Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Nucleic Acids Res ; 52(D1): D413-D418, 2024 Jan 05.
Article in English | MEDLINE | ID: mdl-37956324

ABSTRACT

ChannelsDB 2.0 is an updated database providing structural information about the position, geometry and physicochemical properties of protein channels-tunnels and pores-within deposited biomacromolecular structures from PDB and AlphaFoldDB databases. The newly deposited information originated from several sources. Firstly, we included data calculated using a popular CAVER tool to complement the data obtained using original MOLE tool for detection and analysis of protein tunnels and pores. Secondly, we added tunnels starting from cofactors within the AlphaFill database to enlarge the scope of the database to protein models based on Uniprot. This has enlarged available channel annotations ∼4.6 times as of 1 September 2023. The database stores information about geometrical features, e.g. length and radius, and physico-chemical properties based on channel-lining amino acids. The stored data are interlinked with the available UniProt mutation annotation data. ChannelsDB 2.0 provides an excellent resource for deep analysis of the role of biomacromolecular tunnels and pores. The database is available free of charge: https://channelsdb2.biodata.ceitec.cz.


Subject(s)
Databases, Protein , Proteins , Software , Amino Acids , Proteins/chemistry , Protein Conformation
2.
Bioinformatics ; 39(8)2023 08 01.
Article in English | MEDLINE | ID: mdl-37471591

ABSTRACT

SUMMARY: Access pathways in enzymes are crucial for the passage of substrates and products of catalysed reactions. The process can be studied by computational means with variable degrees of precision. Our in-house approximative method CaverDock provides a fast and easy way to set up and run ligand binding and unbinding calculations through protein tunnels and channels. Here we introduce pyCaverDock, a Python3 API designed to improve user experience with the tool and further facilitate the ligand transport analyses. The API enables users to simplify the steps needed to use CaverDock, from automatizing setup processes to designing screening pipelines. AVAILABILITY AND IMPLEMENTATION: pyCaverDock API is implemented in Python 3 and is freely available with detailed documentation and practical examples at https://loschmidt.chemi.muni.cz/caverdock/.


Subject(s)
Proteins , Software , Ligands
3.
Biotechnol Adv ; 60: 108009, 2022 11.
Article in English | MEDLINE | ID: mdl-35738509

ABSTRACT

Acceleration of chemical reactions by the enzymes optimized using protein engineering represents one of the key pillars of the contribution of biotechnology towards sustainability. Tunnels and channels of enzymes with buried active sites enable the exchange of ligands, ions, and water molecules between the outer environment and active site pockets. The efficient exchange of ligands is a fundamental process of biocatalysis. Therefore, enzymes have evolved a wide range of mechanisms for repetitive conformational changes that enable periodic opening and closing. Protein-ligand interactions are traditionally studied by molecular docking, whereas molecular dynamics is the method of choice for studying conformational changes and ligand transport. However, computational demands make molecular dynamics impractical for screening purposes. Thus, several approximative methods have been recently developed to study interactions between a protein and ligand during the ligand transport process. Apart from identifying the best binding modes, these methods also provide information on the energetics of the transport and identify problematic regions limiting the ligand passage. These methods use approximations to simulate binding or unbinding events rapidly (calculation times from minutes to hours) and provide energy profiles that can be used to rank ligands or pathways. Here we provide a critical comparison of available methods, showcase their results on sample systems, discuss their practical applications in molecular biotechnologies and outline possible future developments.


Subject(s)
Biotechnology , Molecular Dynamics Simulation , Binding Sites , Ligands , Molecular Docking Simulation , Protein Binding , Water
4.
Comput Struct Biotechnol J ; 19: 3187-3197, 2021.
Article in English | MEDLINE | ID: mdl-34104357

ABSTRACT

The new severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causes pathological pulmonary symptoms. Most efforts to develop vaccines and drugs against this virus target the spike glycoprotein, particularly its S1 subunit, which is recognised by angiotensin-converting enzyme 2. Here we use the in-house developed tool CaverDock to perform virtual screening against spike glycoprotein using a cryogenic electron microscopy structure (PDB-ID: 6VXX) and the representative structures of five most populated clusters from a previously published molecular dynamics simulation. The dataset of ligands was obtained from the ZINC database and consists of drugs approved for clinical use worldwide. Trajectories for the passage of individual drugs through the tunnel of the spike glycoprotein homotrimer, their binding energies within the tunnel, and the duration of their contacts with the trimer's three subunits were computed for the full dataset. Multivariate statistical methods were then used to establish structure-activity relationships and select top candidate for movement inhibition. This new protocol for the rapid screening of globally approved drugs (4359 ligands) in a multi-state protein structure (6 states) showed high robustness in the rate of finished calculations. The protocol is universal and can be applied to any target protein with an experimental tertiary structure containing protein tunnels or channels. The protocol will be implemented in the next version of CaverWeb (https://loschmidt.chemi.muni.cz/caverweb/) to make it accessible to the wider scientific community.

5.
Methods Mol Biol ; 2266: 105-124, 2021.
Article in English | MEDLINE | ID: mdl-33759123

ABSTRACT

Interactions between enzymes and small molecules lie in the center of many fundamental biochemical processes. Their analysis using molecular dynamics simulations have high computational demands, geometric approaches fail to consider chemical forces, and molecular docking offers only static information. Recently, we proposed to combine molecular docking and geometric approaches in an application called CaverDock. CaverDock is discretizing enzyme tunnel into discs, iteratively docking with restraints into one disc after another and searching for a trajectory of the ligand passing through the tunnel. Here, we focus on the practical side of its usage describing the whole method: from getting the application, and processing the data through a workflow, to interpreting the results. Moreover, we shared the best practices, recommended how to solve the most common issues, and demonstrated its application on three use cases.


Subject(s)
Drug Discovery/methods , Molecular Docking Simulation/methods , Proteins/chemistry , Arachidonic Acid/chemistry , Binding Sites , Chlorohydrins/chemistry , Cytochrome P-450 Enzyme System/chemistry , Drug Design , Ethanol/analogs & derivatives , Ethanol/chemistry , Ethylene Dibromide/chemistry , Hydrolases/chemistry , Ligands , Molecular Dynamics Simulation , Protein Binding , Software , Structure-Activity Relationship , Thermodynamics
6.
IEEE/ACM Trans Comput Biol Bioinform ; 17(5): 1625-1638, 2020.
Article in English | MEDLINE | ID: mdl-30932844

ABSTRACT

Here we present a novel method for the analysis of transport processes in proteins and its implementation called CaverDock. Our method is based on a modified molecular docking algorithm. It iteratively places the ligand along the access tunnel in such a way that the ligand movement is contiguous and the energy is minimized. The result of CaverDock calculation is a ligand trajectory and an energy profile of transport process. CaverDock uses the modified docking program Autodock Vina for molecular docking and implements a parallel heuristic algorithm for searching the space of possible trajectories. Our method lies in between the geometrical approaches and molecular dynamics simulations. Contrary to the geometrical methods, it provides an evaluation of chemical forces. However, it is far less computationally demanding and easier to set up compared to molecular dynamics simulations. CaverDock will find a broad use in the fields of computational enzymology, drug design, and protein engineering. The software is available free of charge to the academic users at https://loschmidt.chemi.muni.cz/caverdock/.


Subject(s)
Drug Design/methods , Ligands , Molecular Docking Simulation/methods , Proteins , Algorithms , Biological Transport , Protein Binding/physiology , Protein Engineering , Proteins/chemistry , Proteins/metabolism , Proteins/ultrastructure
7.
Front Chem ; 7: 709, 2019.
Article in English | MEDLINE | ID: mdl-31737596

ABSTRACT

Protein tunnels and channels are attractive targets for drug design. Drug molecules that block the access of substrates or release of products can be efficient modulators of biological activity. Here, we demonstrate the applicability of a newly developed software tool CaverDock for screening databases of drugs against pharmacologically relevant targets. First, we evaluated the effect of rigid and flexible side chains on sets of substrates and inhibitors of seven different proteins. In order to assess the accuracy of our software, we compared the results obtained from CaverDock calculation with experimental data previously collected with heat shock protein 90α. Finally, we tested the virtual screening capabilities of CaverDock with a set of oncological and anti-inflammatory FDA-approved drugs with two molecular targets-cytochrome P450 17A1 and leukotriene A4 hydrolase/aminopeptidase. Calculation of rigid trajectories using four processors took on average 53 min per molecule with 90% successfully calculated cases. The screening identified functional tunnels based on the profile of potential energies of binding and unbinding trajectories. We concluded that CaverDock is a sufficiently fast, robust, and accurate tool for screening binding/unbinding processes of pharmacologically important targets with buried functional sites. The standalone version of CaverDock is available freely at https://loschmidt.chemi.muni.cz/caverdock/ and the web version at https://loschmidt.chemi.muni.cz/caverweb/.

8.
Nucleic Acids Res ; 47(W1): W414-W422, 2019 07 02.
Article in English | MEDLINE | ID: mdl-31114897

ABSTRACT

Caver Web 1.0 is a web server for comprehensive analysis of protein tunnels and channels, and study of the ligands' transport through these transport pathways. Caver Web is the first interactive tool allowing both the analyses within a single graphical user interface. The server is built on top of the abundantly used tunnel detection tool Caver 3.02 and CaverDock 1.0 enabling the study of the ligand transport. The program is easy-to-use as the only required inputs are a protein structure for a tunnel identification and a list of ligands for the transport analysis. The automated guidance procedures assist the users to set up the calculation in a way to obtain biologically relevant results. The identified tunnels, their properties, energy profiles and trajectories for ligands' passages can be calculated and visualized. The tool is very fast (2-20 min per job) and is applicable even for virtual screening purposes. Its simple setup and comprehensive graphical user interface make the tool accessible for a broad scientific community. The server is freely available at https://loschmidt.chemi.muni.cz/caverweb.


Subject(s)
Algorithms , Carrier Proteins/chemistry , Computational Biology/methods , User-Computer Interface , Amino Acid Sequence , Animals , Benchmarking , Binding Sites , Carrier Proteins/metabolism , Humans , Internet , Ligands , Molecular Docking Simulation , Protein Binding , Protein Interaction Domains and Motifs , Protein Structure, Quaternary , Protein Structure, Tertiary
9.
Bioinformatics ; 35(23): 4986-4993, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31077297

ABSTRACT

MOTIVATION: Protein tunnels and channels are key transport pathways that allow ligands to pass between proteins' external and internal environments. These functionally important structural features warrant detailed attention. It is difficult to study the ligand binding and unbinding processes experimentally, while molecular dynamics simulations can be time-consuming and computationally demanding. RESULTS: CaverDock is a new software tool for analysing the ligand passage through the biomolecules. The method uses the optimized docking algorithm of AutoDock Vina for ligand placement docking and implements a parallel heuristic algorithm to search the space of possible trajectories. The duration of the simulations takes from minutes to a few hours. Here we describe the implementation of the method and demonstrate CaverDock's usability by: (i) comparison of the results with other available tools, (ii) determination of the robustness with large ensembles of ligands and (iii) the analysis and comparison of the ligand trajectories in engineered tunnels. Thorough testing confirms that CaverDock is applicable for the fast analysis of ligand binding and unbinding in fundamental enzymology and protein engineering. AVAILABILITY AND IMPLEMENTATION: User guide and binaries for Ubuntu are freely available for non-commercial use at https://loschmidt.chemi.muni.cz/caverdock/. The web implementation is available at https://loschmidt.chemi.muni.cz/caverweb/. The source code is available upon request. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Subject(s)
Software , Algorithms , Binding Sites , Ligands , Molecular Docking Simulation , Proteins
10.
Appl Environ Microbiol ; 84(2)2018 01 15.
Article in English | MEDLINE | ID: mdl-29101190

ABSTRACT

The haloalkane dehalogenase enzyme DmmA was identified by marine metagenomic screening. Determination of its crystal structure revealed an unusually large active site compared to those of previously characterized haloalkane dehalogenases. Here we present a biochemical characterization of this interesting enzyme with emphasis on its structure-function relationships. DmmA exhibited an exceptionally broad substrate specificity and degraded several halogenated environmental pollutants that are resistant to other members of this enzyme family. In addition to having this unique substrate specificity, the enzyme was highly tolerant to organic cosolvents such as dimethyl sulfoxide, methanol, and acetone. Its broad substrate specificity, high overexpression yield (200 mg of protein per liter of cultivation medium; 50% of total protein), good tolerance to organic cosolvents, and a broad pH range make DmmA an attractive biocatalyst for various biotechnological applications.IMPORTANCE We present a thorough biochemical characterization of the haloalkane dehalogenase DmmA from a marine metagenome. This enzyme with an unusually large active site shows remarkably broad substrate specificity, high overexpression, significant tolerance to organic cosolvents, and activity under a broad range of pH conditions. DmmA is an attractive catalyst for sustainable biotechnology applications, e.g., biocatalysis, biosensing, and biodegradation of halogenated pollutants. We also report its ability to convert multiple halogenated compounds to corresponding polyalcohols.


Subject(s)
Bacteria/enzymology , Hydrolases/chemistry , Hydrolases/metabolism , Microbial Consortia/physiology , Bacteria/genetics , Bacteria/metabolism , Biocatalysis , Biotechnology , Catalysis , Catalytic Domain , Crystallization , Hydrogen-Ion Concentration , Hydrolases/genetics , Hydrolases/isolation & purification , Kinetics , Metagenome , Microbial Consortia/genetics , Substrate Specificity
11.
Nucleic Acids Res ; 44(W1): W479-87, 2016 07 08.
Article in English | MEDLINE | ID: mdl-27174934

ABSTRACT

HotSpot Wizard 2.0 is a web server for automated identification of hot spots and design of smart libraries for engineering proteins' stability, catalytic activity, substrate specificity and enantioselectivity. The server integrates sequence, structural and evolutionary information obtained from 3 databases and 20 computational tools. Users are guided through the processes of selecting hot spots using four different protein engineering strategies and optimizing the resulting library's size by narrowing down a set of substitutions at individual randomized positions. The only required input is a query protein structure. The results of the calculations are mapped onto the protein's structure and visualized with a JSmol applet. HotSpot Wizard lists annotated residues suitable for mutagenesis and can automatically design appropriate codons for each implemented strategy. Overall, HotSpot Wizard provides comprehensive annotations of protein structures and assists protein engineers with the rational design of site-specific mutations and focused libraries. It is freely available at http://loschmidt.chemi.muni.cz/hotspotwizard.


Subject(s)
Internet , Mutagenesis, Site-Directed/methods , Mutation , Peptide Library , Proteins/chemistry , Proteins/genetics , Software , Amino Acid Substitution , Automation , Biocatalysis , Databases, Protein , Evolution, Molecular , Models, Molecular , Protein Stability , Substrate Specificity
12.
Nanoscale ; 8(20): 10799-805, 2016 May 19.
Article in English | MEDLINE | ID: mdl-27166713

ABSTRACT

A multiferroic tunnel junction (MFTJ) promisingly offers multinary memory states in response to electric- and magnetic-fields, referring to tunneling electroresistance (TER) and tunneling magnetoresistance (TMR), respectively. In spite of recent progress, a substantial number of questions concerning the understanding of these two intertwined phenomena still remain open, e.g. the role of microstructural/chemical asymmetry at the interfaces of the junction and the effect of an electrode material on the MFTJ properties. In this regard, we look into the multiferroic effect of all-complex-oxide MFTJ (La0.7Sr0.3MnO3/Pb(Zr0.3Ti0.7)O3/La0.7Sr0.3MnO3). The results reveal apparent TER-TMR interplay-captured by the reversible electric-field control of the TMR effect. Finally, microscopy analysis on the MFTJ revealed that the observed TER-TMR interplay is perhaps mediated by microstructural and chemical asymmetry in our nominally symmetric MFTJ.

13.
Nat Commun ; 5: 5414, 2014 Nov 17.
Article in English | MEDLINE | ID: mdl-25399545

ABSTRACT

Among recently discovered ferroelectricity-related phenomena, the tunnelling electroresistance (TER) effect in ferroelectric tunnel junctions (FTJs) has been attracting rapidly increasing attention owing to the emerging possibilities of non-volatile memory, logic and neuromorphic computing applications of these quantum nanostructures. Despite recent advances in experimental and theoretical studies of FTJs, many questions concerning their electrical behaviour still remain open. In particular, the role of ferroelectric/electrode interfaces and the separation of the ferroelectric-driven TER effect from electrochemical ('redox'-based) resistance-switching effects have to be clarified. Here we report the results of a comprehensive study of epitaxial junctions comprising BaTiO(3) barrier, La(0.7)Sr(0.3)MnO(3) bottom electrode and Au or Cu top electrodes. Our results demonstrate a giant electrode effect on the TER of these asymmetric FTJs. The revealed phenomena are attributed to the microscopic interfacial effect of ferroelectric origin, which is supported by the observation of redox-based resistance switching at much higher voltages.

14.
Nanoscale ; 5(24): 12598-606, 2013 Dec 21.
Article in English | MEDLINE | ID: mdl-24177268

ABSTRACT

Bipolar switching behaviours of electrochemical metallization (ECM) cells with dual-layer solid electrolytes (SiOx-Ge0.3Se0.7) were analyzed. Type 1 ECM cell, Pt (bottom electrode)/SiOx/Ge0.3Se0.7/Cu (top electrode), exhibited typical eightwise current-voltage (I-V) hysteresis of ECM cells whereas Type 2 ECM cell, Pt (bottom electrode)/Ge0.3Se0.7/SiOx/Cu(top electrode), showed counter-eightwise hysteresis. In addition, absolute off-switching voltage in Type 2 cell is lower than that in Type 1 cell while on-switching voltage in both cells is almost the same. An attempt to understand this electrolyte-stack-sequence-depending switching polarity reversal was made in terms of the ECM cell potential change upon the electrolyte stack sequence and the consequent change in Cu filament growth direction. Relevant experimental evidence for the hypothesis was obtained regarding the switching behaviours. Furthermore, given the switching polarity reversal, feasibility of serial complementary resistive switches was also demonstrated.

SELECTION OF CITATIONS
SEARCH DETAIL
...