Next Article in Journal
Non-Coding RNAs as Potential Targets for Diagnosis and Treatment of Oral Lichen Planus: A Narrative Review
Previous Article in Journal
Placentas from Women with Late-Onset Preeclampsia Exhibit Increased Expression of the NLRP3 Inflammasome Machinery
Previous Article in Special Issue
Overview of the Impact of Pathogenic LRRK2 Mutations in Parkinson’s Disease
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

An Update on the Interplay between LRRK2, Rab GTPases and Parkinson’s Disease

Department of Neuropathology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan
*
Author to whom correspondence should be addressed.
Biomolecules 2023, 13(11), 1645; https://doi.org/10.3390/biom13111645
Submission received: 19 October 2023 / Revised: 10 November 2023 / Accepted: 11 November 2023 / Published: 13 November 2023
(This article belongs to the Special Issue Pathological Roles of LRRK2)

Abstract

:
Over the last decades, research on the pathobiology of neurodegenerative diseases has greatly evolved, revealing potential targets and mechanisms linked to their pathogenesis. Parkinson’s disease (PD) is no exception, and recent studies point to the involvement of endolysosomal defects in PD. The endolysosomal system, which tightly controls a flow of endocytosed vesicles targeted either for degradation or recycling, is regulated by a number of Rab GTPases. Their associations with leucine-rich repeat kinase 2 (LRRK2), a major causative and risk protein of PD, has also been one of the hot topics in the field. Understanding their interactions and functions is critical for unraveling their contribution to PD pathogenesis. In this review, we summarize recent studies on LRRK2 and Rab GTPases and attempt to provide more insight into the interaction of LRRK2 with each Rab and its relationship to PD.

1. Introduction

Since its discovery as the protein responsible for Parkinson’s disease (PD) in the PARK8 locus in 2004 [1,2], leucine-rich repeat kinase 2 (LRRK2) has been one of the main focus molecules associated with this neurodegenerative disease. The physiological roles of LRRK2 have been linked to a myriad of cellular processes, such as several types of autophagy including macroautophagy and mitophagy [3,4], endocytosis and intracellular transport involving the trans-Golgi network (TGN) and other organelles [5,6,7,8], the regulation of microtubules [5,6], interaction with bacterial pathogens [7], regulation of lysosomal homeostasis [8], and much more.
The pathogenic features of LRRK2 have also been studied by analyzing mutations associated with PD, and most, if not all, mutations point towards a similar effect: augmentation of substrate phosphorylation [8,9]. Albeit these findings, the exact mechanism of how defects in LRRK2 lead to PD has been elusive for nearly a score of years now. An auspicious approach to this enigma would be its link to the endolysosomal system and their regulators, Rab GTPases, as a considerable number of findings indicate connections between these [10].
Rab GTPases bind to membranes and utilize their affinity change via their guanine nucleotide binding status to form specific functional domains on their corresponding organelle membranes [11], hence called the master regulators of intracellular vesicular traffic. Some of the first reports that related Rab GTPases to PD were around 20 years ago, when α-synuclein was reported to interact with several Rab GTPases [12] or α-synuclein-induced neuronal loss was rescued by overexpression of Rab1 [13]. Research was boosted when LRRK2 was found to be associated with another candidate PD risk gene, Rab29 (also known as Rab7L1) [10], and by subsequent findings of Rab phosphorylation by LRRK2 in cells [14,15,16,17,18,19]. Recent advances in structural analysis have also revealed several interesting findings and insights about LRRK2 and its functions, with some of them incorporating Rab GTPases as its interactor. In this review, we aim to provide an update of the current findings on LRRK2, both from structural and functional aspects, as well as known interacting Rab GTPases and their functions, hoping to give a better view on LRRK2, Rab GTPases, and PD.

2. Insights from Genetic and Structural Studies of LRRK2

Back in 2002, Funayama et al. reported a family of inherited PD with an unknown causative gene locus, PARK8 [20]. Two years later, two independent groups identified the responsible gene as LRRK2 [1,2]. Since then, genetics have revealed at least seven causative mutations in this gene product: N1437H [21], R1441C [1,22], R1441G [22], R1441H [22], Y1699C [1], G2019S [23], I2020T [1], and numerous other rare variants with unclear causality [24]. The gene product LRRK2 is a 2527-amino-acid multidomain kinase, and the PD-associated mutations lie in the ROC (Ras of complex), COR (C-terminal of ROC), and kinase domains (Figure 1). The ROC domain is a GTP-binding domain that regulates the kinase activity in an intramolecular fashion [25,26], although its GTP-binding state (not GTP-binding capacity) may not be required for its kinase activity [27]. The ROC domain also works as a scaffold for protein interactions [28], whereas the COR domain constitutes an interface for LRRK2 dimerization [29,30]. All the aforementioned LRRK2 mutations result in an increase in phosphorylated substrates [18,31,32], which is not necessarily accompanied by an increased kinase activity of LRRK2 itself [29,30].
Other domains in LRRK2 have no PD-associated mutants allocated to them but are also of importance when considering intermolecular interactions. Armadillo repeats (ARM), ankyrin repeats (ANK), and leucine-rich repeats (LRR) are relatively abundant motifs that form various sizes of scaffolds for protein interaction [33,34,35], whereas WD40 domains are beta-propellers that may interact with DNA as well as proteins [36,37]. The detailed primary structure is depicted in Figure 1A. It might also be worth noting that a portion of endogenous LRRK2 in macrophages is found cleaved at the ANK-LRR interdomain region to produce a C-terminal fragment including the kinase region [38]. Although this fragment may be nonfunctional because the N-terminal membrane-interacting region is lacking, it may also act as dominant negative as it can heterodimerize with full-length LRRK2 [38].
Figure 1. Structures of LRRK2 and PD-associated mutations. (A) Primary structure of LRRK2 and the location of reported PD-associated mutations. ROC: Ras of complex domain, COR: C terminal of ROC domain, WD40: WD40 domain. Domain lengths and borders are based on [39]. (B) Monomeric structure of full-length LRRK2 [30] (PDB ID: 7HLT). (C) Homodimeric structure of full-length LRRK2 [30] (PDB ID: 7HLW). (D) Protomer of filamentous LRRK2 C-terminal half (RCKW) on microtubules [40] (PDB ID: 6VNO). All structures are aligned so that the WD40 domain lies at the top left. Colorization of structures is the same for (AD).
Figure 1. Structures of LRRK2 and PD-associated mutations. (A) Primary structure of LRRK2 and the location of reported PD-associated mutations. ROC: Ras of complex domain, COR: C terminal of ROC domain, WD40: WD40 domain. Domain lengths and borders are based on [39]. (B) Monomeric structure of full-length LRRK2 [30] (PDB ID: 7HLT). (C) Homodimeric structure of full-length LRRK2 [30] (PDB ID: 7HLW). (D) Protomer of filamentous LRRK2 C-terminal half (RCKW) on microtubules [40] (PDB ID: 6VNO). All structures are aligned so that the WD40 domain lies at the top left. Colorization of structures is the same for (AD).
Biomolecules 13 01645 g001
LRRK2 is observed in many conformations in vitro or in cells, with structural models from monomer [30] to homodimer [30,41,42] to filamentous [40,43] and even heteromultimer [44] reported (Table 1, Figure 1B–D). The classical model of LRRK2 is the homodimer model, based on biochemical analysis and crystal structures of ROC domains [45,46]. This model was further confirmed via other methods such as cross-linking and negative stain [47], and further with full-length LRRK2 [30] (Figure 1B).
The monomeric model was first proposed based on biochemical analysis of full-length LRRK2 [50] but was not confirmed until some years ago when the structure of full-length LRRK2 was finally determined via cryoelectron microscopy (cryo-EM) [30] (Figure 1C).
The filamentous LRRK2 model is based on observations of overexpressed PD-associated mutants, which are mostly cytosolic but often form filaments along microtubules [51]. Reports that analyzed this form of LRRK2 bound to microtubules via cryo-EM or cryoelectron tomography (cryo-ET) showed multimer formation via interactions through the WD40 domain with the help of microtubule filaments [40,43] (a protomer of the filament is shown in Figure 1D).
The analyzed structures of monomer or dimer forms of LRRK2 via Cryo-EM were reported to be captured in a kinase-inactive state [30,40], whereas another study utilizing microscale thermophoresis-based biophysical methods reported one of the active forms that bind either Rab8 or Rab10 [52]. Another cryo-EM analysis also observed a LRRK2-Rab29 complex and a tetrameric assembly of this complex, yielding active LRRK2 [44]. Of note, more recent studies have identified Rab12 rather than Rab29 as an activator of LRRK2, as described later, so a heteromultimer complex with Rab12 would also need to be assumed [53,54]. From these reports, one can infer that there are multiple modes of activation, and the activity should be broken down into several aspects. Indeed, we reported that the increased phosphorylation of Rab GTPases by LRRK2 under lysosomal stress can be separated into at least two phases: intrinsic LRRK2 activation and other means of accelerated interaction between LRRK2 and substrate Rab [55].
Interestingly, it has been shown that when LRRK2 is in the monomeric inactive state, interactions between the WD40 domains of LRRK2 and other molecules are blocked by parts of the LRR and ARM domains [30]. Other studies focused on how proteins could interact with LRRK2 via the WD40 domain of LRRK2, such as Syntaxin1 and NSF in synaptic vesicles [56], and these could also be tightly regulated by conformational changes by either protein modifications or proteins binding to LRRK2 in other domains.
Either way, the regulation of LRRK2 activation relies partly on Rab GTPases, and revealing their cooperative mode of action is critical for unraveling the physiological and pathological roles of LRRK2.

3. Rab GTPases and LRRK2

3.1. Rab GTPases and the Endolysosomal System

Rab GTPases are proteins that form a subfamily of the Ras superfamily and are capable of binding cellular membranes via the C-terminal prenylation. Like other small GTPases, they switch their activity, which influences their affinity to specific proteins called effectors by changing their binding state with either GTP (active) or GDP (inactive) with the help of their specific guanine nucleotide exchange factors (GEF) or GTPase-activating protein (GAP). Their functions are associated with a wide variety of intracellular trafficking, ranging from cellular secretory pathways to intracellular degradation pathways involving the endolysosomal system.
The endolysosomal system is part of an intracellular flow of enveloped membrane organelles and, apart from its function in autophagy, serves as a sorting site for substances incorporated by endocytosis. These substances include extracellular materials as well as membrane proteins and cellular membranes themselves. These substances are then either guided towards degradation by lysosomes, returned (or “recycled”) to the plasma membrane, or routed to the TGN. This pathway is known to regulate basic steps of cellular processes such as signaling, adhesion, immunity, nutrient uptake, organelle homeostasis, membrane protein turnover, and much more (reviewed in [57,58,59,60,61,62]).
This route to degradation can be broken up into several parts, with more than one Rab GTPase regulating the routing or maturation of each vesicle. Endocytosed materials are first retained in the early endosome, where Rab5 controls the maturation of the vesicle and other Rab GTPases, such as Rab11, regulate re-routing from the early endosome to other compartments, in this case to the recycling endosomes, another part of the endolysosomal system, and ultimately to the plasma membrane [63,64]. Membranous proteins targeted for degradation start to form vesicles called intraluminal vesicles (ILVs) inside of this early endosome with the help of some specialized protein complexes [57,61]. The formation of ILVs continues throughout the maturation process. Matured early endosomes transform into slightly more acidic vesicles with different lipid compositions, which are called late endosomes. At the late endosomes, Rab7 is the controller of further maturation, and again other Rab GTPases, such as Rab9, account for the re-routing to other compartments [63,65]. Eventually, the late endosomes, with all the reusable proteins routed away to the plasma membrane or trans-Golgi network, gain the ability to fuse with lysosomes with the help of several Rab7 effector proteins, marking the last part of the endolysosomal system [60,63]. In some cells, Rab GTPases also regulate cell-type specific vesicles such as synaptic vesicles in neurons and melanosomes in melanocytes [66,67].

3.2. LRRK2 and Substrate Rab GTPases

As a kinase, LRRK2 phosphorylates a subset of Rab GTPases, e.g., Rab8 and Rab10, in cells. In this section, we review the functions and pathobiology of each Rab GTPase, citing some new reports that may clarify their relationship to LRRK2 and their contribution to PD. The overview of the Rabs with their roles upon phosphorylation is summarized in Figure 2 and Table 2.

3.2.1. Rab8 and Rab10

Rab8 and Rab10 are closely related Rabs, both categorized in the Rab8 subfamily [89] and are the most characterized Rab GTPase in the context of interaction with LRRK2.
Right after the initial report of multiple Rab phosphorylation by LRRK2 [14], Rab10 was found to be a very sensitive marker for assessing LRRK2 activity [90], followed by a quick development of a phospho-specific antibody against Rab10 [68]. To date, numerous studies have incorporated an assessment of this phosphorylation in their studies on LRRK2 kinase activity [24,55,76,77,78,81,82,85,91,92,93,94,95,96,97,98,99]. Moreover, several reports have attempted to utilize phosphorylated Rab10 as a biomarker for upregulated LRRK2 activities in PD or preclinical models, which was shown to be successful to a certain extent, although results vary between studies [100,101,102,103,104,105,106,107,108,109]. Although it is almost established that the phosphorylation state of Rab10 reflects the activity of LRRK2, there are still some function-related ambiguities of this GTPase that have to be straightened out.
Rab10 has been implicated in several modes of transport in a variety of cell types, from general exocytic pathways to neurite or cilia formation and immune responses [110]. LRRK2 has been shown to play a role in these functions via its kinase activity, altering the ability of Rab10 to interact with other proteins. In the context of ciliogenesis, phosphorylated Rab10 binds to RILPL1 at or near centrosomes, inhibiting ciliogenesis [15,71,78,79,80]. This inhibition was also brought about by the retention of Myosin Va at centrioles by the same phosphorylation of Rab10 [77]. In the context of endosomal trafficking, phosphorylation of Rab10 at steady state on early macropinosomal membranes results in a decrease in binding to EHBP1L1, a Rab10 effector. This then inhibits the recycling of macropinosomes, ultimately hindering chemotaxis in macrophage cells [81]. Another effect on endosomal trafficking is in lysosomal stress-induced cells, where Rab10 as well as its effectors EHBP1 and EHBP1L1 regulate lysosomal content release [76]. Other functions include lysosomal tubular budding named LYTL (LYsosomal Tubulation/sorting driven by LRRK2) upon Rab10 phosphorylation by LRRK2 and recruitment of JIP4 [82,83], negative regulation of a lysosomal enzyme CGase upon Rab10 phosphorylation by LRRK2 [84], and induction of lysosomal overload stress and apoptosis after neuronal injury by the same phosphorylation [85].
Rab8, on the other hand, is well characterized as a Rab GTPase, with GEFs Rabin8 and GRAB, GAPs TBC1D30, and other TBC family proteins, and multiple effectors described in the context of anterograde trafficking, endocytic recycling and exocytosis, association with the cytoskeleton, cell shape regulation and migration, ciliogenesis, neurite growth, and much more [111]. Although there have been attempts to utilize the phosphorylation of this GTPase as a biomarker for PD [112], further studies are still required for the assessment of Rab8 phosphorylation, as the currently available antibody against Rab8 phosphorylated at Thr72 (MJF-R20) has been shown to cross-react with phosphorylated Rab3A, Rab10, Rab35, and Rab43 [68].
The first reports on Rab8 functions date back about 30 years when Rab8 was deemed responsible for post-Golgi anterograde trafficking in epithelial and neuronal cells [113,114]. Studies on Rab8’s involvement in neurite formation immediately followed [115] and built the classical view of Rab8 as a controller of neurite formation via anterograde trafficking. Current knowledge on Rab8 in neurite formation includes additional upstream elements, involving various other Rabs and their effectors, such as Rab11 and Rabin8, that activate Rab8 and Rab10 for neurite outgrowth [116], as well as downstream elements such as Cdc42 and tuba that strictly regulate the number of axons formed per cell [117].
Another aspect of Rab8 is its involvement in cilia. Formation of cilia requires Rab8 activation on the centrosome by Rabin8, very much like in neurite formation, and further protein trafficking to the base of primary cilia [111]. Impaired receptor trafficking to cilia via Rab8 dysfunction causes various deficits in functions that require cilia, such as adipocyte differentiation, where Rab8 is responsible for the trafficking of frz2 to the base of primary cilia [118]. Rab10 and Rab13 might have compensatory roles in ciliogenesis as double knockouts of Rab8a and Rab8b were insufficient to cause cilial deficits [119].
These functions of Rab8 rely on proper trafficking from the TGN or recycling endosomes to each responsible compartment, most likely being the central function of Rab8 shared among various cell types.
Some reports have also attempted to delineate the relationship between Rab8 and PD by hypothesizing a direct interaction between Rab8 and α-synuclein, a protein well known to both cause PD and accumulate in PD brains [12,120]. They seem to bind to each other in vitro [12] at the switch region of Rab8 [120], and their binding enhances α-synuclein fibrilization while ameliorating its toxicity [120]. Although the detailed mechanisms are yet to be unraveled, a model that suggests that Rab8, together with optineurin, a Rab8 effector, is involved in the initiation of autophagy near aggregated proteins [121] might also give us insight. Altogether, it is possible that Rab8 is related to the pathogenesis of PD in some way.
Although most of the above-mentioned functions of Rab8 and Rab10 indicate a very close and very much overlapping role for the two Rabs, there are reports that suggest an antagonistic property for them. One report depicts a discrepancy in the role behind ciliogenesis, where overexpression of GFP-tagged Rab8 exhibited an increase in cilia while overexpression of GFP-tagged Rab10 had the opposite effect [79]. Another recent report has also examined the differential functions of Rab8 and Rab10. Although knockouts of either Rab8 or Rab10 show defects in lysosomal or Golgi homeostasis, such as a decrease in lysosomal number or dispersion of the Golgi, a further look into the details revealed some divergent phenotypes: Rab8 knockouts had acidic lysosomes whereas Rab10 knockouts had elevated pH [122]. These reports collectively suggest that Rab8 and Rab10 control Golgi- and endosome-related trafficking from slightly different aspects, some resulting in seemingly opposite phenotypes when suppressed or overexpressed.
Things get more complicated when their kinase LRRK2 cuts in. Some reports suggest that the phosphorylation of Rab8 inhibits its function and causes centrosomal or ciliogenesis deficits [17,71,72] or decreased binding with its effector [73], while other reports indicate new functions such as the recruitment of RILPL2 [74] or alteration of the endosomal pathway [75]. Either way, phosphorylation of Rab8 by LRRK2 seems to alter its binding ability to effectors, just like in Rab10, leading to the observed changes in its function.
We have previously reported a difference in the functions of Rab8 and Rab10 downstream of LRRK2 [76]. Lysosomal enlargement is an outcome of mild lysosomal stress that is not enough to result in lysosomal rupture, and the resultant cellular responses include the deflation of enlarged lysosomes and the promotion of the extracellular release of lysosomal contents. Depletion of Rab8 enhanced lysosomal enlargement, whereas that of Rab10 diminished the exocytosis of lysosomal contents [76]. These lysosomal changes were further controlled by their effectors EHBP1 and EHBP1L1, effectors of Rab10 and Rab8a, as depletion of these effector proteins resulted in both enhanced lysosomal enlargement and lowered lysosomal exocytosis [76]. This may suggest that Rab functions could be defined not only by a single effector but also by the combination of multiple proteins. Indeed, EHBP1 has been reported to bind to both Rab10 and EHD2 to form a ternary complex [123] or to bind active Rab8 and be activated as a scaffold to bind additional proteins such as f-actin [124]. These reports indicate a more complex mode of action: a Rab-and-effector-based recognition of, or an affinity change to, a third factor. Investigating changes in the binding ability of EHBP1/EHBP1L1 upon their binding to Rab8/10 could be worthwhile to further delineate the intertwined functions of the two Rabs.

3.2.2. Rab29

Also known as Rab7L1, Rab29 itself is nominated as a risk factor for PD, encoded in the PARK16 locus [125,126,127]. This GTPase was among the first of all Rabs to have their relationship with LRRK2 uncovered [128,129] and is the Rab that is known to regulate LRRK2 from upstream [16,72,76,87,92,130] in addition to the recently reported Rab12 [53,54] and Rab38 [131]. Rab29 localization at steady state was reported to be at the Golgi, with a small fraction at perinuclear vesicles [132,133]. These Rab29 population at the Golgi was reported to be responsible for the integrity of the TGN and the retrograde trafficking there [18,134]. Golgi fragmentation was also dependent on the recruitment and activation of LRRK2 induced by Rab29 overexpression [16,19], as well as phosphorylation of Rab29 by LRRK2 [18].
The function of Rab29 has attracted attention not only from its relationship to the Golgi but also to the lysosome, as the small population of Rab29 at perinuclear vesicles was found to be lysosomal. Also, Rab29 has been shown to react to lysosomal stress, localizing itself to lysosomes and also co-recruiting and activating LRRK2 [76,87]. Active Rab29 on lysosomes regulates the size of abnormally inflated lysosomes, which is dependent on LRRK2 kinase activity [76,87]. The localization of Rab29 itself to lysosomes also depends on the activity of LRRK2 as well as some PKC isoforms and their ability to phosphorylate Rab29 at corresponding sites [87].
The physiological importance of the function of Rab29 on lysosomes and its relationship with LRRK2, at least in some respects, is confirmed by loss-of-function studies. Knockdown of endogenous Rab29 in macrophage cells causes excessive lysosomal enlargement and inhibits LRRK2 recruitment [76] and activation [55] upon lysosomal stress loading. Consistent with this, Rab29 knockout mice have been shown to exhibit the accumulation of enlarged lysosomes in renal proximal cells, which is strikingly similar to the renal phenotype reported in LRRK2 knockout animals [130]. On the other hand, several other studies have reported few or no phenotypes related to the regulation of LRRK2 activation in Rab29 knockout cells, including MEFs, lung cells [133], and HEK293FT cells [99]. Further studies would be needed to determine whether these differences in results are due to differences in cell types (macrophages vs. other cell types), different experimental conditions (e.g., transient knockdown vs. stable knockout), or other reasons. Considering the observations that LRRK2 is activated once recruited to membranes [92] and that LRRK2 harbors a site that strongly binds phosphorylated Rab substrates other than the site that binds Rab29 [52], Rab29 might be acting as an initiator of LRRK2 activation and not necessarily keeping LRRK2 active.
In any case, Rab29 behaves poorly at steady state [135] but is very reactive to lysosomal stress [76,87], which suggests a role in lysosomal troubleshooting. Although the exact stimulus or conditions that Rab29 reacts to, or their sensor proteins, still need closer investigation, Rab29 is surely a promising key molecule in deciphering lysosomal stress responses, if not PD pathogenesis.

3.2.3. Rab12

Rab12 was first found to regulate a “non-canonical” degradation route from recycling endosomes directly to lysosomes [136], then further allocated to more transfer between the cell surface and Golgi for various cargoes [137,138]. Rab12 is also gradually being understood as a potent marker of LRRK2 activity, as the phosphorylation of Rab12 was reported to be potently induced by PD-associated mutants of LRRK2 [133,139]. The functions of the phosphorylation of this GTPase were not known until very recently; it was found to be responsible for controlling the intracellular localization of lysosomes via an increase in the binding ability to RILPL1 [86]. An unusual point about this phosphorylation is that LRRK2 recognizes GDP-bound Rab12 better than the GTP-bound form, at least in vitro [140]. Rab12 is also implicated in lysosomal repair, as Rab12 accumulates on damaged lysosomes and activates LRRK2 there [53,54]. Rab12-mediated accumulation and activation of LRRK2 on lysosomes during lysosomal damage were enhanced in PD-associated mutants of LRRK2 or even VPS35, even under non-damaged conditions, but were not enhanced beyond wild-type during damage [54], suggestive of a Rab12-dependent lysosomal response mechanism that might be constantly activated in the course of PD pathogenesis with these mutations. This accumulation and activation were brought about by Rab12 binding to LRRK2, and although the binding site on LRRK2 was located in its N-terminal Armadillo domain that includes the binding site of Rab29 or phosphorylated Rab8 or Rab10, the detailed site was different from these molecules [53]. This is indicative of yet another pathway in sensing lysosomal abnormalities apart from Rab29, and as they both augment LRRK2-induced phosphorylations, Rab12 would be a promising GTPase to dig into in terms of the pathogenesis of PD.

3.2.4. Rab35

Rab35 is a Rab GTPase responsible for various cellular processes including exosome release, neurite outgrowth, phagocytosis, cell polarization, immune synapse formation, cytokinesis, and cell migration [141]. These pathways are controlled by either the quick recycling of endocytic cargoes (e.g., T cell receptor (TCR) and MHC complexes for immune synapse formation, podocalyxin for cell polarization) to the plasma membrane or the regulation of actin beneath the plasma membrane to promote changes in cell shape and position [141]. Although the main link to diseases would be between cancer, there are several reports that link this GTPase and LRRK2 to PD.
The effectors of Rab35 include OCRL, MICAL1, and MICAL-L1 [132], which are also effectors of Rab8 or Rab10 as noted above. Not surprisingly, some of the functions of Rab35 overlap with Rab8 and Rab10, which include the recruitment of JIP4 and induction of LYTL upon phosphorylation by LRRK2 [83]. LRRK2-induced phosphorylation of Rab35 was also found to positively regulate the propagation of α-synuclein [88]. This may be caused by either tubulation of lysosomes (LYTL) or the fast recycling of endocytic content, but details need further assessment. Nevertheless, Rab35 may be an important Rab in the development of PD, and possibly, in more initial pathogenic mechanisms.

3.2.5. Rab5

Rab5 is the key GTPase in controlling the maturation of early endosomes. After endocytosis, Rab5 is recruited to the endocytic vesicle by Rab4 or the Rab5 GEF Rabex5 (RABGEF1), and then in turn recruits effector proteins such as EEA1, a tether for fusion with other early endosomes, VPS34, a phosphatidylinositol kinase responsible for converting phosphatidylinositol (PI) to the endosome-enriched lipid phosphatidylinositol-3-phosphate (PI3P), and the Rab7-GEF Mon1-Ccz1, which recruits Rab7 and facilitates transition to late endosomes [60].
Although there are many studies on Rab5, the differences between the three isoforms Rab5a, Rab5b, and Rab5c are not that undeciphered. Rab5c is reported to have a slightly different function from the other two, with little involvement in EGFR recycling [142] or specific involvement in Rac1-dependent cell migration [143]. Some more findings exist in non-mammalian cells, as Rab5a and Rab5b in Leishmania interact with different modes of endocytosis [135], or in yeast, ypt53 (one of the three Rab5 isoforms in yeast) is selectively upregulated under stressed conditions [144]. Further studies are expected.
Rab5 and PD have little connection reported, with implications in Rab5a-mediated uptake of α-synuclein in neurons [145] or clearance in microglia [146]. The latter is the phenotype also seen in LRRK2 knockout mice, which could hint at the possibility of Rab5 interplay in PD pathogenesis or treatment. Other links reported between LRRK2 and Rab5 include a cooperative regulation of neurite outgrowth [147], phosphorylation of all the isoforms of Rab5 by LRRK2 [15], and inactivation of Rab5b [70]. Note that phosphorylation of Rab5 isoforms by LRRK2 has only been observed upon overexpression in cells [15,68] or in vitro [70], and therefore we need to be cautious as to whether they are indeed physiological substrates. Nonetheless, these data could collectively outline a pathway from overactivated pathogenic LRRK2 to irregular inactivation of Rab5 and endosomal deficits, and although further confirmation is needed, Rab5 may be one of the key players behind PD development and progression.

3.2.6. Rab3

Rab3 has four isoforms (Rab3a, Rab3b, Rab3c, Rab3d), and all of the isoforms participate in exocytosis or secretion. They are highly expressed in neurons and secretory cells [66,148]. Their roles in secretion in secretory cells or neurons appear to be redundant, with several knockout studies in mice observing little or no changes in exocytotic activity, but depletion of all Rab3 isoforms results in lethality from respiratory failure [149,150,151,152]. In neurons, Rab3 regulates a specific type of exocytotic vesicles called dense-core vesicles, which are important in neuropeptide release [153]. The four isoforms display different magnitudes of activity, with Rab3a being the most active [153]. Rab3a is also found to be responsible for plasma membrane repair via lysosomal exocytosis [154].
In cancer, these Rabs are associated with increased proliferation and invasion, possibly due to abnormal exosome release [155]. Here, Rab3d is the most popular isoform known to be upregulated in several types of tumors. Although research on Rab3 in non-secretory cells is limited, these perspectives might serve as hints to the functions of Rab3 in non-secretory cells.
There are several reports that associate Rab3a with α-synuclein [156,157,158]. Regulation of synaptic vesicle endocytosis is suggested to be a physiological function of α-synuclein, and disruption of this could very well be a cause of α-synuclein-related neurodegeneration [157].
Although Rab3 was identified as a LRRK2 substrate, there is only a limited number of studies on the interaction between these two proteins. One shows that Rab3a colocalizes with LRRK2 on stressed lysosomes dependent on its kinase activity [76], and another shows that Rab3 is a very weak substrate in LRRK2-G2019S expressing neurons, probably because of different localizations in neurons [159]. There is still quite a large gap between LRRK2, Rab3, and PD, but given the fact that Rab3 is implicated in lysosomal repair and interactions with α-synuclein, Rab3 could possibly be a potential factor in PD pathogenesis.

3.2.7. Rab1

Rab1 is the newest Rab GTPase found to be a substrate of LRRK2 [69]. The classical role of Rab1 is its involvement in ER-Golgi transport and maintenance of the Golgi, but its functions reach out to regulating the localization of endosomes and lysosomes, and consequential cell-surface receptor recycling [160]. Loss of Rab1 results in fragmentation of the Golgi, which is seen in α-synuclein overexpression models [13] or in dopaminergic neurons in the substantia nigra of PD patients [161]. Another aspect of Rab1 is its involvement in autophagy, a cellular process in which cytosolic contents or specific organelles are sequestered in double-membrane autophagosomes and routed toward lysosomal degradation. Rab1 is necessary for the earliest steps in autophagy, possibly through the correct localization of autophagy-initiating proteins such as ATG9 [162,163,164]. Complete knockout of this Rab seems to be lethal [165], possibly due to its wide variety of functions it is responsible for.
It is yet to be determined how the LRRK2-induced phosphorylation affects Rab1 and its functions, but its involvement in PD pathogenesis would certainly be worthwhile to discover.

4. Rab Phosphorylation in Relation to PD

As stated above, almost all familial PD mutations increase Rab phosphorylation in cells, although the most common G2019S mutation may have somewhat different effects than others. That is, the G2019S mutation increases its intrinsic kinase activity more effectively than Rab phosphorylation, whereas the other mutations increase Rab phosphorylation more potently than its kinase activity [24]. This is consistent with the observations in human samples; the increase in Rab10 phosphorylation has been shown in peripheral blood neutrophils of R1441G mutation carriers [106], whereas G2019S mutation appears to have a weaker effect on Rab10 phosphorylation induction, at least in neutrophils [106] and peripheral blood mononuclear cells (PBMCs) [105,108]. These observations implicate slightly different pathomechanisms of PD for G2019S and other familial mutations.
In relevance to the pathomechanism of PD, one should take into account specific cell types in the brain, such as neurons and glia, as LRRK2 and each Rab are known to be expressed relatively ubiquitously in these cells. In analyses using mouse primary neurons and glia, Rab10 phosphorylation is detected in all cell types but is more strongly detected in astrocytes and microglia [105]. In vivo, it has been shown that cholinergic neurons in the striatum of LRRK2 R1441C knock-in mice develop ciliation defects, likely due to Rab10 over-phosphorylation [79], and a similar ciliation phenotype was subsequently observed also in astrocytes of LRRK2 G2019S knock-in mice [166]. Regarding the effects of Rab phosphorylation in microglia, it has been shown that microglial LRRK2 and Rab10 mediate manganese-induced inflammation and neurotoxicity [167]. In addition, since much of the work on LRRK2 and Rab has been conducted using macrophage cells like RAW264.7 cells, these findings would also apply to microglia, which are macrophage-lineage cells.
The actual changes of Rab10 phosphorylation in idiopathic PD (iPD) are also of interest but remain somewhat controversial. It has been shown that Rab10 phosphorylation is elevated in the substantia nigra [168] or PBMCs [108] of iPD patients as compared with healthy subjects. However, other groups have reported that the levels of Rab10 phosphorylation were similar between iPD and controls from the analysis of patients’ cingulate cortex [68] or in PBMCs/neutrophils [100]. Analysis of human urine has shown that urinary Rab10 phosphorylation is just barely higher in iPD than in controls (p = 0.046) [109]. Since the same group reported that the LRRK2 kinase activity itself in urine, as measured by the level of autophosphorylation at Ser1292, increased more clearly in iPD group (p < 0.01) [169], LRRK2 activation state and changes in Rab phosphorylation may not always coincide in PD.
There are some limitations to the above-mentioned analyses, and one should note that the changes of LRRK2 and Rab phosphorylation in PD are not yet at a stage to be fully discussed. First, the well-examined human biofluid samples to date are peripheral blood mononuclear cells and neutrophils, and these peripheral immune cells may exhibit different changes from those in the brain. Second, Rab phosphorylation status may vary greatly among individuals, and further classification of disease based on the type of progression or pathology, as well as other factors, will be necessary. Third, changes in Rab phosphorylation other than Rab10 should also be examined more systematically, but not much has been achieved yet as the generation of phospho-specific antibodies has lagged behind that of phospho-Rab10 antibodies. Fourth, immunoblot analysis after SDS-PAGE is often used for the assessment of Rab phosphorylation, but it would be desirable to develop a simpler and more sensitive assay for Rab phosphorylation detection. Lastly, in studies of PD, it will be required to determine at what time point around the onset of disease Rab phosphorylation is altered, and in which cell types this occurs specifically. Conducting large-scale studies recruiting a large number of PD patient samples, both idiopathic and familial, would address these points and better clarify the contribution of Rab phosphorylation to PD.

5. Conclusions and Future Perspectives

Rab GTPases have been established as the most promising and reliable substrates for LRRK2 since 2016, which has led to significant progress in LRRK2 research. In particular, recent structural and biochemical analyses are revealing more detailed modes of interaction between LRRK2 and Rab, which would advance our understanding of the regulatory mechanisms of Rab phosphorylation.
From a cell biology viewpoint, it would be interesting to further clarify what Rab over-phosphorylation causes in cells. As only a small fraction of Rabs is phosphorylated at steady state, it is likely that the majority of a given pool of Rab proteins can still carry out their normal functions [90]. On the other hand, phosphorylated Rab8a and Rab10 have been shown to bind new effector proteins, i.e., RILPL1/2 and JIP3/4 [15,74,83], suggesting that even a small fraction of them may dominantly affect cellular functions. Indeed, it has been shown that the recruitment of RILPL1 regulates ciliogenesis and centrosomal cohesion [15,71] and that of JIP4 regulates lysosomal tubulation [83] and axonal autophagosome transport [170]. In addition, as noted above, Rab10 phosphorylation by LRRK2 is markedly enhanced under lysosomal stress and is therefore assumed to play important roles in the maintenance of endolysosomes. The effects on disease-causing aggregate-prone proteins are also of interest; the brains of patients with LRRK2 mutations often, but not always, accumulate insoluble α-synuclein and tau to varying degrees, leading to the notion that Rab over-phosphorylation may also affect the metabolism or propagation of these proteins. Indeed, as mentioned above, phosphorylation of Rab35 has been shown to potentially regulate α-synuclein propagation [88]. However, the possibility remains that other Rabs may also be involved in different ways, and it is still unclear how and at what stage Rab over-phosphorylation influences the cellular dynamics of highly insoluble proteins.
With respect to clinical applications, LRRK2 inhibitors including small molecule compounds and antisense oligonucleotides are being developed, and one of them, BIIB122/DNL151, originally developed by Denali Therapeutics Inc, has now proceeded to phase III trials [171]. According to their clinical trial reports, administration of this LRRK2 inhibitor has been shown to markedly reduce Rab10 phosphorylation in PBMCs, as well as phospho-Ser935 LRRK2 in whole blood, total LRRK2 in cerebrospinal fluid (CSF), and a lysosomal lipid di-22:6-bis (monoacylglycerol) phosphate (BMP) in urine, all in a dose-dependent manner [172]. Also, the clinical trial results of another LRRK2 inhibitor, DNL201, were reported earlier, and the results were mostly similar [107]. In addition to LRRK2 inhibitors, some kind of Rab modulators may also be useful if a Rab acting downstream is identified as particularly important. Furthermore, targeting of LRRK2 and Rab phosphorylation in clinical applications may not be limited to PD, since LRRK2 is well-known as a risk gene for several immune/inflammatory diseases, including Crohn’s disease and leprosy. Given that LRRK2 is highly expressed in immune cells, cell type-specific effects of Rab over-phosphorylation would need to be clarified. The full picture of Rab phosphorylation by LRRK2 and its regulatory mechanism will hopefully lead to further elucidation of the pathomechanism of these diseases.

Author Contributions

Conceptualization: T.K. (Tadayuki Komori); Writing—original draft: T.K. (Tadayuki Komori) and T.K. (Tomoki Kuwahara); Writing—review and editing: T.K. (Tadayuki Komori) and T.K. (Tomoki Kuwahara); Visualization: T.K. (Tadayuki Komori); Supervision: T.K. (Tomoki Kuwahara); Funding acquisition: T.K. (Tomoki Kuwahara). All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by JSPS KAKENHI Grant Numbers 22H02949 and 22H04638 (T. Kuwahara).

Acknowledgments

We thank Takeshi Iwatsubo for supervising our activity and our lab members for their helpful suggestions and discussions.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Zimprich, A.; Biskup, S.; Leitner, P.; Lichtner, P.; Farrer, M.; Lincoln, S.; Kachergus, J.; Hulihan, M.; Uitti, R.J.; Calne, D.B.; et al. Mutations in LRRK2 Cause Autosomal-Dominant Parkinsonism with Pleomorphic Pathology. Neuron 2004, 44, 601–607. [Google Scholar] [CrossRef] [PubMed]
  2. Paisán-Ruíz, C.; Jain, S.; Evans, E.W.; Gilks, W.P.; Simón, J.; van der Brug, M.; de Munain, A.L.; Aparicio, S.; Gil, A.M.; Khan, N.; et al. Cloning of the Gene Containing Mutations That Cause PARK8-Linked Parkinson’s Disease. Neuron 2004, 44, 595–600. [Google Scholar] [CrossRef] [PubMed]
  3. Manzoni, C.; Lewis, P.A. Leucine-Rich Repeat Kinase 2 (LRRK2). Adv. Neurobiol. 2017, 14, 89–105. [Google Scholar] [CrossRef] [PubMed]
  4. Singh, F.; Ganley, I.G. Parkinson’s Disease and Mitophagy: An Emerging Role for LRRK2. Biochem. Soc. Trans. 2021, 49, 551–562. [Google Scholar] [CrossRef] [PubMed]
  5. 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. [Google Scholar] [CrossRef]
  6. Boecker, C.A. The Role of LRRK2 in Intracellular Organelle Dynamics. J. Mol. Biol. 2023, 435, 167998. [Google Scholar] [CrossRef]
  7. Herbst, S.; Gutierrez, M.G. LRRK2 in Infection: Friend or Foe? ACS Infect. Dis. 2019, 5, 809–815. [Google Scholar] [CrossRef]
  8. Kuwahara, T.; Iwatsubo, T. The Emerging Functions of LRRK2 and Rab GTPases in the Endolysosomal System. Front. Neurosci. 2020, 14, 227. [Google Scholar] [CrossRef]
  9. Taylor, M.; Alessi, D.R. Advances in Elucidating the Function of Leucine-Rich Repeat Protein Kinase-2 in Normal Cells and Parkinson’s Disease. Curr. Opin. Cell Biol. 2020, 63, 102–113. [Google Scholar] [CrossRef]
  10. Erb, M.L.; Moore, D.J. LRRK2 and the Endolysosomal System in Parkinson’s Disease. J. Park. Dis. 2020, 10, 1271–1291. [Google Scholar] [CrossRef]
  11. Norris, A.; Grant, B.D. Endosomal Microdomains: Formation and Function. Curr. Opin. Cell Biol. 2020, 65, 86–95. [Google Scholar] [CrossRef] [PubMed]
  12. Dalfó, E.; Gómez-Isla, T.; Rosa, J.L.; Bodelón, M.N.; Tejedor, M.C.; Barrachina, M.; Ambrosio, S.; Ferrer, I. Abnormal α-Synuclein Interactions with Rab Proteins in α-Synuclein A30P Transgenic Mice. J. Neuropathol. Exp. Neurol. 2004, 63, 302–313. [Google Scholar] [CrossRef] [PubMed]
  13. Cooper, A.A.; Gitler, A.D.; Cashikar, A.; Haynes, C.M.; Hill, K.J.; Bhullar, B.; Liu, K.; Xu, K.; Strathearn, K.E.; Liu, F.; et al. α-Synuclein Blocks ER-Golgi Traffic and Rab1 Rescues Neuron Loss in Parkinson’s Models. Science 2006, 313, 324–328. [Google Scholar] [CrossRef]
  14. Steger, M.; Tonelli, F.; Ito, G.; Davies, P.; Trost, M.; Vetter, M.; Wachter, S.; Lorentzen, E.; Duddy, G.; Wilson, S.; et al. Phosphoproteomics Reveals That Parkinson’s Disease Kinase LRRK2 Regulates a Subset of Rab GTPases. eLife 2016, 5, e12813. [Google Scholar] [CrossRef] [PubMed]
  15. 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. [Google Scholar] [CrossRef]
  16. Liu, Z.; Bryant, N.; Kumaran, R.; Beilina, A.; Abeliovich, A.; Cookson, M.R.; West, A.B. LRRK2 Phosphorylates Membrane-Bound Rabs and Is Activated by GTP-Bound Rab7L1 to Promote Recruitment to the Trans Golgi Network. Hum. Mol. Genet. 2018, 27, 385–395. [Google Scholar] [CrossRef]
  17. Madero-Pérez, J.; Fdez, E.; Fernández, B.; Ordóñez, A.J.L.; Ramírez, M.B.; Gómez-Suaga, P.; Waschbüsch, D.; Lobbestael, E.; Baekelandt, V.; Nairn, A.C.; et al. Parkinson Disease-Associated Mutations in LRRK2 Cause Centrosomal Defects via Rab8a Phosphorylation. Mol. Neurodegener. 2018, 13, 3. [Google Scholar] [CrossRef]
  18. Fujimoto, T.; Kuwahara, T.; Eguchi, T.; Sakurai, M.; Komori, T.; Iwatsubo, T. Parkinson’s Disease-Associated Mutant LRRK2 Phosphorylates Rab7L1 and Modifies Trans-Golgi Morphology. Biochem. Biophys. Res. Commun. 2018, 495, 1708–1715. [Google Scholar] [CrossRef]
  19. 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. [Google Scholar] [CrossRef]
  20. Funayama, M.; Hasegawa, K.; Kowa, H.; Saito, M.; Tsuji, S.; Obata, F. A New Locus for Parkinson’s Disease (PARK8) Maps to Chromosome 12p11.2–Q13.1. Ann. Neurol. 2002, 51, 296–301. [Google Scholar] [CrossRef]
  21. Aasly, J.O.; Vilariño-Güell, C.; Dachsel, J.C.; Webber, P.J.; West, A.B.; Haugarvoll, K.; Johansen, K.K.; Toft, M.; Nutt, J.G.; Payami, H.; et al. Novel Pathogenic LRRK2 p.Asn1437His Substitution in Familial Parkinson’s Disease. Mov. Disord. 2010, 25, 2156–2163. [Google Scholar] [CrossRef] [PubMed]
  22. Mata, I.F.; Kachergus, J.M.; Taylor, J.P.; Lincoln, S.; Aasly, J.; Lynch, T.; Hulihan, M.M.; Cobb, S.A.; Wu, R.-M.; Lu, C.-S.; et al. Lrrk2 Pathogenic Substitutions in Parkinson’s Disease. Neurogenetics 2005, 6, 171–177. [Google Scholar] [CrossRef] [PubMed]
  23. Kachergus, J.; Mata, I.F.; Hulihan, M.; Taylor, J.P.; Lincoln, S.; Aasly, J.; Gibson, J.M.; Ross, O.A.; Lynch, T.; Wiley, J.; et al. Identification of a Novel LRRK2 Mutation Linked to Autosomal Dominant Parkinsonism: Evidence of a Common Founder across European Populations. Am. J. Hum. Genet. 2005, 76, 672–680. [Google Scholar] [CrossRef] [PubMed]
  24. Kalogeropulou, A.F.; Purlyte, E.; Tonelli, F.; Lange, S.M.; Wightman, M.; Prescott, A.R.; Padmanabhan, S.; Sammler, E.; Alessi, D.R. Impact of 100 LRRK2 Variants Linked to Parkinson’s Disease on Kinase Activity and Microtubule Binding. Biochem. J. 2022, 479, 1759–1783. [Google Scholar] [CrossRef] [PubMed]
  25. Bosgraaf, L.; Haastert, P.J.M.V. Roc, a Ras/GTPase Domain in Complex Proteins. Biochim. Biophys. Acta BBA Mol. Cell Res. 2003, 1643, 5–10. [Google Scholar] [CrossRef]
  26. Guo, L.; Gandhi, P.N.; Wang, W.; Petersen, R.B.; Wilson-Delfosse, A.L.; Chen, S.G. The Parkinson’s Disease-Associated Protein, Leucine-Rich Repeat Kinase 2 (LRRK2), Is an Authentic GTPase Thatstimulates Kinase Activity. Exp. Cell Res. 2007, 313, 3658–3670. [Google Scholar] [CrossRef]
  27. Taymans, J.-M.; Vancraenenbroeck, R.; Ollikainen, P.; Beilina, A.; Lobbestael, E.; Maeyer, M.D.; Baekelandt, V.; Cookson, M.R. LRRK2 Kinase Activity Is Dependent on LRRK2 GTP Binding Capacity but Independent of LRRK2 GTP Binding. PLoS ONE 2011, 6, e23207. [Google Scholar] [CrossRef]
  28. Cogo, S.; Ho, F.Y.; Tosoni, E.; Tomkins, J.E.; Tessari, I.; Iannotta, L.; Montine, T.J.; Manzoni, C.; Lewis, P.A.; Bubacco, L.; et al. The Roc Domain of LRRK2 as a Hub for Protein-Protein Interactions: A Focus on PAK6 and Its Impact on RAB Phosphorylation. Brain Res. 2022, 1778, 147781. [Google Scholar] [CrossRef]
  29. Mills, R.D.; Liang, L.; Lio, D.S.; Mok, Y.; Mulhern, T.D.; Cao, G.; Griffin, M.; Kenche, V.B.; Culvenor, J.G.; Cheng, H. The Roc-COR Tandem Domain of Leucine-rich Repeat Kinase 2 Forms Dimers and Exhibits Conventional Ras-like GTPase Properties. J. Neurochem. 2018, 147, 409–428. [Google Scholar] [CrossRef]
  30. Myasnikov, A.; Zhu, H.; Hixson, P.; Xie, B.; Yu, K.; Pitre, A.; Peng, J.; Sun, J. Structural Analysis of the Full-Length Human LRRK2. Cell 2021, 184, 3519–3527.e10. [Google Scholar] [CrossRef]
  31. Sheng, Z.; Zhang, S.; Bustos, D.; Kleinheinz, T.; Pichon, C.E.; Dominguez, S.L.; Solanoy, H.O.; Drummond, J.; Zhang, X.; Ding, X.; et al. Ser1292 Autophosphorylation Is an Indicator of LRRK2 Kinase Activity and Contributes to the Cellular Effects of PD Mutations. Sci. Transl. Med. 2012, 4, 164ra161. [Google Scholar] [CrossRef]
  32. Christensen, K.; Hentzer, M.; Oppermann, F.S.; Elschenbroich, S.; Dossang, P.; Thirstrup, K.; Egebjerg, J.; Williamson, D.S.; Smith, G.P. LRRK2 Exonic Variants Associated with Parkinson’s Disease Augment Phosphorylation Levels for LRRK2-Ser1292 and Rab10-Thr73. bioRxiv 2018, 447946. [Google Scholar] [CrossRef]
  33. Mosavi, L.K.; Cammett, T.J.; Desrosiers, D.C.; Peng, Z. The Ankyrin Repeat as Molecular Architecture for Protein Recognition. Protein Sci. 2004, 13, 1435–1448. [Google Scholar] [CrossRef] [PubMed]
  34. Tewari, R.; Bailes, E.; Bunting, K.A.; Coates, J.C. Armadillo-Repeat Protein Functions: Questions for Little Creatures. Trends Cell Biol. 2010, 20, 470–481. [Google Scholar] [CrossRef] [PubMed]
  35. Matsushima, N.; Takatsuka, S.; Miyashita, H.; Kretsinger, R.H. Leucine Rich Repeat Proteins: Sequences, Mutations, Structures and Diseases. Protein Pept. Lett. 2019, 26, 108–131. [Google Scholar] [CrossRef] [PubMed]
  36. Xu, C.; Min, J. Structure and Function of WD40 Domain Proteins. Protein Cell 2011, 2, 202–214. [Google Scholar] [CrossRef]
  37. Schapira, M.; Tyers, M.; Torrent, M.; Arrowsmith, C.H. WD40 Repeat Domain Proteins: A Novel Target Class? Nat. Rev. Drug Discov. 2017, 16, 773–786. [Google Scholar] [CrossRef]
  38. Lee, H.; Flynn, R.; Sharma, I.; Haberman, E.; Carling, P.J.; Nicholls, F.J.; Stegmann, M.; Vowles, J.; Haenseler, W.; Wade-Martins, R.; et al. LRRK2 Is Recruited to Phagosomes and Co-Recruits RAB8 and RAB10 in Human Pluripotent Stem Cell-Derived Macrophages. Stem Cell Rep. 2020, 14, 940–955. [Google Scholar] [CrossRef]
  39. Ito, G.; Utsunomiya-Tate, N. Overview of the Impact of Pathogenic LRRK2 Mutations in Parkinson’s Disease. Biomolecules 2023, 13, 845. [Google Scholar] [CrossRef]
  40. Deniston, C.K.; Salogiannis, J.; Mathea, S.; Snead, D.M.; Lahiri, I.; Matyszewski, M.; Donosa, O.; Watanabe, R.; Böhning, J.; Shiau, A.K.; et al. Structure of LRRK2 in Parkinson’s Disease and Model for Microtubule Interaction. Nature 2020, 588, 344–349. [Google Scholar] [CrossRef]
  41. Guaitoli, G.; Gilsbach, B.K.; Raimondi, F.; Gloeckner, C.J. First Model of Dimeric LRRK2: The Challenge of Unrevealing the Structure of a Multidomain Parkinson’s-Associated Protein. Biochem. Soc. Trans. 2016, 44, 1635–1641. [Google Scholar] [CrossRef] [PubMed]
  42. Zhang, P.; Fan, Y.; Ru, H.; Wang, L.; Magupalli, V.G.; Taylor, S.S.; Alessi, D.R.; Wu, H. Crystal Structure of the WD40 Domain Dimer of LRRK2. Proc. Natl. Acad. Sci. USA 2019, 116, 1579–1584. [Google Scholar] [CrossRef] [PubMed]
  43. Watanabe, R.; Buschauer, R.; Böhning, J.; Audagnotto, M.; Lasker, K.; Lu, T.-W.; Boassa, D.; Taylor, S.; Villa, E. The In Situ Structure of Parkinson’s Disease-Linked LRRK2. Cell 2020, 182, 1508–1518.e16. [Google Scholar] [CrossRef]
  44. Zhu, H.; Tonelli, F.; Alessi, D.R.; Sun, J. Structural Basis of Human LRRK2 Membrane Recruitment and Activation. bioRxiv 2022. [Google Scholar] [CrossRef]
  45. Greggio, E.; Zambrano, I.; Kaganovich, A.; Beilina, A.; Taymans, J.-M.; Daniëls, V.; Lewis, P.; Jain, S.; Ding, J.; Syed, A.; et al. The Parkinson Disease-Associated Leucine-Rich Repeat Kinase 2 (LRRK2) Is a Dimer That Undergoes Intramolecular Autophosphorylation*. J. Biol. Chem. 2008, 283, 16906–16914. [Google Scholar] [CrossRef] [PubMed]
  46. Deng, J.; Lewis, P.A.; Greggio, E.; Sluch, E.; Beilina, A.; Cookson, M.R. Structure of the ROC Domain from the Parkinson’s Disease-Associated Leucine-Rich Repeat Kinase 2 Reveals a Dimeric GTPase. Proc. Natl. Acad. Sci. USA 2008, 105, 1499–1504. [Google Scholar] [CrossRef] [PubMed]
  47. 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. [Google Scholar] [CrossRef]
  48. Sejwal, K.; Chami, M.; Rémigy, H.; Vancraenenbroeck, R.; Sibran, W.; Sütterlin, R.; Baumgartner, P.; McLeod, R.; Chartier-Harlin, M.-C.; Baekelandt, V.; et al. Cryo-EM Analysis of Homodimeric Full-Length LRRK2 and LRRK1 Protein Complexes. Sci. Rep. 2017, 7, 8667. [Google Scholar] [CrossRef]
  49. Snead, D.M.; Matyszewski, M.; Dickey, A.M.; Lin, Y.X.; Leschziner, A.E.; Reck-Peterson, S.L. Structural Basis for Parkinson’s Disease-Linked LRRK2’s Binding to Microtubules. Nat. Struct. Mol. Biol. 2022, 29, 1196–1207. [Google Scholar] [CrossRef]
  50. Ito, G.; Iwatsubo, T. Re-Examination of the Dimerization State of Leucine-Rich Repeat Kinase 2: Predominance of the Monomeric Form. Biochem. J. 2012, 441, 987–998. [Google Scholar] [CrossRef]
  51. Kett, L.R.; Boassa, D.; Ho, C.C.-Y.; Rideout, H.J.; Hu, J.; Terada, M.; Ellisman, M.; Dauer, W.T. LRRK2 Parkinson Disease Mutations Enhance Its Microtubule Association. Hum. Mol. Genet. 2012, 21, 890–899. [Google Scholar] [CrossRef] [PubMed]
  52. Vides, E.G.; Adhikari, A.; Chiang, C.Y.; Lis, P.; Purlyte, E.; Limouse, C.; Shumate, J.L.; Spinola-Lasso, E.; Dhekne, H.S.; Alessi, D.R.; et al. A Feed-Forward Pathway Drives LRRK2 Kinase Membrane Recruitment and Activation. eLife 2022, 11, e79771. [Google Scholar] [CrossRef] [PubMed]
  53. Dhekne, H.S.; Tonelli, F.; Yeshaw, W.M.; Chiang, C.Y.; Limouse, C.; Jaimon, E.; Purlyte, E.; Alessi, D.R.; Pfeffer, S.R. Genome-Wide Screen Reveals Rab12 GTPase as a Critical Activator of Parkinson’s Disease-Linked LRRK2 Kinase. eLife 2023, 12, e87098. [Google Scholar] [CrossRef] [PubMed]
  54. Wang, X.; Bondar, V.V.; Davis, O.B.; Maloney, M.T.; Agam, M.; Chin, M.Y.; Ho, A.C.-N.; Ghosh, R.; Leto, D.E.; Joy, D.; et al. Rab12 Is a Regulator of LRRK2 and Its Activation by Damaged Lysosomes. eLife 2023, 12, e87255. [Google Scholar] [CrossRef] [PubMed]
  55. 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. [Google Scholar] [CrossRef]
  56. Piccoli, G.; Onofri, F.; Cirnaru, M.D.; Kaiser, C.J.O.; Jagtap, P.; Kastenmüller, A.; Pischedda, F.; Marte, A.; von Zweydorf, F.; Vogt, A.; et al. Leucine-Rich Repeat Kinase 2 Binds to Neuronal Vesicles through Protein Interactions Mediated by Its C-Terminal WD40 Domain. Mol. Cell Biol. 2014, 34, 2147–2161. [Google Scholar] [CrossRef]
  57. Cullen, P.J.; Steinberg, F. To Degrade or Not to Degrade: Mechanisms and Significance of Endocytic Recycling. Nat. Rev. Mol. Cell Biol. 2018, 19, 679–696. [Google Scholar] [CrossRef]
  58. Scott, C.C.; Vacca, F.; Gruenberg, J. Endosome Maturation, Transport and Functions. Semin. Cell Dev. Biol. 2014, 31, 2–10. [Google Scholar] [CrossRef]
  59. Wang, J.; Fedoseienko, A.; Chen, B.; Burstein, E.; Jia, D.; Billadeau, D.D. Endosomal Receptor Trafficking: Retromer and Beyond. Traffic 2018, 19, 578–590. [Google Scholar] [CrossRef]
  60. Langemeyer, L.; Fröhlich, F.; Ungermann, C. Rab GTPase Function in Endosome and Lysosome Biogenesis. Trends Cell Biol. 2018, 28, 957–970. [Google Scholar] [CrossRef]
  61. Gruenberg, J. Life in the Lumen: The Multivesicular Endosome. Traffic 2020, 21, 76–93. [Google Scholar] [CrossRef]
  62. O’Sullivan, M.J.; Lindsay, A.J. The Endosomal Recycling Pathway—At the Crossroads of the Cell. Int. J. Mol. Sci. 2020, 21, 6074. [Google Scholar] [CrossRef]
  63. Huotari, J.; Helenius, A. Endosome Maturation. EMBO J. 2011, 30, 3481–3500. [Google Scholar] [CrossRef]
  64. Campa, C.C.; Margaria, J.P.; Derle, A.; Giudice, M.D.; Santis, M.C.D.; Gozzelino, L.; Copperi, F.; Bosia, C.; Hirsch, E. Rab11 Activity and PtdIns(3)P Turnover Removes Recycling Cargo from Endosomes. Nat. Chem. Biol. 2018, 14, 801–810. [Google Scholar] [CrossRef]
  65. Bonifacino, J.S.; Rojas, R. Retrograde Transport from Endosomes to the Trans-Golgi Network. Nat. Rev. Mol. Cell Biol. 2006, 7, 568–579. [Google Scholar] [CrossRef]
  66. Mignogna, M.L.; D’Adamo, P. Critical Importance of RAB Proteins for Synaptic Function. Small GTPase 2018, 9, 145–157. [Google Scholar] [CrossRef]
  67. Fukuda, M. Rab GTPases: Key Players in Melanosome Biogenesis, Transport, and Transfer. Pigment. Cell Melanoma Res. 2021, 34, 222–235. [Google Scholar] [CrossRef]
  68. Lis, P.; Burel, S.; Steger, M.; Mann, M.; Brown, F.; Diez, F.; Tonelli, F.; Holton, J.L.; Ho, P.W.; Ho, S.-L.; et al. Development of Phospho-Specific Rab Protein Antibodies to Monitor in Vivo Activity of the LRRK2 Parkinson’s Disease Kinase. Biochem. J. 2018, 475, BCJ20170802. [Google Scholar] [CrossRef]
  69. Nirujogi, R.S.; Tonelli, F.; Taylor, M.; Lis, P.; Zimprich, A.; Sammler, E.; Alessi, D.R. Development of a Multiplexed Targeted Mass Spectrometry Assay for LRRK2-Phosphorylated Rabs and Ser910/Ser935 Biomarker Sites. Biochem. J. 2021, 478, 299–326. [Google Scholar] [CrossRef]
  70. Yun, H.J.; Kim, H.; Ga, I.; Oh, H.; Ho, D.H.; Kim, J.; Seo, H.; Son, I.; Seol, W. An Early Endosome Regulator, Rab5b, Is an LRRK2 Kinase Substrate. J. Biochem. 2015, 157, 485–495. [Google Scholar] [CrossRef]
  71. Ordóñez, A.; Fernández, B.; Fdez, E.; Romo-Lozano, M.; Madero-Pérez, J.; Lobbestael, E.; Baekelandt, V.; Aiastui, A.; Munaín, A.; Melrose, H.L.; et al. RAB8, RAB10 and RILPL1 Contribute to Both LRRK2 Kinase-Mediated Centrosomal Cohesion and Ciliogenesis Deficits. Hum. Mol. Genet. 2019, 28, 3552–3568. [Google Scholar] [CrossRef]
  72. Madero-Pérez, J.; Fernández, B.; Ordóñez, A.J.L.; Fdez, E.; Lobbestael, E.; Baekelandt, V.; Hilfiker, S. RAB7L1-Mediated Relocalization of LRRK2 to the Golgi Complex Causes Centrosomal Deficits via RAB8A. Front. Mol. Neurosci. 2018, 11, 417. [Google Scholar] [CrossRef]
  73. Yu, M.; Arshad, M.; Wang, W.; Zhao, D.; Xu, L.; Zhou, L. LRRK2 Mediated Rab8a Phosphorylation Promotes Lipid Storage. Lipids Health Dis. 2018, 17, 34. [Google Scholar] [CrossRef]
  74. Waschbüsch, D.; Purlyte, E.; Pal, P.; McGrath, E.; Alessi, D.R.; Khan, A.R. Structural Basis for Rab8a Recruitment of RILPL2 via LRRK2 Phosphorylation of Switch 2. Structure 2020, 28, 406–417.e6. [Google Scholar] [CrossRef]
  75. 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. Mutations in LRRK2 Linked to Parkinson Disease Sequester Rab8a to Damaged Lysosomes and Regulate Transferrin-Mediated Iron Uptake in Microglia. PLoS Biol. 2021, 19, e3001480. [Google Scholar] [CrossRef]
  76. Eguchi, T.; Kuwahara, T.; Sakurai, M.; Komori, T.; Fujimoto, T.; Ito, G.; Yoshimura, S.; Harada, A.; Fukuda, M.; Koike, M.; et al. LRRK2 and Its Substrate Rab GTPases Are Sequentially Targeted onto Stressed Lysosomes and Maintain Their Homeostasis. Proc. Natl. Acad. Sci. USA 2018, 115, E9115–E9124. [Google Scholar] [CrossRef]
  77. 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. [Google Scholar] [CrossRef]
  78. Ordóñez, A.J.L.; Fasiczka, R.; Fernández, B.; Naaldijk, Y.; Fdez, E.; Ramírez, M.B.; Phan, S.; Boassa, D.; Hilfiker, S. The LRRK2 Signaling Network Converges on a Centriolar Phospho-Rab10/RILPL1 Complex to Cause Deficits in Centrosome Cohesion and Cell Polarization. Biol. Open 2022, 11, bio059468. [Google Scholar] [CrossRef]
  79. 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. [Google Scholar] [CrossRef]
  80. Sobu, Y.; Wawro, P.S.; Dhekne, H.S.; Yeshaw, W.M.; Pfeffer, S.R. Pathogenic LRRK2 Regulates Ciliation Probability Upstream of Tau Tubulin Kinase 2 via Rab10 and RILPL1 Proteins. Proc. Natl. Acad. Sci. USA 2021, 118, e2005894118. [Google Scholar] [CrossRef]
  81. Liu, Z.; Xu, E.; Zhao, H.T.; Cole, T.; West, A.B. LRRK2 and Rab10 Coordinate Macropinocytosis to Mediate Immunological Responses in Phagocytes. EMBO J. 2020, 39, e104862. [Google Scholar] [CrossRef] [PubMed]
  82. Kluss, J.H.; Beilina, A.; Williamson, C.D.; Lewis, P.A.; Cookson, M.R.; Bonet-Ponce, L. Lysosomal Positioning Regulates Rab10 Phosphorylation at LRRK2+ Lysosomes. Proc. Natl. Acad. Sci. USA 2022, 119, e2205492119. [Google Scholar] [CrossRef]
  83. Bonet-Ponce, L.; Beilina, A.; Williamson, C.D.; Lindberg, E.; Kluss, J.H.; Saez-Atienzar, S.; Landeck, N.; Kumaran, R.; Mamais, A.; Bleck, C.K.E.; et al. LRRK2 Mediates Tubulation and Vesicle Sorting from Lysosomes. Sci. Adv. 2020, 6, eabb2454. [Google Scholar] [CrossRef] [PubMed]
  84. 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. [Google Scholar] [CrossRef] [PubMed]
  85. Li, J.; Wu, M.; Gong, Y.; Tang, J.; Shen, J.; Xu, L.; Dang, B.; Chen, G. Inhibition of LRRK2-Rab10 Pathway Improves Secondary Brain Injury After Surgical Brain Injury in Rats. Front. Surg. 2022, 8, 749310. [Google Scholar] [CrossRef]
  86. Ito, K.; Araki, M.; Katai, Y.; Nishimura, Y.; Imotani, S.; Inoue, H.; Ito, G.; Tomita, T. Pathogenic LRRK2 Compromises the Subcellular Distribution of Lysosomes in a Rab12-RILPL1-dependent Manner. FASEB J. 2023, 37, e22930. [Google Scholar] [CrossRef]
  87. Komori, T.; Kuwahara, T.; Fujimoto, T.; Sakurai, M.; Koyama-Honda, I.; Fukuda, M.; Iwatsubo, T. Phosphorylation of Rab29 at Ser185 Regulates Its Localization and Role in the Lysosomal Stress Response in Concert with LRRK2. J. Cell Sci. 2023, 136, jcs261003. [Google Scholar] [CrossRef]
  88. 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. [Google Scholar] [CrossRef]
  89. Homma, Y.; Hiragi, S.; Fukuda, M. Rab Family of Small GTPases: An Updated View on Their Regulation and Functions. FEBS J. 2021, 288, 36–55. [Google Scholar] [CrossRef]
  90. Ito, G.; Katsemonova, K.; Tonelli, F.; Lis, P.; Baptista, M.; Shpiro, N.; Duddy, G.; Wilson, S.; Ho, P.; Ho, S.-L.; et al. Phos-Tag Analysis of Rab10 Phosphorylation by LRRK2: A Powerful Assay for Assessing Kinase Function and Inhibitors. Biochem. J. 2016, 473, 2671–2685. [Google Scholar] [CrossRef]
  91. Fan, Y.; Howden, A.J.; Sarhan, A.R.; Lis, P.; Ito, G.; Martinez, T.N.; Brockmann, K.; Gasser, T.; Alessi, D.R.; Sammler, E.M. Interrogating Parkinson’s Disease LRRK2 Kinase Pathway Activity by Assessing Rab10 Phosphorylation in Human Neutrophils. Biochem. J. 2017, 475, 23–44. [Google Scholar] [CrossRef] [PubMed]
  92. Gomez, R.C.; Wawro, P.; Lis, P.; Alessi, D.R.; Pfeffer, S.R. Membrane Association but Not Identity Is Required for LRRK2 Activation and Phosphorylation of Rab GTPases. J. Cell Biol. 2019, 218, 4157–4170. [Google Scholar] [CrossRef] [PubMed]
  93. 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. [Google Scholar] [CrossRef] [PubMed]
  94. Iannotta, L.; Biosa, A.; Kluss, J.H.; Tombesi, G.; Kaganovich, A.; Cogo, S.; Plotegher, N.; Civiero, L.; Lobbestael, E.; Baekelandt, V.; et al. Divergent Effects of G2019S and R1441C LRRK2 Mutations on LRRK2 and Rab10 Phosphorylations in Mouse Tissues. Cells 2020, 9, 2344. [Google Scholar] [CrossRef]
  95. Kelly, K.; Chang, A.; Hastings, L.; Abdelmotilib, H.; West, A.B. Genetic Background Influences LRRK2-Mediated Rab Phosphorylation in the Rat Brain. Brain Res. 2021, 1759, 147372. [Google Scholar] [CrossRef]
  96. 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. [Google Scholar] [CrossRef]
  97. Keeney, M.; Hoffman, E.; Greenamyre, J.; Maio, R.D. Measurement of LRRK2 Kinase Activity by Proximity Ligation Assay. Bio-protocol 2021, 11, e4140. [Google Scholar] [CrossRef]
  98. Tasegian, A.; Singh, F.; Ganley, I.G.; Reith, A.D.; Alessi, D.R. Impact of Type II LRRK2 Inhibitors on Signaling and Mitophagy. Biochem. J. 2021, 478, 3555–3573. [Google Scholar] [CrossRef]
  99. Kluss, J.H.; Bonet-Ponce, L.; Lewis, P.A.; Cookson, M.R. Directing LRRK2 to Membranes of the Endolysosomal Pathway Triggers RAB Phosphorylation and JIP4 Recruitment. Neurobiol. Dis. 2022, 170, 105769. [Google Scholar] [CrossRef]
  100. Atashrazm, F.; Hammond, D.; Perera, G.; Bolliger, M.F.; Matar, E.; Halliday, G.M.; Schüle, B.; Lewis, S.J.G.; Nichols, R.J.; Dzamko, N. LRRK2-mediated Rab10 Phosphorylation in Immune Cells from Parkinson’s Disease Patients. Mov. Disord. 2019, 34, 406–415. [Google Scholar] [CrossRef]
  101. Fan, Y.; Tonelli, F.; Padmanabhan, S.; Baptista, M.A.S.; Riley, L.; Smith, D.; Marras, C.; Howden, A.; Alessi, D.R.; Sammler, E. Human Peripheral Blood Neutrophil Isolation for Interrogating the Parkinson’s Associated LRRK2 Kinase Pathway by Assessing Rab10 Phosphorylation. J. Vis. Exp. 2020, e58956. [Google Scholar] [CrossRef]
  102. Karayel, Ö.; Tonelli, F.; Winter, S.V.; Geyer, P.E.; Fan, Y.; Sammler, E.M.; Alessi, D.R.; Steger, M.; Mann, M. Accurate MS-Based Rab10 Phosphorylation Stoichiometry Determination as Readout for LRRK2 Activity in Parkinson’s Disease. Mol. Cell Proteom. 2020, 19, 1546–1560. [Google Scholar] [CrossRef] [PubMed]
  103. Wang, S.; Kelly, K.; Brotchie, J.M.; Koprich, J.B.; West, A.B. Exosome Markers of LRRK2 Kinase Inhibition. npj Park. Dis. 2020, 6, 32. [Google Scholar] [CrossRef] [PubMed]
  104. Bright, J.M.; Carlisle, H.J.; Toda, A.M.A.; Murphy, M.; Molitor, T.P.; Wren, P.; Andruska, K.M.; Liu, E.; Barlow, C. Differential Inhibition of LRRK2 in Parkinson’s Disease Patient Blood by a G2019S Selective LRRK2 Inhibitor. Mov. Disord. 2021, 36, 1362–1371. [Google Scholar] [CrossRef] [PubMed]
  105. Wang, X.; Negrou, E.; Maloney, M.T.; Bondar, V.V.; Andrews, S.V.; Montalban, M.; Llapashtica, C.; Maciuca, R.; Nguyen, H.; Solanoy, H.; et al. Understanding LRRK2 Kinase Activity in Preclinical Models and Human Subjects through Quantitative Analysis of LRRK2 and PT73 Rab10. Sci. Rep. 2021, 11, 12900. [Google Scholar] [CrossRef] [PubMed]
  106. Fan, Y.; Nirujogi, R.S.; Garrido, A.; Ruiz-Martínez, J.; Bergareche-Yarza, A.; Mondragón-Rezola, E.; Vinagre-Aragón, A.; Croitoru, I.; Pagola, A.G.; Markinez, L.P.; et al. R1441G but Not G2019S Mutation Enhances LRRK2 Mediated Rab10 Phosphorylation in Human Peripheral Blood Neutrophils. Acta Neuropathol. 2021, 142, 475–494. [Google Scholar] [CrossRef]
  107. Jennings, D.; Huntwork-Rodriguez, S.; Henry, A.G.; Sasaki, J.C.; Meisner, R.; Diaz, D.; Solanoy, H.; Wang, X.; Negrou, E.; Bondar, V.V.; et al. Preclinical and Clinical Evaluation of the LRRK2 Inhibitor DNL201 for Parkinson’s Disease. Sci. Transl. Med. 2022, 14, eabj2658. [Google Scholar] [CrossRef]
  108. 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 Park. Dis. 2022, 8, 73. [Google Scholar] [CrossRef]
  109. Wang, S.; Unnithan, S.; Bryant, N.; Chang, A.; Rosenthal, L.S.; Pantelyat, A.; Dawson, T.M.; Al-Khalidi, H.R.; West, A.B. Elevated Urinary Rab10 Phosphorylation in Idiopathic Parkinson Disease. Mov. Disord. 2022, 37, 1454–1464. [Google Scholar] [CrossRef]
  110. Chua, C.E.L.; Tang, B.L. Rab 10—A Traffic Controller in Multiple Cellular Pathways and Locations. J. Cell. Physiol. 2018, 233, 6483–6494. [Google Scholar] [CrossRef]
  111. Peränen, J. Rab8 GTPase as a Regulator of Cell Shape. Cytoskeleton 2011, 68, 527–539. [Google Scholar] [CrossRef]
  112. Taymans, J.-M.; Mutez, E.; Sibran, W.; Vandewynckel, L.; Deldycke, C.; Bleuse, S.; Marchand, A.; Sarchione, A.; Leghay, C.; Kreisler, A.; et al. Alterations in the LRRK2-Rab Pathway in Urinary Extracellular Vesicles as Parkinson’s Disease and Pharmacodynamic Biomarkers. npj Park. Dis. 2023, 9, 21. [Google Scholar] [CrossRef]
  113. Huber, L.A.; Pimplikar, S.; Parton, R.G.; Virta, H.; Zerial, M.; Simons, K. Rab8, a Small GTPase Involved in Vesicular Traffic between the TGN and the Basolateral Plasma Membrane. J. Cell Biol. 1993, 123, 35–45. [Google Scholar] [CrossRef] [PubMed]
  114. Huber, L.A.; de Hoop, M.J.; Dupree, P.; Zerial, M.; Simons, K.; Dotti, C. Protein Transport to the Dendritic Plasma Membrane of Cultured Neurons Is Regulated by Rab8p. J. Cell Biol. 1993, 123, 47–55. [Google Scholar] [CrossRef] [PubMed]
  115. Huber, L.A.; Dupree, P.; Dotti, C.G. A Deficiency of the Small GTPase Rab8 Inhibits Membrane Traffic in Developing Neurons. Mol. Cell. Biol. 1995, 15, 918–924. [Google Scholar] [CrossRef] [PubMed]
  116. Homma, Y.; Fukuda, M. Rabin8 Regulates Neurite Outgrowth in Both GEF Activity–Dependent and –Independent Manners. Mol. Biol. Cell. 2016, 27, 2107–2118. [Google Scholar] [CrossRef]
  117. Urrutia, P.J.; Bodaleo, F.; Bórquez, D.A.; Homma, Y.; Rozes-Salvador, V.; Villablanca, C.; Conde, C.; Fukuda, M.; González-Billault, C. Tuba Activates Cdc42 during Neuronal Polarization Downstream of the Small GTPase Rab8a. J. Neurosci. 2021, 41, 1636–1649. [Google Scholar] [CrossRef]
  118. Stypulkowski, E.; Feng, Q.; Joseph, I.; Farrell, V.; Flores, J.; Yu, S.; Sakamori, R.; Sun, J.; Bandyopadhyay, S.; Das, S.; et al. Rab8 Attenuates Wnt Signaling and Is Required for Mesenchymal Differentiation into Adipocytes. J. Biol. Chem. 2021, 296, 100488. [Google Scholar] [CrossRef]
  119. Sato, T.; Iwano, T.; Kunii, M.; Matsuda, S.; Mizuguchi, R.; Jung, Y.; Hagiwara, H.; Yoshihara, Y.; Yuzaki, M.; Harada, R.; et al. Rab8a and Rab8b Are Essential for Several Apical Transport Pathways but Insufficient for Ciliogenesis. J. Cell Sci. 2013, 127, 422–431. [Google Scholar] [CrossRef]
  120. Yin, G.; da Fonseca, T.L.; Eisbach, S.E.; Anduaga, A.M.; Breda, C.; Orcellet, M.L.; Szegő, É.M.; Guerreiro, P.; Lázaro, D.F.; Braus, G.H.; et al. α-Synuclein Interacts with the Switch Region of Rab8a in a Ser129 Phosphorylation-Dependent Manner. Neurobiol. Dis. 2014, 70, 149–161. [Google Scholar] [CrossRef]
  121. Corbier, C.; Sellier, C. C9ORF72 Is a GDP/GTP Exchange Factor for Rab8 and Rab39 and Regulates Autophagy. Small GTPases 2017, 8, 181–186. [Google Scholar] [CrossRef] [PubMed]
  122. Mamais, A.; Sanyal, A.; Fajfer, A.; Zykoski, C.G.; Guldin, M.; Riley-DiPaolo, A.; Subrahmanian, N.; Gibbs, W.; Lin, S.; LaVoie, M.J. The LRRK2 Kinase Substrates Rab8a and Rab10 Contribute Complementary but Distinct Disease-Relevant Phenotypes in Human Neurons. bioRxiv 2023. [Google Scholar] [CrossRef]
  123. Li, Z.; Schulze, R.J.; Weller, S.G.; Krueger, E.W.; Schott, M.B.; Zhang, X.; Casey, C.A.; Liu, J.; Stöckli, J.; James, D.E.; et al. A Novel Rab10-EHBP1-EHD2 Complex Essential for the Autophagic Engulfment of Lipid Droplets. Sci. Adv. 2016, 2, e1601470. [Google Scholar] [CrossRef] [PubMed]
  124. Rai, A.; Bleimling, N.; Vetter, I.R.; Goody, R.S. The Mechanism of Activation of the Actin Binding Protein EHBP1 by Rab8 Family Members. Nat. Commun. 2020, 11, 4187. [Google Scholar] [CrossRef] [PubMed]
  125. Simón-Sánchez, J.; Schulte, C.; Bras, J.M.; Sharma, M.; Gibbs, J.R.; Berg, D.; Paisan-Ruiz, C.; Lichtner, P.; Scholz, S.W.; Hernandez, D.G.; et al. Genome-Wide Association Study Reveals Genetic Risk Underlying Parkinson’s Disease. Nat. Genet. 2009, 41, 1308–1312. [Google Scholar] [CrossRef]
  126. Satake, W.; Nakabayashi, Y.; Mizuta, I.; Hirota, Y.; Ito, C.; Kubo, M.; Kawaguchi, T.; Tsunoda, T.; Watanabe, M.; Takeda, A.; et al. Genome-Wide Association Study Identifies Common Variants at Four Loci as Genetic Risk Factors for Parkinson’s Disease. Nat. Genet. 2009, 41, 1303–1307. [Google Scholar] [CrossRef]
  127. Pihlstrøm, L.; Rengmark, A.; Bjørnarå, K.A.; Dizdar, N.; Fardell, C.; Forsgren, L.; Holmberg, B.; Larsen, J.P.; Linder, J.; Nissbrandt, H.; et al. Fine Mapping and Resequencing of the PARK16 Locus in Parkinson’s Disease. J. Hum. Genet. 2015, 60, 357–362. [Google Scholar] [CrossRef]
  128. MacLeod, D.A.; Rhinn, H.; Kuwahara, T.; Zolin, A.; Di Paolo, G.; McCabe, B.D.; Marder, K.S.; Honig, L.S.; Clark, L.N.; Small, S.A.; et al. RAB7L1 Interacts with LRRK2 to Modify Intraneuronal Protein Sorting and Parkinson’s Disease Risk. Neuron 2013, 77, 425–439. [Google Scholar] [CrossRef]
  129. Beilina, A.; Rudenko, I.N.; Kaganovich, A.; Civiero, L.; Chau, H.; Kalia, S.K.; Kalia, L.V.; Lobbestael, E.; Chia, R.; Ndukwe, K.; et al. Unbiased Screen for Interactors of Leucine-Rich Repeat Kinase 2 Supports a Common Pathway for Sporadic and Familial Parkinson Disease. Proc. Natl. Acad. Sci. USA 2014, 111, 2626–2631. [Google Scholar] [CrossRef]
  130. Kuwahara, T.; Inoue, K.; D’Agati, V.D.; Fujimoto, T.; Eguchi, T.; Saha, S.; Wolozin, B.; Iwatsubo, T.; Abeliovich, A. LRRK2 and RAB7L1 Coordinately Regulate Axonal Morphology and Lysosome Integrity in Diverse Cellular Contexts. Sci. Rep. 2016, 6, 29945. [Google Scholar] [CrossRef]
  131. Unapanta, A.; Shavarebi, F.; Porath, J.; Shen, Y.; Balen, C.; Nguyen, A.; Tseng, J.; Leong, W.S.; Liu, M.; Lis, P.; et al. Endogenous Rab38 Regulates LRRK2’s Membrane Recruitment and Substrate Rab Phosphorylation in Melanocytes. J. Biol. Chem. 2023, 299, 105192. [Google Scholar] [CrossRef] [PubMed]
  132. Helip-Wooley, A.; Thoene, J.G. Sucrose-Induced Vacuolation Results in Increased Expression of Cholesterol Biosynthesis and Lysosomal Genes. Exp. Cell Res. 2004, 292, 89–100. [Google Scholar] [CrossRef] [PubMed]
  133. Kalogeropulou, A.F.; Freemantle, J.B.; Lis, P.; Vides, E.G.; Polinski, N.K.; Alessi, D.R. Endogenous Rab29 Does Not Impact Basal or Stimulated LRRK2 Pathway Activity. Biochem. J. 2020, 477, 4397–4423. [Google Scholar] [CrossRef]
  134. Wang, S.; Ma, Z.; Xu, X.; Wang, Z.; Sun, L.; Zhou, Y.; Lin, X.; Hong, W.; Wang, T. A Role of Rab29 in the Integrity of the Trans-Golgi Network and Retrograde Trafficking of Mannose-6-Phosphate Receptor. PLoS ONE 2014, 9, e96242. [Google Scholar] [CrossRef] [PubMed]
  135. Rivero-Ríos, P.; Romo-Lozano, M.; Fernández, B.; Fdez, E.; Hilfiker, S. Distinct Roles for RAB10 and RAB29 in Pathogenic LRRK2-Mediated Endolysosomal Trafficking Alterations. Cells 2020, 9, 1719. [Google Scholar] [CrossRef]
  136. Matsui, T.; Itoh, T.; Fukuda, M. Small GTPase Rab12 Regulates Constitutive Degradation of Transferrin Receptor. Traffic 2011, 12, 1432–1443. [Google Scholar] [CrossRef]
  137. Rydell, G.E.; Renard, H.; Garcia-Castillo, M.; Dingli, F.; Loew, D.; Lamaze, C.; Römer, W.; Johannes, L. Rab12 Localizes to Shiga Toxin-Induced Plasma Membrane Invaginations and Controls Toxin Transport. Traffic 2014, 15, 772–787. [Google Scholar] [CrossRef]
  138. Wang, J.; Lau, P.K.; Li, C.W.; Guo, Y. The Clathrin Adaptor Complex-1 and Rab12 Regulate Post-Golgi Trafficking of WT Epidermal Growth Factor Receptor (EGFR). J. Biol. Chem. 2023, 299, 102979. [Google Scholar] [CrossRef]
  139. Kluss, J.H.; Mazza, M.C.; Li, Y.; Manzoni, C.; Lewis, P.A.; Cookson, M.R.; Mamais, A. Preclinical Modeling of Chronic Inhibition of the Parkinson’s Disease Associated Kinase LRRK2 Reveals Altered Function of the Endolysosomal System in Vivo. Mol. Neurodegener. 2021, 16, 17. [Google Scholar] [CrossRef]
  140. Ito, G.; Tomita, T.; Utsunomiya-Tate, N. LRRK2-Mediated Phosphorylation and Thermal Stability of Rab12 Are Regulated by Bound Nucleotides. Biochem. Biophys. Res. Commun. 2023, 667, 43–49. [Google Scholar] [CrossRef]
  141. Klinkert, K.; Echard, A. Rab35 GTPase: A Central Regulator of Phosphoinositides and F-actin in Endocytic Recycling and Beyond. Traffic 2016, 17, 1063–1077. [Google Scholar] [CrossRef] [PubMed]
  142. Chen, P.-I.; Kong, C.; Su, X.; Stahl, P.D. Rab5 Isoforms Differentially Regulate the Trafficking and Degradation of Epidermal Growth Factor Receptors*. J. Biol. Chem. 2009, 284, 30328–30338. [Google Scholar] [CrossRef] [PubMed]
  143. Chen, P.-I.; Schauer, K.; Kong, C.; Harding, A.R.; Goud, B.; Stahl, P.D. Rab5 Isoforms Orchestrate a “Division of Labor” in the Endocytic Network; Rab5C Modulates Rac-Mediated Cell Motility. PLoS ONE 2014, 9, e90384. [Google Scholar] [CrossRef] [PubMed]
  144. Schmidt, O.; Weyer, Y.; Fink, M.J.; Müller, M.; Weys, S.; Bindreither, M.; Teis, D. Regulation of Rab5 Isoforms by Transcriptional and Post-transcriptional Mechanisms in Yeast. FEBS Lett. 2017, 591, 2803–2815. [Google Scholar] [CrossRef]
  145. Sung, J.Y.; Kim, J.; Paik, S.R.; Park, J.H.; Ahn, Y.S.; Chung, K.C. Induction of Neuronal Cell Death by Rab5A-Dependent Endocytosis of α-Synuclein*. J. Biol. Chem. 2001, 276, 27441–27448. [Google Scholar] [CrossRef]
  146. 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. [Google Scholar] [CrossRef]
  147. Heo, H.Y.; Kim, K.-S.; Seol, W. Coordinate Regulation of Neurite Outgrowth by LRRK2 and Its Interactor, Rab5. Exp. Neurobiol. 2010, 19, 97–105. [Google Scholar] [CrossRef]
  148. Lang, J. Molecular Mechanisms and Regulation of Insulin Exocytosis as a Paradigm of Endocrine Secretion. Eur. J. Biochem. 1999, 259, 3–17. [Google Scholar] [CrossRef]
  149. Geppert, M.; Bolshakov, V.Y.; Siegelbaum, S.A.; Takei, K.; Camilli, P.D.; Hammer, R.E.; Südhof, T.C. The Role of Rab3A in Neurotransmitter Release. Nature 1994, 369, 493–497. [Google Scholar] [CrossRef]
  150. Geppert, M.; Goda, Y.; Stevens, C.F.; Südhof, T.C. The Small GTP-Binding Protein Rab3A Regulates a Late Step in Synaptic Vesicle Fusion. Nature 1997, 387, 810–814. [Google Scholar] [CrossRef]
  151. Riedel, D.; Antonin, W.; Fernandez-Chacon, R.; de Toledo, G.A.; Jo, T.; Geppert, M.; Valentijn, J.A.; Valentijn, K.; Jamieson, J.D.; Südhof, T.C.; et al. Rab3D Is Not Required for Exocrine Exocytosis but for Maintenance of Normally Sized Secretory Granules. Mol. Cell Biol. 2002, 22, 6487–6497. [Google Scholar] [CrossRef] [PubMed]
  152. Schlüter, O.M.; Schmitz, F.; Jahn, R.; Rosenmund, C.; Südhof, T.C. A Complete Genetic Analysis of Neuronal Rab3 Function. J. Neurosci. 2004, 24, 6629–6637. [Google Scholar] [CrossRef] [PubMed]
  153. Persoon, C.M.; Hoogstraaten, R.I.; Nassal, J.P.; van Weering, J.R.T.; Kaeser, P.S.; Toonen, R.F.; Verhage, M. The RAB3-RIM Pathway Is Essential for the Release of Neuromodulators. Neuron 2019, 104, 1065–1080.e12. [Google Scholar] [CrossRef] [PubMed]
  154. Encarnação, M.; Espada, L.; Escrevente, C.; Mateus, D.; Ramalho, J.; Michelet, X.; Santarino, I.; Hsu, V.W.; Brenner, M.B.; Barral, D.C.; et al. A Rab3a-Dependent Complex Essential for Lysosome Positioning and Plasma Membrane Repair. J. Cell Biol. 2016, 213, 631–640. [Google Scholar] [CrossRef] [PubMed]
  155. Raffaniello, R.D. Rab3 Proteins and Cancer: Exit Strategies. J. Cell. Biochem. 2021, 122, 1295–1301. [Google Scholar] [CrossRef]
  156. Dalfó, E.; Barrachina, M.; Rosa, J.L.; Ambrosio, S.; Ferrer, I. Abnormal α-Synuclein Interactions with Rab3a and Rabphilin in Diffuse Lewy Body Disease. Neurobiol. Dis. 2004, 16, 92–97. [Google Scholar] [CrossRef]
  157. Chen, R.H.C.; Wislet-Gendebien, S.; Samuel, F.; Visanji, N.P.; Zhang, G.; Marsilio, D.; Langman, T.; Fraser, P.E.; Tandon, A. α-Synuclein Membrane Association Is Regulated by the Rab3a Recycling Machinery and Presynaptic Activity. J. Biol. Chem. 2013, 288, 7438–7449. [Google Scholar] [CrossRef]
  158. Lv, G.; Ko, M.S.; Das, T.; Eliezer, D. Molecular and Functional Interactions of Alpha-Synuclein with Rab3a. J. Biol. Chem. 2022, 298, 102239. [Google Scholar] [CrossRef]
  159. Petridi, S.; Middleton, A.C.; Ugbode, C.; Fellgett, A.; Covill, L.; Elliott, C. In Vivo Visual Screen for Dopaminergic Rab ↔ LRRK2-G2019S Interactions in Drosophila Discriminates Rab10 from Rab3. G3 Genes Genomes Genet. 2020, 10, 1903–1914. [Google Scholar] [CrossRef]
  160. Hatoyama, Y.; Homma, Y.; Hiragi, S.; Fukuda, M. Establishment and Analysis of Conditional Rab1 and Rab5 Knockout Cells by Using the Auxin-Inducible Degron System. J. Cell Sci. 2021, 134, jcs259184. [Google Scholar] [CrossRef]
  161. Tomás, M.; Martínez-Alonso, E.; Martínez-Martínez, N.; Cara-Esteban, M.; Martínez-Menárguez, J.A. Fragmentation of the Golgi Complex of Dopaminergic Neurons in Human Substantia Nigra: New Cytopathological Findings in Parkinson’s Disease. Histol. Histopathol. 2020, 36, 47–60. [Google Scholar] [CrossRef] [PubMed]
  162. Winslow, A.R.; Chen, C.-W.; Corrochano, S.; Acevedo-Arozena, A.; Gordon, D.E.; Peden, A.A.; Lichtenberg, M.; Menzies, F.M.; Ravikumar, B.; Imarisio, S.; et al. α-Synuclein Impairs Macroautophagy: Implications for Parkinson’s Disease. J. Cell Biol. 2010, 190, 1023–1037. [Google Scholar] [CrossRef] [PubMed]
  163. Yang, X.-Z.; Li, X.-X.; Zhang, Y.-J.; Rodriguez-Rodriguez, L.; Xiang, M.-Q.; Wang, H.-Y.; Zheng, X.F.S. Rab1 in Cell Signaling, Cancer and Other Diseases. Oncogene 2016, 35, 5699–5704. [Google Scholar] [CrossRef]
  164. Martínez-Menárguez, J.Á.; Martínez-Alonso, E.; Cara-Esteban, M.; Tomás, M. Focus on the Small GTPase Rab1: A Key Player in the Pathogenesis of Parkinson’s Disease. Int. J. Mol. Sci. 2021, 22, 12087. [Google Scholar] [CrossRef]
  165. Homma, Y.; Kinoshita, R.; Kuchitsu, Y.; Wawro, P.S.; Marubashi, S.; Oguchi, M.E.; Ishida, M.; Fujita, N.; Fukuda, M. Comprehensive Knockout Analysis of the Rab Family GTPases in Epithelial Cells. J. Cell Biol. 2019, 218, 2035–2050. [Google Scholar] [CrossRef] [PubMed]
  166. Khan, S.S.; Sobu, Y.; Dhekne, H.S.; Tonelli, F.; Berndsen, K.; Alessi, D.R.; Pfeffer, S.R. Pathogenic LRRK2 Control of Primary Cilia and Hedgehog Signaling in Neurons and Astrocytes of Mouse Brain. eLife 2021, 10, e67900. [Google Scholar] [CrossRef]
  167. Pajarillo, E.; Kim, S.; Digman, A.; Dutton, M.; Son, D.-S.; Aschner, M.; Lee, E. The Role of Microglial LRRK2 Kinase in Manganese-Induced Inflammatory Neurotoxicity via NLRP3 Inflammasome and RAB10-Mediated Autophagy Dysfunction. J. Biol. Chem. 2023, 299, 104879. [Google Scholar] [CrossRef]
  168. Maio, R.D.; Hoffman, E.K.; Rocha, E.M.; Keeney, M.T.; Sanders, L.H.; Miranda, B.R.D.; Zharikov, A.; Laar, A.V.; Stepan, A.F.; Lanz, T.A.; et al. LRRK2 Activation in Idiopathic Parkinson’s Disease. Sci. Transl. Med. 2018, 10, eaar5429. [Google Scholar] [CrossRef]
  169. Fraser, K.B.; Rawlins, A.B.; Clark, R.G.; Alcalay, R.N.; Standaert, D.G.; Liu, N.; Consortium, P.D.B.P.; West, A.B. Ser(P)-1292 LRRK2 in Urinary Exosomes Is Elevated in Idiopathic Parkinson’s Disease. Mov. Disord. 2016, 31, 1543–1550. [Google Scholar] [CrossRef]
  170. 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. 2021, 31, 2140–2154.e6. [Google Scholar] [CrossRef]
  171. Kingwell, K. LRRK2-Targeted Parkinson Disease Drug Advances into Phase III. Nat. Rev. Drug Discov. 2023, 22, 3–5. [Google Scholar] [CrossRef] [PubMed]
  172. Jennings, D.; Huntwork-Rodriguez, S.; Vissers, M.F.J.M.; Daryani, V.M.; Diaz, D.; Goo, M.S.; Chen, J.J.; Maciuca, R.; Fraser, K.; Mabrouk, O.S.; et al. LRRK2 Inhibition by BIIB122 in Healthy Participants and Patients with Parkinson’s Disease. Mov. Disord. 2023, 38, 386–398. [Google Scholar] [CrossRef] [PubMed]
Figure 2. Localizations and functions of LRRK2-phosphorylated Rabs. Green circles with numbers indicate Rabs phosphorylated by LRRK2. LRRK2-phosphorylated Rab12 and Rab29 are recruited to damaged or stressed lysosomes and further activate LRRK2. LRRK2-phosphorylated Rab8 and Rab10, bound to their effector RILPL1, accumulate near centrosomes to inhibit ciliogenesis. Rab8 and Rab10 also act together with their effector EHBP1 or EHBP1L1 to counteract lysosomal inflation or facilitate lysosomal release. LRRK2-phosphorylated Rab10 and Rab35 bind to JIP4 and induce LYTL. LRRK2-phosphorylated Rab12 binds to RILPL1 and moves lysosomes to the perinuclear region. LRRK2-phosphorylated Rab29 alters the morphology of the trans-Golgi. L: Lysosomes. Figure created with BioRender.com.
Figure 2. Localizations and functions of LRRK2-phosphorylated Rabs. Green circles with numbers indicate Rabs phosphorylated by LRRK2. LRRK2-phosphorylated Rab12 and Rab29 are recruited to damaged or stressed lysosomes and further activate LRRK2. LRRK2-phosphorylated Rab8 and Rab10, bound to their effector RILPL1, accumulate near centrosomes to inhibit ciliogenesis. Rab8 and Rab10 also act together with their effector EHBP1 or EHBP1L1 to counteract lysosomal inflation or facilitate lysosomal release. LRRK2-phosphorylated Rab10 and Rab35 bind to JIP4 and induce LYTL. LRRK2-phosphorylated Rab12 binds to RILPL1 and moves lysosomes to the perinuclear region. LRRK2-phosphorylated Rab29 alters the morphology of the trans-Golgi. L: Lysosomes. Figure created with BioRender.com.
Biomolecules 13 01645 g002
Table 1. Studies on leucine-rich repeat kinase 2 (LRRK2) 3D structure.
Table 1. Studies on leucine-rich repeat kinase 2 (LRRK2) 3D structure.
Model of LRRK2MethodPart of LRRK2
Analyzed
InteractionsResolutionPDB IDYearRef.
homodimerCrystallographyLRRK2 ROC domainDimerization of ROC domain2.0 Å2ZEJ2008[46]
homodimerNegative-stain EMFull-length Strep/FLAG-LRRK2-22 ÅN/A2016[47]
homodimerCryo-EMFull-length 3xFLAG-LRRK2-16 ÅN/A2017[48]
homodimerCrystallographyLRRK2 WD40 domainDimerization of WD40 domain2.6 Å6DLO/
6DLP
2019[42]
filamentousCryo-ETFull-length LRRK2 (I2020T)COR:COR
WD40:WD40
14 Å6XR42020[43]
filamentousCryo-EMLRRK2 RCKW 1COR:COR
WD40:WD40
3.5 Å6VNO (6VP6/6VP7
/6VP8)
2020[40]
monomer
homodimer
Cryo-EMFull-length LRRK2COR:COR3.7 Å
3.5 Å
7LHW
7LHT
2021[30]
monomer
dimer
tetramer (all with Rab29)
Cryo-EMFull-length LRRK2LRRK2:Rab29
COR:COR
WD40:kinase
3.5 ÅN/A2022[44]
filamentousCryo-EMLRRK2 RCKW 1COR:COR
WD40:WD40
5.0 Å7THY/
7THZ
2022[49]
1 RCKW: LRRK2 C-terminal half containing ROC, COR, KIN, and WD40 domains.
Table 2. Summary of Rab GTPase phosphorylation by LRRK2.
Table 2. Summary of Rab GTPase phosphorylation by LRRK2.
RabPhosphorylation Site by LRRK2
([68] Unless Otherwise Noted)
Effect of Phosphorylation
Rab1Thr75 [69]Unknown
Rab3Thr86Unknown
Rab5Ser84, Thr6 (in vitro [70])Delay in EGFR degradation [70]
Rab8Thr72Inhibition of ciliogenesis and centrosome cohesion [17,71,72]
Decreased binding with Optineurin [73]
Increased binding with RILPL2 [74]
Mistrafficking of cargo to damaged lysosomes [75]
Suppression of enlargement of stressed lysosomes [76]
Activation of LRRK2 [52]
Rab10Thr73Inhibition of ciliogenesis and centrosome cohesion [15,71,77,78,79,80]
Decreased binding to EHBP1L1 [81]
Induction of LYTL by recruitment of JIP4 [82,83]
Extracellular release of lysosomal contents under lysosomal stress [76]
Lowers GCase activity [84]
Induction of apoptosis after injury [85]
Rab12Ser106Perinuclear clustering of lysosomes via increase in RILPL1 binding [86]
Activation and recruitment of LRRK2 [53,54]
Rab29Thr71, Ser72Regulation of trans-Golgi morphology [18]
Recruitment of Rab29 itself to stressed lysosomes [87]
Rab35Thr72Induction of LYTL by recruitment of JIP4 [83]
Increase in α-synuclein propagation [88]
Rab43Thr82Unknown
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Komori, T.; Kuwahara, T. An Update on the Interplay between LRRK2, Rab GTPases and Parkinson’s Disease. Biomolecules 2023, 13, 1645. https://doi.org/10.3390/biom13111645

AMA Style

Komori T, Kuwahara T. An Update on the Interplay between LRRK2, Rab GTPases and Parkinson’s Disease. Biomolecules. 2023; 13(11):1645. https://doi.org/10.3390/biom13111645

Chicago/Turabian Style

Komori, Tadayuki, and Tomoki Kuwahara. 2023. "An Update on the Interplay between LRRK2, Rab GTPases and Parkinson’s Disease" Biomolecules 13, no. 11: 1645. https://doi.org/10.3390/biom13111645

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