The redox-senescence axis and its therapeutic targeting
Tài liệu tham khảo
Hayflick, 1961, The serial cultivation of human diploid cell strains, Exp. Cell Res., 25, 585, 10.1016/0014-4827(61)90192-6
Harley, 1990, Telomeres shorten during ageing of human fibroblasts, Nature, 345, 458, 10.1038/345458a0
Petrova, 2016, Small molecule compounds that induce cellular senescence, Aging Cell, 15, 999, 10.1111/acel.12518
Hirama, 1995, Role of the cyclin-dependent kinase inhibitors in the development of cancer, Blood, 86, 841, 10.1182/blood.V86.3.841.841
Xiong, 1993, p21 is a universal inhibitor of cyclin kinases, Nature, 366, 701, 10.1038/366701a0
Harper, 1993, The p21 Cdk-interacting protein Cip1 is a potent inhibitor of G1 cyclin-dependent kinases, Cell, 75, 805, 10.1016/0092-8674(93)90499-G
Qian, 2013, Senescence regulation by the p53 protein family, Methods Mol. Biol., 965, 37, 10.1007/978-1-62703-239-1_3
Qian, 2010, Tumor suppression by p53: making cells senescent, Histol. Histopathol., 25, 515
Munoz-Espin, 2013, Programmed cell senescence during mammalian embryonic development, Cell, 155, 1104, 10.1016/j.cell.2013.10.019
Storer, 2013, Senescence is a developmental mechanism that contributes to embryonic growth and patterning, Cell, 155, 1119, 10.1016/j.cell.2013.10.041
Demaria, 2014, An essential role for senescent cells in optimal wound healing through secretion of PDGF-AA, Dev. Cell, 31, 722, 10.1016/j.devcel.2014.11.012
Chiche, 2017, Injury-induced senescence enables in vivo reprogramming in skeletal muscle, Cell Stem Cell, 20, 407, 10.1016/j.stem.2016.11.020
Martinez-Cue, 2020, Cellular senescence in neurodegenerative diseases, Front. Cell. Neurosci., 14, 16, 10.3389/fncel.2020.00016
Baker, 2011, Clearance of p16Ink4a-positive senescent cells delays ageing-associated disorders, Nature, 479, 232, 10.1038/nature10600
Song, 2020, Immune clearance of senescent cells to combat ageing and chronic diseases, Cells, 9, 10.3390/cells9030671
Karin, 2019, Senescent cell turnover slows with age providing an explanation for the Gompertz law, Nat. Commun., 10, 5495, 10.1038/s41467-019-13192-4
Zhang, 2007, Molecular signaling and genetic pathways of senescence: its role in tumorigenesis and aging, J. Cell. Physiol., 210, 567, 10.1002/jcp.20919
Coppe, 2008, Senescence-associated secretory phenotypes reveal cell-nonautonomous functions of oncogenic RAS and the p53 tumor suppressor, PLoS Biol., 6, 2853, 10.1371/journal.pbio.0060301
Coppe, 2010, The senescence-associated secretory phenotype: the dark side of tumor suppression, Annu. Rev. Pathol., 5, 99, 10.1146/annurev-pathol-121808-102144
Coussens, 2002, Inflammation and cancer, Nature, 420, 860, 10.1038/nature01322
Wajapeyee, 2008, Oncogenic BRAF induces senescence and apoptosis through pathways mediated by the secreted protein IGFBP7, Cell, 132, 363, 10.1016/j.cell.2007.12.032
Acosta, 2013, A complex secretory program orchestrated by the inflammasome controls paracrine senescence, Nat. Cell Biol., 15, 978, 10.1038/ncb2784
Eggert, 2016, Distinct functions of senescence-associated immune responses in liver tumor surveillance and tumor progression, Canc. Cell, 30, 533, 10.1016/j.ccell.2016.09.003
Olivieri, 2013, MiR-146a as marker of senescence-associated pro-inflammatory status in cells involved in vascular remodelling, Age (Dordr), 35, 1157, 10.1007/s11357-012-9440-8
Hayashi, 2006, Endothelial cellular senescence is inhibited by nitric oxide: implications in atherosclerosis associated with menopause and diabetes, Proc. Natl. Acad. Sci. U. S. A., 103, 17018, 10.1073/pnas.0607873103
Behrendt, 2002, Endothelial function. From vascular biology to clinical applications, Am. J. Cardiol., 90, 40L, 10.1016/S0002-9149(02)02963-6
Coppe, 2011, Tumor suppressor and aging biomarker p16(INK4a) induces cellular senescence without the associated inflammatory secretory phenotype, J. Biol. Chem., 286, 36396, 10.1074/jbc.M111.257071
Rodier, 2009, Persistent DNA damage signalling triggers senescence-associated inflammatory cytokine secretion, Nat. Cell Biol., 11, 973, 10.1038/ncb1909
Freund, 2011, p38MAPK is a novel DNA damage response-independent regulator of the senescence-associated secretory phenotype, EMBO J., 30, 1536, 10.1038/emboj.2011.69
Rodier, 2011, DNA-SCARS: distinct nuclear structures that sustain damage-induced senescence growth arrest and inflammatory cytokine secretion, J. Cell Sci., 124, 68, 10.1242/jcs.071340
Kuilman, 2008, Oncogene-induced senescence relayed by an interleukin-dependent inflammatory network, Cell, 133, 1019, 10.1016/j.cell.2008.03.039
Chen, 2015, MacroH2A1 and ATM play opposing roles in paracrine senescence and the senescence-associated secretory phenotype, Mol. Cell., 59, 719, 10.1016/j.molcel.2015.07.011
Xu, 2015, JAK inhibition alleviates the cellular senescence-associated secretory phenotype and frailty in old age, Proc. Natl. Acad. Sci. U. S. A., 112, E6301, 10.1073/pnas.1515386112
Wiley, 2016, Mitochondrial dysfunction induces senescence with a distinct secretory phenotype, Cell Metabol., 23, 303, 10.1016/j.cmet.2015.11.011
Dou, 2017, Cytoplasmic chromatin triggers inflammation in senescence and cancer, Nature, 550, 402, 10.1038/nature24050
Yang, 2017, cGAS is essential for cellular senescence, Proc. Natl. Acad. Sci. U. S. A., 114, E4612, 10.1073/pnas.1705499114
Laberge, 2015, MTOR regulates the pro-tumorigenic senescence-associated secretory phenotype by promoting IL1A translation, Nat. Cell Biol., 17, 1049, 10.1038/ncb3195
Zhang, 2018, The senescence-associated secretory phenotype is potentiated by feedforward regulatory mechanisms involving Zscan4 and TAK1, Nat. Commun., 9, 1723, 10.1038/s41467-018-04010-4
Kang, 2015, The DNA damage response induces inflammation and senescence by inhibiting autophagy of GATA4, Science, 349, aaa5612, 10.1126/science.aaa5612
Xue, 2007, Senescence and tumour clearance is triggered by p53 restoration in murine liver carcinomas, Nature, 445, 656, 10.1038/nature05529
Pereira, 2019, Senescent cells evade immune clearance via HLA-E-mediated NK and CD8(+) T cell inhibition, Nat. Commun., 10, 2387, 10.1038/s41467-019-10335-5
Faget, 2019, Unmasking senescence: context-dependent effects of SASP in cancer, Nat. Rev. Canc., 19, 439, 10.1038/s41568-019-0156-2
Rao, 2016, SASP: tumor suppressor or promoter?, Yes! Trends Canc., 2, 676, 10.1016/j.trecan.2016.10.001
Saretzki, 2010, Cellular senescence in the development and treatment of cancer, Curr. Pharmaceut. Des., 16, 79, 10.2174/138161210789941874
Serrano, 1997, Oncogenic ras provokes premature cell senescence associated with accumulation of p53 and p16INK4a, Cell, 88, 593, 10.1016/S0092-8674(00)81902-9
Sarkisian, 2007, Dose-dependent oncogene-induced senescence in vivo and its evasion during mammary tumorigenesis, Nat. Cell Biol., 9, 493, 10.1038/ncb1567
Courtois-Cox, 2008, Many roads lead to oncogene-induced senescence, Oncogene, 27, 2801, 10.1038/sj.onc.1210950
Liu, 2019, Cells exhibiting strong p16 (INK4a) promoter activation in vivo display features of senescence, Proc. Natl. Acad. Sci. U. S. A., 116, 2603, 10.1073/pnas.1818313116
Monasor, 2013, INK4a/ARF limits the expansion of cells suffering from replication stress, Cell Cycle, 12, 1948, 10.4161/cc.25017
Zhang, 1998, ARF promotes MDM2 degradation and stabilizes p53: ARF-INK4a locus deletion impairs both the Rb and p53 tumor suppression pathways, Cell, 92, 725, 10.1016/S0092-8674(00)81401-4
Carlos, 2013, ARF triggers senescence in Brca2-deficient cells by altering the spectrum of p53 transcriptional targets, Nat. Commun., 4, 2697, 10.1038/ncomms3697
Di, 2014, IL-6 secreted from senescent mesenchymal stem cells promotes proliferation and migration of breast cancer cells, PloS One, 9, 10.1371/journal.pone.0113572
Ortiz-Montero, 2017, Senescence-associated IL-6 and IL-8 cytokines induce a self- and cross-reinforced senescence/inflammatory milieu strengthening tumorigenic capabilities in the MCF-7 breast cancer cell line, Cell Commun. Signal., 15, 17, 10.1186/s12964-017-0172-3
Coppe, 2006, Secretion of vascular endothelial growth factor by primary human fibroblasts at senescence, J. Biol. Chem., 281, 29568, 10.1074/jbc.M603307200
Pazolli, 2009, Senescent stromal-derived osteopontin promotes preneoplastic cell growth, Canc. Res., 69, 1230, 10.1158/0008-5472.CAN-08-2970
Farsam, 2016, Senescent fibroblast-derived Chemerin promotes squamous cell carcinoma migration, Oncotarget, 7, 83554, 10.18632/oncotarget.13446
Liu, 2007, Senescent human fibroblasts increase the early growth of xenograft tumors via matrix metalloproteinase secretion, Canc. Res., 67, 3117, 10.1158/0008-5472.CAN-06-3452
Hwang, 2020, Endothelial cells under therapy-induced senescence secrete CXCL11, which increases aggressiveness of breast cancer cells, Canc. Lett., 490, 100, 10.1016/j.canlet.2020.06.019
Tato-Costa, 2016, Therapy-induced cellular senescence induces epithelial-to-mesenchymal transition and increases invasiveness in rectal cancer, Clin. Colorectal Canc., 15, 170, 10.1016/j.clcc.2015.09.003
Sabisz, 2009, Cancer stem cells and escape from drug-induced premature senescence in human lung tumor cells: implications for drug resistance and in vitro drug screening models, Cell Cycle, 8, 3208, 10.4161/cc.8.19.9758
Milanovic, 2018, Senescence-associated reprogramming promotes cancer stemness, Nature, 553, 96, 10.1038/nature25167
Cruickshanks, 2013, Senescent cells harbour features of the cancer epigenome, Nat. Cell Biol., 15, 1495, 10.1038/ncb2879
Chang, 1999, Role of p53 and p21waf1/cip1 in senescence-like terminal proliferation arrest induced in human tumor cells by chemotherapeutic drugs, Oncogene, 18, 4808, 10.1038/sj.onc.1203078
Chang, 1999, A senescence-like phenotype distinguishes tumor cells that undergo terminal proliferation arrest after exposure to anticancer agents, Canc. Res., 59, 3761
Jung, 2019, mTOR kinase leads to PTEN-loss-induced cellular senescence by phosphorylating p53, Oncogene, 38, 1639, 10.1038/s41388-018-0521-8
Iannello, 2013, p53-dependent chemokine production by senescent tumor cells supports NKG2D-dependent tumor elimination by natural killer cells, J. Exp. Med., 210, 2057, 10.1084/jem.20130783
Lujambio, 2013, Non-cell-autonomous tumor suppression by p53, Cell, 153, 449, 10.1016/j.cell.2013.03.020
Saleh, 2019, Tumor cell escape from therapy-induced senescence as a model of disease recurrence after dormancy, Canc. Res., 79, 1044, 10.1158/0008-5472.CAN-18-3437
Saleh, 2019, Tumor cell escape from therapy-induced senescence, Biochem. Pharmacol., 162, 202, 10.1016/j.bcp.2018.12.013
Laud, 2005, Elevated telomere-telomere recombination in WRN-deficient, telomere dysfunctional cells promotes escape from senescence and engagement of the ALT pathway, Genes Dev., 19, 2560, 10.1101/gad.1321305
Dirac, 2003, Reversal of senescence in mouse fibroblasts through lentiviral suppression of p53, J. Biol. Chem., 278, 11731, 10.1074/jbc.C300023200
Was, 2017, Bafilomycin A1 triggers proliferative potential of senescent cancer cells in vitro and in NOD/SCID mice, Oncotarget, 8, 9303, 10.18632/oncotarget.14066
Beausejour, 2003, Reversal of human cellular senescence: roles of the p53 and p16 pathways, EMBO J., 22, 4212, 10.1093/emboj/cdg417
Sies, 1985, Hydroperoxides and thiol oxidants in the study of oxidative stress in intact cells and organs, Oxid. Stress, 1, 73, 10.1016/B978-0-12-642760-8.50008-9
Cui, 2011, Acetylation of H4K12 in porcine oocytes during in vitro aging: potential role of ooplasmic reactive oxygen species, Theriogenology, 75, 638, 10.1016/j.theriogenology.2010.09.031
Sergiev, 2015, Theories of aging: an ever-evolving field, Acta Naturae, 7, 9, 10.32607/20758251-2015-7-1-9-18
Sawada, 1987, Changes in superoxide radical and lipid peroxide formation in the brain, heart and liver during the lifetime of the rat, Mech. Ageing Dev., 41, 125, 10.1016/0047-6374(87)90057-1
Chung, 1999, Modulation of renal xanthine oxidoreductase in aging: gene expression and reactive oxygen species generation, J. Nutr. Health Aging, 3, 19
Liguori, 2018, Oxidative stress, aging, and diseases, Clin. Interv. Aging, 13, 757, 10.2147/CIA.S158513
Korolchuk, 2017, Mitochondria in cell senescence: is mitophagy the weakest link?, EBioMedicine, 21, 7, 10.1016/j.ebiom.2017.03.020
Passos, 2010, Feedback between p21 and reactive oxygen production is necessary for cell senescence, Mol. Syst. Biol., 6, 347, 10.1038/msb.2010.5
Nelson, 2018, The senescent bystander effect is caused by ROS-activated NF-kappaB signalling, Mech. Ageing Dev., 170, 30, 10.1016/j.mad.2017.08.005
Kodama, 2013, ROS-generating oxidases Nox1 and Nox4 contribute to oncogenic Ras-induced premature senescence, Gene Cell., 18, 32, 10.1111/gtc.12015
Ogrunc, 2014, Oncogene-induced reactive oxygen species fuel hyperproliferation and DNA damage response activation, Cell Death Differ., 21, 998, 10.1038/cdd.2014.16
Iskandar, 2016, Synthetic lethality of a novel small molecule against mutant KRAS-expressing cancer cells involves AKT-dependent ROS production, Antioxidants Redox Signal., 24, 781, 10.1089/ars.2015.6362
Allsopp, 1992, Telomere length predicts replicative capacity of human fibroblasts, Proc. Natl. Acad. Sci. U. S. A., 89, 10114, 10.1073/pnas.89.21.10114
Hutter, 2002, Replicative senescence of human fibroblasts: the role of Ras-dependent signaling and oxidative stress, Exp. Gerontol., 37, 1165, 10.1016/S0531-5565(02)00136-5
Saretzki, 2002, Replicative aging, telomeres, and oxidative stress, Ann. N. Y. Acad. Sci., 959, 24, 10.1111/j.1749-6632.2002.tb02079.x
Di Micco, 2008, DNA damage response activation in mouse embryonic fibroblasts undergoing replicative senescence and following spontaneous immortalization, Cell Cycle, 7, 3601, 10.4161/cc.7.22.7152
d'Adda di Fagagna, 2003, A DNA damage checkpoint response in telomere-initiated senescence, Nature, 426, 194, 10.1038/nature02118
Venkatachalam, 2017, Replication stress-induced endogenous DNA damage drives cellular senescence induced by a sub-lethal oxidative stress, Nucleic Acids Res., 45, 10564, 10.1093/nar/gkx684
Macip, 2003, Influence of induced reactive oxygen species in p53-mediated cell fate decisions, Mol. Cell Biol., 23, 8576, 10.1128/MCB.23.23.8576-8585.2003
Macip, 2002, Inhibition of p21-mediated ROS accumulation can rescue p21-induced senescence, EMBO J., 21, 2180, 10.1093/emboj/21.9.2180
Takahashi, 2006, Mitogenic signalling and the p16INK4a-Rb pathway cooperate to enforce irreversible cellular senescence, Nat. Cell Biol., 8, 1291, 10.1038/ncb1491
Sies, 2020, Reactive oxygen species (ROS) as pleiotropic physiological signalling agents, Nat. Rev. Mol. Cell Biol., 21, 363, 10.1038/s41580-020-0230-3
Sies, 2017, Oxidative stress, Annu. Rev. Biochem., 86, 715, 10.1146/annurev-biochem-061516-045037
Jones, 2015, The redox code, Antioxidants Redox Signal., 23, 734, 10.1089/ars.2015.6247
Pervaiz, 2001, Activation of the RacGTPase inhibits apoptosis in human tumor cells, Oncogene, 20, 6263, 10.1038/sj.onc.1204840
Clément, 2003, Decrease in intracellular superoxide sensitizes Bcl-2-overexpressing tumor cells to receptor and drug-induced apoptosis independent of the mitochondria, Cell Death Differ., 10, 1273, 10.1038/sj.cdd.4401302
Chen, 2007, Bcl-2 induces pro-oxidant state by engaging mitochondrial respiration in tumor cells, Cell Death Differ., 14, 1617, 10.1038/sj.cdd.4402165
Tochhawng, 2016, Gelsolin-Cu/ZnSOD interaction alters intracellular reactive oxygen species levels to promote cancer cell invasion, Oncotarget, 7, 52832, 10.18632/oncotarget.10451
Raman, 2020, Peroxynitrite promotes serine-62 phosphorylation-dependent stabilization of the oncoprotein c-Myc, Redox Biol., 34, 10.1016/j.redox.2020.101587
Hirpara, 2020, Superoxide induced inhibition of death receptor signaling is mediated via induced expression of apoptosis inhibitory protein cFLIP, Redox Biol., 30, 10.1016/j.redox.2019.101403
Chong, 2020, Serine-70 phosphorylated Bcl-2 prevents oxidative stress-induced DNA damage by modulating the mitochondrial redox metabolism, Nucleic Acids Res., 48, 12727, 10.1093/nar/gkaa1110
Furukawa, 2007, H2O2 accelerates cellular senescence by accumulation of acetylated p53 via decrease in the function of SIRT1 by NAD+ depletion, Cell. Physiol. Biochem., 20, 10.1159/000104152
Marazita, 2016, Oxidative stress-induced premature senescence dysregulates VEGF and CFH expression in retinal pigment epithelial cells: implications for Age-related Macular Degeneration, Redox Biol., 7, 78, 10.1016/j.redox.2015.11.011
Chen, 1994, Senescence-like growth arrest induced by hydrogen peroxide in human diploid fibroblast F65 cells, Proc. Natl. Acad. Sci. U. S. A., 91, 4130, 10.1073/pnas.91.10.4130
Marazita, 2016, Oxidative stress-induced premature senescence dysregulates VEGF and CFH expression in retinal pigment epithelial cells: implications for age-related macular degeneration, Redox Biol., 7, 78, 10.1016/j.redox.2015.11.011
Chen, 1994, Senescence-like growth arrest induced by hydrogen peroxide in human diploid fibroblast F65 cells, Proc. Natl. Acad. Sci. Unit. States Am., 91, 4130, 10.1073/pnas.91.10.4130
Sin, 2012, Chronic treatment with ginsenoside Rg3 induces Akt-dependent senescence in human glioma cells, Int. J. Oncol., 41, 1669, 10.3892/ijo.2012.1604
Kwak, 2015, Nicotinamide exerts antioxidative effects on senescent cells, Mol. Cell., 38, 229, 10.14348/molcells.2015.2253
Deng, 2003, Bcl2 retards G1/S cell cycle transition by regulating intracellular ROS, Blood, 102, 3179, 10.1182/blood-2003-04-1027
Debidda, 2006, Rac1 GTPase regulates cell genomic stability and senescence, J. Biol. Chem., 281, 38519, 10.1074/jbc.M604607200
Chong, 2019, A feedforward relationship between active Rac1 and phosphorylated Bcl-2 is critical for sustaining Bcl-2 phosphorylation and promoting cancer progression, Canc. Lett., 457, 151, 10.1016/j.canlet.2019.05.009
Ashraf, 2019, Senescence evasion in chemotherapy: a sweet spot for p21, Cell, 178, 267, 10.1016/j.cell.2019.06.025
Roninson, 2003, Tumor cell senescence in cancer treatment, Canc. Res., 63, 2705
Roninson, 2001, If not apoptosis, then what? Treatment-induced senescence and mitotic catastrophe in tumor cells, Drug Resist. Updates, 4, 303, 10.1054/drup.2001.0213
Guillon, 2019, Chemotherapy-induced senescence, an adaptive mechanism driving resistance and tumor heterogeneity, Cell Cycle, 18, 2385, 10.1080/15384101.2019.1652047
Achuthan, 2011, Drug-induced senescence generates chemoresistant stemlike cells with low reactive oxygen species, J. Biol. Chem., 286, 37813, 10.1074/jbc.M110.200675
Elmore, 2005, Evasion of a single-step, chemotherapy-induced senescence in breast cancer cells: implications for treatment response, Clin. Canc. Res., 11, 2637, 10.1158/1078-0432.CCR-04-1462
Collado, 2005, Senescence in premalignant tumours, Nature, 436, 10.1038/436642a
Ewald, 2010, Therapy-induced senescence in cancer, JNCI: J. Natl. Cancer Inst., 102, 1536, 10.1093/jnci/djq364
Campisi, 2011, vol. 21, 354
Beauséjour, 2003, Reversal of human cellular senescence: roles of the p53 and p16 pathways, EMBO J., 22, 4212, 10.1093/emboj/cdg417
Yu, 2003, PUMA mediates the apoptotic response to p53 in colorectal cancer cells, Proc. Natl. Acad. Sci. Unit. States Am., 100, 1931, 10.1073/pnas.2627984100
Chong, 2020, Noncanonical cell fate regulation by bcl-2 proteins, Trends Cell Biol., 30, 537, 10.1016/j.tcb.2020.03.004
Yin, 1995, BCL-2 expression delays drug-induced apoptosis but does not increase clonogenic survival after drug treatment in HeLa cells, Canc. Res., 55, 4922
Crescenzi, 2003, Bcl-2 activates a programme of premature senescence in human carcinoma cells, Biochem. J., 375, 263, 10.1042/bj20030868
Hayward, 2003, Antisense Bcl-xl down-regulation switches the response to topoisomerase I inhibition from senescence to apoptosis in colorectal cancer cells, enhancing global cytotoxicity, Clin. Canc. Res., 9, 2856
Gayle, 2019, Targeting BCL-xL improves the efficacy of bromodomain and extra-terminal protein inhibitors in triple-negative breast cancer by eliciting the death of senescent cells, J. Biol. Chem., 294, 875, 10.1074/jbc.RA118.004712
Pluquet, 2019, Connecting cancer relapse with senescence, Canc. Lett., 463, 50, 10.1016/j.canlet.2019.08.004
Kim, 2019, Senotherapeutics: emerging strategy for healthy aging and age-related disease, BMB Rep., 52, 47, 10.5483/BMBRep.2019.52.1.293
Childs, 2017, Senescent cells: an emerging target for diseases of ageing, Nat. Rev. Drug Discov., 16, 718, 10.1038/nrd.2017.116
Demaria, 2017, Cellular senescence promotes adverse effects of chemotherapy and cancer relapse, Canc. Discov., 7, 165, 10.1158/2159-8290.CD-16-0241
Myrianthopoulos, 2019, Senescence and senotherapeutics: a new field in cancer therapy, Pharmacol. Ther., 193, 31, 10.1016/j.pharmthera.2018.08.006
Kim, 2018, Quercetin and its role in biological functions: an updated review, EXCLI J., 17, 856
Feitelson, 2015, Sustained proliferation in cancer: mechanisms and novel therapeutic targets, Semin. Canc. Biol., 35, S25, 10.1016/j.semcancer.2015.02.006
Murakami, 2008, Multitargeted cancer prevention by quercetin, Canc. Lett., 269, 315, 10.1016/j.canlet.2008.03.046
Zhu, 2015, The Achilles' heel of senescent cells: from transcriptome to senolytic drugs, Aging Cell, 14, 644, 10.1111/acel.12344
Zhu, 2018, Quercetin inhibits renal cyst growth in vitro and via parenteral injection in a polycystic kidney disease mouse model, Food Funct., 9, 389, 10.1039/C7FO01253E
Nguyen, 2017, Quercetin induces apoptosis and cell cycle arrest in triple-negative breast cancer cells through modulation of Foxo3a activity, KOREAN J. PHYSIOL. PHARMACOL., 21, 205, 10.4196/kjpp.2017.21.2.205
Han, 2020, Randomised clinical trial to determine the safety of quercetin supplementation in patients with chronic obstructive pulmonary disease, BMJ Open Respir. Res., 7, 10.1136/bmjresp-2018-000392
Kerimi, 2018, Differential impact of flavonoids on redox modulation, bioenergetics, and cell signaling in normal and tumor cells: a comprehensive review, Antioxidants Redox Signal., 29, 1633, 10.1089/ars.2017.7086
Mostafavi-Pour, 2017, The role of quercetin and vitamin C in Nrf2-dependent oxidative stress production in breast cancer cells, Oncol. Lett., 13, 1965, 10.3892/ol.2017.5619
Hickson, 2019, Senolytics decrease senescent cells in humans: preliminary report from a clinical trial of Dasatinib plus Quercetin in individuals with diabetic kidney disease, EBioMedicine, 47, 446, 10.1016/j.ebiom.2019.08.069
Schafer, 2017, Cellular senescence mediates fibrotic pulmonary disease, Nat. Commun., 8, 14532, 10.1038/ncomms14532
Lehmann, 2017, Senolytic drugs target alveolar epithelial cell function and attenuate experimental lung fibrosis ex vivo, Eur. Respir. J., 50, 10.1183/13993003.02367-2016
Justice, 2019, Senolytics in idiopathic pulmonary fibrosis: results from a first-in-human, open-label, pilot study, EBioMedicine, 40, 554, 10.1016/j.ebiom.2018.12.052
Zhang, 2019, Senolytic therapy alleviates Abeta-associated oligodendrocyte progenitor cell senescence and cognitive deficits in an Alzheimer's disease model, Nat. Neurosci., 22, 719, 10.1038/s41593-019-0372-9
Zhu, 2017, New agents that target senescent cells: the flavone, fisetin, and the BCL-XL inhibitors, A1331852 and A1155463, Aging (Albany NY), 9, 955, 10.18632/aging.101202
Kovacovicova, 2018, Senolytic cocktail Dasatinib+Quercetin (D+Q) does not enhance the efficacy of senescence-inducing chemotherapy in liver cancer, Front. Oncol., 8, 459, 10.3389/fonc.2018.00459
Arai, 2000, Dietary intakes of flavonols, flavones and isoflavones by Japanese women and the inverse correlation between quercetin intake and plasma LDL cholesterol concentration, J. Nutr., 130, 2243, 10.1093/jn/130.9.2243
Kashyap, 2016, Molecular mechanisms of action of quercetin in cancer: recent advances, Tumour Biol., 37, 12927, 10.1007/s13277-016-5184-x
Lall, 2016, Dietary flavonoid fisetin for cancer prevention and treatment, Mol. Nutr. Food Res., 60, 1396, 10.1002/mnfr.201600025
Youns, 2017, The natural flavonoid fisetin inhibits cellular proliferation of hepatic, colorectal, and pancreatic cancer cells through modulation of multiple signaling pathways, PloS One, 12, 10.1371/journal.pone.0169335
Kang, 2014, Fisetin attenuates hydrogen peroxide-induced cell damage by scavenging reactive oxygen species and activating protective functions of cellular glutathione system, In Vitro Cell. Dev. Biol. Anim., 50, 66, 10.1007/s11626-013-9681-6
Yang, 2012, Dietary flavonoid fisetin targets caspase-3-deficient human breast cancer MCF-7 cells by induction of caspase-7-associated apoptosis and inhibition of autophagy, Int. J. Oncol., 40, 469
Li, 2014, The inhibitory effect of intravesical fisetin against bladder cancer by induction of p53 and down-regulation of NF-kappa B pathways in a rat bladder carcinogenesis model, Basic Clin. Pharmacol. Toxicol., 115, 321, 10.1111/bcpt.12229
Yousefzadeh, 2018, Fisetin is a senotherapeutic that extends health and lifespan, EBioMedicine, 36, 18, 10.1016/j.ebiom.2018.09.015
Bezerra, 2013, Overview of the therapeutic potential of piplartine (piperlongumine), Eur. J. Pharmaceut. Sci., 48, 453, 10.1016/j.ejps.2012.12.003
Piska, 2018, Piperlongumine (piplartine) as a lead compound for anticancer agents - synthesis and properties of analogues: a mini-review, Eur. J. Med. Chem., 156, 13, 10.1016/j.ejmech.2018.06.057
Piska, 2019, Synergistic anticancer activity of doxorubicin and piperlongumine on DU-145 prostate cancer cells - the involvement of carbonyl reductase 1 inhibition, Chem. Biol. Interact., 300, 40, 10.1016/j.cbi.2019.01.003
Wang, 2016, Discovery of piperlongumine as a potential novel lead for the development of senolytic agents, Aging (Albany NY), 8, 2915, 10.18632/aging.101100
Zhang, 2018, Oxidation resistance 1 is a novel senolytic target, Aging Cell, 17, 10.1111/acel.12780
Zheng, 2016, Piperlongumine inhibits lung tumor growth via inhibition of nuclear factor kappa B signaling pathway, Sci. Rep., 6, 26357, 10.1038/srep26357
Li, 2019, The curcumin analog EF24 is a novel senolytic agent, Aging (Albany NY), 11, 771, 10.18632/aging.101787
Guerrero, 2019, Cardiac glycosides are broad-spectrum senolytics, Nat. Metab., 1, 1074, 10.1038/s42255-019-0122-z
Triana-Martinez, 2019, Identification and characterization of Cardiac Glycosides as senolytic compounds, Nat. Commun., 10, 4731, 10.1038/s41467-019-12888-x
Zhou, 2019, Digoxin sensitizes gemcitabine-resistant pancreatic cancer cells to gemcitabine via inhibiting Nrf2 signaling pathway, Redox Biol., 22, 10.1016/j.redox.2019.101131
Chang, 2016, Clearance of senescent cells by ABT263 rejuvenates aged hematopoietic stem cells in mice, Nat. Med., 22, 78, 10.1038/nm.4010
Yosef, 2016, Directed elimination of senescent cells by inhibition of BCL-W and BCL-XL, Nat. Commun., 7, 11190, 10.1038/ncomms11190
Zhu, 2016, Identification of a novel senolytic agent, navitoclax, targeting the Bcl-2 family of anti-apoptotic factors, Aging Cell, 15, 428, 10.1111/acel.12445
Mavrogonatou, 2020, The role of senescence in cancer development, Semin. Canc. Biol., 62, 182, 10.1016/j.semcancer.2019.06.018
Grezella, 2018, Effects of senolytic drugs on human mesenchymal stromal cells, Stem Cell Res. Ther., 9, 108, 10.1186/s13287-018-0857-6
Wang, 2017, High-throughput functional genetic and compound screens identify targets for senescence induction in cancer, Cell Rep., 21, 773, 10.1016/j.celrep.2017.09.085
Shahbandi, 2020, BH3 mimetics selectively eliminate chemotherapy-induced senescent cells and improve response in TP53 wild-type breast cancer, Cell Death Differ., 27, 3097, 10.1038/s41418-020-0564-6
Pungsrinont, 2020, Senolytic compounds control a distinct fate of androgen receptor agonist- and antagonist-induced cellular senescent LNCaP prostate cancer cells, Cell Biosci., 10, 59, 10.1186/s13578-020-00422-2
Hantel, 2018, Safety and efficacy of the BCL inhibitors venetoclax and navitoclax in combination with chemotherapy in patients with relapsed/refractory acute lymphoblastic leukemia and lymphoblastic lymphoma, Clin. Lymphoma, Myeloma & Leukemia, 18, S184, 10.1016/j.clml.2018.07.016
Gonzalez-Gualda, 2020, Galacto-conjugation of Navitoclax as an efficient strategy to increase senolytic specificity and reduce platelet toxicity, Aging Cell, 19, 10.1111/acel.13142
He, 2020, Using proteolysis-targeting chimera technology to reduce navitoclax platelet toxicity and improve its senolytic activity, Nat. Commun., 11, 1996, 10.1038/s41467-020-15838-0
Leverson, 2015, Exploiting selective BCL-2 family inhibitors to dissect cell survival dependencies and define improved strategies for cancer therapy, Sci. Transl. Med., 7, 10.1126/scitranslmed.aaa4642
Jeon, 2017, Local clearance of senescent cells attenuates the development of post-traumatic osteoarthritis and creates a pro-regenerative environment, Nat. Med., 23, 775, 10.1038/nm.4324
Li, 2014, Rapamycin: one drug, many effects, Cell Metabol., 19, 373, 10.1016/j.cmet.2014.01.001
Herranz, 2015, mTOR regulates MAPKAPK2 translation to control the senescence-associated secretory phenotype, Nat. Cell Biol., 17, 1205, 10.1038/ncb3225
Correia-Melo, 2019, Rapamycin improves healthspan but not inflammaging in nfkappab1(-/-) mice, Aging Cell, 18, 10.1111/acel.12882
Imrali, 2016, Rapamycin inhibits prostate cancer cell growth through cyclin D1 and enhances the cytotoxic efficacy of cisplatin, Am. J. Canc. Res., 6, 1772
Walters, 2018, mTORC inhibitors as broad-spectrum therapeutics for age-related diseases, Int. J. Mol. Sci., 19, 10.3390/ijms19082325
Ngoi, 2020, Targeting cell metabolism as cancer therapy, Antioxidants Redox Signal., 32, 285, 10.1089/ars.2019.7947
Algire, 2012, Metformin reduces endogenous reactive oxygen species and associated DNA damage, Canc. Prev. Res. (Phila), 5, 536, 10.1158/1940-6207.CAPR-11-0536
Moiseeva, 2013, Metformin inhibits the senescence-associated secretory phenotype by interfering with IKK/NF-kappaB activation, Aging Cell, 12, 489, 10.1111/acel.12075
Sinnett-Smith, 2013, Metformin inhibition of mTORC1 activation, DNA synthesis and proliferation in pancreatic cancer cells: dependence on glucose concentration and role of AMPK, Biochem. Biophys. Res. Commun., 430, 352, 10.1016/j.bbrc.2012.11.010
Samaraweera, 2017, A novel indication for panobinostat as a senolytic drug in NSCLC and HNSCC, Sci. Rep., 7, 1900, 10.1038/s41598-017-01964-1
Filippakopoulos, 2010, Selective inhibition of BET bromodomains, Nature, 468, 1067, 10.1038/nature09504
Tasdemir, 2016, BRD4 connects enhancer remodeling to senescence immune surveillance, Canc. Discov., 6, 612, 10.1158/2159-8290.CD-16-0217
Wakita, 2020, A BET family protein degrader provokes senolysis by targeting NHEJ and autophagy in senescent cells, Nat. Commun., 11, 1935, 10.1038/s41467-020-15719-6
Ozsvari, 2018, Azithromycin and Roxithromycin define a new family of "senolytic" drugs that target senescent human fibroblasts, Aging (Albany NY), 10, 3294, 10.18632/aging.101633
Nogueira-Recalde, 2019, Fibrates as drugs with senolytic and autophagic activity for osteoarthritis therapy, EBioMedicine, 45, 588, 10.1016/j.ebiom.2019.06.049
Sapieha, 2018, Cellular senescence in postmitotic cells: beyond growth arrest, Trends Cell Biol., 28, 595, 10.1016/j.tcb.2018.03.003
von Zglinicki, 2021, Senescence in post-mitotic cells: a driver of aging?, Antioxidants Redox Signal., 34, 308, 10.1089/ars.2020.8048
Clement, 2020, Organismal aging and oxidants beyond macromolecules damage, Proteomics, 20, 10.1002/pmic.201800400