Stem Cell Quiescence versus Senescence: The Key for Healthy Aging
Abstract
BACKGROUND: Aging tissues lose their homeostatic and regenerative capacities, which has been linked to the degeneration of the stem cells such as the tissue-specific stem cells, the stem cell niches, and systemic cues that regulate stem cell activity.
CONTENT: The maintenance of tissue homeostatic and regeneration dependent on its tissue-specific stem cells, that —long-lived cells with the ability to self-renew and differentiate into mature cells. Understanding the molecular mechanisms that governs stem cell survival, self-renewal, quiescence, proliferation, and commitment to specific differentiated cell lineages is critical for identifying the drivers and effectors of age-associated stem cell failure. Such understanding will be critical for the development of therapeutic approaches that can decrease, and possibly reverse and repair the age-related degenerative process in aging tissues.
SUMMARY: The exact mechanisms and reasons of aging process were not fully elucidated until now. Stem cells is one of the keys for maintaining tissues heath and understanding how stem cell decline with age will give us opportunities to find strategy in increasing somatic stem cells regenerative capacity and delay the aging process.
KEYWORDS: adult stem cell, aging, epigenetic, metabolism, quiescence, senescence
Full Text:
PDFReferences
Ermolaeva M, Neri F, Ori A, Rudolph KL. Cellular and epigenetic drivers of stem cell ageing. Nat Rev Mol Cell Biol. 2018; 19: 594–610, CrossRef.
Mohrin M, Bourke E, Alexander D, Warr MR, Barry-Holson K, Le Beau MM, et al. Hematopoietic stem cell quiescence promotes error-prone DNA repair and mutagenesis. Cell Stem Cell. 2010; 7: 174–85, CrossRef.
Sotiropoulou PA, Candi A, Mascré G, De Clercq S, Youssef KK, Lapouge G, 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: 572–82, CrossRef.
Behrens A, van Deursen JM, Rudolph KL, Schumacher B. Impact of genomic damage and ageing on stem cell function. Nat Cell Biol. 2014; 16: 201–7, CrossRef.
Cheung TH, Rando TA. Molecular regulation of stem cell quiescence. Nat Rev Mol Cell Biol. 2013; 14: 329–40, CrossRef.
Oh J, Lee YD, Wagers AJ. Stem cell aging: mechanisms, regulators and therapeutic opportunities. Nat Med. 2014; 20: 870–80, CrossRef.
Liu L, Rando TA. Manifestations and mechanisms of stem cell aging. J Cell Biol. 2011; 193: 257–66, CrossRef.
Blanpain C, Fuchs E. Plasticity of epithelial stem cells in tissue regeneration. Science. 2014; 344: 1242281, CrossRef.
Lucas D, Scheiermann C, Chow A, Kunisaki Y, Bruns I, Barrick C, et al. Chemotherapy-induced bone marrow nerve injury impairs hematopoietic regeneration. Nature Medicine. 2013; 19: 695–703, CrossRef.
Pietras EM, Lakshminarasimhan R, Techner JM, Fong S, Flach J, Binnewies M, et al. Re-entry into quiescence protects hematopoietic stem cells from the killing effect of chronic exposure to type I interferons. J Exp Med. 2014; 211: 245–62, CrossRef.
Lymperi S, Ferraro F, Scadden DT. The HSC niche concept has turned 31: Has our knowledge matured? Ann NY Acad Sci. 2010; 1192: 12–8, CrossRef.
Wang LD, Wagers AJ. Dynamic niches in the origination and differentiation of haematopoietic stem cells. Nat Rev Mol Cell Biol. 2011; 12: 643–55, CrossRef.
Warr MR, Pietras EM, Passegué E. Mechanisms controlling hematopoietic stem cell functions during normal hematopoiesis and hematological malignancies: Mechanisms controlling HSC functions. WIREs Syst Biol Med. 2011; 3: 681–701, CrossRef.
Morrison SJ, Scadden DT. The bone marrow niche for haematopoietic stem cells. Nature. 2014; 505: 327–34, CrossRef.
Adamo L, Naveiras O, Wenzel PL, McKinney-Freeman S, Mack PJ, Gracia-Sancho J, et al. Biomechanical forces promote embryonic haematopoiesis. Nature. 2009; 459: 1131–5, CrossRef.
Shin JW, Buxboim A, Spinler KR, Swift J, Christian DA, Hunter CA, et al. Contractile forces sustain and polarize hematopoiesis from stem and progenitor cells. Cell Stem Cell. 2014; 14: 81–93, CrossRef.
Passegué E, Wagers AJ, Giuriato S, Anderson WC, Weissman IL. Global analysis of proliferation and cell cycle gene expression in the regulation of hematopoietic stem and progenitor cell fates. Journal of Experimental Medicine. 2005; 202: 1599–611, CrossRef.
Essers MAG, Offner S, Blanco-Bose WE, Waibler Z, Kalinke U, Duchosal MA, et al. IFNα activates dormant haematopoietic stem cells in vivo. Nature. 2009; 458: 904–8, CrossRef.
Wilson A, Laurenti E, Oser G, van der Wath RC, Blanco-Bose W, Jaworski M, et al. Hematopoietic stem cells reversibly switch from dormancy to self-renewal during homeostasis and repair. Cell. 2008; 135: 1118–29, CrossRef.
Baldridge MT, King KY, Boles NC, Weksberg DC, Goodell MA. Quiescent haematopoietic stem cells are activated by IFN-γ in response to chronic infection. Nature. 2010; 465: 793–7, CrossRef.
Mendelson A, Frenette PS. Hematopoietic stem cell niche maintenance during homeostasis and regeneration. Nat Med. 2014; 20: 833–46, CrossRef.
Friedenstein AJ, Chailakhyan RK, Latsinik NV, Panasyuk AF, Keiliss-Borok IV. Stromal cells responsible for transferring the microenvironment of the hemopoietic tissues: cloning in vitro and retransplantation in vivo. Transplantation. 1974; 17: 331–40, CrossRef.
Song J, Kiel MJ, Wang Z, Wang J, Taichman RS, Morrison SJ, et al. An in vivo model to study and manipulate the hematopoietic stem cell niche. Blood. 2010; 115: 2592–600, CrossRef.
Sacchetti B, Funari A, Michienzi S, Di Cesare S, Piersanti S, Saggio I, et al. Self-renewing osteoprogenitors in bone marrow sinusoids can organize a hematopoietic microenvironment. Cell. 2007; 131: 324–36, CrossRef.
Pinho S, Lacombe J, Hanoun M, Mizoguchi T, Bruns I, Kunisaki Y, et al. PDGFRα and CD51 mark human Nestin+ sphere-forming mesenchymal stem cells capable of hematopoietic progenitor cell expansion. J Exp Med. 2013; 210: 1351–67, CrossRef.
Méndez-Ferrer S, Michurina TV, Ferraro F, Mazloom AR, MacArthur BD, Lira SA, et al. Mesenchymal and haematopoietic stem cells form a unique bone marrow niche. Nature. 2010; 466: 829–34, CrossRef.
Nolan DJ, Ginsberg M, Israely E, Palikuqi B, Poulos MG, James D, et al. Molecular signatures of tissue-specific microvascular endothelial cell heterogeneity in organ maintenance and regeneration. Dev Cell. 2013; 26: 204–19, CrossRef.
Kobayashi H, Butler JM, O’Donnell R, Kobayashi M, Ding BS, Bonner B, et al. Angiocrine factors from Akt-activated endothelial cells balance self-renewal and differentiation of haematopoietic stem cells. Nat Cell Biol. 2010; 12: 1046–56, CrossRef.
Krause DS, Scadden DT, Preffer FI. The hematopoietic stem cell niche--home for friend and foe? Cytometry B Clin Cytom. 2013; 84: 7–20, CrossRef.
Kennedy M, Firpo M, Choi K, Wall C, Robertson S, Kabrun N, et al. A common precursor for primitive erythropoiesis and definitive haematopoiesis. Nature. 1997; 386: 488–93, CrossRef.
Shalaby F, Rossant J, Yamaguchi TP, Gertsenstein M, Wu XF, Breitman ML, et al. Failure of blood-island formation and vasculogenesis in Flk-1-deficient mice. Nature. 1995; 376: 62–6, CrossRef.
Ono N, Ono W, Mizoguchi T, Nagasawa T, Frenette PS, Kronenberg HM. Vasculature-associated cells expressing nestin in developing bones encompass early cells in the osteoblast and endothelial lineage. Dev Cell. 2014; 29: 330–9, CrossRef.
Chow A, Lucas D, Hidalgo A, Méndez-Ferrer S, Hashimoto D, Scheiermann C, et al. Bone marrow CD169+ macrophages promote the retention of hematopoietic stem and progenitor cells in the mesenchymal stem cell niche. J Exp Med. 2011; 208: 261–71, CrossRef.
Christopher MJ, Rao M, Liu F, Woloszynek JR, Link DC. Expression of the G-CSF receptor in monocytic cells is sufficient to mediate hematopoietic progenitor mobilization by G-CSF in mice. J Exp Med. 2011; 208: 251–60, CrossRef.
Winkler IG, Sims NA, Pettit AR, Barbier V, Nowlan B, Helwani F, et al. Bone marrow macrophages maintain hematopoietic stem cell (HSC) niches and their depletion mobilizes HSCs. Blood. 2010; 116: 4815–28, CrossRef.
Lucas D, Bruns I, Battista M, Mendez-Ferrer S, Magnon C, Kunisaki Y, et al. Norepinephrine reuptake inhibition promotes mobilization in mice: potential impact to rescue low stem cell yields. Blood. 2012; 119: 3962–5, CrossRef.
Lin H. The stem-cell niche theory: lessons from flies. Nat Rev Genet. 2002; 3: 931–40, CrossRef.
Schofield R. The relationship between the spleen colony-forming cell and the haemopoietic stem cell. Blood Cells. 1978; 4: 7–25, PMID.
Geiger H, de Haan G, Florian MC. The ageing haematopoietic stem cell compartment. Nat Rev Immunol. 2013; 13: 376–89, CrossRef.
Köhler A, Schmithorst V, Filippi M-D, Ryan MA, Daria D, Gunzer M, et al. Altered cellular dynamics and endosteal location of aged early hematopoietic progenitor cells revealed by time-lapse intravital imaging in long bones. Blood. 2009; 114: 290–8, CrossRef.
Sato T, Clevers H. Growing self-organizing mini-guts from a single intestinal stem cell: mechanism and applications. Science. 2013; 340: 1190–4, CrossRef.
Boehnke K, Falkowska-Hansen B, Stark HJ, Boukamp P. Stem cells of the human epidermis and their niche: composition and function in epidermal regeneration and carcinogenesis. Carcinogenesis. 2012; 33: 1247–58, CrossRef.
Ito M, Myung P. Dissecting the bulge in hair regeneration. J Clin Invest. 2012; 122: 448–54, CrossRef.
Li L, Clevers H. Coexistence of quiescent and active adult stem cells in mammals. Science. 2010; 327: 542–5, CrossRef.
Orford KW, Scadden DT. Deconstructing stem cell self-renewal: genetic insights into cell-cycle regulation. Nat Rev Genet. 2008; 9: 115–28, CrossRef.
Campisi J. Cellular senescence: putting the paradoxes in perspective. Curr Opin Genet Dev. 2011; 21: 107–12, CrossRef.
Terzi MY, Izmirli M, Gogebakan B. The cell fate: senescence or quiescence. Mol Biol Rep. 2016; 43: 1213–20, CrossRef.
Campisi J. Aging, cellular senescence, and cancer. Annu Rev Physiol. 2013; 75: 685–705, CrossRef.
Campisi J. Senescent cells, tumor suppression, and organismal aging: good citizens, bad neighbors. Cell. 2005; 120: 513–22, CrossRef.
Coppé JP, Desprez PY, Krtolica A, Campisi J. The senescence-associated secretory phenotype: the dark side of tumor suppression. Annu Rev Pathol. 2010; 5: 99–118, CrossRef.
Sousa-Victor P, Gutarra S, García-Prat L, Rodriguez-Ubreva J, Ortet L, Ruiz-Bonilla V, et al. Geriatric muscle stem cells switch reversible quiescence into senescence. Nature. 2014; 506: 316–21, CrossRef.
Janzen V, Forkert R, Fleming HE, Saito Y, Waring MT, Dombkowski DM, et al. Stem-cell ageing modified by the cyclin-dependent kinase inhibitor p16INK4a. Nature. 2006; 443: 421–6, CrossRef.
Coppé JP, Patil CK, Rodier F, Sun Y, Muñoz DP, Goldstein J, et al. Senescence-associated secretory phenotypes reveal cell-nonautonomous functions of oncogenic RAS and the p53 tumor suppressor. PLOS Biol. 2008; 6: 2853–68, CrossRef.
Baker DJ, Wijshake T, Tchkonia T, LeBrasseur NK, Childs BG, van de Sluis B, et al. Clearance of p16 Ink4a -positive senescent cells delays ageing-associated disorders. Nature. 2011; 479: 232–6, CrossRef.
Dhawan J, Rando TA. Stem cells in postnatal myogenesis: molecular mechanisms of satellite cell quiescence, activation and replenishment. Trends Cell Biol. 2005; 15: 666–73, CrossRef.
Burtner CR, Kennedy BK. Progeria syndromes and ageing: what is the connection? Nat Rev Mol Cell Biol. 2010; 11: 567–78, CrossRef.
Arthur ST, Cooley ID. The effect of physiological stimuli on sarcopenia; impact of notch and Wnt signaling on impaired aged skeletal muscle repair. Int J Biol Sci. 2012; 8: 731–60, CrossRef.
Jang YC, Sinha M, Cerletti M, Dall’Osso C, Wagers AJ. Skeletal muscle stem cells: effects of aging and metabolism on muscle regenerative function. Cold Spring Harb Symp Quant Biol. 2011; 76: 101–11, CrossRef.
Renault V, Thornell LE, Eriksson PO, Butler-Browne G, Mouly V, Thorne LE. Regenerative potential of human skeletal muscle during aging. Aging Cell. 2002; 1: 132–9, CrossRef.
Yin H, Price F, Rudnicki MA. Satellite cells and the muscle stem cell niche. Physiol Rev. 2013; 93: 23–67, CrossRef.
Shefer G, Van de Mark DP, Richardson JB, Yablonka-Reuveni Z. Satellite-cell pool size does matter: defining the myogenic potency of aging skeletal muscle. Dev Biol. 2006; 294: 50–66, CrossRef.
Comai G, Tajbakhsh S. Molecular and cellular regulation of skeletal myogenesis. Curr Top Dev Biol. 2014; 110: 1–73, CrossRef.
Montarras D, L’honoré A, Buckingham M. Lying low but ready for action: the quiescent muscle satellite cell. FEBS J. 2013; 280: 4036–50, CrossRef.
Rera M, Bahadorani S, Cho J, Koehler CL, Ulgherait M, Hur JH, et al. Modulation of longevity and tissue homeostasis by the Drosophila PGC-1 homolog. Cell Metab. 2011; 14: 623–34, CrossRef.
Cheng T, Rodrigues N, Shen H, Yang Y, Dombkowski D, Sykes M, et al. Hematopoietic stem cell quiescence maintained by p21cip1/waf1. Science. 2000; 287: 1804–8, CrossRef.
Kippin TE, Martens DJ, van der Kooy D. p21 loss compromises the relative quiescence of forebrain stem cell proliferation leading to exhaustion of their proliferation capacity. Genes Dev. 2005; 19: 756–67, CrossRef.
Chakkalakal JV, Jones KM, Basson MA, Brack AS. The aged niche disrupts muscle stem cell quiescence. Nature. 2012; 490: 355–60, CrossRef.
Hamanaka RB, Chandel NS. Mitochondrial reactive oxygen species regulate cellular signaling and dictate biological outcomes. Trends Biochem Sci. 2010; 35: 505–13, CrossRef.
Owusu-Ansah E, Banerjee U. Reactive oxygen species prime Drosophila haematopoietic progenitors for differentiation. Nature. 2009; 461: 537–41, CrossRef.
Hüttmann A, Liu SL, Boyd AW, Li CL. Functional heterogeneity within rhodamine123lo Hoechst33342lo/sp primitive hemopoietic stem cells revealed by pyronin Y. Exp Hematol. 2001; 29: 1109–16, CrossRef.
Fukada S, Uezumi A, Ikemoto M, Masuda S, Segawa M, Tanimura N, et al. Molecular signature of quiescent satellite cells in adult skeletal muscle. Stem Cells. 2007; 25: 2448–59, CrossRef.
Gerdes J, Schwab U, Lemke H, Stein H. Production of a mouse monoclonal antibody reactive with a human nuclear antigen associated with cell proliferation. Int J Cancer. 1983; 31: 13–20, CrossRef.
Hsu YC, Fuchs E. A family business: stem cell progeny join the niche to regulate homeostasis. Nat Rev Mol Cell Biol. 2012; 13: 103–14, CrossRef.
Rando TA. Stem cells, ageing and the quest for immortality. Nature. 2006; 441: 1080–6, CrossRef.
Kregel KC, Zhang HJ. An integrated view of oxidative stress in aging: basic mechanisms, functional effects, and pathological considerations. Am J Physiol Regul Integr Comp Physiol. 2007; 292: R18–36, CrossRef.
Rossi AP, Rubele S, Pelizzari L, Fantin F, Morgante S, Marchi O, et al. Effects of brisk walking on physical performance and muscle function in community dwelling elderly women. 2017; J Gerontol Geriatr Res. 2017; 6: 1000399, CrossRef.
Klionsky DJ, Emr SD. Autophagy as a regulated pathway of cellular degradation. Science. 2000; 290: 1717–21, CrossRef.
Mortensen M, Soilleux EJ, Djordjevic G, Tripp R, Lutteropp M, Sadighi-Akha E, et al. The autophagy protein Atg7 is essential for hematopoietic stem cell maintenance. J Exp Med. 2011; 208: 455–67, CrossRef.
Cuervo AM, Bergamini E, Brunk UT, Dröge W, Ffrench M, Terman A. Autophagy and aging: the importance of maintaining “clean” cells. Autophagy. 2005; 1: 131–40, CrossRef.
He C, Klionsky DJ. Regulation mechanisms and signaling pathways of autophagy. Annu Rev Genet. 2009; 43: 67–93, CrossRef.
Carnio S, LoVerso F, Baraibar MA, Longa E, Khan MM, Maffei M, et al. Autophagy impairment in muscle induces neuromuscular junction degeneration and precocious aging. Cell Reports. 2014; 8: 1509–21, CrossRef.
Rubinsztein DC, Mariño G, Kroemer G. Autophagy and aging. Cell. 2011; 146: 682–95, CrossRef.
García-Prat L, Martínez-Vicente M, Perdiguero E, Ortet L, Rodríguez-Ubreva J, Rebollo E, et al. Autophagy maintains stemness by preventing senescence. Nature. 2016; 529: 37–42, CrossRef.
Fontana L, Partridge L, Longo VD. Extending healthy life span--from yeast to humans. Science. 2010; 328: 321–6, CrossRef.
Harrison DE, Strong R, Sharp ZD, Nelson JF, Astle CM, Flurkey K, et al. Rapamycin fed late in life extends lifespan in genetically heterogeneous mice. Nature. 2009; 460: 392–5, CrossRef.
Tatar M, Sedivy JM. Mitochondria: masters of epigenetics. Cell. 2016; 165: 1052–4. Tatar M, Sedivy JM. Mitochondria: masters of epigenetics. Cell. 2016; 165: 1052–4, CrossRef.
Berger SL, Sassone-Corsi P. Metabolic signaling to chromatin. Cold Spring Harb Perspect Biol. 2016; 8: a019463, CrossRef.
Merkwirth C, Jovaisaite V, Durieux J, Matilainen O, Jordan SD, Quiros PM, et al. Two conserved histone demethylases regulate mitochondrial stress-induced longevity. Cell. 2016; 165: 1209–23, CrossRef.
Tian Y, Garcia G, Bian Q, Steffen KK, Joe L, Wolff S, et al. Mitochondrial stress induces chromatin reorganization to promote longevity and UPRmt. Cell. 2016; 165: 1197–208, CrossRef.
Han B, Sivaramakrishnan P, Lin C-CJ, Neve IAA, He J, Tay LWR, et al. Microbial genetic composition tunes host longevity. Cell. 2017; 169: 1249–62.e13, CrossRef.
Lin CCJ, Wang MC. Microbial metabolites regulate host lipid metabolism through NR5A–hedgehog signalling. Nat Cell Biol. 2017; 19: 550–7, CrossRef.
Lin YF, Schulz AM, Pellegrino MW, Lu Y, Shaham S, Haynes CM. Maintenance and propagation of a deleterious mitochondrial genome by the mitochondrial unfolded protein response. Nature. 2016; 533: 416–9, CrossRef.
Matilainen O, Quirós PM, Auwerx J. Mitochondria and epigenetics – crosstalk in homeostasis and stress. Trends Cell Biol. 2017; 27: 453–63, CrossRef.
Frederick DW, Loro E, Liu L, Davila A, Chellappa K, Silverman IM, et al. Loss of NAD homeostasis leads to progressive and reversible degeneration of skeletal muscle. Cell Metab. 2016; 24: 269–82, CrossRef.
Cartee GD, Hepple RT, Bamman MM, Zierath JR. Exercise promotes healthy aging of skeletal muscle. Cell Metab. 2016; 23: 1034–47, CrossRef.
López-Otín C, Blasco MA, Partridge L, Serrano M, Kroemer G. The hallmarks of aging. Cell. 2013; 153: 1194–217, CrossRef.
Sen P, Shah PP, Nativio R, Berger SL. Epigenetic mechanisms of longevity and aging. Cell. 2016; 166: 822–39, CrossRef.
Benayoun BA, Pollina EA, Brunet A. Epigenetic regulation of ageing: linking environmental inputs to genomic stability. Nat Rev Mol Cell Biol. 2015; 16: 593–610, CrossRef.
Booth LN, Brunet A. The aging epigenome. Mol Cell. 2016; 62: 728–44, CrossRef.
Pal S, Tyler JK. Epigenetics and aging. Sci Adv. 2016; 2: e1600584, CrossRef.
Ren R, Deng L, Xue Y, Suzuki K, Zhang W, Yu Y, et al. Visualization of aging-associated chromatin alterations with an engineered TALE system. Cell Res. 2017; 27: 483–504, CrossRef.
Zhang W, Li J, Suzuki K, Qu J, Wang P, Zhou J, et al. A Werner syndrome stem cell model unveils heterochromatin alterations as a driver of human aging. Science. 2015; 348: 1160–3, CrossRef.
Liu GH, Barkho BZ, Ruiz S, Diep D, Qu J, Yang SL, et al. Recapitulation of premature ageing with iPSCs from Hutchinson–Gilford progeria syndrome. Nature. 2011; 472: 221–5, CrossRef.
Kubben N, Zhang W, Wang L, Voss TC, Yang J, Qu J, et al. Repression of the antioxidant NRF2 pathway in premature aging. Cell. 2016; 165: 1361–74, CrossRef.
Deng L, Ren R, Wu J, Suzuki K, Izpisua Belmote JC, Liu GH. CRISPR/Cas9 and TALE: beyond cut and paste. Protein Cell. 2015; 6: 157–9, CrossRef.
Ding Z, Sui L, Ren R, Liu Y, Xu X, Fu L, et al. A widely adaptable approach to generate integration-free iPSCs from non-invasively acquired human somatic cells. Protein Cell. 2015; 6: 386–9, CrossRef.
Fu L, Xu X, Ren R, Wu J, Zhang W, Yang J, et al. Modeling xeroderma pigmentosum associated neurological pathologies with patients-derived iPSCs. Protein Cell. 2016; 7: 210–21, CrossRef.
Yang J, Li J, Suzuki K, Liu X, Wu J, Zhang W, et al. Genetic enhancement in cultured human adult stem cells conferred by a single nucleotide recoding. Cell Res. 2017; 27: 1178–81, CrossRef.
Wang H, Diao D, Shi Z, Zhu X, Gao Y, Gao S, et al. SIRT6 Controls hematopoietic stem cell homeostasis through epigenetic regulation of Wnt signaling. Cell Stem Cell. 2016; 18: 495–507, CrossRef.
Buckley SM, Aranda-Orgilles B, Strikoudis A, Apostolou E, Loizou E, Moran-Crusio K, et al. Regulation of pluripotency and cellular reprogramming by the ubiquitin-proteasome system. Cell Stem Cell. 2012; 11: 783–98, CrossRef.
Vilchez D, Boyer L, Morantte I, Lutz M, Merkwirth C, Joyce D, et al. Increased proteasome activity in human embryonic stem cells is regulated by PSMD11. Nature. 2012; 489: 304–8, CrossRef.
Liu F, Lee JY, Wei H, Tanabe O, Engel JD, Morrison SJ, et al. FIP200 is required for the cell-autonomous maintenance of fetal hematopoietic stem cells. Blood. 2010; 116: 4806–14, CrossRef.
Murphy CT, McCarroll SA, Bargmann CI, Fraser A, Kamath RS, Ahringer J, et al. Genes that act downstream of DAF-16 to influence the lifespan of Caenorhabditis elegans. Nature. 2003; 424: 277–83, CrossRef.
Oh SW, Mukhopadhyay A, Dixit BL, Raha T, Green MR, Tissenbaum HA. Identification of direct DAF-16 targets controlling longevity, metabolism and diapause by chromatin immunoprecipitation. Nat Genet. 2006; 38: 251–7, CrossRef.
Demontis F, Perrimon N. FOXO/4E-BP signaling in drosophila muscles regulates organism-wide proteostasis during aging. Cell. 2010; 143: 813–25, CrossRef.
Chen H, Liu X, Chen H, Cao J, Zhang L, Hu X, et al. Role of SIRT1 and AMPK in mesenchymal stem cells differentiation. Ageing Res Rev. 2014; 13: 55–64, CrossRef.
Chen J, Astle CM, Harrison DE. Hematopoietic senescence is postponed and hematopoietic stem cell function is enhanced by dietary restriction. Exp Hematol. 2003; 31: 1097–103. Chen J, Astle CM, Harrison DE. Hematopoietic senescence is postponed and hematopoietic stem cell function is enhanced by dietary restriction. Exp Hematol. 2003; 31: 1097–103, CrossRef.
Cheng CW, Adams GB, Perin L, Wei M, Zhou X, Lam BS, et al. Prolonged fasting reduces IGF-1/PKA to promote hematopoietic stem cell-based regeneration and reverse immunosuppression. Cell Stem Cell. 2014; 14: 810–23, CrossRef.
Salminen A, Kauppinen A, Hiltunen M, Kaarniranta K. Krebs cycle intermediates regulate DNA and histone methylation: epigenetic impact on the aging process. Ageing Res Rev. 2014; 16: 45–65, CrossRef.
Martin-Montalvo A, de Cabo R. Mitochondrial metabolic reprogramming induced by calorie restriction. Antioxid Redox Signal. 2013; 19: 310–20, CrossRef.
López-Lluch G, Hunt N, Jones B, Zhu M, Jamieson H, Hilmer S, et al. Calorie restriction induces mitochondrial biogenesis and bioenergetic efficiency. Proc Natl Acad Sci USA. 2006; 103: 1768–73, CrossRef.
Cerletti M, Jang YC, Finley LWS, Haigis MC, Wagers AJ. Short-term calorie restriction enhances skeletal muscle stem cell function. Cell Stem Cell. 2012; 10: 515–9, CrossRef.
Hekimi S, Lapointe J, Wen Y. Taking a “good” look at free radicals in the aging process. Trends Cell Biol. 2011; 21: 569–76, CrossRef.
Mohrin M, Shin J, Liu Y, Brown K, Luo H, Xi Y, et al. A mitochondrial UPR-mediated metabolic checkpoint regulates hematopoietic stem cell aging. Science. 2015; 347: 1374–7, CrossRef.
Tanaka Y, Okamoto K, Teye K, Umata T, Yamagiwa N, Suto Y, et al. JmjC enzyme KDM2A is a regulator of rRNA transcription in response to starvation. EMBO J. 2010; 29: 1510–22, CrossRef.
Shimazu T, Hirschey MD, Newman J, He W, Shirakawa K, Le Moan N, et al. Suppression of oxidative stress by β-hydroxybutyrate, an endogenous histone deacetylase inhibitor. Science. 2013; 339: 211–4, CrossRef.
Ryall JG, Dell’Orso S, Derfoul A, Juan A, Zare H, Feng X, et al. The NAD+-dependent SIRT1 deacetylase translates a metabolic switch into regulatory epigenetics in skeletal muscle stem cells. Cell Stem Cell. 2015; 16: 171–83, CrossRef.
Collado M, Blasco MA, Serrano M. Cellular senescence in cancer and aging. Cell. 2007; 130: 223–33, CrossRef.
Hentze H, Graichen R, Colman A. Cell therapy and the safety of embryonic stem cell-derived grafts. Trends Biotechnol. 2007; 25: 24–32, CrossRef.
Wang Y, Han ZB, Song YP, Han ZC. Safety of mesenchymal stem cells for clinical application. Stem Cells Int. 2012; 2012: 652034, CrossRef.
Hockemeyer D, Jaenisch R. Induced pluripotent stem cells meet genome editing. Cell Stem Cell. 2016; 18: 573–86, CrossRef.
Yamanaka S. Induced pluripotent stem cells: past, present, and future. Cell Stem Cell. 2012; 10: 678–84, CrossRef.
Pan H, Guan D, Liu X, Li J, Wang L, Wu J, et al. SIRT6 safeguards human mesenchymal stem cells from oxidative stress by coactivating NRF2. Cell Res. 2016; 26: 190–205, CrossRef.
Ren R, Ocampo A, Liu GH, Izpisua Belmonte JC. Regulation of stem cell aging by metabolism and epigenetics. Cell Metab. 2017; 26: 460–74, CrossRef.
DOI: https://doi.org/10.18585/inabj.v13i4.1655
Copyright (c) 2021 The Prodia Education and Research Institute
This work is licensed under a Creative Commons Attribution-NonCommercial 4.0 International License.
Indexed by:
The Prodia Education and Research Institute