Skip to main content

Advertisement

Log in

Mitochondrial quality control in AMD: does mitophagy play a pivotal role?

  • Review
  • Published:
Cellular and Molecular Life Sciences Aims and scope Submit manuscript

Abstract

Age-related macular degeneration (AMD) is the predominant cause of visual loss in old people in the developed world, whose incidence is increasing. This disease is caused by the decrease in macular function, due to the degeneration of retinal pigment epithelium (RPE) cells. The aged retina is characterised by increased levels of reactive oxygen species (ROS), impaired autophagy, and DNA damage that are linked to AMD pathogenesis. Mitophagy, a mitochondria-specific type of autophagy, is an essential part of mitochondrial quality control, the collective mechanism responsible for this organelle’s homeostasis. The abundance of ROS, DNA damage, and the excessive energy consumption in the ageing retina all contribute to the degeneration of RPE cells and their mitochondria. We discuss the role of mitophagy in the cell and argue that its impairment may play a role in AMD pathogenesis. Thus, mitophagy as a potential therapeutic target in AMD and other degenerative diseases is as well explored.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4

Similar content being viewed by others

Abbreviations

AMBRA1:

Activating molecule in beclin-1-regulated autophagy

AMD:

Age-related macular degeneration

AMPK:

AMP-activated protein kinase

ARE:

Antioxidant-binding element

Atg:

Autophagy-related gene

DMT1:

Divalent metal transporter 1

ER:

Endoplasmic reticulum

FPN1:

Ferroportin 1

FPF:

Flavoprotein fluorescence

FtMt:

Mitochondrial ferritin

Gp78:

Glycoprotein 78

HDAC6:

Histone deacetylase 6

HIF-1α:

Hypoxia-inducible factor-1α

Hsp:

Heat-shock protein

IMM:

Inner mitochondrial membrane

Keap1:

Kelch-like ECH-associated protein 1

LIR:

MAPK1LC3-interacting region

MAM:

Mitochondria-associated endoplasmic reticulum membrane

MAPK:

Mitogen-activated protein kinase

MAPK1LC3:

Microtubule-associated protein 1 light-chain 3

Mfn:

Mitofusin

MMP:

Mitochondrial membrane potential

MPT:

Mitochondrial permeability transition

MRC:

Mitochondrial respiratory chain

mtDNA:

Mitochondrial DNA

mTORC1:

Mammalian target of rapamycin complex 1

Mul1:

Mitochondrial E3 ubiquitin ligase

Nrf2:

Nuclear factor (erythroid-derived 2)-related factor 2

OMM:

Outer mitochondrial membrane

PGC-1α:

Peroxisome proliferator-activated receptor gamma coactivator-1α

PI3K:

Phosphatidylinosotole-3-kinase

PINK1:

Phosphatase and tensin homologue-induced putative kinase

POS:

Photoreceptor outer segment

ROS:

Reactive oxygen species

RGC:

Retinal ganglion cell

RPE:

Retinal pigment epithelium

SAMD:

Senescence-associated mitochondrial dysfunction

SASP:

Senescent-associated secretory phenotype

Sirt1:

NAD-dependent deacetylase Sirtuin 1

Smurf1:

SMAD-specific E3 ubiquitin protein ligase 1

SOD:

Superoxide dismutase

SQSTM1/p62:

Sequestosome1/p62

Tfam:

Mitochondrial transcription factor A

TIM:

Translocase of the inner mitochondrial membrane

TOM:

Translocase of the outer mitochondrial membrane

Ub:

Ubiquitin

VDAC:

Voltage-dependent anion channel

VEGF:

Vascular endothelial growth factor

References

  1. Wong WL, Su X, Li X, Cheung CMG, Klein R, Cheng C-Y, Wong TY (2014) Global prevalence of age-related macular degeneration and disease burden projection for 2020 and 2040: a systematic review and meta-analysis. Lancet Glob Health 2(2):e106–e116. https://doi.org/10.1016/S2214-109X(13)70145-1

    Article  PubMed  Google Scholar 

  2. Black JRM, Clark SJ (2015) Age-related macular degeneration: genome-wide association studies to translation. Genet Med 18(4):283–289. https://doi.org/10.1038/gim.2015.70

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  3. Kaarniranta K, Sinha D, Blasiak J, Kauppinen A, Veréb Z, Salminen A, Boulton ME, Petrovski G (2013) Autophagy and heterophagy dysregulation leads to retinal pigment epithelium dysfunction and development of age-related macular degeneration. Autophagy 9(7):973–984. https://doi.org/10.4161/auto.24546

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  4. Bhutto I, Lutty G (2012) Understanding age-related macular degeneration (AMD): relationships between the photoreceptor/retinal pigment epithelium/Bruch’s membrane/choriocapillaris complex. Mol Aspects Med 33(4):295–317. https://doi.org/10.1016/j.mam.2012.04.005

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  5. Kinnunen K, Petrovski G, Moe MC, Berta A, Kaarniranta K (2012) Molecular mechanisms of retinal pigment epithelium damage and development of age-related macular degeneration. Acta Ophthalmol 90(4):299–309. https://doi.org/10.1111/j.1755-3768.2011.02179.x

    Article  PubMed  CAS  Google Scholar 

  6. Klettner A, Roider J (2009) Treating age-related macular degeneration—interaction of VEGF-antagonists with their target. Mini Rev Med Chem 9(9):1127–1135. https://doi.org/10.2174/138955709788922665

    Article  PubMed  CAS  Google Scholar 

  7. Blasiak J, Szaflik JP (2011) DNA damage and repair in age-related macular degeneration. Front Biosci (Landmark Ed) 16(4):1291–1301. https://doi.org/10.2741/3789

    Article  CAS  Google Scholar 

  8. Handa JT (2012) How does the macula protect itself from oxidative stress? Mol Aspects Med 33(4):418–435. https://doi.org/10.1016/j.mam.2012.03.006

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  9. Klettner A (2012) Oxidative stress induced cellular signaling in RPE cells. Front Biosci (Schol Ed) 4(2):392–411. https://doi.org/10.2741/S275

    Article  Google Scholar 

  10. Golestaneh N, Chu Y, Cheng SK, Cao H, Poliakov E, Berinstein DM (2016) Repressed SIRT1/PGC-1α pathway and mitochondrial disintegration in iPSC-derived RPE disease model of age-related macular degeneration. J Transl Med 14(1):344. https://doi.org/10.1186/s12967-016-1101-8

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  11. Golestaneh N, Chu Y, Xiao YY, Stoleru GL, Theos AC (2017) Dysfunctional autophagy in RPE, a contributing factor in age-related macular degeneration. Cell Death Dis 8(1):e2537. https://doi.org/10.1038/cddis.2016.453

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  12. McCord JM, Edeas MA (2005) SOD, oxidative stress and human pathologies: a brief history and a future vision. Biomed Pharmacother 59(4):139–142. https://doi.org/10.1016/j.biopha.2005.03.005

    Article  PubMed  CAS  Google Scholar 

  13. Jarrett SG, Boulton ME (2012) Consequences of oxidative stress in age-related macular degeneration. Mol Aspects Med 33(4):399–417. https://doi.org/10.1016/j.mam.2012.03.009

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  14. Kaarniranta K, Salminen A, Eskelinen EL, Kopitz J (2009) Heat shock proteins as gatekeepers of proteolytic pathways-Implications for age-related macular degeneration (AMD). Ageing Res Rev 8(2):128–139. https://doi.org/10.1016/j.arr.2009.01.001

    Article  PubMed  CAS  Google Scholar 

  15. Mettu PS, Wielgus AR, Ong SS, Cousins SW (2012) Retinal pigment epithelium response to oxidant injury in the pathogenesis of early age-related macular degeneration. Mol Aspects Med 33(4):376–398. https://doi.org/10.1016/j.mam.2012.04.006

    Article  PubMed  CAS  Google Scholar 

  16. Tuo J, Grob S, Zhang K, Chan CC (2012) Genetics of immunological and inflammatory components in age-related macular degeneration. Ocul Immunol Inflamm 20(1):27–36. https://doi.org/10.3109/09273948.2011.628432

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  17. Hyttinen JMT, Błasiak J, Niittykoski M, Kinnunen K, Kauppinen A, Salminen A, Kaarniranta K (2017) DNA damage response and autophagy in the degeneration of retinal pigment epithelial cells—implications for age-related macular degeneration (AMD). Ageing Res Rev 36:64–77. https://doi.org/10.1016/j.arr.2017.03.006

    Article  PubMed  CAS  Google Scholar 

  18. Karunadharma PP, Nordgaard CL, Olsen TW, Ferrington DA (2010) Mitochondrial DNA damage as a potential mechanism for age-related macular degeneration. Investig Ophthalmol Vis Sci 51(11):5470–5479. https://doi.org/10.1167/iovs.10-5429

    Article  Google Scholar 

  19. Terluk MR, Kapphahn RJ, Soukup LM, Gong H, Gallardo C, Montezuma SR, Ferrington DA (2015) Investigating mitochondria as a target for treating age-related macular degeneration. J Neurosci 35(18):7304–7311. https://doi.org/10.1523/jneurosci.0190-15.2015

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  20. Barot M, Gokulgandhi MR, Mitra AK (2011) Mitochondrial dysfunction in retinal diseases. Curr Eye Res 36(12):1069–1077. https://doi.org/10.3109/02713683.2011.607536

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  21. Feher J, Kovacs I, Artico M, Cavallotti C, Papale A, Balacco Gabrieli C (2006) Mitochondrial alterations of retinal pigment epithelium in age-related maculrt degeneration. Neurobiol Aging 27(7):596–607. https://doi.org/10.1016/j.neurobiolaging.2005.05.012

    Article  CAS  Google Scholar 

  22. Jarrett SG, Lin H, Godley BF, Boulton ME (2008) Mitochondrial DNA damage and its potential in the retina degeneration. Prog Retin Eye Res 27(6):596–607. https://doi.org/10.1016/j.preteyeres.2008.09.001

    Article  PubMed  CAS  Google Scholar 

  23. Shang F, Taylor A (2012) Roles for the ubiquitin-proteasome pathway in protein quality control and signaling in the retina: implications in the pathogenesis of age-related macular degeneration. Mol Aspects Med 33(4):446–466. https://doi.org/10.1016/j.mam.2012.04.001

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  24. Reggiori F (2012) Autophagy: new questions from recent answers. ISRN Mol Biol 2012:738718. https://doi.org/10.5402/2012/738718

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  25. Yang Z, Klionsky DJ (2010) Eaten alive: a history of macroautophagy. Nat Cell Biol 12(9):814–822. https://doi.org/10.1038/ncb0910-814

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  26. Hyttinen JMT, Niittykoski M, Salminen A, Kaarniranta K (2013) Maturation of autophagosomes and endosomes: a key role for Rab7. Biochim Biophys Acta 1833(3):503–510. https://doi.org/10.1016/j.bbamcr.2012.11.018

    Article  PubMed  CAS  Google Scholar 

  27. Harnett MM, Pineda MA, Latré de Laté P, Eason RJ, Besteiro S, Harnett W, Langsley G (2017) From Christian de Duve to Yoshinori Ohsumi: more to autophagy than just dining at home. Biomed J 40(1):9–22. https://doi.org/10.1016/j.bj.2016.12.004

    Article  PubMed  Google Scholar 

  28. Yang Z, Klionsky DJ (2010) Mammalian autophagy: core molecular machinery and signalling regulation. Curr Opin Cell Biol 22(2):124–131. https://doi.org/10.1016/j.ceb.2009.11.014

    Article  PubMed  CAS  Google Scholar 

  29. Arjamaa O, Nikinmaa M, Salminen A, Kaarniranta K (2009) Regulatory role of HIF-1alpha in the pathogenesis of age-related macular degeneration (AMD). Ageing Res Rev 8(4):349–358. https://doi.org/10.1016/j.arr.2009.06.002

    Article  PubMed  CAS  Google Scholar 

  30. Salminen A, Kaarniranta K (2009) Regulation of the aging process by autophagy. Trends Mol Med 15(5):217–224. https://doi.org/10.1016/j.molmed.2009.03.004

    Article  PubMed  CAS  Google Scholar 

  31. Krohne TU, Stratmann NK, Kopitz J, Holz FG (2010) Effects of lipid peroxidation products on lipofuscinogenesis and autophagy in human retinal pigment epithelial cells. Exp Eye Res 90(3):465–471. https://doi.org/10.1016/j.exer.2009.12.011

    Article  PubMed  CAS  Google Scholar 

  32. Wang AL, Boulton ME, Dunn WA Jr, Rao HV, Cai J, Lukas TJ, Neufeld AH (2009) Using LC3 to monitor autophagy flux in the retinal pigment epithelium. Autophagy 5(8):1190–1193. https://doi.org/10.4161/auto.5.8.10087

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  33. Kim JY, Zhao H, Martinez J, Doggett TA, Kolesnikov AV, Tang PH, Ablonczy Z, Chan CC, Zhou Z, Green DR, Ferguson TA (2013) Noncanonical autophagy promotes the visual cycle. Cell 154(2):365–376. https://doi.org/10.1016/j.cell.2013.06.012

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  34. Mizushima N, Komatsu M (2011) Autophagy: renovation of cells and tissues. Cell 147(4):728–741. https://doi.org/10.1016/j.cell.2011.10.026

    Article  PubMed  CAS  Google Scholar 

  35. Kaarniranta K (2010) Autophagy-hot topic in AMD. Acta Ophthalmol 88(4):387–388. https://doi.org/10.1111/j.1755-3768.2009.01840.x

    Article  PubMed  Google Scholar 

  36. Mitter SK, Rao HV, Qi X, Cai J, Sugrue A, Dunn WA Jr, Grant MB, Boulton ME (2012) Autophagy in the retina: a potential role in age-related macular degeneration. Adv Exp Med Biol 723:83–90. https://doi.org/10.1007/978-1-4614-0631-0_12

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  37. Ryhänen T, Hyttinen JMT, Kopitz J, Rilla K, Kuusisto E, Mannermaa E, Viiri J, Holmberg CI, Immonen I, Meri S, Parkkinen J, Eskelinen EL, Uusitalo H, Salminen A, Kaarniranta K (2009) Crosstalk between Hsp70 molecular chaperone, lysosomes and proteasomes in autophagy-mediated proteolysis in human retinal pigment epithelial cells. J Cell Mol Med 13(9B):3616–3631. https://doi.org/10.1111/j.1582-4934.2008.00577.x

    Article  PubMed  Google Scholar 

  38. Viiri J, Amadio M, Marchesi N, Hyttinen JMT, Kivinen N, Sironen R, Rilla K, Akhtar S, Provenzani A, D’Agostino VG, Govoni S, Pascale A, Agostini H, Petrovski G, Salminen A, Kaarniranta K (2013) Autophagy activation clears ELAV1/HuR-mediated accumulation of SQSTM1/p62 during proteasomal inhibition in human retinal pigment epithelial cells. PLoS One 8(7):e69563. https://doi.org/10.1371/journal.pone.0069563

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  39. Wang AL, Lukas TJ, Yuan M, Du N, Tso MO, Neufeld AH (2009) Autophagy and exosomes in the aged retinal pigment epithelium: possible relevance to drusen formation and age-related macular degeneration. PLoS One 4(1):e4160. https://doi.org/10.1371/journal.pone.0004160

    Article  PubMed  PubMed Central  Google Scholar 

  40. Kauppinen A, Niskanen H, Suuronen T, Kinnunen K, Salminen A, Kaarniranta K (2012) Oxidative stress activates NLRP3 inflammasomes in ARPE-19 cells-Implications for age-related macular degeneration (AMD). Immunol Lett 147(1–2):29–33. https://doi.org/10.1016/j.imlet.2012.05.005

    Article  PubMed  CAS  Google Scholar 

  41. Salminen A, Kaarniranta K, Kauppinen A (2012) Inflammaging: disturbed interplay between autophagy and inflammasomes. Aging 4(3):166–175. https://doi.org/10.18632/aging.100444

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  42. Boya P, Esteban-Martínez L, Serrano-Puebla A, Gómez-Sintes R, Vilarejo-Zori B (2016) Autophagy in the eye: development, degeneration, and aging. Prog Retin Eye Res 55:206–245. https://doi.org/10.1016/j.preteyeres.2016.08.001

    Article  PubMed  Google Scholar 

  43. Hyttinen JMT, Amadio M, Viiri J, Pascale A, Salminen A, Kaarniranta K (2014) Clearance of misfolded and aggregated proteins by aggrephagy and implication for aggregation diseases. Ageing Res Rev 18:16–28. https://doi.org/10.1016/j.arr.2014.07.002

    Article  PubMed  CAS  Google Scholar 

  44. Novak I (2012) Mitophagy: a complex mechanism of mitochondrial removal. Antiox Redox Signal 17(5):794–802. https://doi.org/10.1089/ars.2011.4407

    Article  CAS  Google Scholar 

  45. He C, Klionsky DJ (2009) Regulation mechanisms and signaling pathways of autophagy. Annu Rev Genet 43:67–93. https://doi.org/10.1146/annurev-genet-102808-114910

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  46. Kroemer G, Mariño G, Levine B (2010) Autophagy and the integrated stress response. Mol Cell 40(2):280–293. https://doi.org/10.1016/j.molcel.2010.09.023

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  47. Scherz-Shouval R, Shvets E, Fass E, Shorer H, Gil L, Elazar Z (2007) Reactive oxygen species are essential for autophagy and specifically regulate the activity of Atg4. EMBO J 26:1749–1760. https://doi.org/10.1038/sj.emboj.7601623

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  48. Hailey DW, Rambold AS, Satpute-Krishnan P, Mitra K, Sougrat R, Kim PK, Lippincott-Schwartz J (2010) Mitochondria supply membranes for autophagosome biogenesis during starvation. Cell 141(4):656–667. https://doi.org/10.1016/j.cell.2010.04.009

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  49. Okamoto K, Kondo-Okamoto N (2012) Mitochondria and autophagy: critical interplay between the two homeostats. Biochim Biophys Acta 1820(5):595–600. https://doi.org/10.1016/j.bbagen.2011.08.001

    Article  PubMed  CAS  Google Scholar 

  50. Funderburk SF, Wang QJ, Yue Z (2010) The beclin 1–VPS34 complex—at the crossroads of autophagy and beyond. Trends Cell Biol 20(6):355–362. https://doi.org/10.1016/j.tcb.2010.03.002

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  51. Cecconi F, Levine B (2008) The role of autophagy in mammalian development: cell makeover rather than cell death. Dev Cell 15(3):344–357. https://doi.org/10.1016/j.devcel.2008.08.012

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  52. Strappazzon F, Nazio F, Corrado M, Cianfanelli V, Romagnoli A, Fimia GM, Campello S, Nardacci R, Piacentini M, Campanella M, Cecconi F (2015) AMBRA1 is able to induce mitophagy via LC3 binding, regardless of PARKIN and p62/SQSTM1. Cell Death Differ 22(3):419–432. https://doi.org/10.1038/cdd.2014.190

    Article  PubMed  CAS  Google Scholar 

  53. Strappazzon F, Vietri-Rudan M, Campello S, Nazio F, Florenzano F, Fimia GM, Piacentini M, Levine B, Cecconi F (2011) Mitochondrial BCL-2 inhibits AMBRA1–induced autophagy. EMBO J 30(7):1195–1208. https://doi.org/10.1038/emboj.2011.49

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  54. Kissová L, Deffieu M, Manon S, Camougrand N (2004) Uth1p is involved in the autophagic degradation of mitochondria. J Biol Chem 279(37):39068–39074. https://doi.org/10.1074/jbc.M406960200

    Article  PubMed  CAS  Google Scholar 

  55. Lemasters JJ (2005) Selective mitochondrial autophagy, or mitophagy, as a targeted defense against oxidative stress, mitochondrial dysfunction, and aging. Rejuvenation Res 8(1):3–5. https://doi.org/10.1089/rej.2005.8.3

    Article  PubMed  CAS  Google Scholar 

  56. Lemasters JJ (2014) Variants of mitochondrial autophagy: types 1 and 2 mitophagy and micromitophagy (type 3). Redox Biol 2:749–754. https://doi.org/10.1016/j.redox.2014.06.004

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  57. Michel S, Wanet A, De Pauw A, Rommelaere G, Arnould T, Renard P (2012) Crosstalk between mitochondrial (dys)function and mitochondrial abundance. J Cell Physiol 227(6):2297–2310. https://doi.org/10.1002/jcp.23021

    Article  PubMed  CAS  Google Scholar 

  58. Picca A, Lezza AMS, Leeuwenburgh C, Pesce V, Calvani R, Landi F, Bernabei R, Marzetti E (2017) Fueling inflamm-aging through mitochondrial dysfunction: mechanisms and molecular targets. Int J Mol Sci 18(5):933. https://doi.org/10.3390/ijms18050933

    Article  PubMed Central  Google Scholar 

  59. Elmore SP, Qian T, Grissom SF, Lemasters JJ (2001) The mitochondrial permeability transition initiates autophagy in rat hepatocytes. FASEB J 15(12):2286–2287. https://doi.org/10.1096/fj.01-0206fje

    Article  PubMed  CAS  Google Scholar 

  60. Kim I, Lemasters JJ (2011) Mitophagy selectively degrades individual damaged mitochondria after photoirradiation. Antiox Redox Signal 14(10):1919–1928. https://doi.org/10.1089/ars.2010.3768

    Article  CAS  Google Scholar 

  61. Esteban-Martínez L, Sierra-Filardi E, McGreal RS, Salazar-Roa M, Mariño G, Seco E, Durand S, Enot D, Graña O, Malumbres M, Cvekl A, Cuervo AM, Kroemer G, Boya P (2017) Programmed mitophagy is essential for the glycolytic switch during cell differentiation. EMBO J 36(12):1688–1706. https://doi.org/10.15252/embj.201695916

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  62. Dengjel J, Abeliovich H (2017) Roles of mitophagy in cellular physiology and development. Cell Tissue Res 367(1):95–109. https://doi.org/10.1007/s00441-016-2472-0

    Article  PubMed  CAS  Google Scholar 

  63. Szymański J, Janikiewicz J, Michalska B, Patalas-Krawczyk P, Perrone M, Ziółkowski W, Duszyński J, Pinton P, Dobrzyń A, Więckowski MR (2017) Interaction of mitochondria with the endoplasmic reticulum and plasma membrane in calcium homeostasis, lipid trafficking and mitochondrial structure. Int J Mol Sci 18(7):1576. https://doi.org/10.3390/ijms18071576

    Article  PubMed Central  Google Scholar 

  64. Hamasaki M, Furuta N, Matsuda A, Nezu A, Yamamoto A, Fujita N, Oomori H, Noda T, Haraguchi T, Hiraoka Y, Amano A, Yoshimori T (2013) Autophagosomes form at ER-mitochondria contact sites. Nature 495(7441):389–393. https://doi.org/10.1038/nature11910

    Article  PubMed  CAS  Google Scholar 

  65. Carreras-Sureda A, Pihán P, Hetz C (2017) The unfolded protein response: at the intersection between endoplasmic reticulum function and mitochondrial bioenergetics. Front Oncol 7:55. https://doi.org/10.3389/fonc.2017.00055

    Article  PubMed  PubMed Central  Google Scholar 

  66. Farré JC, Subramani S (2016) Mechanistic insights into selective autophagy pathways: lessons from yeast. Nat Rev Mol Cell Biol 17(9):537–552. https://doi.org/10.1038/nrm.2016.74

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  67. Mao K, Wang K, Zhao M, Xu T, Klionsky DJ (2011) Two MAPK-signaling pathways are required for mitophagy in Saccharomyces cerevisiae. J Cell Biol 193(4):755–767. https://doi.org/10.1083/jcb.201102092

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  68. Palikaras K, Daskalaki I, Markaki M, Tavernarakis N (2017) Mitophagy and age-related pathologies: development of new therapeutics by targeting mitochondrial turnover. Pharmacol Ther 178:157–174. https://doi.org/10.1016/j.pharmthera.2017.04.005

    Article  PubMed  CAS  Google Scholar 

  69. Geisler S, Holmstrom KM, Skujat D, Fiesel FC, Rothfuss OC, Kahle PJ, Springer W (2010) PINK1/Parkin–mediated mitophagy is dependent on VDAC1 and p62/SQSTM1. Nat Cell Biol 12(2):119–131. https://doi.org/10.1038/ncb2012

    Article  PubMed  CAS  Google Scholar 

  70. 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 (2010) PINK1 stabilized by mitochondrial depolarization recruits Parkin to damaged mitochondria and activates latent Parkin for mitophagy. J Cell Biol 189(2):211–221. https://doi.org/10.1083/jcb.200910140

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  71. Narendra DP, Jin SM, Tanaka A, Suen DF, Gautier CA, Shen J, Cookson MR, Youle RJ (2010) PINK1 is selectively stabilized on impaired mitochondria to activate Parkin. PLoS Biol 8(1):1000298. https://doi.org/10.1371/journal.pbio.1000298

    Article  CAS  Google Scholar 

  72. Pickrell AM, Youle RJ (2015) The roles of PINK1, parkin, and mitochondrial fidelity in Parkinson’s disease. Neuron 85(2):257–273. https://doi.org/10.1016/j.neuron.2014.12.007

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  73. Vives-Bauza C, Zhou C, Huang Y, Cui M, de Vries RL, Kim J, May J, Tocilescu MA, Liu W, Ko HS, Magrané J, Moore DJ, Dawson VL, Grailhe R, Dawson TM, Li C, Tieu K, Przedborski S (2010) PINK1-dependent recruitment of Parkin to mitochondria in mitophagy. Proc Natl Acad Sci USA 107(1):378–383. https://doi.org/10.1073/pnas.0911187107

    Article  PubMed  Google Scholar 

  74. Moore DJ (2006) Parkin: a multifaced ubiquitin ligase. Biochem Soc Trans 34(Pt 5):749–753. https://doi.org/10.1042/BST0340749

    Article  PubMed  CAS  Google Scholar 

  75. Zhou C, Huang Y, Shao Y, May J, Prou D, Perier C, Dauer W, Schon EA, Przedborski S (2008) The kinase domain of mitochondrial PINK1 faces the cytoplasm. Proc Natl Acad Sci USA 105(33):12022–12027. https://doi.org/10.1073/pnas.0802814105

    Article  PubMed  Google Scholar 

  76. Geisler S, Holmström KM, Treis A, Skujat D, Weber SS, Fiesel FC, Kahle PJ, Springer W (2010) The PINK1/Parkin–mediated mitophagy is compromised by PD-associated mutations. Autophagy 6(7):871–878. https://doi.org/10.4161/auto.6.7.13286

    Article  PubMed  CAS  Google Scholar 

  77. Kim Y, Park J, Kim S, Song S, Kwon SK, Lee SH, Kitada T, Kim JM, Chung J (2008) PINK1 controls mitochondrial localization of Parkin through direct phosphorylation. Biochem Biophys Res Commun 377(3):975–980. https://doi.org/10.1016/j.bbrc.2008.10.104

    Article  PubMed  CAS  Google Scholar 

  78. Yamano K, Youle RJ (2013) PINK1 is degraded through the N-end rule pathway. Autophagy 9(11):1758–1769. https://doi.org/10.4161/auto.24633

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  79. Lazarou M (2015) Keeping the immune system in check: a role for mitophagy. Immunol Cell Biol 93(1):3–10. https://doi.org/10.1038/icb.2014.75

    Article  PubMed  CAS  Google Scholar 

  80. Lazarou M, Jin SM, Kane LA, Youle RJ (2012) Role of PINK1 binding to the TOM complex and alternate intracellular membranes in recruitment and activation of the E3 ligase Parkin. Dev Cell 22(2):320–333. https://doi.org/10.1016/j.devcel.2011.12.014

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  81. Zheng Q, Huang C, Guo J, Tan J, Wang C, Tang B, Zhang H (2017) Hsp70 participates in PINK1-mediated mitophagy by regulating the stability of PINK1. Neurosci Lett 662:264–270. https://doi.org/10.1016/j.neulet.2017.10.051

    Article  PubMed  CAS  Google Scholar 

  82. Subrizi A, Toropainen E, Ramsay E, Airaksinen AJ, Kaarniranta K, Urtti A (2015) Oxidative stress protection by exogenous delivery of rhHsp70 chaperone to the retinal pigment epithelium (RPE), a possible therapeutic strategy against RPE degeneration. Pharm Res 32(1):211–221. https://doi.org/10.1007/s11095-014-1456-6

    Article  PubMed  CAS  Google Scholar 

  83. Rüb C, Wilkening A, Voos W (2017) Mitochondnrial quality control by the Pink1/Parkin system. Cell Tissue Res 367(1):111–123. https://doi.org/10.1007/s00441-016-2485-8

    Article  PubMed  CAS  Google Scholar 

  84. Kane LA, Lazarou M, Fogel AI, Li Y, Yamano K, Sarraf SA, Banerjee S, Youle RJ (2014) PINK1 phosphorylates ubiquitin to activate Parkin E3 ubiquitin ligase activity. J Cell Biol 205(2):143–150. https://doi.org/10.1083/jcb.201402104

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  85. Kazlauskaite A, Martínez-Torres RJ, Wilkie S, Kumar A, Peltier J, Gonzalez A, Johnson C, Zhang J, Hope AG, Peggie M, Trost M, van Aalten DM, Alessi DR, Prescott AR, Knebel A, Walden H, Muqit MM (2015) Binding to serine 65-phosphorylated ubiquitin primes Parkin for optimal PINK1-dependent phosphorylation and activation. EMBO Rep 16(8):939–954. https://doi.org/10.15252/embr.201540352

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  86. Springer W, Kahle PJ (2011) Regulation of PINK1-Parkin-mediated mitophagy. Autophagy 7(3):266–278. https://doi.org/10.4161/auto.7.3.14348

    Article  PubMed  CAS  Google Scholar 

  87. Ivankovic D, Chau KY, Schapira AH, Gegg ME (2016) Mitochondrial and lysosomal biogenesis are activated following PINK1/parkin-mediated mitophagy. J Neurochem 136(2):388–402. https://doi.org/10.1111/jnc.13412

    Article  PubMed  CAS  Google Scholar 

  88. Lazarou M, Sliter DA, Kane LA, Sarraf SA, Wang C, Burman JL, Sideris DP, Fogel AI, Youle RJ (2015) The ubiquitin kinase PINK1 recruits autophagy receptors to induce mitophagy. Nature 524(7565):309–314. https://doi.org/10.1038/nature14893

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  89. Choubey V, Cagalinec M, Liiv J, Safiulina D, Hickey MA, Kuum M, Liiv M, Anwar T, Eskelinen EL, Kaasik A (2014) BECN1 is involved in the initiation of mitophagy: it facilitates PARK2 translocation to mitochondria. Autophagy 10(6):1105–1119. https://doi.org/10.4161/auto.28615

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  90. Orvedahl A, Sumpter R Jr, Xiao G, Ng A, Zou Z, Tang Y, Narimatsu M, Gilpin C, Sun Q, Roth M, Forst CV, Wrana JL, Zhang YE, Luby-Phelps K, Xavier RJ, Xie Y, Levine B (2011) Image-based genome-wide siRNA screen identifies selective autophagy factors. Nature 480(7375):113–117. https://doi.org/10.1038/nature10546

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  91. Novak I, Kirkin V, McEwan DG, Zhang J, Wild P, Rozenknop A, Rogov V, Löhr F, Popovic D, Occhipinti A, Reichert AS, Terzic J, Dötsch V, Ney PA, Dikic I (2010) Nix is a selective autophagy receptor for mitochondrial clearance. EMBO Rep 11(1):45–51. https://doi.org/10.1038/embor.2009.256

    Article  PubMed  CAS  Google Scholar 

  92. Zhang J, Ney PA (2009) Role of BNIP3 and NIX in cell death, autophagy, and mitophagy. Cell Death Differ 16:939–946. https://doi.org/10.1038/cdd.2009

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  93. Sandoval H, Thiagarajan P, Dasgupta SK, Schumacher A, Prchal JT, Chen M, Wang J (2008) Essential role for Nix in autophagic maturation of erythroid cells. Nature 454(7201):232–235. https://doi.org/10.1038/nature07006

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  94. Schweers RL, Zhang J, Randall MS, Loyd MR, Li W, Dorsey FC, Kundu M, Opferman JT, Cleveland JL, Miller JL, Ney PA (2007) NIX is required for programmed mitochondrial clearance during reticulocyte maturation. Proc Natl Acad Sci USA 104(49):19500–19505. https://doi.org/10.1073/pnas.0708818104

    Article  PubMed  Google Scholar 

  95. Rozenknop A, Rogov VV, Rogova NY, Löhr F, Güntert P, Dikic I, Dötsch V (2011) Characterization of the interaction of GABARAPL-1 with the LIR motif of NBR1. J Mol Biol 410(3):477–487. https://doi.org/10.1016/j.jmb.2011.05.003

    Article  PubMed  CAS  Google Scholar 

  96. Rogov VV, Suzuki H, Marinković M, Lang V, Kato R, Kawasaki M, Buljubašić M, Šprung M, Rogova N, Wakatsuki S, Hamacher-Brady A, Dötsch V, Dikic I, Brady NR, Novak I (2017) Phosphorylation of the mitochondrial autophagy receptor Nix enhances its interaction with LC3 proteins. Sci Rep 7(1):1131. https://doi.org/10.1038/s41598-017-01258-6

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  97. Ding WX, Ni HM, Li M, Liao Y, Chen X, Stolz DB, Dorn GW 2nd, Yin XM (2010) Nix is critical to two distinct phases of mitophagy, reactive oxygen species-mediated autophagy induction and Parkin-ubiquitin-p62-mediated mitochondrial priming. J Biol Chem 285(36):27879–27890. https://doi.org/10.1074/jbc.M110.119537

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  98. Narendra D, Tanaka A, Suen DF, Youle RJ (2008) Parkin is recruited selectively to impaired mitochondria and promotes their autophagy. J Cell Biol 183(5):795–803. https://doi.org/10.1083/jcb.200809125

    Article  PubMed  PubMed Central  Google Scholar 

  99. Azad MB, Chen Y, Henson ES, Cizeau J, McMillan–Ward E, Israels SJ, Gibson SB (2008) Hypoxia induces autophagic cell death in apoptosis–competent cells through a mechanism involving BNIP3. Autophagy 4(2):195–204. https://doi.org/10.4161/auto.5278

    Article  PubMed  CAS  Google Scholar 

  100. Novak I, Dikic I (2011) Autophagy receptors in developmental clearance of mitochondria. Autophagy 7(3):301–303. https://doi.org/10.4161/auto.7.3.14509

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  101. Zhang H, Bosch-Marce M, Shimoda LA, Tan YS, Baek JH, Wesley JB, Gonzalez FJ, Semenza GL (2008) Mitochondrial autophagy is an HIF-1-dependent adaptive metabolic response to hypoxia. J Biol Chem 283(16):10892–10903. https://doi.org/10.1074/jbc.M800102200

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  102. Liu L, Feng D, Chen G, Chen M, Zheng Q, Song P, Ma Q, Zhu C, Wang R, Qi W, Huang L, Xue P, Li B, Wang X, Jin H, Wang J, Yang F, Liu P, Zhu Y, Sui S, Chen Q (2012) Mitochondrial outer–membrane protein FUNDC1 mediates hypoxia–induced mitophagy in mammalian cells. Nat Cell Biol 14(2):177–185. https://doi.org/10.1038/ncb2422

    Article  PubMed  CAS  Google Scholar 

  103. Wu W, Li W, Chen H, Jiang L, Zhu R, Feng D (2016) FUNDC1 is a novel mitochondrial-associated-membrane (MAM) protein required for hypoxia-induced mitochondrial fission and mitophagy. Autophagy 12(9):1675–1676. https://doi.org/10.1080/15548627.2016.1193656

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  104. Chen G, Han Z, Feng D, Chen Y, Chen L, Wu H, Huang L, Zhou C, Cai X, Fu C, Duan L, Wang X, Liu L, Liu X, Shen Y, Zhu Y, Chen Q (2014) A regulatory signaling loop comprising the PGAM5 phosphatase and CK2 controls receptor-mediated mitophagy. Mol Cell 54(3):362–377. https://doi.org/10.1016/j.molcel.2014.02.034

    Article  PubMed  CAS  Google Scholar 

  105. Bhujabal Z, Birgisdottir ÅB, Sjøttem E, Brenne HB, Øvervatn A, Habisov S, Kirkin V, Lamark T, Johansen T (2017) FKBP8 recruits LC3A to mediate Parkin-independent mitophagy. EMBO Rep 18(6):947–961. https://doi.org/10.15252/embr.201643147

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  106. Fu M, St-Pierre P, Shankar J, Wang PT, Joshi B, Nabi IR (2013) Regulation of mitophagy by the Gp78 E3 ubiquitin ligase. Mol Biol Cell 24(8):1153–1162. https://doi.org/10.1091/mbc.E12-08-0607

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  107. Peng J, Ren KD, Yang J, Luo XJ (2016) Mitochondrial E3 ubiquitin ligase 1: a key enzyme in regulation of mitochondrial dynamics and functions. Mitochondrion 28:49–53. https://doi.org/10.1016/j.mito.2016.03.007

    Article  PubMed  CAS  Google Scholar 

  108. Ambivero CT, Cilenti L, Main S, Zervos AS (2014) Mulan E3 ubiquitin ligase interacts with multiple E2 conjugating enzymes and participates in mitophagy by recruiting GABARAP. Cell Signal 26(12):2921–2929. https://doi.org/10.1016/j.cellsig.2014.09.004

    Article  PubMed  CAS  Google Scholar 

  109. Bernhardt D, Hamann A, Osiewacz HD (2014) The role of mitochondria in fungal aging. Curr Opin Microbiol 22:1–7. https://doi.org/10.1016/j.mib.2014.09.007

    Article  PubMed  CAS  Google Scholar 

  110. Szklarczyk R, Nooteboom M, Osiewacz HD (2014) Control of mitochondrial integrity in ageing and disease. Philos Trans R Soc Lond B Biol Sci 369(1646):20130439. https://doi.org/10.1098/rstb.2013.0439

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  111. Wallace DC (2005) A mitochondrial paradigm of metabolic and degenerative diseases, aging, and cancer: a dawn for evolutionary medicine. Annu Rev Genet 39:359–407. https://doi.org/10.1146/annurev.genet.39.110304.095751

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  112. Payne BA, Chinnery PF (2015) Mitochondrial dysfunction in aging: much progress but many unresolved questions. Biochim Biophys Acta 1847(11):1347–1353. https://doi.org/10.1016/j.bbabio.2015.05.022

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  113. Diot A, Morten K, Poulton J (2016) Mitophagy plays a central role in mitochondrial ageing. Mamm Genome 27(7–8):381–395. https://doi.org/10.1007/s00335-016-9651-x

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  114. Ben-Meir A, Yahalomi S, Moshe B, Shufaro Y, Reubinoff B, Saada A (2015) Coenzyme Q-dependent mitochondrial respiratory chain activity in granulosa cells is reduced with aging. Fertil Steril 104(3):724–727. https://doi.org/10.1016/j.fertnstert.2015.05.023

    Article  PubMed  CAS  Google Scholar 

  115. Chougnet CA, Thacker RI, Shehata HM, Hennies CM, Lehn MA, Lages CS, Janssen EM (2015) Loss of phagocytic and antigen cross-presenting capacity in aging dendritic cells is associated with mitochondrial dysfunction. J Immunol 195(6):2624–2632. https://doi.org/10.4049/jimmunol.1501006

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  116. Kauppila TE, Kauppila JH, Larsson NG (2017) Mammalian mitochondria and aging: an update. Cell Metab 25(1):57–71. https://doi.org/10.1016/j.cmet.2016.09.017

    Article  PubMed  CAS  Google Scholar 

  117. Ferrington DA, Kapphahn RJ, Leary MM, Atilano SR, Terluk MR, Karunadharma P, Chen GK, Ratnapriya R, Swaroop A, Montezuma SR, Kenney MC (2016) Increased retinal mtDNA damage in the CFH variant associated with age-related macular degeneration. Exp Eye Res 145:269–277. https://doi.org/10.1016/j.exer.2016.01.018

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  118. Deininger P (2011) Alu elements: know the SINEs. Genome Biol 12(12):236. https://doi.org/10.1186/gb-2011-12-12-236

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  119. Pandey R, Bhattacharya A, Bhardwaj V, Jha V, Mandal AK, Mukerji M (2016) Alu-miRNA interactions modulate transcript isoform diversity in stress response and reveal signatures of positive selection. Sci Rep 6:32348. https://doi.org/10.1038/srep32348

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  120. Wang W, Wang WH, Azadzoi KM, Dai P, Wang Q, Sun JB, Zhang WT, Shu Y, Yang JH, Yan Z (2016) Alu RNA accumulation in hyperglycemia augments oxidative stress and impairs eNOS and SOD2 expression in endothelial cells. Mol Cell Endocrinol 426:91–100. https://doi.org/10.1016/j.mce.2016.02.008

    Article  PubMed  CAS  Google Scholar 

  121. Kaneko H, Dridi S, Tarallo V, Gelfand BD, Fowler BJ, Cho WG, Kleinman ME, Ponicsan SL, Hauswirth WW, Chiodo VA, Kariko K, Yoo JW, Lee DK, Hadziahmetovic M, Song Y, Misra S, Chaudhuri G, Buaas FW, Braun RE, Hinton DR, Zhang Q, Grossniklaus HE, Provis JM, Madigan MC, Milam AH, Justice NL, Albuquerque RJ, Blandford AD, Bogdanovich S, Hirano Y, Witta J, Fuchs E, Littman DR, Ambati BK, Rudin CM, Chong MM, Provost P, Kugel JF, Goodrich JA, Dunaief JL, Baffi JZ, Ambati J (2011) DICER1 deficit induces Alu RNA toxicity in age-related macular degeneration. Nature 471(7338):325–330. https://doi.org/10.1038/nature09830

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  122. Tarallo V, Hirano Y, Gelfand BD, Dridi S, Kerur N, Kim Y, Cho WG, Kaneko H, Fowler BJ, Bogdanovich S, Albuquerque RJ, Hauswirth WW, Chiodo VA, Kugel JF, Goodrich JA, Ponicsan SL, Chaudhuri G, Murphy MP, Dunaief JL, Ambati BK, Ogura Y, Yoo JW, Lee DK, Provost P, Hinton DR, Núñez G, Baffi JZ, Kleinman ME, Ambati J (2012) DICER1 loss and Alu RNA induce age-related macular degeneration via the NLRP3 inflammasome and MyD88. Cell 149(4):847–859. https://doi.org/10.1016/j.cell.2012.03.036

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  123. Kerur N, Fukuda S, Banerjee D, Kim Y, Fu D, Apicella I, Varshney A, Yasuma R, Fowler BJ, Baghdasaryan E, Marion KM, Huang X, Yasuma T, Hirano Y, Serbulea V, Ambati M, Ambati VL, Kajiwara Y, Ambati K, Hirahara S, Bastos-Carvalho A, Ogura Y, Terasaki H, Oshika T, Kim KB, Hinton DR, Leitinger N, Cambier JC, Buxbaum JD, Kenney MC, Jazwinski SM, Nagai H, Hara I, West AP, Fitzgerald KA, Sadda SR, Gelfand BD, Ambati J (2018) cGAS drives noncanonical-inflammasome activation in age-related macular degeneration. Nat Med 24(1):50–61. https://doi.org/10.1038/nm.4450

    Article  PubMed  CAS  Google Scholar 

  124. Childs BG, Gluscevic M, Baker DJ, Laberge RM, Marquess D, Dananberg J, van Deursen JM (2017) Senescent cells: an emerging target for diseases of ageing. Nat Rev Drug Discov 16(10):718–735. https://doi.org/10.1038/nrd.2017.116

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  125. Korolchuk VI, Miwa S, Carroll B, von Zglinicki T (2017) Mitochondria in cell senescence: is mitophagy the weak link? EBioMedicine 21:7–13. https://doi.org/10.1016/j.ebiom.2017.03.020

    Article  PubMed  PubMed Central  Google Scholar 

  126. Childs BG, Durik M, Baker DJ, van Deursen JM (2015) Cellular senescence in aging and age-related disease: from mechanisms to therapy. Nat Med 21(12):1424–1435. https://doi.org/10.1038/nm.4000

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  127. Coppé J-P, Patil CK, Rodier F, Sun Y, Muñoz DP, Goldstein J, Nelson PS, Desprez PY, Campisi J (2008) Senescence-associated secretory phenotypes reveal cell-nonautonomous functions of oncogenic RAS and the p53 tumor suppressor. PLoS Biol 6(12):2853–2868. https://doi.org/10.1371/journal.pbio.0060301

    Article  PubMed  CAS  Google Scholar 

  128. Passos JF, Saretzki G, Ahmed S, Nelson G, Richter T, Peters H, Wappler I, Birket MJ, Harold G, Schaeuble K, Birch-Machin MA, Kirkwood TB, von Zglinicki T (2007) Mitochondrial dysfunction accounts for the stochastic heterogeneity in telomere-dependent senescence. PLoS Biol 5:e110. https://doi.org/10.1371/journal.pbio.0050110

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  129. Wiley CD, Velarde MC, Lecot P, Liu S, Sarnoski EA, Freund A, Shirakawa K, Lim HW, Davis SS, Ramanathan A, Gerencser AA, Verdin E, Campisi J (2016) Mitochondrial dysfunction induces senescence with a distinct secretory phenotype. Cell Metab 23(2):303–314. https://doi.org/10.1016/j.cmet.2015.11.011

    Article  PubMed  CAS  Google Scholar 

  130. Passos JF, Nelson G, Wang C, Richter T, Simillion C, Proctor CJ, Miwa S, Olijslagers S, Hallinan J, Wipat A, Saretzki G, Rudolph KL, Kirkwood TB, von Zglinicki T (2010) Feedback between p21 and reactive oxygen production is necessary for cell senescence. Mol Syst Biol 6:347. https://doi.org/10.1038/msb.2010.5

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  131. Ferrington DA, Sinha D, Kaarniranta K (2016) Defects in retinal pigment epithelial cell proteolysis and the pathology associated with age-related macular degeneration. Prog Retin Eye Res 51:69–89. https://doi.org/10.1016/j.preteyeres.2015.09.002

    Article  PubMed  CAS  Google Scholar 

  132. Marazita MC, Dugour A, Marquioni-Ramella MD, Figueroa JM, Suburo AM (2016) Oxidative stress-induced premature senescence dysregulates VEGF and CFH expression in retinal pigment epithelial cells: implications for age-related macular degeneration. Redox Biol 7:78–87. https://doi.org/10.1016/j.redox.2015.11.011

    Article  PubMed  CAS  Google Scholar 

  133. Young AR, Narita M, Ferreira M, Kirschner K, Sadaie M, Darot JF, Tavaré S, Arakawa S, Shimizu S, Watt FM, Narita M (2009) Autophagy mediates the mitotic senescence transition. Genes Dev 23(7):798–803. https://doi.org/10.1101/gad.519709

    Article  PubMed  PubMed Central  Google Scholar 

  134. Rubinsztein DC, Mariño G, Kroemer G (2011) Autophagy and aging. Cell 146(5):682–695. https://doi.org/10.1016/j.cell.2011.07.030

    Article  PubMed  CAS  Google Scholar 

  135. Dalle Pezze P, Nelson G, Otten EG, Korolchuk VI, Kirkwood TB, von Zglinicki T, Shanley DP (2014) Dynamic modelling of pathways to cellular senescence reveals strategies for targeted interventions. PLoS Comput Biol 10(8):e1003728. https://doi.org/10.1371/journal.pcbi.1003728

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  136. García-Prat L, Martínez-Vicente M, Perdiguero E, Ortet L, Rodríguez-Ubreva J, Rebollo E, Ruiz-Bonilla V, Gutarra S, Ballestar E, Serrano AL, Sandri M, Muñoz-Cánoves P (2016) Autophagy maintains stemness by preventing senescence. Nature 529(5784):37–42. https://doi.org/10.1038/nature16187

    Article  PubMed  CAS  Google Scholar 

  137. Ahmad T, Sundar IK, Lerner CA, Gerloff J, Tormos AM, Yao H, Rahman I (2015) Impaired mitophagy leads to cigarette smoke stress-induced cellular senescence: implications for chronic obstructive pulmonary disease. FASEB J 29(7):2912–2929. https://doi.org/10.1096/fj.14-268276

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  138. Bueno M, Lai YC, Romero Y, Brands J, St Croix CM, Kamga C, Corey C, Herazo-Maya JD, Sembrat J, Lee JS, Duncan SR, Rojas M, Shiva S, Chu CT, Mora AL (2015) PINK1 deficiency impairs mitochondrial homeostasis and promotes lung fibrosis. J Clin Investig 125(3):521–538. https://doi.org/10.1172/JCI74942

    Article  PubMed  Google Scholar 

  139. Ohtsuji M, Katsuoka F, Kobayashi A, Aburatani H, Hayes JD, Yamamoto M (2008) Nrf1 and Nrf2 play distinct roles in activation of antioxidant response element-dependent genes. J Biol Chem 283(48):33554–33562. https://doi.org/10.1074/jbc.M804597200

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  140. Dodson M, Redmann M, Rajasekaran NS, Darley-Usmar V, Zhang J (2015) KEAP1-NRF2 signalling and autophagy in protection against oxidative and reductive proteotoxicity. Biochem J 469(3):347–355. https://doi.org/10.1042/BJ20150568

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  141. Zhao Z, Chen Y, Wang J, Sternberg P, Freeman ML, Grossniklaus HE, Cai J (2011) Age-related retinopathy in NRF-2-deficient mice. PLoS One 6(4):e19456. https://doi.org/10.1371/journal.pone.0019456

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  142. Phillipson OT (2014) Management of the aging risk factor for Parkinson’s disease. Neurobiol Aging 35(4):847–857. https://doi.org/10.1016/j.neurobiolaging.2013.10.07

    Article  PubMed  Google Scholar 

  143. Ping Z, Zhang LF, Cui YJ, Chang YM, Jiang CW, Meng ZZ, Xu P, Liu HY, Wang DY, Cao XB (2015) The protective effects of salidroside from exhaustive exercise-induced heart injury by enhancing the PGC-1 α -NRF1/NRF2 pathway and mitochondrial respiratory function in rats. Oxid Med Cell Longev 2015:876825. https://doi.org/10.1155/2015/876825

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  144. Iacovelli J, Rowe GC, Khadka A, Diaz-Aguilar D, Spencer C, Arany Z, Saint-Geniez M (2016) PGC-1α induces human RPE oxidative metabolism and antioxidant capacity. Investig Ophthalmol Vis Sci 57(3):1038–1051. https://doi.org/10.1167/iovs.15-17758

    Article  CAS  Google Scholar 

  145. Jäger S, Handschin C, St-Pierre J, Spiegelman BM (2007) AMP-activated protein kinase (AMPK) action in skeletal muscle via direct phosphorylation of PGC-1alpha. Proc Natl Acad Sci USA 104(29):12017–12022. https://doi.org/10.1073/pnas.0705070104

    Article  PubMed  CAS  Google Scholar 

  146. Salminen A, Kaarniranta K, Kauppinen A (2013) Crosstalk between oxidative stress and SIRT1: impact on the aging process. Int J Mol Sci 14(2):3834–3859. https://doi.org/10.3390/ijms14023834

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  147. Egger A, Samardzija M, Sothilingam V, Tanimoto N, Lange C, Salatino S, Fang L, Garcia-Garrido M, Beck S, Okoniewski MJ, Neutzner A, Seeliger MW, Grimm C, Handschin C (2012) PGC-1α determines light damage susceptibility of the murine retina. PLoS One 7(2):e31272. https://doi.org/10.1371/journal.pone.0031272

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  148. Liang FQ, Godley BF (2003) Oxidative stress-induced mitochondrial DNA damage in human retinal pigment epithelial cells: a possible mechanism for RPE aging and age-related macular degeneration. Exp Eye Res 76(4):397–403. https://doi.org/10.1016/S0014-4835(03)00023-X

    Article  PubMed  CAS  Google Scholar 

  149. Sun N, Youle RJ, Finkel T (2016) The mitochondrial basis of aging. Mol Cell 61(5):654–666. https://doi.org/10.1016/j.molcel.2016.01.028

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  150. Rohrer B, Bandyopadhyay M, Beeson C (2016) Reduced metabolic capacity in aged primary retinal pigment epithelium (RPE) is correlated with increased susceptibility to oxidative stress. Adv Exp Med Biol 854:793–798. https://doi.org/10.1007/978-3-319-17121-0_106

    Article  PubMed  CAS  Google Scholar 

  151. He Y, Ge J, Burke JM, Myers RL, Dong ZZ, Tombran-Tink J (2010) Mitochondria impairment correlates with increased sensitivity of aging RPE cells to oxidative stress. J Ocul Biol Dis Inform 3(3):92–108. https://doi.org/10.1007/s12177-011-9061-y

    Article  CAS  Google Scholar 

  152. Gouras P, Ivert L, Neuringer M, Nagasaki T (2016) Mitochondrial elongation in the macular RPE of aging monkeys, evidence of metabolic stress. Graefes Arch Clin Exp Ophthalmol 254(6):1221–1227. https://doi.org/10.1007/s00417-016-3342-x

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  153. Field MG, Comer GM, Kawaji T, Petty HR, Elner VM (2012) Noninvasive imaging of mitochondrial dysfunction in dry age-related macular degeneration. Ophthalmic Surg Lasers Imaging 43(5):358–365. https://doi.org/10.3928/15428877-20120712-02

    Article  PubMed  Google Scholar 

  154. SanGiovanni JP, Arking DE, Iyengar SK, Elashoff M, Clemons TE, Reed GF, Henning AK, Sivakumaran TA, Xu X, DeWan A, Agrón E, Rochtchina E, Sue CM, Wang JJ, Mitchell P, Hoh J, Francis PJ, Klein ML, Chew EY, Chakravarti A (2009) Mitochondrial DNA variants of respiratory complex I that uniquely characterize haplogroup T2 are associated with increased risk of age-related macular degeneration. PLoS One 4(5):e5508. https://doi.org/10.1371/journal.pone.0005508

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  155. Li N, Ragheb K, Lawler G, Sturgis J, Rajwa B, Melendez JA, Robinson JP (2003) Mitochondrial complex I inhibitor rotenone induces apoptosis through enhancing mitochondrial reactive oxygen species production. J Biol Chem 278(10):8516–8525. https://doi.org/10.1074/jbc.M210432200

    Article  PubMed  CAS  Google Scholar 

  156. Lee SY, Oh JS, Rho JH, Jeong NY, Kwon YH, Jeong WJ, Ryu WY, Ahn HB, Park WC, Rho SH, Yoon YG, Jeong SY, Choi YH, Kim HY, Yoo YH (2014) Retinal pigment epithelial cells undergoing mitotic catastrophe are vulnerable to autophagy inhibition. Cell Death Dis 5:e1303. https://doi.org/10.1038/cddis.2014.266

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  157. Blasiak J, Szaflik J, Szaflik JP (2011) Implications of altered iron homeostasis for age-related macular degeneration. Front Biosci (Landmark Ed) 16:1551–1559. https://doi.org/10.2741/3804

    Article  CAS  Google Scholar 

  158. Mena NP, Urrutia PJ, Lourido F, Carrasco CM, Núñez MT (2015) Mitochondrial iron homeostasis and its dysfunctions in neurodegenerative disorders. Mitochondrion 21:92–105. https://doi.org/10.1016/j.mito.2015.02.001

    Article  PubMed  CAS  Google Scholar 

  159. Drysdale J, Arosio P, Invernizzi R, Cazzola M, Volz A, Corsi B, Biasiotto G, Levi S (2002) Mitochondrial ferritin: a new player in iron metabolism. Blood Cells Mol Dis 29(3):376–383. https://doi.org/10.1006/bcmd.2002.0577

    Article  PubMed  CAS  Google Scholar 

  160. Wang L, Yang H, Zhao S, Sato H, Konishi Y, Beach TG, Abdelalim EM, Bisem NJ, Tooyama I (2011) Expression and localization of mitochondrial ferritin mRNA in Alzheimer’s disease cerebral cortex. PLoS One 6(7):e22325. https://doi.org/10.1371/journal.pone.0022325

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  161. Stenirri S, Santambrogio P, Setaccioli M, Erba BG, Pia Manitto M, Rovida E, Ferrari M, Levi S, Cremonesi L (2011) Study of FTMT and ABCA4 genes in a patient affected by age-related macular degeneration: identification and analysis of new mutations. Clin Chem Lab Med 50(6):1021–1029. https://doi.org/10.1515/cclm-2011-0854

    Article  CAS  Google Scholar 

  162. Wang X, Yang H, Yanagisawa D, Bellier JP, Morino K, Zhao S, Liu P, Vigers P, Tooyama I (2016) Mitochondrial ferritin affects mitochondria by stabilizing HIF-1α in retinal pigment epithelium: implications for the pathophysiology of age-related macular degeneration. Neurobiol Aging 47:168–179. https://doi.org/10.1016/j.neurobiolaging.2016.07.025

    Article  PubMed  CAS  Google Scholar 

  163. Sripathi SR, He W, Prigge CL, Sylvester O, Um JY, Powell FL, Neksumi M, Bernstein PS, Choo DW, Bartoli M, Gutsaeva DR, Jahng WJ (2017) Interactome mapping guided by tissue-specific phosphorylation in age-related macular degeneration. Int J Sci Eng Res 8(2):680–699. https://doi.org/10.14299/ijser.2017.02.010

    Article  PubMed  PubMed Central  Google Scholar 

  164. Hyttinen JMT, Petrovski G, Salminen A, Kaarniranta K (2011) 5′-Adenosine monophosphate-activated protein kinase-mammalian target of rapamycin axis as therapeutic target for age-related macular degeneration. Rejuvenation Res 14(6):651–660. https://doi.org/10.1089/rej.2011.1220

    Article  PubMed  CAS  Google Scholar 

  165. Bové J, Martínez-Vicente M, Vila M (2011) Fighting neurodegeneration with rapamycin: mechanistic insights. Nat Rev Neurosci 12(8):437–452. https://doi.org/10.1038/nrn3068

    Article  PubMed  CAS  Google Scholar 

  166. Foretz M, Guigas B, Bertrand L, Pollak M, Viollet B (2014) Metformin: from mechanisms of action to therapies. Cell Metab 20(6):953–966. https://doi.org/10.1016/j.cmet.2014.09.018

    Article  PubMed  CAS  Google Scholar 

  167. Saisho Y (2015) Metformin and inflammation: its potential beyond glucose-lowering effect. Endocr Metab Immune Disord Drug Targets 15(3):196–205. https://doi.org/10.18632/aging.100444

    Article  PubMed  CAS  Google Scholar 

  168. Moiseeva O, Deschênes-Simard X, St-Germain E, Igelmann S, Huot G, Cadar AE, Bourdeau V, Pollak MN, Ferbeyre G (2013) Metformin inhibits the senescence-associated secretory phenotype by interfering with IKK/NF-κB activation. Aging Cell 12(3):489–498. https://doi.org/10.1111/acel.12075

    Article  PubMed  CAS  Google Scholar 

  169. Song YM, Lee WK, Lee YH, Kang ES, Cha BS, Lee BW (2016) Metformin restores Parkin-mediated mitophagy, suppressed by cytosolic p53. Int J Mol Sci 17(1):122. https://doi.org/10.3390/ijms17010122

    Article  PubMed Central  CAS  Google Scholar 

  170. Vousden KH, Ryan KM (2009) p53 and metabolism. Nat Rev Cancer 9(10):691–700. https://doi.org/10.1038/nrc2715

    Article  PubMed  CAS  Google Scholar 

  171. Cantó C, Auwerx J (2009) PGC-1alpha, SIRT1 and AMPK, an energy sensing network that controls energy expenditure. Curr Opin Lipidol 20(2):98–105. https://doi.org/10.1097/MOL.0b013e328328d0a4

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  172. 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 (2010) Caloric restriction and resveratrol promote longevity through the Sirtuin-1-dependent induction of autophagy. Cell Death Dis 1:e10. https://doi.org/10.1038/cddis.2009.8

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  173. Giovannini L, Bianchi S (2017) Role of nutraceutical SIRT1 modulators in AMPK and mTOR pathway: evidence of a synergistic effect. Nutrition 34:82–96. https://doi.org/10.1016/j.nut.2016.09.008

    Article  PubMed  CAS  Google Scholar 

  174. Georgakopoulos ND, Frison M, Alvarez MS, Bertrand H, Wells G, Campanella M (2017) Reversible Keap1 inhibitors are preferential pharmacological tools to modulate cellular mitophagy. Sci Rep 7(1):10303. https://doi.org/10.1038/s41598-017-07679-7

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  175. Morselli E, Mariño G, Bennetzen MV, Eisenberg T, Megalou E, Schroeder S, Cabrera S, Bénit P, Rustin P, Criollo A, Kepp O, Galluzzi L, Shen S, Malik SA, Maiuri MC, Horio Y, López-Otín C, Andersen JS, Tavernarakis N, Madeo F, Kroemer G (2011) Spermidine and resveratrol induce autophagy by distinct pathways converging on the acetylproteome. J Cell Biol 192(4):615–629. https://doi.org/10.1083/jcb.201008167

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  176. Ryu D, Mouchiroud L, Andreux PA, Katsyuba E, Moullan N, Nicolet-Dit-Félix AA, Williams EG, Jha P, Lo Sasso G, Huzard D, Aebischer P, Sandi C, Rinsch C, Auwerx J (2016) Urolithin A induces mitophagy and prolongs lifespan in C. elegans and increases muscle function in rodents. Nat Med 22(8):879–888. https://doi.org/10.1038/nm.4132

    Article  PubMed  CAS  Google Scholar 

  177. Fivenson EM, Lautrup S, Sun N, Scheibye-Knudsen M, Stevnsner T, Nilsen H, Bohr VA, Fang EF (2017) Mitophagy in neurodegeneration and aging. Neurochem Int 109:202–209. https://doi.org/10.1016/j.neuint.2017.02.007

    Article  PubMed  CAS  Google Scholar 

  178. Richter U, Lahtinen T, Marttinen P, Myöhänen M, Greco D, Cannino G, Jacobs HT, Lietzén N, Nyman TA, Battersby BJ (2013) A mitochondrial ribosomal and RNA decay pathway blocks cell proliferation. Curr Biol 23(6):535–541. https://doi.org/10.1016/j.cub.2013.02.019

    Article  PubMed  CAS  Google Scholar 

  179. Sun N, Yun J, Liu J, Malide D, Liu C, Rovira II, Holmström KM, Fergusson MM, Yoo YH, Combs CA, Finkel T (2015) Measuring in vivo mitophagy. Mol Cell 60(4):685–696. https://doi.org/10.1016/j.molcel.2015.10.009

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  180. Hasson SA, Fogel AI, Wang C, MacArthur R, Guha R, Heman-Ackah S, Martin S, Youle RJ, Inglese J (2015) Chemogenomic profiling of endogenous PARK2 expression using a genome-edited coincidence reporter. ACS Chem Biol 10(5):1188–1197. https://doi.org/10.1021/cb5010417

    Article  PubMed  CAS  Google Scholar 

  181. Hertz NT, Berthet A, Sos ML, Thorn KS, Burlingame AL, Nakamura K, Shokat KM (2013) A neo-substrate that amplifies catalytic activity of Parkinson’s-disease-related kinase PINK1. Cell 154(4):737–747. https://doi.org/10.1016/j.cell.2013.07.030

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  182. Blasiak J, Piechota M, Pawlowska E, Szatkowska M, Sikora E, Kaarniranta K (2017) Cellular senescence in age-related macular degeneration: can autophagy and DNA damage response play a role? Oxid Med Cell Longev 2017:5293258. https://doi.org/10.1155/2017/5293258

    Article  PubMed  PubMed Central  Google Scholar 

  183. Kozlowski MR (2012) RPE cell senescence: a key contributor to age-related macular degeneration. Med Hypotheses 78(4):505–510. https://doi.org/10.1016/j.mehy.2012.01.018

    Article  PubMed  Google Scholar 

  184. Knuppertz L, Osiewacz HD (2016) Orchestrating the network of molecular pathways affecting ageing: role of non-selective autophagy and mitophagy. Mech Ageing Dev 153:30–40. https://doi.org/10.1016/j.mad.2016.01.003

    Article  PubMed  CAS  Google Scholar 

Download references

Acknowledgements

The authors thank Mr. Thomas Dunlop for checking the language of the manuscript.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Juha M. T. Hyttinen.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Hyttinen, J.M.T., Viiri, J., Kaarniranta, K. et al. Mitochondrial quality control in AMD: does mitophagy play a pivotal role?. Cell. Mol. Life Sci. 75, 2991–3008 (2018). https://doi.org/10.1007/s00018-018-2843-7

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00018-018-2843-7

Keywords

Navigation