New Trends in Aging Drug Discovery
Abstract
:1. Introduction
2. Cellular Pathways and Targets Involved in Cellular Senescence
3. Diseases Related to Senescence
Compound | Target/Pathway | Clinical Trial Status | |
---|---|---|---|
Age-Related Disease | Registration Number (Phase) | ||
Senolytics | |||
Dasatinib + Quercetin (D + Q) | Numerous (incl. PI3K/AKT and BCL-2) | Alzheimer’s disease | NCT04063124 (1/2) |
NCT05422885 (1/2) | |||
NCT04685590 (2) | |||
NCT04785300 (1/2) | |||
Idiopathic pulmonary fibrosis | NCT02874989 (1) | ||
Skeletal health | NCT04313634 (2) 1 | ||
Chronic kidney disease | NCT02848131 (2) | ||
Frailty | NCT04733534 (2) | ||
Diabetic chronic kidney disease | NCT02848131 (2) | ||
Epigenetic aging | NCT04946383 (2) | ||
Age-related bone loss | NCT04313634 (2) 2 | ||
Fisetin | Numerous (incl. PI3K/AKT, BCL-2, p53, and NF-kB) | Frail elderly syndrome | NCT03675724 (2) |
NCT03430037 (2) | |||
NCT04733534 (2) | |||
Knee osteoarthritis | NCT04210986 (1/2) | ||
NCT04770064 (1/2) | |||
NCT04210986 (1/2) | |||
NCT04815902 (1/2) | |||
UBX0101 | MDM2/p53 | Knee osteoarthritis | NCT04129944 (2) 3 |
NCT04349956 (2) | |||
UBX1325 | BCL-XL | Age-related macular degeneration | NCT05275205 (2) |
NCT04857996 (2) | |||
Curcumin 4,5 and EF-24 | Numerous (incl. Nrf2 and NF-kB) | Cellular models of senescence | |
Cardiac glycosides (ouabain, digoxin 5) | BCL-2, BCL-XL and BCL-W | Preclinical animal models | |
ABT-263 5 (Navitoclax) | BCL-2, BCL-XL and BCL-W | Preclinical animal models | |
Alvespimycin 5 (17-DMAG) | HSP90 | Preclinical animal models | |
Senomorphics | |||
Rapamycin 6 | Mtor (also Nrf2 and NF-κB) | Aging | NCT04488601 (2) |
NCT01649960 (1) | |||
NCT04742777 (2) | |||
NCT02874924 (2) | |||
NCT05237687 (2) | |||
Alzheimer disease | NCT04629495 (2) | ||
Amyotrophic lateral sclerosis | NCT03359538 (2) | ||
Metformin 7 | Numerous (incl. IKK, NF-κB, GPx7, and MBNL1) | Aging | NCT03309007 (3) |
NCT02432287 (4) | |||
NCT04264897 (3) | |||
NCT03451006 (2) | |||
Frailty | NCT03107884 (1) | ||
Muscle atrophy | NCT03107884 (1) | ||
BIRB796 8 | p38MAPK | Healthy 9 | NCT02211885 (1) |
NCT02209805 (1) | |||
RAD001 10 | mTOR | Preclinical animal models | |
NDGA 5 | unknown | Preclinical animal models | |
SR12343 | IKK/NF-κB | Preclinical animal models | |
Ruxolitinib 8,11 | JAK | Preclinical animal models | |
SRT12104 | SIRT1 | Preclinical animal models |
3.1. Neurodegenerative Diseases
3.2. Respiratory Diseases
3.3. Cardiovascular Diseases
3.4. Diabetes
3.5. Musculoskeletal Dysfunctions
3.6. Other Diseases
4. Senotherapeutic Molecules in Preclinical Studies and Clinical Trials
4.1. Senolytics
4.1.1. Natural Products
4.1.2. Repurposed Compounds
4.1.3. Other Senolytic Approaches
4.2. Senomorphics
5. Other Approaches
5.1. Blood Circulating Factors
5.2. The Gut Microbiome
5.3. Immune System
5.4. Geroprotection
6. Conclusions and Future Perspectives
Author Contributions
Funding
Conflicts of Interest
References
- López-Otín, C.; Blasco, M.A.; Partridge, L.; Serrano, M.; Kroemer, G. The hallmarks of aging. Cell 2013, 153, 1194–1217. [Google Scholar] [CrossRef] [PubMed]
- Van Deursen, J.M. The role of senescent cells in ageing. Nature 2014, 509, 439–446. [Google Scholar] [CrossRef] [PubMed]
- Gerdes, E.O.W.; Zhu, Y.; Tchkonia, T.; Kirkland, J.L. Discovery, development, and future application of senolytics: Theories and predictions. FEBS J. 2020, 287, 2418–2427. [Google Scholar] [CrossRef] [PubMed]
- Hayflick, L.; Moorhead, P.S. The serial cultivation of human diploid cell strains. Exp. Cell Res. 1961, 25, 585–621. [Google Scholar] [CrossRef]
- Muñoz-Espín, D.; Rovira, M.; Galiana, I.; Giménez, C.; Lozano-Torres, B.; Paez-Ribes, M.; Llanos, S.; Chaib, S.; Muñoz-Martín, M.; Ucero, A.C.; et al. A versatile drug delivery system targeting senescent cells. EMBO Mol. Med. 2018, 10, e9355. [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]
- Di Micco, R.; Krizhanovsky, V.; Baker, D.; d’Adda di Fagagna, F. Cellular senescence in ageing: From mechanisms to therapeutic opportunities. Nat. Rev. Mol. Cell Biol. 2021, 22, 75–95. [Google Scholar] [CrossRef]
- Zhang, L.; Pitcher, L.E.; Prahalad, V.; Niedernhofer, L.J.; Robbins, P.D. Targeting cellular senescence with senotherapeutics: Senolytics and senomorphics. FEBS J. 2022. [Google Scholar] [CrossRef]
- Baker, D.J.; Childs, B.G.; Durik, M.; Wijers, M.E.; Sieben, C.J.; Zhong, J.; Saltness, R.A.; Jeganathan, K.B.; Verzosa, G.C.; Pezeshki, A.; et al. Naturally occurring p16Ink4a-positive cells shorten healthy lifespan. Nature 2016, 530, 184–189. [Google Scholar] [CrossRef]
- 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]
- Czabotar, P.E.; Lessene, G.; Strasser, A.; Adams, J.M. Control of apoptosis by the BCL-2 protein family: Implications for physiology and therapy. Nat. Rev. Mol. Cell Biol. 2014, 15, 49–63. [Google Scholar] [CrossRef]
- Yosef, R.; Pilpel, N.; Tokarsky-Amiel, R.; Biran, A.; Ovadya, Y.; Cohen, S.; Vadai, E.; Dassa, L.; Shahar, E.; Condiotti, R.; et al. Directed elimination of senescent cells by inhibition of BCL-W and BCL-XL. Nat. Commun. 2016, 7, 11190. [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]
- 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]
- Zhu, Y.; Tchkonia, T.; Fuhrmann-Stroissnigg, H.; Dai, H.M.; Ling, Y.Y.; Stout, M.B.; Pirtskhalava, T.; Giorgadze, N.; Johnson, K.O.; Giles, C.B.; et al. Identification of a novel senolytic agent, navitoclax, targeting the Bcl-2 family of anti-apoptotic factors. Aging Cell 2016, 15, 428–435. [Google Scholar] [CrossRef]
- Cang, S.; Iragavarapu, C.; Savooji, J.; Song, Y.; Liu, D. ABT-199 (venetoclax) and BCL-2 inhibitors in clinical development. J. Hematol. Oncol. 2015, 8, 129. [Google Scholar] [CrossRef]
- Vousden, K.H.; Prives, C. Blinded by the light: The growing complexity of p53. Cell 2009, 137, 413–431. [Google Scholar] [CrossRef]
- Rufini, A.; Tucci, P.; Celardo, I.; Melino, G. Senescence and aging: The critical roles of p53. Oncogene 2013, 32, 5129–5143. [Google Scholar] [CrossRef]
- Bourgeois, B.; Madl, T. Regulation of cellular senescence via the FOXO4-p53 axis. FEBS Lett. 2018, 592, 2083–2097. [Google Scholar] [CrossRef]
- Baar, M.P.; Brandt, R.M.C.; Putavet, D.A.; Klein, J.D.D.; Derks, K.W.J.; Bourgeois, B.R.M.; Stryeck, S.; Rijksen, Y.; van Willigenburg, H.; Feijtel, D.A.; et al. Targeted apoptosis of senescent cells restores tissue homeostasis in response to chemotoxicity and aging. Cell 2017, 169, 132–147.e16. [Google Scholar] [CrossRef]
- Kruse, J.P.; Gu, W. Modes of p53 regulation. Cell 2009, 137, 609–622. [Google Scholar] [CrossRef]
- Jeon, O.H.; Kim, C.; Laberge, R.-M.; Demaria, M.; Rathod, S.; Vasserot, A.P.; Chung, J.W.; Kim, D.H.; Poon, Y.; David, N.; 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. [Google Scholar] [CrossRef]
- He, Y.; Li, W.; Lv, D.; Zhang, X.; Zhang, X.; Ortiz, Y.T.; Budamagunta, V.; Campisi, J.; Zheng, G.; Zhou, D. Inhibition of USP7 activity selectively eliminates senescent cells in part via restoration of p53 activity. Aging Cell 2020, 19, e13117. [Google Scholar] [CrossRef]
- Chauhan, D.; Tian, Z.; Nicholson, B.; Kumar, K.G.; Zhou, B.; Carrasco, R.; McDermott, J.L.; Leach, C.A.; Fulcinniti, M.; Kodrasov, M.P.; et al. A small molecule inhibitor of ubiquitin-specific protease-7 induces apoptosis in multiple myeloma cells and overcomes bortezomib resistance. Cancer Cell 2012, 22, 345–358. [Google Scholar] [CrossRef]
- Fan, Y.H.; Cheng, J.; Vasudevan, S.A.; Dou, J.; Zhang, H.; Patel, R.H.; Ma, I.T.; Rojas, Y.; Zhao, Y.; Yu, Y.; et al. USP7 inhibitor P22077 inhibits neuroblastoma growth via inducing p53-mediated apoptosis. Cell Death Dis. 2013, 4, e867. [Google Scholar] [CrossRef]
- Tavana, O.; Li, D.; Dai, C.; Lopez, G.; Banerjee, D.; Kon, N.; Chen, C.; Califano, A.; Yamashiro, D.J.; Sun, H.; et al. HAUSP deubiquitinates and stabilizes N-Myc in neuroblastoma. Nat. Med. 2016, 22, 1180–1186. [Google Scholar] [CrossRef]
- Fuhrmann-Stroissnigg, H.; Ling, Y.Y.; Zhao, J.; McGowan, S.J.; Zhu, Y.; Brooks, R.W.; Grassi, D.; Gregg, S.Q.; Stripay, J.L.; Dorronsoro, A.; et al. Identification of HSP90 inhibitors as a novel class of senolytics. Nat. Commun. 2017, 8, 422. [Google Scholar] [CrossRef]
- Pluquet, O.; Pourtier, A.; Abbadie, C. The unfolded protein response and cellular senescence. A review in the theme: Cellular mechanisms of endoplasmic reticulum stress signaling in health and disease. Am. J. Physiol. Cell Physiol. 2015, 308, C415–C425. [Google Scholar] [CrossRef]
- Acosta, J.C.; Banito, A.; Wuestefeld, T.; Georgilis, A.; Janich, P.; Morton, J.P.; Athineos, D.; Kang, T.W.; Lasitschka, F.; Andrulis, M.; et al. A complex secretory program orchestrated by the inflammasome controls paracrine senescence. Nat. Cell Biol. 2013, 15, 978–990. [Google Scholar] [CrossRef]
- Coppe, J.P.; Desprez, P.Y.; Krtolica, A.; Campisi, J. The senescence-associated secretory phenotype: The dark side of tumor suppression. Annu. Rev. Pathol. 2010, 5, 99–118. [Google Scholar] [CrossRef]
- Coppe, J.P.; Patil, C.K.; Rodier, F.; Sun, Y.; Munoz, D.P.; Goldstein, J.; Nelson, P.S.; Desprez, P.Y.; Campisi, J. Senescence-associated secretory phenotypes reveal cell-nonautonomous functions of oncogenic RAS and the p53 tumor suppressor. PLoS Biol. 2008, 6, 2853–2868. [Google Scholar] [CrossRef] [PubMed]
- Faget, D.V.; Ren, Q.; Stewart, S.A. Unmasking senescence: Context-dependent effects of SASP in cancer. Nat. Rev. Cancer 2019, 19, 439–453. [Google Scholar] [CrossRef] [PubMed]
- Osorio, F.G.; Soria-Valles, C.; Santiago-Fernandez, O.; Freije, J.M.; Lopez-Otin, C. NF-kappaB signaling as a driver of ageing. Int. Rev. Cell Mol. Biol. 2016, 326, 133–174. [Google Scholar] [PubMed]
- Salminen, A.; Kauppinen, A.; Kaarniranta, K. Emerging role of NF-kappaB signaling in the induction of senescence-associated secretory phenotype (SASP). Cell Signal. 2012, 24, 835–845. [Google Scholar] [CrossRef] [PubMed]
- Herranz, N.; Gallage, S.; Mellone, M.; Wuestefeld, T.; Klotz, S.; Hanley, C.J.; Raguz, S.; Acosta, J.C.; Innes, A.J.; Banito, A.; et al. mTOR regulates MAPKAPK2 translation to control the senescence-associated secretory phenotype. Nat. Cell Biol. 2015, 17, 1205–1217. [Google Scholar] [CrossRef]
- Laberge, R.-M.; Sun, Y.; Orjalo, A.V.; Patil, C.K.; Freund, A.; Zhou, L.; Curran, S.C.; Davalos, A.R.; Wilson-Edell, K.A.; Liu, S.; et al. MTOR regulates the pro-tumorigenic senescence-associated secretory phenotype by promoting IL1A translation. Nat. Cell Biol. 2015, 17, 1049–1061. [Google Scholar] [CrossRef]
- Freund, A.; Patil, C.K.; Campisi, J. p38MAPK is a novel DNA damage response-independent regulator of the senescence-associated secretory phenotype. EMBO J. 2011, 30, 1536–1548. [Google Scholar] [CrossRef]
- Hou, J.; Cui, C.; Kim, S.; Sung, C.; Choi, C. Ginsenoside F1 suppresses astrocytic senescence-associated secretory phenotype. Chem. Biol. Interact. 2018, 283, 75–83. [Google Scholar] [CrossRef]
- Zhao, J.; Zhang, L.; Lu, A.; Han, Y.; Colangelo, D.; Bukata, C.; Scibetta, A.; Yousefzadeh, M.J.; Li, X.; Gurkar, A.U.; et al. ATM is a key driver of NF-kB-dependent DNA-damage-induced senescence, stem cell dysfunction and aging. Aging 2020, 12, 4688–4710. [Google Scholar] [CrossRef]
- Chen, C.; Zhou, M.; Ge, Y.; Wang, X. SIRT1 and aging related signaling pathways. Mech. Ageing Dev. 2020, 187, 111215. [Google Scholar] [CrossRef]
- Moiseeva, O.; Deschenes-Simard, X.; St-Germain, E.; Igelmann, S.; Huot, G.; Cadar, A.E.; Bourdeau, V.; Pollak, M.N.; Ferbeyre, G. Metformin inhibits the senescence-associated secretory phenotype by interfering with IKK/NF-kappaB activation. Aging Cell 2013, 12, 489–498. [Google Scholar] [CrossRef]
- Perrott, K.M.; Wiley, C.D.; Desprez, P.Y.; Campisi, J. Apigenin suppresses the senescence-associated secretory phenotype and paracrine effects on breast cancer cells. Geroscience 2017, 39, 161–173. [Google Scholar] [CrossRef]
- Pitozzi, V.; Mocali, A.; Laurenzana, A.; Giannoni, E.; Cifola, I.; Battaglia, C.; Chiarugi, P.; Dolara, P.; Giovannelli, L. Chronic resveratrol treatment ameliorates cell adhesion and mitigates the inflammatory phenotype in senescent human fibroblasts. J. Gerontol. A Biol. Sci. Med. Sci. 2013, 68, 371–381. [Google Scholar] [CrossRef]
- Xu, M.; Tchkonia, T.; Ding, H.; Ogrodnik, M.; Lubbers, E.R.; Pirtskhalava, T.; White, T.A.; Johnson, K.O.; Stout, M.B.; Mezera, V.; et al. JAK inhibition alleviates the cellular senescence-associated secretory phenotype and frailty in old age. Proc. Natl. Acad. Sci. USA 2015, 112, E6301–E6310. [Google Scholar] [CrossRef]
- Liu, S.; Uppal, H.; Demaria, M.; Desprez, P.Y.; Campisi, J.; Kapahi, P. Simvastatin suppresses breast cancer cell proliferation induced by senescent cells. Sci. Rep. 2015, 5, 17895. [Google Scholar] [CrossRef]
- Crimmins, E.M. Lifespan and healthspan: Past, present, and promise. Gerontologist 2015, 55, 901–911. [Google Scholar] [CrossRef]
- McPhail, S.M. Multimorbidity in chronic disease: Impact on health care resources and costs. Risk Manag. Healthc. Policy 2016, 9, 143–156. [Google Scholar] [CrossRef]
- Noncommunicable Diseases. Available online: https://www.who.int/news-room/fact-sheets/detail/noncommunicable-diseases (accessed on 15 July 2022).
- Boccardi, V.; Mecocci, P. Senotherapeutics: Targeting senescent cells for the main age-related diseases. Mech. Ageing Dev. 2021, 197, 111526. [Google Scholar] [CrossRef]
- Dimri, G.P.; Lee, X.H.; Basile, G.; Acosta, M.; Scott, C.; Roskelley, C.; Medrano, E.E.; Linskens, M.; Rubelj, I.; Pereirasmith, O.; et al. A biomarker that identifies senescent human-cells in culture and in aging skin in-vivo. Proc. Natl. Acad. Sci. USA 1995, 92, 9363–9367. [Google Scholar] [CrossRef]
- Childs, B.G.; Baker, D.J.; Wijshake, T.; Conover, C.A.; Campisi, J.; van Deursen, J.M. Senescent intimal foam cells are deleterious at all stages of atherosclerosis. Science 2016, 354, 472–477. [Google Scholar] [CrossRef]
- Muñoz-Espin, D.; Demaria, M. Senolytics in Disease, Ageing and Longevity; Springer Nature Switzerland AG: Cham, Switzerland, 2020. [Google Scholar]
- Mongelli, A.; Atlante, S.; Barbi, V.; Bachetti, T.; Martelli, F.; Farsetti, A.; Gaetano, C. Treating Senescence like Cancer: Novel Perspectives in Senotherapy of Chronic Diseases. Int. J. Mol. Sci. 2020, 21, 7984. [Google Scholar] [CrossRef] [PubMed]
- Borghesan, M.; Hoogaars, W.M.H.; Varela-Eirin, M.; Talma, N.; Demaria, M. A senescence-centric view of aging: Implications for longevity and disease. Trends Cell Biol. 2020, 30, 777–791. [Google Scholar] [CrossRef] [PubMed]
- He, S.; Sharpless, N.E. Senescence in health and disease. Cell 2017, 169, 1000–1011. [Google Scholar] [CrossRef] [PubMed]
- Kirkland, J.L.; Tchkonia, T. Senolytic drugs: From discovery to translation. J. Intern. Med. 2020, 288, 518–536. [Google Scholar] [CrossRef] [PubMed]
- Roda, A.R.; Serra-Mir, G.; Montoliu-Gaya, L.; Tiessler, L.; Villegas, S. Amyloid-beta peptide and tau protein crosstalk in Alzheimer’s disease. Neural Regen. Res. 2022, 17, 1666–1674. [Google Scholar] [PubMed]
- Armstrong, M.J.; Okun, M.S. Diagnosis and treatment of parkinson disease a review. J. Am. Med. Assoc. 2020, 323, 548–560. [Google Scholar] [CrossRef] [PubMed]
- Si, Z.Z.; Sun, L.L.; Wang, X.D. Evidence and perspectives of cell senescence in neurodegenerative diseases. Biomed. Pharmacother. 2021, 137, 111327. [Google Scholar] [CrossRef]
- Bhat, R.; Crowe, E.P.; Bitto, A.; Moh, M.; Katsetos, C.D.; Garcia, F.U.; Johnson, F.B.; Trojanowski, J.Q.; Sell, C.; Torres, C. Astrocyte senescence as a component of Alzheimer’s disease. PLoS ONE 2012, 7, e45069. [Google Scholar] [CrossRef]
- Chinta, S.J.; Woods, G.; Demaria, M.; Rane, A.; Zou, Y.; McQuade, A.; Rajagopalan, S.; Limbad, C.; Madden, D.T.; Campisi, J.; 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. [Google Scholar] [CrossRef]
- PerezGrovas-Saltijeral, A.; Ochoa-Morales, A.; Miranda-Duarte, A.; Martinez-Ruano, L.; Jara-Prado, A.; Camacho-Molina, A.; Hidalgo-Bravo, A. Telomere length analysis on leukocytes derived from patients with Huntington Disease. Mech. Ageing Dev. 2020, 185, 111185. [Google Scholar] [CrossRef]
- Nicaise, A.M.; Wagstaff, L.J.; Willis, C.M.; Paisie, C.; Chandok, H.; Robson, P.; Fossati, V.; Williams, A.; Crocker, S.J. Cellular senescence in progenitor cells contributes to diminished remyelination potential in progressive multiple sclerosis. Proc. Natl. Acad. Sci. USA 2019, 116, 9030–9039. [Google Scholar] [CrossRef]
- Birger, A.; Ben-Dor, I.; Ottolenghi, M.; Turetsky, T.; Gil, Y.; Sweetat, S.; Perez, L.; Belzer, V.; Casden, N.; Steiner, D.; et al. Human iPSC-derived astrocytes from ALS patients with mutated C9ORF72 show increased oxidative stress and neurotoxicity. EBioMedicine 2019, 50, 274–289. [Google Scholar] [CrossRef]
- Jurk, D.; Wang, C.F.; Miwa, S.; Maddick, M.; Korolchuk, V.; Tsolou, A.; Gonos, E.S.; Thrasivoulou, C.; Saffrey, M.J.; Cameron, K.; et al. Postmitotic neurons develop a p21-dependent senescence-like phenotype driven by a DNA damage response. Aging Cell 2012, 11, 996–1004. [Google Scholar] [CrossRef]
- Musi, N.; Valentine, J.M.; Sickora, K.R.; Baeuerle, E.; Thompson, C.S.; Shen, Q.; Orr, M.E. Tau protein aggregation is associated with cellular senescence in the brain. Aging Cell 2018, 17, e12840. [Google Scholar] [CrossRef]
- Zhang, P.S.; Kishimoto, Y.; Grammatikakis, I.; Gottimukkala, K.; Cutler, R.G.; Zhang, S.L.; Abdelmohsen, K.; Bohr, V.A.; Sen, J.M.; Gorospe, M.; et al. Senolytic therapy alleviates Abeta-associated oligodendrocyte progenitor cell senescence and cognitive deficits in an Alzheimer’s disease model. Nat. Neurosci. 2019, 22, 719–728. [Google Scholar] [CrossRef]
- Han, X.; Zhang, T.; Liu, H.; Mi, Y.; Gou, X. Astrocyte senescence and alzheimer’s disease: A review. Front. Aging Neurosci. 2020, 12, 148. [Google Scholar] [CrossRef]
- Ogrodnik, M.; Evans, S.A.; Fielder, E.; Victorelli, S.; Kruger, P.; Salmonowicz, H.; Weigand, B.M.; Patel, A.D.; Pirtskhalava, T.; Inman, C.L.; et al. Whole-body senescent cell clearance alleviates age-related brain inflammation and cognitive impairment in mice. Aging Cell 2021, 20, e13296. [Google Scholar] [CrossRef]
- Elsallabi, O.; Patruno, A.; Pesce, M.; Cataldi, A.; Carradori, S.; Gallorini, M. Fisetin as a senotherapeutic agent: Biopharmaceutical properties and crosstalk between cell senescence and neuroprotection. Molecules 2022, 27, 738. [Google Scholar] [CrossRef]
- Bussian, T.J.; Aziz, A.; Meyer, C.F.; Swenson, B.L.; van Deursen, J.M.; Baker, D.J. Clearance of senescent glial cells prevents tau-dependent pathology and cognitive decline. Nature 2018, 562, 578–582. [Google Scholar] [CrossRef]
- Go, J.; Ha, T.K.Q.; Seo, J.Y.; Park, T.S.; Ryu, Y.K.; Park, H.Y.; Noh, J.R.; Kim, Y.H.; Hwang, J.H.; Choi, D.H.; et al. Piperlongumine activates Sirtuin1 and improves cognitive function in a murine model of Alzheimer’s disease. J. Funct. Foods 2018, 43, 103–111. [Google Scholar] [CrossRef]
- Katila, N.; Bhurtel, S.; Shadfar, S.; Srivastav, S.; Neupane, S.; Ojha, U.; Jeong, G.S.; Choi, D.Y. Metformin lowers alpha-synuclein phosphorylation and upregulates neurotrophic factor in the MPTP mouse model of Parkinson’s disease. Neuropharmacology 2017, 125, 396–407. [Google Scholar] [CrossRef]
- Van Skike, C.E.; Jahrling, J.B.; Olson, A.B.; Sayre, N.L.; Hussong, S.A.; Ungvari, Z.; Lechleiter, J.D.; Galvan, V. Inhibition of mTOR protects the blood-brain barrier in models of Alzheimer’s disease and vascular cognitive impairment. Am. J. Physiol.-Heart C 2018, 314, H693–H703. [Google Scholar] [CrossRef]
- Tang, Z.; Bereczki, E.; Zhang, H.Y.; Wang, S.; Li, C.X.; Ji, X.Y.; Branca, R.M.; Lehtio, J.; Guan, Z.Z.; Filipcik, P.; et al. Mammalian target of rapamycin (mtor) mediates tau protein dyshomeostasis implicaton fo alzheimer’s disease. J. Biol. Chem. 2013, 288, 15556–15570. [Google Scholar] [CrossRef]
- Ou, Z.R.; Kong, X.J.; Sun, X.D.; He, X.S.; Zhang, L.; Gong, Z.; Huang, J.Y.; Xu, B.A.; Long, D.H.; Li, J.H.; et al. Metformin treatment prevents amyloid plaque deposition and memory impairment in APP/PS1 mice. Brain Behav. Immun. 2018, 69, 351–363. [Google Scholar] [CrossRef]
- Gonzales, M.M.; Krishnamurthy, S.; Garbarino, V.; Daeihagh, A.S.; Gillispie, G.J.; Deep, G.; Craft, S.; Orr, M.E. A geroscience motivated approach to treat Alzheimer’s disease: Senolytics move to clinical trials. Mech. Ageing Dev. 2021, 200, 111589. [Google Scholar] [CrossRef]
- Hamsanathan, S.; Alder, J.K.; Sellares, J.; Rojas, M.; Gurkar, A.U.; Mora, A.L. Cellular senescence: The trojan horse in chronic lung diseases. Am. J. Respir. Cell Mol. Biol. 2019, 61, 21–30. [Google Scholar] [CrossRef]
- Liu, R.M.; Liu, G. Cell senescence and fibrotic lung diseases. Exp. Gerontol. 2020, 132, 110836. [Google Scholar] [CrossRef]
- Araya, J.; Kuwano, K. Cellular senescence-an aging hallmark in chronic obstructive pulmonary disease pathogenesis. Respir. Investig. 2022, 60, 33–44. [Google Scholar] [CrossRef] [PubMed]
- Barnes, P.J. Targeting cellular senescence as a new approach to chronic obstructive pulmonary disease therapy. Curr. Opin. Pharmacol. 2021, 56, 68–73. [Google Scholar] [CrossRef] [PubMed]
- Tuder, R.M. Aging and cigarette smoke: Fueling the fire. Am. J. Respir. Crit. Care Med. 2006, 174, 490–491. [Google Scholar] [CrossRef] [PubMed]
- Leung, J.; Cho, Y.; Lockey, R.F.; Kolliputi, N. The role of aging in idiopathic pulmonary fibrosis. Lung 2015, 193, 605–610. [Google Scholar] [CrossRef]
- Alvarez, D.; Cardenes, N.; Sellares, J.; Bueno, M.; Corey, C.; Hanumanthu, V.S.; Peng, Y.T.; D’Cunha, H.; Sembrat, J.; Nouraie, M.; et al. IPF lung fibroblasts have a senescent phenotype. Am. J. Physiol.-Lung C 2017, 313, L1164–L1173. [Google Scholar] [CrossRef]
- Schafer, M.J.; White, T.A.; Iijima, K.; Haak, A.J.; Ligresti, G.; Atkinson, E.J.; Oberg, A.L.; Birch, J.; Salmonowicz, H.; Zhu, Y.; et al. Cellular senescence mediates fibrotic pulmonary disease. Nat. Commun. 2017, 8, 14532. [Google Scholar] [CrossRef]
- Muller, K.C.; Welker, L.; Paasch, K.; Feindt, B.; Erpenbeck, V.; Hohlfeld, J.; Krug, N.; Nakashima, M.; Branscheid, D.; Magnussen, H.; et al. Lung fibroblasts from patients with emphysema show markers of senescence in vitro. Resp. Res. 2006, 7, 32. [Google Scholar] [CrossRef]
- Lehmann, M.; Korfei, M.; Mutze, K.; Klee, S.; Skronska-Wasek, W.; Alsafadi, H.N.; Ota, C.; Costa, R.; Schiller, H.B.; Lindner, M.; et al. Senolytic drugs target alveolar epithelial cell function and attenuate experimental lung fibrosis ex vivo. Eur. Resp. J. 2017, 50, 1602367. [Google Scholar] [CrossRef]
- Hohmann, M.S.; Habiel, D.M.; Coelho, A.L.; Verri, W.A.; Hogaboam, C.M. Quercetin enhances ligand-induced apoptosis in senescent idiopathic pulmonary fibrosis fibroblasts and reduces lung fibrosis in vivo. Am. J. Resp. Cell Mol. Biol. 2019, 60, 28–40. [Google Scholar] [CrossRef]
- Tsuji, T.; Aoshiba, K.; Nagai, A. Alveolar cell senescence in patients with pulmonary emphysema. Am. J. Resp. Crit. Care Med. 2006, 174, 886–893. [Google Scholar] [CrossRef]
- Baker, J.R.; Donnelly, L.E.; Barnes, P.J. Senotherapy a new horizon for COPD therapy. Chest 2020, 158, 562–570. [Google Scholar] [CrossRef]
- Lamming, D.W.; Ye, L.; Sabatini, D.M.; Baur, J.A. Rapalogs and mTOR inhibitors as anti-aging therapeutics. J. Clin. Investig. 2013, 123, 980–989. [Google Scholar] [CrossRef]
- Polverino, F.; Wu, T.D.; Rojas-Quintero, J.; Wang, X.Y.; Mayo, J.; Tomchaney, M.; Tram, J.; Packard, S.; Zhang, D.; Cleveland, K.H.; et al. Metformin: Experimental and clinical evidence for a potential role in emphysema treatment. Am. J. Resp. Crit. Care Med. 2021, 204, 651–666. [Google Scholar] [CrossRef]
- Hubbard, B.P.; Sinclair, D.A. Small molecule SIRT1 activators for the treatment of aging and age-related diseases. Trends Pharmacol. Sci. 2014, 35, 146–154. [Google Scholar] [CrossRef]
- Easter, M.; Bollenbecker, S.; Barnes, J.W.; Krick, S. Targeting aging pathways in chronic obstructive pulmonary disease. Int. J. Mol. Sci. 2020, 21, 6924. [Google Scholar] [CrossRef] [PubMed]
- Yen, F.S.; Chen, W.S.; Wei, J.C.C.; Hsu, C.C.; Hwu, C.M. Effects of metformin use on total mortality in patients with type 2 diabetes and chronic obstructive pulmonary disease: A matched-subject design. PLoS ONE 2018, 13, e0204859. [Google Scholar] [CrossRef] [PubMed]
- Roth, G.A.; Mensah, G.A.; Johnson, C.O.; Addolorato, G.; Ammirati, E.; Baddour, L.M.; Barengo, N.C.; Beaton, A.Z.; Benjamin, E.J.; Benziger, C.P.; et al. Global burden of cardiovascular diseases and risk factors, 1990–2019 update from the GBD 2019 study. J. Am. Coll. Cardiol. 2020, 76, 2982–3021. [Google Scholar] [CrossRef] [PubMed]
- Wang, J.C.; Bennett, M. Aging and atherosclerosis mechanisms, functional consequences, and potential therapeutics for cellular senescence. Circ. Res. 2012, 111, 245–259. [Google Scholar] [CrossRef] [PubMed]
- Lewis-McDougall, F.C.; Ruchaya, P.J.; Domenjo-Vila, E.; Teoh, T.S.; Prata, L.; Cottle, B.J.; Clark, J.E.; Punjabi, P.P.; Awad, W.; Torella, D.; et al. Aged-senescent cells contribute to impaired heart regeneration. Aging Cell 2019, 18, e12931. [Google Scholar] [CrossRef] [PubMed]
- Anderson, R.; Lagnado, A.; Maggiorani, D.; Walaszczyk, A.; Dookun, E.; Chapman, J.; Birch, J.; Salmonowicz, H.; Ogrodnik, M.; Jurk, D.; et al. Length-independent telomere damage drives post-mitotic cardiomyocyte senescence. EMBO J. 2019, 38, e100492. [Google Scholar] [CrossRef] [PubMed]
- El Hadri, K.; Smith, R.; Duplus, E.; El Amri, C. Inflammation, oxidative stress, senescence in atherosclerosis: Thioredoxine-1 as an emerging therapeutic target. Int. J. Mol. Sci. 2022, 23, 77. [Google Scholar] [CrossRef]
- Hamczyk, M.R.; Villa-Bellosta, R.; Quesada, V.; Gonzalo, P.; Vidak, S.; Nevado, R.M.; Andrés-Manzano, M.J.; Misteli, T.; López-Otín, C.; Andrés, V. Progerin accelerates atherosclerosis by inducing endoplasmic reticulum stress in vascular smooth muscle cells. EMBO Mol. Med. 2019, 11, e9736. [Google Scholar] [CrossRef]
- Macicior, J.; Marcos-Ramiro, B.; Ortega-Gutiérrez, S. Small-molecule therapeutic perspectives for the treatment of progeria. Int. J. Mol. Sci. 2021, 22, 7190. [Google Scholar] [CrossRef]
- Marcos-Ramiro, B.; Gil-Ordóñez, A.; Marín-Ramos, N.I.; Ortega-Nogales, F.J.; Balabasquer, M.; Gonzalo, P.; Khiar-Fernández, N.; Rolas, L.; Barkaway, A.; Nourshargh, S.; et al. Isoprenylcysteine carboxylmethyltransferase-based therapy for Hutchinson-Gilford progeria syndrome. ACS Cent. Sci. 2021, 7, 1300–1310. [Google Scholar] [CrossRef]
- Walaszczyk, A.; Dookun, E.; Redgrave, R.; Tual-Chalot, S.; Victorelli, S.; Spyridopoulos, I.; Owens, A.; Arthur, H.M.; Passos, J.F.; Richardson, G.D. Pharmacological clearance of senescent cells improves survival and recovery in aged mice following acute myocardial infarction. Aging Cell 2019, 18, e12945. [Google Scholar] [CrossRef]
- Roos, C.M.; Zhang, B.; Palmer, A.K.; Ogrodnik, M.B.; Pirtskhalava, T.; Thalji, N.M.; Hagler, M.; Jurk, D.; Smith, L.A.; Casaclang-Verzosa, G.; et al. Chronic senolytic treatment alleviates established vasomotor dysfunction in aged or atherosclerotic mice. Aging Cell 2016, 15, 973–977. [Google Scholar] [CrossRef]
- Owens, W.A.; Walaszczyk, A.; Spyridopoulos, I.; Dookun, E.; Richardson, G.D. Senescence and senolytics in cardiovascular disease: Promise and potential pitfalls. Mech. Ageing Dev. 2021, 198, 111540. [Google Scholar] [CrossRef]
- Sweeney, M.; Cook, S.A.; Gil, J. Therapeutic opportunities for senolysis in cardiovascular disease. FEBS J. 2022. [Google Scholar] [CrossRef]
- Shimizu, I.; Minamino, T. Cellular senescence in cardiac diseases. J. Cardiol. 2019, 74, 313–319. [Google Scholar] [CrossRef]
- Diabetes. Available online: https://www.who.int/health-topics/diabetes-tab=tab_1 (accessed on 15 July 2022).
- Palmer, A.K.; Gustafson, B.; Kirkland, J.L.; Smith, U. Cellular senescence: At the nexus between ageing and diabetes. Diabetologia 2019, 62, 1835–1841. [Google Scholar] [CrossRef]
- Palmer, A.K.; Xu, M.; Zhu, Y.; Pirtskhalava, T.; Weivoda, M.M.; Hachfeld, C.M.; Prata, L.G.; van Dijk, T.H.; Verkade, E.; Casaclang-Verzosa, G.; et al. Targeting senescent cells alleviates obesity-induced metabolic dysfunction. Aging Cell 2019, 18, e12950. [Google Scholar] [CrossRef]
- Aguayo-Mazzucato, C.; Andle, J.; Lee, T.B.; Midha, A.; Talemal, L.; Chipashvili, V.; Hollister-Lock, J.; van Deursen, J.; Weir, G.; Bonner-Weir, S. Acceleration of beta cell aging determines diabetes and senolysis improves disease outcomes. Cell Metab. 2019, 30, 129–142. [Google Scholar] [CrossRef]
- 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]
- 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]
- Thompson, P.J.; Shah, A.; Ntranos, V.; Van Gool, F.; Atkinson, M.; Bhushan, A. Targeted elimination of senescent beta cells prevents type 1 diabetes. Cell Metab. 2019, 29, 1045–1060. [Google Scholar] [CrossRef]
- Khosla, S.; Farr, J.N.; Tchkonia, T.; Kirkland, J.L. The role of cellular senescence in ageing and endocrine disease. Nat. Rev. Endocrinol. 2020, 16, 263–275. [Google Scholar] [CrossRef]
- Barzilai, N.R. Targeting aging with metformin (TAME). Innov. Aging 2017, 1, 743. [Google Scholar] [CrossRef]
- Kloppenburg, M.; Berenbaum, F. Osteoarthritis year in review 2019: Epidemiology and therapy. Osteoarth. Cartil. 2020, 28, 242–248. [Google Scholar] [CrossRef]
- Cruz-Jentoft, A.J.; Sayer, A.A. Sarcopenia. Lancet 2019, 393, 2636–2646. [Google Scholar] [CrossRef]
- Cao, X.; Luo, P.; Huang, J.J.; Liang, C.; He, J.S.; Wang, Z.L.; Shan, D.Y.; Peng, C.; Wu, S. Intraarticular senescent chondrocytes impair the cartilage regeneration capacity of mesenchymal stem cells. Stem Cell Res. Ther. 2019, 10, 86. [Google Scholar] [CrossRef]
- Farr, J.N.; Fraser, D.G.; Wang, H.; Jaehn, K.; Ogrodnik, M.B.; Weivoda, M.M.; Drake, M.T.; Tchkonia, T.; LeBrasseur, N.K.; Kirkland, J.L.; et al. Identification of senescent cells in the bone microenvironment. J. Bone Miner. Res. 2016, 31, 1920–1929. [Google Scholar] [CrossRef]
- Saito, Y.; Chikenji, T.S.; Matsumura, T.; Nakano, M.; Fujimiya, M. Exercise enhances skeletal muscle regeneration by promoting senescence in fibro-adipogenic progenitors. Nat. Commun. 2020, 11, 889. [Google Scholar] [CrossRef] [PubMed]
- Farr, J.N.; Xu, M.; Weivoda, M.M.; Monroe, D.G.; Fraser, D.G.; Onken, J.L.; Negley, B.A.; Sfeir, J.G.; Ogrodnik, M.B.; Hachfeld, C.M.; et al. Targeting cellular senescence prevents age-related bone loss in mice. Nat. Med. 2017, 23, 1072–1079. [Google Scholar] [CrossRef] [PubMed]
- Li, J.; Zhang, B.; Liu, W.X.; Lu, K.; Pan, H.B.; Wang, T.Y.; Oh, C.D.; Yi, D.; Huang, J.; Zhao, L.; et al. Metformin limits osteoarthritis development and progression through activation of AMPK signalling. Ann. Rheum. Dis. 2020, 79, 635–645. [Google Scholar] [CrossRef] [PubMed]
- Takayama, K.; Kawakami, Y.; Kobayashi, M.; Greco, N.; Cummins, J.H.; Matsushita, T.; Kuroda, R.; Kurosaka, M.; Fu, F.H.; Huard, J. Local intra-articular injection of rapamycin delays articular cartilage degeneration in a murine model of osteoarthritis. Arthritis Res. Ther. 2014, 16, 482. [Google Scholar] [CrossRef] [PubMed]
- Wan, M.; Gray-Gaillard, E.F.; Elisseeff, J.H. Cellular senescence in musculoskeletal homeostasis, diseases, and regeneration. Bone Res. 2021, 9, 41. [Google Scholar] [CrossRef] [PubMed]
- Ferreira-Gonzalez, S.; Rodrigo-Torres, D.; Gadd, V.L.; Forbes, S.J. Cellular senescence in liver disease and regeneration. Sem. Liver Dis. 2021, 41, 50–66. [Google Scholar] [CrossRef]
- Meijnikman, A.S.; Herrema, H.; Scheithauer, T.P.M.; Kroon, J.; Nieuwdorp, M.; Groen, A.K. Evaluating causality of cellular senescence in non-alcoholic fatty liver disease. JHEP Rep. 2021, 3, 100301. [Google Scholar] [CrossRef]
- Wang, W.J.; Cai, G.Y.; Chen, X.M. Cellular senescence, senescence-associated secretory phenotype, and chronic kidney disease. Oncotarget 2017, 8, 64520–64533. [Google Scholar] [CrossRef]
- Zhou, B.R.; Wan, Y.; Chen, R.; Zhang, C.M.; Li, X.S.; Meng, F.Y.; Glaser, S.; Wu, N.; Zhou, T.H.; Li, S.W.; et al. The emerging role of cellular senescence in renal diseases. J. Cell. Mol. Med. 2020, 24, 2087–2097. [Google Scholar] [CrossRef] [PubMed]
- Sreekumar, P.G.; Hinton, D.R.; Kannan, R. The emerging role of senescence in ocular disease. Oxid. Med. Cell. Longev. 2020, 2020, 2583601. [Google Scholar] [CrossRef]
- Sargiacomo, C.; Sotgia, F.; Lisanti, M.P. COVID-19 and chronological aging: Senolytics and other anti-aging drugs for the treatment or prevention of corona virus infection? Aging-US 2020, 12, 6511–6517. [Google Scholar] [CrossRef]
- Verdoorn, B.P.; Evans, T.K.; Hanson, G.J.; Zhu, Y.; Prata, L.G.P.L.; Pignolo, R.J.; Atkinson, E.J.; Wissler-Gerdes, E.O.; Kuchel, G.A.; Mannick, J.B.; et al. Fisetin for COVID-19 in skilled nursing facilities: Senolytic trials in the COVID era. J. Am. Geriatr. Soc. 2021, 69, 3023–3033. [Google Scholar] [CrossRef]
- Partridge, L.; Fuentealba, M.; Kennedy, B.K. The quest to slow ageing through drug discovery. Nat. Rev. Drug Discov. 2020, 19, 513–532. [Google Scholar] [CrossRef] [PubMed]
- Formica, J.V.; Regelson, W. Review of the biology of quercetin and related bioflavonoids. Food Chem. Toxicol. 1995, 33, 1061–1080. [Google Scholar] [CrossRef]
- Zhu, Y.; 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] [PubMed]
- Ogrodnik, M.; Miwa, S.; Tchkonia, T.; Tiniakos, D.; Wilson, C.L.; Lahat, A.; Day, C.P.; Burt, A.; Palmer, A.; Anstee, Q.M.; et al. Cellular senescence drives age-dependent hepatic steatosis. Nat. Commun. 2017, 8, 15691. [Google Scholar] [CrossRef] [PubMed]
- Ogrodnik, M.; Zhu, Y.; Langhi, L.G.P.; Tchkonia, T.; Krüger, P.; Fielder, E.; Victorelli, S.; Ruswhandi, R.A.; Giorgadze, N.; Pirtskhalava, T.; et al. Obesity-induced cellular senescence drives anxiety and impairs neurogenesis. Cell Metab. 2019, 29, 1061–1077. [Google Scholar] [CrossRef] [PubMed]
- Sundarraj, K.; Raghunath, A.; Perumal, E. A review on the chemotherapeutic potential of fisetin: In vitro evidences. Biomed. Pharmacother. 2018, 97, 928–940. [Google Scholar] [CrossRef]
- Syed, D.N.; Adhami, V.M.; Khan, N.; Khan, M.I.; Mukhtar, H. Exploring the molecular targets of dietary flavonoid fisetin in cancer. Semin. Cancer Biol. 2016, 40–41, 130–140. [Google Scholar] [CrossRef]
- Yang, D.; Tian, X.; Ye, Y.; Liang, Y.; Zhao, J.; Wu, T.; Lu, N. Identification of GL-V9 as a novel senolytic agent against senescent breast cancer cells. Life Sci. 2021, 272, 119196. [Google Scholar] [CrossRef]
- Li, H.; Hu, P.; Wang, Z.; Wang, H.; Yu, X.; Wang, X.; Qing, Y.; Zhu, M.; Xu, J.; Li, Z.; et al. Mitotic catastrophe and p53-dependent senescence induction in T-cell malignancies exposed to nonlethal dosage of GL-V9. Arch. Toxicol. 2020, 94, 305–323. [Google Scholar] [CrossRef]
- Wang, Y.; Chang, J.; Liu, X.; Zhang, X.; Zhang, S.; Zhang, X.; Zhou, D.; Zheng, G. Discovery of piperlongumine as a potential novel lead for the development of senolytic agents. Aging 2016, 8, 2915–2926. [Google Scholar] [CrossRef]
- Zhang, X.; Zhang, S.; Liu, X.; Wang, Y.; Chang, J.; Zhang, X.; Mackintosh, S.G.; Tackett, A.J.; He, Y.; Lv, D.; et al. Oxidation resistance 1 is a novel senolytic target. Aging Cell 2018, 17, e12780. [Google Scholar] [CrossRef]
- Liu, X.; Wang, Y.; Zhang, X.; Gao, Z.; Zhang, S.; Shi, P.; Zhang, X.; Song, L.; Hendrickson, H.; Zhou, D.; et al. Senolytic activity of piperlongumine analogues: Synthesis and biological evaluation. Bioorg. Med. Chem. 2018, 26, 3925–3938. [Google Scholar] [CrossRef]
- Hewlings, S.J.; Kalman, D.S. Curcumin: A review of its effects on human health. Foods 2017, 6, 92. [Google Scholar] [CrossRef]
- Cherif, H.; Bisson, D.G.; Jarzem, P.; Weber, M.; Ouellet, J.A.; Haglund, L. Curcumin and o-vanillin exhibit evidence of senolytic activity in human IVD Cells in vitro. J. Clin. Med. 2019, 8, 433. [Google Scholar] [CrossRef]
- Li, W.; He, Y.; Zhang, R.; Zheng, G.; Zhou, D. The curcumin analog EF24 is a novel senolytic agent. Aging 2019, 11, 771–782. [Google Scholar] [CrossRef]
- Triana-Martínez, F.; Picallos-Rabina, P.; Da Silva-Álvarez, S.; Pietrocola, F.; Llanos, S.; Rodilla, V.; Soprano, E.; Pedrosa, P.; Ferreirós, A.; Barradas, M.; et al. Identification and characterization of cardiac glycosides as senolytic compounds. Nat. Commun. 2019, 10, 4731. [Google Scholar] [CrossRef]
- Guerrero, A.; Herranz, N.; Sun, B.; Wagner, V.; Gallage, S.; Guiho, R.; Wolter, K.; Pombo, J.; Irvine, E.E.; Innes, A.J.; et al. Cardiac glycosides are broad-spectrum senolytics. Nat. Metab. 2019, 1, 1074–1088. [Google Scholar] [CrossRef]
- Tse, C.; Shoemaker, A.R.; Adickes, J.; Anderson, M.G.; Chen, J.; Jin, S.; Johnson, E.F.; Marsh, K.C.; Mitten, M.J.; Nimmer, P.; et al. ABT-263: A potent and orally bioavailable Bcl-2 family inhibitor. Cancer Res. 2008, 68, 3421–3428. [Google Scholar] [CrossRef]
- Pan, J.; Li, D.; Xu, Y.; Zhang, J.; Wang, Y.; Chen, M.; Lin, S.; Huang, L.; Chung, E.J.; Citrin, D.E.; et al. Inhibition of Bcl-2/xl With ABT-263 Selectively Kills Senescent Type II Pneumocytes and Reverses Persistent Pulmonary Fibrosis Induced by Ionizing Radiation in Mice. Int. J. Radiat. Oncol. Biol. Phys. 2017, 99, 353–361. [Google Scholar] [CrossRef]
- Mellatyar, H.; Talaei, S.; Pilehvar-Soltanahmadi, Y.; Barzegar, A.; Akbarzadeh, A.; Shahabi, A.; Barekati-Mowahed, M.; Zarghami, N. Targeted cancer therapy through 17-DMAG as an Hsp90 inhibitor: Overview and current state of the art. Biomed. Pharmacother. 2018, 102, 608–617. [Google Scholar] [CrossRef]
- Morsli, S.; Doherty, G.J.; Muñoz-Espín, D. Activatable senoprobes and senolytics: Novel strategies to detect and target senescent cells. Mech. Ageing Dev. 2022, 202, 111618. [Google Scholar] [CrossRef]
- González-Gualda, E.; Pàez-Ribes, M.; Lozano-Torres, B.; Macias, D.; Wilson, J.R., III; González-López, C.; Ou, H.-L.; Mirón-Barroso, S.; Zhang, Z.; Lérida-Viso, A.; et al. Galacto-conjugation of Navitoclax as an efficient strategy to increase senolytic specificity and reduce platelet toxicity. Aging Cell 2020, 19, e13142. [Google Scholar] [CrossRef]
- Galiana, I.; Lozano-Torres, B.; Sancho, M.; Alfonso, M.; Bernardos, A.; Bisbal, V.; Serrano, M.; Martínez-Máñez, R.; Orzáez, M. Preclinical antitumor efficacy of senescence-inducing chemotherapy combined with a nanoSenolytic. J. Control. Release 2020, 323, 624–634. [Google Scholar] [CrossRef]
- He, Y.; Zhang, X.; Chang, J.; Kim, H.-N.; Zhang, P.; Wang, Y.; Khan, S.; Liu, X.; Zhang, X.; Lv, D.; et al. Using proteolysis-targeting chimera technology to reduce navitoclax platelet toxicity and improve its senolytic activity. Nat. Commun. 2020, 11, 1996. [Google Scholar] [CrossRef]
- Vézina, C.; Kudelski, A.; Sehgal, S.N. Rapamycin (AY-22,989), a new antifungal antibiotic. I. Taxonomy of the producing streptomycete and isolation of the active principle. J. Antibiot. 1975, 28, 721–726. [Google Scholar] [CrossRef]
- Flynn, J.M.; O’Leary, M.N.; Zambataro, C.A.; Academia, E.C.; Presley, M.P.; Garrett, B.J.; Zykovich, A.; Mooney, S.D.; Strong, R.; Rosen, C.J.; et al. Late-life rapamycin treatment reverses age-related heart dysfunction. Aging Cell 2013, 12, 851–862. [Google Scholar] [CrossRef] [PubMed]
- Majumder, S.; Caccamo, A.; Medina, D.X.; Benavides, A.D.; Javors, M.A.; Kraig, E.; Strong, R.; Richardson, A.; Oddo, S. Lifelong rapamycin administration ameliorates age-dependent cognitive deficits by reducing IL-1β and enhancing NMDA signaling. Aging Cell 2012, 11, 326–335. [Google Scholar] [CrossRef] [PubMed]
- Bjedov, I.; Rallis, C. The target of rapamycin signalling pathway in ageing and lifespan regulation. Genes 2020, 11, 1043. [Google Scholar] [CrossRef] [PubMed]
- Wang, R.; Yu, Z.; Sunchu, B.; Shoaf, J.; Dang, I.; Zhao, S.; Caples, K.; Bradley, L.; Beaver, L.M.; Ho, E.; et al. Rapamycin inhibits the secretory phenotype of senescent cells by a Nrf2-independent mechanism. Aging Cell 2017, 16, 564–574. [Google Scholar] [CrossRef]
- Li, J.; Kim, S.G.; Blenis, J. Rapamycin: One Drug, many effects. Cell Metab. 2014, 19, 373–379. [Google Scholar] [CrossRef]
- Lim, H.; Park, H.; Kim, H.P. Effects of flavonoids on senescence-associated secretory phenotype formation from bleomycin-induced senescence in BJ fibroblasts. Biochem. Pharmacol. 2015, 96, 337–348. [Google Scholar] [CrossRef]
- Harrison, D.E.; Strong, R.; Allison, D.B.; Ames, B.N.; Astle, C.M.; Atamna, H.; Fernandez, E.; Flurkey, K.; Javors, M.A.; Nadon, N.L.; et al. Acarbose, 17-α-estradiol, and nordihydroguaiaretic acid extend mouse lifespan preferentially in males. Aging Cell 2014, 13, 273–282. [Google Scholar] [CrossRef]
- Campbell, J.M.; Bellman, S.M.; Stephenson, M.D.; Lisy, K. Metformin reduces all-cause mortality and diseases of ageing independent of its effect on diabetes control: A systematic review and meta-analysis. Ageing Res. Rev. 2017, 40, 31–44. [Google Scholar] [CrossRef]
- Bannister, C.A.; Holden, S.E.; Jenkins-Jones, S.; Morgan, C.L.; Halcox, J.P.; Schernthaner, G.; Mukherjee, J.; Currie, C.J. Can people with type 2 diabetes live longer than those without? A comparison of mortality in people initiated with metformin or sulphonylurea monotherapy and matched, non-diabetic controls. Diabetes Obes. Metab. 2014, 16, 1165–1173. [Google Scholar] [CrossRef]
- The TAME Trial. Available online: https://www.afar.org/tame-trial (accessed on 15 July 2022).
- Tilstra, J.S.; Robinson, A.R.; Wang, J.; Gregg, S.Q.; Clauson, C.L.; Reay, D.P.; Nasto, L.A.; St Croix, C.M.; Usas, A.; Vo, N.; et al. NF-κB inhibition delays DNA damage–induced senescence and aging in mice. J. Clin. Investig. 2012, 122, 2601–2612. [Google Scholar] [CrossRef]
- Zhang, L.; Zhao, J.; Mu, X.; McGowan, S.J.; Angelini, L.; O’Kelly, R.D.; Yousefzadeh, M.J.; Sakamoto, A.; Aversa, Z.; LeBrasseur, N.K.; et al. Novel small molecule inhibition of IKK/NF-κB activation reduces markers of senescence and improves healthspan in mouse models of aging. Aging Cell 2021, 20, e13486. [Google Scholar] [CrossRef]
- Alimbetov, D.; Davis, T.; Brook, A.J.C.; Cox, L.S.; Faragher, R.G.A.; Nurgozhin, T.; Zhumadilov, Z.; Kipling, D. Suppression of the senescence-associated secretory phenotype (SASP) in human fibroblasts using small molecule inhibitors of p38 MAP kinase and MK2. Biogerontology 2016, 17, 305–315. [Google Scholar] [CrossRef]
- Griveau, A.; Wiel, C.; Ziegler, D.V.; Bergo, M.O.; Bernard, D. The JAK1/2 inhibitor ruxolitinib delays premature aging phenotypes. Aging Cell 2020, 19, e13122. [Google Scholar] [CrossRef]
- Kang, H.T.; Park, J.T.; Choi, K.; Kim, Y.; Choi, H.J.C.; Jung, C.W.; Lee, Y.-S.; Park, S.C. Chemical screening identifies ATM as a target for alleviating senescence. Nat. Chem. Biol. 2017, 13, 616–623. [Google Scholar] [CrossRef]
- Kuk, M.U.; Kim, J.W.; Lee, Y.; Cho, K.A.; Park, J.T.; Park, S.C. Alleviation of senescence via ATM inhibition in accelerated aging models. Mol. Cells 2019, 42, 210–217. [Google Scholar]
- Liu, J.; Jiao, K.; Zhou, Q.; Yang, J.; Yang, K.; Hu, C.; Zhou, M.; Li, Z. Resveratrol alleviates 27-hydroxycholesterol-induced senescence in nerve cells and affects zebrafish locomotor behavior via activation of SIRT1-mediated STAT3 signaling. Oxid. Med. Cell. Longev. 2021, 2021, 6673343. [Google Scholar] [CrossRef]
- Csiszar, A.; Sosnowska, D.; Wang, M.; Lakatta, E.G.; Sonntag, W.E.; Ungvari, Z. Age-associated proinflammatory secretory phenotype in vascular smooth muscle cells from the non-human primate Macaca mulatta: Reversal by resveratrol treatment. J. Gerontol. A Biol. Sci. Med. Sci. 2012, 67, 811–820. [Google Scholar] [CrossRef]
- Mitchell, S.J.; Martin-Montalvo, A.; Mercken, E.M.; Palacios, H.H.; Ward, T.M.; Abulwerdi, G.; Minor, R.K.; Vlasuk, G.P.; Ellis, J.L.; Sinclair, D.A.; et al. The SIRT1 activator SRT1720 extends lifespan and improves health of mice fed a standard diet. Cell Rep. 2014, 6, 836–843. [Google Scholar] [CrossRef]
- Conboy, M.J.; Conboy, I.M.; Rando, T.A. Heterochronic parabiosis: Historical perspective and methodological considerations for studies of aging and longevity. Aging Cell 2013, 12, 525–530. [Google Scholar] [CrossRef]
- Yousefzadeh, M.J.; Wilkinson, J.E.; Hughes, B.; Gadela, N.; Ladiges, W.C.; Vo, N.; Niedernhofer, L.J.; Huffman, D.M.; Robbins, P.D. Heterochronic parabiosis regulates the extent of cellular senescence in multiple tissues. Geroscience 2020, 42, 951–961. [Google Scholar] [CrossRef]
- Conboy, I.M.; Conboy, M.J.; Wagers, A.J.; Girma, E.R.; Weissman, I.L.; Rando, T.A. Rejuvenation of aged progenitor cells by exposure to a young systemic environment. Nature 2005, 433, 760–764. [Google Scholar] [CrossRef]
- Ruckh, J.M.; Zhao, J.W.; Shadrach, J.L.; van Wijngaarden, P.; Rao, T.N.; Wagers, A.J.; Franklin, R.J. Rejuvenation of regeneration in the aging central nervous system. Cell Stem Cell 2012, 10, 96–103. [Google Scholar] [CrossRef]
- Villeda, S.A.; Luo, J.; Mosher, K.I.; Zou, B.; Britschgi, M.; Bieri, G.; Stan, T.M.; Fainberg, N.; Ding, Z.; Eggel, A.; et al. The ageing systemic milieu negatively regulates neurogenesis and cognitive function. Nature 2011, 477, 90–94. [Google Scholar] [CrossRef]
- Villeda, S.A.; Plambeck, K.E.; Middeldorp, J.; Castellano, J.M.; Mosher, K.I.; Luo, J.; Smith, L.K.; Bieri, G.; Lin, K.; Berdnik, D.; et al. Young blood reverses age-related impairments in cognitive function and synaptic plasticity in mice. Nat. Med. 2014, 20, 659–663. [Google Scholar] [CrossRef]
- Huang, Q.; Ning, Y.; Liu, D.; Zhang, Y.; Li, D.; Zhang, Y.; Yin, Z.; Fu, B.; Cai, G.; Sun, X.; et al. A young blood environment decreases aging of senile mice kidneys. J. Gerontol. A Biol. Sci. Med. Sci. 2018, 73, 421–428. [Google Scholar] [CrossRef]
- Salpeter, S.J.; Khalaileh, A.; Weinberg-Corem, N.; Ziv, O.; Glaser, B.; Dor, Y. Systemic regulation of the age-related decline of pancreatic β-cell replication. Diabetes 2013, 62, 2843–2848. [Google Scholar] [CrossRef] [PubMed]
- Baht, G.S.; Silkstone, D.; Vi, L.; Nadesan, P.; Amani, Y.; Whetstone, H.; Wei, Q.; Alman, B.A. Exposure to a youthful circulation rejuvenates bone repair through modulation of β-catenin. Nat. Commun. 2015, 6, 7131. [Google Scholar] [CrossRef] [PubMed]
- Kiss, T.; Nyúl-Tóth, Á.; Gulej, R.; Tarantini, S.; Csipo, T.; Mukli, P.; Ungvari, A.; Balasubramanian, P.; Yabluchanskiy, A.; Benyo, Z.; et al. Old blood from heterochronic parabionts accelerates vascular aging in young mice: Transcriptomic signature of pathologic smooth muscle remodeling. Geroscience 2022, 44, 953–981. [Google Scholar] [CrossRef] [PubMed]
- Kiss, T.; Tarantini, S.; Csipo, T.; Balasubramanian, P.; Nyúl-Tóth, Á.; Yabluchanskiy, A.; Wren, J.D.; Garman, L.; Huffman, D.M.; Csiszar, A.; et al. Circulating anti-geronic factors from heterochonic parabionts promote vascular rejuvenation in aged mice: Transcriptional footprint of mitochondrial protection, attenuation of oxidative stress, and rescue of endothelial function by young blood. Geroscience 2020, 42, 727–748. [Google Scholar] [CrossRef]
- Smith, L.K.; He, Y.; Park, J.S.; Bieri, G.; Snethlage, C.E.; Lin, K.; Gontier, G.; Wabl, R.; Plambeck, K.E.; Udeochu, J.; et al. β2-microglobulin is a systemic pro-aging factor that impairs cognitive function and neurogenesis. Nat. Med. 2015, 21, 932–937. [Google Scholar] [CrossRef]
- Li, R.; Liang, Y.; Lin, B. Accumulation of systematic TPM1 mediates inflammation and neuronal remodeling by phosphorylating PKA and regulating the FABP5/NF-κB signaling pathway in the retina of aged mice. Aging Cell 2022, 21, e13566. [Google Scholar] [CrossRef]
- Castellano, J.M.; Mosher, K.I.; Abbey, R.J.; McBride, A.A.; James, M.L.; Berdnik, D.; Shen, J.C.; Zou, B.; Xie, X.S.; Tingle, M.; et al. Human umbilical cord plasma proteins revitalize hippocampal function in aged mice. Nature 2017, 544, 488–492. [Google Scholar] [CrossRef]
- Iram, T.; Kern, F.; Kaur, A.; Myneni, S.; Morningstar, A.R.; Shin, H.; Garcia, M.A.; Yerra, L.; Palovics, R.; Yang, A.C.; et al. Young CSF restores oligodendrogenesis and memory in aged mice via Fgf17. Nature 2022, 605, 509–515. [Google Scholar] [CrossRef]
- Smith, L.K.; Verovskaya, E.; Bieri, G.; Horowitz, A.M.; von Ungern-Sternberg, S.N.I.; Lin, K.; Seizer, P.; Passegué, E.; Villeda, S.A. The aged hematopoietic system promotes hippocampal-dependent cognitive decline. Aging Cell 2020, 19, e13192. [Google Scholar] [CrossRef]
- Ho, T.T.; Dellorusso, P.V.; Verovskaya, E.V.; Bakker, S.T.; Flach, J.; Smith, L.K.; Ventura, P.B.; Lansinger, O.M.; Hérault, A.; Zhang, S.Y.; et al. Aged hematopoietic stem cells are refractory to bloodborne systemic rejuvenation interventions. J. Exp. Med. 2021, 218, e20210223. [Google Scholar] [CrossRef]
- Levy, M.; Kolodziejczyk, A.A.; Thaiss, C.A.; Elinav, E. Dysbiosis and the immune system. Nat. Rev. Immunol. 2017, 17, 219–232. [Google Scholar] [CrossRef]
- Chaudhari, S.N.; McCurry, M.D.; Devlin, A.S. Chains of evidence from correlations to causal molecules in microbiome-linked diseases. Nat. Chem. Biol. 2021, 17, 1046–1056. [Google Scholar] [CrossRef]
- Helmink, B.A.; Khan, M.A.W.; Hermann, A.; Gopalakrishnan, V.; Wargo, J.A. The microbiome, cancer, and cancer therapy. Nat. Med. 2019, 25, 377–388. [Google Scholar] [CrossRef]
- Papadopoulos, P.D.; Tsigalou, C.; Valsamaki, P.N.; Konstantinidis, T.G.; Voidarou, C.; Bezirtzoglou, E. The emerging role of the gut microbiome in cardiovascular disease: Current knowledge and perspectives. Biomedicines 2022, 10, 948. [Google Scholar] [CrossRef]
- Vallianou, N.; Christodoulatos, G.S.; Karampela, I.; Tsilingiris, D.; Magkos, F.; Stratigou, T.; Kounatidis, D.; Dalamaga, M. Understanding the role of the gut microbiome and microbial metabolites in non-alcoholic fatty liver disease: Current evidence and perspectives. Biomolecules 2022, 12, 56. [Google Scholar] [CrossRef]
- Zhou, Y.; Hu, G.; Wang, M.C. Host and microbiota metabolic signals in aging and longevity. Nat. Chem. Biol. 2021, 17, 1027–1036. [Google Scholar] [CrossRef]
- Claesson, M.J.; Jeffery, I.B.; Conde, S.; Power, S.E.; O’Connor, E.M.; Cusack, S.; Harris, H.M.; Coakley, M.; Lakshminarayanan, B.; O’Sullivan, O.; et al. Gut microbiota composition correlates with diet and health in the elderly. Nature 2012, 488, 178–184. [Google Scholar] [CrossRef]
- Shin, J.; Noh, J.R.; Choe, D.; Lee, N.; Song, Y.; Cho, S.; Kang, E.J.; Go, M.J.; Ha, S.K.; Chang, D.H.; et al. Ageing and rejuvenation models reveal changes in key microbial communities associated with healthy ageing. Microbiome 2021, 9, 240. [Google Scholar] [CrossRef]
- Bárcena, C.; Valdés-Mas, R.; Mayoral, P.; Garabaya, C.; Durand, S.; Rodríguez, F.; Fernández-García, M.T.; Salazar, N.; Nogacka, A.M.; Garatachea, N.; et al. Healthspan and lifespan extension by fecal microbiota transplantation into progeroid mice. Nat. Med. 2019, 25, 1234–1242. [Google Scholar] [CrossRef]
- Biagi, E.; Franceschi, C.; Rampelli, S.; Severgnini, M.; Ostan, R.; Turroni, S.; Consolandi, C.; Quercia, S.; Scurti, M.; Monti, D.; et al. Gut microbiota and extreme longevity. Curr. Biol. 2016, 26, 1480–1485. [Google Scholar] [CrossRef]
- Sato, Y.; Atarashi, K.; Plichta, D.R.; Arai, Y.; Sasajima, S.; Kearney, S.M.; Suda, W.; Takeshita, K.; Sasaki, T.; Okamoto, S.; et al. Novel bile acid biosynthetic pathways are enriched in the microbiome of centenarians. Nature 2021, 599, 458–464. [Google Scholar] [CrossRef]
- Borgoni, S.; Kudryashova, K.S.; Burka, K.; de Magalhães, J.P. Targeting immune dysfunction in aging. Ageing Res. Rev. 2021, 70, 101410. [Google Scholar] [CrossRef]
- Cunha, L.L.; Perazzio, S.F.; Azzi, J.; Cravedi, P.; Riella, L.V. Remodeling of the immune response with aging: Immunosenescence and its potential impact on COVID-19 immune response. Front. Immunol. 2020, 11, 1748. [Google Scholar] [CrossRef]
- 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]
- Martínez, P.; Blasco, M.A. Telomere-driven diseases and telomere-targeting therapies. J. Cell Biol. 2017, 216, 875–887. [Google Scholar] [CrossRef]
- Fischer, F.; Grigolon, G.; Benner, C.; Ristow, M. Evolutionarily conserved transcription factors as regulators of longevity and targets for geroprotection. Physiol. Rev. 2022, 102, 1449–1494. [Google Scholar] [CrossRef]
- Secci, R.; Hartmann, A.; Walter, M.; Grabe, H.J.; Van der Auwera-Palitschka, S.; Kowald, A.; Palmer, D.; Rimbach, G.; Fuellen, G.; Barrantes, I. Biomarkers of geroprotection and cardiovascular health: An overview of omics studies and established clinical biomarkers in the context of diet. Crit. Rev. Food Sci. Nutr. 2021, 1–21. [Google Scholar] [CrossRef]
- Van Praag, H.; Shubert, T.; Zhao, C.; Gage, F.H. Exercise enhances learning and hippocampal neurogenesis in aged mice. J. Neurosci. 2005, 25, 8680–8685. [Google Scholar] [CrossRef]
- Speisman, R.B.; Kumar, A.; Rani, A.; Foster, T.C.; Ormerod, B.K. Daily exercise improves memory, stimulates hippocampal neurogenesis and modulates immune and neuroimmune cytokines in aging rats. Brain Behav. Immun. 2013, 28, 25–43. [Google Scholar] [CrossRef]
- Horowitz, A.M.; Fan, X.; Bieri, G.; Smith, L.K.; Sanchez-Diaz, C.I.; Schroer, A.B.; Gontier, G.; Casaletto, K.B.; Kramer, J.H.; Williams, K.E.; et al. Blood factors transfer beneficial effects of exercise on neurogenesis and cognition to the aged brain. Science 2020, 369, 167–173. [Google Scholar] [CrossRef] [PubMed]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2022 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
Benhamú, B.; Martín-Fontecha, M.; Vázquez-Villa, H.; López-Rodríguez, M.L.; Ortega-Gutiérrez, S. New Trends in Aging Drug Discovery. Biomedicines 2022, 10, 2006. https://doi.org/10.3390/biomedicines10082006
Benhamú B, Martín-Fontecha M, Vázquez-Villa H, López-Rodríguez ML, Ortega-Gutiérrez S. New Trends in Aging Drug Discovery. Biomedicines. 2022; 10(8):2006. https://doi.org/10.3390/biomedicines10082006
Chicago/Turabian StyleBenhamú, Bellinda, Mar Martín-Fontecha, Henar Vázquez-Villa, María L. López-Rodríguez, and Silvia Ortega-Gutiérrez. 2022. "New Trends in Aging Drug Discovery" Biomedicines 10, no. 8: 2006. https://doi.org/10.3390/biomedicines10082006
APA StyleBenhamú, B., Martín-Fontecha, M., Vázquez-Villa, H., López-Rodríguez, M. L., & Ortega-Gutiérrez, S. (2022). New Trends in Aging Drug Discovery. Biomedicines, 10(8), 2006. https://doi.org/10.3390/biomedicines10082006