Epigenetic Clock and Circadian Rhythms in Stem Cell Aging and Rejuvenation
Abstract
:1. Introduction
2. Circadian Clocks and Cell Aging
2.1. Transcriptional and Metabolic Mechanisms of Circadian Rhythms
2.2. Cell Cycle Control
3. Epigenetic Clocks
4. Are Circadian and Epigenetic Clocks Interrelated/Interconnected?
5. Managing the Process of Cellular Aging. Can We Fool the Clock?
5.1. Resident Stem Cells and Aging
5.2. Basic Mechanisms of Stem Cell Aging
5.3. Reprogramming by Pluripotency Factors, Epigenetic Clock and Aging
5.4. Epigenetic Approaches to Stem Cell Rejuvenation
6. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Acknowledgments
Conflicts of Interest
Abbreviations
ARC | Arcuate Nucleus |
AVP | Arginine-Vasopressin Peptide |
b-HLH-PAS | Basic Helix–Loop–Helix/Per-ARNT-SIM |
BMAL1 | Brain and Muscle ARNT-Like 1 |
CBP | CREB Binding Protein |
CCG | Clock-Controlled Genes |
CHK1/2 | Checkpoint Kinase 1/2 |
CK2 | Casein Kinase 2 |
CLOCK | Circadian Locomotor Output Cycles Kaput |
CpG | 5’-Cytosine-phosphate-Guanine-3’ dinucleotide |
Cry1-2 | Cryptochrome1-2 |
DBP | D site of albumin promoter (albumin D-box) binding protein |
EZH2 | Enhancer of Zeste Homolog 2 |
H3K27 | Histone H3 lysine 27 residue |
H3K27me3 | tri-methylated lysine 27 of histone H3 |
H3K36me3 | tri-methylated lysine 36 residue of histone H3 |
H3K4me3 | tri-methylated lysine 4 residue of histone H3 |
H3K9me3 | tri-methylated lysine 9 residue of histone H3 |
HAT | Histone Acetyl Transferase |
HIF | Hypoxia-Inducible Factor |
HSF1 | Heat Shock Transcription Factor 1 |
Jarid1A | histone demethylase Jumonji/ARID Domain-containing protein 1A |
JMJD5 | Jumonji-C (JmjC) Domain-containing protein 5 |
MLL1 | Methyltransferase mixed Lineage Leukemia 1 |
Myc | proto-oncogene MYC |
NAMPT | Nicotinamide phosphoribosyltransferase |
NONO | Non-POU Domain-Containing Octamer-Binding Protein |
p16-Ink4A | Cyclin-Dependent Kinase Inhibitor 2A |
p300 | histone acetyltransferase p300 |
PAI-1 | Plasminogen Activator Inhibitor 1 |
Per1-2 | Period1-2 |
PGC-1α | Peroxisome proliferator-activated receptor-gamma Coactivator -1alpha |
REV-ERBα/β | orphan nuclear-receptor genes Rev-Erbα/β |
ROR α/β | Orphan receptor Response α/β |
RTH | Retinohypothalamic Tract |
SCN | Suprachiasmatic Nucleus |
SIRT | Sirtuin |
TEF | Thyrotroph Embryonic Factor |
TGF-β | Transforming Growth Factor beta |
VIP | Vasoactive Intestinal Peptide |
VMH | Ventromedial Hypothalamus |
Wnt | Wingless-type MMTV integration site family |
References
- Krems, C.; Lührmann, P.M.; Strassburg, A.; Hartmann, B.; Neuhäuser-Berthold, M. Lower resting metabolic rate in the elderly may not be entirely due to changes in body composition. Eur. J. Clin. Nutr. 2005, 59, 255–262. [Google Scholar] [CrossRef]
- Atlantis, E.; Martin, S.A.; Haren, M.T.; Taylor, A.W.; Wittert., G.A. Florey Adelaide Male Aging Study. Lifestyle factors associated with age-related differences in body composition: The Florey Adelaide Male Aging Study. Am. J. Clin. Nutr. 2008, 88, 95–104. [Google Scholar] [CrossRef] [PubMed]
- Huang, Y.L.; Liu, R.Y.; Wang, Q.S.; Van Someren, E.J.; Xu, H.; Zhou, J.N. Age-associated difference in circadian sleep-wake and rest-activity rhythms. Physiol. Behav. 2002, 76, 597–603. [Google Scholar] [CrossRef]
- Haynes, L.; Maue, A.C. Effects of aging on T cell function. Curr. Opin. Immunol. 2009, 21, 414–417. [Google Scholar] [CrossRef]
- Samson, R.D.; Barnes, C.A. Impact of aging brain circuits on cognition. Eur. J. Neurosci. 2013, 37, 1903–1915. [Google Scholar] [CrossRef] [Green Version]
- Gao, H.; Hollyfield, J.G. Aging of the human retina. Differential loss of neurons and retinal pigment epithelial cells. Investig. Ophthalmol. Vis. Sci. 1992, 33, 1–17. [Google Scholar]
- Eckel-Mahan, K.L.; Storm, D.R. Circadian rhythms and memory: Not so simple as cogs and gears. EMBO Rep. 2009, 10, 584–591. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bass, J.; Takahashi, J.S. Circadian integration of metabolism and energetics. Science 2010, 330, 1349–1354. [Google Scholar] [CrossRef] [Green Version]
- Dibner, C.; Schibler, U.; Albrecht, U. The mammalian circadian timing system: Organization and coordination of central and peripheral clocks. Annu. Rev. Physiol. 2010, 72, 517–549. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Buhr, E.D.; Takahashi, J.S. Molecular components of the Mammalian circadian clock. Handb. Exp. Pharmacol. 2013, 2017, 3–27. [Google Scholar] [CrossRef] [Green Version]
- Evans, J.A.; Davidson, A.J. Health consequences of circadian disruption in humans and animal models. Prog. Mol. Biol. Transl. Sci. 2013, 119, 283–323. [Google Scholar] [CrossRef]
- Wyse, C.A.; Selman, C.; Page, M.M.; Coogan, A.N.; Hazlerigg, D.G. Circadian desynchrony and metabolic dysfunction; did light pollution make us fat? Med. Hypotheses 2011, 77, 1139–1144. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Parsons, M.J.; Moffitt, T.E.; Gregory, A.M.; Goldman-Mellor, S.; Nolan, P.M.; Poulton, R.; Caspi, A. Social jetlag, obesity and metabolic disorder: Investigation in a cohort study. Int. J. Obes. 2015, 39, 842–848. [Google Scholar] [CrossRef] [Green Version]
- Hannum, G.; Guinney, J.; Zhao, L.; Zhang, L.; Hughes, G.; Sadda, S.; Klotzle, B.; Bibikova, M.; Fan, J.B.; Gao, Y.; et al. Genome-wide methylation profiles reveal quantitative views of human aging rates. Mol. Cell 2013, 49, 359–367. [Google Scholar] [CrossRef] [Green Version]
- Horvath, S. DNA methylation age of human tissues and cell types (published correction appears in Genome Biol. 2015, 16, 96). Genome Biol. 2013, 14, R115. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Levine, M.E.; Lu, A.T.; Quach, A.; Chen, B.H.; Assimes, T.L.; Bandinelli, S.; Hou, L.; Baccarelli, A.A.; Stewart, J.D.; Li, Y.; et al. An epigenetic biomarker of aging for lifespan and healthspan. Aging 2018, 10, 573–591. [Google Scholar] [CrossRef] [Green Version]
- Field, A.E.; Robertson, N.A.; Wang, T.; Havas, A.; Ideker, T.; Adams, P.D. DNA Methylation Clocks in Aging: Categories, Causes, and Consequences. Mol. Cell 2018, 71, 882–895. [Google Scholar] [CrossRef] [Green Version]
- Booth, L.N.; Brunet, A. The Aging Epigenome. Mol. Cell 2016, 62, 728–744. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Masri, S.; Kinouchi, K.; Sassone-Corsi, P. Circadian clocks, epigenetics, and cancer. Curr. Opin. Oncol. 2015, 27, 50–56. [Google Scholar] [CrossRef] [Green Version]
- Orozco-Solis, R.; Sassone-Corsi, P. Circadian clock: Linking epigenetics to aging. Curr. Opin. Genet. Dev. 2014, 26, 66–72. [Google Scholar] [CrossRef] [Green Version]
- Johnson, C.H. Circadian clocks and cell division: What’s the pacemaker? Cell Cycle 2010, 9, 3864–3873. [Google Scholar] [CrossRef] [Green Version]
- Khapre, R.V.; Samsa, W.E.; Kondratov, R.V. Circadian regulation of cell cycle: Molecular connections between aging and the circadian clock. Ann. Med. 2010, 42, 404–415. [Google Scholar] [CrossRef] [PubMed]
- Bass, J. Circadian topology of metabolism. Nature 2012, 491, 348–356. [Google Scholar] [CrossRef] [PubMed]
- Wijnen, H.; Young, M.W. Interaction of circadian clocks and metabolic rhythms. Annu. Rev. Genet. 2006, 40, 409–448. [Google Scholar] [CrossRef]
- Damiola, F.; Le Minh, N.; Preitner, N.; Kornmann, B.; Fleury-Olela, F.; Schibler, U. Restricted feeding uncouples circadian oscillators in peripheral tissues from the central pacemaker in the suprachiasmatic nucleus. Genes Dev. 2000, 14, 2950–2961. [Google Scholar] [CrossRef] [Green Version]
- Saper, C.B.; Scammell, T.E.; Lu, J. Hypothalamic regulation of sleep and circadian rhythms. Nature 2005, 437, 1257–1263. [Google Scholar] [CrossRef] [PubMed]
- Hatori, M.; Vollmers, C.; Zarrinpar, A.; DiTacchio, L.; Bushong, E.A.; Gill, S.; Leblanc, M.; Chaix, A.; Joens, M.; Fitzpatrick, J.A.; et al. Time-restricted feeding without reducing caloric intake prevents metabolic diseases in mice fed a high-fat diet. Cell Metab. 2012, 15, 848–860. [Google Scholar] [CrossRef] [Green Version]
- Nakamura, T.J.; Nakamura, W.; Yamazaki, S.; Cutler, T.; Colwell, C.S.; Block, G.D. Age-related decline in circadian output. J. Neurosci. 2011, 31, 10201–10205. [Google Scholar] [CrossRef]
- Farajnia, S.; Michel, S.; Deboer, T.; Kudo, T.; Cutler, T.; Colwell, C.S.; Block, G.D. Evidence for neuronal desynchrony in the aged suprachiasmatic nucleus clock. J. Neurosci. 2012, 32, 5891–5899. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Farajnia, S.; Meijer, J.H.; Michel, S. Age-related changes in large-conductance calcium-activated potassium channels in mammalian circadian clock neurons. Neurobiol. Aging 2015, 36, 2176–2183. [Google Scholar] [CrossRef] [Green Version]
- Palomba, M.; Nygård, M.; Florenzano, F.; Bertini, G.; Kristensson, K.; Bentivoglio, M. Decline of the presynaptic network, including GABAergic terminals, in the aging suprachiasmatic nucleus of the mouse. J. Biol. Rhythms 2008, 23, 220–231. [Google Scholar] [CrossRef] [Green Version]
- Duncan, M.J.; Herron, J.M.; Hill, S.A. Aging selectively suppresses vasoactive intestinal peptide messenger RNA expression in the suprachiasmatic nucleus of the Syrian hamster. Brain Res. Mol. Brain Res. 2001, 87, 196–203. [Google Scholar] [CrossRef]
- Cayetanot, F.; Bentivoglio, M.; Aujard, F. Arginine-vasopressin and vasointestinal polypeptide rhythms in the suprachiasmatic nucleus of the mouse lemur reveal aging-related alterations of circadian pacemaker neurons in a non-human primate. Eur. J. Neurosci. 2005, 22, 902–910. [Google Scholar] [CrossRef]
- Maywood, E.S.; Reddy, A.B.; Wong, G.K.; O’Neill, J.S.; O’Brien, J.A.; McMahon, D.G.; Harmar, A.J.; Okamura, H.; Hastings, M.H. Synchronization and maintenance of timekeeping in suprachiasmatic circadian clock cells by neuropeptidergic signaling. Curr. Biol. 2006, 16, 599–605. [Google Scholar] [CrossRef] [Green Version]
- Reppert, S.M.; Weaver, D.R. Coordination of circadian timing in mammals. Nature 2002, 418, 935–941. [Google Scholar] [CrossRef] [PubMed]
- Ueda, H.R.; Hayashi, S.; Chen, W.; Sano, M.; Machida, M.; Shigeyoshi, Y.; Iino, M.; Hashimoto, S. System-level identification of transcriptional circuits underlying mammalian circadian clocks. Nat. Genet. 2005, 37, 187–192. [Google Scholar] [CrossRef] [PubMed]
- Wang, X.; Tian, G.; Li, Z.; Zheng, L. The crosstalk between miRNA and mammalian circadian clock. Curr. Med. Chem. 2015, 22, 1582–1588. [Google Scholar] [CrossRef]
- Godinho, S.I.; Maywood, E.S.; Shaw, L.; Tucci, V.; Barnard, A.R.; Busino, L.; Pagano, M.; Kendall, R.; Quwailid, M.M.; Romero, M.R.; et al. The after-hours mutant reveals a role for Fbxl3 in determining mammalian circadian period. Science 2007, 316, 897–900. [Google Scholar] [CrossRef] [Green Version]
- Aguilar-Arnal, L.; Sassone-Corsi, P. The circadian epigenome: How metabolism talks to chromatin remodeling. Curr. Opin. Cell Biol. 2013, 25, 170–176. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Eckel-Mahan, K.L.; Patel, V.R.; de Mateo, S.; Orozco-Solis, R.; Ceglia, N.J.; Sahar, S.; Dilag-Penilla, S.A.; Dyar, K.A.; Baldi, P.; Sassone-Corsi, P. Reprogramming of the circadian clock by nutritional challenge. Cell 2013, 155, 1464–1478. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Aguilar-Arnal, L.; Hakim, O.; Patel, V.R.; Baldi, P.; Hager, G.L.; Sassone-Corsi, P. Cycles in spatial and temporal chromosomal organization driven by the circadian clock. Nat. Struct. Mol. Biol. 2013, 20, 1206–1213. [Google Scholar] [CrossRef]
- Doi, M.; Hirayama, J.; Sassone-Corsi, P. Circadian regulator CLOCK is a histone acetyltransferase. Cell 2006, 125, 497–508. [Google Scholar] [CrossRef]
- Nakahata, Y.; Grimaldi, B.; Sahar, S.; Hirayama, J.; Sassone-Corsi, P. Signaling to the circadian clock: Plasticity by chromatin remodeling. Curr. Opin. Cell Biol. 2007, 19, 230–237. [Google Scholar] [CrossRef] [PubMed]
- Koike, N.; Yoo, S.H.; Huang, H.C.; Kumar, V.; Lee, C.; Kim, T.K.; Takahashi, J.S. Transcriptional architecture and chromatin landscape of the core circadian clock in mammals. Science 2012, 338, 349–354. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Etchegaray, J.P.; Lee, C.; Wade, P.A.; Reppert, S.M. Rhythmic histone acetylation underlies transcription in the mammalian circadian clock. Nature 2003, 421, 177–182. [Google Scholar] [CrossRef]
- Katada, S.; Sassone-Corsi, P. The histone methyltransferase MLL1 permits the oscillation of circadian gene expression. Nat. Struct. Mol. Biol. 2010, 17, 1414–1421. [Google Scholar] [CrossRef]
- DiTacchio, L.; Le, H.D.; Vollmers, C.; Hatori, M.; Witcher, M.; Secombe, J.; Panda, S. Histone lysine demethylase JARID1a activates CLOCK-BMAL1 and influences the circadian clock. Science 2011, 333, 1881–1885. [Google Scholar] [CrossRef] [Green Version]
- Etchegaray, J.P.; Yang, X.; DeBruyne, J.P.; Peters, A.; Weaver, D.R.; Jenuwein, T.; Reppert, S.M. The polycomb group protein EZH2 is required for mammalian circadian clock function. J. Biol. Chem. 2006, 281, 21209–21215. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jones, M.A.; Covington, M.F.; DiTacchio, L.; Vollmers, C.; Panda, S.; Harmer, S.L. Jumonji domain protein JMJD5 functions in both the plant and human circadian systems. Proc. Natl. Acad. Sci. USA 2010, 107, 21623–21628. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Asher, G.; Gatfield, D.; Stratmann, M.; Reinke, H.; Dibner, C.; Kreppel, F.; Mostoslavsky, R.; Alt, F.W.; Schibler, U. SIRT1 regulates circadian clock gene expression through PER2 deacetylation. Cell 2008, 134, 317–328. [Google Scholar] [CrossRef] [Green Version]
- Nakahata, Y.; Kaluzova, M.; Grimaldi, B.; Sahar, S.; Hirayama, J.; Chen, D.; Guarente, L.P.; Sassone-Corsi, P. The NAD+-dependent deacetylase SIRT1 modulates CLOCK-mediated chromatin remodeling and circadian control. Cell 2008, 134, 329–340. [Google Scholar] [CrossRef] [Green Version]
- Masri, S.; Rigor, P.; Cervantes, M.; Ceglia, N.; Sebastian, C.; Xiao, C.; Roqueta-Rivera, M.; Deng, C.; Osborne, T.F.; Mostoslavsky, R.; et al. Partitioning circadian transcription by SIRT6 leads to segregated control of cellular metabolism. Cell 2014, 158, 659–672. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Carrico, C.; Meyer, J.G.; He, W.; Gibson, B.W.; Verdin, E. The Mitochondrial Acylome Emerges: Proteomics, Regulation by Sirtuins, and Metabolic and Disease Implications. Cell Metab. 2018, 27, 497–512. [Google Scholar] [CrossRef] [Green Version]
- Roth, M.; Chen, W.Y. Sorting out functions of sirtuins in cancer. Oncogene 2014, 33, 1609–1620. [Google Scholar] [CrossRef] [Green Version]
- Ramadori, G.; Fujikawa, T.; Anderson, J.; Berglund, E.D.; Frazao, R.; Michán, S.; Vianna, C.R.; Sinclair, D.A.; Elias, C.F.; Coppari, R. SIRT1 deacetylase in SF1 neurons protects against metabolic imbalance. Cell Metab. 2011, 14, 301–312. [Google Scholar] [CrossRef] [Green Version]
- Ramadori, G.; Fujikawa, T.; Fukuda, M.; Anderson, J.; Morgan, D.A.; Mostoslavsky, R.; Stuart, R.C.; Perello, M.; Vianna, C.R.; Nillni, E.A.; et al. SIRT1 deacetylase in POMC neurons is required for homeostatic defenses against diet-induced obesity. Cell Metab. 2010, 12, 78–87. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chang, H.C.; Guarente, L. SIRT1 mediates central circadian control in the SCN by a mechanism that decays with aging. Cell 2013, 153, 1448–1460. [Google Scholar] [CrossRef] [Green Version]
- Hirayama, J.; Sahar, S.; Grimaldi, B.; Tamaru, T.; Takamatsu, K.; Nakahata, Y.; Sassone-Corsi, P. CLOCK-mediated acetylation of BMAL1 controls circadian function. Nature 2007, 450, 1086–1090. [Google Scholar] [CrossRef]
- Masri, S.; Patel, V.R.; Eckel-Mahan, K.L.; Peleg, S.; Forne, I.; Ladurner, A.G.; Baldi, P.; Imhof, A.; Sassone-Corsi, P. Circadian acetylome reveals regulation of mitochondrial metabolic pathways. Proc. Natl. Acad. Sci. USA 2013, 110, 3339–3344. [Google Scholar] [CrossRef] [Green Version]
- Peek, C.B.; Affinati, A.H.; Ramsey, K.M.; Kuo, H.Y.; Yu, W.; Sena, L.A.; Ilkayeva, O.; Marcheva, B.; Kobayashi, Y.; Omura, C.; et al. Circadian clock NAD+ cycle drives mitochondrial oxidative metabolism in mice. Science 2013, 342, 1243417. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tamaru, T.; Hattori, M.; Ninomiya, Y.; Kawamura, G.; Varès, G.; Honda, K.; Mishra, D.P.; Wang, B.; Benjamin, I.; Sassone-Corsi, P.; et al. ROS stress resets circadian clocks to coordinate pro-survival signals. PLoS ONE 2013, 8, e82006. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gomes, A.P.; Price, N.L.; Ling, A.J.; Moslehi, J.J.; Montgomery, M.K.; Rajman, L.; White, J.P.; Teodoro, J.S.; Wrann, C.D.; Hubbard, B.P.; et al. Declining NAD(+) induces a pseudohypoxic state disrupting nuclear-mitochondrial communication during aging. Cell 2013, 155, 1624–1638. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Laranjeiro, R.; Tamai, T.K.; Peyric, E.; Krusche, P.; Ott, S.; Whitmore, D. Cyclin-dependent kinase inhibitor p20 controls circadian cell-cycle timing. Proc. Natl. Acad. Sci. USA 2013, 110, 6835–6840. [Google Scholar] [CrossRef] [Green Version]
- Matsuo, T.; Yamaguchi, S.; Mitsui, S.; Emi, A.; Shimoda, F.; Okamura, H. Control mechanism of the circadian clock for timing of cell division in vivo. Science 2003, 302, 255–259. [Google Scholar] [CrossRef] [Green Version]
- Feillet, C.; Krusche, P.; Tamanini, F.; Janssens, R.C.; Downey, M.J.; Martin, P.; Teboul, M.; Saito, S.; Lévi, F.A.; Bretschneider, T.; et al. Phase locking and multiple oscillating attractors for the coupled mammalian clock and cell cycle. Proc. Natl. Acad. Sci. USA 2014, 111, 9828–9833. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kelleher, F.C.; Rao, A.; Maguire, A. Circadian molecular clocks and cancer. Cancer Lett. 2014, 342, 9–18. [Google Scholar] [CrossRef]
- Masri, S.; Cervantes, M.; Sassone-Corsi, P. The circadian clock and cell cycle: Interconnected biological circuits. Curr. Opin. Cell Biol. 2013, 25, 730–734. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hunt, T.; Sassone-Corsi, P. Riding tandem: Circadian clocks and the cell cycle. Cell 2007, 129, 461–464. [Google Scholar] [CrossRef] [Green Version]
- Kowalska, E.; Ripperger, J.A.; Hoegger, D.C.; Bruegger, P.; Buch, T.; Birchler, T.; Mueller, A.; Albrecht, U.; Contaldo, C.; Brown, S.A. NONO couples the circadian clock to the cell cycle. Proc. Natl. Acad. Sci. USA 2013, 110, 1592–1599. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Agarwal, M.L.; Agarwal, A.; Taylor, W.R.; Stark, G.R. p53 controls both the G2/M and the G1 cell cycle checkpoints and mediates reversible growth arrest in human fibroblasts. Proc. Natl. Acad. Sci. USA 1995, 92, 8493–8497. [Google Scholar] [CrossRef] [Green Version]
- Miki, T.; Matsumoto, T.; Zhao, Z.; Lee, C.C. p53 regulates Period2 expression and the circadian clock. Nat. Commun. 2013, 4, 2444. [Google Scholar] [CrossRef] [PubMed]
- Bouchard, C.; Thieke, K.; Maier, A.; Saffrich, R.; Hanley-Hyde, J.; Ansorge, W.; Reed, S.; Sicinski, P.; Bartek, J.; Eilers, M. Direct induction of cyclin D2 by Myc contributes to cell cycle progression and sequestration of p27. EMBO J. 1999, 18, 5321–5333. [Google Scholar] [CrossRef]
- Perez-Roger, I.; Kim, S.H.; Griffiths, B.; Sewing, A.; Land, H. Cyclins D1 and D2 mediate myc-induced proliferation via sequestration of p27(Kip1) and p21(Cip1). EMBO J. 1999, 18, 5310–5320. [Google Scholar] [CrossRef] [Green Version]
- Horvath, S.; Raj, K. DNA methylation-based biomarkers and the epigenetic clock theory of ageing. Nat. Rev. Genet. 2018, 19, 371–384. [Google Scholar] [CrossRef] [PubMed]
- López-Otín, C.; Blasco, M.A.; Partridge, L.; Serrano, M.; Kroemer, G. The hallmarks of aging. Cell 2013, 153, 1194–1217. [Google Scholar] [CrossRef] [Green Version]
- Teschendorff, A.E.; Menon, U.; Gentry-Maharaj, A.; Ramus, S.J.; Weisenberger, D.J.; Shen, H.; Campan, M.; Noushmehr, H.; Bell, C.G.; Maxwell, A.P.; et al. Age-dependent DNA methylation of genes that are suppressed in stem cells is a hallmark of cancer. Genome Res. 2010, 20, 440–446. [Google Scholar] [CrossRef] [Green Version]
- Lister, R.; Pelizzola, M.; Dowen, R.H.; Hawkins, R.D.; Hon, G.; Tonti-Filippini, J.; Nery, J.R.; Lee, L.; Ye, Z.; Ngo, Q.M.; et al. Human DNA methylomes at base resolution show widespread epigenomic differences. Nature 2009, 462, 315–322. [Google Scholar] [CrossRef] [Green Version]
- Meissner, A.; Gnirke, A.; Bell, G.W.; Ramsahoye, B.; Lander, E.S.; Jaenisch, R. Reduced representation bisulfite sequencing for comparative high-resolution DNA methylation analysis. Nucl. Acids Res. 2005, 33, 5868–5877. [Google Scholar] [CrossRef] [Green Version]
- Rose, N.R.; Klose, R.J. Understanding the relationship between DNA methylation and histone lysine methylation. Biochim. Biophys. Acta. 2014, 1839, 1362–1372. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Reddington, J.P.; Perricone, S.M.; Nestor, C.E.; Reichmann, J.; Youngson, N.A.; Suzuki, M.; Reinhardt, D.; Dunican, D.S.; Prendergast, J.G.; Mjoseng, H.; et al. Redistribution of H3K27me3 upon DNA hypomethylation results in de-repression of Polycomb target genes. Genome Biol. 2013, 14, R25. [Google Scholar] [CrossRef] [Green Version]
- Berger, S.L.; Sassone-Corsi, P. Metabolic Signaling to Chromatin. Cold Spring Harb. Perspect. Biol. 2016, 8. [Google Scholar] [CrossRef] [PubMed]
- Bocklandt, S.; Lin, W.; Sehl, M.E.; Sánchez, F.J.; Sinsheimer, J.S.; Horvath, S.; Vilain, E. Epigenetic predictor of age. PLoS ONE. 2011, 6, e14821. [Google Scholar] [CrossRef] [Green Version]
- Lu, A.T.; Fei, Z.; Haghani, A.; Robeck, T.; Zoller, J.; Li, C.Z.; Zhang, J.; Ablaeva, J.; Adams, D.; Almunia, J.; et al. Universal DNA methylation age across mammalian tissues. BioRxiv 2021. [Google Scholar] [CrossRef]
- Lu, A.T.; Quach, A.; Wilson, J.G.; Reiner, A.P.; Aviv, A.; Raj, K.; Hou, L.; Baccarelli, A.A.; Li, Y.; Stewart, J.D.; et al. DNA methylation GrimAge strongly predicts lifespan and healthspan. Aging 2019, 11, 303–327. [Google Scholar] [CrossRef]
- Lu, Y.; Brommer, B.; Tian, X.; Krishnan, A.; Meer, M.; Wang, C.; Vera, D.L.; Zeng, Q.; Yu, D.; Bonkowski, M.S.; et al. Reprogramming to recover youthful epigenetic information and restore vision. Nature 2020, 588, 124–129. [Google Scholar] [CrossRef]
- Petkovich, D.A.; Podolskiy, D.I.; Lobanov, A.V.; Lee, S.-G.; Miller, R.A.; Gladyshev, V.N. Using DNA Methylation Profiling to Evaluate Biological Age and Longevity Interventions. Cell Metab. 2017, 25, 954–960. [Google Scholar] [CrossRef] [Green Version]
- Bell, C.G.; Lowe, R.; Adams, P.D.; Baccarelli, A.A.; Beck, S.; Bell, J.T.; Christensen, B.C.; Gladyshev, V.N.; Heijmans, B.T.; Horvath, S.; et al. DNA methylation aging clocks: Challenges and recommendations. Genome Biol. 2019, 20, 249. [Google Scholar] [CrossRef] [Green Version]
- Gravina, S.; Dong, X.; Yu, B.; Vijg, J. Single-cell genome-wide bisulfite sequencing uncovers extensive heterogeneity in the mouse liver methylome. Genome Biol. 2016, 17, 150. [Google Scholar] [CrossRef] [Green Version]
- Guo, H.; Zhu, P.; Wu, X.; Li, X.; Wen, L.; Tang, F. Single-cell methylome landscapes of mouse embryonic stem cells and early embryos analyzed using reduced representation bisulfite sequencing. Genome Res. 2013, 23, 2126–2135. [Google Scholar] [CrossRef] [Green Version]
- Smallwood, S.A.; Lee, H.J.; Angermueller, C.; Krueger, F.; Saadeh, H.; Peat, J.; Andrews, S.R.; Stegle, O.; Reik, W.; Kelsey, G. Single-cell genome-wide bisulfite sequencing for assessing epigenetic heterogeneity. Nat. Methods 2014, 11, 817–820. [Google Scholar] [CrossRef] [PubMed]
- Trapp, A.; Kerepesi, C.; Gladyshev, V.N. Profiling epigenetic age in single cells. bioRxiv 2021. [Google Scholar] [CrossRef]
- Azzi, A.; Dallmann, R.; Casserly, A.; Rehrauer, H.; Patrignani, A.; Maier, B.; Kramer, A.; Brown, S.A. Circadian behavior is light-reprogrammed by plastic DNA methylation. Nat. Neurosci. 2014, 17, 377–382. [Google Scholar] [CrossRef] [PubMed]
- Ripperger, J.A.; Merrow, M. Perfect timing: Epigenetic regulation of the circadian clock. FEBS Lett. 2011, 585, 1406–1411. [Google Scholar] [CrossRef] [Green Version]
- Joska, T.M.; Zaman, R.; Belden, W.J. Regulated DNA methylation and the circadian clock: Implications in cancer. Biology 2014, 3, 560–577. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sharma, S.; Kelly, T.K.; Jones, P.A. Epigenetics in cancer. Carcinogenesis 2010, 31, 27–36. [Google Scholar] [CrossRef]
- Raj, K.; Horvath, S. Current perspectives on the cellular and molecular features of epigenetic ageing. Exp. Biol. Med. 2020, 245, 1532–1542. [Google Scholar] [CrossRef] [PubMed]
- Lehmann, M.; Canatelli-Mallat, M.; Chiavellini, P.; Goya, R.G. A hierarchical model for the control of epigenetic aging in mammals. Ageing Res. Rev. 2020, 62, 101134. [Google Scholar] [CrossRef]
- Fahy, G.M.; Brooke, R.T.; Watson, J.P.; Good, Z.; Vasanawala, S.S.; Maecker, H.; Leipold, M.D.; Lin, D.; Kobor, M.S.; Horvath, S. Reversal of epigenetic aging and immunosenescent trends in humans. Aging Cell 2019, 18, e13028. [Google Scholar] [CrossRef] [Green Version]
- Horvath, S.; Singh, K.; Raj, K.; Khairnar, S.; Sanghavi, A.; Shrivastava, A.; Zoller, J.A.; Li, C.Z.; Herenu, C.B.; Canatelli-Mallat, M.; et al. Reversing age: Dual species measurement of epigenetic age with a single clock. bioRxiv 2020. [Google Scholar] [CrossRef]
- Schultz, M.B.; Sinclair, D.A. When stem cells grow old: Phenotypes and mechanisms of stem cell aging. Development 2016, 143, 3–14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dykstra, B.; Olthof, S.; Schreuder, J.; Ritsema, M.; de Haan, G. Clonal analysis reveals multiple functional defects of aged murine hematopoietic stem cells. J. Exp. Med. 2011, 208, 2691–2703. [Google Scholar] [CrossRef] [Green Version]
- Liang, Y.; Van Zant, G.; Szilvassy, S.J. Effects of aging on the homing and engraftment of murine hematopoietic stem and progenitor cells. Blood 2005, 106, 1479–1487. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Beerman, I.; Bhattacharya, D.; Zandi, S.; Sigvardsson, M.; Weissman, I.L.; Bryder, D.; Rossi, D.J. Functionally distinct hematopoietic stem cells modulate hematopoietic lineage potential during aging by a mechanism of clonal expansion. Proc. Natl. Acad. Sci. USA 2010, 107, 5465–5470. [Google Scholar] [CrossRef] [Green Version]
- Jaiswal, S.; Fontanillas, P.; Flannick, J.; Manning, A.; Grauman, P.V.; Mar, B.G.; Lindsley, R.C.; Mermel, C.H.; Burtt, N.; Chavez, A.; et al. Age-related clonal hematopoiesis associated with adverse outcomes. N. Engl. J. Med. 2014, 371, 2488–2498. [Google Scholar] [CrossRef] [Green Version]
- Rossi, D.J.; Bryder, D.; Zahn, J.M.; Ahlenius, H.; Sonu, R.; Wagers, A.J.; Weissman, I.L. Cell intrinsic alterations underlie hematopoietic stem cell aging. Proc. Natl. Acad. Sci. USA 2005, 102, 9194–9199. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Linton, P.J.; Dorshkind, K. Age-related changes in lymphocyte development and function. Nat. Immunol. 2004, 5, 133–139. [Google Scholar] [CrossRef]
- Lichtman, M.A.; Rowe, J.M. The relationship of patient age to the pathobiology of the clonal myeloid diseases. Semin. Oncol. 2004, 31, 185–197. [Google Scholar] [CrossRef] [PubMed]
- Mareschi, K.; Ferrero, I.; Rustichelli, D.; Aschero, S.; Gammaitoni, L.; Aglietta, M.; Madon, E.; Fagioli, F. Expansion of mesenchymal stem cells isolated from pediatric and adult donor bone marrow. J. Cell Biochem. 2006, 97, 744–754. [Google Scholar] [CrossRef] [Green Version]
- Musina, R.A.; Bekchanova, E.S.; Sukhikh, G.T. Comparison of mesenchymal stem cells obtained from different human tissues. Bull. Exp. Biol. Med. 2005, 139, 504–509. [Google Scholar] [CrossRef]
- Bruder, S.P.; Jaiswal, N.; Haynesworth, S.E. Growth kinetics, self-renewal, and the osteogenic potential of purified human mesenchymal stem cells during extensive subcultivation and following cryopreservation. J. Cell Biochem. 1997, 64, 278–294. [Google Scholar] [CrossRef]
- Watanabe, S.; Kawamoto, S.; Ohtani, N.; Hara, E. Impact of senescence-associated secretory phenotype and its potential as a therapeutic target for senescence-associated diseases. Cancer Sci. 2017, 108, 563–569. [Google Scholar] [CrossRef] [Green Version]
- Noren Hooten, N.; Evans, M.K. Techniques to Induce and Quantify Cellular Senescence. J. Vis. Exp. 2017, 123, 55533. [Google Scholar] [CrossRef]
- Zhai, W.; Yong, D.; El-Jawhari, J.J.; Cuthbert, R.; McGonagle, D.; Win Naing, M.; Jones, E. Identification of senescent cells in multipotent mesenchymal stromal cell cultures: Current methods and future directions. Cytotherapy 2019, 21, 803–819. [Google Scholar] [CrossRef] [PubMed]
- Biteau, B.; Hochmuth, C.E.; Jasper, H. JNK activity in somatic stem cells causes loss of tissue homeostasis in the aging Drosophila gut. Cell Stem Cell 2008, 3, 442–455. [Google Scholar] [CrossRef] [Green Version]
- Choi, N.H.; Kim, J.G.; Yang, D.J.; Kim, Y.S.; Yoo, M.A. Age-related changes in Drosophila midgut are associated with PVF2, a PDGF/VEGF-like growth factor. Aging Cell 2008, 7, 318–334. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Takeda, N.; Jain, R.; LeBoeuf, M.R.; Wang, Q.; Lu, M.M.; Epstein, J.A. Interconversion between intestinal stem cell populations in distinct niches. Science 2011, 334, 1420–1424. [Google Scholar] [CrossRef] [Green Version]
- Martin, K.; Potten, C.S.; Roberts, S.A.; Kirkwood, T.B. Altered stem cell regeneration in irradiated intestinal crypts of senescent mice. J. Cell Sci. 1998, 111, 2297–2303. [Google Scholar] [CrossRef]
- Merlos-Suárez, A.; Barriga, F.M.; Jung, P.; Iglesias, M.; Céspedes, M.V.; Rossell, D.; Sevillano, M.; Hernando-Momblona, X.; da Silva-Diz, V.; Muñoz, P.; et al. The intestinal stem cell signature identifies colorectal cancer stem cells and predicts disease relapse. Cell Stem Cell 2011, 8, 511–524. [Google Scholar] [CrossRef] [Green Version]
- Sherwood, R.I.; Christensen, J.L.; Conboy, I.M.; Conboy, M.J.; Rando, T.A.; Weissman, I.L.; Wagers, A.J. Isolation of adult mouse myogenic progenitors: Functional heterogeneity of cells within and engrafting skeletal muscle. Cell 2004, 119, 543–554. [Google Scholar] [CrossRef] [Green Version]
- Beauchamp, J.R.; Morgan, J.E.; Pagel, C.N.; Partridge, T.A. Dynamics of myoblast transplantation reveal a discrete minority of precursors with stem cell-like properties as the myogenic source. J. Cell Biol. 1999, 144, 1113–1122. [Google Scholar] [CrossRef] [PubMed]
- Brack, A.S.; Bildsoe, H.; Hughes, S.M. Evidence that satellite cell decrement contributes to preferential decline in nuclear number from large fibres during murine age-related muscle atrophy. J. Cell Sci. 2005, 118, 4813–4821. [Google Scholar] [CrossRef] [Green Version]
- Collins, C.A.; Zammit, P.S.; Ruiz, A.P.; Morgan, J.E.; Partridge, T.A. A population of myogenic stem cells that survives skeletal muscle aging. Stem Cells 2007, 25, 885–894. [Google Scholar] [CrossRef] [PubMed]
- Bernet, J.D.; Doles, J.D.; Hall, J.K.; Kelly Tanaka, K.; Carter, T.A.; Olwin, B.B. p38 MAPK signaling underlies a cell-autonomous loss of stem cell self-renewal in skeletal muscle of aged mice. Nat. Med. 2014, 20, 265–271. [Google Scholar] [CrossRef]
- Cosgrove, B.D.; Gilbert, P.M.; Porpiglia, E.; Mourkioti, F.; Lee, S.P.; Corbel, S.Y.; Llewellyn, M.E.; Delp, S.L.; Blau, H.M. Rejuvenation of the muscle stem cell population restores strength to injured aged muscles. Nat. Med. 2014, 20, 255–264. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sousa-Victor, P.; Gutarra, S.; García-Prat, L.; Rodriguez-Ubreva, J.; Ortet, L.; Ruiz-Bonilla, V.; Jardí, M.; Ballestar, E.; González, S.; Serrano, A.L.; et al. Geriatric muscle stem cells switch reversible quiescence into senescence. Nature 2014, 506, 316–321. [Google Scholar] [CrossRef]
- Brack, A.S.; Conboy, M.J.; Roy, S.; Lee, M.; Kuo, C.J.; Keller, C.; Rando, T.A. Increased Wnt signaling during aging alters muscle stem cell fate and increases fibrosis. Science 2007, 317, 807–810. [Google Scholar] [CrossRef]
- Carlson, M.E.; Conboy, M.J.; Hsu, M.; Barchas, L.; Jeong, J.; Agrawal, A.; Mikels, A.J.; Agrawal, S.; Schaffer, D.V.; Conboy, I.M. Relative roles of TGF-beta1 and Wnt in the systemic regulation and aging of satellite cell responses. Aging Cell 2009, 8, 676–689. [Google Scholar] [CrossRef]
- Conboy, I.M.; Conboy, M.J.; Smythe, G.M.; Rando, T.A. Notch-mediated restoration of regenerative potential to aged muscle. Science 2003, 302, 1575–1577. [Google Scholar] [CrossRef]
- Sinha, M.; Jang, Y.C.; Oh, J.; Khong, D.; Wu, E.Y.; Manohar, R.; Miller, C.; Regalado, S.G.; Loffredo, F.S.; Pancoast, J.R.; et al. Restoring systemic GDF11 levels reverses age-related dysfunction in mouse skeletal muscle. Science 2014, 344, 649–652. [Google Scholar] [CrossRef] [Green Version]
- Price, F.D.; von Maltzahn, J.; Bentzinger, C.F.; Dumont, N.A.; Yin, H.; Chang, N.C.; Wilson, D.H.; Frenette, J.; Rudnicki, M.A. Inhibition of JAK-STAT signaling stimulates adult satellite cell function. Nat. Med. 2014, 20, 1174–1181. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jurkowski, M.P.; Bettio, L.; Woo, E.K.; Patten, A.; Yau, S.Y.; Gil-Mohapel, J. Beyond the Hippocampus and the SVZ: Adult Neurogenesis Throughout the Brain. Front. Cell Neurosci. 2020, 14, 576444. [Google Scholar] [CrossRef]
- Fares, J.; Bou Diab, Z.; Nabha, S.; Fares, Y. Neurogenesis in the adult hippocampus: History, regulation, and prospective roles. Int. J. Neurosci. 2019, 129, 598–611. [Google Scholar] [CrossRef]
- Maslov, A.Y.; Barone, T.A.; Plunkett, R.J.; Pruitt, S.C. Neural stem cell detection, characterization, and age-related changes in the subventricular zone of mice. J. Neurosci. 2004, 24, 1726–1733. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ahlenius, H.; Visan, V.; Kokaia, M.; Lindvall, O.; Kokaia, Z. Neural stem and progenitor cells retain their potential for proliferation and differentiation into functional neurons despite lower number in aged brain. J. Neurosci. 2009, 29, 4408–4419. [Google Scholar] [CrossRef] [PubMed]
- Katsimpardi, L.; Litterman, N.K.; Schein, P.A.; Miller, C.M.; Loffredo, F.S.; Wojtkiewicz, G.R.; Chen, J.W.; Lee, R.T.; Wagers, A.J.; Rubin, L.L. Vascular and neurogenic rejuvenation of the aging mouse brain by young systemic factors. Science 2014, 344, 630–634. [Google Scholar] [CrossRef] [Green Version]
- Pineda, J.R.; Daynac, M.; Chicheportiche, A.; Cebrian-Silla, A.; Sii Felice, K.; Garcia-Verdugo, J.M.; Boussin, F.D.; Mouthon, M.A. Vascular-derived TGF-β increases in the stem cell niche and perturbs neurogenesis during aging and following irradiation in the adult mouse brain. EMBO Mol. Med. 2013, 5, 548–562. [Google Scholar] [CrossRef]
- Villeda, S.A.; Plambeck, K.E.; Middeldorp, J.; Castellano, J.M.; Mosher, K.I.; Luo, J.; Smith, L.K.; Bieri, G.; Lin, K.; Berdnik, D.; et al. Young blood reverses age-related impairments in cognitive function and synaptic plasticity in mice. Nat. Med. 2014, 20, 659–663. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Okamoto, M.; Inoue, K.; Iwamura, H.; Terashima, K.; Soya, H.; Asashima, M.; Kuwabara, T. Reduction in paracrine Wnt3 factors during aging causes impaired adult neurogenesis. FASEB J. 2011, 25, 3570–3582. [Google Scholar] [CrossRef]
- Keyes, B.E.; Segal, J.P.; Heller, E.; Lien, W.H.; Chang, C.Y.; Guo, X.; Oristian, D.S.; Zheng, D.; Fuchs, E. Nfatc1 orchestrates aging in hair follicle stem cells. Proc. Natl. Acad. Sci. USA 2013, 110, E4950–E4959. [Google Scholar] [CrossRef] [Green Version]
- Giangreco, A.; Qin, M.; Pintar, J.E.; Watt, F.M. Epidermal stem cells are retained in vivo throughout skin aging. Aging Cell 2008, 7, 250–259. [Google Scholar] [CrossRef] [Green Version]
- Rittié, L.; Stoll, S.W.; Kang., S.; Voorhees., J.J.; Fisher, G.J. Hedgehog signaling maintains hair follicle stem cell phenotype in young and aged human skin. Aging Cell 2009, 8, 738–751. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nishimura, E.K. Melanocyte stem cells: A melanocyte reservoir in hair follicles for hair and skin pigmentation. Pigment. Cell Melanoma Res. 2011, 24, 401–410. [Google Scholar] [CrossRef]
- Inomata, K.; Aoto, T.; Binh, N.T.; Okamoto, N.; Tanimura, S.; Wakayama, T.; Iseki, S.; Hara, E.; Masunaga, T.; Shimizu, H.; et al. Genotoxic stress abrogates renewal of melanocyte stem cells by triggering their differentiation. Cell. 2009, 137, 1088–1099. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Paul, C.; Nagano, M.; Robaire, B. Aging results in molecular changes in an enriched population of undifferentiated rat spermatogonia. Biol. Reprod. 2013, 89, 147. [Google Scholar] [CrossRef] [Green Version]
- Zhang, X.; Ebata, K.T.; Robaire, B.; Nagano, M.C. Aging of male germ line stem cells in mice. Biol. Reprod. 2006, 74, 119–124. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Antonio-Rubio, N.R.; Porras-Gómez, T.J.; Moreno-Mendoza, N. Identification of cortical germ cells in adult ovaries from three phyllostomid bats: Artibeus jamaicensis, Glossophaga soricina and Sturnira lilium. Reprod. Fertil. Dev. 2013, 25, 825–836. [Google Scholar] [CrossRef] [PubMed]
- Inserra, P.I.; Leopardo, N.P.; Willis, M.A.; Freysselinard, A.L.; Vitullo, A.D. Quantification of healthy and atretic germ cells and follicles in the developing and post-natal ovary of the South American plains vizcacha, Lagostomus maximus: Evidence of continuous rise of the germinal reserve. Reproduction 2013, 147, 199–209. [Google Scholar] [CrossRef] [Green Version]
- Hernandez, S.F.; Vahidi, N.A.; Park, S.; Weitzel, R.P.; Tisdale, J.; Rueda, B.R.; Wolff, E.F. Characterization of extracellular DDX4- or Ddx4-positive ovarian cells. Nat. Med. 2015, 21, 1114–1116. [Google Scholar] [CrossRef]
- Zhang, Y.; Yang, Z.; Yang, Y.; Wang, S.; Shi, L.; Xie, W.; Sun, K.; Zou, K.; Wang, L.; Xiong, J.; et al. Production of transgenic mice by random recombination of targeted genes in female germline stem cells. J. Mol. Cell Biol. 2011, 3, 132–141. [Google Scholar] [CrossRef]
- White, Y.A.; Woods, D.C.; Takai, Y.; Ishihara, O.; Seki, H.; Tilly, J.L. Oocyte formation by mitotically active germ cells purified from ovaries of reproductive-age women. Nat. Med. 2012, 18, 413–421. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, H.; Liu, L.; Li, X.; Busayavalasa, K.; Shen, Y.; Hovatta, O.; Gustafsson, J.Å.; Liu, K. Life-long in vivo cell-lineage tracing shows that no oogenesis originates from putative germline stem cells in adult mice. Proc. Natl. Acad. Sci. USA 2014, 111, 17983–17988. [Google Scholar] [CrossRef] [Green Version]
- Zhang, H.; Panula, S.; Petropoulos, S.; Edsgärd, D.; Busayavalasa, K.; Liu, L.; Li, X.; Risal, S.; Shen, Y.; Shao, J.; et al. Adult human and mouse ovaries lack DDX4-expressing functional oogonial stem cells. Nat. Med. 2015, 21, 1116–1118. [Google Scholar] [CrossRef] [Green Version]
- Ferrón, S.R.; Marqués-Torrejón, M.A.; Mira, H.; Flores, I.; Taylor, K.; Blasco, M.A.; Fariñas, I. Telomere shortening in neural stem cells disrupts neuronal differentiation and neuritogenesis. J. Neurosci. 2009, 29, 14394–14407. [Google Scholar] [CrossRef]
- Flores, I.; Canela, A.; Vera, E.; Tejera, A.; Cotsarelis, G.; Blasco, M.A. The longest telomeres: A general signature of adult stem cell compartments. Genes Dev. 2008, 22, 654–667. [Google Scholar] [CrossRef] [Green Version]
- Mather, K.A.; Jorm, A.F.; Parslow, R.A.; Christensen, H. Is telomere length a biomarker of aging? A review. J. Gerontol. A Biol. Sci. Med. Sci. 2011, 66, 202–213. [Google Scholar] [CrossRef] [PubMed]
- Lu, A.T.; Seeboth, A.; Tsai, P.C.; Sun, D.; Quach, A.; Reiner, A.P.; Kooperberg, C.; Ferrucci, L.; Hou, L.; Baccarelli, A.A.; et al. DNA methylation-based estimator of telomere length. Aging 2019, 11, 5895–5923. [Google Scholar] [CrossRef] [PubMed]
- Medawar, P.B. An Unsolved Problem of Biology; HK Lewis: London, UK, 1952. [Google Scholar]
- Beerman, I.; Seita, J.; Inlay, M.A.; Weissman, I.L.; Rossi, D.J. Quiescent hematopoietic stem cells accumulate DNA damage during aging that is repaired upon entry into cell cycle. Cell Stem Cell 2014, 15, 37–50. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rübe, C.E.; Fricke, A.; Widmann, T.A.; Fürst, T.; Madry, H.; Pfreundschuh, M.; Rübe, C. Accumulation of DNA damage in hematopoietic stem and progenitor cells during human aging. PLoS ONE 2011, 6, e17487. [Google Scholar] [CrossRef]
- Flach, J.; Bakker, S.T.; Mohrin, M.; Conroy, P.C.; Pietras, E.M.; Reynaud, D.; Alvarez, S.; Diolaiti, M.E.; Ugarte, F.; Forsberg, E.C.; et al. Replication stress is a potent driver of functional decline in ageing haematopoietic stem cells. Nature 2014, 512, 198–202. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tomasetti, C.; Vogelstein, B. Cancer etiology. Variation in cancer risk among tissues can be explained by the number of stem cell divisions. Science 2015, 347, 78–81. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kanaar, R.; Wyman, C.; Rothstein, R. Quality control of DNA break metabolism: In the ‘end’, it’s a good thing. EMBO J. 2008, 27, 581–588. [Google Scholar] [CrossRef] [Green Version]
- Walter, D.; Lier, A.; Geiselhart, A.; Thalheimer, F.B.; Huntscha, S.; Sobotta, M.C.; Moehrle, B.; Brocks, D.; Bayindir, I.; Kaschutnig, P.; et al. Exit from dormancy provokes DNA-damage-induced attrition in haematopoietic stem cells. Nature 2015, 520, 549–552. [Google Scholar] [CrossRef] [PubMed]
- Cairns, J. Mutation selection and the natural history of cancer. Nature 1975, 255, 197–200. [Google Scholar] [CrossRef]
- Bufalino, M.R.; DeVeale, B.; van der Kooy, D. The asymmetric segregation of damaged proteins is stem cell-type dependent. J. Cell Biol. 2013, 201, 523–530. [Google Scholar] [CrossRef] [Green Version]
- Katajisto, P.; Döhla, J.; Chaffer, C.L.; Pentinmikko, N.; Marjanovic, N.; Iqbal, S.; Zoncu, R.; Chen, W.; Weinberg, R.A.; Sabatini, D.M. Stem cells. Asymmetric apportioning of aged mitochondria between daughter cells is required for stemness. Science 2015, 348, 340–343. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cheng, J.; Türkel, N.; Hemati, N.; Fuller, M.T.; Hunt, A.J.; Yamashita, Y.M. Centrosome misorientation reduces stem cell division during ageing. Nature 2008, 456, 599–604. [Google Scholar] [CrossRef] [Green Version]
- Köhler, A.; Schmithorst, V.; Filippi, M.D.; Ryan, M.A.; Daria, D.; Gunzer, M.; Geiger, H. Altered cellular dynamics and endosteal location of aged early hematopoietic progenitor cells revealed by time-lapse intravital imaging in long bones. Blood 2009, 114, 290–298. [Google Scholar] [CrossRef]
- Cerletti, M.; Jang, Y.C.; Finley, L.W.; Haigis, M.C.; Wagers, A.J. Short-term calorie restriction enhances skeletal muscle stem cell function. Cell Stem Cell 2012, 10, 515–519. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chen, J.; Astle, C.M.; Harrison, D.E. Hematopoietic senescence is postponed and hematopoietic stem cell function is enhanced by dietary restriction. Exp. Hematol. 2003, 31, 1097–1103. [Google Scholar] [CrossRef]
- Mair, W.; McLeod, C.J.; Wang, L.; Jones, D.L. Dietary restriction enhances germline stem cell maintenance. Aging Cell 2010, 9, 916–918. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yilmaz, Ö.H.; Katajisto, P.; Lamming, D.W.; Gültekin, Y.; Bauer-Rowe, K.E.; Sengupta, S.; Birsoy, K.; Dursun, A.; Yilmaz, V.O. mTORC1 in the Paneth cell niche couples intestinal stem-cell function to calorie intake. Nature 2012, 486, 490–495. [Google Scholar] [CrossRef] [Green Version]
- Mihaylova, M.M.; Sabatini, D.M.; Yilmaz, Ö.H. Dietary and metabolic control of stem cell function in physiology and cancer. Cell Stem Cell 2014, 14, 292–305. [Google Scholar] [CrossRef] [Green Version]
- Lamming, D.W.; Ye, L.; Katajisto, P.; Goncalves, M.D.; Saitoh, M.; Stevens, D.M.; Davis, J.G.; Salmon, A.B.; Richardson, A.; Ahima, R.S.; et al. Rapamycin-induced insulin resistance is mediated by mTORC2 loss and uncoupled from longevity. Science 2012, 335, 1638–1643. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kapahi, P.; Kaeberlein, M.; Hansen, M. Dietary restriction and lifespan: Lessons from invertebrate models. Ageing Res. Rev. 2017, 39, 3–14. [Google Scholar] [CrossRef] [PubMed]
- Khapre, R.V.; Kondratova, A.A.; Patel, S.; Dubrovsky, Y.; Wrobel, M.; Antoch, M.P.; Kondratov, R.V. BMAL1-dependent regulation of the mTOR signaling pathway delays aging. Aging 2014, 6, 48–57. [Google Scholar] [CrossRef] [Green Version]
- Bitto, A.; Ito, T.K.; Pineda, V.V.; LeTexier, N.J.; Huang, H.Z.; Sutlief, E.; Tung, H.; Vizzini, N.; Chen, B.; Smith, K.; et al. Transient rapamycin treatment can increase lifespan and healthspan in middle-aged mice. Elife 2016, 5, e16351. [Google Scholar] [CrossRef]
- Urfer, S.R.; Kaeberlein, T.L.; Mailheau, S.; Bergman, P.J.; Creevy, K.E.; Promislow, D.; Kaeberlein, M. A randomized controlled trial to establish effects of short-term rapamycin treatment in 24 middle-aged companion dogs. Geroscience 2017, 39, 117–127. [Google Scholar] [CrossRef]
- Mannick, J.B.; Morris, M.; Hockey, H.P.; Roma, G.; Beibel, M.; Kulmatycki, K.; Watkins, M.; Shavlakadze, T.; Zhou, W.; Quinn, D.; et al. TORC1 inhibition enhances immune function and reduces infections in the elderly. Sci. Transl. Med. 2018, 10, eaaq1564. [Google Scholar] [CrossRef] [Green Version]
- Renault, V.M.; Rafalski, V.A.; Morgan, A.A.; Salih, D.A.; Brett, J.O.; Webb, A.E.; Villeda, S.A.; Thekkat, P.U.; Guillerey, C.; Denko, N.C.; et al. FoxO3 regulates neural stem cell homeostasis. Cell Stem Cell 2009, 5, 527–539. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tothova, Z.; Kollipara, R.; Huntly, B.J.; Lee, B.H.; Castrillon, D.H.; Cullen, D.E.; McDowell, E.P.; Lazo-Kallanian, S.; Williams, I.R.; Sears, C.; et al. FoxOs are critical mediators of hematopoietic stem cell resistance to physiologic oxidative stress. Cell 2007, 128, 325–339. [Google Scholar] [CrossRef] [Green Version]
- Myant, K.B.; Cammareri, P.; McGhee, E.J.; Ridgway, R.A.; Huels, D.J.; Cordero, J.B.; Schwitalla, S.; Kalna, G.; Ogg, E.L.; Athineos, D.; et al. ROS production and NF-κB activation triggered by RAC1 facilitate WNT-driven intestinal stem cell proliferation and colorectal cancer initiation. Cell Stem Cell 2013, 12, 761–773. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tilly, J.L.; Sinclair, D.A. Germline energetics, aging, and female infertility. Cell Metab. 2013, 17, 838–850. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Beerman, I.; Bock, C.; Garrison, B.S.; Smith, Z.D.; Gu, H.; Meissner, A.; Rossi, D.J. Proliferation-dependent alterations of the DNA methylation landscape underlie hematopoietic stem cell aging. Cell Stem Cell 2013, 12, 413–425. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sun, D.; Luo, M.; Jeong, M.; Rodriguez, B.; Xia, Z.; Hannah, R.; Wang, H.; Le, T.; Faull, K.F.; Chen, R.; et al. Epigenomic profiling of young and aged HSCs reveals concerted changes during aging that reinforce self-renewal. Cell Stem Cell 2014, 14, 673–688. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Florian, M.C.; Dörr, K.; Niebel, A.; Daria, D.; Schrezenmeier, H.; Rojewski, M.; Filippi, M.D.; Hasenberg, A.; Gunzer, M.; Scharffetter-Kochanek, K.; et al. Cdc42 activity regulates hematopoietic stem cell aging and rejuvenation. Cell Stem Cell 2012, 10, 520–530. [Google Scholar] [CrossRef] [Green Version]
- Liu, L.; Cheung, T.H.; Charville, G.W.; Hurgo, B.M.; Leavitt, T.; Shih, J.; Brunet, A.; Rando, T.A. Chromatin modifications as determinants of muscle stem cell quiescence and chronological aging. Cell Rep. 2013, 4, 189–204. [Google Scholar] [CrossRef] [Green Version]
- Kofman, A.E.; Huszar, J.M.; Payne, C.J. Transcriptional analysis of histone deacetylase family members reveal similarities between differentiating and aging spermatogonial stem cells. Stem Cell Rev. Rep. 2013, 9, 59–64. [Google Scholar] [CrossRef] [Green Version]
- Takahashi, K.; Yamanaka, S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 2006, 126, 663–676. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Samoylova, E.M.; Baklaushev, V.P. Cell Reprogramming Preserving Epigenetic Age: Advantages and Limitations. Biochemistry 2020, 85, 1035–1047. [Google Scholar] [CrossRef]
- De Boni, L.; Gasparoni, G.; Haubenreich, C.; Tierling, S.; Schmitt, I.; Peitz, M.; Koch, P.; Walter, J.; Wüllner, U.; Brüstle, O. DNA methylation alterations in iPSC- and hESC-derived neurons: Potential implications for neurological disease modeling. Clin. Epigenetics 2018, 10, 13. [Google Scholar] [CrossRef]
- Rando, T.A.; Chang, H.Y. Aging, rejuvenation, and epigenetic reprogramming: Resetting the aging clock. Cell 2012, 148, 46–57. [Google Scholar] [CrossRef] [Green Version]
- Buganim, Y.; Faddah, D.A.; Cheng, A.W.; Itskovich, E.; Markoulaki, S.; Ganz, K.; Klemm, S.L.; van Oudenaarden, A.; Jaenisch, R. Single-cell expression analyses during cellular reprogramming reveal an early stochastic and a late hierarchic phase. Cell 2012, 150, 1209–1222. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Polo, J.M.; Anderssen, E.; Walsh, R.M.; Schwarz, B.A.; Nefzger, C.M.; Lim, S.M.; Borkent, M.; Apostolou, E.; Alaei, S.; Cloutier, J.; et al. A molecular roadmap of reprogramming somatic cells into iPS cells. Cell 2012, 151, 1617–1632. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hansson, J.; Rafiee, M.R.; Reiland, S.; Polo, J.M.; Gehring, J.; Okawa, S.; Huber, W.; Hochedlinger, K.; Krijgsveld, J. Highly coordinated proteome dynamics during reprogramming of somatic cells to pluripotency. Cell Rep. 2012, 2, 1579–1592. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Olova, N.; Simpson, D.J.; Marioni, R.E.; Chandra, T. Partial reprogramming induces a steady decline in epigenetic age before loss of somatic identity. Aging Cell 2019, 18, e12877. [Google Scholar] [CrossRef] [Green Version]
- Ocampo, A.; Reddy, P.; Martinez-Redondo, P.; Platero-Luengo, A.; Hatanaka, F.; Hishida, T.; Li, M.; Lam, D.; Kurita, M.; Beyret, E.; et al. In Vivo Amelioration of Age-Associated Hallmarks by Partial Reprogramming. Cell 2016, 167, 1719–1733. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sheng, C.; Jungverdorben, J.; Wiethoff, H.; Lin, Q.; Flitsch, L.J.; Eckert, D.; Hebisch, M.; Fischer, J.; Kesavan, J.; Weykopf, B.; et al. A stably self-renewing adult blood-derived induced neural stem cell exhibiting patternability and epigenetic rejuvenation. Nat. Commun. 2018, 9, 4047. [Google Scholar] [CrossRef]
- Marion, R.M.; Strati, K.; Li, H.; Tejera, A.; Schoeftner, S.; Ortega, S.; Serrano, M.; Blasco, M.A. Telomeres acquire embryonic stem cell characteristics in induced pluripotent stem cells. Cell Stem Cell 2009, 4, 141–154. [Google Scholar] [CrossRef] [Green Version]
- Suhr, S.T.; Chang, E.A.; Rodriguez, R.M.; Wang, K.; Ross, P.J.; Beyhan, Z.; Murthy, S.; Cibelli, J.B. Telomere dynamics in human cells reprogrammed to pluripotency. PLoS ONE 2009, 4, e8124. [Google Scholar] [CrossRef] [Green Version]
- Prigione, A.; Fauler, B.; Lurz, R.; Lehrach, H.; Adjaye, J. The senescence-related mitochondrial/oxidative stress pathway is repressed in human induced pluripotent stem cells. Stem Cells 2010, 28, 721–733. [Google Scholar] [CrossRef] [PubMed]
- Suhr, S.T.; Chang, E.A.; Tjong, J.; Alcasid, N.; Perkins, G.A.; Goissis, M.D.; Ellisman, M.H.; Perez, G.I.; Cibelli, J.B. Mitochondrial rejuvenation after induced pluripotency. PLoS ONE 2010, 5, e14095. [Google Scholar] [CrossRef] [Green Version]
- Prigione, A.; Hossini, A.M.; Lichtner, B.; Serin, A.; Fauler, B.; Megges, M.; Lurz, R.; Lehrach, H.; Makrantonaki, E.; Zouboulis, C.C.; et al. Mitochondrial-associated cell death mechanisms are reset to an embryonic-like state in aged donor-derived iPS cells harboring chromosomal aberrations. PLoS ONE 2011, 6, e27352. [Google Scholar] [CrossRef] [Green Version]
- Abad, M.; Mosteiro, L.; Pantoja, C.; Cañamero, M.; Rayon, T.; Ors, I.; Graña, O.; Megías, D.; Domínguez, O.; Martínez, D.; et al. Reprogramming in vivo produces teratomas and iPS cells with totipotency features. Nature 2013, 502, 340–345. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sarkar, T.J.; Quarta, M.; Mukherjee, S.; Colville, A.; Paine, P.; Doan, L.; Tran, C.M.; Chu, C.R.; Horvath, S.; Qi, L.S.; et al. Transient non-integrative expression of nuclear reprogramming factors promotes multifaceted amelioration of aging in human cells. Nat. Commun. 2020, 11, 1545. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gill, D.; Parry, A.; Santos, F.; Hernando-Herraez, I.; Stubbs, T.M.; Milagre, I.; Reik, W. Multi-omic rejuvenation of human cells by maturation phase transient reprogramming. bioRxiv 2021. [Google Scholar] [CrossRef]
- Foja, S.; Jung, M.; Harwardt, B.; Riemann, D.; Pelz-Ackermann, O.; Schroeder, I.S. Hypoxia supports reprogramming of mesenchymal stromal cells via induction of embryonic stem cell-specific microRNA-302 cluster and pluripotency-associated genes. Cell Reprogram. 2013, 15, 68–79. [Google Scholar] [CrossRef] [PubMed]
- Hsu, Y.C.; Wu, Y.T.; Tsai, C.L.; Wei, Y.H. Current understanding and future perspectives of the roles of sirtuins in the reprogramming and differentiation of pluripotent stem cells. Exp. Biol. Med. 2018, 243, 563–575. [Google Scholar] [CrossRef]
- Madrigal, M.; Rao, K.S.; Riordan, N.H. A review of therapeutic effects of mesenchymal stem cell secretions and induction of secretory modification by different culture methods. J. Transl. Med. 2014, 12, 260. [Google Scholar] [CrossRef] [Green Version]
- Childs, B.G.; Li, H.; van Deursen, J.M. Senescent cells: A therapeutic target for cardiovascular disease. J. Clin. Investig. 2018, 128, 1217–1228. [Google Scholar] [CrossRef]
- Landgraf, K.; Brunauer, R.; Lepperdinger, G.; Grubeck-Loebenstein, B. The suppressive effect of mesenchymal stromal cells on T cell proliferation is conserved in old age. Transpl. Immunol. 2011, 25, 167–172. [Google Scholar] [CrossRef]
- Zhang, J.; Lv, S.; Liu, X.; Song, B.; Shi, L. Umbilical Cord Mesenchymal Stem Cell Treatment for Crohn’s Disease: A Randomized Controlled Clinical Trial. Gut Liver 2018, 12, 73–78. [Google Scholar] [CrossRef]
- Al Demour, S.; Jafar, H.; Adwan, S.; AlSharif, A.; Alhawari, H.; Alrabadi, A.; Zayed, A.; Jaradat, A.; Awidi, A. Safety and Potential Therapeutic Effect of Two Intracavernous Autologous Bone Marrow Derived Mesenchymal Stem Cells injections in Diabetic Patients with Erectile Dysfunction: An Open Label Phase I Clinical Trial. Urol. Int. 2018, 101, 358–365. [Google Scholar] [CrossRef] [PubMed]
- Iacobaeus, E.; Kadri, N.; Lefsihane, K.; Boberg, E.; Gavin, C.; Törnqvist Andrén, A.; Lilja, A.; Brundin, L.; Blanc, K.L. Short and Long Term Clinical and Immunologic Follow up after Bone Marrow Mesenchymal Stromal Cell Therapy in Progressive Multiple Sclerosis-A Phase I Study. J. Clin. Med. 2019, 8, 2102. [Google Scholar] [CrossRef] [Green Version]
- Gyöngyösi, M.; Wojakowski, W.; Lemarchand, P.; Lunde, K.; Tendera, M.; Bartunek, J.; Marban, E.; Assmus, B.; Henry, T.D.; Traverse, J.H.; et al. Meta-Analysis of Cell-based CaRdiac stUdiEs (ACCRUE) in patients with acute myocardial infarction based on individual patient data. Circ. Res. 2015, 116, 1346–1360. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Abdelmohsen, K.; Gorospe, M. Noncoding RNA control of cellular senescence. Wiley Interdiscip. Rev. RNA 2015, 6, 615–629. [Google Scholar] [CrossRef]
- Ocansey, D.K.W.; Pei, B.; Yan, Y.; Qian, H.; Zhang, X.; Xu, W.; Mao, F. Improved therapeutics of modified mesenchymal stem cells: An update. J. Transl. Med. 2020, 18, 42. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhou, X.; Hong, Y.; Zhang, H.; Li, X. Mesenchymal Stem Cell Senescence and Rejuvenation: Current Status and Challenges. Front. Cell Dev. Biol. 2020, 8, 364. [Google Scholar] [CrossRef]
- Spitzhorn, L.S.; Megges, M.; Wruck, W.; Rahman, M.S.; Otte, J.; Degistirici, Ö.; Meisel, R.; Sorg, R.V.; Oreffo, R.; Adjaye, J. Human iPSC-derived MSCs (iMSCs) from aged individuals acquire a rejuvenation signature. Stem Cell Res. Ther. 2019, 10, 100. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Göbel, C.; Goetzke, R.; Eggermann, T.; Wagner, W. Interrupted reprogramming into induced pluripotent stem cells does not rejuvenate human mesenchymal stromal cells. Sci Rep. 2018, 8, 11676. [Google Scholar] [CrossRef] [Green Version]
- Fernandez-Rebollo, E.; Franzen, J.; Goetzke, R.; Hollmann, J.; Ostrowska, A.; Oliverio, M.; Sieben, T.; Rath, B.; Kornfeld, J.W.; Wagner, W. Senescence-Associated Metabolomic Phenotype in Primary and iPSC-Derived Mesenchymal Stromal Cells. Stem Cell Rep. 2020, 14, 201–209. [Google Scholar] [CrossRef] [Green Version]
- Liang, C.; Liu, Z.; Song, M.; Li, W.; Wu, Z.; Wang, Z.; Wang, Q.; Wang, S.; Yan, K.; Sun, L.; et al. Stabilization of heterochromatin by CLOCK promotes stem cell rejuvenation and cartilage regeneration. Cell Res. 2021, 31, 187–205. [Google Scholar] [CrossRef]
- Jiao, H.; Walczak, B.E.; Lee, M.S.; Lemieux, M.E.; Li, W.J. GATA6 regulates aging of human mesenchymal stem/stromal cells. Stem Cells 2021, 39, 62–77. [Google Scholar] [CrossRef] [PubMed]
- O’Kane, G.M.; Grünwald, B.T.; Jang, G.H.; Masoomian, M.; Picardo, S.; Grant, R.C.; Denroche, R.E.; Zhang, A.; Wang, Y.; Lam, B.; et al. GATA6 Expression Distinguishes Classical and Basal-like Subtypes in Advanced Pancreatic Cancer. Clin. Cancer Res. 2020, 26, 4901–4910. [Google Scholar] [CrossRef] [Green Version]
- Shen, W.; Niu, N.; Lawson, B.; Qi, L.; Zhang, J.; Li, T.; Zhang, H.; Liu, J. GATA6: A new predictor for prognosis in ovarian cancer. Hum. Pathol. 2019, 86, 163–169. [Google Scholar] [CrossRef] [PubMed]
- Fu, L.; Hu, Y.; Song, M.; Liu, Z.; Zhang, W.; Yu, F.X.; Wu, J.; Wang, S.; Izpisua Belmonte, J.C.; Chan, P.; et al. Up-regulation of FOXD1 by YAP alleviates senescence and osteoarthritis. PLoS Biol. 2019, 17, e3000201. [Google Scholar] [CrossRef]
- Deng, L.; Ren, R.; Liu, Z.; Song, M.; Li, J.; Wu, Z.; Ren, X.; Fu, L.; Li, W.; Zhang, W.; et al. Stabilizing heterochromatin by DGCR8 alleviates senescence and osteoarthritis. Nat. Commun. 2019, 10, 3329. [Google Scholar] [CrossRef] [Green Version]
- Ren, X.; Hu, B.; Song, M.; Ding, Z.; Dang, Y.; Liu, Z.; Zhang, W.; Ji, Q.; Ren, R.; Ding, J.; et al. Maintenance of Nucleolar Homeostasis by CBX4 Alleviates Senescence and Osteoarthritis. Cell Rep. 2019, 26, 3643–3656. [Google Scholar] [CrossRef] [Green Version]
- Kornicka, K.; Marycz, K.; Marędziak, M.; Tomaszewski, K.A.; Nicpoń, J. The effects of the DNA methyltranfserases inhibitor 5-Azacitidine on ageing, oxidative stress and DNA methylation of adipose derived stem cells. J. Cell Mol. Med. 2017, 21, 387–401. [Google Scholar] [CrossRef] [PubMed]
- So, A.Y.; Jung, J.W.; Lee, S.; Kim, H.S.; Kang, K.S. DNA methyltransferase controls stem cell aging by regulating BMI1 and EZH2 through microRNAs. PLoS ONE 2011, 6, e19503. [Google Scholar] [CrossRef] [Green Version]
- Gao, B.; Lin, X.; Jing, H.; Fan, J.; Ji, C.; Jie, Q.; Zheng, C.; Wang, D.; Xu, X.; Hu, Y.; et al. Local delivery of tetramethylpyrazine eliminates the senescent phenotype of bone marrow mesenchymal stromal cells and creates an anti-inflammatory and angiogenic environment in aging mice. Aging Cell 2018, 17, e12741. [Google Scholar] [CrossRef] [PubMed]
- Son, M.J.; Kwon, Y.; Son, T.; Cho, Y.S. Restoration of Mitochondrial NAD+ Levels Delays Stem Cell Senescence and Facilitates Reprogramming of Aged Somatic Cells. Stem Cells 2016, 34, 2840–2851. [Google Scholar] [CrossRef]
- Yang, M.; Teng, S.; Ma, C.; Yu, Y.; Wang, P.; Yi, C. Ascorbic acid inhibits senescence in mesenchymal stem cells through ROS and AKT/mTOR signaling. Cytotechnology 2018, 70, 1301–1313. [Google Scholar] [CrossRef]
- Park, S.Y.; Jeong, A.J.; Kim, G.Y.; Jo, A.; Lee, J.E.; Leem, S.H.; Yoon, J.H.; Ye, S.K.; Chung, J.W. Lactoferrin Protects Human Mesenchymal Stem Cells from Oxidative Stress-Induced Senescence and Apoptosis. J. Microbiol. Biotechnol. 2017, 27, 1877–1884. [Google Scholar] [CrossRef] [Green Version]
- Lee, J.H.; Jung, H.K.; Han, Y.S.; Yoon, Y.M.; Yun, C.W.; Sun, H.Y.; Cho, H.W.; Lee, S.H. Antioxidant effects of Cirsium setidens extract on oxidative stress in human mesenchymal stem cells. Mol. Med. Rep. 2016, 14, 3777–3784. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lee, J.H.; Yoon, Y.M.; Song, K.H.; Noh, H.; Lee, S.H. Melatonin suppresses senescence-derived mitochondrial dysfunction in mesenchymal stem cells via the HSPA1L-mitophagy pathway. Aging Cell 2020, 19, e13111. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Seok, J.; Jung, H.S.; Park, S.; Lee, J.O.; Kim, C.J.; Kim, G.J. Alteration of fatty acid oxidation by increased CPT1A on replicative senescence of placenta-derived mesenchymal stem cells. Stem Cell Res. Ther. 2020, 11, 1. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Li, X.; Hong, Y.; He, H.; Jiang, G.; You, W.; Liang, X.; Fu, Q.; Han, S.; Lian, Q.; Zhang, Y. FGF21 Mediates Mesenchymal Stem Cell Senescence via Regulation of Mitochondrial Dynamics. Oxid Med. Cell Longev. 2019, 2019, 4915149. [Google Scholar] [CrossRef]
- Khorraminejad-Shirazi, M.; Sani, M.; Talaei-Khozani, T.; Dorvash, M.; Mirzaei, M.; Faghihi, M.A.; Monabati, A.; Attar, A. AICAR and nicotinamide treatment synergistically augment the proliferation and attenuate senescence-associated changes in mesenchymal stromal cells. Stem Cell Res. Ther. 2020, 11, 45. [Google Scholar] [CrossRef] [PubMed]
- Cohen, H.Y.; Miller, C.; Bitterman, K.J.; Wall, N.R.; Hekking, B.; Kessler, B.; Howitz, K.T.; Gorospe, M.; de Cabo, R.; Sinclair, D.A. Calorie restriction promotes mammalian cell survival by inducing the SIRT1 deacetylase. Science 2004, 305, 390–392. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Someya, S.; Yu, W.; Hallows, W.C.; Xu, J.; Vann, J.M.; Leeuwenburgh, C.; Tanokura, M.; Denu, J.M.; Prolla, T.A. Sirt3 mediates reduction of oxidative damage and prevention of age-related hearing loss under caloric restriction. Cell 2010, 143, 802–812. [Google Scholar] [CrossRef] [Green Version]
- Bonkowski, M.S.; Sinclair, D.A. Slowing ageing by design: The rise of NAD+ and sirtuin-activating compounds. Nat. Rev. Mol. Cell Biol. 2016, 17, 679–690. [Google Scholar] [CrossRef]
- Sun, S.; Qin, W.; Tang, X.; Meng, Y.; Hu, W.; Zhang, S.; Qian, M.; Liu, Z.; Cao, X.; Pang, Q.; et al. Vascular endothelium-targeted Sirt7 gene therapy rejuvenates blood vessels and extends life span in a Hutchinson-Gilford progeria model. Sci. Adv. 2020, 6, eaay5556. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, Y.; Xu, J.; Liu, S.; Lim, M.; Zhao, S.; Cui, K.; Zhang, K.; Wang, L.; Ji, Q.; Han, Z.; et al. Embryonic stem cell-derived extracellular vesicles enhance the therapeutic effect of mesenchymal stem cells. Theranostics 2019, 9, 6976–6990. [Google Scholar] [CrossRef]
- Lu, Y.; Qu, H.; Qi, D.; Xu, W.; Liu, S.; Jin, X.; Song, P.; Guo, Y.; Jia, Y.; Wang, X.; et al. OCT4 maintains self-renewal and reverses senescence in human hair follicle mesenchymal stem cells through the downregulation of p21 by DNA methyltransferases. Stem Cell Res. Ther. 2019, 10, 28. [Google Scholar] [CrossRef]
- Mistriotis, P.; Bajpai, V.K.; Wang, X.; Rong, N.; Shahini, A.; Asmani, M.; Liang, M.S.; Wang, J.; Lei, P.; Liu, S.; et al. NANOG Reverses the Myogenic Differentiation Potential of Senescent Stem Cells by Restoring ACTIN Filamentous Organization and SRF-Dependent Gene Expression. Stem Cells. 2017, 35, 207–221. [Google Scholar] [CrossRef] [PubMed]
- Rong, N.; Mistriotis, P.; Wang, X.; Tseropoulos, G.; Rajabian, N.; Zhang, Y.; Wang, J.; Liu, S.; Andreadis, S.T. Restoring extracellular matrix synthesis in senescent stem cells. FASEB J. 2019, 33, 10954–10965. [Google Scholar] [CrossRef] [Green Version]
- Chen, B.; Sun, Y.; Zhang, J.; Zhu, Q.; Yang, Y.; Niu, X.; Deng, Z.; Li, Q.; Wang, Y. Human embryonic stem cell-derived exosomes promote pressure ulcer healing in aged mice by rejuvenating senescent endothelial cells. Stem Cell Res. Ther. 2019, 10, 142. [Google Scholar] [CrossRef] [Green Version]
- Khanh, V.C.; Yamashita, T.; Ohneda, K.; Tokunaga, C.; Kato, H.; Osaka, M.; Hiramatsu, Y.; Ohneda, O. Rejuvenation of mesenchymal stem cells by extracellular vesicles inhibits the elevation of reactive oxygen species. Sci. Rep. 2020, 10, 17315. [Google Scholar] [CrossRef] [PubMed]
Basic Molecular Factors | Role in Control | Reference |
---|---|---|
Basic Molecular Factors Controlling Circadian Rhythms | ||
CLOCK | Central proteins of the mammalian clockworks; serve as the major transcription activators of b-HLH-PAS (The basic helix–loop–helix/Per-ARNT-SIM) expression, which in turn modulates the transcription of multiple clock-controlled genes (CCGs). | [10] |
BMAL1 | Central proteins of the mammalian clockworks; serve as the major transcription activators of b-HLH-PAS (The basic helix–loop–helix/Per-ARNT-SIM) expression, which in turn modulates the transcription of multiple clock-controlled genes (CCGs). | [10] |
PER | Forms a transcription-translation negative feedback loop for CLOCK/ BMAL1 complex. Translated from the three mammalian homologs of drosophila-per, one of three PER proteins (PER1, PER2, and PER3) dimerizes via its PAS domain with one of two cryptochrome proteins (CRY1 and CRY2) to form a negative element of the clock. This PER/CRY complex moves into the nucleus upon phosphorylation by CK1-epsilon (casein kinase 1 epsilon) and inhibits the CLOCK/ BMAL1 heterodimer, the transcription factor that is bound to the E-boxes of the three per and two cry promoters by basic helix-loop-helix (BHLH) DNA-binding domains. | [35,59,68] |
CRY | Cryptochrome; one of the four groups of mammalian clock genes/proteins that generate a transcription-translation negative-feedback loop (TTFL), along with Period (PER), CLOCK, and BMAL1. In this loop, CLOCK and BMAL1 proteins are transcriptional activators which bind together to the promoters of the Cry and Per genes and activate their transcription. The CRY and PER proteins then bind to each other, enter the nucleus, and inhibit CLOCK-BMAL1-activated transcription. | [35] |
REV-ERBα/β | Occupies the promoter elements of Reb-Erb/ROR. Rev-Erbα plays an important role in regulation of the core circadian clock through repression of the positive clock element Bmal1.The secondary TTFL, featuring Rev-Erbα working in conjunction with Rev-Erbβ and the orphan receptor RORα, is thought to strengthen this primary TTFL by further regulating BMAL1. | [36] |
RORα | Occupies the promoter elements of Reb-Erb/ROR. RORα shares the same response elements as Rev-Erbα but exerts opposite effects on gene transcription; BMAL1 expression is repressed by Rev-Erbα and activated by RORα. | [36] |
p300 | Functions as the co-activator of CLOCK-BMAL1 and launches the cyclic acetylation of histone tails. | [43,44,45] |
CBP | Operates similarly to p300, and has been described to interact with PER2 to repress CLOCK-BMAL1 activity. | [43,44,45] |
MLL1 | Tri-methylates histone H3K4 on the nucleosomes near the circadian gene promoters, which results in transcription activation. | [46,47] |
Jarid1A | A major component of the circadian clock, the upregulation of which at the end of the sleep phase blocks HDAC1 activity. Blocking HDAC1 activity results in an upregulation of CLOCK and BMAL1 and consequent upregulation of PER proteins. The PSF (polypyrimidine tract-binding protein-associated splicing factor) within the PER complex recruits SIN3A, a scaffold for assembly of transcriptional inhibitory complexes, and rhythmically delivers histone deacetylases to the Per1 promoter, which repress Per1 transcription. | [47] |
EZH2 | The catalytic subunit of the Polycomb Repressive Complex 2 (PRC2). As a histone methyltransferase (HMTase), EZH2’s primary function is to methylate Lys-27 on histone 3 (H3K27me) by transferring a methyl group from the cofactor S-adenosyl-L-methionine (SAM). | [48] |
Jmjd5 | Contains a jumonji-C (jmjC) domain that is often found in proteins with histone demethylase activity; indeed, its human ortholog (also known as KDM8) has recently been shown to have this function. Jmjd5 is coregulated with evening-phased clock components and positively affects expression of clock genes expressed at dawn. | [49] |
SIRT1 | Was found to deacetylate a number of proteins downstream of Myc, p53, HIF, TGF-β, and Wnt signaling, and acts as a tumor suppressor or a tumor-promoting factor, depending on the specific biological context. Also, SIRT1 is involved in the adaptation of the organism to nutritional starvation. SIRT1 is required for VMH and ARC neurons to control glucose metabolism and lipid exchange on the periphery; deregulation of these processes is associated with aging. In the context of SCN, SIRT1 modulates the activity of the central pacemaker. | [51,52,53,54,55,56,57,58,62] |
SIRT6 | Is mainly known as a deacetylase of histones H3 and H4, an activity by which it changes chromatin density and regulates gene expression. The enzymatic activity of Sirt6, as well as of the other members of the sirtuins family, is dependent upon the binding of the cofactor nicotinamide adenine dinucleotide (NAD+). | [52] |
SIRT3 | Is believed to set the acetylation rhythms as well as OXPHOS oscillations in liver mitochondria. Overexpression of SIRT3 in cultured cells increases respiration and decreases the production of reactive oxygen species. Activation of the NMNAT2 enzyme, which catalyzes an essential step in the nicotinamide adenine dinucleotide (NAD+) biosynthetic pathway by SIRT3, may be a means of inhibiting axon degeneration and dysfunction. Activation of SIRT3 inhibits the apoptosis leading to age-related macular degeneration. | [60] |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2021 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
Samoilova, E.M.; Belopasov, V.V.; Ekusheva, E.V.; Zhang, C.; Troitskiy, A.V.; Baklaushev, V.P. Epigenetic Clock and Circadian Rhythms in Stem Cell Aging and Rejuvenation. J. Pers. Med. 2021, 11, 1050. https://doi.org/10.3390/jpm11111050
Samoilova EM, Belopasov VV, Ekusheva EV, Zhang C, Troitskiy AV, Baklaushev VP. Epigenetic Clock and Circadian Rhythms in Stem Cell Aging and Rejuvenation. Journal of Personalized Medicine. 2021; 11(11):1050. https://doi.org/10.3390/jpm11111050
Chicago/Turabian StyleSamoilova, Ekaterina M., Vladimir V. Belopasov, Evgenia V. Ekusheva, Chao Zhang, Alexander V. Troitskiy, and Vladimir P. Baklaushev. 2021. "Epigenetic Clock and Circadian Rhythms in Stem Cell Aging and Rejuvenation" Journal of Personalized Medicine 11, no. 11: 1050. https://doi.org/10.3390/jpm11111050
APA StyleSamoilova, E. M., Belopasov, V. V., Ekusheva, E. V., Zhang, C., Troitskiy, A. V., & Baklaushev, V. P. (2021). Epigenetic Clock and Circadian Rhythms in Stem Cell Aging and Rejuvenation. Journal of Personalized Medicine, 11(11), 1050. https://doi.org/10.3390/jpm11111050