Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
EMBO J ; 42(23): e111122, 2023 Dec 01.
Article in English | MEDLINE | ID: mdl-37916890

ABSTRACT

Alpha-synuclein (aSN) is a membrane-associated and intrinsically disordered protein, well known for pathological aggregation in neurodegeneration. However, the physiological function of aSN is disputed. Pull-down experiments have pointed to plasma membrane Ca2+ -ATPase (PMCA) as a potential interaction partner. From proximity ligation assays, we find that aSN and PMCA colocalize at neuronal synapses, and we show that calcium expulsion is activated by aSN and PMCA. We further show that soluble, monomeric aSN activates PMCA at par with calmodulin, but independent of the autoinhibitory domain of PMCA, and highly dependent on acidic phospholipids and membrane-anchoring properties of aSN. On PMCA, the key site is mapped to the acidic lipid-binding site, located within a disordered PMCA-specific loop connecting the cytosolic A domain and transmembrane segment 3. Our studies point toward a novel physiological role of monomeric aSN as a stimulator of calcium clearance in neurons through activation of PMCA.


Subject(s)
Calcium , alpha-Synuclein , Calcium/metabolism , alpha-Synuclein/genetics , alpha-Synuclein/metabolism , Plasma Membrane Calcium-Transporting ATPases/genetics , Plasma Membrane Calcium-Transporting ATPases/chemistry , Plasma Membrane Calcium-Transporting ATPases/metabolism , Cell Membrane/metabolism , Adenosine Triphosphatases/metabolism , Binding Sites
2.
APMIS ; 131(12): 687-697, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37750005

ABSTRACT

Mesenchymal stromal/stem cell (MSC) therapy has been thoroughly tested in preclinical animal models and holds great promise for the treatment of kidney diseases. It is becoming increasingly evident that the efficacy of MSC therapy is dependent on several factors including dosage, the tissue source of MSCs, the route of delivery and timing of administration. In a time where MSC therapy is moving from preclinical research to clinically therapeutic use, the importance of choice of delivery method, modality, and administration route increases. In this review, we provide an overview of the different MSC delivery routes used in preclinical kidney disease models, highlight the recent advances in the field, and summarize studies comparing delivery routes of MSCs to the kidney.


Subject(s)
Kidney Diseases , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Animals , Disease Models, Animal , Mesenchymal Stem Cell Transplantation/methods , Kidney , Kidney Diseases/therapy
3.
PLoS One ; 16(10): e0252635, 2021.
Article in English | MEDLINE | ID: mdl-34613964

ABSTRACT

Accumulation of aggregated alpha-synuclein (α-syn) is believed to play a pivotal role in the pathophysiology of Parkinson's disease (PD) and other synucleinopathies. As a key constituent of Lewy pathology, more than 90% of α-syn in Lewy bodies is phosphorylated at serine-129 (pS129) and hence, it is used extensively as a marker for α-syn pathology. However, the exact role of pS129 remains controversial and the kinase(s) responsible for the phosphorylation have yet to be determined. In this study, we investigated the effect of Polo-like kinase 2 (PLK2) inhibition on formation of pS129 using an ex vivo organotypic brain slice model of synucleinopathy. Our data demonstrated that PLK2 inhibition has no effect on α-syn aggregation, pS129 or inter-neuronal spreading of the aggregated α-syn seen in the organotypic slices. Instead, PLK2 inhibition reduced the soluble pS129 level in the nuclei. The same finding was replicated in an in vivo mouse model of templated α-syn aggregation and in human dopaminergic neurons, suggesting that PLK2 is more likely to be involved in S129-phosphorylation of the soluble physiological fraction of α-syn. We also demonstrated that reduction of nuclear pS129 following PLK2 inhibition for a short time before sample collection improves the signal-to-noise ratio when quantifying pS129 aggregate pathology.


Subject(s)
Phosphorylation/physiology , Protein Aggregates/physiology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Serine/metabolism , alpha-Synuclein/metabolism , Animals , Brain/metabolism , Dopamine/metabolism , Lewy Bodies/metabolism , Mice , Mice, Inbred C57BL , Neurons/metabolism , Parkinson Disease/metabolism
4.
J Neuroinflammation ; 18(1): 177, 2021 Aug 16.
Article in English | MEDLINE | ID: mdl-34399786

ABSTRACT

BACKGROUND: Synucleinopathies are characterized by neurodegeneration and deposition of the presynaptic protein α-synuclein in pathological protein inclusions. Growing evidence suggests the complement system not only has physiological functions in the central nervous system, but also is involved in mediating the pathological loss of synapses in Alzheimer's disease. However, it is not established whether the complement system has a similar role in the diseases Parkinson's disease, Dementia with Lewy bodies, and multiple system atrophy (MSA) that are associated with α-synuclein aggregate pathology. METHODS: To investigate if the complement system has a pathological role in synucleinopathies, we assessed the effect of the complement system on the viability of an α-synuclein expressing cell model and examined direct activation of the complement system by α-synuclein in a plate-based activation assay. Finally, we investigated the levels of the initiator of the classical pathway, C1q, in postmortem brain samples from MSA patients. RESULTS: We demonstrate that α-synuclein activates the classical complement pathway and mediates complement-dependent toxicity in α-synuclein expressing SH-SY5Y cells. The α-synuclein-dependent cellular toxicity was rescued by the complement inhibitors RaCI (inhibiting C5) and Cp20 (inhibiting C3). Furthermore, we observed a trend for higher levels of C1q in the putamen of MSA subjects than that of controls. CONCLUSION: α-Synuclein can activate the classical complement pathway, and the complement system is involved in α-synuclein-dependent cellular cytotoxicity suggesting the system could play a prodegenerative role in synucleinopathies.


Subject(s)
Complement Pathway, Classical/physiology , Inclusion Bodies/metabolism , Visual Cortex/metabolism , alpha-Synuclein/metabolism , Adult , Aged , Aged, 80 and over , Cell Line, Tumor , Female , Humans , Inclusion Bodies/pathology , Male , Middle Aged , Visual Cortex/pathology
5.
Acta Neuropathol ; 142(1): 87-115, 2021 07.
Article in English | MEDLINE | ID: mdl-33978813

ABSTRACT

Pathology consisting of intracellular aggregates of alpha-Synuclein (α-Syn) spread through the nervous system in a variety of neurodegenerative disorders including Parkinson's disease, dementia with Lewy bodies, and multiple system atrophy. The discovery of structurally distinct α-Syn polymorphs, so-called strains, supports a hypothesis where strain-specific structures are templated into aggregates formed by native α-Syn. These distinct strains are hypothesised to dictate the spreading of pathology in the tissue and the cellular impact of the aggregates, thereby contributing to the variety of clinical phenotypes. Here, we present evidence of a novel α-Syn strain induced by the multiple system atrophy-associated oligodendroglial protein p25α. Using an array of biophysical, biochemical, cellular, and in vivo analyses, we demonstrate that compared to α-Syn alone, a substoichiometric concentration of p25α redirects α-Syn aggregation into a unique α-Syn/p25α strain with a different structure and enhanced in vivo prodegenerative properties. The α-Syn/p25α strain induced larger inclusions in human dopaminergic neurons. In vivo, intramuscular injection of preformed fibrils (PFF) of the α-Syn/p25α strain compared to α-Syn PFF resulted in a shortened life span and a distinct anatomical distribution of inclusion pathology in the brain of a human A53T transgenic (line M83) mouse. Investigation of α-Syn aggregates in brain stem extracts of end-stage mice demonstrated that the more aggressive phenotype of the α-Syn/p25α strain was associated with an increased load of α-Syn aggregates based on a Förster resonance energy transfer immunoassay and a reduced α-Syn aggregate seeding activity based on a protein misfolding cyclic amplification assay. When injected unilaterally into the striata of wild-type mice, the α-Syn/p25α strain resulted in a more-pronounced motoric phenotype than α-Syn PFF and exhibited a "tropism" for nigro-striatal neurons compared to α-Syn PFF. Overall, our data support a hypothesis whereby oligodendroglial p25α is responsible for generating a highly prodegenerative α-Syn strain in multiple system atrophy.


Subject(s)
Multiple System Atrophy/genetics , Neurodegenerative Diseases/genetics , Synucleinopathies/pathology , alpha-Synuclein/genetics , Animals , Cell Line , Humans , Inclusion Bodies/pathology , Mice , Mice, Transgenic , Multiple System Atrophy/pathology , Nerve Tissue Proteins/genetics , Oligodendroglia/metabolism , Protein Conformation , Proteostasis Deficiencies/genetics , Substantia Nigra/pathology , alpha-Synuclein/toxicity
6.
J Neurochem ; 158(4): 960-979, 2021 08.
Article in English | MEDLINE | ID: mdl-33991113

ABSTRACT

In Parkinson's disease, dopamine-containing nigrostriatal neurons undergo profound degeneration. Tyrosine hydroxylase (TH) is the rate-limiting enzyme in dopamine biosynthesis. TH increases in vitro formation of reactive oxygen species, and previous animal studies have reported links between cytosolic dopamine build-up and oxidative stress. To examine effects of increased TH activity in catecholaminergic neurons in vivo, we generated TH-over-expressing mice (TH-HI) using a BAC-transgenic approach that results in over-expression of TH with endogenous patterns of expression. The transgenic mice were characterized by western blot, qPCR, and immunohistochemistry. Tissue contents of dopamine, its metabolites, and markers of oxidative stress were evaluated. TH-HI mice had a 3-fold increase in total and phosphorylated TH levels and an increased rate of dopamine synthesis. Coincident with elevated dopamine turnover, TH-HI mice showed increased striatal production of H2 O2 and reduced glutathione levels. In addition, TH-HI mice had elevated striatal levels of the neurotoxic dopamine metabolites 3,4-dihydroxyphenylacetaldehyde and 5-S-cysteinyl-dopamine and were more susceptible than wild-type mice to the effects of amphetamine and methamphetamine. These results demonstrate that increased TH alone is sufficient to produce oxidative stress in vivo, build up autotoxic dopamine metabolites, and augment toxicity.


Subject(s)
Amphetamine/pharmacology , Catecholamines/metabolism , Central Nervous System Stimulants/pharmacology , Oxidative Stress , Tyrosine 3-Monooxygenase/metabolism , 3,4-Dihydroxyphenylacetic Acid/analogs & derivatives , 3,4-Dihydroxyphenylacetic Acid/metabolism , Animals , Dopamine/analogs & derivatives , Dopamine/metabolism , Female , Gene Dosage , Glutathione/metabolism , Humans , Hydrogen Peroxide/metabolism , Male , Mice , Mice, Transgenic , Neurons/drug effects , Tyrosine 3-Monooxygenase/genetics
7.
PLoS One ; 14(10): e0216764, 2019.
Article in English | MEDLINE | ID: mdl-31603909

ABSTRACT

The group of neurodegenerative diseases, Parkinson's disease (PD), dementia with Lewy bodies (DLB), and multiple system atrophy (MSA) all exhibit inclusions containing amyloid-type α-synuclein (α-syn) aggregates within degenerating brain cells. α-syn also exists as soluble oligomeric species that are hypothesized to represent intermediates between its native and aggregated states. These oligomers are present in brain extracts from patients suffering from synucleinopathies and hold great potential as biomarkers. Although easily prepared in vitro, oligomers are metastable and dissociate over time, thereby complicating α-syn oligomer research. Using the small amine-reactive cross-linker, formaldehyde (FA), we successfully stabilized α-syn oligomers without affecting their size, overall structure or antigenicity towards aggregate-conformation specific α-syn antibodies FILA and MJFR-14-6-4-2. Further, cross-linked α-syn oligomers show resistance towards denaturant like urea and SDS treatment and remain fully functional as internal standard in an aggregation-specific enzyme-linked immunosorbent assay (ELISA) despite prior incubation with urea. We propose that FA cross-linked α-syn oligomers could serve as important calibrators to facilitate comparative and standardized α-syn biomarker studies going forward.


Subject(s)
Amyloid/chemistry , Formaldehyde/chemistry , Protein Multimerization , alpha-Synuclein/chemistry , Amyloid/metabolism , Humans , Lewy Body Disease/metabolism , Multiple System Atrophy/metabolism , Parkinson Disease/metabolism , Protein Stability , alpha-Synuclein/metabolism
8.
PLoS One ; 13(4): e0196056, 2018.
Article in English | MEDLINE | ID: mdl-29698510

ABSTRACT

Soluble aggregates of α-synuclein, so-called oligomers, are hypothesized to act as neurotoxic species in Parkinson's disease, Lewy body dementia and multiple systems atrophy, but specific tools to detect these aggregated species are only slowly appearing. We have developed an α-synuclein oligomer ELISA that allows us to detect and compare α-synuclein oligomer levels in different in vivo and in vitro experiments. The ELISA is based on commercially available antibodies and the epitope of the capture antibody MJF14-6-4-2 is folding- and aggregate-dependent and not present on monomers.


Subject(s)
Enzyme-Linked Immunosorbent Assay/methods , Parkinson Disease/pathology , Protein Aggregates/immunology , alpha-Synuclein/analysis , Animals , Antibodies, Monoclonal/immunology , Disease Models, Animal , Epitopes/immunology , Male , Mice , Mice, Transgenic , Parkinson Disease/metabolism , alpha-Synuclein/immunology , alpha-Synuclein/metabolism
9.
EMBO Rep ; 19(5)2018 05.
Article in English | MEDLINE | ID: mdl-29599149

ABSTRACT

Aggregation of α-synuclein is a hallmark of Parkinson's disease and dementia with Lewy bodies. We here investigate the relationship between cytosolic Ca2+ and α-synuclein aggregation. Analyses of cell lines and primary culture models of α-synuclein cytopathology reveal an early phase with reduced cytosolic Ca2+ levels followed by a later Ca2+ increase. Aggregated but not monomeric α-synuclein binds to and activates SERCA in vitro, and proximity ligation assays confirm this interaction in cells. The SERCA inhibitor cyclopiazonic acid (CPA) normalises both the initial reduction and the later increase in cytosolic Ca2+ CPA protects the cells against α-synuclein-aggregate stress and improves viability in cell models and in Caenorhabditis elegans in vivo Proximity ligation assays also reveal an increased interaction between α-synuclein aggregates and SERCA in human brains affected by dementia with Lewy bodies. We conclude that α-synuclein aggregates bind SERCA and stimulate its activity. Reducing SERCA activity is neuroprotective, indicating that SERCA and down-stream processes may be therapeutic targets for treating α-synucleinopathies.


Subject(s)
Calcium/chemistry , Calcium/metabolism , Cytosol/chemistry , Protein Aggregates , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , alpha-Synuclein/metabolism , Animals , Brain/pathology , Caenorhabditis elegans , Cell Line , Cells, Cultured , Endoplasmic Reticulum/metabolism , Humans , Indoles/pharmacology , Lewy Bodies , Male , Mice , Parkinson Disease/pathology , Rats , Sarcoplasmic Reticulum Calcium-Transporting ATPases/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...