Recent Advances in Aging and Immunosenescence: Mechanisms and Therapeutic Strategies
Abstract
:1. Introduction
2. Molecular Mechanisms of Cellular Senescence and Immunosenescence
2.1. Molecular Mechanisms of Cellular Senescence
2.1.1. The cGAS-STING Pathway Promotes Age-Related Inflammation and Accelerates Aging
2.1.2. Endogenous Retroviruses Resurrected in the Human Genome Drive Aging
2.1.3. The CCF-TXNRD1-cGAS Axis Regulates Age-Related Inflammation
2.1.4. The TORC1-S6K-Syx13 Signaling Pathway Regulates Aging Through the Endolysosomal System
2.2. Molecular Mechanisms of Immunosenescence
2.2.1. Increased Expression of CISH in Activated T Cells in Elderly Individuals Causes Immunosenescence
2.2.2. IL-33 Induces Thymic Involution-Associated Naive T Cell Aging
2.2.3. Commensal Bacteria Induce the Aging of Germinal Center B Cells in the Gut
2.3. Regulation of Aging by the Immune System
2.3.1. CD4+ CTLs Eliminate SCs by Targeting Cytomegalovirus Antigens
2.3.2. Immunosenescence Drives Aging in Solid Organs
2.3.3. IgG Leads to Adipose Tissue Fibrosis
2.3.4. APCs Transfer Telomeres to T Cells to Protect T Cells from Aging
3. Strategies for Intervening in Aging
3.1. Intervening in Aging with Small-Molecule Senolytic Drugs
3.1.1. Quercetin
3.1.2. Fisetin
3.2. Intervention in Aging Through Immunological Means
3.2.1. Senolytic CAR T Cells Reverse Age-Related Pathologies
3.2.2. NKG2D-CAR T Cells Eliminate SCs in Aged Animals
3.2.3. Senolytic Vaccination to Remove SCs from the Body
3.2.4. Blocking PD-L1/PD-1 Improves Aging Phenotypes
3.3. Other Intervention Strategies
4. Summary and Prospects
Author Contributions
Funding
Data Availability Statement
Acknowledgments
Conflicts of Interest
References
- Partridge, L.; Deelen, J.; Slagboom, P.E. Facing up to the global challenges of ageing. Nature 2018, 561, 45–56. [Google Scholar] [CrossRef]
- López-Otín, C.; Blasco, M.A.; Partridge, L.; Serrano, M.; Kroemer, G. Hallmarks of aging: An expanding universe. Cell 2023, 186, 243–278. [Google Scholar] [CrossRef] [PubMed]
- Green, C.L.; Lamming, D.W.; Fontana, L. Molecular mechanisms of dietary restriction promoting health and longevity. Nat. Rev. Mol. Cell Biol. 2022, 23, 56–73. [Google Scholar] [CrossRef]
- Calder, P.C.; Ortega, E.F.; Meydani, S.N.; Adkins, Y.; Stephensen, C.B.; Thompson, B.; Zwickey, H. Nutrition, Immunosenescence, and Infectious Disease: An Overview of the Scientific Evidence on Micronutrients and on Modulation of the Gut Microbiota. Adv. Nutr. 2022, 13, S1–S26. [Google Scholar] [CrossRef] [PubMed]
- Guo, J.; Huang, X.; Dou, L.; Yan, M.; Shen, T.; Tang, W.; Li, J. Aging and aging-related diseases: From molecular mechanisms to interventions and treatments. Signal Transduct. Target. Ther. 2022, 7, 391. [Google Scholar] [CrossRef] [PubMed]
- Xu, M.; Pirtskhalava, T.; Farr, J.N.; Weigand, B.M.; Palmer, A.K.; Weivoda, M.M.; Inman, C.L.; Ogrodnik, M.B.; Hachfeld, C.M.; Fraser, D.G.; et al. Senolytics improve physical function and increase lifespan in old age. Nat. Med. 2018, 24, 1246–1256. [Google Scholar] [CrossRef]
- He, S.; Sharpless, N.E. Senescence in Health and Disease. Cell 2017, 169, 1000–1011. [Google Scholar] [CrossRef]
- Sharpless, N.E.; Sherr, C.J. Forging a signature of in vivo senescence. Nat. Rev. Cancer 2015, 15, 397–408. [Google Scholar] [CrossRef]
- Walford, R.L. The Immunologic Theory of Aging. Gerontologist 1964, 4, 195–197. [Google Scholar] [CrossRef]
- North, B.J.; Sinclair, D.A. The Intersection Between Aging and Cardiovascular Disease. Circ. Res. 2012, 110, 1097–1108. [Google Scholar] [CrossRef]
- Amorim, J.A.; Coppotelli, G.; Rolo, A.P.; Palmeira, C.M.; Ross, J.M.; Sinclair, D.A. Mitochondrial and metabolic dysfunction in ageing and age-related diseases. Nat. Rev. Endocrinol. 2022, 18, 243–258. [Google Scholar] [CrossRef] [PubMed]
- Graves, J.S.; Krysko, K.M.; Hua, L.H.; Absinta, M.; Franklin, R.J.M.; Segal, B.M. Ageing and multiple sclerosis. Lancet Neurol. 2023, 22, 66–77. [Google Scholar] [CrossRef] [PubMed]
- Culig, L.; Chu, X.; Bohr, V.A. Neurogenesis in aging and age-related neurodegenerative diseases. Ageing Res. Rev. 2022, 78, 101636. [Google Scholar] [CrossRef]
- Li, X.; Li, C.; Zhang, W.; Wang, Y.; Qian, P.; Huang, H. Inflammation and aging: Signaling pathways and intervention therapies. Signal Transduct. Target. Ther. 2023, 8, 239. [Google Scholar] [CrossRef] [PubMed]
- Baker, D.J.; Wijshake, T.; Tchkonia, T.; Lebrasseur, N.K.; Childs, B.G.; Van De Sluis, B.; Kirkland, J.L.; Van Deursen, J.M. Clearance of p16Ink4a-positive senescent cells delays ageing-associated disorders. Nature 2011, 479, 232–236. [Google Scholar] [CrossRef]
- Wang, B.; Wang, L.; Gasek, N.S.; Zhou, Y.; Kim, T.; Guo, C.; Jellison, E.R.; Haynes, L.; Yadav, S.; Tchkonia, T.; 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. [Google Scholar] [CrossRef]
- Kirkland, J.L.; Tchkonia, T. Cellular Senescence: A Translational Perspective. EBioMedicine 2017, 21, 21–28. [Google Scholar] [CrossRef]
- Zhu, Y.I.; Tchkonia, T.; Pirtskhalava, T.; Gower, A.C.; Ding, H.; Giorgadze, N.; Palmer, A.K.; Ikeno, Y.; Hubbard, G.B.; Lenburg, M.; et al. The Achilles’ heel of senescent cells: From transcriptome to senolytic drugs. Aging Cell 2015, 14, 644–658. [Google Scholar] [CrossRef]
- Amor, C.; Feucht, J.; Leibold, J.; Ho, Y.-J.; Zhu, C.; Alonso-Curbelo, D.; Mansilla-Soto, J.; Boyer, J.A.; Li, X.; Giavridis, T.; et al. Senolytic CAR T cells reverse senescence-associated pathologies. Nature 2020, 583, 127–132. [Google Scholar] [CrossRef]
- Suda, M.; Shimizu, I.; Katsuumi, G.; Yoshida, Y.; Hayashi, Y.; Ikegami, R.; Matsumoto, N.; Yoshida, Y.; Mikawa, R.; Katayama, A.; et al. Senolytic vaccination improves normal and pathological age-related phenotypes and increases lifespan in progeroid mice. Nat. Aging 2021, 1, 1117–1126. [Google Scholar] [CrossRef]
- Wang, T.-W.; Johmura, Y.; Suzuki, N.; Omori, S.; Migita, T.; Yamaguchi, K.; Hatakeyama, S.; Yamazaki, S.; Shimizu, E.; Imoto, S.; et al. Blocking PD-L1–PD-1 improves senescence surveillance and ageing phenotypes. Nature 2022, 611, 358–364. [Google Scholar] [CrossRef] [PubMed]
- Lawless, C.; Jurk, D.; Gillespie, C.S.; Shanley, D.; Saretzki, G.; von Zglinicki, T.; Passos, J.F. A Stochastic Step Model of Replicative Senescence Explains ROS Production Rate in Ageing Cell Populations. PLoS ONE 2012, 7, e32117. [Google Scholar] [CrossRef] [PubMed]
- Liu, X.; Liu, Z.; Wu, Z.; Ren, J.; Fan, Y.; Sun, L.; Cao, G.; Niu, Y.; Zhang, B.; Ji, Q.; et al. Resurrection of endogenous retroviruses during aging reinforces senescence. Cell 2023, 186, 287–304.e26. [Google Scholar] [CrossRef]
- Dong, C.-M.; Wang, X.-L.; Wang, G.-M.; Zhang, W.-J.; Zhu, L.; Gao, S.; Yang, D.-J.; Qin, Y.; Liang, Q.-J.; Chen, Y.-L.; et al. A stress-induced cellular aging model with postnatal neural stem cells. Cell Death Dis. 2014, 5, e1116. [Google Scholar] [CrossRef] [PubMed]
- Hasegawa, T.; Oka, T.; Son, H.G.; Oliver-García, V.S.; Azin, M.; Eisenhaure, T.M.; Lieb, D.J.; Hacohen, N.; Demehri, S. Cytotoxic CD4+ T cells eliminate senescent cells by targeting cytomegalovirus antigen. Cell 2023, 186, 1417–1431.e20. [Google Scholar] [CrossRef]
- Martic, I.; Wedel, S.; Jansen-Dürr, P.; Cavinato, M. A new model to investigate UVB-induced cellular senescence and pigmentation in melanocytes. Mech. Ageing Dev. 2020, 190, 111322. [Google Scholar] [CrossRef]
- Duan, J.; Duan, J.; Zhang, Z.; Tong, T. Irreversible cellular senescence induced by prolonged exposure to H2O2 involves DNA-damage-and-repair genes and telomere shortening. Int. J. Biochem. Cell Biol. 2005, 37, 1407–1420. [Google Scholar] [CrossRef]
- Yang, D.; Sun, B.; Li, S.; Wei, W.; Liu, X.; Cui, X.; Zhang, X.; Liu, N.; Yan, L.; Deng, Y.; et al. NKG2D-CAR T cells eliminate senescent cells in aged mice and nonhuman primates. Sci. Transl. Med. 2023, 15, eadd1951. [Google Scholar] [CrossRef]
- Gulen, M.F.; Samson, N.; Keller, A.; Schwabenland, M.; Liu, C.; Glück, S.; Thacker, V.V.; Favre, L.; Mangeat, B.; Kroese, L.J.; et al. cGAS–STING drives ageing-related inflammation and neurodegeneration. Nature 2023, 620, 374–380. [Google Scholar] [CrossRef]
- Kang, T.-W.; Yevsa, T.; Woller, N.; Hoenicke, L.; Wuestefeld, T.; Dauch, D.; Hohmeyer, A.; Gereke, M.; Rudalska, R.; Potapova, A.; et al. Senescence surveillance of pre-malignant hepatocytes limits liver cancer development. Nature 2011, 479, 547–551. [Google Scholar] [CrossRef]
- Wang, X.; Zheng, Z.; Caviglia, J.M.; Corey, K.E.; Herfel, T.M.; Cai, B.; Masia, R.; Chung, R.T.; Lefkowitch, J.H.; Schwabe, R.F.; et al. Hepatocyte TAZ/WWTR1 Promotes Inflammation and Fibrosis in Nonalcoholic Steatohepatitis. Cell Metab. 2016, 24, 848–862. [Google Scholar] [CrossRef] [PubMed]
- Yoshida, S.; Nakagami, H.; Hayashi, H.; Ikeda, Y.; Sun, J.; Tenma, A.; Tomioka, H.; Kawano, T.; Shimamura, M.; Morishita, R.; et al. The CD153 vaccine is a senotherapeutic option for preventing the accumulation of senescent T cells in mice. Nat. Commun. 2020, 11, 2482. [Google Scholar] [CrossRef]
- Yousefzadeh, M.J.; Flores, R.R.; Zhu, Y.; Schmiechen, Z.C.; Brooks, R.W.; Trussoni, C.E.; Cui, Y.; Angelini, L.; Lee, K.-A.; McGowan, S.J.; et al. An aged immune system drives senescence and ageing of solid organs. Nature 2021, 594, 100–105. [Google Scholar] [CrossRef] [PubMed]
- Zhang, Z.; Tian, X.; Lu, J.Y.; Boit, K.; Ablaeva, J.; Zakusilo, F.T.; Emmrich, S.; Firsanov, D.; Rydkina, E.; Biashad, S.A.; et al. Increased hyaluronan by naked mole-rat Has2 improves healthspan in mice. Nature 2023, 621, 196–205. [Google Scholar] [CrossRef] [PubMed]
- Demaria, M.; Ohtani, N.; Youssef, S.A.; Rodier, F.; Toussaint, W.; Mitchell, J.R.; Laberge, R.-M.; Vijg, J.; Van Steeg, H.; Dollé, M.E.; et al. An Essential Role for Senescent Cells in Optimal Wound Healing through Secretion of PDGF-AA. Dev. Cell 2014, 31, 722–733. [Google Scholar] [CrossRef]
- Alimirah, F.; Pulido, T.; Valdovinos, A.; Alptekin, S.; Chang, E.; Jones, E.; Diaz, D.A.; Flores, J.; Velarde, M.C.; Demaria, M.; et al. Cellular senescence promotes skin carcinogenesis through p38MAPK and p44/p42 MAPK signaling. Cancer Res. 2020, 80, 3606–3619. [Google Scholar] [CrossRef] [PubMed]
- Coppé, J.P.; Desprez, P.Y.; Krtolica, A.; Campisi, J. The Senescence-Associated Secretory Phenotype: The Dark Side of Tumor Suppression. Annu. Rev. Pathol. Mech. Dis. 2010, 5, 99–118. [Google Scholar] [CrossRef]
- Liu, Z.; Liang, Q.; Ren, Y.; Guo, C.; Ge, X.; Wang, L.; Cheng, Q.; Luo, P.; Zhang, Y.; Han, X. Immunosenescence: Molecular mechanisms and diseases. Signal Transduct. Target. Ther. 2023, 8, 200. [Google Scholar] [CrossRef]
- Krizhanovsky, V.; Yon, M.; Dickins, R.A.; Hearn, S.; Simon, J.; Miething, C.; Yee, H.; Zender, L.; Lowe, S.W. Senescence of Activated Stellate Cells Limits Liver Fibrosis. Cell 2008, 134, 657–667. [Google Scholar] [CrossRef]
- Caruso, C.; Accardi, G.; Virruso, C.; Candore, G. Sex, gender and immunosenescence: A key to understand the different lifespan between men and women? Immun. Ageing A 2013, 10, 20. [Google Scholar] [CrossRef]
- Tylutka, A.; Morawin, B.; Gramacki, A.; Zembron-Lacny, A. Lifestyle exercise attenuates immunosenescence; flow cytometry analysis. BMC Geriatr. 2021, 21, 200. [Google Scholar] [CrossRef]
- Savino, W. The Thymus Is a Common Target Organ in Infectious Diseases. PLoS Pathog. 2006, 2, e62. [Google Scholar] [CrossRef] [PubMed]
- Hopfner, K.P.; Hornung, V. Molecular mechanisms and cellular functions of cGAS–STING signalling. Nat. Rev. Mol. Cell Biol. 2020, 21, 501–521. [Google Scholar] [CrossRef] [PubMed]
- Yang, H.; Wang, H.; Ren, J.; Chen, Q.; Chen, Z.J. cGAS is essential for cellular senescence. Proc. Natl. Acad. Sci. USA 2017, 114, E4612–E4620. [Google Scholar] [CrossRef] [PubMed]
- Ablasser, A.; Chen, Z.J. cGAS in action: Expanding roles in immunity and inflammation. Science 2019, 363, eaat8657. [Google Scholar] [CrossRef]
- De Cecco, M.; Ito, T.; Petrashen, A.P.; Elias, A.E.; Skvir, N.J.; Criscione, S.W.; Caligiana, A.; Brocculi, G.; Adney, E.M.; Boeke, J.D.; et al. L1 drives IFN in senescent cells and promotes age-associated inflammation. Nature 2019, 566, 73–78. [Google Scholar] [CrossRef]
- Simon, M.; Van Meter, M.; Ablaeva, J.; Ke, Z.; Gonzalez, R.S.; Taguchi, T.; De Cecco, M.; Leonova, K.I.; Kogan, V.; Helfand, S.L.; et al. LINE1 derepression in aged wild type and SIRT6 deficient mice drives inflammation. Cell Metab. 2019, 29, 871–885.e5. [Google Scholar] [CrossRef]
- Jakobsson, J.; Vincendeau, M. SnapShot: Human endogenous retroviruses. Cell 2022, 185, 400–400.e1. [Google Scholar] [CrossRef]
- Dou, Z.; Ghosh, K.; Vizioli, M.G.; Zhu, J.; Sen, P.; Wangensteen, K.J.; Simithy, J.; Lan, Y.; Lin, Y.; Zhou, Z.; et al. Cytoplasmic chromatin triggers inflammation in senescence and cancer. Nature 2017, 550, 402–406. [Google Scholar] [CrossRef]
- Glück, S.; Guey, B.; Gulen, M.F.; Wolter, K.; Kang, T.-W.; Schmacke, N.A.; Bridgeman, A.; Rehwinkel, J.; Zender, L.; Ablasser, A. Innate immune sensing of cytosolic chromatin fragments through cGAS promotes senescence. Nat. Cell Biol. 2017, 19, 1061–1070. [Google Scholar] [CrossRef]
- Arnér, E.S.J. Chapter Five—Targeting the Selenoprotein Thioredoxin Reductase 1 for Anticancer Therapy [Internet]. In Advances in Cancer Research; Tew, K.D., Galli, F., Eds.; Academic Press: Cambridge, MA, USA, 2017; pp. 139–151. [Google Scholar] [CrossRef]
- Finkel, T.; Holbrook, N.J. Oxidants, oxidative stress and the biology of ageing. Nature 2000, 408, 239–247. [Google Scholar] [CrossRef] [PubMed]
- Hao, X.; Zhao, B.; Towers, M.; Liao, L.; Monteiro, E.L.; Xu, X.; Freeman, C.; Peng, H.; Tang, H.-Y.; Havas, A.; et al. TXNRD1 drives the innate immune response in senescent cells with implications for age-associated inflammation. Nat. Aging 2024, 4, 185–197. [Google Scholar] [CrossRef] [PubMed]
- Bjedov, I.; Toivonen, J.M.; Kerr, F.; Slack, C.; Jacobson, J.; Foley, A.; Partridge, L. Mechanisms of life span extension by rapamycin in the fruit fly Drosophila melanogaster. Cell Metab. 2010, 11, 35–46. [Google Scholar] [CrossRef] [PubMed]
- Selman, C.; Tullet, J.M.A.; Wieser, D.; Irvine, E.; Lingard, S.J.; Choudhury, A.I.; Claret, M.; Al-Qassab, H.; Carmignac, D.; Ramadani, F.; et al. Ribosomal Protein S6 Kinase 1 Signaling Regulates Mammalian Life Span. Science 2009, 326, 140–144. [Google Scholar] [CrossRef]
- Zhang, P.; Catterson, J.H.; Grönke, S.; Partridge, L. Inhibition of S6K lowers age-related inflammation and increases lifespan through the endolysosomal system. Nat. Aging 2024, 4, 491–509. [Google Scholar] [CrossRef]
- Mittelbrunn, M.; Kroemer, G. Hallmarks of T cell aging. Nat. Immunol. 2021, 22, 687–698. [Google Scholar] [CrossRef]
- Franceschi, C.; Garagnani, P.; Parini, P.; Giuliani, C.; Santoro, A. Inflammaging: A new immune–metabolic viewpoint for age-related diseases. Nat. Rev. Endocrinol. 2018, 14, 576–590. [Google Scholar] [CrossRef]
- Ferrucci, L.; Fabbri, E. Inflammageing: Chronic inflammation in ageing, cardiovascular disease, and frailty. Nat. Rev. Cardiol. 2018, 15, 505–522. [Google Scholar] [CrossRef]
- Jin, J.; Mu, Y.; Zhang, H.; Sturmlechner, I.; Wang, C.; Jadhav, R.R.; Xia, Q.; Weyand, C.M.; Goronzy, J.J. CISH impairs lysosomal function in activated T cells resulting in mitochondrial DNA release and inflammaging. Nat. Aging 2023, 3, 600–616. [Google Scholar] [CrossRef]
- Kadouri, N.; Nevo, S.; Goldfarb, Y.; Abramson, J. Thymic epithelial cell heterogeneity: TEC by TEC. Nat. Rev. Immunol. 2020, 20, 239–253. [Google Scholar] [CrossRef]
- Boehm, T.; Swann, J.B. Thymus involution and regeneration: Two sides of the same coin? Nat. Rev. Immunol. 2013, 13, 831–838. [Google Scholar] [CrossRef] [PubMed]
- Stelekati, E.; Wherry, E.J. Chronic Bystander Infections and Immunity to Unrelated Antigens. Cell Host Microbe 2012, 12, 458–469. [Google Scholar] [CrossRef] [PubMed]
- Xu, L.; Wei, C.; Chen, Y.; Wu, Y.; Shou, X.; Chen, W.; Lu, D.; Sun, H.; Li, W.; Yu, B.; et al. IL-33 induces thymic involution-associated naive T cell aging and impairs host control of severe infection. Nat. Commun. 2022, 13, 6881. [Google Scholar] [CrossRef]
- O’Toole, P.W.; Jeffery, I.B. Gut microbiota and aging. Science 2015, 350, 1214–1215. [Google Scholar] [CrossRef]
- Fransen, F.; Van Beek, A.A.; Borghuis, T.; El Aidy, S.; Hugenholtz, F.; van der Gaast-de Jongh, C.; Savelkoul, H.F.J.; De Jonge, M.I.; Boekschoten, M.V.; Smidt, H.; et al. Aged Gut Microbiota Contributes to Systemical Inflammaging after Transfer to Germ-Free Mice. Front. Immunol. 2017, 8, 1385. [Google Scholar] [CrossRef]
- Boehme, M.; Guzzetta, K.E.; Bastiaanssen, T.F.S.; van de Wouw, M.; Moloney, G.M.; Gual-Grau, A.; Spichak, S.; Olavarría-Ramírez, L.; Fitzgerald, P.; Morillas, E.; et al. Microbiota from young mice counteracts selective age-associated behavioral deficits. Nat. Aging 2021, 1, 666–676. [Google Scholar] [CrossRef] [PubMed]
- Kawamoto, S.; Uemura, K.; Hori, N.; Takayasu, L.; Konishi, Y.; Katoh, K.; Matsumoto, T.; Suzuki, M.; Sakai, Y.; Matsudaira, T.; et al. Bacterial induction of B cell senescence promotes age-related changes in the gut microbiota. Nat. Cell Biol. 2023, 25, 865–876. [Google Scholar] [CrossRef]
- Paez-Ribes, M.; González-Gualda, E.; Doherty, G.J.; Muñoz-Espín, D. Targeting senescent cells in translational medicine. EMBO Mol. Med. 2019, 11, e10234. [Google Scholar] [CrossRef]
- Robinson, A.R.; Yousefzadeh, M.J.; Rozgaja, T.A.; Wang, J.; Li, X.; Tilstra, J.S.; Feldman, C.H.; Gregg, S.Q.; Johnson, C.H.; Skoda, E.M.; et al. Spontaneous DNA damage to the nuclear genome promotes senescence, redox imbalance and aging. Redox Biol. 2018, 17, 259–273. [Google Scholar] [CrossRef]
- Zhou, Q.; Wan, Q.; Jiang, Y.; Liu, J.; Qiang, L.; Sun, L. A Landscape of Murine Long Non-Coding RNAs Reveals the Leading Transcriptome Alterations in Adipose Tissue during Aging. Cell Rep. 2020, 31, 107694. [Google Scholar] [CrossRef]
- Palmer, A.K.; Kirkland, J.L. Aging and Adipose Tissue: Potential Interventions for Diabetes and Regenerative Medicine. Exp. Gerontol. 2016, 86, 97–105. [Google Scholar] [CrossRef] [PubMed]
- Roopenian, D.C.; Akilesh, S. FcRn: The neonatal Fc receptor comes of age. Nat. Rev. Immunol. 2007, 7, 715–725. [Google Scholar] [CrossRef]
- Yu, L.; Wan, Q.; Liu, Q.; Fan, Y.; Zhou, Q.; Skowronski, A.A.; Wang, S.; Shao, Z.; Liao, C.-Y.; Ding, L.; et al. IgG is an aging factor that drives adipose tissue fibrosis and metabolic decline. Cell Metab. 2024, 36, 793–807.e5. [Google Scholar] [CrossRef] [PubMed]
- Rossiello, F.; Jurk, D.; Passos, J.F.; d’Adda di Fagagna, F. Telomere dysfunction in ageing and age-related diseases. Nat. Cell Biol. 2022, 24, 135–147. [Google Scholar] [CrossRef] [PubMed]
- Roake, C.M.; Artandi, S.E. Regulation of human telomerase in homeostasis and disease. Nat. Rev. Mol. Cell Biol. 2020, 21, 384–397. [Google Scholar] [CrossRef]
- Lanna, A.; Gomes, D.C.O.; Muller-Durovic, B.; McDonnell, T.; Escors, D.; Gilroy, D.W.; Lee, J.H.; Karin, M.; Akbar, A.N. A sestrin-dependent Erk–Jnk–p38 MAPK activation complex inhibits immunity during aging. Nat. Immunol. 2017, 18, 354–363. [Google Scholar] [CrossRef]
- Lanna, A.; Vaz, B.; D’ambra, C.; Valvo, S.; Vuotto, C.; Chiurchiù, V.; Devine, O.; Sanchez, M.; Borsellino, G.; Akbar, A.N.; et al. An intercellular transfer of telomeres rescues T cells from senescence and promotes long-term immunological memory. Nat. Cell Biol. 2022, 24, 1461–1474. [Google Scholar] [CrossRef]
- Ge, M.; Hu, L.; Ao, H.; Zi, M.; Kong, Q.; He, Y. Senolytic targets and new strategies for clearing senescent cells. Mech. Ageing Dev. 2021, 195, 111468. [Google Scholar] [CrossRef]
- Anand David, A.V.; Arulmoli, R.; Parasuraman, S. Overviews of Biological Importance of Quercetin: A Bioactive Flavonoid. Pharmacogn. Rev. 2016, 10, 84–89. [Google Scholar] [CrossRef]
- Hickson, L.J.; Langhi Prata, L.G.P.; Bobart, S.A.; Evans, T.K.; Giorgadze, N.; Hashmi, S.K.; Herrmann, S.M.; Jensen, M.D.; Jia, Q.; Jordan, K.L.; 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. [Google Scholar] [CrossRef]
- Wagner, J.U.G.; Tombor, L.S.; Malacarne, P.F.; Kettenhausen, L.-M.; Panthel, J.; Kujundzic, H.; Manickam, N.; Schmitz, K.; Cipca, M.; Stilz, K.A.; et al. Aging impairs the neurovascular interface in the heart. Science 2023, 381, 897–906. [Google Scholar] [CrossRef] [PubMed]
- Kashyap, D.; Sharma, A.; Sak, K.; Tuli, H.S.; Buttar, H.S.; Bishayee, A. Fisetin: A bioactive phytochemical with potential for cancer prevention and pharmacotherapy. Life Sci. 2018, 194, 75–87. [Google Scholar] [CrossRef] [PubMed]
- Lall, R.K.; Adhami, V.M.; Mukhtar, H. Dietary flavonoid fisetin for cancer prevention and treatment. Mol. Nutr. Food Res. 2016, 60, 1396–1405. [Google Scholar] [CrossRef] [PubMed]
- Zhu, Y.; Doornebal, E.J.; Pirtskhalava, T.; Giorgadze, N.; Wentworth, M.; Fuhrmann-Stroissnigg, H.; Niedernhofer, L.J.; Robbins, P.D.; Tchkonia, T.; Kirkland, J.L. New agents that target senescent cells: The flavone, fisetin, and the BCL-XL inhibitors, A1331852 and A1155463. Aging 2017, 9, 955–963. [Google Scholar] [CrossRef]
- Yousefzadeh, M.J.; Zhu, Y.; McGowan, S.J.; Angelini, L.; Fuhrmann-Stroissnigg, H.; Xu, M.; Ling, Y.Y.; Melos, K.I.; Pirtskhalava, T.; Inman, C.L.; et al. Fisetin is a senotherapeutic that extends health and lifespan. EBioMedicine 2018, 36, 18–28. [Google Scholar] [CrossRef]
- Chang, J.; Wang, Y.; Shao, L.; Laberge, R.-M.; DeMaria, M.; Campisi, J.; Janakiraman, K.; Sharpless, N.E.; Ding, S.; Feng, W.; et al. Clearance of senescent cells by ABT263 rejuvenates aged hematopoietic stem cells in mice. Nat. Med. 2016, 22, 78–83. [Google Scholar] [CrossRef]
- Childs, B.G.; Gluscevic, M.; Baker, D.J.; Laberge, R.-M.; Marquess, D.; Dananberg, J.; van Deursen, J.M. Senescent cells: An emerging target for diseases of ageing. Nat. Rev. Drug Discov. 2017, 16, 718–735. [Google Scholar] [CrossRef]
- Lu, J.; Jiang, G. The journey of CAR-T therapy in hematological malignancies. Mol. Cancer 2022, 21, 194. [Google Scholar] [CrossRef]
- Müller, F.; Taubmann, J.; Bucci, L.; Wilhelm, A.; Bergmann, C.; Völkl, S.; Aigner, M.; Rothe, T.; Minopoulou, I.; Tur, C.; et al. CD19 CAR T-Cell Therapy in Autoimmune Disease—A Case Series with Follow-up. N. Engl. J. Med. 2024, 390, 687–700. [Google Scholar] [CrossRef]
- Pecher, A.-C.; Hensen, L.; Klein, R.; Schairer, R.; Lutz, K.; Atar, D.; Seitz, C.; Stanger, A.; Schneider, J.; Braun, C.; et al. CD19-Targeting CAR T Cells for Myositis and Interstitial Lung Disease Associated with Antisynthetase Syndrome. JAMA 2023, 329, 2154–2162. [Google Scholar] [CrossRef]
- Amor, C.; Fernández-Maestre, I.; Chowdhury, S.; Ho, Y.-J.; Nadella, S.; Graham, C.; Carrasco, S.E.; Nnuji-John, E.; Feucht, J.; Hinterleitner, C.; et al. Prophylactic and long-lasting efficacy of senolytic CAR T cells against age-related metabolic dysfunction. Res. Sq. 2023; preprint. [Google Scholar] [CrossRef] [PubMed]
- Carapito, R.; Bahram, S. Genetics, genomics, and evolutionary biology of NKG2D ligands. Immunol. Rev. 2015, 267, 88–116. [Google Scholar] [CrossRef] [PubMed]
- Baumeister, S.H.; Murad, J.; Werner, L.; Daley, H.; Trebeden-Negre, H.; Gicobi, J.K.; Schmucker, A.; Reder, J.; Sentman, C.L.; Gilham, D.E.; et al. Phase 1 Trial of Autologous CAR T Cells Targeting NKG2D Ligands in Patients with AML/MDS and Multiple Myeloma. Cancer Immunol. Res. 2019, 7, 100–112. [Google Scholar] [CrossRef] [PubMed]
- Sallman, D.A.; Kerre, T.; Havelange, V.; Poiré, X.; Lewalle, P.; Wang, E.S.; Brayer, J.B.; Davila, M.L.; Moors, I.; Machiels, J.-P.; et al. CYAD-01, an autologous NKG2D-based CAR T-cell therapy, in relapsed or refractory acute myeloid leukaemia and myelodysplastic syndromes or multiple myeloma (THINK): Haematological cohorts of the dose escalation segment of a phase 1 trial. Lancet Haematol. 2023, 10, e191–e202. [Google Scholar] [CrossRef]
- Sagiv, A.; Burton, D.G.A.; Moshayev, Z.; Vadai, E.; Wensveen, F.; Ben-Dor, S.; Golani, O.; Polic, B.; Krizhanovsky, V. NKG2D ligands mediate immunosurveillance of senescent cells. Aging 2016, 8, 328–344. [Google Scholar] [CrossRef]
- Vaccines work. Nat. Commun. 2018, 9, 1666. [CrossRef]
- Morse, M.A.; Gwin, W.R.; Mitchell, D.A. Vaccine Therapies for Cancer: Then and Now. Target. Oncol. 2021, 16, 121–152. [Google Scholar] [CrossRef]
- Shirakawa, K.; Yan, X.; Shinmura, K.; Endo, J.; Kataoka, M.; Katsumata, Y.; Yamamoto, T.; Anzai, A.; Isobe, S.; Yoshida, N.; et al. Obesity accelerates T cell senescence in murine visceral adipose tissue. J. Clin. Investig. 2016, 126, 4626–4639. [Google Scholar] [CrossRef]
- Childs, B.G.; Durik, M.; Baker, D.J.; van Deursen, J.M. Cellular senescence in aging and age-related disease: From mechanisms to therapy. Nat. Med. 2015, 21, 1424–1435. [Google Scholar] [CrossRef]
- Tuttle, C.S.L.; Waaijer, M.E.C.; Slee-Valentijn, M.S.; Stijnen, T.; Westendorp, R.; Maier, A.B. Cellular senescence and chronological age in various human tissues: A systematic review and meta-analysis. Aging Cell 2020, 19, e13083. [Google Scholar] [CrossRef]
- Ishida, Y.; Agata, Y.; Shibahara, K.; Honjo, T. Induced expression of PD-1, a novel member of the immunoglobulin gene superfamily, upon programmed cell death. EMBO J. 1992, 11, 3887–3895. [Google Scholar] [CrossRef] [PubMed]
- Chamoto, K.; Yaguchi, T.; Tajima, M.; Honjo, T. Insights from a 30-year journey: Function, regulation and therapeutic modulation of PD1. Nat. Rev. Immunol. 2023, 23, 682–695. [Google Scholar] [CrossRef] [PubMed]
- Campisi, J.; Kapahi, P.; Lithgow, G.J.; Melov, S.; Newman, J.C.; Verdin, E. From discoveries in ageing research to therapeutics for healthy ageing. Nature 2019, 571, 183–192. [Google Scholar] [CrossRef] [PubMed]
- Calubag, M.F.; Robbins, P.D.; Lamming, D.W. A nutrigeroscience approach: Dietary macronutrients and cellular senescence. Cell Metab. 2024, 36, 1914–1944. [Google Scholar] [CrossRef] [PubMed]
- Speakman, J.R.; Mitchell, S.E. Caloric restriction. Mol. Asp. Med. 2011, 32, 159–221. [Google Scholar] [CrossRef]
- Fontana, L.; Partridge, L.; Longo, V.D. Extending healthy life span—From yeast to humans. Science 2010, 328, 321–326. [Google Scholar] [CrossRef] [PubMed]
- Most, J.; Tosti, V.; Redman, L.M.; Fontana, L. Calorie restriction in humans: An update. Ageing Res. Rev. 2017, 39, 36–45. [Google Scholar] [CrossRef]
- Qu, Q.; Chen, Y.; Wang, Y.; Long, S.; Wang, W.; Yang, H.-Y.; Li, M.; Tian, X.; Wei, X.; Liu, Y.-H.; et al. Lithocholic acid phenocopies anti-ageing effects of calorie restriction. Nature 2024. Epub ahead of print. [Google Scholar] [CrossRef]
- Qu, Q.; Chen, Y.; Wang, Y.; Wang, W.; Long, S.; Yang, H.-Y.; Wu, J.; Li, M.; Tian, X.; Wei, X.; et al. Lithocholic acid binds TULP3 to activate sirtuins and AMPK to slow down ageing. Nature 2024. Epub ahead of print. [Google Scholar] [CrossRef]
- Di Francesco, A.; Di Germanio, C.; Bernier, M.; de Cabo, R. A time to fast. Science 2018, 362, 770–775. [Google Scholar] [CrossRef]
- Hofer, S.J.; Daskalaki, I.; Bergmann, M.; Friščić, J.; Zimmermann, A.; Mueller, M.I.; Abdellatif, M.; Nicastro, R.; Masser, S.; Durand, S.; et al. Spermidine is essential for fasting-mediated autophagy and longevity. Nat. Cell Biol. 2024, 26, 1571–1584. [Google Scholar] [CrossRef]
- Shannon, O.M.; Ashor, A.W.; Scialo, F.; Saretzki, G.; Martin-Ruiz, C.; Lara, J.; Matu, J.; Griffiths, A.; Robinson, N.; Lillà, L.; et al. Mediterranean diet and the hallmarks of ageing. Eur. J. Clin. Nutr. 2021, 75, 1176–1192. [Google Scholar] [CrossRef]
- Tosti, V.; Bertozzi, B.; Fontana, L. Health benefits of the Mediterranean diet: Metabolic and molecular mechanisms. J. Gerontol. A Biol. Sci. Med. Sci. 2018, 73, 318–326. [Google Scholar] [CrossRef] [PubMed]
- Willcox, D.C.; Willcox, B.J.; Todoriki, H.; Suzuki, M. The okinawan diet: Health implications of a low-calorie, nutrient-dense, antioxidant-rich dietary pattern low in glycemic load. J. Am. Coll. Nutr. 2009, 28, 500S–516S. [Google Scholar] [CrossRef] [PubMed]
- Willcox, D.C.; Scapagnini, G.; Willcox, B.J. Healthy aging diets other than the Mediterranean: A focus on the okinawan diet. Mech. Ageing Dev. 2014, 136–137, 148–162. [Google Scholar] [CrossRef] [PubMed]
- Guarente, L.; Sinclair, D.A.; Kroemer, G. Human trials exploring anti-aging medicines. Cell Metab. 2024, 36, 354–376. [Google Scholar] [CrossRef]
- Pernicova, I.; Korbonits, M. Metformin—Mode of action and clinical implications for diabetes and cancer. Nat. Rev. Endocrinol. 2014, 10, 143–156. [Google Scholar] [CrossRef]
- Ma, T.; Tian, X.; Zhang, B.; Li, M.; Wang, Y.; Yang, C.; Wu, J.; Wei, X.; Qu, Q.; Yu, Y.; et al. Low-dose metformin targets the lysosomal AMPK pathway through PEN2. Nature 2022, 603, 159–165. [Google Scholar] [CrossRef]
- Feng, J.; Lu, H.; Ma, W.; Tian, W.; Lu, Z.; Yang, H.; Cai, Y.; Cai, P.; Sun, Y.; Zhou, Z.; et al. Genome-wide CRISPR screen identifies synthetic lethality between DOCK1 inhibition and metformin in liver cancer. Protein Cell 2022, 13, 825–841. [Google Scholar] [CrossRef]
- Yang, Y.; Lu, X.; Liu, N.; Ma, S.; Zhang, H.; Zhang, Z.; Yang, K.; Jiang, M.; Zheng, Z.; Qiao, Y.; et al. Metformin decelerates aging clock in Male monkeys. Cell 2024, 187, 6358–6378.e29. [Google Scholar] [CrossRef]
- Xu, X.; Sun, Y.; Cen, X.; Shan, B.; Zhao, Q.; Xie, T.; Wang, Z.; Hou, T.; Xue, Y.; Zhang, M.; et al. Metformin activates chaperone-mediated autophagy and improves disease pathologies in an alzheimer disease mouse model. Protein Cell 2021, 12, 769–787. [Google Scholar] [CrossRef]
- Tai, S.; Sun, J.; Zhou, Y.; Zhu, Z.; He, Y.; Chen, M.; Yang, H.; Xiao, Y.; Tu, T.; Tang, L.; et al. Metformin suppresses vascular smooth muscle cell senescence by promoting autophagic flux. J. Adv. Res. 2021, 41, 205–218. [Google Scholar] [CrossRef]
- McGaunn, J.; Baur, J.A. Taurine linked with healthy aging. Science 2023, 380, 1010–1011. [Google Scholar] [CrossRef]
- Kurtz, J.A.; VanDusseldorp, T.A.; Doyle, J.A.; Otis, J.S. Taurine in sports and exercise. J. Int. Soc. Sports Nutr. 2021, 18, 39. [Google Scholar] [CrossRef] [PubMed]
- Caine, J.J.; Geracioti, T.D. Taurine, energy drinks, and neuroendocrine effects. Clevel. Clin. J. Med. 2016, 83, 895–904. [Google Scholar] [CrossRef] [PubMed]
- Wen, C.; Li, F.; Zhang, L.; Duan, Y.; Guo, Q.; Wang, W.; He, S.; Li, J.; Yin, Y. Taurine is Involved in Energy Metabolism in Muscles, Adipose Tissue, and the Liver. Mol. Nutr. Food Res. 2019, 63, 1800536. [Google Scholar] [CrossRef]
- Singh, P.; Gollapalli, K.; Mangiola, S.; Schranner, D.; Yusuf, M.A.; Chamoli, M.; Shi, S.L.; Bastos, B.L.; Nair, T.; Riermeier, A.; et al. Taurine deficiency as a driver of aging. Science 2023, 380, eabn9257. [Google Scholar] [CrossRef] [PubMed]
- Cao, T.; Zhang, W.; Wang, Q.; Wang, C.; Ma, W.; Zhang, C.; Ge, M.; Tian, M.; Yu, J.; Jiao, A.; et al. Cancer SLC6A6-mediated taurine uptake transactivates immune checkpoint genes and induces exhaustion in CD8+ T cells. Cell 2024, 187, 2288–2304.e27. [Google Scholar] [CrossRef]
- Sun, S.; Li, J.; Wang, S.; Li, J.; Ren, J.; Bao, Z.; Sun, L.; Ma, X.; Zheng, F.; Ma, S.; et al. CHIT1-positive microglia drive motor neuron ageing in the primate spinal cord. Nature 2023, 624, 611–620. [Google Scholar] [CrossRef]
- Scudellari, M. Ageing research: Blood to blood. Nature 2015, 517, 426–429. [Google Scholar] [CrossRef]
- Schroer, A.B.; Ventura, P.B.; Sucharov, J.; Misra, R.; Chui, M.K.K.; Bieri, G.; Horowitz, A.M.; Smith, L.K.; Encabo, K.; Tenggara, I.; et al. Platelet factors attenuate inflammation and rescue cognition in ageing. Nature 2023, 620, 1071–1079. [Google Scholar] [CrossRef]
- Temple, S. Advancing cell therapy for neurodegenerative diseases. Cell Stem Cell 2023, 30, 512–529. [Google Scholar] [CrossRef] [PubMed]
- Poch, C.M.; Foo, K.S.; De Angelis, M.T.; Jennbacken, K.; Santamaria, G.; Bähr, A.; Wang, Q.-D.; Reiter, F.; Hornaschewitz, N.; Zawada, D.; et al. Migratory and anti-fibrotic programmes define the regenerative potential of human cardiac progenitors. Nat. Cell Biol. 2022, 24, 659–671. [Google Scholar] [CrossRef] [PubMed]
- Shareghi-oskoue, O.; Aghebati-Maleki, L.; Yousefi, M. Transplantation of human umbilical cord mesenchymal stem cells to treat premature ovarian failure. Stem Cell Res. Ther. 2021, 12, 454. [Google Scholar] [CrossRef] [PubMed]
- Genchi, A.; Brambilla, E.; Sangalli, F.; Radaelli, M.; Bacigaluppi, M.; Furlan, R.; Andolfo, A.; Drago, D.; Magagnotti, C.; Scotti, G.M.; et al. Neural stem cell transplantation in patients with progressive multiple sclerosis: An open-label, phase 1 study. Nat. Med. 2023, 29, 75–85. [Google Scholar] [CrossRef]
- Feucht, J.; Abou-El-Enein, M. Senolytic CAR T Cells in Solid Tumors and Age-Related Pathologies. Mol. Ther. 2020, 28, 2108–2110. [Google Scholar] [CrossRef]
- Li, W.; Qin, L.; Feng, R.; Hu, G.; Sun, H.; He, Y.; Zhang, R. Emerging senolytic agents derived from natural products. Mech. Ageing Dev. 2019, 181, 1–6. [Google Scholar] [CrossRef]
- Baker, D.J.; Arany, Z.; Baur, J.A.; Epstein, J.A.; June, C.H. CAR T therapy beyond cancer: The evolution of a living drug. Nature 2023, 619, 707–715. [Google Scholar] [CrossRef]
- Gasek, N.S.; Kuchel, G.A.; Kirkland, J.L.; Xu, M. Strategies for targeting senescent cells in human disease. Nat. Aging 2021, 1, 870–879. [Google Scholar] [CrossRef]
- Wright, W.E.; Shay, J.W. Telomere dynamics in cancer progression and prevention: Fundamental differences in human and mouse telomere biology. Nat. Med. 2000, 6, 849–851. [Google Scholar] [CrossRef]
- Itahana, K.; Campisi, J.; Dimri, G.P. Mechanisms of cellular senescence in human and mouse cells. Biogerontology 2004, 5, 1–10. [Google Scholar] [CrossRef] [PubMed]
- Jain, M.D.; Smith, M.; Shah, N.N. How I treat refractory CRS and ICANS after CAR T-cell therapy. Blood 2023, 141, 2430–2442. [Google Scholar] [CrossRef] [PubMed]
Model Name | Methods | Description |
---|---|---|
Cell Models | ||
Replicative senescence model | Repeat passages until they reach replicative senescence [22]. Human fibroblasts are replicatively senescent in their late passage (LP, p > 23) [23]. | Cell cycle arrest, shortened telomeres, p16-positive, p21-positive, SA-β-Gal positive, and ROS increase. Negative for proliferation markers (BrdU and Ki67) [22]. |
Chemotherapy-induced senescence model | Postnatal NSCs were treated with 8 mM hydroxyurea for 12 h [24]. HCA2 cell senescence induction with RO3306 and nutlin3a [21]. | PD-L1 positive, [21] SA-β-Gal positive, increased ROS, and increased expression of P16, p21, and p53. |
Stress-induced senescence model | Fibroblasts were irradiated with UVA (5 or 10 J) and incubated for one or five days [25]. UVB treatment of melanocytes was performed at a dose of 0.125 J/cm2 [26]. Treatment of HDF with low doses of H2O2 leads to irreversible cellular senescence [27]. | P16-positive, p21-positive, SenTraGor-positive, and SA-β-Gal-positive. Up-regulates the expression of ULBP2 and HLA-II [25]. |
Oncogene-induced senescence model | Human lung fibroblasts were infected with a lentivirus produced from pTomo-KrasG12D-EGFP/pTomo-Teton-P16INK4a-T2A-EGFP [28]. | SA-β-Gal-positive; upregulation of p16 and NKG2DLs expression. |
Animal Models | ||
Natural aging mice | Wild-type C57BL/6J mice cultured to 26 months [29]. | Inflammatory cell accumulation, elevated levels of inflammation in the kidneys and liver, increased microglial cell infiltration, and decreased memory and learning ability. |
Oncogene-induced hepatocyte senescence in mice in vivo [19,30] | Stable delivery of transposable factors expressing oncogenic NrasG12V into mice hepatocytes [30]. | Up-regulation of p21, p16, and uPAR expression and SA-β-Gal-positive [19]. |
Liver fibrosis mice [19,21,31] | C57BL/6N mice were treated CCl4 to induce liver fibrosis [19]. Feeding mice NASH-inducing diets [31]. | Liver fibrosis, liver inflammation, and upregulation of uPAR expression. |
HFD-induced obese mice [20,32] | C57BL/6J mice were fed a HFD (D12492, 60 kcal% fat; Research Diets inc.) [32]. | Senescent T cell accumulation, insulin resistance, increased inflammatory response, and metabolic syndrome. |
Premature immunosenescence mice [33] | Ercc1, which encodes a crucial DNA repair protein, was selectively deleted from mouse haematopoietic cells [33]. | Immune cells are susceptible to endogenous DNA damage, premature onset of immune cell senescence, significant reduction in the proportion of T cells, and impaired immune function. |
Transgenic delayed aging mice [34] | Overexpression of the naked mole rat hyaluronic acid synthase 2 gene (nmrHas2) in mice [34]. | Increased levels of hyaluronic acid, reduced inflammation and oxidative stress, decreased cancer incidence, and increased life expectancy. |
NCT Number | Conditions | Interventions | Clinical Progress | Starting Time | Locations |
---|---|---|---|---|---|
NCT04210986 | Osteoarthritis | Fisetin | Phase 1/2 | 2020 | United States |
NCT05025956 | Femoroacetabular Impingement | Fisetin | Phase 1/2 | 2021 | United States |
NCT04685590 | Alzheimer Disease, Mild Cognitive Impairment | Dasatinib + Quercetin | Phase 2 | 2021 | United States and Spain |
NCT04063124 | Alzheimer Disease | Dasatinib + Quercetin | Phase 1/2 | 2020 | United States |
NCT04815902 | Osteoarthritis | Fisetin, Losartan | Phase 1/2 | 2021 | United States |
NCT05276895 | Osteoarthritis | Quercetin and Fisetin | Not applicable | 2022 | Egypt |
NCT06133634 | Aging, Endothelial Dysfunction, and Arterial Stiffness | Fisetin | Phase 1/2 | 2023 | United States |
NCT06240403 | Heart Failure, Systolic or Diabetes Mellitus, Type-2 | Digoxin | Phase 2 | 2024 | United Kingdom |
NCT02848131 | Chronic Kidney Disease | Dasatinib + Quercetin | Phase 2 | 2016 | United States |
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2025 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Wang, S.; Huo, T.; Lu, M.; Zhao, Y.; Zhang, J.; He, W.; Chen, H. Recent Advances in Aging and Immunosenescence: Mechanisms and Therapeutic Strategies. Cells 2025, 14, 499. https://doi.org/10.3390/cells14070499
Wang S, Huo T, Lu M, Zhao Y, Zhang J, He W, Chen H. Recent Advances in Aging and Immunosenescence: Mechanisms and Therapeutic Strategies. Cells. 2025; 14(7):499. https://doi.org/10.3390/cells14070499
Chicago/Turabian StyleWang, Shuaiqi, Tong Huo, Mingyang Lu, Yueqi Zhao, Jianmin Zhang, Wei He, and Hui Chen. 2025. "Recent Advances in Aging and Immunosenescence: Mechanisms and Therapeutic Strategies" Cells 14, no. 7: 499. https://doi.org/10.3390/cells14070499
APA StyleWang, S., Huo, T., Lu, M., Zhao, Y., Zhang, J., He, W., & Chen, H. (2025). Recent Advances in Aging and Immunosenescence: Mechanisms and Therapeutic Strategies. Cells, 14(7), 499. https://doi.org/10.3390/cells14070499