Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
1.
Biomacromolecules ; 22(7): 2822-2833, 2021 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-34196531

RESUMEN

Protein amyloid nanofibers provide a biocompatible platform for the development of functional nanomaterials. However, the functionalities generated up to date are still limited. Typical building blocks correspond to aggregation-prone proteins and peptides, which must be modified by complex and expensive reactions post-assembly. There is high interest in researching alternative strategies to tailor amyloid-based nanostructures' functionality on demand. In the present study, the biotin-streptavidin system was exploited for this purpose. Prion-inspired heptapeptides (Ac-NYNYNYN-NH2, Ac-QYQYQYQ-NH2, and Ac-SYSYSYS-NH2) were doped with biotin-conjugated counterparts and assembled into amyloid-like fibers under mild conditions. The scaffolds' versatile functionalization was demonstrated by decorating them with different streptavidin conjugates, including gold nanoparticles, quantum dots, and enzymes. In particular, they were functionalized with peroxidase or phosphatase activities using streptavidin conjugated with horseradish peroxidase and alkaline phosphatase, respectively. Modification of amyloid-like nanostructures has generally been restricted to the addition of a single protein moiety. We functionalized the fibrils simultaneously with glucose oxidase and horseradish peroxidase, coupling these activities to build up a nanostructured glucose biosensor. Overall, we present a simple, modular, and multivalent approach for developing amyloid-based nanomaterials functionalized with any desired combination of chemical and biological moieties.


Asunto(s)
Técnicas Biosensibles , Nanopartículas del Metal , Priones , Amiloide , Oro
2.
Bioorg Chem ; 117: 105472, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34775206

RESUMEN

The treatment of Parkinson's disease (PD), the second most common neurodegenerative human disorder, continues to be symptomatic. Development of drugs able to stop or at least slowdown PD progression would benefit several million people worldwide. SynuClean-D is a low molecular weight 2-pyridone-based promising drug candidate that inhibits the aggregation of α-synuclein in human cultured cells and prevents degeneration of dopaminergic neurons in a Caenorhabditis elegans model of PD. Improving SynuClean-D pharmacokinetic/pharmacodynamic properties, performing structure/activity studies and testing its efficacy in mammalian models of PD requires the use of gr-amounts of the compound. However, not enough compound is on sale, and no synthetic route has been reported until now, which hampers the molecule progress towards clinical trials. To circumvent those problems, we describe here an efficient and economical route that enables the synthesis of SynuClean-D with good yields as well as the synthesis of SynuClean-D derivatives. Structure-activity comparison of the new compounds with SynuClean-D reveals the functional groups of the molecule that can be disposed of without activity loss and those that are crucial to interfere with α-synuclein aggregation. Several of the derivatives obtained retain the parent's compound excellent in vitro anti-aggregative activity, without compromising its low toxicity. Computational predictions and preliminary testing indicate that the blood brain barrier (BBB) permeability of SynuClean-D is low. Importantly, several of the newly designed and obtained active derivatives are predicted to display good BBB permeability. The synthetic route developed here will facilitate their synthesis for BBB permeability determination and for efficacy testing in mammalian models of PD.


Asunto(s)
Barrera Hematoencefálica/efectos de los fármacos , Diseño de Fármacos , Enfermedad de Parkinson/tratamiento farmacológico , Piridonas/farmacología , alfa-Sinucleína/antagonistas & inhibidores , Animales , Barrera Hematoencefálica/metabolismo , Caenorhabditis elegans , Relación Dosis-Respuesta a Droga , Estructura Molecular , Enfermedad de Parkinson/metabolismo , Agregado de Proteínas/efectos de los fármacos , Piridonas/síntesis química , Piridonas/química , Relación Estructura-Actividad , alfa-Sinucleína/metabolismo
3.
Proc Natl Acad Sci U S A ; 115(41): 10481-10486, 2018 10 09.
Artículo en Inglés | MEDLINE | ID: mdl-30249646

RESUMEN

Parkinson's disease (PD) is characterized by a progressive loss of dopaminergic neurons, a process that current therapeutic approaches cannot prevent. In PD, the typical pathological hallmark is the accumulation of intracellular protein inclusions, known as Lewy bodies and Lewy neurites, which are mainly composed of α-synuclein. Here, we exploited a high-throughput screening methodology to identify a small molecule (SynuClean-D) able to inhibit α-synuclein aggregation. SynuClean-D significantly reduces the in vitro aggregation of wild-type α-synuclein and the familiar A30P and H50Q variants in a substoichiometric molar ratio. This compound prevents fibril propagation in protein-misfolding cyclic amplification assays and decreases the number of α-synuclein inclusions in human neuroglioma cells. Computational analysis suggests that SynuClean-D can bind to cavities in mature α-synuclein fibrils and, indeed, it displays a strong fibril disaggregation activity. The treatment with SynuClean-D of two PD Caenorhabditis elegans models, expressing α-synuclein either in muscle or in dopaminergic neurons, significantly reduces the toxicity exerted by α-synuclein. SynuClean-D-treated worms show decreased α-synuclein aggregation in muscle and a concomitant motility recovery. More importantly, this compound is able to rescue dopaminergic neurons from α-synuclein-induced degeneration. Overall, SynuClean-D appears to be a promising molecule for therapeutic intervention in Parkinson's disease.


Asunto(s)
Amiloide/efectos de los fármacos , Caenorhabditis elegans/efectos de los fármacos , Neuronas Dopaminérgicas/efectos de los fármacos , Enfermedad de Parkinson/tratamiento farmacológico , Agregación Patológica de Proteínas/tratamiento farmacológico , Bibliotecas de Moléculas Pequeñas/farmacología , alfa-Sinucleína/antagonistas & inhibidores , Amiloide/metabolismo , Animales , Caenorhabditis elegans/metabolismo , Neuronas Dopaminérgicas/metabolismo , Neuronas Dopaminérgicas/patología , Ensayos Analíticos de Alto Rendimiento , Humanos , Neuroblastoma/tratamiento farmacológico , Neuroblastoma/metabolismo , Neuroblastoma/patología , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología , Agregación Patológica de Proteínas/metabolismo , Agregación Patológica de Proteínas/patología , Células Tumorales Cultivadas , alfa-Sinucleína/metabolismo
4.
Biomacromolecules ; 21(6): 2334-2345, 2020 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-32227922

RESUMEN

Amyloids are associated with human disease. However, they are also exploited by nature for functional purposes. Functional amyloids have inspired amyloid-based biomaterials for different nanotechnologies. Early soluble species in the fibrillation pathway seem to be the primary elicitors of cytotoxicity, instead of fibrils. Organisms have evolved dedicated mechanisms to avoid toxicity during the assembly of functional amyloids. In their absence, artificial amyloid-based nanomaterials might also produce toxic intermediates. We show here that even when the building blocks of artificial amyloids are small, polar, and compositionally simple, their early soluble assemblies are extremely cytotoxic, causing cell death through mechanisms identical to those of disease-associated proteins. Our results raise safety concerns about the use of non-natural amyloid-based materials without a rigorous characterization of their fibrillation pathway. Besides, the simple, cheap, and easy to synthesize peptides we use here might turn very useful to understand the molecular determinants behind amyloid cytotoxicity.


Asunto(s)
Amiloidosis , Priones , Amiloide , Proteínas Amiloidogénicas , Humanos , Péptidos
5.
Microb Cell Fact ; 19(1): 117, 2020 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-32487230

RESUMEN

BACKGROUND: Recombinant protein expression in bacteria often leads to the formation of intracellular insoluble protein deposits, a major bottleneck for the production of soluble and active products. However, in recent years, these bacterial protein aggregates, commonly known as inclusion bodies (IBs), have been shown to be a source of stable and active protein for biotechnological and biomedical applications. The formation of these functional IBs is usually facilitated by the fusion of aggregation-prone peptides or proteins to the protein of interest, leading to the formation of amyloid-like nanostructures, where the functional protein is embedded. RESULTS: In order to offer an alternative to the classical amyloid-like IBs, here we develop functional IBs exploiting the coiled-coil fold. An in silico analysis of coiled-coil and aggregation propensities, net charge, and hydropathicity of different potential tags identified the natural homo-dimeric and anti-parallel coiled-coil ZapB bacterial protein as an optimal candidate to form assemblies in which the native state of the fused protein is preserved. The protein itself forms supramolecular fibrillar networks exhibiting only α-helix secondary structure. This non-amyloid self-assembly propensity allows generating innocuous IBs in which the recombinant protein of interest remains folded and functional, as demonstrated using two different fluorescent proteins. CONCLUSIONS: Here, we present a proof of concept for the use of a natural coiled-coil domain as a versatile tool for the production of functional IBs in bacteria. This α-helix-based strategy excludes any potential toxicity drawback that might arise from the amyloid nature of ß-sheet-based IBs and renders highly active and homogeneous submicrometric particles.


Asunto(s)
Proteínas de Ciclo Celular , Proteínas de Escherichia coli , Escherichia coli/metabolismo , Cuerpos de Inclusión , Agregado de Proteínas , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/metabolismo , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , Células HeLa , Humanos , Estructura Secundaria de Proteína , Proteínas Recombinantes de Fusión/metabolismo
6.
PLoS Pathog ; 12(6): e1005711, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27327765

RESUMEN

Biofilms are communities of bacteria that grow encased in an extracellular matrix that often contains proteins. The spatial organization and the molecular interactions between matrix scaffold proteins remain in most cases largely unknown. Here, we report that Bap protein of Staphylococcus aureus self-assembles into functional amyloid aggregates to build the biofilm matrix in response to environmental conditions. Specifically, Bap is processed and fragments containing at least the N-terminus of the protein become aggregation-prone and self-assemble into amyloid-like structures under acidic pHs and low concentrations of calcium. The molten globule-like state of Bap fragments is stabilized upon binding of the cation, hindering its self-assembly into amyloid fibers. These findings define a dual function for Bap, first as a sensor and then as a scaffold protein to promote biofilm development under specific environmental conditions. Since the pH-driven multicellular behavior mediated by Bap occurs in coagulase-negative staphylococci and many other bacteria exploit Bap-like proteins to build a biofilm matrix, the mechanism of amyloid-like aggregation described here may be widespread among pathogenic bacteria.


Asunto(s)
Proteínas Amiloidogénicas/metabolismo , Proteínas Bacterianas/metabolismo , Biopelículas/crecimiento & desarrollo , Animales , Modelos Animales de Enfermedad , Immunoblotting , Ratones , Microscopía Fluorescente , Reacción en Cadena de la Polimerasa , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/metabolismo
7.
Mol Pharm ; 15(9): 3846-3859, 2018 09 04.
Artículo en Inglés | MEDLINE | ID: mdl-30036481

RESUMEN

The aggregation propensity of each particular protein seems to be shaped by evolution according to its natural abundance in the cell. The production and downstream processing of recombinant polypeptides implies attaining concentrations that are orders of magnitude above their natural levels, often resulting in their aggregation; a phenomenon that precludes the marketing of many globular proteins for biomedical or biotechnological applications. Therefore, there is a huge interest in methods aimed to increase the proteins solubility above their natural limits. Here, we demonstrate that an updated version of our AGGRESCAN 3D structural aggregation predictor, that now takes into account protein stability, allows for designing mutations at specific positions in the structure that improve the solubility of proteins without compromising their conformation. Using this approach, we have designed a highly soluble variant of the green fluorescent protein and a human single-domain VH antibody displaying significantly reduced aggregation propensity. Overall, our data indicate that the solubility of unrelated proteins can be easily tuned by in silico-designed nondestabilizing amino acid changes at their surfaces.


Asunto(s)
Proteínas/química , Dicroismo Circular , Cristalografía por Rayos X , Citometría de Flujo , Proteínas Fluorescentes Verdes/química , Immunoblotting , Microscopía Fluorescente , Conformación Proteica , Ingeniería de Proteínas , Estabilidad Proteica , Solubilidad
8.
Int J Mol Sci ; 19(8)2018 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-30042316

RESUMEN

The protein ataxin-3 contains a polyglutamine stretch that triggers amyloid aggregation when it is expanded beyond a critical threshold. This results in the onset of the spinocerebellar ataxia type 3. The protein consists of the globular N-terminal Josephin domain and a disordered C-terminal tail where the polyglutamine stretch is located. Expanded ataxin-3 aggregates via a two-stage mechanism: first, Josephin domain self-association, then polyQ fibrillation. This highlights the intrinsic amyloidogenic potential of Josephin domain. Therefore, much effort has been put into investigating its aggregation mechanism(s). A key issue regards the conformational requirements for triggering amyloid aggregation, as it is believed that, generally, misfolding should precede aggregation. Here, we have assayed the effect of 2,2,2-trifluoroethanol, a co-solvent capable of stabilizing secondary structures, especially α-helices. By combining biophysical methods and molecular dynamics, we demonstrated that both secondary and tertiary JD structures are virtually unchanged in the presence of up to 5% 2,2,2-trifluoroethanol. Despite the preservation of JD structure, 1% of 2,2,2-trifluoroethanol suffices to exacerbate the intrinsic aggregation propensity of this domain, by slightly decreasing its conformational stability. These results indicate that in the case of JD, conformational fluctuations might suffice to promote a transition towards an aggregated state without the need for extensive unfolding, and highlights the important role played by the environment on the aggregation of this globular domain.


Asunto(s)
Amiloide/efectos de los fármacos , Ataxina-3/metabolismo , Agregado de Proteínas/efectos de los fármacos , Proteínas Represoras/metabolismo , Trifluoroetanol/farmacología , Ataxina-3/química , Dicroismo Circular , Humanos , Conformación Molecular , Simulación de Dinámica Molecular , Péptidos/metabolismo , Conformación Proteica/efectos de los fármacos , Dominios Proteicos/efectos de los fármacos , Estabilidad Proteica/efectos de los fármacos , Estructura Secundaria de Proteína/efectos de los fármacos , Estructura Terciaria de Proteína/efectos de los fármacos , Proteínas Represoras/química
9.
Expert Rev Proteomics ; 14(4): 335-350, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28271922

RESUMEN

INTRODUCTION: The aberrant or misfolded forms of the prion protein have been described as the causative agents of rare transmissible spongiform encephalopathies. In addition, proteins associated with frequently occurring neurodegenerative disorders, such as Alzheimer's and Parkinson's, are shown to share prion-like properties and to spread the disease in the brain. Areas covered: Interest in the prion phenomenon has crystallized in a series of computational methods aimed at uncovering prion-like proteins at the proteome level. These programs rely on the identification of sequence signatures similar to those of yeast prions, whose structural conversion is driven by specific domains enriched in glutamine/asparagine residues. A myriad of prion-like candidates, similar to those in yeast, are predicted to exist in organisms across all kingdoms of life. We review here the role of prions, prionoids and prion-like proteins in health and disease, with a special focus on the algorithms and databases developed for their prediction and classification. Expert commentary: Computational approaches provide novel insights into prion-like protein functions, their regulation and their role in disease.


Asunto(s)
Enfermedades Neurodegenerativas/genética , Enfermedades por Prión/genética , Proteínas Priónicas/química , Proteoma/genética , Algoritmos , Biología Computacional , Humanos , Enfermedades Neurodegenerativas/patología , Enfermedades por Prión/patología , Proteínas Priónicas/genética , Conformación Proteica
10.
Int J Mol Sci ; 18(3)2017 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-28257086

RESUMEN

An increasing number of neurodegenerative diseases are being found to be associated with the abnormal accumulation of aggregated proteins in the brain. In Parkinson's disease, this process involves the aggregation of alpha-synuclein (α-syn) into intraneuronal inclusions. Thus, compounds that inhibit α-syn aggregation represent a promising therapeutic strategy as disease-modifying agents for neurodegeneration. The formation of α-syn amyloid aggregates can be reproduced in vitro by incubation of the recombinant protein. However, the in vitro aggregation of α-syn is exceedingly slow and highly irreproducible, therefore precluding fast high throughput anti-aggregation drug screening. Here, we present a simple and easy-to-implement in-plate method for screening large chemical libraries in the search for α-syn aggregation modulators. It allows us to monitor aggregation kinetics with high reproducibility, while being faster and requiring lower protein amounts than conventional aggregation assays. We illustrate how the approach enables the identification of strong aggregation inhibitors in a library of more than 14,000 compounds.


Asunto(s)
Ensayos Analíticos de Alto Rendimiento/métodos , Bibliotecas de Moléculas Pequeñas/farmacología , alfa-Sinucleína/antagonistas & inhibidores , Bases de Datos de Compuestos Químicos , Humanos , Cinética , Enfermedad de Parkinson/metabolismo , Proteínas Recombinantes/farmacología
11.
Int J Mol Sci ; 17(6)2016 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-27322259

RESUMEN

Human Amylin, or islet amyloid polypeptide (hIAPP), is a small hormone secreted by pancreatic ß-cells that forms aggregates under insulin deficiency metabolic conditions, and it constitutes a pathological hallmark of type II diabetes mellitus. In type II diabetes patients, amylin is abnormally increased, self-assembled into amyloid aggregates, and ultimately contributes to the apoptotic death of ß-cells by mechanisms that are not completely understood. We have screened a library of approved drugs in order to identify inhibitors of amylin aggregation that could be used as tools to investigate the role of amylin aggregation in type II diabetes or as therapeutics in order to reduce ß-cell damage. Interestingly, three of the compounds analyzed-benzbromarone, quercetin, and folic acid-are able to slow down amylin fiber formation according to Thioflavin T binding, turbidimetry, and Transmission Electron Microscopy assays. In addition to the in vitro assays, we have tested the effect of these compounds in an amyloid toxicity cell culture model and we have found that one of them, quercetin, has the ability to partly protect cultured pancreatic insulinoma cells from the cytotoxic effect of amylin. Our data suggests that quercetin can contribute to reduce oxidative damage in pancreatic insulinoma ß cells by modulating the aggregation propensity of amylin.


Asunto(s)
Amiloide/antagonistas & inhibidores , Benzbromarona/farmacología , Ácido Fólico/farmacología , Polipéptido Amiloide de los Islotes Pancreáticos/metabolismo , Agregación Patológica de Proteínas/metabolismo , Quercetina/farmacología , Animales , Células HeLa , Humanos , Ratas
12.
Biochim Biophys Acta ; 1843(5): 866-74, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24472658

RESUMEN

Despite genetic variation has the potential to arise new protein functions, spontaneous mutations usually destabilize the native fold. Misfolded proteins tend to form cytotoxic intracellular aggregates, decreasing cell fitness and leading to degenerative disorders in humans. Therefore, it is thought that selection against protein misfolding and aggregation constrains the evolution of protein sequences. However, obtaining experimental data to validate this hypothesis has been traditionally difficult. Here we exploit bacteria as a model organism to address this question. Using variants of the Alzheimer's related Aß42 peptide designed to exhibit different in vivo aggregation propensities we show here that, in cell competition experiments, the most aggregation-prone variants are always purged out from the growing population. Flow cytometry analysis of cellular metabolism and viability demonstrates that this purifying effect responds to a clear correlation between physiological burden and intrinsic aggregation propensity. Interestingly, the fitness cost of aggregation appears to be associated with aggregation rates rather than with overall protein solubility. Accordingly, we show that, by reducing in vivo aggregation rates, the model osmolyte proline is able to buffer the metabolic impact of protein aggregation. Overall, our data provide experimental support for the role of toxic protein aggregation on the cell fitness landscape and the evolution of natural protein sequences.


Asunto(s)
Proteínas Bacterianas/metabolismo , Secuencia de Aminoácidos , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Electroforesis en Gel de Poliacrilamida , Citometría de Flujo , Microscopía Fluorescente , Datos de Secuencia Molecular , Solubilidad
13.
Microb Cell Fact ; 14: 102, 2015 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-26160665

RESUMEN

BACKGROUND: The formation of protein inclusions is connected to the onset of many human diseases. Human RNA binding proteins containing intrinsically disordered regions with an amino acid composition resembling those of yeast prion domains, like TDP-43 or FUS, are being found to aggregate in different neurodegenerative disorders. The structure of the intracellular inclusions formed by these proteins is still unclear and whether these deposits have an amyloid nature or not is a matter of debate. Recently, the aggregation of TDP-43 has been modelled in bacteria, showing that TDP-43 inclusion bodies (IBs) are amorphous but intrinsically neurotoxic. This observation raises the question of whether it is indeed the lack of an ordered structure in these human prion-like protein aggregates the underlying cause of their toxicity in different pathological states. RESULTS: Here we characterize the IBs formed by the human prion-like RNA-processing protein HNRPDL. HNRPDL is linked to the development of limb-girdle muscular dystrophy 1G and shares domain architecture with TDP-43. We show that HNRPDL IBs display characteristic amyloid hallmarks, since these aggregates bind to amyloid dyes in vitro and inside the cell, they are enriched in intermolecular ß-sheet conformation and contain inner amyloid-like fibrillar structure. In addition, despite their ordered structure, HNRPDL IBs are highly neurotoxic. CONCLUSIONS: Our results suggest that at least some of the disorders caused by the aggregation of human prion-like proteins would rely on the formation of classical amyloid assemblies rather than being caused by amorphous aggregates. They also illustrate the power of microbial cell factories to model amyloid aggregation.


Asunto(s)
Amiloide/metabolismo , Bacterias/metabolismo , Cuerpos de Inclusión/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Priones/genética , ARN/metabolismo , Humanos
14.
Microb Cell Fact ; 14: 174, 2015 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-26536866

RESUMEN

BACKGROUND: An increasing number of proteins are being shown to assemble into amyloid structures that lead to pathological states. Among them, mammalian prions outstand due to their ability to transmit the pathogenic conformation, becoming thus infectious. The structural conversion of the cellular prion protein (PrP(C)), into its misfolded pathogenic form (PrP(Sc)) is the central event of prion-driven pathologies. The study of the structural properties of intracellular amyloid aggregates in general and of prion-like ones in particular is a challenging task. In this context, the evidence that the inclusion bodies formed by amyloid proteins in bacteria display amyloid-like structural and functional properties make them a privileged system to model intracellular amyloid aggregation. RESULTS: Here we provide the first demonstration that recombinant murine PrP and its C-terminal domain (90-231) attain amyloid conformations inside bacteria. Moreover, the inclusions formed by these two PrP proteins display conformational diversity, since they differ in fibril morphology, binding affinity to amyloid dyes, stability, resistance to proteinase K digestion and neurotoxicity. CONCLUSIONS: Overall, our results suggest that modelling PrP amyloid formation in microbial cell factories might open an avenue for a better understanding of the structural features modulating the pathogenic impact of this intriguing protein.


Asunto(s)
Amiloide/química , Bacterias/metabolismo , Priones/química , Amiloide/metabolismo , Animales , Benzotiazoles , Endopeptidasa K/metabolismo , Escherichia coli/metabolismo , Cuerpos de Inclusión/metabolismo , Ratones , Microscopía Electrónica de Transmisión , Priones/genética , Priones/metabolismo , Unión Proteica , Pliegue de Proteína , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/química , Proteínas Recombinantes/aislamiento & purificación , Espectroscopía Infrarroja por Transformada de Fourier , Tiazoles/química , Tiazoles/metabolismo
15.
Nanoscale Adv ; 6(10): 2669-2681, 2024 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-38752140

RESUMEN

Protein-based materials have emerged as promising candidates for proton-conducting biomaterials. Therefore, drawing inspiration from the amino acid composition of prion-like domains, we designed short self-assembling peptides incorporating the (X-Tyr) motif, with X representing Asn, Gly and Ser, which form fibrillar structures capable of conducting protons. In this study, we conducted an analysis of the conductivity capacity of these fibers, with a focus on temperature and frequency dependence of conductivity. The loss tangent curves data and the electrode polarization model with the Debye approximation were employed to calculate transport properties, including conductivity, diffusivity, and density of charge carriers. Results revealed the prion-like fibers can transport protons more efficiently than biomaterials and other synthetic proton conducting materials, and that a significant increase in conductivity is observed with fibrillar orientations. The temperature dependence of conductivity of the peptides, measured in wet conditions, showed conductivities following the trend σ(NY7) < σ(GY7) < σ(SY7), in all the range of temperatures studied. The Arrhenius behavior, and the activation energy associated with conductivity followed the trend: Eact (SY7) = 8.2 ± 0.6 kJ mol-1 < Eact (GY7) < 13 ± 5 kJ mol-1 < Eact (NY7) = 31 ± 7 kJ mol-1, in different range of temperatures depending of the peptide. Furthermore, the diffusion coefficient correlated with increasing temperature in GY7 and SY7 fibers for temperatures compress between 20 °C and 80 °C, while NY7 only below 60 °C. However, it is noteworthy that the diffusivity observed in the SY7 peptide is lower, compared to GY7 and NY7 presumably due to its enlarged length. This observation can be attributed to two factors: firstly, the higher conductivity values observed in SY7 compared to GY7 and NY7, and secondly, to the value of relation observed of cations present in the peptide SY7 compared with GY7 and NY7, which in turn is dependent on temperature. In light of these findings, we envision our prion-inspired nanofibers as highly efficient proton-conducting natural biopolymers that are both biocompatible and biodegradable. These properties provide the opportunity for the development of next-generation bioelectrical interfaces and protonic devices.

16.
J Colloid Interface Sci ; 674: 753-765, 2024 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-38955007

RESUMEN

The recent coronavirus disease 2019 (COVID-19) pandemic caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has spurred intense research efforts to develop new materials with antiviral activity. In this study, we genetically engineered amyloid-based nanofibrils for capturing and neutralizing SARS-CoV-2. Building upon the amyloid properties of a short Sup35 yeast prion sequence, we fused it to SARS-CoV-2 receptor-binding domain (RBD) capturing proteins, LCB1 and LCB3. By tuning the reaction conditions, we achieved the spontaneous self-assembly of the Sup35-LCB1 fusion protein into a highly homogeneous and well-dispersed amyloid-like fibrillar material. These nanofibrils exhibited high affinity for the SARS-CoV-2 RBD, effectively inhibiting its interaction with the angiotensin-converting enzyme 2 (ACE2) receptor, the primary entry point for the virus into host cells. We further demonstrate that this functional nanomaterial entraps and neutralizes SARS-CoV-2 virus-like particles (VLPs), with a potency comparable to that of therapeutic antibodies. As a proof of concept, we successfully fabricated patterned surfaces that selectively capture SARS-CoV-2 RBD protein on wet environments. Collectively, these findings suggest that these protein-only nanofibrils hold promise as disinfecting coatings endowed with selective SARS-CoV-2 neutralizing properties to combat viral spread or in the development of sensitive viral sampling and diagnostic tools.


Asunto(s)
Enzima Convertidora de Angiotensina 2 , COVID-19 , Nanofibras , SARS-CoV-2 , SARS-CoV-2/efectos de los fármacos , Humanos , Nanofibras/química , Enzima Convertidora de Angiotensina 2/metabolismo , Enzima Convertidora de Angiotensina 2/química , COVID-19/virología , Proteínas de Saccharomyces cerevisiae/química , Anticuerpos Neutralizantes/inmunología , Amiloide/química , Amiloide/metabolismo , Factores de Terminación de Péptidos
17.
Nat Commun ; 15(1): 4150, 2024 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-38755164

RESUMEN

Age-related neurodegenerative diseases involving amyloid aggregation remain one of the biggest challenges of modern medicine. Alterations in the gastrointestinal microbiome play an active role in the aetiology of neurological disorders. Here, we dissect the amyloidogenic properties of biofilm-associated proteins (BAPs) of the gut microbiota and their implications for synucleinopathies. We demonstrate that BAPs are naturally assembled as amyloid-like fibrils in insoluble fractions isolated from the human gut microbiota. We show that BAP genes are part of the accessory genomes, revealing microbiome variability. Remarkably, the abundance of certain BAP genes in the gut microbiome is correlated with Parkinson's disease (PD) incidence. Using cultured dopaminergic neurons and Caenorhabditis elegans models, we report that BAP-derived amyloids induce α-synuclein aggregation. Our results show that the chaperone-mediated autophagy is compromised by BAP amyloids. Indeed, inoculation of BAP fibrils into the brains of wild-type mice promote key pathological features of PD. Therefore, our findings establish the use of BAP amyloids as potential targets and biomarkers of α-synucleinopathies.


Asunto(s)
Amiloide , Biopelículas , Caenorhabditis elegans , Neuronas Dopaminérgicas , Microbioma Gastrointestinal , Enfermedad de Parkinson , alfa-Sinucleína , Animales , Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/microbiología , Humanos , Biopelículas/crecimiento & desarrollo , Amiloide/metabolismo , alfa-Sinucleína/metabolismo , alfa-Sinucleína/genética , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/microbiología , Enfermedad de Parkinson/patología , Ratones , Neuronas Dopaminérgicas/metabolismo , Autofagia , Enfermedades Neurodegenerativas/metabolismo , Ratones Endogámicos C57BL , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/genética , Encéfalo/metabolismo , Encéfalo/patología , Sinucleinopatías/metabolismo , Sinucleinopatías/patología
18.
ACS Appl Mater Interfaces ; 15(9): 11444-11457, 2023 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-36890692

RESUMEN

The coronavirus disease 2019 (COVID-19) pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection has become a primary health concern. Molecules that prevent viral entry into host cells by interfering with the interaction between SARS-CoV-2 spike (S) protein and the human angiotensin-converting enzyme 2 receptor (ACE2r) opened a promising avenue for virus neutralization. Here, we aimed to create a novel kind of nanoparticle that can neutralize SARS-CoV-2. To this purpose, we exploited a modular self-assembly strategy to engineer OligoBinders, soluble oligomeric nanoparticles decorated with two miniproteins previously described to bind to the S protein receptor binding domain (RBD) with high affinity. The multivalent nanostructures compete with the RBD-ACE2r interaction and neutralize SARS-CoV-2 virus-like particles (SC2-VLPs) with IC50 values in the pM range, preventing SC2-VLPs fusion with the membrane of ACE2r-expressing cells. Moreover, OligoBinders are biocompatible and significantly stable in plasma. Overall, we describe a novel protein-based nanotechnology that might find application in SARS-CoV-2 therapeutics and diagnostics.


Asunto(s)
COVID-19 , Nanopartículas , Humanos , SARS-CoV-2 , Unión Proteica , Proteínas Amiloidogénicas
19.
ACS Nano ; 17(17): 16968-16979, 2023 09 12.
Artículo en Inglés | MEDLINE | ID: mdl-37647583

RESUMEN

Enzymes typically fold into defined 3D protein structures exhibiting a high catalytic efficiency and selectivity. It has been proposed that the earliest enzymes may have arisen from the self-assembly of short peptides into supramolecular amyloid-like structures. Several artificial amyloids have been shown to display catalytic activity while offering advantages over natural enzymes in terms of modularity, flexibility, stability, and reusability. Hydrolases, especially esterases, are the most common artificial amyloid-like nanozymes with some reported to act as carbonic anhydrases (CA). Their hydrolytic activity is often dependent on the binding of metallic cofactors through a coordination triad composed of His residues in the ß-strands, which mimic the arrangement found in natural metalloenzymes. Tyr residues contribute to the coordination of metal ions in the active center of metalloproteins; however, their use has been mostly neglected in the design of metal-containing amyloid-based nanozymes. We recently reported that four different polar prion-inspired heptapeptides spontaneously self-assembled into amyloid fibrils. Their sequences lack His but contain three alternate Tyr residues exposed to solvent. We combine experiments and simulations to demonstrate that the amyloid fibrils formed by these peptides can efficiently coordinate and retain different divalent metal cations, functioning as both metal scavengers and nanozymes. The metallized fibrils exhibit esterase and CA activities without the need for a histidine triad. These findings highlight the functional versatility of prion-inspired peptide assemblies and provide a new sequential context for the creation of artificial metalloenzymes. Furthermore, our data support amyloid-like structures acting as ancestral catalysts at the origin of life.


Asunto(s)
Metaloproteínas , Priones , Amiloide , Péptidos , Proteínas Amiloidogénicas
20.
Eur J Med Chem ; 261: 115837, 2023 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-37837673

RESUMEN

The aggregation of wild-type transthyretin (TTR) and over 130 genetic TTR variants underlies a group of lethal disorders named TTR amyloidosis (ATTR). TTR chemical chaperones are molecules that hold great promise to modify the course of ATTR progression. In previous studies, we combined rational design and molecular dynamics simulations to generate a series of TTR selective kinetic stabilizers displaying exceptionally high affinities. In an effort to endorse the previously developed molecules with optimal pharmacokinetic properties, we conducted structural design optimization, leading to the development of PITB. PITB binds with high affinity to TTR, effectively inhibiting tetramer dissociation and aggregation of both the wild-type protein and the two most prevalent disease-associated TTR variants. Importantly, PITB selectively binds and stabilizes TTR in plasma, outperforming tolcapone, a drug currently undergoing clinical trials for ATTR. Pharmacokinetic studies conducted on mice confirmed that PITB exhibits encouraging pharmacokinetic properties, as originally intended. Furthermore, PITB demonstrates excellent oral bioavailability and lack of toxicity. These combined attributes position PITB as a lead compound for future clinical trials as a disease-modifying therapy for ATTR.


Asunto(s)
Neuropatías Amiloides Familiares , Prealbúmina , Ratones , Animales , Prealbúmina/metabolismo , Neuropatías Amiloides Familiares/tratamiento farmacológico , Neuropatías Amiloides Familiares/metabolismo , Tolcapona/uso terapéutico , Simulación de Dinámica Molecular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA