Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
2.
Mov Disord ; 37(1): 190-195, 2022 01.
Article in English | MEDLINE | ID: mdl-34550621

ABSTRACT

BACKGROUND: Mutations in the GBA gene, which encodes the lysosomal enzyme glucocerebrosidase (GCase), are risk factors for Parkinson's disease (PD). OBJECTIVE: To explore the association between GCase activity, PD phenotype, and probability for prodromal PD among carriers of mutations in the GBA and LRRK2 genes. METHODS: Participants were genotyped for the G2019S-LRRK2 and nine GBA mutations common in Ashkenazi Jews. Performance-based measures enabling the calculation of the Movement Disorder Society (MDS) prodromal probability score were collected. RESULTS: One hundred and seventy PD patients (102 GBA-PD, 38 LRRK2-PD, and 30 idiopathic PD) and 221 non-manifesting carriers (NMC) (129 GBA-NMC, 45 LRRK2-NMC, 15 GBA-LRRK2-NMC, and 32 healthy controls) participated in this study. GCase activity was lower among GBA-PD (3.15 ± 0.85 µmol/L/h), GBA-NMC (3.23 ± 0.91 µmol/L/h), and GBA-LRRK2-NMC (3.20 ± 0.93 µmol/L/h) compared to the other groups of participants, with no correlation to clinical phenotype. CONCLUSIONS: Low GCase activity does not explain the clinical phenotype or risk for prodromal PD in this cohort. © 2021 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.


Subject(s)
Glucosylceramidase , Parkinson Disease , Glucosylceramidase/genetics , Heterozygote , Humans , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/genetics , Mutation/genetics , Parkinson Disease/complications
4.
Neurobiol Dis ; 105: 84-98, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28576704

ABSTRACT

Proteinaceous inclusions in neurons, composed primarily of α-synuclein, define the pathology in several neurodegenerative disorders. Neurons can internalize α-synuclein fibrils that can seed new inclusions from endogenously expressed α-synuclein. The factors contributing to the spread of pathology and subsequent neurodegeneration are not fully understood, and different compositions and concentrations of fibrils have been used in different hosts. Here, we systematically vary the concentration and length of well-characterized α-synuclein fibrils and determine their relative ability to induce inclusions and neurodegeneration in different hosts (primary neurons, C57BL/6J and C3H/HeJ mice, and Sprague Dawley rats). Using dynamic-light scattering profiles and other measurements to determine fibril length and concentration, we find that femptomolar concentrations of fibrils are sufficient to induce robust inclusions in primary neurons. However, a narrow and non-linear dynamic range characterizes fibril-mediated inclusion induction in axons and the soma. In mice, the C3H/HeJ strain is more sensitive to fibril exposures than C57BL/6J counterparts, with more inclusions and dopaminergic neurodegeneration. In rats, injection of fibrils into the substantia nigra pars compacta (SNpc) results in similar inclusion spread and dopaminergic neurodegeneration as injection of the fibrils into the dorsal striatum, with prominent inclusion spread to the amygdala and several other brain areas. Inclusion spread, particularly from the SNpc to the striatum, positively correlates with dopaminergic neurodegeneration. These results define biophysical characteristics of α-synuclein fibrils that induce inclusions and neurodegeneration both in vitro and in vivo, and suggest that inclusion spread in the brain may be promoted by a loss of neurons.


Subject(s)
Dopamine/metabolism , Inclusion Bodies/pathology , Neurodegenerative Diseases/metabolism , alpha-Synuclein/metabolism , alpha-Synuclein/toxicity , Acetylcholinesterase/metabolism , Animals , Corpus Striatum/drug effects , Corpus Striatum/pathology , Disease Models, Animal , Dose-Response Relationship, Drug , Humans , Inclusion Bodies/drug effects , Inclusion Bodies/ultrastructure , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Microscopy, Electron, Transmission , Neurodegenerative Diseases/chemically induced , Neurodegenerative Diseases/pathology , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Neurons/ultrastructure , Phosphopyruvate Hydratase/metabolism , Rats , Rats, Sprague-Dawley , Substantia Nigra/drug effects , Substantia Nigra/metabolism , Substantia Nigra/pathology , Tyrosine 3-Monooxygenase/metabolism , alpha-Synuclein/ultrastructure , tau Proteins/metabolism
5.
NPJ Parkinsons Dis ; 3: 11, 2017.
Article in English | MEDLINE | ID: mdl-28649611

ABSTRACT

Mutations associated with leucine-rich repeat kinase 2 are the most common known cause of Parkinson's disease. The known expression of leucine-rich repeat kinase 2 in immune cells and its negative regulatory function of nuclear factor of activated T cells implicates leucine-rich repeat kinase 2 in the development of the inflammatory environment characteristic of Parkinson's disease. The aim of this study was to determine the expression pattern of leucine-rich repeat kinase 2 in immune cell subsets and correlate it with the immunophenotype of cells from Parkinson's disease and healthy subjects. For immunophenotyping, blood cells from 40 Parkinson's disease patients and 32 age and environment matched-healthy control subjects were analyzed by flow cytometry. Multiplexed immunoassays were used to measure cytokine output of stimulated cells. Leucine-rich repeat kinase 2 expression was increased in B cells (p = 0.0095), T cells (p = 0.029), and CD16+ monocytes (p = 0.01) of Parkinson's disease patients compared to healthy controls. Leucine-rich repeat kinase 2 induction was also increased in monocytes and dividing T cells in Parkinson's disease patients compared to healthy controls. In addition, Parkinson's disease patient monocytes secreted more inflammatory cytokines compared to healthy control, and cytokine expression positively correlated with leucine-rich repeat kinase 2 expression in T cells from Parkinson's disease but not healthy controls. Finally, the regulatory surface protein that limits T-cell activation signals, CTLA-4 (cytotoxic T-lymphocyte-associated protein 4), was decreased in Parkinson's disease compared to HC in T cells (p = 0.029). In sum, these findings suggest that leucine-rich repeat kinase 2 has a regulatory role in immune cells and Parkinson's disease. Functionally, the positive correlations between leucine-rich repeat kinase 2 expression levels in T-cell subsets, cytokine expression and secretion, and T-cell activation states suggest that targeting leucine-rich repeat kinase 2 with therapeutic interventions could have direct effects on immune cell function.

6.
Mov Disord ; 31(10): 1543-1550, 2016 10.
Article in English | MEDLINE | ID: mdl-27297049

ABSTRACT

BACKGROUND: Mutations in Leucine-rich repeat kinase 2 (LRRK2) enhance levels of the autophosphorylated LRRK2 protein and are the most common known cause of inherited Parkinson's disease (PD). LRRK2 has been further implicated in susceptibility to idiopathic PD in genetic association studies. OBJECTIVE: The objective of this study was to compare autophosphorylated Ser(P)-1292 LRRK2 levels from biobanked urine samples with clinical data in PD patients and controls. METHODS: Ser(P)-1292 LRRK2 levels were measured from urine exosome fractions from 79 PD patients and 79 neurologically healthy controls enrolled in the Parkinson Disease Biomarker Program at the University of Alabama at Birmingham. RESULTS: Ser(P)-1292 LRRK2 levels were higher in men than women (P < .0001) and elevated in PD patients when compared with controls (P = .0014). Ser(P)-1292 LRRK2 levels were higher in PD cases with worse cognition and correlated with poor performance in MoCA (r = -0.2679 [-0.4628 to -0.0482]), MDS-UPDRS subscales 1 and 2 (r = 0.2239 [0.0014-0.4252], 0.3404 [0.1276-0.5233], respectively), Epworth Sleepiness Scale (r = 0.3215 [0.1066-0.5077]), and Modified Schwab and England Activities of Daily Living Scales (r = -0.4455 [-0.6078 to -0.2475]). Ser(P)-1292 LRRK2 levels predicted those with worse cognitive impairment in PD patients with some success (c = 0.73). CONCLUSIONS: Urinary exosome Ser(P)-1292 LRRK2 levels are elevated in idiopathic PD and correlated with the severity of cognitive impairment and difficultly in accomplishing activities of daily living. These results implicate biochemical changes in LRRK2 in idiopathic PD. © 2016 International Parkinson and Movement Disorder Society.


Subject(s)
Cognitive Dysfunction/physiopathology , Exosomes/metabolism , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/urine , Parkinson Disease/physiopathology , Parkinson Disease/urine , Severity of Illness Index , Adult , Aged , Biomarkers/urine , Cognitive Dysfunction/etiology , Female , Humans , Male , Middle Aged , Parkinson Disease/complications , Phosphorylation
7.
Neurology ; 86(11): 994-9, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26865512

ABSTRACT

OBJECTIVE: To test whether phosphorylated Ser-1292 LRRK2 levels in urine exosomes predicts LRRK2 mutation carriers (LRRK2+) and noncarriers (LRRK2-) with Parkinson disease (PD+) and without Parkinson disease (PD-). METHODS: LRRK2 protein was purified from urinary exosomes collected from participants in 2 independent cohorts. The first cohort included 14 men (LRRK2+/PD+, n = 7; LRRK2-/PD+, n = 4; LRRK2-/PD-, n = 3). The second cohort included 62 men (LRRK2-/PD-, n = 16; LRRK2+/PD-, n = 16; LRRK2+/PD+, n = 14; LRRK2-/PD+, n = 16). The ratio of Ser(P)-1292 LRRK2 to total LRRK2 was compared between LRRK2+/PD+ and LRRK2- in the first cohort and between LRRK2 G2019S carriers with and without PD in the second cohort. RESULTS: LRRK2+/PD+ had higher ratios of Ser(P)-1292 LRRK2 to total LRRK2 than LRRK2-/PD- (4.8-fold, p < 0.001) and LRRK2-/PD+ (4.6-fold, p < 0.001). Among mutation carriers, those with PD had higher Ser(P)-1292 LRRK2 to total LRRK2 than those without PD (2.2-fold, p < 0.001). Ser(P)-1292 LRRK2 levels predicted symptomatic from asymptomatic carriers with an area under the receiver operating characteristic curve of 0.844. CONCLUSION: Elevated ratio of phosphorylated Ser-1292 LRRK2 to total LRRK2 in urine exosomes predicted LRRK2 mutation status and PD risk among LRRK2 mutation carriers. Future studies may explore whether interventions that reduce this ratio may also reduce PD risk.


Subject(s)
Heterozygote , Parkinsonian Disorders/genetics , Parkinsonian Disorders/urine , Phenotype , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/urine , Aged , Animals , Biomarkers/urine , Cohort Studies , Cross-Sectional Studies , HEK293 Cells , Humans , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 , Male , Mice , Middle Aged , Parkinsonian Disorders/diagnosis , Phosphorylation/physiology , Pilot Projects
8.
Am J Respir Cell Mol Biol ; 54(3): 359-69, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26222144

ABSTRACT

Proteases are important regulators of pulmonary remodeling and airway inflammation. Recently, we have characterized the enzyme prolyl endopeptidase (PE), a serine peptidase, as a critical protease in the generation of the neutrophil chemoattractant tripeptide Pro-Gly-Pro (PGP) from collagen. However, PE has been characterized as a cytosolic enzyme, and the mechanism mediating PE release extracellularly remains unknown. We examined the role of exosomes derived from airway epithelia as a mechanism for PE release and the potential extracellular signals that regulate the release of these exosomes. We demonstrate a specific regulatory pathway of exosome release from airway epithelia and identify PE as novel exosome cargo. LPS stimulation of airway epithelial cells induces release of PE-containing exosomes, which is significantly attenuated by small interfering RNA depletion of Toll-like receptor 4 (TLR4). These differences were recapitulated upon intratracheal LPS administration in mice competent versus deficient for TLR4 signaling. Finally, sputum samples from subjects with cystic fibrosis colonized with Pseudomonas aeruginosa demonstrate elevated exosome content and increased PE levels. This TLR4-based mechanism highlights the first report of nonstochastic release of exosomes in the lung and couples TLR4 activation with matrikine generation. The increased quantity of these proteolytic exosomes in the airways of subjects with chronic lung disease highlights a new mechanism of injury and inflammation in the pathogenesis of pulmonary disorders.


Subject(s)
Bronchi/enzymology , Cystic Fibrosis/enzymology , Epithelial Cells/enzymology , Exosomes/enzymology , Mitochondrial Proteins/metabolism , Serine Endopeptidases/metabolism , Toll-Like Receptor 4/metabolism , Adult , Animals , Bronchi/drug effects , Bronchi/microbiology , Case-Control Studies , Cell Line , Cystic Fibrosis/genetics , Cystic Fibrosis/microbiology , Dose-Response Relationship, Drug , Epithelial Cells/drug effects , Epithelial Cells/microbiology , Exosomes/drug effects , Exosomes/microbiology , Female , Humans , Lipopolysaccharides/pharmacology , Male , Mice, Inbred C3H , Mice, Knockout , Prolyl Oligopeptidases , Pseudomonas aeruginosa/isolation & purification , RNA Interference , Signal Transduction , Toll-Like Receptor 4/agonists , Toll-Like Receptor 4/genetics , Transfection , Young Adult
9.
J Biol Chem ; 290(32): 19433-44, 2015 Aug 07.
Article in English | MEDLINE | ID: mdl-26078453

ABSTRACT

Therapeutic approaches to slow or block the progression of Parkinson disease (PD) do not exist. Genetic and biochemical studies implicate α-synuclein and leucine-rich repeat kinase 2 (LRRK2) in late-onset PD. LRRK2 kinase activity has been linked to neurodegenerative pathways. However, the therapeutic potential of LRRK2 kinase inhibitors is not clear because significant toxicities have been associated with one class of LRRK2 kinase inhibitors. Furthermore, LRRK2 kinase inhibitors have not been tested previously for efficacy in models of α-synuclein-induced neurodegeneration. To better understand the therapeutic potential of LRRK2 kinase inhibition in PD, we evaluated the tolerability and efficacy of a LRRK2 kinase inhibitor, PF-06447475, in preventing α-synuclein-induced neurodegeneration in rats. Both wild-type rats as well as transgenic G2019S-LRRK2 rats were injected intracranially with adeno-associated viral vectors expressing human α-synuclein in the substantia nigra. Rats were treated with PF-06447475 or a control compound for 4 weeks post-viral transduction. We found that rats expressing G2019S-LRRK2 have exacerbated dopaminergic neurodegeneration and inflammation in response to the overexpression of α-synuclein. Both neurodegeneration and neuroinflammation associated with G2019S-LRRK2 expression were mitigated by LRRK2 kinase inhibition. Furthermore, PF-06447475 provided neuroprotection in wild-type rats. We could not detect adverse pathological indications in the lung, kidney, or liver of rats treated with PF-06447475. These results demonstrate that pharmacological inhibition of LRRK2 is well tolerated for a 4-week period of time in rats and can counteract dopaminergic neurodegeneration caused by acute α-synuclein overexpression.


Subject(s)
Antiparkinson Agents/pharmacology , Parkinson Disease/drug therapy , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Pyrimidines/pharmacology , Pyrroles/pharmacology , alpha-Synuclein/genetics , Animals , Dependovirus/genetics , Disease Models, Animal , Female , Gene Expression Regulation/drug effects , Genetic Vectors , Humans , Injections, Intraventricular , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 , Male , Parkinson Disease/genetics , Parkinson Disease/metabolism , Parkinson Disease/pathology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction , Substantia Nigra/drug effects , Substantia Nigra/metabolism , Substantia Nigra/pathology , alpha-Synuclein/metabolism
10.
J Biol Chem ; 289(47): 32937-51, 2014 Nov 21.
Article in English | MEDLINE | ID: mdl-25228699

ABSTRACT

Pathogenic mutations in the LRRK2 gene can cause late-onset Parkinson disease. The most common mutation, G2019S, resides in the kinase domain and enhances activity. LRRK2 possesses the unique property of cis-autophosphorylation of its own GTPase domain. Because high-resolution structures of the human LRRK2 kinase domain are not available, we used novel high-throughput assays that measured both cis-autophosphorylation and trans-peptide phosphorylation to probe the ATP-binding pocket. We disclose hundreds of commercially available activity-selective LRRK2 kinase inhibitors. Some compounds inhibit cis-autophosphorylation more strongly than trans-peptide phosphorylation, and other compounds inhibit G2019S-LRRK2 more strongly than WT-LRRK2. Through exploitation of structure-activity relationships revealed through high-throughput analyses, we identified a useful probe inhibitor, SRI-29132 (11). SRI-29132 is exquisitely selective for LRRK2 kinase activity and is effective in attenuating proinflammatory responses in macrophages and rescuing neurite retraction phenotypes in neurons. Furthermore, the compound demonstrates excellent potency, is highly blood-brain barrier-permeant, but suffers from rapid first-pass metabolism. Despite the observed selectivity of SRI-29132, docking models highlighted critical interactions with residues conserved in many protein kinases, implying a unique structural configuration for the LRRK2 ATP-binding pocket. Although the human LRRK2 kinase domain is unstable and insoluble, we demonstrate that the LRRK2 homolog from ameba can be mutated to approximate some aspects of the human LRRK2 ATP-binding pocket. Our results provide a rich resource for LRRK2 small molecule inhibitor development. More broadly, our results provide a precedent for the functional interrogation of ATP-binding pockets when traditional approaches to ascertain structure prove difficult.


Subject(s)
Adenosine Triphosphate/chemistry , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/metabolism , Protein Structure, Tertiary , Adenosine Triphosphate/metabolism , Amino Acid Sequence , Animals , Binding Sites/genetics , Biocatalysis/drug effects , Blotting, Western , Cell Line, Tumor , Cells, Cultured , Hep G2 Cells , Humans , Kinetics , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 , Mice , Models, Molecular , Molecular Sequence Data , Molecular Structure , Mutation , Phosphorylation/drug effects , Protein Binding , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/genetics , Pyridazines/chemistry , Pyridazines/metabolism , Pyridazines/pharmacology , Sequence Homology, Amino Acid , Small Molecule Libraries/chemistry , Small Molecule Libraries/metabolism , Small Molecule Libraries/pharmacology , Structure-Activity Relationship , Triazoles/chemistry , Triazoles/metabolism , Triazoles/pharmacology
11.
J Neurosci ; 34(1): 10-21, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-24381264

ABSTRACT

Serine phosphorylation of AMPA receptor (AMPAR) subunits GluA1 and GluA2 modulates AMPAR trafficking during long-term changes in strength of hippocampal excitatory transmission required for normal learning and memory. The post-translational addition and removal of O-linked ß-N-acetylglucosamine (O-GlcNAc) also occurs on serine residues. This, together with the high expression of the enzymes O-GlcNAc transferase (OGT) and ß-N-acetylglucosamindase (O-GlcNAcase), suggests a potential role for O-GlcNAcylation in modifying synaptic efficacy and cognition. Furthermore, because key synaptic proteins are O-GlcNAcylated, this modification may be as important to brain function as phosphorylation, yet its physiological significance remains unknown. We report that acutely increasing O-GlcNAcylation in Sprague Dawley rat hippocampal slices induces an NMDA receptor and protein kinase C-independent long-term depression (LTD) at hippocampal CA3-CA1 synapses (O-GcNAc LTD). This LTD requires AMPAR GluA2 subunits, which we demonstrate are O-GlcNAcylated. Increasing O-GlcNAcylation interferes with long-term potentiation, and in hippocampal behavioral assays, it prevents novel object recognition and placement without affecting contextual fear conditioning. Our findings provide evidence that O-GlcNAcylation dynamically modulates hippocampal synaptic function and learning and memory, and suggest that altered O-GlcNAc levels could underlie cognitive dysfunction in neurological diseases.


Subject(s)
Acetylglucosaminidase/metabolism , Hippocampus/metabolism , Long-Term Synaptic Depression/physiology , N-Acetylglucosaminyltransferases/metabolism , Receptors, AMPA/metabolism , Synapses/metabolism , Acylation/physiology , Aged, 80 and over , Animals , Female , Humans , Male , Mice, 129 Strain , Mice, Mutant Strains , Organ Culture Techniques , Rats , Rats, Sprague-Dawley
12.
Hum Mol Genet ; 22(24): 4988-5000, 2013 Dec 15.
Article in English | MEDLINE | ID: mdl-23886663

ABSTRACT

Mutations in the leucine-rich repeat kinase 2 (LRRK2) gene cause late-onset Parkinson's disease (PD). Emerging evidence suggests a role for LRRK2 in the endocytic pathway. Here, we show that LRRK2 is released in extracellular microvesicles (i.e. exosomes) from cells that natively express LRRK2. LRRK2 localizes to collecting duct epithelial cells in the kidney that actively secrete exosomes into urine. Purified urinary exosomes contain LRRK2 protein that is both dimerized and phosphorylated. We provide a quantitative proteomic profile of 1673 proteins in urinary exosomes and find that known LRRK2 interactors including 14-3-3 are some of the most abundant exosome proteins. Disruption of the 14-3-3 LRRK2 interaction with a 14-3-3 inhibitor or through acute LRRK2 kinase inhibition potently blocks LRRK2 release in exosomes, but familial mutations in LRRK2 had no effect on secretion. LRRK2 levels were overall comparable but highly variable in urinary exosomes derived from PD cases and age-matched controls, although very high LRRK2 levels were detected in some PD affected cases. We further characterized LRRK2 exosome release in neurons and macrophages in culture, and found that LRRK2-positive exosomes circulate in cerebral spinal fluid (CSF). Together, these results define a pathway for LRRK2 extracellular release, clarify one function of the LRRK2 14-3-3 interaction and provide a foundation for utilization of LRRK2 as a biomarker in clinical trials.


Subject(s)
14-3-3 Proteins/metabolism , Exosomes/metabolism , Protein Serine-Threonine Kinases/metabolism , Animals , Epithelial Cells/metabolism , Humans , Kidney Tubules, Collecting/metabolism , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 , Macrophages/metabolism , Male , Mice , Mice, Knockout , Models, Biological , Mutation , Neurons/metabolism , Protein Binding , Protein Serine-Threonine Kinases/cerebrospinal fluid , Protein Serine-Threonine Kinases/genetics , Protein Transport , Rats , Rats, Transgenic
SELECTION OF CITATIONS
SEARCH DETAIL
...