16 research outputs found

    CaverDock: Software tool for fast screening of un/binding of ligands in protein engineering

    Get PDF
    Protein tunnels, channels and gates are important for enzymatic catalysis and also represent attractive targets for rational protein design and drug design [1]. Drug molecules blocking the access of natural substrate or release of products are very efficient modulators of biological activity. Here we demonstrate the application of newly in-house developed software tool CaverDock [2,3] for the analysis of the transport of ligands through tunnels in biomolecular targets. Caverdock is a new addition to the Caver Suite [4-6]. We performed virtual screening of large databases of drugs against two pharmacologically relevant targets. We have used FDA-approved drugs for both targets. Oncological drugs (133 molecules), taken from the NIH website, and anti-inflammatory (56 molecules), taken from the Drugbank website, as the libraries of ligands for the two molecular targets: (i) cytochrome P450 17A1 and (ii) leukotriene A4 hydrolase/aminopeptidase. Moreover, we will also show the unbinding of the 2,3-dichloropropan-1-ol product from a buried active site of an haloalkane dehalogenase and its variant. With this study we identified hot-spots that may be used for directed evolution or site-directed mutagenesis to create new variants for faster 2,3-dichloropropan-1-ol release [7]. Finally, we will show the difference on ligand transportation when a protein is in an open and closed conformations [8]. We will show how CaverDock tackles the problem of protein flexibility. 1. Marques, S.M., et al., 2017: Enzyme Tunnels and Gates as Relevant Targets in Drug Design. Medicinal Research Reviews 37: 1095-1139. 2. Vavra, O., et al., 2019: CaverDock 1.0: A New Tool for Analysis of Ligand Binding and Unbinding Based on Molecular Docking. Bioinformatics (under review). 3. Filipovic, J., et al, 2019: A Novel Method for Analysis of Ligand Binding and Unbinding Based on Molecular Docking. Transactions on Computational Biology and Bioinformatics (under review) 4. Chovancova, E., et al., 2012: CAVER 3.0: A Tool for Analysis of Transport Pathways in Dynamic Protein Structures. PLOS Computational Biology 8: e1002708. 5. Jurcik, A., et al., 2018: CAVER Analyst 2.0: Analysis and Visualization of Channels and Tunnels in Protein Structures and Molecular Dynamics Trajectories. Bioinformatics 34: 3586-3588. 6. Stourac, J., et al., 2019: Caver Web 1.0: Identification of Tunnels and Channels in Proteins and Analysis of Ligand Transport. Nucleic Acids Research (under review). 7. Marques, S.M., et al., 2019: Computational Study of Protein-Ligand Unbinding for Enzyme Engineering. Frontiers in Chemistry 6: 650. 8. Kokkonen, P., et al., 2018: Molecular Gating of an Engineered Enzyme Captured in Real Time. Journal of the American Chemical Society 140: 17999–18008

    Strategies and software tools for engineering protein tunnels and dynamical gates

    Get PDF
    Improvements in the catalytic activity, substrate specificity or enantioselectivity of enzymes are traditionally achieved by modification of enzymes’ active sites. We have recently proposed that the enzyme engineering endeavors should target both the active sites and the access tunnels/channels [1,2]. Using the model enzymes haloalkane dehalogenases, we have demonstrated that engineering of access tunnels provides enzymes with significantly improved catalytic properties [3] and stability [4]. User-friendly software tools Caver [5], Caver Analyst [6], CaverDock [7] and Caver Web [8], have been developed for the computational design of protein tunnels/channels; FireProt [9] and HotSpot Wizard [10] for automated design of stabilizing mutations and smart libraries. Using these tools we were able to introduce a new tunnel to a protein structure and tweak its conformational dynamics. This engineering strategy has led to improved catalytic efficiency [2], enhanced promiscuity or even a functional switch (unpublished). Our concepts and software tools are widely applicable to various enzymes with known structures and buried active sites. 1. Damborsky, J., et al., 2009: Computational Tools for Designing and Engineering Biocatalysts. Current Opinion in Chemical Biology 13: 26-34. 2. Prokop, Z., et al., 2012: Engineering of Protein Tunnels: Keyhole-lock-key Model for Catalysis by the Enzymes with Buried Active Sites. Protein Engineering Handbook, Wiley-VCH, Weinheim, pp. 421-464. 3. Brezovsky, J., et al., 2016: Engineering a de Novo Transport Tunnel. ACS Catalysis 6: 7597-7610. 4. Koudelakova, T., et al., 2013: Engineering Enzyme Stability and Resistance to an Organic Cosolvent by Modification of Residues in the Access Tunnel. Angewandte Chemie 52: 1959-1963. 5. Chovancova, E., et al., 2012: CAVER 3.0: A Tool for Analysis of Transport Pathways in Dynamic Protein Structures. PLOS Computational Biology 8: e1002708. 6. Jurcik, A., et al., 2018: CAVER Analyst 2.0: Analysis and Visualization of Channels and Tunnels in Protein Structures and Molecular Dynamics Trajectories. Bioinformatics 34: 3586-3588. 7. Vavra, O., et al., 2019: CaverDock 1.0: A New Tool for Analysis of Ligand Binding and Unbinding Based on Molecular Docking. Bioinformatics (under review). 8. Stourac, J., et al. 2019: Caver Web 1.0: Identification of Tunnels and Channels in Proteins and Analysis of Ligand Transport. Nucleic Acids Research (under review). 9. Musil, M., et al., 2017: FireProt: Web Server for Automated Design of Thermostable Proteins. Nucleic Acids Research 45: W393-W399. 10. Sumbalova, L. et al., 2018: HotSpot Wizard 3.0: Automated Design of Site-Specific Mutations and Smart Libraries in Protein Engineering. Nucleic Acids Research 46: W356-W362

    CAVERDOCK: A new tool for analysis of ligand binding and unbinding based on molecular docking

    Get PDF
    Understanding the protein-ligand interactions is crucial for engineering improved catalysts. The interaction of a protein and a ligand molecule often takes place in enzymes active site. Such functional sites may be buried inside the protein core, and therefore a transport of a ligand from outside environment to the protein inside needs to be understood. Here we present the CaverDock [1], implementing a novel method for analysis of these important transport processes. Our method is based on a modified molecular docking algorithm. It iteratively places the ligand along the tunnel in such a way that the ligand movement is contiguous and its energy is minimized. The output of the calculation is ligand trajectory and energy profile of transport process. CaverDock uses a modified version of the program AutoDock Vina [2] for molecular docking and implements a parallel heuristic algorithm to search the space of possible trajectories. Our method lies in between of geometrical approaches and molecular dynamics simulations. Contrary to geometrical methods, it provides an evaluation of chemical forces. However, it is not as computationally demanding as the methods based on molecular dynamics. The typical input of CaverDock requires setup for molecular docking and tunnel geometry obtained from Caver [3]. Typical computational time is in dozens of minutes at a single node, allowing virtual screening of a large pool of molecules. We demonstrate CaverDock usability by comparison of a ligand trajectory in different tunnels of wild type and engineered proteins; and computation of energetic profiles for a large set of substrates and inhibitors. CaverDock is available from the web site http://www.caver.cz. 1. Vavra, O., Filipovic, J., Plhak, J., Bednar, D., Marques, S., Brezovsky, J., Matyska, L., Damborsky, J., CAVERDOCK: A New Tool for Analysis of Ligand Binding and Unbinding Based on Molecular Docking. PLOS Computational Biology (submitted). 2. Trott, O., Olson, A.J., AutoDock Vina: Improving the Speed and Accuracy of Docking with a New scoring function, efficient optimization and multithreading, Journal of Computational Chemistry 31: 455-461, 2010. 3. Chovancova, E., Pavelka, A., Benes, P., Strnad, O., Brezovsky, J., Kozlikova, B., Gora, A., Sustr, V., Klvana, M., Medek, P., Biedermannova, L., Sochor, J., Damborsky, J., 2012: CAVER 3.0: A Tool for Analysis of Transport Pathways in Dynamic Protein Structures. PLOS Computational Biology 8: e1002708

    Understanding the dynamic evolution of cyclic variability at the operating limits of HCCI engines with negative valve overlap.

    Get PDF
    ABSTRACT An experimental study is performed for homogeneous charge compression ignition (HCCI) combustion focusing on late phasing conditions with high cyclic variability (CV) approaching misfire. High CV limits the feasible operating range and the objective is to understand and quantify the dominating effects of the CV in order to enable controls for widening the operating range of HCCI. A combustion analysis method is developed for explaining the dynamic coupling in sequences of combustion cycles where important variables are residual gas temperature, combustion efficiency, heat release during re-compression, and unburned fuel mass. The results show that the unburned fuel mass carries over to the re-compression and to the next cycle creating a coupling between cycles, in addition to the well known temperature coupling, that is essential for understanding and predicting the HCCI behavior at lean conditions with high CV

    Precise Implementation of CDTA

    Get PDF
    The Current Differencing Transconductance Amplifier (CDTA) is a popular active building block for analog signal processing. Since it is not available as a commercial IC, most of the CDTA applications are verified only via simulations or the CDTA is implemented by commercial circuits. The paper deals with CDTA implementation, emphasizing the accuracy, linearity, and dynamic range of the processed signals, and low power consumption. Such a CDTA can be useful wherever these requirements are more relevant than the speed and bandwidth, typically for implementing hardware emulators of memristive systems

    The Future Possibilities and Security Challenges of City Digitalization

    No full text
    To adapt to current changes, such as globalization, climate change, and demographic growth, modern cities must embrace the digitalization of city management. In this paper, we examine a concept for digitalizing a city based on Rotterdam’s digital twin showcase. Data-processing frameworks for different sources of data are presented. Security risks and the potential of smart cities for military usage are discussed. Lastly, using an example of available datasets for air quality and public lighting consumption, functions of the Rotterdam digital twin are compared with the Brno city digital platform. It was found that every city has its approach to digitalization, and it is probably impossible to unify every city’s digitalization process. This means that the digitalization of the city is very individual. Both systems in their current form make it possible to visualize the collected data about the city. So far, however, these systems do not use advanced functions such as AI-assisted decision-making and prediction of various events in the city. Even so, they are a source of very interesting data that can be used by third parties

    Screening of World Approved Drugs against Highly Dynamical Spike Glycoprotein SARS-CoV-2 using CaverDock and Machine Learning

    No full text
    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 simulations. 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 molecules. This new protocol for rapid screening of globally approved drugs (4359 ligands) in a multi-state protein structure (6 states) required a total of 26,148 calculations and showed high robustness. 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 communit

    Morphology, Molecular Phylogeny, and Ecology of Binucleata daphniae n. g., n. sp (Fungi: Microsporidia), a Parasite of Daphnia magna Straus, 1820 (Crustacea: Branchiopoda)

    No full text
    We describe a new microsporidian species Binucleata daphniae, n. g., n. sp., that infects the integument cells lining the hemocoele cavity of the carapace and the postabdomen of the cladoceran Daphnia magna Straus. Infected cells filled with spores accumulate as large clusters in the carapace cavity and heavily infected hosts are detected by their opaque appearance. Despite the parasite's presence, infected Daphnia grow and molt, but have a reduced fecundity. During the parasite's life cycle, chain-like meronts with isolated nuclei are formed, giving rise to binucleate presporonts, the most frequently observed, characteristic developmental stage. In sporogony, the nuclei of the presporont separate, divide, and eight spores enclosed in a thin-walled sporophorous vesicle are formed. Spores are 4.9 x 2.5 mu m in size (fresh) and have an anisofilar polar filament with eight coils. DNA sequence analysis places B. daphniae in a clade of microsporidians that parasitize crustaceans and mosquitoes and have assumed complex life cycles. Binucleata daphniae, however, has a simple and direct life cycle and can be transferred to naive hosts and maintained as persistent infections in populations of its host D. magna. We propose that B. daphniae has simplified its life cycle by losing its secondary host, rendering it unique in this clade.status: publishe

    Long Distance Military Fiber-Optic Polarization Sensor Improved by an Optical Amplifier

    Get PDF
    The ever-increasing demands for the use of fiber-optic sensors powered by long optical fibers is forcing developers to solve problems associated with powering these remote sensors. Due to their non-electric character, these sensors are suitable for many uses, including military applications. The Army of the Czech Republic is very interested in this type of optical fiber sensor as it fulfils the significant prerequisites for use in military areas. However, the army’s requirements are challenging because they require long supply cables in which there is significant attenuation of optical power. At the same time, there is a need for high sensitivity. The subject of our research team’s work was to use amplifiers to power these sensors. The army already uses this type of sensor for short distances as it cannot ignite a gas mixture with an explosive concentration and thus meet the strict requirements of the explosion-poof standard. The novelty of our research lies in the discovered measurement technique that allows the sensors to be powered remotely and in the saving of optical fibers by utilizing duplex communication with a circulator. Furthermore, the research presents an innovative approach to the optimization of the entire sensor by using a bidirectional, sensory, polarization-maintaining optical fiber. The proposed sensor was first verified in laboratory conditions at the Optoelectronics Laboratory of the University of Defense in Brno, and further tests were carried out in the military training areas of Boletice and Březina in the Czech Republic, which is a member of North Atlantic Treaty Organization
    corecore