Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
J Proteome Res ; 22(6): 1695-1711, 2023 06 02.
Article in English | MEDLINE | ID: mdl-37158322

ABSTRACT

The proteogenomic search pipeline developed in this work has been applied for reanalysis of 40 publicly available shotgun proteomic datasets from various human tissues comprising more than 8000 individual LC-MS/MS runs, of which 5442 .raw data files were processed in total. This reanalysis was focused on searching for ADAR-mediated RNA editing events, their clustering across samples of different origins, and classification. In total, 33 recoded protein sites were identified in 21 datasets. Of those, 18 sites were detected in at least two datasets, representing the core human protein editome. In agreement with prior artworks, neural and cancer tissues were found to be enriched with recoded proteins. Quantitative analysis indicated that recoding the rate of specific sites did not directly depend on the levels of ADAR enzymes or targeted proteins themselves, rather it was governed by differential and yet undescribed regulation of interaction of enzymes with mRNA. Nine recoding sites conservative between humans and rodents were validated by targeted proteomics using stable isotope standards in the murine brain cortex and cerebellum, and an additional one was validated in human cerebrospinal fluid. In addition to previous data of the same type from cancer proteomes, we provide a comprehensive catalog of recoding events caused by ADAR RNA editing in the human proteome.


Subject(s)
Proteogenomics , Proteomics , Humans , Animals , Mice , RNA/metabolism , RNA Editing , Chromatography, Liquid , Tandem Mass Spectrometry , Proteome/genetics , Proteome/metabolism , Adenosine/metabolism , Inosine/genetics , Inosine/metabolism
2.
Biomedicines ; 11(1)2022 Dec 27.
Article in English | MEDLINE | ID: mdl-36672569

ABSTRACT

Dysregulation of intraocular pressure (IOP) is one of the main risk factors for glaucoma. γ-synuclein is a member of the synuclein family of widely expressed synaptic proteins within the central nervous system that are implicated in certain types of neurodegeneration. γ-synuclein expression and localization changes in the retina and optic nerve of patients with glaucoma. However, the mechanisms by which γ-synuclein could contribute to glaucoma are poorly understood. We assessed the presence of autoantibodies to γ-synuclein in the blood serum of patients with primary open-angle glaucoma (POAG) by immunoblotting. A positive reaction was detected for five out of 25 patients (20%) with POAG. Autoantibodies to γ-synuclein were not detected in a group of patients without glaucoma. We studied the dynamics of IOP in response to IOP regulators in knockout mice (γ-KO) to understand a possible link between γ-synuclein dysfunction and glaucoma-related pathophysiological changes. The most prominent decrease of IOP in γ-KO mice was observed after the instillation of 1% phenylephrine and 10% dopamine. The total protein concentration in tear fluid of γ-KO mice was approximately two times higher than that of wild-type mice, and the activity of neurodegeneration-linked protein α2-macroglobulin was reduced. Therefore, γ-synuclein dysfunction contributes to pathological processes in glaucoma, including dysregulation of IOP.

3.
Mol Brain ; 13(1): 75, 2020 05 11.
Article in English | MEDLINE | ID: mdl-32393371

ABSTRACT

Previous studies of the alpha-synuclein null mutant mice on the C57Bl6 genetic background have revealed reduced number of dopaminergic neurons in their substantia nigra pars compacta (SNpc). However, the presence in genomes of the studied mouse lines of additional genetic modifications that affect expression of genes located in a close proximity to the alpha-synuclein-encoding Snca gene makes these data open to various interpretations. To unambiguously demonstrate that the absence of alpha-synuclein is the primary cause of the observed deficit of dopaminergic neurons, we employed a recently produced constituent alpha-synuclein knockout mouse line B6(Cg)-Sncatm1.2Vlb/J. The only modification introduced to the genome of these mice is a substitution of the first coding exon and adjusted short intronic fragments of the Snca gene by a single loxP site. We compared the number of dopaminergic neurons in the SNpc of this line, previously studied B6(Cg)-Sncatm1Rosl/J line and wild type littermate mice. A similar decrease was observed in both knockout lines when compared with wild type mice. In a recently published study we revealed no loss of dopaminergic neurons following conditional inactivation of the Snca gene in neurons of adult mice. Taken together, these results strongly suggest that alpha-synuclein is required for efficient survival or maturation of dopaminergic neurons in the developing SNpc but is dispensable for survival of mature SNpc dopaminergic neurons.


Subject(s)
Dopaminergic Neurons/metabolism , Pars Compacta/metabolism , Substantia Nigra/metabolism , alpha-Synuclein/metabolism , Animals , Cell Survival/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Pars Compacta/growth & development , Substantia Nigra/cytology , Substantia Nigra/growth & development , alpha-Synuclein/genetics
4.
Neurobiol Aging ; 91: 76-87, 2020 07.
Article in English | MEDLINE | ID: mdl-32224067

ABSTRACT

The etiology and pathogenesis of Parkinson's disease (PD) are tightly linked to the gain-of-function of α-synuclein. However, gradual accumulation of α-synuclein aggregates in dopaminergic neurons of substantia nigra pars compacta (SNpc) leads to the depletion of the functional pool of soluble α-synuclein, and therefore, creates loss-of-function conditions, particularly in presynaptic terminals of these neurons. Studies of how this late-onset depletion of a protein involved in many important steps of neurotransmission contributes to PD progression and particularly, to worsening the nigrostriatal pathology at late stages of the disease are limited and obtained data, are controversial. Recently, we produced a mouse line for conditional knockout of the gene encoding α-synuclein, and here we used its tamoxifen-inducible pan-neuronal inactivation to study consequences of the adult-onset (from the age of 6 months) and late-onset (from the age of 12 months) α-synuclein depletion to the nigrostriatal system. No significant changes of animal balance/coordination, the number of dopaminergic neurons in the SNpc and the content of dopamine and its metabolites in the striatum were observed after adult-onset α-synuclein depletion, but in aging (18-month-old) late-onset depleted mice we found a significant reduction of major dopamine metabolites without changes to the content of dopamine itself. Our data suggest that this might be caused, at least partially, by reduced expression of aldehyde dehydrogenase ALDH1a1 and could lead to the accumulation of toxic intermediates of dopamine catabolism. By extrapolating our findings to a potential clinical situation, we suggest that therapeutic downregulation of α-synuclein expression in PD patients is a generally safe option as it should not cause adverse side effects on the functionality of their nigrostriatal system. However, if started in aged patients, this type of therapy might trigger slight functional changes of the nigrostriatal system with potentially unwanted additive effect to already existing pathology.


Subject(s)
Aging/genetics , Aging/pathology , Corpus Striatum/metabolism , Corpus Striatum/pathology , Gene Knockout Techniques , Parkinson Disease/etiology , Parkinson Disease/genetics , Substantia Nigra/metabolism , Substantia Nigra/pathology , alpha-Synuclein/genetics , alpha-Synuclein/metabolism , Aldehyde Dehydrogenase 1 Family/genetics , Aldehyde Dehydrogenase 1 Family/metabolism , Animals , Disease Models, Animal , Dopamine/metabolism , Down-Regulation , Gene Expression/genetics , Mice, Inbred C57BL , Mice, Knockout , Molecular Targeted Therapy , Parkinson Disease/therapy , Retinal Dehydrogenase/genetics , Retinal Dehydrogenase/metabolism , Synaptic Transmission/genetics
5.
Genes Brain Behav ; 18(8): e12607, 2019 11.
Article in English | MEDLINE | ID: mdl-31437340

ABSTRACT

Multiple clinical and experimental evidences suggest that amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration (FTLD) are members of a disease continuum. Pathological inclusions of fused in sarcoma (FUS) protein have been observed in subsets of patients with these diseases but their anatomical distribution is different for two diseases. These structures are present in motor neurons in ALS cases but in cortical neurons in FTLD cases. Expression of a C-terminally truncated form of human FUS causes an early onset and progressive motor neuron pathology in transgenic mice but only when these neurons express a certain level of this protein. Severe motor dysfunction and early lethality of mice with expression above this level prevent their use for studies of FTLD-related pathology caused by expression of this form of FUS. In the present study, we used another line of mice expressing the same protein but not developing any signs of motor system dysfunction due to substantially lower level of transgene expression in motor neurons. In a set of tests 5-month old mice displayed certain behavioural abnormalities, including increased impulsivity, decreased anxiety and compromised social interaction, which recapitulate behaviour characteristics typically seen in FTLD patients.


Subject(s)
Behavior, Animal , Frontotemporal Dementia/genetics , RNA-Binding Protein FUS/genetics , Animals , Conditioning, Classical , Frontal Lobe/metabolism , Frontal Lobe/pathology , Humans , Male , Mice , Mice, Inbred C57BL , Motor Neurons/metabolism , Movement , Social Behavior , Transgenes
6.
PeerJ ; 6: e4779, 2018.
Article in English | MEDLINE | ID: mdl-29785351

ABSTRACT

Lesion of the dopaminergic neurons of the nigrostriatal system is a key feature of Parkinson's disease (PD). Alpha-synuclein is a protein that is a major component of Lewy bodies, histopathological hallmarks of PD, and is involved in regulation of dopamine (DA) neurotransmission. Previous studies of knockout mice have shown that inactivation of alpha-synuclein gene can lead to the reduction in number of DA neurons in the substantia nigra (SN). DA neurons of the SN are known to be the most affected in PD patients whereas DA neurons of neighboring ventral tegmental area (VTA) are much less susceptible to degeneration. Here we have studied the dynamics of changes in TH-positive cell numbers in the SN and VTA during a critical period of their embryonic development in alpha-synuclein knockout mice. This precise study of DA neurons during development of the SN revealed that not only is the number of DA neurons reduced by the end of the period of ontogenic selection, but that the way these neurons are formed is altered in alpha-synuclein knockout mice. At the same time, DA neurons in the VTA are not affected. Alpha-synuclein exerts a modulating effect on the formation of DA neurons in the SN and has no effect on the formation of DA neurons in VTA, the structure that is much less susceptible to degeneration in a brain with PD, suggesting a potential role of alpha-synuclein in the development of the population of DA neurons in substantia nigra.

7.
Neurotox Res ; 29(4): 551-7, 2016 May.
Article in English | MEDLINE | ID: mdl-26842600

ABSTRACT

Intracerebral or intraperitoneal injections of brain extracts from the Alzheimer's disease patients result in the acceleration of cerebral ß-amyloidosis in transgenic mice. Earlier, we have found that intravenous injections of synthetic full-length amyloid-ß (Aß) comprising the isomerized Asp7 trigger cerebral ß-amyloidosis. In vitro studies have shown that isomerization of Asp7 promotes zinc-induced oligomerization of the Aß metal-binding domain (Aß1-16). Here we report that single intracerebral injection of the peptide Aß1-16 with isomerized Asp7 (isoAß1-16) but not the injection of Aß1-16 significantly increases amyloid burden in 5XFAD transgenic mice. Our results provide evidence for a role of isoAß1-16 as a minimal seeding agent of Aß aggregation in vivo.


Subject(s)
Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/pharmacology , Amyloidosis/chemically induced , Aspartic Acid/metabolism , Peptide Fragments/pharmacology , Plaque, Amyloid/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Amyloid beta-Peptides/administration & dosage , Amyloid beta-Protein Precursor/genetics , Amyloidosis/genetics , Analysis of Variance , Animals , Disease Models, Animal , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation/genetics , Peptide Fragments/administration & dosage , Presenilin-1/genetics
8.
Article in English | MEDLINE | ID: mdl-25991062

ABSTRACT

Mutations to the RNA binding protein, fused in sarcoma (FUS) occur in ∼5% of familial ALS and FUS-positive cytoplasmic inclusions are commonly observed in these patients. Altered RNA metabolism is increasingly implicated in ALS, yet it is not understood how the specificity with which FUS interacts with RNA in the cytoplasm can affect its aggregation in vivo. To further understand this, we expressed, in mice, a form of FUS (FUS ΔRRMcyt) that lacked the RNA recognition motif (RRM), thought to impart specificity to FUS-RNA interactions, and carried an ALS-associated point mutation, R522G, retaining the protein in the cytoplasm. Here we report the phenotype and results of histological assessment of the brain of transgenic mice expressing this isoform of FUS. Results demonstrated that neuronal expression of FUS ΔRRMcyt caused early lethality often preceded by severe tremor. Large FUS-positive cytoplasmic inclusions were found in many brain neurons; however, neither neuronal loss nor neuroinflammatory response was observed. In conclusion, the extensive FUS proteinopathy and severe phenotype of these mice suggests that affecting the interactions of FUS with RNA in vivo may augment its aggregation in the neuronal cytoplasm and the severity of disease processes.


Subject(s)
Cytoplasm/genetics , Lethargy/genetics , Neurons/pathology , RNA-Binding Protein FUS/genetics , Sequence Deletion , Amino Acid Motifs , Animals , Brain/pathology , Disease Models, Animal , Disease Progression , Glial Fibrillary Acidic Protein/metabolism , Inclusion Bodies/metabolism , Inclusion Bodies/pathology , Lethargy/complications , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurons/metabolism , Phosphopyruvate Hydratase/metabolism , RNA-Binding Protein FUS/metabolism , Tremor/genetics , Tremor/pathology , Tremor/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL
...