Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
EMBO Mol Med ; 16(7): 1657-1674, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38839930

ABSTRACT

Synucleinopathies such as Parkinson's disease (PD) are defined by the accumulation and aggregation of the α-synuclein protein in neurons, glia and other tissues. We have previously shown that destabilization of α-synuclein tetramers is associated with familial PD due to SNCA mutations and demonstrated brain-region specific alterations of α-synuclein multimers in sporadic PD patients following the classical Braak spreading theory. In this study, we assessed relative levels of disordered and higher-ordered multimeric forms of cytosolic α-synuclein in blood from familial PD with G51D mutations and sporadic PD patients. We used an adapted in vitro-cross-linking protocol for human EDTA-whole blood. The relative levels of higher-ordered α-synuclein tetramers were diminished in blood from familial PD and sporadic PD patients compared to controls. Interestingly, the relative amount of α-synuclein tetramers was already decreased in asymptomatic G51D carriers, supporting the hypothesis that α-synuclein multimer destabilization precedes the development of clinical PD. Our data, therefore suggest that measuring α-synuclein tetramers in blood may have potential as a facile biomarker assay for early detection and quantitative tracking of PD progression.


Subject(s)
Parkinson Disease , alpha-Synuclein , Humans , alpha-Synuclein/metabolism , alpha-Synuclein/blood , Parkinson Disease/blood , Parkinson Disease/metabolism , Parkinson Disease/genetics , Aged , Male , Female , Middle Aged , Protein Multimerization , Protein Aggregates
3.
Sci Rep ; 13(1): 14911, 2023 09 09.
Article in English | MEDLINE | ID: mdl-37689718

ABSTRACT

The spinocerebellar ataxias (SCA) comprise a group of inherited neurodegenerative diseases. SCA3 is the most common form, caused by the expansion of CAG repeats within the ataxin 3 (ATXN3) gene. The mutation results in the expression of an abnormal protein, containing long polyglutamine (polyQ) stretches. The polyQ stretch confers a toxic gain of function and leads to misfolding and aggregation of ATXN3 in neurons. Thus, modulators of ATXN3 expression could potentially ameliorate the pathology in SCA3 patients. Therefore, we generated a CRISPR/Cas9 modified ATXN3-Exon4-Luciferase (ATXN3-LUC) genomic fusion- and control cell lines to perform a reporter cell line-based high-throughput screen comprising 2640 bioactive compounds, including the FDA approved drugs. We found no unequivocal inhibitors of, but identified statins as activators of the LUC signal in the ATXN3-LUC screening cell line. We further confirmed that Simvastatin treatment of wild type SK-N-SH cells increases ATXN3 mRNA and protein levels which likely results from direct binding of the activated sterol regulatory element binding protein 1 (SREBP1) to the ATXN3 promotor. Finally, we observed an increase of normal and expanded ATXN3 protein levels in a patient-derived cell line upon Simvastatin treatment, underscoring the potential medical relevance of our findings.


Subject(s)
Hydroxymethylglutaryl-CoA Reductase Inhibitors , Spinocerebellar Ataxias , Humans , Ataxin-3/genetics , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Neurons , Simvastatin
4.
Neurol Res Pract ; 5(1): 49, 2023 Sep 28.
Article in English | MEDLINE | ID: mdl-37759276

ABSTRACT

Headache is one of the most common neurological manifestations of COVID-19, but it is unclear whether chronic headache as a symptom of Post-COVID-19 is associated with ongoing CNS damage. We compared cerebrospinal fluid (CSF) levels of markers of CNS damage and inflammation in Post-COVID-19 patients with persistent headache to hospitalized acute COVID-19 patients with neurological symptoms and to non-COVID-19 disease-controls. CSF levels of neurofilament light chain, Ubiquitin carboxyl-terminal hydrolase L1 and Tau were similar in patients with persistent headache in post-COVID-19 compared to acute COVID-19 patients and all control groups. Levels of glial fibrillary astrocytic protein were lower in patients with persistent headache in post-COVID-19 compared to some control groups of patients with neurological disease. Therefore, our pilot study of CSF markers indicates that persistent post-COVID-19 headache is not a sign of underlying neuronal damage or glial activation.

5.
Neurol Res Pract ; 4(1): 53, 2022 Oct 31.
Article in English | MEDLINE | ID: mdl-36310154

ABSTRACT

Recent studies have indicated that long-term neurological sequelae after COVID-19 are not accompanied by an increase of canonical biomarkers of central nervous system injury in blood, but subgroup stratifications are lacking. This is a particular concern in chronic headache, which can be a leading symptom of Post-COVID diseases associated with neuronal damage such as vasculitis or autoimmune encephalitis. We here compared patients with mild Post-COVID-19 syndrome and persistent headache (persistent Post-COVID-19 headache) lasting longer than 12 weeks after the initial serological diagnosis, to patients with mild and severe COVID-19 and COVID-19-negative controls. Levels of neurofilament light chain and glial fibrillary astrocytic protein, i.e. markers of neuronal damage and reactive astrogliosis, were lower in blood from patients with persistent Post-COVID-19 headache compared to patients with severe COVID-19. Hence, our pilot serological study indicates that long-term Post-COVID-19 headache may not be a sign of underlying neuronal damage or neuroinflammation.

6.
Acta Neuropathol ; 143(4): 453-469, 2022 04.
Article in English | MEDLINE | ID: mdl-35141810

ABSTRACT

The protein α-synuclein, a key player in Parkinson's disease (PD) and other synucleinopathies, exists in different physiological conformations: cytosolic unfolded aggregation-prone monomers and helical aggregation-resistant multimers. It has been shown that familial PD-associated missense mutations within the α-synuclein gene destabilize the conformer equilibrium of physiologic α-synuclein in favor of unfolded monomers. Here, we characterized the relative levels of unfolded and helical forms of cytosolic α-synuclein in post-mortem human brain tissue and showed that the equilibrium of α-synuclein conformations is destabilized in sporadic PD and DLB patients. This disturbed equilibrium is decreased in a brain region-specific manner in patient samples pointing toward a possible "prion-like" propagation of the underlying pathology and forms distinct disease-specific patterns in the two different synucleinopathies. We are also able to show that a destabilization of multimers mechanistically leads to increased levels of insoluble, pathological α-synuclein, while pharmacological stabilization of multimers leads to a "prion-like" aggregation resistance. Together, our findings suggest that these disease-specific patterns of α-synuclein multimer destabilization in sporadic PD and DLB are caused by both regional neuronal vulnerability and "prion-like" aggregation transmission enabled by the destabilization of local endogenous α-synuclein protein.


Subject(s)
Lewy Body Disease , Parkinson Disease , Prions , Synucleinopathies , Brain/pathology , Humans , Lewy Bodies/pathology , Lewy Body Disease/pathology , Parkinson Disease/pathology , Prions/metabolism , alpha-Synuclein/metabolism
7.
Sci Rep ; 11(1): 19857, 2021 10 06.
Article in English | MEDLINE | ID: mdl-34615900

ABSTRACT

Multiplications, mutations and dysregulation of the alpha synuclein gene (SNCA) are associated with the demise of dopaminergic neurons and are considered to play important roles in the pathogenesis of familial and sporadic forms of Parkinson's disease. Regulation of SNCA expression might thus be an appropriate target for treatment. We aimed to identify specific modulators of SNCA transcription, generated CRISPR/Cas9 modified SNCA-GFP-luciferase (LUC) genomic fusion- and control cell lines and screened a library of 1649 bioactive compounds, including the FDA approved drugs. We found no inhibitors but three selective activators which increased SNCA mRNA and protein levels.


Subject(s)
Drug Discovery , Gene Expression Regulation/drug effects , High-Throughput Screening Assays , alpha-Synuclein/genetics , Cell Line , DNA Methylation , Drug Discovery/methods , Gene Expression , Genes, Reporter , Histones/metabolism , Humans , Small Molecule Libraries , alpha-Synuclein/metabolism
9.
Brain Commun ; 2(1): fcaa010, 2020.
Article in English | MEDLINE | ID: mdl-32280944

ABSTRACT

Since researchers identified α-synuclein as the principal component of Lewy bodies and Lewy neurites, studies have suggested that it plays a causative role in the pathogenesis of dementia with Lewy bodies and other 'synucleinopathies'. While α-synuclein dyshomeostasis likely contributes to the neurodegeneration associated with the synucleinopathies, few direct biochemical analyses of α-synuclein from diseased human brain tissue currently exist. In this study, we analysed sequential protein extracts from a substantial number of patients with neuropathological diagnoses of dementia with Lewy bodies and corresponding controls, detecting a shift of cytosolic and membrane-bound physiological α-synuclein to highly aggregated forms. We then fractionated aqueous extracts (cytosol) from cerebral cortex using non-denaturing methods to search for soluble, disease-associated high molecular weight species potentially associated with toxicity. We applied these fractions and corresponding insoluble fractions containing Lewy-type aggregates to several reporter assays to determine their bioactivity and cytotoxicity. Ultimately, high molecular weight cytosolic fractions enhances phospholipid membrane permeability, while insoluble, Lewy-associated fractions induced morphological changes in the neurites of human stem cell-derived neurons. While the concentrations of soluble, high molecular weight α-synuclein were only slightly elevated in brains of dementia with Lewy bodies patients compared to healthy, age-matched controls, these observations suggest that a small subset of soluble α-synuclein aggregates in the brain may drive early pathogenic effects, while Lewy body-associated α-synuclein can drive neurotoxicity.

10.
Front Neurosci ; 13: 276, 2019.
Article in English | MEDLINE | ID: mdl-31024227

ABSTRACT

Parkinson's disease (PD) is the second most common neurodegenerative disorder next to Alzheimer's disease. Most PD cases are considered to be sporadic and despite considerable scientific effort, the underlying cause(s) still remain(s) enigmatic. In particular, it is unknown to which extent epigenetic alterations contribute to the pathophysiology of this devastating disorder. This is partly due to the fact that appropriate PD models are not yet available. Moreover, epigenetic patterns and mechanisms are species specific and murine systems reflect only a few of the idiosyncrasies of human neurons. For several years now, patient-specific stem cell-derived neural and non-neural cells have been employed to overcome this limitation allowing the analysis and establishment of humanized disease models for PD. Thus, several studies tried to dissect epigenetic alterations such as aberrant DNA methylation or microRNA patterns using lund human mesencephalic cell lines or neurons derived from (patient-specific) induced pluripotent stem cells. These studies demonstrate that human neurons have the potential to be used as model systems for the study of epigenetic modifications in PD such as characterizing epigenetic changes, correlating epigenetic changes to gene expression alterations and hopefully using these insights for the development of novel therapeutics. However, more research is required to define the epigenetic (age-associated) landscape of human in vitro neurons and compare these to native neurons before they can be established as suitable models for epigenetic studies in PD. In this review, we summarize the knowledge about epigenetic studies performed on human neuronal PD models, and we discuss advantages and current limitations of these (stem cell-derived) neuronal models for the study of epigenetic alterations in PD.

11.
Clin Epigenetics ; 10: 13, 2018.
Article in English | MEDLINE | ID: mdl-29422978

ABSTRACT

Background: Genetic predisposition and epigenetic alterations are both considered to contribute to sporadic neurodegenerative diseases (NDDs) such as Parkinson's disease (PD). Since cell reprogramming and the generation of induced pluripotent stem cells (iPSCs) are themselves associated with major epigenetic remodeling, it remains unclear to what extent iPSC-derived neurons lend themselves to model epigenetic disease-associated changes. A key question to be addressed in this context is whether iPSC-derived neurons exhibit epigenetic signatures typically observed in neurons derived from non-reprogrammed human embryonic stem cells (hESCs). Results: Here, we compare mature neurons derived from hESC and isogenic human iPSC generated from hESC-derived neural stem cells. Genome-wide 450 K-based DNA methylation and HT12v4 gene array expression analyses were complemented by a deep analysis of selected genes known to be involved in NDD. Our studies show that DNA methylation and gene expression patterns of isogenic hESC- and iPSC-derived neurons are markedly preserved on a genome-wide and single gene level. Conclusions: Overall, iPSC-derived neurons exhibit similar DNA methylation patterns compared to isogenic hESC-derived neurons. Further studies will be required to explore whether the epigenetic patterns observed in iPSC-derived neurons correspond to those detectable in native brain neurons.


Subject(s)
DNA Methylation , Human Embryonic Stem Cells/cytology , Induced Pluripotent Stem Cells/cytology , Neurons/chemistry , Cell Differentiation , Cells, Cultured , Epigenesis, Genetic , Gene Expression Profiling , Human Embryonic Stem Cells/chemistry , Humans , Induced Pluripotent Stem Cells/chemistry , Oligonucleotide Array Sequence Analysis , Sequence Analysis, DNA , alpha-Synuclein/genetics
12.
Mov Disord Clin Pract ; 4(4): 612-615, 2017.
Article in English | MEDLINE | ID: mdl-30363444

ABSTRACT

BACKGROUND: Spinal cord stimulation (SCS) and deep-brain stimulation reportedly improve refractory orthostatic tremor (OT). No comparative data exist assessing subperceptional versus perceptional SCS with sham stimulation in patients with OT. METHODS: Two patients who had refractory OT were assessed at baseline and 3 months after SCS implantation using 3 different SCS modes: paraesthesia-free burst SCS (40 Hz), sham SCS, and paraesthesia-evoking tonic SCS (100-130 Hz). Surface electromyography, standing time, stimulation parameters, and any adverse events were prospectively recorded. RESULTS: Improved standing time was observed under burst and tonic mode versus stimulation off and compared with baseline in both patients (patient 1: baseline = 22-second; burst SCS [subperceptional]/ standing time = 2.8 minutes; stimulation off/ standing time = 28 seconds; and tonic SCS [perceptional]/ standing time = 1.2 minutes; patient 2: baseline = 47-second; burst SCS [subperceptional]/ standing time = 3.1 minutes; stimulation off/ standing time = 48 seconds; and tonic SCS [perceptional]/ standing time = 1.1 minute). The electromyography frequency demonstrated a decline in tremor frequency. CONCLUSION: Burst as a novel SCS paradigm may be co-considered in patients with refractory OT before more invasive deep-brain stimulation.

13.
Neurobiol Aging ; 36(12): 3334.e7-3334.e11, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26422361

ABSTRACT

DNA methylation patterns change with age, and aging itself is a major confounding risk factor for Parkinson's disease (PD). Duplication and triplication, that is, increased expression of the α-synuclein (SNCA) gene, cause familial PD, and demethylation of SNCA intron 1 has been shown to result in increased expression of SNCA. We thus hypothesized that age-related alterations of SNCA methylation might underly the increased susceptibility toward PD in later life. The present study sought to determine (1) whether alterations of SNCA intron 1 methylation occurred during aging, (2) whether the methylation pattern differed between men and women, and (3) whether purified neurons compared with non-neuronal cells exhibited different methylation patterns. The analysis of DNA from brain tissue and fluorescence activated cell sorting-sorted purified neurons of 41 individuals revealed only a minor increase of SNCA intron 1 DNA methylation levels in presumably healthy individuals during aging but no significant difference between men and women. Interestingly enough, methylation of SNCA intron 1 was higher in neurons compared with non-neuronal cells, although non-neuronal cells express lower levels of SNCA. Therefore, the normal pattern of SNCA methylation during aging should not result in increased expression of α-synuclein protein. It is thus likely that additional, yet not identified, mechanisms contribute to the tissue specificity of SNCA expression and the presumed dysregulation in PD.


Subject(s)
Aging/genetics , DNA Methylation/genetics , Genetic Association Studies , Introns/genetics , alpha-Synuclein/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Brain/metabolism , Child , Child, Preschool , Female , Humans , Infant , Infant, Newborn , Male , Middle Aged , Parkinson Disease/genetics , Risk Factors , Young Adult , alpha-Synuclein/metabolism
14.
Acta Neuropathol ; 124(4): 517-29, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22744790

ABSTRACT

The current classification of human sporadic prion diseases recognizes six major phenotypic subtypes with distinctive clinicopathological features, which largely correlate at the molecular level with the genotype at the polymorphic codon 129 (methionine, M, or valine, V) in the prion protein gene and with the size of the protease-resistant core of the abnormal prion protein, PrP(Sc) (i.e. type 1 migrating at 21 kDa and type 2 at 19 kDa). We previously demonstrated that PrP(Sc) typing by Western blotting is a reliable means of strain typing and disease classification. Limitations of this approach, however, particularly in the interlaboratory setting, are the association of PrP(Sc) types 1 or 2 with more than one clinicopathological phenotype, which precludes definitive case classification if not supported by further analysis, and the difficulty of fully recognizing cases with mixed phenotypic features. In this study, we tested the inter-rater reliability of disease classification based only on histopathological criteria. Slides from 21 cases covering the whole phenotypic spectrum of human sporadic prion diseases, and also including two cases of variant Creutzfeldt-Jakob disease (CJD), were distributed blindly to 13 assessors for classification according to given instructions. The results showed good-to-excellent agreement between assessors in the classification of cases. In particular, there was full agreement (100 %) for the two most common sporadic CJD subtypes and variant CJD, and very high concordance in general for all pure phenotypes and the most common subtype with mixed phenotypic features. The present data fully support the basis for the current classification of sporadic human prion diseases and indicate that, besides molecular PrP(Sc) typing, histopathological analysis permits reliable disease classification with high interlaboratory accuracy.


Subject(s)
Prion Diseases/classification , Prion Diseases/pathology , Consensus , Europe , Humans , Immunohistochemistry , Observer Variation , Phenotype , Prion Diseases/genetics , Reproducibility of Results , United States
15.
Am J Physiol Endocrinol Metab ; 302(9): E1044-54, 2012 May 15.
Article in English | MEDLINE | ID: mdl-22318954

ABSTRACT

This study aimed to elucidate the role of the AT(2) receptor (AT(2)R), which is expressed and upregulated in the adrenal zona glomerulosa (ZG) under conditions of increased aldosterone production. We developed a novel transgenic rat (TGR; TGRCXmAT(2)R) that overexpresses the AT(2)R in the adrenal gland, heart, kidney, brain, skeletal muscle, testes, lung, spleen, aorta, and vein. As a consequence the total angiotensin II (Ang II) binding sites increased 7.8-fold in the kidney, 25-fold in the heart, and twofold in the adrenals. The AT(2)R number amounted to 82-98% of total Ang II binding sites. In the ZG of TGRCXmAT(2)R, the AT(2)R density was elevated threefold relative to wild-type (WT) littermates, whereas AT(1)R density remained unchanged. TGRCXmAT(2)R rats were viable and exhibited normal reproduction, blood pressure, and kidney function. Notably, a slightly but significantly reduced body weight and a moderate increase in plasma urea were observed. With respect to adrenal function, 24-h urinary and plasma aldosterone concentrations were unaffected in TGRCXmAT(2)R at baseline. Three and 14 days of Ang II infusion (300 ng·min(-1)·kg(-1)) increased plasma aldosterone levels in WT and in TGR. These changes were completely abolished by the AT(1)R blocker losartan. Of note, glomerulosa cell proliferation, as indicated by the number of Ki-67-positive glomerulosa cells, was stimulated by Ang II in TGR and WT rats; however, this increase was significantly attenuated in TGR overexpressing the AT(2)R. In conclusion, AT(2)R in the adrenal ZG inhibits Ang II-induced cell proliferation but has no obvious lasting effect on the regulation of the aldosterone production at the investigated stages.


Subject(s)
Aldosterone/physiology , Models, Animal , Rats, Transgenic , Receptor, Angiotensin, Type 2/metabolism , Zona Glomerulosa/physiology , Angiotensin II/physiology , Animals , Cell Proliferation , Gene Expression Regulation/physiology , Rats , Up-Regulation , Zona Glomerulosa/cytology
SELECTION OF CITATIONS
SEARCH DETAIL