Download as pdf or txt
Download as pdf or txt
You are on page 1of 8

Neurochemistry International 109 (2017) 202e209

Contents lists available at ScienceDirect

Neurochemistry International
journal homepage: www.elsevier.com/locate/nci

Mitophagy in neurodegeneration and aging


Elayne M. Fivenson a, Sofie Lautrup a, b, Nuo Sun c, Morten Scheibye-Knudsen d,
Tinna Stevnsner b, Hilde Nilsen e, Vilhelm A. Bohr a, d, **, Evandro F. Fang a, *
a
Laboratory of Molecular Gerontology, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
b
Danish Aging Research Center, Department of Molecular Biology and Genetics, University of Aarhus, 8000 Aarhus C, Denmark
c
Center for Molecular Medicine, National Heart Lung and Blood Institute, NIH, Bethesda, MD 20892, USA
d
Center for Healthy Aging, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark
e
Department of Clinical Molecular Biology, University of Oslo and Akershus University Hospital, 1478 Lørenskog, Norway

a r t i c l e i n f o a b s t r a c t

Article history: Mitochondrial dysfunction contributes to normal aging and a wide spectrum of age-related diseases,
Received 10 February 2017 including neurodegenerative disorders such as Parkinson's disease and Alzheimer's disease. It is
Accepted 16 February 2017 important to maintain a healthy mitochondrial population which is tightly regulated by proteolysis and
Available online 21 February 2017
mitophagy. Mitophagy is a specialized form of autophagy that regulates the turnover of damaged and
dysfunctional mitochondria, organelles that function in producing energy for the cell in the form of ATP
and regulating energy homeostasis. Mechanistic studies on mitophagy across species highlight a so-
phisticated and integrated cellular network that regulates the degradation of mitochondria. Strategies
directed at maintaining a healthy mitophagy level in aged individuals might have beneficial effects. In
this review, we provide an updated mechanistic overview of mitophagy pathways and discuss the role of
reduced mitophagy in neurodegeneration. We also highlight potential translational applications of
mitophagy-inducing compounds, such as NADþ precursors and urolithins.
Published by Elsevier Ltd.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 202
2. Overview of mitophagy pathways . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 203
2.1. Mitophagy in mammals . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 203
2.2. Mitophagy in C. elegans . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 205
3. Mitophagy in neurodegeneration and aging . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 205
4. Sirtuins and mitophagy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 205
4.1. Linking energy sensing to mitochondrial homeostasis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 206
4.2. Nuclear to mitochondrial signaling and mitophagy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 206
5. Therapeutic interventions that target mitophagy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 206
6. Conclusions and future perspectives . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 207
Acknowledgments . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 207
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 207

1. Introduction

* Corresponding author. Aging is associated with a loss of physiological integrity,


** Corresponding author. Laboratory of Molecular Gerontology, National Institute including an imbalance in proteostasis and an increase in mito-
on Aging, National Institutes of Health, Baltimore, MD 21224, USA. chondrial dysfunction, which can be caused by compromised
E-mail addresses: BohrV@grc.nia.nih.gov (V.A. Bohr), evandro.fang@nih.gov,
autophagy and its subtype mitochondrial autophagy, termed
evandrofeifang@yahoo.com (E.F. Fang).

http://dx.doi.org/10.1016/j.neuint.2017.02.007
0197-0186/Published by Elsevier Ltd.
E.M. Fivenson et al. / Neurochemistry International 109 (2017) 202e209 203

mitophagy. Autophagy is the process by which cellular components (Lazarou et al., 2015). Parkin-mediated mitophagy can be sup-
are degraded and recycled within the cell. Autophagy can refer to pressed by deubiquitination of its substrates. USP8 (Ubiquitin
the nonspecific, cell-wide degradation of organelles or misfolded Specific Peptidase 8) was found to regulate mitophagy in human
proteins in nutrient-starved conditions, as well as the removal of cell lines (Durcan et al., 2014), while the reduction of USP30 (Bingol
specific damaged or superfluous organelles. Aging and age-related et al., 2014) and USP15 (Cornelissen et al., 2014) can increase
pathologies are associated with reductions in autophagy mitophagy and rescue mitochondrial phenotypes in fly models.
(Rubinsztein et al., 2011), and emerging evidence suggests that the Mutations in Parkin (Kitada et al., 1998) and PINK1 (Valente et al.,
upregulation of autophagy may delay the onset and ameliorate the 2004) lead to the autosomal recessive form of Parkinson's disease.
symptoms of age-related phenotypes (Madeo et al., 2010). A PINK1-/- mice exhibit mitochondrial dysfunction, including reduced
reduction in autophagy leads to neurodegeneration in mice (Hara mitochondrial respiration in the striatum at 3e4 months of age.
et al., 2006; Park et al., 2016; Komatsu et al., 2006) and is This dysfunction was only observed in control mice at 24 months of
thought to contribute to several neurodegenerative diseases in age, indicating PINK1-/- mice are sensitive to age-related damage
humans (Menzies et al., 2015). Mitochondria, classically referred to (Gautier et al., 2008). While Parkin/- mice exhibit a reduction in
as the “powerhouse” of the cell, produce cellular energy in the form proteins involved in mitochondrial function and oxidative stress
of ATP. However, a large body of work has established additional responses, as well as mild metabolic phenotypes (Palacino et al.,
and synergistic roles of the mitochondria in the regulation of 2004), they do not exhibit severe mitochondrial dysfunction or
cellular homeostasis (Wallace, 2013). Mitochondrial dysfunction is neuronal death, indicating the existence of Parkin-independent
considered a hallmark of aging (Lo  pez-Otín et al., 2013), and is mitochondrial clearance pathway(s) (Perez and Palmiter, 2005).
implicated in apoptosis, senescence, genome instability, inflam- However, when Parkin/- mice are crossed with a PolG mutant
mation, and metabolic disorders (Wallace, 2013; Scheibye-Knudsen mouse, characterized by increased mtDNA damage, the resulting
et al., 2014). The term “mitophagy” was first coined by Dr. Lemas- phenotypes include mitochondrial dysfunction and dopaminergic
ters in 2005 (Lemasters, 2005). Since then, mitophagy has been neuronal loss characteristic of Parkinson's disease (Pickrell et al.,
linked to various diseases, including neurodegenerative disorders 2015), indicating Parkin-deficient mice are susceptible to
(Palikaras and Tavernarakis, 2012) such as Parkinson's (Ryan et al., increased mtDNA damage.
2015), Huntington's (Khalil et al., 2015), and Alzheimer's (Ye et al., The PINK1/Parkin-dependent mitophagy pathway has been
2015), as well as normal physiological aging (Sun et al., 2016). In extensively studied in Drosophila (Greene et al., 2003; Yang et al.,
this review, we summarize recent findings linking mitophagy to 2006; Deng et al., 2008; Poole et al., 2008; Yang et al., 2008).
aging and neurodegeneration. We discuss connections between Knockout of Pink1 in Drosophila results in sterility, mitochondrial
mitochondrial turnover and genomic stability, explore therapeutic defects, increased sensitivity to stress (Clark et al., 2006), dopa-
interventions targeting mitophagy pathways, and highlight minergic neuronal loss and locomotive dysfunction (Park et al.,
emerging avenues of research in the field. 2006). These phenotypes are ameliorated by the overexpression
of Parkin, suggesting that PINK1 and Parkin function in the same
2. Overview of mitophagy pathways pathway to regulate mitochondrial maintenance (Clark et al.,
2006).
Accumulating evidence demonstrates that mitophagy functions Several mitophagy proteins have been reported to work in a
in fertilization and development, maintaining health throughout Parkin-independent manner, including AMBRA1, Bcl2-L-13,
life, and preventing age-related disease. Molecular mechanisms of FUNDC1, MUL1, and Nix/BNIP3L. AMBRA1 (the Activating Mole-
mitophagy have been intensively investigated in multiple species cule in Beclin1-Regulated Autophagy) functions in the autophagy
(Fig. 1). Mitophagy can either specifically eliminate damaged pathway, while also playing a pivotal role in embryogenesis and
mitochondria or clear all mitochondria during specialized devel- the development of the nervous system (Fimia et al., 2007).
opmental stages (fertilization and blood cell maturation) or star- Interestingly, AMBRA1 induces mitophagy independently of
vation (phosphoinositide 3-kinase/PI3K-dependent). Here we Parkin and p62/SQSTM1 (a major autophagy protein) through
summarize current knowledge of different mitophagy pathways in direct binding with LC3 (Strappazzon et al., 2015). The BH do-
mammalian cells, C. elegans, and yeast. mains of Bcl2-L-13 can independently induce mitochondrial
fragmentation, while the WXXI motif binds to LC3 and facilitates
2.1. Mitophagy in mammals mitophagy. In the absence of Parkin, Bcl2-L-13 retains its
mitophagy-inducing function (Murakawa et al., 2015). The inte-
Under nutrient-rich conditions, damaged and superfluous gral mitochondrial outer-membrane protein FUNDC1 was origi-
mitochondria are selectively degraded to maintain mitochondrial nally reported as a major player in hypoxia-induced mitophagy
homeostasis. One of the most studied pathways in clearing (Liu et al., 2012). FUNDC1 can regulate mitophagy both through
damaged mitochondria in mammalian cells is the PINK1/Parkin recruitment of LC3 via its typical LC3-binding motif Y (18)xxL
pathway (recently reviewed by (Youle and Narendra, 2011; Pickrell (21), and by regulation of mitochondrial dynamics through
and Youle, 2015)) (Fig. 1A). PINK1-Parkin-dependent mitophagy is interaction with fission and fusion proteins (DNM1L/DRP1 and
initiated when a decrease in mitochondrial membrane potential OPA1, respectively) (Liu et al., 2012; Chen et al., 2016). MUL1
caused by mitochondrial damage leads to the stabilization of the (mitochondrial ubiquitin ligase 1) is an E3 protein ligase that
ubiquitin kinase (PTEN)-induced kinase 1 (PINK1) on the outer localizes onto the mitochondrial outer membrane during
mitochondrial membrane. Here, it phosphorylates ubiquitin, lead- mitophagy (Yun et al., 2014). In PINK1 or parkin deficient
ing to the recruitment of the E3 ubiquitin ligase Parkin. PINK1 Drosophila, MUL1 can ameliorate disease phenotypes through
phosphorylation activates Parkin, which polyubiquitinates mito- the ubiquitin-dependent degradation of Mitofusin (Yun et al.,
chondrial proteins, leading to their association with the ubiquitin- 2014). MUL1 can also cleave sperm-derived mitochondria in a
binding domains of autophagy receptors and the formation of the PINK1-dependent manner in zygotes (Rojansky et al., 2016)
autophagosome. The autophagosome then fuses with the lysosome, (detailed below). Additionally, Nix/BNIP3L can induce mitophagy
leading to degradation of the mitochondria (Lazarou et al., 2015). in a PINK1/Parkin-independent manner (Song et al., 2016) (also
Alternatively, PINK1 can recruit autophagy receptors directly in a shown below). Some proteins involved in Parkin-independent
Parkin-independent manner, leading to low levels of mitophagy mitophagy pathways, such as Ambra1, Nix, and FUNDC1, also
204 E.M. Fivenson et al. / Neurochemistry International 109 (2017) 202e209

Fig. 1. Overview of Mitophagy Pathways. A. PINK-1/Parkin-mediated pathway. PINK-1 is stabilized on the outer mitochondrial membrane of damaged mitochondria where it
phosphorylates ubiquitin, leading to the activation of parkin. Parkin polyubiquitinates mitochondrial proteins, leading to the association with autophagy receptors and the for-
mation of the autophagosome. The autophagosome then fuses with the lysosome, leading to degradation. Alternatively, PINK-1 can recruit autophagy receptors directly in a parkin-
independent manner, leading to low levels of mitophagy. B. Reticulocyte mitophagy. Upon maturation signal, NIX localizes to the outer mitochondrial membrane of reticulocyte
mitochondria. NIX binds to LC3, leading to the formation of the autophagosome. C. Mammalian sperm mitophagy. Ubiquitinated paternal sperm associates with VCP. VCP facilitates
the presentation of ubiquitinated (Ub) sperm mitochondrial proteins to the 26S proteasome, leading to degradation. Synchronously, SQSTM1 binds to other ubiquitinated
mitochondrial proteins leading to sequestration within an autophagosome. D. Mitochondria-derived vesicles. Unfolded oxidized proteins lead to aggregation. Cardiolipin is locally
oxidized to phosphatidic acid, leading to membrane curvature. PINK-1 is localized to the outer mitochondrial membrane where it recruits parkin. The vesicle is formed and the
cargo is transported to the lysosome for degradation. E. C. elegans somatic tissue. PINK-1 is stabilized on the OMM of damaged/superfluous mitochondria, leading to the
recruitment of parkin. Parkin ubiquitinates DCT-1 (homolog of NIX/BNIP3L), which associates with LGG-1 (homolog of LC3), leading to the formation of the autophagosome. F.
C. elegans sperm. Loss of membrane integrity triggers the release of endonuclease G from inner mitochondrial membrane space to the mitochondrial matrix, where it degrades
mtDNA. G. Yeast. Association of OMM protein Atg32 to isolation membrane-bound Atg8 directly or through its interaction with Atg11 recruits targeted mitochondria to the
autophagosome.

have roles in traditional Parkin-dependent mitophagy (Van Elimination of sperm mitochondria in metazoan cells may require a
Humbeeck et al., 2011; Ding et al., 2010; Wu et al., 2014). Thus, combined action of p62-dependent autophagy and valosin-
in mammalian cells, there are several mitophagy pathways containing protein (VCP)-mediated ubiquitination of sperm mito-
which can compensate for the loss of others. chondria to the 26S proteasome (Fig. 1C).
In addition to the clearance of damaged mitochondria, mam- In addition to the recycling of entire mitochondria, the isolation
mals have also developed specialized mitophagy pathways to and digestion of specific damaged proteins within mitochondria
eliminate mitochondria during developmental stages such as can also occur. In this pathway, oxidized proteins in the mito-
fertilization and blood cell maturation. During the process of chondrial matrix are engulfed in mitochondrial-derived vesicles
reticulocyte maturation into red blood cells, mitochondria are (MDVs) and transported to the lysosomes (Sugiura et al., 2014;
completely cleared by the Nix/BNIP3L-dependent mitophagy Soubannier et al., 2012) or to peroxisomes (Neuspiel et al., 2008).
pathway (Sandoval et al., 2008) (Fig. 1B). Nix is a member of the Bcl- MDVs contain oxidized proteins, which may indicate protein
2 family, and it induces mitophagy by decreasing mitochondrial damage or trigger protein aggregation (Sugiura et al., 2014). MDV
membrane potential. transport to the lysosome is dependent on PINK1 and Parkin
Mitochondria are maternally inherited. After fertilization, (McLelland et al., 2014); however, this process is independent of
paternal mitochondria are selectively degraded leaving only other autophagic proteins such as ATG5 and LC3. MDVs may be an
maternally-derived oocyte mitochondria. Deficiency in this process early response to protein damage within the mitochondria in an
leads to embryonic lethality, signifying its evolutionary significance attempt to correct mitochondrial damage before complete degra-
(Rojansky et al., 2016; Song et al., 2016; Zhou et al., 2016). dation via mitophagy (Sugiura et al., 2014) (Fig. 1D).
E.M. Fivenson et al. / Neurochemistry International 109 (2017) 202e209 205

2.2. Mitophagy in C. elegans linked to Parkinson's disease (PD) (see section 2.1), a neurodegen-
erative disorder characterized by a loss of dopaminergic neurons in
Decades of research in cross-species studies suggests that many the substantia nigra as well as the accumulation of Lewy bodies, a
of the cellular pathways involved in neuronal function and aging form of protein aggregates.
are evolutionally conserved. In C. elegans, mitophagy plays impor- Mitochondrial dysfunction, including reduced membrane po-
tant roles in lifespan and healthspan (Palikaras et al., 2015). The tential and respiration, is also associated with Huntington's disease
C. elegans mitochondrial protein DCT-1 is a putative orthologue to (HD) (Bossy-Wetzel et al., 2008). HD is an autosomal dominant
the human mitophagy protein Nix/BNIP3L. DCT-1 works down- neurodegenerative disorder caused by an expansion of CAG repeats
stream of the PINK-1/Parkin pathway and is upregulated by the in the huntingtin gene, leading to the accumulation of protein ag-
oxidative stress response transcription factor SKN-1 (ortholog of gregates and neuronal death. It has been reported that HD mouse
mammalian NRF2) and the FOXO transcription factor DAF-16 models have reduced activity of PGC-1a, the master regulator of
(ortholog of mammalian FOXO3) (Palikaras et al., 2015) (Fig. 1E). mitochondrial biogenesis (Weydt et al., 2006) and recently found to
Additionally, as in mammals, C. elegans sperm-derived mitochon- be important in the formation of hippocampal dendritic spines and
dria are eliminated after fertilization. Recent work in C. elegans synapses (Cheng et al., 2012). A reduction in mitophagy may also
suggests that CPS-6, a mitochondrial endonuclease G (Song et al., contribute to mitochondrial dysfunction in HD. Human Hunting-
2016; Zhou et al., 2016), is required to degrade the paternal mito- tin's transgene (HTT) mice crossed with the mt-keima mouse
chondrial genome (Fig. 1F). Further research on the mechanisms of indicated reduced mitophagy in the dentate gyrus (Sun et al., 2015).
autophagy and mitophagy in C. elegans will accelerate both GAPDH, a protein which catalyzes the sixth step of glycolysis, also
mechanistic and translational studies in the fields of biological functions in the direct engulfment of damaged mitochondria by
aging and neurodegeneration. lysosomes. In normal cells, expression of expanded polyglutamine
tracts oxidized inactive GAPDH (iGAPDH) to induce micro-
3. Mitophagy in neurodegeneration and aging mitophagy. Mechanistically, iGAPDH signals the induction of
direct engulfment of damaged mitochondria into lysosomes.
Mitochondria are key players in a wide range of pathways However, expanded polyglutamine repeats (mutant Huntingtin)
involved in cellular and tissue homeostasis and aging, including inhibit GAPDH-induced mitophagy through abnormal interaction
cellular senescence, stem cell function, inflammation, nuclear gene with GAPDH (Hwang et al., 2015). Hwang et al. reported a reduction
expression, epigenetics, calcium regulation, apoptosis, telomere in GAPDH-induced mitophagy in HD cell models due to the inac-
maintenance, and genomic stability (Scheibye-Knudsen et al., 2014; tivation of GAPDH by polyglutamine expansion (Hwang et al.,
Sun et al., 2016; Fang et al., 2016a). Mitochondrial dysfunction is a 2015).
hallmark of aging (Lo  pez-Otín et al., 2013), demonstrated by the Alzheimer's disease is the most prevalent form of neuro-
cross-species accumulation of dysfunctional mitochondria with age degeneration in aging populations. It is characterized by progres-
(Wallace, 1999). Mutations (Hudson et al., 1998; Fayet et al., 2002) sive cognitive decline, memory loss, and neuronal death in the
and deletions (Corral-Debrinski et al., 1992) in the mitochondrial cerebral cortex. Brain sections from Alzheimer's patients are char-
genome accumulate with age, leading to bioenergetic dysfunction acterized by neurofibrillary tangles composed of hyper-
and age-related pathologies (Bratic and Larsson, 2013; Larsson, phosphorylated tau protein and plaques of amyloid beta.
2010). Mitochondrial dysfunction can also induce senescence Mitochondrial dysfunction is also implicated in the disease pa-
(termed mitochondrial dysfunction-induced senescence thology (Casley et al., 2002; Kerr et al., 2017). The activation of
(“MiDAS”)), recently shown to contribute extensively to age-related SIRT1 (See “Sirtuins and Mitophagy”) can reduce the amount of
pathologies (Wiley et al., 2016). beta-amyloid in brain, similar to the reduction mediated by caloric
In C. elegans, mitochondria accumulate with age, which may be restriction (Qin et al., 2006).
due to a reduction in the clearing of damaged mitochondria via In addition, compromised mitophagy may also induce amyo-
mitophagy (Palikaras et al., 2015). Furthermore, induction of trophic lateral sclerosis (ALS), a fatal neurological disease caused by
mitophagy is required for the lifespan extension of several long- dysfunction of the motor neurons. ALS-related OPTN and TBK1 gene
lived C. elegans mutants including the IGF/IGF-1 mutants. Life- mutations can cause compromised mitophagy (Moore and
span extension in C. elegans due to mild mitochondrial dysfunction Holzbaur, 2016). Taken together, these studies suggest that
is also mediated by mitophagy (Schiavi et al., 2015). Sun et al. reduced mitophagy contributes to several neurodegenerative dis-
developed the mt-keima mouse to measure mitophagy in vivo and eases and age-related neurogenerative decline.
demonstrated that mitophagy declines by approximately 70% in the
dentate gyrus, a region of the brain important for memory and 4. Sirtuins and mitophagy
learning, between 3 and 21-month old mice (Sun et al., 2015). They
also observed an increase in mitophagy in the PolG mutator mouse, The conserved sirtuin (silent information regulator) family of
associating an increase in mtDNA damage with an increase in proteins are a group of NADþ dependent deacetylases that have
mitophagy (Trifunovic et al., 2004). The authors speculate that it is important roles in metabolism and aging (Houtkooper et al., 2012).
not an increase in mitochondrial damage that contributes to The mammalian sirtuin family consists of SIRT1-7. SIRT3-5 are
mitochondrial-driven age-related pathologies, but rather a decline predominately targeted to the mitochondria, SIRT1, SIRT6 and
in the cellular capacity to mitigate the damage via mitophagy (Sun SIRT7 are mainly nuclear, and SIRT2 is cytoplasmic. Several excel-
et al., 2015). lent reviews have discussed the properties of sirtuins and their role
Neurons have an exceptionally high demand for ATP, which is in disease (Imai and Guarente, 2014, 2016; Haigis and Sinclair,
needed for axonal transport, maintenance of ion gradients and 2010). Sirtuin activity is generally correlated with an increase in
membrane potential, and generation of synaptic vesicles. ATP lifespan and healthspan in yeast (Howitz et al., 2003), flies (Bauer
production in neurons relies predominately on oxidative phos- et al., 2009), worms (Tissenbaum and Guarente, 2001), and mice
phorylation. Therefore, mitochondrial health is essential for (Kanfi et al., 2012; Boily et al., 2008). This is in part due to increased
neuronal function (Rugarli and Langer, 2012). Given these unique stress response. For example, the increased resistance to oxidative
features of neurons, the nervous system may be especially sensitive stress in mouse neurons following exercise is mediated by mito-
to mitochondrial damage. Dysfunctional mitophagy is closely chondrial SIRT3 (Cheng et al., 2016). Furthermore, huntingtin and
206 E.M. Fivenson et al. / Neurochemistry International 109 (2017) 202e209

epilepsy mouse models lacking SIRT3 were more susceptible to XPC models, a DNA repair disorder not associated with neuro-
neurodegeneration (Cheng et al., 2016). Sirtuin enzymatic activity degeneration. Taken together, these results indicate that cellular
is dependent on cellular NADþ levels, which decline with age across levels of NADþ can modulate mitochondrial homeostasis and
species (Canto et al., 2015). In rats, the NADþ/NADH ratio decreases genome stability and may provide therapeutic targets for age-
in all organs by middle age compared to young rats (Braidy et al., related neurodegeneration and neurodegenerative disease.
2011). Similar findings were reported in human skin samples SIRT1 can also regulate mitochondrial homeostasis in a PGC-1a
(Massudi et al., 2012). Cellular levels of NADþ and SIRT1 activity independent manner. Gomes et al. demonstrated that when nu-
have recently gained attention for their role in regulating mito- clear NADþ declines with age, SIRT1 activity is reduced and the
chondrial biogenesis and mitophagy, and may provide novel ther- transcription factor HIF-1a is stabilized (Gomes et al., 2013). The
apeutic targets for neurodegeneration and age-related disease. authors suggest that chronic activation of HIF-1a leads to a
“pseudohypoxic state” in which the transcription of
4.1. Linking energy sensing to mitochondrial homeostasis mitochondrially-encoded genes is reduced, leading to an imbal-
ance between nuclear and mitochondrial transcription, and
Mitochondrial turnover and mitochondrial biogenesis must be eventually mitochondrial dysfunction. Interestingly, recent work
tightly balanced in the cell to maintain energy homeostasis. This is by Jain et al. demonstrated that increased HIF activity due to
in part regulated by the AMP-activated protein kinase (AMPK). hypoxia ameliorated several features and dramatically increased
AMPK functions as an energy sensor, stimulating ATP production the lifespan of mouse models of Leigh Syndrome (Jain et al., 2016).
and suppressing ATP consumption in conditions of high cellular Thus, the dual role of the HIF pathway to regulate mitochondrial
AMP/ATP ratio. AMPK promotes mitophagy through multiple health may be context-dependent and remains to be fully
mechanisms. First, AMPK phosphorylation of ULK1/ULK2 elucidated.
(mammalian orthologs of yeast Atg1) is required for mitophagy
(Egan et al., 2011). Since AMPK can increase intracellular NADþ 5. Therapeutic interventions that target mitophagy
levels (Canto et al., 2009), it may also promote SIRT1-dependent
mitophagy (Fang et al., 2014, 2016b; Fang and Bohr, 2016). SIRT1 Given that mitochondrial dysfunction and compromised auto-
activity enhances mitophagy via FOXO signaling and promotes phagy/mitophagy can induce a wide spectrum of diseases,
mitochondrial biogenesis by activating PGC-1a (Fang et al., 2016a). including neurodegenerative diseases and premature aging, phar-
This pathway links energy sensing to mitochondrial homeostasis macological maintenance of mitophagy may have broad trans-
(Mihaylova and Shaw, 2011). lational applications. First, direct activators of mitophagy
machinery, such as modulators of PINK1 (Hertz et al., 2013) and
4.2. Nuclear to mitochondrial signaling and mitophagy Parkin (Hasson et al., 2015) have been shown to increase mitoph-
agy. Activation of sirtuins through sirtuin activating compounds
Nuclear to mitochondrial signaling regulates mitochondrial (STACs), has also been shown to be a viable strategy for increasing
function, and can have profound impacts on aging and disease mitophagy. Drugs such as resveratrol (Howitz et al., 2003) and
(Fang et al., 2016a). This signaling is mediated by a network of metformin (Song et al., 2015), both of which increase mitophagy,
sirtuins. NADþ is a key metabolite consumed by sirtuins and by the fall into this class. Sirtuin activity can also be mediated through
DNA damage response protein poly (ADP-ribose) polymerase 1 supplementation of NADþ precursors such as NR and NMN, or via
(PARP1). PARP1 detects breaks in DNA and recruits DNA repair inhibitors of NADþ-consumers such as PARP, both of which have
proteins through a process called PARylation in which PAR chains been found to improve several facets of age-related decline,
are generated and NADþ is consumed (Fang et al., 2016a; Kameshita including neurodegeneration (Bonkowski and Sinclair, 2016).
et al., 1984). PARP1 may also participate in the detection of G4 Recently, Mills et al. reported that long term treatment of NMN
structures (Scheibye-Knudsen et al., 2016). The accumulation of delayed the onset of age-related decline in wildtype mice through
nuclear DNA damage, which occurs with age (Chow and Herrup, enhanced energy metabolism, suppression of weight increase, and
2015), can lead to persistent activation of PARP1, which depletes other features (Mills et al., 2016). Mitochondrial dysfunction and
cellular levels of NADþ and reduces sirtuin activity (Gibson and defective mitophagy contribute to the severe neurodegeneration
Kraus, 2012). In C. elegans, when the ortholog of PARP-1, pme-1, phenotypes of several DNA repair deficient premature aging dis-
was knocked down, mitochondrial function improved due to eases, such as CS, XPA, and AT. Intriguingly, NADþ replenishment
increased NADþ levels and SIR-2.1 (the C. elegans ortholog of SIRT1) can ameliorate disease phenotypes and improve both lifespan and
activity and through UPRmt activation. This resulted in lifespan healthspan in C. elegans and mouse disease models.
extension and delayed onset of age-related phenotypes Mitophagy can also be induced by bioactive natural compounds,
(Mouchiroud et al., 2013). such as antibiotics and plant metabolites. Actinonin is a naturally
In addition to normal physiological aging, DNA repair-deficient occurring antibiotic found to increase mitophagy in neuronal stem
neurodegenerative progeria diseases, including Cockayne Syn- cells derived from the mt-Keima mouse (Sun et al., 2015). The
drome (CS), Xeroderma Pigmentosum group A (XPA) (Fang et al., mechanism through which actinonin induces mitophagy may be
2014), and Ataxia Telangiectasia (AT) (Fang et al., 2016b), exhibit linked to the depletion of mitochondrial ribosomes and RNA decay
clinical features similar to classic mitochondrial disorders such as (Richter et al., 2013), although the connection between this
mitochondrial encephalomyopathy, lactic acidosis, stroke-like epi- pathway and mitochondrial quality control remains to be
sodes (MELAS) and Myoclonic epilepsy with ragged-red fibers completely elucidated. Ryu et al. recently characterized a novel
(MERRF) syndromes (Scheibye-Knudsen et al., 2013). XPA and AT mitophagy-inducer Urolithin A (Park et al., 2016), a metabolite
demonstrate mitochondrial dysfunction due to reduced SIRT1 ac- derived from pomegranates. Urolithin A extends the lifespan of
tivity, stemming from an increase in DNA damage, persistent acti- C. elegans and improves muscle function in mice through mito-
vation of PARP1, and depletion of NADþ (Fang and Bohr, 2016; Chen chondrial maintenance via upregulation of mitophagy (Ryu et al.,
et al., 2015). For example, treatment with PARP inhibitors and 2016). More work remains to be done to deduce the mechanism
supplementation of NADþ precursors attenuated mitochondrial behind which Urolithin A increases mitophagy. In conclusion, based
dysfunction in XPA models and extended lifespan in xpa-1 worms on the availability of different mitophagy screening animal models,
(Fang et al., 2014). Interestingly, similar results were not found in including C. elegans and mice, we expect an increase in the
E.M. Fivenson et al. / Neurochemistry International 109 (2017) 202e209 207

Fig. 2. Mitophagy in neurodegeneration and aging.

discovery of new mitophagy-inducing compounds which will whose published articles we were unable to cite owing to space
propel further potential clinical studies. limitations. We thank Dr. Beverly Baptiste and Mr. Jesse Kerr for
critical reading of the manuscript. We thank Marc Raley for some of
6. Conclusions and future perspectives the art works. This research was supported by the Intramural
Research Program of the National Institute on Aging, including a
A growing body of research implicates mitophagy in neurode- 2015e2016 NIA intra-laboratory grant (EFF/VAB). SL and TS were
generative disease and age-related neurological decline, and pro- supported by the Velux Foundation.
vides new avenues for therapeutic intervention (Fig. 2). Although
mitochondrial dysfunction has long been implicated in the pa- References
thology of neurodegenerative diseases such as Parkinson's, Hun-
tington's, Alzheimer's, and others, decreased levels of mitophagy Bauer, J.H., et al., 2009. dSir2 and Dmp53 interact to mediate aspects of CR-
have only recently been recognized as key contributor to disease dependent life span extension in D. melanogaster. Aging 1 (1), 38.
Bingol, B., et al., 2014. The mitochondrial deubiquitinase USP30 opposes parkin-
pathogenesis. Thus, the role of mitochondrial maintenance must be mediated mitophagy. Nature 510 (7505), 370e375.
elucidated to better understand the mitochondrial phenotypes in Boily, G., et al., 2008. SirT1 regulates energy metabolism and response to caloric
neurodegeneration. Work remains to elucidate the coordination of restriction in mice. PLoS One 3 (3), e1759.
Bonkowski, M.S., Sinclair, D.A., 2016. Slowing ageing by design: the rise of NADþ
mitophagy with other mitochondria quality control pathways, such and sirtuin-activating compounds. Nat. Rev. Mol. Cell Biol. 17 (11), 679e690.
as the UPRmt. For instance, it is interesting to consider the different Bossy-Wetzel, E., Petrilli, A., Knott, A.B., 2008. Mutant huntingtin and mitochondrial
layers of mitochondrial maintenance in response to stress. In the dysfunction. Trends Neurosci. 31 (12), 609e616.
Braidy, N., et al., 2011. Age related changes in NADþ metabolism oxidative stress
context of a single mitochondrion, the UPRmt may serve as a first and Sirt1 activity in wistar rats. PLoS One 6 (4), e19194.
line of defense against damage, progressing to degradation of Bratic, A., Larsson, N.-G., 2013. The role of mitochondria in aging. J. Clin. Investig.
damaged proteins via mitochondria-derived vesicles, followed by 123 (3), 951e957.
Canto, C., et al., 2009. AMPK regulates energy expenditure by modulating NADþ
recycling of the entire organelle via mitophagy. One could even
metabolism and SIRT1 activity. Nature 458 (7241), 1056e1060.
consider mitochondrial dysfunction induced senescence (MiDAS) Canto, C., Menzies, K.J., Auwerx, J., 2015. NADþ metabolism and the control of
as the next phase, eventually leading to neurodegeneration and energy homeostasis: a balancing act between mitochondria and the nucleus.
Cell metab. 22 (1), 31e53.
other age-related pathologies. Additionally, the roles and molecular
Casley, C., et al., 2002. b-Amyloid fragment 25e35 causes mitochondrial dysfunc-
mechanisms of mitophagy in stem cell renewal and function tion in primary cortical neurons. Neurobiol. Dis. 10 (3), 258e267.
deserve further investigation. In summary, further mechanistic Chen, S., et al., 2015. RAD6 promotes homologous recombination repair by acti-
studies of mitophagy may not only improve our understanding of vating the autophagy-mediated degradation of heterochromatin protein HP1.
Mol. Cell Biol. 35 (2), 406e416.
the mechanisms of aging, neurodegeneration, and other diseases, Chen, M., et al., 2016. Mitophagy receptor FUNDC1 regulates mitochondrial dy-
but also suggest novel therapeutic strategies to combat multiple namics and mitophagy. Autophagy 12 (4), 689e702.
devastating neurodegenerative diseases, and possibly even delay Cheng, A., et al., 2012. Involvement of PGC-1a in the formation and maintenance of
neuronal dendritic spines. Nat. Commun. 3, 1250.
biological aging. Cheng, A., et al., 2016. Mitochondrial SIRT3 mediates adaptive responses of neurons
Mitophagy can be inhibited by a reduction of autophagic protein to exercise and metabolic and excitatory challenges. Cell metab. 23 (1),
activity or by depletion of metabolites such as NADþ due to 128e142.
Chow, H.-m., Herrup, K., 2015. Genomic integrity and the ageing brain. Nat. Rev.
increased DNA damage and consumption or due to a decline in Neurosci. 16 (11), 672e684.
NADþ synthesis. A reduction in mitophagy leads to an accumulation Clark, I.E., et al., 2006. Drosophila pink1 is required for mitochondrial function and
of dysfunctional mitochondria, which can induce a variety of interacts genetically with parkin. Nature 441 (7097), 1162e1166.
Cornelissen, T., et al., 2014. The deubiquitinase USP15 antagonizes Parkin-mediated
damage contributing to neurodegeneration and aging. These pa- mitochondrial ubiquitination and mitophagy. Hum. Mol. Genet. 23 (19),
thologies can be restored by targeting mitophagy through genetic 5227e5242.
editing, mitophagy-inducing compounds, and NADþ Corral-Debrinski, M., et al., 1992. Mitochondrial DNA deletions in human brain:
regional variability and increase with advanced age. Nat. Genet. 2 (4), 324e329.
supplementation.
Deng, H., et al., 2008. The Parkinson's disease genes pink1 and parkin promote
mitochondrial fission and/or inhibit fusion in Drosophila. Proc. Natl. Acad. Sci.
Acknowledgments U. S. A. 105 (38), 14503e14508.
Ding, W.X., et al., 2010. Nix is critical to two distinct phases of mitophagy, reactive
oxygen species-mediated autophagy induction and Parkin-ubiquitin-p62-
We acknowledge the valuable work of the many investigators mediated mitochondrial priming. J. Biol. Chem. 285 (36), 27879e27890.
208 E.M. Fivenson et al. / Neurochemistry International 109 (2017) 202e209

Durcan, T.M., et al., 2014. USP8 regulates mitophagy by removing K6-linked ubiq- growth, autophagy and metabolism. Nat. Cell Biol. 13 (9), 1016e1023.
uitin conjugates from parkin. EMBO J. e201489729. Mills, K.F., et al., 2016. Long-term administration of nicotinamide mononucleotide
Egan, D.F., et al., 2011. Phosphorylation of ULK1 (hATG1) by AMP-activated protein mitigates age-associated physiological decline in mice. Cell Metab. 24 (6),
kinase connects energy sensing to mitophagy. Science 331 (6016), 456e461. 795e806.
Fang, E.F., Bohr, V.A., 2016. NADþ: the convergence of DNA repair and mitophagy. Moore, A.S., Holzbaur, E.L., 2016. Dynamic recruitment and activation of ALS-
Autophagy 1e2. associated TBK1 with its target optineurin are required for efficient mitoph-
Fang, E.F., et al., 2014. Defective mitophagy in XPA via PARP-1 hyperactivation and agy. Proc. Natl. Acad. Sci. U. S. A. 113 (24). E3349-E3358.
NAD(þ)/SIRT1 reduction. Cell 157 (4), 882e896. Mouchiroud, L., et al., 2013. The NADþ/sirtuin pathway modulates longevity
Fang, E.F., et al., 2016. Nuclear DNA damage signalling to mitochondria in ageing. through activation of mitochondrial UPR and FOXO signaling. Cell 154 (2),
Nat. Rev. Mol. Cell Biol. 17 (5), 308e321. 430e441.
Fang, E.F., et al., 2016. NADþ replenishment improves lifespan and healthspan in Murakawa, T., et al., 2015. Bcl-2-like protein 13 is a mammalian Atg32 homologue
Ataxia telangiectasia models via mitophagy and DNA repair. Cell Metab. 24 (4), that mediates mitophagy and mitochondrial fragmentation. Nat. Commun. 6,
566e581. 7527.
Fayet, G., et al., 2002. Ageing muscle: clonal expansions of mitochondrial DNA point Neuspiel, M., et al., 2008. Cargo-selected transport from the mitochondria to per-
mutations and deletions cause focal impairment of mitochondrial function. oxisomes is mediated by vesicular carriers. Curr. Biol. 18 (2), 102e108.
Neuromuscul. Disord. 12 (5), 484e493. Palacino, J.J., et al., 2004. Mitochondrial dysfunction and oxidative damage in
Fimia, G.M., et al., 2007. Ambra1 regulates autophagy and development of the parkin-deficient mice. J. Biol. Chem. 279 (18), 18614e18622.
nervous system. Nature 447 (7148), 1121e1125. Palikaras, K., Tavernarakis, N., 2012. Mitophagy in neurodegeneration and aging.
Gautier, C.A., Kitada, T., Shen, J., 2008. Loss of PINK1 causes mitochondrial functional Front. Genet. 3, 297.
defects and increased sensitivity to oxidative stress. Proc. Natl. Acad. Sci. 105 Palikaras, K., Lionaki, E., Tavernarakis, N., 2015. Coordination of mitophagy and
(32), 11364e11369. mitochondrial biogenesis during ageing in C. elegans. Nature 521 (7553),
Gibson, B.A., Kraus, W.L., 2012. New insights into the molecular and cellular func- 525e528.
tions of poly (ADP-ribose) and PARPs. Nat. Rev. Mol. Cell Biol. 13 (7), 411e424. Park, J., et al., 2006. Mitochondrial dysfunction in Drosophila PINK1 mutants is
Gomes, A.P., et al., 2013. Declining NADþ induces a pseudohypoxic state disrupting complemented by parkin. Nature 441 (7097), 1157e1161.
nuclear-mitochondrial communication during aging. Cell 155 (7), 1624e1638. Park, D., et al., 2016. Resveratrol induces autophagy by directly inhibiting mTOR
Greene, J.C., et al., 2003. Mitochondrial pathology and apoptotic muscle degener- through ATP competition. Sci. Rep. 6.
ation in Drosophila parkin mutants. Proc. Natl. Acad. Sci. U. S. A. 100 (7), Perez, F.A., Palmiter, R.D., 2005. Parkin-deficient mice are not a robust model of
4078e4083. parkinsonism. Proc. Natl. Acad. Sci. U. S. A. 102 (6), 2174e2179.
Haigis, M.C., Sinclair, D.A., 2010. Mammalian sirtuins: biological insights and disease Pickrell, A.M., Youle, R.J., 2015. The roles of PINK1, parkin, and mitochondrial fidelity
relevance. Annu. Rev. Pathol. 5, 253. in Parkinson's disease. Neuron 85 (2), 257e273.
Hara, T., et al., 2006. Suppression of basal autophagy in neural cells causes neuro- Pickrell, A.M., et al., 2015. Endogenous Parkin preserves dopaminergic substantia
degenerative disease in mice. Nature 441 (7095), 885e889. nigral neurons following mitochondrial DNA mutagenic stress. Neuron 87 (2),
Hasson, S.A., et al., 2015. Chemogenomic profiling of endogenous PARK2 expression 371e381.
using a genome-edited coincidence reporter. ACS Chem. Biol. 10 (5), 1188e1197. Poole, A.C., et al., 2008. The PINK1/Parkin pathway regulates mitochondrial
Hertz, N.T., et al., 2013. A neo-substrate that amplifies catalytic activity of parkin- morphology. Proc. Natl. Acad. Sci. U. S. A. 105 (5), 1638e1643.
son’s-disease-related kinase PINK1. Cell 154 (4), 737e747. Qin, W., et al., 2006. Neuronal SIRT1 activation as a novel mechanism underlying
Houtkooper, R.H., Pirinen, E., Auwerx, J., 2012. Sirtuins as regulators of metabolism the prevention of Alzheimer disease amyloid neuropathology by calorie re-
and healthspan. Nat. Rev. Mol. Cell Biol. 13 (4), 225e238. striction. J. Biol. Chem. 281 (31), 21745e21754.
Howitz, K.T., et al., 2003. Small molecule activators of sirtuins extend Saccharo- Richter, U., et al., 2013. A mitochondrial ribosomal and RNA decay pathway blocks
myces cerevisiae lifespan. Nature 425 (6954), 191e196. cell proliferation. Curr. Biol. 23 (6), 535e541.
Hudson, E., et al., 1998. Age-associated change in mitochondrial DNA damage. Free Rojansky, R., Cha, M.-Y., Chan, D.C., 2016. Elimination of paternal mitochondria in
Radic. Res. 29 (6), 573e579. mouse embryos occurs through autophagic degradation dependent on PARKIN
Hwang, S., Disatnik, M.H., Mochly-Rosen, D., 2015. Impaired GAPDH-induced and MUL1. eLife 5, e17896.
mitophagy contributes to the pathology of Huntington's disease. EMBO Mol. Rubinsztein, D.C., Marin ~ o, G., Kroemer, G., 2011. Autophagy and aging. Cell 146 (5),
Med. 7 (10), 1307e1326. 682e695.
Imai, S.-i., Guarente, L., 2014. NADþ and sirtuins in aging and disease. Trends Cell Rugarli, E.I., Langer, T., 2012. Mitochondrial quality control: a matter of life and
Biol. 24 (8), 464e471. death for neurons. EMBO J. 31 (6), 1336e1349.
Imai, S.-i., Guarente, L., 2016. It takes two to tango: NADþ and sirtuins in aging/ Ryan, B.J., et al., 2015. Mitochondrial dysfunction and mitophagy in Parkinson's:
longevity control. npj Aging Mech. Dis. 2, 16017. from familial to sporadic disease. Trends Biochem. Sci. 40 (4), 200e210.
Jain, I.H., et al., 2016. Hypoxia as a therapy for mitochondrial disease. Science 352 Ryu, D., et al., 2016. Urolithin A induces mitophagy and prolongs lifespan in C.
(6281), 54e61. elegans and increases muscle function in rodents. Nat. Med. 22 (8), 879e888.
Kameshita, I., et al., 1984. Poly (ADP-Ribose) synthetase. Separation and identifi- Sandoval, H., et al., 2008. Essential role for Nix in autophagic maturation of
cation of three proteolytic fragments as the substrate-binding domain, the erythroid cells. Nature 454 (7201), 232e235.
DNA-binding domain, and the automodification domain. J. Biol. Chem. 259 (8), Scheibye-Knudsen, M., et al., 2013. A novel diagnostic tool reveals mitochondrial
4770e4776. pathology in human diseases and aging. Aging (Albany NY) 5 (3), 192e208.
Kanfi, Y., et al., 2012. The sirtuin SIRT6 regulates lifespan in male mice. Nature 483 Scheibye-Knudsen, M., et al., 2014. Protecting the mitochondrial powerhouse.
(7388), 218e221. Trends Cell Biol. 25 (3), 158e170.
Kerr, J.S., et al., 2017. Mitophagy and Alzheimer's disease: cellular and molecular Scheibye-Knudsen, M., et al., 2016. Cockayne syndrome group A and B proteins
mechanisms. Trends Neurosci. http://dx.doi.org/10.1016/j.tins.2017.01.002 (in converge on transcription-linked resolution of non-B DNA. Proc. Natl. Acad. Sci.
press), pii: S0166-2236(17)30005-X, PMID:28190529. U. S. A. 113 (44), 12502e12507.
Khalil, B., et al., 2015. PINK1-induced mitophagy promotes neuroprotection in Schiavi, A., et al., 2015. Iron-starvation-induced mitophagy mediates lifespan
Huntington's disease. Cell death Dis. 6 (1), e1617. extension upon mitochondrial stress in C. elegans. Curr. Biol. 25 (14),
Kitada, T., et al., 1998. Mutations in the parkin gene cause autosomal recessive ju- 1810e1822.
venile parkinsonism. Nature 392 (6676), 605e608. Song, Y.M., et al., 2015. Metformin alleviates hepatosteatosis by restoring SIRT1-
Komatsu, M., et al., 2006. Loss of autophagy in the central nervous system causes mediated autophagy induction via an AMP-activated protein kinase-
neurodegeneration in mice. Nature 441 (7095), 880e884. independent pathway. Autophagy 11 (1), 46e59.
Larsson, N.-G., 2010. Somatic mitochondrial DNA mutations in mammalian aging. Song, W.H., et al., 2016. Autophagy and ubiquitin-proteasome system contribute to
Annu. Rev. Biochem. 79, 683e706. sperm mitophagy after mammalian fertilization. Proc. Natl. Acad. Sci. U. S. A. 113
Lazarou, M., et al., 2015. The ubiquitin kinase PINK1 recruits autophagy receptors to (36). E5261-E5270.
induce mitophagy. Nature 524 (7565), 309e314. Soubannier, V., et al., 2012. A vesicular transport pathway shuttles cargo from
Lemasters, J.J., 2005. Selective mitochondrial autophagy, or mitophagy, as a targeted mitochondria to lysosomes. Curr. Biol. 22 (2), 135e141.
defense against oxidative stress, mitochondrial dysfunction, and aging. Reju- Strappazzon, F., et al., 2015. AMBRA1 is able to induce mitophagy via LC3 binding,
venation Res. 8 (1), 3e5. regardless of PARKIN and p62/SQSTM1. Cell Death Differ. 22 (3), 517.
Liu, L., et al., 2012. Mitochondrial outer-membrane protein FUNDC1 mediates Sugiura, A., et al., 2014. A new pathway for mitochondrial quality control:
hypoxia-induced mitophagy in mammalian cells. Nat. Cell Biol. 14 (2), 177e185. mitochondrial-derived vesicles. EMBO J. e201488104.
pez-Otín, C., et al., 2013. The hallmarks of aging. Cell 153 (6), 1194e1217.
Lo Sun, N., et al., 2015. Measuring in vivo mitophagy. Mol. Cell 60 (4), 685e696.
Madeo, F., Tavernarakis, N., Kroemer, G., 2010. Can autophagy promote longevity? Sun, N., Youle, R.J., Finkel, T., 2016. The mitochondrial basis of aging. Mol. Cell 61 (5),
Nat. Cell Biol. 12 (9), 842e846. 654e666.
Massudi, H., et al., 2012. Age-associated changes in oxidative stress and NADþ Tissenbaum, H.A., Guarente, L., 2001. Increased dosage of a sir-2 gene extends
metabolism in human tissue. PLoS One 7 (7), e42357. lifespan in Caenorhabditis elegans. Nature 410 (6825), 227e230.
McLelland, G.L., et al., 2014. Parkin and PINK1 function in a vesicular trafficking Trifunovic, A., et al., 2004. Premature ageing in mice expressing defective mito-
pathway regulating mitochondrial quality control. EMBO J. e201385902. chondrial DNA polymerase. Nature 429 (6990), 417e423.
Menzies, F.M., Fleming, A., Rubinsztein, D.C., 2015. Compromised autophagy and Valente, E.M., et al., 2004. Hereditary early-onset Parkinson's disease caused by
neurodegenerative diseases. Nat. Rev. Neurosci. 16 (6), 345e357. mutations in PINK1. Science 304 (5674), 1158e1160.
Mihaylova, M.M., Shaw, R.J., 2011. The AMPK signalling pathway coordinates cell Van Humbeeck, C., et al., 2011. Parkin interacts with Ambra1 to induce mitophagy.
E.M. Fivenson et al. / Neurochemistry International 109 (2017) 202e209 209

J. Neurosci. 31 (28), 10249e10261. neuron degeneration caused by inactivation of Drosophila Pink1 is rescued by
Wallace, D.C., 1999. Mitochondrial diseases in man and mouse. Science 283 (5407), Parkin. Proc. Natl. Acad. Sci. U. S. A. 103 (28), 10793e10798.
1482e1488. Yang, Y., et al., 2008. Pink1 regulates mitochondrial dynamics through interaction
Wallace, D.C., 2013. A mitochondrial bioenergetic etiology of disease. J. Clin. with the fission/fusion machinery. Proc. Natl. Acad. Sci. U. S. A. 105 (19),
Investig. 123 (4), 1405e1412. 7070e7075.
Weydt, P., et al., 2006. Thermoregulatory and metabolic defects in Huntington's Ye, X., et al., 2015. Parkin-mediated mitophagy in mutant hAPP neurons and Alz-
disease transgenic mice implicate PGC-1a in Huntington's disease neuro- heimer's disease patient brains. Hum. Mol. Genet. 24 (10), 2938e2951.
degeneration. Cell metab. 4 (5), 349e362. Youle, R.J., Narendra, D.P., 2011. Mechanisms of mitophagy. Nat. Rev. Mol. Cell Biol.
Wiley, C.D., et al., 2016. Mitochondrial dysfunction induces senescence with a 12 (1), 9e14.
distinct secretory phenotype. Cell metab. 23 (2), 303e314. Yun, J., et al., 2014. MUL1 acts in parallel to the PINK1/parkin pathway in regulating
Wu, W., et al., 2014. ULK1 translocates to mitochondria and phosphorylates mitofusin and compensates for loss of PINK1/parkin. Elife 3, e01958.
FUNDC1 to regulate mitophagy. EMBO Rep. 15 (5), 566e575. Zhou, Q., et al., 2016. Mitochondrial endonuclease G mediates breakdown of
Yang, Y., et al., 2006. Mitochondrial pathology and muscle and dopaminergic paternal mitochondria upon fertilization. Science 353 (6297), 394e399.

You might also like