63 results on '"Alexandra Beilina"'
Search Results
2. Mutations in LRRK2 linked to Parkinson disease sequester Rab8a to damaged lysosomes and regulate transferrin-mediated iron uptake in microglia.
- Author
-
Adamantios Mamais, Jillian H Kluss, Luis Bonet-Ponce, Natalie Landeck, Rebekah G Langston, Nathan Smith, Alexandra Beilina, Alice Kaganovich, Manik C Ghosh, Laura Pellegrini, Ravindran Kumaran, Ioannis Papazoglou, George R Heaton, Rina Bandopadhyay, Nunziata Maio, Changyoun Kim, Matthew J LaVoie, David C Gershlick, and Mark R Cookson
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Mutations in leucine-rich repeat kinase 2 (LRRK2) cause autosomal dominant Parkinson disease (PD), while polymorphic LRRK2 variants are associated with sporadic PD. PD-linked mutations increase LRRK2 kinase activity and induce neurotoxicity in vitro and in vivo. The small GTPase Rab8a is a LRRK2 kinase substrate and is involved in receptor-mediated recycling and endocytic trafficking of transferrin, but the effect of PD-linked LRRK2 mutations on the function of Rab8a is poorly understood. Here, we show that gain-of-function mutations in LRRK2 induce sequestration of endogenous Rab8a to lysosomes in overexpression cell models, while pharmacological inhibition of LRRK2 kinase activity reverses this phenotype. Furthermore, we show that LRRK2 mutations drive association of endocytosed transferrin with Rab8a-positive lysosomes. LRRK2 has been nominated as an integral part of cellular responses downstream of proinflammatory signals and is activated in microglia in postmortem PD tissue. Here, we show that iPSC-derived microglia from patients carrying the most common LRRK2 mutation, G2019S, mistraffic transferrin to lysosomes proximal to the nucleus in proinflammatory conditions. Furthermore, G2019S knock-in mice show a significant increase in iron deposition in microglia following intrastriatal LPS injection compared to wild-type mice, accompanied by striatal accumulation of ferritin. Our data support a role of LRRK2 in modulating iron uptake and storage in response to proinflammatory stimuli in microglia.
- Published
- 2021
- Full Text
- View/download PDF
3. Generation of iPSC line from a Parkinson patient with PARK7 mutation and CRISPR-edited Gibco human episomal iPSC line to mimic PARK7 mutation
- Author
-
Melissa Conti Mazza, Alexandra Beilina, Dorien A. Roosen, David Hauser, and Mark R. Cookson
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Mutations in the oncogene PARK7, which codes for DJ-1, have been associated with early-onset autosomal recessive Parkinson’s disease (PD); however, the exact role of DJ-1 in PD remains elusive. Fibroblasts from a PD patient with a uniparental disomy, 1 bp deletion in PARK7 were reprogrammed into the induced pluripotent stem cell (iPSC) line: NIHTVBi015-A. For control purposes, CRISPR-Cas9 editing was used to mimic the mutation in the Gibco Human Episomal iPSC line: TMOi001-A is the control line (A18945) and TMOi001-A-3 is the control-edited line (2B10). All 3 lines exhibit normal karyotyping and expression of pluripotent markers: OCT4, SOX2, and NANOG. These lines provide a translational environment to study DJ-1-related function in PD.
- Published
- 2021
- Full Text
- View/download PDF
4. Sequential screening nominates the Parkinson's disease associated kinase LRRK2 as a regulator of Clathrin-mediated endocytosis
- Author
-
George R. Heaton, Natalie Landeck, Adamantios Mamais, Mike A. Nalls, Jonathon Nixon-Abell, Ravindran Kumaran, Alexandra Beilina, Laura Pellegrini, Yan Li, Kirsten Harvey, and Mark R. Cookson
- Subjects
Parkinson's disease ,LRRK2 ,Clathrin-mediated endocytosis ,Risk factor ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
Mutations in leucine-rich repeat kinase 2 (LRRK2) are an established cause of inherited Parkinson's disease (PD). LRRK2 is expressed in both neurons and glia in the central nervous system, but its physiological function(s) in each of these cell types is uncertain. Through sequential screens, we report a functional interaction between LRRK2 and Clathrin adaptor protein complex 2 (AP2). Analysis of LRRK2 KO tissue revealed a significant dysregulation of AP2 complex components, suggesting LRRK2 may act upstream of AP2. In line with this hypothesis, expression of LRRK2 was found to modify recruitment and phosphorylation of AP2. Furthermore, expression of LRRK2 containing the R1441C pathogenic mutation resulted in impaired clathrin-mediated endocytosis (CME). A decrease in activity-dependent synaptic vesicle endocytosis was also observed in neurons harboring an endogenous R1441C LRRK2 mutation. Alongside LRRK2, several PD-associated genes intersect with membrane-trafficking pathways. To investigate the genetic association between Clathrin-trafficking and PD, we used polygenetic risk profiling from IPDGC genome wide association studies (GWAS) datasets. Clathrin-dependent endocytosis genes were found to be associated with PD across multiple cohorts, suggesting common variants at these loci represent a cumulative risk factor for disease. Taken together, these findings suggest CME is a LRRK2-mediated, PD relevant pathway.
- Published
- 2020
- Full Text
- View/download PDF
5. The Parkinson’s Disease Protein LRRK2 Interacts with the GARP Complex to Promote Retrograde Transport to the trans-Golgi Network
- Author
-
Alexandra Beilina, Luis Bonet-Ponce, Ravindran Kumaran, Jennifer J. Kordich, Morié Ishida, Adamantios Mamais, Alice Kaganovich, Sara Saez-Atienzar, David C. Gershlick, Dorien A. Roosen, Laura Pellegrini, Vlad Malkov, Matthew J. Fell, Kirsten Harvey, Juan S. Bonifacino, Darren J. Moore, and Mark R. Cookson
- Subjects
Leucine-rich repeat kinase 2 ,neurodegeneration ,membrane trafficking ,lysosome ,endosome ,neurodegenerative disease ,Biology (General) ,QH301-705.5 - Abstract
Summary: Mutations in Leucine-rich repeat kinase 2 (LRRK2) cause Parkinson’s disease (PD). However, the precise function of LRRK2 remains unclear. We report an interaction between LRRK2 and VPS52, a subunit of the Golgi-associated retrograde protein (GARP) complex that identifies a function of LRRK2 in regulating membrane fusion at the trans-Golgi network (TGN). At the TGN, LRRK2 further interacts with the Golgi SNAREs VAMP4 and Syntaxin-6 and acts as a scaffolding platform that stabilizes the GARP-SNAREs complex formation. Therefore, LRRK2 influences both retrograde and post-Golgi trafficking pathways in a manner dependent on its GTP binding and kinase activity. This action is exaggerated by mutations associated with Parkinson’s disease and can be blocked by kinase inhibitors. Disruption of GARP sensitizes dopamine neurons to mutant LRRK2 toxicity in C. elegans, showing that these pathways are interlinked in vivo and suggesting a link in PD.
- Published
- 2020
- Full Text
- View/download PDF
6. Hexokinases link DJ-1 to the PINK1/parkin pathway
- Author
-
David N. Hauser, Adamantios Mamais, Melissa M. Conti, Christopher T. Primiani, Ravindran Kumaran, Allissa A. Dillman, Rebekah G. Langston, Alexandra Beilina, Joseph H. Garcia, Alberto Diaz-Ruiz, Michel Bernier, Fabienne C. Fiesel, Xu Hou, Wolfdieter Springer, Yan Li, Rafael de Cabo, and Mark R. Cookson
- Subjects
Proteomics ,RNA-Seq ,Metabolomics ,Systems biology ,Parkinson’s disease ,Mitophagy ,Neurology. Diseases of the nervous system ,RC346-429 ,Geriatrics ,RC952-954.6 - Abstract
Abstract Background Early onset Parkinson’s disease is caused by variants in PINK1, parkin, and DJ-1. PINK1 and parkin operate in pathways that preserve mitochondrial integrity, but the function of DJ-1 and how it relates to PINK1 and parkin is poorly understood. Methods A series of unbiased high-content screens were used to analyze changes at the protein, RNA, and metabolite level in rodent brains lacking DJ-1. Results were validated using targeted approaches, and cellular assays were performed to probe the mechanisms involved. Results We find that in both rat and mouse brains, DJ-1 knockout results in an age-dependent accumulation of hexokinase 1 in the cytosol, away from its usual location at the mitochondria, with subsequent activation of the polyol pathway of glucose metabolism in vivo. Both in the brain and in cultured cells, DJ-1 deficiency is associated with accumulation of the phosphatase PTEN that antagonizes the kinase AKT. In cells, addition of an inhibitor of AKT (MK2206) or addition of a peptide to dissociate association of hexokinases from mitochondria both inhibit the PINK1/parkin pathway, which works to maintain mitochondrial integrity. Conclusion Hexokinases are an important link between three major genetic causes of early onset Parkinson’s disease. Because aging is associated with deregulated nutrient sensing, these results help explain why DJ-1 is associated with age-dependent disease.
- Published
- 2017
- Full Text
- View/download PDF
7. Kinase activity is required for the toxic effects of mutant LRRK2/dardarin
- Author
-
Elisa Greggio, Shushant Jain, Ann Kingsbury, Rina Bandopadhyay, Patrick Lewis, Alice Kaganovich, Marcel P. van der Brug, Alexandra Beilina, Jeff Blackinton, Kelly Jean Thomas, Rili Ahmad, David W. Miller, Sashi Kesavapany, Andrew Singleton, Andrew Lees, Robert J. Harvey, Kirsten Harvey, and Mark R. Cookson
- Subjects
LRRK2 ,Kinase ,Parkinson's disease ,α-Synuclein ,Substantia nigra ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
Mutations in the LRRK2 gene, coding for dardarin, cause dominantly inherited Parkinson's disease (PD). Dardarin is a large protein, and mutations are found throughout the gene including the kinase domain. However, it is not clear if kinase activity is important for the damaging effects of pathogenic mutations. In this study, we noted two cellular phenotypes associated with mutant dardarin. First, pathogenic mutations increase the tendency of dardarin to form inclusion bodies. Secondly, neurons and neuronal cell lines undergo cell death after expression of mutant protein. Manipulating activity by replacing the kinase domain with a ‘kinase-dead’ version blocks inclusion body formation and strongly delays cell death. This predicts that kinase inhibitors will be useful therapeutic agents in patients with LRRK2 mutations and, perhaps, in sporadic PD. We also show that dardarin protein is expressed within human midbrain neurons and that C-terminal epitopes are also found in some Lewy bodies.
- Published
- 2006
- Full Text
- View/download PDF
8. Biochemical characterization of highly purified leucine-rich repeat kinases 1 and 2 demonstrates formation of homodimers.
- Author
-
Laura Civiero, Renée Vancraenenbroeck, Elisa Belluzzi, Alexandra Beilina, Evy Lobbestael, Lauran Reyniers, Fangye Gao, Ivan Micetic, Marc De Maeyer, Luigi Bubacco, Veerle Baekelandt, Mark R Cookson, Elisa Greggio, and Jean-Marc Taymans
- Subjects
Medicine ,Science - Abstract
Leucine-rich repeat kinase 1 and 2 (LRRK1 and LRRK2) are large multidomain proteins containing kinase, GTPase and multiple protein-protein interaction domains, but only mutations in LRRK2 are linked to familial Parkinson's disease (PD). Independent studies suggest that LRRK2 exists in the cell as a complex compatible with the size of a dimer. However, whether this complex is truly a homodimer or a heterologous complex formed by monomeric LRRK2 with other proteins has not been definitively proven due to the limitations in obtaining highly pure proteins suitable for structural characterization. Here, we used stable expression of LRRK1 and LRRK2 in HEK293T cell lines to produce recombinant LRRK1 and LRRK2 proteins of greater than 90% purity. Both purified LRRKs are folded, with a predominantly alpha-helical secondary structure and are capable of binding GTP with similar affinity. Furthermore, recombinant LRRK2 exhibits robust autophosphorylation activity, phosphorylation of model peptides in vitro and ATP binding. In contrast, LRRK1 does not display significant autophosphorylation activity and fails to phosphorylate LRRK2 model substrates, although it does bind ATP. Using these biochemically validated proteins, we show that LRRK1 and LRRK2 are capable of forming homodimers as shown by single-particle transmission electron microscopy and immunogold labeling. These LRRK dimers display an elongated conformation with a mean particle size of 145 Å and 175 Å respectively, which is disrupted by addition of 6M guanidinium chloride. Immunogold staining revealed double-labeled particles also in the pathological LRRK2 mutant G2019S and artificial mutants disrupting GTPase and kinase activities, suggesting that point mutations do not hinder the dimeric conformation. Overall, our findings indicate for the first time that purified and active LRRK1 and LRRK2 can form dimers in their full-length conformation.
- Published
- 2012
- Full Text
- View/download PDF
9. LRRK2 kinase activity is dependent on LRRK2 GTP binding capacity but independent of LRRK2 GTP binding.
- Author
-
Jean-Marc Taymans, Renée Vancraenenbroeck, Petri Ollikainen, Alexandra Beilina, Evy Lobbestael, Marc De Maeyer, Veerle Baekelandt, and Mark R Cookson
- Subjects
Medicine ,Science - Abstract
Leucine rich repeat kinase 2 (LRRK2) is a Parkinson's disease (PD) gene that encodes a large multidomain protein including both a GTPase and a kinase domain. GTPases often regulate kinases within signal transduction cascades, where GTPases act as molecular switches cycling between a GTP bound "on" state and a GDP bound "off" state. It has been proposed that LRRK2 kinase activity may be increased upon GTP binding at the LRRK2 Ras of complex proteins (ROC) GTPase domain. Here we extensively test this hypothesis by measuring LRRK2 phosphorylation activity under influence of GDP, GTP or non-hydrolyzable GTP analogues GTPγS or GMPPCP. We show that autophosphorylation and lrrktide phosphorylation activity of recombinant LRRK2 protein is unaltered by guanine nucleotides, when co-incubated with LRRK2 during phosphorylation reactions. Also phosphorylation activity of LRRK2 is unchanged when the LRRK2 guanine nucleotide binding pocket is previously saturated with various nucleotides, in contrast to the greatly reduced activity measured for the guanine nucleotide binding site mutant T1348N. Interestingly, when nucleotides were incubated with cell lysates prior to purification of LRRK2, kinase activity was slightly enhanced by GTPγS or GMPPCP compared to GDP, pointing to an upstream guanine nucleotide binding protein that may activate LRRK2 in a GTP-dependent manner. Using metabolic labeling, we also found that cellular phosphorylation of LRRK2 was not significantly modulated by nucleotides, although labeling is significantly reduced by guanine nucleotide binding site mutants. We conclude that while kinase activity of LRRK2 requires an intact ROC-GTPase domain, it is independent of GDP or GTP binding to ROC.
- Published
- 2011
- Full Text
- View/download PDF
10. The Parkinson's disease associated LRRK2 exhibits weaker in vitro phosphorylation of 4E-BP compared to autophosphorylation.
- Author
-
Azad Kumar, Elisa Greggio, Alexandra Beilina, Alice Kaganovich, Diane Chan, Jean-Marc Taymans, Benjamin Wolozin, and Mark R Cookson
- Subjects
Medicine ,Science - Abstract
Mutations in the gene encoding Leucine-rich repeat kinase 2 (LRRK2) are the most common cause of inherited Parkinson's disease (PD). LRRK2 is a multi-domain protein kinase containing a central catalytic core and a number of protein-protein interaction domains. An important step forward in the understanding of both the biology and the pathology of LRRK2 would be achieved by identification of its authentic physiological substrates. In the present study we examined phosphorylation of 4E-BP (eukaryotic initiation factor 4E (eIF4E)-binding protein), a recently proposed substrate for LRRKs. We found that LRRK2 is capable of phosphorylating 4E-BP in vitro. The PD related LRRK2-G2019S mutant was approximately 2 fold more active than wild type protein. However, LRRK2 autophosphorylation was stronger than 4E-BP phosphorylation under conditions of molar excess of 4E-BP to LRRK2. We also tested three other kinases (STK3, MAPK14/p38alpha and DAPK2) and found that MAPK14/p38alpha could efficiently phosphorylate 4E-BP at the same site as LRRK2 in vitro. Finally, we did not see changes in 4E-BP phosphorylation levels using inducible expression of LRRK2 in HEK cell lines. We also found that MAPK14/p38alpha phosphorylates 4E-BP in transient overexpression experiments whereas LRRK2 did not. We suggest that increased 4E-BP phosphorylation reported in some systems may be related to p38-mediated cell stress rather than direct LRRK2 activity. Overall, our results suggest that 4E-BP is a relatively poor direct substrate for LRRK2.
- Published
- 2010
- Full Text
- View/download PDF
11. Mitochondrial alterations in PINK1 deficient cells are influenced by calcineurin-dependent dephosphorylation of dynamin-related protein 1.
- Author
-
Anna Sandebring, Kelly Jean Thomas, Alexandra Beilina, Marcel van der Brug, Megan M Cleland, Rili Ahmad, David W Miller, Ibardo Zambrano, Richard F Cowburn, Homira Behbahani, Angel Cedazo-Mínguez, and Mark R Cookson
- Subjects
Medicine ,Science - Abstract
PTEN-induced novel kinase 1 (PINK1) mutations are associated with autosomal recessive parkinsonism. Previous studies have shown that PINK1 influences both mitochondrial function and morphology although it is not clearly established which of these are primary events and which are secondary. Here, we describe a novel mechanism linking mitochondrial dysfunction and alterations in mitochondrial morphology related to PINK1. Cell lines were generated by stably transducing human dopaminergic M17 cells with lentiviral constructs that increased or knocked down PINK1. As in previous studies, PINK1 deficient cells have lower mitochondrial membrane potential and are more sensitive to the toxic effects of mitochondrial complex I inhibitors. We also show that wild-type PINK1, but not recessive mutant or kinase dead versions, protects against rotenone-induced mitochondrial fragmentation whereas PINK1 deficient cells show lower mitochondrial connectivity. Expression of dynamin-related protein 1 (Drp1) exaggerates PINK1 deficiency phenotypes and Drp1 RNAi rescues them. We also show that Drp1 is dephosphorylated in PINK1 deficient cells due to activation of the calcium-dependent phosphatase calcineurin. Accordingly, the calcineurin inhibitor FK506 blocks both Drp1 dephosphorylation and loss of mitochondrial integrity in PINK1 deficient cells but does not fully rescue mitochondrial membrane potential. We propose that alterations in mitochondrial connectivity in this system are secondary to functional effects on mitochondrial membrane potential.
- Published
- 2009
- Full Text
- View/download PDF
12. Lysosomal positioning regulates Rab10 phosphorylation at LRRK2 + lysosomes
- Author
-
Jillian H. Kluss, Alexandra Beilina, Chad D. Williamson, Patrick A. Lewis, Mark R. Cookson, and Luis Bonet-Ponce
- Subjects
Multidisciplinary - Abstract
Genetic variation at the leucine-rich repeat kinase 2 ( LRRK2 ) locus contributes to an enhanced risk of familial and sporadic Parkinson’s disease. Previous data have demonstrated that recruitment to various membranes of the endolysosomal system results in LRRK2 activation. However, the mechanism(s) underlying LRRK2 activation at endolysosomal membranes and the cellular consequences of these events are still poorly understood. Here, we directed LRRK2 to lysosomes and early endosomes, triggering both LRRK2 autophosphorylation and phosphorylation of the direct LRRK2 substrates Rab10 and Rab12. However, when directed to the lysosomal membrane, pRab10 was restricted to perinuclear lysosomes, whereas pRab12 was visualized on both peripheral and perinuclear LRRK2 + lysosomes, suggesting that lysosomal positioning provides additional regulation of LRRK2-dependent Rab phosphorylation. Anterograde transport of lysosomes to the cell periphery by increasing the expression of ARL8B and SKIP or by knockdown of JIP4 blocked the recruitment and phosphorylation of Rab10 by LRRK2. The absence of pRab10 from the lysosomal membrane prevented the formation of a lysosomal tubulation and sorting process we previously named LYTL. Conversely, overexpression of RILP resulted in lysosomal clustering within the perinuclear area and increased LRRK2-dependent Rab10 recruitment and phosphorylation. The regulation of Rab10 phosphorylation in the perinuclear area depends on counteracting phosphatases, as the knockdown of phosphatase PPM1H significantly increased pRab10 signal and lysosomal tubulation in the perinuclear region. Our findings suggest that LRRK2 can be activated at multiple cellular membranes, including lysosomes, and that lysosomal positioning further provides the regulation of some Rab substrates likely via differential phosphatase activity or effector protein presence in nearby cellular compartments.
- Published
- 2022
- Full Text
- View/download PDF
13. Association of a common genetic variant with Parkinson’s disease is mediated by microglia
- Author
-
Rebekah G. Langston, Alexandra Beilina, Xylena Reed, Alice Kaganovich, Andrew B. Singleton, Cornelis Blauwendraat, J. Raphael Gibbs, and Mark R. Cookson
- Subjects
Substantia Nigra ,Humans ,Parkinson Disease ,Microglia ,General Medicine ,Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 ,Article ,Genome-Wide Association Study - Abstract
Studies of multiple neurodegenerative disorders have identified many genetic variants that are associated with risk of disease throughout a lifetime. For example, Parkinson’s disease (PD) risk is attributed in part to both coding mutations in the leucine-rich repeat kinase 2 (LRRK2) gene and to a common noncoding variation in the 5′ region of theLRRK2locus, as identified by genome-wide association studies (GWAS). However, the mechanisms linking GWAS variants to pathogenicity are largely unknown. Here, we found that the influence of PD-associated noncoding variation onLRRK2expression is specifically propagated through microglia and not by other cell types that expressLRRK2in the human brain. We find microglia-specific regulatory chromatin regions that modulate theLRRK2expression in human frontal cortex and substantia nigra and confirm these results in a human-induced pluripotent stem cell–derived microglia model. We showed, using a large-scale clustered regularly interspaced short palindromic repeats interference (CRISPRi) screen, that a regulatory DNA element containing the single-nucleotide variant rs6581593 influences theLRRK2expression in microglia. Our study demonstrates that cell type should be considered when evaluating the role of noncoding variation in disease pathogenesis and sheds light on the mechanism underlying the association of the 5′ region of LRRK2 with PD risk.
- Published
- 2022
- Full Text
- View/download PDF
14. Molecular mechanism of olesoxime-mediated neuroprotection through targeting α-synuclein interaction with mitochondrial VDAC
- Author
-
María Queralt-Martín, Mark R. Cookson, Alexandra Beilina, Amandine Rovini, William M. Rosencrans, Philip A. Gurnev, Sergey M. Bezrukov, and Tatiana K. Rostovtseva
- Subjects
Pharmacology ,0303 health sciences ,Voltage-dependent anion channel ,biology ,030302 biochemistry & molecular biology ,Chromosomal translocation ,Cell Biology ,Proximity ligation assay ,Gating ,Mitochondrion ,Neuroprotection ,Cell biology ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,chemistry.chemical_compound ,chemistry ,biology.protein ,Molecular Medicine ,Olesoxime ,Bacterial outer membrane ,Molecular Biology - Abstract
An intrinsically disordered neuronal protein α-synuclein (αSyn) is known to cause mitochondrial dysfunction, contributing to loss of dopaminergic neurons in Parkinson's disease. Through yet poorly defined mechanisms, αSyn crosses mitochondrial outer membrane and targets respiratory complexes leading to bioenergetics defects. Here, using neuronally differentiated human cells overexpressing wild-type αSyn, we show that the major metabolite channel of the outer membrane, the voltage-dependent anion channel (VDAC), is a pathway for αSyn translocation into the mitochondria. Importantly, the neuroprotective cholesterol-like synthetic compound olesoxime inhibits this translocation. By applying complementary electrophysiological and biophysical approaches, we provide mechanistic insights into the interplay between αSyn, VDAC, and olesoxime. Our data suggest that olesoxime interacts with VDAC β-barrel at the lipid-protein interface thus hindering αSyn translocation through the VDAC pore and affecting VDAC voltage gating. We propose that targeting αSyn translocation through VDAC could represent a key mechanism for the development of new neuroprotective strategies.
- Published
- 2019
- Full Text
- View/download PDF
15. Generation of iPSC line from a Parkinson patient with PARK7 mutation and CRISPR-edited Gibco human episomal iPSC line to mimic PARK7 mutation
- Author
-
Alexandra Beilina, David N. Hauser, Melissa Conti Mazza, Mark R. Cookson, and Dorien A. Roosen
- Subjects
Homeobox protein NANOG ,Mutation ,Oncogene ,QH301-705.5 ,Induced Pluripotent Stem Cells ,Protein Deglycase DJ-1 ,PARK7 ,Parkinson Disease ,Cell Biology ,General Medicine ,Biology ,Fibroblasts ,medicine.disease ,medicine.disease_cause ,Uniparental disomy ,Article ,SOX2 ,medicine ,Cancer research ,CRISPR ,Humans ,Biology (General) ,Induced pluripotent stem cell ,Developmental Biology - Abstract
Mutations in the oncogene PARK7, which codes for DJ-1, have been associated with early-onset autosomal recessive Parkinson’s disease (PD); however, the exact role of DJ-1 in PD remains elusive. Fibroblasts from a PD patient with a uniparental disomy, 1 bp deletion in PARK7 were reprogrammed into the induced pluripotent stem cell (iPSC) line: NIHTVBi015-A. For control purposes, CRISPR-Cas9 editing was used to mimic the mutation in the Gibco Human Episomal iPSC line: TMOi001-A is the control line (A18945) and TMOi001-A-3 is the control-edited line (2B10). All 3 lines exhibit normal karyotyping and expression of pluripotent markers: OCT4, SOX2, and NANOG. These lines provide a translational environment to study DJ-1-related function in PD.
- Published
- 2021
16. Combined Knockout of Lrrk2 and Rab29 Does Not Result in Behavioral Abnormalities in vivo
- Author
-
Alexandra Beilina, Christopher Bishop, Michael Coyle, Ema Karakoleva, Mark R. Cookson, Melissa Conti Mazza, Victoria Nguyen, and Jinhui Ding
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Parkinson's disease ,Biology ,Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 ,Article ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Mice ,0302 clinical medicine ,Dopamine ,Internal medicine ,medicine ,Animals ,Kinase activity ,Protein kinase A ,Mice, Knockout ,Parkinson Disease ,medicine.disease ,LRRK2 ,Corpus Striatum ,nervous system diseases ,Astrogliosis ,030104 developmental biology ,Monoamine neurotransmitter ,Endocrinology ,Knockout mouse ,Mutation ,Neurology (clinical) ,030217 neurology & neurosurgery ,medicine.drug ,Genome-Wide Association Study - Abstract
Background: Coding mutations in the LRRK2 gene, encoding for a large protein kinase, have been shown to cause familial Parkinson’s disease (PD). The immediate biological consequence of LRRK2 mutations is to increase kinase activity, suggesting that inhibition of this enzyme might be useful therapeutically to slow disease progression. Genome-wide association studies have identified the chromosomal loci around LRRK2 and one of its proposed substrates, RAB29, as contributors towards the lifetime risk of sporadic PD. Objective: Considering the evidence for interactions between LRRK2 and RAB29 on the genetic and protein levels, we set out to determine whether there are any consequences on brain function with aging after deletion of both genes. Methods: We generated a double knockout mouse model and performed a battery of motor and non-motor behavioral tests. We then investigated postmortem assays to determine the presence of PD-like pathology, including nigral dopamine cell count, astrogliosis, microgliosis, and striatal monoamine content. Results: Behaviorally, we noted only that 18–24-month Rab29-/- and double (Lrrk2-/-/Rab29-/-) knockout mice had diminished locomotor behavior in open field compared to wildtype mice. However, no genotype differences were seen in the outcomes that represented PD-like pathology. Conclusion: These results suggest that depletion of both LRRK2 and RAB29 is tolerated, at least in mice, and support that this pathway might be able to be safely targeted for therapeutics in humans.
- Published
- 2021
17. Lysosomal positioning regulates Rab10 phosphorylation at LRRK2-positive lysosomes
- Author
-
Mark R. Cookson, Luis Bonet-Ponce, Alexandra Beilina, Jillian H. Kluss, Patrick A. Lewis, and Chad D. Williamson
- Subjects
Endosome ,Chemistry ,Kinase ,Phosphatase ,Autophosphorylation ,Phosphorylation ,Signal transducing adaptor protein ,Rab ,Cellular compartment ,nervous system diseases ,Cell biology - Abstract
Genetic variation at the Leucine-rich repeat kinase 2 (LRRK2) locus contributes to enhanced lifetime risk of familial and sporadic Parkinson’s disease. Previous data have demonstrated that recruitment to various membranes of the endolysosomal system results in LRRK2 activation. However, the mechanism(s) underlying LRRK2 activation at endolysosomal membranes and the cellular consequences of these events are still poorly understood. Here, we directed LRRK2 to lysosomes and early endosomes, triggering both LRRK2 autophosphorylation and phosphorylation of the direct LRRK2 substrates Rab10 and Rab12. However, when directed to the lysosomal membrane, pRab10 was restricted to perinuclear lysosomes, whereas pRab12 was visualized on both peripheral and perinuclear LRRK2-positive lysosomes, suggesting that lysosomal positioning provides additional regulation of LRRK2-dependent Rab phosphorylation. Anterograde transport of lysosomes to the cell periphery by increasing expression of ARL8B and SKIP or by knockdown of the motor adaptor protein JIP4 blocked recruitment and phosphorylation of Rab10 by LRRK2. Conversely, overexpression of the Rab7 effector protein RILP resulted in lysosomal clustering within the perinuclear area and increased LRRK2-dependent Rab10 recruitment and phosphorylation. The regulation of Rab10 phosphorylation in the perinuclear area depends on counteracting phosphatases, as knockdown of phosphatase PPM1H significantly increased pRab10 signal and lysosomal tubulation in the perinuclear region. Our novel findings suggest LRRK2 can be activated at multiple cellular membranes including lysosomes, and that lysosomal positioning further provides regulation of some Rab substrates likely via differential phosphatase activity in nearby cellular compartments.
- Published
- 2020
- Full Text
- View/download PDF
18. LRRK2 mediates tubulation and vesicle sorting from lysosomes
- Author
-
Alexandra Beilina, Ravindran Kumaran, Christopher K. E. Bleck, Natalie Landeck, Yan Li, Jillian H. Kluss, Sara Saez-Atienzar, Eric Lindberg, Adamantios Mamais, Mark R. Cookson, Luis Bonet-Ponce, and Chad D. Williamson
- Subjects
Proteomics ,Lysosomal membrane ,Diseases and Disorders ,Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 ,03 medical and health sciences ,0302 clinical medicine ,Lysosome ,medicine ,Phosphorylation ,Research Articles ,030304 developmental biology ,LRRK2 Gene ,0303 health sciences ,Multidisciplinary ,Chemistry ,Vesicle ,SciAdv r-articles ,Signal transducing adaptor protein ,Cell Biology ,LRRK2 ,nervous system diseases ,Cell biology ,Protein Transport ,medicine.anatomical_structure ,Mutation ,Lysosomes ,030217 neurology & neurosurgery ,Research Article - Abstract
The Parkinson disease kinase LRRK2 translocates to the lysosomal membrane triggering lysosomal sorting through JIP4., Genetic variation around the LRRK2 gene affects risk of both familial and sporadic Parkinson’s disease (PD). However, the biological functions of LRRK2 remain incompletely understood. Here, we report that LRRK2 is recruited to lysosomes after exposure of cells to the lysosome membrane–rupturing agent LLOME. Using an unbiased proteomic screen, we identified the motor adaptor protein JIP4 as an LRRK2 partner at the lysosomal membrane. LRRK2 can recruit JIP4 to lysosomes in a kinase-dependent manner via the phosphorylation of RAB35 and RAB10. Using super-resolution live-cell imaging microscopy and FIB-SEM, we demonstrate that JIP4 promotes the formation of LAMP1-negative tubules that release membranous content from lysosomes. Thus, we describe a new process orchestrated by LRRK2, which we name LYTL (LYsosomal Tubulation/sorting driven by LRRK2), by which lysosomal tubulation is used to release vesicles from lysosomes. Given the central role of the lysosome in PD, LYTL is likely to be disease relevant.
- Published
- 2020
- Full Text
- View/download PDF
19. Pathogenic mutations in LRRK2 sequester Rab8a to damaged lysosomes and regulate transferrin-mediated iron uptake in microglia
- Author
-
Manik C. Ghosh, Alexandra Beilina, Luis Bonet-Ponce, Rebekah G. Langston, Natalie Landeck, Adamantios Mamais, Alice Kaganovich, Ioannis Papazoglou, Jillian H. Kluss, Nunziata Maio, Changyoun Kim, Laura Pellegrini, Ravindran Kumaran, Mark R. Cookson, David C. Gershlick, and Nathan Smith
- Subjects
chemistry.chemical_classification ,Mutation ,Microglia ,biology ,Endocytic cycle ,medicine.disease_cause ,LRRK2 ,nervous system diseases ,Cell biology ,Proinflammatory cytokine ,Ferritin ,medicine.anatomical_structure ,chemistry ,Transferrin ,medicine ,biology.protein ,Kinase activity - Abstract
Mutations in leucine-rich repeat kinase 2 (LRRK2) cause autosomal dominant Parkinson’s disease (PD) while polymorphic LRRK2 variants are associated with sporadic PD. PD-linked mutations increase LRRK2 kinase activity and induce neurotoxicity in vitro and in vivo. The small GTPase Rab8a is a LRRK2 kinase substrate and is involved in receptor-mediated recycling and endocytic trafficking of transferrin, but the effect of PD-linked LRRK2 mutations on the function of Rab8a are poorly understood. Here, we show that gain-of-function mutations in LRRK2 induce sequestration of endogenous Rab8a into lysosomes in cells while pharmacological inhibition of LRRK2 kinase activity reverses this phenotype. Furthermore, we show that LRRK2 mutations drive accumulation of endocytosed transferrin into Rab8a-positive lysosomes leading to a dysregulation of iron transport. LRRK2 has been nominated as an integral part of cellular responses downstream of proinflammatory signals and is activated in microglia in post-mortem PD tissue. Here, we show that iPSC-derived microglia from patients carrying the most common LRRK2 mutation, G2019S, mistraffic transferrin to lysosomes proximal to the nucleus in proinflammatory conditions. Furthermore, G2019S knock-in mice show significant increase in iron deposition in microglia following intrastriatal LPS injection compared to wild type mice, accompanied by striatal accumulation of ferritin. Our data support a role of LRRK2 in modulating iron uptake and storage in response to proinflammatory stimuli in microglia.
- Published
- 2020
- Full Text
- View/download PDF
20. Combined knockout of Lrrk2 and Rab29 does not result in behavioral abnormalities in vivo
- Author
-
Jinhui Ding, Alexandra Beilina, Melissa Conti Mazza, Mark R. Cookson, and Victoria Nguyen
- Subjects
medicine.medical_specialty ,Tyrosine hydroxylase ,Pars compacta ,Substantia nigra ,Striatum ,Biology ,LRRK2 ,nervous system diseases ,Endocrinology ,Dopamine ,Internal medicine ,Knockout mouse ,medicine ,Kinase activity ,medicine.drug - Abstract
Coding mutations in the LRRK2 gene, encoding for a large protein kinase, have been shown to cause familial Parkinson’s disease (PD). The immediate biological consequence of LRRK2 mutations is to increase kinase activity, leading to the suggestion that inhibition of this enzyme might be useful therapeutically to slow disease progression. Genome-wide association studies have identified the chromosomal loci around LRRK2 and one of its proposed substrates, RAB29, as contributors towards the lifetime risk of sporadic PD. Considering the evidence for interactions between LRRK2 and RAB29 on the genetic and protein levels, here we generated a double knockout mouse model and determined whether there are any consequences on brain function with aging. From a battery of motor and non-motor behavioral tests, we noted only that 18-24 month Rab29-/- and double (Lrrk2-/-/Rab29-/-) knockout mice had diminished locomotor behavior in open field compared to wildtype mice. However, no genotype differences were seen in number of substantia nigra pars compacta (SNc) dopamine neurons or in tyrosine hydroxylase levels in the SNc and striatum, which might reflect a PD-like pathology. These results suggest that depletion of both Lrrk2 and Rab29 is tolerated, at least in mice, and support that this pathway might be able to be safely targeted for therapeutics in humans.Significance statementGenetic variation in LRRK2 that result in elevated kinase activity can cause Parkinson’s disease (PD), suggesting LRRK2 inhibition as a therapeutic strategy. RAB29, a substrate of LRRK2, has also been associated with increased PD risk. Evidence exists for an interactive relationship between LRRK2 and RAB29. Mouse models lacking either LRRK2 or RAB29 do not show brain pathologies. We hypothesized that the loss of both targets would result in additive effects across in vivo and post-mortem assessments in aging mice. We found that loss of both LRRK2 and RAB29 did not result in significant behavioral deficits or dopamine neuron loss. This evidence suggests that chronic inhibition of this pathway should be tolerated clinically.
- Published
- 2020
- Full Text
- View/download PDF
21. LRRK2 phosphorylates membrane-bound Rabs and is activated by GTP-bound Rab7L1 to promote recruitment to the trans-Golgi network
- Author
-
Nicole Bryant, Andrew B. West, Alexandra Beilina, Asa Abeliovich, Ravindran Kumaran, Mark R. Cookson, and Zhiyong Liu
- Subjects
0301 basic medicine ,Biology ,Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 ,medicine.disease_cause ,03 medical and health sciences ,symbols.namesake ,0302 clinical medicine ,Genetics ,medicine ,Humans ,Phosphorylation ,Kinase activity ,Molecular Biology ,Genetics (clinical) ,Mutation ,Kinase ,Effector ,GTPase-Activating Proteins ,Autophosphorylation ,Membrane Proteins ,Articles ,General Medicine ,Golgi apparatus ,nervous system diseases ,Cell biology ,rab1 GTP-Binding Proteins ,Protein Transport ,HEK293 Cells ,030104 developmental biology ,rab GTP-Binding Proteins ,symbols ,Guanosine Triphosphate ,Rab ,030217 neurology & neurosurgery ,trans-Golgi Network - Abstract
Human genetic studies implicate LRRK2 and RAB7L1 in susceptibility to Parkinson disease (PD). These two genes function in the same pathway, as knockout of Rab7L1 results in phenotypes similar to LRRK2 knockout, and studies in cells and model organisms demonstrate LRRK2 and Rab7L1 interact in the endolysosomal system. Recently, a subset of Rab proteins have been identified as LRRK2 kinase substrates. Herein, we find that Rab8, Rab10, and Rab7L1 must be membrane and GTP-bound for LRRK2 phosphorylation. LRRK2 mutations that cause PD including R1441C, Y1699C, and G2019S all increase LRRK2 phosphorylation of Rab7L1 four-fold over wild-type LRRK2 in cells, resulting in the phosphorylation of nearly one-third the available Rab7L1 protein in cells. In contrast, the most common pathogenic LRRK2 mutation, G2019S, does not upregulate LRRK2-mediated phosphorylation of Rab8 or Rab10. LRRK2 interaction with membrane and GTP-bound Rab7L1, but not Rab8 or Rab10, results in the activation of LRRK2 autophosphorylation at the serine 1292 position, required for LRRK2 toxicity. Further, Rab7L1 controls the proportion of LRRK2 that is membrane-associated, and LRRK2 mutations enhance Rab7L1-mediated recruitment of LRRK2 to the trans-Golgi network. Interaction studies with the Rab8 and Rab10 GTPase-activating protein TBC1D4/AS160 demonstrate that LRRK2 phosphorylation may block membrane and GTP-bound Rab protein interaction with effectors. These results suggest reciprocal regulation between LRRK2 and Rab protein substrates, where Rab7L1-mediated upregulation of LRRK2 kinase activity results in the stabilization of membrane and GTP-bound Rab proteins that may be unable to interact with Rab effector proteins.
- Published
- 2017
- Full Text
- View/download PDF
22. Correction to: Molecular mechanism of olesoxime-mediated neuroprotection through targeting α-synuclein interaction with mitochondrial VDAC
- Author
-
Philip A. Gurnev, Sergey M. Bezrukov, Alexandra Beilina, Amandine Rovini, María Queralt-Martín, Tatiana K. Rostovtseva, William M. Rosencrans, and Mark R. Cookson
- Subjects
Pharmacology ,Voltage-dependent anion channel ,biology ,Cell Biology ,Neuroprotection ,Article ,Cell biology ,Cellular and Molecular Neuroscience ,chemistry.chemical_compound ,chemistry ,Molecular mechanism ,biology.protein ,Molecular Medicine ,Olesoxime ,α synuclein ,Molecular Biology - Abstract
An intrinsically disordered neuronal protein α-synuclein (αSyn) is known to cause mitochondrial dysfunction, contributing to loss of dopaminergic neurons in Parkinson’s disease. Through yet poorly defined mechanisms, αSyn crosses mitochondrial outer membrane and targets respiratory complexes leading to bioenergetics defects. Here, using neuronally differentiated human cells overexpressing wild-type αSyn, we show that the major metabolite channel of the outer membrane, the voltage-dependent anion channel (VDAC), is a pathway for αSyn translocation into the mitochondria. Importantly, the neuroprotective cholesterol-like synthetic compound olesoxime inhibits this translocation. By applying complementary electrophysiological and biophysical approaches, we provide mechanistic insights into the interplay between αSyn, VDAC, and olesoxime. Our data suggest that olesoxime interacts with VDAC β-barrel at the lipid-protein interface thus hindering αSyn translocation through the VDAC pore and affecting VDAC voltage gating. We propose that targeting αSyn translocation through VDAC could represent a key mechanism for the development of new neuroprotective strategies.
- Published
- 2020
23. Sequential screening nominates the Parkinson's disease associated kinase LRRK2 as a regulator of Clathrin-mediated endocytosis
- Author
-
Laura Pellegrini, George R. Heaton, Adamantios Mamais, Yan Li, Kirsten Harvey, Mark R. Cookson, Alexandra Beilina, Ravindran Kumaran, Mike A. Nalls, Natalie Landeck, and Jonathon Nixon-Abell
- Subjects
0301 basic medicine ,Parkinson's disease ,Adaptor Protein Complex 2 ,Genome-wide association study ,Endocytosis ,medicine.disease_cause ,Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 ,Clathrin ,Article ,lcsh:RC321-571 ,03 medical and health sciences ,Mice ,0302 clinical medicine ,medicine ,Animals ,Humans ,Phosphorylation ,lcsh:Neurosciences. Biological psychiatry. Neuropsychiatry ,030304 developmental biology ,Synaptic vesicle endocytosis ,Neurons ,0303 health sciences ,Mutation ,biology ,Kinase ,LRRK2 ,Parkinson Disease ,Receptor-mediated endocytosis ,Clathrin-mediated endocytosis ,3. Good health ,Cell biology ,nervous system diseases ,030104 developmental biology ,HEK293 Cells ,Neurology ,biology.protein ,Risk factor ,Synaptic Vesicles ,030217 neurology & neurosurgery - Abstract
Mutations in leucine-rich repeat kinase 2 (LRRK2) are an established cause of inherited Parkinson's disease (PD). LRRK2 is expressed in both neurons and glia in the central nervous system, but its physiological function(s) in each of these cell types is uncertain. Through sequential screens, we report a functional interaction between LRRK2 and Clathrin adaptor protein complex 2 (AP2). Analysis of LRRK2 KO tissue revealed a significant dysregulation of AP2 complex components, suggesting LRRK2 may act upstream of AP2. In line with this hypothesis, expression of LRRK2 was found to modify recruitment and phosphorylation of AP2. Furthermore, expression of LRRK2 containing the R1441C pathogenic mutation resulted in impaired clathrin-mediated endocytosis (CME). A decrease in activity-dependent synaptic vesicle endocytosis was also observed in neurons harboring an endogenous R1441C LRRK2 mutation. Alongside LRRK2, several PD-associated genes intersect with membrane-trafficking pathways. To investigate the genetic association between Clathrin-trafficking and PD, we used polygenetic risk profiling from IPDGC genome wide association studies (GWAS) datasets. Clathrin-dependent endocytosis genes were found to be associated with PD across multiple cohorts, suggesting common variants at these loci represent a cumulative risk factor for disease. Taken together, these findings suggest CME is a LRRK2-mediated, PD relevant pathway., Highlights • LRRK2 interacts with components of the clathrin adaptor complex AP2. • LRRK2 KO animals exhibit a significant depletion of AP2 and Clathrin. • The LRRK2-R1441C pathogenic mutation reduces endocytosis in cellular models. • Clathrin-dependent endocytosis genes are associated with Parkinson's disease.
- Published
- 2020
24. A revised 1.6 Å structure of the GTPase domain of the Parkinson’s disease-associated protein LRRK2 provides insights into mechanisms
- Author
-
Victoria A. Engel, Yangshin Park, Neo C. Hoang, Li Wan, Ruslan Sanishvili, Mu Wang, Alexandra Beilina, Mark R. Cookson, Jingling Liao, Misook Oh, Chun-Xiang Wu, Xylena Reed, Yuichiro Takagi, Steven M. Johnson, Mark Federici, Quyen Q. Hoang, and R. Jeremy Nichols
- Subjects
0303 health sciences ,Mutation ,GTP' ,Kinase ,Chemistry ,Dimer ,GDP binding ,GTPase ,medicine.disease_cause ,LRRK2 ,Cell biology ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,G-domain ,medicine ,030217 neurology & neurosurgery ,030304 developmental biology - Abstract
Leucine-rich repeat kinase 2 (LRRK2) is a large 286 kDa multi-domain protein whose mutation is a common cause of Parkinson’s disease (PD). One of the common sites of familial PD-associated mutations occurs at residue Arg-1441 in the GTPase domain of LRRK2. Previously, we reported that the PD-associated mutation R1441H impairs the catalytic activity of the GTPase domain thereby traps it in a persistently "on" state. More recently, we reported that the GTPase domain of LRRK2 exists in a dynamic dimer-monomer equilibrium where GTP binding shifts it to the monomeric conformation while GDP binding shifts it back to the dimeric state. We also reported that all of the PD-associated mutations at Arg-1441, including R1441H, R1441C, and R1441G, impair the nucleotide-dependent dimer-monomer conformational dynamics of the GTPase domain. However, the mechanism of this nucleotide-dependent conformational dynamics and how it is impaired by the mutations at residue Arg-1441 remained unclear. Here, we report a 1.6 Å crystal structure of the GTPase domain of LRRK2. Our structure has revealed a dynamic switch region that can be differentially regulated by GTP and GDP binding. This nucleotide-dependent regulation is impaired when residue Arg-1441 is substituted with the PD-associated mutations due to the loss of its exquisite interactions consisting of two hydrogen bonds and a π-stacking interaction at the dimer interface.Significance StatementMutations in LRRK2 are associated with familial Parkinson’s disease, so understanding its mechanism of actions and how they are changed by the disease-associated mutations is important for developing therapeutic strategies. This paper describes an atomic structure of the G-domain of LRRK2 revealing that the conformational dynamics of the switch regions are potentially important for its normal function. It further shows that a disease-associated mutation could lock the G domain in a persistently active-like conformation, thus perturbing its normal function.
- Published
- 2019
- Full Text
- View/download PDF
25. Molecular mechanism of olesoxime-mediated neuroprotection through targeting α-synuclein interaction with mitochondrial VDAC
- Author
-
Amandine, Rovini, Philip A, Gurnev, Alexandra, Beilina, María, Queralt-Martín, William, Rosencrans, Mark R, Cookson, Sergey M, Bezrukov, and Tatiana K, Rostovtseva
- Subjects
Membrane Potential, Mitochondrial ,Cell Survival ,Voltage-Dependent Anion Channel 1 ,Lipid Bilayers ,Apoptosis ,Protective Agents ,Article ,Mitochondria ,Protein Transport ,Cell Line, Tumor ,alpha-Synuclein ,Humans ,RNA Interference ,RNA, Small Interfering ,Reactive Oxygen Species ,Cholestenones ,Protein Binding - Abstract
An intrinsically disordered neuronal protein α-synuclein (αSyn) is known to cause mitochondrial dysfunction, contributing to loss of dopaminergic neurons in Parkinson's disease. Through yet poorly defined mechanisms, αSyn crosses mitochondrial outer membrane and targets respiratory complexes leading to bioenergetics defects. Here, using neuronally differentiated human cells overexpressing wild-type αSyn, we show that the major metabolite channel of the outer membrane, the voltage-dependent anion channel (VDAC), is a pathway for αSyn translocation into the mitochondria. Importantly, the neuroprotective cholesterol-like synthetic compound olesoxime inhibits this translocation. By applying complementary electrophysiological and biophysical approaches, we provide mechanistic insights into the interplay between αSyn, VDAC, and olesoxime. Our data suggest that olesoxime interacts with VDAC β-barrel at the lipid-protein interface thus hindering αSyn translocation through the VDAC pore and affecting VDAC voltage gating. We propose that targeting αSyn translocation through VDAC could represent a key mechanism for the development of new neuroprotective strategies.
- Published
- 2019
26. The Parkinson's Disease Protein LRRK2 Interacts with the GARP Complex to Promote Retrograde Transport to the trans-Golgi Network
- Author
-
David C. Gershlick, Morié Ishida, Dorien A. Roosen, Vlad Malkov, Luis Bonet-Ponce, Ravindran Kumaran, Matthew J. Fell, Alexandra Beilina, Alice Kaganovich, Kirsten Harvey, Jennifer J. Kordich, Juan S. Bonifacino, Sara Saez-Atienzar, Darren J. Moore, Laura Pellegrini, Adamantios Mamais, and Mark R. Cookson
- Subjects
0301 basic medicine ,Leucine-rich repeat kinase 2 ,Endosome ,Protein subunit ,Vesicular Transport Proteins ,Golgi Apparatus ,Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 ,General Biochemistry, Genetics and Molecular Biology ,Article ,03 medical and health sciences ,symbols.namesake ,Mice ,neurodegenerative disease ,0302 clinical medicine ,medicine ,Animals ,Humans ,Kinase activity ,endosome ,lcsh:QH301-705.5 ,Kinase ,Chemistry ,membrane trafficking ,Neurodegeneration ,neurodegeneration ,Membrane Proteins ,GARP complex ,Parkinson Disease ,Golgi apparatus ,medicine.disease ,LRRK2 ,Cell biology ,nervous system diseases ,Protein Transport ,030104 developmental biology ,lcsh:Biology (General) ,lysosome ,symbols ,030217 neurology & neurosurgery ,Protein Binding ,trans-Golgi Network - Abstract
SUMMARY Mutations in Leucine-rich repeat kinase 2 (LRRK2) cause Parkinson’s disease (PD). However, the precise function of LRRK2 remains unclear. We report an interaction between LRRK2 and VPS52, a subunit of the Golgi-associated retrograde protein (GARP) complex that identifies a function of LRRK2 in regulating membrane fusion at the trans-Golgi network (TGN). At the TGN, LRRK2 further interacts with the Golgi SNAREs VAMP4 and Syntaxin-6 and acts as a scaffolding platform that stabilizes the GARP-SNAREs complex formation. Therefore, LRRK2 influences both retrograde and post-Golgi trafficking pathways in a manner dependent on its GTP binding and kinase activity. This action is exaggerated by mutations associated with Parkinson’s disease and can be blocked by kinase inhibitors. Disruption of GARP sensitizes dopamine neurons to mutant LRRK2 toxicity in C. elegans, showing that these pathways are interlinked in vivo and suggesting a link in PD., Graphical Abstract, In Brief Mutations in LRRK2 are linked to Parkinson’s disease. However, the cellular role of LRRK2 remains elusive. Using a combination of proteomics and imaging techniques, Beilina et al. describe a function of LRRK2 mediating endosome-TGN transport by scaffolding the GARP:Syntaxin-6 interaction, suggesting a connection between GARP and LRRK2 in Parkinson’s disease.
- Published
- 2018
27. Leucine-rich repeat kinase 2 interacts with p21-activated kinase 6 to control neurite complexity in mammalian brain
- Author
-
Elisa Belluzzi, Chris Van den Haute, Umberto Rodella, Alexandra Beilina, Jean-Marc Taymans, Veerle Baekelandt, Evy Lobbestael, Rina Bandopadhyay, Patrick A. Lewis, Mark R. Cookson, Isabella Russo, Giovanni Piccoli, Elisa Greggio, Laura Civiero, Luigi Bubacco, Geshanthi Hondhamuni, Maria Daniela Cirnaru, and Lauran Reyniers
- Subjects
Parkinson's disease ,Protein Serine-Threonine Kinases ,Mitogen-activated protein kinase kinase ,Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 ,Biochemistry ,MAP2K7 ,LRRK2 ,neurodegeneration ,neuronal cyto-skeleton ,p21-activated kinases ,Actin Cytoskeleton ,Animals ,Brain ,Humans ,Leucine ,Mammals ,Mice ,Mutation ,Neurites ,Parkinson Disease ,Protein-Serine-Threonine Kinases ,p21-Activated Kinases ,Cellular and Molecular Neuroscience ,03 medical and health sciences ,0302 clinical medicine ,ASK1 ,Rho-associated protein kinase ,030304 developmental biology ,Molecular Basis of Disease ,0303 health sciences ,MAP kinase kinase kinase ,biology ,Cyclin-dependent kinase 2 ,nervous system diseases ,Cell biology ,biology.protein ,Original Article ,Cyclin-dependent kinase 9 ,ORIGINAL ARTICLES ,neuronal cyto‐skeleton ,p21‐activated kinases ,030217 neurology & neurosurgery - Abstract
Leucine-rich repeat kinase 2 (LRRK2) is a causative gene for Parkinson's disease, but the physiological function and the mechanism(s) by which the cellular activity of LRRK2 is regulated are poorly understood. Here, we identified p21-activated kinase 6 (PAK6) as a novel interactor of the GTPase/ROC domain of LRRK2. p21-activated kinases are serine-threonine kinases that serve as targets for the small GTP binding proteins Cdc42 and Rac1 and have been implicated in different morphogenetic processes through remodeling of the actin cytoskeleton such as synapse formation and neuritogenesis. Using an in vivo neuromorphology assay, we show that PAK6 is a positive regulator of neurite outgrowth and that LRRK2 is required for this function. Analyses of post-mortem brain tissue from idiopathic and LRRK2 G2019S carriers reveal an increase in PAK6 activation state, whereas knock-out LRRK2 mice display reduced PAK6 activation and phosphorylation of PAK6 substrates. Taken together, these results support a critical role of LRRK2 GTPase domain in cytoskeletal dynamics in vivo through the novel interactor PAK6, and provide a valuable platform to unravel the mechanism underlying LRRK2-mediated pathophysiology. We propose p21-activated kinase 6 (PAK6) as a novel interactor of leucine-rich repeat kinase 2 (LRRK2), a kinase involved in Parkinson's disease (PD). In health, PAK6 regulates neurite complexity in the brain and LRRK2 is required for its function, (a) whereas PAK6 is aberrantly activated in LRRK2-linked PD brain (b) suggesting that LRRK2 toxicity is mediated by PAK6. ispartof: Journal of Neurochemistry vol:135 issue:6 pages:1242-56 ispartof: location:England status: published
- Published
- 2015
- Full Text
- View/download PDF
28. Genes associated with Parkinson's disease: regulation of autophagy and beyond
- Author
-
Alexandra Beilina and Mark R. Cookson
- Subjects
0301 basic medicine ,Parkinson's disease ,Disease ,Mitochondrion ,Biology ,Biochemistry ,Article ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,0302 clinical medicine ,Chaperone-mediated autophagy ,Lysosome ,Mitophagy ,Autophagy ,medicine ,Animals ,Humans ,Genetic Predisposition to Disease ,Gene ,Genetics ,Parkinson Disease ,medicine.disease ,Mitochondria ,030104 developmental biology ,medicine.anatomical_structure ,Lysosomes ,Neuroscience ,030217 neurology & neurosurgery - Abstract
Substantial progress has been made in the genetic basis of Parkinson's disease (PD). In particular, by identifying genes that segregate with inherited PD or show robust association with sporadic disease, and by showing the same genes are found on both lists, we have generated an outline of the cause of this condition. Here, we will discuss what those genes tell us about the underlying biology of PD. We specifically discuss the relationships between protein products of PD genes and show that common links include regulation of the autophagy-lysosome system, an important way by which cells recycle proteins and organelles. We also discuss whether all PD genes should be considered to be in the same pathway and propose that in some cases the relationships are closer, whereas in other cases the interactions are more distant and might be considered separate. Beilina and Cookson review the links between genes for Parkinson's disease (red) and the autophagy-lysosomal system. They propose the hypothesis that many of the known PD genes can be assigned to pathways that affect (I) turnover of mitochondria via mitophagy (II) turnover of several vesicular structures via macroautophagy or chaperone-mediated autophagy or (III) general lysosome function. This article is part of a special issue on Parkinson disease.
- Published
- 2015
- Full Text
- View/download PDF
29. Comparative Protein Interaction Network Analysis Identifies Shared and Distinct Functions for the Human ROCO Proteins
- Author
-
James E, Tomkins, Sybille, Dihanich, Alexandra, Beilina, Raffaele, Ferrari, Nicolò, Ilacqua, Mark R, Cookson, Patrick A, Lewis, and Claudia, Manzoni
- Subjects
GTP-Binding Proteins ,Protein Array Analysis ,Humans ,Protein Interaction Maps ,Protein Serine-Threonine Kinases - Abstract
Signal transduction cascades governed by kinases and GTPases are a critical component of the command and control of cellular processes, with the precise outcome partly determined by direct protein-protein interactions (PPIs). Here, we use the human ROCO proteins as a model for investigating PPI signaling events-taking advantage of the unique dual kinase/GTPase activities and scaffolding properties of these multidomain proteins. PPI networks are reported that encompass the human ROCO proteins, developed using two complementary approaches. First, using the recently developed weighted PPI network analysis (WPPINA) pipeline, a confidence-weighted overview of validated ROCO protein interactors is obtained from peer-reviewed literature. Second, novel ROCO PPIs are assessed experimentally via protein microarray screens. The networks derived from these orthologous approaches are compared to identify common elements within the ROCO protein interactome; functional enrichment analysis of this common core of the network identified stress response and cell projection organization as shared functions within this protein family. Despite the presence of these commonalities, the results suggest that many unique interactors and therefore some specialized cellular roles have evolved for different members of the ROCO proteins. Overall, this multi-approach strategy to increase the resolution of protein interaction networks represents a prototype for the utility of PPI data integration in understanding signaling biology.
- Published
- 2017
30. [P4–443]: PARKINSON's DISEASE‐ASSOCIATED MUTATIONS IN LRRK2 CAUSE CENTROSOMAL DEFECTS VIA RAB8A PHOSPHORYLATION
- Author
-
Jesús Madero‐Pérez, Elena Fdez, Belén Fernández, Antonio J. Lara Ordóñez, Marian Blanca Ramírez, Patricia Gómez‐Suaga, Dieter Waschbüsch, Evy Lobbestael, Veerle Baekelandt, Angus C. Nairn, Javier Ruiz‐Martínez, Ana Aiastui, Adolfo López Munaín, Pawel Lis, Thomas Comptdaer, Jean‐Marc Taymans, Marie‐Christine Chartier‐Harlin, Alexandra Beilina, Adriano Gonnelli, Mark R. Cookson, Elisa Greggio, and Sabine Hilfiker
- Subjects
Psychiatry and Mental health ,Cellular and Molecular Neuroscience ,Developmental Neuroscience ,Epidemiology ,Health Policy ,Neurology (clinical) ,Geriatrics and Gerontology - Published
- 2017
- Full Text
- View/download PDF
31. Hexokinases link DJ-1 to the PINK1/parkin pathway
- Author
-
Melissa M. Conti, Allissa Dillman, Rebekah G. Langston, Ravindran Kumaran, Wolfdieter Springer, Yan Li, Christopher T. Primiani, Xu Hou, Alberto Diaz-Ruiz, Fabienne C. Fiesel, Adamantios Mamais, Joseph H. Garcia, Mark R. Cookson, David N. Hauser, Alexandra Beilina, Michel Bernier, and Rafael de Cabo
- Subjects
0301 basic medicine ,Proteomics ,DJ-1 ,Protein Deglycase DJ-1 ,Mitochondrion ,lcsh:Geriatrics ,Parkin ,lcsh:RC346-429 ,chemistry.chemical_compound ,Gene Knockout Techniques ,Mice ,0302 clinical medicine ,Hexokinase ,Mitophagy ,RNA-Seq ,Genetics ,Parkinson Disease ,Cell biology ,Systems biology ,Signal Transduction ,Research Article ,Ubiquitin-Protein Ligases ,PINK1 ,Nutrient sensing ,Biology ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,PTEN ,Animals ,Humans ,Metabolomics ,Rats, Long-Evans ,Molecular Biology ,Protein kinase B ,lcsh:Neurology. Diseases of the nervous system ,AKT ,Rats ,Mice, Inbred C57BL ,lcsh:RC952-954.6 ,030104 developmental biology ,chemistry ,biology.protein ,Parkinson’s disease ,Neurology (clinical) ,Protein Kinases ,030217 neurology & neurosurgery ,HeLa Cells - Abstract
Background Early onset Parkinson’s disease is caused by variants in PINK1, parkin, and DJ-1. PINK1 and parkin operate in pathways that preserve mitochondrial integrity, but the function of DJ-1 and how it relates to PINK1 and parkin is poorly understood. Methods A series of unbiased high-content screens were used to analyze changes at the protein, RNA, and metabolite level in rodent brains lacking DJ-1. Results were validated using targeted approaches, and cellular assays were performed to probe the mechanisms involved. Results We find that in both rat and mouse brains, DJ-1 knockout results in an age-dependent accumulation of hexokinase 1 in the cytosol, away from its usual location at the mitochondria, with subsequent activation of the polyol pathway of glucose metabolism in vivo. Both in the brain and in cultured cells, DJ-1 deficiency is associated with accumulation of the phosphatase PTEN that antagonizes the kinase AKT. In cells, addition of an inhibitor of AKT (MK2206) or addition of a peptide to dissociate association of hexokinases from mitochondria both inhibit the PINK1/parkin pathway, which works to maintain mitochondrial integrity. Conclusion Hexokinases are an important link between three major genetic causes of early onset Parkinson’s disease. Because aging is associated with deregulated nutrient sensing, these results help explain why DJ-1 is associated with age-dependent disease. Electronic supplementary material The online version of this article (10.1186/s13024-017-0212-x) contains supplementary material, which is available to authorized users.
- Published
- 2017
32. Unbiased screen for interactors of leucine-rich repeat kinase 2 supports a common pathway for sporadic and familial Parkinson disease
- Author
-
Mina Ryten, Daniah Trabzuni, J. Ding, Bart Post, Albert Hofman, Jean-Charles Lambert, Olaf Riess, Michele T.M. Hu, Andrew B. Singleton, Stephen Sawcer, X. Huang, Caroline H. Williams-Gray, H. R. Zielke, C Smith, Peter Lichtner, B.P.C. van de Warrenburg, Bernard Ravina, F. Durif, Ellen Sidransky, Mike A. Nalls, Karen E. Morrison, J. R. Gibbs, Robert L. Johnson, Peter Heutink, David J. Burn, Michael Bonin, Sarah Edkins, T. Gasser, Luigi Ferrucci, H. Chau, Sampath Arepalli, Chris C. A. Spencer, Yoav Ben-Shlomo, Honglei Chen, Caroline M. Tanner, Zoltán Bochdanovits, Ruth Chia, Heiko Huber, Kari Stefansson, Dena G. Hernandez, Jean-Marc Taymans, Veerle Baekelandt, Iakov N. Rudenko, Evy Lobbestael, Huw R. Morris, A. Goate, C. Moorby, Lois E. Greene, Manu Sharma, Emma Gray, Ira Shoulson, Janet Brooks, Juan C. Troncoso, K. Shaw, Laura Civiero, Alessandra Biffi, Hans Scheffer, Matthew Moore, Alan B. Zonderman, S. Sveinbjornsdottir, Avazeh Tashakkori-Ghanbaria, Jean-Christophe Corvol, Vincent Plagnol, H. Petursson, Alice Kaganovich, M M Wickremaratchi, Nigel Williams, Thomas Foltynie, Henk W. Berendse, P. Damier, A. Strange, J. M. Cooper, Simon C. Potter, Patricia Limousin, Jiali Gao, Sophie Winder-Rhodes, M. Van Der Brug, Marie Vidailhet, Elisa Greggio, Nicholas W. Wood, Kevin Talbot, M. R. Cookson, Johanna Huttenlocher, J.J. van Hilten, Dan L. Longo, Alisdair McNeill, François Tison, K.D. van Dijk, David N. Hauser, Allissa Dillman, Suneil K. Kalia, Lorraine V. Kalia, Patrick F. Chinnery, Alexis Brice, Kelechi Ndukwe, J. F. Dartigues, M. Gardner, Mohamad Saad, Palmi V. Jonsson, Kailash P. Bhatia, Roger A. Barker, André G. Uitterlinden, Maria Martinez, R. Walker, Elisa Majounie, Fernando Rivadeneira, Joel S. Perlmutter, Panagiotis Deloukas, Bryan J. Traynor, Ese E. Mudanohwo, Grisel Lopez, UM Sheerin, Joanne D. Stockton, Thomas Illig, Andres M. Lozano, Rita Guerreiro, David T. Dexter, Andrew J. Lees, Sean Chong, Gavin Hudson, Cordelia Langford, Günther Deuschl, Ravindran Kumaran, Janice L. Holton, Tamas Revesz, B.R. Bloem, Alexandra Beilina, Clare Elizabeth Harris, Daniela Berg, Anthony H.V. Schapira, Suzanne Lesage, Sean S. O'Sullivan, Albert R. Hollenbeck, James A. Pearson, R. M. A. de Bie, Delia Lorenz, Sarah E. Hunt, Richard O'Brien, Gavin Charlesworth, Maciej B. Olszewski, Stacy Steinberg, Kathrin Brockmann, Carl E Clarke, Patrizia Rizzu, Claudia Schulte, Hreinn Stefansson, Daan C. Velseboer, Omar Gustafsson, Jonathan R. Evans, Alexandra Durr, Javier Simón-Sánchez, Pierre Pollak, H. Z. Munchen, Jose Bras, Carl Counsell, John Hardy, ANS - Amsterdam Neuroscience, Neurology, and Graduate School
- Subjects
Candidate gene ,autophagy ,Blotting, Western ,Golgi Apparatus ,Genome-wide association study ,Protein Serine-Threonine Kinases ,Biology ,Leucine-rich repeat ,Cell Fractionation ,Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 ,Mass Spectrometry ,Commentaries ,Protein Interaction Mapping ,Humans ,Immunoprecipitation ,Genetic Predisposition to Disease ,Transport Vesicles ,Gene ,Adaptor Proteins, Signal Transducing ,DNA Primers ,Genetics ,Analysis of Variance ,Microscopy, Confocal ,Multidisciplinary ,Kinase ,HEK 293 cells ,BAG5 ,Intracellular Signaling Peptides and Proteins ,Brain ,rab7 GTP-Binding Proteins ,Signal transducing adaptor protein ,Parkinson Disease ,Biological Sciences ,Disorders of movement Donders Center for Medical Neuroscience [Radboudumc 3] ,GAK ,LRRK2 ,nervous system diseases ,HEK293 Cells ,Genetic Loci ,rab GTP-Binding Proteins ,Multiprotein Complexes ,trans-Golgi ,Genome-Wide Association Study ,Plasmids - Abstract
Item does not contain fulltext Mutations in leucine-rich repeat kinase 2 (LRRK2) cause inherited Parkinson disease (PD), and common variants around LRRK2 are a risk factor for sporadic PD. Using protein-protein interaction arrays, we identified BCL2-associated athanogene 5, Rab7L1 (RAB7, member RAS oncogene family-like 1), and Cyclin-G-associated kinase as binding partners of LRRK2. The latter two genes are candidate genes for risk for sporadic PD identified by genome-wide association studies. These proteins form a complex that promotes clearance of Golgi-derived vesicles through the autophagy-lysosome system both in vitro and in vivo. We propose that three different genes for PD have a common biological function. More generally, data integration from multiple unbiased screens can provide insight into human disease mechanisms.
- Published
- 2014
- Full Text
- View/download PDF
33. Age-associated changes in gene expression in human brain and isolated neurons
- Author
-
Robert Walker, Alexandra Beilina, Andrew B. Singleton, Colin Smith, Ravindran Kumaran, Mark R. Cookson, Azad Kumar, Daniah Trabzuni, Mina Ryten, Bryan J. Traynor, John Hardy, J. Raphael Gibbs, and Allissa Dillman
- Subjects
Adult ,Male ,Aging ,Adolescent ,Proteome ,Purkinje cell ,Biology ,Article ,Young Adult ,Cerebellum ,Gene expression ,medicine ,Humans ,Tissue Distribution ,Cells, Cultured ,Aged ,Laser capture microdissection ,Aged, 80 and over ,Neurons ,Regulation of gene expression ,Genetics ,General Neuroscience ,Gene Expression Regulation, Developmental ,Human brain ,Middle Aged ,Gene signature ,Frontal Lobe ,medicine.anatomical_structure ,Frontal lobe ,Cerebral cortex ,Female ,Neurology (clinical) ,Geriatrics and Gerontology ,Developmental Biology - Abstract
Previous studies have suggested that there are genes whose expression levels are associated with chronological age. However, which genes show consistent age association across studies, and which are specific to a given organism or tissue remains unresolved. Here, we reassessed this question using 2 independently ascertained series of human brain samples from 2 anatomic regions, the frontal lobe of the cerebral cortex and cerebellum. Using microarrays to estimate gene expression, we found 60 associations between expression and chronological age that were statistically significant and were replicated in both series in at least 1 tissue. There were a greater number of significant associations in the frontal cortex compared with the cerebellum. We then repeated the analysis in a subset of samples using laser capture microdissection to isolate Purkinje neurons from the cerebellum. We were able to replicate 5 gene associations from either frontal cortex or cerebellum in the Purkinje cell dataset, suggesting that there is a subset of genes which have robust changes with aging. Of these, the most consistent and strongest association was with expression of RHBDL3, a rhomboid protease family member. We confirmed several hits using an independent technique (quantitative reverse transcriptase polymerase chain reaction) and in an independent published sample series that used a different array platform. We also interrogated larger patterns of age-related gene expression using weighted gene correlation network analysis. We found several modules that showed significant associations with chronological age and, of these, several that showed negative associations were enriched for genes encoding components of mitochondria. Overall, our results show that there is a distinct and reproducible gene signature for aging in the human brain.
- Published
- 2013
- Full Text
- View/download PDF
34. DJ-1 acts in parallel to the PINK1/parkin pathway to control mitochondrial function and autophagy
- Author
-
Dragan Maric, Angel Cedazo-Minguez, Mark R. Cookson, Jeff Blackinton, Melissa K. McCoy, Marcel P. van der Brug, Anna Sandebring, David Miller, Kelly Jean Thomas, and Alexandra Beilina
- Subjects
Tumor suppressor gene ,Ubiquitin-Protein Ligases ,Protein Deglycase DJ-1 ,PINK1 ,Oxidative phosphorylation ,Biology ,Mitochondrion ,medicine.disease_cause ,Parkin ,Cell Line, Tumor ,Rotenone ,Autophagy ,Genetics ,medicine ,Humans ,Molecular Biology ,Genetics (clinical) ,Oncogene Proteins ,Intracellular Signaling Peptides and Proteins ,Parkinson Disease ,Articles ,General Medicine ,Molecular biology ,Mitochondria ,nervous system diseases ,Oxidative Stress ,mitochondrial fusion ,Mutation ,Protein Kinases ,Oxidative stress - Abstract
Mutations in DJ-1, PINK1 (PTEN-induced putative kinase 1) and parkin all cause recessive parkinsonism in humans, but the relationships between these genes are not clearly defined. One event associated with loss of any of these genes is altered mitochondrial function. Recent evidence suggests that turnover of damaged mitochondria by autophagy might be central to the process of recessive parkinsonism. Here, we show that loss of DJ-1 leads to loss of mitochondrial polarization, fragmentation of mitochondria and accumulation of markers of autophagy (LC3 punctae and lipidation) around mitochondria in human dopaminergic cells. These effects are due to endogenous oxidative stress, as antioxidants will reverse all of them. Similar to PINK1 and parkin, DJ-1 also limits mitochondrial fragmentation in response to the mitochondrial toxin rotenone. Furthermore, overexpressed parkin will protect against loss of DJ-1 and, although DJ-1 does not alter PINK1 mitochondrial phenotypes, DJ-1 is still active against rotenone-induced damage in the absence of PINK1. None of the three proteins complex together using size exclusion chromatography. These data suggest that DJ-1 works in parallel to the PINK1/parkin pathway to maintain mitochondrial function in the presence of an oxidative environment.
- Published
- 2010
- Full Text
- View/download PDF
35. Front Cover: Comparative Protein Interaction Network Analysis Identifies Shared and Distinct Functions for the Human ROCO Proteins
- Author
-
James E. Tomkins, Sybille Dihanich, Alexandra Beilina, Raffaele Ferrari, Nicolò Ilacqua, Mark R. Cookson, Patrick A. Lewis, and Claudia Manzoni
- Subjects
Molecular Biology ,Biochemistry - Published
- 2018
- Full Text
- View/download PDF
36. RNA binding activity of the recessive parkinsonism protein DJ-1 supports involvement in multiple cellular pathways
- Author
-
J. Raphael Gibbs, Nancy M. Bonini, Huaibin Cai, Myriam Gorospe, Chengsong Xie, Ashish Lal, Jeff Blackinton, Jayanth Chandran, Alexandra Beilina, Marcel P. van der Brug, Jinhui Ding, Amanda J. Myers, Ling Yang Hao, Kelly Jean Thomas, Ming Zhan, Rili Ahmad, Mark R. Cookson, Krystyna Mazan-Mamczarz, and Jennifer L. Martindale
- Subjects
MAP Kinase Signaling System ,Molecular Sequence Data ,Protein Deglycase DJ-1 ,Genes, Recessive ,RNA-binding protein ,Mitochondrion ,Biology ,Transfection ,medicine.disease_cause ,Cell Line ,Mice ,Phosphatidylinositol 3-Kinases ,Parkinsonian Disorders ,Gene expression ,medicine ,Animals ,Drosophila Proteins ,Humans ,RNA, Messenger ,RNA, Small Interfering ,Gene ,Mice, Knockout ,Oncogene Proteins ,Genetics ,Multidisciplinary ,Base Sequence ,Parkinsonism ,Intracellular Signaling Peptides and Proteins ,PTEN Phosphohydrolase ,Brain ,RNA-Binding Proteins ,RNA ,Peroxiredoxins ,Biological Sciences ,medicine.disease ,Glutathione ,Recombinant Proteins ,Cell biology ,Oxidative Stress ,Genes, Mitochondrial ,Drosophila ,Oxidative stress - Abstract
Parkinson's disease (PD) is a major neurodegenerative condition with several rare Mendelian forms. Oxidative stress and mitochondrial function have been implicated in the pathogenesis of PD but the molecular mechanisms involved in the degeneration of neurons remain unclear. DJ-1 mutations are one cause of recessive parkinsonism, but this gene is also reported to be involved in cancer by promoting Ras signaling and suppressing PTEN-induced apoptosis. The specific function of DJ-1 is unknown, although it is responsive to oxidative stress and may play a role in the maintenance of mitochondria. Here, we show, using four independent methods, that DJ-1 associates with RNA targets in cells and the brain, including mitochondrial genes, genes involved in glutathione metabolism, and members of the PTEN/PI3K cascade. Pathogenic recessive mutants are deficient in this activity. We show that DJ-1 is sufficient for RNA binding at nanomolar concentrations. Further, we show that DJ-1 binds RNA but dissociates after oxidative stress. These data implicate a single mechanism for the pleiotropic effects of DJ-1 in different model systems, namely that the protein binds multiple RNA targets in an oxidation-dependent manner.
- Published
- 2008
- Full Text
- View/download PDF
37. Structure of the ROC domain from the Parkinson's disease-associated leucine-rich repeat kinase 2 reveals a dimeric GTPase
- Author
-
Alexandra Beilina, Junpeng Deng, Eli Sluch, Elisa Greggio, Mark R. Cookson, and Patrick A. Lewis
- Subjects
Multidisciplinary ,Protein Conformation ,Kinase ,Dimer ,Parkinson Disease ,GTPase ,Biological Sciences ,Protein Serine-Threonine Kinases ,Biology ,Leucine-rich repeat ,Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 ,LRRK2 ,nervous system diseases ,GTP Phosphohydrolases ,Protein Structure, Tertiary ,chemistry.chemical_compound ,chemistry ,Biochemistry ,Domain (ring theory) ,ras Proteins ,Humans ,Kinase activity ,Protein kinase A ,Dimerization - Abstract
Mutations in leucine-rich repeat kinase 2 ( LRRK2 ) are the most common cause of Parkinson's disease (PD). LRRK2 contains a Ras of complex proteins (ROC) domain that may act as a GTPase to regulate its protein kinase activity. The structure of ROC and the mechanism(s) by which it regulates kinase activity are not known. Here, we report the crystal structure of the LRRK2 ROC domain in complex with GDP-Mg 2+ at 2.0-Å resolution. The structure displays a dimeric fold generated by extensive domain-swapping, resulting in a pair of active sites constructed with essential functional groups contributed from both monomers. Two PD-associated pathogenic residues, R1441 and I1371, are located at the interface of two monomers and provide exquisite interactions to stabilize the ROC dimer. The structure demonstrates that loss of stabilizing forces in the ROC dimer is likely related to decreased GTPase activity resulting from mutations at these sites. Our data suggest that the ROC domain may regulate LRRK2 kinase activity as a dimer, possibly via the C-terminal of ROC (COR) domain as a molecular hinge. The structure of the LRRK2 ROC domain also represents a signature from a previously undescribed class of GTPases from complex proteins and results may provide a unique molecular target for therapeutics in PD.
- Published
- 2008
- Full Text
- View/download PDF
38. The R1441C mutation of LRRK2 disrupts GTP hydrolysis
- Author
-
Alexandra Beilina, Elisa Greggio, Acacia K. Baker, Patrick A. Lewis, Mark R. Cookson, and Shushant Jain
- Subjects
GTP' ,Immunoblotting ,Biophysics ,GTPase ,Protein Serine-Threonine Kinases ,Leucine-rich repeat ,Biology ,Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 ,Guanosine Diphosphate ,Biochemistry ,Article ,GTP Phosphohydrolases ,Chlorocebus aethiops ,Animals ,Humans ,Immunoprecipitation ,Kinase activity ,Molecular Biology ,Rho-associated protein kinase ,Kinase ,Hydrolysis ,Parkinson Disease ,Cell Biology ,LRRK2 ,nervous system diseases ,Kinetics ,Amino Acid Substitution ,COS Cells ,Mutation ,Cyclin-dependent kinase 9 ,Guanosine Triphosphate ,Protein Binding - Abstract
Mutations in Leucine Rich Repeat Kinase 2 (LRRK2) are the leading genetic cause of Parkinson's disease (PD). LRRK2 is predicted to contain kinase and GTPase enzymatic domains, with recent evidence suggesting that the kinase activity of LRRK2 is central to the pathogenic process associated with this protein. The GTPase domain of LRRK2 plays an important role in the regulation of kinase activity. To investigate how the GTPase domain might be related to disease, we examined the GTP binding and hydrolysis properties of wild type and a mutant form of LRRK2. We show that LRRK2 immunoprecipitated from cells has a detectable GTPase activity that is disrupted by a familial mutation associated with PD located within the GTPase domain, R1441C.
- Published
- 2007
- Full Text
- View/download PDF
39. Kinase activity is required for the toxic effects of mutant LRRK2/dardarin
- Author
-
Alice Kaganovich, Elisa Greggio, Andrew J. Lees, Jeff Blackinton, Alexandra Beilina, Robert J. Harvey, Kirsten Harvey, Sashi Kesavapany, Rina Bandopadhyay, Andrew B. Singleton, Ann E. Kingsbury, Kelly Jean Thomas, David Miller, Patrick A. Lewis, Mark R. Cookson, Shushant Jain, Rili Ahmad, and Marcel P. van der Brug
- Subjects
Kinase ,DNA, Complementary ,Parkinson's disease ,Mutant ,Protein Serine-Threonine Kinases ,Biology ,Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 ,Inclusion bodies ,lcsh:RC321-571 ,chemistry.chemical_compound ,Mutant protein ,Substantia nigra ,Humans ,Phosphorylation ,Kinase activity ,lcsh:Neurosciences. Biological psychiatry. Neuropsychiatry ,Inclusion Bodies ,Genetics ,LRRK2 Gene ,Alpha-synuclein ,α-Synuclein ,Reverse Transcriptase Polymerase Chain Reaction ,Brain ,Parkinson Disease ,LRRK2 ,Immunohistochemistry ,Amino Acid Substitution ,Neurology ,Protein kinase domain ,chemistry ,Mutation - Abstract
Mutations in the LRRK2 gene, coding for dardarin, cause dominantly inherited Parkinson's disease (PD). Dardarin is a large protein, and mutations are found throughout the gene including the kinase domain. However, it is not clear if kinase activity is important for the damaging effects of pathogenic mutations. In this study, we noted two cellular phenotypes associated with mutant dardarin. First, pathogenic mutations increase the tendency of dardarin to form inclusion bodies. Secondly, neurons and neuronal cell lines undergo cell death after expression of mutant protein. Manipulating activity by replacing the kinase domain with a 'kinase-dead' version blocks inclusion body formation and strongly delays cell death. This predicts that kinase inhibitors will be useful therapeutic agents in patients with LRRK2 mutations and, perhaps, in sporadic PD. We also show that dardarin protein is expressed within human midbrain neurons and that C-terminal epitopes are also found in some Lewy bodies. Published by Elsevier Inc.
- Published
- 2006
40. Mutations in PTEN-induced putative kinase 1 associated with recessive parkinsonism have differential effects on protein stability
- Author
-
Alexandra Beilina, David Miller, Sashi Kesavapany, Mark R. Cookson, Marcel P. van der Brug, Gregory A. Petsko, and Rili Ahmad
- Subjects
Models, Molecular ,Recombinant Fusion Proteins ,MAP3K7 ,Cell Line ,MAP2K7 ,Parkinsonian Disorders ,Chlorocebus aethiops ,Enzyme Stability ,Animals ,Humans ,Point Mutation ,c-Raf ,Kinase activity ,Multidisciplinary ,biology ,Cyclin-dependent kinase 4 ,Cyclin-dependent kinase 2 ,Middle Aged ,Biological Sciences ,Protein kinase R ,Molecular biology ,Mitochondria ,Protein Structure, Tertiary ,COS Cells ,biology.protein ,Cyclin-dependent kinase 7 ,Protein Kinases ,Protein Processing, Post-Translational - Abstract
Several mutations in PTEN-induced putative kinase 1 ( PINK1 ) gene have been reported to be associated with recessive parkinsonism. The encoded protein is predicted to be a Ser/Thr protein kinase targeted to mitochondria. In this study, we have investigated the effects of mutations on PINK1 kinase activity in vitro and on expression levels and localization in mammalian cells. We chose to examine two point mutations: G309D, which was originally reported to be stable and properly localized in cells and L347P, which is of interest because it is present at an appreciable carrier frequency in the Philippines. We were able to confirm kinase activity and produce artificial “kinase-dead” mutants that are stable but lack activity. The L347P mutation grossly destabilizes PINK1 and drastically reduces kinase activity, whereas G309D has much more modest effects on these parameters in vitro . This finding is in line with predictions based on homology modeling. We also examined the localization of PINK1 in transfected mammalian cells by using constructs that were tagged with myc or GFP at either end of the protein. These results show that PINK1 is processed at the N terminus in a manner consistent with mitochondrial import, but the mature protein also exists in the cytosol. The physiological relevance of this observation is not yet clear, but it implies that a portion of PINK1 may be exported after processing in the mitochondria.
- Published
- 2005
- Full Text
- View/download PDF
41. Redistribution of transcription start sites within the FMR1 promoter region with expansion of the downstream CGG-repeat element
- Author
-
Flora Tassone, Paul J. Hagerman, Alexandra Beilina, Parminder Sahota, and Phillip H. Schwartz
- Subjects
Untranslated region ,congenital, hereditary, and neonatal diseases and abnormalities ,Molecular Sequence Data ,Nerve Tissue Proteins ,Biology ,Cell Line ,Fragile X Mental Retardation Protein ,Transformation, Genetic ,Transcription (biology) ,Genetics ,Humans ,RNA, Messenger ,Promoter Regions, Genetic ,Downstream Enhancer ,Molecular Biology ,Gene ,Genetics (clinical) ,DNA Primers ,Base Sequence ,RNA-Binding Proteins ,Promoter ,General Medicine ,Nucleic acid amplification technique ,FMR1 ,nervous system diseases ,Blotting, Southern ,Transcription Initiation Site ,Trinucleotide Repeat Expansion ,Trinucleotide repeat expansion ,Nucleic Acid Amplification Techniques ,Transcription Factors - Abstract
Fragile X syndrome, the most common form of mental impairment, is caused by expansion of a (CGG)n trinucleotide repeat element located in the 5' untranslated region of the fragile X mental retardation 1 (FMR1) gene. Repeat expansion is known to influence both transcription and translation; however, the mechanisms by which the CGG element exerts its effects are not known. In the current work, we have utilized 5'-RLM-RACE to examine the influence of CGG repeat number on the utilization of transcription start sites in normal (n
- Published
- 2004
- Full Text
- View/download PDF
42. Phosphorylation of LRRK2 by casein kinase 1α regulates trans-Golgi clustering via differential interaction with ARHGEF7
- Author
-
Ruth Chia, Alice Kaganovich, Sara Haddock, Adamantios Mamais, Mike A. Nalls, Iakov N. Rudenko, Yan-Yan Li, Mark R. Cookson, Ravindran Kumaran, and Alexandra Beilina
- Subjects
Kinase ,Indoles ,Primary Cell Culture ,General Physics and Astronomy ,Phloroglucinol ,Protein Serine-Threonine Kinases ,Mitogen-activated protein kinase kinase ,Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 ,Article ,General Biochemistry, Genetics and Molecular Biology ,MAP2K7 ,Mice ,Casein kinase 2, alpha 1 ,Animals ,Humans ,Phosphorylation ,RNA, Small Interfering ,Protein Kinase Inhibitors ,Cerebral Cortex ,Mice, Knockout ,Neurons ,Membrane Glycoproteins ,Multidisciplinary ,MAP kinase kinase kinase ,biology ,Cyclin-dependent kinase 2 ,Imidazoles ,Protein interactions ,Casein Kinase Ialpha ,General Chemistry ,Corpus Striatum ,nervous system diseases ,Mice, Inbred C57BL ,HEK293 Cells ,Gene Expression Regulation ,Biochemistry ,Benzamides ,Parkinson’s disease ,Golgi clustering ,biology.protein ,Cyclin-dependent kinase 9 ,Casein kinase 1 ,Casein kinase 2 ,Rho Guanine Nucleotide Exchange Factors ,Signal Transduction ,trans-Golgi Network - Abstract
LRRK2, a gene relevant to Parkinson's disease, encodes a scaffolding protein with both GTPase and kinase activities. LRRK2 protein is itself phosphorylated and therefore is subject to regulation by cell signalling; however, the kinase(s) responsible for this event have not been definitively identified. Here using an unbiased siRNA kinome screen, we identify and validate casein kinase 1α (CK1α) as being responsible for LRRK2 phosphorylation, including in the adult mouse striatum. We further show that LRRK2 recruitment to TGN46-positive Golgi-derived vesicles is modulated by constitutive LRRK2 phosphorylation by CK1α. These effects are mediated by differential protein interactions of LRRK2 with a guanine nucleotide exchange factor, ARHGEF7. These pathways are therefore likely involved in the physiological maintenance of the Golgi in cells, which may play a role in the pathogenesis of Parkinson's disease.
- Published
- 2014
- Full Text
- View/download PDF
43. Differential protein-protein interactions of LRRK1 and LRRK2 indicate roles in distinct cellular signaling pathways
- Author
-
Elisa Greggio, Ciro Iaccarino, Mark R. Cookson, Rita Derua, Yan Li, Elisa Belluzzi, Etienne Waelkens, Maria Grazia Del Giudice, Giorgio Arrigoni, Laura Civiero, Lauran Reyniers, Jean-Marc Taymans, Veerle Baekelandt, Evy Lobbestael, Claudia Crosio, and Alexandra Beilina
- Subjects
Cell signaling ,Endosome ,Kinase ,HEK 293 cells ,Protein Serine-Threonine Kinases ,Biology ,Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 ,Biochemistry ,Article ,nervous system diseases ,Protein–protein interaction ,Cell biology ,Cellular and Molecular Neuroscience ,HEK293 Cells ,Cytoplasm ,Cell Line, Tumor ,biology.protein ,Humans ,Protein Interaction Domains and Motifs ,Epidermal growth factor receptor ,Signal transduction ,Signal Transduction - Abstract
Genetic studies show that LRRK2, and not its closest paralogue LRRK1, is linked to Parkinson's disease. To gain insight into the molecular and cellular basis of this discrepancy, we searched for LRRK1- and LRRK2-specific cellular processes by identifying their distinct interacting proteins. A protein microarray-based interaction screen was performed with recombinant 3xFlag-LRRK1 and 3xFlag-LRRK2 and, in parallel, co-immunoprecipitation followed by mass spectrometry was performed from SH-SY5Y neuroblastoma cell lines stably expressing 3xFlag-LRRK1 or 3xFlag-LRRK2. We identified a set of LRRK1- and LRRK2-specific as well as common interactors. One of our most prominent findings was that both screens pointed to epidermal growth factor receptor (EGF-R) as a LRRK1-specific interactor, while 14-3-3 proteins were LRRK2-specific. This is consistent with phosphosite mapping of LRRK1, revealing phosphosites outside of 14-3-3 consensus binding motifs. To assess the functional relevance of these interactions, SH-SY5Y-LRRK1 and -LRRK2 cell lines were treated with LRRK2 kinase inhibitors that disrupt 14-3-3 binding, or with EGF, an EGF-R agonist. Redistribution of LRRK2, not LRRK1, from diffuse cytoplasmic to filamentous aggregates was observed after inhibitor treatment. Similarly, EGF induced translocation of LRRK1, but not of LRRK2, to endosomes. Our study confirms that LRRK1 and LRRK2 can carry out distinct functions by interacting with different cellular proteins. LRRK1 and LRRK2 (leucine-rich repeat kinase) interaction partners were identified by two different protein-protein interaction screens. These confirmed epidermal growth factor receptor (EGR-R) as a LRRK1-specific interactor, while 14-3-3 proteins were LRRK2-specific. Functional analysis of these interactions and the pathways they mediate shows that LRRK1 and LRRK2 signaling do not intersect, reflective of the differential role of both LRRKs in Parkinson's disease.
- Published
- 2014
44. The group II p21-activated kinases as therapautic targets in LRRK2-related Parkinson’s disease
- Author
-
Laura, Civiero, Elisa, Belluzzi, Alexandra, Beilina, Russo, Isabella, Verle, Baekelandt, Luigi, Bubacco, Mark, R Cookson, Jeanmark, Taymans, and Elisa, Greggio
- Published
- 2013
45. THE GROUP II P21-ACTIVATED KINASES AS THERAPAUTIC TARGETS IN LRRK2-RELATED PARKINSON'S DISEASE
- Author
-
Elisa, Greggio1, Laura, Civiero, Russo, Isabella, Umberto, Rodella, Elisa, Belluzzi, Alexandra, Beilina, Daniela, Cirnaru, Verle, Baekelandt, Luigi, Bubacco, Giovanni, Piccoli, Mark, R Cookson, and Jeanmark, Taymans
- Published
- 2013
46. Assays for Pten-Induced Novel Kinase 1 (PINK1) and Leucine-Rich Repeat Kinase 2 (LRRK2), Kinases Associated with Parkinson’s Disease
- Author
-
Mark R. Cookson and Alexandra Beilina
- Subjects
biology ,MAP kinase kinase kinase ,Cyclin-dependent kinase 4 ,Cyclin-dependent kinase 2 ,biology.protein ,Cancer research ,Cyclin-dependent kinase 9 ,c-Raf ,Kinase activity ,Mitogen-activated protein kinase kinase ,nervous system diseases ,MAP2K7 - Abstract
PTEN-induced novel kinase 1 (PINK1) and leucine-rich repeat kinase 2 (LRRK2) are two protein kinases associated with recessive and dominant forms of parkinsonism, respectively. Mutations in PINK1 cause loss of protein function whereas mutations in LRRK2 are less easily de fi ned but, in some cases, may cause increased kinase activity. Furthermore, LRRK2 kinase activity is being explored as a therapeutic target in PD. Therefore, in both the cases of PINK1 and LRRK2, measuring kinase activity is important but is complicated by the problem that convincing physiological substrates of the two proteins have not yet been found. In this chapter, we will describe in detail the protocols we use in our lab to measure activity and related functions of these two kinases.
- Published
- 2012
- Full Text
- View/download PDF
47. Biochemical Characterization of Highly Purified Leucine-Rich Repeat Kinases 1 and 2 Demonstrates Formation of Homodimers
- Author
-
Elisa Greggio, Alexandra Beilina, Fangye Gao, Luigi Bubacco, Laura Civiero, Mark R. Cookson, Ivan Mičetić, Marc De Maeyer, Elisa Belluzzi, Renée Vancraenenbroeck, Jean-Marc Taymans, Veerle Baekelandt, Evy Lobbestael, Lauran Reyniers, and Yue, Zhenyu
- Subjects
Proteomics ,Protein Folding ,Protein domain ,Biophysics ,lcsh:Medicine ,Plasma protein binding ,Protein Serine-Threonine Kinases ,Leucine-rich repeat ,Leucine-Rich Repeat Proteins ,Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 ,Biochemistry ,Signaling Pathways ,Adenosine Triphosphate ,Protein structure ,Microscopy, Electron, Transmission ,Neurobiology of Disease and Regeneration ,Humans ,Point Mutation ,Phosphorylation ,lcsh:Science ,Protein Interactions ,Biology ,Enzyme Kinetics ,Chromatography ,Multidisciplinary ,Chemistry ,Kinase ,Physics ,Circular Dichroism ,lcsh:R ,Lentivirus ,Autophosphorylation ,Proteins ,Immunogold labelling ,Immunohistochemistry ,Enzymes ,nervous system diseases ,HEK293 Cells ,Microscopy, Fluorescence ,Membrane protein ,Mutation ,lcsh:Q ,Molecular Neuroscience ,Dimerization ,Research Article ,Neuroscience ,Protein Binding - Abstract
Leucine-rich repeat kinase 1 and 2 (LRRK1 and LRRK2) are large multidomain proteins containing kinase, GTPase and multiple protein-protein interaction domains, but only mutations in LRRK2 are linked to familial Parkinson's disease (PD). Independent studies suggest that LRRK2 exists in the cell as a complex compatible with the size of a dimer. However, whether this complex is truly a homodimer or a heterologous complex formed by monomeric LRRK2 with other proteins has not been definitively proven due to the limitations in obtaining highly pure proteins suitable for structural characterization. Here, we used stable expression of LRRK1 and LRRK2 in HEK293T cell lines to produce recombinant LRRK1 and LRRK2 proteins of greater than 90% purity. Both purified LRRKs are folded, with a predominantly alpha-helical secondary structure and are capable of binding GTP with similar affinity. Furthermore, recombinant LRRK2 exhibits robust autophosphorylation activity, phosphorylation of model peptides in vitro and ATP binding. In contrast, LRRK1 does not display significant autophosphorylation activity and fails to phosphorylate LRRK2 model substrates, although it does bind ATP. Using these biochemically validated proteins, we show that LRRK1 and LRRK2 are capable of forming homodimers as shown by single-particle transmission electron microscopy and immunogold labeling. These LRRK dimers display an elongated conformation with a mean particle size of 145 Å and 175 Å respectively, which is disrupted by addition of 6M guanidinium chloride. Immunogold staining revealed double-labeled particles also in the pathological LRRK2 mutant G2019S and artificial mutants disrupting GTPase and kinase activities, suggesting that point mutations do not hinder the dimeric conformation. Overall, our findings indicate for the first time that purified and active LRRK1 and LRRK2 can form dimers in their full-length conformation. ispartof: PLoS One vol:7 issue:8 ispartof: location:United States status: published
- Published
- 2012
48. The Parkinson's disease associated LRRK2 exhibits weaker in vitro phosphorylation of 4E‐BP compared to autophosphorylation
- Author
-
Diane Chan, Alexandra Beilina, Azad Kumar, Ben Wolozin, Alice Kaganovich, Mark R. Cookson, and Elisa Greggio
- Subjects
medicine.medical_specialty ,Parkinson's disease ,Chemistry ,Autophosphorylation ,medicine.disease ,Biochemistry ,LRRK2 ,In vitro ,Endocrinology ,Internal medicine ,Genetics ,medicine ,Phosphorylation ,Molecular Biology ,Biotechnology - Published
- 2010
- Full Text
- View/download PDF
49. The Parkinson's disease associated LRRK2 exhibits weaker in vitro phosphorylation of 4E-BP compared to autophosphorylation
- Author
-
Elisa Greggio, Benjamin Wolozin, Alexandra Beilina, Azad Kumar, Diane Chan, Mark R. Cookson, Alice Kaganovich, and Jean-Marc Taymans
- Subjects
lcsh:Medicine ,Cell Cycle Proteins ,Protein Serine-Threonine Kinases ,Biology ,Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 ,Biochemistry ,Cell Biology/Cell Signaling ,Cell Line ,Humans ,Phosphorylation ,Protein kinase A ,lcsh:Science ,Adaptor Proteins, Signal Transducing ,MAPK14 ,Multidisciplinary ,Kinase ,EIF4E ,Autophosphorylation ,HEK 293 cells ,lcsh:R ,Parkinson Disease ,Phosphoproteins ,LRRK2 ,Molecular biology ,nervous system diseases ,lcsh:Q ,Neuroscience/Neurobiology of Disease and Regeneration ,Research Article - Abstract
Mutations in the gene encoding Leucine-rich repeat kinase 2 (LRRK2) are the most common cause of inherited Parkinson's disease (PD). LRRK2 is a multi-domain protein kinase containing a central catalytic core and a number of protein-protein interaction domains. An important step forward in the understanding of both the biology and the pathology of LRRK2 would be achieved by identification of its authentic physiological substrates. In the present study we examined phosphorylation of 4E-BP (eukaryotic initiation factor 4E (eIF4E)-binding protein), a recently proposed substrate for LRRKs. We found that LRRK2 is capable of phosphorylating 4E-BP in vitro. The PD related LRRK2-G2019S mutant was approximately 2 fold more active than wild type protein. However, LRRK2 autophosphorylation was stronger than 4E-BP phosphorylation under conditions of molar excess of 4E-BP to LRRK2. We also tested three other kinases (STK3, MAPK14/p38alpha and DAPK2) and found that MAPK14/p38alpha could efficiently phosphorylate 4E-BP at the same site as LRRK2 in vitro. Finally, we did not see changes in 4E-BP phosphorylation levels using inducible expression of LRRK2 in HEK cell lines. We also found that MAPK14/p38alpha phosphorylates 4E-BP in transient overexpression experiments whereas LRRK2 did not. We suggest that increased 4E-BP phosphorylation reported in some systems may be related to p38-mediated cell stress rather than direct LRRK2 activity. Overall, our results suggest that 4E-BP is a relatively poor direct substrate for LRRK2. ispartof: Plos One vol:15 issue:1 ispartof: location:United States status: published
- Published
- 2010
50. Mitochondrial Alterations in PINK1 Deficient Cells Are Influenced by Calcineurin-Dependent Dephosphorylation of Dynamin-Related Protein 1
- Author
-
Richard F. Cowburn, Marcel P. van der Brug, Homira Behbahani, Kelly Jean Thomas, Rili Ahmad, Ibardo Zambrano, Megan M. Cleland, Mark R. Cookson, Anna Sandebring, David Miller, Alexandra Beilina, and Angel Cedazo-Minguez
- Subjects
Dynamins ,Cell Survival ,Cell Biology/Neuronal Signaling Mechanisms ,lcsh:Medicine ,PINK1 ,Mitochondrion ,Biology ,Mitochondrial apoptosis-induced channel ,Models, Biological ,Cell Line ,GTP Phosphohydrolases ,Mitochondrial Proteins ,DNM1L ,Rotenone ,Humans ,Phosphorylation ,Inner mitochondrial membrane ,lcsh:Science ,Genetics and Genomics/Genetics of Disease ,Neurological Disorders/Movement Disorders ,Multidisciplinary ,Kinase ,Calcineurin ,lcsh:R ,Molecular biology ,Cell biology ,Mitochondria ,Enzyme Activation ,Phenotype ,Gene Knockdown Techniques ,DNAJA3 ,lcsh:Q ,ATP–ADP translocase ,Microtubule-Associated Proteins ,Protein Kinases ,Research Article - Abstract
PTEN-induced novel kinase 1 (PINK1) mutations are associated with autosomal recessive parkinsonism. Previous studies have shown that PINK1 influences both mitochondrial function and morphology although it is not clearly established which of these are primary events and which are secondary. Here, we describe a novel mechanism linking mitochondrial dysfunction and alterations in mitochondrial morphology related to PINK1. Cell lines were generated by stably transducing human dopaminergic M17 cells with lentiviral constructs that increased or knocked down PINK1. As in previous studies, PINK1 deficient cells have lower mitochondrial membrane potential and are more sensitive to the toxic effects of mitochondrial complex I inhibitors. We also show that wild-type PINK1, but not recessive mutant or kinase dead versions, protects against rotenone-induced mitochondrial fragmentation whereas PINK1 deficient cells show lower mitochondrial connectivity. Expression of dynamin-related protein 1 (Drp1) exaggerates PINK1 deficiency phenotypes and Drp1 RNAi rescues them. We also show that Drp1 is dephosphorylated in PINK1 deficient cells due to activation of the calcium-dependent phosphatase calcineurin. Accordingly, the calcineurin inhibitor FK506 blocks both Drp1 dephosphorylation and loss of mitochondrial integrity in PINK1 deficient cells but does not fully rescue mitochondrial membrane potential. We propose that alterations in mitochondrial connectivity in this system are secondary to functional effects on mitochondrial membrane potential.
- Published
- 2009
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.