In Search for Anti-Aging Strategy: Can We Rejuvenate Our Aging Stem Cells?

Anna Meiliana, Nurrani Mustika Dewi, Andi Wijaya

Abstract


BACKGROUND: Recent evidence suggested that we grow old partly because of our stem cells grow old as a result of mechanisms that suppress the development of cancer over a lifetime. We believe that a further, more precise mechanistic understanding of this process will be required before this knowledge can be translated into human anti-aging therapies.

CONTENT: A diminished capacity to maintain tissue homeostasis is a central physiological characteristic of aging. As stem cells regulate tissue homeostasis, depletion of stem cell reserves and/or diminished stem cell function have been postulated to contribute to aging. It has further been suggested that accumulated DNA damage could be a principal mechanism underlying age-dependent stem cell decline. It is interesting that many of the rejuvenating interventions act on the stem cell compartments, perhaps reflecting shared genetic and biochemical pathways controlling stem cell function and longevity. Strategy to slow down the aging processes is based on caloric restriction refers to a dietary regimen low in calories but without undernutrition. Sirtuin (SIRT)1 and 3, increases longevity by mimicking the beneficial effects of caloric restriction. SIRT3 regulates stress-responsive mitochondrial homeostasis, and more importantly, SIRT3 upregulation rejuvenates aged stem cells in tissues. Resveratrol (3,5,4’-trihydroxystilbene), a natural polyphenol found in grapes and wine, was the most powerful natural activator of SIRT1. In fact, resveratrol treatment has been demonstrated to rescue adult stem cell decline, slow down bodyweight loss, improve trabecular bone structure and mineral density, and significantly extend lifespan.

SUMMARY: Tissue-specific stem cells persist throughout the entire lifespan to repair and maintain tissues, but their self-renewal and differentiation potential become dysregulated with aging. Given that adult stem cells are thought to be central to tissue maintenance and organismal survival, SIRT3 may promote organismal longevity by maintaining the integrity of tissue-speciic stem cells.

KEYWORDS: rejuvenation, aging, stem cell, DNA damage, sirtuin activator


Full Text:

PDF

References


Lopez-Otin C, Blasco MA, Partridge L, Serrano M, Kroemer G. The hallmarks of aging. Cell. 2013; 153: 1194-217, CrossRef.

Della-Morte D, Ricordi C, Rundek T. The fountain of youth: role of sirtuins in aging and regenerative medicine. Regen Med. 2013; 8: 681-3, CrossRef.

Rando TA, Chang HY. Aging, rejuvenation, and epigenetic reprogramming: resetting the aging clock. Cell. 2012; 148: 46-57, CrossRef.

Haigis MC, Yankner BA. The aging stress response. Mol Cell. 2010; 40: 333-44, CrossRef.

Satoh A, Brace CS, Rensing N, Cliften P, Wozniak DF, Herzog ED, et al. Sirt1 extends life span and delays aging in mice through the regulation of Nk2 homeobox 1 in the DMH and LH. Cell Metab. 2013; 18: 416-30, CrossRef.

Chang HC, Guarente L. SIRT1 and other sirtuins in metabolism. Trends Endocrinol Metab. 2014; 25: 138-45, CrossRef.

Guarente L. Aging research-where do we stand and where are we going? Cell. 2014; 159: 15-9, CrossRef.

Brown K, Xie S, Qiu X, Mohrin M, Shin J, Liu Y, et al. SIRT3 reverses aging-associated degeneration. Cell Rep. 2013; 3: 319-27, CrossRef.

Bergmann O, Bhardwaj RD, Bernard S, Zdunek S, Barnabé-Heider F, Walsh S, et al. Evidence for cardiomocyte renewal in humans. Science. 2009; 324: 98-102, CrossRef.

Oberdoerffer P, Sinclair DA. The role of nuclear architecture in genomic instability and aging. Nat Rev Mol Cell Biol. 2007; 8: 692-702, CrossRef.

Fraga MF, Esteller M. Epigenetics and aging: the targets and the marks. Trends Genet. 2007; 23: 413-8, CrossRef.

Shin DM, Kucia M, Ratajczak MZ. Nuclear and chromatin reorganization during cell senescence and aging – a mini-review. Gerontology. 2011; 57: 76-84, CrossRef.

Conboy IM, Rando TA. Aging, stem cells and tissue regeneration: lessons form muscle. Cell Cycle. 2005; 4: 407-10, CrossRef.

Sharpless NE, DePinho RA. Telomeres, stem cells, senescence, and cancer. J Clin Invest. 2004; 113: 160-8, CrossRef.

Bryder D, Rossi DJ, Weissman IL. Hematopoietic stem cells: the paradigmatic tissue specific stem cell. Am J Pathol. 2006; 169: 338-46, CrossRef.

Morrison SJ, Kimble J. Asymmetric and symmetric stem-cell divisions in development and cancer. Nature. 2006; 441: 1068-74, CrossRef.

Meiliana A, Wijaya A. The stem cell hypothesis of aging. Indones Biomed J. 2010; 2: 26-44, CrossRef.

Armstrong L, Al-Aama J, Stojkovic M, Lako M. Concise review: the epigenetic contribution to stem cell ageing: can we rejuvenate our older cells? Stem Cells. 2014; 32: 2291-8, CrossRef.

Raveh-Amit H, Berzsenyi S, Vas V, Ye D, Dinnyes A. Tissue resident stem cells: till death do us part. Biogerontology. 2013; 14: 573-90, CrossRef.

Carlson ME, Conboy IM. Loss of stem cell regenerative capacity within aged niches. Aging Cell. 2007; 6: 371-82, CrossRef.

Sager R. Senescence as a mode of tumor suppression. Environ Health Perspect. 1991; 93: 59-62, CrossRef.

Weinstein BS, Ciszek D. The reserve-capacity hypothesis: evolutionary origins and modern implications of the trade-off between tumor-suppression and tissue-repair. Exp Gerontol. 2002; 37: 615-27, CrossRef.

Pearson BJ, Sanchez Alvarado A. Regeneration, stem cells, and the evolution of tumor suppression. Cold Spring Harb Symp Quant Biol. 2008; 73: 565-72, CrossRef.

Nizetic D, Groet J. Tumorigenesis in Down’s syndrome: big lessons from a small chromosome. Nat Rev Cancer. 2012; 12: 721-32, CrossRef.

Kuranda K, Vargaftig J, de la Rochere P, Dosquet C, Charron D, Bardin F, et al. Age-related changes in human hematopoietic stem/progenitor cells. Aging Cell. 2011; 10: 542-6, CrossRef.

Muller-Sieburg C, Sieburg HB. Stem cell aging: survival of the laziest? Cell Cycle. 2008; 7: 3798-804, CrossRef.

Warren LA, Rossi DJ. Stem cells and aging in the hematopoietic system. Mech Ageing Dev. 2009; 130: 46-53, CrossRef.

Chambers SM, Shaw CA, Gatza C, Fisk CJ, Donehower LA, Goodell MA. Aging hematopoietic stem cells decline in function and exhibit epigenetic dysregulation. PLoS Biol. 2007; 5: e201, 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.

Krishnamurthy J, Ramsey MR, Ligon KL, Torrice C, Koh A, Bonner-Weir S, et al. p16INK4a induces an age-dependent decline in islet regenerative potential. Nature. 2006; 443: 453-7, CrossRef.

Molofsky AV, Slutsky SG, Joseph NM, He S, Pardal R, Krishnamurthy J, et al. Increasing p16INK4a expression decreases forebrain progenitors and neurogenesis during ageing. Nature. 2006; 443: 448-52, CrossRef.

Nishino J, Kim I, Chada K, Morrison SJ. Hmga2 promotes neural stem cell self-renewal in young but not old mice by reducing p16Ink4a and p19Arf Expression. Cell. 2008; 135: 227-39, CrossRef.

Toledano H, D’Alterio C, Czech B, Levine E, Jones DL. The let-7-Imp axis regulates ageing of the Drosophila testis stem-cell niche. Nature. 2012; 485: 605-10, CrossRef.

He S, Nakada D, Morrison SJ. Mechanisms of stem cell self-renewal. Annu Rev Cell Dev Biol. 2009; 25: 377-406, CrossRef.

Sato T, Vries RG, Snippert HJ, van de Wetering M, Barker N, Stange DE, et al. Single Lgr5 stem cells build crypt-villus structures in vitro without a mesenchymal niche. Nature. 2009; 459: 262-5, CrossRef.

Munoz J, Stange DE, Schepers AG, van de Wetering M, Koo BK, Itzkovitz S, et al. The Lgr5 intestinal stem cell signature: robust expression of proposed quiescent ‘+ 4’ cell markers. EMBO J. 2012; 31: 3079-91, CrossRef.

Powell AE, Wang Y, Li Y, Poulin EJ, Means AL, Washington MK, et al. The pan-ErbB negative regulator Lrig1 is an intestinal stem cell marker that functions as a tumor suppressor. Cell. 2012; 149: 146-58, CrossRef.

Reya T, Morrison SJ, Clarke MF, Weissman IL. Stem cells, cancer, and cancer stem cells. Nature. 2001; 414: 105-11, CrossRef.

Liang R, Ghaffari S. Stem cells, redox signaling, and stem cell aging. Antioxid Redox Signal. 2014; 20: 1902-16, CrossRef.

Ahlqvist KJ, Hämäläinen RH, Yatsuga S, Uutela M, Terzioglu M, Götz A, et al. Somatic progenitor cell vulnerability to mitochondrial DNA mutagenesis underlies progeroid phenotypes in Polg mutator mice. Cell Metab. 2012; 15: 100-9, CrossRef.

Bratic A, Larsson NG. The role of mitochondria in aging. J Clin Invest. 2013; 123: 951-7, CrossRef.

Dillin A, Hsu AL, Arantes-Oliveira N, Lehrer-Graiwer J, Hsin H, Fraser AG, et al. Rates of behavior and aging specified by mitochondrial function during development. Science. 2002; 298, 2398-401, CrossRef.

Choudhury AR, Ju Z, Djojosubroto MW, Schienke A, Leche, A, Schaet-zlein S, et al. Cdkn1a deletion improves stem cell function and lifespan of mice with dysfunctional telomeres without accelerating cancer formation. Nat Genet. 2007; 39: 99-105, CrossRef.

Rossi DJ, Bryder D, Seita J, Nussenzweig A, Hoeijmakers J, Weissman IL. Deficiencies in DNA damage repair limit the function of haematopoietic stem cells with age. Nature. 2007; 447: 725-9, CrossRef.

Sahin E, Depinho RA. Linking functional decline of telomeres, mitochondria and stem cells during ageing. Nature. 2010; 464: 520-8, CrossRef.

Lee J, Duan W, Long JM, Ingram DK, Mattson MP. Dietary restriction increases the number of newly generated neural cells, and induces BDNF expression, in the dentate gyrus of rats. J Mol Neurosci. 2000; 15: 99-108, 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, CrossRef.

Mair W, McLeod CJ, Wang L, Jones DL. Dietary restriction enhances germline stem cell maintenance. Aging Cell. 2010; 9: 916-8, CrossRef.

Cerletti M, Jang YC, Finley LW, Haigis MC, Wagers AJ. Short-term calorie restriction enhances skeletal muscle stem cell function. Cell Stem Cell. 2012; 10: 515-9, CrossRef.

Conboy IM, Conboy MJ, Wagers AJ, Girma ER, Weissman IL, Rando TA. Rejuvenation of aged progenitor cells by exposure to a young systemic environment. Nature. 2005; 433: 760-4, CrossRef.

Villeda SA, Luo J, Mosher KI, Zou B, Britschgi M, Bieri G, et al. The ageing systemic milieu negatively regulates neurogenesis and cognitive function. Nature. 2011; 477: 90-4, 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. Nature Rev Genet. 2008; 9: 115-28, CrossRef.

Cheung TH, Rando TA. Molecular regulation of stem cell quiescence. Nat Rev Mol Cell Biol. 2013; 14: 329-40, CrossRef.

Hsu YC, Fuchs E. A family business: stem cell progeny join the niche to regulate homeostasis. Nature Rev Mol Cell Biol. 2012; 13: 103-14, CrossRef.

Florian MC, Geiger H. Concise review: polarity in stem cells, disease, and aging. Stem Cells. 2010; 28: 1623-9, CrossRef.

Morrison SJ, Spradling AC. Stem cells and niches: mechanisms that promote stem cell maintenance throughout life. Cell. 2008; 132: 598-611, CrossRef.

Aichinger E, Kornet N, Friedrich T, Laux T. Plant stem cell niches. Annu Rev Plant Biol. 2012; 63: 615-36, CrossRef.

Schofield R. The relationship between the spleen colony-forming cell and the haemopoietic stem cell. Blood Cells. 1978; 4: 7-25, PMID.

Lander AD, Kimble J, Clevers H, Fuchs E, Montarras D, Buckingham M, et al. What does the concept of the stem cell niche really mean today? BMC Biol. 2012; 10: 19, CrossRef.

Geiger H, de Haan G, Florian C. The ageing haematopoietic stem cell compartment. Nat Rev Immunol. 2013; 13: 376-89, CrossRef.

Lucas D, Scheiermann C, Chow A, Kunisaki Y, Bruns I, Barrick C, et al. Chemotherapy-induced bone marrow nerve injury impairs hematopoietic regeneration. Nat Med. 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, Passegue E. Mechanisms controlling hematopoietic stem cell functions during normal hematopoiesis and hematological malignancies. Wiley Interdiscip Rev 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. J Exp Med. 2005; 202: 1599-611, CrossRef.

Essers MA, 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-gamma 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.

Chamberlain G, Fox J, Ashton B, Middleton J. Concise review: mesenchymal stem cells: their phenotype, differentiation capacity, immunological features, and potential for homing. Stem Cells. 2007; 25: 2739-49, CrossRef.

Noth U, Rackwitz L, Steinert AF, Tuan RS. Cell delivery therapeutics for musculoskeletal regeneration. Adv Drug Deliv Rev. 2010; 62: 765-83, CrossRef.

Chen Y, Shao JZ, Xiang LX, Dong XJ, Zhang GR. Mesenchymal stem cells: a promising candidate in regenerative medicine. Int J Biochem Cell Biol. 2008; 40: 815-20, CrossRef.

Wu XB, Tao R. Hepatocyte differentiation of mesenchymal stem cells. Hepatobiliary Pancreat Dis Int. 2012; 11: 360-71, CrossRef.

Pittenger MF, Mackay AM, Beck SC, Jaiswal RK, Douglas R, Mosca JD, et al. Multilineage potential of adult human mesenchymal stem cells. Science. 1999; 284: 143-7, CrossRef.

Tchkonia T, Zhu Y, van Deursen J, Campisi J, Kirkland JL. Cellular senescence and the senescent secretory phenotype: therapeutic opportunities. J Clin Invest. 2013; 123: 966-72, CrossRef.

Campisi J, d’Adda di Fagagna F. Cellular senescence: when bad things happen to good cells. Nat Rev Mol Cell Biol. 2007; 8: 729-40, CrossRef.

Suram A, Kaplunov J, Patel PL, Ruan H, Cerutti A, Boccardi V, et al. Oncogene-induced telomere dysfunction enforces cellular senescence in human cancer precursor lesions. EMBO J. 2012; 31: 2839-51, CrossRef.

Chen JH, Stoeber K, Kingsbury S, Ozanne SE, Williams GH, Hales CN. Loss of proliferative capacity and induction of senescence in oxidatively stressed human fibroblasts. J Biol Chem. 2004; 279: 49439-46, CrossRef.

Courtois-Cox S, Jones SL, Cichowski K. Many roads lead to oncogene-induced senescence. Oncogene. 2008; 27: 2801-9, CrossRef.

Braig M, Lee S, Loddenkemper C, Rudolph C, Peters AH, Schlegelberger B, et al. Oncogene-induced senescence as an initial barrier in lymphoma development. Nature. 2005; 436: 660-5, CrossRef.

Haigis KM, Sweet-Cordero A. New insights into oncogenic stress. Nat Genet. 2011; 43: 177-8, CrossRef.

Braig M, Schmitt CA. Oncogene-induced senescence: putting the brakes on tumor development. Cancer Res. 2006; 66: 2881-4, CrossRef.

Prieur A, Peeper DS. Cellular senescence in vivo: a barrier to tumorigenesis. Curr Opin Cell Biol. 2008; 20: 150-5, CrossRef.

Campisi J. Cellular senescence as a tumor-suppressor mechanism. Trends Cell Biol. 2001; 11: S27-31, CrossRef.

Guerra C, Collado M, Navas C, Schuhmacher AJ, Hernández-Porras I, Cañamero M, et al. Pancreatitis-induced inflammation contributes to pancreatic cancer by inhibiting oncogene-induced senescence. Cancer Cell. 2011; 19: 728-39, CrossRef.

Jeyapalan JC, Sedivy JM. Cellular senescence and organismal aging. Mech Ageing Dev. 2008; 129: 467-74, CrossRef.

Tchkonia T, Morbeck DE, Von Zglinicki T, Van Deursen J, Lustgarten J, Scrable H, et al. Fat tissue, aging, and cellular senescence. Aging Cell. 2010; 9: 667-84, CrossRef.

Ksiazek K, Mikula-Pietrasik J, Olijslagers S, Jorres A, von Zglinicki T, Witowski J. Vulnerability to oxidative stress and different patterns of senescence in human peritoneal mesothelial cell strains. Am J Physiol Regul Integr Comp Physiol. 2009; 296: R374-82, CrossRef.

Weyemi U, Lagente-Chevallier O, Boufraqech M, Prenois F, Courtin F, Caillou B, et al. ROS-generating NADPH oxidase NOX4 is a critical mediator in oncogenic H-Ras-induced DNA damage and subsequent senescence. Oncogene. 2012; 31: 1117-29, CrossRef.

Passos JF, Nelson G, Wang C, Richter T, Simillion C, Proctor CJ, et al. Feedback between p21 and reactive oxygen production is necessary for cell senescence. Mol Syst Biol. 2010; 6: 347, CrossRef.

Sitte N, Merker K, Von Zglinicki T, Grune T, Davies KJ. Protein oxidation and degradation during cellular senescence of human BJ fibroblasts: part I — effects of proliferative senescence. FASEB J. 2000; 14: 2495-502, CrossRef.

Kuilman T, Michaloglou C, Mooi WJ, Peeper DS. The essence of senescence. Genes Dev. 2010; 24: 2463-79, CrossRef.

Kuilman T, Michaloglou C, Vredeveld LC, Douma S, van Doorn R, Desmet CJ, et al. Oncogene-induced senescence relayed by an interleukin-dependent inflammatory network. Cell. 2008; 133: 1019-31, CrossRef.

Acosta JC, O'Loghlen A, Banito A, Guijarro MV, Augert A, Raguz S, et al. Chemokine signaling via the CXCR2 receptor reinforces senescence. Cell. 2008; 133: 1006-18, CrossRef.

Freund A, Orjalo AV, Desprez PY, Campisi J. Inflammatory networks during cellular senescence: causes and consequences. Trends Mol Med. 2010; 16: 238-46, CrossRef.

Campisi J, Andersen JK, Kapahi P, Melov S. Cellular senescence: a link between cancer and age-related degenerative disease? Semin Cancer Biol. 2011; 21: 354-9, CrossRef.

Rodier F, Campisi J. Four faces of cellular senescence. J Cell Biol. 2011; 192: 547-56, CrossRef.

De Cecco M, Jeyapalan J, Zhao X, Tamamori-Adachi M, Sedivy JM. Nuclear protein accumulation in cellular senescence and organismal aging revealed with a novel single-cell resolution fluorescence microscopy assay. Aging (Albany NY). 2011; 3: 955-67, PMID.

Xue W, Zender L, Miething C, Dickins RA, Hernando E, Krizhanovsky V, et al. Senescence and tumour clearance is triggered by p53 restoration in murine liver carcinomas. Nature. 2007; 445: 656-60, CrossRef.

Krizhanovsky V, Yon M, Dickins RA, Hearn S, Simon J, Miething C, et al. Senescence of activated stellate cells limits liver fibrosis. Cell. 2008; 134: 657-67, CrossRef.

Coppe JP, Patil CK, Rodier F, Krtolica A, Beauséjour CM, Parrinello S, et al. A human-like senescence-associated secretory phenotype is conserved in mouse cells dependent on physiological oxygen. PLoS One. 2010; 5: e9188, 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.

Rodier F, Coppé JP, Patil CK, Hoeijmakers WA, Muñoz DP, Raza SR, et al. Persistent DNA damage signalling triggers senescence-associated inflammatory cytokine secretion. Nat Cell Biol. 2009; 11: 973-9, CrossRef.

Baker DJ, Wijshake T, Tchkonia T, LeBrasseur NK, Childs BG, van de Sluis B, et al. Clearance of p16Ink4a-positive senescent cells delays ageing-associated disorders. Nature. 2011; 479: 232-6, CrossRef.

Kuilman T, Peeper DS. Senescence-messaging secretome: SMS-ing cellular stress. Nature Rev Cancer. 2009; 9: 81-94, CrossRef.

Munoz-Espin D, Serrano M. Cellular senescence: from physiology to pathology. Nat Rev Mol Cell Biol. 2014; 15: 482-96, CrossRef.

Nagaria P, Robert C, Rassool FV. DNA double-strand break response in stem cells: mechanisms to maintain genomic integrity. Biochim Biophys Acta. 2013; 1830: 2345-53, CrossRef.

Beausejour CM, Campisi J. Ageing: balancing regeneration and cancer. Nature. 2006; 443: 404-5, CrossRef.

Rodier F, Campisi J. Four faces of cellular senescence. J Cell Biol. 2011; 192: 547-56, CrossRef.

Liu Y, Johnson SM, Fedoriw Y, Rogers AB, Yuan H, Krishnamurthy J et al. Expression of p16(ink4a) prevents cancer and promotes aging in lymphocytes. Blood. 2011; 117: 3257-67, CrossRef.

Stochaj U, Kodiha M, Shum-Tim D, Colmegna I. Implications of multipotent mesenchymal stromal cell aging. Regen Med. 2013; 8: 211-22, CrossRef.

van Deursen JM. The role of senescent cells in ageing. Nature. 2014; 509: 439-46, CrossRef.

Naylor RM, Baker DJ, van Deursen JM. Senescent cells: A novel therapeutic target for aging and age-related diseases. Clin Pharmacol Ther. 2013; 93: 105-16, CrossRef.

Verzola D, Gandolfo MT, Gaetani G, Ferraris A, Mangerini R, Ferrario F, et al. Accelerated senescence in the kidneys of patients with type 2 diabetic nephropathy. Am J Physiol Renal Physiol. 2008; 295: F1563-73, CrossRef.

De Cabo R, Carmona-Gutierrez D, Bernier M, Hall MN, Madeo F. The search for antiaging interventions: from elixirs to fasting regimens. Cell. 2014; 157: 1515-26, CrossRef.

Willcox BJ, Willcox DC. Caloric restriction, caloric restriction mimetics, and healthy aging in Okinawa: controversies and clinical implications. Curr Opin Clin Nutr Metab Care. 2014; 17: 51-8, CrossRef.

Fontana L, Partridge L, Longo VD. Extending healthy life span - from yeast to humans. Science. 2010; 328: 321-6, CrossRef.

Haigis MC, Guarente LP. Mammalian sirtuin - emerging roles in physiology, aging, and calorie restriction. Genes Dev. 2006; 20: 2913-21, CrossRef.

Hebert AS, Dittenhafer-Reed KE, Yu W, Bailey DJ, Selen ES, Boersma MD, et al. Calorie restriction and SIRT3 trigger global reprogramming of the mitochondrial protein acetylome. Mol Cell. 2013; 49: 186-99, CrossRef.

Kahn BB, Alquier T, Carling D, Hardie DG. 2005. AMP-activated protein kinase: ancient energy gauge provides clues to modern understanding of metabolism. Cell Metab. 2005; 1: 15-25, CrossRef.

Johnson SM, Rabinovitch PS, Kaeberlein M. mTOR is a key modulator of aging and age-related disease. Nature. 2013; 493: 338-45, CrossRef.

Kenyon CJ. The genetics of aging. Nature. 2010; 464: 504-12, CrossRef.

Guarente L. Calorei restriction and sirtuins revisited. Gen Dev. 2013; 27: 2072-85, CrossRef.

Rodgers JT, Lerin C, Haas W, Gygi SP, Spiegelman BM, Puigserver P. Nutrient control of glucose homeostasis through a complex of PGC-1alpha and SIRT1. Nature. 2005; 434: 113-8, CrossRef.

Gerhart-Hines Z, Rodgers JT, Bare O, Lerin C, Kim SH, Mostoslavsky R, et al. Metabolic control of muscle mitochondrial function and fatty acid oxidation through SIRT1/PGC-1alpha. EMBO J. 2007; 26: 1913-23, CrossRef.

Brunet A, Sweeney LB, Sturgill JF, Chua KF, Greer PL, Lin Y, et al. Stress-dependent regulation of FOXO transcription factors by the SIRT1 deacetylase. Science. 2004; 303: 2011-5, CrossRef.

Motta MC, Divecha N, Lemieux M, Kamel C, Chen D, Gu W, et al. Mammalian SIRT1 represses forkhead transcription factors. Cell. 2004; 116: 551-63, CrossRef.

Purushotham A, Schug TT, Xu Q, Surapureddi S, Guo X, Li X. Hepatocyte-specific deletion of SIRT1 alters fatty acid metabolism and results in hepatic steatosis and inflammation. Cell Metab. 2009; 9: 327-38, CrossRef.

Feige JN, Lagouge M, Canto C, Strehle A, Houten SM, Milne JC, et al. Specific SIRT1 activation mimics low energy levels and protects against diet-induced metabolic disorders by enhancing fat oxidation. Cell Metab. 2008; 8: 347-58, CrossRef.

Fulco M, Cen Y, Zhao P, Hoffman EP, McBurney MW, Sauve AA, et al. Glucose restriction inhibits skeletal myoblast differentiation by activating SIRT1 through AMPK-mediated regulation of Nampt. Dev Cell. 2008; 14: 661-73, CrossRef.

Canto C, Gerhart-Hines Z, Feige JN, Lagouge M, Noriega L, Milne JC, et al. AMPK regulates energy expenditure by modulating NAD+ metabolism and SIRT1 activity. Nature. 2009; 458: 1056-60, CrossRef.

Canto C, Jiang LQ, Deshmukh AS, Mataki C, Coste A, Lagouge M, et al. Interdependence of AMPK and SIRT1 for metabolic adaptation to fasting and exercise in skeletal muscle. Cell Metab. 2010; 11: 213-9, CrossRef.

Hou X, Xu S, Maitland-Toolan KA, Sato K, Jiang B, Ido Y, et al. SIRT1 regulates hepatocyte lipid metabolism through activating AMP-activated protein kinase. J Biol Chem. 2008; 283: 20015-26, CrossRef.

Lan F, Cacicedo JM, Ruderman N, Ido Y. SIRT1 modulation of the acetylation status, cytosolic localization, and activity of LKB1. Possible role in AMP-activated protein kinase activation. J Biol Chem. 2008; 283: 27628-35, CrossRef.

Hallows WC, Yu W, Denu JM. Regulation of glycolytic enzyme phosphoglycerate mutase-1 by Sirt1 protein-mediated deacetylation. J Biol Chem. 2012; 287: 3850-8, CrossRef.

Lim JH, Lee YM, Chun YS, Chen J, Kim JE, Park JW. Sirtuin 1 modulates cellular responses to hypoxia by deacetylating hypoxia-inducible factor 1alpha. Mol Cell. 2010; 38: 864-78, CrossRef.

Adler AS, Sinha S, Kawahara TL, Zhang JY, Segal E, Chang HY. Motif module map reveals enforcement of aging by continual NF-kappaB activity. Genes Dev. 2007; 21: 3244-57, CrossRef.

Asher G, Gatfield D, Stratmann M, Reinke H, Dibner C, Kreppel F, et al. SIRT1 regulates circadian clock gene expression through PER2 deacetylation. Cell. 2008; 134: 317-28, CrossRef.

Nakahata Y, Kaluzova M, Grimaldi B, Sahar S, Hirayama J, Chen D, et al. The NAD+-dependent deacetylase SIRT1 modulates CLOCK-mediated chromatin remodeling and circadian control. Cell. 2008; 134: 329-40, CrossRef.

Nakahata Y, Sahar S, Astarita G, Kaluzova M, Sassone-Corsi P. Circadian control of the NAD+ salvage pathway by CLOCK-SIRT1. Science. 2009; 324: 654-7, CrossRef.

Ramsey KM, Yoshino J, Brace CS, Abrassart D, Kobayashi Y, Marcheva B, et al. Circadian clock feedback cycle through NAMPT-mediated NAD+ biosynthesis. Science. 2009; 324: 651-4, CrossRef.

Chang HC, Guarente L. SIRT1 mediates central circadian control in the SCN by a mechanism that decays with aging. Cell. 2013; 153: 1448-60, CrossRef.

Valentinuzzi VS, Scarbrough K, Takahashi JS, Turek FW. Effects of aging on the circadian rhythm of wheel-running activity in C57BL/6 mice. Am J Physiol. 1997; 273: R1957-64, PMID.

Howitz KT, Bitterman KJ, Cohen HY, Lamming DW, Lavu S, Wood JG, et al. Small molecule activators of sirtuins extend Saccharomyces cerevisiae lifespan. Nature. 2003; 425: 191-6, CrossRef.

Jimenez-Gomez Y, Mattison JA, Pearson KJ, Martin-Montalvo A, Palacios HH, Sossong AM, et al. Resveratrol improves adipose insulin signaling and reduces the inflammatory response in adipose tissue of rhesus monkeys on high-fat, high-sugar diet. Cell Metab. 2013; 18: 533-45, CrossRef.

Fiori JL, Shin YK, Kim W, Krzysik-Walker SM, González-Mariscal I, Carlson OD, et al. Resveratrol prevents beta-cell dedifferentiation in nonhuman primates given a high-fat/high-sugar diet. Diabetes. 2013; 62: 3500-13, CrossRef.

Mattison JA, Wang M, Bernier M, Zhang J, Park SS, Maudsley S, et al. Resveratrol prevents high fat/sucrose diet-induced central arterial wall inflammation and stiffening in nonhuman primates. Cell Metab. 2014; 20: 183-90, CrossRef.

Baur JA, Pearson KJ, Price NL, Jamieson HA, Lerin C, Kalra A, et al. Resveratrol improves health and survival of mice on a high-calorie diet. Nature. 2006; 444: 337-42, CrossRef.

Bauer JH, Goupil S, Garber GB, Helfand SL. An accelerated assay for the identification of lifespan-extending interventions in Drosophila melanogaster. Proc Natl Acad Sci USA. 2004; 101: 12980-5, CrossRef.

Baur JA, Sinclair DA. Therapeutic potential of resveratrol: the in vivo evidence. Nat Rev Drug Discov. 2006; 5: 493-506, CrossRef.

Timmers S, Auwerx J, Schrauwen P. The journey of resveratrol from yeast to human. Aging (Albany NY). 2012; 4: 146-58, PMID.

Lagouge M, Argmann C, Gerhart-Hines Z, Meziane H, Lerin C, Daussin F, et al. Resveratrol improves mitochondrial function and protects against metabolic disease by activating SIRT1 and PGC-1alpha. Cell. 2006; 127: 1109-22, CrossRef.

Beher D, Wu J, Cumine S, Kim KW, Lu SC, Atangan L, et al. Resveratrol is not a direct activator of SIRT1 enzyme activity. Chem Biol Drug Des. 2009; 74: 619-24, CrossRef.

Kaeberlein M, McVey M, Guarente L. The SIR2/3/4 complex and SIR2 alone promote longevity in Saccharomyces cerevisiae by two different mechanisms. Genes Dev. 1999; 13: 2570-80, CrossRef.

Mouchiroud L, Houtkooper RH, Moullan N, Katsyuba E, Ryu D, Cantó C, et al. The NAD(+)/Sirtuin Pathway Modulates Longevity through Activation of Mitochondrial UPR and FOXO Signaling. Cell . 2013; 154: 430-41, CrossRef.

Oh J, Lee YD, Wagers AJ. Stem cell aging: mechanisms, regulators and therapeutic opportunities. Nat Med. 2014; 20: 870-80, CrossRef.

Passier R, Mummery C. Origin and use of embryonic and adult stem cells in differentiation and tissue repair. Cardiovasc Res. 2003; 58: 324-35, CrossRef.

Mimeault M, Hauke R, Batra SK. Stem cells: A revolution in therapeutics-recent advances in stem cell biology and their therapeutic applications in regenerative medicine and cancer therapies. Clin Pharmacol Ther. 2007; 82: 252-64, CrossRef.

Saini A, Mastana S, Myers F, Lewis MP. ‘From death, lead me to immortality - Mantra of ageing skeletal muscle. Curr Genomics. 2013; 14: 256-67, CrossRef.

Brack AS, Conboy MJ, Roy S, Lee M, Kuo CJ, Keller C, et al. Increased Wnt signaling during aging alters muscle stem cell fate and increases fibrosis. Science. 2007; 317: 807-10, CrossRef.

Rodier F, Coppé JP, Patil CK, Hoeijmakers WA, Muñoz DP, Raza SR, et al. Persistent DNA damage signalling triggers senescence-associated inflammatory cytokine secretion. Nat Cell Biol. 2009; 11: 973-9, CrossRef.

Kuhn HG, Dickinson-Anson H, Gage FH. Neurogenesis in the dentate gyrus of the adult rat: age-related decrease of neuronal progenitor proliferation. J Neurosci. 1996; 16: 2027-33, PMID.

Maslov AY, Barone TA, Plunkett RJ, Pruitt SC. Neural stem cell detection, characterization, and age-related changes in the subventricular zone of mice. J Neurosci. 2004; 24: 1726-33, CrossRef.

Seib DR, Corsini NS, Ellwanger K, Plaas C, Mateos A, Pitzer C, et al. Loss of Dickkopf-1 restores neurogenesis in old age and counteracts cognitive decline. Cell Stem Cell. 2013; 12: 204-14, CrossRef.

Balaban RS, Nemoto S, Finkel T. Mitochondria, oxidants, and aging. Cell. 2005; 120: 483-95, CrossRef.

Karpac J, Jasper H. Aging: seeking mitonuclear balance. Cell. 2013; 154: 271-3, CrossRef.

Hubbard BP, Gomes AP, Dai H, Li J, Case AW, Considine T, et al. Evidence for a common mechanism of SIRT1 regulation by allosteric activators. Science. 2013; 339: 1216-9, CrossRef.

Dai H, Kustigian L, Carney D, Case A, Considine T, Hubbard BP, et al. SIRT1 activation by small molecules: kinetic and biophysical evidence for direct interaction of enzyme and activator. J Biol Chem. 2010; 285: 32695-703, CrossRef.




DOI: https://doi.org/10.18585/inabj.v7i2.72

Copyright (c) 2015 The Prodia Education and Research Institute

Creative Commons License
This work is licensed under a Creative Commons Attribution-NonCommercial 4.0 International License.

 

Indexed by:

                  

               

                   

 

 

The Prodia Education and Research Institute