Skip to main content
Acta Pharmaceutica Sinica. B logoLink to Acta Pharmaceutica Sinica. B
. 2021 Feb 26;11(10):3015–3034. doi: 10.1016/j.apsb.2021.02.016

Targeting autophagy using small-molecule compounds to improve potential therapy of Parkinson's disease

Kai Zhang a,, Shiou Zhu a,, Jiamei Li a,, Tingting Jiang a, Lu Feng a, Junping Pei a, Guan Wang a,b,, Liang Ouyang a,, Bo Liu a,
PMCID: PMC8546670  PMID: 34729301

Abstract

Parkinson's disease (PD), known as one of the most universal neurodegenerative diseases, is a serious threat to the health of the elderly. The current treatment has been demonstrated to relieve symptoms, and the discovery of new small-molecule compounds has been regarded as a promising strategy. Of note, the homeostasis of the autolysosome pathway (ALP) is closely associated with PD, and impaired autophagy may cause the death of neurons and thereby accelerating the progress of PD. Thus, pharmacological targeting autophagy with small-molecule compounds has been drawn a rising attention so far. In this review, we focus on summarizing several autophagy-associated targets, such as AMPK, mTORC1, ULK1, IMPase, LRRK2, beclin-1, TFEB, GCase, ERRα, C-Abelson, and as well as their relevant small-molecule compounds in PD models, which will shed light on a clue on exploiting more potential targeted small-molecule drugs tracking PD treatment in the near future.

KEY WORDS: Parkinson's disease (PD), Autophagy, Target, Small-molecule compound, PD therapy

Abbreviations: 3-MA, 3-methyladenine; 5-HT2A, Serotonin 2A; 5-HT2C, serotonin 2C; α-syn, α-synuclein; A2A, adenosine 2A; AADC, aromatic amino acid decarboxylase; ALP, autophagy-lysosomal pathway; AMPK, 5ʹAMP-activated protein kinase; ATP13A2, ATPase cation transporting 13A2; ATG, autophagy related protein; ATTEC, autophagosome-tethering compound; AUC, the area under the curve; AUTAC, autophagy targeting chimera; BAF, bafilomycinA1; BBB, blood−brain barrier; CL, clearance rate; CMA, chaperone-mediated autophagy; CNS, central nervous system; COMT, catechol-O-methyltransferase; DA, dopamine; DAT, dopamine transporter; DJ-1, Parkinson protein 7; DR, dopamine receptor; ER, endoplasmic reticulum; ERRα, estrogen-related receptor alpha; F, oral bioavailability; GAPDH, glyceraldehyde 3-phosphate dehydrogenase; GBA, glucocerebrosidase β acid; GWAS, genome-wide association study; HDAC6, histone deacetylase 6; HSPA8, heat shock 70 kDa protein 8; HSC70, heat shock cognate 71 kDa protein; IMPase, inositol monophosphatase; IPPase, inositol polyphosphate 1-phosphatase; Lamp2a, type 2A lysosomal-associated membrane protein; KI, knockin; LAMP2A, lysosome-associated membrane protein 2 A; LC3, light chain 3; LIMP-2, lysosomal integrated membrane protein-2; LRRK2, leucine-rich repeat sequence kinase 2; LRS, leucyl-tRNA synthetase; LUHMES, lund human mesencephalic; mAChR, muscarinic acetylcholine receptor; MAO-B, monoamine oxidase B; MPP+, 1-methyl-4-phenylpyridinium; MPTP, 1-methyl-4-phenyl-1,2,5,6-tetrahydropyridine; mTOR, the mammalian target of rapamycin; MYCBP2, MYC-binding protein 2; NMDA, N-methyl-d-aspartic acid; ONRs, orphan nuclear receptors; Parkin, parkin RBR E3 ubiquitin−protein ligase; PD, Parkinson's disease; PDE4, phosphodiesterase 4; PI3K, phosphatidylinositol 3-kinase; PI3P, phosphatidylinositol 3-phosphate; PINK1, PTEN-induced kinase 1; PLC, phospholipase C; PREP, prolyl oligopeptidase; ROS, reactive oxygen species; SAR, structure–activity relationship; SAS, solvent accessible surface; SN, substantia nigra; SNCA, α-synuclein gene; SYT11, synaptotagmin 11; TFEB, transcription factor EB; TSC2, tuberous sclerosis complex 2; ULK1, UNC-51-like kinase 1; UPS, ubiquitin−proteasome system

Graphical abstract

In this review, we summarize the mechanism of autophagy in the pathogenesis of Parkinson's disease (PD). We focus on several cytoprotective autophagy-regulated targets, such as LRRK2, C-Abelson, GCase, TFEB, ERRα, mTORC1, AMPK, ULK1, beclin-1, and IMPase. At the meantime, their relevant small-molecule compounds in PD models are listed.

Image 1

1. Introduction

Parkinson's disease (PD) is an incurable neurodegenerative disease characterized by dopaminergic neurons loss and Lewy bodies (LB) formed by abnormal aggregation of α-synuclein (α-syn) protein, involving some clinical symptoms like myotonia, cognitive changes, neurological dysfunction, motor retardation as well as posture disorders1. Indeed, PD is considered as a progressive disease, drug therapy is the main choice in clinical treatment. The Parkinson's disease drugs approved by US Food and Drug Administration (FDA) can be roughly divided into four categories including dopaminergic system drugs, serotonergic system drugs, cholinergic system drugs, and others. Aromatic amino acid decarboxylase (AADC) inhibitors, catechol-O-methyltransferase (COMT) inhibitors, monoamine oxidase B (MAO-B) inhibitors, dopamine transporter (DAT) inhibitors and dopamine receptor (DR) agonists affect the dopaminergic system, while 5-hydroxytryptamine (5-HT) 2A and 2C receptor antagonists target serotonergic system. Cholinergic system drugs comprise muscarinic acetylcholine receptor (mAChR) antagonists. And adenosine receptor (A2A) inhibitors and glutamate receptor (NMDA) antagonists are considered as other medications for PD2, 3, 4, 5. As the main strategy is to alleviate the symptoms of patients in a short time, present long-term clinical applications are particularly vulnerable to serious autonomic nervous dysfunction and other insensitive symptoms, failing to achieve satisfactory treatment results6. Among the complex pathogenesis of PD, disequilibrium of autophagy homeostasis acts as a trigger for PD progression while cytoprotective autophagy can save related pathological states in preclinical models of PD.

Autophagy is an evolutionarily conserved process for eukaryotic cells to degrade themselves through lysosomes, which plays a vital role in cell survival, proliferation and death. It may help to maintain the homeostasis of internal environment. Autophagy can degrade most of the cytoplasm and even the entire organelle, including the pathogenic aggregation of intra-neuronal proteins (Lewy bodies) which are composed of α-syn. In this process, the defective protein or organelle is firstly engulfed by the phagocytic vesicle with a lipid bilayer membrane, then the autophagosome emerges. Afterwards, these structures are transported to the lysosome. With the combination of a lysosome, it forms an autophagolysosome to complete the degradation7. Therefore, the application of existing autophagic regulators, as well as the discovery of new small molecules targeting the autolysosome pathway (ALP), have been demonstrated as a great potential on PD treatment.

2. Molecular mechanisms of autolysosome pathway in PD pathogenesis

Autophagy is functionally active within the central nervous system, which maintains the homeostasis of neurons and glial cells8, 9, 10, 11. Highly-differentiated neurons and glial cells are difficult to repair damage and demand more energy for normal activities and autophagic recycling of components than other cells, therefore, requiring sophisticated quality control system9,12.

Indeed, during high-intensity work in the central nervous system (CNS), the ALP is prone to produce dysfunction, while some risk gene mutations of PD also exert deleterious effect on ALP, including some selective autophagy pathways like mitophagy and chaperone-mediated autophagy (CMA). As a result, impaired ALP may promote toxic protein aggregates (like α-syn) accumulation and lead to lysosome dysfunction, delayed clearance of defective mitochondria and increased oxidative stress, which are all considered as stimulators of PD (Fig. 1)13. In a zebra fish PD model treated with MPTP, ATG5 was found decreased, which was correlated with blocked autophagy flux, leading to increase of PD-associated proteins like β-syn, parkin and PINK1, together with loss of dopamine (DA) neurons14.

Figure 1.

Figure 1

The relationship between autophagy and PD: the mutations of casual genes cause some ALP defects and further lead to key pathological states. ALP dysfunction is one of the important pathogenesis of PD. Pharmacologically induced autophagy can rescue the pathological states to a certain extent and play a neuroprotective role.

Recently, with the analysis of genome-wide association study (GWAS), five relative genes of PD, α-syn (SNCA), gene leucine-rich repeat sequence kinase 2 (LRRK2) genes, PTEN-induced kinase 1 (PINK1), Parkin RBR E3 ubiquitin−protein ligase (PARK2, Parkin), and Parkinson protein 7 (PARK7, DJ-1) were identified15. Of note, mutations of these genes are considered as risk factors of PD and one of the pathogenesis mechanisms of PD underlying those genes mutations is the ALP defects.

For example, LRRK2 has captured much attention in PD pathogenesis16, and the relationship of its mutations and autophagy has been extensively investigated and reviewed in recent years17, 18, 19, 20. Accumulating evidence has pointed that the impaired ALP caused by LRRK2 mutations may compose one of its pathogenic mechanisms, whereas the exact mechanisms remain unclear. In general, G2019S, the most frequent mutation in LRRK2, was discovered to trigger progressive dysfunctions of dopamine transporters21. Also, LRRK2 G2019S could phosphorylate and activate leucyl-tRNA synthetase (LRS) and then activating mTORC1, thereby suppressing autophagy22. And the single loss of LRRK2 was reported to not promote selective neurodegeneration, level of α-syn and impairment of ALP, revealing an essential role of LRRK2 in the modulation of ALP pathways23. Besides the traditional autophagy pathways, some latest studies also shed light on its role in regulating selective autophagy associated with PD, such as CMA and mitophagy24. In striatum and cortex of aged LRRK2 R1441G knockin (KI) mice, accumulation of CMA-specific lysosome-associated membrane protein 2 A (LAMP2A) and heat shock 70 kDa protein 8/heat shock cognate 71 kDa protein (HSPA8/HSC70) and increased CMA substrate glyceraldehyde 3-phosphate dehydrogenase (GAPDH) were observed, indicating that LRRK2 mutations may disturb CMA to degrade oligomeric α-syn thereby promoting PD25. Overexpression of kinase active LRRK2 was reported to restrain mitophagy to trigger mitochondrial dysfunction, which was aggravated by the expression of kinase hyperactive LRRK2-G2019S variant and thus accelerate PD progress26.

Importantly, the mutations of SNCA contribute to abnormal aggregates of α-syn (like oligomer and fibrosis forms)27, 28, 29, which was identified to participate in the pathogenesis of PD30, representing the predominant Lewy pathology in PD. These aggregates, which could be degraded by the ALP31,32, may in turn impair mitochondrial, autophagic, and lysosomal functions, aggravating the toxic protein aggregates33. α-Syn overexpression was found to generate ER stress, along with lysosomal pH alkalization, lower LAMP1 levels, and a disruption of lysosomal morphology and distribution, impairing autophagy34. Via abolishing the hydrolase trafficking, the accumulation of α-syn dampens the ability of lysosomes to degrade dopamine in human midbrain dopamine models35. In DA neuron-specific autophagy-deficient mice (autophagy related protein (ATG) 7 knock out), P62 inclusions occurred before synuclein, suggesting the dysfunction of autophagy may be a cause of LB formation36.

Besides, loss of GCase activity due to glucosylceramidase β acid (GBA) mutations impairs the ALP, resulting in increased α-syn levels, which dampen GCase activity in turn37. The L444P heterozygous mutation of GBA, a lysosomal enzyme that degrades glucosylceramide to ceramide and glucose, was reported to generate mitochondrial dysfunction by inhibiting key steps of mitophagy, which underlies part of its pathogenetic roles in PD38.

Additionally, other genes mutations were also proved to link with ALP dysfunction. PINK1 and Parkin mutations were considered to affect mitophagy39,40, which render the delayed clearance of defective mitochondria, exacerbating PD. Both deficiency and mutations of ATPase cation transporting 13A2 (ATP13A2) can damage autophagosome-lysosome fusion through disability of recruiting histone deacetylase 6 (HDAC6) to lysosomes41. In another study, ATP13A2 was found to activate mTORC via the activation of tuberous sclerosis complex 2 (TSC2) caused by its interaction with MYC-binding protein 2 (MYCBP2), and decrease the TEFB-dependent synaptotagmin 11 (SYT11) expression, thus block autophagy42.

As mentioned above, autophagy defects related to sporadic and familial PD may cause the accumulation of protein aggregates and damaged organelles, eventually cell death. Targeting ALP, either chemically or genetically, is proved to be beneficial for neuronal survival, which represents a potential treatment strategy for PD.

3. Targeting autophagy for potential therapy of PD

A variety of small-molecule compounds have been demonstrated in PD models to achieve therapeutic effects by regulating autophagy pathways (Fig. 2 and Table 143, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79). Herein, we discuss some pivotal instances of autophagy-related targets and representative small-molecule compounds.

Figure 2.

Figure 2

Several pharmacological interventions are available to induce cytoautophagy at the nucleation, elongation, fusion, or degradation phase. The figure shows the key targets and representative small-molecule compounds in autophagic regulatory pathways.

Table 1.

Autophagy-modulating compounds in PD.

Target Origin No. Agent Main effect PD model Reported year in PD Ref.
AMPK Natural product 2 Resveratrol Prevented loss of DA neurons; rescued TH and DA levels; improved behavioral abnormalities MPTP treated mice 2008 43
8 Caffeine Reduced p129-α-syn; apoptosis; microglial activation; astrogliosis; increased LC3II/I ratio A53T α-syn transgenic C57BL/6J mice 2001 76
Chemical synthesis 1 Metformin Reduced levels of p-Ser129 α-syn in vivo and in vitro, increased striatal dopaminergic levels α-Syn overexpressing
SH-SY5Y cells and C57BL/6J mice
2014 44
3 A769662 Reduced levels of α-syn inclusions α-Syn overexpressing SH-SY5Y cells 2019 75
4 GSK621 Reduced levels of α-syn inclusions α-Syn overexpressing SH-SY5Y cells 2019 75
5 Rosuvastatin Increased cell viability; increased α-syn clearance Rotenone-exposed SH-SY5Y cells 2017 55
6 FCPR16 Protected neuronal cells against MPP+-induced toxicity and oxidative stress SH-SY5Y cells treated with MPP+ 2018 66
7 Temozolomide Increased α-syn clearance and protected α-syn-induced cytotoxicity α-Syn overexpressing LUHMES cells 74
mTORC1 Natural product 9−11 Rapamycin and Rp analogues (CCI-779 and AP23573) Increased the clearance of WT, A30P and A53T α-syn α-Syn expressing PC12 cells 2010 77
Prevented neuronal death in mice; reduced α-syn accumulation, improved motor function MPTP treated mice; α-syn transgenic mice 50,51
12 Corynoxine Promoted the clearance of wild-type and mutant α-syn via autophagy α-Syn overexpressing PC12 cells 2014 78
13 Loganin Decreased MPTP-induced neurotoxicity MPTP treated PC12 cells 2017 79
Chemical synthesis 14 PI-103 Increased α-syn clearance and protected α-syn-induced cytotoxicity α-Syn overexpressing Lund human mesencephalic (LUHMES) cells 74
ULK1 Chemical synthesis 15 BL-918 Protected against MPTP-induced motor dysfunction and loss of dopaminergic neurons MPP+-treated SH-SY5Y cells 45
IMPase Chemical synthesis 16 Sodium valproate Enhanced autophagy, reduced mitochondrial membrane potential, reduced production of reactive oxygen species and enhanced cell viability Rotenone-induced SH-SY5Y cells 46
17 Carbamazepine Enhanced autophagy, reduced mitochondrial membrane potential, reduced production of reactive oxygen species and enhanced cell viability Rotenone-induced SH-SY5Y cells 46
18 L-690,330 Increased α-syn clearance and protected α-syn-induced cytotoxicity α-Syn overexpressing LUHMES cells 2019 74
LRRK2 Chemical synthesis 19 LRRK2-IN-1 Rescued Lrrk2 G2019S mutant motor injury Drosophila 48
23 GNE-7915 Enhanced DA release and recovery R1441G transgenic mice 47
25 PF-06447475 Reduced neuronal apoptosis and alleviated neurological deficit Weight-drop Sprague–Dawley rats after traumatic brain injury 130
DNL151a 49
DNL201a 49
Beclin-1 Natural product 29 Isorhynchophylline Promoted clearance of wild-type, A53T and A30P α-syn Differentiated human dopaminergic neurons 2014 53
30 Corynoxine B Reversed reduction in LC3-II and BECN1; restored the deficient autophagy induced by SNCA α-Syn overexpressing
PC12 cells
2019 54
31 Glycyrrhizic acid Increased cell viability; up-regulated LC3-II/I and beclin-1 6-OHDA and crticosterone treated SH-SY5Y cells 2018 56
Chemical synthesis 28 KYP-2047 Enhanced autophagy and α-syn clearance; increased in striatal dopamine levels A30P α-syn transgenic mice 52
TFEB Natural product 32−35 Curcumin and Cur analogues (CNB001, C1 and E4) Inhibited the accumulation of α-syn and prevented the accumulation of LB α-Syn overexpressing SH-SY5Y cells 57, 58, 59, 60, 61, 62
36 Trehalose Reduced the loss of SNpc DA neurons and produced a neuroprotective effect Multiple PD related models 2004 63
GCase Chemical synthesis 37 Ambroxola Increased GCase activity and reduced oxidative stress PD fibroblasts with Gba1 mutations 2014 64,65,158
38 Isofagomine Increased GCase levels and activity; lowered ER stress and prevented the loss of motor function PD fibroblasts with Gba1 mutations; Drosophila 67
39 NCGC607 Restored GCase activity and protein levels; reduced
α-syn levels
iPSC-derived macrophages and dopaminergic neurons 68
ERRα Chemical synthesis 40 XCT790 Alleviated dopaminergic neuronal loss; cleared toxic protein aggregates; ameliorated behavioral impairments MPTP treated mice 2018 69,70
c-ABL Chemical synthesis 41 PD180970 Cleared toxic α-syn protein aggregates; alleviated behavioral impairments α-Syn overexpressing SH-SY5Y cells and MPTP induced mice 2019 71
42 Imatinib Reduced expression of c-ABL and p-GSK3β; restored ALP and decreased cells death MPP+-induced SN4741 cells 2014 72
43 Nilotiniba Reduced c-ABL activation, prevented dopamine (DA) neuron loss and behavioral deficits; induced α-syn degradation in vivo and in vitro MPTP-induced mice; α-syn overexpressing primary cultures of mouse cortical neurons and mice 2014 72,73
a

Drugs which have already entered clinical trials. DNL151 (NCT03710707) and DNL201 (NCT04056689) in phase Ⅰ trial while nilotinib (NCT02954978; NCT03205488) and ambroxol (NCT02941822) are in phase Ⅱ trail. Data source: http//clinicaltrials.gov; November 2020.

3.1. Targeting AMPK

5ʹAMP-activated protein kinase (AMPK) is a key factor in the regulation of autophagy and the main sensor of intracellular energy stress, which is able to perceive and respond to the energy changes, and crucial to energy homeostasis80,81. With seven isoforms (α1, α2, β1, β2, γ1, γ2 and γ3), AMPK is a heterotrimeric protein kinase involving the structure of α catalytic subunit, scaffold β subunit, and regulatory γ subunit. In theory, AMPK could form up to 12 possible heterotrimers with different regulatory functions. The catalytic kinase domain was in the α subunit and the main activation site of AMPK was Thr17282. The cell function of AMPK depended on the level of ATP a lot. Alterations in the ratio of AMP/ADP/ATP would lead to the activation or inactivation of AMPK. AMPK activation had a wide range of neuroprotective effects to increase cell survival against many stressors, including starvation, hypoxia, ischemia, and excitotoxicity83, 84, 85, 86. Therefore, AMPK activation has been extensively explored as a neuroprotective strategy for PD treatment.

Hitherto, several small-molecule compounds that can regulate AMPK activity have been identified as their cytoprotective regulation in the PD models. Metformin (1) is the first choice for type 2 diabetes mellitus treatment (oral bioavailability, F: 40%–60%, Fig. 3A). In addition to the anti-diabetic effect, its neuroprotective efficiency in the MPTP-induced PD models was also discovered remarkably in 201443. Yet, this study failed to explain the potential mechanism of its neuroprotection and drug target. Since compound 1 is a well-known AMPK activator, subsequent studies investigated whether metformin prevented DA neurons from MPTP-induced neurodegeneration by activating AMPK87,88. Lu et al.89 showed that 1 directly activated AMPK and initiated downstream protective signals to slow down DA neuronal apoptosis. AMPK inhibitors could eliminate the cytoprotective effect of 1 in SH-SY5Y cells, proving that AMPK activation was necessary for this effect on DA neurons. Moreover, it was also illustrated that 1 could induce autophagy and reduce the accumulation of α-syn. Resveratrol (2), a stilbene found in grapes and red wine, had many benefits, covering reducing oxidative stress, inflammation, and mitochondrial damage, regulating stem cell growth, neuroprotection, and inducing autophagy (Fig. 3A)90, 91, 92, 93. Autophagy and mitochondrial phagocytosis induced by 2 had cytoprotective and antioxidant effects in various cell-based PD models31,44,94, 95, 96. It has been reported that 2 can produce a specific neuroprotection by inducing autophagy to clear α-syn. In overexpressing wild-type and mutant α-syn PC12 cells and rotenone-exposed SH-SY5Y cells, treatment with 2 raised the phosphorylation level of AMPK active site Thr172 and the SIRT1 deacetylation enzymatic activity, representing that 2 enhanced the degradation of α-syn by activating autophagy through the AMPK/SIRT1 signaling pathway44.

Figure 3.

Figure 3

(A) The structures of compounds targeting AMPK. (B) The structure–activity relationship (SAR) of A-769662 and GSK621.

The aforementioned studies of 1 and 2 directly show that they are two commonly used and the most representative autophagy activators targeting AMPK in the PD models. Besides, some small-molecule synthetic compounds with similar neurological effects have been also discovered, such as A-769662 (3), GSK621 (4), rosuvastatin (5), FCPR16 (6) and Temozolomide (7, Fig. 3A)31,55,66,74,97,98. Compound 3 is an AMPK agonist with a structure of thienopyridone, which can reversibly activate AMPK with EC50 values of 116 nmol/L75. The important role of AMPK in the regulation of lipid and glucose metabolism aroused pharmaceutical companies to develop AMPK activators for the treatment of various metabolic diseases. The non-nucleoside thienopyridone compound A-592107 (4-1) with EC50 values of 38 μmol/L was firstly obtained from a molecule database of more than 700 thousand compounds and identified as a direct AMPK activator75. Subsequently, sub-micromolar compound 3 was discovered in the lead compound optimization processes. Compound 3 could inhibit the dephosphorylation of Thr172 and provide moderate allosteric activation, which was similar to the AMP-induced pathway75. Nonetheless, the oral bioavailability of 3 was poor (F = 7%). To improve oral absorption and selectivity, the GlaxoSmithKline (GSK) researchers took advantage of compound 3 as a template to reconstruct and identified a new type of AMPK activators of pyrrolo-pyridine analogs97. Biological replacement of the thiophene ring in 3 with pyrrole reached compound 4-1 imparted it better oral bioavailability (F = 10%). Replacing hydrogen in the pyrrole ring with chlorine failed to improve blood clearance or permeability, but the oral bioavailability could be improved. Afterwards, a series of different functional group substitutions on R1 and R2 increased efficacy and reduced blood clearance. Although the substitution of 3-methoxy-2-phenol at the R2 position slightly reduced oral bioavailability, it could obviously reduce the blood clearance rate. The presence of a cyano group at the R1 position of the pyridone ring resulted in poor permeability owing to its acidity. Replacing cyano group at R1 with phenyl imparted 4-3 better permeability and oral bioavailability97. Compound 4 was derived by changing the pyrrolopyridone core of 4-3 to pyrrolopyrimidone (Fig. 3B). Compound 4 was slightly less effective than 3 in continuously activating AMPK recombinant heterotrimers in vitro, but 4 was more potent than 3 in inducing AMPK activation in cell assays99. In the same study in 2019, Nicolas31 found that the two effective AMPK activators 3 and 4 were able to induce autophagy by activating AMPK and decrease the content of α-syn in over-expressing α-syn SH-SY5Y cells, showing similar neuroprotective effects. Besides, compound 5 is a statin drug, which can competitively inhibit HMG-CoA reductase and is widely used as a cholesterol-lowering drug. A study in 2017 showed that 5 reduced rotenone-induced neurotoxicity in SH-SY5Y cells through autophagy regulation and accelerated the clearance rate of α-syn55. This study published in 2017 provided the first experimental evidence for the neuroprotective role of statins by enhancing the autophagy pathway in the PD models. N-(2-Chlorophenyl)-3-cyclopropylmethoxy-4-difluoromethylbenzamide (5) is a phosphodiesterase 4 (PDE4) inhibitor and plays a beneficial role in the CNS diseases due to its anti-inflammatory and anti-apoptotic characteristics. Compound 5 effectively protects SH-SY5Y neuronal cells from MPP+-induced oxidative damage by activating autophagy of AMPK and 5 has almost no emetic side effects of general PDE4 inhibitors66. Furthermore, the natural product caffeine (8) was found to reduce α-syn-induced autophagy defects by up-regulating AMPK (Fig. 3A)76. Many central mechanisms underlying PD pathology could be regulated by activating AMPK. However, given the broad regulatory role of AMPK, it is necessary to identify specific downstream pathways that are most beneficial for the treatment of PD, so that the pathways can be targeted more selectively without activating other harmful pathways. Consequently, activation of PD-related AMPK-dependent pathways may reduce neurodegeneration and ultimately inhibit PD progression.

3.2. Targeting mTORC1

Rapamycin (9) is one of the most widely applied small molecules in the mTOR dependent method for PD therapy (Fig. 4). Compound 9, an autophagy regulator, is an antifungal macrolide isolated from strains of streptomyces hygroscopicus analogues. Compound 9 binds to the FKBP12 receptor in the cell and forms a complex. Subsequently, the complex can directly integrate with mTOR, which prevents the combination of mTOR and RAPTOR, blocks the signal transduction of mTORC1, and activates autophagy100,101. The biological function of 9 is diverse, such as antifungal, immunosuppression, antitumor, neuroprotection, and anti-aging effect77,102,103. Compound 9 and its derivatives are mainly used as immunosuppressants and anti-tumor drugs in the clinic. Given that mTOR is the central hub of the signal network in the cell, it has been extensively explored. Surprisingly, there is a close relationship between 9 and PD. Compound 9 is proved to increase the formation of autophagosome and the efficiency of α-syn clearance both in vivo and in vitro. Compound 9 can not only prevent the death of dopaminergic neurons but also improve the motor function of transgenic mice77,101,104. Furthermore, treatment with 9 increases the level of the lysosomal structural protein LAMP1 in the mouse brain, demonstrating that it is capable of increasing the lysosomal biogenesis and avoiding the accumulation of autophagosomes and neuronal potential toxicity105. Some analogues of 9, such as CCI-779 (10) and AP23573 (11), were clarified to exert similar neuroprotective effects (Fig. 4)106, 107, 108. Besides, long-term treatment with 9 could inhibit mTORC2, which may stimulate other important cellular pathways such as affecting cell survival mechanisms. Nonetheless, compound 9 can cause a wide range of side effects including oral and respiratory infections, stomatitis, leukopenia, hypertriglyceridemia, hypercholesterolemia, and immunosuppression100,109. Obviously, 9 and its analogues are unsuitable for long-term and high-dose therapy due to their adverse effects. Generally speaking, owing to their profitable action on different PD pathological models, these studies provide important theories on the role of autophagy and prove that inducing autophagy is a superior strategy in experiments.

Figure 4.

Figure 4

The structures of compounds targeting mTOR.

According to the researches on PD therapy, most compounds targeting mTORC1 are natural products including oxindole alkaloids corynoxine (12) and iridoid glycosides glycosides loganin (13, Fig. 4)78,79. In PC12 cells with high expression of wild-type (WT) and mutant (A53T) α-syn, both of their α-syn were significantly degraded after the treatment with 12. The effect was blocked by the autophagy inhibitor 3-MA and the lysosomal inhibitor CQ, suggesting that the clearance action occurred through autophagy78. Compound 12 was identified to reduce phosphorylated mTOR (Ser2448), AKT (Ser473), and p70S6K (Thr389), elucidating that it induced autophagy by inhibiting the AKT/mTOR pathway. In addition, synthetic small molecules targeting mTORC1 also had neuroprotective effects, such as the PI-103 (14) with a structure of pyridylfluoropyrimidine (Fig. 4)74. Compound 14 was initially discovered through high-throughput screening, which had an arylmorpholine pharmacological group to form a key hydrogen bond with the framework Val882 amide of class I PI3K. It could effectively and competitively inhibit all class IA, α, β and δ with IC50 values of 2, 3 and 3 nmol/L respectively which indicated it was a selective inhibitor of class I PI3K110. Subsequently, a study characterized the target selectivity of 14 and proved its effective inhibition of mTORC1 with the IC50 values of 20 nmol/L. Therefore, compound 14 was proved to be a dual-target inhibitor of mTOR and PI3K111.

Among the drugs targeting mTOR, some natural compounds emerged from traditional medicines and achieved promising outcomes in inducing autophagy for PD treatment. Small-molecule compounds are also constantly designed and synthesized. More importantly, it ignites our more attention to keep the balance between mTOR and other cellular pathways to reduce other side effects, hence mTOR inhibitors can be applied more effectively in the treatment of PD.

3.3. Targeting ULK1

UNC-51-like kinase 1 (ULK1), the ortholog of yeast Atg1, is the only serine-threonine kinase and primase, which participates in the formation of early membrane structures of phagocytic vesicles and plays a vital role in the autophagy process112,113. When ULK1 was activated, beclin-1 was phosphorylated respectively at Ser15 and Ser29, triggering the lipid kinase activity of phosphoinositide-3-kinase class 3 (PIK3C3). This upregulated the expression of phosphatidylinositol 3-phosphate (PI3P) and participated in the recruitment of autophagy proteins for autophagosome formation, enabling the autophagy process to eliminate α-syn oligomers produced in the pathogenesis of PD114. A novel activator, BL-918 (15), was able to activate ULK1 effectively45. Firstly, this study applied the solvent accessible surface (SAS) calculation to obtain possible binding sites for the ULK1 activator. After high-throughput screening was used to obtain the lead compound 15-1, its structural optimization was rationally promoted based on the binding site. The bridged oxygen atom was substituted with an ester group that can form another hydrogen bond with Tyr89. The imidazole ring was converted to a piperazine ring to obtain 15-2, which improved the EC50 by 4.8 times and the Emax by 2.1 times. Afterwards, d-(−)-2-phenylglycine skeleton was introduced into the compound structure to obtain 15-3. Compared with 15-2, the ULK1 kinase activity of 15-3 was only slightly increased, while the autophagy activity was promoted by two times. The amino acid residues on the edge of the active pocket were not fully utilized yet, such as Asn86 and Tyr89. As a consequence, the ureido group was used as a linking group to gain the optimal compound 15 (Fig. 5). In contrast to 15-3, the EC50 and Emax of 15 were both improved by 1.7 times and 1.3 times (EC50: 40.80−24.4 nmol/L, Emax: 0.742 ± 0.028−1.000 ± 0.037), and the autophagy activity was also increased by 1.5 times45. Compound 15 could induce cytoprotective autophagy through the ULK1 complex in SH-SY5Y cells largely. Apart from that, it could also exert its neuroprotective role by targeting ULK1-regulated autophagy in the MPTP-induced PD mouse models. This study provided precious experience for designing and synthesizing new compounds that targeted autophagy for PD treatment.

Figure 5.

Figure 5

(A) The structure optimization process of BL-918. (B) The structures of compounds targeting IMPase.

3.4. Targeting IMPase

The regulation of autophagy via the inositol signaling pathway is mTOR-independent115. The G protein-coupled receptor-mediated phospholipase C (PLC) was able to activate this pathway, causing the formation of InsP3 and diacylglycerol. InsP3 bound to the InsP3 receptor on ER and the calcium was released into the cytoplasm. Because of the interaction with 5-phosphatase and inositol polyphosphate 1-phosphatase (IPPase), InsP3 formed inositol phosphate, which was then hydrolyzed to free inositol by inositol monophosphatase (IMPase). The accumulation of intracellular free inositol or InsP3 could inhibit the formation of autophagosomes. Drugs that could lower the inositol would induce autophagy and promote the elimination of autophagy substrates without inhibiting mTORC1 activity115. Besides, there are also two anti-epileptic and mood-stable drugs named valproate (16) and carbamazepine (17, Fig. 5B). A study highlighted that these two drugs were able to inhibit inositol monophosphatase and reduce free inositol and the level of inositol 1,4,5-triphosphate which made them hopeful autophagy enhancers46. Derived from this, in the subsequent rotenone-induced human neuroblastoma SH-SY5Y cell model, not only could both drugs enhance autophagy and reduce rotenone-induced mitosis and apoptosis, but also reduce the mitochondrial membrane potential and the activity of oxygen production and enhance cell viability significantly46. However, a study in 2019 found a contradictory result that compound 17 did not significantly activate autophagy in the PD model of LUHMES cells and failed to protect cells from α-syn-induced toxicity74. Meanwhile, compound 17 did not affect inositol metabolism in the rat cortex after long-term oral medication116. Correspondingly, a case-control study in the United Kingdom indicated that there was no relationship between 17 intake and the risk of PD occurrence117. The two opposite experimental results above showed that 17 can induce autophagy theoretically, but the compound is still not effective enough in neuronal cells yet. It also implied that the effects of drugs in various models may be different, and drug development was a complex and tortuous process. On the contrary, another small-molecule inhibitor of IMPase, L-690,330 (18), produced a promising result in LUHMES cells (Fig. 5B). Compound 18 could inhibit IMPase in LUHMES cells and reduce free inositol levels. It also could activate autophagy and protect cells from α-syn-induced toxicity, which had a good research prospect74. As can be seen from the previous description, drugs targeting the inositol pathway are not necessarily effective enough. And whether they will cause other side effects still need to be continuously verified. Compared with long-term using one single inhibitor, moderate combination therapy with IMPase and mTOR inhibitors may be safer for PD treatment.

3.5. Targeting LRRK2

Raised attention in 2004, LRRK2 was a multidomain protein that displayed dual kinase and GTPase activities. Overexpression or mutation of the LRRK2 gene, especially G2019S missense mutation, will increase the kinase activity, leading to familiar autosomal dominant PD and sporadic PD118. Thus, kinase inhibition is expected to be a therapeutically advantageous target for treating PD119. LRRK2 is inevitably associated with PD through the lysosomal pathway of autophagy120. The chemical inhibition of LRRK2 kinase activity was investigated to stimulate atypical autophagy in the H4 glioma cell line and primary astrocytes, independent of mTOR and ULK1, but dependent on the presence of active beclin-1 complex121. Also, when LRRK2 phosphorylated endophilinA at Ser75, endophilinA induced autophagy and further regulated membrane curvature and promoted autophagosome recruitment122. Given LRRK2 has close connection with autophagy, the inhibition of autophagy which relays on LRRK2 may accelerate neurodegeneration including PD121. In addition, University of Oxford's abundant data supported Lrrk2 siRNA knockdown effectively induced autophagy and prevented cell death caused by starvation conditions123. All these data reveal that LRRK2 is involved in the regulation of autophagy and uses as a hopeful drug target for PD, whereas the exact mechanism of LRRK2 affecting autophagy still needs further investigation.

To date, small-molecular LRRK2 inhibitors have become a hotspot for research in PD. As mentioned above, mutations in LRRK2 can enhance kinase activity. Based on different LRRK2 mutations, various treatments have been developed, such as LRRK2 kinase inhibitors (for G2019S and I2020T mutations), LRRK2 GTPase inhibitors (for R1441G/C/H, Y1699C mutations), and LRRK2 dimer inhibitors (for all mutations)124. Until now, LRRK2 kinase inhibitors are considered as the most prospective candidate drugs. It's generally achieved by manufacturing competing ATP compounds in the ATP binding pocket of LRRK2. The DYG motif of the ATP binding site activating loop is the key to control the transformation of proteins from the active form (DYG-in) to the inactive form (DYG-out), especially the glycine residue (G2019) in the motif makes the conformation more flexible. When G2019 in LRRK2 mutates to serine, the residue may interact with other residues in the ATP binding site (such as Asp1194 in the catalytic ring) to keep the kinase in its active form (Fig. 6A). Simultaneously, D2017 in the DYG motif interacts with ATP β-phosphate. According to this general mode of action, more than 50 specific LRRK2 kinase inhibitors which own different scaffolds with excellent selectivity and potency have been explored, including targeting G2019S mutation47. Given that aminopyrimidine derivatives are the earliest identified and the most widely studied LRRK2 inhibitors, we discuss the SAR of these compounds concretely and deeply. As the first found LRRK2 specific inhibitor, LRRK2-IN-1 (19) could stimulate the dephosphorylation of LRRK2 at Ser910 and Ser935 and had been used as a lead compound to develop selective and blood−brain barrier (BBB) permeable LRRK2 inhibitors (Fig. 6B)47,48. It had an aminopyrimidine scaffold which was proved an effective chemotype for the discovery and optimization of LRRK2 inhibitors. Compound 19 was particularly potent against both the wild-type Lrrk2 (IC50 = 13 nmol/L) and G2019S Lrrk2 mutation (IC50 = 6 nmol/L)48. TAE684 (20) was initially used as an inhibitor of anaplastic lymphoma kinase (ALK), and later found to be a potent inhibitor of LRRK2 kinase activity. It was confirmed with wild-type Lrrk2 and G2019S IC50 reported as 7.8 and 6.1 nmol/L (Fig. 6B)125. Strikingly, compound 20 had favorable selectivity against Lrrk2 A2016T mutant (IC50 = 93.3 nmol/L), in contrast to 19 against Lrrk2 A2016T mutant (IC50 = 2450 nmol/L). The structure of the kinase domain of LRRK2 had not been reported yet. Therefore, researchers constructed the molecular model based on the crystallographic structure of ALK125. It was surmised that the isopropyl sulfone moiety of 20 avoided the steric clash with A2016T residue which may conflict with the anthranilic acid ring of 19 (Fig. 6B)125.

Figure 6.

Figure 6

(A) An overview of ATP binding site of LRRK2 (PDB:6VP6). The kinase is illustrated with dark blue ribbons, and the key amino acid residues are depicted in black solid lines. (B) The SAR of aminopyrimidine derivatives LRRK2 inhibitors. (C) The structures of compounds 2527.

Although these LRRK2 small molecule inhibitors discussed above showed relatively high inhibiting LRRK2 activity, none of them had achieved enough CNS exposure which limited their application in mouse PD models, suggesting urgency of optimizing the BBB penetration ability126. HG-10-102-01 (21, Fig. 6B) was reported as the first brain penetrant LRRK2 inhibitor with aminopyrimidine scaffold and showed more potent inhibitory ability against A2016T + G2019S mutant due to weaker steric clash with A2016T126. With the docking model with 21 and its derivatives, JAK2, the homology model of LRRK2, showed that the C-5 group with increased lipophilicity and size could form van der Waals force with Met1947 gatekeeper side chain better, such as C-5 chlorine127. Based on a more accurate model, it could be deduced that the carbonyl oxygen of the amide group formed weak hydrogen bonds with the guanidine side chain of Arg1957. The aniline ring bound to the flat hydrophobic cleavage bond along the hinge near the opening of the ATP binding site. 4-Morpholinoamide group pointed to the side chain of Phe1883 and C-4 N-methyl filled the hydrophobic cavity127. Compared with 20, removal of 4-anilino substituents retained the inhibitory effect of LRRK2 but improved the permeability of CNS. Ortho-substitution of aniline ring could significantly enhance the selectivity of these compounds. The results showed that small size methoxy substitution was the most ideal127. Further optimization strategy focused on the substituents of the aminopyrimidine C-4 and C-5 position. After analyzing the structure–activity relationships of C-4 and C-5, the optimal C-4/C-5 combination was C-4 N-methyl/C-5 trifluoromethyl substituted aminopyrimidine127. These substitutions led to the discovery of compound 22 (Fig. 6B). Intramolecular hydrogen bonds were formed between the F of the C-5 trifluoromethyl group and the C-4 N-methyl H, as same as the aniline ring N−H and the methoxy group. Simultaneously, the increase of trifluoromethyl lipophilicity was also conducive to the BBB penetration. As the oxidation potential of the pyrimidine ring was relatively lower, trifluoromethyl substitution had a better DMPK distribution than other C-5 substituents. GNE-7915 (23) was identified through the C-5′ substitution with fluorine (Fig. 6B). The substitution of N-ethyl group replacing the C-4 N-methyl group reduced the clearance ratio in vivo. Beyond that, the C-2′ methoxy group occupied the space near Leu1949, which imparted 23 good inhibitory activity. Further structural modification of aminopyrimidine scaffold LRRK2 kinase inhibitors should emphasize reducing the size and improving the penetrating ability of BBB while maintaining the inhibition of LRRK2 activity. The substitution of the aniline ring with pyrazole isomers led to the discovery of GNE-0877 (24, Fig. 6B)47,128,129. Removing amide functional groups and maintaining low molecular weight could effectively enhance brain permeability. Pyrazole substitution also eliminated the formation of potential aniline derived ortho-quinoneimine active metabolites. Through the SAR analysis of a series of gem-disubstituted cyano pyrazole LRRK2 inhibitors, it was found that 24 was more effective against pLRRK2 and water-soluble compared with 23. The two methyl groups on the pyrazole ring had better van der Waals contact with the hydrophobic residue of LRRK2, and the cyano group could form a favorable electrostatic interaction with the side chain of Arg1957, which increased the LRRK2 inhibiting activity of 24. Another several scaffolds also have been developed as the highly potent LRRK2 inhibitors, including pyrrolopyrimidine series represented by PF-06447475 (25), PF-06685360 (26), and 3-pyrimidinyl-indazole series represented by MLi-2 (F = 41%, 27, Fig. 6C)130, 131, 132, 133, 134. Besides, there are some LRRK2 small molecular inhibitors with unpublished structures such as DNL151 and DNL201 (NCT03710707 and NCT04056689). So far, these LRRK2 inhibitors showed great therapeutic effect in preclinical PD models135, 136, 137. For instance, compound 23 showed enhanced DA release and recovery in R1441G transgenic mice models allowing us to ascertain its possibility for treating PD135. LRRK2 inhibitors were also found to rescue Lrrk2 G2019S mutant dopaminergic phenotypes138. Overall, LRRK2 inhibitors play a key role in mediating neurodegenerative phenotypes and certainly fuel our understanding of the efficacy of the LRRK2 inhibitors in treating PD.

Although direct inhibition of ATP binding pocket has been proved to be a relatively successful method for identification of LRRK2 inhibitors, other methods for regulating LRRK2 are also possible. To answer the question of whether LRRK2 inhibitors are effective in the treatment of PD, experts have made great efforts to promote these small molecule inhibitors into clinical application. However, LRRK2 is not a brain-specific protein, highly expressed in the kidney and lung which may be out of target. It's still tough to optimize kinase selectivity, CNS permeability and reduce lung and/or kidney side effects simultaneously. Another obstacle may be that there is no reliable PD model with LRRK2 dysfunction so far, which brings great challenges to clinical trials. Although knockdown of LRRK2 and few LRRK2 inhibitors can induce autophagy and rescue neuron cell death caused by autophagy defects, whether the majority of LRRK2 inhibitors can produce the same effect in autophagy remains to be further characterized139. Denali Therapeutics49 recently reported two LRRK2 small-smolecule inhibitors, DNL151 and DNL201, have entered phase I clinical trials which provide impetus for the further development of LRRK2 small-molecule inhibitors. It can be ascertained that with the unremitting efforts of scientists, the above problems are expected to be overcome. More LRRK2 promising inhibitors will be applied in the clinic, such as 26 and 27 which are in the preclinical studies currently140. In conclusion, the application of LRRK2 kinase inhibitors in the autophagy-lysosome pathway may provide a novel thought for the treatment of PD.

3.6. Targeting beclin-1

Beclin-1, known as ATG6, is highly conserved among eukaryotes and belongs to the ATG protein family. As an important molecule for regulating autophagy, its overexpression can reduce apoptosis and enhance autophagy50,141. Beclin-1 involves the nucleation of autophagic vesicles (the formation of phagocytes) during the process of autophagy induction. To initiate nucleation of vesicles, beclin-1 interacted with its binding partners such as PI3KC3/Vps34, Ambra1, Vps15, and Atg14L. After complexing with beclin-1, PI3KC3/Vps34 was activated to produce its unique product, phosphatidylinositol 3-phosphate, which was in charge of inducing autophagy51. Spencer's team142 suggested that autophagy was activated by beclin-1 which could reduce the accumulation of α-syn in PD. As an inhibitor of the serine protease prolyl oligopeptidase (PREP), KYP-2047 (28) can induce the expression of beclin-1 (Fig. 7A). The number of autophagy markers LC3BII then increased, which further enhanced autophagy and clearance of high molecular weight oligomeric α-syn52. Previous studies pointed out that the 4-phenylbutyryl-l-prolyl-pyrrolidine skeleton of 28 was preferred to reduce the dimerization of α-syn compared with other PREP inhibitors52,143. Moreover, natural products such as isorhynchophylline (29), corynoxine B (30) and glycyrrhizic acid (31) could also strengthen the cytoprotective autophagy by up-regulating the expression of beclin-1 and rescue the survival of neuronal cells (Fig. 7)53,54,56,144,145. Besides autophagy, beclin-1 also regulates apoptosis. In view of the crosstalk between autophagy and apoptosis which involves regulating cell survival and death, beclin-1 possesses great research value146.

Figure 7.

Figure 7

The structures of compounds targeting beclin-1.

3.7. Targeting TFEB

Lysosomal formation and autophagy were promoted when transcription factor EB (TFEB) combined with lysosomal joint expression and regulation (CLEAR) elements147,148. TFEB subcellular localization was regulated by mTOR-mediated phosphorylation thereby affecting TFEB activity149. TFEB could bind to the promoter regions of several autophagy genes and induce autophagosome biogenesis and autophagosome-lysosome fusion148. Interestingly, degradations of numerous autophagy substrates, including α-syn, were enhanced by the overexpression of TFEB. The representative compound targeting TFEB was curcumin (32), a natural pigment derived from the root of the turmeric herb (Fig. 8A). Compound 32 had two aromatic rings involved ortho-methoxyphenol hydroxyl groups, which linked symmetrically with a β-diketone. Experiments showed that 32, as a neuroprotective agent, had beneficial effects on the nervous system and PD models. Firstly, it had strong antioxidant properties and significant anti-inflammatory activity with the bioavailability of57,58. In the meantime, compound 32 could inhibit the aggregation of α-syn and prevent the accumulation of LB in vitro to reduce the toxicity of α-syn oligomers in cells and degeneration of DA neurons59. Besides, 32 was to promote the recovery of autophagy by activating TFEB, leading to the reduction of cellular oxidative stress, neurotoxicity, memory loss, and dyskinesia150,151. Moreover, compound 32 was safe, non-toxic, cheap, and easily available. It could also effectively penetrate the blood−brain barrier and neuronal membrane. Thus, compound 32 was a promising candidate for the treatment of PD. Even though the clinical application of 32 was limited due to its instability and poor metabolic properties. Chemical modification was an effective strategy to improve its biological activity. The β-diketone moiety of 32 appeared to be a series of aldehyde-reductase ketone-specific substrates in vivo, which could be rapidly broken down152. Analogues which replaced the β-diketone part could reduce degradation rate and enhance stability in buffers of different pH in vitro. Meanwhile, half-life (t1/2), clearance rate (CL), the area under the curve (AUC), and other pharmacokinetic properties were dramatically improved153. On this basis, a series of 32 analogues were developed, among which CNB001 (33), curcumin analogues C1 (34), and E4 (35, Fig. 8A) also showed neuroprotective effects in PD models60, 61, 62. Besides compound 32 and its analogs, trehalose (36, Fig. 8B), as an autophagy activator, has been extensively studied in various PD models63. Compound 36 could ameliorate different disease phenotypes. However, the specific autophagy activation pathway and mechanism of 36 were not so clear yet. Recently, compound 36 was reported to induce autophagy through lysosomal-mediated TFEB activation in a model of motor neuron degeneration154. In sum, TFEB has become an emerging target to enhance ALP and up-regulate the TFEB expression in the central nervous system through gene therapy or pharmacological activation.

Figure 8.

Figure 8

(A) Curcumin and its analogues replaced the β-diketone part. (B) The structures of trehalose and XCT790. (C) The structures of compounds targeting GCase.

3.8. Targeting GCase

The regulation of lysosomes has recently become an attractive strategy to selectively stimulate ALP in PD models. One of the remarkable methods is that to target specific lysosomal enzymes directly (such as glucocerebrosidase GCase) to stimulate the lysosomal degradation of α-syn. Much evidence suggests that the level and activity of GCase decreased, and GCase and α-syn levels of the brain are inverse ratio in idiopathic PD105. Therefore, the enhancement of GCase activity may be also applied in the treatment of PD. However, since GCase cannot transverse the blood−brain barrier, the direct application of enzyme replacement therapy is ineffective for PD155. Later, it was proposed to develop small-molecule drug chaperones which could be used as an alternative therapy for PD156,157. Some such drugs were designed to bind to mutant GCase, correcting its misfolding and promote transport to lysosomes, which enabled to improve the GCase activity and the lysosomal function. Ambroxol (37) was a small molecule chaperone used for respiratory diseases (Fig. 8B). McNeill et al.64 found that it could increase GCase activity and reduce oxidative stress in PD fibroblasts with GBA1 mutations. Furthermore, 37 restored levels of cathepsin D, LIMP2, and saposin C, which was crucial to the activity of GCase65. Compound 37 could not only increase the GCase activity significantly in lymphocytes and penetrate the blood−brain barrier but also reduce the level of glycosphingosine in cerebrospinal fluid and improve myoclonus and seizures in patients. The company PRO.MED.CSA recently completed a phase II non-randomized and uncontrolled clinical trial approved by the FDA of 37 (NCT02941822)158. According to reports, oral 37 can be detected in the blood and cerebrospinal fluid of PD patients after 186 days, and the patient did not have any serious adverse reactions. Besides, some other small-molecule chaperones, such as isofagomine (38) displayed similar effects (Fig. 8C)67. Nonetheless, one disadvantage of these chaperones is that they inhibit GCase activity by binding to the catalytic site of the enzyme. Thus, it suggests that the balance between chaperone function and inhibitory activity should be carefully considered when utilizing these compounds, requiring the development of those drugs without binding to the GCase catalytic site159. Consequently, a non-inhibitory GCase small molecular chaperone NCGC607 (39) was identified through high-throughput screening (Fig. 8C). This chaperone has been proved to restore GCase activity and protein levels while reducing α-syn levels in dopaminergic neurons derived from iPSCs (induced pluripotent stem cells)68. These studies of PD have demonstrated that small molecular chaperone enhancers of GCase can improve the lysosomal function and the α-syn clearance, suggesting the great possibility of targeting GCase to selectively stimulate the ALP pathway to treat PD.

3.9. Targeting ERRα

The nuclear receptor (NR) superfamily in eukaryotic cells represents a group of important and diverse transcriptional regulators, part of which are activated by endogenous hormones or ligands in animals160,161. However, a large number of receptors are not activated or regulated by physiological/endogenous ligands which are called orphan nuclear receptors (ONRs). The estrogen-related receptor α (ERRα) is the first ONR to be discovered which participates in the regulation of various biological functions such as energy metabolism and is highly expressed in skeletal muscle, kidney, brain, heart, and other tissues that require energy162, 163, 164. Due to its crucial action in metabolic homeostasis, ERRα has gradually been developed into a considerable target for the treatment of cancer and metabolic diseases. XCT790 (40) is a thiadiazole acrylamide derivative and the most selective inverse agonist for ONR. ERRα has been identified as one of its targets (Fig. 8C)69. Researches elucidated that 40 eliminated α-synuclein aggregated in human neuronal cells in an autophagy-dependent method70. It also showed that autophagy induced by inhibiting the activity of ERRα under nutrient-rich conditions was carried out through an mTOR-independent mechanism. Under the normal condition, ERRα was localized on autophagosomes. After autophagy was induced by compound 40, this localization was lost and accompanied by an increase in the autophagosome biogenesis, indicating that ERRα may regulate autophagy through subcellular localization dynamics of ERRα. Concurrently, in a preclinical mouse model of PD, 40 cleared toxic protein aggregates by inducing autophagy, alleviated motor coordination disorders, and exerted a neuroprotective role on dopaminergic neurons in the substantia nigra70. The data about 21 mentioned above is strong proof that ERRα will be an attractive target for PD.

3.10. Targeting c-ABL

C-Abelson (c-ABL) non-receptor tyrosine kinases can regulate autophagy by promoting the transport and function of lysosomal components165. C-Abelson was shown to be activated in preclinical PD models and brain samples from PD patients. Thus, inhibition of c-ABL displays great potential in PD therapy166,167. PD180970 (41) is a potent ATP-competitive BCR-ABL inhibitor, which could inhibit c-ABL activity at nanomolar concentrations (Fig. 9A)168. Compound 41 also had a neuroprotective effect, which could induce autophagy in an mTOR-independent manner and reduce the toxicity mediated by α-syn in mammalian cells71. In vitro, it was also demonstrated that treatment with 41 could relieve neuroinflammation and regulate the release of cytokines in microglia and neuron-microglia co-cultures. Meanwhile, 41 protected dopaminergic neurons and improved motor behavior defects in preclinical mouse models of PD by inducing autophagy and aggregation clearance71. Imatinib (42) and nilotinib (43) are another two tyrosine kinase inhibitors (TKI) that can selectively inhibit BCR-ABL (Fig. 9A). Both of them have similar structures with the aminopyrimidine mother nucleus. Compound 42, as the first known ATP-competitive inhibitor, is able to inhibit BCR-ABL kinase with high selectivity169. In the early 1990s, the phenylaminopyrimidine derivative 42-1 was identified as a potential lead compound for protein kinase C (PKC) inhibitors, followed by SAR study, and compound 42 was identified consequently. Firstly, adding a pyridyl group to the 3′-position of 42-1 pyrimidine could enhance its cell activity. Then various functional groups were tested as substituents in the benzene ring. It was also found that the amide group had an inhibitory effect on tyrosine kinase. In addition, the analysis of the SAR showed that the presence of a methyl group at the ortho position of the amino group increased the selectivity of BCR-ABL. However, the modified molecule still performed poor solubility in water and oral bioavailability, which were significantly improved by the introduction of N-methylpiperazine groups. At the same time, a spacer benzene ring was introduced between the amide group and N-methylpiperazine, eliminating its mutagenic possibility. Thus, these structural modifications eventually led to the appearance of 42 (Fig. 9B)169. Most of the interactions between 42 and kinases were weak van der Waals force (VdW). Although six high-energy hydrogen bonds (Hb) also formed, each hydrogen bond accounted for a relatively high portion of the total binding energy of kinase inhibitors. If one of the hydrogen bonds was broken because of the mutation of hydrogen bond donor/acceptor residues, its energy contribution would be lost and the competition between the free energy of the dissociated state and the bound state would increase. In case of it, the interaction between 42 and mutant kinase would be lost due to unfavorable thermodynamic factors. This is the reason for the drug resistance of 42, so the second-generation TKI represented by 43 was developed170. Based on the structure of 42, replacing the piperazine ring with 3-methylimidazole and adding trifluoromethyl to the anilinocarbonyl substituent, by inverting the amide linker, were used to increase the number of VdW interactions (Fig. 9B). Thus, the result was that 43 pairs showed higher efficacy to almost all mutations of drug resistance170. The overbased N-methylpiperazinyl group in 42 had stronger desolvation/deprotonation and constraining force of binding surface, while the methylimidazole group in 43 could be stacked in the hydrophobic pocket formed by amino acid residues. The nitrogen atom was exposed to the solvent as well. To be specific, the pyridyl and pyrimidinyl groups of 42 contributed relatively much energy, while the trifluoromethyl and methylimidazole groups of 43 contributed considerably much energy. Therefore, in contrast to 42, mutations linked with pyridyl and pyrimidinyl groups had less effect on the overall affinity of 43. With a better topology between 43 and kinase, the damage of inhibitor binding with the key mutated residues related to Hb interaction could be avoided (Fig. 9C). Despite these modifications, the selectivity and potency of 43 were not destroyed and even improved compared with 42 (IC50 values of 400 and 45 nmol/L, respectively)171. The two compounds also had similar effects in the PD models, which could induce autophagy and increase the clearance rate of α-syn72,83,172. It is worth noting that 43 is undergoing two clinical trials (NCT02954978 and NCT03205488). One is to research the clinical efficacy, and the other is to test the safety and tolerability in PD. Furthermore, a small open-label trial has shown that 43 is safe and can improve the motor function in PD patients73. Splendid clinical manifestations of 43 provide favorable evidence for targeting the autophagy pathway in the treatment of PD.

Figure 9.

Figure 9

(A) The structures of compounds targeting c-ABL. (B) The discovery and structure optimization process of imatinib and nilotinib. (C) The interaction modes of imatinib and nilotinib with c-ABL (PDB:2OIQ,3CS9). The key amino acid residues are illustrated with pink, and imatinib and nilotinib are depicted in blue and yellow respectively. The hydrogen bonds are shown in black dashed lines.

4. Conclusions

In the past few years, the toxic aggregates produced by impaired autophagy has been recognized as one of the pathogenesis of PD, making it possible to target diverse regulators of autophagy (such as AMPK, mTORC1, ULK1, IMPase, LRRK2, beclin-1, TFEB, GCase, ERRα, C-Abelson). A variety of small-molecule activators or inhibitors correlated with such potential druggable targets have been developed, which may play their neuroprotective effects in different types of PD models. However, many of these small-molecule compounds are only used as tool probes for PD research. There is still a big challenge about thorough understandings of autophagic mechanisms in the pathogenesis of PD and transforming the current small molecules into the future clinical practice. The general hypothesis is based upon the fact that autophagic flux decreases in the pathogenesis of PD, illustrating that activation of autophagy is way forward. But we should consider that excessive autophagy can lead to some unique cell death by producing extra degradation products. The effect of complex interactions between autophagy and other intracellular processes call into question that how to induce autophagy precisely without affecting cell homeostasis.

Besides, most of these autophagy pharmacological regulations trigger the whole induction of autophagy, lacking organ specificity and substrate selectivity, which may cause side effects. Considering the cytoprotective function of autophagy on neurons, improving the brain specificity of autophagy is wise. It also requires that small-molecule drugs can penetrate blood–brain barrier better and enrich towards the CNS. The development of LRRK2 inhibitors focuses on this key issue just right. It provides a valuable experience for the discovery of brain osmotic autophagic molecules from a perspective of medicinal chemistry. As for the substrate selectivity, activation of autophagy not only degrades the abnormal aggregation of α-syn, but increases the degradation of other proteins and organelles, thus increasing the pressure of cell survival. α-Syn is the degradation substrate of CMA (a selective autophagy), and targeting CMA has become a new thought of PD drug development33. Interestingly, paradoxical with previous studies, impaired macroautophagy was found not to increase the accumulation of α-syn oligomers, and induced macroautophagy may only benefit in cellular pathology and long-term neuronal survival, and aggravate motor performance through impacting striatal DA dynamics173, which reminds us the questions of when to apply autophagy inducers to which kinds of patients based on their mechanisms and phenotypes.

In addition, some new emerging technologies about autophagy-modulating small molecules have appealed much attention, such as autophagy targeting chimera (AUTAC) and autophagosome-tethering compound (ATTEC)174, 175, 176, 177. Both of these technologies are capable of selectively autophagic degradation of substrates. Although there is no sound evidence that these two technologies can be directly applied to the degradation of α-syn, it may provide a new clue and future direction for exploiting more autophagic modulators in PD therapy.

More recently, autophagy has been considered as one of the predominant aggravators at the late stage of PD progression178, which emphasizing the necessity of combination therapies of inducing autophagy and other interventions targeting gene mutations or neuroinflammation at different phase of PD. Currently, most PD patients take a variety of drugs simultaneously, such as levodopa and MAO-B inhibitors, to limit high medication dose and some adverse events. Therefore, autophagy-modulating drugs combination with themselves, other non-autophagy related anti-PD drugs or gene therapy can be an effective approach to overcome the limitations of autophagy-modulating small molecules and thereby promoting their clinical development. Despite confronting with many difficulties, discovery of more new autophagy-modulating compounds will confirm the practicability of safe candidate drugs and their importance in preventing or limiting inevitable neurodegeneration, which is essential for the successful fight against PD in the near future.

Acknowledgments

This work was financially supported by National Science and Technology Major Project of the Ministry of Science and Technology of the People's Republic of China (No. 2018ZX09735005),National Natural Science Foundation of China (Grant Nos. 81803755, 81673455 and 81922064), Sichuan University Postdoctoral Research and Development Foundation (Grant No. 2020SCU12062, China), and Sichuan Science and Technology Program (Grant No. 2019JDRC0091, China).

Author contributions

Bo Liu, Liang Ouyang and Guan Wang conceived the project and supervised the project. Kai Zhang, Shiou Zhu and Jiamei Li summed up the literature and drafted the manuscript. Tingting Jiang was involved in drawing the figures. Lu Feng collected and organized the inhibitors. Kai Zhang, Guan Wang and Junping Pei proofread the structures and figures. Bo Liu, Liang Ouyang and Shiou Zhu revised the manuscript. All authors approved the final manuscript.

Conflicts of interest

The authors have no conflicts of interest to declare.

Footnotes

Peer review under the responsibility of Chinese Pharmaceutical Association and Institute of Materia Medica, Chinese Academy of Medical Sciences.

Contributor Information

Guan Wang, Email: guan8079@163.com.

Liang Ouyang, Email: ouyangliang@scu.edu.cn.

Bo Liu, Email: liubo2400@163.com.

References

  • 1.Kalia L.V., Lang A.E. Parkinson's disease. Lancet. 2015;386:896–912. doi: 10.1016/S0140-6736(14)61393-3. [DOI] [PubMed] [Google Scholar]
  • 2.Beaulieu J.M., Gainetdinov R.R. The physiology, signaling, and pharmacology of dopamine receptors. Pharmacol Rev. 2011;63:182–217. doi: 10.1124/pr.110.002642. [DOI] [PubMed] [Google Scholar]
  • 3.Doty R.L. Olfactory dysfunction in Parkinson disease. Nat Rev Neurol. 2012;8:329–339. doi: 10.1038/nrneurol.2012.80. [DOI] [PubMed] [Google Scholar]
  • 4.Santangelo R.M., Acker T.M., Zimmerman S.S., Katzman B.M., Strong K.L., Traynelis S.F. Novel NMDA receptor modulators: an update. Expert Opin Ther Pat. 2012;22:1337–1352. doi: 10.1517/13543776.2012.728587. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Shook B.C., Jackson P.F. Adenosine A2A receptor antagonists and Parkinson's disease. ACS Chem Neurosci. 2011;2:555–567. doi: 10.1021/cn2000537. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Valera E., Masliah E. Therapeutic approaches in Parkinson's disease and related disorders. J Neurochem. 2016;139(Suppl):346–352. doi: 10.1111/jnc.13529. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Glick D., Barth S., Macleod K.F. Autophagy: cellular and molecular mechanisms. J Pathol. 2010;221:3–12. doi: 10.1002/path.2697. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Maria Fimia G., Stoykova A., Romagnoli A., Giunta L., Di Bartolomeo S., Nardacci R. Ambra1 regulates autophagy and development of the nervous system. Nature. 2007;447:1121–1125. doi: 10.1038/nature05925. [DOI] [PubMed] [Google Scholar]
  • 9.Komatsu M., Waguri S., Chiba T., Murata S., Iwata J.I., Tanida I. Loss of autophagy in the central nervous system causes neurodegeneration in mice. Nature. 2006;441:880–884. doi: 10.1038/nature04723. [DOI] [PubMed] [Google Scholar]
  • 10.Komatsu M., Qing J.W., Holstein G.R., Friedrich V.L., Iwata J.I., Kominami E. Essential role for autophagy protein Atg7 in the maintenance of axonal homeostasis and the prevention of axonal degeneration. Proc Natl Acad Sci U S A. 2007;104:14489–14494. doi: 10.1073/pnas.0701311104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Wang Q.J., Ding Y., Kohtz S., Mizushima N., Cristea I.M., Rout M.P. Induction of autophagy in axonal dystrophy and degeneration. J Neurosci. 2006;26:8057–8068. doi: 10.1523/JNEUROSCI.2261-06.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Levine B., Klionsky D.J. Autophagy wins the 2016 Nobel Prizein Physiology or Medicine: breakthroughs in baker's yeast fuel advances in biomedical research. Proc Natl Acad Sci U S A. 2017;114:201–205. doi: 10.1073/pnas.1619876114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Son J.H., Shim J.H., Kim K.H., Ha J.Y., Han J.Y. Neuronal autophagy and neurodegenerative diseases. Exp Mol Med. 2012;44:89–98. doi: 10.3858/emm.2012.44.2.031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Hu Z.Y., Chen B., Zhang J.P., Ma Y.Y. Up-regulation of autophagy-related gene 5 (ATG5) protects dopaminergic neurons in a zebrafish model of Parkinson's disease. J Biol Chem. 2017;292:18062–18074. doi: 10.1074/jbc.M116.764795. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Nuytemans K., Theuns J., Cruts M., Van Broeckhoven C. Genetic etiology of Parkinson disease associated with mutations in the SNCA, PARK2, PINK1, PARK7, and LRRK2 genes: a mutation update. Hum Mutat. 2010;31:763–780. doi: 10.1002/humu.21277. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.O’Hara D.M., Pawar G., Kalia S.K., Kalia L.V. LRRK2 and α-synuclein: distinct or synergistic players in Parkinson's disease?. Front Neurosci. 2020;14:577. doi: 10.3389/fnins.2020.00577. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Albanese F., Novello S., Morari M. Autophagy and LRRK2 in the aging brain. Front Neurosci. 2019;13:1–23. doi: 10.3389/fnins.2019.01352. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Erb M.L., Moore D.J. LRRK2 and the endolysosomal system in Parkinson's disease. J Parkinsons Dis. 2020;10:1271–1291. doi: 10.3233/JPD-202138. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Cogo S., Manzoni C., Lewis P.A., Greggio E. Leucine-rich repeat kinase 2 and lysosomal dyshomeostasis in Parkinson disease. J Neurochem. 2020;152:273–283. doi: 10.1111/jnc.14908. [DOI] [PubMed] [Google Scholar]
  • 20.Lamonaca G., Volta M. Alpha-synuclein and LRRK2 in synaptic autophagy: linking early dysfunction to late-stage pathology in Parkinson's disease. Cells. 2020;9:1115. doi: 10.3390/cells9051115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Longo F., Mercatelli D., Novello S., Arcuri L., Brugnoli A., Vincenzi F. Age-dependent dopamine transporter dysfunction and serine129 phospho-α-synuclein overload in G2019S LRRK2 mice. Acta Neuropathol Commun. 2017;5:22. doi: 10.1186/s40478-017-0426-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Ho D.H., Kim H., Nam D., Sim H., Kim J., Kim H.G. LRRK2 impairs autophagy by mediating phosphorylation of leucyl-tRNA synthetase. Cell Biochem Funct. 2018;36:431–442. doi: 10.1002/cbf.3364. [DOI] [PubMed] [Google Scholar]
  • 23.Giaime E., Tong Y., Wagner L.K., Yuan Y., Huang G., Shen J. Age-dependent dopaminergic neurodegeneration and impairment of the autophagy-lysosomal pathway in LRRK-deficient mice. Neuron. 2017;96:796–807. doi: 10.1016/j.neuron.2017.09.036. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Wauters F., Cornelissen T., Imberechts D., Martin S., Koentjoro B., Sue C. LRRK2 mutations impair depolarization-induced mitophagy through inhibition of mitochondrial accumulation of RAB10. Autophagy. 2020;16:203–222. doi: 10.1080/15548627.2019.1603548. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Ho P.W.L., Leung C.T., Liu H., Pang S.Y.Y., Lam C.S.C., Xian J. Age-dependent accumulation of oligomeric SNCA/α-synuclein from impaired degradation in mutant LRRK2 knockin mouse model of Parkinson disease: role for therapeutic activation of chaperone-mediated autophagy (CMA) Autophagy. 2020;16:347–370. doi: 10.1080/15548627.2019.1603545. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Bonello F., Hassoun S.M., Mouton-Liger F., Shin Y.S., Muscat A., Tesson C. LRRK2 impairs PINK1/Parkin-dependent mitophagy via its kinase activity: pathologic insights into Parkinson's disease. Hum Mol Genet. 2019;28:1645–1660. doi: 10.1093/hmg/ddz004. [DOI] [PubMed] [Google Scholar]
  • 27.Alejandra Morato Torres C., Wassouf Z., Zafar F., Sastre D., Outeiro T.F., Schüle B. The role of alpha-synuclein and other Parkinson's genes in neurodevelopmental and neurodegenerative disorders. Int J Mol Sci. 2020;21:1–32. doi: 10.3390/ijms21165724. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Neumann M., Müller V., Kretzschmar H.A., Haass C., Kahle P.J. Regional distribution of proteinase K-resistant α-synuclein correlates with Lewy body disease stage. J Neuropathol Exp Neurol. 2004;63:1225–1235. doi: 10.1093/jnen/63.12.1225. [DOI] [PubMed] [Google Scholar]
  • 29.Peelaerts W., Bousset L., van Der Perren A., Moskalyuk A., Pulizzi R., Giugliano M. α-Synuclein strains cause distinct synucleinopathies after local and systemic administration. Nature. 2015;522:340–344. doi: 10.1038/nature14547. [DOI] [PubMed] [Google Scholar]
  • 30.Spillantini M.G., Schmidt M.L., Lee V.M.Y., Trojanowski J.Q., Jakes R., Goedert M. α-Synuclein in Lewy bodies. Nature. 1997;388:839–840. doi: 10.1038/42166. [DOI] [PubMed] [Google Scholar]
  • 31.Gao J., Perera G., Bhadbhade M., Halliday G.M., Dzamko N. Autophagy activation promotes clearance of α-synuclein inclusions in fibril-seeded human neural cells. J Biol Chem. 2019;294:14241–14256. doi: 10.1074/jbc.RA119.008733. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Pantazopoulou M., Brembati V., Kanellidi A., Bousset L., Melki R., Stefanis L. Distinct alpha-synuclein species induced by seeding are selectively cleared by the lysosome or the proteasome in neuronally differentiated SH-SY5Y cells. J Neurochem. 2021;156:880–896. doi: 10.1111/jnc.15174. [DOI] [PubMed] [Google Scholar]
  • 33.Wong Y.C., Krainc D. α-Synuclein toxicity in neurodegeneration: mechanism and therapeutic strategies. Nat Med. 2017;23:1–13. doi: 10.1038/nm.4269. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Nascimento A.C., Erustes A.G., Reckziegel P., Bincoletto C., Ureshino R.P., Pereira G.J.S. α-Synuclein overexpression induces lysosomal dysfunction and autophagy impairment in human neuroblastoma SH-SY5Y. Neurochem Res. 2020;45:2749–2761. doi: 10.1007/s11064-020-03126-8. [DOI] [PubMed] [Google Scholar]
  • 35.Mazzulli J.R., Zunke F., Isacson O., Studer L., Krainc D. α-Synuclein-induced lysosomal dysfunction occurs through disruptions in protein trafficking in human midbrain synucleinopathy models. Proc Natl Acad Sci U S A. 2016;113:1931–1936. doi: 10.1073/pnas.1520335113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Sato S., Uchihara T., Fukuda T., Noda S., Kondo H., Saiki S. Loss of autophagy in dopaminergic neurons causes Lewy pathology and motor dysfunction in aged mice. Sci Rep. 2018;8:1–10. doi: 10.1038/s41598-018-21325-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Gegg M.E., Verona G., Schapira A.H.V. Glucocerebrosidase deficiency promotes release of α-synuclein fibrils from cultured neurons. Hum Mol Genet. 2020;29:1716–1728. doi: 10.1093/hmg/ddaa085. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Li H., Ham A., Ma T.C., Kuo S.H., Kanter E., Kim D. Mitochondrial dysfunction and mitophagy defect triggered by heterozygous GBA mutations. Autophagy. 2019;15:113–130. doi: 10.1080/15548627.2018.1509818. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Barazzuol L., Giamogante F., Brini M., Calì T. PINK1/Parkin mediated mitophagy, Ca2+ signalling, and ER-mitochondria contacts in Parkinson's disease. Int J Mol Sci. 2020;21:1172. doi: 10.3390/ijms21051772. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Truban D., Hou X., Caulfield T.R., Fiesel F.C., Springer W. PINK1, Parkin, and mitochondrial quality control: what can we learn about Parkinson's disease pathobiology?. J Parkinsons Dis. 2017;7:13–29. doi: 10.3233/JPD-160989. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Wang R., Tan J., Chen T., Han H., Tian R., Tan Y. ATP13A2 facilitates HDAC6 recruitment to lysosome to promote autophagosome–lysosome fusion. J Cell Biol. 2019;218:267–284. doi: 10.1083/jcb.201804165. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Bento C.F., Ashkenazi A., Jimenez-Sanchez M., Rubinsztein D.C. The Parkinson's disease-associated genes ATP13A2 and SYT11 regulate autophagy via a common pathway. Nat Commun. 2016;7:11803. doi: 10.1038/ncomms11803. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Patil S.P., Jain P.D., Ghumatkar P.J., Tambe R., Sathaye S. Neuroprotective effect of metformin in MPTP-induced Parkinson's disease in mice. Neuroscience. 2014;277:747–754. doi: 10.1016/j.neuroscience.2014.07.046. [DOI] [PubMed] [Google Scholar]
  • 44.Wu Y., Li X., Zhu J.X., Xie W., Le W., Fan Z. Resveratrol-activated AMPK/SIRT1/autophagy in cellular models of Parkinson's disease. Neurosignals. 2011;19:163–174. doi: 10.1159/000328516. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Ouyang L., Zhang L., Zhang S., Yao D., Zhao Y., Wang G. Small-molecule activator of UNC-51-like kinase 1 (ULK1) that induces cytoprotective autophagy for Parkinson's disease treatment. J Med Chem. 2018;61:2776–2792. doi: 10.1021/acs.jmedchem.7b01575. [DOI] [PubMed] [Google Scholar]
  • 46.Williams R.S.B., Cheng L., Mudge A.W., Harwood A.J. A common mechanism of action for three mood-stabilizing drugs. Nature. 2002;417:292–295. doi: 10.1038/417292a. [DOI] [PubMed] [Google Scholar]
  • 47.Christensen K.V., Smith G.P., Williamson D.S. Development of LRRK2 inhibitors for the treatment of Parkinson's disease. Prog Med Chem. 2017;56:37–80. doi: 10.1016/bs.pmch.2016.11.002. [DOI] [PubMed] [Google Scholar]
  • 48.Deng X., Dzamko N., Prescott A., Davies P., Liu Q., Yang Q. Characterization of a selective inhibitor of the Parkinson's disease kinase LRRK2. Nat Chem Biol. 2011;7:203–205. doi: 10.1038/nchembio.538. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.West A.B. Achieving neuroprotection with LRRK2 kinase inhibitors in Parkinson disease. Exp Neurol. 2017;298:236–245. doi: 10.1016/j.expneurol.2017.07.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Wirawan E., Lippens S., Vanden Berghe T., Romagnoli A., Fimia G.M., Piacentini M. Beclin 1: a role in membrane dynamics and beyond. Autophagy. 2012;8:6–17. doi: 10.4161/auto.8.1.16645. [DOI] [PubMed] [Google Scholar]
  • 51.Hill S.M., Wrobel L., Rubinsztein D.C. Post-translational modifications of beclin-1 provide multiple strategies for autophagy regulation. Cell Death Differ. 2019;26:617–629. doi: 10.1038/s41418-018-0254-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Savolainen M.H., Richie C.T., Harvey B.K., Männistö P.T., Maguire-Zeiss K.A., Myöhänen T.T. The beneficial effect of a prolyl oligopeptidase inhibitor, KYP-2047, on alpha-synuclein clearance and autophagy in A30P transgenic mouse. Neurobiol Dis. 2014;68:1–15. doi: 10.1016/j.nbd.2014.04.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Lu J.H., Tan J.Q., Durairajan S.S.K., Liu L.F., Zhang Z.H., Ma L. Isorhynchophylline, a natural alkaloid, promotes the degradation of α-synuclein in neuronal cells via inducing autophagy. Autophagy. 2012;8:98–108. doi: 10.4161/auto.8.1.18313. [DOI] [PubMed] [Google Scholar]
  • 54.Yan D., Ma Z., Liu C., Wang C., Deng Y., Liu W. Corynoxine B ameliorates HMGB1-dependent autophagy dysfunction during manganese exposure in SH-SY5Y human neuroblastoma cells. Food Chem Toxicol. 2019;124:336–348. doi: 10.1016/j.fct.2018.12.027. [DOI] [PubMed] [Google Scholar]
  • 55.Kang S.Y., Lee S Bin, Kim H.J., Kim H.T., Yang H.O., Jang W. Autophagic modulation by rosuvastatin prevents rotenone-induced neurotoxicity in an in vitro model of Parkinson's disease. Neurosci Lett. 2017;642:20–26. doi: 10.1016/j.neulet.2017.01.063. [DOI] [PubMed] [Google Scholar]
  • 56.Li Q., Niu C., Zhang X., Dong M. Gastrodin and isorhynchophylline synergistically inhibit MPP+-induced oxidative stress in SH-SY5Y cells by targeting ERK1/2 and GSK-3β pathways: involvement of Nrf2 nuclear translocation. ACS Chem Neurosci. 2018;9:482–493. doi: 10.1021/acschemneuro.7b00247. [DOI] [PubMed] [Google Scholar]
  • 57.Anand P., Kunnumakkara A.B., Newman R.A., Aggarwal B.B. Bioavailability of curcumin: problems and promises. Mol Pharm. 2007;4:807–818. doi: 10.1021/mp700113r. [DOI] [PubMed] [Google Scholar]
  • 58.Cole G.M., Teter B., Frautschy S.A. Neuroprotective effects of curcumin. Adv Exp Med Biol. 2007;595:197–212. doi: 10.1007/978-0-387-46401-5_8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Singh P.K., Kotia V., Ghosh D., Mohite G.M., Kumar A., Maji S.K. Curcumin modulates α-synuclein aggregation and toxicity. ACS Chem Neurosci. 2013;4:393–407. doi: 10.1021/cn3001203. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Jayaraj R.L., Elangovan N., Manigandan K., Singh S., Shukla S. CNB-001 a novel curcumin derivative, guards dopamine neurons in MPTP model of Parkinson's disease. Biomed Res Int. 2014;2014:236182. doi: 10.1155/2014/236182. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Song J.X., Sun Y.R., Peluso I., Zeng Y., Yu X., Lu J.H. A novel curcumin analog binds to and activates TFEB in vitro and in vivo independent of MTOR inhibition. Autophagy. 2016;12:1372–1389. doi: 10.1080/15548627.2016.1179404. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Wang Z., Yang C., Liu J., Tong B.C.K., Zhu Z., Malampati S. A curcumin derivative activates TFEB and protects against parkinsonian neurotoxicity in vitro. Int J Mol Sci. 2020;21:1515. doi: 10.3390/ijms21041515. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Khalifeh M., Barreto G.E., Sahebkar A. Trehalose as a promising therapeutic candidate for the treatment of Parkinson's disease. Br J Pharmacol. 2019;176:1173–1189. doi: 10.1111/bph.14623. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.McNeill A., Magalhaes J., Shen C., Chau K.Y., Hughes D., Mehta A. Ambroxol improves lysosomal biochemistry in glucocerebrosidase mutation-linked Parkinson disease cells. Brain. 2014;137:1481–1495. doi: 10.1093/brain/awu020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Ambrosi G., Ghezzi C., Zangaglia R., Levandis G., Pacchetti C., Blandini F. Ambroxol-induced rescue of defective glucocerebrosidase is associated with increased LIMP-2 and saposin C levels in GBA1 mutant Parkinson's disease cells. Neurobiol Dis. 2015;82:235–242. doi: 10.1016/j.nbd.2015.06.008. [DOI] [PubMed] [Google Scholar]
  • 66.Zhong J., Xie J., Xiao J., Li D., Xu B., Wang X. Inhibition of PDE4 by FCPR16 induces AMPK-dependent autophagy and confers neuroprotection in SH-SY5Y cells and neurons exposed to MPP+-induced oxidative insult. Free Radic Biol Med. 2019;135:87–101. doi: 10.1016/j.freeradbiomed.2019.02.027. [DOI] [PubMed] [Google Scholar]
  • 67.Sanchez-Martinez A., Beavan M., Gegg M.E., Chau K.Y., Whitworth A.J., Schapira A.H.V. Parkinson disease-linked GBA mutation effects reversed by molecular chaperones in human cell and fly models. Sci Rep. 2016;6:31380. doi: 10.1038/srep31380. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Aflaki E., Borger D.K., Moaven N., Stubblefield B.K., Rogers S.A., Patnaik S. A new glucocerebrosidase chaperone reduces α-synuclein and glycolipid levels in iPSC-derived dopaminergic neurons from patients with Gaucher disease and parkinsonism. J Neurosci. 2016;36:7441–7452. doi: 10.1523/JNEUROSCI.0636-16.2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Busch B.B., Stevens W.C., Martin R., Ordentlich P., Zhou S., Sapp D.W. Identification of a selective inverse agonist for the orphan nuclear receptor estrogen-related receptor α. J Med Chem. 2004;47:5593–5596. doi: 10.1021/jm049334f. [DOI] [PubMed] [Google Scholar]
  • 70.Suresh S.N., Chavalmane A.K., Pillai M., Ammanathan V., Vidyadhara D.J., Yarreiphang H. Modulation of autophagy by a small molecule inverse agonist of ERRα is neuroprotective. Front Mol Neurosci. 2018;11:109. doi: 10.3389/fnmol.2018.00109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.SN S, Pandurangi J., Murumalla R., DJ V., Garimella L., Acharya A. Small molecule modulator of aggrephagy regulates neuroinflammation to curb pathogenesis of neurodegeneration. EBioMedicine. 2019;50:260–273. doi: 10.1016/j.ebiom.2019.10.036. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Ren Y., Chen J., Wu X., Gui C., Mao K., Zou F. Role of c-Abl-GSK3β signaling in MPP1-induced autophagy-lysosomal dysfunction. Toxicol Sci. 2018;165:232–243. doi: 10.1093/toxsci/kfy155. [DOI] [PubMed] [Google Scholar]
  • 73.Pagan F., Hebron M., Valadez E.H., Torres-Yaghi Y., Huang X., Mills R.R. Nilotinib effects in Parkinson's disease and dementia with lewy bodies. J Parkinsons Dis. 2016;6:503–517. doi: 10.3233/JPD-160867. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Höllerhage M., Fussi N., Rösler T.W., Wurst W., Behrends C., Höglinger G.U. Multiple molecular pathways stimulating macroautophagy protect from alpha-synuclein-induced toxicity in human neurons. Neuropharmacology. 2019;149:13–26. doi: 10.1016/j.neuropharm.2019.01.023. [DOI] [PubMed] [Google Scholar]
  • 75.Cool B., Zinker B., Chiou W., Kifle L., Cao N., Perham M. Identification and characterization of a small molecule AMPK activator that treats key components of type 2 diabetes and the metabolic syndrome. Cell Metab. 2006;3:403–416. doi: 10.1016/j.cmet.2006.05.005. [DOI] [PubMed] [Google Scholar]
  • 76.Luan Y., Ren X., Zheng W., Zeng Z., Guo Y., Hou Z. Chronic caffeine treatment protects against α-synucleinopathy by reestablishing autophagy activity in the mouse striatum. Front Neurosci. 2018;12:301. doi: 10.3389/fnins.2018.00301. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Malagelada C., Jin Z.H., Jackson-Lewis V., Przedborski S., Greene L.A. Rapamycin protects against neuron death in in vitro and in vivo models of Parkinson's disease. J Neurosci. 2010;30:1166–1175. doi: 10.1523/JNEUROSCI.3944-09.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Chen L.L., Song J.X., Lu J.H., Yuan Z.W., Liu L.F., Durairajan S.S.K. Corynoxine, a natural autophagy enhancer, promotes the clearance of alpha-synuclein via Akt/mTOR pathway. J Neuroimmune Pharmacol. 2014;9:380–387. doi: 10.1007/s11481-014-9528-2. [DOI] [PubMed] [Google Scholar]
  • 79.Xu Y Da, Cui C., Sun M.F., Zhu Y.L., Chu M., Shi Y.W. Neuroprotective effects of loganin on MPTP-induced Parkinson's disease mice: neurochemistry, glial reaction and autophagy studies. J Cell Biochem. 2017;118:3495–3510. doi: 10.1002/jcb.26010. [DOI] [PubMed] [Google Scholar]
  • 80.Bobela W., Nazeeruddin S., Knott G., Aebischer P., Schneider B.L. Modulating the catalytic activity of AMPK has neuroprotective effects against α-synuclein toxicity. Mol Neurodegener. 2017;12:1–19. doi: 10.1186/s13024-017-0220-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Curry D.W., Stutz B., Andrews Z.B., Elsworth J.D. Targeting AMPK signaling as a neuroprotective strategy in Parkinson's disease. J Parkinsons Dis. 2018;8:161–181. doi: 10.3233/JPD-171296. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Carling D. AMPK signalling in health and disease. Curr Opin Cell Biol. 2017;45:31–37. doi: 10.1016/j.ceb.2017.01.005. [DOI] [PubMed] [Google Scholar]
  • 83.Karuppagounder S.S., Brahmachari S., Lee Y., Dawson V.L., Dawson T.M., Ko H.S. The c-Abl inhibitor, nilotinib, protects dopaminergic neurons in a preclinical animal model of Parkinson's disease. Sci Rep. 2014;4:4874. doi: 10.1038/srep04874. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Ashabi G., Khalaj L., Khodagholi F., Goudarzvand M., Sarkaki A Pre-treatment with metformin activates Nrf2 antioxidant pathways and inhibits inflammatory responses through induction of AMPK after transient global cerebral ischemia. Metab Brain Dis. 2015;30:747–754. doi: 10.1007/s11011-014-9632-2. [DOI] [PubMed] [Google Scholar]
  • 85.Culmsee C., Monroe J., Kemp B.E., Mattson M.P. AMP-activated protein kinase is highly expressed in neurons in the developing rat brain and promotes neuronal survival following glucose deprivation. J Mol Neurosci. 2001;17:45–58. doi: 10.1385/JMN:17:1:45. [DOI] [PubMed] [Google Scholar]
  • 86.Li J., Zeng Z., Viollet B., Ronnett G.V., McCullough L.D. Neuroprotective effects of adenosine monophosphate-activated protein kinase inhibition and gene deletion in stroke. Stroke. 2007;38:2992–2999. doi: 10.1161/STROKEAHA.107.490904. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Paudel Y.N., Angelopoulou E., Piperi C., Shaikh M.F., Othman I. Emerging neuroprotective effect of metformin in Parkinson's disease: a molecular crosstalk. Pharmacol Res. 2020;152:104593. doi: 10.1016/j.phrs.2019.104593. [DOI] [PubMed] [Google Scholar]
  • 88.Katila N., Bhurtel S., Park P.H., Hong J.T., Choi D.Y. Activation of AMPK/aPKCζ/CREB pathway by metformin is associated with upregulation of GDNF and dopamine. Biochem Pharmacol. 2020;180:114193. doi: 10.1016/j.bcp.2020.114193. [DOI] [PubMed] [Google Scholar]
  • 89.Lu M., Su C., Qiao C., Bian Y., Ding J., Hu G. Metformin prevents dopaminergic neuron death in MPTP/P-induced mouse model of Parkinson's disease via autophagy and mitochondrial ROS clearance. Int J Neuropsychopharmacol. 2016;19:1–11. doi: 10.1093/ijnp/pyw047. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Caruana M., Cauchi R., Vassallo N. Putative role of red wine polyphenols against brain pathology in Alzheimer's and Parkinson's disease. Front Nutr. 2016;3:31. doi: 10.3389/fnut.2016.00031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Park D., Jeong H., Lee M.N., Koh A., Kwon O., Yang Y.R. Resveratrol induces autophagy by directly inhibiting mTOR through ATP competition. Sci Rep. 2016;6:21772. doi: 10.1038/srep21772. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.ur Rasheed M.S., Tripathi M.K., Mishra A.K., Shukla S., Singh M.P. Resveratrol protects from toxin-induced Parkinsonism: plethora of proofs hitherto petty translational value. Mol Neurobiol. 2016;53:2751–2760. doi: 10.1007/s12035-015-9124-3. [DOI] [PubMed] [Google Scholar]
  • 93.Truong V.L., Jun M., Jeong W.S. Role of resveratrol in regulation of cellular defense systems against oxidative stress. Biofactors. 2018;44:36–49. doi: 10.1002/biof.1399. [DOI] [PubMed] [Google Scholar]
  • 94.Guo Y.J., Dong S.Y., Cui X.X., Feng Y., Liu T., Yin M. Resveratrol alleviates MPTP-induced motor impairments and pathological changes by autophagic degradation of α-synuclein via SIRT1-deacetylated LC3. Mol Nutr Food Res. 2016;60:2161–2175. doi: 10.1002/mnfr.201600111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Lin K.L., Lin K.J., Wang P.W., Chuang J.H., Lin H.Y., Chen S.D. Resveratrol provides neuroprotective effects through modulation of mitochondrial dynamics and ERK1/2 regulated autophagy. Free Radic Res. 2018;52:1371–1386. doi: 10.1080/10715762.2018.1489128. [DOI] [PubMed] [Google Scholar]
  • 96.Pourhanifeh M.H., Shafabakhsh R., Reiter R.J., Asemi Z. The effect of resveratrol on neurodegenerative disorders: possible protective actions against autophagy, apoptosis, inflammation and oxidative stress. Curr Pharm Des. 2019;25:2178–2191. doi: 10.2174/1381612825666190717110932. [DOI] [PubMed] [Google Scholar]
  • 97.Mirguet O., Sautet S., Clément C.A., Toum J., Donche F., Marques C. Discovery of pyridones as oral AMPK direct activators. ACS Med Chem Lett. 2013;4:632–636. doi: 10.1021/ml400157g. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Zhou Z.Z., Ge B.C., Zhong Q.P., Huang C., Cheng Y.F., Yang X.M. Development of highly potent phosphodiesterase 4 inhibitors with anti-neuroinflammation potential: design, synthesis, and structure-activity relationship study of catecholamides bearing aromatic rings. Eur J Med Chem. 2016;124:372–379. doi: 10.1016/j.ejmech.2016.08.052. [DOI] [PubMed] [Google Scholar]
  • 99.Sujobert P., Poulain L., Paubelle E., Zylbersztejn F., Grenier A., Lambert M. Co-activation of AMPK and mTORC1 induces cytotoxicity in acute myeloid leukemia. Cell Rep. 2015;11:1446–1457. doi: 10.1016/j.celrep.2015.04.063. [DOI] [PubMed] [Google Scholar]
  • 100.Bové J., Martínez-Vicente M., Vila M. Fighting neurodegeneration with rapamycin: mechanistic insights. Nat Rev Neurosci. 2011;12:437–452. doi: 10.1038/nrn3068. [DOI] [PubMed] [Google Scholar]
  • 101.Maiese K., Chong Z.Z., Shang Y.C., Wang S. MTOR: on target for novel therapeutic strategies in the nervous system. Trends Mol Med. 2013;19:51–60. doi: 10.1016/j.molmed.2012.11.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Yeh H.S., Yong J. mTOR-coordinated post-transcriptional gene regulations: from fundamental to pathogenic insights. J Lipid Atheroscler. 2020;9:8. doi: 10.12997/jla.2020.9.1.8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Darabi S., Noori-Zadeh A., Rajaei F., Abbaszadeh H.A., Abdollahifar M.A., Bakhtiyari S. Neuroprotective effects of the small-molecule enhancer of rapamycin in the cellular model of Parkinson's disease. Neurophysiology. 2019;51:88–96. [Google Scholar]
  • 104.Liu G.Y., Sabatini D.M. mTOR at the nexus of nutrition, growth, ageing and disease. Nat Rev Mol Cell Biol. 2020;21:183–203. doi: 10.1038/s41580-019-0199-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Dehay B., Bové J., Rodríguez-Muela N., Perier C., Recasens A., Boya P. Pathogenic lysosomal depletion in Parkinson's disease. J Neurosci. 2010;30:12535–12544. doi: 10.1523/JNEUROSCI.1920-10.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Ferree A., Guillily M., Li H., Smith K., Takashima A., Squillace R. Regulation of physiologic actions of LRRK2: focus on autophagy. Neurodegener Dis. 2012;10:238–241. doi: 10.1159/000332599. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Siracusa R., Paterniti I., Cordaro M., Crupi R., Bruschetta G., Campolo M. Neuroprotective effects of temsirolimus in animal models of Parkinson's disease. Mol Neurobiol. 2018;55:2403–2419. doi: 10.1007/s12035-017-0496-4. [DOI] [PubMed] [Google Scholar]
  • 108.Rivero-Ríos P., Madero-Pérez J., Fernández B., Hilfiker S. Targeting the autophagy/lysosomal degradation pathway in Parkinsons disease. Curr Neuropharmacol. 2016;14:238–249. doi: 10.2174/1570159X13666151030103027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Abdel-Maksoud M.S., El-Gamal M.I., Benhalilou D.R., Ashraf S., Mohammed S.A., Oh C. Mechanistic/mammalian target of rapamycin: recent pathological aspects and inhibitors. Med Res Rev. 2019;39:631–664. doi: 10.1002/med.21535. [DOI] [PubMed] [Google Scholar]
  • 110.Knight Z.A., Gonzalez B., Feldman M.E., Zunder E.R., Goldenberg D.D., Williams O. A pharmacological map of the PI3-K family defines a role for p110α in insulin signaling. Cell. 2006;125:733–747. doi: 10.1016/j.cell.2006.03.035. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Fan Q.W., Knight Z.A., Goldenberg D.D., Yu W., Mostov K.E., Stokoe D. A dual PI3 kinase/mTOR inhibitor reveals emergent efficacy in glioma. Cancer Cell. 2006;9:341–349. doi: 10.1016/j.ccr.2006.03.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Wong P.M., Puente C., Ganley I.G., Jiang X. The ULK1 complex sensing nutrient signals for autophagy activation. Autophagy. 2013;9:124–137. doi: 10.4161/auto.23323. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Zhu Z., Yang C., Iyaswamy A., Krishnamoorthi S., Sreenivasmurthy S.G., Liu J. Balancing mTOR signaling and autophagy in the treatment of Parkinson's disease. Int J Mol Sci. 2019;20:728. doi: 10.3390/ijms20030728. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Park J.M., Seo M., Jung C.H., Grunwald D., Stone M., Otto N.M. ULK1 phosphorylates Ser30 of BECN1 in association with ATG14 to stimulate autophagy induction. Autophagy. 2018;14:584–597. doi: 10.1080/15548627.2017.1422851. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Sarkar S., Floto R.A., Berger Z., Imarisio S., Cordenier A., Pasco M. Lithium induces autophagy by inhibiting inositol monophosphatase. J Cell Biol. 2005;170:1101–1111. doi: 10.1083/jcb.200504035. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Elphick M., Taghavi Z., Powell T., Godfrey P.P. Alteration of inositol phospholipid metabolism in rat cortex by lithium but not carbamazepine. Eur J Pharmacol. 1988;156:411–414. doi: 10.1016/0014-2999(88)90287-7. [DOI] [PubMed] [Google Scholar]
  • 117.Skow Á., Douglas I., Smeeth L. The association between Parkinson's disease and anti-epilepsy drug carbamazepine: a case-control study using the UK General Practice Research Database. Br J Clin Pharmacol. 2013;76:816–822. doi: 10.1111/bcp.12100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Lee H.S., Lobbestael E., Vermeire S., Sabino J., Cleynen I. Inflammatory bowel disease and Parkinson's disease: common pathophysiological links. Gut. 2021;70:408–417. doi: 10.1136/gutjnl-2020-322429. [DOI] [PubMed] [Google Scholar]
  • 119.Rudenko I.N., Chia R., Cookson M.R. Is inhibition of kinase activity the only therapeutic strategy for LRRK2-associated Parkinson's disease?. BMC Med. 2012;10:20. doi: 10.1186/1741-7015-10-20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Manzoni C., Lewis P.A. Lrrk2 and autophagy. Adv Neurobiol. 2017;14:89–105. doi: 10.1007/978-3-319-49969-7_5. [DOI] [PubMed] [Google Scholar]
  • 121.Manzoni C., Mamais A., Roosen D.A., Dihanich S., Soutar M.P.M., Plun-Favreau H. MTOR independent regulation of macroautophagy by leucine rich repeat kinase 2 via beclin-1. Sci Rep. 2016;6:35106. doi: 10.1038/srep35106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Soukup S.F., Kuenen S., Vanhauwaert R., Manetsberger J., Hernández-Díaz S., Swerts J. A LRRK2-dependent endophilinA phosphoswitch is critical for macroautophagy at presynaptic terminals. Neuron. 2016;92:829–844. doi: 10.1016/j.neuron.2016.09.037. [DOI] [PubMed] [Google Scholar]
  • 123.Alegre-Abarrategui J., Christian H., Lufino M.M.P., Mutihac R., Venda L.L., Ansorge O. LRRK2 regulates autophagic activity and localizes to specific membrane microdomains in a novel human genomic reporter cellular model. Hum Mol Genet. 2009;18:4022–4034. doi: 10.1093/hmg/ddp346. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Fabbri M., Perez-Lloret S., Rascol O. Therapeutic strategies for Parkinson's disease: promising agents in early clinical development. Expert Opin Investig Drugs. 2020;29:1249–1267. doi: 10.1080/13543784.2020.1814252. [DOI] [PubMed] [Google Scholar]
  • 125.Zhang J., Deng X., Choi H.G., Alessi D.R., Gray N.S. Characterization of TAE684 as a potent LRRK2 kinase inhibitor. Bioorganic Med Chem Lett. 2012;22:1864–1869. doi: 10.1016/j.bmcl.2012.01.084. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Choi H.G., Zhang J., Deng X., Hatcher J.M., Patricelli M.P., Zhao Z. Brain penetrant LRRK2 inhibitor. ACS Med Chem Lett. 2012;3:658–662. doi: 10.1021/ml300123a. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Chen H., Chan B.K., Drummond J., Estrada A.A., Gunzner-Toste J., Liu X. Discovery of selective LRRK2 inhibitors guided by computational analysis and molecular modeling. J Med Chem. 2012;55:5536–5545. doi: 10.1021/jm300452p. [DOI] [PubMed] [Google Scholar]
  • 128.Estrada A.A., Chan B.K., Baker-Glenn C., Beresford A., Burdick D.J., Chambers M. Discovery of highly potent, selective, and brain-penetrant aminopyrazole Leucine-rich repeat kinase 2 (LRRK2) small molecule inhibitors. J Med Chem. 2014;57:921–936. doi: 10.1021/jm401654j. [DOI] [PubMed] [Google Scholar]
  • 129.Fuji R.N., Flagella M., Baca M., Baptista M.A.S., Brodbeck J., Chan B.K. Effect of selective LRRK2 kinase inhibition on nonhuman primate lung. Sci Transl Med. 2015;7:273ra15. doi: 10.1126/scitranslmed.aaa3634. [DOI] [PubMed] [Google Scholar]
  • 130.Henderson J.L., Kormos B.L., Hayward M.M., Coffman K.J., Jasti J., Kurumbail R.G. Discovery and preclinical profiling of 3-[4-(morpholin-4-yl)-7H-pyrrolo[2,3-d]pyrimidin-5-yl]benzonitrile (PF-06447475), a highly potent, selective, brain penetrant, and in vivo active LRRK2 kinase inhibitor. J Med Chem. 2015;58:419–432. doi: 10.1021/jm5014055. [DOI] [PubMed] [Google Scholar]
  • 131.Scott J.D., DeMong D.E., Greshock T.J., Basu K., Dai X., Harris J. Discovery of a 3-(4-pyrimidinyl) indazole (MLi-2), an orally available and selective leucine-rich repeat kinase 2 (LRRK2) inhibitor that reduces brain kinase activity. J Med Chem. 2017;60:2983–2992. doi: 10.1021/acs.jmedchem.7b00045. [DOI] [PubMed] [Google Scholar]
  • 132.Baptista M.A.S., Merchant K., Barrett T., Bhargava S., Bryce D.K., Michael Ellis J. LRRK2 inhibitors induce reversible changes in nonhuman primate lungs without measurable pulmonary deficits. Sci Transl Med. 2020;12 doi: 10.1126/scitranslmed.aav0820. eaav0820. [DOI] [PubMed] [Google Scholar]
  • 133.Andersen M.A., Wegener K.M., Larsen S., Badolo L., Smith G.P., Jeggo R. PFE-360-induced LRRK2 inhibition induces reversible, non-adverse renal changes in rats. Toxicology. 2018;395:15–22. doi: 10.1016/j.tox.2018.01.003. [DOI] [PubMed] [Google Scholar]
  • 134.Zhao Y., Keshiya S., Perera G., Schramko L., Halliday G.M., Dzamko N. LRRK2 kinase inhibitors reduce alpha-synuclein in human neuronal cell lines with the G2019S mutation. Neurobiol Dis. 2020;144:105049. doi: 10.1016/j.nbd.2020.105049. [DOI] [PubMed] [Google Scholar]
  • 135.Qin Q., Zhi L.T., Li X.T., Yue Z.Y., Li G.Z., Zhang H. Effects of LRRK2 inhibitors on nigrostriatal dopaminergic neurotransmission. CNS Neurosci Ther. 2017;23:162–173. doi: 10.1111/cns.12660. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Rui Q., Ni H., Gao F., Dang B., Li D., Gao R. LRRK2 contributes to secondary brain injury through a p38/drosha signaling pathway after traumatic brain injury in rats. Front Cell Neurosci. 2018;12:51. doi: 10.3389/fncel.2018.00051. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Cording A.C., Shiaelis N., Petridi S., Middleton C.A., Wilson L.G., Elliott C.J.H. Targeted kinase inhibition relieves slowness and tremor in a Drosophila model of LRRK2 Parkinson's disease. Npj Park Dis. 2017;3:34. doi: 10.1038/s41531-017-0036-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Bolognin S., Fossépré M., Qing X., Jarazo J., Ščančar J., Moreno E.L. 3D cultures of Parkinson's disease-specific dopaminergic neurons for high content phenotyping and drug testing. Adv Sci. 2019;6:1800927. doi: 10.1002/advs.201800927. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Saez-Atienzar S., Bonet-Ponce L., Blesa J.R., Romero F.J., Murphy M.P., Jordan J. The LRRK2 inhibitor GSK2578215A induces protective autophagy in SH-SY5Y cells: involvement of Drp-1-mediated mitochondrial fission and mitochondrial-derived ROS signaling. Cell Death Dis. 2014;5:e1368. doi: 10.1038/cddis.2014.320. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Tolosa E., Vila M., Klein C., Rascol O. LRRK2 in Parkinson disease: challenges of clinical trials. Nat Rev Neurol. 2020;16:97–107. doi: 10.1038/s41582-019-0301-2. [DOI] [PubMed] [Google Scholar]
  • 141.Qian X., Li X., Cai Q., Zhang C., Yu Q., Jiang Y. Phosphoglycerate kinase 1 phosphorylates beclin-1 to induce autophagy. Mol Cell. 2017;65:917–931. doi: 10.1016/j.molcel.2017.01.027. e6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Spencer B., Potkar R., Trejo M., Rockenstein E., Patrick C., Gindi R. Beclin-1 gene transfer activates autophagy and ameliorates the neurodegenerative pathology in α-synuclein models of Parkinson's and Lewy body diseases. J Neurosci. 2009;29:13578–13588. doi: 10.1523/JNEUROSCI.4390-09.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Savolainen M.H., Yan X., Myöhänen T.T., Huttunen H.J. Prolyl oligopeptidase enhances α-synuclein dimerization via direct protein−protein interaction. J Biol Chem. 2015;290:5117–5126. doi: 10.1074/jbc.M114.592931. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Song J.X., Lu J.H., Liu L.F., Chen L.L., Durairajan S.S.K., Yue Z. HMGB1 is involved in autophagy inhibition caused by SNCA/α-synuclein overexpression : a process modulated by the natural autophagy inducer corynoxine B. Autophagy. 2014;10:144–154. doi: 10.4161/auto.26751. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Yang G., Li J., Cai Y., Yang Z., Li R., Fu W. Glycyrrhizic acid alleviates 6-hydroxydopamine and corticosterone-induced neurotoxicity in SH-SY5Y cells through modulating autophagy. Neurochem Res. 2018;43:1914–1926. doi: 10.1007/s11064-018-2609-5. [DOI] [PubMed] [Google Scholar]
  • 146.Chen Y., Zhang W., Guo X., Ren J., Gao A. The crosstalk between autophagy and apoptosis was mediated by phosphorylation of Bcl-2 and beclin1 in benzene-induced hematotoxicity. Cell Death Dis. 2019;10:1–15. doi: 10.1038/s41419-019-2004-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Palmieri M., Impey S., Kang H., di Ronza A., Pelz C., Sardiello M. Characterization of the CLEAR network reveals an integrated control of cellular clearance pathways. Hum Mol Genet. 2011;20:3852–3866. doi: 10.1093/hmg/ddr306. [DOI] [PubMed] [Google Scholar]
  • 148.Settembre C., Di Malta C., Polito V.A., Arencibia M.G., Vetrini F., Erdin S. TFEB links autophagy to lysosomal biogenesis. Science. 2011;332:1429–1433. doi: 10.1126/science.1204592. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Napolitano G., Esposito A., Choi H., Matarese M., Benedetti V., Di Malta C. mTOR-dependent phosphorylation controls TFEB nuclear export. Nat Commun. 2018;9:3312. doi: 10.1038/s41467-018-05862-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Jiang T.F., Zhang Y.J., Zhou H.Y., Wang H.M., Tian L.P., Liu J. Curcumin ameliorates the neurodegenerative pathology in A53T α-synuclein cell model of Parkinson's disease through the downregulation of mTOR/p70S6K signaling and the recovery of macroautophagy. J Neuroimmune Pharmacol. 2013;8:356–369. doi: 10.1007/s11481-012-9431-7. [DOI] [PubMed] [Google Scholar]
  • 151.Forouzanfar F., Read M.I., Barreto G.E., Sahebkar A. Neuroprotective effects of curcumin through autophagy modulation. IUBMB Life. 2020;72:652–664. doi: 10.1002/iub.2209. [DOI] [PubMed] [Google Scholar]
  • 152.Liang G., Zhou H., Wang Y., Gurley E.C., Feng B., Chen L. Inhibition of LPS-induced production of inflammatory factors in the macrophages by mono-carbonyl analogues of curcumin. J Cell Mol Med. 2009;13:3370–3379. doi: 10.1111/j.1582-4934.2009.00711.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Mosley C.A., Liotta D.C., Snyder J.P. Highly active anticancer curcumin analogues. Adv Exp Med Biol. 2007;595:77–103. doi: 10.1007/978-0-387-46401-5_2. [DOI] [PubMed] [Google Scholar]
  • 154.Rusmini P., Cortese K., Crippa V., Cristofani R., Cicardi M.E., Ferrari V. Trehalose induces autophagy via lysosomal-mediated TFEB activation in models of motoneuron degeneration. Autophagy. 2019;15:631–651. doi: 10.1080/15548627.2018.1535292. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Altarescu G., Hill S., Wiggs E., Jeffries N., Kreps C., Parker C.C. The efficacy of enzyme replacement therapy in patients with chronic neuronopathic gaucher's disease. J Pediatr. 2001;138:539–547. doi: 10.1067/mpd.2001.112171. [DOI] [PubMed] [Google Scholar]
  • 156.Richter F., Fleming S.M., Watson M., Lemesre V., Pellegrino L., Ranes B. A GCase chaperone improves motor function in a mouse model of synucleinopathy. Neurotherapeutics. 2014;11:840–856. doi: 10.1007/s13311-014-0294-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Sawkar A.R., Schmitz M., Zimmer K.P., Reczek D., Edmunds T., Balch W.E. Chemical chaperones and permissive temperatures alter localization of Gaucher disease associated glucocerebrosidase variants. ACS Chem Biol. 2006;1:235–251. doi: 10.1021/cb600187q. [DOI] [PubMed] [Google Scholar]
  • 158.Mullin S., Smith L., Lee K., D’Souza G., Woodgate P., Elflein J. Ambroxol for the treatment of patients with Parkinson disease with and without glucocerebrosidase gene mutations: a nonrandomized, noncontrolled Trial. JAMA Neurol. 2020;77:427–434. doi: 10.1001/jamaneurol.2019.4611. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Blanz J., Saftig P. Parkinson's disease: acid-glucocerebrosidase activity and alpha-synuclein clearance. J Neurochem. 2016;139(Suppl):198–215. doi: 10.1111/jnc.13517. [DOI] [PubMed] [Google Scholar]
  • 160.Weikum E.R., Liu X., Ortlund E.A. The nuclear receptor superfamily: a structural perspective. Protein Sci. 2018;27:1876–1892. doi: 10.1002/pro.3496. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Mazaira G.I., Zgajnar N.R., Lotufo C.M., Daneri-Becerra C., Sivils J.C., Soto O.B. The nuclear receptor field: a historical overview and future challenges. Nucl Recept Res. 2018;5:101320. doi: 10.11131/2018/101320. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Giguère V. Transcriptional control of energy homeostasis by the estrogen-related receptors. Endocr Rev. 2008;29:677–696. doi: 10.1210/er.2008-0017. [DOI] [PubMed] [Google Scholar]
  • 163.Giguère V., Yang N., Segui P., Evans R.M. Identification of a new class of steroid hormone receptors. Nature. 1988;331:91–94. doi: 10.1038/331091a0. [DOI] [PubMed] [Google Scholar]
  • 164.Ranhotra H.S. The estrogen-related receptor alpha: the oldest, yet an energetic orphan with robust biological functions. J Recept Signal Transduct. 2010;30:193–205. doi: 10.3109/10799893.2010.487493. [DOI] [PubMed] [Google Scholar]
  • 165.Yogalingam G., Pendergast A.M. Abl kinases regulate autophagy by promoting the trafficking and function of lysosomal components. J Biol Chem. 2008;283:35941–35953. doi: 10.1074/jbc.M804543200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Ibrahim Abushouk A., Negida A., Abdelsalam Elshenawy R., Zein H., Hammad A.M., Menshawy A. C-Abl inhibition; A novel therapeutic target for Parkinson's disease. CNS Neurol Disord Drug Targets. 2017;16:14–21. doi: 10.2174/1871527316666170602101538. [DOI] [PubMed] [Google Scholar]
  • 167.Fowler A.J., Hebron M., Missner A.A., Wang R., Gao X., Kurd-Misto B.T. Multikinase Abl/DDR/Src inhibition produces optimal effects for tyrosine kinase inhibition in neurodegeneration. Drugs R D. 2019;19:149–166. doi: 10.1007/s40268-019-0266-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Dorsey J.F., Jove R., Kraker A.J., Wu J. The pyrido[2,3-d]pyrimidine derivative PD180970 inhibits p210 (Bcr-Abl) tyrosine kinase and induces apoptosis of K562 leukemic cells. Cancer Res. 2000;60:3127–3131. [PubMed] [Google Scholar]
  • 169.Capdeville R., Buchdunger E., Zimmermann J., Matter A. Glivec (ST1571, imatinib), a rationally developed, targeted anticancer drug. Nat Rev Drug Discov. 2002;1:493–502. doi: 10.1038/nrd839. [DOI] [PubMed] [Google Scholar]
  • 170.Rossari F., Minutolo F., Orciuolo E. Past, present, and future of Bcr-Abl inhibitors: from chemical development to clinical efficacy. J Hematol Oncol. 2018;11:84. doi: 10.1186/s13045-018-0624-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Weisberg E., Manley P.W., Breitenstein W., Brüggen J., Cowan-Jacob S.W., Ray A. Characterization of AMN107, a selective inhibitor of native and mutant Bcr-Abl. Cancer Cell. 2005;7:129–141. doi: 10.1016/j.ccr.2005.01.007. [DOI] [PubMed] [Google Scholar]
  • 172.Mahul-Mellier A.L., Fauvet B., Gysbers A., Dikiy I., Oueslati A., Georgeon S. C-Abl phosphorylates α-synuclein and regulates its degradation: implication for α-synuclein clearance and contribution to the pathogenesis of Parkinson's disease. Hum Mol Genet. 2014;23:2858–2879. doi: 10.1093/hmg/ddt674. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Hunn B.H.M., Vingill S., Threlfell S., Bannerman D.M., Cragg S.J., Wade-martins R. Impairment of macroautophagy in dopamine neurons has opposing effects on Parkinsonian pathology and behavior. Cell Rep. 2019;29:920–931. doi: 10.1016/j.celrep.2019.09.029. e7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Li Z., Wang C., Wang Z., Zhu C., Li J., Sha T. Allele-selective lowering of mutant HTT protein by HTT–LC3 linker compounds. Nature. 2019;575:203–209. doi: 10.1038/s41586-019-1722-1. [DOI] [PubMed] [Google Scholar]
  • 175.Li Z., Zhu C., Ding Y., Fei Y., Lu B. ATTEC: a potential new approach to target proteinopathies. Autophagy. 2020;16:185–187. doi: 10.1080/15548627.2019.1688556. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Takahashi D., Arimoto H. Targeting selective autophagy by AUTAC degraders. Autophagy. 2020;16:765–766. doi: 10.1080/15548627.2020.1718362. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Takahashi D., Moriyama J., Nakamura T., Miki E., Takahashi E., Sato A. AUTACs: cargo-specific degraders using selective autophagy. Mol Cell. 2019;76:797–810. doi: 10.1016/j.molcel.2019.09.009. e10. [DOI] [PubMed] [Google Scholar]
  • 178.Johnson M.E., Stecher B., Labrie V., Brundin L., Brundin P. Triggers, facilitators, and aggravators: redefining Parkinson's disease pathogenesis. Trends Neurosci. 2019;42:4–13. doi: 10.1016/j.tins.2018.09.007. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Acta Pharmaceutica Sinica. B are provided here courtesy of Elsevier

RESOURCES