Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 61.393
Filter
1.
Nat Commun ; 15(1): 4217, 2024 May 17.
Article in English | MEDLINE | ID: mdl-38760359

ABSTRACT

Helix mimicry provides probes to perturb protein-protein interactions (PPIs). Helical conformations can be stabilized by joining side chains of non-terminal residues (stapling) or via capping fragments. Nature exclusively uses capping, but synthetic helical mimics are heavily biased towards stapling. This study comprises: (i) creation of a searchable database of unique helical N-caps (ASX motifs, a protein structural motif with two intramolecular hydrogen-bonds between aspartic acid/asparagine and following residues); (ii) testing trends observed in this database using linear peptides comprising only canonical L-amino acids; and, (iii) novel synthetic N-caps for helical interface mimicry. Here we show many natural ASX motifs comprise hydrophobic triangles, validate their effect in linear peptides, and further develop a biomimetic of them, Bicyclic ASX Motif Mimics (BAMMs). BAMMs are powerful helix inducing motifs. They are synthetically accessible, and potentially useful to a broad section of the community studying disruption of PPIs using secondary structure mimics.


Subject(s)
Amino Acid Motifs , Computational Biology , Computational Biology/methods , Hydrogen Bonding , Peptides/chemistry , Peptides/metabolism , Hydrophobic and Hydrophilic Interactions , Protein Structure, Secondary , Models, Molecular , Amino Acid Sequence , Databases, Protein , Proteins/chemistry , Proteins/metabolism , Aspartic Acid/chemistry
2.
Nat Commun ; 15(1): 4204, 2024 May 17.
Article in English | MEDLINE | ID: mdl-38760374

ABSTRACT

Life continuously transduces energy to perform critical functions using energy stored in reactive molecules like ATP or NADH. ATP dynamically phosphorylates active sites on proteins and thereby regulates their function. Inspired by such machinery, regulating supramolecular functions using energy stored in reactive molecules has gained traction. Enzyme-free, synthetic systems that use dynamic phosphorylation to regulate supramolecular processes have not yet been reported, to our knowledge. Here, we show an enzyme-free reaction cycle that consumes the phosphorylating agent monoamidophosphate by transiently phosphorylating histidine and histidine-containing peptides. The phosphorylated species are labile and deactivate through hydrolysis. The cycle exhibits versatility and tunability, allowing for the dynamic phosphorylation of multiple precursors with a tunable half-life. Notably, we show the resulting phosphorylated products can regulate the peptide's phase separation, leading to active droplets that require the continuous conversion of fuel to sustain. The reaction cycle will be valuable as a model for biological phosphorylation but can also offer insights into protocell formation.


Subject(s)
Peptides , Phosphorylation , Peptides/metabolism , Peptides/chemistry , Histidine/metabolism , Histidine/chemistry , Adenosine Triphosphate/metabolism , Hydrolysis
3.
J Am Chem Soc ; 146(20): 14213-14224, 2024 May 22.
Article in English | MEDLINE | ID: mdl-38739765

ABSTRACT

The formation of an amide bond is an essential step in the synthesis of materials and drugs, and in the assembly of amino acids to form peptides. The mechanism of this reaction has been studied extensively, in particular to understand how it can be catalyzed, but a representation capable of explaining all the experimental data is still lacking. Numerical simulation should provide the necessary molecular description, but the solvent involvement poses a number of challenges. Here, we combine the efficiency and accuracy of neural network potential-based reactive molecular dynamics with the extensive and unbiased exploration of reaction pathways provided by transition path sampling. Using microsecond-scale simulations at the density functional theory level, we show that this method reveals the presence of two competing distinct mechanisms for peptide bond formation between alanine esters in aqueous solution. We describe how both reaction pathways, via a general base catalysis mechanism and via direct cleavage of the tetrahedral intermediate respectively, change with pH. This result contrasts with the conventional mechanism involving a single pathway in which only the barrier heights are affected by pH. We show that this new proposal involving two competing mechanisms is consistent with the experimental data, and we discuss the implications for peptide bond formation under prebiotic conditions and in the ribosome. Our work shows that integrating deep potential molecular dynamics with path sampling provides a powerful approach for exploring complex chemical mechanisms.


Subject(s)
Molecular Dynamics Simulation , Peptides , Water , Water/chemistry , Peptides/chemistry , Density Functional Theory , Hydrogen-Ion Concentration , Alanine/chemistry , Amides/chemistry
4.
Nano Lett ; 24(20): 6102-6111, 2024 May 22.
Article in English | MEDLINE | ID: mdl-38739578

ABSTRACT

Acute lung injury (ALI) is a severe inflammatory lung disease, with high mortality rates. Early intervention by reactive oxygen species (ROS) scavengers could reduce ROS accumulation, break the inflammation expansion chain in alveolar macrophages (AMs), and avoid irreversible damage to alveolar epithelial and endothelial cells. Here, we reported cell-penetrating R9 peptide-modified triangular DNA origami nanostructures (tDONs-R9) as a novel nebulizable drug that could reach the deep alveolar regions and exhibit an enhanced uptake preference of macrophages. tDONs-R9 suppressed the expression of pro-inflammatory cytokines and drove polarization toward the anti-inflammatory M2 phenotype in macrophages. In the LPS-induced ALI mouse model, treatment with nebulized tDONs-R9 alleviated the overwhelming ROS, pro-inflammatory cytokines, and neutrophil infiltration in the lungs. Our study demonstrates that tDONs-R9 has the potential for ALI treatment, and the programmable DNA origami nanostructures provide a new drug delivery platform for pulmonary disease treatment with high delivery efficiency and biosecurity.


Subject(s)
Acute Lung Injury , DNA , Nanostructures , Acute Lung Injury/drug therapy , Acute Lung Injury/pathology , Acute Lung Injury/chemically induced , Animals , Mice , DNA/chemistry , Administration, Inhalation , Nanostructures/chemistry , Reactive Oxygen Species/metabolism , Macrophages, Alveolar/drug effects , Macrophages, Alveolar/metabolism , Cytokines/metabolism , Peptides/chemistry , Nebulizers and Vaporizers , Cell-Penetrating Peptides/chemistry , Disease Models, Animal , Lipopolysaccharides , Drug Delivery Systems , RAW 264.7 Cells
5.
J Agric Food Chem ; 72(20): 11561-11576, 2024 May 22.
Article in English | MEDLINE | ID: mdl-38739709

ABSTRACT

The aim of this study is to validate the activity of hazelnut (Corylus avellana L.)-derived immunoactive peptides inhibiting the main protease (Mpro) of SARS-CoV-2 and further unveil their interaction mechanism using in vitro assays, molecular dynamics (MD) simulations, and binding free energy calculations. In general, the enzymatic hydrolysis components, especially molecular weight < 3 kDa, possess good immune activity as measured by the proliferation ability of mouse splenic lymphocytes and phagocytic activity of mouse peritoneal macrophages. Over 866 unique peptide sequences were isolated, purified, and then identified by nanohigh-performance liquid chromatography/tandem mass spectrometry (NANO-HPLC-MS/MS) from hazelnut protein hydrolysates, but Trp-Trp-Asn-Leu-Asn (WWNLN) and Trp-Ala-Val-Leu-Lys (WAVLK) in particular are found to increase the cell viability and phagocytic capacity of RAW264.7 macrophages as well as promote the secretion of the cytokines nitric oxide (NO), tumor necrosis factor-α (TNF-α), and interleukin-1ß (IL-1ß). Fluorescence resonance energy transfer assay elucidated that WWNLN and WAVLK exhibit excellent inhibitory potency against Mpro, with IC50 values of 6.695 and 16.750 µM, respectively. Classical all-atom MD simulations show that hydrogen bonds play a pivotal role in stabilizing the complex conformation and protein-peptide interaction. Molecular Mechanics/Generalized Born Surface Area (MM/GBSA) calculation indicates that WWNLN has a lower binding free energy with Mpro than WAVLK. Furthermore, adsorption, distribution, metabolism, excretion, and toxicity (ADMET) predictions illustrate favorable drug-likeness and pharmacokinetic properties of WWNLN compared to WAVLK. This study provides a new understanding of the immunomodulatory activity of hazelnut hydrolysates and sheds light on peptide inhibitors targeting Mpro.


Subject(s)
Corylus , Peptides , Mice , Animals , Peptides/chemistry , Peptides/pharmacology , RAW 264.7 Cells , Corylus/chemistry , SARS-CoV-2/drug effects , SARS-CoV-2/immunology , SARS-CoV-2/chemistry , Coronavirus 3C Proteases/chemistry , Coronavirus 3C Proteases/antagonists & inhibitors , Coronavirus 3C Proteases/metabolism , Molecular Dynamics Simulation , Protease Inhibitors/pharmacology , Protease Inhibitors/chemistry , Humans , Plant Proteins/chemistry , Plant Proteins/pharmacology , Plant Proteins/immunology , Macrophages/drug effects , Macrophages/immunology
6.
ACS Chem Biol ; 19(5): 1116-1124, 2024 May 17.
Article in English | MEDLINE | ID: mdl-38695893

ABSTRACT

Borosins are ribosomally synthesized and post-translationally modified peptides (RiPPs) containing backbone α-N-methylations. These modifications confer favorable pharmacokinetic properties including increased membrane permeability and resistance to proteolytic degradation. Previous studies have biochemically and bioinformatically explored several borosins, revealing (1) numerous domain architectures and (2) diverse core regions lacking conserved sequence elements. Due to these characteristics, large-scale computational identification of borosin biosynthetic genes remains challenging and often requires additional, time-intensive manual inspection. This work builds upon previous findings and updates the genome-mining tool RODEO to automatically evaluate borosin biosynthetic gene clusters (BGCs) and identify putative precursor peptides. Using the new RODEO module, we provide an updated analysis of borosin BGCs identified in the NCBI database. From our data set, we bioinformatically predict and experimentally characterize a new fused borosin domain architecture, in which the modified natural product core is encoded N-terminal to the methyltransferase domain. Additionally, we demonstrate that a borosin precursor peptide is a native substrate of shewasin A, a reported aspartyl peptidase with no previously identified substrates. Shewasin A requires post-translational modification of the leader peptide for proteolytic maturation, a feature not previously observed in RiPPs. Overall, this work provides a user-friendly and open-access tool for the analysis of borosin BGCs and we demonstrate its utility to uncover additional biosynthetic strategies within the borosin class of RiPPs.


Subject(s)
Computational Biology , Protein Processing, Post-Translational , Computational Biology/methods , Multigene Family , Amino Acid Sequence , Peptides/chemistry , Peptides/metabolism
7.
Methods Mol Biol ; 2800: 147-165, 2024.
Article in English | MEDLINE | ID: mdl-38709483

ABSTRACT

Molecular forces are increasingly recognized as an important parameter to understand cellular signaling processes. In the recent years, evidence accumulated that also T-cells exert tensile forces via their T-cell receptor during the antigen recognition process. To measure such intercellular pulling forces, one can make use of the elastic properties of spider silk peptides, which act similar to Hookean springs: increased strain corresponds to increased stress applied to the peptide. Combined with Förster resonance energy transfer (FRET) to read out the strain, such peptides represent powerful and versatile nanoscopic force sensing tools. In this paper, we provide a detailed protocol how to synthesize a molecular force sensor for application in T-cell antigen recognition and hands-on guidelines on experiments and analysis of obtained single molecule FRET data.


Subject(s)
Fluorescence Resonance Energy Transfer , Receptors, Antigen, T-Cell , Receptors, Antigen, T-Cell/metabolism , Receptors, Antigen, T-Cell/immunology , Fluorescence Resonance Energy Transfer/methods , Humans , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Single Molecule Imaging/methods , Animals , Peptides/chemistry , Peptides/immunology , Peptides/metabolism , Silk/chemistry
8.
Food Chem ; 451: 139493, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-38703728

ABSTRACT

Iron chelating peptides have been widely utilized as iron supplements due to their excellent absorption capacity, However, the high cost and cumbersome manufacturing process of these peptides significantly limit their industrial application. In this study, fermentation was used for the first time to prepare iron chelating peptides. Bacillus altitudinis 3*1-3 was selected as the most suitable strain from 50 strains. The hydrolysates of fermented scallop skirts showed excellent iron-chelating capacity (9.39 mg/g). Aspartic acid, glutamic acid, and histidine are crucial for the binding of peptides to ferrous ions. The heptapeptide (FEDPEFE) forms six binding bonds with ferrous irons. Compared with ferrous sulfate, peptide-ferrous chelate showed more stability in salt solution and simulated gastrointestinal juice (p < 0.05). Furthermore, the fermentation method could save >50% of the cost compared with the enzymatic method. The results can provide a theoretical basis for the preparation of ferrous-chelated peptides using the fermentation method.


Subject(s)
Bacillus , Fermentation , Iron Chelating Agents , Pectinidae , Peptides , Animals , Pectinidae/chemistry , Pectinidae/metabolism , Pectinidae/microbiology , Peptides/chemistry , Peptides/metabolism , Iron Chelating Agents/chemistry , Iron Chelating Agents/metabolism , Bacillus/metabolism , Bacillus/chemistry , Iron/chemistry , Iron/metabolism
9.
Bioinformatics ; 40(5)2024 May 02.
Article in English | MEDLINE | ID: mdl-38710496

ABSTRACT

MOTIVATION: Peptide therapeutics hinge on the precise interaction between a tailored peptide and its designated receptor while mitigating interactions with alternate receptors is equally indispensable. Existing methods primarily estimate the binding score between protein and peptide pairs. However, for a specific peptide without a corresponding protein, it is challenging to identify the proteins it could bind due to the sheer number of potential candidates. RESULTS: We propose a transformers-based protein embedding scheme in this study that can quickly identify and rank millions of interacting proteins. Furthermore, the proposed approach outperforms existing sequence- and structure-based methods, with a mean AUC-ROC and AUC-PR of 0.73. AVAILABILITY AND IMPLEMENTATION: Training data, scripts, and fine-tuned parameters are available at https://github.com/RoniGurvich/Peptriever. The proposed method is linked with a web application available for customized prediction at https://peptriever.app/.


Subject(s)
Peptides , Protein Binding , Proteins , Software , Peptides/chemistry , Peptides/metabolism , Proteins/chemistry , Proteins/metabolism , Algorithms , Computational Biology/methods , Databases, Protein
10.
Yakugaku Zasshi ; 144(5): 511-519, 2024.
Article in Japanese | MEDLINE | ID: mdl-38692926

ABSTRACT

Nanoparticles, including liposomes and lipid nanoparticles, have garnered global attention due to their potential applications in pharmaceuticals, vaccines, and gene therapies. These particles enable targeted delivery of new drug modalities such as highly active small molecules and nucleic acids. However, for widespread use of nanoparticle-based formulations, it is crucial to comprehensively analyze their characteristics to ensure both efficacy and safety, as well as enable consistent production. In this context, this review focuses on our research using atomic force microscopy (AFM) to study liposomes and lipid nanoparticles. Our work significantly contributes to the capability of AFM to measure various types of liposomes in an aqueous medium, providing valuable insights into the mechanical properties of these nanoparticles. We discuss the applications of this AFM technique in assessing the quality of nanoparticle-based pharmaceuticals and developing membrane-active peptides.


Subject(s)
Liposomes , Microscopy, Atomic Force , Nanoparticles , Microscopy, Atomic Force/methods , Lipids/chemistry , Drug Delivery Systems , Nanoparticle Drug Delivery System/chemistry , Peptides/chemistry
11.
Protein Sci ; 33(6): e4976, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38757374

ABSTRACT

G-protein coupled receptors (GPCRs) are the largest class of membrane proteins encoded in the human genome with high pharmaceutical relevance and implications to human health. These receptors share a prevalent architecture of seven transmembrane helices followed by an intracellular, amphipathic helix 8 (H8) and a disordered C-terminal tail (Ctail). Technological advancements have led to over 1000 receptor structures in the last two decades, yet frequently H8 and the Ctail are conformationally heterogeneous or altogether absent. Here we synthesize a peptide comprising the neurotensin receptor 1 (NTS1) H8 and Ctail (H8-Ctail) to investigate its structural stability, conformational dynamics, and orientation in the presence of detergent and phospholipid micelles, which mimic the membrane. Circular dichroism (CD) and nuclear magnetic resonance (NMR) measurements confirm that zwitterionic 1,2-diheptanoyl-sn-glycero-3-phosphocholine is a potent stabilizer of H8 structure, whereas the commonly-used branched detergent lauryl maltose neopentyl glycol (LMNG) is unable to completely stabilize the helix - even at amounts four orders of magnitude greater than its critical micellar concentration. We then used NMR spectroscopy to assign the backbone chemical shifts. A series of temperature and lipid titrations were used to define the H8 boundaries as F376-R392 from chemical shift perturbations, changes in resonance intensity, and chemical-shift-derived phi/psi angles. Finally, the H8 azimuthal and tilt angles, defining the helix orientation relative of the membrane normal were measured using paramagnetic relaxation enhancement NMR. Taken together, our studies reveal the H8-Ctail region is sensitive to membrane physicochemical properties and is capable of more adaptive behavior than previously suggested by static structural techniques.


Subject(s)
Receptors, Neurotensin , Receptors, Neurotensin/chemistry , Receptors, Neurotensin/metabolism , Receptors, Neurotensin/genetics , Humans , Micelles , Nuclear Magnetic Resonance, Biomolecular , Peptides/chemistry , Peptides/metabolism , Circular Dichroism , Protein Conformation, alpha-Helical , Detergents/chemistry , Models, Molecular
12.
J Am Chem Soc ; 146(19): 12901-12906, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38701349

ABSTRACT

Cholesterol-rich membranes play a pivotal role in cancer initiation and progression, necessitating innovative approaches to target these membranes for cancer inhibition. Here we report the first case of unnatural peptide (1) assemblies capable of depleting cholesterol and inhibiting cancer cells. Peptide 1 self-assembles into micelles and is rapidly taken up by cancer cells, especially when combined with an acute cholesterol-depleting agent (MßCD). Click chemistry has confirmed that 1 depletes cell membrane cholesterol. It localizes in membrane-rich organelles, including the endoplasmic reticulum, Golgi apparatus, and lysosomes. Furthermore, 1 potently inhibits malignant cancer cells, working synergistically with cholesterol-lowering agents. Control experiments have confirmed that C-terminal capping and unnatural amino acid residues (i.e., BiP) are essential for both cholesterol depletion and potent cancer cell inhibition. This work highlights unnatural peptide assemblies as a promising platform for targeting the cell membrane in controlling cell fates.


Subject(s)
Cholesterol , Peptides , Humans , Cholesterol/chemistry , Cholesterol/metabolism , Peptides/chemistry , Peptides/pharmacology , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Cell Line, Tumor , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Proliferation/drug effects
13.
Nature ; 629(8014): 1062-1068, 2024 May.
Article in English | MEDLINE | ID: mdl-38720082

ABSTRACT

Most chemistry and biology occurs in solution, in which conformational dynamics and complexation underlie behaviour and function. Single-molecule techniques1 are uniquely suited to resolving molecular diversity and new label-free approaches are reshaping the power of single-molecule measurements. A label-free single-molecule method2-16 capable of revealing details of molecular conformation in solution17,18 would allow a new microscopic perspective of unprecedented detail. Here we use the enhanced light-molecule interactions in high-finesse fibre-based Fabry-Pérot microcavities19-21 to detect individual biomolecules as small as 1.2 kDa, a ten-amino-acid peptide, with signal-to-noise ratios (SNRs) >100, even as the molecules are unlabelled and freely diffusing in solution. Our method delivers 2D intensity and temporal profiles, enabling the distinction of subpopulations in mixed samples. Notably, we observe a linear relationship between passage time and molecular radius, unlocking the potential to gather crucial information about diffusion and solution-phase conformation. Furthermore, mixtures of biomolecule isomers of the same molecular weight and composition but different conformation can also be resolved. Detection is based on the creation of a new molecular velocity filter window and a dynamic thermal priming mechanism that make use of the interplay between optical and thermal dynamics22,23 and Pound-Drever-Hall (PDH) cavity locking24 to reveal molecular motion even while suppressing environmental noise. New in vitro ways of revealing molecular conformation, diversity and dynamics can find broad potential for applications in the life and chemical sciences.


Subject(s)
Signal-To-Noise Ratio , Solutions , Diffusion , Single Molecule Imaging/methods , Isomerism , Peptides/chemistry , Light
14.
J Nanobiotechnology ; 22(1): 300, 2024 May 30.
Article in English | MEDLINE | ID: mdl-38816719

ABSTRACT

BACKGROUND: Extracellular vesicles (EVs) derived from human adipose-derived mesenchymal stem cells (hADSCs) have shown great therapeutic potential in plastic and reconstructive surgery. However, the limited production and functional molecule loading of EVs hinder their clinical translation. Traditional two-dimensional culture of hADSCs results in stemness loss and cellular senescence, which is unfavorable for the production and functional molecule loading of EVs. Recent advances in regenerative medicine advocate for the use of three-dimensional culture of hADSCs to produce EVs, as it more accurately simulates their physiological state. Moreover, the successful application of EVs in tissue engineering relies on the targeted delivery of EVs to cells within biomaterial scaffolds. METHODS AND RESULTS: The hADSCs spheroids and hADSCs gelatin methacrylate (GelMA) microspheres are utilized to produce three-dimensional cultured EVs, corresponding to hADSCs spheroids-EVs and hADSCs microspheres-EVs respectively. hADSCs spheroids-EVs demonstrate excellent production and functional molecule loading compared with hADSCs microspheres-EVs. The upregulation of eight miRNAs (i.e. hsa-miR-486-5p, hsa-miR-423-5p, hsa-miR-92a-3p, hsa-miR-122-5p, hsa-miR-223-3p, hsa-miR-320a, hsa-miR-126-3p, and hsa-miR-25-3p) and the downregulation of hsa-miR-146b-5p within hADSCs spheroids-EVs show the potential of improving the fate of remaining ear chondrocytes and promoting cartilage formation probably through integrated regulatory mechanisms. Additionally, a quick and innovative pipeline is developed for isolating chondrocyte homing peptide-modified EVs (CHP-EVs) from three-dimensional dynamic cultures of hADSCs spheroids. CHP-EVs are produced by genetically fusing a CHP at the N-terminus of the exosomal surface protein LAMP2B. The CHP + LAMP2B-transfected hADSCs spheroids were cultured with wave motion to promote the secretion of CHP-EVs. A harvesting method is used to enable the time-dependent collection of CHP-EVs. The pipeline is easy to set up and quick to use for the isolation of CHP-EVs. Compared with nontagged EVs, CHP-EVs penetrate the biomaterial scaffolds and specifically deliver the therapeutic miRNAs to the remaining ear chondrocytes. Functionally, CHP-EVs show a major effect on promoting cell proliferation, reducing cell apoptosis and enhancing cartilage formation in remaining ear chondrocytes in the M1 macrophage-infiltrated microenvironment. CONCLUSIONS: In summary, an innovative pipeline is developed to obtain CHP-EVs from three-dimensional dynamic culture of hADSCs spheroids. This pipeline can be customized to increase EVs production and functional molecule loading, which meets the requirements for regulating remaining ear chondrocyte fate in the M1 macrophage-infiltrated microenvironment.


Subject(s)
Chondrocytes , Extracellular Vesicles , Mesenchymal Stem Cells , Peptides , Spheroids, Cellular , Humans , Chondrocytes/metabolism , Chondrocytes/cytology , Extracellular Vesicles/metabolism , Spheroids, Cellular/metabolism , Spheroids, Cellular/cytology , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Peptides/chemistry , Peptides/metabolism , MicroRNAs/metabolism , MicroRNAs/genetics , Macrophages/metabolism , Macrophages/cytology , Cells, Cultured , Microspheres , Tissue Engineering/methods , Cell Culture Techniques, Three Dimensional/methods , Cellular Microenvironment , Ear Cartilage/metabolism , Adipose Tissue/cytology , Adipose Tissue/metabolism , Cell Differentiation
15.
Methods Enzymol ; 697: 151-180, 2024.
Article in English | MEDLINE | ID: mdl-38816122

ABSTRACT

Many self-assembling peptides can form amyloid like structures with different sizes and morphologies. Driven by non-covalent interactions, their aggregation can occur through distinct pathways. Additionally, they can bind metal ions to create enzyme like active sites that allow them to catalyze diverse reactions. Due to the non-crystalline nature of amyloids, it is quite challenging to elucidate their structures using experimental spectroscopic techniques. In this aspect, molecular dynamics (MD) simulations provide a useful tool to derive structures of these macromolecules in solution. They can be further validated by comparing with experimentally measured structural parameters. However, these simulations require a multi-step process starting from the selection of the initial structure to the analysis of MD trajectories. There are multiple force fields, parametrization protocols, equilibration processes, software and analysis tools available for this process. Therefore, it is complicated for non-experts to select the most relevant tools and perform these simulations effectively. In this chapter, a systematic methodology that covers all major aspects of modeling of catalytic peptides is provided in a user-friendly manner. It will be helpful for researchers in this critical area of research.


Subject(s)
Molecular Dynamics Simulation , Peptides , Peptides/chemistry , Software , Protein Conformation , Catalytic Domain , Catalysis
16.
Methods Enzymol ; 697: 247-268, 2024.
Article in English | MEDLINE | ID: mdl-38816125

ABSTRACT

Drawing inspiration from cellular compartmentalization, enzymatic compartments play a pivotal role in bringing enzymes and substrates into confined environments, offering heightened catalytic efficiency and prolonged enzyme lifespan. Previously, we engineered bioinspired enzymatic compartments, denoted as TPE-Q18H@GPs, achieved through the spatiotemporally controllable self-assembly of the catalytic peptide TPE-Q18H within hollow porous glucan particles (GPs). This design strategy allows substrates and products to freely traverse, while retaining enzymatic aggregations. The confined environment led to the formation of catalytic nanofibers, resulting in enhanced substrate binding affinity and a more than two-fold increase in the second-order kinetic constant (kcat/Km) compared to TPE-Q18H nanofibers in a dispersed system. In this work, we will introduce how to synthesize the above-mentioned enzymatic compartments using salt-responsive catalytic peptides and GPs.


Subject(s)
Glucans , Peptides , Glucans/chemistry , Peptides/chemistry , Nanofibers/chemistry , Kinetics , Porosity , Biocatalysis
17.
Methods Enzymol ; 697: 321-343, 2024.
Article in English | MEDLINE | ID: mdl-38816128

ABSTRACT

Peptides that self-assemble exhibit distinct three-dimensional structures and attributes, positioning them as promising candidates for biocatalysts. Exploring their catalytic processes enhances our comprehension of the catalytic actions inherent to self-assembling peptides, laying a theoretical foundation for creating novel biocatalysts. The investigation into the intricate reaction mechanisms of these entities is rendered challenging due to the vast variability in peptide sequences, their aggregated formations, supportive elements, structures of active sites, types of catalytic reactions, and the interplay between these variables. This complexity hampers the elucidation of the linkage between sequence, structure, and catalytic efficiency in self-assembling peptide catalysts. This chapter delves into the latest progress in understanding the mechanisms behind peptide self-assembly, serving as a catalyst in hydrolysis and oxidation reactions, and employing computational analyses. It discusses the establishment of models, selection of computational strategies, and analysis of computational procedures, emphasizing the application of modeling techniques in probing the catalytic mechanisms of peptide self-assemblies. It also looks ahead to the potential future trajectories within this research domain. Despite facing numerous obstacles, a thorough investigation into the structural and catalytic mechanisms of peptide self-assemblies, combined with the ongoing advancement in computational simulations and experimental methodologies, is set to offer valuable theoretical insights for the development of new biocatalysts, thereby significantly advancing the biocatalysis field.


Subject(s)
Biocatalysis , Peptides , Peptides/chemistry , Hydrolysis , Oxidation-Reduction , Catalytic Domain , Molecular Dynamics Simulation , Catalysis , Models, Molecular
18.
Methods Enzymol ; 697: 15-33, 2024.
Article in English | MEDLINE | ID: mdl-38816121

ABSTRACT

Once considered a thermodynamic minimum of the protein fold or as simply by-products of a misfolding process, amyloids are increasingly showing remarkable potential for promoting enzyme-like catalysis. Recent studies have demonstrated a diverse range of catalytic behaviors that amyloids can promote way beyond the hydrolytic behaviors originally reported. We and others have demonstrated the strong propensity of catalytic amyloids to facilitate redox reactions both in the presence and in the absence of metal cofactors. Here, we present a detailed protocol for measuring the oxidative ability of supramolecular peptide assemblies.


Subject(s)
Amyloid , Oxidation-Reduction , Amyloid/chemistry , Amyloid/metabolism , Humans , Catalysis , Peptides/chemistry , Peptides/metabolism , Protein Folding
19.
Methods Enzymol ; 697: 211-245, 2024.
Article in English | MEDLINE | ID: mdl-38816124

ABSTRACT

Among the important questions in supramolecular peptide self-assemblies are their interactions with metallic compounds and ions. In the last decade, intensive efforts have been devoted to understanding the structural properties of these interactions including their dynamical and catalytic impact in natural and de novo systems. Since structural insights from experimental approaches could be particularly challenging, computational chemistry methods are interesting complementary tools. Here, we present the general multiscale strategies we developed and applied for the study of metallopeptide assemblies. These strategies include prediction of metal binding site, docking of metallic moieties, classical and accelerated molecular dynamics and finally QM/MM calculations. The systems of choice for this chapter are, on one side, peptides involved in neurodegenerative diseases and, on the other, de novo fibrillar systems with catalytic properties. Both successes and remaining challenges are highlighted so that the protocol could be apply to other system of this kind.


Subject(s)
Metalloproteins , Molecular Dynamics Simulation , Peptides , Peptides/chemistry , Metalloproteins/chemistry , Binding Sites , Humans , Molecular Docking Simulation/methods , Metals/chemistry , Quantum Theory
20.
Methods Enzymol ; 697: 293-319, 2024.
Article in English | MEDLINE | ID: mdl-38816127

ABSTRACT

Assembly of de novo peptides designed from scratch is in a semi-rational manner and creates artificial supramolecular structures with unique properties. Considering that the functions of various proteins in living cells are highly regulated by their assemblies, building artificial assemblies within cells holds the potential to simulate the functions of natural protein assemblies and engineer cellular activities for controlled manipulation. How can we evaluate the self-assembly of designed peptides in cells? The most effective approach involves the genetic fusion of fluorescent proteins (FPs). Expressing a self-assembling peptide fused with an FP within cells allows for evaluating assemblies through fluorescence signal. When µm-scale assemblies such as condensates are formed, the peptide assemblies can be directly observed by imaging. For sub-µm-scale assemblies, fluorescence correlation spectroscopy analysis is more practical. Additionally, the fluorescence resonance energy transfer (FRET) signal between FPs is valuable evidence of proximity. The decrease in fluorescence anisotropy associated with homo-FRET reveals the properties of self-assembly. Furthermore, by combining two FPs, one acting as a donor and the other as an acceptor, the heteromeric interaction between two different components can be studied through the FRET signal. In this chapter, we provide detailed protocols, from designing and constructing plasmid DNA expressing the peptide-fused protein to analysis of self-assembly in living cells.


Subject(s)
Fluorescence Resonance Energy Transfer , Luminescent Proteins , Peptides , Recombinant Fusion Proteins , Fluorescence Resonance Energy Transfer/methods , Peptides/chemistry , Peptides/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/chemistry , Humans , Luminescent Proteins/genetics , Luminescent Proteins/chemistry , Luminescent Proteins/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Green Fluorescent Proteins/chemistry , Plasmids/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...