scispace - formally typeset
Search or ask a question

Answers from top 3 papers

More filters
Papers (3)Insight
The results indicate that the use of a multiple-ligand representation is superior to a single-conformer concept and reduces the user bias associated with the ligand alignment.
may be involved in ligand binding and could find fertile applications in ligand design.
We also show that positional restraints can be applied to the large ligand using DINC: this is useful when computing a docked conformation of the ligand.

Related Questions

How to do ligand docking in protein?5 answersLigand docking in proteins involves several steps. First, a suitable search strategy and scoring function are needed for efficient and effective docking. One approach is to use AutoDock Vina, which employs an exhaustiveness parameter and a grid box to define the ligand binding site. Another method, FitDock, utilizes a hierarchical multi-feature alignment approach to fit the initial conformation to a given template, resulting in improved docking success rates and faster performance. Additionally, flexible protein-ligand docking can be achieved using a deep learning model based on the prediction of an intermolecular Euclidean distance matrix, which outperforms traditional docking methods. Finally, computational molecular docking can be used to generate a density of binding states, which can be used to calculate binding strengths, conformations, and atomic interactions between the ligand and protein.
What techniques can be used to understand the binding of ligand with the protein?5 answersSeveral techniques can be used to understand the binding of a ligand with a protein. High-resolution structural approaches, such as X-ray crystallography, provide a static view of the protein-ligand complex. Molecular docking simulations, based on force fields like Martini, can accurately predict binding sites and pathways, as well as binding free energies. Big Data analytics techniques, including machine learning and deep learning, can efficiently process a large number of protein-ligand complexes and identify specific protein properties responsible for correct prediction of binding. Differential scanning calorimetry (DSC) can determine the enthalpy change upon protein unfolding and the melting temperature of the protein, providing information on ligand binding. These techniques, along with thermodynamic treatments and experimental designs, can enhance our understanding of protein-ligand interactions.
What is a ligand in biology?5 answersA ligand in biology refers to a molecule that binds to a receptor, enabling communication and interaction between cells and their environment. Ligands can be a wide range of biomolecules, including proteins, carbohydrates, and small molecules. In biological activities, ligands are involved in contact interactions with their complementary partners, such as enzymes and substrates, antigens and immunoglobulins, or neurotransmitters and receptor sites. Ligands can be used in various applications, such as targeting specific cell types or delivering bio-active compounds to cells expressing a particular receptor. Ligand identification methods, such as using oligonucleotides or analogues, can help identify ligands that interact specifically with a target molecule. In the field of chemical genetics, ligands are used to alter protein function and study the consequences of these changes in a cellular or organismal context.
How do you choose ligands for docking?6 answers
How do you prepare ligands for a dock?5 answers
How to select variables for SVM?7 answers

See what other people are reading

How to predict the structure of inorganic metal ion complex by uv spectrum graph?
4 answers
To predict the structure of inorganic metal ion complexes using UV spectrum graphs, various spectroscopic and computational techniques can be employed. Deep UV resonance Raman spectroscopy can provide intrinsic sensitivity and selectivity for characterizing ligand-metal ion complexes. Additionally, machine learning approaches, such as deep graph learning with natural quantum graphs (NatQG), can leverage electronic structure data for transition metal complexes (TMCs) to predict quantum properties like HOMO-LUMO gap and polarizability, surpassing traditional descriptors and achieving high accuracy levels. Graph neural networks (GNNs) can be utilized to learn properties of atomic structures, incorporating angular information crucial for describing complex atomic arrangements in metal complexes, enabling efficient graph encoding of atomic structures for spectroscopy prediction. These combined approaches offer a comprehensive strategy for predicting the structure of inorganic metal ion complexes from UV spectrum graphs.
What is heterocyclic azo ligand?
5 answers
A heterocyclic azo ligand is a type of ligand synthesized by combining heterocyclic compounds with azo groups. These ligands have been extensively studied for their coordination chemistry with various metal ions to form complexes with diverse properties. For example, studies have reported the synthesis of heterocyclic azo ligands derived from compounds like antipyrine, pyrazole, and imidazole, which have shown promising antimicrobial and anticancer activities. The coordination of these ligands with metal ions like Co(II), Cu(II), and Zn(II) has been characterized using techniques such as FT-IR, UV-Vis, and NMR spectroscopy, revealing their tridentate binding behavior and octahedral geometry in the resulting complexes. Additionally, theoretical calculations and molecular docking studies have provided insights into the structural and electronic properties of these ligands and their complexes, further highlighting their potential for future investigations.
What is homocyclic azo ligand?
5 answers
A homocyclic azo ligand refers to a ligand where the azo group (-N=N-) is part of a cyclic structure within the molecule. In the provided research contexts, various homocyclic azo ligands were synthesized and used to form metal complexes. For example, ligands derived from compounds like 5-amino-3-methyl-1-phenylpyrazole, 1-phenyl-2-pyrazolin-5-one, 2-amino-5-methyl thiazole, 4-aminoantipyrine, and 4-aminoacetophenonewere employed to create azo ligands with homocyclic structures. These ligands were then utilized to form complexes with various metal ions like Co(II), Ni(II), Cu(II), Ag(I), and Au(III). The characterization and evaluation of these complexes included techniques such as elemental analysis, spectroscopy, and biological activity testing against different microorganisms and cancer cells.
How is the IC50 calculated for protein-ligand?
5 answers
The IC50 value for a protein-ligand interaction is typically calculated based on the inhibitory concentration at which 50% of the protein's activity is inhibited by the ligand. This value is crucial in assessing the potency of a ligand as an inhibitor. Computational methods like free energy perturbation (FEP) and molecular dynamics (MD) simulations play a significant role in predicting protein-ligand binding affinities, aiding in IC50 determination. Experimental techniques such as isothermal titration calorimetry (ITC) provide essential thermodynamic parameters like affinity (Ka), enthalpy change (∆H), and free energy change (∆G), which are fundamental in understanding the binding process and can indirectly contribute to IC50 determination. Additionally, computational analyses of protein-ligand interactions, focusing on scoring functions and electrostatic interactions, are pivotal in evaluating ligand binding energy and potential as an inhibitor.
What is an intermolecular energy of protein-ligand?
5 answers
The intermolecular energy of a protein-ligand complex refers to the interaction energy between the protein and the ligand, which plays a crucial role in binding affinity and drug design. Various methods, such as dispersion-corrected density functional theory (DFT-D3) and multilayer molecules-in-molecules (MIM) fragmentation-based approaches, are utilized to accurately calculate these interaction energies at a molecular level. These methods allow for the decomposition of total binding affinity into residue-specific contributions, providing insights into the key interactions that determine the binding strength between proteins and ligands. Understanding intermolecular energies is essential for predicting and optimizing the effectiveness of computational drug design by identifying critical residues and interactions that influence binding affinities.
How do multi-functional drugs deal with different IC-50s?
5 answers
Multi-functional drugs address different IC-50s by targeting multiple pathways or receptors simultaneously, enhancing efficacy against complex diseases. These drugs are designed to interact with various targets, allowing for a broader spectrum of action and potentially overcoming variations in IC-50 values for different targets. By utilizing Molecular Docking in Computer-Aided Drug Design, researchers can identify compounds that interact with multiple targets, optimizing therapeutic outcomes. Additionally, the multifunctional drug boxes and cases described in other contexts provide convenient and organized storage for these complex medications, ensuring proper administration and adherence to treatment regimens. Overall, the multifunctional nature of these drugs enables a more comprehensive approach to addressing diseases with varying IC-50 values for different targets.
How does the formation of chalcogen bonds contribute to the stability and reactivity of molecules?
4 answers
The formation of chalcogen bonds plays a crucial role in stabilizing molecules and influencing their reactivity. Chalcogen bonds involve interactions between electrophilic chalcogen atoms (such as sulfur, selenium, or tellurium) and nucleophilic regions, resembling hydrogen and halogen bonds in strength and directionality. These bonds are observed in various molecular contexts, including protein structures, metal complexes, and drug design studies. Chalcogen bonding contributes to structural stability by rigidifying reagents and influencing conformational flexibility. Additionally, chalcogen bonds exhibit a significant role in the interaction energy decomposition of molecules, affecting their preferred conformations and overall stability. The presence of chalcogen bonds, particularly in hydrated sulfur dioxide clusters, showcases a cooperative effect with hydrogen bonding, enhancing the stability of the systems.
What is stripping of ligand?
5 answers
Ligand stripping refers to the process of removing stabilizing ligands from the surface of colloidal nanoparticles (NPs). This process is crucial for controlling the properties of NPs, such as their coverage and reactivity. Ligand stripping can be achieved using poor solvents, which interact with the ligands and facilitate their removal from the NP surface. However, the mechanism behind ligand stripping is complex and involves interactions between the solvent, ligands, and NP surface. Studies have shown that ligand stripping can be influenced by factors like the concentration of the solvent and the enthalpy of mixing of the ligands and solvents. Understanding ligand stripping is essential for various applications of NPs, including self-assembly, optoelectronics, nanomedicine, and catalysis.
What is the IC50 value for minoxodil for 5 alpha reductase inhibitor?
5 answers
The IC50 value for minoxidil as a 5 alpha-reductase inhibitor was found to be 62 nM for stromal cells and 45 nM for epithelial cells in benign prostatic hyperplasia (BPH) tissue. Additionally, a study on indole derivatives showed that compound KF20405 exhibited potent inhibitory activity against rat prostatic 5 alpha-reductase with an IC50 value of 0.48 ± 0.086 nM, indicating high potency as compared to other inhibitors like MK-906. However, the specific IC50 value for minoxidil as a 5 alpha-reductase inhibitor was not directly provided in the available contexts.
Structural based pharmacophore modeling with free software?
5 answers
Structure-based pharmacophore modeling for ligand discovery in G protein-coupled receptors (GPCRs) can be effectively conducted using free software. Studies have shown the successful generation of high-performing pharmacophore models for GPCRs using fragments placed with Multiple Copy Simultaneous Search (MCSS). These models have exhibited high enrichment factors when applied to search databases containing GPCR ligands. Additionally, the use of a cluster-then-predict machine learning workflow has enabled the selection of pharmacophore models with positive predictive values for high enrichment models in both experimentally determined and modeled GPCR structures. Such approaches have proven useful in virtual screening efforts, aiding in the identification of novel ligands for GPCRs with limited known ligands or structures.
Did autodocktools optimized the ligand before docking?
5 answers
AutoDockTools did not directly optimize the ligand before docking. However, AutoDock Vina, a related software, was optimized to enhance ligand-binding affinity predictions by adjusting its parameters. Additionally, a study demonstrated that a convolutional neural network (CNN) trained to identify binding poses could be utilized for pose optimization, outperforming AutoDock Vina in optimizing randomly initialized poses. While AutoDock and AutoDockTools are essential in structure-based drug design, they do not specifically optimize ligands before docking. Instead, they focus on predicting binding modes and affinities, allowing for virtual screening of ligand libraries.