Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
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.
Hum Mol Genet ; 30(23): 2332-2346, 2021 11 16.
Artigo em Inglês | MEDLINE | ID: mdl-34254125

RESUMO

α-Synuclein (αS) has been well-documented to play a role in human synucleinopathies such as Parkinson's disease (PD) and dementia with Lewy bodies (DLB). First, the lesions found in PD/DLB brains-Lewy bodies and Lewy neurites-are rich in aggregated αS. Second, genetic evidence links missense mutations and increased αS expression to familial forms of PD/DLB. Third, toxicity and cellular stress can be caused by αS under certain experimental conditions. In contrast, the homologs ß-synuclein (ßS) and γ-synuclein (γS) are not typically found in Lewy bodies/neurites, have not been clearly linked to brain diseases and have been largely non-toxic in experimental settings. In αS, the so-called non-amyloid-ß component of plaques (NAC) domain, constituting amino acids 61-95, has been identified to be critical for aggregation in vitro. This domain is partially absent in ßS and only incompletely conserved in γS, which could explain why both homologs do not cause disease. However, αS in vitro aggregation and cellular toxicity have not been firmly linked experimentally, and it has been proposed that excess αS membrane binding is sufficient to induce neurotoxicity. Indeed, recent characterizations of Lewy bodies have highlighted the accumulation of lipids and membranous organelles, raising the possibility that ßS and γS could also become neurotoxic if they were more prone to membrane/lipid binding. Here, we increased ßS and γS membrane affinity by strategic point mutations and demonstrate that these proteins behave like membrane-associated monomers, are cytotoxic and form round cytoplasmic inclusions that can be prevented by inhibiting stearoyl-CoA desaturase.


Assuntos
Membrana Celular/metabolismo , Corpos de Inclusão/metabolismo , alfa-Sinucleína/metabolismo , beta-Sinucleína/metabolismo , gama-Sinucleína/metabolismo , Sequência de Aminoácidos , Sequência Conservada , Humanos , Mutagênese , Agregação Patológica de Proteínas , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Multimerização Proteica , Solubilidade , alfa-Sinucleína/química , alfa-Sinucleína/genética , beta-Sinucleína/química , beta-Sinucleína/genética , gama-Sinucleína/química , gama-Sinucleína/genética
3.
Proc Natl Acad Sci U S A ; 117(52): 33254-33262, 2020 12 29.
Artigo em Inglês | MEDLINE | ID: mdl-33318217

RESUMO

A wide range of proteins have been reported to condensate into a dense liquid phase, forming a reversible droplet state. Failure in the control of the droplet state can lead to the formation of the more stable amyloid state, which is often disease-related. These observations prompt the question of how many proteins can undergo liquid-liquid phase separation. Here, in order to address this problem, we discuss the biophysical principles underlying the droplet state of proteins by analyzing current evidence for droplet-driver and droplet-client proteins. Based on the concept that the droplet state is stabilized by the large conformational entropy associated with nonspecific side-chain interactions, we develop the FuzDrop method to predict droplet-promoting regions and proteins, which can spontaneously phase separate. We use this approach to carry out a proteome-level study to rank proteins according to their propensity to form the droplet state, spontaneously or via partner interactions. Our results lead to the conclusion that the droplet state could be, at least transiently, accessible to most proteins under conditions found in the cellular environment.


Assuntos
Proteínas/metabolismo , Proteoma/metabolismo , Aminoácidos/metabolismo , Animais , Entropia , Humanos , Extração Líquido-Líquido , Ligação Proteica , Conformação Proteica , Reprodutibilidade dos Testes , alfa-Sinucleína/química , beta-Sinucleína/química
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 Biol Chem ; 293(28): 11195-11205, 2018 07 13.
Artigo em Inglês | MEDLINE | ID: mdl-29853639

RESUMO

Membrane association of α-synuclein (α-syn), a neuronal protein associated with Parkinson's disease (PD), is involved in α-syn function and pathology. Most previous studies on α-syn-membrane interactions have not used the physiologically relevant N-terminally acetylated (N-acetyl) α-syn form nor the most naturally abundant cellular lipid, i.e. phosphatidylcholine (PC). Here, we report on how PC membrane fluidity affects the conformation and aggregation propensity of N-acetyl α-syn. It is well established that upon membrane binding, α-syn adopts an α-helical structure. Using CD spectroscopy, we show that N-acetyl α-syn transitions from α-helical to disordered at the lipid melting temperature (Tm ). We found that this fluidity sensing is a robust characteristic, unaffected by acyl chain length (Tm = 34-55 °C) and preserved in its homologs ß- and γ-syn. Interestingly, both N-acetyl α-syn membrane binding and amyloid formation trended with lipid order (1,2-dipalmitoyl-sn-glycero-3-phosphocholine (DPPC) > 1,2-dioleoyl-sn-glycero-3-phosphocholine (DOPC)/sphingomyelin/cholesterol (2:2:1) ≥ DOPC), with gel-phase vesicles shortening aggregation kinetics and promoting fibril formation compared to fluid membranes. Furthermore, we found that acetylation enhances binding to PC micelles and small unilamellar vesicles with high curvature (r ∼16-20 nm) and that DPPC binding is reduced in the presence of cholesterol. These results confirmed that the exposure of hydrocarbon chains (i.e. packing defects) is essential for binding to zwitterionic gel membranes. Collectively, our in vitro results suggest that N-acetyl α-syn localizes to highly curved, ordered membranes inside a cell. We propose that age-related changes in membrane fluidity can promote the formation of amyloid fibrils, insoluble materials associated with PD.


Assuntos
Amiloide/química , Bicamadas Lipídicas/química , Fluidez de Membrana , Proteínas de Neoplasias/química , Fosfatidilcolinas/química , alfa-Sinucleína/química , beta-Sinucleína/química , gama-Sinucleína/química , Acetilação , Sequência de Aminoácidos , Amiloide/metabolismo , Colesterol/metabolismo , Humanos , Bicamadas Lipídicas/metabolismo , Micelas , Proteínas de Neoplasias/metabolismo , Fosfatidilcolinas/metabolismo , Ligação Proteica , Conformação Proteica , Homologia de Sequência , alfa-Sinucleína/metabolismo , beta-Sinucleína/metabolismo , gama-Sinucleína/metabolismo
6.
Biochemistry ; 57(41): 5989-5995, 2018 10 16.
Artigo em Inglês | MEDLINE | ID: mdl-30239196

RESUMO

The effects of crowding, using the crowding agent Ficoll 70, and the presence of ß-synuclein on the fibrillation process of α-synuclein were studied by spectroscopic techniques, transmission electron microscopy, and thioflavin T assays. This combined approach, in which all techniques were applied to the same original sample, generated an unprecedented understanding of the effects of these modifying agents on the morphological properties of the fibrils. Separately, crowding gives rise to shorter mutually aligned fibrils, while ß-synuclein leads to branched, short fibrils. The combination of both effects leads to short, branched, mutually aligned fibrils. Moreover, it is shown that the nondestructive technique of vibrational circular dichroism is extremely sensitive to the length and the higher-order morphology of the fibrils.


Assuntos
Amiloide/química , alfa-Sinucleína/química , beta-Sinucleína/química , Amiloide/ultraestrutura , Benzotiazóis/química , Dicroísmo Circular , Humanos , Microscopia de Força Atômica , Microscopia Eletrônica de Transmissão , Estrutura Quaternária de Proteína
7.
Biochemistry ; 57(26): 3830-3848, 2018 07 03.
Artigo em Inglês | MEDLINE | ID: mdl-29851342

RESUMO

The human synuclein (syn) family is comprised of α-, ß-, and γ-syn proteins. α-syn has the highest propensity for aggregation, and its aggregated forms accumulate in Lewy bodies (LB) and Lewy neurites, which are involved in Parkinson's disease (PD). ß- and γ-syn are absent in LB, and their exact role is still enigmatic. ß-syn does not form aggregates under physiological conditions (pH 7.4), while γ-syn is associated with neural and non-neural diseases like breast cancer. Because of their similar regional distribution in the brain, natively unfolded structure, and high degree of sequence homology, studying the effect of the environment on their conformation, interactions, fibrillation, and fibril morphologies has become important. Our studies show that high temperatures, low pH values, and high concentrations increase the rate of fibrillation of α- and γ-syn, while ß-syn forms fibrils only at low pH. Fibril morphologies are strongly dependent on the immediate environment of the proteins. The high molar ratio of ß-syn inhibits the fibrillation in α- and γ-syn. However, preformed seed fibrils of ß- and γ-syn do not affect fibrillation of α-syn. Surface plasmon resonance data show that interactions between α- and ß-syn, ß- and γ-syn, and α- and γ-syn are weak to moderate in nature and can be physiologically significant in counteracting several adverse conditions in the cells that trigger their aggregation. These studies could be helpful in understanding collective human synuclein behavior in various protein environments and in the modulation of the homeostasis between ß-syn and healthy versus corrupt α- and γ-syn that can potentially affect PD pathology.


Assuntos
Proteínas de Neoplasias/química , alfa-Sinucleína/química , beta-Sinucleína/química , gama-Sinucleína/química , Humanos , Corpos de Lewy/química , Corpos de Lewy/metabolismo , Proteínas de Neoplasias/metabolismo , Doença de Parkinson/metabolismo , Agregados Proteicos , Domínios Proteicos , Ressonância de Plasmônio de Superfície , alfa-Sinucleína/metabolismo , beta-Sinucleína/metabolismo , gama-Sinucleína/metabolismo
8.
J Biol Chem ; 292(39): 16368-16379, 2017 09 29.
Artigo em Inglês | MEDLINE | ID: mdl-28710275

RESUMO

α-Synuclein (αS) is the primary protein associated with Parkinson's disease, and it undergoes aggregation from its intrinsically disordered monomeric form to a cross-ß fibrillar form. The closely related homolog ß-synuclein (ßS) is essentially fibril-resistant under cytoplasmic physiological conditions. Toxic gain-of-function by ßS has been linked to dysfunction, but the aggregation behavior of ßS under altered pH is not well-understood. In this work, we compare fibril formation of αS and ßS at pH 7.3 and mildly acidic pH 5.8, and we demonstrate that pH serves as an on/off switch for ßS fibrillation. Using αS/ßS domain-swapped chimera constructs and single residue substitutions in ßS, we localized the switch to acidic residues in the N-terminal and non-amyloid component domains of ßS. Computational models of ßS fibril structures indicate that key glutamate residues (Glu-31 and Glu-61) in these domains may be sites of pH-sensitive interactions, and variants E31A and E61A show dramatically altered pH sensitivity for fibril formation supporting the importance of these charged side chains in fibril formation of ßS. Our results demonstrate that relatively small changes in pH, which occur frequently in the cytoplasm and in secretory pathways, may induce the formation of ßS fibrils and suggest a complex role for ßS in synuclein cellular homeostasis and Parkinson's disease.


Assuntos
Ácido Glutâmico/química , Modelos Moleculares , Agregação Patológica de Proteínas/metabolismo , beta-Sinucleína/metabolismo , Substituição de Aminoácidos , Humanos , Ligação de Hidrogênio , Concentração de Íons de Hidrogênio , Microfibrilas/química , Microfibrilas/metabolismo , Microfibrilas/patologia , Mutagênese Sítio-Dirigida , Doença de Parkinson/metabolismo , Doença de Parkinson/patologia , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Mutação Puntual , Agregação Patológica de Proteínas/genética , Agregação Patológica de Proteínas/patologia , Domínios e Motivos de Interação entre Proteínas , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , alfa-Sinucleína/química , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo , beta-Sinucleína/química , beta-Sinucleína/genética
9.
Inorg Chem ; 56(17): 10387-10395, 2017 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-28820253

RESUMO

Alterations in the levels of copper in brain tissue and formation of α-synuclein (αS)-copper complexes might play a key role in the amyloid aggregation of αS and the onset of Parkinson's disease (PD). Recently, we demonstrated that formation of the high-affinity Cu(I) complex with the N-terminally acetylated form of the protein αS substantially increases and stabilizes local conformations with α-helical secondary structure and restricted motility. In this work, we performed a detailed NMR-based structural characterization of the Cu(I) complexes with the full-length acetylated form of its homologue ß-synuclein (ßS), which is colocalized with αS in vivo and can bind copper ions. Our results show that, similarly to αS, the N-terminal region of ßS constitutes the preferential binding interface for Cu(I) ions, encompassing two independent and noninteractive Cu(I) binding sites. According to these results, ßS binds the metal ion with higher affinity than αS, in a coordination environment that involves the participation of Met-1, Met-5, and Met-10 residues (site 1). Compared to αS, the shift of His from position 50 to 65 in the N-terminal region of ßS does not change the Cu(I) affinity features at that site (site 2). Interestingly, the formation of the high-affinity ßS-Cu(I) complex at site 1 in the N-terminus promotes a short α-helix conformation that is restricted to the 1-5 segment of the AcßS sequence, which differs with the substantial increase in α-helix conformations seen for N-terminally acetylated αS upon Cu(I) complexation. Our NMR data demonstrate conclusively that the differences observed in the conformational transitions triggered by Cu(I) binding to AcαS and AcßS find a correlation at the level of their backbone dynamic properties; added to the potential biological implications of these findings, this fact opens new avenues of investigations into the bioinorganic chemistry of PD.


Assuntos
Complexos de Coordenação/metabolismo , Cobre/metabolismo , Doença de Parkinson/metabolismo , beta-Sinucleína/metabolismo , Acetilação , Sítios de Ligação , Química Bioinorgânica , Complexos de Coordenação/química , Cobre/química , Humanos , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/metabolismo , Ligação Proteica , Conformação Proteica em alfa-Hélice , alfa-Sinucleína/química , alfa-Sinucleína/metabolismo , beta-Sinucleína/química
10.
Biochim Biophys Acta ; 1852(8): 1658-64, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25960149

RESUMO

Aggregation and fibril formation of human alpha-Synuclein (αS) are neuropathological hallmarks of Parkinson's disease and other synucleinopathies. The molecular mechanisms of αS aggregation and fibrillogenesis are largely unknown. Several studies suggested a sequence of events from αS dimerization via oligomerization and pre-fibrillar aggregation to αS fibril formation. In contrast to αS, little evidence suggests that γS can form protein aggregates in the brain, and for ßS its neurotoxic properties and aggregation propensities are controversially discussed. These apparent differences in aggregation behavior prompted us to investigate the first step in Synuclein aggregation, i.e. the formation of dimers or oligomers, by Bimolecular Fluorescence Complementation in cells. This assay showed some Synuclein-specific limitations, questioning its performance on a single cell level. Nevertheless, we unequivocally demonstrate that all Synucleins can interact with each other in a very similar way. Given the divergent aggregation properties of the three Synucleins this suggests that formation of dimers is not predictive for the aggregation of αS, ßS or γS in the aged or diseased brain.


Assuntos
Agregados Proteicos , Agregação Patológica de Proteínas/diagnóstico , Multimerização Proteica , Sinucleínas/metabolismo , Células Cultivadas , Células HEK293 , Células HeLa , Humanos , Microscopia de Fluorescência , Proteínas de Neoplasias/química , Proteínas de Neoplasias/metabolismo , Prognóstico , Agregação Patológica de Proteínas/metabolismo , Isoformas de Proteínas , Sinucleínas/química , alfa-Sinucleína/química , alfa-Sinucleína/metabolismo , beta-Sinucleína/química , beta-Sinucleína/metabolismo , gama-Sinucleína/química , gama-Sinucleína/metabolismo
11.
Chembiochem ; 16(16): 2319-28, 2015 Nov 02.
Artigo em Inglês | MEDLINE | ID: mdl-26338312

RESUMO

Copper binding to α-synuclein (aS) and to amyloid-ß (Ab) has been connected to Parkinson's and Alzheimer's disease (AD), respectively, because Cu ions can modulate the peptide aggregation, and these Cu ⋅ peptide complexes can catalyse the production of reactive oxygen species (ROS). In a significant proportion of AD brains, aggregation of aS and Ab has been detected, and it was proposed that Ab and aS interact with each other. Thus, we investigated the potential interactions of Ab and aS through their binding of copper(I) and copper(II). Additionally, ß-synuclein (bS) was investigated, due to its additional methionine residue, a potential Cu(I) ligand. We found that: 1) the peptides containing the Cu-binding domains Ab1-16, aS1-15 and bS1-15 have similar affinities towards Cu(II) and towards Cu(I), with Ab1-16 being slightly stronger, 2) in the case of Cu(I), the additional Met residue in bS1-15 increased the affinity slightly, 3) the exchange of Cu(I/II) between the two peptides is rapid (≤ ms), 4) a/bS1-15 and Ab1-16 form a heterodimeric complex with Cu(II), 5) Cu(I) probably promotes a transient ternary complex, 6) the different Cu(I/II) coordination of Ab1-16, aS1-15 and bS1-15 impacts the capacity to produce ROS and to oxidise catechol, and 7) when Ab1-16, aS1-15 and Cu are present, the ROS production more closely resembles that by Ab1-16. The work gives insights into the coordination chemistry of these related peptides, and the relevance of coordination differences, the ternary complex and ROS production are discussed.


Assuntos
Peptídeos beta-Amiloides/química , Cobre/química , alfa-Sinucleína/química , beta-Sinucleína/química , Peptídeos beta-Amiloides/metabolismo , Ácido Ascórbico/química , Sítios de Ligação , Catálise , Complexos de Coordenação/química , Cobre/metabolismo , Espectroscopia de Ressonância de Spin Eletrônica , Cinética , Ressonância Magnética Nuclear Biomolecular , Peptídeos/química , Peptídeos/metabolismo , Ligação Proteica , Espécies Reativas de Oxigênio/metabolismo , alfa-Sinucleína/metabolismo , beta-Sinucleína/metabolismo
12.
Inorg Chem ; 54(1): 265-72, 2015 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-25495902

RESUMO

Parkinson's disease (PD) is a neurodegenerative disorder characterized by the presence of abnormal α-synuclein (αS) deposits in the brain. Alterations in homeostasis and metal-induced oxidative stress may play a crucial role in the progression of αS amyloid assembly and pathogenesis of PD. Contrary to αS, ß-synuclein (ßS) is not involved in the PD etiology. However, it has been suggested that the ßS/αS ratio is altered in PD, indicating that a correct balance of these two proteins is implicated in the inhibition of αS aggregation. αS and ßS share similar abilities to coordinate Cu(II). In this study, we investigated and compared the interaction of Cu(I) with the N-terminal portion of ßS and αS by means of NMR, circular dichroism, and X-ray absorption spectroscopies. Our data show the importance of M10K mutation, which induces different Cu(I) chemical environments. Coordination modes 3S1O and 2S2O were identified for ßS and αS, respectively. These new insights into the bioinorganic chemistry of copper and synuclein proteins are a basis to understand the molecular mechanism by which ßS might inhibit αS aggregation.


Assuntos
Cobre/química , Peptídeos/química , alfa-Sinucleína/química , beta-Sinucleína/química , Sequência de Aminoácidos , Cátions Monovalentes , Precipitação Química , Humanos , Lisina/química , Metionina/química , Dados de Sequência Molecular , Mutação , Peptídeos/síntese química , Ligação Proteica , Técnicas de Síntese em Fase Sólida , Soluções , alfa-Sinucleína/genética , beta-Sinucleína/genética
13.
Biochemistry ; 53(46): 7170-83, 2014 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-25389903

RESUMO

α-Synuclein is an intrinsically disordered protein whose aggregation is implicated in Parkinson's disease. A second member of the synuclein family, ß-synuclein, shares significant sequence similarity with α-synuclein but is much more resistant to aggregation. ß-Synuclein is missing an 11-residue stretch in the central non-ß-amyloid component region that forms the core of α-synuclein amyloid fibrils, yet insertion of these residues into ß-synuclein to produce the ßSHC construct does not markedly increase the aggregation propensity. To investigate the structural basis of these different behaviors, quantitative nuclear magnetic resonance data, in the form of paramagnetic relaxation enhancement-derived interatomic distances, are combined with molecular dynamics simulations to generate ensembles of structures representative of the solution states of α-synuclein, ß-synuclein, and ßSHC. Comparison of these ensembles reveals that the differing aggregation propensities of α-synuclein and ß-synuclein are associated with differences in the degree of residual structure in the C-terminus coupled to the shorter separation between the N- and C-termini in ß-synuclein and ßSHC, making protective intramolecular contacts more likely.


Assuntos
Agregados Proteicos , alfa-Sinucleína/química , beta-Sinucleína/química , Sequência de Aminoácidos , Humanos , Simulação de Dinâmica Molecular , Dados de Sequência Molecular , Doença de Parkinson/metabolismo , Agregação Patológica de Proteínas/metabolismo , Estrutura Secundária de Proteína , Alinhamento de Sequência , alfa-Sinucleína/ultraestrutura , beta-Sinucleína/ultraestrutura
14.
J Biol Chem ; 288(3): 1829-40, 2013 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-23184946

RESUMO

Synucleins are a family of presynaptic membrane binding proteins. α-Synuclein, the principal member of this family, is mutated in familial Parkinson disease. To gain insight into the molecular functions of synucleins, we performed an unbiased proteomic screen and identified synaptic protein changes in αßγ-synuclein knock-out brains. We observed increases in the levels of select membrane curvature sensing/generating proteins. One of the most prominent changes was for the N-BAR protein endophilin A1. Here we demonstrate that the levels of synucleins and endophilin A1 are reciprocally regulated and that they are functionally related. We show that all synucleins can robustly generate membrane curvature similar to endophilins. However, only monomeric but not tetrameric α-synuclein can bend membranes. Further, A30P α-synuclein, a Parkinson disease mutant that disrupts protein folding, is also deficient in this activity. This suggests that synucleins generate membrane curvature through the asymmetric insertion of their N-terminal amphipathic helix. Based on our findings, we propose to include synucleins in the class of amphipathic helix-containing proteins that sense and generate membrane curvature. These results advance our understanding of the physiological function of synucleins.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/química , Membrana Celular/química , Lipossomos/química , Doença de Parkinson/metabolismo , alfa-Sinucleína/química , beta-Sinucleína/química , gama-Sinucleína/química , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Química Encefálica , Membrana Celular/metabolismo , Forma Celular , Regulação da Expressão Gênica , Humanos , Lipossomos/metabolismo , Camundongos , Camundongos Knockout , Doença de Parkinson/genética , Dobramento de Proteína , Multimerização Proteica , Estrutura Secundária de Proteína , Proteômica , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Vesículas Sinápticas/química , alfa-Sinucleína/deficiência , alfa-Sinucleína/genética , beta-Sinucleína/deficiência , beta-Sinucleína/genética , gama-Sinucleína/deficiência , gama-Sinucleína/genética
15.
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
16.
Biochem Biophys Res Commun ; 441(2): 308-17, 2013 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-24140056

RESUMO

Over the last decade, many genetic studies have suggested that the synucleins, which are small, natively unfolded proteins, are closely related to Parkinson's disease and cancer. Less is known about the molecular basis of this role. A comprehensive analysis of the evolutionary path of the synuclein protein family may reveal the relationship between evolutionarily conserved residues and protein function or structure. The phylogeny of 252 unique synuclein sequences from 73 organisms suggests that gamma-synuclein is the common ancestor of alpha- and beta-synuclein. Although all three sub-families remain highly conserved, especially at the N-terminal, nearly 15% of the residues in each sub family clearly diverged during evolution, providing crucial guidance for investigations of the different properties of the members of the superfamily. His50 is found to be an alpha-specific conserved residue (91%) and, based on mutagenesis, evolutionarily developed a secondary copper binding site in the alpha synuclein family. Surprisingly, this site is located between two well-known polymorphisms of alpha-synuclein, E46K and A53T, which are linked to early-onset Parkinson's disease, suggesting that the mutation-induced impairment of copper binding could be a mechanism responsible for alpha-synuclein aggregation.


Assuntos
Evolução Molecular , Filogenia , alfa-Sinucleína/classificação , beta-Sinucleína/classificação , gama-Sinucleína/classificação , Sequência de Aminoácidos , Sítios de Ligação , Sequência Conservada , Cobre/química , Humanos , Dados de Sequência Molecular , Alinhamento de Sequência , alfa-Sinucleína/química , alfa-Sinucleína/genética , beta-Sinucleína/química , beta-Sinucleína/genética , gama-Sinucleína/química , gama-Sinucleína/genética
17.
Biochemistry ; 51(44): 8771-8, 2012 Nov 06.
Artigo em Inglês | MEDLINE | ID: mdl-23003198

RESUMO

The aggregation process of α-synuclein, a protein closely associated with Parkinson's disease, is highly sensitive to sequence variations. It is therefore of great importance to understand the factors that define the aggregation propensity of specific mutational variants as well as their toxic behavior in the cellular environment. In this context, we investigated the extent to which the aggregation behavior of α-synuclein can be altered to resemble that of ß-synuclein, an aggregation-resistant homologue of α-synuclein not associated with disease, by swapping residues between the two proteins. Because of the vast number of possible swaps, we have applied a rational design procedure to single out a mutational variant, called α2ß, in which two short stretches of the sequence in the NAC region have been replaced in α-synuclein from ß-synuclein. We find not only that the aggregation rate of α2ß is close to that of ß-synuclein, being much lower than that of α-synuclein, but also that α2ß effectively changes the cellular toxicity of α-synuclein to a value similar to that of ß-synuclein upon exposure of SH-SY5Y cells to preformed oligomers. Remarkably, control experiments on the corresponding mutational variant of ß-synuclein, called ß2α, confirmed that the mutations that we have identified also shift the aggregation behavior of this protein toward that of α-synuclein. These results demonstrate that it is becoming possible to control in quantitative detail the sequence code that defines the aggregation behavior and toxicity of α-synuclein.


Assuntos
Estrutura Quaternária de Proteína , alfa-Sinucleína/química , beta-Sinucleína/química , Sequência de Aminoácidos , Substituição de Aminoácidos , Sobrevivência Celular , Humanos , Dados de Sequência Molecular , Alinhamento de Sequência , Células Tumorais Cultivadas , alfa-Sinucleína/genética , alfa-Sinucleína/toxicidade , beta-Sinucleína/toxicidade
18.
J Inorg Biochem ; 229: 111715, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35074552

RESUMO

Amyloid aggregation of α-synuclein (AS) is one of the hallmarks of Parkinson's disease (PD). Copper ions specifically bind at the N-terminus of AS, accelerating protein aggregation. Its protein homolog ß-synuclein (BS) is also a copper binding protein, but it inhibits AS aggregation. Here, a comparative spectroscopic study of the Cu2+ binding properties of AS and BS has been performed, using electronic absorption, circular dichroism (CD) and electronic paramagnetic resonance (EPR). Our comparative spectroscopic study reveals striking similarities between the Cu2+ binding features of the two proteins. The Cu2+ binding site at the N-terminal group of BS protein, modeled by the BS (1-15) fragment is identical to that of AS; however, its rate of reduction is three times faster as compared to the AS site, consistent with BS having an additional Met residue in its Met1-Xn-Met5-Xn-Met10 motif. The latter is also evident in the cyclic voltammetry studies of the Cu-BS complex. On the other hand, the Cu2+ binding features of the His site in both proteins, as modeled by AS(45-55) and BS(60-70), are identical, indicating that the shift in the His position does not affect its coordination features. Finally, replacement of Glu46 by Ala does not alter Cu2+ binding to the His site, suggesting that the familial PD E46K mutation would not impact copper-induced aggregation. While further studies of the redox activity of copper bound to His50 in AS are required to understand the role of this site in metal-mediated aggregation, our study contributes to a better understanding of the bioinorganic chemistry of PD.


Assuntos
Cobre/metabolismo , alfa-Sinucleína/metabolismo , beta-Sinucleína/metabolismo , Sequência de Aminoácidos , Sítios de Ligação , Histidina/química , Histidina/metabolismo , Metionina/química , Metionina/metabolismo , Ligação Proteica , alfa-Sinucleína/química , beta-Sinucleína/química
19.
Biochemistry ; 50(1): 37-47, 2011 Jan 11.
Artigo em Inglês | MEDLINE | ID: mdl-21117662

RESUMO

Thermodynamic studies in conjunction with EPR confirm that α-synuclein, ß-synuclein, and γ-synuclein bind copper(II) in a high affinity 1:1 stoichiometry. γ-Synuclein demonstrates the highest affinity, in the picomolar range, while α-synuclein and ß-synuclein both bind copper(II) with nanomolar affinity. The copper center on all three proteins demonstrates reversible or partly reversible redox cycling. Various mutations show that the primary coordinating ligand for copper(II) is located within the N-terminal regions between residues 2-9. There is also a contribution from the C-terminus in conjunction with the histidine at position 50 in α-synuclein and position 65 in ß-synuclein, although these regions appear to have little effect on overall coordination stability. These histidines and the C-terminus, however, appear to be critical to the redox engine of the proteins.


Assuntos
Proteínas de Transporte/metabolismo , alfa-Sinucleína/metabolismo , beta-Sinucleína/metabolismo , gama-Sinucleína/metabolismo , Sítios de Ligação , Cobre/metabolismo , Espectroscopia de Ressonância de Spin Eletrônica , Humanos , Mutagênese Sítio-Dirigida , Mutação , Oxirredução , Ligação Proteica , Termodinâmica , alfa-Sinucleína/química , alfa-Sinucleína/genética , beta-Sinucleína/química , beta-Sinucleína/genética , gama-Sinucleína/química , gama-Sinucleína/genética
20.
Biochemistry ; 50(31): 6667-77, 2011 Aug 09.
Artigo em Inglês | MEDLINE | ID: mdl-21699177

RESUMO

C-Terminal Src kinase-homologous kinase (CHK) exerts its tumor suppressor function by phosphorylating the C-terminal regulatory tyrosine of the Src-family kinases (SFKs). The phosphorylation suppresses their activity and oncogenic action. In addition to phosphorylating SFKs, CHK also performs non-SFK-related functions by phosphorylating other cellular protein substrates. To define these non-SFK-related functions of CHK, we used the "kinase substrate tracking and elucidation" method to search for its potential physiological substrates in rat brain cytosol. Our search revealed ß-synuclein as a potential CHK substrate, and Y127 in ß-synuclein as the preferential phosphorylation site. Using peptides derived from ß-synuclein and positional scanning combinatorial peptide library screening, we defined the optimal substrate phosphorylation sequence recognized by the CHK active site to be E-x-[Φ/E/D]-Y-Φ-x-Φ, where Φ and x represent hydrophobic residues and any residue, respectively. Besides ß-synuclein, cellular proteins containing motifs resembling this sequence are potential CHK substrates. Intriguingly, the CHK-optimal substrate phosphorylation sequence bears little resemblance to the C-terminal tail sequence of SFKs, indicating that interactions between the CHK active site and the local determinants near the C-terminal regulatory tyrosine of SFKs play only a minor role in governing specific phosphorylation of SFKs by CHK. Our results imply that recognition of SFKs by CHK is mainly governed by interactions between motifs located distally from the active site of CHK and determinants spatially separate from the C-terminal regulatory tyrosine in SFKs. Thus, besides assisting in the identification of potential CHK physiological substrates, our findings shed new light on how CHK recognizes SFKs and other protein substrates.


Assuntos
Proteínas Tirosina Quinases/química , Homologia Estrutural de Proteína , beta-Sinucleína/química , Domínios de Homologia de src , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Proteína Tirosina Quinase CSK , Domínio Catalítico , Citosol/enzimologia , Citosol/metabolismo , Células HEK293 , Humanos , Dados de Sequência Molecular , Biblioteca de Peptídeos , Fosforilação , Proteínas Tirosina Quinases/metabolismo , Ratos , Especificidade por Substrato , beta-Sinucleína/metabolismo , Quinases da Família src
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA