Next Article in Journal
Effect of Soluble Adenylyl Cyclase (ADCY10) Inhibitors on the LH-Stimulated cAMP Synthesis in Mltc-1 Leydig Cell Line
Next Article in Special Issue
Glucocerebrosidase Gene Therapy Induces Alpha-Synuclein Clearance and Neuroprotection of Midbrain Dopaminergic Neurons in Mice and Macaques
Previous Article in Journal
The Chemistry of Reactive Oxygen Species (ROS) Revisited: Outlining Their Role in Biological Macromolecules (DNA, Lipids and Proteins) and Induced Pathologies
Previous Article in Special Issue
A New Tool to Study Parkinsonism in the Context of Aging: MPTP Intoxication in a Natural Model of Multimorbidity
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Genes Implicated in Familial Parkinson’s Disease Provide a Dual Picture of Nigral Dopaminergic Neurodegeneration with Mitochondria Taking Center Stage

by
Rafael Franco
1,2,†,
Rafael Rivas-Santisteban
1,2,†,
Gemma Navarro
2,3,†,
Annalisa Pinna
4,*,† and
Irene Reyes-Resina
1,†,‡
1
Department Biochemistry and Molecular Biomedicine, University of Barcelona, 08028 Barcelona, Spain
2
Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CiberNed), Instituto de Salud Carlos III, 28031 Madrid, Spain
3
Department Biochemistry and Physiology, School of Pharmacy and Food Sciences, University of Barcelona, 08028 Barcelona, Spain
4
National Research Council of Italy (CNR), Neuroscience Institute–Cagliari, Cittadella Universitaria, Blocco A, SP 8, Km 0.700, 09042 Monserrato (CA), Italy
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Current address: RG Neuroplasticity, Leibniz Institute for Neurobiology, Brenneckestr 6, 39118 Magdeburg, Germany.
Int. J. Mol. Sci. 2021, 22(9), 4643; https://doi.org/10.3390/ijms22094643
Submission received: 29 March 2021 / Revised: 16 April 2021 / Accepted: 20 April 2021 / Published: 28 April 2021
(This article belongs to the Special Issue Neurobiology of Parkinson’s Disease)

Abstract

:
The mechanism of nigral dopaminergic neuronal degeneration in Parkinson’s disease (PD) is unknown. One of the pathological characteristics of the disease is the deposition of α-synuclein (α-syn) that occurs in the brain from both familial and sporadic PD patients. This paper constitutes a narrative review that takes advantage of information related to genes (SNCA, LRRK2, GBA, UCHL1, VPS35, PRKN, PINK1, ATP13A2, PLA2G6, DNAJC6, SYNJ1, DJ-1/PARK7 and FBXO7) involved in familial cases of Parkinson’s disease (PD) to explore their usefulness in deciphering the origin of dopaminergic denervation in many types of PD. Direct or functional interactions between genes or gene products are evaluated using the Search Tool for the Retrieval of Interacting Genes/Proteins (STRING) database. The rationale is to propose a map of the interactions between SNCA, the gene encoding for α-syn that aggregates in PD, and other genes, the mutations of which lead to early-onset PD. The map contrasts with the findings obtained using animal models that are the knockout of one of those genes or that express the mutated human gene. From combining in silico data from STRING-based assays with in vitro and in vivo data in transgenic animals, two likely mechanisms appeared: (i) the processing of native α-syn is altered due to the mutation of genes involved in vesicular trafficking and protein processing, or (ii) α-syn mutants alter the mechanisms necessary for the correct vesicular trafficking and protein processing. Mitochondria are a common denominator since both mechanisms require extra energy production, and the energy for the survival of neurons is obtained mainly from the complete oxidation of glucose. Dopamine itself can result in an additional burden to the mitochondria of dopaminergic neurons because its handling produces free radicals. Drugs acting on G protein-coupled receptors (GPCRs) in the mitochondria of neurons may hopefully end up targeting those receptors to reduce oxidative burden and increase mitochondrial performance. In summary, the analysis of the data of genes related to familial PD provides relevant information on the etiology of sporadic cases and might suggest new therapeutic approaches.

1. Introduction

Parkinson’s disease is a prevalent neurodegenerative disease the first clinical symptoms of which include tremors and motor dysfunction. The number of cases with unknown causes is much higher than the number of familial cases; that is, cases due to known genetic alterations. Years ago, the percentage of cases of inheritable PD was estimated to be around 5%. Recent estimates increased this percentage because of Genetic-oriented research, which led to the discovery of more genes associated with the disease. In a recent multicenter study in a cohort of 1587 cases, mutations were found in 14.1% of patients [1] (see also [2,3]). Although familial cases often display early-onset symptoms, the main risk factor is age with 60 years being considered as the threshold for developing symptoms in sporadic cases. In these cases, many positive (e.g., caffeine) and negative (e.g., toxin exposure) risk factors are known [4,5,6,7,8,9,10,11,12,13], but the exact causes of why dopamine neurons of the substantia nigra die in both familial and sporadic cases remain unknown.
The work of Hornykiewicz and colleagues was instrumental for early therapeutic intervention in PD cases. They discovered the lack of dopamine in certain brain areas as the cause of motor symptoms and noticed that dopamine supplementation was not effective because the compound is unable to cross the blood–brain barrier. They discovered that L-DOPA (levodopa), the precursor of the neurotransmitter, was able to enter the brain and there be converted into dopamine. L-DOPA is still used in PD therapy. The need for chronic treatment and fluctuations in drug levels in the brain may lead to some side effects, mainly dyskinesia [14,15,16,17,18,19]. Side effects may be addressed via surgical procedures. Since the nineties, the technique has been refined and used with success by implanting electrodes that achieve what is known as deep-brain stimulation (DBS) [20,21,22,23,24,25,26,27,28]. Unfortunately, there is no therapeutic intervention that delays disease progression, that is, the neurodegeneration of dopaminergic nigral neurons.
PD results from death of dopamine-producing neurons of the substantia nigra. An imbalance of dopaminergic neurotransmission in this area of the brain leads to motor deficits, which first appear as tremors and difficulty starting to walk and moving limbs precisely. Be it cause or consequence, there is a main characteristic of pathological PD: the appearance of Lewy bodies formed by aggregates of a protein, α-synuclein (α-syn), the function of which has not yet been fully elucidated. PD has analogies and differences with other diseases known as α-synucleinopathies; the two other main α-synucleinopathies are dementia with Lewy bodies and multiple system atrophy (see [29,30,31]). The deposition of α-syn aggregates occurs in the brain of patients with both familial and sporadic PD. Finally, it should be noted that a theory has arisen that postulates that the risk of PD may be increased by some viral infections [32].
Taking into account the familial cases, i.e., mutations in SNCA, LRRK2, GBA, UCHL1, VPS35, PRKN, PINK1, ATP13A2, PLA2G6, DNAJC6, SYNJ1, DJ-1/PARK7 and FBXO7 genes, it appears that (i) mutant forms of α-syn can aggregate and lead to early-onset PD and (ii) that mutations in other genes lead to deposition of non-mutated α-syn and early onset of the EP. The present narrative review aims to propose a scenario in which these players are connected; that is, we would like to obtain information from familial cases that could help decipher the causes of dopaminergic denervation in all types of PD.
The approach is sustained in two qualitatively different datasets. First, a blind approach is used to find the connections between genes that have alterations associated with the disease. First, we selected genes related to PD familial cases (listed in Table 1) to find interactions among them (in a blind-like approach) using the Search Tool for the Retrieval of Interacting Genes/Proteins (STRING). Second, information from transgenic models was analyzed in the search for mechanisms of phenotypic alterations (focused on the CNS and, whenever possible, considering alterations in the substantia nigra/striatum). From these two independent sets of data we finally presented a picture of the potential events that occur in degenerating dopaminergic neurons.

2. STRING Analysis Restricted to Connections Obtained from Experimental, Gene Fusion, Protein Analogy or Proximity Data

STRING 11.0 version was used in the analysis of the potential connections among genes in Table 1. Both software and platform are incorporated into the European “ELIXIR Core Data Resources”; it is freely available and analyses were performed on-line in https://string-db.org (accessed on 4 March 2021).
The STRING default setting for the genes associated with familial PD and listed in Table 1 leads to Figure 1A, in which virtually all genes (or gene products) are interconnected. We have omitted genes that were more recently associated with familial PD but for which less data was available [33,34]. Figure 1A shows that PLA2G6 is linked to UCHL1 and BNAJc6 only by neighborhood, meaning that there are no studies confirming whether or not there is any functional link between them. Obviously, at the bibliographic level, all genes are related to Parkinson’s disease; therefore, we next eliminated the connections derived from the mention of genes in papers related to PD.
At first glance, avoiding the bias due to appearance in PD-related papers, the number of interactions is markedly reduced. On the one hand, restricting connections to those derived from experimental data, gene fusion, protein analogy, or proximity, 5 gene products were found to be interconnected (See Figure 1B). All except the PARK2/PARK7 potential interaction were (according to STRING database) experimentally determined (pink line between elements). On the other hand, an interaction between SYNJ1 and DNAJC6 products was identified. Overall, 6 elements were not connected to each other. If we enrich the pattern, including partners that were not present in the initial analysis, we obtain Figure 1C, in which the 6 elements still remain non-connected. Patterns arising using these 6 genes as individual elements are in the Supplementary Material Figure S1 (settings exclude co-appearance in PD-related papers). Whereas 5 of them showed interrelationships with other proteins, the product of PLA2G6, 85/88 kDa calcium-independent phospholipase A2, did not display any interaction related to experiments, gene fusion, co-expression, co-occurrence or neighborhood, meaning that its role is not yet well deciphered. In contrast, the protein encoded by UCLH1, ubiquitin carboxyl-terminal hydrolase isozyme L1, is an element with relevant links that include experimental data, protein homology and gene and protein coexpression. Actually, the search of mechanisms for non-gene-associated PD cases is the aim of this paper.

3. Lessons from Interactions among PARK2, PARK7, PINK1, LRRK2 and VPS35

The proteins encoded by PARK2, PARK7, PINK1, LRRK2 and VPS35 have a complementary role related to subcellular organelles, mainly the lysosome. Furthermore, practically all gene products participate in protein processing, in which ubiquitination is a relevant mechanism. Ubiquitination was described decades ago as a mechanism for targeting a protein for degradation [35,36,37]. If the processes involving ubiquitination are not completely balanced, the handling of proteins is inadequate, and the cell is damaged. Apart from ubiquitin-protein ligase (PARK2/parkin), other elements also participate in ubiquitination-mediated processes that lead to proteasomal degradation. This view is in agreement with the finding that some inherited PD cases are associated with the PARK7, PARK6, and PARK2 loci [38]. Mutant forms of DJ-1/PARK7 that are associated with PD lead to differential interactions with E3 ubiquitin-protein ligase leading to altering protein processing or leading to oxidative stress [39]. PINK1 is genetically associated with parkin. The discovery first made in Drosophila melanogaster [40,41] was later confirmed in humans [42]. In 2005, Smith and colleagues demonstrated an interaction of leucin-rich repeat kinase 2 (product of LRRK2) with parkin and showed that mutant forms of the kinase may induce neuronal death [43]. Vacuolar protein sorting-associated protein 35 is involved in the vesicular transport of vesicles from mitochondria. Such mitochondrial-derived vesicles are linked to ubiquitination in a complex way (see [44] for details). Furthermore, the ubiquitin carboxyl-terminal hydrolase isozyme L1 (UCHL1) and the F-box component of protein 3 ubiquitin-protein ligase complex (FBXO7), have a role in targeting protein degradation using the ubiquitination pathway.

4. Lessons from Interactions between and from SYNJ1 and DNAJC6

The link between these two genes is quite straightforward. On the one hand, SYNJ1 codes for Synaptojanin-1, which is a quite unspecific phosphatase because it may degrade different phosphoinositides, which are key components on biological membranes. On the other hand, DNAJC6 codes for auxilin, which is a protein phosphatase involved in vesicle handling; in addition, auxilin belongs to a family of chaperones (DNAJ/HSP40). Knowledge of the function of these proteins is only partial, but at present they seem related to early events occurring shortly after endocytosis. Data on their involvement in the trafficking of intracellular vesicles or targeting of proteins for degradation is scant. In fact, genetic deletion of DNAJC6 in mice results in death due to the inability to perform clathrin-mediated endocytosis [45]. A study in Saccharomyces cerevisiae confirms that both the product of Dnajc6 and Synaptojanin-1 are necessary for the uncoating of clathrin, the proper trafficking of endosomes and the delivery of endocytic cargo [46,47].

5. Enriching the Network

STRING is a powerful tool as it permits the enrichment of networks but avoiding the bias of parkinsonism in the enrichment. In doing so, the PARK2/PARK7/PINK1/LRRK2/VPS35 network connects with the SYNJ1/DNAJC6 network. Obviously, this only is possible if more genes, related or not with PD, are considered. The resulting enriched network is in Figure 2.
First of all, Figure 2 shows that only 3 PD-related genes are not connected to the rest (GBA, ATP132A and PLA2G6). Importantly, VPS35 becomes a relevant node that is connected, on the one side to several proteins involved in intracellular vesicular trafficking (vesicular sorting proteins, sorting nexins and Wnt Ligand Secretion Mediator, WLS) and, on the other side, directly to two gene products related to PD (PINK1 and LRRK2) and, indirectly with all other PD-related genes except GBA, ATP132A and PLA2G6. Also of note is that the network involving most of the PD-related genes (Figure 2, see connections involving VPS35) contains elements having a variety of functions in common: ubiquitination, clathrin-mediated endocytosis and phosphoinositide handling. In the far bottom right of Figure 2, α-synuclein (α-syn) appears only connected to ubiquitin-60S ribosomal protein L40 due to protein homology.
In summary, the study of genes related with inherited forms of PD shows that the products of the genes participate in intracellular vesicular transport and in clathrin-mediated endocytosis, which is subsequent to the activation of cell surface G protein-coupled receptors (GPCRs).

6. Lessons from Transgenic Animals

Transgenic animals have been fundamental to understanding disease mechanisms. While in non-neurological diseases many models are based on the genetic inactivation of a gene in neurodegenerative diseases (providing so-called knockout (KO) animals), the approach of overexpressing a mutated or non-mutated form of a human protein is often used. In a set of tables, we present a summary of the data obtained from transgenic animals, both generated for the purpose of better understanding the disease of PD, or previously generated for another purpose, but having provided data on alterations of the functionality of the nervous system.
Data from transgenic animals of other PD-related genes and the relevant references are included in Table 3 and Table 4 and in Supplementary Table S1. After a selection of ad hoc papers, these 3 tables were constructed to summarize data from a significant number of studies indicating for each the transgenic model and the main anomalies found after in vivo or in vitro studies.
In accordance with the leading role of α-syn in synucleinopathies such as PD [48], there are several transgenic animals expressing different versions of the SNCA gene and several studies on them. The results of these studies are interesting despite their bias as the link between α-syn and synucleinopathies is obvious. On the one hand, the lack of a gene (mouse SNCA knockout) produced viable animals with some mild abnormalities. It is the overexpression of the human gene that led to neural alterations. On the other hand, the expression of a non-phosphorylatable (S129A) mutation on an SNCA knockout background did not lead to any significant alteration in neural function. Neural alterations including dopaminergic neuronal death appeared upon overexpression of some gene mutants: often the mutant version included a phosphorylatable amino acid or, the other way around, eliminates a phosphorylatable residue. Findings using such proteins that may be differentially phosphorylated could link α-syn to the PD-associated genes that code for a kinase or a phosphatase. Also of interest was fact that multimers of the protein in samples from a knockout model attenuate synaptic vesicle recycling and that the α-syn spread correlated with dopaminergic neurodegeneration. Apparently unrelated to PD-associated genes was an alteration in the effect of neurotrophic factors that were detected in some of those animals. Finally, we would like to highlight that some of the studies reported alterations in mitochondrial dynamics or function. If mutated α-syn has more or less phosphorylatable sites, this may be related to an energy imbalance; in fact, kinase action requires ATP.
Animals that are KO for the Lrrk2 gene or overexpress human versions (mutant versions included) of this gene, consistently showed synaptogenetic deficiencies in intracellular vesicle trafficking and function and in protein processing. Interestingly, the G2019S mutation of the LRRK2 kinase produced neurodegeneration in a rodent model of human α-syn overexpression. The line expressing the G2019S mutation also displayed alterations in both homeostasis and neuronal morphogenesis.
Table 3. Findings in animal models related to Snca.
Table 3. Findings in animal models related to Snca.
Animal Model (s)In VivoIn VitroExpression LevelMain FindingsRef.
Mice harbouring Sncatm1Nbm mutation (n.s.n) XSnca KO miceModulates microglial activation phenotype
Increased levels of reactive marker proteins
[49]
Mice harbouring Sncatm1Nbm mutation (n.s.n) XSnca KO miceAltered palmitate metabolism[50]
Mice harbouring Sncatm1Nbm mutation (n.s.n) XSnca KO miceMitochondrial lipid abnormality
Electron transport chain impairment
[51]
Mice harbouring Sncatm1Nbm mutation (n.s.n)XXSnca KO miceSynaptic vesicle depletion[52]
C57BL/6N-Sncatm1Mjff/JXXSnca KO miceNon-altered mitochondrial bioenergetics[53]
B6; 129 × 1-Sncatm1Rosl/J XSnca KO miceα-syn restricts RNA viral infections in the brain[54]
B6; 129 × 1-Sncatm1Rosl/J XSnca KO miceROS and NOS-2 decreased in mature
erythrocytes
[55]
B6; 129 × 1-Sncatm1Rosl/J XSnca KO miceNo modification in pale body-like inclusion
Altered proteasome function
[56]
B6; 129 × 1-Sncatm1Rosl/J XSnca KO miceInhibition of intrasynaptic vesicle mobility
Maintains recycling-pool homeostasis
[57]
B6; 129 × 1-Sncatm1Rosl/JX Snca KO miceCognitive impairments[58]
B6; 129 × 1-Sncatm1Rosl/J XSnca KO miceVulnerability of peripheral catecholaminergic neurons to MPTP not regulated by α-synuclein[59]
B6; 129 × 1-Sncatm1Rosl/J XSnca KO miceResistant to mitochondrial toxins[60]
B6; 129 × 1-Sncatm1Rosl/JXXSnca KO miceDeficits in the nigrostriatal dopamine system[61]
B6; 129 × 1-Sncatm1Rosl/J
C57Bl/6JOlaHsd
XXSnca KO miceDecreased impulsivity[62]
B6; 129 × 1-Sncatm1Rosl/J
C57Bl/6JOlaHsd
X Snca KO miceNeuromuscular pathology[63]
B6; 129 × 1-Sncatm1Rosl/J
C57Bl/6JOlaHsd
XXSnca KO miceDecreased reuptake of dopamine in the dorsal striatum[64]
B6; 129 × 1-Sncatm1Rosl/J (Snca KO)
B6-TgHSNCGtm1VLB (Sncg KO)
XX-α-syn (Snca) KO mice
-ɣ-syn (Sncg) KO mice
Altered dopamine metabolism [65]
B6; 129 × 1-Sncatm1Rosl/J (Snca KO)
B6-TgHSNCGtm1VLB (Sncg KO)
XX-α-syn (Snca) KO mice
-ɣ-syn (Sncg) KO mice
-α-ɣ-syn double KO mice
Increased striatal dopamine release
Hyperdopaminergic signs
[66]
Triple-synuclein-KO (TKO)XXα-syn (Snca), ß-syn (Sncb) and ɣ-syn (Sncg) triple KO miceFunctional alterations to the nigrostriatal system[67]
Triple-synuclein-KO (TKO)XXα-syn (Snca), ß-syn (Sncb) and ɣ-syn (Sncg) triple KO miceAltered synaptic vesicle endocytosis
α -, ß -, or γ-synucleins are functionally redundant
[68]
Triple-synuclein-KO (TKO)XXα-syn (Snca), ß-syn (Sncb) and ɣ-syn (Sncg) triple KO miceAge-dependent neuronal dysfunction[69]
Triple-synuclein-KO (TKO)
See paper for further details on model(s)
XXα-syn (Snca), ß-syn (Sncb) and ɣ-syn (Sncg) triple KO mice.
Mice overexpressing human SNCA mutants PARK1/hA30P or PARK4/hα-syn
Effects on presynaptic architecture[70]
B6;129 × 1-Sncatm1Rosl/J
B6.CgTg (SNCA) OVX37Rwm
Sncatm1Rosl/J
XX-Snca KO mice
-SNCA overexpressing mice
Blockade of TrkB neurotrophic effect[71]
B6;DBA-Tg (Thy1-SNCA) 61Ema XSNCA overexpressing miceImpairment of mitochondrial function
Elevated ROS in brain mitochondria
[72]
B6;DBA-Tg (Thy1-SNCA) 61Ema XSNCA overexpressing miceAlterations in corticostriatal synaptic plasticity[73]
B6;DBA-Tg (Thy1-SNCA) 61EmaXXSNCA overexpressing miceAlterations in calcium buffering capacity[74]
(Thy1)-(WT)a-syn
XSNCA overexpressing miceEnhanced axonal degeneration after
peripheral nerve lesion
[75]
B6.Cg-Tg (SNCA) OVX37Rwm Sncatm1Rosl/JXXSNCA overexpressing miceDeficits in dopaminergic transmission precede neuronal loss[76]
B6.Cg-Tg (SNCA) OVX37Rwm Sncatm1Rosl/J XSNCA overexpressing miceImpairment of macroautophagy in dopaminergic neurons[77]
B6; 129 × 1-Sncatm1Rosl/J neurons transfected with SNCA or TsixK-SNCA XCultured neurons from Snca KO mice overexpressing SNCA or TsixK-SNCAα-syn multimers attenuate synaptic vesicle recycling[78]
S129 mutations performed on mice harbouring Sncatm1Nbm mutation (n.s.n.)XXS129D-SNCA (phosphomimetic) or S129A- SNCA (non-phosphorylatable) overexpressing Snca KO miceNo abnormalities detected (in vivo)[79]
FVB;129S6-Sncatm1Nbm Tg (SNCA*A53T) 1Nbm Tg (SNCA*A53T) 2Nbm/J
FVB;129S6-Sncatm1Nbm Tg (SNCA*A30P) 1Nbm Tg (SNCA*A30P) 2Nbm/J
XXA53T-SNCA or A30P-SNCA overexpressing Snca KO miceEnteric nervous system abnormalities[80]
M83KO mice resulting from crossing M83 line with B6; 129X1-Sncatm1Rosl/J line XA30P-SNCA overexpressing Snca KO miceDopaminergic
neurodegeneration
[81]
B6.Cg-Sncatm1Rosl Tg (SNCA*A30P) 192Rwm/JXXA30P-SNCA overexpressing Snca KO miceRegion-specific deficits in dopamine signaling[82]
FVB;129-Tg (Prnp-SNCA*A53T) AAub/JXXA53T-SNCA overexpressing miceDysfunctional neurotransmission
Impaired synaptic plasticity
[83]
FVB;129-Tg (Prnp-SNCA*A53T) AAub/JXXA53T-SNCA overexpressing miceNeuronal dysfunction in the absence of aggregate formation
Behavioral alterations
[84]
NTac:SD-Tg (SNCA*A53T) 268CjliXXA53T-SNCA overexpressing miceDynamic changes in
striatal mGluR1 but not mGluR5
[85]
B6;C3-Tg (Prnp-SNCA*A53T) 83Vle/J
(also known as:A53T α-synuclein transgenic line M83)
XXBrain inoculation with brain homogenates from older Tg mice or with human α-syn fibrils in Tg A53T-SNCA overexpressing miceInoculation initiates a rapidly progressive neurodegenerative α-synucleinopathy[86]
NTac:SD-Tg (SNCA*E46K) 70CJLiXXE46K-SNCA overexpressing ratsEnhanced vulnerability to mitochondrial
impairment
[87]
Double transgenic Uchl1tm1Dgen:Thy1-maSNXXUch-L1 KO +
Snca overexpressing mice
Excess α-syn worsens disease in mice lacking Uch-L1[88]
B6; 129×1-Sncatm1Rosl/J
C57B/6jxSJL F3, Tg5093
X-Snca KO mice
-A53T-SNCA overexpressing mice
α-syn expression levels do not significantly affect proteasome function[89]
See paper for details of the animal model(s) X-A53T-SNCA transfected in WT neurons
-Snca KO mice
Altered fatty acid composition of
dopaminergic neuron
PUFAs enhance α-syn oligomerization
[90]
B6;129-Sncatm1Sud Sncbtm1.1Sud/J
See also paper for further details
X-Cultured neurons from Snca KO mice overexpressing SNCA
-See paper for other models used
Inhibition of synaptic vesicle reclustering after endocytosis[91]
B6;129×1-Sncatm1Rosl/JXXα-syn fibrils gut-injected in Snca KO miceα-syn transneuronal propagation from the gut to the brain[92]
WT vs. KOM2 XGlial cytoplasmic inclusions-α-syn or Lewy Bodies-α-syn injected in WT mice vs. mice that express human α-syn only in oligodendrocites (KOM2)Cellular milieu affects pathology of α-syn[93]
n.s.n.: Non standard nomenclature.
Recent results from genetic and pharmacological experiments in a cell model showed that knocking down LRRK2 using a shRNA-based approach reduces oxidative stress by means of mitophagy [94]. A recent review uncovers the role of mitophagy in PD and lists the genes that may be involved in such phenomenon; many of the above-described PD-related genes may participate in mitophagy regulation (see [95] and references therein).
The number of transgenic animals related to other PD-associated genes is lower. There are, however, a significant number of studies on PRKN, PINK1, and DJ-1-related transgenic lines. Remarkably, Prkn knockout animals are resistant to the toxics usually used to produce dopaminergic neurodegeneration in wild-type rats or mice. In contrast, the Pink1 knockout showed hypersensitivity to 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced dopaminergic neuronal loss and to activation of some of glutamate receptors (both ionotropic and metabotropic), thus affecting excitatory neurotransmission. Studies with transgenic lines also show that the PRKN gene product is involved in protein processing, particularly via ubiquitination. In addition, the double mutant line, so called “TwinkPark” mice showed decreased mitochondrial function and altered membrane potential.
Table 4. Findings in animal models related to Prkn, Lrrk2 and Pink1.
Table 4. Findings in animal models related to Prkn, Lrrk2 and Pink1.
GeneAnimal Model (s)In VivoIn VitroExpression LevelMain FindingsRef.
PRKNB6.129S4-Prkntm1Shn/JXXPrkn KO miceIndependent regulation of parkin ubiquitination and alpha-synuclein clearance[96]
B6.129S4-Prkntm1Shn/JXXPrkn KO miceAccelerated microtubule aging in dopaminergic neurons[97]
B6.129S4-Prkntm1Shn/J
XPrkn KO miceMyotubular atrophy
Impaired mitochondrial function and smaller myofiber area
[98]
B6.129S4-Prkntm1Shn/J XPrkn KO miceParkin mediates the ubiquitination of VPS35
Reduced WASH complexes
[99]
B6.129S4-Prkntm1Shn/JXXPrkn KO miceER stress and induced inflammation levels[100]
B6.129S4-Prkntm1Shn/JXXPrkn KO miceBehavioral impairments
Amplified EtOH-induced dopaminergic neurodegeneration, oxidative stress and apoptosis
Dysfunction of mitochondrial autophagy
[101]
B6.129S4-Prkntm1Shn/J
XPrkn KO miceParkin promotes proteasomal degradation of Synaptotagmin IV[102]
B6.129S4-Prkntm1Shn/JXXPrkn KO miceSNPH Cargo vesicle generation not affected[103]
B6.129S4-Prkntm1Shn/JXXPrkn KO miceExacerbated mitochondrial dysfunction in neurons[104]
B6.129S4-Prkntm1Shn/J
XXPrkn KO miceIncreased sensitivity to myocardial infarction
Reduced survival after the infarction
Reduced mitophagy
[105]
B6.129S4-Prkntm1Shn/JXXPrkn KO miceParkin antagonizes the death potential of FAF1[106]
B6.129S4-Prkntm1Shn/JXXPrkn KO miceAcutely sensitivity to oxidative stress
Inability to maintain Mcl-1 levels
Death of dopaminergic neurons
[107,108]
B6.129S4-Prkntm1Shn/JXXPrkn KO miceAberrant behavioral response to dopamine replacement therapy in PD[109]
B6.129S4-Prkntm1Shn/JXXPrkn KO miceResisted weight gain, steatohepatitis, and insulin resistance
Abolished hepatic fat uptake
[110]
B6.129S4-Prkntm1Shn/JXXPrkn KO miceReductions in the total catecholamine release
Impaired LTP and LTD
Normal levels of dopamine receptors and dopamine transporters
[111]
B6.129S4-Prkntm1Shn/JXXPrkn KO miceRequiring inflammatory stimulus for nigral DA neuron loss[112]
B6.129S4-Prkntm1Shn/JXXPrkn KO miceReduced respiratory capacity mitochondria (in striatal cells)
Delayed weight gain
Lower protection against ROS
[113]
B6.129S4-Prkntm1Shn/JXXPrkn KO miceIncreased extracellular dopamine concentration in the striatum
Deficits in behavioral tests
[114]
Double-mutant “TwinkPark” mice, resulting from crossing B6.129S4-Prkntm1Shn/J line with Twinkledup/+ lineXXPrkn KO (enhanced in the substantia nigra) miceDecrease of mitochondrial DNA
Low mitochondrial function and membrane potential
Neurobehavioral deficits
[115]
Crossing B6.129S4-Prkntm1Shn/J line with a Mcl-1 +/− line (n.s.n.)XXPrkn KO + Mcl-1 +/− miceDopaminergic neuron loss
Motor impairments
[107]
B6.129S4-Prkntm1Shn/J
B6.129S4-Pink1tm1Shn/J
XX-Prkn KO mice
-Pink1 KO mice
Inflammation rescued by STING-mediated action[116]
B6.129S4-Prkntm1Shn/J
B6.129S4-Pink1tm1Shn/J
XX-Prkn KO mice
-Pink1 KO mice
No repression of mitochondrial antigen presentation[117]
B6.129S4-Prkntm1Shn/J
LEH-Pink1tm1sage
X-Prkn KO mice
-Pink1 KO rats
Mitophagy of damaged mitochondria in axons requires PINK1 and Parkin[118]
Crossing
B6.129S4-Prkntm1Shn/J line and
B6.129S4-Pink1tm1Shn/J line
XPrkn/Pink1
double KO mice
Higher levels of ATP synthase
Denervated neuromuscular junctions
[119]
B6.129S4-Prkntm1Shn/J
B6.129S4-Pink1tm1Shn/J
B6.Cg-Park7tm1Shn/J
XX-Prkn KO mice
-Pink1 KO mice
-Dj-1 KO mice
Aberrant striatal synaptic plasticity in rodent models of autosomal recessive PD[120]
Crossing DASYN53 double-transgenic (tetO-SNCA*A53T) E2Cai/J line + DAT-PF-tTA) mice with B6.129S4-Prkntm1Shn/J line or with Pink1tm1Zhzh mutation line (n.s.n.)XXOverexpressing human A53T-SNCA in DA neurons and KO for either Prkn or Pink1
Pervasive mitochondrial macroautophagy defects
Dopamine neuron degeneration
[121]
B6;129-Pink1tm1Aub/J XPink1 KO miceAltered spontaneous EPSCs[122]
B6;129-Pink1tm1Aub/J XPink1 KO miceMitochondrial recruitment of parkin not affected[123]
B6;129-Pink1tm1Aub/JXXPink1 KO miceProgressive mitochondrial dysfunction in absence of neurodegeneration[124]
LRRK2B6.129X1(FVB)-Lrrk2tm1.1Cai/J XLrrk2 KO miceAlterations in protein synthesis
Alterations in degradation pathways
[125]
B6.129X1(FVB)-Lrrk2tm1.1Cai/JXXLrrk2 KO miceLRRK2 regulates synaptogenesis and dopamine receptor activation[126]
B6.129X1(FVB)-Lrrk2tm1.1Cai/J XLrrk2 KO miceLRRK2 regulates ER-Golgi export[127]
B6.129X1(FVB)-Lrrk2tm1.1Cai/JX Lrrk2 KO miceNeurons have more motile axonal and dendritic growth[128]
C57BL/6-Lrrk2tm1Mjfa/JXXLrrk2 KO miceLRRK2 modulates microglial phenotype and dopaminergic neurodegeneration[129]
C57BL/6-Lrrk2tm1Mjfa/J XLrrk2 KO miceStress-Related Gastrointestinal Dysmotility[130]
C57BL/6-Lrrk2tm1Mjfa/J XLrrk2 KO miceLRRK2 is required for Rip2 localization to DCVs[131]
C57BL/6-Lrrk2tm1Mjfa/J XLrrk2 KO miceSignificant increase of ceramide levels
Direct effects on GBA1
[132]
B6;129-Lrrk2tm2.1Shn/JXXLrrk2 KO miceImpairment of Autophagy[133]
B6;129-Lrrk2tm2.1Shn/J
B6;129-Lrrk2tm3.1Shn/J
XLrrk2 KO miceImpairment of protein degradation pathways
Apoptotic cell death
[134]
C57BL/6-Lrrk2tm1.1Mjff/JXXLrrk2 KO miceNo obvious bone alteration phenotypes[135]
B6.Cg-Tg(Lrrk2)6Yue/JXXLrrk2 overexpressing miceAutophagy suppression[136]
B6.FVB-Tg (LRRK2) WT1Mjfa/JXXLRRK2 overexpressing miceBehavioral hypoactivity
Altered dopamine-dependent short-term plasticity
[137]
STOCK Tg (tetO-LRRK2*G2019S) E3Cai/J XG2019S-LRRK2 overexpressing micePerturbed homeostasis
Altered neuronal morphogenesis
[138]
B6.Cg-Tg (Lrrk2*G2019S) 2Yue/J XG2019S-LRRK2 overexpressing miceReduction in lysosomal pH
Increased expression of lysosomal ATPases
[139]
B6.FVB-Tg (LRRK2*G2019S) 1Mjfa/JXXG2019S-LRRK2 overexpressing miceSynapsis gain-of-function effect of the G2019S
mutation
[140]
NTac:SD-Tg (LRRK2*G2019S) 571CJLiXXG2019S-Lrrk2 overexpressing ratsAltered bone marrow myelopoiesis
Peripheral myeloid cell differentiation
[141]
NTac:SD-Tg (LRRK2*G2019S) 571CJLiXXG2019S-Lrrk2 overexpressing ratsEnhanced α-syn gene-induced neurodegeneration[142]
K-14Cre-positive Gbalnl/lnl XGba KO mice (except in skin)Reduced cerebral vascularization[143]
PINK1B6.129S4-Pink1tm1Shn/J XPink1 KO micePink1 is not required for ubiquitination of mitochondrial proteins[144]
B6.129S4-Pink1tm1Shn/JXXPink1 KO miceReduced motor activity
Slower locomotor activity time
Absence of nigrostriatal dopamine loss
[145]
B6.129S4-Pink1tm1Shn/J XPink1 KO miceImpaired mitochondrial trafficking
Fragmented mitochondria
[146]
B6.129S4-Pink1tm1Shn/JXXPink1 KO miceHypersensitivity to MPTP-induced dopaminergic neuronal loss[147]
B6.129S4-Pink1tm1Shn/J XPink1 KO miceNo significant change in Ca2+ currents[148]
B6.129S4-Pink1tm1Shn/JXXPink1 KO micePathological cardiac hypertrophy
Greater levels of oxidative stress
Impaired mitochondrial function
[149]
B6.129S4-Pink1tm1Shn/JXXPink1 KO miceImpairments of corticostriatal LTP and LTD
Impaired dopamine release
[150]
n.s.n. XPink1 KO miceIntestinal infection triggers Parkinson’s disease-like symptoms[151]
Crossing B6;129-Pink1tm1Aub/J line with dOTC lineXXPink1 KO mice overexpressing OTC in DA neuronsEnhanced neurodegeneration in a model of mitochondrial stress[152]
B6.129S4-Pink1tm1Shn/J
B6.129S4-Prkntm1Shn/J
X-Pink1 KO mice
-Prkn KO mice
Enhanced sensitivity to group II mGlu receptor activation[153]
B6.129S4-Pink1tm1Shn/J
B6.129S4-Prkntm1Shn/J
X-Pink1 KO mice
-Prkn KO mice
Reduced mitochondria functions
Altered mitophagy in macrophages
[154]
FVB;129-Pink1tm1Aub Tg(Prnp-SNCA*A53T)AAub/J XA53T-SNCA overexpressing
Pink1 KO mice
Altered mitochondrial biogenesis[155]
FVB;129-Pink1tm1Aub Tg(Prnp-SNCA*A53T)AAub/JXXA53T-SNCA overexpressing
Pink1 KO mice
Exacerbated synucleinopathy[156]
FVB;129-Pink1tm1Aub Tg(Prnp-SNCA*A53T)AAub/JXXA53T-SNCA overexpressing
Pink1 KO mice
Potentiation of neurotoxicity[157]
Atad3afl/fl Mx1CrePink1 −/− mice, resulting from crossing B6.129S4-Pink1tm1Shn/J line with Atad3afl/fl Mx1Cre lineXXPink1 KO + conditional Atad3a KO miceAberrant stem-cell and progenitor homeostasis
Pink1-dependent mitophagy
[158]
B6N.129S6(Cg)-Atp13a2tm1Pjsch/JXXAtp13a2 KO miceAutophagy impairment
Reduced HDAC6 activity
[159]
B6N.129S6(Cg)-Atp13a2tm1Pjsch/J XAtp13a2 KO miceHarmful gliosis[160]
B6N.129S6(Cg)-Atp13a2tm1Pjsch/JXXAtp13a2 KO miceNeuronal ceroid lipofuscinosis
Limited α-syn accumulation
Sensorimotor deficits
[161]
n.s.n.: Non standard nomenclature.
A common factor in all cases is mitochondrial affectation. It should be noted that of all the genes devoted to results on transgenic animals described so far in this section, the most related to mitochondria biogenesis and function is Pink1.
Results related to DJ-1 (PARK7) were quite diverse perhaps due to the fact that some rodent models were available >15 years ago. There is also a Drosophila melanogaster model in which expression of a mutant gene led to motor dysfunction related to oxidative stress. DJ-1 knockout mice displayed oxidative stress and altered regulation of autophagy that may be the consequence of previous findings: mitochondrial dysfunction and increased reactive oxygen species (ROS) production and higher sensitivity to excitotoxicity. At the CNS level, the animals have less dendritic arborization and reduced number of dendritic spines in the medium spiny neurons of the striatum.
Although the GBA glucocerebrosidase gene is a pretty new player in PD, it has been extensively studied for its involvement in hereditary Gaucher’s disease, which is caused by glucocerebrosides in lysosomes. When assessing the neurological abnormalities of the GBA transgenic lines an acceleration of the onset of PD traits was observed (e.g., motor impairment, and a downregulation of some neurotrophic factors).
Results from transgenic lines affecting Uchl1, Atp13a2, Pla2g6, SYNJ1 and FBXO7 genes are limited although they are included in Supplementary Table S1. In all cases the disturbances observed in the transgenic lines overlapped with the traits described earlier in this section. For instance, F-box protein 7 participates with Parkin in mitophagy-associated processes [162] and VPS35 participates in the transport of vesicles from mitochondria to peroxisomes [163].

7. α-Synuclein: Main Character or Supporting Actor?

Of course, α-syn is an important factor in PD and in any other synucleinopathy. However, evidence from gene interaction studies indicates that there is no particular link with other genes related to familial PD. It should be noted the possibility that α-syn may be related to LRRK2. Although no direct evidence is yet available, it is suggested that that the kinase encoded by LRRK2, the physiological function of which is not known, could phosphorylate α-syn and such phosphorylation could have an impact in PD etiopathology (see [164,165]). If it were the cause of the disturbances leading, for instance, to dopaminergic denervation in PD, α-syn would be the main character. Many monogenic diseases appear early in childhood, but this is not the case in early onset PD caused by α-syn mutations. Therefore, it is likely that some α-syn mutants are aggravating the deterioration of mechanisms of homeostasis maintenance that occur with aging. These mechanisms would affect vesicular traffic, protein ubiquitination and protein processing/degradation, eventually leading to production of α-syn aggregates. It is necessary to point out that in familial forms of PD, aggregates occur for both mutated forms of α-syn, when the SNCA gene is affected and non-mutated α-syn, when the gene affected is another one of those in Table 1. One conclusion could be that α-syn is an important actor but not the main one. At the level of PD-related genes, there does not seem to be a main character because every gene product is necessary for the essential functions occurring in the dopaminergic neurons of the substantia nigra. Any mutation of those genes would lead to a gene product that would negatively affect cell function. Such an alteration never causes the disease to appear in the first years of life, thus suggesting that there is a progressive loss of function (failure to appropriately maintain homeostasis) consistent with the progressive character of the disease. Figure 3 displays two sides of an equation in which a mutant α-syn would affect mechanisms that, in turn, would lead to α-syn aggregation, and in which a mutant LRRK2, GBA, UCHL1, VPS35, PRKN, PINK1, ATP13A2, PLA2G6, DNAJC6, SYNJ1, DJ-1/PARK7 or FBXO7 genes would affect mechanisms that in time would affect the processing of nonmutated α-syn, thus leading to α-syn aggregation.
In agreement with the aberrant formation of α-syn aggregates, the vesicular transport or the proteasomal degradation mechanism is defective or overexposed. Although the neurotransmitters(s) involved are not known, the association of some PD-related genes to endocytosis indicates that signal transduction may regulate the processing of α-syn. Could dopaminergic signaling be involved? This is a possibility because of the unique vulnerability of the nigral neurons in PD.
Mitochondria are linked to all of the events mentioned above, either through mitogenesis and mitochondria-derived vesicles or through the provision of energy. In fact, a substantial amount of protein processing, from synthesis to ubiquitination-directed degradation, requires energy, primarily in the form of ATP. Energy is also required for vesicle/endosome sorting and trafficking. Neurons rely primarily on the breakdown of glucose to meet energy requirements, and anaerobic glycolysis would not be sufficient to support all neuronal processes. Therefore, complete oxidation of glucose is required since ATP is obtained mainly through the Krebs’ cycle and the electron-transport chain. As shown in Figure 3, mitochondria are center stage in all events involving the PD-related gene products. In fact, there are several studies linking proteins of Table 2 to mitochondrial action. One example from data obtained in Drosophila melanogaster demonstrated that loss-of-function PINK1 results in motor deficits due to neurodegeneration mediated by mitochondrial dysfunction. The authors concluded: “our genetic evidence clearly establishes that Parkin and PINK1 act in a common pathway in maintaining mitochondrial integrity and function in both muscles and dopaminergic neurons” [41]. Similar results were more recently obtained in a cell model and in primary cells obtained from patients. Loss-of-function of PINK1 in HeLa cells leads to altered mitochondrion function and morphology. Cells from patients with mutated PINK1 show altered mitochondrial morphology [42]. Another example is provided by [166], who showed that endoplasmic reticulum-mitochondrion interactions are regulated by LRRK2 and by PERK products via protein ubiquitination pathways. It is worth mentioning the MitoPark mice, in which a mutated gene for a mitochondrial transcription factor in dopaminergic neurons led to the defective expression of proteins encoded by mitochondrial DNA and a phenotype consistent with parkinsonism, including motor deficits and progressive neurodegeneration [167]. Because the MitoPark mice exhibited deficits in the electron transport chain, it raised the question of why nigral dopaminergic neurons or their mitochondria were more vulnerable than other neurons. A differential expression of α-syn in different neuronal populations may be a reasonable hypothesis. However, in our opinion, the dopamine metabolism should be taken into account because it adds extra oxidative stress to dopaminergic neurons. Indeed, dopamine leads to the production of a quinone, aminochrome (the precursor of neuromelanin), which significantly contributes to the dark color of the substantia nigra in humans. The reaction of dopamine with molecular oxygen can lead to other quinones and free radicals, an oxidative load that alters mitochondrial function and can produce autophagy in dopaminergic neurons [168,169].

8. Concluding Remarks

Information available from genes related to familial PD led to a scenario that may be useful for explaining neurodegeneration in sporadic cases. Apart from a potential link between the kinase coded by LRRK2 and the phosphorylation of α-Syn, which is not yet substantiated, there is no direct relationship between the gene for this protein (α-Syn) and all the other genes displayed in Table 1. Hence, a common factor in familial and sporadic cases may be a progressive loss of efficacy in the mechanisms of vesicle traffic and protein handling, especially in degradation by the proteasome and the lysosome. The loss is quicker in familial cases as there is an excess burden due to difficulties in processing mutant forms of α-Syn or to difficulties in processing α-Syn by mutant components of the protein-processing machinery. For those processes, neuronal energy in the form of ATP is dependent of the full oxidation of glucose (i.e., involvement of mitochondrial Krebs’s cycle and electron chain transport). The higher vulnerability of dopaminergic neurons may come from the need of appropriate α-Syn processing and also from the oxidative stress produced by dopamine metabolism. It is reasonable to speculate that mitochondria copes with the supply of additional energy and with oxidative stress to the point where the demands for energy and antioxidant actions can no longer be satisfied, and cell death occurs: earlier in familial cases but later in sporadic cases. Hence, it can be speculated that any action to help mitochondria would be useful for preventing neurodegeneration. In general, to improve mitochondrial performance, coenzyme Q is useful; in fact, there is a correlation between oxidative stress and the deficit of this molecule in centenarians [170,171,172]. However, better options for improving the antioxidant machinery in the central nervous system are needed. The discovery of G protein-coupled receptors (GPCRs) in the mitochondria of neurons [173,174,175] may end up helping target those receptors to reduce oxidative burden or increase mitochondrial performance. Drugs acting on GPCRs constitute 35–45% of all approved therapeutic drugs and antagonists of receptors that are targets of neuroprotection (e.g., the adenosine A2A receptor) may help. Could it be that the decreased risk of suffering from neurodegenerative diseases after consuming the adenosine-receptor antagonists caffeine (coffee and cola drinks) and theophylline (tea) [8,176,177,178,179,180,181,182,183,184,185,186,187,188,189] is due to an effect on neuronal mitochondria?

Supplementary Materials

The following are available online at https://www.mdpi.com/article/10.3390/ijms22094643/s1, Figure S1: STRING analysis of connections of either SNCA (A), GBA (B), FBXO7 (C), ATP13A2 (D) and UCHL1 (E). Table S1: Findings in animal models related to Gba, Uchl1, Vps35, Atp13a2, Pla2g6, Dnajc6, Synj1, DJ-1/Park7 and Fbxo7.

Author Contributions

R.F. and A.P. designed the paper. R.F., R.R.-S., G.N., A.P. and I.R.-R. retrieved information for the literature, selected the papers and summarized their data. The tables were constructed by A.P., I.R.-R. and R.R.-S. The figures were constructed by R.F. and R.R.-S., R.F. and G.N. wrote the first draft. All co-authors contributed equally to the paper. All authors have read and agreed to the published version of the manuscript.

Funding

This research was supported by a grant (#RTI2018-09204-B-I00) from the Spanish Ministerio de Ciencia, Innovación y Universidades (MCIU) and Spanish Agencia Estatal de Investigación (AEI); it includes UE FEDER funds.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

Work supported by a grant (#RTI2018-09204-B-I00) from the Spanish Ministerio de Ciencia, Innovación y Universidades (MCIU) and Spanish Agencia Estatal de Investigación (AEI); it includes UE FEDER funds. The research group of the University of Barcelona is considered of excellence (grup consolidat #2017 SGR 1497) by the Regional Catalonian Government, which does not provide any specific funding for reagents or for payment of services or Open Access fees).

Conflicts of Interest

Authors declare no conflict of interests.

References

  1. Lesage, S.; Lunati, A.; Houot, M.; Ben Romdhan, S.; Clot, F.; Tesson, C.; Mangone, G.; Toullec, B.L.; Courtin, T.; Larcher, K.; et al. Characterization of Recessive Parkinson Disease in a Large Multicenter Study. Ann. Neurol. 2020, 88, 843–850. [Google Scholar] [CrossRef] [PubMed]
  2. Deng, H.; Wang, P.; Jankovic, J. The genetics of Parkinson disease. Ageing Res. Rev. 2018, 42, 72–85. [Google Scholar] [CrossRef] [PubMed]
  3. Emamzadeh, F.N.; Surguchov, A. Parkinson’s disease: Biomarkers, treatment, and risk factors. Front. Neurosci. 2018, 12, 612. [Google Scholar] [CrossRef] [PubMed]
  4. Oñatibia-Astibia, A.; Franco, R.; Martínez-Pinilla, E. Health benefits of methylxanthines in neurodegenerative diseases. Mol. Nutr. Food Res. 2017, 61, 1600670. [Google Scholar] [CrossRef] [PubMed]
  5. Simon, D.K.; Wu, C.; Tilley, B.C.; Lohmann, K.; Klein, C.; Payami, H.; Wills, A.M.; Aminoff, M.J.; Bainbridge, J.; Dewey, R.; et al. Caffeine, creatine, GRIN2A and Parkinson’s disease progression. J. Neurol. Sci. 2017, 375, 355–359. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Sipetic, S.B.; Vlajinac, H.D.; Maksimovic, J.M.; Marinkovic, J.M.; Dzoljic, E.D.; Ratkov, I.S.; Kostic, V.S. Cigarette smoking, coffee intake and alcohol consumption preceding Parkinson’s disease: A case-control study. Acta Neuropsychiatr. 2012, 24, 109–114. [Google Scholar] [CrossRef] [PubMed]
  7. Ross, G.W.; Petrovitch, H. Current Evidence for Neuroprotective Effects of Nicotine and Caffeine Against Parkinsons Disease. Drugs Aging 2001, 18, 797–806. [Google Scholar] [CrossRef]
  8. Ross, G.W.; RD, A.; Petrovitch, H.; Al, E.; Abbott, R.D.; Petrovitch, H.; Morens, D.M.; Grandinetti, A.; Tung, K.H.; Tanner, C.M.; et al. Association of coffee and caffeine intake with the risk of parkinson disease. JAMA 2000, 283, 2674–2679. [Google Scholar] [CrossRef]
  9. Jiménez-Jiménez, F.J.; Mateo, D.; Giméanez-Roldan, S. Premorbid smoking, alcohol consumption, and coffee drinking habits in Parkinson’s disease: A case-control study. Mov. Disord. 1992, 7, 339–344. [Google Scholar] [CrossRef]
  10. Bronstein, J.; Carvey, P.; Chen, H.; Cory-Slechta, D.; DiMonte, D.; Duda, J.; English, P.; Goldman, S.; Grate, S.; Hanssen, J.; et al. Meeting report: Consensus Statement—Parkinson’s disease and the environment: Collaborative on health and the environment and Parkinson’s action network (CHE PAN) conference 26–28 June 2007. Environ. Health Perspect. 2009, 117, 117–121. [Google Scholar] [CrossRef]
  11. Chen, J.-F. Adenosine receptor control of cognition in normal and disease. Int. Rev. Neurobiol. 2014, 119, 257–307. [Google Scholar] [PubMed]
  12. Elbaz, A.; Carcaillon, L.; Kab, S.; Moisan, F. Epidemiology of Parkinson’s disease. Rev. Neurol. 2016, 172, 14–26. [Google Scholar] [CrossRef] [PubMed]
  13. Blauwendraat, C.; Nalls, M.A.; Singleton, A.B. The genetic architecture of Parkinson’s disease. Lancet Neurol. 2020, 19, 170–178. [Google Scholar] [CrossRef]
  14. Birkmayer, W.; Hornykiewicz, O. The L-dihydroxyphenylalanine (L-DOPA) effect in Parkinson’s syndrome in man: On the pathogenesis and treatment of Parkinson akinesis. Arch. Psychiatr. Nervenkr. Z. Gesamte Neurol. Psychiatr. 1962, 203, 560–574. [Google Scholar] [CrossRef]
  15. Hornykiewicz, O. Die topische Lokalization und des Verhalten von Noradrenalin und Dopamin (3-Hydroxytyramin) in der Substantia nigra der normalen und Parkinson Kranken Menschen. Wien. Klin. Wochschr. 1963, 75, 309–312. [Google Scholar]
  16. Birkmayer, W.; Hornykiewicz, O. Additional experimental studies on L-DOPA in Parkinson’s syndrome and reserpine parkinsonism. Arch. Psychiatr. Nervenkr. 1964, 206, 367–381. [Google Scholar] [CrossRef]
  17. Holzer, G.; Hornykiewicz, O. Über den Dopamin-(Hydroxytyramin-)Stoffwechsel im Gehirn der Ratte. Naunyn-Schmiedeberg’s Arch. Exp. Pathol. Pharmakol. 1959, 237, 27–33. [Google Scholar]
  18. Hornykiewicz, O. The discovery of dopamine deficiency in the parkinsonian brain. J. Neural Transm. Suppl. 2006, 70, 9–15. [Google Scholar]
  19. Olanow, C.W.; Agid, Y.; Mizuno, Y.; Albanese, A.; Bonuccelli, U.; Bonucelli, U.; Damier, P.; De Yebenes, J.; Gershanik, O.; Guttman, M.; et al. Levodopa in the treatment of Parkinson’s disease: Current controversies. Mov. Disord. 2004, 19, 997–1005. [Google Scholar] [CrossRef]
  20. Walter, B.L.; Vitek, J.L. Surgical treatment for Parkinson’s disease. Lancet Neurol. 2004, 3, 719–728. [Google Scholar] [CrossRef]
  21. Conley, S.C.; Kirchner, J.T. Medical and surgical treatment of Parkinson’s disease: Strategies to slow symptom progression and improve quality of life. Postgrad. Med. 1999, 106, 41–52. [Google Scholar] [CrossRef] [PubMed]
  22. Durif, F. Treating and preventing levodopa-induced dyskinesias: Current and future strategies. Drugs Aging 1999, 14, 337–345. [Google Scholar] [CrossRef] [PubMed]
  23. Kumar, R.; Lozano, A.M.; Montgomery, E.; Lang, A.E. Pallidotomy and deep brain stimulation of the pallidum and subthalamic nucleus in advanced Parkinson’s disease. Mov. Disord. 1998, 13 (Suppl. 1), 73–82. [Google Scholar]
  24. Arle, J.E.; Alterman, R.L. Surgical options in Parkinson’s disease. Med. Clin. N. Am. 1999, 83, 483–498. [Google Scholar] [CrossRef]
  25. Limousin-Dowsey, P.; Pollak, P.; Blercom, N.; Krack, P.; Benazzouz, A.; Benabid, A.-L. Thalamic, subthalamic nucleus and internal pallidum stimulation in Parkinson’s disease. J. Neurol. 1999, 246, II42–II45. [Google Scholar] [CrossRef] [Green Version]
  26. Gross, C.E.; Boraud, T.; Guehl, D.; Bioulac, B.; Bezard, E. From experimentation to the surgical treatment of Parkinson’s disease: Prelude or suite in basal ganglia research? Prog. Neurobiol. 1999, 59, 509–532. [Google Scholar] [CrossRef]
  27. Gross, R.E.; Lozano, A.M. Advances in neurostimulation for movement disorders. Neurol. Res. 2000, 22, 247–258. [Google Scholar] [CrossRef]
  28. Caparros-Lefebvre, D.; Blond, S.; Vermersch, P.; Pecheux, N.; Guieu, J.D.; Petit, H. Chronic thalamic stimulation improves tremor and levodopa induced dyskinesias in Parkinson’s disease. J. Neurol. Neurosurg. Psychiatry 1993, 56, 268–273. [Google Scholar] [CrossRef] [PubMed]
  29. Goedert, M.; Spillantini, M.G.; Del Tredici, K.; Braak, H. 100 years of Lewy pathology. Nat. Rev. Neurol. 2012, 9, 13–24. [Google Scholar] [CrossRef] [PubMed]
  30. McKeith, I.G.; Burn, D. Spectrum of Parkinson’s disease, Parkinson’s dementia, and Lewy body dementia. Neurol. Clin. 2000, 18, 865–883. [Google Scholar] [CrossRef]
  31. McCann, H.; Stevens, C.H.; Cartwright, H.; Halliday, G.M. α-Synucleinopathy phenotypes. Park. Relat. Disord. 2014, 20, S62–S67. [Google Scholar] [CrossRef] [Green Version]
  32. Smeyne, R.J.; Noyce, A.J.; Byrne, M.; Savica, R.; Marras, C. Infection and risk of Parkinson’s disease. J. Parkinsons. Dis. 2021, 11, 31–43. [Google Scholar] [CrossRef] [PubMed]
  33. Lunati, A.; Lesage, S.; Brice, A. The genetic landscape of Parkinson’s disease. Rev. Neurol. 2018, 174, 628–643. [Google Scholar] [CrossRef] [PubMed]
  34. Puschmann, A. New Genes Causing Hereditary Parkinson’s Disease or Parkinsonism. Curr. Neurol. Neurosci. Rep. 2017, 17, 1–11. [Google Scholar] [CrossRef] [Green Version]
  35. Haas, A.L.; Warms, J.V.; Hershko, A.; Rose, I.A. Ubiquitin-activating enzyme. Mechanism and role in protein-ubiquitin conjugation. J. Biol. Chem. 1982, 257, 2543–2548. [Google Scholar] [CrossRef]
  36. Ciechanover, A.; Finley, D.; Varshavsky, A. The ubiquitin-mediated proteolytic pathway and mechanisms of energy-dependent intracellular protein degradation. J. Cell. Biochem. 1984, 24, 27–53. [Google Scholar] [CrossRef]
  37. Hershko, A.; Leshinsky, E.; Ganoth, D.; Heller, H. ATP-dependent degradation of ubiquitin-protein conjugates. Proc. Natl. Acad. Sci. USA 1984, 81, 1619–1623. [Google Scholar] [CrossRef] [Green Version]
  38. Bonifati, V.; Dekker, M.C.J.; Vanacore, N.; Fabbrini, G.; Squitieri, F.; Marconi, R.; Antonini, A.; Brustenghi, P.; Dalla Libera, A.; De Mari, M.; et al. Autosomal recessive early onset parkinsonism is linked to three loci: PARK2, PARK6, and PARK. Neurol. Sci. 2002, 23, 59–60. [Google Scholar] [CrossRef] [PubMed]
  39. Moore, D.J.; Zhang, L.; Troncoso, J.; Lee, M.K.; Hattori, N.; Mizuno, Y.; Dawson, T.M.; Dawson, V.L. Association of DJ-1 and parkin mediated by pathogenic DJ-1 mutations and oxidative stress. Hum. Mol. Genet. 2005, 14, 71–84. [Google Scholar] [CrossRef] [Green Version]
  40. Clark, I.E.; Dodson, M.W.; Jiang, C.; Cao, J.H.; Huh, J.R.; Seol, J.H.; Yoo, S.J.; Hay, B.A.; Guo, M. Drosophila pink1 is required for mitochondrial function and interacts genetically with parkin. Nature 2006, 441, 1162–1166. [Google Scholar] [CrossRef] [PubMed]
  41. Park, J.; Lee, S.B.; Lee, S.; Kim, Y.; Song, S.; Kim, S.; Bae, E.; Kim, J.; Shong, M.; Kim, J.M.; et al. Mitochondrial dysfunction in Drosophila PINK1 mutants is complemented by parkin. Nature 2006, 441, 1157–1161. [Google Scholar] [CrossRef]
  42. Exner, N.; Treske, B.; Paquet, D.; Holmström, K.; Schiesling, C.; Gispert, S.; Carballo-Carbajal, I.; Berg, D.; Hoepken, H.H.; Gasser, T.; et al. Loss-of-function of human PINK1 results in mitochondrial pathology and can be rescued by parkin. J. Neurosci. 2007, 27, 12413–12418. [Google Scholar] [CrossRef] [Green Version]
  43. Smith, W.W.; Pei, Z.; Jiang, H.; Moore, D.J.; Liang, Y.; West, A.B.; Dawson, V.L.; Dawson, T.M.; Ross, C.A. Leucine-rich repeat kinase 2 (LRRK2) interacts with parkin, and mutant LRRK2 induces neuronal degeneration. Proc. Natl. Acad. Sci. USA 2005, 102, 18676–18681. [Google Scholar] [CrossRef] [Green Version]
  44. Neuspiel, M.; Schauss, A.C.; Braschi, E.; Zunino, R.; Rippstein, P.; Rachubinski, R.A.; Andrade-Navarro, M.A.; McBride, H.M. Cargo-Selected Transport from the Mitochondria to Peroxisomes Is Mediated by Vesicular Carriers. Curr. Biol. 2008, 18, 102–108. [Google Scholar] [CrossRef] [Green Version]
  45. Lee, D.W.; Zhao, X.; Yim, Y.I.; Eisenberg, E.; Greene, L.E. Essential role of cyclin-G-associated kinase (auxilin-2) in developing and mature mice. Mol. Biol. Cell 2008, 19, 2766–2776. [Google Scholar] [CrossRef] [Green Version]
  46. Pishvaee, B.; Costaguta, G.; Yeung, B.G.; Ryazantsev, S.; Greener, T.; Greene, L.E.; Eisenberg, E.; McCaffery, J.M.; Payne, G.S. A yeast DNA J protein required for uncoating of clathrin-coated vesicles in vivo. Nat. Cell Biol. 2000, 2, 958–963. [Google Scholar] [CrossRef] [PubMed]
  47. Toret, C.P.; Lee, L.; Sekiya-Kawasaki, M.; Drubin, D.G. Multiple pathways regulate endocytic coat disassembly in Saccharomyces cerevisiae for optimal downstream trafficking. Traffic 2008, 9, 848–859. [Google Scholar] [CrossRef] [PubMed]
  48. Surguchev, A.A.; Surguchov, A. Synucleins and gene expression: Ramblers in a crowd or cops regulating traffic? Front. Mol. Neurosci. 2017, 10, 224. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Austin, S.A.; Floden, A.M.; Murphy, E.J.; Combs, C.K. α-Synuclein Expression Modulates Microglial Activation Phenotype. J. Neurosci. 2006, 26, 10558–10563. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  50. Golovko, M.Y.; Faergeman, N.J.; Cole, N.B.; Castagnet, P.I.; Nussbaum, R.L.; Murphy, E.J. α-Synuclein Gene Deletion Decreases Brain Palmitate Uptake and Alters the Palmitate Metabolism in the Absence of R -Synuclein Palmitate Binding. Biochemistry 2005, 44, 8251–8259. [Google Scholar] [CrossRef]
  51. Ellis, C.E.; Murphy, E.J.; Mitchell, D.C.; Golovko, M.Y.; Scaglia, F.; Barcelo, G.C.; Nussbaum, R.L. Mitochondrial Lipid Abnormality and Electron Transport Chain Impairment in Mice Lacking α-Synuclein. Mol. Cell. Biol. 2005, 25, 10190–10201. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Cabin, D.E.; Shimazu, K.; Murphy, D.; Cole, N.B.; Gottschalk, W.; Mcilwain, K.L.; Orrison, B.; Chen, A.; Ellis, C.E.; Paylor, R.; et al. Synaptic Vesicle Depletion Correlates with Attenuated Synaptic α-Synuclein. J. Neurosci. 2002, 22, 8797–8807. [Google Scholar] [CrossRef] [Green Version]
  53. Pathak, D.; Berthet, A.; Bendor, J.T.; Yu, K.; Sellnow, R.C.; Orr, L.; Nguyen, M.K.; Edwards, R.H.; Manfredsson, F.P.; Nakamura, K. Loss of α-Synuclein Does Not Affect Mitochondrial Bioenergetics in Rodent Neurons. Eneuro 2017, 4. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Beatman, E.L.; Massey, A.; Shives, K.D.; Burrack, K.S.; Chamanian, M.; Morrison, T.E. α-Synuclein Expression Restricts RNA Viral Infections in the Brain. J. Virol. 2016, 90, 2767–2782. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Renella, R.; Schlehe, J.S.; Selkoe, D.J.; Williams, D.A.; LaVoie, M.J. Genetic deletion of the GATA1-regulated protein α-synuclein reduces oxidative stress and nitric oxide synthase levels in mature erythrocytes. Am. J. Hematol. 2014, 89, 974–977. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Paine, S.M.L.; Anderson, G.; Bedford, K.; Lawler, K.; Mayer, R.J.; Lowe, J.; Bedford, L. Pale Body-Like Inclusion Formation and Neurodegeneration following Depletion of 26S Proteasomes in Mouse Brain Neurones are Independent of α-Synuclein. PLoS ONE 2013, 8, e54711. [Google Scholar] [CrossRef] [PubMed]
  57. Scott, D.; Roy, S. α-Synuclein Inhibits Intersynaptic Vesicle Mobility and Maintains Recycling-Pool Homeostasis. J. Neurosci. 2012, 32, 10129–10135. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Kokhan, V.S.; Afanasyeva, M.A.; Van, G.I. α-Synuclein knockout mice have cognitive impairments. Behav. Brain Res. 2012, 231, 226–230. [Google Scholar] [CrossRef]
  59. Cano-jaimez, M.; Pérez-sánchez, F.; Milán, M.; Buendía, P.; Ambrosio, S.; Fariñas, I. Neurobiology of Disease Vulnerability of peripheral catecholaminergic neurons to MPTP is not regulated by α-synuclein. Neurobiol. Dis. 2010, 38, 92–103. [Google Scholar] [CrossRef]
  60. Klivenyi, P.; Siwek, D.; Gardian, G.; Yang, L.; Starkov, A.; Cleren, C.; Ferrante, R.J.; Kowall, N.W.; Abeliovich, A.; Beal, M.F. Mice lacking α-synuclein are resistant to mitochondrial toxins. Neurobiol. Dis. 2006, 21, 541–548. [Google Scholar] [CrossRef]
  61. Abeliovich, A.; Schmitz, Y.; Farin, I.; Choi-lundberg, D.; Ho, W.; Castillo, P.E.; Shinsky, N.; Manuel, J.; Verdugo, G.; Armanini, M.; et al. Mice Lacking α-Synuclein Display Functional Deficits in the Nigrostriatal Dopamine System. Neuron 2000, 25, 239–252. [Google Scholar] [CrossRef] [Green Version]
  62. Peña-Oliver, Y.; Buchman, V.L.; Dalley, J.W.; Robbins, T.W.; Schumann, G.; Ripley, T.L.; King, S.L.; Stephens, D.N. Deletion of α-synuclein decreases impulsivity in mice. Genes Brain Behav. 2012, 11, 137–146. [Google Scholar] [CrossRef] [PubMed]
  63. Pelkonen, A.; Yavich, L. Neuroscience Letters Neuromuscular pathology in mice lacking α-synuclein. Neurosci. Lett. 2011, 487, 350–353. [Google Scholar] [CrossRef]
  64. Chadchankar, H.; Ihalainen, J.; Tanila, H.; Yavich, L. Decreased reuptake of dopamine in the dorsal striatum in the absence of α-synuclein. Brain Res. 2011, 1382, 37–44. [Google Scholar] [CrossRef] [PubMed]
  65. Al-Wandi, A.; Ninkina, N.; Millership, S.; Williamson, S.J.M.; Jones, P.A.; Buchman, V.L. Absence of α-synuclein affects dopamine metabolism and synaptic markers in the striatum of aging mice. Neurobiol. Aging 2010, 31, 796–804. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Senior, S.L.; Ninkina, N.; Deacon, R.; Bannerman, D.; Vladimir, L.; Cragg, S.J.; Wade-martins, R. Europe PMC Funders Group Increased striatal dopamine release and hyperdopaminergic-like behaviour in mice lacking both α-synuclein and gamma- synuclein. J. Neurosci. 2011, 27, 947–957. [Google Scholar]
  67. Anwar, S.; Peters, O.; Millership, S.; Ninkina, N.; Doig, N.; Connor-robson, N.; Threlfell, S.; Kooner, G.; Deacon, R.M.; Bannerman, D.M.; et al. Functional Alterations to the Nigrostriatal System in Mice Lacking All Three Members of the Synuclein Family. J. Neurosci. 2011, 31, 7264–7274. [Google Scholar] [CrossRef] [PubMed]
  68. Vargas, K.J.; Makani, S.; Davis, T.; Westphal, C.H.; Castillo, P.E.; Chandra, S.S. Synucleins Regulate the Kinetics of Synaptic Vesicle Endocytosis. J. Neurosci. 2014, 34, 9364–9376. [Google Scholar] [CrossRef]
  69. Greten-Harrison, B.; Polydoro, M.; Morimoto-Tomita, M.; Diao, L.; Williams, A.M. αβγ -Synuclein triple knockout mice reveal age-dependent neuronal dysfunction. Proc. Natl. Acad. Sci. USA 2010, 107, 19573–19578. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  70. Vargas, K.J.; Schrod, N.; Davis, T.; Fernandez-Busnadiego, R.; Taguchi, Y.V.; Laugks, U.; Lucic, V.; Chandra, S.S. Synucleins Have Multiple Effects on Presynaptic Architecture. Cell Rep. 2017, 18, 161–173. [Google Scholar] [CrossRef]
  71. Su, S.; Zhang, Z.; Liu, X.; Manfredsson, F.P.; Benskey, M.J.; Cao, X.; Xu, J. TrkB neurotrophic activities are blocked by α-synuclein, triggering dopaminergic cell death in Parkinson’s disease. Proc. Natl. Acad. Sci. USA 2017, 114, 10773–10778. [Google Scholar]
  72. Sarafian, T.A.; Ryan, C.M.; Souda, P.; Masliah, E.; Kar, U.K.; Vinters, H.V.; Mathern, G.W.; Faull, K.F.; Whitelegge, J.P.; Watson, J.B. Impairment of Mitochondria in Adult Mouse Brain Overexpressing Predominantly Full-Length, N-Terminally Acetylated Human α-Synuclein. PLoS ONE 2013, 8, e63557. [Google Scholar] [CrossRef] [PubMed]
  73. Watson, J.B.; Hatami, A.; David, H.; Masliah, E.; Roberts, K.; Evans, C.E.; Levine, M.S. Aleterations in corticostriatal synaptic plasticity in mice overexpressing human α-synuclein. NSC 2009, 159, 501–513. [Google Scholar]
  74. Reznichenko, L.; Cheng, Q.; Nizar, K.; Gratiy, S.L.; Saisan, P.A.; Rockenstein, E.M.; Patrick, C.; Spencer, B.; Desplats, P.; Dale, A.M.; et al. In Vivo Alterations in Calcium Buffering Capacity in Transgenic Mouse Model of Synucleinopathy. J. Neurosci. 2012, 32, 9992–9998. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Siebert, H.; Kahle, P.J.; Kramer, M.L.; Isik, T.; Schlüter, O.M.; Schulz-Schaeffer, W.J.; Brück, W. Over-expression of α-synuclein in the nervous system enhances axonal degeneration after peripheral nerve lesion in a transgenic mouse strain. J. Neurochem. 2010, 114, 1007–1018. [Google Scholar] [CrossRef] [PubMed]
  76. Janezic, S.; Threlfell, S.; Dodson, P.D.; Dowie, M.J.; Taylor, T.N.; Potgieter, D.; Parkkinen, L.; Senior, S.L.; Anwar, S.; Ryan, B.; et al. Deficits in dopaminergic transmission precede neuron loss and dysfunction in a new Parkinson model. Proc. Natl. Acad. Sci. USA 2013, 110, E4016–E4025. [Google Scholar] [CrossRef] [Green Version]
  77. Hunn, B.H.M.; Vingill, S.; Threlfell, S.; Bannerman, D.M.; Cragg, S.J.; Wade-martins, R.; Hunn, B.H.M.; Vingill, S.; Threlfell, S.; Alegre-abarrategui, J.; et al. Impairment of Macroautophagy in Dopamine Neurons Has Opposing Effects on Parkinsonian Pathology and Behavior Article Impairment of Macroautophagy in Dopamine Neurons Has Opposing Effects on Parkinsonian Pathology and Behavior. Cell Rep. 2019, 29, 920–931.e7. [Google Scholar] [CrossRef] [Green Version]
  78. Wang, L.; Das, U.; Scott, D.A.; Tang, Y.; McLean, P.J.; Roy, S. α-Synuclein multimers cluster synaptic vesicles and attenuate recycling. Curr. Biol. 2014, 24, 2319–2326. [Google Scholar] [CrossRef] [Green Version]
  79. Escobar, V.D.; Kuo, Y.-M.; Orrison, B.M.; Giasson, B.I.; Nussbaum, R.L. Transgenic mice expressing S129 phosphorylation mutations in α-synuclein. Neurosci. Lett. 2014, 563, 96–100. [Google Scholar] [CrossRef] [Green Version]
  80. Kuo, Y.; Li, Z.; Jiao, Y.; Gaborit, N.; Pani, A.K.; Orrison, B.M.; Bruneau, B.G.; Giasson, B.I.; Smeyne, R.J.; Gershon, M.D.; et al. Extensive enteric nervous system abnormalities in mice transgenic for artificial chromosomes containing Parkinson disease-associated α-synuclein gene mutations precede central nervous system changes. Hum. Mol. Genet. 2010, 19, 1633–1650. [Google Scholar] [CrossRef]
  81. Gao, H.; Kotzbauer, P.T.; Uryu, K.; Leight, S.; Trojanowski, J.Q.; Lee, V.M. Neuroinflammation and Oxidation / Nitration of α-Synuclein Linked to Dopaminergic Neurodegeneration. J. Neurosci. 2008, 28, 7687–7698. [Google Scholar] [CrossRef]
  82. Taylor, T.N.; Potgieter, D.; Anwar, S.; Senior, S.L.; Janezic, S.; Threlfell, S.; Ryan, B.; Parkkinen, L.; Deltheil, T.; Cioroch, M.; et al. Region-specific deficits in dopamine, but not norepinephrine, signaling in a novel A30P α-synuclein BAC transgenic mouse. Neurobiol. Dis. 2014, 62, 193–207. [Google Scholar] [CrossRef] [Green Version]
  83. Kurz, A.; Double, K.L.; Lastres-becker, I.; Tozzi, A.; Tantucci, M.; Nuber, S.; Schlaudraff, F.; Bockhart, V.; Bonin, M.; Ferna, J. A53T-α-Synuclein Overexpression Impairs Dopamine Signaling and Striatal Synaptic Plasticity in Old Mice. PLoS ONE 2010, 5, e11464. [Google Scholar] [CrossRef]
  84. Gispert, S.; Turco, D.; Garrett, L.; Chen, A.; Bernard, D.J.; Hamm-clement, J.; Korf, H.; Deller, T.; Braak, H.; Auburger, G.; et al. Transgenic mice expressing mutant A53T human α-synuclein show neuronal dysfunction in the absence of aggregate formation. Mol. Cell. Neurosci. 2003, 24, 419–429. [Google Scholar] [CrossRef]
  85. Yamasaki, T.; Fujinaga, M.; Kawamura, K.; Furutsuka, K.; Nengaki, N.; Shimoda, Y.; Shiomi, S.; Takei, M.; Hashimoto, H.; Yui, J.; et al. Dynamic Changes in Striatal mGluR1 But Not mGluR5 during Pathological Progression of Parkinson’s Disease in Human Alpha-Synuclein A53T Transgenic Rats: A Multi-PET Imaging Study. J. Neurosci. 2016, 36, 375–384. [Google Scholar] [CrossRef] [Green Version]
  86. Luk, K.C.; Kehm, V.M.; Zhang, B.; Brien, P.O.; Trojanowski, J.Q.; Lee, V.M.Y. Intracerebral inoculation of pathological α-synuclein initiates a rapidly progressive neurodegenerative α-synucleinopathy in mice. J. Exp. Med. 2012, 209, 975–986. [Google Scholar] [CrossRef] [Green Version]
  87. Cannon, J.R.; Geghman, K.D.; Tapias, V.; Sew, T.; Dail, M.K.; Li, C.; Greenamyre, J.T. Expression of human E46K-mutated α-synuclein in BAC-transgenic rats replicates early-stage Parkinson’s disease features and enhances vulnerability to mitochondrial impairment. Exp. Neurol. 2013, 240, 44–56. [Google Scholar] [CrossRef] [Green Version]
  88. Shimshek, D.R.; Schweizer, T.; Schmid, P.; Van Der Putten, P.H. Excess α-synuclein worsens disease in mice lacking ubiquitin carboxy-terminal hydrolase L1. Sci. Rep. 2012, 2, 1–8. [Google Scholar] [CrossRef] [PubMed]
  89. Martín-Clemente, B.; Alvarez-Castelao, B.; Mayo, I.; Sierra, A.B.; Díaz, V.; Milán, M.; Fariñas, I.; Gómez-Isla, T.; Ferrer, I.; Castaño, J.G. α-Synuclein expression levels do not significantly affect proteasome function and expression in mice and stably transfected PC12 cell lines. J. Biol. Chem. 2004, 279, 52984–52990. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  90. Sharon, R.; Bar-joseph, I.; Mirick, G.E.; Serhan, C.N.; Selkoe, D.J. Altered Fatty Acid Composition of Dopaminergic Neurons Expressing α-Synuclein and Human Brains with α-Synucleinopathies. J. Biol. Chem. 2003, 278, 49874–49881. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  91. Nemani, V.M.; Lu, W.; Berge, V.; Nakamura, K.; Onoa, B.; Lee, M.K.; Chaudhry, F.A.; Nicoll, R.A.; Edwards, R.H. Increased Expression of α-Synuclein Reduces Neurotransmitter Release by Inhibiting Synaptic Vesicle Reclustering after Endocytosis. Neuron 2010, 65, 66–79. [Google Scholar] [CrossRef] [Green Version]
  92. Kim, S.; Kwon, S.; Kam, T.; Dawson, V.L.; Dawson, T.M.; Ko, H.S.; Kim, S.; Kwon, S.; Kam, T.; Panicker, N.; et al. Transneuronal Propagation of Pathologic a -Synuclein from the Gut to the Brain Models Parkinson’ s Disease Article Transneuronal Propagation of Pathologic α-Synuclein from the Gut to the Brain Models Parkinson’ s Disease. Neuron 2019, 103, 627–641.e7. [Google Scholar] [CrossRef]
  93. Peng, C.; Gathagan, R.J.; Covell, D.J.; Medellin, C.; Stieber, A.; Robinson, J.L.; Zhang, B.; Pitkin, R.M.; Olufemi, M.F.; Luk, K.C.; et al. α -synuclein strains in α -synucleinopathies. Nature 2018, 557, 558–563. [Google Scholar] [CrossRef]
  94. Lin, J.; Zheng, X.; Zhang, Z.; Zhuge, J.; Shao, Z.; Huang, C.; Jin, J.; Chen, X.; Chen, Y.; Wu, Y.; et al. Inhibition of LRRK2 restores parkin-mediated mitophagy and attenuates intervertebral disc degeneration. Osteoarthr. Cartil. 2021, 29, 579–591. [Google Scholar] [CrossRef] [PubMed]
  95. Wang, X.L.; Feng, S.T.; Wang, Y.T.; Yuan, Y.H.; Li, Z.P.; Chen, N.H.; Wang, Z.Z.; Zhang, Y. Mitophagy, a Form of Selective Autophagy, Plays an Essential Role in Mitochondrial Dynamics of Parkinson’s Disease. Cell. Mol. Neurobiol. 2021. [Google Scholar] [CrossRef]
  96. Liu, X.; Hebron, M.; Shi, W.; Lonskaya, I.; Moussa, C.E.H. Ubiquitin specific protease-13 independently regulates parkin ubiquitination and α-synuclein clearance in alpha-synucleinopathies. Hum. Mol. Genet. 2019, 28, 548–560. [Google Scholar] [CrossRef] [PubMed]
  97. Cartelli, D.; Amadeo, A.; Calogero, A.M.; Casagrande, F.V.M.; De Gregorio, C.; Gioria, M.; Kuzumaki, N.; Costa, I.; Sassone, J.; Ciammola, A.; et al. Parkin absence accelerates microtubule aging in dopaminergic neurons. Neurobiol. Aging 2018, 61, 66–74. [Google Scholar] [CrossRef] [PubMed]
  98. Peker, N.; Donipadi, V.; Sharma, M.; McFarlane, C.; Kambadur, R. Loss of Parkin impairs mitochondrial function and leads to muscle atrophy. Am. J. Physiol. Cell Physiol. 2018, 315, C164–C185. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  99. Williams, E.T.; Glauser, L.; Tsika, E.; Jiang, H.; Islam, S.; Moore, D.J. Parkin mediates the ubiquitination of VPS35 and modulates retromer-dependent endosomal sorting. Hum. Mol. Genet. 2018, 27, 3189–3205. [Google Scholar] [CrossRef]
  100. Singh, K.; Han, K.; Tilve, S.; Wu, K.; Geller, H.M.; Sack, M.N. Parkin targets NOD2 to regulate astrocyte endoplasmic reticulum stress and inflammation. Glia 2018, 66, 2427–2437. [Google Scholar] [CrossRef]
  101. Hwang, C.J.; Kim, Y.E.; Son, D.J.; Park, M.H.; Choi, D.Y.; Park, P.H.; Hellström, M.; Han, S.B.; Oh, K.W.; Park, E.K.; et al. Parkin deficiency exacerbate ethanol-induced dopaminergic neurodegeneration by P38 pathway dependent inhibition of autophagy and mitochondrial function. Redox Biol. 2017, 11, 456–468. [Google Scholar] [CrossRef]
  102. Kabayama, H.; Tokushige, N.; Takeuchi, M.; Kabayama, M.; Fukuda, M.; Mikoshiba, K. Parkin promotes proteasomal degradation of synaptotagmin IV by accelerating polyubiquitination. Mol. Cell. Neurosci. 2017, 80, 89–99. [Google Scholar] [CrossRef]
  103. Lin, M.Y.; Cheng, X.T.; Tammineni, P.; Xie, Y.; Zhou, B.; Cai, Q.; Sheng, Z.H. Releasing Syntaphilin Removes Stressed Mitochondria from Axons Independent of Mitophagy under Pathophysiological Conditions. Neuron 2017, 94, 595–610.e6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Pickrell, A.M.; Huang, C.H.; Kennedy, S.R.; Ordureau, A.; Sideris, D.P.; Hoekstra, J.G.; Harper, J.W.; Youle, R.J. Endogenous Parkin Preserves Dopaminergic Substantia Nigral Neurons following Mitochondrial DNA Mutagenic Stress. Neuron 2015, 87, 371–381. [Google Scholar] [CrossRef] [Green Version]
  105. Kubli, D.A.; Zhang, X.; Lee, Y.; Hanna, R.A.; Quinsay, M.N.; Nguyen, C.K.; Jimenez, R.; Petrosyans, S.; Murphy, A.N.; Gustafsson, Å.B. Parkin protein deficiency exacerbates cardiac injury and reduces survival following myocardial infarction. J. Biol. Chem. 2013, 288, 915–926. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Sul, J.W.; Park, M.Y.; Shin, J.; Kim, Y.R.; Yoo, S.E.; Kong, Y.Y.; Kwon, K.S.; Lee, Y.H.; Kim, E. Accumulation of the parkin substrate, FAF1, plays a key role in the dopaminergic neurodegeneration. Hum. Mol. Genet. 2013, 22, 1558–1573. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Ekholm-Reed, S.; Baker, R.; Campos, A.R.; Stouffer, D.; Henze, M.; Wolf, D.A.; Loring, J.F.; Thomas, E.A.; Reed, S.I. Reducing Mcl-1 gene dosage induces dopaminergic neuronal loss and motor impairments in Park2 knockout mice. Commun. Biol. 2019, 2, 1–7. [Google Scholar] [CrossRef] [Green Version]
  108. Ekholm-Reed, S.; Goldberg, M.S.; Schlossmacher, M.G.; Reed, S.I. Parkin-Dependent Degradation of the F-Box Protein Fbw7 Promotes Neuronal Survival in Response to Oxidative Stress by Stabilizing Mcl. Mol. Cell. Biol. 2013, 33, 3627–3643. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  109. Berthet, A.; Bezard, E.; Porras, G.; Fasano, S.; Barroso-Chinea, P.; Dehay, B.; Martinez, A.; Thiolat, M.L.; Nosten-Bertrand, M.; Giros, B.; et al. L-DOPA impairs proteasome activity in parkinsonism through D 1 dopamine receptor. J. Neurosci. 2012, 32, 681–691. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  110. Kim, K.Y.; Stevens, M.V.; Akter, M.H.; Rusk, S.E.; Huang, R.J.; Cohen, A.; Noguchi, A.; Springer, D.; Bocharov, A.V.; Eggerman, T.L.; et al. Parkin is a lipid-responsive regulator of fat uptake in mice and mutant human cells. J. Clin. Investig. 2011, 121, 3701–3712. [Google Scholar] [CrossRef]
  111. Kitada, T.; Pisani, A.; Karouani, M.; Haburcak, M.; Martella, G.; Tscherter, A.; Platania, P.; Wu, B.; Pothos, E.N.; Shen, J. Impaired dopamine release and synaptic plasticity in the striatum of Parkin-/- mice. J. Neurochem. 2009, 110, 613–621. [Google Scholar] [CrossRef] [PubMed]
  112. Frank-Cannon, T.C.; Tran, T.; Ruhn, K.A.; Martinez, T.N.; Hong, J.; Marvin, M.; Hartley, M.; Treviño, I.; O’Brien, D.E.; Casey, B.; et al. Parkin deficiency increases vulnerability to inflammation-related nigral degeneration. J. Neurosci. 2008, 28, 10825–10834. [Google Scholar] [CrossRef] [PubMed]
  113. Palacino, J.J.; Sagi, D.; Goldberg, M.S.; Krauss, S.; Motz, C.; Wacker, M.; Klose, J.; Shen, J. Mitochondrial Dysfunction and Oxidative Damage in parkin-deficient Mice. J. Biol. Chem. 2004, 279, 18614–18622. [Google Scholar] [CrossRef] [Green Version]
  114. Goldberg, M.S.; Fleming, S.M.; Palacino, J.J.; Cepeda, C.; Lam, H.A.; Bhatnagar, A.; Meloni, E.G.; Wu, N.; Ackerson, L.C.; Klapstein, G.J.; et al. Parkin-deficient Mice Exhibit Nigrostriatal Deficits but not Loss of Dopaminergic Neurons. J. Biol. Chem. 2003, 278, 43628–43635. [Google Scholar] [CrossRef] [Green Version]
  115. Song, L.; McMackin, M.; Nguyen, A.; Cortopassi, G. Parkin deficiency accelerates consequences of mitochondrial DNA deletions and Parkinsonism. Neurobiol. Dis. 2017, 100, 30–38. [Google Scholar] [CrossRef]
  116. Sliter, D.A.; Martinez, J.; Hao, L.; Chen, X.; Sun, N.; Fischer, T.D.; Burman, J.L.; Li, Y.; Zhang, Z.; Narendra, D.P.; et al. Parkin and PINK1 mitigate STING-induced inflammation. Nature 2018, 561, 258–262. [Google Scholar] [CrossRef]
  117. Matheoud, D.; Sugiura, A.; Bellemare-Pelletier, A.; Laplante, A.; Rondeau, C.; Chemali, M.; Fazel, A.; Bergeron, J.J.; Trudeau, L.-E.; Burelle, Y.; et al. Parkinson’s Disease-Related Proteins PINK1 and Parkin Repress Mitochondrial Antigen Presentation. Cell 2016, 166, 314–327. [Google Scholar] [CrossRef] [Green Version]
  118. Ashrafi, G.; Schlehe, J.S.; LaVoie, M.J.; Schwarz, T.L. Mitophagy of damaged mitochondria occurs locally in distal neuronal axons and requires PINK1 and Parkin. J. Cell Biol. 2014, 206, 655–670. [Google Scholar] [CrossRef]
  119. Rogers, R.S.; Tungtur, S.; Tanaka, T.; Nadeau, L.L.; Badawi, Y.; Wang, H.; Ni, H.M.; Ding, W.X.; Nishimune, H. Impaired mitophagy plays a role in denervation of neuromuscular junctions in ALS mice. Front. Neurosci. 2017, 11, 1–17. [Google Scholar] [CrossRef] [PubMed]
  120. Madeo, G.; Martella, G.; Schirinzi, T.; Ponterio, G.; Shen, J.; Bonsi, P.; Pisani, A. Aberrant striatal synaptic plasticity in monogenic parkinsonisms. Neuroscience 2012, 211, 126–135. [Google Scholar] [CrossRef] [PubMed]
  121. Chen, L.; Xie, Z.; Turkson, S.; Zhuang, X. A53T human α-synuclein overexpression in transgenic mice induces pervasive mitochondria macroautophagy defects preceding dopamine neuron degeneration. J. Neurosci. 2015, 35, 890–905. [Google Scholar] [CrossRef]
  122. Pearlstein, E.; Michel, F.J.; Save, L.; Ferrari, D.C.; Hammond, C. Abnormal development of glutamatergic synapses afferent to dopaminergic neurons of the pink1–/– mouse model of Parkinson’s disease. Front. Cell. Neurosci. 2016, 10, 1–13. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Kubli, D.A.; Cortez, M.Q.; Moyzis, A.G.; Najor, R.H.; Lee, Y.; Gustafsson, Å.B. PINK1 is dispensable for mitochondrial recruitment of parkin and activation of mitophagy in cardiac myocytes. PLoS ONE 2015, 10, e0130707. [Google Scholar] [CrossRef] [PubMed]
  124. Gispert, S.; Ricciardi, F.; Kurz, A.; Azizov, M.; Hoepken, H.H.; Becker, D.; Voos, W.; Leuner, K.; Müller, W.E.; Kudin, A.P.; et al. Parkinson phenotype in aged PINK1-deficient mice is accompanied by progressive mitochondrial dysfunction in absence of neurodegeneration. PLoS ONE 2009, 4, e5777. [Google Scholar] [CrossRef]
  125. Pellegrini, L.; Hauser, D.N.; Li, Y.; Mamais, A.; Beilina, A.; Kumaran, R.; Wetzel, A.; Nixon-Abell, J.; Heaton, G.; Rudenko, I.; et al. Proteomic analysis reveals co-ordinated alterations in protein synthesis and degradation pathways in LRRK2 knockout mice. Hum. Mol. Genet. 2018, 27, 3257–3271. [Google Scholar] [CrossRef] [PubMed]
  126. Parisiadou, L.; Yu, J.; Sgobio, C.; Xie, C.; Liu, G.; Sun, L.; Gu, X.-L.; Lin, X.; Crowley, N.A.; Lovinger, D.M.; et al. LRRK2 regulates synaptogenesis and dopamine receptor activation through modulation of PKA activity. Nat. Neurosci. 2014, 17, 367–376. [Google Scholar] [CrossRef] [PubMed]
  127. Cho, H.J.; Yu, J.; Xie, C.; Rudrabhatla, P.; Chen, X.; Wu, J.; Parisiadou, L.; Liu, G.; Sun, L.; Ma, B.; et al. Leucine-rich repeat kinase 2 regulates Sec16A at ER exit sites to allow ER –Golgi export. EMBO J. 2014, 33, 2314–2331. [Google Scholar] [CrossRef] [Green Version]
  128. Sepulveda, B.; Mesias, R.; Li, X.; Yue, Z.; Benson, D.L. Short- and Long-Term Effects of LRRK2 on Axon and Dendrite Growth. PLoS ONE 2013, 8, e61986. [Google Scholar] [CrossRef] [Green Version]
  129. Dwyer, Z.; Rudyk, C.; Thompson, A.; Farmer, K.; Fenner, B.; Fortin, T.; Derksen, A.; Sun, H.; Hayley, S. Leucine-rich repeat kinase-2 (LRRK2) modulates microglial phenotype and dopaminergic neurodegeneration. Neurobiol. Aging 2020, 91, 45–55. [Google Scholar] [CrossRef] [PubMed]
  130. Maekawa, T.; Tsushima, H.; Kawakami, F.; Kawashima, R.; Kodo, M.; Imai, M.; Ichikawa, T. Leucine-rich repeat kinase 2 is associated with activation of the paraventricular nucleus of the hypothalamus and stress-related gastrointestinal dysmotility. Front. Neurosci. 2019, 13, 1–10. [Google Scholar] [CrossRef]
  131. Wang, H.; Zhang, X.; Zuo, Z.; Zhang, Q.; Pan, Y.; Zeng, B.; Li, W.; Wei, H.; Liu, Z. Rip2 Is Required for Nod2-Mediated Lysozyme Sorting in Paneth Cells. J. Immunol. 2017, 198, 3729–3736. [Google Scholar] [CrossRef] [Green Version]
  132. Ferrazza, R.; Cogo, S.; Melrose, H.; Bubacco, L.; Greggio, E.; Guella, G.; Civiero, L.; Plotegher, N. LRRK2 deficiency impacts ceramide metabolism in brain. Biochem. Biophys. Res. Commun. 2016, 478, 1141–1146. [Google Scholar] [CrossRef] [Green Version]
  133. 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.e6. [Google Scholar] [CrossRef] [Green Version]
  134. Tong, Y.; Yamaguchi, H.; Giaime, E.; Boyle, S.; Kopan, R.; Kelleher, R.J.; Shen, J. Loss of leucine-rich repeat kinase 2 causes impairment of protein degradation pathways, accumulation of α-synuclein, and apoptotic cell death in aged mice. Proc. Natl. Acad. Sci. USA 2010, 107, 9879–9884. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Xing, W.; Liu, J.; Cheng, S.; Vogel, P.; Mohan, S.; Brommage, R. Targeted disruption of leucine-rich repeat kinase 1 but not leucine-rich repeat kinase 2 in mice causes severe osteopetrosis. J. Bone Miner. Res. 2013, 28, 1962–1974. [Google Scholar] [CrossRef] [PubMed]
  136. Takagawa, T.; Kitani, A.; Fuss, I.; Levine, B.; Brant, S.R.; Tajima, M.; Nakamura, S.; Strober, W. Dectin-1–Induced immunity in a mouse model of colitis. Sci. Transl. Med. 2019, 10, 1–25. [Google Scholar]
  137. Beccano-kelly, D.A.; Volta, M.; Munsie, L.N.; Paschall, S.A. LRRK2 overexpression alters glutamatergic presynaptic plasticity, striatal dopamine tone, postsynaptic signal transduction, behavioural hypoactivity and memory deficits. Hum. Mol. Genet. 2014, 24, 1336–1349. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  138. Parisiadou, L.; Xie, C.; Hyun, J.C.; Lin, X.; Gu, X.L.; Long, C.X.; Lobbestael, E.; Baekelandt, V.; Taymans, J.M.; Sun, L.; et al. Phosphorylation of ezrin/radixin/moesin proteins by LRRK2 promotes the rearrangement of actin cytoskeleton in neuronal morphogenesis. J. Neurosci. 2009, 29, 13971–13980. [Google Scholar] [CrossRef]
  139. Henry, A.G.; Aghamohammadzadeh, S.; Samaroo, H.; Chen, Y.; Mou, K.; Needle, E.; Hirst, W.D. Pathogenic LRRK2 mutations, through increased kinase activity, produce enlarged lysosomes with reduced degradative capacity and increase ATP13A2 expression. Hum. Mol. Genet. 2015, 24, 6013–6028. [Google Scholar] [CrossRef] [Green Version]
  140. Beccano-Kelly, D.A.; Kuhlmann, N.; Tatarnikov, I.; Volta, M.; Munsie, L.N.; Chou, P.; Cao, L.P.; Han, H.; Tapia, L.; Farrer, M.J.; et al. Synaptic function is modulated by LRRK2 and glutamate release is increased in cortical neurons of G2019S LRRK2 knock-in mice. Front. Cell. Neurosci. 2014, 8, 1–11. [Google Scholar] [CrossRef] [Green Version]
  141. Park, J.; Lee, J.-W.; Cooper, S.C.; Broxmeyer, H.E.; Cannon, J.R.; Kim, C.H. Parkinson disease–associated LRRK2 G2019S transgene disrupts marrow myelopoiesis and peripheral Th17 response. J. Leukoc. Biol. 2017, 102, 1093–1102. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Daher, J.P.L.; Abdelmotilib, H.A.; Hu, X.; Volpicelli-Daley, L.A.; Moehle, M.S.; Fraser, K.B.; Needle, E.; Chen, Y.; Steyn, S.J.; Galatsis, P.; et al. Leucine-rich repeat kinase 2 (LRRK2) pharmacological inhibition abates α-synuclein gene-induced neurodegeneration. J. Biol. Chem. 2015, 290, 19433–19444. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Smith, N.J.; Fuller, M.; Saville, J.T.; Cox, T.M. Reduced cerebral vascularization in experimental neuronopathic Gaucher disease. J. Pathol. 2018, 244, 120–128. [Google Scholar] [CrossRef] [PubMed]
  144. Yamada, T.; Dawson, T.M.; Yanagawa, T.; Iijima, M.; Sesaki, H. SQSTM1/p62 promotes mitochondrial ubiquitination independently of PINK1 and PRKN/parkin in mitophagy. Autophagy 2019, 15, 2012–2018. [Google Scholar] [CrossRef] [PubMed]
  145. Kelm-Nelson, C.A.; Brauer, A.F.L.; Barth, K.J.; Lake, J.M.; Sinnen, M.L.K.; Stehula, F.J.; Muslu, C.; Marongiu, R.; Kaplitt, M.G.; Ciucci, M.R. Characterization of early-onset motor deficits in the Pink1−/− mouse model of Parkinson disease. Brain Res. 2018, 1680, 1–12. [Google Scholar] [CrossRef]
  146. Das Banerjee, T.; Dagda, R.Y.; Dagda, M.; Chu, C.T.; Rice, M.; Vazquez-Mayorga, E.; Dagda, R.K. PINK1 regulates mitochondrial trafficking in dendrites of cortical neurons through mitochondrial PKA. J. Neurochem. 2017, 142, 545–559. [Google Scholar] [CrossRef] [Green Version]
  147. Haque, M.E.; Mount, M.P.; Safarpour, F.; Abdel-Messih, E.; Callaghan, S.; Mazerolle, C.; Kitada, T.; Slack, R.S.; Wallace, V.; Shen, J.; et al. Inactivation of Pink1 gene in Vivo sensitizes dopamine-producing neurons to 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) and can be rescued by autosomal recessive Parkinson disease genes, Parkin or DJ-1. J. Biol. Chem. 2012, 287, 23162–23170. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  148. Martella, G.; Madeo, G.; Schirinzi, T.; Tassone, A.; Sciamanna, G.; Spadoni, F.; Stefani, A.; Shen, J.; Pisani, A.; Bonsi, P. Altered profile and D2-dopamine receptor modulation of high voltage-activated calcium current in striatal medium spiny neurons from animal models of Parkinson’s disease. Neuroscience 2011, 177, 240–251. [Google Scholar] [CrossRef]
  149. Billia, F.; Hauck, L.; Konecny, F.; Rao, V.; Shen, J.; Mak, T.W. PTEN-inducible kinase 1 (PINK1)/Park6 is indispensable for normal heart function. Proc. Natl. Acad. Sci. USA 2011, 108, 9572–9577. [Google Scholar] [CrossRef] [Green Version]
  150. Kitada, T.; Pisani, A.; Porter, D.R.; Yamaguchi, H.; Tscherter, A.; Martella, G.; Bonsi, P.; Zhang, C.; Pothos, E.N.; Shen, J. Impaired dopamine release and synaptic plasticity in the striatum of PINK1-deficient mice. Proc. Natl. Acad. Sci. USA 2007, 104, 11441–11446. [Google Scholar] [CrossRef] [Green Version]
  151. Matheoud, D.; Cannon, T.; Voisin, A.; Penttinen, A.M.; Ramet, L.; Fahmy, A.M.; Ducrot, C.; Laplante, A.; Bourque, M.J.; Zhu, L.; et al. Intestinal infection triggers Parkinson’s disease-like symptoms in Pink1 −/− mice. Nature 2019, 571, 565–569. [Google Scholar] [CrossRef]
  152. Moisoi, N.; Fedele, V.; Edwards, J.; Martins, L.M. Loss of PINK1 enhances neurodegeneration in a mouse model of Parkinson’s disease triggered by mitochondrial stress. Neuropharmacology 2014, 77, 350–357. [Google Scholar] [CrossRef]
  153. Martella, G.; Platania, P.; Vita, D.; Sciamanna, G.; Cuomo, D.; Tassone, A.; Tscherter, A.; Kitada, T.; Bonsi, P.; Shen, J.; et al. Enhanced sensitivity to group II mGlu receptor activation at corticostriatal synapses in mice lacking the familial parkinsonism-linked genes PINK1 or Parkin. Exp. Neurol. 2009, 215, 388–396. [Google Scholar] [CrossRef] [Green Version]
  154. Bhatia, D.; Chung, K.P.; Nakahira, K.; Patino, E.; Rice, M.C.; Torres, L.K.; Muthukumar, T.; Choi, A.M.K.; Akchurin, O.M.; Choi, M.E. Mitophagy-dependent macrophage reprogramming protects against kidney fibrosis. JCI Insight 2019, 4, 1–20. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. Auburger, G.; Gispert, S.; Brehm, N. Methyl-arginine profile of brain from aged PINK1-KO+A53T-SNCA mice suggests altered mitochondrial biogenesis. Parkinsons. Dis. 2016, 2016, 4686185. [Google Scholar] [CrossRef] [Green Version]
  156. Cabin, D.E.; Gispert-Sanchez, S.; Murphy, D.; Auburger, G.; Myers, R.R.; Nussbaum, R.L. Exacerbated synucleinopathy in mice expressing A53T SNCA on a Snca null background. Neurobiol. Aging 2005, 26, 25–35. [Google Scholar] [CrossRef]
  157. Gispert, S.; Brehm, N.; Weil, J.; Seidel, K.; Rüb, U.; Kern, B.; Walter, M.; Roeper, J.; Auburger, G. Potentiation of neurotoxicity in double-mutant mice with Pink1 ablation and A53T-SNCA overexpression. Hum. Mol. Genet. 2015, 24, 1061–1076. [Google Scholar] [CrossRef] [Green Version]
  158. Jin, G.; Xu, C.; Zhang, X.; Long, J.; Rezaeian, A.H.; Liu, C.; Furth, M.E.; Kridel, S.; Pasche, B.; Bian, X.-W.; et al. Atad3a suppresses Pink1-dependent mitophagy to maintain homeostasis of hematopoietic progenitor cells. Nat. Immunol. 2018, 19, 29–40. [Google Scholar] [CrossRef] [PubMed]
  159. Wang, R.; Tan, J.; Chen, T.; Han, H.; Tian, R.; Tan, Y.; Wu, Y.; Cui, J.; Chen, F.; Li, J.; et al. ATP13A2 facilitates HDAC6 recruitment to lysosome to promote autophagosome–lysosome fusion. J. Cell Biol. 2019, 218, 267–284. [Google Scholar] [CrossRef] [PubMed]
  160. Rayaprolu, S.; Seven, Y.B.; Howard, J.; Duffy, C.; Altshuler, M.; Moloney, C.; Giasson, B.I.; Lewis, J. Partial loss of ATP13A2 causes selective gliosis independent of robust lipofuscinosis. Mol. Cell. Neurosci. 2018, 92, 17–26. [Google Scholar] [CrossRef] [PubMed]
  161. Schultheis, P.J.; Fleming, S.M.; Clippinger, A.K.; Lewis, J.; Tsunemi, T.; Giasson, B.; Dickson, D.W.; Mazzulli, J.R.; Bardgett, M.E.; Haik, K.L.; et al. Atp13a2-deficient mice exhibit neuronal ceroid lipofuscinosis, limited α-synuclein accumulation and age-dependent sensorimotor deficits. Hum. Mol. Genet 2013, 22, 2067–2082. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  162. Burchell, V.S.; Nelson, D.E.; Sanchez-Martinez, A.; Delgado-Camprubi, M.; Ivatt, R.M.; Pogson, J.H.; Randle, S.J.; Wray, S.; Lewis, P.A.; Houlden, H.; et al. The Parkinson’s disease-linked proteins Fbxo7 and Parkin interact to mediate mitophagy. Nat. Neurosci. 2013, 16, 1257–1265. [Google Scholar] [CrossRef] [Green Version]
  163. Braschi, E.; Goyon, V.; Zunino, R.; Mohanty, A.; Xu, L.; McBride, H.M. Vps35 mediates vesicle transport between the mitochondria and peroxisomes. Curr. Biol. 2010, 20, 1310–1315. [Google Scholar] [CrossRef] [Green Version]
  164. Outeiro, T.F.; Harvey, K.; Dominguez-Meijide, A.; Gerhardt, E. LRRK2, alpha-synuclein, and tau: Partners in crime or unfortunate bystanders? Biochem. Soc. Trans. 2019, 47, 827–838. [Google Scholar] [CrossRef]
  165. Cresto, N.; Gardier, C.; Gubinelli, F.; Gaillard, M.C.; Liot, G.; West, A.B.; Brouillet, E. The unlikely partnership between LRRK2 and α-synuclein in Parkinson’s disease. Eur. J. Neurosci. 2019, 49, 339–363. [Google Scholar] [CrossRef]
  166. Toyofuku, T.; Okamoto, Y.; Ishikawa, T.; Sasawatari, S.; Kumanogoh, A. LRRK 2 regulates endoplasmic reticulum–mitochondrial tethering through the PERK -mediated ubiquitination pathway. EMBO J. 2020, 39, e100875. [Google Scholar] [CrossRef] [PubMed]
  167. Ekstrand, M.I.; Galter, D. The MitoPark Mouse—An animal model of Parkinson’s disease with impaired respiratory chain function in dopamine neurons. Parkinsonism Relat. Disord. 2009, 15 (Suppl. 3), S185–S188. [Google Scholar] [CrossRef]
  168. Zhang, S.; Wang, R.; Wang, G. Impact of Dopamine Oxidation on Dopaminergic Neurodegeneration. ACS Chem. Neurosci. 2019, 10, 945–953. [Google Scholar] [CrossRef] [PubMed]
  169. Muñoz, P.; Huenchuguala, S.; Paris, I.; Segura-Aguilar, J. Dopamine Oxidation and Autophagy. Parkinsons. Dis. 2012, 2012, 920953. [Google Scholar] [CrossRef] [PubMed]
  170. Kanďár, R. The ratio of oxidized and reduced forms of selected antioxidants as a possible marker of oxidative stress in humans. Biomed. Chromatogr. 2016, 30, 13–28. [Google Scholar] [CrossRef] [PubMed]
  171. Nagase, M.; Yamamoto, Y.; Matsumoto, N.; Arai, Y.; Hirose, N. Increased oxidative stress and coenzyme Q10 deficiency in centenarians. J. Clin. Biochem. Nutr. 2018, 63, 129–136. [Google Scholar] [CrossRef] [Green Version]
  172. Sourris, K.C.; Harcourt, B.E.; Tang, P.H.; Morley, A.L.; Huynh, K.; Penfold, S.A.; Coughlan, M.T.; Cooper, M.E.; Nguyen, T.-V.; Ritchie, R.H.; et al. Ubiquinone (coenzyme Q10) prevents renal mitochondrial dysfunction in an experimental model of type 2 diabetes. Free Radic. Biol. Med. 2012, 52, 716–723. [Google Scholar] [CrossRef] [PubMed]
  173. Bénard, G.; Massa, F.; Puente, N.; Lourenço, J.; Bellocchio, L.; Soria-Gómez, E.; Matias, I.; Delamarre, A.; Metna-Laurent, M.; Cannich, A.; et al. Mitochondrial CB1 receptors regulate neuronal energy metabolism. Nat. Neurosci. 2012, 15, 558–564. [Google Scholar] [CrossRef]
  174. Melser, S.; Zottola, A.C.P.; Serrat, R.; Puente, N.; Grandes, P.; Marsicano, G.; Hebert-Chatelain, E. Functional Analysis of Mitochondrial CB1 Cannabinoid Receptors (mtCB1) in the Brain. Methods Enzymol. 2017, 593, 143–174. [Google Scholar]
  175. Valenzuela, R.; Costa-Besada, M.A.; Iglesias-Gonzalez, J.; Perez-Costas, E.; Villar-Cheda, B.; Garrido-Gil, P.; Melendez-Ferro, M.; Soto-Otero, R.; Lanciego, J.L.; Henrion, D.; et al. Mitochondrial angiotensin receptors in dopaminergic neurons. Role in cell protection and aging-related vulnerability to neurodegeneration. Cell Death Dis. 2016, 7, e2427. [Google Scholar] [CrossRef] [Green Version]
  176. Lindsay, J.; Laurin, D.; Verreault, R.; Hébert, R.; Helliwell, B.; Hill, G.B.; McDowell, I. Risk factors for Alzheimer’s disease: A prospective analysis from the Canadian Study of Health and Aging. Am. J. Epidemiol. 2002, 156, 445–453. [Google Scholar] [CrossRef] [Green Version]
  177. Espinosa, J.; Rocha, A.; Nunes, F.; Costa, M.S.; Schein, V.; Kazlauckas, V.; Kalinine, E.; Souza, D.O.; Cunha, R.A.; Porciúncula, L.O. Caffeine consumption prevents memory impairment, neuronal damage, and adenosine A2A receptors upregulation in the hippocampus of a rat model of sporadic dementia. J. Alzheimer’s Dis. JAD 2013, 34, 509–518. [Google Scholar] [CrossRef] [PubMed]
  178. Cunha, R.A. Cafeína, receptores de adenosina, memoria y enfermedad de Alzheimer. Med. Clin. 2008, 131, 790–795. [Google Scholar] [CrossRef]
  179. Canas, P.M.; Porciuncula, L.O.; Cunha, G.M.A.; Silva, C.G.; Machado, N.J.; Oliveira, J.M.A.; Oliveira, C.R.; Cunha, R.A. Adenosine A2A Receptor Blockade Prevents Synaptotoxicity and Memory Dysfunction Caused by -Amyloid Peptides via p38 Mitogen-Activated Protein Kinase Pathway. J. Neurosci. 2009, 29, 14741–14751. [Google Scholar] [CrossRef]
  180. Eskelinen, M.H.; Kivipelto, M. Caffeine as a protective factor in dementia and Alzheimer’s disease. J. Alzheimer’s Dis. 2010, 20, S167–S174. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  181. Eskelinen, M.H.; Ngandu, T.; Tuomilehto, J.; Soininen, H.; Kivipelto, M. Midlife coffee and tea drinking and the risk of late-life dementia: A population-based CAIDE study. J. Alzheimer’s Dis. 2009, 16, 85–91. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  182. Pedata, F.; Pugliese, A.M.; Melani, A.; Gianfriddo, M. A2A receptors in neuroprotection of dopaminergic neurons. Neurology 2003, 61, S49–S50. [Google Scholar] [CrossRef] [PubMed]
  183. Franco, R.; Navarro, G. Adenosine A2A receptor antagonists in neurodegenerative diseases: Huge potential and huge challenges. Front. Psychiatry 2018, 9, 1–5. [Google Scholar] [CrossRef] [Green Version]
  184. Franco, R. Café y salud mental. Aten. Primaria 2009, 41, 578–581. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  185. Abbas, M.M.; Xu, Z.; Tan, L.C.S. Epidemiology of Parkinson’s Disease-East Versus West. Mov. Disord. Clin. Pract. 2018, 5, 14–28. [Google Scholar] [CrossRef]
  186. Ascherio, A.; Schwarzschild, M.A. The epidemiology of Parkinson’s disease: Risk factors and prevention. Lancet Neurol. 2016, 15, 1257–1272. [Google Scholar] [CrossRef]
  187. Ribeiro, J.A.; Sebastiao, A.M. Caffeine and adenosine. J. Alzheimer’s Dis. 2010, 20, S3–S15. [Google Scholar] [CrossRef] [Green Version]
  188. Qi, H.; Li, S. Dose-response meta-analysis on coffee, tea and caffeine consumption with risk of Parkinson’s disease. Geriatr. Gerontol. Int. 2014, 14, 430–439. [Google Scholar] [CrossRef]
  189. Liu, R.; Guo, X.; Park, Y.; Huang, X.; Sinha, R.; Freedman, N.D.; Hollenbeck, A.R.; Blair, A.; Chen, H. Caffeine intake, smoking, and risk of parkinson disease in men and women. Am. J. Epidemiol. 2012, 175, 1200–1207. [Google Scholar] [CrossRef] [Green Version]
Figure 1. STRING analysis of connections between (human) genes in Table 1. Panel (A): using by default settings. Panel (B). Restricted settings (mainly leaving aside connections driven by PD-related literature; see text for details). Panel (C): Interaction-enriched connection pattern (see text for details). Line color code: sky blue, known interactions from curated databases; magenta, experimentally determined interactions; green, predicted from neighborhood; red, predicted from gene fusions; blue, predicted from gene co-occurrence; pastel green, textmining; black, coexpression; and clear violet, protein homology.
Figure 1. STRING analysis of connections between (human) genes in Table 1. Panel (A): using by default settings. Panel (B). Restricted settings (mainly leaving aside connections driven by PD-related literature; see text for details). Panel (C): Interaction-enriched connection pattern (see text for details). Line color code: sky blue, known interactions from curated databases; magenta, experimentally determined interactions; green, predicted from neighborhood; red, predicted from gene fusions; blue, predicted from gene co-occurrence; pastel green, textmining; black, coexpression; and clear violet, protein homology.
Ijms 22 04643 g001
Figure 2. Enriched STRING analysis of connections between (human) genes in Table 1. Line color code; sky blue, known interactions from curated databases; magenta, experimentally determined interactions; green, predicted from neighborhood; red, predicted from gene fusions; blue, predicted from gene co-occurrence; pastel green, textmining; black, coexpression; and clear violet, protein homology.
Figure 2. Enriched STRING analysis of connections between (human) genes in Table 1. Line color code; sky blue, known interactions from curated databases; magenta, experimentally determined interactions; green, predicted from neighborhood; red, predicted from gene fusions; blue, predicted from gene co-occurrence; pastel green, textmining; black, coexpression; and clear violet, protein homology.
Ijms 22 04643 g002
Figure 3. Mechanisms of dyshomeostasis of dopaminergic neurons. Panel (A). Mutations in SNCA leads to aggregation of mutated α-synuclein that affects the mechanisms of protein handling and vesicle transport, thus dysbalancing mitochondrial dynamics and function. Panel (B). Mutations in genes that affect protein processing, vesicle transport and mitochondrial function leads to aggregation of non-mutated α-synuclein. Red dots represent ATP molecules, mainly synthesized in the mitochondria of neurons, that are needed for all the processes depicted in the schemes.
Figure 3. Mechanisms of dyshomeostasis of dopaminergic neurons. Panel (A). Mutations in SNCA leads to aggregation of mutated α-synuclein that affects the mechanisms of protein handling and vesicle transport, thus dysbalancing mitochondrial dynamics and function. Panel (B). Mutations in genes that affect protein processing, vesicle transport and mitochondrial function leads to aggregation of non-mutated α-synuclein. Red dots represent ATP molecules, mainly synthesized in the mitochondria of neurons, that are needed for all the processes depicted in the schemes.
Ijms 22 04643 g003
Table 1. Genes considered in this review and having mutations likely related to early-onset PD. See description of gene products in Table 2.
Table 1. Genes considered in this review and having mutations likely related to early-onset PD. See description of gene products in Table 2.
GeneInheritanceProposed Disease MechanismDisease OnsetMutationFrequencyConfidence as
Actual PD Gene
Year of
Discovery
Animal Model
EarlyLate
SNCADGoF or overexpressionOften with
dementia
Missense or
multiplication
Very rareVery high1997,2003+
LRRK2DGoFXMissensecommonVery high2004+
GBADLikely LoFXMissense or LoFcommonVery high2009+
UCHL1DLoFNAMissenseunclearLow1998+
VPS35DLoFXMissenseVery rareVery high2011+
PRKNRLoFXMissense or LoFrareVery high1998+
PINK1RLoFXMissense or loss
of function
rareVery high2004+
ATP13A2RLoFAtypical PDMissense or LoFVery rareVery high2006+
PLA2G6RLoFXMissense or LoFrareVery high2009NA
DNAJC6RLoFXMissense o LoFVery rareHigh2012NA
SYNJ1RLoF(often) Atypical PDMissense or loss
of function
Very rareVery high2013NA
DJ-1/PARK7RLoFXMissenseVery rareVery High2003+
FBXO7RLoFXMissenseVery rareVery High2008NA
D: Dominant; R: Recessive; NA: Not available; GoF: Gain-of-function; LoF: Loss-of-function.
Table 2. Description of the products of the genes in Table 1. Function retrieved from https://string-db.org/ (accessed on 10 April 2021).
Table 2. Description of the products of the genes in Table 1. Function retrieved from https://string-db.org/ (accessed on 10 April 2021).
GeneEnsembl IdentifierProteinFunction (Information Obtained from STRING Data Base)
SNCA ENSP00000338345 α-synuclein Involved in regulating dopamine release and transport. Induces the fibrillization of tau protein. Reduces neuronal responsiveness to different apoptotic stimuli, thus promoting a decreased caspase-3 activation.
LRRK2 ENSP00000298910 Leucine-rich repeat serine/threonine-protein kinase 2 Regulates autophagy in a positive way by means of the calcium- dependent activation of the CaMKK/AMPK pathway, also involving the activation of nicotinic acid adenine dinucleotide phosphate (NAADP) receptors, increases in lysosomal pH, and release of Ca++ from lysosomes.
GBA ENSP00000314508 Glucosylceramidase beta Involved in the hydrolization of glucocerebroside. Localized in lysosomes.
UCHL1 ENSP00000284440 Ubiquitin carboxyl-terminal hydrolase isozyme L1 Deubiquitinating enzyme, generates ubiquitin monomers. Might prevent the degradation of monoubiquitin in lysosomes. Its expression is highly specific to neurons and to cells of the diffuse neuroendocrine system and their tumors.
VPS35 ENSP00000299138 Vacuolar protein sorting-associated protein 35 Involved in autophagy. Is part of the retromer cargo-selective complex (CSC), which is responsible for transporting select cargo proteins between vesicular structures (e.g., endosomes, lysosomes, vacuoles) and the Golgi apparatus.
PRKN ENSP00000355865 E3 ubiquitin-protein ligase parkin Ubiquitin ligase; covalently binds ubiquitin residues onto proteins. Involved in the removal of abnormally folded or damaged proteins thanks to ‘Lys-63’-linked polyubiquitination of misfolded proteins.
PINK1 ENSP00000364204 Serine/threonine-protein kinase PINK1 Localized in mitochondria. Protects cells against stress-induced mitochondrial dysfunction by phosphorylating mitochondrial proteins. By means of activation and translocation of PRKN participates in the clearance of damaged mitochondria via selective autophagy (mitophagy).
ATP13A2 ENSP00000327214 Cation-transporting ATPase 13A2 ATPase involved in the transport of divalent transition metal cations and the maintenance of neuronal integrity. It is necessary for a correct lysosomal and mitochondrial maintenance.
PLA2G6 ENSP00000333142 85/88 kDa calcium-independent phospholipase A2 Involved in the release of fatty acids from phospholipids. Implicated in normal phospholipid remodeling. It has also been involved in NO- or vasopressin-induced arachidonic acid release and in leukotriene and prostaglandin production.
DNAJC6 ENSP00000360108 Putative tyrosine-protein phosphatase auxilin Promotes uncoating of clathrin-coated vesicles by recruiting HSPA8/HSC70 to clathrin-coated vesicles. Involved in clathrin-mediated endocytosis in neurons.
SYNJ1 ENSP00000409667 Synaptojanin-1 Phosphoinositide phosphatase, regulates levels of membrane phosphatidylinositol-4,5-bisphosphate (PIP2). Involved in the rearrangement of actin filaments downstream of tyrosine kinase and ASH/GRB2 by means of hydrolyzing PIP2 bound to actin regulatory proteins.
DJ-1/ PARK7 ENSP00000418770 Protein/nucleic acid deglycase DJ-1 Under an oxidative condition, via its chaperone activity, inhibits the aggregation of α-synuclein, thus functioning as a redox-sensitive chaperone and as a sensor for oxidative stress. Deglycates proteins and nucleotides, and the Maillard adducts formed between amino groups of proteins or nucleotides and reactive carbonyl groups of glyoxals.
FBXO7 ENSP00000266087 F-box only protein 7 Part of a SCF (SKP1-CUL1-F- box protein) E3 ubiquitin-protein ligase complex involved in protein ubiquitination. Role in the clearance of damaged mitochondria (mitophagy).
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Franco, R.; Rivas-Santisteban, R.; Navarro, G.; Pinna, A.; Reyes-Resina, I. Genes Implicated in Familial Parkinson’s Disease Provide a Dual Picture of Nigral Dopaminergic Neurodegeneration with Mitochondria Taking Center Stage. Int. J. Mol. Sci. 2021, 22, 4643. https://doi.org/10.3390/ijms22094643

AMA Style

Franco R, Rivas-Santisteban R, Navarro G, Pinna A, Reyes-Resina I. Genes Implicated in Familial Parkinson’s Disease Provide a Dual Picture of Nigral Dopaminergic Neurodegeneration with Mitochondria Taking Center Stage. International Journal of Molecular Sciences. 2021; 22(9):4643. https://doi.org/10.3390/ijms22094643

Chicago/Turabian Style

Franco, Rafael, Rafael Rivas-Santisteban, Gemma Navarro, Annalisa Pinna, and Irene Reyes-Resina. 2021. "Genes Implicated in Familial Parkinson’s Disease Provide a Dual Picture of Nigral Dopaminergic Neurodegeneration with Mitochondria Taking Center Stage" International Journal of Molecular Sciences 22, no. 9: 4643. https://doi.org/10.3390/ijms22094643

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop