Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
Acta Neuropathol ; 147(1): 67, 2024 04 06.
Article in English | MEDLINE | ID: mdl-38581586

ABSTRACT

Transcription factor EB (TFEB) is a master regulator of genes involved in the maintenance of autophagic and lysosomal homeostasis, processes which have been implicated in the pathogenesis of GBA-related and sporadic Parkinson's disease (PD), and dementia with Lewy bodies (DLB). TFEB activation results in its translocation from the cytosol to the nucleus. Here, we investigated TFEB subcellular localization and its relation to intracellular alpha-synuclein (aSyn) accumulation in post-mortem human brain of individuals with either incidental Lewy body disease (iLBD), GBA-related PD/DLB (GBA-PD/DLB) or sporadic PD/DLB (sPD/DLB), compared to control subjects. We analyzed nigral dopaminergic neurons using high-resolution confocal and stimulated emission depletion (STED) microscopy and semi-quantitatively scored the TFEB subcellular localization patterns. We observed reduced nuclear TFEB immunoreactivity in PD/DLB patients compared to controls, both in sporadic and GBA-related cases, as well as in iLBD cases. Nuclear depletion of TFEB was more pronounced in neurons with Ser129-phosphorylated (pSer129) aSyn accumulation in all groups. Importantly, we observed previously-unidentified TFEB-immunopositive perinuclear clusters in human dopaminergic neurons, which localized at the Golgi apparatus. These TFEB clusters were more frequently observed and more severe in iLBD, sPD/DLB and GBA-PD/DLB compared to controls, particularly in pSer129 aSyn-positive neurons, but also in neurons lacking detectable aSyn accumulation. In aSyn-negative cells, cytoplasmic TFEB clusters were more frequently observed in GBA-PD/DLB and iLBD patients, and correlated with reduced GBA enzymatic activity as well as increased Braak LB stage. Altered TFEB distribution was accompanied by a reduction in overall mRNA expression levels of selected TFEB-regulated genes, indicating a possible early dysfunction of lysosomal regulation. Overall, we observed cytoplasmic TFEB retention and accumulation at the Golgi in cells without apparent pSer129 aSyn accumulation in iLBD and PD/DLB patients. This suggests potential TFEB impairment at the early stages of cellular disease and underscores TFEB as a promising therapeutic target for synucleinopathies.


Subject(s)
Lewy Body Disease , Humans , alpha-Synuclein/metabolism , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Brain/pathology , Dopaminergic Neurons/metabolism , Lewy Bodies/pathology , Lewy Body Disease/pathology
2.
Cells ; 12(10)2023 05 11.
Article in English | MEDLINE | ID: mdl-37408202

ABSTRACT

Growing evidence suggests a crucial role of neuroinflammation in the pathophysiology of Parkinson's disease (PD). Neuroinflammation is linked to the accumulation and aggregation of a-synuclein (αSyn), the primary pathological hallmark of PD. Toll-like receptors 4 (TLR4) can have implications in the development and progression of the pathology. In this study, we analyzed the expression of TLR4 in the substantia nigra (SN) and medial temporal gyrus (GTM) of well-characterized PD patients and age-matched controls. We also assessed the co-localization of TLR4 with pSer129 αSyn. Using qPCR, we observed an upregulation of TLR4 expression in the SN and GTM in PD patients compared to controls, which was accompanied by a reduction in αSyn expression likely due to the depletion of dopaminergic (DA) cells. Additionally, using immunofluorescence and confocal microscopy, we observed TLR4-positive staining and co-localization with pSer129-αSyn in Lewy bodies of DA neurons in the SN, as well as in pyramidal neurons in the GTM of PD donors. Furthermore, we observed a co-localization of TLR4 and Iba-1 in glial cells of both SN and GTM. Our findings provide evidence for the increased expression of TLR4 in the PD brain and suggest that the interaction between TLR4 and pSer129-αSyn could play a role in mediating the neuroinflammatory response in PD.


Subject(s)
Parkinson Disease , Humans , alpha-Synuclein/metabolism , Lewy Bodies/metabolism , Neuroinflammatory Diseases , Parkinson Disease/metabolism , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism
3.
Commun Biol ; 3(1): 101, 2020 03 05.
Article in English | MEDLINE | ID: mdl-32139796

ABSTRACT

The molecular mechanisms underlying caudal-to-rostral progression of Lewy body pathology in Parkinson's disease remain poorly understood. Here, we identified transcriptomic signatures across brain regions involved in Braak Lewy body stages in non-neurological adults from the Allen Human Brain Atlas. Among the genes that are indicative of regional vulnerability, we found known genetic risk factors for Parkinson's disease: SCARB2, ELOVL7, SH3GL2, SNCA, BAP1, and ZNF184. Results were confirmed in two datasets of non-neurological subjects, while in two datasets of Parkinson's disease patients we found altered expression patterns. Co-expression analysis across vulnerable regions identified a module enriched for genes associated with dopamine synthesis and microglia, and another module related to the immune system, blood-oxygen transport, and endothelial cells. Both were highly expressed in regions involved in the preclinical stages of the disease. Finally, alterations in genes underlying these region-specific functions may contribute to the selective regional vulnerability in Parkinson's disease brains.


Subject(s)
Brain/pathology , Gene Expression Profiling , Lewy Bodies/genetics , Lewy Bodies/pathology , Parkinson Disease/genetics , Parkinson Disease/pathology , Transcriptome , Adolescent , Adult , Aged , Aged, 80 and over , Case-Control Studies , Databases, Genetic , Disease Progression , Female , Genetic Association Studies , Genetic Predisposition to Disease , Humans , Male , Middle Aged , Phenotype , Risk Assessment , Risk Factors , Young Adult
4.
Mol Neurobiol ; 56(2): 1344-1355, 2019 Feb.
Article in English | MEDLINE | ID: mdl-29948939

ABSTRACT

Mutations in the GBA gene, encoding the lysosomal hydrolase glucocerebrosidase (GCase), are the most common known genetic risk factor for Parkinson's disease (PD) and dementia with Lewy bodies (DLB). The present study aims to gain more insight into changes in lysosomal activity in different brain regions of sporadic PD and DLB patients, screened for GBA variants. Enzymatic activities of GCase, ß-hexosaminidase, and cathepsin D were measured in the frontal cortex, putamen, and substantia nigra (SN) of a cohort of patients with advanced PD and DLB as well as age-matched non-demented controls (n = 15/group) using fluorometric assays. Decreased activity of GCase (- 21%) and of cathepsin D (- 15%) was found in the SN and frontal cortex of patients with PD and DLB compared to controls, respectively. Population stratification was applied based on GBA genotype, showing substantially lower GCase activity (~ - 40%) in GBA variant carriers in all regions. GCase activity was further significantly decreased in the SN of PD and DLB patients without GBA variants in comparison to controls without GBA variants. Our results show decreased GCase activity in brains of PD and DLB patients with and without GBA variants, most pronounced in the SN. The results of our study confirm findings from previous studies, suggesting a role for GCase in GBA-associated as well as sporadic PD and DLB.


Subject(s)
Dementia/genetics , Lewy Bodies/metabolism , Lewy Body Disease/genetics , Lysosomes/metabolism , Parkinson Disease/genetics , Aged , Aged, 80 and over , Female , Genotype , Humans , Lewy Bodies/pathology , Male , Middle Aged , Substantia Nigra/metabolism
5.
Nat Neurosci ; 22(7): 1099-1109, 2019 07.
Article in English | MEDLINE | ID: mdl-31235907

ABSTRACT

Parkinson's disease, the most common age-related movement disorder, is a progressive neurodegenerative disease with unclear etiology. Key neuropathological hallmarks are Lewy bodies and Lewy neurites: neuronal inclusions immunopositive for the protein α-synuclein. In-depth ultrastructural analysis of Lewy pathology is crucial to understanding pathogenesis of this disease. Using correlative light and electron microscopy and tomography on postmortem human brain tissue from Parkinson's disease brain donors, we identified α-synuclein immunopositive Lewy pathology and show a crowded environment of membranes therein, including vesicular structures and dysmorphic organelles. Filaments interspersed between the membranes and organelles were identifiable in many but not all α-synuclein inclusions. Crowding of organellar components was confirmed by stimulated emission depletion (STED)-based super-resolution microscopy, and high lipid content within α-synuclein immunopositive inclusions was corroborated by confocal imaging, Fourier-transform coherent anti-Stokes Raman scattering infrared imaging and lipidomics. Applying such correlative high-resolution imaging and biophysical approaches, we discovered an aggregated protein-lipid compartmentalization not previously described in the Parkinsons' disease brain.


Subject(s)
Intracellular Membranes/ultrastructure , Lewy Bodies/ultrastructure , Lewy Body Disease/pathology , Membrane Lipids/analysis , Organelles/ultrastructure , Parkinson Disease/pathology , alpha-Synuclein/analysis , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Hippocampus/chemistry , Hippocampus/ultrastructure , Humans , Imaging, Three-Dimensional , Lewy Bodies/chemistry , Lewy Body Disease/metabolism , Mesencephalon/chemistry , Mesencephalon/ultrastructure , Microscopy, Confocal , Microscopy, Electron/methods , Microscopy, Fluorescence , Parkinson Disease/metabolism , Substantia Nigra/chemistry , Substantia Nigra/ultrastructure , Exome Sequencing
6.
Lancet Neurol ; 17(7): 597-608, 2018 07.
Article in English | MEDLINE | ID: mdl-29887161

ABSTRACT

BACKGROUND: Most patients with Parkinson's disease, Parkinson's disease dementia, and dementia with Lewy bodies do not carry mutations in known disease-causing genes. The aim of this study was to identify a novel gene implicated in the development of these disorders. METHODS: Our study was done in three stages. First, we did genome-wide linkage analysis of an Italian family with dominantly inherited Parkinson's disease to identify the disease locus. Second, we sequenced the candidate gene in an international multicentre series of unrelated probands who were diagnosed either clinically or pathologically with Parkinson's disease, Parkinson's disease dementia, or dementia with Lewy bodies. As a control, we used gene sequencing data from individuals with abdominal aortic aneurysms (who were not examined neurologically). Third, we enrolled an independent series of patients diagnosed clinically with Parkinson's disease and controls with no signs or family history of Parkinson's disease, Parkinson's disease dementia, or dementia with Lewy bodies from centres in Portugal, Sardinia, and Taiwan, and screened them for specific variants. We also did mRNA and brain pathology studies in three patients from the international multicentre series carrying disease-associated variants, and we did functional protein studies in in-vitro models, including neurons from induced pluripotent stem-like cells. FINDINGS: Molecular studies were done between Jan 1, 2008, and Dec 31, 2017. In the initial kindred of ten affected Italian individuals (mean age of disease onset 59·8 years [SD 8·7]), we detected significant linkage of Parkinson's disease to chromosome 14 and nominated LRP10 as the disease-causing gene. Among the international series of 660 probands, we identified eight individuals (four with Parkinson's disease, two with Parkinson's disease dementia, and two with dementia with Lewy bodies) who carried different, rare, potentially pathogenic LRP10 variants; one carrier was found among 645 controls with abdominal aortic aneurysms. In the independent series, two of these eight variants were detected in three additional Parkinson's disease probands (two from Sardinia and one from Taiwan) but in none of the controls. Of the 11 probands from the international and independent cohorts with LRP10 variants, ten had a positive family history of disease and DNA was available from ten affected relatives (in seven of these families). The LRP10 variants were present in nine of these ten relatives, providing independent-albeit limited-evidence of co-segregation with disease. Post-mortem studies in three patients carrying distinct LRP10 variants showed severe Lewy body pathology. Of nine variants identified in total (one in the initial family and eight in stage 2), three severely affected LRP10 expression and mRNA stability (1424+5delG, 1424+5G→A, and Ala212Serfs*17, shown by cDNA analysis), four affected protein stability (Tyr307Asn, Gly603Arg, Arg235Cys, and Pro699Ser, shown by cycloheximide-chase experiments), and two affected protein localisation (Asn517del and Arg533Leu; shown by immunocytochemistry), pointing to loss of LRP10 function as a common pathogenic mechanism. INTERPRETATION: Our findings implicate LRP10 gene defects in the development of inherited forms of α-synucleinopathies. Future elucidation of the function of the LRP10 protein and pathways could offer novel insights into mechanisms, biomarkers, and therapeutic targets. FUNDING: Stichting ParkinsonFonds, Dorpmans-Wigmans Stichting, Erasmus Medical Center, ZonMw-Memorabel programme, EU Joint Programme Neurodegenerative Disease Research (JPND), Parkinson's UK, Avtal om Läkarutbildning och Forskning (ALF) and Parkinsonfonden (Sweden), Lijf and Leven foundation, and cross-border grant of Alzheimer Netherlands-Ligue Européene Contre la Maladie d'Alzheimer (LECMA).


Subject(s)
LDL-Receptor Related Proteins/genetics , Lewy Body Disease/genetics , Parkinson Disease/genetics , Brain/pathology , Chromosomes, Human, Pair 14/genetics , Dementia/epidemiology , Dementia/etiology , Dementia/genetics , Family , Female , Genetic Linkage , Genome-Wide Association Study , Heterozygote , Humans , Italy , Lewy Body Disease/epidemiology , Male , Middle Aged , Parkinson Disease/complications , Parkinson Disease/epidemiology , Pedigree , Pluripotent Stem Cells/metabolism , RNA, Messenger/chemistry , RNA, Messenger/genetics
7.
Mol Neurodegener ; 12(1): 11, 2017 01 25.
Article in English | MEDLINE | ID: mdl-28122627

ABSTRACT

Converging evidence from genetic, pathological and experimental studies have increasingly suggested an important role for autophagy impairment in Parkinson's Disease (PD). Genetic studies have identified mutations in genes encoding for components of the autophagy-lysosomal pathway (ALP), including glucosidase beta acid 1 (GBA1), that are associated with increased risk for developing PD. Observations in PD brain tissue suggest an aberrant regulation of autophagy associated with the aggregation of α-synuclein (α-syn). As autophagy is one of the main systems involved in the proteolytic degradation of α-syn, pharmacological enhancement of autophagy may be an attractive strategy to combat α-syn aggregation in PD. Here, we review the potential of autophagy enhancement as disease-modifying therapy in PD based on preclinical evidence. In particular, we provide an overview of the molecular regulation of autophagy and targets for pharmacological modulation within the ALP. In experimental models, beneficial effects on multiple pathological processes involved in PD, including α-syn aggregation, cell death, oxidative stress and mitochondrial dysfunction, have been demonstrated using the autophagy enhancers rapamycin and lithium. However, selectivity of these agents is limited, while upstream ALP signaling proteins are involved in many other pathways than autophagy. Broad stimulation of autophagy may therefore cause a wide spectrum of dose-dependent side-effects, suggesting that its clinical applicability is limited. However, recently developed agents selectively targeting core ALP components, including Transcription Factor EB (TFEB), lysosomes, GCase as well as chaperone-mediated autophagy regulators, exert more specific effects on molecular pathogenetic processes causing PD. To conclude, the targeted manipulation of downstream ALP components, rather than broad autophagy stimulation, may be an attractive strategy for the development of novel pharmacological therapies in PD. Further characterization of dysfunctional autophagy in different stages and molecular subtypes of PD in combination with the clinical translation of downstream autophagy regulation offers exciting new avenues for future drug development.


Subject(s)
Autophagy/drug effects , Neuroprotective Agents/pharmacology , Parkinson Disease/drug therapy , Animals , Humans
8.
PLoS One ; 10(6): e0128651, 2015.
Article in English | MEDLINE | ID: mdl-26087293

ABSTRACT

Subjects with incidental Lewy body disease (iLBD) may represent the premotor stage of Parkinson's disease (PD). To elucidate molecular mechanisms underlying neuronal dysfunction and alpha-synuclein pathology in the premotor phase of PD, we investigated the transcriptome of the substantia nigra (SN) of well-characterized iLBD, PD donors and age-matched controls with Braak alpha-synuclein stage ranging from 0-6. In Braak alpha-synuclein stages 1 and 2, we observed deregulation of pathways linked to axonal degeneration, immune response and endocytosis, including axonal guidance signaling, mTOR signaling, EIF2 signaling and clathrin-mediated endocytosis in the SN. In Braak stages 3 and 4, we observed deregulation of pathways involved in protein translation and cell survival, including mTOR and EIF2 signaling. In Braak stages 5 and 6, we observed deregulation of dopaminergic signaling, axonal guidance signaling and thrombin signaling. Throughout the progression of PD pathology, we observed a deregulation of mTOR, EIF2 and regulation of eIF4 and p70S6K signaling in the SN. Our results indicate that molecular mechanisms related to axonal dysfunction, endocytosis and immune response are an early event in PD pathology, whereas mTOR and EIF2 signaling are impaired throughout disease progression. These pathways may hold the key to altering the disease progression in PD.


Subject(s)
Axons/physiology , Endocytosis/physiology , Parkinson Disease/physiopathology , Substantia Nigra/physiopathology , Aged , Case-Control Studies , Disease Progression , Eukaryotic Initiation Factor-2/physiology , Female , Humans , In Situ Hybridization , Lewy Body Disease/physiopathology , Male , Oligonucleotide Array Sequence Analysis , Parkinson Disease/immunology , Signal Transduction/physiology , Substantia Nigra/immunology , TOR Serine-Threonine Kinases/physiology
9.
Blood ; 106(7): 2259-68, 2005 Oct 01.
Article in English | MEDLINE | ID: mdl-15941905

ABSTRACT

Bone marrow cells (BMCs) could correct some pathologic conditions of the central nervous system (CNS) if these cells would effectively repopulate the brain. One such condition is G(M1)-gangliosidosis, a neurodegenerative glycosphingolipidosis due to deficiency of lysosomal beta-galactosidase (beta-gal). In this disease, abnormal build up of G(M1)-ganglioside in the endoplasmic reticulum of brain cells results in calcium imbalance, induction of an unfolded protein response (UPR), and neuronal apoptosis. These processes are accompanied by the activation/proliferation of microglia and the production of inflammatory cytokines. Here we demonstrate that local neuroinflammation promotes the selective activation of chemokines, such as stromal-cell-derived factor 1 (SDF-1), macrophage inflammatory protein 1-alpha (MIP-1alpha), and MIP-1beta, which chemoattract genetically modified BMCs into the CNS. Mice that underwent bone marrow transplantation showed increased beta-gal activity in different brain regions and reduced lysosomal storage. Decreased production of chemokines and effectors of the UPR as well as restoration of neurologic functions accompanied this phenotypic reversion. Our results suggest that beta-gal-expressing bone marrow (BM)-derived cells selectively migrate to the CNS under a gradient of chemokines and become a source of correcting enzyme to deficient neurons. Thus, a disease condition such as G(M1)-gangliosidosis, which is characterized by neurodegeneration and neuroinflammation, may influence the response of the CNS to ex vivo gene therapy.


Subject(s)
Bone Marrow Cells/metabolism , Central Nervous System/metabolism , Chemokines/metabolism , Gangliosidosis, GM1/metabolism , Animals , Apoptosis , Bone Marrow Cells/cytology , Brain/metabolism , Calcium/metabolism , Cell Movement , Cell Proliferation , Cell Transplantation , Chemokine CCL3 , Chemokine CCL4 , Chemokine CXCL12 , Chemokines, CXC/metabolism , Chromatography, Thin Layer , Down-Regulation , Endoplasmic Reticulum/metabolism , Enzyme-Linked Immunosorbent Assay , Gangliosidosis, GM1/genetics , Genetic Therapy/methods , Genetic Vectors , Green Fluorescent Proteins/metabolism , Humans , Immunohistochemistry , Inflammation , Lysosomes/enzymology , Macrophage Inflammatory Proteins/metabolism , Mice , Mice, Transgenic , Microscopy, Fluorescence , Neurodegenerative Diseases/pathology , Neurons/pathology , Protein Folding , RNA/chemistry , Reverse Transcriptase Polymerase Chain Reaction , Ribonucleases/metabolism , Time Factors , Up-Regulation , beta-Galactosidase/metabolism
10.
Mol Cell ; 15(5): 753-66, 2004 Sep 10.
Article in English | MEDLINE | ID: mdl-15350219

ABSTRACT

GM1-ganglioside (GM1) is a major sialoglycolipid of neuronal membranes that, among other functions, modulates calcium homeostasis. Excessive accumulation of GM1 due to deficiency of lysosomal beta-galactosidase (beta-gal) characterizes the neurodegenerative disease GM1-gangliosidosis, but whether the accumulation of GM1 is directly responsible for CNS pathogenesis was unknown. Here we demonstrate that activation of an unfolded protein response (UPR) associated with the upregulation of BiP and CHOP and the activation of JNK2 and caspase-12 leads to neuronal apoptosis in the mouse model of GM1-gangliosidosis. GM1 loading of wild-type neurospheres recapitulated the phenotype of beta-gal-/- cells and activated this pathway by depleting ER calcium stores, which ultimately culminated in apoptosis. Activation of UPR pathways did not occur in mice double deficient for beta-gal and ganglioside synthase, beta-gal-/-/GalNAcT-/-, which do not accumulate GM1. These findings suggest that the UPR can be induced by accumulation of the sialoglycolipid GM1 and this causes a novel mechanism of neuronal apoptosis.


Subject(s)
G(M1) Ganglioside/metabolism , Gangliosidosis, GM1/metabolism , Nerve Degeneration/metabolism , Neurons/metabolism , Protein Folding , Animals , Animals, Newborn , Apoptosis/genetics , CCAAT-Enhancer-Binding Proteins/metabolism , Calcium/metabolism , Caspase 12 , Caspases/metabolism , Cell Death/genetics , Cells, Cultured , Disease Models, Animal , Endoplasmic Reticulum Chaperone BiP , Gangliosidosis, GM1/genetics , Gangliosidosis, GM1/physiopathology , Heat-Shock Proteins/metabolism , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 9 , Mitogen-Activated Protein Kinases/metabolism , Molecular Chaperones/metabolism , N-Acetylgalactosaminyltransferases/metabolism , Nerve Degeneration/genetics , Nerve Degeneration/physiopathology , Neurons/pathology , Transcription Factor CHOP , Transcription Factors/metabolism , beta-Galactosidase/deficiency , beta-Galactosidase/genetics , Polypeptide N-acetylgalactosaminyltransferase
SELECTION OF CITATIONS
SEARCH DETAIL