoft, T.; Lukas, J.; Bartek, J. Retinoblastoma Pathway Defects Show Differential Ability to Activate the Constitutive DNA Damage Response in Human Tumorigenesis. Cancer Res. 2006, 66, 10258–10263. [Google Scholar] [CrossRef] [Green Version]
  141. Dankort, D.; Filenova, E.; Collado, M.; Serrano, M.; Jones, K.; McMahon, M. A New Mouse Model to Explore the Initiation, Progression, and Therapy of BRAFV600E-Induced Lung Tumors. Genes Dev. 2007, 21, 379–384. [Google Scholar] [CrossRef] [PubMed]
  142. Miyauchi, H.; Minamino, T.; Tateno, K.; Kunieda, T.; Toko, H.; Komuro, I. Akt Negatively Regulates the in Vitro Lifespan of Human Endothelial Cells via a P53/P21-Dependent Pathway. EMBO J. 2004, 23, 212–220. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Dimri, G.P.; Itahana, K.; Acosta, M.; Campisi, J. Regulation of a Senescence Checkpoint Response by the E2F1 Transcription Factor and P14ARF Tumor Suppressor. Mol. Cell. Biol. 2000, 20, 273–285. [Google Scholar] [CrossRef] [Green Version]
  144. Schosserer, M.; Grillari, J.; Breitenbach, M. The Dual Role of Cellular Senescence in Developing Tumors and Their Response to Cancer Therapy. Front. Oncol. 2017, 7, 278. [Google Scholar] [CrossRef] [Green Version]
  145. Alimirah, F.; Pulido, T.; Valdovinos, A.; Alptekin, S.; Chang, E.; Jones, E.; Diaz, D.A.; Flores, J.; Velarde, M.C.; Demaria, M.; et al. Cellular Senescence Promotes Skin Carcinogenesis through P38MAPK and P44/42MAPK Signaling. Cancer Res. 2020, 80, 3606–3619. [Google Scholar] [CrossRef] [PubMed]
  146. Cahu, J.; Bustany, S.; Sola, B. Senescence-Associated Secretory Phenotype Favors the Emergence of Cancer Stem-like Cells. Cell Death Dis. 2012, 3, e446–e448. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  147. Laberge, R.; Sun, Y.; Orjalo, A.V.; Patil, C.K.; Freund, A.; Zhou, L.; Curran, S.C.; Davalos, A.R.; Wilson-edell, K.A.; Liu, S.; et al. MTOR Regulates the Pro-Tumorigenic Senescence-Associated Secretory Phenotype by Promoting IL1A Translation. Nat. Cell Biol. 2015, 17, 1049–1061. [Google Scholar] [CrossRef]
  148. Eggert, T.; Wolter, K.; Ji, J.; Ma, C.; Yevsa, T.; Klotz, S.; Medina-Echeverz, J.; Longerich, T.; Forgues, M.; Reisinger, F.; et al. Distinct Functions of Senescence-Associated Immune Responses in Liver Tumor Surveillance and Tumor Progression. Cancer Cell 2016, 30, 533–547. [Google Scholar] [CrossRef] [Green Version]
  149. Bent, E.H.; Gilbert, L.A.; Hemann, M.T. A Senescence Secretory Switch Mediated by PI3K/AKT/MTOR Activation Controls Chemoprotective Endothelial Secretory Responses. Genes Dev. 2016, 30, 1811–1821. [Google Scholar] [CrossRef] [Green Version]
  150. Hubackova, S.; Krejcikova, K.; Bartek, J.; Hodny, Z. IL1- and TGFβ-Nox4 Signaling, Oxidative Stress and DNA Damage Response Are Shared Features of Replicative, Oncogene-Induced, and Drug-Induced Paracrine ‘Bystander Senescence’. Aging 2012, 4, 932–951. [Google Scholar] [CrossRef]
  151. Shannon, C.L.; Klausner, J.D. The Growing Epidemic of Sexually Transmitted Infections in Adolescents: A Neglected Population. Curr. Opin. Pediatr. 2019, 30, 137–143. [Google Scholar] [CrossRef] [PubMed]
  152. Clifford, G.M.; Tully, S.; Franceschi, S. Carcinogenicity of Human Papillomavirus (HPV) Types in HIV-Positive Women: A Meta-Analysis from HPV Infection to Cervical Cancer. Clin. Infect. Dis. 2017, 64, 1228–1235. [Google Scholar] [CrossRef] [PubMed]
  153. de Martel, C.; Plummer, M.; Vignat, J.; Franceschi, S. Worldwide Burden of Cancer Attributable to HPV by Site, Country and HPV Type. Int. J. Cancer 2017, 141, 664–670. [Google Scholar] [CrossRef] [Green Version]
  154. Wang, J.; Tang, D.; Wang, K.; Wang, J.; Zhang, Z.; Chen, Y.; Zhang, X.; Ma, C. HPV Genotype Prevalence and Distribution during 2009-2018 in Xinjiang, China: Baseline Surveys Prior to Mass HPV Vaccination. BMC Womens Health 2019, 19, 90. [Google Scholar] [CrossRef] [Green Version]
  155. Delany-Moretlwe, S.; Kelley, K.F.; James, S.; Scorgie, F.; Subedar, H.; Dlamini, N.R.; Pillay, Y.; Naidoo, N.; Chikandiwa, A.; Rees, H. Human Papillomavirus Vaccine Introduction in South Africa: Implementation Lessons from an Evaluation of the National School-Based Vaccination Campaign. Glob. Health Sci. Pract. 2018, 6, 428–438. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Trus, B.L.; Roden, R.B.S.; Greenstone, H.L.; Vrhel, M.; Schiller, J.T.; Booy, F.P. Novel Structural Features of Bovine Papillomavirus Capsid Revealed by a Three-Dimensional Reconstruction to 9 A Resolution. Nat. Struct. Biol. 1997, 4, 413–420. [Google Scholar] [CrossRef] [PubMed]
  157. DiGiuseppe, S.; Bienkowska-Haba, M.; Guion, L.G.M.; Keiffer, T.R.; Sapp, M. Human Papillomavirus Major Capsid Protein L1 Remains Associated with the Incoming Viral Genome throughout the Entry Process. J. Virol. 2017, 91, e00537-17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  158. Godi, A.; Bissett, S.L.; Masloh, S.; Fleury, M.; Li, S.; Zhao, Q.; Xia, N.; Cocuzza, C.E.; Beddows, S. Impact of Naturally Occurring Variation in the Human Papillomavirus 52 Capsid Proteins on Recognition by Type-Specific Neutralising Antibodies. J. Gen. Virol. 2019, 100, 237–245. [Google Scholar] [CrossRef]
  159. Pouyanfard, S.; Spagnoli, G.; Bulli, L.; Balz, K.; Yang, F.; Odenwald, C.; Seitz, H.; Mariz, F.C.; Bolchi, A.; Ottonello, S.; et al. Minor Capsid Protein L2 Polytope Induces Broad Protection against Oncogenic and Mucosal Human Papillomaviruses. J. Virol. 2018, 92, e01930-17. [Google Scholar] [CrossRef] [Green Version]
  160. Castro-Muñoz, L.J.; Manzo-Merino, J.; Muñoz-Bello, J.O.; Olmedo-Nieva, L.; Cedro-Tanda, A.; Alfaro-Ruiz, L.A.; Hidalgo-Miranda, A.; Madrid-Marina, V.; Lizano, M. The Human Papillomavirus (HPV) E1 Protein Regulates the Expression of Cellular Genes Involved in Immune Response. Sci. Rep. 2019, 9, 13620. [Google Scholar] [CrossRef] [Green Version]
  161. Graham, S.V. Human Papillomavirus E2 Protein: Linking Replication, Transcription, and RNA Processing. J. Virol. 2016, 90, 8384–8388. [Google Scholar] [CrossRef]
  162. Chojnacki, M.; Melendy, T. The HPV E2 Transcriptional Transactivation Protein Stimulates Cellular DNA Polymerase Epsilon. Viruses 2018, 10, 321. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Knight, G.L.; Pugh, A.G.; Yates, E.; Bell, I.; Wilson, R.; Moody, C.A.; Laimins, L.A.; Roberts, S. A Cyclin-Binding Motif in Human Papillomavirus Type 18 (HPV18) E1^E4 Is Necessary for Association with CDK-Cyclin Complexes and G2/M Cell Cycle Arrest of Keratinocytes, but Is Not Required for Differentiation-Dependent Viral Genome Amplification or L1 Cap. Virology 2011, 412, 196–210. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. Ren, S.; Gaykalova, D.A.; Guo, T.; Favorov, A.V.; Fertig, E.J.; Tamayo, P.; Callejas-Valera, J.L.; Allevato, M.; Gilardi, M.; Santos, J.; et al. HPV E2, E4, E5 Drive Alternative Carcinogenic Pathways in HPV Positive Cancers. Oncogene 2020, 39, 6327–6339. [Google Scholar] [CrossRef] [PubMed]
  165. Bryan, J.T.; Han, A.; Fife, K.H.; Brown, D.R. The Human Papillomavirus Type 11 E1E4 Protein Is Phosphorylated in Genital Epithelium. Virology 2000, 268, 430–439. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  166. Ilahi, N.E.; Bhatti, A. Impact of HPV E5 on Viral Life Cycle via EGFR Signaling. Microb. Pathog. 2020, 139, 103923. [Google Scholar] [CrossRef]
  167. Baz-Martínez, M.; Da Silva-Álvarez, S.; Rodríguez, E.; Guerra, J.; El Motiam, A.; Vidal, A.; Garciá-Caballero, T.; González-Barcia, M.; Sánchez, L.; Munõz-Fontela, C.; et al. Cell Senescence Is an Antiviral Defense Mechanism. Sci. Rep. 2016, 6, 37007. [Google Scholar] [CrossRef] [Green Version]
  168. Kim, J.A.; Seong, R.K.; Shin, O.S. Enhanced Viral Replication by Cellular Replicative Senescence. Immune Netw. 2016, 16, 286–295. [Google Scholar] [CrossRef] [Green Version]
  169. Velasco, J.A.; Stevens, C.W.; Esteban, J.M.; Ruthsatz, M.K.J.; Ramsamooj, P.; Dritschilo, A.; Notario, V. Modulation of Proliferation and Tumorigenic Potential of Cervical-Carcinoma Cells by the Expression of Sense and Antisense P53. Int. J. Oncol. 1995, 7, 883–888. [Google Scholar] [CrossRef]
  170. Holt, S.; Gollahon, L.; Willingham, T.; Barbosa, M.; Shay, J. P53 Levels in Human Mammary Epithelial Cells Expressing Wild-Type and Mutant Human Papillomavirus Type 16 (HPV-16) E6 Proteins. Int. J. Oncol. 1996, 8, 263–270. [Google Scholar] [CrossRef]
  171. Litaker, J.R.; Pan, J.; Cheung, Y.C.; Zhang, D.K.; Liu, Y.; Wong, S.C.H.; Wan, T.S.K.; Tsao, S.W. Expression Profile of Senescence-Associated Beta-Galactosidase and Activation of Telomerase in Human Ovarian Surface Epithelial Cells Undergoing Immortalization. Int. J. Oncol. 1998, 13, 951–956. [Google Scholar] [CrossRef] [PubMed]
  172. Filatov, L.; Golubovskaya, V.; Hurt, J.C.; Byrd, L.L.; Phillips, J.M.; Kaufmann, W.K. Chromosomal Instability Is Correlated with Telomere Erosion and Inactivation of G2 Checkpoint Function in Human Fibroblasts Expressing Human Papillomavirus Type 16 E6 Oncoprotein. Oncogene 1998, 16, 1825–1838. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  173. Helt, A.-M.; Funk, J.O.; Galloway, D.A. Inactivation of Both the Retinoblastoma Tumor Suppressor and P21 by the Human Papillomavirus Type 16 E7 Oncoprotein Is Necessary to Inhibit Cell Cycle Arrest in Human Epithelial Cells. J. Virol. 2002, 76, 10559–10568. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  174. Wells, S.I. Papillomavirus E2 Induces Senescence in HPV-Positive Cells via PRB- and P21CIP-Dependent Pathways. EMBO J. 2000, 19, 5762–5771. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  175. Wells, S.I.; Aronow, B.J.; Wise, T.M.; Williams, S.S.; Couget, J.A.; Howley, P.M. Transcriptome Signature of Irreversible Senescence in Human Papillomavirus-Positive Cervical Cancer Cells. Proc. Natl. Acad. Sci. USA 2003, 100, 7093–7098. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  176. Thierry, F.; Howley, P.M. Functional Analysis of E2-Mediated Repression of the HPV18 P105 Promoter. New Biol. 1991, 3, 90–100. [Google Scholar]
  177. Goodwin, E.C.; DiMaio, D. Induced Senescence in HeLa Cervical Carcinoma Cells Containing Elevated Telomerase Activity and Extended Telomeres. Cell Growth Differ. 2001, 12, 525–534. [Google Scholar]
  178. Lee, C.J.; Suh, E.J.; Kang, H.T.; Im, J.S.; Um, S.J.; Park, J.S.; Hwang, E.S. Induction of Senescence-like State and Suppression of Telomerase Activity through Inhibition of HPV E6/E7 Gene Expression in Cells Immortalized by HPV16 DNA. Exp. Cell Res. 2002, 277, 173–182. [Google Scholar] [CrossRef]
  179. DeFilippis, R.A.; Goodwin, E.C.; Wu, L.; DiMaio, D. Endogenous Human Papillomavirus E6 and E7 Proteins Differentially Regulate Proliferation, Senescence, and Apoptosis in HeLa Cervical Carcinoma Cells. J. Virol. 2003, 77, 1551–1563. [Google Scholar] [CrossRef] [Green Version]
  180. Gu, W.; Putral, L.; Hengst, K.; Minto, K.; Saunders, N.A.; Leggatt, G.; McMillan, N.A.J. Inhibition of Cervical Cancer Cell Growth in Vitro and in Vivo with Lentiviral-Vector Delivered Short Hairpin RNA Targeting Human Papillomavirus E6 and E7 Oncogenes. Cancer Gene Ther. 2006, 13, 1023–1032. [Google Scholar] [CrossRef]
  181. Celegato, M.; Messa, L.; Goracci, L.; Mercorelli, B.; Bertagnin, C.; Spyrakis, F.; Suarez, I.; Cousido-Siah, A.; Travé, G.; Banks, L.; et al. A Novel Small-Molecule Inhibitor of the Human Papillomavirus E6-P53 Interaction That Reactivates P53 Function and Blocks Cancer Cells Growth. Cancer Lett. 2020, 470, 115–125. [Google Scholar] [CrossRef] [PubMed]
  182. Braun, J.A.; Herrmann, A.L.; Blase, J.I.; Frensemeier, K.; Bulkescher, J.; Scheffner, M.; Galy, B.; Hoppe-Seyler, K.; Hoppe-Seyler, F. Effects of the Antifungal Agent Ciclopirox in HPV-Positive Cancer Cells: Repression of Viral E6/E7 Oncogene Expression and Induction of Senescence and Apoptosis. Int. J. Cancer 2020, 146, 461–474. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  183. Inturi, R.; Jemth, P. CRISPR/Cas9-Based Inactivation of Human Papillomavirus Oncogenes E6 or E7 Induces Senescence in Cervical Cancer Cells. Virology 2021, 562, 92–102. [Google Scholar] [CrossRef] [PubMed]
  184. Itahana, K.; Zou, Y.; Itahana, Y.; Martinez, J.-L.; Beausejour, C.; Jacobs, J.J.L.; van Lohuizen, M.; Band, V.; Campisi, J.; Dimri, G.P. Control of the Replicative Life Span of Human Fibroblasts by P16 and the Polycomb Protein Bmi-1. Mol. Cell. Biol. 2003, 23, 389–401. [Google Scholar] [CrossRef] [Green Version]
  185. Ren, C.; Cheng, X.; Lu, B.; Yang, G. Activation of Interleukin-6/Signal Transducer and Activator of Transcription 3 by Human Papillomavirus Early Proteins 6 Induces Fibroblast Senescence to Promote Cervical Tumourigenesis through Autocrine and Paracrine Pathways in Tumour Microenvironment. Eur. J. Cancer 2013, 49, 3889–3899. [Google Scholar] [CrossRef]
  186. Von Keyserling, H.; Kühn, W.; Schneider, A.; Bergmann, T.; Kaufmann, A.M. P16INK4a and P14ARF MRNA Expression in Pap Smears Is Age-Related. Mod. Pathol. 2012, 25, 465–470. [Google Scholar] [CrossRef] [Green Version]
  187. Feng, W.; Xiao, J.; Zhang, Z.; Rosen, D.G.; Brown, R.E.; Liu, J.; Duan, X. Senescence and Apoptosis in Carcinogenesis of Cervical Squamous Carcinoma. Mod. Pathol. 2007, 20, 961–966. [Google Scholar] [CrossRef] [Green Version]
  188. Zhu, Y.; Tchkonia, T.; Pirtskhalava, T.; Gower, A.C.; Ding, H.; Giorgadze, N.; Palmer, A.K.; Ikeno, Y.; Hubbard, G.B.; Hara, S.P.O.; et al. The Achilles’ Heel of Senescent Cells: From Transcriptome to Senolytic Drugs. Aging Cell 2015, 14, 644–658. [Google Scholar] [CrossRef]
  189. van Deursen, J.M. Senolytic Therapies for Healthy Longevity. Science 2019, 364, 636–637. [Google Scholar] [CrossRef]
  190. Roos, C.M.; Zhang, B.; Palmer, A.K.; Ogrodnik, M.B.; Pirtskhalava, T.; Thalji, N.M.; Hagler, M.; Jurk, D.; Smith, L.A.; Casaclang-Verzosa, G.; et al. Chronic Senolytic Treatment Alleviates Established Vasomotor Dysfunction in Aged or Atherosclerotic Mice. Aging Cell 2016, 15, 973–977. [Google Scholar] [CrossRef]
  191. Jeon, O.H.; Kim, C.; Laberge, R.-M.; Demaria, M.; Rathod, S.; Vasserot, A.P.; Chung, J.W.; Kim, D.H.; Poon, Y.; David, N.; et al. Local Clearance of Senescent Cells Attenuates the Development of Post-Traumatic Osteoarthritis and Creates a pro-Regenerative Environment. Nat. Med. 2017, 23, 712–781. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  192. Nogueira-Recalde, U.; Lorenzo-Gómez, I.; Blanco, F.J.; Loza, M.I.; Grassi, D.; Shirinsky, V.; Shirinsky, I.; Lotz, M.; Robbins, P.D.; Domínguez, E.; et al. Fibrates as Drugs with Senolytic and Autophagic Activity for Osteoarthritis Therapy. EBioMedicine 2019, 45, 588–605. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  193. Bussian, T.J.; Aziz, A.; Meyer, C.F.; Swenson, B.L.; van Deursen, J.M.; Baker, D.J. Clearance of Senescent Glial Cells Prevents Tau-Dependent Pathology and Cognitive Decline. Nature 2018, 562, 578–582. [Google Scholar] [CrossRef] [PubMed]
  194. Zhang, P.; Kishimoto, Y.; Grammatikakis, I.; Gottimukkala, K.; Cutler, R.G.; Zhang, S.; Abdelmohsen, K.; Bohr, V.A.; Sen, J.M.; Gorospe, M.; et al. Senolytic Therapy Alleviates Aβ-Associated Oligodendrocyte Progenitor Cell Senescence and Cognitive Deficits in an Alzheimer’s Disease Model. Nat. Neurosci. 2019, 22, 719–728. [Google Scholar] [CrossRef] [PubMed]
  195. Acklin, S.; Zhang, M.; Du, W.; Zhao, X.; Plotkin, M.; Chang, J.; Campisi, J.; Zhou, D.; Xia, F. Depletion of Senescent-like Neuronal Cells Alleviates Cisplatin-Induced Peripheral Neuropathy in Mice. Sci. Rep. 2020, 10, 14170. [Google Scholar] [CrossRef]
  196. Pan, J.; Li, D.; Xu, Y.; Zhang, J.; Wang, Y.; Chen, M.; Lin, S.; Huang, L.; Chung, E.J.; Citrin, D.E.; et al. Inhibition of Bcl-2/Xl With ABT-263 Selectively Kills Senescent Type II Pneumocytes and Reverses Persistent Pulmonary Fibrosis Induced by Ionizing Radiation in Mice. Int. J. Radiat. Oncol. Biol. Phys. 2017, 99, 353–361. [Google Scholar] [CrossRef] [PubMed]
  197. Palmer, A.K.; Xu, M.; Zhu, Y.; Pirtskhalava, T.; Weivoda, M.M.; Hachfeld, C.M.; Prata, L.G.; van Dijk, T.H.; Verkade, E.; Casaclang-Verzosa, G.; et al. Targeting Senescent Cells Alleviates Obesity-Induced Metabolic Dysfunction. Aging Cell 2019, 18, e12950. [Google Scholar] [CrossRef]
  198. Chaib, S.; Tchkonia, T.; Kirkland, J.L. Cellular Senescence and Senolytics: The Path to the Clinic. Nat. Med. 2022, 28, 1556–1568. [Google Scholar] [CrossRef]
  199. Justice, J.N.; Nambiar, A.M.; Tchkonia, T.; LeBrasseur, N.K.; Pascual, R.; Hashmi, S.K.; Prata, L.; Masternak, M.M.; Kritchevsky, S.B.; Musi, N.; et al. Senolytics in Idiopathic Pulmonary Fibrosis: Results from a First-in-Human, Open-Label, Pilot Study. EBioMedicine 2019, 40, 554–563. [Google Scholar] [CrossRef] [Green Version]
  200. Hickson, L.T.J.; Langhi Prata, L.G.P.; Bobart, S.A.; Evans, T.K.; Giorgadze, N.; Hashmi, S.K.; Herrmann, S.M.; Jensen, M.D.; Jia, Q.; Jordan, K.L.; et al. Corrigendum to “Senolytics Decrease Senescent Cells in Humans: Preliminary Report from a Clinical Trial of Dasatinib plus Quercetin in Individuals with Diabetic Kidney Disease” EBioMedicine 47 (2019) 446-456. EBioMedicine 2020, 52, 102595. [Google Scholar] [CrossRef]
  201. Short, S.; Fielder, E.; Miwa, S.; von Zglinicki, T. Senolytics and Senostatics as Adjuvant Tumour Therapy. EBioMedicine 2019, 41, 683–692. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  202. Kahlem, P.; Dörken, B.; Schmitt, C.A.; Kahlem, P.; Dörken, B.; Schmitt, C.A. Cellular Senescence in Cancer Treatment: Friend or Foe ? J. Clin. Investig. 2004, 113, 169–174. [Google Scholar] [CrossRef] [Green Version]
  203. Saleh, T.; Carpenter, V.; Tyutyunyk-Massey, L.; Murray, G.; Leverson, J.; Souers, A.; Alotaibi, M.; Faber, A.; Reed, J.; Harada, H.; et al. Clearance of Therapy-induced Senescent Tumor Cells by the Senolytic ABT-263 via Interference with BCL-XL-BAX Interaction. Mol. Oncol. 2020, 14, 2504–2519. [Google Scholar] [CrossRef] [PubMed]
  204. Shahbandi, A.; Rao, S.G.; Anderson, A.Y.; Frey, W.D.; Olayiwola, J.O.; Ungerleider, N.A.; Jackson, J.G. BH3 Mimetics Selectively Eliminate Chemotherapy-Induced Senescent Cells and Improve Response in TP53 Wild-Type Breast Cancer. Cell Death Differ. 2020, 27, 3097–3116. [Google Scholar] [CrossRef] [PubMed]
  205. Wang, L.; Lankhorst, L.; Bernards, R. Exploiting Senescence for the Treatment of Cancer. Nat. Rev. Cancer 2022, 22, 340–355. [Google Scholar] [CrossRef]
  206. 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] [Green Version]
  207. Demaria, M.; Leary, M.N.O.; Chang, J.; Shao, L.; Liu, S.; Alimirah, F.; Koenig, K.; Le, C.; Mitin, N.; Deal, A.M.; et al. Cellular Senescence Promotes Adverse Effects of Chemotherapy and Cancer Relapse. Cancer Discov. 2017, 7, 165–177. [Google Scholar] [CrossRef] [Green Version]
  208. Carpenter, V.J.; Saleh, T.; Gewirtz, D.A. Senolytics for Cancer Therapy: Is All That Glitters Really Gold? Cancers 2021, 13, 723. [Google Scholar] [CrossRef]
  209. Chang, J.; Wang, Y.; Shao, L.; Laberge, R.; Demaria, M.; Campisi, J.; Janakiraman, K.; Sharpless, N.E.; Ding, S.; Feng, W.; et al. Clearance of Senescent Cells by ABT263 Rejuvenates Aged Hematopoietic Stem Cells in Mice. Nat. Med. 2016, 22, 78–83. [Google Scholar] [CrossRef] [Green Version]
  210. Selt, F.; Hohloch, J.; Hielscher, T.; Sahm, F.; Capper, D.; Korshunov, A.; Usta, D.; Brabetz, S.; Ridinger, J.; Ecker, J.; et al. Establishment and Application of a Novel Patient-Derived KIAA1549: BRAF-Driven Pediatric Pilocytic Astrocytoma Model for Preclinical Drug Testing. Oncotarget 2017, 8, 11460–11479. [Google Scholar] [CrossRef] [Green Version]
  211. Buhl, J.L.; Selt, F.; Hielscher, T.; Guiho, R.; Ecker, J.; Sahm, F.; Ridinger, J.; Riehl, D.; Usta, D.; Ismer, B.; et al. The Senescence-Associated Secretory Phenotype Mediates Oncogene-Induced Senescence in Pediatric Pilocytic Astrocytoma. Clin. Cancer Res. 2019, 25, 1851–1866. [Google Scholar] [CrossRef] [PubMed]
  212. Wang, Y.; Chang, J.; Liu, X.; Zhang, X.; Zhang, S.; Zhang, X.; Zhou, D.; Zheng, G. Discovery of Piperlongumine as a Potential Novel Lead for the Development of Senolytic Agents. Aging 2016, 8, 2915–2926. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  213. Guerrero, A.; Herranz, N.; Sun, B.; Wagner, V.; Gallage, S.; Guiho, R.; Wolter, K.; Pombo, J.; Irvine, E.E.; Innes, A.J.; et al. Cardiac Glycosides Are Broad-Spectrum Senolytics. Nat. Metab. 2019, 1, 1074–1088. [Google Scholar] [CrossRef] [PubMed]
  214. Mario Gonzalez-Meljem, J.; Haston, S.; Carreno, G.; Apps, J.R.; Pozzi, S.; Stache, C.; Kaushal, G.; Virasami, A.; Panousopoulos, L.; Neda Mousavy-Gharavy, S.; et al. Stem Cell Senescence Drives Age-Attenuated Induction of Pituitary Tumours in Mouse Models of Paediatric Craniopharyngioma. Nat. Commun. 2017, 8, 1819. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  215. L’Hôte, V.; Courbeyrette, R.; Pinna, G.; Cintrat, J.-C.; Pavec, G.L.; Delaunay-Moisan, A.; Thuret, J.-Y.; Moisan, A.D.; Mann, C. Ouabain and Chloroquine Trigger Senolysis of BRAF-V600E-Induced Senescent Cells by Targeting Autophagy. Aging Cell 2021, 20, e13447. [Google Scholar] [CrossRef]
  216. Denholm, M.; Rintoul, R.C.; Muñoz-Espín, D. SARS-CoV-2-Induced Senescence as a Potential Therapeutic Target. Eur. Respir. J. 2022, 60, 2201101. [Google Scholar] [CrossRef]
  217. Camell, C.D.; Yousefzadeh, M.J.; Zhu, Y.; Langhi Prata, L.G.P.; Huggins, M.A.; Pierson, M.; Zhang, L.; O’Kelly, R.D.; Pirtskhalava, T.; Xun, P.; et al. Senolytics Reduce Coronavirus-Related Mortality in Old Mice. Science 2021, 373, eabe4832. [Google Scholar] [CrossRef]
  218. Lee, S.; Yu, Y.; Trimpert, J.; Benthani, F.; Mairhofer, M.; Richter-Pechanska, P.; Wyler, E.; Belenki, D.; Kaltenbrunner, S.; Pammer, M.; et al. Virus-Induced Senescence Is a Driver and Therapeutic Target in COVID-19. Nature 2021, 599, 283–289. [Google Scholar] [CrossRef]
  219. Pham, A.M.; Ortiz, L.E.; Lukacher, A.E.; Kwun, H.J. Cellular Senescence Preserves Viral Genome Maintenance. bioRxiv 2022. [Google Scholar] [CrossRef]
  220. Seoane, R.; Vidal, S.; Bouzaher, Y.H.; El Motiam, A.; Rivas, C. The Interaction of Viruses with the Cellular Senescence Response. Biology 2020, 9, 455. [Google Scholar] [CrossRef]
  221. Giannakoulis, V.G.; Dubovan, P.; Papoutsi, E.; Kataki, A.; Koskinas, J. Senescence in HBV-, HCV- and NAFLD- Mediated Hepatocellular Carcinoma and Senotherapeutics: Current Evidence and Future Perspective. Cancers 2021, 13, 4732. [Google Scholar] [CrossRef] [PubMed]
  222. Szaniawski, M.A.; Spivak, A.M. Senotherapeutics and HIV-1 Persistence. Curr. HIV/AIDS Rep. 2020, 17, 219–225. [Google Scholar] [CrossRef] [PubMed]
  223. Hoppe-Seyler, K.; Herrmann, A.L.; Däschle, A.; Kuhn, B.J.; Strobel, T.D.; Lohrey, C.; Bulkescher, J.; Krijgsveld, J.; Hoppe-Seyler, F. Effects of Metformin on the Virus/Host Cell Crosstalk in Human Papillomavirus-positive Cancer Cells. Int. J. Cancer 2021, 149, 1137–1149. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Proposed Model for The Contribution of HPV-Induced Senescence to Cervical Cancer. Oncogenic variants of the Human Papilloma Virus (HPV) infect basal squamous epithelial cells of the cervix ①. Fortunately, the majority of infections resolve spontaneously and are cleared within two years ②. however, approximately 1% of infected patients can develop cervical cancer ③. The oncogenic potential of HPV is primarily attributed to the oncoproteins E6 and E7, which lead to the degradation/inactivation of the tumor suppressor genes p53 ④ and ⑤ Rb, respectively. As a defense mechanism, several HPV-infected cells undergo apoptosis and are eliminated. Alternatively, cervical epithelial cells may undergo senescence as a tumor suppressor mechanism. Primarily, cells that have become dependent on E6- or E7-mediated suppression of cell cycle control can undergo senescence as a consequence to E2-mediated inactivation of E6 or E7 ⑥. Otherwise, when a naïve, virus-free cell is infected with HPV, cells undergo senescence, which is likely to represent an Oncogene-induced Senescence (OIS) variant. Oncogene-induced senescent cells infected with HPV accumulate as a component of the cervical premalignant lesion (CIN) being generated ⑦. Only those cells that manage to escape OIS or those under constant pro-tumorigenic stimulation by the SASP, are capable of progressing into malignant phenotypes ⑧. Alternatively, we propose the use of senolytics, compounds that selectively eliminate senescent cells, to interfere with the accumulation of premalignant senescent cells in cervical lesions, as a novel pharmacological approach to interfere with the development of HPV-induced cervical cancer ⑨.
Figure 1. Proposed Model for The Contribution of HPV-Induced Senescence to Cervical Cancer. Oncogenic variants of the Human Papilloma Virus (HPV) infect basal squamous epithelial cells of the cervix ①. Fortunately, the majority of infections resolve spontaneously and are cleared within two years ②. however, approximately 1% of infected patients can develop cervical cancer ③. The oncogenic potential of HPV is primarily attributed to the oncoproteins E6 and E7, which lead to the degradation/inactivation of the tumor suppressor genes p53 ④ and ⑤ Rb, respectively. As a defense mechanism, several HPV-infected cells undergo apoptosis and are eliminated. Alternatively, cervical epithelial cells may undergo senescence as a tumor suppressor mechanism. Primarily, cells that have become dependent on E6- or E7-mediated suppression of cell cycle control can undergo senescence as a consequence to E2-mediated inactivation of E6 or E7 ⑥. Otherwise, when a naïve, virus-free cell is infected with HPV, cells undergo senescence, which is likely to represent an Oncogene-induced Senescence (OIS) variant. Oncogene-induced senescent cells infected with HPV accumulate as a component of the cervical premalignant lesion (CIN) being generated ⑦. Only those cells that manage to escape OIS or those under constant pro-tumorigenic stimulation by the SASP, are capable of progressing into malignant phenotypes ⑧. Alternatively, we propose the use of senolytics, compounds that selectively eliminate senescent cells, to interfere with the accumulation of premalignant senescent cells in cervical lesions, as a novel pharmacological approach to interfere with the development of HPV-induced cervical cancer ⑨.
Ijms 23 15512 g001
Table 1. Examples of the most frequently described hallmarks of cellular senescence.
Table 1. Examples of the most frequently described hallmarks of cellular senescence.
HallmarkDescriptionReference
Growth arrestUpregulation of p21Cip1[32]
Upregulation of p16INK4a[33]
Downregulation of ribosomal biogenesis[34,35]
Morphological changesLarge and flattened appearance[36]
Suborganellar damageTelomere dysfunction[37]
Persistent activation of the DNA damage repair response (DDR)[28]
DNA-SCARSs[18]
Proteosomal activity[22]
Accumulation of reactive oxygen species (ROS)[38]
Enhanced lysosomal biogenesis (SA-β-galactosidase)[26]
Accumulation of protein aggregates (lipofuscin)[39,40]
Mitochondrial dysfunction[41]
Epigenetic changes; SAHFHistone edits (H3K9Me3, HP-1, γH2AX)[42,43]
The SASPGrowth factors[44]
Chemokines[29]
Cytokines[45]
Angiogenic factors[46]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Saleh, T.; Khasawneh, A.I.; Himsawi, N.; Abu-Raideh, J.; Ejeilat, V.; Elshazly, A.M.; Gewirtz, D.A. Senolytic Therapy: A Potential Approach for the Elimination of Oncogene-Induced Senescent HPV-Positive Cells. Int. J. Mol. Sci. 2022, 23, 15512. https://doi.org/10.3390/ijms232415512

AMA Style

Saleh T, Khasawneh AI, Himsawi N, Abu-Raideh J, Ejeilat V, Elshazly AM, Gewirtz DA. Senolytic Therapy: A Potential Approach for the Elimination of Oncogene-Induced Senescent HPV-Positive Cells. International Journal of Molecular Sciences. 2022; 23(24):15512. https://doi.org/10.3390/ijms232415512

Chicago/Turabian Style

Saleh, Tareq, Ashraf I. Khasawneh, Nisreen Himsawi, Jumana Abu-Raideh, Vera Ejeilat, Ahmed M. Elshazly, and David A. Gewirtz. 2022. "Senolytic Therapy: A Potential Approach for the Elimination of Oncogene-Induced Senescent HPV-Positive Cells" International Journal of Molecular Sciences 23, no. 24: 15512. https://doi.org/10.3390/ijms232415512

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