Changes in Regenerative Capacity through Lifespan
Abstract
:1. Introduction
2. Changes in Regenerative Ability through Phylogeny, Ontogeny and Aging
2.1. Variations during Development
2.2. Variations during Aging
2.3. The Exceptions to the Rule
3. Factors that Affect Regenerative Capacity during Aging
3.1. Genomic Instability
3.2. Telomere Shortening
3.3. Mitochondrial Dysfunction and Oxidative Damage
3.4. Epigenetic Modifications
3.5. Alterations in Proteostasis
3.6. Metabolic Changes
3.7. Extrinsic Signals: Local, Systemic and Blood-Borne Factors
4. A Link between Cellular Senescence, Aging and Regenerative Capacity
5. Avenues for Therapeutic Interventions
6. Conclusions
Acknowledgments
Conflicts of Interest
References
- Sharpless, N.E.; DePinho, R.A. Telomeres, stem cells, senescence, and cancer. J. Clin. Investig. 2004, 113, 160–168. [Google Scholar] [CrossRef] [PubMed]
- Finch, C.E. Longevity, Senescence and the Genome; The University of Chicago Press: Chicago, IL, USA, 1994. [Google Scholar]
- Lopez-Otin, C.; Blasco, M.A.; Partridge, L.; Serrano, M.; Kroemer, G. The hallmarks of aging. Cell 2013, 153, 1194–1217. [Google Scholar] [CrossRef] [PubMed]
- Seifert, A.W.; Voss, S.R. Revisiting the relationship between regenerative ability and aging. BMC Biol. 2013, 11, 2. [Google Scholar] [CrossRef] [PubMed]
- Porrello, E.R.; Mahmoud, A.I.; Simpson, E.; Hill, J.A.; Richardson, J.A.; Olson, E.N.; Sadek, H.A. Transient regenerative potential of the neonatal mouse heart. Science 2011, 331, 1078–1080. [Google Scholar] [CrossRef] [PubMed]
- Tanaka, E.M.; Ferretti, P. Considering the evolution of regeneration in the central nervous system. Nat. Rev. Neurosci. 2009, 10, 713–723. [Google Scholar] [CrossRef] [PubMed]
- Taylor, R.R.; Forge, A. Hair cell regeneration in sensory epithelia from the inner ear of a urodele amphibian. J. Comp. Neurol. 2005, 484, 105–120. [Google Scholar] [CrossRef] [PubMed]
- Walters, B.J.; Zuo, J. Postnatal development, maturation and aging in the mouse cochlea and their effects on hair cell regeneration. Hear. Res. 2013, 297, 68–83. [Google Scholar] [CrossRef] [PubMed]
- Borgens, R.B. Mice regrow the tips of their foretoes. Science 1982, 217, 747–750. [Google Scholar] [CrossRef] [PubMed]
- Reginelli, A.D.; Wang, Y.Q.; Sassoon, D.; Muneoka, K. Digit tip regeneration correlates with regions of msx1 (hox 7) expression in fetal and newborn mice. Development 1995, 121, 1065–1076. [Google Scholar] [PubMed]
- Monaghan, J.R.; Stier, A.C.; Michonneau, F.; Smith, M.D.; Pasch, B.; Maden, M.; Seifert, A.W. Experimentally induced metamorphosis in axolotls reduces regenerative rate and fidelity. Regeneration 2014, 1, 2–14. [Google Scholar] [CrossRef]
- Suetsugu-Maki, R.; Maki, N.; Nakamura, K.; Sumanas, S.; Zhu, J.; del Rio-Tsonis, K.; Tsonis, P.A. Lens regeneration in axolotl: New evidence of developmental plasticity. BMC Biol. 2012, 10, 103. [Google Scholar] [CrossRef] [PubMed]
- Sahin, E.; Depinho, R.A. Linking functional decline of telomeres, mitochondria and stem cells during ageing. Nature 2010, 464, 520–528. [Google Scholar] [CrossRef] [PubMed]
- Sousounis, K.; Baddour, J.A.; Tsonis, P.A. Aging and regeneration in vertebrates. Curr. Top. Dev. Biol. 2014, 108, 217–246. [Google Scholar] [PubMed]
- Beggs, M.L.; Nagarajan, R.; Taylor-Jones, J.M.; Nolen, G.; Macnicol, M.; Peterson, C.A. Alterations in the TGFbeta signaling pathway in myogenic progenitors with age. Aging Cell 2004, 3, 353–361. [Google Scholar] [CrossRef] [PubMed]
- Gibson, M.C.; Schultz, E. Age-related differences in absolute numbers of skeletal muscle satellite cells. Muscle Nerve 1983, 6, 574–580. [Google Scholar] [CrossRef] [PubMed]
- Conboy, I.M.; Rando, T.A. Heterochronic parabiosis for the study of the effects of aging on stem cells and their niches. Cell Cycle 2012, 11, 2260–2267. [Google Scholar] [CrossRef] [PubMed]
- Chambers, S.M.; Goodell, M.A. Hematopoietic stem cell aging: Wrinkles in stem cell potential. Stem Cell Rev. 2007, 3, 201–211. [Google Scholar] [CrossRef] [PubMed]
- Chambers, S.M.; Shaw, C.A.; Gatza, C.; Fisk, C.J.; Donehower, L.A.; Goodell, M.A. Aging hematopoietic stem cells decline in function and exhibit epigenetic dysregulation. PLoS Biol. 2007, 5, 201. [Google Scholar] [CrossRef] [PubMed]
- De Haan, G.; Nijhof, W.; van Zant, G. Mouse strain-dependent changes in frequency and proliferation of hematopoietic stem cells during aging: Correlation between lifespan and cycling activity. Blood 1997, 89, 1543–1550. [Google Scholar] [PubMed]
- Rossi, D.J.; Bryder, D.; Seita, J.; Nussenzweig, A.; Hoeijmakers, J.; Weissman, I.L. Deficiencies in DNA damage repair limit the function of haematopoietic stem cells with age. Nature 2007, 447, 725–729. [Google Scholar] [CrossRef] [PubMed]
- Zheng, H.; Martin, J.A.; Duwayri, Y.; Falcon, G.; Buckwalter, J.A. Impact of aging on rat bone marrow-derived stem cell chondrogenesis. J. Gerontol. A 2007, 62, 136–148. [Google Scholar] [CrossRef]
- Stenderup, K.; Justesen, J.; Clausen, C.; Kassem, M. Aging is associated with decreased maximal life span and accelerated senescence of bone marrow stromal cells. Bone 2003, 33, 919–926. [Google Scholar] [CrossRef] [PubMed]
- Zhou, S.; Greenberger, J.S.; Epperly, M.W.; Goff, J.P.; Adler, C.; Leboff, M.S.; Glowacki, J. Age-related intrinsic changes in human bone-marrow-derived mesenchymal stem cells and their differentiation to osteoblasts. Aging Cell 2008, 7, 335–343. [Google Scholar] [CrossRef] [PubMed]
- Lee-Liu, D.; Edwards-Faret, G.; Tapia, V.S.; Larrain, J. Spinal cord regeneration: Lessons for mammals from non-mammalian vertebrates. Genesis 2013, 51, 529–544. [Google Scholar] [CrossRef] [PubMed]
- Mattson, M.P.; Magnus, T. Ageing and neuronal vulnerability. Nat. Rev. Neurosci. 2006, 7, 278–294. [Google Scholar] [CrossRef] [PubMed]
- He, L.; Yadgarov, A.; Sharif, S.; McCluskey, L.P. Aging profoundly delays functional recovery from gustatory nerve injury. Neuroscience 2012, 209, 208–218. [Google Scholar] [CrossRef] [PubMed]
- Dor, Y.; Brown, J.; Martinez, O.I.; Melton, D.A. Adult pancreatic beta-cells are formed by self-duplication rather than stem-cell differentiation. Nature 2004, 429, 41–46. [Google Scholar] [CrossRef] [PubMed]
- Chera, S.; Baronnier, D.; Ghila, L.; Cigliola, V.; Jensen, J.N.; Gu, G.; Furuyama, K.; Thorel, F.; Gribble, F.M.; Reimann, F.; et al. Diabetes recovery by age-dependent conversion of pancreatic delta-cells into insulin producers. Nature 2014, 514, 503–507. [Google Scholar] [CrossRef] [PubMed]
- Thorel, F.; Nepote, V.; Avril, I.; Kohno, K.; Desgraz, R.; Chera, S.; Herrera, P.L. Conversion of adult pancreatic alpha-cells to beta-cells after extreme beta-cell loss. Nature 2010, 464, 1149–1154. [Google Scholar] [CrossRef] [PubMed]
- Rankin, M.M.; Kushner, J.A. Adaptive beta-cell proliferation is severely restricted with advanced age. Diabetes 2009, 58, 1365–1372. [Google Scholar] [CrossRef] [PubMed]
- Chen, H.; Gu, X.; Su, I.H.; Bottino, R.; Contreras, J.L.; Tarakhovsky, A.; Kim, S.K. Polycomb protein ezh2 regulates pancreatic beta-cell ink4a/arf expression and regeneration in diabetes mellitus. Genes Dev. 2009, 23, 975–985. [Google Scholar] [CrossRef] [PubMed]
- Dhawan, S.; Tschen, S.I.; Bhushan, A. Bmi-1 regulates the ink4a/arf locus to control pancreatic beta-cell proliferation. Genes Dev. 2009, 23, 906–911. [Google Scholar] [CrossRef] [PubMed]
- Avrahami, D.; Li, C.; Zhang, J.; Schug, J.; Avrahami, R.; Rao, S.; Stadler, M.B.; Burger, L.; Schubeler, D.; Glaser, B.; et al. Aging-dependent demethylation of regulatory elements correlates with chromatin state and improved beta cell function. Cell Metab. 2015, 22, 619–632. [Google Scholar] [CrossRef] [PubMed]
- Heiss, C.; Keymel, S.; Niesler, U.; Ziemann, J.; Kelm, M.; Kalka, C. Impaired progenitor cell activity in age-related endothelial dysfunction. J. Am. Coll. Cardiol. 2005, 45, 1441–1448. [Google Scholar] [CrossRef] [PubMed]
- Rauscher, F.M.; Goldschmidt-Clermont, P.J.; Davis, B.H.; Wang, T.; Gregg, D.; Ramaswami, P.; Pippen, A.M.; Annex, B.H.; Dong, C.; Taylor, D.A. Aging, progenitor cell exhaustion, and atherosclerosis. Circulation 2003, 108, 457–463. [Google Scholar] [CrossRef] [PubMed]
- Galliot, B.; Chera, S. The hydra model: Disclosing an apoptosis-driven generator of wnt-based regeneration. Trends Cell Biol. 2010, 20, 514–523. [Google Scholar] [CrossRef] [PubMed]
- Newmark, P.A.; Sanchez Alvarado, A. Not your father’s planarian: A classic model enters the era of functional genomics. Nat. Rev. Genet. 2002, 3, 210–219. [Google Scholar] [CrossRef] [PubMed]
- Watanabe, H.; Hoang, V.T.; Mattner, R.; Holstein, T.W. Immortality and the base of multicellular life: Lessons from cnidarian stem cells. Semin. Cell Dev. Biol. 2009, 20, 1114–1125. [Google Scholar] [CrossRef] [PubMed]
- Brøndsted, H.V. An Excellent Compendium of Experiments Carried Out on Planarians with an Extensive Bibliography of the Classic Literature; Pergamon: London, UK, 1969. [Google Scholar]
- Gemberling, M.; Bailey, T.J.; Hyde, D.R.; Poss, K.D. The zebrafish as a model for complex tissue regeneration. Trends Genet. 2013, 29, 611–620. [Google Scholar] [CrossRef] [PubMed]
- Kizil, C.; Kaslin, J.; Kroehne, V.; Brand, M. Adult neurogenesis and brain regeneration in zebrafish. Dev. Neurobiol. 2012, 72, 429–461. [Google Scholar] [CrossRef] [PubMed]
- Itou, J.; Kawakami, H.; Burgoyne, T.; Kawakami, Y. Life-long preservation of the regenerative capacity in the fin and heart in zebrafish. Biol. Open 2012, 1, 739–746. [Google Scholar] [CrossRef] [PubMed]
- Shao, J.; Chen, D.; Ye, Q.; Cui, J.; Li, Y.; Li, L. Tissue regeneration after injury in adult zebrafish: The regenerative potential of the caudal fin. Dev. Dyn. 2011, 240, 1271–1277. [Google Scholar] [CrossRef] [PubMed]
- Azevedo, A.S.; Grotek, B.; Jacinto, A.; Weidinger, G.; Saude, L. The regenerative capacity of the zebrafish caudal fin is not affected by repeated amputations. PLoS ONE 2011, 6, e22820. [Google Scholar] [CrossRef] [PubMed]
- LeClair, E.E.; Topczewski, J. Development and regeneration of the zebrafish maxillary barbel: A novel study system for vertebrate tissue growth and repair. PLoS ONE 2010, 5, e8737. [Google Scholar] [CrossRef] [PubMed]
- Nachtrab, G.; Czerwinski, M.; Poss, K.D. Sexually dimorphic fin regeneration in zebrafish controlled by androgen/gsk3 signaling. Curr. Biol. 2011, 21, 1912–1917. [Google Scholar] [CrossRef] [PubMed]
- Brockes, J.P.; Kumar, A. Comparative aspects of animal regeneration. Annu. Rev. Cell Dev. Biol. 2008, 24, 525–549. [Google Scholar] [CrossRef] [PubMed]
- Gardiner, D.M.; Endo, T.; Bryant, S.V. The molecular basis of amphibian limb regeneration: Integrating the old with the new. Semin. Cell Dev. Biol. 2002, 13, 345–352. [Google Scholar] [CrossRef] [PubMed]
- Yun, M.H.; Gates, P.B.; Brockes, J.P. Regulation of p53 is critical for vertebrate limb regeneration. Proc. Natl. Acad. Sci. USA 2013, 110, 17392–17397. [Google Scholar] [CrossRef] [PubMed]
- Eguchi, G.; Eguchi, Y.; Nakamura, K.; Yadav, M.C.; Millan, J.L.; Tsonis, P.A. Regenerative capacity in newts is not altered by repeated regeneration and ageing. Nat. Commun. 2011, 2, 384. [Google Scholar] [CrossRef] [PubMed]
- Sandoval-Guzman, T.; Wang, H.; Khattak, S.; Schuez, M.; Roensch, K.; Nacu, E.; Tazaki, A.; Joven, A.; Tanaka, E.M.; Simon, A. Fundamental differences in dedifferentiation and stem cell recruitment during skeletal muscle regeneration in two salamander species. Cell Stem Cell 2014, 14, 174–187. [Google Scholar] [CrossRef] [PubMed]
- Margotta, V.; Filoni, S.; Merante, A.; Chimenti, C. Analysis of morphogenetic potential of caudal spinal cord in triturus carnifex adults (urodele amphibians) subjected to repeated tail amputations. Ital. J. Anat. Embryol. 2002, 107, 127–144. [Google Scholar] [PubMed]
- 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]
- Rossi, D.J.; Seita, J.; Czechowicz, A.; Bhattacharya, D.; Bryder, D.; Weissman, I.L. Hematopoietic stem cell quiescence attenuates DNA damage response and permits DNA damage accumulation during aging. Cell Cycle 2007, 6, 2371–2376. [Google Scholar] [CrossRef] [PubMed]
- Sotiropoulou, P.A.; Candi, A.; Mascre, G.; de Clercq, S.; Youssef, K.K.; Lapouge, G.; Dahl, E.; Semeraro, C.; Denecker, G.; Marine, J.C.; et al. Bcl-2 and accelerated DNA repair mediates resistance of hair follicle bulge stem cells to DNA-damage-induced cell death. Nat. Cell Biol. 2010, 12, 572–582. [Google Scholar] [CrossRef] [PubMed]
- 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]
- Nijnik, A.; Woodbine, L.; Marchetti, C.; Dawson, S.; Lambe, T.; Liu, C.; Rodrigues, N.P.; Crockford, T.L.; Cabuy, E.; Vindigni, A.; et al. DNA repair is limiting for haematopoietic stem cells during ageing. Nature 2007, 447, 686–690. [Google Scholar] [CrossRef] [PubMed]
- Sousa-Victor, P.; Gutarra, S.; Garcia-Prat, L.; Rodriguez-Ubreva, J.; Ortet, L.; Ruiz-Bonilla, V.; Jardi, M.; Ballestar, E.; Gonzalez, S.; Serrano, A.L.; et al. Geriatric muscle stem cells switch reversible quiescence into senescence. Nature 2014, 506, 316–321. [Google Scholar] [CrossRef] [PubMed]
- Wang, J.; Sun, Q.; Morita, Y.; Jiang, H.; Gross, A.; Lechel, A.; Hildner, K.; Guachalla, L.M.; Gompf, A.; Hartmann, D.; et al. A differentiation checkpoint limits hematopoietic stem cell self-renewal in response to DNA damage. Cell 2012, 148, 1001–1014. [Google Scholar] [CrossRef] [PubMed]
- Oh, J.; Lee, Y.D.; Wagers, A.J. Stem cell aging: Mechanisms, regulators and therapeutic opportunities. Nat. Med. 2014, 20, 870–880. [Google Scholar] [CrossRef] [PubMed]
- Cheung, H.H.; Liu, X.; Canterel-Thouennon, L.; Li, L.; Edmonson, C.; Rennert, O.M. Telomerase protects werner syndrome lineage-specific stem cells from premature aging. Stem Cell Rep. 2014, 2, 534–546. [Google Scholar] [CrossRef] [PubMed]
- 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]
- Mohrin, M.; Bourke, E.; Alexander, D.; Warr, M.R.; Barry-Holson, K.; Le Beau, M.M.; Morrison, C.G.; Passegue, E. Hematopoietic stem cell quiescence promotes error-prone DNA repair and mutagenesis. Cell Stem Cell 2010, 7, 174–185. [Google Scholar] [CrossRef] [PubMed]
- 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]
- Alt, E.U.; Senst, C.; Murthy, S.N.; Slakey, D.P.; Dupin, C.L.; Chaffin, A.E.; Kadowitz, P.J.; Izadpanah, R. Aging alters tissue resident mesenchymal stem cell properties. Stem Cell Res. 2012, 8, 215–225. [Google Scholar] [CrossRef] [PubMed]
- Sousounis, K.; Looso, M.; Maki, N.; Ivester, C.J.; Braun, T.; Tsonis, P.A. Transcriptome analysis of newt lens regeneration reveals distinct gradients in gene expression patterns. PLoS ONE 2013, 8, e61445. [Google Scholar] [CrossRef] [PubMed]
- Janzen, V.; Forkert, R.; Fleming, H.E.; Saito, Y.; Waring, M.T.; Dombkowski, D.M.; Cheng, T.; DePinho, R.A.; Sharpless, N.E.; Scadden, D.T. Stem-cell ageing modified by the cyclin-dependent kinase inhibitor p16ink4a. Nature 2006, 443, 421–426. [Google Scholar] [CrossRef] [PubMed]
- Krishnamurthy, J.; Ramsey, M.R.; Ligon, K.L.; Torrice, C.; Koh, A.; Bonner-Weir, S.; Sharpless, N.E. P16ink4a induces an age-dependent decline in islet regenerative potential. Nature 2006, 443, 453–457. [Google Scholar] [CrossRef] [PubMed]
- 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] [PubMed]
- Signer, R.A.; Morrison, S.J. Mechanisms that regulate stem cell aging and life span. Cell Stem Cell 2013, 12, 152–165. [Google Scholar] [CrossRef] [PubMed]
- Lane, D.P. Cancer. P53, guardian of the genome. Nature 1992, 358, 15–16. [Google Scholar] [CrossRef] [PubMed]
- Vousden, K.H.; Lu, X. Live or let die: The cell’s response to p53. Nat. Rev. Cancer 2002, 2, 594–604. [Google Scholar] [CrossRef] [PubMed]
- Dumble, M.; Moore, L.; Chambers, S.M.; Geiger, H.; van Zant, G.; Goodell, M.A.; Donehower, L.A. The impact of altered p53 dosage on hematopoietic stem cell dynamics during aging. Blood 2007, 109, 1736–1742. [Google Scholar] [CrossRef] [PubMed]
- Gannon, H.S.; Donehower, L.A.; Lyle, S.; Jones, S.N. Mdm2-p53 signaling regulates epidermal stem cell senescence and premature aging phenotypes in mouse skin. Dev. Biol. 2011, 353, 1–9. [Google Scholar] [CrossRef] [PubMed]
- Lee, J.Y.; Nakada, D.; Yilmaz, O.H.; Tothova, Z.; Joseph, N.M.; Lim, M.S.; Gilliland, D.G.; Morrison, S.J. Mtor activation induces tumor suppressors that inhibit leukemogenesis and deplete hematopoietic stem cells after pten deletion. Cell Stem Cell 2010, 7, 593–605. [Google Scholar] [CrossRef] [PubMed]
- Maier, B.; Gluba, W.; Bernier, B.; Turner, T.; Mohammad, K.; Guise, T.; Sutherland, A.; Thorner, M.; Scrable, H. Modulation of mammalian life span by the short isoform of p53. Genes Dev. 2004, 18, 306–319. [Google Scholar] [CrossRef] [PubMed]
- Tyner, S.D.; Venkatachalam, S.; Choi, J.; Jones, S.; Ghebranious, N.; Igelmann, H.; Lu, X.; Soron, G.; Cooper, B.; Brayton, C.; et al. P53 mutant mice that display early ageing-associated phenotypes. Nature 2002, 415, 45–53. [Google Scholar] [CrossRef] [PubMed]
- Carrasco-Garcia, E.; Arrizabalaga, O.; Serrano, M.; Lovell-Badge, R.; Matheu, A. Increased gene dosage of ink4/arf and p53 delays age-associated central nervous system functional decline. Aging Cell 2015, 14, 710–714. [Google Scholar] [CrossRef] [PubMed]
- Matheu, A.; Maraver, A.; Klatt, P.; Flores, I.; Garcia-Cao, I.; Borras, C.; Flores, J.M.; Vina, J.; Blasco, M.A.; Serrano, M. Delayed ageing through damage protection by the arf/p53 pathway. Nature 2007, 448, 375–379. [Google Scholar] [CrossRef] [PubMed]
- Hayflick, L.; Moorhead, P.S. The serial cultivation of human diploid cell strains. Exp. Cell Res. 1961, 25, 585–621. [Google Scholar] [CrossRef]
- Blasco, M.A. Telomere length, stem cells and aging. Natl. Chem. Biol. 2007, 3, 640–649. [Google Scholar] [CrossRef] [PubMed]
- 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]
- Vaziri, H.; Dragowska, W.; Allsopp, R.C.; Thomas, T.E.; Harley, C.B.; Lansdorp, P.M. Evidence for a mitotic clock in human hematopoietic stem cells: Loss of telomeric DNA with age. Proc. Natl. Acad. Sci. USA 1994, 91, 9857–9860. [Google Scholar] [CrossRef] [PubMed]
- Gomes, N.M.; Ryder, O.A.; Houck, M.L.; Charter, S.J.; Walker, W.; Forsyth, N.R.; Austad, S.N.; Venditti, C.; Pagel, M.; Shay, J.W.; et al. Comparative biology of mammalian telomeres: Hypotheses on ancestral states and the roles of telomeres in longevity determination. Aging Cell 2011, 10, 761–768. [Google Scholar] [CrossRef] [PubMed]
- Meyne, J.; Ratliff, R.L.; Moyzis, R.K. Conservation of the human telomere sequence (TTAGGG)n among vertebrates. Proc. Natl. Acad. Sci. USA 1989, 86, 7049–7053. [Google Scholar] [CrossRef] [PubMed]
- De Lange, T. How telomeres solve the end-protection problem. Science 2009, 326, 948–952. [Google Scholar] [CrossRef] [PubMed]
- Aubert, G.; Lansdorp, P.M. Telomeres and aging. Physiol. Rev. 2008, 88, 557–579. [Google Scholar] [CrossRef] [PubMed]
- Cong, Y.S.; Wright, W.E.; Shay, J.W. Human telomerase and its regulation. Microbiol. Mol. Biol. Rev. 2002, 66, 407–425. [Google Scholar] [CrossRef] [PubMed]
- Mitchell, J.R.; Wood, E.; Collins, K. A telomerase component is defective in the human disease dyskeratosis congenita. Nature 1999, 402, 551–555. [Google Scholar] [PubMed]
- Lee, H.W.; Blasco, M.A.; Gottlieb, G.J.; Horner, J.W., 2nd; Greider, C.W.; DePinho, R.A. Essential role of mouse telomerase in highly proliferative organs. Nature 1998, 392, 569–574. [Google Scholar] [PubMed]
- Ferron, S.; Mira, H.; Franco, S.; Cano-Jaimez, M.; Bellmunt, E.; Ramirez, C.; Farinas, I.; Blasco, M.A. Telomere shortening and chromosomal instability abrogates proliferation of adult but not embryonic neural stem cells. Development 2004, 131, 4059–4070. [Google Scholar] [CrossRef] [PubMed]
- Ferron, S.R.; Marques-Torrejon, M.A.; Mira, H.; Flores, I.; Taylor, K.; Blasco, M.A.; Farinas, I. Telomere shortening in neural stem cells disrupts neuronal differentiation and neuritogenesis. J. Neurosci. 2009, 29, 14394–14407. [Google Scholar] [CrossRef] [PubMed]
- Rudolph, K.L.; Chang, S.; Lee, H.W.; Blasco, M.; Gottlieb, G.J.; Greider, C.; DePinho, R.A. Longevity, stress response, and cancer in aging telomerase-deficient mice. Cell 1999, 96, 701–712. [Google Scholar] [CrossRef]
- Chin, L.; Artandi, S.E.; Shen, Q.; Tam, A.; Lee, S.L.; Gottlieb, G.J.; Greider, C.W.; DePinho, R.A. P53 deficiency rescues the adverse effects of telomere loss and cooperates with telomere dysfunction to accelerate carcinogenesis. Cell 1999, 97, 527–538. [Google Scholar] [CrossRef]
- Flores, I.; Blasco, M.A. A p53-dependent response limits epidermal stem cell functionality and organismal size in mice with short telomeres. PLoS ONE 2009, 4, e4934. [Google Scholar] [CrossRef] [PubMed]
- Jaskelioff, M.; Muller, F.L.; Paik, J.H.; Thomas, E.; Jiang, S.; Adams, A.C.; Sahin, E.; Kost-Alimova, M.; Protopopov, A.; Cadinanos, J.; et al. Telomerase reactivation reverses tissue degeneration in aged telomerase-deficient mice. Nature 2011, 469, 102–106. [Google Scholar] [CrossRef] [PubMed]
- Bar, C.; Bernardes de Jesus, B.; Serrano, R.; Tejera, A.; Ayuso, E.; Jimenez, V.; Formentini, I.; Bobadilla, M.; Mizrahi, J.; de Martino, A.; et al. Telomerase expression confers cardioprotection in the adult mouse heart after acute myocardial infarction. Nat. Commun. 2014, 5, 5863. [Google Scholar] [CrossRef] [PubMed]
- Francis, N.; Gregg, T.; Owen, R.; Ebert, T.; Bodnar, A. Lack of age-associated telomere shortening in long- and short-lived species of sea urchins. FEBS Lett. 2006, 580, 4713–4717. [Google Scholar] [CrossRef] [PubMed]
- Tan, T.C.; Rahman, R.; Jaber-Hijazi, F.; Felix, D.A.; Chen, C.; Louis, E.J.; Aboobaker, A. Telomere maintenance and telomerase activity are differentially regulated in asexual and sexual worms. Proc. Natl. Acad. Sci. USA 2012, 109, 4209–4214. [Google Scholar] [CrossRef] [PubMed]
- Bernardes de Jesus, B.; Vera, E.; Schneeberger, K.; Tejera, A.M.; Ayuso, E.; Bosch, F.; Blasco, M.A. Telomerase gene therapy in adult and old mice delays aging and increases longevity without increasing cancer. EMBO Mol. Med. 2012, 4, 691–704. [Google Scholar] [CrossRef] [PubMed]
- Harman, D. The biologic clock: The mitochondria? J. Am. Geriatr. Soc. 1972, 20, 145–147. [Google Scholar] [CrossRef] [PubMed]
- Doonan, R.; McElwee, J.J.; Matthijssens, F.; Walker, G.A.; Houthoofd, K.; Back, P.; Matscheski, A.; Vanfleteren, J.R.; Gems, D. Against the oxidative damage theory of aging: Superoxide dismutases protect against oxidative stress but have little or no effect on life span in caenorhabditis elegans. Genes Dev. 2008, 22, 3236–3241. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Andziak, B.; O’Connor, T.P.; Qi, W.; DeWaal, E.M.; Pierce, A.; Chaudhuri, A.R.; van Remmen, H.; Buffenstein, R. High oxidative damage levels in the longest-living rodent, the naked mole-rat. Aging Cell 2006, 5, 463–471. [Google Scholar] [CrossRef] [PubMed]
- Ninov, N.A.; Yun, M.H. Current advances in tissue repair and regeneration: The future is bright. Regeneration 2015, 2, 84–91. [Google Scholar] [CrossRef]
- Vriz, S.; Reiter, S.; Galliot, B. Cell death: A program to regenerate. Curr. Top. Dev. Biol. 2014, 108, 121–151. [Google Scholar] [PubMed]
- Gauron, C.; Rampon, C.; Bouzaffour, M.; Ipendey, E.; Teillon, J.; Volovitch, M.; Vriz, S. Sustained production of ros triggers compensatory proliferation and is required for regeneration to proceed. Sci. Rep. 2013, 3, 2084. [Google Scholar] [CrossRef] [PubMed]
- Love, N.R.; Chen, Y.; Ishibashi, S.; Kritsiligkou, P.; Lea, R.; Koh, Y.; Gallop, J.L.; Dorey, K.; Amaya, E. Amputation-induced reactive oxygen species are required for successful xenopus tadpole tail regeneration. Nat. Cell Biol. 2013, 15, 222–228. [Google Scholar] [CrossRef] [PubMed]
- Niethammer, P.; Grabher, C.; Look, A.T.; Mitchison, T.J. A tissue-scale gradient of hydrogen peroxide mediates rapid wound detection in zebrafish. Nature 2009, 459, 996–999. [Google Scholar] [CrossRef] [PubMed]
- Pirotte, N.; Stevens, A.S.; Fraguas, S.; Plusquin, M.; Van Roten, A.; Van Belleghem, F.; Paesen, R.; Ameloot, M.; Cebria, F.; Artois, T.; et al. Reactive oxygen species in planarian regeneration: An upstream necessity for correct patterning and brain formation. Oxid. Med. Cell. Longev. 2015, 2015, 392476. [Google Scholar] [CrossRef] [PubMed]
- Razzell, W.; Evans, I.R.; Martin, P.; Wood, W. Calcium flashes orchestrate the wound inflammatory response through duox activation and hydrogen peroxide release. Curr. Biol. 2013, 23, 424–429. [Google Scholar] [CrossRef] [PubMed]
- Ito, K.; Suda, T. Metabolic requirements for the maintenance of self-renewing stem cells. Nat. Rev. Mol. Cell Biol. 2014, 15, 243–256. [Google Scholar] [CrossRef] [PubMed]
- Edgar, D.; Shabalina, I.; Camara, Y.; Wredenberg, A.; Calvaruso, M.A.; Nijtmans, L.; Nedergaard, J.; Cannon, B.; Larsson, N.G.; Trifunovic, A. Random point mutations with major effects on protein-coding genes are the driving force behind premature aging in mtdna mutator mice. Cell Metab. 2009, 10, 131–138. [Google Scholar] [CrossRef] [PubMed]
- Ameur, A.; Stewart, J.B.; Freyer, C.; Hagstrom, E.; Ingman, M.; Larsson, N.G.; Gyllensten, U. Ultra-deep sequencing of mouse mitochondrial DNA: Mutational patterns and their origins. PLoS Genet. 2011, 7, e1002028. [Google Scholar] [CrossRef] [PubMed]
- Norddahl, G.L.; Pronk, C.J.; Wahlestedt, M.; Sten, G.; Nygren, J.M.; Ugale, A.; Sigvardsson, M.; Bryder, D. Accumulating mitochondrial DNA mutations drive premature hematopoietic aging phenotypes distinct from physiological stem cell aging. Cell Stem Cell 2011, 8, 499–510. [Google Scholar] [CrossRef] [PubMed]
- Ahlqvist, K.J.; Hamalainen, R.H.; Yatsuga, S.; Uutela, M.; Terzioglu, M.; Gotz, A.; Forsstrom, S.; Salven, P.; Angers-Loustau, A.; Kopra, O.H.; et al. Somatic progenitor cell vulnerability to mitochondrial DNA mutagenesis underlies progeroid phenotypes in polg mutator mice. Cell Metab. 2012, 15, 100–109. [Google Scholar] [CrossRef] [PubMed]
- Yahata, T.; Takanashi, T.; Muguruma, Y.; Ibrahim, A.A.; Matsuzawa, H.; Uno, T.; Sheng, Y.; Onizuka, M.; Ito, M.; Kato, S.; et al. Accumulation of oxidative DNA damage restricts the self-renewal capacity of human hematopoietic stem cells. Blood 2011, 118, 2941–2950. [Google Scholar] [CrossRef] [PubMed]
- Kroemer, G.; Galluzzi, L.; Brenner, C. Mitochondrial membrane permeabilization in cell death. Physiol. Rev. 2007, 87, 99–163. [Google Scholar] [CrossRef] [PubMed]
- Marzetti, E.; Wohlgemuth, S.E.; Lees, H.A.; Chung, H.Y.; Giovannini, S.; Leeuwenburgh, C. Age-related activation of mitochondrial caspase-independent apoptotic signaling in rat gastrocnemius muscle. Mech. Ageing Dev. 2008, 129, 542–549. [Google Scholar] [CrossRef]
- Pollina, E.A.; Brunet, A. Epigenetic regulation of aging stem cells. Oncogene 2011, 30, 3105–3126. [Google Scholar] [CrossRef] [PubMed]
- Maegawa, S.; Hinkal, G.; Kim, H.S.; Shen, L.; Zhang, L.; Zhang, J.; Zhang, N.; Liang, S.; Donehower, L.A.; Issa, J.P. Widespread and tissue specific age-related DNA methylation changes in mice. Genome Res. 2010, 20, 332–340. [Google Scholar] [CrossRef] [PubMed]
- Singhal, R.P.; Mays-Hoopes, L.L.; Eichhorn, G.L. DNA methylation in aging of mice. Mech. Ageing Dev. 1987, 41, 199–210. [Google Scholar] [CrossRef]
- Wilson, V.L.; Smith, R.A.; Ma, S.; Cutler, R.G. Genomic 5-methyldeoxycytidine decreases with age. J. Biol. Chem. 1987, 262, 9948–9951. [Google Scholar] [PubMed]
- Schellenberg, A.; Lin, Q.; Schuler, H.; Koch, C.M.; Joussen, S.; Denecke, B.; Walenda, G.; Pallua, N.; Suschek, C.V.; Zenke, M.; et al. Replicative senescence of mesenchymal stem cells causes DNA-methylation changes which correlate with repressive histone marks. Aging 2011, 3, 873–888. [Google Scholar] [PubMed]
- 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]
- Ma, D.K.; Jang, M.H.; Guo, J.U.; Kitabatake, Y.; Chang, M.L.; Pow-Anpongkul, N.; Flavell, R.A.; Lu, B.; Ming, G.L.; Song, H. Neuronal activity-induced gadd45b promotes epigenetic DNA demethylation and adult neurogenesis. Science 2009, 323, 1074–1077. [Google Scholar] [CrossRef] [PubMed]
- Trowbridge, J.J.; Orkin, S.H. DNA methylation in adult stem cells: New insights into self-renewal. Epigenetics 2010, 5, 189–193. [Google Scholar] [CrossRef] [PubMed]
- Trowbridge, J.J.; Snow, J.W.; Kim, J.; Orkin, S.H. DNA methyltransferase 1 is essential for and uniquely regulates hematopoietic stem and progenitor cells. Cell Stem Cell 2009, 5, 442–449. [Google Scholar] [CrossRef] [PubMed]
- Wu, H.; Coskun, V.; Tao, J.; Xie, W.; Ge, W.; Yoshikawa, K.; Li, E.; Zhang, Y.; Sun, Y.E. Dnmt3a-dependent nonpromoter DNA methylation facilitates transcription of neurogenic genes. Science 2010, 329, 444–448. [Google Scholar] [CrossRef] [PubMed]
- Zhao, X.; Ueba, T.; Christie, B.R.; Barkho, B.; McConnell, M.J.; Nakashima, K.; Lein, E.S.; Eadie, B.D.; Willhoite, A.R.; Muotri, A.R.; et al. Mice lacking methyl-cpg binding protein 1 have deficits in adult neurogenesis and hippocampal function. Proc. Natl. Acad. Sci. USA 2003, 100, 6777–6782. [Google Scholar] [CrossRef] [PubMed]
- Stewart, S.; Tsun, Z.Y.; Izpisua Belmonte, J.C. A histone demethylase is necessary for regeneration in zebrafish. Proc. Natl. Acad. Sci. USA 2009, 106, 19889–19894. [Google Scholar] [CrossRef] [PubMed]
- Hamada, Y.; Bando, T.; Nakamura, T.; Ishimaru, Y.; Mito, T.; Noji, S.; Tomioka, K.; Ohuchi, H. Regenerated leg segment patterns are regulated epigenetically by histone h3k27 methylation in the cricket gryllus bimaculatus. Development 2015, 142, 2916–2927. [Google Scholar] [CrossRef] [PubMed]
- Brown, K.; Xie, S.; Qiu, X.; Mohrin, M.; Shin, J.; Liu, Y.; Zhang, D.; Scadden, D.T.; Chen, D. Sirt3 reverses aging-associated degeneration. Cell Rep. 2013, 3, 319–327. [Google Scholar] [CrossRef] [PubMed]
- Cardus, A.; Uryga, A.K.; Walters, G.; Erusalimsky, J.D. Sirt6 protects human endothelial cells from DNA damage, telomere dysfunction, and senescence. Cardiovasc. Res. 2013, 97, 571–579. [Google Scholar] [CrossRef] [PubMed]
- Oellerich, M.F.; Potente, M. Foxos and sirtuins in vascular growth, maintenance, and aging. Circ. Res. 2012, 110, 1238–1251. [Google Scholar] [CrossRef] [PubMed]
- Burnett, C.; Valentini, S.; Cabreiro, F.; Goss, M.; Somogyvari, M.; Piper, M.D.; Hoddinott, M.; Sutphin, G.L.; Leko, V.; McElwee, J.J.; et al. Absence of effects of sir2 overexpression on lifespan in c. Elegans and drosophila. Nature 2011, 477, 482–485. [Google Scholar] [CrossRef] [PubMed]
- Park, I.K.; Qian, D.; Kiel, M.; Becker, M.W.; Pihalja, M.; Weissman, I.L.; Morrison, S.J.; Clarke, M.F. Bmi-1 is required for maintenance of adult self-renewing haematopoietic stem cells. Nature 2003, 423, 302–305. [Google Scholar] [CrossRef] [PubMed]
- Molofsky, A.V.; He, S.; Bydon, M.; Morrison, S.J.; Pardal, R. Bmi-1 promotes neural stem cell self-renewal and neural development but not mouse growth and survival by repressing the p16ink4a and p19arf senescence pathways. Genes Dev. 2005, 19, 1432–1437. [Google Scholar] [CrossRef] [PubMed]
- Nishino, J.; Kim, I.; Chada, K.; Morrison, S.J. Hmga2 promotes neural stem cell self-renewal in young but not old mice by reducing p16ink4a and p19arf expression. Cell 2008, 135, 227–239. [Google Scholar] [CrossRef] [PubMed]
- Ugalde, A.P.; Kwarciak, A.; Caravia, X.M.; López-Otín, C.; Ramsay, A.J. The emergence of geromirs: A group of micrornas implicated in aging. In Micrornas in Medicine; Lawrie, C.H., Ed.; John Wiley & Sons, Inc.: Hoboken, NJ, USA, 2013. [Google Scholar]
- De Lencastre, A.; Pincus, Z.; Zhou, K.; Kato, M.; Lee, S.S.; Slack, F.J. Micrornas both promote and antagonize longevity in C. elegans. Curr. Biol. 2010, 20, 2159–2168. [Google Scholar] [CrossRef] [PubMed]
- Liu, N.; Landreh, M.; Cao, K.; Abe, M.; Hendriks, G.J.; Kennerdell, J.R.; Zhu, Y.; Wang, L.S.; Bonini, N.M. The microrna mir-34 modulates ageing and neurodegeneration in drosophila. Nature 2012, 482, 519–523. [Google Scholar] [CrossRef] [PubMed]
- Gomez-Cabello, D.; Adrados, I.; Palmero, I. Microrna biogenesis and senescence. Aging 2013, 5, 721–722. [Google Scholar] [PubMed]
- Thatcher, E.J.; Paydar, I.; Anderson, K.K.; Patton, J.G. Regulation of zebrafish fin regeneration by micrornas. Proc. Natl. Acad. Sci. USA 2008, 105, 18384–18389. [Google Scholar] [CrossRef] [PubMed]
- Yin, V.P.; Thomson, J.M.; Thummel, R.; Hyde, D.R.; Hammond, S.M.; Poss, K.D. Fgf-dependent depletion of microrna-133 promotes appendage regeneration in zebrafish. Genes Dev. 2008, 22, 728–733. [Google Scholar] [CrossRef] [PubMed]
- Sehm, T.; Sachse, C.; Frenzel, C.; Echeverri, K. Mir-196 is an essential early-stage regulator of tail regeneration, upstream of key spinal cord patterning events. Dev. Biol. 2009, 334, 468–480. [Google Scholar] [CrossRef] [PubMed]
- Witman, N.; Heigwer, J.; Thaler, B.; Lui, W.O.; Morrison, J.I. Mir-128 regulates non-myocyte hyperplasia, deposition of extracellular matrix and islet1 expression during newt cardiac regeneration. Dev. Biol. 2013, 383, 253–263. [Google Scholar] [CrossRef] [PubMed]
- Menghini, R.; Casagrande, V.; Cardellini, M.; Martelli, E.; Terrinoni, A.; Amati, F.; Vasa-Nicotera, M.; Ippoliti, A.; Novelli, G.; Melino, G.; et al. Microrna 217 modulates endothelial cell senescence via silent information regulator 1. Circulation 2009, 120, 1524–1532. [Google Scholar] [CrossRef] [PubMed]
- Yamakuchi, M.; Ferlito, M.; Lowenstein, C.J. Mir-34a repression of sirt1 regulates apoptosis. Proc. Natl. Acad. Sci. USA 2008, 105, 13421–13426. [Google Scholar] [CrossRef] [PubMed]
- Saez, I.; Vilchez, D. The mechanistic links between proteasome activity, aging and age-related diseases. Curr. Genom. 2014, 15, 38–51. [Google Scholar] [CrossRef] [PubMed]
- Morimoto, R.I. Proteotoxic stress and inducible chaperone networks in neurodegenerative disease and aging. Genes Dev. 2008, 22, 1427–1438. [Google Scholar] [CrossRef] [PubMed]
- Rubinsztein, D.C.; Marino, G.; Kroemer, G. Autophagy and aging. Cell 2011, 146, 682–695. [Google Scholar] [CrossRef] [PubMed]
- Ben-Zvi, A.; Miller, E.A.; Morimoto, R.I. Collapse of proteostasis represents an early molecular event in caenorhabditis elegans aging. Proc. Natl. Acad. Sci. USA 2009, 106, 14914–14919. [Google Scholar] [CrossRef] [PubMed]
- Tomaru, U.; Takahashi, S.; Ishizu, A.; Miyatake, Y.; Gohda, A.; Suzuki, S.; Ono, A.; Ohara, J.; Baba, T.; Murata, S.; et al. Decreased proteasomal activity causes age-related phenotypes and promotes the development of metabolic abnormalities. Am. J. Pathol. 2012, 180, 963–972. [Google Scholar] [CrossRef] [PubMed]
- Mortensen, M.; Soilleux, E.J.; Djordjevic, G.; Tripp, R.; Lutteropp, M.; Sadighi-Akha, E.; Stranks, A.J.; Glanville, J.; Knight, S.; Jacobsen, S.E.; et al. The autophagy protein atg7 is essential for hematopoietic stem cell maintenance. J. Exp. Med. 2011, 208, 455–467. [Google Scholar] [CrossRef] [PubMed]
- Mortensen, M.; Watson, A.S.; Simon, A.K. Lack of autophagy in the hematopoietic system leads to loss of hematopoietic stem cell function and dysregulated myeloid proliferation. Autophagy 2011, 7, 1069–1070. [Google Scholar] [CrossRef] [PubMed]
- Serrano, M. Dissecting the role of mtor complexes in cellular senescence. Cell Cycle 2012, 11, 2231–2232. [Google Scholar] [CrossRef] [PubMed]
- Xu, S.; Cai, Y.; Wei, Y. Mtor signaling from cellular senescence to organismal aging. Aging Dis. 2014, 5, 263–273. [Google Scholar] [PubMed]
- Iglesias-Bartolome, R.; Patel, V.; Cotrim, A.; Leelahavanichkul, K.; Molinolo, A.A.; Mitchell, J.B.; Gutkind, J.S. Mtor inhibition prevents epithelial stem cell senescence and protects from radiation-induced mucositis. Cell Stem Cell 2012, 11, 401–414. [Google Scholar] [CrossRef] [PubMed]
- Laberge, R.M.; Sun, Y.; Orjalo, A.V.; Patil, C.K.; Freund, A.; Zhou, L.; Curran, S.C.; Davalos, A.R.; Wilson-Edell, K.A.; Liu, S.; et al. Mtor regulates the pro-tumorigenic senescence-associated secretory phenotype by promoting il1a translation. Nat. Cell Biol. 2015, 17, 1049–1061. [Google Scholar] [CrossRef] [PubMed]
- Rafalski, V.A.; Mancini, E.; Brunet, A. Energy metabolism and energy-sensing pathways in mammalian embryonic and adult stem cell fate. J. Cell Sci. 2012, 125, 5597–5608. [Google Scholar] [CrossRef] [PubMed]
- Chen, C.; Liu, Y.; Liu, R.; Ikenoue, T.; Guan, K.L.; Zheng, P. Tsc-mtor maintains quiescence and function of hematopoietic stem cells by repressing mitochondrial biogenesis and reactive oxygen species. J. Exp. Med. 2008, 205, 2397–2408. [Google Scholar] [CrossRef] [PubMed]
- Castilho, R.M.; Squarize, C.H.; Chodosh, L.A.; Williams, B.O.; Gutkind, J.S. Mtor mediates wnt-induced epidermal stem cell exhaustion and aging. Cell Stem Cell 2009, 5, 279–289. [Google Scholar] [CrossRef] [PubMed]
- Yilmaz, O.H.; Katajisto, P.; Lamming, D.W.; Gultekin, Y.; Bauer-Rowe, K.E.; Sengupta, S.; Birsoy, K.; Dursun, A.; Yilmaz, V.O.; Selig, M.; et al. Mtorc1 in the paneth cell niche couples intestinal stem-cell function to calorie intake. Nature 2012, 486, 490–495. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cheng, C.W.; Adams, G.B.; Perin, L.; Wei, M.; Zhou, X.; Lam, B.S.; da Sacco, S.; Mirisola, M.; Quinn, D.I.; Dorff, T.B.; et al. Prolonged fasting reduces igf-1/pka to promote hematopoietic-stem-cell-based regeneration and reverse immunosuppression. Cell Stem Cell 2014, 14, 810–823. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Schmuck, E.G.; Mulligan, J.D.; Saupe, K.W. Caloric restriction attenuates the age-associated increase of adipose-derived stem cells but further reduces their proliferative capacity. Age 2011, 33, 107–118. [Google Scholar] [CrossRef] [PubMed]
- 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]
- Fontana, L.; Partridge, L. Promoting health and longevity through diet: From model organisms to humans. Cell 2015, 161, 106–118. [Google Scholar] [CrossRef] [PubMed]
- Dorr, J.R.; Yu, Y.; Milanovic, M.; Beuster, G.; Zasada, C.; Dabritz, J.H.; Lisec, J.; Lenze, D.; Gerhardt, A.; Schleicher, K.; et al. Synthetic lethal metabolic targeting of cellular senescence in cancer therapy. Nature 2013, 501, 421–425. [Google Scholar] [CrossRef] [PubMed]
- Kuhlow, D.; Florian, S.; von Figura, G.; Weimer, S.; Schulz, N.; Petzke, K.J.; Zarse, K.; Pfeiffer, A.F.; Rudolph, K.L.; Ristow, M. Telomerase deficiency impairs glucose metabolism and insulin secretion. Aging 2010, 2, 650–658. [Google Scholar] [PubMed]
- Carlson, M.E.; Conboy, I.M. Loss of stem cell regenerative capacity within aged niches. Aging Cell 2007, 6, 371–382. [Google Scholar] [CrossRef] [PubMed]
- Chakkalakal, J.V.; Jones, K.M.; Basson, M.A.; Brack, A.S. The aged niche disrupts muscle stem cell quiescence. Nature 2012, 490, 355–360. [Google Scholar] [CrossRef] [PubMed]
- 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] [PubMed]
- Biteau, B.; Jasper, H. Egf signaling regulates the proliferation of intestinal stem cells in drosophila. Development 2011, 138, 1045–1055. [Google Scholar] [CrossRef] [PubMed]
- Miranda, C.J.; Braun, L.; Jiang, Y.; Hester, M.E.; Zhang, L.; Riolo, M.; Wang, H.; Rao, M.; Altura, R.A.; Kaspar, B.K. Aging brain microenvironment decreases hippocampal neurogenesis through wnt-mediated survivin signaling. Aging Cell 2012, 11, 542–552. [Google Scholar] [CrossRef] [PubMed]
- Conboy, I.M.; Conboy, M.J.; Wagers, A.J.; Girma, E.R.; Weissman, I.L.; Rando, T.A. Rejuvenation of aged progenitor cells by exposure to a young systemic environment. Nature 2005, 433, 760–764. [Google Scholar] [CrossRef] [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] [PubMed]
- Kumar, A.; Brockes, J.P. Nerve dependence in tissue, organ, and appendage regeneration. Trends Neurosci. 2012, 35, 691–699. [Google Scholar] [CrossRef] [PubMed]
- Geraudie, J.; Singer, M. Necessity of an adequate nerve supply for regeneration of the amputated pectoral fin in the teleost fundulus. J. Exp. Zool. 1985, 234, 367–374. [Google Scholar] [CrossRef] [PubMed]
- Brownell, I.; Guevara, E.; Bai, C.B.; Loomis, C.A.; Joyner, A.L. Nerve-derived sonic hedgehog defines a niche for hair follicle stem cells capable of becoming epidermal stem cells. Cell Stem Cell 2011, 8, 552–565. [Google Scholar] [CrossRef] [PubMed]
- Yamazaki, S.; Ema, H.; Karlsson, G.; Yamaguchi, T.; Miyoshi, H.; Shioda, S.; Taketo, M.M.; Karlsson, S.; Iwama, A.; Nakauchi, H. Nonmyelinating schwann cells maintain hematopoietic stem cell hibernation in the bone marrow niche. Cell 2011, 147, 1146–1158. [Google Scholar] [CrossRef] [PubMed]
- Mahmoud, A.I.; O’Meara, C.C.; Gemberling, M.; Zhao, L.; Bryant, D.M.; Zheng, R.; Gannon, J.B.; Cai, L.; Choi, W.Y.; Egnaczyk, G.F.; et al. Nerves regulate cardiomyocyte proliferation and heart regeneration. Dev. Cell 2015, 34, 387–399. [Google Scholar] [CrossRef] [PubMed]
- Verdu, E.; Ceballos, D.; Vilches, J.J.; Navarro, X. Influence of aging on peripheral nerve function and regeneration. J. Peripher. Nerv. Syst. 2000, 5, 191–208. [Google Scholar] [CrossRef] [PubMed]
- Guilak, F.; Cohen, D.M.; Estes, B.T.; Gimble, J.M.; Liedtke, W.; Chen, C.S. Control of stem cell fate by physical interactions with the extracellular matrix. Cell Stem Cell 2009, 5, 17–26. [Google Scholar] [CrossRef] [PubMed]
- Wood, L.K.; Kayupov, E.; Gumucio, J.P.; Mendias, C.L.; Claflin, D.R.; Brooks, S.V. Intrinsic stiffness of extracellular matrix increases with age in skeletal muscles of mice. J. Appl. Physiol. 2014, 117, 363–369. [Google Scholar] [CrossRef] [PubMed]
- Gilbert, P.M.; Havenstrite, K.L.; Magnusson, K.E.; Sacco, A.; Leonardi, N.A.; Kraft, P.; Nguyen, N.K.; Thrun, S.; Lutolf, M.P.; Blau, H.M. Substrate elasticity regulates skeletal muscle stem cell self-renewal in culture. Science 2010, 329, 1078–1081. [Google Scholar] [CrossRef] [PubMed]
- Lacraz, G.; Rouleau, A.J.; Couture, V.; Sollrald, T.; Drouin, G.; Veillette, N.; Grandbois, M.; Grenier, G. Increased stiffness in aged skeletal muscle impairs muscle progenitor cell proliferative activity. PLoS ONE 2015, 10, e0136217. [Google Scholar] [CrossRef] [PubMed]
- Carlson, B.M.; Faulkner, J.A. Muscle transplantation between young and old rats: Age of host determines recovery. Am. J. Physiol. 1989, 256, 1262–1266. [Google Scholar]
- 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] [PubMed]
- Loffredo, F.S.; Steinhauser, M.L.; Jay, S.M.; Gannon, J.; Pancoast, J.R.; Yalamanchi, P.; Sinha, M.; Dall’Osso, C.; Khong, D.; Shadrach, J.L.; et al. Growth differentiation factor 11 is a circulating factor that reverses age-related cardiac hypertrophy. Cell 2013, 153, 828–839. [Google Scholar] [CrossRef] [PubMed]
- Ruckh, J.M.; Zhao, J.W.; Shadrach, J.L.; van Wijngaarden, P.; Rao, T.N.; Wagers, A.J.; Franklin, R.J. Rejuvenation of regeneration in the aging central nervous system. Cell Stem Cell 2012, 10, 96–103. [Google Scholar] [CrossRef] [PubMed]
- Villeda, S.A.; Luo, J.; Mosher, K.I.; Zou, B.; Britschgi, M.; Bieri, G.; Stan, T.M.; Fainberg, N.; Ding, Z.; Eggel, A.; et al. The ageing systemic milieu negatively regulates neurogenesis and cognitive function. Nature 2011, 477, 90–94. [Google Scholar] [CrossRef] [PubMed]
- 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] [PubMed]
- Elabd, C.; Cousin, W.; Upadhyayula, P.; Chen, R.Y.; Chooljian, M.S.; Li, J.; Kung, S.; Jiang, K.P.; Conboy, I.M. Oxytocin is an age-specific circulating hormone that is necessary for muscle maintenance and regeneration. Nat. Commun. 2014, 5, 4082. [Google Scholar] [CrossRef] [PubMed]
- Fletcher, T.J. The induction of male sexual behavior in red deer (cervus elaphus) by the administration of testosterone to hinds and estradiol-17beta to stags. Horm. Behav. 1978, 11, 74–88. [Google Scholar] [CrossRef]
- Price, J.S.; Allen, S.; Faucheux, C.; Althnaian, T.; Mount, J.G. Deer antlers: A zoological curiosity or the key to understanding organ regeneration in mammals? J. Anat. 2005, 207, 603–618. [Google Scholar] [CrossRef] [PubMed]
- Price, J.S.; Oyajobi, B.O.; Oreffo, R.O.; Russell, R.G. Cells cultured from the growing tip of red deer antler express alkaline phosphatase and proliferate in response to insulin-like growth factor-i. J. Endocrinol. 1994, 143, 9–16. [Google Scholar] [CrossRef]
- Arnold, L.; Henry, A.; Poron, F.; Baba-Amer, Y.; van Rooijen, N.; Plonquet, A.; Gherardi, R.K.; Chazaud, B. Inflammatory monocytes recruited after skeletal muscle injury switch into antiinflammatory macrophages to support myogenesis. J. Exp. Med. 2007, 204, 1057–1069. [Google Scholar] [CrossRef] [PubMed]
- Munoz-Canoves, P.; Serrano, A.L. Macrophages decide between regeneration and fibrosis in muscle. Trends Endocrinol. Metab. 2015, 26, 449–450. [Google Scholar] [CrossRef] [PubMed]
- Kyritsis, N.; Kizil, C.; Zocher, S.; Kroehne, V.; Kaslin, J.; Freudenreich, D.; Iltzsche, A.; Brand, M. Acute inflammation initiates the regenerative response in the adult zebrafish brain. Science 2012, 338, 1353–1356. [Google Scholar] [CrossRef] [PubMed]
- Petrie, T.A.; Strand, N.S.; Yang, C.T.; Rabinowitz, J.S.; Moon, R.T. Macrophages modulate adult zebrafish tail fin regeneration. Development 2014, 141, 2581–2591. [Google Scholar] [CrossRef] [PubMed]
- Godwin, J.W.; Pinto, A.R.; Rosenthal, N.A. Macrophages are required for adult salamander limb regeneration. Proc. Natl. Acad. Sci. USA 2013, 110, 9415–9420. [Google Scholar] [CrossRef] [PubMed]
- Chung, H.Y.; Cesari, M.; Anton, S.; Marzetti, E.; Giovannini, S.; Seo, A.Y.; Carter, C.; Yu, B.P.; Leeuwenburgh, C. Molecular inflammation: Underpinnings of aging and age-related diseases. Ageing Res. Rev. 2009, 8, 18–30. [Google Scholar] [CrossRef] [PubMed]
- Weng, N.P. Aging of the immune system: How much can the adaptive immune system adapt? Immunity 2006, 24, 495–499. [Google Scholar] [CrossRef] [PubMed]
- Jurk, D.; Wilson, C.; Passos, J.F.; Oakley, F.; Correia-Melo, C.; Greaves, L.; Saretzki, G.; Fox, C.; Lawless, C.; Anderson, R.; et al. Chronic inflammation induces telomere dysfunction and accelerates ageing in mice. Nat. Commun. 2014, 2, 4172. [Google Scholar] [CrossRef] [PubMed]
- Hoenicke, L.; Zender, L. Immune surveillance of senescent cells-biological significance in cancer- and non-cancer pathologies. Carcinogenesis 2012, 33, 1123–1126. [Google Scholar] [CrossRef] [PubMed]
- Kang, T.W.; Yevsa, T.; Woller, N.; Hoenicke, L.; Wuestefeld, T.; Dauch, D.; Hohmeyer, A.; Gereke, M.; Rudalska, R.; Potapova, A.; et al. Senescence surveillance of pre-malignant hepatocytes limits liver cancer development. Nature 2011, 479, 547–551. [Google Scholar] [CrossRef] [PubMed]
- Fulop, T.; Franceschi, C.; Hirokawa, K.; Pawelec, G. Handbook on Immunosenescence; Springer Science+Business Media B.V.: Dordrecht, The Netherlands, 2009. [Google Scholar]
- Yun, M.H.; Davaapil, H.; Brockes, J.P. Recurrent turnover of senescent cells during regeneration of a complex structure. eLife 2015, 4, e05505. [Google Scholar] [CrossRef] [PubMed]
- Cabreiro, F.; Gems, D. Worms need microbes too: Microbiota, health and aging in caenorhabditis elegans. EMBO Mol. Med. 2013, 5, 1300–1310. [Google Scholar] [CrossRef] [PubMed]
- Collins, S.M.; Surette, M.; Bercik, P. The interplay between the intestinal microbiota and the brain. Nat. Rev. Microbiol. 2012, 10, 735–742. [Google Scholar] [CrossRef] [PubMed]
- Buchon, N.; Broderick, N.A.; Chakrabarti, S.; Lemaitre, B. Invasive and indigenous microbiota impact intestinal stem cell activity through multiple pathways in drosophila. Genes Dev. 2009, 23, 2333–2344. [Google Scholar] [CrossRef] [PubMed]
- Nigro, G.; Rossi, R.; Commere, P.H.; Jay, P.; Sansonetti, P.J. The cytosolic bacterial peptidoglycan sensor nod2 affords stem cell protection and links microbes to gut epithelial regeneration. Cell Host Microbe 2014, 15, 792–798. [Google Scholar] [CrossRef] [PubMed]
- Van Deursen, J.M. The role of senescent cells in ageing. Nature 2014, 509, 439–446. [Google Scholar] [CrossRef] [PubMed]
- Beausejour, C.M.; Krtolica, A.; Galimi, F.; Narita, M.; Lowe, S.W.; Yaswen, P.; Campisi, J. Reversal of human cellular senescence: Roles of the p53 and p16 pathways. EMBO J. 2003, 22, 4212–4222. [Google Scholar] [CrossRef] [PubMed]
- Collins, C.J.; Sedivy, J.M. Involvement of the ink4a/arf gene locus in senescence. Aging Cell 2003, 2, 145–150. [Google Scholar] [CrossRef] [PubMed]
- Jacobs, J.J.; de Lange, T. Significant role for p16ink4a in p53-independent telomere-directed senescence. Curr. Biol. 2004, 14, 2302–2308. [Google Scholar] [CrossRef] [PubMed]
- Schmitt, C.A.; Fridman, J.S.; Yang, M.; Lee, S.; Baranov, E.; Hoffman, R.M.; Lowe, S.W. A senescence program controlled by p53 and p16ink4a contributes to the outcome of cancer therapy. Cell 2002, 109, 335–346. [Google Scholar] [CrossRef]
- Shay, J.W.; Pereira-Smith, O.M.; Wright, W.E. A role for both rb and p53 in the regulation of human cellular senescence. Exp. Cell Res. 1991, 196, 33–39. [Google Scholar] [CrossRef]
- Dimri, G.P.; Lee, X.H.; Basile, G.; Acosta, M.; Scott, C.; Roskelley, C.; Medrano, E.E.; Linskens, M.; Rubelj, I.; Pereirasmith, O.; et al. A biomarker that identifies senescent human-cells in culture and in aging skin in vivo. Proc. Natl. Acad. Sci. USA 1995, 92, 9363–9367. [Google Scholar] [CrossRef] [PubMed]
- Acosta, J.C.; O’Loghlen, A.; Banito, A.; Guijarro, M.V.; Augert, A.; Raguz, S.; Fumagalli, M.; da Costa, M.; Brown, C.; Popov, N.; et al. Chemokine signaling via the cxcr2 receptor reinforces senescence. Cell 2008, 133, 1006–1018. [Google Scholar] [CrossRef] [PubMed]
- Coppe, J.P.; Patil, C.K.; Rodier, F.; Sun, Y.; Munoz, D.P.; Goldstein, J.; Nelson, P.S.; Desprez, P.Y.; Campisi, J. Senescence-associated secretory phenotypes reveal cell-nonautonomous functions of oncogenic ras and the p53 tumor suppressor. PLoS Biol. 2008, 6, 2853–2868. [Google Scholar] [PubMed]
- Kuilman, T.; Michaloglou, C.; Vredeveld, L.C.; Douma, S.; van Doorn, R.; Desmet, C.J.; Aarden, L.A.; Mooi, W.J.; Peeper, D.S. Oncogene-induced senescence relayed by an interleukin-dependent inflammatory network. Cell 2008, 133, 1019–1031. [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]
- Nelson, G.; Wordsworth, J.; Wang, C.; Jurk, D.; Lawless, C.; Martin-Ruiz, C.; von Zglinicki, T. A senescent cell bystander effect: Senescence-induced senescence. Aging Cell 2012, 11, 345–349. [Google Scholar] [CrossRef] [PubMed]
- Wang, C.; Jurk, D.; Maddick, M.; Nelson, G.; Martin-Ruiz, C.; von Zglinicki, T. DNA damage response and cellular senescence in tissues of aging mice. Aging Cell 2009, 8, 311–323. [Google Scholar] [CrossRef] [PubMed]
- Yang, H.; Fogo, A.B. Cell senescence in the aging kidney. J. Am. Soc. Nephrol. 2010, 21, 1436–1439. [Google Scholar] [CrossRef] [PubMed]
- Baker, D.J.; Wijshake, T.; Tchkonia, T.; LeBrasseur, N.K.; Childs, B.G.; van de Sluis, B.; Kirkland, J.L.; van Deursen, J.M. Clearance of p16ink4a-positive senescent cells delays ageing-associated disorders. Nature 2011, 479, 232–236. [Google Scholar] [CrossRef] [PubMed]
- Campisi, J. Cellular senescence: Putting the paradoxes in perspective. Curr. Opin. Genet. Dev. 2011, 21, 107–112. [Google Scholar] [CrossRef] [PubMed]
- Braun, H.; Schmidt, B.M.; Raiss, M.; Baisantry, A.; Mircea-Constantin, D.; Wang, S.; Gross, M.L.; Serrano, M.; Schmitt, R.; Melk, A. Cellular senescence limits regenerative capacity and allograft survival. J. Am. Soc. Nephrol. 2012, 23, 1467–1473. [Google Scholar] [CrossRef] [PubMed]
- Zhou, F.; Onizawa, S.; Nagai, A.; Aoshiba, K. Epithelial cell senescence impairs repair process and exacerbates inflammation after airway injury. Respir. Res. 2011, 12, 78. [Google Scholar] [CrossRef] [PubMed]
- 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]
- Munoz-Espin, D.; Canamero, M.; Maraver, A.; Gomez-Lopez, G.; Contreras, J.; Murillo-Cuesta, S.; Rodriguez-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]
- Storer, M.; Mas, A.; Robert-Moreno, A.; Pecoraro, M.; Ortells, M.C.; di Giacomo, V.; Yosef, R.; Pilpel, N.; Krizhanovsky, V.; Sharpe, J.; et al. Senescence is a developmental mechanism that contributes to embryonic growth and patterning. Cell 2013, 155, 1119–1130. [Google Scholar] [CrossRef] [PubMed]
- Demaria, M.; Ohtani, N.; Youssef, S.A.; Rodier, F.; Toussaint, W.; Mitchell, J.R.; Laberge, R.M.; Vijg, J.; van Steeg, H.; Dolle, M.E.; et al. An essential role for senescent cells in optimal wound healing through secretion of pdgf-aa. Dev. Cell 2014, 31, 722–733. [Google Scholar] [CrossRef] [PubMed]
- Jun, J.I.; Lau, L.F. Cellular senescence controls fibrosis in wound healing. Aging 2010, 2, 627–631. [Google Scholar] [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] [PubMed]
- Zhu, F.; Li, Y.; Zhang, J.; Piao, C.; Liu, T.; Li, H.H.; Du, J. Senescent cardiac fibroblast is critical for cardiac fibrosis after myocardial infarction. PLoS ONE 2013, 8, e74535. [Google Scholar] [CrossRef] [PubMed]
- Zhu, Y.; Tchkonia, T.; Pirtskhalava, T.; Gower, A.C.; Ding, H.; Giorgadze, N.; Palmer, A.K.; Ikeno, Y.; Hubbard, G.B.; Lenburg, M.; et al. The achilles’ heel of senescent cells: From transcriptome to senolytic drugs. Aging Cell 2015, 14. [Google Scholar] [CrossRef] [PubMed]
- Wahlestedt, M.; Norddahl, G.L.; Sten, G.; Ugale, A.; Frisk, M.A.; Mattsson, R.; Deierborg, T.; Sigvardsson, M.; Bryder, D. An epigenetic component of hematopoietic stem cell aging amenable to reprogramming into a young state. Blood 2013, 121, 4257–4264. [Google Scholar] [CrossRef] [PubMed]
- Nishimura, T.; Kaneko, S.; Kawana-Tachikawa, A.; Tajima, Y.; Goto, H.; Zhu, D.; Nakayama-Hosoya, K.; Iriguchi, S.; Uemura, Y.; Shimizu, T.; et al. Generation of rejuvenated antigen-specific T cells by reprogramming to pluripotency and redifferentiation. Cell Stem Cell 2013, 12, 114–126. [Google Scholar] [CrossRef] [PubMed]
- Vizcardo, R.; Masuda, K.; Yamada, D.; Ikawa, T.; Shimizu, K.; Fujii, S.; Koseki, H.; Kawamoto, H. Regeneration of human tumor antigen-specific T cells from ipscs derived from mature CD8+ T cells. Cell Stem Cell 2013, 12, 31–36. [Google Scholar] [CrossRef] [PubMed]
- Crompton, J.G.; Rao, M.; Restifo, N.P. Memoirs of a reincarnated t cell. Cell Stem Cell 2013, 12, 6–8. [Google Scholar] [CrossRef] [PubMed]
- Artandi, S.E.; Blau, H.M.; de Haan, G.; Geiger, H.; Goodell, M.A.; Jones, L.; Levine, R.L.; Munoz-Canoves, P.; Rodewald, H.R.; Wagers, A.; et al. Stem cells and aging: What’s next? Cell Stem Cell 2015, 16, 578–581. [Google Scholar] [PubMed]
- Tanaka, E.M.; Reddien, P.W. The cellular basis for animal regeneration. Dev. Cell 2011, 21, 172–185. [Google Scholar] [CrossRef] [PubMed]
- Loof, S.; Straube, W.L.; Drechsel, D.; Tanaka, E.M.; Simon, A. Plasticity of mammalian myotubes upon stimulation with a thrombin-activated serum factor. Cell Cycle 2007, 6, 1096–1101. [Google Scholar] [CrossRef] [PubMed]
- Yun, M.H.; Gates, P.B.; Brockes, J.P. Sustained erk activation underlies reprogramming in regeneration-competent salamander cells and distinguishes them from their mammalian counterparts. Stem Cell Rep. 2014, 3, 15–23. [Google Scholar] [CrossRef] [PubMed]
- Maki, N.; Suetsugu-Maki, R.; Tarui, H.; Agata, K.; del Rio-Tsonis, K.; Tsonis, P.A. Expression of stem cell pluripotency factors during regeneration in newts. Dev. Dyn. 2009, 238, 1613–1616. [Google Scholar] [CrossRef] [PubMed]
- Gentile, L.; Cebria, F.; Bartscherer, K. The planarian flatworm: An in vivo model for stem cell biology and nervous system regeneration. Dis. Model Mech. 2011, 4, 12–19. [Google Scholar] [CrossRef] [PubMed]
- Wagner, D.E.; Wang, I.E.; Reddien, P.W. Clonogenic neoblasts are pluripotent adult stem cells that underlie planarian regeneration. Science 2011, 332, 811–816. [Google Scholar] [CrossRef] [PubMed]
© 2015 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 license (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Yun, M.H. Changes in Regenerative Capacity through Lifespan. Int. J. Mol. Sci. 2015, 16, 25392-25432. https://doi.org/10.3390/ijms161025392
Yun MH. Changes in Regenerative Capacity through Lifespan. International Journal of Molecular Sciences. 2015; 16(10):25392-25432. https://doi.org/10.3390/ijms161025392
Chicago/Turabian StyleYun, Maximina H. 2015. "Changes in Regenerative Capacity through Lifespan" International Journal of Molecular Sciences 16, no. 10: 25392-25432. https://doi.org/10.3390/ijms161025392
APA StyleYun, M. H. (2015). Changes in Regenerative Capacity through Lifespan. International Journal of Molecular Sciences, 16(10), 25392-25432. https://doi.org/10.3390/ijms161025392