Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Biol Chem ; 298(10): 102430, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-36037966

RESUMO

Methionine/valine polymorphism at position 129 of the human prion protein, huPrP, is tightly associated with the pathogenic phenotype, disease progress, and age of onset of neurodegenerative diseases such as Creutzfeldt-Jakob disease or Fatal Familial Insomnia. This raises the question of whether and how the amino acid type at position 129 influences the structural properties of huPrP, affecting its folding, stability, and amyloid formation behavior. Here, our detailed biophysical characterization of the 129M and 129V variants of recombinant full-length huPrP(23-230) by amyloid formation kinetics, CD spectroscopy, molecular dynamics simulations, and sedimentation velocity analysis reveals differences in their aggregation propensity and oligomer content, leading to deviating pathways for the conversion into amyloid at acidic pH. We determined that the 129M variant exhibits less secondary structure content before amyloid formation and higher resistance to thermal denaturation compared to the 129V variant, whereas the amyloid conformation of both variants shows similar thermal stability. Additionally, our molecular dynamics simulations and rigidity analyses at the atomistic level identify intramolecular interactions responsible for the enhanced monomer stability of the 129M variant, involving more frequent minimum distances between E196 and R156, forming a salt bridge. Removal of the N-terminal half of the 129M full-length variant diminishes its differences compared to the 129V full-length variant and highlights the relevance of the flexible N terminus in huPrP. Taken together, our findings provide insight into structural properties of huPrP and the effects of the amino acid identity at position 129 on amyloid formation behavior.


Assuntos
Amiloide , Amiloidose , Síndrome de Creutzfeldt-Jakob , Insônia Familiar Fatal , Polimorfismo Genético , Proteínas Priônicas , Humanos , Amiloide/genética , Amiloide/química , Amiloidose/genética , Síndrome de Creutzfeldt-Jakob/genética , Metionina/genética , Proteínas Priônicas/química , Proteínas Priônicas/genética , Dobramento de Proteína , Valina/genética , Insônia Familiar Fatal/genética
2.
Chemistry ; 28(38): e202201698, 2022 Jul 06.
Artigo em Inglês | MEDLINE | ID: mdl-35701098

RESUMO

Invited for the cover of this issue is the group of Prof. Hamilton at New York University. The image depicts how cucurbit[7]uril inhibits islet amyloid polypeptide self-assembly that rescues rat insulinoma cells (a pancreatic ß-cell model) from assembly-associated cytotoxicity. Read the full text of the article at 10.1002/chem.202200456.


Assuntos
Células Secretoras de Insulina , Polipeptídeo Amiloide das Ilhotas Pancreáticas , Amiloide , Animais , Hidrocarbonetos Aromáticos com Pontes/farmacologia , Compostos Heterocíclicos com 2 Anéis , Humanos , Imidazóis/farmacologia , Imidazolidinas , Compostos Macrocíclicos , Ratos
3.
Nat Commun ; 13(1): 2363, 2022 05 02.
Artigo em Inglês | MEDLINE | ID: mdl-35501361

RESUMO

Chaperones, as modulators of protein conformational states, are key cellular actors to prevent the accumulation of fibrillar aggregates. Here, we integrated kinetic investigations with structural studies to elucidate how the ubiquitous co-chaperonin prefoldin inhibits diabetes associated islet amyloid polypeptide (IAPP) fibril formation. We demonstrated that both human and archaeal prefoldin interfere similarly with the IAPP fibril elongation and secondary nucleation pathways. Using archaeal prefoldin model, we combined nuclear magnetic resonance spectroscopy with electron microscopy to establish that the inhibition of fibril formation is mediated by the binding of prefoldin's coiled-coil helices to the flexible IAPP N-terminal segment accessible on the fibril surface and fibril ends. Atomic force microscopy demonstrates that binding of prefoldin to IAPP leads to the formation of lower amounts of aggregates, composed of shorter fibrils, clustered together. Linking structural models with observed fibrillation inhibition processes opens perspectives for understanding the interference between natural chaperones and formation of disease-associated amyloids.


Assuntos
Polipeptídeo Amiloide das Ilhotas Pancreáticas , Chaperonas Moleculares , Amiloide/metabolismo , Chaperoninas , Humanos , Chaperonas Moleculares/metabolismo
4.
Chemistry ; 28(38): e202200456, 2022 Jul 06.
Artigo em Inglês | MEDLINE | ID: mdl-35532096

RESUMO

Two "hot segments" within an islet amyloid polypeptide are responsible for its self-assembly, which in turn is linked to the decline of ß-cells in type 2 diabetes (T2D). A readily available water-soluble, macrocyclic host, cucurbit[7]uril (CB[7]), effectively inhibits islet amyloid polypeptide (IAPP) aggregation through ion-dipole and hydrophobic interactions with different residues of the monomeric peptide in its random-coil conformation. A HSQC NMR study shows that CB[7] likely modulates IAPP self-assembly by interacting with and masking major residues present in the "hot segments" at the N terminus. CB[7] also prevents the formation of toxic oligomers and inhibits seed-catalyzed fibril proliferation. Importantly, CB[7] recovers rat insulinoma cells (RIN-m) from IAPP-assembly associated cytotoxicity.


Assuntos
Diabetes Mellitus Tipo 2 , Células Secretoras de Insulina , Amiloide/química , Animais , Compostos Heterocíclicos com 2 Anéis , Imidazolidinas , Polipeptídeo Amiloide das Ilhotas Pancreáticas/química , Compostos Macrocíclicos , Ratos
5.
Nat Struct Mol Biol ; 27(7): 660-667, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32541895

RESUMO

Amyloid deposits consisting of fibrillar islet amyloid polypeptide (IAPP) in pancreatic islets are associated with beta-cell loss and have been implicated in type 2 diabetes (T2D). Here, we applied cryo-EM to reconstruct densities of three dominant IAPP fibril polymorphs, formed in vitro from synthetic human IAPP. An atomic model of the main polymorph, built from a density map of 4.2-Å resolution, reveals two S-shaped, intertwined protofilaments. The segment 21-NNFGAIL-27, essential for IAPP amyloidogenicity, forms the protofilament interface together with Tyr37 and the amidated C terminus. The S-fold resembles polymorphs of Alzheimer's disease (AD)-associated amyloid-ß (Aß) fibrils, which might account for the epidemiological link between T2D and AD and reports on IAPP-Aß cross-seeding in vivo. The results structurally link the early-onset T2D IAPP genetic polymorphism (encoding Ser20Gly) with the AD Arctic mutation (Glu22Gly) of Aß and support the design of inhibitors and imaging probes for IAPP fibrils.


Assuntos
Polipeptídeo Amiloide das Ilhotas Pancreáticas/química , Doença de Alzheimer/fisiopatologia , Substituição de Aminoácidos , Peptídeos beta-Amiloides/química , Microscopia Crioeletrônica , Diabetes Mellitus Tipo 2 , Humanos , Concentração de Íons de Hidrogênio , Polipeptídeo Amiloide das Ilhotas Pancreáticas/genética , Polipeptídeo Amiloide das Ilhotas Pancreáticas/metabolismo , Modelos Moleculares , Simulação de Dinâmica Molecular , Conformação Proteica
6.
RSC Chem Biol ; 1(4): 225-232, 2020 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-34458762

RESUMO

Membrane-catalysed misfolding of islet amyloid polypeptide is associated with the death of ß-cells in type II diabetes (T2D). Most active compounds so far reported require high doses for inhibition of membrane bound IAPP fibrillation. Here, we describe a naphthalimide-appended oligopyridylamide-based α-helical mimetic, DM 1, for targeting membrane bound IAPP. DM 1 completely inhibits the aggregation of IAPP at doses of 0.2 equivalents. DM 1 is also effective at similarly low doses for inhibition of seed-catalyzed secondary nucleation. An NMR based study demonstrates that DM 1 modulates IAPP self-assembly by stabilizing and/or perturbing the N-terminus helix conformation. DM 1 at substoichiometric doses rescues rat insulinoma cells from IAPP-mediated cytotoxicity. Most importantly, 0.2 equivalents of DM 1 disaggregate preformed oligomers and fibrils and can reverse cytotoxicity by modulating toxic preformed oligomers and fibrils of IAPP into non-toxic conformations.

7.
J Biol Chem ; 294(18): 7388-7402, 2019 05 03.
Artigo em Inglês | MEDLINE | ID: mdl-30862675

RESUMO

The hepatitis C virus (HCV) nonstructural protein 5A (NS5A) plays a key role in viral replication and virion assembly, and the regulation of the assembly process critically depends on phosphorylation of both serine and threonine residues in NS5A. We previously identified SRC proto-oncogene, nonreceptor tyrosine kinase (c-Src), as an essential host component of the HCV replication complex consisting of NS5A, the RNA-dependent RNA polymerase NS5B, and c-Src. Pulldown assays revealed an interaction between NS5A and the Src homology 2 (SH2) domain of c-Src; however, the precise binding mode remains undefined. In this study, using a variety of biochemical and biophysical techniques, along with molecular dynamics simulations, we demonstrate that the interaction between NS5A and the c-Src SH2 domain strictly depends on an intact phosphotyrosine-binding competent SH2 domain and on tyrosine phosphorylation within NS5A. Detailed analysis of c-Src SH2 domain binding to a panel of phosphorylation-deficient NS5A variants revealed that phosphorylation of Tyr-93 located within domain 1 of NS5A, but not of any other tyrosine residue, is crucial for complex formation. In line with these findings, effective replication of subgenomic HCV replicons as well as production of infectious virus particles in mammalian cell culture models were clearly dependent on the presence of tyrosine at position 93 of NS5A. These findings indicate that phosphorylated Tyr-93 in NS5A plays an important role during viral replication by facilitating NS5A's interaction with the SH2 domain of c-Src.


Assuntos
Hepacivirus/fisiologia , Tirosina/metabolismo , Proteínas não Estruturais Virais/metabolismo , Replicação Viral , Quinases da Família src/metabolismo , Linhagem Celular Tumoral , Humanos , Fosforilação , Ligação Proteica , Proto-Oncogene Mas , Proteínas não Estruturais Virais/química , Domínios de Homologia de src
8.
J Biol Chem ; 293(41): 15748-15764, 2018 10 12.
Artigo em Inglês | MEDLINE | ID: mdl-30131337

RESUMO

Alzheimer's disease (AD) is a progressive neurodegenerative disorder that affects millions of people worldwide. One AD hallmark is the aggregation of ß-amyloid (Aß) into soluble oligomers and insoluble fibrils. Several studies have reported that oligomers rather than fibrils are the most toxic species in AD progression. Aß oligomers bind with high affinity to membrane-associated prion protein (PrP), leading to toxic signaling across the cell membrane, which makes the Aß-PrP interaction an attractive therapeutic target. Here, probing this interaction in more detail, we found that both full-length, soluble human (hu) PrP(23-230) and huPrP(23-144), lacking the globular C-terminal domain, bind to Aß oligomers to form large complexes above the megadalton size range. Following purification by sucrose density-gradient ultracentrifugation, the Aß and huPrP contents in these heteroassemblies were quantified by reversed-phase HPLC. The Aß:PrP molar ratio in these assemblies exhibited some limited variation depending on the molar ratio of the initial mixture. Specifically, a molar ratio of about four Aß to one huPrP in the presence of an excess of huPrP(23-230) or huPrP(23-144) suggested that four Aß units are required to form one huPrP-binding site. Of note, an Aß-binding all-d-enantiomeric peptide, RD2D3, competed with huPrP for Aß oligomers and interfered with Aß-PrP heteroassembly in a concentration-dependent manner. Our results highlight the importance of multivalent epitopes on Aß oligomers for Aß-PrP interactions and have yielded an all-d-peptide-based, therapeutically promising agent that competes with PrP for these interactions.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Fragmentos de Peptídeos/metabolismo , Peptídeos/metabolismo , Proteínas Priônicas/metabolismo , Peptídeos beta-Amiloides/química , Sítios de Ligação , Humanos , Tamanho da Partícula , Fragmentos de Peptídeos/química , Peptídeos/química , Proteínas Priônicas/química , Ligação Proteica , Multimerização Proteica , Estereoisomerismo
9.
Cell Signal ; 50: 121-130, 2018 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-29964150

RESUMO

A pathological hallmark of Alzheimer's disease (AD) is the aggregation of amyloid-ß peptides (Aß) into fibrils, leading to deposits in cerebral parenchyma and vessels known as cerebral amyloid angiopathy (CAA). Platelets are major players of hemostasis but are also implicated in AD. Recently we provided strong evidence for a direct contribution of platelets to AD pathology. We found that monomeric Aß40 binds through its RHDS sequence to integrin αIIbß3, and promotes the formation of fibrillar Aß aggregates by the secretion of adenosine diphosphate (ADP) and the chaperone protein clusterin (CLU) from platelets. Here we investigated the molecular mechanisms of Aß binding to integrin αIIbß3 by using Aß11 and Aß16 peptides. These peptides include the RHDS binding motif important for integrin binding but lack the central hydrophobic core and the C-terminal sequence of Aß. We observed platelet adhesion to truncated N-terminal Aß11 and Aß16 peptides that was not mediated by integrin αIIbß3. Thus, no integrin outside-in signaling and reduced CLU release was detected. Accordingly, platelet mediated Aß fibril formation was not observed. Taken together, the RHDS motif of Aß is not sufficient for Aß binding to platelet integrin αIIbß3 and platelet mediated Aß fibril formation but requires other recognition or binding motifs important for platelet mediated processes in CAA. Thus, increased understanding of the molecular mechanisms of Aß binding to platelet integrin αIIbß3 is important to understand the role of platelets in amyloid pathology.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Amiloide/metabolismo , Plaquetas/metabolismo , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , Transdução de Sinais/fisiologia , Difosfato de Adenosina/metabolismo , Doença de Alzheimer/metabolismo , Angiopatia Amiloide Cerebral/metabolismo , Clusterina/metabolismo , Humanos
10.
Sci Rep ; 7(1): 16275, 2017 11 24.
Artigo em Inglês | MEDLINE | ID: mdl-29176708

RESUMO

While amyloid-ß protein (Aß) aggregation into insoluble plaques is one of the pathological hallmarks of Alzheimer's disease (AD), soluble oligomeric Aß has been hypothesized to be responsible for synapse damage, neurodegeneration, learning, and memory deficits in AD. Here, we investigate the in vitro and in vivo efficacy of the D-enantiomeric peptide RD2, a rationally designed derivative of the previously described lead compound D3, which has been developed to efficiently eliminate toxic Aß42 oligomers as a promising treatment strategy for AD. Besides the detailed in vitro characterization of RD2, we also report the results of a treatment study of APP/PS1 mice with RD2. After 28 days of treatment we observed enhancement of cognition and learning behaviour. Analysis on brain plaque load did not reveal significant changes, but a significant reduction of insoluble Aß42. Our findings demonstrate that RD2 was significantly more efficient in Aß oligomer elimination in vitro compared to D3. Enhanced cognition without reduction of plaque pathology in parallel suggests that synaptic malfunction due to Aß oligomers rather than plaque pathology is decisive for disease development and progression. Thus, Aß oligomer elimination by RD2 treatment may be also beneficial for AD patients.


Assuntos
Placa Amiloide/patologia , Peptídeos beta-Amiloides/química , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Cognição/efeitos dos fármacos , Modelos Animais de Doenças , Feminino , Camundongos , Peptídeos/química , Peptídeos/uso terapêutico , Placa Amiloide/tratamento farmacológico
11.
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
12.
Sci Rep ; 6: 33474, 2016 09 19.
Artigo em Inglês | MEDLINE | ID: mdl-27641459

RESUMO

In type 2 diabetes, the formation of islet amyloid consisting of islet amyloid polypeptide (IAPP) is associated with reduction in ß-cell mass and contributes to the failure of islet cell transplantation. Rational design of inhibitors of IAPP amyloid formation has therapeutic potential, but is hampered by the lack of structural information on inhibitor complexes of the conformationally flexible, aggregation-prone IAPP. Here we characterize a ß-hairpin conformation of IAPP in complex with the engineered binding protein ß-wrapin HI18. The ß-strands correspond to two amyloidogenic motifs, 12-LANFLVH-18 and 22-NFGAILS-28, which are connected by a turn established around Ser-20. Besides backbone hydrogen bonding, the IAPP:HI18 interaction surface is dominated by non-polar contacts involving hydrophobic side chains of the IAPP ß-strands. Apart from monomers, HI18 binds oligomers and fibrils and inhibits IAPP aggregation and toxicity at low substoichiometric concentrations. The IAPP ß-hairpin can serve as a molecular recognition motif enabling control of IAPP aggregation.


Assuntos
Polipeptídeo Amiloide das Ilhotas Pancreáticas/química , Polipeptídeo Amiloide das Ilhotas Pancreáticas/metabolismo , Agregados Proteicos , Motivos de Aminoácidos , Sequência de Aminoácidos , Amiloide/química , Peptídeos beta-Amiloides/química , Humanos , Modelos Moleculares , Agregados Proteicos/efeitos dos fármacos , Ligação Proteica/efeitos dos fármacos , Estrutura Secundária de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/farmacologia , Serina/química , alfa-Sinucleína/química
13.
PLoS One ; 11(9): e0161243, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27607147

RESUMO

Amyloid deposits formed from islet amyloid polypeptide (IAPP) are a hallmark of type 2 diabetes mellitus and are known to be cytotoxic to pancreatic ß-cells. The molecular structure of the fibrillar form of IAPP is subject of intense research, and to date, different models exist. We present results of solid-state NMR experiments on fibrils of recombinantly expressed and uniformly 13C, 15N-labeled human IAPP in the non-amidated, free acid form. Complete sequential resonance assignments and resulting constraints on secondary structure are shown. A single set of chemical shifts is found for most residues, which is indicative of a high degree of homogeneity. The core region comprises three to four ß-sheets. We find that the central 23-FGAILS-28 segment, which is of critical importance for amyloid formation, is part of the core region and forms a ß-strand in our sample preparation. The eight N-terminal amino acid residues of IAPP, forming a ring-like structure due to a disulfide bridge between residues C2 and C7, appear to be well defined but with an increased degree of flexibility. This study supports the elucidation of the structural basis of IAPP amyloid formation and highlights the extent of amyloid fibril polymorphism.


Assuntos
Amiloide/química , Polipeptídeo Amiloide das Ilhotas Pancreáticas/química , Espectroscopia de Ressonância Magnética , Proteínas Recombinantes/química , Cromatografia Líquida de Alta Pressão , Cromatografia de Fase Reversa , Temperatura Baixa , Difusão , Humanos , Microscopia de Força Atômica , Estrutura Secundária de Proteína , Reprodutibilidade dos Testes , Relação Estrutura-Atividade
14.
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
15.
Sci Signal ; 9(429): ra52, 2016 05 24.
Artigo em Inglês | MEDLINE | ID: mdl-27221710

RESUMO

Cerebral amyloid angiopathy (CAA) is a vascular dysfunction disorder characterized by deposits of amyloid-ß (Aß) in the walls of cerebral vessels. CAA and Aß deposition in the brain parenchyma contribute to dementia and Alzheimer's disease (AD). We investigated the contribution of platelets, which accumulate at vascular Aß deposits, to CAA. We found that synthetic monomeric Aß40 bound through its RHDS (Arg-His-Asp-Ser) sequence to integrin αIIbß3, which is the receptor for the extracellular matrix protein fibrinogen, and stimulated the secretion of adenosine diphosphate (ADP) and the chaperone protein clusterin from platelets. Clusterin promoted the formation of fibrillar Aß aggregates, and ADP acted through its receptors P2Y1 and P2Y12 on platelets to enhance integrin αIIbß3 activation, further increasing the secretion of clusterin and Aß40 binding to platelets. Platelets from patients with Glanzmann's thrombasthenia, a bleeding disorder in which platelets have little or dysfunctional αIIbß3, indicated that the abundance of this integrin dictated Aß-induced clusterin release and platelet-induced Aß aggregation. The antiplatelet agent clopidogrel, which irreversibly inhibits P2Y12, inhibited Aß aggregation in platelet cultures; in transgenic AD model mice, this drug reduced the amount of clusterin in the circulation and the incidence of CAA. Our findings indicate that activated platelets directly contribute to CAA by promoting the formation of Aß aggregates and that Aß, in turn, activates platelets, creating a feed-forward loop. Thus, antiplatelet therapy may alleviate fibril formation in cerebral vessels of AD patients.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Plaquetas/citologia , Clusterina/metabolismo , Integrinas/metabolismo , Adolescente , Adulto , Doença de Alzheimer/metabolismo , Animais , Encéfalo/metabolismo , Circulação Cerebrovascular , Cruzamentos Genéticos , Progressão da Doença , Feminino , Fibrinogênio/metabolismo , Genótipo , Humanos , Integrina alfa2/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Pessoa de Meia-Idade , Simulação de Dinâmica Molecular , Fosforilação , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , Transdução de Sinais , Adulto Jovem
16.
PLoS One ; 11(4): e0153035, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27105346

RESUMO

The aggregation of amyloid-ß (Aß) is postulated to be the crucial event in Alzheimer's disease (AD). In particular, small neurotoxic Aß oligomers are considered to be responsible for the development and progression of AD. Therefore, elimination of thesis oligomers represents a potential causal therapy of AD. Starting from the well-characterized d-enantiomeric peptide D3, we identified D3 derivatives that bind monomeric Aß. The underlying hypothesis is that ligands bind monomeric Aß and stabilize these species within the various equilibria with Aß assemblies, leading ultimately to the elimination of Aß oligomers. One of the hereby identified d-peptides, DB3, and a head-to-tail tandem of DB3, DB3DB3, were studied in detail. Both peptides were found to: (i) inhibit the formation of Thioflavin T-positive fibrils; (ii) bind to Aß monomers with micromolar affinities; (iii) eliminate Aß oligomers; (iv) reduce Aß-induced cytotoxicity; and (v) disassemble preformed Aß aggregates. The beneficial effects of DB3 were improved by DB3DB3, which showed highly enhanced efficacy. Our approach yielded Aß monomer-stabilizing ligands that can be investigated as a suitable therapeutic strategy against AD.


Assuntos
Peptídeos beta-Amiloides/química , Oligopeptídeos/química , Sequência de Aminoácidos , Animais , Linhagem Celular Tumoral , Ensaio de Imunoadsorção Enzimática , Ratos
17.
PLoS One ; 11(2): e0147470, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26840229

RESUMO

Alzheimer´s disease is the most prominent type of dementia and currently no causative treatment is available. According to recent studies, oligomeric species of the amyloid beta (Aß) peptide appear to be the most toxic Aß assemblies. Aß monomers, however, may be not toxic per se and may even have a neuroprotective role. Here we describe a competitive mirror image phage display procedure that allowed us to identify preferentially Aß1-42 monomer binding and thereby stabilizing peptides, which destabilize and thereby eliminate toxic oligomer species. One of the peptides, called Mosd1 (monomer specific d-peptide 1), was characterized in more detail. Mosd1 abolished oligomers from a mixture of Aß1-42 species, reduced Aß1-42 toxicity in cell culture, and restored the physiological phenotype in neuronal cells stably transfected with the gene coding for human amyloid precursor protein.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Peptídeos beta-Amiloides/toxicidade , Técnicas de Visualização da Superfície Celular , Peptídeos/farmacologia , Agregados Proteicos/efeitos dos fármacos , Agregação Patológica de Proteínas , Secretases da Proteína Precursora do Amiloide/metabolismo , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Humanos
18.
Protein Eng Des Sel ; 27(12): 473-9, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25332193

RESUMO

Aggregation of the protein α-synuclein (α-syn) has been implicated in Parkinson's disease and other neurodegenerative disorders, collectively referred to as synucleinopathies. The ß-wrapin AS69 is a small engineered binding protein to α-syn that stabilizes a ß-hairpin conformation of monomeric α-syn and inhibits α-syn aggregation at substoichiometric concentrations. AS69 is a homodimer whose subunits are linked via a disulfide bridge between their single cysteine residues, Cys-28. Here we show that expression of a functional dimer as a single polypeptide chain is achievable by head-to-tail linkage of AS69 subunits. Choice of a suitable linker is essential for construction of head-to-tail dimers that exhibit undiminished α-syn affinity compared with the solely disulfide-linked dimer. We characterize AS69-GS3, a head-to-tail dimer with a glycine-serine-rich linker, under oxidized and reduced conditions in order to evaluate the impact of the Cys28-disulfide bond on structure, stability and α-syn binding. Formation of the disulfide bond causes compaction of AS69-GS3, increases its thermostability, and is a prerequisite for high-affinity binding to α-syn. Comparison of AS69-GS3 and AS69 demonstrates that head-to-tail linkage promotes α-syn binding by affording accelerated disulfide bond formation.


Assuntos
Dissulfetos/química , Peptídeos/química , alfa-Sinucleína/química , Sequência de Aminoácidos , Escherichia coli/genética , Escherichia coli/metabolismo , Expressão Gênica , Humanos , Cinética , Dados de Sequência Molecular , Peptídeos/genética , Peptídeos/metabolismo , Ligação Proteica , Engenharia de Proteínas , Dobramento de Proteína , Multimerização Proteica , Estrutura Secundária de Proteína , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Relação Estrutura-Atividade , Termodinâmica
19.
J Biotechnol ; 191: 221-7, 2014 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-24928165

RESUMO

Human islet amyloid polypeptide (IAPP) is the major component of pancreatic amyloid deposits in type 2 diabetes. The structural conversion of IAPP from a monomeric state into amyloid assemblies is the subject of intense research. Recombinant production of IAPP is, however, difficult due to its extreme aggregation propensity. Here we describe a novel strategy for expression of IAPP in Escherichia coli, based on an engineered protein tag, which sequesters IAPP monomers and prevents IAPP aggregation. The IAPP-binding protein HI18 was selected by phage display from a ß-wrapin library. Fusion of HI18 to IAPP enabled the soluble expression of the construct. IAPP was cleaved from the fusion construct and purified to homogeneity with a yield of 3mg of isotopically labeled peptide per liter of culture. In the monomeric state, IAPP was largely disordered as evidenced by far-UV CD and liquid-state NMR spectroscopy but competent to form amyloid fibrils according to atomic force microscopy. These results demonstrate the ability of the engineered ß-wrapin HI18 for shielding the hydrophobic sequence of IAPP during expression and purification. Fusion of aggregation-inhibiting ß-wrapins is a suitable approach for the recombinant production of aggregation-prone proteins.


Assuntos
Sequência de Aminoácidos/genética , Diabetes Mellitus Tipo 2/genética , Polipeptídeo Amiloide das Ilhotas Pancreáticas/biossíntese , Agregação Patológica de Proteínas/genética , Escherichia coli/genética , Regulação da Expressão Gênica , Humanos , Polipeptídeo Amiloide das Ilhotas Pancreáticas/genética , Espectroscopia de Ressonância Magnética , Microscopia de Força Atômica , Dobramento de Proteína , Estrutura Secundária de Proteína
20.
J Biol Chem ; 289(1): 74-88, 2014 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-24273164

RESUMO

The cancer-associated, centrosomal adaptor protein TACC3 (transforming acidic coiled-coil 3) and its direct effector, the microtubule polymerase chTOG (colonic and hepatic tumor overexpressed gene), play a crucial function in centrosome-driven mitotic spindle assembly. It is unclear how TACC3 interacts with chTOG. Here, we show that the C-terminal TACC domain of TACC3 and a C-terminal fragment adjacent to the TOG domains of chTOG mediate the interaction between these two proteins. Interestingly, the TACC domain consists of two functionally distinct subdomains, CC1 (amino acids (aa) 414-530) and CC2 (aa 530-630). Whereas CC1 is responsible for the interaction with chTOG, CC2 performs an intradomain interaction with the central repeat region of TACC3, thereby masking the TACC domain before effector binding. Contrary to previous findings, our data clearly demonstrate that Aurora-A kinase does not regulate TACC3-chTOG complex formation, indicating that Aurora-A solely functions as a recruitment factor for the TACC3-chTOG complex to centrosomes and proximal mitotic spindles. We identified with CC1 and CC2, two functionally diverse modules within the TACC domain of TACC3 that modulate and mediate, respectively, TACC3 interaction with chTOG required for spindle assembly and microtubule dynamics during mitotic cell division.


Assuntos
Aurora Quinase A/metabolismo , Proteínas de Transporte/metabolismo , Centrossomo/metabolismo , Proteínas Fetais/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Mitose/fisiologia , Animais , Aurora Quinase A/genética , Proteínas de Transporte/genética , Proteínas Fetais/genética , Células HEK293 , Células HeLa , Humanos , Camundongos , Proteínas Associadas aos Microtúbulos/genética , Complexos Multiproteicos/genética , Complexos Multiproteicos/metabolismo , Ligação Proteica/fisiologia , Estrutura Terciária de Proteína
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA