Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
NPJ Parkinsons Dis ; 10(1): 12, 2024 Jan 08.
Article in English | MEDLINE | ID: mdl-38191886

ABSTRACT

Parkinson´s disease (PD) is a common neurodegenerative movement disorder and leucine-rich repeat kinase 2 (LRRK2) is a promising therapeutic target for disease intervention. However, the ability to stratify patients who will benefit from such treatment modalities based on shared etiology is critical for the success of disease-modifying therapies. Ciliary and centrosomal alterations are commonly associated with pathogenic LRRK2 kinase activity and can be detected in many cell types. We previously found centrosomal deficits in immortalized lymphocytes from G2019S-LRRK2 PD patients. Here, to investigate whether such deficits may serve as a potential blood biomarker for PD which is susceptible to LRKK2 inhibitor treatment, we characterized patient-derived cells from distinct PD cohorts. We report centrosomal alterations in peripheral cells from a subset of early-stage idiopathic PD patients which is mitigated by LRRK2 kinase inhibition, supporting a role for aberrant LRRK2 activity in idiopathic PD. Centrosomal defects are detected in R1441G-LRRK2 and G2019S-LRRK2 PD patients and in non-manifesting LRRK2 mutation carriers, indicating that they accumulate prior to a clinical PD diagnosis. They are present in immortalized cells as well as in primary lymphocytes from peripheral blood. These findings indicate that analysis of centrosomal defects as a blood-based patient stratification biomarker may help nominate idiopathic PD patients who will benefit from LRRK2-related therapeutics.

2.
STAR Protoc ; 4(1): 102024, 2023 03 17.
Article in English | MEDLINE | ID: mdl-36856766

ABSTRACT

The present protocol allows for quantification of inter-centrosome distances in G2 phase cells by confocal fluorescence microscopy to determine centrosome cohesion deficits. We describe transfection and immunofluorescence approaches followed by image acquisition and analysis of inter-centrosome distances. This protocol is for adherent A549 cells transiently overexpressing pathogenic LRRK2 and for immortalized murine embryonic fibroblasts endogenously expressing LRRK2 but is amenable to any other cultured cell type as well. For complete details on the use and execution of this protocol, please refer to Fdez et al.1 and Lara Ordóñez et al.2.


Subject(s)
Centrosome , Coleoptera , Animals , Mice , Humans , Cell Line , A549 Cells , Microscopy, Confocal
3.
Cell Death Dis ; 13(12): 1047, 2022 12 16.
Article in English | MEDLINE | ID: mdl-36522443

ABSTRACT

Increased brain iron content has been consistently reported in sporadic Parkinson's disease (PD) patients, and an increase in cytosolic free iron is known to cause oxidative stress and cell death. However, whether iron also accumulates in susceptible brain areas in humans or in mouse models of familial PD remains unknown. In addition, whilst the lysosome functions as a critical intracellular iron storage organelle, little is known about the mechanisms underlying lysosomal iron release and how this process is influenced by lysosome biogenesis and/or lysosomal exocytosis. Here, we report an increase in brain iron content also in PD patients due to the common G2019S-LRRK2 mutation as compared to healthy age-matched controls, whilst differences in iron content are not observed in G2019S-LRRK2 knockin as compared to control mice. Chemically triggering iron overload in cultured cells causes cytotoxicity via the endolysosomal release of iron which is mediated by TRPML1. TFEB expression reverts the iron overload-associated cytotoxicity by causing lysosomal exocytosis, which is dependent on a TRPML1-mediated increase in cytosolic calcium levels. Therefore, approaches aimed at increasing TFEB levels, or pharmacological TRPML1 activation in conjunction with iron chelation may prove beneficial against cell death associated with iron overload conditions such as those associated with PD.


Subject(s)
Iron Overload , Transient Receptor Potential Channels , Humans , Mice , Animals , Iron/metabolism , Transient Receptor Potential Channels/genetics , Transient Receptor Potential Channels/metabolism , Calcium/metabolism , Lysosomes/metabolism , Iron Overload/metabolism , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism
4.
Biol Open ; 11(8)2022 08 15.
Article in English | MEDLINE | ID: mdl-35776681

ABSTRACT

The Parkinson's-disease-associated LRRK2 kinase phosphorylates multiple Rab GTPases including Rab8 and Rab10, which enhances their binding to RILPL1 and RILPL2. The nascent interaction between phospho-Rab10 and RILPL1 blocks ciliogenesis in vitro and in the intact brain, and interferes with the cohesion of duplicated centrosomes in dividing cells. We show here that regulators of the LRRK2 signaling pathway including vps35 and PPM1H converge upon causing centrosomal deficits. The cohesion alterations do not require the presence of other LRRK2 kinase substrates including Rab12, Rab35 and Rab43 or the presence of RILPL2. Rather, they depend on the RILPL1-mediated centrosomal accumulation of phosphorylated Rab10. RILPL1 localizes to the subdistal appendage of the mother centriole, followed by recruitment of the LRRK2-phosphorylated Rab proteins to cause the centrosomal defects. The centrosomal alterations impair cell polarization as monitored by scratch wound assays which is reverted by LRRK2 kinase inhibition. These data reveal a common molecular pathway by which enhanced LRRK2 kinase activity impacts upon centrosome-related events to alter the normal biology of a cell.


Subject(s)
Centrioles , Centrosome , Centrioles/metabolism , Centrosome/metabolism , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/genetics , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/metabolism , Phosphorylation , Signal Transduction
5.
iScience ; 25(6): 104476, 2022 Jun 17.
Article in English | MEDLINE | ID: mdl-35721463

ABSTRACT

Mutations in LRRK2 increase its kinase activity and cause Parkinson's disease. LRRK2 phosphorylates a subset of Rab proteins which allows for their binding to RILPL1. The phospho-Rab/RILPL1 interaction causes deficits in ciliogenesis and interferes with the cohesion of duplicated centrosomes. We show here that centrosomal deficits mediated by pathogenic LRRK2 can also be observed in patient-derived iPS cells, and we have used transiently transfected cell lines to identify the underlying mechanism. The LRRK2-mediated centrosomal cohesion deficits are dependent on both the GTP conformation and phosphorylation status of the Rab proteins. Pathogenic LRRK2 does not displace proteinaceous linker proteins which hold duplicated centrosomes together, but causes the centrosomal displacement of CDK5RAP2, a protein critical for centrosome cohesion. The LRRK2-mediated centrosomal displacement of CDK5RAP2 requires RILPL1 and phospho-Rab proteins, which stably associate with centrosomes. These data provide fundamental information as to how pathogenic LRRK2 alters the normal physiology of a cell.

6.
J Parkinsons Dis ; 12(5): 1423-1447, 2022.
Article in English | MEDLINE | ID: mdl-35599495

ABSTRACT

BACKGROUND: Coding variation in the Leucine rich repeat kinase 2 gene linked to Parkinson's disease (PD) promotes enhanced activity of the encoded LRRK2 kinase, particularly with respect to autophosphorylation at S1292 and/or phosphorylation of the heterologous substrate RAB10. OBJECTIVE: To determine the inter-laboratory reliability of measurements of cellular LRRK2 kinase activity in the context of wildtype or mutant LRRK2 expression using published protocols. METHODS: Benchmark western blot assessments of phospho-LRRK2 and phospho-RAB10 were performed in parallel with in situ immunological approaches in HEK293T, mouse embryonic fibroblasts, and lymphoblastoid cell lines. Rat brain tissue, with or without adenovirus-mediated LRRK2 expression, and human brain tissues from subjects with or without PD, were also evaluated for LRRK2 kinase activity markers. RESULTS: Western blots were able to detect extracted LRRK2 activity in cells and tissue with pS1292-LRRK2 or pT73-RAB10 antibodies. However, while LRRK2 kinase signal could be detected at the cellular level with over-expressed mutant LRRK2 in cell lines, we were unable to demonstrate specific detection of endogenous cellular LRRK2 activity in cell culture models or tissues that we evaluated. CONCLUSION: Further development of reliable methods that can be deployed in multiple laboratories to measure endogenous LRRK2 activities are likely required, especially at cellular resolution.


Subject(s)
Parkinson Disease , rab GTP-Binding Proteins , Animals , Fibroblasts/metabolism , HEK293 Cells , Humans , Leucine/genetics , Leucine/metabolism , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/genetics , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/metabolism , Mice , Mutation , Parkinson Disease/genetics , Parkinson Disease/metabolism , Phosphorylation , Rats , Reproducibility of Results , rab GTP-Binding Proteins/genetics , rab GTP-Binding Proteins/metabolism
7.
Cells ; 9(7)2020 07 17.
Article in English | MEDLINE | ID: mdl-32709066

ABSTRACT

Mutations in the gene encoding leucine-rich repeat kinase 2 (LRRK2) cause familial Parkinson's disease, and sequence variations are associated with the sporadic form of the disease. LRRK2 phosphorylates a subset of RAB proteins implicated in secretory and recycling trafficking pathways, including RAB8A and RAB10. Another RAB protein, RAB29, has been reported to recruit LRRK2 to the Golgi, where it stimulates its kinase activity. Our previous studies revealed that G2019S LRRK2 expression or knockdown of RAB8A deregulate epidermal growth factor receptor (EGFR) trafficking, with a concomitant accumulation of the receptor in a RAB4-positive recycling compartment. Here, we show that the G2019S LRRK2-mediated EGFR deficits are mimicked by knockdown of RAB10 and rescued by expression of active RAB10. By contrast, RAB29 knockdown is without effect, but expression of RAB29 also rescues the pathogenic LRRK2-mediated trafficking deficits independently of Golgi integrity. Our data suggest that G2019S LRRK2 deregulates endolysosomal trafficking by impairing the function of RAB8A and RAB10, while RAB29 positively modulates non-Golgi-related trafficking events impaired by pathogenic LRRK2.


Subject(s)
Endosomes/metabolism , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/metabolism , Lysosomes/metabolism , rab GTP-Binding Proteins/metabolism , Epidermal Growth Factor/metabolism , Gene Knockdown Techniques , Golgi Apparatus/metabolism , HeLa Cells , Humans , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/genetics , Mutation/genetics , Protein Transport , Proteolysis , rab4 GTP-Binding Proteins/metabolism , rab7 GTP-Binding Proteins
8.
Biochem J ; 476(19): 2797-2813, 2019 10 15.
Article in English | MEDLINE | ID: mdl-31527116

ABSTRACT

Leucine-rich repeat kinase 2 (LRRK2) is a promising therapeutic target for the treatment of Parkinson's disease (PD), and orally bioavailable, brain penetrant and highly potent LRRK2 kinase inhibitors are in early stages of clinical testing. Detection of LRRK2 phosphorylation, as well as phosphorylation of Rab10, a LRRK2 kinase substrate, have been proposed as target engagement biomarkers for LRRK2 inhibitor clinical trials. However, these readouts do not seem able to stratify patients based on enhanced LRRK2 kinase activity. Here, we describe a robust cell biological assay based on centrosomal cohesion alterations which were observed in peripheral blood mononuclear cell-derived lymphoblastoid cell lines (LCLs) from patients with G2019S LRRK2 mutations as compared with healthy controls, and could also be detected in a subset of sporadic PD patient samples. We suggest that LCLs may be a valuable resource for LRRK2 research, and that determination of centrosomal cohesion deficits may assist in the stratification of a subset of sporadic PD patients.


Subject(s)
Centrosome/metabolism , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/metabolism , Leukocytes, Mononuclear/metabolism , Parkinson Disease/metabolism , Adult , Aged , Biomarkers/metabolism , Cell Line, Tumor , Female , Humans , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/antagonists & inhibitors , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/genetics , Leukocytes, Mononuclear/pathology , Male , Middle Aged , Phosphorylation
9.
Hum Mol Genet ; 28(21): 3552-3568, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31428781

ABSTRACT

Mutations in the LRRK2 kinase are the most common cause of familial Parkinson's disease, and variants increase risk for the sporadic form of the disease. LRRK2 phosphorylates multiple RAB GTPases including RAB8A and RAB10. Phosphorylated RAB10 is recruited to centrosome-localized RILPL1, which may interfere with ciliogenesis in a disease-relevant context. Our previous studies indicate that the centrosomal accumulation of phosphorylated RAB8A causes centrosomal cohesion deficits in dividing cells, including in peripheral patient-derived cells. Here, we show that both RAB8 and RAB10 contribute to the centrosomal cohesion deficits. Pathogenic LRRK2 causes the centrosomal accumulation not only of phosho-RAB8 but also of phospho-RAB10, and the effects on centrosomal cohesion are dependent on RAB8, RAB10 and RILPL1. Conversely, the pathogenic LRRK2-mediated ciliogenesis defects correlate with the centrosomal accumulation of both phospho-RAB8 and phospho-RAB10. LRRK2-mediated centrosomal cohesion and ciliogenesis alterations are observed in patient-derived peripheral cells, as well as in primary astrocytes from mutant LRRK2 mice, and are reverted upon LRRK2 kinase inhibition. These data suggest that the LRRK2-mediated centrosomal cohesion and ciliogenesis defects are distinct cellular readouts of the same underlying phospho-RAB8/RAB10/RILPL1 nexus and highlight the possibility that either centrosomal cohesion and/or ciliogenesis alterations may serve as cellular biomarkers for LRRK2-related PD.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Centrosome/metabolism , Ciliopathies/metabolism , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/metabolism , rab GTP-Binding Proteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Ciliopathies/enzymology , Ciliopathies/genetics , Humans , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/genetics , Phosphorylation , rab GTP-Binding Proteins/genetics
10.
Front Mol Neurosci ; 11: 417, 2018.
Article in English | MEDLINE | ID: mdl-30483055

ABSTRACT

Mutations in the LRRK2 gene cause autosomal-dominant Parkinson's disease (PD), and both LRRK2 as well as RAB7L1 have been implicated in increased susceptibility to idiopathic PD. RAB7L1 has been shown to increase membrane-association and kinase activity of LRRK2, and both seem to be mechanistically implicated in the same pathway. Another RAB protein, RAB8A, has been identified as a prominent LRRK2 kinase substrate, and our recent work demonstrates that aberrant LRRK2-mediated phosphorylation of RAB8A leads to centrosomal alterations. Here, we show that RAB7L1 recruits LRRK2 to the Golgi complex, which causes accumulation of phosphorylated RAB8A in a pericentrosomal/centrosomal location as well as centrosomal deficits identical to those observed with pathogenic LRRK2. The centrosomal alterations induced by wildtype LRRK2 in the presence of RAB7L1 depend on Golgi integrity. This is in contrast to pathogenic LRRK2 mutants, which cause centrosomal deficits independent of Golgi integrity or largely independent on RAB7L1 expression. Furthermore, centrosomal alterations in the presence of wildtype LRRK2 and RAB7L1 are at least in part mediated by aberrant LRRK2-mediated RAB8A phosphorylation, as abolished by kinase inhibitors and reduced upon knockdown of RAB8A. These results indicate that pathogenic LRRK2, as well as increased levels of RAB7L1, cause centrosomal deficits in a manner dependent on aberrant RAB8A phosphorylation and centrosomal/pericentrosomal accumulation, suggesting that centrosomal cohesion deficits may comprise a useful cellular readout for a broader spectrum of the disease.

11.
Mol Neurodegener ; 13(1): 3, 2018 01 23.
Article in English | MEDLINE | ID: mdl-29357897

ABSTRACT

BACKGROUND: Mutations in LRRK2 are a common genetic cause of Parkinson's disease (PD). LRRK2 interacts with and phosphorylates a subset of Rab proteins including Rab8a, a protein which has been implicated in various centrosome-related events. However, the cellular consequences of such phosphorylation remain elusive. METHODS: Human neuroblastoma SH-SY5Y cells stably expressing wildtype or pathogenic LRRK2 were used to test for polarity defects in the context of centrosomal positioning. Centrosomal cohesion deficits were analyzed from transiently transfected HEK293T cells, as well as from two distinct peripheral cell types derived from LRRK2-PD patients. Kinase assays, coimmunoprecipitation and GTP binding/retention assays were used to address Rab8a phosphorylation by LRRK2 and its effects in vitro. Transient transfections and siRNA experiments were performed to probe for the implication of Rab8a and its phosphorylated form in the centrosomal deficits caused by pathogenic LRRK2. RESULTS: Here, we show that pathogenic LRRK2 causes deficits in centrosomal positioning with effects on neurite outgrowth, cell polarization and directed migration. Pathogenic LRRK2 also causes deficits in centrosome cohesion which can be detected in peripheral cells derived from LRRK2-PD patients as compared to healthy controls, and which are reversed upon LRRK2 kinase inhibition. The centrosomal cohesion and polarity deficits can be mimicked when co-expressing wildtype LRRK2 with wildtype but not phospho-deficient Rab8a. The centrosomal defects induced by pathogenic LRRK2 are associated with a kinase activity-dependent increase in the centrosomal localization of phosphorylated Rab8a, and are prominently reduced upon RNAi of Rab8a. CONCLUSIONS: Our findings reveal a new function of LRRK2 mediated by Rab8a phosphorylation and related to various centrosomal defects.


Subject(s)
Centrosome/metabolism , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/metabolism , Parkinson Disease/genetics , Parkinson Disease/metabolism , rab GTP-Binding Proteins/metabolism , Cell Line , Humans , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/genetics , Parkinson Disease/pathology , Phosphorylation
12.
Hum Mol Genet ; 26(14): 2747-2767, 2017 07 15.
Article in English | MEDLINE | ID: mdl-28453723

ABSTRACT

Mutations in leucine-rich repeat kinase 2 (LRRK2) comprise the most common cause of familial Parkinson's disease (PD), and sequence variants modify risk for sporadic PD. Previous studies indicate that LRRK2 interacts with microtubules (MTs) and alters MT-mediated vesicular transport processes. However, the molecular determinants within LRRK2 required for such interactions have remained unknown. Here, we report that most pathogenic LRRK2 mutants cause relocalization of LRRK2 to filamentous structures which colocalize with a subset of MTs, and an identical relocalization is seen upon pharmacological LRRK2 kinase inhibition. The pronounced colocalization with MTs does not correlate with alterations in LRRK2 kinase activity, but rather with increased GTP binding. Synthetic mutations which impair GTP binding, as well as LRRK2 GTP-binding inhibitors profoundly interfere with the abnormal localization of both pathogenic mutant as well as kinase-inhibited LRRK2. Conversely, addition of a non-hydrolyzable GTP analog to permeabilized cells enhances the association of pathogenic or kinase-inhibited LRRK2 with MTs. Our data elucidate the mechanism underlying the increased MT association of select pathogenic LRRK2 mutants or of pharmacologically kinase-inhibited LRRK2, with implications for downstream MT-mediated transport events.


Subject(s)
Guanosine Triphosphate/metabolism , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/metabolism , Parkinson Disease/metabolism , GTP Phosphohydrolases/metabolism , GTP-Binding Proteins/genetics , GTP-Binding Proteins/metabolism , Genetic Variation , Guanosine Triphosphate/genetics , HEK293 Cells , Humans , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/antagonists & inhibitors , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/genetics , Microtubules/genetics , Microtubules/metabolism , Mutation , Parkinson Disease/genetics , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Signal Transduction
13.
Biochem Soc Trans ; 45(1): 141-146, 2017 02 08.
Article in English | MEDLINE | ID: mdl-28202667

ABSTRACT

Mutations in the Leucine-Rich Repeat Kinase 2 (LRRK2) gene are intimately linked to both familial and sporadic Parkinson's disease. LRRK2 is a large protein kinase able to bind and hydrolyse GTP. A wealth of in vitro studies have established that the distinct pathogenic LRRK2 mutants differentially affect those enzymatic activities, either causing an increase in kinase activity without altering GTP binding/GTP hydrolysis, or displaying no change in kinase activity but increased GTP binding/decreased GTP hydrolysis. Importantly, recent studies have shown that all pathogenic LRRK2 mutants display increased kinase activity towards select kinase substrates when analysed in intact cells. To understand those apparently discrepant results, better insight into the cellular role(s) of normal and pathogenic LRRK2 is crucial. Various studies indicate that LRRK2 regulates numerous intracellular vesicular trafficking pathways, but the mechanism(s) by which the distinct pathogenic mutants may equally interfere with such pathways has largely remained elusive. Here, we summarize the known alterations in the catalytic activities of the distinct pathogenic LRRK2 mutants and propose a testable working hypothesis by which the various mutants may affect membrane trafficking events in identical ways by culminating in increased phosphorylation of select substrate proteins known to be crucial for membrane trafficking between specific cellular compartments.


Subject(s)
GTP Phosphohydrolases/metabolism , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/metabolism , Microtubules/metabolism , Signal Transduction , Animals , Biocatalysis , Humans , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/genetics , Models, Biological , Mutation , Phosphorylation
14.
Biochem Soc Trans ; 45(1): 147-154, 2017 02 08.
Article in English | MEDLINE | ID: mdl-28202668

ABSTRACT

Leucine-rich repeat kinase 2 (LRRK2) is a key player in the pathogenesis of Parkinson's disease. Mutations in LRRK2 are associated with increased kinase activity that correlates with cytotoxicity, indicating that kinase inhibitors may comprise promising disease-modifying compounds. However, before embarking on such strategies, detailed knowledge of the cellular deficits mediated by pathogenic LRRK2 in the context of defined and pathologically relevant kinase substrates is essential. LRRK2 has been consistently shown to impair various intracellular vesicular trafficking events, and recent studies have shown that LRRK2 can phosphorylate a subset of proteins that are intricately implicated in those processes. In light of these findings, we here review the link between cellular deficits in intracellular trafficking pathways and the LRRK2-mediated phosphorylation of those newly identified substrates.


Subject(s)
Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/metabolism , Parkinson Disease/enzymology , Transport Vesicles/metabolism , rab GTP-Binding Proteins/metabolism , Amino Acid Sequence , Animals , Humans , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/genetics , Models, Biological , Mutation , Parkinson Disease/genetics , Parkinson Disease/metabolism , Phosphorylation , Sequence Homology, Amino Acid , Substrate Specificity , rab GTP-Binding Proteins/genetics
15.
Curr Protein Pept Sci ; 18(7): 677-686, 2017.
Article in English | MEDLINE | ID: mdl-26965688

ABSTRACT

Mutations in LRRK2 comprise the most common cause for familial Parkinson's disease (PD), and variations increase risk for sporadic disease, implicating LRRK2 in the entire disease spectrum. LRRK2 is a large protein harbouring both GTPase and kinase domains which display measurable catalytic activity. Most pathogenic mutations increase the kinase activity, with increased activity being cytotoxic under certain conditions. These findings have spurred great interest in drug development approaches, and various specific LRRK2 kinase inhibitors have been developed. However, LRRK2 is a largely ubiquitously expressed protein, and inhibiting its function in some non-neuronal tissues has raised safety liability issues for kinase inhibitor approaches. Therefore, understanding the cellular and cell type-specific role(s) of LRRK2 has become of paramount importance. This review will highlight current knowledge on the precise biochemical activities of normal and pathogenic LRRK2, and highlight the most common proposed cellular roles so as to gain a better understanding of the cell type-specific effects of LRRK2 modulators.


Subject(s)
Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/genetics , Mutation , Neurons/metabolism , Parkinson Disease/genetics , Aminopyridines/pharmacology , Antiparkinson Agents/pharmacology , Autophagy/drug effects , Autophagy/genetics , Benzamides/pharmacology , Endocytosis/drug effects , Gene Expression , Humans , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/antagonists & inhibitors , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/metabolism , Neurons/drug effects , Neurons/pathology , Parkinson Disease/drug therapy , Parkinson Disease/metabolism , Parkinson Disease/pathology , Protein Domains , Protein Kinase Inhibitors/pharmacology , alpha-Synuclein/genetics , alpha-Synuclein/metabolism , rab GTP-Binding Proteins/genetics , rab GTP-Binding Proteins/metabolism , rab1 GTP-Binding Proteins/genetics , rab1 GTP-Binding Proteins/metabolism , rab7 GTP-Binding Proteins
16.
Autophagy ; 12(9): 1487-506, 2016 09.
Article in English | MEDLINE | ID: mdl-27383256

ABSTRACT

Various neurodegenerative disorders are associated with increased brain iron content. Iron is known to cause oxidative stress, which concomitantly promotes cell death. Whereas endolysosomes are known to serve as intracellular iron storage organelles, the consequences of increased iron on endolysosomal functioning, and effects on cell viability upon modulation of endolysosomal iron release remain largely unknown. Here, we show that increasing intracellular iron causes endolysosomal alterations associated with impaired autophagic clearance of intracellular protein aggregates, increased cytosolic oxidative stress and increased cell death. These effects are subject to regulation by NAADP, a potent second messenger reported to target endolysosomal TPCNs (2-pore channels). Consistent with endolysosomal iron storage, cytosolic iron levels are modulated by NAADP, and increased cytosolic iron is detected when overexpressing active, but not inactive TPCNs, indicating that these channels can modulate endolysosomal iron release. Cell death triggered by altered intralysosomal iron handling is abrogated in the presence of an NAADP antagonist or when inhibiting RAB7A activity. Taken together, our results suggest that increased endolysosomal iron causes cell death associated with increased cytosolic oxidative stress as well as autophagic impairments, and these effects are subject to modulation by endolysosomal ion channel activity in a RAB7A-dependent manner. These data highlight alternative therapeutic strategies for neurodegenerative disorders associated with increased intracellular iron load.


Subject(s)
Endosomes/metabolism , Iron Overload , Lysosomes/metabolism , NADP/analogs & derivatives , rab GTP-Binding Proteins/metabolism , Animals , Apoptosis , Autophagy , Calcium/metabolism , Calcium Channels/metabolism , Calcium Signaling/physiology , Cell Line, Tumor , Cell Survival , Cytosol/metabolism , DNA/chemistry , Green Fluorescent Proteins/metabolism , HEK293 Cells , HeLa Cells , Humans , Iron/chemistry , NADP/chemistry , Neurodegenerative Diseases/metabolism , Oxidative Stress , PC12 Cells , Rats , rab7 GTP-Binding Proteins
17.
Hum Mol Genet ; 23(25): 6779-96, 2014 Dec 20.
Article in English | MEDLINE | ID: mdl-25080504

ABSTRACT

Mutations in the leucine-rich repeat kinase 2 (LRRK2) gene cause late-onset autosomal dominant Parkinson's disease (PD), and sequence variations at the LRRK2 locus are associated with increased risk for sporadic PD. LRRK2 contains both GTPase and kinase domains flanked by protein interaction motifs, and mutations associated with familial PD have been described for both catalytic domains. LRRK2 has been implicated in diverse cellular processes, and recent evidence pinpoints to an important role for LRRK2 in modulating a variety of intracellular membrane trafficking pathways. However, the underlying mechanisms are poorly understood. Here, by studying the classical, well-understood, degradative trafficking pathway of the epidermal growth factor receptor (EGFR), we show that LRRK2 regulates endocytic membrane trafficking in an Rab7-dependent manner. Mutant LRRK2 expression causes a slight delay in early-to-late endosomal trafficking, and a pronounced delay in trafficking out of late endosomes, which become aberrantly elongated into tubules. This is accompanied by a delay in EGFR degradation. The LRRK2-mediated deficits in EGFR trafficking and degradation can be reverted upon coexpression of active Rab7 and of a series of proteins involved in bridging the EGFR to Rab7 on late endosomes. Effector pulldown assays indicate that pathogenic LRRK2 decreases Rab7 activity both in cells overexpressing LRRK2, as well as in fibroblasts from pathogenic mutant LRRK2 PD patients when compared with healthy controls. Together, these findings provide novel insights into a previously unknown regulation of Rab7 activity by mutant LRRK2 which impairs membrane trafficking at very late stages of the endocytic pathway.


Subject(s)
Endosomes/metabolism , ErbB Receptors/metabolism , Parkinson Disease/metabolism , Protein Serine-Threonine Kinases/metabolism , rab GTP-Binding Proteins/metabolism , Brain/metabolism , Brain/pathology , Case-Control Studies , Endosomes/ultrastructure , ErbB Receptors/genetics , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Expression Regulation , HEK293 Cells , HeLa Cells , Humans , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 , Parkinson Disease/genetics , Parkinson Disease/pathology , Plasmids , Primary Cell Culture , Protein Serine-Threonine Kinases/genetics , Protein Structure, Tertiary , Protein Transport , Proteolysis , Signal Transduction , Transfection , rab GTP-Binding Proteins/genetics , rab7 GTP-Binding Proteins
18.
Front Mol Neurosci ; 7: 53, 2014.
Article in English | MEDLINE | ID: mdl-24987329

ABSTRACT

Parkinson's disease (PD) is a major health problem affecting millions of people worldwide. Recent studies provide compelling evidence that altered Ca(2) (+) homeostasis may underlie disease pathomechanism and be an inherent feature of all vulnerable neurons. The downstream effects of altered Ca(2) (+) handling in the distinct subcellular organelles for proper cellular function are beginning to be elucidated. Here, we summarize the evidence that vulnerable neurons may be exposed to homeostatic Ca(2) (+) stress which may determine their selective vulnerability, and suggest how abnormal Ca(2) (+) handling in the distinct intracellular compartments may compromise neuronal health in the context of aging, environmental, and genetic stress. Gaining a better understanding of the varied effects of Ca(2) (+) dyshomeostasis may allow novel combinatorial therapeutic strategies to slow PD progression.

19.
Parkinsons Dis ; 2012: 324521, 2012.
Article in English | MEDLINE | ID: mdl-23251830

ABSTRACT

Parkinson's disease is a debilitating neurodegenerative disorder, and its molecular etiopathogenesis remains poorly understood. The discovery of monogenic forms has significantly advanced our understanding of the molecular mechanisms underlying PD, as it allows generation of cellular and animal models carrying the mutant gene to define pathological pathways. Mutations in leucine-rich repeat kinase 2 (LRRK2) cause dominantly inherited PD, and variations increase risk, indicating that LRRK2 is an important player in both genetic and sporadic forms of the disease. G2019S, the most prominent pathogenic mutation, maps to the kinase domain and enhances enzymatic activity of LRRK2, which in turn seems to correlate with cytotoxicity. Since kinases are druggable targets, this has raised great hopes that disease-modifying therapies may be developed around modifying LRRK2 enzymatic activity. Apart from cytotoxicity, changes in autophagy have been consistently reported in the context of G2019S mutant LRRK2. Here, we will discuss current knowledge about mechanism(s) by which mutant LRRK2 may regulate autophagy, which highlights additional putative therapeutic targets.

20.
J Cell Sci ; 123(Pt 14): 2473-80, 2010 Jul 15.
Article in English | MEDLINE | ID: mdl-20571052

ABSTRACT

Neurosecretion involves fusion of vesicles with the plasma membrane. Such membrane fusion is mediated by the SNARE complex, which is composed of the vesicle-associated protein synaptobrevin (VAMP2), and the plasma membrane proteins syntaxin-1A and SNAP-25. Although clearly important at the point of membrane fusion, the precise structural and functional requirements for the transmembrane domains (TMDs) of SNAREs in bringing about neurosecretion remain largely unknown. Here, we used a bimolecular fluorescence complementation (BiFC) approach to study SNARE protein interactions involving TMDs in vivo. VAMP2 molecules were found to dimerise through their TMDs in intact cells. Dimerisation was abolished when replacing a glycine residue in the centre of the TMD with residues of increasing molecular volume. However, such mutations still were fully competent in bringing about membrane-fusion events, suggesting that dimerisation of the VAMP2 TMDs does not have an important functional role. By contrast, a series of deletion or insertion mutants in the C-terminal half of the TMD were largely deficient in supporting neurosecretion, whereas mutations in the N-terminal half did not display severe secretory deficits. Thus, structural length requirements, largely confined to the C-terminal half of the VAMP2 TMD, seem to be essential for SNARE-mediated membrane-fusion events in cells.


Subject(s)
Membrane Fusion , Multiprotein Complexes/metabolism , Neurosecretion , SNARE Proteins/metabolism , Vesicle-Associated Membrane Protein 2/metabolism , Amino Acid Sequence , Animals , Cell Membrane/metabolism , Membrane Fusion/genetics , Microscopy, Fluorescence , Multiprotein Complexes/genetics , Mutagenesis, Site-Directed , Mutation/genetics , Neurosecretion/genetics , PC12 Cells , Protein Multimerization/genetics , Protein Structure, Tertiary/genetics , Rats , SNARE Proteins/genetics , Synaptosomal-Associated Protein 25/metabolism , Syntaxin 1/metabolism , Vesicle-Associated Membrane Protein 2/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...