Next Article in Journal
Machine Learning Model Discriminate Ischemic Heart Disease Using Breathome Analysis
Previous Article in Journal
Obesity-Related Inflammation Reduces Treatment Sensitivity and Promotes Aggressiveness in Luminal Breast Cancer Modulating Oxidative Stress and Mitochondria
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Suppressing the Aging Phenotype of Mesenchymal Stromal Cells: Are We Ready for Clinical Translation?

Department of Surgical Sciences, CIR-Dental School, University of Turin, 10126 Turin, Italy
*
Author to whom correspondence should be addressed.
Biomedicines 2024, 12(12), 2811; https://doi.org/10.3390/biomedicines12122811
Submission received: 2 October 2024 / Revised: 6 December 2024 / Accepted: 9 December 2024 / Published: 11 December 2024

Abstract

:
Mesenchymal stem/stromal cells (MSCs) are involved in the maintenance and regeneration of a large variety of tissues due to their stemness and multi-lineage differentiation capability. Harnessing these advantageous features, a flurry of clinical trials have focused on MSCs to treat different pathologies, but only few protocols have received regulatory approval so far. Among the various causes hindering MSCs’ efficacy is the emergence of cellular senescence, which has been correlated with specific characteristics, such as morphological and epigenetic alterations, DNA damage, ROS production, mitochondrial dysfunction, telomere shortening, non-coding RNAs, loss of proteostasis, and a peculiar senescence-associated secretory phenotype. Several strategies have been investigated for delaying or even hopefully reverting the onset of senescence, as assessed by the senescent phenotype of MSCs. Here, the authors reviewed the most updated literature on the potential causes of senescence, with a particular emphasis on the current and future therapeutic approaches aimed at reverting senescence and/or extending the functional lifespan of stem cells.

Graphical Abstract

1. Background

Mesenchymal stem cells, also known as mesenchymal stromal cells (MSCs), are multipotent non-hematopoietic stem cells that represent a reservoir indispensable for the homeostatic maintenance and regeneration of a large variety of tissues throughout their individual lifespans, owing to their capacity of keeping their “stemness” while differentiating into the required histotype [1]. Since they are ubiquitous in the human body [2], MSCs have been defined through “minimal criteria” by Dominici et al. in 2006 as follows: (a) tissue culture plastic-adherent; (b) positive (≥95%) for surface antigen markers CD105, CD90, and CD73 while also being negative (≤2%) for CD45, CD34, CD14 or CD11b, CD79α or CD19, and HLA-DR; and (c) capable of differentiating to adipocytes, chondroblasts, and osteoblasts [3]. MSCs are also well recognized for their unique immunomodulatory functions, which allow them to exert regulatory activity on the immune microenvironment. Specifically, MSCs have been demonstrated to inhibit the proliferation of T cells and immune defense responses, underscoring their immunosuppressive properties.
In the human body, there are different sources of MSCs: bone marrow (BM), umbilical cord, and adipose tissue are particularly rich in MSCs, but the oral cavity also harbors a variety of MSCs [4]. In the past decade, about 77% of clinical trials dealing with MSCs regarded BMSCs and ASCs [5]. Notwithstanding the remarkable number of studies available, only a few MSC-based protocols have received regulatory approval [6]. Among the possible causes of this shortcoming are cells’ heterogeneity, considerably limiting the standardization of therapies and the onset of senescence [7]. Recent studies report that senescent MSCs show reduced regenerative potential, impaired migration, and altered paracrine secretion, promoting tissue damage [8]. Transplanted senescent cells exhibit less efficient therapeutic potential function and may be detrimental [9], posing a major issue for MSC treatments. Thus, preventing, or at least reducing, stem cell senescence is emerging as a paramount task to improve the efficacy of future clinical applications.

2. Senescence Features of MSCs

Cellular senescence regards properly mitotic cells and is usually defined as a “cell state triggered by stressful insults and certain physiological processes, characterized by a prolonged and generally irreversible cell-cycle arrest with secretory features, macromolecular damage, and altered metabolism” [10]. Senescent cells are resistant to apoptosis and show alterations in cellular and tissue repair mechanisms [11]. For instance, autophagy, a highly conserved evolutionary process controlling cellular homeostasis, has been associated with aging and a range of pathological conditions [12,13] since it is essential for the degradation and recycling of senescent or damaged organelles and macromolecules via lysosomal pathways [14]. As a physiological homeostatic mechanism, senescence is implicated in tissue repair [12], while its impairment leads to inflammatory diseases associated with aging such as osteoarthritis and Alzheimer’s disease [15,16]. Senescence is important to control cell growth as it protects against tumorigenesis by preventing cells with stress-induced damage from entering the replicative cycle [17].
In the following paragraphs, we will focus more carefully on different features of MSC senescence, which are shown in Figure 1.

2.1. Surface Markers and Morphological Changes

In cell culture, MSCs have a morphology characterized by a spindle-shaped cell body with a few long, thin processes, a large nucleus, and a differentiated nucleolus (Figure 2A). Senescent MSCs modify their morphology, becoming more enlarged and flatted with small nuclei, a granular cytoplasm, augmented actin stress fibers and losing their spindle-shaped characteristics. The senescence-associated morphological changes have been linked to the status of the scaffolding protein caveolin-1 (CAV-1), which is thought to regulate actin stress fiber formation [18]. It is widely recognized that MSCs tend to differentiate preferentially towards adipose tissue during senescence. Peroxisome proliferator-activated receptor-γ (PPAR-γ), an adipogenesis transcription factor, increases during senescence, thereby directing MSC differentiation towards adipogenesis. In contrast, Wnt/β-catenin (WNT) signaling can inhibit adipogenesis and promote MSCs’ differentiation into osteoblasts, positioning WNT signaling as a critical element in the adipogenesis and osteogenesis balance, as well as in regulating cellular senescence [19]. WNT signaling is downregulated according to age, but an increased activation of WNT signaling can also induce MSCs’ senescence.
In 1961, Hayflick and Moorhead described cellular senescence for the first time by reporting that human diploid fibroblasts derived from fetuses lost the ability to proliferate and degenerate after about 50 sub-cultivations and one year in culture [20]. Prolonged culture can lead to cellular senescence, which can significantly impair MSCs’ function in terms of their immunomodulatory function, proliferation rate, and expression of surface antigens, even prior to their expected replicative limit. Indeed, senescent MSCs downregulate the expression of alkaline phosphatase (ALP) and osteocalcin, correlating with the observed decrease efficiency in bone formation during long-term culture [19]. STRO-1, CD106, and CD146 were expressed by MSCs and downregulated in vitro and/or in vivo during senescence, allowing for the identification of senescent surface markers on MSCs. Positive staining for SA-β-gal activity is universally known as a feature of senescent cells (Figure 2B); indeed, β-gal activity in senescent cells is significantly enhanced due to the increased lysosomal activity and altered cytosolic pH [21], along with the persistent expression of p53, p21, and p16 proteins (Figure 2C) [22].
CD264+ MSCs display elevated SA-β-gal activity, decreased proliferation rates, and reduced differentiation potential, whereas CD295+ MSCs exhibit lower proliferative capacity, with CD295/LEPR specifically identifying a distinct subpopulation of cells undergoing apoptosis [23]. The concomitant decrease in the adhesion molecule CD146 and the higher expression of decoy TRAIL receptor CD264 have been associated with senescent MSCs [24]. Recently, autofluorescence has also been reported as a feature of senescence [25]. Finally, senescent MSCs display nuclear changes, such as the loss of Lamin B1, that even precede SASP and SA-β-gal activity and the onset of senescence-associated heterochromatic foci (SAHF). These punctate nuclear structures are probably condensed chromosomes that depend on p53 and Rb activation [26].

2.2. ROS Production and DNA Damage

MSCs accumulate DNA damage over time because of various stressors, such as genotoxic agents, nutrient deprivation, hypoxia, and mitochondrial dysfunction [10]. Both endogenous (oncogene activation, chronic inflammation, and oxidative stress driven by the accumulation of reactive oxygen species (ROS) and exogenous mutagens (such as irradiation) are known contributors to DNA damage [27]. ROS, such as peroxide, superoxide, and hydroxyl radical, are normally found in cells and play a fundamental role in cellular signaling to perform physiological functions. Nonetheless, high ROS concentration drives MSCs toward senescence through different pathological mechanisms. Cell cycle arrest in senescent MSCs is influenced by oxidative stress, such as the release of DNA-damaging ROS [28]. Natural defense mechanisms against ROS, which are physiological products of the aerobic metabolism, may not be sufficient to avoid senescence owing to oxidative stress. Generally, the regulation of ROS levels and oxidative repair mechanisms become less efficient with aging, leading to ROS accumulation, molecular damage, and ultimately MSC senescence [29]. Senescent MSCs, in turn, produce more ROS, creating a positive feedback loop in which the more ROS are present, the worse the senescence and the consequent molecular damage [30]. Antioxidants scavenge local ROS, delaying the onset of senescence; when aged MSCs are treated with reduced glutathione and melatonin, they reacquire early-passage stemness and migratory capabilities, lowering the expression of p16 and p53 [31].
Consistently, hypoxic culture promotes the inhibition of MSCs’ senescence with the retention of stem cell properties compared to normoxia, and more importantly, without increasing tumorigenicity. ROS can stimulate the MAPK pathway both directly and indirectly, and the inhibition of the p38 MAPK pathway has been shown to prevent senescence [32]. Indeed, the activation of this pathway promotes p53 phosphorylation, allowing ROS to induce senescence. Additionally, the phosphatidylinositol-3-kinase (PI3K)-protein kinase B (AKT) signaling pathway is also implicated in MSC senescence [33,34]. This activation subsequently induces the transcription of target genes, such as the mechanistic target of rapamycin 1 (mTORC1), forkhead box protein O3 (FOXO3), and p53, ultimately leading to MSC senescence [35]. Since hypoxic culture conditions decrease the senescent phenotype by mimicking the biological niche of MSCs, hypoxic pre-conditioning was proposed to rejuvenate MSCs before transplantation [36].
The DNA damage response network regulates cell cycle arrest, which is a hallmark of MSC senescence and is primarily governed by the p21CIP1/WAF1 and p16 INK4A signaling pathways, which are crucial regulators of genes involved in the control of cell growth and proliferation, ultimately leading to cell cycle arrest and the prevention of re-entry into the cell cycle by damaged and/or senescent cells [37]. p21 is a cyclin-dependent kinase (CDK) inhibitor that is induced by DNA damage and transcriptionally activated by p53. The expression levels of p16 and p21 are upregulated both in vitro and in vivo during cellular senescence [38]. The activation of p21 reduces the phosphorylation of retinoblastoma protein (RB), allowing RB to retain its function and continue to suppress the E2 transcription factor [39,40]. Cell cycle withdrawal has also been described in quiescence, which is reversible upon proper stimulation, and in terminal differentiation with apparent differences. In particular circumstances, such as in tumors, a subset of malignant senescent cells upregulate stemness genes and can re-enter the cell cycle, inducing a much more aggressive phenotype of tumor cells [41,42].
An abundance of ROS not only triggers the DNA damage response, but also induces a range of other detrimental effects, such as mitochondrial dysfunction, telomere attrition, and protein degradation, which contribute to cellular senescence.

2.3. Mitochondrial Dysfunction

Mitochondria exert a crucial role in cellular senescence as they are essential for cellular respiration, and their dysfunction is associated with abnormal NAD+/NADH and ATP/ADP ratios, which are associated with MSCs’ senescence. An imbalance in the ATP/ADP ratio may activate 5ʹ-AMP-activated protein kinase (AMPK), inducing senescence through the p53/p21 pathway. Mitophagy regulates the selective degradation of damaged or dysfunctional mitochondria. Insufficient mitophagy has been implicated in senescence-related cellular injuries through the accumulation of damaged mitochondria and the consequent metabolic dysfunction. Recent findings indicate that reduced PARKIN (an E3 ubiquitin ligase involved in mitophagy) translocation to damaged mitochondria, through the p53 pathway, leads to defective mitophagy, hindering the clearance of damaged mitochondria. Additionally, ROS accumulation and sirtuin (SIRT) deficiency have been shown to inhibit PARKIN-mediated mitophagy, further contributing to cellular senescence [43,44].
It is established that elevated ROS levels cause increased mitochondrial DNA (mtDNA) damage, and due to the lack of efficient repair mechanisms, mtDNA is more susceptible to mutations than nuclear DNA. Age-related mtDNA abnormalities contribute to an augment of ROS production, creating a vicious cycle that disrupts the balance between ROS and antioxidants [45,46]. Another theory suggests that ROS-induced DNA damage stimulates mitochondrial biogenesis, leading to a higher number of mitochondria and further increasing mitochondrial ROS production, thus perpetuating another vicious cycle [47].

2.4. Telomeres Shortening

“Mammalian telomeres are composed of tandem repeats of TTAGGGn DNA sequences associated with a six-member protein Shelterin complex that facilitates the formation of a lariat-like structure (the t-loop) to shield the exposed chromosome ends of telomeric DNA from DNA damage machinery” [48]. Progressive telomere shortening occurs in all dividing normal somatic cells, and even stem cells, although not at the same rate [49]. This phenomenon originates from the incomplete lagging-strand DNA synthesis during DNA replication, owing to the inability of standard DNA polymerase to replicate fully linear DNA in the absence of telomerase [50], a ribonucleoprotein enzyme (whose catalytic component is represented by the telomerase reverse transcriptase TERT, named hTERT in humans) that is responsible for telomere elongation.
When telomere shortening reaches a critical length, due to the decrease in telomere capping proteins, DNA damage response pathways arrest cell proliferation [51]. If telomere-binding proteins are sufficient to inhibit DNA repair and avoid fusions [52], a permanent DNA damage-induced proliferative arrest occurs, initiating cell senescence. Activation of the DNA damage response at telomeres leads to the formation of telomere-associated DDR foci (TAFs), which are markers of cellular senescence in cultured cells [50]. In their inspiring magistral review, Rosiello et al. [50] proposed a comprehensive perspective on how persistent DRR at the telomers may explain both replicative cellular senescence caused by critically short telomere and the senescence-like state occurring in non-replicating cells. Hence, the somehow simplistic concept of telomere shortening (classically describing replicative senescence) has been overcome by a more complex “telomere-centric” etiopathological mechanism unifying several aging hallmarks, which is also in agreement with the findings of Chakravarti et al. [53].

2.5. Epigenetic Alterations

Histone modification and DNA methylation are epigenetic events implicated in cellular senescence and may drive alterations associated with MSC senescence. Histone modification plays a significant role in influencing the transcriptional activities of nearby genomic regions. Histone deacetylases (HDACs), a group of enzymes that modulate histone acetylation levels, determine whether chromatin is in a relaxed euchromatin state associated with an open configuration and active transcription or a condensed heterochromatin state and an inactive conformation. HDAC inhibitors can accelerate aging by activating the transcription of p21 through the acetylation of histones H3 and H4 [54]. Moreover, HDAC inhibitors can regulate the cellular senescence gene CDKN2A via multiple microRNAs [54]. During senescence, EZH2, the histone lysine methyltransferase that allows the trimethylation of lysine 27 on histone H3 (H3K27me3), is downregulated, resulting in the loss of H3K27me3 at the CDKN2A locus with the consequent upregulation of p16 and the activation of genes of the senescence-associated secretory phenotype (SASP) [55]. Indeed, the role of EZH2 seems broader as its downregulation was proven to induce senescence by activating a DDR in proliferating cells before the reduction in the levels of H3K27me3 marks [55]. DNA methylation rules the gene expression inactivation of the X-chromosome, transposon silencing, cell specification, and cellular identity maintenance [56,57].
Changes in the DNA methylation profile also regulate the self-renewal and differentiation of adult stem cells [58]. During differentiation, DNA methylation increases, leading to the silencing of pluripotency-associated genes and other genes involved in non-specific cell type differentiation [59]. The methylation status is generally regulated by DNA methyltransferases (DNMTs), which include three major members: DNMT1, which maintains the methylation pattern during DNA replication, and DNMT3A and DNMT3B, which are responsible for de novo methylation. The expression levels of DNMT1 and DNMT3B are significantly reduced during senescence [60].

2.6. Senescence-Associated Secretory Phenotype (SASP)

Senescent MSCs can be distinguished from healthy cells based on their immunogenicity [61]. Senescent cells exhibit SASP, i.e., a multitude of secretory factors, including inflammatory and matrix-modeling signaling molecules, released with autocrine and paracrine effects. The SASP mediates the conversion of the anti-inflammatory MSC phenotype to the proinflammatory one [62]. Senescent MSCs increase the production of many cytokines and factors, including IL-1, IL-3, IL-4, IL-6, IL-8, IL-17, interferon-β (IFN- β), epidermal growth factor (EGF), fibroblast growth factor-2 (FGF-2), FGF-4, and FGF-8, hepatocyte growth factor (HGF), insulin growth factor-1 (IGF-1), platelet-derived growth factor (PDGF), transforming growth factor-β (TGF-β), and vascular endothelial growth factors (VEGFs) [24]. Among the factors contributing to the SASP of aged MSCs are the persistent activation of TLR signaling [63] and the increased adipogenesis associated with aging [64].
SASP and the immune system have a complex interaction. While SASP factors are known to recruit immune cells, such as macrophages, T lymphocytes, natural killer cells, and neutrophils, which promote the clearance of senescent cells [65], aged MSCs exhibit an altered immune-suppressive function. In physiological conditions, MSCs promote the polarization of macrophages towards an anti-inflammatory phenotype, M2 [66]; conversely, aged MSCs promote a shift in macrophage from the M2 to M1 (pro-inflammatory) phenotype, inducing inflammation [67]. MSCs exert strong immunosuppressive action on T cells, while aged MSCs show an impaired capability to reduce the proliferation of T cells [68,69].
SASP factors can act on neighboring cells through paracrine mechanisms to accelerate senescence. Indeed, non-coding RNA can be both contained in SASP and secreted in exosomes, contributing to the induction of cellular senescence in young cells [70,71]. Exosomes play a dual critical role in aging and cellular senescence, either promoting aging through SASP or exerting anti-aging effects when secreted by young and healthy MSCs [72]. Indeed, extracellular vesicles (EVs) from young MSCs have also been shown to rejuvenate hematopoietic stem cells by transferring autophagy-related and lineage commitment-related mRNAs [73]. EVs can transport pericentromeric non-coding RNAs into neighboring cells, impairing the DNA binding of the CCCTC-binding factor to modify chromosomal accessibility, stimulating an SASP-like inflammatory response [74].
Recent studies have highlighted the significant role of non-coding RNA in the aging and differentiation of MSCs [75]. For instance, miR-486-5p and miRNA-155-5p are elevated in the serum and MSCs of aged human donors but not in those of younger donors [70,76]. miR-486-5p inhibits osteogenic and adipogenic differentiation and induces MSC senescence through the targeted inhibition of SIRT1 expression [76]. miRNA-155-5p induces MSC senescence through mitochondrial dysfunction in an AMPK-dependent manner. Its inhibition has been shown to mitigate cardiac impairment in an aged mouse model, suggesting a potential target for rejuvenating MSCs [77]. The expression of miR-335 in MSCs derived from older subjects has been shown to affect MSC senescence by inhibiting AP-1 activity [78]. MiR-1292 has been shown to regulate senescence and osteogenesis through the Wnt/β-catenin signaling pathway [79].
The miR-17-92 cluster is mainly an oncogenic miRNA cluster [80] that is decreased in senescent adipose tissue-derived MSCs, contributing to the increase in p21 expression, which is associated with senescence. Moreover, the miR-17-92 cluster also affected the oxidative homeostasis of MSCs by regulating thioredoxin-interacting protein, which is induced by oxidative stress and inhibits the antioxidant protein thioredoxin [81].

2.7. Loss of Proteostasis

The proteostasis network is a macromolecular system that coordinates the synthesis, folding, disaggregation, and degradation of proteins. The proteasome system is also responsible for removing normal and damaged proteins, participating in the aging mechanism and longevity regulation. Thus, the loss of proteostasis has deep consequences for aging and age-related diseases. During aging, the aggregation of proteostasis network components can elicit reduced folding capacity, aberrant transcriptional procedures, and the accumulation of misfolded species [82]. Proteostasis impairment also depends on the aging-related pause of ribosome-associated quality control [83]. The autophagy–lysosomal system and the ubiquitin–proteasomal system (UPS) are two crucial pathways for removing misfolded proteins and damaged organelles in the muscles, as revied by Fernando et al. [84]. Aging often shifts the balance between the protein lifecycle in organisms, leading to pathologic conditions. For instance, UPS dysregulation is associated with the aging process and several aging-related diseases in mammals [85].

3. MSCs’ Rejuvenation Strategies

Existing research has focused on new therapeutic strategies based on molecules capable of preventing the senescent status of MSCs. MSCs derived from elder donors exhibit reduced proliferation, differentiation capacities, and stemness compared to those from younger donors, indicating an inverse relationship between the functional capacity of MSCs and their divisional paths. Moreover, it is relevant and beneficial to delay senescence as much as possible, since a remarkable cell expansion is mandatory in regenerative protocols to treat a person who needs one to two million cells per kilogram of body mass, and the yield of MSCs in their niche is estimated to be as low as 1 in every 50 million cells [86]. Therefore, understanding the molecular mechanisms underlying MSC senescence is crucial for optimizing the therapeutic potential of MSCs.
A series of approaches are described in the following section focusing on the techniques that are likely to achieve clinical translation (Table 1).

3.1. Anti-Aging Molecules/Drugs

3.1.1. ROS Reduction Through Antioxidants

Ascorbic acid is a prototypical antioxidant and an mTOR signaling inhibitor. It has been proven to be capable of greatly extending the MSC expansion limit, inhibiting ROS production through AKT/mTOR [87].
The N-acetyl derivative of the amino acid l-cysteine, N-acetyl-L-cysteine (NAC), has a well-established reducing activity, behaving like a reduced glutathione (GSH) precursor (by breaking thiolated proteins and releasing free thiols), which is a substrate of many antioxidant enzymes, and it can also directly act as an antioxidant. Notably, in the case of the significant depletion of endogenous GSH, NAC can act as a direct antioxidant for some oxidant species such as NO2 and hydrogen oxide radicals [88]. Indeed, it has been shown that MSCs, depleted of GSH through a specific inhibitor, died for apoptosis due to H2O2-induced oxidative stress [89]. During the procedure of a bone marrow MSC transplant in a mouse model, the generation of oxidative stress, which causes inflammation, represents a critical issue for the effectiveness of cell therapies. Thus, the use of NAC to inhibit this inflammatory process was investigated, showing that the pre-treatment of MSCs with NAC induced a significant increase in intracellular GSH levels, preventing oxidative stress, reducing apoptosis and senescence, and promoting osteodifferentiation [90].
Supplementation with vitamin E, such as the synthetic compound α-tocopherol-acetate (α-TOA), has been shown to help maintain the proliferative capability of MSCs. It seems that α-TOA exhibits a low-oxygen-concentration-mimicking effect, reducing mitochondrial oxygen consumption. In an in vivo animal model, supplementation with vitamin E guarantees levels of α-TOA which can sustain MSCs [91].
MSC senescence can also be addressed by reducing ROS production with lactoferrin. When senescent human MSCs, induced by hydrogen peroxide exposure, were treated with lactoferrin, the protein effectively suppressed hydrogen peroxide-induced intercellular ROS and apoptosis [92]. This suggests that lactoferrin holds promise as an antioxidant and a potential enhancer of the immunomodulatory potency of MSCs, mitigating the senescence effects triggered by ROS.
Moreover, various strategies have been developed to mitigate ROS-mediated oxidative stress in MSCs, employing drugs such as metformin [93], 5-azacytidine [94], and resveratrol [95]. Metformin is being trialed in humans as the first geroprotective drug, and in adipose-derived MSCs, it reduced replicative senescence likely owing to its ROS scavenging activity [96], which is consistent with the reported activation of AMP-activated protein kinase implicated in mitochondrial homeostasis. Indeed, cell rejuvenation is possibly attained by restoring mitochondrial function. High levels of scientific evidence regarding their antioxidant properties are available, although not specifically in MSCs, for fullerol [97], fucoidan [98], and carvediol [99]. The possible translation of the advantageous properties of pigment epithelium-derived factor (PEDF) (a serpin member of the superfamily of serine protease inhibitors [100], that is prevalently studied in the ophthalmic field) to MSCs is also encouraged. Anecdotally, exendin-4 preconditioning was reported to attenuate apoptosis induced by H2O2 in ASCs [101], similarly to Cirsium setidens [102]. Furthermore, exposure to basic fibroblast growth factor (bFGF) was described as beneficial in reducing liver ischemia–reperfusion injury owing to its antioxidant properties [103]. Ginsenoside RG1 has been shown to extend the lifespan, enhance the proliferation, and promote the colony formation of bone marrow stromal cells. The treatment of bone marrow mononuclear cells with ginsenoside Rg1 reduced apoptotic- and SA-β-gal-positive cells, accompanied with decreased ROS generation and improved colony-forming capacity [104].
Although all these agents have demonstrated effectiveness in reducing senescence in MSCs, caution is suggested as their prolonged usage may affect differentiation capacity. Indeed, at low intrinsic levels, ROS are beneficial for MSCs’ osteogenesis, and their excessive removal can result in a paradoxical induction of senescence in proliferating cells [105] as well as in the suppression of adipogenic potential [106].
Finally, among nutraceuticals (which are naturally occurring biomolecules found in food and other natural sources) that have demonstrated anti-senescence effects [107,108], polyphenols exhibit significant antioxidant and anti-inflammatory properties. Polyphenols are broadly categorized into flavonoids and non-flavonoids and have been shown to exert anti-SASP effects by downregulating oxidative stress and inflammatory pathways. It has been reported that MSCs’ stemness could be promoted by some food-derived nutrients [108,109].

3.1.2. Autophagy Regulation

The autophagy–lysosomal pathway is important in maintaining cellular equilibrium, and targeting this pathway has emerged as a promising anti-aging strategy by either suppressing cellular senescence or inducing apoptosis in senescent cells. Regulating the autophagy level is also a way to rejuvenate senescent MSCs. Treatment with the autophagy inhibitor rapamycin considerably downregulated SASP in senescent MSCs. Among the most promising anti-senescent drugs, rapamycin is a potent inhibitor of the mechanistic Target Of Rapamycin Complex One (mTORC1) protein kinase, which showed anti-aging properties such as the potential to restore differentiation and proliferation, rescue nuclear membrane deformation, revert morphological changes, and activate SA-β-gal and p53/p21 expression in diverse model systems, including stem cells derived from a progeroid mouse model [110]. It is noteworthy that these beneficial effects rely on the inhibition of mTORC1, while most unwanted side effects are due to the inhibition of mTORC2, which has paved the way for exploring a therapeutic strategy selectively targeting mTORC1 [111]. Moreover, the overexpression of macrophage migration inhibitory factor (MIF) has been demonstrated to rejuvenate aged MSCs by activating autophagy [112]. Different clinical trials test the safety and efficacy of rapamycin in attenuating the aging process, but to date, no human data have supported its use as a geroprotector, which is a remarkable translational knowledge gap [111].

3.1.3. Mitochondria Targeting

As regards the drugs targeting mitochondria with senotherapeutic potential, besides the antioxidant mitoquinone [113], it is worth mentioning melatonin. When compared to untreated controls, melatonin-treated senescent MSCs seem to possess enhanced therapeutic potential, sustaining better ischemic recovery and neo-vascularization in murine models [114]. From a molecular point of view, melatonin increases sirtuin 1 (SIRT1) expression while inhibiting ROS accumulation, and it also activates mitophagy which can clear damaged mitochondria [114]. Sirtuins, a family of NAD-dependent HDACs, regulate glucose and insulin metabolism, protein homeostasis, and circadian rhythms, resulting in the pivotal delay of cellular senescence. To date, three sirtuin members (SIRT1, SIRT3, and SIRT6) have been identified as potential anti-aging factors.
Likewise, other molecules promoting SIRT1 activity, such as resveratrol or nicotinamide adenine dinucleotide (NAD), ameliorate the “stemness” properties of early-passage MSCs while rescuing the functional impairment of senescent cells [115]. Unfortunately, this outcome is only achieved at high drug concentrations or through prolonged use. To counteract this pitfall, Wang et al. recently proposed a “targeting nanoplatform with a strong affinity for senescent MSCs through conjugation with anti-Kremen 1”, which may be useful in future nano-therapeutics [116].
The precise mechanisms underlying the anti-aging effects of SIRT1 and SIRT3 are not fully elucidated but are believed to involve the maintenance of genomic stability. The depletion of SIRT3 accelerates aging and inhibits MSC differentiation into osteoblasts and adipocytes. In late-passage MSCs, the overexpression of SIRT3 can reduce oxidative stress, restore their differentiation capacity, and mitigate senescence [117]. SIRT6 specifically deacetylates histone lysine residues H3K9, H3K18, and H3K56, which maintain genomic integrity through the formation of a repressive heterochromatin structure [118,119].

3.1.4. DNA demethylation

Epigenetics refers to heritable changes in gene expression that occur during cell division without alterations in the DNA sequence itself [120]. Chemical modifications to the DNA molecule and associated histone proteins can influence the chromatin structure, thereby altering the accessibility of transcription factors to gene regulatory regions. These modifications are critical for maintaining cells in an undifferentiated state or guiding them toward specific cell fate decisions. DNA methylation is mediated by DNMT enzymes, with DNMT1 functioning as a maintenance methyltransferase, ensuring the propagation of methylation patterns at replication forks [121]. DNMT3A and DNMT3B are responsible for de novo DNA methylation, introducing methyl groups to previously unmethylated CpG sites [112]. Conversely, active DNA demethylation involves the oxidation of methylcytosine to hydroxymethylcytosine, a process catalyzed by Ten-eleven Translocation (TET) enzymes [122]. DNMT inhibitors are a category of anti-senescence drugs that have been acquiring interest in the past few years. RG108 is a small molecule specifically designed to inhibit the catalytic activity of DNMTs [123]. Its demonstrated capacity to reactivate several tumor suppressor genes, coupled with its lack of cytotoxicity [123,124], positions RG108 as a promising candidate for epigenetic modulation therapies in regenerative protocols. The effect of RG108 has been studied in the treatment of aging-related diseases, demonstrating its beneficial effects in restoring aberrant DNA methylation patterns in human MSCs [125]. Subsequently, it was further shown that the senescence phenotype driven by excessive DNMT expression observed in amyotrophic lateral sclerosis (ALS) could be mitigated by RG108 treatment [126]. The authors proposed that RG108 may enhance the therapeutic efficacy of hMSCs in stem cell therapy, as it was shown to restore cell function by improving stem cell potency, enhancing cell migration, providing protection against oxidative stress, and reducing senescence. Also, on bone marrow derived-MSCs of swine origin, RG108 exerted an effect on pluripotency gene expression, apoptosis, and senescence [127]. These data were confirmed in Periodontal Ligament-Derived Stem Cells (PDLSCs) derived from patients with periodontitis, which show some senescent characteristics. Indeed, treatment with RG108 was able to suppress the senescent phenotype of PDLSCs harvested from patients with periodontitis by inducing the downregulation of p16 and p21, increasing the expression of stemness genes, such as OCT4, and rescuing multi-differentiation ability [128].

3.1.5. Senolytics: The Elimination of Senescent Cells

Senescent MSCs are more resistant to apoptotic stimuli; thus, senolytic drugs effectively eliminate senescent cells by targeting their anti-apoptotic pathways, and they represent a further pharmacological strategy to treat many age-related diseases [129,130].
Proteomic and transcriptomic analyses identified many senescent cell anti-apoptotic pathways (SCAPs), which are more expressed in senescent than non-senescent cells and account for the apoptosis resistance of senescent cells [131].
The BCL-2 gene family contains the main regulators of programmed cell death [132]; indeed, its members are upregulated during cellular senescence and downregulated or inhibited when promoting the apoptosis of senescent cells. Several BCL-2 inhibitors have been identified as promising senolytic agents [133,134,135], but their use is restricted to clinical trials since they can induce significant side effects [136]. Dasatinib, an FDA-approved anti-cancer drug, acts as a tyrosine kinase inhibitor, effectively reducing cell proliferation and migration while inducing apoptosis [137], and it kills senescent human fat cell progenitors, while quercetin is more effective against senescent human endothelial cells and mouse bone marrow MSCs [131]. Senolytic flavonoids, such as quercetin and fisetin, induce apoptosis by blocking members of the BCL-2 family such BCLxL, as well as HIF-1 and other SCAP network elements [138]. Quercetin showed a wide range of biological activities, including interactions with specific isoforms of PI3K and members of the BCL-2 family. Moreover, quercetin alone or associated with Dasatinib has been proven to reduce the number of senescent mouse bone marrow MSCs [131]. Fisetin attenuates senescence markers while maintaining the differentiation potential of adipose-derived mesenchymal stem cells [139].
Other mechanisms of apoptotic resistance, dependent on different proteins or pathways, are activated in senescent cells; thus, new molecular targets have been developed for the development of therapies to clear senescent cells. Recently, p53 and its associated regulatory networks have emerged as other targets for the development of senolytic therapies. The transcription factor p53 has a fundamental role in various biological processes, such as the regulation of cell growth and apoptosis with both the initiation and maintenance of senescence, DNA repair, and cellular stress response [140].
The E3 ligase murine double minute 2 (MDM2) is a negative regulator of p53, promoting its degradation via the proteasome. As a small-molecule inhibitor of the MDM2/p53 interaction, RG7112 is used to restore p53 activity, inducing senolysis in senescent intervertebral disc cells, reducing SASP factors such as IFN-γ, IL-6, and CCL24 in vitro [141].
Ubiquitin-specific peptidase 7 (USP7) deubiquitinates MDM2, preventing its degradation via the ubiquitin–proteasome system, thus offering an alternative approach to stabilization and upregulation [142]. USP7 inhibitors have shown the ability to selectively eliminate senescent cells by reducing MDM2 levels and increasing p53 activity, ultimately leading to the induction of pro-apoptotic proteins [143].
Finally, proteolysis-targeting chimeras (PROTACs) are bifunctional molecules consisting of a ligand that binds a target protein and another ligand that recruits an E3 ubiquitin ligase connected by an optimized linker. PROTACs function by bringing the target protein and E3 ligase into close proximity, leading to the ubiquitination and subsequent degradation of the target protein by the ubiquitin–proteasome system [142]. Unlike traditional inhibitors which inhibit the activity of the target proteins, PROTACs act in a catalytic manner to degrade the target [144]. This approach has been applied to enhance the efficacy and specificity of senolytic therapies. For instance, a novel PROTAC-based senolytic, PZ15227, that binds to BCL-XL, demonstrated great efficacy in clearing senescent cells [145]. A second generation of senolytic drugs has been identified through high-throughput library screens and includes antibody−drug conjugates, lysosomal- and SA-β-gal-activated pro-drugs, sodium–potassium pump (Na+/K+-ATPase), and immune-mediated clearance by CAR T cells [135].

3.1.6. Senomorphics: Suppression of MSC SASP

The initial hypothesis that MSCs contribute to tissue regeneration primarily through homing to injury sites and subsequent differentiation has been reconsidered due to the short implantation time of MSCs, which is typically insufficient to exert a substantial impact. It is now recognized that the survival rate of MSCs is less than 1% one week after systemic administration [146,147,148]. However, MSCs exert significant biological effects by promoting cell-to-cell interactions and cellular proliferation, largely through the secretion of paracrine factors such as growth factors and cytokines. Consequently, the MSC secretome may represent a novel avenue in medical biotechnology. Agents suppressing SASP production are called senomorphics, and they have been studied and are currently under investigation for their potential capability to prevent or treat age-related diseases and to extend healthspan [149]. Nonetheless, SASP inhibitors need continuous treatment to exert SASP suppression; thus, they could show more side effects compared to senolytic agents, which are taken on an intermittent schedule.
SASP inhibitors target pathways such as p38 MAPK, JAK/STAT, and interleukins. p38MAPK is an MAPK family member and regulates SASP; thus, p38MAPK inhibitors potently suppress SASP expression in senescent cells [150].
The JAK/STAT pathway regulates cytokine production [151]. JAK inhibitors repress the SASP of adipocyte progenitors by reducing systemic inflammation associated with aging [151].
Senescent normal and dysplastic oral keratinocytes release different levels of pro-inflammatory cytokines, and treatment with Rho kinase inhibitors, such as Y-27632, can reduce the dysfunctional levels of these cytokines without affecting permanent cell growth arrest [152].
Table 1. Therapeutic strategies for anti-MSC senescence. This table presents an overview of various therapeutic strategies aimed at retarding the onset of MSC senescence, which ranges from in vivo studies to clinical trials (Phase I–III). Each category outlines the targeted senescence feature and the mechanism of action and provides the current evidence level.
Table 1. Therapeutic strategies for anti-MSC senescence. This table presents an overview of various therapeutic strategies aimed at retarding the onset of MSC senescence, which ranges from in vivo studies to clinical trials (Phase I–III). Each category outlines the targeted senescence feature and the mechanism of action and provides the current evidence level.
Therapeutic Strategy Mechanism of ActionLevel of Evidence
ROS Reduction
Ascorbic AcidInhibition of AKT/mTOR pathwayIn Vivo [88]
LactoferrinSuppression of hydrogen peroxide-induced intercellular ROS and apoptosisIn Vivo [93]
Ginsenoside RG1Reduction in SA-β-gal-positive cells and apoptotic cells, decreased ROS generationIn Vivo [105]
Autophagy Regulation
RapamycinInhibition of mTORC1 protein kinaseClinical Trial (I-II) [111]
MetforminROS scavenging action, linked to activation of AMP-activated protein kinaseClinical Trial (II-III) [153]
Mitochondrial Targeting
MelatoninIncrease in SIRT1 expression, inhibition of ROS accumulation, activation of mitophagyClinical Trial (I-II) [154,155]
Nutraceutical
PolyphenolsAntioxidant and anti-inflammatory properties (SASP reduction)Clinical Trial (II-III) [155]
MethylsulfonylmethaneInduction of MSC differentiationClinical Trial (I-II) [156]
DNA demethylation
RG108DNMT inhibitorsIn Vitro [157]
Senolytic Drugs
DasatinibBCL-2 family inhibitorsClinical Trial (I-II) [158]
QuercetinBCL-2 family inhibitorsClinical Trial (I-II) [158]
FisetinBCL-2 family inhibitorsClinical Trial (I) [159,160]
RG7112Restores p53 activity and reduces SASP factorsClinical Trial (I) [161]
USP7 inhibitorsUSP7 inhibition, whichh leads to reduced MDM2 levels and increased p53 activityIn Vivo [143]
PROTACsLeads to E3 ubiquitination and subsequent degradation (PZ15227)In Vivo [144]
Senomorphic Drugs
SASP InhibitorsSuppression of p38 MAPK, JAK/STAT, and Rho/Kinase inhibitor pathwaysClinical Trial (I-II) [160]
Non-coding RNAs
MiR-335Inhibition of AP-1 activityIn Vivo [78]
MiR-1292Regulation of Wnt/β-catenin signaling pathwayIn Vivo [79]
MiR-17-92Contributes to increased p21 expression andaffects oxidative homeostasisIn Vivo [82]
miR-486-5pInduces MSC senescence by targeted inhibition of SIRT1 expression, induces mitochondrial dysfunction in AMPK-dependent mannerIn Vivo [162]
miRNA-155-5pInduces mitochondrial dysfunction in AMPK-dependent mannerIn Vivo [163]

4. Conclusions

Although MSCs are widely regarded as a powerful and safe therapeutic option, their actual translation to clinics has been limited to date. Key hurdles to MSC usage are cellular senescence and a natural response to accumulated stress-induced damage. To address this shortcoming, researchers have been working on several strategies capable of delaying or even hopefully reverting the senescent phenotypes of MSCs. In the present review, the authors focused on the recent advancements in the study of therapeutic anti-senescence approaches for MSCs. More studies are needed to elucidate the physiological mechanisms underpinning stem cell aging, aiming at developing novel methods that may extend the functional lifespan of stem cells in a safe, reproducible, effective, and tunable way. Indeed, the off-target effects of anti-senescent therapies may even include oncogenesis and thus require the uttermost attention to safety issues besides efficacy evidence.

Author Contributions

Conceptualization, I.R. and F.M.; methodology, I.R. and M.V.; investigation, I.R. and M.V.; data curation, I.R. and F.M.; writing—original draft preparation, I.R. and M.V.; writing—review and editing, I.R. and F.M.; funding acquisition, F.M. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by PRIN Concerto (Prot. 2020BN5ZW9) and CRT Foundation (CRT2021).

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Poliwoda, S.; Noor, N.; Downs, E.; Schaaf, A.; Cantwell, A.; Ganti, L.; Kaye, A.D.; Mosel, L.I.; Carroll, C.B.; Viswanath, O.; et al. Stem Cells: A Comprehensive Review of Origins and Emerging Clinical Roles in Medical Practice. Orthop. Rev. 2022, 14, 37498. [Google Scholar] [CrossRef]
  2. Kuci, S.; Kuci, Z.; Latifi-Pupovci, H.; Niethammer, D.; Handgretinger, R.; Schumm, M.; Bruchelt, G.; Bader, P.; Klingebiel, T. Adult Stem Cells as an Alternative Source of Multipotential (Pluripotential) Cells in Regenerative Medicine. Curr. Stem Cell Res. Ther. 2009, 4, 107–117. [Google Scholar] [CrossRef]
  3. Dominici, M.; Le Blanc, K.; Mueller, I.; Slaper-Cortenbach, I.; Marini, F.; Krause, D.; Deans, R.; Keating, A.; Prockop, D.; Horwitz, E. Minimal Criteria for Defining Multipotent Mesenchymal Stromal Cells. The International Society for Cellular Therapy Position Statement. Cytotherapy 2006, 8, 315–317. [Google Scholar] [CrossRef]
  4. Roato, I.; Chinigo, G.; Genova, T.; Munaron, L.; Mussano, F. Oral Cavity as a Source of Mesenchymal Stem Cells Useful for Regenerative Medicine in Dentistry. Biomedicines 2021, 9, 1085. [Google Scholar] [CrossRef]
  5. Zhou, T.; Yuan, Z.; Weng, J.; Pei, D.; Du, X.; He, C.; Lai, P. Challenges and Advances in Clinical Applications of Mesenchymal Stromal Cells. J. Hematol. Oncol. 2021, 14, 24. [Google Scholar] [CrossRef]
  6. Levy, O.; Kuai, R.; Siren, E.M.J.; Bhere, D.; Milton, Y.; Nissar, N.; De Biasio, M.; Heinelt, M.; Reeve, B.; Abdi, R.; et al. Shattering Barriers toward Clinically Meaningful MSC Therapies. Sci. Adv. 2020, 6, eaba6884. [Google Scholar] [CrossRef]
  7. Wright, A.; Arthaud-Day, M.L.; Weiss, M.L. Therapeutic Use of Mesenchymal Stromal Cells: The Need for Inclusive Characterization Guidelines to Accommodate All Tissue Sources and Species. Front. Cell Dev. Biol. 2021, 9, 632717. [Google Scholar] [CrossRef]
  8. O’Hagan-Wong, K.; Nadeau, S.; Carrier-Leclerc, A.; Apablaza, F.; Hamdy, R.; Shum-Tim, D.; Rodier, F.; Colmegna, I. Increased IL-6 Secretion by Aged Human Mesenchymal Stromal Cells Disrupts Hematopoietic Stem and Progenitor Cells’ Homeostasis. Oncotarget 2016, 7, 13285–13296. [Google Scholar] [CrossRef]
  9. Wang, B.; Liu, Z.; Chen, V.P.; Wang, L.; Inman, C.L.; Zhou, Y.; Guo, C.; Tchkonia, T.; Rowe, D.W.; Kuchel, G.A.; et al. Transplanting Cells from Old but Not Young Donors Causes Physical Dysfunction in Older Recipients. Aging Cell 2020, 19, e13106. [Google Scholar] [CrossRef]
  10. 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]
  11. Hu, L.; Li, H.; Zi, M.; Li, W.; Liu, J.; Yang, Y.; Zhou, D.; Kong, Q.P.; Zhang, Y.; He, Y. Why Senescent Cells Are Resistant to Apoptosis: An Insight for Senolytic Development. Front. Cell Dev. Biol. 2022, 10, 822816. [Google Scholar] [CrossRef]
  12. Green, D.R.; Galluzzi, L.; Kroemer, G. Mitochondria and the Autophagy-Inflammation-Cell Death Axis in Organismal Aging. Science 2011, 333, 1109–1112. [Google Scholar] [CrossRef]
  13. Giampieri, F.; Afrin, S.; Forbes-Hernandez, T.Y.; Gasparrini, M.; Cianciosi, D.; Reboredo-Rodriguez, P.; Varela-Lopez, A.; Quiles, J.L.; Battino, M. Autophagy in Human Health and Disease: Novel Therapeutic Opportunities. Antioxid. Redox Signal 2019, 30, 577–634. [Google Scholar] [CrossRef]
  14. Rastaldo, R.; Vitale, E.; Giachino, C. Dual Role of Autophagy in Regulation of Mesenchymal Stem Cell Senescence. Front. Cell Dev. Biol. 2020, 8, 276. [Google Scholar] [CrossRef]
  15. McHugh, D.; Gil, J. Senescence and Aging: Causes, Consequences, and Therapeutic Avenues. J. Cell Biol. 2018, 217, 65–77. [Google Scholar] [CrossRef]
  16. 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]
  17. Storer, M.; Mas, A.; Robert-Moreno, A.; Pecoraro, M.; Ortells, M.C.; Di Giacomo, V.; Yosef, R.; Pilpel, N.; Krizhanovsky, V.; Sharpe, J.; et al. Senescence Is a Developmental Mechanism That Contributes to Embryonic Growth and Patterning. Cell 2013, 155, 1119–1130. [Google Scholar] [CrossRef]
  18. Volonte, D.; Galbiati, F. Caveolin-1, a Master Regulator of Cellular Senescence. Cancer Metastasis Rev. 2020, 39, 397–414. [Google Scholar] [CrossRef]
  19. Xu, C.; Wang, J.; Zhu, T.; Shen, Y.; Tang, X.; Fang, L.; Xu, Y. Cross-Talking Between PPAR and WNT Signaling and Its Regulation in Mesenchymal Stem Cell Differentiation. Curr. Stem Cell Res. Ther. 2016, 11, 247–254. [Google Scholar] [CrossRef]
  20. Hayflick, L.; Moorhead, P.S. The Serial Cultivation of Human Diploid Cell Strains. Exp. Cell Res. 1961, 25, 585–621. [Google Scholar] [CrossRef]
  21. Gonzalez-Gualda, E.; Baker, A.G.; Fruk, L.; Munoz-Espin, D. A Guide to Assessing Cellular Senescence in Vitro and in Vivo. FEBS J. 2021, 288, 56–80. [Google Scholar] [CrossRef]
  22. Galbiati, A.; Beausejour, C.; d’Adda di Fagagna, F. A Novel Single-Cell Method Provides Direct Evidence of Persistent DNA Damage in Senescent Cells and Aged Mammalian Tissues. Aging Cell 2017, 16, 422–427. [Google Scholar] [CrossRef]
  23. Zhang, Y.; Oliveira, A.N.; Hood, D.A. The Intersection of Exercise and Aging on Mitochondrial Protein Quality Control. Exp. Gerontol. 2020, 131, 110824. [Google Scholar] [CrossRef]
  24. Lee, S.S.; Vu, T.T.; Weiss, A.S.; Yeo, G.C. Stress-Induced Senescence in Mesenchymal Stem Cells: Triggers, Hallmarks, and Current Rejuvenation Approaches. Eur. J. Cell Biol. 2023, 102, 151331. [Google Scholar] [CrossRef]
  25. Bertolo, A.; Baur, M.; Guerrero, J.; Potzel, T.; Stoyanov, J. Autofluorescence Is a Reliable in Vitro Marker of Cellular Senescence in Human Mesenchymal Stromal Cells. Sci. Rep. 2019, 9, 2074. [Google Scholar] [CrossRef]
  26. Yoon, K.B.; Park, K.R.; Kim, S.Y.; Han, S.Y. Induction of Nuclear Enlargement and Senescence by Sirtuin Inhibitors in Glioblastoma Cells. Immune Netw. 2016, 16, 183–188. [Google Scholar] [CrossRef]
  27. Schumacher, B.; Pothof, J.; Vijg, J.; Hoeijmakers, J.H.J. The Central Role of DNA Damage in the Ageing Process. Nature 2021, 592, 695–703. [Google Scholar] [CrossRef]
  28. Zhou, X.; Hong, Y.; Zhang, H.; Li, X. Mesenchymal Stem Cell Senescence and Rejuvenation: Current Status and Challenges. Front. Cell Dev. Biol. 2020, 8, 364. [Google Scholar] [CrossRef]
  29. Yang, S.R.; Park, J.R.; Kang, K.S. Reactive Oxygen Species in Mesenchymal Stem Cell Aging: Implication to Lung Diseases. Oxid. Med. Cell Longev. 2015, 2015, 486263. [Google Scholar] [CrossRef]
  30. Weng, Z.; Wang, Y.; Ouchi, T.; Liu, H.; Qiao, X.; Wu, C.; Zhao, Z.; Li, L.; Li, B. Mesenchymal Stem/Stromal Cell Senescence: Hallmarks, Mechanisms, and Combating Strategies. Stem Cells Transl. Med. 2022, 11, 356–371. [Google Scholar] [CrossRef]
  31. Liao, N.; Shi, Y.; Zhang, C.; Zheng, Y.; Wang, Y.; Zhao, B.; Zeng, Y.; Liu, X.; Liu, J. Antioxidants Inhibit Cell Senescence and Preserve Stemness of Adipose Tissue-Derived Stem Cells by Reducing ROS Generation during Long-Term in Vitro Expansion. Stem Cell Res. Ther. 2019, 10, 306. [Google Scholar] [CrossRef]
  32. Jin, H.J.; Lee, H.J.; Heo, J.; Lim, J.; Kim, M.; Kim, M.K.; Nam, H.Y.; Hong, G.H.; Cho, Y.S.; Choi, S.J.; et al. Senescence-Associated MCP-1 Secretion Is Dependent on a Decline in BMI1 in Human Mesenchymal Stromal Cells. Antioxid. Redox Signal 2016, 24, 471–485. [Google Scholar] [CrossRef]
  33. Abraham, A.G.; O’Neill, E. PI3K/Akt-Mediated Regulation of P53 in Cancer. Biochem. Soc. Trans. 2014, 42, 798–803. [Google Scholar] [CrossRef]
  34. Zhang, X.S.; Zhang, X.; Wu, Q.; Li, W.; Zhang, Q.R.; Wang, C.X.; Zhou, X.M.; Li, H.; Shi, J.X.; Zhou, M.L. Astaxanthin Alleviates Early Brain Injury Following Subarachnoid Hemorrhage in Rats: Possible Involvement of Akt/Bad Signaling. Mar. Drugs 2014, 12, 4291–4310. [Google Scholar] [CrossRef]
  35. Jiramongkol, Y.; Lam, E.W. FOXO Transcription Factor Family in Cancer and Metastasis. Cancer Metastasis Rev. 2020, 39, 681–709. [Google Scholar] [CrossRef]
  36. Liu, J.; He, J.; Ge, L.; Xiao, H.; Huang, Y.; Zeng, L.; Jiang, Z.; Lu, M.; Hu, Z. Hypoxic Preconditioning Rejuvenates Mesenchymal Stem Cells and Enhances Neuroprotection Following Intracerebral Hemorrhage via the MiR-326-Mediated Autophagy. Stem Cell Res. Ther. 2021, 12, 413. [Google Scholar] [CrossRef]
  37. Baker, D.J.; Childs, B.G.; Durik, M.; Wijers, M.E.; Sieben, C.J.; Zhong, J.; Saltness, R.A.; Jeganathan, K.B.; Verzosa, G.C.; Pezeshki, A.; et al. Naturally Occurring P16(Ink4a)-Positive Cells Shorten Healthy Lifespan. Nature 2016, 530, 184–189. [Google Scholar] [CrossRef]
  38. Huang, Y.; Corbley, M.J.; Tang, Z.; Yang, L.; Peng, Y.; Zhang, Z.Y.; Tong, T.J. Down-Regulation of P21WAF1 Promotes Apoptosis in Senescent Human Fibroblasts: Involvement of Retinoblastoma Protein Phosphorylation and Delay of Cellular Aging. J. Cell Physiol. 2004, 201, 483–491. [Google Scholar] [CrossRef]
  39. Sanidas, I.; Morris, R.; Fella, K.A.; Rumde, P.H.; Boukhali, M.; Tai, E.C.; Ting, D.T.; Lawrence, M.S.; Haas, W.; Dyson, N.J. A Code of Mono-Phosphorylation Modulates the Function of RB. Mol. Cell 2019, 73, 985–1000.e6. [Google Scholar] [CrossRef]
  40. Wells, J.; Graveel, C.R.; Bartley, S.M.; Madore, S.J.; Farnham, P.J. The Identification of E2F1-Specific Target Genes. Proc. Natl. Acad. Sci. USA 2002, 99, 3890–3895. [Google Scholar] [CrossRef]
  41. Galanos, P.; Vougas, K.; Walter, D.; Polyzos, A.; Maya-Mendoza, A.; Haagensen, E.J.; Kokkalis, A.; Roumelioti, F.M.; Gagos, S.; Tzetis, M.; et al. Chronic P53-Independent P21 Expression Causes Genomic Instability by Deregulating Replication Licensing. Nat. Cell Biol. 2016, 18, 777–789. [Google Scholar] [CrossRef] [PubMed]
  42. Milanovic, M.; Fan, D.N.Y.; Belenki, D.; Däbritz, J.H.M.; Zhao, Z.; Yu, Y.; Dörr, J.R.; Dimitrova, L.; Lenze, D.; Monteiro Barbosa, I.A.; et al. Senescence-Associated Reprogramming Promotes Cancer Stemness. Nature 2018, 553, 96–100. [Google Scholar] [CrossRef] [PubMed]
  43. Hoshino, A.; Mita, Y.; Okawa, Y.; Ariyoshi, M.; Iwai-Kanai, E.; Ueyama, T.; Ikeda, K.; Ogata, T.; Matoba, S. Cytosolic P53 Inhibits Parkin-Mediated Mitophagy and Promotes Mitochondrial Dysfunction in the Mouse Heart. Nat. Commun. 2013, 4, 2308. [Google Scholar] [CrossRef] [PubMed]
  44. Zhang, F.; Peng, W.; Zhang, J.; Dong, W.; Wu, J.; Wang, T.; Xie, Z. P53 and Parkin Co-Regulate Mitophagy in Bone Marrow Mesenchymal Stem Cells to Promote the Repair of Early Steroid-Induced Osteonecrosis of the Femoral Head. Cell Death Dis. 2020, 11, 42. [Google Scholar] [CrossRef] [PubMed]
  45. Chistiakov, D.A.; Sobenin, I.A.; Revin, V.V.; Orekhov, A.N.; Bobryshev, Y.V. Mitochondrial Aging and Age-Related Dysfunction of Mitochondria. Biomed. Res. Int. 2014, 2014, 238463. [Google Scholar] [CrossRef]
  46. Somasundaram, I.; Jain, S.M.; Blot-Chabaud, M.; Pathak, S.; Banerjee, A.; Rawat, S.; Sharma, N.R.; Duttaroy, A.K. Mitochondrial Dysfunction and Its Association with Age-Related Disorders. Front. Physiol. 2024, 15, 1384966. [Google Scholar] [CrossRef]
  47. Summer, R.; Shaghaghi, H.; Schriner, D.; Roque, W.; Sales, D.; Cuevas-Mora, K.; Desai, V.; Bhushan, A.; Ramirez, M.I.; Romero, F. Activation of the MTORC1/PGC-1 Axis Promotes Mitochondrial Biogenesis and Induces Cellular Senescence in the Lung Epithelium. Am. J. Physiol. Lung Cell Mol. Physiol. 2019, 316, L1049–L1060. [Google Scholar] [CrossRef]
  48. Shay, J.W.; Wright, W.E. Telomeres and Telomerase: Three Decades of Progress. Nat. Rev. Genet. 2019, 20, 299–309. [Google Scholar] [CrossRef]
  49. Harley, C.B.; Futcher, A.B.; Greider, C.W. Telomeres Shorten during Ageing of Human Fibroblasts. Nature 1990, 345, 458–460. [Google Scholar] [CrossRef]
  50. Rossiello, F.; Jurk, D.; Passos, J.F.; d’Adda di Fagagna, F. Telomere Dysfunction in Ageing and Age-Related Diseases. Nat. Cell Biol. 2022, 24, 135–147. [Google Scholar] [CrossRef]
  51. di Fagagna, A.; Reaper, P.M.; Clay-Farrace, L.; Fiegler, H.; Carr, P.; von Zglinicki, T.; Saretzki, G.; Carter, N.P.; Jackson, S.P. A DNA Damage Checkpoint Response in Telomere-Initiated Senescence. Nature 2003, 426, 194–198. [Google Scholar] [CrossRef]
  52. Cesare, A.J.; Karlseder, J. A Three-State Model of Telomere Control over Human Proliferative Boundaries. Curr. Opin. Cell Biol. 2012, 24, 731–738. [Google Scholar] [CrossRef]
  53. Chakravarti, D.; LaBella, K.A.; DePinho, R.A. Telomeres: History, Health, and Hallmarks of Aging. Cell 2021, 184, 306–322. [Google Scholar] [CrossRef]
  54. Lee, S.; Park, J.R.; Seo, M.S.; Roh, K.H.; Park, S.B.; Hwang, J.W.; Sun, B.; Seo, K.; Lee, Y.S.; Kang, S.K.; et al. Histone Deacetylase Inhibitors Decrease Proliferation Potential and Multilineage Differentiation Capability of Human Mesenchymal Stem Cells. Cell Prolif. 2009, 42, 711–720. [Google Scholar] [CrossRef]
  55. Ito, T.; Teo, Y.V.; Evans, S.A.; Neretti, N.; Sedivy, J.M. Regulation of Cellular Senescence by Polycomb Chromatin Modifiers through Distinct DNA Damage- and Histone Methylation-Dependent Pathways. Cell Rep. 2018, 22, 3480–3492. [Google Scholar] [CrossRef]
  56. Smith, Z.D.; Meissner, A. DNA Methylation: Roles in Mammalian Development. Nat. Rev. Genet. 2013, 14, 204–220. [Google Scholar] [CrossRef]
  57. Suzuki, M.M.; Bird, A. DNA Methylation Landscapes: Provocative Insights from Epigenomics. Nat. Rev. Genet. 2008, 9, 465–476. [Google Scholar] [CrossRef]
  58. Cheng, Y.; Xie, N.; Jin, P.; Wang, T. DNA Methylation and Hydroxymethylation in Stem Cells. Cell Biochem. Funct. 2015, 33, 161–173. [Google Scholar] [CrossRef] [PubMed]
  59. Nagae, G.; Isagawa, T.; Shiraki, N.; Fujita, T.; Yamamoto, S.; Tsutsumi, S.; Nonaka, A.; Yoshiba, S.; Matsusaka, K.; Midorikawa, Y.; et al. Tissue-Specific Demethylation in CpG-Poor Promoters during Cellular Differentiation. Hum. Mol. Genet. 2011, 20, 2710–2721. [Google Scholar] [CrossRef] [PubMed]
  60. So, A.Y.; Jung, J.W.; Lee, S.; Kim, H.S.; Kang, K.S. DNA Methyltransferase Controls Stem Cell Aging by Regulating BMI1 and EZH2 through MicroRNAs. PLoS ONE 2011, 6, e19503. [Google Scholar] [CrossRef] [PubMed]
  61. Sagiv, A.; Biran, A.; Yon, M.; Simon, J.; Lowe, S.W.; Krizhanovsky, V. Granule Exocytosis Mediates Immune Surveillance of Senescent Cells. Oncogene 2013, 32, 1971–1977. [Google Scholar] [CrossRef]
  62. Gnani, D.; Crippa, S.; Della Volpe, L.; Rossella, V.; Conti, A.; Lettera, E.; Rivis, S.; Ometti, M.; Fraschini, G.; Bernardo, M.E.; et al. An Early-Senescence State in Aged Mesenchymal Stromal Cells Contributes to Hematopoietic Stem and Progenitor Cell Clonogenic Impairment through the Activation of a pro-Inflammatory Program. Aging Cell 2019, 18, e12933. [Google Scholar] [CrossRef] [PubMed]
  63. Lepperdinger, G. Inflammation and Mesenchymal Stem Cell Aging. Curr. Opin. Immunol. 2011, 23, 518–524. [Google Scholar] [CrossRef]
  64. Starr, M.E.; Evers, B.M.; Saito, H. Age-Associated Increase in Cytokine Production during Systemic Inflammation: Adipose Tissue as a Major Source of IL-6. J. Gerontol. A Biol. Sci. Med. Sci. 2009, 64, 723–730. [Google Scholar] [CrossRef]
  65. Kale, A.; Sharma, A.; Stolzing, A.; Desprez, P.Y.; Campisi, J. Role of Immune Cells in the Removal of Deleterious Senescent Cells. Immun. Ageing 2020, 17, 16. [Google Scholar] [CrossRef]
  66. Shin, T.H.; Kim, H.S.; Kang, T.W.; Lee, B.C.; Lee, H.Y.; Kim, Y.J.; Shin, J.H.; Seo, Y.; Won Choi, S.; Lee, S.; et al. Human Umbilical Cord Blood-Stem Cells Direct Macrophage Polarization and Block Inflammasome Activation to Alleviate Rheumatoid Arthritis. Cell Death Dis. 2016, 7, e2524. [Google Scholar] [CrossRef]
  67. Oishi, Y.; Manabe, I. Macrophages in Age-Related Chronic Inflammatory Diseases. NPJ Aging Mech. Dis. 2016, 2, 16018. [Google Scholar] [CrossRef]
  68. Kizilay Mancini, O.; Lora, M.; Shum-Tim, D.; Nadeau, S.; Rodier, F.; Colmegna, I. A Proinflammatory Secretome Mediates the Impaired Immunopotency of Human Mesenchymal Stromal Cells in Elderly Patients with Atherosclerosis. Stem Cells Transl. Med. 2017, 6, 1132–1140. [Google Scholar] [CrossRef] [PubMed]
  69. Zhang, Y.; Ravikumar, M.; Ling, L.; Nurcombe, V.; Cool, S.M. Age-Related Changes in the Inflammatory Status of Human Mesenchymal Stem Cells: Implications for Cell Therapy. Stem Cell Rep. 2021, 16, 694–707. [Google Scholar] [CrossRef]
  70. Terlecki-Zaniewicz, L.; Lammermann, I.; Latreille, J.; Bobbili, M.R.; Pils, V.; Schosserer, M.; Weinmullner, R.; Dellago, H.; Skalicky, S.; Pum, D.; et al. Small Extracellular Vesicles and Their MiRNA Cargo Are Anti-Apoptotic Members of the Senescence-Associated Secretory Phenotype. Aging 2018, 10, 1103–1132. [Google Scholar] [CrossRef] [PubMed]
  71. Yang, L.; Li, Y.; Gong, R.; Gao, M.; Feng, C.; Liu, T.; Sun, Y.; Jin, M.; Wang, D.; Yuan, Y.; et al. The Long Non-Coding RNA-ORLNC1 Regulates Bone Mass by Directing Mesenchymal Stem Cell Fate. Mol. Ther. 2019, 27, 394–410. [Google Scholar] [CrossRef]
  72. Cai, J.; Qi, H.; Yao, K.; Yao, Y.; Jing, D.; Liao, W.; Zhao, Z. Non-Coding RNAs Steering the Senescence-Related Progress, Properties, and Application of Mesenchymal Stem Cells. Front. Cell Dev. Biol. 2021, 9, 650431. [Google Scholar] [CrossRef]
  73. Wang, L.; Wei, J.; Da Fonseca Ferreira, A.; Wang, H.; Zhang, L.; Zhang, Q.; Bellio, M.A.; Chu, X.M.; Khan, A.; Jayaweera, D.; et al. Rejuvenation of Senescent Endothelial Progenitor Cells by Extracellular Vesicles Derived from Mesenchymal Stromal Cells. JACC Basic. Transl. Sci. 2020, 5, 1127–1141. [Google Scholar] [CrossRef]
  74. Miyata, K.; Imai, Y.; Hori, S.; Nishio, M.; Loo, T.M.; Okada, R.; Yang, L.; Nakadai, T.; Maruyama, R.; Fujii, R.; et al. Pericentromeric Noncoding RNA Changes DNA Binding of CTCF and Inflammatory Gene Expression in Senescence and Cancer. Proc. Natl. Acad. Sci. USA 2021, 118, e2025647118. [Google Scholar] [CrossRef]
  75. Rossi, M.; Gorospe, M. Noncoding RNAs Controlling Telomere Homeostasis in Senescence and Aging. Trends Mol. Med. 2020, 26, 422–433. [Google Scholar] [CrossRef]
  76. Kim, Y.J.; Hwang, S.H.; Lee, S.Y.; Shin, K.K.; Cho, H.H.; Bae, Y.C.; Jung, J.S. MiR-486-5p Induces Replicative Senescence of Human Adipose Tissue-Derived Mesenchymal Stem Cells and Its Expression Is Controlled by High Glucose. Stem Cells Dev. 2012, 21, 1749–1760. [Google Scholar] [CrossRef]
  77. Hong, Y.; He, H.; Jiang, G.; Zhang, H.; Tao, W.; Ding, Y.; Yuan, D.; Liu, J.; Fan, H.; Lin, F.; et al. MiR-155-5p Inhibition Rejuvenates Aged Mesenchymal Stem Cells and Enhances Cardioprotection Following Infarction. Aging Cell 2020, 19, e13128. [Google Scholar] [CrossRef]
  78. Tome, M.; Sepulveda, J.C.; Delgado, M.; Andrades, J.A.; Campisi, J.; Gonzalez, M.A.; Bernad, A. MiR-335 Correlates with Senescence/Aging in Human Mesenchymal Stem Cells and Inhibits Their Therapeutic Actions through Inhibition of AP-1 Activity. Stem Cells 2014, 32, 2229–2244. [Google Scholar] [CrossRef]
  79. Fan, J.; An, X.; Yang, Y.; Xu, H.; Fan, L.; Deng, L.; Li, T.; Weng, X.; Zhang, J.; Chunhua Zhao, R. MiR-1292 Targets FZD4 to Regulate Senescence and Osteogenic Differentiation of Stem Cells in TE/SJ/Mesenchymal Tissue System via the Wnt/Beta-Catenin Pathway. Aging Dis. 2018, 9, 1103–1121. [Google Scholar] [CrossRef] [PubMed]
  80. Mogilyansky, E.; Rigoutsos, I. The MiR-17/92 Cluster: A Comprehensive Update on Its Genomics, Genetics, Functions and Increasingly Important and Numerous Roles in Health and Disease. Cell Death Differ. 2013, 20, 1603–1614. [Google Scholar] [CrossRef] [PubMed]
  81. Cen, Y.; Qi, J.; Chen, L.; Xia, C.; Zheng, M.; Liu, Y.; Lou, G. Decreased MiR-17-92 Cluster Correlates with Senescence Features, Disrupted Oxidative Homeostasis, and Impaired Therapeutic Efficacy of Mesenchymal Stem Cells. Am. J. Physiol. Cell Physiol. 2023, 325, C443–C455. [Google Scholar] [CrossRef] [PubMed]
  82. Klaips, C.L.; Jayaraj, G.G.; Hartl, F.U. Pathways of Cellular Proteostasis in Aging and Disease. J. Cell Biol. 2018, 217, 51–63. [Google Scholar] [CrossRef] [PubMed]
  83. Stein, K.C.; Morales-Polanco, F.; van der Lienden, J.; Rainbolt, T.K.; Frydman, J. Ageing Exacerbates Ribosome Pausing to Disrupt Cotranslational Proteostasis. Nature 2022, 601, 637–642. [Google Scholar] [CrossRef] [PubMed]
  84. Fernando, R.; Drescher, C.; Nowotny, K.; Grune, T.; Castro, J.P. Impaired Proteostasis during Skeletal Muscle Aging. Free Radic. Biol. Med. 2019, 132, 58–66. [Google Scholar] [CrossRef] [PubMed]
  85. Korovila, I.; Hugo, M.; Castro, J.P.; Weber, D.; Hohn, A.; Grune, T.; Jung, T. Proteostasis, Oxidative Stress and Aging. Redox Biol. 2017, 13, 550–567. [Google Scholar] [CrossRef]
  86. Murphy, M.B.; Moncivais, K.; Caplan, A.I. Mesenchymal Stem Cells: Environmentally Responsive Therapeutics for Regenerative Medicine. Exp. Mol. Med. 2013, 45, e54. [Google Scholar] [CrossRef]
  87. Yang, M.; Teng, S.; Ma, C.; Yu, Y.; Wang, P.; Yi, C. Ascorbic Acid Inhibits Senescence in Mesenchymal Stem Cells through ROS and AKT/MTOR Signaling. Cytotechnology 2018, 70, 1301–1313. [Google Scholar] [CrossRef]
  88. Aldini, G.; Altomare, A.; Baron, G.; Vistoli, G.; Carini, M.; Borsani, L.; Sergio, F. N-Acetylcysteine as an Antioxidant and Disulphide Breaking Agent: The Reasons Why. Free Radic. Res. 2018, 52, 751–762. [Google Scholar] [CrossRef]
  89. Bonilla-Porras, A.R.; Jimenez-Del-Rio, M.; Velez-Pardo, C. N-Acetyl-Cysteine Blunts 6-Hydroxydopamine- and l-Buthionine-Sulfoximine-Induced Apoptosis in Human Mesenchymal Stromal Cells. Mol. Biol. Rep. 2019, 46, 4423–4435. [Google Scholar] [CrossRef]
  90. Watanabe, J.; Yamada, M.; Niibe, K.; Zhang, M.; Kondo, T.; Ishibashi, M.; Egusa, H. Preconditioning of Bone Marrow-Derived Mesenchymal Stem Cells with N-Acetyl-L-Cysteine Enhances Bone Regeneration via Reinforced Resistance to Oxidative Stress. Biomaterials 2018, 185, 25–38. [Google Scholar] [CrossRef]
  91. Loncaric, D.; Rodriguez, L.; Debeissat, C.; Touya, N.; Labat, V.; Villacreces, A.; Bouzier-Sore, A.-K.; Pasquet, J.-M.; Brunet De La Grange, P.; Vlaski-Lafarge, M.; et al. α-Tocopherol Acetate Attenuates Mitochondrial Oxygen Consumption and Maintains Primitive Cells within Mesenchymal Stromal Cell Population. Stem Cell Rev. Rep. 2021, 17, 1390–1405. [Google Scholar] [CrossRef] [PubMed]
  92. Park, S.Y.; Jeong, A.J.; Kim, G.Y.; Jo, A.; Lee, J.E.; Leem, S.H.; Yoon, J.H.; Ye, S.K.; Chung, J.W. Lactoferrin Protects Human Mesenchymal Stem Cells from Oxidative Stress-Induced Senescence and Apoptosis. J. Microbiol. Biotechnol. 2017, 27, 1877–1884. [Google Scholar] [CrossRef] [PubMed]
  93. Liu, X.; Li, Z.; Liu, L.; Zhang, P.; Wang, Y.; Ding, G. Metformin-Mediated Effects on Mesenchymal Stem Cells and Mechanisms: Proliferation, Differentiation and Aging. Front. Pharmacol. 2024, 15, 1465697. [Google Scholar] [CrossRef]
  94. Kornicka, K.; Szlapka-Kosarzewska, J.; Smieszek, A.; Marycz, K. 5-Azacytydine and Resveratrol Reverse Senescence and Ageing of Adipose Stem Cells via Modulation of Mitochondrial Dynamics and Autophagy. J. Cell Mol. Med. 2019, 23, 237–259. [Google Scholar] [CrossRef]
  95. Chen, Y.; Li, D.; Zhang, X.; Ma, Q.; Xu, Y.; Luo, Z. Azacytidine-Induced Hypomethylation Delays Senescence and Coloration in Harvested Strawberries by Stimulating Antioxidant Enzymes and Modulating Abscisate Metabolism to Minimize Anthocyanin Overproduction. Food Chem. 2023, 407, 135189. [Google Scholar] [CrossRef]
  96. Acar, M.B.; Ayaz-Guner, S.; Gunaydin, Z.; Karakukcu, M.; Peluso, G.; Di Bernardo, G.; Ozcan, S.; Galderisi, U. Proteomic and Biological Analysis of the Effects of Metformin Senomorphics on the Mesenchymal Stromal Cells. Front. Bioeng. Biotechnol. 2021, 9, 730813. [Google Scholar] [CrossRef]
  97. Arifa, R.D.N.; de Paula, T.P.; Lima, R.L.; Brito, C.B.; Andrade, M.E.R.; Cardoso, V.N.; Pinheiro, M.V.B.; Ladeira, L.O.; Krambrock, K.; Teixeira, M.M.; et al. Anti-Inflammatory and Antioxidant Effects of the Nanocomposite Fullerol Decrease the Severity of Intestinal Inflammation Induced by Gut Ischemia and Reperfusion. Eur. J. Pharmacol. 2021, 898, 173984. [Google Scholar] [CrossRef] [PubMed]
  98. Laeliocattleya, R.A.; Yunianta, Y.; Risjani, Y.; Wulan, S.N. Characterization of “novel Fucoidan” Extracted from Brown Seaweed (Sargassum echinocarpum J. Ag.) Using Ultrasound-Assisted Extraction (UAE) and Its Potential Antioxidant Activity. Nat. Prod. Res. 2023, 19, 1–13. [Google Scholar] [CrossRef] [PubMed]
  99. Lysko, P.G.; Webb, C.L.; Gu, J.L.; Ohlstein, E.H.; Ruffolo, R.R., Jr.; Yue, T.L. A Comparison of Carvedilol and Metoprolol Antioxidant Activities in Vitro. J. Cardiovasc. Pharmacol. 2000, 36, 277–281. [Google Scholar] [CrossRef]
  100. Yoshida, Y.; Yamagishi, S.; Matsui, T.; Nakamura, K.; Imaizumi, T.; Yoshimura, K.; Yamakawa, R. Positive Correlation of Pigment Epithelium-Derived Factor and Total Antioxidant Capacity in Aqueous Humour of Patients with Uveitis and Proliferative Diabetic Retinopathy. Br. J. Ophthalmol. 2007, 91, 1133–1134. [Google Scholar] [CrossRef]
  101. Zhou, H.; Yang, J.; Xin, T.; Li, D.; Guo, J.; Hu, S.; Zhou, S.; Zhang, T.; Zhang, Y.; Han, T.; et al. Exendin-4 Protects Adipose-Derived Mesenchymal Stem Cells from Apoptosis Induced by Hydrogen Peroxide through the PI3K/Akt-Sfrp2 Pathways. Free Radic. Biol. Med. 2014, 77, 363–375. [Google Scholar] [CrossRef] [PubMed]
  102. Lee, J.H.; Jung, H.K.; Han, Y.S.; Yoon, Y.M.; Yun, C.W.; Sun, H.Y.; Cho, H.W.; Lee, S.H. Antioxidant Effects of Cirsium Setidens Extract on Oxidative Stress in Human Mesenchymal Stem Cells. Mol. Med. Rep. 2016, 14, 3777–3784. [Google Scholar] [CrossRef] [PubMed]
  103. Chen, X.; Tong, G.; Chen, S. Basic Fibroblast Growth Factor Protects against Liver Ischemia-Reperfusion Injury via the Nrf2/Hippo Signaling Pathway. Tissue Cell 2022, 79, 101921. [Google Scholar] [CrossRef] [PubMed]
  104. Zeng, Y.; Hu, W.; Jing, P.; Chen, X.; Wang, Z.; Wang, L.; Wang, Y. The Regulation of Ginsenoside Rg1 upon Aging of Bone Marrow Stromal Cell Contribute to Delaying Senescence of Bone Marrow Mononuclear Cells (BMNCs). Life Sci. 2018, 209, 63–68. [Google Scholar] [CrossRef] [PubMed]
  105. Kornienko, J.S.; Smirnova, I.S.; Pugovkina, N.A.; Ivanova, J.S.; Shilina, M.A.; Grinchuk, T.M.; Shatrova, A.N.; Aksenov, N.D.; Zenin, V.V.; Nikolsky, N.N.; et al. High Doses of Synthetic Antioxidants Induce Premature Senescence in Cultivated Mesenchymal Stem Cells. Sci. Rep. 2019, 9, 1296. [Google Scholar] [CrossRef]
  106. Zhang, Y.; Marsboom, G.; Toth, P.T.; Rehman, J. Mitochondrial Respiration Regulates Adipogenic Differentiation of Human Mesenchymal Stem Cells. PLoS ONE 2013, 8, e77077. [Google Scholar] [CrossRef]
  107. Dong, Y.; Guha, S.; Sun, X.; Cao, M.; Wang, X.; Zou, S. Nutraceutical Interventions for Promoting Healthy Aging in Invertebrate Models. Oxid. Med. Cell Longev. 2012, 2012, 718491. [Google Scholar] [CrossRef]
  108. Gurau, F.; Baldoni, S.; Prattichizzo, F.; Espinosa, E.; Amenta, F.; Procopio, A.D.; Albertini, M.C.; Bonafe, M.; Olivieri, F. Anti-Senescence Compounds: A Potential Nutraceutical Approach to Healthy Aging. Ageing Res. Rev. 2018, 46, 14–31. [Google Scholar] [CrossRef]
  109. Luo, J.; Si, H.; Jia, Z.; Liu, D. Dietary Anti-Aging Polyphenols and Potential Mechanisms. Antioxidants 2021, 10, 283. [Google Scholar] [CrossRef]
  110. Nie, D.; Zhang, J.; Zhou, Y.; Sun, J.; Wang, W.; Wang, J.H. Rapamycin Treatment of Tendon Stem/Progenitor Cells Reduces Cellular Senescence by Upregulating Autophagy. Stem Cells Int. 2021, 2021, 6638249. [Google Scholar] [CrossRef]
  111. Konopka, A.R.; Lamming, D.W.; Investigators, R.P.; Investigators, E. Blazing a Trail for the Clinical Use of Rapamycin as a GeroprotecTOR. Geroscience 2023, 45, 2769–2783. [Google Scholar] [CrossRef] [PubMed]
  112. Zhang, Y.; Zhu, W.; He, H.; Fan, B.; Deng, R.; Hong, Y.; Liang, X.; Zhao, H.; Li, X.; Zhang, F. Macrophage Migration Inhibitory Factor Rejuvenates Aged Human Mesenchymal Stem Cells and Improves Myocardial Repair. Aging 2019, 11, 12641–12660. [Google Scholar] [CrossRef]
  113. Ye, G.; Xie, Z.; Zeng, H.; Wang, P.; Li, J.; Zheng, G.; Wang, S.; Cao, Q.; Li, M.; Liu, W.; et al. Oxidative Stress-Mediated Mitochondrial Dysfunction Facilitates Mesenchymal Stem Cell Senescence in Ankylosing Spondylitis. Cell Death Dis. 2020, 11, 775. [Google Scholar] [CrossRef] [PubMed]
  114. Lee, J.H.; Yoon, Y.M.; Song, K.H.; Noh, H.; Lee, S.H. Melatonin Suppresses Senescence-Derived Mitochondrial Dysfunction in Mesenchymal Stem Cells via the HSPA1L-Mitophagy Pathway. Aging Cell 2020, 19, e13111. [Google Scholar] [CrossRef] [PubMed]
  115. Hu, C.; Li, L. The Application of Resveratrol to Mesenchymal Stromal Cell-Based Regenerative Medicine. Stem Cell Res. Ther. 2019, 10, 307. [Google Scholar] [CrossRef]
  116. Wang, Y.; Xie, F.; He, Z.; Che, L.; Chen, X.; Yuan, Y.; Liu, C. Senescence-Targeted and NAD(+)-Dependent SIRT1-Activated Nanoplatform to Counteract Stem Cell Senescence for Promoting Aged Bone Regeneration. Small 2023, e2304433. [Google Scholar] [CrossRef]
  117. Denu, R.A. SIRT3 Enhances Mesenchymal Stem Cell Longevity and Differentiation. Oxid. Med. Cell Longev. 2017, 2017, 5841716. [Google Scholar] [CrossRef]
  118. Michishita, E.; McCord, R.A.; Berber, E.; Kioi, M.; Padilla-Nash, H.; Damian, M.; Cheung, P.; Kusumoto, R.; Kawahara, T.L.; Barrett, J.C.; et al. SIRT6 Is a Histone H3 Lysine 9 Deacetylase That Modulates Telomeric Chromatin. Nature 2008, 452, 492–496. [Google Scholar] [CrossRef]
  119. Yang, B.; Zwaans, B.M.; Eckersdorff, M.; Lombard, D.B. The Sirtuin SIRT6 Deacetylates H3 K56Ac in Vivo to Promote Genomic Stability. Cell Cycle 2009, 8, 2662–2663. [Google Scholar] [CrossRef]
  120. Tang, M.; Xu, W.; Wang, Q.; Xiao, W.; Xu, R. Potential of DNMT and Its Epigenetic Regulation for Lung Cancer Therapy. Curr. Genomics 2009, 10, 336–352. [Google Scholar] [CrossRef]
  121. Klose, R.J.; Bird, A.P. Genomic DNA Methylation: The Mark and Its Mediators. Trends Biochem. Sci. 2006, 31, 89–97. [Google Scholar] [CrossRef]
  122. Tahiliani, M.; Koh, K.P.; Shen, Y.; Pastor, W.A.; Bandukwala, H.; Brudno, Y.; Agarwal, S.; Iyer, L.M.; Liu, D.R.; Aravind, L.; et al. Conversion of 5-Methylcytosine to 5-Hydroxymethylcytosine in Mammalian DNA by MLL Partner TET1. Science 2009, 324, 930–935. [Google Scholar] [CrossRef]
  123. Brueckner, B.; Garcia Boy, R.; Siedlecki, P.; Musch, T.; Kliem, H.C.; Zielenkiewicz, P.; Suhai, S.; Wiessler, M.; Lyko, F. Epigenetic Reactivation of Tumor Suppressor Genes by a Novel Small-Molecule Inhibitor of Human DNA Methyltransferases. Cancer Res. 2005, 65, 6305–6311. [Google Scholar] [CrossRef] [PubMed]
  124. Stresemann, C.; Brueckner, B.; Musch, T.; Stopper, H.; Lyko, F. Functional Diversity of DNA Methyltransferase Inhibitors in Human Cancer Cell Lines. Cancer Res. 2006, 66, 2794–2800. [Google Scholar] [CrossRef] [PubMed]
  125. Oh, Y.S.; Jeong, S.G.; Cho, G.W. Anti-Senescence Effects of DNA Methyltransferase Inhibitor RG108 in Human Bone Marrow Mesenchymal Stromal Cells. Biotechnol. Appl. Biochem. 2015, 62, 583–590. [Google Scholar] [CrossRef] [PubMed]
  126. Oh, Y.S.; Kim, S.H.; Cho, G.W. Functional Restoration of Amyotrophic Lateral Sclerosis Patient-Derived Mesenchymal Stromal Cells Through Inhibition of DNA Methyltransferase. Cell Mol. Neurobiol. 2016, 36, 613–620. [Google Scholar] [CrossRef]
  127. Li, Q.; Zhai, Y.; Man, X.; Zhang, S.; An, X. Inhibition of DNA Methyltransferase by RG108 Promotes Pluripotency-Related Character of Porcine Bone Marrow Mesenchymal Stem Cells. Cell Reprogram 2020, 22, 82–89. [Google Scholar] [CrossRef]
  128. Roato, I.; Baima, G.; Orrico, C.; Mosca Balma, A.; Alotto, D.; Romano, F.; Ferracini, R.; Aimetti, M.; Mussano, F. Senescent Markers Expressed by Periodontal Ligament-Derived Stem Cells (PDLSCs) Harvested from Patients with Periodontitis Can Be Rejuvenated by RG108. Biomedicines 2023, 11, 2535. [Google Scholar] [CrossRef]
  129. Campisi, J.; Kapahi, P.; Lithgow, G.J.; Melov, S.; Newman, J.C.; Verdin, E. From Discoveries in Ageing Research to Therapeutics for Healthy Ageing. Nature 2019, 571, 183–192. [Google Scholar] [CrossRef]
  130. 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. 2021, 22, 75–95. [Google Scholar] [CrossRef]
  131. 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, 644–658. [Google Scholar] [CrossRef] [PubMed]
  132. Hockenbery, D.; Nunez, G.; Milliman, C.; Schreiber, R.D.; Korsmeyer, S.J. Bcl-2 Is an Inner Mitochondrial Membrane Protein That Blocks Programmed Cell Death. Nature 1990, 348, 334–336. [Google Scholar] [CrossRef] [PubMed]
  133. Yosef, R.; Pilpel, N.; Tokarsky-Amiel, R.; Biran, A.; Ovadya, Y.; Cohen, S.; Vadai, E.; Dassa, L.; Shahar, E.; Condiotti, R.; et al. Directed Elimination of Senescent Cells by Inhibition of BCL-W and BCL-XL. Nat. Commun. 2016, 7, 11190. [Google Scholar] [CrossRef] [PubMed]
  134. Tse, C.; Shoemaker, A.R.; Adickes, J.; Anderson, M.G.; Chen, J.; Jin, S.; Johnson, E.F.; Marsh, K.C.; Mitten, M.J.; Nimmer, P.; et al. ABT-263: A Potent and Orally Bioavailable Bcl-2 Family Inhibitor. Cancer Res. 2008, 68, 3421–3428. [Google Scholar] [CrossRef] [PubMed]
  135. 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]
  136. Kirkland, J.L.; Tchkonia, T. Senolytic Drugs: From Discovery to Translation. J. Intern. Med. 2020, 288, 518–536. [Google Scholar] [CrossRef]
  137. 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, 147–157. [Google Scholar] [CrossRef]
  138. Zhu, Y.; Doornebal, E.J.; Pirtskhalava, T.; Giorgadze, N.; Wentworth, M.; Fuhrmann-Stroissnigg, H.; Niedernhofer, L.J.; Robbins, P.D.; Tchkonia, T.; Kirkland, J.L. New Agents That Target Senescent Cells: The Flavone, Fisetin, and the BCL-X(L) Inhibitors, A1331852 and A1155463. Aging 2017, 9, 955–963. [Google Scholar] [CrossRef]
  139. Mullen, M.; Nelson, A.L.; Goff, A.; Billings, J.; Kloser, H.; Huard, C.; Mitchell, J.; Hambright, W.S.; Ravuri, S.; Huard, J. Fisetin Attenuates Cellular Senescence Accumulation During Culture Expansion of Human Adipose-Derived Stem Cells. Stem Cells 2023, 41, 698–710. [Google Scholar] [CrossRef]
  140. Rufini, A.; Tucci, P.; Celardo, I.; Melino, G. Senescence and Aging: The Critical Roles of P53. Oncogene 2013, 32, 5129–5143. [Google Scholar] [CrossRef]
  141. Cherif, H.; Bisson, D.G.; Mannarino, M.; Rabau, O.; Ouellet, J.A.; Haglund, L. Senotherapeutic Drugs for Human Intervertebral Disc Degeneration and Low Back Pain. eLife 2020, 9, e54693. [Google Scholar] [CrossRef] [PubMed]
  142. Li, M.; Chen, D.; Shiloh, A.; Luo, J.; Nikolaev, A.Y.; Qin, J.; Gu, W. Deubiquitination of P53 by HAUSP Is an Important Pathway for P53 Stabilization. Nature 2002, 416, 648–653. [Google Scholar] [CrossRef] [PubMed]
  143. He, Y.; Li, W.; Lv, D.; Zhang, X.; Zhang, X.; Ortiz, Y.T.; Budamagunta, V.; Campisi, J.; Zheng, G.; Zhou, D. Inhibition of USP7 Activity Selectively Eliminates Senescent Cells in Part via Restoration of P53 Activity. Aging Cell 2020, 19, e13117. [Google Scholar] [CrossRef] [PubMed]
  144. Liu, Z.; Hu, M.; Yang, Y.; Du, C.; Zhou, H.; Liu, C.; Chen, Y.; Fan, L.; Ma, H.; Gong, Y.; et al. An Overview of PROTACs: A Promising Drug Discovery Paradigm. Mol. Biomed. 2022, 3, 46. [Google Scholar] [CrossRef]
  145. 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]
  146. Eggenhofer, E.; Hoogduijn, M.J. Mesenchymal Stem Cell-Educated Macrophages. Transplant. Res. 2012, 1, 12. [Google Scholar] [CrossRef] [PubMed]
  147. Parekkadan, B.; Milwid, J.M. Mesenchymal Stem Cells as Therapeutics. Annu. Rev. Biomed. Eng. 2010, 12, 87–117. [Google Scholar] [CrossRef]
  148. Toma, C.; Pittenger, M.F.; Cahill, K.S.; Byrne, B.J.; Kessler, P.D. Human Mesenchymal Stem Cells Differentiate to a Cardiomyocyte Phenotype in the Adult Murine Heart. Circulation 2002, 105, 93–98. [Google Scholar] [CrossRef]
  149. Xu, M.; Pirtskhalava, T.; Farr, J.N.; Weigand, B.M.; Palmer, A.K.; Weivoda, M.M.; Inman, C.L.; Ogrodnik, M.B.; Hachfeld, C.M.; Fraser, D.G.; et al. Senolytics Improve Physical Function and Increase Lifespan in Old Age. Nat. Med. 2018, 24, 1246–1256. [Google Scholar] [CrossRef]
  150. Alimbetov, D.; Davis, T.; Brook, A.J.; Cox, L.S.; Faragher, R.G.; 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, 305–315. [Google Scholar] [CrossRef]
  151. 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]
  152. Niklander, S.; Bandaru, D.; Lambert, D.W.; Hunter, K.D. ROCK Inhibition Modulates the Senescence-Associated Secretory Phenotype (SASP) in Oral Keratinocytes. FEBS Open Bio 2020, 10, 2740–2749. [Google Scholar] [CrossRef] [PubMed]
  153. Osorio-Llanes, E.; Villamizar-Villamizar, W.; Ospino Guerra, M.C.; Díaz-Ariza, L.A.; Castiblanco-Arroyave, S.C.; Medrano, L.; Mengual, D.; Belón, R.; Castellar-López, J.; Sepúlveda, Y.; et al. Effects of Metformin on Ischemia/Reperfusion Injury: New Evidence and Mechanisms. Pharmaceuticals 2023, 16, 1121. [Google Scholar] [CrossRef] [PubMed]
  154. Sadeghi, M.; Khosrawi, S.; Heshmat-Ghahdarijani, K.; Gheisari, Y.; Roohafza, H.; Mansoorian, M.; Hoseini, S.G. Effect of Melatonin on Heart Failure: Design for a Double-Blinded Randomized Clinical Trial. ESC Heart Fail. 2020, 7, 3142–3150. [Google Scholar] [CrossRef]
  155. El Oirdi, M. Harnessing the Power of Polyphenols: A New Frontier in Disease Prevention and Therapy. Pharmaceuticals 2024, 17, 692. [Google Scholar] [CrossRef]
  156. Toguchi, A.; Noguchi, N.; Kanno, T.; Yamada, A. Methylsulfonylmethane Improves Knee Quality of Life in Participants with Mild Knee Pain: A Randomized, Double-Blind, Placebo-Controlled Trial. Nutrients 2023, 15, 2995. [Google Scholar] [CrossRef]
  157. Schneeberger, Y.; Stenzig, J.; Hübner, F.; Schaefer, A.; Reichenspurner, H.; Eschenhagen, T. Pharmacokinetics of the Experimental Non-Nucleosidic DNA Methyl Transferase Inhibitor N-Phthalyl-l-Tryptophan (RG 108) in Rats. Basic. Clin. Pharmacol. Toxicol. 2016, 118, 327–332. [Google Scholar] [CrossRef]
  158. 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]
  159. Ji, J.; Bae, M.; Wong, F.L.; Crespi, C.M.; Yee, L.D.; Sedrak, M.S. A Phase II Randomized Double-Blind Placebo-Controlled Study of Fisetin to Improve Physical Function in Frail Older Breast Cancer Survivors (TROFFi). J. Clin. Oncol. 2024, 42, TPS1645. [Google Scholar] [CrossRef]
  160. Kozlowski, M.; Ladurner, A.G. ATM, MacroH2A.1, and SASP: The Checks and Balances of Cellular Senescence. Mol. Cell 2015, 59, 713–715. [Google Scholar] [CrossRef]
  161. Pellot Ortiz, K.I.; Rechberger, J.S.; Nonnenbroich, L.F.; Daniels, D.J.; Sarkaria, J.N. MDM2 Inhibition in the Treatment of Glioblastoma: From Concept to Clinical Investigation. Biomedicines 2023, 11, 1879. [Google Scholar] [CrossRef] [PubMed]
  162. Li, Z.; Liu, C.; Li, S.; Li, T.; Li, Y.; Wang, N.; Bao, X.; Xue, P.; Liu, S. BMSC-Derived Exosomes Inhibit Dexamethasone-Induced Muscle Atrophy via the MiR-486-5p/FoxO1 Axis. Front. Endocrinol. 2021, 12, 681267. [Google Scholar] [CrossRef] [PubMed]
  163. Shahin, H.; Belcastro, L.; Das, J.; Perdiki Grigoriadi, M.; Saager, R.B.; Steinvall, I.; Sjöberg, F.; Olofsson, P.; Elmasry, M.; El-Serafi, A.T. MicroRNA-155 Mediates Multiple Gene Regulations Pertinent to the Role of Human Adipose-Derived Mesenchymal Stem Cells in Skin Regeneration. Front. Bioeng. Biotechnol. 2024, 12, 1328504. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Features of senescent MSCs. Senescence is associated with different mechanisms, such as morphological alterations, ROS production, DNA damage, mitochondrial dysfunction, telomere shortening, epigenetic modifications, non-coding RNA control, loss of proteostasis, and SASP production, which lead to typical phenotype of senescent MSCs, expressing p16, p21, β-gal, and SASP.
Figure 1. Features of senescent MSCs. Senescence is associated with different mechanisms, such as morphological alterations, ROS production, DNA damage, mitochondrial dysfunction, telomere shortening, epigenetic modifications, non-coding RNA control, loss of proteostasis, and SASP production, which lead to typical phenotype of senescent MSCs, expressing p16, p21, β-gal, and SASP.
Biomedicines 12 02811 g001
Figure 2. Senescent MSCs. The images show PDLSCs isolated from periodontitis patients, cultured in vitro, with a typical spindle-shaped cell body (A) and stained positive for β-gal (B). A confocal analysis showing nuclei (blue) and the co-expression of p21 (green) and OCT4 (red); magnification: 63× (C).
Figure 2. Senescent MSCs. The images show PDLSCs isolated from periodontitis patients, cultured in vitro, with a typical spindle-shaped cell body (A) and stained positive for β-gal (B). A confocal analysis showing nuclei (blue) and the co-expression of p21 (green) and OCT4 (red); magnification: 63× (C).
Biomedicines 12 02811 g002
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

Roato, I.; Visca, M.; Mussano, F. Suppressing the Aging Phenotype of Mesenchymal Stromal Cells: Are We Ready for Clinical Translation? Biomedicines 2024, 12, 2811. https://doi.org/10.3390/biomedicines12122811

AMA Style

Roato I, Visca M, Mussano F. Suppressing the Aging Phenotype of Mesenchymal Stromal Cells: Are We Ready for Clinical Translation? Biomedicines. 2024; 12(12):2811. https://doi.org/10.3390/biomedicines12122811

Chicago/Turabian Style

Roato, Ilaria, Matteo Visca, and Federico Mussano. 2024. "Suppressing the Aging Phenotype of Mesenchymal Stromal Cells: Are We Ready for Clinical Translation?" Biomedicines 12, no. 12: 2811. https://doi.org/10.3390/biomedicines12122811

APA Style

Roato, I., Visca, M., & Mussano, F. (2024). Suppressing the Aging Phenotype of Mesenchymal Stromal Cells: Are We Ready for Clinical Translation? Biomedicines, 12(12), 2811. https://doi.org/10.3390/biomedicines12122811

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