Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 466
Filtrer
1.
Int J Biol Macromol ; : 135997, 2024 Sep 27.
Article de Anglais | MEDLINE | ID: mdl-39343253

RÉSUMÉ

This study examined two oleosins of 17 kDa and 15 kDa isolated from Yuzhi white sesame seeds through oil body extraction. The allergens were identified as oleosin H1 (Ses i 4) and oleosin L (Ses i 5) using SDS-PAGE, dot blot analysis, and LC-MS/MS. PCR analysis revealed high sequence homology for the oleosin proteins in the sesame seeds. Utilizing AlphaFold2, bioinformatics tools, and protein-protein docking, the structure and function of these oleosins were analyzed. Ten potential B cell epitope peptides were predicted and mapped onto the α-helix and random coil-dominated oleosome membrane conformation. IgE binding simulations identified key epitopes, B3 (FLTSGAFGL) and B4 (RGVQEGTLY) for oleosin H1, and B8 (GGFGVAALSV) and B9 (DQLESAKTKL) for oleosin L. Mutational analysis highlighted Glu135, Phe102, Tyr128, Tyr139, Gly136, and Gly132 in oleosin H1, and Leu120, Lys119, and Leu113 in oleosin L as critical residues for binding stability, providing insights into the sensitization mechanism of these epitopes. The integration of bioinformatics and immunoinformatics in this study has contributed to a deeper understanding of the allergy properties of sesame oleosins.

2.
Mol Pharm ; 2024 Sep 09.
Article de Anglais | MEDLINE | ID: mdl-39251364

RÉSUMÉ

Parkinson's disease (PD) is an idiopathic neurodegenerative disorder with the second-highest prevalence rate behind Alzheimer's disease. The pathophysiological hallmarks of PD are both degeneration of dopaminergic neurons in the substantia nigra pars compacta and the inclusion of misfolded α-synuclein (α-syn) aggregates known as Lewy bodies. Despite decades of research for potential PD treatments, none have been developed, and developing new therapeutic agents is a time-consuming and expensive process. Computational methods can be used to investigate the properties of drug candidates currently undergoing clinical trials to determine their theoretical efficiency at targeting α-syn. Monoclonal antibodies (mAbs) are biological drugs with high specificity, and Prasinezumab (PRX002) is an mAb currently in Phase II, which targets the C-terminus (AA 118-126) of α-syn. We utilized BioLuminate and PyMol for the structure prediction and preparation of the fragment antigen-binding (Fab) region of PRX002 and 34 different conformations of α-syn. Protein-protein docking simulations were performed using PIPER, and 3 of the docking poses were selected based on the best fit. Molecular dynamics simulations were conducted on the docked protein structures in triplicate for 1000 ns, and hydrogen bonds and electrostatic and hydrophobic interactions were analyzed using MDAnalysis to determine which residues were interacting and how often. Hydrogen bonds were shown to form frequently between the HCDR2 region of PRX002 and α-syn. Free energy was calculated to determine the binding affinity. The predicted binding affinity shows a strong antibody-antigen attraction between PRX002 and α-syn. RMSD was calculated to determine the conformational change of these regions throughout the simulation. The mAb's developability was determined using computational screening methods. Our results demonstrate the efficiency and developability of this therapeutic agent.

4.
Heliyon ; 10(16): e35943, 2024 Aug 30.
Article de Anglais | MEDLINE | ID: mdl-39229544

RÉSUMÉ

Memory loss is becoming an increasingly significant health problem, largely due to Alzheimer's disease (AD), which disrupts the brain in several ways, including causing inflammation and weakening the body's defenses. This study explores the potential of medicinal plants as a source of novel therapeutic agents for AD. First, we tested various plant extracts against acetylcholinesterase (AChE) in vitro, following molecular docking simulations with key AD-related protein targets such as MAO-B, P-gp, GSK-3ß, and CD14. Rosemary extract was found to be the most inhibitory towards AChE. The compounds found in rosemary (oleanolic acid), sage (pinocembrin), and cinnamon (italicene) showed promise in potentially binding to MAO-B. These chemicals may interact with a key protein in the brain and alter the production and removal of amyloid-ß. Luteolin (from rosemary), myricetin (from sage), chamigrene, and italicene (from cinnamon) exhibited potential for inhibiting tau aggregation. Additionally, ursolic acid found in rosemary, sage, and chamigrene from cinnamon could modulate CD14 activity. For the first time, our findings shed light on the intricate interplay between neuroinflammation, neuroprotective mechanisms, and the immune system's role in AD. Further research is needed to validate the in vivo efficacy and safety of these plant-derived compounds, as well as their interactions with key protein targets, which could lead to the development of novel AD therapeutics.

5.
J Mol Biol ; 436(17): 168540, 2024 Sep 01.
Article de Anglais | MEDLINE | ID: mdl-39237205

RÉSUMÉ

Protein interactions are essential for cellular processes. In recent years there has been significant progress in computational prediction of 3D structures of individual protein chains, with the best-performing algorithms reaching sub-Ångström accuracy. These techniques are now finding their way into the prediction of protein interactions, adding to the existing modeling approaches. The community-wide Critical Assessment of Predicted Interactions (CAPRI) has been a catalyst for the development of procedures for the structural modeling of protein assemblies by organizing blind prediction experiments. The predicted structures are assessed against unpublished experimentally determined structures using a set of metrics with proven robustness that have been established in the CAPRI community. In addition, several advanced benchmarking databases provide targets against which users can test docking and assembly modeling software. These include the Protein-Protein Docking Benchmark, the CAPRI Scoreset, and the Dockground database, all developed by members of the CAPRI community. Here we present CAPRI-Q, a stand-alone model quality assessment tool, which can be freely downloaded or used via a publicly available web server. This tool applies the CAPRI metrics to assess the quality of query structures against given target structures, along with other popular quality metrics such as DockQ, TM-score and l-DDT, and classifies the models according to the CAPRI model quality criteria. The tool can handle a variety of protein complex types including those involving peptides, nucleic acids, and oligosaccharides. The source code is freely available from https://gitlab.in2p3.fr/cmsb-public/CAPRI-Q and its web interface through the Dockground resource at https://dockground.compbio.ku.edu/assessment/.


Sujet(s)
Bases de données de protéines , Conformation des protéines , Protéines , Logiciel , Protéines/composition chimique , Modèles moléculaires , Biologie informatique/méthodes , Simulation de docking moléculaire , Algorithmes , Cartographie d'interactions entre protéines/méthodes , Liaison aux protéines
6.
Bioinformatics ; 2024 Sep 30.
Article de Anglais | MEDLINE | ID: mdl-39348157

RÉSUMÉ

MOTIVATION: Antibody-antigen complex modelling is an important step in computational workflows for therapeutic antibody design. While experimentally determined structures of both antibody and the cognate antigen are often not available, recent advances in machine learning-driven protein modelling have enabled accurate prediction of both antibody and antigen structures. Here, we analyse the ability of protein-protein docking tools to use machine learning generated input structures for information-driven docking. RESULTS: In an information-driven scenario, we find that HADDOCK can generate accurate models of antibody-antigen complexes using an ensemble of antibody structures generated by machine learning tools and AlphaFold2 predicted antigen structures. Targeted docking using knowledge of the complementary determining regions on the antibody and some information about the targeted epitope allows the generation of high quality models of the complex with reduced sampling, resulting in a computationally cheap protocol that outperforms the ZDOCK baseline. AVAILABILITY: The source code of HADDOCK3 is freely available at github.com/haddocking/haddock3. The code to generate and analyse the data is available at github.com/haddocking/ai-antibodies. The full runs, including docking models from all modules of a workflow have been deposited in our lab collection (data.sbgrid.org/labs/32/1139) at the SBGRID data repository. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.

7.
Biochimie ; 2024 Aug 10.
Article de Anglais | MEDLINE | ID: mdl-39134296

RÉSUMÉ

Mycoses infect millions of people annually across the world. The most common mycosis agent, Candida albicans is responsible for a great deal of illness and death. C. albicans infection is becoming more widespread and the current antifungals polyenes, triazoles, and echinocandins are less efficient against it. Investigating antifungal peptides (AFPs) as therapeutic is gaining momentum. Therefore, we used MALDI-TOF/MS analysis to identify AFPs and protein-protein docking to analyze their interactions with the C. albicans target protein. Some microorganisms with strong antifungal action against C. albicans were selected for the isolation of AFPs. Using MALDI-TOF/MS, we identified 3 AFPs Chitin binding protein (ACW83017.1; Bacillus licheniformis), the bifunctional protein GlmU (BBQ13478.1; Stenotrophomonas maltophilia), and zinc metalloproteinase aureolysin (BBA25172.1; Staphylococcus aureus). These AFPs showed robust interactions with C. albicans target protein Sap5. We deciphered some important residues in identified APFs and highlighted interaction with Sap5 through hydrogen bonds, protein-protein interactions, and salt bridges using protein-protein docking and MD simulations. The three discovered AFPs-Sap5 complexes exhibit different levels of stability, as seen by the RMSD analysis and interaction patterns. Among protein-protein interactions, the remarkable stability of the BBQ25172.1-2QZX complex highlights the role of salt bridges and hydrogen bonds. Identified AFPs could be further studied for developing successful antifungal candidates and peptide-based new antifungal therapeutic strategies as fresh insights into addressing antifungal resistance also.

8.
J Biol Chem ; 300(8): 107575, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39013537

RÉSUMÉ

Adaptation to the shortage in free amino acids (AA) is mediated by 2 pathways, the integrated stress response (ISR) and the mechanistic target of rapamycin (mTOR). In response to reduced levels, primarily of leucine or arginine, mTOR in its complex 1 configuration (mTORC1) is suppressed leading to a decrease in translation initiation and elongation. The eIF2α kinase general control nonderepressible 2 (GCN2) is activated by uncharged tRNAs, leading to induction of the ISR in response to a broader range of AA shortage. ISR confers a reduced translation initiation, while promoting the selective synthesis of stress proteins, such as ATF4. To efficiently adapt to AA starvation, the 2 pathways are cross-regulated at multiple levels. Here we identified a new mechanism of ISR/mTORC1 crosstalk that optimizes survival under AA starvation, when mTORC1 is forced to remain active. mTORC1 activation during acute AA shortage, augmented ATF4 expression in a GCN2-dependent manner. Under these conditions, enhanced GCN2 activity was not dependent on tRNA sensing, inferring a different activation mechanism. We identified a labile physical interaction between GCN2 and mTOR that results in a phosphorylation of GCN2 on serine 230 by mTOR, which promotes GCN2 activity. When examined under prolonged AA starvation, GCN2 phosphorylation by mTOR promoted survival. Our data unveils an adaptive mechanism to AA starvation, when mTORC1 evades inhibition.


Sujet(s)
Facteur de transcription ATF-4 , Complexe-1 cible mécanistique de la rapamycine , Protein-Serine-Threonine Kinases , Stress physiologique , Sérine-thréonine kinases TOR , Sérine-thréonine kinases TOR/métabolisme , Phosphorylation , Facteur de transcription ATF-4/métabolisme , Facteur de transcription ATF-4/génétique , Complexe-1 cible mécanistique de la rapamycine/métabolisme , Protein-Serine-Threonine Kinases/métabolisme , Protein-Serine-Threonine Kinases/génétique , Humains , Animaux , Souris , Acides aminés/métabolisme , Adaptation physiologique , Complexes multiprotéiques/métabolisme , Complexes multiprotéiques/génétique , ARN de transfert/métabolisme , ARN de transfert/génétique , Cellules HEK293
9.
Methods Mol Biol ; 2780: 45-68, 2024.
Article de Anglais | MEDLINE | ID: mdl-38987463

RÉSUMÉ

Proteins are the fundamental organic macromolecules in living systems that play a key role in a variety of biological functions including immunological detection, intracellular trafficking, and signal transduction. The docking of proteins has greatly advanced during recent decades and has become a crucial complement to experimental methods. Protein-protein docking is a helpful method for simulating protein complexes whose structures have not yet been solved experimentally. This chapter focuses on major search tactics along with various docking programs used in protein-protein docking algorithms, which include: direct search, exhaustive global search, local shape feature matching, randomized search, and broad category of post-docking approaches. As backbone flexibility predictions and interactions in high-resolution protein-protein docking remain important issues in the overall optimization context, we have put forward several methods and solutions used to handle backbone flexibility. In addition, various docking methods that are utilized for flexible backbone docking, including ATTRACT, FlexDock, FLIPDock, HADDOCK, RosettaDock, FiberDock, etc., along with their scoring functions, algorithms, advantages, and limitations are discussed. Moreover, what progress in search technology is expected, including not only the creation of new search algorithms but also the enhancement of existing ones, has been debated. As conformational flexibility is one of the most crucial factors affecting docking success, more work should be put into evaluating the conformational flexibility upon binding for a particular case in addition to developing new algorithms to replace the rigid body docking and scoring approach.


Sujet(s)
Algorithmes , Simulation de docking moléculaire , Liaison aux protéines , Protéines , Simulation de docking moléculaire/méthodes , Protéines/composition chimique , Protéines/métabolisme , Logiciel , Conformation des protéines , Biologie informatique/méthodes , Bases de données de protéines , Cartographie d'interactions entre protéines/méthodes
10.
Methods Mol Biol ; 2780: 15-26, 2024.
Article de Anglais | MEDLINE | ID: mdl-38987461

RÉSUMÉ

Protein-protein docking is considered one of the most important techniques supporting experimental proteomics. Recent developments in the field of computer science helped to improve this computational technique so that it better handles the complexity of protein nature. Sampling algorithms are responsible for the generation of numerous protein-protein ensembles. Unfortunately, a primary docking output comprises a set of both near-native poses and decoys. Application of the efficient scoring function helps to differentiate poses with the most favorable properties from those that are very unlikely to represent a natural state of the complex. This chapter explains the importance of sampling and scoring in the process of protein-protein docking. Moreover, it summarizes advances in the field.


Sujet(s)
Algorithmes , Simulation de docking moléculaire , Liaison aux protéines , Protéines , Simulation de docking moléculaire/méthodes , Protéines/composition chimique , Protéines/métabolisme , Biologie informatique/méthodes , Conformation des protéines , Cartographie d'interactions entre protéines/méthodes , Logiciel , Protéomique/méthodes
11.
Methods Mol Biol ; 2780: 69-89, 2024.
Article de Anglais | MEDLINE | ID: mdl-38987464

RÉSUMÉ

Molecular docking is used to anticipate the optimal orientation of a particular molecule to a target to form a stable complex. It makes predictions about the 3D structure of any complex based on the binding characteristics of the ligand and the target receptor usually a protein. It is an exceptionally useful tool, which is used as a model to study how ligands attach to proteins. Docking can also be used for studying the interaction of ligands and proteins to analyze inhibitory efficacy. The ligand may also be a protein, making it possible to study interactions between two different proteins using the numerous docking tools available for basic research on protein interactions. The protein-protein docking is a crucial approach to understanding the protein interactions and predicting the structure of protein complexes that have not yet been experimentally determined. Moreover, the protein-protein interactions can predict the function of target proteins and the drug-like properties of molecules. Therefore, protein docking assists in uncovering insights into protein interactions and also aids in a better understanding of molecular pathways/mechanisms. This chapter comprehends the various tools for protein-protein docking (pairwise and multiple), including their methodologies and analysis of output as results.


Sujet(s)
Simulation de docking moléculaire , Liaison aux protéines , Cartographie d'interactions entre protéines , Protéines , Protéines/composition chimique , Protéines/métabolisme , Ligands , Cartographie d'interactions entre protéines/méthodes , Logiciel , Biologie informatique/méthodes , Conformation des protéines , Sites de fixation , Bases de données de protéines
12.
Methods Mol Biol ; 2780: 3-14, 2024.
Article de Anglais | MEDLINE | ID: mdl-38987460

RÉSUMÉ

Despite the development of methods for the experimental determination of protein structures, the dissonance between the number of known sequences and their solved structures is still enormous. This is particularly evident in protein-protein complexes. To fill this gap, diverse technologies have been developed to study protein-protein interactions (PPIs) in a cellular context including a range of biological and computational methods. The latter derive from techniques originally published and applied almost half a century ago and are based on interdisciplinary knowledge from the nexus of the fields of biology, chemistry, and physics about protein sequences, structures, and their folding. Protein-protein docking, the main protagonist of this chapter, is routinely treated as an integral part of protein research. Herein, we describe the basic foundations of the whole process in general terms, but step by step from protein representations through docking methods and evaluation of complexes to their final validation.


Sujet(s)
Simulation de docking moléculaire , Liaison aux protéines , Protéines , Simulation de docking moléculaire/méthodes , Protéines/composition chimique , Protéines/métabolisme , Logiciel , Cartographie d'interactions entre protéines/méthodes , Conformation des protéines , Biologie informatique/méthodes
13.
Methods Mol Biol ; 2780: 129-138, 2024.
Article de Anglais | MEDLINE | ID: mdl-38987467

RÉSUMÉ

Protein-protein interactions (PPIs) provide valuable insights for understanding the principles of biological systems and for elucidating causes of incurable diseases. One of the techniques used for computational prediction of PPIs is protein-protein docking calculations, and a variety of software has been developed. This chapter is a summary of software and databases used for protein-protein docking.


Sujet(s)
Bases de données de protéines , Simulation de docking moléculaire , Cartographie d'interactions entre protéines , Protéines , Logiciel , Cartographie d'interactions entre protéines/méthodes , Protéines/composition chimique , Protéines/métabolisme , Biologie informatique/méthodes , Liaison aux protéines , Humains
14.
Methods Mol Biol ; 2780: 107-126, 2024.
Article de Anglais | MEDLINE | ID: mdl-38987466

RÉSUMÉ

An exponential increase in the number of publications that address artificial intelligence (AI) usage in life sciences has been noticed in recent years, while new modeling techniques are constantly being reported. The potential of these methods is vast-from understanding fundamental cellular processes to discovering new drugs and breakthrough therapies. Computational studies of protein-protein interactions, crucial for understanding the operation of biological systems, are no exception in this field. However, despite the rapid development of technology and the progress in developing new approaches, many aspects remain challenging to solve, such as predicting conformational changes in proteins, or more "trivial" issues as high-quality data in huge quantities.Therefore, this chapter focuses on a short introduction to various AI approaches to study protein-protein interactions, followed by a description of the most up-to-date algorithms and programs used for this purpose. Yet, given the considerable pace of development in this hot area of computational science, at the time you read this chapter, the development of the algorithms described, or the emergence of new (and better) ones should come as no surprise.


Sujet(s)
Algorithmes , Biologie informatique , Apprentissage machine , Simulation de docking moléculaire , Protéines , Protéines/composition chimique , Protéines/métabolisme , Simulation de docking moléculaire/méthodes , Biologie informatique/méthodes , Liaison aux protéines , Cartographie d'interactions entre protéines/méthodes , Humains , Conformation des protéines , Logiciel
15.
Methods Mol Biol ; 2780: 91-106, 2024.
Article de Anglais | MEDLINE | ID: mdl-38987465

RÉSUMÉ

Concerted interactions between all the cell components form the basis of biological processes. Protein-protein interactions (PPIs) constitute a tremendous part of this interaction network. Deeper insight into PPIs can help us better understand numerous diseases and lead to the development of new diagnostic and therapeutic strategies. PPI interfaces, until recently, were considered undruggable. However, it is now believed that the interfaces contain "hot spots," which could be targeted by small molecules. Such a strategy would require high-quality structural data of PPIs, which are difficult to obtain experimentally. Therefore, in silico modeling can complement or be an alternative to in vitro approaches. There are several computational methods for analyzing the structural data of the binding partners and modeling of the protein-protein dimer/oligomer structure. The major problem with in silico structure prediction of protein assemblies is obtaining sufficient sampling of protein dynamics. One of the methods that can take protein flexibility and the effects of the environment into account is Molecular Dynamics (MD). While sampling of the whole protein-protein association process with plain MD would be computationally expensive, there are several strategies to harness the method to PPI studies while maintaining reasonable use of resources. This chapter reviews known applications of MD in the PPI investigation workflows.


Sujet(s)
Simulation de docking moléculaire , Simulation de dynamique moléculaire , Liaison aux protéines , Protéines , Simulation de docking moléculaire/méthodes , Protéines/composition chimique , Protéines/métabolisme , Cartographie d'interactions entre protéines/méthodes , Conformation des protéines , Humains , Logiciel , Biologie informatique/méthodes
16.
Methods Mol Biol ; 2780: 203-255, 2024.
Article de Anglais | MEDLINE | ID: mdl-38987471

RÉSUMÉ

Despite the recent advances in the determination of high-resolution membrane protein (MP) structures, the structural and functional characterization of MPs remains extremely challenging, mainly due to the hydrophobic nature, low abundance, poor expression, purification, and crystallization difficulties associated with MPs. Whereby the major challenges/hurdles for MP structure determination are associated with the expression, purification, and crystallization procedures. Although there have been significant advances in the experimental determination of MP structures, only a limited number of MP structures (approximately less than 1% of all) are available in the Protein Data Bank (PDB). Therefore, the structures of a large number of MPs still remain unresolved, which leads to the availability of widely unplumbed structural and functional information related to MPs. As a result, recent developments in the drug discovery realm and the significant biological contemplation have led to the development of several novel, low-cost, and time-efficient computational methods that overcome the limitations of experimental approaches, supplement experiments, and provide alternatives for the characterization of MPs. Whereby the fine tuning and optimizations of these computational approaches remains an ongoing endeavor.Computational methods offer a potential way for the elucidation of structural features and the augmentation of currently available MP information. However, the use of computational modeling can be extremely challenging for MPs mainly due to insufficient knowledge of (or gaps in) atomic structures of MPs. Despite the availability of numerous in silico methods for 3D structure determination the applicability of these methods to MPs remains relatively low since all methods are not well-suited or adequate for MPs. However, sophisticated methods for MP structure predictions are constantly being developed and updated to integrate the modifications required for MPs. Currently, different computational methods for (1) MP structure prediction, (2) stability analysis of MPs through molecular dynamics simulations, (3) modeling of MP complexes through docking, (4) prediction of interactions between MPs, and (5) MP interactions with its soluble partner are extensively used. Towards this end, MP docking is widely used. It is notable that the MP docking methods yet few in number might show greater potential in terms of filling the knowledge gap. In this chapter, MP docking methods and associated challenges have been reviewed to improve the applicability, accuracy, and the ability to model macromolecular complexes.


Sujet(s)
Bases de données de protéines , Protéines membranaires , Simulation de docking moléculaire , Protéines membranaires/composition chimique , Protéines membranaires/métabolisme , Simulation de docking moléculaire/méthodes , Liaison aux protéines , Conformation des protéines , Biologie informatique/méthodes
17.
Methods Mol Biol ; 2780: 327-343, 2024.
Article de Anglais | MEDLINE | ID: mdl-38987476

RÉSUMÉ

The chapter emphasizes the importance of understanding protein-protein interactions in cellular mechanisms and highlights the role of computational modeling in predicting these interactions. It discusses sequence-based approaches such as evolutionary trace (ET), correlated mutation analysis (CMA), and subtractive correlated mutation (SCM) for identifying crucial amino acid residues, considering interface conservation or evolutionary changes. The chapter also explores methods like differential ET, hidden-site class model, and spatial cluster detection (SCD) for interface specificity and spatial clustering. Furthermore, it examines approaches combining structural and sequential methodologies and evaluates modeled predictions through initiatives like critical assessment of prediction of interactions (CAPRI). Additionally, the chapter provides an overview of various software programs used for molecular docking, detailing their search, sampling, refinement and scoring stages, along with innovative techniques and tools like normal mode analysis (NMA) and adaptive Poisson-Boltzmann solver (APBS) for electrostatic calculations. These computational and experimental approaches are crucial for unraveling protein-protein interactions and aid in developing potential therapeutics for various diseases.


Sujet(s)
Biologie informatique , Simulation de docking moléculaire , Liaison aux protéines , Protéines , Logiciel , Biologie informatique/méthodes , Protéines/métabolisme , Protéines/composition chimique , Cartographie d'interactions entre protéines/méthodes , Humains , Mutation , Algorithmes , Conformation des protéines
18.
Methods Mol Biol ; 2780: 289-302, 2024.
Article de Anglais | MEDLINE | ID: mdl-38987474

RÉSUMÉ

Accurate prediction and evaluation of protein-protein complex structures is of major importance to understand the cellular interactome. Predicted complex structures based on deep learning approaches or traditional docking methods require often structural refinement and rescoring for realistic evaluation. Standard molecular dynamics (MD) simulations are time-consuming and often do not structurally improve docking solutions. Better refinement can be achieved with our recently developed replica-exchange-based scheme employing different levels of repulsive biasing between proteins in each replica simulation (RS-REMD). The bias acts specifically on the intermolecular interactions based on an increase in effective pairwise van der Waals radii without changing interactions within each protein or with the solvent. It allows for an improvement of the predicted protein-protein complex structure and simultaneous realistic free energy scoring of protein-protein complexes. The setup of RS-REMD simulations is described in detail including the application on two examples (all necessary scripts and input files can be obtained from https://gitlab.com/TillCyrill/mmib ).


Sujet(s)
Simulation de docking moléculaire , Simulation de dynamique moléculaire , Protéines , Protéines/composition chimique , Simulation de docking moléculaire/méthodes , Liaison aux protéines , Logiciel , Conformation des protéines , Biologie informatique/méthodes
19.
Methods Mol Biol ; 2780: 281-287, 2024.
Article de Anglais | MEDLINE | ID: mdl-38987473

RÉSUMÉ

G-protein-coupled receptors (GPCRs), the largest family of human membrane proteins, play a crucial role in cellular control and are the target of approximately one-third of all drugs on the market. Targeting these complexes with selectivity or formulating small molecules capable of modulating receptor-receptor interactions could potentially offer novel avenues for drug discovery, fostering the development of more refined and safer pharmacotherapies. Due to the lack of experimentally derived X-ray crystallography spectra of GPCR oligomers, there is growing evidence supporting the development of new in silico approaches for predicting GPCR self-assembling structures. The significance of GPCR oligomerization, the challenges in modeling these structures, and the potential of protein-protein docking algorithms to address these challenges are discussed. The study also underscores the use of various software solutions for modeling GPCR oligomeric structures and presents practical cases where these techniques have been successfully applied.


Sujet(s)
Simulation de docking moléculaire , Multimérisation de protéines , Récepteurs couplés aux protéines G , Logiciel , Récepteurs couplés aux protéines G/composition chimique , Récepteurs couplés aux protéines G/métabolisme , Simulation de docking moléculaire/méthodes , Humains , Liaison aux protéines , Algorithmes , Cristallographie aux rayons X/méthodes , Conformation des protéines , Modèles moléculaires
20.
bioRxiv ; 2024 Jul 13.
Article de Anglais | MEDLINE | ID: mdl-39026849

RÉSUMÉ

The oligomerization of protein macromolecules on cell membranes plays a fundamental role in regulating cellular function. From modulating signal transduction to directing immune response, membrane proteins (MPs) play a crucial role in biological processes and are often the target of many pharmaceutical drugs. Despite their biological relevance, the challenges in experimental determination have hampered the structural availability of membrane proteins and their complexes. Computational docking provides a promising alternative to model membrane protein complex structures. Here, we present Rosetta-MPDock, a flexible transmembrane (TM) protein docking protocol that captures binding-induced conformational changes. Rosetta-MPDock samples large conformational ensembles of flexible monomers and docks them within an implicit membrane environment. We benchmarked this method on 29 TM-protein complexes of variable backbone flexibility. These complexes are classified based on the root-mean-square deviation between the unbound and bound states (RMSDUB) as: rigid (RMSDUB <1.2 Å), moderately-flexible (RMSDUB ∈ [1.2, 2.2) Å), and flexible targets (RMSDUB > 2.2 Å). In a local docking scenario, i.e. with membrane protein partners starting ≈10 Å apart embedded in the membrane in their unbound conformations, Rosetta-MPDock successfully predicts the correct interface (success defined as achieving 3 near-native structures in the 5 top-ranked models) for 67% moderately flexible targets and 60% of the highly flexible targets, a substantial improvement from the existing membrane protein docking methods. Further, by integrating AlphaFold2-multimer for structure determination and using Rosetta-MPDock for docking and refinement, we demonstrate improved success rates over the benchmark targets from 64% to 73%. Rosetta-MPDock advances the capabilities for membrane protein complex structure prediction and modeling to tackle key biological questions and elucidate functional mechanisms in the membrane environment. The benchmark set and the code is available for public use at github.com/Graylab/MPDock.

SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE