- Review
- Open access
- Published:
Cellular senescence in Alzheimer’s disease: from physiology to pathology
Translational Neurodegeneration volume 13, Article number: 55 (2024)
Abstract
Alzheimer’s disease (AD) is one of the most common neurodegenerative disorders, characterized by the accumulation of Aβ and abnormal tau hyperphosphorylation. Despite substantial efforts in development of drugs targeting Aβ and tau pathologies, effective therapeutic strategies for AD remain elusive. Recent attention has been paid to the significant role of cellular senescence in AD progression. Mounting evidence suggests that interventions targeting cellular senescence hold promise in improving cognitive function and ameliorating hallmark pathologies in AD. This narrative review provides a comprehensive summary and discussion of the physiological roles, characteristics, biomarkers, and commonly employed in vivo and in vitro models of cellular senescence, with a particular focus on various cell types in the brain, including astrocytes, microglia, oligodendrocyte precursor cells, neurons, and endothelial cells. The review further delves into factors influencing cellular senescence in AD and emphasizes the significance of targeting cellular senescence as a promising approach for AD treatment, which includes the utilization of senolytics and senomorphics.
Background
Cellular senescence was first discovered almost 60 years ago in multiple strains of fibroblasts cultured in vitro [1, 2]. It was initially identified as a stable exit from the cell cycle caused by limited proliferation of normal human fibroblasts [1, 2]. Currently, cellular senescence is defined as an irreversible cell cycle arrest in response to either intrinsic or extrinsic insults, including telomere shortening and aberrant oncogenic activation (e.g., overexpression of HrasV12), mitochondrial dysfunction, oxidative and genotoxic stress, irradiation, and chemotherapeutic agents [3]. Therefore, senescence is also considered a defense mechanism against unnecessary cellular damage [4]. Cellular senescence is closely associated with aging [5]. As previously reported, hallmarks of aging are divided into three main categories: (1) primary hallmarks, causes of age-related cellular damage; (2) antagonistic responses in response to cellular damage; and (3) integrative hallmarks, consequences of the responses and culprits of the clinical phenotype [5]. Senescence belongs to the antagonistic class [5]. However, increasing evidence shows that the accumulation of senescent cells has deleterious effects on the tissue microenvironment by secreting pro-inflammatory and matrix-degrading molecules (known as senescence-associated secretory phenotype, SASP) [6,7,8]. SASP contributes to the pathological progression of aging-related neurodegenerative disease [6,7,8].
Aging is the most significant risk factor for AD [9]. As the most common form of dementia among older people [6], AD-associated cellular senescence has recently attracted increasing attention [6, 7, 10]. AD treatment strategies in the past 30 years were mainly focused on the pathological hallmarks, i.e., extracellular beta-amyloid (Aβ) accumulation and intraneuronal abnormal tau hyperphosphorylation [11]. However, there are currently no effective pharmacotherapeutic approaches for AD prevention and treatment [12]. Recent studies suggest that astrocytes, microglia, and oligodendrocyte progenitor cells (OPCs) exhibit a senescence phenotype in the Aβ plaque environment [7, 13, 14]. These studies provide a promising new avenue for studying senolytic strategies in AD.
In this narrative review, we provide a comprehensive overview and discussion of the physiological roles, characteristics, biomarkers, and models of cellular senescence, with a specific focus on various brain cell types, including astrocytes, microglia, OPCs, neurons, and endothelial cells. We synthesize existing knowledge from both primary research and relevant reviews to explore the factors influencing cellular senescence in AD and highlight the therapeutic potential of targeting cellular senescence, including the use of senolytics and senomorphics.
Understanding cellular senescence: definitions, classifications, and key characteristics
Definition and classification of cellular senescence
Cellular senescence is classified into replicative senescence and stress-induced premature senescence (SIPS) [15]. It is characterized by a stable cell cycle arrest triggered by developmental signals or by intrinsic and extrinsic stimuli [15]. Replicative senescence refers to the process of normal, nonmalignant cells undergoing cell cycle arrest after a limited number of divisions [16]. Key factors leading to replicative senescence include sustained proliferation, telomerase deficiency, and telomere shortening [15]. Most primary cells have low telomerase levels, leading to cell growth arrest after several vigorous generations due to telomere shortening and chromosomal fusions [17]. Supporting this concept, some studies showed that telomere shortening leads to p53 activation, a key mediator of cellular senescence, which further promotes senescence [18, 19]. External and internal stresses also induce cellular senescence, which is termed SIPS and occurs independently of telomerase length [20]. Chemical or physical stimuli-induced stress, including oxidative stress, inflammation, mitochondrial dysfunction, suboptimal culture conditions, and various genotoxic stressors, lead to SIPS [21]. For instance, superoxide radicals, ultraviolet radiation, and hypoxia can induce cellular senescence in cultured cells [22,23,24]. Replicative senescence and SIPS share characteristics such as growth arrest, telomere shortening, DNA damage, p53 activation from DNA damage accumulation, and altered expression of proteins involved in cell-cycle regulation [25, 26]. Thus, SIPS is frequently used as a model to study normal aging [25].
Key features of cellular senescence
Growth arrest
As mentioned previously, one of the hallmarks of cellular senescence is the irreversible cell cycle arrest [3]. Senescent cells can not initiate DNA replication despite adequate growth conditions and do not respond to growth factors or mitogenic stimuli [27]. The growth arrest is induced by the expression of cyclin-dependent kinase (CDK) inhibitors described below in detail. Like the terminally differentiated cells, the senescent cells are irreversibly withdrawn from the cell cycle. However, unlike differentiated cells, senescent cells are not specialized effector cells and have no specific physiological function [28]. Growth arrest is a potent tumor-suppressive mechanism, but it is not conducive to neuronal survival and damage repair [29].
Apoptosis resistance
Factors that determine cell fate choice between apoptosis and senescence remain unclear. However, cell type, intensity and nature of the stress/stimuli/damage, and changes of cellular apoptosis-related proteins may determine the direction of cell fate [27]. In contrast to apoptotic cells, senescent cells exhibit anti-apoptotic properties [30]. Senescent cells display significantly increased levels of anti-apoptotic proteins BCL-W and BCL-XL, which result in apoptosis resistance [31]. The anti-apoptotic properties induce an accumulation of senescent cells without function, contributing to normal aging and promoting the development of age-related disorders [32].
SASP
Although senescent cells are growth-arrested, they are still in a metabolically active state and display a hyper-secretory phenotype termed SASP [8]. SASP is a fundamental feature of cellular senescence [33]. In AD, SASP occurs in different cell lines and leads to secretion of various pro-inflammatory cytokines, including IL-6, IL-8, TNF-α, TGF-β, and IL-1β [7]. The excessive release of pro-inflammatory cytokines exacerbates pathological changes in AD, including Aβ accumulation, neurofibrillary tangles, neuronal loss, and neuronal degeneration [7, 34].
DNA damage response (DDR)
DDR is initiated by DNA damage or telomere shortening. In addition to arresting cell-cycle progression, cells respond to DNA damage by attempting to repair DNA damage via DDR pathways in aging. Progressive telomere shortening also triggers a DDR, and the markers of DDR are localized to the telomeres [35]. The DDR includes formation of DNA damage foci at either uncapped telomeres or persistent DNA strand breaks, which contribute to cellular senescence [36]. Indeed, cellular senescence can be considered a permanent DNA-damage response state [37].
Biomarkers of cellular senescence
p16INK4a in cellular senescence
The p16INK4a gene, also called CDKN2a (cyclin-dependent kinase inhibitor 2A), is located on chromosome 4 in mice and chromosome 9, band p21.3 in humans [38]. The p16INK4a gene encodes two proteins, p16INK4a and p14arf [39]. The p16INK4a gene, first identified as MTS1 (multi-tumor suppressor 1) in1994, is inactivated or mutated in nearly 50% of human cancers [40], which suggests an essential role of p16INK4a in the cell cycle [41]. The cell cycle has four stages called G1, S, G2, and M phases [42]. Cell cycle checkpoints regulate progression through these four phases [43]. The G1 checkpoint controls the G1-to-S transition, and p16INK4a is a critical component of the G1 checkpoint [39, 44]. p16INK4a binds to CDK4 and CDK6, inhibits their activity, and prevents retinoblastoma (Rb) tumor suppressor protein phosphorylation. The dephosphorylated status of Rb physically associates with E2Fs (E2F1-E2F3) and blocks their transactivation domain [45]. Moreover, the interaction between the E2F and Rb transactivation domain inhibits the expression of E2F1 target genes that are crucial for G1/S transition [45, 46]. Therefore, the increased level of p16INK4a can induce G1 cell cycle arrest in aged and stressed tissues [47]. Clearance of p16INK4a-positive senescent cells protects against AD and other aging-associated disorders [7, 48, 49]. Now, p16INK4a has been regarded as a marker of aging and cellular senescence, although the currently available antibodies poorly detect p16INK4a in mice [50].
p21CIP1/WAF1/SDI1 in cellular senescence
p21CIP1/WAF1/SDI1 is another potent CDK inhibitor [51]. The p21-encoding gene was initially found as a target of p53 to mediate its role as a tumor suppressor [52]. p21 interacts with CDK2-associated complexes and inhibits the cell cycle from entering the S phase [51]. Therefore, as a downstream target of p53, p21CIP1/WAF1/SDI1 is activated to trigger cell cycle arrest and contributes to cell cycle arrest, apoptosis, and differentiation [51]. Consistently, p21CIP1/WAF1/SDI1 is closely associated with stress or danger cell responses in multiple tissues [53]. For example, p21 triggers the cell cycle growth arrest in the lung to modulate alveolar inflammation and destruction [53]. As a sensor of cellular stress, p21CIP1/WAF1/SDI1 in cellular senescence provides a logical explanation of how p21 is involved in arrested tissue repair but contributes to tumor suppression.
Senescence-associated beta-galactosidase (SA-β-gal) in cellular senescence
SA-β-gal is the most extensively utilized biomarker for cellular senescence [54]. Increased SA-β-gal activity can be detected as cells enter the senescent state [55]. A quantitative assay of SA-β-gal activity found that the normalized β-gal activity in senescent cells is two folds that in pre-senescent cells [56]. Therefore, assays measuring SA-β-gal activity offer approaches to detect cellular senescence. Several substrates for SA-β-gal are currently widely used in the assays, including 5-bromo-4-chloro-3-indolyl-β-D-galactopyranoside (known as x-gal), C12FDG, and CellEvent senescence green reagent [57,58,59]. The x-gal and C12FDG can be converted into a blue precipitate by SA-β-gal and used as an indicator for cellular senescence [58, 59].
Lipofuscin in cellular senescence
Lipofuscin is a nondegradable aggregate of oxidation products of covalently cross-linked proteins, lipids, and metals within lysosomes [60]. Lipofuscin accumulates with age and is considered a hallmark of senescent cells [61]. Lipofuscin is an autofluorescent material that can be detected using fluorescence microscopy [60]. Besides, Sudan Black B and the biotinylated chemical compound derived from Sudan Black B have been described as a method of detecting lipofuscin based on lipid detection [62].
Cellular senescence in AD: contributing factors and cell types affected
Factors contributing to cellular senescence in AD
Under pathological conditions of AD, cellular senescence induces the formation of SASP, leading to the accumulation of senescent cells. This process is accompanied by excessive release of reactive oxygen species (ROS) and pro-inflammatory factors, exacerbating mitochondrial dysfunction, oxidative stress, and inflammatory responses. Moreover, it accelerates amyloid-beta (Aβ) deposition and triggers excessive phosphorylation of tau, forming a vicious cycle between cellular senescence and AD pathology (Fig. 1). In addition, altered proteostasis, DNA damage and repair, telomerase deficiency, and telomere shortening also contribute to cellular senescence in AD [63,64,65]. The following paragraphs review and discuss factors influencing cellular senescence in AD.
Pathological vicious cycle of cellular senescence in AD. Under pathological conditions of AD, cellular senescence induces SASP, leading to accumulation of senescent cells. This process is accompanied by excessive release of reactive oxygen species and pro-inflammatory factors, exacerbating mitochondrial dysfunction, oxidative stress, and inflammatory responses. Moreover, it accelerates Aβ deposition and excessive phosphorylation of tau, forming a vicious cycle between cellular senescence and AD pathology. SASP, senescence-associated secretory phenotype
Mitochondrial dysfunction
Mitochondrial dysfunction is a hallmark in neurodegenerative diseases and brain injuries [66, 67]. Disrupted mitochondrial structure, abnormal function, and impaired biogenesis have been observed in AD [66, 67]. Specifically, an imbalance between mitochondrial fusion and fission leads to excessive fragmentation, evidenced by decreased fusion-associated proteins (e.g., MFN1, MFN2, OPA1) and increased fission-associated proteins (e.g., FIS1, MFF, MIEF, DRP1) in the cortex and hippocampus [67, 68]. Additionally, defects of oxidative phosphorylation caused by impairment of mitochondrial enzymes such as Complexes II and IV, contribute to reduced ATP production [67, 68]. Impaired mitochondrial trafficking and defective mitophagy have also been observed in AD [69, 70].
Mitochondrial dysfunction is closely linked to cellular senescence [71]. Senescent cells exhibit reduced mitochondrial membrane potential and defects in respiratory chain complexes [71]. Deficiency of complex I assembly factors alone can induce cell senescence [72]. Indeed, mitochondrial DNA is essential for electron transport and mitochondrial function [73]. Mitochondrial genomic instability induces cellular senescence when mitochondrial DNA mutations are accompanied by increased ROS [73]. Additionally, mitochondrial content is associated with cellular senescence [71, 74]. Both in vivo and in vitro studies have observed elevated mitochondrial mass in senescence, possibly as a compensatory mechanism for mitochondrial dysfunction [75, 76]. Consistent with this, reducing mitochondrial mass significantly inhibits cellular senescence, suggesting that targeting mitochondria may help alleviate senescence [74]. Mitochondrial Ca2+ overload also mediates the essential role of mitochondria in cellular senescence [77]. Increasing evidence shows that mitochondrial Ca2+ overload contributes to AD pathology [77]. The mitochondrial Ca2+ overload causes decreased membrane potential and excessive production of ROS [71, 77]. Subsequently, the excessively released ROS causes DNA damage and telomere shortening, activating the p53 pathway and cellular senescence [64]. These findings suggest that mitochondrial dysfunction is a potential intervention target for alleviating cellular senescence in AD.
Oxidative stress
As mentioned previously, mitochondrial dysfunction-induced oxidative stress causes genomic DNA damage and activates cellular senescence-associated pathways [64]. Therefore, suppressing oxidative stress may be a way to inhibit cellular senescence [78]. According to previous studies, anti-oxidative substances and approaches potentially improve age-related neurodegenerative diseases by targeting cellular senescence [34, 66, 79]. For instance, curcumin, a natural phenol with antioxidant activities, exhibits therapeutic potential for AD treatment [80]. An in vitro study showed that curcumin significantly ameliorates hydrogen peroxide-induced endothelial cellular senescence [81]. Nicotinamide, a vitamin B3 derivative, significantly alleviates the replicative senescence of human fibroblasts by reducing ROS production [82]. Photobiomodulation, a non-invasive low-level laser therapy, directly improves mitochondrial function by promoting cytochrome c oxidase activity, which reduces electron leakage, thereby decreasing oxidative stress and mitigating the senescence of bone marrow mesenchymal stem cells [79]. Further, a previous study found that cellular ROS in neurons could induce cellular senescence by activating the IL-6/STAT3 pathway [83], and that inhibiting ROS production or the IL-6/STAT3 pathway alleviated cellular senescence [83]. These findings suggest that oxidative stress or its downstream targets may contribute to cellular senescence.
Neuroinflammation
AD is also accompanied by a robust inflammatory response characterized by excessive release of pro-inflammatory factors and overactivation of glial cells [66]. The neuroinflammatory response is triggered by the accumulation of Aβ, tau, and damage-associated molecular patterns (etc., mitochondrial constituents) [66]. At the early stage, glial cells, including astrocytes and microglia, are activated in response to the pathological changes [66, 84]. During this stage, they are involved in the clearance of pathological molecules through microglial phagocytosis and glymphatic clearance pathway [85, 86]. However, as the disease progresses, microglia and astrocytes fail to remove pathological molecules [66, 84], and chronically activated microglia and astrocytes excessively release pro-inflammatory cytokines and ROS, causing detrimental effects [66]. Indeed, the chronic activation of glial cells and excessive release of pro-inflammatory cytokines exacerbate mitochondrial dysfunction, oxidative stress, and accumulation of pathological molecules, forming a vicious cycle and contributing to cellular senescence [66]. A previous study observed decreased nicotinamide adenine dinucleotide (NAD+) levels and an increased neuroinflammatory response in AD mouse brains [87]. Exogenous NAD+ supplementation alleviates microglial and astrocytic activation and reduces pro-inflammatory cytokine release [87]. Interestingly, the decreased neuroinflammation is accompanied by attenuated senescence of brain cells, including microglia, astrocytes, neurons, and OPCs [87]. In fact, the pro-inflammatory secretory phenotype SASP, the hallmark of senescent cells, releases significantly increased pro-inflammatory cytokines, exacerbating pathological changes and cellular senescence, which highlights the intimate association between inflammation and cellular senescence [15].
Altered proteostasis
Proteostasis or protein homeostasis refers to appropriate protein synthesis, folding, post-translational modification, trafficking, and degradation [63]. Altered proteostasis is a common feature of several neurodegenerative diseases and has been extensively studied in AD [88,89,90]. Defective protein synthesis caused by dysfunctional mRNA translation control has been discovered as an early event and a trigger or a mediator of AD pathogenesis [91]. In addition, alterations of mechanistic target of rapamycin complex 1 (mTORC1) signaling can cause changes in proteostasis in AD [92, 93]. As part of mTORC1, the mechanistic target of rapamycin (mTOR) is a central nutrient-sensitive regulator and regulates cellular proliferation, metabolism, and protein synthesis [93]. mTOR exists in 2 complexes, mTORC1 and mTORC2, with different cellular functions [94]. mTORC1 plays a major role in protein synthesis and degradation [93]. mTORC1 is overactivated in AD and contributes to AD progression [95]. For example, although protein translation inhibition is observed in AD, overactivation of mTORC1 causes detrimental excessive synaptic protein synthesis [96]. Moreover, elevated mTORC1 activity suppresses autophagy in AD, consistent with the impaired autophagy and Aβ clearance in AD patients and animal models [95, 97].
Recent evidence indicates that altered proteostasis and cellular senescence share several pathogenic mechanisms, and cellular senescence works as an adaptive stress response to altered proteostasis [63]. In a previous study, prolonged neuronal culture displayed cellular senescence and altered proteostasis [63]. However, attenuation of the altered proteostasis with a mTOR antagonist alleviated the proteotoxic stress and reduced cellular senescence, suggesting that altered proteostasis contributes to cellular senescence [63]. Indeed, altered proteostasis is one of the typical features of cellular senescence [98]. A change of cellular state from normal functioning to the senescent state needs the participation of proteome alteration [98]. Furthermore, it should be noted that although stress sensing is remarkably promoted in human senescent cells, the stress adaptation at the gene transcription level is reduced in these cells because of the altered proteostasis [99].
DNA damage and repair
Emerging evidence supports the essential role of DNA damage and repair in healthy aging and various neurodegenerative diseases, including AD and multiple sclerosis [100, 101]. DNA repair maintains genomic stability during healthy aging and alleviates DNA damage [102]. In AD, increased DNA damage and dysfunctional DNA repair further facilitate AD pathogenesis [15, 103]. Increasing evidence supports that DNA damage is a crucial pathological cause of and an early event in AD [15, 104, 105]. The excessive release of ROS causes DNA–protein and DNA-DNA cross-links, formation of modified bases and DNA adducts, and double-stranded DNA breaks [105]. In response to DNA damage, DDR is initiated, which involves a series of events including DNA damage detection (damage sensors), repair, and cellular fate decision [106]. In aging, DNA damage and the activation of the DDR can influence cell fate decisions, leading to outcomes such as cell death or cellular senescence [107]. In AD, different cellular stressors (e.g., oxidative stress, DNA damage, and mitochondrial stress) combined with proteinopathy lead to DNA damage and a DDR cascade that causes senescence [108]. DNA damage contributes to the activation of DDR machinery and the transcriptional regulation of the INK4/ARF locus [109,110,111]. The DDR machinery activates the p53-p21CIP1 axis, followed by inhibition of CDK2 activity, resulting in hyperphosphorylation of RB and cell cycle exit [109, 110, 112]. Moreover, DNA damage also induces the expression of INK4/ARF genes [113]. Under physiological conditions, the expression of INK4-ARF is arrested by epigenetic factors and polycomb proteins [114]. However, when INK4-ARF is activated, the increased level of ARF prevents p53 degradation and causes increased expression of p16INK4a, followed by inhibition of CDK4 and CDK6 and long-lasting cell cycle arrest [114]. In fact, DDR is a shared characteristic between cellular senescence and telomere shortening in aging [115]. Therefore, inefficient DNA repair under pathological conditions and aging results in cellular senescence and cell-cycle arrest [115].
Taupathy
Tau hyperphosphorylation is one of the typical pathologies in AD [116]. Tau is a tubulin-binding protein that promotes microtubule assembly and stabilization under physiological conditions [117]. The stabilization of microtubules is essential for fundamental metabolic and biochemical activities, including cell division, intracellular nutrient transport, cell motility, and cellular shape maintenance [66, 118]. However, under pathological conditions, tau hyperphosphorylation contributes to abnormal metabolic and biochemical activities in AD [66, 118, 119]. In AD, the abnormally phosphorylated tau dissociates from and destabilizes microtubules, and aggregates into oligomers, paired helical filaments, and intracellular neurofibrillary tangles [66, 118, 120]. There is increasing evidence for the close association between tauopathy and cellular senescence [121,122,123]. For example, a previous study found that the neurofibrillary tangle (NFT)-bearing neurons exhibited a senescence-associated transcriptomic profile in post-mortem AD brain tissues and tau-transgenic mice [122]. More interestingly, a senolytic therapy significantly ameliorated the NFT burden in the transgenic mice, suggesting the close association between hyperphosphorylated tau aggregation and cellular senescence [122]. Further, like neurons, astrocytes bearing pathological tau oligomers in human AD brains also display a senescence-like phenotype. Mechanistically, tau oligomers trigger the nucleo-cytoplasmic translocation of high mobility group box 1 (HMGB1) [121]. HMGB1 release causes the formation of SASP, which induces senescence in adjacent cells [121]. Treatment with HMGB1 inhibitors significantly abolished the tau oligomer-induced cellular senescence [121]. Furthermore, senescent microglia are closely associated with tauopathy and AD progression, although the underlying mechanisms remain unclear [124]. These findings suggest that tauopathy is involved in the cellular senescence in AD.
Aβ
Like hyperphosphorylated tau, oligomeric Aβ exposure induces cellular senescence in AD [125]. Aβ is generated from amyloid precursor protein (APP), a transmembrane protein [66]. APP is sequentially cleaved under physiological conditions by α- and γ-secretases, generating C-terminal fragments (p3, AICD, and CTF83) and soluble amyloid precursor protein α [66]. However, under pathological conditions, APP undergoes sequential cleavages by β- and γ-secretases to produce neurotoxic Aβ [66]. The excessive Aβ accumulation results in extracellular Aβ aggregation into amyloid plaques [66]. Numerous studies have found that Aβ accumulation triggers excessive activation of glial cells, neuroinflammation, oxidative stress, and mitochondrial dysfunction [66, 85, 126], all contributing to cellular senescence in AD [125]. For example, Aβ oligomers are able to induce senescence of OPCs and neurons through Aβ functional receptors and some proteins that regulate neuroinflammation and metabolic pathways [125, 127]. For instance, an in vitro study found that Aβ1-42 oligomers induce neural stem/progenitor cell senescence, as evidenced by significantly increased p16 and SA-β-gal [127]. During this process, formylpeptide receptor 2, one of the Aβ1-42 functional receptors, activates its downstream ROS-p38MAPK signaling, which limits neural stem/progenitor cell function and leads to failure of neurogenesis [127]. In another in vitro study, Aβ1-42 oligomers induced neuronal senescence by suppressing the sirtuin-1 level, while preservation of the sirtuin-1 level significantly attenuated Aβ-induced cell senescence [125]. These findings suggest that Aβ accumulation contributes to cellular senescence in AD.
Telomerase deficiency and telomere shortening
Telomerase, a ribonucleoprotein complex, is a telomere-terminal transferase necessary for maintaining telomere length by adding guanine-rich repetitive sequences to chromosome ends [128]. In AD, Aβ accumulation inhibits telomerase activity and causes telomere shortening [129]. Telomere shortening is one of the hallmarks of aging, wherein progressive telomere shortening eventually causes replicative senescence [130]. In aging and aging-related diseases including AD, telomerase deficiency and telomere shortening compromise neurogenesis and cause cellular senescence [15, 131]. In contrast, telomerase reverse transcriptase (TERT), a key component of telomerase, could preserve telomere length and alleviate or reverse brain tissue degeneration and cellular senescence in AD [132]. Interestingly, a previous study found that human neurons with TERT expression had no hyperphosphorylated tau detected, while neurons with tau pathology were not detected with TERT staining [133]. In addition, TERT-deficient animals show AD-like phenotypes and increased tau pathology [133, 134]. As tauopathy is closely associated with cellular senescence [121, 122], the telomerase deficiency may also contribute to cellular senescence by exacerbating tau pathology in AD [134]. Therefore, telomerase deficiency can cause both replicative senescence through telomere shortening and stress-induced premature senescence by exacerbating tau pathology, oxidative stress, and neuroinflammation.
Cell types affected by senescence in AD
Cellular senescence, as an important player in the pathological progression of AD, has been identified in various cell types within AD brains [135]. In the following, we will introduce cellular senescence in the AD brain.
Astrocyte senescence in AD
Astrocytes are the most abundant glial cells in the central nervous system [136]. They play an essential role in maintaining the homeostasis of brain function, including synaptic support, axon guidance, and regulation of the blood–brain barrier (BBB) and blood flow [137,138,139]. As a critical regulator of inflammatory responses, astrocytes respond to pathological changes and exert anti-inflammatory effects at the early stage of AD. However, astrocytes are polarized into neurotoxic phenotypes at the late AD stage and exacerbate AD pathology [140].
Astrocytes undergo both replicative senescence and stress-induced senescence, with a series of senescent characteristics and markers, including growth arrest, increased p53 and p21 expression, and elevated SA-β-gal activity in vitro and in vivo [141, 142]. Studies in primary human and rodent astrocytes found that various stimuli and factors (such as H2O2, ionizing radiation, and proteasome inhibition) can lead to stress-induced senescence in astrocytes with classical senescence features [143, 144]. Senescent astrocytes display multiple classic senescent characteristics [143, 144]. First, senescent astrocytes exhibit cellular arrest both in vitro and in vivo [145]. As mentioned previously, the upregulation of p21WAF1 by p53 inhibits CDK2 activity and initiates cell cycle arrest [52]. Furthermore, downregulation or inhibition of CDK4 and CDK6 by p16INK4a blocks the astrocytic entry to the S phase and mediates permanent cell cycle arrest [47]. Second, senescent astrocytes exhibit nuclear changes. A previous autopsy study found prominent typical chromatin structural changes (senescence-associated heterochromatic foci) and increased levels of histone modification (e.g., γH2AX) in astrocytes of AD patients [146]. The nuclear changes are associated with decreased expression of proliferation-related genes, leading to cell cycle arrest [145]. Third, astrocyte senescence is also characterized by the generation of SASP factors [147]. The SASP secretes pro-inflammatory factors, promotes the senescence state, and enhances age-related neurogenerative disease [144, 148].
The neuroprotective function of astrocytes is compromised in AD [149, 150]. Cellular senescence is one of the reasons that contribute to astrocyte dysfunction in AD [149, 150]. Senescent astrocytes lack normal function, acquire the SASP, and secrete inflammatory cytokines and proteases that affect the function of neighboring cells [13]. The population of p16INK4a-positive astrocytes is significantly increased in AD patients, suggesting prominent astrocyte senescence [13]. An in vitro study in human astrocyte cultures confirmed that Aβ1-42 peptides could elevate SA-β-gal activity and p16INK4a expression, suggesting that excessive production of Aβ1-42 is involved in astrocytic senescent response [13]. In addition, the senescent astrocytes release multiple inflammatory cytokines, including IL-6, RANTES, IL-8, and ICAM-1, inducing a vicious circle [13]. Emerging evidence indicates that inhibiting astrocyte senescence or clearing senescent astrocytes could prevent AD initiation or slow AD progression [10, 151]. For example, a previous study using primary human astrocytes showed that inhibiting astrocyte senescence by administering the p53 isoform Δ133p53 may alleviate AD pathologies and confer neuroprotective effects [151]. Furthermore, eliminating senescent astrocytes via the genetic approach and pharmacological drugs (e.g., ABT263, dasatinib, and quercetin) alleviates Aβ plaque deposition, reduces neuroinflammation, and improves cognitive function in an ex vivo model of AD and in transgenic p16-3MR mice with cognitive deficits [152, 153]. These findings suggest that astrocyte senescence is a potential target for alleviating AD pathology.
Microglial senescence
Microglia, the primary immune cells in brain, play essential roles in neuroinflammation and brain infection [154, 155]. Moreover, they modulate synaptic pruning and eliminate unnecessary synapses in adulthood [156]. Excessive synaptic pruning mediated by adult brain microglia contributes to neurodegenerative diseases and related behavioral deficits [156]. Indeed, microglia exhibit multiple roles in AD initiation and development [67, 104, 154]. The increasing deposition of Aβ initiates the activation of microglia at early stages of AD to remove Aβ deposition through phagocytosis [34, 157]. However, with the progression of AD, especially at the late stage, the capability of microglia to clear amyloid plaques is compromised and they transform to a pro-inflammatory phenotype that contributes to AD progression [158]. Microglial activation is now understood to be a spectrum rather than a binary M1/M2 classification [159]. Microglia can be categorized into specific subsets such as disease-associated microglia (DAM), homeostatic microglia, intermediate states that exhibit characteristics of both pro-inflammatory and anti-inflammatory responses, and other subsets with disease-related genes and RNA signatures observed by single-cell RNA sequencing [155, 160,161,162]. At the early stage of AD, microglia predominantly exhibit a homeostatic or anti-inflammatory phenotype that facilitates Aβ clearance, while in the late stages, they transform to a disease-associated or pro-inflammatory state with diminished phagocytic capacity [162,163,164,165,166,167]. Regulation of microglial phagocytosis and microglial recruitment around amyloid plaques provides a potential approach for AD treatment [168, 169].
In addition, the DAM can secrete many same pro-inflammatory molecules as those found in the SASP, complicating the identification of senescent microglia [155, 160,161,162,163,164,165,166,167]. Several criteria can be considered to differentiate senescent microglia from DAM: (1) persistent inflammatory profile: senescent microglia maintain a chronic pro-inflammatory state, unlike the transient inflammatory responses seen in DAM [170]; (2) senescence markers: increased expression of senescence markers such as p16INK4a, p21CIP1/WAF1, and β-galactosidase activity can help identify senescent microglia [170]; (3) functional decline: senescent microglia often exhibit reduced phagocytic activity and impaired responses to injury or infection [170, 171]; (4) epigenetic changes: senescent cells may undergo specific epigenetic modifications, such as changes in DNA methylation and histone modification patterns, which can be applied as additional markers [172].
Senescent microglia have also been found in multiple brain regions in AD [154, 173]. In AD-like mouse models and post-mortem human brain samples, microglial senescence is detected in the cortex and hippocampus, contributing to Aβ pathology [154]. The generation of microglial senescence is correlated with sustained microglial proliferation at the early AD stage [154]. The replicative senescence is characterized by telomere shortening and elevated SA-β-gal activity, similar to other cellular senescence [154]. Specifically, senescent microglia are characterized by a decreased phagocytic ability, resulting in deficiency in Aβ clearance and Aβ accumulation in AD mouse models [154]. In addition, increased accumulation of extracellular Aβ and abnormal intracellular tau contribute to myelin loss at the early AD stage [34, 174]. Microglia are essential for phagocytosis and degradation of myelin debris [156]. However, when the myelin debris accumulation exceeds the degradation capacity of microglial lysosomes, microglia may become senescent and promote the release of pro-inflammatory cytokines [109]. The impaired lysosomal degradative capacity and the sustained secretion of inflammatory factors further lead to increased myelin debris and Aβ accumulation [109]. The excessive production of myelin debris and overload of myelin residues within microglia in turn lead to senescence. These changes disrupt microglial production of factors required for oligodendrocyte-dependent remyelination, suggesting that microglia senescence disrupts neuronal repair [109, 175].
Increasing evidence supports inhibiting cellular senescence as a new therapeutic paradigm for AD [154]. In an AD-like mouse model, inhibiting microglial proliferation at the early stage hinders microglial senescence and attenuates AD-associated pathology including Aβ accumulation and synaptic damage [154]. The MAPTP301SPS19 transgenic mice exhibit a significant increase in the expression of senescence marker p16INK4a at the early AD stage [10], preceding the initiation of aggressive tauopathy [10]. Eliminating the p16INK4a-expressing glial cells, including senescent microglia, prevents neurofibrillary tangle deposition and mitigates cognitive deficits [10].
OPC senescence
OPCs, also known as NG2-glia, oligodendrocyte precursor cells, or synantocytes [176], are the fourth major glial subtype in the brain and the source of oligodendrocytes [177]. Oligodendrocytes are the primary myelin-forming cells in the central nervous system (CNS) [176]. As the primary source of oligodendrocytes, OPCs are essential for myelination and myelin repair [176]. During the early development of brain tissue, OPCs migrate along the cerebral vasculature and spread to the entire brain [178]. The long-distance migration of OPCs supports the generation of oligodendrocytes and neuronal myelination [178]. OPC migration is guided by several signaling molecules, including Sonic hedgehog, bone morphogenic proteins, and Wnt proteins [179]. Similarly, several growth factors facilitate OPC migration, including hepatocyte growth factor, fibroblast growth factor, and vascular endothelial growth factor [179]. Once OPCs reach the specific brain region, the cells detach from the endothelium and differentiate into pre-oligodendrocytes. As oligodendrocytes mature, they contact neuronal axons and initiate myelin assembly [179, 180]. Indeed, the OPCs retain the ability to proliferate and differentiate throughout adulthood [181]. Following acute demyelinating injury or in demyelination-related neurodegenerative diseases, the OPCs migrate to the lesion area and differentiate into oligodendrocytes, contributing to myelin repair [182]. Dysfunction of OPCs contributes to demyelination and compromised remyelination in aging and neurodegenerative diseases [183]. The compromised remyelination with aging is characterized by poor response of OPCs to differentiation signals and weakened recruitment of OPCs to the damaged area [183]. Improving remyelination by rejuvenating OPCs has shown therapeutic potential for neurodegenerative diseases [183].
In AD, loss of OPCs occurs at an early stage and has been recognized as an early sign of AD pathology [176]. Elevated OPC senescence and functional disruption have been detected in the aging brain [176]. In addition, OPC senescence and demyelination were found to be aggravated in a transgenic AD mouse model [176]. Senescent OPC-related SASP contributes to local neuroinflammation and is closely associated with neuronal damage [7]. The SASP exacerbates neuronal damage and AD pathology by releasing pro-inflammatory factors [7]. The unfavorable inflammatory microenvironment in turn impairs neurogenesis and OPC differentiation and increases Aβ accumulation [176, 184, 185]. Myelin disruption is a typical change in AD and directly leads to memory deficits [174]. In 3 × Tg-AD mice, loss of myelin and OPCs is detected as early as 6 months of age, suggesting that the dysfunction of OPCs at the early stage of AD is not induced by replicative exhaustion but possibly by OPC senescence [176]. Indeed, OPCs around the senile plaques display the senescence phenotype in AD patients and AD animal models, suggesting that Aβ exposure causes OPC senescence [7].
Evidence from transgenic AD mice suggests that senolytic treatment targeting OPC senescence can alleviate typical AD pathological changes, including Aβ accumulation and hippocampal-dependent cognitive deficits [7]. Treatment with FDA-approved ‘senolytic’ compounds dasatinib and quercetin significantly alleviates pro-inflammatory cytokines, reducing Aβ accumulation in APP/PS1 AD mice [7]. In addition, treatments that promote OPC generation and differentiation exhibit promising therapeutic effects in AD [186, 187]. Preserving differentiation and reducing senescence of OPCs rescued short-term memory deficits and enhanced myelin formation in an AD mouse model [186]. As mentioned, the OPCs migrate along cerebral vasculature, spread to the entire brain, and contribute to remyelination [180]. Therefore, the loss of migration of senescent OPCs may contribute to perivascular clustering and impaired remyelination, although more studies are still needed [176, 188].
Neuronal senescence
Neurons are the fundamental units and the primary executor of signal transmission within the brain [189]. However, neuronal cells are often subjected to physiological and pathological stress, leading to elevated levels of DNA damage and an activated DDR [190]. The DNA damage causes aberrant cell-cycle re-entry for some neurons and leads to neuronal apoptosis [190]. However, the neuronal DDR promotes cell-cycle arrest and induces a senescence-like phenotype in neurons, which helps protect them from apoptotic death [190]. The surviving senescence-like neurons have all the features of cellular senescence, including mitochondrial dysfunction, metabolic dysfunction, and excessive release of pro-oxidant and pro-inflammatory cytokines [190].
The adult CNS neurons are often considered permanently postmitotic; however, this notion may not fully capture the complexities of their cell cycle regulation [191,192,193]. These highly specialized cells must consistently monitor their cell cycle status. Relaxation of this vigilance can trigger re-entry into the cell cycle, resulting in an altered and vulnerable state that often results in cell death [191,192,193]. Notably, neurons susceptible to neurodegeneration are also at an increased risk of initiating cell cycle processes characterized by expression of cell cycle proteins and DNA replication. This connection between cell cycle dynamics and neuronal death is fundamental to the pathology of various neurodegenerative diseases [191,192,193]. Depending on their maturation stage, neurons that do not adequately suppress cell cycle re-entry face different outcomes [191,192,193]. In younger, differentiating neurons, re-initiating the cell cycle can lead to cell death within hours, a process that can be prevented through cell cycle inhibition [191,192,193]. In contrast, adult neurons that re-enter the cell cycle may express cell cycle proteins and replicate portions of their genome. However, the cell cycle often remains prolonged, where they neither complete the cell cycle nor undergo cell death for an extended period [191,192,193].
Therefore, it is worth noting that defining neuronal senescence is a challenge in non-proliferating cells, such as post-mitotic and terminally differentiated neurons [194]. Historically, neurons were believed to escape from senescence, as senescence was thought to occur only in dividing cells. Consequently, most studies on brain cell senescence focused on glial cells [13, 154]. However, studies have demonstrated that post-mitotic neurons also undergo senescence, characterized by expression of markers like SA-β-gal, MCP-1, γ-H2AX, and 4-HNE, leading to the concept of amitosenescence [195, 196]. Long-term cultured neurons exhibit hallmark features of senescence earlier than glial cells, including increased SA-β-gal activity, SASP, DNA damage, and astrogliosis, which can be mitigated by senolytic agents such as resveratrol [197]. Neuronal senescence involves sustained DNA damage, high ROS production, altered transcription, increased p21CIP1 expression, and activation of the p38MAPK pathway [198]. Therefore, despite the challenges in defining senescence in neurons, a combination of alternative markers and functional changes may help determine whether neurons are truly senescent. These indicators include all the aforementioned cellular senescence markers.
Consistent with other cells within the brain [154], neuronal senescence is considered a source of oxidative stress and neuroinflammation, suggesting the potential contribution of neuronal senescence in aging and neurodegenerative diseases [198]. In a previous study, more than 97% of the senescent excitatory neurons overlapped with abnormal tau in AD post-mortem human brain tissue, suggesting the close association between neuronal senescence and typical AD pathology [199]. Consistent with these findings, in an AD transgenic mouse model, the expression of Cdkn2a was significantly increased in the NFT-containing neurons [122]. Furthermore, the cellular senescence markers, including p21, Cdkn2a, and p16INK4a, were colocalized with NFTs in the brain of a tau transgenic mouse model and elevated in human AD, suggesting the existence of neuronal senescence in both human and animal models [122]. Moreover, a study using the 5 × FAD mouse model further specified the cell types of senescence within the hippocampus [200]. Co-staining of p16 and other cellular markers confirmed that p16 was mainly located within neurons, and in a small proportion of astrocytes and microglia [200]. Notably, neuronal senescence occurs even at the early stage of AD, suggesting that neuronal senescence can be considered an indicator and a typical pathological characteristic of AD [200]. In vitro experiments confirmed the elevation of p16 expression and established the association between Aβ and neuronal senescence in cultured primary neurons [200].
Increasing evidence suggests that inhibiting neuronal senescence or clearing senescent neurons may be a possible treatment for AD [200]. In a tau-transgenic mouse model, clearance of senescent neurons significantly attenuated AD pathologies, including ventricular enlargement, neurofibrillary tangle burden, neuronal damage, and neuronal degeneration [200]. A recent study found that AD is characterized by elevated post-mitotic neuronal senescence in humans [194]. The senescent neurons release pro-inflammatory cytokines and cause a cascade of brain inflammation at the late AD stage [194]. However, reducing neuronal senescence to normal levels could ameliorate neuroinflammation and slow AD progression [194].
Endothelial cellular senescence
Brain endothelial cells are the major component of the microvasculature that forms the BBB, protecting the brain against toxins and pathogens and restricting the access of soluble and cellular substances from the blood into the brain [201]. As the main component of BBB, brain endothelial cells contribute to regulating local cerebral blood flow and neurovascular unit function [202, 203]. However, the brain endothelial cells undergo significant changes with age and experience senescence-associated stress during aging, including excessive oxidative stress, increased innate immunity, accumulation of DNA damage, and telomere shortening [204]. As an exquisite sensor of aging-associated circulatory cues, brain endothelial cells and BBB are potential therapeutic targets for AD [201, 205].
Increasing studies indicate that the senescence of endothelial cells contributes to the dysfunction of neurovascular units and BBB [204, 206]. Both in vivo and in vitro studies have demonstrated endothelial cell senescence induced by external stimuli and aging [207]. For example, γ-irradiation causes rat primary brain endothelial cells to acquire SASP, as evidenced by significantly increased DNA damage and upregulated expression of p16 and p53 [207]. These changes impair the angiogenic capacity [207]. In aged mice, significantly increased expression of senescence core genes, senescence effector genes, and SASP genes was detected in the brain, accompanied by markedly increased senescent endothelial cells [208]. The underlying mechanisms for endothelial cell senescence include oxidative stress, DNA damage, mitochondrial dysfunction, and inflammation [209, 210].
Mounting evidence suggests that endothelial cell senescence contributes to AD progression and pathogenesis [211]. A positive feedback relationship has been reported between NFT formation and cellular senescence [122]. In the prefrontal cortex of AD patients, expression of endothelial senescence-associated genes is elevated, contributing to AD-related BBB dysfunction and cerebral blood flow impairment [211]. Endothelial senescence has also been detected in the in vitro cell cultures [212]. In vitro studies showed that neurotoxic Aβ exposure induces endothelial cell senescence [213]. Although the causal relationship between endothelial cell senescence and AD remains unclear [109], DNA damage and senescence in the endothelial cells may lead to neurovascular unit dysfunction in elderly individuals with AD [214]. However, to the best of our knowledge, no studies have specifically investigated the effects of clearing senescent endothelial cells in AD.
Targeting cellular senescence with senotherapeutics in AD
As mentioned previously, senescent cells acquire SASP in AD and other neurodegenerative diseases, releasing many inflammatory cytokines and matrix-degrading molecules [7, 215]. Therefore, accumulation of senescent cells induces chronic activation of the immune system, resulting in reduced senescent cell clearance and increased accumulation of senescent cells, thereby forming a vicious cycle [215]. Several potential therapeutic approaches that target senescent cells by clearing senescent cells or selectively blocking SASP using natural or synthetic compounds have been studied in animals and in clinical trials [7, 215]. These therapeutics include natural or synthetic compounds that are members of senolytics and senomorphics.
Senolytics
Senolytics are small molecules that selectively clear senescent cells through cellular apoptosis [216]. Therefore, components that affect anti-apoptotic pathways may serve as senolytics. For example, Bcl-2 family inhibitors, BH3 mimetics, Hsp90 inhibitors, p53 binding inhibitors, HDAC inhibitors, p38MAPK inhibitors, and JAK/STAT inhibitors are widely used senolytics (Table 1) [215, 217,218,219,220,221,222,223]. Senolytics significantly alleviate AD pathology [7, 215]. For instance, dasatinib (a protein tyrosine kinase inhibitor) and quercetin (a natural compound that serves as a senomorphic) co-administration effectively removes senescent OPCs, alleviates neuroinflammation, attenuates Aβ accumulation, and improves cognitive function, suggesting the great potential of senescent cell clearance in AD therapy [7].
In a tau-transgenic mouse model that displays late-stage pathology with a senescence-associated transcriptomic profile [122], dasatinib and quercetin (DQ) senolytic treatment for 12 weeks (two consecutive days every other week) significantly reduced NFT density, neuronal loss, and neurodegeneration, suggesting a strong association between cellular senescence and AD pathology [122]. Moreover, in APP/PS1 AD mice, Aβ plaque-associated OPCs display a senescence-like phenotype, suggesting that OPCs around the senile plaques lose their function in neuronal repair and demyelination [7]. Similarly, DQ senolytic treatment significantly alleviates AD pathology, as evidenced by attenuated Aβ accumulation and cognitive impairment [7]. Further mechanistic studies found that the DQ senolytic treatment ameliorates Aβ load by clearing senescent OPCs from the plaque environment and reducing SASP-induced excessive neuroinflammation, suggesting that senolytic treatment is a potential therapeutic approach for AD [7].
In addition to animal studies (Table 2), several senolytic therapies are currently undergoing clinical trials [224, 225]. In a phase I feasibility trial, oral QD administration was proven safe, with dasatinib showing a good profile of BBB penetration [224]. Although cognitive and neuroimaging endpoints did not show improvement after treatment, CSF levels of senescence-related cytokines and chemokines displayed a trend of decrease and CSF Aβ levels showed a trend of increase. These findings suggest the potential of QD in treating AD under safety, tolerability, and feasibility [224].
Senomorphics
Senomorphics are molecules that inhibit or block some of the characteristics of SASP without killing senescent cells [215]. Currently, therapies using senomorphics adopt two major therapeutic strategies [215], targeting the SASP-associated pathways (PI3k/Akt, JAK/STAT, and mTOR pathways, etc.) and transcription factors (NF-κB, C/EBP β, and STAT3 factors, etc.) [226], and neutralizing the SASP factors, including inflammatory cytokines and matrix-degrading molecules [226]. For example, monoclonal antibodies against IL-6, NF-κB, and IL-8 effectively alleviate SASP-induced chronic inflammation [226, 227]. However, senomorphics display apparent defects [215]. For example, senomorphics should be administered chronically to achieve sufficient effectiveness [215]. However, long-term senomorphics administration may inhibit pathways and transcription factors essential for maintaining tissue homeostasis [215]. According to previous studies, the most widely studied senomorphics suppress SASP by targeting NF-κB, IL-1α, mTOR, JAK/STAT, and p38MAPK pathways [228, 229].
Rapamycin
Rapamycin isolated from the bacterium Streptomyces hygroscopicus is one of the most widely used senomorphics [230]. Rapamycin was initially developed as an anti-fungal agent and later applied to prevent organ rejection and treat lymphangioleiomyomatosis [231]. Currently, rapamycin is one of the most widely established senomorphics that alleviate cellular senescence and the characteristics of SASP [229]. Rapamycin reduces cellular senescence mainly by inhibiting the mTOR signaling [232]. The mTOR contains two main functional complexes: mTORC1 and mTORC2 [233]. Increased mTORC1 activity has been found to cause phenotypes of cellular senescence, and inhibiting mTORC1 activity is currently one of the best-known pharmacological approaches to attenuate cellular senescence and increase lifespan [234, 235]. As an acute inhibitor of mTORC1, rapamycin inhibits translation and cell growth by inhibiting the phosphorylation of downstream substrates, including eukaryotic translation initiation factor 4E-binding protein 1 (4E-BP1) and S6 kinase 1 (S6K1) [236]. 4E-BP1 and S6K1 are involved in cell growth, proliferation, and migration by regulating mRNA translation and protein synthesis [237, 238]. Although many studies have demonstrated beneficial effects of rapamycin in alleviating AD pathology, including reducing Aβ accumulation [239], tau hyperphosphorylation [240] and neuroinflammation [241], and improving cognitive dysfunction [242], the effects of rapamycin remain controversial. For example, a previous study found that treating AD mice with rapamycin causes reduction of Aβ clearance [243]. Therefore, more studies investigating the effects of rapamycin on AD are warranted.
Metformin
Metformin is another widely studied senomorphics, which was initially developed for treating type 2 diabetes [229]. Metformin alleviates cellular senescence and SASP by inhibiting nuclear translocation of NF-κB and preventing IκB and IKKα/β phosphorylation, which is AMPK-independent [244]. However, another study found that metformin attenuates stress-induced cellular senescence and restores almost all senescence-related functions in an AMPK-dependent manner [245]. This discrepancy may be caused by differences in the types of cell and senescence. Increasing studies have attempted to repurpose metformin to AD therapy [246,247,248]. Clinical evidence suggests that metformin could reduce the risk of AD in elderly diabetic patients and exert beneficial effects in patients with cognitive impairment [249,250,251]. Moreover, animal studies showed that metformin could alleviate typical AD pathology, including tau pathology, Aβ pathology, neuronal loss and dysfunction, and neuroinflammation [252,253,254,255]. Mechanistically, metformin markedly promotes the expression of neurotrophic factors (e.g., BDNF and NgF), which exert neuroprotective effects and significantly improve synaptic plasticity [256, 257]. Moreover, metformin could attenuate Aβ deposition in AD by increasing insulin-degrading enzyme (an Aβ-degrading peptidase) levels [258]. Metformin also decreases Aβ production by upregulating AMPK expression and suppressing the activation of mTOR, P65 NF-κB, and S6K [254]. As mentioned previously, the activation of mTOR, NF-κB, and S6K is a typical change in cellular senescence. Therefore, the effects of metformin on mTOR, NF-κB, and S6K in AD indicate that metformin could alleviate cellular senescence in AD [254]. The decreased pro-inflammatory cytokines following metformin treatment further support the attenuation of SASP in AD [254]. These findings suggest that metformin, as a widely studied senomorphics, holds a promising therapeutic potential in alleviating cellular senescence and AD pathology.
NF-κB inhibitors
NF-κB is one of the main inducers of SASP [259]. The mammalian NF-κB family has five members: RelA (p65), RelB, c-Rel, NF-κB1 (p50/p105), and NF-κB2 (p52/p100) [260]. There are two types of NF-κB signaling pathways, canonical and noncanonical. Activation of p65 homodimers or c-Rel/p65 heterodimer causes the activation of the canonical NF-κB pathway in response to pro-inflammatory cytokines and bacterial products [260]. The noncanonical pathway is activated in response to stimuli involved in lymphoid organogenesis and causes activation of the p52/RelB heterodimer [261]. According to previous studies, NF-κB plays an indispensable role in cellular senescence [262, 263]. For instance, chronic activation of the canonical IKK/NF-κB signaling pathway and p65 phosphorylation increases the expression of neuroinflammatory markers and the senescence of oligodendrocytes, leading to white matter loss and neurological deficits [262]. Several signaling pathways are involved in the activation of NF-κB signaling and the induction of SASP, including DNA damage, p38MAPK signaling pathway, RIG-1 signaling pathway, TGF-β-TAK1 pathway, HMGB1 proteins, and ceramide signaling [259, 264]. Ataxia-telangiectasia mutated (ATM) kinase, a critical protein involved in DDR and DNA repair, causes activation of NF-κB and induces NF-κB-dependent, DNA damage-induced senescence [264]. Therefore, NF-κB inhibitors are potential senomorphic agents to inhibit SASP [226, 244]. Monoclonal antibodies or metformin can inhibit NF-κB and alleviate SASP-induced chronic inflammation [226].
ATM inhibitors
ATM, an essential protein involved in DNA damage and repair, promotes cellular senescence. ATM inhibitors can counteract cellular senescence [264]. In addition to inducing cellular senescence by activating NF-κB, ATM also disrupts the removal of defective mitochondria by autophagy [265, 266]. The accumulation of defective mitochondria induces inflammatory response and metabolic dysfunction, exacerbating cellular senescence [265, 267]. In contrast, ATM inhibition preserves mitochondrial function and promotes metabolic reprogramming, which is essential in alleviating cellular senescence during aging [268]. In a previous study, ATM depletion attenuates SASP in cellular senescence by reducing secretion of a large subset of major pro-inflammatory cytokines, including IL-6 and IL-8 [269]. These findings suggest that ATM inhibitors are promising strategies against SASP in aging and aging-associated neurodegenerative disorders [269]. KU-55933 and KU-60019 are the most widely studied ATM inhibitors [268, 270], which attenuate senescent markers and alleviate SASP [264, 268].
p38MAPK inhibitors
p38MAPK is a member of the mitogen-activated protein kinase (MAPK) family, and can be rapidly activated by phosphorylation in response to DNA damage, pro-inflammatory cytokines, and other extracellular stress [271]. p38MAPK activation is essential in stress-induced cellular senescence [218, 272]. For example, X-irradiation-induced genotoxic stress significantly increases p38MAPK phosphorylation and cellular senescence, wherein p38MAPK activation is necessary for SASP and the secretion of some SASP components [218]. Constitutive p38MAPK activation is sufficient to induce arrest of cell proliferation, increased expression of cellular senescence markers (e.g., SA-β gal activity), and SASP [218]. In contrast, p38MAPK inhibition alleviates cellular senescence and mitigates SASP component levels [218]. Moreover, further analysis revealed that the role of p38MAPK in cellular senescence and SASP is primarily achieved by increasing NF‐κB transcriptional activity [218]. Notably, although p38MAPK activation is involved in DNA damage-induced cellular senescence, p38MAPK activation induces SASP independently of the DDR signaling [218].
Other senomorphics
As mentioned, monoclonal antibodies against SASP-released inflammatory cytokines could alleviate SASP by neutralizing the released pro-inflammatory cytokines [226, 227]. Likewise, the non-steroidal anti-inflammatory drug Aspirin effectively reduces cellular senescence [125, 273]. For example, Aspirin could alleviate Aβ-induced cell senescence and DNA damage in human neurons and neural stem cells by rescuing SIRT1, suggesting Aspirin as a potential senomorphic drug in alleviating cellular senescence in AD [125, 274]. Moreover, rescuing SIRT1 has been shown to rescue Aβ-induced neuronal senescence and alleviate senescence-associated DNA damage. Additionally, the JAK-STAT signaling pathway, which is involved in the production of inflammatory cytokines, plays a critical role in cellular senescence and SASP [219, 220, 275]. Consistently, numerous studies confirmed that JAK/STAT inhibitors, including AG490, momelotinib, and INCB18424, could alleviate cellular senescence and SASP [219, 220]. In addition, senescent cells display increased mitochondrial and cytosolic Ca2+ levels, and Ca2+ channel inhibitors alleviate the intracellular Ca2+ rise-induced exacerbation of cellular senescence [276, 277]. Furthermore, there is additional evidence suggesting that the ERK pathway inhibitor simvastatin can suppress cellular senescence, although more evidence is needed to clarify its role in cellular senescence within the CNS [278].
Conclusion
Cellular senescence in the brain is influenced by multiple factors, including mitochondrial dysfunction, oxidative stress, neuroinflammation, altered proteostasis, DNA damage, taupathy, Aβ accumulation, telomerase deficiency, telomere shortening, aging, and other stressors such as brain injury (Fig. 2). These factors lead to stress-induced senescence and replicative senescence. Moreover, the factors inducing cellular senescence contribute to cellular aging in AD, affecting various cell types including astrocytes, microglia, OPCs, neurons, and endothelial cells [145, 173, 176, 200]. With the progression of AD, the accumulated senescent cells acquire SASP and secrete numerous chemokines and pro-inflammatory cytokines, including pro-inflammatory factors, ROS, and matrix-degrading enzymes, to exacerbate AD pathology [279]. In addition, the loss of normal functions of the senescent cells in the brain tissue induces synaptic dysfunction, impaired remyelination, BBB breakdown, impaired remyelination, and impaired removal of neuronal debris, metabolic waste, and cell fragments [13, 109, 280, 281] (Fig. 2). Several strategies have been proposed to alleviate cellular senescence or SASP, including promoting apoptosis of senescent cells [7, 282], inhibiting oxidative stress [283], suppressing the cellular senescent-associated pathways [218, 268], destroying senescent cells using oncolytic viruses [284], or using monoclonal antibodies against SASP-released cytokines [226, 227]. Targeting cellular senescence with senotherapeutics has emerged as a promising approach to mitigating AD progression [125, 225, 274]. Senolytic drugs that selectively eliminate senescent cells, and senomorphic agents that suppress SASP without inducing cell death, are being investigated for their potential to alleviate AD pathology and improve cognitive function [216, 285]. Although increasing preclinical studies have demonstrated the efficacy of senotherapeutics in ameliorating AD-related phenotypes in animal models [7, 215], the safety and effectiveness of senotherapeutic interventions in human patients remain unclear. In addition, while this review offers an in-depth examination of these topics, we also acknowledge the limitations and gaps that remain in the current literature. For instance, not all instances of cellular senescence may occur simultaneously across cell types, and there is currently no direct evidence linking cellular senescence to the pathogenic mechanisms of AD. Further research is still needed to elucidate the specific mechanisms by which cellular senescence contributes to AD pathogenesis, and to optimize the safety and efficacy of senotherapeutic interventions in AD patients.
Summary of cellular senescence in AD. Cellular senescence, including stress-induced senescence and replicative senescence in the brain, is induced by multiple factors. Moreover, the factors inducing cellular senescence affect various cell types in AD, including astrocytes, microglia, oligodendrocyte precursor cells (OPCs), neurons, and endothelial cells. The senescent cells further exacerbate AD pathology. Senotherapeutics, which include senolytics and senomorphics, are strategies to alleviate cellular senescence. Senolytics promote the apoptosis of senescent cells, clearing them from the system, while senomorphics mitigate the SASP (senescence-associated secretory phenotype), thereby attenuating cellular senescence. Clearing cellular senescence holds promise for alleviating AD pathology
Moreover, it should be noted that while cellular senescence is associated with various pathological pathways and brain cell types in AD, the impact of senescence is not uniform across these cell types. Senescent astrocytes, microglia, neurons, OPCs, and endothelial cells exhibit distinct roles in AD pathology, contributing to disease progression through unique mechanisms. This highlights the complexity of senescence-related changes in AD and underscores the need for targeted approaches considering the diverse roles of senescent cells across the brain. In addition, as mentioned previously, aging is a critical state that increases the risk of developing AD. Cellular senescence associated with aging can lead to cumulative effects on brain health, contributing to neurodegenerative processes. The age-related senescent changes can exacerbate AD pathology by impairing neuronal function, increasing neuroinflammation, and disrupting homeostasis in the brain. Therefore, it is crucial to consider the interplay between aging and cellular senescence in the context of AD to fully understand its pathogenesis and identify potential therapeutic targets.
Finally, the relationship between cellular senescence and AD is complex, as the observed changes in senescent cells may be both causes and effects of AD pathogenesis. This bidirectional relationship complicates our understanding of the underlying mechanisms. Therapeutic strategies targeting cellular senescence in AD are still under investigation. Future research is essential to elucidate the causal relationships between cellular senescence and AD, which may provide insights into more effective therapeutic approaches.
Limitations
While this review discusses the potential role of cellular senescence in AD pathology, it is important to acknowledge several limitations. First, there is currently a lack of experimental evidence demonstrating a clear causal relationship between cellular senescence and AD progression in human patients. Second, this narrative review does not follow a systematic review methodology, and no formal search strategy was employed. This may introduce selection bias in the sources cited and potentially limit the comprehensiveness of the literature reviewed. It is also important to note that while this narrative review focuses primarily on cellular senescence and apoptosis, other mechanisms, such as aberrant re-entry into the cell cycle, may also play a significant role in AD pathology. Finally, this review highlights several key factors that contribute to cellular senescence, such as oxidative stress, mitochondrial dysfunction, and telomere shortening; however, the relative weight and interactions of these factors in different cell types in the context of AD remain unclear. Further research is required to delineate these complex interactions and how they vary across different stages of AD.
Availability of data and material
Not applicable.
Abbreviations
- AD:
-
Alzheimer’s disease
- OPC:
-
Oligodendrocyte precursor cell
- SASP:
-
Senescence-associated secretory phenotype
- Aβ:
-
Beta-amyloid
- SIPS:
-
Stress-induced premature senescence
- SA-β-gal:
-
Senescence-associated beta-galactosidase
- NSC:
-
Neural stem cell
- CNS:
-
Central nervous system
- NFT:
-
Neurofibrillary tangle
- BBB:
-
Blood-brain barrier
- ROS:
-
Reactive oxygen species
- NAD + :
-
Nicotinamide adenine dinucleotide
- mTORC1:
-
Rapamycin complex 1
- mTOR:
-
Mammalian target of rapamycin
- DDR:
-
DNA damage response
- CDK2:
-
Cyclin-dependent kinase 2
- Rb:
-
Retinoblastoma
- HMGB1:
-
High mobility group box 1
- APP:
-
Amyloid precursor protein
- TERT:
-
Telomerase reverse transcriptase
- DQ:
-
Dasatinib and quercetin
- MAPK:
-
Mitogen-activated protein kinase
References
Hayflick L. The limited in vitro lifetime of human diploid cell strains. Exp Cell Res. 1965;37:614–36.
Hayflick L, Moorhead PS. The serial cultivation of human diploid cell strains. Exp Cell Res. 1961;25:585–621.
Herranz N, Gil J. Mechanisms and functions of cellular senescence. J Clin Invest. 2018;128:1238–46.
Baz-Martinez M, Da Silva-Alvarez S, Rodriguez E, Guerra J, El Motiam A, Vidal A, et al. Cell senescence is an antiviral defense mechanism. Sci Rep. 2016;6:37007.
Aunan JR, Watson MM, Hagland HR, Soreide K. Molecular and biological hallmarks of ageing. Br J Surg. 2016;103:e29-46.
Masaldan S, Belaidi AA, Ayton S, Bush AI. Cellular senescence and iron dyshomeostasis in Alzheimer’s disease. Pharmaceuticals (Basel). 2019;12:93.
Zhang P, Kishimoto Y, Grammatikakis I, Gottimukkala K, Cutler RG, Zhang S, et al. Senolytic therapy alleviates Abeta-associated oligodendrocyte progenitor cell senescence and cognitive deficits in an Alzheimer’s disease model. Nat Neurosci. 2019;22:719–28.
Coppe JP, Desprez PY, Krtolica A, Campisi J. The senescence-associated secretory phenotype: the dark side of tumor suppression. Annu Rev Pathol. 2010;5:99–118.
Guerreiro R, Bras J. The age factor in Alzheimer’s disease. Genome Med. 2015;7:106.
Bussian TJ, Aziz A, Meyer CF, Swenson BL, van Deursen JM, Baker DJ. Clearance of senescent glial cells prevents tau-dependent pathology and cognitive decline. Nature. 2018;562:578–82.
Cao J, Hou J, Ping J, Cai D. Advances in developing novel therapeutic strategies for Alzheimer’s disease. Mol Neurodegener. 2018;13:64.
Yiannopoulou KG, Papageorgiou SG. Current and future treatments for Alzheimer’s disease. Ther Adv Neurol Disord. 2013;6:19–33.
Bhat R, Crowe EP, Bitto A, Moh M, Katsetos CD, Garcia FU, et al. Astrocyte senescence as a component of Alzheimer’s disease. PLoS ONE. 2012;7:e45069.
Hu Y, Huang Y, Xing S, Chen C, Shen D, Chen J. Abeta promotes CD38 expression in senescent microglia in Alzheimer’s disease. Biol Res. 2022;55:10.
Liu RM. Aging, cellular senescence, and Alzheimer’s disease. Int J Mol Sci. 2022;23:1989.
Campisi J. The biology of replicative senescence. Eur J Cancer. 1997;33:703–9.
Serrano M, Blasco MA. Putting the stress on senescence. Curr Opin Cell Biol. 2001;13:748–53.
Herbig U, Jobling WA, Chen BP, Chen DJ, Sedivy JM. Telomere shortening triggers senescence of human cells through a pathway involving ATM, p53, and p21(CIP1), but not p16(INK4a). Mol Cell. 2004;14:501–13.
El Maï M, Marzullo M, Pimenta I, de Castro M, Ferreira G. Opposing p53 and mTOR/AKT promote an in vivo switch from apoptosis to senescence upon telomere shortening in zebrafish. Elife. 2020:9:e54935.
Lorenzini A, Sell C. Enhanced stress-induced senescence-response may increase species lifespan. Aging (Albany NY). 2021;13:15694–6.
Coleman PR, Hahn CN, Grimshaw M, Lu Y, Li X, Brautigan PJ, et al. Stress-induced premature senescence mediated by a novel gene, SENEX, results in an anti-inflammatory phenotype in endothelial cells. Blood. 2010;116:4016–24.
Wang Z, Wei D, Xiao H. Methods of cellular senescence induction using oxidative stress. Methods Mol Biol. 2013;1048:135–44.
Kim CO, Huh AJ, Han SH, Kim JM. Analysis of cellular senescence induced by lipopolysaccharide in pulmonary alveolar epithelial cells. Arch Gerontol Geriatr. 2012;54:e35-41.
Martic I, Wedel S, Jansen-Durr P, Cavinato M. A new model to investigate UVB-induced cellular senescence and pigmentation in melanocytes. Mech Ageing Dev. 2020;190:111322.
de Magalhaes JP, Passos JF. Stress, cell senescence and organismal ageing. Mech Ageing Dev. 2018;170:2–9.
Vaziri H, Benchimol S. From telomere loss to p53 induction and activation of a DNA-damage pathway at senescence: the telomere loss/DNA damage model of cell aging. Exp Gerontol. 1996;31:295–301.
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.
Hernandez-Segura A, Nehme J, Demaria M. Hallmarks of cellular senescence. Trends Cell Biol. 2018;28:436–53.
Campisi J. Aging, cellular senescence, and cancer. Annu Rev Physiol. 2013;75:685–705.
Salminen A, Ojala J, Kaarniranta K. Apoptosis and aging: increased resistance to apoptosis enhances the aging process. Cell Mol Life Sci. 2011;68:1021–31.
Yosef R, Pilpel N, Tokarsky-Amiel R, Biran A, Ovadya Y, Cohen S, et al. Directed elimination of senescent cells by inhibition of BCL-W and BCL-XL. Nat Commun. 2016;7:11190.
Baar MP, Brandt RMC, Putavet DA, Klein JDD, Derks KWJ, Bourgeois BRM, et al. Targeted apoptosis of senescent cells restores tissue homeostasis in response to chemotoxicity and aging. Cell. 2017;169(132–147): e116.
Cuollo L, Antonangeli F, Santoni A, Soriani A. The senescence-associated secretory phenotype (SASP) in the challenging future of cancer therapy and age-related diseases. Biology (Basel). 2020;9:485.
Yang L, Wu C, Parker E, Li Y, Dong Y, Tucker L, et al. Non-invasive photobiomodulation treatment in an Alzheimer Disease-like transgenic rat model. Theranostics. 2022;12:2205–31.
d’Adda di Fagagna F. Living on a break: cellular senescence as a DNA-damage response. Nat Rev Cancer. 2008;8:512–22.
Wang C, Jurk D, Maddick M, Nelson G, Martin-Ruiz C, von Zglinicki T. DNA damage response and cellular senescence in tissues of aging mice. Aging Cell. 2009;8:311–23.
von Zglinicki T, Saretzki G, Ladhoff J, d’Adda di Fagagna F, Jackson SP. Human cell senescence as a DNA damage response. Mech Ageing Dev. 2005;126:111–7.
Dessy E, Rossi E, Berenzi A, Tironi A, Benetti A, Grigolato P. Chromosome 9 instability and alterations of p16 gene in squamous cell carcinoma of the lung and in adjacent normal bronchi: FISH and immunohistochemical study. Histopathology. 2008;52:475–82.
Rayess H, Wang MB, Srivatsan ES. Cellular senescence and tumor suppressor gene p16. Int J Cancer. 2012;130:1715–25.
Li J, Poi MJ, Tsai MD. Regulatory mechanisms of tumor suppressor P16(INK4A) and their relevance to cancer. Biochemistry. 2011;50:5566–82.
Bruce JL, Hurford RK Jr, Classon M, Koh J, Dyson N. Requirements for cell cycle arrest by p16INK4a. Mol Cell. 2000;6:737–42.
Khamchun S, Thongboonkerd V. Cell cycle shift from G0/G1 to S and G2/M phases is responsible for increased adhesion of calcium oxalate crystals on repairing renal tubular cells at injured site. Cell Death Discov. 2018;4:106.
Bohnsack BL, Hirschi KK. Nutrient regulation of cell cycle progression. Annu Rev Nutr. 2004;24:433–53.
Anzola M, Cuevas N, Lopez-Martinez M, Martinez de Pancorbo M, Burgos JJ. p16INK4A gene alterations are not a prognostic indicator for survival in patients with hepatocellular carcinoma undergoing curative hepatectomy. J Gastroenterol Hepatol. 2004;19:397–405.
Giacinti C, Giordano A. RB and cell cycle progression. Oncogene. 2006;25:5220–7.
Lee C, Chang JH, Lee HS, Cho Y. Structural basis for the recognition of the E2F transactivation domain by the retinoblastoma tumor suppressor. Genes Dev. 2002;16:3199–212.
D’Arcangelo D, Tinaburri L, Dellambra E. The role of p16(INK4a) pathway in human epidermal stem cell self-renewal, aging and cancer. Int J Mol Sci. 2017;18:15915.
Patil P, Dong Q, Wang D, Chang J, Wiley C, Demaria M, et al. Systemic clearance of p16(INK4a) -positive senescent cells mitigates age-associated intervertebral disc degeneration. Aging Cell. 2019;18:e12927.
Baker DJ, Wijshake T, Tchkonia T, LeBrasseur NK, Childs BG, van de Sluis B, et al. Clearance of p16Ink4a-positive senescent cells delays ageing-associated disorders. Nature. 2011;479:232–6.
Sharpless NE, Sherr CJ. Forging a signature of in vivo senescence. Nat Rev Cancer. 2015;15:397–408.
Chen X, Zhang W, Gao YF, Su XQ, Zhai ZH. Senescence-like changes induced by expression of p21(waf1/Cip1) in NIH3T3 cell line. Cell Res. 2002;12:229–33.
El-Deiry WS, Tokino T, Velculescu VE, Levy DB, Parsons R, Trent JM, et al. WAF1, a potential mediator of p53 tumor suppression. Cell. 1993;75(4):817–25.
Tuder RM, Yun JH, Graham BB. Cigarette smoke triggers code red: p21CIP1/WAF1/SDI1 switches on danger responses in the lung. Am J Respir Cell Mol Biol. 2008;39:1–6.
Kritsilis M, Rizou SV, Koutsoudaki PN, Evangelou K, Gorgoulis VG, Papadopoulos D. Ageing, cellular senescence and neurodegenerative disease. Int J Molecul Sci. 2018;19(10):2937.
Piechota M, Sunderland P, Wysocka A, Nalberczak M, Sliwinska MA, Radwanska K, et al. Is senescence-associated beta-galactosidase a marker of neuronal senescence? Oncotarget. 2016;7:81099–109.
Gary RK, Kindell SM. Quantitative assay of senescence-associated beta-galactosidase activity in mammalian cell extracts. Anal Biochem. 2005;343:329–34.
Velasquez ZD, Lopez-Osorio S, Waiger D, Manosalva C, Pervizaj-Oruqaj L, Herold S, et al. Eimeria bovis infections induce G(1) cell cycle arrest and a senescence-like phenotype in endothelial host cells. Parasitology. 2021;148:341–53.
Cahu J, Sola B. A sensitive method to quantify senescent cancer cells. J Vis Exp. 2013. https://doiorg.publicaciones.saludcastillayleon.es/10.3791/50494-v.
Yang NC, Hu ML. A fluorimetric method using fluorescein di-beta-D-galactopyranoside for quantifying the senescence-associated beta-galactosidase activity in human foreskin fibroblast Hs68 cells. Anal Biochem. 2004;325:337–43.
Salmonowicz H, Passos JF. Detecting senescence: a new method for an old pigment. Aging Cell. 2017;16:432–4.
Terman A, Brunk UT. Lipofuscin. Int J Biochem Cell Biol. 2004;36:1400–4.
Evangelou K, Lougiakis N, Rizou SV, Kotsinas A, Kletsas D, Munoz-Espin D, et al. Robust, universal biomarker assay to detect senescent cells in biological specimens. Aging Cell. 2017;16:192–7.
Ishikawa S, Ishikawa F. Proteostasis failure and cellular senescence in long-term cultured postmitotic rat neurons. Aging Cell. 2020;19:e13071.
Macip S, Igarashi M, Fang L, Chen A, Pan ZQ, Lee SW, et al. Inhibition of p21-mediated ROS accumulation can rescue p21-induced senescence. EMBO J. 2002;21:2180–8.
Jaskelioff M, Muller FL, Paik JH, Thomas E, Jiang S, Adams AC, et al. Telomerase reactivation reverses tissue degeneration in aged telomerase-deficient mice. Nature. 2011;469:102–6.
Wu C, Yang L, Feng S, Zhu L, Yang L, Liu TC, et al. Therapeutic non-invasive brain treatments in Alzheimer’s disease: recent advances and challenges. Inflamm Regen. 2022;42:31.
Yang L, Youngblood H, Wu C, Zhang Q. Mitochondria as a target for neuroprotection: role of methylene blue and photobiomodulation. Transl Neurodegener. 2020;9:19.
Yang L, Dong Y, Wu C, Li Y, Guo Y, Yang B, et al. Photobiomodulation preconditioning prevents cognitive impairment in a neonatal rat model of hypoxia-ischemia. J Biophotonics. 2019;12:e201800359.
de la Cueva M, Antequera D, Ordonez-Gutierrez L, Wandosell F, Camins A, Carro E, et al. Amyloid-beta impairs mitochondrial dynamics and autophagy in Alzheimer’s disease experimental models. Sci Rep. 2022;12:10092.
Calkins MJ, Manczak M, Mao P, Shirendeb U, Reddy PH. Impaired mitochondrial biogenesis, defective axonal transport of mitochondria, abnormal mitochondrial dynamics and synaptic degeneration in a mouse model of Alzheimer’s disease. Hum Mol Genet. 2011;20:4515–29.
Miwa S, Kashyap S, Chini E, von Zglinicki T. Mitochondrial dysfunction in cell senescence and aging. J Clin Invest. 2022;132(13):e158447.
Miwa S, Jow H, Baty K, Johnson A, Czapiewski R, Saretzki G, et al. Low abundance of the matrix arm of complex I in mitochondria predicts longevity in mice. Nat Commun. 2014;5:3837.
Park SY, Choi B, Cheon H, Pak YK, Kulawiec M, Singh KK, et al. Cellular aging of mitochondrial DNA-depleted cells. Biochem Biophys Res Commun. 2004;325:1399–405.
Correia-Melo C, Marques FD, Anderson R, Hewitt G, Hewitt R, Cole J, et al. Mitochondria are required for pro-ageing features of the senescent phenotype. EMBO J. 2016;35:724–42.
Rizza S, Cardaci S, Montagna C, Di Giacomo G, De Zio D, Bordi M, et al. S-nitrosylation drives cell senescence and aging in mammals by controlling mitochondrial dynamics and mitophagy. Proc Natl Acad Sci U S A. 2018;115:E3388–97.
Passos JF, Saretzki G, Ahmed S, Nelson G, Richter T, Peters H, et al. Mitochondrial dysfunction accounts for the stochastic heterogeneity in telomere-dependent senescence. PLoS Biol. 2007;5:e110.
Noh SE, Lee SJ, Lee TG, Park KS, Kim JH. Inhibition of cellular senescence hallmarks by mitochondrial transplantation in senescence-induced ARPE-19 cells. Neurobiol Aging. 2023;121:157–65.
Wei W, Ji S. Cellular senescence: Molecular mechanisms and pathogenicity. J Cell Physiol. 2018;233:9121–35.
Eroglu B, Genova E, Zhang Q, Su Y, Shi X, Isales C, et al. Photobiomodulation has rejuvenating effects on aged bone marrow mesenchymal stem cells. Sci Rep. 2021;11:13067.
Alamro AA, Alsulami EA, Almutlaq M, Alghamedi A, Alokail M, Haq SH. Therapeutic potential of Vitamin D and curcumin in an in vitro model of Alzheimer disease. J Cent Nerv Syst Dis. 2020;12:1179573520924311.
Sun Y, Hu X, Hu G, Xu C, Jiang H. Curcumin attenuates hydrogen peroxide-induced premature senescence via the activation of SIRT1 in human umbilical vein endothelial cells. Biol Pharm Bull. 2015;38:1134–41.
Kwak JY, Ham HJ, Kim CM, Hwang ES. Nicotinamide exerts antioxidative effects on senescent cells. Mol Cells. 2015;38:229–35.
Liu J, Liu Y, Chen J, Hu C, Teng M, Jiao K, et al. The ROS-mediated activation of IL-6/STAT3 signaling pathway is involved in the 27-hydroxycholesterol-induced cellular senescence in nerve cells. Toxicol In Vitro. 2017;45:10–8.
Grazioli S, Pugin J. Mitochondrial damage-associated molecular patterns: from inflammatory signaling to human diseases. Front Immunol. 2018;9:832.
Feng S, Wu C, Zou P, Deng Q, Chen Z, Li M, et al. High-intensity interval training ameliorates Alzheimer’s disease-like pathology by regulating astrocyte phenotype-associated AQP4 polarization. Theranostics. 2023;13:3434–50.
Pan XD, Zhu YG, Lin N, Zhang J, Ye QY, Huang HP, et al. Microglial phagocytosis induced by fibrillar beta-amyloid is attenuated by oligomeric beta-amyloid: implications for Alzheimer’s disease. Mol Neurodegener. 2011;6:45.
Hou Y, Wei Y, Lautrup S, Yang B, Wang Y, Cordonnier S, et al. NAD(+) supplementation reduces neuroinflammation and cell senescence in a transgenic mouse model of Alzheimer’s disease via cGAS-STING. Proc Natl Acad Sci U S A. 2021;118(37):e2011226118.
Morrone Parfitt G, Coccia E, Goldman C, Whitney K, Reyes R, Sarrafha L, et al. Disruption of lysosomal proteolysis in astrocytes facilitates midbrain organoid proteostasis failure in an early-onset Parkinson’s disease model. Nat Commun. 2024;15:447.
Peng Y, Kim MJ, Hullinger R, O’Riordan KJ, Burger C, Pehar M, et al. Improved proteostasis in the secretory pathway rescues Alzheimer’s disease in the mouse. Brain. 2016;139:937–52.
Cui K, Li C, Fang G. Aerobic exercise delays Alzheimer’s disease by regulating mitochondrial proteostasis in the cerebral cortex and hippocampus. Life (Basel). 2023;13:1204.
Cozachenco D, Ribeiro FC, Ferreira ST. Defective proteostasis in Alzheimer’s disease. Ageing Res Rev. 2023;85:101862.
Gourmaud S, Stewart DA, Irwin DJ, Roberts N, Barbour AJ, Eberwine G, et al. The role of mTORC1 activation in seizure-induced exacerbation of Alzheimer’s disease. Brain. 2022;145:324–39.
Zhang Y, Nicholatos J, Dreier JR, Ricoult SJ, Widenmaier SB, Hotamisligil GS, et al. Coordinated regulation of protein synthesis and degradation by mTORC1. Nature. 2014;513:440–3.
Switon K, Kotulska K, Janusz-Kaminska A, Zmorzynska J, Jaworski J. Molecular neurobiology of mTOR. Neuroscience. 2017;341:112–53.
Paccalin M, Pain-Barc S, Pluchon C, Paul C, Besson MN, Carret-Rebillat AS, et al. Activated mTOR and PKR kinases in lymphocytes correlate with memory and cognitive decline in Alzheimer’s disease. Dement Geriatr Cogn Disord. 2006;22:320–6.
Ding Q, Markesbery WR, Chen Q, Li F, Keller JN. Ribosome dysfunction is an early event in Alzheimer’s disease. J Neurosci. 2005;25:9171–5.
Li L, Zhang S, Zhang X, Li T, Tang Y, Liu H, et al. Autophagy enhancer carbamazepine alleviates memory deficits and cerebral amyloid-beta pathology in a mouse model of Alzheimer’s disease. Curr Alzheimer Res. 2013;10:433–41.
Martinez-Cue C, Rueda N. Cellular senescence in neurodegenerative diseases. Front Cell Neurosci. 2020;14:16.
Sabath N, Levy-Adam F, Younis A, Rozales K, Meller A, Hadar S, et al. Cellular proteostasis decline in human senescence. Proc Natl Acad Sci U S A. 2020;117:31902–13.
Foret MK, Orciani C, Welikovitch LA, Huang C, Cuello AC, Do CS. Early oxidative stress and DNA damage in Abeta-burdened hippocampal neurons in an Alzheimer’s-like transgenic rat model. Commun Biol. 2024;7:861.
Ghiasian M, Khamisabadi F, Kheiripour N, Karami M, Haddadi R, Ghaleiha A, et al. Effects of crocin in reducing DNA damage, inflammation, and oxidative stress in multiple sclerosis patients: a double-blind, randomized, and placebo-controlled trial. J Biochem Mol Toxicol. 2019;33:e22410.
Rivera M, Wu Q, Hamerlik P, Hjelmeland AB, Bao S, Rich JN. Acquisition of meiotic DNA repair regulators maintain genome stability in glioblastoma. Cell Death Dis. 2015;6:e1732.
Hou Y, Lautrup S, Cordonnier S, Wang Y, Croteau DL, Zavala E, et al. NAD(+) supplementation normalizes key Alzheimer’s features and DNA damage responses in a new AD mouse model with introduced DNA repair deficiency. Proc Natl Acad Sci U S A. 2018;115:E1876–85.
Wu C, Yang L, Tucker D, Dong Y, Zhu L, Duan R, et al. Beneficial effects of exercise pretreatment in a sporadic Alzheimer’s rat model. Med Sci Sports Exerc. 2018;50:945–56.
Lovell MA, Markesbery WR. Oxidative DNA damage in mild cognitive impairment and late-stage Alzheimer’s disease. Nucleic Acids Res. 2007;35:7497–504.
Hewitt G, Jurk D, Marques FD, Correia-Melo C, Hardy T, Gackowska A, et al. Telomeres are favoured targets of a persistent DNA damage response in ageing and stress-induced senescence. Nat Commun. 2012;3:708.
Schumacher B, Pothof J, Vijg J, Hoeijmakers JHJ. The central role of DNA damage in the ageing process. Nature. 2021;592:695–703.
Tokoda H. Key points in clinical training: at the operating room. Kurinikaru Sutadi. 1989;10:630–1.
Saez-Atienzar S, Masliah E. Cellular senescence and Alzheimer disease: the egg and the chicken scenario. Nat Rev Neurosci. 2020;21:433–44.
Kang C, Xu Q, Martin TD, Li MZ, Demaria M, Aron L, et al. The DNA damage response induces inflammation and senescence by inhibiting autophagy of GATA4. Science. 2015;349(6255):aaa5612.
Farooq U, Saravanan B, Islam Z, Walavalkar K, Singh AK, Jayani RS, et al. An interdependent network of functional enhancers regulates transcription and EZH2 loading at the INK4a/ARF locus. Cell Rep. 2021;34:108898.
Kim WY, Sharpless NE. The regulation of INK4/ARF in cancer and aging. Cell. 2006;127:265–75.
Le O, Palacio L, Bernier G, Batinic-Haberle I, Hickson G, Beausejour C. INK4a/ARF expression impairs neurogenesis in the brain of irradiated mice. Stem Cell Reports. 2018;10:1721–33.
Eto H, Kishi Y, Yakushiji-Kaminatsui N, Sugishita H, Utsunomiya S, Koseki H, et al. The Polycomb group protein Ring1 regulates dorsoventral patterning of the mouse telencephalon. Nat Commun. 2020;11:5709.
Lou Z, Chen J. Cellular senescence and DNA repair. Exp Cell Res. 2006;312:2641–6.
Korte N, Barkaway A, Wells J, Freitas F, Sethi H, Andrews SP, et al. Inhibiting Ca(2+) channels in Alzheimer’s disease model mice relaxes pericytes, improves cerebral blood flow and reduces immune cell stalling and hypoxia. Nat Neurosci. 2024. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41593-024-01753-w.
Kadavath H, Hofele RV, Biernat J, Kumar S, Tepper K, Urlaub H, et al. Tau stabilizes microtubules by binding at the interface between tubulin heterodimers. Proc Natl Acad Sci U S A. 2015;112:7501–6.
Akhmanova A, Kapitein LC. Mechanisms of microtubule organization in differentiated animal cells. Nat Rev Mol Cell Biol. 2022;23:541–58.
Zhang X, Zhang YW, Liu S, Bulloj A, Tong GG, Zhang Z, et al. Tumor suppressor PTEN affects tau phosphorylation: deficiency in the phosphatase activity of PTEN increases aggregation of an FTDP-17 mutant Tau. Mol Neurodegener. 2006;1:7.
Drummond E, Pires G, MacMurray C, Askenazi M, Nayak S, Bourdon M, et al. Phosphorylated tau interactome in the human Alzheimer’s disease brain. Brain. 2020;143:2803–17.
Gaikwad S, Puangmalai N, Bittar A, Montalbano M, Garcia S, McAllen S, et al. Tau oligomer induced HMGB1 release contributes to cellular senescence and neuropathology linked to Alzheimer’s disease and frontotemporal dementia. Cell Rep. 2021;36:109419.
Musi N, Valentine JM, Sickora KR, Baeuerle E, Thompson CS, Shen Q, et al. Tau protein aggregation is associated with cellular senescence in the brain. Aging Cell. 2018;17:e12840.
Hussong SA, Banh AQ, Van Skike CE, Dorigatti AO, Hernandez SF, Hart MJ, et al. Soluble pathogenic tau enters brain vascular endothelial cells and drives cellular senescence and brain microvascular dysfunction in a mouse model of tauopathy. Nat Commun. 2023;14:2367.
Streit WJ, Braak H, Xue QS, Bechmann I. Dystrophic (senescent) rather than activated microglial cells are associated with tau pathology and likely precede neurodegeneration in Alzheimer’s disease. Acta Neuropathol. 2009;118:475–85.
Li Y, Lu J, Hou Y, Huang S, Pei G. Alzheimer’s amyloid-beta accelerates human neuronal cell senescence which could be rescued by sirtuin-1 and aspirin. Front Cell Neurosci. 2022;16:906270.
Son SM, Cha MY, Choi H, Kang S, Choi H, Lee MS, et al. Insulin-degrading enzyme secretion from astrocytes is mediated by an autophagy-based unconventional secretory pathway in Alzheimer disease. Autophagy. 2016;12:784–800.
He N, Jin WL, Lok KH, Wang Y, Yin M, Wang ZJ. Amyloid-beta(1–42) oligomer accelerates senescence in adult hippocampal neural stem/progenitor cells via formylpeptide receptor 2. Cell Death Dis. 2013;4:e924.
Rolyan H, Scheffold A, Heinrich A, Begus-Nahrmann Y, Langkopf BH, Holter SM, et al. Telomere shortening reduces Alzheimer’s disease amyloid pathology in mice. Brain. 2011;134:2044–56.
Chiu WT, Shen SC, Yang LY, Chow JM, Wu CY, Chen YC. Inhibition of HSP90-dependent telomerase activity in amyloid beta-induced apoptosis of cerebral endothelial cells. J Cell Physiol. 2011;226:2041–51.
Azarm K, Bhardwaj A, Kim E, Smith S. Persistent telomere cohesion protects aged cells from premature senescence. Nat Commun. 2020;11:3321.
Palmos AB, Duarte RRR, Smeeth DM, Hedges EC, Nixon DF, Thuret S, et al. Telomere length and human hippocampal neurogenesis. Neuropsychopharmacology. 2020;45:2239–47.
Yu X, Liu MM, Zheng CY, Liu YT, Wang Z, Wang ZY. Telomerase reverse transcriptase and neurodegenerative diseases. Front Immunol. 2023;14:1165632.
Whittemore K, Derevyanko A, Martinez P, Serrano R, Pumarola M, Bosch F, et al. Telomerase gene therapy ameliorates the effects of neurodegeneration associated to short telomeres in mice. Aging (Albany NY). 2019;11:2916–48.
Spilsbury A, Miwa S, Attems J, Saretzki G. The role of telomerase protein TERT in Alzheimer’s disease and in tau-related pathology in vitro. J Neurosci. 2015;35:1659–74.
Guerrero A, De Strooper B, Arancibia-Carcamo IL. Cellular senescence at the crossroads of inflammation and Alzheimer’s disease. Trends Neurosci. 2021;44:714–27.
Blackburn D, Sargsyan S, Monk PN, Shaw PJ. Astrocyte function and role in motor neuron disease: a future therapeutic target? Glia. 2009;57:1251–64.
Chung WS, Allen NJ, Eroglu C. Astrocytes control synapse formation, function, and elimination. Cold Spring Harb Perspect Biol. 2015;7:a020370.
Abbott NJ, Ronnback L, Hansson E. Astrocyte-endothelial interactions at the blood-brain barrier. Nat Rev Neurosci. 2006;7:41–53.
Buosi AS, Matias I, Araujo APB, Batista C, Gomes FCA. Heterogeneity in synaptogenic profile of astrocytes from different brain regions. Mol Neurobiol. 2018;55:751–62.
Taylor X, Cisternas P, You Y, You Y, Xiang S, Marambio Y, et al. A1 reactive astrocytes and a loss of TREM2 are associated with an early stage of pathology in a mouse model of cerebral amyloid angiopathy. J Neuroinflammation. 2020;17:223.
Lye JJ, Latorre E, Lee BP, Bandinelli S, Holley JE, Gutowski NJ, et al. Astrocyte senescence may drive alterations in GFAPalpha, CDKN2A p14(ARF), and TAU3 transcript expression and contribute to cognitive decline. Geroscience. 2019;41:561–73.
Willis CM, Nicaise AM, Bongarzone ER, Givogri M, Reiter CR, Heintz O, et al. astrocyte support for oligodendrocyte differentiation can be conveyed via extracellular vesicles but diminishes with age. Sci Rep. 2020;10:828.
Bitto A, Sell C, Crowe E, Lorenzini A, Malaguti M, Hrelia S, et al. Stress-induced senescence in human and rodent astrocytes. Exp Cell Res. 2010;316:2961–8.
Zhang W, Yang X, Liu J, Pan Y, Zhang M, Chen L. Senescent phenotype of astrocytes leads to activation of BV2 microglia and N2a neuronal cells death. Molecules. 2022;27(18):5925.
Han X, Zhang T, Liu H, Mi Y, Gou X. Astrocyte senescence and Alzheimer’s disease: a review. Front Aging Neurosci. 2020;12:148.
Myung NH, Zhu X, Kruman II, Castellani RJ, Petersen RB, Siedlak SL, et al. Evidence of DNA damage in Alzheimer disease: phosphorylation of histone H2AX in astrocytes. Age (Dordr). 2008;30:209–15.
Gao L, Zheng WG, Wu XK, Du GH, Qin XM. Baicalein delays H(2)O(2)-induced astrocytic senescence through inhibition of senescence-associated secretory phenotype (SASP), suppression of JAK2/STAT1/NF-kappaB pathway, and regulation of leucine metabolism. ACS Chem Neurosci. 2021;12:2320–35.
Birch J, Gil J. Senescence and the SASP: many therapeutic avenues. Genes Dev. 2020;34:1565–76.
Ungerleider K, Beck J, Lissa D, Turnquist C, Horikawa I, Harris BT, et al. Astrocyte senescence and SASP in neurodegeneration: tau joins the loop. Cell Cycle. 2021;20:752–64.
Gonzalez-Reyes RE, Nava-Mesa MO, Vargas-Sanchez K, Ariza-Salamanca D, Mora-Munoz L. Involvement of astrocytes in alzheimer’s disease from a neuroinflammatory and oxidative stress perspective. Front Mol Neurosci. 2017;10:427.
Turnquist C, Horikawa I, Foran E, Major EO, Vojtesek B, Lane DP, et al. p53 isoforms regulate astrocyte-mediated neuroprotection and neurodegeneration. Cell Death Differ. 2016;23:1515–28.
Davis N, Mota BC, Stead L, Palmer EOC, Lombardero L, Rodriguez-Puertas R, et al. Pharmacological ablation of astrocytes reduces Abeta degradation and synaptic connectivity in an ex vivo model of Alzheimer’s disease. J Neuroinflammation. 2021;18:73.
Yabluchanskiy A, Tarantini S, Balasubramanian P, Kiss T, Csipo T, Fulop GA, et al. Pharmacological or genetic depletion of senescent astrocytes prevents whole brain irradiation-induced impairment of neurovascular coupling responses protecting cognitive function in mice. Geroscience. 2020;42:409–28.
Hu Y, Fryatt GL, Ghorbani M, Obst J, Menassa DA, Martin-Estebane M, et al. Replicative senescence dictates the emergence of disease-associated microglia and contributes to Abeta pathology. Cell Rep. 2021;35:109228.
Yang L, Dong Y, Wu C, Youngblood H, Li Y, Zong X, et al. Effects of prenatal photobiomodulation treatment on neonatal hypoxic ischemia in rat offspring. Theranostics. 2021;11:1269–94.
Ding X, Wang J, Huang M, Chen Z, Liu J, Zhang Q, et al. Loss of microglial SIRPalpha promotes synaptic pruning in preclinical models of neurodegeneration. Nat Commun. 2021;12:2030.
Leng F, Edison P. Neuroinflammation and microglial activation in Alzheimer disease: Where do we go from here? Nat Rev Neurol. 2021;17:157–72.
Gong Q, Wang Y, Wang X, Pan H, Yan C. Baicalein promotes the microglia M2 polarization and suppresses apoptosis by targeting HMOX1/PDE4D to alleviate Alzheimer’s disease. Immunobiology. 2023;228:152761.
Ransohoff RM. A polarizing question: do M1 and M2 microglia exist? Nat Neurosci. 2016;19:987–91.
Pettas S, Karagianni K, Kanata E, Chatziefstathiou A, Christoudia N, Xanthopoulos K, et al. Profiling microglia through single-cell RNA sequencing over the course of development, aging, and disease. Cells. 2022;11:2383.
Olah M, Menon V, Habib N, Taga MF, Ma Y, Yung CJ, et al. Single cell RNA sequencing of human microglia uncovers a subset associated with Alzheimer’s disease. Nat Commun. 2020;11:6129.
Deczkowska A, Keren-Shaul H, Weiner A, Colonna M, Schwartz M, Amit I. Disease-associated microglia: a universal immune sensor of neurodegeneration. Cell. 2018;173:1073–81.
Malm TM, Jay TR, Landreth GE. The evolving biology of microglia in Alzheimer’s disease. Neurotherapeutics. 2015;12:81–93.
Mandrekar-Colucci S, Karlo JC, Landreth GE. Mechanisms underlying the rapid peroxisome proliferator-activated receptor-gamma-mediated amyloid clearance and reversal of cognitive deficits in a murine model of Alzheimer’s disease. J Neurosci. 2012;32:10117–28.
Shen Z, Bao X, Wang R. Clinical PET imaging of microglial activation: implications for microglial therapeutics in alzheimer’s disease. Front Aging Neurosci. 2018;10:314.
Thomas AL, Lehn MA, Janssen EM, Hildeman DA, Chougnet CA. Naturally-aged microglia exhibit phagocytic dysfunction accompanied by gene expression changes reflective of underlying neurologic disease. Sci Rep. 2022;12:19471.
Podlesny-Drabiniok A, Marcora E, Goate AM. Microglial phagocytosis: a disease-associated process emerging from alzheimer’s disease genetics. Trends Neurosci. 2020;43:965–79.
Iaccarino HF, Singer AC, Martorell AJ, Rudenko A, Gao F, Gillingham TZ, et al. Gamma frequency entrainment attenuates amyloid load and modifies microglia. Nature. 2016;540:230–5.
Lucin KM, O’Brien CE, Bieri G, Czirr E, Mosher KI, Abbey RJ, et al. Microglial beclin 1 regulates retromer trafficking and phagocytosis and is impaired in Alzheimer’s disease. Neuron. 2013;79:873–86.
Ng PY, McNeely TL, Baker DJ. Untangling senescent and damage-associated microglia in the aging and diseased brain. FEBS J. 2023;290:1326–39.
Ritzel RM, Patel AR, Pan S, Crapser J, Hammond M, Jellison E, et al. Age- and location-related changes in microglial function. Neurobiol Aging. 2015;36:2153–63.
Cheng LQ, Zhang ZQ, Chen HZ, Liu DP. Epigenetic regulation in cell senescence. J Mol Med (Berl). 2017;95:1257–68.
Shaerzadeh F, Phan L, Miller D, Dacquel M, Hachmeister W, Hansen C, et al. Microglia senescence occurs in both substantia nigra and ventral tegmental area. Glia. 2020;68:2228–45.
Depp C, Sun T, Sasmita AO, Spieth L, Berghoff SA, Nazarenko T, et al. Myelin dysfunction drives amyloid-beta deposition in models of Alzheimer’s disease. Nature. 2023;618:349–57.
Koellhoffer EC, McCullough LD, Ritzel RM. Old maids: aging and its impact on microglia function. Int J Mol Sci. 2017;18:769.
Vanzulli I, Papanikolaou M, De-La-Rocha IC, Pieropan F, Rivera AD, Gomez-Nicola D, et al. Disruption of oligodendrocyte progenitor cells is an early sign of pathology in the triple transgenic mouse model of Alzheimer’s disease. Neurobiol Aging. 2020;94:130–9.
Dawson MR, Polito A, Levine JM, Reynolds R. NG2-expressing glial progenitor cells: an abundant and widespread population of cycling cells in the adult rat CNS. Mol Cell Neurosci. 2003;24:476–88.
Tsai HH, Niu J, Munji R, Davalos D, Chang J, Zhang H, et al. Oligodendrocyte precursors migrate along vasculature in the developing nervous system. Science. 2016;351:379–84.
van Tilborg E, de Theije CGM, van Hal M, Wagenaar N, de Vries LS, Benders MJ, et al. Origin and dynamics of oligodendrocytes in the developing brain: Implications for perinatal white matter injury. Glia. 2018;66:221–38.
Maki T. Novel roles of oligodendrocyte precursor cells in the developing and damaged brain. Clin Exp Neuroimmunol. 2017;8(1):33–42.
Beiter RM, Rivet-Noor C, Merchak AR, Bai R, Johanson DM, Slogar E, et al. Evidence for oligodendrocyte progenitor cell heterogeneity in the adult mouse brain. Sci Rep. 2022;12:12921.
Seo JH, Miyamoto N, Hayakawa K, Pham LD, Maki T, Ayata C, et al. Oligodendrocyte precursors induce early blood-brain barrier opening after white matter injury. J Clin Invest. 2013;123:782–6.
Neumann B, Baror R, Zhao C, Segel M, Dietmann S, Rawji KS, et al. Metformin restores CNS remyelination capacity by rejuvenating aged stem cells. Cell Stem Cell. 2019;25(473–485):e478.
Lau V, Ramer L, Tremblay ME. An aging, pathology burden, and glial senescence build-up hypothesis for late onset Alzheimer’s disease. Nat Commun. 2023;14:1670.
Yang L, Tucker D, Dong Y, Wu C, Lu Y, Li Y, et al. Photobiomodulation therapy promotes neurogenesis by improving post-stroke local microenvironment and stimulating neuroprogenitor cells. Exp Neurol. 2018;299:86–96.
Xie YY, Pan TT, Xu DE, Huang X, Tang Y, Huang W, et al. Clemastine ameliorates myelin deficits via preventing senescence of oligodendrocytes precursor cells in alzheimer’s disease model mouse. Front Cell Dev Biol. 2021;9:733945.
Cui X, Guo YE, Fang JH, Shi CJ, Suo N, Zhang R, et al. Donepezil, a drug for Alzheimer’s disease, promotes oligodendrocyte generation and remyelination. Acta Pharmacol Sin. 2019;40:1386–93.
Niu J, Tsai HH, Hoi KK, Huang N, Yu G, Kim K, et al. Aberrant oligodendroglial-vascular interactions disrupt the blood-brain barrier, triggering CNS inflammation. Nat Neurosci. 2019;22:709–18.
The principles of nerve cell communication. Alcohol Health Res World. 1997;21:107–8.
Fielder E, von Zglinicki T, Jurk D. The DNA damage response in neurons: Die by apoptosis or survive in a senescence-like state? J Alzheimers Dis. 2017;60:S107–31.
Chauhan M, Modi PK, Sharma P. Aberrant activation of neuronal cell cycle caused by dysregulation of ubiquitin ligase Itch results in neurodegeneration. Cell Death Dis. 2020;11:441.
Yang Y, Herrup K. Cell division in the CNS: protective response or lethal event in post-mitotic neurons? Biochim Biophys Acta. 2007;1772:457–66.
Herrup K, Yang Y. Cell cycle regulation in the postmitotic neuron: Oxymoron or new biology? Nat Rev Neurosci. 2007;8:368–78.
Herdy JR, Traxler L, Agarwal RK, Karbacher L, Schlachetzki JCM, Boehnke L, et al. Increased post-mitotic senescence in aged human neurons is a pathological feature of Alzheimer’s disease. Cell Stem Cell. 2022;29(1637–1652):e1636.
Sahu MR, Rani L, Subba R, Mondal AC. Cellular senescence in the aging brain: a promising target for neurodegenerative diseases. Mech Ageing Dev. 2022;204:111675.
Wengerodt D, Schmeer C, Witte OW, Kretz A. Amitosenescence and pseudomitosenescence: putative new players in the aging process. Cells. 2019;8:1546.
Bigagli E, Luceri C, Scartabelli T, Dolara P, Casamenti F, Pellegrini-Giampietro DE, et al. Long-term neuroglial cocultures as a brain aging model: hallmarks of senescence, microrna expression profiles, and comparison with in vivo models. J Gerontol A Biol Sci Med Sci. 2016;71:50–60.
Jurk D, Wang C, Miwa S, Maddick M, Korolchuk V, Tsolou A, et al. Postmitotic neurons develop a p21-dependent senescence-like phenotype driven by a DNA damage response. Aging Cell. 2012;11:996–1004.
Dehkordi SK, Walker J, Sah E, Bennett E, Atrian F, Frost B, et al. Profiling senescent cells in human brains reveals neurons with CDKN2D/p19 and tau neuropathology. Nat Aging. 2021;1:1107–16.
Wei Z, Chen XC, Song Y, Pan XD, Dai XM, Zhang J, et al. Amyloid beta protein aggravates neuronal senescence and cognitive deficits in 5XFAD mouse model of alzheimer’s disease. Chin Med J (Engl). 2016;129:1835–44.
Chen MB, Yang AC, Yousef H, Lee D, Chen W, Schaum N, et al. Brain endothelial cells are exquisite sensors of age-related circulatory cues. Cell Rep. 2020;30(4418–4432):e4414.
Matsui Y, Muramatsu F, Nakamura H, Noda Y, Matsumoto K, Kishima H, et al. Brain-derived endothelial cells are neuroprotective in a chronic cerebral hypoperfusion mouse model. Commun Biol. 2024;7:338.
Wang H, Yang H, Shi Y, Xiao Y, Yin Y, Jiang B, et al. Reconstituting neurovascular unit with primary neural stem cells and brain microvascular endothelial cells in three-dimensional matrix. Brain Pathol. 2021;31:e12940.
Sah E, Krishnamurthy S, Ahmidouch MY, Gillispie GJ, Milligan C, Orr ME. The cellular senescence stress response in post-mitotic brain cells: cell survival at the expense of tissue degeneration. Life (Basel). 2021;11:229.
Uprety A, Kang Y, Kim SY. Blood-brain barrier dysfunction as a potential therapeutic target for neurodegenerative disorders. Arch Pharm Res. 2021;44:487–98.
Tarantini S, Tran CHT, Gordon GR, Ungvari Z, Csiszar A. Impaired neurovascular coupling in aging and Alzheimer’s disease: Contribution of astrocyte dysfunction and endothelial impairment to cognitive decline. Exp Gerontol. 2017;94:52–8.
Ungvari Z, Podlutsky A, Sosnowska D, Tucsek Z, Toth P, Deak F, et al. Ionizing radiation promotes the acquisition of a senescence-associated secretory phenotype and impairs angiogenic capacity in cerebromicrovascular endothelial cells: role of increased DNA damage and decreased DNA repair capacity in microvascular radiosensitivity. J Gerontol A Biol Sci Med Sci. 2013;68:1443–57.
Kiss T, Nyul-Toth A, Balasubramanian P, Tarantini S, Ahire C, DelFavero J, et al. Single-cell RNA sequencing identifies senescent cerebromicrovascular endothelial cells in the aged mouse brain. Geroscience. 2020;42:429–44.
Miyao M, Cicalese S, Kawai T, Cooper HA, Boyer MJ, Elliott KJ, et al. Involvement of senescence and mitochondrial fission in endothelial cell pro-inflammatory phenotype induced by angiotensin II. Int J Mol Sci. 2020;21:3112.
Graves SI, Baker DJ. Implicating endothelial cell senescence to dysfunction in the ageing and diseased brain. Basic Clin Pharmacol Toxicol. 2020;127:102–10.
Ting KK, Coleman P, Kim HJ, Zhao Y, Mulangala J, Cheng NC, et al. Vascular senescence and leak are features of the early breakdown of the blood-brain barrier in Alzheimer’s disease models. Geroscience. 2023;45:3307–31.
Singh Angom R, Wang Y, Wang E, Pal K, Bhattacharya S, Watzlawik JO, et al. VEGF receptor-1 modulates amyloid beta 1–42 oligomer-induced senescence in brain endothelial cells. FASEB J. 2019;33:4626–37.
Sun R, He T, Pan Y, Katusic ZS. Effects of senescence and angiotensin II on expression and processing of amyloid precursor protein in human cerebral microvascular endothelial cells. Aging (Albany NY). 2018;10:100–14.
Garwood CJ, Simpson JE, Al Mashhadi S, Axe C, Wilson S, Heath PR, et al. DNA damage response and senescence in endothelial cells of human cerebral cortex and relation to Alzheimer’s neuropathology progression: a population-based study in the medical research council cognitive function and ageing study (MRC-CFAS) cohort. Neuropathol Appl Neurobiol. 2014;40:802–14.
Lagoumtzi SM, Chondrogianni N. Senolytics and senomorphics: natural and synthetic therapeutics in the treatment of aging and chronic diseases. Free Radic Biol Med. 2021;171:169–90.
Kirkland JL, Tchkonia T. Senolytic drugs: from discovery to translation. J Intern Med. 2020;288:518–36.
Wang Y, Liu L, Zhou D. Inhibition of p38 MAPK attenuates ionizing radiation-induced hematopoietic cell senescence and residual bone marrow injury. Radiat Res. 2011;176:743–52.
Freund A, Patil CK, Campisi J. p38MAPK is a novel DNA damage response-independent regulator of the senescence-associated secretory phenotype. EMBO J. 2011;30:1536–48.
Chen M, Xiao L, Dai G, Lu P, Zhang Y, Li Y, et al. Inhibition of JAK-STAT signaling pathway alleviates age-related phenotypes in tendon stem/progenitor cells. Front Cell Dev Biol. 2021;9:650250.
Xu M, Tchkonia T, Ding H, Ogrodnik M, Lubbers ER, Pirtskhalava T, et al. JAK inhibition alleviates the cellular senescence-associated secretory phenotype and frailty in old age. Proc Natl Acad Sci U S A. 2015;112:E6301-6310.
Fuhrmann-Stroissnigg H, Ling YY, Zhao J, McGowan SJ, Zhu Y, Brooks RW, et al. Identification of HSP90 inhibitors as a novel class of senolytics. Nat Commun. 2017;8:422.
Nieto M, Konisgberg M, Silva-Palacios A. Quercetin and dasatinib, two powerful senolytics in age-related cardiovascular disease. Biogerontology. 2024;25:71–82.
Gulej R, Nyul-Toth A, Ahire C, DelFavero J, Balasubramanian P, Kiss T, et al. Elimination of senescent cells by treatment with Navitoclax/ABT263 reverses whole brain irradiation-induced blood-brain barrier disruption in the mouse brain. Geroscience. 2023;45:2983–3002.
Gonzales MM, Garbarino VR, Kautz TF, Palavicini JP, Lopez-Cruzan M, Dehkordi SK, et al. Senolytic therapy in mild Alzheimer’s disease: a phase 1 feasibility trial. Nat Med. 2023;29:2481–8.
Gonzales MM, Garbarino VR, Marques Zilli E, Petersen RC, Kirkland JL, Tchkonia T, et al. Senolytic therapy to modulate the progression of alzheimer’s disease (SToMP-AD): a pilot clinical trial. J Prev Alzheimers Dis. 2022;9:22–9.
von Kobbe C. Targeting senescent cells: approaches, opportunities, challenges. Aging (Albany NY). 2019;11:12844–61.
Kuilman T, Michaloglou C, Vredeveld LC, Douma S, van Doorn R, Desmet CJ, et al. Oncogene-induced senescence relayed by an interleukin-dependent inflammatory network. Cell. 2008;133:1019–31.
Thompson EL, Pitcher LE, Niedernhofer LJ, Robbins PD. Targeting cellular senescence with senotherapeutics: development of new approaches for skin care. Plast Reconstr Surg. 2022;150:12S-19S.
Zhang L, Pitcher LE, Prahalad V, Niedernhofer LJ, Robbins PD. Targeting cellular senescence with senotherapeutics: senolytics and senomorphics. FEBS J. 2023;290:1362–83.
Vezina C, Kudelski A, Sehgal SN. Rapamycin (AY-22,989), a new antifungal antibiotic. I. Taxonomy of the producing streptomycete and isolation of the active principle. J Antibiot (Tokyo). 1975;28:721–6.
Martel RR, Klicius J, Galet S. Inhibition of the immune response by rapamycin, a new antifungal antibiotic. Can J Physiol Pharmacol. 1977;55:48–51.
Yoon MS. The role of mammalian target of rapamycin (mTOR) in insulin signaling. Nutrients. 2017;9:1176.
Li X, Yang Z, Nie W, Jiang J, Li S, Li Z, et al. Exosomes derived from cardiac progenitor cells attenuate CVB3-induced apoptosis via abrogating the proliferation of CVB3 and modulating the mTOR signaling pathways. Cell Death Dis. 2019;10:691.
Carroll B, Nelson G, Rabanal-Ruiz Y, Kucheryavenko O, Dunhill-Turner NA, Chesterman CC, et al. Persistent mTORC1 signaling in cell senescence results from defects in amino acid and growth factor sensing. J Cell Biol. 2017;216:1949–57.
Joseph GA, Wang SX, Jacobs CE, Zhou W, Kimble GC, Tse HW, et al. Partial inhibition of mTORC1 in aged rats counteracts the decline in muscle mass and reverses molecular signaling associated with sarcopenia. Mol Cell Biol. 2019;39:e00141.
Lamming DW. Inhibition of the mechanistic target of rapamycin (mTOR)-Rapamycin and beyond. Cold Spring Harb Perspect Med. 2016;6:a025924.
Yang L, Miao L, Liang F, Huang H, Teng X, Li S, et al. The mTORC1 effectors S6K1 and 4E-BP play different roles in CNS axon regeneration. Nat Commun. 2014;5:5416.
Zhou H, Huang S. Role of mTOR signaling in tumor cell motility, invasion and metastasis. Curr Protein Pept Sci. 2011;12:30–42.
Chen J, Long Z, Li Y, Luo M, Luo S, He G. Alteration of the Wnt/GSK3beta/beta-catenin signalling pathway by rapamycin ameliorates pathology in an Alzheimer’s disease model. Int J Mol Med. 2019;44:313–23.
Liu Y, Su Y, Wang J, Sun S, Wang T, Qiao X, et al. Rapamycin decreases tau phosphorylation at Ser214 through regulation of cAMP-dependent kinase. Neurochem Int. 2013;62:458–67.
Hou SJ, Zhang SX, Li Y, Xu SY. Rapamycin responds to Alzheimer’s disease: a potential translational therapy. Clin Interv Aging. 2023;18:1629–39.
Spilman P, Podlutskaya N, Hart MJ, Debnath J, Gorostiza O, Bredesen D, et al. Inhibition of mTOR by rapamycin abolishes cognitive deficits and reduces amyloid-beta levels in a mouse model of Alzheimer’s disease. PLoS ONE. 2010;5:e9979.
Shi Q, Chang C, Saliba A, Bhat MA. Microglial mTOR activation upregulates Trem2 and enhances beta-amyloid plaque clearance in the 5XFAD alzheimer’s disease model. J Neurosci. 2022;42:5294–313.
Moiseeva O, Deschenes-Simard X, St-Germain E, Igelmann S, Huot G, Cadar AE, et al. Metformin inhibits the senescence-associated secretory phenotype by interfering with IKK/NF-kappaB activation. Aging Cell. 2013;12:489–98.
Le Pelletier L, Mantecon M, Gorwood J, Auclair M, Foresti R, Motterlini R, et al. Metformin alleviates stress-induced cellular senescence of aging human adipose stromal cells and the ensuing adipocyte dysfunction. Elife. 2021;10:e62635.
Ale Mahmoud Mehraban R, Babaei P, Rohampour K, Jafari A, Golipoor Z. Metformin improves memory via AMPK/mTOR-dependent route in a rat model of Alzheimer’s disease. Iran J Basic Med Sci. 2024;27:360–5.
Cho SY, Kim EW, Park SJ, Phillips BU, Jeong J, Kim H, et al. Reconsidering repurposing: long-term metformin treatment impairs cognition in Alzheimer’s model mice. Transl Psychiatry. 2024;14:34.
Ha J, Choi DW, Kim KJ, Cho SY, Kim H, Kim KY, et al. Association of metformin use with Alzheimer’s disease in patients with newly diagnosed type 2 diabetes: a population-based nested case-control study. Sci Rep. 2021;11:24069.
Zheng J, Xu M, Walker V, Yuan J, Korologou-Linden R, Robinson J, et al. Evaluating the efficacy and mechanism of metformin targets on reducing Alzheimer’s disease risk in the general population: a Mendelian randomisation study. Diabetologia. 2022;65:1664–75.
Sluggett JK, Koponen M, Bell JS, Taipale H, Tanskanen A, Tiihonen J, et al. Metformin and risk of Alzheimer’s disease among community-dwelling people with diabetes: a national case-control study. J Clin Endocrinol Metab. 2020;105:e963.
Campbell JM, Stephenson MD, de Courten B, Chapman I, Bellman SM, Aromataris E. Metformin use associated with reduced risk of dementia in patients with diabetes: a systematic review and meta-analysis. J Alzheimers Dis. 2018;65:1225–36.
Pilipenko V, Narbute K, Pupure J, Langrate IK, Muceniece R, Klusa V. Neuroprotective potential of antihyperglycemic drug metformin in streptozocin-induced rat model of sporadic Alzheimer’s disease. Eur J Pharmacol. 2020;881:173290.
Lu XY, Huang S, Chen QB, Zhang D, Li W, Ao R, et al. Metformin ameliorates Abeta pathology by insulin-degrading enzyme in a transgenic mouse model of Alzheimer’s disease. Oxid Med Cell Longev. 2020;2020:2315106.
Ou Z, Kong X, Sun X, He X, Zhang L, Gong Z, et al. Metformin treatment prevents amyloid plaque deposition and memory impairment in APP/PS1 mice. Brain Behav Immun. 2018;69:351–63.
Chen F, Dong RR, Zhong KL, Ghosh A, Tang SS, Long Y, et al. Antidiabetic drugs restore abnormal transport of amyloid-beta across the blood-brain barrier and memory impairment in db/db mice. Neuropharmacology. 2016;101:123–36.
Wang Y, Zhao J, Guo FL, Gao X, Xie X, Liu S, et al. Metformin ameliorates synaptic defects in a mouse model of AD by inhibiting Cdk5 activity. Front Cell Neurosci. 2020;14:170.
Houshmand F, Barati M, Golab F, Ramezani-Sefidar S, Tanbakooie S, Tabatabaei M, et al. Metformin-induced AMPK activation stimulates remyelination through induction of neurotrophic factors, downregulation of NogoA and recruitment of Olig2+ precursor cells in the cuprizone murine model of multiple sclerosis. Daru. 2019;27:583–92.
Kurochkin IV, Guarnera E, Berezovsky IN. Insulin-degrading enzyme in the fight against alzheimer’s disease. Trends Pharmacol Sci. 2018;39:49–58.
Salminen A, Kauppinen A, Kaarniranta K. Emerging role of NF-kappaB signaling in the induction of senescence-associated secretory phenotype (SASP). Cell Signal. 2012;24:835–45.
Hayden MS, Ghosh S. NF-kappaB, the first quarter-century: remarkable progress and outstanding questions. Genes Dev. 2012;26:203–34.
Sun SC. Non-canonical NF-kappaB signaling pathway. Cell Res. 2011;21:71–85.
Schlett JS, Mettang M, Skaf A, Schweizer P, Errerd A, Mulugeta EA, et al. NF-kappaB is a critical mediator of post-mitotic senescence in oligodendrocytes and subsequent white matter loss. Mol Neurodegener. 2023;18:24.
Rovillain E, Mansfield L, Caetano C, Alvarez-Fernandez M, Caballero OL, Medema RH, et al. Activation of nuclear factor-kappa B signalling promotes cellular senescence. Oncogene. 2011;30:2356–66.
Zhao J, Zhang L, Lu A, Han Y, Colangelo D, Bukata C, et al. ATM is a key driver of NF-kappaB-dependent DNA-damage-induced senescence, stem cell dysfunction and aging. Aging (Albany NY). 2020;12:4688–710.
Kang HT, Park JT, Choi K, Kim Y, Choi HJC, Jung CW, et al. Chemical screening identifies ATM as a target for alleviating senescence. Nat Chem Biol. 2017;13:616–23.
Valentin-Vega YA, Maclean KH, Tait-Mulder J, Milasta S, Steeves M, Dorsey FC, et al. Mitochondrial dysfunction in ataxia-telangiectasia. Blood. 2012;119:1490–500.
Stagni V, Ferri A, Cirotti C, Barila D. ATM kinase-dependent regulation of autophagy: A key player in senescence? Front Cell Dev Biol. 2020;8:599048.
Kuk MU, Kim JW, Lee YS, Cho KA, Park JT, Park SC. Alleviation of senescence via atm inhibition in accelerated aging models. Mol Cells. 2019;42:210–7.
Rodier F, Coppe JP, Patil CK, Hoeijmakers WA, Munoz DP, Raza SR, et al. Persistent DNA damage signalling triggers senescence-associated inflammatory cytokine secretion. Nat Cell Biol. 2009;11:973–9.
Harris BRE, Zhang Y, Tao J, Shen R, Zhao X, Cleary MP, et al. ATM inhibitor KU-55933 induces apoptosis and inhibits motility by blocking GLUT1-mediated glucose uptake in aggressive cancer cells with sustained activation of Akt. FASEB J. 2021;35:e21264.
Koffel R, Meshcheryakova A, Warszawska J, Hennig A, Wagner K, Jorgl A, et al. Monocytic cell differentiation from band-stage neutrophils under inflammatory conditions via MKK6 activation. Blood. 2014;124:2713–24.
Alimbetov D, Davis T, Brook AJ, Cox LS, Faragher RG, Nurgozhin T, et al. Suppression of the senescence-associated secretory phenotype (SASP) in human fibroblasts using small molecule inhibitors of p38 MAP kinase and MK2. Biogerontology. 2016;17:305–15.
Feng M, Kim J, Field K, Reid C, Chatzistamou I, Shim M. Aspirin ameliorates the long-term adverse effects of doxorubicin through suppression of cellular senescence. FASEB Bioadv. 2019;1:579–90.
Li R, Li Y, Zuo H, Pei G, Huang S, Hou Y. Alzheimer’s amyloid-beta accelerates cell senescence and suppresses SIRT1 in human neural stem cells. Biomolecules. 2024;14:189.
Zhang W, Xu M, Chen F, Su Y, Yu M, Xing L, et al. Targeting the JAK2-STAT3 pathway to inhibit cGAS-STING activation improves neuronal senescence after ischemic stroke. Exp Neurol. 2023;368:114474.
Yu X, Li X, Jiang G, Wang X, Chang HC, Hsu WH, et al. Isradipine prevents rotenone-induced intracellular calcium rise that accelerates senescence in human neuroblastoma SH-SY5Y cells. Neuroscience. 2013;246:243–53.
Martin N, Zhu K, Czarnecka-Herok J, Vernier M, Bernard D. Regulation and role of calcium in cellular senescence. Cell Calcium. 2023;110:102701.
Liu S, Uppal H, Demaria M, Desprez PY, Campisi J, Kapahi P. Simvastatin suppresses breast cancer cell proliferation induced by senescent cells. Sci Rep. 2015;5:17895.
Huang W, Hickson LJ, Eirin A, Kirkland JL, Lerman LO. Cellular senescence: the good, the bad and the unknown. Nat Rev Nephrol. 2022;18:611–27.
Zou P, Wu C, Liu TC, Duan R, Yang L. Oligodendrocyte progenitor cells in Alzheimer’s disease: from physiology to pathology. Transl Neurodegener. 2023;12:52.
Bryant AG, Hu M, Carlyle BC, Arnold SE, Frosch MP, Das S, et al. Cerebrovascular senescence is associated with tau pathology in alzheimer’s disease. Front Neurol. 2020;11:575953.
Gasek NS, Kuchel GA, Kirkland JL, Xu M. Strategies for targeting senescent cells in human disease. Nat Aging. 2021;1:870–9.
Faraonio R. Oxidative stress and cell senescence process. Antioxidants (Basel). 2022;11:1718.
Seoane R, Vidal S, Bouzaher YH, El Motiam A, Rivas C. The interaction of viruses with the cellular senescence response. Biology (Basel). 2020;9:455.
Vasil’eva LA, Zotova EG, Aseev D. The role of intracortical inhibition in forming homo- and heterosensory interaction on the neurons of the sensorimotor cortex in kittens. Neirofiziologiia. 1988;20:234–43.
Mohamad Anuar NN, Nor Hisam NS, Liew SL, Ugusman A. Clinical review: navitoclax as a pro-apoptotic and anti-fibrotic agent. Front Pharmacol. 2020;11:564108.
Shen J, Wang Q, Mao Y, Gao W, Duan S. Targeting the p53 signaling pathway in cancers: molecular mechanisms and clinical studies. MedComm. 2020;2023(4):e288.
Chen Q, Sun X, Luo X, Wang J, Hu J, Feng Y. PIK3R3 inhibits cell senescence through p53/p21 signaling. Cell Death Dis. 2020;11:798.
Smoots SG, Schreiber AR, Jackson MM, Bagby SM, Dominguez ATA, Dus ED, et al. Overcoming doxorubicin resistance in triple-negative breast cancer using the class I-targeting HDAC inhibitor bocodepsin/OKI-179 to promote apoptosis. Breast Cancer Res. 2024;26:35.
Park JH, Lee NK, Lim HJ, Ji ST, Kim YJ, Jang WB, et al. Pharmacological inhibition of mTOR attenuates replicative cell senescence and improves cellular function via regulating the STAT3-PIM1 axis in human cardiac progenitor cells. Exp Mol Med. 2020;52:615–28.
Blagosklonny MV. Rapamycin for longevity: opinion article. Aging (Albany NY). 2019;11:8048–67.
Hsu CM, Lin PM, Tsai YT, Tsai MS, Tseng CH, Lin SF, et al. NVP-BEZ235, a dual PI3K-mTOR inhibitor, suppresses the growth of FaDu hypopharyngeal squamous cell carcinoma and has a synergistic effect with Cisplatin. Cell Death Discov. 2018;4:57.
Leontieva OV, Blagosklonny MV. Gerosuppression by pan-mTOR inhibitors. Aging (Albany NY). 2016;8:3535–51.
Tezil T, Chamoli M, Ng CP, Simon RP, Butler VJ, Jung M, et al. Lifespan-increasing drug nordihydroguaiaretic acid inhibits p300 and activates autophagy. NPJ Aging Mech Dis. 2019;5:7.
Demirci D, Dayanc B, Mazi FA, Senturk S. The Jekyll and Hyde of cellular senescence in cancer. Cells. 2021;10:208.
Chauhan A, Ojha R, Semwal DK, Mishra SP, Semwal RB. Combined treatment with CCI779 and SB203580 induces cellular senescence in renal cell carcinoma cell line via p53 pathway. Anticancer Agents Med Chem. 2017;17:1545–54.
Chibaya L, Snyder J, Ruscetti M. Senescence and the tumor-immune landscape: implications for cancer immunotherapy. Semin Cancer Biol. 2022;86:827–45.
Yasuda S, Horinaka M, Iizumi Y, Goi W, Sukeno M, Sakai T. Oridonin inhibits SASP by blocking p38 and NF-kappaB pathways in senescent cells. Biochem Biophys Res Commun. 2022;590:55–62.
van de Veerdonk FL, Renga G, Pariano M, Bellet MM, Servillo G, Fallarino F, et al. Anakinra restores cellular proteostasis by coupling mitochondrial redox balance to autophagy. J Clin Invest. 2022;132(2):e144983.
Gu J, Wang J, Liu X, Sai K, Mai J, Xing F, et al. IL-6 derived from therapy-induced senescence facilitates the glycolytic phenotype in glioblastoma cells. Am J Cancer Res. 2021;11:458–78.
Prattichizzo F, Giuliani A, Recchioni R, Bonafe M, Marcheselli F, De Carolis S, et al. Anti-TNF-alpha treatment modulates SASP and SASP-related microRNAs in endothelial cells and in circulating angiogenic cells. Oncotarget. 2016;7:11945–58.
Griveau A, Wiel C, Ziegler DV, Bergo MO, Bernard D. The JAK1/2 inhibitor ruxolitinib delays premature aging phenotypes. Aging Cell. 2020;19:e13122.
Krzystyniak A, Wesierska M, Petrazzo G, Gadecka A, Dudkowska M, Bielak-Zmijewska A, et al. Combination of dasatinib and quercetin improves cognitive abilities in aged male Wistar rats, alleviates inflammation and changes hippocampal synaptic plasticity and histone H3 methylation profile. Aging (Albany NY). 2022;14:572–95.
Ogrodnik M, Evans SA, Fielder E, Victorelli S, Kruger P, Salmonowicz H, et al. Whole-body senescent cell clearance alleviates age-related brain inflammation and cognitive impairment in mice. Aging Cell. 2021;20:e13296.
Acknowledgements
Not applicable.
Funding
This study was supported by the National Natural Science Foundation of China (32300959 and 32100918), the China Postdoctoral Science Foundation (2021M690060 and 2022T150227), Guangzhou Scientific Research Grant (SL2024A04J00578) and the SCNU Young Faculty Development Program (22KJ04).
Author information
Authors and Affiliations
Contributions
JZ reviewed the literature and drafted the manuscript. JZ, CW, and LY prepared the figures. LY revised the manuscript. All authors read and approved the final manuscript.
Corresponding author
Ethics declarations
Ethics approval and consent to participate
Not applicable.
Consent for publication
Not applicable.
Competing interests
The authors declare that they have no competing interests.
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.
About this article
Cite this article
Zhu, J., Wu, C. & Yang, L. Cellular senescence in Alzheimer’s disease: from physiology to pathology. Transl Neurodegener 13, 55 (2024). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s40035-024-00447-4
Received:
Accepted:
Published:
DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s40035-024-00447-4