Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 106
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Bioconjug Chem ; 2024 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-38865349

RESUMEN

Peptide fibrillization is crucial in biological processes such as amyloid-related diseases and hormone storage, involving complex transitions between folded, unfolded, and aggregated states. We here employ light to induce reversible transitions between aggregated and nonaggregated states of a peptide, linked to the parathyroid hormone (PTH). The artificial light-switch 3-{[(4-aminomethyl)phenyl]diazenyl}benzoic acid (AMPB) is embedded into a segment of PTH, the peptide PTH25-37, to control aggregation, revealing position-dependent effects. Through in silico design, synthesis, and experimental validation of 11 novel PTH25-37-derived peptides, we predict and confirm the amyloid-forming capabilities of the AMPB-containing peptides. Quantum-chemical studies shed light on the photoswitching mechanism. Solid-state NMR studies suggest that ß-strands are aligned parallel in fibrils of PTH25-37, while in one of the AMPB-containing peptides, ß-strands are antiparallel. Simulations further highlight the significance of π-π interactions in the latter. This multifaceted approach enabled the identification of a peptide that can undergo repeated phototriggered transitions between fibrillated and defibrillated states, as demonstrated by different spectroscopic techniques. With this strategy, we unlock the potential to manipulate PTH to reversibly switch between active and inactive aggregated states, representing the first observation of a photostimulus-responsive hormone.

2.
Plant Physiol ; 192(1): 504-526, 2023 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-36493393

RESUMEN

Organisms require micronutrients, and Arabidopsis (Arabidopsis thaliana) IRON-REGULATED TRANSPORTER1 (IRT1) is essential for iron (Fe2+) acquisition into root cells. Uptake of reactive Fe2+ exposes cells to the risk of membrane lipid peroxidation. Surprisingly little is known about how this is avoided. IRT1 activity is controlled by an intracellular variable region (IRT1vr) that acts as a regulatory protein interaction platform. Here, we describe that IRT1vr interacted with peripheral plasma membrane SEC14-Golgi dynamics (SEC14-GOLD) protein PATELLIN2 (PATL2). SEC14 proteins bind lipophilic substrates and transport or present them at the membrane. To date, no direct roles have been attributed to SEC14 proteins in Fe import. PATL2 affected root Fe acquisition responses, interacted with ROS response proteins in roots, and alleviated root lipid peroxidation. PATL2 had high affinity in vitro for the major lipophilic antioxidant vitamin E compound α-tocopherol. Molecular dynamics simulations provided insight into energetic constraints and the orientation and stability of the PATL2-ligand interaction in atomic detail. Hence, this work highlights a compelling mechanism connecting vitamin E with root metal ion transport at the plasma membrane with the participation of an IRT1-interacting and α-tocopherol-binding SEC14 protein.


Asunto(s)
Proteínas de Arabidopsis , Arabidopsis , Proteínas de Arabidopsis/metabolismo , Vitamina E/metabolismo , alfa-Tocoferol , Transporte Biológico , Arabidopsis/genética , Arabidopsis/metabolismo , Raíces de Plantas/metabolismo , Regulación de la Expresión Génica de las Plantas
3.
Proc Natl Acad Sci U S A ; 118(39)2021 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-34544868

RESUMEN

Mounting evidence suggests that the neuronal cell membrane is the main site of oligomer-mediated neuronal toxicity of amyloid-ß peptides in Alzheimer's disease. To gain a detailed understanding of the mutual interference of amyloid-ß oligomers and the neuronal membrane, we carried out microseconds of all-atom molecular dynamics (MD) simulations on the dimerization of amyloid-ß (Aß)42 in the aqueous phase and in the presence of a lipid bilayer mimicking the in vivo composition of neuronal membranes. The dimerization in solution is characterized by a random coil to ß-sheet transition that seems on pathway to amyloid aggregation, while the interactions with the neuronal membrane decrease the order of the Aß42 dimer by attenuating its propensity to form a ß-sheet structure. The main lipid interaction partners of Aß42 are the surface-exposed sugar groups of the gangliosides GM1. As the neurotoxic activity of amyloid oligomers increases with oligomer order, these results suggest that GM1 is neuroprotective against Aß-mediated toxicity.


Asunto(s)
Péptidos beta-Amiloides/química , Péptidos beta-Amiloides/metabolismo , Amiloide/química , Membrana Celular/metabolismo , Gangliósido G(M1)/metabolismo , Neuronas/metabolismo , Multimerización de Proteína , Humanos , Membrana Dobles de Lípidos/metabolismo , Simulación de Dinámica Molecular , Unión Proteica , Conformación Proteica
4.
Proteins ; 2023 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-37964477

RESUMEN

Among the various factors controlling the amyloid aggregation process, the influences of ions on the aggregation rate and the resulting structures are important aspects to consider, which can be studied by molecular simulations. There is a wide variety of protein force fields and ion models, raising the question of which model to use in such studies. To address this question, we perform molecular dynamics simulations of Aß16-22 , a fragment of the Alzheimer's amyloid ß peptide, using different protein force fields, AMBER99SB-disp (A99-d) and CHARMM36m (C36m), and different ion parameters. The influences of NaCl and CaCl2 at various concentrations are studied and compared with the systems without the addition of ions. Our results indicate a sensitivity of the peptide-ion interactions to the different ion models. In particular, we observe a strong binding of Ca2+ to residue E22 with C36m and also with the Åqvist ion model used together with A99-d, which slightly affects the monomeric Aß16-22 structures and the aggregation rate, but significantly affects the oligomer structures formed in the aggregation simulations. For example, at high Ca2+ concentrations, there was a switch from an antiparallel to a parallel ß-sheet. Such ionic influences are of biological relevance because local ion concentrations can change in vivo and could help explain the polymorphism of amyloid fibrils.

5.
J Am Chem Soc ; 145(33): 18340-18354, 2023 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-37555670

RESUMEN

The amyloid-ß (Aß) peptide is associated with the development of Alzheimer's disease and is known to form highly neurotoxic prefibrillar oligomeric aggregates, which are difficult to study due to their transient, low-abundance, and heterogeneous nature. To obtain high-resolution information about oligomer structure and dynamics as well as relative populations of assembly states, we here employ a combination of native ion mobility mass spectrometry and molecular dynamics simulations. We find that the formation of Aß oligomers is dependent on the presence of a specific ß-hairpin motif in the peptide sequence. Oligomers initially grow spherically but start to form extended linear aggregates at oligomeric states larger than those of the tetramer. The population of the extended oligomers could be notably increased by introducing an intramolecular disulfide bond, which prearranges the peptide in the hairpin conformation, thereby promoting oligomeric structures but preventing conversion into mature fibrils. Conversely, truncating one of the ß-strand-forming segments of Aß decreased the hairpin propensity of the peptide and thus decreased the oligomer population, removed the formation of extended oligomers entirely, and decreased the aggregation propensity of the peptide. We thus propose that the observed extended oligomer state is related to the formation of an antiparallel sheet state, which then nucleates into the amyloid state. These studies provide increased mechanistic understanding of the earliest steps in Aß aggregation and suggest that inhibition of Aß folding into the hairpin conformation could be a viable strategy for reducing the amount of toxic oligomers.


Asunto(s)
Enfermedad de Alzheimer , Péptidos beta-Amiloides , Humanos , Péptidos beta-Amiloides/química , Conformación Proteica , Simulación de Dinámica Molecular , Fragmentos de Péptidos/química
6.
Chem Rev ; 121(13): 8285-8307, 2021 07 14.
Artículo en Inglés | MEDLINE | ID: mdl-34137605

RESUMEN

This review will focus on the process of amyloid-type protein aggregation. Amyloid fibrils are an important hallmark of protein misfolding diseases and therefore have been investigated for decades. Only recently, however, atomic or near-atomic resolution structures have been elucidated from various in vitro and ex vivo obtained fibrils. In parallel, the process of fibril formation has been studied in vitro under highly artificial but comparatively reproducible conditions. The review starts with a summary of what is known and speculated from artificial in vitro amyloid-type protein aggregation experiments. A partially hypothetic fibril selection model will be described that may be suitable to explain why amyloid fibrils look the way they do, in particular, why at least all so far reported high resolution cryo-electron microscopy obtained fibril structures are in register, parallel, cross-ß-sheet fibrils that mostly consist of two protofilaments twisted around each other. An intrinsic feature of the model is the prion-like nature of all amyloid assemblies. Transferring the model from the in vitro point of view to the in vivo situation is not straightforward, highly hypothetic, and leaves many open questions that need to be addressed in the future.


Asunto(s)
Amiloide/química , Proteínas Amiloidogénicas/química , Priones/química , Agregado de Proteínas , Amiloide/ultraestructura , Proteínas Amiloidogénicas/ultraestructura , Animales , Microscopía por Crioelectrón , Humanos , Priones/ultraestructura
7.
Chem Rev ; 121(4): 2545-2647, 2021 02 24.
Artículo en Inglés | MEDLINE | ID: mdl-33543942

RESUMEN

Protein misfolding and aggregation is observed in many amyloidogenic diseases affecting either the central nervous system or a variety of peripheral tissues. Structural and dynamic characterization of all species along the pathways from monomers to fibrils is challenging by experimental and computational means because they involve intrinsically disordered proteins in most diseases. Yet understanding how amyloid species become toxic is the challenge in developing a treatment for these diseases. Here we review what computer, in vitro, in vivo, and pharmacological experiments tell us about the accumulation and deposition of the oligomers of the (Aß, tau), α-synuclein, IAPP, and superoxide dismutase 1 proteins, which have been the mainstream concept underlying Alzheimer's disease (AD), Parkinson's disease (PD), type II diabetes (T2D), and amyotrophic lateral sclerosis (ALS) research, respectively, for many years.


Asunto(s)
Amiloide/química , Amiloide/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/química , Péptidos beta-Amiloides/metabolismo , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/metabolismo , Esclerosis Amiotrófica Lateral/patología , Animales , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patología , Humanos , Polipéptido Amiloide de los Islotes Pancreáticos/química , Polipéptido Amiloide de los Islotes Pancreáticos/metabolismo , Modelos Moleculares , Enfermedades Neurodegenerativas/patología , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología , Agregación Patológica de Proteínas , Deficiencias en la Proteostasis/metabolismo , Superóxido Dismutasa-1/química , Superóxido Dismutasa-1/metabolismo , alfa-Sinucleína/química , alfa-Sinucleína/metabolismo , Proteínas tau/química , Proteínas tau/metabolismo
8.
Methods ; 206: 18-26, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35934194

RESUMEN

Intrinsically disordered proteins (IDPs) do not fold into a unique three-dimensional structure but sample different configurations of different probabilities that further change with the surrounding of the IDPs. The structural heterogeneity and dynamics of IDPs pose a challenge for the characterization of their structures by experimental techniques only. Molecular dynamics (MD) simulations provide a powerful complement to experimental approaches for that purpose. However, MD simulations on the micro- to millisecond timescale generate a lot of data of protein motions, necessitating advanced post-processing techniques to extract the relevant information. Here, we demonstrate how transition networks created from MD trajectories allow revealing the configurational ensemble and structural interconversions of IDPs, using the amyloid-ß peptide as example. The construction of transition networks relies on molecular descriptors as input, and we show how the choice of descriptors influences the resulting transition network. The transition networks are generated with the open-source Python script ATRANET, and we explain the usage of ATRANET by providing a detailed workflow and exemplary analysis for amyloid-ß, which can be easily generalized to other IDPs and even protein aggregation.


Asunto(s)
Proteínas Intrínsecamente Desordenadas , Péptidos beta-Amiloides , Proteínas Intrínsecamente Desordenadas/química , Simulación de Dinámica Molecular , Agregado de Proteínas , Conformación Proteica
9.
Int J Mol Sci ; 24(14)2023 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-37510997

RESUMEN

The aggregation of amyloid-ß (Aß) peptides, particularly of Aß1-42, has been linked to the pathogenesis of Alzheimer's disease. In this study, we focus on the conformational change of Aß1-42 in the presence of glycosaminoglycans (GAGs) and 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine (POPC) lipids using molecular dynamics simulations. We analyze the conformational changes that occur in Aß by extracting the key structural features that are then used to generate transition networks. Using the same three features per network highlights the transitions from intrinsically disordered states ubiquitous in Aß1-42 in solution to more compact states arising from stable ß-hairpin formation when Aß1-42 is in the vicinity of a GAG molecule, and even more compact states characterized by a α-helix or ß-sheet structures when Aß1-42 interacts with a POPC lipid cluster. We show that the molecular mechanisms underlying these transitions from disorder to order are different for the Aß1-42/GAG and Aß1-42/POPC systems. While in the latter the hydrophobicity provided by the lipid tails facilitates the folding of Aß1-42, in the case of GAG there are hardly any intermolecular Aß1-42-GAG interactions. Instead, GAG removes sodium ions from the peptide, allowing stronger electrostatic interactions within the peptide that stabilize a ß-hairpin. Our results contribute to the growing knowledge of the role of GAGs and lipids in the conformational preferences of the Aß peptide, which in turn influences its aggregation into toxic oligomers and amyloid fibrils.


Asunto(s)
Enfermedad de Alzheimer , Glicosaminoglicanos , Humanos , Péptidos beta-Amiloides/química , Simulación de Dinámica Molecular , Amiloide/química , Fragmentos de Péptidos/química
10.
Proteins ; 90(11): 1811-1824, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35475513

RESUMEN

Sickle cell disease is a hemoglobinopathy resulting from a point mutation from glutamate to valine at position six of the ß-globin chains of hemoglobin. This mutation gives rise to pathological aggregation of the sickle hemoglobin and, as a result, impaired oxygen binding, misshapen and short-lived erythrocytes, and anemia. We aim to understand the structural effects caused by the single Glu6Val mutation leading to protein aggregation. To this end, we perform multiscale molecular dynamics simulations employing atomistic and coarse-grained models of both wild-type and sickle hemoglobin. We analyze the dynamics of hemoglobin monomers and dimers, study the aggregation of wild-type and sickle hemoglobin into decamers, and analyze the protein-protein interactions in the resulting aggregates. We find that the aggregation of sickle hemoglobin is driven by both hydrophobic and electrostatic protein-protein interactions involving the mutation site and surrounding residues, leading to an extended interaction area and thus stable aggregates. The wild-type protein can also self-assemble, which, however, results from isolated interprotein salt bridges that do not yield stable aggregates. This knowledge can be exploited for the development of sickle hemoglobin-aggregation inhibitors.


Asunto(s)
Hemoglobina Falciforme , Agregado de Proteínas , Glutamatos , Hemoglobina Falciforme/genética , Hemoglobina Falciforme/metabolismo , Hemoglobinas/química , Oxígeno/metabolismo , Valina , Globinas beta
11.
Bioorg Chem ; 111: 104862, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33862474

RESUMEN

For the COVID-19 pandemic caused by SARS-CoV-2, there are currently no effective drugs or vaccines to treat this coronavirus infection. In this study, we focus on the main protease enzyme of SARS-CoV-2, 3CLpro, which is critical for viral replication. We employ explicit solvent molecular dynamics simulations of about 150 compounds docked into 3CLpro's binding site and that had emerged as good main protease ligands from our previous in silico screening of over 1.2 million compounds. By incoporating protein dynamics and applying a range of structural descriptors, such as the ability to form specific contacts with the catalytic dyad residues of 3CLpro and the structural fluctuations of the ligands in the binding site, we are able to further refine our compound selection. Fourteen compounds including estradiol shown to be the most promising based on our calculations were procured and screened against recombinant 3CLpro in a fluorescence assay. Eight of these compounds have significant activity in inhibiting the SARS-CoV-2 main protease. Among these are corilagin, a gallotannin, and lurasidone, an antipsychotic drug, which emerged as the most promising natural product and drug, respectively, and might thus be candidates for drug repurposing for the treatment of COVID-19. In addition, we also tested the inhibitory activity of testosterone, and our results reveal testosterone as possessing moderate inhibitory potency against the 3CLpro enzyme, which may thus provide an explanation why older men are more severely affected by COVID-19.


Asunto(s)
Proteasas 3C de Coronavirus/antagonistas & inhibidores , Inhibidores de Proteasas/metabolismo , SARS-CoV-2/enzimología , Bibliotecas de Moléculas Pequeñas/metabolismo , Antivirales/metabolismo , Sitios de Unión , Proteasas 3C de Coronavirus/metabolismo , Pruebas de Enzimas , Ligandos , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Unión Proteica
12.
Int J Mol Sci ; 22(21)2021 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-34768961

RESUMEN

The increasing recognition of the biochemical importance of glycosaminoglycans (GAGs) has in recent times made them the center of attention of recent research investigations. It became evident that subtle conformational factors play an important role in determining the relationship between the chemical composition of GAGs and their activity. Therefore, a thorough understanding of their structural flexibility is needed, which is addressed in this work by means of all-atom molecular dynamics (MD) simulations. Four major GAGs with different substitution patterns, namely hyaluronic acid as unsulphated GAG, heparan-6-sulphate, chondroitin-4-sulphate, and chondroitin-6-sulphate, were investigated to elucidate the influence of sulphation on the dynamical features of GAGs. Moreover, the effects of increasing NaCl and KCl concentrations were studied as well. Different structural parameters were determined from the MD simulations, in combination with a presentation of the free energy landscape of the GAG conformations, which allowed us to unravel the conformational fingerprints unique to each GAG. The largest effects on the GAG structures were found for sulphation at position 6, as well as binding of the metal ions in the absence of chloride ions to the carboxylate and sulphate groups, which both increase the GAG conformational flexibility.


Asunto(s)
Glicosaminoglicanos/química , Conformación de Carbohidratos , Secuencia de Carbohidratos , Sulfatos de Condroitina/química , Sulfatos de Condroitina/metabolismo , Glicosaminoglicanos/metabolismo , Heparitina Sulfato/química , Heparitina Sulfato/metabolismo , Humanos , Ácido Hialurónico/química , Ácido Hialurónico/metabolismo , Enlace de Hidrógeno , Interacciones Hidrofóbicas e Hidrofílicas , Simulación de Dinámica Molecular , Estructura Molecular , Cloruro de Potasio/química , Cloruro de Sodio/química , Sulfatos/química
13.
PLoS Comput Biol ; 15(10): e1007193, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31589600

RESUMEN

Guanylate binding proteins (GBPs) belong to the dynamin-related superfamily and exhibit various functions in the fight against infections. The functions of the human guanylate binding protein 1 (hGBP1) are tightly coupled to GTP hydrolysis and dimerization. Despite known crystal structures of the hGBP1 monomer and GTPase domain dimer, little is known about the dynamics of hGBP1. To gain a mechanistic understanding of hGBP1, we performed sub-millisecond multi-resolution molecular dynamics simulations of both the hGBP1 monomer and dimer. We found that hGBP1 is a highly flexible protein that undergoes a hinge motion similar to the movements observed for other dynamin-like proteins. Another large-scale motion was observed for the C-terminal helix α13, providing a molecular view for the α13-α13 distances previously reported for the hGBP1 dimer. Most of the loops of the GTPase domain were found to be flexible, revealing why GTP binding is needed for hGBP1 dimerization to occur.


Asunto(s)
Biología Computacional/métodos , Proteínas de Unión al GTP/fisiología , Algoritmos , Sitios de Unión , Simulación por Computador , Dimerización , Dinaminas , GTP Fosfohidrolasas/metabolismo , Guanosina Trifosfato/metabolismo , Humanos , Cinética , Simulación de Dinámica Molecular , Movimiento (Física) , Unión Proteica , Dominios y Motivos de Interacción de Proteínas/fisiología , Programas Informáticos
14.
Eur Biophys J ; 49(2): 175-191, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-32123956

RESUMEN

Protein misfolding and subsequent self-association are complex, intertwined processes, resulting in development of a heterogeneous population of aggregates closely related to many chronic pathological conditions including Type 2 Diabetes Mellitus and Alzheimer's disease. To address this issue, here, we develop a theoretical model in the general framework of linear stability analysis. According to this model, self-assemblies of peptides with pronounced conformational flexibility may become, under particular conditions, unstable and spontaneously evolve toward an alternating array of partially ordered and disordered monomers. The predictions of the theory were verified by atomistic molecular dynamics (MD) simulations of islet amyloid polypeptide (IAPP) used as a paradigm of aggregation-prone polypeptides (proteins). Simulations of dimeric, tetrameric, and hexameric human-IAPP self-assemblies at physiological electrolyte concentration reveal an alternating distribution of the smallest domains (of the order of the peptide mean length) formed by partially ordered (mainly ß-strands) and disordered (turns and coil) arrays. Periodicity disappears upon weakening of the inter-peptide binding, a result in line with the predictions of the theory. To further probe the general validity of our hypothesis, we extended the simulations to other peptides, the Aß(1-40) amyloid peptide, and the ovine prion peptide as well as to other proteins (SOD1 dimer) that do not belong to the broad class of intrinsically disordered proteins. In all cases, the oligomeric aggregates show an alternate distribution of partially ordered and disordered monomers. We also carried out Surface Enhanced Raman Scattering (SERS) measurements of hIAPP as an experimental validation of both the theory and in silico simulations.


Asunto(s)
Polipéptido Amiloide de los Islotes Pancreáticos/química , Desnaturalización Proteica , Pliegue de Proteína , Coloides/química , Simulación por Computador , Electrólitos , Humanos , Cinética , Modelos Teóricos , Simulación de Dinámica Molecular , Péptidos/química , Multimerización de Proteína , Estructura Secundaria de Proteína , Reproducibilidad de los Resultados , Solventes , Espectrometría Raman , Termodinámica
15.
J Chem Inf Model ; 60(12): 6462-6475, 2020 12 28.
Artículo en Inglés | MEDLINE | ID: mdl-33174726

RESUMEN

The progress toward understanding the molecular basis of Alzheimers's disease is strongly connected to elucidating the early aggregation events of the amyloid-ß (Aß) peptide. Molecular dynamics (MD) simulations provide a viable technique to study the aggregation of Aß into oligomers with high spatial and temporal resolution. However, the results of an MD simulation can only be as good as the underlying force field. A recent study by our group showed that none of the common force fields can distinguish between aggregation-prone and nonaggregating peptide sequences, producing a similar and in most cases too fast aggregation kinetics for all peptides. Since then, new force fields specially designed for intrinsically disordered proteins such as Aß were developed. Here, we assess the applicability of these new force fields to studying peptide aggregation using the Aß16-22 peptide and mutations of it as test case. We investigate their performance in modeling the monomeric state, the aggregation into oligomers, and the stability of the aggregation end product, i.e., the fibrillar state. A main finding is that changing the force field has a stronger effect on the simulated aggregation pathway than changing the peptide sequence. Also the new force fields are not able to reproduce the experimental aggregation propensity order of the peptides. Dissecting the various energy contributions shows that AMBER99SB-disp overestimates the interactions between the peptides and water, thereby inhibiting peptide aggregation. More promising results are obtained with CHARMM36m and especially its version with increased protein-water interactions. It is thus recommended to use this force field for peptide aggregation simulations and base future reparameterizations on it.


Asunto(s)
Proteínas Intrínsecamente Desordenadas , Simulación de Dinámica Molecular , Amiloide , Péptidos beta-Amiloides , Cinética , Fragmentos de Péptidos
16.
Bioorg Chem ; 101: 104012, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32683138

RESUMEN

Aggregation of amyloid peptides results in severe neurodegenerative diseases. While the fibril structures of Aß40 and Aß42 have been described recently, resolution of the aggregation pathway and evaluation of potent inhibitors still remains elusive, in particular in view of the hairpin-region of Aß40. We here report the preparation of beta-turn mimetic conjugates containing synthetic turn mimetic structures in the turn region of Aß40 and Aß16-35, replacing 2 amino acids in the turn-region G25 - K28. The structure of the turn mimic induces both, acceleration of fibrillation and the complete inhibition of fibrillation, confirming the importance of the turn region on the aggregation. Replacing position G25-S26 provided the best inhibition effect for both beta-turn mimetics, the bicyclic BTD 1 and the aromatic TAA 2, while positions N27-K28 and V24-G25 showed only weaker or no inhibitory effects. When comparing different turn mimetics at the same position (G25-S26), conjugate 1a bearing the BTD turn showed the best inhibition of Aß40 aggregation, while 5-amino-valeric acid 4a showed the weakest effect. Thus there is a pronounced impact on fibrillation with the chemical nature of the embedded beta-turn-mimic: the conformationally constrained turns 1 and 2 lead to a significantly reduced fibrillation, even inhibiting fibrillation of native Aß40 when added in amounts down to 1/10, whereas the more flexible beta-turn-mimics 4-amino-benzoic acid 3a and 5-amino-valeric acid 4a lead to enhanced fibrillation. Toxicity-testing of the most successful conjugate showed only minor toxicity in cell-viability assays using the N2a cell line. Structural downsizing lead to the short fragment BTD/peptide Aß16-35 as inhibitor of the aggregation of Aß40, opening large potential for further small peptide based inhibitors.


Asunto(s)
Péptidos beta-Amiloides/antagonistas & inhibidores , Imitación Molecular , Aminoácidos/química , Péptidos beta-Amiloides/química , Biopolímeros/química , Línea Celular , Supervivencia Celular/efectos de los fármacos , Humanos , Simulación de Dinámica Molecular
17.
Biochem J ; 476(21): 3161-3182, 2019 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-31689351

RESUMEN

Guanylate-binding proteins (GBPs) constitute a family of interferon-inducible guanosine triphosphatases (GTPases) that are key players in host defense against intracellular pathogens ranging from protozoa to bacteria and viruses. So far, human GBP1 and GBP5 as well as murine GBP2 (mGBP2) have been biochemically characterized in detail. Here, with murine GBP7 (mGBP7), a GBP family member with an unconventional and elongated C-terminus is analyzed. The present study demonstrates that mGBP7 exhibits a concentration-dependent GTPase activity and an apparent GTP turnover number of 20 min-1. In addition, fluorescence spectroscopy analyses reveal that mGBP7 binds GTP with high affinity (KD = 0.22 µM) and GTPase activity assays indicate that mGBP7 hydrolyzes GTP to GDP and GMP. The mGBP7 GTPase activity is inhibited by incubation with γ-phosphate analogs and a K51A mutation interfering with GTP binding. SEC-MALS analyses give evidence that mGBP7 forms transient dimers and that this oligomerization pattern is not influenced by the presence of nucleotides. Moreover, a structural model for mGBP7 is provided by homology modeling, which shows that the GTPase possesses an elongated C-terminal (CT) tail compared with the CaaX motif-containing mGBP2 and human GBP1. Molecular dynamics simulations indicate that this tail has transmembrane characteristics and, interestingly, confocal microscopy analyses reveal that the CT tail is required for recruitment of mGBP7 to the parasitophorous vacuole of Toxoplasma gondii.


Asunto(s)
Proteínas de Unión al GTP/química , Proteínas de Unión al GTP/metabolismo , Secuencias de Aminoácidos , Animales , Proteínas de Unión al GTP/genética , Guanosina Trifosfato/química , Guanosina Trifosfato/metabolismo , Humanos , Cinética , Ratones , Simulación de Dinámica Molecular , Dominios Proteicos , Toxoplasma/fisiología , Toxoplasmosis/enzimología , Toxoplasmosis/genética , Toxoplasmosis/metabolismo , Toxoplasmosis/parasitología
18.
Chem Soc Rev ; 48(14): 3946-3996, 2019 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-31192324

RESUMEN

One of the grand challenges of biophysical chemistry is to understand the principles that govern protein misfolding and aggregation, which is a highly complex process that is sensitive to initial conditions, operates on a huge range of length- and timescales, and has products that range from protein dimers to macroscopic amyloid fibrils. Aberrant aggregation is associated with more than 25 diseases, which include Alzheimer's, Parkinson's, Huntington's, and type II diabetes. Amyloid aggregation has been extensively studied in the test tube, therefore under conditions that are far from physiological relevance. Hence, there is dire need to extend these investigations to in vivo conditions where amyloid formation is affected by a myriad of biochemical interactions. As a hallmark of neurodegenerative diseases, these interactions need to be understood in detail to develop novel therapeutic interventions, as millions of people globally suffer from neurodegenerative disorders and type II diabetes. The aim of this review is to document the progress in the research on amyloid formation from a physicochemical perspective with a special focus on the physiological factors influencing the aggregation of the amyloid-ß peptide, the islet amyloid polypeptide, α-synuclein, and the hungingtin protein.


Asunto(s)
Amiloide/química , Agregado de Proteínas , Agregación Patológica de Proteínas , Animales , Humanos
19.
Molecules ; 25(14)2020 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-32668701

RESUMEN

We use state-of-the-art computer-aided drug design (CADD) techniques to identify prospective inhibitors of the main protease enzyme, 3CLpro of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causing COVID-19. From our screening of over one million compounds including approved drugs, investigational drugs, natural products, and organic compounds, and a rescreening protocol incorporating enzyme dynamics via ensemble docking, we have been able to identify a range of prospective 3CLpro inhibitors. Importantly, some of the identified compounds had previously been reported to exhibit inhibitory activities against the 3CLpro enzyme of the closely related SARS-CoV virus. The top-ranking compounds are characterized by the presence of multiple bi- and monocyclic rings, many of them being heterocycles and aromatic, which are flexibly linked allowing the ligands to adapt to the geometry of the 3CLpro substrate site and involve a high amount of functional groups enabling hydrogen bond formation with surrounding amino acid residues, including the catalytic dyad residues H41 and C145. Among the top binding compounds we identified several tyrosine kinase inhibitors, which include a bioflavonoid, the group of natural products that binds best to 3CLpro. Another class of compounds that decently binds to the SARS-CoV-2 main protease are steroid hormones, which thus may be endogenous inhibitors and might provide an explanation for the age-dependent severity of COVID-19. Many of the compounds identified by our work show a considerably stronger binding than found for reference compounds with in vitro demonstrated 3CLpro inhibition and anticoronavirus activity. The compounds determined in this work thus represent a good starting point for the design of inhibitors of SARS-CoV-2 replication.


Asunto(s)
Betacoronavirus/enzimología , Infecciones por Coronavirus/tratamiento farmacológico , Descubrimiento de Drogas , Neumonía Viral/tratamiento farmacológico , Inhibidores de Proteasas/farmacología , Proteínas no Estructurales Virales/antagonistas & inhibidores , Sitios de Unión , COVID-19 , Simulación por Computador , Proteasas 3C de Coronavirus , Cisteína Endopeptidasas , Diseño de Fármacos , Humanos , Concentración 50 Inhibidora , Ligandos , Modelos Moleculares , Estructura Molecular , Pandemias , SARS-CoV-2 , Programas Informáticos , Termodinámica
20.
J Chem Inf Model ; 59(5): 1782-1805, 2019 05 28.
Artículo en Inglés | MEDLINE | ID: mdl-30933519

RESUMEN

Monomers and oligomers of the amyloid-ß peptide aggregate to form the fibrils found in the brains of Alzheimer's disease patients. These monomers and oligomers are largely disordered and can interact with transition metal ions, affecting the mechanism and kinetics of amyloid-ß aggregation. Due to the disordered nature of amyloid-ß, its rapid aggregation, as well as solvent and paramagnetic effects, experimental studies face challenges in the characterization of transition metal ions bound to amyloid-ß monomers and oligomers. The details of the coordination chemistry between transition metals and amyloid-ß obtained from experiments remain debated. Furthermore, the impact of transition metal ion binding on the monomeric or oligomeric amyloid-ß structures and dynamics are still poorly understood. Computational chemistry studies can serve as an important complement to experimental studies and can provide additional knowledge on the binding between amyloid-ß and transition metal ions. Many research groups conducted first-principles calculations, ab initio molecular dynamics simulations, quantum mechanics/classical mechanics simulations, and classical molecular dynamics simulations for studying the interplay between transition metal ions and amyloid-ß monomers and oligomers. This review summarizes the current understanding of transition metal interactions with amyloid-ß obtained from computational chemistry studies. We also emphasize the current view of the coordination chemistry between transition metal ions and amyloid-ß. This information represents an important foundation for future metal ion chelator and drug design studies aiming to combat Alzheimer's disease.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Cobre/metabolismo , Hierro/metabolismo , Zinc/metabolismo , Péptidos beta-Amiloides/química , Animales , Química Computacional , Cobre/química , Humanos , Iones/química , Iones/metabolismo , Hierro/química , Simulación de Dinámica Molecular , Zinc/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA