Skip Navigation
Skip to contents

Diabetes Metab J : Diabetes & Metabolism Journal

Search
OPEN ACCESS

Articles

Page Path
HOME > Diabetes Metab J > Volume 37(5); 2013 > Article
Review
Pathophysiology The Interplay between Autophagy and Aging
Jong-Ok Pyo, Seung-Min Yoo, Yong-Keun Jung
Diabetes & Metabolism Journal 2013;37(5):333-339.
DOI: https://doi.org/10.4093/dmj.2013.37.5.333
Published online: October 17, 2013
  • 3,661 Views
  • 37 Download
  • 25 Crossref
  • 34 Scopus

Bio-MAX Institute, Seoul National University School of Biological Sciences, Seoul, Korea.

Corresponding author: Yong-Keun Jung. Global Research Laboratory, Bio-MAX Institute, Seoul National University School of Biological Sciences, 1 Gwanak-ro, Gwanak-gu, Seoul 151-747, Korea. ykjung@snu.ac.kr

Copyright © 2013 Korean Diabetes Association

This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/3.0/) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

  • Numerous studies have established a link between autophagy and aging; however, the relationship has not been clearly defined. Aging is a very complex process caused by the accumulation of various factors due to the gradual failure of cellular maintenance. Recent studies have shown that autophagy reduces the stress responses induced by starvation, reactive oxygen species, and the accumulation of intracellular proteins and organelles through cytoprotection, clearance of damaged mitochondria, and lysosomal degradation. Here, we summarize our current understanding of the relationship between autophagy and the aging process.
Autophagy is a catabolic process in eukaryotic cells that delivers cytosolic substrates to the lysosome for degradation. Autophagy consists of several steps: 1) induction and nucleation, 2) elongation of phagophores and sequestration of cytosolic components through autophagosome formation, 3) transport to the lysosome, 4) degradation, and 5) utilization of degradation products [1]. Despite the simplicity of this process, autophagy plays a role in various physiological processes, including cell growth, cell differentiation, cell survival, and immune responses, as well as in pathological diseases, including cancer, neurodegeneration, and metabolic syndrome [2]. Numerous studies also suggest that autophagy is closely related to aging through its housekeeping function in the degradation and recycling of cytoplasmic components and damaged organelles. Recent studies have emphasized the role of autophagy as a key regulator of the aging process, especially through the removal of damaged mitochondria. Autophagy is also associated with well-known longevity-promoting signals, including caloric restriction (CR), insulin/insulin-like growth factor (IGF)-1 signaling, and the p53 pathway [3]. Therefore, it is clear that autophagy contributes to the overall health and lifespan of individual cells as well as the whole organism. Here, we describe and discuss the likely relationships between autophagy and aging, as well as the potential molecular mechanism underlying the regulation of aging by autophagy.
Cells are constantly exposed to internal and external stresses, which can cause problems for the maintenance of homeostasis and functional integrity and can eventually lead to cell death. Therefore, understanding the protective pathway against death induced by a variety of stresses is useful for deciphering the aging process [4]. An important function of autophagy is a self-limited survival strategy, which is a cytoprotective mechanism against starvation and oxidative stress due to mitochondrial dysfunction, aggregate-prone proteins, and pathogens [5]. Under prolonged starvation, autophagy plays a crucial role in maintaining an amino acid pool for survival [2]. Mice with a knockout of atg5, an essential gene for membrane elongation in autophagy, are normal at birth but show severe nutrient and energy insufficiency within 10 hours after birth and have significantly shorter survival times than wild-type mice under starvation conditions [6]. Moreover, beclin-1 or atg8 RNAi-mediated inhibition of autophagy in Caenorhabditis elegans causes a failure to survive during dauer diapause [7]. This implies that recycling via autophagy is critical for the maintenance of cellular energy homeostasis and survival.
Recent studies suggest that well-known aging regulators, such as SIRT1 and DAF-16/FOXO, are required to induce autophagy under starvation conditions. For example, expression of SIRT1, an NAD+-dependent deacetylase [8], is increased during starvation [9] and activates autophagy by deacetylating Atg5, Atg7, and Atg8 under starvation conditions in both nematodes and mammals [10,11]. Moreover, SIRT1 increases the median and maximum lifespan of C. elegans. The transcription factor DAF-16/FOXO, which is known to extend lifespan through its cytoprotective effects, is also involved in autophagy activation [12]. Deficiency of p53, a tumor suppressor that influences lifespan, can also induce autophagy in mammalian systems [13]. These findings point to a molecular connection between the aging process and the cytoprotective activity of autophagy.
Autophagy may contribute to lifespan expansion through regulation of reactive oxygen species (ROS) production. It is commonly known that even a tolerable level of ROS induces autophagy, which in turn reduces oxidative damage in cancer, cardiac, and neurodegenerative disease [14,15]. For example, starvation increases ROS, which regulates autophagy through adenosine monophosphate-activated protein kinase (AMPK) activation [16]. ROS itself accumulates in Atg5- and Atg7-knockout mice [17] and in Atg5- and Atg10-knockdown cells under starvation conditions [18]. Interestingly, ATG4, an essential protease in the autophagy process, was identified as a direct target for oxidation by hydrogen peroxide, which activates autophagy [19]. Numerous studies have shown direct evidence that continuous oxidative stress occurs during the aging process [20], which might disrupt protein turnover [21]. In fact, enhanced autophagy eliminates oxidative stress through enhanced lysosome activity and alteration of the luminal pH, and/or direct regulation of lysosomal enzymes [22] and removal of damaged mitochondria [23]. While the relationship between autophagy and ROS production is still a subject of debate, autophagy apparently functions to remove ROS generated by starvation and/or aging. Taken together, these studies clearly demonstrate that autophagy plays a critical role in the aging process through the regulation of ROS.
Aging is characterized by inefficiency and even failure of cellular maintenance, repair, and turnover mechanisms, which results in the accumulation of damaged substances or organelles in aged cells [24]. The primary function of autophagy is protein and organelle turnover for the maintenance of homeostasis. During aging, protein turnover slows down, and aggregate-prone proteins, such as mutant α-synuclein, tau, and mutant huntingtin which cause Parkinson, Alzheimer, and Huntington diseases, respectively, accumulate [25]. In fact, ATG5, ATG7, and BECN-1 expression is down-regulated in the aged human brain [26]; therefore, autophagy activity is likely down-regulated with aging. Rapamycin, an inhibitor of mammalian target of rapamycin (mTOR) and autophagy inducer, boosts the clearance of mutant huntingtin fragments and attenuates toxicity in cells [27]. Similarly, rapamycin reduces tau toxicity in Drosophila melanogaster as well as the appearance of aggregates and cell death associated with poly-glutamine expansion in D. melanogaster and mammals [28,29]. In contrast, inhibition of autophagy increases mutant huntingtin aggregates [30,31]. In Alzheimer disease (AD), an AD-causing mutation in presenilin disrupts autophagy activity [32], and deletion of beclin-1 leads to neurodegeneration and amyloid β accumulation via decreased neuronal autophagy and disruption of lysosomes [33].
The proteolytic activity of the ubiquitin proteasome system (UPS), which is the major degradation pathway for short-lived and soluble proteins, declines during aging [34-36]. Similar to the early aging phenotype of animals with UPS impairment [36,37], a genetic model with reduced autophagy activity also exhibits an accelerated aging phenotype [38]. These observations led to the proposal that increased proteolytic activity of autophagy and UPS may delay aging [39,40]. Consistently, genetic ablation of p62, an ubiquitin- and LC3-binding protein that is involved in the clearance of ubiquitin-conjugates [41], enhances the formation of ubiquitin-positive protein aggregates, resulting in liver injury and neurodegeneration in autophagy-deficient mice [17] and accelerated presentation of ageing phenotypes [42]. More recently, Pyo et al. [43] presented direct evidence that overexpression of Atg5 in mice activates autophagy and significantly extends lifespan. Therefore, it is generally believed that degradation of misfolded and aggregate-prone proteins-which are apparently responsible for aging and aging-related diseases when they accumulate-is accomplished through the modulation of autophagy. While impairment of UPS might induce compensatory activation of autophagy in cells [44,45], their crosstalk in the aging process of animal model is not yet clear.
One of the major functions of autophagy is the degradation of excess or injured organelles [1]. Accumulated mitochondrial damage leads to mitochondrial dysfunction, a common feature of aging that involves changes in mitochondria membrane potential, ROS and adenosine triphosphate (ATP) production, and calcium homeostasis. Under such conditions, cells usually overcome the damage by inducing autophagy to remove the dysregulated mitochondria [46]. Mitophagy, a mitochondrial-selective type of autophagy, removes the damaged mitochondria for quality control. This process was first described in yeast, where ATG32 was discovered as a key receptor in mitophagy that interacts with ATG8/LC3. Extensive studies in mammalian systems revealed that mitophagy has unique machinery and is mediated by PINK1, an outer mitochondrial membrane (OMM) kinase, and PARKIN, a cytosolic E3 ubiquitin ligase [47]. Upon induction of mitophagy, cytosolic PARKIN is recruited to the damaged mitochondria destined for degradation by PINK1 on the OMM, which then induces mitophagy to remove the damaged mitochondria [48].
Despite the expectation of a connection between mitophagy and aging, there is little evidence showing a direct correlation. However, a limited number of studies have demonstrated an association between mitophagy and aging. A defect in mitophagy is apparently associated with Parkinson disease caused by loss of function mutations in PINK1 or PARK2. Several mutations in parkin and PINK1 in Parkinson disease display defects in parkin-induced mitophagy [49,50]. Interestingly, parkin-knockout mice have reduced lifespan and receive less neuroprotection against aging [51]. In addition, Uth1p, one of the four youth proteins on the OMM, participates in mitophagy and prolongs the lifespan of yeast under starvation through regulation of oxidative stress [52]. Deficiency in CISD2, which is involved in mammalian lifespan control and is a causative gene for Wolfram syndrome 2, leads to mitochondrial degeneration, which appears to induce autophagy [53]. In addition to damaged mitochondria quality control, mitophagy is also involved in the steady-state turnover of mitochondria which removes undamaged mitochondrial during developmental processes. In most mammals, mature red blood cells lack mitochondria, which are removed by the mitophagy-mediated action of NIP3-like protein X during maturation [54]. Therefore, the effect of mitophagy on aging should be determined through distinct studies.
Although there are many sites of ROS production in the cell, mitochondria are a major source of ROS. Because mitochondrial proteins and mitochondrial DNA are easily exposed to oxidative damage, and mitochondrial damage in turn stimulates ROS generation, this continuous process eventually allows cells to age and die [55]. The murine lifespan is extended by the overexpression of catalase in mitochondria [56] and by superoxide dismutase/catalase mimetics and detoxification of mitochondrial ROS [57]. Although it is becoming clear that mitophagy is critical for the quality control of mitochondria, a clear connection among mitophagy, mitochondrial quality control, turnover, and the aging process remains to be fully addressed.
Among the many factors that influence lifespan, such as oxidative stress, DNA damage, genetic programming, and the environment, CR is the best-known factor that extends lifespan in various organisms and reduces the pathogenesis of age-related diseases, including diabetes, cardiovascular disease, cancer, and neurodegeneration [58,59]. It has been reported that CR induces a maximum rate of autophagy proteolysis in rat liver [60]. Interestingly, CR can extend lifespan in model organisms by inducing autophagy and reducing mTOR and protein kinase A (PKA) activity, and protein kinase B (PKB)/Sch9 signaling [61]. Autophagy is required for lifespan extension of an eat-2 deletion mutant, a CR model in C. elegans [62], and knockdown of beclin-1 and atg7 suppresses the longevity associated with CR [63]. Conversely, forced activation of autophagy in Atg5 transgenic mice is accompanied by less obesity than their age-matched littermates [43]. These observations illustrate that the atg genes, which are essential in autophagy, are also critical for lifespan extension by CR, indicating a strong correlation between CR and autophagy.
In addition, more evidence has shown a functional correlation between autophagy and energy levels. SIRT1, which is essential for lifespan extension through CR via the insulin/IGF pathway in mice [64], is also a potent autophagy inducer [9]. AMPK, another energy sensor, functions to extend lifespan in C. elegans and induces autophagy in human cell lines [65]. Sch9 serine/threonine protein kinase, a nutrient sensor, is also involved in lifespan extension and modulation of autophagy in yeast [66]. Based on these findings, we know better than before that the established signals regulating the aging process and energy level can also coordinate autophagy activity, drawing more attention to the role of autophagy in lifespan control.
In this review, we have elucidated the functional relationship between autophagy and aging while encompassing the recent discoveries. Autophagy is now believed to be a pathway regulating healthy aging and lifespan, although the relationship between the subordinate and the superior is not clear. Increasing evidence illustrates that many aging pathways can control autophagy activity and that autophagy can regulate aging in various organisms. Although a detailed molecular mechanism linking autophagy to aging is not yet clear, the crucial roles of autophagy in aging seem to be associated with the removal of factors affecting the aging process: for example, cytoprotection against stresses, such as ROS, and the clearance of misfolded or aggregate prone-proteins and damaged subcellular organelles, such as the mitochondria. In particular, the understanding that the regulation of aging by CR is apparently associated with autophagy is a considerable advancement. There are also accumulating reports demonstrating autophagy control through genetic or pharmacological manipulation. Nonetheless, we currently lack a suitable chemical or genetic modulator that selectively activates autophagy that could be used as a lifespan extender in mammals. Therefore, the discovery of an autophagy regulator is the best strategy to pursue in healthy aging and improve lifespan.
Acknowledgements
Dr. Jong-Ok Pyo and Seung-Min Yoo were partly supported by the Brain Korea 21 program. This work was supported by the grants of Global Research Laboratory program (Yong-Keun Jung) and Basic Research for Woman Scientists (Jong-Ok Pyo) of the Korea Research Foundation.

No potential conflict of interest relevant to this article was reported.

  • 1. Yoshimori T. Autophagy: a regulated bulk degradation process inside cells. Biochem Biophys Res Commun 2004;313:453-458. ArticlePubMed
  • 2. Ravikumar B, Sarkar S, Davies JE, Futter M, Garcia-Arencibia M, Green-Thompson ZW, Jimenez-Sanchez M, Korolchuk VI, Lichtenberg M, Luo S, Massey DC, Menzies FM, Moreau K, Narayanan U, Renna M, Siddiqi FH, Underwood BR, Winslow AR, Rubinsztein DC. Regulation of mammalian autophagy in physiology and pathophysiology. Physiol Rev 2010;90:1383-1435. ArticlePubMed
  • 3. Madeo F, Tavernarakis N, Kroemer G. Can autophagy promote longevity? Nat Cell Biol 2010;12:842-846. ArticlePubMedPDF
  • 4. Warner HR, Hodes RJ, Pocinki K. What does cell death have to do with aging? J Am Geriatr Soc 1997;45:1140-1146. ArticlePubMed
  • 5. Kroemer G, Levine B. Autophagic cell death: the story of a misnomer. Nat Rev Mol Cell Biol 2008;9:1004-1010. ArticlePubMedPMCPDF
  • 6. Kuma A, Hatano M, Matsui M, Yamamoto A, Nakaya H, Yoshimori T, Ohsumi Y, Tokuhisa T, Mizushima N. The role of autophagy during the early neonatal starvation period. Nature 2004;432:1032-1036. ArticlePubMedPDF
  • 7. Edinger AL, Cinalli RM, Thompson CB. Rab7 prevents growth factor-independent survival by inhibiting cell-autonomous nutrient transporter expression. Dev Cell 2003;5:571-582. ArticlePubMed
  • 8. Frye RA. Characterization of five human cDNAs with homology to the yeast SIR2 gene: Sir2-like proteins (sirtuins) metabolize NAD and may have protein ADP-ribosyltransferase activity. Biochem Biophys Res Commun 1999;260:273-279. ArticlePubMed
  • 9. Barzilai N, Ferrucci L. Insulin resistance and aging: a cause or a protective response? J Gerontol A Biol Sci Med Sci 2012;67:1329-1331. ArticlePubMed
  • 10. Lee IH, Cao L, Mostoslavsky R, Lombard DB, Liu J, Bruns NE, Tsokos M, Alt FW, Finkel T. A role for the NAD-dependent deacetylase Sirt1 in the regulation of autophagy. Proc Natl Acad Sci U S A 2008;105:3374-3379. ArticlePubMedPMC
  • 11. Morselli E, Maiuri MC, Markaki M, Megalou E, Pasparaki A, Palikaras K, Criollo A, Galluzzi L, Malik SA, Vitale I, Michaud M, Madeo F, Tavernarakis N, Kroemer G. Caloric restriction and resveratrol promote longevity through the Sirtuin-1-dependent induction of autophagy. Cell Death Dis 2010;1:e10ArticlePubMedPMCPDF
  • 12. Melendez A, Talloczy Z, Seaman M, Eskelinen EL, Hall DH, Levine B. Autophagy genes are essential for dauer development and life-span extension in C. elegans. Science 2003;301:1387-1391. ArticlePubMed
  • 13. Tasdemir E, Maiuri MC, Galluzzi L, Vitale I, Djavaheri-Mergny M, D'Amelio M, Criollo A, Morselli E, Zhu C, Harper F, Nannmark U, Samara C, Pinton P, Vicencio JM, Carnuccio R, Moll UM, Madeo F, Paterlini-Brechot P, Rizzuto R, Szabadkai G, Pierron G, Blomgren K, Tavernarakis N, Codogno P, Cecconi F, Kroemer G. Regulation of autophagy by cytoplasmic p53. Nat Cell Biol 2008;10:676-687. ArticlePubMedPMCPDF
  • 14. Scherz-Shouval R, Elazar Z. Regulation of autophagy by ROS: physiology and pathology. Trends Biochem Sci 2011;36:30-38. ArticlePubMed
  • 15. Moore MN. Autophagy as a second level protective process in conferring resistance to environmentally-induced oxidative stress. Autophagy 2008;4:254-256. ArticlePubMed
  • 16. Li L, Chen Y, Gibson SB. Starvation-induced autophagy is regulated by mitochondrial reactive oxygen species leading to AMPK activation. Cell Signal 2013;25:50-65. ArticlePubMed
  • 17. Komatsu M, Waguri S, Koike M, Sou YS, Ueno T, Hara T, Mizushima N, Iwata J, Ezaki J, Murata S, Hamazaki J, Nishito Y, Iemura S, Natsume T, Yanagawa T, Uwayama J, Warabi E, Yoshida H, Ishii T, Kobayashi A, Yamamoto M, Yue Z, Uchiyama Y, Kominami E, Tanaka K. Homeostatic levels of p62 control cytoplasmic inclusion body formation in autophagy-deficient mice. Cell 2007;131:1149-1163. ArticlePubMed
  • 18. Takamura A, Komatsu M, Hara T, Sakamoto A, Kishi C, Waguri S, Eishi Y, Hino O, Tanaka K, Mizushima N. Autophagy-deficient mice develop multiple liver tumors. Genes Dev 2011;25:795-800. ArticlePubMedPMC
  • 19. Scherz-Shouval R, Shvets E, Fass E, Shorer H, Gil L, Elazar Z. Reactive oxygen species are essential for autophagy and specifically regulate the activity of Atg4. EMBO J 2007;26:1749-1760. ArticlePubMedPMC
  • 20. Sohal RS, Mockett RJ, Orr WC. Mechanisms of aging: an appraisal of the oxidative stress hypothesis. Free Radic Biol Med 2002;33:575-586. PubMed
  • 21. Szweda PA, Camouse M, Lundberg KC, Oberley TD, Szweda LI. Aging, lipofuscin formation, and free radical-mediated inhibition of cellular proteolytic systems. Ageing Res Rev 2003;2:383-405. ArticlePubMed
  • 22. Terman A, Brunk UT. Aging as a catabolic malfunction. Int J Biochem Cell Biol 2004;36:2365-2375. ArticlePubMed
  • 23. Rubinsztein DC, Marino G, Kroemer G. Autophagy and aging. Cell 2011;146:682-695. ArticlePubMed
  • 24. Grune T. Oxidative stress, aging and the proteasomal system. Biogerontology 2000;1:31-40. PubMed
  • 25. Levine B, Kroemer G. Autophagy in the pathogenesis of disease. Cell 2008;132:27-42. ArticlePubMedPMC
  • 26. Lipinski MM, Zheng B, Lu T, Yan Z, Py BF, Ng A, Xavier RJ, Li C, Yankner BA, Scherzer CR, Yuan J. Genome-wide analysis reveals mechanisms modulating autophagy in normal brain aging and in Alzheimer's disease. Proc Natl Acad Sci U S A 2010;107:14164-14169. ArticlePubMedPMC
  • 27. Ravikumar B, Berger Z, Vacher C, O'Kane CJ, Rubinsztein DC. Rapamycin pre-treatment protects against apoptosis. Hum Mol Genet 2006;15:1209-1216. ArticlePubMed
  • 28. Ravikumar B, Duden R, Rubinsztein DC. Aggregate-prone proteins with polyglutamine and polyalanine expansions are degraded by autophagy. Hum Mol Genet 2002;11:1107-1117. ArticlePubMed
  • 29. Ravikumar B, Vacher C, Berger Z, Davies JE, Luo S, Oroz LG, Scaravilli F, Easton DF, Duden R, O'Kane CJ, Rubinsztein DC. Inhibition of mTOR induces autophagy and reduces toxicity of polyglutamine expansions in fly and mouse models of Huntington disease. Nat Genet 2004;36:585-595. ArticlePubMedPDF
  • 30. Lee H, Noh JY, Oh Y, Kim Y, Chang JW, Chung CW, Lee ST, Kim M, Ryu H, Jung YK. IRE1 plays an essential role in ER stress-mediated aggregation of mutant huntingtin via the inhibition of autophagy flux. Hum Mol Genet 2012;21:101-114. ArticlePubMed
  • 31. Shibata M, Lu T, Furuya T, Degterev A, Mizushima N, Yoshimori T, MacDonald M, Yankner B, Yuan J. Regulation of intracellular accumulation of mutant Huntingtin by Beclin 1. J Biol Chem 2006;281:14474-14485. ArticlePubMed
  • 32. Nixon RA. Autophagy, amyloidogenesis and Alzheimer disease. J Cell Sci 2007;120(Pt 23):4081-4091. ArticlePubMedPDF
  • 33. Pickford F, Masliah E, Britschgi M, Lucin K, Narasimhan R, Jaeger PA, Small S, Spencer B, Rockenstein E, Levine B, Wyss-Coray T. The autophagy-related protein beclin 1 shows reduced expression in early Alzheimer disease and regulates amyloid beta accumulation in mice. J Clin Invest 2008;118:2190-2199. PubMedPMC
  • 34. Hamer G, Matilainen O, Holmberg CI. A photoconvertible reporter of the ubiquitin-proteasome system in vivo. Nat Methods 2010;7:473-478. ArticlePubMedPDF
  • 35. Liu J, Ma J. Fates-shifted is an F-box protein that targets Bicoid for degradation and regulates developmental fate determination in Drosophila embryos. Nat Cell Biol 2011;13:22-29. ArticlePubMedPDF
  • 36. Min JN, Whaley RA, Sharpless NE, Lockyer P, Portbury AL, Patterson C. CHIP deficiency decreases longevity, with accelerated aging phenotypes accompanied by altered protein quality control. Mol Cell Biol 2008;28:4018-4025. ArticlePubMedPMCPDF
  • 37. Shim SM, Lee WJ, Kim Y, Chang JW, Song S, Jung YK. Role of S5b/PSMD5 in proteasome inhibition caused by TNF-α/NFκB in higher eukaryotes. Cell Rep 2012;2:603-615. ArticlePubMed
  • 38. Vellai T, Takacs-Vellai K, Sass M, Klionsky DJ. The regulation of aging: does autophagy underlie longevity? Trends Cell Biol 2009;19:487-494. ArticlePubMedPMC
  • 39. Bergamini E, Cavallini G, Donati A, Gori Z. The role of macroautophagy in the ageing process, anti-ageing intervention and age-associated diseases. Int J Biochem Cell Biol 2004;36:2392-2404. ArticlePubMed
  • 40. Rubinsztein DC. The roles of intracellular protein-degradation pathways in neurodegeneration. Nature 2006;443:780-786. ArticlePubMedPDF
  • 41. Ichimura Y, Kominami E, Tanaka K, Komatsu M. Selective turnover of p62/A170/SQSTM1 by autophagy. Autophagy 2008;4:1063-1066. ArticlePubMed
  • 42. Kwon J, Han E, Bui CB, Shin W, Lee J, Lee S, Choi YB, Lee AH, Lee KH, Park C, Obin MS, Park SK, Seo YJ, Oh GT, Lee HW, Shin J. Assurance of mitochondrial integrity and mammalian longevity by the p62-Keap1-Nrf2-Nqo1 cascade. EMBO Rep 2012;13:150-156. ArticlePubMedPMCPDF
  • 43. Pyo JO, Yoo SM, Ahn HH, Nah J, Hong SH, Kam TI, Jung S, Jung YK. Overexpression of Atg5 in mice activates autophagy and extends lifespan. Nat Commun 2013;4:2300ArticlePubMedPDF
  • 44. Pandey UB, Nie Z, Batlevi Y, McCray BA, Ritson GP, Nedelsky NB, Schwartz SL, DiProspero NA, Knight MA, Schuldiner O, Padmanabhan R, Hild M, Berry DL, Garza D, Hubbert CC, Yao TP, Baehrecke EH, Taylor JP. HDAC6 rescues neurodegeneration and provides an essential link between autophagy and the UPS. Nature 2007;447:859-863. ArticlePubMedPDF
  • 45. Ding WX, Ni HM, Gao W, Yoshimori T, Stolz DB, Ron D, Yin XM. Linking of autophagy to ubiquitin-proteasome system is important for the regulation of endoplasmic reticulum stress and cell viability. Am J Pathol 2007;171:513-524. ArticlePubMedPMC
  • 46. Ventura N, Rea SL, Testi R. Long-lived C. elegans mitochondrial mutants as a model for human mitochondrial-associated diseases. Exp Gerontol 2006;41:974-991. ArticlePubMed
  • 47. Narendra DP, Jin SM, Tanaka A, Suen DF, Gautier CA, Shen J, Cookson MR, Youle RJ. PINK1 is selectively stabilized on impaired mitochondria to activate Parkin. PLoS Biol 2010;8:e1000298ArticlePubMedPMC
  • 48. Jin SM, Youle RJ. PINK1- and Parkin-mediated mitophagy at a glance. J Cell Sci 2012;125(Pt 4):795-799. ArticlePubMedPMCPDF
  • 49. Vives-Bauza C, Zhou C, Huang Y, Cui M, de Vries RL, Kim J, May J, Tocilescu MA, Liu W, Ko HS, Magrane J, Moore DJ, Dawson VL, Grailhe R, Dawson TM, Li C, Tieu K, Przedborski S. PINK1-dependent recruitment of Parkin to mitochondria in mitophagy. Proc Natl Acad Sci U S A 2010;107:378-383. ArticlePubMed
  • 50. Matsuda N, Sato S, Shiba K, Okatsu K, Saisho K, Gautier CA, Sou YS, Saiki S, Kawajiri S, Sato F, Kimura M, Komatsu M, Hattori N, Tanaka K. PINK1 stabilized by mitochondrial depolarization recruits Parkin to damaged mitochondria and activates latent Parkin for mitophagy. J Cell Biol 2010;189:211-221. ArticlePubMedPMCPDF
  • 51. Solano RM, Casarejos MJ, Menendez-Cuervo J, Rodriguez-Navarro JA, Garcia de, Mena MA. Glial dysfunction in parkin null mice: effects of aging. J Neurosci 2008;28:598-611. ArticlePubMedPMC
  • 52. Kissova I, Deffieu M, Manon S, Camougrand N. Uth1p is involved in the autophagic degradation of mitochondria. J Biol Chem 2004;279:39068-39074. ArticlePubMed
  • 53. Chen YF, Wu CY, Kirby R, Kao CH, Tsai TF. A role for the CISD2 gene in lifespan control and human disease. Ann N Y Acad Sci 2010;1201:58-64. ArticlePubMed
  • 54. Zhang J, Randall MS, Loyd MR, Dorsey FC, Kundu M, Cleveland JL, Ney PA. Mitochondrial clearance is regulated by Atg7-dependent and -independent mechanisms during reticulocyte maturation. Blood 2009;114:157-164. ArticlePubMedPMCPDF
  • 55. Harman D. The biologic clock: the mitochondria? J Am Geriatr Soc 1972;20:145-147. ArticlePubMed
  • 56. Schriner SE, Linford NJ, Martin GM, Treuting P, Ogburn CE, Emond M, Coskun PE, Ladiges W, Wolf N, Van Remmen H, Wallace DC, Rabinovitch PS. Extension of murine life span by overexpression of catalase targeted to mitochondria. Science 2005;308:1909-1911. ArticlePubMed
  • 57. Melov S, Ravenscroft J, Malik S, Gill MS, Walker DW, Clayton PE, Wallace DC, Malfroy B, Doctrow SR, Lithgow GJ. Extension of life-span with superoxide dismutase/catalase mimetics. Science 2000;289:1567-1569. ArticlePubMed
  • 58. Omodei D, Fontana L. Calorie restriction and prevention of age-associated chronic disease. FEBS Lett 2011;585:1537-1542. ArticlePubMedPMCPDF
  • 59. Colman RJ, Anderson RM, Johnson SC, Kastman EK, Kosmatka KJ, Beasley TM, Allison DB, Cruzen C, Simmons HA, Kemnitz JW, Weindruch R. Caloric restriction delays disease onset and mortality in rhesus monkeys. Science 2009;325:201-204. ArticlePubMedPMC
  • 60. Ward WF. Food restriction enhances the proteolytic capacity of the aging rat liver. J Gerontol 1988;43:B121-B124. ArticlePubMed
  • 61. Kennedy BK, Steffen KK, Kaeberlein M. Ruminations on dietary restriction and aging. Cell Mol Life Sci 2007;64:1323-1328. ArticlePubMedPDF
  • 62. Jia K, Thomas C, Akbar M, Sun Q, Adams-Huet B, Gilpin C, Levine B. Autophagy genes protect against Salmonella typhimurium infection and mediate insulin signaling-regulated pathogen resistance. Proc Natl Acad Sci U S A 2009;106:14564-14569. ArticlePubMedPMC
  • 63. Toth ML, Sigmond T, Borsos E, Barna J, Erdelyi P, Takacs-Vellai K, Orosz L, Kovacs AL, Csikos G, Sass M, Vellai T. Longevity pathways converge on autophagy genes to regulate life span in Caenorhabditis elegans. Autophagy 2008;4:330-338. ArticlePubMed
  • 64. Chen D, Steele AD, Lindquist S, Guarente L. Increase in activity during calorie restriction requires Sirt1. Science 2005;310:1641ArticlePubMed
  • 65. Liang J, Shao SH, Xu ZX, Hennessy B, Ding Z, Larrea M, Kondo S, Dumont DJ, Gutterman JU, Walker CL, Slingerland JM, Mills GB. The energy sensing LKB1-AMPK pathway regulates p27(kip1) phosphorylation mediating the decision to enter autophagy or apoptosis. Nat Cell Biol 2007;9:218-224. ArticlePubMedPDF
  • 66. Yorimitsu T, Zaman S, Broach JR, Klionsky DJ. Protein kinase A and Sch9 cooperatively regulate induction of autophagy in Saccharomyces cerevisiae. Mol Biol Cell 2007;18:4180-4189. ArticlePubMedPMC

Figure & Data

References

    Citations

    Citations to this article as recorded by  
    • Dysregulation of autophagy activation induced by atorvastatin contributes to new-onset diabetes mellitus in western diet-fed mice
      Juhee Kim, Minjune Kim, Minjeong Kim, Young-Hye You, Youngmi Song, Byung-Wan Lee
      Metabolism.2024; 153: 155795.     CrossRef
    • The impact of zinc on the molecular signaling pathways in the diabetes disease
      Keyvan Asghari, Zahra Shargh, Sina Fatehfar, Leila Chodari, Parsa Sameei
      Journal of Trace Elements in Medicine and Biology.2022; 72: 126985.     CrossRef
    • Antiaging agents: safe interventions to slow aging and healthy life span extension
      Ji-Kai Liu
      Natural Products and Bioprospecting.2022;[Epub]     CrossRef
    • Glycans in autophagy, endocytosis and lysosomal functions
      Fulvio Reggiori, Hans-Joachim Gabius, Massimo Aureli, Winfried Römer, Sandro Sonnino, Eeva-Liisa Eskelinen
      Glycoconjugate Journal.2021; 38(5): 625.     CrossRef
    • Reduced cardiomyocyte Na+ current in the age‐dependent murine Pgc‐1β−/− model of ventricular arrhythmia
      Shiraz Ahmad, Haseeb Valli, Robert Smyth, Anita Y. Jiang, Kamalan Jeevaratnam, Hugh R. Matthews, Christopher L.‐H. Huang
      Journal of Cellular Physiology.2019; 234(4): 3921.     CrossRef
    • Autophagy in Human Health and Disease: Novel Therapeutic Opportunities
      Francesca Giampieri, Sadia Afrin, Tamara Y. Forbes-Hernandez, Massimiliano Gasparrini, Danila Cianciosi, Patricia Reboredo-Rodriguez, Alfonso Varela-Lopez, Jose L. Quiles, Maurizio Battino
      Antioxidants & Redox Signaling.2019; 30(4): 577.     CrossRef
    • Molybdenum Disulfide Nanoparticles Resist Oxidative Stress-Mediated Impairment of Autophagic Flux and Mitigate Endothelial Cell Senescence and Angiogenic Dysfunctions
      Sunkui Ke, Youlin Lai, Tong Zhou, Lihuang Li, Yange Wang, Lei Ren, Shefang Ye
      ACS Biomaterials Science & Engineering.2018; 4(2): 663.     CrossRef
    • Phenformin inhibits cell proliferation and induces cell apoptosis and autophagy in cholangiocarcinoma
      Shuyang Hu, Qing Ouyang, Qingbao Cheng, Jinghan Wang, Feiling Feng, Liang Qiao, Wei Gan, Yang Shi, Demin Wu, Xiaoqing Jiang
      Molecular Medicine Reports.2018;[Epub]     CrossRef
    • Ventricular pro-arrhythmic phenotype, arrhythmic substrate, ageing and mitochondrial dysfunction in peroxisome proliferator activated receptor-γ coactivator-1β deficient (Pgc-1β) murine hearts
      Shiraz Ahmad, Haseeb Valli, Karan R. Chadda, James Cranley, Kamalan Jeevaratnam, Christopher L.-H. Huang
      Mechanisms of Ageing and Development.2018; 173: 92.     CrossRef
    • From Christian de Duve to Yoshinori Ohsumi: More to autophagy than just dining at home
      Margaret M. Harnett, Miguel A. Pineda, Perle Latré de Laté, Russell J. Eason, Sébastien Besteiro, William Harnett, Gordon Langsley
      Biomedical Journal.2017; 40(1): 9.     CrossRef
    • Metformin Restores Parkin-Mediated Mitophagy, Suppressed by Cytosolic p53
      Young Song, Woo Lee, Yong-ho Lee, Eun Kang, Bong-Soo Cha, Byung-Wan Lee
      International Journal of Molecular Sciences.2016; 17(1): 122.     CrossRef
    • Selected reaction monitoring mass spectrometry for relative quantification of proteins involved in cellular life and death processes
      Rune Isak Dupont Birkler, Zahra Nochi, Niels Gregersen, Johan Palmfeldt
      Journal of Chromatography B.2016; 1035: 49.     CrossRef
    • Dehydroepiandrosterone prevents linoleic acid-induced endothelial cell senescence by increasing autophagy
      Min Jung Lee, Eun Hee Kim, Sang Ah. Lee, Yu Mi Kang, Chang Hee Jung, Hae Kyeong Yoon, So Mi Seol, Yoo La Lee, Woo Je Lee, Joong-Yeol Park
      Metabolism.2015; 64(9): 1134.     CrossRef
    • Successful aging: Advancing the science of physical independence in older adults
      Stephen D. Anton, Adam J. Woods, Tetso Ashizawa, Diana Barb, Thomas W. Buford, Christy S. Carter, David J. Clark, Ronald A. Cohen, Duane B. Corbett, Yenisel Cruz-Almeida, Vonetta Dotson, Natalie Ebner, Philip A. Efron, Roger B. Fillingim, Thomas C. Foster
      Ageing Research Reviews.2015; 24: 304.     CrossRef
    • Autophagy in acute leukemias: A double-edged sword with important therapeutic implications
      Cecilia Evangelisti, Camilla Evangelisti, Francesca Chiarini, Annalisa Lonetti, Francesca Buontempo, Luca M. Neri, James A. McCubrey, Alberto M. Martelli
      Biochimica et Biophysica Acta (BBA) - Molecular Cell Research.2015; 1853(1): 14.     CrossRef
    • Autophagy: A housekeeper in cardiorenal metabolic health and disease
      Guanghong Jia, James R. Sowers
      Biochimica et Biophysica Acta (BBA) - Molecular Basis of Disease.2015; 1852(2): 219.     CrossRef
    • Autophagy and Lipid Droplets in the Liver
      Nuria Martinez-Lopez, Rajat Singh
      Annual Review of Nutrition.2015; 35(1): 215.     CrossRef
    • Lysosomal Two-pore Channel Subtype 2 (TPC2) Regulates Skeletal Muscle Autophagic Signaling
      Pei-Hui Lin, Pu Duann, Shinji Komazaki, Ki Ho Park, Haichang Li, Mingzhai Sun, Mathew Sermersheim, Kristyn Gumpper, John Parrington, Antony Galione, A. Mark Evans, Michael X. Zhu, Jianjie Ma
      Journal of Biological Chemistry.2015; 290(6): 3377.     CrossRef
    • Zinc and autophagy
      Juan P. Liuzzi, Liang Guo, Changwon Yoo, Tiffanie S. Stewart
      BioMetals.2014; 27(6): 1087.     CrossRef
    • Autophagy in the eye: Implications for ocular cell health
      Laura S. Frost, Claire H. Mitchell, Kathleen Boesze-Battaglia
      Experimental Eye Research.2014; 124: 56.     CrossRef
    • Can Enhanced Autophagy Be Associated with Human Longevity? Serum Levels of the Autophagy Biomarker Beclin-1 Are Increased in Healthy Centenarians
      Enzo Emanuele, Piercarlo Minoretti, Fabian Sanchis-Gomar, Helios Pareja-Galeano, Yusuf Yilmaz, Nuria Garatachea, Alejandro Lucia
      Rejuvenation Research.2014; 17(6): 518.     CrossRef
    • Responses and adaptations of intervertebral disc cells to microenvironmental stress: a possible central role of autophagy in the adaptive mechanism
      Libo Jiang, Fenglai Yuan, Xiaofan Yin, Jian Dong
      Connective Tissue Research.2014; 55(5-6): 311.     CrossRef
    • STOP accelerating lung aging for the treatment of COPD
      Kazuhiro Ito, Nicolas Mercado
      Experimental Gerontology.2014; 59: 21.     CrossRef
    • In search of antiaging modalities: Evaluation of mTOR‐ and ROS/DNA damage‐signaling by cytometry
      Zbigniew Darzynkiewicz, Hong Zhao, H. Dorota Halicka, Jiangwei Li, Yong‐Syu Lee, Tze‐Chen Hsieh, Joseph M. Wu
      Cytometry Part A.2014; 85(5): 386.     CrossRef
    • Induction of Covalently Crosslinked p62 Oligomers with Reduced Binding to Polyubiquitinated Proteins by the Autophagy Inhibitor Verteporfin
      Elizabeth Donohue, Aruna D. Balgi, Masaaki Komatsu, Michel Roberge, Srinivasa M. Srinivasula
      PLoS ONE.2014; 9(12): e114964.     CrossRef

    • PubReader PubReader
    • Cite
      CITE
      export Copy
      Close
      Download Citation
      Download a citation file in RIS format that can be imported by all major citation management software, including EndNote, ProCite, RefWorks, and Reference Manager.

      Format:
      • RIS — For EndNote, ProCite, RefWorks, and most other reference management software
      • BibTeX — For JabRef, BibDesk, and other BibTeX-specific software
      Include:
      • Citation for the content below
      The Interplay between Autophagy and Aging
      Diabetes Metab J. 2013;37(5):333-339.   Published online October 17, 2013
      Close
    • XML DownloadXML Download

    Diabetes Metab J : Diabetes & Metabolism Journal