Next Article in Journal
Enhancement of Methane Catalysis Rates in Methylosinus trichosporium OB3b
Next Article in Special Issue
Natural Products from Plants and Algae for Treatment of Alzheimer’s Disease: A Review
Previous Article in Journal
Differentiating Inhibitors of Closely Related Protein Kinases with Single- or Multi-Target Activity via Explainable Machine Learning and Feature Analysis
Previous Article in Special Issue
Tau Protein Modulates Perineuronal Extracellular Matrix Expression in the TauP301L-acan Mouse Model
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Parkin as a Molecular Bridge Linking Alzheimer’s and Parkinson’s Diseases?

by
Frédéric Checler
* and
Cristine Alves da Costa
*
IPMC, UMR7275 CNRS-UCA, INSERM, Labex DistALZ, 660 Route des Lucioles, 06560 Valbonne, France
*
Authors to whom correspondence should be addressed.
Biomolecules 2022, 12(4), 559; https://doi.org/10.3390/biom12040559
Submission received: 8 February 2022 / Revised: 4 April 2022 / Accepted: 7 April 2022 / Published: 9 April 2022

Abstract

:
Alzheimer’s (AD) and Parkinson’s (PD) diseases are two distinct age-related pathologies that are characterized by various common dysfunctions. They are referred to as proteinopathies characterized by ubiquitinated protein accumulation and aggregation. This accumulation is mainly due to altered lysosomal and proteasomal clearing processes and is generally accompanied by ER stress disturbance, autophagic and mitophagic defects, mitochondrial structure and function alterations and enhanced neuronal cell death. Genetic approaches aimed at identifying molecular triggers responsible for familial forms of AD or PD have helped to understand the etiology of their sporadic counterparts. It appears that several proteins thought to contribute to one of these pathologies are also likely to contribute to the other. One such protein is parkin (PK). Here, we will briefly describe anatomical lesions and genetic advances linked to AD and PD as well as the main cellular processes commonly affected in these pathologies. Further, we will focus on current studies suggesting that PK could well participate in AD and thereby act as a molecular bridge between these two pathologies. In particular, we will focus on the transcription factor function of PK and its newly described transcriptional targets that are directly related to AD- and PD-linked cellular defects.

Graphical Abstract

1. Alzheimer’s Disease in Brief

Alzheimer’s disease (AD) is the main age-related disease. Sporadic AD cases are characterized by a slow, progressive and irreversible degeneration, ultimately leading to mental disability and death [1,2]. The main macroscopic cerebral lesions consist in senile plaques, which are extracellular deposits mainly composed of amyloid β peptides (Aβ), and neurofibrillary tangles, which are intracellular lesions filled with hyperphosphorylated Tau proteins [3,4]. Additionally, exacerbated oxidative and ER stress/unfolded protein responses [5,6,7], alterations in mitochondrial structure, function and clearance (mitophagy) [8,9], autophagy [10,11], endolysosomal function [11,12,13,14] as well as synaptic loss [15,16,17] have been also observed at early stages in AD-affected brains. At later stages, although this has been discussed, apoptotic neuronal lesions and more particularly p53-dependent cell death have also been documented [18,19,20,21].
A few percent of AD cases are of familial origin and are due to mutations that segregate in an autosomal dominant manner [22,23]. The genes bearing these mutations have been identified and encode for the β-amyloid precursor protein (βAPP) and two family-related proteins referred to as presenilins 1 and 2 [24,25,26,27]. Genetic clues, anatomical stigmata and post-mortem tissue analyses have led to the proposal of the two main current etiological hypotheses. The hypothesis centered around Aβ, namely, the amyloid cascade hypothesis [28,29], proposes that Aβ accumulation is the early trigger of the degenerative process. This is strongly supported by the fact that mutations in βAPP (the precursor of Aβ) and in presenilins/γ-secretase (the enzyme releasing Aβ from βAPP [30,31]) affect the physiopathological processing of βAPP [32,33,34,35,36]. Alternatively, the tau-related hypothesis has been reinforced not only by the study of tau-related preclinical models [37] but also indirectly, by the consistent failure of pharmacological or immunological therapeutic strategies aimed at interfering with Aβ production or accumulation [38]. More recently, with the reconciliation of undoubtful genetic data and the failure of most Aβ-centric clinical trials, it has been proposed that additional proteases [39,40,41,42,43,44] and βAPP-related products distinct from Aβ could participate in the etiology of AD [40,41,45,46,47,48,49]. The latter postulate agrees well with the observations that some of these catabolites are indeed at the center of gravity of cellular dysfunctions observed early in AD [9,50,51,52].

2. Parkinson’s Disease in Brief

Parkinson’s disease is the second most frequent age-related pathology. PD is characterized by motor and non-motor symptoms [53,54,55]. It is a progressive and irreversible pathology characterized macroscopically by a drastic neuronal loss in the pars compacta of the substantia nigra, leading to an alteration in dopaminergic transmission [56,57,58,59]. At the histological level, PD is mainly characterized by intracellular inclusions named Lewy bodies that are mainly filled with aggregated α-synuclein [60,61]. PD is mainly idiopathic but there exist relatively rare genetic cases that could be due to either autosomal dominant or autosomal recessive transmission [62,63]. Monogenic forms due to a single mutation with Mendelian inheritance account for about 5–10 percent of PD cases.
More than fifteen chromosomal loci referred to as PARKs have been identified [64,65,66]. Of note, causative genes associated with a subset of loci are still pending definitive identification. Alternatively, the direct influence of some identified genes on the PD etiology remains to be definitely established. Confirmed genes responsible for the autosomal dominant form of PD have been identified as PARK1 (the SNCA gene encoding α-synuclein [67,68,69,70], PARK8 (LRRK2 [71]), PARK13 (HTRA2 [72]) and PARK17 (VPR35 [73]). Autosomal recessive genes include PARK2 (Parkin [74,75,76]), PARK6 (PINK1 [77,78,79]), PARK7 (DJ1 [80]) and PARK9 (ATP13A2 [81]). Known functions of the above proteins clearly indicate that the etiology of PD is complex, although they point to ER stress [82,83], mitophagy [84,85], autophagy [86,87,88] and mitochondrial [89] and lysosomal [90,91,92,93] defects, as well as neuronal death exacerbation [94,95,96].

3. Alzheimer’s and Parkinson’s Diseases: Two Distinct Pathologies with Common Dysfunctions

3.1. Proteasomal Dysfunction

As stated above, AD and PD are proteinopathies, i.e., pathologies in which several generally misfolded proteins accumulate and are poorly cleared. On the whole, it has been reported that, whatever the nature of the aggregate components, they are all highly toxic [97]. This reduced propensity to proteolytic degradation is directly related to the drastic alteration of the ubiquitin–proteasomal system triggered by aggregates [98]. The proteasomal machinery, which underlies one of the major cellular degradative pathways, is altered in AD [99,100]. This conclusion has been supported by the observation of accumulated ubiquitin in both senile plaques and neurofibrillary tangles [101] and by the fact that proteasomal activity was reduced upon aging in an AD mouse model [102]. Indeed, it was demonstrated that Aβ peptides physically interact with the proteasome and inhibit its activity [103]. Accordingly, Aβ load is inversely correlated with proteasome activity in AD [104].
Several studies also reported proteasomal defects in PD [105,106] which lead to the disruption of α-synuclein clearance [107,108]. The key role of proteasomal degradation is indirectly supported by the fact that parkin acts as a ubiquitin-ligase [109] and promotes the degradation of proteins involved in mitochondrial structure [110] and autophagic processes [111] (see below).

3.2. Oxidative Stress Dysfunction

Aberrant production of oxidated proteins, lipids or nucleic acids and/or defects in cellular anti-oxidant defenses occurring during aging or in pathological conditions [112,113] have been consistently observed in AD and PD [114]. Proteins, lipids, DNA and carbohydrates are increased in the AD brain and advanced glycation end-products have been described in plaques [115]. Data concerning the cellular antioxidant portfolio, including several catalases, reductases and peroxidases, appear to show significant decreases in terms of both mRNA levels [116] and activities [117].
Excessive oxidative stress has also been documented in PD [118,119,120]. Particularly compelling was the recent systematic review indicating that oxidative stress markers could be recovered in cerebrospinal fluid and blood in PD-affected patients [120]. Of interest, this study reported a significant decrease in enzymatic and non-enzymatic antioxidants [120].

3.3. ER Stress and Unfolded Protein Response

The endoplasmic reticulum is where protein biogenesis and post-translational and structural modifications occur [121]. When neo-synthesized proteins are not correctly folded, they normally undergo proteasomal-mediated clearance via a cellular process referred to as ER-associated degradation (ERAD) [122,123]. This cellular program is linked to the unfolded protein response (UPR) that is directly initiated by ER stress [124] and involves three cellular pathways involving either pancreatic ER kinase (PERK1), activating transcription factor 6 (ATF6) or inositol-requiring enzyme-1 (IRE1α) [125]. Thus, in neurodegenerative diseases, accumulated toxic proteins could derive from combined exacerbated misfolding and proteasomal defects [126,127,128].
Activations of ER stress and UPR have been extensively documented in AD [6,7,17,129]. Thus, elevated ER stress response has been observed in APP/PS1 double transgenic mice as indicated by enhanced levels of GRP78 and phosphorylated forms of ERK and eIF2a [130]. Further, deletion of endogenous PERK prevents synaptic defects and cognitive alterations in APP/PS1 mice [131]. It should be noted that these alterations were not observed in APP Ki Tg mice that harbor a single mutation in APP [132]. However, in support of a genuine relationship between AD-related stigmata and ER stress/UPR alterations, it has been shown that intracerebral administration of Aβ42 alters the ER stress-associated proteins GRP78 and eIF2α in adult mouse brains [133]. Further, several studies reported ER stress-mediated effects of Aβ oligomers in various cellular models and AD brain samples [134,135,136]. Importantly, ruling out the possibility of an artifactual phenotype linked to overexpression, Aβ also accumulates in the ER and triggers UPR in iPSCs derived from sporadic and familial AD differentiated into neural cells [137]. Finally, it has been demonstrated that Tau blocks the ERAD pathway [138] and that the UPR is activated in pre-tangles in the AD hippocampus [5]. Although far from being exhaustive, these studies strongly suggest ER stress/UPR exacerbation in AD.
ER stress is also drastically altered in PD [83,139,140,141]. Several proteins responsible for familial PD have been documented as effectors of the ER stress machinery. α-Synuclein physically interacts with ER chaperones such as GRP78 [142] and phosphorylated α-synuclein perturbs ER–Golgi trafficking by altering several ER exit factors such as RAB1 and SNARES [143]. Further, in dopaminergic neurons, α-synuclein activates PERK and ATF6 pathways, while, conversely, GRP78 overexpression rescues α-synuclein-associated toxicity by elevating tyrosine hydroxylase and restoring dopamine levels in the substantia nigra [144].
ER stress also increases parkin protein and mRNA levels and the ER stress-mediated increase in parkin levels is prevented by ATF4 deletion in cells as well as in mice, likely because it functionally impairs ATF4 binding to the parkin promoter region [145]. Further, parkin overexpression protects against ER stress, while, conversely, parkin depletion sensitizes cells to ER stress [146]. It has been proposed that parkin could control ER stress response via its ubiquitin-ligase activity. Thus, Imai and colleagues showed that the parkin-associated endothelin receptor-like receptor (PAEL-R) behaves as a parkin substrate. An ubiquitinated insoluble form of PAEL-R occurs in familial PD-affected brains and its accumulation is prevented by parkin-mediated degradation [147].
XBP1s is a transcription factor that results from a stress-induced non-conventional splicing of XBP1 by Ire1α [148]. It has been recently demonstrated that there exists a functional interplay between XBP1s and PINK1. Thus, XBP1s activates the transcription of PINK1 in various cellular models, including primary cultured neurons, as well as in mouse brains [149], and thereby triggers a pro-mitophagic phenotype that was fully abolished by PINK1 depletion [149]. On the other hand, PINK1 was shown to phosphorylate XBP1s and modulate its transcriptional activity by governing its shuttling to the nucleus [149]. Of importance, PINK1-induced phosphorylation of XBP1s occurs at sites reminiscent of those phosphorylated in the substantia nigra of sporadic PD-affected brains [149].
A few papers concern LRRK2 that is mutated in autosomal dominant forms of PD (see above). LRRK2 is localized at the ER, suggesting a possible contribution to the control of ER stress response. Indeed, in C-elegans, LRRK2 protects dopaminergic neurons from 6-hydroxydopamine-associated toxicity, likely by promoting GRP78 enhanced expression [150].

3.4. Mitochondria Dysfunction

Most oxygen is consumed by mitochondria in the cytochrome oxygen chain. As stated above, in aging and neurodegenerative conditions, it is acknowledged that reactive oxygen species (ROS) formation is increased. Thus, there exists a loop wherein mitochondria are both producers and targets of ROS. In neurodegenerative conditions, it is therefore not surprising that reports have consistently indicated drastic alterations in mitochondrial structure and function. This has been particularly well documented in recent reviews [151,152,153,154].
Mitochondria dysfunctions are early and prominent alterations in the brains of patients with AD [9,155]. A bioinformatic analysis indicated that the mitochondrial oxidative phosphorylation system (OXPHOS) pathway was consistently altered in AD [156]. Particularly, a reduction of complex I of OXPHOS was observed in the hippocampus of AD-affected patients [157]. Recently, it was documented that mitochondria size alterations and cristae disorganization were observed together with increased ROS species production in AD cell models [9]. This was confirmed in various AD transgenic mice [9]. On the whole, AD is associated with mitochondrial defects that affect their biogenesis and dynamics, their transport, their contact with the endoplasmic reticulum [154,158,159] and their clearance by mitophagy (see below).
Mitochondrial dysfunction is also a feature of PD [89,160]. Multiple pathways directly related to mitochondria have been documented [161] that include increased ROS production [162], abnormally elevated intracellular calcium concentrations and reduced ATP formation [163,164,165]. Indeed, most of the proteins responsible for both autosomal and recessive cases of PD trigger drastic mitochondrial alterations. Amongst myriad works documenting these alterations, one could notice that DJ1 downregulation increases mitochondrial fragmentation and reduces mitochondrial potential [166]. LRRK2 regulates mitochondrial dynamics [167] and its pathogenic mutations impair the ability of MIRO to stop mitochondrial damage [168]. α-Synuclein is partly localized in mitochondria in PD-affected post-mortem brains [169] and disrupts mitochondrial function by increasing ROS [170]. Finally, the PINK1–parkin axis is recognized as the major system aimed at controlling mitochondrial homeostasis by mitophagy [171].

3.5. Autophagy/Mitophagy

Autophagy is a key cellular process by which cellular homeostasis is maintained. Mitophagy is an autophagic process that concerns the control of mitochondrial function and quantity. Homeostatic mitochondrial equilibrium between biogenesis and clearance by mitophagy is necessary for a wide spectrum of intracellular functions, including, among others, embryogenic development, cellular differentiation and death, as well as inflammation. In mammalian cells, it is admitted that the main pathway to regulate mitochondrial homeostasis involves PINK1 and parkin cooperation aimed at selectively clearing defective mitochondria [172,173].
As expected from the key role of mitochondria, mitophagy dysfunction is at the center of gravity of neurodegenerative diseases and mitophagy defects have been well documented in AD [174,175,176]. It has been shown that mitophagy reversed Aβ- and Tau-induced memory defects in C. elegans and that it lowered the load of insoluble Aβ40 and Aβ42 in a mouse model (APP/PS1) of AD [177]. Conversely, Aβ and phosphorylated Tau trigger defective mitophagy in an age-dependent manner, resulting in a severe augmentation of the number of mitochondria accompanied by a significant reduction in their mean size [178]. Besides canonical Aβ, additional fragments derived from APP have been shown to trigger drastic mitochondrial dysfunctions. Thus, the β-secretase-derived APP-CTF (C99) was recently shown to affect mitophagy in both cell and animal AD models, as well as in sporadic AD brains, as illustrated by mitochondrial size alteration and cristae disorganization in neuroblastoma cells [9]. Accordingly, C99 accumulation triggers mitophagy failure, as shown by the alteration of canonical proteins involved in mitophagy. Thus, enhanced conversion and accumulation of LC3, resistance to degradation of p62 and altered PINK1-mediated recruitment of parkin in mitochondria have been documented [9]. These cellular observations were confirmed in young 3xTg-AD mice (a widely used mouse model [179]) and in AAV-C99-infected mice [9]. These alterations were also reported in sporadic AD brain samples [9]. Of importance, a very recent study confirmed this set of data in induced neural stem cells (iNSCs) derived from AD-affected patient fibroblasts [180]. In iNSCs, C99 accumulates in mitochondrial domains and triggers mitophagy failure, as illustrated by increased LC3II and p62 proteins as well as by altered PINK1-mediated parkin recruitment to mitochondria [180].
As parkin and PINK1 are two proteins mutations in which account for a subset of autosomal recessive cases of PD, it is not surprising that mitophagy dysfunction is also a key cellular process affected in PD [84,85,181,182]. PINK1 accumulates at the outer membrane of dysfunctional mitochondria and recruits parkin which, in turn, ubiquitinates several proteins, including ubiquitin, the ubiquitination of which is central to autophagosome formation. This function in mitochondrial quality control results in poorly detectable levels of PINK1 due to a continuous import/degradation process. This virtuous circle is impacted in PD. A rate-limiting fission step in the mitophagy process implies that parkin–PINK 1 interplay should control the levels of inner and outer mitochondrial membrane proteins. In this context, it is noteworthy that mitofusins 1 and 2 are ubiquitinated in a parkin/PINK1-dependent manner [183,184] and that PINK1 displays the ability to phosphorylate dynamin-related-protein 1 (Drp-1) [185]. Several mechanistic explanations have been provided to link PINK1 with mitochondrial defects. One of the most common hypotheses proposes that increased fission and mitophagy could be accounted for by the ability of PINK1 to phosphorylate Protein kinase A. This kinase phosphorylates and activates Drp-1 at its serine 656 residue and a constitutively dephosphorylated Drp1 mutant (Ser- > Ala 656) triggers mitochondrial fragmentation and increased cell vulnerability [186]. Thus, a PD-linked loss of function of PINK1 could increase the dephosphorylated form of Drp-1 and thus increase fission and mitophagy [187,188]. Another interesting study indicated that, in contrast to previous anticipations, parkin could also act upstream to PINK1. Thus, Goiran and colleagues showed that parkin controlled PINK1 transcription via a FOXO3a-dependent mechanism in various transgenic and knock-out mice [189]. Thus, pathogenic mutations in both parkin and PINK1 could have a direct role in the alteration of the mitophagic process that takes place in PD.

3.6. Cell Death

Several types of cell death can occur. Necroptosis is a form of necrosis leading to unprogrammed cell death [190]. Programmed cell death, also named apoptosis, is a normal process that allows cellular homeostasis and the destruction of damaged cells. When apoptosis is exacerbated, neurodegeneration triggers neuronal loss and subsequent cerebral defects. The extent of apoptosis varies according to the brain pathology examined. It has been convincingly demonstrated in PD [95,191]. Among a bulk of studies, p53 has been documented as the prominent contributor to cell death in various degenerative diseases [20,192], including PD [193,194]. Thus, it has been shown that p53-like immunoreactivity is increased in PD-affected brains [195] and that p53 contributes to 6-hydroxydopamine-induced cell death, an experimental model of PD-like pathology [196,197,198]. Furthermore, selective depletion of p53 in dopaminergic neurons protects against neuronal loss [199]. It should be noted that most proteins responsible for familial cases of PD interact functionally with p53. α- and β-synucleins lower p53-dependent apoptosis [94,200,201], a phenotype abolished by 6-hydroxydopamine [108] while, p53 up-regulates α-synuclein transcription [202] and promotes its aggregation [203], thus underlining a functional interplay between α-synuclein and p53 [193]. Parkin can repress p53 transcription and this is abolished by PD-related mutations [204]. Further, parkin translocation to the nucleus and physical interaction with the p53 promoter are prevented by S-nitrosylation [205], a post-translational process exacerbated in PD-affected brains [206]. As is the case for α-synuclein, parkin also behaves as a transcriptional target of p53 [207,208]. DJ1 prevents p53-induced mitochondrial impairment [209] and is regulated in ER stress conditions by a molecular cascade involving parkin, p53 and the transcription factor XBP1s [210]. Finally, PINK1 behaves as a repressed transcriptional target of nuclear p53 [211], while, conversely, PINK1 can directly bind to and phosphorylate p53 and thereby control the autophagic process in hepatic cancer stem cells [212].
Apoptosis in AD is less consensual [213,214]. However as is the case with PD, p53 appears to play a key role in AD-related cell death stigmata [215]. Thus, p53 expression is increased in AD brains [216], correlates with the age of patients [217] and undergoes several AD-linked post-translational and conformational modifications [217,218]. Also important, in AD, apoptotic neurons display intracellular Aβ [19], which can regulate p53 promoter transactivation [219]. Further, there exists an intricate network of functional interactions between p53 and members of the γ-secretase complex [220]. Presenilins [221] and its presenilinase-derived C-terminal fragment [222] control p53-dependent cell death in AD. This phenotype is exacerbated by AD-related mutations [223]. Other components of the γ-secretase complex, namely, Pen-2, Aph-1 and nicastrin [224,225], all control p53-dependent cell death in either presenilins-dependent [226] or independent [227] processes. Finally, p53 also regulates the expression of βAPP [228]. Conversely, various APP-derived C-terminal fragments derived from γ-secretase [229] (AICD) or caspase 3 [230] (C31) cleavages can induce enhanced p53 promoter transcription [221,231] and translation [232] or toxicity [233,234].

4. Parkin, a Molecular Link between Alzheimer’s and Parkinson’s Diseases?

Parkin has been characterized as a pleiotropic protein involved in multiple cellular functions [76]. Since it was initially described as a ubiquitin-ligase [109] and thus as being committed to feeding the proteasomal machinery with ubiquitinated unfolded/misfolded proteins, parkin has been proposed as a key regulator of proteinopathies characterizing most neurodegenerative diseases [235,236]. Since parkin mutations are generally responsible for autosomal recessive familial cases of PD, particular attention has been paid to delineating parkin-associated dysfunction in this pathology.
However, several works have indicated that parkin could also contribute to AD. First, a macroscopic analysis of AD-related lesions indicated that parkin co-localized with both senile plaques and Aβ-related vascular lesions. Further, parkin expression also appears to be high in astrocytes surrounding both lesions [237]. These observations per se were obviously not sufficient to postulate a key role for parkin in AD, and the mechanistic explanation underlying these anatomical observations was initially lacking. According to the main ubiquitin-ligase function of parkin, one could envision that parkin could have a protective function with respect to AD-related triggers, mainly Aβ peptides and tau proteins, by governing their clearance via enhanced proteasomal-driven degradation [238] or by mitophagy [239].
Several studies indicated that parkin-mediated mitophagy could be altered in AD and that parkin manipulation could potentially rescue AD-like defects. First, Ye and colleagues reported a mitophagic dysfunction in cultured neurons expressing FAD-linked APP mutations as well as in AD-affected brains of patients and that gradual depletion of cytosolic parkin occurs as AD progresses [240]. In agreement with this, the activation of parkin-mediated mitophagy by rapamycin was found to be able to improve cognitive and synaptic plasticity in APP/PS1 mice [241]. Further, Martin-Maestro and colleagues demonstrated that in skin fibroblasts of sporadic AD-affected patients parkin expression was diminished and was accompanied by impaired mitophagy. Of note, parkin overexpression was able to rescue mitophagy failure and lowered ubiquitinated protein loads in these cells [242].
The above studies emphasized the role of parkin in mitophagic defects taking place in AD but did not delineate the precise molecular AD-related targets underlying this dysfunction. A series of studies indicated that both Aβ and Tau could alter parkin-mediated mitophagy and that, conversely, manipulating the mitophagic process could alleviate Aβ- and Tau-related toxicity. First, mitophagy blocks Aβ- and Tau-related pathology in iPSC-derived human neurons as well as in C-elegans and mouse models of AD [177]. Interestingly, parkin overexpression restores mitophagy and thereby lowers Aβ-related mitochondrial defects in human cells [243]. In agreement with this, parkin was shown to clear defective mitochondria and ubiquitinated Aβ in the widely used 3xTg-AD triple transgenic mouse model [244]. Further, parkin was shown to prevent cortical atrophy and Aβ-linked brain metabolism defects in this mouse model [245] and reverse intracellular Aβ accumulation in human neuroblastomas and primary cultured neurons [246]. Of utmost importance, parkin overexpression improves hippocampal long-term potentiation and lowers Aβ load in APP/PS1 transgenic mice crossed with mice overexpressing parkin [247].
These above-described models are often overexpressing systems. In order to preclude erroneous conclusions based on an overload of parkin, it was important to note that depletion of endogenous parkin led to the same conclusions. Thus, parkin knock-out cells are more sensitive to intracellular Aβ-induced toxicity [248]. On the other hand, Aβ oligomers were shown to reduce the mitochondrial GTPase Miro1 and trigger autophagy [249], while neurons prepared from parkin-null mice remain resistant to Aβ42-induced toxicity [250]. This set of data altogether indicates an intricate interplay between parkin and Aβ by which parkin could protect cells against Aβ-induced toxicity, while Aβ itself could interfere with parkin-mediated mitophagy.
A parkin-associated phenotype protective against Aβ toxicity has also been indirectly confirmed by pharmacological approaches in various AD-related models. Thus, the neuroprotective vegetal compound β-asarone improved learning and memory in Aβ42-injected rats by increasing PINK1-parkin expressions and lowering Aβ42 loads [251]. This agreed with the decrease in Aβ and augmented autophagy observed in a PC12 cell model [252]. In the same cell line, Panax notoginseng saponins protect against Aβ toxicity by promoting parkin-mediated mitophagy [253]. Finally, nilotinib, an inhibitor of Abl tyrosine kinase, triggers increased autophagic and endogenous parkin levels which are accompanied by Aβ clearance [254].
There exists a network of evidence also indicating that Tau proteins could affect parkin expression and function and that, conversely, endogenous parkin could control Tau. So, intracellular accumulation of Tau, which corresponds to one of the anatomical stigmata in AD brains, is concomitant to mitophagy deficits, as illustrated by increased mitophagy markers. The direct link between these two observations was forged by the demonstration that the overexpression of human Tau induced mitophagy defects in human cells, primary cultured neurons and mouse brains. These changes were accompanied by increased mitochondrial membrane potential and, even more interestingly, by decreased levels of parkin and PINK1 [255]. These observations were corroborated by Cummins and colleagues who confirmed the ability of wild-type as well as a mutant Tau (P301L) to impair mitophagy in neuroblastomas by affecting parkin recruitment to defective mitochondria [256].
Parkin was shown to counteract the alteration of wild-type Tau expression and hyperphosphorylation in human neuroblastoma cells [257]. This could be a general phenotype aspecifically linked to most tauopathies. However, the fact that parkin-associated modification of Tau occurs in the presence of Aβ42 [257] and the recent demonstration that parkin reductions in TS65DN mice (a mouse model of down syndrome [258] in which a triplication of chromosome 21 encoding βAPP occurs in humans) occur before Tau hyperphosphorylation [259] argues in favor of a more selective participation of parkin in Tau modifications taking place in AD. This was corroborated by a series of studies in which the contributions of endogenous parkin to Tau-related pathologies were reported. Thus, in a mouse model in which overexpression of human mutated Tau was combined with parkin gene deletion, severe neuropathological alterations were observed, including parkin-dependent selective expression of the mutated Tau transgene in neuronal cell bodies, increased Tau phosphorylation and cerebral atrophy [260]. In this mouse model, the deletion of parkin enhances neuronal lesions in the substantia nigra [261], causes cerebral amyloidosis [262] and triggers aggregation of hyperphophorylated Tau in the cortex and hippocampus [263].
Overall, the above-described data strongly support a significant contribution of parkin to AD pathology (Figure 1). This is based on consistent observations derived from multiple cells, animal models and AD-affected brains. Although distinct, these various approaches genuinely identify parkin-mediated contributions to AD, as was supported by a very recent and important study which used a transcriptomic approach to examine the molecular signatures associated with various models of AD. De Bastiani and colleagues analyzed putative analogies and discrepancies between the transcriptomes of three mouse models, namely, 5xFAD, APP/PS1 and human Aβ knocked-in, as well as those of early- and late-onset sporadic cases of AD (De Bastiani et al. bioRxiv, https://doi.org/10.1101/2021.06.09.447404). By means of a regulatory network-based approach aimed at identifying common master regulators (MRs), the authors showed that 5xFAD and APP/PS1 mice shared more MRs with early-onset than with late-onset AD cases, while the hAβ-KI mouse profile did not discriminate between early and late AD cases. However, when considering the 17 MRs overlapping in more than 4 models, only SOX9 and PARK2 were altered across animal/human models. This agreed well with a previous transcriptional analysis delineating PARK2 as a MR in the human AD brain hippocampus [264]. Of note, in De Bastiani et al.’s study, PARK2 appeared to be repressed, thus suggesting a lowering of parkin expression/activity in AD (De Bastiani et al. bioRxiv, https://doi.org/10.1101/2021.06.09.447404). This could be interpreted in several ways. At first sight, the lowering of parkin/ubiquitin ligase activity could be in agreement with decreased Aβ/Tau degradation in AD (see above). On the other hand, parkin is also a transcription factor [204] that can up-regulate presenilin1/γ-secretase [265] and thereby increase Aβ production. Thus, the repression of parkin documented by De Bastiani and colleagues would paradoxically lower γ-secretase-mediated Aβ production. This is not antinomic with the pathogenic process taking place in AD. Thus, we showed that the precursor of Aβ (C99) is very toxic [52], affects many of the cellular pathways altered in AD (see above) [9,14], occurs very early in mice [266] and accumulates in vulnerable neurons in the human brain [267]. Accordingly, we showed that γ-secretase inhibitors potentiate the recovery of intracellular C99 [268] and proposed γ-secretase as a beneficial C99 inactivating enzyme [38]. In this context, the consistent repression of PARK2 and its associated decrease in presenilin1/γ-secretase expression/activity observed in all models examined by De Bastiani and colleagues would yield enhanced levels of C99, in agreement with our C99-centric etiological hypothesis for AD.
The contribution of parkin’s transcriptional function to AD pathology has been further supported by several studies. First, as stated above, besides its ubiquitin-ligase activity, parkin acts as transcription factor [204,269] and up-regulates presenilin 1/γ-secretase [265]. γ-secretase-mediated proteolysis of C99 not only generates Aβ but also the APP intracellular domain (AICD) that acts as a transcription factor [229] able to modulate the expression of several key proteins directly or indirectly contributing to AD pathology [221,231,270,271]. We showed that parkin controls PINK1-linked mitophagy via a presenilin-mediated pathway involving an AICD-FOXO1 functional interaction [189]. Further, it has been shown that the transcription factor XBP1s acts as a key contributor to memory and cognitive functions in AD [272]. Thus, XBP1s is able to rescue hippocampal synaptic plasticity alterations and memory defects in mouse models of AD [273]. As we showed that parkin can control XBP1s activity in a p53-dependent manner [210], one can envision that parkin-mediated improvement in AD-related cognitive defects could, at least partly, involve the control of the XBP1s pathway by parkin.

5. Future Perspectives

Besides its well documented contribution to PD, there exists a consistent network of indirect and direct anatomical and functional evidence suggesting that parkin could well contribute to AD pathology. This protein adds to the general picture that proteinopathies harbor common cellular defects and that studying a protein thought to contribute to a given neurodegenerative disease could help to understand another brain pathology. As a corollary, this should open putative novel therapeutic tracks. Sticking to this proposal, parkin could well be envisioned as an additional target to fight the onset and/or progression of AD.

Author Contributions

Both F.C. and C.A.d.C. contributed to the writing, review and editing of the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the Labex DISTALZ (Development of Innovative Strategies for a Translational Approach of Alzheimer’s disease; ANR-11LABX-0009), by Fondation de France (WB-2020-27248-Maladie de Parkinson-00107076) and by the ANR (grant ANR-20-CE16-008).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

ADAlzheimer’s disease
PDParkinson’s disease
EREndoplasmic reticulum
PINK1Phosphatase and TENsin-induced putative kinase 1
XBP1sSpliced form of X-box binding protein 1
Ab peptidesAmyloid b peptides
PARKDesignated genes involved in familial PD
HTRA2High temperature requirement protein A2
LRRK2Leucine rich repeat kinase 2
ATP13A2ATPase type 13A2
ERADER-associated degradation
UPRUnfolded protein response
ATF6Activating transcription factor 6
PERKPancreatic ER kinase
GRP7878 Kd Glucose-regulated protein
(IRE1a)Inositol-requiring enzyme-1 alpha
eIF2 aEukaryotic initiation factor 2 alpha
iPSCsInducible pluripotent stem cells
PAEL-RParkin-associated endothelin receptor-like receptor
OXPHOSMitochondrial oxidative phosphorylation system
(ROS)Reactive oxygen species
MIROMitochondrial Rho
(Drp-1)Dynamin-related-protein 1
FOXO3aForkhead box O3
PEN-2Presenilin enhancer 2
APH-1Anterior pharynx-defective 1

References

  1. Selkoe, D.J. Alzheimer’s disease. Cold Spring Harb. Perspect. Biol. 2011, 3, 1–16. [Google Scholar] [CrossRef] [PubMed]
  2. Knopman, D.S.; Amieva, H.; Petersen, R.C.; Chetelat, G.; Holtzman, D.M.; Hyman, B.T.; Nixon, R.A.; Jones, D.T. Alzheimer disease. Nat. Rev. Dis. Primers 2021, 7, 33. [Google Scholar] [CrossRef] [PubMed]
  3. Serrano-Pozo, A.; Frosch, M.P.; Masliah, E.; Hyman, B.T. Neuropathological alterations in Alzheimer disease. Cold Spring Harb. Perspect. Med. 2011, 1, a006189. [Google Scholar] [CrossRef] [PubMed]
  4. DeTure, M.A.; Dickson, D.W. The neuropathological diagnosis of Alzheimer’s disease. Mol. Neurodegener. 2019, 14, 32. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Hoozemans, J.J.; van Haastert, E.S.; Nijholt, D.A.; Rozemuller, A.J.; Eikelenboom, P.; Scheper, W. The unfolded protein response is activated in pretangle neurons in Alzheimer’s disease hippocampus. Am. J. Pathol. 2009, 174, 1241–1251. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Uddin, M.S.; Tewari, D.; Sharma, G.; Kabir, M.T.; Barreto, G.E.; Bin-Jumah, M.N.; Perveen, A.; Abdel-Daim, M.M.; Ashraf, G.M. Molecular Mechanisms of ER Stress and UPR in the Pathogenesis of Alzheimer’s Disease. Mol. Neurobiol. 2020, 57, 2902–2919. [Google Scholar] [CrossRef]
  7. Huang, H.C.; Tang, D.; Lu, S.Y.; Jiang, Z.F. Endoplasmic reticulum stress as a novel neuronal mediator in Alzheimer’s disease. Neurol. Res. 2015, 37, 366–374. [Google Scholar] [CrossRef]
  8. Perez Ortiz, J.M.; Swerdlow, R.H. Mitochondrial dysfunction in Alzheimer’s disease: Role in pathogenesis and novel therapeutic opportunities. Br. J. Pharmacol. 2019, 176, 3489–3507. [Google Scholar] [CrossRef]
  9. Vaillant-Beuchot, L.; Mary, A.; Pardossi-Piquard, R.; Bourgeois, A.; Lauritzen, I.; Eysert, F.; Kinoshita, P.F.; Cazareth, J.; Badot, C.; Fragaki, K.; et al. Accumulation of amyloid precursor protein C-terminal fragments triggers mitochondrial structure, function, and mitophagy defects in Alzheimer’s disease models and human brains. Acta Neuropathol. 2021, 141, 39–65. [Google Scholar] [CrossRef]
  10. Nixon, R.A.; Wegiel, J.; Kumar, A.; Yu, W.H.; Peterhoff, C.; Cataldo, A.; Cuervo, A.M. Extensive involvement of autophagy in Alzheimer disease: An immuno-electron microscopy study. J. Neuropathol. Exp. Neurol. 2005, 64, 113–122. [Google Scholar] [CrossRef] [Green Version]
  11. Hung, C.; Livesey, F.J. Endolysosome and autophagy dysfunction in Alzheimer disease. Autophagy 2021, 17, 3882–3883. [Google Scholar] [CrossRef] [PubMed]
  12. Nixon, R.A. Amyloid precursor protein and endosomal-lysosomal dysfunction in Alzheimer’s disease: Inseparable partners in a multifactorial disease. FASEB J. 2017, 31, 2729–2743. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. van Weering, J.R.T.; Scheper, W. Endolysosome and Autolysosome Dysfunction in Alzheimer’s Disease: Where Intracellular and Extracellular Meet. CNS Drugs 2019, 33, 639–648. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Lauritzen, I.; Pardossi-Piquard, R.; Bourgeois, A.; Pagnotta, S.; Biferi, M.G.; Barkats, M.; Lacor, P.; Klein, W.; Bauer, C.; Checler, F. Intraneuronal aggregation of the beta-CTF fragment of APP (C99) induces Abeta-independent lysosomal-autophagic pathology. Acta Neuropathol. 2016, 132, 257–276. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Subramanian, J.; Savage, J.C.; Tremblay, M.E. Synaptic Loss in Alzheimer’s Disease: Mechanistic Insights Provided by Two-Photon in vivo Imaging of Transgenic Mouse Models. Front. Cell Neurosci. 2020, 14, 592607. [Google Scholar] [CrossRef]
  16. Colom-Cadena, M.; Spires-Jones, T.; Zetterberg, H.; Blennow, K.; Caggiano, A.; DeKosky, S.T.; Fillit, H.; Harrison, J.E.; Schneider, L.S.; Scheltens, P.; et al. The clinical promise of biomarkers of synapse damage or loss in Alzheimer’s disease. Alzheimer’s Res. Ther. 2020, 12, 21. [Google Scholar] [CrossRef]
  17. Lee, J.H.; Won, S.M.; Suh, J.; Son, S.J.; Moon, G.J.; Park, U.J.; Gwag, B.J. Induction of the unfolded protein response and cell death pathway in Alzheimer’s disease, but not in aged Tg2576 mice. Exp. Mol. Med. 2010, 42, 386–394. [Google Scholar] [CrossRef] [Green Version]
  18. Dickson, D.W. Apoptotic mechanisms in Alzheimer neurofibrillary degeneration: Cause or effect? J. Clin. Investig. 2004, 114, 23–27. [Google Scholar] [CrossRef] [Green Version]
  19. Chui, D.H.; Dobo, E.; Makifuchi, T.; Akiyama, H.; Kawakatsu, S.; Petit, A.; Checler, F.; Araki, W.; Takahashi, K.; Tabira, T. Apoptotic neurons in Alzheimer’s disease frequently show intracellular Abeta42 labeling. J. Alzheimer’s Dis. 2001, 3, 231–239. [Google Scholar] [CrossRef]
  20. Checler, F.; Alves da Costa, C. p53 in neurodegenerative diseases and brain cancers. Pharmacol. Ther. 2014, 142, 99–113. [Google Scholar] [CrossRef]
  21. Tajbakhsh, A.; Read, M.; Barreto, G.E.; Avila-Rodriguez, M.; Gheibi-Hayat, S.M.; Sahebkar, A. Apoptotic neurons and amyloid-beta clearance by phagocytosis in Alzheimer’s disease: Pathological mechanisms and therapeutic outlooks. Eur. J. Pharmacol. 2021, 895, 173873. [Google Scholar] [CrossRef] [PubMed]
  22. St George-Hyslop, P.H.; Tanzi, R.E.; Polinsky, R.J.; Haines, J.L.; Nee, L.; Watkins, P.c.; Myers, R.H.; Feldman, R.G.; Pollen, D.; Drachman, D.; et al. The genetic defect causing familial Alzheimer’s disease maps on chromosome 21. Science 1987, 235, 885–890. [Google Scholar] [CrossRef] [PubMed]
  23. Bertram, L.; Tanzi, R.E. The genetics of Alzheimer’s disease. Prog. Mol. Biol. Transl. Sci. 2012, 107, 79–100. [Google Scholar] [CrossRef] [PubMed]
  24. St George-Hyslop, P.; Haines, J.; Rogaev, E.; Mortilla, M.; Vaula, G.; Pericak-Vance, M.; Foncin, J.F.; Montesi, M.; Bruni, A.; Sorbi, S.; et al. Genetic evidence for a novel fami!lial Alzheimer’s disease locus on chromosome 14. Nat. Genet. 1992, 2, 330–334. [Google Scholar] [CrossRef]
  25. Rogaev, E.I.; Sherrington, R.; Rogaeva, E.A.; Levesque, G.; Ikeda, M.; Liang, Y.; Chi, H.; Lin, C.; Holman, K.; Tsuda, T.; et al. Familial Alzheimer’s disease in kindreds with missense mutations in a gene on chromosome 1 related to the Alzheimer’s disease type 3 gene. Nature 1995, 376, 775–778. [Google Scholar] [CrossRef]
  26. Levy-Lahad, E.; Wijsman, E.M.; Nemens, E.; Anderson, L.; Goddard, A.B.; Weber, J.L.; Bird, T.D.; Schellenberg, G.D. A familial Alzheimer’s disease locus on chromosome 1. Science 1995, 269, 970–973. [Google Scholar] [CrossRef]
  27. Sherrington, R.; Rogaev, E.I.; Liang, Y.; Rogaeva, E.A.; Levesque, G.; Ikeda, M.; Chi, H.; Lin, C.; Li, G.; Holman, K.; et al. Cloning of a gene bearing missense mutations in early-onset familial Alzheimer’s disease. Nature 1995, 375, 754–760. [Google Scholar] [CrossRef]
  28. Hardy, J.A.; Higgins, G.A. Alzheimer’s disease: The amyloid cascade hypothesis. Science 1992, 256, 184–185. [Google Scholar] [CrossRef]
  29. Tanzi, R.E.; Bertram, L. Twenty years of the Alzheimer’s disease amyloid hypothesis: A genetic approach. Cell 2005, 120, 545–555. [Google Scholar] [CrossRef]
  30. De Strooper, B.; Saftig, P.; Craessaerts, K.; Vanderstichele, H.; Guhde, G.; Annaert, W.; Von Figura, K.; Van Leuven, F. Deficiency of presenilin-1 inhibits the normal cleavage of amyloid precursor protein. Nature 1998, 391, 387–390. [Google Scholar] [CrossRef]
  31. Herreman, A.; Serneels, L.; Annaert, W.; Collen, D.; Schoonjans, L.; De Strooper, B. Total inactivation of g-secretase activity in presenilin-deficient embryonic stem cells. Nat. Cell. Biol. 2000, 2, 461–462. [Google Scholar] [CrossRef] [PubMed]
  32. Thinakaran, G. The role of presenilins in Alzheimers’ disease. J. Clin. Investig. 1999, 104, 1321–1327. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Vassar, R.; Citron, M. Ab-generating enzymes: Recent advances in b- and g-secretases research. Neuron 2000, 27, 419–422. [Google Scholar] [CrossRef] [Green Version]
  34. Li, N.; Liu, K.; Qiu, Y.; Ren, Z.; Dai, R.; Deng, Y.; Qing, H. Effect of Presenilin Mutations on APP Cleavage; Insights into the Pathogenesis of FAD. Front. Aging Neurosci. 2016, 8, 51. [Google Scholar] [CrossRef] [Green Version]
  35. Ancolio, K.; Marambaud, P.; Dauch, P.; Checler, F. α-secretase-derived product of b-amyloid precursor protein is decreased by presenilin 1 mutations linked to familial Alzheimer’s disease. J. Neurochem. 1997, 69, 2494–2499. [Google Scholar] [CrossRef]
  36. Kelleher, R.J., 3rd; Shen, J. Presenilin-1 mutations and Alzheimer’s disease. Proc. Natl. Acad. Sci. USA 2017, 114, 629–631. [Google Scholar] [CrossRef] [Green Version]
  37. Sexton, C.; Snyder, H.; Beher, D.; Boxer, A.L.; Brannelly, P.; Brion, J.P.; Buee, L.; Cacace, A.M.; Chetelat, G.; Citron, M.; et al. Current directions in tau research: Highlights from Tau 2020. Alzheimer’s Dement. 2021. [Google Scholar] [CrossRef]
  38. Checler, F.; Afram, E.; Pardossi-Piquard, R.; Lauritzen, I. Is gamma-secretase a beneficial inactivating enzyme of the toxic APP C-terminal fragment C99? J. Biol. Chem. 2021, 296, 100489. [Google Scholar] [CrossRef]
  39. Garcia-Gonzalez, L.; Pilat, D.; Baranger, K.; Rivera, S. Emerging Alternative Proteinases in APP Metabolism and Alzheimer’s Disease Pathogenesis: A Focus on MT1-MMP and MT5-MMP. Front. Aging Neurosci. 2019, 11, 244. [Google Scholar] [CrossRef]
  40. Baranger, K.; Marchalant, Y.; Bonnet, A.E.; Crouzin, N.; Carrete, A.; Paumier, J.M.; Py, N.A.; Bernard, A.; Bauer, C.; Charrat, E.; et al. MT5-MMP is a new pro-amyloidogenic proteinase that promotes amyloid pathology and cognitive decline in a transgenic mouse model of Alzheimer’s disease. Cell Mol. Life Sci. 2016, 73, 217–236. [Google Scholar] [CrossRef] [Green Version]
  41. Willem, M.; Tahirovic, S.; Busche, M.A.; Ovsepian, S.V.; Chafai, M.; Kootar, S.; Hornburg, D.; Evans, L.D.; Moore, S.; Daria, A.; et al. η-Secretase processing of APP inhibits neuronal activity in the hippocampus. Nature 2015, 526, 443–447. [Google Scholar] [CrossRef] [PubMed]
  42. Becker-Pauly, C.; Pietrzik, C.U. The Metalloprotease Meprin beta Is an Alternative beta-Secretase of APP. Front. Mol. Neurosci. 2016, 9, 159. [Google Scholar] [CrossRef] [PubMed]
  43. Valverde, A.; Dunys, J.; Lorivel, T.; Debayle, D.; Gay, A.S.; Caillava, C.; Chami, M.; Checler, F. Dipeptidyl peptidase 4 contributes to Alzheimer’s disease-like defects in a mouse model and is increased in sporadic Alzheimer’s disease brains. J. Biol. Chem. 2021, 297, 100963. [Google Scholar] [CrossRef] [PubMed]
  44. Valverde, A.; Dunys, J.; Lorivel, T.; Debayle, D.; Gay, A.S.; Lacas-Gervais, S.; Roques, B.P.; Chami, M.; Checler, F. Aminopeptidase A contributes to biochemical, anatomical and cognitive defects in Alzheimer’s disease (AD) mouse model and is increased at early stage in sporadic AD brain. Acta Neuropathol. 2021, 141, 823–839. [Google Scholar] [CrossRef]
  45. Dunys, J.; Valverde, A.; Checler, F. Are N- and C-terminally truncated Abeta species key pathological triggers in Alzheimer’s disease? J. Biol. Chem. 2018, 293, 15419–15428. [Google Scholar] [CrossRef] [Green Version]
  46. Wirths, O.; Zampar, S.; Weggen, S. N-Terminally Truncated Abeta Peptide Variants in Alzheimer’s Disease. In Alzheimer’s Disease; Wisniewski, T., Ed.; Exon Publications: Brisbane, Australia, 2019. [Google Scholar] [CrossRef] [Green Version]
  47. Bayer, T.A. N-Truncated Abeta Starting at Position Four-Biochemical Features, Preclinical Models, and Potential as Drug Target in Alzheimer’s Disease. Front. Aging Neurosci. 2021, 13, 710579. [Google Scholar] [CrossRef]
  48. Jiang, Y.; Mullaney, K.A.; Peterhoff, C.M.; Che, S.; Schmidt, S.D.; Boyer-Boiteau, A.; Ginsberg, S.D.; Cataldo, A.M.; Mathews, P.M.; Nixon, R.A. Alzheimer’s-related endosome dysfunction in Down syndrome is Abeta-independent but requires APP and is reversed by BACE-1 inhibition. Proc. Natl. Acad. Sci. USA 2010, 107, 1630–1635. [Google Scholar] [CrossRef] [Green Version]
  49. Bourgeois, A.; Lauritzen, I.; Lorivel, T.; Bauer, C.; Checler, F.; Pardossi-Piquard, R. Intraneuronal accumulation of C99 contributes to synaptic alterations, apathy-like behavior, and spatial learning deficits in 3xTgAD and 2xTgAD mice. Neurobiol. Aging 2018, 71, 21–31. [Google Scholar] [CrossRef]
  50. Kwart, D.; Gregg, A.; Scheckel, C.; Murphy, E.A.; Paquet, D.; Duffield, M.; Fak, J.; Olsen, O.; Darnell, R.B.; Tessier-Lavigne, M. A Large Panel of Isogenic APP and PSEN1 Mutant Human iPSC Neurons Reveals Shared Endosomal Abnormalities Mediated by APP beta-CTFs, Not Abeta. Neuron 2019, 104, 1022. [Google Scholar] [CrossRef] [Green Version]
  51. Miranda, A.M.; Lasiecka, Z.M.; Xu, Y.; Neufeld, J.; Shahriar, S.; Simoes, S.; Chan, R.B.; Oliveira, T.G.; Small, S.A.; Di Paolo, G. Neuronal lysosomal dysfunction releases exosomes harboring APP C-terminal fragments and unique lipid signatures. Nat. Commun. 2018, 9, 291. [Google Scholar] [CrossRef] [Green Version]
  52. Lauritzen, I.; Pardossi-Piquard, R.; Bourgeois, A.; Becot, A.; Checler, F. Does Intraneuronal Accumulation of Carboxyl-terminal Fragments of the Amyloid Precursor Protein Trigger Early Neurotoxicity in Alzheimer’s Disease? Curr. Alzheimer Res. 2019, 16, 453–457. [Google Scholar] [CrossRef] [PubMed]
  53. Poewe, W.; Seppi, K.; Tanner, C.M.; Halliday, G.M.; Brundin, P.; Volkmann, J.; Schrag, A.E.; Lang, A.E. Parkinson disease. Nat. Rev. Dis. Primers 2017, 3, 17013. [Google Scholar] [CrossRef] [PubMed]
  54. Marino, B.L.B.; de Souza, L.R.; Sousa, K.P.A.; Ferreira, J.V.; Padilha, E.C.; da Silva, C.; Taft, C.A.; Hage-Melim, L.I.S. Parkinson’s Disease: A Review from Pathophysiology to Treatment. Mini Rev. Med. Chem. 2020, 20, 754–767. [Google Scholar] [CrossRef] [PubMed]
  55. Bloem, B.R.; Okun, M.S.; Klein, C. Parkinson’s disease. Lancet 2021, 397, 2284–2303. [Google Scholar] [CrossRef]
  56. Drui, G.; Carnicella, S.; Carcenac, C.; Favier, M.; Bertrand, A.; Boulet, S.; Savasta, M. Loss of dopaminergic nigrostriatal neurons accounts for the motivational and affective deficits in Parkinson’s disease. Mol. Psychiatry 2014, 19, 358–367. [Google Scholar] [CrossRef] [Green Version]
  57. Grosch, J.; Winkler, J.; Kohl, Z. Early Degeneration of Both Dopaminergic and Serotonergic Axons-A Common Mechanism in Parkinson’s Disease. Front. Cell Neurosci. 2016, 10, 293. [Google Scholar] [CrossRef] [Green Version]
  58. Masato, A.; Plotegher, N.; Boassa, D.; Bubacco, L. Impaired dopamine metabolism in Parkinson’s disease pathogenesis. Mol. Neurodegener. 2019, 14, 35. [Google Scholar] [CrossRef] [Green Version]
  59. Surmeier, D.J. Determinants of dopaminergic neuron loss in Parkinson’s disease. FEBS J. 2018, 285, 3657–3668. [Google Scholar] [CrossRef] [Green Version]
  60. Spillantini, M.G.; Goedert, M.; Crowther, R.A.; Murrell, J.R.; Farlow, M.R.; Ghetti, B. Familial multiple system tauopathy with presenile dementia: A disease with abundant neuronal and glial tau filaments. Proc. Natl. Acad. Sci. USA 1997, 94, 4113–4118. [Google Scholar] [CrossRef] [Green Version]
  61. Xu, L.; Pu, J. Alpha-Synuclein in Parkinson’s Disease: From Pathogenetic Dysfunction to Potential Clinical Application. Parkinson’s Dis. 2016, 2016, 1720621. [Google Scholar] [CrossRef] [Green Version]
  62. Lill, C.M. Genetics of Parkinson’s disease. Mol. Cell Probes 2016, 30, 386–396. [Google Scholar] [CrossRef]
  63. Day, J.O.; Mullin, S. The Genetics of Parkinson’s Disease and Implications for Clinical Practice. Genes 2021, 12, 1006. [Google Scholar] [CrossRef] [PubMed]
  64. Klein, C.; Westenberger, A. Genetics of Parkinson’s disease. Cold Spring Harb. Perspect. Med. 2012, 2, a008888. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Blauwendraat, C.; Nalls, M.A.; Singleton, A.B. The genetic architecture of Parkinson’s disease. Lancet Neurol. 2020, 19, 170–178. [Google Scholar] [CrossRef]
  66. Bandres-Ciga, S.; Diez-Fairen, M.; Kim, J.J.; Singleton, A.B. Genetics of Parkinson’s disease: An introspection of its journey towards precision medicine. Neurobiol. Dis. 2020, 137, 104782. [Google Scholar] [CrossRef] [PubMed]
  67. Polymeropoulos, M.H.; Higgins, L.S.; Golbe, L.; Johnson, W.G.; Ide, S.E.; Di Iorio, G.; Sanges, G.; Stenroos, E.S.; Pho, L.T.; Shaffer, A.A.; et al. A gene for Parkinson’s disease maps to 4q21-q23. Science 1996, 274, 1197–1199. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Athanassiadou, A.; Voutsinas, G.; Psiouri, L.; Leroy, E.; Polymeropoulos, M.H.; Ilias, A.; Maniatis, G.M.; Papapetropoulos, T. Genetic analysis of families with Parkinson disease that carry the Ala53Thr mutation in the gene encoding alpha-synuclein. Am. J. Hum. Genet. 1999, 65, 555–558. [Google Scholar] [CrossRef] [Green Version]
  69. Krüger, R.; Kuhn, W.; Müller, T.; Woitalla, D.; Graeber, M.; Kösel, S.; Przuntek, H.; Epplen, J.T.; Schöls, L.; Rriess, O. Ala30Pro mutation in the gene encoding a-synuclein in Parkinson’s disease. Nat. Genet. 1998, 18, 106–108. [Google Scholar] [CrossRef]
  70. Zarranz, J.J.; Alegre, J.; Gomez-Esteban, J.C.; Lezcano, E.; Ros, R.; Ampuero, I.; Vidal, L.; Hoenicka, J.; Rodriguez, O.; Atares, B.; et al. The new mutation, E46K, of a-syunuclein, causes Parkinson and Lewy bodies dementia. Ann. Neurol. 2004, 55, 164–173. [Google Scholar] [CrossRef]
  71. Nuytemans, K.; Theuns, J.; Cruts, M.; Van Broeckhoven, C. Genetic etiology of Parkinson disease associated with mutations in the SNCA, PARK2, PINK1, PARK7, and LRRK2 genes: A mutation update. Hum. Mutat. 2010, 31, 763–780. [Google Scholar] [CrossRef] [Green Version]
  72. Strauss, K.M.; Martins, L.M.; Plun-Favreau, H.; Marx, F.P.; Kautzmann, S.; Berg, D.; Gasser, T.; Wszolek, Z.; Müller, T.; Borneman, A.; et al. Loss of function mutations in the gene encoding Omi/HtrA2 in Parkinson’s disease. Hum. Mol. Gen. 2005, 14, 2099–2111. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Vilarino-Guell, C.; Wider, C.; Ross, O.A.; Dachsel, J.C.; Kachergus, J.M.; Lincoln, S.J.; Soto-Ortolaza, A.I.; Cobb, S.A.; Wilhoite, G.J.; Bacon, J.A.; et al. VPS35 mutations in Parkinson disease. Am. J. Hum. Genet. 2011, 89, 162–167. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Lücking, C.B.; Dürr, A.; Bonifati, V.; Vaughan, J.; De Michele, G.; Gasser, T.; Harhangi, B.S.; Meco, G.; Denèfle, P.; Wood, N.W.; et al. Association between early-onset Parkinson’s disease and mutations in the Parkin gene. New Eng. J. Med. 2000, 342, 1560–1567. [Google Scholar] [CrossRef] [PubMed]
  75. Klein, C.; Lohmann-Hedrich, K. Impact of recent genetic findings in Parkinson’s disease. Curr. Opin. Neurol. 2007, 20, 453–464. [Google Scholar] [CrossRef]
  76. Corti, O.; Lesage, S.; Brice, A. What Genetics Tells us About the Causes and Mechanisms of Parkinson’s Disease. Physiol. Rev. 2011, 91, 1161–1218. [Google Scholar] [CrossRef]
  77. Rogaeva, E.; Johnson, J.; Lang, A.E.; Gulick, C.; Gwinn-Hardy, K.; Kawarai, T.; Sato, C.; Morgan, A.; Werner, J.; Nussbaum, R.; et al. Analysis of the PINK1 gene in a large cohort of cases with Parkinson disease. Arch. Neurol. 2004, 61, 1898–1904. [Google Scholar] [CrossRef]
  78. Bonifati, V.; Rohe, C.F.; Breedveld, G.J.; Fabrizio, E.; De Mari, M.; Tassorelli, C.; Tavella, A.; Marconi, R.; Nicholl, D.J.; Chien, H.F.; et al. Early-onset parkinsonism associated with PINK1 mutations: Frequency, genotypes, and phenotypes. Neurology 2005, 65, 87–95. [Google Scholar] [CrossRef]
  79. Valente, E.M.; Abou-Sleiman, P.M.; Caputo, V.; Muqit, M.M.; Harvey, K.; Gispert, S.; Ali, Z.; Del Turco, D.; Bentivoglio, A.R.; Healy, D.G.; et al. Hereditary early-onset Parkinson’s disease caused by mutations in PINK1. Science 2004, 304, 1158–1160. [Google Scholar] [CrossRef] [Green Version]
  80. Bonifati, V.; Rizzu, P.; Squitieri, F.; Krieger, E.; Vanacore, N.; Swieten, J.C.v.; Brice, A.; Duijn, C.M.v.; Oostra, B.A.; Meco, G.; et al. DJ-1 (PARK7), a novel gene for autosomal recessive, early onset parkinsonism. Neurol. Sci. 2003, 24, 159–160. [Google Scholar] [CrossRef] [Green Version]
  81. Ramirez, A.; Heimbach, A.; Grundemann, J.; Stiller, B.; Hampshire, D.; Cid, L.P.; Goebel, I.; Mubaidin, A.F.; Wriekat, A.L.; Roeper, J.; et al. Hereditary parkinsonism with dementia is caused by mutations in ATP13A2, encoding a lysosomal type 5 P-type ATPase. Nat. Genet. 2006, 38, 1184–1191. [Google Scholar] [CrossRef]
  82. Omura, T.; Kaneko, M.; Okuma, Y.; Matsubara, K.; Nomura, Y. Endoplasmic reticulum stress and Parkinson’s disease: The role of HRD1 in averting apoptosis in neurodegenerative disease. Oxid. Med. Cell. Longev. 2013, 2013, 239854. [Google Scholar] [CrossRef] [PubMed]
  83. Costa, C.A.D.; Manaa, W.E.; Duplan, E.; Checler, F. The Endoplasmic Reticulum Stress/Unfolded Protein Response and Their Contributions to Parkinson’s Disease Physiopathology. Cells 2020, 9, 2495. [Google Scholar] [CrossRef] [PubMed]
  84. Dernie, F. Mitophagy in Parkinson’s disease: From pathogenesis to treatment target. Neurochem. Int. 2020, 138, 104756. [Google Scholar] [CrossRef] [PubMed]
  85. Liu, J.; Liu, W.; Li, R.; Yang, H. Mitophagy in Parkinson’s Disease: From Pathogenesis to Treatment. Cells 2019, 8, 712. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Lynch-Day, M.A.; Mao, K.; Wang, K.; Zhao, M.; Klionsky, D.J. The role of autophagy in Parkinson’s disease. Cold Spring Harb. Perspect. Med. 2012, 2, a009357. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Hou, X.; Watzlawik, J.O.; Fiesel, F.C.; Springer, W. Autophagy in Parkinson’s Disease. J. Mol. Biol. 2020, 432, 2651–2672. [Google Scholar] [CrossRef]
  88. Cerri, S.; Blandini, F. Role of Autophagy in Parkinson’s Disease. Curr. Med. Chem. 2019, 26, 3702–3718. [Google Scholar] [CrossRef]
  89. Park, J.S.; Davis, R.L.; Sue, C.M. Mitochondrial Dysfunction in Parkinson’s Disease: New Mechanistic Insights and Therapeutic Perspectives. Curr. Neurol. Neurosci. Rep. 2018, 18, 21. [Google Scholar] [CrossRef] [Green Version]
  90. Dehay, B.; Martinez-Vicente, M.; Caldwell, G.A.; Caldwell, K.A.; Yue, Z.; Cookson, M.R.; Klein, C.; Vila, M.; Bezard, E. Lysosomal impairment in Parkinson’s disease. Mov. Disord. 2013, 28, 725–732. [Google Scholar] [CrossRef]
  91. Nguyen, M.; Wong, Y.C.; Ysselstein, D.; Severino, A.; Krainc, D. Synaptic, Mitochondrial, and Lysosomal Dysfunction in Parkinson’s Disease. Trends Neurosci. 2019, 42, 140–149. [Google Scholar] [CrossRef]
  92. Klein, A.D.; Mazzulli, J.R. Is Parkinson’s disease a lysosomal disorder? Brain 2018, 141, 2255–2262. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Burbulla, L.F.; Song, P.; Mazzulli, J.R.; Zampese, E.; Wong, Y.C.; Jeon, S.; Santos, D.P.; Blanz, J.; Obermaier, C.D.; Strojny, C.; et al. Dopamine oxidation mediates mitochondrial and lysosomal dysfunction in Parkinson’s disease. Science 2017, 357, 1255–1261. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Alves Da Costa, C.; Paitel, E.; Vincent, B.; Checler, F. Alpha-synuclein lowers p53-dependent apoptotic response of neuronal cells. Abolishment by 6-hydroxydopamine and implication for Parkinson’s disease. J. Biol. Chem. 2002, 277, 50980–50984. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Venderova, K.; Park, D.S. Programmed cell death in Parkinson’s disease. Cold Spring Harb. Perspect. Med. 2012, 2, a009365. [Google Scholar] [CrossRef]
  96. Michel, P.P.; Hirsch, E.C.; Hunot, S. Understanding Dopaminergic Cell Death Pathways in Parkinson Disease. Neuron 2016, 90, 675–691. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Bucciantini, M.; Giannoni, E.; Chiti, F.; Baroni, F.; Formigli, L.; Zurdo, J.; Taddei, N.; Ramponi, G.; Dobson, C.M.; Stefani, M. Inherent toxicity of aggregates implies a common mechanism for protein misfolding diseases. Nature 2002, 416, 507–511. [Google Scholar] [CrossRef]
  98. Bence, N.F.; Sampat, R.M.; Kopito, R.R. Impairment of the ubiquitin-proteasome system by protein aggregation. Science 2001, 292, 1552–1555. [Google Scholar] [CrossRef]
  99. Checler, F.; Alves da Costa, C.; Ancolio, K.; Lopez-Perez, E.; Marambaud, P. Role of the Proteasome in Alzheimer’s disease. Biochim. Biophys. Acta 2000, 1502, 133–138. [Google Scholar] [CrossRef] [Green Version]
  100. Bonet-Costa, V.; Pomatto, L.C.; Davies, K.J. The Proteasome and Oxidative Stress in Alzheimer’s Disease. Antioxid. Redox Signal. 2016, 25, 886–901. [Google Scholar] [CrossRef] [Green Version]
  101. Perry, G.; Friedman, R.; Shaw, G.; Chau, V. Ubiquitin is detected in neurofibrillary tangles and senile plaque neurites of Alzheimer’s disease brains. Proc. Natl. Acad. Sci. USA 1987, 84, 3033–3036. [Google Scholar] [CrossRef] [Green Version]
  102. Almeida, C.G.; Takahashi, R.H.; Gouras, G.K. Beta-amyloid accumulation impairs multivesicular body sorting by inhibiting the ubiquitin-proteasome system. J. Neurosci. 2006, 26, 4277–4288. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Gregori, L.; Hainfeld, J.F.; Simon, M.N.; Goldgaber, D. Binding of amyloid b protein to the 20S proteasome. J. Biol. Chem. 1997, 272, 58–62. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Lopez Salon, M.; Pasquini, L.; Moreno, B.; Pasquini, J.M.; Soto, E. Relationship between b-amyloid degradation and the 26S proteasome in neural cells. Exp. Neurol. 2003, 180, 131–143. [Google Scholar] [CrossRef]
  105. McNaught, K.S.; Belizaire, R.; Isacson, O.; Jenner, P.; Olanow, C.W. Altered proteasomal function in sporadic Parkinson’s disease. Exp. Neurol. 2003, 179, 38–46. [Google Scholar] [CrossRef] [PubMed]
  106. Bi, M.; Du, X.; Jiao, Q.; Chen, X.; Jiang, H. Expanding the role of proteasome homeostasis in Parkinson’s disease: Beyond protein breakdown. Cell Death Dis. 2021, 12, 154. [Google Scholar] [CrossRef] [PubMed]
  107. Bennett, D.A.; Leurgans, S. Is there a link between cancer and Alzheimer disease? Neurology 2010, 74, 100–101. [Google Scholar] [CrossRef] [PubMed]
  108. Alves da Costa, C.; Dunys, J.; Brau, F.; Wilk, S.; Cappai, R.; Checler, F. 6-Hydroxydopamine but not 1-methyl-4-phenylpyridinium abolishes alpha-synuclein anti-apoptotic phenotype by inhibiting its proteasomal degradation and by promoting its aggregation. J. Biol. Chem. 2006, 281, 9824–9831. [Google Scholar] [CrossRef] [Green Version]
  109. Shimura, H.; Hattori, N.; Kubo, S.; Mizuno, Y.; Asakawa, S.; Minoshima, S.; Shimizu, N.; Iwai, K.; Chiba, T.; Tanaka, K.; et al. Familial Parkinson disease gene product, parkin, is a ubiquitin-protein ligase. Nat. Genet. 2000, 25, 302–305. [Google Scholar] [CrossRef]
  110. Yoshii, S.R.; Kishi, C.; Ishihara, N.; Mizushima, N. Parkin mediates proteasome-dependent protein degradation and rupture of the outer mitochondrial membrane. J. Biol. Chem. 2011, 286, 19630–19640. [Google Scholar] [CrossRef] [Green Version]
  111. Song, P.; Li, S.; Wu, H.; Gao, R.; Rao, G.; Wang, D.; Chen, Z.; Ma, B.; Wang, H.; Sui, N.; et al. Parkin promotes proteasomal degradation of p62: Implication of selective vulnerability of neuronal cells in the pathogenesis of Parkinson’s disease. Protein Cell 2016, 7, 114–129. [Google Scholar] [CrossRef] [Green Version]
  112. Betteridge, D.J. What is oxidative stress? Metab. Clin. Exp. 2000, 49, 3–8. [Google Scholar] [CrossRef]
  113. Sies, H.; Berndt, C.; Jones, D.P. Oxidative Stress. Annu. Rev. Biochem. 2017, 86, 715–748. [Google Scholar] [CrossRef] [PubMed]
  114. Markesbery, W.R. The role of oxidative stress in Alzheimer disease. Arch. Neurol. 1999, 56, 1449–1452. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Gella, A.; Durany, N. Oxidative stress in Alzheimer disease. Cell Adhes. Migr. 2009, 3, 88–93. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Aksenov, M.Y.; Tucker, H.M.; Nair, P.; Aksenova, M.V.; Butterfield, D.A.; Estus, S.; Markesbery, W.R. The expression of key oxidative stress-handling genes in different brain regions in Alzheimer’s disease. J. Mol. Neurosci. 1998, 11, 151–164. [Google Scholar] [CrossRef]
  117. Youssef, P.; Chami, B.; Lim, J.; Middleton, T.; Sutherland, G.T.; Witting, P.K. Evidence supporting oxidative stress in a moderately affected area of the brain in Alzheimer’s disease. Sci. Rep. 2018, 8, 11553. [Google Scholar] [CrossRef] [Green Version]
  118. Dias, V.; Junn, E.; Mouradian, M.M. The role of oxidative stress in Parkinson’s disease. J. Parkinson’s Dis. 2013, 3, 461–491. [Google Scholar] [CrossRef] [Green Version]
  119. Blesa, J.; Trigo-Damas, I.; Quiroga-Varela, A.; Jackson-Lewis, V.R. Oxidative stress and Parkinson’s disease. Front. Neuroanat. 2015, 9, 91. [Google Scholar] [CrossRef] [Green Version]
  120. Wei, Z.; Li, X.; Li, X.; Liu, Q.; Cheng, Y. Oxidative Stress in Parkinson’s Disease: A Systematic Review and Meta-Analysis. Front. Mol. Neurosci. 2018, 11, 236. [Google Scholar] [CrossRef]
  121. Hotamisligil, G.S.; Davis, R.J. Cell Signaling and Stress Responses. Cold Spring Harb. Perspect. Biol. 2016, 8, a006072. [Google Scholar] [CrossRef] [Green Version]
  122. Wu, X.; Rapoport, T.A. Mechanistic insights into ER-associated protein degradation. Curr. Opin. Cell Biol. 2018, 53, 22–28. [Google Scholar] [CrossRef] [PubMed]
  123. Sun, Z.; Brodsky, J.L. Protein quality control in the secretory pathway. J. Cell Biol. 2019, 218, 3171–3187. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Frakes, A.E.; Dillin, A. The UPR(ER): Sensor and Coordinator of Organismal Homeostasis. Mol. Cell 2017, 66, 761–771. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Hetz, C.; Zhang, K.; Kaufman, R.J. Mechanisms, regulation and functions of the unfolded protein response. Nat. Rev. Mol. Cell Biol. 2020, 21, 421–438. [Google Scholar] [CrossRef] [PubMed]
  126. Hetz, C.; Saxena, S. ER stress and the unfolded protein response in neurodegeneration. Nat. Rev. Neurol. 2017, 13, 477–491. [Google Scholar] [CrossRef] [PubMed]
  127. Kurtishi, A.; Rosen, B.; Patil, K.S.; Alves, G.W.; Moller, S.G. Cellular Proteostasis in Neurodegeneration. Mol. Neurobiol. 2019, 56, 3676–3689. [Google Scholar] [CrossRef]
  128. Ghemrawi, R.; Khair, M. Endoplasmic Reticulum Stress and Unfolded Protein Response in Neurodegenerative Diseases. Int. J. Mol. Sci. 2020, 21, 6127. [Google Scholar] [CrossRef]
  129. Hoozemans, J.J.; Veerhuis, R.; Van Haastert, E.S.; Rozemuller, J.M.; Baas, F.; Eikelenboom, P.; Scheper, W. The unfolded protein response is activated in Alzheimer’s disease. Acta Neuropathol. 2005, 110, 165–172. [Google Scholar] [CrossRef]
  130. Barbero-Camps, E.; Fernandez, A.; Baulies, A.; Martinez, L.; Fernandez-Checa, J.C.; Colell, A. Endoplasmic reticulum stress mediates amyloid beta neurotoxicity via mitochondrial cholesterol trafficking. Am. J. Pathol. 2014, 184, 2066–2081. [Google Scholar] [CrossRef]
  131. Ma, T.; Trinh, M.A.; Wexler, A.J.; Bourbon, C.; Gatti, E.; Pierre, P.; Cavener, D.R.; Klann, E. Suppression of eIF2alpha kinases alleviates Alzheimer’s disease-related plasticity and memory deficits. Nat. Neurosci. 2013, 16, 1299–1305. [Google Scholar] [CrossRef] [Green Version]
  132. Hashimoto, S.; Saido, T.C. Critical review: Involvement of endoplasmic reticulum stress in the aetiology of Alzheimer’s disease. Open Biol. 2018, 8, 180024. [Google Scholar] [CrossRef] [PubMed]
  133. Goswami, P.; Afjal, M.A.; Akhter, J.; Mangla, A.; Khan, J.; Parvez, S.; Raisuddin, S. Involvement of endoplasmic reticulum stress in amyloid beta (1-42)-induced Alzheimer’s like neuropathological process in rat brain. Brain Res. Bull. 2020, 165, 108–117. [Google Scholar] [CrossRef] [PubMed]
  134. Nishitsuji, K.; Tomiyama, T.; Ishibashi, K.; Ito, K.; Teraoka, R.; Lambert, M.P.; Klein, W.L.; Mori, H. The E693Delta mutation in amyloid precursor protein increases intracellular accumulation of amyloid beta oligomers and causes endoplasmic reticulum stress-induced apoptosis in cultured cells. Am. J. Pathol. 2009, 174, 957–969. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Alberdi, E.; Wyssenbach, A.; Alberdi, M.; Sanchez-Gomez, M.V.; Cavaliere, F.; Rodriguez, J.J.; Verkhratsky, A.; Matute, C. Ca(2+) -dependent endoplasmic reticulum stress correlates with astrogliosis in oligomeric amyloid beta-treated astrocytes and in a model of Alzheimer’s disease. Aging Cell 2013, 12, 292–302. [Google Scholar] [CrossRef] [Green Version]
  136. Castillo-Carranza, D.L.; Zhang, Y.; Guerrero-Munoz, M.J.; Kayed, R.; Rincon-Limas, D.E.; Fernandez-Funez, P. Differential activation of the ER stress factor XBP1 by oligomeric assemblies. Neurochem. Res. 2012, 37, 1707–1717. [Google Scholar] [CrossRef] [Green Version]
  137. Kondo, T.; Asai, M.; Tsukita, K.; Kutoku, Y.; Ohsawa, Y.; Sunada, Y.; Imamura, K.; Egawa, N.; Yahata, N.; Okita, K.; et al. Modeling Alzheimer’s disease with iPSCs reveals stress phenotypes associated with intracellular Abeta and differential drug responsiveness. Cell Stem Cell 2013, 12, 487–496. [Google Scholar] [CrossRef] [Green Version]
  138. Abisambra, J.F.; Jinwal, U.K.; Blair, L.J.; O’Leary, J.C., 3rd; Li, Q.; Brady, S.; Wang, L.; Guidi, C.E.; Zhang, B.; Nordhues, B.A.; et al. Tau accumulation activates the unfolded protein response by impairing endoplasmic reticulum-associated degradation. J. Neurosci. 2013, 33, 9498–9507. [Google Scholar] [CrossRef] [Green Version]
  139. Mou, Z.; Yuan, Y.H.; Zhang, Z.; Song, L.K.; Chen, N.H. Endoplasmic reticulum stress, an important factor in the development of Parkinson’s disease. Toxicol Lett 2020, 324, 20–29. [Google Scholar] [CrossRef]
  140. Lindholm, D.; Wootz, H.; Korhonen, L. ER stress and neurodegenerative diseases. Cell Death Differ. 2006, 13, 385–392. [Google Scholar] [CrossRef]
  141. Kovaleva, V.; Saarma, M. Endoplasmic Reticulum Stress Regulators: New Drug Targets for Parkinson’s Disease. J. Parkinson’s Dis. 2021, 11, S219–S228. [Google Scholar] [CrossRef]
  142. Bellucci, A.; Navarria, L.; Zaltieri, M.; Falarti, E.; Bodei, S.; Sigala, S.; Battistin, L.; Spillantini, M.; Missale, C.; Spano, P. Induction of the unfolded protein response by alpha-synuclein in experimental models of Parkinson’s disease. J. Neurochem. 2011, 116, 588–605. [Google Scholar] [CrossRef] [PubMed]
  143. Sugeno, N.; Takeda, A.; Hasegawa, T.; Kobayashi, M.; Kikuchi, A.; Mori, F.; Wakabayashi, K.; Itoyama, Y. Serine 129 Phosphorylation of {alpha}-Synuclein Induces Unfolded Protein Response-mediated Cell Death. J. Biol. Chem. 2008, 283, 23179–23188. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Gorbatyuk, M.S.; Shabashvili, A.; Chen, W.; Meyers, C.; Sullivan, L.F.; Salganik, M.; Lin, J.H.; Lewin, A.S.; Muzyczka, N.; Gorbatyuk, O.S. Glucose regulated protein 78 diminishes alpha-synuclein neurotoxicity in a rat model of Parkinson disease. Mol. Ther. J. Am. Soc. Gene Ther. 2012, 20, 1327–1337. [Google Scholar] [CrossRef] [PubMed]
  145. Bouman, L.; Schlierf, A.; Lutz, A.K.; Shan, J.; Deinlein, A.; Kast, J.; Galehdar, Z.; Palmisano, V.; Patenge, N.; Berg, D.; et al. Parkin is transcriptionally regulated by ATF4: Evidence for an interconnection between mitochondrial stress and ER stress. Cell Death Differ. 2011, 18, 769–782. [Google Scholar] [CrossRef] [Green Version]
  146. Imai, Y.; Soda, M.; Takahashi, R. Parkin suppresses unfolded protein stress-induced cell deth through its E3 ubiquitin-protein ligase activity. J. Biol. Chem. 2002, 275, 35661–35664. [Google Scholar] [CrossRef] [Green Version]
  147. Imai, Y.; Soda, M.; Inoue, H.; Hattori, N.; Mizuno, Y.; Takahashi, R. An unfolded putative transmembrane polypeptide, which can lead to endoplasmic reticulum stress, is a substrate of parkin. Cell 2001, 105, 891–902. [Google Scholar] [CrossRef] [Green Version]
  148. Yoshida, H.; Matsui, T.; Yamamoto, A.; Okada, T.; Mori, K. XBP1 mRNA is induced by ATF6 and spliced by IRE1 in response to ER stress to produce a highly active transcription factor. Cell 2001, 107, 881–891. [Google Scholar] [CrossRef] [Green Version]
  149. El Manaa, W.; Duplan, E.; Goiran, T.; Lauritzen, I.; Vaillant Beuchot, L.; Lacas-Gervais, S.; Morais, V.A.; You, H.; Qi, L.; Salazar, M.; et al. Transcription- and phosphorylation-dependent control of a functional interplay between XBP1s and PINK1 governs mitophagy and potentially impacts Parkinson disease pathophysiology. Autophagy 2021, 17, 4363–4385. [Google Scholar] [CrossRef]
  150. Yuan, Y.; Cao, P.; Smith, M.A.; Kramp, K.; Huang, Y.; Hisamoto, N.; Matsumoto, K.; Hatzoglou, M.; Jin, H.; Feng, Z. Dysregulated LRRK2 signaling in response to endoplasmic reticulum stress leads to dopaminergic neuron degeneration in C. elegans. PLoS ONE 2011, 6, e22354. [Google Scholar] [CrossRef] [Green Version]
  151. Wang, W.; Zhao, F.; Ma, X.; Perry, G.; Zhu, X. Mitochondria dysfunction in the pathogenesis of Alzheimer’s disease: Recent advances. Mol. Neurodegener. 2020, 15, 30. [Google Scholar] [CrossRef]
  152. Weidling, I.W.; Swerdlow, R.H. Mitochondria in Alzheimer’s disease and their potential role in Alzheimer’s proteostasis. Exp. Neurol. 2020, 330, 113321. [Google Scholar] [CrossRef] [PubMed]
  153. Bell, S.M.; Barnes, K.; De Marco, M.; Shaw, P.J.; Ferraiuolo, L.; Blackburn, D.J.; Venneri, A.; Mortiboys, H. Mitochondrial Dysfunction in Alzheimer’s Disease: A Biomarker of the Future? Biomedicines 2021, 9, 63. [Google Scholar] [CrossRef] [PubMed]
  154. Calvo-Rodriguez, M.; Bacskai, B.J. Mitochondria and Calcium in Alzheimer’s Disease: From Cell Signaling to Neuronal Cell Death. Trends Neurosci. 2021, 44, 136–151. [Google Scholar] [CrossRef] [PubMed]
  155. Swerdlow, R.H. Mitochondria and Mitochondrial Cascades in Alzheimer’s Disease. J. Alzheimer’s Dis. 2018, 62, 1403–1416. [Google Scholar] [CrossRef] [Green Version]
  156. Zhang, L.; Guo, X.Q.; Chu, J.F.; Zhang, X.; Yan, Z.R.; Li, Y.Z. Potential hippocampal genes and pathways involved in Alzheimer’s disease: A bioinformatic analysis. Genet. Mol. Res. 2015, 14, 7218–7232. [Google Scholar] [CrossRef]
  157. Mastroeni, D.; Khdour, O.M.; Delvaux, E.; Nolz, J.; Olsen, G.; Berchtold, N.; Cotman, C.; Hecht, S.M.; Coleman, P.D. Nuclear but not mitochondrial-encoded oxidative phosphorylation genes are altered in aging, mild cognitive impairment, and Alzheimer’s disease. Alzheimer’s Dement. 2017, 13, 510–519. [Google Scholar] [CrossRef] [Green Version]
  158. Area-Gomez, E.; Schon, E.A. Mitochondria-associated ER membranes and Alzheimer disease. Curr. Opin. Genet. Dev. 2016, 38, 90–96. [Google Scholar] [CrossRef] [Green Version]
  159. Eysert, F.; Kinoshita, P.F.; Mary, A.; Vaillant-Beuchot, L.; Checler, F.; Chami, M. Molecular Dysfunctions of Mitochondria-Associated Membranes (MAMs) in Alzheimer’s Disease. Int. J. Mol. Sci. 2020, 21, 9521. [Google Scholar] [CrossRef]
  160. Chen, C.; Turnbull, D.M.; Reeve, A.K. Mitochondrial Dysfunction in Parkinson’s Disease-Cause or Consequence? Biology 2019, 8, 38. [Google Scholar] [CrossRef] [Green Version]
  161. Ganguly, G.; Chakrabarti, S.; Chatterjee, U.; Saso, L. Proteinopathy, oxidative stress and mitochondrial dysfunction: Cross talk in Alzheimer’s disease and Parkinson’s disease. Drug Des. Devel. Ther. 2017, 11, 797–810. [Google Scholar] [CrossRef] [Green Version]
  162. Seet, R.C.; Lee, C.Y.; Lim, E.C.; Tan, J.J.; Quek, A.M.; Chong, W.L.; Looi, W.F.; Huang, S.H.; Wang, H.; Chan, Y.H.; et al. Oxidative damage in Parkinson disease: Measurement using accurate biomarkers. Free Radic. Biol. Med. 2010, 48, 560–566. [Google Scholar] [CrossRef] [PubMed]
  163. Yadava, N.; Nicholls, D.G. Spare respiratory capacity rather than oxidative stress regulates glutamate excitotoxicity after partial respiratory inhibition of mitochondrial complex I with rotenone. J. Neurosci. 2007, 27, 7310–7317. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. Gandhi, S.; Wood-Kaczmar, A.; Yao, Z.; Plun-Favreau, H.; Deas, E.; Klupsch, K.; Downward, J.; Latchman, D.S.; Tabrizi, S.J.; Wood, N.W.; et al. PINK1-associated Parkinson’s disease is caused by neuronal vulnerability to calcium-induced cell death. Mol. Cell 2009, 33, 627–638. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Prasuhn, J.; Davis, R.L.; Kumar, K.R. Targeting Mitochondrial Impairment in Parkinson’s Disease: Challenges and Opportunities. Front. Cell Dev. Biol. 2020, 8, 615461. [Google Scholar] [CrossRef] [PubMed]
  166. Wang, X.; Petrie, T.G.; Liu, Y.; Liu, J.; Fujioka, H.; Zhu, X. Parkinson’s disease-associated DJ-1 mutations impair mitochondrial dynamics and cause mitochondrial dysfunction. J. Neurochem. 2012, 121, 830–839. [Google Scholar] [CrossRef]
  167. Wang, X.; Yan, M.H.; Fujioka, H.; Liu, J.; Wilson-Delfosse, A.; Chen, S.G.; Perry, G.; Casadesus, G.; Zhu, X. LRRK2 regulates mitochondrial dynamics and function through direct interaction with DLP1. Hum. Mol. Genet. 2012, 21, 1931–1944. [Google Scholar] [CrossRef] [Green Version]
  168. Hsieh, C.H.; Shaltouki, A.; Gonzalez, A.E.; Bettencourt da Cruz, A.; Burbulla, L.F.; St Lawrence, E.; Schule, B.; Krainc, D.; Palmer, T.D.; Wang, X. Functional Impairment in Miro Degradation and Mitophagy Is a Shared Feature in Familial and Sporadic Parkinson’s Disease. Cell Stem Cell 2016, 19, 709–724. [Google Scholar] [CrossRef] [Green Version]
  169. Devi, L.; Raghavendran, V.; Prabhu, B.M.; Avadhani, N.G.; Anandatheerthavarada, H.K. Mitochondrial import and accumulation of alpha-synuclein impair complex I in human dopaminergic neuronal cultures and Parkinson disease brain. J. Biol. Chem. 2008, 283, 9089–9100. [Google Scholar] [CrossRef] [Green Version]
  170. Ludtmann, M.H.R.; Angelova, P.R.; Horrocks, M.H.; Choi, M.L.; Rodrigues, M.; Baev, A.Y.; Berezhnov, A.V.; Yao, Z.; Little, D.; Banushi, B.; et al. alpha-synuclein oligomers interact with ATP synthase and open the permeability transition pore in Parkinson’s disease. Nat. Commun. 2018, 9, 2293. [Google Scholar] [CrossRef] [Green Version]
  171. Ge, P.; Dawson, V.L.; Dawson, T.M. PINK1 and Parkin mitochondrial quality control: A source of regional vulnerability in Parkinson’s disease. Mol. Neurodegener. 2020, 15, 20. [Google Scholar] [CrossRef] [Green Version]
  172. Eiyama, A.; Okamoto, K. PINK1/Parkin-mediated mitophagy in mammalian cells. Curr. Opin. Cell Biol. 2015, 33, 95–101. [Google Scholar] [CrossRef] [PubMed]
  173. Nguyen, T.N.; Padman, B.S.; Lazarou, M. Deciphering the Molecular Signals of PINK1/Parkin Mitophagy. Trends Cell Biol. 2016, 26, 733–744. [Google Scholar] [CrossRef] [PubMed]
  174. Kerr, J.S.; Adriaanse, B.A.; Greig, N.H.; Mattson, M.P.; Cader, M.Z.; Bohr, V.A.; Fang, E.F. Mitophagy and Alzheimer’s Disease: Cellular and Molecular Mechanisms. Trends Neurosci. 2017, 40, 151–166. [Google Scholar] [CrossRef] [Green Version]
  175. Cai, Q.; Jeong, Y.Y. Mitophagy in Alzheimer’s Disease and Other Age-Related Neurodegenerative Diseases. Cells 2020, 9, 150. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  176. Pradeepkiran, J.A.; Reddy, P.H. Defective mitophagy in Alzheimer’s disease. Ageing Res. Rev. 2020, 64, 101191. [Google Scholar] [CrossRef] [PubMed]
  177. Fang, E.F.; Hou, Y.; Palikaras, K.; Adriaanse, B.A.; Kerr, J.S.; Yang, B.; Lautrup, S.; Hasan-Olive, M.M.; Caponio, D.; Dan, X.; et al. Mitophagy inhibits amyloid-beta and tau pathology and reverses cognitive deficits in models of Alzheimer’s disease. Nat. Neurosci. 2019, 22, 401–412. [Google Scholar] [CrossRef]
  178. Reddy, P.H.; Oliver, D.M. Amyloid Beta and Phosphorylated Tau-Induced Defective Autophagy and Mitophagy in Alzheimer’s Disease. Cells 2019, 8, 488. [Google Scholar] [CrossRef] [Green Version]
  179. Oddo, S.; Caccamo, A.; Shepherd, J.D.; Murphy, G.; Golde, T.E.; Kayed, R.; Metherate, R.; Mattson, M.P.; Akbari, Y.; LaFerla, F. Triple-transgenic model of Alzheimer’s disease with plaques and tangles: Intracellular Ab and synaptic dysfunction. Neuron 2003, 39, 409–421. [Google Scholar] [CrossRef] [Green Version]
  180. Lee, S.E.; Kwon, D.; Shin, N.; Kong, D.; Kim, N.G.; Kim, H.Y.; Kim, M.J.; Choi, S.W.; Kang, K.S. Accumulation of APP-CTF induces mitophagy dysfunction in the iNSCs model of Alzheimer’s disease. Cell Death Discov. 2022, 8, 1. [Google Scholar] [CrossRef]
  181. Malpartida, A.B.; Williamson, M.; Narendra, D.P.; Wade-Martins, R.; Ryan, B.J. Mitochondrial Dysfunction and Mitophagy in Parkinson’s Disease: From Mechanism to Therapy. Trends Biochem. Sci. 2021, 46, 329–343. [Google Scholar] [CrossRef]
  182. Pickrell, A.M.; Youle, R.J. The roles of PINK1, parkin, and mitochondrial fidelity in Parkinson’s disease. Neuron 2015, 85, 257–273. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  183. Gegg, M.E.; Cooper, J.M.; Chau, K.Y.; Rojo, M.; Schapira, A.H.; Taanman, J.W. Mitofusin 1 and mitofusin 2 are ubiquitinated in a PINK1/parkin-dependent manner upon induction of mitophagy. Hum. Mol. Genet. 2010, 19, 4861–4870. [Google Scholar] [CrossRef] [PubMed]
  184. Glauser, L.; Sonnay, S.; Stafa, K.; Moore, D.J. Parkin promotes the ubiquitination and degradation of the mitochondrial fusion factor mitofusin 1. J. Neurochem. 2011, 118, 636–645. [Google Scholar] [CrossRef] [PubMed]
  185. Han, H.; Tan, J.; Wang, R.; Wan, H.; He, Y.; Yan, X.; Guo, J.; Gao, Q.; Li, J.; Shang, S.; et al. PINK1 phosphorylates Drp1(S616) to regulate mitophagy-independent mitochondrial dynamics. EMBO Rep. 2020, 21, e48686. [Google Scholar] [CrossRef]
  186. Cribbs, J.T.; Strack, S. Reversible phosphorylation of Drp1 by cyclic AMP-dependent protein kinase and calcineurin regulates mitochondrial fission and cell death. EMBO Rep. 2007, 8, 939–944. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  187. Dagda, R.K.; Cherra, S.J., 3rd; Kulich, S.M.; Tandon, A.; Park, D.; Chu, C.T. Loss of PINK1 function promotes mitophagy through effects on oxidative stress and mitochondrial fission. J. Biol. Chem. 2009, 284, 13843–13855. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  188. Sandebring, A.; Dehvari, N.; Perez-Manso, M.; Thomas, K.J.; Karpilovski, E.; Cookson, M.R.; Cowburn, R.F.; Cedazo-Minguez, A. Parkin deficiency disrupts calcium homeostasis by modulating phospholipase C signalling. FEBS J. 2009, 276, 5041–5052. [Google Scholar] [CrossRef] [Green Version]
  189. Goiran, T.; Duplan, E.; Chami, M.; Bourgeois, A.; El Manaa, W.; Rouland, L.; Dunys, J.; Lauritzen, I.; You, H.; Stambolic, V.; et al. beta-Amyloid Precursor Protein Intracellular Domain Controls Mitochondrial Function by Modulating Phosphatase and Tensin Homolog-Induced Kinase 1 Transcription in Cells and in Alzheimer Mice Models. Biol. Psychiatry 2018, 83, 416–427. [Google Scholar] [CrossRef] [Green Version]
  190. Weinlich, R.; Oberst, A.; Beere, H.M.; Green, D.R. Necroptosis in development, inflammation and disease. Nat. Rev. Mol. Cell Biol. 2017, 18, 127–136. [Google Scholar] [CrossRef]
  191. Erekat, N.S. Apoptosis and its Role in Parkinson’s Disease. In Parkinson’s Disease: Pathogenesis and Clinical Aspects; Stoker, T.B., Greenland, J.C., Eds.; Exon Publications: Brisbane, Australia, 2018. [Google Scholar] [CrossRef] [Green Version]
  192. Szybinska, A.; Lesniak, W. P53 Dysfunction in Neurodegenerative Diseases-The Cause or Effect of Pathological Changes? Aging Dis. 2017, 8, 506–518. [Google Scholar] [CrossRef] [Green Version]
  193. Alves da Costa, C.; Checler, F. Apoptosis in Parkinson’s disease: Is p53 the missing link between genetic and sporadic Parkinsonism? Cell Signal 2011, 23, 963–968. [Google Scholar] [CrossRef] [PubMed]
  194. Nair, V.D. Activation of p53 signaling initiates apoptotic death in a cellular model of Parkinson’s disease. Apoptosis 2006, 11, 955–966. [Google Scholar] [CrossRef] [PubMed]
  195. Mogi, M.; Kondo, T.; Mizuno, Y.; Nagatsu, T. p53 protein, interferon-gamma, and NF-kappaB levels are elevated in the parkinsonian brain. Neurosci. Lett. 2007, 414, 94–97. [Google Scholar] [CrossRef] [PubMed]
  196. Komarov, P.G.; Komarova, E.A.; Kondratov, R.V.; Christov-Tselkov, K.; Coon, J.S.; Chernov, M.V.; Gudkov, A.V. A chemical inhibitor of p53 that protects mice from the side effects of cancer therapy. Science 1999, 285, 1733–1737. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  197. Biswas, S.C.; Ryu, E.J.; Park, C.H.; Malagelada, C.; Greene, L.A. Puma and p53 play required roles in death-evoked in a cellular model of Parkinson disease. Neurochem. Res. 2005, 30, 839–845. [Google Scholar] [CrossRef] [PubMed]
  198. Alves da Costa, C.; Duplan, E.; Checler, F. α-synuclein and p53 functional interplay in physiopathological contexts. Oncotarget 2017, 8, 9001–9002. [Google Scholar] [CrossRef] [Green Version]
  199. Qi, X.; Davis, B.; Chiang, Y.H.; Filichia, E.; Barnett, A.; Greig, N.H.; Hoffer, B.; Luo, Y. Dopaminergic neuron-specific deletion of p53 gene is neuroprotective in an experimental Parkinson’s disease model. J. Neurochem. 2016, 138, 746–757. [Google Scholar] [CrossRef] [Green Version]
  200. Alves da Costa, C.; Ancolio, K.; Checler, F. Wild-type but not Parkinson’s disease-related Ala53Thr-a-synuclein protect neuronal cells from apoptotic stimuli. J. Biol. Chem 2000, 275, 24065–24069. [Google Scholar] [CrossRef] [Green Version]
  201. da Costa, C.A.; Masliah, E.; Checler, F. Beta-synuclein displays an antiapoptotic p53-dependent phenotype and protects neurons from 6-hydroxydopamine-induced caspase 3 activation: Cross-talk with alpha-synuclein and implication for Parkinson’s disease. J. Biol. Chem. 2003, 278, 37330–37335. [Google Scholar] [CrossRef] [Green Version]
  202. Duplan, E.; Giordano, C.; Checler, F.; Alves da Costa, C. Direct alpha-synuclein promoter transactivation by the tumor suppressor p53. Mol. Neurodegener. 2016, 11, 13. [Google Scholar] [CrossRef] [Green Version]
  203. Chen, H.Y.; Lin, C.H.; Teng, S.C. Stress-induced p53 drives BAG5 cochaperone expression to control alpha-synuclein aggregation in Parkinson’s disease. Aging 2020, 12, 20702–20727. [Google Scholar] [CrossRef] [PubMed]
  204. da Costa, C.A.; Sunyach, C.; Giaime, E.; West, A.; Corti, O.; Brice, A.; Safe, S.; Abou-Sleiman, P.M.; Wood, N.W.; Takahashi, H.; et al. Transcriptional repression of p53 by parkin and impairment by mutations associated with autosomal recessive juvenile Parkinson’s disease. Nat. Cell Biol. 2009, 11, 1370–1375. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  205. Sunico, C.R.; Nakamura, T.; Rockenstein, E.; Mante, M.; Adame, A.; Chan, S.F.; Newmeyer, T.F.; Masliah, E.; Nakanishi, N.; Lipton, S.A. S-Nitrosylation of parkin as a novel regulator of p53-mediated neuronal cell death in sporadic Parkinson’s disease. Mol. Neurodegener. 2013, 8, 29. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  206. Yao, D.; Gu, Z.; Nakamura, T.; Shi, Z.-Q.; Ma, Y.; Gaston, B.; Palmer, L.A.; Rockenstein, E.M.; Zhang, Z.; Masliah, E.; et al. Nitrosative stress linked to sporadic Parkinson’s disease: S-nitrosylation of parkin regulates its E3 ubiquitin ligase activity. Proc. Natl. Acad. Sci. USA 2004, 101, 10810–10814. [Google Scholar] [CrossRef] [Green Version]
  207. Viotti, J.; Duplan, E.; Caillava, C.; Condat, J.; Goiran, T.; Giordano, C.; Marie, Y.; Idbaih, A.; Delattre, J.Y.; Honnorat, J.; et al. Glioma tumor grade correlates with parkin depletion in mutant p53-linked tumors and results from loss of function of p53 transcriptional activity. Oncogene 2014, 33, 1764–1775. [Google Scholar] [CrossRef] [Green Version]
  208. Zhang, C.; Lin, M.; Wu, R.; Wang, X.; Yang, B.; Levine, A.J.; Hu, W.; Feng, Z. Parkin, a p53 target gene, mediates the role of p53 in glucose metabolism and the Warburg effect. Proc. Natl. Acad. Sci. USA 2011, 108, 16259–16264. [Google Scholar] [CrossRef] [Green Version]
  209. Ottolini, D.; Cali, T.; Negro, A.; Brini, M. The Parkinson disease-related protein DJ-1 counteracts mitochondrial impairment induced by the tumour suppressor protein p53 by enhancing endoplasmic reticulum-mitochondria tethering. Hum. Mol. Genet. 2013, 22, 2152–2168. [Google Scholar] [CrossRef]
  210. Duplan, E.; Giaime, E.; Viotti, J.; Sevalle, J.; Corti, O.; Brice, A.; Ariga, H.; Qi, L.; Checler, F.; da Costa, C.A. ER-stress-associated functional link between Parkin and DJ-1 via a transcriptional cascade involving the tumor suppressor p53 and the spliced X-box binding protein XBP-1. J. Cell Sci. 2013, 126, 2124–2133. [Google Scholar] [CrossRef] [Green Version]
  211. Goiran, T.; Duplan, E.; Rouland, L.; El Manaa, W.; Lauritzen, I.; Dunys, J.; You, H.; Checler, F.; Alves da Costa, C. Nuclear p53-mediated repression of autophagy involves PINK1 transcriptional down-regulation. Cell Death Differ. 2018, 25, 873–884. [Google Scholar] [CrossRef] [Green Version]
  212. Liu, K.; Lee, J.; Kim, J.Y.; Wang, L.; Tian, Y.; Chan, S.T.; Cho, C.; Machida, K.; Chen, D.; Ou, J.J. Mitophagy Controls the Activities of Tumor Suppressor p53 to Regulate Hepatic Cancer Stem Cells. Mol. Cell 2017, 68, 281–292.e5. [Google Scholar] [CrossRef] [Green Version]
  213. Zhu, X.; Raina, A.K.; Perry, G.; Smith, M.A. Apoptosis in Alzheimer disease: A mathematical improbability. Curr. Alzheimer Res. 2006, 3, 393–396. [Google Scholar] [CrossRef] [PubMed]
  214. Obulesu, M.; Lakshmi, M.J. Apoptosis in Alzheimer’s disease: An understanding of the physiology, pathology and therapeutic avenues. Neurochem. Res. 2014, 39, 2301–2312. [Google Scholar] [CrossRef] [PubMed]
  215. Abate, G.; Frisoni, G.B.; Bourdon, J.C.; Piccirella, S.; Memo, M.; Uberti, D. The pleiotropic role of p53 in functional/dysfunctional neurons: Focus on pathogenesis and diagnosis of Alzheimer’s disease. Alzheimer’s Res. Ther. 2020, 12, 160. [Google Scholar] [CrossRef] [PubMed]
  216. Kitamura, Y.; Shimohama, S.; Kamoshima, W.; Matsuoka, Y.; Nomura, Y.; Taniguchi, T. Changes of p53 in the brains patients with Alzheimer disease. Biochem. Biophys. Res. Comm. 1997, 232, 418–421. [Google Scholar] [CrossRef] [PubMed]
  217. Lanni, C.; Racchi, M.; Mazzini, G.; Ranzenigo, A.; Polotti, R.; Sinforiani, E.; Olivari, L.; Barcikowska, M.; Styczynska, M.; Kuznicki, J.; et al. Conformationally altered p53: A novel Alzheimer’s disease marker? Mol. Psychiatry 2008, 13, 641–647. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  218. Chen, L.; Liu, S.; Tao, Y. Regulating tumor suppressor genes: Post-translational modifications. Signal Transduct. Target. Ther. 2020, 5, 90. [Google Scholar] [CrossRef] [PubMed]
  219. Ohyagi, Y.; Asahara, H.; Chui, D.H.; Tsuruta, Y.; Sakae, N.; Miyoshi, K.; Yamada, T.; Kikuchi, H.; Taniwaki, T.; Murai, H.; et al. Intracellular Abeta42 activates p53 promoter: A pathway to neurodegeneration in Alzheimer’s disease. FASEB J. 2005, 19, 255–257. [Google Scholar] [CrossRef] [PubMed]
  220. Checler, F.; Dunys, J.; Pardossi-Piquard, R.; Alves da Costa, C. p53 Is Regulated by and Regulates Members of the gamma-Secretase Complex. Neurodegener. Dis. 2010, 7, 50–55. [Google Scholar] [CrossRef]
  221. Alves da Costa, C.; Sunyach, C.; Pardossi-Piquard, R.; Sevalle, J.; Vincent, B.; Boyer, N.; Kawarai, T.; Girardot, N.; St George-Hyslop, P.; Checler, F. Presenilin-dependent gamma-secretase-mediated control of p53-associated cell death in Alzheimer’s disease. J. Neurosci. 2006, 26, 6377–6385. [Google Scholar] [CrossRef] [Green Version]
  222. Alves da Costa, C.; Mattson, M.P.; Ancolio, K.; Checler, F. The C-terminal fragment of presenilin 2 triggers p53-mediated staurosporine-induced apoptosis, a function independent of the presenilinase-derived N-terminal counterpart. J. Biol. Chem. 2003, 278, 12064–12069. [Google Scholar] [CrossRef] [Green Version]
  223. Alves da Costa, C.; Paitel, E.; Mattson, M.P.; Amson, R.; Telerman, A.; Ancolio, K.; Checler, F. Wild-type and mutated presenilins 2 trigger p53-dependent apoptosis and down-regulate presenilin 1 expression in HEK293 human cells and in murine neurons. Proc. Natl. Acad. Sci. USA 2002, 99, 4043–4048. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  224. Takasugi, N.; Tomita, T.; Hayashi, I.; Tsuruoka, M.; Niimura, M.; Takahashi, Y.; Thinakaran, G.; Iwatsubo, T. The role of presenilin cofactors in the g-secretase complex. Nature 2003, 422, 438–441. [Google Scholar] [CrossRef] [PubMed]
  225. Edbauer, D.; Winkler, E.; Regula, J.T.; Pesold, B.; Steiner, H.; Haass, C. Reconstitution of g-secretase activity. Nat. Cell Biol. 2003, 5, 486–488. [Google Scholar] [CrossRef] [PubMed]
  226. Dunys, J.; Kawarai, T.; Sevalle, J.; Dolcini, V.; St George-Hyslop, P.; Alves da Costa, C.; Checler, F. p53-dependent Aph-1 and Pen-2 anti-apoptotic phenotype requires the integrity of the gamma-secretase complex but is independent of its activity. J. Biol. Chem. 2007, 282, 10516–10525. [Google Scholar] [CrossRef] [Green Version]
  227. Pardossi-Piquard, R.; Dunys, J.; Giaime, E.; Guillot-Sestier, M.V.; St George-Hyslop, P.; Checler, F.; Alves da Costa, C. p53-dependent control of cell death by nicastrin: Lack of requirement for presenilin-dependent gamma-secretase complex. J. Neurochem. 2009, 109, 225–237. [Google Scholar] [CrossRef]
  228. Cuesta, A.; Zambrano, A.; Royo, M.; Pascual, A. The tumor suppressor p53 regulates the expression of the beta-amyloid precursor protein. Biochem. J. 2008, 418, 643–650. [Google Scholar] [CrossRef] [Green Version]
  229. Pardossi-Piquard, R.; Checler, F. The physiology of the beta-amyloid precursor protein intracellular domain AICD. J. Neurochem. 2012, 120 (Suppl. 1), 109–124. [Google Scholar] [CrossRef]
  230. Lu, D.C.; Rabizadeh, S.; Chandra, S.; Shayya, R.F.; Ellerby, L.M.; Ye, X.; Salvesen, G.S.; Koo, E.H.; Bredesen, D.E. A second cytotoxic proteolytic peptide derived from amyloid beta-protein precursor. Nat. Med. 2000, 6, 397–404. [Google Scholar] [CrossRef]
  231. Ozaki, T.; Li, Y.; Kikuchi, H.; Tomita, T.; Iwatsubo, T.; Nakagawara, A. The intracellular domain of the amyloid precursor protein (AICD) enhances the p53-mediated apoptosis. Biochem. Biophys. Res. Commun. 2006, 351, 57–63. [Google Scholar] [CrossRef]
  232. Li, M.; Pehar, M.; Liu, Y.; Bhattacharyya, A.; Zhang, S.C.; O’Riordan, K.J.; Burger, C.; D’Adamio, L.; Puglielli, L. The amyloid precursor protein (APP) intracellular domain regulates translation of p44, a short isoform of p53, through an IRES-dependent mechanism. Neurobiol. Aging 2015, 36, 2725–2736. [Google Scholar] [CrossRef] [Green Version]
  233. Dumanchin-Njock, C.; Alves da Costa, C.A.; Mercken, L.; Pradier, L.; Checler, F. The caspase-derived C-terminal fragment of betaAPP induces caspase-independent toxicity and triggers selective increase of Abeta42 in mammalian cells. J. Neurochem. 2001, 78, 1153–1161. [Google Scholar] [CrossRef] [PubMed]
  234. Park, S.A.; Shaked, G.M.; Bredesen, D.E.; Koo, E.H. Mechanism of cytotoxicity mediated by the C31 fragment of the Amyloid Precursor Protein. Biochem. Biophys. Res. Commun. 2009, 388, 450–455. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  235. Khandelwal, P.J.; Moussa, C.E. The Relationship between Parkin and Protein Aggregation in Neurodegenerative Diseases. Front. Psychiatry 2010, 1, 15. [Google Scholar] [CrossRef] [PubMed]
  236. Zhang, C.W.; Hang, L.; Yao, T.P.; Lim, K.L. Parkin Regulation and Neurodegenerative Disorders. Front. Aging Neurosci. 2015, 7, 248. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  237. Witte, M.E.; Bol, J.G.; Gerritsen, W.H.; van der Valk, P.; Drukarch, B.; van Horssen, J.; Wilhelmus, M.M. Parkinson’s disease-associated parkin colocalizes with Alzheimer’s disease and multiple sclerosis brain lesions. Neurobiol. Dis. 2009, 36, 445–452. [Google Scholar] [CrossRef]
  238. Kumar, D.; Kumar, P. Abeta, Tau, and alpha-Synuclein aggregation and integrated role of PARK2 in the regulation and clearance of toxic peptides. Neuropeptides 2019, 78, 101971. [Google Scholar] [CrossRef]
  239. Goudarzi, S.; Hosseini, A.; Abdollahi, M.; Haghi-Aminjan, H. Insights Into Parkin-Mediated Mitophagy in Alzheimer’s Disease: A Systematic Review. Front. Aging Neurosci. 2021, 13, 674071. [Google Scholar] [CrossRef]
  240. Ye, X.; Sun, X.; Starovoytov, V.; Cai, Q. Parkin-mediated mitophagy in mutant hAPP neurons and Alzheimer’s disease patient brains. Hum. Mol. Genet. 2015, 24, 2938–2951. [Google Scholar] [CrossRef]
  241. Wang, H.; Fu, J.; Xu, X.; Yang, Z.; Zhang, T. Rapamycin Activates Mitophagy and Alleviates Cognitive and Synaptic Plasticity Deficits in a Mouse Model of Alzheimer’s Disease. J. Gerontol. Ser. A 2021, 76, 1707–1713. [Google Scholar] [CrossRef]
  242. Martin-Maestro, P.; Gargini, R.; Perry, G.; Avila, J.; Garcia-Escudero, V. PARK2 enhancement is able to compensate mitophagy alterations found in sporadic Alzheimer’s disease. Hum. Mol. Genet. 2016, 25, 792–806. [Google Scholar] [CrossRef] [Green Version]
  243. Wang, H.; Zhang, T.; Ge, X.; Chen, J.; Zhao, Y.; Fu, J. Parkin overexpression attenuates Abeta-induced mitochondrial dysfunction in HEK293 cells by restoring impaired mitophagy. Life Sci. 2020, 244, 117322. [Google Scholar] [CrossRef] [PubMed]
  244. Khandelwal, P.J.; Herman, A.M.; Hoe, H.S.; Rebeck, G.W.; Moussa, C.E. Parkin mediates beclin-dependent autophagic clearance of defective mitochondria and ubiquitinated Abeta in AD models. Hum. Mol. Genet. 2011, 20, 2091–2102. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  245. Algarzae, N.; Hebron, M.; Miessau, M.; Moussa, C.E. Parkin prevents cortical atrophy and Abeta-induced alterations of brain metabolism: (1)(3)C NMR and magnetic resonance imaging studies in AD models. Neuroscience 2012, 225, 22–34. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  246. Rosen, K.M.; Moussa, C.E.; Lee, H.K.; Kumar, P.; Kitada, T.; Qin, G.; Fu, Q.; Querfurth, H.W. Parkin reverses intracellular beta-amyloid accumulation and its negative effects on proteasome function. J. Neurosci. Res. 2010, 88, 167–178. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  247. Hong, X.; Liu, J.; Zhu, G.; Zhuang, Y.; Suo, H.; Wang, P.; Huang, D.; Xu, J.; Huang, Y.; Yu, M.; et al. Parkin overexpression ameliorates hippocampal long-term potentiation and beta-amyloid load in an Alzheimer’s disease mouse model. Hum. Mol. Genet. 2014, 23, 1056–1072. [Google Scholar] [CrossRef] [Green Version]
  248. Rosen, K.M.; Veereshwarayya, V.; Moussa, C.E.; Fu, Q.; Goldberg, M.S.; Schlossmacher, M.G.; Shen, J.; Querfurth, H.W. Parkin protects against mitochondrial toxins and beta-amyloid accumulation in skeletal muscle cells. J. Biol. Chem. 2006, 281, 12809–12816. [Google Scholar] [CrossRef] [Green Version]
  249. Kam, M.K.; Lee, D.G.; Kim, B.; Huh, J.W.; Lee, H.J.; Park, Y.H.; Lee, D.S. Amyloid-beta oligomers induce Parkin-mediated mitophagy by reducing Miro1. Biochem J 2020, 477, 4581–4597. [Google Scholar] [CrossRef]
  250. Solano, R.M.; Casarejos, M.J.; Gomez, A.; Perucho, J.; de Yebenes, J.G.; Mena, M.A. Parkin null cortical neuronal/glial cultures are resistant to amyloid-beta1-42 toxicity: A role for autophagy? J. Alzheimer’s Dis. 2012, 32, 57–76. [Google Scholar] [CrossRef] [Green Version]
  251. Han, Y.; Wang, N.; Kang, J.; Fang, Y. beta-Asarone improves learning and memory in Abeta1-42-induced Alzheimer’s disease rats by regulating PINK1-Parkin-mediated mitophagy. Metab. Brain Dis. 2020, 35, 1109–1117. [Google Scholar] [CrossRef]
  252. Wang, N.; Wang, H.; Li, L.; Li, Y.; Zhang, R. beta-Asarone Inhibits Amyloid-beta by Promoting Autophagy in a Cell Model of Alzheimer’s Disease. Front. Pharmacol. 2019, 10, 1529. [Google Scholar] [CrossRef]
  253. Jiang, Y.; Li, H.; Huang, P.; Li, S.; Li, B.; Huo, L.; Zhong, J.; Pan, Z.; Li, Y.; Xia, X. Panax notoginseng saponins protect PC12 cells against Abeta induced injury via promoting parkin-mediated mitophagy. J. Ethnopharmacol. 2022, 285, 114859. [Google Scholar] [CrossRef] [PubMed]
  254. Lonskaya, I.; Hebron, M.L.; Desforges, N.M.; Schachter, J.B.; Moussa, C.E. Nilotinib-induced autophagic changes increase endogenous parkin level and ubiquitination, leading to amyloid clearance. J. Mol. Med. 2014, 92, 373–386. [Google Scholar] [CrossRef] [PubMed]
  255. Hu, Y.; Li, X.C.; Wang, Z.H.; Luo, Y.; Zhang, X.; Liu, X.P.; Feng, Q.; Wang, Q.; Yue, Z.; Chen, Z.; et al. Tau accumulation impairs mitophagy via increasing mitochondrial membrane potential and reducing mitochondrial Parkin. Oncotarget 2016, 7, 17356–17368. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  256. Cummins, N.; Tweedie, A.; Zuryn, S.; Bertran-Gonzalez, J.; Gotz, J. Disease-associated tau impairs mitophagy by inhibiting Parkin translocation to mitochondria. EMBO J. 2019, 38, e99360. [Google Scholar] [CrossRef]
  257. Moussa, C.E. Parkin attenuates wild-type tau modification in the presence of beta-amyloid and alpha-synuclein. J. Mol. Neurosci. 2009, 37, 25–36. [Google Scholar] [CrossRef]
  258. Baxter, L.L.; Moran, T.H.; Richtsmeier, J.T.; Troncoso, J.; Reeves, R.H. Discovery and genetic localization of Down syndrome cerebellar phenotypes using the Ts65Dn mouse. Hum. Mol. Genet. 2000, 9, 195–202. [Google Scholar] [CrossRef] [Green Version]
  259. Henrique, A.M.; Gianetti, N.G.; Ferrari, M.F.R. Parkin is downregulated among autophagy-related proteins prior to hyperphosphorylation of Tau in TS65DN mice. Biochem. Biophys. Res. Commun. 2021, 561, 59–64. [Google Scholar] [CrossRef]
  260. Guerrero, R.; Navarro, P.; Gallego, E.; Avila, J.; de Yebenes, J.G.; Sanchez, M.P. Park2-null/tau transgenic mice reveal a functional relationship between parkin and tau. J. Alzheimer’s Dis. 2008, 13, 161–172. [Google Scholar] [CrossRef]
  261. Menendez, J.; Rodriguez-Navarro, J.A.; Solano, R.M.; Casarejos, M.J.; Rodal, I.; Guerrero, R.; Sanchez, M.P.; Avila, J.; Mena, M.A.; de Yebenes, J.G. Suppression of Parkin enhances nigrostriatal and motor neuron lesion in mice over-expressing human-mutated tau protein. Hum. Mol. Genet. 2006, 15, 2045–2058. [Google Scholar] [CrossRef] [Green Version]
  262. Rodriguez-Navarro, J.A.; Gomez, A.; Rodal, I.; Perucho, J.; Martinez, A.; Furio, V.; Ampuero, I.; Casarejos, M.J.; Solano, R.M.; de Yebenes, J.G.; et al. Parkin deletion causes cerebral and systemic amyloidosis in human mutated tau over-expressing mice. Hum. Mol. Genet. 2008, 17, 3128–3143. [Google Scholar] [CrossRef] [Green Version]
  263. Guerrero, R.; Navarro, P.; Gallego, E.; Garcia-Cabrero, A.M.; Avila, J.; Sanchez, M.P. Hyperphosphorylated tau aggregates in the cortex and hippocampus of transgenic mice with mutant human FTDP-17 Tau and lacking the PARK2 gene. Acta Neuropathol. 2009, 117, 159–168. [Google Scholar] [CrossRef] [PubMed]
  264. Vargas, D.M.; De Bastiani, M.A.; Zimmer, E.R.; Klamt, F. Alzheimer’s disease master regulators analysis: Search for potential molecular targets and drug repositioning candidates. Alzheimer’s Res. Ther. 2018, 10, 59. [Google Scholar] [CrossRef] [PubMed]
  265. Duplan, E.; Sevalle, J.; Viotti, J.; Goiran, T.; Bauer, C.; Renbaum, P.; Levy-Lahad, E.; Gautier, C.A.; Corti, O.; Leroudier, N.; et al. Parkin differently regulates presenilin-1 and presenilin-2 functions by direct control of their promoter transcription. J. Mol. Cell Biol. 2013, 5, 132–142. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  266. Lauritzen, I.; Pardossi-Piquard, R.; Bauer, C.; Brigham, E.; Abraham, J.D.; Ranaldi, S.; Fraser, P.; St-George-Hyslop, P.; Le Thuc, O.; Espin, V.; et al. The beta-secretase-derived C-terminal fragment of betaAPP, C99, but not Abeta, is a key contributor to early intraneuronal lesions in triple-transgenic mouse hippocampus. J. Neurosci. 2012, 32, 16243–16255. [Google Scholar] [CrossRef]
  267. Pulina, M.V.; Hopkins, M.; Haroutunian, V.; Greengard, P.; Bustos, V. C99 selectively accumulates in vulnerable neurons in Alzheimer’s disease. Alzheimer’s Dement. 2020, 16, 273–282. [Google Scholar] [CrossRef]
  268. Lauritzen, I.; Becot, A.; Bourgeois, A.; Pardossi-Piquard, R.; Biferi, M.G.; Barkats, M.; Checler, F. Targeting gamma-secretase triggers the selective enrichment of oligomeric APP-CTFs in brain extracellular vesicles from Alzheimer cell and mouse models. Transl. Neurodegener. 2019, 8, 35. [Google Scholar] [CrossRef] [Green Version]
  269. Rouland, L.; Duplan, E.; Ramos Dos Santos, L.; Bernardin, A.; Katula, K.S.; Manfioletti, G.; Idbaih, A.; Checler, F.; Alves da Costa, C. Therapeutic potential of parkin as a tumor suppressor via transcriptional control of cyclins in glioblastoma cell and animal models. Theranostics 2021, 11, 10047–10063. [Google Scholar] [CrossRef]
  270. Pardossi-Piquard, R.; Petit, A.; Kawarai, T.; Sunyach, C.; Alves da Costa, C.; Vincent, B.; Ring, S.; D’Adamio, L.; Shen, J.; Muller, U.; et al. Presenilin-dependent transcriptional control of the Abeta-degrading enzyme neprilysin by intracellular domains of betaAPP and APLP. Neuron 2005, 46, 541–554. [Google Scholar] [CrossRef] [Green Version]
  271. Ghosal, K.; Vogt, D.L.; Liang, M.; Shen, Y.; Lamb, B.T.; Pimplikar, S.W. Alzheimer’s disease-like pathological features in transgenic mice expressing the APP intracellular domain. Proc. Natl. Acad. Sci. USA 2009, 106, 18367–18372. [Google Scholar] [CrossRef] [Green Version]
  272. Cisse, M.; Duplan, E.; Checler, F. The transcription factor XBP1 in memory and cognition: Implications in Alzheimer disease. Mol. Med. 2017, 22, 905–917. [Google Scholar] [CrossRef]
  273. Cisse, M.; Duplan, E.; Lorivel, T.; Dunys, J.; Bauer, C.; Meckler, X.; Gerakis, Y.; Lauritzen, I.; Checler, F. The transcription factor XBP1s restores hippocampal synaptic plasticity and memory by control of the Kalirin-7 pathway in Alzheimer model. Mol. Psychiatry 2017, 22, 1562–1575. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Influence of parkin reduction in XBP1S-linked unfolded protein response (UPR), p53-dependent cell death, long-term potentiation and long-term depression (synaptic plasticity), mitophagy, catabolism of Aβ and Tau proteins in mouse primary cultured neurons as well as in AD-affected human fibroblasts and sporadic AD-affected brains.
Figure 1. Influence of parkin reduction in XBP1S-linked unfolded protein response (UPR), p53-dependent cell death, long-term potentiation and long-term depression (synaptic plasticity), mitophagy, catabolism of Aβ and Tau proteins in mouse primary cultured neurons as well as in AD-affected human fibroblasts and sporadic AD-affected brains.
Biomolecules 12 00559 g001
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Checler, F.; Alves da Costa, C. Parkin as a Molecular Bridge Linking Alzheimer’s and Parkinson’s Diseases? Biomolecules 2022, 12, 559. https://doi.org/10.3390/biom12040559

AMA Style

Checler F, Alves da Costa C. Parkin as a Molecular Bridge Linking Alzheimer’s and Parkinson’s Diseases? Biomolecules. 2022; 12(4):559. https://doi.org/10.3390/biom12040559

Chicago/Turabian Style

Checler, Frédéric, and Cristine Alves da Costa. 2022. "Parkin as a Molecular Bridge Linking Alzheimer’s and Parkinson’s Diseases?" Biomolecules 12, no. 4: 559. https://doi.org/10.3390/biom12040559

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