P16INK4A—More Than a Senescence Marker
Abstract
:1. Introduction
2. The P16 Gene
3. P16 Function and Regulation of Expression
4. Role of p16 in Different Tissues and Organs: Cancer, Physiology, and Pathophysiology
4.1. In the Skin
4.2. In the Bones
4.3. In the Lungs
4.4. In the Brain
4.5. In the Heart
4.6. In the Kidney
4.7. In the Liver
Process | Model and/or Tissue | Potential Role/Function | References |
---|---|---|---|
Intervertebral disc damage | Mouse and human intervertebral disc tissues | P16 is a potential therapeutic target for intervertebral disc damage relief. | [48] |
Wound healing | P16-3MR model (fibroblasts, endothelial cells, and keratinocytes) | Accelerate wound closure and re-epithelialization | [43] |
Tumorigenesis and tumor suppression | Skin, bone, lung, liver, and brain cancer patients tissues and immunohistopathology | Implicated in tumor development, progression, and metastasis Predictive and prognostic marker Therapeutic target Increases chemosensitivity | [39,40,41,42,72,105,106,107,108,111,112,134,135,136,150,151,152,153,168,169,170,205,206] |
Stem cell self-renewal and differentiation | Skin, lung, bone, brain, and heart stem cells | Balanced expression of p16 is a prerequisite for stem cells proliferation and differentiation. Therapeutic approach for maintenance of regenerative capacity | [46,92,101,115,116,117,118,119,138,149,155,156,179] |
Cellular senescence | Primary mouse fibroblasts and melanocytes | Target for oncogene-induced senescence bypass and aging | [37,38] |
Bone homeostasis | P16-3MR and p16-INK-ATTAC mouse model | Maintenance of bone mass Orchestration of osteoblast and osteoclast function | [123,132,133] |
Bone fracture healing | Geriatric Mouse model (p16-/- and WT) | P16-deletion stimulated osteoblastogenesis and vascularization and accelerated bone fracture healing | [138] |
Muscle injury | Acute muscle injury (AIM) mouse model | Tissue regeneration | [45] |
Osteoporosis | Ovariectomized p16-/- and WT mice | Potential therapeutic target to prevent estrogen-induced osteoporosis | [124,125,126,127,137] |
COPD | Lung alveolar and lung epithelial cells in mice and human | Implicated in COPD severity Potential therapeutic target | [140,141,142,149] |
Cervical SCC and pulmonary SCC | Human cancer patients | Discriminating biomarker | [150] |
Oxidative stress | Fibroblasts, keratinocytes, and melanocytes | P16 regulates oxidative stress and ROS production as pRB-independent tumor suppression mechanism | [72,204] |
Mitochondrial biogenesis | Primary mouse fibroblasts, human melanocytes, A375 melanoma cells | P16 balances mitochondrial structure and function | [111] |
Alzheimer’s disease (AD) | Alzheimer’s disease patients and mouse model | Implicated in AD severity and development Therapeutic target | [159,160,161,166] |
Lung injury | P16-/- and WT mouse model (lung epithelium) | P16 protects against lungs injury | [154] |
Cardiac fibrosis | p16-CreERT2-tdTomato mouse model | P16-positive cells removal induces cardiac fibrosis | [83] |
Myocardial Infarction | Mice | Indispensable for maintenance of cardiac function and cardiac remodeling after infarction | [181] |
Glucose metabolism and homeostasis | Super-INK4A/ARF mice model | Prevented the development of glucose intolerance with aging Protective role against age-induced insulin resistance | [198] |
Liver fibrosis | INK-ATTAC mouse model | Therapeutic approach for treatment of liver fibrosis | [83,88,204] |
Fat metabolism | Mouse model and primary hepatocytes | Regulate fasting-induced fatty acid oxidation and lipid droplet accumulation in the liver | [201] |
Development | Young mice brain, heart, kidney, and liver | Dynamic p16 expression detected in embryonic mice organs, reflecting a potential role in embryonic development | [120] |
5. The Role of p16 in Development
6. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Conflicts of Interest
References
- Stone, S.; Jiang, P.; Dayananth, P.; Tavtigian, S.V.; Katcher, H.; Parry, D.; Peters, G.; Kamb, A. Complex structure and regulation of the P16 (MTS1) locus. Cancer Res. 1995, 55, 2988–2994. [Google Scholar]
- Mao, L.; Merlo, A.; Bedi, G.; Shapiro, G.I.; Edwards, C.D.; Rollins, B.J.; Sidransky, D. A novel p16INK4A transcript. Cancer Res. 1995, 55, 2995–2997. [Google Scholar] [PubMed]
- Serrano, M.; Hannon, G.J.; Beach, D. A new regulatory motif in cell-cycle control causing specific inhibition of cyclin D/CDK4. Nature 1993, 366, 704–707. [Google Scholar] [CrossRef] [PubMed]
- Rayess, H.; Wang, M.B.; Srivatsan, E.S. Cellular senescence and tumor suppressor gene p16. Int. J. Cancer 2012, 130, 1715–1725. [Google Scholar] [CrossRef] [PubMed]
- Krishnamurthy, J.; Torrice, C.; Ramsey, M.R.; Kovalev, G.I.; Al-Regaiey, K.; Su, L.; Sharpless, N.E. Ink4a/Arf expression is a biomarker of aging. J. Clin. Investig. 2004, 114, 1299–1307. [Google Scholar] [CrossRef]
- López-Domínguez, J.A.; Rodríguez-López, S.; Ahumada-Castro, U.; Desprez, P.-Y.; Konovalenko, M.; Laberge, R.-M.; Cárdenas, C.; Villalba, J.M.; Campisi, J. Cdkn1a transcript variant 2 is a marker of aging and cellular senescence. Aging 2021, 13, 13380–13392. [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]
- 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]
- Ohtani, N.; Mann, D.J.; Hara, E. Cellular senescence: Its role in tumor suppression and aging. Cancer Sci. 2009, 100, 792–797. [Google Scholar] [CrossRef]
- Understanding the Dynamics of the Aging Process National Institute on Aging. Available online: http://www.nia.nih.gov/about/aging-strategic-directions-research/understanding-dynamics-aging (accessed on 28 July 2022).
- Tchkonia, T.; Kirkland, J.L. Aging, Cell Senescence, and Chronic Disease: Emerging Therapeutic Strategies. JAMA 2018, 320, 1319–1320. [Google Scholar] [CrossRef]
- Hou, Y.; Dan, X.; Babbar, M.; Wei, Y.; Hasselbalch, S.G.; Croteau, D.L.; Bohr, V.A. Ageing as a risk factor for neurodegenerative disease. Nat. Rev. Neurol. 2019, 15, 565–581. [Google Scholar] [CrossRef] [PubMed]
- North, B.J.; Sinclair, D.A. The Intersection Between Aging and Cardiovascular Disease. Circ. Res. 2012, 110, 1097–1108. [Google Scholar] [CrossRef] [PubMed]
- Ramly, E.; Kaafarani, H.M.A.; Velmahos, G.C. The effect of aging on pulmonary function: Implications for monitoring and support of the surgical and trauma patient. Surg. Clin. North Am. 2015, 95, 53–69. [Google Scholar] [CrossRef] [PubMed]
- Meyer, B.R. Renal function in aging. J. Am. Geriatr. Soc. 1989, 37, 791–800. [Google Scholar] [CrossRef]
- Tung, S.; Iqbal, J. Evolution, Aging, and Osteoporosis. Ann. N. Y. Acad. Sci. 2007, 1116, 499–506. [Google Scholar] [CrossRef]
- Misra, D.; Seo, P.H.; Cohen, H.J. Aging and cancer. Clin. Adv. Hematol. Oncol. 2004, 2, 457–465. [Google Scholar]
- Liguori, I.; Russo, G.; Curcio, F.; Bulli, G.; Aran, L.; Della-Morte, D.; Gargiulo, G.; Testa, G.; Cacciatore, F.; Bonaduce, D.; et al. Oxidative stress, aging, and diseases. Clin. Interv. Aging 2018, 13, 757–772. [Google Scholar] [CrossRef]
- Michaud, M.; Balardy, L.; Moulis, G.; Gaudin, C.; Peyrot, C.; Vellas, B.; Cesari, M.; Nourhashemi, F. Proinflammatory cytokines, aging, and age-related diseases. J. Am. Med. Dir. Assoc. 2013, 14, 877–882. [Google Scholar] [CrossRef]
- Burhans, W.C.; Weinberger, M. DNA replication stress, genome instability and aging. Nucleic Acids Res. 2007, 35, 7545–7556. [Google Scholar] [CrossRef]
- Best, B.P. Nuclear DNA Damage as a Direct Cause of Aging. Rejuvenation Res. 2009, 12, 199–208. [Google Scholar] [CrossRef]
- Freitas, A.A.; de Magalhães, J.P. A review and appraisal of the DNA damage theory of ageing. Mutat. Res./Rev. Mutat. Res. 2011, 728, 12–22. [Google Scholar] [CrossRef] [PubMed]
- Campisi, J.; d’Adda di Fagagna, F. Cellular senescence: When bad things happen to good cells. Nat. Rev. Mol. Cell Biol. 2007, 8, 729–740. [Google Scholar] [CrossRef] [PubMed]
- Hayflick, L. The limited in vitro lifetime of human diploid cell strains. Exp. Cell Res. 1965, 37, 614–636. [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] [PubMed]
- 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] [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, e301. [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]
- He, S.; Sharpless, N.E. Senescence in Health and Disease. Cell 2017, 169, 1000–1011. [Google Scholar] [CrossRef]
- Gorgoulis, V.; Adams, P.D.; Alimonti, A.; Bennett, D.C.; Bischof, O.; Bishop, C.; Campisi, J.; Collado, M.; Evangelou, K.; Ferbeyre, G.; et al. Cellular Senescence: Defining a Path Forward. Cell 2019, 179, 813–827. [Google Scholar] [CrossRef]
- Collins, C.J.; Sedivy, J.M. Involvement of the INK4a/Arf gene locus in senescence. Aging Cell 2003, 2, 145–150. [Google Scholar] [CrossRef]
- Takeuchi, S.; Takahashi, A.; Motoi, N.; Yoshimoto, S.; Tajima, T.; Yamakoshi, K.; Hirao, A.; Yanagi, S.; Fukami, K.; Ishikawa, Y.; et al. Intrinsic Cooperation between p16INK4a and p21Waf1/Cip1 in the Onset of Cellular Senescence and Tumor Suppression In vivo. Cancer Res. 2010, 70, 9381–9390. [Google Scholar] [CrossRef] [PubMed]
- Parry, D.; Bates, S.; Mann, D.J.; Peters, G. Lack of cyclin D-Cdk complexes in Rb-negative cells correlates with high levels of p16INK4/MTS1 tumour suppressor gene product. EMBO J. 1995, 14, 503–511. [Google Scholar] [CrossRef] [PubMed]
- Rhinn, M.; Ritschka, B.; Keyes, W.M. Cellular senescence in development, regeneration and disease. Dev. Camb. Engl. 2019, 146, dev151837. [Google Scholar] [CrossRef] [PubMed]
- Shamloo, B.; Usluer, S. p21 in Cancer Research. Cancers 2019, 11, 1178. [Google Scholar] [CrossRef] [PubMed]
- Coppé, J.-P.; Rodier, F.; Patil, C.K.; Freund, A.; Desprez, P.-Y.; Campisi, J. Tumor suppressor and aging biomarker p16(INK4a) induces cellular senescence without the associated inflammatory secretory phenotype. J. Biol. Chem. 2011, 286, 36396–36403. [Google Scholar] [CrossRef]
- Buj, R.; Leon, K.E.; Anguelov, M.A.; Aird, K.M. Suppression of p16 alleviates the senescence-associated secretory phenotype. Aging 2021, 13, 3290–3312. [Google Scholar] [CrossRef]
- Buj, R.; Chen, C.-W.; Dahl, E.S.; Leon, K.E.; Kuskovsky, R.; Maglakelidze, N.; Navaratnarajah, M.; Zhang, G.; Doan, M.T.; Jiang, H.; et al. Suppression of p16 Induces mTORC1-Mediated Nucleotide Metabolic Reprogramming. Cell Rep. 2019, 28, 1971–1980e8. [Google Scholar] [CrossRef]
- Damsky, W.; Micevic, G.; Meeth, K.; Muthusamy, V.; Curley, D.P.; Santhanakrishnan, M.; Erdelyi, I.; Platt, J.T.; Huang, L.; Theodosakis, N.; et al. mTORC1 activation blocks BrafV600E-induced growth arrest but is insufficient for melanoma formation. Cancer Cell 2015, 27, 41–56. [Google Scholar] [CrossRef]
- Dankort, D.; Filenova, E.; Collado, M.; Serrano, M.; Jones, K.; McMahon, M. A new mouse model to explore the initiation, progression, and therapy of BRAFV600E-induced lung tumors. Genes Dev. 2007, 21, 379–384. [Google Scholar] [CrossRef]
- Goel, V.K.; Ibrahim, N.; Jiang, G.; Singhal, M.; Fee, S.; Flotte, T.; Westmoreland, S.; Haluska, F.S.; Hinds, P.W.; Haluska, F.G. Melanocytic nevus-like hyperplasia and melanoma in transgenic BRAFV600E mice. Oncogene 2009, 28, 2289–2298. [Google Scholar] [CrossRef]
- Haferkamp, S.; Becker, T.M.; Scurr, L.L.; Kefford, R.F.; Rizos, H. p16INK4a-induced senescence is disabled by melanoma-associated mutations. Aging Cell 2008, 7, 733–745. [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.T.; 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] [PubMed] [Green Version]
- Natarajan, E.; Omobono, J.D.; Jones, J.C.; Rheinwald, J.G. Co-expression of p16INK4A and laminin 5 by keratinocytes: A wound-healing response coupling hypermotility with growth arrest that goes awry during epithelial neoplastic progression. J. Investig. Dermatol. Symp. Proc. 2005, 10, 72–85. [Google Scholar] [CrossRef] [PubMed]
- Chikenji, T.S.; Saito, Y.; Konari, N.; Nakano, M.; Mizue, Y.; Otani, M.; Fujimiya, M. p16INK4A-expressing mesenchymal stromal cells restore the senescence–clearance–regeneration sequence that is impaired in chronic muscle inflammation. eBioMedicine 2019, 44, 86–97. [Google Scholar] [CrossRef] [PubMed]
- D’Arcangelo, D.; Tinaburri, L.; Dellambra, E. The Role of p16INK4a Pathway in Human Epidermal Stem Cell Self-Renewal, Aging and Cancer. Int. J. Mol. Sci. 2017, 18, 1591. [Google Scholar] [CrossRef] [PubMed]
- 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]
- Che, H.; Li, J.; Li, Y.; Ma, C.; Liu, H.; Qin, J.; Dong, J.; Zhang, Z.; Xian, C.J.; Miao, D.; et al. p16 deficiency attenuates intervertebral disc degeneration by adjusting oxidative stress and nucleus pulposus cell cycle. eLife 2020, 9, e52570. [Google Scholar] [CrossRef]
- Li, Y.; Zhao, H.; Huang, X.; Tang, J.; Zhang, S.; Li, Y.; Liu, X.; He, L.; Ju, Z.; Lui, K.O.; et al. Embryonic senescent cells re-enter cell cycle and contribute to tissues after birth. Cell Res. 2018, 28, 775–778. [Google Scholar] [CrossRef]
- Hosako, H.; Francisco, L.E.; Martin, G.S.; Mirkes, P.E. The roles of p53 and p21 in normal development and hyperthermia-induced malformations. Birth Defects Res. B. Dev. Reprod. Toxicol. 2009, 86, 40–47. [Google Scholar] [CrossRef]
- 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]
- Muñoz-Espín, D.; Cañamero, M.; Maraver, A.; Gómez-López, G.; Contreras, J.; Murillo-Cuesta, S.; Rodríguez-Baeza, A.; Varela-Nieto, I.; Ruberte, J.; Collado, M.; et al. Programmed cell senescence during mammalian embryonic development. Cell 2013, 155, 1104–1118. [Google Scholar] [CrossRef] [PubMed]
- Zindy, F.; Soares, H.; Herzog, K.H.; Morgan, J.; Sherr, C.J.; Roussel, M.F. Expression of INK4 inhibitors of cyclin D-dependent kinases during mouse brain development. Cell Growth Differ. 1997, 8, 1139–1150. [Google Scholar] [PubMed]
- Zindy, F.; Quelle, D.E.; Roussel, M.F.; Sherr, C.J. Expression of the p16INK4a tumor suppressor versus other INK4 family members during mouse development and aging. Oncogene 1997, 15, 203–211. [Google Scholar] [CrossRef]
- Quelle, D.E.; Zindy, F.; Ashmun, R.A.; Sherr, C.J. Alternative reading frames of the INK4a tumor suppressor gene encode two unrelated proteins capable of inducing cell cycle arrest. Cell 1995, 83, 993–1000. [Google Scholar] [CrossRef] [PubMed]
- Serrano, M. The tumor suppressor protein p16INK4a. Exp. Cell Res. 1997, 237, 7–13. [Google Scholar] [CrossRef] [PubMed]
- Komata, T.; Kanzawa, T.; Takeuchi, H.; Germano, I.M.; Schreiber, M.; Kondo, Y.; Kondo, S. Antitumour effect of cyclin-dependent kinase inhibitors (p16(INK4A), p18(INK4C), p19(INK4D), p21(WAF1/CIP1) and p27(KIP1)) on malignant glioma cells. Br. J. Cancer 2003, 88, 1277–1280. [Google Scholar] [CrossRef] [PubMed]
- Li, J.; Poi, M.J.; Tsai, M.-D. Regulatory mechanisms of tumor suppressor P16(INK4A) and their relevance to cancer. Biochemistry 2011, 50, 5566–5582. [Google Scholar] [CrossRef]
- Cilluffo, D.; Barra, V.; Di Leonardo, A. P14ARF: The Absence that Makes the Difference. Genes 2020, 11, 824. [Google Scholar] [CrossRef]
- Liggett, W.H.; Sidransky, D. Role of the p16 tumor suppressor gene in cancer. J. Clin. Oncol. 1998, 16, 1197–1206. [Google Scholar] [CrossRef]
- Barnum, K.J.; O’Connell, M.J. Cell Cycle Regulation by Checkpoints. Methods Mol. Biol. 2014, 1170, 29–40. [Google Scholar] [CrossRef]
- Weinberg, R.A. The Cat and Mouse Games That Genes, Viruses, and Cells Play. Cell 1997, 88, 573–575. [Google Scholar] [CrossRef]
- Ahlander, J.; Bosco, G. The RB/E2F pathway and regulation of RNA processing. Biochem. Biophys. Res. Commun. 2009, 384, 280–283. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sherr, C.J.; Roberts, J.M. Living with or without cyclins and cyclin-dependent kinases. Genes Dev. 2004, 18, 2699–2711. [Google Scholar] [CrossRef] [PubMed]
- Sun, P.; Nallar, S.C.; Raha, A.; Kalakonda, S.; Velalar, C.N.; Reddy, S.P.; Kalvakolanu, D.V. GRIM-19 and p16(INK4a) synergistically regulate cell cycle progression and E2F1-responsive gene expression. J. Biol. Chem. 2010, 285, 27545–27552. [Google Scholar] [CrossRef]
- Li, Y.; Nichols, M.A.; Shay, J.W.; Xiong, Y. Transcriptional repression of the D-type cyclin-dependent kinase inhibitor p16 by the retinoblastoma susceptibility gene product pRb. Cancer Res. 1994, 54, 6078–6082. [Google Scholar]
- Sanchez-Cespedes, M.; Reed, A.L.; Buta, M.; Wu, L.; Westra, W.H.; Herman, J.G.; Yang, S.C.; Jen, J.; Sidransky, D. Inactivation of the INK4A/ARF locus frequently coexists with TP53 mutations in non-small cell lung cancer. Oncogene 1999, 18, 5843–5849. [Google Scholar] [CrossRef]
- Sparmann, A.; van Lohuizen, M. Polycomb silencers control cell fate, development and cancer. Nat. Rev. Cancer 2006, 6, 846–856. [Google Scholar] [CrossRef]
- Schwartz, Y.B.; Pirrotta, V. Polycomb silencing mechanisms and the management of genomic programmes. Nat. Rev. Genet. 2007, 8, 9–22. [Google Scholar] [CrossRef]
- Cao, R.; Zhang, Y. The functions of E(Z)/EZH2-mediated methylation of lysine 27 in histone H3. Curr. Opin. Genet. Dev. 2004, 14, 155–164. [Google Scholar] [CrossRef]
- Bracken, A.P.; Kleine-Kohlbrecher, D.; Dietrich, N.; Pasini, D.; Gargiulo, G.; Beekman, C.; Theilgaard-Mönch, K.; Minucci, S.; Porse, B.T.; Marine, J.-C.; et al. The Polycomb group proteins bind throughout the INK4A-ARF locus and are disassociated in senescent cells. Genes Dev. 2007, 21, 525–530. [Google Scholar] [CrossRef]
- Jenkins, N.C.; Liu, T.; Cassidy, P.; Leachman, S.A.; Boucher, K.M.; Goodson, A.G.; Samadashwily, G.; Grossman, D. The p16INK4A tumor suppressor regulates cellular oxidative stress. Oncogene 2011, 30, 265–274. [Google Scholar] [CrossRef] [PubMed]
- Kim, J.; Wong, P.K.Y. Oxidative stress is linked to ERK1/2-p16 signaling-mediated growth defect in ATM-deficient astrocytes. J. Biol. Chem. 2009, 284, 14396–14404. [Google Scholar] [CrossRef] [PubMed]
- Nishiwaki, E.; Turner, S.L.; Harju, S.; Miyazaki, S.; Kashiwagi, M.; Koh, J.; Serizawa, H. Regulation of CDK7-carboxyl-terminal domain kinase activity by the tumor suppressor p16(INK4A) contributes to cell cycle regulation. Mol. Cell. Biol. 2000, 20, 7726–7734. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Choi, B.Y.; Choi, H.S.; Ko, K.; Cho, Y.-Y.; Zhu, F.; Kang, B.S.; Ermakova, S.P.; Ma, W.-Y.; Bode, A.M.; Dong, Z. The tumor suppressor p16(INK4a) prevents cell transformation through inhibition of c-Jun phosphorylation and AP-1 activity. Nat. Struct. Mol. Biol. 2005, 12, 699–707. [Google Scholar] [CrossRef] [PubMed]
- Serizawa, H. Cyclin-dependent kinase inhibitor p16INK4A inhibits phosphorylation of RNA polymerase II by general transcription factor TFIIH. J. Biol. Chem. 1998, 273, 5427–5430. [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]
- Lin, A.W.; Barradas, M.; Stone, J.C.; van Aelst, L.; Serrano, M.; Lowe, S.W. Premature senescence involving p53 and p16 is activated in response to constitutive MEK/MAPK mitogenic signaling. Genes Dev. 1998, 12, 3008–3019. [Google Scholar] [CrossRef]
- Graves, B.J.; Petersen, J.M. Specificity within the ets family of transcription factors. Adv. Cancer Res. 1998, 75, 1–57. [Google Scholar] [CrossRef]
- Ohtani, N.; Zebedee, Z.; Huot, T.J.G.; Stinson, J.A.; Sugimoto, M.; Ohashi, Y.; Sharrocks, A.D.; Peters, G.; Hara, E. Opposing effects of Ets and Id proteins on p16INK4a expression during cellular senescence. Nature 2001, 409, 1067–1070. [Google Scholar] [CrossRef]
- Xiu, M.; Kim, J.; Sampson, E.; Huang, C.-Y.; Davis, R.J.; Paulson, K.E.; Yee, A.S. The Transcriptional Repressor HBP1 Is a Target of the p38 Mitogen-Activated Protein Kinase Pathway in Cell Cycle Regulation. Mol. Cell. Biol. 2003, 23, 8890–8901. [Google Scholar] [CrossRef]
- Barradas, M.; Anderton, E.; Acosta, J.C.; Li, S.; Banito, A.; Rodriguez-Niedenführ, M.; Maertens, G.; Banck, M.; Zhou, M.-M.; Walsh, M.J.; et al. Histone demethylase JMJD3 contributes to epigenetic control of INK4a/ARF by oncogenic RAS. Genes Dev. 2009, 23, 1177–1182. [Google Scholar] [CrossRef] [PubMed]
- Grosse, L.; Wagner, N.; Emelyanov, A.; Molina, C.; Lacas-Gervais, S.; Wagner, K.-D.; Bulavin, D.V. Defined p16High Senescent Cell Types Are Indispensable for Mouse Healthspan. Cell Metab. 2020, 32, 87–99e6. [Google Scholar] [CrossRef] [PubMed]
- Matheu, A.; Pantoja, C.; Efeyan, A.; Criado, L.M.; Martín-Caballero, J.; Flores, J.M.; Klatt, P.; Serrano, M. Increased gene dosage of Ink4a/Arf results in cancer resistance and normal aging. Genes Dev. 2004, 18, 2736–2746. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- 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]
- Chandra, A.; Lagnado, A.B.; Farr, J.N.; Doolittle, M.; Tchkonia, T.; Kirkland, J.L.; LeBrasseur, N.K.; Robbins, P.D.; Niedernhofer, L.J.; Ikeno, Y.; et al. Targeted clearance of p21- but not p16-positive senescent cells prevents radiation-induced osteoporosis and increased marrow adiposity. Aging Cell 2022, 21, e13602. [Google Scholar] [CrossRef]
- Sturmlechner, I.; Zhang, C.; Sine, C.C.; van Deursen, E.-J.; Jeganathan, K.B.; Hamada, N.; Grasic, J.; Friedman, D.; Stutchman, J.T.; Can, I.; et al. p21 produces a bioactive secretome that places stressed cells under immunosurveillance. Science 2021, 374, eabb3420. [Google Scholar] [CrossRef]
- 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]
- 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]
- 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, 1061–1077e8. [Google Scholar] [CrossRef]
- 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]
- Lewis-McDougall, F.C.; Ruchaya, P.J.; Domenjo-Vila, E.; Shin Teoh, T.; 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]
- 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] [PubMed]
- Cohen, C.; Le Goff, O.; Soysouvanh, F.; Vasseur, F.; Tanou, M.; Nguyen, C.; Amrouche, L.; Le Guen, J.; Saltel-Fulero, O.; Meunier, T.; et al. Glomerular endothelial cell senescence drives age-related kidney disease through PAI-1. EMBO Mol. Med. 2021, 13, e14146. [Google Scholar] [CrossRef] [PubMed]
- Patil, P.; Dong, Q.; Wang, D.; Chang, J.; Wiley, C.; Demaria, M.; Lee, J.; Kang, J.; Niedernhofer, L.J.; Robbins, P.D.; et al. Systemic clearance of p16INK4a -positive senescent cells mitigates age-associated intervertebral disc degeneration. Aging Cell 2019, 18, e12927. [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]
- Jeon, O.H.; Mehdipour, M.; Gil, T.-H.; Kang, M.; Aguirre, N.W.; Robinson, Z.R.; Kato, C.; Etienne, J.; Lee, H.G.; Alimirah, F.; et al. Systemic induction of senescence in young mice after single heterochronic blood exchange. Nat. Metab. 2022, 4, 995–1006. [Google Scholar] [CrossRef]
- Blanpain, C.; Fuchs, E. Epidermal Stem Cells of the Skin. Annu. Rev. Cell Dev. Biol. 2006, 22, 339–373. [Google Scholar] [CrossRef]
- Kligman, A.M. Perspectives and Problems in Cutaneous Gerontology. J. Investig. Dermatol. 1979, 73, 39–46. [Google Scholar] [CrossRef]
- Montagna, W.; Carlisle, K. Structural Changes in Aging Human Skin. J. Investig. Dermatol. 1979, 73, 47–53. [Google Scholar] [CrossRef]
- Mimeault, M.; Batra, S.K. Recent advances on skin-resident stem/progenitor cell functions in skin regeneration, aging and cancers and novel anti-aging and cancer therapies. J. Cell. Mol. Med. 2010, 14, 116–134. [Google Scholar] [CrossRef]
- Debacq-Chainiaux, F.; Borlon, C.; Pascal, T.; Royer, V.; Eliaers, F.; Ninane, N.; Carrard, G.; Friguet, B.; de Longueville, F.; Boffe, S.; et al. Repeated exposure of human skin fibroblasts to UVB at subcytotoxic level triggers premature senescence through the TGF-β1 signaling pathway. J. Cell Sci. 2005, 118, 743–758. [Google Scholar] [CrossRef] [PubMed]
- Lewis, D.A.; Yi, Q.; Travers, J.B.; Spandau, D.F. UVB-induced Senescence in Human Keratinocytes Requires a Functional Insulin-like Growth Factor-1 Receptor and p53. Mol. Biol. Cell 2008, 19, 1346–1353. [Google Scholar] [CrossRef] [PubMed]
- McCart, E.A.; Thangapazham, R.L.; Lombardini, E.D.; Mog, S.R.; Panganiban, R.A.M.; Dickson, K.M.; Mansur, R.A.; Nagy, V.; Kim, S.-Y.; Selwyn, R.; et al. Accelerated senescence in skin in a murine model of radiation-induced multi-organ injury. J. Radiat. Res. 2017, 58, 636–646. [Google Scholar] [CrossRef] [Green Version]
- Nobori, T.; Miura, K.; Wu, D.J.; Lois, A.; Takabayashi, K.; Carson, D.A. Deletions of the cyclin-dependent kinase-4 inhibitor gene in multiple human cancers. Nature 1994, 368, 753–756. [Google Scholar] [CrossRef]
- Scholes, A.G.; Liloglou, T.; Maloney, P.; Hagan, S.; Nunn, J.; Hiscott, P.; Damato, B.E.; Grierson, I.; Field, J.K. Loss of heterozygosity on chromosomes 3, 9, 13, and 17, including the retinoblastoma locus, in uveal melanoma. Investig. Ophthalmol. Vis. Sci. 2001, 42, 2472–2477. [Google Scholar]
- Castellano, M.; Pollock, P.M.; Walters, M.K.; Sparrow, L.E.; Down, L.M.; Gabrielli, B.G.; Parsons, P.G.; Hayward, N.K. CDKN2A/p16 is inactivated in most melanoma cell lines. Cancer Res. 1997, 57, 4868–4875. [Google Scholar]
- Funk, J.O.; Schiller, P.I.; Barrett, M.T.; Wong, D.J.; Kind, P.; Sander, C.A. p16INK4a expression is frequently decreased and associated with 9p21 loss of heterozygosity in sporadic melanoma. J. Cutan. Pathol. 1998, 25, 291–296. [Google Scholar] [CrossRef] [PubMed]
- Straume, O.; Sviland, L.; Akslen, L.A. Loss of nuclear p16 protein expression correlates with increased tumor cell proliferation (Ki-67) and poor prognosis in patients with vertical growth phase melanoma. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2000, 6, 1845–1853. [Google Scholar]
- Mihic-Probst, D.; Mnich, C.D.; Oberholzer, P.A.; Seifert, B.; Sasse, B.; Moch, H.; Dummer, R. p16 expression in primary malignant melanoma is associated with prognosis and lymph node status. Int. J. Cancer 2006, 118, 2262–2268. [Google Scholar] [CrossRef]
- Tyagi, E.; Liu, B.; Li, C.; Liu, T.; Rutter, J.; Grossman, D. Loss of p16INK4A stimulates aberrant mitochondrial biogenesis through a CDK4/Rb-independent pathway. Oncotarget 2017, 8, 55848–55862. [Google Scholar] [CrossRef]
- Al-Khalaf, H.H.; Mohideen, P.; Nallar, S.C.; Kalvakolanu, D.V.; Aboussekhra, A. The cyclin-dependent kinase inhibitor p16INK4a physically interacts with transcription factor Sp1 and cyclin-dependent kinase 4 to transactivate microRNA-141 and microRNA-146b-5p spontaneously and in response to ultraviolet light-induced DNA damage. J. Biol. Chem. 2013, 288, 35511–35525. [Google Scholar] [CrossRef]
- Jun, J.-I.; Lau, L.F. The matricellular protein CCN1 induces fibroblast senescence and restricts fibrosis in cutaneous wound healing. Nat. Cell Biol. 2010, 12, 676–685. [Google Scholar] [CrossRef] [PubMed]
- Pavey, S.; Conroy, S.; Russell, T.; Gabrielli, B. Ultraviolet Radiation Induces p16CDKN2A Expression in Human Skin1. Cancer Res. 1999, 59, 4185–4189. [Google Scholar] [PubMed]
- Adam, R.C.; Fuchs, E. The Yin and Yang of Chromatin Dynamics In Stem Cell Fate Selection. Trends Genet. 2016, 32, 89–100. [Google Scholar] [CrossRef] [PubMed]
- Perdigoto, C.N.; Valdes, V.J.; Bardot, E.S.; Ezhkova, E. Epigenetic regulation of epidermal differentiation. Cold Spring Harb. Perspect. Med. 2014, 4, a015263. [Google Scholar] [CrossRef] [PubMed]
- Botchkarev, V.A.; Gdula, M.R.; Mardaryev, A.N.; Sharov, A.A.; Fessing, M.Y. Epigenetic regulation of gene expression in keratinocytes. J. Investig. Dermatol. 2012, 132, 2505–2521. [Google Scholar] [CrossRef] [Green Version]
- Eckert, R.L.; Adhikary, G.; Rorke, E.A.; Chew, Y.C.; Balasubramanian, S. Polycomb group proteins are key regulators of keratinocyte function. J. Investig. Dermatol. 2011, 131, 295–301. [Google Scholar] [CrossRef]
- Avgustinova, A.; Benitah, S.A. Epigenetic control of adult stem cell function. Nat. Rev. Mol. Cell Biol. 2016, 17, 643–658. [Google Scholar] [CrossRef]
- Safwan-Zaiter, H.; Wagner, N.; Michiels, J.-F.; Wagner, K.-D. Dynamic Spatiotemporal Expression Pattern of the Senescence-Associated Factor p16Ink4a in Development and Aging. Cells 2022, 11, 541. [Google Scholar] [CrossRef]
- Langlands, K.; Down, G.A.; Kealey, T. Id proteins are dynamically expressed in normal epidermis and dysregulated in squamous cell carcinoma. Cancer Res. 2000, 60, 5929–5933. [Google Scholar]
- Chen, X.; Wang, Z.; Duan, N.; Zhu, G.; Schwarz, E.M.; Xie, C. Osteoblast-Osteoclast Interactions. Connect. Tissue Res. 2018, 59, 99–107. [Google Scholar] [CrossRef] [PubMed]
- Jilka, R.L.; O’Brien, C.A. The Role of Osteocytes in Age-Related Bone Loss. Curr. Osteoporos. Rep. 2016, 14, 16–25. [Google Scholar] [CrossRef] [PubMed]
- Almeida, M.; Han, L.; Martin-Millan, M.; Plotkin, L.I.; Stewart, S.A.; Roberson, P.K.; Kousteni, S.; O’Brien, C.A.; Bellido, T.; Parfitt, A.M.; et al. Skeletal involution by age-associated oxidative stress and its acceleration by loss of sex steroids. J. Biol. Chem. 2007, 282, 27285–27297. [Google Scholar] [CrossRef]
- Glatt, V.; Canalis, E.; Stadmeyer, L.; Bouxsein, M.L. Age-related changes in trabecular architecture differ in female and male C57BL/6J mice. J. Bone Miner. Res. 2007, 22, 1197–1207. [Google Scholar] [CrossRef] [PubMed]
- Piemontese, M.; Almeida, M.; Robling, A.G.; Kim, H.-N.; Xiong, J.; Thostenson, J.D.; Weinstein, R.S.; Manolagas, S.C.; O’Brien, C.A.; Jilka, R.L. Old age causes de novo intracortical bone remodeling and porosity in mice. JCI Insight 2017, 2, 93771. [Google Scholar] [CrossRef]
- Ucer, S.; Iyer, S.; Kim, H.-N.; Han, L.; Rutlen, C.; Allison, K.; Thostenson, J.D.; de Cabo, R.; Jilka, R.L.; O’Brien, C.; et al. The Effects of Aging and Sex Steroid Deficiency on the Murine Skeleton Are Independent and Mechanistically Distinct. J. Bone Miner. Res. 2017, 32, 560–574. [Google Scholar] [CrossRef]
- Marie, P.J. Bone Cell Senescence: Mechanisms and Perspectives. J. Bone Miner. Res. 2014, 29, 1311–1321. [Google Scholar] [CrossRef]
- Baron, R.; Kneissel, M. WNT signaling in bone homeostasis and disease: From human mutations to treatments. Nat. Med. 2013, 19, 179–192. [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]
- Kim, H.-N.; Chang, J.; Shao, L.; Han, L.; Iyer, S.; Manolagas, S.C.; O’Brien, C.A.; Jilka, R.L.; Zhou, D.; Almeida, M. DNA damage and senescence in osteoprogenitors expressing Osx1 may cause their decrease with age. Aging Cell 2017, 16, 693–703. [Google Scholar] [CrossRef]
- 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]
- Kim, H.-N.; Chang, J.; Iyer, S.; Han, L.; Campisi, J.; Manolagas, S.C.; Zhou, D.; Almeida, M. Elimination of senescent osteoclast progenitors has no effect on the age-associated loss of bone mass in mice. Aging Cell 2019, 18, e12923. [Google Scholar] [CrossRef] [PubMed]
- Peng, S.; Chen, X.; Huang, C.; Yang, C.; Situ, M.; Zhou, Q.; Ling, Y.; Huang, H.; Huang, M.; Zhang, Y.; et al. UBE2S as a novel ubiquitinated regulator of p16 and β-catenin to promote bone metastasis of prostate cancer. Int. J. Biol. Sci. 2022, 18, 3528–3543. [Google Scholar] [CrossRef] [PubMed]
- Harris, A.S.; Thomas, R.G.; Passant, C.D. Do patients with p16-positive oropharyngeal squamous cell carcinoma get more bone metastasis than p16-negative patients? J. Laryngol. Otol. 2018, 132, 429–433. [Google Scholar] [CrossRef]
- Righi, A.; Gambarotti, M.; Sbaraglia, M.; Sisto, A.; Ferrari, S.; Dei Tos, A.P.; Picci, P. p16 expression as a prognostic and predictive marker in high-grade localized osteosarcoma of the extremities: An analysis of 357 cases. Hum. Pathol. 2016, 58, 15–23. [Google Scholar] [CrossRef]
- Li, J.; Karim, M.A.; Che, H.; Geng, Q.; Miao, D. Deletion of p16 prevents estrogen deficiency-induced osteoporosis by inhibiting oxidative stress and osteocyte senescence. Am. J. Transl. Res. 2020, 12, 672–683. [Google Scholar]
- Ding, Q.; Liu, H.; Liu, L.; Ma, C.; Qin, H.; Wei, Y.; Ren, Y. Deletion of p16 accelerates fracture healing in geriatric mice. Am. J. Transl. Res. 2021, 13, 11107. [Google Scholar]
- Mercado, N.; Ito, K.; Barnes, P.J. Accelerated ageing of the lung in COPD: New concepts. Thorax 2015, 70, 482–489. [Google Scholar] [CrossRef]
- Fukuchi, Y. The aging lung and chronic obstructive pulmonary disease: Similarity and difference. Proc. Am. Thorac. Soc. 2009, 6, 570–572. [Google Scholar] [CrossRef]
- John-Schuster, G.; Günter, S.; Hager, K.; Conlon, T.M.; Eickelberg, O.; Yildirim, A.Ö. Inflammaging increases susceptibility to cigarette smoke-induced COPD. Oncotarget 2016, 7, 30068–30083. [Google Scholar] [CrossRef]
- Meiners, S.; Eickelberg, O.; Königshoff, M. Hallmarks of the ageing lung. Eur. Respir. J. 2015, 45, 807–827. [Google Scholar] [CrossRef]
- Selman, M.; Buendía-Roldán, I.; Pardo, A. Aging and Pulmonary Fibrosis. Rev. Investig. Clin. 2016, 68, 75–83. [Google Scholar]
- Venosa, A. Senescence in Pulmonary Fibrosis: Between Aging and Exposure. Front. Med. 2020, 7, 606462. [Google Scholar] [CrossRef] [PubMed]
- Kheradmand, F.; You, R.; Hee Gu, B.; Corry, D.B. Cigarette Smoke and DNA Cleavage Promote Lung Inflammation and Emphysema. Trans. Am. Clin. Climatol. Assoc. 2017, 128, 222–233. [Google Scholar]
- Nyunoya, T.; Monick, M.M.; Klingelhutz, A.L.; Glaser, H.; Cagley, J.R.; Brown, C.O.; Matsumoto, E.; Aykin-Burns, N.; Spitz, D.R.; Oshima, J.; et al. Cigarette Smoke Induces Cellular Senescence via Werner’s Syndrome Protein Down-regulation. Am. J. Respir. Crit. Care Med. 2009, 179, 279–287. [Google Scholar] [CrossRef]
- Nyunoya, T.; Monick, M.M.; Klingelhutz, A.; Yarovinsky, T.O.; Cagley, J.R.; Hunninghake, G.W. Cigarette Smoke Induces Cellular Senescence. Am. J. Respir. Cell Mol. Biol. 2006, 35, 681–688. [Google Scholar] [CrossRef] [PubMed]
- Rashid, K.; Sundar, I.K.; Gerloff, J.; Li, D.; Rahman, I. Lung cellular senescence is independent of aging in a mouse model of COPD/emphysema. Sci. Rep. 2018, 8, 9023. [Google Scholar] [CrossRef]
- Cottage, C.T.; Peterson, N.; Kearley, J.; Berlin, A.; Xiong, X.; Huntley, A.; Zhao, W.; Brown, C.; Migneault, A.; Zerrouki, K.; et al. Targeting p16-induced senescence prevents cigarette smoke-induced emphysema by promoting IGF1/Akt1 signaling in mice. Commun. Biol. 2019, 2, 307. [Google Scholar] [CrossRef]
- Wang, C.-W.; Wu, T.-I.; Yu, C.-T.; Wu, Y.-C.; Teng, Y.-H.; Chin, S.-Y.; Lai, C.-H.; Chen, T.-C. Usefulness of p16 for differentiating primary pulmonary squamous cell carcinoma from cervical squamous cell carcinoma metastatic to the lung. Am. J. Clin. Pathol. 2009, 131, 715–722. [Google Scholar] [CrossRef]
- Belinsky, S.A.; Nikula, K.J.; Palmisano, W.A.; Michels, R.; Saccomanno, G.; Gabrielson, E.; Baylin, S.B.; Herman, J.G. Aberrant methylation of p16(INK4a) is an early event in lung cancer and a potential biomarker for early diagnosis. Proc. Natl. Acad. Sci. USA 1998, 95, 11891–11896. [Google Scholar] [CrossRef]
- Okamoto, A.; Hussain, S.P.; Hagiwara, K.; Spillare, E.A.; Rusin, M.R.; Demetrick, D.J.; Serrano, M.; Hannon, G.J.; Shiseki, M.; Zariwala, M.; et al. Mutations in the p16INK4/MTS1/CDKN2, p15INK4B/MTS2, and p18 Genes in Primary and Metastatic Lung Cancer1. Cancer Res. 1995, 55, 1448–1451. [Google Scholar] [PubMed]
- Tong, J.; Sun, X.; Cheng, H.; Zhao, D.; Ma, J.; Zhen, Q.; Cao, Y.; Zhu, H.; Bai, J. Expression of p16 in non-small cell lung cancer and its prognostic significance: A meta-analysis of published literatures. Lung Cancer 2011, 74, 155–163. [Google Scholar] [CrossRef] [PubMed]
- p16 Regulation of Lung Epithelial Cell Growth, Repair after Injury and Transformation—ProQuest. Available online: https://www.proquest.com/openview/c4b38b5ea3ce9a758bbacff031039f5c/1?pq-origsite=gscholar&cbl=18750&diss=y (accessed on 28 July 2022).
- De Mochel, N.R.; Cheong, K.N.; Cassandras, M.; Wang, C.; Krasilnikov, M.; Matatia, P.; Molofsky, A.; Campisi, J.; Peng, T. Sentinel p16INK4a+ cells in the basement membrane form a reparative niche in the lung 2020. bioRxiv 2020. [CrossRef]
- Molofsky, A.V.; Slutsky, S.G.; Joseph, N.M.; He, S.; Pardal, R.; Krishnamurthy, J.; Sharpless, N.E.; Morrison, S.J. Increasing p16INK4a expression decreases forebrain progenitors and neurogenesis during ageing. Nature 2006, 443, 448–452. [Google Scholar] [CrossRef]
- Song, P.; An, J.; Zou, M.-H. Immune Clearance of Senescent Cells to Combat Ageing and Chronic Diseases. Cells 2020, 9, 671. [Google Scholar] [CrossRef] [PubMed]
- Lok, K.; Zhao, H.; Shen, H.; Wang, Z.; Gao, X.; Zhao, W.; Yin, M. Characterization of the APP/PS1 mouse model of Alzheimer’s disease in senescence accelerated background. Neurosci. Lett. 2013, 557, 84–89. [Google Scholar] [CrossRef] [PubMed]
- Dorigatti, A.O.; Riordan, R.; Yu, Z.; Ross, G.; Wang, R.; Reynolds-Lallement, N.; Magnusson, K.; Galvan, V.; Perez, V.I. Brain cellular senescence in mouse models of Alzheimer’s disease. GeroScience 2022, 44, 1157–1168. [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]
- Ramsden, M.; Kotilinek, L.; Forster, C.; Paulson, J.; McGowan, E.; SantaCruz, K.; Guimaraes, A.; Yue, M.; Lewis, J.; Carlson, G.; et al. Age-Dependent Neurofibrillary Tangle Formation, Neuron Loss, and Memory Impairment in a Mouse Model of Human Tauopathy (P301L). J. Neurosci. 2005, 25, 10637–10647. [Google Scholar] [CrossRef]
- Mv, S.; Hv, V. Astrocytes: Biology and pathology. Acta Neuropathol. 2010, 119, 7–35. [Google Scholar] [CrossRef]
- Halassa, M.M.; Fellin, T.; Haydon, P.G. Tripartite synapses: Roles for astrocytic purines in the control of synaptic physiology and behavior. Neuropharmacology 2009, 57, 343–346. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pertusa, M.; García-Matas, S.; Rodriguez-Farre, E.; Sanfeliu, C.; Cristòfol, R. Astrocytes aged in vitro show a decreased neuroprotective capacity. J. Neurochem. 2007, 101, 794–805. [Google Scholar] [CrossRef] [PubMed]
- Cohen, J.; Torres, C. Astrocyte senescence: Evidence and significance. Aging Cell 2019, 18, e12937. [Google Scholar] [CrossRef] [PubMed]
- 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]
- Jen, J.; Harper, J.W.; Bigner, S.H.; Bigner, D.D.; Papadopoulos, N.; Markowitz, S.; Willson, J.K.V.; Kinzler, K.W.; Vogelstein, B. Deletion of p16 and p15 Genes in Brain Tumors1. Cancer Res. 1994, 54, 6353–6358. [Google Scholar]
- Ueki, K.; Ono, Y.; Henson, J.W.; Efird, J.T.; von Deimling, A.; Louis, D.N. CDKN2/p16 or RB Alterations Occur in the Majority of Glioblastomas and Are Inversely Correlated1. Cancer Res. 1996, 56, 150–153. [Google Scholar]
- Park, J.W.; Kang, J.; Lim, K.Y.; Kim, H.; Kim, S.-I.; Won, J.K.; Park, C.-K.; Park, S.-H. The prognostic significance of p16 expression pattern in diffuse gliomas. J. Pathol. Transl. Med. 2021, 55, 102–111. [Google Scholar] [CrossRef]
- Fueyo, J.; Gomez-Manzano, C.; Puduvalli, V.K.; Martin-Duque, P.; Perez-Soler, R.; Levin, V.A.; Yung, W.K.; Kyritsis, A.P. Adenovirus-mediated p16 transfer to glioma cells induces G1 arrest and protects from paclitaxel and topotecan: Implications for therapy. Int. J. Oncol. 1998, 12, 665–674. [Google Scholar] [CrossRef]
- Kranenburg, O.; van der Eb, A.J.; Zantema, A. Cyclin D1 is an essential mediator of apoptotic neuronal cell death. EMBO J. 1996, 15, 46–54. [Google Scholar] [CrossRef]
- Kfoury, N.; Sun, T.; Yu, K.; Rockwell, N.; Tinkum, K.L.; Qi, Z.; Warrington, N.M.; McDonald, P.; Roy, A.; Weir, S.J.; et al. Cooperative p16 and p21 action protects female astrocytes from transformation. Acta Neuropathol. Commun. 2018, 6, 12. [Google Scholar] [CrossRef]
- Yabluchanskiy, A.; Tarantini, S.; Balasubramanian, P.; Kiss, T.; Csipo, T.; Fülöp, G.A.; Lipecz, A.; Ahire, C.; DelFavero, J.; Nyul-Toth, A.; et al. Pharmacological or genetic depletion of senescent astrocytes prevents whole brain irradiation–induced impairment of neurovascular coupling responses protecting cognitive function in mice. GeroScience 2020, 42, 409–428. [Google Scholar] [CrossRef] [PubMed]
- Qian, J.; Wang, X.; Cao, J.; Zhang, W.; Lu, C.; Chen, X. Dihydromyricetin attenuates D-galactose-induced brain aging of mice via inhibiting oxidative stress and neuroinflammation. Neurosci. Lett. 2021, 756, 135963. [Google Scholar] [CrossRef]
- Torella, D.; Rota, M.; Nurzynska, D.; Musso, E.; Monsen, A.; Shiraishi, I.; Zias, E.; Walsh, K.; Rosenzweig, A.; Sussman, M.A.; et al. Cardiac stem cell and myocyte aging, heart failure, and insulin-like growth factor-1 overexpression. Circ. Res. 2004, 94, 514–524. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chimenti, C.; Kajstura, J.; Torella, D.; Urbanek, K.; Heleniak, H.; Colussi, C.; Di Meglio, F.; Nadal-Ginard, B.; Frustaci, A.; Leri, A.; et al. Senescence and Death of Primitive Cells and Myocytes Lead to Premature Cardiac Aging and Heart Failure. Circ. Res. 2003, 93, 604–613. [Google Scholar] [CrossRef] [PubMed]
- Kajstura, J.; Pertoldi, B.; Leri, A.; Beltrami, C.-A.; Deptala, A.; Darzynkiewicz, Z.; Anversa, P. Telomere Shortening Is an in Vivo Marker of Myocyte Replication and Aging. Am. J. Pathol. 2000, 156, 813–819. [Google Scholar] [CrossRef]
- Matthews, C.; Gorenne, I.; Scott, S.; Figg, N.; Kirkpatrick, P.; Ritchie, A.; Goddard, M.; Bennett, M. Vascular Smooth Muscle Cells Undergo Telomere-Based Senescence in Human Atherosclerosis. Circ. Res. 2006, 99, 156–164. [Google Scholar] [CrossRef] [PubMed]
- Cianflone, E.; Torella, M.; Biamonte, F.; De Angelis, A.; Urbanek, K.; Costanzo, F.S.; Rota, M.; Ellison-Hughes, G.M.; Torella, D. Targeting Cardiac Stem Cell Senescence to Treat Cardiac Aging and Disease. Cells 2020, 9, 1558. [Google Scholar] [CrossRef] [PubMed]
- Epstein, J.A. A Time to Press Reset and Regenerate Cardiac Stem Cell Biology. JAMA Cardiol. 2019, 4, 95–96. [Google Scholar] [CrossRef]
- Shi, J.; Sun, J.; Liu, L.; Shan, T.; Meng, H.; Yang, T.; Wang, S.; Wei, T.; Chen, B.; Ma, Y.; et al. P16ink4a overexpression ameliorates cardiac remodeling of mouse following myocardial infarction via CDK4/pRb pathway. Biochem. Biophys. Res. Commun. 2022, 595, 62–68. [Google Scholar] [CrossRef]
- Joosten, S.A.; van Ham, V.; Nolan, C.E.; Borrias, M.C.; Jardine, A.G.; Shiels, P.G.; van Kooten, C.; Paul, L.C. Telomere Shortening and Cellular Senescence in a Model of Chronic Renal Allograft Rejection. Am. J. Pathol. 2003, 162, 1305–1312. [Google Scholar] [CrossRef]
- Chkhotua, A.B.; Gabusi, E.; Altimari, A.; D’Errico, A.; Yakubovich, M.; Vienken, J.; Stefoni, S.; Chieco, P.; Yussim, A.; Grigioni, W.F. Increased expression of p16(INK4a) and p27(Kip1) cyclin-dependent kinase inhibitor genes in aging human kidney and chronic allograft nephropathy. Am. J. Kidney Dis. 2003, 41, 1303–1313. [Google Scholar] [CrossRef]
- Melk, A.; Schmidt, B.M.W.; Vongwiwatana, A.; Rayner, D.C.; Halloran, P.F. Increased Expression of Senescence-Associated Cell Cycle Inhibitor p16INK4a in Deteriorating Renal Transplants and Diseased Native Kidney. Am. J. Transplant. 2005, 5, 1375–1382. [Google Scholar] [CrossRef] [PubMed]
- Melk, A.; Schmidt, B.M.W.; Takeuchi, O.; Sawitzki, B.; Rayner, D.C.; Halloran, P.F. Expression of p16INK4a and other cell cycle regulator and senescence associated genes in aging human kidney. Kidney Int. 2004, 65, 510–520. [Google Scholar] [CrossRef] [Green Version]
- Sis, B.; Tasanarong, A.; Khoshjou, F.; Dadras, F.; Solez, K.; Halloran, P.F. Accelerated expression of senescence associated cell cycle inhibitor p16INK4A in kidneys with glomerular disease. Kidney Int. 2007, 71, 218–226. [Google Scholar] [CrossRef]
- Westhoff, J.H.; Hilgers, K.F.; Steinbach, M.P.; Hartner, A.; Klanke, B.; Amann, K.; Melk, A. Hypertension induces somatic cellular senescence in rats and humans by induction of cell cycle inhibitor p16INK4a. Hypertension 2008, 52, 123–129. [Google Scholar] [CrossRef] [PubMed]
- Liu, J.; Yang, J.-R.; Chen, X.-M.; Cai, G.-Y.; Lin, L.-R.; He, Y.-N. Impact of ER stress-regulated ATF4/p16 signaling on the premature senescence of renal tubular epithelial cells in diabetic nephropathy. Am. J. Physiol.-Cell Physiol. 2015, 308, C621–C630. [Google Scholar] [CrossRef]
- Gu, X.; Peng, C.-Y.; Lin, S.-Y.; Qin, Z.-Y.; Liang, J.-L.; Chen, H.-J.; Hou, C.-X.; Wang, R.; Du, Y.-Q.; Jin, J.-L.; et al. P16INK4a played a critical role in exacerbating acute tubular necrosis in acute kidney injury. Am. J. Transl. Res. 2019, 11, 3850–3861. [Google Scholar]
- Jin, J.; Tao, J.; Gu, X.; Yu, Z.; Wang, R.; Zuo, G.; Li, Q.; Lv, X.; Miao, D. P16 INK4a Deletion Ameliorated Renal Tubulointerstitial Injury in a Stress-induced Premature Senescence Model of Bmi-1 Deficiency. Sci. Rep. 2017, 7, 7502. [Google Scholar] [CrossRef]
- Baiocchi, L.; Glaser, S.; Francis, H.; Kennedy, L.; Felli, E.; Alpini, G.; Gracia-Sancho, J. Impact of Aging on Liver Cells and Liver Disease: Focus on the Biliary and Vascular Compartments. Hepatol. Commun. 2021, 5, 1125–1137. [Google Scholar] [CrossRef]
- Kim, H.; Kisseleva, T.; Brenner, D.A. Aging and liver disease. Curr. Opin. Gastroenterol. 2015, 31, 184–191. [Google Scholar] [CrossRef]
- Aravinthan, A.D.; Alexander, G.J.M. Senescence in chronic liver disease: Is the future in aging? J. Hepatol. 2016, 65, 825–834. [Google Scholar] [CrossRef] [PubMed]
- Zhu, C.; Ikemoto, T.; Utsunomiya, T.; Yamada, S.; Morine, Y.; Imura, S.; Arakawa, Y.; Takasu, C.; Ishikawa, D.; Shimada, M. Senescence-related genes possibly responsible for poor liver regeneration after hepatectomy in elderly patients. J. Gastroenterol. Hepatol. 2014, 29, 1102–1108. [Google Scholar] [CrossRef] [PubMed]
- Sawada, N. Hepatocytes from old rats retain responsiveness of c-myc expression to EGF in primary culture but do not enter S phase. Exp. Cell Res. 1989, 181, 584–588. [Google Scholar] [CrossRef]
- Maeso-Díaz, R.; Ortega-Ribera, M.; Fernández-Iglesias, A.; Hide, D.; Muñoz, L.; Hessheimer, A.J.; Vila, S.; Francés, R.; Fondevila, C.; Albillos, A.; et al. Effects of aging on liver microcirculatory function and sinusoidal phenotype. Aging Cell 2018, 17, e12829. [Google Scholar] [CrossRef] [PubMed]
- Omori, S.; Wang, T.-W.; Johmura, Y.; Kanai, T.; Nakano, Y.; Kido, T.; Susaki, E.A.; Nakajima, T.; Shichino, S.; Ueha, S.; et al. Generation of a p16 Reporter Mouse and Its Use to Characterize and Target p16high Cells In Vivo. Cell Metab. 2020, 32, 814–828e6. [Google Scholar] [CrossRef]
- González-Navarro, H.; Vinué, Á.; Sanz, M.J.; Delgado, M.; Pozo, M.A.; Serrano, M.; Burks, D.J.; Andrés, V. Increased dosage of Ink4/Arf protects against glucose intolerance and insulin resistance associated with aging. Aging Cell 2013, 12, 102–111. [Google Scholar] [CrossRef]
- Pal, A.; Potjer, T.P.; Thomsen, S.K.; Ng, H.J.; Barrett, A.; Scharfmann, R.; James, T.J.; Bishop, D.T.; Karpe, F.; Godsland, I.F.; et al. Loss-of-Function Mutations in the Cell-Cycle Control Gene CDKN2A Impact on Glucose Homeostasis in Humans. Diabetes 2016, 65, 527–533. [Google Scholar] [CrossRef]
- Bantubungi, K.; Hannou, S.-A.; Caron-Houde, S.; Vallez, E.; Baron, M.; Lucas, A.; Bouchaert, E.; Paumelle, R.; Tailleux, A.; Staels, B. Cdkn2a/p16Ink4a regulates fasting-induced hepatic gluconeogenesis through the PKA-CREB-PGC1α pathway. Diabetes 2014, 63, 3199–3209. [Google Scholar] [CrossRef]
- Deleye, Y.; Cotte, A.K.; Hannou, S.A.; Hennuyer, N.; Bernard, L.; Derudas, B.; Caron, S.; Legry, V.; Vallez, E.; Dorchies, E.; et al. CDKN2A/p16INK4a suppresses hepatic fatty acid oxidation through the AMPKα2-SIRT1-PPARα signaling pathway. J. Biol. Chem. 2020, 295, 17310–17322. [Google Scholar] [CrossRef]
- Zhang, X.; Xu, G.B.; Zhou, D.; Pan, Y.-X. High-fat diet modifies expression of hepatic cellular senescence gene p16(INK4a) through chromatin modifications in adult male rats. Genes Nutr. 2018, 13, 6. [Google Scholar] [CrossRef]
- Kyritsi, K.; Francis, H.; Zhou, T.; Ceci, L.; Wu, N.; Yang, Z.; Meng, F.; Chen, L.; Baiocchi, L.; Kundu, D.; et al. Downregulation of p16 Decreases Biliary Damage and Liver Fibrosis in the Mdr2−/− Mouse Model of Primary Sclerosing Cholangitis. Gene Expr. 2020, 20, 89–103. [Google Scholar] [CrossRef] [PubMed]
- Lv, F.; Li, N.; Kong, M.; Wu, J.; Fan, Z.; Miao, D.; Xu, Y.; Ye, Q.; Wang, Y. CDKN2a/p16 Antagonizes Hepatic Stellate Cell Activation and Liver Fibrosis by Modulating ROS Levels. Front. Cell Dev. Biol. 2020, 8, 176. [Google Scholar] [CrossRef] [PubMed]
- Zang, J.-J.; Xie, F.; Xu, J.-F.; Qin, Y.-Y.; Shen, R.-X.; Yang, J.-M.; He, J. P16 gene hypermethylation and hepatocellular carcinoma: A systematic review and meta-analysis. World J. Gastroenterol. 2011, 17, 3043–3048. [Google Scholar] [CrossRef] [PubMed]
- Wong, I.H.N.; Dennis Lo, Y.M.; Zhang, J.; Liew, C.-T.; Ng, M.H.L.; Wong, N.; Lai, P.B.S.; Lau, W.Y.; Hjelm, N.M.; Johnson, P.J. Detection of Aberrant p16 Methylation in the Plasma and Serum of Liver Cancer Patients1. Cancer Res. 1999, 59, 71–73. [Google Scholar] [PubMed]
- Wagner, K.-D.; Wagner, N. The Senescence Markers p16INK4A, p14ARF/p19ARF, and p21 in Organ Development and Homeostasis. Cells 2022, 11, 1966. [Google Scholar] [CrossRef] [PubMed]
- Vasey, D.B.; Roland Wolf, C.; Brown, K.; Whitelaw, C.B.A. Spatial p21 expression profile in the mid-term mouse embryo. Transgenic Res. 2011, 20, 23–28. [Google Scholar] [CrossRef]
- Melk, A.; Kittikowit, W.; Sandhu, I.; Halloran, K.M.; Grimm, P.; Schmidt, B.M.W.; Halloran, P.F. Cell senescence in rat kidneys in vivo increases with growth and age despite lack of telomere shortening. Kidney Int. 2003, 63, 2134–2143. [Google Scholar] [CrossRef]
- Rhinn, M.; Zapata-Bodalo, I.; Klein, A.; Plassat, J.-L.; Knauer-Meyer, T.; Keyes, W.M. Aberrant induction of p19Arf-mediated cellular senescence contributes to neurodevelopmental defects. PLoS Biol. 2022, 20, e3001664. [Google Scholar] [CrossRef]
- Cheong, C.; Sung, Y.H.; Lee, J.; Choi, Y.S.; Song, J.; Kee, C.; Lee, H.-W. Role of INK4a locus in normal eye development and cataract genesis. Mech. Ageing Dev. 2006, 127, 633–638. [Google Scholar] [CrossRef]
- Nacher, V.; Carretero, A.; Navarro, M.; Ayuso, E.; Ramos, D.; Luppo, M.; Rodríguez, A.; Mendes, L.; Herrero-Fresneda, I.; Ruberte, J. Endothelial Cell Transduction in Primary Cultures from Regressing Mesonephros. Cells Tissues Organs 2010, 191, 84–95. [Google Scholar] [CrossRef]
- Domínguez-Bautista, J.A.; Acevo-Rodríguez, P.S.; Castro-Obregón, S. Programmed Cell Senescence in the Mouse Developing Spinal Cord and Notochord. Front. Cell Dev. Biol. 2021, 9, 587096. [Google Scholar] [CrossRef]
- Wagner, N.; Ninkov, M.; Vukolic, A.; Cubukcuoglu Deniz, G.; Rassoulzadegan, M.; Michiels, J.-F.; Wagner, K.-D. Implications of the Wilms’ Tumor Suppressor Wt1 in Cardiomyocyte Differentiation. Int. J. Mol. Sci. 2021, 22, 94346. [Google Scholar] [CrossRef] [PubMed]
- Wagner, K.-D.; El Maï, M.; Ladomery, M.; Belali, T.; Leccia, N.; Michiels, J.-F.; Wagner, N. Altered VEGF Splicing Isoform Balance in Tumor Endothelium Involves Activation of Splicing Factors Srpk1 and Srsf1 by the Wilms’ Tumor Suppressor Wt1. Cells 2019, 8, 41. [Google Scholar] [CrossRef]
- El Maï, M.; Wagner, K.-D.; Michiels, J.-F.; Ambrosetti, D.; Borderie, A.; Destree, S.; Renault, V.; Djerbi, N.; Giraud-Panis, M.-J.; Gilson, E.; et al. The Telomeric Protein TRF2 Regulates Angiogenesis by Binding and Activating the PDGFRβ Promoter. Cell Rep. 2014, 9, 1047–1060. [Google Scholar] [CrossRef] [Green Version]
- Wagner, K.-D.; Cherfils-Vicini, J.; Hosen, N.; Hohenstein, P.; Gilson, E.; Hastie, N.D.; Michiels, J.-F.; Wagner, N. The Wilms’ tumour suppressor Wt1 is a major regulator of tumour angiogenesis and progression. Nat. Commun. 2014, 5, 5852. [Google Scholar] [CrossRef] [PubMed]
- Wagner, N.; Morrison, H.; Pagnotta, S.; Michiels, J.-F.; Schwab, Y.; Tryggvason, K.; Schedl, A.; Wagner, K.-D. The podocyte protein nephrin is required for cardiac vessel formation. Hum. Mol. Genet. 2011, 20, 2182–2194. [Google Scholar] [CrossRef]
- Dimri, G.P. The search for biomarkers of aging: Next stop INK4a/ARF locus. Sci. Aging Knowl. Environ. 2004, 2004, pe40. [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]
- González-Gualda, E.; Baker, A.G.; Fruk, L.; Muñoz-Espín, D. A guide to assessing cellular senescence in vitro and in vivo. FEBS J. 2021, 288, 56–80. [Google Scholar] [CrossRef]
- Basisty, N.; Kale, A.; Jeon, O.H.; Kuehnemann, C.; Payne, T.; Rao, C.; Holtz, A.; Shah, S.; Sharma, V.; Ferrucci, L.; et al. A proteomic atlas of senescence-associated secretomes for aging biomarker development. PLoS Biol. 2020, 18, e3000599. [Google Scholar] [CrossRef] [PubMed]
- Frescas, D.; Hall, B.M.; Strom, E.; Virtuoso, L.P.; Gupta, M.; Gleiberman, A.S.; Rydkina, E.; Balan, V.; Vujcic, S.; Chernova, O.B.; et al. Murine mesenchymal cells that express elevated levels of the CDK inhibitor p16(Ink4a) in vivo are not necessarily senescent. Cell Cycle 2017, 16, 1526–1533. [Google Scholar] [CrossRef] [PubMed]
- Wissler Gerdes, E.O.; Misra, A.; Netto, J.M.E.; Tchkonia, T.; Kirkland, J.L. Strategies for late phase preclinical and early clinical trials of senolytics. Mech. Ageing Dev. 2021, 200, 111591. [Google Scholar] [CrossRef] [PubMed]
- Chaib, S.; Tchkonia, T.; Kirkland, J.L. Cellular senescence and senolytics: The path to the clinic. Nat. Med. 2022, 28, 1556–1568. [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
Safwan-Zaiter, H.; Wagner, N.; Wagner, K.-D. P16INK4A—More Than a Senescence Marker. Life 2022, 12, 1332. https://doi.org/10.3390/life12091332
Safwan-Zaiter H, Wagner N, Wagner K-D. P16INK4A—More Than a Senescence Marker. Life. 2022; 12(9):1332. https://doi.org/10.3390/life12091332
Chicago/Turabian StyleSafwan-Zaiter, Hasan, Nicole Wagner, and Kay-Dietrich Wagner. 2022. "P16INK4A—More Than a Senescence Marker" Life 12, no. 9: 1332. https://doi.org/10.3390/life12091332
APA StyleSafwan-Zaiter, H., Wagner, N., & Wagner, K.-D. (2022). P16INK4A—More Than a Senescence Marker. Life, 12(9), 1332. https://doi.org/10.3390/life12091332