Journal List > Hanyang Med Rev > v.33(2) > 1044179

Choi and Kim: Autophagy in Redox Signalling

Abstract

Autophagy is a major catabolic process that is involved in cellular degradation of unnecessary or dysfunctional cellular components via the lysosomal machinery. Autophagy is involved in a variety of biological processes such as programmed cell death, removal of damaged organelles and development of different tissue-specific functions. In recent experiments, the role of autophagy as an important mediator of the pathological response to redox signalling of cellular damage has been elucidated and expanded. Oxidative stresses to the cellular system induces autophagy as a means to selectively remove oxidatively modified macromolecules and organelles. Reactive oxygen species (ROS) are highly reactive oxygen free radicals that are produced as by-products of cellular metabolism, primarily by mitochondria and NADPH oxidases. ROS can be beneficial or harmful to cells and tissues by depending on their concentration and location. ROS function as redox messengers in intracellular signalling at physiologically low level, whereas excess ROS can induce oxidative modification of cellular macromolecules and eventually promote cell death. Thus, the interface of autophagy-related oxidative stress adaptation and cell death is important for understanding redox biology and pathogenesis. In this review, we describe the basic mechanism and function of autophagy in the context of response to oxidative stress and redox signalling in pathogenesis.

Figures and Tables

Fig. 1
Schematic diagram of the steps of autophagy. Autophagy is initiated by the formation of the isolation membrane. The concerted action of the autophagy core machinery proteins at the phagophore assembly site (PAS) is thought to lead to the expansion of the isolation membrane into an autophagosome (vesicle elongation). Ubiquitinylated proteins may be directly targeted for degradation via the autophagic pathway. The p62 protein interacts with ubiquitinylated-damaged proteins in cells. The complex is then selectively tied to the autophagosome through the interaction between p62 and light chain 3-II (LC3-II). When the outer membrane of the autophagosome fuses with a lysosome, it forms an autophagolysosome. Finally, the sequestered material is degraded inside the autophagolyosome and recycled.
hmr-33-83-g001
Fig. 2
Reactive oxygen species-induced autophagy. ROS production can induce protein aggregates, generated by the mitochondrial electron transport chain activities and by NADPH oxidase. These ROS, which include hydrogen superoxide (O2-), peroxide (H2O2), nitric oxide (NO), peroxynitrite (ONOO-), can damage proteins and celluar organelles to stimulate autophagy. Atg4 Cys81 thiol modification is involved in H2O2-induced autophagy. S-nitrosation of inhibitor of κB kinase β (IKKβ) and c-Jun N-terminal kinase1 (JNK1) are involved in NO-induced inhibition of autophagy. Hypoxia-induced ROS also activates autophagic pathway by hypoxia inducible factors (HIFs) and their adapt proteins. Moreover, autophagy can stimulate the ROS generation to enhance the autophagic signalling.
hmr-33-83-g002
Table 1
Roles of autophagy in disease and pathogenesis
hmr-33-83-i001

References

1. Cuervo AM. Autophagy: in sickness and in health. Trends Cell Biol. 2004; 14:70–77.
crossref
2. Levine B, Klionsky DJ. Development by self-digestion: molecular mechanisms and biological functions of autophagy. Dev Cell. 2004; 6:463–477.
3. Sridhar S, Botbol Y, Macian F, Cuervo AM. Autophagy and disease: always two sides to a problem. J Pathol. 2012; 226:255–273.
crossref
4. Shacka JJ, Roth KA, Zhang J. The autophagy-lysosomal degradation pathway: role in neurodegenerative disease and therapy. Front Biosci. 2008; 13:718–736.
crossref
5. Gottlieb RA, Carreira RS. Autophagy in health and disease. 5. Mitophagy as a way of life. Am J Physiol Cell Physiol. 2010; 299:C203–C210.
crossref
6. He C, Klionsky DJ. Regulation mechanisms and signaling pathways of autophagy. Annu Rev Genet. 2009; 43:67–93.
crossref
7. Chen Y, Klionsky DJ. The regulation of autophagy - unanswered questions. J Cell Sci. 2011; 124:161–170.
crossref
8. Kabeya Y, Mizushima N, Ueno T, Yamamoto A, Kirisako T, Noda T, et al. LC3, a mammalian homologue of yeast Apg8p, is localized in autophagosome membranes after processing. EMBO J. 2000; 19:5720–5728.
crossref
9. Bjorkoy G, Lamark T, Brech A, Outzen H, Perander M, Overvatn A, et al. p62/SQSTM1 forms protein aggregates degraded by autophagy and has a protective effect on huntingtin-induced cell death. J Cell Biol. 2005; 171:603–614.
crossref
10. Gueraud F, Atalay M, Bresgen N, Cipak A, Eckl PM, Huc L, et al. Chemistry and biochemistry of lipid peroxidation products. Free Radic Res. 2010; 44:1098–1124.
crossref
11. Murphy MP. How mitochondria produce reactive oxygen species. Biochem J. 2009; 417:1–13.
crossref
12. 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.
crossref
13. Inoki K, Zhu T, Guan KL. TSC2 mediates cellular energy response to control cell growth and survival. Cell. 2003; 115:577–590.
crossref
14. Kim J, Kundu M, Viollet B, Guan KL. AMPK and mTOR regulate autophagy through direct phosphorylation of Ulk1. Nat Cell Biol. 2011; 13:132–141.
crossref
15. Egan DF, Shackelford DB, Mihaylova MM, Gelino S, Kohnz RA, Mair W, et al. Phosphorylation of ULK1 (hATG1) by AMP-activated protein kinase connects energy sensing to mitophagy. Science. 2011; 331:456–461.
crossref
16. Emerling BM, Weinberg F, Snyder C, Burgess Z, Mutlu GM, Viollet B, et al. Hypoxic activation of AMPK is dependent on mitochondrial ROS but independent of an increase in AMP/ATP ratio. Free Radic Biol Med. 2009; 46:1386–1391.
crossref
17. Alexander A, Cai SL, Kim J, Nanez A, Sahin M, MacLean KH, et al. ATM signals to TSC2 in the cytoplasm to regulate mTORC1 in response to ROS. Proc Natl Acad Sci U S A. 2010; 107:4153–4158.
crossref
18. Sims GP, Rowe DC, Rietdijk ST, Herbst R, Coyle AJ. HMGB1 and RAGE in inflammation and cancer. Annu Rev Immunol. 2010; 28:367–388.
crossref
19. Tang D, Kang R, Cheh CW, Livesey KM, Liang X, Schapiro NE, et al. HMGB1 release and redox regulates autophagy and apoptosis in cancer cells. Oncogene. 2010; 29:5299–5310.
crossref
20. Bellot G, Garcia-Medina R, Gounon P, Chiche J, Roux D, Pouyssegur J, et al. Hypoxia-induced autophagy is mediated through hypoxia-inducible factor induction of BNIP3 and BNIP3L via their BH3 domains. Mol Cell Biol. 2009; 29:2570–2581.
crossref
21. Byun YJ, Kim SK, Kim YM, Chae GT, Jeong SW, Lee SB. Hydrogen peroxide induces autophagic cell death in C6 glioma cells via BNIP3-mediated suppression of the mTOR pathway. Neurosci Lett. 2009; 461:131–135.
crossref
22. Li Y, Wang Y, Kim E, Beemiller P, Wang CY, Swanson J, et al. Bnip3 mediates the hypoxia-induced inhibition on mammalian target of rapamycin by interacting with Rheb. J Biol Chem. 2007; 282:35803–35813.
crossref
23. Hanna RA, Quinsay MN, Orogo AM, Giang K, Rikka S, Gustafsson AB. Microtubule-associated protein 1 light chain 3 (LC3) interacts with Bnip3 protein to selectively remove endoplasmic reticulum and mitochondria via autophagy. J Biol Chem. 2012; 287:19094–19104.
crossref
24. Wasserman WW, Fahl WE. Functional antioxidant responsive elements. Proc Natl Acad Sci U S A. 1997; 94:5361–5366.
25. Ishii T, Itoh K, Yamamoto M. Roles of Nrf2 in activation of antioxidant enzyme genes via antioxidant responsive elements. Methods Enzymol. 2002; 348:182–190.
26. Itoh K, Wakabayashi N, Katoh Y, Ishii T, Igarashi K, Engel JD, et al. Keap1 represses nuclear activation of antioxidant responsive elements by Nrf2 through binding to the amino-terminal Neh2 domain. Genes Dev. 1999; 13:76–86.
crossref
27. Li PF, Dietz R, von Harsdorf R. p53 regulates mitochondrial membrane potential through reactive oxygen species and induces cytochrome c-independent apoptosis blocked by Bcl-2. EMBO J. 1999; 18:6027–6036.
crossref
28. Bensaad K, Tsuruta A, Selak MA, Vidal MN, Nakano K, Bartrons R, et al. TIGAR, a p53-inducible regulator of glycolysis and apoptosis. Cell. 2006; 126:107–120.
crossref
29. Bensaad K, Cheung EC, Vousden KH. Modulation of intracellular ROS levels by TIGAR controls autophagy. EMBO J. 2009; 28:3015–3026.
crossref
30. Crighton D, Wilkinson S, O'Prey J, Syed N, Smith P, Harrison PR, et al. DRAM, a p53-induced modulator of autophagy, is critical for apoptosis. Cell. 2006; 126:121–134.
crossref
31. Budanov AV. Stress-responsive sestrins link p53 with redox regulation and mammalian target of rapamycin signaling. Antioxid Redox Signal. 2011; 15:1679–1690.
crossref
32. McCray BA, Taylor JP. The role of autophagy in age-related neurodegeneration. Neurosignals. 2008; 16:75–84.
crossref
33. Gal J, Strom AL, Kwinter DM, Kilty R, Zhang J, Shi P, et al. Sequestosome 1/p62 links familial ALS mutant SOD1 to LC3 via an ubiquitin-independent mechanism. J Neurochem. 2009; 111:1062–1073.
crossref
34. Hara T, Nakamura K, Matsui M, Yamamoto A, Nakahara Y, Suzuki-Migishima R, et al. Suppression of basal autophagy in neural cells causes neurodegenerative disease in mice. Nature. 2006; 441:885–889.
crossref
35. Lee JH, Yu WH, Kumar A, Lee S, Mohan PS, Peterhoff CM, et al. Lysosomal proteolysis and autophagy require presenilin 1 and are disrupted by Alzheimer-related PS1 mutations. Cell. 2010; 141:1146–1158.
crossref
36. Jin S. Autophagy, mitochondrial quality control, and oncogenesis. Autophagy. 2006; 2:80–84.
crossref
37. Jung HS, Chung KW, Won Kim J, Kim J, Komatsu M, Tanaka K, et al. Loss of autophagy diminishes pancreatic beta cell mass and function with resultant hyperglycemia. Cell Metab. 2008; 8:318–324.
crossref
38. Petersen KF, Dufour S, Befroy D, Garcia R, Shulman GI. Impaired mitochondrial activity in the insulin-resistant offspring of patients with type 2 diabetes. N Engl J Med. 2004; 350:664–671.
crossref
39. Jung HS, Lee MS. Role of autophagy in diabetes and mitochondria. Ann N Y Acad Sci. 2010; 1201:79–83.
crossref
40. Nishino I, Fu J, Tanji K, Yamada T, Shimojo S, Koori T, et al. Primary LAMP-2 deficiency causes X-linked vacuolar cardiomyopathy and myopathy (Danon disease). Nature. 2000; 406:906–910.
crossref
41. Zhu H, Tannous P, Johnstone JL, Kong Y, Shelton JM, Richardson JA, et al. Cardiac autophagy is a maladaptive response to hemodynamic stress. J Clin Invest. 2007; 117:1782–1793.
crossref
42. Nakai A, Yamaguchi O, Takeda T, Higuchi Y, Hikoso S, Taniike M, et al. The role of autophagy in cardiomyocytes in the basal state and in response to hemodynamic stress. Nat Med. 2007; 13:619–624.
crossref
43. Huang C, Andres AM, Ratliff EP, Hernandez G, Lee P, Gottlieb RA. Preconditioning involves selective mitophagy mediated by Parkin and p62/SQSTM1. PLoS One. 2011; 6:e20975.
crossref
44. Deretic V. Multiple regulatory and effector roles of autophagy in immunity. Curr Opin Immunol. 2009; 21:53–62.
crossref
45. Orvedahl A, Levine B. Eating the enemy within: autophagy in infectious diseases. Cell Death Differ. 2009; 16:57–69.
crossref
46. Lee HK, Lund JM, Ramanathan B, Mizushima N, Iwasaki A. Autophagy-dependent viral recognition by plasmacytoid dendritic cells. Science. 2007; 315:1398–1401.
crossref
47. Jounai N, Takeshita F, Kobiyama K, Sawano A, Miyawaki A, Xin KQ, et al. The Atg5 Atg12 conjugate associates with innate antiviral immune responses. Proc Natl Acad Sci U S A. 2007; 104:14050–14055.
crossref
48. Dengjel J, Schoor O, Fischer R, Reich M, Kraus M, Muller M, et al. Autophagy promotes MHC class II presentation of peptides from intracellular source proteins. Proc Natl Acad Sci U S A. 2005; 102:7922–7927.
crossref
49. Miller BC, Zhao Z, Stephenson LM, Cadwell K, Pua HH, Lee HK, et al. The autophagy gene ATG5 plays an essential role in B lymphocyte development. Autophagy. 2008; 4:309–314.
crossref
50. Nedjic J, Aichinger M, Emmerich J, Mizushima N, Klein L. Autophagy in thymic epithelium shapes the T-cell repertoire and is essential for tolerance. Nature. 2008; 455:396–400.
crossref
TOOLS
Similar articles