Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 158
Filtrar
1.
J Mol Biol ; 433(13): 166993, 2021 06 25.
Artigo em Inglês | MEDLINE | ID: mdl-33865867

RESUMO

It is known that insulin-degrading-enzyme (IDE) plays a crucial role in the clearance of Alzheimer's amyloid-ß (Aß). The cysteine-free IDE mutant (cf-E111Q-IDE) is catalytically inactive against insulin, but its effect on Aß degradation is unknown that would help in the allosteric modulation of the enzyme activity. Herein, the degradation of Aß(1-40) by cf-E111Q-IDE via a non-chaperone mechanism is demonstrated by NMR and LC-MS, and the aggregation of fragmented peptides is characterized using fluorescence and electron microscopy. cf-E111Q-IDE presented a reduced effect on the aggregation kinetics of Aß(1-40) when compared with the wild-type IDE. Whereas LC-MS and diffusion ordered NMR spectroscopy revealed the generation of Aß fragments by both wild-type and cf-E111Q-IDE. The aggregation propensities and the difference in the morphological phenotype of the full-length Aß(1-40) and its fragments are explained using multi-microseconds molecular dynamics simulations. Notably, our results reveal that zinc binding to Aß(1-40) inactivates cf-E111Q-IDE's catalytic function, whereas zinc removal restores its function as evidenced from high-speed AFM, electron microscopy, chromatography, and NMR results. These findings emphasize the catalytic role of cf-E111Q-IDE on Aß degradation and urge the development of zinc chelators as an alternative therapeutic strategy that switches on/off IDE's function.


Assuntos
Doença de Alzheimer/metabolismo , Insulisina/metabolismo , Proteínas Mutantes/metabolismo , Sequência de Aminoácidos , Peptídeos beta-Amiloides/química , Peptídeos beta-Amiloides/metabolismo , Peptídeos beta-Amiloides/ultraestrutura , Biocatálise , Cromatografia Líquida de Alta Pressão , Humanos , Insulisina/química , Insulisina/genética , Espectrometria de Massas , Microscopia Eletrônica de Transmissão , Simulação de Dinâmica Molecular , Proteínas Mutantes/química , Proteínas Mutantes/genética , Mutação de Sentido Incorreto , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/metabolismo , Fragmentos de Peptídeos/ultraestrutura , Ligação Proteica , Proteólise , Especificidade por Substrato , Zinco/química , Zinco/metabolismo
2.
Biochem Biophys Res Commun ; 534: 950-956, 2021 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-33143872

RESUMO

Peptide sequence modulates amyloid fibril formation and triggers Alzheimer's disease. The N-terminal region of amyloid peptide is disordered and lack any specific secondary structure. An ionic interaction of Aß1-11 with factor XII is critical for the activation of the contact system in Alzheimer's disease. In this study, we report the self-assembly of fluctuating N-terminal Aß1-11 into nanotubes using atomic force micrography, transmission electron microscopy, circular dichroism studies and molecular modeling studies. The effect of four polyphenols: baicalein, rutin, vanillin and cyanidin-3-O-glucoside (C3G) was also explored on the amyloid fibril inhibitor perspective using amyloid specific dye Thioflavin T (ThT). AFM micrographs suggested the self-assembly of Aß1-11 into nanotubes after three weeks of incubation. Microwave treatment results in the conformational variation of disordered structure to ß-sheet rich amyloid fibrils. The presence of salts (sodium and potassium chloride) induces the structural transformation of Aß1-11 to super-helix. Fluorescence spectroscopy studies using ThT suggested differential inhibition of amyloid fibrils formation in the presence of polyphenols. Molecular modeling studies suggested that binding of polyphenols to Aß1-11 through hydrophobic interaction (Phe4 and Tyr 10) and hydrogen bonding (Glu3 and Arg5) play a substantial role in stabilizing Aß1-11-polyphenols complex. In the presence of polyphenols, Aß1-11 transforms to hybrid nanostructures thus hindering amyloid fibril formation. These results provide structural insights and importance of the N-terminal residues in the Aß1-42 self-assembly mechanism.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Fragmentos de Peptídeos/metabolismo , Doença de Alzheimer/tratamento farmacológico , Peptídeos beta-Amiloides/análise , Peptídeos beta-Amiloides/antagonistas & inibidores , Peptídeos beta-Amiloides/ultraestrutura , Humanos , Modelos Moleculares , Fragmentos de Peptídeos/análise , Fragmentos de Peptídeos/antagonistas & inibidores , Fragmentos de Peptídeos/ultraestrutura , Polifenóis/farmacologia , Estrutura Secundária de Proteína/efeitos dos fármacos
3.
PLoS Biol ; 18(11): e3000925, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-33216759

RESUMO

Lifeact is a short actin-binding peptide that is used to visualize filamentous actin (F-actin) structures in live eukaryotic cells using fluorescence microscopy. However, this popular probe has been shown to alter cellular morphology by affecting the structure of the cytoskeleton. The molecular basis for such artefacts is poorly understood. Here, we determined the high-resolution structure of the Lifeact-F-actin complex using electron cryo-microscopy (cryo-EM). The structure reveals that Lifeact interacts with a hydrophobic binding pocket on F-actin and stretches over 2 adjacent actin subunits, stabilizing the DNase I-binding loop (D-loop) of actin in the closed conformation. Interestingly, the hydrophobic binding site is also used by actin-binding proteins, such as cofilin and myosin and actin-binding toxins, such as the hypervariable region of TccC3 (TccC3HVR) from Photorhabdus luminescens and ExoY from Pseudomonas aeruginosa. In vitro binding assays and activity measurements demonstrate that Lifeact indeed competes with these proteins, providing an explanation for the altering effects of Lifeact on cell morphology in vivo. Finally, we demonstrate that the affinity of Lifeact to F-actin can be increased by introducing mutations into the peptide, laying the foundation for designing improved actin probes for live cell imaging.


Assuntos
Actinas/química , Proteínas dos Microfilamentos/química , Actinas/metabolismo , Actinas/ultraestrutura , Animais , Toxinas Bacterianas/química , Sítios de Ligação , Ligação Competitiva , Cofilina 1/química , Cofilina 1/ultraestrutura , Microscopia Crioeletrônica , Corantes Fluorescentes/química , Células HEK293 , Humanos , Interações Hidrofóbicas e Hidrofílicas , Técnicas In Vitro , Proteínas dos Microfilamentos/metabolismo , Proteínas dos Microfilamentos/ultraestrutura , Microscopia Confocal , Modelos Moleculares , Miosinas/química , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/ultraestrutura , Engenharia de Proteínas , Domínios e Motivos de Interação entre Proteínas , Coelhos , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/ultraestrutura , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/ultraestrutura
4.
Biochem Biophys Res Commun ; 533(1): 125-131, 2020 11 26.
Artigo em Inglês | MEDLINE | ID: mdl-32943189

RESUMO

Revealing the aggregation and fibrillation process of variant amyloid proteins is critical for understanding the molecular mechanism of related amyloidosis diseases. Here we characterized the fibrillation morphology and kinetics of type 2 diabetes (T2D) related human islet amyloid polypeptide (hIAPP1-37) fibril formation process using negative staining transmission electron microscopy (NS-TEM), cryo-electron microscopy (cryo-EM) analysis, and 3D cryo-electron tomography (cryo-ET) reconstruction, together with circular dichroism (CD) and Thioflavin-T (ThT) assays. Our results showed that various amyloid fibrils can be observed at different time points of hIAPP1-37 fibrillization process, while the winding of protofibrils presents in different growth stages, which suggests a synchronous process of hIAPP1-37 amyloid fibrillization. This work provides insights into the understanding of hIAPP1-37 amyloid aggregation process and the pathogenesis of Type 2 diabetes disease.


Assuntos
Amiloide/metabolismo , Polipeptídeo Amiloide das Ilhotas Pancreáticas/metabolismo , Amiloide/química , Amiloide/ultraestrutura , Microscopia Crioeletrônica , Diabetes Mellitus Tipo 2/metabolismo , Humanos , Polipeptídeo Amiloide das Ilhotas Pancreáticas/química , Polipeptídeo Amiloide das Ilhotas Pancreáticas/ultraestrutura , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/metabolismo , Fragmentos de Peptídeos/ultraestrutura , Agregação Patológica de Proteínas/metabolismo
5.
Commun Biol ; 3(1): 484, 2020 09 02.
Artigo em Inglês | MEDLINE | ID: mdl-32879439

RESUMO

Self-assembly of proteins into amyloid fibrils is a hallmark of various diseases, including Alzheimer's disease (AD) and Type-2 diabetes Mellitus (T2DM). Aggregation of specific peptides, like Aß42 in AD and hIAPP in T2DM, causes cellular dysfunction resulting in the respective pathology. While these amyloidogenic proteins lack sequence homology, they all contain aromatic amino acids in their hydrophobic core that play a major role in their self-assembly. Targeting these aromatic residues by small molecules may be an attractive approach for inhibiting amyloid aggregation. Here, various biochemical and biophysical techniques revealed that a panel of tryptophan-galactosylamine conjugates significantly inhibit fibril formation of Aß42 and hIAPP, and disassemble their pre-formed fibrils in a dose-dependent manner. They are also not toxic to mammalian cells and can reduce the cytotoxicity induced by Aß42 and hIAPP aggregates. These tryptophan-galactosylamine conjugates can therefore serve as a scaffold for the development of therapeutics towards AD and T2DM.


Assuntos
Peptídeos beta-Amiloides/toxicidade , Amiloide/metabolismo , Galactosamina/metabolismo , Polipeptídeo Amiloide das Ilhotas Pancreáticas/toxicidade , Fragmentos de Peptídeos/toxicidade , Agregados Proteicos , Triptofano/metabolismo , Sequência de Aminoácidos , Amiloide/ultraestrutura , Peptídeos beta-Amiloides/química , Peptídeos beta-Amiloides/ultraestrutura , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Humanos , Concentração Inibidora 50 , Polipeptídeo Amiloide das Ilhotas Pancreáticas/química , Polipeptídeo Amiloide das Ilhotas Pancreáticas/ultraestrutura , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/ultraestrutura , Agregados Proteicos/efeitos dos fármacos
6.
Sci Rep ; 10(1): 10332, 2020 06 25.
Artigo em Inglês | MEDLINE | ID: mdl-32587293

RESUMO

Atypical low-oxidation-state iron phases in Alzheimer's disease (AD) pathology are implicated in disease pathogenesis, as they may promote elevated redox activity and convey toxicity. However, the origin of low-oxidation-state iron and the pathways responsible for its formation and evolution remain unresolved. Here we investigate the interaction of the AD peptide ß-amyloid (Aß) with the iron storage protein ferritin, to establish whether interactions between these two species are a potential source of low-oxidation-state iron in AD. Using X-ray spectromicroscopy and electron microscopy we found that the co-aggregation of Aß and ferritin resulted in the conversion of ferritin's inert ferric core into more reactive low-oxidation-states. Such findings strongly implicate Aß in the altered iron handling and increased oxidative stress observed in AD pathogenesis. These amyloid-associated iron phases have biomarker potential to assist with disease diagnosis and staging, and may act as targets for therapies designed to lower oxidative stress in AD tissue.


Assuntos
Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Ferritinas/metabolismo , Ferro/metabolismo , Fragmentos de Peptídeos/metabolismo , Doença de Alzheimer/diagnóstico , Peptídeos beta-Amiloides/ultraestrutura , Biomarcadores/química , Biomarcadores/metabolismo , Ferritinas/química , Ferritinas/ultraestrutura , Humanos , Ferro/química , Microscopia Eletrônica de Transmissão e Varredura , Oxirredução , Estresse Oxidativo , Fragmentos de Peptídeos/ultraestrutura , Agregados Proteicos , Espectrometria por Raios X
7.
Sci Rep ; 10(1): 10356, 2020 06 25.
Artigo em Inglês | MEDLINE | ID: mdl-32587390

RESUMO

Human pancreatic islet amyloid polypeptide (hIAPP) and beta amyloid (Aß) can accumulate in Type 2 diabetes (T2D) and Alzheimer's disease (AD) brains and evidence suggests that interaction between the two amyloidogenic proteins can lead to the formation of heterocomplex aggregates. However, the structure and consequences of the formation of these complexes remains to be determined. The main objective of this study was to characterise the different types and morphology of Aß-hIAPP heterocomplexes and determine if formation of such complexes exacerbate neurotoxicity. We demonstrate that hIAPP promotes Aß oligomerization and formation of small oligomer and large aggregate heterocomplexes. Co-oligomerized Aß42-hIAPP mixtures displayed distinct amorphous structures and a 3-fold increase in neuronal cell death as compared to Aß and hIAPP alone. However, in contrast to hIAPP, non-amyloidogenic rat amylin (rIAPP) reduced oligomer Aß-mediated neuronal cell death. rIAPP exhibited reductions in Aß induced neuronal cell death that was independent of its ability to interact with Aß and form heterocomplexes; suggesting mediation by other pathways. Our findings reveal distinct effects of IAPP peptides in modulating Aß aggregation and toxicity and provide new insight into the potential pathogenic effects of Aß-IAPP hetero-oligomerization and development of IAPP based therapies for AD and T2D.


Assuntos
Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Diabetes Mellitus Tipo 2/patologia , Polipeptídeo Amiloide das Ilhotas Pancreáticas/metabolismo , Neurônios/patologia , Fragmentos de Peptídeos/metabolismo , Agregação Patológica de Proteínas/patologia , Doença de Alzheimer/etiologia , Peptídeos beta-Amiloides/toxicidade , Peptídeos beta-Amiloides/ultraestrutura , Animais , Encéfalo/patologia , Linhagem Celular Tumoral , Diabetes Mellitus Tipo 2/complicações , Humanos , Polipeptídeo Amiloide das Ilhotas Pancreáticas/toxicidade , Polipeptídeo Amiloide das Ilhotas Pancreáticas/ultraestrutura , Microscopia Eletrônica de Transmissão , Ressonância Magnética Nuclear Biomolecular , Pâncreas/metabolismo , Fragmentos de Peptídeos/toxicidade , Fragmentos de Peptídeos/ultraestrutura , Agregados Proteicos , Multimerização Proteica , Ratos
8.
Proc Natl Acad Sci U S A ; 117(21): 11265-11273, 2020 05 26.
Artigo em Inglês | MEDLINE | ID: mdl-32439711

RESUMO

The nucleation of Alzheimer-associated Aß peptide monomers can be catalyzed by preexisting Aß fibrils. This leads to autocatalytic amplification of aggregate mass and underlies self-replication and generation of toxic oligomers associated with several neurodegenerative diseases. However, the nature of the interactions between the monomeric species and the fibrils during this key process, and indeed the ultrastructural localization of the interaction sites have remained elusive. Here we used NMR and optical spectroscopy to identify conditions that enable the capture of transient species during the aggregation and secondary nucleation of the Aß42 peptide. Cryo-electron microscopy (cryo-EM) images show that new aggregates protrude from the entire length of the progenitor fibril. These protrusions are morphologically distinct from the well-ordered fibrils dominating at the end of the aggregation process. The data provide direct evidence that self-replication through secondary nucleation occurs along the sides of fibrils, which become heavily decorated under the current solution conditions (14 µM Aß42, 20 mM sodium phosphate, 200 µM EDTA, pH 6.8).


Assuntos
Peptídeos beta-Amiloides/metabolismo , Peptídeos beta-Amiloides/ultraestrutura , Fragmentos de Peptídeos/metabolismo , Fragmentos de Peptídeos/ultraestrutura , Doença de Alzheimer/patologia , Amiloide/metabolismo , Amiloide/ultraestrutura , Peptídeos beta-Amiloides/química , Benzotiazóis/química , Benzotiazóis/metabolismo , Microscopia Crioeletrônica , Humanos , Concentração de Íons de Hidrogênio , Cinética , Espectroscopia de Ressonância Magnética , Fragmentos de Peptídeos/química , Imagem com Lapso de Tempo
9.
FEBS J ; 287(10): 2134-2149, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-31659864

RESUMO

Celiac disease (CeD) is a highly prevalent chronic immune-mediated enteropathy developed in genetically predisposed individuals after ingestion of a group of wheat proteins (called gliadins and glutenins). The 13mer α-gliadin peptide, p31-43, induces proinflammatory responses, observed by in vitro assays and animal models, that may contribute to innate immune mechanisms of CeD pathogenesis. Since a cellular receptor for p31-43 has not been identified, this raises the question of whether this peptide could mediate different biological effects. In this work, we aimed to characterize the p31-43 secondary structure by different biophysical and in silico techniques. By dynamic light scattering and using an oligomer/fibril-sensitive fluorescent probe, we showed the presence of oligomers of this peptide in solution. Furthermore, atomic force microscopy analysis showed p31-43 oligomers with different height distribution. Also, peptide concentration had a very strong influence on peptide self-organization process. Oligomers gradually increased their size at lower concentration. Whereas, at higher ones, oligomers increased their complexity, forming branched structures. By CD, we observed that p31-43 self-organized in a polyproline II conformation in equilibrium with ß-sheets-like structures, whose pH remained stable in the range of 3-8. In addition, these findings were supported by molecular dynamics simulation. The formation of p31-43 nanostructures with increased ß-sheet structure may help to explain the molecular etiopathogenesis in the induction of proinflammatory effects and subsequent damage at the intestinal mucosa in CeD.


Assuntos
Doença Celíaca/tratamento farmacológico , Gliadina/farmacologia , Imunidade Inata/efeitos dos fármacos , Fragmentos de Peptídeos/farmacologia , Células CACO-2 , Doença Celíaca/genética , Doença Celíaca/imunologia , Doença Celíaca/patologia , Gliadina/genética , Gliadina/imunologia , Gliadina/ultraestrutura , Humanos , Imunidade Inata/imunologia , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/imunologia , Microscopia de Força Atômica , Conformação Molecular/efeitos dos fármacos , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/imunologia , Fragmentos de Peptídeos/ultraestrutura , Peptídeos/química , Peptídeos/imunologia , Peptídeos/farmacologia , Conformação Proteica em Folha beta , Soluções/química , Água/química
10.
Eur J Med Chem ; 158: 393-404, 2018 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-30227353

RESUMO

One of the pathologic hallmarks in Alzheimer's disease (AD) is extracellular senile plaques composed of amyloid-ß (Aß) fibrils. Blocking Aß self-assembly or disassembling Aß aggregates by small molecules would be potential therapeutic strategies to treat AD. In this study, we synthesized a series of rationally designed divalent compounds and examined their effects on Aß fibrillization. A divalent amide (2) derived from two molecules of caffeic acid with a propylenediamine linker of ∼5.0 Šin length, which is close to the distance of adjacent ß sheets in Aß fibrils, showed good potency to inhibit Aß(1-42) fibrillization. Furthermore, compound 2 effectively dissociated the Aß(1-42) preformed fibrils. The cytotoxicity induced by Aß(1-42) aggregates in human neuroblastoma was reduced in the presence of 2, and feeding 2 to Aß transgenic C. elegans rescued the paralysis phenotype. In addition, the binding and stoichiometry of 2 to Aß(1-40) were demonstrated by using electrospray ionization-traveling wave ion mobility-mass spectrometry, while molecular dynamic simulation was conducted to gain structural insights into the Aß(1-40)-2 complex.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Ácidos Cafeicos/química , Ácidos Cafeicos/farmacologia , Fragmentos de Peptídeos/metabolismo , Doença de Alzheimer/tratamento farmacológico , Amidas/química , Amidas/farmacologia , Amidas/uso terapêutico , Peptídeos beta-Amiloides/ultraestrutura , Animais , Caenorhabditis elegans , Ácidos Cafeicos/uso terapêutico , Humanos , Modelos Moleculares , Fragmentos de Peptídeos/ultraestrutura , Multimerização Proteica/efeitos dos fármacos
11.
J Biol Chem ; 293(10): 3734-3746, 2018 03 09.
Artigo em Inglês | MEDLINE | ID: mdl-29358329

RESUMO

Huntingtin N-terminal fragments (Htt-NTFs) with expanded polyglutamine tracts form a range of neurotoxic aggregates that are associated with Huntington's disease. Here, we show that aggregation of Htt-NTFs, irrespective of polyglutamine length, yields at least three phases (designated M, S, and F) that are delineated by sharp concentration thresholds and distinct aggregate sizes and morphologies. We found that monomers and oligomers make up the soluble M phase, ∼25-nm spheres dominate in the soluble S phase, and long, linear fibrils make up the insoluble F phase. Previous studies showed that profilin, an abundant cellular protein, reduces Htt-NTF aggregation and toxicity in cells. We confirm that profilin achieves its cellular effects through direct binding to the C-terminal proline-rich region of Htt-NTFs. We show that profilin preferentially binds to Htt-NTF M-phase species and destabilizes aggregation and phase separation by shifting the concentration boundaries for phase separation to higher values through a process known as polyphasic linkage. Our experiments, aided by coarse-grained computer simulations and theoretical analysis, suggest that preferential binding of profilin to the M-phase species of Htt-NTFs is enhanced through a combination of specific interactions between profilin and polyproline segments and auxiliary interactions between profilin and polyglutamine tracts. Polyphasic linkage may be a general strategy that cells utilize to regulate phase behavior of aggregation-prone proteins. Accordingly, detailed knowledge of phase behavior and an understanding of how ligands modulate phase boundaries may pave the way for developing new therapeutics against a variety of aggregation-prone proteins.


Assuntos
Proteína Huntingtina/metabolismo , Modelos Moleculares , Profilinas/metabolismo , Agregação Patológica de Proteínas/prevenção & controle , Substituição de Aminoácidos , Sítios de Ligação , Fluorescência , Humanos , Proteína Huntingtina/química , Proteína Huntingtina/genética , Proteína Huntingtina/ultraestrutura , Processamento de Imagem Assistida por Computador , Ligantes , Microscopia Eletrônica de Transmissão , Mutação , Coloração Negativa , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Fragmentos de Peptídeos/ultraestrutura , Ácido Poliglutâmico/química , Ácido Poliglutâmico/genética , Ácido Poliglutâmico/metabolismo , Profilinas/química , Profilinas/genética , Profilinas/ultraestrutura , Domínios Proteicos Ricos em Prolina , Agregação Patológica de Proteínas/metabolismo , Agregação Patológica de Proteínas/patologia , Estabilidade Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/ultraestrutura , Espalhamento a Baixo Ângulo , Solubilidade , Termodinâmica , Triptofano/química
12.
ACS Chem Neurosci ; 8(9): 1889-1900, 2017 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-28581708

RESUMO

Amyloid-beta (Aß) oligomers are thought to be causative for the development and progression of Alzheimer's disease (AD). Starting from the Aß oligomer eliminating d-enantiomeric peptide D3, we developed and applied a two-step procedure based on peptide microarrays to identify D3 derivatives with increased binding affinity and specificity for monomeric Aß(1-42) to further enhance the Aß oligomer elimination efficacy. Out of more than 1000 D3 derivatives, we selected seven novel d-peptides, named ANK1 to ANK7, and characterized them in more detail in vitro. All ANK peptides bound to monomeric Aß(1-42), eliminated Aß(1-42) oligomers, inhibited Aß(1-42) fibril formation, and reduced Aß(1-42)-induced cytotoxicity more efficiently than D3. Additionally, ANK6 completely inhibited the prion-like propagation of preformed Aß(1-42) seeds and showed a nonsignificant tendency for improving memory performance of tg-APPSwDI mice after i.p. application for 4 weeks. This supports the hypothesis that stabilization of Aß monomers and thereby induced elimination of Aß oligomers is a suitable therapeutic strategy.


Assuntos
Peptídeos beta-Amiloides/antagonistas & inibidores , Peptídeos beta-Amiloides/metabolismo , Fármacos Neuroprotetores/farmacologia , Nootrópicos/farmacologia , Oligopeptídeos/farmacologia , Fragmentos de Peptídeos/antagonistas & inibidores , Fragmentos de Peptídeos/metabolismo , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Doença de Alzheimer/psicologia , Peptídeos beta-Amiloides/toxicidade , Peptídeos beta-Amiloides/ultraestrutura , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Animais Geneticamente Modificados , Ligação Competitiva , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Descoberta de Drogas , Feminino , Humanos , Camundongos Endogâmicos C57BL , Análise em Microsséries , Fragmentos de Peptídeos/toxicidade , Fragmentos de Peptídeos/ultraestrutura , Placa Amiloide/tratamento farmacológico , Placa Amiloide/metabolismo , Placa Amiloide/patologia , Presenilina-1/genética , Presenilina-1/metabolismo , Agregação Patológica de Proteínas/tratamento farmacológico , Agregação Patológica de Proteínas/metabolismo , Agregação Patológica de Proteínas/patologia , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/toxicidade , Proteínas Recombinantes/ultraestrutura
13.
Neurosci Bull ; 33(4): 405-412, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28555357

RESUMO

Accumulation and aggregation of ß-amyloid (Aß) peptides result in neuronal death, leading to cognitive dysfunction in Alzheimer's disease. The self-assembled Aß molecules form various intermediate aggregates including oligomers that are more toxic to neurons than the mature aggregates, including fibrils. Thus, one strategy to alleviate Aß toxicity is to facilitate the conversion of Aß intermediates to larger aggregates such as fibrils. In this study, we designed a peptide named A3 that significantly enhanced the formation of amorphous aggregates of Aß by accelerating the aggregation kinetics. Thioflavin T fluorescence experiments revealed an accelerated aggregation of Aß monomers, accompanying reduced Aß cytotoxicity. Transgenic Caenorhabditis elegans over-expressing amyloid precursor protein exhibited paralysis due to the accumulation of Aß oligomers, and this phenotype was attenuated by feeding the animals with A3 peptide. These findings suggest that the Aß aggregation-promotion effect can potentially be useful for developing strategies to reduce Aß toxicity.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/toxicidade , Fragmentos de Peptídeos/metabolismo , Agregação Patológica de Proteínas/induzido quimicamente , Peptídeos beta-Amiloides/genética , Peptídeos beta-Amiloides/ultraestrutura , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Animais Geneticamente Modificados , Caenorhabditis elegans , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Humanos , Cinética , Neuroblastoma/patologia , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/ultraestrutura , Agregação Patológica de Proteínas/genética , Transfecção
14.
Eur J Med Chem ; 133: 184-196, 2017 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-28388521

RESUMO

A series of new donepezil derivatives were designed synthesized and evaluated as multifunctional cholinesterase inhibitors against Alzheimer's disease (AD). In vitro studies showed that most of them exhibited significant potency to inhibit acetylcholinesterase and self-induced ß-amyloid (Aß) aggregation, and moderate antioxidant activity. Especially, compound 5b presented the greatest ability to inhibit cholinesterase (IC50, 1.9 nM for eeAChE and 0.8 nM for hAChE), good inhibition of Aß aggregation (53.7% at 20 µM) and good antioxidant activity (0.54 trolox equivalents). Kinetic and molecular modeling studies indicated that compound 5b was a mixed-type inhibitor, binding simultaneously to the catalytic active site (CAS) and the peripheral anionic site (PAS) of AChE. In addition, compound 5b could reduce PC12 cells death induced by oxidative stress and Aß (1-42). Moreover, in vivo experiments showed that compound 5b was nontoxic and tolerated at doses up to 2000 mg/kg. These results suggested that compound 5b might be an excellent multifunctional agent for AD treatment.


Assuntos
Acetilcolinesterase/metabolismo , Peptídeos beta-Amiloides/antagonistas & inibidores , Inibidores da Colinesterase/química , Inibidores da Colinesterase/farmacologia , Indanos/química , Indanos/farmacologia , Fragmentos de Peptídeos/antagonistas & inibidores , Piperidinas/química , Piperidinas/farmacologia , Agregados Proteicos/efeitos dos fármacos , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Peptídeos beta-Amiloides/ultraestrutura , Animais , Barreira Hematoencefálica/metabolismo , Sobrevivência Celular , Inibidores da Colinesterase/farmacocinética , Donepezila , Desenho de Fármacos , Electrophorus , Humanos , Indanos/farmacocinética , Camundongos , Simulação de Acoplamento Molecular , Fármacos Neuroprotetores/química , Fármacos Neuroprotetores/farmacologia , Células PC12 , Fragmentos de Peptídeos/metabolismo , Fragmentos de Peptídeos/ultraestrutura , Piperidinas/farmacocinética , Compostos de Piridínio/química , Compostos de Piridínio/farmacocinética , Compostos de Piridínio/farmacologia , Ratos , Relação Estrutura-Atividade
15.
EMBO Mol Med ; 9(5): 703-715, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28356312

RESUMO

Alzheimer's disease (AD) is the most common neurodegenerative disease. Imbalance between the production and clearance of amyloid ß (Aß) peptides is considered to be the primary mechanism of AD pathogenesis. This amyloid hypothesis is supported by the recent success of the human anti-amyloid antibody aducanumab, in clearing plaque and slowing clinical impairment in prodromal or mild patients in a phase Ib trial. Here, a peptide combining polyarginines (polyR) (for charge repulsion) and a segment derived from the core region of Aß amyloid (for sequence recognition) was designed. The efficacy of the designed peptide, R8-Aß(25-35), on amyloid reduction and the improvement of cognitive functions were evaluated using APP/PS1 double transgenic mice. Daily intranasal administration of PEI-conjugated R8-Aß(25-35) peptide significantly reduced Aß amyloid accumulation and ameliorated the memory deficits of the transgenic mice. Intranasal administration is a feasible route for peptide delivery. The modular design combining polyR and aggregate-forming segments produced a desirable therapeutic effect and could be easily adopted to design therapeutic peptides for other proteinaceous aggregate-associated diseases.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Peptídeos beta-Amiloides/uso terapêutico , Encéfalo/efeitos dos fármacos , Disfunção Cognitiva/tratamento farmacológico , Fragmentos de Peptídeos/uso terapêutico , Peptídeos/uso terapêutico , Administração Intranasal , Doença de Alzheimer/complicações , Doença de Alzheimer/patologia , Amiloide/antagonistas & inibidores , Amiloide/ultraestrutura , Peptídeos beta-Amiloides/administração & dosagem , Peptídeos beta-Amiloides/antagonistas & inibidores , Peptídeos beta-Amiloides/química , Peptídeos beta-Amiloides/ultraestrutura , Animais , Encéfalo/patologia , Linhagem Celular , Cognição/efeitos dos fármacos , Disfunção Cognitiva/complicações , Disfunção Cognitiva/patologia , Modelos Animais de Doenças , Feminino , Transtornos da Memória/complicações , Transtornos da Memória/tratamento farmacológico , Transtornos da Memória/patologia , Camundongos Endogâmicos C57BL , Fragmentos de Peptídeos/administração & dosagem , Fragmentos de Peptídeos/antagonistas & inibidores , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/ultraestrutura , Peptídeos/administração & dosagem , Peptídeos/química
16.
J Alzheimers Dis ; 57(3): 857-871, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28282805

RESUMO

The 24-residue peptide humanin (HN) has been proposed as a peptide-based inhibitor able to interact directly with amyloid-ß (Aß) oligomers and interfere with the formation and/or biological properties of toxic Aß species. When administered exogenously, HN, or its synthetic S14G-derivative (HNG), exerted multiple cytoprotective effects, counteracting the Aß-induced toxicity. Whether these peptides interact directly with Aß, particularly with the soluble oligomeric assemblies, remains largely unknown. We here investigated the ability of HN and HNG to interact directly with highly aggregating Aß42, and interfere with the formation and toxicity of its oligomers. Experiments were run in cell-free conditions and in vivo in a transgenic C. elegans strain in which the Aß toxicity was specifically due to oligomeric species. Thioflavin-T assay indicated that both HN and HNG delay the formation and reduce the final amount of Aß42 fibrils. In vitro surface plasmon resonance studies indicated that they interact with Aß42 oligomers favoring the formation of amorphous larger assemblies, observed with turbidity and electron microscopy. In vivo studies indicated that both HN and HNG decrease the relative abundance of A11-positive prefibrillar oligomers as well as OC-positive fibrillar oligomers and had similar protective effects. However, while HN possibly decreased the oligomers by promoting their assembly into larger aggregates, the reduction of oligomers caused by HNG can be ascribed to a marked decrease of the total Aß levels, likely the consequence of the HNG-induced overexpression of the Aß-degrading enzyme neprilysin. These findings provide information on the mechanisms underlying the anti-oligomeric effects of HN and HNG and illustrate the role of S14G substitution in regulating the in vivo mechanism of action.


Assuntos
Peptídeos beta-Amiloides/toxicidade , Regulação da Expressão Gênica/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/uso terapêutico , Paralisia/induzido quimicamente , Paralisia/tratamento farmacológico , Peptídeos beta-Amiloides/química , Peptídeos beta-Amiloides/metabolismo , Peptídeos beta-Amiloides/ultraestrutura , Animais , Animais Geneticamente Modificados , Caenorhabditis elegans , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Dicroísmo Circular/métodos , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/química , Peptídeos e Proteínas de Sinalização Intracelular/farmacologia , Microscopia de Força Atômica , Microscopia Eletrônica de Transmissão , Neprilisina/genética , Neprilisina/metabolismo , Paralisia/metabolismo , Fragmentos de Peptídeos/farmacologia , Fragmentos de Peptídeos/ultraestrutura , Ressonância de Plasmônio de Superfície
17.
J Alzheimers Dis ; 56(3): 991-1005, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28106549

RESUMO

The goals of this work were i) to identify the interactions of amyloid-ß (Aß)42 under monomeric, oligomeric, and fibrillar forms with the extracellular matrix (ECM) and receptors, ii) to determine the influence of Aß42 supramolecular organization on these interactions, and iii) to identify the molecular functions, biological processes, and pathways targeted by Aß42 in the ECM. The ECM and cell surface partners of Aß42 and its supramolecular forms were identified with protein and glycosaminoglycan (GAG) arrays (81 molecules in triplicate) probed by surface plasmon resonance imaging. The number of partners of Aß42 increased upon its multimerization, ranging from 4 for the peptide up to 53 for the fibrillar aggregates. The peptide interacted only with ECM proteins but their percentage among Aß42 partners decreased upon multimerization. Aß42 and its supramolecular forms recognized different molecular features on their partners, and the partners of Aß42 fibrillar forms were enriched in laminin IV-A, N-terminal, and EGF-like domains. Aß42 oligomerization triggered interactions with receptors, whereas Aß42 fibrillogenesis promoted binding to GAGs, proteoglycans, enzymes, and growth factors and the ability to interact with perineuronal nets. Fibril aggregation bind to further membrane proteins including tumor endothelial marker-8, syndecan-4, and discoidin-domain receptor-2. The partners of the Aß42 supramolecular forms are enriched in proteins contributing to cell growth and/or maintenance, involved in integrin cell surface interactions and expressed in kidney cancer, preadipocytes, and dentin. In conclusion, the supramolecular assembly of Aß42 governs its ability to interact in vitro with ECM proteins, remodeling and crosslinking ECM enzymes, proteoglycans, and receptors.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Matriz Extracelular/metabolismo , Fragmentos de Peptídeos/metabolismo , Amiloide/metabolismo , Amiloide/ultraestrutura , Peptídeos beta-Amiloides/ultraestrutura , Células HEK293 , Humanos , Microscopia Eletrônica , Fragmentos de Peptídeos/ultraestrutura , Ligação Proteica , Multimerização Proteica , Ressonância de Plasmônio de Superfície
18.
Brain Res ; 1648(Pt A): 485-495, 2016 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-27531183

RESUMO

One pathological hallmark of Alzheimer's disease (AD) is the accumulation of amyloid-ß peptide (Aß) in the affected brain. While there are numerous deleterious effects of Aß accumulation, there is general agreement that a sustained inflammatory response to aggregated Aß contributes to progressive neurodegeneration in AD and microglial cells play a significant role in this process. Our laboratory and others have shown that small soluble aggregates of Aß activate a microglia-mediated inflammatory response. One component of the response involves internalization of extracellular Aß, and this process is likely very sensitive to Aß structure. In this study we analyzed the proclivity of microglia for internalization of Aß42 monomers and protofibrils using fluorescently-labeled Aß. Both Aß42 species were labeled directly via amino linkage with an Alexa Fluor 488 tetrafluorophenyl ester (AF488-TFP) and then isolated individually by chromatography. Aß42 protofibrils retained their size and morphological properties after labeling but monomers had a much higher stoichiometry of labeling compared to protofibrils. Primary murine microglia internalized AF488-Aß42 protofibrils rapidly and in significant amounts compared to AF488-Aß42 monomers. Microglial internalization of protofibrils was dependent on time and concentration, and corresponded with tumor necrosis factor α secretion. In competition studies, unlabeled Aß42 protofibril internalization, detected by immunostaining, did not diminish AF488-protofibril uptake. Internalized AF488-Aß42 protofibrils were found widely dispersed in the cytosol with some lysosomal accumulation but little degradation. These studies highlight the sensitivity that microglia exhibit to Aß structure in the internalization process and emphasize their affinity for soluble Aß protofibrils.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Microglia/metabolismo , Fragmentos de Peptídeos/metabolismo , Peptídeos beta-Amiloides/química , Peptídeos beta-Amiloides/ultraestrutura , Animais , Células Cultivadas , Camundongos , Camundongos Endogâmicos C57BL , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/ultraestrutura , Fator de Necrose Tumoral alfa/metabolismo
19.
ACS Chem Neurosci ; 7(8): 1088-96, 2016 08 17.
Artigo em Inglês | MEDLINE | ID: mdl-27240424

RESUMO

Alzheimer's disease (AD) is a neurodegenerative disorder and the most common type of dementia. Until now, there is no curative therapy available. Previously, we selected the amyloid-beta (Aß) targeting peptide D3 consisting of 12 d-enantiomeric amino acid residues by mirror image phage display as a potential drug candidate for the treatment of AD. In the current approach, we investigated the optimization potential of linear D3 with free C-terminus (D3COOH) by chemical modifications. First, the impact of the net charge was investigated and second, cyclization was introduced which is a well-known tool for the optimization of peptides for enhanced target affinity. Following this strategy, three D3 derivatives in addition to D3COOH were designed: C-terminally amidated linear D3 (D3CONH2), cyclic D3 (cD3), and cyclic D3 with an additional arginine residue (cD3r) to maintain the net charge of linear D3CONH2. These four compounds were compared to each other according to their binding affinities to Aß(1-42), their efficacy to eliminate cytotoxic oligomers, and consequently their potency to neutralize Aß(1-42) oligomer induced neurotoxicity. D3CONH2 and cD3r versions with equally increased net charge showed superior properties over D3COOH and cD3, respectively. The cyclic versions showed superior properties compared to their linear version with equal net charge, suggesting cD3r to be the most efficient compound among these four. Indeed, treatment of the transgenic AD mouse model Tg-SwDI with cD3r significantly enhanced spatial memory and cognition of these animals as revealed by water maze performance. Therefore, charge increase and cyclization imply suitable modification steps for an optimization approach of the Aß targeting compound D3.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Peptídeos beta-Amiloides/toxicidade , Fragmentos de Peptídeos/metabolismo , Fragmentos de Peptídeos/toxicidade , Peptídeos Cíclicos/química , Peptídeos Cíclicos/uso terapêutico , Doença de Alzheimer/complicações , Doença de Alzheimer/genética , Peptídeos beta-Amiloides/ultraestrutura , Precursor de Proteína beta-Amiloide/genética , Animais , Linhagem Celular Tumoral , Transtornos Cognitivos/tratamento farmacológico , Transtornos Cognitivos/etiologia , Citocinas/metabolismo , Modelos Animais de Doenças , Humanos , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Conformação Molecular/efeitos dos fármacos , Mutação/genética , Neuroblastoma/patologia , Oligopeptídeos/química , Fragmentos de Peptídeos/ultraestrutura , Peptídeos Cíclicos/metabolismo , Placa Amiloide/metabolismo , Placa Amiloide/patologia , Placa Amiloide/ultraestrutura , Ligação Proteica/efeitos dos fármacos , Estereoisomerismo
20.
ACS Chem Neurosci ; 7(2): 218-26, 2016 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-26629788

RESUMO

Alzheimer's disease (AD) is a neurodegenerative disease characterized by extracellular deposits of amyloid ß protein (Aß) in the brain. The conversion of soluble monomers to amyloid Aß fibrils is a complicated process and involves several transient oligomeric species, which are widely believed to be highly toxic and play a crucial role in the etiology of AD. The development of inhibitors to prevent formation of small and midsized oligomers is a promising strategy for AD treatment. In this work, we employ ion mobility spectrometry (IMS), transmission electron microscopy (TEM), and molecular dynamics (MD) simulations to elucidate the structural modulation promoted by two potential inhibitors of Aß oligomerization, cucurbit[7]uril (CB[7]) and 1,2,3,4,6-penta-O-galloyl-ß-d-glucopyranose (PGG), on early oligomer and fibril formation of the Aß25-35 fragment. One and two CB[7] molecules bind to Aß25-35 monomers and dimers, respectively, and suppress aggregation by remodeling early oligomer structures and inhibiting the formation of higher-order oligomers. On the other hand, nonselective binding was observed between PGG and Aß25-35. The interactions between PGG and Aß25-35, surprisingly, enhanced the formation of Aß aggregates by promoting extended Aß25-35 conformations in both homo- and hetero-oligomers. When both ligands were present, the inhibitory effect of CB[7] overrode the stimulatory effect of PGG on Aß25-35 aggregation, suppressing the formation of large amyloid oligomers and eliminating the structural conversion from isotropic to ß-rich topologies induced by PGG. Our results provide mechanistic insights into CB[7] and PGG action on Aß oligomerization. They also demonstrate the power of the IMS technique to investigate mechanisms of multiple small-molecule agents on the amyloid formation process.


Assuntos
Peptídeos beta-Amiloides/síntese química , Peptídeos beta-Amiloides/metabolismo , Hidrocarbonetos Aromáticos com Pontes/farmacologia , Taninos Hidrolisáveis/farmacologia , Imidazóis/farmacologia , Fragmentos de Peptídeos/síntese química , Fragmentos de Peptídeos/metabolismo , Peptídeos beta-Amiloides/química , Peptídeos beta-Amiloides/ultraestrutura , Simulação por Computador , Humanos , Espectrometria de Massas , Microscopia Eletrônica de Transmissão , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/ultraestrutura , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA