Abstract
Recent animal and clinical studies report promising results for the therapeutic utilization of stem cells in regenerative medicine. Mesenchymal stem cells (MSCs), with their pluripotent nature, have advantages over embryonic stem cells in terms of their availability and feasibility. However, their proliferative activity is destined to slow by replicative senescence, and the limited proliferative potential of MSCs not only hinders the preparation of sufficient cells for in vivo application, but also draws a limitation on their potential for differentiation. This calls for the development of safe and efficient means to increase the proliferative as well as differentiation potential of MSCs. Recent advances have led to a better understanding of the underlying mechanisms and significance of cellular senescence, facilitating ways to manipulate the replicative lifespan of a variety of primary cells, including MSCs. This paper introduces a class of proteins that function as senescence suppressors. Like tumor suppressors, these proteins are lost in senescence, while their forced expression delays the onset of senescence. Moreover, treatments that increase the expression or the activity of senescence suppressors, therefore, cause expansion of the replicative and differentiation potential of MSCs. The nature of the activities and putative underlying mechanisms of the senescence suppressors will be discussed to facilitate their evaluation.
Keywords: Mesenchymal stem cell (MSC), Stem cell therapy, Replicative senescence, Differentiation, Replicative lifespan extension, ECM
Introduction
Mesenchymal stem cells (or marrow stem cells; MSCs) are able to differentiate to become osteoblasts, chondrocytes, adipocytes, myoblasts, and neurons. This pluripotency makes them excellent resources for cell replacement therapy or stem cell therapy, an approach to regenerate the functions of damaged tissues or organs by supplying stem cells. MSCs have advantage over embryonic stem cells in many aspects, such as availability and feasibility and low risk of cancer development. However, the low cancer risk, by nature, also raises the problem of a limitation to the number of available cells. The in vitro life span of stem cells isolated from human bone marrow (BMSCs) reportedly ranges from approximately 20–40 doublings, with Banfi et al. [1] reporting 22–23 doublings while Bruder et al. [2] reported 38 doublings, which is possibly dependent on many variables, including donor age and the efficiency of MSC preparation. In fact, current MSC preparations used in therapies are mixtures of heterogeneous stromal cells that include a subpopulation of stem cells. In order to be useful for therapy, MSCs should be expanded in vitro to a number sufficient for transplantation. In a consensus, at least 1 × 106/kg or 108 total cells are required to be transplanted for a successful MSC therapy [3]. This number of cells is reached after 27 doublings from a single cell, which consumes a large portion of the proliferative potential. Even if the population does not reach the end of its life span, i.e., replicative senescence, many cells in the late passage population show slow proliferation and express a senescence phenotype. Transplantation of such cells would not produce good results and may even be harmful. Senescence also imposes a limit to the stemness capacity of MSCs in differentiation and homing. As cells approach the end of their life span, they tend to lose osteogenic potential and gain in adipogenesis, the phenomenon termed “osteogenic to adipogenic shift” [1, 4–7]. Late passage MSCs express smaller amounts of homing receptors [8, 9] dropping down the efficacy of therapy.
Replicative senescence becomes more problematic when cell therapy is considered for aged individuals for whom the practice would be in greater demand. BMSCs from older human donors have a lower growth rate and capacity as well as decreased potential for differentiation [10–13]. The decreased proliferation is due to an increase in gate-keeping tumor suppressor activity and likely functions in reducing the incidence of cancer in aging tissues, but it also causes a decrease in regenerative capacity [14, 15]. This would certainly impose a limitation on the applicability of stem cell therapy in aged donors.
Replicative senescence may simply be viewed as an outcome of the chronic activation of DNA damage response triggered from short and deformed telomeres. p53-Rb and p53-Rb2 (probably functions more importantly than p53-Rb in human MSC), gate-keeping tumor suppressor pathways, play central roles in senescence imposition by linking damage sensing and execution of cell cycle arrest [17–19]. DNA damage is exacerbated by reactive oxygen species (ROS), and ROS-mediated single-strand breaks in telomeres have been shown to cause earlier imposition of senescence [16]. ROS are produced at a high level by damaged mitochondria, causing even higher levels of damage [20]. Thus, the cellular ROS level increases exponentially during continued cell cultivation in vitro [21]. Therefore, attenuating the accumulation of ROS or damage to cellular components can be an excellent strategy to slow down the cellular progression to replicative senescence. Indeed, cultivation of human fibroblasts and MSCs under low oxygen tension results in decreased levels of DNA damage and an extended replicative lifespan [22–24].
A number of proteins that function in damage response or defense against oxidative stress are involved in modulation of the lifespan of human primary cells. Some of these proteins, when overexpressed in human MSCs or human fibroblasts, induce substantial expansion of the proliferative potential and delay replicative senescence to an extent worthy of practical consideration. Interestingly, they are commonly lost during the progression to senescence. In this review, these proteins are termed as “senescence suppressors” and are introduced as promising targets for manipulation to enhance the proliferation and differentiation capacities of MSCs.
Senescence suppressor proteins
Tumor suppressor proteins such as p53 and Rb are frequently lost in cancer cells. Their forced expression puts a strong brake on the proliferation of cancer cells by causing either senescence or apoptosis. Therefore, tumor suppressors are characterized as proteins whose expression or activity needs to be attenuated for a cell to become cancerous [21]. An analogy can be found for cellular senescence. Certain proteins are highly expressed in early passage primary cells, but their expression declines during continued proliferation and is lost at senescence. Furthermore, their forced expression delays senescence and extends replicative lifespan, while the attenuated expression or activity leads to the development of the senescence phenotype. Here, these are analogously termed senescence suppressors, despite a difference in the etiology. Unlike tumor suppressors, the change in the levels of senescence suppressors is not known to be caused by loss of function mutations, and therefore, cellular senescence does not arise as an outcome of genetic selection against them. Importantly, the forced expression of a list of senescence suppressors has been shown to allow MSCs to maintain the phenotype of actively proliferating cells and the property of stemness.
Nrf2: master regulator of antioxidative defenses
Nf-E2 related factor 2 (Nrf2) is a transcription factor that, in response to oxidative stress, activates various antioxidant responsive element (ARE)-dependent genes encoding cellular redox regulators such as NAD(P)H quinone oxidoreductase 1, glutathione S-transferase, and peroxiredoxins [26]. It also activates the expression of multiple subunits of the 20S proteasome [27, 28]. Nrf2 has been shown to critically control stem cell survival under oxidative damage, which is likely mediated through cellular redox and proteome homeostasis by the aforementioned proteins [29–31].
The activity of Nrf2 declines during replicative senescence and aging [27, 32], and this has been proposed to contribute to the age-associated decline of antioxidative capacity. Furthermore, Nrf2 silencing causes premature senescence in human fibroblasts [27], while its activation induces lifespan extension of human fibroblasts and BMSCs [27, 31]. Considering that BMSCs are highly sensitive to oxidative stress [30], Nrf2 activation (through certain dietary phytochemicals and synthetic chemicals [33–35]) may be an excellent strategy to extend the lifespan of BMSCs in vitro.
20S proteasome: protein homeostasis
The 26S proteasome complex removes extra or damaged proteins. In cells at late passage or those isolated from aged donors, highly oxidized proteins are abundantly present and frequently cross-linked to each other, forming aggregates that are resistant to proteasome-mediated proteolysis [36]. Hydrophobic surface patches appear to be a major recognition motif in the substrates of proteasomes, and their oxidative modification is thought to help the proteins escape proteolysis [37, 38]. Such a change in susceptibility to proteolysis has been suggested to contribute to senescence and various age-related neural disorders such as Parkinson’s disease [39, 40]. Proteasome dysfunction or reduced activity of the 20S proteasome, the active component of 26S proteasome complex, has been observed in cells at late passage or from aged donors [31, 40–42]. Meanwhile, inhibition of proteasome activity using antagonists, such as MG132, induced senescence in early passage MSCs [41, 43].
While proteasome antagonists have attracted attention for their antitumoral effects [44], proteasome agonists may be the subjects of further focus for their anti-senescence effects. A triterpenoid, 18α-glycyrrhetinic acid (18α-GA) increased the level of the 20S proteasome subunit and its activity probably through the activation of Nrf2 [27, 45]. The treatment of human fibroblasts with 18α-GA provided cytoprotection against oxidative stress in a manner dependent on proteasome activity. It also caused substantial extension of the replicative lifespan in human fibroblasts and BMSCs [27, 31], and helped neuronal cells maintain expression of stemness markers [31].
SIRT1: promotion of cell survival and differentiation in response to genomic stress
SIRT1, an NAD+-dependent protein deacetylase, is a major mediator in the effect of calorie restriction and plays a preventive role in many aging-associated disorders [46, 47]. It also exerts positive effects on lifespan and differentiation of various stem cells. SIRT1 expression is reduced in senescent stem cells [48] and fibroblasts [49–52]. In addition, its knockdown suppresses cell growth and accelerates senescence of MSCs, while its overexpression delays the onset of senescence and the loss of differentiation capacity [51].
The effects of SIRT1 on the cellular lifespan appear to manifest through multiple different ways. First, it modulates p53 activity. Deacetylation by SIRT1 antagonizes p53 transcriptional activity [53]. It also induces p53 translocation to mitochondria [54], thereby attenuating the biological response to DNA damage [55, 56]. An outcome of this may be the attenuation of stress-induced premature senescence. SIRT1 activation prevented premature senescence induced by oxidative stress [57], oncogene expression [53, 58], or PML upregulation [53]. SIRT1 may also extend cellular lifespan by suppressing triggering of the growth-arrest signal derived from telomere shortening or dysfunction. SIRT1 overexpression attenuated telomere shortening, while its deficiency caused increased chromosome fragility at telomeres and telomere dysfunction in MEFs [59]. Moreover, it upregulates the hTERT gene, the catalytic subunit of telomerase (possibly through c-Myc activation [60]) [61]. SIRT1 is also involved in the management of oxidative stress. It activates FOXO3a, which upregulates the expression of antioxidant proteins such as MnSOD, catalase, thioredoxin 2, thioredoxin reductase 2, and peroxiredoxins [62], and has been shown to protect against cigarette smoke-induced oxidative stress in a FOXO3-dependent manner [63]. In addition, SIRT1 is involved in basal level autophagy. Through deacetylation, it activates key molecules in autophagosome formation such as Atg5, Atg7, and Atg8 [64]. It was shown that calorie restriction-mediated SIRT1 activation in the kidney of aged mice attenuates renal dysfunction possibly through the activation of autophagic disposal of damaged mitochondria [65]. These suggest that SIRT1 activation can promote stem cell longevity by facilitating better maintenance of mitochondria quality through augmentation of basal level autophagy.
Intriguingly, SIRT1 also imposes a limit to cellular lifespan. This appears to be a response to chronic genotoxic stress. SIRT1-deficient MEFs showed increased replicative lifespan and proliferative capacity under chronic sub-lethal oxidative stress [66]. In these cells, the level of p19ARF, which stabilizes p53, was low, and its increase during serial passage was attenuated, resulting in a reduction in p53 protein levels. SIRT1 antagonizes REGγ, a proteasome activator, which promotes degradation of p19ARF along with other cyclin-dependent kinase inhibitors (CDKIs), p21WAF1 and p16INK4a [67]. Therefore, the absence of SIRT1 might cause reduction in the levels of CDKI and p53. Together, these findings suggest that, in chronic sub-lethal genotoxic stress such as that generated during serial cultivation under high oxygen conditions, SIRT1 potentiates p53-growth-inhibitory pathway and causes early onset of cell cycle arrest and senescence. These contradictory findings on the effect of SIRT1 on cellular lifespan suggest that cells have evolved diverse responses to cope with different levels of genotoxic insult.
SIRT1 appears to modulate stem cell differentiation primarily by antagonizing p53 suppression of the Nanog gene, which functions in self-renewal and maintenance of a differentiated state [54, 68]. In addition, the direction of differentiation may also be affected by SIRT1. Under oxidative stress, SIRT1 deacetylates histones at the promoter region of Mash1, which is responsible for the activation of a neuron-specific transcriptome [69], thereby switching neural differentiation to gliogenesis [70, 71]. Whether SIRT1 exerts this kind of regulatory effect on other types of differentiation is not known.
NAMPT and nicotinamide: regulators of NAD redox and SIRT1 activity
Nicotinamide phosphoribosyltransferase (Nampt/PBEF/Visfatin) converts nicotinamide (NAM) to nicotinamide adenine mononucleotide (NMN), in the rate-limiting step of the salvage pathway for NAD+ biosynthesis [72]. It thereby provides cells the key coenzyme for metabolism. The concomitant increase in the NAD+/NADH ratio also affects the activity of several key enzymes including SIRT1 [73, 74]. The level of Nampt protein in smooth muscle cells decreases after continued proliferation, and antagonist-mediated downregulation of its activity causes premature senescence [75]. Meanwhile, transduction of the Nampt gene lengthened the lifespan of smooth muscle cells (extension of 34–71 % depending on the cell strains) and fibroblasts. This effect is likely to be mediated by SIRT1 activation, since the expression of the dominant-negative form of SIRT1 abrogated the effect of Nampt overexpression [75]. Furthermore, the lifespan-extending effect of SIRT1 overexpression was significantly potentiated by Nampt overexpression [76].
Meanwhile, prolonged administration of 5-mM NAM effectively extends the replicative lifespan of human fibroblasts and keratinocytes [77]. The lifespan was extended by 60 %, while the increase in ROS was substantially delayed. In the NAM-treated cells, mitochondrial autophagy was augmented while the level of mitochondrial ROS decreased. These changes were reproduced by the treatment of SIRT1 activators, and abrogated by co-treatment with FK866 [77, 78], an antagonist of Nampt, indicating that at least a part of the effect of NAM is mediated by an enhancement of cellular levels of NAD+ or the ratio of NAD+/NADH, and, thereby, activation of SIRT1. Meanwhile, in a recent study, NAM and its methyl derivative, methylnicotinamide extended the lifespan of Caenorhabditis elegans even in the absence of SIR-2.1 [79] suggesting a SIRT1-independent effect of NAM. NAM has been reported to have strong antioxidant activity [80], and whether this contributes to the prolongevity effect of NAM that needs to be determined.
Klotho: attenuation of senescence-associated inflammation
A defect in the Klotho gene causes premature aging [81], and the anti-aging effect of the Klotho gene has been demonstrated in mice [82]. α-Klotho protein, an obligatory co-receptor for FGF23, a hormone involved in Ca2+ and phosphate homeostasis [82, 83], was shown to interact with retinoic acid-inducible gene-I (RIG-1) protein [84] and inhibit RIG-1-mediated expression of IL-6 and IL-8, which are key mediators of inflammation and factors involved in the senescence-associated secretory phenotype (SASP) [85]. Expression of Klotho declines in senescent cells while that of RIG-1 increases inversely, possibly contributing to the expression of SASP and senescence-associated inflammation in senescent cells [84]. Extension of this line of investigation on Klotho may help clarify the possible link between replicative lifespan and SASP. Meanwhile, knocking down Klotho expression caused premature senescence in a p53-dependent manner, suggesting that Klotho normally suppresses senescence by interfering with the damage accumulation or the p53-damage response pathway [86].
HO-1, BVR-A, and Nrf2: primary antioxidative defense network
Heme oxygenase-1 degrades heme to produce biliverdin and carbon monoxide [87]. It upregulates the expression of the key anti-inflammatory modulators IL-10 and IL-1Ra (IL-1R antagonist) [88], and thereby, plays an anti-inflammatory role. For instance, HO-1 protected osteoarthritis chondrocytes, synoviocytes, and osteoblasts against damage caused by cytokine-induced inflammation and apoptosis [89, 90]. HO-1 also exerts strong positive effects on the osteogenesis. Its overexpression potentiated BMSCs and periodontal ligament cells for osteogenic differentiation while suppressing adipogenesis [91]. It also prevented the expression of the senescence phenotype as well as the reduction of bone formation in osteoblasts treated by IL-1β, a potent pro-inflammatory molecule [92]. Therefore, HO-1 manipulation would certainly provide a means to block the osteogenic to adipogenic shift that takes place during MSC expansion in vitro.
Bilirubin is an important intrinsic antioxidant that quenches inflammation-induced free radicals by being oxidized to biliverdin. Biliverdin reductase A (BVR-A) converts biliverdin to bilirubin [93]. In addition, it facilitates the expression of HO-1 by forming a transcription factor along with hematin [94]. Therefore, BVR-A constitutes a strong cellular antioxidative defense along with HO-1. The upregulation of the HO-1/BVR-A antioxidant system has been identified as one of the early responses to counteract oxidative stress in the brain and proposed as a useful mechanism to counteract Alzheimer disease-induced oxidative damage [95]. The activity of BVR-A declines in aging and its upregulation upon oxidative stress is impaired in senescence [96, 97], possibly due to the decrease in Nrf2 activity, which upregulates the expression HO-1 [98].
PPARγ and δ: transcriptional activation of antioxidative and anti-inflammatory functions
Peroxisome proliferator-activated receptors (PPAR)γ and δ, members of the PPAR transcription factor family, have antioxidative and anti-inflammatory functions through the expression of a battery of downstream antioxidative proteins [99, 100]. PPARγ induces the expression of Cu/Zn-SOD [101] and Klotho [102]. It also inhibits inflammatory gene expression by directly or indirectly interfering with the pro-inflammatory transcription factor NF-kB [103]. Expression of PPARγ decreases during aging of rats [104], while its activation rescues the functions of tissues damaged by oxidation or inflammation [104] and reduces the senescence phenotype in fibroblasts in inflammatory skin [105]. Meanwhile, PPARδ induces the expression of glutathione peroxidase 1 (GPx1), thioredoxin 1 (Trx1), MnSOD, and HO-1 [106]. It also suppresses the inflammation of vascular smooth muscle cells (VSMCs) through upregulation of TGF-β1, an anti-inflammatory mediator [107, 108]. PPAR δ activation attenuates ROS generation and prevents premature senescence of VSMCs [109–111]. Overall, activation of the PPAR transcription factors by agonists such as thiazolidinedione compounds (for PPARγ) [112, 113] and GW501516 (for PPARδ) [109] may be a strategy worthy of trial for functional expansion of MSCs in vitro.
Most of the antioxidative genes induced by PPARγ are also activated by Nrf2 [114]. Further, Nrf2 modulates PPARγ expression by binding to a putative ARE in its promoter [115]. This suggests a possible link between the pathways of these transcription factors for cellular antioxidant function.
Prion protein: enhancing cellular antioxidation capacity
Cellular prion protein, PrPC, is expressed in multipotent neural precursors and plays roles in neurogenesis [116], while certain variants, collectively designated PrPSC, are culprits of Creutzfeldt-Jakob disease (CJD) and other transmissible spongiform encephalopathies [117]. PrPC expression decreases in MSCs during expansion in vitro, and its knockdown causes a significant reduction in proliferation as well as differentiation [118]. Meanwhile, maintenance of its expression resulted in an extended lifespan [108]. The antioxidant activity of PrPC has been recognized [119–121]. PrPC increases cellular SOD activity through increased incorporation of copper into CuZn-SOD, or the formation of a complex with copper that mimics cellular SOD activity at the cell surface [122, 123]. It also causes a substantial increase in the activity of glutathione reductase and, thereby, the level of glutathione [123], although the underlying mechanism is not known. A chemical named 3/689 has been shown to cause sustained expression of PrPC protein [118], while inhibiting PrPSC accumulation during continued cell passage in human MSC [124]. In addition, this treatment extended the replicative lifespan of hMSCs (near 25 % increase in PD number) and increased their engraftment efficiency after bone marrow transplant in a manner dependent on PrPC expression [118]. An increase in SOD2 activity was observed in the late passage hMSC treated with the chemical [118].
Statins: senescence suppressor-like effectors modulating cell cycle and cellular oxidative level
Statins (HMG-CoA reductase inhibitors) such as atorvastatin and mevastatin were shown to inhibit the onset of senescence as well as apoptosis in endothelial progenitor cells (EPCs) [125]. Atorvastatin treatment of human EPCs was found to activate the PI3-kinase/Akt pathway, which is known to regulate the senescence of endothelial cells [126] and also to mediate the angiogenic effect of VEGF by conferring endothelial cell survival and neovascularization [127, 128]. PI3K/Akt activation-mediated suppression of FOXO4 and its downstream gene, p27Kip1, a CDK inhibitor, has been suggested to be involved in this effect [129–132]. Statins may also delay senescence through antioxidative effects. Atorvastatin downregulates the expression of NOX1 and p22phox, which are key subunits of NAD(P)H oxidase, a predominant producer of superoxides in the endothelium [133, 134], and upregulates the expression of catalase and SIRT1 in rat aortic vascular smooth muscle cells [135] and PPAR-α/β/δ/γ in the rat heart [136]. Furthermore, atorvastatin has been shown to delay the onset of replicative senescence of endothelial cells caused by homocysteine-induced inactivation of telomerase [137]. Meanwhile, simvastatin has been reported to induce melanoma cell senescence, possibly by increasing oxidative stress [138]. This finding suggest that certain statins may have pro-oxidative effects.
Extracellular matrix (ECM): cell adhesion signaling
Culture substratum affects the growth and differentiation of MSCs. The proliferative capacity of human BMSCs increased by near eight doublings when cultured on a plate coated with ECM-gelR (Sigma-Aldrich), a commercially available mixture of basement membrane proteins [139]. ECM-gelR plate also enhanced differentiation and attenuated the adipogenic switch in late passage MSCs. Individual ECM components, fibronectin, laminin-1, laminin-5, and collagen IV were also effective, albeit to smaller extents. Meanwhile, treatment with antibodies against α6 integrin/CD49f blocked the lifespan-extending effect of ECM-gelR [139]. This indicates an outside-in signaling generated by the ECM-integrin interaction that positively affects the proliferation of MSCs. No detail of the underlying mechanism has yet been determined, although an increase in telomerase activity has been reported [140]. Meanwhile, a senescence-promoting role for integrin β4 has also been proposed in vascular endothelial cells [141]. Knocking down integrin β4, which is highly expressed in senescent HUVECs, suppressed ROS increase and senescence [141, 142].
Cellular rejuvenation has been made by utilizing the ECM of the cells from young animals. In one study, the cell-free ECM was generated by cultivating BMSCs from 3-month-old mice and removing them by treatment with Triton X-100 and NH4OH. This young ECM, unlike the one generated from BMSCs of 18-month-old mice (old ECM), supported self-renewal and bone formation of BMSCs isolated from old mice [143]. Importantly, cultivation on the young ECM dramatically attenuated the increase of ROS during continued passage. The young ECM was found to contain higher amounts of collagen and lower amounts of phosphate. Whether these affect the proliferation of BMSC is not known, but their ratio at least appears to affect the osteogenic differentiation [143]. In addition, calcium phosphate particles impair the viability and proliferation of osteoblast progenitor cells [144]. ECM of early passage cells may work similarly. Senescent fibroblasts, when plated on the ECM of early passage fibroblasts, resumed proliferation and gained young cell properties (lower level ROS and longer telomeres) for several doublings [145]. These findings, together, suggest that the way cells attach to ECM not only determines their capacities for proliferation and differentiation, but also modulates the growth-inhibitory machinery that is already in force.
The change in the levels of senescence suppressors
Protein homeostasis is important for cell function and physiology. Cells change their fates through subtle alteration in the level of certain proteins. For example, both the upregulation and downregulation of brahma-related gene 1 (BRG1), a component of chromatin remodeling complex that controls cell cycle control, apoptosis, and differentiation [146], induces senescence in MSCs [147]. This suggests that, for certain proteins, manipulations that affect their expression or activity may cause unexpected or unwanted effects, and therefore, need to be attempted with caution.
A hallmark of senescence suppressors is the decline in their levels during continued passaging of primary cells. Whether this is the outcome of prolonged oxidative damage or driven by an innate program induced by complex signaling processes during prolonged cell proliferation is unknown. A variety of mechanisms are known to involved in the decrease in the levels of senescence suppressor proteins. For example, the decline in the SIRT1 level is caused by changes at multiple different points during pre- and post-translational processes. The increased level of p53 at late passages attenuates SIRT1 expression by inactivating the SIRT1 promoter [148] and/or upregulating the expression of miR-34a [149] that binds to SIRT1 mRNA and facilitates its degradation [150]. SIRT1 mRNA is also destabilized upon dissociation of RBP HuR from its AU-rich elements [150, 151], which is caused by RBP HuR phosphorylation by Chk2 [152], a member of the DNA damage signaling pathway. Meanwhile, the SIRT1 protein level decreases in EPCs because of cleavage by cathepsins B, S, and L [153] which are released from lysosomes upon loss of membrane integrity caused by ROS in senescence [154]. Finally, oxidative stress-induced carbonylation of cysteine residues causes proteasome-mediated degradation of SIRT1 protein [155]. Some conclusions can be derived from these findings: the levels of senescence suppressors may be modulated by multiple mechanisms, and most of these mechanisms are strongly associated with oxidative damage and damage signaling. (In the case with SIRT1, the association of oxidative damage and its signaling has not been demonstrated only in the case with miR-34a). These indications may be highly relevant for safety concerning prolonged modulation of senescence suppressor levels in practical applications. If such decreases are simply a passive outcome of oxidative damage, the reversal of their expression can be safely adopted as a strategy for MSC therapy. However, more caution is required if an innate program drives the change in senescence suppressor levels. Therefore, the mechanisms underlying the decreases in senescence suppressor levels warrant further in-depth investigation.
Concluding remarks
A class of proteins which can be classified as senescence suppressors by definition, are not included in this review. Bmi-1 and Wip1 (PPM1D) commonly extends cellular lifespan by interfering with the damage-response pathways. Bmi-1 suppresses the expression of p16INK4a and p19Arf, the two major inhibitors of cell cycle progression [156–158]. Its overexpression extends the replicative lifespan of primary cells [159–161], but has also been shown to induce immortalization of epithelial cells and MSCs [162, 163] and bears a risk for tumorigenesis [164, 165]. Wip1 abrogates the ATM/p53-DNA-damage checkpoint function [166] and the p38MAPK–p16INKa pathway [167] by dephosphorylating key members in the pathways. Overexpression of Wip1 in chondrocytes and hMSCs attenuated the growth-inhibitory response to oxidative stress (but not the level of ROS itself) [168, 169] and delayed the onset of replicative senescence in MSCs [168]. Its sustained activation, however, has also been suggested to promote tumorigenesis [166]. The primary function of these damage-response pathways is to prevent accumulation of mutations, and therefore, the modulation of the individual components in the pathway would cause cancer development [170]. However, the senescence suppressors introduced here appear to act differently, i.e., reduce the level of oxidative stress either by enhancing the cellular antioxidative capacity or promoting damage removal as summarized in Fig. 1. Cultivation of cells under low oxygen tension has been shown to extend the replicative lifespan of human MSCs and fibroblasts [22, 23, 171], providing a rationale for approaches to reduce the level of oxidative stress as an effective strategy for stem cell expansion. Meanwhile, the cellular glutathione/glutathione disulfide (GSH/GSSG) and cysteine/cystine (Cys/CySS) redox couples control oxidative stress [172]. These can be manipulated to reduce oxidative stress by adding glutathione or cysteine in culture medium. Variation in the GSH/GSSG and Cys/CySS redox, however, also affects cell-signaling pathways [173], and it is not known if the long-term manipulation of these redox couples indeed causes an extension of cellular replicative lifespan.
Finally, the results of a series of studies on telomerase-mediated extension of MSC lifespan have revealed that the differentiation potential of MSCs is tightly associated with their proliferation potential, i.e., once the proliferation potential is secured, multipotency can be maintained [174–176]. This suggests that a manipulation that extends the lifespan of MSCs, once the risk can be ruled out for cell immortalization, might be adopted without much concern for its effect on differentiation. To date, individual senescence suppressors have not been compared in terms of their effect on expanding the replicative capacity of MSCs. Although each of the discussed proteins can be activated to obtain certain levels of effect, some are predicted to be more potent than others. Nrf2 acts by inducing a family of antioxidant enzymes and transcription factors targeting antioxidant genes. It also increases proteasome activity, thereby exerting a strong effect on both prevention and removal of damage. Importantly, there are chemical agonists available for Nrf2 (Table 1). SIRT1 may also function in cellular longevity through multiple pathways. It facilitates induction of antioxidative proteins while suppressing chronic damage signaling. Furthermore, SIRT1 activates autophagy that prevents damage accumulation and helps cells avoid damage-induced senescence [177, 178]. A family of SIRT1 activators have also been examined in vivo [179], although their effects have not been clearly determined because of certain off-target effects [180]. However, they can also be readily utilized for lifespan extension of MSCs in vitro.
Table 1.
Proteins | Effects and mechanisms | Inducers or agonists |
---|---|---|
Nrf2 | Reduction of oxidative stress by inducing antioxidant enzymes | Dimethyl fumarate [181], α-lipoic acid [182, 183], sulforaphane [184], bardoxolone methyl [185] |
Protection from inflammation-caused damage by inducing anti-inflammatory cytokines | ||
Damage removal by inducing 20S proteasome | ||
PrPC | Reduction of oxidative stress by potentiating CuZn-SOD and glutathione reductase | 3/689 [118] |
BVR-A | Reduction of oxidative stress by producing bilirubin and inducing HO-1 | |
HO-1 | Protection from inflammation-caused damage by inducing anti-inflammatory cytokines | Atrial natriuretic peptide [186] |
Forms HO-1/BVR-A antioxidant loop | ||
α-Klotho | Protection from inflammation-caused damage by suppressing pro-inflammatory cytokine expression | Rosiglitazone through PPAR γ [112] |
PPAR γ & PPARδ | Reduction of oxidative stress by inducing antioxidative and anti- inflammatory enzymes | Rosiglitazone [112], Pioglitazone [113] for PPAR γ; GW501516 [109] for PPARδ |
20S proteasome | Damage removal | 18α-glycyrrhetinic acid [27, 45] |
SIRT1 | Reduction of oxidative stress by inducing antioxidant enzymes via FOXO3 activation | SIRT1 activators [179] |
Suppression of damage signaling by inhibiting p53 | ||
Telomere maintenance by hTERT induction | ||
Damage removal through autophagy activation | ||
NAMPT | SIRT1 activation by increasing NAD+/NADH ratio | Nicotinamide works by potentiating Nampt effect, but, also has antioxidative effect |
ECM materials | Increased cell survival signaling and decreased ROS production. | |
Cellular rejuvenation (by young cell ECM) | ||
Statins | Reduction of oxidative stress by downregulating NAD(P)H oxidase and upregulating catalase | Mevastatin [129] atorvastatin [135] but, simvastatin has pro-senescence effect [138] |
Suppression of damage signaling by inhibiting CDK inhibitor | ||
Upregulation of SIRT1 and PPAR α/β/δ/γ | ||
Bmi-1a | Suppression of tumor suppressor function: repression of INK4a-Arf locus | |
Wip1a | Suppression of DNA damage signaling | |
hTERTa | Damage reduction by telomere maintenance (not-senescence supprossor) |
a Bm-1, Wip1, and hTERT are not suitable for in vitro expansion of MSC for stem cell therapy since their activation causes cell immortalization which can lead to tumorigenesis in vivo
Acknowledgments
This work is supported by National Research Foundation of Korea (NRF-2013R1A2A2A01015144).
Abbreviations
- MSCs
Mesenchymal stem cells
- BMSCs
Bone marrow stem cells
- ROS
Reactive oxygen species
- ARE
Antioxidant responsive element
- 18α-GA
18α-Glycyrrhetinic acid
- MEF
Mouse embryonic fibroblast
- CDKI
Cyclin-dependent kinase inhibitor
- Nampt
Nicotinamide phosphoribosyltransferase
- NAM
Nicotinamide
- HO-1
Heme oxygenase-1
- BVR-A
Biliverdin reductase A
- PPAR
Peroxisome proliferator-activated receptor
- VSMC
Vascular smooth muscle cell
- EPC
Endothelial progenitor cell
- HUVEC
Human umbilical vein endothelial cell
- ECM
Extracellular matrix
References
- 1.Banfi A, Muraglia A, Dozin B, et al. Proliferation kinetics and differentiation potential of ex vivo expanded human bone marrow stromal cells: Implications for their use in cell therapy. Exp Hematol. 2000;28:707–715. doi: 10.1016/s0301-472x(00)00160-0. [DOI] [PubMed] [Google Scholar]
- 2.Bruder SP, Jaiswal N, Haynesworth SE. Growth kinetics, self-renewal, and the osteogenic potential of purified human mesenchymal stem cells during extensive subcultivation and following cryopreservation. J Cell Biochem. 1997;64:278–294. doi: 10.1002/(sici)1097-4644(199702)64:2<278::aid-jcb11>3.0.co;2-f. [DOI] [PubMed] [Google Scholar]
- 3.Ringdén O, Uzunel M, Rasmusson I, et al. Mesenchymal stem cells for treatment of therapy-resistant graft-versus-host disease. Transplantation. 2006;81:1390–1397. doi: 10.1097/01.tp.0000214462.63943.14. [DOI] [PubMed] [Google Scholar]
- 4.Stenderup K, Justesen J, Clausen C, Kassem M. Aging is associated with decreased maximal life span and accelerated senescence of bone marrow stromal cells. Bone. 2003;33:919–926. doi: 10.1016/j.bone.2003.07.005. [DOI] [PubMed] [Google Scholar]
- 5.Tokalov SV, Gruener S, Schindler S, et al. A number of bone marrow mesenchymal stem cells but neither phenotype nor differentiation capacities changes with age of rats. Mol Cells. 2007;24:255–260. [PubMed] [Google Scholar]
- 6.Rombouts WJ, Ploemacher RE. Primary murine MSC show highly efficient homing to the bone marrow but lose homing ability following culture. Leukemia. 2003;17:160–170. doi: 10.1038/sj.leu.2402763. [DOI] [PubMed] [Google Scholar]
- 7.Digirolamo CM, Stokes D, Colter D, et al. Propagation and senescence of human marrow stromal cells in culture: a simple colony-forming assay identifies samples with the greatest potential to propagate and differentiate. Br J Haematol. 1999;107:275–281. doi: 10.1046/j.1365-2141.1999.01715.x. [DOI] [PubMed] [Google Scholar]
- 8.Sethe S, Scutt A, Stolzing A. Aging of mesenchymal stem cells. Ageing Res Rev. 2006;5:91–116. doi: 10.1016/j.arr.2005.10.001. [DOI] [PubMed] [Google Scholar]
- 9.Honczarenko M, Le Y, Swierkowski M, et al. Human bone marrow stromal cells express a distinct set of biologically functional chemokine receptors. Stem Cells. 2006;24:1030–1041. doi: 10.1634/stemcells.2005-0319. [DOI] [PubMed] [Google Scholar]
- 10.Brohlin M, Kingham PJ, Novikova LN, et al. Aging effect on neurotrophic activity of human mesenchymal stem cells. PLoS One. 2012;7:e45052. doi: 10.1371/journal.pone.0045052. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Gruber HE, Somayaji S, Riley F, et al. Human adipose-derived mesenchymal stem cells: serial passaging, doubling time and cell senescence. Biotech Histochem. 2012;87:303–311. doi: 10.3109/10520295.2011.649785. [DOI] [PubMed] [Google Scholar]
- 12.Erickson IE, van Veen SC, Sengupta S, et al. Cartilage matrix formation by bovine mesenchymal stem cells in three-dimensional culture is age-dependent. Clin Orthop Relat Res. 2011;469:2744–2753. doi: 10.1007/s11999-011-1869-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.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;129:163–173. doi: 10.1016/j.mad.2007.12.002. [DOI] [PubMed] [Google Scholar]
- 14.Janzen V, Forkert R, Fleming HE, et al. Stem-cell ageing modified by the cyclin-dependent kinase inhibitor p16INK4a. Nature. 2006;443:421–426. doi: 10.1038/nature05159. [DOI] [PubMed] [Google Scholar]
- 15.Molofsky AV, Slutsky SG, Joseph NM, et al. Increasing p16INK4a expression decreases forebrain progenitors and neurogenesis during ageing. Nature. 2006;443:448–452. doi: 10.1038/nature05091. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.von Zglinicki T, Martin-Ruiz C, Saretzki G. Telomeres, cell senescence and human ageing. Signal Transduct. 2005;3:103–114. [Google Scholar]
- 17.Hwang ES. Replicative senescence and senescence-like state induced in cancer-derived cells. Mech Ageing Dev. 2002;123:1681–1694. doi: 10.1016/s0047-6374(02)00102-1. [DOI] [PubMed] [Google Scholar]
- 18.Helmbold H, Galderisi U, Bohn W. The switch from pRb/p105 to Rb2/p130 in DNA damage and cellular senescence. J Cell Physiol. 2012;227:508–513. doi: 10.1002/jcp.22786. [DOI] [PubMed] [Google Scholar]
- 19.Alessio N, Bohn W, Rauchberger V, et al. Silencing of RB1 but not of RB2/P130 induces cellular senescence and impairs the differentiation potential of human mesenchymal stem cells. Cell Mol Life Sci. 2013;70:1637–1651. doi: 10.1007/s00018-012-1224-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Hutter E, Unterluggauer H, Uberall F, et al. Replicative senescence of human fibroblasts: the role of Ras-dependent signaling and oxidative stress. Exp Gerontol. 2002;37:1165–11674. doi: 10.1016/s0531-5565(02)00136-5. [DOI] [PubMed] [Google Scholar]
- 21.Ozawa T. Genetic and functional changes in mitochondria associated with aging. Physiol Rev. 1997;77:425–464. doi: 10.1152/physrev.1997.77.2.425. [DOI] [PubMed] [Google Scholar]
- 22.Packer L, Fuehr K. Low O2 concentration extends the life span of cultured human diploid cells. Nature. 1977;267:423–425. doi: 10.1038/267423a0. [DOI] [PubMed] [Google Scholar]
- 23.Saito H, Hammond AT, Moses RE. The effect of low O2 tension on the in vitro-replicative life span of human diploid fibroblast cells and their transformed derivatives. Exp Cell Res. 1995;217:272–279. doi: 10.1006/excr.1995.1087. [DOI] [PubMed] [Google Scholar]
- 24.Estrada JC, Albo C, Benguría A, et al. Culture of human mesenchymal stem cells at low oxygen tension improves growth and genetic stability by activating glycolysis. Cell Death Differ. 2012;19:743–755. doi: 10.1038/cdd.2011.172. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Cowell JK. The nuclear oncoproteins: RB and p53. Semin Cancer Biol. 1990;1:437–446. [PubMed] [Google Scholar]
- 26.Kensler TW, Wakabayashi N, Biswal S. Cell survival responses to environmental stresses via the Keap1-Nrf2-ARE pathway. Annu Rev Pharmacol Toxicol. 2007;47:89–116. doi: 10.1146/annurev.pharmtox.46.120604.141046. [DOI] [PubMed] [Google Scholar]
- 27.Kapeta S, Chondrogianni N, Gonos ES. Nuclear erythroid factor 2-mediated proteasome activation delays senescence in human fibroblasts. J Biol Chem. 2010;285:8171–8184. doi: 10.1074/jbc.M109.031575. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Kwak MK, Wakabayashi N, Greenlaw JL, et al. Antioxidants enhance mammalian proteasome expression through the Keap1-Nrf2 signaling pathway. Mol Cell Biol. 2003;23:8786–8794. doi: 10.1128/MCB.23.23.8786-8794.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Li J, Johnson D, Calkins M, et al. Stabilization of Nrf2 by tBHQ confers protection against oxidative stress-induced cell death in human neural stem cells. Toxicol Sci. 2005;83:313–328. doi: 10.1093/toxsci/kfi027. [DOI] [PubMed] [Google Scholar]
- 30.Haneline LS. Redox regulation of stem and progenitor cells. Antioxid Redox Signal. 2008;10:1849–1852. doi: 10.1089/ars.2008.2141. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Lu L, Song HF, Zhang WG, et al. Potential role of 20S proteasome in maintaining stem cell integrity of human bone marrow stromal cells in prolonged culture expansion. Biochem Biophys Res Commun. 2012;422:121–127. doi: 10.1016/j.bbrc.2012.04.119. [DOI] [PubMed] [Google Scholar]
- 32.Dröge W, Schipper HM. Oxidative stress and aberrant signaling in aging and cognitive decline. Aging Cell. 2007;6:361–370. doi: 10.1111/j.1474-9726.2007.00294.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Milani P, Ambrosi G, Gammoh O, et al. SOD1 and DJ-1 converge at Nrf2 pathway: a clue for antioxidant therapeutic potential in neurodegeneration. Oxid Med Cell Longev. 2013;2013:836760. doi: 10.1155/2013/836760. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Barbagallo I, Galvano F, Frigiola A, et al. Potential therapeutic effects of natural heme oxygenase-1 inducers in cardiovascular diseases. Antioxid Redox Signal. 2013;18:507–521. doi: 10.1089/ars.2011.4360. [DOI] [PubMed] [Google Scholar]
- 35.Su ZY, Shu L, Khor TO, et al. A perspective on dietary phytochemicals and cancer chemoprevention: oxidative stress, nrf2, and epigenomics. Top Curr Chem. 2013;329:133–162. doi: 10.1007/128_2012_340. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Davies KJ. Degradation of oxidized proteins by the 20S proteasome. Biochimie. 2003;83:301–310. doi: 10.1016/s0300-9084(01)01250-0. [DOI] [PubMed] [Google Scholar]
- 37.Demasi M, Davies KJ. Proteasome inhibitors induce intracellular protein aggregation and cell death by an oxygen-dependent mechanism. FEBS Lett. 2003;542:89–94. doi: 10.1016/s0014-5793(03)00353-3. [DOI] [PubMed] [Google Scholar]
- 38.Grune T, Merker K, Sandig G, Davies KJ. Selective degradation of oxidatively modified protein substrates by the proteasome. Biochem Biophys Res Commun. 2003;305:709–718. doi: 10.1016/s0006-291x(03)00809-x. [DOI] [PubMed] [Google Scholar]
- 39.Lehman NL. The ubiquitin proteasome system in neuropathology. Acta Neuropathol. 2009;118:329–347. doi: 10.1007/s00401-009-0560-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Bulteau AL, Petropoulos I, Friguet B. Age-related alterations of proteasome structure and function in aging epidermis. Exp Gerontol. 2000;35:767–777. doi: 10.1016/s0531-5565(00)00136-4. [DOI] [PubMed] [Google Scholar]
- 41.Chondrogianni N, Stratford FL, Trougakos IP, et al. Central role of the proteasome in senescence and survival of human fibroblasts: induction of a senescence-like phenotype upon its inhibition and resistance to stress upon its activation. J Biol Chem. 2003;278:28026–28037. doi: 10.1074/jbc.M301048200. [DOI] [PubMed] [Google Scholar]
- 42.Hwang JS, Hwang JS, Chang I, Kim S. Age-associated decrease in proteasome content and activities in human dermal fibroblasts: restoration of normal level of proteasome subunits reduces aging markers in fibroblasts from elderly persons. J Gerontol A Biol Sci Med Sci. 2007;62:490–499. doi: 10.1093/gerona/62.5.490. [DOI] [PubMed] [Google Scholar]
- 43.Chondrogianni N, Gonos ES. Proteasome inhibition induces a senescence-like phenotype in primary human fibroblasts cultures. Biogerontology. 2004;5:55–61. doi: 10.1023/b:bgen.0000017687.55667.42. [DOI] [PubMed] [Google Scholar]
- 44.Adams J, Palombella VJ, Sausville EA, et al. Proteasome inhibitors: a novel class of potent and effective antitumor agents. Cancer Res. 1999;59:2615–2622. [PubMed] [Google Scholar]
- 45.Chapple SJ, Siow RC, Mann GE. Crosstalk between Nrf2 and the proteasome: therapeutic potential of Nrf2 inducers in vascular disease and aging. Int J Biochem Cell Biol. 2012;44:1315–1320. doi: 10.1016/j.biocel.2012.04.021. [DOI] [PubMed] [Google Scholar]
- 46.Lavu S, Boss O, Elliott PJ, Lambert PD. Sirtuins–novel therapeutic targets to treat age-associated diseases. Nat Rev Drug Discov. 2008;7:841–853. doi: 10.1038/nrd2665. [DOI] [PubMed] [Google Scholar]
- 47.Park S, Mori R, Shimokawa I. Do sirtuins promote mammalian longevity?: A Critical review on its relevance to the longevity effect induced by calorie restriction. Mol Cells. 2013;35:474–480. doi: 10.1007/s10059-013-0130-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Saunders LR, Sharma AD, Tawney J, et al. miRNAs regulate SIRT1 expression during mouse embryonic stem cell differentiation and in adult mouse tissues. Aging. 2010;2:415–431. doi: 10.18632/aging.100176. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Sasaki T, Maier B, Bartke A, Scrable H. Progressive loss of SIRT1 with cell cycle withdrawal. Aging Cell. 2006;5:413–422. doi: 10.1111/j.1474-9726.2006.00235.x. [DOI] [PubMed] [Google Scholar]
- 50.Huang J, Gan Q, Han L, et al. SIRT1 overexpression antagonizes cellular senescence with activated ERK/S6k1 signaling in human diploid fibroblasts. PLoS One. 2008;3:e1710. doi: 10.1371/journal.pone.0001710. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Yuan HF, Zhai C, Yan XL, et al. SIRT1 is required for long-term growth of human mesenchymal stem cells. J Mol Med (Berl) 2012;90:389–400. doi: 10.1007/s00109-011-0825-4. [DOI] [PubMed] [Google Scholar]
- 52.Peng CH, Chang YL, Kao CL, et al. SirT1–a sensor for monitoring self-renewal and aging process in retinal stem cells. Sensors (Basel) 2010;10:6172–6194. doi: 10.3390/s100606172. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Langley E, Pearson M, Faretta M, et al. Human SIR2 deacetylates p53 and antagonizes PML/p53-induced cellular senescence. EMBO J. 2002;21:2383–2396. doi: 10.1093/emboj/21.10.2383. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Han MK, Song EK, Guo Y, et al. SIRT1 regulates apoptosis and Nanog expression in mouse embryonic stem cells by controlling p53 subcellular localization. Cell Stem Cell. 2008;2:241–251. doi: 10.1016/j.stem.2008.01.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Vaziri H, Dessain SK, Ng Eaton E, et al. hSIR2(SIRT1) functions as an NAD-dependent p53 deacetylase. Cell. 2001;107:149–159. doi: 10.1016/s0092-8674(01)00527-x. [DOI] [PubMed] [Google Scholar]
- 56.Luo J, Nikolaev AY, Imai S, et al. Negative control of p53 by Sir2alpha pro Nemoto motes cell survival under stress. Cell. 2001;107:137–148. doi: 10.1016/s0092-8674(01)00524-4. [DOI] [PubMed] [Google Scholar]
- 57.Ota H, Akishita M, Eto M, et al. Sirt1 modulates premature senescence-like phenotype in human endothelial cells. J Mol Cell Cardiol. 2007;43:571–579. doi: 10.1016/j.yjmcc.2007.08.008. [DOI] [PubMed] [Google Scholar]
- 58.Menssen A, Hydbring P, Kapelle K, et al. The c-MYC oncoprotein, the NAMPT enzyme, the SIRT1-inhibitor DBC1, and the SIRT1 deacetylase form a positive feedback loop. Proc Natl Acad Sci USA. 2012;109:E187–E196. doi: 10.1073/pnas.1105304109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Palacios JA, Herranz D, De Bonis ML, et al. SIRT1 contributes to telomere maintenance and augments global homologous recombination. J Cell Biol. 2010;191:1299–1313. doi: 10.1083/jcb.201005160. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Zhang Y, Chen L, Yang S, Fang D. E2F1: a potential negative regulator of hTERT transcription in normal cells upon activation of oncogenic c-Myc. Med Sci Monit. 2012;18:RA12–RA15. doi: 10.12659/MSM.882192. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Yamashita S, Ogawa K, Ikei T, et al. SIRT1 prevents replicative senescence of normal human umbilical cord fibroblast through potentiating the transcription of human telomerase reverse transcriptase gene. Biochem Biophys Res Commun. 2012;417:630–634. doi: 10.1016/j.bbrc.2011.12.021. [DOI] [PubMed] [Google Scholar]
- 62.Olmos Y, Sánchez-Gómez FJ, Wild B, et al. SirT1 regulation of antioxidant genes is dependent on the formation of a FoxO3a/PGC-1α complex. Antioxid Redox Signal. 2013;19:1507–1521. doi: 10.1089/ars.2012.4713. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Yao H, Sundar IK, Ahmad T, et al. SIRT1 protects against cigarette smoke-induced lung oxidative stress via a FOXO3-dependent mechanism. Am J Physiol Lung Cell Mol Physiol. 2014;306:L816–L828. doi: 10.1152/ajplung.00323.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Lee IH, Cao L, Mostoslavsky R, et al. A role for the NAD-dependent deacetylase Sirt1 in the regulation of autophagy. Proc Natl Acad Sci USA. 2008;105:3374–3379. doi: 10.1073/pnas.0712145105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Kume S, Uzu T, Horiike K, et al. Calorie restriction enhances cell adaptation to hypoxia through Sirt1-dependent mitochondrial autophagy in mouse aged kidney. J Clin Invest. 2010;120:1043–1055. doi: 10.1172/JCI41376. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Chua KF, Mostoslavsky R, Lombard DB, et al. Mammalian SIRT1 limits replicative life span in response to chronic genotoxic stress. Cell Metab. 2005;2:67–76. doi: 10.1016/j.cmet.2005.06.007. [DOI] [PubMed] [Google Scholar]
- 67.Chen X, Barton LF, Chi Y, et al. Ubiquitin-independent degradation of cell-cycle inhibitors by the REGγ proteasome. Mol Cell. 2007;26:843–852. doi: 10.1016/j.molcel.2007.05.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Lin T, Chao C, Saito S, et al. p53 induces differentiation of mouse embryonic stem cells by suppressing Nanog expression. Nat Cell Biol. 2005;7:165–171. doi: 10.1038/ncb1211. [DOI] [PubMed] [Google Scholar]
- 69.Verma-Kurvari S, Savage T, Gowan K, Johnson JE. Lineage-specific regulation of the neural differentiation gene MASH1. Dev Biol. 1996;180:605–617. doi: 10.1006/dbio.1996.0332. [DOI] [PubMed] [Google Scholar]
- 70.Prozorovski T, Schulze-Topphoff U, Glumm R, et al. Sirt1 contributes critically to the redox-dependent fate of neural progenitors. Nat Cell Biol. 2008;10:385–394. doi: 10.1038/ncb1700. [DOI] [PubMed] [Google Scholar]
- 71.Libert S, Cohen D, Guarente L. Neurogenesis directed by Sirt1. Nat Cell Biol. 2008;10:373–374. doi: 10.1038/ncb0408-373. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Fukuhara A, Matsuda M, Nishizawa M, et al. Visfatin: a protein secreted by visceral fat that mimics the effects of insulin. Science. 2005;307:426–430. doi: 10.1126/science.1097243. [DOI] [PubMed] [Google Scholar]
- 73.Bitterman KJ, Anderson RM, Cohen HY, et al. Inhibition of silencing and accelerated aging by nicotinamide, a putative negative regulator of yeast sir2 and human SIRT1. J Biol Chem. 2002;277:45099–45107. doi: 10.1074/jbc.M205670200. [DOI] [PubMed] [Google Scholar]
- 74.Araki T, Sasaki Y, Milbrandt J. Increased nuclear NAD biosynthesis and SIRT1 activation prevent axonal degeneration. Science. 2004;305:1010–1013. doi: 10.1126/science.1098014. [DOI] [PubMed] [Google Scholar]
- 75.van der Veer E, Ho C, O’Neil C, et al. Extension of human cell lifespan by nicotinamide phosphoribosyltransferase. J Biol Chem. 2007;282:10841–10845. doi: 10.1074/jbc.C700018200. [DOI] [PubMed] [Google Scholar]
- 76.Ho C, van der Veer E, Akawi O, Pickering JG. SIRT1 markedly extends replicative lifespan if the NAD + salvage pathway is enhanced. FEBS Lett. 2009;583:3081–3085. doi: 10.1016/j.febslet.2009.08.031. [DOI] [PubMed] [Google Scholar]
- 77.Kang HT, Hwang ES. Nicotinamide enhances mitochondria quality through autophagy activation in human cells. Aging Cell. 2009;8:426–438. doi: 10.1111/j.1474-9726.2009.00487.x. [DOI] [PubMed] [Google Scholar]
- 78.Jang SY, Kang HT, Hwang ES. Nicotinamide-induced mitophagy: event mediated by high NAD +/NADH ratio and SIRT1 protein activation. J Biol Chem. 2012;287:19304–19314. doi: 10.1074/jbc.M112.363747. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Schmeisser K, Mansfeld J, Kuhlow D, et al. Role of sirtuins in lifespan regulation is linked to methylation of nicotinamide. Nat Chem Biol. 2013;9:693–700. doi: 10.1038/nchembio.1352. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Kamat JP, Devasagayam TP. Nicotinamide (vitamin B3) as an effective antioxidant against oxidative damage in rat brain mitochondria. Redox Rep. 1999;4:179–184. doi: 10.1179/135100099101534882. [DOI] [PubMed] [Google Scholar]
- 81.Kuro-o M, Matsumura Y, Aizawa H, et al. Mutation of the mouse klotho gene leads to a syndrome resembling ageing. Nature. 1997;390:45–51. doi: 10.1038/36285. [DOI] [PubMed] [Google Scholar]
- 82.Kurosu H, Ogawa Y, Miyoshi M, et al. Regulation of fibroblast growth factor-23 signaling by klotho. J Biol Chem. 2006;281(10):6120–6123. doi: 10.1074/jbc.C500457200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Urakawa I, Yamazaki Y, Shimada T, et al. Klotho converts canonical FGF receptor into a specific receptor for FGF23. Nature. 2006;444:770–774. doi: 10.1038/nature05315. [DOI] [PubMed] [Google Scholar]
- 84.Liu F, Wu S, Ren H, Gu J. Klotho suppresses RIG-I-mediated senescence-associated inflammation. Nat Cell Biol. 2011;13:254–262. doi: 10.1038/ncb2167. [DOI] [PubMed] [Google Scholar]
- 85.Kuilman T, Michaloglou C, Vredeveld LC, et al. Oncogene-induced senescence relayed by an interleukin-dependent inflammatory network. Cell. 2008;133:1019–1031. doi: 10.1016/j.cell.2008.03.039. [DOI] [PubMed] [Google Scholar]
- 86.de Oliveira RM. Klotho RNAi induces premature senescence of human cells via a p53/p21 dependent pathway. FEBS Lett. 2006;580:5753–5758. doi: 10.1016/j.febslet.2006.09.036. [DOI] [PubMed] [Google Scholar]
- 87.Kikuchi G, Yoshida T, Noguchi M. Heme oxygenase and heme degradation. Biochem Biophys Res Commun. 2005;338:558–567. doi: 10.1016/j.bbrc.2005.08.020. [DOI] [PubMed] [Google Scholar]
- 88.Piantadosi CA, Withers CM, Bartz RR, et al. Heme oxygenase-1 couples activation of mitochondrial biogenesis to anti-inflammatory cytokine expression. J Biol Chem. 2011;286:16374–16385. doi: 10.1074/jbc.M110.207738. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Guillen MI, Megias J, Gomar F, Alcaraz MJ. Heme oxygenase-1 regulates catabolic and anabolic processes in osteoarthritic chondrocytes. J Pathol. 2008;214:515–522. doi: 10.1002/path.2313. [DOI] [PubMed] [Google Scholar]
- 90.Garcia-Arnandis I, Guillen MI, Castejon MA, et al. Heme oxygenase-1 down-regulates high mobility group box 1 and matrix metalloproteinases in osteoarthritic synoviocytes. Rheumatology. 2010;49:854–861. doi: 10.1093/rheumatology/kep463. [DOI] [PubMed] [Google Scholar]
- 91.Barbagallo I, Vanella A, Peterson SJ, et al. Overexpression of heme oxygenase-1 increases human osteoblast stem cell differentiation. J Bone Miner Metab. 2010;28:276–288. doi: 10.1007/s00774-009-0134-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Clérigues V, Guillén MI, Castejón MA, et al. Heme oxygenase-1 mediates protective effects on inflammatory, catabolic and senescence responses induced by interleukin-1β in osteoarthritic osteoblasts. Biochem Pharmacol. 2012;83:395–405. doi: 10.1016/j.bcp.2011.11.024. [DOI] [PubMed] [Google Scholar]
- 93.Baranano DE, Rao M, Ferris CD, Snyder SH. Biliverdin reductase: a major physiologic cytoprotectant. Proc Natl Acad Sci USA. 2002;99:16093–16098. doi: 10.1073/pnas.252626999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Tudor C, Lerner-Marmarosh N, Engelborghs Y, et al. Biliverdin reductase is a transporter of haem into the nucleus and is essential for regulation of HO-1 gene expression by haematin. Biochem J. 2008;413:405–416. doi: 10.1042/BJ20080018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Barone E, Di Domenico F, Sultana R, et al. Heme oxygenase-1 posttranslational modifications in the brain of subjects with Alzheimer disease and mild cognitive impairment. Free Radic Biol Med. 2012;52:2292–2301. doi: 10.1016/j.freeradbiomed.2012.03.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Kim SY, Park SC. Physiological antioxidative network of the bilirubin system in aging and age-related diseases. Front Pharmacol. 2012;3:45. doi: 10.3389/fphar.2012.00045. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Kim SY, Kang HT, Choi HR, Park SC. Biliverdin reductase A in the prevention of cellular senescence against oxidative stress. Exp Mol Med. 2011;43:15–23. doi: 10.3858/emm.2011.43.1.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Alam J, Cook JL. Transcriptional regulation of the heme oxygenase-1 gene via the stress response element pathway. Curr Pharm Des. 2003;9:2499–2511. doi: 10.2174/1381612033453730. [DOI] [PubMed] [Google Scholar]
- 99.Diep QN, Amiri F, Touyz RM, et al. PPARalpha activator effects on Ang II-induced vascular oxidative stress and inflammation. Hypertension. 2002;40:866–871. doi: 10.1161/01.hyp.0000037969.41360.cc. [DOI] [PubMed] [Google Scholar]
- 100.Tao L, Liu HR, Gao E, et al. Antioxidative, antinitrative, and vasculoprotective effects of a peroxisome proliferator-activated receptor-gamma agonist in hypercholesterolemia. Circulation. 2003;108:2805–2811. doi: 10.1161/01.CIR.0000097003.49585.5E. [DOI] [PubMed] [Google Scholar]
- 101.Umeji K, Umemoto S, Itoh S, et al. Cu/Zn superoxide dismutase, PPAR-gamma, and aortic stiffness in hypercholesterolemia. Am J Physiol Heart Circ Physiol. 2006;291:H2522–H2532. doi: 10.1152/ajpheart.01198.2005. [DOI] [PubMed] [Google Scholar]
- 102.Zhang H, Li Y, Fan Y, et al. Klotho is a target gene of PPAR-gamma. Kidney Int. 2008;74:732–739. doi: 10.1038/ki.2008.244. [DOI] [PubMed] [Google Scholar]
- 103.Ricote M, Glass CK. PPARs and molecular mechanisms of transrepression. Biochim Biophys Acta. 2007;1771:926–935. doi: 10.1016/j.bbalip.2007.02.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Nicolakakis N, Aboulkassim T, Ongali B, et al. Complete rescue of cerebrovascular function in aged Alzheimer’s disease transgenic mice by antioxidants and pioglitazone, a peroxisome proliferator-activated receptor gamma agonist. J Neurosci. 2008;28:9287–9296. doi: 10.1523/JNEUROSCI.3348-08.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Briganti S, Flori E, Mastrofrancesco A, et al. Azelaic acid reduced senescence-like phenotype in photo-irradiated human dermal fibroblasts: possible implication of PPARγ. Exp Dermatol. 2013;22:41–47. doi: 10.1111/exd.12066. [DOI] [PubMed] [Google Scholar]
- 106.Kim HJ, Ham SA, Paek KS, et al. Transcriptional up-regulation of antioxidant genes by PPARδ inhibits angiotensin II-induced premature senescence in vascular smooth muscle cells. Biochem Biophys Res Commun. 2011;406:564–569. doi: 10.1016/j.bbrc.2011.02.091. [DOI] [PubMed] [Google Scholar]
- 107.Kim HJ, Ham SA, Kim SU, et al. Transforming growth factor-beta1 is a molecular target for the peroxisome proliferator-activated receptor delta. Circ Res. 2008;102:193–200. doi: 10.1161/CIRCRESAHA.107.158477. [DOI] [PubMed] [Google Scholar]
- 108.Lim HA, Lee EK, Kim JM, et al. PPARγ activation by baicalin suppresses NF-κB-mediated inflammation in aged rat kidney. Biogerontology. 2012;13:133–145. doi: 10.1007/s10522-011-9361-4. [DOI] [PubMed] [Google Scholar]
- 109.Kim HJ, Ham SA, Kim MY, et al. PPARδ coordinates angiotensin II-induced senescence in vascular smooth muscle cells through PTEN-mediated inhibition of superoxide generation. J Biol Chem. 2011;286:44585–44593. doi: 10.1074/jbc.M111.222562. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Ham SA, Hwang JS, Yoo T, et al. Ligand-activated PPARδ inhibits UVB-induced senescence of human keratinocytes via PTEN-mediated inhibition of superoxide production. Biochem J. 2012;444:27–38. doi: 10.1042/BJ20111832. [DOI] [PubMed] [Google Scholar]
- 111.Kim MY, Kang ES, Ham SA, et al. The PPARδ-mediated inhibition of angiotensin II-induced premature senescence in human endothelial cells is SIRT1-dependent. Biochem Pharmacol. 2012;84:1627–1634. doi: 10.1016/j.bcp.2012.09.008. [DOI] [PubMed] [Google Scholar]
- 112.Chen LJ, Cheng MF, Ku PM. Lin JW (2014) Rosiglitazone increases cerebral klotho expression to reverse baroreflex in type 1-like diabetic rats. Biomed Res Int. 2014;2014(309151):13. doi: 10.1155/2014/309151. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Yang HC, Deleuze S, Zuo Y, et al. The PPARgamma agonist pioglitazone ameliorates aging-related progressive renal injury. J Am Soc Nephrol. 2009;20:2380–2388. doi: 10.1681/ASN.2008111138. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Zhang T, Wang F, Xu HX, et al. Activation of nuclear factor erythroid 2-related factor 2 and PPARγ plays a role in the genistein-mediated attenuation of oxidative stress-induced endothelial cell injury. Br J Nutr. 2013;109:223–235. doi: 10.1017/S0007114512001110. [DOI] [PubMed] [Google Scholar]
- 115.Cho HY, Gladwell W, Wang X, et al. Nrf2-regulated PPARγ expression is critical to protection against acute lung injury in mice. Am J Respir Crit Care Med. 2010;182:170–182. doi: 10.1164/rccm.200907-1047OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Steele AD, Emsley JG, Ozdinler PH, et al. Prion protein (PrPc) positively regulates neural precursor proliferation during developmental and adult mammalian neurogenesis. Proc Natl Acad Sci USA. 2006;103:3416–3421. doi: 10.1073/pnas.0511290103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Chakraborty C, Nandi S, Jana S. Prion disease: a deadly disease for protein misfolding. Current Pharmaceutical Biotechnology. 2005;6:167–177. doi: 10.2174/1389201053642321. [DOI] [PubMed] [Google Scholar]
- 118.Mohanty ST, Cairney CJ, Chantry AD, et al. A small molecule modulator of prion protein increases human mesenchymal stem cell lifespan, ex vivo expansion, and engraftment to bone marrow in NOD/SCID mice. Stem Cells. 2012;30:1134–1143. doi: 10.1002/stem.1065. [DOI] [PubMed] [Google Scholar]
- 119.Brown DR, Besinger A. Prion protein expression and superoxide dismutase activity. Biochem J. 1998;334:423–429. doi: 10.1042/bj3340423. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Brown DR, Wong BS, Hafiz F, et al. Normal prion protein has an activity like that of superoxide dismutase. Biochem J. 1999;344(Pt 1):1–5. [PMC free article] [PubMed] [Google Scholar]
- 121.Malaisé M, Schätzl HM, Bürkle A. The octarepeat region of prion protein, but not the TM1 domain, is important for the antioxidant effect of prion protein. Free Radic Biol Med. 2008;45:1622–1630. doi: 10.1016/j.freeradbiomed.2008.08.024. [DOI] [PubMed] [Google Scholar]
- 122.Kramer ML, Kratzin HD, Schmidt B, et al. Prion protein binds copper within the physiological concentration range. J Biol Chem. 2001;276:16711–16719. doi: 10.1074/jbc.M006554200. [DOI] [PubMed] [Google Scholar]
- 123.Rachidi W, Vilette D, Guiraud P, et al. Expression of prion protein increases cellular copper binding and antioxidant enzyme activities but not copper delivery. J Biol Chem. 2003;278:9064–9072. doi: 10.1074/jbc.M211830200. [DOI] [PubMed] [Google Scholar]
- 124.Thompson MJ, Borsenberger V, Louth JC, et al. Design, synthesis, and structure-activity relationship of indole-3-glyoxylamide libraries possessing highly potent activity in a cell line model of prion disease. J Med Chem. 2009;52:7503–7511. doi: 10.1021/jm900920x. [DOI] [PubMed] [Google Scholar]
- 125.Hill JM, Zalos G, Halcox JP, et al. Circulating endothelial progenitor cells, vascular function, and cardiovascular risk. N Engl J Med. 2003;348:593–600. doi: 10.1056/NEJMoa022287. [DOI] [PubMed] [Google Scholar]
- 126.Kawamoto A, Gwon HC, Iwaguro H, et al. Therapeutic potential of ex vivo expanded endothelial progenitor cells for myocardial ischemia. Circulation. 2001;103:634–637. doi: 10.1161/01.cir.103.5.634. [DOI] [PubMed] [Google Scholar]
- 127.Llevadot J, Murasawa S, Kureishi Y, et al. HMG-CoA reductase inhibitor mobilizes bone marrow-derived endothelial progenitor cells. J Clin Invest. 2001;108:399–405. doi: 10.1172/JCI13131. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Kureishi Y, Luo Z, Shiojima I, et al. The HMG-CoA reductase inhibitor simvastatin activates the protein kinase Akt and promotes agiogenesis in normocholesterolemic animals. Nat Med. 2000;6:1004–1010. doi: 10.1038/79510. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Assmus B, Urbich C, Aicher A, et al. HMG-CoA reductase inhibitors reduce senescence and increase proliferation of endothelial progenitor cells via regulation of cell cycle regulatory genes. Circ Res. 2003;92:1049–1055. doi: 10.1161/01.RES.0000070067.64040.7C. [DOI] [PubMed] [Google Scholar]
- 130.Breitschopf K, Zeiher AM, Dimmeler S. Proatherosclerotic factors induce telomerase inactivation in endothelial cells through an Akt-dependent mechanism. FEBS Lett. 2001;493:21–25. doi: 10.1016/s0014-5793(01)02272-4. [DOI] [PubMed] [Google Scholar]
- 131.Dimmeler S, Aicher A, Vasa M, et al. HMG-CoA reductase inhibitors (statins) increase endothelial progenitor cells via the PI 3-kinase/Akt pathway. J Clin Invest. 2001;108:391–397. doi: 10.1172/JCI13152. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Chandramohan V, Jeay S, Pianetti S, Sonenshein GE. Reciprocal control of Forkhead box O 3a and c-Myc via the phosphatidylinositol 3-kinase pathway coordinately regulates p27Kip1 levels. J Immunol. 2004;172:5522–5527. doi: 10.4049/jimmunol.172.9.5522. [DOI] [PubMed] [Google Scholar]
- 133.Wassmann S, Laufs U, Bäumer AT, et al. HMG-CoA reductase inhibitors improve endothelial dysfunction in normocholesterolemic hypertension via reduced production of reactive oxygen species. Hypertension. 2001;37:1450–1457. doi: 10.1161/01.hyp.37.6.1450. [DOI] [PubMed] [Google Scholar]
- 134.Wassmann S, Laufs U, Muller K, et al. Cellular antioxidant effects of atorvastatin in vitro and in vivo. Arterioscler Thromb Vasc Biol. 2002;22:300–305. doi: 10.1161/hq0202.104081. [DOI] [PubMed] [Google Scholar]
- 135.Ota H, Eto M, Kano MR, et al. Induction of endothelial nitric oxide synthase, SIRT1, and catalase by statins inhibits endothelial senescence through the Akt pathway. Arterioscler Thromb Vasc Biol. 2010;30:2205–2211. doi: 10.1161/ATVBAHA.110.210500. [DOI] [PubMed] [Google Scholar]
- 136.Sung B, Park S, Yu BP, Chung HY. Modulation of PPAR in aging, inflammation, and calorie restriction. J Gerontol A Biol Sci Med Sci. 2004;59:997–1006. doi: 10.1093/gerona/59.10.b997. [DOI] [PubMed] [Google Scholar]
- 137.Zhu JH, Chen JZ, Wang XX, et al. Homocysteine accelerates senescence and reduces proliferation of endothelial progenitor cells. J Mol Cell Cardiol. 2006;40:648–652. doi: 10.1016/j.yjmcc.2006.01.011. [DOI] [PubMed] [Google Scholar]
- 138.Guterres FA, Martinez GR, Rocha ME, Winnischofer SM. Simvastatin rises reactive oxygen species levels and induces senescence in human melanoma cells by activation of p53/p21 pathway. Exp Cell Res. 2013;319:2977–2988. doi: 10.1016/j.yexcr.2013.07.026. [DOI] [PubMed] [Google Scholar]
- 139.Lindner U, Kramer J, Behrends J, et al. Improved proliferation and differentiation capacity of human mesenchymal stromal cells cultured with basement-membrane extracellular matrix proteins. Cytotherapy. 2010;12(8):992–1005. doi: 10.3109/14653249.2010.510503. [DOI] [PubMed] [Google Scholar]
- 140.Lai Y, Sun Y, Skinner CM, et al. Reconstitution of marrow-derived extracellular matrix ex vivo: a robust culture system for expanding large-scale highly functional human mesenchymal stem cells. Stem Cells Dev. 2010;19:1095–1107. doi: 10.1089/scd.2009.0217. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Liu X, Yin D, Zhang Y, et al. Vascular endothelial cell senescence mediated by integrin beta4 in vitro. FEBS Lett. 2007;581:5337–5342. doi: 10.1016/j.febslet.2007.10.027. [DOI] [PubMed] [Google Scholar]
- 142.Sun C, Liu X, Qi L, et al. Modulation of vascular endothelial cell senescence by integrin β4. J Cell Physiol. 2010;225:673–681. doi: 10.1002/jcp.22262. [DOI] [PubMed] [Google Scholar]
- 143.Sun Y, Li W, Lu Z, et al. Rescuing replication and osteogenesis of aged mesenchymal stem cells by exposure to a young extracellular matrix. FASEB J. 2011;25:1474–1485. doi: 10.1096/fj.10-161497. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Pioletti DP, Takei H, Lin T, et al. The effects of calcium phosphate cement particles on osteoblast functions. Biomaterials. 2000;21:1103–1114. doi: 10.1016/s0142-9612(99)00250-1. [DOI] [PubMed] [Google Scholar]
- 145.Choi HR, Cho KA, Kang HT, et al. Restoration of senescent human diploid fibroblasts by modulation of the extracellular matrix. Aging Cell. 2011;10:148–157. doi: 10.1111/j.1474-9726.2010.00654.x. [DOI] [PubMed] [Google Scholar]
- 146.de la Serna IL, Ohkawa Y, Imbalzano AN. Chromatin remodelling in mammalian differentiation: lessons from ATP-dependent remodellers. Nat Rev Genet. 2006;7:461–473. doi: 10.1038/nrg1882. [DOI] [PubMed] [Google Scholar]
- 147.Alessio N, Squillaro T, Cipollaro M, et al. The BRG1 ATPase of chromatin remodeling complexes is involved in modulation of mesenchymal stem cell senescence through RB-P53 pathways. Oncogene. 2010;29:5452–5463. doi: 10.1038/onc.2010.285. [DOI] [PubMed] [Google Scholar]
- 148.Nemoto S, Fergusson MM, Finkel T. Nutrient availability regulates SIRT1 through a forkhead-dependent pathway. Science. 2004;306:2105–2108. doi: 10.1126/science.1101731. [DOI] [PubMed] [Google Scholar]
- 149.Chang TC, Wentzel EA, Kent OA, et al. Transactivation of miR-34a by p53 broadly influences gene expression and promotes apoptosis. Mol Cell. 2007;26:745–752. doi: 10.1016/j.molcel.2007.05.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Yamakuchi M. MicroRNA regulation of SIRT1. Front Physiolo. 2012 doi: 10.3389/fphys.2012.00068. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Abdelmohsen K, Srikantan S, Kuwano Y, Gorospe M. miR-519 reduces cell proliferation by lowering RNA-binding protein HuR levels. Proc. Natl. Acad. Sci. U.S.A. 2008;105:20297–20302. doi: 10.1073/pnas.0809376106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Abdelmohsen K, Pullmann R, Jr, Lal A, et al. Phosphorylation of HuR by Chk2 regulates SIRT1 expression. Mol Cell. 2007;25:543–557. doi: 10.1016/j.molcel.2007.01.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Chen J, Xavier S, Moskowitz-Kassai E, et al. Cathepsin cleavage of sirtuin 1 in endothelial progenitor cells mediates stress-induced premature senescence. Am J Pathol. 2012;180:973–983. doi: 10.1016/j.ajpath.2011.11.033. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Park JH, Yi HW, DiMaio D, Hwang ES. Heterogeneous upregulation of lysosomal genes in human fibroblasts and cancer cells undergoing senescence. Korean J Genet. 2008;29:521–527. [Google Scholar]
- 155.Caito S, Rajendrasozhan S, Cook S, et al. SIRT1 is a redox-sensitive deacetylase that is post-translationally modified by oxidants and carbonyl stress. FASEB J. 2010;24:3145–3159. doi: 10.1096/fj.09-151308. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Lessard J, Sauvageau G. Bmi-1 determines the proliferative capacity of normal and leukaemic stem cells. Nature. 2003;423:255–260. doi: 10.1038/nature01572. [DOI] [PubMed] [Google Scholar]
- 157.Molofsky AV, He S, Bydon M, et al. Bmi-1 promotes neural stem cell self-renewal and neural development but not mouse growth and survival by repressing the p16Ink4a and p19Arf senescence pathways. Genes Dev. 2005;19:1432–1437. doi: 10.1101/gad.1299505. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Jacobs JJ, Kieboom K, Marino S, et al. The oncogene and Polycomb-group gene bmi-1 regulates cell proliferation and senescence through the ink4a locus. Nature. 1999;397:164–168. doi: 10.1038/16476. [DOI] [PubMed] [Google Scholar]
- 159.Itahana K, Zou Y, Itahana Y, et al. Control of the replicative life span of human fibroblasts by p16 and the polycomb protein Bmi-1. Mol Cell Biol. 2003;23:389–401. doi: 10.1128/MCB.23.1.389-401.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Wang Y, Guan Y, Wang F, et al. Bmi-1 regulates self-renewal, proliferation and senescence of human fetal neural stem cells in vitro. Neurosci Lett. 2010;476(2):74–78. doi: 10.1016/j.neulet.2010.04.006. [DOI] [PubMed] [Google Scholar]
- 161.He S, Iwashita T, Buchstaller J, et al. Bmi-1 over-expression in neural stem/progenitor cells increases proliferation and neurogenesis in culture but has little effect on these functions in vivo. Dev Biol. 2009;328:257–272. doi: 10.1016/j.ydbio.2009.01.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Dimri GP, Martinez JL, Jacobs JJ, et al. The Bmi-1 oncogene induces telomerase activity and immortalizes human mammary epithelial cells. Cancer Res. 2002;62:4736–4745. [PubMed] [Google Scholar]
- 163.Zhang X, Soda Y, Takahashi K, et al. Successful immortalization of mesenchymal progenitor cells derived from human placenta and the differentiation abilities of immortalized cells. Biochem Biophys Res Commun. 2006;351:853–859. doi: 10.1016/j.bbrc.2006.10.125. [DOI] [PubMed] [Google Scholar]
- 164.Jacobs JJ, Scheijen B, Voncken JW, et al. Bmi-1 collaborates with c-Myc in tumorigenesis by inhibiting c-Myc-induced apoptosis via INK4a/ARF. Genes Dev. 1999;13:2678–2690. doi: 10.1101/gad.13.20.2678. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.Jiang L, Li J, Song L. Bmi-1, stem cells and cancer. Acta Biochim Biophys Sin (Shanghai) 2009;41:527–534. doi: 10.1093/abbs/gmp040. [DOI] [PubMed] [Google Scholar]
- 166.Bulavin DV, Demidov ON, Saito S, et al. Amplification of PPM1D in human tumors abrogates p53 tumor-suppressor activity. Nat Genet. 2002;31:210–215. doi: 10.1038/ng894. [DOI] [PubMed] [Google Scholar]
- 167.Bulavin DV, Phillips C, Nannenga B, et al. Inactivation of the Wip1 phosphatase inhibits mammary tumorigenesis through p38 MAPK-mediated activation of the p16(Ink4a)-p19(Arf) pathway. Nat Genet. 2004;36:343–350. doi: 10.1038/ng1317. [DOI] [PubMed] [Google Scholar]
- 168.Lee JS, Lee MO, Moon BH, et al. Senescent growth arrest in mesenchymal stem cells is bypassed by Wip1-mediated downregulation of intrinsic stress signaling pathways. Stem Cells. 2009;27:1963–1975. doi: 10.1002/stem.121. [DOI] [PubMed] [Google Scholar]
- 169.Cha BH, Lee JS, Kim SW, et al. The modulation of the oxidative stress response in chondrocytes by Wip1 and its effect on senescence and dedifferentiation during in vitro expansion. Biomaterials. 2013;34:2380–2388. doi: 10.1016/j.biomaterials.2012.12.009. [DOI] [PubMed] [Google Scholar]
- 170.Rossi DJ, Jamieson CH, Weissman IL. Stems cells and the pathways to aging and cancer. Cell. 2008;132:681–696. doi: 10.1016/j.cell.2008.01.036. [DOI] [PubMed] [Google Scholar]
- 171.Parrinello S, Samper E, Krtolica A, et al. Oxygen sensitivity severely limits the replicative lifespan of murine fibroblasts. Nat Cell Biol. 2003;5:741–747. doi: 10.1038/ncb1024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Dröge W. Aging-related changes in the thiol/disulfide redox state: implications for the use of thiol antioxidants. Exp Gerontol. 2002;37:1333–1345. doi: 10.1016/s0531-5565(02)00175-4. [DOI] [PubMed] [Google Scholar]
- 173.Giles GI. The redox regulation of thiol dependent signaling pathways in cancer. Curr Pharm Des. 2006;12:4427–4443. doi: 10.2174/138161206779010549. [DOI] [PubMed] [Google Scholar]
- 174.Simonsen JL, Rosada C, Serakinci N, et al. Telomerase expression extends the proliferative life-span and maintains the osteogenic potential of human bone marrow stromal cells. Nat Biotechnol. 2002;20(6):592–596. doi: 10.1038/nbt0602-592. [DOI] [PubMed] [Google Scholar]
- 175.Abdallah BM, Haack-Sørensen M, Burns JS, et al. Maintenance of differentiation potential of human bone marrow mesenchymal stem cells immortalized by human telomerase reverse transcriptase gene despite extensive proliferation. Biochem Biophys Res Commun. 2005;326(3):527–538. doi: 10.1016/j.bbrc.2004.11.059. [DOI] [PubMed] [Google Scholar]
- 176.Liang XJ, Chen XJ, Yang DH, et al. Differentiation of human umbilical cord mesenchymal stem cells into hepatocyte-like cells by hTERT gene transfection in vitro. Cell Biol Int. 2012;36(2):215–221. doi: 10.1042/CBI20110350. [DOI] [PubMed] [Google Scholar]
- 177.Stroikin Y, Dalen H, Brunk UT, Terman A. Testing the “garbage” accumulation theory of ageing: mitotic activity protects cells from death induced by inhibition of autophagy. Biogerontology. 2005;6:39–47. doi: 10.1007/s10522-004-7382-y. [DOI] [PubMed] [Google Scholar]
- 178.Giordano S, Darley-Usmar V, Zhang J. Autophagy as an essential cellular antioxidant pathway in neurodegenerative disease. Redox Biol. 2013;2:82–90. doi: 10.1016/j.redox.2013.12.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.Hubbard BP, Sinclair DA. Small molecule SIRT1 activators for the treatment of aging and age-related diseases. Trends Pharmacol Sci. 2014;35:146–154. doi: 10.1016/j.tips.2013.12.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180.Pacholec M, Bleasdale JE, Chrunyk B, et al. SRT1720, SRT2183, SRT1460, and resveratrol are not direct activators of SIRT1. J Biol Chem. 2010;285:8340–8351. doi: 10.1074/jbc.M109.088682. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181.Fox RJ, Kita M, Cohan SL, et al. BG-12 (dimethyl fumarate): a review of mechanism of action, efficacy, and safety. Curr Med Res Opin. 2014;30:251–262. doi: 10.1185/03007995.2013.849236. [DOI] [PubMed] [Google Scholar]
- 182.Suh JH, Shenvi SV, Dixon BM, et al. Decline in transcriptional activity of Nrf2 causes age-related loss of glutathione synthesis, which is reversible with lipoic acid. Proc Natl Acad Sci USA. 2004;101:3381–3386. doi: 10.1073/pnas.0400282101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183.Lii CK, Liu KL, Cheng YP, et al. Sulforaphane and alpha-lipoic acid upregulate the expression of the pi class of glutathione S-transferase through c-jun and Nrf2 activation. J Nutr. 2010;140:885–892. doi: 10.3945/jn.110.121418. [DOI] [PubMed] [Google Scholar]
- 184.Zhao XD, Zhou YT, Lu XJ. Sulforaphane enhances the activity of the Nrf2-ARE pathway and attenuates inflammation in OxyHb-induced rat vascular smooth muscle cells. Inflamm Res. 2013;62:857–863. doi: 10.1007/s00011-013-0641-0. [DOI] [PubMed] [Google Scholar]
- 185.Dinkova-Kostova AT, Liby KT, Stephenson KK, et al. Extremely potent triterpenoid inducers of the phase 2 response: correlations of protection against oxidant and inflammatory stress. Proc Natl Acad Sci USA. 2005;102:4584–4589. doi: 10.1073/pnas.0500815102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186.Kiemer AK, Bildner N, Weber NC, Vollmar AM. Characterization of heme oxygenase 1 (heat shock protein 32) induction by atrial natriuretic peptide in human endothelial cells. Endocrinology. 2003;144:802–812. doi: 10.1210/en.2002-220610. [DOI] [PubMed] [Google Scholar]