Abstract
Purpose of review
Chronic kidney disease (CKD) is often viewed as an accelerated and premature ageing of the kidney, as they share common pathological features characterized by cellular senescence. In this review, we summarize the experimental evidence linking cellular senescence to the pathobiology of kidney ageing and CKD, and discuss the strategies for targeting senescent cells in developing therapeutics for ageing-related kidney disorders.
Recent findings
Kidney ageing and CKD are featured with increased cellular senescence, an irreversible state of cell cycle arrest and the cessation of cell division. Senescent cells secrete a diverse array of proinflammatory and profibrotic factors known as senescence-associated secretory phenotype (SASP). Secondary senescence can be induced by primary senescent cells via a mechanism involving direct contact or the SASP. Various senolytic therapies aiming to selectively remove senescent cells in vivo have been developed. Senostatic approaches to suppress senescence or inhibit SASP, as well as nutrient signaling regulators are also be validated in animal models of ageing.
Summary
These recent studies provide experimental evidence supporting the notion that accumulation of senescent cells and their associated SASP is a main driver leading to structural and functional organ degeneration in CKD and other ageing-related disorder.
Keywords: Cellular senescence, renal ageing, senescence-associated secretory phenotype, chronic kidney disease, senotherapy
INTRODUCTION
In the past several decades, the elderly population has significantly increased worldwide, thanks to the rise of average life expectancy. This imposes an enormous challenge on healthcare systems and has unprecedented health, economic and social implications on a global scale [1,2]. Ageing is associated with structural and functional degeneration in major organs of the body including the kidneys. The incidence and prevalence of chronic kidney disease (CKD) are exceedingly higher in the elderly[3]. Extensive studies have shown that ageing and CKD share many common stressors and have similar underlying mechanisms, such as cellular senescence, oxidative stress, DNA damage, mitochondrial dysfunction, telomere shortening and hyperactive Wnt/β-catenin signaling [4]*.
On cellular level, ageing is characterized by cellular senescence, which is an irreversible state of cell cycle arrest and the cessation of cell division [5]*. Cellular senescence plays a critical role in regulating diverse biological events under different circumstances [6,7]. In general, acute and transient cellular senescence could be beneficial by making positive contributions to many biological processes such as embryogenesis, tissue repair and tumor suppression. However, chronic and sustained senescence after injury inevitably leads to the accumulation of senescent cells and the release of a host of proinflammatory and profibrotic factors known as the senescence-associated secretory phenotype (SASP) [8]. Cellular senescence often precede the manifestation of morphologic lesions and kidney insufficiency, indicating that it is involved in the onset of kidney ageing and the pathogenesis of CKD [9]. In this review, we concisely summarize the experimental evidence linking cellular senescence to the pathobiology of renal ageing and CKD, and discuss recent advances on understanding of the mechanism underlying cellular senescence. We also highlight potential strategies for targeting senescent cells in developing therapeutics for age-related kidney disorders.
MAJOR FEATURES IN THE AGEING KIDNEY
Ageing kidneys manifest significant changes in their morphology and function. On the macrostructural scale, kidney cortical volume decreases with age. Surface roughness develops and grows, and the frequency of forming renal cysts increases during the ageing process. On the microstructural scale, ageing kidneys are characterized by a series of pathological features resembling that found in CKD, such as tubular atrophy, inflammation, interstitial fibrosis, glomerulosclerosis, vascular rarefaction and arteriosclerosis. Kidney ageing is also accompanied by decreased number of the nephrons, with the remaining nephrons being hypertrophic [10]. Estimated glomerular filtration rate (eGFR), approximated by urinary creatinine clearance, declines at approximately 0.75 ml/min/1.73 m2 per year in a longitudinal study of relatively healthy individuals [10,11]. In many aspects, ageing not only leads to renal lesions and functional deficiency but also causes an increased susceptibility to kidney injury [12].
MOUSE MODELS USED IN KIDNEY AGEING
For experimental studies, a suitable animal model is essential for interrogating the key pathways of ageing. The mouse models used in ageing studies could be classified into natural ageing and accelerated ageing (Table 1). Mice with age older than 20 months are often regarded as naturally aged, and they typically present with age-related phenotypes such as muscle weakness, cardiac hypertrophy, reduced cognitive function. Kidneys of the naturally aged mice are manifested with mild glomerulosclerosis, tubular atrophy, inflammation and renal fibrosis, and these lesions get worsened progressively with age (Table 1).
Table1.
Experimental models of ageing
Model | Gross condition | Renal manifestation |
---|---|---|
Naturally aged mice | ||
Naturally aged mice [45,72] | Muscle weakness, cardiac hypertrophy, reduced cognitive function. | Glomerulosclerosis, tubular atrophy, inflammation, renal fibrosis. |
Accelerated aged mice
(chemically and radiation-induced) |
||
D-galactose-induced aged mice [48] | Neurodegeneration, osteoporosis, hearing loss, muscle weakness, weight loss. | Tubular atrophy, inflammation, renal fibrosis. |
γ-Ray irradiation-induced aging mice [73] | Decreased white blood cell, red blood cell and platelet counts. | Renal fib rosis. |
Accelerated aged mice
(genetical modified mice) |
||
SAMP6 [14] | Osteoporosis, increased adipose tissue, hepatic lipidosis. | Renal fibrosis. |
SAMP8 [14] | Neurodegeneration, retinal degeneration, cardiac fibrosis. | Renal fibrosis. |
Klotho null mice [74] | Short lifespan, infertility, growth retardation, reduced cognitive function, arteriosclerosis, osteoporosis, ectopic calcification, skin atrophy and emphysema. | Renal interstitial fibrosis, renal vascular wall thickening, renal ectopic calcification. |
Zmpste24 deficient mice [75] | Growth retardation, alopecia, muscle weakness, rib fractures, dilated cardiomyopathy, muscular dystrophy, lipodystrophy. | Unknow |
Ercc1-/Δ mice [76] | Growth retardation, liver failure, increased mortality, spleen hypoplasia, dystonia, ataxia, sarcopenia, kyphosis, uraemic encephalopathy. | Renal failure, glomerulosclerosis, proteinuria, renal interstitial fibrosis, tubular atrophy. |
Xpd TTD/ TTD mice [77] | Osteoporosis, kyphosis, osteosclerosis, cachexia, infertility, short lifespan. | Impaired renal function, inflammation. |
Abbreviation: SAMP6, senescence-accelerated mouse prone 6; Zmpste24, zinc metalloproteinase Ste24; Ercc1-/Δ, hypomorphic mice with a reduced level of the ERCC1-XPF DNA repair endonuclease; XPD, xeroderma pigmentosum group D.
There are also numerous models with accelerated ageing (Table 1). Mouse models of accelerated ageing could be either induced chemically by D-galactose or irradiation-induced by γ-ray. The D-galactose-induced aged mice display tubular atrophy, renal inflammation and interstitial fibrosis in the kidneys. They also present with premature ageing with neurodegeneration, osteoporosis, hearing loss, muscle weakness and weight loss. The γ-ray irradiation-induced aged mice present with decreased counts of white blood cell, red blood cell and platelets in the circulation, as well as fibrotic lesions in the kidney. These models are characterized by an increased cellular senescence, which undoubtedly contributes to the premature ageing phenotypes in these models.
Another type of accelerated ageing models is induced by gene mutations in the genetically modified mice (Table 1). Of them, Klotho-null mice are the most well characterized and widely used model. Klotho is anti-ageing protein, and its loss in mice is associated with short lifespan, infertility, growth retardation, reduced cognitive function, arteriosclerosis, osteoporosis and ectopic calcification. Renal manifestations of Klotho−/− mice include interstitial fibrosis, renal vascular wall thickening, renal ectopic calcification. Mouse models of progeroid syndrome, a group of rare genetic disorders that mimic physiological aging, are also used in ageing studies [13]. These mice have increased numbers of senescent cells that contribute to premature ageing. For example, the senescence-accelerated mouse prone 6 and 8 (SAMP6 and SAMP8) mice develop a variety of age-associated diseases, such as retinal degeneration, renal fibrosis, immune dysfunction and degenerative joint disorder [14]. Excision repair cross complementing 1 (Ercc1) -/Δ mice, another model of human progeroid syndrome, are impaired in several DNA repair systems and develop age-dependent, all kinds of features of ageing such as motor abnormalities, sarcopenia, cachexia, overall frailty, liver failure, proteinuria and renal fibrosis, and have a shortened life span of 6–7 months. Mice with deficiency of Sirtuin 6 (Sirt6), a NAD+-dependent histone deacetylase and ADP-ribose transferase enzyme, also exhibit premature ageing and suffer from ageing-associated CKD and other degenerative disorders [15]. These studies support the notion that accumulation of DNA damage and genotoxic stress, as well as loss of anti-ageing proteins, contributes to kidney aging and other age-related disorders.
HALLMARKS OF CELLULAR SENESCENCE
There are several hallmarks that could be used to identify cellular senescence. Senescent cells often possess characteristic features of an enlarged size, increased expression of β-galactosidase, accumulation of lipofuscin granules and senescence-associated heterochromatin, among others [16]. Senescent cells also produce and secrete a series of soluble factors known as SASP, which consist of a diverse array of proinflammatory cytokines, chemokines, growth factors and extracellular matrix remodeling factors, such as interleukin-1β (IL-1β), IL-6, IL-8, C-X-C motif chemokine ligand 1 (CXCL1), transforming growth factor-β1 (TGF-β1), plasminogen activator inhibitor-1 (PAI-1) and monocyte chemoattractant protein-1 (MCP-1) [17]. The SASP components could be harnessed for assessing senescence state [18].
The acidic senescence-associated β-galactosidase (SA-β-gal) activity reflects an increased β-galactosidase activity in lysosomes, which becomes the best described and most used marker to identify senescent cells [19,20]. The expression levels of cyclin-dependent kinase (CDK) inhibitors, including p16INK4a (hereafter referred as p16), p21CIP1 (hereafter referred as p21), p19ARF (in mouse), p14ARF (in human), p27KIP1 and p15INK4b are prominently elevated in the senescent cells [4]. Furthermore, accumulation of senescence-associated DNA damage foci (SADF) and senescence-associated heterochromatic foci (SAHF) in the nucleus is also described as senescence biomarkers [21]. Overexpression of SAHF-associated proteins can induce cellular senescence and repress those genes related to cell proliferation [22]. The γ-H2AX, generated by phosphorylation of the histone H2AX, is also defined to reflect the magnitude of senescence [23]. Besides, down-regulation of cell proliferation marker Ki67 and structural nuclear Lamin B1 protein (LMNB1) is also utilized as senescence-associated markers [24].
Recent studies have shown that early growth response 2 (Egr2) is a direct transcriptional activator of the p16/pRb (the retinoblastoma tumor suppressor protein) and p53/p21 pathways, and serves as a novel marker of cellular senescence [25]. In addition, tRNA-derived fragments is a potential biomarker for evaluating senescent cells [26]. The human positive cofactor 4 (PC4) is a sensitive marker and can disrupt proteostasis resulting in age-related organ dysfunction [27]. The histone acetyltransferase KAT7, a genetic and epigenetic marker of senescence, is upregulated in murine models of ageing [28]. Cdkn1a transcript variant 2 is accumulated and activated during ageing and could be used for the diagnosis of ageing-related diseases [29].
It should be stressed that all these biomarkers are neither specific nor unique for senescent cells. In addition, not all cells with these markers exhibit senescent pathology and not all senescent cells express p16 [30]. A single-cell RNA sequencing reveals that cells in response to the same stressors display distinct senescent subtypes [31]. Therefore, multiple biomarkers are recommended to be used in concert for identifying senescent cells in aged kidneys.
DIFFERENT TYPES OF CELL SENESCENCE
Cellular senescence could be divided into two main subtypes, namely acute senescence and chronic senescence. Acute senescence occurs after extracellular cues target a specific group of cells in the tissue. Acute senescence can positively exert beneficial effects by constituting a programmed mechanism that regulates embryogenesis, limits tumorigenesis, promotes wound healing and improves tissue repair [32]. Acute senescent cells are subsequently cleaned by infiltrated macrophage in a timely fashion. When the immune system cannot remove senescent cells at the rate at which senescent cells are being produced, an inevitable outcome is that the accumulation of these cells leads to a disruption in tissue homeostasis. Chronic senescence is typically induced by tissue injury, extracellular stressors or macromolecular damage. As such, chronic senescence usually play a critical role in reducing cell renewal and repopulation, triggering inflammation and fibrotic responses via secreting SASP, thereby promoting CKD progression [8].
Senescent cells, if not be removed appropriately and timely, can propagate and expand via a process causing secondary senescence. Primary senescent cells are induced by detrimental factors such as defective DNA repair, oncogene activation, telomere shortening and others. Primary senescent cells produce distinct paracrine and endocrine signals, leading to other non-senescent cells to undergo secondary senescence. Secondary senescence has been demonstrated to occur in response to SASP secretion through two broad types of mechanisms [33]*. Firstly, SASP factors have been shown to be involved in spreading senescence via a paracrine fashion, which is also called paracrine senescence. Secondly, primary senescent cells are capable of inducing senescence by a direct cell-to-cell contacts, which is defined as juxtacrine senescence. Secondary senescence allows a small number of senescent cells to expand and make it possible to spread to distant locations, leading to age-related diseases.
MECHANISM OF CELLULAR SENESCENCE
The mechanism of cellular senescence remains incompletely understood. There are a wide variety of extracellular stressors and triggers that can induce cellular senescence. These stressors act through diverse mechanisms, eventually leading to permanent cell cycle arrest (Fig. 1).
Figure 1. Major signaling pathways that regulate cellular senescence.
A wide variety of stressors induce cellular senescence via diverse signaling pathways. Many of these stressors such as DNA damage, oxidative stress and nephrotoxins trigger DNA damage response (DDR), leading to activation of ATM/ATR kinases, which induces G2/M phase cell cycle arrest via checkpoint kinase 1 and 2 (CHK1/CHK2) or G1/S phase arrest via p53/p2-mediated inhibition of CDK2. Wnt/β-catenin stimulates p53 and p16 to promote cellular senescence, while anti-ageing proteins Klotho and Sirt1 hamper senescence by blocking these signal pathways. RAAS, renin angiotensin aldosterone system; ATR, ATM and Rad3-related.
Increased expression of cyclin-dependent kinase inhibitors
Numerous signal pathways are involved in cellular senescence, primarily including the p53/p21 and p16 pathways (Fig. 1). The ATM (ataxia telangiectasia mutated) protein kinase, p53, p21 and p16 are activated and induced in response to stress-triggered DNA damage [4]. This cascade of events eventually leads to inhibition of the phosphorylation of cyclin-dependent kinase (CDK) complexes and Rb, resulting in the cessation of cell proliferation and cellular senescence. P53/p21 pathway is thought to be implicated in the onset of cellular senescence, whereas p16 signaling is largely involved in the initiation and maintenance of senescent phenotype [18].
Down-regulation of anti-ageing proteins
There are some intrinsic anti-ageing factors that prevent cellular senescence and premature ageing. Among them, Klotho and Sirt1 are the two most studied proteins. Klotho is an interesting anti-ageing protein that halts cellular senescence. It is a single-pass transmembrane protein and expressed primarily in the kidney [34]. It has been shown that serum level of Klotho decreases with physiological ageing, indicating its potential as a marker of ageing [34]. Mice with Klotho deficiency manifest an accelerated ageing phenotype, characterized by short lifespan, infertility, growth retardation, reduced cognitive function and ectopic calcification. On the contrary, overexpression of Klotho in transgenic mice leads to an increased longevity and prevents ageing-related disorders [35]. Klotho exerts suppressive effects on the p53/p21 and Wnt/β-catenin signaling [36,37]. Klotho also protects against mitochondrial dysfunction in aged cells and CKD by attenuating Wnt/β-catenin signaling [38].
Sirt1, an intrinsic inhibitor of ageing, is capable of deacetylating both histone and non-histone proteins such as FOXO, p53 and NF-κB [39,40]. Therefore, Sirt1 have a broad impact on a variety of key signaling pathways associated with cellular senescence and ageing. It has been reported that podocyte-specific Sirt1 depletion in mice manifests age-related sclerosis in glomerulus [41]. Sirt1 has anti-ageing properties and reverses senescence in endothelial cells, and ablation of Sirt1 in these cells results in accelerated senescence phenotype through acetylating p53. In contrast, overexpression of Sirt1 prevents cellular senescence via restraining p53 activity [42]. Furthermore, it has been shown that sodium tetrasulfide (Na2S4) directly sulfhydrated Sirt1, thereby inhibiting p65 NF-κB and STAT3 phosphorylation/acetylation and alleviating diabetic renal lesions [43].
Other regulatory mechanisms
Recent studies have uncovered several novel signal pathways that play a role in exacerbating renal ageing. For example, Wnt/β-catenin is shown to take part in mediating cellular senescence and age-related renal fibrosis [44,45]. Wnt9a/β-catenin activation is associated with renal tubular senescence and renal fibrosis in diseased kidneys, which is accompanied by an increased expression of p16, p53 and p21, and enhanced SA-β-gal activity in renal tubular epithelia [46]. Recent studies identify the calcium-activated chloride channel accessory 1 (CLCA1) as a novel mediator of kidney injury in aging through TMEM16A/Cl− current pathway [47]. The IκB kinases including IKKε, IKKα and IKKβ are key regulators of cellular senescence and renal ageing, which is regulated by extracellular vesicles (EVs)38. Cannabinoid receptor 2 (CB2) could accelerate mitochondrial damage in renal tubular cells and exerts a detrimental influence on renal ageing [48]. The proper functioning of the circadian clockwork is crucial to prevent ageing. Conversely, ageing reprograms the circadian clock output and disturbs sleep-wake cycles, causing a lowered capacity to synchronize circadian rhythms in peripheral tissues [49,50]. Recent evidence suggests that dysbiosis of intestinal flora and changes of microbial metabolism exhaust reparative potentials in the cell and lead to ageing [51]. Phenotypic discrepancies of senescent cells created by metabolic and epigenomic reprogram may open up new approaches for the treatment of age-related diseases [52].
THERAPEUTIC STRATEGIES FOR KIDNEY AGEING
Given the critical role of cellular senescence in kidney ageing and diseases, it is conceivable that senescence could be used as a therapeutic target for the treatment of CKD. In recent years, novel therapeutic approaches have been developed to halt ageing-associated organ dysfunction via targeting senescent cells [53]. There are three main strategies to pharmacologically regulate senescence for therapeutic intervention. Table 2 lists these three types of anti-ageing remedies that have been validated in a variety of experimental models of ageing, including senolytic, senostatic and nutrient signaling regulators.
Table 2.
Therapeutic strategies targeting cellular senescence.
Senotherapy | Agents |
---|---|
Senolytics | Dasatinib and quercetin [78], fisetin [79], catechins [80], uPAR-targeted CAR-T [58], ABT263 [55], A1331852 [81], A1155463 [81], ABT737 [81], alvespimycin (17-DMAG) [76], FOXO4-DRI [56], panobinostat [82], BPTES [61] |
Senostatics | SS31 [83], klotho [62], mitoQ [84], SKQ1 [85], melatonin [86], ICG001 [45], Na2S4 [43], ganoderma lucidum [87], ruxolitinib [75], MSC-derived EVs [64], SIRT1 activator [65], PPAR-γ agonists [66], metformin [67], rapamycin [88] |
Nutrient regulators | Calorie restriction [69,89], exercise [70], resveratrol [90], curcumin [91], spermidine [92], barbarum [93], DHA [71]. |
Senolytic is an approach or agent that selectively induces death of senescent cells, thereby eliminating them in the affected organs (Table 2). Studies from animal experiments show that senolytic agents can reverse ageing phenotypes and improve kidney function [30,54]. For example, ABT263 (navitoclax), an inhibitor of Bcl-2 family, has been confirmed to have the ability to eliminate senescent cells in aged mice and preserve tissue regeneration [55]. The anti-senescence peptide FOXO4-DRI is potent remedy for the treatment of kidney disease by binding with FOXO4 to disrupt the p53-FOXO4 interaction, thereby releasing p53 into the cytosol and triggering p53-mediated apoptosis [56]. A peptide that removes p16high cells can attenuate nonalcoholic steatohepatitis-related hepatic lipidosis and immune cell infiltration in p16-CreERT2-tdTomato mouse model [57]. Recent studies show that urokinase-type plasminogen activator receptor (uPAR)-targeted chimeric antigen receptor (CAR) T cells can remove senescent cells. Such uPAR-targeted CAR T cells have therapeutic effect on senescence-associated organ dysfunction via selectively clearing uPAR-expressing senescent cells in murine models [58]**. Similarly, eliminating p16-positive senescent cells in the p16-3MR (tri-modal reporter) and the INK-ATTAC (apoptosis through targeted activation of caspase 8) models also promotes would healing and prevents ageing-associated disorder [59,60]. Inhibition of kidney-type glutaminase (KGA)-dependent glutaminolysis in aged mice attenuates ageing and exhibits a therapeutic benefit in age-associated tissue degeneration [61]*. These experimental studies suggest that senolysis can be harnessed as a therapeutic strategy for the treatment of ageing-related diseases.
In addition to senolytic, a related concept is the so-called senostatic, which means to suppress senescence or the SASP generated by senescent cells. Therapies inhibiting the SASP have proven to be effective in experimental animal models. For example, high phosphate induces kidney tubular epithelial cell senescence, and Klotho have the potential to hamper tubular epithelial senescence [62]. A recent study has indicated that mesenchymal stem cells (MSC)-derived EVs suppress the progression of ageing by blocking cellular senescence [63,64]. Sirt1 activator and peroxisome proliferator-activated receptor-γ (PPAR-γ) agonists have been used for prevention and therapies of ageing-related renal deficiency [65,66]. Metformin exerts an anti-ageing effect by targeting senescent MSCs in CKD [67].
There are nutrient signaling regulators, which can regulate ageing-related diseases and promote health. Numerous studies show that caloric restriction (CR) prolongs lifespan in laboratory animals [68,69]. Moderate CR can be used to halt the age-related kidney dysfunction by inhibiting the endothelin-1 (ET-1) expression [68]. In aged mice, physical exercise reduces triacylglycerol and restores plasma lipids homeostasis by regulating lipid metabolism [70]. Dehydroascorbic acid (DHA) retards or even reverses renal impairment in mouse models of ageing [71].
CONCLUSION
Over the past several years, growing evidence has shown that aged kidneys and CKD share many pathological features and move to similar destination. Accumulation of senescent cells and their associated SASP has been identified as a main driver leading to structural and functional degeneration in CKD and ageing-related disorder of other organs. We have gained a wealth of new knowledge and better understanding on the role, triggers and mechanisms of cellular senescence in ageing and ageing-associated degenerative diseases.
Future studies are warranted to explore new characteristics and delicate regulation of cellular senescence and define the specific molecular targets of senescent cells. Single cell RNA sequencing and relevant bioinformatics analyses need to be employed to facilitate our understanding of the heterogeneity and dynamics of senescent cells in the affected organs. Whether primary and secondary senescence displays any divergence in response to current senotherapy remains an open question. As emerging evidence points to senescent cells as a promising target for therapeutic intervention, this provides an unprecedented opportunity for developing more effective senotherapies to fight against CKD and other ageing-related disorders. It is hopeful that future therapeutics based on targeting cellular senescence will be translated into the clinic for improving the life of millions of patients with ageing-related diseases.
KEY POINTS.
Aged kidney and CKD share common pathological features characterized by cellular senescence, an irreversible state of cell cycle arrest and the cessation of cell division.
There are several models of ageing, including naturally aged and accelerated aged mice, for interrogating the signal pathways involved in cellular senescence and evaluating therapeutic efficacy.
Senescent cells secrete a host of proinflammatory and profibrotic factors known as senescence-associated secretory phenotype (SASP), which can lead neighboring healthy cells to undergo secondary senescence.
Key senescence signaling pathways include the p53/p21 and p16 routes, leading to inhibition of the phosphorylation of cyclin-dependent kinase complexes and cell cycle arrest.
Senescent cells are a promising target for senotherapy, an approach to specifically eliminate or inhibit cellular senescence.
Financial support and sponsorship
This work was supported by the National Natural Science Foundation of China grant 81521003 and 81920108007, and National Institutes of Health grant DK064005.
Footnotes
Conflicts of interest
There are no conflicts of interest.
REFERENCES AND RECOMMENDED READING
Papers of particular interest, published within the annual period of review, have been highlighted as:
• of special interest
•• of outstanding interest
- 1.Partridge L, Deelen J, Slagboom PE: Facing up to the global challenges of ageing. Nature 2018; 561: 45–56. [DOI] [PubMed] [Google Scholar]
- 2.Beard JR, Bloom DE: Towards a comprehensive public health response to population ageing. Lancet 2015; 385: 658–661. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Liu P, Quinn RR, Lam NN et al. : Accounting for age in the definition of chronic kidney disease. JAMA Intern Med 2021; 181: 1359–1366. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. •.Xu J, Zhou L, Liu Y: Cellular senescence in kidney fibrosis: Pathologic significance and therapeutic strategies. Front Pharmacol 2020; 11: 601325. [DOI] [PMC free article] [PubMed] [Google Scholar]; This is a comprehensive review on cellular senescence in kidney fibrosis and chronic kidney disease. It also highlights recent advance on the thepeutic intervention of ageing-related diseases.
- 5. •.Di Micco R, Krizhanovsky V, Baker D et al. : Cellular senescence in ageing: From mechanisms to therapeutic opportunities. Nat Rev Mol Cell Biol 2021; 22: 75–95. [DOI] [PMC free article] [PubMed] [Google Scholar]; This is a more-detailed, broader review of the mechanisms of cellular senescence in ageing. It provides an overview of cellular senescence as a therapeutic target in ageing-associated organ dysfunction.
- 6.Kowald A, Passos JF, Kirkwood TBL: On the evolution of cellular senescence. Aging cell 2020; 19: e13270. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Gasek NS, Kuchel GA, Kirkland JL et al. : Strategies for targeting senescent cells in human disease. Nat Aging 2021; 1: 870–879. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.van Deursen JM: The role of senescent cells in ageing. Nature 2014; 509: 439–446. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Li Z, Wang Z: Aging kidney and aging-related disease. Adv Exp Med Biol 2018; 1086: 169–187. [DOI] [PubMed] [Google Scholar]
- 10.Hommos MS, Glassock RJ, Rule AD: Structural and functional changes in human kidneys with healthy aging. J Am Soc Nephrol 2017; 28: 2838–2844. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Denic A, Glassock RJ, Rule AD: Structural and functional changes with the aging kidney. Adv Chronic Kidney Dis 2016; 23: 19–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Schmitt R, Cantley LG: The impact of aging on kidney repair. Am J Physiol Renal Physiol 2008; 294: F1265–F1272. [DOI] [PubMed] [Google Scholar]
- 13.Yousefzadeh MJ, Melos KI, Angelini L et al. : Mouse models of accelerated cellular senescence. Methods Mol Biol 2019; 1896: 203–230. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Beck J, Horikawa I, Harris C: Cellular senescence: Mechanisms, morphology, and mouse models. Vet Pathol 2020; 57: 747–757. [DOI] [PubMed] [Google Scholar]
- 15.Mostoslavsky R, Chua KF, Lombard DB et al. : Genomic instability and aging-like phenotype in the absence of mammalian sirt6. Cell 2006; 124: 315–329. [DOI] [PubMed] [Google Scholar]
- 16.Bernardes de Jesus B, Blasco MA: Assessing cell and organ senescence biomarkers. Circ Res 2012; 111: [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Birch J, Gil J: Senescence and the sasp: Many therapeutic avenues. Genes Dev 2020; 34: 1565–1576. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Docherty MH, Baird DP, Hughes J et al. : Cellular senescence and senotherapies in the kidney: Current evidence and future directions. Front Pharmacol 2020; 11: 755. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Hernandez-Segura A, Nehme J, Demaria M: Hallmarks of cellular senescence. Trends Cell Biol 2018; 28: 436–453. [DOI] [PubMed] [Google Scholar]
- 20.Kurz DJ, Decary S, Hong Y et al. : Senescence-associated (beta)-galactosidase reflects an increase in lysosomal mass during replicative ageing of human endothelial cells. J Cell Sci 2000; 113: 3613–3622. [DOI] [PubMed] [Google Scholar]
- 21.d’Adda di Fagagna F, Reaper PM, Clay-Farrace L et al. : A DNA damage checkpoint response in telomere-initiated senescence. Nature 2003; 426: 194–198. [DOI] [PubMed] [Google Scholar]
- 22.Narita M, Narita M, Krizhanovsky V et al. : A novel role for high-mobility group a proteins in cellular senescence and heterochromatin formation. Cell 2006; 126: 503–514. [DOI] [PubMed] [Google Scholar]
- 23.d’Adda di Fagagna F: Living on a break: Cellular senescence as a DNA-damage response. Nat Rev Cancer 2008; 8: 512–522. [DOI] [PubMed] [Google Scholar]
- 24.Wang AS, Ong PF, Chojnowski A et al. : Loss of lamin b1 is a biomarker to quantify cellular senescence in photoaged skin. Sci Rep 2017; 7: 15678. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Tyler EJ, Gutierrez Del Arroyo A, Hughes BK et al. : Early growth response 2 (egr2) is a novel regulator of the senescence programme. Aging cell 2021; 20: e13318. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Yuan Y, Li J, He Z et al. : Trna-derived fragments as new hallmarks of aging and age-related diseases. Aging Dis 2021; 12: 1304–1322. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Chen L, Liao F, Wu J et al. : Acceleration of ageing via disturbing mtor-regulated proteostasis by a new ageing-associated gene pc4. Aging Cell 2021; 20: e13370. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Wang W, Zheng Y, Sun S et al. : A genome-wide crispr-based screen identifies as a driver of cellular senescence. Sci Transl Med 2021; 13: [DOI] [PubMed] [Google Scholar]
- 29.López-Domínguez JA, Rodríguez-López S, Ahumada-Castro U et al. : Transcript variant 2 is a marker of aging and cellular senescence. Aging 2021; 13: 13380–13392. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Kirkland JL, Tchkonia T: Senolytic drugs: From discovery to translation. J Intern Med 2020; 288: 518–536. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Chen W, Wang X, Wei G et al. : Single-cell transcriptome analysis reveals six subpopulations reflecting distinct cellular fates in senescent mouse embryonic fibroblasts. Front Genet 2020; 11: 867. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Muñoz-Espín D, Cañamero M, Maraver A et al. : Programmed cell senescence during mammalian embryonic development. Cell 2013; 155: 1104–1118. [DOI] [PubMed] [Google Scholar]
- 33. •.Admasu TD, Rae M, Stolzing A: Dissecting primary and secondary senescence to enable new senotherapeutic strategies. Ageing Res Rev 2021; 70: 101412. [DOI] [PubMed] [Google Scholar]; This paper summarizes the difference between primary senescence and secondary senescence. Secondary senescence can spread and accumulate senescent cells.
- 34.Xu Y, Sun Z: Molecular basis of klotho: From gene to function in aging. Endocr Rev 2015; 36: 174–193. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Kurosu H, Yamamoto M, Clark JD et al. : Suppression of aging in mice by the hormone klotho. Science 2005; 309: 1829–1833. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Sopjani M, Rinnerthaler M, Kruja J et al. : Intracellular signaling of the aging suppressor protein klotho. Curr Mol Med 2015; 15: 27–37. [DOI] [PubMed] [Google Scholar]
- 37.Kadoya H, Satoh M, Nishi Y et al. : Klotho is a novel therapeutic target in peritoneal fibrosis via wnt signaling inhibition. Nephrol Dial Transplant 2020; 35: 773–781. [DOI] [PubMed] [Google Scholar]
- 38.Miao J, Huang J, Luo C et al. : Klotho retards renal fibrosis through targeting mitochondrial dysfunction and cellular senescence in renal tubular cells. Physiol Rep 2021; 9: e14696. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Brunet A, Sweeney LB, Sturgill JF et al. : Stress-dependent regulation of foxo transcription factors by the sirt1 deacetylase. Science 2004; 303: 2011–2015. [DOI] [PubMed] [Google Scholar]
- 40.Yeung F, Hoberg JE, Ramsey CS et al. : Modulation of nf-kappab-dependent transcription and cell survival by the sirt1 deacetylase. EMBO J 2004; 23: 2369–2380. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Chuang PY, Cai W, Li X et al. : Reduction in podocyte sirt1 accelerates kidney injury in aging mice. Am J Physiol Renal Physiol 2017; 313: F621–F628. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Ota H, Akishita M, Eto M et al. : Sirt1 modulates premature senescence-like phenotype in human endothelial cells. J Mol Cell Cardiol 2007; 43: 571–579. [DOI] [PubMed] [Google Scholar]
- 43.Sun H-J, Xiong S-P, Cao X et al. : Polysulfide-mediated sulfhydration of sirt1 prevents diabetic nephropathy by suppressing phosphorylation and acetylation of p65 nf-κb and stat3. Redox Biol 2021; 38: 101813. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Kim DY, Lee M, Kim EJ: Involvement of klotho, tnf-α and adams in radiation-induced senescence of renal epithelial cells. Mol Med Rep 2021; 23: [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Miao J, Liu J, Niu J et al. : Wnt/β-catenin/ras signaling mediates age-related renal fibrosis and is associated with mitochondrial dysfunction. Aging cell 2019; 18: e13004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Luo C, Zhou S, Zhou Z et al. : Wnt9a promotes renal fibrosis by accelerating cellular senescence in tubular epithelial cells. J Am Soc Nephrol 2018; 29: 1238–1256. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Lee HJ, Donati A, Feliers D et al. : Chloride channel accessory 1 integrates chloride channel activity and mtorc1 in aging-related kidney injury. Aging cell 2021; 20: e13407. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Zhou S, Ling X, Meng P et al. : Cannabinoid receptor 2 plays a central role in renal tubular mitochondrial dysfunction and kidney ageing. J Cell Mol Med 2021; 25: 8957–8972. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Benitah SA, Welz PS: Circadian regulation of adult stem cell homeostasis and aging. Cell Stem Cell 2020; 26: 817–831. [DOI] [PubMed] [Google Scholar]
- 50.Welz P-S, Benitah SA: Molecular connections between circadian clocks and aging. J Mol Biol 2020; 432: 3661–3679. [DOI] [PubMed] [Google Scholar]
- 51.Wu C-S, Muthyala SDV, Klemashevich C et al. : Age-dependent remodeling of gut microbiome and host serum metabolome in mice. Aging 2021; 13: 6330–6345. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Nakao M, Tanaka H, Koga T: Cellular senescence variation by metabolic and epigenomic remodeling. Trends Cell Biol 2020; 30: 919–922. [DOI] [PubMed] [Google Scholar]
- 53.Zhu M, Meng P, Ling X et al. : Advancements in therapeutic drugs targeting of senescence. Ther Adv Chronic Dis 2020; 11: 2040622320964125. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Wang M: Senolytics for kidney repair. Nat Rev Nephrol 2021; 17: 512. [DOI] [PubMed] [Google Scholar]
- 55.Mylonas KJ, O’Sullivan ED, Humphries D et al. : Cellular senescence inhibits renal regeneration after injury in mice, with senolytic treatment promoting repair. Sci Transl Med 2021; 13: [DOI] [PubMed] [Google Scholar]
- 56.Zhang C, Xie Y, Chen H et al. : Foxo4-dri alleviates age-related testosterone secretion insufficiency by targeting senescent leydig cells in aged mice. Aging 2020; 12: 1272–1284. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Omori S, Wang T-W, Johmura Y et al. : Generation of a p16 reporter mouse and its use to characterize and target p16 cells in vivo. Cell Metab 2020; 32: [DOI] [PubMed] [Google Scholar]
- 58. ••.Amor C, Feucht J, Leibold J et al. : Senolytic car t cells reverse senescence-associated pathologies. Nature 2020; 583: 127–132. [DOI] [PMC free article] [PubMed] [Google Scholar]; This study shows that urokinase-type plasminogen activator receptor (uPAR)-targeted chimeric antigen receptor (CAR) T cells can remove senescent cells. Senolytic CAR T cells have the potential to be remedy for senescence-associated diseases.
- 59.Demaria M, Ohtani N, Youssef SA et al. : An essential role for senescent cells in optimal wound healing through secretion of pdgf-aa. Dev Cell 2014; 31: 722–733. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Baker DJ, Wijshake T, Tchkonia T et al. : Clearance of p16ink4a-positive senescent cells delays ageing-associated disorders. Nature 2011; 479: 232–236. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. •.Johmura Y, Yamanaka T, Omori S et al. : Senolysis by glutaminolysis inhibition ameliorates various age-associated disorders. Science 2021; 371: 265–270. [DOI] [PubMed] [Google Scholar]; This paper shows that aged-associated lesions in mice can be ameliorated by inhibiting kidney-type glutaminase (KGA)-dependent glutaminolysis. Glutaminolysis inhibition displays a promising therapeutic effect on ageing and age-associated diseases.
- 62.Maique J, Flores B, Shi M et al. : High phosphate induces and klotho attenuates kidney epithelial senescence and fibrosis. Front Pharmacol 2020; 11: 1273. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Dorronsoro A, Santiago FE, Grassi D et al. : Mesenchymal stem cell-derived extracellular vesicles reduce senescence and extend health span in mouse models of aging. Aging cell 2021; 20: e13337. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Lei Q, Gao F, Liu T et al. : Extracellular vesicles deposit to rejuvenate aged bone marrow-derived mesenchymal stem cells and slow age-related degeneration. Sci Transl Med 2021; 13: [DOI] [PubMed] [Google Scholar]
- 65.Han X, Ding C, Sang X et al. : Targeting sirtuin1 to treat aging-related tissue fibrosis: From prevention to therapy. Pharmacol Ther 2021; 107983. [DOI] [PubMed] [Google Scholar]
- 66.Xu L, Ma X, Verma N et al. : Pparγ agonists delay age-associated metabolic disease and extend longevity. Aging cell 2020; 19: e13267. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Kim H, Yu MR, Lee H et al. : Metformin inhibits chronic kidney disease-induced DNA damage and senescence of mesenchymal stem cells. Aging cell 2021; 20: e13317. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Wang X-H, Ao Q-G, Cheng Q-L: Caloric restriction inhibits renal artery ageing by reducing endothelin-1 expression. Ann Transl Med 2021; 9: 979. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Fontana L, Partridge L: Promoting health and longevity through diet: From model organisms to humans. Cell 2015; 161: 106–118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Kim KU, Yoon KJ, Park S et al. : Exercise-induced recovery of plasma lipids perturbed by ageing with nanoflow uhplc-esi-ms/ms. Anal Bioanal Chem 2020; 412: 8003–8014. [DOI] [PubMed] [Google Scholar]
- 71.Forman K, Martínez F, Cifuentes M et al. : Dehydroascorbic acid, the oxidized form of vitamin c, improves renal histology and function in old mice. J Cell Physiol 2020; 235: 9773–9784. [DOI] [PubMed] [Google Scholar]
- 72.Chen K, Wang S, Sun QW et al. : Klotho deficiency causes heart aging via impairing the nrf2-gr pathway. Circ Res 2021; 128: 492–507. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Tang Y-L, Zhou Y, Wang Y-P et al. : Ginsenoside rg1 protects against sca-1 hsc/hpc cell aging by regulating the sirt1-foxo3 and sirt3-sod2 signaling pathways in a γ-ray irradiation-induced aging mice model. Exp Ther Med 2020; 20: 1245–1252. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Kuro-o M, Matsumura Y, Aizawa H et al. : Mutation of the mouse klotho gene leads to a syndrome resembling ageing. Nature 1997; 390: 45–51. [DOI] [PubMed] [Google Scholar]
- 75.Griveau A, Wiel C, Ziegler DV et al. : The jak1/2 inhibitor ruxolitinib delays premature aging phenotypes. Aging cell 2020; 19: e13122. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Fuhrmann-Stroissnigg H, Ling YY, Zhao J et al. : Identification of hsp90 inhibitors as a novel class of senolytics. Nat Commun 2017; 8: 422. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.de Boer J, Andressoo JO, de Wit J et al. : Premature aging in mice deficient in DNA repair and transcription. Science 2002; 296: 1276–1279. [DOI] [PubMed] [Google Scholar]
- 78.Hickson LJ, Langhi Prata LGP, Bobart SA et al. : Senolytics decrease senescent cells in humans: Preliminary report from a clinical trial of dasatinib plus quercetin in individuals with diabetic kidney disease. EBioMedicine 2019; 47: 446–456. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Yousefzadeh MJ, Zhu Y, McGowan SJ et al. : Fisetin is a senotherapeutic that extends health and lifespan. EBioMedicine 2018; 36: 18–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Kumar R, Sharma A, Kumari A et al. : Epigallocatechin gallate suppresses premature senescence of preadipocytes by inhibition of pi3k/akt/mtor pathway and induces senescent cell death by regulation of bax/bcl-2 pathway. Biogerontology 2019; 20: 171–189. [DOI] [PubMed] [Google Scholar]
- 81.Zhu Y, Tchkonia T, Fuhrmann-Stroissnigg H et al. : Identification of a novel senolytic agent, navitoclax, targeting the bcl-2 family of anti-apoptotic factors. Aging cell 2016; 15: 428–435. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Samaraweera L, Adomako A, Rodriguez-Gabin A et al. : A novel indication for panobinostat as a senolytic drug in nsclc and hnscc. Sci Rep 2017; 7: 1900. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Lee F-Y, Shao P-L, Wallace CG et al. : Combined therapy with ss31 and mitochondria mitigates myocardial ischemia-reperfusion injury in rats. Int J Mol Sci 2018; 19: [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Xiao L, Xu X, Zhang F et al. : The mitochondria-targeted antioxidant mitoq ameliorated tubular injury mediated by mitophagy in diabetic kidney disease via nrf2/pink1. Redox Biol 2017; 11: 297–311. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Anisimov VN, Egorov MV, Krasilshchikova MS et al. : Effects of the mitochondria-targeted antioxidant skq1 on lifespan of rodents. Aging 2011; 3: 1110–1119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Chuang J-I, Pan IL, Hsieh C-Y et al. : Melatonin prevents the dynamin-related protein 1-dependent mitochondrial fission and oxidative insult in the cortical neurons after 1-methyl-4-phenylpyridinium treatment. J Pineal Res 2016; 61: 230–240. [DOI] [PubMed] [Google Scholar]
- 87.Sudheesh NP, Ajith TA, Ramnath V et al. : Therapeutic potential of ganoderma lucidum (fr.) p. Karst. Against the declined antioxidant status in the mitochondria of post-mitotic tissues of aged mice. Clin Nutr 2010; 29: 406–412. [DOI] [PubMed] [Google Scholar]
- 88.Harrison DE, Strong R, Sharp ZD et al. : Rapamycin fed late in life extends lifespan in genetically heterogeneous mice. Nature 2009; 460: 392–395. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Abbott A: Reduced-calorie diet shows signs of slowing ageing in people. Nature 2018; 555: 570–571. [DOI] [PubMed] [Google Scholar]
- 90.Howitz KT, Bitterman KJ, Cohen HY et al. : Small molecule activators of sirtuins extend saccharomyces cerevisiae lifespan. Nature 2003; 425: 191–196. [DOI] [PubMed] [Google Scholar]
- 91.Liao VH-C, Yu C-W, Chu Y-J et al. : Curcumin-mediated lifespan extension in caenorhabditis elegans. Mech Ageing Dev 2011; 132: 480–487. [DOI] [PubMed] [Google Scholar]
- 92.Yue F, Li W, Zou J et al. : Spermidine prolongs lifespan and prevents liver fibrosis and hepatocellular carcinoma by activating map1s-mediated autophagy. Cancer Res 2017; 77: 2938–2951. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Hsieh F-C, Hung C-T, Cheng K-C et al. : Protective effects of extracts on uvb-induced damage in human retinal pigment epithelial cells accompanied by attenuating ros and DNA damage. Oxid Med Cell Longev 2018; 2018: 4814928. [DOI] [PMC free article] [PubMed] [Google Scholar]