Subscriber access provided by Nottingham Trent University
Review
A Cleaning Crew: The Pursuit of Autophagy in Parkinson’s Disease Pathik Parekh, Nishant Sharma, Anagha Gadepalli, Abhishek Shahane, Monika Sharma, and Amit Khairnar ACS Chem. Neurosci., Just Accepted Manuscript • DOI: 10.1021/acschemneuro.9b00244 • Publication Date (Web): 06 Aug 2019 Downloaded from pubs.acs.org on August 7, 2019
Just Accepted “Just Accepted” manuscripts have been peer-reviewed and accepted for publication. They are posted online prior to technical editing, formatting for publication and author proofing. The American Chemical Society provides “Just Accepted” as a service to the research community to expedite the dissemination of scientific material as soon as possible after acceptance. “Just Accepted” manuscripts appear in full in PDF format accompanied by an HTML abstract. “Just Accepted” manuscripts have been fully peer reviewed, but should not be considered the official version of record. They are citable by the Digital Object Identifier (DOI®). “Just Accepted” is an optional service offered to authors. Therefore, the “Just Accepted” Web site may not include all articles that will be published in the journal. After a manuscript is technically edited and formatted, it will be removed from the “Just Accepted” Web site and published as an ASAP article. Note that technical editing may introduce minor changes to the manuscript text and/or graphics which could affect content, and all legal disclaimers and ethical guidelines that apply to the journal pertain. ACS cannot be held responsible for errors or consequences arising from the use of information contained in these “Just Accepted” manuscripts.
is published by the American Chemical Society. 1155 Sixteenth Street N.W., Washington, DC 20036 Published by American Chemical Society. Copyright © American Chemical Society. However, no copyright claim is made to original U.S. Government works, or works produced by employees of any Commonwealth realm Crown government in the course of their duties.
Page 1 of 37 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
ACS Chemical Neuroscience
1
+-Full Title: A Cleaning Crew: The Pursuit of Autophagy in Parkinson’s Disease
2
Author names: Pathik Parekh1, Nishant Sharma1, Anagha Gadepalli1, Abhishekh Sahane1, Monika
3
Sharma1, Amit Khairnar1*
4
Author Affiliations:
5
1
6
Research (NIPER), Ahmedabad, Gandhinagar, Gujarat, India.
7
*Corresponding
8
Amit Khairnar, Ph. D
9
Assistant Professor,
Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and
author:
10
National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad,
11
Palaj, Gandhinagar-382355, Gujarat, India.
12
Phone: +91-9284349396
13
Email:
[email protected],
[email protected] 14 15 16 17 18 19 20 21 22 23 24 25
ACS Paragon Plus Environment
ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
1
Abbreviations:
2
ATG – Autophagy-related genes
3
Hsc70 – Heat shock cognate 70KDa protein
4
CHIP - Carboxy-terminus of Hsc70-interacting protein
5
AP- Autophagosome
6
ER- Endoplasmic Reticulum
7
ULK1- Unc-51 Like Autophagy Activating Kinase 1
8
FIP200- Focal adhesion kinase family-interacting protein of 200 kDa
9
VPS34 - Vacuolar protein sorting 34
10
PI3KC3 - Class-III Phosphatidyl-inositol-3-kinase-catalytic subunit-3
11
LC3I/II – Light chain kinase 3-I/II
12
CMA- Chaperone-mediated autophagy
13
MA- Macroautophagy
14
ALP- Autophagy-lysosomal pathway
15
PFFs – Pre-formed fibrils
16
KO – Knock-out
17
Rab1 – Ras superfamily of small G protein-1
18
ATP13A2 – Type 5 P-type ATPase
19
DLB – Dementia with Lewy-bodies
20
Bcl-2 – B-cell lymphoma 2
21
WT – Wild-type
22
WIPI2 – WD repeat domain phosphoinositide-interacting protein 2
23
MAPK/ERK – Mitogen-activated protein kinase/ Extracellular signal-regulated kinase
24
mTOR- Mammalian target of Rapamycin
25
CaMKK-β – Calcium/calmodulin-dependent protein kinase kinase- β
26
AMPK- 5'-adenosine monophosphate-activated protein kinase
27
Keap-1 – Kelch Like ECH Associated Protein-1
28
GSK3ß – Glycogen synthase kinase 3- ß
29
MPTP – 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine
ACS Paragon Plus Environment
Page 2 of 37
Page 3 of 37 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
1
6-OHDA – 6-Hydroxydopamine
2
TFEB – Transcription factor-EB
ACS Chemical Neuroscience
3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25
ACS Paragon Plus Environment
ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Page 4 of 37
1
Abstract:
2
Parkinson’s disease (PD) is the 2ndmost common neurodegenerative disorder, neuropathologically
3
depicted by the aggregation of misfolded α-synuclein (α-syn) protein and appears to be central to the
4
onset and progression of PD pathology. Evidence from pioneering studies has highly advocated the
5
existence of impaired autophagy pathways in the brains of Parkinson’s disease patients. Autophagy is
6
an evolutionary conserved, homeostatic machinery for minimizing abnormal protein aggregates and
7
for facilitating organelle turnover. Any aberration in constitutive autophagy activity results in the
8
aggregation of misfolded α-syn which in turn, may further inhibit their own degradation- leading to a
9
vicious cycle of neuronal death. Despite the plethora of available literature, there are still lacunas
10
existing in our understanding related to the exact cellular interplay between autophagy impairment and
11
α-syn accumulation mediated neurotoxicity. In this context, clearance of aggregated α-syn via
12
upregulation of the autophagy-lysosomal pathway could provide a pharmacologically viable approach
13
in the treatment of PD. This present review highlights about basics of autophagy, detrimental crosstalk
14
of α-syn and chaperone-mediated autophagy, α-syn and macroautophagy and it also depicts the
15
interaction between α-syn and novel targets: LRRK2 and mTOR, followed by the role of autophagy in
16
PD from the therapeutic perspective. More importantly, it further updates the reader’s understanding
17
on various newer therapeutic avenues which may provide disease modification via promoting
18
clearance of toxic α-syn through activation of autophagy.
19
Keywords: Parkinson’s disease, Autophagy, α-synuclein, LRRK2, Chaperone-mediated autophagy,
20
Macroautophagy, Neurotoxicity
21 22 23 24 25 26 27 28 29
ACS Paragon Plus Environment
Page 5 of 37 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
ACS Chemical Neuroscience
1
1. Introduction:
2
Parkinson’s disease (PD) is the 2nd-most common neurodegenerative disorder preceded by
3
Alzheimer’s disease, affecting 1 - 2% population over the age of 65 years[1]. Typically, it is
4
characterized by progressive loss of specific sub-region of dopaminergic (DAnergic) neurons in
5
substantia nigra-pars compacta (SNpc) of the brain which in turn give rise to the cardinal motor
6
features of PD i.e. resting tremor, bradykinesia, muscle rigidity, and postural instability[2]. This, of
7
course, is an oversimplified view of the disorder. In actuality, PD patients also suffer from a
8
constellation of non-motor symptoms beyond cardinal motor symptoms which includes hyposmia,
9
autonomic dysfunction, depression, sleep disturbances, cognitive abnormalities and gastrointestinal
10
dysfunction[3]. Today, it is well-accepted fact that non-motor symptoms occur due to extensive extra-
11
nigral pathology that appears approximately 20 years before the clinical onset of PD motor
12
symptoms[4].
13
At the cellular level, neuronal loss is accompanied by aggregation of misfolded α-syn within the
14
intracytoplasmic inclusions known as Lewy bodies[5]. α-synuclein (α-syn) is a monomeric protein
15
encoded by SNCA gene which has been found to play an important role in synaptic plasticity and
16
vesicle trafficking [6, 7]. Till date, more than 22 genes have been found to be associated with the PD
17
[8]. Most of the cases of PD are sporadic, only 5% are of familial PD which is associated with
18
autosomal dominant or autosomal recessive mutation[9]. Indeed, identification of autosomal dominant
19
mutations such as A30P, A53T, E46K, G51D, H51Q mutations as well as duplication and triplication
20
in SNCA gene has markedly broadened our current understanding related to the pathogenesis of
21
familial PD [6, 7]. Interestingly, it has been reported that increased levels of a toxic oligomeric form
22
of α-syn (aggregated α-syn) are the pre-requisite for the severity of disease[10]. In addition, there is a
23
large body of evidence suggesting increased expression of α-syn mRNA levels in case of sporadic PD
24
patients in comparison to control. Overall, these observations point to a pivotal involvement of α-syn
25
overexpression to cause genetic PD and, that a subtle aggregation of α-syn may be central to the
26
pathogenesis of PD. As the degradation of α-syn is under the control of cellular cleaning crew that is
27
macroautophagy (MA) and chaperone-mediated autophagy (CMA), any dysfunction in the removal of
28
α-syn might be the one reason responsible for its aggregation. Therefore, enhancing α-syn elimination
29
by autophagy induction may represent a viable therapeutic strategy for the treatment of PD and other
30
synucleopathies.
31
A most common response to the food abstinence is activation of autophagy via Autophagy-Lysosomal
32
Pathway (ALP) in which cell starts to digest its own components in order to recycle the nutrients from
33
dying cells. Autophagy contributes to the maintenance of the nutritional status of the cell in the fasting
34
condition and also helps in the removal or clearance of misfolded proteins or damaged organelles[11].
ACS Paragon Plus Environment
ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Page 6 of 37
1
Christian de Duve, the discoverer of the Lysosome gave birth to the new research field of autophagy
2
and received the Nobel prize in physiology or medicine in 1974[12]. However, the research on
3
autophagy didn’t receive that much attention at that time. In 1993, the laboratory of Yoshinori Ohsumi
4
conducted ground-breaking experiments of genetic analyses of apg mutants in yeast and identified 15
5
APG genes (Autophagy-related genes) involved in autophagy [13, 14]. From that point, research on
6
autophagy was tremendously up taken and the role of autophagy in human health was explored. Along
7
with it, there is unequivocal evidence that suggests impairment in the ALP in PD initiates misfolding
8
of α-syn and dysfunction in protein clearance system converse to form two interconnected concepts
9
related to the cellular mechanism of PD pathology. From the last decade research on PD has been
10
shifted on understanding the mechanism behind the clearance of toxic α-syn via focusing on
11
autophagy. So, by keeping this viewpoint in mind, this review begins with existing knowledge on the
12
complex process of autophagy.
13 14
2. Autophagy
15
Autophagy is an intracellular degradation process which involves degradation of cytoplasmic contents
16
including both misfolded proteins and damaged organelles inside the lysosome or vacuole (in case of
17
yeast) [15]. It is followed by subsequent re-entry of breakdown products into the cytosol in order to
18
promote recycling of the cellular constituents and to get energy from it and maintain the viability of
19
the cells under stressful conditions [16]. Thus, the autophagy is important in two aspects: first is the
20
removal of toxic waste from the cells and second is to promote the recycling of nutrients from damaged
21
organelles and proteins [17]. Based on the method of delivery of cargo to the lysosome, autophagy can
22
be categorized into three classes: Chaperone-mediated autophagy (CMA), Macroautophagy (MA),
23
Microautophagy.
24
Autophagy was considered as nonselective “in bulk” degradation process before the discovery of the
25
CMA. Discovery of CMA made the first evidence that autophagy may be selective as only proteins
26
having KFERQ motif (about 40% of proteins) can be selectively degraded by CMA[18]. Basically,
27
any misfolded proteins like α-syn having KFERQ motif are identified by chaperone cytosolic Hsc70
28
and co-chaperones Hsp70-interacting protein (CHIP), Hsp40, Hsp70-Hsp90 organizing protein (HOP)
29
[19]. Once identified they form complex and then targeted to the lysosomal surface where it will bind
30
to the lysosomal surface receptor, LAMP2A (Lysosome-associated membrane protein 2A). This is the
31
first identified component of lysosome which is involved in the CMA[20]. LAMP2 mRNA undergoes
32
alternative splicing and thus exists as three isoforms: LAMP2A, LAMP2B, LAMP2C[21] but, the
33
LAMP2A is selectively involved in the autophagy [22]. All isoforms differ in their luminal, cytosolic
ACS Paragon Plus Environment
Page 7 of 37 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
ACS Chemical Neuroscience
1
and transmembrane region[23] and further gets internalized by the lysosome and promote its
2
degradation via lysosomal hydrolases.
3
Macroautophagy (MA) (hereafter referred as autophagy), is preserved intracellular degradation
4
pathway involves directing substrates like damaged or superfluous organelles (mitochondria,
5
peroxisomes), aggregated proteins, aggregation-prone proteins, etc. towards the lysosome via double-
6
membrane vesicles known as autophagosome (AP). Phagophore is cup-shaped double-membrane
7
vesicle which is the perceivable herald of AP. Edges of phagophore extend and fuse with each other
8
and lead to the formation of AP. Mechanism of MA has been divided into different stages: initiation,
9
elongation, and maturation of AP. Noteworthy progress has been made on the identification of proteins
10
that controls the biogenesis of AP. Although, how AP is built up of and what could be the source of
11
the AP membrane are prime questions in this area. Many studies advocate relationship between ER
12
and autophagy structures and put forth the hypothesis that membranes of AP are derived from the
13
ER[24]. However, ER is not the only source, but, mitochondria, ER-mitochondrial contact sites, Golgi
14
apparatus, Plasma membrane, and ER-Golgi intermediate compartment are also involved in the
15
biogenesis of AP as they supply lipids to the growing isolation membrane[25] and its underlying
16
molecular mechanism is still unclear. There are lot many genes as well as proteins which are involved
17
in the biogenesis of AP, most of them are conserved from yeast to human. Atg1 is the autophagy-
18
related gene-1 which is also known as ULK1 in mammals and reported to form complex in
19
combination with Atg13 and FIP200. Further, Hosokawa et. al found that Atg101, anAtg13-binding
20
protein molds tetrameric complex in association with Atg13 and FIP200 and upregulates autophagy
21
towards nutrient deficiency[26]. The activity of ULK1 kinase is essential for recruitment of VPS34
22
(vacuolar protein sorting 34) to the phagophore[27]. VPS34 is class-III PI3KC3 (Class-III
23
Phosphatidyl-inositol-3-kinase-catalytic subunit-3) complex which is required for the formation of the
24
phagophore. After completion of the initiation process, it further leads to elongation of AP, which is
25
mainly controlled by the ubiquitin-like conjugation system. This system consists of Atg7 [ubiquitin-
26
activating-enzyme (E1)-like] and Atg10 [ubiquitin-conjugating enzyme (E2)-like]. Both Atg7 and
27
Atg10 assists in the conjugation of Atg12 with Atg5 and forms complex which ultimately forms
28
another complex Atg12-Atg5 with Atg16L, an E3-ubiquitin ligase and promotes the conversion of
29
LC3 to LC3I and finally LC3I to LC3II causing elongation of AP [28]. Furthermore, maturation of AP
30
involves fabrication of AP throughout the cytoplasm followed by the trafficking of AP by means of
31
dynein machinery on the microtubules to carry AP into the juxtaposition of the lysosome[29]. After
32
fusion of AP with a lysosome, contents of AP will get degraded into free amino acid by lysosomal
33
hydrolases. The primitive function of autophagy is to foster the cells against starvation or any other
34
stress conditions which are preserved from yeast to human. Autophagy has a precious role in health as
ACS Paragon Plus Environment
ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Page 8 of 37
1
well as in several diseases. Defective autophagy may be responsible for the development of pathology
2
in various neurodegenerative disorders. For example, aggregation of Tau protein and α-syn in
3
Alzheimer’s disease and Parkinson’s disease respectively. Refer to the previous work for more detailed
4
molecular mechanisms related to autophagy [28].
5
The idea that membrane of lysosome undergoes invagination to confine cytosolic components to
6
degrade them was put forth in early days and the process is commonly known as microautophagy. The
7
phrase microautophagy is reserved for removal of damaged or misfolded proteins and superfluous
8
organelles that are directly engulfed by lysosomes or vacuoles in case of yeast. In early years it was
9
known that microautophagy is not selective but it was proven that cargo can be distinguished in case
10
of microautophagy also and it gave genesis to new terms like micromitophagy, microlipophagy,
11
micropexophagy. As the link between microautophagy and α-syn is not clearly established, discussing
12
microautophagy is beyond the scope of this article.
13 14
3. α-syn and cleaning crew: single-player and multiple cross-talks
15
3.1.α-syn and CMA: Detrimental cross-talk
16
The cross-talk between α-syn and CMA has been of great interest and witnessed to have detrimental
17
outcomes from the last several years. It has been observed monomers and dimers of α-syn can be easily
18
degraded by CMA however, α-syn oligomers can’t be degraded by CMA.[30]. Both in vivo and in
19
vitro studies have found the increased levels of LAMP2A and decreased levels of α-syn aggregation
20
which supports the notion that proper functioning of CMA is required to mitigate neurotoxicity
21
imparted by α-syn aggregation.[31-33]. Thus, impairment of CMA leads to the formation of α-syn
22
oligomers and development of α-syn pathology[32, 34]. Not only α-syn oligomers but several other
23
factors like a mutation in LRRK2 (leucine-rich repeat kinase 2) and VPS35 (vacuolar protein sorting-
24
associated protein 35) and post-translational modifications (PTMs) also impair CMA and thus leads to
25
α-syn aggregation. Mutations in the leucine-rich repeat kinase 2 (LRRK2) including G2019S mutant
26
inhibited the translocation complex at the lysosome membrane and blocked CMA[35]. PTMs like
27
nitration and oxidation of α-syn promotes mild impairment of CMA. Nevertheless, phosphorylation of
28
α-syn and/or dopamine-induced oligomerization of α-syn completely impairs the CMA[30].
29
Interestingly, it has been reported that activity of VPS35 in DAnergic neurons is essential for
30
endosome-to-Golgi retrieval of LAMP2A, a receptor of CMA that is critical for degradation of α-syn
31
and deficiency as well as mutation of VPS35 lead to impairment of LAMP2A and subsequently α-syn
32
aggregation[36]. Impairment of CMA by overexpression of WT α-syn or A53T-α-syn induces
33
translocation of Myocyte enhancer factor 2D (MEF2D) which is one of the neuronal survival markers
ACS Paragon Plus Environment
Page 9 of 37 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
ACS Chemical Neuroscience
1
from the nucleus to the cytosol and induces neuronal death[37]. Apart from in-vivo studies, clinical
2
studies with post-mortem brains of PD patients also suggested impairment of CMA which can be seen
3
from decreased levels of LAMP2A and Hsc70 in the SN and amygdala, which was correlated with the
4
α-syn aggregation [38]. Moreover, mRNA levels of LAMP2 isoforms and protein expression of
5
LAMP2B, 2C were not altered significantly in the initial stages of PD compared to control. However,
6
protein expression of LAMP2A was significantly up-regulated that has been linked with α-syn
7
aggregation suggesting impairment of CMA in PD[38].
8
Recently research is focused on the role of microRNA (miRNA) in CMA in PD. miRNAs are the
9
conserved non-protein coding sequence that emerged as a key post-transcriptional regulator of gene
10
expression. In general, it has been observed that miRNAs promote degradation of α-syn either directly
11
targeting α-syn or indirectly via activation of CMA (Zhao L et al., 2019). miRNA-133b is found to be
12
involved in the degradation of α-syn through CMA. Kim et. al reported a significant decrease in the
13
level of miRNA-133b in PD patients which is normally present in the DAnergic neurons of the
14
midbrain of healthy person [39]. While most of the other miRNAs (miRNA-320A) were found to
15
inhibit the degradation of α-syn and thus it alters the normal state of protein to aggregated state[40].
16
Alvarez-Erviti, L and group recently with the help of luciferase reporter assay in SHSY5Y cells
17
reported that increased levels of, miRNA-21, miRNA-224, miRNA-373, miRNA-379 caused a
18
reduction in levels of LAMP2A whereas increased levels of miRNA-26b, miRNA-106a, miRNA-301b
19
reduced levels of Hsc70 which was correlated with an increase in the level of α-syn. [41]. Recently,
20
it was reported that miRNA-7 promotes clearance of α-syn aggregates and pre-formed fibrils (PFFs)
21
via activation of CMA [42]. Recent studies reported that inhibition of glucocerebrosidase (GCase), a
22
lysosomal hydrolase lead to α-syn aggregation via blockade of CMA [38]. However, the role of GCase
23
in PD is not only limited to the blockage of CMA. Recently, Gegg et. al has described that inhibition
24
of GCase may alleviate ER stress, mitochondrial dysfunction and neuroinflammation, thus lead to the
25
induction of PD pathology[43]. As we have discussed crosstalk between α-syn and CMA and its
26
detrimental consequences. Now it is quite clear that α-syn aggregation impairs the CMA and vice
27
versa. As inhibition of CMA shifts total burden of clearance of α-syn on MA, for this reason, we will
28
discuss how α-syn interacts with the MA. Please refer Fig.1 for diagrammatic representation.
29 30 31 32 33 34
ACS Paragon Plus Environment
ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Page 10 of 37
1 2 3 4 5 6 7 8 9 10 11 12
Figure 1: Illustration representing crosstalk in between α-syn and chaperone-mediated
13
autophagy (CMA): Under pathological conditions:1) A30P/A53T-mutant α-syn binds with
14
LAMP2A with the high affinity 2) mutated LRRK2 inhibits translocation complex at lysosome surface
15
3) post-translational modifications (PTMs) like nitration and oxidation as well as phosphorylated α-
16
syn directly inhibits CMA 4) deficiency of VPS35 impairs retrieval of LAMP2A while miRNAs
17
21,224,373,379 directly inhibits LAMP2A 5) Exposure of dopamine to WT-α-syn promotes
18
aggregation of α-syn which on turn inhibits LAMP2A 6)miRNA 26b,301b,106a reduces levels of
19
Hsc70 and blocks downstream cascade. Mechanisms 1-6 ultimately impairs CMA and leads to α-syn
20
aggregation. Under physiological conditions: 7) Monomeric α-syn binds with chaperone cytosolic
21
Hsc70 and forms complex which further targeted to LAMP2A, further internalized into the lysosome
22
where α-syn interacts with the lysosomal hydrolase, cathepsin-D and ultimately undergoes breakdown.
23 24
3.2.α-syn and macroautophagy: Deliberating crosstalk
25
Several studies enlightened involvement of oligomeric/fibrillar form of α-syn in the pathogenesis of
26
PD. It is evident that MA is involved in the degradation of the mainly oligomeric or fibrillar form of
27
α-syn. Thus, it becomes foremost to understand how MA is intricated in the clearance of aggregated
28
α-syn. Most importantly both in vitro and in vivo studies suggests that clearance of mutated α-syn is
29
mainly governed by Beclin-1, which is an important component of the MA pathway[44]. Beclin-1 is
30
the autophagic protein which is encoded by the BECN1 gene, which interacts with Bcl-2 and plays an
31
important role in the regulation of autophagy. Till date, various activators, as well as inhibitors, have
32
been identified related to macroautophagy (MA) which are involved in the degradation of α-syn.
33
Besides, the clearance of α-syn is found to be dependent on the type of activators/inhibitors used, few
34
studies suggested MA activator rapamycin and Trehalose selectively targeted E46K mutated α-syn
ACS Paragon Plus Environment
Page 11 of 37 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
ACS Chemical Neuroscience
1
through inhibition of JNK1/Bcl-2 pathway[45]. Eventually, the blockade in this pathway leads to
2
accumulation of oligomeric or fibrillar of α-syn [46]. However, it is still ambiguous whether α-syn is
3
accumulated only due to impairment of clearance of α-syn due to inhibition of MA or not. As exposure
4
of PFFs of α-syn to HEK293 induced α-syn accumulation and finally lead to impairment of MA [47].
5
Exhaustively studies suggest clearance of α-syn via MA relies on its exact conformational state like
6
whether α-syn is in the monomeric, oligomeric or fibrillar state. Ahmed et al developed a transgenic
7
knock out (KO) mice lacking Atg -7, an important autophagic protein that is responsible for the
8
formation of AP. The same study ascertained that KO of Atg-7 leads to impairment in the autophagy,
9
age-related DAnergic neuronal loss in striatum, α-syn accumulation and up-regulation of LRRK-2
10
proteins [48, 49] Not only this but also PTMs in α-syn like sumoylation and phosphorylation are
11
critical factor for its clearance by MA[50-52].
12
As the final destination of cargo in the MA is the lysosomes, so straightway AP containing cargo (here,
13
α-syn) enters into the lysosomes and once inside the lysosomes, it is mostly degraded by lysosomal
14
hydrolase specifically cathepsin-D (CTSD)[53]. The role of CTSD in the degradation of α-syn has
15
been confirmed by a study where they have used CTSD KO mice which showed increased
16
accumulation of oligomeric α-syn [54]. Furthermore, mislocalization of CTSD has been observed due
17
to a mutation in VPS35 which also promoted to the accumulation of α-syn[55]. Intriguingly, a mutation
18
in VPS35 has been already linked to the development of familial PD [56]. Not only CTSD but, VPS35
19
mutation also induces mislocalization of Atg9 and hampers the biogenesis of AP which can cause a
20
decrease in α-syn clearance. [57]. Apart from this, studies with mammalian cells found α-syn
21
accumulation also inhibited Rab1 protein, involved in the localization of Atg9 and ultimately inhibits
22
AP biogenesis[58].PARK9, a lysosomal gene was found to be mutated in autosomal PD which encodes
23
for Type 5 P-type ATPase (ATP13A2) protein which is involved in exosomal biogenesis. Kong SM
24
has reported reduced levels of α-syn aggregation due to elevated expression of ATP13A2 which
25
induced externalization of α-syn through exosomes[59]. Thus, the above study supports the transfer of
26
α-syn from dying neurons to healthy neurons might be occurring through the exosomal secretion of α-
27
syn [60]. Furthermore, it has been evident by the observation that expression of ATP13A2 was found
28
to be elevated in surviving DAnergic neurons of SNpc in sporadic PD suggesting that secreted α-syn
29
through exosomes might be taken up by surviving neurons. [59, 61]. Distinctive studies reported
30
increased secretion of α-syn either mediated by exosomes[62, 63]or by exophagy[64] upon the
31
inhibition of MA or lysosomal dysfunction. Above studies suggest debilitating equilibrium exists
32
between α-syn aggregation and its secretion which is mainly mediated by MA leads to the development
33
of PD pathology.
ACS Paragon Plus Environment
ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Page 12 of 37
1
Multitudinous studies described an increased number of AP in the brains of PD patients[65]. Similarly,
2
increased levels of LC3-II, a marker of AP biogenesis has been observed in PD and DLB patients[66,
3
67]. It is clear from the above studies thatα-syn accumulation might be one reason for increased AP
4
formation. However, it might be possible that lysosomal dysfunction dampens the clearance of α-syn
5
aggregates. High mobility group box 1 (HMGB1) has been identified as a novel target to promote
6
clearance of α-syn via activation of autophagy. Inversely, the α-syn aggregation has also been found
7
to disturb binding of HMGB1 to Beclin1 and promotes binding of Beclin1 to Bcl-2 which ultimately
8
impairs the autophagic process[68, 69].
9
Taken together all studies advocates that α-syn may act as a single-player and multiple roles in the
10
process of modulation of autophagy which relies on the levels of α-syn, the conformation of α-syn
11
whether it is in monomeric form or oligomeric, species whether it is WT or mutated α-syn or cellular
12
environment. We have summarized that instead of the presence of conflicting evidence, there is no
13
doubt about the existence of robust crosstalk in between α-syn and autophagy pathways (MA) and thus
14
may enable the development of therapeutics. Refer Fig.2 for diagrammatic representation.
15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30
Figure 2: Illustration representing the crosstalk in between α-syn and macroautophagy: A)
31
Under physiological conditions: 1) α-syn overexpression/mutation 2) A53T α-syn 3) post-
32
translational modifications like SUMOylation and phosphorylation 4) monomeric α-syn can be
33
effectively cleared out from the cells as it upregulates MA. Under pathological conditions: 1)
34
Deletion of Atg-7 2) E46K α-syn inhibits MA via inhibition of JNK1/Bcl-2 pathway 3) Exposure of
ACS Paragon Plus Environment
Page 13 of 37 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
ACS Chemical Neuroscience
1
pre-formed fibrils of α-syn inhibits MA as it promotes aggregation of WT-α-syn4)α-syn aggregation
2
disturbs binding of HMGB1 to Beclin1 and promotes binding of Beclin1 to Bcl-2 which ultimately
3
impairs the MA 5) α-syn aggregation also inhibits the MA via inhibition of Rab1, protein regulates
4
localization of Atg9 required for biogenesis of AP 6) VPS35 mutation also inhibits AP biogenesis via
5
inhibition of Rab1 7) VPS35 mutation also promotes aggregation of α-syn via mislocalization of
6
cathepsin-D, a lysosomal hydrolase involved in degradation of α-syn and ultimately leads to
7
aggregation of α-syn.
8 9
3.3. α-syn and LRRK2: Partners of crime????
10
Leucine-rich repeat kinase 2 (LRRK2) is one of the most important genetic contributors to the
11
development of PD. Several mutations in LRRK2 has been reported in patients of PD suggests LRRK2
12
undergoes mutation in familial PD. Specifically, mutations in the coding region (G2019S, R1441G,
13
R1441H, R1441C, Y1699C, I2020T) or polymorphism in coding, as well as non-coding region, are a
14
major cause of autosomal dominant PD or sporadic PD respectively[70, 71]. Continuing further, these
15
mutations can be identified due to the presence of pleomorphic characteristic in patients of mutant
16
LRRK2, which generally absents in the patients of sporadic PD[72]. However, it is merely impossible
17
to differentiate patients of LRRK2-associated PD from sporadic PD. LRRK2 has been reported to have
18
multiple roles ranging from autophagy, mitochondrial maintenance, immune system-related activity,
19
protein synthesis, etc. Out of which, the role of LRRK2 in the context of autophagy has been widely
20
studied. LRRK2 is a large, multi-domain protein having both GTPase and kinase activity. LRRK2
21
regulates the process of autophagy through its kinase activity. In that direction, Manzoni et. al studied
22
the role of LRRK2 in autophagy by taking primary fibroblasts from individuals having mutations in
23
the functional domain of LRRK2 and found that LRRK2-mutation impaired autophagy that is evident
24
from the altered ratio of LC3-II/I, p62 and WIPI2 positive puncta, an autophagy marker [73].
25
Interestingly, LRRK2 KO mice showed increased accumulation of α-syn, as the deficiency of LRRK2
26
impairs protein degradation pathways [74]. Moreover, inhibition of LRRK2 was associated with the
27
impaired degradation of AP, as it facilitated downregulation of Rab7, a major component involved in
28
AP degradation [75]. However, this defective degradation of AP may be due to impaired microtubule
29
mediated trafficking of AP towards the lysosome[76]suggests an evident role of LRRK2 and its kinase
30
activity in the process of autophagy [77].
31
Most recurrent mutation involved in LRRK2 that has been seen in 40% of familial PD patients is
32
G2019S, moreover, mutation I2020T also has been reported in the kinase domain of LRRK2. G2019S-
33
LRRK2 upregulates MA via activation of mitogen-activated protein kinase/ extracellular signal-
ACS Paragon Plus Environment
ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Page 14 of 37
1
regulated protein kinase (MAPK/ERK) pathway[78] while in another study mutant-LRRK2 (R1441G,
2
G2019S, Y1699C-LRRK2) has been shown to downregulate MA via inactivation of mTOR[73].
3
LRRK2 has been studied from so many times by using different approaches like knockdown,
4
overexpression of wild-type (WT) or mutant LRRK2 in the different model systems. Firstly
5
overexpression of WT-LRRK2 has been reported to activate MA via activation of CaMKK-β/AMPK
6
(calcium/calmodulin-dependent protein kinase kinase-β/ 5’-Adenosine monophosphate-activated
7
protein kinase) pathway[79]. However, overexpression of G2019S-LRRK2 has been associated with
8
reduced degradation capacity as it reduces lysosomal PH and increased expression of ATP13A2 a gene
9
associated with Parkinsonian syndrome[80]. In addition, Park et. al reported interplay between
10
LRRK2 and p62 and observed that LRRK2 promotes phosphorylation of p62 which weakens the
11
interaction of p62 with keap1 and ultimately upregulates autophagy[81]. Recently, it has been reported
12
that G2019S-LRRK2 impairs autophagy as it mediates phosphorylation of leucyl-tRNΑ-synthetase,
13
the substrate of LRRK2 [82].
14
Above studies suggests the involvement of link between LRRK2 and autophagy in PD. However, the
15
link between α-syn and LRRK2 and how α-syn aggregation affects LRRK2 has been poorly
16
understood. But it has been answered by a study using brains of PD patients carrying LRRK2 mutation,
17
showed the presence of α-syn positive Lewy bodies (LBs) suggests that α-syn aggregation is in part
18
under the control of LRRK2[70] and overexpression of the same in the A53T mutated mice has been
19
reported to enhance cytotoxicity mediated by α-syn aggregation. Inhibition of LRRK2 has also been
20
reported to protect the DAnergic neurons from α-syn mediated toxicity[83]. But, the inhibition
21
mediated by the α-syn aggregation has been shown to be well tolerated and can protect the
22
degeneration of DAnergic neurons for a time period not more than 4-week[84]. Nuclear factor
23
erythroid 2-related factor (Nrf2), a well-known anti-oxidant gene, has been identified as a novel target
24
that mitigates toxicity induced by α-syn and LRRK2 by promoting neuronal protein homeostasis in a
25
time-dependent manner[85]. Nrf2 enhances the accumulation of LRRK2 in the LBs and thus it
26
decreases the levels of mutant LRRK2 in other places of neurons[85]. LRRK2 has been reported to be
27
involved in the propagation of α-syn mediated by phosphorylation of RAB35 and the same has been
28
confirmed by LRRK2 inhibitor resulted in decreased α-syn aggregation[86]. Taken together all studies
29
suggest that LRRK2 is a negative regulator of autophagy and thereby promotes α-syn aggregation,
30
imparts neurotoxicity and provides indications that α-syn and LRRK2 might act as partners of crime.
31 32 33
ACS Paragon Plus Environment
Page 15 of 37 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
ACS Chemical Neuroscience
1
4. The interplay between α-syn and mTOR: Double-edged sword
2
The etiology and pathogenesis behind the PD still have to be uncovered. From so many years, it has
3
been considered in the literature that many signaling mechanisms including mTOR get disturbed in
4
case of PD[87]. Mutant forms of α-syn have been found in familial PD patients [23], as well as oxidized
5
forms of α-syn [40] have been observed in patients of sporadic PD and DLB which are shown to block
6
autophagy by deregulation of mTOR. Indeed, most of the neurotoxins like rotenone, 6-OHDA, MPTP
7
exert its neurotoxicity by arresting the activation of mTOR in PD [88, 89]. On that account, compelling
8
evidence is in support of the neuroprotective role of mTOR activation, in a various model of PD. In
9
that accordance, a separate study done by Choi et al. revealed that phylogenetically conserved Oxi-α
10
(novel activator of mTOR) rescues oxidative stress-induced death of DAnergic neurons by inhibiting
11
autophagy and subsequent accumulation of autophagic vacuoles [90]. Above these studies are in
12
support of the neuroprotective role of mTOR activation.
13
Howbeit, transcriptome analysis in the SN of post mortem brains of well-characterized PD patients
14
suggested increased levels of mTOR throughout the progression of PD pathology that advocates the
15
neurotoxic role of mTOR activation[91]. Likewise, administration of L-DOPA facilitates dopamine
16
D1 receptor-mediated activation of mTOR selectively in the GABAergic medium spiny neurons of the
17
striatum, associated with the development of dyskinesia in the mouse model and is abrogated by
18
mTOR inhibitor rapamycin.[92].Intriguingly, these findings collectively raise the possibility that
19
inhibition of mTOR might able to provide the neuroprotection via activation of autophagy.
20
Interestingly, feeding mTOR inhibitor rapamycin to adult Drosophila produces longevity in
21
Drosophila life span previously exposed to paraquat and the underlying mechanism is the activation
22
of autophagy via inhibition of mTOR [93]. Further, MANF a mesencephalic astrocyte-derived
23
neurotrophic factor showed its neuroprotective and neurorestorative effect selectively to the DAnergic
24
neurons against 6-OHDA-induced neurotoxicity in SH-SY5Y cells at even very low concentration.
25
This potential mechanism was partly executed through the downregulation of p-AMPK and
26
mTOR[94]The research focused on traditional Chinese medicine, Loganin suggested that Loganin
27
protects zebrafish from MPTP-induced neurotoxicity via inhibition of PI3K/Akt/mTOR signaling
28
mechanism[95].
29
Additionally, Pérez-Revuelta, et al. delineate the direct relationship between mTOR, α-syn
30
aggregation and autophagy by using SH-SY5Y and HeLa cells and found that activation of mTOR
31
fosters aggregation of α-syn via inhibition of protein phosphatase 2A (PP2A) [96]. PP2A is an enzyme
32
belongs to a family of phosphatase which promotes phosphorylation of Beclin-1 and aids in autophagy.
33
In value addition, wang et. al perceived that moxibustion lowers the expression of p-mTOR and
34
p70S6K leads to significant improvement in behavioral performance, TH immunoreactivity, and levels
ACS Paragon Plus Environment
ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Page 16 of 37
1
of LC3-II as well as decreased immunoreactivity towards α-syn in SN of rotenone-induced PD rat
2
model[97].MANF facilitates the autophagy-mediated clearance of misfolded α-syn prevents DA
3
neuronal degeneration in human α-synuclein-induced C. elegans PD model. This protective effect was
4
found to be mitigated by silencing 26 autophagy-related genes using RNAi [98]. An explorative study
5
by using uric acid on PC12 cells overexpressing A53T mutant SNCA have found to decrease
6
aggregation of α-syn. This effect might be related to downregulation of mTOR along with upregulation
7
of LC3II and p62. By treatment with 3-BDO (3-benzyl-5-((2-nitrophenoxy) methyl)-dihydrofuran-2-
8
(3H)-one), mTOR activator this effect was found to be reversed [99]. Taken into consideration that
9
interplay between α-syn and mTOR might be threatening and has detrimental consequences. However,
10
it still remains debatable whether activation of mTOR is neurotoxic or neuroprotective. Taken together
11
these lines of evidence suggest that mTOR acts as a double-edged sword which may play a beneficial
12
or detrimental role in PD.
13 14
5. Targeting Autophagy-Lysosomal Pathway (ALP): Implications of Promising targets and
15
therapeutic intervention
16
Pioneering studies have been done to explore the influence of different therapeutic agents on the
17
autophagy-lysosomal pathway (ALP) by using different models of PD. The succeeding section will
18
emphasize on the modulation of autophagy by various therapeutics in the clinical and preclinical
19
studies. These therapeutics have been reported to modulate autophagy by mTOR-dependent pathway,
20
mTOR-independent pathway, and other mechanisms. Key targets for all of these pathways are different
21
i.e. in case of the mTOR-dependent pathway, key targets are mTOR, AMPK, Beclin-1while for
22
mTOR-independent pathway key target is inositol monophosphatase (IMPase). Recent studies on the
23
c-Abl kinase, GSK3ß and Nrf2have significantly gained interest as potential targets.
24
Starting with the mTOR-dependent pathway; Ganoderma Lucidum extract (GLE)(by restored AMPK,
25
mTOR, ULK1 and normalized LC3-II/LC3-I ratio)[100], Resveratrol (by activation go AMPK via
26
SIRT1 which enhances autophagy)[101]and Rosuvastatin[102] fosters autophagy by virtue of
27
activation of AMPK. Drugs like Rapamycin, Moxibustion and Loganin were found to alleviate
28
neurotoxicity in PD based animal models via inhibition of mTOR[92, 95, 97]. Interestingly, Metformin
29
was found to exert neuroprotective effect by using both the above approaches (activation of AMPK
30
and inhibition of mTOR) which resulted in the activation of autophagy. This was reflected by
31
downregulated levels of p-ser129, α-syn and mTOR along with upregulated AMPK and LC3-II
32
levels[96, 103, 104]. Taking into account the activation of Beclin-1, Prolyloligopeptidase (PREP)and
33
its inhibitor KYP-2047was found to ameliorate PD condition through activation of autophagy via
ACS Paragon Plus Environment
Page 17 of 37 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
ACS Chemical Neuroscience
1
activation of Beclin-1[105]. Similar effects using caffeine was found to mitigate α-syn induced defects
2
in the autophagy via activation of Beclin-1[42].
3
Up till now, several therapeutics have been developed to target mTOR-independent pathway focusing
4
on Inositol-monophosphatase (IMPase), responsible for cleavage of inositol-monophosphate (IMP) to
5
inositol (Ins) which ultimately downregulates autophagy. in-vitro and in-vivo studies carried out by
6
using SH-SY5Y, PC12 cells and MPTP-induced mouse model of PD respectively suggests that lithium
7
chloride and valproic acid promotes autophagy via inhibition of IMPase as it is evident from decreased
8
levels of IMPase and increased levels of LC3-II [106-108].
9
Recently, other targets have emerged which plays an important role in the modulation of autophagy
10
i.e. c-Abl, GSK3ß, Nrf2. Among them, c-Abl is activated non-receptor tyrosine kinase Abelson,
11
promotes α-syn aggregation via activation of GSK3ß, in turn, causes phosphorylation of α-syn. In
12
2018, Ren and group demonstrated the role of c-Abl-GSK3β Signalling in MPP+-Induced ALP
13
dysfunction. Study results revealed that inhibition of c-Abl by STI-571 rescued the function of ALP
14
through facilitating the nuclear translocation of TFEB and diminished the phosphorylation of GSK3β
15
at Tyr216 which was previously enhanced under MPP+ treatment in SN4741 cells and in primary
16
midbrain neurons ultimately imparted protection against MPP+-induced neuronal cell death [109].
17
Further continuing with GSK3ß, lithium chloride and 6-Bio, direct inhibitors of GSK3ß has been
18
shown to decrease α-syn aggregation as it inhibits GSK3ß and also activates autophagy that can be
19
seen from increased levels of LC3-II [110, 111]. Using Adeno-associated virus serotype 1/2
20
(AAV1/2)-based rat model of PD, Qing et al highlighted the ability of autophagy enhancer Trehalose
21
and suggested that Trehalose at the concentration of 2% prevents/reverse the α-syn aggregation via
22
activation of autophagy as it increases levels of LC3-II [112]. Completely different, Geraniol has been
23
reported to activate autophagy via activation of ATG genes (ATG5, 7, 12) directly in SK-N-SH cells
24
exposed to rotenone [113]. While ITC-3, Naringin, Curcumin, Safranal, Tanshinone-IIa, Pinostrobin
25
has been reported to promote activate Nrf2, an anti-oxidant enzyme and counteracts neurotoxicity in
26
different models of PD as shown in Table 1. Taking together all the studies suggests that AMPK-
27
mTOR-Beclin1 axis, c-Abl kinase, GSK3β, and Nrf2 might serve as potential therapeutic targets to
28
counteract neurotoxicity induced by α-syn due to their autophagic potential. Refer Fig.3 for
29
diagrammatic representation.
30 31 32 33 34
ACS Paragon Plus Environment
ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Page 18 of 37
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19
Figure 3: Illustration representing molecular mechanisms regulating the process of autophagy
20
and therapeutic agents acting on different targets for promoting autophagy via activation of
21
autophagy pathway:
22
Molecular mechanisms regulating the process of autophagy: A) Amino acid depletion as well as
23
low amount of energy levels can be detected by AMPK through LKB1, activates TSC1/2, can lead to
24
inhibition of mTORC1 facilitates autophagy via activation of Beclin1 and ULK1 and also promotes
25
dephosphorylation of TFEB leads to migrate into the nucleus and promotes biogenesis of lysosomes
26
and AP via promoting transcription of ATG genes and lysosomal genes B) Insulin and growth
27
hormones acts on growth hormone receptors and thereby activates class-I PI3K, can activates Akt and
28
thus activates mTORC1 and inhibits autophagy C) Cellular stress leads to activation of p53 which
29
activates PTEN leads to inhibition of PI3K and activates autophagy D) ROS generation due to stress
30
promotes oligomerization of monomeric α-syn which phosphorylates NRF2 and promotes
31
ubiquitination followed by degradation of NRF2 leads to enhanced levels of ROS, triggers
32
inflammatory cascade and inhibits autophagy E) GSK3β also inhibits the autophagy via
33
phosphorylation of Nrf2.
ACS Paragon Plus Environment
Page 19 of 37 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
ACS Chemical Neuroscience
1
Therapeutic agents acting on different targets for promoting autophagy via activation of
2
autophagy pathway: Therapeutic agents upregulates autophagy via 1) activation of AMPK
3
2)Activation of Beclin-1 3) Inhibition of mTOR 4) Enhancing biogenesis of lysosome via
4
dephosphorylation of TFEB 5) Inhibition of c-Abl involved in activation of GSK3β and thereby
5
inhibits degradation of Nrf2 6) Directly inhibiting GSK3β 7) Activation of Nrf2.
6
Table 1: List of therapeutic agents: modulation of the autophagy pathway Agent
Target
Effect
Animal/
Model
cell line
Induction
Reference
(dose) mTOR-dependent Pathway Ganoderma
AMPK
Improved
locomotor C57BL/6J
lucidum
performance,
extract (GLE)
TH+-neurones in SNpc, increased
(400 mg/kg/d) Ganoderma
increased mice levels
[100]
(20mg/kg)
of
AMPK, mTOR, ULK1 AMPK
Increased
cell Neuro-2a
lucidum
viability,increased
extract (GLE)
expression
of
MPTP (1nM)
[100]
Rotenone
[102]
cells
AMPK,
mTOR, ULK1
(800μg/ml) Rosuvastatin
MPTP
AMPK
Upregulated Beclin-1,
(40μM)
AMPK, SH-SY5Y
LC3-II,
cell cells
viability, increased α-syn
(200nM)
clearance Metformin (50mg/kg)
AMPK
Improved coordination, TH+-neurons,
motor Wildtype elevated and LC3-II, C1k1+/-
AMPK expression
mutant mice
ACS Paragon Plus Environment
MPTP (25mg/kg)
[103]
ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Metformin
AMPK
Page 20 of 37
Increased levels of LC3- Murine
MPP+
II,
(1000μM)
AMPK,
while DAnergic
[103]
decreased expression of MN9D p62 Metformin
AMPK
(100mg/kg)
cells
Attenuated induced
MPTP- AMPK
loss
positive
of
TH- KO mice
MPTP
[104]
(30mg/kg)
neurons,
Restored
DOPAC:DA
ratio, Decreased MPTPinduced gliosis
Metformin
AMPK
(1.5,2 mM) Metformin
Reduced levels of p- SH-SY5Y ser129 α-syn
AMPK
(5g/kg)
α-syn
[96]
cells
Reduced levels of p- C57BL/6J
[96]
ser129 α-syn in mouse mice brain
Resveratrol
AMPK/
Increased
oxidative Cultured
SIRT1
capacity of mitochondria, skin
mediated
cellular
model
ATP
content, fibroblasts
reduced oxidative stress KYP-2047 (PREP inhibitor) (5mg/kg) Glycyrrhizic acid (500μM)
Beclin-1 Reduced
levels
Increased levels of DA,
KO of
PARKIN [105]
syn transgenic mice
Beclin-1 Increased cell viability, SH-SY5Y LC3-II/I, Beclin-1
[101]
of A30P Α-
oligomeric α-syn,
HVA in the striatum
siRNA
cells
6-OHDA
Corticosterone (50μM)
ACS Paragon Plus Environment
and [114]
Page 21 of 37 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
ACS Chemical Neuroscience
Caffeine (1g/L Beclin-1 Reduced in
drinking
p129-α-syn, C57BL/6J
apoptosis,
water)
activation,
microglial mice
A53T
α-syn [42]
(3.25μl)
astrogliosis,
increased LC3II/I ratio, Beclin-1 expression MANF
mTOR/
Increased
mTOR, SH-SY5Y
AMPK
decreased
AMPK, cells
6-OHDA
[94]
decreased apoptosis Loganin
mTOR
Suppression
of PC12 cells MPTP
[95]
PI3K/Akt/mTOR signaling,
decreased
MPTP-induced neurotoxicity Rapamycin
mTORC Decreased levels of p- α-syn
and analogues
1
ser129 α-syn
[96]
overexpre ssing SHSY5Y cells
Rapamycin
mTORC Alleviated
and analogues
1
cell
Rescued
death, SH-SY5Y
rotenone
[92]
L-DOPA cells
induced dyskinesia Rapamycin
mTORC Alleviated cell death
α-syn
and analogues
1
transgenic
[115]
rats Moxibustion
mTOR
Suppresses
Sprague-
mTOR/p70S6K,
Dawley
Activated
autophagy,
rats
increased α-syn clearance
ACS Paragon Plus Environment
Rotenone (2mg/ml, s.c.)
[97]
ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
2-
TFEB
Page 22 of 37
Promotes clearance of α- Transfecte
hydroxypropyl
syn
via
-β-
autophagy
cyclodextrin
activation
[116]
of d Human H4 neuroglio ma
cells
for α-syn
mTOR-independent Pathway Lithium
Inositol
chloride
monoph osphatas
Rotenone (200nM)
SH-SY5Y
Increased cell [106]
cells
viability, reduced
e
mitochondrial dysfunction, increased ration of LC3II/I
Lithium
Inositol
chloride
monoph osphatas
Rotenone (200nM)
SH-SY5Y
Increased cell [106]
cells
viability, reduced
e
mitochondrial dysfunction, increased ration of LC3II/I
Valproic
Inositol
(20mic/ml)
monoph
acid Lithium
and osphatas
MPTP (30mg/kg)
C57BL/6J
Rescues
mice
DAnergic
e
neuronal degeneration,
carbonate
decreased loss
(10mic/ml)
of
ACS Paragon Plus Environment
DOPAC
[107]
Page 23 of 37 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
ACS Chemical Neuroscience
after exposure of MPTP Lithium
Inositol
----------
A53T
& Promotes
[108]
monoph
A30P
clearance
osphatas
mutant α- mutant α-syn,
e
syn
of
expressing PC12 cells Other Mechanisms STI-571 (5μM)
Activate MPP+
SN4741
Reduced
d
cells
expression
nonrece
(200μM)
[109] of
c-Abl and p-
ptor
GSK3ß,
tyrosine
restored
kinase
Autophagy-
Abelson
lysosomal
(c-Abl)
pathway
& GSK3ß Geraniol (60nM)
ATG
Rotenone (100nM)
genes
SK-N-SH
Enhanced cell [113]
cells
viability, reduced ROS, restored MMP, reduced
ER-
stress Lithium
GSK3β
Methamphetamine
PC12 cells Increased cell [110]
chloride Or
viability,
GSK3β
siRNA
ACS Paragon Plus Environment
ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Trehalose (2- -------
AAV A53T- α-syn
Rat
5%)
Page 24 of 37
Increased
[112]
levels of LC3II,
reduced
levels of α-syn aggregates 6-Bio
GSK3β
MPP+
(50μM)
SH-SY5Y
Reduced levels [111]
cells
of
p-GSK3β,
enhanced LC3II, activation of autophagy, increased
the
clearance of αsyn 6-Bio
GSK3β
(5mg/kg)
MPTP (20mg/kg)
C57BL/6J
Enhanced
[111]
mice
autophagy, increased
the
clearance of αsyn, improved motor functions ITC-3 (30mg/kg)
Nrf2
MPTP (20mg/kg)
C57BL/6J
Increased
mice
expression nuclear
[117] of
Nrf2-
dependent oxidative enzymes, decreased activation
of
microglia and TNF-α, improved
ACS Paragon Plus Environment
Page 25 of 37 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
ACS Chemical Neuroscience
motor functions ITC-3
Nrf2
MPP+/BH4
(1μM)
BV-2
Suppressed
[117]
microglial
pro-
cells
inflammatory cytokines, Increased expression nuclear
of
Nrf2,
HO-1, NQO1, GCL gene Curcumin
Nrf2
(100mg/kg)
Rotenone
Lewis rats
(1ml/kg/day)
Decreased
[118]
ROS,
MDA
levels
while
increased GSH, Nrf2 Safranal
Nrf2
(50μg/ml)
Rotenone (100nM)
Primary
Suppressed
[119]
dopaminer apoptosis and gic cells
ROS generation induced
by
rotenone, inhibited Keap1
and
activated Nrf2, HO-1, NQO1, GCLc Naringin
Nrf2
3-nitropropionic acid
PC12 cells Increased cell [120] viability, increased antioxidant enzymes,
ACS Paragon Plus Environment
ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Page 26 of 37
decreased apoptosis, restored MMP Tanshinone-
Nrf2
IIA (20 μg/ml)
6-OHDA (100μM)
SH-SY5Y
Decreased
Cells
apoptosis,
[121]
increased expression
of
Antioxidant related enzyme (ARE)regulated gene expression, increased nuclear Nrf2 Pinostrobin
Nrf2
(125μM)
MPTP
Zebrafish
Rescued loss of [122] dopaminergic
(360μM)
neurons, improved locomotion,
Pinostrobin (25μM)
Nrf2
MPP+ (1.5mM)
SH-SY5Y
Improved cell [122]
cells
viability, increased levels
of
nuclear Nrf2 1 2
6. Concluding Remarks and Future directions:
3
Recent studies reinforce that the turnover of α-syn is the main touchstone of PD as it manifests
4
aggregation of α-syn as part of its pathology. A lot of advances have been done to understand the
5
pathophysiology of PD but still, we do not have a complete depiction regarding the pathophysiology
6
of PD. Many studies have been done which strongly advocates the role of autophagy in PD but still it
7
is quite ambiguous whether autophagy is neuroprotective or not due to its complex nature. In the
ACS Paragon Plus Environment
Page 27 of 37 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
ACS Chemical Neuroscience
1
closure, visionary studies employing autophagy promoting agents catered decisive insight regarding
2
the role of autophagy and the importance of autophagy activation in PD. Different agents which may
3
found to promote clearance of toxic α-syn by targeting the autophagy-lysosomal pathway, GSK3β,
4
IMPase, c-Abl, mTOR, etc might be suitable therapeutic agents to halt the progression of PD
5
pathology. Howbeit, the clinical application of autophagy promoting agents is defined because of their
6
non-selectivity and property of double-edged sword, hitherto they are not the primary choice in the
7
treatment of PD. Therefore, advancement and deep understanding of molecular mechanisms of
8
autophagy in PD is the pre-requisite and it may lead to the development of agents which may promote
9
the clearance of toxic α-syn thereby combating PD pathology. As the field of autophagy has gained
10
focus in recent years and as it is growing expeditiously, updates may be expected soon.
11 12
7. Conflict of Interests:
13
The authors declare no conflict of interest
14 15
8. Funding Sources:
16
This supplement was supported by the seed fund of the National Institute of Pharmaceutical Education
17
and Research (NIPER), Ahmedabad and Ministry of Chemical and Fertilizers, Government of India.
18 19
9. Author contribution:
20 21 22
PP, NS and AK contribute to conception and design. PP did a rigorous literature search. PP, NS, AG, AS, MS and AK participated in revising the manuscript critically for important intelligence part. PP, NS, AK wrote the manuscript. AK supervised the project.
23 24 25 26 27 28 29 30 31 32 33 34
ACS Paragon Plus Environment
ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51
Page 28 of 37
References 1. 2. 3. 4. 5. 6. 7. 8. 9. 10. 11. 12. 13. 14. 15. 16. 17. 18. 19. 20. 21. 22. 23. 24. 25.
Tanner, C.M. and S.M. Goldman, Epidemiology of Parkinson's disease. Neurologic clinics, 1996. 14(2): p. 317-335. MartÌ, M.J., E. Tolosa, and J. Campdelacreu, Clinical overview of the synucleinopathies. Movement disorders: official journal of the Movement Disorder Society, 2003. 18(S6): p. 21-27. Swick, T.J., Parkinson's disease and sleep/wake disturbances. Parkinson’s Disease, 2012. 2012. Munhoz, R.P., et al., Non-motor signs in Parkinson’s disease: a review. Arquivos de neuro-psiquiatria, 2015. 73(5): p. 454-462. Spillantini, M.G., et al., α-Synuclein in Lewy bodies. Nature, 1997. 388(6645): p. 839. Singleton, A., et al., α-Synuclein locus triplication causes Parkinson's disease. Science, 2003. 302(5646): p. 841-841. Chartier-Harlin, M.-C., et al., α-synuclein locus duplication as a cause of familial Parkinson's disease. The Lancet, 2004. 364(9440): p. 1167-1169. Wales, P., et al., Limelight on alpha-synuclein: pathological and mechanistic implications in neurodegeneration. Journal of Parkinson's disease, 2013. 3(4): p. 415-459. Klein, C. and A. Westenberger, Genetics of Parkinson’s disease. Cold Spring Harbor perspectives in medicine, 2012. 2(1): p. a008888. Ross, O.A., et al., Genomic investigation of α-synuclein multiplication and parkinsonism. Annals of Neurology: Official Journal of the American Neurological Association and the Child Neurology Society, 2008. 63(6): p. 743-750. Levine, B. and G. Kroemer, Autophagy in the pathogenesis of disease. Cell, 2008. 132(1): p. 27-42. De Duve, C., et al., Tissue fractionation studies. 6. Intracellular distribution patterns of enzymes in rat-liver tissue. Biochemical Journal, 1955. 60(4): p. 604. Sabatini, D.D. and M. Adesnik, Christian de Duve: Explorer of the cell who discovered new organelles by using a centrifuge. Proceedings of the National Academy of Sciences, 2013. 110(33): p. 1323413235. Tsukada, M. and Y. Ohsumi, Isolation and characterization of autophagy-defective mutants of Saccharomyces cerevisiae. FEBS letters, 1993. 333(1-2): p. 169-174. Deter, R.L., P. Baudhuin, and C. de Duve, Participation of lysosomes in cellular autophagy induced in rat liver by glucagon. The Journal of cell biology, 1967. 35(2): p. C11. Masclaux-Daubresse, C., Q. Chen, and M. Havé, Regulation of nutrient recycling via autophagy. Current opinion in plant biology, 2017. 39: p. 8-17. Mizushima, N. and D.J. Klionsky, Protein turnover via autophagy: implications for metabolism. Annu. Rev. Nutr., 2007. 27: p. 19-40. Chiang, H. and J.F. Dice, Peptide sequences that target proteins for enhanced degradation during serum withdrawal. Journal of Biological Chemistry, 1988. 263(14): p. 6797-6805. Agarraberes, F.A. and J.F. Dice, A molecular chaperone complex at the lysosomal membrane is required for protein translocation. Journal of cell science, 2001. 114(13): p. 2491-2499. Cuervo, A.M. and J.F. Dice, A receptor for the selective uptake and degradation of proteins by lysosomes. Science, 1996. 273(5274): p. 501-503. Hatem, C.L., N.R. Gough, and D.M. Fambrough, Multiple mRNAs encode the avian lysosomal membrane protein LAMP-2, resulting in alternative transmembrane and cytoplasmic domains. Journal of Cell Science, 1995. 108(5): p. 2093-2100. Cuervo, A. and J. Dice, Unique properties of lamp2a compared to other lamp2 isoforms. Journal of cell science, 2000. 113(24): p. 4441-4450. Li, J. and S.R. Pfeffer, Lysosomal membrane glycoproteins bind cholesterol and contribute to lysosomal cholesterol export. Elife, 2016. 5: p. e21635. Hayashi-Nishino, M., et al., A subdomain of the endoplasmic reticulum forms a cradle for autophagosome formation. Nature cell biology, 2009. 11(12): p. 1433. Wei, Y., et al., Origin of the Autophagosome Membrane in Mammals. BioMed research international, 2018. 2018.
ACS Paragon Plus Environment
Page 29 of 37 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49
26. 27. 28. 29. 30. 31. 32. 33. 34. 35. 36.
37. 38. 39. 40. 41. 42. 43. 44. 45. 46. 47. 48.
ACS Chemical Neuroscience
Hosokawa, N., et al., Atg101, a novel mammalian autophagy protein interacting with Atg13. Autophagy, 2009. 5(7): p. 973-979. Itakura, E. and N. Mizushima, Characterization of autophagosome formation site by a hierarchical analysis of mammalian Atg proteins. Autophagy, 2010. 6(6): p. 764-776. Ravikumar, B., et al., Mammalian macroautophagy at a glance. Journal of cell science, 2009. 122(11): p. 1707-1711. Jahreiss, L., F.M. Menzies, and D.C. Rubinsztein, The itinerary of autophagosomes: from peripheral formation to kiss-and-run fusion with lysosomes. Traffic, 2008. 9(4): p. 574-587. Martinez-Vicente, M., et al., Dopamine-modified α-synuclein blocks chaperone-mediated autophagy. The Journal of clinical investigation, 2008. 118(2): p. 777-788. Mak, S.K., et al., Lysosomal degradation of α-synuclein in vivo. Journal of Biological Chemistry, 2010. 285(18): p. 13621-13629. Vogiatzi, T., et al., Wild type a-synuclein is degraded by chaperone mediated autophagy and macroautophagy in neuronal cells. Journal of Biological Chemistry, 2008. Xilouri, M., et al., Boosting chaperone-mediated autophagy in vivo mitigates α-synuclein-induced neurodegeneration. Brain, 2013. 136(7): p. 2130-2146. Alvarez-Erviti, L., et al., Chaperone-mediated autophagy markers in Parkinson disease brains. Archives of neurology, 2010. 67(12): p. 1464-1472. Sala, G., et al., Role of chaperone-mediated autophagy dysfunctions in the pathogenesis of Parkinson’s disease. Frontiers in molecular neuroscience, 2016. 9: p. 157. Tang, F.-L., et al., VPS35 in dopamine neurons is required for endosome-to-Golgi retrieval of Lamp2a, a receptor of chaperone-mediated autophagy that is critical for α-synuclein degradation and prevention of pathogenesis of Parkinson's disease. Journal of Neuroscience, 2015. 35(29): p. 1061310628. Yang, Q., et al., Regulation of neuronal survival factor MEF2D by chaperone-mediated autophagy. Science, 2009. 323(5910): p. 124-127. Murphy, K.E., et al., Reduced glucocerebrosidase is associated with increased α-synuclein in sporadic Parkinson’s disease. Brain, 2014. 137(3): p. 834-848. Kim, J., et al., A MicroRNA feedback circuit in midbrain dopamine neurons. Science, 2007. 317(5842): p. 1220-1224. Li, G., et al., Targeted suppression of chaperone-mediated autophagy by miR-320a promotes αsynuclein aggregation. International journal of molecular sciences, 2014. 15(9): p. 15845-15857. Alvarez-Erviti, L., et al., Influence of microRNA deregulation on chaperone-mediated autophagy and α-synuclein pathology in Parkinson's disease. Cell death & disease, 2013. 4(3): p. e545. Luan, Y.-n., et al., Chronic caffeine treatment protects against α-synucleinopathy by reestablishing autophagy activity in the mouse striatum. Frontiers in neuroscience, 2018. 12: p. 301. Gegg, M.E. and A.H. Schapira, The role of glucocerebrosidase in Parkinson disease pathogenesis. The FEBS journal, 2018. 285(19): p. 3591-3603. Spencer, B., et al., Beclin 1 gene transfer activates autophagy and ameliorates the neurodegenerative pathology in α-synuclein models of Parkinson's and Lewy body diseases. Journal of Neuroscience, 2009. 29(43): p. 13578-13588. Yan, J.-q., et al., E46K Mutant α-Synuclein Is Degraded by Both Proteasome and Macroautophagy Pathway. Molecules, 2018. 23(11): p. 2839. Salvador, N., et al., Import of a cytosolic protein into lysosomes by chaperone-mediated autophagy depends on its folding state. Journal of Biological Chemistry, 2000. 275(35): p. 27447-27456. Tanik, S.A., et al., Lewy body-like α-synuclein aggregates resist degradation and impair macroautophagy. Journal of Biological Chemistry, 2013. 288(21): p. 15194-15210. Ahmed, I., et al., Development and characterization of a new Parkinson's disease model resulting from impaired autophagy. Journal of Neuroscience, 2012. 32(46): p. 16503-16509.
ACS Paragon Plus Environment
ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50
49. 50. 51. 52. 53. 54. 55. 56. 57. 58. 59. 60. 61. 62. 63. 64. 65. 66. 67. 68. 69.
Page 30 of 37
Friedman, L.G., et al., Disrupted autophagy leads to dopaminergic axon and dendrite degeneration and promotes presynaptic accumulation of α-synuclein and LRRK2 in the brain. Journal of Neuroscience, 2012. 32(22): p. 7585-7593. Oueslati, A., et al., Polo-like kinase 2 regulates selective autophagic α-synuclein clearance and suppresses its toxicity in vivo. Proceedings of the National Academy of Sciences, 2013. 110(41): p. E3945-E3954. Tenreiro, S., et al., Phosphorylation modulates clearance of alpha-synuclein inclusions in a yeast model of Parkinson's disease. PLoS genetics, 2014. 10(5): p. e1004302. Shahpasandzadeh, H., et al., Interplay between sumoylation and phosphorylation for protection against α-synuclein inclusions. Journal of Biological Chemistry, 2014. 289(45): p. 31224-31240. Sevlever, D., P. Jiang, and S.-H.C. Yen, Cathepsin D is the main lysosomal enzyme involved in the degradation of α-synuclein and generation of its carboxy-terminally truncated species. Biochemistry, 2008. 47(36): p. 9678-9687. Cullen, V., et al., Cathepsin D expression level affects alpha-synuclein processing, aggregation, and toxicity in vivo. Molecular brain, 2009. 2(1): p. 5. Miura, E., et al., VPS35 dysfunction impairs lysosomal degradation of α-synuclein and exacerbates neurotoxicity in a Drosophila model of Parkinson's disease. Neurobiology of disease, 2014. 71: p. 113. Zimprich, A., et al., A mutation in VPS35, encoding a subunit of the retromer complex, causes lateonset Parkinson disease. The American Journal of Human Genetics, 2011. 89(1): p. 168-175. Zavodszky, E., et al., Mutation in VPS35 associated with Parkinson’s disease impairs WASH complex association and inhibits autophagy. Nature communications, 2014. 5: p. 3828. Winslow, A.R., et al., α-Synuclein impairs macroautophagy: implications for Parkinson’s disease. The Journal of cell biology, 2010. 190(6): p. 1023-1037. Kong, S.M., et al., Parkinson's disease-linked human PARK9/ATP13A2 maintains zinc homeostasis and promotes α-Synuclein externalization via exosomes. Human molecular genetics, 2014. 23(11): p. 2816-2833. Angot, E., et al., Alpha-synuclein cell-to-cell transfer and seeding in grafted dopaminergic neurons in vivo. PLoS One, 2012. 7(6): p. e39465. Lopes da Fonseca, T., R. Pinho, and T.F. Outeiro, A familial ATP13A2 mutation enhances alphasynuclein aggregation and promotes cell death. Human molecular genetics, 2016. 25(14): p. 29592971. Alvarez-Erviti, L., et al., Lysosomal dysfunction increases exosome-mediated alpha-synuclein release and transmission. Neurobiology of disease, 2011. 42(3): p. 360-367. Danzer, K.M., et al., Exosomal cell-to-cell transmission of alpha synuclein oligomers. Molecular neurodegeneration, 2012. 7(1): p. 42. Ejlerskov, P., et al., Tubulin polymerization-promoting protein (TPPP/p25α) promotes unconventional secretion of α-synuclein through exophagy by impairing autophagosome-lysosome fusion. Journal of Biological Chemistry, 2013. 288(24): p. 17313-17335. Anglade, P., et al., Apoptosis and autophagy in nigral neurons of patients with Parkinson's disease. Histology and histopathology, 1997. 12(1): p. 25-32. Klucken, J., et al., Alpha-synuclein aggregation involves a bafilomycin A1-sensitive autophagy pathway. Autophagy, 2012. 8(5): p. 754-766. Wu, G., et al., Altered expression of autophagic genes in the peripheral leukocytes of patients with sporadic Parkinson's disease. Brain research, 2011. 1394: p. 105-111. Song, J.-X., et al., HMGB1 is involved in autophagy inhibition caused by SNCA/α-synuclein overexpression: a process modulated by the natural autophagy inducer corynoxine B. Autophagy, 2014. 10(1): p. 144-154. Yan, J.-Q., et al., Overexpression of human E46K mutant α-synuclein impairs macroautophagy via inactivation of JNK1-Bcl-2 pathway. Molecular neurobiology, 2014. 50(2): p. 685-701.
ACS Paragon Plus Environment
Page 31 of 37 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50
70. 71. 72. 73. 74. 75. 76. 77. 78. 79. 80. 81. 82. 83. 84. 85. 86. 87. 88. 89. 90. 91. 92.
ACS Chemical Neuroscience
Zimprich, A., et al., Mutations in LRRK2 cause autosomal-dominant parkinsonism with pleomorphic pathology. Neuron, 2004. 44(4): p. 601-607. Hardy, J., Genetic analysis of pathways to Parkinson disease. Neuron, 2010. 68(2): p. 201-206. Haugarvoll, K. and Z.K. Wszolek, Clinical features of LRRK2 parkinsonism. Parkinsonism & related disorders, 2009. 15: p. S205-S208. Manzoni, C., et al., Pathogenic Parkinson’s disease mutations across the functional domains of LRRK2 alter the autophagic/lysosomal response to starvation. Biochemical and biophysical research communications, 2013. 441(4): p. 862-866. Tong, Y., et al., Loss of leucine-rich repeat kinase 2 causes impairment of protein degradation pathways, accumulation of α-synuclein, and apoptotic cell death in aged mice. Proceedings of the National Academy of Sciences, 2010. 107(21): p. 9879-9884. Esteves, A., et al., The upshot of LRRK2 inhibition to Parkinson’s disease paradigm. Molecular neurobiology, 2015. 52(3): p. 1804-1820. Arduíno, D.M., et al., Mitochondrial metabolism in Parkinson's disease impairs quality control autophagy by hampering microtubule-dependent traffic. Human molecular genetics, 2012. 21(21): p. 4680-4702. Schapansky, J., et al., Membrane recruitment of endogenous LRRK2 precedes its potent regulation of autophagy. Human molecular genetics, 2014. 23(16): p. 4201-4214. Plowey, E.D., et al., Role of autophagy in G2019S-LRRK2-associated neurite shortening in differentiated SH-SY5Y cells. Journal of neurochemistry, 2008. 105(3): p. 1048-1056. Gomez-Suaga, P., et al., Leucine-rich repeat kinase 2 regulates autophagy through a calciumdependent pathway involving NAADP. Human molecular genetics, 2011. 21(3): p. 511-525. Henry, A.G., et al., Pathogenic LRRK2 mutations, through increased kinase activity, produce enlarged lysosomes with reduced degradative capacity and increase ATP13A2 expression. Human molecular genetics, 2015. 24(21): p. 6013-6028. Park, S., et al., Interplay between leucine-rich repeat kinase 2 (LRRK2) and p62/SQSTM-1 in selective autophagy. PloS one, 2016. 11(9): p. e0163029. Ho, D.H., et al., LRRK2 impairs autophagy by mediating phosphorylation of leucyl-tRNA synthetase. Cell biochemistry and function, 2018. 36(8): p. 431-442. Daher, J.P., et al., Abrogation of α-synuclein–mediated dopaminergic neurodegeneration in LRRK2deficient rats. Proceedings of the National Academy of Sciences, 2014. 111(25): p. 9289-9294. Daher, J.P., et al., Leucine-rich repeat kinase 2 (LRRK2) pharmacological inhibition abates α-synuclein gene-induced neurodegeneration. Journal of Biological Chemistry, 2015. 290(32): p. 19433-19444. Skibinski, G., et al., Nrf2 mitigates LRRK2-and α-synuclein–induced neurodegeneration by modulating proteostasis. Proceedings of the National Academy of Sciences, 2017. 114(5): p. 1165-1170. Bae, E.-J., et al., LRRK2 kinase regulates α-synuclein propagation via RAB35 phosphorylation. Nature communications, 2018. 9(1): p. 3465. Bockaert, J. and P. Marin, mTOR in brain physiology and pathologies. Physiological reviews, 2015. 95(4): p. 1157-1187. Zhou, Q., et al., Rotenone induction of hydrogen peroxide inhibits mTOR-mediated S6K1 and 4EBP1/eIF4E pathways, leading to neuronal apoptosis. Toxicological sciences, 2014. 143(1): p. 81-96. Xu, Y., et al., Activation of AMPK and inactivation of Akt result in suppression of mTOR-mediated S6K1 and 4E-BP1 pathways leading to neuronal cell death in in vitro models of Parkinson's disease. Cellular signalling, 2014. 26(8): p. 1680-1689. Choi, K.C., et al., A novel mTOR activating protein protects dopamine neurons against oxidative stress by repressing autophagy related cell death. Journal of neurochemistry, 2010. 112(2): p. 366-376. Dijkstra, A.A., et al., Evidence for immune response, axonal dysfunction and reduced endocytosis in the substantia nigra in early stage Parkinson’s disease. PLoS One, 2015. 10(6): p. e0128651. Santini, E., et al., Inhibition of mTOR signaling in Parkinson’s disease prevents L-DOPA–induced dyskinesia. Sci. Signal., 2009. 2(80): p. ra36-ra36.
ACS Paragon Plus Environment
ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49
93. 94. 95. 96. 97. 98. 99. 100. 101. 102. 103. 104. 105. 106. 107. 108. 109. 110. 111. 112.
Page 32 of 37
Bjedov, I., et al., Mechanisms of life span extension by rapamycin in the fruit fly Drosophila melanogaster. Cell metabolism, 2010. 11(1): p. 35-46. Zhang, J., et al., Mesencephalic astrocyte-derived neurotrophic factor alleviated 6-OHDA-induced cell damage via ROS-AMPK/mTOR mediated autophagic inhibition. Experimental gerontology, 2017. 89: p. 45-56. Yao, L., et al., Unexpected Neuroprotective Effects of Loganin on 1-Methyl-4-Phenyl-1, 2, 3, 6-Tetrahydropyridine-Induced Neurotoxicity and Cell Death in Zebrafish. Journal of cellular biochemistry, 2017. 118(3): p. 615-628. Pérez-Revuelta, B., et al., Metformin lowers Ser-129 phosphorylated α-synuclein levels via mTORdependent protein phosphatase 2A activation. Cell death & disease, 2014. 5(5): p. e1209. Wang, S.-j., et al., Effect of moxibustion on mTOR-mediated autophagy in rotenone-induced Parkinson's disease model rats. Neural regeneration research, 2018. 13(1): p. 112. Zhang, Z., et al., MANF protects dopamine neurons and locomotion defects from a human αsynuclein induced Parkinson's disease model in C. elegans by regulating ER stress and autophagy pathways. Experimental neurology, 2018. 308: p. 59-71. Sheng, Y.-L., et al., Urate promotes SNCA/α-synuclein clearance via regulating mTOR-dependent macroautophagy. Experimental neurology, 2017. 297: p. 138-147. Ren, Z.-l., et al., Ganoderma lucidum extract ameliorates MPTP-induced parkinsonism and protects dopaminergic neurons from oxidative stress via regulating mitochondrial function, autophagy, and apoptosis. Acta pharmacologica Sinica, 2018: p. 1. Ferretta, A., et al., Effect of resveratrol on mitochondrial function: implications in parkin-associated familiar Parkinson's disease. Biochimica et Biophysica Acta (BBA)-Molecular Basis of Disease, 2014. 1842(7): p. 902-915. Kang, S.Y., et al., Autophagic modulation by rosuvastatin prevents rotenone-induced neurotoxicity in an in vitro model of Parkinson’s disease. Neuroscience letters, 2017. 642: p. 20-26. Yan, Q., et al., Activation of AMPK/mTORC1-mediated autophagy by metformin reverses Clk1 deficiency-sensitized dopaminergic neuronal death. Molecular pharmacology, 2017: p. mol. 117.109512. Bayliss, J.A., et al., Metformin prevents nigrostriatal dopamine degeneration independent of AMPK activation in dopamine neurons. PloS one, 2016. 11(7): p. e0159381. Savolainen, M.H., et al., The beneficial effect of a prolyl oligopeptidase inhibitor, KYP-2047, on alphasynuclein clearance and autophagy in A30P transgenic mouse. Neurobiology of disease, 2014. 68: p. 1-15. Hou, L., et al., Lithium protects dopaminergic cells from rotenone toxicity via autophagy enhancement. BMC neuroscience, 2015. 16(1): p. 82. Li, X.-z., et al., Therapeutic effects of valproate combined with lithium carbonate on MPTP-induced parkinsonism in mice: possible mediation through enhanced autophagy. International Journal of Neuroscience, 2012. 123(2): p. 73-79. Sarkar, S., et al., Lithium induces autophagy by inhibiting inositol monophosphatase. J Cell Biol, 2005. 170(7): p. 1101-1111. Ren, Y., et al., Role of c-Abl-GSK3β signaling in MPP+-induced autophagy-lysosomal dysfunction. Toxicological Sciences, 2018. Li, L., et al., Role of GSK3β/α-synuclein axis in methamphetamine-induced neurotoxicity in PC12 cells. Toxicology Research, 2018. 7(2): p. 221-234. Suresh, S., et al., A novel autophagy modulator 6-Bio ameliorates SNCA/α-synuclein toxicity. Autophagy, 2017. 13(7): p. 1221-1234. He, Q., et al., Treatment with trehalose prevents behavioral and neurochemical deficits produced in an AAV α-synuclein rat model of Parkinson’s disease. Molecular neurobiology, 2016. 53(4): p. 22582268.
ACS Paragon Plus Environment
Page 33 of 37 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26
113. 114. 115. 116. 117. 118. 119. 120. 121. 122.
ACS Chemical Neuroscience
Rekha, K.R. and R.I. Sivakamasundari, Geraniol Protects Against the Protein and Oxidative Stress Induced by Rotenone in an In Vitro Model of Parkinson’s Disease. Neurochemical research, 2018: p. 1-16. Yang, G., et al., Glycyrrhizic Acid Alleviates 6-Hydroxydopamine and Corticosterone-Induced Neurotoxicity in SH-SY5Y Cells Through Modulating Autophagy. Neurochemical research, 2018. 43(10): p. 1914-1926. Decressac, M., et al., TFEB-mediated autophagy rescues midbrain dopamine neurons from αsynuclein toxicity. Proceedings of the National Academy of Sciences, 2013: p. 201305623. Kilpatrick, K., et al., Genetic and chemical activation of TFEB mediates clearance of aggregated αsynuclein. PloS one, 2015. 10(3): p. e0120819. Lee, J.A., et al., A novel compound ITC-3 activates the Nrf2 signaling and provides neuroprotection in Parkinson’s disease models. Neurotoxicity research, 2015. 28(4): p. 332-345. Cui, Q., X. Li, and H. Zhu, Curcumin ameliorates dopaminergic neuronal oxidative damage via activation of the Akt/Nrf2 pathway. Molecular medicine reports, 2016. 13(2): p. 1381-1388. Pan, P., L. Qiao, and X. Wen, Safranal prevents rotenone-induced oxidative stress and apoptosis in an in vitro model of Parkinson's disease through regulating Keap1/Nrf2 signaling pathway. Cellular and Molecular Biology, 2016. 62(14): p. 11-17. Kulasekaran, G. and S. Ganapasam, Neuroprotective efficacy of naringin on 3-nitropropionic acidinduced mitochondrial dysfunction through the modulation of Nrf2 signaling pathway in PC12 cells. Molecular and cellular biochemistry, 2015. 409(1-2): p. 199-211. Zhang, X., et al., Tanshinone IIA protects dopaminergic neurons against 6-hydroxydopamine-induced neurotoxicity through miR-153/NF-E2-related factor 2/antioxidant response element signaling pathway. Neuroscience, 2015. 303: p. 489-502. Li, C., et al., Pinostrobin Exerts Neuroprotective Actions in Neurotoxin-Induced Parkinson’s Disease Models through Nrf2 Induction. Journal of agricultural and food chemistry, 2018. 66(31): p. 83078318.
27 28
Table of Contents Graphic (TOC)
29
ACS Paragon Plus Environment
ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
llustration representing crosstalk in between α-syn and chaperone-mediated autophagy (CMA)
ACS Paragon Plus Environment
Page 34 of 37
Page 35 of 37 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
ACS Chemical Neuroscience
Illustration representing the crosstalk in between α-syn and macroautophagy 414x254mm (300 x 300 DPI)
ACS Paragon Plus Environment
ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Illustration representing molecular mechanisms regulating the process of autophagy and therapeutic agents acting on different targets for promoting autophagy via activation of autophagy pathway 303x193mm (300 x 300 DPI)
ACS Paragon Plus Environment
Page 36 of 37
Page 37 of 37 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
ACS Chemical Neuroscience
309x174mm (150 x 150 DPI)
ACS Paragon Plus Environment