Abstract
Aging is a major risk factor for cardiovascular diseases (CVDs) and accumulating evidence indicates that biological aging has a significant effect on the onset and progression of CVDs. In recent years, therapies targeting senescent cells (senotherapies), particularly senolytics that selectively eliminate senescent cells, have been developed and show promise for treating geriatric syndromes and age-associated diseases, including CVDs. In 2 pilot studies published in 2019 the senolytic combination, dasatinib plus quercetin, improved physical function in patients with idiopathic pulmonary fibrosis and eliminated senescent cells from adipose tissue in patients with diabetic kidney disease. More than 30 clinical trials using senolytics are currently underway or planned. In preclinical CVD models, senolytics appear to improve heart failure, ischemic heart disease, valvular heart disease, atherosclerosis, aortic aneurysm, vascular dysfunction, dialysis arteriovenous fistula patency, and pre-eclampsia. Because senotherapies are completely different strategies from existing treatment paradigms, they might alleviate diseases for which there are no current effective treatments or they could be used in addition to current therapies to enhance efficacy. Moreover, senotherapies might delay, prevent, alleviate or treat multiple diseases in the elderly and reduce polypharmacy, because senotherapies target fundamental aging mechanisms. We comprehensively summarize the preclinical evidence about senotherapies for CVDs and discuss future prospects for their clinical application.
Keywords: Atherosclerosis, Cellular senescence, Diabetes, Senescence-associated secretory phenotype (SASP), Senolytics
Cardiovascular diseases (CVDs) remain the leading cause of morbidity and mortality among older people globally, claiming an estimated 17.9 million lives each year.1 The prevalence of the majority of CVDs such as heart attacks, coronary artery disease, hypertension, atherosclerosis, stroke, heart failure (HF), valvular heart diseases, aortic aneurysms, and arrhythmias, including atrial fibrillation (AF), increases steadily with patient age. Although the pathogenesis of CVDs is multifactorial and risk factors for CVDs such as diabetes and hypertension increase with age, aging appears to be an independent risk factor.2,3 Although CVD mortality rates are declining,4 despite aggressive control of risk factors as recommended in recent guidelines, the prevalence of CVDs has not improved dramatically and may be due to failure to intervene against a key underlying cause of CVD: biological aging.
Age-associated diseases and geriatric syndromes including CVDs are, in part, a consequence of biological aging. Targeting the hallmarks of biological aging may be a root-cause therapeutic approach for delaying the onset or alleviating multiple age-associated conditions; that is, the “geroscience hypothesis”.4 Moreover, intervening against any one of the biological aging mechanisms that underlie age-associated diseases and geriatric syndromes may affect many or most of the rest, a unitary theory of fundamental aging mechanisms.5
Senotherapies
The 3 approaches being investigated as potential senotherapies in preclinical models are: (1) elimination of persisting senescent cells (senolytics), (2) in vivo direct reprogramming of senescent cells, and (3) suppression of release of the senescence-associated secretory phenotype (SASP) factors by senescent cells (senomorphics) (Figure).
Figure.

Contributions of senescent cells to cardiovascular diseases (CVDs). Senescent cells accumulate during aging and are related to many forms of stress including DNA damage, mechanical/shear stress, reactive metabolites (e.g., reactive oxygen species), angiotensin II, hyperinsulinemia, and oxidized low-density lipoprotein. The cell cycle becomes arrested and so reduces the risk that damaged dysfunctional cells proliferate and give rise to cancers. However, persistent dysfunctional senescent cells can continue to release tissue-damaging SASP factors that induce inflammation, contributing to the pathology of CVDs. There are currently 3 strategies being investigated to attenuate unfavorable consequences of persistent, pro-inflammatory senescent cells: (1) senolytics that induce death of those senescent cells that are pro-apoptotic; (2) reprogramming to alter epigenetic programming through the Yamanaka factors (OSKM) to revert senescent cells into a non-senescent state; and (3) senomorphics that inhibit production of tissue-destructive SASP factors from senescent cells while not directly eliminating the senescent cells themselves. Metformin, rapamycin, and ruxolitinib are among the senomorphics in clinical use for various indications.
Senolytics was proposed as a therapeutic strategy of inducing cell death in senescent cells. The first senolytic, a combination of dasatinib and quercetin (D+Q), reduced the senescent cell burden in naturally aged mice,6–9 and alleviated age-related metabolic dysfunction.10 A causal role for cellular senescence in the generation of age-related phenotypes was further demonstrated by showing that transplantation of small numbers of senescent cells into middle-aged mice can cause frailty-like physical dysfunction and multiple age-associated diseases.11 These findings support the potential of senolytics as a root-cause treatment for many age-associated disorders and diseases. Based on the first study of senolytics, more have since been identified4,10,12–31 (Table 1). To date, senolytics appear to alleviate multiple conditions, including cardiac dysfunction,5,7,32–35 vascular diseases,34,35 frailty,36–38 cognitive impairment,39–43 renal dysfunction,44 pulmonary diseases,38 osteoporosis,28,45,46 and hepatic steatosis.47
Table 1.
List of Senotherapies
| Senolytic | Target molecules | Localization |
|---|---|---|
| Dasatinib | Src kinase, Tyrosine kinase, etc | Plasma membrane |
| Quercetin | Bcl-2 family, p53/p21/serpine, PI3K/AKT | Cytoplasm, Mitochondria, Nuclei |
| Fisetin | PI3K/AKT | Cytoplasm |
| Navitoclax (ABT-263) | Bcl-2 family (Bcl-2, Bcl-xl, Bcl-w) | Cytoplasm, Mitochondria |
| PZ15227 | Bcl-xl | Cytoplasm, Mitochondria |
| TW-37 | Bcl-2 family (Bcl-2, Bcl-xl, Mcl-1) | Cytoplasm, Mitochondria |
| ABT-737 | Bcl-xl, Bcl-w | Cytoplasm, Mitochondria |
| Luteolin | Bcl-2 family (Bcl-xl) | Cytoplasm, Mitochondria |
| Curcumin, Curcumin analog EF24 | Bcl-2 family (Bcl-xl) | Cytoplasm, Mitochondria |
| A1331852 | Bcl-2 family (Bcl-xl) | Cytoplasm, Mitochondria |
| A1155463 | Bcl-2 family (Bcl-xl) | Cytoplasm, Mitochondria |
| Piperlongumine | p53/p21, Bcl-2 family (PUMA) | Cytoplasm, Mitochondria, Nuclei |
| FOXO4-DRI | FOXO-p53 interaction | Nuclei |
| Nutlin3a | p53 | Nuclei |
| UBX0101 | p53-MDM2 / p21 | Nuclei |
| Panobinostat | Histon deacetylase | Nuclei |
| ARV825 | BET family protein, NHEJ | Cytoplasm |
| AT-406 | IAP1/2/XIAP | Cytoplasm |
| Fenofibrate | PPARa | Cytoplasm |
| Alvespimycin (17-DMAG) | HSP90 | Cytoplasm |
| Tanespimycin (17-AAG) | HSP90 | Cytoplasm |
| Geldanamycin | HSP90 | Cytoplasm |
| Digoxin (cardiac glycosides) | Na+/K+ ATPase pump | Plasma membrane |
| BPTES | GLS1 | Lysosome |
| Galactose-modified duocarmycin (GMD) | Lysosomal β-galactosidase | Lysosome |
| SSK1-Gemcitabine | Lysosomal β-galactosidase | Lysosome |
| 2-DG | Lysosomal V-ATPases | Lysosome |
| GPNMB vaccine | GPNMB | Plasma membrane, lysosome |
| CD153 vaccine | CD153 | Plasma membrane |
| ADC against B2M | β2 microglobulin (B2M) | Plasma membrane |
| uPAR specific CAR T | Urokinase-type plasminogen activator receptor (uPAR) | Plasma membrane |
| Senomorphics | Target molecules | Localization |
| Rapamycin | mTOR | Cytoplasm |
| Metformin | AMPK | Cytoplasm |
| Ruxolitinib | JAK 1/2 | Cytoplasm |
| NBD peptide/mimetics | NF-κB | Cytoplasm |
2-DG, 2-deoxy-d-glucose; 17-DMAG, 17-dimethylaminoethylamino-17-demethoxygeldanamycin; NHEJ, non-homologous end joining.
Another strategy to mitigate the adverse effects of senescent cells is to reprogram them using the Yamanaka factors, Oct4, Sox2, Klf4, and c-Myc (OSKM), the discovery of which led to a Nobel prize in 2012.48 This approach allows cells, including senescent cells, to be converted to a state akin to pluripotency that might be applicable to counteract the effect of biological aging,49–55 as well as regenerative medicine and development of new drugs. This field is evolving, and the long-term effects and the risk for adverse events have not yet been fully elucidated. Enabling potentially cancerous mutation-harboring senescent cells to escape replicative arrest is a concern.
The third approach to targeting cellular senescence is to use SASP inhibitors, senomorphics. This approach is different from the other two, because it does not target senescent cells themselves but rather the release of detrimental, inflammatory SASP factors by senescent cells. Several agents that are senomorphic, including metformin, ruxolitinib, and rapamycin, are in clinical use, with data available from post-marketing surveillance56 and retrospective studies.57–64 A clinical trial using low-dose rapamycin for coronary artery disease (Cardiac Rehabilitation And Rapamycin in Elderly [CARE]) was conducted to test the safety and feasibility of low-dose rapamycin and effects on both SASP and frailty in elderly subjects undergoing cardiac rehabilitation. Intriguingly, rapamycin suppressed some of markers of senescent cells in adipose tissue, although it failed to alleviate frailty.65 Another clinical trial, TAME (Targeting Aging with Metformin), will test if metformin delays the appearance of a second age-associated disease.59,66 A challenge in using senomorphic drugs is modulating potential off-target effects on non-senescent cells and potential side effects from suppressing the physiological inflammation that may be necessary for resolving certain acute diseases or promoting tissue repair. Additionally, senomorphic treatment allows senescent cells themselves to persist, potentially necessitating more continuous administration of SASP inhibitors that senolytics, which could increase off-target and side effects.
Preclinical Evidence of Senolytics for CVDs
In preclinical studies of CVDs there is mounting evidence of the potential benefits of senolytics (Table 2). D+Q,7 quercetin,67,68 and navitoclax69,70 have improved cardiac function in aged mice and several HF models. In older humans, diastolic dysfunction, also known as HF with preserved ejection fraction (HFpEF), is a major problem.71–74 An effect of senolytics on HFpEF may be also expected. In addition, D+Q prolongs survival of cardiac allografts from old mice,69 with a reduction in senescent cell burden and inflammation, suggesting senolytics may be of benefit in the case of organ transplantation or cell-based therapies to increase their viability.75–77 Moreover, D+Q, by eliminating senescent cells, improved human induced pluripotent stem (iPS) cells-derived cardiomyocyte survival.78 Senolytics may be potentially useful in ischemic heart disease. D+Q activated approximately 10% of cardiac resident cardiac progenitor cells (CPCs) and increased differentiation from CPCs,32 in association with an improvement of cardiac repair.79 Navitoclax alleviated increased the left ventricular EF, enhanced myocardial vascularization in an ischemia-reperfusion model in mice and rats, and increased survival after myocardial infarction in aged mice.33,80,81 Navitoclax-coated stents were associated with reduced stenotic area in rabbits on a high cholesterol diet.82
Table 2.
Preclinical Studies of Senolytics for Cardiovascular Diseases
| Disease targeted | Results of senolytic therapy | Senolytics | Senolytic model | Disease model |
|---|---|---|---|---|
| Heart failure | Alleviated impaired ejection fraction Activated resident CPCs and increased cardiomyocyte proliferation Reduced myocardial hypertrophy and fibrosis |
D+Q7,32 Navitoclax33 |
INK-ATTAC 32,33 | Aged (mouse) |
| Attenuated cardiac systolic dysfunction Normalized hypertrophy Reduced cardiac fat and fibrosis Increased proliferation of CPCs and cardiac repair |
Q67,68 D+Q79 Navitoclax70,119 |
High fat diet-induced Angiotensin II (Ang II)-induced Doxorubicin (DOX)-induced (Mouse or rat) |
||
| Heart transplantation | Prolonged the survival of cardiac allografts from aged mice | D+Q69 | Aged (mouse) | |
| Hypertension | Improved vascular relaxation Alleviated aortic calcification and improved vasomotor function |
D+Q7,34 | INK-ATTAC 34 | Aged (mouse) |
| Pulmonary arterial hypertension | Improved hemodynamic and structural changes associated with severe PAH | Navitoclax97 | MCT and shunt-induced PAH (rat) | |
| Ischemic heart diseases | Reduced myocardial hypertrophy and fibrosis Alleviated impaired ejection fraction Increased myocardial vascularization Increased survival after myocardial infarction |
D+Q120 Navitoclax81,121,122 |
Ischemia-reperfusion or myocardial infarction model (mouse or rat) | |
| Atherosclerosis | Reduced atherosclerotic plaque formation and instability Increased thickness of the fibrous cap |
Navitoclax35,91,95 GPNMB-vaccine26 Digoxin93 17-DMAG94 BPTES24 |
INK-ATTAC
35,95
p16-3MR 35,91,95 |
Ldlr KO or ApoE KO (mouse) |
| Coronary artery stent restenosis | Reduced stenosis area of senolytic-coated stent | Navitoclax82 | Cholesterol diet (rabbit) | |
| Abdominal aortic aneurysm | Reduced size of AAA | D+Q96 | Angiotensin II (Ang II) administrated in aged (mouse) | |
| Atrial fibrillation | Ameliorated atrial fibrosis Shorten AF duration |
Fisetin89 Q88 |
Myocardial infarction (rat) Isoproterenol (rat) |
|
| Chronic kidney disease | Decreased senescent cells burden | D+Q99 | Dialysis arteriovenous fistula (mouse) | |
| Diabetic retinal angiopathy | Attenuated ischemic retinopathy | UBX1967100 | INK-ATTAC 100 | Oxygen-induced retinopathy (mouse) Eye tissue (human) |
The incidence of AF increases with aging.83,84 Senescent cell accumulation in the atrium with aging has been demonstrated in rodents and humans.85–87 There are 2 preclinical studies with either quercetin88 or fisetin89 that demonstrated potential alleviation of AF and fibrosis, although in those studies the drugs were used as antifibrotics and senescent cell removal was not specifically examined. The results obtained in rodent models may not fully reflect the underlying mechanisms of AF in humans, because AF is not a naturally occurring condition in rodents.90
Senolytic therapies have also been tested in preclinical models of vascular diseases with promising results. Senolytics such as anti-Gpnmb vaccination, navitoclax, digoxin, BPTES, and 17-DMAG (alvespimycin) have reduced atherosclerotic plaque formation and inflammation,22–24,35,91–94 alleviated fibrous capsule thinning of atherosclerotic plaques,95 and attenuated vascular calcification.34 D+Q might attenuate hypertension in the elderly because it improves vasorelaxation and vasomotor function in aged mice.7,34 Also, in aged mice with abdominal aortic aneurysms (AAA), D+Q reduced the size of the AAA.96 In addition to these effects, senolytics has been tested for efficacy in the diseases such as pulmonary arterial hypertension (PAH),97,98 hemodialysis shunt survival,99 and ischemic retinopathy100 (Table 2).
Clinical Trials of Senolytics
More than 30 clinical trials of senolytics, including phase 2 randomized, double-blinded, placebo-controlled trials, are already underway or planned based on the promising results in preclinical experiments.4 D+Q was used in 2 pilot studies of senolytics because 1 approach is to use senolytic compounds already approved for other uses or natural products with established safety data in the early studies and already favorable results have been reported.101,102 In the field of CVDs, translation of antisenescence drugs into the clinic has lagged, although, there are current studies planned or beginning with senolytic drugs targeting cardiac and arterial diseases.103 One is the Q-CABG study, testing oral quercetin monotherapy as a senolytic in patients undergoing coronary artery bypass graft (CABG) surgery. More clinical studies are being planned, including for HF with different causes, arrhythmias such as AF, and maintaining arteriovenous dialysis fistula patency, among other CVDs.
Future Directions
Senolytics, as a novel class of drugs, might be effective in diseases for which no good treatments currently exist, such as HFpEF. Senolytics may have the potential for synergistic beneficial effects if combined with current therapies that are known to improve the prognosis of CVDs.104 The effectiveness of senolytics might also be enhanced when used in combination with other interventions, such as lifestyle changes (e.g., caloric restriction, exercise), that target interlinked fundamental aging processes. Senolytics might also alleviate more than one pathology associated with aging at the same time. Such multimorbidity is often a problem for older patients with CVDs.105,106
The senotherapy field is new and there are important steps that need to be taken to efficiently discover new, safe senolytics and senomorphics and translate them into agents for clinical use. Preclinical and early clinical trials suggested short-term safety and target engagement for senolytics, but it will be important to conduct further studies to test for potential long-term side effects. Another need is to identify gerodiagnostic biomarkers that can be tested in body fluids such as blood, urine, saliva or tears to monitor senescent cell abundance. Among such gerodiagnostics are CD3-positive T cells expressing p16INK4a in peripheral blood,107,108 and α-Klotho in urine.109 SASP factors in circulating blood, tissue or cells might also be used for estimating senescent cell burden. Although a single SASP factor may not be highly sensitive or specific, combinations of such indicators might be better.69,101,102,110,111 In addition to protein and peptide SASP factors, blood non-coding nucleotide SASP factors (microRNAs [miRNA], short non-coding RNAs,112,113 small extracellular vesicles,114,115 and mitochondrial DNAs69) are emerging as gerodiagnostic markers. One difficulty is that senescent cells are highly heterogeneous: they release different factors depending on the cell type that became senescent, the inducer of senescence, how long the cell has been senescent, and the tissue microenvironment.116–118
Conclusions
Senotherapies are an emerging strategy for intervening against fundamental aging mechanisms to potentially prevent and treat multiple disorders and diseases and extend health span. Because senotherapies allow targeting of the underlying causes of aging, rather than treating disorders and diseases individually, they could reduce polypharmacy, which is a cause of the attendant risks of adverse events and drug interactions. Senolytics are particularly attractive because they seem to be effective even when given intermittently, despite having short elimination half-lives, thereby minimizing potential toxic effects.
Although clinical studies of senotherapies for CVDs are limited so far, promising evidence from preclinical studies suggests senolytic agents might become a novel strategy for delaying, preventing, alleviating or treating CVDs. Further research is needed to identify the combinations of gerodiagnostic markers for monitoring target engagement during clinical trials and potentially for identifying which senolytics to use, when, and for how long to customize treatment for each patient. It must be borne in mind that treatments that appear to be highly promising in rodents frequently fail in clinical trials, especially in the cases of disorders such as AF, which do not occur naturally in rodents. It will be important to conduct validation studies for these diseases in large animal CVD models such rabbits, dogs, pigs, and monkeys. Because clinical studies of senolytics and senomorphics have already begun for other indications, it will be helpful to closely monitor cardiovascular effects and side effects in those studies. If eventually shown to be safe and effective, senolytics might one day be indicated for a number of CVDs.
Acknowledgments
This work was supported by the US National Institutes of Health (grants R37AG013925, R33AG061456, R01AG072301, R01AG061414, R01DK120292, R01AG68048, R01AG 64165, P01AG062413, and UH3AG056933), the Connor Fund, Robert J. and Theresa W. Ryan, the Noaber Foundation, a Grant-in-Aid for Scientific Research (A) (20H00533) and Grant-in-Aid for Challenging Research (Pioneering) (22K18389) from MEXT, AMED-CREST (JP22 gm1110012), and the Moonshot Research and Development Program (22zf0127003s0201).
Footnotes
Disclosure Summary
M.S., G.K. have no disclosures. T.M. is a member of Circulation Journal’s Editorial Team. T.T., J.L.K. have a financial interest related to this research, including patents and pending patents covering senolytic drugs and their uses that are held by Mayo Clinic. This research has been reviewed by the Mayo Clinic Conflict of Interest Review Board and was conducted in compliance with Mayo Clinic conflict of interest policies.
References
- 1.Cardiovascular diseases. https://www.who.int/health-topics/cardiovascular-diseases#tab=tab_1 (accessed October 7, 2023).
- 2.Rodgers JL, Jones J, Bolleddu SI, Vanthenapalli S, Rodgers LE, Shah K, et al. Cardiovascular risks associated with gender and aging. J Cardiovasc Dev Dis 2019; 6: 19, doi: 10.3390/jcdd6020019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.North BJ, Sinclair DA. The intersection between aging and cardiovascular disease. Circ Res 2012; 110: 1097–1108, doi: 10.1161/circresaha.111.246876. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Chaib S, Tchkonia T, Kirkland JL. Cellular senescence and senolytics: The path to the clinic. Nat Med 2022, doi: 10.1038/s41591-022-01923-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Tchkonia T, Palmer AK, Kirkland JL. New horizons: Novel approaches to enhance healthspan through targeting cellular senescence and related aging mechanisms. J Clin Endocrinol Metab 2021; 106: e1481–e1487, doi: 10.1210/clinem/dgaa728. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Wang E Senescent human fibroblasts resist programmed cell death, and failure to suppress bcl2 is involved. Cancer Res 1995; 55: 2284–2292. [PubMed] [Google Scholar]
- 7.Zhu Y, Tchkonia T, Pirtskhalava T, Gower AC, Ding H, Giorgadze N, et al. The Achilles’ heel of senescent cells: From transcriptome to senolytic drugs. Aging Cell 2015; 14: 644–658, doi: 10.1111/acel.12344. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Kirkland JL, Tchkonia T. Senolytic drugs: From discovery to translation. J Intern Med 2020; 288: 518–536, doi: 10.1111/joim.13141. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Wissler Gerdes EO, Zhu Y, Tchkonia T, Kirkland JL. Discovery, development, and future application of senolytics: Theories and predictions. FEBS J 2020; 287: 2418–2427, doi: 10.1111/febs.15264. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Xu M, Palmer AK, Ding H, Weivoda MM, Pirtskhalava T, White TA, et al. Targeting senescent cells enhances adipogenesis and metabolic function in old age. Elife 2015; 4: e12997, doi: 10.7554/eLife.12997. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Xu M, Pirtskhalava T, Farr JN, Weigand BM, Palmer AK, Weivoda MM, et al. Senolytics improve physical function and increase lifespan in old age. Nat Med 2018; 24: 1246–1256, doi: 10.1038/s41591-018-0092-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Chang J, Wang Y, Shao L, Laberge RM, Demaria M, Campisi J, et al. Clearance of senescent cells by ABT263 rejuvenates aged hematopoietic stem cells in mice. Nat Med 2016; 22: 78–83, doi: 10.1038/nm.4010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Zhu Y, Tchkonia T, Fuhrmann-Stroissnigg H, Dai HM, Ling YY, Stout MB, 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: 10.1111/acel.12445. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Zhu Y, Doornebal EJ, Pirtskhalava T, Giorgadze N, Wentworth M, Fuhrmann-Stroissnigg H, et al. New agents that target senescent cells: The flavone, fisetin, and the BCL-X(L) inhibitors, A1331852 and A1155463. Aging (Albany NY) 2017; 9: 955–963, doi: 10.18632/aging.101202. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Yousefzadeh MJ, Zhu Y, McGowan SJ, Angelini L, Fuhrmann-Stroissnigg H, Xu M, et al. Fisetin is a senotherapeutic that extends health and lifespan. EBioMedicine 2018; 36: 18–28, doi: 10.1016/j.ebiom.2018.09.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Wang Y, Chang J, Liu X, Zhang X, Zhang S, Zhang X, et al. Discovery of piperlongumine as a potential novel lead for the development of senolytic agents. Aging (Albany NY) 2016; 8: 2915–2926, doi: 10.18632/aging.101100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Xu Q, Fu Q, Li Z, Liu H, Wang Y, Lin X, et al. The flavonoid procyanidin C1 has senotherapeutic activity and increases lifespan in mice. Nat Metab 2021; 3: 1706–1726, doi: 10.1038/s42255-021-00491-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.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, doi: 10.1016/j.cell.2017.02.031. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Kirkland JL, Tchkonia T. Cellular senescence: A translational perspective. EBioMedicine 2017; 21: 21–28, doi: 10.1016/j.ebiom.2017.04.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.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: 232–236, doi: 10.1038/nature10600. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.He S, Sharpless NE. Senescence in health and disease. Cell 2017; 169: 1000–1011, doi: 10.1016/j.cell.2017.05.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Triana-Martínez F, Picallos-Rabina P, Da Silva-Álvarez S, Pietrocola F, Llanos S, Rodilla V, et al. Identification and characterization of cardiac glycosides as senolytic compounds. Nat Commun 2019; 10: 4731, doi: 10.1038/s41467-019-12888-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Guerrero A, Herranz N, Sun B, Wagner V, Gallage S, Guiho R, et al. Cardiac glycosides are broad-spectrum senolytics. Nat Metab 2019; 1: 1074–1088, doi: 10.1038/s42255-019-0122-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Johmura Y, Yamanaka T, Omori S, Wang TW, Sugiura Y, Matsumoto M, et al. Senolysis by glutaminolysis inhibition ameliorates various age-associated disorders. Science 2021; 371: 265–270, doi: 10.1126/science.abb5916. [DOI] [PubMed] [Google Scholar]
- 25.Guerrero A, Guiho R, Herranz N, Uren A, Withers DJ, Martínez-Barbera JP, et al. Galactose-modified duocarmycin prodrugs as senolytics. Aging Cell 2020; 19: e13133, doi: 10.1111/acel.13133. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Suda M, Shimizu I, Katsuumi G, Yoshida Y, Hayashi Y, Ikegami R, et al. Senolytic vaccination improves normal and pathological age-related phenotypes and increases lifespan in progeroid mice. Nat Aging 2021; 1: 1117–1126, doi: 10.1038/s43587-021-00151-2. [DOI] [PubMed] [Google Scholar]
- 27.Palmer AK, Tchkonia T, Kirkland JL. Targeting cellular senescence in metabolic disease. Mol Metab 2022; 66: 101601, doi: 10.1016/j.molmet.2022.101601. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Farr JN, Xu M, Weivoda MM, Monroe DG, Fraser DG, Onken JL, et al. Targeting cellular senescence prevents age-related bone loss in mice. Nat Med 2017; 23: 1072–1079, doi: 10.1038/nm.4385. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Yoshida S, Nakagami H, Hayashi H, Ikeda Y, Sun J, Tenma A, et al. The CD153 vaccine is a senotherapeutic option for preventing the accumulation of senescent T cells in mice. Nat Commun 2020; 11: 2482, doi: 10.1038/s41467-020-16347-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Poblocka M, Bassey AL, Smith VM, Falcicchio M, Manso AS, Althubiti M, et al. Targeted clearance of senescent cells using an antibody-drug conjugate against a specific membrane marker. Sci Rep 2021; 11: 20358, doi: 10.1038/s41598-021-99852-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Amor C, Feucht J, Leibold J, Ho YJ, Zhu C, Alonso-Curbelo D, et al. Senolytic CAR T cells reverse senescence-associated pathologies. Nature 2020; 583: 127–132, doi: 10.1038/s41586-020-2403-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Lewis-McDougall FC, Ruchaya PJ, Domenjo-Vila E, Shin Teoh T, Prata L, Cottle BJ, et al. Aged-senescent cells contribute to impaired heart regeneration. Aging Cell 2019; 18: e12931, doi: 10.1111/acel.12931. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Anderson R, Lagnado A, Maggiorani D, Walaszczyk A, Dookun E, Chapman J, et al. Length-independent telomere damage drives post-mitotic cardiomyocyte senescence. EMBO J 2019; 38: e100492, doi: 10.15252/embj.2018100492. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Roos CM, Zhang B, Palmer AK, Ogrodnik MB, Pirtskhalava T, Thalji NM, et al. Chronic senolytic treatment alleviates established vasomotor dysfunction in aged or atherosclerotic mice. Aging Cell 2016; 15: 973–977, doi: 10.1111/acel.12458. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Childs BG, Baker DJ, Wijshake T, Conover CA, Campisi J, van Deursen JM. Senescent intimal foam cells are deleterious at all stages of atherosclerosis. Science 2016; 354: 472–477, doi: 10.1126/science.aaf6659. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Wang B, Wang L, Gasek NS, Zhou Y, Kim T, Guo C, et al. An inducible p21-Cre mouse model to monitor and manipulate p21-highly-expressing senescent cells in vivo. Nat Aging 2021; 1: 962–973, doi: 10.1038/s43587-021-00107-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Xu M, Bradley EW, Weivoda MM, Hwang SM, Pirtskhalava T, Decklever T, et al. Transplanted senescent cells induce an osteoarthritis-like condition in mice. J Gerontol A Biol Sci Med Sci 2017; 72: 780–785, doi: 10.1093/gerona/glw154. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Schafer MJ, White TA, Iijima K, Haak AJ, Ligresti G, Atkinson EJ, et al. Cellular senescence mediates fibrotic pulmonary disease. Nat Commun 2017; 8: 14532, doi: 10.1038/ncomms14532. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Ogrodnik M, Zhu Y, Langhi LGP, Tchkonia T, Krüger P, Fielder E, et al. Obesity-induced cellular senescence drives anxiety and impairs neurogenesis. Cell Metab 2019; 29: 1061–1077. e1068, doi: 10.1016/j.cmet.2018.12.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.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, doi: 10.1111/acel.13296. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Fatt MP, Tran LM, Vetere G, Storer MA, Simonetta JV, Miller FD, et al. Restoration of hippocampal neural precursor function by ablation of senescent cells in the aging stem cell niche. Stem Cell Rep 2022; 17: 259–275, doi: 10.1016/j.stemcr.2021.12.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.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: 578–582, doi: 10.1038/s41586-018-0543-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Chinta SJ, Woods G, Demaria M, Rane A, Zou Y, McQuade A, et al. Cellular senescence is induced by the environmental neurotoxin paraquat and contributes to neuropathology linked to Parkinson’s disease. Cell Rep 2018; 22: 930–940, doi: 10.1016/j.celrep.2017.12.092. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Kim SR, Puranik AS, Jiang K, Chen X, Zhu XY, Taylor I, et al. Progressive Cellular senescence mediates renal dysfunction in ischemic nephropathy. J Am Soc Nephrol 2021; 32: 1987–2004, doi: 10.1681/asn.2020091373. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.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, doi: 10.1111/acel.12927. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Jeon OH, Kim C, Laberge RM, Demaria M, Rathod S, Vasserot AP, et al. Local clearance of senescent cells attenuates the development of post-traumatic osteoarthritis and creates a pro-regenerative environment. Nat Med 2017; 23: 775–781, doi: 10.1038/nm.4324. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Ogrodnik M, Miwa S, Tchkonia T, Tiniakos D, Wilson CL, Lahat A, et al. Cellular senescence drives age-dependent hepatic steatosis. Nat Commun 2017; 8: 15691, doi: 10.1038/ncomms15691. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 2006; 126: 663–676, doi: 10.1016/j.cell.2006.07.024. [DOI] [PubMed] [Google Scholar]
- 49.Lapasset L, Milhavet O, Prieur A, Besnard E, Babled A, Aït-Hamou N, et al. Rejuvenating senescent and centenarian human cells by reprogramming through the pluripotent state. Genes Dev 2011; 25: 2248–2253, doi: 10.1101/gad.173922.111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Liu GH, Barkho BZ, Ruiz S, Diep D, Qu J, Yang SL, et al. Recapitulation of premature ageing with iPSCs from Hutchinson-Gilford progeria syndrome. Nature 2011; 472: 221–225, doi: 10.1038/nature09879. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Zhang J, Lian Q, Zhu G, Zhou F, Sui L, Tan C, et al. A human iPSC model of Hutchinson Gilford Progeria reveals vascular smooth muscle and mesenchymal stem cell defects. Cell Stem Cell 2011; 8: 31–45, doi: 10.1016/j.stem.2010.12.002. [DOI] [PubMed] [Google Scholar]
- 52.Abad M, Mosteiro L, Pantoja C, Cañamero M, Rayon T, Ors I, et al. Reprogramming in vivo produces teratomas and iPS cells with totipotency features. Nature 2013; 502: 340–345, doi: 10.1038/nature12586. [DOI] [PubMed] [Google Scholar]
- 53.Ohnishi K, Semi K, Yamamoto T, Shimizu M, Tanaka A, Mitsunaga K, et al. Premature termination of reprogramming in vivo leads to cancer development through altered epigenetic regulation. Cell 2014; 156: 663–677, doi: 10.1016/j.cell.2014.01.005. [DOI] [PubMed] [Google Scholar]
- 54.Ocampo A, Reddy P, Martinez-Redondo P, Platero-Luengo A, Hatanaka F, Hishida T, et al. In vivo amelioration of age-associated hallmarks by partial reprogramming. Cell 2016; 167: 1719–1733.e1712, doi: 10.1016/j.cell.2016.11.052. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Browder KC, Reddy P, Yamamoto M, Haghani A, Guillen IG, Sahu S, et al. In vivo partial reprogramming alters age-associated molecular changes during physiological aging in mice. Nat Aging 2022; 2: 243–253, doi: 10.1038/s43587-022-00183-2. [DOI] [PubMed] [Google Scholar]
- 56.Moiseeva O, Deschênes-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-κB activation. Aging Cell 2013; 12: 489–498, doi: 10.1111/acel.12075. [DOI] [PubMed] [Google Scholar]
- 57.UK Prospective Diabetes Study (UKPDS) Group. Effect of intensive blood-glucose control with metformin on complications in overweight patients with type 2 diabetes (UKPDS 34). Lancet 1998; 352: 854–865. [PubMed] [Google Scholar]
- 58.Knowler WC, Barrett-Connor E, Fowler SE, Hamman RF, Lachin JM, Walker EA, et al. Reduction in the incidence of type 2 diabetes with lifestyle intervention or metformin. N Engl J Med 2002; 346: 393–403, doi: 10.1056/NEJMoa012512. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Barzilai N, Crandall JP, Kritchevsky SB, Espeland MA. Metformin as a tool to target aging. Cell Metab 2016; 23: 1060–1065, doi: 10.1016/j.cmet.2016.05.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Ramos FJ, Chen SC, Garelick MG, Dai DF, Liao CY, Schreiber KH, et al. Rapamycin reverses elevated mTORC1 signaling in lamin A/C-deficient mice, rescues cardiac and skeletal muscle function, and extends survival. Sci Transl Med 2012; 4: 144ra103, doi: 10.1126/scitranslmed.3003802. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Wang G, Zhu X, Xie W, Han P, Li K, Sun Z, et al. Rad as a novel regulator of excitation-contraction coupling and beta-adrenergic signaling in heart. Circ Res 2010; 106: 317–327, doi: 10.1161/circresaha.109.208272. [DOI] [PubMed] [Google Scholar]
- 62.Laberge RM, Sun Y, Orjalo AV, Patil CK, Freund A, Zhou L, et al. MTOR regulates the pro-tumorigenic senescence-associated secretory phenotype by promoting IL1A translation. Nat Cell Biol 2015; 17: 1049–1061, doi: 10.1038/ncb3195. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.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, doi: 10.1371/journal.pone.0009979. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Hurez V, Dao V, Liu A, Pandeswara S, Gelfond J, Sun L, et al. Chronic mTOR inhibition in mice with rapamycin alters T, B, myeloid, and innate lymphoid cells and gut flora and prolongs life of immune-deficient mice. Aging Cell 2015; 14: 945–956, doi: 10.1111/acel.12380. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Singh M, Jensen MD, Lerman A, Kushwaha S, Rihal CS, Gersh BJ, et al. Effect of low-dose rapamycin on senescence markers and physical functioning in older adults with coronary artery disease: Results of a pilot study. J Frailty Aging 2016; 5: 204–207, doi: 10.14283/jfa.2016.112. [DOI] [PubMed] [Google Scholar]
- 66.Justice JN, Ferrucci L, Newman AB, Aroda VR, Bahnson JL, Divers J, et al. A framework for selection of blood-based biomarkers for geroscience-guided clinical trials: Report from the TAME Biomarkers Workgroup. Geroscience 2018; 40: 419–436, doi: 10.1007/s11357-018-0042-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Yu S, Kim SR, Jiang K, Ogrodnik M, Zhu XY, Ferguson CM, et al. Quercetin reverses cardiac systolic dysfunction in mice fed with a high-fat diet: Role of angiogenesis. Oxid Med Cell Longev 2021; 2021: 8875729, doi: 10.1155/2021/8875729. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Panchal SK, Poudyal H, Brown L. Quercetin ameliorates cardiovascular, hepatic, and metabolic changes in diet-induced metabolic syndrome in rats. J Nutr 2012; 142: 1026–1032, doi: 10.3945/jn.111.157263. [DOI] [PubMed] [Google Scholar]
- 69.Iske J, Seyda M, Heinbokel T, Maenosono R, Minami K, Nian Y, et al. Senolytics prevent mt-DNA-induced inflammation and promote the survival of aged organs following transplantation. Nat Commun 2020; 11: 4289, doi: 10.1038/s41467-020-18039-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Jia K, Dai Y, Liu A, Li X, Wu L, Lu L, et al. Senolytic agent navitoclax inhibits angiotensin II-induced heart failure in mice. J Cardiovasc Pharmacol 2020; 76: 452–460, doi: 10.1097/fjc.0000000000000878. [DOI] [PubMed] [Google Scholar]
- 71.Ho JE, Enserro D, Brouwers FP, Kizer JR, Shah SJ, Psaty BM, et al. Predicting Heart failure with preserved and reduced ejection fraction: The International Collaboration on Heart Failure Subtypes. Circ Heart Fail 2016; 9: e003116, doi: 10.1161/circheartfailure.115.003116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Dunlay SM, Roger VL, Redfield MM. Epidemiology of heart failure with preserved ejection fraction. Nat Rev Cardiol 2017; 14: 591–602, doi: 10.1038/nrcardio.2017.65. [DOI] [PubMed] [Google Scholar]
- 73.Wong CM, Hawkins NM, Jhund PS, MacDonald MR, Solomon SD, Granger CB, et al. Clinical characteristics and outcomes of young and very young adults with heart failure: The CHARM programme (Candesartan in Heart Failure Assessment of Reduction in Mortality and Morbidity). J Am Coll Cardiol 2013; 62: 1845–1854, doi: 10.1016/j.jacc.2013.05.072. [DOI] [PubMed] [Google Scholar]
- 74.Gerber Y, Weston SA, Redfield MM, Chamberlain AM, Manemann SM, Jiang R, et al. A contemporary appraisal of the heart failure epidemic in Olmsted County, Minnesota, 2000 to 2010. JAMA Intern Med 2015; 175: 996–1004, doi: 10.1001/jamainternmed.2015.0924. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Tateno K, Minamino T, Moriya J, Katada A, Yokoyama M, Miura K, et al. Cell therapy for cardiovascular diseases. Ann Vasc Dis 2008; 1: 66–79, doi: 10.3400/avd.AVDsr00108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Oda M, Toba K, Ozawa T, Kato K, Yanagawa T, Ikarashi N, et al. Establishment of culturing system for ex-vivo expansion of angiogenic immature erythroid cells, and its application for treatment of patients with chronic severe lower limb ischemia. J Mol Cell Cardiol 2010; 49: 347–353, doi: 10.1016/j.yjmcc.2010.04.001. [DOI] [PubMed] [Google Scholar]
- 77.Tateno K, Minamino T, Toko H, Akazawa H, Shimizu N, Takeda S, et al. Critical roles of muscle-secreted angiogenic factors in therapeutic neovascularization. Circ Res 2006; 98: 1194–1202, doi: 10.1161/01.Res.0000219901.13974.15. [DOI] [PubMed] [Google Scholar]
- 78.Sunderland P, Alshammari L, Ambrose E, Torella D, Ellison-Hughes GM. Senolytics rejuvenate the reparative activity of human cardiomyocytes and endothelial cells. J Cardiovasc Aging 2023; 3: 21, doi: 10.20517/jca.2023.07. [DOI] [Google Scholar]
- 79.Marino F, Scalise M, Salerno N, Salerno L, Molinaro C, Cappetta D, et al. Diabetes-induced cellular senescence and senescence-associated secretory phenotype impair cardiac regeneration and function independently of age. Diabetes 2022; 71: 1081–1098, doi: 10.2337/db21-0536. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Dookun E, Walaszczyk A, Redgrave R, Palmowski P, Tual-Chalot S, Suwana A, et al. Clearance of senescent cells during cardiac ischemia-reperfusion injury improves recovery. Aging Cell 2020; 19: e13249, doi: 10.1111/acel.13249. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Walaszczyk A, Dookun E, Redgrave R, Tual-Chalot S, Victorelli S, Spyridopoulos I, et al. Pharmacological clearance of senescent cells improves survival and recovery in aged mice following acute myocardial infarction. Aging Cell 2019; 18: e12945, doi: 10.1111/acel.12945. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Kim C, Lee SG, Lim S, Jung M, Kwon SP, Hong J, et al. A senolytic-eluting coronary stent for the prevention of in-stent restenosis. ACS Biomater Sci Eng 2022; 8: 1921–1929, doi: 10.1021/acsbiomaterials.1c01611. [DOI] [PubMed] [Google Scholar]
- 83.Lakshminarayan K, Solid CA, Collins AJ, Anderson DC, Herzog CA. Atrial fibrillation and stroke in the general medicare population: A 10-year perspective (1992 to 2002). Stroke 2006; 37: 1969–1974, doi: 10.1161/01.Str.0000230607.07928.17. [DOI] [PubMed] [Google Scholar]
- 84.Go AS, Hylek EM, Phillips KA, Chang Y, Henault LE, Selby JV, et al. Prevalence of diagnosed atrial fibrillation in adults: national implications for rhythm management and stroke prevention: The AnTicoagulation and Risk Factors in Atrial Fibrillation (ATRIA) Study. JAMA 2001; 285: 2370–2375, doi: 10.1001/jama.285.18.2370. [DOI] [PubMed] [Google Scholar]
- 85.Jesel L, Abbas M, Park SH, Matsushita K, Kindo M, Hasan H, et al. Atrial fibrillation progression is associated with cell senescence burden as determined by p53 and p16 expression. J Clin Med 2019; 9: 36, doi: 10.3390/jcm9010036. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Adili A, Zhu X, Cao H, Tang X, Wang Y, Wang J, et al. Atrial fibrillation underlies cardiomyocyte senescence and contributes to deleterious atrial remodeling during disease progression. Aging Dis 2022; 13: 298–312, doi: 10.14336/ad.2021.0619. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Xie J, Chen Y, Hu C, Pan Q, Wang B, Li X, et al. Premature senescence of cardiac fibroblasts and atrial fibrosis in patients with atrial fibrillation. Oncotarget 2017; 8: 57981–57990, doi: 10.18632/oncotarget.19853. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Wang H, Jiang W, Hu Y, Wan Z, Bai H, Yang Q, et al. Quercetin improves atrial fibrillation through inhibiting TGF-β/Smads pathway via promoting MiR-135b expression. Phytomedicine 2021; 93: 153774, doi: 10.1016/j.phymed.2021.153774. [DOI] [PubMed] [Google Scholar]
- 89.Liu L, Gan S, Li B, Ge X, Yu H, Zhou H. Fisetin alleviates atrial inflammation, remodeling, and vulnerability to atrial fibrillation after myocardial infarction. Int Heart J 2019; 60: 1398–1406, doi: 10.1536/ihj.19-131. [DOI] [PubMed] [Google Scholar]
- 90.Schüttler D, Bapat A, Kääb S, Lee K, Tomsits P, Clauss S, et al. Animal models of atrial fibrillation. Circ Res 2020; 127: 91–110, doi: 10.1161/circresaha.120.316366. [DOI] [PubMed] [Google Scholar]
- 91.Garrido AM, Kaistha A, Uryga AK, Oc S, Foote K, Shah A, et al. Efficacy and limitations of senolysis in atherosclerosis. Cardiovasc Res 2022; 118: 1713–1727, doi: 10.1093/cvr/cvab208. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Suda M, Shimizu I, Katsuumi G, Hsiao CL, Yoshida Y, Matsumoto N, et al. Glycoprotein nonmetastatic melanoma protein B regulates lysosomal integrity and lifespan of senescent cells. Sci Rep 2022; 12: 6522, doi: 10.1038/s41598-022-10522-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Shi H, Mao X, Zhong Y, Liu Y, Zhao X, Yu K, et al. Digoxin reduces atherosclerosis in apolipoprotein E-deficient mice. Br J Pharmacol 2016; 173: 1517–1528, doi: 10.1111/bph.13453. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Lazaro I, Oguiza A, Recio C, Mallavia B, Madrigal-Matute J, Blanco J, et al. Targeting HSP90 ameliorates nephropathy and atherosclerosis through suppression of NF-κB and STAT signaling pathways in diabetic mice. Diabetes 2015; 64: 3600–3613, doi: 10.2337/db14-1926. [DOI] [PubMed] [Google Scholar]
- 95.Childs BG, Zhang C, Shuja F, Sturmlechner I, Trewartha S, Velasco RF, et al. Senescent cells suppress innate smooth muscle cell repair functions in atherosclerosis. Nat Aging 2021; 1: 698–714, doi: 10.1038/s43587-021-00089-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Parvizi M, Franchi F, Arendt BK, Ebtehaj S, Rodriguez-Porcel M, Lanza IR. Senolytic agents lessen the severity of abdominal aortic aneurysm in aged mice. Exp Gerontol 2021; 151: 111416, doi: 10.1016/j.exger.2021.111416. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.van der Feen DE, Bossers GPL, Hagdorn QAJ, Moonen JR, Kurakula K, Szulcek R, et al. Cellular senescence impairs the reversibility of pulmonary arterial hypertension. Sci Transl Med 2020; 12: eaaw4974, doi: 10.1126/scitranslmed.aaw4974. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Wakasugi T, Shimizu I, Yoshida Y, Hayashi Y, Ikegami R, Suda M, et al. Role of smooth muscle cell p53 in pulmonary arterial hypertension. PLoS One 2019; 14: e0212889, doi: 10.1371/journal.pone.0212889. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Nath KA, O’Brien DR, Croatt AJ, Grande JP, Ackerman AW, Nath MC, et al. The murine dialysis fistula model exhibits a senescence phenotype: Pathobiological mechanisms and therapeutic potential. Am J Physiol Renal Physiol 2018; 315: F1493–F1499, doi: 10.1152/ajprenal.00308.2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Crespo-Garcia S, Tsuruda PR, Dejda A, Ryan RD, Fournier F, Chaney SY, et al. Pathological angiogenesis in retinopathy engages cellular senescence and is amenable to therapeutic elimination via BCL-xL inhibition. Cell Metab 2021; 33: 818–832. e817, doi: 10.1016/j.cmet.2021.01.011. [DOI] [PubMed] [Google Scholar]
- 101.Justice JN, Nambiar AM, Tchkonia T, LeBrasseur NK, Pascual R, Hashmi SK, et al. Senolytics in idiopathic pulmonary fibrosis: Results from a first-in-human, open-label, pilot study. EBioMedicine 2019; 40: 554–563, doi: 10.1016/j.ebiom.2018.12.052. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Hickson LJ, Langhi Prata LGP, Bobart SA, Evans TK, Giorgadze N, Hashmi SK, 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: 10.1016/j.ebiom.2019.08.069. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Dagher O, Mury P, Noly PE, Fortier A, Lettre G, Thorin E, et al. Design of a randomized placebo-controlled trial to evaluate the anti-inflammatory and senolytic effects of quercetin in patients undergoing coronary artery bypass graft surgery. Front Cardiovasc Med 2021; 8: 741542, doi: 10.3389/fcvm.2021.741542. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Suda M, Shimizu I, Yoshida Y, Hayashi Y, Ikegami R, Katsuumi G, et al. Inhibition of dipeptidyl peptidase-4 ameliorates cardiac ischemia and systolic dysfunction by up-regulating the FGF-2/EGR-1 pathway. PLoS One 2017; 12: e0182422, doi: 10.1371/journal.pone.0182422. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Forman DE, Maurer MS, Boyd C, Brindis R, Salive ME, Horne FM, et al. Multimorbidity in older adults with cardiovascular disease. J Am Coll Cardiol 2018; 71: 2149–2161, doi: 10.1016/j.jacc.2018.03.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Dunlay SM, Chamberlain AM. Multimorbidity in older patients with cardiovascular disease. Curr Cardiovasc Risk Rep 2016; 10: 3, doi: 10.1007/s12170-016-0491-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Liu Y, Sanoff HK, Cho H, Burd CE, Torrice C, Ibrahim JG, et al. Expression of p16(INK4a) in peripheral blood T-cells is a biomarker of human aging. Aging Cell 2009; 8: 439–448, doi: 10.1111/j.1474-9726.2009.00489.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Englund DA, Sakamoto AE, Fritsche CM, Heeren AA, Zhang X, Kotajarvi BR, et al. Exercise reduces circulating biomarkers of cellular senescence in humans. Aging Cell 2021; 20: e13415, doi: 10.1111/acel.13415. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Zhu Y, Prata L, Gerdes EOW, Netto JME, Pirtskhalava T, Giorgadze N, et al. Orally-active, clinically-translatable senolytics restore α-Klotho in mice and humans. EBioMedicine 2022; 77: 103912, doi: 10.1016/j.ebiom.2022.103912. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Tchkonia T, Zhu Y, van Deursen J, Campisi J, Kirkland JL. Cellular senescence and the senescent secretory phenotype: Therapeutic opportunities. J Clin Invest 2013; 123: 966–972, doi: 10.1172/jci64098. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Saul D, Kosinsky RL, Atkinson EJ, Doolittle ML, Zhang X, LeBrasseur NK, et al. A new gene set identifies senescent cells and predicts senescence-associated pathways across tissues. Nat Commun 2022; 13: 4827, doi: 10.1038/s41467-022-32552-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Munk R, Panda AC, Grammatikakis I, Gorospe M, Abdelmohsen K. Senescence-associated microRNAs. Int Rev Cell Mol Biol 2017; 334: 177–205, doi: 10.1016/bs.ircmb.2017.03.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Suh N. MicroRNA controls of cellular senescence. BMB Rep 2018; 51: 493–499, doi: 10.5483/BMBRep.2018.51.10.209. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Misawa T, Hitomi K, Miyata K, Tanaka Y, Fujii R, Chiba M, et al. Identification of novel senescent markers in small extracellular vesicles. Int J Mol Sci 2023; 24: 2421, doi: 10.3390/ijms24032421. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Tanaka Y, Takahashi A. Senescence-associated extracellular vesicle release plays a role in senescence-associated secretory phenotype (SASP) in age-associated diseases. J Biochem 2021; 169: 147–153, doi: 10.1093/jb/mvaa109. [DOI] [PubMed] [Google Scholar]
- 116.Mehdizadeh M, Aguilar M, Thorin E, Ferbeyre G, Nattel S. The role of cellular senescence in cardiac disease: Basic biology and clinical relevance. Nat Rev Cardiol 2022; 19: 250–264, doi: 10.1038/s41569-021-00624-2. [DOI] [PubMed] [Google Scholar]
- 117.Hu C, Zhang X, Teng T, Ma ZG, Tang QZ. Cellular senescence in cardiovascular diseases: A systematic review. Aging Dis 2022; 13: 103–128, doi: 10.14336/ad.2021.0927. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Suda M, Paul KH, Minamino T, Miller JD, Lerman A, Ellison-Hughes GM, et al. Senescent cells: A therapeutic target in cardiovascular diseases. Cells 2023; 12: 1296, doi: 10.3390/cells12091296. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Lérida-Viso A, Estepa-Fernández A, Morellá-Aucejo Á, Lozano-Torres B, Alfonso M, Blandez JF, et al. Pharmacological senolysis reduces doxorubicin-induced cardiotoxicity and improves cardiac function in mice. Pharmacol Res 2022; 183: 106356, doi: 10.1016/j.phrs.2022.106356. [DOI] [PubMed] [Google Scholar]
- 120.Salaami O, Kuo CL, Drake MT, Kuchel GA, Kirkland JL, Pignolo RJ. Antidiabetic effects of the senolytic agent dasatinib. Mayo Clin Proc 2021; 96: 3021–3029, doi: 10.1016/j.mayocp.2021.06.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Dookun E, Passos JF, Arthur HM, Richardson GD. Therapeutic potential of senolytics in cardiovascular disease. Cardiovasc Drugs Ther 2022; 36: 187–196, doi: 10.1007/s10557-020-07075-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Lee JR, Park BW, Park JH, Lim S, Kwon SP, Hwang JW, et al. Local delivery of a senolytic drug in ischemia and reperfusion-injured heart attenuates cardiac remodeling and restores impaired cardiac function. Acta Biomater 2021; 135: 520–533, doi: 10.1016/j.actbio.2021.08.028. [DOI] [PubMed] [Google Scholar]
