You are on page 1of 13

The Role of Autophagy in Parkinson’s Disease

Melinda A. Lynch-Day, Kai Mao, Ke Wang, Mantong Zhao, and Daniel J. Klionsky
University of Michigan, Life Sciences Institute, Ann Arbor, Michigan 48109
Correspondence: klionsky@umich.edu

Great progress has been made toward understanding the pathogenesis of Parkinson’s disease
(PD) during the past two decades, mainly as a consequence of the discovery of specific gene
mutations contributing to the onset of PD. Recently, dysregulation of the autophagy pathway
has been observed in the brains of PD patients and in animal models of PD, indicating the
emerging role of autophagy in this disease. Indeed, autophagy is increasingly implicated
in a number of pathophysiologies, including various neurodegenerative diseases. This
article will lead you through the connection between autophagy and PD by introducing
the concept and physiological function of autophagy, and the proteins related to autosomal
dominant and autosomal recessive PD, particularly a-synuclein and PINK1-PARKIN, as they
pertain to autophagy.
www.perspectivesinmedicine.org

here seem to be various causes of Parkin- AUTOPHAGY


T son’s disease (PD), yet the pathogenesis of
this disease appears to be converging on com- Main Types of Autophagy
mon themes—oxidative stress, mitochondrial Autophagy is an evolutionarily conserved ca-
dysfunction, and protein aggregation—all of tabolic process that mediates the degradation
which are tightly linked to autophagy, a highly of long-lived proteins and dysfunctional or
conserved cellular homeostatic process essential superfluous organelles in eukaryotic cells. Au-
for bulk degradation of cytoplasmic contents. tophagy is induced by various adverse condi-
In particular, the recent identification of auto- tions such as limited nutrients, low oxygen lev-
somal dominant and autosomal recessive muta- els, and decreased energy supply, and its action
tions in familial PD has revealed the involve- results in the release of degradation products,
ment of the corresponding gene products in especially amino acids, back into the cytoplasm
autophagy. Although autophagy has commonly to be used in essential biosynthetic pathways.
been regarded as an adaptive response to nu- According to the different pathways by
trient deprivation, increasing evidence indicates which cargo is delivered to the lysosome or va-
that basal, constitutive autophagy is essential cuole, autophagy can be divided into three
for neuronal survival and that its dysregulation main types (Fig. 1): macroautophagy, microau-
leads to neurodegeneration. tophagy, and chaperone-mediated autophagy

Editor: Serge Przedborski


Additional Perspectives on Parkinson’s Disease available at www.perspectivesinmedicine.org
Copyright # 2012 Cold Spring Harbor Laboratory Press; all rights reserved; doi: 10.1101/cshperspect.a009357
Cite this article as Cold Spring Harb Perspect Med 2012;2:a009357

1
M.A. Lynch-Day et al.

Peroxisome

Microautophagy
Lysosome (micropexophagy)

Autophagosome
Chaperone-mediated
autophagy

hsc70
LAMP-2A
Unfolded
substrate protein
Macroautophagy
Permease

Lysosomal
hydrolase
Phagophore
Mitochondrion
Autolysosome

Figure 1. Schematic model of the three main types of autophagy. The modes of autophagy differ depending on
the nature of the substrate and the site of sequestration. In chaperone-mediated autophagy, the substrates con-
tain a KFERQ-consensus motif, are unfolded by HSC70 chaperones, and translocate directly across the lysosome
membrane via interaction with a LAMP-2A oligomer. There are various types of microautophagy-like processes
including micropexophagy and micromitophagy, the selective degradation of peroxisomes and mitochondria,
respectively. Again, sequestration occurs at the lysosome-limiting membrane, but the substrates do not have to
www.perspectivesinmedicine.org

be unfolded. Macroautophagy uses a double-membrane phagophore to sequester the cargo. Essentially any cy-
toplasmic component can be enwrapped by a phagophore, which expands into an autophagosome. Fusion with
the lysosome allows the cargo to be degraded, and the resulting macromolecules are released into the cytosol
through permeases, allowing them to be reused for anabolic processes.

(CMA). CMA involves direct translocation of lases access to the inner autophagosome mem-
unfolded proteins across the lysosome mem- brane and its cargo, which is degraded and re-
brane. Chaperone proteins mediate this process cycled. In contrast to the ubiquitin-26S pro-
by binding to cytosolic substrates that enter the teasome system, macroautophagy can mediate
lysosome through interaction with a receptor/ nonselective and bulk degradation of cytoplas-
channel on the lysosomal membrane (Majeski mic contents, including entire organelles (Shin-
and Dice 2004). Microautophagy describes the tani et al. 2002; Kanki et al. 2009). Among the
process of direct uptake of cytoplasmic materials three main types of autophagy, macroautophagy
at the lysosome surface by invagination of the is the best characterized process and will here-
lysosome membrane. After vesicles containing after be referred to as autophagy.
the cytosolic substrates pinch off into the lyso-
somal lumen, they are rapidly degraded (Kunz
Selective Autophagy
et al. 2004). In contrast, during macroautophagy,
portions of the cytoplasm are engulfed by a dou- In some cases, autophagy displays substrate
ble-membrane phagophore that expands into a specificity, even though autophagy is often con-
cytosolic vesicle called an autophagosome; the sidered to be a nonselective pathway for the
completed autophagosome is targeted to the degradation of bulk cytoplasmic components.
lysosome in mammalian cells or the vacuole in Indeed, the unique feature of the autophagy
yeast (Klionsky 2005). The outer membrane of process where the initial sequestering compart-
the autophagosome subsequently fuses with the ment expands into an autophagosome allows
lysosomal/vacuolar membrane, allowing hydro- for flexible cargo selection. For example, in the

2 Cite this article as Cold Spring Harb Perspect Med 2012;2:a009357


Autophagy and Parkinson’s Disease

cytoplasm to vacuole targeting (Cvt) pathway, regulating the localization of other Atg proteins
autophagy fulfills a biosynthetic role by deliver- such as Atg9, an integral membrane protein that
ing three vacuolar hydrolases, a-mannosidase cycles back and forth between the site of phag-
(Ams1) (Hutchins and Klionsky 2001), amino- ophore nucleation/autophagosome formation,
peptidase I (Ape1) (Klionsky et al. 1992), and as- termed the phagophore assembly site (PAS),
partyl aminopeptidase (Ape4) (Yuga et al. 2011) and other peripheral locations (Reggiori et al.
to their final destination, the vacuole. In ad- 2004). Due to its subcellular itinerary and its
dition, superfluous or damaged organelles and characterization as a membrane protein, Atg9
misfolded or aggregated proteins are selectively is thought to be responsible for the transport
targeted for degradation by autophagy. Differ- of donor membrane that contributes to auto-
ent terms are used depending on the cargo, for phagosome formation.
example, “mitophagy” for selective autophagic The main product of the ubiquitin-like con-
degradation of mitochondria, “pexophagy” for jugation systems is the covalent attachment of
peroxisomes, “reticulophagy” for the endoplas- Atg8 to phosphatidylethanolamine (PE) (Geng
mic reticulum (ER), and “ribophagy” for ribo- and Klionsky 2008). Atg8 – PE becomes associ-
somes (Klionsky et al. 2007). Among them, ated with both the inner and outer membrane
mitophagy has been increasingly implicated in of the phagophore. Once the autophagosome
the pathogenesis of PD through the PINK1- is complete, Atg8 is cleaved off of PE from the
PARKIN-mediated pathway. outer membrane, whereas the Atg8 on the inner
membrane remains associated with PE and is
degraded in the vacuole. Atg8 controls the size
Molecular Mechanisms of Autophagy
of the forming autophagosome (Xie et al. 2008),
The molecular understanding of autophagy was and is also involved in cargo recognition during
www.perspectivesinmedicine.org

initiated by pioneering work in yeast utilizing selective autophagy (Okamoto et al. 2009; Shin-
genetic screens that led to the discovery of au- tani et al. 2002).
tophagy-related (ATG) genes, followed by the
identification of homologs in higher eukary-
Physiological Functions and
otes. The corresponding Atg proteins can be
Connections to PD
divided into four major groups: the Atg1/unc-
51-like kinase (ULK) complex, two ubiquitin- Although autophagy is primarily a starvation re-
like protein (Atg12 and Atg8/LC3) conjuga- sponse in yeast, in higher eukaryotic organisms,
tion systems, the class III phosphatidylinositol autophagy is involved in a wide range of phy-
3-kinase (PtdIns3K)/Vps34 complex I, and the siological and pathological processes, includ-
Atg9/mATG9 transmembrane protein system ing responses to nutrient deprivation, develop-
(Xie and Klionsky 2007). The target of rapa- ment, intracellular clearance, suppression of
mycin (TOR), a master regulator of nutrient tumor formation, aging, cell death and survival,
and growth factor signaling, is one of the critical and immunity (Huang and Klionsky 2007). As a
components involved in controlling the induc- primary protective mechanism that maintains
tion of autophagy (He and Klionsky 2009). In nutrient and energy homeostasis in response to
most cell types, TOR activity is necessary and stress, dysregulation of autophagy underlies the
sufficient to suppress autophagy under favor- pathophysiologies of many diseases. Increasing
able growth conditions, primarily in response evidence suggests that dysregulation of auto-
to nitrogen. Other kinases, including protein phagy results in the accumulation of abnormal
kinase A, AMPK/Snf1, and Pho85, modulate proteins and/or damaged organelles, which is
autophagy in response to various types of stress. commonly observed in neurodegenerative di-
Both the Atg1/ULK complex and the mem- seases, such as Alzheimer, Huntington’s, and
brane protein Atg9 function early in the process Parkinson’s diseases (Banerjee et al. 2010). Of
of autophagosome formation. Atg1 is a target of note, autophagy is the only known mechanism
the Tor signaling pathway and acts in part by that eukaryotic cells possess to degrade protein

Cite this article as Cold Spring Harb Perspect Med 2012;2:a009357 3


M.A. Lynch-Day et al.

aggregates and damaged organelles that cannot be This CMA receptor with the aid of a lysosomal
processed by the proteasome. Recent studies from lumenal HSC70 transports a-synuclein into
transgenic mice, animal, and cell models of PD the lysosome where it is degraded by proteases.
suggest the involvement of proteins genetically Mutant forms of the protein prevent its degra-
linked to autosomal dominant PD, particularly dation by the CMA pathway resulting in toxic
a-synuclein and LRRK2, in the autophagy path- aggregation in the cytoplasm as was seen in
way (Bandyopadhyay and Cuervo 2007; Alegre- cell culture and postmortem tissues (Martinez-
Abarrategui and Wade-Martins 2009). In addi- Vicente et al. 2008). Autophagy can partially
tion, proteins related to recessive PD, such as compensate for the lack of CMA-mediated deg-
PINK1 and PARKIN, have an important role in radation, but may cause autophagic cell death
the process of mitophagy. under stress conditions. Cells expressing mutant
a-synuclein can be characterized by an increase
EARLY DISCOVERIES: a-SYNUCLEIN in cell death, accumulation of autophagosomes,
AND AUTOPHAGY and a loss of ability to store catecholamine along
a-Synuclein and PD with a failure to release dopamine (Stefanis et al.
2001).
a-Synuclein was found to localize to the presy- All mutant forms of a-synuclein vary in the
naptic terminals in the central nervous system degree to which they hamper the lysosomal/
and is involved in vesicular release (Clayton CMA degradation pathway and thus have differ-
and George 1998, 1999; Spencer et al. 2009). It ent levels of toxicity. In cell culture studies, the
is a natively unfolded protein, but can be found A53T and A30P mutants of the protein bind
in several aberrant conformational states in- more strongly to the LAMP-2A receptor than the
cluding an oligomer, a protofibril, and an amy- wild-type form, but fail to be transported across
www.perspectivesinmedicine.org

loid fibril (Martinez-Vicente et al. 2008). a-Syn- the lysosomal membrane (Martinez-Vicente et
uclein was identified as a component of Lewy al. 2008). The mutants act as receptor inhibitors,
bodies, cytosolic inclusions that are a patholog- preventing other CMA targets from binding.
ical trait of PD (Spillantini et al. 1997, 1998). This leads to a complete block in CMA resulting
Studies of familial cases of autophagy reveal in a higher degree of toxicity. Overexpression of
two separate autosomal dominant mutations the wild-type protein is matched by an increase
in the a-synuclein gene: A53T and A30P (Poly- in expression of the CMA receptor protein
meropoulos et al. 1997). In addition to the LAMP-2A, but high levels of protein expression
point mutations, several posttranslational mod- lead to the formation of oligomeric forms that
ifications such as phosphorylation, ubiquitina- cannot be degraded by CMA (Vogiatzi et al.
tion, nitration, oxidation, and dopamine-de- 2008; Xilouri et al. 2009). This results in a toxicity
pendent adduct formation also create toxic level that is lower than the familial point mutant
forms of the protein (Martinez-Vicente et al. forms. Similar intermediate levels of toxicity are
2008). seen in certain posttranslationally modified ver-
sions of a-synuclein as was illustrated in cell and
Chaperone-Mediated Autophagy
post-mortem tissues. Phosphorylated, ubiquiti-
a-Synuclein in its native form is degraded by nated, nitrated, and oxidized forms are less sus-
chaperone-mediated autophagy (Cuervo et al. ceptible to CMA degradation than the nonmodi-
2004). The protein contains a 15 amino acid fied protein, but they do not block CMA in its
sequence that consists of imperfect yet overlap- entirety. It is thought that these modifications
ping variations of the KFERQ CMA recogni- promote higher-order oligomers that cannot be
tion motif. The chaperone protein HSC70 rec- broken down and degraded. This is reflected in
ognizes the pentapeptide sequence motif and the observation of all of these modifications in cy-
binds to a-synuclein. a-Synuclein then binds tosolic aggregates.
to the lysosomal-associated membrane protein Modification of the protein with dopamine
type 2A (LAMP-2A) at the lysosomal membrane. gives a phenotype that more closely resembles

4 Cite this article as Cold Spring Harb Perspect Med 2012;2:a009357


Autophagy and Parkinson’s Disease

the point mutations. a-Synulcein can be modi- a-Synuclein and Autophagy


fied with oxidized dopamine through a nonco-
valent interaction. Dopamine a-synuclein (DA- As noted previously, inhibition of CMA by
a-syn) inhibits not only its own degradation, aberrant a-synuclein leads to an increase in au-
but it also blocks CMA activity in general. This tophagy. This appears to be a compensatory re-
defect can be seen in a variety of experimental sponse, but rather than leading to cell survival,
conditions including isolated lysosomes, dopa- the induction of autophagy can be detrimental
minergic cell lines, and visceral motor neurons. causing autophagic cell death. Blocking autoph-
The CMA defect results from DA-a-syn expres- agy by knocking down the autophagy protein
sion because it is not seen in a-synuclein dele- Atg5 in cells expressing the A53T a-synuclein
tion cells, it is not observed with the dopamine mutant can rescue the cell from toxicity-in-
insensitive form of a-synuclein, and it is repro- duced cell death (Xilouri et al. 2009). However,
duced with isolated lysosomes when presented autophagy-induced neuronal death is not al-
with DA-a-syn. The complete blockage of CMA ways the outcome. One study suggests that the
creates a high level of toxicity (Martinez-Vicente signaling pathway for activation of autophagy
et al. 2008). What is interesting about the DA-a- may be important as to whether or not autoph-
syn modification is that it is the dopaminergic agy will be protective or detrimental. Autophagy
substantia nigra and the norepinephrine-releasing is mainly initiated through the mTOR signal-
locus coeruleus neurons that are killed first in the ing pathway either directly or indirectly through
progression of PD. Both of these types of neurons the autophagy protein Atg1. An additional sig-
contain cytosolic dopamine and produce neuro- naling pathway for initiation of autophagy is
melanin (a product of dopamine modifications). the Vps34-Beclin 1 complex. It is this secondary
The toxic DA-a-syn form of the protein may ex- signaling pathway that appears to promote cell
www.perspectivesinmedicine.org

plain why those two types of neurons are particu- survival. For example, a reduction in a-syn-
larly sensitive. uclein accumulation is seen when Beclin 1 is
The blockage of CMA activity with mutant overexpressed. In addition, Beclin 1 overexpres-
forms of a-synuclein not only results in the di- sion decreases cell death and increases autoph-
rect buildup of toxicity in the neuron through agy activity observed through enhanced lysoso-
the formation of aggregates, but it also prevents mal degradation (Spencer et al. 2009).
the protective activity of the protein myocyte en- Not only does aberrant a-synuclein inhibit
hancer factor 2D (MEF2D). MEF2D, atranscrip- CMA, but it also inhibits autophagy through
tion factor, is an important player in neuronal RAB1A and omegasome formation as seen in
survival. Patients with PD show an increase of both cell and mouse models (Winslow et al.
this protein in brain neurons, and a genetic poly- 2010). RAB1A is a GTPase involved in the early
morphism of a related protein (MEF2A) has secretory pathway, specifically ER-to-Golgi trans-
been linked to Alzheimer disease. CMA-depen- port. The early secretory pathway is important
dent degradation regulates MEF2D activity. for autophagy, and inhibition of the secretory
MEF2D is continuously shuttled to the cytosol pathway blocks autophagy. Furthermore, RAB
from the nucleus where it interacts with hsc70. proteins can play a role in autophagy indepen-
In cells, when CMA is inhibited, an inactive dent of the secretory pathway, as seen with Ypt1
form of the protein accumulates in the cytosol in yeast (Lynch-Day et al. 2010) and RAB1A in
and the amount of protein in the nucleus drops. mammalian cells (Huang et al. 2011). a-Syn-
This inactive form can no longer bind DNA. uclein overexpression blocks autophagosome for-
Wild-type and mutant forms of a-synuclein mation, inhibits secretion, and increases Golgi
prevent binding between HSC70 and MEF2D fragmentation. Overexpression of RAB1A rescues
(Yangetal.2009).Thissuggeststhatnotonlydoes this defect. The block in autophagy due to a-syn-
a-synuclein promote neuronal death through uclein overexpression occurs early in the pathway,
the formation of aggregates, but it also promotes before autophagosome formation, suggesting an
cell death by inhibiting cell survival proteins. effect on ATG9, which is the only transmembrane

Cite this article as Cold Spring Harb Perspect Med 2012;2:a009357 5


M.A. Lynch-Day et al.

protein required for autophagy. It is thought that RECENT STUDIES: CONTROVERSIES


ATG9 is responsible for the transport of mem- ABOUND
brane to the site of autophagosome formation
In this section, we will discuss new yet contro-
and thus acts early in the process. ATG9 normally
versial areas of research with regard to PD and
forms puncta at a perinuclear location (the site of
autophagy. Recent studies have focused on the
autophagosome formation) and at the trans-
hypothesis of mitochondrial dysfunction as a
Golgi network in mammalian cells. When a-syn-
cause of the disease. These studies have resulted
uclein is overexpressed, ATG9 is mislocalized and
in some interesting data, but to date there is no
is diffuse throughout the cytoplasm of the cell.
clear indication as to whether mitochondrial
The same phenotype is seen with the knock-
dysfunction is a cause of Parkinson or is rather
down of RAB1A. One preautophagosome struc-
correlated with the progression of the disease.
ture that branches off of the ER is the omegasome,
which generates an autophagosome, at least un-
der some circumstances. Omegasome formation
The Hypothesis: Mitochondria
is reduced in cells that overexpress a-synuclein
Dysfunction in PD
and in cells that have reduced RAB1A protein
levels. a-Synuclein blocks autophagy by inhibit- Mitochondria are essential organelles that pro-
ing the activity of RAB1A, which results in the vide .90% of the energy in all eukaryotic cells
mislocalization of ATG9 and inhibition of auto- through oxidative phosphorylation (McBride
phagosome formation (Winslow et al. 2010). et al. 2006). Mitochondria are also involved in
various other processes such as calcium ho-
meostasis (Celsi et al. 2009) and regulation of
a-Synuclein and Mitophagy
apoptosis (Keeble and Gilmore 2007). However,
www.perspectivesinmedicine.org

More recent studies of a-synuclein in PD have mitochondria are also the major source of cellu-
shown a relationship between its aberrant ex- lar reactive oxygen species (ROS). Normal levels
pression and mitophagy. Mitochondrial dys- of ROS can be tolerated because of cellular anti-
function is another characteristic of PD and will oxidants, whereas in pathological situations of
be described in more detail later in this article. mitochondrial respiratory defect, dramatic ROS
However, a connection has been made between production exceeds the capability of antioxidant
the activation of autophagy by aberrant a-syn- protection and causes severe damage to a wide
uclein expression and mitochondrial dysfunc- range of cellular components including mito-
tion. In cells expressing the A53T a-synuclein chondria. Accumulation of this damage is re-
mutant, there is an observed increase in colocal- lated to aging, cancer, and recently to neurode-
ization between autophagosomes and normal, generative diseases such as PD (Wallace 2005).
polarized mitochondria. In addition, there is a PD is characterized primarily by the selective
decrease in the number and length of mitochon- loss of dopaminergic neurons in the substantia
dria in these cells. Similar results are seen when nigra pars compacta leading to a dopamine def-
wild-type a-synuclein is overexpressed; how- icit in the striatum. Recent evidence suggests
ever, the phenotype is not as severe. The increase that mitochondria dysfunction may play a role
in mitochondria clearance in these cell lines is in the pathogenesis of both sporadic PD and fa-
dependent on mitochondrial fragmentation and milial Parkinsonism. One current model sug-
on the protein PARKIN (Choubey et al. 2011). gests that mitochondrial dysfunction results
PARKIN, currently another large area of focus from damage to complex I of the mitochon-
for autophagy and PD research, will be discussed drial electron transport chain (Schuler and
in the next section of this article. However, the Casida 2001). Indeed, some studies have shown
role of a-synuclein in the promotion of mito- complex I activities to be significantly reduced
phagy of polarized mitochondria suggests that in post-mortem substantia nigra of PD patients
there may be a connection between it and PAR- (Schapira et al. 1989; Schapira 1993). There are
KIN in the promotion of the disease. several lines of evidence that suggest that in-

6 Cite this article as Cold Spring Harb Perspect Med 2012;2:a009357


Autophagy and Parkinson’s Disease

creased oxidative damage and ATP depletion may functions as the receptor for the sequestration of
cause dopaminergic neuronal cell death, but the mitochondria into an autophagosome (Kanki
hypothesis linking PD and complex I deficiency et al. 2009; Okamoto et al. 2009). During mi-
is still highly debatable, and the question of cau- tophagy, Atg32 is recognized by an adaptor pro-
sation versus correlation remains to be answered. tein, Atg11, which is proposed to play a role in
Familial variants of PD account for up to mediating cargo recognition and transport to
10% of all cases (Gasser 2009). In familial PD, the phagophore assembly site (PAS), the nucleat-
several genes have been linked to autosomal re- ing structure for generation of the phagophore
cessive (PARK2, PARK6, PARK7) or dominant (Yorimitsu and Klionsky 2005). Mitochondrial
(LRRK2) Parkinson (Hardy et al. 2009). These fragments containing Atg32 are then enwrapped
genes have been linked to mitochondrial func- by the expanding phagophore, ultimately being
tion and several very recent studies have demon- incorporated into an autophagosome. The de-
strated that the corresponding gene products tailed mechanism of this process is still under
are involved in the selective removal of damaged study.
mitochondria through autophagy (Narendra et In higher eukaryotes, autophagy also plays a
al. 2008, 2010b). Thus, these proteins may pro- critical role in degrading mitochondria. In fact,
vide a link between mitophagy and PD. mitochondria were first detected inside an auto-
phagosome in the 1950s (Eskelinen et al. 2011);
however, a molecular understanding of this pro-
Mitophagy: Autophagic Mitochondrial
cess is occurring only now. Studies suggest that
Removal
the selective removal of mitochondria, espe-
As discussed above, autophagy can be highly cially damaged mitochondria, is part of an
specific. During autophagy, the phagophore important homeostatic pathway for organelle
www.perspectivesinmedicine.org

gradually expands and engulfs a portion of the quality control. Since mitochondria function
cytoplasm, or specific cargos, to form the dou- is compromised in some PD models, a defect
ble-membrane autophagosome (Nair and Kli- in mitochondria quality control may play a crit-
onsky 2005). The diameter of a typical autopha- ical role in the pathogenesis of PD.
gosome is approximately 500 nm (Xie et al.
2008); however, the mechanism of autophago-
Mitophagy and PD
some formation, involving the sequential ex-
pansion of the phagophore, provides autophagy As mentioned above, several genes related to PD
with the capacity to sequester essentially any cel- have been recently reported to participate in
lular components, including entire organelles, the removal of damaged mitochondria through
and deliver them into the lysosome for degra- autophagy. The PARK2 gene has been reported
dation. to be mutated in nearly 50% of autosomal reces-
Pioneering studies in yeast have demon- sive, and 10%– 15% of sporadic early-onset PD.
strated that autophagic degradation of mito- PARKIN, the gene product of PARK2 is a pri-
chondria, mitophagy, can be a highly selective marily cytosolic ubiquitin E3 ligase that con-
and tightly regulated process (Kanki et al. 2009; tains a ubiquitin-like domain, two RING finger
Mao et al. 2011). In yeast cells, mitophagy fits domains, and a conserved region between the
with the common model of a receptor-adaptor RING domains (Schapira 2008). PARKIN has
system for the selective degradation of a specific been previously reported to function in the
cargo by autophagy; a tag on the cargo is recog- cytosol, in the ER, on mitochondrial targets,
nized by a receptor and/or adapter, which links and at the plasma membrane; however, no clear
the cargo with the autophagy machinery via in- evidence had linked PARKIN function to the
teraction with Atg8 (Shintani et al. 2002; Wang pathogenesis of PD. Recent studies from Richard
and Klionsky 2011). In the case of mitophagy, Youle’s group and others, however, have pro-
yeast genetic screens discovered a mitochondrial vided a model for PARKIN’s role in eliminating
outer membrane resident protein, Atg32, which impaired mitochondria (Narendra et al. 2008):

Cite this article as Cold Spring Harb Perspect Med 2012;2:a009357 7


M.A. Lynch-Day et al.

PARKIN is specifically recruited to damaged mi- 2010; Ziviani et al. 2010). After translocation to
tochondria and promotes their autophagic deg- mitochondria, PARKIN ubquitinates MFN1/2
radation (Gasser 2009). At steady state, PARKIN causing their degradation, which facilitates mi-
is cytosolic. However, treatment of PARKIN- tochondrial fission; normal fission may be neces-
overexpessing cells with the mitochondrial un- sary for efficient mitophagy. However, if MFN1/2
coupler carbonyl cyanide m-chlorophenylhy- are the only substrates of PARKIN, the latter
drazone (CCCP) leads to a rapid and significant might play a role in facilitating, but not activating
relocation of PARKIN to mitochondria, fol- mitophagy; it is thought that mitochondrial fis-
lowed by substantial mitochondria loss from sion is required, but not sufficient to initiate mi-
the treated cells. The loss of mitochondria is de- tophagy. Therefore, to determine the real role of
pendent on the expression of PARKIN and the PARKIN in mitophagy, some other specific sub-
presence of autophagic proteins, demonstrating strates of PARKIN, if they exist, have to be iden-
that degradation of mitochondria is through tified. Along these lines, a recent study from
autophagy. Extensive mitochondria fragmenta- David Chan’s group suggests that PARKIN acti-
tion is observed following CCCP treatment, in vates the ubiquitin-proteasome system, which re-
a PARKIN-independent manner. Microscopy sults in the ubiquitination of a large number of
studies show that PARKIN is selectively re- mitochondrial proteins (Chan et al. 2011).
cruited to mitochondria fragments that have
decreased or no membrane potential, suggesting
The Role of p62 in PARKIN-Dependent
a role for PARKIN in distinguishing between
Mitophagy
healthy and damaged mitochondria. Further
observation shows that these PARKIN-marked p62 connects ubiquitinated proteins to LC3 for
mitochondrial fragments are LC3 (a mamma- autophagic degradation (Pankiv et al. 2007). As
www.perspectivesinmedicine.org

lian homolog of yeast Atg8) positive, further accumulation of p62 is strikingly elevated when
demonstrating that clearance of damaged mi- autophagy is blocked, it is widely used as an
tochondria occurs through autophagy. Over- autophagy marker. The loss of mitochondri-
expressed PARKIN is also recruited to mito- al membrane potential promotes the accumu-
chondria upon an increase in complex one- lation of p62 on clustered mitochondria in a
dependent ROS, which follows treatment with PARKIN-dependent manner. Whether p62 is re-
the herbicide paraquat, a toxin frequently used quired for mitophagy, however, is controversial
to induce a PD phenotype in some animal and and further studies are needed to determine its
cell culture models (Terzioglu and Galter 2008). role (Geisler et al. 2010; Narendra et al. 2010a).

Mitochondrial Targets of PARKIN PINK1


The translocation of PARKIN to mitochondria PARKIN interacts with another PD-related pro-
is an indispensable step in PARKIN-dependent tein, PTEN-induced kinase 1 (PINK1), a mito-
mitophagy. Therefore, the identification of mi- chondrial membrane-anchored kinase. In Dro-
tochondrial targets of PARKIN is significant for sophila melanogaster, the phenotype resulting
elucidating the underlying mechanism of this from the loss of PINK1 is rescued on overexpres-
cellular activity. Although the mitochondrial sion of PARKIN; however, loss of PARKIN is not
voltage-dependent anion channel 1 (VDAC1) rescued by the overexpression of PINK1 (Clark
was reported to be ubquitinated by PARKIN in et al. 2006; Yang et al. 2006), suggesting that
HeLa cells, this ubquitination does not seem to PINK1 acts upstream of PARKIN. Subsequent
be required for mitochondrial clustering or mi- studies show that PINK1 plays a role in the re-
tophagy (Geisler et al. 2010; Narendra et al. cruitment of PARKIN (Geisler et al. 2010; Mat-
2010a,b). The other putative mitochondrial tar- suda et al. 2010; Narendra et al. 2010b; Vives-
gets of PARKIN include the mitochondrial fu- Bauza et al. 2010). Expression of PINK1 on in-
sion proteins MFN1 and MFN2 (Gegg et al. dividual mitochondria is regulated by voltage-

8 Cite this article as Cold Spring Harb Perspect Med 2012;2:a009357


Autophagy and Parkinson’s Disease

dependent proteolysis; thus, low levels of PINK1 length PINK1 is the only functional form. Full-
are maintained on healthy, polarized mitochon- length PINK1 is rapidly degraded in normal con-
dria. In steady-state cells, PINK1 is imported ditions, but accumulates in dysfunctional mito-
into the mitochondrial inner membrane in a chondriato activate mitophagy when mitochon-
membrane potential-dependent manner. When dria lose their membrane potential (Matsuda
imported into the inner membrane, the mi- et al. 2010; Narendra et al. 2010a,b). These latter
tochondrial inner membrane rhomboid prote- results imply that truncated PINK1 is an inter-
ase presenilin-associated rhomboidlike protein mediate product destined for degradation. As
(PARL) mediates the cleavage of PINK1 (Jin et different isoforms of PINK1 are related to differ-
al. 2010). Upon mitochondria depolarization, ent cellular locations and functions and might
PINK1 import into the inner membrane is im- respond to different stresses, further studies are
paired, leading to a rapid PINK1 accumulation still needed to elucidate this issue.
on the outer membrane of damaged mitochon-
dria. PINK1 accumulation on mitochondria is
both necessary and sufficient for PARKIN re- Protective Function of PINK1 in Different
cruitment to mitochondria. How recruited Animal Models
PARKIN on damaged mitochondria can pro- Although the significant role of PINK1 in neu-
mote their degradation is still under extensive ron protection is clear, an apparent difference
investigation. An intriguing possibility is that of displayed phenotypes is observed between fly
PARKIN may mediate the ubiquitination of cer- and mouse models when PINK1 is depleted. In
tain substrates on mitochondria, and the ubiq- Pink1 mutant Drosophila, the obvious pheno-
uitinated substrates may serve as a recognition types, including loss of dopaminergic neurons,
target for p62/SQSTM1, a ubiquitin-binding
www.perspectivesinmedicine.org

reduced life span, mitochondrial impairment,


protein that interacts with LC3 and is proposed and mobility abnormalities, are strikingly sim-
to play a role in cargo recruitment to the phag- ilar to the PD pathology in humans (Clark et al.
ophore (Gegg et al. 2010; Geisler et al. 2010). 2006; Park et al. 2006). However, dopaminergic
Importantly, several follow-up studies show that neurons remain normal in the park6 2/2 mouse,
disease-associated PARK2 and PARK6 muta- which implies an even more complicated mech-
tions result in defective mitophagy, thereby im- anism of PINK1 function in PD (Kitada et al.
plicating mitophagy defects in the development 2007).
of PD (Lee et al. 2010; Matsuda et al. 2010; Na-
rendra et al. 2010b).
Other PD-Related Genes and Autophagy
Although 95% of PD cases are sporadic, iden-
Functions of Different Isoforms of PINK1
tification of genes responsible for monogenic
PINK1 has at least two isoforms: a full-length forms has improved our knowledge of this neu-
form and an N-terminally truncated form (Bei- rodegenerative disease. In addition to SNCA
lina et al. 2005; Silvestri et al. 2005). PINK1 cleav- (encoding a-synuclein), PARK2, and PARK6,
age is mediated by the mitochondrial protease two other monogenic PD-related genes, encod-
rhomboid-7/PARL in flies and mammalian cells ing the leucine-rich repeat kinase 2 (LRRK2)
(Whitworth et al. 2008; Deas et al. 2010; Jin et al. and DJ-1, also play a role in autophagy or mito-
2010). However, which is the functional iso- chondrial dynamics.
form of PINK1 remains unclear. Early work indi- Mutation of the gene encoding LRRK2 is re-
cated the cleaved PINK1 might be the functional sponsible for an autosomal dominant form of
form, as the expression of a cytoplasmic, cleaved PD. LRRK2 is mainly localized in membrane
PINK1 is sufficient to protect neurons from mi- microdomains, multivesicular bodies, and auto-
tochondrial stress by MPTP (1-methyl-4-phe- phagic vesicles. Mutation or depletion of LRRK2
nyl-1,2,3,6-tetrahydropyridine) (Haque et al. results in autophagy impairment and the ac-
2008). In contrast, recent work suggests that full- cumulation of the autophagy marker proteins

Cite this article as Cold Spring Harb Perspect Med 2012;2:a009357 9


M.A. Lynch-Day et al.

LC3 and p62 (Alegre-Abarrategui et al. 2009; mendous advances in our understanding of the
Tong et al. 2010). In contrast, DJ-1 was identified causes of PD. Novel genes causing familial PD
as mediating autosomal recessive PD. Recent have been discovered, and have been shown to
studies also made a link between autophagy and be involved in the autophagy pathway, one of
DJ-1, as depletion of DJ-1 in both human neuro- the major proteolytic systems that maintain
blastoma cells and Drosophila results in mito- cellular protein homeostasis. Because autoph-
chondrial dysfunction and impaired autophagy agy is part of the cell’s homeostatic machinery,
(Hao et al. 2010; Thomas et al. 2010). maintaining a proper level of autophagy is im-
These findings imply that mitochondria portant for minimizing abnormal protein ag-
and autophagy might play a significant role or gregates and for facilitating organelle turnover.
even be the convergence points for different Discovery of therapeutic agents that boost au-
monogenic PD-related mutations that give rise tophagic activity or that directly maintain mi-
to similar symptoms. An alternative possibility tochondrial homeostasis, could potentially re-
is that these PD-related gene products might duce neuronal loss and slow down disease
function together. However, to date, only PINK1 progression. A better understanding of the reg-
and PARKIN have been shown to genetically ulatory mechanism of autophagy in the patho-
and physically interact, especially in modulating genesis of PD will enable the identification of
neuron protection, mitochondrial function, and possible methods for clinical intervention.
mitophagy. Thus, even though many studies
have begun to uncover the connections among ACKNOWLEDGMENTS
PINK1, PARKIN, and mitochondria, several
controversies remain to be resolved. This work was supported by NIH Grant GM5-
3396 to D.J.K.
www.perspectivesinmedicine.org

BASIC SCIENCE RESEARCH AND


CLINICAL TREATMENT REFERENCES

Based on the significant roles of mitochondria Alegre-Abarrategui J, Wade-Martins R. 2009. Parkinson dis-
ease, LRRK2 and the endocytic-autophagic pathway. Au-
and autophagy in PD, maintaining and stabiliz- tophagy 5: 1208–1210.
ing mitochondrial function or promoting the Alegre-Abarrategui J, Christian H, Lufino MM, Mutihac R,
degradation of damaged mitochondria might Venda LL, Ansorge O, Wade-Martins R. 2009. LRRK2
regulates autophagic activity and localizes to specific
benefit the protection of dopaminergic neurons. membrane microdomains in a novel human genomic re-
Data on the possible connection between defects porter cellular model. Hum Mol Genet 18: 4022– 4034.
in mitophagy and PD suggest that modulation Bandyopadhyay U, Cuervo AM. 2007. Chaperone-mediated
of autophagy might be one avenue for treating autophagy in aging and neurodegeneration: Lessons from
a-synuclein. Exp Gerontol 42: 120–128.
some types of this disease. However, autophagy
Banerjee R, Beal MF, Thomas B. 2010. Autophagy in neuro-
is described as a double-edged sword, because degenerative disorders: Pathogenic roles and therapeutic
both reduced and excessive autophagy can be implications. Trends Neurosci 33: 541 –549.
detrimental; therefore, simply upregulating au- Beilina A, Van Der Brug M, Ahmad R, Kesavapany S, Miller
DW, Petsko GA, Cookson MR. 2005. Mutations in
tophagy is not a practical course of action, and PTEN-induced putative kinase 1 associated with reces-
the application of autophagy-inducing drugs sive parkinsonism have differential effects on protein
must be undertaken with extreme caution. stability. Proc Natl Acad Sci 102: 5703–5708.
Brewer GJ, Wallimann TW. 2000. Protective effect of the en-
ergy precursor creatine against toxicity of glutamate and
CONCLUDING REMARKS b-amyloid in rat hippocampal neurons. J Neurochem 74:
1968–1978.
The turnover of proteins has been the focus Celsi F, Pizzo P, Brini M, Leo S, Fotino C, Pinton P, Rizzuto
of attention across neurodegenerative diseases, R. 2009. Mitochondria, calcium and cell death: A deadly
triad in neurodegeneration. Biochim Biophys Acta 1787:
given that many, if not all, of these diseases 335 –344.
show characteristic protein aggregation as part Chan NC, Salazar AM, Pham AH, Sweredoski MJ, Kolawa
of their cellular pathology. There have been tre- NJ, Graham RL, Hess S, Chan DC. 2011. Broad activation

10 Cite this article as Cold Spring Harb Perspect Med 2012;2:a009357


Autophagy and Parkinson’s Disease

of the ubiquitin-proteasome system by Parkin is critical PI(3)P-enriched domains of the endoplasmic reticulum
for mitophagy. Hum Mol Genet 20: 1726– 1737. and requires Rab1 GTPase. Autophagy 7: 17– 26.
Choubey V, Safiulina D, Vaarmann A, Cagalinec M, Wareski Hutchins MU, Klionsky DJ. 2001. Vacuolar localization of
P, Kuum M, Zharkovsky A, Kaasik A. 2011. Mutant A53T oligomeric a-mannosidase requires the cytoplasm to va-
a-synuclein induces neuronal death by increasing mito- cuole targeting and autophagy pathway components in
chondrial autophagy. J Biol Chem 286: 10814– 10824. Saccharomyces cerevisiae. J Biol Chem 276: 20491– 20498.
Clark IE, Dodson MW, Jiang C, Cao JH, Huh JR, Seol JH, Jin SM, Lazarou M, Wang C, Kane LA, Narendra DP, Youle
Yoo SJ, Hay BA, Guo M. 2006. Drosophila pink1 is re- RJ. 2010. Mitochondrial membrane potential regulates
quired for mitochondrial function and interacts geneti- PINK1 import and proteolytic destabilization by PARL.
cally with parkin. Nature 441: 1162–1166. J Cell Biol 191: 933– 942.
Clayton DF, George JM. 1998. The synucleins: A family of Kanki T, Wang K, Cao Y, Baba M, Klionsky DJ. 2009. Atg32 is
proteins involved in synaptic function, plasticity, neuro- a mitochondrial protein that confers selectivity during
degeneration and disease. Trends Neurosci 21: 249– 254. mitophagy. Dev Cell 17: 98–109.
Clayton DF, George JM. 1999. Synucleins in synaptic plasti- Keeble JA, Gilmore AP. 2007. Apoptosis commitment—
city and neurodegenerative disorders. J Neurosci Res 58: Translating survival signals into decisions on mitochon-
120–129. dria. Cell Res 17: 976 –984.
Cuervo AM, Stefanis L, Fredenburg R, Lansbury PT, Sulzer Kitada T, Pisani A, Porter DR, Yamaguchi H, Tscherter A,
D. 2004. Impaired degradation of mutant a-synuclein Martella G, Bonsi P, Zhang C, Pothos EN, Shen J. 2007.
by chaperone-mediated autophagy. Science 305: 1292– Impaired dopamine release and synaptic plasticity in
1295. the striatum of PINK1-deficient mice. Proc Natl Acad
Deas E, Plun-Favreau H, Gandhi S, Desmond H, Kjaer S, Sci 104: 11441–11446.
Loh SH, Renton AE, Harvey RJ, Whitworth AJ, Martins Klionsky DJ. 2005. The molecular machinery of autophagy:
LM, et al. 2010. PINK1 cleavage at position A103 by Unanswered questions. J Cell Sci 118: 7 –18.
the mitochondrial protease PARL. Hum Mol Genet 20: Klionsky DJ, Cueva R, Yaver DS. 1992. Aminopeptidase I
867–879. of Saccharomyces cerevisiae is localized to the vacuole in-
Eskelinen E-L, Reggiori F, Baba M, Kovacs AL, Seglen PO. dependent of the secretory pathway. J Cell Biol 119:
2011. Seeing is believing. The impact of electron micro- 287 –299.
scopy on autophagy research. Autophagy 7: 935– 956.
www.perspectivesinmedicine.org

Klionsky DJ, Cuervo AM, Dunn WA Jr, Levine B, van der


Ferrante RJ, Andreassen OA, Jenkins BG, Dedeoglu A, Klei I, Seglen PO. 2007. How shall I eat thee? Autophagy
Kuemmerle S, Kubilus JK, Kaddurah-Daouk R, Hersch 3: 413– 416.
SM, Beal MF. 2000. Neuroprotective effects of creatine Kunz JB, Schwarz H, Mayer A. 2004. Determination of four
in a transgenic mouse model of Huntington’s disease. J sequential stages during microautophagy in vitro. J Biol
Neurosci 20: 4389– 4397. Chem 279: 9987– 9996.
Gasser T. 2009. Mendelian forms of Parkinson’s disease. Lambert AJ, Brand MD. 2004. Inhibitors of the quinone-
Biochim Biophys Acta 1792: 587 –596. binding site allow rapid superoxide production from mi-
Gegg ME, Cooper JM, Chau KY, Rojo M, Schapira AH, tochondrial NADH:ubiquinone oxidoreductase (com-
Taanman JW. 2010. Mitofusin 1 and mitofusin 2 are plex I). J Biol Chem 279: 39414–39420.
ubiquitinated in a PINK1/parkin-dependent manner Lee JY, Nagano Y, Taylor JP, Lim KL, Yao TP. 2010. Disease-
upon induction of mitophagy. Hum Mol Genet 19: causing mutations in parkin impair mitochondrial ubiq-
4861– 4870. uitination, aggregation, and HDAC6-dependent mitoph-
Geisler S, Holmstrom KM, Skujat D, Fiesel FC, Rothfuss agy. J Cell Biol 189: 671–679.
OC, Kahle PJ, Springer W. 2010. PINK1/Parkin- Littarru GP, Tiano L. 2007. Bioenergetic and antioxidant
mediated mitophagy is dependent on VDAC1 and p62/ properties of coenzyme Q10: Recent developments. Mol
SQSTM1. Nat Cell Biol 12: 119– 131. Biotechnol 37: 31–37.
Hao LY, Giasson BI, Bonini NM. 2010. DJ-1 is critical for mi- Lynch-Day MA, Bhandari D, Menon S, Huang J, Cai H, Bar-
tochondrial function and rescues PINK1 loss of function. tholomew CR, Brumell JH, Ferro-Novick S, Klionsky DJ.
Proc Natl Acad Sci 107: 9747–9752. 2010. Trs85 directs a Ypt1 GEF, TRAPPIII, to the phago-
Haque ME, Thomas KJ, D’Souza C, Callaghan S, Kitada T, phore to promote autophagy. Proc Natl Acad Sci 107:
Slack RS, Fraser P, Cookson MR, Tandon A, Park DS. 7811–7816.
2008. Cytoplasmic Pink1 activity protects neurons from Majeski AE, Dice JF. 2004. Mechanisms of chaperone-
dopaminergic neurotoxin MPTP. Proc Natl Acad Sci mediated autophagy. Int J Biochem Cell Biol 36: 2435–
105: 1716–1721. 2444.
Hardy J, Lewis P, Revesz T, Lees A, Paisan-Ruiz C. 2009. The Mao K, Wang K, Zhao M, Xu T, Klionsky DJ. 2011. Two
genetics of Parkinson’s syndromes: A critical review. Curr MAPK signaling pathways are required for mitophagy
Opin Genet Dev 19: 254 –265. in Saccharomyces cerevisiae. J Cell Biol 193: 755– 767.
Huang J, Klionsky DJ. 2007. Autophagy and human disease. Martinez-Vicente M, Tallóczy Z, Kaushik S, Massey AC,
Cell Cycle 6: 1837–1849. Mazzulli J, Mosharov EV, Hodara R, Fredenburg R, Wu
Huang J, Birmingham CL, Shahnazari S, Shiu J, Zheng YT, DC, Follenzi A, et al. 2008. Dopamine-modified a-synu-
Smith AC, Campellone KG, Heo WD, Gruenheid S, clein blocks chaperone-mediated autophagy. J Clin Invest
Meyer T, et al. 2011. Antibacterial autophagy occurs at 118: 777–788.

Cite this article as Cold Spring Harb Perspect Med 2012;2:a009357 11


M.A. Lynch-Day et al.

Matsuda N, Sato S, Shiba K, Okatsu K, Saisho K, Gautier Shintani T, Huang W-P, Stromhaug PE, Klionsky DJ. 2002.
CA, Sou YS, Saiki S, Kawajiri S, Sato F, et al. 2010. Mechanism of cargo selection in the cytoplasm to va-
PINK1 stabilized by mitochondrial depolarization re- cuole targeting pathway. Dev Cell 3: 825–837.
cruits Parkin to damaged mitochondria and activates la- Silvestri L, Caputo V, Bellacchio E, Atorino L, Dallapiccola
tent Parkin for mitophagy. J Cell Biol 189: 211–221. B, Valente EM, Casari G. 2005. Mitochondrial import
McBride HM, Neuspiel M, Wasiak S. 2006. Mitochondria: and enzymatic activity of PINK1 mutants associated to
More than just a powerhouse. Curr Biol 16: R551–R560. recessive parkinsonism. Hum Mol Genet 14: 3477–3492.
Naderi J, Somayajulu-Nitu M, Mukerji A, Sharda P, Sikorska Spencer B, Potkar R, Trejo M, Rockenstein E, Patrick C,
M, Borowy-Borowski H, Antonsson B, Pandey S. 2006. Gindi R, Adame A, Wyss-Coray T, Masliah E. 2009. Beclin
Water-soluble formulation of Coenzyme Q10 inhibits 1 gene transfer activates autophagy and ameliorates the
Bax-induced destabilization of mitochondria in mam- neurodegenerative pathology in a-synuclein models of
malian cells. Apoptosis 11: 1359–1369. Parkinson’s and Lewy body diseases. J Neurosci 29:
Nair U, Klionsky DJ. 2005. Molecular mechanisms and reg- 13578– 13588.
ulation of specific and nonspecific autophagy pathways Spillantini MG, Schmidt ML, Lee VM-Y, Trojanowski JQ,
in yeast. J Biol Chem 280: 41785–41788. Jakes R, Goedert M. 1997. a-Synuclein in Lewy bodies.
Narendra D, Tanaka A, Suen DF, Youle RJ. 2008. Parkin is re- Nature 388: 839 –840.
cruited selectively to impaired mitochondria and pro- Spillantini MG, Crowther RA, Jakes R, Hasegawa M, Goe-
motes their autophagy. J Cell Biol 183: 795– 803. dert M. 1998. a-Synuclein in filamentous inclusions of
Narendra D, Kane LA, Hauser DN, Fearnley IM, Youle RJ. Lewy bodies from Parkinson’s disease and dementia
2010a. p62/SQSTM1 is required for Parkin-induced mi- with Lewy bodies. Proc Natl Acad Sci 95: 6469–6473.
tochondrial clustering but not mitophagy; VDAC1 is dis- Stefanis L, Larsen KE, Rideout HJ, Sulzer D, Greene LA.
pensable for both. Autophagy 6: 1090– 1106. 2001. Expression of A53T mutant but not wild-type
Narendra DP, Jin SM, Tanaka A, Suen DF, Gautier CA, Shen a-synuclein in PC12 cells induces alterations of the
J, Cookson MR, Youle RJ. 2010b. PINK1 is selectively ubiquitin-dependent degradation system, loss of dopa-
stabilized on impaired mitochondria to activate Parkin. mine release, and autophagic cell death. J Neurosci 21:
PLoS Biol 8: e1000298. 9549–9560.
Okamoto K, Kondo-Okamoto N, Ohsumi Y. 2009. Mito- Szeto HH. 2008. Development of mitochondria-targeted
chondria-anchored receptor Atg32 mediates degradation aromatic-cationic peptides for neurodegenerative dis-
www.perspectivesinmedicine.org

of mitochondria via selective autophagy. Dev Cell 17: eases. Ann NY Acad Sci 1147: 112 –121.
87–97. Terzioglu M, Galter D. 2008. Parkinson’s disease: Genetic
Pankiv S, Clausen TH, Lamark T, Brech A, Bruun JA, Outzen versus toxin-induced rodent models. FEBS J 275: 1384–
H, Overvatn A, Bjorkoy G, Johansen T. 2007. p62/SQ- 1391.
STM1 binds directly to Atg8/LC3 to facilitate degrada- Thomas KJ, McCoy MK, Blackinton J, Beilina A, van der
tion of ubiquitinated protein aggregates by autophagy. J Brug M, Sandebring A, Miller D, Maric D, Cedazo-
Biol Chem 282: 24131–24145. Minguez A, Cookson MR. 2010. DJ-1 acts in parallel to
Papucci L, Schiavone N, Witort E, Donnini M, Lapucci A, the PINK1/parkin pathway to control mitochondrial
Tempestini A, Formigli L, Zecchi-Orlandini S, Orlandini function and autophagy. Hum Mol Genet 20: 40– 50.
G, Carella G, et al. 2003. Coenzyme q10 prevents apopto- Tong Y, Yamaguchi H, Giaime E, Boyle S, Kopan R, Kelleher
sis by inhibiting mitochondrial depolarization inde- RJ III, Shen J. 2010. Loss of leucine-rich repeat kinase 2
pendently of its free radical scavenging property. J Biol causes impairment of protein degradation pathways, ac-
Chem 278: 28220– 28228. cumulation of a-synuclein, and apoptotic cell death in
Park J, Lee SB, Lee S, Kim Y, Song S, Kim S, Bae E, Kim J, aged mice. Proc Natl Acad Sci 107: 9879– 9884.
Shong M, Kim J-M, et al. 2006. Mitochondrial dysfunc- Vives-Bauza C, Zhou C, Huang Y, Cui M, de Vries RL, Kim J,
tion in Drosophila PINK1 mutants is complemented by May J, Tocilescu MA, Liu W, Ko HS, et al. 2010. PINK1-
parkin. Nature 441: 1157–1161. dependent recruitment of Parkin to mitochondria in mi-
Polymeropoulos MH, Lavedan C, Leroy E, Ide SE, Dehejia A, tophagy. Proc Natl Acad Sci 107: 378– 383.
Dutra A, Pike B, Root H, Rubenstein J, Boyer R, et al. 1997. Vogiatzi T, Xilouri M, Vekrellis K, Stefanis L. 2008. Wild type
Mutation in the a-synuclein gene identified in families a-synuclein is degraded by chaperone-mediated autoph-
with Parkinson’s disease. Science 276: 2045–2047. agy and macroautophagy in neuronal cells. J Biol Chem
Schapira AH. 1993. Mitochondrial complex I deficiency in 283: 23542–23556.
Parkinson’s disease. Adv Neurol 60: 288 –291. Wallace DC. 2005. A mitochondrial paradigm of metabolic
Schapira AH. 2008. Mitochondria in the aetiology and and degenerative diseases, aging, and cancer: A dawn for
pathogenesis of Parkinson’s disease. Lancet Neurol 7: evolutionary medicine. Annu Rev Genet 39: 359– 407.
97–109. Wang K, Klionsky DJ. 2011. Mitochondria removal by au-
Schapira AH, Cooper JM, Dexter D, Jenner P, Clark JB, tophagy. Autophagy 7: 297 –300.
Marsden CD. 1989. Mitochondrial complex I deficiency Whitworth AJ, Lee JR, Ho VM, Flick R, Chowdhury R,
in Parkinson’s disease. Lancet 1: 1269. McQuibban GA. 2008. Rhomboid-7 and HtrA2/Omi
Schuler F, Casida JE. 2001. Functional coupling of PSSTand act in a common pathway with the Parkinson’s disease
ND1 subunits in NADH:ubiquinone oxidoreductase es- factors Pink1 and Parkin. Dis Model Mech 1: 168– 174.
tablished by photoaffinity labeling. Biochim Biophys Acta Winslow AR, Chen C-W, Corrochano S, Acevedo-Arozena
1506: 79–87. A, Gordon DE, Peden AA, Lichtenberg M, Menzies FM,

12 Cite this article as Cold Spring Harb Perspect Med 2012;2:a009357


Autophagy and Parkinson’s Disease

Ravikumar B, Imarisio S, et al. 2010. a-Synuclein impairs by chaperone-mediated autophagy. Science 323: 124–
macroautophagy: Implications for Parkinson’s disease. J 127.
Cell Biol 190: 1023–1037. Yorimitsu T, Klionsky DJ. 2005. Atg11 links cargo to the
Xie Z, Nair U, Klionsky DJ. 2008. Atg8 controls phagophore vesicle-forming machinery in the cytoplasm to vacuole
expansion during autophagosome formation. Mol Biol targeting pathway. Mol Biol Cell 16: 1593–1605.
Cell 19: 3290–3298. Yuga M, Gomi K, Klionsky DJ, Shintani T. 2011. Aspartyl
Xilouri M, Vogiatzi T, Vekrellis K, Park D, Stefanis L. 2009. aminopeptidase is imported from the cytoplasm to the
Abberant a-synuclein confers toxicity to neurons in vacuole by selective autophagy in Saccharomyces cerevi-
part through inhibition of chaperone-mediated autoph- siae. J Biol Chem 286: 13704–13713.
agy. PLoS One 4: e5515. Zhao K, Zhao GM, Wu D, Soong Y, Birk AV, Schiller PW,
Yang Y, Gehrke S, Imai Y, Huang Z, Ouyang Y, Wang J-W, Szeto HH. 2004. Cell-permeable peptide antioxidants
Yang L, Beal MF, Vogel H, Lu B. 2006. Mitochondrial targeted to inner mitochondrial membrane inhibit mito-
pathology and muscle and dopaminergic neuron chondrial swelling, oxidative cell death, and reperfusion
degeneration caused by inactivation of Drosophila injury. J Biol Chem 279: 34682– 34690.
Pink1 is rescued by Parkin. Proc Natl Acad Sci 103: Ziviani E, Tao RN, Whitworth AJ. 2010. Drosophila parkin
10793–10798. requires PINK1 for mitochondrial translocation and
Yang Q, She H, Gearing M, Colla E, Lee M, Shacka JJ, Mao Z. ubiquitinates mitofusin. Proc Natl Acad Sci 107: 5018–
2009. Regulation of neuronal survival factor MEF2D 5023.
www.perspectivesinmedicine.org

Cite this article as Cold Spring Harb Perspect Med 2012;2:a009357 13

You might also like