With aging, a portion of cells, including mesenchymal stem cells (MSCs), become senescent, and these senescent cells accumulate and promote various age-related diseases. Therefore, the older age group has become a major population for MSC therapy, which is aimed at improving tissue regeneration and function of the aged body. However, the application of MSC therapy is often unsatisfying in the aged group. One reasonable conjecture for this correlation is that aging microenvironment reduces the number and function of MSCs. Cellular senescence also plays an important role in MSC function impairment. Thus, it is necessary to explore the relationship between senescence and MSCs for improving the application of MSCs in the elderly. Here, we present the influence of aging on MSCs and the characteristics and functional changes of senescent MSCs. Furthermore, current therapeutic strategies for improving MSC therapy in the elderly group are also discussed.

Aging has a significant impact on human health and increased risk of human diseases, such as cardiovascular disorders, diabetes, and neurodegenerative diseases. For example, axonal regeneration was less effective in aged mice than young mice when nerve conduit and autologous nerve were transplanted [1].

Mesenchymal stem cells (MSCs) have great potential for the treatment of degenerative diseases, such as osteoporosis, large bone defects, and nerve defects. It is reported that axonal regeneration using nerve conduits was improved when induced pluripotent stem cell derived neurospheres were transplanted in the aged mice [1]. Furthermore, MSC therapy has been widely and successfully applied in various diseases, including liver injury, multiple sclerosis, graft-versus-host disease, Crohn’s disease, diabetes mellitus, and myocardial infarction. These achievements relied on the following characteristics of MSCs: (1) multiple differentiation potential: it was the first detailed description in 1999, including osteogenesis, chondrogenesis, and adipogenesis [2]; (2) replication capacity: upon stimulation by biological signals, MSCs are activated and recruited to injured sites to proliferate, regenerate, and replace damaged tissues [3, 4]; (3) low immunogenicity: MSCs have vague cell surface markers, which could cause less host immune response. Therefore, MSC therapies are likely candidates for use in aged individuals to restore the loss of tissue homeostasis caused by degenerative disease.

However, the application of MSC therapy is less effective in the elderly group than in the young group. For example, bone regeneration impaired in older mice when human muscle-derived stem cells were transplanted into the defect bone area [5].

This phenomenon indicated close relationship between aging and stem cells. On the one hand, with aging, a portion of stem cells become senescent, that is, characterized by permanent growth arrest, resistance to apoptosis, macromolecular damage, and altered metabolism [6]. These senescent cells (SNCs) promote various age-related diseases. On the other hand, aging microenvironment, including a senescence-associated secretory phenotype (SASP), impairs the function of transplanted MSCs [7-9], which is a major factor that contributes to the unsatisfactory therapeutic effect of MSC therapy.

In this review, we discuss the effect of aging microenvironment on the biological properties of MSCs and their therapeutic potential, as well as the characteristics and functional changes of senescent MSCs. Possible techniques to improve the therapeutic effect of MSCs and anti-senescence in the older population are discussed as well. The aim of this review is promoting the therapeutic effect of MSCs in the elderly group.

It is generally recognized that a causal link has been established between senescence and aging [10]. With aging, SNCs accumulate in various tissues and organs which – once reaching a sufficient number – may drive age-related diseases. However, they are different. Generally, senescence is a cellular program mainly triggered by stressful stimuli, including DNA damage, oncogene activation, and telomere shortening [6], while aging is an individual condition caused by the passage of time. Therefore, SNCs can be detected at any physical age from embryogenesis to adulthood, regardless of organismal age. Besides, more than 80% of the cells in the older age group are not SNCs. However, young groups with premature phenotype might contain more SNSs even at a much younger age. For example, similar to Werner syndrome in human, mice with mutants in the Werner helicase and telomerase develop clinical symptoms of premature aging once they reach adulthood [11].

Cellular senescence is a cell state characterized by cell cycle arrest, antiapoptosis, and special secretory features related to age-related diseases [6]. According to stimulating factors, cellular senescence can be divided into replicative senescence which occurs by replication and premature senescence which is caused by stressful stimuli [12].

Characteristics of SNCs

Cell Cycle Arrest

Different from quiescence and terminal differentiation, SNCs lost their ability to divide to some extent but remained active and metabolically active [13]. The senescence growth arrest is generally irreversible. However, SNCs can re-enter cell cycle in some contexts. Cyclin-dependent kinase inhibitors (CDKi) p16 and p21 are the 2 main signaling pathways that initiate and maintain cell cycle arrest by activating the transcriptional regulator retinoblastoma protein. Mitogen-activated protein kinases (MAPKs) [14] and P53 [15] are 2 upstream enzymes of CDKi and also promote growth arrest in SNCs by controlling CDKi.

Resistance to Apoptosis

SNCs are antiapoptotic and thus block programmed cell death [16, 17]. P53 signaling is a main pathway modulating the expression of a wide variety of molecules and genes related to senescence or apoptosis. It regulates extrinsic pathways and intrinsic pathways, 2 major apoptotic pathways. Extrinsic pathways involve particular “death” receptors from the tumor necrosis factor receptor family and death-inducing signaling complex, which lead to a cascade of activation of caspases and induce apoptosis. Intrinsic apoptotic pathways are dominated by the B-cell lymphoma-2 (BCL-2) family of proteins, which govern the release of cytochrome C from the mitochondria.

Secretory Features

SNCs secrete distinctive bioactive factors, including pro-inflammatory cytokines and chemokines, growth modulators, and proteases, collectively termed SASP [6]. SNCs do not exist as a single, discrete cell but are diverse: SNCs of different origins and stimuli secrete different SASP factors, which drive the pathogenesis of diseases and trigger cell apoptosis through different senescence mechanisms.

The mechanisms related to SASP regulation remain incompletely understood. Several transcription factors associated with regulation of SASP have been reported, mainly through the action of transcription factor nuclear factor-kappa B (NF-κB) and CCAAT/enhancer-binding proteins-β [18]. Besides, there are some factors involved in regulating SASP through the NF-κB pathway, such as p38-MAPKs [14], mTOR [19], and transcription factor GATA4 [20]. Cyclic GMP-AMP synthase/stimulator of interferon genes triggers the secretion of SASP factors, thereby promoting paracrine senescence [21].

Methods for Detecting SNCs

Although some markers of identification of SNCs are more universal, such as senescence-associated β-galactosidase [22], currently, no single marker has been found to specifically identify SNCs [23] due to many factors affecting marker specificity, including cell type, tissue developmental stage, and stimulus. Remarkably, with the development of technology and concept, more and more novel methods have been used for identification of SNCs. Cell cycle arrest is a main feature of SNCs, suggesting high expression of cyclin kinase inhibitors, such as P15 [24-26], P16, P19 [27-29], P21, P27 [24, 30, 31], P53 [27, 32, 33], and P57 [26, 29, 30], and less expression of proliferation markers, such as bromodeoxyuridine [34, 35] and ki67 [28, 36, 37]. Furthermore, markers of apoptosis such as P53, decoy receptor 2, and BHLHB2 gene [37-39] are related to senescence.

Senescence triggers the production and accumulation of distinct heterochromatic structures and thus results in DNA damage. Senescence-associated distension of satellites [6, 35, 40, 41] assay and telomere-associated DNA damage response foci [42, 43] assay have also been recognized as specific markers of senescence in vivo. Other markers of DNA damage include γ-H2A.X [36, 41, 42], telomere dysfunction-induced foci [44-46], terminal restriction fragmentation [47], senescence-associated heterochromatic foci [48-50], and high-mobility group box 1 [36, 51, 52].

Therefore, it is necessary to combine various kinds of above methods, as well as a special SASP. There is still a long way before finding the gold standard for identifying SNCs.

MSC therapy has shown great application prospects in cell and animal experiments. However, the effect of stem cell therapy is currently unsatisfactory in clinic. Some scholars suspect that host age may be responsible for it. In most cell and animal experiments, the hosts are young individuals. While clinically, the elderly are the main recipients of MSC therapy. It is reported that bone regeneration impaired in older mice compared with young mice when human muscle-derived stem cells were transplanted into the defected bone area [5]. Similarly, after 6 months of repairing the skull defects by fetal bone marrow stromal cells (fetal BMSCs) with the decalcified bone matrix scaffolds, the repair effect of the skull defect in old goats is worse compared to young goats [53]. Furthermore, serum from old mice has a negative impact on muscle satellite cells (a kind of MSCs) and neurogenesis in young mice [7]. Serum from old rat reduces the proliferation and survival of bone marrow-derived MSCs and promotes its senescence [8]. What is more, exposure to young system environment can rejuvenate aging progenitor cells [54].

To sum up, it is reasonable to doubt that aging environment has adverse effects on the function of MSCs. However, mechanisms of aging microenvironment on MSCs are incompletely understood. It has been reported that the age-associated functional degeneration of stem cell is induced by the surrounding molecules to a significant extent, in addition to cell-intrinsic mechanisms [55]. In this part, we discuss how aging microenvironment affects MSCs (shown in Fig. 1), and what effects does it have on MSCs.

Fig. 1.

Effect of senescent microenvironment on MSCs. Senescent microenvironment plays a role in MSCs through SASP, EVs derived from SNCs, and cell-cell contact. MSCs, mesenchymal stem cells; SNCs, senescent cells; SASP, senescence-associated secretory phenotype; EVs, extracellular vesicles.

Fig. 1.

Effect of senescent microenvironment on MSCs. Senescent microenvironment plays a role in MSCs through SASP, EVs derived from SNCs, and cell-cell contact. MSCs, mesenchymal stem cells; SNCs, senescent cells; SASP, senescence-associated secretory phenotype; EVs, extracellular vesicles.

Close modal

Senescence-Associated Secretory Phenotype

SASP contains various age-associated components; these components mediate paracrine senescence and affect the function of MSCs, including proliferation ability, clonal formation ability, differentiation potential, immune characteristics, telomerase activity, cell migration ability, and adhesion. The SASP can create a chronic sterile inflammatory environment which is mainly mediated by NF-κB signaling and thus drive stem cell dysfunction and aberrant remodeling [56]. Pro-inflammatory factors, such as IL-1β and IL-6, induce the expression of p53 and p21, increase oxidative and DNA damage and thus cause senescence of IMR90 cells [57]. In addition, the early SASP regulates p15INK4b and p21CIP1 and induces senescence in nearby cells by transforming growth factor-β (TGF-β) family members [57-59].

Age-related insulin-like growth factor-binding protein 4 and 7, a kind of chemokines, could directly induce a senescent phenotype in P1 MSCs [60] and inhibit osteogenic differentiation of rat MSCs [61]. SASP is not always harmful. For example, intestinal stem cells after heavy ion radiation in mice and cells undergoing OIS show an increased expression of vascular endothelial growth factor (VEGF, one of the SASP components) [57, 62]. However, after pretreating with VEGF, the stem cells show a remodeling ability and attenuate age-related decay of adult hippocampal neurogenesis. In addition, MSCs release VEGF, activate the Jagged-1/Notch-1 signaling pathway, inhibit secretion of TGF-β1, and thus rescue rat embryonic myoblasts from doxorubicin-induced senescence [63]. Furthermore, TNF-α-exposed MSCs exhibit upregulation of SASP molecules, including IL-6 and IL-8 and MCP-1 [64]. However, IL-8 exerts a chemotactic effect on human bone marrow MSC [65] and enhances the angiogenic and proliferation potential of MSCs [66, 67]. However, no positive influence of SASP on MSCs has been reported so far.

Extracellular Vesicles

Acting in a similar manner as SASP, extracellular vesicles (EVs), including exosomes from the endosomal compartment, and endosomes, microvesicles, and apoptotic bodies shredded from the plasma membrane, have recently been described as a new mechanism of cell-cell communication. These lipid-based carriers shuttle between cells, delivering their microRNAs, messenger RNAs, DNA, and proteins to target cells via endocytosis and membrane fusion.

A senescence-associated increase in EV secretion was first described by Lehmann et al. [68] and was reported to induce senescence of adjacent cells subsequently [69-73] and to promote the proliferation and inflammation of cancer cells [74]. With aging, SNC-derived microRNA-183-5p suppresses MSC proliferation and induces its senescence [70]. Moreover, microvesicular miR-31 secreted by SNCs inhibits the osteogenic ability of MSCs [69]. Very long-chain C24:1 ceramide, which is associated with cellular senescence and apoptosis, can induce senescence of MSCs [71]. In addition, SNC-derived EVs exhibit antiapoptotic activity by targeting 5 common proapoptotic mediators (PTEN, P53, APAF-1, CDKN1B, and MYC) [75]. However, it is still unknown which of these miRNAs exert this effect.

EVs are actually a heterogeneous population that can be classified based on their origin [76, 77]. EVs from different cells or tissue have different RNAs or proteins; therefore, it is very important to study EVs from different origins and its pathway to promote senescence for exploring the mechanism of senescence and alleviating age-related diseases. Terlecki-Zaniewicz et al. [75] identified senescence-specific differences in miRNA composition of EVs in senescent human dermal fibroblasts, with an increase in miR-23a-5p and miR-137 and a decrease in miR-625-3p, miR-766-3p, miR-199b-5p, miR-381-3p, and miR-17-3p. EVs from senescent human umbilical vein endothelial cells were enriched in miR-21-5p and miR-217, which target 2 key enzymes in methylation pattern maintenance DNMT1 and SIRT1. Thus, it promotes senescence signal transmission, affects DNA methylation, and modulates senescence features [73].

Given that the function of EVs secreted by SNCs seems to be similar to SASP, increasing with aging, inducing senescence of adjacent cells, and resisting apoptosis, some researchers support that EVs are part of the SASP [75]. However, it is not clear whether EVs play a beneficial role in tissue homeostasis in vivo, such as wound healing or tissue regeneration [78].

Cell Junction

For SNCs, secretory factors are the main mechanism of information transmission between cells, but not the only mechanism. Cell junction is an important way of cell communication between adjacent cells. It has been proved that gap junction-mediated cell-cell contact increases the formation rate of DNA damage foci and steady-state foci levels in bystander cells [79] and induces DNA damage of neighboring cells, thus resulting in its senescence. Anat Biran et al. found that intercellular protein transfer is dependent on cell-cell contact mediated by actin polymerization and regulated primarily by CDC42 [80]. On all these counts; aging microenvironment exacerbates the inflammatory response and decreases the number, biological function, and immune modulation activity of MSCs.

Except that an aging microenvironment hinders the function of transplanted MSCs, senescence of autologous and transplanted MSCs contribute to an unsatisfied therapeutic effect [81]. For example, transplanting adipose-derived MSCs from aged mice, not from young mice, induces physical dysfunction in older recipients [82]. In addition, the adverse effect caused by senescent MSCs is also proved by vitro experiments. Muscle-derived stem/progenitor cells came from old mice or premature aging models of mice show a decreased regeneration function [83]. It implied the importance of evaluating the impact of senescence on biological properties of MSCs and monitoring activities of MSCs preparations before application.

Senescent MSCs are usually characterized by an enlarged, more granular, and flat fried egg morphology, with constrained nuclei and a granular cytoplasm. During aging, the quantity of BM-MSCs with osteogenic potential decreases [84, 85], which may contribute to age-related diseases. Also, self-senescence will affect the quality of MSCs, including proliferation ability, clonal formation ability, differentiation potential, immune characteristics, telomerase activity, cell migration ability, and adhesion [86]. The aforementioned reasons may explain the dissatisfied therapeutic effect of stem cells.

Specifically, aged MSCs exhibit a reduced cell proliferation and osteogenic ability (alkaline phosphatase activity, extracellular matrix mineralization, and osteogenesis-related genes), while an increased adipogenic differentiation ability (adipocyte protein 2, resistin, and lipid accumulation) [87], which contributes to decreased bone formation [88] and results in osteoporosis [89-92] and dissatisfied ability of osseointegration [87]. What is more, senescent MSCs impair basal and calcium-regulated functions of the CXCR4/SDF-1 axis in bone marrow-derived cells and thus exhibit decreased vasculogenesis ability [93]. Senescent MSCs have been proved to have metabolic inflexibility with impairment of proteasome activity and the autophagic flux [94]. They also exhibit a significant decline in the cell colony number, one of the most convenient and common predictive indicators of MSC senescence [95].

Autologous MSC therapies are preferable due to tissue regeneration ability and biosafety concerns (immune rejection) in recent years. Besides, increasing evidence suggests that MSCs may not be immune-privileged [96, 97]. Unfortunately, the appliance of MSCs usually requires in vitro expansion, which exposes cells to replicative senescence. Therapies based on autologous MSCs have been hindered due to decreased quantity and impaired quality of MSCs with aging, as described previously [84]. How to rejuvenate senescent MSCs and improve the function of MSCs is a focus of scholars in regenerative medicine.

Targeting MSCs or Its Derivatives

Improving the Quality of MSCs

Several lines of evidence support the feasibility of extracting high-quality autologous MSCs from older donors. (1) It has been proved that it is the clonal composition of stem cell populations, instead of individual stem cells, that changed with aging. (2) MSCs derived from the elderly population consist of mixtures of cells which contain not only SNCs but also healthy cells [98-100]. (3) With aging and some pathological conditions, SNCs accumulates in various tissues and organs, and the proportion is between 1 and 20% [101]. Once reached a sufficient number, SNCs may do harm to adjacent cells and tissues and drive age-related diseases. Injecting a small number of aging cells (less than 0.03% of all cells in the whole body) into mice can shorten the life span of mice and cause dysfunction of the body [101]. Travis J. Block first isolated a subset of “youthful” cells from elderly donors and then expanded these cells on a “young microenvironment” and thus harvested high-quality MSCs from elderly populations, which suggests that it may be able to store masses of high-quality autologous MSCs from the elderly population for treatment of age-related diseases [102].

Bioactivator-Treated MSCs

Due to long-term culture in vitro, human-derived MSCs were induced cellular senescence, which called replicative senescence. Treated by FGF-2, these cells show a suppressed expression level of p21, p53, and p16 mRNA, as well as decreased percentages of G1 cell growth arrest [103].

Macrophage migration inhibitory factor is an endocrine molecule used to restrict abnormal movement of macrophages. Pretreatment with migration inhibitory factor improved the proliferation, cell viability, trophic effects, telomere length, and telomerase activity of senescent MSCs, which may be due to oxidative stress inhibition and phosphatidylinositol 3-kinase-RAC-α serine/threonine-protein kinase (Akt) signaling pathway activation [104].

Melatonin clears free radicals and reactive species and stimulates a number of antioxidant enzymes and thus protects macromolecules from oxidative damage, possibly by acting on membrane-bound receptors MT1 and MT2 and activating RK/MAPK and phosphatidylinositol 3-kinase/Akt signaling pathways [105].

Resveratrol-treated adult stem cells rescue the quantitative decline of BMSCs, improves bone structural organization, increases mineral density, and significantly alleviates progeroid features and prolongs the life span in Zmpste24−/− mice (a kind of premature aging mice, responsible for prelamin A maturation) [106]. Liu et al. reported that it is associated with enhanced binding between SIRT1 and prelamin A by resveratrol.

Interestingly, IL-6 maintains MSC “stemness,” which may act through the ERK1/2 pathway [107]. The IL-6 and the IL-6 receptor complex activates the downstream signal transducer and activator of transcription (STAT) 3 pathway, thereby promoting osteogenic differentiation of BM-MSCs [108]. However, IL-6 is a pro-inflammatory factor, which can enhance senescence. Therefore, the effects and underlying mechanisms of IL-6 in MSCs and the interaction between these mechanisms require further investigation.

Cell-Free Therapy

Despite some success, many concerns have gained attention regarding the curative effect of aged MSCs because of their poor survival, low retention [109-111], replicative aging, and instability in clinical outcomes [112]. Emerging evidence suggests that much of the therapeutic benefit related to MSC therapy may owe to the biological activities of factors and molecules secreted by these cells. Growth factors, cytokines, and different components of the extracellular matrix have been widely studied as a cell-free therapeutic.

Bioactivator

Growth factors and cytokines can be used to alleviate inflammation, enhance angiogenesis, reduce fibrosis, and promote tissue generation. For example, TGF-β stimulates both Smad3-dependent and independent activation of macrophages, promotes secretion of VEGF and TGF-β1, and protects against adverse cardiac tissue remodeling [113]. These bioactivators usually cooperate with scaffold materials for delivery and sustained release, which contributes to prolonging the impact on surrounding tissues. SDF-1 facilitates cell homing and thus promotes defected cartilage repair when using radially oriented scaffolds to deliver SDF-1 locally [114]. Compared to conventional patch material, VEGF- and bFGF-embedded nanofiber matrices were confirmed to promote vascularization and thus restore cardiac function after ischemia [115].

However, these bioactivators fail to exert an abiding impact on clinical application due to their short half-lives and instability. It is reported that most of these limitations could be overcome by directly delivering gene, instead of the protein [116].

EVs of MSCs

Although several different target genes controlled by some specific signaling pathways and mechanisms were found in MSCs and EVs, MSCs and their EVs shared similar miRNA components [117, 118]. It seems that EVs have a long-term effect on surrounding tissues when used in vitro.

EVs are powerful biological entities released by cells and exhibit anti-inflammatory, antiapoptotic effects, and pro-angiogenetic ability, thus promoting changes in their targets. It has been reported that MSC-EVs play a positive role in various diseases, such as acute kidney injury [119], lung injury [120-123], liver injury [124], neuroinflammation [125], allergic airway inflammation [126], acute graft-versus-host disease [127], autoimmune diseases [128], hair follicle growth [129], bone formation [130], and osseointegration between titanium surfaces and bone [131].

The current EV-based therapy is limited by the purity of EV preparations [132, 133]. The 2 commonly used methods are ultracentrifugation and differential centrifugation [109]. However, repeated ultracentrifugation not only reduces particle yield and recovery but also damages isolated vesicles, reducing their quality [133, 134]. Protein precipitation protocols are frequently used as well. Richard J. Lobb confirmed that ultrafiltration was far more time-efficient than ultracentrifugation [133]. Ultrafiltration combined with ultracentrifugation is a highly purified method because it removes non-EV-associated proteins from EVs [135]. Besides, some scholars have integrated many methods that separated free secreted proteins from EVs [136] (e.g., immunoaffinity isolation [137], polymer-based precipitation, density gradient, anion exchange [138], and microfluidics [139]). It is essential to standardize and optimize EV purification protocols.

Apoptotic body is a member of EVs. It has been reported that apoptotic cells may stimulate progenitor or stem cell proliferation, thus replacing damaged cells and promoting tissue regeneration [140, 141]. Subsequently, Liu et al. [142] showed that coculture of apoptotic bodies from MSCs from normal mouse bone marrow with MRL/lpr and cas3-/-mouse MSCs in vitro improves proliferation, osteogenic differentiation, new bone formation, and adipogenic differentiation. In addition, apoptotic bodies are capable of maintaining MSC homeostasis by directly regulating the WNT/β-catenin pathway [142, 143]. In this sense, apoptotic bodies from MSCs are promising and potential tools in tissue engineering. How to accelerate its clinical application still needs efforts.

Targeting Senescence

Another way to reinforce MSC therapy in aging is preventing senescence or reducing SNC burden. Senescence does harm to the quantity and quality of MSCs and the etiology of numerous tissues; therefore, disrupting or preventing senescence and its deleterious effects may be a promising way to delay health decline during aging. There are 4 strategies to alleviating these effects: suppressing the formation of SNCs, eliminating already formed SNCs, inhibiting the secretion of SASP, and cleaning up secreted SASP. Here, we discuss each of aforementioned therapeutic strategies of senescence, summarize the latest progress of these strategies, and emphasize their relative advantages and side effects.

Inhibit the Formation Mechanism of SNCs

Arguably, one of the most effective ways to target senescence is to start from the origin – preventing the production of SNCs, which is mainly based on epigenetics currently. However, interfering with epigenetics of senescence may lead to cancer [144]. Interventions targeting the formation of SNCs, such as knocking down the expression of p16Ink4a or p53 or short-term cyclic expression of Oct4, Sox2, Klf4, and c-Myc in mice, ameliorate aging features but promote the development of cancer [144, 145]. Based on Ocampo [144], Y. Lu et al. transplant, 3 of above genes that had nothing to do with cancer cells were used to devise a drug which controls the expression of these genes. This research regenerated nerve from injured eye cells and alleviated age-related vision loss in mice [146], but it needs safety assessment and considerable refinement before it can be deployed safely in humans.

To date, no study targeting the epigenetic of senescence has been conducted in human. One pivotal problem concerning inhibiting generation signaling of SNCs is how to prevent the formation of SNCs without increasing the risk of cancer and other side effects.

Eliminating SNCs

Eliminating SNCs is the most widely studied method to target senescence, which delays several pathologies and increases healthy life span [147, 148]. Some agents have been put into clinical practice. Among them, most products are mainly by inducing apoptosis and activating the immune system to eliminate SNCs.

The BCL-2 gene family plays a central role in modulating programmed cell death by the intrinsic apoptosis pathway [149-152]. The multiregional (BH1-4) anti-apoptotic proteins, such as BCL-2, BCL-XL, BCL-W, myeloid cell leukemia 1, and A1, antagonize proapoptotic BH3-only proteins and suppress the essential apoptosis effectors such as BCL-2 antagonist killer 1 and BCL-2-associated X protein, which leads to the release of cytochrome C, thus driving programmed cell death [153, 154].

ABT-737, the first discovered BCL-2 and BCL-XL inhibitor [155], has been widely used to study and define apoptosis biology. ABT-737 efficiently removes DNA damage-induced SNCs in the lungs as well as P53 activation-induced SNCs in the epidermis [156], enhancing liver regeneration [157]. However, the poor oral absorption of ABT-737 contributed to the emerging of its second-generation orally available analog, navitoclax (also known as ABT-263). Navitoclax, a BCL-2 and BCL-xl dual inhibitor, is one of the most effective and broad-spectrum BCL-2 family senolytic discovered so far [158-161]. Unfortunately, because platelets solely depend on BCL-xl for survival [149, 162-164], BCL-xl inhibition, including ABT263 and other small molecular inhibitors, induces platelet apoptosis and leads to severe thrombocytopenia [149, 162, 163]. Yonghan He et al. modified navitoclax using proteolysis-targeting chimera technology, improved senolytic activity, and, importantly, reduced platelet toxicity [161, 165, 166]. Unlike BCL-xl, BCL-2 is dispensable for generation and survival of platelet in mice and humans [166, 167]. Inhibition of BCL-2 does not induce thrombocytopenia, such as ABT199 (also known as venetoclax) [164]. How to decrease clinical side effects or improve ability to eliminate BCL inhibitor deserves more exploration.

Dasatinib (D) is a synthetic small-molecule inhibitor of Src family tyrosine kinase (TYK), which is used to treat chronic myelogenous leukemia that is positive for the Philadelphia chromosome. D promotes apoptosis caused by dependence receptors, such as the ephrins, in part by inhibiting Src family TYK [168, 169]. Quercetin (Q) is a natural bioflavonoid and antioxidant, which exists in many plants. SNCs with a wide range of cell types are selectively eliminated by treatment with the D plus Q (D + Q) [42]. Interestingly, D nor Q alone can only kill certain cell types of SNCs, while the combination of D and Q can achieve additive benefits [170]. Furthermore, D + Q has been proved to remove naturally occurring SNCs in human adipose tissue, accompanied by an increase in cleaved caspase-3 [101], confirming that D + Q is effective to kill human SNCs partly by inducing apoptosis. However, whether other pathways have such effects still needs further study. D + Q has made significant progress in clinical trials, such as age-related osteoporosis, diabetic chronic kidney disease [171], and idiopathic pulmonary fibrosis [172], indicating D + Q has great prospects as a senolytic.

Fisetin, a natural flavonoid, is another senolytic, which acts in part by inhibiting BCL-2 family members, such as BCL-xL and HIF-1a. Fisetin has been shown to eliminate SNCs in vivo [159, 173], while not hurting non-SNCs [174]. Importantly, no adverse effects of fisetin have been reported, even when administrated at high doses [175].

Furthermore, other senolytics, such as UBX0101, A1331852, or A1155463, are being studied [176, 177]. However, research results are less impressive. Unity, a pioneer biotechnology company in the research of senolytic drugs in the USA announced that its flagship product UBX0101 failed to beat placebo in a phase II clinical trial of osteoarthritis involving 183 people. Several major concerns remain to be solved, including possible side effects and optimal drug delivery mode before we can apply these treatments in clinic.

The immune system clear SNCs is an emerging therapeutic opportunity [178]. Natural killer cells, macrophages, B cells, T cells, neutrophils, and mast cells are involved. Some agents have been proposed to clear SNCs, such as DC vaccines and development of Abs against MDA-vimentin [179, 180]. Besides, SNCs derived from cell types outside the formal immune system could essentially be considered as an integral component of the immune system. For example, senescent stellate cells stimulate T cells, activate potently natural killer cell proliferation in vivo, which in turn kill senescent stellate cells and appear to contribute to limitation of liver fibrosis [181, 182].

Inhibition of SASP Secretion Pathway

The Janus kinase (JAK)/STAT pathway is a common pathway of many cytokine signal transduction, which is widely involved in cell proliferation, differentiation, apoptosis, and inflammation. It is highly activated in SNCs than non-SNCs. The JAK family consists of 4 members, including JAK1, JAK2, JAK3, and TYK2. These 4 members act on downstream STAT proteins to regulate a series of biological reactions. Among them, JAK1 and 2 play important roles in inflammatory signaling and other endocrine signaling.

Inhibition of the JAK pathway downregulates the secretion of SASP, including IL-6, MCP-1, and TNF-α, and alleviates age-related dysfunction [182-185]. For example, JAK inhibitors have been found to alleviate senescence of BMSCs, enhance osteogenic differentiation [186], enhance muscle stem cell function, promote muscle regeneration [187], improve antitumor responses by reprogramming the SASP in SNCs [188], and promote hair growth. Furthermore, some agents have been put into clinical implementation. Ruxolitinib, a specific JAK1/2 inhibitor, has been approved by the Food and Drug Administration (FDA) for the treatment of myelofibrosis.

In addition to JAK inhibitors, other compounds have been proved to inhibit inflammation-related SASP factors targeting the NF-κB pathway. For example, metformin, a commonly prescribed drug for treating type 2 diabetes, has decreased the secretion of SASP factors of oncogene-induced SNCs by inhibiting NF-κB activation. It has been proved to inhibit inflammatory reaction, increase insulin sensitivity, reduce lipotoxicity, and alleviate physical dysfunction in aged mice. Glucocorticoids and rapamycin (an MTOR inhibitor) have decreased IL1A production, which establishes and maintains the SASP by activating NF-κB, thus decreased the secretion of inflammation-related SASP factors, postpone aging, and extend life span.

SNCs derived from different cell types have overlapping but distinct molecular signatures. Different senolytics target specific SASP molecules. How to select senolytics in disease and how to alleviate side effects of senolytics must be considered.

Inhibiting Individual Components of the SASP

Blocking of individual components of SASP components or their receptors, such as TGF-β or its receptor, has the advantage of a clear target and therefore potentially decreases the risk of off-target effects. However, a drawback of this method is that only one of many SASP factors can be targeted at a time. Up to now, no research of such agents in aging or senescence has been implemented.

Although therapies targeted at SNCs and their secretions have gained some achievements, these methods are limited to eliminate SNCs or suppress their SASP, without reversing them to non-SNCs. The possibility of turning SNCs into normal and functional cells is worth exploring.

As mentioned previously, aging microenvironment impairs the function of MSCs, induces cellular senescence, undermines various tissue and organ, and promotes age-associated diseases. However, senescence is not always harmful but offers a variety of benefits, such as tumor suppression [189-191], embryonic development [192, 193], and wound healing [194-196]. One thing to be considered is whether suppressing or removing SNCs can promote tumors or delay wound healing. It is highly valuable to investigate how to minimize the harmful effects of eliminating SNCs.

Advanced age is associated with dysfunction of different tissues and organs and pharmacokinetic changes, most of which are converted into drug accumulation in the body, leads to a higher possibility of drug toxicity [197]. Furthermore, the elderly are more susceptible to the adverse effects of polypharmacy. The increase in the number of medications increases the possibility of interaction between clinically important drugs and adverse drug effects [198]. Interaction between senolytics and MSC therapies and its influence on body needs further research.

In addition, systemic or local administration, intermittent or continuous administration, and how to minimize the side effect must be addressed for the optimization of senolytics usage in the aged people. It is reported that intermittent administration to eliminate SNCs (such as once every 2 weeks or even once every month) has been proved to effectively delay the aging process and has undetectable side effects [40, 42, 101].

In conclusion, MSC therapy has achieved great success and entered clinical trials. The potential of therapeutically targeting SNCs and their secretions is beginning to emerge. However, senescence mechanism and regulatory genes of aging still need further research. It remains unknown whether there is any gene or molecule that can reverse SNCs to normal and functional cells without causing cancer. In this case, it will make milestone progress in the alleviation of aging-related diseases and extend the life with high quality.

The authors have no conflicts of interest to disclose.

Key R&D projects in Hunan Province: mechanism of miR-287 on osteogenic differentiation of bone marrow mesenchymal stem cells from the alveolar cleft and its early clinical diagnosis and treatment.

Yueying Zhou conceptualized and revised the review; Huan Chen wrote the review; Ousheng Liu and Sijia Chen contributed to the revision of the review.

1.
Yokoi
T
,
Uemura
T
,
Takamatsu
K
,
Shintani
K
,
Onode
E
,
Okada
M
,
Bioabsorbable nerve conduits coated with induced pluripotent stem cell-derived neurospheres enhance axonal regeneration in sciatic nerve defects in aged mice
.
J Biomed Mater Res B Appl Biomater
.
2018
;
106
(
5
):
1752
8
. .
2.
Pittenger
MF
,
Mackay
AM
,
Beck
SC
,
Jaiswal
RK
,
Douglas
R
,
Mosca
JD
,
Multilineage potential of adult human mesenchymal stem cells
.
Science
.
1999
;
284
(
5411
):
143
7
. .
3.
Caplan
AI
.
MSCs: the sentinel and safe-guards of injury
.
J Cell Physiol
.
2016
;
231
(
7
):
1413
6
. .
4.
Park
D
,
Spencer
JA
,
Koh
BI
,
Kobayashi
T
,
Fujisaki
J
,
Clemens
TL
,
Endogenous bone marrow MSCs are dynamic, fate-restricted participants in bone maintenance and regeneration
.
Cell Stem Cell
.
2012
;
10
(
3
):
259
72
. .
5.
Gao
X
,
Lu
A
,
Tang
Y
,
Schneppendahl
J
,
Liebowitz
AB
,
Scibetta
AC
,
Influences of donor and host age on human muscle-derived stem cell-mediated bone regeneration
.
Stem Cell Res Ther
.
2018
;
9
(
1
):
316
. .
6.
Gorgoulis
V
,
Adams
PD
,
Alimonti
A
,
Bennett
DC
,
Bischof
O
,
Bishop
C
,
Cellular senescence: defining a path forward
.
Cell
.
2019
;
179
(
4
):
813
27
. .
7.
Zhang
DY
,
Wang
HJ
,
Tan
YZ
.
Wnt/β-catenin signaling induces the aging of mesenchymal stem cells through the DNA damage response and the p53/p21 pathway
.
PLoS One
.
2011
;
6
(
6
):
e21397
. .
8.
Bickford
PC
,
Kaneko
Y
,
Grimmig
B
,
Pappas
C
,
Small
B
,
Sanberg
CD
,
Nutraceutical intervention reverses the negative effects of blood from aged rats on stem cells
.
Age (Dordr)
.
2015
;
37
(
5
):
103
; .
9.
Rebo
J
,
Mehdipour
M
,
Gathwala
R
,
Causey
K
,
Liu
Y
,
Conboy
MJ
,
A single heterochronic blood exchange reveals rapid inhibition of multiple tissues by old blood
.
Nat Commun
.
2016
;
7
:
13363
. .
10.
Hanks
S
,
Coleman
K
,
Reid
S
,
Plaja
A
,
Firth
H
,
Fitzpatrick
D
,
Constitutional aneuploidy and cancer predisposition caused by biallelic mutations in BUB1B
.
Nat Genet
.
2004
;
36
(
11
):
1159
61
. .
11.
Oshima
J
,
Sidorova
JM
,
Monnat
RJ
.
Werner syndrome: clinical features, pathogenesis and potential therapeutic interventions
.
Ageing Res Rev
.
2017
;
33
:
105
14
. .
12.
Mohamad Kamal
NS
,
Safuan
S
,
Shamsuddin
S
,
Foroozandeh
P
.
Aging of the cells: insight into cellular senescence and detection methods
.
Eur J Cell Biol
.
2020
;
99
(
6
):
151108
. .
13.
Karatza
C
,
Stein
WD
,
Shall
S
.
Kinetics of in vitro ageing of mouse embryo fibroblasts
.
J Cell Sci
.
1984
;
65
:
163
75
. .
14.
Anerillas
C
,
Abdelmohsen
K
,
Gorospe
M
.
Regulation of senescence traits by MAPKs
.
GeroScience
.
2020
;
42
(
2
):
397
408
. .
15.
Sun
X
,
Liu
B
,
Wang
J
,
Li
J
,
Ji
WY
.
Inhibition of p21-activated kinase 4 expression suppresses the proliferation of Hep-2 laryngeal carcinoma cells via activation of the ATM/Chk1/2/p53 pathway
.
Int J Oncol
.
2013
;
42
(
2
):
683
9
. .
16.
Burton
DG
,
Faragher
RG
.
Cellular senescence: from growth arrest to immunogenic conversion
.
Age (Dordr)
.
2015
;
37
(
2
):
27
. .
17.
Childs
BG
,
Baker
DJ
,
Kirkland
JL
,
Campisi
J
,
van Deursen
JM
.
Senescence and apoptosis: dueling or complementary cell fates?
EMBO Rep
.
2014
;
15
(
11
):
1139
53
. .
18.
Lopes-Paciencia
S
,
Saint-Germain
E
,
Rowell
MC
,
Ruiz
AF
,
Kalegari
P
,
Ferbeyre
G
.
The senescence-associated secretory phenotype and its regulation
.
Cytokine
.
2019
;
117
:
15
22
. .
19.
Herranz
N
,
Gallage
S
,
Mellone
M
,
Wuestefeld
T
,
Klotz
S
,
Hanley
CJ
,
mTOR regulates MAPKAPK2 translation to control the senescence-associated secretory phenotype
.
Nat Cell Biol
.
2015
;
17
(
9
):
1205
17
. .
20.
Kang
C
,
Xu
Q
,
Martin
TD
,
Li
MZ
,
Demaria
M
,
Aron
L
,
The DNA damage response induces inflammation and senescence by inhibiting autophagy of GATA4
.
Science
.
2015
;
349
(
6255
):
aaa5612
. .
21.
Glück
S
,
Guey
B
,
Gulen
MF
,
Wolter
K
,
Kang
T-W
,
Schmacke
NA
,
Innate immune sensing of cytosolic chromatin fragments through cGAS promotes senescence
.
Nat Cell Biol
.
2017
;
19
(
9
):
1061
70
. .
22.
Dimri
GP
,
Lee
X
,
Basile
G
,
Acosta
M
,
Scott
G
,
Roskelley
C
,
A biomarker that identifies senescent human cells in culture and in aging skin in vivo
.
Proc Natl Acad Sci USA
.
1995
;
92
(
20
):
9363
7
. .
23.
Sharpless
NE
,
Sherr
CJ
.
Forging a signature of in vivo senescence
.
Nat Rev Cancer
.
2015
;
15
(
7
):
397
408
. .
24.
Liu
S
,
Wang
X
,
Zhao
Q
,
Liu
S
,
Zhang
H
,
Shi
J
,
Senescence of human skin-derived precursors regulated by Akt-FOXO3-p27(KIP1)/p15(INK⁴b) signaling
.
Cell Mol Life Sci
.
2015
;
72
(
15
):
2949
60
.
25.
Bai
H
,
Gao
Y
,
Hoyle
DL
,
Cheng
T
,
Wang
ZZ
.
Suppression of transforming growth factor-β signaling delays cellular senescence and preserves the function of endothelial cells derived from human pluripotent stem cells
.
Stem Cells Transl Med
.
2017
;
6
(
2
):
589
600
. .
26.
He
J
,
Kallin
EM
,
Tsukada
Y
,
Zhang
Y
.
The H3K36 demethylase Jhdm1b/Kdm2b regulates cell proliferation and senescence through p15(Ink4b)
.
Nat Struct Mol Biol
.
2008
;
15
(
11
):
1169
75
. .
27.
Wang
ZL
,
Chen
LB
,
Qiu
Z
,
Chen
XB
,
Liu
Y
,
Li
J
,
Ginsenoside Rg1 ameliorates testicular senescence changes in Dgalinduced aging mice via antiinflammatory and antioxidative mechanisms
.
Mol Med Rep
.
2018
;
17
(
5
):
6269
76
. .
28.
Novais
EJ
,
Diekman
BO
,
Shapiro
IM
,
Risbud
MV
.
p16Ink4a deletion in cells of the intervertebral disc affects their matrix homeostasis and senescence associated secretory phenotype without altering onset of senescence
.
Matrix Biol
.
2019
;
82
:
54
70
. .
29.
Hilpert
M
,
Legrand
C
,
Bluteau
D
,
Balayn
N
,
Betems
A
,
Bluteau
O
,
p19 INK4d controls hematopoietic stem cells in a cell-autonomous manner during genotoxic stress and through the microenvironment during aging
.
Stem cell reports
.
2014
;
3
(
6
):
1085
102
. .
30.
Tang
H
,
Fan
X
,
Xing
J
,
Liu
Z
,
Jiang
B
,
Dou
Y
,
NSun2 delays replicative senescence by repressing p27 (KIP1) translation and elevating CDK1 translation
.
Aging (Albany NY)
.
2015
;
7
(
12
):
1143
58
. .
31.
Sang
B
,
Zhang
YY
,
Guo
ST
,
Kong
LF
,
Cheng
Q
,
Liu
GZ
,
Dual functions for OVAAL in initiation of RAF/MEK/ERK prosurvival signals and evasion of p27-mediated cellular senescence
.
Proc Natl Acad Sci USA
.
2018
;
115
(
50
):
E11661
70
. .
32.
Beck
J
,
Turnquist
C
,
Horikawa
I
,
Harris
C
.
Targeting cellular senescence in cancer and aging: roles of p53 and its isoforms
.
Carcinogenesis
.
2020
;
41
(
8
):
1017
29
. .
33.
Ke
Y
,
Li
D
,
Zhao
M
,
Liu
C
,
Liu
J
,
Zeng
A
,
Gut flora-dependent metabolite Trimethylamine-N-oxide accelerates endothelial cell senescence and vascular aging through oxidative stress
.
Free Radic Biol Med
.
2018
;
116
:
88
100
. .
34.
Castro-Garcia
P
,
Díaz-Moreno
M
,
Gil-Gas
C
,
Fernández-Gómez
FJ
,
Honrubia-Gómez
P
,
Álvarez-Simón
CB
,
Defects in subventricular zone pigmented epithelium-derived factor niche signaling in the senescence-accelerated mouse prone-8
.
FASEB J
.
2015
;
29
(
4
):
1480
92
. .
35.
Swanson
EC
,
Manning
B
,
Zhang
H
,
Lawrence
JB
.
Higher-order unfolding of satellite heterochromatin is a consistent and early event in cell senescence
.
J Cell Biol
.
2013
;
203
(
6
):
929
42
. .
36.
Biran
A
,
Zada
L
,
Abou Karam
P
,
Vadai
E
,
Roitman
L
,
Ovadya
Y
,
Quantitative identification of senescent cells in aging and disease
.
Aging Cell
.
2017
;
16
(
4
):
661
71
. .
37.
Zhu
Y
,
Xu
L
,
Zhang
J
,
Hu
X
,
Liu
Y
,
Yin
H
,
Sunitinib induces cellular senescence via p53/Dec1 activation in renal cell carcinoma cells
.
Cancer Sci
.
2013
;
104
(
8
):
1052
61
. .
38.
Pare
R
,
Soon
PS
,
Shah
A
,
Lee
CS
.
Differential expression of senescence tumour markers and its implications on survival outcomes of breast cancer patients
.
PLoS One
.
2019
;
14
(
4
):
e0214604
. .
39.
Campo-Trapero
J
,
Cano-Sánchez
J
,
Palacios-Sánchez
B
,
Llamas-Martínez
S
,
Lo Muzio
L
,
Bascones-Martínez
A
.
Cellular senescence in oral cancer and precancer and treatment implications: a review
.
Acta Oncol
.
2008
;
47
(
8
):
1464
74
. .
40.
Farr
JN
,
Xu
M
,
Weivoda
MM
,
Monroe
DG
,
Fraser
DG
,
Onken
JL
,
Targeting cellular senescence prevents age-related bone loss in mice
.
Nat Med
.
2017
;
23
(
9
):
1072
9
. .
41.
Aquino-Martinez
R
,
Rowsey
JL
,
Fraser
DG
,
Eckhardt
BA
,
Khosla
S
,
Farr
JN
,
LPS-induced premature osteocyte senescence: implications in inflammatory alveolar bone loss and periodontal disease pathogenesis
.
Bone
.
2020
;
132
:
115220
. .
42.
Schafer
MJ
,
White
TA
,
Iijima
K
,
Haak
AJ
,
Ligresti
G
,
Atkinson
EJ
,
Cellular senescence mediates fibrotic pulmonary disease
.
Nat Commun
.
2017
;
8
:
14532
. .
43.
Hallam
D
,
Wan
T
,
Saretzki
G
.
Dietary restriction mitigates age-related accumulation of DNA damage, but not all changes in mouse corneal epithelium
.
Exp Gerontol
.
2015
;
67
:
72
9
. .
44.
Qian
W
,
Kumar
N
,
Roginskaya
V
,
Fouquerel
E
,
Opresko
PL
,
Shiva
S
,
Chemoptogenetic damage to mitochondria causes rapid telomere dysfunction
.
Proc Natl Acad Sci USA
.
2019
;
116
(
37
):
18435
44
. .
45.
Takai
H
,
Smogorzewska
A
,
de Lange
T
.
DNA damage foci at dysfunctional telomeres
.
Curr Biol
.
2003
;
13
(
17
):
1549
56
. .
46.
Farr
JN
,
Fraser
DG
,
Wang
H
,
Jaehn
K
,
Ogrodnik
MB
,
Weivoda
MM
,
Identification of senescent cells in the bone microenvironment
.
J Bone Miner Res
.
2016
;
31
(
11
):
1920
9
. .
47.
Ameh
OI
,
Okpechi
IG
,
Dandara
C
,
Kengne
AP
.
Association between telomere length, chronic kidney disease, and renal traits: a systematic review
.
OMICS
.
2017
;
21
(
3
):
143
55
. .
48.
Narita
M
,
Nũnez
S
,
Heard
E
,
Narita
M
,
Lin
AW
,
Hearn
SA
,
Rb-mediated heterochromatin formation and silencing of E2F target genes during cellular senescence
.
Cell
.
2003
;
113
(
6
):
703
16
. .
49.
Shi
X
,
Tian
B
,
Liu
L
,
Gao
Y
,
Ma
C
,
Mwichie
N
,
Rb protein is essential to the senescence-associated heterochromatic foci formation induced by HMGA2 in primary WI38 cells
.
J Genet Genomics
.
2013
;
40
(
8
):
391
8
. .
50.
Zhang
Y
,
Gao
Y
,
Zhao
L
,
Han
L
,
Lu
Y
,
Hou
P
,
Mitogen-activated protein kinase p38 and retinoblastoma protein signalling is required for DNA damage-mediated formation of senescence-associated heterochromatic foci in tumour cells
.
FEBS J
.
2013
;
280
(
18
):
4625
39
. .
51.
Lee
JJ
,
Park
IH
,
Rhee
WJ
,
Kim
HS
,
Shin
JS
.
HMGB1 modulates the balance between senescence and apoptosis in response to genotoxic stress
.
FASEB J
.
2019
;
33
(
10
):
10942
53
. .
52.
Lu
H
,
Zhang
Z
,
Barnie
PA
,
Su
Z
.
Dual faced HMGB1 plays multiple roles in cardiomyocyte senescence and cardiac inflammatory injury
.
Cytokine Growth Factor Rev
.
2019
;
47
:
74
82
. .
53.
Wu
D
,
Wang
Z
,
Zheng
Z
,
Geng
Y
,
Zhang
Z
,
Li
Q
,
Effects of physiological aging factor on bone tissue engineering repair based on fetal BMSCs
.
J Transl Med
.
2018
;
16
(
1
):
324
. .
54.
Conboy
IM
,
Conboy
MJ
,
Wagers
AJ
,
Girma
ER
,
Weissman
IL
,
Rando
TA
.
Rejuvenation of aged progenitor cells by exposure to a young systemic environment
.
Nature
.
2005
;
433
(
7027
):
760
4
. .
55.
Liu
L
,
Rando
TA
.
Manifestations and mechanisms of stem cell aging
.
J Cell Biol
.
2011
;
193
(
2
):
257
66
. .
56.
Khosla
S
,
Farr
JN
,
Tchkonia
T
,
Kirkland
JL
.
The role of cellular senescence in ageing and endocrine disease
.
Nat Rev Endocrinol
.
2020
;
16
(
5
):
263
75
. .
57.
Acosta
JC
,
Banito
A
,
Wuestefeld
T
,
Georgilis
A
,
Janich
P
,
Morton
JP
,
A complex secretory program orchestrated by the inflammasome controls paracrine senescence
.
Nat Cell Biol
.
2013
;
15
(
8
):
978
90
. .
58.
Hoare
M
,
Ito
Y
,
Kang
TW
,
Weekes
MP
,
Matheson
NJ
,
Patten
DA
,
NOTCH1 mediates a switch between two distinct secretomes during senescence
.
Nat Cell Biol
.
2016
;
18
(
9
):
979
92
. .
59.
Debacq-Chainiaux
F
,
Borlon
C
,
Pascal
T
,
Royer
V
,
Eliaers
F
,
Ninane
N
,
Repeated exposure of human skin fibroblasts to UVB at subcytotoxic level triggers premature senescence through the TGF-beta1 signaling pathway
.
J Cell Sci
.
2005
;
118
:
743
58
. .
60.
Severino
V
,
Alessio
N
,
Farina
A
,
Sandomenico
A
,
Cipollaro
M
,
Peluso
G
,
Insulin-like growth factor binding proteins 4 and 7 released by senescent cells promote premature senescence in mesenchymal stem cells
.
Cell Death Dis
.
2013
;
4
:
e911
. .
61.
Wu
J
,
Wang
C
,
Miao
X
,
Wu
Y
,
Yuan
J
,
Ding
M
,
Age-related insulin-like growth factor binding protein-4 overexpression inhibits osteogenic differentiation of rat mesenchymal stem cells
.
Cell Physiol Biochem
.
2017
;
42
(
2
):
640
50
. .
62.
Kumar
S
,
Suman
S
,
Fornace
AJ
,
Datta
K
.
Intestinal stem cells acquire premature senescence and senescence associated secretory phenotype concurrent with persistent DNA damage after heavy ion radiation in mice
.
Aging (Albany NY)
.
2019
;
11
(
12
):
4145
58
. .
63.
Chen
L
,
Xia
W
,
Hou
M
.
Mesenchymal stem cells attenuate doxorubicininduced cellular senescence through the VEGF/Notch/TGFβ signaling pathway in H9c2 cardiomyocytes
.
Int J Mol Med
.
2018
;
42
(
1
):
674
84
. .
64.
Suvakov
S
,
Cubro
H
,
White
WM
,
Butler Tobah
YS
,
Weissgerber
TL
,
Jordan
KL
,
Targeting senescence improves angiogenic potential of adipose-derived mesenchymal stem cells in patients with preeclampsia
.
Biol Sex Differ
.
2019
;
10
(
1
):
49
. .
65.
Mishima
Y
,
Lotz
M
.
Chemotaxis of human articular chondrocytes and mesenchymal stem cells
.
J Orthop Res
.
2008
;
26
(
10
):
1407
12
. .
66.
Hou
Y
,
Ryu
CH
,
Jun
JA
,
Kim
SM
,
Jeong
CH
,
Jeun
SS
.
IL-8 enhances the angiogenic potential of human bone marrow mesenchymal stem cells by increasing vascular endothelial growth factor
.
Cell Biol Int
.
2014
;
38
(
9
):
1050
9
. .
67.
Jeon
BJ
,
Yang
Y
,
Kyung Shim
S
,
Yang
HM
,
Cho
D
,
Ik Bang
S
.
Thymosin beta-4 promotes mesenchymal stem cell proliferation via an interleukin-8-dependent mechanism
.
Exp Cell Res
.
2013
;
319
(
17
):
2526
34
. .
68.
Lehmann
BD
,
Paine
MS
,
Brooks
AM
,
McCubrey
JA
,
Renegar
RH
,
Wang
R
,
Senescence-associated exosome release from human prostate cancer cells
.
Cancer Res
.
2008
;
68
(
19
):
7864
71
. .
69.
Weilner
S
,
Schraml
E
,
Wieser
M
,
Messner
P
,
Schneider
K
,
Wassermann
K
,
Secreted microvesicular miR-31 inhibits osteogenic differentiation of mesenchymal stem cells
.
Aging Cell
.
2016
;
15
(
4
):
744
54
. .
70.
Davis
C
,
Dukes
A
,
Drewry
M
,
Helwa
I
,
Johnson
MH
,
Isales
CM
,
MicroRNA-183-5p Increases with Age in Bone-Derived Extracellular Vesicles, Suppresses Bone Marrow Stromal (Stem) Cell Proliferation, and Induces Stem Cell Senescence
.
Tissue Eng Part A
.
2017
;
23
:
1231
40
. .
71.
Khayrullin
A
,
Krishnan
P
,
Martinez-Nater
L
,
Mendhe
B
,
Fulzele
S
,
Liu
Y
,
Very long-chain C24:1 ceramide is increased in serum extracellular vesicles with aging and can induce senescence in bone-derived mesenchymal stem cells
.
Cells
.
2019
;
8
(
1
). .
72.
Borghesan
M
,
Fafián-Labora
J
,
Eleftheriadou
O
,
Carpintero-Fernández
P
,
Paez-Ribes
M
,
Vizcay-Barrena
G
,
Small extracellular vesicles are key regulators of non-cell autonomous intercellular communication in senescence via the interferon protein IFITM3
.
Cell Rep
.
2019
;
27
(
13
):
3956
e6
. .
73.
Mensà
E
,
Guescini
M
,
Giuliani
A
,
Bacalini
MG
,
Ramini
D
,
Corleone
G
,
Small extracellular vesicles deliver miR-21 and miR-217 as pro-senescence effectors to endothelial cells
.
J Extracell Vesicles
.
2020
;
9
(
1
):
1725285
. .
74.
Takasugi
M
.
Emerging roles of extracellular vesicles in cellular senescence and aging
.
Aging Cell
.
2018
;
17
(
2
). .
75.
Terlecki-Zaniewicz
L
,
Lämmermann
I
,
Latreille
J
,
Bobbili
MR
,
Pils
V
,
Schosserer
M
,
Small extracellular vesicles and their miRNA cargo are anti-apoptotic members of the senescence-associated secretory phenotype
.
Aging (Albany NY)
.
2018
;
10
(
5
):
1103
32
. .
76.
Bobrie
A
,
Colombo
M
,
Krumeich
S
,
Raposo
G
,
Théry
C
.
Diverse subpopulations of vesicles secreted by different intracellular mechanisms are present in exosome preparations obtained by differential ultracentrifugation
.
J Extracell Vesicles
.
2012
;
1
. .
77.
Kowal
J
,
Arras
G
,
Colombo
M
,
Jouve
M
,
Morath
JP
,
Primdal-Bengtson
B
,
Proteomic comparison defines novel markers to characterize heterogeneous populations of extracellular vesicle subtypes
.
Proc Natl Acad Sci USA
.
2016
;
113
(
8
):
E968
77
. .
78.
Raposo
G
,
Stoorvogel
W
.
Extracellular vesicles: exosomes, microvesicles, and friends
.
J Cell Biol
.
2013
;
200
(
4
):
373
83
. .
79.
Nelson
G
,
Wordsworth
J
,
Wang
C
,
Jurk
D
,
Lawless
C
,
Martin-Ruiz
C
,
A senescent cell bystander effect: senescence-induced senescence
.
Aging Cell
.
2012
;
11
(
2
):
345
9
. .
80.
Biran
A
,
Perelmutter
M
,
Gal
H
,
Burton
DG
,
Ovadya
Y
,
Vadai
E
,
Senescent cells communicate via intercellular protein transfer
.
Genes Dev
.
2015
;
29
(
8
):
791
802
. .
81.
Yamaguchi
S
,
Horie
N
,
Satoh
K
,
Ishikawa
T
,
Mori
T
,
Maeda
H
,
Age of donor of human mesenchymal stem cells affects structural and functional recovery after cell therapy following ischaemic stroke
.
J Cereb Blood Flow Metab
.
2018
;
38
(
7
):
1199
212
. .
82.
Wang
B
,
Liu
Z
,
Chen
VP
,
Wang
L
,
Inman
CL
,
Zhou
Y
,
Transplanting cells from old but not young donors causes physical dysfunction in older recipients
.
Aging Cell
.
2020
;
19
(
3
):
e13106
. .
83.
Lavasani
M
,
Robinson
AR
,
Lu
A
,
Song
M
,
Feduska
JM
,
Ahani
B
,
Muscle-derived stem/progenitor cell dysfunction limits healthspan and lifespan in a murine progeria model
.
Nat Commun
.
2012
;
3
:
608
. .
84.
Baker
N
,
Boyette
LB
,
Tuan
RS
.
Characterization of bone marrow-derived mesenchymal stem cells in aging
.
Bone
.
2015
;
70
:
37
47
. .
85.
D'Ippolito
G
,
Schiller
PC
,
Ricordi
C
,
Roos
BA
,
Howard
GA
.
Age-related osteogenic potential of mesenchymal stromal stem cells from human vertebral bone marrow
.
J Bone Miner Res
.
1999
;
14
(
7
):
1115
22
. .
86.
Lin
H
,
Sohn
J
,
Shen
H
,
Langhans
MT
,
Tuan
RS
.
Bone marrow mesenchymal stem cells: aging and tissue engineering applications to enhance bone healing
.
Biomaterials
.
2019
;
203
:
96
110
. .
87.
Abuna
RP
,
Stringhetta-Garcia
CT
,
Fiori
LP
,
Dornelles
RC
,
Rosa
AL
,
Beloti
MM
.
Aging impairs osteoblast differentiation of mesenchymal stem cells grown on titanium by favoring adipogenesis
.
J Appl Oral Sci
.
2016
;
24
(
4
):
376
82
. .
88.
Marędziak
M
,
Marycz
K
,
Tomaszewski
KA
,
Kornicka
K
,
Henry
BM
.
The Influence of Aging on the Regenerative Potential of Human Adipose Derived Mesenchymal Stem Cells
.
Stem Cells Int
.
2016
;
2016
:
2152435
. .
89.
Bonyadi
M
,
Waldman
SD
,
Liu
D
,
Aubin
JE
,
Grynpas
MD
,
Stanford
WL
.
Mesenchymal progenitor self-renewal deficiency leads to age-dependent osteoporosis in Sca-1/Ly-6A null mice
.
Proc Natl Acad Sci USA
.
2003
;
100
(
10
):
5840
5
. .
90.
Moerman
EJ
,
Teng
K
,
Lipschitz
DA
,
Lecka-Czernik
B
.
Aging activates adipogenic and suppresses osteogenic programs in mesenchymal marrow stroma/stem cells: the role of PPAR-gamma2 transcription factor and TGF-beta/BMP signaling pathways
.
Aging Cell
.
2004
;
3
(
6
):
379
89
. .
91.
Miura
Y
,
Miura
M
,
Gronthos
S
,
Allen
MR
,
Cao
C
,
Uveges
TE
,
Defective osteogenesis of the stromal stem cells predisposes CD18-null mice to osteoporosis
.
Proc Natl Acad Sci USA
.
2005
;
102
(
39
):
14022
7
. .
92.
Infante
A
,
Rodríguez
CI
.
Osteogenesis and aging: lessons from mesenchymal stem cells
.
Stem Cell Res Ther
.
2018
;
9
(
1
):
244
. .
93.
Shao
H
,
Xu
Q
,
Wu
Q
,
Ma
Q
,
Salgueiro
L
,
Wang
J
,
Defective CXCR4 expression in aged bone marrow cells impairs vascular regeneration
.
J Cell Mol Med
.
2011
;
15
(
10
):
2046
56
. .
94.
Neri
S
,
Borzì
RM
.
Molecular mechanisms contributing to mesenchymal stromal cell aging
.
Biomolecules
.
2020
;
10
(
2
). .
95.
Stolzing
A
,
Jones
E
,
McGonagle
D
,
Scutt
A
.
Age-related changes in human bone marrow-derived mesenchymal stem cells: consequences for cell therapies
.
Mech Ageing Dev
.
2008
;
129
(
3
):
163
73
. .
96.
Huang
XP
,
Sun
Z
,
Miyagi
Y
,
McDonald Kinkaid
H
,
Zhang
L
,
Weisel
RD
,
Differentiation of allogeneic mesenchymal stem cells induces immunogenicity and limits their long-term benefits for myocardial repair
.
Circulation
.
2010
;
122
(
23
):
2419
29
. .
97.
Katoh
M
.
Canonical and non-canonical WNT signaling in cancer stem cells and their niches: Cellular heterogeneity, omics reprogramming, targeted therapy and tumor plasticity (Review)
.
Int J Oncol
.
2017
;
51
(
5
):
1357
69
. .
98.
Zhou
S
,
Greenberger
JS
,
Epperly
MW
,
Goff
JP
,
Adler
C
,
Leboff
MS
,
Age-related intrinsic changes in human bone-marrow-derived mesenchymal stem cells and their differentiation to osteoblasts
.
Aging Cell
.
2008
;
7
(
3
):
335
43
. .
99.
van Deursen
J
.
The role of senescent cells in ageing
.
Nature
.
2014
;
509
(
7501
):
439
46
. .
100.
Campisi
J
.
Aging, cellular senescence, and cancer
.
Annu Rev Physiol
.
2013
;
75
:
685
705
. .
101.
Xu
M
,
Pirtskhalava
T
,
Farr
JN
,
Weigand
BM
,
Palmer
AK
,
Weivoda
MM
,
Senolytics improve physical function and increase lifespan in old age
.
Nat Med
.
2018
;
24
(
8
):
1246
56
. .
102.
Block
TJ
,
Marinkovic
M
,
Tran
ON
,
Gonzalez
AO
,
Marshall
A
,
Dean
DD
,
Restoring the quantity and quality of elderly human mesenchymal stem cells for autologous cell-based therapies
.
Stem Cell Res Ther
.
2017
;
8
(
1
):
239
. .
103.
Ito
T
,
Sawada
R
,
Fujiwara
Y
,
Seyama
Y
,
Tsuchiya
T
.
FGF-2 suppresses cellular senescence of human mesenchymal stem cells by down-regulation of TGF-beta2
.
Biochem Biophys Res Commun
.
2007
;
359
(
1
):
108
14
. .
104.
Xia
W
,
Hou
M
.
Macrophage migration inhibitory factor rescues mesenchymal stem cells from doxorubicin-induced senescence though the PI3K-Akt signaling pathway
.
Int J Mol Med
.
2018
;
41
(
2
):
1127
37
. .
105.
Reiter
RJ
,
Acuña-Castroviejo
D
,
Tan
DX
,
Burkhardt
S
.
Free radical-mediated molecular damage. Mechanisms for the protective actions of melatonin in the central nervous system
.
Ann NY Acad Sci
.
2001
;
939
:
200
15
.
106.
Liu
B
,
Ghosh
S
,
Yang
X
,
Zheng
H
,
Liu
X
,
Wang
Z
,
Resveratrol rescues SIRT1-dependent adult stem cell decline and alleviates progeroid features in laminopathy-based progeria
.
Cell Metab
.
2012
;
16
(
6
):
738
50
. .
107.
Pricola
KL
,
Kuhn
NZ
,
Haleem-Smith
H
,
Song
Y
,
Tuan
RS
.
Interleukin-6 maintains bone marrow-derived mesenchymal stem cell stemness by an ERK1/2-dependent mechanism
.
J Cell Biochem
.
2009
;
108
(
3
):
577
88
. .
108.
Xie
Z
,
Tang
S
,
Ye
G
,
Wang
P
,
Li
J
,
Liu
W
,
Interleukin-6/interleukin-6 receptor complex promotes osteogenic differentiation of bone marrow-derived mesenchymal stem cells
.
Stem Cell Res Ther
.
2018
;
9
(
1
):
13
. .
109.
Khan
M
,
Nickoloff
E
,
Abramova
T
,
Johnson
J
,
Verma
SK
,
Krishnamurthy
P
,
Embryonic stem cell-derived exosomes promote endogenous repair mechanisms and enhance cardiac function following myocardial infarction
.
Circ Res
.
2015
;
117
(
1
):
52
64
. .
110.
Müller-Ehmsen
J
,
Whittaker
P
,
Kloner
RA
,
Dow
JS
,
Sakoda
T
,
Long
TI
,
Survival and development of neonatal rat cardiomyocytes transplanted into adult myocardium
.
J Mol Cell Cardiol
.
2002
;
34
(
2
):
107
16
. .
111.
Pagani
FD
,
DerSimonian
H
,
Zawadzka
A
,
Wetzel
K
,
Edge
AS
,
Jacoby
DB
,
Autologous skeletal myoblasts transplanted to ischemia-damaged myocardium in humans. Histological analysis of cell survival and differentiation
.
J Am Coll Cardiol
.
2003
;
41
(
5
):
879
88
. .
112.
Kim
N
,
Cho
SG
.
New strategies for overcoming limitations of mesenchymal stem cell-based immune modulation
.
Int J Stem Cells
.
2015
;
8
(
1
):
54
68
. .
113.
Chen
B
,
Huang
S
,
Su
Y
,
Wu
YJ
,
Hanna
A
,
Brickshawana
A
,
Macrophage Smad3 protects the infarcted heart, stimulating phagocytosis and regulating inflammation
.
Circ Res
.
2019
;
125
(
1
):
55
70
. .
114.
Chen
P
,
Tao
J
,
Zhu
S
,
Cai
Y
,
Mao
Q
,
Yu
D
,
Radially oriented collagen scaffold with SDF-1 promotes osteochondral repair by facilitating cell homing
.
Biomaterials
.
2015
;
39
:
114
23
. .
115.
Lakshmanan
R
,
Kumaraswamy
P
,
Krishnan
UM
,
Sethuraman
S
.
Engineering a growth factor embedded nanofiber matrix niche to promote vascularization for functional cardiac regeneration
.
Biomaterials
.
2016
;
97
:
176
95
. .
116.
Giacca
M
,
Zacchigna
S
.
VEGF gene therapy: therapeutic angiogenesis in the clinic and beyond
.
Gene Ther
.
2012
;
19
(
6
):
622
9
. .
117.
Zou
XY
,
Yu
Y
,
Lin
S
,
Zhong
L
,
Sun
J
,
Zhang
G
,
Comprehensive miRNA analysis of human umbilical cord-derived mesenchymal stromal cells and extracellular vesicles
.
Kidney Blood Press Res
.
2018
;
43
(
1
):
152
61
. .
118.
Zhao
Y
,
Zhu
X
,
Zhang
L
,
Ferguson
CM
,
Song
T
,
Jiang
K
,
Mesenchymal stem/stromal cells and their extracellular vesicle progeny decrease injury in poststenotic swine kidney through different mechanisms
.
Stem Cells Dev
.
2020
;
29
(
18
):
1190
200
. .
119.
Ranghino
A
,
Bruno
S
,
Bussolati
B
,
Moggio
A
,
Dimuccio
V
,
Tapparo
M
,
The effects of glomerular and tubular renal progenitors and derived extracellular vesicles on recovery from acute kidney injury
.
Stem Cell Res Ther
.
2017
;
8
(
1
):
24
. .
120.
Morrison
TJ
,
Jackson
MV
,
Cunningham
EK
,
Kissenpfennig
A
,
McAuley
DF
,
O’Kane
CM
,
Mesenchymal stromal cells modulate macrophages in clinically relevant lung injury models by extracellular vesicle mitochondrial transfer
.
Am J Respir Crit Care Med
.
2017
;
196
(
10
):
1275
86
. .
121.
Stone
ML
,
Zhao
Y
,
Robert Smith
J
,
Weiss
ML
,
Kron
IL
,
Laubach
VE
,
Mesenchymal stromal cell-derived extracellular vesicles attenuate lung ischemia-reperfusion injury and enhance reconditioning of donor lungs after circulatory death
.
Respir Res
.
2017
;
18
(
1
):
212
. .
122.
Potter
DR
,
Miyazawa
BY
,
Gibb
SL
,
Deng
X
,
Togaratti
PP
,
Croze
RH
,
Mesenchymal stem cell-derived extracellular vesicles attenuate pulmonary vascular permeability and lung injury induced by hemorrhagic shock and trauma
.
J Trauma Acute Care Surg
.
2018
;
84
(
2
):
245
56
. .
123.
Lonati
C
,
Bassani
GA
,
Brambilla
D
,
Leonardi
P
,
Carlin
A
,
Maggioni
M
,
Mesenchymal stem cell-derived extracellular vesicles improve the molecular phenotype of isolated rat lungs during ischemia/reperfusion injury
.
J Heart Lung Transplant
.
2019
;
38
(
12
):
1306
16
. .
124.
Mardpour
S
,
Hassani
SN
,
Mardpour
S
,
Sayahpour
F
,
Vosough
M
,
Ai
J
,
Extracellular vesicles derived from human embryonic stem cell-MSCs ameliorate cirrhosis in thioacetamide-induced chronic liver injury
.
J Cell Physiol
.
2018
;
233
(
12
):
9330
44
. .
125.
Dabrowska
S
,
Andrzejewska
A
,
Strzemecki
D
,
Muraca
M
,
Janowski
M
,
Lukomska
B
.
Human bone marrow mesenchymal stem cell-derived extracellular vesicles attenuate neuroinflammation evoked by focal brain injury in rats
.
J Neuroinflammation
.
2019
;
16
(
1
):
216
. .
126.
Fang
SB
,
Zhang
HY
,
Meng
XC
,
Wang
C
,
He
BX
,
Peng
YQ
,
Small extracellular vesicles derived from human MSCs prevent allergic airway inflammation via immunomodulation on pulmonary macrophages
.
Cell Death Dis
.
2020
;
11
(
6
):
409
. .
127.
Dal Collo
G
,
Adamo
A
,
Gatti
A
,
Tamellini
E
,
Bazzoni
R
,
Takam Kamga
P
,
Functional dosing of mesenchymal stromal cell-derived extracellular vesicles for the prevention of acute graft-versus-host-disease
.
Stem Cells
.
2020
;
38
(
5
):
698
711
. .
128.
Sharma
J
,
Hampton
JM
,
Valiente
GR
,
Wada
T
,
Steigelman
H
,
Young
MC
,
Therapeutic development of mesenchymal stem cells or their extracellular vesicles to inhibit autoimmune-mediated inflammatory processes in systemic lupus erythematosus
.
Front Immunol
.
2017
;
8
:
526
. .
129.
Rajendran
RL
,
Gangadaran
P
,
Bak
SS
,
Oh
JM
,
Kalimuthu
S
,
Lee
HW
,
Extracellular vesicles derived from MSCs activates dermal papilla cell in vitro and promotes hair follicle conversion from telogen to anagen in mice
.
Sci Rep
.
2017
;
7
(
1
):
15560
. .
130.
Otsuru
S
,
Desbourdes
L
,
Guess
AJ
,
Hofmann
TJ
,
Relation
T
,
Kaito
T
,
Extracellular vesicles released from mesenchymal stromal cells stimulate bone growth in osteogenesis imperfecta
.
Cytotherapy
.
2018
;
20
(
1
):
62
73
. .
131.
Wang
X
,
Shah
FA
,
Vazirisani
F
,
Johansson
A
,
Palmquist
A
,
Omar
O
,
Exosomes influence the behavior of human mesenchymal stem cells on titanium surfaces
.
Biomaterials
.
2020
;
230
:
119571
. .
132.
Forteza-Genestra
MA
,
Antich-Rosselló
M
,
Calvo
J
,
Gayà
A
,
Monjo
M
,
Ramis
JM
.
Purity determines the effect of extracellular vesicles derived from mesenchymal stromal cells
.
Cells
.
2020
;
9
(
2
). .
133.
Lobb
RJ
,
Becker
M
,
Wen
SW
,
Wong
CS
,
Wiegmans
AP
,
Leimgruber
A
,
Optimized exosome isolation protocol for cell culture supernatant and human plasma
.
J Extracell Vesicles
.
2015
;
4
:
27031
. .
134.
Lamparski
HG
,
Metha-Damani
A
,
Yao
JY
,
Patel
S
,
Hsu
DH
,
Ruegg
C
,
Production and characterization of clinical grade exosomes derived from dendritic cells
.
J Immunol Methods
.
2002
;
270
(
2
):
211
26
. .
135.
Liu
S
,
Mahairaki
V
,
Bai
H
,
Ding
Z
,
Li
J
,
Witwer
KW
,
Highly purified human extracellular vesicles produced by stem cells alleviate aging cellular phenotypes of senescent human cells
.
Stem Cells
.
2019
;
37
(
6
):
779
90
. .
136.
Szatanek
R
,
Baran
J
,
Siedlar
M
,
Baj-Krzyworzeka
M
.
Isolation of extracellular vesicles: determining the correct approach (Review)
.
Int J Mol Med
.
2015
;
36
(
1
):
11
7
. .
137.
Nakai
W
,
Yoshida
T
,
Diez
D
,
Miyatake
Y
,
Nishibu
T
,
Imawaka
N
,
A novel affinity-based method for the isolation of highly purified extracellular vesicles
.
Sci Rep
.
2016
;
6
:
33935
. .
138.
Gardiner
C
,
Di Vizio
D
,
Sahoo
S
,
Théry
C
,
Witwer
KW
,
Wauben
M
,
Techniques used for the isolation and characterization of extracellular vesicles: results of a worldwide survey
.
J Extracell Vesicles
.
2016
;
5
:
32945
. .
139.
Gholizadeh
S
,
Shehata Draz
M
,
Zarghooni
M
,
Sanati-Nezhad
A
,
Ghavami
S
,
Shafiee
H
,
Microfluidic approaches for isolation, detection, and characterization of extracellular vesicles: current status and future directions
.
Biosens Bioelectron
.
2017
;
91
:
588
605
. .
140.
Zhan
SS
,
Jiang
JX
,
Wu
J
,
Halsted
C
,
Friedman
SL
,
Zern
MA
,
Phagocytosis of apoptotic bodies by hepatic stellate cells induces NADPH oxidase and is associated with liver fibrosis in vivo
.
Hepatology
.
2006
;
43
(
3
):
435
43
. .
141.
Li
F
,
Huang
Q
,
Chen
J
,
Peng
Y
,
Roop
DR
,
Bedford
JS
,
Apoptotic cells activate the “phoenix rising” pathway to promote wound healing and tissue regeneration
.
Sci Signal
.
2010
;
3
(
110
):
ra13
. .
142.
Liu
D
,
Kou
X
,
Chen
C
,
Liu
S
,
Liu
Y
,
Yu
W
,
Circulating apoptotic bodies maintain mesenchymal stem cell homeostasis and ameliorate osteopenia via transferring multiple cellular factors
.
Cell Res
.
2018
;
28
(
9
):
918
33
. .
143.
Brock
CK
,
Wallin
ST
,
Ruiz
OE
,
Samms
KM
,
Mandal
A
,
Sumner
EA
,
Stem cell proliferation is induced by apoptotic bodies from dying cells during epithelial tissue maintenance
.
Nat Commun
.
2019
;
10
(
1
):
1044
. .
144.
Ocampo
A
,
Reddy
P
,
Martinez-Redondo
P
,
Platero-Luengo
A
,
Hatanaka
F
,
Hishida
T
,
In vivo amelioration of age-associated hallmarks by partial reprogramming
.
Cell
.
2016
;
167
(
7
):
1719
e12
.
145.
Sharpless
NE
,
Bardeesy
N
,
Lee
KH
,
Carrasco
D
,
Castrillon
DH
,
Aguirre
AJ
,
Loss of p16Ink4a with retention of p19Arf predisposes mice to tumorigenesis
.
Nature
.
2001
;
413
(
6851
):
86
91
. .
146.
Ledford
H
.
Reversal of biological clock restores vision in old mice
.
Nature
.
2020
;
588
(
7837
):
209
. .
147.
Baker
DJ
,
Wijshake
T
,
Tchkonia
T
,
LeBrasseur
NK
,
Childs
BG
,
van de Sluis
B
,
Clearance of p16Ink4a-positive senescent cells delays ageing-associated disorders
.
Nature
.
2011
;
479
(
7372
):
232
6
. .
148.
Baker
DJ
,
Childs
BG
,
Durik
M
,
Wijers
ME
,
Sieben
CJ
,
Zhong
J
,
Naturally occurring p16(Ink4a)-positive cells shorten healthy lifespan
.
Nature
.
2016
;
530
(
7589
):
184
9
. .
149.
Ashkenazi
A
,
Fairbrother
WJ
,
Leverson
JD
,
Souers
AJ
.
From basic apoptosis discoveries to advanced selective BCL-2 family inhibitors
.
Nat Rev Drug Discov
.
2017
;
16
(
4
):
273
84
. .
150.
Shamas-Din
A
,
Brahmbhatt
H
,
Leber
B
,
Andrews
DW
.
BH3-only proteins: Orchestrators of apoptosis
.
Biochim Biophys Acta
.
2011
;
1813
(
4
):
508
20
. .
151.
Chipuk
JE
,
Moldoveanu
T
,
Llambi
F
,
Parsons
MJ
,
Green
DR
.
The BCL-2 family reunion
.
Mol Cell
.
2010
;
37
(
3
):
299
310
. .
152.
Czabotar
PE
,
Lessene
G
,
Strasser
A
,
Adams
JM
.
Control of apoptosis by the BCL-2 protein family: implications for physiology and therapy
.
Nat Rev Mol Cell Biol
.
2014
;
15
(
1
):
49
63
. .
153.
Luna-Vargas
MP
,
Chipuk
JE
.
Physiological and pharmacological control of BAK, BAX, and Beyond
.
Trends Cell Biol
.
2016
;
26
(
12
):
906
17
. .
154.
O'Neill
KL
,
Huang
K
,
Zhang
J
,
Chen
Y
,
Luo
X
.
Inactivation of prosurvival Bcl-2 proteins activates Bax/Bak through the outer mitochondrial membrane
.
Genes Dev
.
2016
;
30
(
8
):
973
88
. .
155.
Oltersdorf
T
,
Elmore
SW
,
Shoemaker
AR
,
Armstrong
RC
,
Augeri
DJ
,
Belli
BA
,
An inhibitor of Bcl-2 family proteins induces regression of solid tumours
.
Nature
.
2005
;
435
(
7042
):
677
81
. .
156.
Yosef
R
,
Pilpel
N
,
Tokarsky-Amiel
R
,
Biran
A
,
Ovadya
Y
,
Cohen
S
,
Directed elimination of senescent cells by inhibition of BCL-W and BCL-XL
.
Nat Commun
.
2016
;
7
:
11190
. .
157.
Birch
J
,
Gil
J
.
Blunting senescence boosts liver regeneration
.
Genes Dev
.
2020
;
34
:
463
4
. .
158.
Chang
J
,
Wang
Y
,
Shao
L
,
Laberge
RM
,
Demaria
M
,
Campisi
J
,
Clearance of senescent cells by ABT263 rejuvenates aged hematopoietic stem cells in mice
.
Nat Med
.
2016
;
22
(
1
):
78
83
. .
159.
Zhu
Y
,
Tchkonia
T
,
Fuhrmann-Stroissnigg
H
,
Dai
HM
,
Ling
YY
,
Stout
MB
,
Identification of a novel senolytic agent, navitoclax, targeting the Bcl-2 family of anti-apoptotic factors
.
Aging Cell
.
2016
;
15
(
3
):
428
35
. .
160.
Childs
BG
,
Baker
DJ
,
Wijshake
T
,
Conover
CA
,
Campisi
J
,
van Deursen
JM
.
Senescent intimal foam cells are deleterious at all stages of atherosclerosis
.
Science
.
2016
;
354
(
6311
):
472
7
. .
161.
Tse
C
,
Shoemaker
AR
,
Adickes
J
,
Anderson
MG
,
Chen
J
,
Jin
S
,
ABT-263: a potent and orally bioavailable Bcl-2 family inhibitor
.
Cancer Res
.
2008
;
68
(
9
):
3421
8
. .
162.
Leverson
JD
,
Phillips
DC
,
Mitten
MJ
,
Boghaert
ER
,
Diaz
D
,
Tahir
SK
,
Exploiting selective BCL-2 family inhibitors to dissect cell survival dependencies and define improved strategies for cancer therapy
.
Sci Transl Med
.
2015
;
7
(
279
):
279ra40
. .
163.
Gandhi
L
,
Camidge
DR
,
Ribeiro de Oliveira
M
,
Bonomi
P
,
Gandara
D
,
Khaira
D
,
Phase I study of Navitoclax (ABT-263), a novel Bcl-2 family inhibitor, in patients with small-cell lung cancer and other solid tumors
.
J Clin Oncol
.
2011
;
29
(
7
):
909
16
. .
164.
Souers
AJ
,
Leverson
JD
,
Boghaert
ER
,
Ackler
SL
,
Catron
ND
,
Chen
J
,
ABT-199, a potent and selective BCL-2 inhibitor, achieves antitumor activity while sparing platelets
.
Nat Med
.
2013
;
19
(
2
):
202
8
. .
165.
He
Y
,
Zhang
X
,
Chang
J
,
Kim
HN
,
Zhang
P
,
Wang
Y
,
Using proteolysis-targeting chimera technology to reduce navitoclax platelet toxicity and improve its senolytic activity
.
Nat Commun
.
2020
;
11
(
1
):
1996
. .
166.
Zhang
H
,
Nimmer
PM
,
Tahir
SK
,
Chen
J
,
Fryer
RM
,
Hahn
KR
,
Bcl-2 family proteins are essential for platelet survival
.
Cell Death Differ
.
2007
;
14
(
5
):
943
51
. .
167.
Debrincat
MA
,
Pleines
I
,
Lebois
M
,
Lane
RM
,
Holmes
ML
,
Corbin
J
,
BCL-2 is dispensable for thrombopoiesis and platelet survival
.
Cell Death Dis
.
2015
;
6
:
e1721
. .
168.
Breccia
M
,
Alimena
G
.
Activity and safety of dasatinib as second-line treatment or in newly diagnosed chronic phase chronic myeloid leukemia patients
.
BioDrugs
.
2011
;
25
(
3
):
147
57
. .
169.
Chan
CM
,
Jing
X
,
Pike
LA
,
Zhou
Q
,
Lim
DJ
,
Sams
SB
,
Targeted inhibition of Src kinase with dasatinib blocks thyroid cancer growth and metastasis
.
Clin Cancer Res
.
2012
;
18
(
13
):
3580
91
. .
170.
Zhu
Y
,
Tchkonia
T
,
Pirtskhalava
T
,
Gower
AC
,
Ding
H
,
Giorgadze
N
,
The Achilles’ heel of senescent cells: from transcriptome to senolytic drugs
.
Aging Cell
.
2015
;
14
(
4
):
644
58
. .
171.
Hickson
LJ
,
Langhi Prata
LGP
,
Bobart
SA
,
Evans
TK
,
Giorgadze
N
,
Hashmi
SK
,
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
56
. .
172.
Justice
JN
,
Gregory
H
,
Tchkonia
T
,
LeBrasseur
NK
,
Kirkland
JL
,
Kritchevsky
SB
,
Cellular senescence biomarker p16INK4a+ cell burden in thigh adipose is associated with poor physical function in older women
.
J Gerontol A Biol Sci Med Sci
.
2018
;
73
(
7
):
939
45
. .
173.
Currais
A
,
Farrokhi
C
,
Dargusch
R
,
Armando
A
,
Quehenberger
O
,
Schubert
D
,
Fisetin reduces the impact of aging on behavior and physiology in the rapidly aging SAMP8 mouse
.
J Gerontol A Biol Sci Med Sci
.
2018
;
73
(
3
):
299
307
. .
174.
Yousefzadeh
MJ
,
Zhu
Y
,
McGowan
SJ
,
Angelini
L
,
Fuhrmann-Stroissnigg
H
,
Xu
M
,
Fisetin is a senotherapeutic that extends health and lifespan
.
EBioMedicine
.
2018
;
36
:
18
28
. .
175.
Maher
P
.
How fisetin reduces the impact of age and disease on CNS function
.
Front Biosci (Schol Ed)
.
2015
;
7
:
58
82
. .
176.
Jeon
OH
,
Kim
C
,
Laberge
RM
,
Demaria
M
,
Rathod
S
,
Vasserot
AP
,
Local clearance of senescent cells attenuates the development of post-traumatic osteoarthritis and creates a pro-regenerative environment
.
Nat Med
.
2017
;
23
(
6
):
775
81
. .
177.
Zhu
Y
,
Doornebal
EJ
,
Pirtskhalava
T
,
Giorgadze
N
,
Wentworth
M
,
Fuhrmann-Stroissnigg
H
,
New agents that target senescent cells: the flavone, fisetin, and the BCL-X inhibitors, A1331852 and A1155463
.
Aging
.
2017
;
9
(
3
):
955
63
.
178.
Prata
LGPL
,
Ovsyannikova
IG
,
Tchkonia
T
,
Kirkland
JL
.
Senescent cell clearance by the immune system: Emerging therapeutic opportunities
.
Semin Immunol
.
2018
;
40
:
101275
. .
179.
Weiland
T
,
Lampe
J
,
Essmann
F
,
Venturelli
S
,
Berger
A
,
Bossow
S
,
Enhanced killing of therapy-induced senescent tumor cells by oncolytic measles vaccine viruses
.
Int J Cancer
.
2014
;
134
(
1
):
235
43
. .
180.
Burton
DGA
,
Stolzing
A
.
Cellular senescence: Immunosurveillance and future immunotherapy
.
Ageing Res Rev
.
2018
;
43
:
17
25
. .
181.
Krizhanovsky
V
,
Yon
M
,
Dickins
RA
,
Hearn
S
,
Simon
J
,
Miething
C
,
Senescence of activated stellate cells limits liver fibrosis
.
Cell
.
2008
;
134
(
4
):
657
67
. .
182.
Xu
M
,
Tchkonia
T
,
Ding
H
,
Ogrodnik
M
,
Lubbers
ER
,
Pirtskhalava
T
,
JAK inhibition alleviates the cellular senescence-associated secretory phenotype and frailty in old age
.
Proc Natl Acad Sci USA
.
2015
;
112
(
46
):
E6301
10
. .
183.
Yang
LW
,
Song
M
,
Li
YL
,
Liu
YP
,
Liu
C
,
Han
L
,
L-Carnitine inhibits the senescence-associated secretory phenotype of aging adipose tissue by JNK/p53 pathway
.
Biogerontology
.
2019
;
20
(
2
):
203
11
. .
184.
Griveau
A
,
Wiel
C
,
Ziegler
DV
,
Bergo
MO
,
Bernard
D
.
The JAK1/2 inhibitor ruxolitinib delays premature aging phenotypes
.
Aging Cell
.
2020
;
19
(
4
):
e13122
. .
185.
Xu
M
,
Palmer
AK
,
Ding
H
,
Weivoda
MM
,
Pirtskhalava
T
,
White
TA
,
Targeting senescent cells enhances adipogenesis and metabolic function in old age
.
eLife
.
2015
;
4
:
e12997
. .
186.
Wu
W
,
Fu
J
,
Gu
Y
,
Wei
Y
,
Ma
P
,
Wu
J
.
JAK2/STAT3 regulates estrogen-related senescence of bone marrow stem cells
.
J Endocrinol
.
2020
;
245
(
1
):
141
53
. .
187.
Price
FD
,
von Maltzahn
J
,
Bentzinger
CF
,
Dumont
NA
,
Yin
H
,
Chang
NC
,
Inhibition of JAK-STAT signaling stimulates adult satellite cell function
.
Nat Med
.
2014
;
20
(
10
):
1174
81
. .
188.
Toso
A
,
Revandkar
A
,
Di Mitri
D
,
Guccini
I
,
Proietti
M
,
Sarti
M
,
Enhancing chemotherapy efficacy in Pten-deficient prostate tumors by activating the senescence-associated antitumor immunity
.
Cell Rep
.
2014
;
9
(
1
):
75
89
. .
189.
Eggert
T
,
Wolter
K
,
Ji
J
,
Ma
C
,
Yevsa
T
,
Klotz
S
,
Distinct functions of senescence-associated immune responses in liver tumor surveillance and tumor progression
.
Cancer cell
.
2016
;
30
(
4
):
533
47
. .
190.
Xue
W
,
Zender
L
,
Miething
C
,
Dickins
RA
,
Hernando
E
,
Krizhanovsky
V
,
Senescence and tumour clearance is triggered by p53 restoration in murine liver carcinomas
.
Nature
.
2007
;
445
(
7128
):
656
60
. .
191.
Ventura
A
,
Kirsch
DG
,
McLaughlin
ME
,
Tuveson
DA
,
Grimm
J
,
Lintault
L
,
Restoration of p53 function leads to tumour regression in vivo
.
Nature
.
2007
;
445
(
7128
):
661
5
. .
192.
Rajagopalan
S
,
Long
EO
.
Cellular senescence induced by CD158d reprograms natural killer cells to promote vascular remodeling
.
Proc Natl Acad Sci USA
.
2012
;
109
(
50
):
20596
601
. .
193.
Storer
M
,
Mas
A
,
Robert-Moreno
A
,
Pecoraro
M
,
Ortells
MC
,
Di Giacomo
V
,
Senescence is a developmental mechanism that contributes to embryonic growth and patterning
.
Cell
.
2013
;
155
(
5
):
1119
30
. .
194.
Jun
JI
,
Lau
LF
.
The matricellular protein CCN1 induces fibroblast senescence and restricts fibrosis in cutaneous wound healing
.
Nat Cell Biol
.
2010
;
12
(
7
):
676
85
. .
195.
Demaria
M
,
Ohtani
N
,
Youssef
SA
,
Rodier
F
,
Toussaint
W
,
Mitchell
JR
,
An essential role for senescent cells in optimal wound healing through secretion of PDGF-AA
.
Dev Cell
.
2014
;
31
(
6
):
722
33
. .
196.
Krishnamurthy
J
,
Ramsey
MR
,
Ligon
KL
,
Torrice
C
,
Koh
A
,
Bonner-Weir
S
,
p16INK4a induces an age-dependent decline in islet regenerative potential
.
Nature
.
2006
;
443
(
7110
):
453
7
. .
197.
Koren
G
,
Nordon
G
,
Radinsky
K
,
Shalev
V
.
Clinical pharmacology of old age
.
Expert Rev Clin Pharmacol
.
2019
;
12
(
8
):
749
55
. .
198.
Wimmer
BC
,
Cross
AJ
,
Jokanovic
N
,
Wiese
MD
,
George
J
,
Johnell
K
,
Clinical outcomes associated with medication regimen complexity in older people: a systematic review
.
J Am Geriatr Soc
.
2017
;
65
(
4
):
747
53
. .
Open Access License / Drug Dosage / Disclaimer
This article is licensed under the Creative Commons Attribution-NonCommercial 4.0 International License (CC BY-NC). Usage and distribution for commercial purposes requires written permission. Drug Dosage: The authors and the publisher have exerted every effort to ensure that drug selection and dosage set forth in this text are in accord with current recommendations and practice at the time of publication. However, in view of ongoing research, changes in government regulations, and the constant flow of information relating to drug therapy and drug reactions, the reader is urged to check the package insert for each drug for any changes in indications and dosage and for added warnings and precautions. This is particularly important when the recommended agent is a new and/or infrequently employed drug. Disclaimer: The statements, opinions and data contained in this publication are solely those of the individual authors and contributors and not of the publishers and the editor(s). The appearance of advertisements or/and product references in the publication is not a warranty, endorsement, or approval of the products or services advertised or of their effectiveness, quality or safety. The publisher and the editor(s) disclaim responsibility for any injury to persons or property resulting from any ideas, methods, instructions or products referred to in the content or advertisements.