Next Article in Journal
Diagnostic and Prognostic Value of PACAP in Multiple Myeloma
Next Article in Special Issue
Structural Insight into the Amino Acid Environment of the Two-Domain Laccase’s Trinuclear Copper Cluster
Previous Article in Journal
Correlation of the High-Resolution Computed Tomography Patterns of Intrathoracic Sarcoidosis with Serum Levels of SAA, CA 15.3, SP-D, and Other Biomarkers of Interstitial Lung Disease
Previous Article in Special Issue
The Effect of Prohibitins on Mitochondrial Function during Octopus tankahkeei Spermiogenesis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Exploring the Communication of the SASP: Dynamic, Interactive, and Adaptive Effects on the Microenvironment

1
Laboratory of Biochemistry and Cell Biology (URBC), Namur Research Institute for Life Sciences (NARILIS), University of Namur, 5000 Namur, Belgium
2
University of Lille, CNRS, Inserm, Pasteur Institute of Lille, UMR9020-U1277-CANTHER-Cancer Heterogeneity Plasticity and Resistance to Therapies, 59000 Lille, France
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(13), 10788; https://doi.org/10.3390/ijms241310788
Submission received: 30 April 2023 / Revised: 20 June 2023 / Accepted: 25 June 2023 / Published: 28 June 2023
(This article belongs to the Collection Feature Paper Collection in Biochemistry)

Abstract

:
Cellular senescence is a complex cell state that can occur during physiological ageing or after exposure to stress signals, regardless of age. It is a dynamic process that continuously evolves in a context-dependent manner. Senescent cells interact with their microenvironment by producing a heterogenous and plastic secretome referred to as the senescence-associated secretory phenotype (SASP). Hence, understanding the cross-talk between SASP and the microenvironment can be challenging due to the complexity of signal exchanges. In this review, we first aim to update the definition of senescence and its associated biomarkers from its discovery to the present day. We detail the regulatory mechanisms involved in the expression of SASP at multiple levels and develop how SASP can orchestrate microenvironment modifications, by focusing on extracellular matrix modifications, neighboring cells’ fate, and intercellular communications. We present hypotheses on how these microenvironmental events may affect dynamic changes in SASP composition in return. Finally, we discuss the various existing approaches to targeting SASP and clarify what is currently known about the biological effects of these modified SASPs on the cellular environment.

1. Introduction

Cellular senescence was first described as a stable and irreversible cellular state, in which cells permanently stop proliferating while remaining metabolically active. Since then, our understanding and definition of cellular senescence and its various roles have constantly evolved. Currently, senescence is presented as a complex and partially heterogeneous phenotype that can occur in response to the exhaustion of the proliferative capacity of the cell, and/or as a result of exposure to intrinsic and extrinsic stressors [1,2,3,4,5].
Since there is no universal marker for senescence, a combination of specific biochemical markers and phenotypic features is necessary to identify senescent cells. However, there is no consensus on the number and type of markers required to identify senescent cells, as one senescent cell is not equivalent to another [6]. Despite this, multiple pieces of evidence in the literature suggest that senescent cells share several characteristics, including a strong or prolonged growth arrest, an altered metabolism [3,4], and a specific senescence-associated secretory phenotype (SASP) [7,8,9,10]. Although SASP is a hallmark shared by various types of senescence, it is heterogeneous and can evolve in conjunction with changes in gene and protein expression, impacting biochemical features, trafficking, and intercellular signaling [3]. The diversity and the dynamic nature of SASP make it a complex process to understand, which is well demonstrated by its close association with both beneficial and detrimental effects depending on the physiological context [1,11,12,13,14,15].
These diverse effects are commonly linked with key SASP proteins whose secretion is increased in different senescence models. However, the variable component of SASP, which is modulated according to context, is often neglected in current knowledge, despite its importance.
Here, we first present the complexity of the senescence phenotype and the new biomarkers that have recently been revealed. Then, we describe the heterogeneity and plasticity of SASP composition, as well as its different levels of regulation, and highlight its contribution to both ageing and cancer. Additionally, we emphasize the importance of further investigating the complex crosstalk between the microenvironment and the SASP. Finally, we discuss how controlling SASP might be a suitable approach to treating age-related diseases.

1.1. The Senescent Phenotype: A Large Definition

Cellular senescence was identified more than 60 years ago by Hayflick and Moorhead who observed in vitro that normal human fibroblasts grown in optimal culture conditions have a limited proliferative capacity [16]. Nowadays, the “Hayflick limit” is widely accepted, especially since the discovery of telomere shortening-induced genomic instability [17]. This type of senescence is therefore referred to as replicative senescence (RS).
In recent years, progress has been made to suggest that senescence could be an adaptive stress response, resulting in the persistence of irreparable damages and mainly in a prolonged cell division arrest. Indeed, senescence can be induced by the activation of oncogenes or repression of tumor suppressor genes, known as oncogene-induced senescence (OIS) [18], or by repeated exposures to oxidative or genotoxic stress, known as stress-induced premature senescence (SIPS) [19]. Since the range of potential senescence-inducing stresses is wide, it is commonly labeled based on the nature of the inducer such as irradiation-induced senescence (IRIS) or therapy-induced senescence (TIS). One common feature of these different senescence inducers is the generation of irreparable DNA damage at the origin of the sustained cell cycle arrest. However, senescence induction can occur independently of DNA damage, such as senescence induced by sodium butyrate, a histone deacetylase inhibitor (HDACi) [20], or by mitochondrial dysfunction, known as MiDAS (Mitochondrial Dysfunction-Associated Senescence), which is associated with decreased NAD+/NADH ratios [21]. Finally, cellular senescence can also be triggered by epigenetic modifications, perturbed proteostasis, and autophagy impairment [22].

1.2. Biomarkers and Characteristics of Senescence

The scientific community has faced challenges in identifying robust and specific markers that characterize the senescent state, likely due to the heterogeneity of cellular senescence. While growth arrest is the main characteristic of senescent cells, it is not sufficient to distinguish them from other non-proliferative cell states, such as quiescence or terminal differentiation. Interestingly, prolonged quiescence induces a lack of response to proliferative stimuli and progressively leads to senescence [23]. Nevertheless, the establishment of the senescent phenotype is accompanied by a set of features and alterations that are now accepted as “standard hallmarks” (Figure 1).
To ensure the senescent state of a cell, several of these hallmarks must be validated in combination.

1.2.1. Standard Hallmarks

As displayed in Figure 1, the cell cycle arrest that characterizes the senescent cell often depends on the activation of two main pathways, p53/p21WAF1 and/or p16INK4/pRb. Senescent cells are enlarged and adopt a flat shape and their nuclei undergo structural and functional alterations [3]. These alterations include a decreased expression of lamin B1, a structural protein of the nuclear lamina, impairment of their structural integrity due to epigenetic alterations, and the formation of condensed heterochromatin regions, termed senescence-associated heterochromatic foci (SAHF) [24,25,26,27]. Senescent cells also exhibit alterations in histones (de)methylations and persistent DNA damage [28,29]. Their metabolic profile is deregulated, mainly through mitochondrial dysfunction, increased production of reactive oxygen species, elevated lysosomal activity (resulting in the SA-βgal activity), increased autophagy, and activation of the AMPK signaling pathway [30]. Apoptosis resistance via the upregulation of ephrins and anti-apoptotic proteins of the Bcl-2 family is also used as a senescence biomarker [31]. Finally, senescent cells have a particular secretory phenotype including soluble pro-inflammatory factors as well as growth factors, regulatory components of the MEC, bioactive lipids, and extracellular vesicles referred to as the senescence-associated secretory phenotype and further developed in Section 2.
While these biomarkers are commonly encountered in the literature, it is not unusual to observe variations in their expression and effectors.

1.2.2. Other Pathways Involved in the Senescence-Mediated Cell Cycle Arrest

The cell cycle arrest mediated by the p53, p21WAF1, and p16INK4 results in the hypophosphorylation of pRb. This leads to the subsequent sequestration of E2F and prevents the expression of genes necessary for cell cycle progression. Interestingly, the p53/p21WAF1 pathway is also involved in assembling the repressive DREAM complex, which serves the same purpose, thereby reinforcing the role played by p53 [32,33]. However, other cell cycle regulators are also involved in this senescence-associated cell cycle arrest. Additionally, the actors of the cell cycle arrest can vary over time, with some being expressed at the onset of senescence and others during its late phases. As an example, in glyoxal-induced senescent keratinocytes, the cell cycle arrest is first mediated by the protein kinase B-FOXO3a-p27KIP1 pathway but is sustained over time by the p16INK4/pRb pathway [34]. Furthermore, in therapy-induced senescence in positive breast cancer cell lines, the tyrosine kinase inhibitor Lapatinib can induce senescence by increasing the expression of p27KIP1 and p15IKK4b [35] and in prostate cancer cells, supraphysiological androgen levels regulate the establishment of senescence through p15IKK4b [36]. Thus, it is important to extend the study of the expression of other proteins involved in cell cycle arrest and to verify their expression over time. It would be beneficial to decipher the actors involved in the cell cycle arrest and their expression patterns over time according to the cell type and senescence inducer. Currently, it is still unclear which pathways act concurrently and whether there are differences in the hierarchy and kinetics of the events, as well as which pathways are dispensable. It is worth noting that while the senescence-associated cell cycle arrest has historically been considered “irreversible”, primarily studied in fibroblasts, under certain conditions and contexts, both tumoral and normal senescent cells can resume proliferation and thus escape senescence [37,38].

1.2.3. Surfaceome

Since any cell population is inherently heterogeneous, the induction of senescence only affects a fraction of the population, which therefore becomes enriched in senescent cells. Therefore, the identification, sorting, and targeting of these senescent cells using surface markers is of crucial importance. Althubiti et al. [39] were the first to perform proteomic analyses of plasma membrane-associated proteins in an EJ bladder cancer cell line with regulating expression of p21WAF1 or p16INK4. They identified 107 potential protein markers and demonstrated that a set of 10 proteins (DEP1, NTAL, EBP50, STX4, VAMP3, ARMX3, B2MG, LANCL1, VPS26A, and PLD3), when combined, could serve as markers of senescence and facilitate the detection of senescent cells in various human tissues [39]. Since this first analysis, other proteins that contribute to the surfaceome of senescent cells have been described such as the dipeptidyl peptidase 4 (DPP4) [40] and the urokinase-type plasminogen activator receptor uPAR [41]. B2MG, DPP4, and uPAR are therefore used as targets for the specific clearance of senescent cells in vitro and in vivo [41,42,43].

1.2.4. Alterations of Nuclear Shape and Nucleus-Cytoplasm Exchanges

Senescence is also associated with alterations of the nuclear architecture. Besides the well-known modification of the nuclear lamina partly linked to lamin B1 loss [44], other nuclear features including nuclear matrix, nucleolus, heterochromatin, and even nuclear shape and size can also be altered [45]. In particular, the increased nuclear shape is a promising biomarker to predict the senescent state [46]. By using machine learning algorithms, the group of Heckenbach demonstrated that nuclear morphology could predict senescence following different senescent inducers, in different cell types and species. Deep learning could therefore be used as a tool to predict the occurrence of senescent cells in clinical contexts and to study their potential negative effects, paving the way for the prevention of the harmful effects of senescence [47].
Moreover, the nucleus–cytoplasm exchanges are altered in senescence, as the transcription-export (TREX) machinery and the nucleocytoplasmic trafficking (NCT) are downregulated [48,49]. Interference in exchanges between the nucleus and the cytoplasm of senescent cells leads to a reduction in the transmission of extrinsic signals toward the nucleus and alters the nucleus-to-cytoplasm protein–RNA transport, resulting in the establishment of a “nuclear barrier” [48,49].

1.2.5. UPR and ER Control Quality

The endoplasmic reticulum (ER) is a dynamic structure playing a major role in the folding, synthesis, processing, and quality control of secreted and transmembrane proteins. When ER homeostasis is perturbed, an adaptive mechanism called UPR (Unfolded Protein Response) aims to restore ER homeostasis and promote cell survival [50]. It is now well accepted that different cell types undergoing senescence upon various inducers promote UPR activation [47,51]. We and other groups have shown evidence that the different arms of the UPR pathway may control several senescence hallmarks. Indeed, UPR inducers promote a premature senescence phenotype [52,53]. Moreover, the ATF6α arm of the UPR controls cell size and enlargement in replicative senescent fibroblasts [52]. In addition, the expression of autophagic component MAP1LC3-II in glioblastoma cells undergoing therapy induced-senescence is triggered by the PERK/ATF4 arm [54], and in Hras-induced senescence, production of SASP induces the UPR activation via proteotoxic stress [55,56]. The role of the UPR regarding SASP is discussed in Section 3.
How senescence affects the other ER quality control systems, namely, the ERAD (Endoplasmic reticulum-associated protein degradation) and ER-phagy, is poorly understood. To date, only one study has reported a relationship between ER-phagy and senescence. The authors demonstrated activation of the ER-phagy mediated by FAM134B, an ER-phagy receptor, upon advanced glycation end product (AGE) stress-induced senescence in primary human nucleus pulposus (NP) cells [57]. They also found that enhancement of ER-phagy by FAM134B overexpression reduces the percentage of SA-β Gal positive cells, while the suppression of FAM134B exacerbates it upon AGEs stress in NP cells [57]. The role of ERAD and ER-phagy deserves to be further explored in senescence.

2. Characteristics of SASP

During its lifespan, a cell can release a range of molecules into its surrounding extracellular environment. The secretory profile is constantly influenced by intrinsic characteristics, which depend on the cell type and its differentiation stage, as well as extrinsic factors such as the change in the cellular environment. Indeed, a clear remodeling of the secretory profile can be observed in cells undergoing senescence [58]. Examining the secretome of senescent cells reveals modifications in the levels of soluble, insoluble, and extracellular vesicle (EV)-related components. Senescence establishment can cause these components to be either exacerbated or partially depleted, and can also lead to the secretion of new components when compared to proliferative cells [59].

2.1. Reported SASP Factors

Senescent cells exhibit a distinct and dynamic secretome different from their exponentially growing counterparts [60]. This SASP is complex and is composed of hundreds of different proteins and non-protein signaling molecules [60,61]. Despite the diversity of the factors secreted, a core protein secretome can be distinguished from the soluble part of SASP (sSASP) (Table 1). In human primary fibroblasts subjected to various senescence inducers (IR, RAS, atazanavir (ATV)), this core protein sSASP includes, among others, STC1 (stanniocalcin 1), chemokines such as CXCL1 (C-X-C motif ligand 1), and proteases such as MMP-1 (matrix metalloproteinase 1) [60]. Hemostasis-related factors, another class of bioactive compounds, show a marked increase in secretion into the extracellular medium by senescent human primary fibroblasts when exposed to different inducers (IR, doxorubicin (DOX), and MiDAS) [61]. Non-protein signaling molecules, including various bioactive oxidized lipid metabolites, prostaglandins, and nitric oxide can also be found enriched in the sSASP of senescent cells [62,63,64,65]. While empirical research has focused on soluble factors secreted by senescent cells, new studies show evidence that EVs are also a substantial and effective part of SASP [66]. EVs are lipid membrane vesicles containing cytosol from the secreting cells and are released by multiple cell types. According to their origin, biological function, and secretion, EVs can be classified into two main subtypes: exosomes and microvesicles (MVs) [67]. In this context, it has been evidenced in multiple cell types that EV secretion increases after exposure to different senescence inducers, with changes in cargo composition including proteins and genomic content such as microRNAs (miRNAs) and lipids [63,68,69].
The core secretome is a concept based on a limited number of established cellular models used to study senescence, such as human primary fibroblasts, and represents only a portion of the complete picture. Hence, the application of a core SASP should be considered as a tool to evaluate the acquisition of the senescent phenotype in a standardized manner, rather than a way to generalize the effects of SASP on the cellular microenvironment.
Indeed, the bioactive effects of SASP may be more closely linked to specific and possibly subtle variations in the secretome that result from the combination of a specific cell type and a particular senescence inducer, rather than the shared components.

2.2. SASP Heterogeneity and Plasticity

Coppé and colleagues demonstrated initially that only a subset of SASP proteins was shared between fibroblasts and prostate epithelial cells upon irradiation-induced senescence (IRIS) [9]. A subsequent large-scale proteomic analysis of SASP then revealed only 58 shared SASP factors between fibroblasts and renal epithelial cells in IRIS [60]. When considering other proteomic studies on various cell types and senescence inducers, such as UVA-induced senescent keratinocytes and IRIS mesenchymal stem cells [71,72], the number of shared SASP factors drops to 19, suggesting that only a handful of proteins are commonly secreted across all types of senescent cells. Given the differences in experimental parameters such as EV isolation methods, detection techniques, and time points assessed after senescence induction, comparing these studies remains challenging.
Secondly, some SASP factors are secreted at different times depending on the cell type. For instance, in IRIS, the IL- gene is overexpressed on days 10 and 20 in fibroblasts, but only on day 10 in keratinocytes, and on day 20 in melanocytes [73]. This point emphasized SASP plasticity over time. Moreover, we showed an increased secretion of IL-6 and IL-8 in UVB-induced senescent keratinocytes on day 3 that disappeared on day 7 following senescence induction [70]. In addition, HDFs in replicative senescence also exhibit two distinct phases of SASP gene expressions. The first phase is often characterized by an overexpression of SASP genes related to inflammation, such as IL-6 and IL-8, while the second phase is more characterized by changes in the expression of genes associated with extracellular matrix modulation, such as MMPs [74]. This is related to the temporal regulation of SASP, further developed in Section 3. Further experiments are required to better understand the kinetics of SASP factors and their physiological relevance in the short and long term.
Thirdly, SASP composition is also influenced by the senescence inducers. Senescent IMR-90 fibroblasts present a different secretome profile depending on whether the senescence was induced by X-rays, ATZ, or RAS overexpression [60]. Similar results were observed on mesenchymal stem cells in senescence induced by oxidative stress, doxorubicin treatment, X-ray irradiation, or replicative exhaustion [72].
Finally, the matrix and cellular microenvironment of senescent cells can impact their secretome composition. The substrate stiffness impacts the NF-κB phosphorylation status in UV-induced senescence in fibroblasts [75], suggesting that the ECM composition could have an impact on SASP composition. Moreover, it has been demonstrated that co-cultivating squamous cell carcinoma and RS fibroblasts exacerbate some SASP gene expressions, such as IL-8, IL-, or CCL2, in RS fibroblasts [76]. These findings demonstrate the need of developing cellular models that consider the influence of the ECM and other cell types within the same tissue. Such models would allow a better understanding of the complex interplay between SASP and its microenvironment, which can affect SASP composition over time.

3. Regulation of SASP

The regulation of SASP involves transcriptional, post-transcriptional, epigenetic, and translational mechanisms. In addition, the secretion of SASP components is regulated through intracellular trafficking, and many compartments of secretion are altered during senescence. These alterations could potentially affect the dynamic and heterogeneous composition of SASP.

3.1. Transcriptional Regulation

Multiple signaling pathways have been identified to activate transcription factors that play a crucial role in regulating the expression of inflammatory cytokines.
First, there is a clear link between the expression of SASP and the DNA Damage Response (DDR) pathway, as several DDR proteins (ATM, Chk2, and NBS1) are necessary for the initiation and maintenance of the cytokine response in IRIS fibroblasts [29]. It has recently been described that in the absence of DNA damage, such as after sodium butyrate treatment, the SASP of fibroblasts still relies on the non-canonical activation of DDR and the accumulation of ATM, MRE11, and NF-kB on chromatin [77]. However, the expression of SASP factors seems independent of the cell-cycle regulators p53 and pRb, as their invalidation or inactivation even promotes IL-6 secretion [9,29]. On the other hand, it has been recently demonstrated that the knockdown of p16 in fibroblasts decreases the expression of IL-6 and IL-8 in OIS and etoposide-induced senescence [78].
Then, the NF-κB and C/EBPβ transcription factors were identified to be involved in the regulation of CXCR2 ligands expression, including IL8 in fibroblasts in OIS [79]. The activation of NF-κB in fibroblasts has been demonstrated to depend on GATA4, whose regulation by p62 is suppressed during senescence but mediated by DDR-related ATM and ATR [80]. NOTCH1 plays a dual role in the regulation of SASP. It is positively associated with early SASP expression in OIS in fibroblasts but then represses late SASP expression by suppressing C/EBPβ expression [81].
Regarding the regulation of inflammatory cytokine expression, the activation of another signaling pathway involved in inflammation, JAK/STAT, has also been demonstrated in a PTEN-deficient prostate cancer mouse model [82], as well as in senescent preadipocytes [83].
Finally, the cGAS/STING pathway has been highlighted to be involved in the regulation of inflammatory SASP factors, notably, IL-6 and CXCL10 secretion, via NF-κB activation in vitro and in vivo [84], following the detection of cytoplasmic chromatin fragments (CCFs) [85] associated with a loss of nuclear integrity following Lamin B1 (LMNB1) downregulation [86]. It has recently been demonstrated that COX2 plays an important role in regulating the expression of several inflammatory SASP components in OIS through an autocrine feedback loop involving prostaglandin E2 (PGE2) binding to EP4, but the downstream pathways of PGE2 and EP4 remain unknown. Nevertheless, the COX2 pathway is thought to be able to activate major SASP transcriptional regulators, such as NF-κB, C/EBPβ, and GATA4 [87].

3.2. Post-Transcriptional Regulation

While early SASP is mainly regulated at the transcriptional level, its long-term SASP expression is mainly driven by post-transcriptional mechanisms. This has been demonstrated by the lack of impact of actinomycin D treatment, an inhibitor of transcription, on the expression of several SASP factors [88]. P38MAPK appears to be an important factor in the temporal regulation of SASP. If it is first activated after the induction of senescence, it enables the expression of SASP factors, such as IL-6 and IL-8, through NF-κB activation in IRIS fibroblasts [89]. It is also involved in the subsequent post-transcriptional regulation of SASP by restricting the binding of AUF1 to the 3′-UTRs of several SASP mRNAs, including IL-6 and IL-8, thereby preventing their destabilization, as demonstrated in bleomycin-induced senescent fibroblasts [88]. The mTOR pathway is also involved in the post-transcriptional regulation of SASP. Specifically, mTOR activates the translation of MK2 (or MAPKAPK2), which can phosphorylate and inhibit the RNA-binding protein ZFP36L1, also involved in the destabilization of several SASP mRNAs [90]. The mTORC1 kinase has also been shown to modulate senescence-induced inflammation and SASP [91].
As previously mentioned, studies on the regulation of SASP have primarily focused on the transcriptional and post-transcriptional regulation of inflammatory cytokines. However, there is a limited understanding of the regulatory mechanisms underlying other SASP factors, such as growth factors and proteases. A recent study on fibroblasts has described that E2F4, TEAD1, and AP-1 transcription factors are major regulators of RS [92]. Moreover, AP-1 is involved in the expression of IL-6, IL-, and MMP-10, as their expression is abrogated when expressing a dominant-negative isoform of c-Jun, one of the subunits of AP-1, during OIS in fibroblasts [93]. Future studies focusing on the regulation of non-inflammatory SASP factors would be valuable.

3.3. Epigenetic Regulation

The physical clustering of SASP genes, such as MMPs (MMP-1, -3, -10, and -12) or chemokines (CXCLs and CCLs), suggests that the regulation of their expression may depend, at least in part, on broader changes in chromatin conformation [94]. Indeed, several histone variants can influence the expression of SASP genes. For example, the relocation of the macroH2A1 histone variant away from SASP genes following ER stress response-mediated activation of ATM in fibroblasts in OIS is involved in the maintenance of SASP gene expression [56]. Moreover, the increased expression of histone variant H2A.J in fibroblasts undergoing etoposide-induced senescence enhances the expression of multiple genes associated with inflammation and immune response. This effect is likely attributed to the interaction of H2A.J with other factors [95]. In addition, nuclear HMGBs bind to DNA, facilitating the access of transcription factors to promoter regions. In fibroblasts in RS or IRIS, HMGB1 can be released into the extracellular space and act as an alarmin to activate NF-kB, which subsequently upregulates the expression of pro-inflammatory target genes [96]. Furthermore, HMGB2 preferentially localizes to SASP gene regions during OIS in fibroblasts, protecting them from being incorporated in transcriptionally repressed SAHF regions [97].

3.4. Secretory Control: Compartments of Secretion and Vesicular Trafficking

Even though most organelles are morphologically or functionally affected during senescence, their proportion increases in senescent cells due to various signaling defects. In addition to nuclear and mitochondrial dysfunction, the endoplasmic reticulum, Golgi apparatus, and lysosomal compartments are strongly involved in the generation, processing, and release of SASP factors [91].
The ER is the site of membrane biosynthesis used in secretory and excretory pathways. It is responsible for folding and maturating secreted proteins, making it the first compartment of secretion. Recently, it has been proposed that ER stress and the subsequent activation of the unfolded protein response upon senescence could contribute to the modified secretome of senescent cells [98]. While there are multiple connections between the UPR and inflammation [99,100], the UPR and normal or tumoral secretome [101,102,103], as well as the UPR and direct control of MMPs [104], the data directly linking ER stress with SASP are scarce. Our group demonstrated that knocking-down ATF6 in RS fibroblasts decreased IL-6 mRNA levels [57]. Chen et al. [56] proposed that UPR induction in RAS-mediated senescence led to macroH2A1 expression, which, in turn, induces the expression of various SASP-associated genes in fibroblasts. Moreover, Dorr et al. [55] suggested that OIS and TIS induce proteotoxic stress and UPR activation to ensure SASP production. Nevertheless, the exact role of ER and UPR must be further addressed to confirm a central role in the establishment and composition of SASP.
The Golgi structure is also altered in senescent cells [105]. These alterations can not only be mediated by the translocation of a G protein γ subunit from the plasma membrane to the Golgi [106] but also by the impaired expression of the vacuolar ATPase ATP6V0A2, which acidifies organelles such as Golgi, endosomes, or lysosomes [107]. This results in deep changes in post-translational glycosylation in the Golgi, impacting SASP compounds. In addition, the trans-Golgi network (TGN) appears to be increased in senescent cells. A blockade of TGN components such as the protein kinase D1 (PKD1) is associated with the intracellular accumulation of some SASP factors including IL-6 and IL8 in OIS fibroblasts [108].
Lysosomes are at the crossroads of endocytic and exocytic pathways, and their increased abundance in senescent cells may be associated with the exacerbation or deregulation of these pathways. Besides their partnership with the Golgi apparatus and the endosomal compartments, lysosomes are also important for the clearance of cytoplasmic chromatin fragments (CCFs). CCFs may leak from the nucleus in the cytoplasm of senescent cells and induce an SASP; both CCFs and SASP inductions would be related to a retrograde mitochondrial–nucleus signaling pathway associated with the mitochondrial increase in ROS species [91].
In melanoma cells, the lysosomal exocytosis mediated by the small GTPase RAB27A has also been shown to be upregulated in TIS and to participate in SASP factors secretion, including the chemokines CCL-2 and CXCL-12 [109]. Along with this enhanced lysosomal secretion, senescent cells exhibit a remodeling of their lysosomal proteome with selective enrichment in some lysosomal resident proteins such as those implicated in vesicular transport and fusion [109].
Small EV and exosome secretions are now part of the specific secretory phenotype. The release of senescence-associated exosomes is linked to RAB27A expression, as silencing of RAB27A leads to decreased exosome secretion in fibroblasts undergoing RS or OIS [110]. Rab27 GTPases are associated with the connection of multivesicular endosomes and the secretion of exosomes [111]. The enhanced biogenesis of EVs and their release by senescent cells have been demonstrated to be associated with the extent of DNA damage generated by the senescence inducer, as well as the activation of the ceramide synthetic pathway [112]. EVs and exosomes also contribute to SASP and its paracrine impact. For example, EVs from senescent stromal cells can enhance the proliferation of cancer cells by promoting the activation of the ephrin-A2 tyrosine kinase receptor, which interacts with overexpressed ephrin-A1 on the surface of the cancer cells, thereby boosting an Erk-dependent proliferation pathway [110,113]. In addition to being components of SASP, the release of senescence-associated EVs seems to be a mechanism used by senescent cells to discard cytoplasmic chromatin DNA fragments (CCFs), thus limiting DNA damage accumulation caused by major stress exposure and potentially modulating SASP [110,112]. Finally, EVs and exosomes are opening a new era of a multifunctional SASP due to their wide range of potential contents, as well as the specificities of the cellular niches and partnerships in which they operate.

4. Senescence and SASP In Vivo

Senescent cells accumulate in tissues with age. A meta-analysis showed that even if the proportion of senescent cells in 14 different human tissues is correlated with chronological age, it varies depending on the tissue type and the senescence marker used [114]. Moreover, the accumulation of senescent cells is also detected at pathological sites due to various stress signals regardless of age [115]. As a result, there is a wide diversity of senescent cells throughout the body. Furthermore, several studies have indicated that the elimination of senescent cells using transgenic mice, such as the INK-ATTAC and p16-3MR mouse models that both specifically target the elimination of p16-positive cells [15,116], or through the use of small pharmacological molecules called senolytics (which kill senescent cells) or senomorphics (which suppress some or all of their phenotype/properties) has shown improvements in healthspan, alleviated several age-associated conditions, delayed tumor formation, and mitigated the side effects of chemotherapy [117,118,119,120]. These findings highlight senescence as a significant contributor to ageing and associated pathologies. Therefore, SASP profiles may contribute to developing senescence biomarkers in human plasma or other biofluids, as well as assessing the efficacy of senescence-targeted therapies (see Section 6). Basisty and colleagues defined in senescent culture cells a core SASP including GDF15, STC1, SERPINE1/PAI-1, and MMP1, which are also reported to be significantly increased among the plasma markers of ageing in humans [60]. Another study showed that doxorubicin-induced senescence enriched the SERPINE1/PAI-1 SASP factor in plasma in vivo [61]. However, these markers can also serve as biomarkers for several diseases such as cardiovascular, metabolic, neurodegenerative, and malignant diseases, regardless of age. This makes them indicators of a “state of ageing” rather than a chronological accumulation of senescent cells. Surprisingly, the production of SASP factor IL-6 is increased in in vitro senescent models, but the circulating levels of IL-6 are not significantly different between young and elderly subjects [121,122]. Interestingly, Markov et al. identified, by using machine learning on a human cohort, immune biomarkers to predict brain ageing and suggested that intervention on these biomarkers could prevent brain ageing [123]. Regarding the use of senolytics for treating human cellular senescence-associated diseases, it is worth noting that the SASP factors assayed or detected can vary significantly from one study to another [124,125]. These variations are undoubtedly specific to the origin of senescent cells, which makes them difficult to use as markers in clinical settings. Further studies are needed to be able to robustly consider SASP factors, or a subset of them, as reliable biomarkers and to determine how SASP could be effectively translated into clinical applications.

5. Pleiotropic Roles of SASP

Due to its diverse composition, SASP can have pleiotropic effects on the cellular environment, which can be either beneficial or deleterious. Several recent publications have extensively covered this topic [10,58,126]. However, in this section, we focus on certain physiological and pathophysiological consequences that SASP can cause. Specifically, we examine how SASP influences extracellular matrix remodeling, intercellular communication modification, ageing, and the development of cancer.

5.1. Extracellular Matrix Remodeling

Collagen alterations in the dermal ECM have been associated with the decline in human skin structure and function during ageing. This emphasizes the overexpression of cysteine-rich protein 61 (CCN1) and MMP-1 expression in the SASP of senescent fibroblasts [127]. Changes in ECM composition and ECM-degrading molecules produced by SASP also disrupt elastin and collagen fiber networks and basement membranes in ageing tissue [128]. Therefore, SASP-related changes in ECM components have a notable effect on cell functions and fates by altering the tortuosity of collagen or by increasing the stiffness of the ECM as ageing progresses.
In the context of tissue injury, senescent cells can play a role in regeneration by accelerating wound healing or limiting fibrosis. For instance, the transient secretion of PDGF-AA (Platelet-derived growth factor AA) from senescent fibroblasts is necessary for effective healing following skin injury [15]. Strikingly, the short-term presence of miR-23a-3p in EVs derived from senescent fibroblasts allows a faster wound closure of epidermal keratinocytes [129]. However, to date, few studies have investigated the link between senescent cells and ECM since the matricellular protein CCN1 has been shown to induce ROS-induced senescence in fibroblasts during wound healing [130]. In addition, the secretome of senescent HSCs (hepatic stellate cells) plays an important role in fibrotic degradation and maintenance of liver tissue homeostasis [131]. Moreover, the elimination of senescent p16High LSECs (Liver Sinusoïd Endothelial Cells) in mice induces fibrosis [132]. Recently, efforts have been made to better characterize the changes in the matrisome of senescent cells and their effects on the environment. Hierbert and colleagues [133] described that the activation of Nrf2 in fibroblasts triggers the production of a senescence-promoting ECM via the expression and secretion of certain ECM proteins, such as PAI-1. This can accelerate wound closure and promote re-epithelization in vivo. In addition, Nrf2 inhibition in fibroblasts reduces the production of collagen I and alters ECM deposition [134]. Furthermore, when muscle stem cells are seeded onto decellularized ECM maintained by senescent fibroblasts, their responses and functions are affected, resulting in enhanced expression of fibrogenic markers and reduced myogenic markers [135].

5.2. Tumor Suppression and Promotion

While cellular senescence is widely recognized as an anti-tumor barrier, there is growing evidence to suggest that senescence may also have a tumor-promoting role.
Senescent cells have been observed at sites of benign tumors, such as prostatic hyperplasia and melanocytic naevi [136,137]. The factors secreted by these cells influence the tissue microenvironment and impact cellular differentiation and proliferation, notably in cancer cells [136,138]. Moreover, the first studies pointing out the role of the cellular microenvironment in the promotion of cancer progression highlighted the role of Carcinoma-Associated Fibroblasts (CAFs) in prostate cancer progression [139]. It has been subsequently reported that senescent fibroblasts share many features with CAFs, and can have a similar impact on the differentiation of epithelial cells initiated by cancer, and on tumor growth both in vitro and in vivo [13,140]. Co-culture systems and xenograft models have shown that SASP from senescent fibroblasts promotes the tumorigenesis of premalignant epithelial cells [141], induces epithelial–mesenchymal transition (EMT), and increases tumor vascularization, which suggests pro-tumorigenic properties [98].
We and others have shown that Normal Human Mammary Epithelial Cells (HMECs) and Normal Human Epidermal Keratinocytes (NHEKs) can spontaneously and systematically escape from the senescent state [142]. Some rare senescent cells re-enter the cell cycle via a process called Post-Senescence Neoplastic Escape (PSNE) with characteristics similar to those observed during the early stages of tumor initiation [52,143,144,145,146]. More importantly, when xenografted in nude mice, these PSNE cells developed into disseminated skin lesions such as hyperplasia and small non-melanoma skin carcinoma, evidencing their tumorigenic potential [144,145]. SASP from senescent dermal fibroblasts promotes neoplastic escape from normal human keratinocytes and increases markers of EMT as well as the migration of emerging cells [145]. This was attributed to the activation of the membrane PAR-1/Thrombin receptor by MMPs among SASP of senescent fibroblasts [145]. Furthermore, a recent study has identified a BDNF-TrkB axis as being associated with the role of SASP of aged fibroblasts in promoting EMT initiation in primary keratinocytes from aged donors [147].
Another point is that the alteration in the secretion of ECM components and regulators by senescent prostate cells generates a favorable environment for tumor development [148]. UVB-induced senescent fibroblasts were shown to produce an ECM that promotes proliferative signaling pathways of preneoplastic HaCaT epidermal keratinocytes [149]. Enhanced collagen deposition has been described along breast cancer progression, with dysregulated architecture and increased reticulation via abnormal expression of lysyl oxidase and MMP-resistant collagen isoforms, contributing to carcinoma progression [150]. Moreover, increased collagen matrix stiffness has been shown to control the cell fate of normal breast cells in 3D models. The increase in collagen concentration led to the overexpression of the α6β4 integrin pair, profound disruption of the architecture from regular normal-like acini and tubes to a tumor-like mass of increasing size, and decreased differentiation [151].
Taken together, the combination of SASP factors responsible for the pro-tumorigenic effects remains poorly understood. However, progress has been made in defining how context-dependent (such as cellular partnerships, specific soluble proteins, or membrane receptors) can influence the effects of SASP in cancer promotion [152].

5.3. Senescence Induction and Reinforcement

In addition, SASP primarily influences the induction and reinforcement of senescence. It is now clear that senescent cells maintain their phenotype through an autocrine positive feedback loop in which the main factors identified are cytokines such as IL-6 and IL-8 [79,153]. Similarly, the same SASP factors and many others such as TGF-β family ligands, VEGF, and chemokines such as CCL2 and CCL20 also play an important role in inducing paracrine senescence in neighboring cells [154]. The intensity of SASP can impact local homeostasis paracrine through signals that propagate the senescent state, exacerbating local stress, and inducing ROS-mediated damage in neighboring cells. This is the so-called SMS effect of SASP. Hence, conditioned media (CM) of cells exposed to UV radiations (UVA, B, and C) initiate bystander DNA damage in non-exposed neighboring cells [155]. Moreover, since the recent interest in deciphering the role of eSASP, new studies have shown the important contribution of microvesicles in the propagation of the senescent phenotype, for example, via the transfer of interferon or miRNA cargo factors [156,157].

5.4. Other Functions of SASP

SASP can modulate the fate of neighboring cells in several ways and can even impact the differentiation of surrounding cells.
For example, Wiley et al. [21] have shown that CM harvested from fibroblasts whose mtDNA has been depleted (rho0) can block adipogenesis in preadipocytes but promote keratinocyte differentiation. A recent study also showed that TGF-β secreted by senescent cells can influence the differentiation of T helper cells during the response to influenza infection in mice [158]. A proteomic analysis of CM of fibroblasts in IRIS identified a role for SASP in hemostasis, platelet activation, and degranulation [61]. Moreover, transient exposure of primary mouse keratinocytes to SASP of OIS keratinocytes led to enhanced plasticity via the increased expression of stemness markers and better regenerative capacities in vivo, while long-term exposure promoted senescence, reducing regenerative stimuli [159]. Therefore, SASP may induce cellular plasticity and tissue regeneration capacities according to its intensity and duration, and may promote cellular reprogramming in neighboring cells [160]. In parallel, ECM stiffness is also of particular importance in stem cell response and can lead to considerable changes in cell signaling, shape, and differentiation status [161]. Decellularized matrices form cardiac explants of donors of various ages, differentially impacting the cell fates and functions of IPSC-derived cardiomyocytes (ICMs) cultured on top of the matrices. In fact, matrices from young donors can enhance the proliferation and functions of young ICMs, while matrices from aged donors promoted their senescence [162]. In addition, SASP can reduce muscle stem cell expansion. In damaged muscle, senescent cells altered their normal niche to create an age-related-inflamed microenvironment that impairs regeneration [163].
Finally, Saul et al. [164] conducted a bioinformatic analysis and identified a panel of genes, called SenMayo, which is enriched in elderly vs. young women. They evaluated the applicability of SenMayo across tissues and species. Using their tool, they were able to evaluate intercellular communication patterns of senescent cells with other cells in their microenvironment at the single-cell level. Overall, they showed that senescent hematopoietic and mesenchymal cells interact with neighboring cells mainly through the Macrophage Migration Inhibitory Factor (MIF) pathway.

6. Developing Strategies to Block SASP or Its Specific Effects

Given the importance of senescence in physiological processes, it is reasonable to think that there is a threshold beyond which the accumulation of senescent cells induces a microenvironment conducive to the development of pathologies via SASP. The accumulation of senescent cells can also occur when the immune system ages, altering the ability of immune cells to clear senescent cells.
Elimination of senescent cells by senolytics demonstrated a contributive role of senescent cells in ageing and age-related diseases [165] and paved the way for the development of senotherapeutic approaches. Therefore, over the past 5 years, senotherapeutic research has emerged to slow down the ageing phenotypes. Current senotherapeutic strategies targeting senescent cells are mainly based on drugs that specifically kill senescent cells (senolytics) and components that suppress the detrimental effects of SASP without inducing senescent cell death (senomorphics, also known as senostatics) [166,167,168,169,170,171,172]. Other senotherapeutic strategies include prodrugs, protein degraders, nanocarriers, and immunotherapies [173]. It is worth noting that a recent study showed that eliminating senescent cells by using chimeric antigen receptor (CAR) T cells that specifically target senescence-specific surface antigens such as uPAR improved the survival of mice with lung adenocarcinoma and restored tissue homeostasis in a chemical-induced liver fibrosis mouse model [41]. Emerging preclinical evidence has highlighted the significant potential of these approaches [27,28,124]. However, further analyses are necessary to rule out the potential adverse effects of long-term administration. Additionally, there are ongoing efforts to evaluate combinations of senotherapies in individuals with multiple age-related diseases [174].
Nevertheless, in this section, we will not cover all senotherapeutic strategies, especially as excellent reviews have recently been published on senolytic developments [171,175,176,177], but rather focus on those with senomorphic activities (Table 2), based on their ability to block SASP components.
A first strategy would consist in using neutralizing antibodies, recognizing and blocking specific surface proteins upregulated at senescence. Secretion of IL-6 has been decreased in senescent HUVECs and fibroblasts treated with anti-TNFα or anti-ephrin B2 antibodies, respectively [178,179]. Several other surface proteins are known to play a role in the regulation of SASP profiles, including SCAMP4, Notch, and CD36 [40,81,180]. However, it has not yet been reported that the use of neutralizing antibodies targeting SCAMP4, Notch, or CD36 can impact the composition of SASP and, therefore, arrive at a conclusion regarding their senomorphic properties. In a model of bleomycin-induced senescence, the secretion of certain SASP factors (including IL-6 and IL-8) can be directly inhibited with neutralizing antibodies such as those against the membrane-bound IL-1α [181]. It would be interesting to investigate the impact of other neutralizing antibodies directed against other major SASP factors such as circulating IL-1β-, IL-6, or their receptors [182], on their abilities to alter the chemical composition of SASP, impair SASP-mediated effects, and attenuate other features of senescence in different cell types.
A second strategy would be to use pharmacological and natural compounds. Many senomorphics are polyphenols (including flavonoids, phenolic acids, lignans, and stilbenesenes) that possess antioxidant activities, but their modes of action have not been thoroughly studied. Other senomorphics are plant extracts consisting of a mixture of terpenes, alkaloids, and polyphenols. The biological effects of these compounds are multiple, ranging from the activation of antioxidant enzymes to the reduction in interleukin or MMP expression, and the inhibition of MAPKs. Data in Table 2 show that most senomorphics modulate the senescent phenotypes to disrupt the proinflammatory nature of senescent cells.
Table 2. Senomorphics that block SASP components at the secreted level only (measured by ELISA, antibody arrays, and multiplex array). Arrows mean that the secretion of all written factors is decreased upon treatment with the corresponding compound, compared to untreated senescent cells.
Table 2. Senomorphics that block SASP components at the secreted level only (measured by ELISA, antibody arrays, and multiplex array). Arrows mean that the secretion of all written factors is decreased upon treatment with the corresponding compound, compared to untreated senescent cells.
CompoundFunctionCell TypeInducerEffect on SASP FactorsReferences
Adalimumab
(monoclonal antibody)
TNFα inhibitorHUVECsReplicative senescenceIL-6 ↓[178]
Anti-ephrin B2 antibody
(clone B11)
Ephrin B2 inhibitorHuman fibroblastsChemical-induced senescence
Irradiation-induced senescence
IL-6 ↓[179]
Apigenin (flavonoid)NF-κB inhibitorBJ fibroblastsBleomycin-induced senescenceIL-6; IL-8; IL-1β ↓[183]
Avenanthramicine CAMPK activator
p38/NF-κB inhibitor
Human fibroblasts (HDFs)Replicative SenescenceIL-6; IL-8; TGF-β ↓[184]
BIRB796p38 inhibitorHuman fibroblasts (NHDFs)Replicative SenescenceIL-6 ↓[185]
Cortisol/corticosteroneGlucocorticoidsHuman fibroblasts (HCA2)X-irradiation induced senescenceIL-6 ↓[186]
Hydroxytyrosol
(olive phenolic compound)
NF-κB inhibitorHuman fibroblasts (NHDFs
, MRC5)
Replicative senescenceIL-6; MMP-2; MMP-9 ↓[187]
IPI-504HSP90 inhibitorARPE-19H2O2-induced senescenceIL1-β; IL-8 ↓[188]
Isatis tinctoria L. Leaf extract (ITE)mTOR/MAPK/NF-κB inhibitorHuman fibroblasts (HDFs)Replicative SenescenceIL-6; IL1-β; IL-8 ↓[189]
Kaempferol (flavonoid)NF-kB inhibitorBJ fibroblastsBleomycin-induced senescenceIL-6; IL-8; IL-1β ↓[183]
LamivudineNucleoside reverse transcriptase inhibitorHuman fibroblastsReplicative SenescenceIFN-1 ↓[190]
MetforminSeveral pathwaysHuman HNSCC cell line Cal27LY2835219 (CDK4/6 inhibitor)- induced senescenceNT3; MCP-1; IL-6; IL-8; GRO; IGFBP1; BMP4; BLC ↓[191]
MetforminSeveral pathwaysPrimary VSMCs from the aortas of elderly patientsAng II-induced premature senescenceMMP-2; IL-6; TGFβ ↓[192]
Mix of bioCurcumin, Polydatin and liposomal-b-caryophylleneSeveral pathwaysHUVECsReplicative Senescence
Doxorubine-induced senescence
IL-6; IL-1β ↓[193]
MK2.IIIMK2 kinase inhibitorHuman fibroblasts (NHDFs)Replicative SenescenceIL-6 ↓[185]
Oleuropein
(olive phenolic compound)
NF-κB inhibitorHuman fibroblasts (NHDFs)
MRC5
Replicative senescence
γ-irradiation-induced senescence
IL-6; MMP-2; MMP-9↓
IL-6; IL-8; MCP-1; RANTES ↓
[187,194]
SimvastatinHMG-CoA reductase inhibitorNormal Human Fibroblasts (HCA2)γ-irradiation-induced senescenceIL-6 ↓[195]
RapamycinmTOR inhibitorNormal Human Fibroblasts (HCA2)γ-irradiation-induced senescenceIL-6; CSF2; CCL7; CCL8; IGF1; TGFB3; IL-8; BMP4; IL-10 ↓[196]
RapamycinmTOR inhibitorMurine MEFsH2O2-induced senescenceTNFα; LIX; Leptin R; MIP-1α[197]
ResveratrolSIRT1 activator
NF-κB inhibitor
NRF2 activator
Arterial VSMCs derived from aged rhesus monkeysChronological ageMCP-1; TNFα, VEGF[198]
RuxolitinibJAK1/2 inhibitorPreadipocytes from healthy human kidney transplant donorsγ-irradiation-induced senescence
Replicative senescence
IL-6; GM-CSF; G-CSF; IL-10; CXCL-1; MIP-1a; IL-8, MCP-1; RANTES, MCP-3; PAI-1; MIP-1β; TNFα; IFN-α2; IL-1α; VEGF; CCL-11; PDGF-AA
IL-6; IL-8; MCP-1; PAI-1 ↓
[83]
SB203580p38 inhibitorHuman fibroblasts (NHDFs)
Normal Human Fibroblasts (HCA2)
Normal Human Fibroblasts (HCA2)
Replicative Senescence
γ-irradiation-induced senescence
Ras-induced senescence
IL-6 ↓
GRO; IL-6; IL-8; MCP-2; MCP-1; GCP-2; GM-CSF; IL-10; GDNF; IGFBP4, CNTF; GROα; TGF-β1, Angiogenin; IL-2; Eotaxin; IL-7; MIG; IL-1α; TNFα; IL-5; TNFβ; Sgp130; Osteoprotegerin
IL-6; IL-8; GM-CSF ↓
GM-CSF; IL-6; GRO; MIP-1α; IL-1β; ENA78; GROα; IL-8; MCP-3; HGF; ICAM3; MIP-1β; uPAR; Dtk; IGF-1SR; IL-1α; Sgp130; IL-12 p40; IL-4; TIMP1; IL-11; PIGF; IL-15; IL-2; RANTES; IL-2 Rα; Oncostatin M; GDNF; MIP-3α; IL-12 p70; Thrombopoietin
[89,185]
Silybum marianum flower extract (SMFE)UnknownHuman fibroblasts (HDFs)Replicative SenescenceIL-6; MMP-1 ↓[199]
SR9009Reduces ROS level via the activation of the NRF2 pathwayHuman fibroblasts (HDFs)Doxorubicin-induced senescenceIL-1α; IL-1β ↓[200]
UR-13756p38 inhibitorHuman fibroblasts (NHDFs)Replicative SenescenceIL-6 ↓[185]
Wogonin (flavonoid)NF-κB inhibitorBJ fibroblastsBleomycin-induced senenscenIL-6; IL-8; IL-1β ↓[183]
Zileuton5-LO inhibitorHuman fibroblasts (HDFs)γ-irradiation-induced senescenceIL-6 ↓[201]
Most studies, however, have only assessed a few SASP major factors (such as IL-6, IL-1β, and MMPs) following senomorphic treatments, which is not representative of SASP as a whole. Moreover, the impact of senomorphics on the secretion of ECM components, microvesicles, and complex lipids remains largely unexplored. Senomorphics can act on multiple targets depending on the context, the nature, and the model of senescence. In some cases, we cannot rule out that they might even increase the secretion of some detrimental factors. This raises the concern that SASP resulting from senomorphic treatment should probably be less deleterious and should be considered as modified rather than non-senescent-like. In addition, few studies are using CM from senescent cells treated with senomorphics to examine the biological effects of the modified SASP (such as the pro-tumoral impact or differentiation) on other cell types. A study demonstrated that CM from senescent HUVECs treated with anti-TNFα reduced the migration and mammosphere formation of MCF7 cells compared to the CM obtained from untreated senescent HUVECs [178]. Other authors showed that CM from senescent preadipocytes treated with a JAK inhibitor (ruxolitinib) prevented macrophage migration, reduced inflammation in non-senescent preadipocytes [83], and attenuated osteoclast differentiation [202]. CM collected from simvastatin-treated senescent fibroblasts suppressed breast cancer cell proliferation by reducing the phosphorylation of ERK1/2 [195]. Furthermore, CM of senescent cancer cells treated with metformin attenuated the sphere-forming ability of growing cancer cells [191]. Metformin has also been shown to reverse the decrease in the migration of senescent VSMCs [192]. Therefore, in the absence of more extensive data, it is difficult to assess the real effectiveness of senomorphics on SASP.

7. Conclusions and Future Directions

Despite the efforts to characterize the senescence phenotype and the composition of SASPs, understanding the biological effects at a given time for a given cell type and senescence inducer remains challenging. Senescent cells are very heterogeneous, therefore, comparing the effects and status of senescence between senescent cells is a complex operation. Indeed, senescence evolves based on the microenvironment and time elapsed. This modulation suggests that the factors responsible for initiating senescence may not be the same as those involved in maintaining the senescent phenotype. In other words, the “initial/early” senescence is not the same as the “terminal/late” senescence. Once senescence is reached, senescent cells can correct some characteristics of their phenotype via the microenvironmental context. This indicated that the tissue’s physiological and pathological context contributes to the extent, intensity, and regulation of senescence. In turn, the new information received can prompt senescent cells to adapt and produce an SASP that is more attuned to the context, meaning that the communication of the senescent cells is not unidirectional (Figure 2). This gives rise to a large variety of SASPs, and the different combinations of SASP components could generate distinct « barcodes » that may dictate SASP-mediated responses. For this reason, machine learning might be a promising tool to help the better characterization of SASPs [203].
Although efforts have been made to understand the effects of secreted factors, particularly, pro-inflammatory factors, little is known about the effects of changes in the ECM as well as direct cell-to-cell contacts. Secreted factors are only one of the means by which cells communicate with their matrix and cellular microenvironment [13]. Moreover, most studies on senescence have been carried out on fibroblasts, a model that, in essence, does not give rise to direct intercellular communication. Furthermore, the precise role of each class of secreted molecules in mediating the biological effects of SASP remains unclear. While the pro-inflammatory portion is known to be related to the pro-oncogenic effects of SASP, the role of secreted matrix proteins is not well understood. Models of decellularized matrices from senescent cells could help to fill this gap in our knowledge.
Beyond the fact that SASP may evolve and that we lack knowledge on temporal aspects, data on the transcriptomic, proteomic, and secretomic concordance are necessary. There are limited secretomic data available to support conclusions about the effects of SASP. In addition, co-culture models could be of great interest for modeling cellular interactions and the evolution of SASP, as the presence of other cell types can influence the secretome of senescent cells, either by slowing it down or stimulating its production. Several studies have attempted to define a core SASP, which is very useful to understand the common aspects following different senescence inducers or cell types. However, from a therapeutic point of view, it would be more interesting to understand what differentiates two cell types that have undergone the same senescence inducer. As an example, when two SASPs have a 70% similarity, it raises questions about the potential impact of the remaining 30% difference. This highlights the potential for the selective use of SASP blockers. Regarding SASP blocking molecules, their senomorphic properties have been essentially based on their ability to inhibit the transcription of SASP genes. However, concordance between transcriptomics, proteomics, and secretomics has not been demonstrated for all SASP genes. Further experiments are needed to validate this concordance as well as data using CM from senescent cells treated with senomorphics.
In conclusion, there is still much to be discovered regarding how SASP communicates with its environment and network, and it is only through multidisciplinary approaches that we can better understand and decode them. Finally, it is still challenging to critically evaluate the use of SASP in vivo, as modulating its components has demonstrated limited reliability on human health. To be continued in the next “What SASP” discussion.

Funding

This work was supported by the Centre National de la Recherche Scientifique, the Institut National de la Santé et de la Recherche Médicale, the Université de Lille, the Ligue contre le Cancer (Comité du Nord), the Institut Pasteur de Lille, and the Contrat Plan État Région—CTRL (Centre Transdisciplinaire de Recherche sur la Longévité) and the Contrat de Plan Etat Region CPER Cancer 2015–2020 to O.P. JG is a recipient of a co-funded FSR fellowship (UNamur) and doctoral fellowship (Univ Lille), IB is a recipient of an FSR fellowship (UNamur). FDC is a research associate at Fonds de la Recherche Scientifique-FNRS.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The data presented in this study are available in the article.

Conflicts of Interest

The authors declare that they have no competing interest.

References

  1. Roger, L.; Tomas, F.; Gire, V. Mechanisms and Regulation of Cellular Senescence. Int. J. Mol. Sci. 2021, 22, 13173. [Google Scholar] [CrossRef] [PubMed]
  2. Niklander, S.E.; Lambert, D.W.; Hunter, K.D. Senescent Cells in Cancer: Wanted or Unwanted Citizens. Cells 2021, 10, 3315. [Google Scholar] [CrossRef] [PubMed]
  3. Hernandez-Segura, A.; Nehme, J.; Demaria, M. Hallmarks of Cellular Senescence. Trends Cell Biol. 2018, 28, 436–453. [Google Scholar] [CrossRef] [PubMed]
  4. Abbadie, C.; Pluquet, O.; Pourtier, A. Epithelial Cell Senescence: An Adaptive Response to Pre-Carcinogenic Stresses? Cell. Mol. Life Sci. 2017, 74, 4471–4509. [Google Scholar] [CrossRef]
  5. Toussaint, O.; Medrano, E.E.; Von Zglinicki, T. Cellular and Molecular Mechanisms of Stress-Induced Premature Senescence (SIPS) of Human Diploid Fibroblasts and Melanocytes. Exp. Gerontol. 2000, 35, 927–945. [Google Scholar] [CrossRef]
  6. Toutfaire, M.; Bauwens, E.; Debacq-Chainiaux, F. The Impact of Cellular Senescence in Skin Ageing: A Notion of Mosaic and Therapeutic Strategies. Biochem. Pharmacol. 2017, 142, 1–12. [Google Scholar] [CrossRef]
  7. Malaquin, N.; Martinez, A.; Rodier, F. Keeping the Senescence Secretome under Control: Molecular Reins on the Senescence-Associated Secretory Phenotype. Exp. Gerontol. 2016, 82, 39–49. [Google Scholar] [CrossRef]
  8. Nacarelli, T.; Liu, P.; Zhang, R. Epigenetic Basis of Cellular Senescence and Its Implications in Aging. Genes 2017, 8, 343. [Google Scholar] [CrossRef] [Green Version]
  9. Coppé, J.P.; Patil, C.K.; Rodier, F.; Sun, Y.; Muñoz, D.P.; Goldstein, J.; Nelson, P.S.; Desprez, P.Y.; Campisi, J. Senescence-Associated Secretory Phenotypes Reveal Cell-Nonautonomous Functions of Oncogenic RAS and the P53 Tumor Suppressor. PLoS Biol. 2008, 6, e301. [Google Scholar] [CrossRef]
  10. Birch, J.; Gil, J. Senescence and the SASP: Many Therapeutic Avenues. Genes Dev. 2020, 34, 1565–1576. [Google Scholar] [CrossRef]
  11. Campisi, J.; Andersen, J.K.; Kapahi, P.; Melov, S. Cellular Senescence: A Link between Cancer and Age-Related Degenerative Disease? Semin. Cancer Biol. 2011, 21, 354. [Google Scholar] [CrossRef] [Green Version]
  12. Yoshimoto, S.; Loo, T.M.; Atarashi, K.; Kanda, H.; Sato, S.; Oyadomari, S.; Iwakura, Y.; Oshima, K.; Morita, H.; Hattori, M.; et al. Obesity-Induced Gut Microbial Metabolite Promotes Liver Cancer through Senescence Secretome. Nature 2013, 499, 97–101. [Google Scholar] [CrossRef]
  13. Krtolica, A.; Parrinello, S.; Lockett, S.; Desprez, P.Y.; Campisi, J. Senescent Fibroblasts Promote Epithelial Cell Growth and Tumorigenesis: A Link between Cancer and Aging. Proc. Natl. Acad. Sci. USA 2001, 98, 12072–12077. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Muñoz-Espín, D.; Cañamero, M.; Maraver, A.; Gómez-López, G.; Contreras, J.; Murillo-Cuesta, S.; Rodríguez-Baeza, A.; Varela-Nieto, I.; Ruberte, J.; Collado, M.; et al. Programmed Cell Senescence during Mammalian Embryonic Development. Cell 2013, 155, 1104. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Demaria, M.; Ohtani, N.; Youssef, S.A.; Rodier, F.; Toussaint, W.; Mitchell, J.R.; Laberge, R.M.; Vijg, J.; VanSteeg, H.; Dollé, M.E.T.; et al. An Essential Role for Senescent Cells in Optimal Wound Healing through Secretion of PDGF-AA. Dev. Cell 2014, 31, 722–733. [Google Scholar] [CrossRef] [Green Version]
  16. Hayflick, L.; Moorhead, P.S. The Serial Cultivation of Human Diploid Cell Strains. Exp. Cell Res. 1961, 25, 585–621. [Google Scholar] [CrossRef]
  17. Maciejowski, J.; De Lange, T. Telomeres in Cancer: Tumour Suppression and Genome Instability. Nat. Rev. Mol. Cell Biol. 2017, 18, 175–186. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Gorgoulis, V.G.; Halazonetis, T.D. Oncogene-Induced Senescence: The Bright and Dark Side of the Response. Curr. Opin. Cell Biol. 2010, 22, 816–827. [Google Scholar] [CrossRef]
  19. Ott, C.; Jung, T.; Grune, T.; Höhn, A. SIPS as a Model to Study Age-Related Changes in Proteolysis and Aggregate Formation. Mech. Ageing Dev. 2018, 170, 72–81. [Google Scholar] [CrossRef]
  20. Pazolli, E.; Alspach, E.; Milczarek, A.; Prior, J.; Piwnica-Worms, D.; Stewart, S.A. Chromatin Remodeling Underlies the Senescence- Associated Secretory Phenotype of Tumor Stromal Fibroblasts That Supports Cancer Progression. Cancer Res. 2012, 72, 2251–2261. [Google Scholar] [CrossRef] [Green Version]
  21. 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. [Google Scholar] [CrossRef] [Green Version]
  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. Fujimaki, K.; Yao, G. Cell Dormancy Plasticity: Quiescence Deepens into Senescence through a Dimmer Switch. Physiol. Genomics 2020, 52, 558–562. [Google Scholar] [CrossRef]
  24. Sen, P.; Shah, P.P.; Nativio, R.; Berger, S.L. Epigenetic Mechanisms of Longevity and Aging. Cell 2016, 166, 822–839. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. O’Sullivan, R.J.; Kubicek, S.; Schreiber, S.L.; Karlseder, J. Reduced Histone Biosynthesis and Chromatin Changes Arising from a Damage Signal at Telomeres. Nat. Struct. Mol. Biol. 2010, 17, 1218–1225. [Google Scholar] [CrossRef] [Green Version]
  26. Rai, T.S.; Cole, J.J.; Nelson, D.M.; Dikovskaya, D.; Faller, W.J.; Vizioli, M.G.; Hewitt, R.N.; Anannya, O.; McBryan, T.; Manoharan, I.; et al. HIRA Orchestrates a Dynamic Chromatin Landscape in Senescence and Is Required for Suppression of Neoplasia. Genes Dev. 2014, 28, 2712–2725. [Google Scholar] [CrossRef] [Green Version]
  27. Schmeer, C.; Kretz, A.; Wengerodt, D.; Stojiljkovic, M.; Witte, O.W. Dissecting Aging and Senescence—Current Concepts and Open Lessons. Cells 2019, 8, 1446. [Google Scholar] [CrossRef] [Green Version]
  28. Sidler, C.; Kovalchuk, O.; Kovalchuk, I. Epigenetic Regulation of Cellular Senescence and Aging. Front. Genet. 2017, 8, 138. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. Rodier, F.; Coppé, J.P.; 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 Signaling Triggers Senescence-Associated Inflammatory Cytokine Secretion. Nat. Cell Biol. 2009, 11, 973. [Google Scholar] [CrossRef] [PubMed]
  30. Wiley, C.D.; Campisi, J. The Metabolic Roots of Senescence: Mechanisms and Opportunities for Intervention. Nat. Metab. 2021, 3, 1290–1301. [Google Scholar] [CrossRef]
  31. Childs, B.G.; Baker, D.J.; Kirkland, J.L.; Campisi, J.; Deursen, J.M. Senescence and Apoptosis: Dueling or Complementary Cell Fates? EMBO Rep. 2014, 15, 1139–1153. [Google Scholar] [CrossRef] [Green Version]
  32. Quaas, M.; Müller, G.A.; Engeland, K. P53 Can Repress Transcription of Cell Cycle Genes through a P21 WAF1/CIP1-Dependent Switch from MMB to DREAM Protein Complex Binding at CHR Promoter Elements. Cell Cycle 2012, 11, 4661–4672. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Kumari, R.; Jat, P. Mechanisms of Cellular Senescence: Cell Cycle Arrest and Senescence Associated Secretory Phenotype. Front. Cell Dev. Biol. 2021, 9, 485. [Google Scholar] [CrossRef] [PubMed]
  34. Halkoum, R.; Salnot, V.; Capallere, C.; Plaza, C.; L’honoré, A.; Pays, K.; Friguet, B.; Nizard, C.; Petropoulos, I. Glyoxal Induces Senescence in Human Keratinocytes through Oxidative Stress and Activation of the Protein Kinase B/FOXO3a/P27KIP1 Pathway. J. Investig. Dermatol. 2022, 142, 2068–2078.e7. [Google Scholar] [CrossRef] [PubMed]
  35. McDermott, M.S.J.; Conlon, N.; Browne, B.C.; Szabo, A.; Synnott, N.C.; O’brien, N.A.; Duffy, M.J.; Crown, J.; O’donovan, N. HER2-Targeted Tyrosine Kinase Inhibitors Cause Therapy-Induced-Senescence in Breast Cancer Cells. Cancers 2019, 11, 197. [Google Scholar] [CrossRef] [Green Version]
  36. Mirzakhani, K.; Kallenbach, J.; Rasa, S.M.M.; Ribaudo, F.; Ungelenk, M.; Ehsani, M.; Gong, W.; Gassler, N.; Leeder, M.; Grimm, M.O.; et al. The Androgen Receptor—LncRNASAT1-AKT-P15 Axis Mediates Androgen-Induced Cellular Senescence in Prostate Cancer Cells. Oncogene 2021, 41, 943–959. [Google Scholar] [CrossRef]
  37. Saleh, T.; Tyutyunyk-Massey, L.; Gewirtz, D.A. Tumor Cell Escape from Therapy-Induced Senescence as a Model of Disease Recurrence after Dormancy. Cancer Res. 2019, 79, 1044–1046. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Martínez-Zamudio, R.I.; Stefa, A.; Nabuco Leva Ferreira Freitas, J.A.; Vasilopoulos, T.; Simpson, M.; Doré, G.; Roux, P.F.; Galan, M.A.; Chokshi, R.J.; Bischof, O.; et al. Escape from Oncogene-Induced Senescence Is Controlled by POU2F2 and Memorized by Chromatin Scars. Cell Genom. 2023, 3, 100293. [Google Scholar] [CrossRef]
  39. Althubiti, M.; Lezina, L.; Carrera, S.; Jukes-Jones, R.; Giblett, S.M.; Antonov, A.; Barlev, N.; Saldanha, G.S.; Pritchard, C.A.; Cain, K.; et al. Characterization of Novel Markers of Senescence and Their Prognostic Potential in Cancer. Cell Death Dis. 2014, 5, e1528. [Google Scholar] [CrossRef] [Green Version]
  40. Kim, K.M.; Noh, J.H.; Bodogai, M.; Martindale, J.L.; Pandey, P.R.; Yang, X.; Biragyn, A.; Abdelmohsen, K.; Gorospe, M. SCAMP4 Enhances the Senescent Cell Secretome. Genes Dev. 2018, 32, 909–914. [Google Scholar] [CrossRef]
  41. Amor, C.; Feucht, J.; Leibold, J.; Ho, Y.-J.; Zhu, C.; Alonso-Curbelo, D.; Mansilla-Soto, J.; Boyer, J.A.; Li, X.; Giavridis, T.; et al. Senolytic CAR T Cells Reverse Senescence-Associated Pathologies. Nature 2020, 583, 127. [Google Scholar] [CrossRef] [PubMed]
  42. Poblocka, M.; Bassey, A.L.; Smith, V.M.; Falcicchio, M.; Manso, A.S.; Althubiti, M.; Sheng, X.B.; Kyle, A.; Barber, R.; Frigerio, M.; et al. Targeted Clearance of Senescent Cells Using an Antibody-Drug Conjugate against a Specific Membrane Marker. Sci. Rep. 2021, 11, 20358. [Google Scholar] [CrossRef] [PubMed]
  43. Rossi, M.; Abdelmohsen, K. The Emergence of Senescent Surface Biomarkers as Senotherapeutic Targets. Cells 2021, 10, 1740. [Google Scholar] [CrossRef] [PubMed]
  44. Freund, A.; Laberge, R.M.; Demaria, M.; Campisi, J. Lamin B1 Loss Is a Senescence-Associated Biomarker. Mol. Biol. Cell 2012, 23, 2066. [Google Scholar] [CrossRef]
  45. Pathak, R.U.; Soujanya, M.; Mishra, R.K. Deterioration of Nuclear Morphology and Architecture: A Hallmark of Senescence and Aging. Ageing Res. Rev. 2021, 67, 101264. [Google Scholar] [CrossRef]
  46. Heckenbach, I.; Mkrtchyan, G.V.; Ben Ezra, M.; Bakula, D.; Sture Madsen, J.; Hasle Nielsen, M.; Oró, D.; Osborne, B.; Covarrubias, A.J.; Laura Idda, M.; et al. Nuclear Morphology Is a Deep Learning Biomarker of Cellular Senescence. Nat. Aging 2022, 2, 742–755. [Google Scholar] [CrossRef]
  47. Abbadie, C.; Pluquet, O. Unfolded Protein Response (UPR) Controls Major Senescence Hallmarks. Trends Biochem. Sci. 2020, 45, 371–374. [Google Scholar] [CrossRef]
  48. Kim, S.Y.; Yang, E.J.; Lee, S.B.; Lee, Y.S.; Cho, K.A.; Park, S.C. Global Transcriptional Downregulation of TREX and Nuclear Trafficking Machinery as Pan-Senescence Phenomena: Evidence from Human Cells and Tissues. Exp. Mol. Med. 2020, 52, 1351–1359. [Google Scholar] [CrossRef]
  49. Park, S.S.; Choi, Y.W.; Kim, J.H.; Kim, H.S.; Park, T.J. Senescent Tumor Cells: An Overlooked Adversary in the Battle against Cancer. Exp. Mol. Med. 2021, 53, 1834–1841. [Google Scholar] [CrossRef] [PubMed]
  50. Kaufman, R.J. Stress Signaling from the Lumen of the Endoplasmic Reticulum: Coordination of Gene Transcriptional and Translational Controls. Genes Dev. 1999, 13, 1211–1233. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  51. Pluquet, O.; Pourtier, A.; Abbadie, C. The Unfolded Protein Response and Cellular Senescence. A Review in the Theme: Cellular Mechanisms of Endoplasmic Reticulum Stress Signaling in Health and Disease. Am. J. Physiol. Cell Physiol. 2015, 308, 415–425. [Google Scholar] [CrossRef] [Green Version]
  52. Druelle, C.; Drullion, C.; Deslé, J.; Martin, N.; Saas, L.; Cormenier, J.; Malaquin, N.; Huot, L.; Slomianny, C.; Bouali, F.; et al. ATF6α Regulates Morphological Changes Associated with Senescence in Human Fibroblasts. Oncotarget 2016, 7, 67699. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Kim, H.S.; Kim, Y.; Lim, M.J.; Park, Y.-G.; Park, S.I.; Sohn, J. The P38-Activated ER Stress-ATF6α Axis Mediates Cellular Senescence. FASEB J. 2019, 33, 2422–2434. [Google Scholar] [CrossRef] [Green Version]
  54. Wang, J.; Qi, Q.; Zhou, W.; Feng, Z.; Huang, B.; Chen, A.; Zhang, D.; Li, W.; Zhang, Q.; Jiang, Z.; et al. Inhibition of Glioma Growth by Flavokawain B Is Mediated through Endoplasmic Reticulum Stress Induced Autophagy. Autophagy 2018, 14, 2007–2022. [Google Scholar] [CrossRef] [Green Version]
  55. Dörr, J.R.; Yu, Y.; Milanovic, M.; Beuster, G.; Zasada, C.; Däbritz, J.H.M.; Lisec, J.; Lenze, D.; Gerhardt, A.; Schleicher, K.; et al. Synthetic Lethal Metabolic Targeting of Cellular Senescence in Cancer Therapy. Nature 2013, 501, 421–425. [Google Scholar] [CrossRef]
  56. Chen, H.; Ruiz, P.D.; McKimpson, W.M.; Novikov, L.; Kitsis, R.N.; Gamble, M.J. MacroH2A1 and ATM Play Opposing Roles in Paracrine Senescence and the Senescence-Associated Secretory Phenotype. Mol. Cell 2015, 59, 719–731. [Google Scholar] [CrossRef] [Green Version]
  57. Lu, S.; Song, Y.; Luo, R.; Li, S.; Li, G.; Wang, K.; Liao, Z.; Wang, B.; Ke, W.; Xiang, Q.; et al. Ferroportin-Dependent Iron Homeostasis Protects against Oxidative Stress-Induced Nucleus Pulposus Cell Ferroptosis and Ameliorates Intervertebral Disc Degeneration In Vivo. Oxid. Med. Cell. Longev. 2021, 2021, 6670497. [Google Scholar] [CrossRef]
  58. Tan, Q.; Liang, N.; Zhang, X.; Li, J. Dynamic Aging: Channeled Through Microenvironment. Front. Physiol. 2021, 12, 702276. [Google Scholar] [CrossRef] [PubMed]
  59. Matos, L.; Gouveia, A.M.; Almeida, H. ER Stress Response in Human Cellular Models of Senescence. J. Gerontol. Ser. A 2015, 70, 924–935. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Basisty, N.; Kale, A.; Jeon, O.H.; Kuehnemann, C.; Payne, T.; Rao, C.; Holtz, A.; Shah, S.; Sharma, V.; Ferrucci, L.; et al. A Proteomic Atlas of Senescence-Associated Secretomes for Aging Biomarker Development. PLoS Biol. 2020, 18, e3000599. [Google Scholar] [CrossRef] [Green Version]
  61. Wiley, C.D.; Liu, S.; Limbad, C.; Zawadzka, A.M.; Beck, J.; Demaria, M.; Artwood, R.; Alimirah, F.; Lopez-Dominguez, J.A.; Kuehnemann, C.; et al. SILAC Analysis Reveals Increased Secretion of Hemostasis-Related Factors by Senescent Cells. Cell Rep. 2019, 28, 3329–3337.e5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Ni, C.; Narzt, M.S.; Nagelreiter, I.M.; Zhang, C.F.; Larue, L.; Rossiter, H.; Grillari, J.; Tschachler, E.; Gruber, F. Autophagy Deficient Melanocytes Display a Senescence Associated Secretory Phenotype That Includes Oxidized Lipid Mediators. Int. J. Biochem. Cell Biol. 2016, 81, 375–382. [Google Scholar] [CrossRef] [PubMed]
  63. Narzt, M.S.; Pils, V.; Kremslehner, C.; Nagelreiter, I.M.; Schosserer, M.; Bessonova, E.; Bayer, A.; Reifschneider, R.; Terlecki-Zaniewicz, L.; Waidhofer-Söllner, P.; et al. Epilipidomics of Senescent Dermal Fibroblasts Identify Lysophosphatidylcholines as Pleiotropic Senescence-Associated Secretory Phenotype (SASP) Factors. J. Investig. Dermatol. 2021, 141, 993–1006.e15. [Google Scholar] [CrossRef]
  64. Wiley, C.D.; Sharma, R.; Davis, S.S.; Lopez-Dominguez, J.A.; Mitchell, K.P.; Wiley, S.; Alimirah, F.; Kim, D.E.; Payne, T.; Rosko, A.; et al. Oxylipin Biosynthesis Reinforces Cellular Senescence and Allows Detection of Senolysis. Cell Metab. 2021, 33, 1124–1136.e5. [Google Scholar] [CrossRef] [PubMed]
  65. Hattori, H.; Takaoka, K.; Ueta, M.; Oshitani, M.; Tamaoka, J.; Noguchi, K.; Kishimoto, H. Senescent RAW264.7 Cells Exhibit Increased Production of Nitric Oxide and Release Inducible Nitric Oxide Synthase in Exosomes. Mol. Med. Rep. 2021, 24, 1–9. [Google Scholar] [CrossRef]
  66. Takasugi, M. Emerging Roles of Extracellular Vesicles in Cellular Senescence and Aging. Aging Cell 2018, 17, e12734. [Google Scholar] [CrossRef] [Green Version]
  67. Abels, E.R.; Breakefield, X.O. Introduction to Extracellular Vesicles: Biogenesis, RNA Cargo Selection, Content, Release, and Uptake. Cell. Mol. Neurobiol. 2016, 36, 301–312. [Google Scholar] [CrossRef] [Green Version]
  68. Wallis, R.; Mizen, H.; Bishop, C.L. The Bright and Dark Side of Extracellular Vesicles in the Senescence-Associated Secretory Phenotype. Mech. Ageing Dev. 2020, 189, 111263. [Google Scholar] [CrossRef]
  69. Lee, A.H.; Ghosh, D.; Koh, I.L.; Dawson, M.R. Senescence-Associated Exosomes Transfer MiRNA-Induced Fibrosis to Neighboring Cells. Aging 2023, 15, 1237–1256. [Google Scholar] [CrossRef]
  70. Bauwens, E.; Parée, T.; Meurant, S.; Bouriez, I.; Hannart, C.; Wéra, A.-C.; Khelfi, A.; Fattaccioli, A.; Burteau, S.; Demazy, C.; et al. Senescence Induced by UVB in Keratinocytes Impairs Amino Acids Balance. J. Investig. Dermatol. 2022, 143, 554–565.e9. [Google Scholar] [CrossRef]
  71. Valerio, H.P.; Ravagnani, F.G.; Ronsein, G.E.; Di Mascio, P. A Single Dose of Ultraviolet-A Induces Proteome Remodeling and Senescence in Primary Human Keratinocytes. Sci. Rep. 2021, 11, 23355. [Google Scholar] [CrossRef] [PubMed]
  72. Özcan, S.; Alessio, N.; Acar, M.B.; Mert, E.; Omerli, F.; Peluso, G.; Galderisi, U. Unbiased Analysis of Senescence Associated Secretory Phenotype (SASP) to Identify Common Components Following Different Genotoxic Stresses. Aging 2016, 8, 1316. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. 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. [Google Scholar] [CrossRef] [Green Version]
  74. Kim, Y.M.; Byun, H.O.; Jee, B.A.; Cho, H.; Seo, Y.H.; Kim, Y.S.; Park, M.H.; Chung, H.Y.; Woo, H.G.; Yoon, G. Implications of Time-Series Gene Expression Profiles of Replicative Senescence. Aging Cell 2013, 12, 622–634. [Google Scholar] [CrossRef]
  75. Yao, X.; Li, H.; Chen, L.; Tan, L.P. UV-Induced Senescence of Human Dermal Fibroblasts Restrained by Low-Stiffness Matrix by Inhibiting NF-ΚB Activation. Eng. Regen. 2022, 3, 365–373. [Google Scholar] [CrossRef]
  76. Toutfaire, M.; Dumortier, E.; Fattaccioli, A.; Van Steenbrugge, M.; Proby, C.M.; Debacq-Chainiaux, F. Unraveling the Interplay between Senescent Dermal Fibroblasts and Cutaneous Squamous Cell Carcinoma Cell Lines at Different Stages of Tumorigenesis. Int. J. Biochem. Cell Biol. 2018, 98, 113–126. [Google Scholar] [CrossRef] [Green Version]
  77. Malaquin, N.; Olivier, M.; Martinez, A.; Nadeau, S.; Sawchyn, C.; Coppé, J.; Cardin, G.; Mallette, F.A.; Campisi, J.; Rodier, F. Non-canonical ATM/MRN Activities Temporally Define the Senescence Secretory Program. EMBO Rep. 2020, 21, e50718. [Google Scholar] [CrossRef]
  78. Buj, R.; Leon, K.E.; Anguelov, M.A.; Aird, K.M. Suppression of P16 Alleviates the Senescence-Associated Secretory Phenotype. Aging 2021, 13, 3290–3312. [Google Scholar] [CrossRef] [PubMed]
  79. Acosta, J.C.; O’Loghlen, A.; Banito, A.; Guijarro, M.V.; Augert, A.; Raguz, S.; Fumagalli, M.; Da Costa, M.; Brown, C.; Popov, N.; et al. Chemokine Signaling via the CXCR2 Receptor Reinforces Senescence. Cell 2008, 133, 1006–1018. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  80. Kang, C.; Xu, Q.; Martin, T.D.; Li, M.Z.; Demaria, M.; Aron, L.; Lu, T.; Yankner, B.A.; Campisi, J.; Elledge, S.J. The DNA Damage Response Induces Inflammation and Senescence by Inhibiting Autophagy of GATA4. Science 2015, 349, aaa5612. [Google Scholar] [CrossRef] [Green Version]
  81. Hoare, M.; Ito, Y.; Kang, T.W.; Weekes, M.P.; Matheson, N.J.; Patten, D.A.; Shetty, S.; Parry, A.J.; Menon, S.; Salama, R.; et al. NOTCH1 Mediates a Switch between Two Distinct Secretomes during Senescence. Nat. Cell Biol. 2016, 18, 979. [Google Scholar] [CrossRef] [Green Version]
  82. Toso, A.; Revandkar, A.; DiMitri, D.; Guccini, I.; Proietti, M.; Sarti, M.; Pinton, S.; Zhang, J.; Kalathur, M.; Civenni, G.; et al. Enhancing Chemotherapy Efficacy in Pten-Deficient Prostate Tumors by Activating the Senescence-Associated Antitumor Immunity. Cell Rep. 2014, 9, 75–89. [Google Scholar] [CrossRef]
  83. Xu, M.; Tchkonia, T.; Ding, H.; Ogrodnik, M.; Lubbers, E.R.; Pirtskhalava, T.; White, T.A.; Johnson, K.O.; Stout, M.B.; Mezera, V.; et al. JAK Inhibition Alleviates the Cellular Senescence-Associated Secretory Phenotype and Frailty in Old Age. Proc. Natl. Acad. Sci. USA 2015, 112, E6301–E6310. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Glück, S.; Guey, B.; Gulen, M.F.; Wolter, K.; Kang, T.W.; Schmacke, N.A.; Bridgeman, A.; Rehwinkel, J.; Zender, L.; Ablasser, A. Innate Immune Sensing of Cytosolic Chromatin Fragments through CGAS Promotes Senescence. Nat. Cell Biol. 2017, 19, 1061–1070. [Google Scholar] [CrossRef] [PubMed]
  85. Dou, Z.; Ghosh, K.; Vizioli, M.G.; Zhu, J.; Sen, P.; Wangensteen, K.J.; Simithy, J.; Lan, Y.; Lin, Y.; Zhou, Z.; et al. Cytoplasmic Chromatin Triggers Inflammation in Senescence and Cancer. Nature 2017, 550, 402–406. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Sadaie, M.; Salama, R.; Carroll, T.; Tomimatsu, K.; Chandra, T.; Young, A.R.J.; Narita, M.; Pérez-Mancera, P.A.; Bennett, D.C.; Chong, H.; et al. Redistribution of the Lamin B1 Genomic Binding Profile Affects Rearrangement of Heterochromatic Domains and SAHF Formation during Senescence. Genes Dev. 2013, 27, 1800–1808. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Gonçalves, S.; Yin, K.; Ito, Y.; Chan, A.; Olan, I.; Gough, S.; Cassidy, L.; Serrao, E.; Smith, S.; Young, A.; et al. COX2 Regulates Senescence Secretome Composition and Senescence Surveillance through PGE2. Cell Rep. 2021, 34, 108860. [Google Scholar] [CrossRef]
  88. Alspach, E.; Flanagan, K.C.; Luo, X.; Ruhland, M.K.; Huang, H.; Pazolli, E.; Donlin, M.J.; Marsh, T.; Piwnica-Worms, D.; Monahan, J.; et al. P38MAPK Plays a Crucial Role in Stromal-Mediated Tumorigenesis. Cancer Discov. 2014, 4, 716–729. [Google Scholar] [CrossRef] [Green Version]
  89. Freund, A.; Patil, C.K.; Campisi, J. P38MAPK Is a Novel DNA Damage Response-Independent Regulator of the Senescence-Associated Secretory Phenotype. EMBO J. 2011, 30, 1536–1548. [Google Scholar] [CrossRef] [Green Version]
  90. Herranz, N.; Gallage, S.; Mellone, M.; Wuestefeld, T.; Klotz, S.; Hanley, C.J.; Raguz, S.; Acosta, J.C.; Innes, A.J.; Banito, A.; et al. MTOR Regulates MAPKAPK2 Translation to Control the Senescence-Associated Secretory Phenotype. Nat. Cell Biol. 2015, 17, 1205–1217. [Google Scholar] [CrossRef] [Green Version]
  91. Machado-Oliveira, G.; Ramos, C.; Marques, A.R.A.; Vieira, O.V. Cell Senescence, Multiple Organelle Dysfunction and Atherosclerosis. Cells 2020, 9, 2146. [Google Scholar] [CrossRef] [PubMed]
  92. Wang, Y.; Liu, L.; Song, Y.; Yu, X.; Deng, H. Unveiling E2F4, TEAD1 and AP-1 as Regulatory Transcription Factors of the Replicative Senescence Program by Multi-Omics Analysis. Protein Cell 2022, 13, 742–759. [Google Scholar] [CrossRef] [PubMed]
  93. Martínez-Zamudio, R.I.; Roux, P.F.; de Freitas, J.A.N.L.F.; Robinson, L.; Doré, G.; Sun, B.; Belenki, D.; Milanovic, M.; Herbig, U.; Schmitt, C.A.; et al. AP-1 Imprints a Reversible Transcriptional Programme of Senescent Cells. Nat. Cell Biol. 2020, 22, 842–855. [Google Scholar] [CrossRef] [PubMed]
  94. 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. [Google Scholar] [CrossRef] [Green Version]
  95. Contrepois, K.; Coudereau, C.; Benayoun, B.A.; Schuler, N.; Roux, P.F.; Bischof, O.; Courbeyrette, R.; Carvalho, C.; Thuret, J.Y.; Ma, Z.; et al. Histone Variant H2A.J Accumulates in Senescent Cells and Promotes Inflammatory Gene Expression. Nat. Commun. 2017, 8, 14995. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Davalos, A.R.; Kawahara, M.; Malhotra, G.K.; Schaum, N.; Huang, J.; Ved, U.; Beausejour, C.M.; Coppe, J.P.; Rodoer, F.; Campisi, J. P53-Dependent Release of Alarmin HMGB1 Is a Central Mediator of Senescent Phenotypes. J. Cell Biol. 2013, 201, 613–629. [Google Scholar] [CrossRef] [Green Version]
  97. Aird, K.M.; Iwasaki, O.; Kossenkov, A.V.; Tanizawa, H.; Fatkhutdinov, N.; Bitler, B.G.; Le, L.; Alicea, G.; Yang, T.L.; Johnson, F.B.; et al. HMGB2 Orchestrates the Chromatin Landscape of Senescence-Associated Secretory Phenotype Gene Loci. J. Cell Biol. 2016, 215, 325–334. [Google Scholar] [CrossRef] [Green Version]
  98. Pluquet, O.; Abbadie, C. Cellular Senescence and Tumor Promotion: Role of the Unfolded Protein Response. Adv. Cancer Res. 2021, 150, 285–334. [Google Scholar] [CrossRef]
  99. Dandekar, A.; Mendez, R.; Zhang, K. Cross Talk between ER Stress, Oxidative Stress, and Inflammation in Health and Disease. Methods Mol. Biol. 2015, 1292, 205–214. [Google Scholar] [CrossRef]
  100. Schmitz, R.; Wright, G.W.; Huang, D.W.; Johnson, C.A.; Phelan, J.D.; Wang, J.Q.; Roulland, S.; Kasbekar, M.; Young, R.M.; Shaffer, A.L.; et al. Genetics and Pathogenesis of Diffuse Large B-Cell Lymphoma. N. Engl. J. Med. 2018, 378, 1396. [Google Scholar] [CrossRef]
  101. Smith, H.L.; Freeman, O.J.; Butcher, A.J.; Holmqvist, S.; Humoud, I.; Schätzl, T.; Hughes, D.T.; Verity, N.C.; Swinden, D.P.; Hayes, J.; et al. Astrocyte Unfolded Protein Response Induces a Specific Reactivity State That Causes Non-Cell-Autonomous Neuronal Degeneration. Neuron 2020, 105, 855–866.e5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Logue, S.E.; McGrath, E.P.; Cleary, P.; Greene, S.; Mnich, K.; Almanza, A.; Chevet, E.; Dwyer, R.M.; Oommen, A.; Legembre, P.; et al. Inhibition of IRE1 RNase Activity Modulates the Tumor Cell Secretome and Enhances Response to Chemotherapy. Nat. Commun. 2018, 9, 3267. [Google Scholar] [CrossRef] [Green Version]
  103. Rubio-Patiño, C.; Bossowski, J.P.; Chevet, E.; Ricci, J.E. Reshaping the Immune Tumor Microenvironment Through IRE1 Signaling. Trends Mol. Med. 2018, 24, 607–614. [Google Scholar] [CrossRef]
  104. Zhu, H.; Chen, X.; Chen, B.; Chen, B.; Song, W.; Sun, D.; Zhao, Y. Activating Transcription Factor 4 Promotes Esophageal Squamous Cell Carcinoma Invasion and Metastasis in Mice and Is Associated with Poor Prognosis in Human Patients. PLoS ONE 2014, 9, e103882. [Google Scholar] [CrossRef] [Green Version]
  105. Despres, J.; Ramdani, Y.; di Giovanni, M.; Bénard, M.; Zahid, A.; Montero-Hadjadje, M.; Yvergnaux, F.; Saguet, T.; Driouich, A.; Follet-Gueye, M.L. Replicative Senescence of Human Dermal Fibroblasts Affects Structural and Functional Aspects of the Golgi Apparatus. Exp. Dermatol. 2019, 28, 922–932. [Google Scholar] [CrossRef]
  106. Cho, J.H.; Saini, D.K.; Karunarathne, W.K.A.; Kalyanaraman, V.; Gautam, N. Alteration of Golgi Structure in Senescent Cells and Its Regulation by a G Protein γ Subunit. Cell. Signal 2011, 23, 785–793. [Google Scholar] [CrossRef] [Green Version]
  107. Udono, M.; Fujii, K.; Harada, G.; Tsuzuki, Y.; Kadooka, K.; Zhang, P.; Fujii, H.; Amano, M.; Nishimura, S.I.; Tashiro, K.; et al. Impaired ATP6V0A2 Expression Contributes to Golgi Dispersion and Glycosylation Changes in Senescent Cells. Sci. Rep. 2015, 5, 6–10. [Google Scholar] [CrossRef] [Green Version]
  108. Yuanyuan, S.; Sun, Z.; Xu, C.; Li, G. Protein Kinase D1-Mediated Classical Protein Secretory Pathway Regulates Oncogene Ras-Induced Senescent Response. Artic. J. Cell Sci. 2018, 131, jcs207217. [Google Scholar] [CrossRef] [Green Version]
  109. Rovira, M.; Sereda, R.; Pladevall-Morera, D.; Ramponi, V.; Marin, I.; Maus, M.; Madrigal-Matute, J.; Díaz, A.; García, F.; Muñoz, J.; et al. The Lysosomal Proteome of Senescent Cells Contributes to the Senescence Secretome. Aging Cell 2022, 21, e13707. [Google Scholar] [CrossRef] [PubMed]
  110. Takahashi, A.; Okada, R.; Nagao, K.; Kawamata, Y.; Hanyu, A.; Yoshimoto, S.; Takasugi, M.; Watanabe, S.; Kanemaki, M.T.; Obuse, C.; et al. ARTICLE Exosomes Maintain Cellular Homeostasis by Excreting Harmful DNA from Cells. Nat. Commun. 2017, 8, 15287. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  111. Ostrowski, M.; Carmo, N.B.; Krumeich, S.; Fanget, I.; Raposo, G.; Savina, A.; Moita, C.F.; Schauer, K.; Hume, A.N.; Freitas, R.P.; et al. Rab27a and Rab27b Control Different Steps of the Exosome Secretion Pathway. Nat. Cell Biol. 2010, 12, 19–30. [Google Scholar] [CrossRef] [Green Version]
  112. Hitomi, K.; Okada, R.; Loo, T.M.; Miyata, K.; Nakamura, A.J.; Takahashi, A. DNA Damage Regulates Senescence-Associated Extracellular Vesicle Release via the Ceramide Pathway to Prevent Excessive Inflammatory Responses. Int. J. Mol. Sci. 2020, 21, 3720. [Google Scholar] [CrossRef]
  113. Misawa, T.; Tanaka, Y.; Okada, R.; Takahashi, A. Biology of Extracellular Vesicles Secreted from Senescent Cells as Senescence-Associated Secretory Phenotype Factors. Geriatr. Gerontol. Int. 2020, 20, 539–546. [Google Scholar] [CrossRef]
  114. Tuttle, C.S.L.; Waaijer, M.E.C.; Slee-Valentijn, M.S.; Stijnen, T.; Westendorp, R.; Maier, A.B. Cellular Senescence and Chronological Age in Various Human Tissues: A Systematic Review and Meta-Analysis. Aging Cell. 2020, 19, e13083. [Google Scholar] [CrossRef] [Green Version]
  115. Song, S.; Lam, E.W.F.; Tchkonia, T.; Kirkland, J.L.; Sun, Y. Senescent Cells: Emerging Targets for Human Aging and Age-Related Diseases. Trends Biochem. Sci. 2020, 45, 578–592. [Google Scholar] [CrossRef] [PubMed]
  116. Kaur, G.; Sundar, I.K.; Rahman, I. P16-3MR: A Novel Model to Study Cellular Senescence in Cigarette Smoke-Induced Lung Injuries. Int. J. Mol. Sci. 2021, 22, 4834. [Google Scholar] [CrossRef] [PubMed]
  117. Baker, D.J.; Wijshake, T.; Tchkonia, T.; Lebrasseur, N.K.; Childs, B.G.; Van De Sluis, B.; Kirkland, J.L.; Van Deursen, J.M. Clearance of P16 Ink4a-Positive Senescent Cells Delays Ageing-Associated Disorders. Nature 2011, 479, 232–236. [Google Scholar] [CrossRef] [Green Version]
  118. Baker, D.J.; Childs, B.G.; Durik, M.; Wijers, M.E.; Sieben, C.J.; Zhong, J.; Saltness, R.A.; Jeganathan, K.B.; Casaclang Verzosa, G.; Pezeshki, A.; et al. Naturally Occurring P16 Ink4a-Positive Cells Shorten Healthy Lifespan. Nature 2016, 530, 184–189. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  119. Chang, J.; Wang, Y.; Shao, L.; Laberge, R.M.; 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]
  120. 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.e16. [Google Scholar] [CrossRef] [Green Version]
  121. Beharka, A.A.; Meydani, M.; Wu, D.; Leka, L.S.; Meydani, A.; Meydani, S.N. Interleukin-6 Production Does Not Increase with Age. J. Gerontol. Ser. A Biol. Sci. Med. Sci. 2001, 56, B81–B88. [Google Scholar] [CrossRef]
  122. Jeon, O.H.; Mehdipour, M.; Gil, T.H.; Kang, M.; Aguirre, N.W.; Robinson, Z.R.; Kato, C.; Etienne, J.; Lee, H.G.; Alimirah, F.; et al. Systemic Induction of Senescence in Young Mice after Single Heterochronic Blood Exchange. Nat. Metab. 2022, 4, 995–1006. [Google Scholar] [CrossRef]
  123. Markov, N.T.; Lindbergh, C.A.; Staffaroni, A.M.; Perez, K.; Stevens, M.; Nguyen, K.; Murad, N.F.; Fonseca, C.; Campisi, J.; Kramer, J.F.D. Age-Related Brain Atrophy Is Not a Homogenous Process: Different Functional Brain Networks Associate Differentially with Aging and Blood Factors. Proc. Natl. Acad. Sci. USA 2017, 120, 2017. [Google Scholar] [CrossRef]
  124. Zhu, Y.; Prata, L.G.P.L.; Gerdes, E.O.W.; Netto, J.M.E.; Pirtskhalava, T.; Giorgadze, N.; Tripathi, U.; Inman, C.L.; Johnson, K.O.; Xue, A.; et al. Orally-Active, Clinically-Translatable Senolytics Restore α-Klotho in Mice and Humans. eBioMedicine 2022, 77, 103912. [Google Scholar] [CrossRef]
  125. 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. Senolytics Decrease Senescent Cells in Humans: Preliminary Report from a Clinical Trial of Dasatinib plus Quercetin in Individuals with Diabetic Kidney Disease. eBioMedicine 2019, 47, 446–456. [Google Scholar] [CrossRef] [Green Version]
  126. Hoare, M.; Narita, M. The Power Behind the Throne: Senescence and the Hallmarks of Cancer. Annu. Rev. Cancer Biol. 2018, 2, 175–194. [Google Scholar] [CrossRef] [Green Version]
  127. Quan, T.; Fisher, G.J. Role of Age-Associated Alterations of the Dermal Extracellular Matrix Microenvironment in Human Skin Aging. Gerontology 2015, 61, 427. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  128. Frantz, C.; Stewart, K.M.; Weaver, V.M. The Extracellular Matrix at a Glance. J. Cell Sci. 2010, 123, 4195–4200. [Google Scholar] [CrossRef] [Green Version]
  129. Terlecki-Zaniewicz, L.; Pils, V.; Bobbili, M.R.; Lämmermann, I.; Perrotta, I.; Grillenberger, T.; Schwestka, J.; Weiß, K.; Pum, D.; Arcalis, E.; et al. Extracellular Vesicles in Human Skin: Cross-Talk from Senescent Fibroblasts to Keratinocytes by MiRNAs. J. Investig. Dermatol. 2019, 139, 2425–2436.e5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  130. Jun, J.-I.; Lau, L.F. The Matricellular Protein CCN1/CYR61 Induces Fibroblast Senescence and Restricts Fibrosis in Cutaneous Wound Healing. Nat. Cell Biol. 2010, 12, 676. [Google Scholar] [CrossRef] [PubMed]
  131. Lujambio, A.; Akkari, L.; Simon, J.; Grace, D.; Tschaharganeh, D.F.; Bolden, J.E.; Zhao, Z.; Thapar, V.; Joyce, J.A.; Krizhanovsky, V.; et al. Non-Cell-Autonomous Tumor Suppression by P53. Cell 2013, 153, 449–460. [Google Scholar] [CrossRef] [Green Version]
  132. Grosse, L.; Wagner, N.; Emelyanov, A.; Molina, C.; Lacas-Gervais, S.; Wagner, K.D.; Bulavin, D.V. Defined P16High Senescent Cell Types Are Indispensable for Mouse Healthspan. Cell Metab. 2020, 32, 87–99.e6. [Google Scholar] [CrossRef] [PubMed]
  133. Hiebert, P.; Wietecha, M.S.; Cangkrama, M.; Haertel, E.; Mavrogonatou, E.; Stumpe, M.; Steenbock, H.; Grossi, S.; Beer, H.D.; Angel, P.; et al. Nrf2-Mediated Fibroblast Reprogramming Drives Cellular Senescence by Targeting the Matrisome. Dev. Cell 2018, 46, 145–161.e10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Salamito, M.; Gillet, B.; Syx, D.; Vaganay, E.; Malbouyres, M.; Cerutti, C.; Tissot, N.; Exbrayat-Héritier, C.; Perez, P.; Jones, C.; et al. NRF2 Shortage in Human Skin Fibroblasts Dysregulates Matrisome Gene Expression and Affects Collagen Fibrillogenesis. J. Investig. Dermatol. 2023, 143, 386–397.e12. [Google Scholar] [CrossRef]
  135. Stearns-Reider, K.M.; D’Amore, A.; Beezhold, K.; Rothrauff, B.; Cavalli, L.; Wagner, W.R.; Vorp, D.A.; Tsamis, A.; Shinde, S.; Zhang, C.; et al. Aging of the Skeletal Muscle Extracellular Matrix Drives a Stem Cell Fibrogenic Conversion. Aging Cell 2017, 16, 518–528. [Google Scholar] [CrossRef] [Green Version]
  136. Castro, P.; Giri, D.; Lamb, D.; Ittmann, M. Cellular Senescence in the Pathogenesis of Benign Prostatic Hyperplasia. Prostate 2003, 55, 30–38. [Google Scholar] [CrossRef]
  137. 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]
  138. Takasugi, M.; Okada, R.; Takahashi, A.; Virya Chen, D.; Watanabe, S.; Hara, E. Small Extracellular Vesicles Secreted from Senescent Cells Promote Cancer Cell Proliferation through EphA2. Nat. Commun. 2017, 8, 15729. [Google Scholar] [CrossRef] [Green Version]
  139. Olumi, A.; Grossfeld, G.; Hayward, S.; Carroll, P.; Cunha, G.; Hein, P.; Tlsty, T. Carcinoma-Associated Fibroblasts Direct Tumor Progression of Initiated Human Prostatic Epithelium. Cancer Res. 1999, 59, 5002–5011. [Google Scholar] [CrossRef] [PubMed]
  140. Parrinello, S.; Coppe, J.P.; Krtolica, A.; Campisi, J. Stromal-Epithelial Interactions in Aging and Cancer: Senescent Fibroblasts Alter Epithelial Cell Differentiation. J. Cell Sci. 2005, 118 Pt 3, 485. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  141. Krtolica, A.; Ortiz De Solorzano, C.; Lockert, S.; Campisi, J. Quantification of Epithelial Cells in Coculture with Fibroblasts by Fluorescence Image Analysis. Cytometry 2002, 49, 73–82. [Google Scholar] [CrossRef] [PubMed]
  142. Gosselin, K.; Martien, S.; Pourtier, A.; Vercamer, C.; Ostoich, P.; Morat, L.; Sabatier, L.; Duprez, L.; T’kint De Roodenbeke, C.; Gilson, E.; et al. Senescence-Associated Oxidative DNA Damage Promotes the Generation of Neoplastic Cells. Cancer Res 2009, 69, 7917–7942. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Romanov, S.R.; Kozakiewicz, B.K.; Holst, C.R.; Stampfer, M.R.; Haupt, L.M.; Tlsty, T.D. Normal Human Mammary Epithelial Cells Spontaneously Escape Senescence and Acquire Genomic Changes. Nature 2001, 409, 633–637. [Google Scholar] [CrossRef]
  144. Gosselin, K.; Deruy, E.; Martien, S.; Vercamer, C.; Bouali, F.; Dujardin, T.; Slomianny, C.; Houel-Renault, L.; Chelli, F.; De Launoit, Y.; et al. Senescent Keratinocytes Die by Autophagic Programmed Cell Death. Am. J. Pathol. 2009, 174, 423–435. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Malaquin, N.; Vercamer, C.; Bouali, F.; Martien, S.; Deruy, E.; Wernert, N.; Chwastyniak, M.; Pinet, F.; Abbadie, C.; Pourtier, A. Senescent Fibroblasts Enhance Early Skin Carcinogenic Events via a Paracrine MMP-PAR-1 Axis. PLoS ONE 2013, 8, e63607. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Martin, N.; Salazar-Cardozo, C.; Vercamer, C.; Ott, L.; Marot, G.; Slijepcevic, P.; Abbadie, C.; Pluquet, O. Identification of a Gene Signature of a Pre-Transformation Process by Senescence Evasion in Normal Human Epidermal Keratinocytes. Mol. Cancer 2014, 13, 151. [Google Scholar] [CrossRef] [Green Version]
  147. Tinaburri, L.; Valente, C.; Teson, M.; Minafò, Y.A.; Cordisco, S.; Guerra, L.; Dellambra, E. The Secretome of Aged Fibroblasts Promotes EMT-like Phenotype in Primary Keratinocytes from Elderly Donors through BDNF-TrkB Axis. J. Investig. Dermatol. 2021, 141, 1052–1062.e12. [Google Scholar] [CrossRef]
  148. Sprenger, C.C.; Plymate, S.R.; Reed, M.J. Aging-Related Alterations in the Extracellular Matrix Modulate the Microenvironment and Influence Tumor Progression. Int. J. Cancer 2010, 127, 2739–2748. [Google Scholar] [CrossRef] [Green Version]
  149. Kang, J.; Chen, W.; Xia, J.; Li, Y.; Yang, B.; Chen, B.; Sun, W.; Song, X.; Xiang, W.; Wang, X.; et al. Extracellular Matrix Secreted by Senescent Fibroblasts Induced by UVB Promotes Cell Proliferation in HaCaT Cells through PI3K/AKT and ERK Signaling Pathways. Int. J. Mol. Med. 2008, 21, 777–784. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  150. Egeblad, M.; Rasch, M.G.; Weaver, V.M. Dynamic Interplay between the Collagen Scaffold and Tumor Evolution. Curr. Opin. Cell Biol. 2010, 22, 697–706. [Google Scholar] [CrossRef] [Green Version]
  151. Paszek, M.J.; Zahir, N.; Johnson, K.R.; Lakins, J.N.; Rozenberg, G.I.; Gefen, A.; Reinhart-King, C.A.; Margulies, S.S.; Dembo, M.; Boettiger, D.; et al. Tensional Homeostasis and the Malignant Phenotype. Cancer Cell 2005, 8, 241–254. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  152. Faget, D.V.; Ren, Q.; Stewart, S.A. Unmasking Senescence: Context-Dependent Effects of SASP in Cancer. Nat. Rev. Cancer 2019, 19, 439–453. [Google Scholar] [CrossRef] [PubMed]
  153. 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] [PubMed] [Green Version]
  154. Acosta, J.C.; Banito, A.; Wuestefeld, T.; Georgilis, A.; Janich, P.; Morton, J.P.; Athineos, D.; Kang, T.W.; Lasitschka, F.; Andrulis, M.; et al. A Complex Secretory Program Orchestrated by the Inflammasome Controls Paracrine Senescence. Nat. Cell Biol. 2013, 15, 978. [Google Scholar] [CrossRef]
  155. Dickey, J.S.; Baird, B.J.; Redon, C.E.; Sokolov, M.V.; Sedelnikova, O.A.; Bonner, W.M. Intercellular Communication of Cellular Stress Monitored by γ-H2AX Induction. Carcinogenesis 2009, 30, 1686–1695. [Google Scholar] [CrossRef] [Green Version]
  156. Borghesan, M.; Fafiá N-Labora, J.; Eleftheriadou, O.; Vossenkamper, A.; Muñ Oz-Espin, D.; O’, A.; Correspondence, L. Small Extracellular Vesicles Are Key Regulators of Non-Cell Autonomous Intercellular Communication in Senescence via the Interferon Protein IFITM3. Cell Rep. 2019, 27, 3956–3971. [Google Scholar] [CrossRef] [Green Version]
  157. Fulzele, S.; Mendhe, B.; Khayrullin, A.; Johnson, M.; Kaiser, H.; Liu, Y.; Isales, C.M.; Hamrick, M.W. Muscle-Derived MiR-34a Increases with Age in Circulating Extracellular Vesicles and Induces Senescence of Bone Marrow Stem Cells. Aging 2019, 11, 1791–1803. [Google Scholar] [CrossRef]
  158. Lorenzo, E.C.; Torrance, B.L.; Keilich, S.R.; Al-Naggar, I.; Harrison, A.; Xu, M.; Bartley, J.M.; Haynes, L. Senescence-induced Changes in CD4 T Cell Differentiation Can Be Alleviated by Treatment with Senolytics. Aging Cell 2022, 21, e13525. [Google Scholar] [CrossRef]
  159. Ritschka, B.; Storer, M.; Mas, A.; Heinzmann, F.; Ortells, M.C.; Morton, J.P.; Sansom, O.J.; Zender, L.; Keyes, W.M. The Senescence-Associated Secretory Phenotype Induces Cellular Plasticity and Tissue Regeneration. Genes Dev. 2017, 31, 172–183. [Google Scholar] [CrossRef] [Green Version]
  160. Mosteiro, L.; Pantoja, C.; Alcazar, N.; Marión, R.M.; Chondronasiou, D.; Rovira, M.; Fernandez-Marcos, P.J.; Muñoz-Martin, M.; Blanco-Aparicio, C.; Pastor, J.; et al. Tissue Damage and Senescence Provide Critical Signals for Cellular Reprogramming in Vivo. Science 2016, 354, aaf4445. [Google Scholar] [CrossRef]
  161. Watt, F.M.; Fujiwara, H. Cell-Extracellular Matrix Interactions in Normal and Diseased Skin. Cold Spring Harb. Perspect. Biol. 2011, 3, a005124. [Google Scholar] [CrossRef] [Green Version]
  162. Ozcebe, S.G.; Bahcecioglu, G.; Yue, X.S.; Zorlutuna, P. Effect of Cellular and ECM Aging on Human IPSC-Derived Cardiomyocyte Performance, Maturity and Senescence. Biomaterials 2021, 268, 120554. [Google Scholar] [CrossRef]
  163. Moiseeva, V.; Cisneros, A.; Sica, V.; Deryagin, O.; Lai, Y.; Jung, S.; Andrés, E.; An, J.; Segalés, J.; Ortet, L.; et al. Senescence Atlas Reveals an Aged-like Inflamed Niche That Blunts Muscle Regeneration. Nature 2022, 613, 169–178. [Google Scholar] [CrossRef] [PubMed]
  164. Saul, D.; Kosinsky, R.L.; Atkinson, E.J.; Doolittle, M.L.; Zhang, X.; LeBrasseur, N.K.; Pignolo, R.J.; Robbins, P.D.; Niedernhofer, L.J.; Ikeno, Y.; et al. A New Gene Set Identifies Senescent Cells and Predicts Senescence-Associated Pathways across Tissues. Nat. Commun. 2022, 13, 4827. [Google Scholar] [CrossRef] [PubMed]
  165. Zhu, Y.; Tchkonia, T.; Pirtskhalava, T.; Gower, A.C.; Ding, H.; Giorgadze, N.; Palmer, A.K.; Ikeno, Y.; Hubbard, G.B.; Lenburg, M.; et al. The Achilles’ Heel of Senescent Cells: From Transcriptome to Senolytic Drugs. Aging Cell 2015, 14, 644. [Google Scholar] [CrossRef]
  166. Kim, E.C.; Kim, J.R. Senotherapeutics: Emerging Strategy for Healthy Aging and Age-Related Disease. BMB Rep. 2019, 52, 47–55. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  167. Zhang, L.; Pitcher, L.E.; Prahalad, V.; Niedernhofer, L.J.; Robbins, P.D. Targeting Cellular Senescence with Senotherapeutics: Senolytics and Senomorphics. FEBS J. 2023, 290, 1362–1383. [Google Scholar] [CrossRef]
  168. Niedernhofer, L.J.; Robbins, P.D. Senotherapeutics for Healthy Ageing. Nat. Rev. Drug Discov. 2018, 17, 377. [Google Scholar] [CrossRef] [Green Version]
  169. Sun, Y.; Li, Q.; Kirkland, J.L. Targeting Senescent Cells for a Healthier Longevity: The Roadmap for an Era of Global Aging. Life Med. 2022, 1, 103–119. [Google Scholar] [CrossRef]
  170. Wissler Gerdes, E.O.; Zhu, Y.; Tchkonia, T.; Kirkland, J.L. Discovery, Development, and Future Application of Senolytics: Theories and Predictions. FEBS J. 2020, 287, 2418–2427. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  171. 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]
  172. Pignolo, R.J.; Passos, J.F.; Khosla, S.; Tchkonia, T.; Kirkland, J.L. Reducing Senescent Cell Burden in Aging and Disease. Trends Mol. Med. 2020, 26, 630–638. [Google Scholar] [CrossRef]
  173. Zhang, L.; Gao, S.; White, Z.; Dai, Y.; Malik, A.B.; Rehman, J. Single-Cell Transcriptomic Profiling of Lung Endothelial Cells Identifies Dynamic Inflammatory and Regenerative Subpopulations. JCI Insight 2022, 7, 6. [Google Scholar] [CrossRef]
  174. Wissler Gerdes, E.O.; Misra, A.; Netto, J.M.E.; Tchkonia, T.; Kirkland, J.L. Strategies for Late Phase Preclinical and Early Clinical Trials of Senolytics. Mech. Ageing Dev. 2021, 200, 111591. [Google Scholar] [CrossRef]
  175. Kirkland, J.L.; Tchkonia, T. Senolytic Drugs: From Discovery to Translation. J. Intern. Med. 2020, 288, 518–536. [Google Scholar] [CrossRef] [PubMed]
  176. Morsli, S.; Doherty, G.J.; Muñoz-Espín, D. Activatable Senoprobes and Senolytics: Novel Strategies to Detect and Target Senescent Cells. Mech. Ageing Dev. 2022, 202, 111618. [Google Scholar] [CrossRef] [PubMed]
  177. Paez-Ribes, M.; González-Gualda, E.; Doherty, G.J.; Muñoz-Espín, D. Targeting Senescent Cells in Translational Medicine. EMBO Mol. Med. 2019, 11, e10234. [Google Scholar] [CrossRef] [PubMed]
  178. Prattichizzo, F.; Giuliani, A.; Recchioni, R.; Bonafè, M.; Marcheselli, F.; De Carolis, S.; Campanati, A.; Giuliodori, K.; Rippo, M.R.; Brugè, F.; et al. Anti-TNF-α Treatment Modulates SASP and SASP-Related MicroRNAs in Endothelial Cells and in Circulating Angiogenic Cells. Oncotarget 2016, 7, 11945–11958. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  179. Lister, R.; Chrysovergi, M.; Lagares, D. Ephrin-B2 Neutralizing Antibody Suppresses the SASP in Senescent Fibroblasts and Treats Lung Fibrosis in Aged Mice. Am. Thorac. Soc. 2022, 216, 147059. [Google Scholar]
  180. Chong, M.; Yin, T.; Chen, R.; Xiang, H.; Yuan, L.; Ding, Y.; Pan, C.C.; Tang, Z.; Alexander, P.B.; Jia, Y.; et al. CD36 Initiates the Secretory Phenotype during the Establishment of Cellular Senescence. EMBO Rep. 2018, 19, e45274. [Google Scholar] [CrossRef]
  181. Orjalo, A.V.; Bhaumik, D.; Gengler, B.K.; Scott, G.K.; Campisi, J. Cell Surface-Bound IL-1α Is an Upstream Regulator of the Senescence-Associated IL-6/IL-8 Cytokine Network. Proc. Natl. Acad. Sci. USA 2009, 106, 17031–17036. [Google Scholar] [CrossRef] [Green Version]
  182. Di Micco, R.; Krizhanovsky, V.; Baker, D.; d’Adda di Fagagna, F. Cellular Senescence in Ageing: From Mechanisms to Therapeutic Opportunities. Nat. Rev. Mol. Cell Biol. 2020, 22, 75–95. [Google Scholar] [CrossRef]
  183. Lim, H.; Park, H.; Kim, H.P. Effects of Flavonoids on Senescence-Associated Secretory Phenotype Formation from Bleomycin-Induced Senescence in BJ Fibroblasts. Biochem. Pharmacol. 2015, 96, 337–348. [Google Scholar] [CrossRef] [PubMed]
  184. Lim, J.S.; Lee, D.Y.; Kim, H.S.; Park, S.C.; Oh, W.K.; Cho, K.A. Identification of a novel senomorphic agent, avenanthramide C, via the suppression of the senescence-associated secretory phenotype. Mech. Ageing Dev. 2020, 192, 111355. [Google Scholar] [CrossRef]
  185. Alimbetov, D.; Davis, T.; Brook, A.J.C.; Cox, L.S.; Faragher, R.G.A.; Nurgozhin, T.; Zhumadilov, Z.; Kipling, D. Suppression of the Senescence-Associated Secretory Phenotype (SASP) in Human Fibroblasts Using Small Molecule Inhibitors of P38 MAP Kinase and MK2. Biogerontology 2016, 17, 305. [Google Scholar] [CrossRef] [Green Version]
  186. Laberge, R.M.; Zhou, L.; Sarantos, M.R.; Rodier, F.; Freund, A.; de Keizer, P.L.J.; Liu, S.; Demaria, M.; Cong, Y.S.; Kapahi, P.; et al. Glucocorticoids Suppress Selected Components of the Senescence-Associated Secretory Phenotype. Aging Cell 2012, 11, 569–578. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  187. Menicacci, B.; Cipriani, C.; Margheri, F.; Mocali, A.; Giovannelli, L. Modulation of the Senescence-Associated Inflammatory Phenotype in Human Fibroblasts by Olive Phenols. Int. J. Mol. Sci. 2017, 18, 2275. [Google Scholar] [CrossRef] [Green Version]
  188. Chen, D.D.; Peng, X.; Wang, Y.; Jiang, M.; Xue, M.; Shang, G.; Liu, X.; Jia, X.; Liu, B.; Lu, Y.; et al. HSP90 Acts as a Senomorphic Target in Senescent Retinal Pigmental Epithelial Cells. Aging 2021, 13, 21547. [Google Scholar] [CrossRef] [PubMed]
  189. Signaling, B.; Woo, J.; Shin, S.; Ji, H.; Ryu, D.; Cho, E.; Kim, Y.; Kim, J.; Park, D.; Jung, E. Isatis Tinctoria L. Leaf Extract Inhibits Replicative Senescence. Nutrients 2022, 14, 1979. [Google Scholar]
  190. De Cecco, M.; Ito, T.; Petrashen, A.P.; Elias, A.E.; Skvir, N.J.; Criscione, S.W.; Caligiana, A.; Brocculi, G.; Adney, E.M.; Boeke, J.D.; et al. L1 Drives IFN in Senescent Cells and Promotes Age-Associated Inflammation. Nature 2019, 566, 73–78. [Google Scholar] [CrossRef]
  191. Hu, Q.; Peng, J.; Jiang, L.; Li, W.; Su, Q.; Zhang, J.; Li, H.; Song, M.; Cheng, B.; Xia, J.; et al. Metformin as a Senostatic Drug Enhances the Anticancer Ef Fi Cacy of CDK4/6 Inhibitor in Head and Neck Squamous Cell Carcinoma. Cell Death Dis. 2020, 11, 925. [Google Scholar] [CrossRef]
  192. Tai, S.; Sun, J.; Zhou, Y.; Zhu, Z.; He, Y.; Chen, M.; Yang, H.; Xiao, Y.; Tu, T.; Tang, L.; et al. Metformin Suppresses Vascular Smooth Muscle Cell Senescence by Promoting Autophagic Flux. J. Adv. Res. 2022, 41, 205–218. [Google Scholar] [CrossRef]
  193. Matacchione, G.; Gurău, F.; Silvestrini, A.; Tiboni, M.; Mancini, L.; Valli, D.; Rippo, M.R.; Recchioni, R.; Marcheselli, F.; Carnevali, O.; et al. Anti-SASP and Anti-Inflammatory Activity of Resveratrol, Curcumin and β-Caryophyllene Association on Human Endothelial and Monocytic Cells. Biogerontology 2021, 22, 297. [Google Scholar] [CrossRef]
  194. Frediani, E.; Scavone, F.; Laurenzana, A.; Chillà, A.; Tortora, K.; Cimmino, I.; Leri, M.; Bucciantini, M.; Mangoni, M.; Fibbi, G.; et al. Olive Phenols Preserve Lamin B1 Expression Reducing CGAS/STING/NF κ B-Mediated SASP in Ionizing Radiation-Induced Senescence. J. Cell. Mol. Med. 2022, 26, 2337–2350. [Google Scholar] [CrossRef]
  195. Liu, S.; Uppal, H.; Demaria, M.; Desprez, P.Y.; Campisi, J.; Kapahi, P. Simvastatin Suppresses Breast Cancer Cell Proliferation Induced by Senescent Cells. Sci. Rep. 2015, 5, 17895. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  196. Laberge, R.M.; 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. [Google Scholar] [CrossRef] [PubMed]
  197. Wang, R.; Yu, Z.; Sunchu, B.; Shoaf, J.; Dang, I.; Zhao, S.; Caples, K.; Bradley, L.; Beaver, L.M.; Ho, E.; et al. Rapamycin Inhibits the Secretory Phenotype of Senescent Cells by a Nrf2-independent Mechanism. Aging Cell 2017, 16, 564. [Google Scholar] [CrossRef] [PubMed]
  198. Csiszar, A.; Sosnowska, D.; Wang, M.; Lakatta, E.G.; Sonntag, W.E.; Ungvari, Z. Age-Associated Proinflammatory Secretory Phenotype in Vascular Smooth Muscle Cells From the Non-Human Primate Macaca Mulatta: Reversal by Resveratrol Treatment. Journals Gerontol. Ser. A 2012, 67, 811–820. [Google Scholar] [CrossRef]
  199. Woo, J.; Shin, S.; Cho, E.; Ryu, D.; Garandeau, D.; Chajra, H.; Fréchet, M.; Park, D.; Jung, E. Senotherapeutic-like Effect of Silybum Marianum Flower Extract Revealed on Human Skin Cells. PLoS ONE 2021, 16, e0260545. [Google Scholar] [CrossRef] [PubMed]
  200. Gao, L.; Wang, Y.H.; Liu, Z.H.; Sun, Y.; Cai, P.; Jing, Q. Identification of a Small Molecule SR9009 That Activates NRF2 to Counteract Cellular Senescence. Aging Cell 2021, 20, e13483. [Google Scholar] [CrossRef]
  201. Park, M.; Na, J.; Kwak, S.Y.; Park, S.; Kim, H.; Lee, S.J.; Jang, W.S.; Lee, S.B.; Jang, W.I.; Jang, H.; et al. Zileuton Alleviates Radiation-Induced Cutaneous Ulcers via Inhibition of Senescence-Associated Secretory Phenotype in Rodents. Int. J. Mol. Sci. 2022, 23, 8390. [Google Scholar] [CrossRef] [PubMed]
  202. Farr, J.N.; Xu, M.; Weivoda, M.M.; Monroe, D.G.; Fraser, D.G.; Onken, J.L.; Negley, B.A.; Sfeir, J.G.; Ogrodnik, M.B.; Hachfeld, C.M.; et al. Targeting Cellular Senescence Prevents Age-Related Bone Loss in Mice. Nat. Med. 2017, 23, 1072–1079. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  203. Hughes, B.K.; Wallis, R.; Bishop, C.L. Yearning for Machine Learning: Applications for the Classification and Characterisation of Senescence. Cell Tissue Res. 2023, 1–6. [Google Scholar] [CrossRef] [PubMed]
Figure 1. The updating hallmarks of senescence. The scheme compiles the major hallmarks of senescent cells, which are classified into five characteristic groups: cell cycle arrest, shape and cell surface proteins, nuclear alterations, organelle and metabolic dysfunctions, and senescence-associated secretory phenotype. The light and dark brown portions indicate the well-described standard hallmarks and the more recent ones, respectively. CDKIs, cyclin-dependent kinase inhibitors; UPR, unfolded protein response; ER, endoplasmic reticulum; ERAD, ER-associated degradation. This figure was created using BioRender.com.
Figure 1. The updating hallmarks of senescence. The scheme compiles the major hallmarks of senescent cells, which are classified into five characteristic groups: cell cycle arrest, shape and cell surface proteins, nuclear alterations, organelle and metabolic dysfunctions, and senescence-associated secretory phenotype. The light and dark brown portions indicate the well-described standard hallmarks and the more recent ones, respectively. CDKIs, cyclin-dependent kinase inhibitors; UPR, unfolded protein response; ER, endoplasmic reticulum; ERAD, ER-associated degradation. This figure was created using BioRender.com.
Ijms 24 10788 g001
Figure 2. The crosstalk between the senescent cells and their microenvironment through the SASP. The core SASP is defined as the common SASP components shared between different senescent cells regardless of the cell type, senescence inducer, micro-environment, or time-point. The final composition of SASP (colored dots) is the combination of the core SASP and the variable SASP, which is defined by the four different factors mentioned above. Altogether, the whole SASP will influence different physiological and pathophysiological processes, such as cell differentiation, micro-environment deregulation, paracrine induced-senescence, and age-related pathologies (left panel). However, the question remains, are those modulations of the cellular environment influencing, in turn, the composition of SASP, and to what extent the core and variable SASP are affected (right panel)?
Figure 2. The crosstalk between the senescent cells and their microenvironment through the SASP. The core SASP is defined as the common SASP components shared between different senescent cells regardless of the cell type, senescence inducer, micro-environment, or time-point. The final composition of SASP (colored dots) is the combination of the core SASP and the variable SASP, which is defined by the four different factors mentioned above. Altogether, the whole SASP will influence different physiological and pathophysiological processes, such as cell differentiation, micro-environment deregulation, paracrine induced-senescence, and age-related pathologies (left panel). However, the question remains, are those modulations of the cellular environment influencing, in turn, the composition of SASP, and to what extent the core and variable SASP are affected (right panel)?
Ijms 24 10788 g002
Table 1. Common SASP proteins shared between different cell types under different inducers of senescence.
Table 1. Common SASP proteins shared between different cell types under different inducers of senescence.
SASP FactorCell TypeInducerDetectionReferences
IL-6Human fibroblasts (WI-38, IM90, BJ)
Prostate epithelial cells (PrECs)
Human keratinocytes (NHEK)
Irradiation-induced senescence
Replicative Senescence
Ras overexpression
UVB-induced senescence
Antibodies array
ELISA
[9,29,70]
CXCL1-2-3Human fibroblasts (IMR90, HCA2)
Prostate epithelial cells (PrECs)
Irradiation-induced senescence
Replicative Senescence
Antibodies array[9,29]
IL-8Human fibroblasts (WI-38, IM90, BJ),
Prostate epithelial cells (PrECs)
Renal epithelial cells (ATCC)
Human keratinocytes (NHEK)
Irradiation-induced senescence
Replicative Senescence
UVB-induced senescence
Glyoxal-induced senescence
Antibodies array
Mass spectrometry
ELISA
[9,29,34,60,70]
IGFBP-2Human fibroblasts (IMR90, WI-38, HCA-2, BJ)
Renal epithelial cells (ATCC)
Irradiation-induced senescenceAntibodies array
Mass spectrometry
[29,60]
IL-7Human fibroblasts (WI-38, HCA-2, BJ)
Prostate epithelial cells (PrECs)
Irradiation-induced senescenceAntibodies array[9]
GDF15Human fibroblasts (IMR-90)
Epithelial renal cells (ATCC)
Irradiation-induced senescence
Replicative Senescence
Mass spectrometry[60]
Macrophage migration inhibitory factor (MIF)Human fibroblasts (IMR-90, WI-38)
Epithelial renal cells (ATCC)
Human keratinocytes (NHEK)
Prostate epithelial cells (PrECs)
Bone marrow MSC
Irradiation-induced senescence
UVA-induced senescence
RAS overexpression
Chemical-induced senescence (ATZ)
H2O2-induced senescence
Mass spectrometry
Antibodies array
[9,60,71,72]
Filamin BHuman fibroblasts (IMR-90)
Epithelial renal cells (ATCC)
Human keratinocytes (NHEK)
Prostate epithelial cells (PrECs)
Bone marrow MSC
Irradiation-induced senescence
UVA-induced senescence
RAS overexpression
Chemical-induced senescence (ATZ)
H2O2-induced senescence
Mass spectrometry[9,60,71,72]
Cathepsin DMesenchymal stem cellsReplicative Senescence
Chemical-induced senescence (doxorubicin)
H2O2-induced senescence
Irradiation-induced senescence
Mass spectrometry[72]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Giroud, J.; Bouriez, I.; Paulus, H.; Pourtier, A.; Debacq-Chainiaux, F.; Pluquet, O. Exploring the Communication of the SASP: Dynamic, Interactive, and Adaptive Effects on the Microenvironment. Int. J. Mol. Sci. 2023, 24, 10788. https://doi.org/10.3390/ijms241310788

AMA Style

Giroud J, Bouriez I, Paulus H, Pourtier A, Debacq-Chainiaux F, Pluquet O. Exploring the Communication of the SASP: Dynamic, Interactive, and Adaptive Effects on the Microenvironment. International Journal of Molecular Sciences. 2023; 24(13):10788. https://doi.org/10.3390/ijms241310788

Chicago/Turabian Style

Giroud, Joëlle, Inès Bouriez, Hugo Paulus, Albin Pourtier, Florence Debacq-Chainiaux, and Olivier Pluquet. 2023. "Exploring the Communication of the SASP: Dynamic, Interactive, and Adaptive Effects on the Microenvironment" International Journal of Molecular Sciences 24, no. 13: 10788. https://doi.org/10.3390/ijms241310788

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