You are on page 1of 11

REVIEW

The Vicious Cycle Between α-Synuclein Aggregation and


Autophagic-Lysosomal Dysfunction
Giovanni Bellomo, PhD,1 Silvia Paciotti, PhD,2,3 Leonardo Gatticchi, PhD,3 and Lucilla Parnetti, MD, PhD2*

1
Magnetic Resonance Center (CERM), University of Florence, Sesto Fiorentino (FI), Italy
2
Laboratory of Clinical Neurochemistry, Section of Neurology, University of Perugia, Perugia (PG), Italy
3
Department of Experimental Medicine, University of Perugia, Perugia (PG), Italy

A B S T R A C T : The accumulation and misfolding of pathway dysfunctions’ role in the pathogenesis and pro-
α-synuclein (α-syn) represent the main pathological hall- gression of synucleinopathies opened new perspectives
mark of PD. Overexpression of α-syn and failure of cellu- for novel possible therapeutic strategies. In this article,
lar protein degradation systems play a major role in the evidences and mechanisms of the reciprocal rela-
α-syn aggregation. The discovery of PD-associated genes tion between autophagic-lysosomal pathway impair-
related to the autophagic-lysosomal pathway, such as ment and misfolded α-syn aggregation and propagation
VPS35, LRRK2, GBA1, SMPD1, GALC, ASAH1, SCARB2, are reviewed, together with the most promising com-
CTSD, CTSB, and GLA, confirms the involvement of cellu- pounds targeting autophagic-lysosomal pathway resto-
lar clearance systems dysfunction in PD pathogenesis. Of ration as a disease-modifying strategy for PD treatment.
importance, lysosomal enzyme activity is altered both in © 2019 International Parkinson and Movement Disorder
genetic and sporadic PD. Decreased lysosomal enzymes Society
activities were measured in the same brain regions where
α-syn accumulates, suggesting that a crosstalk between Key Words: alpha-synuclein; Parkinson’s disease; lyso-
α-syn aggregation and autophagic-lysosomal impairment some; GCase; cathepsin-D; autophagy
may exist. The understanding of autophagic-lysosomal

Parkinson’s disease (PD) is an age-related neurodegen- elements.2 Although the presence of LBs in SN has been
erative disease with a complex etiology and varying associated to the death of dopaminergic neurons,3,4
clinical manifestations. The accumulation, misfolding, smaller aggregates rather than LBs, like protofibrils and
and aggregation of the intrinsically disordered protein, soluble oligomers, are currently considered to be the
α-synuclein (α-syn), and its progressive deposition in most toxic species in the amyloidogenic pathway of
large intracellular aggregates, named Lewy bodies (LBs), α-syn.5-7 Among the causes of uncontrolled α-syn accu-
represent the main pathological hallmark of most genetic mulation and aggregation, overexpression of α-syn and
and sporadic forms of PD.1 These inclusions were failure of cellular protein degradation systems play a
recently found to wrap and encase vesicles, lysosomes, major role.8 Mutations in genes associated to the
dysmorphic mitochondria, and disrupted cytoskeletal autophagic-lysosomal pathway (ALP) are recognized as
genetic causes or risks factors for PD onset.9 Autophagy
-*Correspondence
- - - - - - - - - - - - - - -to:- - Prof.
- - - - Lucilla
- - - - - -Parnetti,
- - - - - - -Laboratory
- - - - - - - - -of- -Clinical
- - - - - -Neu-
----- is a catabolic process in which dysfunctional cytoplasmic
components (organelles and proteins) are rerouted
rochemistry, Section of Neurology, University of Perugia, Piazzale Lucio
Severi 1/8, 06132 Perugia (PG), Italy; E-mail: lucilla.parnetti@unipg.it toward the lysosomal environment for degradation.
Relevant conflicts of interest/financial disclosures: Nothing to Three types of autophagy have been identified based on
report. the pathway by which the substrates reach the lysosomal
Full financial disclosures and author roles may be found in the online compartment: macroautophagy, microautophagy, and
version of this article. chaperone-mediated autophagy (CMA).10
Received: 17 June 2019; Revised: 31 August 2019; Accepted: 27 Sep- Macroautophagy is the most well-established type of
tember 2019 autophagy, which is characterized by the formation of
Published online 15 November 2019 in Wiley Online Library double-membrane vesicles (autophagosomes) enclosing
(wileyonlinelibrary.com). DOI: 10.1002/mds.27895 bulky materials. Autophagosomes fuse with lysosomes

34 Movement Disorders, Vol. 35, No. 1, 2020


α- S Y N U C L E I N A N D A U T O P H A G I C - L Y S O S O M A L V I C I O U S C Y C L E

and release their content into the lysosomal lumen, VPS35 encodes vacuolar protein sorting-associated
where acid hydrolases have the task of degrading it. The protein 35, which is involved in the endosomal-lysosomal
master regulator of this process is mTOR (mammalian trafficking associated to autophagy. Mutations on VPS35
target of rapamycin), a 298-kDa serine/threonine kinase, cause a rare form of autosomal-dominant PD.30 Another
which negatively modulates macroautophagy through gene which has been defined causative of PD is LRRK2,
the regulation of autophagy-related genes (Atg-genes). which encodes for leucine-rich repeat kinase 2 (LRRK2).
An mTOR-dependent pathway also inhibits the translo- Mutations on LRRK2 are the most common known
cation from the cytosol to the nucleus of transcriptional genetic causes for PD. The most prevalent pathogenic
factor EB (TFEB),11,12 which plays a prominent role in mutation, G2019S, increases LRRK2 kinase activity and
the biogenesis and function of lysosomes. impairs the autophagy process, leading to a significant
In microautophagy, cytosolic substrates are directly accumulation of α-syn, as assessed by in in vitro and
taken up by the lysosomes and released onto the lyso- in vivo experiments.31-33
somal lumen. GBA1 gene encodes for β-glucocerebrosidase (GCase), a
Differently from macroautophagy and microautophagy, lysosomal acidic hydrolase involved in glycosphingolipid
CMA is a highly selective catabolic process. Only specific catabolism. Whereas GBA1 homozygous mutations cause
soluble cytosolic proteins containing a KFERQ-like Gaucher’s disease (GD), a lysosomal storage disorder, het-
amino-acidic sequence are brought to the lysosomes by erozygous GBA1 mutation carriers have a 5- to 6-fold
CMA. Tagged proteins are recognized by a chaperone increased risk to develop PD.34 Some researchers suggest
complex involving the heat shock cognate protein of that GBA1 mutations should be considered causative fac-
70 kDa (Hsc70). The complex is translocated to the lyso- tors for PD.35 However, the penetrance of PD in GBA1
somes and internalized by the lysosomal-associated mem- mutation carriers is low: 7.6%, 13.7%, 21.4%, and
brane protein 2a (LAMP2a) receptor. Macroautophagy 29.7% at 50, 60, 70, and 80 years, under a dominant
and CMA are major degradation pathways for α-syn13,14; model, with no relevant differences among N370S carriers,
therefore, the knowledge of the relation between α-syn L444P carriers, and carriers of rarer mutations.35 This
misfolding and ALP dysfunctions may provide new incomplete penetrance suggests that other genetic and/or
insights in the pathogenesis of synucleinopathies and in environmental factors are necessary for carriers of GBA1
the discovery of novel therapeutic targets. mutations to manifest α-syn oligomerization and to
develop PD.36 However, a more severe disease phenotype
has been associated to the burden of carried GBA1
mutations.37,38
Mutations in Genes Encoding Recently, variants in other lysosomal genes, even
from the same pathway of GBA1, were also associated
Autophagic-Lysosomal Proteins with PD susceptibility.9
Interfere with PD Pathogenic Single-nucleotide polymorphisms (SNPs) on SCARB2,
Mechanisms which encodes for lysosomal integral membrane protein
2 (LIMP-2), a protein responsible for GCase trafficking,
Genetic forms of PD are associated to variants in sev- have been also shown to be linked to the onset of LB
eral genes, which range from highly penetrant rare pathologies.39,40 Although no relevant changes in GCase
familial mutations, considered causative of PD (SNCA, activities were recently found in dried blood spots of PD
PRKN, LRRK2, PINK1, DJ-1, and VPS35)15 to com- patients carrying SCARB2 SNPs versus controls,41
mon genetic risk factors and genes that confer suscepti- homozygous mutations are known to be responsible for
bility to PD (GBA1, SMPD1, GALC, ASAH1, the onset of lysosomal disorders with a systemic reduc-
SCARB2, CTSD, CTSB, and GLA).15-19 The protein tion in GCase activity.42 Conversely, overexpression of
products of some of these genes play pivotal roles in LIMP-2 enhanced α-syn clearance and improved lyso-
regulating autophagy-lysosome function and vesicular somal activity of GCase in different cell lines over-
trafficking. Moreover, mutations on some of these regu- expressing α-syn.43
lators has been demonstrated to directly or indirectly α-Syn degradation can be achieved following two
favor α-syn accumulation processes.20-22 main degradative routes, that is, ALP44-48 and ubiquitin-
In particular, the identification of familial cases with proteasome system.44,49 Variants in genes, which induce
multiplications of SNCA (the gene encoding for α-syn) dysfunction at multiple levels of ALP, gradually lead to
revealed a strong correlation between α-syn accumula- the impairment of protein degradation that may favor
tion and aggregation, and disease severity.23-25 Although the development of PD. This mechanism creates a
the physiological role of α-syn is not fully characterized, vicious cycle linking α-syn accumulation to ALP dys-
it has been shown that it plays important roles in the function and vice versa. The causal links among GBA1
supply of synaptic vesicles in presynaptic terminals26,27 and other ALP-related mutations, PD, and α-syn
and in the movement of microtubules.28,29 decreased clearance are still unclear and currently under

Movement Disorders, Vol. 35, No. 1, 2020 35


B E L L O M O E T A L

investigation; some hypotheses, based on recent scientific patients, both in GBA1 mutation carriers and noncar-
evidences, are described below. riers. Decreased levels of LAMP1, CTSD, and heat shock
protein 73 were also observed in PD nigral neurons when
compared to age-matched controls.64 This decrease was
The issue of ALP Alterations in significantly greater in nigral neurons that contained
Sporadic PD α-syn inclusions. This pattern was recapitulated in rats
with targeted overexpression of mutant α-syn in the SN,
To date, familiar forms of PD account for only approxi- which resulted in dopamine cell loss with α-syn inclu-
mately 5% of patients. GBA1 mutations, which confer sions. Significantly reduced α-Gal A and CTSD activities
susceptibility to develop PD, are the most common were found in temporal cortex of PD patients in advanced
genetic risk factors and account for 10% to 15%, a per- stage. The activities of these two enzymes were inversely
centage that increases to 25% in the Ashkenazi Jewish correlated with levels of p129S–α-syn and high-molecu-
population.15,50,51 In PD, sporadic forms represent the lar-weight (hMW) α-syn species.65 Of importance, also
vast majority, being the consequence of the aging process cathepsin-B (CTSB) activity correlated negatively with
itself and the combination of harmful environmental fac- α-syn hMW species.65 In SN and amygdala of sporadic
tors added on a susceptible genetic background. Such PD patients, HSC70 and LAMP2A were found signifi-
convergence of factors leads to an ineffective response cantly reduced with respect to both controls and
against the accumulation and aggregation of α-syn.52 Of Alzheimer’s disease patients.66 Reduction of LAMP2A
importance, the activity of some lysosomal enzymes is protein was also observed in early-stage PD anterior cin-
found significantly reduced in PD versus controls both in gulate cortex. Lower levels LAMP2A directly correlated
the presence and in absence of genetic risk factors. Two with the increased levels of α-syn and decreased levels of
studies carried out between 200753 and 200954 showed HSC70 in the same PD samples, as well as with the accu-
decreased activities of GCase in cerebrospinal fluid (CSF) mulation of cytosolic CMA substrate proteins.67 In the
of PD and DLB (dementia with Lewy bodies) patients SN of PD patients, decreased levels of LAMP1 and
with respect to controls. Subsequent observations in increased levels of the autophagosomal marker, light
larger series55,56 not only confirmed the decrease of CSF chain 3 (LC3)-II, were also found.64,68 LC3 colocalized
GCase activity, but also showed the reduction of with α-syn in the halo of LB, and it was also detected in
cathepsin-D (CTSD), independently of the GBA1 carrier α-syn-positive Lewy neurites. Increased levels of mTOR
status. The reduced activity of GCase in biofluids of both protein were found in the temporal cortex of patients
GBA1 mutation carriers and sporadic PD patients has with DLB, particularly in neurons displaying α-syn accu-
been observed also in dried blood spots.37 mulation.69 The evidence of decreased activity/levels of
Similarly, reduced activity of the lysosomal enzyme, different lysosomal enzymes and lysosome-associated pro-
α-galactosidase A (α-Gal A; deficient in Fabry’s disease), teins in sporadic PD, concomitantly to α-syn accumula-
was found in dried blood spots of PD patients com- tion in the same brain regions,60,64,65 seems to indicate a
pared to controls, both excluding GBA1 and LRRK2 relationship between lysosomal enzymes failure and
G2019S mutation carriers and considering the entire synucleinopathies.
cohort.57 Decreased activities of α-Gal A have been also
measured in the leukocytes of sporadic PD patients.58
In brain tissue, GCase activity and content have been Dysfunction of the ALP Pathway Leads to
shown to be reduced in the SN and other brain regions in Aggregation and Propagation of Misfolded
different studies, both in GBA1 mutation carriers and α-Syn in Cellular and Animal Models
sporadic PD patients.59-61 Importantly, Murphy and col- In several animal models, as well as in patients affected
leagues60 showed that the reduced activity is more evident by lysosomal storage disorders, such as GD, GM2
at the early PD stage, and that the brain regions in which gangliosidosis, Niemann-Pick type C, β-galactosialidosis,
GCase activity is most affected are also those in which NCL10, and Krabbe’s disease, α-syn accumulation has
α-syn accumulates the most. Chiasserini and colleagues62 been observed.70-72 Moreover, in GD patients, levels of
reported decreased GCase activity in brain tissues of non- plasma oligomeric α-syn were found higher with respect
GBA1-mutation carrier PD and DLB patients, with the to controls73 and negatively associated with leucocyte
SN and caudate being the most affected regions. Reduced GCase activity.74 Increased plasma oligomeric α-syn
GCase activity was also found in the cingulate cortex of levels were also found in a small group of patients
PD patients without GBA1 mutations.63 Moors and col- affected by rarer lysosomal storage disorders, namely
leagues61 found significantly decreased GCase activity in Niemann-Pick type C1 and C2 disease, Krabbe’s dis-
the SN of both PD and DLB patients; a similar, though ease, and Wolman’s disease.73
not significant, trend was also observed in the frontal cor- In cellular models (non-neuronal cells, differentiated
tex and putamen. In this series, CTSD activity was signifi- neuroblastoma cells, and primary cortical neurons),
cantly decreased in the frontal cortex of DLB and PD blocking the ALP with bafilomycin A1 results in the

36 Movement Disorders, Vol. 35, No. 1, 2020


α- S Y N U C L E I N A N D A U T O P H A G I C - L Y S O S O M A L V I C I O U S C Y C L E

accumulation of α-syn aggregates.44,75 Similarly, inhibi- increased α-syn levels. Furthermore, GCase knockdown
tion of macroautophagy by 3-methyladenine led to an and concomitant expression of WT α-syn and A53T α-syn
increase of both endogenous and overexpressed wild- enhanced α-syn-mediated neurotoxicity through formation
type (WT) α-syn in PC12 cells.13 Increased monomeric of soluble and insoluble hMW species. A fibrillization-
and aggregated α-syn levels were also found in an incompetent mutant (Δ71-82 α-syn) also accumulated, but
Atg7-depleted mouse model.76,77 showed no toxicity. Accumulation of both soluble and
α-Syn accumulation was also observed in dopaminer- oligomeric insoluble α-syn was observed also in brains of
gic neurons of VPS35 mutant mice.78 Experiments car- GD mouse models (4L/PS-NA), as compared to WT mice.
ried out in neuroblastoma cells showed that expression In GD patients, α-syn dimer/monomer ratio positively cor-
of the R524W VPS35 mutant strongly impairs recruit- related with levels of glucosylceramide (GlcCer, the GCase
ment of retromer-dependent interacting proteins and the substrate) and the glucosylceramide/ceramide (GlcCer/Cer,
trafficking of cation-independent mannose-6-phosphate Cer is the GCase product) ratio in red blood cells.90 GlcCer
receptor, which resulted in higher levels of α-syn-positive stabilizes toxic oligomeric α-syn intermediates,63,91-93
aggregates when compared to WT or P316S VPS35.79 which have been shown to diffuse through membranes94
Also β-hexosaminidase (β-hex) activity seems to influ- and to inhibit the opening of the proteasome gate.95 These
ence α-syn clearance. In a mouse model of Sandhoff dis- features may be relevant for the cell-to-cell transmission of
ease, α-syn accumulation in neurons was observed aggregates and protein misfolding cascade.
along with the accumulation GM2 ganglioside.80 About the link between GCase reduced activity and
Among all the lysosomal enzymes, cathepsins have been α-syn accumulation, another putative causal factor might
found to be directly involved in the degradation of be represented by the reduction of ceramide (Cer) levels
monomeric and aggregated α-syn.48,81-84 In particular, in endosomal and lysosomal compartments, as a conse-
membrane-bound CTSD is able to fully degrade mono- quence of GCase failure. Given that Cer binds CTSD and
meric and aggregated α-syn, whereas CTSB and cathepsin- triggers its cleavage to the catalytic active form,96,97 lower
L (CTSL) can degrade α-syn monomers and fibrils in their levels of Cer may also lead to a decreased capacity of the
free state.48 However, in murine models overexpressing lysosome to degrade α-syn98; this mechanism is schema-
A53T α-syn, the latter degradation pathway (CTSB and tized in Figure 1. Altered Cer levels have been found in
CTSL) has recently shown to produce highly aggregation- PD. In particular, Cer levels were increased in plasma
prone C-terminus truncated forms of α-syn.85 samples of PD patients with respect to controls.99-101
GCase is not directly involved in α-syn degradation, but Conversely, lower levels of Cer as well as its altered chain
its reduced activity/availability promotes the accumulation composition have been described in the anterior cingulate
and aggregation of α-syn in different cellular cultures and cortex of PD patients with respect to controls.60,102 As a
animal models.63,86-89 Mazzulli and colleagues63 demon- support for this link between Cer and PD, the inhibition
strated, in human cortical neurons, that GCase depletion of acid ceramidase (encoded by ASAH1) by carmofur
compromises protein degradation capacity, leading to resulted in increased Cer levels in GCase-deficient cells as

FIG. 1. (A) Graphical summary of the mechanisms by which GCase dysfunctions may lead to misfolded α-syn aggregation and propagation.
(B) Putative mechanisms by which α-syn aggregates may impair GCase activity leading to a bidirectional pathogenic loop. [Color figure can be viewed
at wileyonlinelibrary.com]

Movement Disorders, Vol. 35, No. 1, 2020 37


B E L L O M O E T A L

well as reduced α-syn levels in GBA1-PD–derived dopa- impair ALP functioning, thus leading to a positive feed-
minergic neurons.103 It is also worthy to note that vari- back loop forward α-syn aggregation.46 In mammalian
ants of acid sphingomyelinase (aSMase; encoded by cells and in transgenic mice, overexpression of WT and
SMPD1)98,104-106 and galactosylceramidase (GALC)17 are both A30P and A53T mutated α-syn leads to inhibition
recognized as susceptibility factors for PD and other syn- of macroautophagy,120,121 which is caused by a reduc-
ucleinopathies. Together with GCase, these two enzymes tion in autophagosome formation through α-syn/Rab1a
catalyze the production of Cer within the lysosome. interaction.120 Furthermore, in α-syn transgenic mice,
aSMase belongs to the sphingomyelinase family, responsi- intracerebral infusion of rapamycin, an inhibitor of
ble for catalyzing the breakdown of sphingomyelin to Cer mTOR, resulted in reduced accumulation of α-syn and
and phosphorylcholine. A recent study showed that in amelioration of associated neurodegenerative alter-
reduced aSMase activity in blood samples is associated to ations.69 Interestingly, the A30P and A53P mutated
a 3.5- to 5.8-year earlier onset of PD.107 α-syn forms bind LAMP2A with higher affinity with
GALC catalyzes the hydrolysis of different substrates respect to WT α-syn.13,66,122 Binding of mutated α-syn
(i.e., galactosylceramide and galactosylsphingosine). with LAMP2A blocks CMA with the consequent accu-
Galactosylsphingosine levels were higher in the cerebral mulation of hMW α-syn and formation of insoluble
cortex of PD patients compared to age-matched con- α-syn oligomers.13 Similarly, also post-translational
trols.71 Furthermore, in vitro experiments have shown modifications of α-syn alter its clearance. The most
that galactosylsphingosine accelerates aggregation of prominent effects were caused by phosphorylation and
α-syn in a dose-dependent manner.108 exposure to dopamine, whereas oxidation and nitration
Thus, in PD, low activity of GCase, aSMase, and only slightly alter CMA processes.123
GALC, by decreasing Cer levels, may cause a mild reduc- In 2011, Mazzulli and colleagues63 demonstrated that
tion of CTSD activity. As a consequence, the slowdown expression of both WT and A53T α-syn in H4 neuro-
in α-syn degradation may favor its accumulation in cyto- glioma cells, human GD dopaminergic neurons and pri-
plasm, endosomes, and lysosomes.109,110 In lysosomes, mary cortical neurons resulted in a significant decrease
acidic pH further promotes α-syn aggregation.111,112 in GCase activity compared to controls, which pro-
Together with α-syn accumulation, ALP dysfunctions voked substrate accumulation and enlargement of lyso-
have also been demonstrated to promote the exocytosis somes. Conversely, in the same study, expression of a
and cell-to-cell propagation of α-syn aggregates.110,113-118 fibrillization incompetent mutant (Δ71-82 α-syn) did
α-Syn exocytosis has been shown to be related to a not affect GCase lysosomal activity. The cause of the
bafilomycin A1–sensitive autophagy pathway in human decreased activity may be addressed to an impaired
neuroblastoma and primary neuron cell lines113,114 and trafficking of GCase (attributable to the accumulation
by inhibition of macroautophagy in immortalized of α-syn) through the endoplasmic reticulum/Golgi
human neuronal cells.116 Depletion of GCase and the apparatus that impedes its correct localization in the
haploinsufficiency of CTSD in human neuroblastoma lysosome. This mechanism was observed both in H4
also promoted the secretion of vesicles containing cells63 and in SH-SY5Y cells overexpressing α-syn.59
aggregated α-syn.117,118 These exosomes demonstrated Subsequently, in a series of investigations, the direct
to be able to transfer aggregates from one cell to another interaction between monomeric α-syn and GCase was
with high efficiency.114 Moreover, both internalization of demonstrated in vitro.124-126 The GCase-α-syn complex
free exogenous extracellular α-syn aggregates in neuronal was shown to decrease GCase activity in lysosome-like
cells and accumulation in lysosomes has been confirmed environments. However, no information is available
and characterized.110,115 The enhancement of aggregated about the possible direct interaction between GCase
α-syn exocytosis may represent a compensatory mecha- and α-syn fibrils or oligomers.
nism aimed at reducing the intracellular α-syn burden to The inhibition of ALP attributable to WT-α-syn was
prevent neuronal cell death.116 On the other hand, ALP further investigated: by exposing HEK293 mammalian
dysfunctions may also contribute in the progressive cells, and primary cortical neurons from α-syn
spreading of α-syn misfolding pathology by promoting expressing mice, to prefibrillar α-syn aggregates, the
recurring cycles of release and uptake of oligomeric and obtained LB-like structures could not be degraded by
fibrillar α-syn, leading to a prion-like propagation.47,119 the ALP because of failure of autophagosomes clear-
ance.127 Accumulation of α-syn aggregates was also
shown to cause lysosome rupture in human glioblas-
Internalization and Aggregation of Misfolded toma cell lines,109 probably as a consequence of exces-
α-Syn Leads to Dysfunction of the ALP in sive substrate accumulation as well as the pore-forming
Cellular and Animal Models ability of α-syn oligomers.94,128 Recently, α-syn-
As a complementary mechanism mirroring what is triggered ALP dysfunction has been reported also by
described in the previous section, the internaliza- Hoffmann and colleagues.110 According to this investi-
tion110,115 and/or accumulation of misfolded α-syn may gation, ALP can be impaired by exposure to exogenous

38 Movement Disorders, Vol. 35, No. 1, 2020


α- S Y N U C L E I N A N D A U T O P H A G I C - L Y S O S O M A L V I C I O U S C Y C L E

oligomeric and fibrillar α-syn both in human glioblas- neurons located in the SN as well as the axon terminals
toma H4 cells and in rat primary neurons. Accumulation located in the striatum.45
of α-syn aggregates within the lysosomal environment Recently, a mitochondrial pyruvate carrier (MPC) inhibi-
resulted in reduced CTSD activity and enlargement of tor, MSDC-0160, has been also tested in different models of
lysosomes. As hypothesized in the previous paragraph, in PD.148 In an α-syn-based Caenorhabditis elegans model,
GBA1-, GALC-, and SMPD1-related PD, decreased MSDC-0160 reduced α-syn toxicity, preventing neuronal
CTSD activity may also be an indirect effect, given that loss. In mice treated with 1-methyl-4-phenylpyridinium
the lack of Cer may compromise the activity and (MPP+), MSDC-0160 modulated mTOR signaling and
post-translational maturation of CTSD.96,97 Taken protected against the insult caused by this neurotoxin.148
together, these findings support the hypothesis that PD Furthermore, in MPTP-treated mice, administration of
onset may be linked to ALP dysfunction, leading to α-syn MSDC-0160 protected nigrostriatal neurons, reduced neu-
accumulation/aggregation and vice versa. roinflammation, and improved locomotor behavior. Similar
results were also obtained in a slowly progressive Engrailed1
(En1+/−) genetic mouse model of PD, where the long-term
targeting of MPC preserved motor function, rescued the
The ALP as a Possible Therapeutic nigrostriatal pathway, and reduced neuroinflammation.148
Target For PD Some studies have also investigated the effects of
Abelson tyrosine kinase (c-Abl) inhibition on Beclin-
Several compounds activating the autophagic cascade 1-mediated autophagic α-syn clearance, throughout
have been shown to reduce α-syn toxicity in cellular treatment with Nilotinib, which is usually used in
and animal models.129 Interesting results have been patients with Philadelphia chromosome–positive chronic
obtained by using trehalose, a disaccharide able to acti- myelogenous leukemia.149,150 Nilotinib appeared to be
vate autophagy processes130-132 and regulate the activ- potentially effective in treating motor and nonmotor
ity of TFEB, the main transcriptional regulator of the symptoms in patients with PD and DLB.151,152 Cur-
coordinated lysosomal expression and regulation net- rently, two trials investigating safety, pharmacokinetics,
work.133,134 The therapeutic effects of trehalose have and pharmacodynamics of Nilotinib in patients affected
been demonstrated in several in vitro and in vivo by PD and PD with dementia (ClinicalTrials.gov ID:
models of PD and other neurodegenerative dis- NCT02954978), as well as the ability of this molecule to
eases.110,133,135-140 Hoffman and colleagues showed pass the blood–brain barrier and to reach its target, are
that exposure of H4 cells and primary neurons to treha- ongoing (ClinicalTrials.gov ID:NCT03205488).
lose, before α-syn exposure, prevents lysosomal enlarge- Given the growing number of findings reporting
ment and formation of massive α-syn aggregates.110,141 decreased GCase activity as a possible cause of
In rats overexpressing A53T α-syn, oral administration synucleinopathy and neuronal dysfunction observed in
of trehalose significantly reduced α-syn accumulation PD, this enzyme got attention as a possible therapeutic
and aggregation in the nigrostriatal system and miti- target. Different studies in mouse models of PD showed
gated motor asymmetry142 and dopaminergic neurons that augmentation of GCase activity, throughout
neurodegeneration.139 Similar effects were observed in adeno-associated virus–mediated expression of GCase,
a neurotoxin MPTP-induced PD mouse model.140 reduced α-syn accumulation and protected from α-syn-
Recently, trehalose has been tested in parallel on rat induced neurodegeneration by enhancing lysosomal
and nonhuman primate PD models overexpressing activity.153,154 However, development of GBA1 gene
A53T α-syn.143 In both models, trehalose prevented the therapy as a therapeutic approach for PD must be still
development of striatal dopamine loss and behavioral deeper investigated before its clinical use would be
deficits. Considering the safety of trehalose in possible.
humans144 and its long half-life in the brain, where Small molecular chaperones able to increase GCase
levels of trehalase are low,145 further investigations activity are also under investigation. Ambroxol is a
aimed at evaluating the efficacy of this disaccharide in mucolytic agent approved by the European Medicines
PD treatment and/or prevention are recommended. Agency, which is able to stabilize and increase GCase
The therapeutic potential of overexpressing proteins activity by increasing the levels of LIMP-2 and Saposin-
actively involved in the ALP has also been evaluated. C (GCase cofactor).155 Ambroxol has been shown to
Both the overexpression and chemical activation of improve α-syn clearance also through the indirect acti-
TFEB in H4 cells and in rats overexpressing α-syn vation of TFEB.156 To date, an increasing number of
decreased the formation of α-syn oligomers146 and studies have been carried out in different cellular and
protected midbrain dopaminergic neurons from α-syn animal models to support the therapeutic potential of
toxicity.147 Similarly, overexpression of LAMP2 in rats Ambroxol as a disease-modifying treatment for syn-
and cellular models of PD restored CMA, reduced ucleinopathies.157-159 Furthermore, two phase 2 clinical
α-syn aggregated species, and increased the viability of trials evaluating the safety and efficacy of this drug in

Movement Disorders, Vol. 35, No. 1, 2020 39


B E L L O M O E T A L

PD and PD with dementia patients are currently ongo- References


ing (ClinicalTrials.gov ID: NCT02941822 and
1. Spillantini MG, Schmidt ML, Lee VMY, Trojanowski JQ, Jakes R,
NCT02914366).159 Other molecular chaperones, such Goedert M. α-Synuclein in Lewy bodies. Nature 1997;388:
as Isofagomine, NCGC758, and NCGC607, seem capa- 839–840.
ble to increase GCase activity. However, further studies 2. Shahmoradian SH, Lewis AJ, Genoud C, et al. Lewy pathology in
are needed in order to define their actual efficacy in PD Parkinson’s disease consists of crowded organelles and lipid mem-
branes. Nat Neurosci 2019;22:1099–1109.
patients.160-162
3. Osterberg VR, Spinelli KJ, Weston LJ, Luk KC, Woltjer RL,
Given that accumulation of glycosphingolipids seems to Unni VK. Progressive aggregation of alpha-synuclein and selective
influence α-syn aggregation,91 substrate reduction has degeneration of lewy inclusion-bearing neurons in a mouse model
of parkinsonism. Cell Rep 2015;10:1252–1260.
been suggested as a therapeutic approach alternative to
GCase activation. The use of inhibitors able to block the 4. Rey NL, George S, Steiner JA, et al. Spread of aggregates after
olfactory bulb injection of α-synuclein fibrils is associated with
activity of the enzyme, glucosylceramide synthase (GCS), early neuronal loss and is reduced long term. Acta Neuropathol.
reduces accumulation of glucosylceramide attributed to 2018;135:65–83.
GBA1 mutations, reduces accumulation of aggregated 5. Conway KA, Lee SJ, Rochet JC, Ding TT, Williamson RE,
α-syn, and ameliorates behavioral outcomes in mouse Lansbury PT. Acceleration of oligomerization, not fibrillization, is
a shared property of both α-synuclein mutations linked to early-
models of synucleinopathy.163 A phase II, double-blind, onset Parkinson’s disease: Implications for pathogenesis and ther-
placebo-controlled study to assess the safety of the GCS apy. Proc Natl Acad Sci U S A 2000;97:571–576.
antagonist, Venglustat (GZ/SAR402671), in early-stage 6. Volles MJ, Lee SJ, Rochet JC, et al. Vesicle permeabilization by
PD patients carrying the GBA1 mutation is ongoing protofibrillar alpha-synuclein: implications for the pathogenesis
and treatment of Parkinson’s disease. Biochemistry 2001;40:
(ClinicalTrials.gov ID: NCT02906020).164 This trial will 7812–7819.
help in understanding whether GCS inhibition may repre- 7. Ingelsson M. Alpha-synuclein oligomers-neurotoxic molecules in
sent a suitable strategy for treating PD. Parkinson’s disease and other Lewy body disorders. Front Neurosci
2016;10:408.
8. Jackson MP, Hewitt EW. Cellular proteostasis: degradation of
misfolded proteins by lysosomes. Essays Biochem 2016;60:
Conclusions and Perspectives 173–180.
9. Karimi-Moghadam A, Charsouei S, Bell B, Jabalameli MR.
In this review, multiple evidences of the bidirectional Parkinson disease from Mendelian forms to genetic susceptibility:
relation between ALP dysfunction and α-syn aggrega- new molecular insights into the neurodegeneration process. Cell
Mol Neurobiol 2018;38:1153–1178.
tion in PD pathogenesis and progression are exposed.
10. Galluzzi L, Baehrecke EH, Ballabio A, et al. Molecular definitions
Lysosomal proteins have emerged as promising thera- of autophagy and related processes. EMBO J. 2017;36:1811–1836.
peutic targets for PD and other synucleinopathies. 11. Vega-Rubin-de-Celis S, Peña-Llopis S, Konda M, Brugarolas J.
However, the causal link between mutations on GBA1, Multistep regulation of TFEB by MTORC1. Autophagy 2017;13:
SMPD1, GALC, ASAH1, GLA, and SCARB2 and 464–472.
α-syn accumulation and aggregation is still unclear. In 12. Settembre C, Zoncu R, Medina DL, et al. A lysosome-to-nucleus
signalling mechanism senses and regulates the lysosome via mTOR
this review, we considered a hypothesis that takes into and TFEB. EMBO J 2012;31:1095–1108.
account the effect of Cer, whose levels can be altered by
13. Vogiatzi T, Xilouri M, Vekrellis K, Stefanis L. Wild type alpha-
mutations on some of the cited genes, on the matura- synuclein is degraded by chaperone-mediated autophagy and mac-
tion and activation of CTSD that may result in a roautophagy in neuronal cells. J Biol Chem 2008;283:
23542–23556.
decreased degradation of α-syn. Given the inability of
14. Mak SK, McCormack AL, Manning-Bo g AB, Cuervo AM, Di
the ALP to degrade the toxic species α-syn, formation Monte DA. Lysosomal degradation of α-synuclein in vivo. J Biol
of LB could, in this context, represent a last move of Chem 2010;285:13621–13629.
the cell to store the excess of α-syn and avoid excessive 15. Klein C, Westenberger A. Genetics of Parkinson’s disease. Cold
proteolytic stress.64,165 However, further investigations Spring Harb Perspect Med 2012;2:a008888.
are needed to better clarify these aspects. Vice versa, 16. Kim CY, Alcalay RN. Genetic forms of Parkinson’s disease. Semin
also the mechanisms of GCase inhibition by α-syn olig- Neurol. 2017;37:135–146.

omers and fibrils should be further investigated to 17. Chang D, Nalls MA, Hallgrímsdóttir IB, et al. A meta-analysis of
genome-wide association studies identifies 17 new Parkinson’s dis-
design new therapies and better understand the link ease risk loci. Nat Genet 2017;49:1511–1516.
between lysosomal dysfunctions and sporadic 18. Robak LA, Jansen IE, van Rooij J, et al. Excessive burden of lyso-
PD. Clinical trials with compounds able to restore ALP somal storage disorder gene variants in Parkinson’s disease. Brain
functions are currently ongoing. More in vitro and 2017;140:3191–3203.
in vivo studies trying to push to the limit of the ability 19. Wise AH, Yang A, Naik H, et al. Parkinson’s disease prevalence in
Fabry disease: a survey study. Mol Genet Metab Rep 2018;14:
of lysosomes and lysosomal enzymes to “digest” fibrils 27–30.
and oligomers or prevent their formation47 are highly
20. Klein AD, Mazzulli JR. Is Parkinson’s disease a lysosomal disor-
recommended in order to produce further evidence for der? Brain 2018;141:2255–2262.
the potential of ALP activation as a therapeutic target 21. Creed RB, Goldberg MS. Analysis of α-synuclein pathology in
for PD treatment. PINK1 knockout rat brains. Front Neurosci 2018;12:1034.

40 Movement Disorders, Vol. 35, No. 1, 2020


α- S Y N U C L E I N A N D A U T O P H A G I C - L Y S O S O M A L V I C I O U S C Y C L E

22. Xu CY, Kang WY, Chen YM, et al. DJ-1 Inhibits α-synuclein 45. Xilouri M, Brekk OR, Landeck N, et al. Boosting chaperone-
aggregation by regulating chaperone-mediated autophagy. Front mediated autophagy in vivo mitigates α-synuclein-induced neu-
Aging Neurosci 2017;9:308. rodegeneration. Brain 2013;136:2130–2146.
23. Stefanis L. α-Synuclein in Parkinson’s disease. Cold Spring Harb 46. Cuervo AM, Stefanis L, Fredenburg R, Lansbury PT, Sulzer D.
Perspect Med 2012;2:a009399. Impaired degradation of mutant alpha-synuclein by chaperone-
24. Kojovic M, Sheerin UM, Rubio-Agusti I, et al. Young-onset parkin- mediated autophagy. Science 2004;305:1292–1295.
sonism due to homozygous duplication of α-synuclein in a consan- 47. Sacino AN, Brooks MMT, Chakrabarty P, et al. Proteolysis of
guineous family. Mov Disord 2012;27:1827–1829. α-synuclein fibrils in the lysosomal pathway limits induction of
25. Mutez E, Leprêtre F, Le Rhun E, et al. SNCA locus duplication car- inclusion pathology. J Neurochem 2017;140:662–678.
riers: from genetics to Parkinson disease phenotypes. Hum Mutat 48. McGlinchey RP, Lee JC. Cysteine cathepsins are essential in lyso-
2011;32:E2079–E2090. somal degradation of α-synuclein. Proc Natl Acad Sci U S A 2015;
26. Murphy DD, Rueter SM, Trojanowski JQ, Lee VM. Synucleins are 112:9322–9327.
developmentally expressed, and alpha-synuclein regulates the size 49. Zhang NY, Tang Z, Liu CW. α-synuclein protofibrils inhibit 26 S
of the presynaptic vesicular pool in primary hippocampal neurons. proteasome-mediated protein degradation: Understanding the cyto-
J Neurosci 2000;20:3214–3220. toxicity of protein protofibrils in neurodegenerative disease patho-
27. Cooper AA, Gitler AD, Cashikar A, et al. α-Synuclein blocks genesis. J Biol Chem 2008;283:20288–20298.
ER-Golgi traffic and Rab1 rescues neuron loss in Parkinson’s 50. Schapira AHV. Glucocerebrosidase and Parkinson disease: recent
models. Science 2006;313:324–328. advances. Mol Cell Neurosci 2015;66:37–42.
28. Alim MA, Ma QL, Takeda K, et al. Demonstration of a role for 51. Thomas B, Beal MF. Parkinson’s disease. Hum Mol Genet 2007;
alpha-synuclein as a functional microtubule-associated protein. 16:R183–R194.
J Alzheimers Dis 2004;6:435–442; discussion, 443–449.
52. Del Tredici K, Braak H. Review: sporadic Parkinson’s disease:
29. Cartelli D, Aliverti A, Barbiroli A, et al. α-Synuclein is a novel
development and distribution of α-synuclein pathology. Neuro-
microtubule dynamase. Sci Rep 2016;6:33289.
pathol Appl Neurobiol 2016;42:33–50.
30. Gan-Or Z, Dion PA, Rouleau GA. Genetic perspective on the role
of the autophagy-lysosome pathway in Parkinson disease. 53. Balducci C, Pierguidi L, Persichetti E, et al. Lysosomal hydrolases
Autophagy 2015;11:1443–1457. in cerebrospinal fluid from subjects with Parkinson’s disease. Mov
Disord 2007;22:1481–1484.
31. di Domenico A, Carola G, Calatayud C, et al. Patient-specific
iPSC-derived astrocytes contribute to non-cell-autonomous neu- 54. Parnetti L, Balducci C, Pierguidi L, et al. Cerebrospinal fluid
rodegeneration in Parkinson’s disease. Stem Cell Reports 2019;12: β-glucocerebrosidase activity is reduced in Dementia with Lewy
213–229. Bodies. Neurobiol Dis 2009;34:484–486.

32. Ho DH, Kim H, Nam D, et al. LRRK2 impairs autophagy by 55. Parnetti L, Chiasserini D, Persichetti E, et al. Cerebrospinal fluid
mediating phosphorylation of leucyl-tRNA synthetase. Cell Bio- lysosomal enzymes and alpha-synuclein in Parkinson’s disease.
chem Funct 2018;36:431–442. Mov Disord 2014;29:1019–1027.

33. Orenstein SJ, Kuo SH, Tasset I, et al. Interplay of LRRK2 with 56. Parnetti L, Paciotti S, Eusebi P, et al. Cerebrospinal fluid
chaperone-mediated autophagy. Nat Neurosci 2013;16:394–406. β-glucocerebrosidase activity is reduced in Parkinson’s disease
patients. Mov Disord 2017;32:1423–1431.
34. Sidransky E, Nalls MA, Aasly JO, et al. Multicenter analysis of
glucocerebrosidase mutations in Parkinson’s disease. N Engl J Med 57. Alcalay RN, Wolf P, Levy OA, et al. Alpha galactosidase A activity
2009;361:1651–1661. in Parkinson’s disease. Neurobiol Dis 2018;112:85–90.
35. Anheim M, Elbaz A, Lesage S, et al. Penetrance of Parkinson dis- 58. Wu G, Yan B, Wang X, et al. Decreased activities of lysosomal acid
ease in glucocerebrosidase gene mutation carriers. Neurology alpha-D-galactosidase A in the leukocytes of sporadic Parkinson’s
2012;78:417–420. disease. J Neurol Sci 2008;271:168–173.
36. Choi JH, Stubblefield B, Cookson MR, et al. Aggregation of 59. Gegg ME, Burke D, Heales SJR, et al. Glucocerebrosidase defi-
α-synuclein in brain samples from subjects with glucocerebrosidase ciency in substantia nigra of Parkinson disease brains. Ann Neurol
mutations. Mol Genet Metab 2011;104:185–188. 2012;72:455–463.
37. Alcalay RN, Levy OA, Waters CC, et al. Glucocerebrosidase activ- 60. Murphy KE, Gysbers AM, Abbott SK, et al. Reduced
ity in Parkinson’s disease with and without GBA mutations. Brain glucocerebrosidase is associated with increased α-synuclein in spo-
2015;138:2648–2658. radic Parkinson’s disease. Brain 2014;137:834–848.
38. Thaler A, Gurevich T, Bar Shira A, et al. A “dose” effect of muta- 61. Moors TE, Paciotti S, Ingrassia A, et al. Characterization of brain
tions in the GBA gene on Parkinson’s disease phenotype. Parkin- lysosomal activities in GBA-related and sporadic Parkinson’s dis-
sonism Relat Disord 2017;36:47–51. ease and dementia with Lewy bodies. Mol Neurobiol 2019;56:
39. Michelakakis H, Xiromerisiou G, Dardiotis E, et al. Evidence of an 1344–1355.
association between the scavenger receptor class B member 2 gene 62. Chiasserini D, Paciotti S, Eusebi P, et al. Selective loss of
and Parkinson’s disease. Mov Disord 2012;27:400–405. glucocerebrosidase activity in sporadic Parkinson’s disease and
40. Hopfner F, Schulte EC, Mollenhauer B, et al. The role of SCARB2 dementia with Lewy bodies. Mol Neurodegener 2015;10:15.
as susceptibility factor in Parkinson’s disease. Mov Disord 2013; 63. Mazzulli JR, Xu YH, Sun Y, et al. Gaucher disease
28:538–540. glucocerebrosidase and α-synuclein form a bidirectional pathogenic
41. Alcalay RN, Levy OA, Wolf P, et al. SCARB2 variants and loop in synucleinopathies. Cell 2011;146:37–52.
glucocerebrosidase activity in Parkinson’s disease. NPJ Parkinsons 64. Chu Y, Dodiya H, Aebischer P, Olanow CW, Kordower JH. Alter-
Dis 2016;2:16004. ations in lysosomal and proteasomal markers in Parkinson’s dis-
42. Zeigler M, Meiner V, Newman JP, et al. A novel SCARB2 muta- ease: relationship to alpha-synuclein inclusions. Neurobiol Dis
tion in progressive myoclonus epilepsy indicated by reduced 2009;35:385–398.
β-glucocerebrosidase activity. J Neurol Sci 2014;339:210–213. 65. Nelson MP, Boutin M, Tse TE, et al. The lysosomal enzyme alpha-
43. Rothaug M, Zunke F, Mazzulli JR, et al. LIMP-2 expression is crit- Galactosidase A is deficient in Parkinson’s disease brain in association
ical for β-glucocerebrosidase activity and α-synuclein clearance. with the pathologic accumulation of alpha-synuclein. Neurobiol Dis
Proc Natl Acad Sci U S A 2014;111:15573–15578. 2018;110:68–81.
44. Webb JL, Ravikumar B, Atkins J, Skepper JN, Rubinsztein DC. 66. Alvarez-Erviti L, Rodriguez-Oroz MC, Cooper JM, et al. Chaper-
α-Synuclein is degraded by both autophagy and the proteasome. one-mediated autophagy markers in Parkinson disease brains. Arch
J Biol Chem 2003;278:25009–25013. Neurol 2010;67:1464–1472.

Movement Disorders, Vol. 35, No. 1, 2020 41


B E L L O M O E T A L

67. Murphy KE, Gysbers AM, Abbott SK, et al. Lysosomal-associated 87. Suzuki M, Fujikake N, Takeuchi T, et al. Glucocerebrosidase defi-
membrane protein 2 isoforms are differentially affected in early ciency accelerates the accumulation of proteinase K-resistant
Parkinson’s disease. Mov Disord 2015;30:1639–1647. α-synuclein and aggravates neurodegeneration in a Drosophila
model of Parkinson’s disease. Hum Mol Genet 2015;24:
68. Dehay B, Bové J, Rodríguez-Muela N, et al. Pathogenic Lysosomal
6675–6686.
depletion in Parkinson’s disease. J Neurosci 2010;30:
12535–12544. 88. Yun SP, Kim D, Kim S, et al. α-Synuclein accumulation and GBA
deficiency due to L444P GBA mutation contributes to MPTP-
69. Crews L, Spencer B, Desplats P, et al. Selective molecular alter-
induced parkinsonism. Mol Neurodegener 2018;13:1.
ations in the autophagy pathway in patients with Lewy body dis-
ease and in models of alpha-synucleinopathy. PLoS One 2010;5: 89. Abul Khair SB, Dhanushkodi NR, Ardah MT, Chen W, Yang Y,
e9313. Haque ME. Silencing of glucocerebrosidase gene in Drosophila
70. Shachar T, Lo Bianco C, Recchia A, Wiessner C, Raas- enhances the aggregation of Parkinson’s disease associated
Rothschild A, Futerman AH. Lysosomal storage disorders and α-synuclein mutant A53T and affects locomotor activity. Front
Parkinson’s disease: Gaucher disease and beyond. Mov Disord Neurosci 2018;12:81.
2011;26:1593–1604. 90. Moraitou M, Dermentzaki G, Dimitriou E, et al. α-Synuclein
71. Marshall MS, Jakubauskas B, Bogue W, et al. Analysis of age- dimerization in erythrocytes of Gaucher disease patients: correla-
related changes in psychosine metabolism in the human brain. tion with lipid abnormalities and oxidative stress. Neurosci Lett
PLoS One 2018;13:e0193438. 2016;613:1–5.

72. Suzuki K, Iseki E, Togo T, et al. Neuronal and glial accumulation 91. Taguchi YV, Liu J, Ruan J, et al. Glucosylsphingosine promotes
of alpha- and beta-synucleins in human lipidoses. Acta Neuro- α-synuclein pathology in mutant GBA-associated Parkinson’s dis-
pathol 2007;114:481–489. ease. J Neurosci 2017;37:9617–9631.

73. Pchelina SN, Nuzhnyi EP, Emelyanov AK, et al. Increased plasma 92. McGlinchey R, Lee J. Effect of glucocerebrosidase and its substrate,
oligomeric alpha-synuclein in patients with lysosomal storage dis- glucosylceramide on α-synuclein aggregation and degradation.
eases. Neurosci Lett 2014;583:188–193. FASEB J 2015;29:564.7.

74. Nuzhnyi E, Emelyanov A, Boukina T, et al. Plasma oligomeric 93. Zunke F, Moise AC, Belur NR, et al. Reversible conformational
alpha-synuclein is associated with glucocerebrosidase activity in conversion of α-synuclein into toxic assemblies by
Gaucher disease. Mov Disord 2015;30:989–991. glucosylceramide. Neuron 2018;97:92-107.e10.
75. Klucken J, Poehler AM, Ebrahimi-Fakhari D, et al. Alpha-synuclein 94. Fusco G, Chen SW, Williamson PTF, et al. Structural basis of mem-
aggregation involves a bafilomycin A1-sensitive autophagy path- brane disruption and cellular toxicity by α-synuclein oligomers. Sci-
way. Autophagy 2012;8:754–766. ence 2017;358:1440–1443.
76. Ahmed I, Liang Y, Schools S, Dawson VL, Dawson TM, Savitt JM. 95. Thibaudeau TA, Anderson RT, Smith DM. A common mechanism
Development and characterization of a new Parkinson’s disease of proteasome impairment by neurodegenerative disease-associated
model resulting from impaired autophagy. J Neurosci 2012;32: oligomers. Nat Commun 2018;9:1097.
16503–16509.
96. Heinrich M, Wickel M, Schneider-Brachert W, et al. Cathepsin D
77. Friedman LG, Lachenmayer ML, Wang J, et al. Disrupted targeted by acid sphingomyelinase-derived ceramide. EMBO J
autophagy leads to dopaminergic axon and dendrite degeneration 1999;18:5252–5263.
and promotes presynaptic accumulation of α-synuclein and LRRK2
97. Liu F, Li X, Lu C, et al. Ceramide activates lysosomal cathepsin B
in the brain. J Neurosci 2012;32:7585–7593.
and cathepsin D to attenuate autophagy and induces ER stress to
78. Tang FL, Erion JR, Tian Y, et al. VPS35 in dopamine neurons is suppress myeloid-derived suppressor cells. Oncotarget 2016;7:
required for endosome-to-Golgi retrieval of Lamp2a, a receptor of 83907–83925.
chaperone-mediated autophagy that is critical for α-synuclein deg-
radation and prevention of pathogenesis of Parkinson’s disease. 98. Gan-Or Z, Liong C, Alcalay RN. GBA-associated Parkinson’s dis-
J Neurosci 2015;35:10613–10628. ease and other synucleinopathies. Curr Neurol Neurosci Rep 2018;
18:44.
79. Follett J, Bugarcic A, Yang Z, et al. Parkinson disease-linked
Vps35 R524W mutation impairs the endosomal association of 99. Mielke MM, Maetzler W, Haughey NJ, et al. Plasma ceramide and
retromer and induces α-synuclein aggregation. J Biol Chem 2016; glucosylceramide metabolism is altered in sporadic Parkinson’s dis-
291:18283–18298. ease and associated with cognitive impairment: a pilot study. PLoS
One 2013;8:e73094.
80. Suzuki K, Iseki E, Katsuse O, et al. Neuronal accumulation of
alpha- and beta-synucleins in the brain of a GM2 gangliosidosis 100. Guedes LC, Chan RB, Gomes MA, et al. Serum lipid alterations in
mouse model. Neuroreport 2003;14:551–554. GBA-associated Parkinson’s disease. Parkinsonism Relat Disord
2017;44:58–65.
81. Sevlever D, Jiang P, Yen SHC. Cathepsin D is the main lysosomal
enzyme involved in the degradation of α-synuclein and generation 101. Kurz J, Parnham MJ, Geisslinger G, Schiffmann S. Ceramides as
of its carboxy-terminally truncated species. Biochemistry 2008;47: novel disease biomarkers. Trends Mol Med 2019;25:20–32.
9678–9687. 102. Abbott SK, Li H, Muñoz SS, et al. Altered ceramide acyl chain
82. Qiao L, Hamamichi S, Caldwell KA, et al. Lysosomal enzyme length and ceramide synthase gene expression in Parkinson’s dis-
cathepsin D protects against alpha-synuclein aggregation and toxic- ease. Mov Disord 2014;29:518–526.
ity. Mol Brain 2008;1:17. 103. Kim MJ, Jeon S, Burbulla LF, Krainc D. Acid ceramidase inhibition
83. Cullen V, Lindfors M, Ng J, et al. Cathepsin D expression level ameliorates α-synuclein accumulation upon loss of GBA1 function.
affects alpha-synuclein processing, aggregation, and toxicity Hum Mol Genet 2018;27:1972–1988.
in vivo. Mol Brain 2009;2:5. 104. Clark LN, Chan R, Cheng R, et al. Gene-wise association of vari-
84. Stoka V, Turk V, Turk B. Lysosomal cathepsins and their regula- ants in four lysosomal storage disorder genes in neu-
tion in aging and neurodegeneration. Ageing Res Rev 2016;32: ropathologically confirmed Lewy body disease. PLoS One 2015;10:
22–37. e0125204.
85. McGlinchey RP, Lacy SM, Huffer KE, Tayebi N, Sidransky E, 105. Dagan E, Schlesinger I, Ayoub M, Mory A, Nassar M, Kurolap A,
Lee JC. C-terminal α-synuclein truncations are linked to cysteine et al. The contribution of Niemann-Pick SMPD1 mutations to
cathepsin activity in Parkinson’s disease. J Biol Chem 2019;294: Parkinson disease in Ashkenazi Jews. Parkinsonism Relat Disord
9973–9984. 2015;21:1067–1071.
86. Bae EJ, Yang NY, Lee C, et al. Loss of glucocerebrosidase 1 activity 106. Gan-Or Z, Ozelius LJ, Bar-Shira A, et al. The p.L302P mutation in
causes lysosomal dysfunction and α-synuclein aggregation. Exp the lysosomal enzyme gene SMPD1 is a risk factor for Parkinson
Mol Med 2015;47:e153. disease. Neurology 2013;80:1606–1610.

42 Movement Disorders, Vol. 35, No. 1, 2020


α- S Y N U C L E I N A N D A U T O P H A G I C - L Y S O S O M A L V I C I O U S C Y C L E

107. Alcalay RN, Mallett V, Vanderperre B, et al. SMPD1 mutations, 128. Stefanovic AND, Stöckl MT, Claessens MMAE, Subramaniam V.
activity, and α-synuclein accumulation in Parkinson’s disease. Mov α-Synuclein oligomers distinctively permeabilize complex model
Disord 2019;34:526–535. membranes. FEBS J 2014;281:2838–2850.
108. Smith BR, Santos MB, Marshall MS, et al. Neuronal inclusions of 129. Moors TE, Hoozemans JJM, Ingrassia A, et al. Therapeutic poten-
α-synuclein contribute to the pathogenesis of Krabbe disease. tial of autophagy-enhancing agents in Parkinson’s disease. Mol
J Pathol 2014;232:509–521. Neurodegener 2017;12:11.
109. Freeman D, Cedillos R, Choyke S, et al. Alpha-synuclein induces 130. Tanji K, Miki Y, Maruyama A, et al. Trehalose intake induces
lysosomal rupture and cathepsin dependent reactive oxygen species chaperone molecules along with autophagy in a mouse model of
following endocytosis. PLoS One 2013;8:e62143. Lewy body disease. Biochem Biophys Res Commun 2015;465:
746–752.
110. Hoffmann AC, Minakaki G, Menges S, et al. Extracellular aggre-
gated alpha synuclein primarily triggers lysosomal dysfunction in 131. Sarkar S, Davies JE, Huang Z, Tunnacliffe A, Rubinsztein DC.
neural cells prevented by trehalose. Sci Rep 2019;9:544. Trehalose, a novel mTOR-independent autophagy enhancer, accel-
erates the clearance of mutant huntingtin and alpha-synuclein.
111. Grey M, Dunning CJ, Gaspar R, et al. Acceleration of α-synuclein J Biol Chem 2007;282:5641–5652.
aggregation by exosomes. J Biol Chem 2015;290:2969–2982.
132. Rodríguez-Navarro JA, Rodríguez L, Casarejos MJ, et al. Treha-
112. Paciotti S, Bellomo G, Gatticchi L, Parnetti L. Are we ready for lose ameliorates dopaminergic and tau pathology in parkin
detecting α-synuclein prone to aggregation in patients? The case of deleted/tau overexpressing mice through autophagy activation.
“protein-misfolding cyclic amplification” and “real-time quaking- Neurobiol Dis 2010;39:423–438.
induced conversion” as diagnostic tools. Front Neurol. 2018;
9:415. 133. Palmieri M, Pal R, Nelvagal HR, et al. mTORC1-independent
TFEB activation via Akt inhibition promotes cellular clearance in
113. Jang A, Lee HJ, Suk JE, Jung JW, Kim KP, Lee SJ. Non-classical neurodegenerative storage diseases. Nat Commun 2017;8:14338.
exocytosis of alpha-synuclein is sensitive to folding states and pro-
134. Rusmini P, Cortese K, Crippa V, et al. Trehalose induces
moted under stress conditions. J Neurochem 2010;113:1263–1274.
autophagy via lysosomal-mediated TFEB activation in models of
114. Alvarez-Erviti L, Seow Y, Schapira AH, et al. Lysosomal dysfunc- motoneuron degeneration. Autophagy 2019;15:631–651.
tion increases exosome-mediated alpha-synuclein release and trans-
135. Hosseinpour-Moghaddam K, Caraglia M, Sahebkar A. Autophagy
mission. Neurobiol Dis 2011;42:360–367.
induction by trehalose: Molecular mechanisms and therapeutic
115. Apetri MM, Harkes R, Subramaniam V, Canters GW, Schmidt T, impacts. J Cell Physiol 2018;233:6524–6543.
Aartsma TJ. Direct observation of α-synuclein amyloid aggregates 136. Emanuele E. Can trehalose prevent neurodegeneration? Insights
in endocytic vesicles of neuroblastoma cells. PLOS One 2016;11: from experimental studies. Curr Drug Targets 2014;15:551–557.
e0153020.
137. Wu F, Xu HD, Guan JJ, et al. Rotenone impairs autophagic flux
116. Fussi N, Höllerhage M, Chakroun T, et al. Exosomal secretion of and lysosomal functions in Parkinson’s disease. Neuroscience
α-synuclein as protective mechanism after upstream blockage of 2015;284:900–911.
macroautophagy. Cell Death Dis 2018;9:757.
138. Kaur S, Nazir A. Potential role of protein stabilizers in ameliora-
117. Bae EJ, Yang NY, Song M, et al. Glucocerebrosidase regulates per- tion of Parkinson’s disease and associated effects in transgenic
petual cell-to-cell transmission of α-synuclein. Nat Commun 2014; Caenorhabditis elegans model expressing alpha-synuclein. RSC
5:4755. Adv 2015;5:77706–77715.
118. Bae EJ, Yang NY, Lee C, Kim S, Lee HJ, Lee SJ. Haploinsufficiency 139. He Q, Koprich JB, Wang Y, et al. Treatment with trehalose pre-
of cathepsin D leads to lysosomal dysfunction and promotes cell- vents behavioral and neurochemical deficits produced in an AAV
to-cell transmission of α-synuclein aggregates. Cell Death Dis α-synuclein rat model of Parkinson’s disease. Mol Neurobiol 2016;
2015;6:e1901. 53:2258–2268.
119. Steiner JA, Angot E, Brundin P. A deadly spread: cellular mecha- 140. Pupyshev AB, Tikhonova MA, Akopyan AA, Tenditnik MV,
nisms of α-synuclein transfer. Cell Death Differ 2011;18: Dubrovina NI, Korolenko TA. Therapeutic activation of
1425–1433. autophagy by combined treatment with rapamycin and trehalose in
a mouse MPTP-induced model of Parkinson’s disease. Pharmacol
120. Winslow AR, Chen CW, Corrochano S, et al. α-Synuclein impairs
Biochem Behav 2019;177:1–11.
macroautophagy: implications for Parkinson’s disease. J Cell Biol
2010;190:1023–1037. 141. Parnetti L, Bellomo G. The issue of waste disposal in Parkinson’s
disease pathogenesis. Mov Disord 2019;34:985–985.
121. Erustes AG, Stefani FY, Terashima JY, et al. Overexpression of
α-synuclein in an astrocyte cell line promotes autophagy inhibition 142. Whishaw IQ, Kolb B (eds). The Behavior of the Laboratory Rat: A
and apoptosis. J Neurosci Res 2018;96:160–171. Handbook with Tests. New York, NY: Oxford University Press;
2005.
122. Xilouri M, Vogiatzi T, Vekrellis K, Park D, Stefanis L. Abberant
alpha-synuclein confers toxicity to neurons in part through inhibi- 143. Howson PA, Johnston TH, Ravenscroft P, Hill MP, Su J,
tion of chaperone-mediated autophagy. PLoS One 2009;4:e5515. Brotchie JM, Koprich JB. Beneficial effects of trehalose on striatal
dopaminergic deficits in rodent and primate models of
123. Martinez-Vicente M, Talloczy Z, Kaushik S, et al. Dopamine- synucleinopathy in Parkinson’s disease. J Pharmacol Exp Ther
modified alpha-synuclein blocks chaperone-mediated autophagy. 2019;369:364–374.
J Clin Invest 2008;118:777–788.
144. Richards AB, Krakowka S, Dexter LB, Schmid H,
124. Yap TL, Gruschus JM, Velayati A, et al. α-Synuclein interacts with Wolterbeek APM, Waalkens-Berendsen DH, et al. Trehalose: a
glucocerebrosidase providing a molecular link between Parkinson review of properties, history of use and human tolerance, and
and Gaucher diseases. J Biol Chem 2011;286:28080–28088. results of multiple safety studies. Food Chem Toxicol 2002;40:
125. Yap TL, Velayati A, Sidransky E, Lee JC. Membrane-bound 871–898.
α-synuclein interacts with glucocerebrosidase and inhibits enzyme 145. Sacktor B. Trehalase and the transport of glucose in the mamma-
activity. Mol Genet Metab 2013;108:56–64. lian kidney and intestine. Proc Natl Acad Sci U S A 1968;60:
126. Yap TL, Jiang Z, Heinrich F, et al. Structural features of 1007–1014.
membrane-bound glucocerebrosidase and α-synuclein probed by 146. Kilpatrick K, Zeng Y, Hancock T, Segatori L. Genetic and chemi-
neutron reflectometry and fluorescence spectroscopy. J Biol Chem. cal activation of TFEB mediates clearance of aggregated α-syn-
2015;290:744–754. uclein. PLoS One 2015;10:e0120819.
127. Tanik SA, Schultheiss CE, Volpicelli-Daley LA, Brunden KR, 147. Decressac M, Mattsson B, Weikop P, Lundblad M, Jakobsson J,
Lee VMY. Lewy body-like α-synuclein aggregates resist degrada- Björklund A. TFEB-mediated autophagy rescues midbrain dopa-
tion and impair macroautophagy. J Biol Chem 2013;288: mine neurons from α-synuclein toxicity. Proc Natl Acad Sci U S A
15194–15210. 2013;110:E1817–E1826.

Movement Disorders, Vol. 35, No. 1, 2020 43


B E L L O M O E T A L

148. Ghosh A, Tyson T, George S, et al. Mitochondrial pyruvate carrier 157. Yang SY, Beavan M, Chau KY, Taanman JW, Schapira AHV. A
regulates autophagy, inflammation, and neurodegeneration in human neural crest stem cell-derived dopaminergic neuronal model
experimental models of Parkinsons disease. Sci Transl Med 2016; recapitulates biochemical abnormalities in GBA1 mutation carriers.
8:368ra174. Stem Cell Reports 2017;8:728–742.
149. Karuppagounder SS, Brahmachari S, Lee Y, Dawson VL, 158. Migdalska-Richards A, Daly L, Bezard E, Schapira AHV.
Dawson TM, Ko HS. The c-Abl inhibitor, nilotinib, protects dopa- Ambroxol effects in glucocerebrosidase and α-synuclein transgenic
minergic neurons in a preclinical animal model of Parkinson’s dis- mice. Ann Neurol 2016;80:766–775.
ease. Sci Rep 2014;4:4874.
159. Migdalska-Richards A, Ko WKD, Li Q, Bezard E, Schapira AHV.
150. Hebron ML, Lonskaya I, Moussa CEH. Nilotinib reverses loss of
Oral ambroxol increases brain glucocerebrosidase activity in a
dopamine neurons and improves motor behavior via autophagic
nonhuman primate. Synapse 2017;71:e21967.
degradation of α-synuclein in Parkinson’s disease models. Human
molecular genetics. Hum Mol Genet 2013;22:3315–3328. 160. Sanchez-Martinez A, Beavan M, Gegg ME, Chau KY,
151. Pagan FL, Hebron ML, Wilmarth B, et al. Pharmacokinetics and Whitworth AJ, Schapira AH. Parkinson disease-linked GBA muta-
pharmacodynamics of a single dose Nilotinib in individuals with tion effects reversed by molecular chaperones in human cell and fly
Parkinson’s disease. Pharmacol Res Perspect 2019;7:e00470. models. Sci Rep 2016;6:31380.
152. Pagan F, Hebron M, Valadez EH, et al. Nilotinib effects in 161. Mazzulli JR, Zunke F, Tsunemi T, Toker NJ, Jeon S, Burbulla LF,
Parkinson’s disease and dementia with Lewy bodies. J Parkinsons et al. Activation of β-glucocerebrosidase reduces pathological
Dis 2016;6:503–517. α-synuclein and restores lysosomal function in Parkinson’s patient
153. Rocha EM, Smith GA, Park E, et al. Glucocerebrosidase gene ther- midbrain neurons. J Neurosci. 2016;36:7693–7706.
apy prevents α-synucleinopathy of midbrain dopamine neurons. 162. Aflaki E, Borger DK, Moaven N, et al. A new glucocerebrosidase
Neurobiol Dis 2015;82:495–503. chaperone reduces α-synuclein and glycolipid levels in iPSC-derived
154. Sardi SP, Clarke J, Viel C, et al. Augmenting CNS dopaminergic neurons from patients with Gaucher disease and par-
glucocerebrosidase activity as a therapeutic strategy for parkinson- kinsonism. J Neurosci 2016;36:7441–7452.
ism and other Gaucher-related synucleinopathies. Proc Natl Acad
163. Sardi SP, Viel C, Clarke J, et al. Glucosylceramide synthase inhibi-
Sci U S A 2013;110:3537–3542.
tion alleviates aberrations in synucleinopathy models. Proc Natl
155. Ambrosi G, Ghezzi C, Zangaglia R, Levandis G, Pacchetti C, Acad Sci U S A 2017;114:2699–2704.
Blandini F. Ambroxol-induced rescue of defective
glucocerebrosidase is associated with increased LIMP-2 and 164. Judith Peterschmitt M, Gasser T, Isaacson S, et al. Safety, tolerabil-
saposin C levels in GBA1 mutant Parkinson’s disease cells. Neuro- ity and pharmacokinetics of oral venglustat in Parkinson disease
biol Dis 2015;82:235–242. patients with a GBA mutation. Mol Genet Metab 2019;126:S117.
156. McNeill A, Magalhaes J, Shen C, et al. Ambroxol improves lyso- 165. Olanow CW, Perl DP, DeMartino GN, McNaught KSP. Lewy-
somal biochemistry in glucocerebrosidase mutation-linked body formation is an aggresome-related process: a hypothesis. Lan-
Parkinson disease cells. Brain 2014;137:1481–1495. cet Neurol 2004;3:496–503.

44 Movement Disorders, Vol. 35, No. 1, 2020

You might also like