Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.398
Filter
1.
Int J Mol Sci ; 25(17)2024 Aug 31.
Article in English | MEDLINE | ID: mdl-39273438

ABSTRACT

With emerging genetic association studies, new genes and pathways are revealed as causative factors in the development of Parkinson's disease (PD). However, many of these PD genes are poorly characterized in terms of their function, subcellular localization, and interaction with other components in cellular pathways. This represents a major obstacle towards a better understanding of the molecular causes of PD, with deeper molecular studies often hindered by a lack of high-quality, validated antibodies for detecting the corresponding proteins of interest. In this study, we leveraged the nanoluciferase-derived LgBiT-HiBiT system by generating a cohort of tagged PD genes in both induced pluripotent stem cells (iPSCs) and iPSC-derived neuronal cells. To promote luminescence signals within cells, a master iPSC line was generated, in which LgBiT expression is under the control of a doxycycline-inducible promoter. LgBiT could bind to HiBiT when present either alone or when tagged onto different PD-associated proteins encoded by the genes GBA1, GPNMB, LRRK2, PINK1, PRKN, SNCA, VPS13C, and VPS35. Several HiBiT-tagged proteins could already generate luminescence in iPSCs in response to the doxycycline induction of LgBiT, with the enzyme glucosylceramidase beta 1 (GCase), encoded by GBA1, being one such example. Moreover, the GCase chaperone ambroxol elicited an increase in the luminescence signal in HiBiT-tagged GBA1 cells, correlating with an increase in the levels of GCase in dopaminergic cells. Taken together, we have developed and validated a Doxycycline-inducible luminescence system to serve as a sensitive assay for the quantification, localization, and activity of HiBiT-tagged PD-associated proteins with reliable sensitivity and efficiency.


Subject(s)
Induced Pluripotent Stem Cells , Parkinson Disease , Parkinson Disease/genetics , Parkinson Disease/metabolism , Parkinson Disease/pathology , Humans , Induced Pluripotent Stem Cells/metabolism , Glucosylceramidase/genetics , Glucosylceramidase/metabolism , alpha-Synuclein/metabolism , alpha-Synuclein/genetics , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/genetics , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/metabolism , Neurons/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Vesicular Transport Proteins/genetics , Vesicular Transport Proteins/metabolism , Protein Kinases
2.
Biol Direct ; 19(1): 77, 2024 Sep 05.
Article in English | MEDLINE | ID: mdl-39237967

ABSTRACT

BACKGROUND: GALNTs (UDP-GalNAc; polypeptide N-acetylgalactosaminyltransferases) initiate mucin-type O-GalNAc glycosylation by adding N-GalNAc to protein serine/threonine residues. Abnormalities in O-GalNAc glycosylation are involved in various disorders such as Parkinson's disease (PD), a neurodegenerative disorder. GALNT9 is potentially downregulated in PD patients. METHODS: To determine whether GALNT9 enrichment ameliorates cytotoxicity related to PD-like variations, a pcDNA3.1-GALNT9 plasmid was constructed and transfected into SH-SY5Y cells to establish a GALNT9-overexpressing cell model. RESULTS: Downregulation of GALNT9 and O-GalNAc glycosylation was confirmed in our animal and cellular models of PD-like variations. GALNT9 supplementation greatly attenuated cytotoxicity induced by MPP+ (1-Methyl-4-phenylpyridinium iodide) since it led to increased levels of tyrosine hydroxylase and dopamine, reduced rates of apoptosis, and significantly ameliorated MPP+-induced mitochondrial dysfunction by alleviating abnormal levels of mitochondrial membrane potential and reactive oxygen species. A long-lasting mPTP (mitochondrial permeability transition pores) opening and calcium efflux resulted in significantly lower activity in the cytochrome C-associated apoptotic pathway and mitophagy process, signifying that GALNT9 supplementation maintained neuronal cell health under MPP+ exposure. Additionally, it was found that glycans linked to proteins influenced the formation of protein aggregates containing α-synuclein, and GALNT9 supplement dramatically reduced such insoluble protein aggregations under MPP+ treatment. Glial GALNT9 predominantly appears under pathological conditions like PD-like variations. CONCLUSIONS: GALNT9 enrichment improved cell survival, and glial GALNT9 potentially represents a pathogenic index for PD patients. This study provides insights into the development of therapeutic strategies for the treatment of PD.


Subject(s)
1-Methyl-4-phenylpyridinium , Mitochondria , N-Acetylgalactosaminyltransferases , Polypeptide N-acetylgalactosaminyltransferase , alpha-Synuclein , N-Acetylgalactosaminyltransferases/metabolism , N-Acetylgalactosaminyltransferases/genetics , alpha-Synuclein/metabolism , alpha-Synuclein/genetics , Humans , Mitochondria/metabolism , Mitochondria/drug effects , Animals , 1-Methyl-4-phenylpyridinium/toxicity , 1-Methyl-4-phenylpyridinium/pharmacology , Protein Aggregates , Parkinson Disease/metabolism , Parkinson Disease/genetics , Cell Line, Tumor , Mice , Apoptosis/drug effects , Reactive Oxygen Species/metabolism , Glycosylation , Membrane Potential, Mitochondrial/drug effects , Male
3.
Alzheimers Res Ther ; 16(1): 201, 2024 Sep 10.
Article in English | MEDLINE | ID: mdl-39256877

ABSTRACT

BACKGROUND: The term rapidly progressive dementia (RPD) with Lewy bodies (rpDLB) is used for DLB patients who develop a rapidly progressive neurological syndrome and have reduced survival. Here, we characterise the clinical, neuropathological, and molecular characteristics of a large rpDLB neuropathological series. METHODS: We included all RPD patients with a disease duration < 4 years submitted to our prion disease referral centre between 2003 and 2022 who showed Lewy body pathology (LBP) in limbic or neocortical stages as primary neuropathological diagnosis, had no systemic condition justifying the rapid deterioration and were previously neurologically unimpaired. Clinical features were retrieved and compared with Creutzfeldt-Jakob disease (CJD) and rapidly progressive Alzheimer's disease (rpAD) cohorts. Neuropathological and genetic (whole exome sequencing, APOE genotyping, and C9orf72 repeat expansion analysis) characteristics of rpDLB patients were systematically investigated. We scored semi-quantitatively the LBP load and performed a α-synuclein (αSyn) RT-QuIC seeding amplification assay (SAA) on cerebrospinal fluid (CSF) and tenfold serially diluted brain homogenates from different brain areas in rpDLB patients and typical long-lasting Lewy body disease (LBD) with dementia patients as control group. RESULTS: RpDLB patients were older (p = 0.047) and presented more cognitive fluctuations (p = 0.005), visual hallucinations (p = 0.020), neuropsychiatric symptoms (p = 0.006) and seizures (p = 0.032), and fewer cerebellar (p < 0.001) and visual (p = 0.004) signs than CJD ones. Delirium onset was more common than in both CJD (p < 0.001) and rpAD (p = 0.008). Atypical LBD signs (pyramidal, myoclonus, akinetic mutism) were common. All tested patients were positive by CSF αSyn SAA. Concomitant pathologies were common, with only four cases showing relatively "pure" LBP. LBP load and αSyn seeding activity measured through αSyn RT-QuIC SAA were not significantly different between rpDLB patients and typical LBD. We found a likely pathogenic variant in GBA in one patient. CONCLUSIONS: Our results indicate that: 1) rpDLB exhibits a distinct clinical signature (2) CSF αSyn SAA is a reliable diagnostic test; 3) rpDLB is a heterogeneous neuropathological entity that can be underlain by both widespread pure LBP, or multiple copathologies 4) rpDLB is likely not sustained by distinct αSyn conformational strains; 5) genetic defects may, at least occasionally, contribute to the poor prognosis in these patients.


Subject(s)
Disease Progression , Lewy Body Disease , Humans , Lewy Body Disease/genetics , Lewy Body Disease/pathology , Lewy Body Disease/cerebrospinal fluid , Female , Male , Aged , Aged, 80 and over , Middle Aged , Brain/pathology , alpha-Synuclein/cerebrospinal fluid , alpha-Synuclein/genetics , alpha-Synuclein/metabolism , Creutzfeldt-Jakob Syndrome/genetics , Creutzfeldt-Jakob Syndrome/pathology , Creutzfeldt-Jakob Syndrome/cerebrospinal fluid , Creutzfeldt-Jakob Syndrome/diagnosis , Lewy Bodies/pathology
4.
PLoS Pathog ; 20(9): e1012517, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39264912

ABSTRACT

The cellular prion protein, PrPC, has been postulated to function as a receptor for α-synuclein, potentially facilitating cell-to-cell spreading and/or toxicity of α-synuclein aggregates in neurodegenerative disorders such as Parkinson's disease. Previously, we generated the "Salt (S)" and "No Salt (NS)" strains of α-synuclein aggregates that cause distinct pathological phenotypes in M83 transgenic mice overexpressing A53T-mutant human α-synuclein. To test the hypothesis that PrPC facilitates the propagation of α-synuclein aggregates, we produced M83 mice that either express or do not express PrPC. Following intracerebral inoculation with the S or NS strain, the absence of PrPC in M83 mice did not prevent disease development and had minimal influence on α-synuclein strain-specified attributes such as the extent of cerebral α-synuclein deposition, selective targeting of specific brain regions and cell types, the morphology of induced α-synuclein deposits, and the structural fingerprints of protease-resistant α-synuclein aggregates. Likewise, there were no appreciable differences in disease manifestation between PrPC-expressing and PrPC-lacking M83 mice following intraperitoneal inoculation of the S strain. Interestingly, intraperitoneal inoculation with the NS strain resulted in two distinct disease phenotypes, indicative of α-synuclein strain evolution, but this was also independent of PrPC expression. Overall, these results suggest that PrPC plays at most a minor role in the propagation, neuroinvasion, and evolution of α-synuclein strains in mice that express A53T-mutant human α-synuclein. Thus, other putative receptors or cell-to-cell propagation mechanisms may have a larger effect on the spread of α-synuclein aggregates during disease.


Subject(s)
Synucleinopathies , alpha-Synuclein , Animals , Humans , Mice , alpha-Synuclein/metabolism , alpha-Synuclein/genetics , Brain/metabolism , Brain/pathology , Disease Models, Animal , Mice, Transgenic , PrPC Proteins/metabolism , PrPC Proteins/genetics , Synucleinopathies/metabolism , Synucleinopathies/pathology
5.
Biochim Biophys Acta Mol Basis Dis ; 1870(8): 167494, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39233262

ABSTRACT

SNCA/PARK1 encodes α-synuclein, which is associated with familial Parkinson's disease. Despite its abundance in presynaptic terminals, the aggregation mechanism of α-synuclein and its relationship with Parkinson's disease have not yet been elucidated. Moreover, the ultrastructures of α-synuclein localization sites in neuronal presynaptic terminals remain unclear. Therefore, we herein generated transgenic mice expressing human α-synuclein tagged with mKate2 (hSNCA-mKate2 mice). These mice exhibited normal growth and fertility and had no motor dysfunction relative to their wild-type littermates, even at one year old. α-Synuclein-mKate2 accumulated in presynaptic terminals, particularly between Purkinje cells in the cerebellum and neurons in cerebellar nuclei. α-Synuclein-mKate2 was associated with the presynaptic marker, synaptophysin. In-resin CLEM and immunoelectron or electron microscopy revealed that α-synuclein-mKate2 localized on the surface of synaptic vesicles that were tightly arranged and assembled to form large synaptic pools in the cerebellum with negligible effects on the active zone. These results suggest that α-synuclein-associated ultrastructures in the presynaptic terminals of hSNCA-mKate2 mice reflect the structures of α-synuclein-assembled synaptic vesicle pools, and the size of vesicle pools increased. This transgenic mouse model will be a valuable tool for studying α-synuclein-associated synaptic vesicle pools.


Subject(s)
Mice, Transgenic , Presynaptic Terminals , Synaptic Vesicles , alpha-Synuclein , Animals , alpha-Synuclein/metabolism , alpha-Synuclein/genetics , Presynaptic Terminals/metabolism , Presynaptic Terminals/ultrastructure , Synaptic Vesicles/metabolism , Synaptic Vesicles/ultrastructure , Mice , Humans , Purkinje Cells/metabolism , Purkinje Cells/ultrastructure , Parkinson Disease/metabolism , Parkinson Disease/pathology , Cerebellum/metabolism , Cerebellum/ultrastructure , Synaptophysin/metabolism , Synaptophysin/genetics , Male
6.
Bull Exp Biol Med ; 177(4): 412-417, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39259467

ABSTRACT

The behavioral effects of α-synuclein oligomers were studied at various times after its chronic intranasal administration to 75-day-old C57BL/6J mice in comparison with the dynamics of changes in the transcriptional activity of caspases genes (Casp9, Casp8, and Casp3) in the hippocampus, frontal cortex, and cerebellum. The negative effects of α-synuclein oligomers on exploratory activity and short-term memory in the novel object recognition test were most pronounced after 90 days from the end of administration, while after 1 and 270 days, partial compensation of the studied cognitive functions was observed. Analysis of the expression of caspase genes suggests that early compensatory mechanisms are associated with suppression of the effector caspase-3 gene expression along with increased activity of the genes encoding initiator caspases-9 and -8. Late compensation processes are associated with a decrease in the activity of initiator caspases in the frontal cortex and cerebellum.


Subject(s)
Caspase 3 , Caspase 8 , Caspase 9 , Cerebellum , Cognitive Dysfunction , Hippocampus , Mice, Inbred C57BL , alpha-Synuclein , Animals , Mice , Caspase 3/genetics , Caspase 3/metabolism , alpha-Synuclein/genetics , alpha-Synuclein/metabolism , Cognitive Dysfunction/genetics , Cognitive Dysfunction/metabolism , Caspase 8/genetics , Caspase 8/metabolism , Caspase 9/genetics , Caspase 9/metabolism , Hippocampus/metabolism , Cerebellum/metabolism , Frontal Lobe/metabolism , Male , Disease Models, Animal , Parkinsonian Disorders/genetics , Parkinsonian Disorders/metabolism , Memory, Short-Term/drug effects
7.
Reprod Fertil Dev ; 362024 Sep.
Article in English | MEDLINE | ID: mdl-39283977

ABSTRACT

Context Understanding of central nervous system mechanisms related to age-related infertility remains limited. Fibril α-synuclein, distinct from its monomer form, is implicated in age-related diseases and propagates among neurons akin to prions. Aims We compared α-synuclein expression in gonadotropin-releasing hormone-expressing neurons (GnRH neurons) in the pre-optic area, arcuate nucleus, and median eminence of healthy heifers and aged cows to determine its role in age-related infertility. Methods We analysed mRNA and protein expression, along with fluorescent immunohistochemistry for GnRH and α-synuclein, followed by Congo red staining to detect amyloid deposits, and confocal microscopy. Key results Both mRNA and protein expressions of α-synuclein were confirmed by reverse transcription-polymerase chain reaction (RT-PCR) and western blots in bovine cortex, hippocampus, and anterior and posterior hypothalamus tissues. Significant differences in α-synuclein mRNA expression were observed in the cortex and hippocampus between young and old cows. Western blots showed five bands of α-synuclein, probably reflecting monomer, dimer, and oligomers, in the cortex, hippocampus, hypothalamus tissues, and there were significant differences in some bands between young and old cows. Bright-field and polarised light microscopy did not detect obvious amyloid deposition in aged hypothalami; however, higher-sensitive confocal microscopy unveiled strong positive signal of Congo red and α-synuclein in GnRH neurons in aged hypothalami. Additionally, α-synuclein expression was detected in immortalised GnRH neurons, GT1-7 cells. Conclusion Alpha-synuclein was expressed in GnRH neurons, and some differences were observed between young and old hypothalami. Implications Alpha-synuclein may play an important role in aging-related infertility.


Subject(s)
Aging , Gonadotropin-Releasing Hormone , Hypothalamus , Neurons , alpha-Synuclein , Animals , Gonadotropin-Releasing Hormone/metabolism , Gonadotropin-Releasing Hormone/genetics , Cattle , alpha-Synuclein/metabolism , alpha-Synuclein/genetics , Neurons/metabolism , Hypothalamus/metabolism , Female , Aging/metabolism , RNA, Messenger/metabolism
8.
Proc Natl Acad Sci U S A ; 121(39): e2406479121, 2024 Sep 24.
Article in English | MEDLINE | ID: mdl-39284050

ABSTRACT

Parkinson's disease (PD) is typically a sporadic late-onset disorder, which has made it difficult to model in mice. Several transgenic mouse models bearing mutations in SNCA, which encodes alpha-Synuclein (α-Syn), have been made, but these lines do not express SNCA in a physiologically accurate spatiotemporal pattern, which limits the ability of the mice to recapitulate the features of human PD. Here, we generated knock-in mice bearing the G51D SNCA mutation. After establishing that their motor symptoms begin at 9 mo of age, we then sought earlier pathologies. We assessed the phosphorylation at Serine 129 of α-Syn in different tissues and detected phospho-α-Syn in the olfactory bulb and enteric nervous system at 3 mo of age. Olfactory deficit and impaired gut transit followed at 6 mo, preceding motor symptoms. The SncaG51D mice thus parallel the progression of human PD and will enable us to study PD pathogenesis and test future therapies.


Subject(s)
Disease Models, Animal , Gene Knock-In Techniques , Parkinson Disease , alpha-Synuclein , Animals , alpha-Synuclein/metabolism , alpha-Synuclein/genetics , Mice , Parkinson Disease/genetics , Parkinson Disease/metabolism , Parkinson Disease/physiopathology , Parkinson Disease/pathology , Mice, Transgenic , Phosphorylation , Olfaction Disorders/genetics , Olfaction Disorders/metabolism , Olfaction Disorders/physiopathology , Olfactory Bulb/metabolism , Olfactory Bulb/pathology , Gastrointestinal Diseases/genetics , Gastrointestinal Diseases/metabolism , Gastrointestinal Diseases/pathology , Enteric Nervous System/metabolism , Enteric Nervous System/physiopathology , Humans , Male
9.
Int J Mol Sci ; 25(17)2024 Aug 25.
Article in English | MEDLINE | ID: mdl-39273169

ABSTRACT

Parkinson's disease (PD) is a multifactorial, chronic, and progressive neurodegenerative disorder inducing movement alterations as a result of the loss of dopaminergic (DAergic) neurons of the pars compacta in the substantia nigra and protein aggregates of alpha synuclein (α-Syn). Although its etiopathology agent has not yet been clearly established, environmental and genetic factors have been suggested as the major contributors to the disease. Mutations in the glucosidase beta acid 1 (GBA1) gene, which encodes the lysosomal glucosylceramidase (GCase) enzyme, are one of the major genetic risks for PD. We found that the GBA1 K198E fibroblasts but not WT fibroblasts showed reduced catalytic activity of heterozygous mutant GCase by -70% but its expression levels increased by 3.68-fold; increased the acidification of autophagy vacuoles (e.g., autophagosomes, lysosomes, and autolysosomes) by +1600%; augmented the expression of autophagosome protein Beclin-1 (+133%) and LC3-II (+750%), and lysosomal-autophagosome fusion protein LAMP-2 (+107%); increased the accumulation of lysosomes (+400%); decreased the mitochondrial membrane potential (∆Ψm) by -19% but the expression of Parkin protein remained unperturbed; increased the oxidized DJ-1Cys106-SOH by +900%, as evidence of oxidative stress; increased phosphorylated LRRK2 at Ser935 (+1050%) along with phosphorylated α-synuclein (α-Syn) at pathological residue Ser129 (+1200%); increased the executer apoptotic protein caspase 3 (cleaved caspase 3) by +733%. Although exposure of WT fibroblasts to environmental neutoxin rotenone (ROT, 1 µM) exacerbated the autophagy-lysosomal system, oxidative stress, and apoptosis markers, ROT moderately increased those markers in GBA1 K198E fibroblasts. We concluded that the K198E mutation endogenously primes skin fibroblasts toward autophagy dysfunction, OS, and apoptosis. Our findings suggest that the GBA1 K198E fibroblasts are biochemically and molecularly equivalent to the response of WT GBA1 fibroblasts exposed to ROT.


Subject(s)
Apoptosis , Autophagy , Fibroblasts , Glucosylceramidase , Mitochondria , Oxidative Stress , Glucosylceramidase/metabolism , Glucosylceramidase/genetics , Humans , Fibroblasts/metabolism , Autophagy/genetics , Mitochondria/metabolism , Parkinson Disease/metabolism , Parkinson Disease/genetics , Parkinson Disease/pathology , Skin/metabolism , Skin/pathology , Lysosomes/metabolism , alpha-Synuclein/metabolism , alpha-Synuclein/genetics , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/metabolism , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/genetics , Mutation
10.
PeerJ ; 12: e18100, 2024.
Article in English | MEDLINE | ID: mdl-39285918

ABSTRACT

Genetically modified organisms are commonly used in disease research and agriculture but the precise genomic alterations underlying transgenic mutations are often unknown. The position and characteristics of transgenes, including the number of independent insertions, influences the expression of both transgenic and wild-type sequences. We used long-read, Oxford Nanopore Technologies (ONT) to sequence and assemble two transgenic strains of Caenorhabditis elegans commonly used in the research of neurodegenerative diseases: BY250 (pPdat-1::GFP) and UA44 (GFP and human α-synuclein), a model for Parkinson's research. After scaffolding to the reference, the final assembled sequences were ∼102 Mb with N50s of 17.9 Mb and 18.0 Mb, respectively, and L90s of six contiguous sequences, representing chromosome-level assemblies. Each of the assembled sequences contained more than 99.2% of the Nematoda BUSCO genes found in the C. elegans reference and 99.5% of the annotated C. elegans reference protein-coding genes. We identified the locations of the transgene insertions and confirmed that all transgene sequences were inserted in intergenic regions, leaving the organismal gene content intact. The transgenic C. elegans genomes presented here will be a valuable resource for Parkinson's research as well as other neurodegenerative diseases. Our work demonstrates that long-read sequencing is a fast, cost-effective way to assemble genome sequences and characterize mutant lines and strains.


Subject(s)
Animals, Genetically Modified , Caenorhabditis elegans , Nanopore Sequencing , Transgenes , Caenorhabditis elegans/genetics , Animals , Transgenes/genetics , Animals, Genetically Modified/genetics , Nanopore Sequencing/methods , alpha-Synuclein/genetics , Genome, Helminth , Mutagenesis, Insertional , Humans , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism
11.
Sci Adv ; 10(37): eadk3700, 2024 Sep 13.
Article in English | MEDLINE | ID: mdl-39259788

ABSTRACT

Aggregated α-synuclein (α-SYN) proteins, encoded by the SNCA gene, are hallmarks of Lewy body disease (LBD), affecting multiple brain regions. However, the specific mechanisms underlying α-SYN pathology in cortical neurons, crucial for LBD-associated dementia, remain unclear. Here, we recapitulated α-SYN pathologies in human induced pluripotent stem cells (iPSCs)-derived cortical organoids generated from patients with LBD with SNCA gene triplication. Single-cell RNA sequencing, combined with functional and molecular validation, identified synaptic and mitochondrial dysfunction in excitatory neurons exhibiting high expression of the SNCA gene, aligning with observations in the cortex of autopsy-confirmed LBD human brains. Furthermore, we screened 1280 Food and Drug Administration-approved drugs and identified four candidates (entacapone, tolcapone, phenazopyridine hydrochloride, and zalcitabine) that inhibited α-SYN seeding activity in real-time quaking-induced conversion assays with human brains, reduced α-SYN aggregation, and alleviated mitochondrial dysfunction in SNCA triplication organoids and excitatory neurons. Our findings establish human cortical LBD models and suggest potential therapeutic drugs targeting α-SYN aggregation for LBD.


Subject(s)
Induced Pluripotent Stem Cells , Lewy Body Disease , Organoids , alpha-Synuclein , Humans , alpha-Synuclein/metabolism , alpha-Synuclein/genetics , Organoids/metabolism , Organoids/drug effects , Organoids/pathology , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/cytology , Lewy Body Disease/pathology , Lewy Body Disease/genetics , Lewy Body Disease/metabolism , Lewy Body Disease/drug therapy , Mitochondria/metabolism , Mitochondria/drug effects , Mitochondria/genetics , Neurons/metabolism , Neurons/drug effects , Neurons/pathology , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Cerebral Cortex/drug effects , Drug Evaluation, Preclinical
12.
Cell Mol Life Sci ; 81(1): 403, 2024 Sep 14.
Article in English | MEDLINE | ID: mdl-39276174

ABSTRACT

Insulin resistance (IR) was found to be a critical element in the pathogenesis of Parkinson's disease (PD), facilitating abnormal α-synuclein (α-Syn) aggregation in neurons and thus promoting PD development. However, how IR contributes to abnormal α-Syn aggregation remains ill-defined. Here, we analyzed six PD postmortem brain transcriptome datasets to reveal module genes implicated in IR-mediated α-Syn aggregation. In addition, we induced IR in cultured dopaminergic (DA) neurons overexpressing α-Syn to identify IR-modulated differentially expressed genes (DEGs). Integrated analysis of data from PD patients and cultured neurons revealed 226 genes involved in α-Syn aggregation under IR conditions, of which 53 exhibited differential expression between PD patients and controls. Subsequently, we conducted an integrated analysis of the 53 IR-modulated genes employing transcriptome data from PD patients with different Braak stages and DA neuron subclasses with varying α-Syn aggregation scores. Protein tyrosine phosphatase receptor type O (PTPRO) was identified to be closely associated with PD progression and α-Syn aggregation. Experimental validation in a cultured PD cell model confirmed that both mRNA and protein of PTPRO were reduced under IR conditions, and the downregulation of PTPRO significantly facilitated α-Syn aggregation and cell death. Collectively, our findings identified PTPRO as a key regulator in IR-mediated α-Syn aggregation and uncovered its prospective utility as a therapeutic target in PD patients with IR.


Subject(s)
Insulin Resistance , Parkinson Disease , alpha-Synuclein , alpha-Synuclein/metabolism , alpha-Synuclein/genetics , Humans , Parkinson Disease/metabolism , Parkinson Disease/genetics , Parkinson Disease/pathology , Insulin Resistance/genetics , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , Animals , Transcriptome , Male , Receptor-Like Protein Tyrosine Phosphatases, Class 3/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 3/genetics , Female , Protein Aggregation, Pathological/metabolism , Protein Aggregation, Pathological/genetics
13.
J Neuroinflammation ; 21(1): 216, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-39218899

ABSTRACT

Parkinson's disease (PD) is characterized by neuroinflammation, progressive loss of dopaminergic neurons, and accumulation of α-synuclein (α-Syn) into insoluble aggregates called Lewy pathology. The Line 61 α-Syn mouse is an established preclinical model of PD; Thy-1 is used to promote human α-Syn expression, and features of sporadic PD develop at 9-18 months of age. To accelerate the PD phenotypes, we injected sonicated human α-Syn preformed fibrils (PFFs) into the striatum, which produced phospho-Syn (p-α-Syn) inclusions in the substantia nigra pars compacta and significantly increased MHC Class II-positive immune cells. Additionally, there was enhanced infiltration and activation of innate and adaptive immune cells in the midbrain. We then used this new model, Line 61-PFF, to investigate the effect of inhibiting the JAK/STAT signaling pathway, which is critical for regulation of innate and adaptive immune responses. After administration of the JAK1/2 inhibitor AZD1480, immunofluorescence staining showed a significant decrease in p-α-Syn inclusions and MHC Class II expression. Flow cytometry showed reduced infiltration of CD4+ T-cells, CD8+ T-cells, CD19+ B-cells, dendritic cells, macrophages, and endogenous microglia into the midbrain. Importantly, single-cell RNA-Sequencing analysis of CD45+ cells from the midbrain identified 9 microglia clusters, 5 monocyte/macrophage (MM) clusters, and 5 T-cell (T) clusters, in which potentially pathogenic MM4 and T3 clusters were associated with neuroinflammatory responses in Line 61-PFF mice. AZD1480 treatment reduced cell numbers and cluster-specific expression of the antigen-presentation genes H2-Eb1, H2-Aa, H2-Ab1, and Cd74 in the MM4 cluster and proinflammatory genes such as Tnf, Il1b, C1qa, and C1qc in the T3 cluster. Together, these results indicate that inhibiting the JAK/STAT pathway suppresses the activation and infiltration of innate and adaptive cells, reducing neuroinflammation in the Line 61-PFF mouse model.


Subject(s)
Disease Models, Animal , Neuroinflammatory Diseases , Parkinson Disease , STAT Transcription Factors , Signal Transduction , alpha-Synuclein , Animals , Mice , Neuroinflammatory Diseases/metabolism , Neuroinflammatory Diseases/drug therapy , alpha-Synuclein/metabolism , alpha-Synuclein/genetics , Signal Transduction/drug effects , Signal Transduction/physiology , STAT Transcription Factors/metabolism , STAT Transcription Factors/antagonists & inhibitors , STAT Transcription Factors/genetics , Parkinson Disease/metabolism , Parkinson Disease/pathology , Parkinson Disease/immunology , Humans , Mice, Transgenic , Mice, Inbred C57BL , Janus Kinases/metabolism , Janus Kinases/antagonists & inhibitors , Parkinsonian Disorders/metabolism , Parkinsonian Disorders/pathology , Parkinsonian Disorders/immunology , Pyrimidines/pharmacology
14.
JAMA Neurol ; 81(9): 905-906, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-39102226

ABSTRACT

This Viewpoint cautions against premature adoption of the α-synuclein seed amplification assay as a biomarker test for Parkinson disease in general neurology practice.


Subject(s)
alpha-Synuclein , Humans , alpha-Synuclein/genetics , alpha-Synuclein/metabolism , Parkinson Disease/genetics , Parkinson Disease/diagnosis , Parkinsonian Disorders/genetics , Parkinsonian Disorders/diagnosis
15.
Neurosci Lett ; 839: 137936, 2024 Sep 14.
Article in English | MEDLINE | ID: mdl-39151573

ABSTRACT

Synucleins, including α-synuclein (α-syn), ß-syn, and γ-syn, have been implicated in various synucleinopathies, notably Parkinson's disease (PD), which has generated increased interest in understanding their roles. Although α-syn and ß-syn have contrasting neuropathological consequences, the precise role of γ-syn remains unclear. This study validated non-motor symptoms, specifically anxiety-like behavior, along with the degradation of dopaminergic (DAergic) neurons in the nigrostriatal system and DAergic neurites in the prefrontal cortex and hippocampus of rats infused with striatal 6-hydroxydopamine (6-OHDA). Our study further investigated the alterations in γ-syn expression levels in the prefrontal cortices and hippocampi of these 6-OHDA-treated rats, aiming to establish foundational insights into the neuropathophysiology of DA depletion, a central feature of PD. Our findings revealed a significant increase in the expression of γ-syn mRNA and protein in these brain regions, in contrast to unaltered α- and ß-syn expression levels. This suggests a distinct role of γ-syn within the neurobiological milieu under conditions of DA deficiency. Overall, our data shed light on the neurobiological changes observed in the hemiparkinsonian rat model induced with 6-OHDA, underscoring the potential significance of γ-syn in PD pathology.


Subject(s)
Dopamine , Hippocampus , Oxidopamine , Prefrontal Cortex , Up-Regulation , gamma-Synuclein , Animals , Prefrontal Cortex/metabolism , Oxidopamine/toxicity , Male , Hippocampus/metabolism , Dopamine/metabolism , gamma-Synuclein/metabolism , gamma-Synuclein/genetics , Rats , Rats, Sprague-Dawley , Parkinsonian Disorders/metabolism , Parkinsonian Disorders/chemically induced , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , Corpus Striatum/metabolism , Disease Models, Animal , alpha-Synuclein/metabolism , alpha-Synuclein/genetics
16.
Pathol Res Pract ; 261: 155511, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39094523

ABSTRACT

Parkinson's disease is one of the vital neurodegenerative ailments attributed to a rise in Alpha-synuclein proteins leading to the advancement of motor and cognitive deterioration. Interestingly, in PD lncRNAs, miRNAs and siRNAs are also key regulators of SNCA and alpha-synuclein aggregation. This review will focus on the roles of these three types of small RNAs in trebling the development of PD through regulating SNCA expression or alpha-synuclein protein mediating the RNA from acting. Parkinson's disease is defined by the build-up of alpha-synuclein protein resulting predominantly from the elevated expression level of the SNCA gene. Non-coding RNAs have gained broad appeal as fundamental modulators of gene expression and protein aggregation dynamics, with significant implications on the aetiology of PD. LncRNAs modulate SNCA transcription and edit epigenetic modifications, while miRNA target mRNA is involved in the stability and translation of count alpha-synuclein. Considering all these data, siRNAs can achieve the precise gene silencing effect that directly induces the downregulation of SNCA mRNA. This review also summarizes some recent reports about the interaction between these ncRNAs with the SNCA gene and alpha-synuclein protein, each through its independent in addition to synergistic mechanisms. This review highlights the possibility of therapeutic interventions to perturb SNCA expression to prevent alpha-synuclein aggregation via targeting ncRNAs that might be spun off novel drug development for PD.


Subject(s)
Parkinson Disease , alpha-Synuclein , alpha-Synuclein/metabolism , alpha-Synuclein/genetics , Humans , Parkinson Disease/genetics , Parkinson Disease/metabolism , RNA, Untranslated/genetics , RNA, Untranslated/metabolism , Animals , MicroRNAs/genetics , MicroRNAs/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Gene Expression Regulation
17.
Int J Biol Macromol ; 277(Pt 4): 134417, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39098688

ABSTRACT

Alpha-synuclein, encoded by the SNCA gene, is a pivotal protein implicated in the pathogenesis of synucleinopathies, including Parkinson's disease. Current approaches for modulating alpha-synuclein levels involve antisense nucleotides, siRNAs, and small molecules targeting SNCA's 5'-UTR mRNA. Here, we propose a groundbreaking strategy targeting G-quadruplex structures to effectively modulate SNCA gene expression and lowering alpha-synuclein amount. Novel G-quadruplex sequences, identified on the SNCA gene's transcription starting site and 5'-UTR of SNCA mRNAs, were experimentally confirmed for their stability through biophysical assays and in vitro experiments on human genomic DNA. Biological validation in differentiated SH-SY5Y cells revealed that well-known G-quadruplex ligands remarkably stabilized these structures, inducing the modulation of SNCA mRNAs expression, and the effective decrease in alpha-synuclein amount. Besides, a novel peptide nucleic acid conjugate, designed to selectively disrupt of G-quadruplex within the SNCA gene promoter, caused a promising lowering of both SNCA mRNA and alpha-synuclein protein. Altogether our findings highlight G-quadruplexes' key role as intriguing biological targets in achieving a notable and successful reduction in alpha-synuclein expression, pointing to a novel approach against synucleinopathies.


Subject(s)
G-Quadruplexes , alpha-Synuclein , alpha-Synuclein/genetics , alpha-Synuclein/metabolism , Humans , RNA, Messenger/genetics , RNA, Messenger/metabolism , Cell Line, Tumor , Promoter Regions, Genetic , Gene Expression Regulation/drug effects , 5' Untranslated Regions/genetics , Parkinson Disease/genetics , Parkinson Disease/metabolism , Peptide Nucleic Acids/pharmacology , Peptide Nucleic Acids/chemistry
18.
Neurobiol Dis ; 200: 106647, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39187209

ABSTRACT

In this study, we develop and validate a new Parkinson's disease (PD) mouse model that can be used to better understand how the disease progresses and to test the effects of new, potentially disease-modifying, PD therapies. Our central hypothesis is that mitochondrial dysfunction intercalates with misfolded α-synuclein (α-syn) accumulation in a vicious cycle, leading to the loss of nigral neurons. Our hypothesis builds on the concept that PD involves multiple molecular insults, including mitochondrial dysfunction and aberrant α-syn handling. We predicted that mitochondrial deficits, due to heterozygous loss of Engrailed-1 (En1+/-), combined with bilateral injections of pathogenic α-syn fibrils (PFFs), will act to generate a highly relevant PD model - the En1/SYN model. Here, En1+/- mice received bilateral intrastriatal stereotaxic injections of either PBS or α-syn fibrils and were analyzed using automated behavioral tests and deep learning-assisted histological analysis at 2, 4, and 6 months post-injection. We observed significant and progressive Lewy body-like inclusion pathology in the amygdala, motor cortex, and cingulate cortex, as well as the loss of tyrosine hydroxylase-positive (TH+) cells in the substantia nigra. The En1/SYN model also exhibited significant motor impairments at 6 months post-injection, which were however not exacerbated as we had expected. Still, this model has a comprehensive number of PD-like phenotypes and is therefore superior when compared to the α-syn PFF or En1+/- models alone.


Subject(s)
Homeodomain Proteins , Parkinson Disease , alpha-Synuclein , Animals , Male , Mice , alpha-Synuclein/metabolism , alpha-Synuclein/genetics , Artificial Intelligence , Disease Models, Animal , Heterozygote , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Mice, Inbred C57BL , Parkinson Disease/metabolism , Parkinson Disease/pathology , Parkinson Disease/genetics , Substantia Nigra/metabolism , Substantia Nigra/pathology , Substantia Nigra/drug effects , Synucleinopathies/pathology , Synucleinopathies/metabolism , Synucleinopathies/genetics
19.
Proc Natl Acad Sci U S A ; 121(34): e2315006121, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-39133842

ABSTRACT

Amyloid formation by α-synuclein (αSyn) occurs in Parkinson's disease, multiple system atrophy, and dementia with Lewy bodies. Deciphering the residues that regulate αSyn amyloid fibril formation will not only provide mechanistic insight but may also reveal targets to prevent and treat disease. Previous investigations have identified several regions of αSyn to be important in the regulation of amyloid formation, including the non-amyloid-ß component (NAC), P1 region (residues 36 to 42), and residues in the C-terminal domain. Recent studies have also indicated the importance of the N-terminal region of αSyn for both its physiological and pathological roles. Here, the role of residues 2 to 7 in the N-terminal region of αSyn is investigated in terms of their ability to regulate amyloid fibril formation in vitro and in vivo. Deletion of these residues (αSynΔN7) slows the rate of fibril formation in vitro and reduces the capacity of the protein to be recruited by wild-type (αSynWT) fibril seeds, despite cryo-EM showing a fibril structure consistent with those of full-length αSyn. Strikingly, fibril formation of αSynΔN7 is not induced by liposomes, despite the protein binding to liposomes with similar affinity to αSynWT. A Caenorhabditis elegans model also showed that αSynΔN7::YFP forms few puncta and lacks motility and lifespan defects typified by expression of αSynWT::YFP. Together, the results demonstrate the involvement of residues 2 to 7 of αSyn in amyloid formation, revealing a target for the design of amyloid inhibitors that may leave the functional role of the protein in membrane binding unperturbed.


Subject(s)
Amyloid , Caenorhabditis elegans , alpha-Synuclein , alpha-Synuclein/metabolism , alpha-Synuclein/genetics , alpha-Synuclein/chemistry , Amyloid/metabolism , Caenorhabditis elegans/metabolism , Animals , Humans , Lipids/chemistry , Parkinson Disease/metabolism , Parkinson Disease/genetics , Parkinson Disease/pathology
20.
Elife ; 122024 Aug 28.
Article in English | MEDLINE | ID: mdl-39196271

ABSTRACT

The aggregation of the protein α-synuclein is closely associated with several neurodegenerative disorders and as such the structures of the amyloid fibril aggregates have high scientific and medical significance. However, there are dozens of unique atomic-resolution structures of these aggregates, and such a highly polymorphic nature of the α-synuclein fibrils hampers efforts in disease-relevant in vitro studies on α-synuclein amyloid aggregation. In order to better understand the factors that affect polymorph selection, we studied the structures of α-synuclein fibrils in vitro as a function of pH and buffer using cryo-EM helical reconstruction. We find that in the physiological range of pH 5.8-7.4, a pH-dependent selection between Type 1, 2, and 3 polymorphs occurs. Our results indicate that even in the presence of seeds, the polymorph selection during aggregation is highly dependent on the buffer conditions, attributed to the non-polymorph-specific nature of secondary nucleation. We also uncovered two new polymorphs that occur at pH 7.0 in phosphate-buffered saline. The first is a monofilament Type 1 fibril that highly resembles the structure of the juvenile-onset synucleinopathy polymorph found in patient-derived material. The second is a new Type 5 polymorph that resembles a polymorph that has been recently reported in a study that used diseased tissues to seed aggregation. Taken together, our results highlight the shallow amyloid energy hypersurface that can be altered by subtle changes in the environment, including the pH which is shown to play a major role in polymorph selection and in many cases appears to be the determining factor in seeded aggregation. The results also suggest the possibility of producing disease-relevant structure in vitro.


Subject(s)
Amyloid , alpha-Synuclein , alpha-Synuclein/chemistry , alpha-Synuclein/metabolism , alpha-Synuclein/genetics , Hydrogen-Ion Concentration , Amyloid/chemistry , Amyloid/metabolism , Humans , Cryoelectron Microscopy , Protein Aggregates , Protein Aggregation, Pathological
SELECTION OF CITATIONS
SEARCH DETAIL