Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 3621 2022-10-21 10:49:00 |
2 format Meta information modification 3621 2022-10-24 05:49:21 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Ravinther, A.I.;  Dewadas, H.D.;  Tong, S.R.;  Foo, C.N.;  Lin, Y.;  Chien, C.;  Lim, Y.M. Molecular Pathways Involved in LRRK2-Linked Parkinson’s Disease. Encyclopedia. Available online: https://encyclopedia.pub/entry/30692 (accessed on 27 July 2024).
Ravinther AI,  Dewadas HD,  Tong SR,  Foo CN,  Lin Y,  Chien C, et al. Molecular Pathways Involved in LRRK2-Linked Parkinson’s Disease. Encyclopedia. Available at: https://encyclopedia.pub/entry/30692. Accessed July 27, 2024.
Ravinther, Ailyn Irvita, Hemaniswarri Dewi Dewadas, Shi Ruo Tong, Chai Nien Foo, Yu-En Lin, Cheng-Ting Chien, Yang Mooi Lim. "Molecular Pathways Involved in LRRK2-Linked Parkinson’s Disease" Encyclopedia, https://encyclopedia.pub/entry/30692 (accessed July 27, 2024).
Ravinther, A.I.,  Dewadas, H.D.,  Tong, S.R.,  Foo, C.N.,  Lin, Y.,  Chien, C., & Lim, Y.M. (2022, October 21). Molecular Pathways Involved in LRRK2-Linked Parkinson’s Disease. In Encyclopedia. https://encyclopedia.pub/entry/30692
Ravinther, Ailyn Irvita, et al. "Molecular Pathways Involved in LRRK2-Linked Parkinson’s Disease." Encyclopedia. Web. 21 October, 2022.
Molecular Pathways Involved in LRRK2-Linked Parkinson’s Disease
Edit

Parkinson’s disease is one of the most common neurodegenerative diseases affecting the ageing population. Studies have found that mutations in Leucine-rich-repeat-kinase 2 (LRRK2) are the most common cause of familial Parkinson’s disease (PD). Moreover, aberrant (higher) LRRK2 kinase activity has an influence in idiopathic PD as well. 

Parkinson’s disease mechanism LRRK2

1. Introduction

Parkinson’s disease (PD) is the second most common neuropathology after Alzheimer’s disease [1]. It has the fastest-growing prevalence and most deaths among neurological disorders, affecting nearly 6.1 million ageing people in 2016 globally, which is more than doubled from the past generation [2]. PD is a multisystem disease that affects the nervous system, especially the central nervous system (CNS), with the pathological characteristic of losing more than 50% of the dopaminergic neurons in the substantia nigra pars compacta [3][4]. Dopaminergic neurons are the main source of producing rich dopamine (DA). The depletion of DA manifests in the progressive degeneration of motor mobility with the clinical symptoms of bradykinesia, rigidity, rest tremor, and postural instability [1][5]. In addition, the neuropathology of PD also exhibits heterogeneous clinical symptoms in non-motor function (deficit in olfactory, depression and cognition, rapid eye movement sleep behaviour disorder, constipation, and central autonomic control) at all stages of the PD pathological process, even predating the motor symptoms [6].
PD is considered as multifactorial neuropathology with complex aetiology, which was found to be highly correlated with ageing, abnormal alpha-synuclein (α-Syn) accumulation in dopaminergic neurons, exposure to polluted environment, and genetic susceptibility [1][3][7]. However, the risk of PD pathogenesis was found to interplay with multiple different factors and eventually give rise to different clinical symptoms. More often, the disease mechanisms may not be completely identical in all PD patients. Hence, the complexity of PD disease increases the difficulty to reveal a clearer pathogenesis pathway.
To date, there are only approximately 10–15% of patients reported as family history of PD symptoms, while the remaining 85–90% of the PD population are classified as sporadic PD [1][8]. More than 20 genes were found to be involved in familial PD, in which the penetrant mutation is rare, with an occurrence of 2% in the PD population [7][8][9]. These mutations present a broad range of risks and demonstrate heterogeneous mechanisms associated with PD. Among the PD causative genes, the 51-exon Leucine-rich repeat kinase (LRRK2) gene is the most common mutation, where the variants are reported in 0.7% of all the people showing PD symptoms [8]. Hence, a missense mutation in LRRK2 G2019S received the most attention because it appears to be the most penetrant mutation in the autosomal dominant PD and sporadic PD, with an influence of 40% and 10%, respectively [4][5][10]. Moreover, the occurrence of LRRK2 variants affecting a wide span of diverse ethnicities in different geographic distributions further define its role of genetic susceptibility in the PD population [11].
There are nearly 100 mutations identified at the different domains of LRRK2, in which some are related to the pathogenesis of PD, including G2019S, R1441C/G/H, I2020T, and Y1699C. The mutation sites are highly conserved and segregate in families with PD disease [5][8][10]. These mutations lead to gain-of-function mechanisms, where kinase activity of LRRK2 will increase to phosphorylate a group of Rab proteins in different subcellular localities, thus affecting downstream pathways to drive PD pathogenesis [14]. Interestingly, in in vitro and in vivo studies, knocking out LRRK2 recapitulated aspects of PD pathogenesis such as dopaminergic neuron loss, accumulation of α-Syn, impairment of protein degradation, and dysregulation of autophagy [15][16][17][18]. However, there is a lack of evidence of phenotypic impact, as there was no increase in the risk of PD in humans carrying loss of function variants of LRRK2 [19].
Many studies also show that the aberrant function of LRRK2 to PD is not necessary by mutation alone, instead, some upstream factors such as Rab29 and α-Syn could exacerbate the activity of LRRK2 [14][20][21]. Therefore, evidence suggests that the mutant LRRK2 is inter-related with other proteins and impacts diverse cellular biological processes, increasing the complexity and enigma of the understanding of PD pathogenesis.

2. Main Mechanisms of LRRK2-Associated PD

The main finding in pre-clinical studies of LRRK2-PD is that elevated LRRK2 kinase activity or phosphorylation of targets is linked to various aspects of pathogenesis.
Firstly, LRRK2 causes α-Syn neurotoxicity by decreased clearance of α-Syn and increased propagation and aggregate formation of α-Syn in a kinase-dependent manner. In the transgenic models with G2019S, α-Syn neurotoxicity induction is reversible with LRRK2 kinase inhibition, but not reversible in non-transgenic models [22]. Intriguingly, in models of synucleinopathy, LRRK2 inhibition can restore neuronal loss and motor deficits but worsens α-Syn clearance [23]. The similarity between these two studies is that LRRK2 inhibition is ineffective in preventing α-Syn neurotoxicity in the absence of pathogenic LRRK2. This suggests that though LRRK2 hyperactivation is present in idiopathic PD, it is not a main contributor to α-Syn neurotoxicity, but rather other activities lead of LRRK2 lead to neurodegeneration. The differences in these findings could be attributed to different mouse models. The former utilized an α-Syn PFF PD mouse model, whereas the latter utilized human α-Syn overexpressing transgenic mice. While the studies in this field have attributed neurotoxicity to α-Syn inclusion, some studies suggest α-Syn aggregation is a protective mechanism [24]. Rather than being a causative factor, Lewy body pathology could be an accompaniment of neuronal death. Therefore, the reversal of neurotoxicity with LRRK2 kinase inhibition could be associated with other exacerbated activities of LRRK2.
LRRK2 kinase activity modulates calcineurin-independent and calcineurin-dependent pathways [23][25]. LRRK2 impacts both inflammatory pathways, as it can phosphorylate RCAN1, which is an inhibitor of calcineurin and activates NFATc2, a downstream substrate of calcineurin [26]. Other inflammatory pathways influenced by kinase activities of LRRK2 include upregulation of AGE-RAGE, which activates the NF-κB signalling pathway [27]. The NF-κB signalling pathway is indirectly upregulated in microglia by LRRK2, as LRRK2 decreases NF-κB inhibitory signalling by downregulating PKA activity [28].
In addition to contributing to inflammatory pathways, glia to neuron crosstalk induces neurotoxicity through other ways. In both astrocytes and microglia, both WT and pathogenic LRRK2 had a decreased ability to clear α-Syn [29][30]. This accumulation of α-Syn in astrocytes can be linked to how LRRK2-G2019S compromises CMA and macroautophagy [31]. LRRK2 G2019S astrocytes had a decreased ability in responding to oxidative stress through distinct means, SERCA inactivation and the SHP (receptor)-PIAS1 (protein)-XBP1 (transcription factor) pathway [32][33]. In LRRK2 KO microglia, there was a decrease in antioxidant response after treatment with pre-formed fibrils of αSYN [34]. These three results suggest that diversion of typical activities of LRRK2, i.e., the hyperactivation of kinase, will affect LRRK2 redox signalling. LRRK2 also facilitates microglial activation in a kinase-dependent manner [35]. A newer study found that LRRK2 G2019S impacts microglia response to inflammation by Rab8a function in iron uptake and transport [36].
Mitochondrial dysfunction and ER stress are the main mechanisms in PD. LRRK2 causes it through two main ways: calcium dyshomeostasis and defects in the mitochondria life cycle. Calcium dyshomeostasis in the mitochondria and ER was exacerbated by LRRK2 mutants G2019S, Y1699C, and R1441C and by the knockout of LRRK2 [37][38][39]. This shows that elevated LRRK2 activity will not exclusively impact calcium homeostasis, as a loss of physiological LRRK2 activity impacts calcium homeostasis as well. Mutations of LRRK2 such as G2019S and R1441G cause defects in the mitochondrial life cycle by the inhibition of mitophagy [40][41][42]. In two of the three studies, one involving G2019S and another with R1441G, kinase inhibition was unable to reverse the defects in mitophagy, suggesting that factors aside from kinase hyperactivity reduce mitophagy [40][42]. The R1441G study involved LRRK2 mutant mice, whereas the G2019S studies used patient fibroblasts. The difference in response to inhibition of kinase activity despite the utility of similar models could be because of differences in monitoring of mitophagy in human primary fibroblasts. The former study focused on measuring Miro1 intensity as opposed to mitophagy induction [40][41]. In general, the life cycle of the mitochondria is affected by LRRK2 because expression of the enzymatic core of LRRK2 alone (kinase and COR domain) leads to reduced mitochondrial biogenesis [43].
Many downstream substrates of LRRK2 are involved in vesicle trafficking. A delicate balance of activities of LRRK2 is needed in physiological conditions for vesicular transport, particularly in microtubule-mediated vesicular transport events [44]. The increase in phosphorylation of downstream factors of LRRK2 such as SJN1, auxilin, and Synapsin 1 is linked to dysfunction in synaptic vesicle trafficking [45][46][47]. Defects in autophagy and lysosomal processes are caused by LRRK2 kinase hyperactivity, disassociation of normal binding partners of LRRK2, altered recruitment of AV proteins, and phosphorylation of Rab10.
Downstream Rab GTPases of LRRK2, including Rab10, play a pertinent role in LRRK2-PD pathology. From the studies in this field, Rab5, Rab7, Rab10, Rab29, and Rab35 were linked to various aspects of PD pathogenesis in a kinase-dependent manner. Rab5 and Rab35 were linked to α-Syn neurotoxicity by the downregulation of endosomal formation [29][48][49]. The negative regulation of CME was due to Rab5, Rab7, and Rab10 [50]. Lysosomal stress and impaired autolysosome formation was linked to Rab10 and Rab29, respectively [51][52]. In addition, the interference of ciliogenesis was mediated by binding of RILPL1 to pRab10 [53][54]. A separate study found that pRab8a and pRab10 were associated with RILPL1 and pRab8a and pRab12 was linked to RILPL2-related cilia defects [55]. Other PD-related phenotypes linked to Rab GTPases include impaired mitophagy. The phosphorylation of Rab10 prevented a necessary accumulation of Rab10 at the mitochondria that was necessary to induce mitophagy [56]. Of the five Rab GTPases implicated in LRRK2-PD, Rab10 has a role in several mechanisms and was found to be relevant in other forms of familial PD, including PINK1 and GBA1 [56][57]. This suggests a convergence of pathways with Rab10 as a substrate. Furthermore, pRab10 elevation is detectable in clinical samples of both LRRK2-PD and iPD patients [58]. Studies have been conducted to investigate the mechanisms that influence the phosphorylation of Rab10 by LRRK2. PPM1H, a phosphatase, was found to be a negative regulator of Rab10 phosphorylation [59]. The differential distribution of PPM1H in tissues could be an explanation for the variation of pRab10 in clinical samples [58].
Lastly, a downstream target of LRRK2 is tau. Previous studies have shown the in-vitro hyperphosphorylation of tau by LRRK2 [60][61][62]. Expression of LRRK2 G2019S in Drosophila promoted the phosphorylation of tau by the recruitment of glycogen synthase kinase 3β (GSK3β) [63]. The accumulation of insoluble and phosphorylated tau was observed in a transgenic LRRK2 mouse model of tauopathy [62]. Studies in this field reveal contrasting findings to previous knowledge of LRRK2 and tau. Two studies, despite different cell types, show that tau pathology observed in LRRK2-PD is kinase-independent [64][65]. Rather than the LRRK2 kinase activity contributing to tau neurotoxicity, it is attributed to proteosomal impairment and actin polymerization [64][66]. Two separate in vivo studies found that the oligomerization of LRRK2 promoted by its mutant forms resulted in overstabilization of F-actin, which has consequences for the accumulation and spread of proteins such as tau and α-Syn [66][67]. Overall, despite a lack of consensus that the kinase activities of LRRK2 directly impact tau pathology, there is evidence that LRRK2 promotes the spread of tau pathology.

3. LRRK2 Role in iPD and Familial PD

Studies reported in this field show that in the presence or absence of pathogenic variants, LRRK2 can contribute to the pathogenesis of PD. Comparisons were drawn from SH-SY5Y cells transfected with LRRK2 mutants, fibroblasts of LRRK2-PD patients, and post mortem brains of sporadic PD patients. Elevated protein levels associated with increased susceptibility to mitochondrial calcium dysregulation were observed for all groups, suggesting a shared mechanism between LRRK2-PD and sporadic PD [37]. Furthermore, kinase inhibition was able to reduce translational impairment in fibroblasts derived from both LRRK2 familial and sporadic PD patients [68]. It was also demonstrated that in patients with idiopathic PD, there was enhanced kinase activity of WT LRRK2 which was associated with abnormalities in mitochondria and lysosomal function [69]. Therefore, targeting LRRK2 activities may have broad therapeutic utility in idiopathic PD, not only in those who carry a LRRK2 mutation.

4. Potential Biomarkers in LRRK2-PD

The most significant biomarker in the detection of PD would be the increased kinase activity of LRRK2. LRRK2 kinase activity can be quantified by its autophosphorylation at serine1292 or the levels of downstream pRab10. In a Norwegian cohort, elevated levels of s1292 were identified in male patients that were carriers of G2019S [70]. These elevated pS1292 levels were found in brain and urinary exosomes. Future studies are needed to investigate pS1292 levels in larger, more diverse cohorts and to explore other LRRK2-PD mutations.
In this field, some studies highlight the possibility of novel biomarkers in LRRK2-associated PD. In vitro and in vivo studies have identified the overexpression of Rab35 and linked it to α-Syn pathology [48][49]. In addition to that, elevated mtDNA damage was found in iPD and LRRK2-PD patient fibroblasts [71]. Impairment of translation was also observed in fibroblasts of iPD and LRRK2-G2019S patients [68]. Further studies with human cohorts are needed to assess if these biomarkers are detectable in patient serum.

5. Therapeutic Strategies for LRRK2-Associated PD

5.1. Direct Inhibition of LRRK2

Direct inhibition of LRRK2 kinase activity has a potential to impact other forms of familial PD. In this field, LRRK2 G2019S and GBA1-N370S-derived patient astrocytes shared hallmarks of PD [72]. Pre-clinical studies have shown that LRRK2 kinase inhibition can restore defects in GBA1 D490V mutant astrocytes [73]. The same group found that while defects caused by loss of GBA1 function can be attenuated by LRRK2 inhibition, the activity of the enzyme coded by GBA1, β-Glucocerebrosidase (GCase), is unaffected [74]. However, in that study, instead of mutant GBA1, GBA1-heterozygous-null iPSC-derived neurons were used, whereas in DA neurons derived from LRRK2-PD patients with G2019S or R1441C mutations, a reduction in GCase activity was observed in a manner mediated by Rab10 [75]. In contrast to the previous study, GCase activity in DA neurons with LRRK2 or GBA1 mutations can be increased with LRRK2 kinase inhibition. A study in 2022 corroborated these findings to some extent. In G2019S iPSC-derived neurons, GCase protein level was reduced, but in patient-derived fibroblasts and peripheral blood mononuclear cells, a positive correlation between LRRK2 kinase and GCase activity was found [57]. These studies indicate that LRRK2 kinase plays a role in regulating GCase activity, though there are differences specific to cell type. As these studies collectively suggest an interplay between these two PD-related genes, a case for GCase activation in LRRK2-PD can be also made.
While most of the studies in this field focus on the role of LRRK2 G2019S in PD, other variants of LRRK2 that are not directly linked to the kinase function of LRRK2 play a role in the pathogenesis of PD. Like G2019S, R1441G in the GTPase domain can cause mitochondrial defects through Drp1 [42]. Protein synthesis deficiency, vesicle trafficking dysfunction, and autolysosomal dysfunction are linked to R1441C/G mutations. Hence, inhibitors of the GTP binding activities of LRRK2 have been proposed as a different approach. However, at present only pre-clinical work justifies its therapeutic potential. In SH-SY5Y cells expressing LRRK2 R1441C, a GTPase domain mutation, impairments were seen in the neurite transport of mitochondria and lysosomes. Treatment with GTP-binding inhibitors were able to prevent these defects [76]. GTP-binding inhibitors could also reduce kinase activity. LRRK2 GTP-binding inhibitors 68 and Fx2149 promoted LRRK2 ubiquitination, increased ubiquitinated aggregation, and contributed to an aggresomal response. This could be linked to improved clearance of protein aggregates [77].
An alternative to reducing LRRK2 activity would be to reduce total levels of LRRK2 protein. Pre-clinical studies have shown that LRRK2 antisense oligonucleotides (ASO) can ameliorate α-Syn pathology, reduce elevated levels of ER Ca2+ and defects in mitophagy, and reduce locomotor deficits [78][79][80]. Furthermore, treatment of LRRK2 ASO in transgenic mice expressing LRRK2 G2019S or WT LRRK2 was able to decrease phosphorylation of Rab10 and correct autophagic defects caused by LRRK2 [80]. Currently, there is an ongoing phase I clinical trial for BIIB09, a LRRK2 ASO [81].
Studies exploring the physiological role of LRRK2 have described a role in immune response—T-cell function as well as the innate immune response [82][83][84]. LRRK2 kinase activity as a negative regulator of macroautophagy has been previously described [85]. Therefore, inhibition of LRRK2 activities or total reduction in protein could dysregulate immune or vesicle trafficking functions. Therefore, apart from direct inhibition of LRRK2 activity and protein level, potential pharmacological interventions involving inhibition or upregulation of LRRK2 targets could be considered.

5.2. Indirect Inhibition of LRRK2

Inhibition of signalling pathways linked to neurodegeneration could be considered as a potential therapeutic intervention. There are signalling pathways activated or upregulated by LRRK2 linked to neurodegeneration. ASK1, an upstream regulator of P38 and JNK, when directly phosphorylated by LRRK2, results in neuronal apoptosis [86]. Therefore, ASK1 inhibitors which are currently undergoing clinical trials, some in phase III, could be used in the context of PD or broadly in neurodegenerative diseases [87]. Another inhibitor that is proposed are JNK inhibitors. These inhibitors, mainly in phase I trials, are being developed as anti-tumorigenic agents [88]. JNK inhibitors may be used in combination with other PD drugs. A study found that pathogenic LRRK2 results in JNK activation which led to motor impairment and neuronal death in a Drosophila model [89]. JNK inhibition alone only partially attenuated neurotoxicity [89]. Hence, combination therapy of LRRK2 inhibitors and JNK inhibitors would be more effective. Another signalling pathway that can be targeted is the AGE-RAGE intracellular signalling. G2019 enhances the interaction between AGE and RAGE, leading to inflammation and oxidative stress [27]. In a pre-clinical rat study, an anti-RAGE antibody was found to be able to block systemic inflammatory responses [90]. However, as it was unable to cross the blood–brain barrier, it may have limited applicability in the context of PD.
Upstream to LRRK2, there are genes causing its aberrant expression. Some in vitro studies have shown that the knockdown of these genes can attenuate neurotoxicity in LRRK2-associated PD [91][92]. Two long non-coding RNA, HOTAIR and MALAT1, increase the expression of LRRK2, thereby increasing its activity as well. Since gene knockdown is not feasible in humans, alternatives need to be explored. Both MALAT1 and HOTAIR have been proposed as therapeutic targets in cancer. Anti-lncRNAs were able to suppress HOTAIR activity in targeting solid tumours in vivo [93]. To suppress MALAT1 activity, siRNAs were able to elicit a degradation of MALAT1 [94]. A similar approach could be used to inhibit these lncRNA in PD models to assess its applicability. TXNIP is synergistic with LRRK2 and causes ER stress by preventing the release of antioxidants and is linked to α-Syn pathology. In human 3D midbrain organoids, inhibition of TXNIP was found to reduce LRRK2-induced phenotypes [95].
Another strategy is targeting downstream mechanisms such as mitochondrial dysfunctions. Firstly, pre-clinical work has shown that the reduction in Miro levels can attenuate neurotoxicity, as that allows damaged mitochondria to be degraded [96]. Secondly, the suppression of PERK can prevent degradation of ER–mitochondria contacts, which is necessary for mitochondrial health. In a mouse model for frontotemporal dementia, PERK inhibition was able to prevent further neuronal loss and lower levels of phosphorylated tau [97]. Thirdly, SERCA is a potential target as well. LRRK2 deactivates SERCA by direct association, which leads to ER stress and mitochondrial dysfunction [32]. As kinase inhibition of LRRK2 does not activate SERCA, another approach must be conducted, such as the pharmacological activation of SERCA through an allosteric activator. Current pre-clinical work utilizes SERCA activators for diseases such as type-2 diabetes and Duchenne Muscular Dystrophy [98][99]. Lastly, Drp1 inhibition could be a strategy as well to prevent neuronal death linked to mitochondrial dysfunction. A pre-clinical study found that Drp1 inhibition protected against neurotoxicity [100].
Antisense oligonucleotides (ASOs) are a potential strategy to reduce the total protein levels of LRRK2 and other interacting proteins. ASOs for α-Syn are being studied in pre-clinical models and have been found to prevent neurodegeneration associated with LRRK2 [101]. Another protein target implicated in LRRK2-PD, tau, which is abundant in carries of LRRK2 mutations, could also be addressed with a similar approach [102]. The use of tau ASOs could be necessary to address tau pathology in LRRK2-PD, as some studies have shown that it is LRRK2 kinase-independent [64][65].
While many of these targets are synergistic with LRRK2, there are some that are antagonistic targets. In upregulating the activity or levels of these targets, it may reverse or reduce the damage caused by overactivated LRRK2. There were two targets found to be upstream of LRRK2: Prx2 and Fbxl18. Prx2 was found to be an upstream inhibitor of LRRK2, as it could reduce pRab10 levels, a downstream effector of LRRK2 [103]. It is effectively an inhibitor of the kinase activities of LRRK2. Fbxl18 is able to target phosphorylated LRRK2 for degradation, which may be beneficial as self-phosphorylation of LRRK2 is also reflective of its kinase activities [104]. There were also factors that were not upstream of LRRK2 but could mediate the downstream effects of LRRK2. In pre-clinical studies, the upregulation of NCLX was able to help with Ca2+ influx caused by LRRK2 [38]. SP1 is a factor associated with α-Syn that has opposite effects on neuronal health compared to mutant LRRK2 [105]. Upregulating the activity of SP1 could be considered as a strategy in pre-clinical work. Lastly, the restoration of auxilin or DNAJC6 was able to prevent synaptic vesicle endocytosis dysfunction caused by LRRK2 [46]. Clathrin-mediated endocytosis (CME) is a key process mediated by auxilin; this could explain the functional antagonism between LRRK2 and auxilin [106].

References

  1. Kouli, A.; Torsney, K.M.; Kuan, W.-L. Parkinson’s Disease: Etiology, Neuropathology, and Pathogenesis. In Parkinson’s Disease: Pathogenesis and Clinical Aspects; Stoker, T.B., Greenland, J.C., Eds.; Exon Publications: Brisbane, Queensland, Australia, 2018; ISBN 978-0-9944381-6-4.
  2. GBD 2016 Parkinson’s Disease Collaborators. Global, Regional, and National Burden of Parkinson’s Disease, 1990–2016: A Systematic Analysis for the Global Burden of Disease Study 2016. Lancet Neurol. 2018, 17, 939–953.
  3. Maiti, P.; Manna, J.; Dunbar, G.L. Current Understanding of the Molecular Mechanisms in Parkinson’s Disease: Targets for Potential Treatments. Transl. Neurodegener. 2017, 6, 28.
  4. Hur, E.-M.; Lee, B.D. LRRK2 at the Crossroad of Aging and Parkinson’s Disease. Genes 2021, 12, 505.
  5. Ren, C.; Ding, Y.; Wei, S.; Guan, L.; Zhang, C.; Ji, Y.; Wang, F.; Yin, S.; Yin, P. G2019S Variation in LRRK2: An Ideal Model for the Study of Parkinson’s Disease? Front. Hum. Neurosci. 2019, 13, 306.
  6. Chaudhuri, K.R.; Odin, P. The Challenge of Non-Motor Symptoms in Parkinson’s Disease. Prog. Brain Res. 2010, 184, 325–341.
  7. Klein, C.; Westenberger, A. Genetics of Parkinson’s Disease. Cold Spring Harb. Perspect. Med. 2012, 2, a008888.
  8. Tran, J.; Anastacio, H.; Bardy, C. Genetic Predispositions of Parkinson’s Disease Revealed in Patient-Derived Brain Cells. NPJ Parkinson’s Dis. 2020, 6, 8.
  9. Blauwendraat, C.; Nalls, M.A.; Singleton, A.B. The Genetic Architecture of Parkinson’s Disease. Lancet Neurol. 2020, 19, 170–178.
  10. Biskup, S.; West, A.B. Zeroing in on LRRK2-Linked Pathogenic Mechanisms in Parkinson’s Disease. Biochim. Biophys. Acta 2009, 1792, 625–633.
  11. Shu, L.; Zhang, Y.; Sun, Q.; Pan, H.; Tang, B. A Comprehensive Analysis of Population Differences in LRRK2 Variant Distribution in Parkinson’s Disease. Front. Aging Neurosci. 2019, 11, 13.
  12. Guaitoli, G.; Raimondi, F.; Gilsbach, B.K.; Gómez-Llorente, Y.; Deyaert, E.; Renzi, F.; Li, X.; Schaffner, A.; Jagtap, P.K.A.; Boldt, K.; et al. Structural Model of the Dimeric Parkinson’s Protein LRRK2 Reveals a Compact Architecture Involving Distant Interdomain Contacts. Proc. Natl. Acad. Sci. USA 2016, 113, E4357–E4366.
  13. 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.
  14. Berwick, D.C.; Heaton, G.R.; Azeggagh, S.; Harvey, K. LRRK2 Biology from Structure to Dysfunction: Research Progresses, but the Themes Remain the Same. Mol. Neurodegener. 2019, 14, 49.
  15. Tong, Y.; Yamaguchi, H.; Giaime, E.; Boyle, S.; Kopan, R.; Kelleher, R.J.; Shen, J. Loss of Leucine-Rich Repeat Kinase 2 Causes Impairment of Protein Degradation Pathways, Accumulation of Alpha-Synuclein, and Apoptotic Cell Death in Aged Mice. Proc. Natl. Acad. Sci. USA 2010, 107, 9879–9884.
  16. Giaime, E.; Tong, Y.; Wagner, L.K.; Yuan, Y.; Huang, G.; Shen, J. Age-Dependent Dopaminergic Neurodegeneration and Impairment of the Autophagy-Lysosomal Pathway in LRRK-Deficient Mice. Neuron 2017, 96, 796–807.
  17. Albanese, F.; Mercatelli, D.; Finetti, L.; Lamonaca, G.; Pizzi, S.; Shimshek, D.R.; Bernacchia, G.; Morari, M. Constitutive Silencing of LRRK2 Kinase Activity Leads to Early Glucocerebrosidase Deregulation and Late Impairment of Autophagy in Vivo. Neurobiol. Dis. 2021, 159, 105487.
  18. Huang, G.; Bloodgood, D.W.; Kang, J.; Shahapal, A.; Chen, P.; Kaganovsky, K.; Kim, J.-I.; Ding, J.B.; Shen, J. Motor Impairments and Dopaminergic Defects Caused by Loss of Leucine-Rich Repeat Kinase Function in Mice. J. Neurosci. 2022, 42, 4755–4765.
  19. Blauwendraat, C.; Reed, X.; Kia, D.A.; Gan-Or, Z.; Lesage, S.; Pihlstrøm, L.; Guerreiro, R.; Gibbs, J.R.; Sabir, M.; Ahmed, S.; et al. Frequency of Loss of Function Variants in LRRK2 in Parkinson Disease. JAMA Neurol. 2018, 75, 1416–1422.
  20. Purlyte, E.; Dhekne, H.S.; Sarhan, A.R.; Gomez, R.; Lis, P.; Wightman, M.; Martinez, T.N.; Tonelli, F.; Pfeffer, S.R.; Alessi, D.R. Rab29 Activation of the Parkinson’s Disease-Associated LRRK2 Kinase. EMBO J. 2018, 37, 1–18.
  21. O’Hara, D.M.; Pawar, G.; Kalia, S.K.; Kalia, L.V. LRRK2 and α-Synuclein: Distinct or Synergistic Players in Parkinson’s Disease? Front. Neurosci. 2020, 14, 577.
  22. Henderson, M.X.; Sengupta, M.; McGeary, I.; Zhang, B.; Olufemi, M.F.; Brown, H.; Trojanowski, J.Q.; Lee, V.M.Y. LRRK2 Inhibition Does Not Impart Protection from α-Synuclein Pathology and Neuron Death in Non-Transgenic Mice. Acta Neuropathol. Commun. 2019, 7, 28.
  23. Kim, C.; Beilina, A.; Smith, N.; Li, Y.; Kim, M.; Kumaran, R.; Kaganovich, A.; Mamais, A.; Adame, A.; Iba, M.; et al. LRRK2 Mediates Microglial Neurotoxicity via NFATc2 in Rodent Models of Synucleinopathies. Sci. Transl. Med. 2020, 12, eaay0399.
  24. Melachroinou, K.; Leandrou, E.; Valkimadi, P.-E.; Memou, A.; Hadjigeorgiou, G.; Stefanis, L.; Rideout, H.J. Activation of FADD-Dependent Neuronal Death Pathways as a Predictor of Pathogenicity for LRRK2 Mutations. PLoS ONE 2016, 11, e0166053.
  25. Han, K.A.; Yoo, L.; Sung, J.Y.; Chung, S.A.; Um, J.W.; Kim, H.; Seol, W.; Chung, K.C. Leucine-Rich Repeat Kinase 2 (LRRK2) Stimulates IL-1β-Mediated Inflammatory Signaling through Phosphorylation of RCAN1. Front. Cell. Neurosci. 2017, 11, 125.
  26. Martínez-Martínez, S.; Genescà, L.; Rodríguez, A.; Raya, A.; Salichs, E.; Were, F.; López-Maderuelo, M.D.; Redondo, J.M.; de la Luna, S. The RCAN Carboxyl End Mediates Calcineurin Docking-Dependent Inhibition via a Site That Dictates Binding to Substrates and Regulators. Proc. Natl. Acad. Sci. USA 2009, 106, 6117–6122.
  27. Cho, H.J.; Xie, C.; Cai, H. AGE-Induced Neuronal Cell Death Is Enhanced in G2019S LRRK2 Mutation with Increased RAGE Expression. Transl. Neurodegener. 2018, 7, 1.
  28. Russo, I.; Di Benedetto, G.; Kaganovich, A.; Ding, J.; Mercatelli, D.; Morari, M.; Cookson, M.R.; Bubacco, L.; Greggio, E. Leucine-Rich Repeat Kinase 2 Controls Protein Kinase A Activation State through Phosphodiesterase 4. J. Neuroinflammation 2018, 15, 297.
  29. Maekawa, T.; Sasaoka, T.; Azuma, S.; Ichikawa, T.; Melrose, H.L.; Farrer, M.J.; Obata, F. Leucine-Rich Repeat Kinase 2 (LRRK2) Regulates α-Synuclein Clearance in Microglia. BMC Neurosci. 2016, 17, 77.
  30. Streubel-Gallasch, L.; Giusti, V.; Sandre, M.; Tessari, I.; Plotegher, N.; Giusto, E.; Masato, A.; Iovino, L.; Battisti, I.; Arrigoni, G.; et al. Parkinson’s Disease-Associated LRRK2 Interferes with Astrocyte-Mediated Alpha-Synuclein Clearance. Mol. Neurobiol. 2021, 58, 3119–3140.
  31. Di Domenico, A.; Carola, G.; Calatayud, C.; Pons-Espinal, M.; Muñoz, J.P.; Richaud-Patin, Y.; Fernandez-Carasa, I.; Gut, M.; Faella, A.; Parameswaran, J.; et al. Patient-Specific IPSC-Derived Astrocytes Contribute to Non-Cell-Autonomous Neurodegeneration in Parkinson’s Disease. Stem Cell Rep. 2019, 12, 213–229.
  32. Lee, J.H.; Han, J.-H.; Kim, H.; Park, S.M.; Joe, E.-H.; Jou, I. Parkinson’s Disease-Associated LRRK2-G2019S Mutant Acts through Regulation of SERCA Activity to Control ER Stress in Astrocytes. Acta Neuropathol. Commun. 2019, 7, 68.
  33. Lee, J.H.; Han, J.-H.; Joe, E.-H.; Jou, I. Small Heterodimer Partner (SHP) Aggravates ER Stress in Parkinson’s Disease-Linked LRRK2 Mutant Astrocyte by Regulating XBP1 SUMOylation. J. Biomed. Sci. 2021, 28, 51.
  34. Russo, I.; Kaganovich, A.; Ding, J.; Landeck, N.; Mamais, A.; Varanita, T.; Biosa, A.; Tessari, I.; Bubacco, L.; Greggio, E.; et al. Transcriptome Analysis of LRRK2 Knock-out Microglia Cells Reveals Alterations of Inflammatory- and Oxidative Stress-Related Pathways upon Treatment with α-Synuclein Fibrils. Neurobiol. Dis. 2019, 129, 67–78.
  35. Ho, D.H.; Je, A.R.; Lee, H.; Son, I.; Kweon, H.-S.; Kim, H.-G.; Seol, W. LRRK2 Kinase Activity Induces Mitochondrial Fission in Microglia via Drp1 and Modulates Neuroinflammation. Exp. Neurobiol. 2018, 27, 171–180.
  36. Mamais, A.; Kluss, J.H.; Bonet-Ponce, L.; Landeck, N.; Langston, R.G.; Smith, N.; Beilina, A.; Kaganovich, A.; Ghosh, M.C.; Pellegrini, L.; et al. Correction: Mutations in LRRK2 Linked to Parkinson Disease Sequester Rab8a to Damaged Lysosomes and Regulate Transferrin-Mediated Iron Uptake in Microglia. PLoS Biol. 2022, 20, e3001621.
  37. Verma, M.; Callio, J.; Otero, P.A.; Sekler, I.; Wills, Z.P.; Chu, C.T. Mitochondrial Calcium Dysregulation Contributes to Dendrite Degeneration Mediated by PD/LBD-Associated LRRK2 Mutants. J. Neurosci. 2017, 37, 11151–11165.
  38. Ludtmann, M.H.R.; Kostic, M.; Horne, A.; Gandhi, S.; Sekler, I.; Abramov, A.Y. LRRK2 Deficiency Induced Mitochondrial Ca2+ Efflux Inhibition Can Be Rescued by Na+/Ca2+/Li+ Exchanger Upregulation. Cell Death Dis. 2019, 10, 265.
  39. Kim, J.W.; Yin, X.; Jhaldiyal, A.; Khan, M.R.; Martin, I.; Xie, Z.; Perez-Rosello, T.; Kumar, M.; Abalde-Atristain, L.; Xu, J.; et al. Defects in MRNA Translation in LRRK2-Mutant HiPSC-Derived Dopaminergic Neurons Lead to Dysregulated Calcium Homeostasis. Cell Stem Cell 2020, 27, 633–645.
  40. Hsieh, C.-H.; Shaltouki, A.; Gonzalez, A.E.; Bettencourt da Cruz, A.; Burbulla, L.F.; St Lawrence, E.; Schüle, 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.
  41. Bonello, F.; Hassoun, S.-M.; Mouton-Liger, F.; Shin, Y.S.; Muscat, A.; Tesson, C.; Lesage, S.; Beart, P.M.; Brice, A.; Krupp, J.; et al. LRRK2 Impairs PINK1/Parkin-Dependent Mitophagy via Its Kinase Activity: Pathologic Insights into Parkinson’s Disease. Hum. Mol. Genet. 2019, 28, 1645–1660.
  42. Liu, H.; Ho, P.W.-L.; Leung, C.-T.; Pang, S.Y.-Y.; Chang, E.E.S.; Choi, Z.Y.-K.; Kung, M.H.-W.; Ramsden, D.B.; Ho, S.-L. Aberrant Mitochondrial Morphology and Function Associated with Impaired Mitophagy and DNM1L-MAPK/ERK Signaling Are Found in Aged Mutant Parkinsonian LRRK2R1441G Mice. Autophagy 2021, 17, 3196–3220.
  43. Aufschnaiter, A.; Kohler, V.; Walter, C.; Tosal-Castano, S.; Habernig, L.; Wolinski, H.; Keller, W.; Vögtle, F.-N.; Büttner, S. The Enzymatic Core of the Parkinson’s Disease-Associated Protein LRRK2 Impairs Mitochondrial Biogenesis in Aging Yeast. Front. Mol. Neurosci. 2018, 11, 205.
  44. Blanca Ramírez, M.; Lara Ordóñez, A.J.; Fdez, E.; Madero-Pérez, J.; Gonnelli, A.; Drouyer, M.; Chartier-Harlin, M.-C.; Taymans, J.-M.; Bubacco, L.; Greggio, E.; et al. GTP Binding Regulates Cellular Localization of Parkinson’s Disease-Associated LRRK2. Hum. Mol. Genet. 2017, 26, 2747–2767.
  45. Pan, P.-Y.; Li, X.; Wang, J.; Powell, J.; Wang, Q.; Zhang, Y.; Chen, Z.; Wicinski, B.; Hof, P.; Ryan, T.A.; et al. Parkinson’s Disease-Associated LRRK2 Hyperactive Kinase Mutant Disrupts Synaptic Vesicle Trafficking in Ventral Midbrain Neurons. J. Neurosci. 2017, 37, 11366–11376.
  46. Nguyen, M.; Krainc, D. LRRK2 Phosphorylation of Auxilin Mediates Synaptic Defects in Dopaminergic Neurons from Patients with Parkinson’s Disease. Proc. Natl. Acad. Sci. USA 2018, 115, 5576–5581.
  47. Marte, A.; Russo, I.; Rebosio, C.; Valente, P.; Belluzzi, E.; Pischedda, F.; Montani, C.; Lavarello, C.; Petretto, A.; Fedele, E.; et al. Leucine-Rich Repeat Kinase 2 Phosphorylation on Synapsin I Regulates Glutamate Release at Pre-Synaptic Sites. J. Neurochem. 2019, 150, 264–281.
  48. Bae, E.-J.; Kim, D.-K.; Kim, C.; Mante, M.; Adame, A.; Rockenstein, E.; Ulusoy, A.; Klinkenberg, M.; Jeong, G.R.; Bae, J.R.; et al. LRRK2 Kinase Regulates α-Synuclein Propagation via RAB35 Phosphorylation. Nat. Commun. 2018, 9, 3465.
  49. Chiu, C.-C.; Yeh, T.-H.; Lai, S.-C.; Weng, Y.-H.; Huang, Y.-C.; Cheng, Y.-C.; Chen, R.-S.; Huang, Y.-Z.; Hung, J.; Chen, C.-C.; et al. Increased Rab35 Expression Is a Potential Biomarker and Implicated in the Pathogenesis of Parkinson’s Disease. Oncotarget 2016, 7, 54215–54227.
  50. Connor-Robson, N.; Booth, H.; Martin, J.G.; Gao, B.; Li, K.; Doig, N.; Vowles, J.; Browne, C.; Klinger, L.; Juhasz, P.; et al. An Integrated Transcriptomics and Proteomics Analysis Reveals Functional Endocytic Dysregulation Caused by Mutations in LRRK2. Neurobiol. Dis. 2019, 127, 512–526.
  51. Kuwahara, T.; Funakawa, K.; Komori, T.; Sakurai, M.; Yoshii, G.; Eguchi, T.; Fukuda, M.; Iwatsubo, T. Roles of Lysosomotropic Agents on LRRK2 Activation and Rab10 Phosphorylation. Neurobiol. Dis. 2020, 145, 105081.
  52. Boecker, C.A.; Goldsmith, J.; Dou, D.; Cajka, G.G.; Holzbaur, E.L.F. Increased LRRK2 Kinase Activity Alters Neuronal Autophagy by Disrupting the Axonal Transport of Autophagosomes. Curr. Biol. CB 2021, 31, 2140–2154.
  53. Dhekne, H.S.; Yanatori, I.; Gomez, R.C.; Tonelli, F.; Diez, F.; Schüle, B.; Steger, M.; Alessi, D.R.; Pfeffer, S.R. A Pathway for Parkinson’s Disease LRRK2 Kinase to Block Primary Cilia and Sonic Hedgehog Signaling in the Brain. Elife 2018, 7, e40202.
  54. Dhekne, H.S.; Yanatori, I.; Vides, E.G.; Sobu, Y.; Diez, F.; Tonelli, F.; Pfeffer, S.R. LRRK2-Phosphorylated Rab10 Sequesters Myosin Va with RILPL2 during Ciliogenesis Blockade. Life Sci. Alliance 2021, 4, e202101050.
  55. Steger, M.; Diez, F.; Dhekne, H.S.; Lis, P.; Nirujogi, R.S.; Karayel, O.; Tonelli, F.; Martinez, T.N.; Lorentzen, E.; Pfeffer, S.R.; et al. Systematic Proteomic Analysis of LRRK2-Mediated Rab GTPase Phosphorylation Establishes a Connection to Ciliogenesis. Elife 2017, 6, e31012.
  56. Wauters, F.; Cornelissen, T.; Imberechts, D.; Martin, S.; Koentjoro, B.; Sue, C.; Vangheluwe, P.; Vandenberghe, W. LRRK2 Mutations Impair Depolarization-Induced Mitophagy through Inhibition of Mitochondrial Accumulation of RAB10. Autophagy 2020, 16, 203–222.
  57. Kedariti, M.; Frattini, E.; Baden, P.; Cogo, S.; Civiero, L.; Ziviani, E.; Zilio, G.; Bertoli, F.; Aureli, M.; Kaganovich, A.; et al. LRRK2 Kinase Activity Regulates GCase Level and Enzymatic Activity Differently Depending on Cell Type in Parkinson’s Disease. NPJ Parkinson’s Dis. 2022, 8, 92.
  58. Petropoulou-Vathi, L.; Simitsi, A.; Valkimadi, P.-E.; Kedariti, M.; Dimitrakopoulos, L.; Koros, C.; Papadimitriou, D.; Papadimitriou, A.; Stefanis, L.; Alcalay, R.N.; et al. Distinct Profiles of LRRK2 Activation and Rab GTPase Phosphorylation in Clinical Samples from Different PD Cohorts. NPJ Parkinson’s Dis. 2022, 8, 73.
  59. Berndsen, K.; Lis, P.; Yeshaw, W.M.; Wawro, P.S.; Nirujogi, R.S.; Wightman, M.; Macartney, T.; Dorward, M.; Knebel, A.; Tonelli, F.; et al. PPM1H Phosphatase Counteracts LRRK2 Signaling by Selectively Dephosphorylating Rab Proteins. Elife 2019, 8, e50416.
  60. Kawakami, F.; Yabata, T.; Ohta, E.; Maekawa, T.; Shimada, N.; Suzuki, M.; Maruyama, H.; Ichikawa, T.; Obata, F. LRRK2 Phosphorylates Tubulin-Associated Tau but Not the Free Molecule: LRRK2-Mediated Regulation of the Tau-Tubulin Association and Neurite Outgrowth. PLoS ONE 2012, 7, e30834.
  61. Ujiie, S.; Hatano, T.; Kubo, S.-I.; Imai, S.; Sato, S.; Uchihara, T.; Yagishita, S.; Hasegawa, K.; Kowa, H.; Sakai, F.; et al. LRRK2 I2020T Mutation Is Associated with Tau Pathology. Parkinsonism Relat. Disord. 2012, 18, 819–823.
  62. Bailey, R.M.; Covy, J.P.; Melrose, H.L.; Rousseau, L.; Watkinson, R.; Knight, J.; Miles, S.; Farrer, M.J.; Dickson, D.W.; Giasson, B.I.; et al. LRRK2 Phosphorylates Novel Tau Epitopes and Promotes Tauopathy. Acta Neuropathol. (Berl.) 2013, 126, 809–827.
  63. Lin, C.-H.; Tsai, P.-I.; Wu, R.-M.; Chien, C.-T. LRRK2 G2019S Mutation Induces Dendrite Degeneration through Mislocalization and Phosphorylation of Tau by Recruiting Autoactivated GSK3ß. J. Neurosci. 2010, 30, 13138–13149.
  64. Guerreiro, P.S.; Gerhardt, E.; Lopes da Fonseca, T.; Bähr, M.; Outeiro, T.F.; Eckermann, K. LRRK2 Promotes Tau Accumulation, Aggregation and Release. Mol. Neurobiol. 2016, 53, 3124–3135.
  65. Henderson, M.X.; Changolkar, L.; Trojanowski, J.Q.; Lee, V.M.Y. LRRK2 Kinase Activity Does Not Alter Cell-Autonomous Tau Pathology Development in Primary Neurons. J. Park. Dis. 2021, 11, 1187–1196.
  66. Bardai, F.H.; Ordonez, D.G.; Bailey, R.M.; Hamm, M.; Lewis, J.; Feany, M.B. Lrrk Promotes Tau Neurotoxicity through Dysregulation of Actin and Mitochondrial Dynamics. PLoS Biol. 2018, 16, e2006265.
  67. Sarkar, S.; Bardai, F.; Olsen, A.L.; Lohr, K.M.; Zhang, Y.-Y.; Feany, M.B. Oligomerization of Lrrk Controls Actin Severing and α-Synuclein Neurotoxicity in Vivo. Mol. Neurodegener. 2021, 16, 33.
  68. Deshpande, P.; Flinkman, D.; Hong, Y.; Goltseva, E.; Siino, V.; Sun, L.; Peltonen, S.; Elo, L.L.; Kaasinen, V.; James, P.; et al. Protein Synthesis Is Suppressed in Sporadic and Familial Parkinson’s Disease by LRRK2. FASEB J. 2020, 34, 14217–14233.
  69. Di Maio, R.; Hoffman, E.K.; Rocha, E.M.; Keeney, M.T.; Sanders, L.H.; De Miranda, B.R.; Zharikov, A.; Van Laar, A.; Stepan, A.F.; Lanz, T.A.; et al. LRRK2 Activation in Idiopathic Parkinson’s Disease. Sci. Transl. Med. 2018, 10, eaar5429.
  70. Wang, S.; Liu, Z.; Ye, T.; Mabrouk, O.S.; Maltbie, T.; Aasly, J.; West, A.B. Elevated LRRK2 Autophosphorylation in Brain-Derived and Peripheral Exosomes in LRRK2 Mutation Carriers. Acta Neuropathol. Commun. 2017, 5, 86.
  71. Podlesniy, P.; Puigròs, M.; Serra, N.; Fernández-Santiago, R.; Ezquerra, M.; Tolosa, E.; Trullas, R. Accumulation of Mitochondrial 7S DNA in Idiopathic and LRRK2 Associated Parkinson’s Disease. EBioMedicine 2019, 48, 554–567.
  72. Sonninen, T.-M.; Hämäläinen, R.H.; Koskuvi, M.; Oksanen, M.; Shakirzyanova, A.; Wojciechowski, S.; Puttonen, K.; Naumenko, N.; Goldsteins, G.; Laham-Karam, N.; et al. Metabolic Alterations in Parkinson’s Disease Astrocytes. Sci. Rep. 2020, 10, 14474.
  73. Sanyal, A.; DeAndrade, M.P.; Novis, H.S.; Lin, S.; Chang, J.; Lengacher, N.; Tomlinson, J.J.; Tansey, M.G.; LaVoie, M.J. Lysosome and Inflammatory Defects in GBA1-Mutant Astrocytes Are Normalized by LRRK2 Inhibition. Mov. Disord. 2020, 35, 760–773.
  74. Sanyal, A.; Novis, H.S.; Gasser, E.; Lin, S.; LaVoie, M.J. LRRK2 Kinase Inhibition Rescues Deficits in Lysosome Function Due to Heterozygous GBA1 Expression in Human IPSC-Derived Neurons. Front. Neurosci. 2020, 14, 442.
  75. Ysselstein, D.; Nguyen, M.; Young, T.J.; Severino, A.; Schwake, M.; Merchant, K.; Krainc, D. LRRK2 Kinase Activity Regulates Lysosomal Glucocerebrosidase in Neurons Derived from Parkinson’s Disease Patients. Nat. Commun. 2019, 10, 5570.
  76. Thomas, J.M.; Li, T.; Yang, W.; Xue, F.; Fishman, P.S.; Smith, W.W. 68 and FX2149 Attenuate Mutant LRRK2-R1441C-Induced Neural Transport Impairment. Front. Aging Neurosci. 2016, 8, 337.
  77. Thomas, J.M.; Wang, X.; Guo, G.; Li, T.; Dai, B.; Nucifora, L.G.; Nucifora, F.C.; Liu, Z.; Xue, F.; Liu, C.; et al. GTP-binding Inhibitors Increase LRRK2-linked Ubiquitination and Lewy Body-like Inclusions. J. Cell. Physiol. 2020, 235, 7309–7320.
  78. Zhao, H.T.; John, N.; Delic, V.; Ikeda-Lee, K.; Kim, A.; Weihofen, A.; Swayze, E.E.; Kordasiewicz, H.B.; West, A.B.; Volpicelli-Daley, L.A. LRRK2 Antisense Oligonucleotides Ameliorate α-Synuclein Inclusion Formation in a Parkinson’s Disease Mouse Model. Mol. Ther. Nucleic Acids 2017, 8, 508–519.
  79. Korecka, J.A.; Talbot, S.; Osborn, T.M.; de Leeuw, S.M.; Levy, S.A.; Ferrari, E.J.; Moskites, A.; Atkinson, E.; Jodelka, F.M.; Hinrich, A.J.; et al. Neurite Collapse and Altered ER Ca2+ Control in Human Parkinson Disease Patient IPSC-Derived Neurons with LRRK2 G2019S Mutation. Stem Cell Rep. 2019, 12, 29–41.
  80. Korecka, J.A.; Thomas, R.; Hinrich, A.J.; Moskites, A.M.; Macbain, Z.K.; Hallett, P.J.; Isacson, O.; Hastings, M.L. Splice-Switching Antisense Oligonucleotides Reduce LRRK2 Kinase Activity in Human LRRK2 Transgenic Mice. Mol. Ther. Nucleic Acids 2020, 21, 623–635.
  81. Biogen. A Phase 1 Single- and Multiple-Ascending-Dose Study to Assess the Safety, Tolerability, and Pharmacokinetics of BIIB094 Administered Intrathecally to Adults With Parkinson’s Disease (REASON). J. Parkinson’s Dis. 2019, 12. Available online: https://www.clinicaltrials.gov/ct2/show/NCT03976349 (accessed on 6 January 2022).
  82. Cook, D.A.; Kannarkat, G.T.; Cintron, A.F.; Butkovich, L.M.; Fraser, K.B.; Chang, J.; Grigoryan, N.; Factor, S.A.; West, A.B.; Boss, J.M.; et al. LRRK2 Levels in Immune Cells Are Increased in Parkinson’s Disease. NPJ Parkinson’s Dis. 2017, 3, 11.
  83. Yan, R.; Liu, Z. LRRK2 Enhances Nod1/2-Mediated Inflammatory Cytokine Production by Promoting Rip2 Phosphorylation. Protein Cell 2017, 8, 55–66.
  84. Nazish, I.; Arber, C.; Piers, T.M.; Warner, T.T.; Hardy, J.A.; Lewis, P.A.; Pocock, J.M.; Bandopadhyay, R. Abrogation of LRRK2 Dependent Rab10 Phosphorylation with TLR4 Activation and Alterations in Evoked Cytokine Release in Immune Cells. Neurochem. Int. 2021, 147, 105070.
  85. Manzoni, C.; Mamais, A.; Roosen, D.A.; Dihanich, S.; Soutar, M.P.M.; Plun-Favreau, H.; Bandopadhyay, R.; Hardy, J.; Tooze, S.A.; Cookson, M.R.; et al. MTOR Independent Regulation of Macroautophagy by Leucine Rich Repeat Kinase 2 via Beclin-1. Sci. Rep. 2016, 6, 35106.
  86. Yoon, J.-H.; Mo, J.-S.; Kim, M.-Y.; Ann, E.-J.; Ahn, J.-S.; Jo, E.-H.; Lee, H.-J.; Lee, Y.C.; Seol, W.; Yarmoluk, S.M.; et al. LRRK2 Functions as a Scaffolding Kinase of ASK1-Mediated Neuronal Cell Death. Biochim. Biophys. Acta Mol. Cell Res. 2017, 1864, 2356–2368.
  87. Ogier, J.M.; Nayagam, B.A.; Lockhart, P.J. ASK1 Inhibition: A Therapeutic Strategy with Multi-System Benefits. J. Mol. Med. 2020, 98, 335–348.
  88. Wu, Q.; Wu, W.; Jacevic, V.; Franca, T.C.C.; Wang, X.; Kuca, K. Selective Inhibitors for JNK Signalling: A Potential Targeted Therapy in Cancer. J. Enzyme Inhib. Med. Chem. 2020, 35, 574–583.
  89. Yang, D.; Thomas, J.M.; Li, T.; Lee, Y.; Liu, Z.; Smith, W.W. The Drosophila Hep Pathway Mediates Lrrk2-Induced Neurodegeneration. Biochem. Cell Biol. 2018, 96, 441–449.
  90. Gasparotto, J.; Ribeiro, C.T.; Bortolin, R.C.; Somensi, N.; Fernandes, H.S.; Teixeira, A.A.; Guasselli, M.O.R.; Agani, C.A.J.O.; Souza, N.C.; Grings, M.; et al. Anti-RAGE Antibody Selectively Blocks Acute Systemic Inflammatory Responses to LPS in Serum, Liver, CSF and Striatum. Brain. Behav. Immun. 2017, 62, 124–136.
  91. Wang, S.; Zhang, X.; Guo, Y.; Rong, H.; Liu, T. The Long Noncoding RNA HOTAIR Promotes Parkinson’s Disease by Upregulating LRRK2 Expression. Oncotarget 2017, 8, 24449–24456.
  92. Chen, Q.; Huang, X.; Li, R. LncRNA MALAT1/MiR-205-5p Axis Regulates MPP+-Induced Cell Apoptosis in MN9D Cells by Directly Targeting LRRK2. Am. J. Transl. Res. 2018, 10, 563–572.
  93. Özeş, A.R.; Wang, Y.; Zong, X.; Fang, F.; Pilrose, J.; Nephew, K.P. Therapeutic Targeting Using Tumor Specific Peptides Inhibits Long Non-Coding RNA HOTAIR Activity in Ovarian and Breast Cancer. Sci. Rep. 2017, 7, 894.
  94. Amodio, N.; Raimondi, L.; Juli, G.; Stamato, M.A.; Caracciolo, D.; Tagliaferri, P.; Tassone, P. MALAT1: A Druggable Long Non-Coding RNA for Targeted Anti-Cancer Approaches. J. Hematol. Oncol. 2018, 11, 63.
  95. Kim, H.; Park, H.J.; Choi, H.; Chang, Y.; Park, H.; Shin, J.; Kim, J.; Lengner, C.J.; Lee, Y.K.; Kim, J. Modeling G2019S-LRRK2 Sporadic Parkinson’s Disease in 3D Midbrain Organoids. Stem Cell Rep. 2019, 12, 518–531.
  96. Hsieh, C.-H.; Li, L.; Vanhauwaert, R.; Nguyen, K.T.; Davis, M.D.; Bu, G.; Wszolek, Z.K.; Wang, X. Miro1 Marks Parkinson’s Disease Subset and Miro1 Reducer Rescues Neuron Loss in Parkinson’s Models. Cell Metab. 2019, 30, 1131–1140.
  97. Radford, H.; Moreno, J.A.; Verity, N.; Halliday, M.; Mallucci, G.R. PERK Inhibition Prevents Tau-Mediated Neurodegeneration in a Mouse Model of Frontotemporal Dementia. Acta Neuropathol. (Berl.) 2015, 130, 633–642.
  98. Mengeste, A.M.; Lund, J.; Katare, P.; Ghobadi, R.; Bakke, H.G.; Lunde, P.K.; Eide, L.; Mahony, G.O.; Göpel, S.; Peng, X.-R.; et al. The Small Molecule SERCA Activator CDN1163 Increases Energy Metabolism in Human Skeletal Muscle Cells. Curr. Res. Pharmacol. Drug Discov. 2021, 2, 100060.
  99. Nogami, K.; Maruyama, Y.; Sakai-Takemura, F.; Motohashi, N.; Elhussieny, A.; Imamura, M.; Miyashita, S.; Ogawa, M.; Noguchi, S.; Tamura, Y.; et al. Pharmacological Activation of SERCA Ameliorates Dystrophic Phenotypes in Dystrophin-Deficient Mdx Mice. Hum. Mol. Genet. 2021, 30, 1006–1019.
  100. Grohm, J.; Kim, S.-W.; Mamrak, U.; Tobaben, S.; Cassidy-Stone, A.; Nunnari, J.; Plesnila, N.; Culmsee, C. Inhibition of Drp1 Provides Neuroprotection in Vitro and in Vivo. Cell Death Differ. 2012, 19, 1446–1458.
  101. Cole, T.A.; Zhao, H.; Collier, T.J.; Sandoval, I.; Sortwell, C.E.; Steece-Collier, K.; Daley, B.F.; Booms, A.; Lipton, J.; Welch, M.; et al. α-Synuclein Antisense Oligonucleotides as a Disease-Modifying Therapy for Parkinson’s Disease. JCI Insight 2021, 6, e135633.
  102. Henderson, M.X.; Sengupta, M.; Trojanowski, J.Q.; Lee, V.M.Y. Alzheimer’s Disease Tau Is a Prominent Pathology in LRRK2 Parkinson’s Disease. Acta Neuropathol. Commun. 2019, 7, 183.
  103. Yan, K.; Zhang, W.; Han, X.; Chang, F.; Liu, Y. Inhibitory Role of Peroxiredoxin 2 in LRRK2 Kinase Activity Induced Cellular Pathogenesis. J. Biomed. Res. 2019, 34, 103–113.
  104. Ding, X.; Barodia, S.K.; Ma, L.; Goldberg, M.S. Fbxl18 Targets LRRK2 for Proteasomal Degradation and Attenuates Cell Toxicity. Neurobiol. Dis. 2017, 98, 122–136.
  105. Liu, J.; Li, T.; Thomas, J.M.; Pei, Z.; Jiang, H.; Engelender, S.; Ross, C.A.; Smith, W.W. Synphilin-1 Attenuates Mutant LRRK2-Induced Neurodegeneration in Parkinson’s Disease Models. Hum. Mol. Genet. 2016, 25, 672–680.
  106. Yim, Y.-I.; Sun, T.; Wu, L.-G.; Raimondi, A.; De Camilli, P.; Eisenberg, E.; Greene, L.E. Endocytosis and Clathrin-Uncoating Defects at Synapses of Auxilin Knockout Mice. Proc. Natl. Acad. Sci. USA 2010, 107, 4412–4417.
More
Information
Subjects: Neurosciences
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , , , , , ,
View Times: 426
Revisions: 2 times (View History)
Update Date: 24 Oct 2022
1000/1000
Video Production Service