Nano-Based Theranostic Tools for the Detection and Elimination of Senescent Cells
Abstract
1. Cellular Senescence
2. Nano-Based Delivery Systems for Diagnostic and Therapeutic Purposes
3. Senolytics and Senotherapy
4. Nanomaterials for the Clearance of Senescent Cells
5. Conclusions
Author Contributions
Funding
Acknowledgments
Conflicts of Interest
Abbreviations
| 17-AAG | tanespimycin |
| 17-DMAG | alvespimycin |
| AKT | protein kinase B |
| AMPK | AMP-activated protein kinase |
| B2M | Beta-2 microglobulin |
| Bcl-2 | B cell lymphoma 2 family |
| BETd | BET family protein degrader |
| CAR | Chimeric antigen receptor |
| CCL-16 | Monotactin-1 |
| CDKI | Cyclin-dependent kinase inhibitor |
| CS | Cellular senescence |
| DAMPs | Damage-associated molecular patterns |
| DDR | DNA damage response |
| DNA | Deoxyribonucleic acid |
| Dox | Doxorubicin |
| DSAs | Docetaxel-tannic acid self-assemblies |
| EGF | Endothelial growth factor |
| EGF | Epidermal growth factor |
| EVs | Extracellular vesicles |
| FOXO4 | Forkhead box protein O4 |
| Gal | Galacto-oligosaccharide |
| Gal6 | 6-mer galacto-oligosaccharide |
| HAECs | Human aortic endothelial cells |
| HDF | Human dermal fibroblasts |
| HIF-1α | Hypoxia inducible factor 1α |
| HSP90 | Chaperone heat shock protein 90 |
| IFN-β | Interferon beta |
| IGF-1 | Insulin-like growth factor 1 |
| IL-6 | Interleukin 6 |
| IL-8 | Interleukin 8 |
| IL-1β | Interleukin 1β |
| JAK | Janus kinase |
| Lac-PEG | Lactose-polyethylene glycol |
| LR | Rapamycin-loaded PEGylated liposomes |
| MAPK | Mitogen-activated protein kinase |
| MCP-1 | Monocyte chemoattractant protein 1 |
| MIP-1α | Macrophage inflammatory protein 1α |
| miRNA | MicroRNA |
| MMPs | Metalloproteinases |
| MoS2 | Molybdenum disulfide |
| MNPQ | Quercetin surface-functionalized Fe3O4 nanoparticles |
| MSN | Mesoporous silica nanoparticles |
| mTOR | Mammalian target of rapamycin |
| nanoMIPs | Molecularly-imprinted polymer nanoparticles |
| Nav | Navitoclax |
| NF | Nuclear factor |
| NHEJ | Non-homologous end joining |
| NPs | Nanoparticles |
| OIS | Oncogene-induced senescence |
| p16 | Cyclin-dependent kinase inhibitor 2A |
| p21 | Cyclin-dependent kinase inhibitor 1A |
| p53 | Tumor suppressor protein p53 |
| PEG | Polyethylene glycol |
| PI3K | Phosphatidylinositol-3-kinase |
| Rapa | Rapamycin |
| Rb | Retinoblastoma |
| RNA | Ribonucleic acid |
| ROS | Reactive oxygen species |
| SAHF | Senescence-associated heterochromatin foci |
| SASP | Senescence-associated secretory phenotype |
| SA-β-gal | Senescence associated β-galactosidase |
| SCAPs | Senescent cell anti-apoptotic pathways |
| SCs | Senescent cells |
| SIPS | Stress-induced premature senescence |
| siRNA | Small interfering RNA |
| STAT | Signal transducer and activator of transcription |
| uPAR | Urokinase-type plasminogen activator receptor |
References
- Rodier, F.; Campisi, J. Four faces of cellular senescence. J. Cell Biol. 2011, 192, 547–556. [Google Scholar] [CrossRef] [PubMed]
- Herranz, N.; Gil, J. Mechanisms and functions of cellular senescence. J. Clin. Investig. 2018, 128, 1238–1246. [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]
- Shay, J.W.; Wright, W.E. Telomeres and telomerase: Three decades of progress. Nat. Rev. Genet. 2019, 20, 299–309. [Google Scholar] [CrossRef]
- Bodnar, A.G. Extension of Life-Span by Introduction of Telomerase into Normal Human Cells. Science 1998, 279, 349–352. [Google Scholar] [CrossRef]
- Dimri, G.P.; Lee, X.; Basile, G.; Acosta, M.; Scott, G.; Roskelley, C.; Medrano, E.E.; Linskens, M.; Rubelj, I.; Pereira-Smith, O. 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]
- Chen, Q.; Fischer, A.; Reagan, J.D.; Yan, L.J.; Ames, B.N. Oxidative DNA damage and senescence of human diploid fibroblast cells. Proc. Natl. Acad. Sci. USA 1995, 92, 4337–4341. [Google Scholar] [CrossRef]
- Kuilman, T.; Michaloglou, C.; Mooi, W.J.; Peeper, D.S. The essence of senescence. Genes Dev. 2010, 24, 2463–2479. [Google Scholar] [CrossRef]
- Toussaint, O.; Medrano, E.E.; von Zglinicki, T. Cellular and molecular mechanisms of stress-induced premature senescence (SIPS) of human diploid fibroblasts and melanocytes. Exp. Gerontol. 2000, 35, 927–945. [Google Scholar] [CrossRef]
- Serrano, M.; Lin, A.W.; McCurrach, M.E.; Beach, D.; Lowe, S.W. Oncogenic ras Provokes Premature Cell Senescence Associated with Accumulation of p53 and p16INK4a. Cell 1997, 88, 593–602. [Google Scholar] [CrossRef]
- Collado, M.; Serrano, M. Senescence in tumours: Evidence from mice and humans. Nat. Rev. Cancer 2010, 10, 51–57. [Google Scholar] [CrossRef] [PubMed]
- Storer, M.; Mas, A.; Robert-Moreno, A.; Pecoraro, M.; Ortells, M.C.; Di Giacomo, V.; Yosef, R.; Pilpel, N.; Krizhanovsky, V.; Sharpe, J.; et al. Senescence Is a Developmental Mechanism that Contributes to Embryonic Growth and Patterning. Cell 2013, 155, 1119–1130. [Google Scholar] [CrossRef] [PubMed]
- Muñoz-Espín, D.; Serrano, M. Cellular senescence: From physiology to pathology. Nat. Rev. Mol. Cell Biol. 2014, 15, 482–496. [Google Scholar] [CrossRef] [PubMed]
- Tchkonia, T.; Morbeck, D.E.; Von Zglinicki, T.; Van Deursen, J.; Lustgarten, J.; Scrable, H.; Khosla, S.; Jensen, M.D.; Kirkland, J.L. Fat tissue, aging, and cellular senescence: Fat tissue and aging. Aging Cell 2010, 9, 667–684. [Google Scholar] [CrossRef] [PubMed]
- Naylor, R.M.; Baker, D.J.; van Deursen, J.M. Senescent cells: A novel therapeutic target for aging and age-related diseases. Clin. Pharmacol. Ther. 2013, 93, 105–116. [Google Scholar] [CrossRef] [PubMed]
- van Deursen, J.M. The role of senescent cells in ageing. Nature 2014, 509, 439–446. [Google Scholar] [CrossRef] [PubMed]
- He, S.; Sharpless, N.E. Senescence in Health and Disease. Cell 2017, 169, 1000–1011. [Google Scholar] [CrossRef] [PubMed]
- Kritsilis, M.; Rizou, V.S.; Koutsoudaki, P.N.; Evangelou, K.; Gorgoulis, V.G.; Papadopoulos, D. Ageing, Cellular Senescence and Neurodegenerative Disease. Int. J. Mol. Sci. 2018, 19, 2937. [Google Scholar] [CrossRef] [PubMed]
- 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] [PubMed]
- Wei, W.; Ji, S. Cellular senescence: Molecular mechanisms and pathogenicity. J. Cell. Physiol. 2018, 233, 9121–9135. [Google Scholar] [CrossRef] [PubMed]
- Sikora, E.; Bielak-Zmijewska, A.; Mosieniak, G. Targeting normal and cancer senescent cells as a strategy of senotherapy. Ageing Res. Rev. 2019, 55, 100941. [Google Scholar] [CrossRef] [PubMed]
- Terzi, M.Y.; Izmirli, M.; Gogebakan, B. The cell fate: Senescence or quiescence. Mol. Biol. Rep. 2016, 43, 1213–1220. [Google Scholar] [CrossRef] [PubMed]
- Sharpless, N.E.; Sherr, C.J. Forging a signature of in vivo senescence. Nat. Rev. Cancer 2015, 15, 397–408. [Google Scholar] [CrossRef] [PubMed]
- 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] [PubMed]
- von Kobbe, C. Targeting senescent cells: Approaches, opportunities, challenges. Aging 2019, 11, 12844–12861. [Google Scholar] [CrossRef] [PubMed]
- Coppé, J.-P.; Patil, C.K.; Rodier, F.; Sun, Y.; Muñoz, 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]
- Kuilman, T.; Peeper, D.S. Senescence-messaging secretome: SMS-ing cellular stress. Nat. Rev. Cancer 2009, 9, 81–94. [Google Scholar] [CrossRef]
- Rodier, F.; Coppé, J.-P.; Patil, C.K.; Hoeijmakers, W.A.M.; Muñoz, D.P.; Raza, S.R.; Freund, A.; Campeau, E.; Davalos, A.R.; Campisi, J. Persistent DNA damage signalling triggers senescence-associated inflammatory cytokine secretion. Nat. Cell Biol. 2009, 11, 973–979. [Google Scholar] [CrossRef]
- Coppé, 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]
- Kirkland, J.L.; Tchkonia, T. Cellular Senescence: A Translational Perspective. EBioMedicine 2017, 21, 21–28. [Google Scholar] [CrossRef]
- Panda, A.C.; Abdelmohsen, K.; Gorospe, M. SASP regulation by noncoding RNA. Mech. Ageing Dev. 2017, 168, 37–43. [Google Scholar] [CrossRef] [PubMed]
- 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] [PubMed]
- Yun, M.H. Cellular senescence in tissue repair: Every cloud has a silver lining. Int. J. Dev. Biol. 2018, 62, 591–604. [Google Scholar] [CrossRef] [PubMed]
- Acosta, J.C.; O’Loghlen, A.; Banito, A.; Guijarro, M.V.; Augert, A.; Raguz, S.; Fumagalli, M.; Da Costa, M.; Brown, C.; Popov, N.; et al. Chemokine Signaling via the CXCR2 Receptor Reinforces Senescence. Cell 2008, 133, 1006–1018. [Google Scholar] [CrossRef]
- Krtolica, A.; Parrinello, S.; Lockett, S.; Desprez, P.-Y.; Campisi, J. Senescent fibroblasts promote epithelial cell growth and tumorigenesis: A link between cancer and aging. Proc. Natl. Acad. Sci. USA 2001, 98, 12072. [Google Scholar] [CrossRef]
- Vizioli, M.G.; Adams, P.D. Senescence Can Be BETter without the SASP? Cancer Discov. 2016, 6, 576–578. [Google Scholar] [CrossRef]
- Soto-Gamez, A.; Demaria, M. Therapeutic interventions for aging: The case of cellular senescence. Drug Discov. Today 2017, 22, 786–795. [Google Scholar] [CrossRef]
- Sakoda, K.; Yamamoto, M.; Negishi, Y.; Liao, J.K.; Node, K.; Izumi, Y. Simvastatin Decreases IL-6 and IL-8 Production in Epithelial Cells. J. Dent. Res. 2006, 85, 520–523. [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]
- Hoare, M.; Ito, Y.; Kang, T.-W.; Weekes, M.P.; Matheson, N.J.; Patten, D.A.; Shetty, S.; Parry, A.J.; Menon, S.; Salama, R.; et al. NOTCH1 mediates a switch between two distinct secretomes during senescence. Nat. Cell Biol. 2016, 18, 979–992. [Google Scholar] [CrossRef]
- Nelson, G.; Wordsworth, J.; Wang, C.; Jurk, D.; Lawless, C.; Martin-Ruiz, C.; von Zglinicki, T. A senescent cell bystander effect: Senescence-induced senescence. Aging Cell 2012, 11, 345–349. [Google Scholar] [CrossRef] [PubMed]
- Lehmann, B.D.; Paine, M.S.; Brooks, A.M.; McCubrey, J.A.; Renegar, R.H.; Wang, R.; Terrian, D.M. Senescence-Associated Exosome Release from Human Prostate Cancer Cells. Cancer Res. 2008, 68, 7864–7871. [Google Scholar] [CrossRef] [PubMed]
- Fuhrmann-Stroissnigg, H.; Niedernhofer, L.J.; Robbins, P.D. Hsp90 inhibitors as senolytic drugs to extend healthy aging. Cell Cycle Georget. Tex 2018, 17, 1048–1055. [Google Scholar] [CrossRef] [PubMed]
- Sajja, H.; East, M.; Mao, H.; Wang, Y.; Nie, S.; Yang, L. Development of Multifunctional Nanoparticles for Targeted Drug Delivery and Noninvasive Imaging of Therapeutic Effect. Curr. Drug Discov. Technol. 2009, 6, 43–51. [Google Scholar] [CrossRef]
- Obeid, M.A.; Al Qaraghuli, M.M.; Alsaadi, M.; Alzahrani, A.R.; Niwasabutra, K.; Ferro, V.A. Delivering natural products and biotherapeutics to improve drug efficacy. Ther. Deliv. 2017, 8, 947–956. [Google Scholar] [CrossRef]
- Patra, J.K.; Das, G.; Fraceto, L.F.; Campos, E.V.R.; Rodriguez-Torres, M.P.; Acosta-Torres, L.S.; Diaz-Torres, L.A.; Grillo, R.; Swamy, M.K.; Sharma, S.; et al. Nano based drug delivery systems: Recent developments and future prospects. J. Nanobiotechnol. 2018, 16, 71. [Google Scholar] [CrossRef]
- Chenthamara, D.; Subramaniam, S.; Ramakrishnan, S.G.; Krishnaswamy, S.; Essa, M.M.; Lin, F.-H.; Qoronfleh, M.W. Therapeutic efficacy of nanoparticles and routes of administration. Biomater. Res. 2019, 23, 20. [Google Scholar] [CrossRef]
- Farokhzad, O.; Langer, R. Nanomedicine: Developing smarter therapeutic and diagnostic modalities. Adv. Drug Deliv. Rev. 2006, 58, 1456–1459. [Google Scholar] [CrossRef]
- Nie, S.; Xing, Y.; Kim, G.J.; Simons, J.W. Nanotechnology Applications in Cancer. Annu. Rev. Biomed. Eng. 2007, 9, 257–288. [Google Scholar] [CrossRef]
- Seigneuric, R.; Markey, L.; Nuyten, S.A.D.; Dubernet, C.; Evelo, T.A.C.; Finot, E.; Garrido, C. From Nanotechnology to Nanomedicine: Applications to Cancer Research. Curr. Mol. Med. 2010, 10, 640–652. [Google Scholar] [CrossRef]
- Kolahalam, L.A.; Kasi Viswanath, I.V.; Diwakar, B.S.; Govindh, B.; Reddy, V.; Murthy, Y.L.N. Review on nanomaterials: Synthesis and applications. Mater. Today Proc. 2019, 18, 2182–2190. [Google Scholar] [CrossRef]
- Ventola, C.L. The nanomedicine revolution: Part 1: Emerging concepts. P T Peer-Rev. J. Formul. Manag. 2012, 37, 512–525. [Google Scholar]
- Salatin, S.; Yari Khosroushahi, A. Overviews on the cellular uptake mechanism of polysaccharide colloidal nanoparticles. J. Cell. Mol. Med. 2017, 21, 1668–1686. [Google Scholar] [CrossRef] [PubMed]
- Kargozar, S.; Mozafari, M. Nanotechnology and Nanomedicine: Start small, think big. Mater. Today Proc. 2018, 5, 15492–15500. [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. [Google Scholar] [CrossRef]
- Pelaz, B.; del Pino, P.; Maffre, P.; Hartmann, R.; Gallego, M.; Rivera-Fernández, S.; de la Fuente, J.M.; Nienhaus, G.U.; Parak, W.J. Surface Functionalization of Nanoparticles with Polyethylene Glycol: Effects on Protein Adsorption and Cellular Uptake. ACS Nano 2015, 9, 6996–7008. [Google Scholar] [CrossRef]
- Schöttler, S.; Becker, G.; Winzen, S.; Steinbach, T.; Mohr, K.; Landfester, K.; Mailänder, V.; Wurm, F.R. Protein adsorption is required for stealth effect of poly(ethylene glycol)- and poly(phosphoester)-coated nanocarriers. Nat. Nanotechnol. 2016, 11, 372–377. [Google Scholar] [CrossRef]
- Kolhar, P.; Anselmo, A.C.; Gupta, V.; Pant, K.; Prabhakarpandian, B.; Ruoslahti, E.; Mitragotri, S. Using shape effects to target antibody-coated nanoparticles to lung and brain endothelium. Proc. Natl. Acad. Sci. USA 2013, 110, 10753–10758. [Google Scholar] [CrossRef]
- Gao, H.; Yang, Z.; Zhang, S.; Cao, S.; Shen, S.; Pang, Z.; Jiang, X. Ligand modified nanoparticles increases cell uptake, alters endocytosis and elevates glioma distribution and internalization. Sci. Rep. 2013, 3, 2534. [Google Scholar] [CrossRef]
- Gao, W.; Zhang, L. Coating nanoparticles with cell membranes for targeted drug delivery. J. Drug Target. 2015, 23, 619–626. [Google Scholar] [CrossRef]
- Suk, J.S.; Xu, Q.; Kim, N.; Hanes, J.; Ensign, L.M. PEGylation as a strategy for improving nanoparticle-based drug and gene delivery. Adv. Drug Deliv. Rev. 2016, 99, 28–51. [Google Scholar] [CrossRef] [PubMed]
- Zavaleta, C.; Ho, D.; Chung, E.J. Theranostic Nanoparticles for Tracking and Monitoring Disease State. SLAS Technol. Transl. Life Sci. Innov. 2018, 23, 281–293. [Google Scholar] [CrossRef] [PubMed]
- Ramanathan, S.; Archunan, G.; Sivakumar, M.; Tamil Selvan, S.; Fred, A.L.; Kumar, S.; Gulyás, B.; Padmanabhan, P. Theranostic applications of nanoparticles in neurodegenerative disorders. Int. J. Nanomed. 2018, 13, 5561–5576. [Google Scholar] [CrossRef] [PubMed]
- Haley, B.; Frenkel, E. Nanoparticles for drug delivery in cancer treatment. Urol. Oncol. Semin. Orig. Investig. 2008, 26, 57–64. [Google Scholar] [CrossRef] [PubMed]
- Chen, F.; Ehlerding, E.B.; Cai, W. Theranostic nanoparticles. J. Nucl. Med. 2014, 55, 1919–1922. [Google Scholar] [CrossRef] [PubMed]
- Chen, G.; Roy, I.; Yang, C.; Prasad, P.N. Nanochemistry and Nanomedicine for Nanoparticle-based Diagnostics and Therapy. Chem. Rev. 2016, 116, 2826–2885. [Google Scholar] [CrossRef] [PubMed]
- Riccardi, C.; Fàbrega, C.; Grijalvo, S.; Vitiello, G.; D’Errico, G.; Eritja, R.; Montesarchio, D. AS1411-decorated niosomes as effective nanocarriers for Ru(iii)-based drugs in anticancer strategies. J. Mater. Chem. B 2018, 6, 5368–5384. [Google Scholar] [CrossRef] [PubMed]
- Squillaro, T.; Cimini, A.; Peluso, G.; Giordano, A.; Melone, M.A.B. Nano-delivery systems for encapsulation of dietary polyphenols: An experimental approach for neurodegenerative diseases and brain tumors. Biochem. Pharmacol. 2018, 154, 303–317. [Google Scholar] [CrossRef]
- Di Martino, P.; Censi, R.; Gigliobianco, M.R.; Zerrillo, L.; Magnoni, F.; Agas, D.; Quaglia, W.; Lupidi, G. Nano-medicine Improving the Bioavailability of Small Molecules for the Prevention of Neurodegenerative Diseases. Curr. Pharm. Des. 2017, 23, 1897–1908. [Google Scholar] [CrossRef]
- McClements, D.J.; Xiao, H. Designing food structure and composition to enhance nutraceutical bioactivity to support cancer inhibition. Semin. Cancer Biol. 2017, 46, 215–226. [Google Scholar] [CrossRef]
- Squillaro, T.; Peluso, G.; Melone, M.A.B. Nanotechnology-Based Polyphenol Delivery: A Novel Therapeutic Strategy for the Treatment of Age-Related Neurodegenerative Disorder. Austin Aging Res. 2017, 1, 1004. [Google Scholar]
- Li, C.; Zhang, J.; Zu, Y.J.; Nie, S.F.; Cao, J.; Wang, Q.; Nie, S.P.; Deng, Z.Y.; Xie, M.Y.; Wang, S. Biocompatible and biodegradable nanoparticles for enhancement of anti-cancer activities of phytochemicals. Chin. J. Nat. Med. 2015, 13, 641–652. [Google Scholar] [CrossRef]
- Thomas, O.S.; Weber, W. Overcoming Physiological Barriers to Nanoparticle Delivery-Are We There Yet? Front. Bioeng. Biotechnol. 2019, 7, 415. [Google Scholar] [CrossRef] [PubMed]
- Clark, A.J.; Davis, M.E. Increased brain uptake of targeted nanoparticles by adding an acid-cleavable linkage between transferrin and the nanoparticle core. Proc. Natl. Acad. Sci. USA 2015, 112, 12486–12491. [Google Scholar] [CrossRef] [PubMed]
- Wang, W.; Sun, C.; Mao, L.; Ma, P.; Liu, F.; Yang, J.; Gao, Y. The biological activities, chemical stability, metabolism and delivery systems of quercetin: A review. Trends Food Sci. Technol. 2016, 56, 21–38. [Google Scholar] [CrossRef]
- Abdelwahab, S.I.; Sheikh, B.Y.; Taha, M.M.E.; How, C.W.; Abdullah, R.; Yagoub, U.; El-Sunousi, R.; Eid, E.E.M. Thymoquinone-loaded nanostructured lipid carriers: Preparation, gastroprotection, in vitro toxicity, and pharmacokinetic properties after extravascular administration. Int. J. Nanomed. 2013, 8, 2163–2172. [Google Scholar] [CrossRef]
- Sharma, M.; Sharma, R.; Jain, D.K. Nanotechnology Based Approaches for Enhancing Oral Bioavailability of Poorly Water Soluble Antihypertensive Drugs. Scientifica 2016, 2016, 8525679. [Google Scholar] [CrossRef]
- Caster, J.M.; Patel, A.N.; Zhang, T.; Wang, A. Investigational nanomedicines in 2016: A review of nanotherapeutics currently undergoing clinical trials: Investigational nanomedicines in 2016. Wiley Interdiscip. Rev. Nanomed. Nanobiotechnol. 2017, 9, e1416. [Google Scholar] [CrossRef]
- Mura, S.; Nicolas, J.; Couvreur, P. Stimuli-responsive nanocarriers for drug delivery. Nat. Mater. 2013, 12, 991–1003. [Google Scholar] [CrossRef]
- Liu, J.; Huang, Y.; Kumar, A.; Tan, A.; Jin, S.; Mozhi, A.; Liang, X.-J. pH-Sensitive nano-systems for drug delivery in cancer therapy. Biotechnol. Adv. 2014, 32, 693–710. [Google Scholar] [CrossRef]
- Wang, X.; Teng, Z.; Wang, H.; Wang, C.; Liu, Y.; Tang, Y.; Wu, J.; Sun, J.; Wang, H.; Wang, J.; et al. Increasing the cytotoxicity of doxorubicin in breast cancer MCF-7 cells with multidrug resistance using a mesoporous silica nanoparticle drug delivery system. Int. J. Clin. Exp. Pathol. 2014, 7, 1337–1347. [Google Scholar] [PubMed]
- Hamidu, A.; Mokrish, A.; Mansor, R.; Razak, I.S.A.; Danmaigoro, A.; Jaji, A.Z.; Bakar, Z.A. Modified methods of nanoparticles synthesis in pH-sensitive nano-carriers production for doxorubicin delivery on MCF-7 breast cancer cell line. Int. J. Nanomed. 2019, 14, 3615–3627. [Google Scholar] [CrossRef]
- Muñoz-Espín, D. Nanocarriers targeting senescent cells. Transl. Med. Aging 2019, 3, 1–5. [Google Scholar] [CrossRef]
- Lee, B.Y.; Han, J.A.; Im, J.S.; Morrone, A.; Johung, K.; Goodwin, E.C.; Kleijer, W.J.; DiMaio, D.; Hwang, E.S. Senescence-associated β-galactosidase is lysosomal β-galactosidase. Aging Cell 2006, 5, 187–195. [Google Scholar] [CrossRef]
- Agostini, A.; Mondragón, L.; Bernardos, A.; Martínez-Máñez, R.; Marcos, M.D.; Sancenón, F.; Soto, J.; Costero, A.; Manguan-García, C.; Perona, R.; et al. Targeted Cargo Delivery in Senescent Cells Using Capped Mesoporous Silica Nanoparticles. Angew. Chem. Int. Ed. 2012, 51, 10556–10560. [Google Scholar] [CrossRef]
- Ovadya, Y.; Krizhanovsky, V. Strategies targeting cellular senescence. J. Clin. Investig. 2018, 128, 1247–1254. [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 p16(Ink4a)-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]
- Kirkland, J.L.; Tchkonia, T. Clinical strategies and animal models for developing senolytic agents. Exp. Gerontol. 2015, 68, 19–25. [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]
- 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] [PubMed]
- González-Gualda, E.; Pàez-Ribes, M.; Lozano-Torres, B.; Macias, D.; Wilson, J.R.; 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, e13142. [Google Scholar] [CrossRef] [PubMed]
- Justice, J.N.; Nambiar, A.M.; Tchkonia, T.; LeBrasseur, N.K.; Pascual, R.; Hashmi, S.K.; Prata, L.; Masternak, M.M.; Kritchevsky, S.B.; Musi, N.; et al. Senolytics in idiopathic pulmonary fibrosis: Results from a first-in-human, open-label, pilot study. EBioMedicine 2019, 40, 554–563. [Google Scholar] [CrossRef] [PubMed]
- 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] [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]
- 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]
- 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]
- 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]
- 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]
- Samaraweera, L.; Adomako, A.; Rodriguez-Gabin, A.; McDaid, H.M. A Novel Indication for Panobinostat as a Senolytic Drug in NSCLC and HNSCC. Sci. Rep. 2017, 7, 1900. [Google Scholar] [CrossRef] [PubMed]
- 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] [PubMed]
- Kirkland, J.L.; Tchkonia, T.; Zhu, Y.; Niedernhofer, L.J.; Robbins, P.D. The Clinical Potential of Senolytic Drugs. J. Am. Geriatr. Soc. 2017, 65, 2297–2301. [Google Scholar] [CrossRef] [PubMed]
- 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] [PubMed]
- Wakita, M.; Takahashi, A.; Sano, O.; Loo, T.M.; Imai, Y.; Narukawa, M.; Iwata, H.; Matsudaira, T.; Kawamoto, S.; Ohtani, N.; et al. A BET family protein degrader provokes senolysis by targeting NHEJ and autophagy in senescent cells. Nat. Commun. 2020, 11, 1935. [Google Scholar] [CrossRef] [PubMed]
- Mariño, G.; Niso-Santano, M.; Baehrecke, E.H.; Kroemer, G. Self-consumption: The interplay of autophagy and apoptosis. Nat. Rev. Mol. Cell Biol. 2014, 15, 81–94. [Google Scholar] [CrossRef]
- Thapa, R.K.; Nguyen, H.T.; Jeong, J.-H.; Kim, J.R.; Choi, H.-G.; Yong, C.S.; Kim, J.O. Progressive slowdown/prevention of cellular senescence by CD9-targeted delivery of rapamycin using lactose-wrapped calcium carbonate nanoparticles. Sci. Rep. 2017, 7, 43299. [Google Scholar] [CrossRef]
- Ke, S.; Lai, Y.; Zhou, T.; Li, L.; Wang, Y.; Ren, L.; Ye, S. Molybdenum Disulfide Nanoparticles Resist Oxidative Stress-Mediated Impairment of Autophagic Flux and Mitigate Endothelial Cell Senescence and Angiogenic Dysfunctions. ACS Biomater. Sci. Eng. 2018, 4, 663–674. [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]
- Ekpenyong-Akiba, A.E.; Canfarotta, F.; Abd, H.B.; Poblocka, M.; Casulleras, M.; Castilla-Vallmanya, L.; Kocsis-Fodor, G.; Kelly, M.E.; Janus, J.; Althubiti, M.; et al. Detecting and targeting senescent cells using molecularly imprinted nanoparticles. Nanoscale Horiz. 2019, 4, 757–768. [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] [PubMed]
- Riessland, M.; Kolisnyk, B.; Kim, T.W.; Cheng, J.; Ni, J.; Pearson, J.A.; Park, E.J.; Dam, K.; Acehan, D.; Ramos-Espiritu, L.S.; et al. Loss of SATB1 Induces p21-Dependent Cellular Senescence in Post-mitotic Dopaminergic Neurons. Cell Stem. Cell 2019, 25, 514–530.e8. [Google Scholar] [CrossRef] [PubMed]
- Salmonowicz, H.; Passos, J.F. Detecting senescence: A new method for an old pigment. Aging Cell 2017, 16, 432–434. [Google Scholar] [CrossRef] [PubMed]
- Muñoz-Espin, D.; Demaria, M. Senolytics in Disease, Ageing and Longevity, 1st ed.; Springer Nature Switzerland: Cham, Switzerland, 2020; pp. 163–180. [Google Scholar]
- Xu, T.; Cai, Y.; Zhong, X.; Zhang, L.; Zheng, D.; Gao, Z.; Pan, X.; Wang, F.; Chen, M.; Yang, Z. β-Galactosidase instructed supramolecular hydrogelation for selective identification and removal of senescent cells. Chem. Commun. 2019, 55, 7175–7178. [Google Scholar] [CrossRef]
- Hildebrand, D.; Lehle, S.; Borst, A.; Haferkamp, S.; Essmann, F.; Schulze-Osthoff, K. α-Fucosidase as a novel convenient biomarker for cellular senescence. Cell Cycle 2013, 12, 1922–1927. [Google Scholar] [CrossRef]
- Lozano-Torres, B.; Blandez, J.F.; Galiana, I.; García-Fernández, A.; Alfonso, M.; Marcos, M.D.; Orzáez, M.; Sancenón, F.; Martínez-Máñez, R. Real-Time In Vivo Detection of Cellular Senescence through the Controlled Release of the NIR Fluorescent Dye Nile Blue. Angew. Chem. Int. Ed. 2020, 59, 15152–15156. [Google Scholar] [CrossRef]
- Nguyen, H.T.; Thapa, R.K.; Shin, B.S.; Jeong, J.-H.; Kim, J.-R.; Yong, C.S.; Kim, J.O. CD9 monoclonal antibody-conjugated PEGylated liposomes for targeted delivery of rapamycin in the treatment of cellular senescence. Nanotechnology 2017, 28, 095101. [Google Scholar] [CrossRef]
- Nagesh, P.K.B.; Chowdhury, P.; Hatami, E.; Kumari, S.; Kashyap, V.K.; Tripathi, M.K.; Wagh, S.; Meibohm, B.; Chauhan, S.C.; Jaggi, M.; et al. Cross-Linked Polyphenol-Based Drug Nano-Self-Assemblies Engineered to Blockade Prostate Cancer Senescence. ACS Appl. Mater. Interfaces 2019, 11, 38537–38554. [Google Scholar] [CrossRef]
- Lewinska, A.; Adamczyk-Grochala, J.; Bloniarz, D.; Olszowka, J.; Kulpa-Greszta, M.; Litwinienko, G.; Tomaszewska, A.; Wnuk, M.; Pazik, R. AMPK-mediated senolytic and senostatic activity of quercetin surface-functionalized Fe3O4 nanoparticles during oxidant-induced senescence in human fibroblasts. Redox Biol. 2020, 28, 101337. [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]




| Nanomaterial | Biologically- Active Component | Concentration | Senescence Model | In Vitro/In Vivo Model | Senolytic Effects and Mechanism | Ref. |
|---|---|---|---|---|---|---|
| LR-CD9mAb CD9 monoclonal antibody conjugated to PEGylated liposomes | Rapa | 25 nM | Human dermal fibroblasts (HDF), doxorubicin-induced senescence | In vitro | Anti-senescence activity (increased proliferative potential, decreased β-galactosidase activity and p53/p21 expression, and increased cell migration) | [118] |
| CD9-Lac/CaCO3/Rapa NPs CD9 monoclonal antibody-conjugated lactose-wrapped calcium carbonate nanoparticles loaded with rapamycin | Rapa | 0.2 mg of rapamycin per mg of CaCO3 NPs | Human dermal fibroblasts (HDF), replicative and doxorubicin-induced senescence | In vitro | Anti-senescence activity (decreased β-galactosidase activity and p53/p21/CD9/cyclin D1 expression, increased cell proliferation and cell migration ability, decreased population doubling time, and prevention of G1 cell cycle arrest) | [107] |
| MoS2 NPs molybdenum disulfide mesoporous silica nanoparticles | - | 50 μg/mL | Human aortic endothelial cells (HAECs), stress-induced premature senescence | In vitro | Anti-senescence activity (decreased γ−H2AX phosphorylation, repressed upregulation of p16, p21 and p53, activation of autophagy, improved autophagic flux, and prevention of lysosomal and mitochondrial dysfunction) | [108] |
| GalNP(dox) 6-mer galacto-oligosaccharide encapsulated doxorubicin | Dox | 1 mg/kg | Mouse, bleomycin-induced lung fibrosis | In vivo | Anti-senescence activity (improved lung function) | [109] |
| GalNP(nav) 6-mer galacto-oligosaccharide encapsulated navitoclax | Nav | 0.06 mg/mL | Melanoma (SK-MEL-103), palbociclib-induced senescence | In vitro | Senolytic activity (apoptosis of senescent cells) | |
| GalNP(dox) 6-mer galacto-oligosaccharide encapsulated doxorubicin | Dox | 1 mg/kg | Mouse-bearing SK-MEL-103 tumor xenografts, palbociclib-induced tumor senescence | In vivo | Clearance of senescent cells and induced regression of tumor xenografts | |
| GalNP(nav) 6-mer galacto-oligosaccharide encapsulated navitoclax | Nav | |||||
| DSAs Docetaxel-tannic acid self-assemblies (DSAs)-based nanoparticles | Docetaxel | 2.5-5 nM | Prostate cancer cells (C4-2 and PC- 3) | In vitro | Senolytic activity (inhibition of senescence-related TGFβR1, FOXO1, and p21 proteins and activation of apoptosis) | [119] |
| 30 mg/kg | Mouse-bearing PC-3 tumor xenografts | In vivo | Clearance of senescent cells (induced regression of tumor xenografts by blockade of TGFβR1/p21-mediated senescence signaling and activation of apoptosis) | |||
| NanoMIPs molecularly-imprinted nanoparticles | Dasatinib | 10 μM dasatinib-conjugated B2M nanoMIPs | Bladder cancer cells with a tetracycline (tet)-regulatable p16 expression systems (EJp16) | In vitro | Senolytic activity (decreased number of senescent cancer cells) | [110] |
| MNPQ quercetin surface-functionalized Fe3O4 nanoparticles | Quercetin | 5 μg/mL | Human foreskin fibroblasts (BJ), hydrogen peroxide-induced senescence | In vitro | Senolytic and senostatic activity (AMPK activation, induction of non-apoptotic cell death, and inhibition of SASP components, namely IL-6 and IFN-β) | [120] |
| GalNP(nav) | Nav | 40 mg GalNP (Nav)/kg (≈2.5 mg/kg of free navitoclax) | Triple-negative breast cancer mouse model, palbociclib-induced senescence | In vivo | Senolytic activity (inhibited tumor growth, reduced metastasis, and limited systemic toxicity of navitoclax, and apoptosis of senescent cancer cells) | [121] |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2020 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 (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Adamczyk-Grochala, J.; Lewinska, A. Nano-Based Theranostic Tools for the Detection and Elimination of Senescent Cells. Cells 2020, 9, 2659. https://doi.org/10.3390/cells9122659
Adamczyk-Grochala J, Lewinska A. Nano-Based Theranostic Tools for the Detection and Elimination of Senescent Cells. Cells. 2020; 9(12):2659. https://doi.org/10.3390/cells9122659
Chicago/Turabian StyleAdamczyk-Grochala, Jagoda, and Anna Lewinska. 2020. "Nano-Based Theranostic Tools for the Detection and Elimination of Senescent Cells" Cells 9, no. 12: 2659. https://doi.org/10.3390/cells9122659
APA StyleAdamczyk-Grochala, J., & Lewinska, A. (2020). Nano-Based Theranostic Tools for the Detection and Elimination of Senescent Cells. Cells, 9(12), 2659. https://doi.org/10.3390/cells9122659

