Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 6.279
Filtrar
1.
Nanoscale ; 16(16): 8074-8089, 2024 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-38563405

RESUMO

Amyloid aggregation is implicated in the pathogenesis of various neurodegenerative disorders, such as Alzheimer's disease (AD) and Parkinson's disease (PD). It is critical to develop high-performance drugs to combat amyloid-related diseases. Most identified nanomaterials exhibit limited biocompatibility and therapeutic efficacy. In this work, we used a solvent-free carbonization process to prepare new photo-responsive carbon nanodots (CNDs). The surface of the CNDs is densely packed with chemical groups. CNDs with large, conjugated domains can interact with proteins through π-π stacking and hydrophobic interactions. Furthermore, CNDs possess the ability to generate singlet oxygen species (1O2) and can be used to oxidize amyloid. The hydrophobic interaction and photo-oxidation can both influence amyloid aggregation and disaggregation. Thioflavin T (ThT) fluorescence analysis and circular dichroism (CD) spectroscopy indicate that CNDs can block the transition of amyloid from an α-helix structure to a ß-sheet structure. CNDs demonstrate efficacy in alleviating cytotoxicity induced by Aß42 and exhibit promising blood-brain barrier (BBB) permeability. CNDs have small size, low biotoxicity, good fluorescence and photocatalytic properties, and provide new ideas for the diagnosis and treatment of amyloid-related diseases.


Assuntos
Peptídeos beta-Amiloides , Carbono , Carbono/química , Peptídeos beta-Amiloides/metabolismo , Peptídeos beta-Amiloides/química , Humanos , Catálise , Oxigênio Singlete/metabolismo , Oxigênio Singlete/química , Barreira Hematoencefálica/metabolismo , Animais , Agregados Proteicos/efeitos dos fármacos , Pontos Quânticos/química , Amiloide/química , Amiloide/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/metabolismo , Interações Hidrofóbicas e Hidrofílicas
2.
ACS Appl Mater Interfaces ; 16(15): 18268-18284, 2024 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-38564419

RESUMO

The essential amino acid histidine plays a central role in the manifestation of several metabolic processes, including protein synthesis, enzyme-catalysis, and key biomolecular interactions. However, excess accumulation of histidine causes histidinemia, which shows brain-related medical complications, and the molecular mechanism of such histidine-linked complications is largely unknown. Here, we show that histidine undergoes a self-assembly process, leading to the formation of amyloid-like cytotoxic and catalytically active nanofibers. The kinetics of histidine self-assembly was favored in the presence of Mg(II) and Co(II) ions. Molecular dynamics data showed that preferential noncovalent interactions dominated by H-bonds between histidine molecules facilitate the formation of histidine nanofibers. The histidine nanofibers induced amyloid cross-seeding reactions in several proteins and peptides including pathogenic Aß1-42 and brain extract components. Further, the histidine nanofibers exhibited oxidase activity and enhanced the oxidation of neurotransmitters. Cell-based studies confirmed the cellular internalization of histidine nanofibers in SH-SY5Y cells and subsequent cytotoxic effects through necrosis and apoptosis-mediated cell death. Since several complications including behavioral abnormality, developmental delay, and neurological disabilities are directly linked to abnormal accumulation of histidine, our findings provide a foundational understanding of the mechanism of histidine-related complications. Further, the ability of histidine nanofibers to catalyze amyloid seeding and oxidation reactions is equally important for both biological and materials science research.


Assuntos
Nanofibras , Nanoestruturas , Neuroblastoma , Humanos , Histidina , Peptídeos/química , Nanofibras/química , Amiloide/química , Peptídeos beta-Amiloides/química
3.
Phys Chem Chem Phys ; 26(15): 11880-11892, 2024 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-38568008

RESUMO

Recent experiments have revealed that adenosine triphosphate (ATP) suppresses the fibrillation of amyloid peptides - a process closely linked to neurodegenerative diseases such as Alzheimer's and Parkinson's. Apart from the adsorption of ATP onto amyloid peptides, the molecular understanding is still limited, leaving the underlying mechanism for the fibrillation suppression by ATP largely unclear, especially in regards to the molecular energetics. Here we provide an explanation at the molecular scale by quantifying the free energies using all-atom molecular dynamics simulations. We found that the changes of the free energies due to the addition of ATP lead to a significant equilibrium shift towards monomeric peptides in agreement with experiments. Despite ATP being a highly charged species, the decomposition of the free energies reveals that the van der Waals interactions with the peptide are decisive in determining the relative stabilization of the monomeric state. While the phosphate moiety exhibits strong electrostatic interactions, the compensation by the water solvent results in a minor, overall Coulomb contribution. Our quantitative analysis of the free energies identifies which intermolecular interactions are responsible for the suppression of the amyloid fibril formation by ATP and offers a promising method to analyze the roles of similarly complex cosolvents in aggregation processes.


Assuntos
Amiloide , Peptídeos , Amiloide/química , Peptídeos/química , Água/química , Entropia , Solventes/química , Simulação de Dinâmica Molecular , Proteínas Amiloidogênicas , Peptídeos beta-Amiloides/química , Fragmentos de Peptídeos/química
4.
J Phys Chem B ; 128(14): 3320-3328, 2024 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-38447080

RESUMO

Protein self-assembly plays an important role in biological systems, accounting for the formation of mesoscopic structures that can be highly symmetric as in the capsid of viruses or disordered as in molecular condensates or exhibit a one-dimensional fibrillar morphology as in amyloid fibrils. Deposits of the latter in tissues of individuals with degenerative diseases like Alzheimer's and Parkinson's has motivated extensive efforts to understand the sequence of molecular events accounting for their formation. These studies aim to identify on-pathway intermediates that may be the targets for therapeutic intervention. This detailed knowledge of fibril formation remains obscure, in part due to challenges with experimental analyses of these processes. However, important progress is being achieved for short amyloid peptides due to advances in our ability to perform completely unbiased all-atom simulations of the self-assembly process. This perspective discusses recent developments, their implications, and the hurdles that still need to be overcome to further advance the field.


Assuntos
Peptídeos beta-Amiloides , Amiloide , Humanos , Amiloide/química , Peptídeos beta-Amiloides/química
5.
J Alzheimers Dis ; 98(3): 755-772, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38489183

RESUMO

Alzheimer's disease (AD) is an extremely complex and heterogeneous pathology influenced by many factors contributing to its onset and progression, including aging, amyloid-beta (Aß) plaques, tau fibril accumulation, inflammation, etc. Despite promising advances in drug development, there is no cure for AD. Although there have been substantial advancements in understanding the pathogenesis of AD, there have been over 200 unsuccessful clinical trials in the past decade. In recent years, immunotherapies have been at the forefront of these efforts. Immunotherapy alludes to the immunological field that strives to identify disease treatments via the enhancement, suppression, or induction of immune responses. Interestingly, immunotherapy in AD is a relatively new approach for non-infectious disease. At present, antibody therapy (passive immunotherapy) that targets anti-Aß aimed to prevent the fibrillization of Aß peptides and disrupt pre-existing fibrils is a predominant AD immunotherapy due to the continuous failure of active immunotherapy for AD. The most rational and safe strategies will be those targeting the toxic molecule without triggering an abnormal immune response, offering therapeutic advantages, thus making clinical trial design more efficient. This review offers a concise overview of immunotherapeutic strategies, including active and passive immunotherapy for AD. Our review encompasses approved methods and those presently under investigation in clinical trials, while elucidating the recent challenges, complications, successes, and potential treatments. Thus, immunotherapies targeting Aß throughout the disease progression using a mutant oligomer-Aß stimulated dendritic cell vaccine may offer a promising therapy in AD.


Assuntos
Doença de Alzheimer , Humanos , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/química , Imunoterapia , Imunização Passiva , Inflamação
6.
J Chem Phys ; 160(12)2024 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-38516974

RESUMO

Alzheimer's disease is a detrimental neurological disorder caused by the formation of amyloid fibrils due to the aggregation of amyloid-ß peptide. The primary therapeutic approaches for treating Alzheimer's disease are targeted to prevent this amyloid fibril formation using potential inhibitor molecules. The discovery of such inhibitor molecules poses a formidable challenge to the design of anti-amyloid drugs. This study investigates the effect of caffeine on dimer formation of the full-length amyloid-ß using a combined approach of all-atom, explicit water molecular dynamics simulations and the three-dimensional reference interaction site model theory. The change in the hydration free energy of amyloid-ß dimer, with and without the inhibitor molecules, is calculated with respect to the monomeric amyloid-ß, where the hydration free energy is decomposed into energetic and entropic components, respectively. Dimerization is accompanied by a positive change in the partial molar volume. Dimer formation is spontaneous, which implies a decrease in the hydration free energy. However, a reverse trend is observed for the dimer with inhibitor molecules. It is observed that the negatively charged residues primarily contribute for the formation of the amyloid-ß dimer. A residue-wise decomposition reveals that hydration/dehydration of the side-chain atoms of the charged amino acid residues primarily contribute to dimerization.


Assuntos
Doença de Alzheimer , Humanos , Doença de Alzheimer/metabolismo , Cafeína/farmacologia , Peptídeos beta-Amiloides/química , Amiloide , Simulação de Dinâmica Molecular , Fragmentos de Peptídeos/química
7.
J Chem Inf Model ; 64(6): 2035-2044, 2024 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-38427576

RESUMO

Effective scaffolding of immunogens is crucial for generating conformationally selective antibodies through active immunization, particularly in the treatment of protein misfolding diseases such as Alzheimer's and Parkinson's disease. Previous computational work has revealed that a disorder-prone region of the tau protein, when in a stacked form, is predicted to structurally resemble a small, soluble protofibril, having conformational properties similar to those of experimental in vitro tau oligomers. Such an oligomeric structural mimic has the potential to serve as a vaccine immunogen design for Alzheimer's disease. In this study, we developed a cyclization scaffolding method in Rosetta, in which multiple cyclic peptides are stacked into a protofibril. Cyclization results in significant stabilization of protofibril-like structures by constraining the conformational space. Applying this method to the disorder-prone region of the tau fibril, we evaluated the metastability of the cyclized tau immunogen using molecular dynamics simulations, and we identified sequences of two cyclic constructs having high metastability in the protofibril. We then assessed their thermodynamic stability by computing the free energy required to separate a distal chain from the rest of the stacked structure. Our computational results, based on molecular dynamics simulations and free energy calculations, demonstrate that two cyclized constructs, cyclo-(VKSEKLDFKDRVQSKIFyN) and cyclo-(VKSEKLDFKDRVQSKIYvG) (lowercase letters indicate d-form amino acids), possess significantly increased thermodynamic stability in the protofibril over an uncyclized linear construct VKSEKLDFKDRVQSKI. The cyclization scaffolding approach proposed here holds promise as a means to effectively design immunogens for protein misfolding diseases, particularly those involving liposome-conjugated peptide constructs.


Assuntos
Doença de Alzheimer , Deficiências na Proteostase , Vacinas , Humanos , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/química , Proteínas tau/metabolismo , Ciclização , Simulação de Dinâmica Molecular
8.
Methods Mol Biol ; 2754: 193-203, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38512668

RESUMO

Alzheimer's disease (AD) is characterized by the abnormal accumulation of disordered protein, that is, extracellular senile plaques of amyloid-ß (Aß) and intracellular neurofibrillary tangles of Tau. Tau protein has gained the attention in recent years owing to the ability to propagate in a "prion-like" nature. The disordered protein Tau possesses a high positive charge, which allows its binding to anionic proteins and factors. The native disorder of proteins attends the ß-sheet structure from its random-coiled conformation upon charge compensation by various polyanionic agents such as heparin, RNA, etc. Anionic lipids such as arachidonic acid (AA) and oleic acid (OA) are also one of the factors which can induce aggregation of Tau in physiological conditions. The free units of Tau protein can bind to lipid membranes through its repeat domain (RD), the anionic side chains of the membrane lipids induce aggregation of Tau by reducing the activation barrier. In this study, we investigated the role of α-linolenic acid (ALA) as an inducing agent for Tau aggregation in vitro conditions. Omega-3 fatty acids bear a capacity to reduce the pathology of Tau by downregulating the Tau phosphorylation pathway. We have studied by using various biochemical or biophysical methods the potency of ALA as an aggregating agent for Tau. We have implemented different techniques such as SDS-PAGE, transmission electron microscopy, CD spectroscopy to evaluated higher-order aggregates of Tau upon induction by ALA.


Assuntos
Doença de Alzheimer , Proteínas tau , Humanos , Proteínas tau/metabolismo , Ácido alfa-Linolênico/farmacologia , Ácido alfa-Linolênico/metabolismo , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/química , Emaranhados Neurofibrilares/metabolismo
9.
Food Funct ; 15(8): 4233-4245, 2024 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-38517352

RESUMO

The aggregation of ß-amyloid (Aß) peptides to form amyloid plaques is one of the primary hallmarks for Alzheimer's disease (AD). Dietary flavonoid supplements containing hesperetin have an ability to decline the risk of developing AD, but the molecular mechanism is still unclear. In this work, hesperetin, a flavanone abundant in citrus fruits, has been proven to prevent the formation of Aß aggregates and depolymerized preformed fibrils in a concentration-dependent fashion. Hesperetin inhibited the conformational conversion from the natural structure to a ß-sheet-rich conformation. It was found that hesperetin significantly reduced the cytotoxicity and relieved oxidative stress eventuated by Aß aggregates in a concentration-dependent manner. Additionally, the beneficial effects of hesperetin were confirmed in Caenorhabditis elegans, including the inhibition of the formation and deposition of Aß aggregates and extension of their lifespan. Finally, the results of molecular dynamics simulations showed that hesperetin directly interacted with an Aß42 pentamer mainly through strong non-polar and electrostatic interactions, which destroyed the structural stability of the preformed pentamer. To summarize, hesperetin exhibits great potential as a prospective dietary supplement for preventing and improving AD.


Assuntos
Peptídeos beta-Amiloides , Caenorhabditis elegans , Hesperidina , Hesperidina/farmacologia , Hesperidina/química , Peptídeos beta-Amiloides/metabolismo , Peptídeos beta-Amiloides/toxicidade , Peptídeos beta-Amiloides/química , Animais , Caenorhabditis elegans/efeitos dos fármacos , Humanos , Doença de Alzheimer/metabolismo , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/prevenção & controle , Amiloide/metabolismo , Simulação de Dinâmica Molecular , Estresse Oxidativo/efeitos dos fármacos , Agregados Proteicos/efeitos dos fármacos
10.
ACS Nano ; 18(12): 8798-8810, 2024 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-38478911

RESUMO

Synthetic peptides that self-assemble into cross-ß fibrils are versatile building blocks for engineered biomaterials due to their modularity and biocompatibility, but their structural and morphological similarities to amyloid species have been a long-standing concern for their translation. Further, their polymorphs are difficult to characterize by using spectroscopic and imaging techniques that rely on ensemble averaging to achieve high resolution. Here, we utilize Nile red (NR), an amyloidophilic fluorogenic probe, and single-molecule orientation-localization microscopy (SMOLM) to characterize fibrils formed by the designed amphipathic enantiomers KFE8L and KFE8D and the pathological amyloid-beta peptide Aß42. Importantly, NR SMOLM reveals the helical (bilayer) ribbon structure of both KFE8 and Aß42 and quantifies the precise tilt of the fibrils' inner and outer backbones in relevant buffer conditions without the need for covalent labeling or sequence mutations. SMOLM also distinguishes polymorphic branched and curved morphologies of KFE8, whose backbones exhibit much more heterogeneity than those of typical straight fibrils. Thus, SMOLM is a powerful tool to interrogate the structural differences and polymorphism between engineered and pathological cross-ß-rich fibrils.


Assuntos
Peptídeos beta-Amiloides , Microscopia , Conformação Proteica em Folha beta , Estrutura Secundária de Proteína , Peptídeos beta-Amiloides/química , Amiloide/química
11.
ACS Chem Neurosci ; 15(6): 1125-1134, 2024 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-38416693

RESUMO

Oligomeric assemblies of the amyloid ß peptide (Aß) have been investigated for over two decades as possible neurotoxic agents in Alzheimer's disease. However, due to their heterogeneous and transient nature, it is not yet fully established which of the structural features of these oligomers may generate cellular damage. Here, we study distinct oligomer species formed by Aß40 (the 40-residue form of Aß) in the presence of four different metal ions (Al3+, Cu2+, Fe2+, and Zn2+) and show that they differ in their structure and toxicity in human neuroblastoma cells. We then describe a correlation between the size of the oligomers and their neurotoxic activity, which provides a type of structure-toxicity relationship for these Aß40 oligomer species. These results provide insight into the possible role of metal ions in Alzheimer's disease by the stabilization of Aß oligomers.


Assuntos
Doença de Alzheimer , Peptídeos beta-Amiloides , Humanos , Peptídeos beta-Amiloides/química , Metais , Íons , Fragmentos de Peptídeos/química
12.
J Inorg Biochem ; 253: 112480, 2024 04.
Artigo em Inglês | MEDLINE | ID: mdl-38309203

RESUMO

Amyloid beta (Aß) peptides and copper (Cu) ions are each involved in critical biological processes including antimicrobial activity, regulation of synaptic function, angiogenesis, and others. Aß binds to Cu and may play a role in Cu trafficking. Aß peptides exist in isoforms that vary at their C-and N-termini; variation at the N-terminal sequence affects Cu binding affinity, structure, and redox activity by providing different sets of coordinating groups to the metal ion. Several N-terminal isoforms have been detected in human brain tissues including Aß1-40/42, Aß3-42, pEAß3-42, Aß4-42, Aß11-40 and pEAß11-40 (where pE denotes an N-terminal pyroglutamic acid). Several previous works have individually investigated the affinity and structure of Cu(I) bound to some of these isoforms' metal binding domains. However, the disparately reported values are apparent constants collected under different sets of conditions, and thus an integrated comparison cannot be made. The work presented here provides the Cu(I) coordination structure and binding affinities of these six biologically relevant Aß isoforms determined in parallel using model peptides of the Aß metal binding domains (Aß1-16, Aß3-16, pEAß3-16, Aß4-16, Aß11-16 and pEAß11-16). The binding affinities of Cu(I)-Aß complexes were measured using solution competition with ferrozine (Fz) and bicinchoninic acid (BCA), two colorimetric Cu(I) indicators in common use. The Cu(I) coordination structures were characterized by X-ray absorption spectroscopy. The data presented here facilitate comparison of the isoforms' Cu-binding interactions and contribute to our understanding of the role of Aß peptides as copper chelators in healthy and diseased brains.


Assuntos
Peptídeos beta-Amiloides , Cobre , Humanos , Peptídeos beta-Amiloides/química , Cobre/química , Isoformas de Proteínas , Íons , Quelantes
13.
J Phys Chem B ; 128(7): 1647-1655, 2024 Feb 22.
Artigo em Inglês | MEDLINE | ID: mdl-38334278

RESUMO

Amyloid ß (Aß) is a hallmark protein of Alzheimer's disease. One physiologically important Aß variant is formed by initial N-terminal truncation at a glutamic acid position (either E3 or E11), which is subsequently cyclized to a pyroglutamate (either pE3 or pE11). Both forms have been found in high concentrations in the core of amyloid plaques and are likely of high importance in the pathology of Alzheimer's disease. However, the molecular structure of the fibrils of these variants is not entirely clear. Solid-state NMR spectroscopy studies have reported a molecular contact between Gly25 and Ile31, which would disagree with the conventional hairpin model of wildtype (WT-)Aß1-40 fibrils, most often described in the literature. We investigated the conformation of the monomeric unit of pE3-Aß3-40 and pE11-Aß11-40 (and for comparison also wildtype (WT)-Aß1-40) fibrils to find out whether the hairpin or a newly suggested extended structure dominates the structure of the Aß monomers in these fibrils. To this end, solid-state NMR spectroscopy was applied probing the inter-residual contacts between Phe19/Leu34, Ala21/Leu34, and especially Gly25/Ile31 using suitable isotopic labeling schemes. In the second part, the flexible turn of the Aß40 peptides was replaced by a (3-(3-aminomethyl)phenylazo)phenylacetic acid (AMPP)-based photoswitch, which can predefine the peptide conformation to either an extended (trans) or hairpin (cis) conformation. This enables simultaneous spectroscopic assessment of the conformation of the AMPP-photoswitch, allowing in situ structural investigations during fibrillation in contrast to structural techniques such as NMR spectroscopy or cryo-EM, which can only be applied to stable conformers. Both methods confirm an extended structure for the peptidic monomers in fibrils of all investigated Aß variants. Especially the Gly25/Ile31 contact is a decisive indicator for the extended structure along with the characteristic absorption spectra of trans-AMPP-Aß.


Assuntos
Doença de Alzheimer , Peptídeos beta-Amiloides , Humanos , Peptídeos beta-Amiloides/química , Doença de Alzheimer/metabolismo , Conformação Molecular , Estrutura Molecular , Espectroscopia de Ressonância Magnética , Amiloide , Fragmentos de Peptídeos/química
14.
J Mol Graph Model ; 129: 108732, 2024 06.
Artigo em Inglês | MEDLINE | ID: mdl-38412813

RESUMO

Recent evidence from in vitro and in vivo studies has shown that anthocyanins and anthocyanidins can reduce and inhibit the amyloid beta (Aß) species, one of the hallmarks of Alzheimer's disease (AD). However, their inhibition mechanisms on Aß species at molecular details remain elusive. Therefore, in the present study, molecular modelling methods were employed to investigate their inhibitory mechanisms on Aß(1-42) peptide. The results highlighted that anthocyanidins effectively inhibited the conformational transitions of helices into beta-sheet (ß-sheet) conformation within Aß(1-42) peptide by two different mechanisms: 1) the obstruction of two terminals from coming into contact due to the binding of anthocyanidins with residues of N- and second hydrophobic core (SHC)-C-terminals, and 2) the prevention of the folding process due to the binding of anthocyanidin with the central polar (Asp23 and Lys28) and native helix (Asp23, Lys28, and Leu34) residues. These new findings on the inhibition of ß-sheet formation by targeting both N- and SHC-C-terminals, and the long-established target, D23-K28 salt bridge residues, not with the conventional central hydrophobic core (CHC) as reported in the literature, might aid in designing more potent inhibitors for AD treatment.


Assuntos
Doença de Alzheimer , Peptídeos beta-Amiloides , Humanos , Peptídeos beta-Amiloides/química , Antocianinas/farmacologia , Simulação de Dinâmica Molecular , Simulação de Acoplamento Molecular , Fragmentos de Peptídeos/química , Doença de Alzheimer/metabolismo
15.
J Mol Biol ; 436(6): 168464, 2024 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-38311235

RESUMO

Alzheimer's disease (AD) is linked with the self-association of the amyloid-ß peptide (Aß) into oligomers and fibrils. The brain is a lipid rich environment for Aß to assemble, while the brain membrane composition varies in an age dependent manner, we have therefore monitored the influence of lipid bilayer composition on the kinetics of Aß40 fibril assembly. Using global-fitting models of fibril formation kinetics, we show that the microscopic rate constant for primary nucleation is influenced by variations in phospholipid composition. Anionic phospholipids and particularly those with smaller headgroups shorten fibril formation lag-times, while zwitterionic phospholipids tend to extend them. Using a physiological vesicle model, we show cellular derived exosomes accelerate Aß40 and Aß42 fibril formation. Two distinct effects are observed, the presence of even small amounts of any phospholipid will impact the slope of the fibril growth curve. While subsequent additions of phospholipids only affect primary nucleation with the associated change in lag-times. Heightened anionic phospholipids and cholesterol levels are associated with aging and AD respectively, both these membrane components strongly accelerate primary nucleation during Aß assembly, making a link between disrupted lipid metabolism and Alzheimer's disease.


Assuntos
Doença de Alzheimer , Exossomos , Fragmentos de Peptídeos , Fosfolipídeos , Humanos , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/química , Exossomos/metabolismo , Cinética , Fragmentos de Peptídeos/química , Fosfolipídeos/química
16.
Proc Natl Acad Sci U S A ; 121(7): e2220075121, 2024 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-38335256

RESUMO

Self-replication of amyloid fibrils via secondary nucleation is an intriguing physicochemical phenomenon in which existing fibrils catalyze the formation of their own copies. The molecular events behind this fibril surface-mediated process remain largely inaccessible to current structural and imaging techniques. Using statistical mechanics, computer modeling, and chemical kinetics, we show that the catalytic structure of the fibril surface can be inferred from the aggregation behavior in the presence and absence of a fibril-binding inhibitor. We apply our approach to the case of Alzheimer's A[Formula: see text] amyloid fibrils formed in the presence of proSP-C Brichos inhibitors. We find that self-replication of A[Formula: see text] fibrils occurs on small catalytic sites on the fibril surface, which are far apart from each other, and each of which can be covered by a single Brichos inhibitor.


Assuntos
Peptídeos beta-Amiloides , Amiloide , Peptídeos beta-Amiloides/química , Amiloide/química , Simulação por Computador , Fragmentos de Peptídeos/química , Cinética
17.
J Phys Chem B ; 128(8): 1843-1853, 2024 Feb 29.
Artigo em Inglês | MEDLINE | ID: mdl-38359305

RESUMO

The misfolding and aggregation of amyloid-ß (Aß) peptides play a pivotal role in the pathogenesis of Alzheimer's disease (AD). Aß40 and Aß42, the two primary isoforms of Aß, can not only self-aggregate into homogeneous aggregates but also coaggregate to form mixed fibrils. Epigallocatechin-3-gallate (EGCG), a prominent tea polyphenol, has shown the capability to prevent the self-aggregation of Aß40 and Aß42 peptides and disaggregate their homogeneous fibrils. However, its effects on the cofibrillation of Aß40 and Aß42 have not yet been explored. Here, we employed molecular dynamic simulations to investigate the effects of EGCG on the coaggregation of Aß40 and Aß42, as well as on their mixed fibril. Our findings indicated that EGCG effectively inhibits the codimerization of Aß40 and Aß42 primarily by impeding the interchain interaction between the two isoforms. The key binding sites for EGCG on Aß40 and Aß42 are the polar residues and aromatic residues, engaging in hydrogen-bond , π-π, and cation-π interactions with EGCG. Additionally, EGCG disaggregates the Aß40-Aß42 mixed fibril by reducing its long-range interaction through similar binding sites and interactions as those between EGCG and Aß40-Aß42 heterodimers. Our research reveals the comprehensive inhibition and disaggregation effects of EGCG on the cofibrillation of Aß isoforms, which provides further support for the development of EGCG as an effective antiaggregation agent for AD.


Assuntos
Doença de Alzheimer , Catequina/análogos & derivados , Fragmentos de Peptídeos , Humanos , Fragmentos de Peptídeos/química , Peptídeos beta-Amiloides/química , Doença de Alzheimer/metabolismo , Isoformas de Proteínas
18.
Biophys Chem ; 307: 107165, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38309218

RESUMO

A major hallmark of Alzheimer's disease is the accumulation of aggregated amyloid ß peptide (Aß) in the brain. Here we develop a solubility assay for proteins and measure the solubility of Aß40. In brief, the method utilizes 96-well filter plates to separate monomeric Aß from aggregated Aß, and the small species are quantified with the amine reactive dye o-phthalaldehyde (OPA). This procedure ensures that solubility is measured for unlabeled species, and makes the assay high-throughput and inexpensive. We demonstrate that the filter plates successfully separate fibrils from monomer, with negligible monomer adsorption, and that OPA can quantify Aß peptides in a concentration range from 40 nM to 20 µM. We also show that adding a methionine residue to the N-terminus of Aß1-40 decreases the solubility by <3-fold. The method will facilitate further solubility studies, and contribute to the understanding of the thermodynamics of amyloid fibril formation.


Assuntos
Doença de Alzheimer , Peptídeos beta-Amiloides , Humanos , Peptídeos beta-Amiloides/química , Fragmentos de Peptídeos/química , Solubilidade , Doença de Alzheimer/metabolismo , Termodinâmica , Amiloide/química
19.
Sensors (Basel) ; 24(4)2024 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-38400369

RESUMO

Amyloid plays a critical role in the pathogenesis of Alzheimer's disease (AD) and can aggregate to form oligomers and fibrils in the brain. There is increasing evidence that highly toxic amyloid-ß oligomers (AßOs) lead to tau protein aggregation, hyperphosphorylation, neuroinflammation, neuronal loss, synaptic loss, and dysfunction. Although the effects of AßOs on neurons have been investigated using conventional biochemical experiments, there are no established criteria for electrical evaluation. To this end, we explored electrophysiological changes in mouse hippocampal neurons (HT22) following exposure to AßOs and/or naringenin (Nar, a flavonoid compound) using electrical impedance spectroscopy (EIS). AßO-induced HT22 showed a decreased impedance amplitude and increased phase angle, and the addition of Nar reversed these changes. The characteristic frequency was markedly increased with AßO exposure, which was also reversed by Nar. The AßOs decreased intranuclear and cytoplasmic resistance and increased nucleus resistance and extracellular capacitance. Overall, the innovative construction of the eight-element CPE-equivalent circuit model further reflects that the pseudo-capacitance of the cell membrane and cell nucleus was increased in the AßO-induced group. This study conclusively revealed that AßOs induce cytotoxic effects by disrupting the resistance characteristics of unit membranes. The results further support that EIS is an effective technique for evaluating AßO-induced neuronal damage and microscopic electrical distinctions in the sub-microscopic structure of reactive cells.


Assuntos
Doença de Alzheimer , Peptídeos beta-Amiloides , Camundongos , Animais , Peptídeos beta-Amiloides/química , Impedância Elétrica , Doença de Alzheimer/patologia , Neurônios/metabolismo , Sinapses/metabolismo , Sinapses/patologia
20.
J Am Chem Soc ; 146(9): 6045-6052, 2024 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-38394622

RESUMO

Many cellular coassemblies of proteins and polynucleotides facilitate liquid-liquid phase separation (LLPS) and the subsequent self-assembly of disease-associated amyloid fibrils within the liquid droplets. Here, we explore the dynamics of coupled phase and conformational transitions of model adenosine triphosphate (ATP)-binding peptides, ACC1-13Kn, consisting of the potent amyloidogenic fragment of insulin's A-chain (ACC1-13) merged with oligolysine segments of various lengths (Kn, n = 16, 24, 40). The self-assembly of ATP-stabilized amyloid fibrils is preceded by LLPS for peptides with sufficiently long oligolysine segments. The two-component droplets and fibrils are in dynamic equilibria with free ATP and monomeric peptides, which makes them susceptible to ATP-hydrolyzing apyrase and ACC1-13Kn-digesting proteinase K. Both enzymes are capable of rapid disassembly of amyloid fibrils, producing either monomers of the peptide (apyrase) or free ATP released together with cleaved-off oligolysine segments (proteinase K). In the latter case, the enzyme-sequestered Kn segments form subsequent droplets with the co-released ATP, resulting in an unusual fibril-to-droplet transition. In support of the highly dynamic nature of the aggregate-monomer equilibria, addition of superstoichiometric amounts of free peptide to the ACC1-13Kn-ATP coaggregate causes its disassembly. Our results show that the droplet state is not merely an intermediate phase on the pathway to the amyloid aggregate but may also constitute the final phase of a complex amyloidogenic protein misfolding scenario rich in highly degraded protein fragments incompetent to transition again into fibrils.


Assuntos
Trifosfato de Adenosina , Apirase , Endopeptidase K , Peptídeos , Amiloide/química , Peptídeos beta-Amiloides/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA