Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Int J Mol Sci ; 25(15)2024 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-39125972

RESUMO

In this work, we explored the intrinsic disorder status of the three members of the synuclein family of proteins-α-, ß-, and γ-synucleins-and showed that although all three human synucleins are highly disordered, the highest levels of disorder are observed in γ-synuclein. Our analysis of the peculiarities of the amino acid sequences and modeled 3D structures of the human synuclein family members revealed that the pathological mutations A30P, E46K, H50Q, A53T, and A53E associated with the early onset of Parkinson's disease caused some increase in the local disorder propensity of human α-synuclein. A comparative sequence-based analysis of the synuclein proteins from various evolutionary distant species and evaluation of their levels of intrinsic disorder using a set of commonly used bioinformatics tools revealed that, irrespective of their origin, all members of the synuclein family analyzed in this study were predicted to be highly disordered proteins, indicating that their intrinsically disordered nature represents an evolutionary conserved and therefore functionally important feature. A detailed functional disorder analysis of the proteins in the interactomes of the human synuclein family members utilizing a set of commonly used disorder analysis tools showed that the human α-synuclein interactome has relatively higher levels of intrinsic disorder as compared with the interactomes of human ß- and γ- synucleins and revealed that, relative to the ß- and γ-synuclein interactomes, α-synuclein interactors are involved in a much broader spectrum of highly diversified functional pathways. Although proteins interacting with three human synucleins were characterized by highly diversified functionalities, this analysis also revealed that the interactors of three human synucleins were involved in three common functional pathways, such as the synaptic vesicle cycle, serotonergic synapse, and retrograde endocannabinoid signaling. Taken together, these observations highlight the critical importance of the intrinsic disorder of human synucleins and their interactors in various neuronal processes.


Assuntos
alfa-Sinucleína , Humanos , alfa-Sinucleína/metabolismo , alfa-Sinucleína/química , alfa-Sinucleína/genética , Proteínas Intrinsicamente Desordenadas/metabolismo , Proteínas Intrinsicamente Desordenadas/química , Proteínas Intrinsicamente Desordenadas/genética , Sequência de Aminoácidos , beta-Sinucleína/metabolismo , beta-Sinucleína/genética , beta-Sinucleína/química , gama-Sinucleína/metabolismo , gama-Sinucleína/genética , Doença de Parkinson/metabolismo , Doença de Parkinson/genética , Sinucleínas/metabolismo , Sinucleínas/genética , Modelos Moleculares , Mutação
2.
Alzheimers Dement ; 19(11): 4896-4907, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37052206

RESUMO

INTRODUCTION: ß-synuclein is an emerging blood biomarker to study synaptic degeneration in Alzheimer´s disease (AD), but its relation to amyloid-ß (Αß) pathology is unclear. METHODS: We investigated the association of plasma ß-synuclein levels with [18F] flutemetamol positron emission tomography (PET) in patients with AD dementia (n = 51), mild cognitive impairment (MCI-Aß+ n = 18, MCI- Aß- n = 30), non-AD dementias (n = 22), and non-demented controls (n = 5). RESULTS: Plasma ß-synuclein levels were higher in Aß+ (AD dementia, MCI-Aß+) than in Aß- subjects (non-AD dementias, MCI-Aß-) with good discrimination of Aß+ from Aß- subjects and prediction of Aß status in MCI individuals. A positive correlation between plasma ß-synuclein and Aß PET was observed in multiple cortical regions across all lobes. DISCUSSION: Plasma ß-synuclein demonstrated discriminative properties for Aß PET positive and negative subjects. Our data underline that ß-synuclein is not a direct marker of Aß pathology and suggest different longitudinal dynamics of synaptic degeneration versus amyloid deposition across the AD continuum. HIGHLIGHTS: Blood and CSF ß-synuclein levels are higher in Aß+ than in Aß- subjects. Blood ß-synuclein level correlates with amyloid PET positivity in multiple regions. Blood ß-synuclein predicts Aß status in MCI individuals.


Assuntos
Doença de Alzheimer , Disfunção Cognitiva , Humanos , beta-Sinucleína , Encéfalo/patologia , Doença de Alzheimer/diagnóstico por imagem , Doença de Alzheimer/patologia , Tomografia por Emissão de Pósitrons/métodos , Peptídeos beta-Amiloides/metabolismo , Disfunção Cognitiva/diagnóstico por imagem , Disfunção Cognitiva/patologia , Biomarcadores
3.
Alzheimers Dement ; 19(11): 5095-5102, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37186338

RESUMO

INTRODUCTION: ß-Synuclein is an emerging synaptic blood biomarker for Alzheimer's disease (AD) but differences in ß-synuclein levels in preclinical AD and its association with amyloid and tau pathology have not yet been studied. METHODS: We measured plasma ß-synuclein levels in cognitively unimpaired individuals with positive Aß-PET (i.e., preclinical AD, N = 48) or negative Aß-PET (N = 61), Aß-positive patients with mild cognitive impairment (MCI, N = 36), and Aß-positive AD dementia (N = 85). Amyloid (A) and tau (T) pathology were assessed by [18 F]flutemetamol and [18 F]RO948 PET. RESULTS: Plasma ß-synuclein levels were higher in preclinical AD and even higher in MCI and AD dementia. Stratification according to amyloid/tau pathology revealed higher ß-synuclein in A+ T- and A+ T+ subjects compared with A- T- . Plasma ß-synuclein levels were related to tau and Aß pathology and associated with temporal cortical thinning and cognitive impairment. DISCUSSION: Our data indicate that plasma ß-synuclein might track synaptic dysfunction, even during the preclinical stages of AD. HIGHLIGHTS: Plasma ß-synuclein is already higher in preclinical AD. Plasma ß-synuclein is higher in MCI and AD dementia than in preclinical AD. Aß- and tau-PET SUVRs are associated with plasma ß-synuclein levels. Plasma ß-synuclein is already higher in tau-PET negative subjects. Plasma ß-synuclein is related to temporal cortical atrophy and cognitive impairment.


Assuntos
Doença de Alzheimer , Disfunção Cognitiva , Humanos , Doença de Alzheimer/patologia , Proteínas tau , Peptídeos beta-Amiloides , beta-Sinucleína , Disfunção Cognitiva/patologia , Biomarcadores , Amiloide , Proteínas Amiloidogênicas , Tomografia por Emissão de Pósitrons
4.
J Biol Chem ; 296: 100271, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33428933

RESUMO

Aggregation of α-synuclein (αS) leads to the hallmark neuropathology of Parkinson's disease (PD) and related synucleinopathies. αS has been described to exist in both cytosolic and membrane-associated forms, the relative abundance of which has remained unsettled. To study αS under the most relevant conditions by a quantitative method, we cultured and matured rodent primary cortical neurons for >17 days and determined αS cytosol:membrane distribution via centrifugation-free sequential extractions based on the weak ionic detergent digitonin. We noticed that at lower temperatures (4 °C or room temperature), αS was largely membrane-associated. At 37 °C, however, αS solubility was markedly increased. In contrast, the extraction of control proteins (GAPDH, cytosolic; calnexin, membrane) was not affected by temperature. When we compared the relative distribution of the synuclein homologs αS and ß-synuclein (ßS) under various conditions that differed in temperature and digitonin concentration (200-1200 µg/ml), we consistently found αS to be more membrane-associated than ßS. Both proteins, however, exhibited temperature-dependent membrane binding. Under the most relevant conditions (37 °C and 800 µg/ml digitonin, i.e., the lowest digitonin concentration that extracted cytosolic GAPDH to near completion), cytosolic distribution was 49.8% ± 9.0% for αS and 63.6% ± 6.6% for ßS. PD-linked αS A30P was found to be largely cytosolic, confirming previous studies that had used different methods. Our work highlights the dynamic nature of cellular synuclein behavior and has important implications for protein-biochemical and cell-biological studies of αS proteostasis, such as testing the effects of genetic and pharmacological manipulations.


Assuntos
Membrana Celular/genética , Neurônios/metabolismo , Doença de Parkinson/genética , alfa-Sinucleína/genética , beta-Sinucleína/genética , Sequência de Aminoácidos/genética , Animais , Membrana Celular/química , Humanos , Lentivirus/genética , Neurônios/química , Doença de Parkinson/imunologia , Doença de Parkinson/patologia , Cultura Primária de Células , Agregados Proteicos/genética , Agregados Proteicos/imunologia , Agregação Patológica de Proteínas/genética , Ligação Proteica/genética , Ratos , Temperatura , alfa-Sinucleína/química , alfa-Sinucleína/imunologia , alfa-Sinucleína/isolamento & purificação , beta-Sinucleína/química , beta-Sinucleína/imunologia , beta-Sinucleína/isolamento & purificação
5.
J Neurochem ; 156(5): 674-691, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-32730640

RESUMO

A contribution of α-Synuclein (α-Syn) to etiology of Parkinson´s disease (PD) and Dementia with Lewy bodies (DLB) is currently undisputed, while the impact of the closely related ß-Synuclein (ß-Syn) on these disorders remains enigmatic. ß-Syn has long been considered to be an attenuator of the neurotoxic effects of α-Syn, but in a rodent model of PD ß-Syn induced robust neurodegeneration in dopaminergic neurons of the substantia nigra. Given that dopaminergic nigral neurons are selectively vulnerable to neurodegeneration in PD, we now investigated if dopamine can promote the neurodegenerative potential of ß-Syn. We show that in cultured rodent and human neurons a dopaminergic neurotransmitter phenotype substantially enhanced ß-Syn-induced neurodegeneration, irrespective if dopamine is synthesized within neurons or up-taken from extracellular space. Nuclear magnetic resonance interaction and thioflavin-T incorporation studies demonstrated that dopamine and its oxidized metabolites 3,4-dihydroxyphenylacetaldehyde (DOPAL) and dopaminochrome (DCH) directly interact with ß-Syn, thereby enabling structural and functional modifications. Interaction of DCH with ß-Syn inhibits its aggregation, which might result in increased levels of neurotoxic oligomeric ß-Syn. Since protection of outer mitochondrial membrane integrity prevented the additive neurodegenerative effect of dopamine and ß-Syn, such oligomers might act at a mitochondrial level similar to what is suggested for α-Syn. In conclusion, our results suggest that ß-Syn can play a significant pathophysiological role in etiology of PD through its interaction with dopamine metabolites and thus should be re-considered as a disease-relevant factor, at least for those symptoms of PD that depend on degeneration of nigral dopaminergic neurons.


Assuntos
Dopamina/metabolismo , Neurônios Dopaminérgicos/metabolismo , Doenças Neurodegenerativas/metabolismo , beta-Sinucleína/metabolismo , beta-Sinucleína/toxicidade , Animais , Células Cultivadas , Neurônios Dopaminérgicos/efeitos dos fármacos , Neurônios Dopaminérgicos/patologia , Feminino , Humanos , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Pluripotentes Induzidas/patologia , Pessoa de Meia-Idade , Doenças Neurodegenerativas/induzido quimicamente , Doenças Neurodegenerativas/patologia , Gravidez , Ratos , Ratos Wistar
6.
Clin Proteomics ; 17: 13, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32390785

RESUMO

BACKGROUND: Parkinson's disease (PD) is the second most prevalent neurodegenerative disorder. Biomarkers that can help monitor the progression of PD or response to disease-modifying agents will be invaluable in making appropriate therapeutic decisions. Further, biomarkers that could be used to distinguish PD from other related disorders with PD-like symptoms will be useful for accurate diagnosis and treatment. C-Abl tyrosine kinase is activated in PD resulting in increased phosphorylation of the tyrosine residue at position 39 (Y39) of α-synuclein (α-syn) (pY39 α-syn), which contributes to the death of dopaminergic neurons. Because pY39 α-syn may be pathogenic, monitoring pY39 α-syn could allow us to diagnose presymptomatic PD and help monitor disease progression and response to treatment. We sought to investigate if increased phosphorylation of pY39 α-syn can be detected in the cerebrospinal fluid (CSF) of PD patients by targeted mass spectrometry. METHODS: Here, we report a two-step enrichment method in which phosphotyrosine peptides were first enriched with an anti-phosphotyrosine antibody followed by a second round of enrichment by titanium dioxide (TiO2) beads to detect EGVLpYVGSK sequence derived from tyrosine 39 region of α- and ß-synuclein (αß-syn). Accurate quantification was achieved by adding a synthetic heavy version of pY39 αß-syn peptide before enzymatic digestion. RESULTS: Using the developed enrichment methods and optimized parallel reaction monitoring (PRM) assays, we detected pY39 αß-syn peptide in human CSF and demonstrated that the ratio of pY39 αß-syn to Y39 αß-syn was significantly increased in the CSF of patients with PD. CONCLUSIONS: We anticipate that this optimized two-step enrichment-based PRM detection method will help monitor c-Abl activation in PD patients and can also be used to quantify other phosphotyrosine peptides of low abundance in biological samples.

7.
Int J Mol Sci ; 21(8)2020 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-32325870

RESUMO

Dementia with Lewy bodies (DLB) is the second most prevalent neurodegenerative dementia after Alzheimer's disease, and is pathologically characterized by formation of intracellular inclusions called Lewy bodies, the major constituent of which is aggregated α-synuclein (αS). Currently, neither a mechanistic etiology nor an effective disease-modifying therapy for DLB has been established. Although two missense mutations of ß-synuclein (ßS), V70M and P123H, were identified in sporadic and familial DLB, respectively, the precise mechanisms through which ßS mutations promote DLB pathogenesis remain elusive. To further clarify such mechanisms, we investigated transgenic (Tg) mice expressing P123H ßS, which develop progressive neurodegeneration in the form of axonal swelling and non-motor behaviors, such as memory dysfunction and depression, which are more prominent than motor deficits. Furthermore, cross-breeding of P123H ßS Tg mice with αS Tg mice worsened the neurodegenerative phenotype presumably through the pathological cross-seeding of P123H ßS with αS. Collectively, we predict that ßS misfolding due to gene mutations might be pathogenic. In this paper, we will discuss the possible involvement of amyloidogenic evolvability in the pathogenesis of DLB based on our previous papers regarding the P123H ßS Tg mice. Given that stimulation of αS evolvability by P123H ßS may underlie neuropathology in our mouse model, more radical disease-modifying therapy might be derived from the evolvability mechanism. Additionally, provided that altered ßS were involved in the pathogenesis of sporadic DLB, the P123H ßS Tg mice could be used for investigating the mechanism and therapy of DLB.


Assuntos
Proteínas Amiloidogênicas/genética , Proteínas Amiloidogênicas/metabolismo , Doença por Corpos de Lewy/etiologia , Doença por Corpos de Lewy/metabolismo , beta-Sinucleína/genética , beta-Sinucleína/metabolismo , Alelos , Substituição de Aminoácidos , Animais , Gerenciamento Clínico , Modelos Animais de Doenças , Suscetibilidade a Doenças , Expressão Gênica , Humanos , Doença por Corpos de Lewy/patologia , Doença por Corpos de Lewy/terapia , Camundongos , Camundongos Transgênicos , Mutação
8.
Int J Mol Sci ; 19(2)2018 Jan 24.
Artigo em Inglês | MEDLINE | ID: mdl-29364143

RESUMO

α-Synuclein (αS) is an intrinsically disordered protein that is associated with Parkinson's disease (PD) through its ability to self-assemble into oligomers and fibrils. Inhibition of this oligomerization cascade is an interesting approach to developing therapeutical strategies and ß-synuclein (ßS) has been described as a natural negative regulator of this process. However, the biological background and molecular mechanisms by which this inhibition occurs is unclear. Herein, we focused on assessing the effect of ßS on the aggregation of five αS pathological mutants linked to early-onset PD (A30P, E46K, H50Q, G51D and A53T). By coupling single molecule fluorescence spectroscopy to a cell-free protein expression system, we validated the ability of ßS to act as a chaperone of αS, effectively inhibiting its aggregation. Interestingly, we found that ßS does so in a selective manner, i.e., is a more effective inhibitor for certain αS pathological mutants-A30P and G51D-as compared to E46K, H50Q and A53T. Moreover, two-color coincidence experiments proved that this discrepancy is due to a preferential incorporation of ßS into smaller oligomers of αS. This was validated by showing that the chaperoning effect was lost when proteins were mixed after being expressed individually. This study highlights the potential of fluorescence spectroscopy to deconstruct αS aggregation cascade and its interplay with ßS.


Assuntos
Agregados Proteicos , Agregação Patológica de Proteínas , alfa-Sinucleína/metabolismo , beta-Sinucleína/metabolismo , Sistema Livre de Células , Imunofluorescência , Expressão Gênica , Genes Reporter , Humanos , Mutação , Doença de Parkinson/etiologia , Doença de Parkinson/metabolismo , Doença de Parkinson/patologia , Ligação Proteica , Multimerização Proteica , alfa-Sinucleína/química , alfa-Sinucleína/genética , beta-Sinucleína/genética
9.
Neurosci Lett ; 833: 137826, 2024 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-38768940

RESUMO

Synucleins are pivotal in neurodegenerative conditions. Beta-synuclein (ß-synuclein) is part of the synuclein protein family alongside alpha-synuclein (α-synuclein) and gamma-synuclein (γ-synuclein). These proteins, found mainly in brain tissue and cancers, are soluble and unstructured. ß-synuclein shares significant similarity with α-synuclein, especially in their N-terminus, with a 90% match. However, their aggregation tendencies differ significantly. While α-synuclein aggregation is believed to be counteracted by ß-synuclein, which occurs in conditions like Parkinson's disease, ß-synuclein may counteract α-synuclein's toxic effects on the nervous system, offering potential treatment for neurodegenerative diseases. Under normal circumstances, ß-synuclein may guard against disease by interacting with α-synuclein. Yet, in pathological environments with heightened levels or toxic substances, it might contribute to disease. Our research aims to explore potential harmful mutations in the ß-synuclein using computational tools to predict their destabilizing impact on protein structure. Consensus analysis revealed rs1207608813 (A63P), rs1340051870 (S72F), and rs1581178262 (G36C) as deleterious. These findings highlight the intricate relationship between nsSNPs and protein function, shedding light on their potential implications in disease pathways. Understanding the structural consequences of nsSNPs is crucial for elucidating their role in pathogenesis and developing targeted therapeutic interventions. Our results offer a robust computational framework for identifying neurodegenerative disorder-related mutations from SNP datasets, potentially reducing the costs associated with experimental characterization.


Assuntos
Polimorfismo de Nucleotídeo Único , beta-Sinucleína , beta-Sinucleína/genética , beta-Sinucleína/metabolismo , beta-Sinucleína/química , Humanos , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo , alfa-Sinucleína/química , Predisposição Genética para Doença , Mutação , Conformação Proteica
10.
ACS Chem Neurosci ; 15(13): 2445-2453, 2024 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-38905183

RESUMO

Parkinson's disease (PD) is the second most common age-related neurodegenerative disease in the world, and synuclein is closely related to the onset and progression of PD. Synuclein is considered a therapeutic target for PD. Recent studies have found that abnormal aggregation of α-synuclein (α-Syn) in the brains of PD patients leads to mitochondrial dysfunction and neuroinflammation. Research in the field of neuroscience has confirmed that ß-synuclein (ß-Syn) also plays a role in Parkinson's disease. However, there has been little research on the role mechanisms and interactions between ß-Syn and α-Syn in PD. Therefore, the purpose of this study is to clarify the relationship between α-Syn, ß-Syn, and PD and to explore the roles and interactions of ß-Syn and α-Syn in PD.


Assuntos
Doença de Parkinson , alfa-Sinucleína , beta-Sinucleína , Doença de Parkinson/metabolismo , alfa-Sinucleína/metabolismo , Humanos , beta-Sinucleína/metabolismo , Animais , Encéfalo/metabolismo
11.
Brain Struct Funct ; 229(5): 1279-1298, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38703218

RESUMO

ß-synuclein, a member of the synuclein family, is frequently co-expressed with α-synuclein in the neural system, where it serves to inhibit abnormal aggregation of α-synuclein in neurodegenerative diseases. Beyond its role in pathological conditions, ß-synuclein plays various functions independently of α-synuclein. In our investigation, we discovered a broader expression of ß-synuclein in the mouse retina compared to α-synuclein. This widespread pattern implies its potential significance in the retina. Through detailed examination via light- and electron-microscopic immunocytochemistry, we identified ß-synuclein expression from the inner segment (IS) and outer segment (OS) of photoreceptor cells to the ganglion cell layer (GCL). Our findings unveiled unique features, including ß-synuclein immunoreactive IS and OS of cones, higher expression in cone pedicles than in rod spherules, absence in horizontal cells, limited expression in cone bipolar dendrites and somas, higher expression in cone bipolar terminals, presence in most amacrine cells, and expression in almost majority of somas in GCL with an absence in intrinsically photosensitive retinal ganglion cell (ipRGCs) processes. Notably, all cholinergic amacrine cells express high ß- but not α-synuclein, while dopaminergic amacrine cells express α-synuclein exclusively. These distinctive expression patterns offer valuable insights for further exploration into the functions of ß-synuclein and its potential role in synuclein pathology within the retina.


Assuntos
Camundongos Endogâmicos C57BL , Retina , Células Ganglionares da Retina , alfa-Sinucleína , beta-Sinucleína , Animais , Masculino , Camundongos , alfa-Sinucleína/metabolismo , Células Amácrinas/metabolismo , beta-Sinucleína/metabolismo , Retina/metabolismo , Células Bipolares da Retina/metabolismo , Células Ganglionares da Retina/metabolismo
12.
Biology (Basel) ; 12(2)2023 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-36829434

RESUMO

Synucleins consist of three proteins exclusively expressed in vertebrates. α-Synuclein (αS) has been identified as the main proteinaceous aggregate in Lewy bodies, a pathological hallmark of many neurodegenerative diseases. Less is understood about ß-synuclein (ßS) and γ-synuclein (γS), although it is known ßS can interact with αS in vivo to inhibit aggregation. Likewise, both γS and ßS can inhibit αS's propensity to aggregate in vitro. In the central nervous system, ßS and αS, and to a lesser extent γS, are highly expressed in the neural presynaptic terminal, although they are not strictly located there, and emerging data have shown a more complex expression profile. Synapse loss and astrocyte atrophy are early aspects of degenerative diseases of the brain and correlate with disease progression. Synucleins appear to be involved in synaptic transmission, and astrocytes coordinate and organize synaptic function, with excess αS degraded by astrocytes and microglia adjacent to the synapse. ßS and γS have also been observed in the astrocyte and may provide beneficial roles. The astrocytic responsibility for degradation of αS as well as emerging evidence on possible astrocytic functions of ßS and γS, warrant closer inspection on astrocyte-synuclein interactions at the synapse.

13.
Prion ; 17(1): 29-34, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-36785484

RESUMO

Although multiple sclerosis (MS) and multiple system atrophy (MSA) are both characterized by impaired oligodendrocytes (OLs), the aetiological relevance remains obscure. Given inherent stressors affecting OLs, the objective of the present study was to discuss the possible role of amyloidogenic evolvability (aEVO) in these conditions. Hypothetically, in aEVO, protofibrils of amyloidogenic proteins (APs), including ß-synuclein and ß-amyloid, might form in response to diverse stressors in parental brain. Subsequently, the AP protofibrils might be transmitted to offspring via germ cells in a prion-like fashion. By virtue of the stress information conferred by protofibrillar APs, the OLs in offspring's brain might be more resilient to forthcoming stressors, perhaps reducing MS risk. aEVO could be comparable to a gene for the inheritance of acquired characteristics. On the contrary, during ageing, MSA risk is increased through antagonistic pleiotropy. Consistently, the expression levels of APs are reduced in MS, but are increased in MSA compared to controls. Furthermore, ß-synuclein, the non-amyloidogenic homologue of ß-synuclein, might exert a buffering effect on aEVO, and abnormal ß-synuclein could also increase MS and MSA disease activity. Collectively, a better understanding of the role of aEVO in the OL diseases might lead to novel interventions for such chronic degenerative conditions.


Assuntos
Esclerose Múltipla , Atrofia de Múltiplos Sistemas , Humanos , Atrofia de Múltiplos Sistemas/genética , Atrofia de Múltiplos Sistemas/metabolismo , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo , beta-Sinucleína/metabolismo , Esclerose Múltipla/genética , Esclerose Múltipla/metabolismo , Encéfalo/metabolismo , Proteínas Amiloidogênicas/genética , Proteínas Amiloidogênicas/metabolismo
14.
Int J Mol Sci ; 13(9): 11584-11592, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23109872

RESUMO

α-Synucleinopathies are neurodegenerative disorders that are characterized by progressive decline of motor and non-motor dysfunctions. α-Synuclein (αS) has been shown to play a causative role in neurodegeneration, but the pathogenic mechanisms are still unclear. Thus, there are no radical therapies that can halt or reverse the disease's progression. ß-Synuclein (ßS), the non-amyloidogenic homologue of αS, ameliorates the neurodegeneration phenotype of αS in transgenic (tg) mouse models, as well as in cell free and cell culture systems, which suggests that ßS might be a negative regulator of neurodegeneration caused by αS, and that "loss of function" of ßS might be involved in progression of α-synucleinopathies. Alternatively, it is possible that "toxic gain of function" of wild type ßS occurs during the pathogenesis of sporadic α-synucleinopathies, since tg mice expressing dementia with Lewy bodies-linked P123H ßS develop progressive neurodegeneration phenotypes, such as axonal pathology and dementia. In this short review, we emphasize the aspects of "toxic gain of function" of wild type ßS during the pathogenesis of sporadic α-synucleinopathies.


Assuntos
Corpos de Lewy/genética , Doença por Corpos de Lewy/genética , alfa-Sinucleína/genética , beta-Sinucleína/genética , Animais , Encéfalo/patologia , Progressão da Doença , Camundongos , Camundongos Transgênicos , Mutação de Sentido Incorreto/genética
15.
Alzheimers Res Ther ; 14(1): 179, 2022 11 30.
Artigo em Inglês | MEDLINE | ID: mdl-36451155

RESUMO

Synaptic degeneration is an early event closely associated with the course of Alzheimer's disease (AD). The identification of synaptic blood biomarkers is, therefore, of great interest and clinical relevance. The levels of most synaptic proteins are increased in the cerebrospinal fluid (CSF) of patients with AD, but their detection in blood is hitherto either unavailable or not very informative. This paradigm is related to their low concentration, their peripheral origin, or the presence of highly abundant blood proteins that hinder detection. In recent years, significant progress has been made in detecting the presynaptic protein ß-synuclein. This mini-review summarizes the results that highlight the role of ß-synuclein as a candidate blood marker for synaptic degeneration in AD.


Assuntos
Doença de Alzheimer , beta-Sinucleína , Humanos , Doença de Alzheimer/diagnóstico , Biomarcadores
16.
Antioxidants (Basel) ; 10(6)2021 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-34206096

RESUMO

The formation of multiple proteoforms by post-translational modifications (PTMs) enables a single protein to acquire distinct functional roles in its biological context. Oxidation of methionine residues (Met) is a common PTM, involved in physiological (e.g., signaling) and pathological (e.g., oxidative stress) states. This PTM typically maps at multiple protein sites, generating a heterogeneous population of proteoforms with specific biophysical and biochemical properties. The identification and quantitation of the variety of oxidized proteoforms originated under a given condition is required to assess the exact molecular nature of the species responsible for the process under investigation. In this work, the binding and oxidation of human ß-synuclein (BS) by dopamine (DA) has been explored. Native mass spectrometry (MS) has been employed to analyze the interaction of BS with DA. In a second step, top-down fragmentation of the intact protein from denaturing conditions has been performed to identify and quantify the distinct proteoforms generated by DA-induced oxidation. The analysis of isobaric proteoforms is approached by a combination of electron-transfer dissociation (ETD) at each extent of modification, quantitation of methionine-containing fragments and combinatorial analysis of the fragmentation products by multiple linear regression. This procedure represents a promising approach to systematic assessment of proteoforms variety and their relative abundance. The method can be adapted, in principle, to any protein containing any number of methionine residues, allowing for a full structural characterization of the protein oxidation states.

17.
Biomolecules ; 11(2)2021 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-33672048

RESUMO

Gaucher disease (GD), the most common lysosomal storage disorder (LSD), is caused by autosomal recessive mutations of the glucocerebrosidase gene, GBA1. In the majority of cases, GD has a non-neuropathic chronic form with adult onset (GD1), while other cases are more acute and severer neuropathic forms with early onset (GD2/3). Currently, no radical therapies are established for GD2/3. Notably, GD1, but not GD2/3, is associated with increased risk of Parkinson's disease (PD), the elucidation of which might provide a clue for novel therapeutic strategies. In this context, the objective of the present study is to discuss that the evolvability of α-synuclein (αS) might be differentially involved in GD subtypes. Hypothetically, aging-associated PD features with accumulation of αS, and the autophagy-lysosomal dysfunction might be an antagonistic pleiotropy phenomenon derived from αS evolvability in the development in GD1, without which neuropathies like GD2/3 might be manifested due to the autophagy-lysosomal dysfunction. Supposing that the increased severity of GD2/3 might be attributed to the decreased activity of αS evolvability, suppressing the expression of ß-synuclein (ßS), a potential buffer against αS evolvability, might be therapeutically efficient. Of interest, a similar view might be applicable to Niemann-Pick type C (NPC), another LSD, given that the adult type of NPC, which is comorbid with Alzheimer's disease, exhibits milder medical symptoms compared with those of infantile NPC. Thus, it is predicted that the evolvability of amyloid ß and tau, might be beneficial for the adult type of NPC. Collectively, a better understanding of amyloidogenic evolvability in the pathogenesis of LSD may inform rational therapy development.


Assuntos
Doença de Gaucher/metabolismo , Doença de Gaucher/terapia , alfa-Sinucleína/metabolismo , Amiloide/metabolismo , Autofagia , Encéfalo/metabolismo , Doença de Gaucher/genética , Glucosilceramidase/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Lisossomos/metabolismo , Modelos Biológicos , Mutação , Proteína C1 de Niemann-Pick , Doença de Parkinson/genética , Doença de Parkinson/metabolismo , Doença de Parkinson/terapia , Espécies Reativas de Oxigênio , Risco , Fatores de Risco , Resultado do Tratamento , beta-Sinucleína/metabolismo
18.
ACS Chem Neurosci ; 11(18): 2836-2848, 2020 09 16.
Artigo em Inglês | MEDLINE | ID: mdl-32833434

RESUMO

Synucleinopathies are a class of neurodegenerative diseases, including Parkinson's disease (PD), Dementia with Lewy bodies (DLB), and Multiple System Atrophy (MSA). The common pathological hallmark of synucleinopathies is the filamentous α-synuclein (α-Syn) aggregates along with membrane components in cytoplasmic inclusions in the brain. ß-Synuclein (ß-Syn), an isoform of α-Syn, inhibits α-Syn aggregation and prevents its neurotoxicity, suggesting the neuroprotective nature of ß-Syn. However, this notion changed with the discovery of disease-associated ß-Syn mutations, V70M and P123H, in patients with DLB. It is still unclear how these missense mutations alter the structural and amyloidogenic properties of ß-Syn, leading to neurodegeneration. Here, we characterized the biophysical properties and investigated the effect of mutations on ß-Syn fibrillation under different conditions. V70M and P123H show high membrane binding affinity compared to wild-type ß-Syn, suggesting their potential role in membrane interactions. ß-Syn and its mutants do not aggregate under normal physiological conditions; however, the proteins undergo self-polymerization in a slightly acidic microenvironment and/or in the presence of an inducer, forming long unbranched amyloid fibrils similar to α-Syn. Strikingly, V70M and P123H mutants exhibit accelerated fibrillation compared to native ß-Syn under these conditions. NMR study further revealed that these point mutations induce local perturbations at the site of mutation in ß-Syn. Overall, our data provide insight into the biophysical properties of disease-associated ß-Syn mutations and demonstrate that these mutants make the native protein more susceptible to aggregation in an altered microenvironment.


Assuntos
Doença de Parkinson , beta-Sinucleína , Amiloide , Humanos , Mutação/genética , Doença de Parkinson/genética , alfa-Sinucleína/genética , beta-Sinucleína/genética
19.
Mol Brain ; 11(1): 38, 2018 07 06.
Artigo em Inglês | MEDLINE | ID: mdl-29976232

RESUMO

AIM: Maturation abnormalities of the brain cells have been suggested in several neuropsychiatric disorders, including schizophrenia, bipolar disorder, autism spectrum disorders, and epilepsy. In this study, we examined the expression patterns of neuronal maturation markers in the brain of a mouse model of dementia with Lewy body-linked mutant ß-synuclein (ßS), especially in the hippocampus, to explore whether such brain abnormalities occur in neurodegenerative disorders as well. METHODS: Quantitative PCR (qPCR) and immunohistochemical analyses were performed using the hippocampus of 14-month-old P123H ßS transgenic (Tg) mice to evaluate the expression of molecular markers for maturation of dentate granule cells. RESULTS: Based on qPCR results, expression of Tdo2 and Dsp (markers of mature granule cells) was decreased and that of Drd1a (a marker of immature granule cells) was increased in the hippocampus of P123H ßS Tg mice compared to that in wild-type controls. Immunohistochemical analysis revealed decreased expression of mature granule cell markers Calb1 and Gria1, along with increased expression of the microglial marker Iba1, in the hippocampal dentate gyrus region of P123H ßS Tg mice. P123H ßS Tg mice exhibited immature-like neuronal molecular expression patterns and microgliosis in the hippocampus. Pseudo-immaturity of dentate granule cells, associated with neuroinflammation, may be a shared endophenotype in the brains of at least a subgroup of patients with neuropsychiatric disorders and neurodegenerative diseases.


Assuntos
Demência/genética , Hipocampo/metabolismo , Hipocampo/patologia , Corpos de Lewy/genética , Corpos de Lewy/patologia , Mutação/genética , beta-Sinucleína/genética , Animais , Giro Denteado/metabolismo , Giro Denteado/patologia , Modelos Animais de Doenças , Camundongos Transgênicos , Fenótipo , beta-Sinucleína/metabolismo
20.
Neuropsychopharmacol Rep ; 38(2): 95-97, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-30106256

RESUMO

AIM: We previously generated transgenic (Tg) mice that expressed P123H ß-synuclein (ßS), a dementia with Lewy body-linked mutant ßS. Notably, these mice recapitulated neurodegenerative features of Lewy body disease, reflected by motor dysfunction, greater protein aggregation, and memory impairment. Since recent studies suggested that non-motor symptoms, such as depression, might be manifested in the prodromal stage of Lewy body disease, the main objective of the present study was to investigate the early expression of behavior in P123H ßS Tg mice. METHODS: Nest building, locomotor activity, and depressive-like behavior were assessed using 6- to 10-month-old male and female P123H ßS Tg and wildtype mice. KEY RESULTS: P123H ßS Tg mice exhibited hyperlocomotor activity in a novel environment, a decrease in mobility time in the tail suspension test, and impairments in nest building. CONCLUSIONS: Importantly, these non-motor behaviors were manifested before the onset of motor dysfunction, suggesting that P123H ßS Tg mice could be a valid model for investigating the early phase of Lewy body disease.


Assuntos
Depressão/genética , Doença por Corpos de Lewy/genética , Mutação de Sentido Incorreto , beta-Sinucleína/genética , Animais , Depressão/fisiopatologia , Feminino , Doença por Corpos de Lewy/fisiopatologia , Locomoção , Masculino , Camundongos , Comportamento de Nidação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA